PMID-sentid Pub_year Sent_text comp_official_name comp_offset protein_name organism prot_offset 33522686-13 2021 CONCLUSION: Verbascoside mitigated the cell proliferation and aggressiveness of prostate cancer via downregulation of TGF-beta-associated EMT progression through HMGB1/RAGE suppression. acteoside 12-24 high mobility group box 1 Homo sapiens 162-167 34054222-8 2021 EGCG also shows protective effects against 1) cytokine storm-associated acute lung injury/acute respiratory distress syndrome, 2) thrombosis via suppressing tissue factors and activating platelets, 3) sepsis by inactivating redox-sensitive HMGB1, and 4) lung fibrosis through augmenting Nrf2 and suppressing NF-kappaB. epigallocatechin gallate 0-4 high mobility group box 1 Homo sapiens 240-245 3405231-2 1988 One of the satellite binding proteins (SBP1) purified by column chromatography using DEAE-, phospho- and DNA-cellulose steps interacted also with adenovirus 5 replication enhancer (ARE), another protein (SBP2) was separated during phosphocellulose chromatography from ARE-binding protein. 2-diethylaminoethanol 85-89 high mobility group box 1 Homo sapiens 39-43 5164097-2 1971 These are a simple synthesis of the substrate precursor HMG-3-(14)C anhydride and a double-label ((14)C and (3)H) method for determining the amount of mevalonate-3-(14)C that is formed from the substrate. Mevalonic Acid 151-161 high mobility group box 1 Homo sapiens 56-61 34007314-3 2021 In the present study, the exact molecular mechanism underlying HMGB1-mediated drug resistance in MM was explored using three chemotherapy-resistant MM cells (RPMI8226/ADR, RPMI8226/BOR and RPMI8226/DEX) that were successfully established. rpmi8226 158-166 high mobility group box 1 Homo sapiens 63-68 34007314-3 2021 In the present study, the exact molecular mechanism underlying HMGB1-mediated drug resistance in MM was explored using three chemotherapy-resistant MM cells (RPMI8226/ADR, RPMI8226/BOR and RPMI8226/DEX) that were successfully established. rpmi8226 172-180 high mobility group box 1 Homo sapiens 63-68 34007314-3 2021 In the present study, the exact molecular mechanism underlying HMGB1-mediated drug resistance in MM was explored using three chemotherapy-resistant MM cells (RPMI8226/ADR, RPMI8226/BOR and RPMI8226/DEX) that were successfully established. Boron 181-184 high mobility group box 1 Homo sapiens 63-68 34007314-3 2021 In the present study, the exact molecular mechanism underlying HMGB1-mediated drug resistance in MM was explored using three chemotherapy-resistant MM cells (RPMI8226/ADR, RPMI8226/BOR and RPMI8226/DEX) that were successfully established. rpmi8226 172-180 high mobility group box 1 Homo sapiens 63-68 34007314-3 2021 In the present study, the exact molecular mechanism underlying HMGB1-mediated drug resistance in MM was explored using three chemotherapy-resistant MM cells (RPMI8226/ADR, RPMI8226/BOR and RPMI8226/DEX) that were successfully established. Dextromethorphan 198-201 high mobility group box 1 Homo sapiens 63-68 34007314-4 2021 Reverse transcription-quantitative polymerase chain reaction revealed that the three chemotherapy-resistant MM cells exhibited a higher release of HMGB1 compared with the parental RPMI8226 cells. rpmi8226 180-188 high mobility group box 1 Homo sapiens 147-152 33986864-10 2021 HMGB1 knockdown increased nicotinamide adenine nucleotide phosphate oxidase-mediated ROS production and induced DNA damage via the MAPK signaling pathway, which may promote apoptosis and radiosensitivity after radiation in TE-1 cells. Niacinamide 26-38 high mobility group box 1 Homo sapiens 0-5 33986864-10 2021 HMGB1 knockdown increased nicotinamide adenine nucleotide phosphate oxidase-mediated ROS production and induced DNA damage via the MAPK signaling pathway, which may promote apoptosis and radiosensitivity after radiation in TE-1 cells. Reactive Oxygen Species 85-88 high mobility group box 1 Homo sapiens 0-5 33716257-4 2021 Ethanol consumption leads to the activation of neuroimmune signaling in the central nervous system through many subtypes of Toll-like receptors (TLRs), as well as release of their endogenous agonists (high-mobility group protein B1 (HMGB1), S100 protein, heat shock proteins (HSPs), and extracellular matrix degradation proteins). Ethanol 0-7 high mobility group box 1 Homo sapiens 201-231 34054124-10 2021 CONCLUSIONS This study demonstrated that the rs1360485 SNP of the HMGB1 gene is associated with susceptibility of NEC in neonates, and the rs1360485 genotypes TC and CC may affect HMGB1 expression and are associated with the survival prognosis of neonates with NEC. Technetium 159-161 high mobility group box 1 Homo sapiens 66-71 34054124-10 2021 CONCLUSIONS This study demonstrated that the rs1360485 SNP of the HMGB1 gene is associated with susceptibility of NEC in neonates, and the rs1360485 genotypes TC and CC may affect HMGB1 expression and are associated with the survival prognosis of neonates with NEC. Technetium 159-161 high mobility group box 1 Homo sapiens 180-185 34024230-9 2022 Knockdown of HMGB1 expression rescued LMP, restored the degradative capacity of autophagy, decreased the expression of inflammatory factors and VEGF (vascular endothelial growth factor), and protected against apoptosis in RPE cells in the early stages of DR. (2~{s},3~{r},4~{s})-2-[(2~{s},3~{r})-1,3-Bis(Oxidanyl)-1-Oxidanylidene-Butan-2-Yl]-4-[(3~{s},5~{s})-5-(Dimethylcarbamoyl)pyrrolidin-3-Yl]sulfanyl-3-Methyl-3,4-Dihydro-2~{h}-Pyrrole-5-Carboxylic Acid 38-41 high mobility group box 1 Homo sapiens 13-18 33716257-4 2021 Ethanol consumption leads to the activation of neuroimmune signaling in the central nervous system through many subtypes of Toll-like receptors (TLRs), as well as release of their endogenous agonists (high-mobility group protein B1 (HMGB1), S100 protein, heat shock proteins (HSPs), and extracellular matrix degradation proteins). Ethanol 0-7 high mobility group box 1 Homo sapiens 233-238 33621742-0 2021 Corrigendum to "Paclitaxel alleviates the sepsis-induced acute kidney injury via lnc-MALAT1/miR-370-3p/HMGB1 axis" [Life Sci. Paclitaxel 16-26 high mobility group box 1 Homo sapiens 103-108 33975537-0 2021 Therapeutic administration of etoposide coincides with reduced systemic HMGB1 levels in macrophage activation syndrome. Etoposide 30-39 high mobility group box 1 Homo sapiens 72-77 33107103-4 2021 Different isoforms of HMGB1 present with distinctive physiological functions in ECM-fully-reduced HMGB1 (all thiol) acts as the initial damage signal to recruit circulating myeloid cells, disulfide HMGB1 behaves as a cytokine to activate macrophages and neutrophils, and both signals are turned off when HMGB1 is terminally oxidized into the final sulfonate form. Sulfhydryl Compounds 109-114 high mobility group box 1 Homo sapiens 22-27 33107103-4 2021 Different isoforms of HMGB1 present with distinctive physiological functions in ECM-fully-reduced HMGB1 (all thiol) acts as the initial damage signal to recruit circulating myeloid cells, disulfide HMGB1 behaves as a cytokine to activate macrophages and neutrophils, and both signals are turned off when HMGB1 is terminally oxidized into the final sulfonate form. Sulfhydryl Compounds 109-114 high mobility group box 1 Homo sapiens 98-103 33107103-4 2021 Different isoforms of HMGB1 present with distinctive physiological functions in ECM-fully-reduced HMGB1 (all thiol) acts as the initial damage signal to recruit circulating myeloid cells, disulfide HMGB1 behaves as a cytokine to activate macrophages and neutrophils, and both signals are turned off when HMGB1 is terminally oxidized into the final sulfonate form. Sulfhydryl Compounds 109-114 high mobility group box 1 Homo sapiens 98-103 33107103-4 2021 Different isoforms of HMGB1 present with distinctive physiological functions in ECM-fully-reduced HMGB1 (all thiol) acts as the initial damage signal to recruit circulating myeloid cells, disulfide HMGB1 behaves as a cytokine to activate macrophages and neutrophils, and both signals are turned off when HMGB1 is terminally oxidized into the final sulfonate form. Sulfhydryl Compounds 109-114 high mobility group box 1 Homo sapiens 98-103 33107103-4 2021 Different isoforms of HMGB1 present with distinctive physiological functions in ECM-fully-reduced HMGB1 (all thiol) acts as the initial damage signal to recruit circulating myeloid cells, disulfide HMGB1 behaves as a cytokine to activate macrophages and neutrophils, and both signals are turned off when HMGB1 is terminally oxidized into the final sulfonate form. Disulfides 188-197 high mobility group box 1 Homo sapiens 22-27 33107103-4 2021 Different isoforms of HMGB1 present with distinctive physiological functions in ECM-fully-reduced HMGB1 (all thiol) acts as the initial damage signal to recruit circulating myeloid cells, disulfide HMGB1 behaves as a cytokine to activate macrophages and neutrophils, and both signals are turned off when HMGB1 is terminally oxidized into the final sulfonate form. Disulfides 188-197 high mobility group box 1 Homo sapiens 98-103 33107103-4 2021 Different isoforms of HMGB1 present with distinctive physiological functions in ECM-fully-reduced HMGB1 (all thiol) acts as the initial damage signal to recruit circulating myeloid cells, disulfide HMGB1 behaves as a cytokine to activate macrophages and neutrophils, and both signals are turned off when HMGB1 is terminally oxidized into the final sulfonate form. Disulfides 188-197 high mobility group box 1 Homo sapiens 98-103 33107103-4 2021 Different isoforms of HMGB1 present with distinctive physiological functions in ECM-fully-reduced HMGB1 (all thiol) acts as the initial damage signal to recruit circulating myeloid cells, disulfide HMGB1 behaves as a cytokine to activate macrophages and neutrophils, and both signals are turned off when HMGB1 is terminally oxidized into the final sulfonate form. Disulfides 188-197 high mobility group box 1 Homo sapiens 98-103 33107103-4 2021 Different isoforms of HMGB1 present with distinctive physiological functions in ECM-fully-reduced HMGB1 (all thiol) acts as the initial damage signal to recruit circulating myeloid cells, disulfide HMGB1 behaves as a cytokine to activate macrophages and neutrophils, and both signals are turned off when HMGB1 is terminally oxidized into the final sulfonate form. Alkanesulfonates 348-357 high mobility group box 1 Homo sapiens 22-27 33107103-4 2021 Different isoforms of HMGB1 present with distinctive physiological functions in ECM-fully-reduced HMGB1 (all thiol) acts as the initial damage signal to recruit circulating myeloid cells, disulfide HMGB1 behaves as a cytokine to activate macrophages and neutrophils, and both signals are turned off when HMGB1 is terminally oxidized into the final sulfonate form. Alkanesulfonates 348-357 high mobility group box 1 Homo sapiens 98-103 33107103-4 2021 Different isoforms of HMGB1 present with distinctive physiological functions in ECM-fully-reduced HMGB1 (all thiol) acts as the initial damage signal to recruit circulating myeloid cells, disulfide HMGB1 behaves as a cytokine to activate macrophages and neutrophils, and both signals are turned off when HMGB1 is terminally oxidized into the final sulfonate form. Alkanesulfonates 348-357 high mobility group box 1 Homo sapiens 98-103 33107103-4 2021 Different isoforms of HMGB1 present with distinctive physiological functions in ECM-fully-reduced HMGB1 (all thiol) acts as the initial damage signal to recruit circulating myeloid cells, disulfide HMGB1 behaves as a cytokine to activate macrophages and neutrophils, and both signals are turned off when HMGB1 is terminally oxidized into the final sulfonate form. Alkanesulfonates 348-357 high mobility group box 1 Homo sapiens 98-103 33107103-6 2021 Antagonists such as ethyl pyruvate inhibit HMGB1 by interfering with its cytoplasmic exportation, while others such as glycyrrhizin directly bind to HMGB1 and render it unavailable for its receptors. ethyl pyruvate 20-34 high mobility group box 1 Homo sapiens 43-48 33107103-6 2021 Antagonists such as ethyl pyruvate inhibit HMGB1 by interfering with its cytoplasmic exportation, while others such as glycyrrhizin directly bind to HMGB1 and render it unavailable for its receptors. Glycyrrhizic Acid 119-131 high mobility group box 1 Homo sapiens 149-154 33864964-0 2021 HMGB1 expression in leukocytes as a biomarker of cellular damage induced by [99mTc]Tc-HMPAO-labelling procedure: A quality control study. Technetium Tc 99m Exametazime 83-91 high mobility group box 1 Homo sapiens 0-5 33864964-11 2021 CONCLUSIONS: The evaluation of HMGB1 levels in WBC-CE from each subject after radiolabelling with [99mTc]Tc-HMPAO did not show significant changes compared to the cold cellular sample. Technetium Tc 99m Exametazime 105-113 high mobility group box 1 Homo sapiens 31-36 33946401-0 2021 Meta-Analysis of Methamphetamine Modulation on Amyloid Precursor Protein through HMGB1 in Alzheimer"s Disease. Methamphetamine 17-32 high mobility group box 1 Homo sapiens 81-86 33965862-6 2021 Furthermore, the HMGB1/p62/LC3II/LC3I and p53/SSAT/beta-catenin pathways were mainly involved in the inhibition of 13b-induced HCC metastasis by targeting polyamine transporters (PTs) overexpressed in HCC. CHEMBL3741121 115-118 high mobility group box 1 Homo sapiens 17-22 33946401-6 2021 Simulated METH exposure increased APP expression through HMGB1 at a confidence of p < 0.00001 (Z-score = 7.64, two-tailed). Methamphetamine 10-14 high mobility group box 1 Homo sapiens 57-62 33946401-9 2021 Our analyses suggest the involvement of HMGB1 signaling pathway in METH-induced modulation of APP as a potential casual factor of AD. Methamphetamine 67-71 high mobility group box 1 Homo sapiens 40-45 33871066-0 2021 Clinical application of heparin in the treatment of severe acute pancreatitis -- new discovery of the HMGB1 pathway. Heparin 24-31 high mobility group box 1 Homo sapiens 102-107 33903607-6 2021 Histological examination of the HMGB-1 treated group showed a vast area covered by lamellar and woven bone surrounding the beta-TCP granule remnants. beta-tricalcium phosphate 123-131 high mobility group box 1 Homo sapiens 32-38 33903887-0 2021 [Dexmedetomidine alleviates hepatic ischemia-reperfusion injury by regulating MALAT1/miR-126-5p/HMGB1 axis]. Dexmedetomidine 1-16 high mobility group box 1 Homo sapiens 96-101 33903887-11 2021 The results suggest that Dex protects hepatocytes from HIRI via regulating MALAT1/miR-126-5p/HMGB1 axis. Dexmedetomidine 25-28 high mobility group box 1 Homo sapiens 93-98 33617946-11 2021 CONCLUSION: G-Rg1 had a protective effect against LPS- and D-gal-induced ALF both in vitro and in vivo, which might be related to inhibited HMGB1-mediated TLR4-NF-kappaB Pathway. g-rg1 12-17 high mobility group box 1 Homo sapiens 140-145 33854044-3 2021 Using a phenotypic screening strategy with recombinant HMGB1 and peritoneal macrophages, we discovered that FeTPPS, a small molecule selectively inhibits HMGB1-mediated caspase-11 activation. fetpps 108-114 high mobility group box 1 Homo sapiens 55-60 33854044-3 2021 Using a phenotypic screening strategy with recombinant HMGB1 and peritoneal macrophages, we discovered that FeTPPS, a small molecule selectively inhibits HMGB1-mediated caspase-11 activation. fetpps 108-114 high mobility group box 1 Homo sapiens 154-159 33854044-5 2021 Treatment of FeTPPS significantly attenuates HMGB1- and caspase-11-mediated immune responses, organ damage, and lethality in endotoxemia and bacterial sepsis. fetpps 13-19 high mobility group box 1 Homo sapiens 45-50 33836651-13 2021 Compared with the C group, the levels of HMGB1, TNF-alpha, and IL-6 were significantly decreased 1 and 3 days after surgery in the QLB group (P < 0.05). N-Octyl(oxyethylene)(3)ethanol 131-134 high mobility group box 1 Homo sapiens 41-46 33618227-1 2021 To screen for specific transcription factors (TFs) that induce expression of the HMGB1 promoter in response to stimulation by Ang-II. ang-ii 126-132 high mobility group box 1 Homo sapiens 81-86 33618227-5 2021 However, compared with NC, invasion and migration by HMGB1 siRNA HCC cells stimulated by Ang-II were not altered; the expression of E-cadherin and vimentin was also unaltered. ang-ii 89-95 high mobility group box 1 Homo sapiens 53-58 33496921-8 2021 Moreover, the inhibitory effect of circ_0081146 knockdown on the progression of GC cells were reversed by silencing miR-144 or HMGB1 overexpression. circ_0081146 35-47 high mobility group box 1 Homo sapiens 127-132 33453094-0 2021 HMGB1 is a key factor for tamoxifen resistance and has the potential to predict the efficacy of CDK4/6 inhibitors in breast cancer. Tamoxifen 26-35 high mobility group box 1 Homo sapiens 0-5 33453094-9 2021 Moreover, we demonstrated that HMGB1 promoted tamoxifen resistance by combining with Toll-like receptor 4 (TLR4) and activating nuclear factor kappa B (NF-kappaB) pathway. Tamoxifen 46-55 high mobility group box 1 Homo sapiens 31-36 33453094-10 2021 CDK4/6 inhibitors could down-regulate the expression of HMGB1 and suppress TLR4-NF-kappaB pathway, in turn reverse tamoxifen resistance. Tamoxifen 115-124 high mobility group box 1 Homo sapiens 56-61 33453094-11 2021 These results illuminated the critical role of HMGB1 in the process of tamoxifen resistance, explained the mechanism of CDK4/6 inhibitors reversing tamoxifen resistance, and suggested the feasibility of HMGB1 serving as a potential biomarker for screening sensitive patients of CDK4/6 inhibitors. Tamoxifen 71-80 high mobility group box 1 Homo sapiens 47-52 33166075-4 2021 Animal and human studies have found cigarette smoke exposure upregulates RAGE expression, suggesting that the HMGB1-RAGE pathway might be involved in maternal nicotine-induced lung injury. Nicotine 159-167 high mobility group box 1 Homo sapiens 110-115 33660567-5 2021 Treatment with PF-43 prevented the downregulation of the epithelial barrier by HMGB1 and inflammatory cytokines in 2D culture. pf-43 15-20 high mobility group box 1 Homo sapiens 79-84 33660567-6 2021 Treatment with PF-43 prevented the epithelial hyperpermeability induced by HMGB1 and inflammatory cytokines in 2.5D culture. pf-43 15-20 high mobility group box 1 Homo sapiens 75-80 33660567-7 2021 In 2.5D culture, treatment with PF-43 inhibited the decreases of angulin-1/LSR, TRIC, pJNK, pAMPK and pMAPK induced by HMGB1 and the inflammatory cytokines. pf-43 32-37 high mobility group box 1 Homo sapiens 119-124 33617946-0 2021 Gensenoside Rg1 protects against lipopolysaccharide- and d-galactose-induced acute liver failure via suppressing HMGB1-mediated TLR4-NF-kappaB pathway. Galactose 57-68 high mobility group box 1 Homo sapiens 113-118 33617946-11 2021 CONCLUSION: G-Rg1 had a protective effect against LPS- and D-gal-induced ALF both in vitro and in vivo, which might be related to inhibited HMGB1-mediated TLR4-NF-kappaB Pathway. Galactose 59-64 high mobility group box 1 Homo sapiens 140-145 33691128-3 2021 propose that heparin provides protection during gram-negative sepsis by dampening harmful CASP11-dependent signaling through inhibition of HMGB1- and heparanase-mediated cytosolic delivery of LPS. Heparin 13-20 high mobility group box 1 Homo sapiens 139-144 33841665-6 2021 RESULTS: Ectopic HESCs showed markedly decreased cell viability with the increased release of HMGB-1 following treatment with H2O2. Hydrogen Peroxide 126-130 high mobility group box 1 Homo sapiens 94-100 33461096-0 2021 Reactive oxygen species induce Cys106-mediated anti-parallel HMGB1 dimerization that protects against DNA damage. Oxygen 9-15 high mobility group box 1 Homo sapiens 61-66 33461096-3 2021 As a redox-sensitive protein, HMGB1 contains three cysteine residues: Cys23, Cys45, and Cys106. Cysteine 51-59 high mobility group box 1 Homo sapiens 30-35 33461096-5 2021 HMGB1 dimerization was positively modulated by CuCl2 and H2O2. cupric chloride 47-52 high mobility group box 1 Homo sapiens 0-5 33461096-5 2021 HMGB1 dimerization was positively modulated by CuCl2 and H2O2. Hydrogen Peroxide 57-61 high mobility group box 1 Homo sapiens 0-5 33461096-7 2021 Treatment of HEK293T cells with CuCl2 and H2O2 enhanced the oxidative self-dimerization of HMGB1, whereas this dimerization was inhibited in mutant HMGB1C106A cells. cupric chloride 32-37 high mobility group box 1 Homo sapiens 91-96 33461096-7 2021 Treatment of HEK293T cells with CuCl2 and H2O2 enhanced the oxidative self-dimerization of HMGB1, whereas this dimerization was inhibited in mutant HMGB1C106A cells. cupric chloride 32-37 high mobility group box 1 Homo sapiens 148-153 33461096-7 2021 Treatment of HEK293T cells with CuCl2 and H2O2 enhanced the oxidative self-dimerization of HMGB1, whereas this dimerization was inhibited in mutant HMGB1C106A cells. Hydrogen Peroxide 42-46 high mobility group box 1 Homo sapiens 91-96 33729484-0 2021 miR-92d-3p suppresses the progression of diabetic nephropathy renal fibrosis by inhibiting the C3/HMGB1/TGF-beta1 pathway. mir-92d-3p 0-10 high mobility group box 1 Homo sapiens 98-103 33729484-9 2021 miR-92d-3p inhibited inflammatory factor production by C3 in the HK-2 cells and the activation of the C3/HMGB1/TGF-beta1 pathway and EMT by C3 and TGF-beta1. mir-92d-3p 0-10 high mobility group box 1 Homo sapiens 105-110 33729484-10 2021 miR-92d-3p suppressed the progression of DN renal fibrosis by inhibiting the activation of the C3/HMGB1/TGF-beta1 pathway and EMT. mir-92d-3p 0-10 high mobility group box 1 Homo sapiens 98-103 33731757-0 2021 Disulfide HMGB1 acts via TLR2/4 receptors to reduce the numbers of oligodendrocyte progenitor cells after traumatic injury in vitro. Disulfides 0-9 high mobility group box 1 Homo sapiens 10-15 33561388-5 2021 Heparin or a chemically modified form of heparin without anticoagulant function inhibited the alarmin HMGB1-lipopolysaccharide (LPS) interaction and prevented the macrophage glycocalyx degradation by heparanase. Heparin 0-7 high mobility group box 1 Homo sapiens 102-107 33561388-5 2021 Heparin or a chemically modified form of heparin without anticoagulant function inhibited the alarmin HMGB1-lipopolysaccharide (LPS) interaction and prevented the macrophage glycocalyx degradation by heparanase. Heparin 41-48 high mobility group box 1 Homo sapiens 102-107 33658363-5 2021 Further characterizing HMGB1 as a sialic acid-binding lectin, we found that optimal binding takes place at normal blood pH and is markedly reduced when pH is adjusted with lactic acid to levels found in sepsis. N-Acetylneuraminic Acid 34-45 high mobility group box 1 Homo sapiens 23-28 33500702-0 2021 miR-142-3p reduces the viability of human cervical cancer cells by negatively regulating the cytoplasmic localization of HMGB1. mir-142-3p 0-10 high mobility group box 1 Homo sapiens 121-126 33658363-5 2021 Further characterizing HMGB1 as a sialic acid-binding lectin, we found that optimal binding takes place at normal blood pH and is markedly reduced when pH is adjusted with lactic acid to levels found in sepsis. Lactic Acid 172-183 high mobility group box 1 Homo sapiens 23-28 33658363-6 2021 Glycan array studies confirmed the binding of HMGB1 to sialylated glycan sequences typically found on plasma glycoproteins, with binding again being dependent on zinc and normal blood pH. Polysaccharides 0-6 high mobility group box 1 Homo sapiens 46-51 33658363-6 2021 Glycan array studies confirmed the binding of HMGB1 to sialylated glycan sequences typically found on plasma glycoproteins, with binding again being dependent on zinc and normal blood pH. Polysaccharides 66-72 high mobility group box 1 Homo sapiens 46-51 33314408-0 2021 H19/miR-107 / HMGB1 axis sensitizes laryngeal squamous cell carcinoma to cisplatin by suppressing autophagy in vitro and in vivo. Cisplatin 73-82 high mobility group box 1 Homo sapiens 14-19 33500702-9 2021 Transfection with a miR-142-3p inhibitor increased cytoplasmic HMGB1 expression in HeLa cells, as shown by western blot analysis, while transfection with miR-142-3p mimics decreased the cytoplasmic expression of HMGB1 in HeLa cells. mir-142-3p 20-30 high mobility group box 1 Homo sapiens 63-68 33500702-9 2021 Transfection with a miR-142-3p inhibitor increased cytoplasmic HMGB1 expression in HeLa cells, as shown by western blot analysis, while transfection with miR-142-3p mimics decreased the cytoplasmic expression of HMGB1 in HeLa cells. mir-142-3p 20-30 high mobility group box 1 Homo sapiens 212-217 33500702-9 2021 Transfection with a miR-142-3p inhibitor increased cytoplasmic HMGB1 expression in HeLa cells, as shown by western blot analysis, while transfection with miR-142-3p mimics decreased the cytoplasmic expression of HMGB1 in HeLa cells. mir-142-3p 154-164 high mobility group box 1 Homo sapiens 212-217 33500702-10 2021 Therefore, miR-142-3p negatively regulated HMGB1 levels in cervical cancer cells. mir-142-3p 11-21 high mobility group box 1 Homo sapiens 43-48 33500702-11 2021 These findings indicated that miR-142-3p inhibited the proliferation of human cervical cancer cells, at least in part, by negatively regulating the cytoplasmic localization of HMGB1. mir-142-3p 30-40 high mobility group box 1 Homo sapiens 176-181 32426849-0 2021 Disulfide-HMGB1 Drives Ischemia-Reperfusion Injury in Human Liver Transplantation. Disulfides 0-9 high mobility group box 1 Homo sapiens 10-15 32426849-4 2021 Portal blood immediately following allograft reperfusion (liver flush, LF) had increased total HMGB1, but only LF from patients with histopathological IRI had increased disulfide-HMGB1 and induced TLR4-dependent TNFalpha production by macrophages. Disulfides 169-178 high mobility group box 1 Homo sapiens 179-184 32426849-5 2021 Disulfide HMGB1 levels increased concomitantly with IRI severity. Disulfides 0-9 high mobility group box 1 Homo sapiens 10-15 32426849-6 2021 IRI+ pre-reperfusion biopsies contained macrophages with hyperacetylated, lysosomal disulfide-HMGB1 that increased post-reperfusion at sites of injury, paralleling increased histone acetyltransferase GTF3C4 and decreased histone deacetylase HDAC5 expression. Disulfides 84-93 high mobility group box 1 Homo sapiens 94-99 32426849-7 2021 Purified disulfide-HMGB1 or IRI+ blood stimulated further production of disulfide-HMGB1 and increased pro-inflammatory molecule and cytokine expression in macrophages via a positive feedback loop. Disulfides 9-18 high mobility group box 1 Homo sapiens 19-24 32426849-7 2021 Purified disulfide-HMGB1 or IRI+ blood stimulated further production of disulfide-HMGB1 and increased pro-inflammatory molecule and cytokine expression in macrophages via a positive feedback loop. Disulfides 9-18 high mobility group box 1 Homo sapiens 82-87 32426849-7 2021 Purified disulfide-HMGB1 or IRI+ blood stimulated further production of disulfide-HMGB1 and increased pro-inflammatory molecule and cytokine expression in macrophages via a positive feedback loop. Disulfides 72-81 high mobility group box 1 Homo sapiens 19-24 32426849-7 2021 Purified disulfide-HMGB1 or IRI+ blood stimulated further production of disulfide-HMGB1 and increased pro-inflammatory molecule and cytokine expression in macrophages via a positive feedback loop. Disulfides 72-81 high mobility group box 1 Homo sapiens 82-87 32426849-8 2021 CONCLUSIONS: These data identify disulfide-HMGB1 as a mechanistic biomarker of, and therapeutic target for, minimizing sterile inflammation during human liver IRI. Disulfides 33-42 high mobility group box 1 Homo sapiens 43-48 33741691-4 2021 This nuclear retention of HMGB1 may also be a mediator of the anti-inflammatory effect of melatonin therapy in COVID-19-complementing melatonin"s suppression of nuclear factor kappa B activity and upregulation of nuclear factor erythroid 2-related factor 2. Melatonin 90-99 high mobility group box 1 Homo sapiens 26-31 33741691-4 2021 This nuclear retention of HMGB1 may also be a mediator of the anti-inflammatory effect of melatonin therapy in COVID-19-complementing melatonin"s suppression of nuclear factor kappa B activity and upregulation of nuclear factor erythroid 2-related factor 2. Melatonin 134-143 high mobility group box 1 Homo sapiens 26-31 33732708-7 2021 The higher cell viability observed in the HMGB1 knocked-down group after stimulation with H2O2 indicated the possible negative effect of HMGB1 on cellular lifespan. Hydrogen Peroxide 90-94 high mobility group box 1 Homo sapiens 42-47 33565572-0 2021 HMGB1 regulates ferroptosis through Nrf2 pathway in mesangial cells in response to high glucose. Glucose 88-95 high mobility group box 1 Homo sapiens 0-5 33565572-2 2021 The present study explored the role of high-mobility group box-1 (HMGB1) on the regulation of ferroptosis in mesangial cells in response to high glucose. Glucose 145-152 high mobility group box 1 Homo sapiens 39-64 33565572-2 2021 The present study explored the role of high-mobility group box-1 (HMGB1) on the regulation of ferroptosis in mesangial cells in response to high glucose. Glucose 145-152 high mobility group box 1 Homo sapiens 66-71 33565572-5 2021 Suppression of HMGB1 restored cellular proliferation, prevented ROS and LDH generation, decreased ACSL4, PTGS2, and NOX1, and increased GPX4 levels in mesangial cells. Reactive Oxygen Species 64-67 high mobility group box 1 Homo sapiens 15-20 33565572-6 2021 Furthermore, nuclear factor E2-related factor 2 (Nrf2) was decreased in DN patients and high glucose mediated translocation of HMGB1 in mesangial cells. Glucose 93-100 high mobility group box 1 Homo sapiens 127-132 33565572-7 2021 Knockdown of HMGB1 suppressed high glucose-induced activation of TLR4/NF-kappaB axis and promoted Nrf2 expression as well as its downstream targets including HO-1, NQO-1, GCLC and GCLM. Glucose 35-42 high mobility group box 1 Homo sapiens 13-18 33565572-8 2021 Collectively, these findings suggest that HMGB1 regulates glucose-induced ferroptosis via Nrf2 pathway in mesangial cells. Glucose 58-65 high mobility group box 1 Homo sapiens 42-47 33716687-6 2021 Galantamine treatment also recovered the AIE-increased expression of the proinflammatory receptors TLR4 and RAGE, the endogenous TLR4/RAGE agonist HMGB1, and the transcription activation marker pNF-kappaB p65. Galantamine 0-11 high mobility group box 1 Homo sapiens 147-152 33716687-10 2021 Together, these data suggest that AIE induces HMGB1-TLR4/RAGE-pNF-kappaB p65 signals, causing the loss of cholinergic phenotype (i.e., ChAT, TrkA, and p75NTR) through epigenetic histone transcription silencing that result in the loss of the BFCN phenotype that can be prevented and restored by galantamine. Galantamine 294-305 high mobility group box 1 Homo sapiens 46-51 33732708-7 2021 The higher cell viability observed in the HMGB1 knocked-down group after stimulation with H2O2 indicated the possible negative effect of HMGB1 on cellular lifespan. Hydrogen Peroxide 90-94 high mobility group box 1 Homo sapiens 137-142 33509758-0 2021 [Dihydromyricetin inhibits proliferation and migration of gastric cancer cells through regulating Akt/STAT3 signaling pathways and HMGB1 expression]. dihydromyricetin 1-17 high mobility group box 1 Homo sapiens 131-136 33624300-6 2021 We observed increased ACE2 expression with both HMGB1 and hyperglycemia treatment in A549 as well as H441 cells, which were blunted by TCBN treatment. triciribine 135-139 high mobility group box 1 Homo sapiens 48-53 33611821-7 2021 Depletion of microglia prior to ethanol treatment prevented pro-inflammatory activity of EtOH-MVs, as did incubation of EtOH-MVs with the HMGB1 inhibitor glycyrrhizin. Ethanol 120-124 high mobility group box 1 Homo sapiens 138-143 33611821-7 2021 Depletion of microglia prior to ethanol treatment prevented pro-inflammatory activity of EtOH-MVs, as did incubation of EtOH-MVs with the HMGB1 inhibitor glycyrrhizin. Glycyrrhizic Acid 154-166 high mobility group box 1 Homo sapiens 138-143 33611821-8 2021 Ethanol caused HMGB1 secretion from cultured BV2 microglia in MVs through activation of PI3 kinase. Ethanol 0-7 high mobility group box 1 Homo sapiens 15-20 33664952-4 2021 Vitamin D played a role in inhibiting HMGB1 secretion in the animal study. Vitamin D 0-9 high mobility group box 1 Homo sapiens 38-43 33664952-11 2021 Conclusions: Strong negative correlation between VDR and HMGB1 in different immunodeficiency statuses suggesting an important role of vitamin D in inflammation control in HIV infection. Vitamin D 134-143 high mobility group box 1 Homo sapiens 57-62 32374414-4 2021 Endothelial thrombomodulin (TM) and recombinant human soluble TM, TMalpha, dramatically promote thrombin-dependent degradation of HMGB1, and systemic administration of TMalpha prevents and reverses various HMGB1-dependent pathological pain. tmalpha 66-73 high mobility group box 1 Homo sapiens 130-135 32666385-0 2021 Involvement of HMGB1 in vemurafenib resistance in thyroid cancer cells harboring BRAF (V600E) mutation by regulating excessive autophagy. Vemurafenib 24-35 high mobility group box 1 Homo sapiens 15-20 32666385-3 2021 Here, we sought to elucidate the function of high mobility group box 1 (HMGB1) in vemurafenib resistance in thyroid cancer harboring BRAF mutation. Vemurafenib 82-93 high mobility group box 1 Homo sapiens 45-70 32666385-3 2021 Here, we sought to elucidate the function of high mobility group box 1 (HMGB1) in vemurafenib resistance in thyroid cancer harboring BRAF mutation. Vemurafenib 82-93 high mobility group box 1 Homo sapiens 72-77 32666385-5 2021 Then, BCPAP cells were transfected with recombinant HMGB1 plasmids, and vemurafenib-resistant BCPAP-R cells were treated with si-HMGB1. Vemurafenib 72-83 high mobility group box 1 Homo sapiens 129-134 32666385-6 2021 The efficacy of HMGB1 on vemurafenib resistance was evaluated by detecting cell viability, apoptosis, and caspase-3 activity. Vemurafenib 25-36 high mobility group box 1 Homo sapiens 16-21 32666385-8 2021 RESULTS: Lower expression of HMGB1 was observed in BRAF-mutant BCPAP cells that had high sensitivity to vemurafenib. Vemurafenib 104-115 high mobility group box 1 Homo sapiens 29-34 32666385-9 2021 Overexpression of HMGB1 attenuated BCPAP cell sensitivity to vemurafenib by increasing cell viability and decreasing cell apoptosis and caspase-3 activity. Vemurafenib 61-72 high mobility group box 1 Homo sapiens 18-23 32666385-10 2021 Intriguingly, higher expression of HMGB1 was confirmed in vemurafenib-resistant BCPAP-R cells. Vemurafenib 58-69 high mobility group box 1 Homo sapiens 35-40 32666385-11 2021 Moreover, knockdown of HMGB1 sensitized BCPAP-R cells to vemurafenib resistance. Vemurafenib 57-68 high mobility group box 1 Homo sapiens 23-28 32666385-13 2021 Importantly, targeting HMGB1 suppressed vemurafenib-induced autophagy. Vemurafenib 40-51 high mobility group box 1 Homo sapiens 23-28 32666385-15 2021 CONCLUSIONS: These findings highlight that HMGB1-mediated autophagy may account for vemurafenib resistance in thyroid cancer harboring BRAF mutation, implying a promising approach to overcome vemurafenib resistance in vemurafenib-mutant thyroid carcinomas. Vemurafenib 84-95 high mobility group box 1 Homo sapiens 43-48 32666385-15 2021 CONCLUSIONS: These findings highlight that HMGB1-mediated autophagy may account for vemurafenib resistance in thyroid cancer harboring BRAF mutation, implying a promising approach to overcome vemurafenib resistance in vemurafenib-mutant thyroid carcinomas. Vemurafenib 192-203 high mobility group box 1 Homo sapiens 43-48 32666385-15 2021 CONCLUSIONS: These findings highlight that HMGB1-mediated autophagy may account for vemurafenib resistance in thyroid cancer harboring BRAF mutation, implying a promising approach to overcome vemurafenib resistance in vemurafenib-mutant thyroid carcinomas. Vemurafenib 192-203 high mobility group box 1 Homo sapiens 43-48 33614649-0 2021 Temozolomide Treatment Induces HMGB1 to Promote the Formation of Glioma Stem Cells via the TLR2/NEAT1/Wnt Pathway in Glioblastoma. Temozolomide 0-12 high mobility group box 1 Homo sapiens 31-36 33614649-4 2021 Our study showed that TMZ treatment promoted GSCs formation by glioma cells; TMZ treatment of biopsy-derived glioblastoma multiforme cells upregulated HMGB1; HMGB1 altered gene expression profile of glioma cells with respect to mRNA, lncRNA and miRNA. Temozolomide 22-25 high mobility group box 1 Homo sapiens 158-163 33614649-4 2021 Our study showed that TMZ treatment promoted GSCs formation by glioma cells; TMZ treatment of biopsy-derived glioblastoma multiforme cells upregulated HMGB1; HMGB1 altered gene expression profile of glioma cells with respect to mRNA, lncRNA and miRNA. Temozolomide 77-80 high mobility group box 1 Homo sapiens 151-156 33614649-4 2021 Our study showed that TMZ treatment promoted GSCs formation by glioma cells; TMZ treatment of biopsy-derived glioblastoma multiforme cells upregulated HMGB1; HMGB1 altered gene expression profile of glioma cells with respect to mRNA, lncRNA and miRNA. Temozolomide 77-80 high mobility group box 1 Homo sapiens 158-163 33614649-5 2021 Furthermore, our results showed that TMZ-induced HMGB1 increased the formation of GSCs and when HMGB1 was downregulated, TMZ-mediated GSCs formation was attenuated. Temozolomide 37-40 high mobility group box 1 Homo sapiens 49-54 33614649-5 2021 Furthermore, our results showed that TMZ-induced HMGB1 increased the formation of GSCs and when HMGB1 was downregulated, TMZ-mediated GSCs formation was attenuated. Temozolomide 37-40 high mobility group box 1 Homo sapiens 96-101 33614649-5 2021 Furthermore, our results showed that TMZ-induced HMGB1 increased the formation of GSCs and when HMGB1 was downregulated, TMZ-mediated GSCs formation was attenuated. Temozolomide 121-124 high mobility group box 1 Homo sapiens 49-54 33614649-5 2021 Furthermore, our results showed that TMZ-induced HMGB1 increased the formation of GSCs and when HMGB1 was downregulated, TMZ-mediated GSCs formation was attenuated. Temozolomide 121-124 high mobility group box 1 Homo sapiens 96-101 33614649-8 2021 In conclusion, TMZ treatment upregulates HMGB1, which promotes the formation of GSCs via the TLR2/NEAT1/Wnt pathway. Temozolomide 15-18 high mobility group box 1 Homo sapiens 41-46 33614649-9 2021 Blocking HMGB1-mediated GSCs formation could serve as a potential therapeutic target for preventing TMZ resistance in GBM patients. Temozolomide 100-103 high mobility group box 1 Homo sapiens 9-14 33308900-0 2021 Astragaloside IV antagonizes M2 phenotype macrophage polarization-evoked ovarian cancer cell malignant progression by suppressing the HMGB1-TLR4 axis. astragaloside 0-13 high mobility group box 1 Homo sapiens 134-139 33308900-7 2021 During this process, administration with astragaloside IV restrained the high expression of high-mobility group box1 (HMGB1) and TLR4 in macrophages co-cultured with ovarian cancer cells, concomitant with decreases in release of M2 marker TGF-beta, MMP-9 and IL-10. astragaloside 41-54 high mobility group box 1 Homo sapiens 92-116 33308900-7 2021 During this process, administration with astragaloside IV restrained the high expression of high-mobility group box1 (HMGB1) and TLR4 in macrophages co-cultured with ovarian cancer cells, concomitant with decreases in release of M2 marker TGF-beta, MMP-9 and IL-10. astragaloside 41-54 high mobility group box 1 Homo sapiens 118-123 33308900-9 2021 Noticeably, exogenous HMGB1 overturned the inhibitory efficacy of astragaloside IV against macrophage M2 polarization-evoked malignant potential in ovarian cancer cells. astragaloside 66-79 high mobility group box 1 Homo sapiens 22-27 33308900-10 2021 Together, these findings suggest that astragaloside IV may protect against M2 macrophages-evoked malignancy in ovarian cancer cells by suppressing the HMGB1-TLR4 signaling. astragaloside 38-51 high mobility group box 1 Homo sapiens 151-156 33053467-0 2021 Amorphous silica nanoparticles induce inflammation via activation of NLRP3 inflammasome and HMGB1/TLR4/MYD88/NF-kb signaling pathway in HUVEC cells. Silicon Dioxide 10-16 high mobility group box 1 Homo sapiens 92-97 33053467-5 2021 In addition, SiO2 NPs were found to promote the translocation and release of high-mobility group box 1 (HMGB1) from nucleus to cytoplasm, which was demonstrated to be regulated by ROS and NOD-like receptor pyrin domain containing 3 (NLRP3) inflammasome. Reactive Oxygen Species 180-183 high mobility group box 1 Homo sapiens 77-102 33053467-5 2021 In addition, SiO2 NPs were found to promote the translocation and release of high-mobility group box 1 (HMGB1) from nucleus to cytoplasm, which was demonstrated to be regulated by ROS and NOD-like receptor pyrin domain containing 3 (NLRP3) inflammasome. Reactive Oxygen Species 180-183 high mobility group box 1 Homo sapiens 104-109 33559243-7 2021 Treatment of cholangiocytes with ethyl pyruvate suppressed the release of HMGB1. ethyl pyruvate 33-47 high mobility group box 1 Homo sapiens 74-79 33655086-8 2021 Second, curcumin protects from lethal pneumonia and ARDS via targeting NF-kappaB, inflammasome, IL-6 trans signal, and HMGB1 pathways. Curcumin 8-16 high mobility group box 1 Homo sapiens 119-124 33535837-8 2021 Furthermore, upregulation of high mobility group box protein 1 (HMGB1) protein levels and downregulation of miR-129-5p levels were observed in vivo and in vitro following sufentanil treatment. Sufentanil 171-181 high mobility group box 1 Homo sapiens 29-62 33535837-8 2021 Furthermore, upregulation of high mobility group box protein 1 (HMGB1) protein levels and downregulation of miR-129-5p levels were observed in vivo and in vitro following sufentanil treatment. Sufentanil 171-181 high mobility group box 1 Homo sapiens 64-69 33535837-11 2021 CONCLUSION: Sufentanil regulated the miR-129-5p/HMGB1 axis to enhance IkappaB-alpha expression, suggesting that sufentanil represents a candidate drug for ALI protection and providing avenues for clinical treatment. Sufentanil 12-22 high mobility group box 1 Homo sapiens 48-53 33535837-11 2021 CONCLUSION: Sufentanil regulated the miR-129-5p/HMGB1 axis to enhance IkappaB-alpha expression, suggesting that sufentanil represents a candidate drug for ALI protection and providing avenues for clinical treatment. Sufentanil 112-122 high mobility group box 1 Homo sapiens 48-53 32216030-0 2021 Adsorption kinetics for high mobility group box 1 (HMGB1) protein in a polyacrylonitrile hemofiltration membrane. polyacrylonitrile 71-88 high mobility group box 1 Homo sapiens 24-49 32216030-0 2021 Adsorption kinetics for high mobility group box 1 (HMGB1) protein in a polyacrylonitrile hemofiltration membrane. polyacrylonitrile 71-88 high mobility group box 1 Homo sapiens 51-56 32216030-2 2021 We previously reported that a polyacrylonitrile (AN69ST) membrane rapidly adsorbed HMGB1. polyacrylonitrile 30-47 high mobility group box 1 Homo sapiens 83-88 33515401-3 2021 Our study illustrates an example of drug repositioning of Biapenem (BIPM), a carbapenem antibiotic, for the modulation of HMGB1-induced septic responses. biapenem 58-66 high mobility group box 1 Homo sapiens 122-127 33515401-3 2021 Our study illustrates an example of drug repositioning of Biapenem (BIPM), a carbapenem antibiotic, for the modulation of HMGB1-induced septic responses. biapenem 68-72 high mobility group box 1 Homo sapiens 122-127 33515401-3 2021 Our study illustrates an example of drug repositioning of Biapenem (BIPM), a carbapenem antibiotic, for the modulation of HMGB1-induced septic responses. Carbapenems 77-87 high mobility group box 1 Homo sapiens 122-127 33509758-5 2021 Treatment with dihydromyricetin obviously suppressed the proliferation and migration of BGC-823 cells, significantly reduced the expression levels of cyclin D1, cyclin E1 and Ncadherin, enhanced E-cadherin expression, inhibited the phosphorylation of Akt and stat3, and downregulated HMGB1 expression in the cells. dihydromyricetin 15-31 high mobility group box 1 Homo sapiens 284-289 33509758-7 2021 CONCLUSIONS: Dihydromyricetin inhibits the proliferation and migration of BGC-823 cells through suppressing the activation of Akt/stat3 signaling pathways and HMGB1 expression. dihydromyricetin 13-29 high mobility group box 1 Homo sapiens 159-164 33440377-5 2021 Acrylonitrile 69-based oXiris membranes can remove endotoxin and high-mobility group box 1 protein. Acrylonitrile 0-13 high mobility group box 1 Homo sapiens 65-90 33573541-1 2021 Oleanolic acid can inhibit edema and exhibit obvious inhibitory activity to inflammatory by activating of the pituitary-adrenal cortical system, inhibiting the synthesis or release of PGs, inhibiting endotoxin-mediated release of HMGB1 by endothelial cells or regulating MAPK, PI3K/Akt/NF-kappaB/ICAM-1/JAK/STAT signaling pathways, etc. Oleanolic Acid 0-14 high mobility group box 1 Homo sapiens 230-235 33242463-5 2021 Mechanistically, tumor HIF1alpha signaling activated by chemo-induced ROS drives the transcription of HMGB1 to promote more macrophage infiltration into CRC tumor. ros 70-73 high mobility group box 1 Homo sapiens 102-107 33505216-10 2021 GA is capable of inhibiting toll-like receptors as well as high-mobility group box 1. Glycyrrhetinic Acid 0-2 high mobility group box 1 Homo sapiens 59-84 33422068-2 2021 Ring-stage parasite mRNA, measured by sbp1 quantitative reverse-transcriptase PCR (qRT-PCR), was previously reported to persist after ACT treatment and hypothesized to reflect temporary arrest of the growth of ring-stage parasites (dormancy) following exposure to artemisinins. Artemisinins 264-276 high mobility group box 1 Homo sapiens 38-42 32852122-9 2021 HMGB1 facilitated the proliferation of iNKT cells co-cultured with DCs and macrophages, which was found to be inhibited by heparin. Heparin 123-130 high mobility group box 1 Homo sapiens 0-5 33414419-0 2021 Gefitinib initiates sterile inflammation by promoting IL-1beta and HMGB1 release via two distinct mechanisms. Gefitinib 0-9 high mobility group box 1 Homo sapiens 67-72 33414419-3 2021 Here, we provide evidence that gefitinib elicits pro-inflammatory responses by promoting mature-interleukin-1beta (IL-1beta) and high-mobility group box 1 (HMGB1) release. Gefitinib 31-40 high mobility group box 1 Homo sapiens 129-154 33414419-3 2021 Here, we provide evidence that gefitinib elicits pro-inflammatory responses by promoting mature-interleukin-1beta (IL-1beta) and high-mobility group box 1 (HMGB1) release. Gefitinib 31-40 high mobility group box 1 Homo sapiens 156-161 33414419-5 2021 Notably, gefitinib also stimulated HMGB1 release, which is, however, not mediated by the NLRP3 inflammasome. Gefitinib 9-18 high mobility group box 1 Homo sapiens 35-40 33414419-6 2021 On the other hand, gefitinib-driven mtROS promoted the accumulation of gammaH2AX, a hallmark of DNA damage, leading to the activation of poly (ADP-ribose) polymerase-1 (PARP-1) and subsequent active release of HMGB1. Gefitinib 19-28 high mobility group box 1 Homo sapiens 210-215 33414419-7 2021 Together our results reveal the potential ability of gefitinib to initiate sterile inflammation via two distinct mechanisms, and identified IL-1beta and HMGB1 as key determinants of gefitinib-induced inflammation that may provide insights into gefitinib-induced interstitial pneumonitis. Gefitinib 182-191 high mobility group box 1 Homo sapiens 153-158 33414419-7 2021 Together our results reveal the potential ability of gefitinib to initiate sterile inflammation via two distinct mechanisms, and identified IL-1beta and HMGB1 as key determinants of gefitinib-induced inflammation that may provide insights into gefitinib-induced interstitial pneumonitis. Gefitinib 182-191 high mobility group box 1 Homo sapiens 153-158 33485134-13 2021 were analyzed for remission rates after first anthracycline based induction therapy, in those who did not experience a complete remission significantly enhanced HMGB1 surface expression was seen (98 vs. 94%; p < 0.05; n = 20). Anthracyclines 46-59 high mobility group box 1 Homo sapiens 161-166 32902022-0 2021 MiR-381-3p inhibits high glucose-induced vascular smooth muscle cell proliferation and migration by targeting HMGB1. mir-381-3p 0-10 high mobility group box 1 Homo sapiens 110-115 31888389-2 2021 Collismycin C inhibited the HMGB1 release and downregulated HMGB1-mediated inflammatory responses in human endothelial cells. collismycin A 0-13 high mobility group box 1 Homo sapiens 28-33 31888389-2 2021 Collismycin C inhibited the HMGB1 release and downregulated HMGB1-mediated inflammatory responses in human endothelial cells. collismycin A 0-13 high mobility group box 1 Homo sapiens 60-65 32902022-0 2021 MiR-381-3p inhibits high glucose-induced vascular smooth muscle cell proliferation and migration by targeting HMGB1. Glucose 25-32 high mobility group box 1 Homo sapiens 110-115 32902022-13 2021 CONCLUSION: MiR-381-3p targets HMGB1 to suppress the inflammation, oxidative stress, proliferation and migration of high-glucose-induced VSMCs by targeting HMGB1. Glucose 121-128 high mobility group box 1 Homo sapiens 31-36 32902022-13 2021 CONCLUSION: MiR-381-3p targets HMGB1 to suppress the inflammation, oxidative stress, proliferation and migration of high-glucose-induced VSMCs by targeting HMGB1. Glucose 121-128 high mobility group box 1 Homo sapiens 156-161 33347772-6 2021 Results: GLY protected against loss of cell viability induced by HG and significantly reduced HMGB1, IL-1beta, TLR2, TLR4, NLRP3, COX2, SOD2, HO-1, GPX2, and GR1. Glycyrrhizic Acid 9-12 high mobility group box 1 Homo sapiens 94-99 33128580-3 2021 Mechanistic investigation revealed that transforming growth factor beta-induced (TGFBI) acts as a potential downstream target of HMGB1 and lentivirus-mediated knockdown of TGFBI expression impaired phorbol-12-myristate-13-acetate (PMA) and all-trans retinoic acid (ATRA)-induced myeloid differentiation of AML cell lines. Tetradecanoylphorbol Acetate 198-229 high mobility group box 1 Homo sapiens 129-134 33128580-3 2021 Mechanistic investigation revealed that transforming growth factor beta-induced (TGFBI) acts as a potential downstream target of HMGB1 and lentivirus-mediated knockdown of TGFBI expression impaired phorbol-12-myristate-13-acetate (PMA) and all-trans retinoic acid (ATRA)-induced myeloid differentiation of AML cell lines. Tetradecanoylphorbol Acetate 231-234 high mobility group box 1 Homo sapiens 129-134 33128580-3 2021 Mechanistic investigation revealed that transforming growth factor beta-induced (TGFBI) acts as a potential downstream target of HMGB1 and lentivirus-mediated knockdown of TGFBI expression impaired phorbol-12-myristate-13-acetate (PMA) and all-trans retinoic acid (ATRA)-induced myeloid differentiation of AML cell lines. Tretinoin 240-263 high mobility group box 1 Homo sapiens 129-134 33128580-3 2021 Mechanistic investigation revealed that transforming growth factor beta-induced (TGFBI) acts as a potential downstream target of HMGB1 and lentivirus-mediated knockdown of TGFBI expression impaired phorbol-12-myristate-13-acetate (PMA) and all-trans retinoic acid (ATRA)-induced myeloid differentiation of AML cell lines. Tretinoin 265-269 high mobility group box 1 Homo sapiens 129-134 33128580-4 2021 On the other hand, chidamide, an orally histone deacetylase inhibitor, decreases HMGB1 expression significantly in AML cells with concomitant upregulation of TGFBI expression, and confers therapeutic effect on AML by inducing cell differentiation, apoptosis and inhibiting cell proliferation. N-(2-amino-5-fluorobenzyl)-4-(N-(pyridine-3-acrylyl)aminomethyl)benzamide 19-28 high mobility group box 1 Homo sapiens 81-86 33128580-5 2021 In conclusion, our findings provide additional insights that HMGB1 is a promising therapeutic target of AML, and also present experimental evidence for the clinical application of chidamide as a novel agent in AML therapy by downregulating HMGB1 expression. N-(2-amino-5-fluorobenzyl)-4-(N-(pyridine-3-acrylyl)aminomethyl)benzamide 180-189 high mobility group box 1 Homo sapiens 240-245 33128580-8 2021 Chidamide, a selective HDAC inhibitor, confers promising therapeutic effect for AML via downregulating HMGB1 expression. N-(2-amino-5-fluorobenzyl)-4-(N-(pyridine-3-acrylyl)aminomethyl)benzamide 0-9 high mobility group box 1 Homo sapiens 103-108 33030958-10 2021 We identified that HMGB1 secreted by breast cancer cells promotes fibroblast activation via RAGE/aerobic glycolysis, and activated fibroblasts enhance breast cancer cell metastasis through increased lactate. Lactic Acid 199-206 high mobility group box 1 Homo sapiens 19-24 33313438-7 2020 Importantly, genetic (using AGER siRNA) or pharmacological (using glycyrrhizin, chloroquine, hydroxychloroquine, and FPS-ZM1) inhibition of the HMGB1-AGER pathway blocks ACE2 expression. Glycyrrhizic Acid 66-78 high mobility group box 1 Homo sapiens 144-149 33420996-0 2021 Immunoaffinity-Based Liquid Chromatography Mass Spectrometric Assay to Accurately Quantify the Protein Concentration of HMGB1 in EDTA Plasma. Edetic Acid 129-133 high mobility group box 1 Homo sapiens 120-125 33086122-3 2020 The pharmacological activities essentially derive from the capacity of DMC to interact with the protein targets HMGB1 and AMPK. 2',4'-dihydroxy-6'-methoxy-3',5'-dimethylchalcone 71-74 high mobility group box 1 Homo sapiens 112-117 33086122-4 2020 Upon binding to HMGB1, DMC inhibits the nucleocytoplasmic transfer of the protein and its extracellular secretion, thereby blocking its alarmin function. 2',4'-dihydroxy-6'-methoxy-3',5'-dimethylchalcone 23-26 high mobility group box 1 Homo sapiens 16-21 33086122-6 2020 AMPK activation by DMC reinforces inhibition of HMGB1, to further reduce the release of the alarmin protein, likely contributing to the anticancer effects. 2',4'-dihydroxy-6'-methoxy-3',5'-dimethylchalcone 19-22 high mobility group box 1 Homo sapiens 48-53 33086122-7 2020 The characterization of a tight control of DMC over the AMPK-HMGB1 axis not only helps to explain the known activities of DMC but also suggests opportunities to use this chalcone to treat other pathological conditions such as the acute respiratory distress syndrome (which affects patients with COVID-19). 2',4'-dihydroxy-6'-methoxy-3',5'-dimethylchalcone 43-46 high mobility group box 1 Homo sapiens 61-66 33086122-7 2020 The characterization of a tight control of DMC over the AMPK-HMGB1 axis not only helps to explain the known activities of DMC but also suggests opportunities to use this chalcone to treat other pathological conditions such as the acute respiratory distress syndrome (which affects patients with COVID-19). 2',4'-dihydroxy-6'-methoxy-3',5'-dimethylchalcone 122-125 high mobility group box 1 Homo sapiens 61-66 33086122-7 2020 The characterization of a tight control of DMC over the AMPK-HMGB1 axis not only helps to explain the known activities of DMC but also suggests opportunities to use this chalcone to treat other pathological conditions such as the acute respiratory distress syndrome (which affects patients with COVID-19). Chalcone 170-178 high mobility group box 1 Homo sapiens 61-66 33380312-0 2020 Retraction Note to: Redox modification of cysteine residues regulates the cytokine activity of high mobility group box-1 (HMGB1). Cysteine 42-50 high mobility group box 1 Homo sapiens 95-120 33380312-0 2020 Retraction Note to: Redox modification of cysteine residues regulates the cytokine activity of high mobility group box-1 (HMGB1). Cysteine 42-50 high mobility group box 1 Homo sapiens 122-127 33321563-0 2021 Synergistic Tumoricidal Effects of Alpha-Lipoic Acid and Radiotherapy on Human Breast Cancer Cells Via HMGB1. Thioctic Acid 35-52 high mobility group box 1 Homo sapiens 103-108 33321563-8 2021 In addition, ALA significantly enhanced radiation-induced cellular senescence, which was shown by increased HMGB1 expression in the cytosol fraction compared to the control, increased p53 expression compared to the control, activation of p38 as well as NF-kappaB, and G2/M cell cycle arrest. Thioctic Acid 13-16 high mobility group box 1 Homo sapiens 108-113 33313438-7 2020 Importantly, genetic (using AGER siRNA) or pharmacological (using glycyrrhizin, chloroquine, hydroxychloroquine, and FPS-ZM1) inhibition of the HMGB1-AGER pathway blocks ACE2 expression. Chloroquine 80-91 high mobility group box 1 Homo sapiens 144-149 33313438-7 2020 Importantly, genetic (using AGER siRNA) or pharmacological (using glycyrrhizin, chloroquine, hydroxychloroquine, and FPS-ZM1) inhibition of the HMGB1-AGER pathway blocks ACE2 expression. Hydroxychloroquine 93-111 high mobility group box 1 Homo sapiens 144-149 33288764-4 2020 Here, we show that gemcitabine, a standard chemotherapy for various solid tumors, triggers hallmark immunostimualtory DAMP release (e.g., calreticulin, HSP70, and HMGB1); however, is unable to induce ICD. gemcitabine 19-30 high mobility group box 1 Homo sapiens 163-168 33287126-0 2020 Role of HMGB1/TLR4 Axis in Ischemia/Reperfusion-Impaired Extracellular Glutamate Clearance in Primary Astrocytes. Glutamic Acid 71-80 high mobility group box 1 Homo sapiens 8-13 33287126-4 2020 (2) Methods: We investigated the mechanism of the HMGB1/TLR4 axis in extracellular glutamate clearance in primary astrocytes exposed to ischemia/reperfusion by using OGD/R (oxygen-glucose deprivation/reoxygenation) model. Glutamic Acid 83-92 high mobility group box 1 Homo sapiens 50-55 33287126-5 2020 (3) Results: OGD/R insult activated the HMGB1/TLR4 axis for reducing the activity of glutamate clearance by inhibiting GLAST (glutamate aspartate transporter) expression in primary astrocytes. Glutamic Acid 85-94 high mobility group box 1 Homo sapiens 40-45 33287126-6 2020 Interestingly, OGD/R-untreated astrocytes showed impairment of glutamate clearance after exposure to exogenous HMGB1 or conditioned medium from OGD/R-treated astrocytes culture. Glutamic Acid 63-72 high mobility group box 1 Homo sapiens 111-116 33287126-7 2020 Inhibition of HMGB1 or TLR4 effectively prevented impaired glutamate clearance, which was induced by OGD/R, exogenous HMGB1, or conditioned medium from OGD/R-treated astrocytes. Glutamic Acid 59-68 high mobility group box 1 Homo sapiens 14-19 33287126-7 2020 Inhibition of HMGB1 or TLR4 effectively prevented impaired glutamate clearance, which was induced by OGD/R, exogenous HMGB1, or conditioned medium from OGD/R-treated astrocytes. Glutamic Acid 59-68 high mobility group box 1 Homo sapiens 118-123 33287126-8 2020 Furthermore, glycyrrhizic acid attenuated OGD/R-induced impairment of astrocytic glutamate clearance mediated by the HMGB1-TLR4 axis. Glycyrrhizic Acid 13-30 high mobility group box 1 Homo sapiens 117-122 33287126-8 2020 Furthermore, glycyrrhizic acid attenuated OGD/R-induced impairment of astrocytic glutamate clearance mediated by the HMGB1-TLR4 axis. Glutamic Acid 81-90 high mobility group box 1 Homo sapiens 117-122 33274007-0 2020 Protective Effect of miR-204 on Doxorubicin-Induced Cardiomyocyte Injury via HMGB1. Doxorubicin 32-43 high mobility group box 1 Homo sapiens 77-82 33012199-6 2020 Moreover, only human pulmonary artery smooth muscle cell death induced a release of dimeric HMGB1, found to be mitochondrial reactive oxygen species dependent, and TLR4 (toll-like receptor 4) activation. Reactive Oxygen Species 125-148 high mobility group box 1 Homo sapiens 92-97 33049495-8 2020 Juglanin also inhibits the inflammatory response by suppressing OSS-induced expressions of IL-1beta, MCP-1, and HMGB1. juglanin 0-8 high mobility group box 1 Homo sapiens 112-117 32998017-0 2020 Paclitaxel alleviates the sepsis-induced acute kidney injury via lnc-MALAT1/miR-370-3p/HMGB1 axis. Paclitaxel 0-10 high mobility group box 1 Homo sapiens 87-92 32998017-7 2020 What"s more, miR-370-3p targeted HMGB1 which was induced in serum and cells of sepsis. mir-370-3p 13-23 high mobility group box 1 Homo sapiens 33-38 32998017-8 2020 Knockdown of miR-370-3p inhibited the expression of HMGB1 and suppressed the proliferation but promoted the apoptosis of HK-2 cells injured by LPS as well as the expression of TNF-alpha, IL-6 and IL-1beta. mir-370-3p 13-23 high mobility group box 1 Homo sapiens 52-57 32998017-9 2020 Besides, paclitaxel restrained the expression of HMGB1 via regulating lnc-MALAT1/miR-370-3p axis. Paclitaxel 9-19 high mobility group box 1 Homo sapiens 49-54 33274007-13 2020 In summary, our study showed that miR-204 protected against DOX-induced cardiac injury via the inhibition of HMGB1, and increasing miR-204 expression may be a new treatment option for patients with DOX-induced cardiac injury. Doxorubicin 60-63 high mobility group box 1 Homo sapiens 109-114 33274007-13 2020 In summary, our study showed that miR-204 protected against DOX-induced cardiac injury via the inhibition of HMGB1, and increasing miR-204 expression may be a new treatment option for patients with DOX-induced cardiac injury. Doxorubicin 198-201 high mobility group box 1 Homo sapiens 109-114 33091813-0 2020 Paeonol promotes the phagocytic ability of macrophages through confining HMGB1 to the nucleus. paeonol 0-7 high mobility group box 1 Homo sapiens 73-78 33091813-5 2020 We found that paeonol could inhibit the translocation of HMGB1 from the nucleus to the cytoplasm, it may have an impact on phagocytosis. paeonol 14-21 high mobility group box 1 Homo sapiens 57-62 33091813-8 2020 Our results showed that paeonol promotes the phagocytosis of macrophages through confining HMGB1 to the nucleus. paeonol 24-31 high mobility group box 1 Homo sapiens 91-96 32998017-10 2020 SIGNIFICANCE: Paclitaxel could protect against LPS-induced AKI via the regulation of lnc-MALAT1/miR-370-3p/HMGB1 axis and the expression of TNF-alpha, IL-6 and IL-1beta, revealing that paclitaxel might act as a therapy drug in reducing sepsis-associated AKI. Paclitaxel 14-24 high mobility group box 1 Homo sapiens 107-112 33174035-10 2020 miR-143-3p overexpression attenuated the Der p1-induced inflammatory response and apoptosis of BEAS-2B cells by targeting high mobility group box 1 (HMGB1). mir-143-3p 0-10 high mobility group box 1 Homo sapiens 122-147 33174035-10 2020 miR-143-3p overexpression attenuated the Der p1-induced inflammatory response and apoptosis of BEAS-2B cells by targeting high mobility group box 1 (HMGB1). mir-143-3p 0-10 high mobility group box 1 Homo sapiens 149-154 33324614-0 2020 Discovery of 5,5"-Methylenedi-2,3-Cresotic Acid as a Potent Inhibitor of the Chemotactic Activity of the HMGB1 CXCL12 Heterocomplex Using Virtual Screening and NMR Validation. 5,5"-methylenedi-2,3-cresotic acid 13-47 high mobility group box 1 Homo sapiens 105-110 33324614-7 2020 We identified 5,5"-methylenedi-2,3-cresotic acid (2a) as a binder of both HMGB1 and CXCL12; 2a also targets the HMGB1 CXCL12 heterocomplex. 5,5"-methylenedi-2,3-cresotic acid 14-48 high mobility group box 1 Homo sapiens 74-79 33324614-7 2020 We identified 5,5"-methylenedi-2,3-cresotic acid (2a) as a binder of both HMGB1 and CXCL12; 2a also targets the HMGB1 CXCL12 heterocomplex. 5,5"-methylenedi-2,3-cresotic acid 14-48 high mobility group box 1 Homo sapiens 112-117 33182652-5 2020 EW-7197, AG-1478 and the TNFalpha antibody prevented hyperpermeability induced by HMGB1 and inflammatory cytokines in 2.5D culture. NSC334073 0-2 high mobility group box 1 Homo sapiens 82-87 32931793-13 2020 Inhibition of HMGB1 expression correlated with increased basal phospholipid production and SP-B expression in the lung lining. Phospholipids 63-75 high mobility group box 1 Homo sapiens 14-19 32898630-5 2020 In vitro studies showed that combination of laser assisted indocyanine green-mediated photothermal therapy and doxorubicin-mediated chemotherapy effectively eradicated cancer cells and resulted in the highest level of damage-associated molecular pattern presentation (calreticulin, high mobility group box 1, and adenosine triphosphate) compared to the individual treatments alone. Indocyanine Green 59-76 high mobility group box 1 Homo sapiens 282-307 32898630-5 2020 In vitro studies showed that combination of laser assisted indocyanine green-mediated photothermal therapy and doxorubicin-mediated chemotherapy effectively eradicated cancer cells and resulted in the highest level of damage-associated molecular pattern presentation (calreticulin, high mobility group box 1, and adenosine triphosphate) compared to the individual treatments alone. Doxorubicin 111-122 high mobility group box 1 Homo sapiens 282-307 33182652-7 2020 EW-7197, AG-1478 and the TNFalpha antibody prevented the increase in cell metabolism induced by HMGB1 and inflammatory cytokines in 2D culture. NSC334073 0-2 high mobility group box 1 Homo sapiens 96-101 32860758-2 2020 The high mobility group Box 1 (HMGB1) protein is a DNA-binding protein which exerts robust inflammatory actions, however, its role in ketoprofen-induced gastric damage has not been explored. Ketoprofen 134-144 high mobility group box 1 Homo sapiens 4-29 31504866-12 2020 We noted a negative correlation between HMGB1, NGF, Il-6, and lidocaine concentrations after surgery. Lidocaine 62-71 high mobility group box 1 Homo sapiens 40-45 33158052-0 2020 Morin Hydrate Sensitizes Hepatoma Cells and Xenograft Tumor towards Cisplatin by Downregulating PARP-1-HMGB1 Mediated Autophagy. morin 0-13 high mobility group box 1 Homo sapiens 103-108 33158052-6 2020 In addition, CP-induced PARP-1 activation led to ADP-ribosylation of HMGB1, which consequently developed autophagy as evident by the LC3I/II ratio. Adenosine Diphosphate 49-52 high mobility group box 1 Homo sapiens 69-74 33158052-8 2020 CP-Mh abrogates the PARP-1 expression and significantly reduced HMGB1-cytoplasmic translocation with subsequent inhibition of the HMGB1-Beclin1 complex formation. cp-mh 0-5 high mobility group box 1 Homo sapiens 64-69 33158052-8 2020 CP-Mh abrogates the PARP-1 expression and significantly reduced HMGB1-cytoplasmic translocation with subsequent inhibition of the HMGB1-Beclin1 complex formation. cp-mh 0-5 high mobility group box 1 Homo sapiens 130-135 33106070-8 2020 In addition, DHA decreased protein expression of TNF receptor, receptor interacting protein kinase 1, RIP3 and phosphorylation of mixed lineage kinase-like protein, phosphoglycerate mutase family 5, dynamin-related protein 1 and high mobility group box-1 protein. Docosahexaenoic Acids 13-16 high mobility group box 1 Homo sapiens 229-254 33455114-1 2020 PURPOSE: To explore the role of micro ribonucleic acid (miR)-218 in cervical cancer (CC) and the regulatory mechanism between the high mobility group box 1 (HMGB1)/receptor for advanced glycation end-product (RAGE) pathway and miR-218. micro ribonucleic acid 32-54 high mobility group box 1 Homo sapiens 157-162 33455114-8 2020 Moreover, miR-218 was observed to directly regulate the HMGB1/RAGE signaling pathway in a targeted manner to affect the proliferation and migration of CC cells. mir-218 10-17 high mobility group box 1 Homo sapiens 56-61 33455114-9 2020 CONCLUSIONS: MiR-218 inhibits the HMGB1/RAGE pathway to suppress the proliferation, migration and invasion of CC cells. mir-218 13-20 high mobility group box 1 Homo sapiens 34-39 33070867-0 2020 Retraction notice to "Molecular forms of HMGB1 and keratin-18 as mechanistic biomarkers for mode of cell death and prognosis during clinical acetaminophen hepatotoxicity": J Hepatol 56(2012)1070-1079. Acetaminophen 141-154 high mobility group box 1 Homo sapiens 41-46 33012373-0 2020 Retraction Notice to "Ketamine Reduces LPS-Induced HMGB1 Through HO-1 and Nrf2/ p38 MAPK" [Journal of Surgical Research 194 (2015) 599-613]. Ketamine 22-30 high mobility group box 1 Homo sapiens 51-56 32860758-2 2020 The high mobility group Box 1 (HMGB1) protein is a DNA-binding protein which exerts robust inflammatory actions, however, its role in ketoprofen-induced gastric damage has not been explored. Ketoprofen 134-144 high mobility group box 1 Homo sapiens 31-36 33017561-7 2021 Additionally, geldanamycin, an HSP90AA1 inhibitor, reduced HMGB1 secretion. geldanamycin 14-26 high mobility group box 1 Homo sapiens 59-64 33193406-3 2020 This study investigates whether increased HMGB1 levels are found in the intestinal mucosa of ulcerative colitis (UC) patients, and whether an anti-HMGB1 neutralizing-antibody (HnAb) can inhibit the intestinal inflammation elicited by dextran sulfate sodium (DSS) in mice. Dextran Sulfate 234-256 high mobility group box 1 Homo sapiens 147-152 33193406-3 2020 This study investigates whether increased HMGB1 levels are found in the intestinal mucosa of ulcerative colitis (UC) patients, and whether an anti-HMGB1 neutralizing-antibody (HnAb) can inhibit the intestinal inflammation elicited by dextran sulfate sodium (DSS) in mice. Dextran Sulfate 258-261 high mobility group box 1 Homo sapiens 147-152 33076532-0 2020 Nonhistone Proteins HMGB1 and HMGB2 Differentially Modulate the Response of Human Embryonic Stem Cells and the Progenitor Cells to the Anticancer Drug Etoposide. Etoposide 151-160 high mobility group box 1 Homo sapiens 20-25 33076532-1 2020 HMGB1 and HMGB2 proteins are abundantly expressed in human embryonic stem cells (hESCs) and hESC-derived progenitor cells (neuroectodermal cells, hNECs), though their functional roles in pluripotency and the mechanisms underlying their differentiation in response to the anticancer drug etoposide remain to be elucidated. Etoposide 287-296 high mobility group box 1 Homo sapiens 0-5 33076532-3 2020 The objective of this work was to determine whether HMGB1/2 proteins could modulate the sensitivity of hESCs and hESC-derived progenitor cells (hNECs) to etoposide. Etoposide 154-163 high mobility group box 1 Homo sapiens 52-59 33076532-4 2020 We observed that HMGB1 KD knockdown (KD) and, to a lesser extent, HMGB2 KD enhanced the sensitivity of hESCs to etoposide. Etoposide 112-121 high mobility group box 1 Homo sapiens 17-22 33076532-5 2020 Enhanced accumulation of 53BP1 on telomeres was detected by confocal microscopy in both untreated and etoposide-treated HMGB1 KD hESCs and hNECs, indicating that the loss of HMGB1 could destabilize telomeres. Etoposide 102-111 high mobility group box 1 Homo sapiens 120-125 33076532-5 2020 Enhanced accumulation of 53BP1 on telomeres was detected by confocal microscopy in both untreated and etoposide-treated HMGB1 KD hESCs and hNECs, indicating that the loss of HMGB1 could destabilize telomeres. Etoposide 102-111 high mobility group box 1 Homo sapiens 174-179 33076532-10 2020 Collectively, we have demonstrated that HMGB1 or HMGB2 differentially modulate the impact of etoposide treatment on human embryonic stem cells and their progenitor cells, suggesting possible strategies for the enhancement of the efficacy of this anticancer drug. Etoposide 93-102 high mobility group box 1 Homo sapiens 40-45 33056943-0 2020 Epigallocatechin Gallate (EGCG) Improves Anti-Angiogenic State, Cell Viability, and Hypoxia-Induced Endothelial Dysfunction by Downregulating High Mobility Group Box 1 (HMGB1) in Preeclampsia. epigallocatechin gallate 0-24 high mobility group box 1 Homo sapiens 142-167 33056943-0 2020 Epigallocatechin Gallate (EGCG) Improves Anti-Angiogenic State, Cell Viability, and Hypoxia-Induced Endothelial Dysfunction by Downregulating High Mobility Group Box 1 (HMGB1) in Preeclampsia. epigallocatechin gallate 0-24 high mobility group box 1 Homo sapiens 169-174 33056943-0 2020 Epigallocatechin Gallate (EGCG) Improves Anti-Angiogenic State, Cell Viability, and Hypoxia-Induced Endothelial Dysfunction by Downregulating High Mobility Group Box 1 (HMGB1) in Preeclampsia. epigallocatechin gallate 26-30 high mobility group box 1 Homo sapiens 142-167 33056943-0 2020 Epigallocatechin Gallate (EGCG) Improves Anti-Angiogenic State, Cell Viability, and Hypoxia-Induced Endothelial Dysfunction by Downregulating High Mobility Group Box 1 (HMGB1) in Preeclampsia. epigallocatechin gallate 26-30 high mobility group box 1 Homo sapiens 169-174 33056943-2 2020 This study assessed whether epigallocatechin gallate (EGCG) could reduce the production of anti-angiogenic factors, improve cell viability, and suppress endothelial dysfunction in vitro via regulating high mobility group box 1 (HMGB1) in preeclampsia. epigallocatechin gallate 28-52 high mobility group box 1 Homo sapiens 201-226 33056943-2 2020 This study assessed whether epigallocatechin gallate (EGCG) could reduce the production of anti-angiogenic factors, improve cell viability, and suppress endothelial dysfunction in vitro via regulating high mobility group box 1 (HMGB1) in preeclampsia. epigallocatechin gallate 28-52 high mobility group box 1 Homo sapiens 228-233 33056943-2 2020 This study assessed whether epigallocatechin gallate (EGCG) could reduce the production of anti-angiogenic factors, improve cell viability, and suppress endothelial dysfunction in vitro via regulating high mobility group box 1 (HMGB1) in preeclampsia. epigallocatechin gallate 54-58 high mobility group box 1 Homo sapiens 201-226 33056943-2 2020 This study assessed whether epigallocatechin gallate (EGCG) could reduce the production of anti-angiogenic factors, improve cell viability, and suppress endothelial dysfunction in vitro via regulating high mobility group box 1 (HMGB1) in preeclampsia. epigallocatechin gallate 54-58 high mobility group box 1 Homo sapiens 228-233 33056943-5 2020 RESULTS EGCG inhibited HMGB1 expression in hypoxic trophoblast cells in a dose-dependent manner. epigallocatechin gallate 8-12 high mobility group box 1 Homo sapiens 23-28 33056943-6 2020 In addition, EGCG relieved anti-angiogenic state and endothelial dysfunction in hypoxic trophoblast cells by downregulating HMGB1. epigallocatechin gallate 13-17 high mobility group box 1 Homo sapiens 124-129 33056943-7 2020 Moreover, EGCG dose-dependently promoted cell proliferation by downregulating HMGB1. epigallocatechin gallate 10-14 high mobility group box 1 Homo sapiens 78-83 33056943-8 2020 CONCLUSIONS Taken together, our data show the protective role of EGCG in preeclampsia and revealed EGCG-mediated effects on the production of anti-angiogenic factors, cell viability, and endothelial dysfunction through downregulating HMGB1. epigallocatechin gallate 99-103 high mobility group box 1 Homo sapiens 234-239 32999071-4 2020 We previously demonstrated that, upon asbestos exposure, HM and reactive macrophages releases the high mobility group box 1 (HMGB1) protein that becomes detectable in the tissues near asbestos deposits where HMGB1 triggers chronic inflammation. Asbestos 38-46 high mobility group box 1 Homo sapiens 125-130 32999071-4 2020 We previously demonstrated that, upon asbestos exposure, HM and reactive macrophages releases the high mobility group box 1 (HMGB1) protein that becomes detectable in the tissues near asbestos deposits where HMGB1 triggers chronic inflammation. Asbestos 38-46 high mobility group box 1 Homo sapiens 208-213 32999071-4 2020 We previously demonstrated that, upon asbestos exposure, HM and reactive macrophages releases the high mobility group box 1 (HMGB1) protein that becomes detectable in the tissues near asbestos deposits where HMGB1 triggers chronic inflammation. Asbestos 184-192 high mobility group box 1 Homo sapiens 125-130 32999071-4 2020 We previously demonstrated that, upon asbestos exposure, HM and reactive macrophages releases the high mobility group box 1 (HMGB1) protein that becomes detectable in the tissues near asbestos deposits where HMGB1 triggers chronic inflammation. Asbestos 184-192 high mobility group box 1 Homo sapiens 208-213 33023630-8 2020 RESULTS: DEX, but not IgG, significantly inhibited apoptosis caused by inflammatory stimuli, resulting in effective reduction of HMGB1 and IL-1alpha protein release by HCAECs. Dexamethasone 9-12 high mobility group box 1 Homo sapiens 129-134 33017561-11 2021 The multivesicular body formation inhibitor GW4869 dramatically decreased HMGB1 secretion under HMGB1 secretion-inducing conditions. GW 4869 44-50 high mobility group box 1 Homo sapiens 74-79 33017561-11 2021 The multivesicular body formation inhibitor GW4869 dramatically decreased HMGB1 secretion under HMGB1 secretion-inducing conditions. GW 4869 44-50 high mobility group box 1 Homo sapiens 96-101 32436353-0 2020 Hypoxia-responsive miR-141-3p is involved in the progression of breast cancer via mediating HMGB1/HIF-1alpha signaling pathway. mir-141-3p 19-29 high mobility group box 1 Homo sapiens 92-97 32388677-4 2020 Mechanistically, in the GC cells under the 5-FU treatment, reactive oxygen species is accumulated and then induces the activation of HIF1alpha signaling to drive the expression of high-mobility group box 1, which leads to more macrophage"s infiltration into GC tumor. Fluorouracil 43-47 high mobility group box 1 Homo sapiens 180-205 32388677-4 2020 Mechanistically, in the GC cells under the 5-FU treatment, reactive oxygen species is accumulated and then induces the activation of HIF1alpha signaling to drive the expression of high-mobility group box 1, which leads to more macrophage"s infiltration into GC tumor. Oxygen 68-74 high mobility group box 1 Homo sapiens 180-205 32623356-0 2020 N-glycosylation of High Mobility Group Box 1 protein (HMGB1) modulates the interaction with glycyrrhizin: A molecular modeling study. Glycyrrhizic Acid 92-104 high mobility group box 1 Homo sapiens 54-59 32623356-5 2020 In cells, HMGB1 is N-glycosylated at three asparagine residues located in boxes A and B, and these N-glycans are essential for the nucleocytoplasmic transport of the protein. Nitrogen 19-20 high mobility group box 1 Homo sapiens 10-15 32623356-5 2020 In cells, HMGB1 is N-glycosylated at three asparagine residues located in boxes A and B, and these N-glycans are essential for the nucleocytoplasmic transport of the protein. Asparagine 43-53 high mobility group box 1 Homo sapiens 10-15 32623356-5 2020 In cells, HMGB1 is N-glycosylated at three asparagine residues located in boxes A and B, and these N-glycans are essential for the nucleocytoplasmic transport of the protein. n-glycans 99-108 high mobility group box 1 Homo sapiens 10-15 32623356-7 2020 Here we have investigated the effect of the N-glycosylation of HMGB1 on its interaction with GLR using molecular modelling, after incorporation of three N-glycans on a Human HMGB1 structure (PDB code 2YRQ). Nitrogen 44-45 high mobility group box 1 Homo sapiens 63-68 32623356-7 2020 Here we have investigated the effect of the N-glycosylation of HMGB1 on its interaction with GLR using molecular modelling, after incorporation of three N-glycans on a Human HMGB1 structure (PDB code 2YRQ). n-glycans 153-162 high mobility group box 1 Homo sapiens 63-68 32623356-13 2020 The effects of the N-glycans are mostly indirect, but in one case a direct contact with the drug, via a carbohydrate-carbohydrate interaction, was observed with 18beta-GLR bound to Box-B of glycosylated HMGB1. n-glycans 19-28 high mobility group box 1 Homo sapiens 203-208 32623356-13 2020 The effects of the N-glycans are mostly indirect, but in one case a direct contact with the drug, via a carbohydrate-carbohydrate interaction, was observed with 18beta-GLR bound to Box-B of glycosylated HMGB1. Carbohydrates 104-116 high mobility group box 1 Homo sapiens 203-208 32623356-13 2020 The effects of the N-glycans are mostly indirect, but in one case a direct contact with the drug, via a carbohydrate-carbohydrate interaction, was observed with 18beta-GLR bound to Box-B of glycosylated HMGB1. Carbohydrates 117-129 high mobility group box 1 Homo sapiens 203-208 32623356-14 2020 For the first time, it is shown (at least in silico) that N-glycosylation, one of the many post-translational modifications of HMGB1, can affect drug binding. Nitrogen 58-59 high mobility group box 1 Homo sapiens 127-132 32623356-0 2020 N-glycosylation of High Mobility Group Box 1 protein (HMGB1) modulates the interaction with glycyrrhizin: A molecular modeling study. Nitrogen 0-1 high mobility group box 1 Homo sapiens 19-44 32623356-0 2020 N-glycosylation of High Mobility Group Box 1 protein (HMGB1) modulates the interaction with glycyrrhizin: A molecular modeling study. Nitrogen 0-1 high mobility group box 1 Homo sapiens 54-59 32623356-0 2020 N-glycosylation of High Mobility Group Box 1 protein (HMGB1) modulates the interaction with glycyrrhizin: A molecular modeling study. Glycyrrhizic Acid 92-104 high mobility group box 1 Homo sapiens 19-44 32945389-7 2020 The results of the present study demonstrated that the expression levels of circ_0010283 and HMGB1 were significantly upregulated in ox-LDL-induced VSMCs compared with those in VSMCs without ox-LDL induction, whereas the expression of miR-370-3p was downregulated. mir-370-3p 235-245 high mobility group box 1 Homo sapiens 93-98 32945389-9 2020 In addition, miR-370-3p was demonstrated to be a target of circ_0010283 and to target HMGB1; thus, circ_0010283 regulated HMGB1 expression via miR-370-3p. mir-370-3p 13-23 high mobility group box 1 Homo sapiens 86-91 32945389-9 2020 In addition, miR-370-3p was demonstrated to be a target of circ_0010283 and to target HMGB1; thus, circ_0010283 regulated HMGB1 expression via miR-370-3p. mir-370-3p 13-23 high mobility group box 1 Homo sapiens 122-127 32945389-11 2020 Additionally, the miR-370-3p-mediated suppressive effects on cell viability and migration were rescued by overexpression of HMGB1. mir-370-3p 18-28 high mobility group box 1 Homo sapiens 124-129 32436353-2 2020 The present study was aimed to investigate whether hypoxia-responsive miR-141-3p was implicated in the pathogenesis of breast cancer via mediating HMGB1/HIF-1alpha signaling pathway. mir-141-3p 70-80 high mobility group box 1 Homo sapiens 147-152 32436353-10 2020 Mechanistically, hypoxia-related HMGB1/HIF-1alpha signaling pathway might be a possible target of miR-141-3p to prevent the development of breast cancer. mir-141-3p 98-108 high mobility group box 1 Homo sapiens 33-38 32436353-11 2020 CONCLUSIONS: Our finding provides a new mechanism that miR-141-3p could prevent hypoxia-induced breast tumorigenesis by post-transcriptional repression of HMGB1/HIF-1alpha signaling pathway. mir-141-3p 55-65 high mobility group box 1 Homo sapiens 155-160 31872339-0 2020 Glycyrrhizin Prevents Hemorrhagic Transformation and Improves Neurological Outcome in Ischemic Stroke with Delayed Thrombolysis Through Targeting Peroxynitrite-Mediated HMGB1 Signaling. Glycyrrhizic Acid 0-12 high mobility group box 1 Homo sapiens 169-174 32857676-5 2020 Pretreatment with EW-7197 prevented the changes of all tight junction molecules induced by HMGB1. vactosertib 18-25 high mobility group box 1 Homo sapiens 91-96 32857676-6 2020 In 2.5D Matrigel culture, treatment with HMGB1 induced permeability of FITC-dextran (FD-4) into the lumen, whereas pretreatment with EW-7197 prevented the hyperpermeability of FD-4 into the lumen caused by HMGB1. fluorescein isothiocyanate dextran 71-83 high mobility group box 1 Homo sapiens 41-46 32857676-6 2020 In 2.5D Matrigel culture, treatment with HMGB1 induced permeability of FITC-dextran (FD-4) into the lumen, whereas pretreatment with EW-7197 prevented the hyperpermeability of FD-4 into the lumen caused by HMGB1. vactosertib 133-140 high mobility group box 1 Homo sapiens 206-211 31872339-0 2020 Glycyrrhizin Prevents Hemorrhagic Transformation and Improves Neurological Outcome in Ischemic Stroke with Delayed Thrombolysis Through Targeting Peroxynitrite-Mediated HMGB1 Signaling. Peroxynitrous Acid 146-159 high mobility group box 1 Homo sapiens 169-174 31872339-12 2020 In conclusion, peroxynitrite-mediated HMGB1/TLR2 signaling contributes to hemorrhagic transformation, and glycyrrhizin could be a potential adjuvant therapy to attenuate hemorrhagic transformation, possibly through inhibiting the ONOO-/HMGB1/TLR2 signaling cascades. Peroxynitrous Acid 15-28 high mobility group box 1 Homo sapiens 38-43 31872339-12 2020 In conclusion, peroxynitrite-mediated HMGB1/TLR2 signaling contributes to hemorrhagic transformation, and glycyrrhizin could be a potential adjuvant therapy to attenuate hemorrhagic transformation, possibly through inhibiting the ONOO-/HMGB1/TLR2 signaling cascades. Peroxynitrous Acid 15-28 high mobility group box 1 Homo sapiens 236-241 31872339-2 2020 However, the roles of ONOO- in mediating HMGB1 expression and its impacts on hemorrhagic transformation (HT) in ischemic brain injury with delayed t-PA treatment remain unclear. tetramethylammonium peroxynitrite 22-26 high mobility group box 1 Homo sapiens 41-46 31872339-12 2020 In conclusion, peroxynitrite-mediated HMGB1/TLR2 signaling contributes to hemorrhagic transformation, and glycyrrhizin could be a potential adjuvant therapy to attenuate hemorrhagic transformation, possibly through inhibiting the ONOO-/HMGB1/TLR2 signaling cascades. Glycyrrhizic Acid 106-118 high mobility group box 1 Homo sapiens 236-241 31872339-3 2020 In the present study, we tested the hypothesis that ONOO- could directly mediate the activation and release of HMGB1 in ischemic brains with delayed t-PA treatment. tetramethylammonium peroxynitrite 52-56 high mobility group box 1 Homo sapiens 111-116 31872339-4 2020 With clinical studies, we found that plasma nitrotyrosine (NT, a surrogate marker of ONOO-) was positively correlated with HMGB1 level in acute ischemic stroke patients. Nitrotyrosine 44-57 high mobility group box 1 Homo sapiens 123-128 32446815-4 2020 The role of HMGB1 in DNA damage in an aflatoxin-induced lung inflammatory environment was investigated in this study. Aflatoxins 38-47 high mobility group box 1 Homo sapiens 12-17 31872339-4 2020 With clinical studies, we found that plasma nitrotyrosine (NT, a surrogate marker of ONOO-) was positively correlated with HMGB1 level in acute ischemic stroke patients. nt 59-61 high mobility group box 1 Homo sapiens 123-128 31872339-4 2020 With clinical studies, we found that plasma nitrotyrosine (NT, a surrogate marker of ONOO-) was positively correlated with HMGB1 level in acute ischemic stroke patients. tetramethylammonium peroxynitrite 85-89 high mobility group box 1 Homo sapiens 123-128 31872339-7 2020 ONOO- donor SIN-1 directly induced expression of HMGB1 and its receptor TLR2 in naive rat brains in vivo and induced HMGB1 in brain microvascular endothelial b.End3 cells in vitro. tetramethylammonium peroxynitrite 0-4 high mobility group box 1 Homo sapiens 49-54 31872339-8 2020 Those results suggest that ONOO- could activate HMGB1/TLR2/MMP-9 signaling. tetramethylammonium peroxynitrite 27-31 high mobility group box 1 Homo sapiens 48-53 31872339-9 2020 We then addressed whether glycyrrhizin, a natural HMGB1 inhibitor, could inhibit ONOO- production and the antioxidant properties of glycyrrhizin contribute to the inhibition of HMGB1 and the neuroprotective effects on attenuating hemorrhagic transformation in ischemic stroke with delayed t-PA treatment. Glycyrrhizic Acid 26-38 high mobility group box 1 Homo sapiens 50-55 31872339-9 2020 We then addressed whether glycyrrhizin, a natural HMGB1 inhibitor, could inhibit ONOO- production and the antioxidant properties of glycyrrhizin contribute to the inhibition of HMGB1 and the neuroprotective effects on attenuating hemorrhagic transformation in ischemic stroke with delayed t-PA treatment. Glycyrrhizic Acid 26-38 high mobility group box 1 Homo sapiens 177-182 31872339-10 2020 Glycyrrhizin treatment downregulated the expressions of NADPH oxidase p47 phox and p67 phox and iNOS, inhibited superoxide and ONOO- production, reduced the expression of HMGB1, TLR2, MMP-9, preserved type IV collagen and claudin-5 in ischemic brains. Glycyrrhizic Acid 0-12 high mobility group box 1 Homo sapiens 171-176 32963505-5 2020 TNF-alpha-treated human periodontal ligament stem cell (hPDLSC) model was established, and was administrated with 1, 2 or 5 mM glycyrrhizin for 24 h. After treatment, the expression of HMGB1and inflammatory cytokines was monitored. Glycyrrhizic Acid 127-139 high mobility group box 1 Homo sapiens 185-190 32553625-0 2020 Dimethyl fumarate prevents osteoclastogenesis by decreasing NFATc1 expression, inhibiting of erk and p38 MAPK phosphorylation, and suppressing of HMGB1 release. Dimethyl Fumarate 0-17 high mobility group box 1 Homo sapiens 146-151 32553625-8 2020 We also found that DMF inhibited the extracellular release of high mobility group box 1, associated with an up-regulation of heme oxygenase-1, likely mediated through Nrf2 activation. Dimethyl Fumarate 19-22 high mobility group box 1 Homo sapiens 62-87 32963505-7 2020 Glycyrrhizin significantly prevented TNF-alpha-induced expression of HMGB1 (691.5 +- 136.4 vs 142.8 +- 57.3 pg/mL), IL-6 (388.1 +- 85.2 vs 189.4 +- 61.2 pg/mL) and IL-1beta (176.3 +- 47.2 vs 53.9 +- 25.7 pg/mL) in hPDLSC. Glycyrrhizic Acid 0-12 high mobility group box 1 Homo sapiens 69-74 32954194-5 2020 We also found that ropivacaine could inhibit the secretion of the high-mobility group box 1 protein and improve cell viability. Ropivacaine 19-30 high mobility group box 1 Homo sapiens 66-91 33013386-14 2020 In conclusion, we showed for the first time a hepatoprotective effect for AU in liver IRI, which acted by inhibiting the HMGB1/TLR-4/NF-kappaB signaling pathway, oxidative stress, and apoptosis. aucubin 74-76 high mobility group box 1 Homo sapiens 121-126 32320818-6 2020 CONCLUSION: LncRNA ZFAS1 could bind to miR-129 to promote HMGB1 expression, thereby affecting the endocrine disturbance, proliferation and apoptosis of ovarian granulosa cells in PCOS. mir-129 39-46 high mobility group box 1 Homo sapiens 58-63 32833553-8 2020 Knockdown or block of HMGB1 relieved the CCl4-induced liver fibrosis. Carbon Tetrachloride 41-45 high mobility group box 1 Homo sapiens 22-27 32497617-2 2020 HMGB1 is a danger associated protein pattern receptor which can sense high glucose as a stressor. Glucose 75-82 high mobility group box 1 Homo sapiens 0-5 32437849-3 2020 RGG-motif proteins Scd6 and Sbp1 are translation-repressors and decapping-activators that localize to and affect the assembly of RNA granules in response to sodium azide stress. Sodium Azide 157-169 high mobility group box 1 Homo sapiens 28-32 32874136-8 2020 Additionally, in both the rabbit trauma model and cellular injury model, CAA combined with high-dosage of VB6 inhibited the expression of inflammatory factors (IL-6, TNF-alpha and IL-1beta) and proteins (HMGB1, TLR4 and p-p65) in HMGB1/TLR4/NF-kappaB pathway. caa 73-76 high mobility group box 1 Homo sapiens 204-209 32771683-6 2020 High Mobility Group Box 1 (HMGB1) is a mediator of lethality in trauma and sepsis and our mechanistic studies revealed that disulfide and oxidized forms of HMGB1 bind to the gp91phox subunit of NOX2, and thus decrease the neutrophil respiratory burst and bacterial killing. Disulfides 124-133 high mobility group box 1 Homo sapiens 0-25 32771683-6 2020 High Mobility Group Box 1 (HMGB1) is a mediator of lethality in trauma and sepsis and our mechanistic studies revealed that disulfide and oxidized forms of HMGB1 bind to the gp91phox subunit of NOX2, and thus decrease the neutrophil respiratory burst and bacterial killing. Disulfides 124-133 high mobility group box 1 Homo sapiens 27-32 32874136-0 2020 Compound amino acid combined with high-dose vitamin B6 attenuate traumatic coagulopathy via inhibiting inflammation by HMGB1/TLR4/NF-kappaB pathway. Vitamin B 6 44-54 high mobility group box 1 Homo sapiens 119-124 32553931-0 2020 Magnoflorine inhibits the malignant phenotypes and increases cisplatin sensitivity of osteosarcoma cells via regulating miR-410-3p/HMGB1/NF-kappaB pathway. magnoflorine 0-12 high mobility group box 1 Homo sapiens 131-136 32553931-8 2020 To explore the potential mechanism, we assayed the influence of magnoflorine on the miR-410-3p/HMGB1/NF-kappaB signaling pathway. magnoflorine 64-76 high mobility group box 1 Homo sapiens 95-100 32553931-8 2020 To explore the potential mechanism, we assayed the influence of magnoflorine on the miR-410-3p/HMGB1/NF-kappaB signaling pathway. mir-410 84-91 high mobility group box 1 Homo sapiens 95-100 32553931-12 2020 Magnoflorine significantly suppressed HMGB1 expression and NF-kappaB activation, but upregulated miR-410-3p level. magnoflorine 0-12 high mobility group box 1 Homo sapiens 38-43 32553931-13 2020 Overexpression of HMGB1 promoted NF-kappaB activation and reversed the effects of magnoflorine on the viability, invasion, EMT and cisplatin sensitivity of OS cells. magnoflorine 82-94 high mobility group box 1 Homo sapiens 18-23 32553931-13 2020 Overexpression of HMGB1 promoted NF-kappaB activation and reversed the effects of magnoflorine on the viability, invasion, EMT and cisplatin sensitivity of OS cells. Cisplatin 131-140 high mobility group box 1 Homo sapiens 18-23 32553931-15 2020 And miR-410-3p inhibitor abrogated the influence of magnoflorine on HMGB1 expression in OS cells. mir-410-3p 4-14 high mobility group box 1 Homo sapiens 68-73 32553931-15 2020 And miR-410-3p inhibitor abrogated the influence of magnoflorine on HMGB1 expression in OS cells. magnoflorine 52-64 high mobility group box 1 Homo sapiens 68-73 32553931-16 2020 SIGNIFICANCE: Magnoflorine inhibited the malignant phenotypes and increased cisplatin sensitivity of OS cells via modulating miR-410-3p/HMGB1/NF-kappaB pathway. magnoflorine 14-26 high mobility group box 1 Homo sapiens 136-141 32874136-8 2020 Additionally, in both the rabbit trauma model and cellular injury model, CAA combined with high-dosage of VB6 inhibited the expression of inflammatory factors (IL-6, TNF-alpha and IL-1beta) and proteins (HMGB1, TLR4 and p-p65) in HMGB1/TLR4/NF-kappaB pathway. caa 73-76 high mobility group box 1 Homo sapiens 230-235 32874136-9 2020 Most importantly, over-expression of HMGB1 reversed the effect of CAA and VB6 in HUVECs and EA.hy926 cells injury model. caa 66-69 high mobility group box 1 Homo sapiens 37-42 32867128-13 2020 HMGB1 silencing causes loss of response to paclitaxel, whereas silencing of HMGB2 slightly increases sensitivity to olaparib. Paclitaxel 43-53 high mobility group box 1 Homo sapiens 0-5 32874136-10 2020 Conclusion: CAA combined with high-dosage of VB6 alleviated TC and inhibited the expression and secretion of inflammatory factors by inhibiting HMGB1-mediated TLR4/NF-kappaB pathway. caa 12-15 high mobility group box 1 Homo sapiens 144-149 32867128-14 2020 Silencing of either HMGB1 or HMGB2 increases sensitivity to carboplatin. Carboplatin 60-71 high mobility group box 1 Homo sapiens 20-25 32824279-0 2020 N-(2"-Hydroxyphenyl)-2-Propylpentanamide (HO-AAVPA) Inhibits HDAC1 and Increases the Translocation of HMGB1 Levels in Human Cervical Cancer Cells. N-(2-hydroxyphenyl)-2-propylpentanamide 0-40 high mobility group box 1 Homo sapiens 102-107 32824279-6 2020 Furthermore, an increase in ROS production was observed after the treatment with HO-AAVPA, which also could contribute to HMGB1 translocation. ros 28-31 high mobility group box 1 Homo sapiens 122-127 32754040-0 2020 Berberine Ameliorates Subarachnoid Hemorrhage Injury via Induction of Sirtuin 1 and Inhibiting HMGB1/Nf-kappaB Pathway. Berberine 0-9 high mobility group box 1 Homo sapiens 95-100 32724408-12 2020 In conclusion, miR-505-3p inhibited the development of glioma by targeting HMGB1 and regulating AKT expression. mir-505-3p 15-25 high mobility group box 1 Homo sapiens 75-80 32275943-0 2020 HMGB1 release promotes paclitaxel resistance in castration-resistant prostate cancer cells via activating c-Myc expression. Paclitaxel 23-33 high mobility group box 1 Homo sapiens 0-5 32275943-3 2020 In this study, we reported that PTX-induced constant HMGB1 expression and release confers to PTX resistance in mCRPC cells via activating and sustaining c-Myc signaling. Paclitaxel 32-35 high mobility group box 1 Homo sapiens 53-58 32275943-3 2020 In this study, we reported that PTX-induced constant HMGB1 expression and release confers to PTX resistance in mCRPC cells via activating and sustaining c-Myc signaling. Paclitaxel 93-96 high mobility group box 1 Homo sapiens 53-58 32275943-4 2020 PTX upregulated HMGB1 expression and triggered its release in human mCRPC cells. Paclitaxel 0-3 high mobility group box 1 Homo sapiens 16-21 32275943-5 2020 Silencing HMGB1 by RNAi and blocking HMGB1 release by glycyrrhizin or HMGB1 neutralizing antibody sensitized the response of PTX-resistant mCRPC cells to PTX. Glycyrrhizic Acid 54-66 high mobility group box 1 Homo sapiens 37-42 32275943-5 2020 Silencing HMGB1 by RNAi and blocking HMGB1 release by glycyrrhizin or HMGB1 neutralizing antibody sensitized the response of PTX-resistant mCRPC cells to PTX. Glycyrrhizic Acid 54-66 high mobility group box 1 Homo sapiens 37-42 32275943-5 2020 Silencing HMGB1 by RNAi and blocking HMGB1 release by glycyrrhizin or HMGB1 neutralizing antibody sensitized the response of PTX-resistant mCRPC cells to PTX. Paclitaxel 125-128 high mobility group box 1 Homo sapiens 10-15 32275943-5 2020 Silencing HMGB1 by RNAi and blocking HMGB1 release by glycyrrhizin or HMGB1 neutralizing antibody sensitized the response of PTX-resistant mCRPC cells to PTX. Paclitaxel 125-128 high mobility group box 1 Homo sapiens 37-42 32275943-5 2020 Silencing HMGB1 by RNAi and blocking HMGB1 release by glycyrrhizin or HMGB1 neutralizing antibody sensitized the response of PTX-resistant mCRPC cells to PTX. Paclitaxel 125-128 high mobility group box 1 Homo sapiens 37-42 32275943-5 2020 Silencing HMGB1 by RNAi and blocking HMGB1 release by glycyrrhizin or HMGB1 neutralizing antibody sensitized the response of PTX-resistant mCRPC cells to PTX. Paclitaxel 154-157 high mobility group box 1 Homo sapiens 37-42 32275943-5 2020 Silencing HMGB1 by RNAi and blocking HMGB1 release by glycyrrhizin or HMGB1 neutralizing antibody sensitized the response of PTX-resistant mCRPC cells to PTX. Paclitaxel 154-157 high mobility group box 1 Homo sapiens 37-42 32275943-8 2020 Therefore, HMGB1/c-Myc axis is critical in the development of PTX resistance, and targeting HMGB1/c-Myc axis would counteract PTX resistance in mCRPC cells. Paclitaxel 62-65 high mobility group box 1 Homo sapiens 11-16 32275943-8 2020 Therefore, HMGB1/c-Myc axis is critical in the development of PTX resistance, and targeting HMGB1/c-Myc axis would counteract PTX resistance in mCRPC cells. Paclitaxel 126-129 high mobility group box 1 Homo sapiens 92-97 32282121-7 2020 Finally, we proved that miR-141-3p decreased the level of MUAC5AC in LPS-treated NECs through regulating HMGB1. mir-141-3p 24-34 high mobility group box 1 Homo sapiens 105-110 32282121-8 2020 In conclusion, miR-141-3p inhibited LPS-induced MUAC5AC production and the apoptosis of LPS-treated NECs by targeting HMGB1. mir-141-3p 15-25 high mobility group box 1 Homo sapiens 118-123 32722545-9 2020 Several molecules, such as glycyrrhizin (GA), have proven effective in reducing diabetic damage to the retina through the inhibition of HMGB1. Glycyrrhizic Acid 27-39 high mobility group box 1 Homo sapiens 136-141 32722545-9 2020 Several molecules, such as glycyrrhizin (GA), have proven effective in reducing diabetic damage to the retina through the inhibition of HMGB1. Gallium 41-43 high mobility group box 1 Homo sapiens 136-141 32683852-11 2020 Regarding the mechanism, circFANCL served as a sponge of miR-337-3p that was a tumor suppressor in glioma and circFANCL targeted miR-337-3p to regulate HMGB1 that was a target gene of miR-337-3p. circfancl 25-34 high mobility group box 1 Homo sapiens 152-157 32683852-11 2020 Regarding the mechanism, circFANCL served as a sponge of miR-337-3p that was a tumor suppressor in glioma and circFANCL targeted miR-337-3p to regulate HMGB1 that was a target gene of miR-337-3p. circfancl 110-119 high mobility group box 1 Homo sapiens 152-157 32683852-12 2020 Furthermore, HMGB1 down-regulation was responsible for the repression of glioma progression caused by knockdown of circFANCL. circfancl 115-124 high mobility group box 1 Homo sapiens 13-18 32683852-13 2020 CONCLUSIONS: Through the illustration of oncogenic function of circFANCL in glioma by the miR-337-3p/HMGB1 axis, we believed that circFANCL might be a great target in the early diagnosis and late treatment of glioma. circfancl 63-72 high mobility group box 1 Homo sapiens 101-106 32683852-13 2020 CONCLUSIONS: Through the illustration of oncogenic function of circFANCL in glioma by the miR-337-3p/HMGB1 axis, we believed that circFANCL might be a great target in the early diagnosis and late treatment of glioma. circfancl 130-139 high mobility group box 1 Homo sapiens 101-106 32471717-6 2020 Similarly, inhibition for ERK (PD98059), NF-kappaB (BAY11-7082) and c-Jun (c-Jun peptide), respectively, also suppressed the HMGB1 cytoplasm translocation. 2-(2-amino-3-methoxyphenyl)-4H-1-benzopyran-4-one 31-38 high mobility group box 1 Homo sapiens 125-130 32754040-6 2020 Moreover, berberine significantly inhibited high mobile group box 1 (HMGB1)/nuclear factor-kappaB (Nf-kappaB)-dependent pathway and enhanced sirtuin 1 (SIRT1) expression after SAH. Berberine 10-19 high mobility group box 1 Homo sapiens 44-67 32754040-6 2020 Moreover, berberine significantly inhibited high mobile group box 1 (HMGB1)/nuclear factor-kappaB (Nf-kappaB)-dependent pathway and enhanced sirtuin 1 (SIRT1) expression after SAH. Berberine 10-19 high mobility group box 1 Homo sapiens 69-74 32754040-7 2020 Treatment with ex527, a selective SIRT1 inhibitor, reversed berberine-induced SIRT1 activation and inhibitory effects on HMGB1/Nf-kappaB activation. 6-chloro-2,3,4,9-tetrahydro-1H-carbazole-1-carboxamide 15-20 high mobility group box 1 Homo sapiens 121-126 32754040-9 2020 Taken together, these findings suggest that berberine provides beneficial effects against SAH-triggered cerebral inflammation by inhibiting HMGB1/Nf-kappaB pathway, which may be modulated by SIRT1 activation. Berberine 44-53 high mobility group box 1 Homo sapiens 140-145 32469087-7 2020 Concurrently, HMGB1 impaired insulin sensitivities by attenuating the abundance of insulin receptor substrate-1, Akt phosphorylation, GLUT4 translocation, and glucose uptake in granulosa cells, which were reversed by blocking autophagy pathways with siRNA-mediated knockdown of ATG7 or with chloroquine and bafilomycin A1, the lysosome inhibitors. Glucose 159-166 high mobility group box 1 Homo sapiens 14-19 32469087-7 2020 Concurrently, HMGB1 impaired insulin sensitivities by attenuating the abundance of insulin receptor substrate-1, Akt phosphorylation, GLUT4 translocation, and glucose uptake in granulosa cells, which were reversed by blocking autophagy pathways with siRNA-mediated knockdown of ATG7 or with chloroquine and bafilomycin A1, the lysosome inhibitors. Chloroquine 291-302 high mobility group box 1 Homo sapiens 14-19 32469087-7 2020 Concurrently, HMGB1 impaired insulin sensitivities by attenuating the abundance of insulin receptor substrate-1, Akt phosphorylation, GLUT4 translocation, and glucose uptake in granulosa cells, which were reversed by blocking autophagy pathways with siRNA-mediated knockdown of ATG7 or with chloroquine and bafilomycin A1, the lysosome inhibitors. bafilomycin A1 307-321 high mobility group box 1 Homo sapiens 14-19 32347316-6 2020 RESULTS: In hydrogen peroxide (H2O2)-stimulated HL7702 cells, HBx triggered the release of pro-inflammatory mediators apoptosis-associated speck-like protein containing a CARD (ASC), interleukin (IL)-1beta, IL-18, and high-mobility group box 1 (HMGB1); activated NLRP3; and initiated pro-inflammatory cell death (pyroptosis). Hydrogen Peroxide 12-29 high mobility group box 1 Homo sapiens 218-243 32054355-0 2020 HMGB1 is increased in adolescents with polycystic ovary syndrome (PCOS) and decreases after treatment with myo-inositol (MYO) in combination with alpha-lipoic acid (ALA). 5-O-methyl-myo-inositol 107-119 high mobility group box 1 Homo sapiens 0-5 32054355-0 2020 HMGB1 is increased in adolescents with polycystic ovary syndrome (PCOS) and decreases after treatment with myo-inositol (MYO) in combination with alpha-lipoic acid (ALA). 5-O-methyl-myo-inositol 121-124 high mobility group box 1 Homo sapiens 0-5 32054355-0 2020 HMGB1 is increased in adolescents with polycystic ovary syndrome (PCOS) and decreases after treatment with myo-inositol (MYO) in combination with alpha-lipoic acid (ALA). Thioctic Acid 146-163 high mobility group box 1 Homo sapiens 0-5 32054355-0 2020 HMGB1 is increased in adolescents with polycystic ovary syndrome (PCOS) and decreases after treatment with myo-inositol (MYO) in combination with alpha-lipoic acid (ALA). Thioctic Acid 165-168 high mobility group box 1 Homo sapiens 0-5 32054355-4 2020 The aim of this study was to verify changes in HMGB1, in metabolic and endocrine parameters in adolescents with PCOS compared with controls and after treatment with a combination of MYO + ALA. 5-O-methyl-myo-inositol 182-185 high mobility group box 1 Homo sapiens 47-52 32054355-4 2020 The aim of this study was to verify changes in HMGB1, in metabolic and endocrine parameters in adolescents with PCOS compared with controls and after treatment with a combination of MYO + ALA. Thioctic Acid 188-191 high mobility group box 1 Homo sapiens 47-52 32416454-0 2020 Glycyrrhetinic acid alleviates hepatic inflammation injury in viral hepatitis disease via a HMGB1-TLR4 signaling pathway. Glycyrrhetinic Acid 0-19 high mobility group box 1 Homo sapiens 92-97 32606157-6 2020 RESULTS: HMGB1 levels increased with increasing dietary cholesterol, and a negative correlation was found between HMGB1 levels and thrombus formation time. Cholesterol 56-67 high mobility group box 1 Homo sapiens 9-14 32416454-6 2020 We also employed a murine hepatitis virus (MHV) infection model to illustrate that GA treatment inhibited activation of hepatic inflammatory responses by blocking high-mobility group box 1 (HMGB1) cytokine activity. Glycyrrhetinic Acid 83-85 high mobility group box 1 Homo sapiens 163-188 32347316-6 2020 RESULTS: In hydrogen peroxide (H2O2)-stimulated HL7702 cells, HBx triggered the release of pro-inflammatory mediators apoptosis-associated speck-like protein containing a CARD (ASC), interleukin (IL)-1beta, IL-18, and high-mobility group box 1 (HMGB1); activated NLRP3; and initiated pro-inflammatory cell death (pyroptosis). Hydrogen Peroxide 12-29 high mobility group box 1 Homo sapiens 245-250 32416454-6 2020 We also employed a murine hepatitis virus (MHV) infection model to illustrate that GA treatment inhibited activation of hepatic inflammatory responses by blocking high-mobility group box 1 (HMGB1) cytokine activity. Glycyrrhetinic Acid 83-85 high mobility group box 1 Homo sapiens 190-195 32416454-8 2020 Thus, our findings characterize GA as a hepatoprotective therapy agent in hepatic infectious disease not only by suppressing HMGB1 release and blocking HMGB1 cytokine activity, but also via an underlying viral-induced HMGB1-TLR4 immunological regulation axis that occurs during the cytokine storm. Glycyrrhetinic Acid 32-34 high mobility group box 1 Homo sapiens 125-130 32347316-6 2020 RESULTS: In hydrogen peroxide (H2O2)-stimulated HL7702 cells, HBx triggered the release of pro-inflammatory mediators apoptosis-associated speck-like protein containing a CARD (ASC), interleukin (IL)-1beta, IL-18, and high-mobility group box 1 (HMGB1); activated NLRP3; and initiated pro-inflammatory cell death (pyroptosis). Hydrogen Peroxide 31-35 high mobility group box 1 Homo sapiens 218-243 32416454-8 2020 Thus, our findings characterize GA as a hepatoprotective therapy agent in hepatic infectious disease not only by suppressing HMGB1 release and blocking HMGB1 cytokine activity, but also via an underlying viral-induced HMGB1-TLR4 immunological regulation axis that occurs during the cytokine storm. Glycyrrhetinic Acid 32-34 high mobility group box 1 Homo sapiens 152-157 32347316-6 2020 RESULTS: In hydrogen peroxide (H2O2)-stimulated HL7702 cells, HBx triggered the release of pro-inflammatory mediators apoptosis-associated speck-like protein containing a CARD (ASC), interleukin (IL)-1beta, IL-18, and high-mobility group box 1 (HMGB1); activated NLRP3; and initiated pro-inflammatory cell death (pyroptosis). Hydrogen Peroxide 31-35 high mobility group box 1 Homo sapiens 245-250 32416454-8 2020 Thus, our findings characterize GA as a hepatoprotective therapy agent in hepatic infectious disease not only by suppressing HMGB1 release and blocking HMGB1 cytokine activity, but also via an underlying viral-induced HMGB1-TLR4 immunological regulation axis that occurs during the cytokine storm. Glycyrrhetinic Acid 32-34 high mobility group box 1 Homo sapiens 152-157 32602358-0 2021 Glycyrrhizin, an HMGB1 inhibitor, Suppresses Interleukin-1beta-Induced Inflammatory Responses in Chondrocytes from Patients with Osteoarthritis. Glycyrrhizic Acid 0-12 high mobility group box 1 Homo sapiens 17-22 32612147-8 2020 8-NitroG formation in A549 cells was also largely suppressed by small interfering RNA (siRNA) for high-mobility group box-1 (HMGB1), receptor for advanced glycation and end products (AGER, RAGE) and Toll-like receptor 9 (TLR9). 8-nitrog 0-8 high mobility group box 1 Homo sapiens 98-123 32612147-8 2020 8-NitroG formation in A549 cells was also largely suppressed by small interfering RNA (siRNA) for high-mobility group box-1 (HMGB1), receptor for advanced glycation and end products (AGER, RAGE) and Toll-like receptor 9 (TLR9). 8-nitrog 0-8 high mobility group box 1 Homo sapiens 125-130 32612147-9 2020 These results suggest that indium compounds induce inflammation-mediated DNA damage in lung epithelial cells via the HMGB1-RAGE-TLR9 pathway. Indium 27-33 high mobility group box 1 Homo sapiens 117-122 32602358-8 2021 HMGB1 inhibition by glycyrrhizin improved cell viability of chondrocytes treated with IL-1beta. Glycyrrhizic Acid 20-32 high mobility group box 1 Homo sapiens 0-5 32602358-9 2021 Glycyrrhizin suppressed IL-1beta-induced upregulation of HMGB1 and inflammatory mediators and cytokines, including PGE2, NO, proinflammatory cytokines, and MMPs. Glycyrrhizic Acid 0-12 high mobility group box 1 Homo sapiens 57-62 32602358-3 2021 This study aimed to investigate how inhibition of HMGB1 by glycyrrhizin might affect inflammatory responses and viability of OA patient-derived chondrocytes treated with IL-1beta. Glycyrrhizic Acid 59-71 high mobility group box 1 Homo sapiens 50-55 32423802-8 2020 Treatment with EW-7197, a transforming growth factor-beta (TGF-beta) type I receptor kinase inhibitor, prevented all the effects of HMGB1 in Calu-3 cells. vactosertib 15-22 high mobility group box 1 Homo sapiens 132-137 32600307-0 2020 The alpha7 nicotinic acetylcholine receptor agonist, GTS-21, attenuates hyperoxia-induced acute inflammatory lung injury by alleviating the accumulation of HMGB1 in the airways and the circulation. 3-(2,4-dimethoxybenzylidene)anabaseine 53-59 high mobility group box 1 Homo sapiens 156-161 32600316-5 2020 This comprehension was recently reinforced by results reported in Molecular Medicine by Sitapara and coworkers demonstrating that administration of an alpha7 nicotinic acetylcholine receptor agonist attenuated hyperoxia-induced acute inflammatory lung injury by alleviating the accumulation of HMGB1 in the airways and the circulation. sitapara 88-96 high mobility group box 1 Homo sapiens 294-299 32423802-10 2020 The effects of HMGB1 contribute to TGF-beta signaling and EW-7197 shows potential for use in therapy for HMGB1-induced airway inflammation. vactosertib 58-65 high mobility group box 1 Homo sapiens 105-110 32587593-5 2020 We currently investigated that peroxiredoxins I and II (PrxI/II) induce the intramolecular disulfide bond formation of HMGB1 in the nucleus. Disulfides 91-100 high mobility group box 1 Homo sapiens 119-124 32636835-2 2020 HMGB1 binds to its specific receptors not only to activate the nuclear factor (NF)-kappaB and mitogen-activated protein kinase (MAPK) pathways but also to regulate the activation of the phosphatidylinositol 3"-kinase/protein kinase B/mammalian target of the rapamycin (PI3K/AKT/mTOR) pathway. Sirolimus 258-267 high mobility group box 1 Homo sapiens 0-5 32321806-0 2020 PCV2 Induces Reactive Oxygen Species To Promote Nucleocytoplasmic Translocation of the Viral DNA Binding Protein HMGB1 To Enhance Its Replication. Reactive Oxygen Species 13-36 high mobility group box 1 Homo sapiens 113-118 32321806-4 2020 Here, we demonstrate that nuclear HMGB1 negatively regulated PCV2 replication as shown by overexpression of HMGB1 or blockage of its nucleocytoplasmic translocation with ethyl pyruvate. ethyl pyruvate 170-184 high mobility group box 1 Homo sapiens 34-39 32321806-8 2020 Elevation of reactive oxygen species (ROS) induced by PCV2 infection was closely associated with cytosolic translocation of nuclear HMGB1. Reactive Oxygen Species 13-36 high mobility group box 1 Homo sapiens 132-137 32321806-8 2020 Elevation of reactive oxygen species (ROS) induced by PCV2 infection was closely associated with cytosolic translocation of nuclear HMGB1. Reactive Oxygen Species 38-41 high mobility group box 1 Homo sapiens 132-137 32321806-9 2020 Treatment of PCV2-infected cells with ethyl pyruvate or N-acetylcysteine downregulated PCV2-induced ROS production, suppressed nucleocytoplasmic HMGB1 translocation, and decreased PCV2 replication. ethyl pyruvate 38-52 high mobility group box 1 Homo sapiens 145-150 32321806-9 2020 Treatment of PCV2-infected cells with ethyl pyruvate or N-acetylcysteine downregulated PCV2-induced ROS production, suppressed nucleocytoplasmic HMGB1 translocation, and decreased PCV2 replication. Acetylcysteine 56-72 high mobility group box 1 Homo sapiens 145-150 32321806-13 2020 PCV2 then upregulates host reactive oxygen species (ROS) to prevent sequestration of its genome by expelling nuclear HMGB1 into the cytosol. Reactive Oxygen Species 27-50 high mobility group box 1 Homo sapiens 117-122 32321806-13 2020 PCV2 then upregulates host reactive oxygen species (ROS) to prevent sequestration of its genome by expelling nuclear HMGB1 into the cytosol. Reactive Oxygen Species 52-55 high mobility group box 1 Homo sapiens 117-122 32321806-15 2020 This study also provides insight into the justification and pharmacological basis of antioxidants as an adjunct therapy in PCV2 infection or possibly other diseases caused by the viruses that deploy the ROS-HMGB1 interaction favoring their replication. Reactive Oxygen Species 203-206 high mobility group box 1 Homo sapiens 207-212 32587593-6 2020 Disulfide HMGB1 is preferentially transported out of the nucleus by binding to the nuclear exportin chromosome-region maintenance 1 (CRM1). Disulfides 0-9 high mobility group box 1 Homo sapiens 10-15 32587593-7 2020 We determined the kinetics of HMGB1 oxidation in bone marrow-derived macrophage as early as a few minutes after lipopolysaccharide treatment, peaking at 4 h while disulfide HMGB1 accumulation was observed within the cells, starting to secrete in the late time point. Disulfides 163-172 high mobility group box 1 Homo sapiens 173-178 32419317-5 2020 THP-1 cells were induced to differentiate into M1 macrophages, then challenged with either CC-5013 or an HMGB1 inhibitor, glycyrrhizin. Glycyrrhizic Acid 122-134 high mobility group box 1 Homo sapiens 105-110 32642410-0 2020 MiR-142-3p enhances chemosensitivity of breast cancer cells and inhibits autophagy by targeting HMGB1. mir-142-3p 0-10 high mobility group box 1 Homo sapiens 96-101 32642410-6 2020 Moreover, overexpression of HMGB1 dramatically reversed the promotion of apoptosis and inhibition of autophagy mediated by miR-142-3p up-regulation. mir-142-3p 123-133 high mobility group box 1 Homo sapiens 28-33 32642410-7 2020 In conclusion, miR-142-3p overexpression may inhibit autophagy and promote the drug sensitivity of breast cancer cells to DOX by targeting HMGB1. mir-142-3p 15-25 high mobility group box 1 Homo sapiens 139-144 32642410-7 2020 In conclusion, miR-142-3p overexpression may inhibit autophagy and promote the drug sensitivity of breast cancer cells to DOX by targeting HMGB1. Doxorubicin 122-125 high mobility group box 1 Homo sapiens 139-144 32642410-8 2020 The miR-142-3p/HMGB1 axis might be a novel target to regulate the drug resistance of breast cancer patients. mir-142-3p 4-14 high mobility group box 1 Homo sapiens 15-20 32251024-6 2020 Experiments with anti-N-methyl-D-aspartate antibodies show that HMGB1 and C1q were essential to activate microglia which, in turn, remodel damaged neurons in vivo. N-Methylaspartate 22-42 high mobility group box 1 Homo sapiens 64-69 32169518-7 2020 HMGB1 concentration in plasma was higher in the URSA group than the control group. ursa 48-52 high mobility group box 1 Homo sapiens 0-5 32277430-8 2020 Furthermore, reduction in the levels of high-mobility group box 1 protein, inhibitor of kappa B protein-alpha, matrix metalloproteinase-9 and C-reactive protein are some other important hesperidin-derived hepatoprotective mechanisms. Hesperidin 186-196 high mobility group box 1 Homo sapiens 40-65 32633407-8 2020 Lipopolysaccharide (LPS) was used in stimulating the macrophages to activate inflammatory response and recombined human HMGB1 was used to resist the function of GL to explore whether GL acted via the target of HMGB1. Glycyrrhizic Acid 161-163 high mobility group box 1 Homo sapiens 120-125 32359123-5 2020 Serum HMGB1 level was also positively correlated with HbA1c and negatively correlated with nitric oxide (NO) in diabetic patients. Nitric Oxide 91-103 high mobility group box 1 Homo sapiens 6-11 32169518-8 2020 Furthermore, the levels of HMGB1 of subjects with URSA could be reduced by administrating low doses of aspirin (ASPL). Aspirin 103-110 high mobility group box 1 Homo sapiens 27-32 32514250-4 2020 To observe the relationship between HMGB1, GPX4 and inflammation or ROS, Western blot assays were adopted. Reactive Oxygen Species 68-71 high mobility group box 1 Homo sapiens 36-41 32514250-8 2020 Furthermore, by utilizing siHMGB1 and its inhibitor, our discoveries demonstrate that HMGB1 knockdown can inhibit inflammation and reactive oxygen species (ROS) accumulation via NF-kB. Reactive Oxygen Species 131-154 high mobility group box 1 Homo sapiens 28-33 32514250-8 2020 Furthermore, by utilizing siHMGB1 and its inhibitor, our discoveries demonstrate that HMGB1 knockdown can inhibit inflammation and reactive oxygen species (ROS) accumulation via NF-kB. Reactive Oxygen Species 156-159 high mobility group box 1 Homo sapiens 28-33 32523550-7 2020 In line with the supposed switch to a pro-inflammatory phenotype known as the senescence associated secretory phenotype (SASP), we found that both RS and DS upregulated IL-1beta and released HMGB-1 from the nucleus, while RS also showed IL-6 upregulation. ds 154-156 high mobility group box 1 Homo sapiens 191-197 32466385-12 2020 Treatment with PROG reduced HMGB1 release and NLRP3 inflammasome activation, and enhanced autophagy in stressed and unstressed ischemic animals. Progesterone 15-19 high mobility group box 1 Homo sapiens 28-33 32489326-0 2020 LncRNA NNT-AS1 contributes to the cisplatin resistance of cervical cancer through NNT-AS1/miR-186/HMGB1 axis. Cisplatin 34-43 high mobility group box 1 Homo sapiens 98-103 31978425-6 2020 Additionally, our findings demonstrate that nobiletin potently ameliorated IL-21-induced increased production of reactive oxygen species and 4-hydroxynonenal, increased expression of interleukin 6 (IL-6), tumor necrosis factor-alpha (TNF-alpha), and high-mobility group box 1 (HMGB1), and decreased mitochondrial membrane potential. nobiletin 44-53 high mobility group box 1 Homo sapiens 250-275 31978425-6 2020 Additionally, our findings demonstrate that nobiletin potently ameliorated IL-21-induced increased production of reactive oxygen species and 4-hydroxynonenal, increased expression of interleukin 6 (IL-6), tumor necrosis factor-alpha (TNF-alpha), and high-mobility group box 1 (HMGB1), and decreased mitochondrial membrane potential. nobiletin 44-53 high mobility group box 1 Homo sapiens 277-282 32536835-0 2020 Aloin promotes cell apoptosis by targeting HMGB1-TLR4-ERK axis in human melanoma cells. alloin 0-5 high mobility group box 1 Homo sapiens 43-48 32536835-7 2020 Furthermore, HMGB1 release was significantly inhibited in melanoma cancer cells treated with ALO. alloin 93-96 high mobility group box 1 Homo sapiens 13-18 32536835-8 2020 Knockdown of HMGB1 could enhance melanoma cell death that is induced by ALO treatment. alloin 72-75 high mobility group box 1 Homo sapiens 13-18 32523355-15 2020 Conclusion: HMGB1 promotes NHEJ via interaction with Ku70 resulting in resistance to IR and cisplatin. Cisplatin 92-101 high mobility group box 1 Homo sapiens 12-17 32523355-16 2020 Inhibition of HMGB1 by glycyrrhizin is a potential therapeutic regimen to treat cisplatin and IR resistant NPC patients. Glycyrrhizic Acid 23-35 high mobility group box 1 Homo sapiens 14-19 32523355-16 2020 Inhibition of HMGB1 by glycyrrhizin is a potential therapeutic regimen to treat cisplatin and IR resistant NPC patients. Cisplatin 80-89 high mobility group box 1 Homo sapiens 14-19 32403440-5 2020 KEY RESULTS: Incubation of human CM with HMGB-1 or histones is associated with changes in calcium handling, a reduction of cell viability and a substantial reduction of the mitochondrial respiratory capacity. Calcium 90-97 high mobility group box 1 Homo sapiens 41-47 32380958-4 2020 Disulfide-HMGB1 triggers TLR4 receptors generating pro-inflammatory cytokine release. Disulfides 0-9 high mobility group box 1 Homo sapiens 10-15 32380958-9 2020 Extracellular HMGB1 generally exists in vivo bound to other molecules, including PAMPs and DAMPs. dinitrophenyl-aminopropyl-methylamine 91-96 high mobility group box 1 Homo sapiens 14-19 32431618-0 2020 Quercetin Attenuates d-GaLN-Induced L02 Cell Damage by Suppressing Oxidative Stress and Mitochondrial Apoptosis via Inhibition of HMGB1. Quercetin 0-9 high mobility group box 1 Homo sapiens 130-135 32431618-4 2020 Quercetin (Que), as an antioxidant, is a potential phytochemical with hepatocyte protection and is also considered to be an inhibitor of HMGB1. Quercetin 0-9 high mobility group box 1 Homo sapiens 137-142 32431618-4 2020 Quercetin (Que), as an antioxidant, is a potential phytochemical with hepatocyte protection and is also considered to be an inhibitor of HMGB1. Quercetin 0-3 high mobility group box 1 Homo sapiens 137-142 32431618-6 2020 The present study explored whether the hepatoprotective effect of Que antagonizes HMGB1, and subsequent molecular signaling events. Quercetin 66-69 high mobility group box 1 Homo sapiens 82-87 32431618-9 2020 Further research showed that after transfection with HMGB1 short hairpin RNA (shRNA), cell viability was improved, and intracellular ROS production and apoptosis were suppressed. Reactive Oxygen Species 133-136 high mobility group box 1 Homo sapiens 53-58 32431618-10 2020 When co-treated with Que, the expression of HMGB1 was decreased significantly, the expression of proteins in the corresponding signal pathway were further reduced, and the production of ROS and apoptosis were further suppressed. Quercetin 21-24 high mobility group box 1 Homo sapiens 44-49 32431618-10 2020 When co-treated with Que, the expression of HMGB1 was decreased significantly, the expression of proteins in the corresponding signal pathway were further reduced, and the production of ROS and apoptosis were further suppressed. Reactive Oxygen Species 186-189 high mobility group box 1 Homo sapiens 44-49 32431618-12 2020 Taken together, these results indicate that Que significantly improves d-GaLN-induced cellular damage by inhibiting oxidative stress and mitochondrial apoptosis via inhibiting HMGB1. Quercetin 44-47 high mobility group box 1 Homo sapiens 176-181 32456685-1 2020 BACKGROUND: Chemotherapeutics can stimulate immune antitumor response by inducing immunogenic cell death (ICD), which is activated by Damage-Associated Molecular Patterns (DAMPs) like the exposure of calreticulin (CRT) on the cell surface, the release of ATP and the secretion of High Mobility Group Box 1 (HMGB1). Adenosine Triphosphate 255-258 high mobility group box 1 Homo sapiens 280-305 32456685-1 2020 BACKGROUND: Chemotherapeutics can stimulate immune antitumor response by inducing immunogenic cell death (ICD), which is activated by Damage-Associated Molecular Patterns (DAMPs) like the exposure of calreticulin (CRT) on the cell surface, the release of ATP and the secretion of High Mobility Group Box 1 (HMGB1). Adenosine Triphosphate 255-258 high mobility group box 1 Homo sapiens 307-312 32514327-5 2020 Consistent with experimental data, the acidic tail was predicted to adopt an extended conformation that allows it to make a range of hydrogen-bonding and electrostatic interactions with the box-like domains that stabilize the overall structure of HMGB1. Hydrogen 133-141 high mobility group box 1 Homo sapiens 247-252 32523355-11 2020 Mutational analysis revealed that serine 155 of Ku70 was required for its direct interaction with HMGB1. Serine 34-40 high mobility group box 1 Homo sapiens 98-103 32523355-13 2020 Deficiency of HMGB1 sensitized wild-type (WT) and resistant NPC cells to IR and cisplatin. Cisplatin 80-89 high mobility group box 1 Homo sapiens 14-19 32523355-14 2020 Glycyrrhizin, which is HMGB1 inhibitor, impaired DNA binding of HMGB1 and exhibited excellent synergy with IR and cisplatin. Glycyrrhizic Acid 0-12 high mobility group box 1 Homo sapiens 23-28 32523355-14 2020 Glycyrrhizin, which is HMGB1 inhibitor, impaired DNA binding of HMGB1 and exhibited excellent synergy with IR and cisplatin. Glycyrrhizic Acid 0-12 high mobility group box 1 Homo sapiens 64-69 32014690-6 2020 Here we found that TDI induced pyroptosis in 16HBE cells, as evidenced by enhanced expressions of caspase-1 and elevated levels of LDH, IL-1beta and HMGB1. Toluene 2,4-Diisocyanate 19-22 high mobility group box 1 Homo sapiens 149-154 32588826-4 2020 Ethanol consumption leads to activation of neuroimmune signaling in the central nervous system through many types of Toll-like receptors (TLRs), as well as the release of their endogenous agonists (HMGB1 protein, S100 protein, heat shock proteins, extracellular matrix breakdown proteins). Ethanol 0-7 high mobility group box 1 Homo sapiens 198-203 32252560-8 2020 In this regard, glycyrrhetic acid (GA), the active component of Glycyrrhiza glabra, can efficiently block HMGB1. Glycyrrhetinic Acid 16-33 high mobility group box 1 Homo sapiens 106-111 32252560-8 2020 In this regard, glycyrrhetic acid (GA), the active component of Glycyrrhiza glabra, can efficiently block HMGB1. Glycyrrhetinic Acid 35-37 high mobility group box 1 Homo sapiens 106-111 32246277-6 2020 Radiotherapy induces so-called "immunogenic cell death", which involves cell surface translocation of calreticulin and extracellular release of high-mobility group protein box 1 (HMGB-1) and adenosine-5"-triphosphate (ATP). Adenosine Triphosphate 218-221 high mobility group box 1 Homo sapiens 179-185 31846033-0 2020 Corrigendum to "Carbon Nanomaterials Stimulate HMGB1 Release From Macrophages and Induce Cell Migration and Invasion". Carbon 16-22 high mobility group box 1 Homo sapiens 47-52 32440094-10 2020 Results: Our findings demonstrate that laquinimod reduced the expression of key inflammatory cytokines and chemokines, including IL-6, MCP-1, and HMGB1. laquinimod 39-49 high mobility group box 1 Homo sapiens 146-151 32489453-1 2020 Purpose: We aimed to investigate the association of single-nucleotide polymorphisms (SNPs) in HMGB1, REV3L, and NFE2L2 with prognosis in lung cancer patients with platinum-based chemotherapy. Platinum 163-171 high mobility group box 1 Homo sapiens 94-99 32489453-4 2020 Results: The results revealed that patients carrying TC or CC genotype in REV3L rs462779 (HR=0.67, 95% CI=0.51-0.90, P=0.007) and AG or GG genotype in HMGB1 rs1045411 (HR=0.61, 95% CI=0.38-0.99, P=0.046) had a better overall survival. Technetium 53-55 high mobility group box 1 Homo sapiens 151-156 32489453-9 2020 The REV3L rs462779 and HMGB1 rs1045411 may serve as prognosis markers in lung cancer patients with platinum-based chemotherapy. Platinum 99-107 high mobility group box 1 Homo sapiens 23-28 32340172-0 2020 Glycyrrhizin Inhibits PEDV Infection and Proinflammatory Cytokine Secretion via the HMGB1/TLR4-MAPK p38 Pathway. Glycyrrhizic Acid 0-12 high mobility group box 1 Homo sapiens 84-89 32340172-8 2020 Collectively, our data indicated that GLY inhibited PEDV infection and decreased proinflammatory cytokine secretion via the HMGB1/TLR4-mitogen-activated protein kinase (MAPK) p38 pathway. Glycyrrhizic Acid 38-41 high mobility group box 1 Homo sapiens 124-129 31928132-6 2020 We further demonstrated that SIRT6 and PARP1 activation were required for chemotherapy-induced ADP-ribosylation of HMGB1 in leukemic cells and then influenced the acetylation of HMGB1, finally promoting the autophagy of leukemic cells mediated by HMGB1 translocation. Adenosine Diphosphate 95-98 high mobility group box 1 Homo sapiens 115-120 32328193-0 2020 Self-enforcing HMGB1/NF-kappaB/HIF-1alpha Feedback Loop Promotes Cisplatin Resistance in Hepatocellular Carcinoma Cells. Cisplatin 65-74 high mobility group box 1 Homo sapiens 15-20 32328193-6 2020 Further, we showed that HMGB1 has an important role in mediating cisplatin resistance via an HMGB1/ nuclear factor kappa-B (NF-kappaB)/ hypoxia inducible factor-1alpha (HIF-1alpha) feedback loop. Cisplatin 65-74 high mobility group box 1 Homo sapiens 24-29 32328193-6 2020 Further, we showed that HMGB1 has an important role in mediating cisplatin resistance via an HMGB1/ nuclear factor kappa-B (NF-kappaB)/ hypoxia inducible factor-1alpha (HIF-1alpha) feedback loop. Cisplatin 65-74 high mobility group box 1 Homo sapiens 93-98 32328193-7 2020 The study findings reveal an unappreciated molecular mechanism of HMGB1-mediated cisplatin resistance and may provide a new clue in cancer therapy. Cisplatin 81-90 high mobility group box 1 Homo sapiens 66-71 31928132-0 2020 SIRT6-PARP1 is involved in HMGB1 polyADP-ribosylation and acetylation and promotes chemotherapy-induced autophagy in leukemia. Adenosine Diphosphate 33-40 high mobility group box 1 Homo sapiens 27-32 32331368-5 2020 In Pgp-expressing/doxorubicin-resistant cells, Tariquidar and R-3 increased doxorubicin accumulation and toxicity, reduced Pgp activity, and increased CRT translocation and ATP and HMGB1 release. tariquidar 47-57 high mobility group box 1 Homo sapiens 181-186 32035144-4 2020 Our results showed that hesperetin significantly relieved the symptoms of DSS -induced colitis and increased the expressions of zonula occludens-1 (ZO-1), occludin and mucin2 (MUC-2) as well as the decrease of tumor necrosis factor-alpha (TNF-alpha), interleukin (IL)-1beta, IL-18, HMGB1 and IL-6. hesperetin 24-34 high mobility group box 1 Homo sapiens 282-287 32328059-7 2020 Caspase-11/GsdmD was responsible for HMGB1 translocation from nucleus to the cytoplasm via calcium changing-induced phosphorylation of calcium-calmodulin kinase kinase (camkk)beta during endotoxemia. Calcium 91-98 high mobility group box 1 Homo sapiens 37-42 32328059-10 2020 These findings provide a novel mechanism that TLR4 signaling results in an increase in caspase-11 expression, as well as increased exosome release, while caspase-11/GsdmD activation/cleavage leads to accumulation of HMGB1 in the cytoplasm through a process associated with the release of calcium from the endoplasmic reticulum and camkkbeta activation. Calcium 288-295 high mobility group box 1 Homo sapiens 216-221 32329842-1 2020 OBJECTIVE: The aim of this study was to investigate the effects of micro ribonucleic acid (miR)-129-5p on the proliferation and apoptosis of gastric cancer cells via targeted repression on the expression of high mobility group protein B1 (HMGB1). micro ribonucleic acid 67-89 high mobility group box 1 Homo sapiens 207-237 32329842-1 2020 OBJECTIVE: The aim of this study was to investigate the effects of micro ribonucleic acid (miR)-129-5p on the proliferation and apoptosis of gastric cancer cells via targeted repression on the expression of high mobility group protein B1 (HMGB1). micro ribonucleic acid 67-89 high mobility group box 1 Homo sapiens 239-244 32329842-1 2020 OBJECTIVE: The aim of this study was to investigate the effects of micro ribonucleic acid (miR)-129-5p on the proliferation and apoptosis of gastric cancer cells via targeted repression on the expression of high mobility group protein B1 (HMGB1). methionylisoleucylarginine 91-94 high mobility group box 1 Homo sapiens 207-237 32329842-1 2020 OBJECTIVE: The aim of this study was to investigate the effects of micro ribonucleic acid (miR)-129-5p on the proliferation and apoptosis of gastric cancer cells via targeted repression on the expression of high mobility group protein B1 (HMGB1). methionylisoleucylarginine 91-94 high mobility group box 1 Homo sapiens 239-244 32329842-1 2020 OBJECTIVE: The aim of this study was to investigate the effects of micro ribonucleic acid (miR)-129-5p on the proliferation and apoptosis of gastric cancer cells via targeted repression on the expression of high mobility group protein B1 (HMGB1). -129-5p 95-102 high mobility group box 1 Homo sapiens 207-237 32329842-1 2020 OBJECTIVE: The aim of this study was to investigate the effects of micro ribonucleic acid (miR)-129-5p on the proliferation and apoptosis of gastric cancer cells via targeted repression on the expression of high mobility group protein B1 (HMGB1). -129-5p 95-102 high mobility group box 1 Homo sapiens 239-244 32095984-10 2020 Uric acid and HMGB1 siRNA inhibited HMGB1 acetylation to prevent its transport from the nucleus to the cytoplasm. Uric Acid 0-9 high mobility group box 1 Homo sapiens 36-41 32095984-12 2020 These data indicated that uric acid may prevent cell injury mainly by inhibiting HMGB1 acetylation to regulate TLR4/NF-kappaB pathways and reduce the levels of inflammatory factors. Uric Acid 26-35 high mobility group box 1 Homo sapiens 81-86 32095984-0 2020 The protective effect of uric acid in reducing TLR4/NF-kappaB activation through the inhibition of HMGB1 acetylation in a model of ischemia-reperfusion injury in vitro. Uric Acid 25-34 high mobility group box 1 Homo sapiens 99-104 31802434-0 2020 HMGB1 Is a Therapeutic Target and Biomarker in Diazepam-Refractory Status Epilepticus with Wide Time Window. Diazepam 47-55 high mobility group box 1 Homo sapiens 0-5 31802434-2 2020 Here, we investigated the proinflammatory cytokine high mobility group box-1 (HMGB1) as a candidate therapeutic target for diazepam (DZP)-refractory SE. Diazepam 123-131 high mobility group box 1 Homo sapiens 51-76 31858421-0 2020 HMGB1 Mediates Paraquat-Induced Neuroinflammatory Responses via Activating RAGE Signaling Pathway. Paraquat 15-23 high mobility group box 1 Homo sapiens 0-5 31802434-2 2020 Here, we investigated the proinflammatory cytokine high mobility group box-1 (HMGB1) as a candidate therapeutic target for diazepam (DZP)-refractory SE. Diazepam 123-131 high mobility group box 1 Homo sapiens 78-83 31858421-3 2020 High-mobility group box 1 (HMGB1) has been proven to be relevant to the neuroinflammation involved in PD; however, whether and how HMGB1 exerts modulatory effects in nervous system upon PQ exposure remain elusive. Paraquat 186-188 high mobility group box 1 Homo sapiens 131-136 31858421-4 2020 Therefore, the present study investigated the underlying association between HMGB1 and PQ exposure in SH-SY5Y cells, which is a well-established in vitro model for PD research. Paraquat 87-89 high mobility group box 1 Homo sapiens 77-82 31802434-2 2020 Here, we investigated the proinflammatory cytokine high mobility group box-1 (HMGB1) as a candidate therapeutic target for diazepam (DZP)-refractory SE. Diazepam 133-136 high mobility group box 1 Homo sapiens 51-76 31858421-5 2020 We observed that HMGB1 was markedly increased in a concentration and time-dependent manner upon PQ exposure, and the elevated HMGB1 could be translocated into cytosol and then released to the extracellular milieu of SH-SY5Y cells. Paraquat 96-98 high mobility group box 1 Homo sapiens 17-22 31802434-2 2020 Here, we investigated the proinflammatory cytokine high mobility group box-1 (HMGB1) as a candidate therapeutic target for diazepam (DZP)-refractory SE. Diazepam 133-136 high mobility group box 1 Homo sapiens 78-83 31858421-7 2020 These results suggested that HMGB1 is involved in the PQ-induced neuron death via activating RAGE signaling pathways and promoting neuroinflammatory responses. Paraquat 54-56 high mobility group box 1 Homo sapiens 29-34 31802434-4 2020 Exogenous HMGB1 was sufficient to directly induce DZP-refractory SE in nonrefractory SE. Diazepam 50-53 high mobility group box 1 Homo sapiens 10-15 31802434-5 2020 Neutralization of HMGB1 with an anti-HMGB1 monoclonal antibody decreased the incidence of SE and alleviated the severity of seizure activity in DZP-refractory SE, which was mediated by a Toll-like receptor 4 (TLR4)-dependent pathway. Diazepam 144-147 high mobility group box 1 Homo sapiens 18-23 31802434-5 2020 Neutralization of HMGB1 with an anti-HMGB1 monoclonal antibody decreased the incidence of SE and alleviated the severity of seizure activity in DZP-refractory SE, which was mediated by a Toll-like receptor 4 (TLR4)-dependent pathway. Diazepam 144-147 high mobility group box 1 Homo sapiens 37-42 31802434-6 2020 Importantly, anti-HMGB1 mAb reversed DZP-refractory SE with a wide time window, extending the therapeutic window from 30 to 180 min. Diazepam 37-40 high mobility group box 1 Homo sapiens 18-23 31802434-7 2020 Furthermore, we found the upregulation of plasma HMGB1 level is closely correlated with the therapeutic response of anti-HMGB1 mAb in DZP-refractory SE. Diazepam 134-137 high mobility group box 1 Homo sapiens 49-54 31802434-7 2020 Furthermore, we found the upregulation of plasma HMGB1 level is closely correlated with the therapeutic response of anti-HMGB1 mAb in DZP-refractory SE. Diazepam 134-137 high mobility group box 1 Homo sapiens 121-126 31802434-8 2020 All these results indicated that HMGB1 is a potential therapeutic target and a useful predictive biomarker in DZP-refractory SE. Diazepam 110-113 high mobility group box 1 Homo sapiens 33-38 32221312-0 2020 New insights in acetaminophen toxicity: HMGB1 contributes by itself to amplify hepatocyte necrosis in vitro through the TLR4-TRIF-RIPK3 axis. Acetaminophen 16-29 high mobility group box 1 Homo sapiens 40-45 32098696-0 2020 Propofol upregulates miR-320a and reduces HMGB1 by downregulating ANRIL to inhibit PTC cell malignant behaviors. Propofol 0-8 high mobility group box 1 Homo sapiens 42-47 32098696-15 2020 CONCLUSION: Propofol upregulated miR-320a and reduced HMGB1 by downregulating ANRIL and inactivating the Wnt/beta-catenin and NF-kappaB pathways, thus preventing PTC cell malignant behaviors. Propofol 12-20 high mobility group box 1 Homo sapiens 54-59 31962109-7 2020 Phoenixin-14 attenuated oxidative stress via downregulation of ROS and NOX1 and inhibited HMGB1 expression. phoenixin 0-9 high mobility group box 1 Homo sapiens 90-95 32221312-1 2020 Extracellular release of HMGB1 contributes to acetaminophen-induced liver injury. Acetaminophen 46-59 high mobility group box 1 Homo sapiens 25-30 32221312-3 2020 Here we studied, in vitro, the role of HMGB1 in amplifying the acetaminophen-induced hepatocyte necrosis process. Acetaminophen 63-76 high mobility group box 1 Homo sapiens 39-44 32221312-5 2020 We confirmed that addition of acetaminophen induced HepaRG cell death and HMGB1 release. Acetaminophen 30-43 high mobility group box 1 Homo sapiens 74-79 32221312-6 2020 We showed that inhibition of HMGB1 decreased acetaminophen-induced HepaRG cell death, suggesting a feedforward effect. Acetaminophen 45-58 high mobility group box 1 Homo sapiens 29-34 32221312-11 2020 Our data support that released HMGB1 from acetaminophen-stressed hepatocytes induced necrosis of neighboring hepatocytes by TLR4-TRIF-RIPK3- pathway. Acetaminophen 42-55 high mobility group box 1 Homo sapiens 31-36 32221312-12 2020 This in vitro study gives new insights in the role of HMGB1 in the amplification of acetaminophen-induced toxicity. Acetaminophen 84-97 high mobility group box 1 Homo sapiens 54-59 32161598-4 2020 In our study, we observed that low-dose gemcitabine treatment enhanced the immunogenicity of lung cancer by increasing the exposure of calreticulin, high mobility group box 1, and upregulating expression of NKG2D ligands. gemcitabine 40-51 high mobility group box 1 Homo sapiens 149-174 32265930-5 2020 Disulfide HMGB1 activates the TLR4 complex by binding to MD-2. Disulfides 0-9 high mobility group box 1 Homo sapiens 10-15 32188725-4 2020 Here, we found that a heparan sulfate (HS) octadecasaccharide (18-mer-HP or hepatoprotective 18-mer) displays potent hepatoprotection by targeting the HMGB1/RAGE axis. Heparitin Sulfate 22-37 high mobility group box 1 Homo sapiens 151-156 32188725-4 2020 Here, we found that a heparan sulfate (HS) octadecasaccharide (18-mer-HP or hepatoprotective 18-mer) displays potent hepatoprotection by targeting the HMGB1/RAGE axis. Heparitin Sulfate 39-41 high mobility group box 1 Homo sapiens 151-156 32188725-4 2020 Here, we found that a heparan sulfate (HS) octadecasaccharide (18-mer-HP or hepatoprotective 18-mer) displays potent hepatoprotection by targeting the HMGB1/RAGE axis. octadecasaccharide 43-61 high mobility group box 1 Homo sapiens 151-156 32188725-6 2020 Furthermore, purified syndecan-1, but not syndecan-1 core protein, binds to HMGB1, suggesting that HMGB1 binds to HS polysaccharide side chains of syndecan-1. Heparitin Sulfate 114-116 high mobility group box 1 Homo sapiens 99-104 32188725-6 2020 Furthermore, purified syndecan-1, but not syndecan-1 core protein, binds to HMGB1, suggesting that HMGB1 binds to HS polysaccharide side chains of syndecan-1. Polysaccharides 117-131 high mobility group box 1 Homo sapiens 99-104 32169964-0 2020 Expression of Concern: HMGB1 and Its Hyperacetylated Isoform are Sensitive and Specific Serum Biomarkers to Detect Asbestos Exposure and to Identify Mesothelioma Patients. Asbestos 115-123 high mobility group box 1 Homo sapiens 23-28 31879132-0 2020 Corrigendum to "HMGB1 contributes to adriamycin-induced cardiotoxicity via up-regulating autophagy" [Toxicol. Doxorubicin 37-47 high mobility group box 1 Homo sapiens 16-21 32208365-4 2020 Correlations between metformin concentration and high-mobility group box 1 (HMGB1) and miR-142-3p levels were assessed. Metformin 21-30 high mobility group box 1 Homo sapiens 76-81 32208365-7 2020 HMGB1 gene expression correlated negatively with metformin concentration, whereas miR-142-3p expression correlated positively with metformin concentration. Metformin 49-58 high mobility group box 1 Homo sapiens 0-5 32208365-8 2020 In addition, mimic-induced miR-142-3p elevation resulted in decreased HMGB1 and LC3II levels and elevated p62 levels in the high-glucose condition, whereas miR-142-3p knockdown had the reverse effects, and metformin abolished those effects. Glucose 129-136 high mobility group box 1 Homo sapiens 70-75 32208365-9 2020 Metformin inhibits high glucose-induced VSMC hyperproliferation and increased migration by inducing miR-142-3p-mediated inhibition of HMGB1 expression via the HMGB1-autophagy related pathway. Metformin 0-9 high mobility group box 1 Homo sapiens 134-139 32208365-9 2020 Metformin inhibits high glucose-induced VSMC hyperproliferation and increased migration by inducing miR-142-3p-mediated inhibition of HMGB1 expression via the HMGB1-autophagy related pathway. Metformin 0-9 high mobility group box 1 Homo sapiens 159-164 32208365-9 2020 Metformin inhibits high glucose-induced VSMC hyperproliferation and increased migration by inducing miR-142-3p-mediated inhibition of HMGB1 expression via the HMGB1-autophagy related pathway. Glucose 24-31 high mobility group box 1 Homo sapiens 134-139 32208365-9 2020 Metformin inhibits high glucose-induced VSMC hyperproliferation and increased migration by inducing miR-142-3p-mediated inhibition of HMGB1 expression via the HMGB1-autophagy related pathway. Glucose 24-31 high mobility group box 1 Homo sapiens 159-164 32191778-0 2020 Retraction: Inhibition of HMGB1-Induced Angiogenesis by Cilostazol via SIRT1 Activation in Synovial Fibroblasts from Rheumatoid Arthritis. Cilostazol 56-66 high mobility group box 1 Homo sapiens 26-31 31913693-6 2020 In normal SAECs, HMGB-1 increased amiloride-sensitive Isc and ENaC Po from 0.15+-0.03 to 0.28+-0.04; p<0.01. Amiloride 34-43 high mobility group box 1 Homo sapiens 17-23 32089069-5 2020 Upregulation of miR-204 and downregulation of HMGB1 could repress TLR4/NF-kappaB pathway activation, degraded insulin release and testosterone (T) leveland ascended ovarian coefficient, boosted cell proliferation and restrained apoptosis of granulosa cells. Testosterone 130-142 high mobility group box 1 Homo sapiens 46-51 32036391-7 2020 HMGB1 can inhibit mutant huntingtin aggregation, protecting against polyglutamine-induced neurotoxicity and acting as a chaperon-like molecule, possibly via autophagy regulation. polyglutamine 68-81 high mobility group box 1 Homo sapiens 0-5 31270920-2 2020 Previous studies showed that HMGB1 is increased in the lung and circulation of patients with acute exacerbation of idiopathic pulmonary fibrosis (AE-IPF). ae-ipf 146-152 high mobility group box 1 Homo sapiens 29-34 31270920-9 2020 Higher levels of HMGB1 were associated with earlier onset of AE in stable IPF patients and with shorter survival in AE-IPF patients (P = 0.030 and 0.001, respectively). ae-ipf 116-122 high mobility group box 1 Homo sapiens 17-22 32296583-13 2020 Conclusions: Our results suggest that NETs produced by TINs mediate the crosstalk between glioma progression and the tumor microenvironment by regulating the HMGB1/RAGE/IL-8 axis. tins 55-59 high mobility group box 1 Homo sapiens 158-163 31924647-12 2020 NSG mice with human hematopoietic system injected with the HMGB1 antagonist glycyrrhizin verified the in vitro effects of HMGB1. Glycyrrhizic Acid 76-88 high mobility group box 1 Homo sapiens 59-64 31924647-12 2020 NSG mice with human hematopoietic system injected with the HMGB1 antagonist glycyrrhizin verified the in vitro effects of HMGB1. Glycyrrhizic Acid 76-88 high mobility group box 1 Homo sapiens 122-127 31642155-5 2020 Vitamin D downstream signalling has also been checked in placenta (VDR, CYP27B1, Cathelicidin LL37) along with expression of inflammatory markers (S100A8, HMGB1, TLR2, pNF-kappaB) using Western blotting and immunohistochemistry. Vitamin D 0-9 high mobility group box 1 Homo sapiens 155-160 32103987-6 2020 Results: Hypoxia enhanced the expressions of NRF-1/TFAM, boosted mtDNA copy number and ATP content and increased the number of mitochondria in pancreatic cancer cells while induction was suppressed by a knockdown of HMGB1. Adenosine Triphosphate 87-90 high mobility group box 1 Homo sapiens 216-221 32103987-9 2020 A knockdown of HMGB1 attenuated hypoxia with AICAR (an AMPK activator)-induced expression of NRF-1, TFAM, PGC-1alpha, SIRT1 and the proteins of complexes I& III and reduced the acetylation level of PGC-1alpha/SIRT1 activity. Adenosine Monophosphate 156-159 high mobility group box 1 Homo sapiens 15-20 31056833-0 2020 Neuronal HMGB1 in nucleus accumbens regulates cocaine reward memory. Cocaine 46-53 high mobility group box 1 Homo sapiens 9-14 31056833-4 2020 Here, we show that cocaine exposure induced the translocation and release of HMGB1 in the nucleus accumbens (NAc) neurons. Cocaine 19-26 high mobility group box 1 Homo sapiens 77-82 31056833-5 2020 Gain and loss of HMGB1 in the NAc bidirectionally regulate cocaine-induced conditioned place preference. Cocaine 59-66 high mobility group box 1 Homo sapiens 17-22 31056833-6 2020 From the nucleus to the cytosol, HMGB1 binds to glutamate receptor subunits (GluA2/GluN2B) on the membrane, which regulates cocaine-induced synaptic adaptation and the formation of cocaine-related memory. Cocaine 124-131 high mobility group box 1 Homo sapiens 33-38 31056833-6 2020 From the nucleus to the cytosol, HMGB1 binds to glutamate receptor subunits (GluA2/GluN2B) on the membrane, which regulates cocaine-induced synaptic adaptation and the formation of cocaine-related memory. Cocaine 181-188 high mobility group box 1 Homo sapiens 33-38 32019497-0 2020 Expression of Concern to: Redox modification of cysteine residues regulates the cytokine activity of high mobility group box-1 (HMGB1). Cysteine 48-56 high mobility group box 1 Homo sapiens 101-126 32019497-0 2020 Expression of Concern to: Redox modification of cysteine residues regulates the cytokine activity of high mobility group box-1 (HMGB1). Cysteine 48-56 high mobility group box 1 Homo sapiens 128-133 32117622-0 2020 Glycyrrhizin suppresses epithelial-mesenchymal transition by inhibiting high-mobility group box1 via the TGF-beta1/Smad2/3 pathway in lung epithelial cells. Glycyrrhizic Acid 0-12 high mobility group box 1 Homo sapiens 72-96 32117622-2 2020 Glycyrrhizin, an active component extracted from licorice plant, has been reported to treat a variety of inflammatory reactions through inhibiting high-mobility group box1 (HMGB1), which has been suggested to be a significant mediator in EMT process. Glycyrrhizic Acid 0-12 high mobility group box 1 Homo sapiens 147-171 32117622-2 2020 Glycyrrhizin, an active component extracted from licorice plant, has been reported to treat a variety of inflammatory reactions through inhibiting high-mobility group box1 (HMGB1), which has been suggested to be a significant mediator in EMT process. Glycyrrhizic Acid 0-12 high mobility group box 1 Homo sapiens 173-178 32117622-11 2020 Notably, we found that glycyrrhizin treatment effectively suppressed TGF-beta1-induced EMT process by inhibiting HMGB1. Glycyrrhizic Acid 23-35 high mobility group box 1 Homo sapiens 113-118 32117622-14 2020 Glycyrrhizin significantly blocked the phosphorylation of Smad2/3 stimulated either by TGF-beta1 or by ectopic HMGB1 in A549 and BEAS-2B cells. Glycyrrhizic Acid 0-12 high mobility group box 1 Homo sapiens 111-116 32117622-16 2020 Importantly, glycyrrhizin can effectively block Smad2/3 signaling pathway through inhibiting HMGB1, thereby suppressing the EMT progress. Glycyrrhizic Acid 13-25 high mobility group box 1 Homo sapiens 93-98 31802650-11 2020 Inhibition of miR-34a or overexpression of HMGB1 could effectively reverse elevated 5-FU sensitivity upon NEAT1 knockdown. Fluorouracil 84-88 high mobility group box 1 Homo sapiens 43-48 32175381-4 2020 However, whether curcumin could effectively inhibit inflammation through the suppression of HMGB1 production or HMGB1-mediated inflammatory responses in Abeta-activated microglia is still unclear. Curcumin 17-25 high mobility group box 1 Homo sapiens 92-97 32175381-4 2020 However, whether curcumin could effectively inhibit inflammation through the suppression of HMGB1 production or HMGB1-mediated inflammatory responses in Abeta-activated microglia is still unclear. Curcumin 17-25 high mobility group box 1 Homo sapiens 112-117 31916113-0 2020 Restoration of calcium-induced differentiation potential and tight junction formation in HaCaT keratinocytes by functional attenuation of overexpressed high mobility group box-1 protein. Calcium 15-22 high mobility group box 1 Homo sapiens 152-177 31916113-5 2020 In contrast, dipotassium glycyrrhizate (GK2), a soluble analogue of HMGB1-blocker Glycyrrhizin, down-regulated interferon-beta, a typical inflammatory cytokine induced by secreted HMGB1, and accelerated differentiation responses to the calcium treatment in these cells. glycyrrhizic acid methyl ester 13-38 high mobility group box 1 Homo sapiens 68-73 31916113-5 2020 In contrast, dipotassium glycyrrhizate (GK2), a soluble analogue of HMGB1-blocker Glycyrrhizin, down-regulated interferon-beta, a typical inflammatory cytokine induced by secreted HMGB1, and accelerated differentiation responses to the calcium treatment in these cells. glycyrrhizic acid methyl ester 13-38 high mobility group box 1 Homo sapiens 180-185 31916113-5 2020 In contrast, dipotassium glycyrrhizate (GK2), a soluble analogue of HMGB1-blocker Glycyrrhizin, down-regulated interferon-beta, a typical inflammatory cytokine induced by secreted HMGB1, and accelerated differentiation responses to the calcium treatment in these cells. Glycyrrhizic Acid 82-94 high mobility group box 1 Homo sapiens 68-73 31916113-5 2020 In contrast, dipotassium glycyrrhizate (GK2), a soluble analogue of HMGB1-blocker Glycyrrhizin, down-regulated interferon-beta, a typical inflammatory cytokine induced by secreted HMGB1, and accelerated differentiation responses to the calcium treatment in these cells. Glycyrrhizic Acid 82-94 high mobility group box 1 Homo sapiens 180-185 31916113-5 2020 In contrast, dipotassium glycyrrhizate (GK2), a soluble analogue of HMGB1-blocker Glycyrrhizin, down-regulated interferon-beta, a typical inflammatory cytokine induced by secreted HMGB1, and accelerated differentiation responses to the calcium treatment in these cells. Calcium 236-243 high mobility group box 1 Homo sapiens 68-73 32560618-0 2020 Antigene and Antiproliferative Effects of Triplex-Forming Oligonucleotide (TFO) Targeted on hmgb1 Gene in Human Hepatoma Cells. triplex-forming oligonucleotide 42-73 high mobility group box 1 Homo sapiens 92-97 31733186-7 2020 Additionally, Linagliptin inhibited AGEs-induced production of high mobility group box chromosomal protein 1 (HMGB-1), and the expression of matrix metalloproteases (MMPs)-2 and -9. linagliptin 14-25 high mobility group box 1 Homo sapiens 110-116 32096202-11 2020 Melatonin repressed the HMGB1, TNF-alpha, and iNOS mRNA expression, NO production, and nuclear factor kappaB (NF-kappaB) activation in OGD/R challenged SH SY5Y cells. Melatonin 0-9 high mobility group box 1 Homo sapiens 24-29 33308096-6 2020 Brucine also triggered cell stress and induced features of ICD, including calreticulin (CRT) exposure and high-mobility group box 1 (HMGB1) release in MDA-MB-231 and CT26 cancer cells. brucine 0-7 high mobility group box 1 Homo sapiens 106-131 33308096-6 2020 Brucine also triggered cell stress and induced features of ICD, including calreticulin (CRT) exposure and high-mobility group box 1 (HMGB1) release in MDA-MB-231 and CT26 cancer cells. brucine 0-7 high mobility group box 1 Homo sapiens 133-138 32560618-6 2020 RESULTS: Treatment of HepG2 cell with specific Triplex-forming Oligonucleotide significantly downregulated HMGB1 expression level at mRNA and protein levels by 50%, while the classical anticancer drugs, actinomycin/adriamycin as positive controls showed 65% and the combination of TFO and drug decreased by 70%. triplex-forming oligonucleotide 47-78 high mobility group box 1 Homo sapiens 107-112 32560618-6 2020 RESULTS: Treatment of HepG2 cell with specific Triplex-forming Oligonucleotide significantly downregulated HMGB1 expression level at mRNA and protein levels by 50%, while the classical anticancer drugs, actinomycin/adriamycin as positive controls showed 65% and the combination of TFO and drug decreased by 70%. Dactinomycin 203-214 high mobility group box 1 Homo sapiens 107-112 32560618-6 2020 RESULTS: Treatment of HepG2 cell with specific Triplex-forming Oligonucleotide significantly downregulated HMGB1 expression level at mRNA and protein levels by 50%, while the classical anticancer drugs, actinomycin/adriamycin as positive controls showed 65% and the combination of TFO and drug decreased by 70%. Doxorubicin 215-225 high mobility group box 1 Homo sapiens 107-112 32560618-6 2020 RESULTS: Treatment of HepG2 cell with specific Triplex-forming Oligonucleotide significantly downregulated HMGB1 expression level at mRNA and protein levels by 50%, while the classical anticancer drugs, actinomycin/adriamycin as positive controls showed 65% and the combination of TFO and drug decreased by 70%. tfo 281-284 high mobility group box 1 Homo sapiens 107-112 32560618-9 2020 CONCLUSION: Therefore, the ability of hmgb1 targeted triplex-forming oligonucleotide in combination with triplex selective anticancer drug holds promise in the treatment of malignancies associated with hmgb1 overexpression. triplex-forming oligonucleotide 53-84 high mobility group box 1 Homo sapiens 38-43 32560618-0 2020 Antigene and Antiproliferative Effects of Triplex-Forming Oligonucleotide (TFO) Targeted on hmgb1 Gene in Human Hepatoma Cells. tfo 75-78 high mobility group box 1 Homo sapiens 92-97 32560618-9 2020 CONCLUSION: Therefore, the ability of hmgb1 targeted triplex-forming oligonucleotide in combination with triplex selective anticancer drug holds promise in the treatment of malignancies associated with hmgb1 overexpression. triplex-forming oligonucleotide 53-84 high mobility group box 1 Homo sapiens 202-207 31585890-5 2020 This protective effect of simvastatin was largely due to improved lysosome function that attenuated lysosome injury-mediated Nlrp3 inflammasome activation and subsequent release of high mobility group box protein-1 (HMGB1). Simvastatin 26-37 high mobility group box 1 Homo sapiens 181-214 31676289-0 2020 MSU Crystals induce sterile IL-1beta secretion via P2X7 receptor activation and HMGB1 release. Uric Acid 0-3 high mobility group box 1 Homo sapiens 80-85 31676289-11 2020 Moreover, MSU treatment induced HMGB1 release; pre-treatment with P2X7 antagonist reduced the amount of HMGB1 in cell supernatants. Uric Acid 10-13 high mobility group box 1 Homo sapiens 32-37 31676289-11 2020 Moreover, MSU treatment induced HMGB1 release; pre-treatment with P2X7 antagonist reduced the amount of HMGB1 in cell supernatants. Uric Acid 10-13 high mobility group box 1 Homo sapiens 104-109 31676289-12 2020 CONCLUSIONS: IL-1beta secretion induced by MSU depends on P2X7 receptor activation and involves HMGB1 release. Uric Acid 43-46 high mobility group box 1 Homo sapiens 96-101 31676289-13 2020 GENERAL SIGNIFICANCE: We propose that cell activation caused by MSU crystals induces peritoneal macrophages and THP-1 cells to release ATP and HMGB1, causing IL-1beta secretion via P2X7 receptor activation. Uric Acid 64-67 high mobility group box 1 Homo sapiens 143-148 32115500-0 2020 Docetaxel Upregulates HMGB1 Levels in Non-small Cell Lung Cancer. Docetaxel 0-9 high mobility group box 1 Homo sapiens 22-27 32115500-6 2020 On the other hand, plasma levels of high-mobility group box 1 (HMGB1), a damage-associated molecular pattern (DAMP) protein, showed significant increase after docetaxel treatment. Docetaxel 159-168 high mobility group box 1 Homo sapiens 36-61 32115500-6 2020 On the other hand, plasma levels of high-mobility group box 1 (HMGB1), a damage-associated molecular pattern (DAMP) protein, showed significant increase after docetaxel treatment. Docetaxel 159-168 high mobility group box 1 Homo sapiens 63-68 32115500-9 2020 These findings imply that docetaxel could be involved in anti-tumor immunity via HMGB1. Docetaxel 26-35 high mobility group box 1 Homo sapiens 81-86 33268671-0 2020 (-)-Epigallocatechin-3-gallate inhibits RANKL-induced osteoclastogenesis via downregulation of NFATc1 and suppression of HO-1-HMGB1-RAGE pathway. epigallocatechin gallate 0-30 high mobility group box 1 Homo sapiens 126-131 33268671-8 2020 We also found that EGCG upregulated heme oxygenase-1 (HO-1) and suppressed the extracellular release of high-mobility group box 1 (HMGB1). epigallocatechin gallate 19-23 high mobility group box 1 Homo sapiens 104-129 33268671-8 2020 We also found that EGCG upregulated heme oxygenase-1 (HO-1) and suppressed the extracellular release of high-mobility group box 1 (HMGB1). epigallocatechin gallate 19-23 high mobility group box 1 Homo sapiens 131-136 33268671-9 2020 In addition, EGCG decreased the expression of the receptor for advanced glycation end products (RAGE), which is the receptor of HMGB1, in osteoclastogenesis. epigallocatechin gallate 13-17 high mobility group box 1 Homo sapiens 128-133 33268671-10 2020 In summary, our study showed that EGCG could inhibit osteoclast differentiation through the downregulation of NFATc1 and the suppression of the HO-1-HMGB1-RAGE pathway. epigallocatechin gallate 34-38 high mobility group box 1 Homo sapiens 149-154 31585890-5 2020 This protective effect of simvastatin was largely due to improved lysosome function that attenuated lysosome injury-mediated Nlrp3 inflammasome activation and subsequent release of high mobility group box protein-1 (HMGB1). Simvastatin 26-37 high mobility group box 1 Homo sapiens 216-221 31875024-4 2019 A bioinformatics analysis predicted the binding of HMGB1 by the miR-548 cluster. mir-548 64-71 high mobility group box 1 Homo sapiens 51-56 31769590-6 2020 Blockade of HMGB1 with chicken anti-HMGB1 polyclonal antibody (anti-HMGB1) or glycyrrhizin (Gly) attenuated the increase of LC3B-II and Beclin1, migration and p65 phosphorylation, suggesting the involvement of HMGB1 in autophagy, migration and NF-kappaB activation of lung macrophages. Glycyrrhizic Acid 92-95 high mobility group box 1 Homo sapiens 12-17 31897190-6 2020 The results of the current study indicated that HMGB1 was localized to the nucleus and the cytoplasm, and it was determined to combine with the condensed chromosomes of proliferating cells in paraformaldehyde (PFA)-fixed glioma tissues. paraform 192-208 high mobility group box 1 Homo sapiens 48-53 31897190-6 2020 The results of the current study indicated that HMGB1 was localized to the nucleus and the cytoplasm, and it was determined to combine with the condensed chromosomes of proliferating cells in paraformaldehyde (PFA)-fixed glioma tissues. paraform 210-213 high mobility group box 1 Homo sapiens 48-53 31897190-7 2020 However, HMGB1 was also associated with interphase (but not mitotic chromosomes) when fixed with chilled methanol and 5% (v/v) acetic acid or PFA in vitro. Methanol 105-113 high mobility group box 1 Homo sapiens 9-14 31897190-7 2020 However, HMGB1 was also associated with interphase (but not mitotic chromosomes) when fixed with chilled methanol and 5% (v/v) acetic acid or PFA in vitro. Acetic Acid 127-138 high mobility group box 1 Homo sapiens 9-14 31897190-7 2020 However, HMGB1 was also associated with interphase (but not mitotic chromosomes) when fixed with chilled methanol and 5% (v/v) acetic acid or PFA in vitro. paraform 142-145 high mobility group box 1 Homo sapiens 9-14 31806570-5 2020 Here, we found that the specific agonist of CB2R, GW405833 (GW) could induce intracellular HMGB1 degradation without influencing HMGB1 mRNA in peritoneal macrophages. 1-(2,3-dichlorobenzoyl)-5-methoxy-2-methyl-(2-(mopholin-4-yl)ethyl)-1H-indole 50-58 high mobility group box 1 Homo sapiens 91-96 31806570-5 2020 Here, we found that the specific agonist of CB2R, GW405833 (GW) could induce intracellular HMGB1 degradation without influencing HMGB1 mRNA in peritoneal macrophages. 1-(2,3-dichlorobenzoyl)-5-methoxy-2-methyl-(2-(mopholin-4-yl)ethyl)-1H-indole 50-52 high mobility group box 1 Homo sapiens 91-96 31806570-6 2020 Then we observed that autophagy inhibitor 3-methyladenine (3-MA) but not proteasome inhibitor MG-132 (MG) could block GW-induced HMGB1 degradation, which indicated that the autophagy-lysosome but not the ubiquitination pathway was involved in this process. 3-methyladenine 42-57 high mobility group box 1 Homo sapiens 129-134 31806570-6 2020 Then we observed that autophagy inhibitor 3-methyladenine (3-MA) but not proteasome inhibitor MG-132 (MG) could block GW-induced HMGB1 degradation, which indicated that the autophagy-lysosome but not the ubiquitination pathway was involved in this process. 3-methyladenine 59-63 high mobility group box 1 Homo sapiens 129-134 31806570-6 2020 Then we observed that autophagy inhibitor 3-methyladenine (3-MA) but not proteasome inhibitor MG-132 (MG) could block GW-induced HMGB1 degradation, which indicated that the autophagy-lysosome but not the ubiquitination pathway was involved in this process. 1-(2,3-dichlorobenzoyl)-5-methoxy-2-methyl-(2-(mopholin-4-yl)ethyl)-1H-indole 118-120 high mobility group box 1 Homo sapiens 129-134 31806570-8 2020 It also observed that inhibition of autophagy blocked GW-induced nuclear translocation of HMGB1 in macrophages. 1-(2,3-dichlorobenzoyl)-5-methoxy-2-methyl-(2-(mopholin-4-yl)ethyl)-1H-indole 54-56 high mobility group box 1 Homo sapiens 90-95 31897149-0 2020 Poly (ADP-ribosylation) of HMGB1 facilitates its acetylation and promotes HMGB1 translocation-associated chemotherapy-induced autophagy in leukaemia cells. poly If 0-4 high mobility group box 1 Homo sapiens 27-32 31897149-0 2020 Poly (ADP-ribosylation) of HMGB1 facilitates its acetylation and promotes HMGB1 translocation-associated chemotherapy-induced autophagy in leukaemia cells. poly If 0-4 high mobility group box 1 Homo sapiens 74-79 31897149-0 2020 Poly (ADP-ribosylation) of HMGB1 facilitates its acetylation and promotes HMGB1 translocation-associated chemotherapy-induced autophagy in leukaemia cells. Adenosine Diphosphate 6-9 high mobility group box 1 Homo sapiens 27-32 31897149-0 2020 Poly (ADP-ribosylation) of HMGB1 facilitates its acetylation and promotes HMGB1 translocation-associated chemotherapy-induced autophagy in leukaemia cells. Adenosine Diphosphate 6-9 high mobility group box 1 Homo sapiens 74-79 31897149-6 2020 It was further demonstrated that poly (ADP-ribosylation) of HMGB1 facilitates its acetylation, thereby inducing HMGB1 translocation and ultimately promoting chemotherapy-induced autophagy in leukaemic cells. poly If 33-37 high mobility group box 1 Homo sapiens 60-65 31897149-6 2020 It was further demonstrated that poly (ADP-ribosylation) of HMGB1 facilitates its acetylation, thereby inducing HMGB1 translocation and ultimately promoting chemotherapy-induced autophagy in leukaemic cells. poly If 33-37 high mobility group box 1 Homo sapiens 112-117 31897149-6 2020 It was further demonstrated that poly (ADP-ribosylation) of HMGB1 facilitates its acetylation, thereby inducing HMGB1 translocation and ultimately promoting chemotherapy-induced autophagy in leukaemic cells. Adenosine Diphosphate 39-42 high mobility group box 1 Homo sapiens 60-65 31897149-6 2020 It was further demonstrated that poly (ADP-ribosylation) of HMGB1 facilitates its acetylation, thereby inducing HMGB1 translocation and ultimately promoting chemotherapy-induced autophagy in leukaemic cells. Adenosine Diphosphate 39-42 high mobility group box 1 Homo sapiens 112-117 31905926-3 2019 Comparison of CDDP-sensitive cases with CDDP-resistant cases, the expression of c-met, HMGB1, and PCDHB9 was correlated with CDDP resistance. Cisplatin 14-18 high mobility group box 1 Homo sapiens 87-92 31905926-3 2019 Comparison of CDDP-sensitive cases with CDDP-resistant cases, the expression of c-met, HMGB1, and PCDHB9 was correlated with CDDP resistance. Cisplatin 40-44 high mobility group box 1 Homo sapiens 87-92 31905926-3 2019 Comparison of CDDP-sensitive cases with CDDP-resistant cases, the expression of c-met, HMGB1, and PCDHB9 was correlated with CDDP resistance. Cisplatin 40-44 high mobility group box 1 Homo sapiens 87-92 31905926-4 2019 Among them, the expression of HMGB1 showed the most significant correlation with CDDP resistance in multivariate analysis. Cisplatin 81-85 high mobility group box 1 Homo sapiens 30-35 31905926-5 2019 Treatment of TMK-1 and MKN74 human gastric cancer cell lines with ethyl pyruvate (EP) or tanshinone IIA (TAN), which are reported to inhibit HMGB1 signaling, showed a 4-5-fold increase in inhibition by CDDP. ethyl pyruvate 66-80 high mobility group box 1 Homo sapiens 141-146 31905926-5 2019 Treatment of TMK-1 and MKN74 human gastric cancer cell lines with ethyl pyruvate (EP) or tanshinone IIA (TAN), which are reported to inhibit HMGB1 signaling, showed a 4-5-fold increase in inhibition by CDDP. ethyl pyruvate 82-84 high mobility group box 1 Homo sapiens 141-146 31905926-5 2019 Treatment of TMK-1 and MKN74 human gastric cancer cell lines with ethyl pyruvate (EP) or tanshinone IIA (TAN), which are reported to inhibit HMGB1 signaling, showed a 4-5-fold increase in inhibition by CDDP. tanshinone 89-103 high mobility group box 1 Homo sapiens 141-146 31905926-5 2019 Treatment of TMK-1 and MKN74 human gastric cancer cell lines with ethyl pyruvate (EP) or tanshinone IIA (TAN), which are reported to inhibit HMGB1 signaling, showed a 4-5-fold increase in inhibition by CDDP. tanshinone 105-108 high mobility group box 1 Homo sapiens 141-146 31905926-6 2019 Treatment with EP or TAN also suppressed the expression of TLR4 and MyD88 in the HMGB1 signal transduction pathway and suppressed the activity of NFkappaB in both cell lines. ethyl pyruvate 15-17 high mobility group box 1 Homo sapiens 81-86 31905926-6 2019 Treatment with EP or TAN also suppressed the expression of TLR4 and MyD88 in the HMGB1 signal transduction pathway and suppressed the activity of NFkappaB in both cell lines. tanshinone 21-24 high mobility group box 1 Homo sapiens 81-86 31905926-7 2019 These results suggest that the expression of these cancer metastasis-related genes is also related to anticancer drug resistance and that suppression of HMGB1 may be particularly useful for CDDP sensitization. Cisplatin 190-194 high mobility group box 1 Homo sapiens 153-158 31875024-9 2019 Moreover, forced expression of miR-548 suppressed HMGB1 and inflammatory cytokines in hAECs, which increased when treated with lipopolysaccharide. mir-548 31-38 high mobility group box 1 Homo sapiens 50-55 31875024-10 2019 These results suggest miR-548 can alter the inflammatory responses in hAECs, and might be involved in the pathogenesis of preterm birth by regulating HMGB1. mir-548 22-29 high mobility group box 1 Homo sapiens 150-155 31810939-3 2019 Previously, we identified an anticancer candidate, papaverine, that inhibited the HMGB1-RAGE interaction. Papaverine 51-61 high mobility group box 1 Homo sapiens 82-87 31844158-4 2019 Acetylated HMGB1 (Ac-HMGB1) secreted by geminin-overexpressing cells activates RAGE and CXCR4 expression on mesenchymal stem cells (MSCs) located in tumor stroma. Actinium 0-2 high mobility group box 1 Homo sapiens 11-16 31844158-4 2019 Acetylated HMGB1 (Ac-HMGB1) secreted by geminin-overexpressing cells activates RAGE and CXCR4 expression on mesenchymal stem cells (MSCs) located in tumor stroma. Actinium 0-2 high mobility group box 1 Homo sapiens 21-26 31106593-6 2019 Furthermore, salicin inhibits IL-1beta-induced production of pro-inflammatory cytokines such as tumor necrosis factor-alpha (TNF-alpha), interleukin-6 (IL-6), and monocyte chemoattractant protein-1 (MCP-1), vascular adhesion molecules such as intercellular cell adhesion molecule-1 (iCAM-1) and vascular cell adhesion molecule 1 (VCAM-1), and high-mobility group protein 1 (HMGB-1). salicin 13-20 high mobility group box 1 Homo sapiens 374-380 30945564-8 2019 Mechanistically, we show that vildagliptin suppresses FFA-induced expression of proteins of the NLRP3 inflammasome complex, including NLRP3, ASC, p20 and HMGB-1, and mitigates FFA-induced inactivation of the AMPK pathway. Vildagliptin 30-42 high mobility group box 1 Homo sapiens 154-160 30945564-8 2019 Mechanistically, we show that vildagliptin suppresses FFA-induced expression of proteins of the NLRP3 inflammasome complex, including NLRP3, ASC, p20 and HMGB-1, and mitigates FFA-induced inactivation of the AMPK pathway. Fatty Acids, Nonesterified 54-57 high mobility group box 1 Homo sapiens 154-160 30945564-11 2019 Collectively, we conclude that the protective role of vildagliptin in endothelial cells is mediated via suppression of the AMPK-NLRP3 inflammasome-HMGB-1 axis pathway. Vildagliptin 54-66 high mobility group box 1 Homo sapiens 147-153 31666085-2 2019 Endothelial thrombomodulin (TM) promotes thrombin-induced degradation of HMGB1, and TMalpha, a recombinant human soluble TM, abolishes peripheral HMGB1-induced allodynia in mice. tmalpha 84-91 high mobility group box 1 Homo sapiens 146-151 31514535-5 2019 We found that cilostazol significantly reduced NLRP3 inflammasome activation, as well as the activity of other related and harmful factors, including oxidative stress, expression of NADPH oxidase 4 (NOX-4), thioredoxin-interacting protein (TxNIP), high mobility group box 1 (HMGB-1), interleukin 1beta (IL-1beta) and IL-18. Cilostazol 14-24 high mobility group box 1 Homo sapiens 248-273 31514535-5 2019 We found that cilostazol significantly reduced NLRP3 inflammasome activation, as well as the activity of other related and harmful factors, including oxidative stress, expression of NADPH oxidase 4 (NOX-4), thioredoxin-interacting protein (TxNIP), high mobility group box 1 (HMGB-1), interleukin 1beta (IL-1beta) and IL-18. Cilostazol 14-24 high mobility group box 1 Homo sapiens 275-281 31418997-0 2019 Long noncoding RNA XIST enhances ethanol-induced hepatic stellate cells autophagy and activation via miR-29b/HMGB1 axis. Ethanol 33-40 high mobility group box 1 Homo sapiens 109-114 31418997-12 2019 Collectively, XIST enhances ethanol-induced HSCs autophagy and activation via miR-29b/HMGB1 axis. Ethanol 28-35 high mobility group box 1 Homo sapiens 86-91 31611261-7 2019 Mechanistically, hresistin promoted HMGB1 posttranslational lysine acetylation by preserving the NAD+-dependent deacetylase sirtuin (Sirt) 1 in human macrophages. tyrosyl-lysine 60-66 high mobility group box 1 Homo sapiens 36-41 31849690-4 2019 In this pathological process, PM2.5 particles, excessive reactive oxygen species (ROS) derived from PM2.5, and certain components in PM2.5, such as ions and polyaromatic hydrocarbons (PAHs), have been implicated as potential EMT mediators that are linked to the activation of transforming growth factor beta (TGF-beta)/SMADs, NF-kappaB, growth factor (GF)/extracellular signal-regulated protein kinase (ERK), GF/phosphatidylinositol 3-kinase (PI3K)/Akt, wingless/integrated (Wnt)/beta-catenin, Notch, Hedgehog, high mobility group box B1 (HMGB1)-receptor for advanced glycation end-products (RAGE), and aryl hydrocarbon receptor (AHR) signaling cascades and to cytoskeleton rearrangement. Hydrocarbons 184-188 high mobility group box 1 Homo sapiens 511-537 31849690-4 2019 In this pathological process, PM2.5 particles, excessive reactive oxygen species (ROS) derived from PM2.5, and certain components in PM2.5, such as ions and polyaromatic hydrocarbons (PAHs), have been implicated as potential EMT mediators that are linked to the activation of transforming growth factor beta (TGF-beta)/SMADs, NF-kappaB, growth factor (GF)/extracellular signal-regulated protein kinase (ERK), GF/phosphatidylinositol 3-kinase (PI3K)/Akt, wingless/integrated (Wnt)/beta-catenin, Notch, Hedgehog, high mobility group box B1 (HMGB1)-receptor for advanced glycation end-products (RAGE), and aryl hydrocarbon receptor (AHR) signaling cascades and to cytoskeleton rearrangement. Hydrocarbons 184-188 high mobility group box 1 Homo sapiens 539-544 31570601-0 2019 HMGB1-C1q complexes regulate macrophage function by switching between leukotriene and specialized proresolving mediator biosynthesis. Leukotrienes 70-81 high mobility group box 1 Homo sapiens 0-5 31570601-6 2019 Here, we show that HMGB1 induces leukotriene production through a RAGE-dependent pathway, while HMGB1 plus C1q induces specialized proresolving lipid mediators lipoxin A4, resolvin D1, and resolvin D2 through a RAGE- and LAIR-1-dependent pathway. Leukotrienes 33-44 high mobility group box 1 Homo sapiens 19-24 31087368-3 2019 Here, we reported that high mobility group box 1 (HMGB1) was highly elevated in high glucose (HG)-treated mesangial cells, and induced the phosphorylation, nuclear translocation, and DNA binding activity of NF-kappaB via toll-like receptor 4 (TLR4). Glucose 85-92 high mobility group box 1 Homo sapiens 23-48 31087368-3 2019 Here, we reported that high mobility group box 1 (HMGB1) was highly elevated in high glucose (HG)-treated mesangial cells, and induced the phosphorylation, nuclear translocation, and DNA binding activity of NF-kappaB via toll-like receptor 4 (TLR4). Glucose 85-92 high mobility group box 1 Homo sapiens 50-55 31629951-0 2019 XIAOPI formula promotes breast cancer chemosensitivity via inhibiting CXCL1/HMGB1-mediated autophagy. xiaopi formula 0-14 high mobility group box 1 Homo sapiens 76-81 31629951-11 2019 Taken together, XIAOPI formula is a potential adjuvant drug via inhibiting CXCL1/HMGB1-mediated autophagy for breast cancer treatment with good safety. xiaopi formula 16-30 high mobility group box 1 Homo sapiens 81-86 31353785-2 2019 HMGB1 is implicated in hyperglycemia and excess glucose in trophoblast. Glucose 48-55 high mobility group box 1 Homo sapiens 0-5 31545873-0 2019 Protective effect of glycyrrhizic acid on cerebral ischemia/reperfusion injury via inhibiting HMGB1-mediated TLR4/NF-kappaB pathway. Glycyrrhizic Acid 21-38 high mobility group box 1 Homo sapiens 94-99 31545873-7 2019 Moreover, GA inhibited the expressions of high-mobility group protein box-1 (HMGB1)-mediated TLR4/NF-kappaB pathway. Glycyrrhizic Acid 10-12 high mobility group box 1 Homo sapiens 42-75 31545873-7 2019 Moreover, GA inhibited the expressions of high-mobility group protein box-1 (HMGB1)-mediated TLR4/NF-kappaB pathway. Glycyrrhizic Acid 10-12 high mobility group box 1 Homo sapiens 77-82 31545398-8 2019 The present study also demonstrated that bexarotene exerted an anti-inflammatory effect by downregulating expression of interleukin (IL)-6, IL-8, monocyte chemoattractant protein-1, and high mobility group box-1. Bexarotene 41-51 high mobility group box 1 Homo sapiens 186-211 31664305-0 2019 Upregulation of miR-129-5p increases the sensitivity to Taxol through inhibiting HMGB1-mediated cell autophagy in breast cancer MCF-7 cells. Paclitaxel 56-61 high mobility group box 1 Homo sapiens 81-86 31754297-0 2019 miR- 26a Sensitizes Melanoma Cells To Dabrafenib Via Targeting HMGB1-Dependent Autophagy Pathways. dabrafenib 38-48 high mobility group box 1 Homo sapiens 63-68 31754297-2 2019 Methods: In our study, we conducted a variety of studies, including quantitative PCR, Western blot, and autophagy and apoptosis assays to investigate the involvement of miR-26a and HMGB1 in modulation of dabrafenib sensitivity in human melanoma cell lines. dabrafenib 204-214 high mobility group box 1 Homo sapiens 181-186 31754297-3 2019 Results: Our studies revealed that the expressions of miR-26a and HMGB1 were altered in two melanoma cell lines after dabrafenib treatment. dabrafenib 118-128 high mobility group box 1 Homo sapiens 66-71 31664305-10 2019 We found that interference of HMGB1 enhanced the chemosensitivity of Taxol by inhibiting autophagy and inducing apoptosis in MCF-7 cells. Paclitaxel 69-74 high mobility group box 1 Homo sapiens 30-35 31754297-4 2019 Additionally, dabrafenib caused autophagy in melanoma and this autophagic process was regulated by miR-26a via modifying HMGB1 expression. dabrafenib 14-24 high mobility group box 1 Homo sapiens 121-126 31754297-5 2019 Furthermore, silencing HMGB1-inhibited autophagy induced by dabrafenib in melanoma cells. dabrafenib 60-70 high mobility group box 1 Homo sapiens 23-28 31664305-11 2019 Taken together, our findings suggested that miR-129-5p increased the chemosensitivity of MCF-7 cells to Taxol through suppressing autophagy and enhancing apoptosis by inhibiting HMGB1. Paclitaxel 104-109 high mobility group box 1 Homo sapiens 178-183 31754297-6 2019 Last, we verified that treatment with a miR-26a mimic and HMGB1 shRNA could increase the efficacy of dabrafenib in melanoma cells. dabrafenib 101-111 high mobility group box 1 Homo sapiens 58-63 31664305-12 2019 Using miR-129-5p/HMGB1/autophagy-based therapeutic strategies may be a potential treatment for overcoming Taxol resistance in breast cancer. Paclitaxel 106-111 high mobility group box 1 Homo sapiens 17-22 31754297-7 2019 Conclusion: Taken together, we showed that miR-26a is involved in the regulation of dabrafenib efficacy via a HMGB1-dependent autophagy pathway in melanoma cells. dabrafenib 84-94 high mobility group box 1 Homo sapiens 110-115 31695615-9 2019 The plasma levels of IL-1beta, IL-6, TNF-alpha, HMGB1, and netrin-1 were significantly reduced with the treatment of minocycline. Minocycline 117-128 high mobility group box 1 Homo sapiens 48-53 31284735-10 2019 Indeed, although WT MEF entered senescence accompanied by p21 increase, HMGB1 KO underwent apoptosis with p21 decrease by Dox treatment. Doxorubicin 122-125 high mobility group box 1 Homo sapiens 72-77 31636696-3 2019 In addition, resveratrol treatment stimulated cell surface exposure of calreticulin, HMGB1 secretion and ATP release. resveratrol 13-24 high mobility group box 1 Homo sapiens 85-90 31418171-2 2019 HMGB1 has several redox states: reduced HMGB1 recruits inflammatory cells to injured tissues forming a heterocomplex with CXCL12 and signaling via its receptor CXCR4; disulfide-containing HMGB1 binds to TLR4 and promotes inflammatory responses. Disulfides 167-176 high mobility group box 1 Homo sapiens 0-5 31418171-3 2019 Here we show that diflunisal, an aspirin-like nonsteroidal anti-inflammatory drug (NSAID) that has been in clinical use for decades, specifically inhibits in vitro and in vivo the chemotactic activity of HMGB1 at nanomolar concentrations, at least in part by binding directly to both HMGB1 and CXCL12 and disrupting their heterocomplex. Diflunisal 18-28 high mobility group box 1 Homo sapiens 204-209 31418171-3 2019 Here we show that diflunisal, an aspirin-like nonsteroidal anti-inflammatory drug (NSAID) that has been in clinical use for decades, specifically inhibits in vitro and in vivo the chemotactic activity of HMGB1 at nanomolar concentrations, at least in part by binding directly to both HMGB1 and CXCL12 and disrupting their heterocomplex. Diflunisal 18-28 high mobility group box 1 Homo sapiens 284-289 31418171-3 2019 Here we show that diflunisal, an aspirin-like nonsteroidal anti-inflammatory drug (NSAID) that has been in clinical use for decades, specifically inhibits in vitro and in vivo the chemotactic activity of HMGB1 at nanomolar concentrations, at least in part by binding directly to both HMGB1 and CXCL12 and disrupting their heterocomplex. Aspirin 33-40 high mobility group box 1 Homo sapiens 204-209 31284735-7 2019 In senescent B16-F10, SK-MEL-24, and LS174T cells treated with Dox, p21 levels were increased by persistent HMGB1 expression. Doxorubicin 63-66 high mobility group box 1 Homo sapiens 108-113 31558702-6 2019 Mechanistically, HMGB1 binding with TLR2 receptor functions in a paracrine manner to affect CD133- pancreatic cancer cells dedifferentiation via activating Hippo-YAP pathway and HIF-1alpha expression in oxygen independent manner in vitro and in vivo. Oxygen 203-209 high mobility group box 1 Homo sapiens 17-22 30998983-6 2019 Furthermore, cultured normal human melanocytes could release HMGB1 under treatment with hydrogen peroxide. Hydrogen Peroxide 88-105 high mobility group box 1 Homo sapiens 61-66 31525576-0 2019 Cucurbitacin E ameliorates lipopolysaccharide-evoked injury, inflammation and MUC5AC expression in bronchial epithelial cells by restraining the HMGB1-TLR4-NF-kappaB signaling. cucurbitacin E 0-14 high mobility group box 1 Homo sapiens 145-150 31547018-0 2019 HMGB1-triggered inflammation inhibition of notoginseng leaf triterpenes against cerebral ischemia and reperfusion injury via MAPK and NF-kappaB signaling pathways. Triterpenes 60-71 high mobility group box 1 Homo sapiens 0-5 31527653-7 2019 After ex vivo trauma and/or CHX/TNF stimulation, addition of zVAD further enhanced expression of necroptosis-related markers as well as release of PGE2 and nitric oxide, which was in line with increased cell death and subsequent release of intracellular HMGB1 and dsDNA in CHX/TNF stimulated chondrocytes. benzyloxycarbonylvalyl-alanyl-aspartyl fluoromethyl ketone 61-65 high mobility group box 1 Homo sapiens 254-259 31607859-5 2019 HMGB1 was then passed through the impaired cell membrane to upregulate the receptor for advanced glycation end-products (RAGE), nuclear factor (NF)-kappaB, and inflammatory factors such as interleukin-6 (IL-6), interleukin-1 (IL-1beta), as well as NACHT, LRR and PYD domains-containing protein 3 (NLRP3). Nalp5 protein, mouse 248-253 high mobility group box 1 Homo sapiens 0-5 31351069-7 2019 We also show that orexin A inhibits high glucose-induced expression of TxNIP, which is crucial to the activation of the NLRP3 inflammasome, as well as that of HMGB1. Glucose 41-48 high mobility group box 1 Homo sapiens 159-164 31630719-6 2019 A significant positive correlation was found both between total cholesterol levels and HMGB1 values (r=0.846, p=0.000) and between LDL cholesterol and HMBG1 values (r=0.663, p=0.001). Cholesterol 64-75 high mobility group box 1 Homo sapiens 87-92 31369761-2 2019 The aim of current study is to investigate whether melatonin would be protective in doxorubicin-induced beta (beta) cell toxicity via HMGB1/TLR2/TLR4/MAPK/NF-kB signaling pathway. Melatonin 51-60 high mobility group box 1 Homo sapiens 134-139 31369761-2 2019 The aim of current study is to investigate whether melatonin would be protective in doxorubicin-induced beta (beta) cell toxicity via HMGB1/TLR2/TLR4/MAPK/NF-kB signaling pathway. Doxorubicin 84-95 high mobility group box 1 Homo sapiens 134-139 31510019-7 2019 Co-culture blunted ethanol-induced high mobility group box protein 1 (HMGB1)-TLR responses, corresponding with reduced ethanol induction of several proinflammatory NFkappaB target genes. Ethanol 19-26 high mobility group box 1 Homo sapiens 35-68 31510019-7 2019 Co-culture blunted ethanol-induced high mobility group box protein 1 (HMGB1)-TLR responses, corresponding with reduced ethanol induction of several proinflammatory NFkappaB target genes. Ethanol 19-26 high mobility group box 1 Homo sapiens 70-75 31252267-13 2019 In conclusion, SNHG14 silencing suppressed NSCLC progression at least partly by miR-34a/HMGB1 axis in vitroand promoted NSCLC cell sensitivity to CDDP, highlighting that SNHG14 might be a potential target for NSCLC therapy. Cisplatin 146-150 high mobility group box 1 Homo sapiens 88-93 30238832-5 2019 From our findings, we hypothesize that disulfide A-box fragment binds as an anchor toward the TLR4-MD-2 but does not facilitate the TLR4 dimer formation, thereby competing with the HMGb1-binding site and preventing HMGb1-induced signaling and downstream inflammation, whereas the pro-inflammatory B-box fragment retains the MD-2 active conformation and binds to both TLR4 proteins in the complex to aid TLR4 dimer formation, which activates the intracellular signaling for downstream inflammatory pathways and cytokine release. Disulfides 39-48 high mobility group box 1 Homo sapiens 181-186 30238832-5 2019 From our findings, we hypothesize that disulfide A-box fragment binds as an anchor toward the TLR4-MD-2 but does not facilitate the TLR4 dimer formation, thereby competing with the HMGb1-binding site and preventing HMGb1-induced signaling and downstream inflammation, whereas the pro-inflammatory B-box fragment retains the MD-2 active conformation and binds to both TLR4 proteins in the complex to aid TLR4 dimer formation, which activates the intracellular signaling for downstream inflammatory pathways and cytokine release. Disulfides 39-48 high mobility group box 1 Homo sapiens 215-220 30647411-6 2019 This protective effect of NecroX-7 was mediated by inhibition of HMGB1 release and downregulation of mitochondrial oxidative stress. necrox-7 26-34 high mobility group box 1 Homo sapiens 65-70 31079281-6 2019 ROS increase the synthesis of Abeta, high-mobility group box 1(HMGB1), and S100 that interacts with RAGE to produce additional ROS resulting in enhancement of AD pathology. Reactive Oxygen Species 0-3 high mobility group box 1 Homo sapiens 37-62 31079281-6 2019 ROS increase the synthesis of Abeta, high-mobility group box 1(HMGB1), and S100 that interacts with RAGE to produce additional ROS resulting in enhancement of AD pathology. Reactive Oxygen Species 0-3 high mobility group box 1 Homo sapiens 63-68 31695540-10 2019 Serum HMGB1 levels were positively correlated with aspartate aminotransferase (rs =0.48, p<0.01, Spearman"s rank correlation coefficient) and negatively correlated with fibrinogen (rs = -0.475, p=0.011) and hemoglobin (rs = -0.465, p=0.013). Aspartic Acid 51-60 high mobility group box 1 Homo sapiens 6-11 31079281-6 2019 ROS increase the synthesis of Abeta, high-mobility group box 1(HMGB1), and S100 that interacts with RAGE to produce additional ROS resulting in enhancement of AD pathology. Reactive Oxygen Species 127-130 high mobility group box 1 Homo sapiens 37-62 31079281-6 2019 ROS increase the synthesis of Abeta, high-mobility group box 1(HMGB1), and S100 that interacts with RAGE to produce additional ROS resulting in enhancement of AD pathology. Reactive Oxygen Species 127-130 high mobility group box 1 Homo sapiens 63-68 31129968-6 2019 Ectopic expression of RIP3 promoted cisplatin-induced HepG2/DDP cells death, HMGB1 and LDH release. Cisplatin 36-45 high mobility group box 1 Homo sapiens 77-82 31146014-0 2019 Hyperbaric oxygen inhibits the HMGB1/RAGE signaling pathway by upregulating Mir-107 expression in human osteoarthritic chondrocytes. Oxygen 11-17 high mobility group box 1 Homo sapiens 31-36 31507595-3 2019 In suppressing activated T cells, it first sequesters the pro-inflammatory high mobility group Box 1 (HMGB1) protein, which facilitates its binding to the inhibitory sialic acid-binding immunoglobulin-like lectin-10 (Siglec-10) receptor. N-Acetylneuraminic Acid 166-177 high mobility group box 1 Homo sapiens 75-100 31507595-3 2019 In suppressing activated T cells, it first sequesters the pro-inflammatory high mobility group Box 1 (HMGB1) protein, which facilitates its binding to the inhibitory sialic acid-binding immunoglobulin-like lectin-10 (Siglec-10) receptor. N-Acetylneuraminic Acid 166-177 high mobility group box 1 Homo sapiens 102-107 31484595-3 2019 This study aimed to investigate the role of high mobility group box protein 1 (HMGB1) and the toll-like receptor 4 (TLR4) pathway in normal human gastric epithelial (GES-1) cells under clopidogrel exposure. Clopidogrel 185-196 high mobility group box 1 Homo sapiens 44-77 31484595-3 2019 This study aimed to investigate the role of high mobility group box protein 1 (HMGB1) and the toll-like receptor 4 (TLR4) pathway in normal human gastric epithelial (GES-1) cells under clopidogrel exposure. Clopidogrel 185-196 high mobility group box 1 Homo sapiens 79-84 31484595-5 2019 A laser scanning confocal microscope was used to determine the distribution of HMGB1 and TLR4 in clopidogrel-induced injury. Clopidogrel 97-108 high mobility group box 1 Homo sapiens 79-84 31484595-8 2019 We found the expression of HMGB1 and TLR4 in the cytoplasm increased after clopidogrel administration. Clopidogrel 75-86 high mobility group box 1 Homo sapiens 27-32 31484595-9 2019 Besides, inhibited TLR4, which is a receptor of HMGB1, prevented the injury and occludin reducing caused by clopidogrel challenge. Clopidogrel 108-119 high mobility group box 1 Homo sapiens 48-53 31484595-10 2019 Furthermore, blocking HMGB1 or TLR4 activity by ethyl pyruvate (HMGB1 inhibitor) or cli-095(TLR4 inhibitor) can partially diminish the activation of p38MAPK. ethyl pyruvate 48-62 high mobility group box 1 Homo sapiens 22-27 31484595-10 2019 Furthermore, blocking HMGB1 or TLR4 activity by ethyl pyruvate (HMGB1 inhibitor) or cli-095(TLR4 inhibitor) can partially diminish the activation of p38MAPK. ethyl pyruvate 48-62 high mobility group box 1 Homo sapiens 64-69 31484595-10 2019 Furthermore, blocking HMGB1 or TLR4 activity by ethyl pyruvate (HMGB1 inhibitor) or cli-095(TLR4 inhibitor) can partially diminish the activation of p38MAPK. ethyl 6-(N-(2-chloro-4-fluorophenyl)sulfamoyl)cyclohex-1-ene-1-carboxylate 84-91 high mobility group box 1 Homo sapiens 22-27 31484595-11 2019 Gastric mucosal damages observed by clopidogrel challenge are associated with HMGB1, TLR4, through p38MAPK signal pathway. Clopidogrel 36-47 high mobility group box 1 Homo sapiens 78-83 31507379-8 2019 In agreement, in vivo HMGB-1 blockage with glycyrrhizin, immediately after pilocarpine-induced status epilepticus (SE), reduced neuronal degeneration, reactive astrogliosis and microgliosis in the long term. Glycyrrhizic Acid 43-55 high mobility group box 1 Homo sapiens 22-28 31507379-8 2019 In agreement, in vivo HMGB-1 blockage with glycyrrhizin, immediately after pilocarpine-induced status epilepticus (SE), reduced neuronal degeneration, reactive astrogliosis and microgliosis in the long term. Pilocarpine 75-86 high mobility group box 1 Homo sapiens 22-28 31450620-4 2019 Suppression of HMGB1 with glycyrrhizin has therapeutic benefits in fibrotic diseases. Glycyrrhizic Acid 26-38 high mobility group box 1 Homo sapiens 15-20 31552388-10 2019 Furthermore, HMGB1-mediated inflammation in depressive-like behaviors may be involved in Nod-like receptor family pyrin domain containing three (NLRP3) inflammasome and proinflammatory cytokines, abnormal kynurenine pathway and imbalance between neuroprotective and neurotoxic factors. hydroxykynurenine glucoside 205-215 high mobility group box 1 Homo sapiens 13-18 31552388-11 2019 Conclusions: We found that stress-induced inflammation mediated by HMGB1 may affect motivational deficits through regulating dopamine pathway in corticostriatal neurocircuitry. Dopamine 125-133 high mobility group box 1 Homo sapiens 67-72 31450620-12 2019 TGF-beta, Smad2/3, ERK1/2, and HMGB1 were decreased in glycyrrhizin-treated keloids. Glycyrrhizic Acid 55-67 high mobility group box 1 Homo sapiens 31-36 31481891-6 2019 Raltegravir ameliorated pulmonary fibrosis by reducing the pathology score, collagen deposition, and expression of alpha-smooth muscle actin, NLRP3, HMGB1, TLR4, inhibitor of kappa B, p-NF-kappaB, HIF-1alpha, collagen I, and collagen III. Raltegravir Potassium 0-11 high mobility group box 1 Homo sapiens 149-154 31190067-7 2019 Ker/ODC conditioned medium (CM) stimulated As-HaCaT but not parental HaCaT tumorsphere formation, and this was inhibited by glycyrrhizin, an inhibitor of HMGB1, and by TAK242, an inhibitor of the HMGB1 receptor TLR4. lysylglutamylarginine 0-3 high mobility group box 1 Homo sapiens 154-159 31190067-7 2019 Ker/ODC conditioned medium (CM) stimulated As-HaCaT but not parental HaCaT tumorsphere formation, and this was inhibited by glycyrrhizin, an inhibitor of HMGB1, and by TAK242, an inhibitor of the HMGB1 receptor TLR4. lysylglutamylarginine 0-3 high mobility group box 1 Homo sapiens 196-201 31190067-7 2019 Ker/ODC conditioned medium (CM) stimulated As-HaCaT but not parental HaCaT tumorsphere formation, and this was inhibited by glycyrrhizin, an inhibitor of HMGB1, and by TAK242, an inhibitor of the HMGB1 receptor TLR4. Glycyrrhizic Acid 124-136 high mobility group box 1 Homo sapiens 154-159 31190067-7 2019 Ker/ODC conditioned medium (CM) stimulated As-HaCaT but not parental HaCaT tumorsphere formation, and this was inhibited by glycyrrhizin, an inhibitor of HMGB1, and by TAK242, an inhibitor of the HMGB1 receptor TLR4. ethyl 6-(N-(2-chloro-4-fluorophenyl)sulfamoyl)cyclohex-1-ene-1-carboxylate 168-174 high mobility group box 1 Homo sapiens 196-201 31190067-11 2019 These results show that polyamine-dependent release of HMGB1 promotes the expansion of stem cell-like subpopulations in arsenic-transformed keratinocytes while also increasing their invasiveness, suggesting that polyamines may be a potential therapeutic target for the prevention and treatment of arsenic-initiated skin cancers. Polyamines 24-33 high mobility group box 1 Homo sapiens 55-60 31190067-11 2019 These results show that polyamine-dependent release of HMGB1 promotes the expansion of stem cell-like subpopulations in arsenic-transformed keratinocytes while also increasing their invasiveness, suggesting that polyamines may be a potential therapeutic target for the prevention and treatment of arsenic-initiated skin cancers. Arsenic 120-127 high mobility group box 1 Homo sapiens 55-60 31190067-11 2019 These results show that polyamine-dependent release of HMGB1 promotes the expansion of stem cell-like subpopulations in arsenic-transformed keratinocytes while also increasing their invasiveness, suggesting that polyamines may be a potential therapeutic target for the prevention and treatment of arsenic-initiated skin cancers. Polyamines 212-222 high mobility group box 1 Homo sapiens 55-60 31078691-6 2019 As with RU486, ADX mitigated the effects of stress on HMGB1 and CD200R1. Adenylyl sulfate 15-18 high mobility group box 1 Homo sapiens 54-59 31218684-7 2019 Furthermore, higher amounts of HMGB1 was found in the plasma of BTI patients. biotinamide 64-67 high mobility group box 1 Homo sapiens 31-36 31497226-0 2019 Glycyrrhizin suppresses inflammation and cell apoptosis by inhibition of HMGB1 via p38/p-JUK signaling pathway in attenuating intervertebral disc degeneration. Glycyrrhizic Acid 0-12 high mobility group box 1 Homo sapiens 73-78 31497226-2 2019 Previous studies have shown that glycyrrhizin (GL) is a valid inhibitor of the high-mobility group box-1 gene (HMGB1) which expressed much higher in an inflammatory condition. Glycyrrhizic Acid 33-45 high mobility group box 1 Homo sapiens 79-104 31497226-2 2019 Previous studies have shown that glycyrrhizin (GL) is a valid inhibitor of the high-mobility group box-1 gene (HMGB1) which expressed much higher in an inflammatory condition. Glycyrrhizic Acid 33-45 high mobility group box 1 Homo sapiens 111-116 31497226-2 2019 Previous studies have shown that glycyrrhizin (GL) is a valid inhibitor of the high-mobility group box-1 gene (HMGB1) which expressed much higher in an inflammatory condition. Glycyrrhizic Acid 47-49 high mobility group box 1 Homo sapiens 79-104 31497226-2 2019 Previous studies have shown that glycyrrhizin (GL) is a valid inhibitor of the high-mobility group box-1 gene (HMGB1) which expressed much higher in an inflammatory condition. Glycyrrhizic Acid 47-49 high mobility group box 1 Homo sapiens 111-116 31497226-3 2019 However, it is not known whether GL protects against IDD by the inhibition of HMGB1. Glycyrrhizic Acid 33-35 high mobility group box 1 Homo sapiens 78-83 31497226-11 2019 These results suggested that GL prevented NP degradation via restraining inflammation and cell apoptosis by inhibition of HMGB1 via p38/p-JNK signaling pathway. Glycyrrhizic Acid 29-31 high mobility group box 1 Homo sapiens 122-127 30938057-7 2019 Serum HMGB1 in LN patients correlated positively to the SLE Disease Activity Index (P < 0.0001), and 24 hours urinary proteins and negatively to creatinine clearance (P < 0.001). Creatinine 145-155 high mobility group box 1 Homo sapiens 6-11 31162203-12 2019 In contrast, a second high mobility group box 1 protein wave peaking 3-6 hours after trauma in the most severely injured and physiologically deranged patients was consistently the most important predictor of outcome in our multivariable models, rendering all other predictor variables insignificant except for smaller contributions from age and sex, and of admission base excess for maximal creatinine concentration. Creatinine 391-401 high mobility group box 1 Homo sapiens 22-47 31082723-0 2019 Melatonin alleviates circadian rhythm disruption exacerbating DSS-induced colitis by inhibiting the distribution of HMGB1 in intestinal tissues. Melatonin 0-9 high mobility group box 1 Homo sapiens 116-121 31082723-3 2019 However, no exact mechanism has been illustrated among melatonin, disrupted circadian rhythm and inflammatory bowel disease, as well as regarding the effect of melatonin on HMGB1. Melatonin 160-169 high mobility group box 1 Homo sapiens 173-178 31082723-5 2019 CRD aggravated DSS-induced colitis by worsening colonic inflammation and tissue injury, as well as by enhancing HMGB1 translocation, which could be reversed by ethyl pyruvate, an HMGB1 antagonist. ethyl pyruvate 160-174 high mobility group box 1 Homo sapiens 112-117 31082723-5 2019 CRD aggravated DSS-induced colitis by worsening colonic inflammation and tissue injury, as well as by enhancing HMGB1 translocation, which could be reversed by ethyl pyruvate, an HMGB1 antagonist. ethyl pyruvate 160-174 high mobility group box 1 Homo sapiens 179-184 31082723-6 2019 Moreover, melatonin treatment attenuated these disorders and the shuttling of HMGB1 in the intestinal epithelial cells (IECs), the effect of which could be partly reversed by the melatonin antagonist luzindole. Melatonin 10-19 high mobility group box 1 Homo sapiens 78-83 31082723-6 2019 Moreover, melatonin treatment attenuated these disorders and the shuttling of HMGB1 in the intestinal epithelial cells (IECs), the effect of which could be partly reversed by the melatonin antagonist luzindole. Melatonin 179-188 high mobility group box 1 Homo sapiens 78-83 31082723-6 2019 Moreover, melatonin treatment attenuated these disorders and the shuttling of HMGB1 in the intestinal epithelial cells (IECs), the effect of which could be partly reversed by the melatonin antagonist luzindole. luzindole 200-209 high mobility group box 1 Homo sapiens 78-83 31082723-7 2019 The protective role of melatonin may be tightly related to the translocation of HMGB1 in IECs. Melatonin 23-32 high mobility group box 1 Homo sapiens 80-85 30860623-6 2019 RESULTS: In vitro PUVA treatment induced the expression of several damage-associated molecular patterns (DAMPs) by dying T cells (calreticulin, high-mobility group box-1, and to a lesser extent heat shock proteins 70 and 90), especially upon T cell activation, leading to their phagocytosis by macrophages and dendritic cells (DCs). puva 18-22 high mobility group box 1 Homo sapiens 144-169 31331356-9 2019 HMGB1-mediated AMPK, mTOR and p70S6K phosphorylation (p-AMPK, p-mTOR & p-p70S6K) were detected by Western blot. Adenosine Monophosphate 70-73 high mobility group box 1 Homo sapiens 0-5 31173265-0 2019 Ethyl pyruvate suppresses the growth, invasion and migration and induces the apoptosis of non-small cell lung cancer cells via the HMGB1/RAGE axis and the NF-kappaB/STAT3 pathway. ethyl pyruvate 0-14 high mobility group box 1 Homo sapiens 131-136 31173265-1 2019 As an inhibitor of high mobility group protein B1 (HMGB1), ethyl pyruvate (EP) has been associated with various inflammatory diseases. ethyl pyruvate 59-73 high mobility group box 1 Homo sapiens 19-49 31173265-1 2019 As an inhibitor of high mobility group protein B1 (HMGB1), ethyl pyruvate (EP) has been associated with various inflammatory diseases. ethyl pyruvate 59-73 high mobility group box 1 Homo sapiens 51-56 31331356-3 2019 Here the authors intended to identify the role of HMGB1 in Hank"s balanced salt solution (HBSS)-induced autophagy, explore NIS protein degradation through a autophagy-lysosome pathway in thyroid cancer cells and elucidate the possible molecular mechanisms. hank"s balanced salt solution 59-88 high mobility group box 1 Homo sapiens 50-55 31145943-0 2019 Inhibition of lncRNA TUG1 upregulates miR-142-3p to ameliorate myocardial injury during ischemia and reperfusion via targeting HMGB1- and Rac1-induced autophagy. mir-142-3p 38-48 high mobility group box 1 Homo sapiens 127-132 31331356-12 2019 RESULTS: HMGB1 was a critical regulator of autophagy-mediated NIS degradation in HBSS-treated FTC-133/TPC-1 cells. Nickel 62-65 high mobility group box 1 Homo sapiens 9-14 31336567-0 2019 High Mobility Group Box 1 Mediates TMAO-Induced Endothelial Dysfunction. trimethyloxamine 35-39 high mobility group box 1 Homo sapiens 0-25 31336567-4 2019 The present study tested whether TMAO associated endothelial dysfunction results via HMGB1 activation. trimethyloxamine 33-37 high mobility group box 1 Homo sapiens 85-90 31331356-3 2019 Here the authors intended to identify the role of HMGB1 in Hank"s balanced salt solution (HBSS)-induced autophagy, explore NIS protein degradation through a autophagy-lysosome pathway in thyroid cancer cells and elucidate the possible molecular mechanisms. hbss 90-94 high mobility group box 1 Homo sapiens 50-55 31331356-12 2019 RESULTS: HMGB1 was a critical regulator of autophagy-mediated NIS degradation in HBSS-treated FTC-133/TPC-1 cells. hbss 81-85 high mobility group box 1 Homo sapiens 9-14 31336567-5 2019 Biochemical and RT-PCR analysis showed that TMAO increased the HMGB1 expression in a dose-dependent manner in endothelial cells. trimethyloxamine 44-48 high mobility group box 1 Homo sapiens 63-68 31331356-14 2019 HMGB1-knockdown dramatically suppressed autophagy, NIS degradation and boosted iodide uptake in HBSS-treated cells. Nickel 51-54 high mobility group box 1 Homo sapiens 0-5 31336567-6 2019 However, prior treatment with glycyrrhizin, an HMGB1 binder, abolished the TMAO-induced HMGB1 production in endothelial cells. Glycyrrhizic Acid 30-42 high mobility group box 1 Homo sapiens 47-52 31331356-14 2019 HMGB1-knockdown dramatically suppressed autophagy, NIS degradation and boosted iodide uptake in HBSS-treated cells. Iodides 79-85 high mobility group box 1 Homo sapiens 0-5 31336567-6 2019 However, prior treatment with glycyrrhizin, an HMGB1 binder, abolished the TMAO-induced HMGB1 production in endothelial cells. Glycyrrhizic Acid 30-42 high mobility group box 1 Homo sapiens 88-93 31331356-14 2019 HMGB1-knockdown dramatically suppressed autophagy, NIS degradation and boosted iodide uptake in HBSS-treated cells. hbss 96-100 high mobility group box 1 Homo sapiens 0-5 31336567-6 2019 However, prior treatment with glycyrrhizin, an HMGB1 binder, abolished the TMAO-induced HMGB1 production in endothelial cells. trimethyloxamine 75-79 high mobility group box 1 Homo sapiens 47-52 31336567-6 2019 However, prior treatment with glycyrrhizin, an HMGB1 binder, abolished the TMAO-induced HMGB1 production in endothelial cells. trimethyloxamine 75-79 high mobility group box 1 Homo sapiens 88-93 31331356-15 2019 Moreover, HBSS enhanced ROS-sustained autophagy and promoted the cytosolic translocation of HMGB1. hbss 10-14 high mobility group box 1 Homo sapiens 92-97 31336567-11 2019 In conclusion, our results demonstrate that HMGB1 is one of the important mediators of TMAO-induced endothelial dysfunction. trimethyloxamine 87-91 high mobility group box 1 Homo sapiens 44-49 31331356-16 2019 A knockdown of HMGB1 suppressed LC3-II conversion and NIS degradation via an AMPK/mTOR-dependent signal pathway through a regulation of ROS generation, rather than ATP. Nickel 54-57 high mobility group box 1 Homo sapiens 15-20 31331356-16 2019 A knockdown of HMGB1 suppressed LC3-II conversion and NIS degradation via an AMPK/mTOR-dependent signal pathway through a regulation of ROS generation, rather than ATP. Reactive Oxygen Species 136-139 high mobility group box 1 Homo sapiens 15-20 31331356-16 2019 A knockdown of HMGB1 suppressed LC3-II conversion and NIS degradation via an AMPK/mTOR-dependent signal pathway through a regulation of ROS generation, rather than ATP. Adenosine Triphosphate 164-167 high mobility group box 1 Homo sapiens 15-20 31331356-17 2019 Furthermore, these data were further supported by our in vivo experiment of xenografts formed by HMGB1 knockdown cells reverting the uptake of 99mTcO4- as compared with control shRNA-transfected cells in hunger group. Sodium Pertechnetate Tc 99m 143-150 high mobility group box 1 Homo sapiens 97-102 31331356-18 2019 CONCLUSIONS: Acting as a critical regulator of autophagy-mediated NIS degradation via ROS/AMPK/mTOR pathway, HMGB1is a potential intervention target of radioiodine therapy in thyroid cancer. Nickel 66-69 high mobility group box 1 Homo sapiens 109-114 31331356-18 2019 CONCLUSIONS: Acting as a critical regulator of autophagy-mediated NIS degradation via ROS/AMPK/mTOR pathway, HMGB1is a potential intervention target of radioiodine therapy in thyroid cancer. Reactive Oxygen Species 86-89 high mobility group box 1 Homo sapiens 109-114 31331356-18 2019 CONCLUSIONS: Acting as a critical regulator of autophagy-mediated NIS degradation via ROS/AMPK/mTOR pathway, HMGB1is a potential intervention target of radioiodine therapy in thyroid cancer. Iodine-131 152-163 high mobility group box 1 Homo sapiens 109-114 31189308-4 2019 Bioassay results showed that these oligomers significantly reversed the multidrug resistance of MCF-7/doxorubicin (DOX) cells and increased the sensitivity of U2 OS cells to DOX by downregulating the HMGB1 expression. Doxorubicin 115-118 high mobility group box 1 Homo sapiens 200-205 31189308-4 2019 Bioassay results showed that these oligomers significantly reversed the multidrug resistance of MCF-7/doxorubicin (DOX) cells and increased the sensitivity of U2 OS cells to DOX by downregulating the HMGB1 expression. Doxorubicin 174-177 high mobility group box 1 Homo sapiens 200-205 31311167-2 2019 De-regulation of the poly(ADP-ribose) polymerase 1 (PARP1)/high-mobility group box 1 (HMGB1) signaling pathway has been proposed as an important mechanism involved in cisplatin-resistance. Cisplatin 167-176 high mobility group box 1 Homo sapiens 59-84 31324798-5 2019 beta-Lapachone-induced high mobility group box 1 (HMGB1) release activates the host TLR4/MyD88/type I interferon pathway and Batf3 dendritic cell-dependent cross-priming to bridge innate and adaptive immune responses against the tumor. beta-lapachone 0-14 high mobility group box 1 Homo sapiens 23-48 31324798-5 2019 beta-Lapachone-induced high mobility group box 1 (HMGB1) release activates the host TLR4/MyD88/type I interferon pathway and Batf3 dendritic cell-dependent cross-priming to bridge innate and adaptive immune responses against the tumor. beta-lapachone 0-14 high mobility group box 1 Homo sapiens 50-55 31311167-2 2019 De-regulation of the poly(ADP-ribose) polymerase 1 (PARP1)/high-mobility group box 1 (HMGB1) signaling pathway has been proposed as an important mechanism involved in cisplatin-resistance. Cisplatin 167-176 high mobility group box 1 Homo sapiens 86-91 31311167-8 2019 The cisplatin/Morin hydrate combination was effective in the reversal of the HepG2DR cell resistance via suppression of PARP1-mediated autophagy by regulating the HMGB1 and microtubule-associated protein 1A/1B light chain 3B (LC3) I/II. Cisplatin 4-13 high mobility group box 1 Homo sapiens 163-168 30977640-10 2019 PCB29-pQ-induced high-mobility group box 1 (HMGB1) release and subsequent binding to its receptors [toll-like receptor 2 (TLR2), TLR4, TLR9, and receptor for advanced glycation end products (RAGE)] are essential for the activation of NLRP3 inflammasome. 2,3,5-trichloro-6-phenyl-(1,4)benzoquinone 0-8 high mobility group box 1 Homo sapiens 17-42 31284269-9 2019 Our data indicate that A2E-induced upregulation of HMGB1 Caveolin-1 and HMGB1 release may relate to RPE cell senescence and play a role in the pathogenesis of AMD. N-retinylidene-N-retinylethanolamine 23-26 high mobility group box 1 Homo sapiens 51-56 31284269-9 2019 Our data indicate that A2E-induced upregulation of HMGB1 Caveolin-1 and HMGB1 release may relate to RPE cell senescence and play a role in the pathogenesis of AMD. N-retinylidene-N-retinylethanolamine 23-26 high mobility group box 1 Homo sapiens 72-77 30613954-4 2019 Methods Serum HMGB1 was quantified in Malawian nevirapine-induced hypersensitivity, Taiwanese SJS/TEN and Spanish SJS/TEN cohorts. Nevirapine 47-57 high mobility group box 1 Homo sapiens 14-19 31115516-0 2019 Fr-HMGB1 and ds-HMGB1 activate the kynurenine pathway via different mechanisms in association with depressive-like behavior. Kynurenine 35-45 high mobility group box 1 Homo sapiens 3-8 31115516-0 2019 Fr-HMGB1 and ds-HMGB1 activate the kynurenine pathway via different mechanisms in association with depressive-like behavior. Kynurenine 35-45 high mobility group box 1 Homo sapiens 16-21 31115516-2 2019 In the present study, the induction of depression via the kynurenine pathway by different redox states of HMGB1 was investigated in vivo and in vitro. Kynurenine 58-68 high mobility group box 1 Homo sapiens 106-111 31115516-5 2019 Following intracerebroventricular injection of ds- and fr-HMGB1, behavioral tests were performed, revealing the presentation of depressive-like behavior, and essential proteins in the kynurenine pathway were demonstrated to be upregulated at the mRNA level, suggesting that ds- and fr-HMGB1 contributed to the development of this behavior via the kynurenine pathway. Kynurenine 184-194 high mobility group box 1 Homo sapiens 58-63 31115516-5 2019 Following intracerebroventricular injection of ds- and fr-HMGB1, behavioral tests were performed, revealing the presentation of depressive-like behavior, and essential proteins in the kynurenine pathway were demonstrated to be upregulated at the mRNA level, suggesting that ds- and fr-HMGB1 contributed to the development of this behavior via the kynurenine pathway. Kynurenine 184-194 high mobility group box 1 Homo sapiens 285-290 31115516-5 2019 Following intracerebroventricular injection of ds- and fr-HMGB1, behavioral tests were performed, revealing the presentation of depressive-like behavior, and essential proteins in the kynurenine pathway were demonstrated to be upregulated at the mRNA level, suggesting that ds- and fr-HMGB1 contributed to the development of this behavior via the kynurenine pathway. Kynurenine 347-357 high mobility group box 1 Homo sapiens 58-63 31115516-6 2019 ds-HMGB1 directly activated the kynurenine pathway and cytokines such as tumor necrosis factor-alpha (TNF-alpha) and interleukin-1beta (IL-1beta) in the hippocampal tissue. Kynurenine 32-42 high mobility group box 1 Homo sapiens 3-8 31115516-7 2019 Conversely, fr-HMGB1 upregulated the aforementioned factors only following treatment with H2O2. Hydrogen Peroxide 90-94 high mobility group box 1 Homo sapiens 15-20 31115516-8 2019 These findings indicated that ds-HMGB1 induced depression in a manner associated with the kynurenine pathway, whereas oxidation of fr-HMGB1 evoked activation of the kynurenine pathway, resulting in depressive behavior. Kynurenine 90-100 high mobility group box 1 Homo sapiens 33-38 31115516-8 2019 These findings indicated that ds-HMGB1 induced depression in a manner associated with the kynurenine pathway, whereas oxidation of fr-HMGB1 evoked activation of the kynurenine pathway, resulting in depressive behavior. Kynurenine 165-175 high mobility group box 1 Homo sapiens 134-139 31441407-0 2019 [Effect of unfractionated heparin on high mobility group box 1-mediated expression of vascular endothelial cadherin]. Heparin 26-33 high mobility group box 1 Homo sapiens 37-62 31441407-1 2019 OBJECTIVE: To observe the damage of high mobility group box 1 (HMGB1) on human umbilical vein endothelial cell (HUVEC) barrier permeability and the protective effect of unfractionated heparin (UFH), and to explore the down-regulated protection effect mechanism of UFH on HMGB1-mediated vascular endothelial cadherin (VE-cadherin) expression. Heparin 264-267 high mobility group box 1 Homo sapiens 36-61 31441407-1 2019 OBJECTIVE: To observe the damage of high mobility group box 1 (HMGB1) on human umbilical vein endothelial cell (HUVEC) barrier permeability and the protective effect of unfractionated heparin (UFH), and to explore the down-regulated protection effect mechanism of UFH on HMGB1-mediated vascular endothelial cadherin (VE-cadherin) expression. Heparin 264-267 high mobility group box 1 Homo sapiens 63-68 31298304-11 2019 RESULTS: In the presence of high glucose, hRECs cells proliferation was significantly reduced, Caspase-3 activity was enhanced, LDH and ROS levels were increased, SOD activity was declined with increased expression of HMGB-1, NF-kappaB, VEGF, as well as secretion of TNF-alpha and IL-1beta compared with control group (p < 0.05). Glucose 33-40 high mobility group box 1 Homo sapiens 218-224 31298304-11 2019 RESULTS: In the presence of high glucose, hRECs cells proliferation was significantly reduced, Caspase-3 activity was enhanced, LDH and ROS levels were increased, SOD activity was declined with increased expression of HMGB-1, NF-kappaB, VEGF, as well as secretion of TNF-alpha and IL-1beta compared with control group (p < 0.05). ros 136-139 high mobility group box 1 Homo sapiens 218-224 30946496-5 2019 HMGB1 function is also regulated by the redox status of critical cysteine residues within the protein, and is cell-type dependent. Cysteine 65-73 high mobility group box 1 Homo sapiens 0-5 31247032-13 2019 MiR-19 only regulates the proliferation of HASM cells induced by HMGB1, but not PDGF, EGF, TGF-beta1. mir-19 0-6 high mobility group box 1 Homo sapiens 65-70 31247032-14 2019 Furthermore, we demonstrated that miR-19 contributed to the promoting effects of HMGB1 on ASM cells by targeting PTEN 3"-UTR. mir-19 34-40 high mobility group box 1 Homo sapiens 81-86 31333815-4 2019 Here, we used multi-microsecond molecular dynamics (MD) simulations to elucidate the effect of the disulfide bond on the structure and dynamics of HMGB1. Disulfides 99-108 high mobility group box 1 Homo sapiens 147-152 31333815-5 2019 The results of the MD simulations show that the presence or lack of the disulfide bond between Cys23 and Cys45 modulates the conformational space explored by HMGB1, making the reduced protein more suitable to form a complex with CXCL12. Disulfides 72-81 high mobility group box 1 Homo sapiens 158-163 30942231-7 2019 For this, we employ a three-dimensional colon cancer spheroid model and show for the first time that the treatment with KP1339, a ruthenium-based complex, triggers an ICD signature hallmarked by phosphorylation of PERK and eIF2alpha, exposure of calreticulin on the cell membrane, release of high mobility group box 1 and secretion of ATP. Ruthenium 130-139 high mobility group box 1 Homo sapiens 292-317 30977640-10 2019 PCB29-pQ-induced high-mobility group box 1 (HMGB1) release and subsequent binding to its receptors [toll-like receptor 2 (TLR2), TLR4, TLR9, and receptor for advanced glycation end products (RAGE)] are essential for the activation of NLRP3 inflammasome. 2,3,5-trichloro-6-phenyl-(1,4)benzoquinone 0-8 high mobility group box 1 Homo sapiens 44-49 30956264-0 2019 Triptolide Suppresses Growth of Breast Cancer by Targeting HMGB1 in Vitro and in Vivo. triptolide 0-10 high mobility group box 1 Homo sapiens 59-64 31281467-5 2019 We found that after adjusting for other covariates, NSCLC patients who smoked and who respectively had CG, CT, and TC heterozygotes of HMGB1 rs2249825, rs1045411, and rs1360485, were at lower risk of developing mutant EGFR, compared to those patients with wild-type homozygotes. cysteinylglycine 103-105 high mobility group box 1 Homo sapiens 135-140 31281467-5 2019 We found that after adjusting for other covariates, NSCLC patients who smoked and who respectively had CG, CT, and TC heterozygotes of HMGB1 rs2249825, rs1045411, and rs1360485, were at lower risk of developing mutant EGFR, compared to those patients with wild-type homozygotes. Carbon Tetrachloride 107-109 high mobility group box 1 Homo sapiens 135-140 31281467-5 2019 We found that after adjusting for other covariates, NSCLC patients who smoked and who respectively had CG, CT, and TC heterozygotes of HMGB1 rs2249825, rs1045411, and rs1360485, were at lower risk of developing mutant EGFR, compared to those patients with wild-type homozygotes. Technetium 115-117 high mobility group box 1 Homo sapiens 135-140 31281467-6 2019 Moreover, significant inverse associations between the CG and CG + GG genotypes of HMGB1 rs2249825 and the EGFR hotspot mutation, an exon 19 in-frame deletion, were also observed among NSCLC patients. cysteinylglycine 55-57 high mobility group box 1 Homo sapiens 83-88 31281467-6 2019 Moreover, significant inverse associations between the CG and CG + GG genotypes of HMGB1 rs2249825 and the EGFR hotspot mutation, an exon 19 in-frame deletion, were also observed among NSCLC patients. cysteinylglycine 62-64 high mobility group box 1 Homo sapiens 83-88 30956264-6 2019 Further studies evidenced that triptolide treatment not only inhibited HMGB1 mRNA expression, but also decreased supernatant level of HMGB1 in vitro. triptolide 31-41 high mobility group box 1 Homo sapiens 134-139 30956264-3 2019 The therapeutic effect of triptolide on HMGB1 has not been reported. triptolide 26-36 high mobility group box 1 Homo sapiens 40-45 30956264-8 2019 As well, triptolide enhanced the anti-proliferative activity of ethyl pyruvate (EP) (HMGB1 inhibitor). triptolide 9-19 high mobility group box 1 Homo sapiens 85-90 30956264-8 2019 As well, triptolide enhanced the anti-proliferative activity of ethyl pyruvate (EP) (HMGB1 inhibitor). ethyl pyruvate 64-78 high mobility group box 1 Homo sapiens 85-90 30956264-4 2019 Thus, our study aims to clarify the role of HMGB1 in triptolide-induced anti-growth effect on breast cancer in vitro and in vivo. triptolide 53-63 high mobility group box 1 Homo sapiens 44-49 30956264-9 2019 Furthermore, downstream correlation factors (Toll-like receptor 4 (TLR4) and phosphorylated-nuclear factor-kappaB (NF-kappaB) p65) of HMGB1 were significantly decreased in vitro after triptolide treatment. triptolide 184-194 high mobility group box 1 Homo sapiens 134-139 30956264-6 2019 Further studies evidenced that triptolide treatment not only inhibited HMGB1 mRNA expression, but also decreased supernatant level of HMGB1 in vitro. triptolide 31-41 high mobility group box 1 Homo sapiens 71-76 30956264-11 2019 Meanwhile, immunohistochemical analyses showed that triptolide treatment significantly decreased the level of cytoplasmic HMGB1 and TLR4 expression, whereas the expression of NF-kappaB p65 was relatively higher in cytoplasm, and conversely lower in nucleus as compared to the control group. triptolide 52-62 high mobility group box 1 Homo sapiens 122-127 30958608-0 2019 Dexmedetomidine protects against high mobility group box 1-induced cellular injury by inhibiting pyroptosis. Dexmedetomidine 0-15 high mobility group box 1 Homo sapiens 33-58 30956264-12 2019 Collectively, these results demonstrate that triptolide suppresses the growth of breast cancer cells via reduction of HMGB1 expression in vitro and in vivo, which may provide new insights into the treament of breast cancer. triptolide 45-55 high mobility group box 1 Homo sapiens 118-123 30853521-0 2019 N4-acetylcytidine is required for sustained NLRP3 inflammasome activation via HMGB1 pathway in microglia. N-acetylcytidine 0-17 high mobility group box 1 Homo sapiens 78-83 30853521-3 2019 Here we reported that N4-acetylcytidine (N4A), a nucleoside metabolite, activated microglia and sustained NLRP3 inflammasome activation by inducing HMGB1 signaling. N-acetylcytidine 22-39 high mobility group box 1 Homo sapiens 148-153 30958608-6 2019 We found that DEX protected against HMGB1-induced cell death of BMDMs. Dexmedetomidine 14-17 high mobility group box 1 Homo sapiens 36-41 30958608-9 2019 Taken together, these findings demonstrate the protective effect of DEX in mediating HMGB1-induced cellular injury, thus indicating that DEX may be a potential therapeutic candidate for the management of infection and trauma-derived inflammation. Dexmedetomidine 68-71 high mobility group box 1 Homo sapiens 85-90 30853521-3 2019 Here we reported that N4-acetylcytidine (N4A), a nucleoside metabolite, activated microglia and sustained NLRP3 inflammasome activation by inducing HMGB1 signaling. CHEMBL3099812 41-44 high mobility group box 1 Homo sapiens 148-153 30958608-9 2019 Taken together, these findings demonstrate the protective effect of DEX in mediating HMGB1-induced cellular injury, thus indicating that DEX may be a potential therapeutic candidate for the management of infection and trauma-derived inflammation. Dexmedetomidine 137-140 high mobility group box 1 Homo sapiens 85-90 30742880-8 2019 HMGB1 and p53 both serve as transcription factors that play crucial roles in the regulation of PCB29-pQ-induced autophagy and apoptosis. 2,3,5-trichloro-6-phenyl-(1,4)benzoquinone 95-103 high mobility group box 1 Homo sapiens 0-5 30370641-5 2019 Bleomycin (BLM)-induced PF in Nrf2-knockout (Nrf2-/- ) and wild-type (WT) mice and transforming growth factor beta1 (TGF-beta1)-induced EMT in rat type II alveolar epithelial cell line (RLE-6TN) and human alveolar epithelial cell line (A549) were established to observe the relationship among Nrf2, HMGB1, and EMT by western blot and immunohistochemistry. Bleomycin 0-9 high mobility group box 1 Homo sapiens 299-304 30742880-0 2019 Polychlorinated biphenyl quinone-induced signaling transition from autophagy to apoptosis is regulated by HMGB1 and p53 in human hepatoma HepG2 cells. polychlorinated biphenyl quinone 0-32 high mobility group box 1 Homo sapiens 106-111 31177924-0 2019 Indomethacin down-regulating HMGB1 and TNF-alpha to prevent pancreatitis after endoscopic retrograde cholangiopancreatography. Indomethacin 0-12 high mobility group box 1 Homo sapiens 29-34 31177924-11 2019 The levels of HMGB1 and TNF-alpha were significantly decreased in indomethacin group at 3 (p < .0001) and 24 h (p < .0001) after ERCP, compared to the control group. Indomethacin 66-78 high mobility group box 1 Homo sapiens 14-19 31177924-13 2019 Conclusions: Rectal indomethacin could significantly reduce the risk of PEP by down-regulating the levels of HMGB1 and TNF-alpha. Indomethacin 20-32 high mobility group box 1 Homo sapiens 109-114 31100066-3 2019 Previously, we identified an inhibitor of HMGB1/RAGE interaction, papaverine (a non-narcotic opium alkaloid), using a unique drug design system and drug repositioning approach. Papaverine 66-76 high mobility group box 1 Homo sapiens 42-47 31100066-3 2019 Previously, we identified an inhibitor of HMGB1/RAGE interaction, papaverine (a non-narcotic opium alkaloid), using a unique drug design system and drug repositioning approach. Opiate Alkaloids 93-107 high mobility group box 1 Homo sapiens 42-47 31100066-5 2019 HMGB1 supplementation in the culture medium promoted tumor cell growth in T98G cells, and this effect was canceled by papaverine. Papaverine 118-128 high mobility group box 1 Homo sapiens 0-5 31100066-7 2019 As an HMGB1/RAGE inhibitor, papaverine also significantly inhibited cell proliferation in U87MG and T98G cells. Papaverine 28-38 high mobility group box 1 Homo sapiens 6-11 30742880-9 2019 PCB29-pQ not only enhanced the expression of HMGB1 and p53 but also promoted their binding and cytosolic translocation. 2,3,5-trichloro-6-phenyl-(1,4)benzoquinone 0-8 high mobility group box 1 Homo sapiens 45-50 30742880-10 2019 Interestingly, HMGB1 rather than p53 plays a primary role in 5 muM of PCB29-pQ-induced autophagy in the nucleus; however, p53 promoted apoptosis to a great extent in the cytosol at the dose of 15 muM PCB29-pQ. 2,3,5-trichloro-6-phenyl-(1,4)benzoquinone 70-78 high mobility group box 1 Homo sapiens 15-20 30742880-10 2019 Interestingly, HMGB1 rather than p53 plays a primary role in 5 muM of PCB29-pQ-induced autophagy in the nucleus; however, p53 promoted apoptosis to a great extent in the cytosol at the dose of 15 muM PCB29-pQ. 2,3,5-trichloro-6-phenyl-(1,4)benzoquinone 200-208 high mobility group box 1 Homo sapiens 15-20 30742880-11 2019 Together, HMGB1 and p53 provided a subtle balance between autophagy and apoptosis, thus determining the fate of PCB29-pQ-treated cells. 2,3,5-trichloro-6-phenyl-(1,4)benzoquinone 112-120 high mobility group box 1 Homo sapiens 10-15 30990031-3 2019 Quantitative mass spectrometry results unambiguously revealed the predominant binding of HMGB1 and HMGB2, the well-established specific binders of 1,2-cisplatin-cross-linked DNA, to the cisplatin-cross-linked ODN, thus validating the accuracy and reliability of our strategy. 1,2-cisplatin 147-160 high mobility group box 1 Homo sapiens 89-94 31123414-0 2019 Downregulation of MALAT1 alleviates saturated fatty acid-induced myocardial inflammatory injury via the miR-26a/HMGB1/TLR4/NF-kappaB axis. Fatty Acids 36-56 high mobility group box 1 Homo sapiens 112-117 30990031-3 2019 Quantitative mass spectrometry results unambiguously revealed the predominant binding of HMGB1 and HMGB2, the well-established specific binders of 1,2-cisplatin-cross-linked DNA, to the cisplatin-cross-linked ODN, thus validating the accuracy and reliability of our strategy. Cisplatin 151-160 high mobility group box 1 Homo sapiens 89-94 30712242-0 2019 Cytosolic HMGB1 Mediates Autophagy Activation in an Emulsified Isoflurane Anesthesia Cell Model. Isoflurane 63-73 high mobility group box 1 Homo sapiens 10-15 30767669-12 2019 NEW & NOTEWORTHY This study shows for the first time to our knowledge that the plasma level of high mobility group box 1 (HMGB1) is elevated in hyperhomocysteinemia (HHcy) patients, and homocysteine promotes expression and secretion of HMGB1 partially regulated by neuropilin-1 in endothelial cells, which is involved in endothelial inflammation. Adenosine Monophosphate 5-8 high mobility group box 1 Homo sapiens 99-124 30767669-12 2019 NEW & NOTEWORTHY This study shows for the first time to our knowledge that the plasma level of high mobility group box 1 (HMGB1) is elevated in hyperhomocysteinemia (HHcy) patients, and homocysteine promotes expression and secretion of HMGB1 partially regulated by neuropilin-1 in endothelial cells, which is involved in endothelial inflammation. Adenosine Monophosphate 5-8 high mobility group box 1 Homo sapiens 126-131 30767669-12 2019 NEW & NOTEWORTHY This study shows for the first time to our knowledge that the plasma level of high mobility group box 1 (HMGB1) is elevated in hyperhomocysteinemia (HHcy) patients, and homocysteine promotes expression and secretion of HMGB1 partially regulated by neuropilin-1 in endothelial cells, which is involved in endothelial inflammation. Homocysteine 153-165 high mobility group box 1 Homo sapiens 99-124 30767669-12 2019 NEW & NOTEWORTHY This study shows for the first time to our knowledge that the plasma level of high mobility group box 1 (HMGB1) is elevated in hyperhomocysteinemia (HHcy) patients, and homocysteine promotes expression and secretion of HMGB1 partially regulated by neuropilin-1 in endothelial cells, which is involved in endothelial inflammation. Homocysteine 153-165 high mobility group box 1 Homo sapiens 126-131 30822402-2 2019 KC1 and KC3, but not KC2, significantly reduced HMGB1 release in lipopolysaccharide (LPS)-activated human umbilical vein endothelial cells (HUVECs) and attenuated the cecal ligation and puncture (CLP)-induced release of HMGB1. kc1 0-3 high mobility group box 1 Homo sapiens 48-53 30822402-2 2019 KC1 and KC3, but not KC2, significantly reduced HMGB1 release in lipopolysaccharide (LPS)-activated human umbilical vein endothelial cells (HUVECs) and attenuated the cecal ligation and puncture (CLP)-induced release of HMGB1. kc1 0-3 high mobility group box 1 Homo sapiens 220-225 30822402-2 2019 KC1 and KC3, but not KC2, significantly reduced HMGB1 release in lipopolysaccharide (LPS)-activated human umbilical vein endothelial cells (HUVECs) and attenuated the cecal ligation and puncture (CLP)-induced release of HMGB1. UNII-426P9HSR8I 8-11 high mobility group box 1 Homo sapiens 48-53 30822402-2 2019 KC1 and KC3, but not KC2, significantly reduced HMGB1 release in lipopolysaccharide (LPS)-activated human umbilical vein endothelial cells (HUVECs) and attenuated the cecal ligation and puncture (CLP)-induced release of HMGB1. UNII-426P9HSR8I 8-11 high mobility group box 1 Homo sapiens 220-225 30712242-4 2019 However, the effect of intracellular HMGB1 during emulsified isoflurane (EI) exposure is poorly understood. Isoflurane 61-71 high mobility group box 1 Homo sapiens 37-42 30712242-4 2019 However, the effect of intracellular HMGB1 during emulsified isoflurane (EI) exposure is poorly understood. aziridine 73-75 high mobility group box 1 Homo sapiens 37-42 31040377-8 2019 Finally, blocking the receptor for advanced glycation end-products, a canonical HMGB1 receptor, inhibited HMGB1-induced PGE2 production and hair shaft elongation. Dinoprostone 120-124 high mobility group box 1 Homo sapiens 80-85 30875490-6 2019 Further, a small dose of TMAO significantly increased TF expression and nuclear translocation of NF-kappaB with the synergistic action of low-dose of pro-atherosclerotic factors, such as TNF-alpha and HMGB1. trimethyloxamine 25-29 high mobility group box 1 Homo sapiens 201-206 31040377-0 2019 HMGB1 promotes hair growth via the modulation of prostaglandin metabolism. Prostaglandins 49-62 high mobility group box 1 Homo sapiens 0-5 31040377-7 2019 HMGB1 also stimulated prostaglandin E2 (PGE2) secretion from hDPCs. Dinoprostone 22-38 high mobility group box 1 Homo sapiens 0-5 31040377-7 2019 HMGB1 also stimulated prostaglandin E2 (PGE2) secretion from hDPCs. Dinoprostone 40-44 high mobility group box 1 Homo sapiens 0-5 31040377-8 2019 Finally, blocking the receptor for advanced glycation end-products, a canonical HMGB1 receptor, inhibited HMGB1-induced PGE2 production and hair shaft elongation. Dinoprostone 120-124 high mobility group box 1 Homo sapiens 106-111 31040377-9 2019 Our results suggest that HMGB1 promotes hair growth via PGE2 secretion from hDPCs. Dinoprostone 56-60 high mobility group box 1 Homo sapiens 25-30 31105713-2 2019 The dipotassium glycyrrhizate (DPG), a salt of the glycoconjugated triterpene glycyrrhizin, has been shown to inhibit the High Mobility Group Box 1 (HMGB1) protein, an allarmin strongly implicated in the pathogenesis of most inflammatory and auto-immune disorders. Glycyrrhizic Acid 4-29 high mobility group box 1 Homo sapiens 122-147 31105713-2 2019 The dipotassium glycyrrhizate (DPG), a salt of the glycoconjugated triterpene glycyrrhizin, has been shown to inhibit the High Mobility Group Box 1 (HMGB1) protein, an allarmin strongly implicated in the pathogenesis of most inflammatory and auto-immune disorders. Glycyrrhizic Acid 4-29 high mobility group box 1 Homo sapiens 149-154 31105713-2 2019 The dipotassium glycyrrhizate (DPG), a salt of the glycoconjugated triterpene glycyrrhizin, has been shown to inhibit the High Mobility Group Box 1 (HMGB1) protein, an allarmin strongly implicated in the pathogenesis of most inflammatory and auto-immune disorders. Glycyrrhizic Acid 31-34 high mobility group box 1 Homo sapiens 122-147 31105713-2 2019 The dipotassium glycyrrhizate (DPG), a salt of the glycoconjugated triterpene glycyrrhizin, has been shown to inhibit the High Mobility Group Box 1 (HMGB1) protein, an allarmin strongly implicated in the pathogenesis of most inflammatory and auto-immune disorders. Glycyrrhizic Acid 31-34 high mobility group box 1 Homo sapiens 149-154 31105713-2 2019 The dipotassium glycyrrhizate (DPG), a salt of the glycoconjugated triterpene glycyrrhizin, has been shown to inhibit the High Mobility Group Box 1 (HMGB1) protein, an allarmin strongly implicated in the pathogenesis of most inflammatory and auto-immune disorders. Salts 39-43 high mobility group box 1 Homo sapiens 122-147 31105713-2 2019 The dipotassium glycyrrhizate (DPG), a salt of the glycoconjugated triterpene glycyrrhizin, has been shown to inhibit the High Mobility Group Box 1 (HMGB1) protein, an allarmin strongly implicated in the pathogenesis of most inflammatory and auto-immune disorders. Salts 39-43 high mobility group box 1 Homo sapiens 149-154 31105713-2 2019 The dipotassium glycyrrhizate (DPG), a salt of the glycoconjugated triterpene glycyrrhizin, has been shown to inhibit the High Mobility Group Box 1 (HMGB1) protein, an allarmin strongly implicated in the pathogenesis of most inflammatory and auto-immune disorders. Triterpenes 67-77 high mobility group box 1 Homo sapiens 122-147 31105713-2 2019 The dipotassium glycyrrhizate (DPG), a salt of the glycoconjugated triterpene glycyrrhizin, has been shown to inhibit the High Mobility Group Box 1 (HMGB1) protein, an allarmin strongly implicated in the pathogenesis of most inflammatory and auto-immune disorders. Triterpenes 67-77 high mobility group box 1 Homo sapiens 149-154 30975096-7 2019 METHODS: Endocytosis was studied in cultured macrophages using Alexa Fluor-labeled HMGB1 or complexes of HMGB1 and Alexa Fluor-labeled LPS in the presence of an anti-HMGB1 monoclonal antibody (mAb), recombinant HMGB1 box A protein, acetylcholine, the nicotinic acetylcholine receptor subtype alpha 7 (alpha7 nAChR) agonist GTS-21, or a dynamin-specific inhibitor of endocytosis. alexa fluor 63-74 high mobility group box 1 Homo sapiens 83-88 31114162-0 2019 The molecular mechanisms of Aloin induce gastric cancer cells apoptosis by targeting High Mobility Group Box 1. alloin 28-33 high mobility group box 1 Homo sapiens 85-110 31114162-4 2019 We investigated the specific molecular mechanisms by which ALO-induced apoptosis by targeting HMGB1 in gastric cancer cells. alloin 59-62 high mobility group box 1 Homo sapiens 94-99 31114162-13 2019 Blocking these signalling pathways by special inhibitors and HMGB1 knockdown could enhance ALO-induced HGC-27 cell apoptosis. alloin 91-94 high mobility group box 1 Homo sapiens 61-66 31114162-14 2019 Conclusion: ALO- induced HGC-27 cell apoptosis by down-regulating expressions of HMGB1 and RAGE, inhibiting HMGB1 release and then suppressing rhHMGB1-induced activation of Akt-mTOR-P70S6K and ERK-P90RSK-CREB signalling pathways. alloin 12-15 high mobility group box 1 Homo sapiens 81-86 31114162-14 2019 Conclusion: ALO- induced HGC-27 cell apoptosis by down-regulating expressions of HMGB1 and RAGE, inhibiting HMGB1 release and then suppressing rhHMGB1-induced activation of Akt-mTOR-P70S6K and ERK-P90RSK-CREB signalling pathways. alloin 12-15 high mobility group box 1 Homo sapiens 108-113 31105821-6 2019 Notably, our findings indicate that sitagliptin possesses an anti-inflammatory effect against H/R-induced expression of IL-6, IL-8, and TNF-alpha as well as secretion of HMGB1. Sitagliptin Phosphate 36-47 high mobility group box 1 Homo sapiens 170-175 30936310-0 2019 Correction for Bandala-Sanchez et al., CD52 glycan binds the proinflammatory B box of HMGB1 to engage the Siglec-10 receptor and suppress human T cell function. Polysaccharides 44-50 high mobility group box 1 Homo sapiens 86-91 30970518-4 2019 In two AML cell lines and primary AML cells from two patients, etoposide induced necroptosis via cIAP1/2 degradation when caspase activity was inhibited by Z-VAD-fmk, but treatment with extracellular HMGB1 prevented this necroptosis. Etoposide 63-72 high mobility group box 1 Homo sapiens 200-205 31105999-0 2019 HMGB1 regulates erastin-induced ferroptosis via RAS-JNK/p38 signaling in HL-60/NRASQ61L cells. erastin 16-23 high mobility group box 1 Homo sapiens 0-5 31105999-5 2019 Erastin enhanced ROS levels, thereby promoting cytosolic translocation of HMGB1 and enhancing cell death. erastin 0-7 high mobility group box 1 Homo sapiens 74-79 31105999-5 2019 Erastin enhanced ROS levels, thereby promoting cytosolic translocation of HMGB1 and enhancing cell death. ros 17-20 high mobility group box 1 Homo sapiens 74-79 31105999-6 2019 Knockdown of HMGB1 decreased erastin-induced ROS generation and cell death in an iron-mediated lysosomal pathway in HL-60/NRASQ61L cells. ros 45-48 high mobility group box 1 Homo sapiens 13-18 31105999-6 2019 Knockdown of HMGB1 decreased erastin-induced ROS generation and cell death in an iron-mediated lysosomal pathway in HL-60/NRASQ61L cells. Iron 81-85 high mobility group box 1 Homo sapiens 13-18 30832473-0 2019 Glycyrrhizin Attenuates the Process of Epithelial-to-Mesenchymal Transition by Modulating HMGB1 Initiated Novel Signaling Pathway in Prostate Cancer Cells. Glycyrrhizic Acid 0-12 high mobility group box 1 Homo sapiens 90-95 30784882-5 2019 More importantly, cRGD target liposomes of the thymidine conjugate markedly promoted the early detection of immunogenic cell death markers including ATP, HMGB1 and calreticulin. Thymidine 47-56 high mobility group box 1 Homo sapiens 154-159 30871870-4 2019 In NCM460 cells, under the normal glucose state, proliferation increased by overexpression of HMGB1. Glucose 34-41 high mobility group box 1 Homo sapiens 94-99 30871870-5 2019 Under a high glucose state, increased expression of HMGB1 was accompanied with a release from cell nuclei into the cytoplasm and extracellular matrix. Glucose 13-20 high mobility group box 1 Homo sapiens 52-57 30871870-7 2019 And overexpression of HMGB1 reversed the suppressing effect of butyrate on abnormally increased proliferation. Butyrates 63-71 high mobility group box 1 Homo sapiens 22-27 30871870-8 2019 Conclusively, butyrate suppressed the abnormally increased proliferation in colonic epithelial cells under diabetic state by targeting HMGB1. Butyrates 14-22 high mobility group box 1 Homo sapiens 135-140 30306713-0 2019 High Efficiency Removal of Cytokines and HMGB-1 by Continuous Hemofiltration With a Dual Layered Polyethersulfone Membrane: An Ex Vivo Study. polyether sulfone 97-113 high mobility group box 1 Homo sapiens 41-47 30306713-2 2019 While hemofiltration has been shown to remove cytokines, removal of cytokines and HMGB-1 by hemofiltration using a polyethersulfone membrane has not been reported. polyether sulfone 115-131 high mobility group box 1 Homo sapiens 82-88 30306713-11 2019 Continuous hemofiltration with a polyethersulfone membrane achieved high efficiency removal of cytokines and HMGB-1, without excessive removal of albumin. polyether sulfone 33-49 high mobility group box 1 Homo sapiens 109-115 30871870-0 2019 Butyrate suppresses abnormal proliferation in colonic epithelial cells under diabetic state by targeting HMGB1. Butyrates 0-8 high mobility group box 1 Homo sapiens 105-110 30832473-4 2019 Glycyrrhizin is a novel pharmacological inhibitor of HMGB1 that may repress prostate cancer metastasis. Glycyrrhizic Acid 0-12 high mobility group box 1 Homo sapiens 53-58 30832473-5 2019 This research was aimed to investigate the effect of glycyrrhizin on inhibition of HMGB1-induced epithelial-to-mesenchymal transition (EMT), a key step of tumor metastasis, in prostate cancer cells. Glycyrrhizic Acid 53-65 high mobility group box 1 Homo sapiens 83-88 30912672-11 2021 Stimulation via HMGB1 significantly increased the migration of CPCs. cpcs 63-67 high mobility group box 1 Homo sapiens 16-21 30832473-8 2019 Furthermore, HMGB1 facilitated cell migration and invasion via downstream signaling, whereas HMGB1 targeting by 10 mM ethyl pyruvate effectively inhibited EMT characteristics. ethyl pyruvate 118-132 high mobility group box 1 Homo sapiens 93-98 30832473-9 2019 Interestingly, cell migration capacity induced by HMGB1 in DU145 cells was abolished in a dose-dependent effect of 25-200 muM glycyrrhizin treatment. Glycyrrhizic Acid 126-138 high mobility group box 1 Homo sapiens 50-55 30832473-10 2019 In conclusion, glycyrrhizin successfully inhibited HMGB1-induced EMT phenomenon, which suggested that glycyrrhizin may serves as a therapeutic agent for metastatic prostate cancer. Glycyrrhizic Acid 15-27 high mobility group box 1 Homo sapiens 51-56 30832473-10 2019 In conclusion, glycyrrhizin successfully inhibited HMGB1-induced EMT phenomenon, which suggested that glycyrrhizin may serves as a therapeutic agent for metastatic prostate cancer. Glycyrrhizic Acid 102-114 high mobility group box 1 Homo sapiens 51-56 31019652-6 2019 miR-122 expression was downregulated while SBP1 expression was upregulated under TNBS-induced colitis or oxidative stress. Trinitrobenzenesulfonic Acid 81-85 high mobility group box 1 Homo sapiens 43-47 31019652-7 2019 Pre-miR-122 or siRNA SBP1 (si-SBP1) treatment ameliorated acute TNBS-induced colitis and H2O2-induced oxidative stress. Hydrogen Peroxide 89-93 high mobility group box 1 Homo sapiens 21-25 31019652-7 2019 Pre-miR-122 or siRNA SBP1 (si-SBP1) treatment ameliorated acute TNBS-induced colitis and H2O2-induced oxidative stress. Hydrogen Peroxide 89-93 high mobility group box 1 Homo sapiens 30-34 30686534-5 2019 In contrast, genetic ablation (using ATG5-/- or ATG7-/- cells) or pharmacologic inhibition (the administration of bafilomycin A1 or chloroquine) of autophagy was found to block ferroptosis activator-induced HMGB1 release. bafilomycin A1 114-128 high mobility group box 1 Homo sapiens 207-212 30686534-5 2019 In contrast, genetic ablation (using ATG5-/- or ATG7-/- cells) or pharmacologic inhibition (the administration of bafilomycin A1 or chloroquine) of autophagy was found to block ferroptosis activator-induced HMGB1 release. Chloroquine 132-143 high mobility group box 1 Homo sapiens 207-212 30686534-4 2019 Both type I and II ferroptosis activators, including erastin, sorafenib, RSL3, and FIN56, induce HMGB1 release in cancer and noncancer cells. erastin 53-60 high mobility group box 1 Homo sapiens 97-102 30686534-4 2019 Both type I and II ferroptosis activators, including erastin, sorafenib, RSL3, and FIN56, induce HMGB1 release in cancer and noncancer cells. Sorafenib 62-71 high mobility group box 1 Homo sapiens 97-102 30227219-4 2019 The present study assessed fecal HMGB1 levels in IBD patients and compared the effects of similar doses of Bifidobacterium longum (Bif) versus VSL#3 on HMGB1 levels in 2,4,6-trinitrobenzene sulfonic acid (TNBS)-induced murine colitis. Trinitrobenzenesulfonic Acid 169-204 high mobility group box 1 Homo sapiens 153-158 30686534-4 2019 Both type I and II ferroptosis activators, including erastin, sorafenib, RSL3, and FIN56, induce HMGB1 release in cancer and noncancer cells. RSL3 73-77 high mobility group box 1 Homo sapiens 97-102 30227219-9 2019 In Caco-2 cells, HMGB1 reduced transepithelial electrical resistance, zonula occludins-1 protein expression, and increased paracellular permeability of FITC-dextran; the opposite was found with both probiotic treatments. fluorescein isothiocyanate dextran 152-164 high mobility group box 1 Homo sapiens 17-22 30260006-10 2019 Treatment with Pingchuanning decoction activated PI3K/Akt/mTOR pathway and inhibited HMGB1/TLR4/NF-kappaB pathway, which could be overturned by LY294002, a PI3K antagonist, or rapamycin (Rapa), an autophagy inducer. 2-(4-morpholinyl)-8-phenyl-4H-1-benzopyran-4-one 144-152 high mobility group box 1 Homo sapiens 85-90 30634142-0 2019 Glycyrrhizin attenuates hepatic ischemia-reperfusion injury by suppressing HMGB1-dependent GSDMD-mediated kupffer cells pyroptosis. Glycyrrhizic Acid 0-12 high mobility group box 1 Homo sapiens 75-80 30634142-3 2019 Glycyrrhizin, a natural anti-inflammatory and antiviral triterpene in clinical use, was recently referred to have ability to prevent I/R induced liver injury by inhibiting HMGB1 expression and activity. Glycyrrhizic Acid 0-12 high mobility group box 1 Homo sapiens 172-177 30260006-10 2019 Treatment with Pingchuanning decoction activated PI3K/Akt/mTOR pathway and inhibited HMGB1/TLR4/NF-kappaB pathway, which could be overturned by LY294002, a PI3K antagonist, or rapamycin (Rapa), an autophagy inducer. Sirolimus 187-191 high mobility group box 1 Homo sapiens 85-90 30260006-10 2019 Treatment with Pingchuanning decoction activated PI3K/Akt/mTOR pathway and inhibited HMGB1/TLR4/NF-kappaB pathway, which could be overturned by LY294002, a PI3K antagonist, or rapamycin (Rapa), an autophagy inducer. Sirolimus 176-185 high mobility group box 1 Homo sapiens 85-90 30716337-0 2019 KLF4 sensitizes the colon cancer cell HCT-15 to cisplatin by altering the expression of HMGB1 and hTERT. Cisplatin 48-57 high mobility group box 1 Homo sapiens 88-93 30732500-5 2019 RESULTS: High HMGB-1 levels were associated with high cortisol levels. Hydrocortisone 54-62 high mobility group box 1 Homo sapiens 14-20 30716337-6 2019 Immunoblotting was used to analyze the change in expression hTERT and HMGB1 involved in KLF4 mediated cisplatin resistance. Cisplatin 102-111 high mobility group box 1 Homo sapiens 70-75 30887719-10 2019 The stimulation study with retenone or LPS suggested that there were mutual regulations between NF-kappaB and HMGB1. retenone 27-35 high mobility group box 1 Homo sapiens 110-115 30716337-10 2019 Immuno-blotting showed that KLF4 positively regulates expression of the survival proteins hTERT and HMGB1 while in presence of cisplatin, expression of HMGB1 and hTERT is negatively regulated by KLF4. Cisplatin 127-136 high mobility group box 1 Homo sapiens 152-157 30318858-6 2019 The use of RIPC in recipients only did not allow for reversal of diabetes, with increased serum HMGB1 at day 1; the three other groups demonstrated significantly better outcomes. ripc 11-15 high mobility group box 1 Homo sapiens 96-101 30891101-4 2019 We conclude that novel strategies targeting HMGB1 may suppress MM cells and interfere with asbestos-induced inflammation. Asbestos 91-99 high mobility group box 1 Homo sapiens 44-49 31037139-0 2019 H2Se Induces Reductive Stress in HepG2 Cells and Activates Cell Autophagy by Regulating the Redox of HMGB1 Protein under Hypoxia. hydrogen selenide 0-4 high mobility group box 1 Homo sapiens 101-106 31037139-7 2019 The redox of HMGB1 protein were analyzed by non-reducing sodium dodecyl sulfate polyacrylamide gel electrophoresis. Sodium Dodecyl Sulfate 57-79 high mobility group box 1 Homo sapiens 13-18 31037139-7 2019 The redox of HMGB1 protein were analyzed by non-reducing sodium dodecyl sulfate polyacrylamide gel electrophoresis. polyacrylamide 80-94 high mobility group box 1 Homo sapiens 13-18 31037139-11 2019 H2Se could interrupt the disulfide bond in HMGB1 and promote its secretion. Disulfides 25-34 high mobility group box 1 Homo sapiens 43-48 30796203-0 2019 Tumor-derived exosomal HMGB1 promotes esophageal squamous cell carcinoma progression through inducing PD1+ TAM expansion. Tamoxifen 107-110 high mobility group box 1 Homo sapiens 23-28 30629986-8 2019 While HMGB1 and ROS production increased with PHMG-P concentration, the addition of anionic compounds with PHMG-P reduced the ROS production and HMGB1 release. polyhexamethyleneguanidine 46-52 high mobility group box 1 Homo sapiens 6-11 30629986-8 2019 While HMGB1 and ROS production increased with PHMG-P concentration, the addition of anionic compounds with PHMG-P reduced the ROS production and HMGB1 release. polyhexamethyleneguanidine 107-113 high mobility group box 1 Homo sapiens 145-150 30538136-6 2019 The RAG1 mutation, R401W, places a bulky tryptophan opposite the binding site for HMG Box A at both 12- and 23-spacer recombination signal sequences, disrupting stable binding of HMGB1. Tryptophan 41-51 high mobility group box 1 Homo sapiens 179-184 30538136-7 2019 Replacement of R401W with leucine and then lysine progressively restores HMGB1 binding, correlating with increased RAG cutting and recombination in vivo. Leucine 26-33 high mobility group box 1 Homo sapiens 73-78 30538136-7 2019 Replacement of R401W with leucine and then lysine progressively restores HMGB1 binding, correlating with increased RAG cutting and recombination in vivo. Lysine 43-49 high mobility group box 1 Homo sapiens 73-78 30538129-0 2019 High-mobility group box 1 links sensing of reactive oxygen species by huntingtin to its nuclear entry. Reactive Oxygen Species 43-66 high mobility group box 1 Homo sapiens 0-25 30859087-6 2019 Results: We found increased staining for HMGB1 in the dermis of psoriatic plaques in comparison to uninvolved skin of patients with PV. pv 132-134 high mobility group box 1 Homo sapiens 41-46 30744691-0 2019 Cancer-cell-secreted CXCL11 promoted CD8+ T cells infiltration through docetaxel-induced-release of HMGB1 in NSCLC. Docetaxel 71-80 high mobility group box 1 Homo sapiens 100-105 30744691-4 2019 Functional experiments were carried out to check whether docetaxel (DOC), a chemotherapeutic agent, modifies the expression of HMGB1 and CXCL11, and influences the infiltration properties of CD8+ T cells to the tumor microenvironment. Docetaxel 57-66 high mobility group box 1 Homo sapiens 127-132 30744691-4 2019 Functional experiments were carried out to check whether docetaxel (DOC), a chemotherapeutic agent, modifies the expression of HMGB1 and CXCL11, and influences the infiltration properties of CD8+ T cells to the tumor microenvironment. Docetaxel 68-71 high mobility group box 1 Homo sapiens 127-132 30744691-8 2019 DOC treatment significantly increased HMGB1 release in an ROS-dependent manner. Docetaxel 0-3 high mobility group box 1 Homo sapiens 38-43 30744691-8 2019 DOC treatment significantly increased HMGB1 release in an ROS-dependent manner. ros 58-61 high mobility group box 1 Homo sapiens 38-43 30744691-12 2019 CONCLUSIONS: Our results demonstrate that DOC induces CD8+ T cell recruitment to the tumor microenvironment by enhancing the secretion of HMGB1 and CXCL11, thus improving the anti-tumor efficacy, indicating that modulating the HMGB1-CXCL11 axis might be helpful for NSCLC treatment. Docetaxel 42-45 high mobility group box 1 Homo sapiens 138-143 30744691-12 2019 CONCLUSIONS: Our results demonstrate that DOC induces CD8+ T cell recruitment to the tumor microenvironment by enhancing the secretion of HMGB1 and CXCL11, thus improving the anti-tumor efficacy, indicating that modulating the HMGB1-CXCL11 axis might be helpful for NSCLC treatment. Docetaxel 42-45 high mobility group box 1 Homo sapiens 227-232 30538129-7 2019 We also found that HMGB1 interacts with the huntingtin N17 region and that this interaction is enhanced by the presence of ROS and phosphorylation of critical serine residues in the N17 region. Reactive Oxygen Species 123-126 high mobility group box 1 Homo sapiens 19-24 30538129-7 2019 We also found that HMGB1 interacts with the huntingtin N17 region and that this interaction is enhanced by the presence of ROS and phosphorylation of critical serine residues in the N17 region. Serine 159-165 high mobility group box 1 Homo sapiens 19-24 30538129-8 2019 We conclude that HMGB1 is a huntingtin N17/PY-NLS ROS-dependent interactor, and this protein bridging is essential for relaying ROS sensing by huntingtin to its nuclear entry during ROS stress. Reactive Oxygen Species 50-53 high mobility group box 1 Homo sapiens 17-22 30538129-8 2019 We conclude that HMGB1 is a huntingtin N17/PY-NLS ROS-dependent interactor, and this protein bridging is essential for relaying ROS sensing by huntingtin to its nuclear entry during ROS stress. Reactive Oxygen Species 128-131 high mobility group box 1 Homo sapiens 17-22 30538129-8 2019 We conclude that HMGB1 is a huntingtin N17/PY-NLS ROS-dependent interactor, and this protein bridging is essential for relaying ROS sensing by huntingtin to its nuclear entry during ROS stress. Reactive Oxygen Species 128-131 high mobility group box 1 Homo sapiens 17-22 30675709-0 2019 Dexmedetomidine Preconditioning Protects Cardiomyocytes Against Hypoxia/Reoxygenation-Induced Necroptosis by Inhibiting HMGB1-Mediated Inflammation. Dexmedetomidine 0-15 high mobility group box 1 Homo sapiens 120-125 30728013-0 2019 Ethyl pyruvate reduces organic dust-induced airway inflammation by targeting HMGB1-RAGE signaling. ethyl pyruvate 0-14 high mobility group box 1 Homo sapiens 77-82 30728013-12 2019 Anti-HMGB1 treatment reduced ODE-induced NF-kappaB p65 expression, IL-6, ROS and RNS but augmented TGF-beta1 and IL-10 levels. ros 73-76 high mobility group box 1 Homo sapiens 5-10 30728013-12 2019 Anti-HMGB1 treatment reduced ODE-induced NF-kappaB p65 expression, IL-6, ROS and RNS but augmented TGF-beta1 and IL-10 levels. Radon 81-84 high mobility group box 1 Homo sapiens 5-10 30838095-0 2019 Correction: N-(2 -Hydroxyphenyl)-2-propylpentanamide (OH-VPA), a histone deacetylase inhibitor, induces the release of nuclear HMGB1 and modifies ROS levels in HeLa cells. N-(2-hydroxyphenyl)-2-propylpentanamide 12-52 high mobility group box 1 Homo sapiens 127-132 30838095-0 2019 Correction: N-(2 -Hydroxyphenyl)-2-propylpentanamide (OH-VPA), a histone deacetylase inhibitor, induces the release of nuclear HMGB1 and modifies ROS levels in HeLa cells. oh-vpa 54-60 high mobility group box 1 Homo sapiens 127-132 30675709-4 2019 Therefore, we speculated that DEX preconditioning may suppress H/R-induced necroptosis by inhibiting expression of HMGB1 in cardiomyocytes. Dexmedetomidine 30-33 high mobility group box 1 Homo sapiens 115-120 30675709-4 2019 Therefore, we speculated that DEX preconditioning may suppress H/R-induced necroptosis by inhibiting expression of HMGB1 in cardiomyocytes. r 65-66 high mobility group box 1 Homo sapiens 115-120 30675709-7 2019 We also found that silencing expression of HMGB1 reinforced the protective effects of DEX preconditioning and overexpression of HMGB1 counteracted the protective effects of DEX preconditioning. Dexmedetomidine 86-89 high mobility group box 1 Homo sapiens 43-48 30675709-7 2019 We also found that silencing expression of HMGB1 reinforced the protective effects of DEX preconditioning and overexpression of HMGB1 counteracted the protective effects of DEX preconditioning. Dexmedetomidine 173-176 high mobility group box 1 Homo sapiens 128-133 30675709-8 2019 Thus, we concluded that DEX preconditioning inhibits H/R-induced necroptosis by inhibiting expression of HMGB1 in cardiomyocytes. Dexmedetomidine 24-27 high mobility group box 1 Homo sapiens 105-110 30675709-8 2019 Thus, we concluded that DEX preconditioning inhibits H/R-induced necroptosis by inhibiting expression of HMGB1 in cardiomyocytes. r 55-56 high mobility group box 1 Homo sapiens 105-110 30578841-0 2019 Corynoxine B ameliorates HMGB1-dependent autophagy dysfunction during manganese exposure in SH-SY5Y human neuroblastoma cells. corynoxine B 0-12 high mobility group box 1 Homo sapiens 25-30 30496780-5 2019 SB1 and SB2 significantly reduced the release of HMGB1 in lipopolysaccharide (LPS)-activated primary human umbilical vein endothelial cells (HUVECS) via the SIRT1-mediated deacetylation of HMGB1. sb2 8-11 high mobility group box 1 Homo sapiens 49-54 30496780-5 2019 SB1 and SB2 significantly reduced the release of HMGB1 in lipopolysaccharide (LPS)-activated primary human umbilical vein endothelial cells (HUVECS) via the SIRT1-mediated deacetylation of HMGB1. sb2 8-11 high mobility group box 1 Homo sapiens 189-194 30578841-8 2019 Our results showed that Cory B ameliorated Mn-induced autophagic dysregulation and neurotoxicity partly by dissociating HMGB1 from alpha-synuclein and inhibiting mTOR signaling. corynoxine B 24-30 high mobility group box 1 Homo sapiens 120-125 30578841-0 2019 Corynoxine B ameliorates HMGB1-dependent autophagy dysfunction during manganese exposure in SH-SY5Y human neuroblastoma cells. Manganese 70-79 high mobility group box 1 Homo sapiens 25-30 30676239-6 2019 DOX suppressed the emission of HMGB1 while Nano-DOX stimulated HMGB1 emission which was attenuated when autophagy was repressed. Doxorubicin 0-3 high mobility group box 1 Homo sapiens 31-36 30527499-0 2019 Corrigendum to Beneficial Effects of Dantrolene on Sepsis-Induced Diaphragmatic Dysfunction Are Associated With Downregulation of High-Mobility Group Box 1 and Calpain-Caspase-3 Proteolytic Pathway [Journal of Surgical Research 200(2) (2016) 637-647]. Dantrolene 37-47 high mobility group box 1 Homo sapiens 130-155 30732222-11 2019 Correlation analysis showed that serum HMGB1 levels were positive significant correlated with the Birmingham Vasculitis Activity Score, hypersensitive C reactive protein (Hs-CRP), serum creatinine (Scr) and 24-hour proteinuria (all P < .05). Creatinine 186-196 high mobility group box 1 Homo sapiens 39-44 30676239-6 2019 DOX suppressed the emission of HMGB1 while Nano-DOX stimulated HMGB1 emission which was attenuated when autophagy was repressed. Doxorubicin 48-51 high mobility group box 1 Homo sapiens 63-68 30676239-7 2019 Blocking of HMGB1 emission mitigated autophagy and enhanced apoptosis in Nano-DOX-treated GC. Doxorubicin 78-81 high mobility group box 1 Homo sapiens 12-17 30676239-8 2019 Exogenously administered HMGB1 promoted autophagy and protected against apoptosis in both Nano-DOX-treated GC and DOX-treated GC. Doxorubicin 95-98 high mobility group box 1 Homo sapiens 25-30 30676239-8 2019 Exogenously administered HMGB1 promoted autophagy and protected against apoptosis in both Nano-DOX-treated GC and DOX-treated GC. Doxorubicin 114-117 high mobility group box 1 Homo sapiens 25-30 30676239-10 2019 Nano-DOX initiates a mutual reinforcement loop between autophagy and HMGB1 in GC and thereby protects GC against apoptosis. Doxorubicin 5-8 high mobility group box 1 Homo sapiens 69-74 30906660-7 2019 Here, we describe for the first time production of the in vitro hallmarks of immunogenic cell death - ecto-calreticulin and secreted ATP and HMGB1 protein - by cells in response to treatment with antibody-drug conjugates bearing a maytansine payload. Maytansine 231-241 high mobility group box 1 Homo sapiens 141-146 30782274-12 2019 Glycyrrhizin inhibited the mRNA expression of HMGB1 and the mRNA and protein expression of TLR4 and NF-kappaB and then reduced the mRNA expression of IL-8 (P<0.05). Glycyrrhizic Acid 0-12 high mobility group box 1 Homo sapiens 46-51 30782274-14 2019 The HMGB1 blocker glycyrrhizin can inhibit activation of the TLR4/NF-kappaB signaling pathway and the secretion of inflammatory factors. Glycyrrhizic Acid 18-30 high mobility group box 1 Homo sapiens 4-9 30704538-0 2019 Upregulation of miR-107 expression following hyperbaric oxygen treatment suppresses HMGB1/RAGE signaling in degenerated human nucleus pulposus cells. Oxygen 56-62 high mobility group box 1 Homo sapiens 84-89 30704538-3 2019 In this study, we investigated the regulation of the miR-107/HMGB1/RAGE pathway in degenerated nucleus pulposus cells (NPCs) after hyperbaric oxygen (HBO) treatment. Oxygen 142-148 high mobility group box 1 Homo sapiens 61-66 30391318-0 2019 Effect of hyperbaric oxygen therapy on HMGB1/NF-kappaB expression and prognosis of acute spinal cord injury: A randomized clinical trial. Oxygen 21-27 high mobility group box 1 Homo sapiens 39-44 30502394-0 2019 gamma-Mangostin alleviates liver fibrosis through Sirtuin 3-superoxide-high mobility group box 1 signaling axis. gamma-mangostin 0-15 high mobility group box 1 Homo sapiens 71-96 30502394-0 2019 gamma-Mangostin alleviates liver fibrosis through Sirtuin 3-superoxide-high mobility group box 1 signaling axis. Superoxides 59-70 high mobility group box 1 Homo sapiens 71-96 30030153-5 2019 After intermittent UVB exposure, melanocytes treated with the JAK inhibitor ruxolitinib reduced expression of HMGB1 and MX1 as well as activation of JAK1 (pJAK1) and signal transducer and activator of transcription 1 (pSTAT1). ruxolitinib 76-87 high mobility group box 1 Homo sapiens 110-115 30966773-3 2019 Aloin was administered after LPS or HMGB1 challenge, and the antiseptic activity of aloin was determined from measurements of permeability, activation of pro-inflammatory proteins and production of markers for tissue injury in HMGB1-activated human umbilical vein endothelial cells (HUVECs) and a cecal ligation and puncture (CLP)-induced sepsis mouse model. alloin 84-89 high mobility group box 1 Homo sapiens 227-232 30474802-6 2019 Elevated plasma HMGB1 concentrations can serve as early and sensitive mechanistic biomarker for clinical acetaminophen hepatotoxicity. Acetaminophen 105-118 high mobility group box 1 Homo sapiens 16-21 30474802-8 2019 In addition, HMGB1 mediates 80% of gut bacterial translocation (BT) during acetaminophen toxicity. Acetaminophen 75-88 high mobility group box 1 Homo sapiens 13-18 30502088-0 2019 MiR-216a-5p protects 16HBE cells from H2O2-induced oxidative stress through targeting HMGB1/NF-kB pathway. mir-216a-5p 0-11 high mobility group box 1 Homo sapiens 86-91 30502088-0 2019 MiR-216a-5p protects 16HBE cells from H2O2-induced oxidative stress through targeting HMGB1/NF-kB pathway. Hydrogen Peroxide 38-42 high mobility group box 1 Homo sapiens 86-91 30502088-4 2019 A significantly elevation of HMGB1 protein expression and a reduction of miR-216a-5p expression were observed in children with asthma as well as in H2O2 stimulated 16HBE cells. Hydrogen Peroxide 148-152 high mobility group box 1 Homo sapiens 29-34 30502088-6 2019 MiR-216a-5p repressed HMGB1 protein expression in H2O2 induced 16HBE cells. mir-216a-5p 0-11 high mobility group box 1 Homo sapiens 22-27 30502088-6 2019 MiR-216a-5p repressed HMGB1 protein expression in H2O2 induced 16HBE cells. Hydrogen Peroxide 50-54 high mobility group box 1 Homo sapiens 22-27 30502088-9 2019 In conclusion, these results suggested that miR-216a-5p functions as a negative regulator of H2O2 induced 16HBE cell injury through targeting HMGB1/NF-kB pathway, provided a potential therapeutic target for asthma. Hydrogen Peroxide 93-97 high mobility group box 1 Homo sapiens 142-147 30630344-6 2019 The antiseptic activity of ginsenoside Rh1 was determined by measuring the permeability, leukocyte adhesion and migration, activation of pro-inflammatory proteins in HMGB1-activated human umbilical vein endothelial cells (HUVECs) and mice, and the survival rate in a sepsis mouse model. ginsenoside Rh1 27-42 high mobility group box 1 Homo sapiens 166-171 30551543-5 2019 Concomitant treatment of CoQ10 & methotrexate caused improvement in histological picture of the lung and liver tissues, liver function and oxidative stress biomarkers, modulation of autophagy genes [mammalian target of rapamycin (m-TOR), Microtubule-associated proteins 1 A/1B light chain 3 (MAP1LC3B), and Sequestosome 1 ubiquitin-binding protein p62 (p62/SQSTM1)] with simultaneous reduction in High Mobility Group Protein B1 (HMGB1). coenzyme Q10 25-30 high mobility group box 1 Homo sapiens 401-431 30551543-5 2019 Concomitant treatment of CoQ10 & methotrexate caused improvement in histological picture of the lung and liver tissues, liver function and oxidative stress biomarkers, modulation of autophagy genes [mammalian target of rapamycin (m-TOR), Microtubule-associated proteins 1 A/1B light chain 3 (MAP1LC3B), and Sequestosome 1 ubiquitin-binding protein p62 (p62/SQSTM1)] with simultaneous reduction in High Mobility Group Protein B1 (HMGB1). coenzyme Q10 25-30 high mobility group box 1 Homo sapiens 433-438 30551543-5 2019 Concomitant treatment of CoQ10 & methotrexate caused improvement in histological picture of the lung and liver tissues, liver function and oxidative stress biomarkers, modulation of autophagy genes [mammalian target of rapamycin (m-TOR), Microtubule-associated proteins 1 A/1B light chain 3 (MAP1LC3B), and Sequestosome 1 ubiquitin-binding protein p62 (p62/SQSTM1)] with simultaneous reduction in High Mobility Group Protein B1 (HMGB1). Adenosine Monophosphate 32-35 high mobility group box 1 Homo sapiens 401-431 30551543-5 2019 Concomitant treatment of CoQ10 & methotrexate caused improvement in histological picture of the lung and liver tissues, liver function and oxidative stress biomarkers, modulation of autophagy genes [mammalian target of rapamycin (m-TOR), Microtubule-associated proteins 1 A/1B light chain 3 (MAP1LC3B), and Sequestosome 1 ubiquitin-binding protein p62 (p62/SQSTM1)] with simultaneous reduction in High Mobility Group Protein B1 (HMGB1). Adenosine Monophosphate 32-35 high mobility group box 1 Homo sapiens 433-438 30551543-5 2019 Concomitant treatment of CoQ10 & methotrexate caused improvement in histological picture of the lung and liver tissues, liver function and oxidative stress biomarkers, modulation of autophagy genes [mammalian target of rapamycin (m-TOR), Microtubule-associated proteins 1 A/1B light chain 3 (MAP1LC3B), and Sequestosome 1 ubiquitin-binding protein p62 (p62/SQSTM1)] with simultaneous reduction in High Mobility Group Protein B1 (HMGB1). Methotrexate 37-49 high mobility group box 1 Homo sapiens 401-431 30551543-5 2019 Concomitant treatment of CoQ10 & methotrexate caused improvement in histological picture of the lung and liver tissues, liver function and oxidative stress biomarkers, modulation of autophagy genes [mammalian target of rapamycin (m-TOR), Microtubule-associated proteins 1 A/1B light chain 3 (MAP1LC3B), and Sequestosome 1 ubiquitin-binding protein p62 (p62/SQSTM1)] with simultaneous reduction in High Mobility Group Protein B1 (HMGB1). Methotrexate 37-49 high mobility group box 1 Homo sapiens 433-438 29895396-5 2019 In the present study, HMGB-1 levels in serum samples collected from six-week-old piglets infected intra-nasally with 2 x 105.75 TCID50/mL of Indian PRRSV (Ind-297221/2013) was estimated by ELISA up to 21 days post infection (dpi). 3-aminodiphenyleneiodium 225-228 high mobility group box 1 Homo sapiens 22-28 29895396-7 2019 Mean HMGB-1 concentration in serum was found to be significantly elevated in PRRSV infected piglets on 6 dpi as compared to uninfected control piglets. 3-aminodiphenyleneiodium 105-108 high mobility group box 1 Homo sapiens 5-11 31266378-0 2019 Curcumin inhibits the lymphangiogenesis of gastric cancer cells by inhibiton of HMGB1/VEGF-D signaling. Curcumin 0-8 high mobility group box 1 Homo sapiens 80-85 31266378-3 2019 However, the molecular mechanisms by which curcumin regulates HMGB1-mediated lymphangiogenesis in gastric cancer remain unclear. Curcumin 43-51 high mobility group box 1 Homo sapiens 62-67 31266378-5 2019 The effects of curcumin on HMGB1 and VEGF-D expression were examined by reverse transcription polymerase chain reaction (RT-PCR) and western blot analysis. Curcumin 15-23 high mobility group box 1 Homo sapiens 27-32 31266378-7 2019 The mRNA and protein expression levels of HMGB1 and VEGF-D were significantly eliminated by curcumin administration. Curcumin 92-100 high mobility group box 1 Homo sapiens 42-47 31266378-8 2019 Pre-treatment with the recombinant HMGB1 (rHMGB1) markedly abolished curcumin-reduced VEGF-D expression. Curcumin 69-77 high mobility group box 1 Homo sapiens 35-40 31266378-9 2019 Our findings suggested that curcumin might exert anti-lymphangiogenesis in gastric cancer by inhibition of HMGB1/VEGF-D signaling. Curcumin 28-36 high mobility group box 1 Homo sapiens 107-112 30954689-6 2019 Moreover, 5-LO expression, accompanied by production of leukotrienes was markedly increased in HMGB1-stimulated VSMCs, which was attenuated in cells deficient of TLR2 or RAGE. Leukotrienes 56-68 high mobility group box 1 Homo sapiens 95-100 30273982-9 2019 Inhibition of TLR-4 signaling, with TAK-242, completely abrogated HMGB1 induced IL-6 and MMP-1 expression, whereas inhibition of TLR-2, with O-vanillin, or RAGE, with FPS-ZM1, had mild inhibitory effects. ethyl 6-(N-(2-chloro-4-fluorophenyl)sulfamoyl)cyclohex-1-ene-1-carboxylate 36-43 high mobility group box 1 Homo sapiens 66-71 31727240-5 2019 Thus, exclusively in the presence of biotin, which liberates HMGB1-SBP-GFP from its nuclear hook, immunogenic cell death (ICD) inducers such as anthracyclines are able to cause the nucleo-cytoplasmic translocation of HMGB1-SBP-GFP. Biotin 37-43 high mobility group box 1 Homo sapiens 61-66 31727240-5 2019 Thus, exclusively in the presence of biotin, which liberates HMGB1-SBP-GFP from its nuclear hook, immunogenic cell death (ICD) inducers such as anthracyclines are able to cause the nucleo-cytoplasmic translocation of HMGB1-SBP-GFP. Biotin 37-43 high mobility group box 1 Homo sapiens 217-222 31727240-5 2019 Thus, exclusively in the presence of biotin, which liberates HMGB1-SBP-GFP from its nuclear hook, immunogenic cell death (ICD) inducers such as anthracyclines are able to cause the nucleo-cytoplasmic translocation of HMGB1-SBP-GFP. Anthracyclines 144-158 high mobility group box 1 Homo sapiens 61-66 31727240-5 2019 Thus, exclusively in the presence of biotin, which liberates HMGB1-SBP-GFP from its nuclear hook, immunogenic cell death (ICD) inducers such as anthracyclines are able to cause the nucleo-cytoplasmic translocation of HMGB1-SBP-GFP. Anthracyclines 144-158 high mobility group box 1 Homo sapiens 217-222 30954689-5 2019 Both of these effects were markedly attenuated in cells pretreated with zileuton (1-10 muM), a 5-LO inhibitor, as well as in cells transfected with 5-LO siRNA, suggesting a potential involvement of 5-LO signaling in HMGB1-mediated RAGE expression in VSMCs. zileuton 72-80 high mobility group box 1 Homo sapiens 216-221 29600445-9 2018 Moreover, heat-shock treatment of GBCCLs induced calreticulin translocation and release of HMGB1 and ATP, both known to act as danger signals. gbccls 34-40 high mobility group box 1 Homo sapiens 91-96 30204458-10 2018 By dividing the distribution of HMGB1 levels into quartiles, serum HMGB1 levels were increased gradually with the increase of testosterone levels (p<0.05), whereas the FMD levels decreased (p<0.05). Testosterone 126-138 high mobility group box 1 Homo sapiens 32-37 30204458-10 2018 By dividing the distribution of HMGB1 levels into quartiles, serum HMGB1 levels were increased gradually with the increase of testosterone levels (p<0.05), whereas the FMD levels decreased (p<0.05). Testosterone 126-138 high mobility group box 1 Homo sapiens 67-72 30204458-12 2018 The absolute changes in HMGB1 showed a positive correlation with the changes in testosterone (p<0.05) and negative correlation with the changes in FMD (p<0.05) in patients with PCOS during the course of metformin therapy. Testosterone 80-92 high mobility group box 1 Homo sapiens 24-29 30204458-12 2018 The absolute changes in HMGB1 showed a positive correlation with the changes in testosterone (p<0.05) and negative correlation with the changes in FMD (p<0.05) in patients with PCOS during the course of metformin therapy. Metformin 209-218 high mobility group box 1 Homo sapiens 24-29 30481260-9 2018 Furthermore, our study illustrated that atorvastatin played its role in CPC viability and proliferation by modulating the expression of HMGB1 through the MEG3/miR-22 pathway. Atorvastatin 40-52 high mobility group box 1 Homo sapiens 136-141 30662657-2 2018 Isoliquiritigenin (ISL) as a small molecular from licorice, is able to inhibit the expression of HMGB1. isoliquiritigenin 0-17 high mobility group box 1 Homo sapiens 97-102 30662657-2 2018 Isoliquiritigenin (ISL) as a small molecular from licorice, is able to inhibit the expression of HMGB1. isoliquiritigenin 19-22 high mobility group box 1 Homo sapiens 97-102 30587909-4 2018 Glycyrrhizin binds to HMGB1, inhibiting cytokine activities, thus potentially improving DED. Glycyrrhizic Acid 0-12 high mobility group box 1 Homo sapiens 22-27 30481260-0 2018 Atorvastatin protects cardiac progenitor cells from hypoxia-induced cell growth inhibition via MEG3/miR-22/HMGB1 pathway. Atorvastatin 0-12 high mobility group box 1 Homo sapiens 107-112 30325653-0 2018 Glycyrrhizin, a Potential Drug for Autoimmune Encephalomyelitis by Inhibiting High-Mobility Group Box 1. Glycyrrhizic Acid 0-12 high mobility group box 1 Homo sapiens 78-103 30476719-7 2018 Bipartite recognition of heptamer and nonamer, flexibly linked nonamer-binding domain dimer relatively to the heptamer recognition region dimer, and RSS plasticity and bending by HMGB1 together contribute to the molecular basis of the 12/23 rule in the RAG molecular machine. RSS 149-152 high mobility group box 1 Homo sapiens 179-184 30325653-3 2018 Glycyrrhizin (GL), a major constituent of licorice root, can inhibit the proinflammatory bioactivities of HMGB1. Glycyrrhizic Acid 0-12 high mobility group box 1 Homo sapiens 106-111 30325653-3 2018 Glycyrrhizin (GL), a major constituent of licorice root, can inhibit the proinflammatory bioactivities of HMGB1. Glycyrrhizic Acid 14-16 high mobility group box 1 Homo sapiens 106-111 30468092-7 2018 In particular, ONOO- and its derivatives could mediate neurovascular unit damages and induce the BBB disruption and HT possibly through interacting with different cellular signalling pathways including matrix metalloproteinase (MMPs), high mobility group Box 1 (HMGB1), toll-like receptor2/4, poly(ADP-ribose) polymerase, Src, ROCK, and GSK-3beta. onoo 15-19 high mobility group box 1 Homo sapiens 235-260 30468092-7 2018 In particular, ONOO- and its derivatives could mediate neurovascular unit damages and induce the BBB disruption and HT possibly through interacting with different cellular signalling pathways including matrix metalloproteinase (MMPs), high mobility group Box 1 (HMGB1), toll-like receptor2/4, poly(ADP-ribose) polymerase, Src, ROCK, and GSK-3beta. onoo 15-19 high mobility group box 1 Homo sapiens 262-267 30328477-0 2018 PKA regulates HMGB1 through activation of IGFBP-3 and SIRT1 in human retinal endothelial cells cultured in high glucose. Glucose 112-119 high mobility group box 1 Homo sapiens 14-19 30328477-11 2018 High glucose inhibited SIRT1 levels and increased cytoplasmic HMGB1 in REC. Glucose 5-12 high mobility group box 1 Homo sapiens 62-67 30338917-0 2018 HMGB1 deficiency reduces H2 O2 -induced oxidative damage in human melanocytes via the Nrf2 pathway. Hydrogen Peroxide 25-30 high mobility group box 1 Homo sapiens 0-5 30338917-7 2018 H2 O2 treatment increased cytoplasmic translocation and extracellular release of HMGB1. Hydrogen Peroxide 0-5 high mobility group box 1 Homo sapiens 81-86 30338917-10 2018 The expression of Nrf2 and its downstream antioxidant genes was downregulated after the supernatant of H2 O2 -treated NHEMs was added to HMGB1-deficient cells. Hydrogen Peroxide 103-108 high mobility group box 1 Homo sapiens 137-142 30680297-11 2018 Conclusion: Our results indicated that the levels of IL-17, IL-33, and IL-6 in supernatants from patients" cultured T cells were increased after stimulation with HMGB-1 following employing quercetin. Quercetin 189-198 high mobility group box 1 Homo sapiens 162-168 30680297-1 2018 Statement of the Problem: Quercetin is a pharmacological flavonoid that can inhibit high mobility group box1 (HMGB1) protein, a non-histone nuclear protein that is implicated in inflammation. Quercetin 26-35 high mobility group box 1 Homo sapiens 84-108 30118903-2 2018 Here we showed that ALA-PDT significantly upregulated HMGB1 while downregulated miR-34a expression levels in cervical cancer tissues, and the percentages of mature DCs(mDCs) were increased in ALA-PDT treated patients" peripheral blood. Alanine 20-23 high mobility group box 1 Homo sapiens 54-59 30680297-1 2018 Statement of the Problem: Quercetin is a pharmacological flavonoid that can inhibit high mobility group box1 (HMGB1) protein, a non-histone nuclear protein that is implicated in inflammation. Quercetin 26-35 high mobility group box 1 Homo sapiens 110-115 30680297-1 2018 Statement of the Problem: Quercetin is a pharmacological flavonoid that can inhibit high mobility group box1 (HMGB1) protein, a non-histone nuclear protein that is implicated in inflammation. Flavonoids 57-66 high mobility group box 1 Homo sapiens 84-108 30680297-1 2018 Statement of the Problem: Quercetin is a pharmacological flavonoid that can inhibit high mobility group box1 (HMGB1) protein, a non-histone nuclear protein that is implicated in inflammation. Flavonoids 57-66 high mobility group box 1 Homo sapiens 110-115 30680297-5 2018 Purpose: The current study aimed to compare blocking of HMGB1 function and stimulation of HMGB1 function with quercetin and investigate the effects of the blockage on T helper 17 (Th17) cells and mitogen-activated protein kinase Toll-like receptor 4 (MAPK-TLR4) signaling pathway. Quercetin 110-119 high mobility group box 1 Homo sapiens 90-95 30680297-9 2018 Results: The level of these cytokines decreased; moreover, western blot data showed that quercetin could decrease MAPK signaling pathway by means of inhibition of HMGB1 on T cells. Quercetin 89-98 high mobility group box 1 Homo sapiens 163-168 30118903-6 2018 By co-culturing cervical cancer cell lines with immature DCs(imDCs) in the Transwell systems, we found that ALA-PDT induced HMGB1 exosomes could promote DCs maturation, which could be reversed by silencing HMGB1 in HPV-positive cervical cancer cells. Alanine 108-111 high mobility group box 1 Homo sapiens 124-129 30118903-6 2018 By co-culturing cervical cancer cell lines with immature DCs(imDCs) in the Transwell systems, we found that ALA-PDT induced HMGB1 exosomes could promote DCs maturation, which could be reversed by silencing HMGB1 in HPV-positive cervical cancer cells. Alanine 108-111 high mobility group box 1 Homo sapiens 206-211 30055307-9 2018 However, HMGB1 overexpression attenuated the protective effect of miR-106 on HUVECs in high glucose conditions. Glucose 92-99 high mobility group box 1 Homo sapiens 9-14 30591798-7 2018 The results from RT-PCR analysis revealed a significant reduction in the expression of genes involved in inflammation including, HMGB1, IL-6 and IL-8 on treatment of DU-145 cells with crocetin. crocetin 184-192 high mobility group box 1 Homo sapiens 129-134 30055307-12 2018 Taken together, these data collectively suggested that miR-106 was a potential molecular target for inhibiting high glucose-induced inflammation and apoptosis by targeting HMGB1. Glucose 116-123 high mobility group box 1 Homo sapiens 172-177 30055307-5 2018 The results showed that the expression of HMGB1 was increased in human umbilical vein endothelial cells (HUVECs) after exposure to high glucose (25 mM). Glucose 136-143 high mobility group box 1 Homo sapiens 42-47 30055307-6 2018 Downregulation of HMGB1 attenuated the high glucose-induced antiangiogenesis of HUVECs, and the decrease expression of HMGB1 inhibiting HUVEC apoptosis and inflammatory factor expression. Glucose 44-51 high mobility group box 1 Homo sapiens 18-23 30464464-0 2018 A novel disulfide bond-mediated cleavable RGD-modified PAMAM nanocomplex containing nuclear localization signal HMGB1 for enhancing gene transfection efficiency. Disulfides 8-17 high mobility group box 1 Homo sapiens 112-117 30464464-0 2018 A novel disulfide bond-mediated cleavable RGD-modified PAMAM nanocomplex containing nuclear localization signal HMGB1 for enhancing gene transfection efficiency. Poly(amidoamine) 55-60 high mobility group box 1 Homo sapiens 112-117 30464464-7 2018 A pDNA/HMGB1/PAMAM-SS-PEG-RGD (DHP) nanocomplex was prepared via electrostatic interaction and characterized. pamam-ss-peg-rgd 13-29 high mobility group box 1 Homo sapiens 7-12 30464464-10 2018 Intracellular trafficking and in vitro expression study indicated that the DHP nanocomplex escaped from lysosomes and the disulfide bonds between PAMAM and PEG cleaved due to the high concentration of GSH in the cytoplasm, pDNA consequently became exclusively located in the nucleus under the guidance of HMGB1, thereby promoting the red fluorescence protein (RFP) expression. Disulfides 122-131 high mobility group box 1 Homo sapiens 305-310 30464464-10 2018 Intracellular trafficking and in vitro expression study indicated that the DHP nanocomplex escaped from lysosomes and the disulfide bonds between PAMAM and PEG cleaved due to the high concentration of GSH in the cytoplasm, pDNA consequently became exclusively located in the nucleus under the guidance of HMGB1, thereby promoting the red fluorescence protein (RFP) expression. Poly(amidoamine) 146-151 high mobility group box 1 Homo sapiens 305-310 30464464-10 2018 Intracellular trafficking and in vitro expression study indicated that the DHP nanocomplex escaped from lysosomes and the disulfide bonds between PAMAM and PEG cleaved due to the high concentration of GSH in the cytoplasm, pDNA consequently became exclusively located in the nucleus under the guidance of HMGB1, thereby promoting the red fluorescence protein (RFP) expression. Polyethylene Glycols 156-159 high mobility group box 1 Homo sapiens 305-310 30464464-10 2018 Intracellular trafficking and in vitro expression study indicated that the DHP nanocomplex escaped from lysosomes and the disulfide bonds between PAMAM and PEG cleaved due to the high concentration of GSH in the cytoplasm, pDNA consequently became exclusively located in the nucleus under the guidance of HMGB1, thereby promoting the red fluorescence protein (RFP) expression. Glutathione 201-204 high mobility group box 1 Homo sapiens 305-310 30307477-5 2018 In this work, we found that the expression of high-mobility group box 1(HMGB1) was upregulated in CPCs under hypoxia conditions. cpcs 98-102 high mobility group box 1 Homo sapiens 46-71 30307477-5 2018 In this work, we found that the expression of high-mobility group box 1(HMGB1) was upregulated in CPCs under hypoxia conditions. cpcs 98-102 high mobility group box 1 Homo sapiens 72-77 30307477-7 2018 Additionally, mitogen-activated protein kinase (MAPK) signaling pathway was found to be involved in regulating DNMT1/HMGB1-mediated CPC apoptosis in hypoxia process. cpc 132-135 high mobility group box 1 Homo sapiens 117-122 30175447-10 2018 RESULTS: Excess glucose triggered a trophoblast sterile inflammatory IL-8 and antimigratory response through HMGB1 activation of TLR4. Glucose 16-23 high mobility group box 1 Homo sapiens 109-114 29178411-5 2018 Additionally, plasma oleoylethanolamide positively correlated with plasma levels of high mobility group box-1, which is a danger-associated molecular pattern and an endogenous TLR4 agonist, specifically in female alcohol binge drinkers. oleoylethanolamide 21-39 high mobility group box 1 Homo sapiens 84-109 29178411-5 2018 Additionally, plasma oleoylethanolamide positively correlated with plasma levels of high mobility group box-1, which is a danger-associated molecular pattern and an endogenous TLR4 agonist, specifically in female alcohol binge drinkers. Alcohols 213-220 high mobility group box 1 Homo sapiens 84-109 30175447-0 2018 Excess glucose induce trophoblast inflammation and limit cell migration through HMGB1 activation of Toll-Like receptor 4. Glucose 7-14 high mobility group box 1 Homo sapiens 80-85 30214593-5 2018 Therefore, the probable involvement of HMGB1 in the development of carboplatin resistance in ovarian cancer SKOV3 cells was investigated. Carboplatin 67-78 high mobility group box 1 Homo sapiens 39-44 30257412-0 2018 MiR-1284 enhances sensitivity of cervical cancer cells to cisplatin via downregulating HMGB1. Cisplatin 58-67 high mobility group box 1 Homo sapiens 87-92 30257412-16 2018 CONCLUSION: miR-1284 enhances sensitivity of cervical cancer cells to cisplatin via targeting HMGB1. Cisplatin 70-79 high mobility group box 1 Homo sapiens 94-99 30367066-6 2018 SMART in combination with imaging of the release of nuclear DAMPs and Live-Cell Imaging for Secretion activity (LCI-S) reveals two different modes of the release of High Mobility Group Box 1 from necroptotic cells. lci-s 112-117 high mobility group box 1 Homo sapiens 165-190 30086328-0 2018 High-mobility group box 1 protein-mediated necroptosis contributes to dasatinib-induced cardiotoxicity. Dasatinib 70-79 high mobility group box 1 Homo sapiens 0-25 30086328-9 2018 What"s more, unlike the inflammation-associated necroptosis, dasatinib-triggered necroptosis was dependent on intracellular instead of secreted High-mobility group box 1 (HMGB1) protein. Dasatinib 61-70 high mobility group box 1 Homo sapiens 144-169 30086328-9 2018 What"s more, unlike the inflammation-associated necroptosis, dasatinib-triggered necroptosis was dependent on intracellular instead of secreted High-mobility group box 1 (HMGB1) protein. Dasatinib 61-70 high mobility group box 1 Homo sapiens 171-176 30086328-10 2018 Collectively, our study revealed that dasatinib-induced cardiotoxicity acted via leading cardiomyocytes to HMGB1-mediated necroptosis, indicating a viable strategy for prevention of dasatinib-induced cardiotoxicity. Dasatinib 38-47 high mobility group box 1 Homo sapiens 107-112 30086328-10 2018 Collectively, our study revealed that dasatinib-induced cardiotoxicity acted via leading cardiomyocytes to HMGB1-mediated necroptosis, indicating a viable strategy for prevention of dasatinib-induced cardiotoxicity. Dasatinib 182-191 high mobility group box 1 Homo sapiens 107-112 30119194-9 2018 Furthermore, the SH-SY5Y cells were exposed to high glucose stimulation, FMN (2.5, 5 and 10 muM) treatment, and glycyrrhizin, the selective inhibitor of HMGB1. Glycyrrhizic Acid 112-124 high mobility group box 1 Homo sapiens 153-158 30119194-13 2018 The inhibition of HMGB1 by glycyrrhizin also confirmed the involvement of HMGB1/TLR4/NF-kappaB/NLRP3 pathway in high glucose-induced SH-SY5Y cells. Glycyrrhizic Acid 27-39 high mobility group box 1 Homo sapiens 18-23 30119194-13 2018 The inhibition of HMGB1 by glycyrrhizin also confirmed the involvement of HMGB1/TLR4/NF-kappaB/NLRP3 pathway in high glucose-induced SH-SY5Y cells. Glycyrrhizic Acid 27-39 high mobility group box 1 Homo sapiens 74-79 30119194-13 2018 The inhibition of HMGB1 by glycyrrhizin also confirmed the involvement of HMGB1/TLR4/NF-kappaB/NLRP3 pathway in high glucose-induced SH-SY5Y cells. Glucose 117-124 high mobility group box 1 Homo sapiens 18-23 30119194-13 2018 The inhibition of HMGB1 by glycyrrhizin also confirmed the involvement of HMGB1/TLR4/NF-kappaB/NLRP3 pathway in high glucose-induced SH-SY5Y cells. Glucose 117-124 high mobility group box 1 Homo sapiens 74-79 30119194-14 2018 In summary, the results suggested that FMN exhibited the protective effect on STZ-induced cognitive impairment possibly via the mediation of HMGB1/TLR4/NF-kappaB signaling and NLRP3 inflammasome. Streptozocin 78-81 high mobility group box 1 Homo sapiens 141-146 30313098-2 2018 Experimental animal studies suggested that increased extracellular histone and high morbidity group box-1 (HMGB1) levels might contribute to ALI development. ali 141-144 high mobility group box 1 Homo sapiens 79-105 30313098-2 2018 Experimental animal studies suggested that increased extracellular histone and high morbidity group box-1 (HMGB1) levels might contribute to ALI development. ali 141-144 high mobility group box 1 Homo sapiens 107-112 30132524-0 2018 Renoprotection of dapagliflozin in human renal proximal tubular cells via the inhibition of the high mobility group box 1-receptor for advanced glycation end products-nuclear factor-kappaB signaling pathway. dapagliflozin 18-31 high mobility group box 1 Homo sapiens 96-121 30114640-7 2018 In addition, GSK"872 could also decrease the protein levels of RIPK3 and MLKL, and cytoplasmic translocation and expression of HMGB1, an important pro-inflammatory protein. gsk"872 13-20 high mobility group box 1 Homo sapiens 127-132 30314759-5 2018 Subsequently, HMGB1 permeabilized the phospholipid bilayer in the acidic environment of lysosomes. Phospholipids 38-50 high mobility group box 1 Homo sapiens 14-19 30405740-10 2018 MAN induced declines of both HMGB1/TLR4/p-p65 and TNF-alpha were substantially reversed by cotreatment with nicotinamide mononucleotide or NAD. Nicotinamide Mononucleotide 108-135 high mobility group box 1 Homo sapiens 29-34 30405740-10 2018 MAN induced declines of both HMGB1/TLR4/p-p65 and TNF-alpha were substantially reversed by cotreatment with nicotinamide mononucleotide or NAD. NAD 139-142 high mobility group box 1 Homo sapiens 29-34 30119256-0 2018 Synergy of theophylline reduces necrotic effect of berberine, induces cell cycle arrest and PARP, HMGB1, Bcl-2 family mediated apoptosis in MDA-MB-231 breast cancer cells. Theophylline 11-23 high mobility group box 1 Homo sapiens 98-103 30119256-6 2018 Combined treatment of Berberine and theophylline reduced extracellular level of HMGB1 and down regulated HMGB1 and MMP-9 mRNA expression. Berberine 22-31 high mobility group box 1 Homo sapiens 80-85 30119256-6 2018 Combined treatment of Berberine and theophylline reduced extracellular level of HMGB1 and down regulated HMGB1 and MMP-9 mRNA expression. Berberine 22-31 high mobility group box 1 Homo sapiens 105-110 30119256-6 2018 Combined treatment of Berberine and theophylline reduced extracellular level of HMGB1 and down regulated HMGB1 and MMP-9 mRNA expression. Theophylline 36-48 high mobility group box 1 Homo sapiens 80-85 30119256-6 2018 Combined treatment of Berberine and theophylline reduced extracellular level of HMGB1 and down regulated HMGB1 and MMP-9 mRNA expression. Theophylline 36-48 high mobility group box 1 Homo sapiens 105-110 30119256-7 2018 The results of flow cytometry using annexin/PI staining of the cells, HMGB1 release, and poly ADP ribose polymerase cleavage demonstrated that theophylline attenuated necrotic effect of berberine and increased the level of apoptotic cell death. Theophylline 143-155 high mobility group box 1 Homo sapiens 70-75 30214593-12 2018 Annexin V-fluorescein isothiocyanate/propidium iodide staining demonstrated that HMGB1 silencing enhanced the effects of carboplatin in inducing the apoptosis of SKOV3-Carb cells relative to HMGB1 non-silenced control cells. Propidium 37-53 high mobility group box 1 Homo sapiens 81-86 30214593-12 2018 Annexin V-fluorescein isothiocyanate/propidium iodide staining demonstrated that HMGB1 silencing enhanced the effects of carboplatin in inducing the apoptosis of SKOV3-Carb cells relative to HMGB1 non-silenced control cells. Carboplatin 121-132 high mobility group box 1 Homo sapiens 81-86 30214593-13 2018 The results of the present study suggested that HMGB1 may be involved in the development of carboplatin resistance in ovarian cancer SKOV3 cells and that HMGB1 silencing may induce the sensitization of carboplatin-resistant ovarian cancer cells to carboplatin. Carboplatin 92-103 high mobility group box 1 Homo sapiens 48-53 30214593-13 2018 The results of the present study suggested that HMGB1 may be involved in the development of carboplatin resistance in ovarian cancer SKOV3 cells and that HMGB1 silencing may induce the sensitization of carboplatin-resistant ovarian cancer cells to carboplatin. Carboplatin 202-213 high mobility group box 1 Homo sapiens 154-159 30214593-13 2018 The results of the present study suggested that HMGB1 may be involved in the development of carboplatin resistance in ovarian cancer SKOV3 cells and that HMGB1 silencing may induce the sensitization of carboplatin-resistant ovarian cancer cells to carboplatin. Carboplatin 202-213 high mobility group box 1 Homo sapiens 154-159 30214593-14 2018 Therefore, HMGB1 may be considered as a potent therapeutic target for increasing the sensitivity of ovarian cancer cells to carboplatin in order to improve the treatment and prognosis of ovarian cancer. Carboplatin 124-135 high mobility group box 1 Homo sapiens 11-16 30214593-6 2018 HMGB1 has been reported to be overexpressed in carboplatin-resistant SKOV3-Carb cells compared with control SKOV3 cells. Carboplatin 47-58 high mobility group box 1 Homo sapiens 0-5 30214593-10 2018 An MTT assay revealed that the proliferation of HMGB1-silenced SKOV3 and SKOV3-Carb cells were decreased compared with the proliferation of non-silenced control cells. monooxyethylene trimethylolpropane tristearate 3-6 high mobility group box 1 Homo sapiens 48-53 30214593-11 2018 Additionally, HMGB1 protein expression levels in SKOV3 cells, but not in SKOV3-Carb cells, were decreased in response to carboplatin treatment. Carboplatin 121-132 high mobility group box 1 Homo sapiens 14-19 30283452-2 2018 The alarmin HMGB1, in its reduced form, can complex with CXCL12 enhancing its activity on monocytes via the chemokine receptor CXCR4, while the form containing a disulfide bond, by binding to TLR2 or TLR4, initiates a cascade of events leading to production of cytokines and chemokines. Disulfides 162-171 high mobility group box 1 Homo sapiens 12-17 28885675-4 2018 In addition, we also investigated the regulatory role of HMGB1 in the sensitivity of NSCLC cells to cisplatin. Cisplatin 100-109 high mobility group box 1 Homo sapiens 57-62 28885675-7 2018 Then the influence by HMGB1 on the chemosensitivity of lung cancer A549 cells was examined with MTT assay and colony forming assay for the A549 cells post the treatment with cisplatin or (and) HMGB1. monooxyethylene trimethylolpropane tristearate 96-99 high mobility group box 1 Homo sapiens 22-27 28885675-7 2018 Then the influence by HMGB1 on the chemosensitivity of lung cancer A549 cells was examined with MTT assay and colony forming assay for the A549 cells post the treatment with cisplatin or (and) HMGB1. Cisplatin 174-183 high mobility group box 1 Homo sapiens 22-27 28885675-10 2018 Recombinant HMGB1 reduced the sensitivity of A549 cells to cisplatin in vitro. Cisplatin 59-68 high mobility group box 1 Homo sapiens 12-17 28885675-12 2018 HMGB1 reduced the sensitivity of NSCLC A549 cells to cisplatin in vitro. Cisplatin 53-62 high mobility group box 1 Homo sapiens 0-5 30283452-3 2018 So far, the possibility that the CXCL12/HMGB1 heterocomplex could be maintained in chronic inflammation was debated, due to the release of reactive oxygen species. Reactive Oxygen Species 139-162 high mobility group box 1 Homo sapiens 40-45 30064906-4 2018 In LPS-induced sepsis model in vivo, administration of KPT330 increased survival rate and ameliorated LPS-induced lung injury, with suppressed levels of TNF-alpha, IL-6 and HMGB1 in the circulation and decreased macrophage and PMN subpopulations in peritoneal cavity. selinexor 55-61 high mobility group box 1 Homo sapiens 173-178 30166062-0 2018 High-mobility group box 1 is responsible for monosodium urate crystal-induced inflammation in human U937 macrophages. Uric Acid 45-61 high mobility group box 1 Homo sapiens 0-25 30166062-2 2018 We performed this study to investigate the role of HMGB1 in the pathogenesis of uric acid-induced inflammation in human U937 macrophages. Uric Acid 80-89 high mobility group box 1 Homo sapiens 51-56 30166062-8 2018 Stimulation of U937 cells with MSU crystals induced translocation of HMGB1 from the nucleus to the cytoplasm and its extracellular release. Uric Acid 31-34 high mobility group box 1 Homo sapiens 69-74 30166062-10 2018 Antioxidants, such as N-acetyl-l-cysteine and quercetin, markedly inhibited the nuclear-to-cytoplasmic translocation of HMGB1 and its release into the extracellular milieu. Acetylcysteine 22-41 high mobility group box 1 Homo sapiens 120-125 30166062-10 2018 Antioxidants, such as N-acetyl-l-cysteine and quercetin, markedly inhibited the nuclear-to-cytoplasmic translocation of HMGB1 and its release into the extracellular milieu. Quercetin 46-55 high mobility group box 1 Homo sapiens 120-125 30166062-11 2018 In conclusion, HMGB1, regulated by the enzymatic activity of caspase-1, is a crucial mediator in uric acid-induced inflammation. Uric Acid 97-106 high mobility group box 1 Homo sapiens 15-20 30208922-0 2018 Retraction Note: HMGB1: a novel protein that induced platelets active and aggregation via Toll-like receptor-4, NF-kappaB and cGMP dependent mechanisms. Cyclic GMP 126-130 high mobility group box 1 Homo sapiens 17-22 30064906-5 2018 In vitro investigations showed that KPT330 dose-dependently inhibited LPS-triggered proinflammatory cytokines production including TNF-alpha, IL-6 and HMGB1 in macrophages. selinexor 36-42 high mobility group box 1 Homo sapiens 151-156 30064906-6 2018 Furthermore, KPT330 treatment significantly suppressed TNF-alpha and IL-6 mRNA expression and inhibited HMGB1 necleocytoplasmic translocation by inhibiting CRM1 distribution. selinexor 13-19 high mobility group box 1 Homo sapiens 104-109 29901427-8 2018 Ferulic acid significantly ameliorated HUVEC radiation injury, as evidenced by increases in cell viability and angiogenesis and decreases in G2/M cell cycle arrest and levels of high mobility group box 1 protein (HMGB1), interleukin (IL)-6 and IL-8. ferulic acid 0-12 high mobility group box 1 Homo sapiens 178-203 29947767-0 2018 High linear energy transfer carbon-ion irradiation increases the release of the immune mediator high mobility group box 1 from human cancer cells. Carbon 28-34 high mobility group box 1 Homo sapiens 96-121 30279967-0 2018 N-(2"-Hydroxyphenyl)-2-propylpentanamide (OH-VPA), a histone deacetylase inhibitor, induces the release of nuclear HMGB1 and modifies ROS levels in HeLa cells. N-(2-hydroxyphenyl)-2-propylpentanamide 0-40 high mobility group box 1 Homo sapiens 115-120 30279967-0 2018 N-(2"-Hydroxyphenyl)-2-propylpentanamide (OH-VPA), a histone deacetylase inhibitor, induces the release of nuclear HMGB1 and modifies ROS levels in HeLa cells. oh-vpa 42-48 high mobility group box 1 Homo sapiens 115-120 30279967-6 2018 Our results show that OH-VPA induces nuclear to cytoplasmic translocation of HMGB1, as demonstrated by confocal microscopy observations and infrared spectra that revealed high quantities of acetylated HMGB1 in HeLa cells. oh-vpa 22-28 high mobility group box 1 Homo sapiens 77-82 30279967-6 2018 Our results show that OH-VPA induces nuclear to cytoplasmic translocation of HMGB1, as demonstrated by confocal microscopy observations and infrared spectra that revealed high quantities of acetylated HMGB1 in HeLa cells. oh-vpa 22-28 high mobility group box 1 Homo sapiens 201-206 28840951-6 2018 The immune/inflammatory changes were more prominent in female drinkers, who showed elevated levels of alcohol danger-associated molecules, such as high mobility group box 1, indicating that there are sex-related differences in the peripheral inflammatory response to alcohol. Alcohols 102-109 high mobility group box 1 Homo sapiens 147-172 28840951-6 2018 The immune/inflammatory changes were more prominent in female drinkers, who showed elevated levels of alcohol danger-associated molecules, such as high mobility group box 1, indicating that there are sex-related differences in the peripheral inflammatory response to alcohol. Alcohols 267-274 high mobility group box 1 Homo sapiens 147-172 29741224-5 2018 Subsequently, HMGB1 inhibitors can reduce the ANG II-elicited polarize of macrophage. Angiotensin II 46-52 high mobility group box 1 Homo sapiens 14-19 27900730-13 2018 The interactions between DNA and HMGB1 are defined by hydrogen bonds and van der Waals contacts. Hydrogen 54-62 high mobility group box 1 Homo sapiens 33-38 29901427-8 2018 Ferulic acid significantly ameliorated HUVEC radiation injury, as evidenced by increases in cell viability and angiogenesis and decreases in G2/M cell cycle arrest and levels of high mobility group box 1 protein (HMGB1), interleukin (IL)-6 and IL-8. ferulic acid 0-12 high mobility group box 1 Homo sapiens 213-218 30157804-13 2018 HMGB1 had good diagnostic ability to differentiate RMPP with AUC of 0.876, sensitivity of 0.833, and specificity of 0.824 compared with TNF-alpha and IL-6. rmpp 51-55 high mobility group box 1 Homo sapiens 0-5 30157804-17 2018 CONCLUSIONS: HMGB1 is a good diagnostic biomarker for differentiating RMPP and NRMPP. rmpp 70-74 high mobility group box 1 Homo sapiens 13-18 30157958-7 2018 HMGB1 knockdown in MM cells enhanced the inhibitory effect of chemotherapy with dexamethasone (Dex) via apoptosis induction. Dexamethasone 80-93 high mobility group box 1 Homo sapiens 0-5 30157958-7 2018 HMGB1 knockdown in MM cells enhanced the inhibitory effect of chemotherapy with dexamethasone (Dex) via apoptosis induction. Dexamethasone 95-98 high mobility group box 1 Homo sapiens 0-5 30157804-4 2018 The present study aimed to explore the role and clinical significance of HMGB1 in children with RMPP and the potential mechanism of HMGB1 expression. rmpp 96-100 high mobility group box 1 Homo sapiens 73-78 30157804-12 2018 HMGB1, TNF-alpha, and IL-6 levels were significantly higher in RMPP cases compared with NRMPP cases (all p < 0.05). rmpp 63-67 high mobility group box 1 Homo sapiens 0-5 30157804-19 2018 Further in vitro and in vivo studies are needed for the development of a new treatment strategy to inhibit the HMGB1 pathway, thereby preventing the inflammation in RMPP. rmpp 165-169 high mobility group box 1 Homo sapiens 111-116 30157958-8 2018 Furthermore, downregulation of HMGB1 activated the mTOR pathway, inhibited autophagy and increased DNA damage induced by Dex by modulating expression of related genes. Dexamethasone 121-124 high mobility group box 1 Homo sapiens 31-36 30157958-10 2018 CONCLUSIONS: Our research shows that HMGB1 participates in autophagy and DNA damage repair and that downregulation of HMGB1 enhances the sensitivity of MM cells to Dex, suggesting that HMGB1 may serve as a target for MM treatment. Dexamethasone 164-167 high mobility group box 1 Homo sapiens 118-123 30096430-0 2018 WITHDRAWN: Paeoniflorin attenuates LPS-induced inflammation in nucleus pulposus cells via Nrf-2/HO-1/HMGB1/NF-kappaB pathway. peoniflorin 11-23 high mobility group box 1 Homo sapiens 101-106 30157958-10 2018 CONCLUSIONS: Our research shows that HMGB1 participates in autophagy and DNA damage repair and that downregulation of HMGB1 enhances the sensitivity of MM cells to Dex, suggesting that HMGB1 may serve as a target for MM treatment. Dexamethasone 164-167 high mobility group box 1 Homo sapiens 118-123 30210495-2 2018 In cultured primary bone marrow-derived cells (BMDCs) stimulated with HMGB1, the number of cells with macrophage-like morphology was markedly increased in association with an increased expression of CD11b/Mac-1, which were attenuated in cells pre-treated with Zileuton, a 5-LO inhibitor as well as in 5-LO-deficient BMDCs. zileuton 260-268 high mobility group box 1 Homo sapiens 70-75 30126243-8 2018 The study on the recognition and binding of the HMGB-1 protein to cisplatin or Pd1 modified DNA probes have shown that HMG proteins are less involved in the palladium agent cytotoxicity. Cisplatin 66-75 high mobility group box 1 Homo sapiens 48-54 29031614-8 2018 Interfering with disulfide HMGB1-activated cell signaling mediates significant therapeutic effects in epilepsy models. Disulfides 17-26 high mobility group box 1 Homo sapiens 27-32 30081847-0 2018 Intermittent high glucose-induced oxidative stress modulates retinal pigmented epithelial cell autophagy and promotes cell survival via increased HMGB1. Glucose 18-25 high mobility group box 1 Homo sapiens 146-151 30081847-7 2018 The expression of HMGB1 was detected by immunohistochemistry.Cells were pre-incubated with HMGB1 inhibitor ethyl pyruvate (EP) ,then detected the expression pattern of autophagic markers and level of cellular ROS. ethyl pyruvate 107-121 high mobility group box 1 Homo sapiens 91-96 29803174-0 2018 miR-505 enhances doxorubicin-induced cytotoxicity in hepatocellular carcinoma through repressing the Akt pathway by directly targeting HMGB1. Doxorubicin 17-28 high mobility group box 1 Homo sapiens 135-140 29803174-8 2018 We found that HMGB1 knockdown and miR-505 overexpression exacerbated ADM-induced cell viability inhibition, enhanced ADM-induced apoptosis, and increased caspase-3 activity in ADM-treated HCC cells. Doxorubicin 69-72 high mobility group box 1 Homo sapiens 14-19 29930000-0 2018 Inorganic Polyphosphate Amplifies High Mobility Group Box 1-Mediated Von Willebrand Factor Release and Platelet String Formation on Endothelial Cells. inorganic polyphosphate 0-23 high mobility group box 1 Homo sapiens 34-59 29763588-0 2018 Glycyrrhizin ameliorates inflammatory pain by inhibiting microglial activation-mediated inflammatory response via blockage of the HMGB1-TLR4-NF-kB pathway. Glycyrrhizic Acid 0-12 high mobility group box 1 Homo sapiens 130-135 29704473-8 2018 With regard to cardiovascular risk, levels of serum HMGB1 were positively associated with 10-year CHD risk (beta coefficient 0.506, 95% CI 0.030 to 0.983, p = .037), independent of patients" undiagnosed abnormal glucose regulation. Glucose 212-219 high mobility group box 1 Homo sapiens 52-57 29763588-8 2018 Furthermore, glycyrrhizin dampened the activation of subsequent TLR4-NF-kappaB pathway in LPS-stimulated microglia, which was abrogated by HMGB1 elevation. Glycyrrhizic Acid 13-25 high mobility group box 1 Homo sapiens 139-144 29737584-0 2018 Metformin increases the cytotoxicity of oxaliplatin in human DLD-1 colorectal cancer cells through down-regulating HMGB1 expression. Metformin 0-9 high mobility group box 1 Homo sapiens 115-120 29737584-0 2018 Metformin increases the cytotoxicity of oxaliplatin in human DLD-1 colorectal cancer cells through down-regulating HMGB1 expression. Oxaliplatin 40-51 high mobility group box 1 Homo sapiens 115-120 29737584-7 2018 In this study, we examined whether HMGB1 plays a role in the OXA- and/or metformin-induced cytotoxic effect on CRC cells. Metformin 73-82 high mobility group box 1 Homo sapiens 35-40 29737584-8 2018 The results showed that treatment with OXA increased HMGB1 expression in the ERK1/2- and Akt-dependent manners in DLD-1 cells. Oxaliplatin 39-42 high mobility group box 1 Homo sapiens 53-58 29737584-11 2018 Compared to a single agent, OXA combined with metformin administration resulted in cytotoxicity and cell growth inhibition synergistically, accompanied with reduced HMGB1 level. Oxaliplatin 28-31 high mobility group box 1 Homo sapiens 165-170 29737584-11 2018 Compared to a single agent, OXA combined with metformin administration resulted in cytotoxicity and cell growth inhibition synergistically, accompanied with reduced HMGB1 level. Metformin 46-55 high mobility group box 1 Homo sapiens 165-170 29676481-5 2018 Furthermore, therapeutic targeting of HMGB1 with either anti-HMGB1 antibodies or selective inhibitors, such as glycyrrhizin, has been shown to inhibit neurodegeneration in animal models. Glycyrrhizic Acid 111-123 high mobility group box 1 Homo sapiens 38-43 29997173-3 2018 CD52-Fc bound specifically to the proinflammatory Box B domain of HMGB1, and this in turn promoted binding of the CD52 N-linked glycan, in alpha-2,3 sialic acid linkage with galactose, to Siglec-10. n-linked glycan 119-134 high mobility group box 1 Homo sapiens 66-71 29715512-0 2018 HMGB1 contributes to adriamycin-induced cardiotoxicity via up-regulating autophagy. Doxorubicin 21-31 high mobility group box 1 Homo sapiens 0-5 29909245-0 2018 Corrigendum to "Immunosuppressant cytoprotection correlates with HMGB1 suppression in primary astrocyte cultures exposed to combined oxygen-glucose deprivation" [Pharmacol. oxygen-glucose 133-147 high mobility group box 1 Homo sapiens 65-70 30061484-8 2018 Sulphated K5 carbohydrate fragments inhibited RAGE binding to amyloid beta-peptide and HMGB1. Carbohydrates 13-25 high mobility group box 1 Homo sapiens 87-92 30061484-11 2018 Further, glycyrrhizin inhibited HMGB1 and HMGB1 A-box binding to heparin. Glycyrrhizic Acid 9-21 high mobility group box 1 Homo sapiens 32-37 30061484-11 2018 Further, glycyrrhizin inhibited HMGB1 and HMGB1 A-box binding to heparin. Glycyrrhizic Acid 9-21 high mobility group box 1 Homo sapiens 42-47 30061484-11 2018 Further, glycyrrhizin inhibited HMGB1 and HMGB1 A-box binding to heparin. Heparin 65-72 high mobility group box 1 Homo sapiens 32-37 30061484-11 2018 Further, glycyrrhizin inhibited HMGB1 and HMGB1 A-box binding to heparin. Heparin 65-72 high mobility group box 1 Homo sapiens 42-47 30123419-3 2018 In this study we established a representative panel of patient derived xenograft (PDX) mouse models from patients with renal cell carcinomas and determined serum levels of high mobility group B1 (HMGB1) protein under treatment with sunitinib, pazopanib, sorafenib, axitinib, temsirolimus and bevacizumab. Sorafenib 254-263 high mobility group box 1 Homo sapiens 196-201 30123419-3 2018 In this study we established a representative panel of patient derived xenograft (PDX) mouse models from patients with renal cell carcinomas and determined serum levels of high mobility group B1 (HMGB1) protein under treatment with sunitinib, pazopanib, sorafenib, axitinib, temsirolimus and bevacizumab. Axitinib 265-273 high mobility group box 1 Homo sapiens 196-201 30123419-3 2018 In this study we established a representative panel of patient derived xenograft (PDX) mouse models from patients with renal cell carcinomas and determined serum levels of high mobility group B1 (HMGB1) protein under treatment with sunitinib, pazopanib, sorafenib, axitinib, temsirolimus and bevacizumab. temsirolimus 275-287 high mobility group box 1 Homo sapiens 196-201 29997173-3 2018 CD52-Fc bound specifically to the proinflammatory Box B domain of HMGB1, and this in turn promoted binding of the CD52 N-linked glycan, in alpha-2,3 sialic acid linkage with galactose, to Siglec-10. N-Acetylneuraminic Acid 149-160 high mobility group box 1 Homo sapiens 66-71 29997173-3 2018 CD52-Fc bound specifically to the proinflammatory Box B domain of HMGB1, and this in turn promoted binding of the CD52 N-linked glycan, in alpha-2,3 sialic acid linkage with galactose, to Siglec-10. Galactose 174-183 high mobility group box 1 Homo sapiens 66-71 29743526-4 2018 It was further shown that HQH administration reduced cisplatin-induced release and nuclear-cytoplasmic translocation of HMGB1 and inactivated its downstream signaling molecules, TLR4 and NFkappaB, in renal tubular cells; as a result, HQH repressed cisplatin-induced TNF-alpha production. Cisplatin 53-62 high mobility group box 1 Homo sapiens 120-125 29751935-0 2018 Down-Regulation of miR-218-5p Promotes Apoptosis of Human Umbilical Vein Endothelial Cells Through Regulating High-Mobility Group Box-1 in Henoch-Schonlein Purpura. mir-218-5p 19-29 high mobility group box 1 Homo sapiens 110-135 29751935-6 2018 The association between miR-218-5p and HMGB1 was determined by luciferase assay. mir-218-5p 24-34 high mobility group box 1 Homo sapiens 39-44 29751935-11 2018 Furthermore, the miR-218-5p mimic demonstrated an inhibitory effect on the apoptosis of HUVECs co-cultured in PBMC supernatant, which was reversed by over expression of HMGB1. mir-218-5p 17-27 high mobility group box 1 Homo sapiens 169-174 29573352-3 2018 The results showed that administration of 7.5 mg/kg alpha-HMGB1 1 h after IAV (A/Puerto Rico/8/34) inoculation significantly alleviated brain edema at 48 h after IAV inoculation, as confirmed by the suppression of Evans Blue dye leakage and matrix metallopeptidase-9 mRNA expression in the brain. Evans Blue 214-228 high mobility group box 1 Homo sapiens 58-63 29795370-7 2018 HMGB1 dose-dependently enhanced the expression of CD80 and CD86 on DCs in vitro, and intravenous or oral administration of glycyrrhizin, an HMGB1 inhibitor, significantly suppressed activation of mucosal DCs and induction of intestinal OVA-specific CTLs and IgA by oral CT administration. Glycyrrhizic Acid 123-135 high mobility group box 1 Homo sapiens 0-5 29795370-7 2018 HMGB1 dose-dependently enhanced the expression of CD80 and CD86 on DCs in vitro, and intravenous or oral administration of glycyrrhizin, an HMGB1 inhibitor, significantly suppressed activation of mucosal DCs and induction of intestinal OVA-specific CTLs and IgA by oral CT administration. Glycyrrhizic Acid 123-135 high mobility group box 1 Homo sapiens 140-145 29899164-0 2018 miRNA-1284, a regulator of HMGB1, inhibits cell proliferation and migration in osteosarcoma. mirna-1284 0-10 high mobility group box 1 Homo sapiens 27-32 29773200-0 2018 Lithium chloride attenuates mitomycin C induced necrotic cell death in MDA-MB-231 breast cancer cells via HMGB1 and Bax signaling. Lithium Chloride 0-16 high mobility group box 1 Homo sapiens 106-111 29773200-0 2018 Lithium chloride attenuates mitomycin C induced necrotic cell death in MDA-MB-231 breast cancer cells via HMGB1 and Bax signaling. Mitomycin 28-39 high mobility group box 1 Homo sapiens 106-111 29773200-8 2018 LiCl combined with mitomycin C significantly down-regulated HMGB1, MMP9 and Bcl-2 gene expression but significantly increased the level of Bax protein. Lithium Chloride 0-4 high mobility group box 1 Homo sapiens 60-65 29773200-8 2018 LiCl combined with mitomycin C significantly down-regulated HMGB1, MMP9 and Bcl-2 gene expression but significantly increased the level of Bax protein. Mitomycin 19-30 high mobility group box 1 Homo sapiens 60-65 29773200-9 2018 In addition, the content of HMGB1 in the nuclei decreased and pretreatment with LiCl reduced the content of HMGB1 release induced by MMC. Lithium Chloride 80-84 high mobility group box 1 Homo sapiens 108-113 29773200-9 2018 In addition, the content of HMGB1 in the nuclei decreased and pretreatment with LiCl reduced the content of HMGB1 release induced by MMC. Mitomycin 133-136 high mobility group box 1 Homo sapiens 108-113 29791130-10 2018 The concentration of HMGB1 in citrated and EDTA-treated plasma from the same healthy control subjects was below the limit of detection of our assay (1 ng/mL), confirming that HMGB1 in serum arises when blood is allowed to clot. Edetic Acid 43-47 high mobility group box 1 Homo sapiens 21-26 29791130-10 2018 The concentration of HMGB1 in citrated and EDTA-treated plasma from the same healthy control subjects was below the limit of detection of our assay (1 ng/mL), confirming that HMGB1 in serum arises when blood is allowed to clot. Edetic Acid 43-47 high mobility group box 1 Homo sapiens 175-180 29292841-6 2018 In vitro, the exposure of normal human bronchial epithelial cells to HMGB1 resulted in the upregulation of GRP75, proinflammatory cytokine production, enhanced ER-Mitochondrial Ca2+ transfer, and enhancement of reactive oxygen species (ROS). Reactive Oxygen Species 236-239 high mobility group box 1 Homo sapiens 69-74 29356044-9 2018 High expression levels of HMGB1 as total (P = 0.0011) and cytoplasmic score (P = 0.0462) were related to a worse disease-specific survival (DSS) in the entire cohort and in the clinicopathological subgroups. dss 140-143 high mobility group box 1 Homo sapiens 26-31 29292841-6 2018 In vitro, the exposure of normal human bronchial epithelial cells to HMGB1 resulted in the upregulation of GRP75, proinflammatory cytokine production, enhanced ER-Mitochondrial Ca2+ transfer, and enhancement of reactive oxygen species (ROS). Reactive Oxygen Species 211-234 high mobility group box 1 Homo sapiens 69-74 29292841-8 2018 Sequentially, pretreatment with 2-APB, SKF960365 (SKF) and Ru360 which inhibit ER-Mitochondrial Ca2+ transfer significantly lowered HMGB1-induced the generation of ROS and the release of Th2 cytokines in 16HBE cells. 2-aminoethoxydiphenyl borate 32-37 high mobility group box 1 Homo sapiens 132-137 29292841-8 2018 Sequentially, pretreatment with 2-APB, SKF960365 (SKF) and Ru360 which inhibit ER-Mitochondrial Ca2+ transfer significantly lowered HMGB1-induced the generation of ROS and the release of Th2 cytokines in 16HBE cells. Reactive Oxygen Species 164-167 high mobility group box 1 Homo sapiens 132-137 29329282-3 2018 The role of HMGB1 in RSV infection was explored using glycyrrhizin, a selective HMGB1 inhibitor.ResultsRSV infection strongly induced HMGB1 expression both in vitro and in vivo. Glycyrrhizic Acid 54-66 high mobility group box 1 Homo sapiens 80-85 29395576-3 2018 OBJECTIVE: To evaluate the effect of reduced-HMGB1 (previously termed chemoattractive-HMGB1) on polyinosine-polycytidylic acid [poly(I:C)]-induced inflammation in normal human keratinocytes (NHKs). polyinosine-polycytidylic acid 96-126 high mobility group box 1 Homo sapiens 86-91 29395576-5 2018 An immunoprecipitation was performed to know whether HMGB1 could bind to poly(I:C), and immunofluorescence staining and flow cytometric analysis were performed to check whether reduced-HMGB interferes with cellular uptake of poly(I:C) translocation (possibly by endocytosis). Poly I-C 73-81 high mobility group box 1 Homo sapiens 53-58 29395576-5 2018 An immunoprecipitation was performed to know whether HMGB1 could bind to poly(I:C), and immunofluorescence staining and flow cytometric analysis were performed to check whether reduced-HMGB interferes with cellular uptake of poly(I:C) translocation (possibly by endocytosis). Poly I-C 73-82 high mobility group box 1 Homo sapiens 53-58 29395576-6 2018 RESULTS: Application of exogenous HMGB1 before, but not after, exerted a suppressive effect on poly(I:C)-induced inflammation in NHKs. Poly I-C 95-104 high mobility group box 1 Homo sapiens 34-39 29395576-7 2018 In addition, reduced-HMGB1, but not disulfide-HMGB1, exerted a suppressive effect on poly(I:C)-induced inflammation in NHKs, suggesting the importance of the redox status of exogenous HMGB1. Poly I-C 85-94 high mobility group box 1 Homo sapiens 21-26 29395576-9 2018 Disulfide-HMGB1 formed a complex with poly(I:C), as did reduced- and oxidized-HMGB1, albeit to a lesser extent. Disulfides 0-9 high mobility group box 1 Homo sapiens 10-15 29395576-11 2018 CONCLUSION: These findings suggest that pre-treatment with reduced-HMGB1 ameliorates poly(I:C)-mediated inflammation in NHKs. Poly I-C 85-94 high mobility group box 1 Homo sapiens 67-72 29395576-0 2018 Reduced-HMGB1 suppresses poly(I:C)-induced inflammation in keratinocytes. Poly I-C 25-34 high mobility group box 1 Homo sapiens 8-13 29395576-3 2018 OBJECTIVE: To evaluate the effect of reduced-HMGB1 (previously termed chemoattractive-HMGB1) on polyinosine-polycytidylic acid [poly(I:C)]-induced inflammation in normal human keratinocytes (NHKs). polyinosine-polycytidylic acid 96-126 high mobility group box 1 Homo sapiens 45-50 29329282-3 2018 The role of HMGB1 in RSV infection was explored using glycyrrhizin, a selective HMGB1 inhibitor.ResultsRSV infection strongly induced HMGB1 expression both in vitro and in vivo. Glycyrrhizic Acid 54-66 high mobility group box 1 Homo sapiens 80-85 29329282-4 2018 Glycyrrhizin dose-dependently inhibited HMGB1 upregulation in both RSV-infected immortalized and primary human bronchial epithelial cells, and this effect was associated with significant reduction of viral replication.ConclusionOur data suggest that HMGB1 expression increases during RSV replication. Glycyrrhizic Acid 0-12 high mobility group box 1 Homo sapiens 40-45 29329282-4 2018 Glycyrrhizin dose-dependently inhibited HMGB1 upregulation in both RSV-infected immortalized and primary human bronchial epithelial cells, and this effect was associated with significant reduction of viral replication.ConclusionOur data suggest that HMGB1 expression increases during RSV replication. Glycyrrhizic Acid 0-12 high mobility group box 1 Homo sapiens 250-255 29531138-4 2018 In vitro analysis indicated that HMGB1, granulocyte-macrophage colony-stimulating factor (GM-CSF), and interleukin-1beta (IL-1beta) were secreted in response to tumor necrosis factor-alpha (TNF-alpha) stimuli in human gingival epithelial cells (HGECs) and human monocytic leukemia cells (THP-1) treated with phorbol myristate acetate. Tetradecanoylphorbol Acetate 308-333 high mobility group box 1 Homo sapiens 33-38 29922279-0 2018 Staphylococcus aureus Phenol-Soluble Modulins alpha1-alpha3 Act as Novel Toll-Like Receptor (TLR) 4 Antagonists to Inhibit HMGB1/TLR4/NF-kappaB Signaling Pathway. Phenol 22-28 high mobility group box 1 Homo sapiens 123-128 29853785-6 2018 The plasma high-mobility group box 1 level decreased significantly from 51.7 (58.1) to 33.9 (45.0) ng.ml-1 (p < 0.05) in the dexmedetomidine group, which was not observed in the saline group. Dexmedetomidine 128-143 high mobility group box 1 Homo sapiens 11-36 29850505-7 2018 The matrix metalloproteinase-2 (MMP-2) expression and activity were upregulated after treatment with HMGB1, while the upregulated expression of MMP-2 stimulated by HMGB1 in lung cancer cells was significantly reduced with the blockage of si-p65. Silicon 45-47 high mobility group box 1 Homo sapiens 101-106 29850505-7 2018 The matrix metalloproteinase-2 (MMP-2) expression and activity were upregulated after treatment with HMGB1, while the upregulated expression of MMP-2 stimulated by HMGB1 in lung cancer cells was significantly reduced with the blockage of si-p65. Silicon 45-47 high mobility group box 1 Homo sapiens 164-169 29330155-0 2018 Amlexanox Inhibits Cerebral Ischemia-Induced Delayed Astrocytic High-Mobility Group Box 1 Release and Subsequent Brain Damage. amlexanox 0-9 high mobility group box 1 Homo sapiens 64-89 29436639-6 2018 GL suppressed HMGB1-induced monocyte migration and increased HMGB1-inhibited monocyte apoptosis. Glycyrrhizic Acid 0-2 high mobility group box 1 Homo sapiens 61-66 29393419-0 2018 HMGB1 mediates microglia activation via the TLR4/NF-kappaB pathway in coriaria lactone induced epilepsy. coriaria lactone 70-86 high mobility group box 1 Homo sapiens 0-5 29393419-8 2018 The potential toxic effect of HMGB1 on HM cells was evaluated by MTT and 5-ethynyl-2-deoxyuridine assays. monooxyethylene trimethylolpropane tristearate 65-68 high mobility group box 1 Homo sapiens 30-35 29393419-8 2018 The potential toxic effect of HMGB1 on HM cells was evaluated by MTT and 5-ethynyl-2-deoxyuridine assays. 5-ethynyl-2'-deoxyuridine 73-97 high mobility group box 1 Homo sapiens 30-35 29436639-0 2018 Glycyrrhizin affects monocyte migration and apoptosis by blocking HMGB1 signaling. Glycyrrhizic Acid 0-12 high mobility group box 1 Homo sapiens 66-71 29436639-3 2018 Glycyrrhizin (GL), an effective component of licorice, weakens the proinflammatory effect of HMGB1. Glycyrrhizic Acid 0-12 high mobility group box 1 Homo sapiens 93-98 29436639-3 2018 Glycyrrhizin (GL), an effective component of licorice, weakens the proinflammatory effect of HMGB1. Glycyrrhizic Acid 14-16 high mobility group box 1 Homo sapiens 93-98 29436639-4 2018 The present study investigated the effect of GL on the migration and apoptosis of monocytes associated with HMGB1 signaling. Glycyrrhizic Acid 45-47 high mobility group box 1 Homo sapiens 108-113 29436639-6 2018 GL suppressed HMGB1-induced monocyte migration and increased HMGB1-inhibited monocyte apoptosis. Glycyrrhizic Acid 0-2 high mobility group box 1 Homo sapiens 14-19 29330155-4 2018 HMGB1 depletion from neuronal nuclei was observed within 3 hours after transient middle cerebral artery occlusion (tMCAO), whereas the intracerebroventricular (i.c.v.) tmcao 115-120 high mobility group box 1 Homo sapiens 0-5 29330155-5 2018 pretreatment with amlexanox blocked HMGB1 release from neurons, resulting in HMGB1 redistribution in the nuclei and cytoplasm. amlexanox 18-27 high mobility group box 1 Homo sapiens 36-41 29330155-5 2018 pretreatment with amlexanox blocked HMGB1 release from neurons, resulting in HMGB1 redistribution in the nuclei and cytoplasm. amlexanox 18-27 high mobility group box 1 Homo sapiens 77-82 29330155-6 2018 HMGB1 was selectively released from astrocytes 27 hours after tMCAO and this HMGB1 release was blocked by late treatment with amlexanox (i.c.v.) tmcao 62-67 high mobility group box 1 Homo sapiens 0-5 29330155-6 2018 HMGB1 was selectively released from astrocytes 27 hours after tMCAO and this HMGB1 release was blocked by late treatment with amlexanox (i.c.v.) amlexanox 126-135 high mobility group box 1 Homo sapiens 0-5 29330155-6 2018 HMGB1 was selectively released from astrocytes 27 hours after tMCAO and this HMGB1 release was blocked by late treatment with amlexanox (i.c.v.) amlexanox 126-135 high mobility group box 1 Homo sapiens 77-82 29330155-8 2018 Proximity extension assay revealed that the HMGB1 level was elevated in the CSF at 3 and 27 hours after tMCAO. tmcao 104-109 high mobility group box 1 Homo sapiens 44-49 29330155-11 2018 with anti-HMGB1 antibody 24 hours after tMCAO also ameliorated ischemic brain damage 48 hours after tMCAO. tmcao 40-45 high mobility group box 1 Homo sapiens 10-15 29330155-11 2018 with anti-HMGB1 antibody 24 hours after tMCAO also ameliorated ischemic brain damage 48 hours after tMCAO. tmcao 100-105 high mobility group box 1 Homo sapiens 10-15 29330155-12 2018 Thus, the inhibition of brain damage by late treatment with amlexanox or anti-HMGB1 antibody indicates that late HMGB1 release plays a role in the maintenance of stroke-induced brain damage, and the inhibition of this release would be a novel therapeutic target for protection of ischemic brain damage. amlexanox 60-69 high mobility group box 1 Homo sapiens 113-118 29541234-3 2018 Papaverine significantly inhibited RAGE-dependent nuclear factor kappa-B activation driven by high mobility group box-1, a RAGE ligand. Papaverine 0-10 high mobility group box 1 Homo sapiens 94-119 29673663-9 2018 (3) HMGB1 and RAGE immunoreactivity in pPROM placenta TMA were increased in the cytoplasm of syncytiotrophoblast (STB), extravillous trophoblast (EVT) and mesenchymal cells, while NF-kappaBp65 was enhanced in the nucleus of STB and EVT. EVT 146-149 high mobility group box 1 Homo sapiens 4-9 29673663-9 2018 (3) HMGB1 and RAGE immunoreactivity in pPROM placenta TMA were increased in the cytoplasm of syncytiotrophoblast (STB), extravillous trophoblast (EVT) and mesenchymal cells, while NF-kappaBp65 was enhanced in the nucleus of STB and EVT. EVT 232-235 high mobility group box 1 Homo sapiens 4-9 29665925-5 2018 Meanwhile, the protein expression levels of HMGB1 in peripheral blood serum and mononuclear cells(PBMNC) of 25 patients with chronic and refractory ITP were detected by ELISA and Western blot. Inosine Triphosphate 148-151 high mobility group box 1 Homo sapiens 44-49 29665925-10 2018 The HMGB1 expression positive rate in spleen of ITP patients was significantly higher than that in control group (85.0% vs 15.0%)(P<0.001). Inosine Triphosphate 48-51 high mobility group box 1 Homo sapiens 4-9 29665925-13 2018 CONCLUSION: The splenectomy has been found to be effective therapeutic method for patients with ITP, the HMGB1 highly express in the spleen of the patients with chronic refractory ITP, but negatively correlats with the therapeutic outcome after splenectomy. Inosine Triphosphate 96-99 high mobility group box 1 Homo sapiens 105-110 29540727-7 2018 In addition, 1 ng/ml HMGB1 increased the percentage of filopodia formation using phalloidin staining. Phalloidine 81-91 high mobility group box 1 Homo sapiens 21-26 29599781-13 2018 Finally, we detected several subunits of the proteasome (PSMB9, PSMB10) as well as the danger signal HMGB1 exclusively within apoptotic cell-released LEVs. levs 150-154 high mobility group box 1 Homo sapiens 101-106 29588304-7 2018 Nevertheless, according to fluorescence-activated cell sorter analysis, VLD rivaroxaban treatment resulted in a significantly reduced expression of platelet HMGB-1, whereas P-selectin exposure was not affected. Rivaroxaban 76-87 high mobility group box 1 Homo sapiens 157-163 29535197-14 2018 Towards development of a new therapeutic modality, we show herein that blocking serum HMGB1 levels by Eritoran improves lung pathology in influenza B virus-infected cotton rats. eritoran 102-110 high mobility group box 1 Homo sapiens 86-91 29361549-3 2018 In this study, we studied the effect of inflachromene (ICM), a novel HMGB1 secretion inhibitor, on autophagy. inflachromene 40-53 high mobility group box 1 Homo sapiens 69-74 29316268-9 2018 Pharmacological inhibition of HMGB1 Cyt translocation with ethyl pyruvate prevented HBx-induced autophagy. ethyl pyruvate 59-73 high mobility group box 1 Homo sapiens 30-35 29197622-4 2018 Mechanistic study showed that dioscin significantly up-regulated the expression levels of Sirt3, SOD2, and then suppressed inflammation by decreasing the expression levels of NF-kB, HMGB1, c-Jun, c-Fos, COX2, TNF-alpha, IL-1beta and IL-6. dioscin 30-37 high mobility group box 1 Homo sapiens 182-187 29536989-0 2018 Corrigendum: 1,25-Dihydroxyvitamin D Inhibits LPS-Induced High-Mobility Group Box 1 (HMGB1) Secretion via Targeting the NF-E2-Related Factor 2-Hemeoxygenase-1-HMGB1 Pathway in Macrophages. 1,25-dihydroxyvitamin D 13-36 high mobility group box 1 Homo sapiens 58-83 29536989-0 2018 Corrigendum: 1,25-Dihydroxyvitamin D Inhibits LPS-Induced High-Mobility Group Box 1 (HMGB1) Secretion via Targeting the NF-E2-Related Factor 2-Hemeoxygenase-1-HMGB1 Pathway in Macrophages. 1,25-dihydroxyvitamin D 13-36 high mobility group box 1 Homo sapiens 85-90 29536989-0 2018 Corrigendum: 1,25-Dihydroxyvitamin D Inhibits LPS-Induced High-Mobility Group Box 1 (HMGB1) Secretion via Targeting the NF-E2-Related Factor 2-Hemeoxygenase-1-HMGB1 Pathway in Macrophages. 1,25-dihydroxyvitamin D 13-36 high mobility group box 1 Homo sapiens 159-164 29447008-7 2018 Expert opinion: Blocking excessive amounts of extracellular HMGB1, particularly the disulfide isoform, offers an attractive clinical opportunity to ameliorate systemic inflammatory diseases. Disulfides 84-93 high mobility group box 1 Homo sapiens 60-65 29328447-2 2018 Western blot analysis revealed that vincristine (VCR), etoposide (ETO) and carboplatin (CBP) significantly increased the expression of HMGB1 in Weri-Rb-1 and Y79 cells compared with the untreated control (P<0.01). Vincristine 36-47 high mobility group box 1 Homo sapiens 135-140 29328447-2 2018 Western blot analysis revealed that vincristine (VCR), etoposide (ETO) and carboplatin (CBP) significantly increased the expression of HMGB1 in Weri-Rb-1 and Y79 cells compared with the untreated control (P<0.01). Etoposide 55-64 high mobility group box 1 Homo sapiens 135-140 29328447-2 2018 Western blot analysis revealed that vincristine (VCR), etoposide (ETO) and carboplatin (CBP) significantly increased the expression of HMGB1 in Weri-Rb-1 and Y79 cells compared with the untreated control (P<0.01). Etoposide 66-69 high mobility group box 1 Homo sapiens 135-140 29328447-2 2018 Western blot analysis revealed that vincristine (VCR), etoposide (ETO) and carboplatin (CBP) significantly increased the expression of HMGB1 in Weri-Rb-1 and Y79 cells compared with the untreated control (P<0.01). Carboplatin 75-86 high mobility group box 1 Homo sapiens 135-140 29197515-6 2018 In addition, Fingolimod treatment reduced serum levels of HMGB1 by 71.6% (P = 0.02) and pentosidine serum levels by 41.3% (P = 0.12). Fingolimod Hydrochloride 13-23 high mobility group box 1 Homo sapiens 58-63 29448108-3 2018 In this study, glycyrrhizin (GL), an inhibitor of HMGB1, was used to investigate whether the inhibition of HMGB1 could modulate microglia/macrophage polarization after TBI. Glycyrrhizic Acid 15-27 high mobility group box 1 Homo sapiens 50-55 29448108-3 2018 In this study, glycyrrhizin (GL), an inhibitor of HMGB1, was used to investigate whether the inhibition of HMGB1 could modulate microglia/macrophage polarization after TBI. Glycyrrhizic Acid 15-27 high mobility group box 1 Homo sapiens 107-112 29448108-3 2018 In this study, glycyrrhizin (GL), an inhibitor of HMGB1, was used to investigate whether the inhibition of HMGB1 could modulate microglia/macrophage polarization after TBI. Glycyrrhizic Acid 29-31 high mobility group box 1 Homo sapiens 50-55 29448108-4 2018 The results showed that treatment with GL improved the neurological function recovery, reduced the lesion volume, and inhibited the release and expression of HMGB1 after TBI. Glycyrrhizic Acid 39-41 high mobility group box 1 Homo sapiens 158-163 29448108-6 2018 In conclusion, the results suggested that GL attenuated TBI by inhibiting M1 phenotype while inducing M2 phenotype activation of microglia/macrophages, at least partly through inhibiting HMGB1. Glycyrrhizic Acid 42-44 high mobility group box 1 Homo sapiens 187-192 28816009-4 2018 After atazanavir sulphate treatment, in A549 cells and HPMECs, the expression of vimentin, HMGB1, Toll-like receptor 4 (TLR-4) and p-NF-kappaB decreased, while the expression of E-cadherin and VE-cadherin increased. Atazanavir Sulfate 6-25 high mobility group box 1 Homo sapiens 91-96 29447234-0 2018 Folic acid derived-P5779 mimetics regulate DAMP-mediated inflammation through disruption of HMGB1:TLR4:MD-2 axes. Folic Acid 0-10 high mobility group box 1 Homo sapiens 92-97 28816009-6 2018 Proliferation of HLF-1 was reduced after atazanavir treatment, meanwhile the expression of hypoxia-inducible factor-1alpha (HIF-1alpha), prolyl hydroxylase domain protein 2 (PHD-2), HMGB1, TLR-9, p-NF-kappaB, collagen I and collagen III was decreased. Atazanavir Sulfate 41-51 high mobility group box 1 Homo sapiens 182-187 28816009-8 2018 In summary, our study supports the proposal that atazanavir sulphate may have a therapeutic potential in reducing the progression of pulmonary fibrosis by suppressing HMGB1/TLR signalling. Atazanavir Sulfate 49-68 high mobility group box 1 Homo sapiens 167-172 28033959-0 2018 Assessment of binding properties of Actinomycin-D to 21nt DNA segment of hmgb1 gene promoter using spectroscopic and calorimetric techniques. Dactinomycin 36-49 high mobility group box 1 Homo sapiens 73-78 29366441-7 2018 Furthermore, hypoxia-induced inflammation by HMGB1 translocation into the cytoplasm results in the release of IL-8 through a ROS-dependent mechanism in upper airway epithelium. ros 125-128 high mobility group box 1 Homo sapiens 45-50 29191973-9 2018 The combined inhibition of IL6R and HMGB1 may serve as a new treatment for enzalutamide resistance in patients with advanced or metastatic prostate cancer. enzalutamide 75-87 high mobility group box 1 Homo sapiens 36-41 29083089-7 2018 Furthermore, both U0126 (ErK inhibitor) and SP600125 (JNK inhibitor) significantly suppressed the enhanced production of HIF-1alpha and VEGF induced by HMGB1. pyrazolanthrone 44-52 high mobility group box 1 Homo sapiens 152-157 29083089-7 2018 Furthermore, both U0126 (ErK inhibitor) and SP600125 (JNK inhibitor) significantly suppressed the enhanced production of HIF-1alpha and VEGF induced by HMGB1. U 0126 18-23 high mobility group box 1 Homo sapiens 152-157 29375570-0 2017 From Human Megakaryocytes to Platelets: Effects of Aspirin on High-Mobility Group Box 1/Receptor for Advanced Glycation End Products Axis. Aspirin 51-58 high mobility group box 1 Homo sapiens 62-87 29241031-0 2018 The protective effect of dexmedetomidine on LPS-induced acute lung injury through the HMGB1-mediated TLR4/NF-kappaB and PI3K/Akt/mTOR pathways. Dexmedetomidine 25-40 high mobility group box 1 Homo sapiens 86-91 29241031-9 2018 Additionally, treatment with DEX inhibited the expression of HMGB1, TLR4, MyD88, p-IkappaB, p-NF-kappaB, p-PI3K, p-Akt and p-mTOR in vivo and in vitro. Dexmedetomidine 29-32 high mobility group box 1 Homo sapiens 61-66 29241031-11 2018 Treatment with glycyrrhizin, an inhibitor of HMGB1, confirmed that HMGB1 was involved in the mechanism of DEX on LPS-induced ALI. Glycyrrhizic Acid 15-27 high mobility group box 1 Homo sapiens 45-50 29241031-11 2018 Treatment with glycyrrhizin, an inhibitor of HMGB1, confirmed that HMGB1 was involved in the mechanism of DEX on LPS-induced ALI. Glycyrrhizic Acid 15-27 high mobility group box 1 Homo sapiens 67-72 29241031-11 2018 Treatment with glycyrrhizin, an inhibitor of HMGB1, confirmed that HMGB1 was involved in the mechanism of DEX on LPS-induced ALI. Dexmedetomidine 106-109 high mobility group box 1 Homo sapiens 45-50 29241031-11 2018 Treatment with glycyrrhizin, an inhibitor of HMGB1, confirmed that HMGB1 was involved in the mechanism of DEX on LPS-induced ALI. Dexmedetomidine 106-109 high mobility group box 1 Homo sapiens 67-72 29386662-6 2018 DHA-treated breast cancer cells triggered increased caspase-1and gasdermin D activation, enhanced IL-1beta secretion, translocated HMGB1 towards the cytoplasm, and membrane pore formation when compared to untreated cells, suggesting DHA induces pyroptosis programmed cell death in breast cancer cells. Docosahexaenoic Acids 0-3 high mobility group box 1 Homo sapiens 131-136 30154286-1 2018 OBJECTIVE: To determine mechanisms for the role of high mobility group box-1 (HMGB1) to platinum based chemo-sensitivity in cervical cancer. Platinum 88-96 high mobility group box 1 Homo sapiens 51-76 30154286-1 2018 OBJECTIVE: To determine mechanisms for the role of high mobility group box-1 (HMGB1) to platinum based chemo-sensitivity in cervical cancer. Platinum 88-96 high mobility group box 1 Homo sapiens 78-83 29568761-0 2018 Glycyrrhizin Suppresses the Growth of Human NSCLC Cell Line HCC827 by Downregulating HMGB1 Level. Glycyrrhizic Acid 0-12 high mobility group box 1 Homo sapiens 85-90 29568761-4 2018 And high level of HMGB1 promoted the migration and invasion of lung cancer cells, which was suppressed by glycyrrhizin. Glycyrrhizic Acid 106-118 high mobility group box 1 Homo sapiens 18-23 29568761-5 2018 Moreover, glycyrrhizin reduced the activity of JAK/STAT signaling pathway, which is the upstream regulator of HMGB1. Glycyrrhizic Acid 10-22 high mobility group box 1 Homo sapiens 110-115 29375570-4 2017 Aspirin "in vivo" and "in vitro" not only reduces HMGB1 and receptor for advanced glycation end products expression on MKs and PLTs but also drives the movement of HMGB1 from MKs into PLTs and PLT-derived MV. Aspirin 0-7 high mobility group box 1 Homo sapiens 50-55 29375570-4 2017 Aspirin "in vivo" and "in vitro" not only reduces HMGB1 and receptor for advanced glycation end products expression on MKs and PLTs but also drives the movement of HMGB1 from MKs into PLTs and PLT-derived MV. Aspirin 0-7 high mobility group box 1 Homo sapiens 164-169 29203538-3 2018 We previously showed that HMGB1 with reduced cysteines is a chemoattractant, whereas a disulfide bond makes it a proinflammatory cytokine. Cysteine 45-54 high mobility group box 1 Homo sapiens 26-31 29317708-5 2018 By screening a panel of Cx43 mimetic peptides, we discovered that one cysteine-containing peptide, P5 (ENVCYD), effectively attenuated hemichannel activities, and significantly suppressed endotoxin-induced release of ATP and HMGB1 in vitro. Cysteine 70-78 high mobility group box 1 Homo sapiens 225-230 29203538-4 2018 Here we report that fully reduced HMGB1 orchestrates muscle and liver regeneration via CXCR4, whereas disulfide HMGB1 and its receptors TLR4/MD-2 and RAGE (receptor for advanced glycation end products) are not involved. Disulfides 102-111 high mobility group box 1 Homo sapiens 112-117 29203538-6 2018 The nonoxidizable HMGB1 mutant 3S, in which serines replace cysteines, promotes muscle and liver regeneration more efficiently than the wild-type protein and without exacerbating inflammation by selectively interacting with CXCR4. Serine 44-51 high mobility group box 1 Homo sapiens 18-23 29203538-6 2018 The nonoxidizable HMGB1 mutant 3S, in which serines replace cysteines, promotes muscle and liver regeneration more efficiently than the wild-type protein and without exacerbating inflammation by selectively interacting with CXCR4. Cysteine 60-69 high mobility group box 1 Homo sapiens 18-23 30198432-0 2018 miR-142-3p Inhibits the Metastasis of Hepatocellular Carcinoma Cells by Regulating HMGB1 Gene Expression. mir-142-3p 0-10 high mobility group box 1 Homo sapiens 83-88 30205729-0 2018 HMGB1 represses the anti-cancer activity of sunitinib by governing TP53 autophagic degradation via its nucleus-to-cytoplasm transport. Sunitinib 44-53 high mobility group box 1 Homo sapiens 0-5 30205729-7 2018 The inhibition of HMGB1 sensitized cancer cells to sunitinib. Sunitinib 51-60 high mobility group box 1 Homo sapiens 18-23 30205729-9 2018 In conclusion, our data identify an alternative HMGB1-mediated TP53 protein turnover mechanism that participates in the resistance of sunitinib and suggest HMGB1 as a potential therapeutic target for improving clinical outcomes of sunitinib. Sunitinib 134-143 high mobility group box 1 Homo sapiens 48-53 30205729-9 2018 In conclusion, our data identify an alternative HMGB1-mediated TP53 protein turnover mechanism that participates in the resistance of sunitinib and suggest HMGB1 as a potential therapeutic target for improving clinical outcomes of sunitinib. Sunitinib 231-240 high mobility group box 1 Homo sapiens 48-53 30205729-9 2018 In conclusion, our data identify an alternative HMGB1-mediated TP53 protein turnover mechanism that participates in the resistance of sunitinib and suggest HMGB1 as a potential therapeutic target for improving clinical outcomes of sunitinib. Sunitinib 231-240 high mobility group box 1 Homo sapiens 156-161 29719300-0 2018 Unfractionated Heparin Alleviates Human Lung Endothelial Barrier Dysfunction Induced by High Mobility Group Box 1 Through Regulation of P38-GSK3beta-Snail Signaling Pathway. Heparin 15-22 high mobility group box 1 Homo sapiens 88-113 29719300-12 2018 Moreover, HMGB1 activated p38/GSK3beta/Snail signaling pathway and treatment with p38 inhibitor SB203580 abolished its biological effects. SB 203580 96-104 high mobility group box 1 Homo sapiens 10-15 29719300-13 2018 In addition, we found that UFH was able to reverse the effect of HMGB1 on EndoMT and endothelial permeability through inhibition of p38 signaling in a dose-dependent manner. Heparin 27-30 high mobility group box 1 Homo sapiens 65-70 29244233-5 2018 RESULTS: DAB caused neuronal pannexin-1 large pore opening and activation of the downstream inflammatory pathway as shown by procaspase-1 cleavage and HMGB1 release from neurons. 1,4-dideoxy-1,4-iminoarabinitol 9-12 high mobility group box 1 Homo sapiens 151-156 30497069-6 2018 HMGB1 activated p38 mitogen-activated protein kinase (p38 MAPK) and ADAM17, while HMGB1-induced ADAM17 activation was inhibited by SB203580, a p38 MAPK inhibitor. SB 203580 131-139 high mobility group box 1 Homo sapiens 82-87 30497069-7 2018 HMGB1-induced ectodomain shedding of RAGE and TLR4 was prevented by siRNA depletion of ADAM17 as well as TAPI-2, an inhibitor of ADAM family, and SB203580. TAPI-2 105-111 high mobility group box 1 Homo sapiens 0-5 30497069-7 2018 HMGB1-induced ectodomain shedding of RAGE and TLR4 was prevented by siRNA depletion of ADAM17 as well as TAPI-2, an inhibitor of ADAM family, and SB203580. SB 203580 146-154 high mobility group box 1 Homo sapiens 0-5 29138861-3 2018 In the present study, the expression of HMGB1 and calreticulin was evaluated by immunohistochemistry in pre-treatment biopsy specimens and surgically resected specimens, which were obtained from patients with breast cancer (n=52) and esophageal squamous cell carcinoma (ESCC) (n=8) who had been treated with neoadjuvant chemotherapy (NAC). nac 334-337 high mobility group box 1 Homo sapiens 40-45 28555325-0 2018 Erratum to: Serum HMGB1 concentrations at 4 weeks is a useful predictor of extreme poor prognosis for advanced hepatocellular carcinoma treated with sorafenib and hepatic arterial infusion chemotherapy. Sorafenib 149-158 high mobility group box 1 Homo sapiens 18-23 28474222-0 2018 Serum HMGB1 concentrations at 4 weeks is a useful predictor of extreme poor prognosis for advanced hepatocellular carcinoma treated with sorafenib and hepatic arterial infusion chemotherapy. Sorafenib 137-146 high mobility group box 1 Homo sapiens 6-11 28474222-4 2018 METHODS: Serum HMGB1 concentrations were measured in 71 and 72 patients with advanced HCC treated with sorafenib and HAIC, respectively, to assess their usefulness for prediction of the response to the treatment and prognosis. Sorafenib 103-112 high mobility group box 1 Homo sapiens 15-20 28474222-8 2018 CONCLUSIONS: These results suggest that serum HMGB1 at 4 weeks after the start of treatment might be a useful biomarker with added value to the conventional tumor marker and radiologic responses to predict poor overall survival in patients with advanced HCC treated with sorafenib or HAIC. Sorafenib 271-280 high mobility group box 1 Homo sapiens 46-51 29138861-5 2018 As a result, both HMGB1 and calreticulin expression levels were significantly upregulated after NAC in both breast cancer and ESCC tissues. nac 96-99 high mobility group box 1 Homo sapiens 18-23 29283384-4 2017 Treatment of the HMGB1 inhibitor glycyrrhizic acid (GA) showed an opposing effect on both activities. Glycyrrhizic Acid 33-50 high mobility group box 1 Homo sapiens 17-22 28986506-4 2018 Furthermore, methylation of arginines in the RGG domain abolishes the protein-protein interaction and the inhibitory effect of Sbp1 on translation initiation of Pab1 mRNA. Arginine 28-37 high mobility group box 1 Homo sapiens 127-131 32476912-8 2018 HMGB1 level varied in different diagnoses: the highest level was detected in QuantiFERON TB-positive subjects (median: 30.2) and hypersensitivity pneumonitis (median: 33.2), followed by pulmonary sarcoidosis (median: 16.8) and idiopathic pulmonary fibrosis (median: 8.8). quantiferon tb 77-91 high mobility group box 1 Homo sapiens 0-5 29283384-4 2017 Treatment of the HMGB1 inhibitor glycyrrhizic acid (GA) showed an opposing effect on both activities. Glycyrrhizic Acid 52-54 high mobility group box 1 Homo sapiens 17-22 29258482-4 2017 Thus, this study aimed to investigate the dose- and time-dependent effects of herbal medicines to protect against AKI in cisplatin-induced human kidney 2 (HK-2) cells by assessing the activities of high-mobility group box protein 1 (HMGB1), neutrophil gelatinase-associated lipocalin (NGAL) and kidney injury molecule-1 (KIM-1). Cisplatin 121-130 high mobility group box 1 Homo sapiens 233-238 29246127-0 2017 High mobility group box 1 promotes sorafenib resistance in HepG2 cells and in vivo. Sorafenib 35-44 high mobility group box 1 Homo sapiens 0-25 29246127-7 2017 This study investigated the association between HMGB1 and sorafenib resistance in HCC. Sorafenib 58-67 high mobility group box 1 Homo sapiens 48-53 29246127-10 2017 The subcellular localization of HMGB1 in HepG2 cells following sorafenib treatment was measured by western blotting and confocal microscopy. Sorafenib 63-72 high mobility group box 1 Homo sapiens 32-37 29246127-12 2017 RESULTS: The HMGB1 knockdown cells exhibited a significantly higher apoptotic level and lower cell viability than the normal HMGB1 expressing cells following the sorafenib treatment. Sorafenib 162-171 high mobility group box 1 Homo sapiens 13-18 29246127-12 2017 RESULTS: The HMGB1 knockdown cells exhibited a significantly higher apoptotic level and lower cell viability than the normal HMGB1 expressing cells following the sorafenib treatment. Sorafenib 162-171 high mobility group box 1 Homo sapiens 125-130 29246127-13 2017 In addition, the cell viability observed in the HMGB1 overexpressing cells was higher than that observed in the control cells following the sorafenib intervention. Sorafenib 140-149 high mobility group box 1 Homo sapiens 48-53 29246127-14 2017 Sorafenib had a better tumour inhibition effect in the HMGB1 knockdown group in vivo. Sorafenib 0-9 high mobility group box 1 Homo sapiens 55-60 29246127-15 2017 The amount of mitochondrial HMGB1 decreased, while the amount of cytosolic HMGB1 increased following the exposure to sorafenib. Sorafenib 117-126 high mobility group box 1 Homo sapiens 28-33 29246127-15 2017 The amount of mitochondrial HMGB1 decreased, while the amount of cytosolic HMGB1 increased following the exposure to sorafenib. Sorafenib 117-126 high mobility group box 1 Homo sapiens 75-80 29246127-16 2017 Altogether, HMGB1 translocated from the mitochondria to the cytoplasm outside the mitochondria following the exposure of HepG2 cells to sorafenib. Sorafenib 136-145 high mobility group box 1 Homo sapiens 12-17 29246127-17 2017 CONCLUSIONS: A novel potential role of HMGB1 in the regulation of sorafenib therapy resistance in HCC was observed. Sorafenib 66-75 high mobility group box 1 Homo sapiens 39-44 29246127-18 2017 The knockdown of HMGB1 restores sensitivity to sorafenib and enhances HepG2 cell death, while HMGB1 overexpression blunts these effects. Sorafenib 47-56 high mobility group box 1 Homo sapiens 17-22 29246127-19 2017 The translocation of HMGB1 from the mitochondria to the cytosol following sorafenib treatment provides new insight into sorafenib resistance in HCC. Sorafenib 74-83 high mobility group box 1 Homo sapiens 21-26 29246127-19 2017 The translocation of HMGB1 from the mitochondria to the cytosol following sorafenib treatment provides new insight into sorafenib resistance in HCC. Sorafenib 120-129 high mobility group box 1 Homo sapiens 21-26 29259392-7 2017 Significant positive correlations were observed between the levels of HMGB1 and the levels of 8-OHdG (r = 0.422; p = 0.001) and sVAP-1 (r = 0.354; p = 0.004) and between the levels of 8-OHdG and the levels of sVAP-1 (r = 0.598; p<0.0001). 8-ohdg 94-100 high mobility group box 1 Homo sapiens 70-75 29259392-7 2017 Significant positive correlations were observed between the levels of HMGB1 and the levels of 8-OHdG (r = 0.422; p = 0.001) and sVAP-1 (r = 0.354; p = 0.004) and between the levels of 8-OHdG and the levels of sVAP-1 (r = 0.598; p<0.0001). 8-ohdg 184-190 high mobility group box 1 Homo sapiens 70-75 29259392-13 2017 Treatment of HRMECs with HMGB1 increased leukocyte adhesion and induced the upregulation of 8-OHdG and HO-1 and the membranous translocation of VAP-1. 8-ohdg 92-98 high mobility group box 1 Homo sapiens 25-30 28970183-3 2017 A chemical hypoxia mimetic agent, cobalt chloride, induced SIRT1 downregulation, HMGB1 nucleocytoplasmic relocation and extracellular release from ARPE-19 cells, implicating its autocrine function. cobaltous chloride 34-49 high mobility group box 1 Homo sapiens 81-86 28970183-4 2017 Resveratrol treatment significantly reduced secretion of HMGB1 from ARPE-19 cells exposed to hypoxia. Resveratrol 0-11 high mobility group box 1 Homo sapiens 57-62 29285170-7 2017 The results of western blot analysis indicated that the expression levels of HMGB1, TGF-beta1 and NF-kappaB were significantly higher in the CAN group, compared with the normal group (P<0.05), and the expression levels increased with the progression of CAN grade. can 141-144 high mobility group box 1 Homo sapiens 77-82 29285170-11 2017 HMGB1, TGF-beta1 and NF-kappaB may form a signaling pathway that leads to the occurrence of CAN, which induces renal interstitial fibrosis. can 92-95 high mobility group box 1 Homo sapiens 0-5 29113279-6 2017 Meanwhile, administration of gemcitabine to HIPC cells increased the expression of high mobility group box1 (HMGB1). gemcitabine 29-40 high mobility group box 1 Homo sapiens 109-114 29181774-6 2017 Two of the most commonly used ABAs in pharmaceutical vaccine formulations, aluminium oxyhydroxide and aluminium hydroxyphosphate, induced a vigorous extracellular expression of the two DAMP molecules calreticulin and HMGB1. abas 30-34 high mobility group box 1 Homo sapiens 217-222 29181774-6 2017 Two of the most commonly used ABAs in pharmaceutical vaccine formulations, aluminium oxyhydroxide and aluminium hydroxyphosphate, induced a vigorous extracellular expression of the two DAMP molecules calreticulin and HMGB1. aluminium oxyhydroxide 75-97 high mobility group box 1 Homo sapiens 217-222 29181774-6 2017 Two of the most commonly used ABAs in pharmaceutical vaccine formulations, aluminium oxyhydroxide and aluminium hydroxyphosphate, induced a vigorous extracellular expression of the two DAMP molecules calreticulin and HMGB1. aluminium hydroxyphosphate 102-128 high mobility group box 1 Homo sapiens 217-222 29149067-0 2017 The HMGB1/RAGE Pro-Inflammatory Axis in the Human Placenta: Modulating Effect of Low Molecular Weight Heparin. Heparin 102-109 high mobility group box 1 Homo sapiens 4-9 29149067-2 2017 Moreover, we investigated, in physiological placental tissue, the ability of Low Molecular Weight Heparin (LMWH) to modify HMGB1 structural conformation thus inhibiting RAGE binding and HMGB1/RAGE axis inflammatory activity. Heparin 98-105 high mobility group box 1 Homo sapiens 123-128 29149067-2 2017 Moreover, we investigated, in physiological placental tissue, the ability of Low Molecular Weight Heparin (LMWH) to modify HMGB1 structural conformation thus inhibiting RAGE binding and HMGB1/RAGE axis inflammatory activity. Heparin 98-105 high mobility group box 1 Homo sapiens 186-191 29149067-2 2017 Moreover, we investigated, in physiological placental tissue, the ability of Low Molecular Weight Heparin (LMWH) to modify HMGB1 structural conformation thus inhibiting RAGE binding and HMGB1/RAGE axis inflammatory activity. Heparin, Low-Molecular-Weight 107-111 high mobility group box 1 Homo sapiens 123-128 29149067-2 2017 Moreover, we investigated, in physiological placental tissue, the ability of Low Molecular Weight Heparin (LMWH) to modify HMGB1 structural conformation thus inhibiting RAGE binding and HMGB1/RAGE axis inflammatory activity. Heparin, Low-Molecular-Weight 107-111 high mobility group box 1 Homo sapiens 186-191 29149067-5 2017 Physiological term placental explants were treated by 0.5 U LMWH for 24 or 48 h. HMGB1 and RAGE expression and association were evaluated in LMWH explants by RAGE immunoprecipitation followed by HMGB1 immunoblot. Heparin, Low-Molecular-Weight 141-145 high mobility group box 1 Homo sapiens 81-86 29149067-8 2017 24 h LMWH treatment significantly up-regulated HMGB1 expression but inhibited HMGB1/RAGE complex formation in physiological explants. Heparin, Low-Molecular-Weight 5-9 high mobility group box 1 Homo sapiens 47-52 29149067-8 2017 24 h LMWH treatment significantly up-regulated HMGB1 expression but inhibited HMGB1/RAGE complex formation in physiological explants. Heparin, Low-Molecular-Weight 5-9 high mobility group box 1 Homo sapiens 78-83 29149067-11 2017 Our data depicted a new molecular mechanism through which LMWH exerts its anti-inflammatory effect on PE placentae, underlying the importance of HMGB1/RAGE axis in PE inflammatory response. Heparin, Low-Molecular-Weight 58-62 high mobility group box 1 Homo sapiens 145-150 29296182-0 2017 MicroRNA-410-3p attenuates gemcitabine resistance in pancreatic ductal adenocarcinoma by inhibiting HMGB1-mediated autophagy. gemcitabine 27-38 high mobility group box 1 Homo sapiens 100-105 29296182-6 2017 We also found that miR-410-3p attenuated the gemcitabine resistance of PDAC by targeting the 3"-UTR of HMGB1. gemcitabine 45-56 high mobility group box 1 Homo sapiens 103-108 29296182-6 2017 We also found that miR-410-3p attenuated the gemcitabine resistance of PDAC by targeting the 3"-UTR of HMGB1. pdac 71-75 high mobility group box 1 Homo sapiens 103-108 29296182-7 2017 Moreover, our study clearly demonstrated that miR-410-3p enhanced chemosensitivity to gemcitabine via inhibiting HMGB1-induced autophagy during chemotherapy in PDAC cells. gemcitabine 86-97 high mobility group box 1 Homo sapiens 113-118 29201182-0 2017 The efficacy of gastrodin in combination with folate and vitamin B12 on patients with epilepsy after stroke and its effect on HMGB-1, IL-2 and IL-6 serum levels. gastrodin 16-25 high mobility group box 1 Homo sapiens 126-132 29201182-1 2017 This study evaluated the efficacy of gastrodin in combination with folate (FOL) and vitamin-B12 (V-B12) on patients with epilepsy after stroke (EAS) and its effect on high-mobility group protein B1 (HMGB-1), interleukin-1 (IL-1), and IL-6 serum levels. gastrodin 37-46 high mobility group box 1 Homo sapiens 167-197 29201182-1 2017 This study evaluated the efficacy of gastrodin in combination with folate (FOL) and vitamin-B12 (V-B12) on patients with epilepsy after stroke (EAS) and its effect on high-mobility group protein B1 (HMGB-1), interleukin-1 (IL-1), and IL-6 serum levels. gastrodin 37-46 high mobility group box 1 Homo sapiens 199-205 29113279-0 2017 HMGB1-mediated autophagy confers resistance to gemcitabine in hormone-independent prostate cancer cells. gemcitabine 47-58 high mobility group box 1 Homo sapiens 0-5 29113279-6 2017 Meanwhile, administration of gemcitabine to HIPC cells increased the expression of high mobility group box1 (HMGB1). gemcitabine 29-40 high mobility group box 1 Homo sapiens 83-107 29113279-7 2017 Furthermore, the gemcitabine-induced autophagy response was attenuated in stable HIPC cells harboring HMGB1 shRNA. gemcitabine 17-28 high mobility group box 1 Homo sapiens 102-107 29113279-8 2017 Notably, the HIPC cells stably transfected with HMGB1 shRNA or treated with autophagy inhibitors were more sensitive to gemcitabine compared with the control group. gemcitabine 120-131 high mobility group box 1 Homo sapiens 48-53 29113279-9 2017 These data suggested that inhibition of HMGB1 increased the sensitivity to gemcitabine by decreasing autophagy response in HIPC cells. gemcitabine 75-86 high mobility group box 1 Homo sapiens 40-45 29085368-0 2017 1,25-Dihydroxyvitamin D Inhibits LPS-Induced High-Mobility Group Box 1 (HMGB1) Secretion via Targeting the NF-E2-Related Factor 2-Hemeoxygenase-1-HMGB1 Pathway in Macrophages. 1,25-dihydroxyvitamin D 0-23 high mobility group box 1 Homo sapiens 45-70 29085368-0 2017 1,25-Dihydroxyvitamin D Inhibits LPS-Induced High-Mobility Group Box 1 (HMGB1) Secretion via Targeting the NF-E2-Related Factor 2-Hemeoxygenase-1-HMGB1 Pathway in Macrophages. 1,25-dihydroxyvitamin D 0-23 high mobility group box 1 Homo sapiens 72-77 29085368-0 2017 1,25-Dihydroxyvitamin D Inhibits LPS-Induced High-Mobility Group Box 1 (HMGB1) Secretion via Targeting the NF-E2-Related Factor 2-Hemeoxygenase-1-HMGB1 Pathway in Macrophages. 1,25-dihydroxyvitamin D 0-23 high mobility group box 1 Homo sapiens 146-151 28975966-2 2017 MATERIALS AND METHODS: The effect of HMGB1 on the proliferation of the two cell types was examined using the MTT assay under environmental hypoxia (incubation with 1.5% oxygen) to simulate the condition of pulmonary arterial hypertension in the body. monooxyethylene trimethylolpropane tristearate 109-112 high mobility group box 1 Homo sapiens 37-42 29085368-6 2017 When siHO-1 or an HO-1 inhibitor are used, the effect of 1,25(OH)2D3 on inhibition of HMGB1 secretion is suppressed. Calcitriol 57-68 high mobility group box 1 Homo sapiens 86-91 29018504-2 2017 We also report data on the use of 18-beta-glycyrrhetic acid (GA), which has been shown able to inhibit the pro-inflammatory activities of HMGB1, in young patients affected by allergic rhinitis and complaining of nasal obstruction as main symptom. Glycyrrhetinic Acid 34-59 high mobility group box 1 Homo sapiens 138-143 29018504-2 2017 We also report data on the use of 18-beta-glycyrrhetic acid (GA), which has been shown able to inhibit the pro-inflammatory activities of HMGB1, in young patients affected by allergic rhinitis and complaining of nasal obstruction as main symptom. Glycyrrhetinic Acid 61-63 high mobility group box 1 Homo sapiens 138-143 29018504-8 2017 GA inhibits HMGB1 chemotactic and mitogenic function by a scavenger mechanism on extracellular HMGB1 accumulation stimulated by lipopolysaccharides in vitro. Glycyrrhetinic Acid 0-2 high mobility group box 1 Homo sapiens 12-17 29018504-8 2017 GA inhibits HMGB1 chemotactic and mitogenic function by a scavenger mechanism on extracellular HMGB1 accumulation stimulated by lipopolysaccharides in vitro. Glycyrrhetinic Acid 0-2 high mobility group box 1 Homo sapiens 95-100 27501713-7 2017 The biphasic biological property of extracellular HMGB1 may be related to the redox modifications of its cysteine residues. Cysteine 105-113 high mobility group box 1 Homo sapiens 50-55 28474284-5 2017 This review focuses on the interactions and contribution to the pathogenesis and progression of OA through the DAMPs: high-mobility group box 1 (HMGB-1), the receptor for advanced glycation end-products (RAGE), the alarmin proteins S100A8 and S100A9, and heparan sulfate. Heparitin Sulfate 255-270 high mobility group box 1 Homo sapiens 145-151 28474284-9 2017 Heparan sulfate has been shown to facilitate the binding of HMGB-1 to RAGE and could play a role in the progression of OA. Heparitin Sulfate 0-15 high mobility group box 1 Homo sapiens 60-66 28741799-5 2017 RESULT: Baseline HMGB-1 was significantly higher in the BHT (+) group than in the BHT (-) group. Butylated Hydroxytoluene 56-59 high mobility group box 1 Homo sapiens 17-23 28741799-5 2017 RESULT: Baseline HMGB-1 was significantly higher in the BHT (+) group than in the BHT (-) group. Butylated Hydroxytoluene 82-85 high mobility group box 1 Homo sapiens 17-23 28741799-6 2017 Thereafter, HMGB-1 in the BHT (+) group significantly decreased at 24 h intervals, reaching the reference range by 2 days of age. Butylated Hydroxytoluene 26-29 high mobility group box 1 Homo sapiens 12-18 28741799-7 2017 In the BHT (+) group, when patients were classified into clinically significant neurological disorder due to HIE (+) and (-) according to MRI, the neurological disorder (+) group had higher mean HMGB-1. Butylated Hydroxytoluene 7-10 high mobility group box 1 Homo sapiens 195-201 28741799-8 2017 CONCLUSIONS: In HIE, HMGB-1 differs according to the presence of BHT, suggesting that HMGB-1 measurement soon after birth might be useful for determining BHT necessity and short-term outcome. Butylated Hydroxytoluene 65-68 high mobility group box 1 Homo sapiens 21-27 28741799-8 2017 CONCLUSIONS: In HIE, HMGB-1 differs according to the presence of BHT, suggesting that HMGB-1 measurement soon after birth might be useful for determining BHT necessity and short-term outcome. Butylated Hydroxytoluene 65-68 high mobility group box 1 Homo sapiens 86-92 28741799-8 2017 CONCLUSIONS: In HIE, HMGB-1 differs according to the presence of BHT, suggesting that HMGB-1 measurement soon after birth might be useful for determining BHT necessity and short-term outcome. Butylated Hydroxytoluene 154-157 high mobility group box 1 Homo sapiens 21-27 28741799-8 2017 CONCLUSIONS: In HIE, HMGB-1 differs according to the presence of BHT, suggesting that HMGB-1 measurement soon after birth might be useful for determining BHT necessity and short-term outcome. Butylated Hydroxytoluene 154-157 high mobility group box 1 Homo sapiens 86-92 29033853-0 2017 Proinflammatory Effect of High Glucose Concentrations on HMrSV5 Cells via the Autocrine Effect of HMGB1. Glucose 31-38 high mobility group box 1 Homo sapiens 98-103 29033853-5 2017 Aim: In this study, we aimed to explore the effect and underlying mechanism of endogenous HMGB1 in high-glucose-induced MC injury. Glucose 104-111 high mobility group box 1 Homo sapiens 90-95 29033853-9 2017 Results: The major findings of these analyses were: (1) MCs secreted HMGB1 from the nucleus during exposure to high glucose levels; HMGB1 acted in an autocrine fashion on the MCs to promote the production of MCP-1 and IL-8; (2) HMGB1 had little effect on high-glucose-induced apoptosis of the MCs; and (3) HMGB1-mediated MCP-1 and IL-8 production depended on the activation of MAPK signaling pathways. mcs 56-59 high mobility group box 1 Homo sapiens 69-74 29033853-9 2017 Results: The major findings of these analyses were: (1) MCs secreted HMGB1 from the nucleus during exposure to high glucose levels; HMGB1 acted in an autocrine fashion on the MCs to promote the production of MCP-1 and IL-8; (2) HMGB1 had little effect on high-glucose-induced apoptosis of the MCs; and (3) HMGB1-mediated MCP-1 and IL-8 production depended on the activation of MAPK signaling pathways. Glucose 116-123 high mobility group box 1 Homo sapiens 69-74 28692534-7 2017 NDE from neuropsychologically impaired study participants had significantly higher levels of high-mobility group box 1 (HMGB1), NF-L, and amyloid beta proteins compared with neuropsychologically normal individuals. NDE 0-3 high mobility group box 1 Homo sapiens 93-118 28692534-7 2017 NDE from neuropsychologically impaired study participants had significantly higher levels of high-mobility group box 1 (HMGB1), NF-L, and amyloid beta proteins compared with neuropsychologically normal individuals. NDE 0-3 high mobility group box 1 Homo sapiens 120-125 28741799-1 2017 BACKGROUND: We measured changes in the blood level of high-mobility group box-1 (HMGB-1) at 24 h intervals in neonates treated with brain/body hypothermia (body hypothermia therapy: BHT) for hypoxic-ischemic encephalopathy (HIE), to evaluate the usefulness of HMGB-1 level for determining outcomes. Butylated Hydroxytoluene 182-185 high mobility group box 1 Homo sapiens 54-79 28741799-1 2017 BACKGROUND: We measured changes in the blood level of high-mobility group box-1 (HMGB-1) at 24 h intervals in neonates treated with brain/body hypothermia (body hypothermia therapy: BHT) for hypoxic-ischemic encephalopathy (HIE), to evaluate the usefulness of HMGB-1 level for determining outcomes. Butylated Hydroxytoluene 182-185 high mobility group box 1 Homo sapiens 81-87 29033853-9 2017 Results: The major findings of these analyses were: (1) MCs secreted HMGB1 from the nucleus during exposure to high glucose levels; HMGB1 acted in an autocrine fashion on the MCs to promote the production of MCP-1 and IL-8; (2) HMGB1 had little effect on high-glucose-induced apoptosis of the MCs; and (3) HMGB1-mediated MCP-1 and IL-8 production depended on the activation of MAPK signaling pathways. mcs 175-178 high mobility group box 1 Homo sapiens 132-137 29033853-9 2017 Results: The major findings of these analyses were: (1) MCs secreted HMGB1 from the nucleus during exposure to high glucose levels; HMGB1 acted in an autocrine fashion on the MCs to promote the production of MCP-1 and IL-8; (2) HMGB1 had little effect on high-glucose-induced apoptosis of the MCs; and (3) HMGB1-mediated MCP-1 and IL-8 production depended on the activation of MAPK signaling pathways. mcs 175-178 high mobility group box 1 Homo sapiens 132-137 29033853-9 2017 Results: The major findings of these analyses were: (1) MCs secreted HMGB1 from the nucleus during exposure to high glucose levels; HMGB1 acted in an autocrine fashion on the MCs to promote the production of MCP-1 and IL-8; (2) HMGB1 had little effect on high-glucose-induced apoptosis of the MCs; and (3) HMGB1-mediated MCP-1 and IL-8 production depended on the activation of MAPK signaling pathways. mcs 175-178 high mobility group box 1 Homo sapiens 132-137 29033853-9 2017 Results: The major findings of these analyses were: (1) MCs secreted HMGB1 from the nucleus during exposure to high glucose levels; HMGB1 acted in an autocrine fashion on the MCs to promote the production of MCP-1 and IL-8; (2) HMGB1 had little effect on high-glucose-induced apoptosis of the MCs; and (3) HMGB1-mediated MCP-1 and IL-8 production depended on the activation of MAPK signaling pathways. Glucose 260-267 high mobility group box 1 Homo sapiens 132-137 29033853-9 2017 Results: The major findings of these analyses were: (1) MCs secreted HMGB1 from the nucleus during exposure to high glucose levels; HMGB1 acted in an autocrine fashion on the MCs to promote the production of MCP-1 and IL-8; (2) HMGB1 had little effect on high-glucose-induced apoptosis of the MCs; and (3) HMGB1-mediated MCP-1 and IL-8 production depended on the activation of MAPK signaling pathways. Glucose 260-267 high mobility group box 1 Homo sapiens 132-137 29033853-9 2017 Results: The major findings of these analyses were: (1) MCs secreted HMGB1 from the nucleus during exposure to high glucose levels; HMGB1 acted in an autocrine fashion on the MCs to promote the production of MCP-1 and IL-8; (2) HMGB1 had little effect on high-glucose-induced apoptosis of the MCs; and (3) HMGB1-mediated MCP-1 and IL-8 production depended on the activation of MAPK signaling pathways. Glucose 260-267 high mobility group box 1 Homo sapiens 132-137 29033853-9 2017 Results: The major findings of these analyses were: (1) MCs secreted HMGB1 from the nucleus during exposure to high glucose levels; HMGB1 acted in an autocrine fashion on the MCs to promote the production of MCP-1 and IL-8; (2) HMGB1 had little effect on high-glucose-induced apoptosis of the MCs; and (3) HMGB1-mediated MCP-1 and IL-8 production depended on the activation of MAPK signaling pathways. mcs 175-178 high mobility group box 1 Homo sapiens 132-137 29033853-9 2017 Results: The major findings of these analyses were: (1) MCs secreted HMGB1 from the nucleus during exposure to high glucose levels; HMGB1 acted in an autocrine fashion on the MCs to promote the production of MCP-1 and IL-8; (2) HMGB1 had little effect on high-glucose-induced apoptosis of the MCs; and (3) HMGB1-mediated MCP-1 and IL-8 production depended on the activation of MAPK signaling pathways. mcs 175-178 high mobility group box 1 Homo sapiens 132-137 29033853-9 2017 Results: The major findings of these analyses were: (1) MCs secreted HMGB1 from the nucleus during exposure to high glucose levels; HMGB1 acted in an autocrine fashion on the MCs to promote the production of MCP-1 and IL-8; (2) HMGB1 had little effect on high-glucose-induced apoptosis of the MCs; and (3) HMGB1-mediated MCP-1 and IL-8 production depended on the activation of MAPK signaling pathways. mcs 175-178 high mobility group box 1 Homo sapiens 132-137 29033853-10 2017 In conclusion, endogenous HMGB1 plays an important role in the inflammatory reaction induced by high glucose on MCs via mitogen-activated protein kinase (MAPK) signaling pathways, but it seems to have little effect on high-glucose-induced apoptosis. Glucose 101-108 high mobility group box 1 Homo sapiens 26-31 29033853-10 2017 In conclusion, endogenous HMGB1 plays an important role in the inflammatory reaction induced by high glucose on MCs via mitogen-activated protein kinase (MAPK) signaling pathways, but it seems to have little effect on high-glucose-induced apoptosis. mcs 112-115 high mobility group box 1 Homo sapiens 26-31 28514198-6 2017 The current study further indicated that piceatannol decreased the expression of sbp-1 (encodes an ortholog of mammalian sterol regulatory element-binding protein) and its target gene fasn-1 (encodes an ortholog of fatty acid synthase) as well as increased the expression of hosl-1 (encodes an ortholog of hormone-sensitive lipase) in glucose-treated worms. 3,3',4,5'-tetrahydroxystilbene 41-52 high mobility group box 1 Homo sapiens 81-86 28185215-10 2017 Importantly, SA up-regulated a series of apoptosis-related gene expression and reduced the mRNA level of HMGB1. Salicylic Acid 13-15 high mobility group box 1 Homo sapiens 105-110 28514198-6 2017 The current study further indicated that piceatannol decreased the expression of sbp-1 (encodes an ortholog of mammalian sterol regulatory element-binding protein) and its target gene fasn-1 (encodes an ortholog of fatty acid synthase) as well as increased the expression of hosl-1 (encodes an ortholog of hormone-sensitive lipase) in glucose-treated worms. Glucose 335-342 high mobility group box 1 Homo sapiens 81-86 28161237-5 2017 Here we report that LMWHs induce a profound change in the ability of human neutrophils to generate NETs and to mobilize the content of the primary granules in response to unrelated inflammatory stimuli, such as IL-8, PMA and HMGB1. Heparin, Low-Molecular-Weight 20-25 high mobility group box 1 Homo sapiens 225-230 28858636-9 2017 OS and senescence caused the translocation of HMGB1 and cffTF from AECs" nuclei to cytoplasm compared to untreated cells, which was inhibited by antioxidant N-acetyl cysteine (NAC). Acetylcysteine 157-174 high mobility group box 1 Homo sapiens 46-51 28858636-9 2017 OS and senescence caused the translocation of HMGB1 and cffTF from AECs" nuclei to cytoplasm compared to untreated cells, which was inhibited by antioxidant N-acetyl cysteine (NAC). Acetylcysteine 176-179 high mobility group box 1 Homo sapiens 46-51 28677786-8 2017 Furthermore, LPS-stimulation significantly upregulated HMGB-1 secretion via the c-Jun N-terminal kinase (JNK) signaling pathway in RAW264.7 macrophages, whereas pretreatment with the JNK inhibitor SP600125 markedly downregulated LPS-induced HMGB-1 levels. pyrazolanthrone 197-205 high mobility group box 1 Homo sapiens 55-61 29798353-3 2017 The effect of HMGB1 on fluorescein isothiocyanatedextran 4 kDa (FD4) permeability of NECs was measured. fluorescein isothiocyanatedextran 4 kda 23-62 high mobility group box 1 Homo sapiens 14-19 28912678-6 2017 These findings demonstrated that ROS might be located upstream of IL-17A and HMGB1 so that ROS can regulate HMGB1/IL-17A expression to affect the p53 and PI3K/Akt signaling pathways and therefore promote the occurrence of apoptosis in microglial cells. ros 33-36 high mobility group box 1 Homo sapiens 77-82 28912678-6 2017 These findings demonstrated that ROS might be located upstream of IL-17A and HMGB1 so that ROS can regulate HMGB1/IL-17A expression to affect the p53 and PI3K/Akt signaling pathways and therefore promote the occurrence of apoptosis in microglial cells. ros 33-36 high mobility group box 1 Homo sapiens 108-113 28912678-6 2017 These findings demonstrated that ROS might be located upstream of IL-17A and HMGB1 so that ROS can regulate HMGB1/IL-17A expression to affect the p53 and PI3K/Akt signaling pathways and therefore promote the occurrence of apoptosis in microglial cells. ros 91-94 high mobility group box 1 Homo sapiens 77-82 28912678-6 2017 These findings demonstrated that ROS might be located upstream of IL-17A and HMGB1 so that ROS can regulate HMGB1/IL-17A expression to affect the p53 and PI3K/Akt signaling pathways and therefore promote the occurrence of apoptosis in microglial cells. ros 91-94 high mobility group box 1 Homo sapiens 108-113 28732809-8 2017 RESULTS: Oleacein at the concentrations of 10 and 20 microM significantly (P < 0.001) decreased secretion of HMGB1 (up 90%), MMP-9 (up to 80%), MMP-9/NGAL complex (up to 80%) and TF (more than 90%) from the treated plaque, as compared to control. oleacein 9-17 high mobility group box 1 Homo sapiens 112-117 28677729-13 2017 In-depth IPA analysis revealed that CENPU was associated with the HMGB1 signaling pathway. ipa 9-12 high mobility group box 1 Homo sapiens 66-71 28167116-3 2017 anti-HMGB1 mAb attenuated both acute seizure models (maximal electroshock seizure, pentylenetetrazole-induced and kindling-induced), and chronic epilepsy model (kainic acid-induced) in a dose-dependent manner. Pentylenetetrazole 83-101 high mobility group box 1 Homo sapiens 5-10 28167116-3 2017 anti-HMGB1 mAb attenuated both acute seizure models (maximal electroshock seizure, pentylenetetrazole-induced and kindling-induced), and chronic epilepsy model (kainic acid-induced) in a dose-dependent manner. Kainic Acid 161-172 high mobility group box 1 Homo sapiens 5-10 28912641-6 2017 Moreover, we also discuss our recent findings that its endogenous ligand, high-mobility group box 1 (HMGB1), is released from dying OLs and exerts autocrine trophic effects on OLs and myelin sheath under ischemic condition. N-biotin-C-Co4(mu3-O)4(Py)4(H2O)4-beta-alanine 132-135 high mobility group box 1 Homo sapiens 74-99 28912641-6 2017 Moreover, we also discuss our recent findings that its endogenous ligand, high-mobility group box 1 (HMGB1), is released from dying OLs and exerts autocrine trophic effects on OLs and myelin sheath under ischemic condition. N-biotin-C-Co4(mu3-O)4(Py)4(H2O)4-beta-alanine 132-135 high mobility group box 1 Homo sapiens 101-106 28912641-6 2017 Moreover, we also discuss our recent findings that its endogenous ligand, high-mobility group box 1 (HMGB1), is released from dying OLs and exerts autocrine trophic effects on OLs and myelin sheath under ischemic condition. N-biotin-C-Co4(mu3-O)4(Py)4(H2O)4-beta-alanine 176-179 high mobility group box 1 Homo sapiens 74-99 28912641-6 2017 Moreover, we also discuss our recent findings that its endogenous ligand, high-mobility group box 1 (HMGB1), is released from dying OLs and exerts autocrine trophic effects on OLs and myelin sheath under ischemic condition. N-biotin-C-Co4(mu3-O)4(Py)4(H2O)4-beta-alanine 176-179 high mobility group box 1 Homo sapiens 101-106 28652214-4 2017 Serum concentrations of vWF and HMGB1 were significantly higher in all the patients than in the healthy controls, both prior to doxycycline treatment and on day 7 of doxycycline treatment (p<0.01). Doxycycline 166-177 high mobility group box 1 Homo sapiens 32-37 28159648-2 2017 Microglia, the innate immune cells of the brain, and neurons respond to alcohol, signaling through Toll-like receptors (TLRs), high-mobility group box 1 (HMGB1), miRNAs, pro-inflammatory cytokines and their associated receptors involved in signaling between microglia, other glia and neurons. Alcohols 72-79 high mobility group box 1 Homo sapiens 127-152 28159648-2 2017 Microglia, the innate immune cells of the brain, and neurons respond to alcohol, signaling through Toll-like receptors (TLRs), high-mobility group box 1 (HMGB1), miRNAs, pro-inflammatory cytokines and their associated receptors involved in signaling between microglia, other glia and neurons. Alcohols 72-79 high mobility group box 1 Homo sapiens 154-159 28159648-3 2017 Repeated cycles of alcohol and stress cause a progressive, persistent induction of HMGB1, miRNA and TLR receptors in brain that appear to underlie the progressive and persistent loss of behavioral control, increased impulsivity and anxiety, as well as craving, coupled with increasing ventral striatal responses that promote reward seeking behavior and increase risk of developing alcohol use disorders. Alcohols 19-26 high mobility group box 1 Homo sapiens 83-88 28159648-3 2017 Repeated cycles of alcohol and stress cause a progressive, persistent induction of HMGB1, miRNA and TLR receptors in brain that appear to underlie the progressive and persistent loss of behavioral control, increased impulsivity and anxiety, as well as craving, coupled with increasing ventral striatal responses that promote reward seeking behavior and increase risk of developing alcohol use disorders. Alcohols 381-388 high mobility group box 1 Homo sapiens 83-88 28575153-11 2017 Oxidative stress during epileptogenesis was associated with de novo brain and blood generation of disulfide high mobility group box 1 (HMGB1), a neuroinflammatory molecule implicated in seizure mechanisms. Disulfides 98-107 high mobility group box 1 Homo sapiens 108-133 29089150-0 2017 Hemin Reduces HMGB1 Release by UVB in an AMPK/HO-1-dependent Pathway in Human Keratinocytes HaCaT Cells. Hemin 0-5 high mobility group box 1 Homo sapiens 14-19 29089150-2 2017 We hypothesized that hemin might reduce HMGB1 release through the induction of HO-1 in UVB-induced HaCaTs. Hemin 21-26 high mobility group box 1 Homo sapiens 40-45 29089150-3 2017 METHODS: The effects of hemin on the release of HMGB1 in UVB exposure were evaluated. Hemin 24-29 high mobility group box 1 Homo sapiens 48-53 29089150-6 2017 HMGB1 release by UVB was significantly reduced by hemin, N-acetyl-cysteine and DPI (NADPH oxidase inhibitor). Hemin 50-55 high mobility group box 1 Homo sapiens 0-5 29089150-6 2017 HMGB1 release by UVB was significantly reduced by hemin, N-acetyl-cysteine and DPI (NADPH oxidase inhibitor). Acetylcysteine 57-74 high mobility group box 1 Homo sapiens 0-5 29089150-6 2017 HMGB1 release by UVB was significantly reduced by hemin, N-acetyl-cysteine and DPI (NADPH oxidase inhibitor). 3-aminodiphenyleneiodium 79-82 high mobility group box 1 Homo sapiens 0-5 29089150-9 2017 The inhibitory effects of UVB-induced HMGB1 release by hemin were significantly reversed not only with pharmacological inhibitors of AMPK (compound c) or HO-1 (ZnPPIX) but also through transfection of small interfering RNAs (siRNAs) for AMPK or HO-1. zinc protoporphyrin 160-166 high mobility group box 1 Homo sapiens 38-43 29089150-12 2017 CONCLUSIONS: It is concluded that the increased activity of HO-1/AMPK and scavenging ROS are, at least in part, responsible for the inhibition of UVB-induced HMGB1 release in keratinocyte HaCaTs. Reactive Oxygen Species 85-88 high mobility group box 1 Homo sapiens 158-163 28575153-11 2017 Oxidative stress during epileptogenesis was associated with de novo brain and blood generation of disulfide high mobility group box 1 (HMGB1), a neuroinflammatory molecule implicated in seizure mechanisms. Disulfides 98-107 high mobility group box 1 Homo sapiens 135-140 28575153-12 2017 Drug-induced reduction of oxidative stress prevented disulfide HMGB1 generation, thus highlighting a potential novel mechanism contributing to therapeutic effects. Disulfides 53-62 high mobility group box 1 Homo sapiens 63-68 28216372-3 2017 HBx triggered an increase of cytoplasmic calcium and activated CAMKK/CAMKIV pathway, leading to subsequent translocation and release of HMGB1. Calcium 41-48 high mobility group box 1 Homo sapiens 136-141 28294475-0 2017 Sodium tanshinone IIA sulfonate prevents hypoxic trophoblast-induced endothelial cell dysfunction via targeting HMGB1 release. tanshinone II A sodium sulfonate 0-31 high mobility group box 1 Homo sapiens 112-117 28294475-3 2017 In the present study, we investigated the protective effect of sodium tanshinone IIA sulfonate (STS), the soluble form of tanshinone IIA isolated from danshen, against hypoxic trophoblast HMGB1-induced human umbilical vein endothelial cell (HUVEC) dysfunction. tanshinone II A sodium sulfonate 63-94 high mobility group box 1 Homo sapiens 188-193 28294475-3 2017 In the present study, we investigated the protective effect of sodium tanshinone IIA sulfonate (STS), the soluble form of tanshinone IIA isolated from danshen, against hypoxic trophoblast HMGB1-induced human umbilical vein endothelial cell (HUVEC) dysfunction. tanshinone II A sodium sulfonate 96-99 high mobility group box 1 Homo sapiens 188-193 28294475-6 2017 In conclusion, our study suggests that STS is an effective agent against hypoxic trophoblast-induced cell injury of HUVEC via targeting HMGB1 release and forms the basis of the development of such a compound in treating PE. tanshinone II A sodium sulfonate 39-42 high mobility group box 1 Homo sapiens 136-141 28504645-6 2017 The pathologic disulfide HMGB1 isoform progressively increased in blood before epilepsy onset and prospectively identified animals that developed the disease. Disulfides 15-24 high mobility group box 1 Homo sapiens 25-30 28504645-7 2017 Consistent with animal data, we observed early expression of disulfide HMGB1 in patients with newly diagnosed epilepsy, and its persistence was associated with subsequent seizures. Disulfides 61-70 high mobility group box 1 Homo sapiens 71-76 28504645-8 2017 In contrast with patients with well-controlled epilepsy, patients with chronic, drug-refractory epilepsy persistently expressed the acetylated, disulfide HMGB1 isoforms. Disulfides 144-153 high mobility group box 1 Homo sapiens 154-159 28251435-7 2017 Moreover, HMGB1-mediated EPC apoptosis and CHOP expression were dramatically suppressed by PERK shRNA or a specific eIF2alpha inhibitor (salubrinal). salubrinal 137-147 high mobility group box 1 Homo sapiens 10-15 28444390-6 2017 High-mobility group box 1 protein (HMGB1) and lactate dehydrogenase release as well as adenosine triphosphate depletion occurred in a drug-, time-, and concentration-dependent manner with SMX-NO and flucloxacillin, whereas isoniazid and amoxicillin were nontoxic. 4-nitrososulfamethoxazole 188-194 high mobility group box 1 Homo sapiens 0-25 28444390-6 2017 High-mobility group box 1 protein (HMGB1) and lactate dehydrogenase release as well as adenosine triphosphate depletion occurred in a drug-, time-, and concentration-dependent manner with SMX-NO and flucloxacillin, whereas isoniazid and amoxicillin were nontoxic. 4-nitrososulfamethoxazole 188-194 high mobility group box 1 Homo sapiens 35-40 28444390-6 2017 High-mobility group box 1 protein (HMGB1) and lactate dehydrogenase release as well as adenosine triphosphate depletion occurred in a drug-, time-, and concentration-dependent manner with SMX-NO and flucloxacillin, whereas isoniazid and amoxicillin were nontoxic. Floxacillin 199-213 high mobility group box 1 Homo sapiens 0-25 28444390-6 2017 High-mobility group box 1 protein (HMGB1) and lactate dehydrogenase release as well as adenosine triphosphate depletion occurred in a drug-, time-, and concentration-dependent manner with SMX-NO and flucloxacillin, whereas isoniazid and amoxicillin were nontoxic. Floxacillin 199-213 high mobility group box 1 Homo sapiens 35-40 28444390-6 2017 High-mobility group box 1 protein (HMGB1) and lactate dehydrogenase release as well as adenosine triphosphate depletion occurred in a drug-, time-, and concentration-dependent manner with SMX-NO and flucloxacillin, whereas isoniazid and amoxicillin were nontoxic. Isoniazid 223-232 high mobility group box 1 Homo sapiens 0-25 28444390-6 2017 High-mobility group box 1 protein (HMGB1) and lactate dehydrogenase release as well as adenosine triphosphate depletion occurred in a drug-, time-, and concentration-dependent manner with SMX-NO and flucloxacillin, whereas isoniazid and amoxicillin were nontoxic. Isoniazid 223-232 high mobility group box 1 Homo sapiens 35-40 28444390-6 2017 High-mobility group box 1 protein (HMGB1) and lactate dehydrogenase release as well as adenosine triphosphate depletion occurred in a drug-, time-, and concentration-dependent manner with SMX-NO and flucloxacillin, whereas isoniazid and amoxicillin were nontoxic. Amoxicillin 237-248 high mobility group box 1 Homo sapiens 0-25 28444390-6 2017 High-mobility group box 1 protein (HMGB1) and lactate dehydrogenase release as well as adenosine triphosphate depletion occurred in a drug-, time-, and concentration-dependent manner with SMX-NO and flucloxacillin, whereas isoniazid and amoxicillin were nontoxic. Amoxicillin 237-248 high mobility group box 1 Homo sapiens 35-40 28444390-8 2017 The disulfide isoform of HMGB1 stimulated dendritic cell cytokine release and enhanced the priming of naive T-cells. Disulfides 4-13 high mobility group box 1 Homo sapiens 25-30 28584116-7 2017 Knockdown of HMGB1 or application of glycyrrhizin, a specific HMGB1 inhibitor, aggravated OGD-induced OL death, and recombinant HMGB1 application reduced the extent of OL death in a TLR2-dependent manner. Glycyrrhizic Acid 37-49 high mobility group box 1 Homo sapiens 62-67 28584116-7 2017 Knockdown of HMGB1 or application of glycyrrhizin, a specific HMGB1 inhibitor, aggravated OGD-induced OL death, and recombinant HMGB1 application reduced the extent of OL death in a TLR2-dependent manner. Glycyrrhizic Acid 37-49 high mobility group box 1 Homo sapiens 62-67 28216372-7 2017 CONCLUSION: HBx promotes the progression of HCC through translocation and secretion of HMGB1 from tumor cells via calcium dependent cascades. Calcium 114-121 high mobility group box 1 Homo sapiens 87-92 28373282-0 2017 Metformin directly binds the alarmin HMGB1 and inhibits its proinflammatory activity. Metformin 0-9 high mobility group box 1 Homo sapiens 37-42 28373282-4 2017 Here we identified HMGB1 as a novel metformin-binding protein by affinity purification using a biotinylated metformin analogue. Metformin 36-45 high mobility group box 1 Homo sapiens 19-24 28373282-5 2017 Metformin directly bound to the C-terminal acidic tail of HMGB1. Metformin 0-9 high mobility group box 1 Homo sapiens 58-63 28373282-6 2017 Both in vitro and in vivo, metformin inhibited inflammatory responses induced by full-length HMGB1 but not by HMGB1 lacking the acidic tail. Metformin 27-36 high mobility group box 1 Homo sapiens 93-98 28373282-7 2017 In an acetaminophen-induced acute liver injury model in which HMGB1 released from injured cells exacerbates the initial injury, metformin effectively reduced liver injury and had no additional inhibitory effects when the extracellular HMGB1 was blocked by anti-HMGB1-neutralizing antibody. Metformin 128-137 high mobility group box 1 Homo sapiens 235-240 28373282-7 2017 In an acetaminophen-induced acute liver injury model in which HMGB1 released from injured cells exacerbates the initial injury, metformin effectively reduced liver injury and had no additional inhibitory effects when the extracellular HMGB1 was blocked by anti-HMGB1-neutralizing antibody. Metformin 128-137 high mobility group box 1 Homo sapiens 235-240 28373282-8 2017 In summary, we report for the first time that metformin suppresses inflammation by inhibiting the extracellular activity of HMGB1. Metformin 46-55 high mobility group box 1 Homo sapiens 124-129 29069735-0 2017 HMGB1-mediated autophagy attenuates gemcitabine-induced apoptosis in bladder cancer cells involving JNK and ERK activation. gemcitabine 36-47 high mobility group box 1 Homo sapiens 0-5 28426082-0 2017 Deciphering of interactions between platinated DNA and HMGB1 by hydrogen/deuterium exchange mass spectrometry. Hydrogen 64-72 high mobility group box 1 Homo sapiens 55-60 28426082-0 2017 Deciphering of interactions between platinated DNA and HMGB1 by hydrogen/deuterium exchange mass spectrometry. Deuterium 73-82 high mobility group box 1 Homo sapiens 55-60 28426082-1 2017 A high mobility group box 1 (HMGB1) protein has been reported to recognize both 1,2-intrastrand crosslinked DNA by cisplatin (1,2-cis-Pt-DNA) and monofunctional platinated DNA using trans-[PtCl2(NH3)(thiazole)] (1-trans-PtTz-DNA). Cisplatin 115-124 high mobility group box 1 Homo sapiens 2-27 28426082-1 2017 A high mobility group box 1 (HMGB1) protein has been reported to recognize both 1,2-intrastrand crosslinked DNA by cisplatin (1,2-cis-Pt-DNA) and monofunctional platinated DNA using trans-[PtCl2(NH3)(thiazole)] (1-trans-PtTz-DNA). Cisplatin 115-124 high mobility group box 1 Homo sapiens 29-34 28426082-1 2017 A high mobility group box 1 (HMGB1) protein has been reported to recognize both 1,2-intrastrand crosslinked DNA by cisplatin (1,2-cis-Pt-DNA) and monofunctional platinated DNA using trans-[PtCl2(NH3)(thiazole)] (1-trans-PtTz-DNA). ptcl2(nh3) 189-199 high mobility group box 1 Homo sapiens 2-27 28426082-1 2017 A high mobility group box 1 (HMGB1) protein has been reported to recognize both 1,2-intrastrand crosslinked DNA by cisplatin (1,2-cis-Pt-DNA) and monofunctional platinated DNA using trans-[PtCl2(NH3)(thiazole)] (1-trans-PtTz-DNA). ptcl2(nh3) 189-199 high mobility group box 1 Homo sapiens 29-34 28426082-1 2017 A high mobility group box 1 (HMGB1) protein has been reported to recognize both 1,2-intrastrand crosslinked DNA by cisplatin (1,2-cis-Pt-DNA) and monofunctional platinated DNA using trans-[PtCl2(NH3)(thiazole)] (1-trans-PtTz-DNA). Thiazoles 200-208 high mobility group box 1 Homo sapiens 2-27 28426082-1 2017 A high mobility group box 1 (HMGB1) protein has been reported to recognize both 1,2-intrastrand crosslinked DNA by cisplatin (1,2-cis-Pt-DNA) and monofunctional platinated DNA using trans-[PtCl2(NH3)(thiazole)] (1-trans-PtTz-DNA). Thiazoles 200-208 high mobility group box 1 Homo sapiens 29-34 28426082-3 2017 In the present work, we described a hydrogen/deuterium exchange mass spectrometry (HDX-MS) method in combination with docking simulation to decipher the interactions of platinated DNA with domain A of HMGB1. Hydrogen 36-44 high mobility group box 1 Homo sapiens 201-206 28426082-3 2017 In the present work, we described a hydrogen/deuterium exchange mass spectrometry (HDX-MS) method in combination with docking simulation to decipher the interactions of platinated DNA with domain A of HMGB1. Deuterium 45-54 high mobility group box 1 Homo sapiens 201-206 29069735-8 2017 Interestingly, suppressing HMGB1 expression attenuated gemcitabine-induced ERK and JNK activation and Bcl-2 phosphorylation. gemcitabine 55-66 high mobility group box 1 Homo sapiens 27-32 29069735-9 2017 Thus, our results suggest that while gemcitabine kills bladder cancer cells through apoptosis, a cytoprotective autophagy is also induced involving HMGB1-mediated JNK and ERK to counteract the cytotoxicity of gemcitabine, and intervention targeting this pathway may improve the anticancer efficacy of gemcitabine against bladder cancer. gemcitabine 209-220 high mobility group box 1 Homo sapiens 148-153 29069735-9 2017 Thus, our results suggest that while gemcitabine kills bladder cancer cells through apoptosis, a cytoprotective autophagy is also induced involving HMGB1-mediated JNK and ERK to counteract the cytotoxicity of gemcitabine, and intervention targeting this pathway may improve the anticancer efficacy of gemcitabine against bladder cancer. gemcitabine 209-220 high mobility group box 1 Homo sapiens 148-153 29069735-4 2017 While gemcitabine induced apoptotic cell death, it also induced HMGB1 expression and autophagy in bladder cancer T24 and BIU-87 cells. gemcitabine 6-17 high mobility group box 1 Homo sapiens 64-69 29069735-5 2017 Suppressing HMGB1 expression with siRNA strongly potentiated gemcitabine-induced apoptosis. gemcitabine 61-72 high mobility group box 1 Homo sapiens 12-17 29069735-6 2017 HMGB1 siRNA or autophagy inhibitors suppressed gemcitabine-induced autophagy. gemcitabine 47-58 high mobility group box 1 Homo sapiens 0-5 28210782-6 2017 Multiple TLRs, HMGB1, and miRNAs are induced in the brain by stress, alcohol, and other drugs of abuse and are increased in the postmortem human alcoholic brain. Alcohols 69-76 high mobility group box 1 Homo sapiens 15-20 27908811-12 2017 CONCLUSIONS: Removal or isolation of AL via surgical intervention significantly decreases serum HMGB-1 levels. Aluminum 37-39 high mobility group box 1 Homo sapiens 96-102 28407046-4 2017 Methods and results: Expression of RAGE and its activating ligands, S100/calgranulins and high mobility group box 1 (HMGB1), were increased in both human and mouse pulmonary arterial smooth muscle cells (PASMCs) under hypoxic conditions and were also strikingly upregulated in pulmonary arteries in hypoxia plus SU5416 (HySu)-induced PAH in mice. pasmcs 204-210 high mobility group box 1 Homo sapiens 90-115 28407046-4 2017 Methods and results: Expression of RAGE and its activating ligands, S100/calgranulins and high mobility group box 1 (HMGB1), were increased in both human and mouse pulmonary arterial smooth muscle cells (PASMCs) under hypoxic conditions and were also strikingly upregulated in pulmonary arteries in hypoxia plus SU5416 (HySu)-induced PAH in mice. pasmcs 204-210 high mobility group box 1 Homo sapiens 117-122 27943400-5 2017 The injection of HMGB1 into the IMQ-treated skin further aggravated the psoriasis-like disease, enhanced the infiltration of CD3+ T cells, myeloperoxidase+ neutrophils and CD11c+ dendritic cells, increased the number of gammadelta T cells, and upregulated the mRNA expression of interleukin (IL)-6, tumor necrosis factor (TNF)-alpha, interferon (IFN)-gamma and IL-17 compared with the PBS injection. Imiquimod 32-35 high mobility group box 1 Homo sapiens 17-22 27943400-5 2017 The injection of HMGB1 into the IMQ-treated skin further aggravated the psoriasis-like disease, enhanced the infiltration of CD3+ T cells, myeloperoxidase+ neutrophils and CD11c+ dendritic cells, increased the number of gammadelta T cells, and upregulated the mRNA expression of interleukin (IL)-6, tumor necrosis factor (TNF)-alpha, interferon (IFN)-gamma and IL-17 compared with the PBS injection. pbs 385-388 high mobility group box 1 Homo sapiens 17-22 27624778-0 2017 ROS-dependent HMGB1 secretion upregulates IL-8 in upper airway epithelial cells under hypoxic condition. Reactive Oxygen Species 0-3 high mobility group box 1 Homo sapiens 14-19 27624778-4 2017 Reactive oxygen species (ROS) level was evaluated to estimate the translocation mechanism of HMGB1. Reactive Oxygen Species 0-23 high mobility group box 1 Homo sapiens 93-98 27624778-4 2017 Reactive oxygen species (ROS) level was evaluated to estimate the translocation mechanism of HMGB1. Reactive Oxygen Species 25-28 high mobility group box 1 Homo sapiens 93-98 27624778-7 2017 Hypoxia induced translocation of HMGB1 into the extracellular area and it was dependent on ROS produced by dual oxidase 2. Reactive Oxygen Species 91-94 high mobility group box 1 Homo sapiens 33-38 27624778-11 2017 We suggest that HMGB1 is secreted in hypoxic condition via ROS-dependent mechanism and secreted HMGB1 participates in IL-8 upregulation mediating inflammatory response. Reactive Oxygen Species 59-62 high mobility group box 1 Homo sapiens 16-21 28404891-3 2017 HMGB1 and the pro-inflammatory cytokines IL-1beta and TNF-alpha were gradually released from NB4 and HL-60 cells treated with ATRA and/or ATO. Tretinoin 126-130 high mobility group box 1 Homo sapiens 0-5 28404891-3 2017 HMGB1 and the pro-inflammatory cytokines IL-1beta and TNF-alpha were gradually released from NB4 and HL-60 cells treated with ATRA and/or ATO. Arsenic Trioxide 138-141 high mobility group box 1 Homo sapiens 0-5 28404891-8 2017 Treatment with a HMGB1-neutralizing antibody reduced secretion of TNF-alpha and IL-1beta, arrested the elevation of ICAM-1 and blunted the activation of ERK1/2 in ATRA-induced NB4 cells. Tretinoin 163-167 high mobility group box 1 Homo sapiens 17-22 28616398-7 2017 We also found that PDT with G-chlorin induced immunogenic cell death which is characterized by the secretion, release, or surface exposure of damage-associated molecular patterns (DAMPs), including calreticulin (CRT) and the high-mobility group box 1 (HMGB1) protein. g-chlorin 28-37 high mobility group box 1 Homo sapiens 225-250 28469775-0 2017 Melatonin promoted renal regeneration in folic acid-induced acute kidney injury via inhibiting nucleocytoplasmic translocation of HMGB1 in tubular epithelial cells. Melatonin 0-9 high mobility group box 1 Homo sapiens 130-135 28469775-0 2017 Melatonin promoted renal regeneration in folic acid-induced acute kidney injury via inhibiting nucleocytoplasmic translocation of HMGB1 in tubular epithelial cells. Folic Acid 41-51 high mobility group box 1 Homo sapiens 130-135 28469775-2 2017 High-mobility group box 1 (HMGB1) is a novel member of the damage-associated molecular pattern (DAMP) family, and has been verified to be an inflammatory cytokine mediating AKI induced by I/R and cisplatin. Cisplatin 196-205 high mobility group box 1 Homo sapiens 0-25 28469775-2 2017 High-mobility group box 1 (HMGB1) is a novel member of the damage-associated molecular pattern (DAMP) family, and has been verified to be an inflammatory cytokine mediating AKI induced by I/R and cisplatin. Cisplatin 196-205 high mobility group box 1 Homo sapiens 27-32 28469775-4 2017 In this study, we sought to identify the role of melatonin on folic acid induced AKI and its association with HMGB1. Melatonin 49-58 high mobility group box 1 Homo sapiens 110-115 28469775-7 2017 This protective role of melatonin was closely related to the inhibition of nucleocytoplasmic translocation of HMGB1 in TECs. Melatonin 24-33 high mobility group box 1 Homo sapiens 110-115 28186988-0 2017 HMGB1 targeting by ethyl pyruvate suppresses malignant phenotype of human mesothelioma. ethyl pyruvate 19-33 high mobility group box 1 Homo sapiens 0-5 28186988-4 2017 Ethyl pyruvate (EP), the ethyl ester of pyruvic acid, has been shown to be an effective HMGB1 inhibitor in inflammation-related diseases and several cancers. ethyl pyruvate 0-14 high mobility group box 1 Homo sapiens 88-93 28186988-4 2017 Ethyl pyruvate (EP), the ethyl ester of pyruvic acid, has been shown to be an effective HMGB1 inhibitor in inflammation-related diseases and several cancers. ethyl pyruvate 16-18 high mobility group box 1 Homo sapiens 88-93 28186988-4 2017 Ethyl pyruvate (EP), the ethyl ester of pyruvic acid, has been shown to be an effective HMGB1 inhibitor in inflammation-related diseases and several cancers. Pyruvic Acid 40-52 high mobility group box 1 Homo sapiens 88-93 28616398-7 2017 We also found that PDT with G-chlorin induced immunogenic cell death which is characterized by the secretion, release, or surface exposure of damage-associated molecular patterns (DAMPs), including calreticulin (CRT) and the high-mobility group box 1 (HMGB1) protein. g-chlorin 28-37 high mobility group box 1 Homo sapiens 252-257 28089782-9 2017 In the supernatant of co-cultured, but not single-cultured HepG2 and Huh-7 cells, o-anisidine caused increases of damage-associated proteins, such as HMGB1 (high mobility group box-1) protein. 2-anisidine 82-93 high mobility group box 1 Homo sapiens 150-155 28685526-0 2017 Glycyrrhetinic acid suppressed hmgb1 release by up-regulation of Sirt6 in nasal inflammation. Glycyrrhetinic Acid 0-19 high mobility group box 1 Homo sapiens 31-36 28685526-7 2017 In an in vitro study we used the 18-beta-stereoisomer of GTA to enhance Sirt6 expression levels, inhibiting through this mechanism the translocation of HMGB1 protein from nucleus and reversing its extracellular accumulation stimulated by lipopolysaccharides. Glycyrrhetinic Acid 57-60 high mobility group box 1 Homo sapiens 152-157 28089782-9 2017 In the supernatant of co-cultured, but not single-cultured HepG2 and Huh-7 cells, o-anisidine caused increases of damage-associated proteins, such as HMGB1 (high mobility group box-1) protein. 2-anisidine 82-93 high mobility group box 1 Homo sapiens 157-182 28024939-11 2017 RAGE or ER stress knockdown reduced the up-regulation of monocyte chemoattractant protein-1 (MCP-1) and tumor necrosis factor-alpha (TNF-alpha) in human AVICs exposed to HMGB1.These novel findings demonstrate that RAGE deficiency protects against aortic valve calcification in high cholesterol diet-fed ApoE-/- mice via inhibition of ER stress. Cholesterol 282-293 high mobility group box 1 Homo sapiens 170-175 28443467-3 2017 In this study, the effect of irinotecan on high-mobility group protein B1 and MMP9 content, gene expression, cell cycle, and cell growth in human breast cancer cells (MCF-7) was investigated. Irinotecan 29-39 high mobility group box 1 Homo sapiens 43-73 28443467-11 2017 From the results, it is concluded that overexpression of high-mobility group protein B1 in the presence of irinotecan precedes breast cancer cells into apoptosis and in this response the binding of irinotecan to chromatin or high-mobility group protein B1 may condense/aggregate chromatin, preventing high-mobility group protein B1 release from chromatin. Irinotecan 107-117 high mobility group box 1 Homo sapiens 57-87 28443467-11 2017 From the results, it is concluded that overexpression of high-mobility group protein B1 in the presence of irinotecan precedes breast cancer cells into apoptosis and in this response the binding of irinotecan to chromatin or high-mobility group protein B1 may condense/aggregate chromatin, preventing high-mobility group protein B1 release from chromatin. Irinotecan 198-208 high mobility group box 1 Homo sapiens 57-87 28396833-11 2017 HMGB1, sRAGE, IL6 and TGFbeta1 levels in BALF were also reduced by baicalin treatment. baicalin 67-75 high mobility group box 1 Homo sapiens 0-5 28396833-13 2017 Furthermore, expression levels of HMGB1, RAGE, IL6 and TGFbeta1 in lung tissue were dramatically decreased by baicalin in a dosage-dependent manner. baicalin 110-118 high mobility group box 1 Homo sapiens 34-39 28049065-4 2017 Herein, we demonstrated that nucleic acid binding polymers, e.g., polyethylenimine (PEI) and polyamidoamine dendrimers, immobilized onto electrospun microfiber mesh can effectively capture various DAMPs, such as extracellular DNAs and high mobility group box 1 (HMGB1). Polymers 50-58 high mobility group box 1 Homo sapiens 235-260 28049065-4 2017 Herein, we demonstrated that nucleic acid binding polymers, e.g., polyethylenimine (PEI) and polyamidoamine dendrimers, immobilized onto electrospun microfiber mesh can effectively capture various DAMPs, such as extracellular DNAs and high mobility group box 1 (HMGB1). Polymers 50-58 high mobility group box 1 Homo sapiens 262-267 28049065-4 2017 Herein, we demonstrated that nucleic acid binding polymers, e.g., polyethylenimine (PEI) and polyamidoamine dendrimers, immobilized onto electrospun microfiber mesh can effectively capture various DAMPs, such as extracellular DNAs and high mobility group box 1 (HMGB1). Polyethyleneimine 66-82 high mobility group box 1 Homo sapiens 235-260 28049065-4 2017 Herein, we demonstrated that nucleic acid binding polymers, e.g., polyethylenimine (PEI) and polyamidoamine dendrimers, immobilized onto electrospun microfiber mesh can effectively capture various DAMPs, such as extracellular DNAs and high mobility group box 1 (HMGB1). Polyethyleneimine 66-82 high mobility group box 1 Homo sapiens 262-267 28049065-4 2017 Herein, we demonstrated that nucleic acid binding polymers, e.g., polyethylenimine (PEI) and polyamidoamine dendrimers, immobilized onto electrospun microfiber mesh can effectively capture various DAMPs, such as extracellular DNAs and high mobility group box 1 (HMGB1). Polyethyleneimine 84-87 high mobility group box 1 Homo sapiens 235-260 28049065-4 2017 Herein, we demonstrated that nucleic acid binding polymers, e.g., polyethylenimine (PEI) and polyamidoamine dendrimers, immobilized onto electrospun microfiber mesh can effectively capture various DAMPs, such as extracellular DNAs and high mobility group box 1 (HMGB1). Polyethyleneimine 84-87 high mobility group box 1 Homo sapiens 262-267 27393275-0 2017 Urinary MCP-1 HMGB1 increased in calcium nephrolithiasis patients and the influence of hypercalciuria on the production of the two cytokines. Calcium 33-40 high mobility group box 1 Homo sapiens 14-19 27393275-9 2017 Results showed that urinary MCP-1 and HMGB1 increased in calcium nephrolithiasis patients and hypercalciuria might affect the identical pathways (through the reactive oxygen species) with other factors in stimulating the production of MCP-1 and HMGB1 in vivo. Calcium 57-64 high mobility group box 1 Homo sapiens 38-43 27393275-9 2017 Results showed that urinary MCP-1 and HMGB1 increased in calcium nephrolithiasis patients and hypercalciuria might affect the identical pathways (through the reactive oxygen species) with other factors in stimulating the production of MCP-1 and HMGB1 in vivo. Reactive Oxygen Species 158-181 high mobility group box 1 Homo sapiens 245-250 28218519-2 2017 Herein, a simple and sensitive sensor system was tailored for real-time probing of the dynamic molecular recognition between cisplatin-damaged-DNA (cisPt-DNA) and a cellular responsive protein, high-mobility-group box 1 (HMGB1). Cisplatin 125-134 high mobility group box 1 Homo sapiens 194-219 28218519-2 2017 Herein, a simple and sensitive sensor system was tailored for real-time probing of the dynamic molecular recognition between cisplatin-damaged-DNA (cisPt-DNA) and a cellular responsive protein, high-mobility-group box 1 (HMGB1). Cisplatin 125-134 high mobility group box 1 Homo sapiens 221-226 28049065-4 2017 Herein, we demonstrated that nucleic acid binding polymers, e.g., polyethylenimine (PEI) and polyamidoamine dendrimers, immobilized onto electrospun microfiber mesh can effectively capture various DAMPs, such as extracellular DNAs and high mobility group box 1 (HMGB1). Poly(amidoamine) 93-107 high mobility group box 1 Homo sapiens 235-260 28012970-0 2017 Isoliquiritigenin inhibits TNF-alpha-induced release of high-mobility group box 1 through activation of HDAC in human intestinal epithelial HT-29 cells. isoliquiritigenin 0-17 high mobility group box 1 Homo sapiens 56-81 28049065-4 2017 Herein, we demonstrated that nucleic acid binding polymers, e.g., polyethylenimine (PEI) and polyamidoamine dendrimers, immobilized onto electrospun microfiber mesh can effectively capture various DAMPs, such as extracellular DNAs and high mobility group box 1 (HMGB1). Poly(amidoamine) 93-107 high mobility group box 1 Homo sapiens 262-267 28012822-12 2017 Pharmacological inhibition of HMGB1 cytoplasmic translocation by agents such as ethyl pyruvate limits starvation-induced autophagy. ethyl pyruvate 80-94 high mobility group box 1 Homo sapiens 30-35 28216608-8 2017 The multivariate statistical analysis indicated an iron-related 10-protein panel effective in separating non-cancerous from cancerous lesions including STAT5, STAT5_pY694, myeloid differentiation factor 88 (MYD88), CD74, iron exporter ferroportin (FPN), high mobility group box 1 (HMGB1), STAT3_pS727, TFRC, ferritin heavy chain (FTH), and ferritin light chain (FTL). Iron 51-55 high mobility group box 1 Homo sapiens 254-279 28216608-8 2017 The multivariate statistical analysis indicated an iron-related 10-protein panel effective in separating non-cancerous from cancerous lesions including STAT5, STAT5_pY694, myeloid differentiation factor 88 (MYD88), CD74, iron exporter ferroportin (FPN), high mobility group box 1 (HMGB1), STAT3_pS727, TFRC, ferritin heavy chain (FTH), and ferritin light chain (FTL). Iron 51-55 high mobility group box 1 Homo sapiens 281-286 27579780-9 2017 Additionally, pretreatment with resveratrol, a selective SIRT1 activator, abrogated IFN-gamma-induced HMGB1 translocation and its release. Resveratrol 32-43 high mobility group box 1 Homo sapiens 102-107 27579780-11 2017 In contrast, blocking HMGB1 release or activity by resveratrol and HMGB1-neutralizing antibody prevents IFN-gamma-induced phenotypic modulation of VSMCs. Resveratrol 51-62 high mobility group box 1 Homo sapiens 22-27 27579780-11 2017 In contrast, blocking HMGB1 release or activity by resveratrol and HMGB1-neutralizing antibody prevents IFN-gamma-induced phenotypic modulation of VSMCs. vsmcs 147-152 high mobility group box 1 Homo sapiens 22-27 27579780-11 2017 In contrast, blocking HMGB1 release or activity by resveratrol and HMGB1-neutralizing antibody prevents IFN-gamma-induced phenotypic modulation of VSMCs. vsmcs 147-152 high mobility group box 1 Homo sapiens 67-72 28118842-0 2017 Microglial-derived miRNA let-7 and HMGB1 contribute to ethanol-induced neurotoxicity via TLR7. Ethanol 55-62 high mobility group box 1 Homo sapiens 35-40 27838489-9 2017 HMGB1 inhibition by ethyl pyruvate or blockade by neutralizing antibodies significantly decreased the phosphorylation of STAT3, p38 and IkappaBalpha, the production of IL-1beta and TNF-alpha, and the islet injury in wild-type islets after exposure to H/R and significantly improved early islet graft failure. ethyl pyruvate 20-34 high mobility group box 1 Homo sapiens 0-5 28000879-11 2017 Treatment with NF-kappaB inhibitor (BAY11-7082) and receptor for advanced glycation end products (RAGE) antagonist (anti-RAGE) significantly suppressed HMGB1-mediated epithelial-to-mesenchymal transition in the HeLa cervical cancer cells. 3-(4-methylphenylsulfonyl)-2-propenenitrile 36-46 high mobility group box 1 Homo sapiens 152-157 28197072-4 2017 Here in this study, we investigated the interaction between HMGB1 and alpha-synuclein in rotenone-induced PD cell models and their roles in autophagy flux. Rotenone 89-97 high mobility group box 1 Homo sapiens 60-65 28197072-5 2017 Results revealed elevated expression and cytosolic translocation of endogenous HMGB1 upon rotenone exposure. Rotenone 90-98 high mobility group box 1 Homo sapiens 79-84 28197072-8 2017 Based on these findings, we propose that HMGB1 is involved in rotenone-induced dopaminergic cell death via interacting with alpha-synuclein, perturbing the autophagy process, aggravating protein aggregation and finally propelling dopaminergic neurons to move from morbidity to mortality. Rotenone 62-70 high mobility group box 1 Homo sapiens 41-46 27840243-5 2017 Further investigation found that simvastatin reversed the TAM-mediated down-regulation of Gfi-1 and up-regulation of CTGF and HMGB1. Simvastatin 33-44 high mobility group box 1 Homo sapiens 126-131 27840243-5 2017 Further investigation found that simvastatin reversed the TAM-mediated down-regulation of Gfi-1 and up-regulation of CTGF and HMGB1. tam 58-61 high mobility group box 1 Homo sapiens 126-131 28118842-10 2017 Further, ethanol increased let-7b binding to the danger signaling molecule high mobility group box-1 (HMGB1) in MVs, while reducing let-7 binding to classical chaperone protein argonaute (Ago2). Ethanol 9-16 high mobility group box 1 Homo sapiens 75-100 28118842-10 2017 Further, ethanol increased let-7b binding to the danger signaling molecule high mobility group box-1 (HMGB1) in MVs, while reducing let-7 binding to classical chaperone protein argonaute (Ago2). Ethanol 9-16 high mobility group box 1 Homo sapiens 102-107 28118842-10 2017 Further, ethanol increased let-7b binding to the danger signaling molecule high mobility group box-1 (HMGB1) in MVs, while reducing let-7 binding to classical chaperone protein argonaute (Ago2). let-7 27-32 high mobility group box 1 Homo sapiens 75-100 28118842-10 2017 Further, ethanol increased let-7b binding to the danger signaling molecule high mobility group box-1 (HMGB1) in MVs, while reducing let-7 binding to classical chaperone protein argonaute (Ago2). let-7 27-32 high mobility group box 1 Homo sapiens 102-107 28118842-12 2017 CONCLUSIONS: Our results identify that ethanol induces neuroimmune pathology involving the release of let-7b/HMGB1 complexes in microglia-derived microvesicles. Ethanol 39-46 high mobility group box 1 Homo sapiens 109-114 28116308-0 2017 Uric Acid Induces Endothelial Dysfunction by Activating the HMGB1/RAGE Signaling Pathway. Uric Acid 0-9 high mobility group box 1 Homo sapiens 60-65 28270074-4 2017 OBJECTIVE: In the present study, we have mobilized a proximal twenty one base pair nucleotide (21RY) which is in the promoter region (-55 to-75) of hmgb1 gene and targeted it with triplex forming oligonucleotide (TFO) in combination with two widely used chemotherapeutic drugs, actinomycin (ACT) and adriamycin (ADM). triplex forming oligonucleotide 180-211 high mobility group box 1 Homo sapiens 148-153 28270074-4 2017 OBJECTIVE: In the present study, we have mobilized a proximal twenty one base pair nucleotide (21RY) which is in the promoter region (-55 to-75) of hmgb1 gene and targeted it with triplex forming oligonucleotide (TFO) in combination with two widely used chemotherapeutic drugs, actinomycin (ACT) and adriamycin (ADM). tfo 213-216 high mobility group box 1 Homo sapiens 148-153 28270074-4 2017 OBJECTIVE: In the present study, we have mobilized a proximal twenty one base pair nucleotide (21RY) which is in the promoter region (-55 to-75) of hmgb1 gene and targeted it with triplex forming oligonucleotide (TFO) in combination with two widely used chemotherapeutic drugs, actinomycin (ACT) and adriamycin (ADM). Dactinomycin 278-289 high mobility group box 1 Homo sapiens 148-153 28270074-4 2017 OBJECTIVE: In the present study, we have mobilized a proximal twenty one base pair nucleotide (21RY) which is in the promoter region (-55 to-75) of hmgb1 gene and targeted it with triplex forming oligonucleotide (TFO) in combination with two widely used chemotherapeutic drugs, actinomycin (ACT) and adriamycin (ADM). Doxorubicin 300-310 high mobility group box 1 Homo sapiens 148-153 28270074-4 2017 OBJECTIVE: In the present study, we have mobilized a proximal twenty one base pair nucleotide (21RY) which is in the promoter region (-55 to-75) of hmgb1 gene and targeted it with triplex forming oligonucleotide (TFO) in combination with two widely used chemotherapeutic drugs, actinomycin (ACT) and adriamycin (ADM). Doxorubicin 312-315 high mobility group box 1 Homo sapiens 148-153 28241714-0 2017 [Molecular mechanisms of HMGB1 extracellular secretion in asbestos-induced malignant mesothelioma]. Asbestos 58-66 high mobility group box 1 Homo sapiens 25-30 28059131-7 2017 Our in vivo and in vitro studies show dioscin protects hippocampus from endotoxemia induced cascade neuro-inflammation through neurotransmitter 5-HT and HMGB-1/TLR4 signaling pathway, which accounts for the dioscin therapeutic effect in behavioral tests. dioscin 38-45 high mobility group box 1 Homo sapiens 153-159 28059131-7 2017 Our in vivo and in vitro studies show dioscin protects hippocampus from endotoxemia induced cascade neuro-inflammation through neurotransmitter 5-HT and HMGB-1/TLR4 signaling pathway, which accounts for the dioscin therapeutic effect in behavioral tests. dioscin 207-214 high mobility group box 1 Homo sapiens 153-159 27585400-0 2017 2-O, 3-O Desulfated Heparin Blocks High Mobility Group Box 1 Release by Inhibition of p300 Acetyltransferase Activity. 2-o, 3-o desulfated heparin 0-27 high mobility group box 1 Homo sapiens 35-60 28116308-2 2017 UA increases high mobility group box chromosomal protein 1 (HMGB1) expression and extracellular release in endothelial cells. Uric Acid 0-2 high mobility group box 1 Homo sapiens 60-65 28116308-5 2017 UA inhibited endothelial nitric oxide synthase (eNOS) expression and nitric oxide (NO) production in HUVECs, increased intracellular HMGB1 expression and extracellular HMGB1 secretion, and upregulated RAGE expression. Uric Acid 0-2 high mobility group box 1 Homo sapiens 133-138 28116308-5 2017 UA inhibited endothelial nitric oxide synthase (eNOS) expression and nitric oxide (NO) production in HUVECs, increased intracellular HMGB1 expression and extracellular HMGB1 secretion, and upregulated RAGE expression. Uric Acid 0-2 high mobility group box 1 Homo sapiens 168-173 28116308-9 2017 Our results suggest that high concentrations of UA cause endothelial dysfunction via the HMGB1/RAGE signaling pathway. Uric Acid 48-50 high mobility group box 1 Homo sapiens 89-94 28478444-4 2017 Lipofectamine 2000 was used to transfect with miR-34a mimics, miR-34a inhibitor, si-HMGB1, pcDNA 3.1-HMGB1, and corresponding controls. Lipofectamine 0-18 high mobility group box 1 Homo sapiens 84-89 28478444-4 2017 Lipofectamine 2000 was used to transfect with miR-34a mimics, miR-34a inhibitor, si-HMGB1, pcDNA 3.1-HMGB1, and corresponding controls. Lipofectamine 0-18 high mobility group box 1 Homo sapiens 101-106 28052268-5 2017 Once mucosal barrier function is disrupted due to NSAID-induced prostaglandin deficiency and mitochondrial malfunction, lipopolysaccharide from luminal gram-negative bacteria and high mobility group box 1 from the injured epithelial cells activate toll-like receptor 4-signaling pathway and nucleotide-binding oligomerization domain-like receptor family, pyrin domain-containing 3 inflammasome; this leads to the release of proinflammatory cytokines such as tumor necrosis factor-alpha and interleukin-1beta. Prostaglandins 64-77 high mobility group box 1 Homo sapiens 179-204 27488419-0 2017 Every Cloud Has a Silver Lining: Proneurogenic Effects of Abeta Oligomers and HMGB-1 via Activation of the RAGE-NF-kappaB Axis. Silver 18-24 high mobility group box 1 Homo sapiens 78-84 27931589-6 2016 The serum levels of high-mobility group box 1 protein showed a peak at 8 hours after occlusion in control group, and this elevation was significantly blunted by 4.5 mg/kg NecroX-7 treatment. necrox-7 171-179 high mobility group box 1 Homo sapiens 20-45 27908419-5 2017 The selenium levels in the tissues were correlated to the genotype of the SELENOP selenium carrier protein, but not to other proteins whose levels have been reported to be responsive to selenium availability, including GPX1, SELENOF and SBP1. Selenium 4-12 high mobility group box 1 Homo sapiens 237-241 29249869-7 2017 The patients with POCD had higher serum interleukin 1beta (IL-1beta), serum amyloid A (SAA), S100 calcium-binding protein beta (S-100beta), and high mobility group box-1 protein (HMGB-1) levels at 1 and 24 h postoperatively than patients without POCD. pocd 18-22 high mobility group box 1 Homo sapiens 144-169 29249869-7 2017 The patients with POCD had higher serum interleukin 1beta (IL-1beta), serum amyloid A (SAA), S100 calcium-binding protein beta (S-100beta), and high mobility group box-1 protein (HMGB-1) levels at 1 and 24 h postoperatively than patients without POCD. pocd 18-22 high mobility group box 1 Homo sapiens 179-185 27560926-7 2017 High glucose induced a significant upregulation of HMGB1 and pSTAT-3 upregulation and nuclear translocation in retinal Muller cells. Glucose 5-12 high mobility group box 1 Homo sapiens 51-56 27560926-8 2017 GA co-treatment normalized the high-glucose-induced upregulation of HMGB1 and pSTAT-3 upregulation and nuclear translocation in Muller cells. Gallium 0-2 high mobility group box 1 Homo sapiens 68-73 27560926-8 2017 GA co-treatment normalized the high-glucose-induced upregulation of HMGB1 and pSTAT-3 upregulation and nuclear translocation in Muller cells. Glucose 36-43 high mobility group box 1 Homo sapiens 68-73 26239884-3 2017 Here, we examined the effects of methylthiouracil (MTU), used as antithyroid drug, by monitoring the effects on lipopolysaccharide (LPS)- or cecal ligation and puncture (CLP)-mediated release of HMGB1, and on the modulation of HMGB1-mediated inflammatory responses. Methylthiouracil 33-49 high mobility group box 1 Homo sapiens 195-200 26239884-3 2017 Here, we examined the effects of methylthiouracil (MTU), used as antithyroid drug, by monitoring the effects on lipopolysaccharide (LPS)- or cecal ligation and puncture (CLP)-mediated release of HMGB1, and on the modulation of HMGB1-mediated inflammatory responses. Methylthiouracil 33-49 high mobility group box 1 Homo sapiens 227-232 26239884-3 2017 Here, we examined the effects of methylthiouracil (MTU), used as antithyroid drug, by monitoring the effects on lipopolysaccharide (LPS)- or cecal ligation and puncture (CLP)-mediated release of HMGB1, and on the modulation of HMGB1-mediated inflammatory responses. Methylthiouracil 51-54 high mobility group box 1 Homo sapiens 195-200 26239884-3 2017 Here, we examined the effects of methylthiouracil (MTU), used as antithyroid drug, by monitoring the effects on lipopolysaccharide (LPS)- or cecal ligation and puncture (CLP)-mediated release of HMGB1, and on the modulation of HMGB1-mediated inflammatory responses. Methylthiouracil 51-54 high mobility group box 1 Homo sapiens 227-232 26239884-5 2017 MTU inhibited the release of HMGB1 and downregulated HMGB1-dependent inflammatory responses in human endothelial cells. Methylthiouracil 0-3 high mobility group box 1 Homo sapiens 29-34 26239884-5 2017 MTU inhibited the release of HMGB1 and downregulated HMGB1-dependent inflammatory responses in human endothelial cells. Methylthiouracil 0-3 high mobility group box 1 Homo sapiens 53-58 26239884-7 2017 In addition, treatment with MTU reduced CLP-induced release of HMGB1 and sepsis-related mortality and pulmonary injury. Methylthiouracil 28-31 high mobility group box 1 Homo sapiens 63-68 27983705-6 2016 Our results indicated an efficient reduction in pro-inflammatory factors (TNFalpha, IL-6, MCP-1, HMGB1) upon culture with riboflavin supplementation (500-1000 nM), accompanied by elevation in anti-inflammatory adiponectin and IL-10. Riboflavin 122-132 high mobility group box 1 Homo sapiens 97-102 27075010-8 2016 Urine protein to urine creatinine ratio (P/C) correlated with urinary HMGB-1 (r = 0.52, p < 0.001), but across the classes this was true only for membranous disease (r = 0.71, p = 0.022, proliferative, p = 0.63; mixed, p = 0.34). Creatinine 23-33 high mobility group box 1 Homo sapiens 70-76 27959427-0 2017 Inhibiting the cytoplasmic location of HMGB1 reverses cisplatin resistance in human cervical cancer cells. Cisplatin 54-63 high mobility group box 1 Homo sapiens 39-44 27959427-3 2017 When human cervical cancer cells were treated with 10 microg/ml of cisplatin for 24 and 48 h, high mobility group box 1 (HMGB1) protein expression levels significantly increased in a time-dependent manner. Cisplatin 67-76 high mobility group box 1 Homo sapiens 94-119 27959427-3 2017 When human cervical cancer cells were treated with 10 microg/ml of cisplatin for 24 and 48 h, high mobility group box 1 (HMGB1) protein expression levels significantly increased in a time-dependent manner. Cisplatin 67-76 high mobility group box 1 Homo sapiens 121-126 27959427-4 2017 Comparisons between cisplatin-sensitive HeLa cells and cisplatin-resistant HeLa/DDP cells revealed higher levels of HMGB1 in HeLa/DDP cells than in HeLa cells. Cisplatin 20-29 high mobility group box 1 Homo sapiens 116-121 27959427-4 2017 Comparisons between cisplatin-sensitive HeLa cells and cisplatin-resistant HeLa/DDP cells revealed higher levels of HMGB1 in HeLa/DDP cells than in HeLa cells. Cisplatin 55-64 high mobility group box 1 Homo sapiens 116-121 27959427-6 2017 Analysis of the distribution of cellular components revealed that HMGB1 translocation from the nucleus to cytoplasm contributed to cisplatin resistance. Cisplatin 131-140 high mobility group box 1 Homo sapiens 66-71 27959427-7 2017 This was further confirmed by demonstration that ethyl pyruvate treatment suppressed the cytoplasmic translocation of HMGB1, resulting in inhibition of HeLa cell proliferation. ethyl pyruvate 49-63 high mobility group box 1 Homo sapiens 118-123 27959427-8 2017 Furthermore, endogenous HMGB1 was inhibited with HMGB1-specific short hairpin (sh)RNA, and MTT assay results showed that interference with HMGB1 expression reduced cell viability and potentially reversed cisplatin resistance in HeLa cells. Cisplatin 204-213 high mobility group box 1 Homo sapiens 24-29 27959427-11 2017 Thus, cytoplasmic HMGB1 translocation and HMGB1-induced cell autophagy are proposed to contribute to cisplatin resistance by inhibiting apoptosis of cervical cancer cells. Cisplatin 101-110 high mobility group box 1 Homo sapiens 18-23 27959427-11 2017 Thus, cytoplasmic HMGB1 translocation and HMGB1-induced cell autophagy are proposed to contribute to cisplatin resistance by inhibiting apoptosis of cervical cancer cells. Cisplatin 101-110 high mobility group box 1 Homo sapiens 42-47 27717108-6 2016 The augmenting effects of glycyrrhizin were also observed in other synthetic HMGB1 inhibitors, gabexate mesilate, nafamostat, and sivelestat. Glycyrrhizic Acid 26-38 high mobility group box 1 Homo sapiens 77-82 27779688-0 2016 [Retracted] Inhibitory effects of ethyl pyruvate administration on human gastric cancer growth via regulation of the HMGB1-RAGE and Akt pathways in vitro and in vivo. ethyl pyruvate 34-48 high mobility group box 1 Homo sapiens 117-122 28024504-5 2016 RESULTS: Before treatment, the plasma HMGB1 level in ITP group was significantly higher than that in control group (t=4.259, P<0.01), while after treatment it significantly decreased and close to level in control group (t=1.267, P>0.05). Inosine Triphosphate 53-56 high mobility group box 1 Homo sapiens 38-43 27836008-8 2016 The degree of TAM infiltration was higher in peritumoral tissues with high HMGB1 expression than in peritumoral tissues with low HMGB1 expression (p < 0.001). tam 14-17 high mobility group box 1 Homo sapiens 75-80 27897164-0 2016 HMGB1-mediated autophagy decreases sensitivity to oxymatrine in SW982 human synovial sarcoma cells. oxymatrine 50-60 high mobility group box 1 Homo sapiens 0-5 27897164-10 2016 Therefore, we believe that combining OMT with an inhibitor of autophagy or HMGB1 may make OMT more effective in the treatment of human synovial sarcoma. oxymatrine 90-93 high mobility group box 1 Homo sapiens 75-80 27771306-3 2016 We found that the overexpression of HMGB1 A-box significantly decreased the IL-1beta-stimulated the production of MMP-1, MMP-3 and MMP-9, and also reduced the elevated levels of inducible nitric oxide synthase (iNOS) and cyclooxygenase-2 (COX-2) associated with the inhibition of prostaglandin E2 (PGE2) and nitric oxide (NO) production in IL-1beta-stimulated chondrocytes. Dinoprostone 280-296 high mobility group box 1 Homo sapiens 36-41 27771306-3 2016 We found that the overexpression of HMGB1 A-box significantly decreased the IL-1beta-stimulated the production of MMP-1, MMP-3 and MMP-9, and also reduced the elevated levels of inducible nitric oxide synthase (iNOS) and cyclooxygenase-2 (COX-2) associated with the inhibition of prostaglandin E2 (PGE2) and nitric oxide (NO) production in IL-1beta-stimulated chondrocytes. Dinoprostone 298-302 high mobility group box 1 Homo sapiens 36-41 27771306-3 2016 We found that the overexpression of HMGB1 A-box significantly decreased the IL-1beta-stimulated the production of MMP-1, MMP-3 and MMP-9, and also reduced the elevated levels of inducible nitric oxide synthase (iNOS) and cyclooxygenase-2 (COX-2) associated with the inhibition of prostaglandin E2 (PGE2) and nitric oxide (NO) production in IL-1beta-stimulated chondrocytes. Nitric Oxide 188-200 high mobility group box 1 Homo sapiens 36-41 27836008-8 2016 The degree of TAM infiltration was higher in peritumoral tissues with high HMGB1 expression than in peritumoral tissues with low HMGB1 expression (p < 0.001). tam 14-17 high mobility group box 1 Homo sapiens 129-134 27826297-8 2016 In contrast to the effects of LPS treatment, the alarmin high-mobility group protein B1 acts in synergy with polyI:C to promote TSLP and IL33 expression. Poly I-C 109-116 high mobility group box 1 Homo sapiens 57-87 27911399-7 2016 Here, we use a TFO covalently conjugated on the 5" end to a 4"-hydroxymethyl-4,5",8-trimethylpsoralen (HMT) psoralen to generate a site-specific ICL on a mutation-reporter plasmid to use as a tool to study the architectural modification, processing, and repair of complex DNA lesions by HMGB1 in human cells. tfo 15-18 high mobility group box 1 Homo sapiens 287-292 27911399-7 2016 Here, we use a TFO covalently conjugated on the 5" end to a 4"-hydroxymethyl-4,5",8-trimethylpsoralen (HMT) psoralen to generate a site-specific ICL on a mutation-reporter plasmid to use as a tool to study the architectural modification, processing, and repair of complex DNA lesions by HMGB1 in human cells. hydroxymethyltrioxsalen 60-101 high mobility group box 1 Homo sapiens 287-292 27911399-7 2016 Here, we use a TFO covalently conjugated on the 5" end to a 4"-hydroxymethyl-4,5",8-trimethylpsoralen (HMT) psoralen to generate a site-specific ICL on a mutation-reporter plasmid to use as a tool to study the architectural modification, processing, and repair of complex DNA lesions by HMGB1 in human cells. hydroxymethyltrioxsalen 103-106 high mobility group box 1 Homo sapiens 287-292 27911399-8 2016 We describe experimental techniques to prepare TFO-directed ICLs on reporter plasmids, and to interrogate the association of HMGB1 with the TFO-directed ICLs in a cellular context using chromatin immunoprecipitation assays. tfo 140-143 high mobility group box 1 Homo sapiens 125-130 26431816-0 2016 HMGB1 is responsible for Amitriptyline-mediated cardiac protection from ischemic-reperfusion injury. Amitriptyline 25-38 high mobility group box 1 Homo sapiens 0-5 27899980-0 2016 Treatment effects of oxaliplatin combined with gemcitabine on colorectal cancer and its influence on HMGB1 expression. Oxaliplatin 21-32 high mobility group box 1 Homo sapiens 101-106 27899980-0 2016 Treatment effects of oxaliplatin combined with gemcitabine on colorectal cancer and its influence on HMGB1 expression. gemcitabine 47-58 high mobility group box 1 Homo sapiens 101-106 27899980-1 2016 In the present study, we analyzed the supra-additive effect of oxaliplatin in combination with gemcitabine on terminal colorectal cancer and its influence on high-mobility group box 1 (HMGB1) expression. Oxaliplatin 63-74 high mobility group box 1 Homo sapiens 158-183 27899980-1 2016 In the present study, we analyzed the supra-additive effect of oxaliplatin in combination with gemcitabine on terminal colorectal cancer and its influence on high-mobility group box 1 (HMGB1) expression. Oxaliplatin 63-74 high mobility group box 1 Homo sapiens 185-190 27899980-12 2016 We conclude that the reduced expression rate of HMGB1 in terminal colorectal cancer cases was probably related to the effects of oxaliplatin combined with gemcitabine. Oxaliplatin 129-140 high mobility group box 1 Homo sapiens 48-53 27899980-12 2016 We conclude that the reduced expression rate of HMGB1 in terminal colorectal cancer cases was probably related to the effects of oxaliplatin combined with gemcitabine. gemcitabine 155-166 high mobility group box 1 Homo sapiens 48-53 27807322-8 2016 : Results: Compared with the control group, the cell activity was significantly decreased and the level of serum HMGB1 was significantly increased in the chemotherapeutic (VCR, VP-16, Ara-C, ADM and As2O3) groups (all P<0.05). Cytarabine 185-190 high mobility group box 1 Homo sapiens 114-119 27807322-8 2016 : Results: Compared with the control group, the cell activity was significantly decreased and the level of serum HMGB1 was significantly increased in the chemotherapeutic (VCR, VP-16, Ara-C, ADM and As2O3) groups (all P<0.05). Doxorubicin 192-195 high mobility group box 1 Homo sapiens 114-119 27807322-8 2016 : Results: Compared with the control group, the cell activity was significantly decreased and the level of serum HMGB1 was significantly increased in the chemotherapeutic (VCR, VP-16, Ara-C, ADM and As2O3) groups (all P<0.05). Arsenic Trioxide 200-205 high mobility group box 1 Homo sapiens 114-119 27899819-0 2016 4-cholesten-3-one suppresses lung adenocarcinoma metastasis by regulating translocation of HMGB1, HIF1alpha and Caveolin-1. cholest-4-en-3-one 0-17 high mobility group box 1 Homo sapiens 91-96 27704361-5 2016 Our results found that the risk of susceptibility to cervical invasive cancer was 1.85 (95 % CI 1.12-3.04; p = 0.016) in women with TC and 1.99 (95 % CI 1.24-3.23; p = 0.005) in women with TC/CC after adjusting for age, using TT as a comparison reference in HMGB1 SNP rs1412125. Technetium 132-134 high mobility group box 1 Homo sapiens 258-263 27704361-8 2016 In conclusion, Taiwanese women with TC or TC/CC in HMGB1 SNP rs1412125 as well as CG or CG/GG in rs2249825 were susceptible to the development of cervical invasive cancer. Technetium 36-38 high mobility group box 1 Homo sapiens 51-56 27704361-8 2016 In conclusion, Taiwanese women with TC or TC/CC in HMGB1 SNP rs1412125 as well as CG or CG/GG in rs2249825 were susceptible to the development of cervical invasive cancer. Technetium 42-44 high mobility group box 1 Homo sapiens 51-56 27451951-0 2016 DNA-HMGB1 interaction: The nuclear aggregates of polyamine mediation. Polyamines 49-58 high mobility group box 1 Homo sapiens 4-9 27383136-5 2016 Interestingly, with the increasing concentration of DDP, HMGB1 was gradually located into cytoplasm in cisplatin-sensitive A549 cells. Cisplatin 103-112 high mobility group box 1 Homo sapiens 57-62 27383136-6 2016 Moreover, interference with endogenous HMGB1 sensitized the effects of chemotherapeutic drugs, including 5-Fu, DDP, and OXA. Fluorouracil 105-109 high mobility group box 1 Homo sapiens 39-44 27473010-9 2016 CONCLUSION: MCZ may have preventive roles in the clinical management of IP in ELBWI by the suppression of IL-8 and HMGB-1. Miconazole 12-15 high mobility group box 1 Homo sapiens 115-121 27413111-0 2016 From the Cover: Tetrachlorobenzoquinone Exerts Neurological Proinflammatory Activity by Promoting HMGB1 Release, Which Induces TLR4 Clustering within the Lipid Raft. tetrachlorobenzoquinone 16-39 high mobility group box 1 Homo sapiens 98-103 27413111-5 2016 Indeed, this study demonstrated that TCBQ induces the secretion/translocation of HMGB1, which in turn activates its receptors, TLR family gene (especially TLR4) and receptor for advanced glycation end-products (RAGE) expressions. tetrachlorobenzoquinone 37-41 high mobility group box 1 Homo sapiens 81-86 27413111-9 2016 In summary, our current findings revealed a previously unknown mechanism of TCBQ-induced neurological inflammation related to HMGB1-TLR4 signaling. tetrachlorobenzoquinone 76-80 high mobility group box 1 Homo sapiens 126-131 27924158-0 2016 Lycorine Downregulates HMGB1 to Inhibit Autophagy and Enhances Bortezomib Activity in Multiple Myeloma. lycorine 0-8 high mobility group box 1 Homo sapiens 23-28 27924158-4 2016 We identified High mobility group box 1 (HMGB1), an important regulator of autophagy, as the most aberrantly expressed protein after lycorine treatment and as a critical mediator of lycorine activity. lycorine 133-141 high mobility group box 1 Homo sapiens 14-39 27924158-4 2016 We identified High mobility group box 1 (HMGB1), an important regulator of autophagy, as the most aberrantly expressed protein after lycorine treatment and as a critical mediator of lycorine activity. lycorine 133-141 high mobility group box 1 Homo sapiens 41-46 27924158-4 2016 We identified High mobility group box 1 (HMGB1), an important regulator of autophagy, as the most aberrantly expressed protein after lycorine treatment and as a critical mediator of lycorine activity. lycorine 182-190 high mobility group box 1 Homo sapiens 14-39 27924158-4 2016 We identified High mobility group box 1 (HMGB1), an important regulator of autophagy, as the most aberrantly expressed protein after lycorine treatment and as a critical mediator of lycorine activity. lycorine 182-190 high mobility group box 1 Homo sapiens 41-46 27924158-7 2016 Mechanistically, proteasomal degradation of HMGB1 by lycorine inhibits the activation of MEK-ERK thereby decreases phosphorylation of Bcl-2 resulting in constitutive association of Bcl-2 with Beclin-1. lycorine 53-61 high mobility group box 1 Homo sapiens 44-49 27924158-8 2016 In addition, we observed higher HMGB1 expression in bortezomib resistant cells and the combination of bortezomib plus lycorine was highly efficient in vitro and in vivo myeloma models as well as in re-sensitizing resistant cells to bortezomib. Bortezomib 52-62 high mobility group box 1 Homo sapiens 32-37 27924158-8 2016 In addition, we observed higher HMGB1 expression in bortezomib resistant cells and the combination of bortezomib plus lycorine was highly efficient in vitro and in vivo myeloma models as well as in re-sensitizing resistant cells to bortezomib. Bortezomib 102-112 high mobility group box 1 Homo sapiens 32-37 27924158-8 2016 In addition, we observed higher HMGB1 expression in bortezomib resistant cells and the combination of bortezomib plus lycorine was highly efficient in vitro and in vivo myeloma models as well as in re-sensitizing resistant cells to bortezomib. lycorine 118-126 high mobility group box 1 Homo sapiens 32-37 27924158-8 2016 In addition, we observed higher HMGB1 expression in bortezomib resistant cells and the combination of bortezomib plus lycorine was highly efficient in vitro and in vivo myeloma models as well as in re-sensitizing resistant cells to bortezomib. Bortezomib 102-112 high mobility group box 1 Homo sapiens 32-37 27568461-8 2016 HMGB1 levels were significantly decreased after sildenafil therapy for 6months (P<0.01). Sildenafil Citrate 48-58 high mobility group box 1 Homo sapiens 0-5 27568461-9 2016 CONCLUSIONS: Our study suggests that serum HMGB1 level may be used as a biomarker to identify PAH in CHD patients, assess pulmonary vascular remodeling, and evaluate the treatment response to sildenafil. Sildenafil Citrate 192-202 high mobility group box 1 Homo sapiens 43-48 27198223-0 2016 PKCalpha and HMGB1 antagonistically control hydrogen peroxide-induced poly-ADP-ribose formation. Hydrogen Peroxide 44-61 high mobility group box 1 Homo sapiens 13-18 27198223-0 2016 PKCalpha and HMGB1 antagonistically control hydrogen peroxide-induced poly-ADP-ribose formation. poly-adp 70-78 high mobility group box 1 Homo sapiens 13-18 27198223-8 2016 Together, these results identify PKCalpha and HMGB1 as important co-regulators involved in H2O2-induced PAR formation, a finding that may have important relevance for oxidative stress-associated pathophysiological conditions. Hydrogen Peroxide 91-95 high mobility group box 1 Homo sapiens 46-51 27512095-6 2016 Results demonstrated that either IL-1beta or HMGB1 promoted the release of the inflammatory cytokines such as prostaglandin E2 (PGE2), TNF-alpha, IL-6 and IL-8 in human IVD cells. Dinoprostone 110-126 high mobility group box 1 Homo sapiens 45-50 27512095-6 2016 Results demonstrated that either IL-1beta or HMGB1 promoted the release of the inflammatory cytokines such as prostaglandin E2 (PGE2), TNF-alpha, IL-6 and IL-8 in human IVD cells. Dinoprostone 128-132 high mobility group box 1 Homo sapiens 45-50 27725858-5 2016 The targeting effect of HMGB1 by miR-218 was measured with luciferase reporter assay. mir-218 33-40 high mobility group box 1 Homo sapiens 24-29 27899819-8 2016 4-cholesten-3-one-induced autophagy facilitated the release of HMGB1 from nuclei to cytoplasm, blocking nuclear translocation of HIF1alpha and activation of MMP2 and MMP9. cholest-4-en-3-one 0-17 high mobility group box 1 Homo sapiens 63-68 27285673-5 2016 The aim of this study was to investigate whether the expression of HMGB1differed between early onset and late onset preeclampsia or severe and mild preeclampsia and whether its expression correlated with the levels of uric acid. Uric Acid 218-227 high mobility group box 1 Homo sapiens 67-72 27093475-7 2016 Moreover, CS extract induced cellular senescence in cultured human airway epithelial cells, represented by induced senescence-associated beta-galactosidase activity, inhibited cell proliferation, increased p21 expression, and increased release of high-mobility group box 1 and IL-6. Cesium 10-12 high mobility group box 1 Homo sapiens 247-272 27457217-3 2016 We showed recently that an epirubicin/docetaxel therapy is associated with an increase in the cell death marker high-mobility group box 1 protein (HMGB1) in the circulation. Epirubicin 27-37 high mobility group box 1 Homo sapiens 112-137 27457217-3 2016 We showed recently that an epirubicin/docetaxel therapy is associated with an increase in the cell death marker high-mobility group box 1 protein (HMGB1) in the circulation. Epirubicin 27-37 high mobility group box 1 Homo sapiens 147-152 27457217-3 2016 We showed recently that an epirubicin/docetaxel therapy is associated with an increase in the cell death marker high-mobility group box 1 protein (HMGB1) in the circulation. Docetaxel 38-47 high mobility group box 1 Homo sapiens 112-137 27457217-3 2016 We showed recently that an epirubicin/docetaxel therapy is associated with an increase in the cell death marker high-mobility group box 1 protein (HMGB1) in the circulation. Docetaxel 38-47 high mobility group box 1 Homo sapiens 147-152 27285673-8 2016 The expression of HMGB1 in preeclamptic placentae or in first trimester and term placentae that had been treated with uric acid was measured. Uric Acid 118-127 high mobility group box 1 Homo sapiens 18-23 27285673-10 2016 The circulating levels of uric acid were significantly increased in preeclampsia and correlated with the expression of HMGB1. Uric Acid 26-35 high mobility group box 1 Homo sapiens 119-124 27602152-11 2016 The results revealed that the oncogenic cluster miRs 221/222 were more highly expressed by the most undifferentiated cell line [SK-N-BE(2)] compared with the the less tumorigenic cell line (SH-SY5Y) and that exogenous HMGB1 increases this expression. sk-n-be 128-135 high mobility group box 1 Homo sapiens 218-223 27522665-0 2016 Methotrexate affects HMGB1 expression in rheumatoid arthritis, and the downregulation of HMGB1 prevents rheumatoid arthritis progression. Methotrexate 0-12 high mobility group box 1 Homo sapiens 21-26 27522665-2 2016 Recent studies have shown that methotrexate (MTX) may inhibit the expression of HMGB1. Methotrexate 31-43 high mobility group box 1 Homo sapiens 80-85 27522665-2 2016 Recent studies have shown that methotrexate (MTX) may inhibit the expression of HMGB1. Methotrexate 45-48 high mobility group box 1 Homo sapiens 80-85 27522665-3 2016 This study examined whether HMGB1 might be involved in the treatment of RA using MTX. Methotrexate 81-84 high mobility group box 1 Homo sapiens 28-33 27522665-10 2016 At the tissue level, HMGB1 expression in synovial membrane did not differ significantly between the OA and RA-MTX groups, but was significantly lower in these groups than in the RA-noMTX group. Methotrexate 110-113 high mobility group box 1 Homo sapiens 21-26 27522665-13 2016 These results showed that MTX reduced HMGB1 expression in RA synovial tissues, and through the downregulation of HMGB1 expression in tissues, MTX may slow disease progression of RA. Methotrexate 26-29 high mobility group box 1 Homo sapiens 38-43 27522665-13 2016 These results showed that MTX reduced HMGB1 expression in RA synovial tissues, and through the downregulation of HMGB1 expression in tissues, MTX may slow disease progression of RA. Methotrexate 142-145 high mobility group box 1 Homo sapiens 113-118 27602152-12 2016 In addition, HMGB1 modulates PTEN expression via miR-221/222, as demonstrated by transiently blocking miR-221/222 with anti-sense oligonucleotides. Oligonucleotides 130-146 high mobility group box 1 Homo sapiens 13-18 27682500-0 2016 [The wle of high mobility group protein B1(HMGB-1) on inflammatony responese and pulmonary fibrosis induced by silica dusty]. Silicon Dioxide 111-117 high mobility group box 1 Homo sapiens 12-42 27378565-7 2016 Noteworthy, salicylic acid, a metabolite of aspirin, has been recently found to inhibit HMGB1. Salicylic Acid 12-26 high mobility group box 1 Homo sapiens 88-93 27378565-7 2016 Noteworthy, salicylic acid, a metabolite of aspirin, has been recently found to inhibit HMGB1. Aspirin 44-51 high mobility group box 1 Homo sapiens 88-93 27378565-9 2016 Notably, high levels of serum HMGB1, in particular of the hyper-acetylated and disulfide isoforms, are sensitive disease biomarkers and are associated with different disease stages. Disulfides 79-88 high mobility group box 1 Homo sapiens 30-35 27544687-6 2016 We also found that both glycyrrhizin-mediated inhibition of HMGB1 and intracerebroventricular neutralizing antibody treatments before middle cerebral artery occlusion onset diminish infarct volume. Glycyrrhizic Acid 24-36 high mobility group box 1 Homo sapiens 60-65 27622063-8 2016 We show that both platinum drugs triggered translocation of calreticulin and HSP70, as well as the release of ATP and HMGB1. Platinum 18-26 high mobility group box 1 Homo sapiens 118-123 26888251-9 2016 HMGB1 was significantly increased in RECs and Muller cells grown in high-glucose culture conditions, which was subsequently reduced with Compound 49b treatment. Glucose 73-80 high mobility group box 1 Homo sapiens 0-5 26888251-10 2016 Our findings suggest that high glucose may increase HMGB1 levels that lead to increased TLR4 signaling. Glucose 31-38 high mobility group box 1 Homo sapiens 52-57 27553758-10 2016 Direct in vitro BBB integrity assessment further revealed a significant and concentration-dependent increase in paracellular permeability to dextrans by HMGB1 or alpha-thrombin, possibly through disruption of zona occludins-1 bands. Dextrans 141-149 high mobility group box 1 Homo sapiens 153-158 27682500-0 2016 [The wle of high mobility group protein B1(HMGB-1) on inflammatony responese and pulmonary fibrosis induced by silica dusty]. Silicon Dioxide 111-117 high mobility group box 1 Homo sapiens 43-49 26546554-9 2016 It was observed that aluminum potentially enhanced protein levels of PERK, EIF2alpha, caspase 9, caspase 3, and inflammatory markers like NF-kappaB, NLRP3, HMGB1, and nitric oxide (NO). Aluminum 21-29 high mobility group box 1 Homo sapiens 156-161 27434276-12 2016 Ex vivo, recombinant-HMGB1 potentiated cytokine release (e.g. TNF-alpha) when combined with lipoarabinomannan. lipoarabinomannan 92-109 high mobility group box 1 Homo sapiens 21-26 26845344-8 2016 The functional consequence of HMGB1 depletion in HCT116 and INS1 cells was reduced insulin and TCF7L2 mRNA expression, TCF7L2 transcriptional activity and glucose stimulated insulin secretion. Glucose 155-162 high mobility group box 1 Homo sapiens 30-35 27474498-7 2016 administration of the anti-HMGB1-neutralizing antibody or recombinant human soluble thrombomodulin (rhsTM), known to inactivate HMGB1, prevented the development of hyperalgesia and/or allodynia induced by paclitaxel or vincristine in rats. Paclitaxel 205-215 high mobility group box 1 Homo sapiens 27-32 27474498-7 2016 administration of the anti-HMGB1-neutralizing antibody or recombinant human soluble thrombomodulin (rhsTM), known to inactivate HMGB1, prevented the development of hyperalgesia and/or allodynia induced by paclitaxel or vincristine in rats. Paclitaxel 205-215 high mobility group box 1 Homo sapiens 128-133 27474498-7 2016 administration of the anti-HMGB1-neutralizing antibody or recombinant human soluble thrombomodulin (rhsTM), known to inactivate HMGB1, prevented the development of hyperalgesia and/or allodynia induced by paclitaxel or vincristine in rats. Vincristine 219-230 high mobility group box 1 Homo sapiens 27-32 27174706-5 2016 In addition, we evaluated the expression of the high mobility group box-1 protein (HMGB1), a chromatin remodelling protein involved in DSBs repair and obesity. dsbs 135-139 high mobility group box 1 Homo sapiens 48-73 26482581-2 2016 Recent findings suggest that the redox state of HMGB1 is a critical molecular feature of HMGB1 such that the reduced form (fr-HMGB1) is chemotactic, while the disulfide form (ds-HMGB1) is pro-inflammatory. Disulfides 159-168 high mobility group box 1 Homo sapiens 48-53 26482581-2 2016 Recent findings suggest that the redox state of HMGB1 is a critical molecular feature of HMGB1 such that the reduced form (fr-HMGB1) is chemotactic, while the disulfide form (ds-HMGB1) is pro-inflammatory. Disulfides 159-168 high mobility group box 1 Homo sapiens 89-94 26482581-2 2016 Recent findings suggest that the redox state of HMGB1 is a critical molecular feature of HMGB1 such that the reduced form (fr-HMGB1) is chemotactic, while the disulfide form (ds-HMGB1) is pro-inflammatory. Disulfides 159-168 high mobility group box 1 Homo sapiens 89-94 26482581-2 2016 Recent findings suggest that the redox state of HMGB1 is a critical molecular feature of HMGB1 such that the reduced form (fr-HMGB1) is chemotactic, while the disulfide form (ds-HMGB1) is pro-inflammatory. Disulfides 159-168 high mobility group box 1 Homo sapiens 89-94 26482581-16 2016 Consistent with prior reports, the present findings demonstrate that the disulfide form of HMGB1 not only potentiates the neuroinflammatory response to a subsequent immune challenge in vivo, but also potentiates the sickness response to that challenge. Disulfides 73-82 high mobility group box 1 Homo sapiens 91-96 26482581-19 2016 Taken together, the present results suggest that the redox state of HMGB1 is a critical determinant of the priming properties of HMGB1 such that the disulfide form of HMGB1 induces a primed immunophenotype in the CNS, which may result in an exacerbated neuroinflammatory response upon exposure to a subsequent pro-inflammatory stimulus. Disulfides 149-158 high mobility group box 1 Homo sapiens 68-73 26482581-19 2016 Taken together, the present results suggest that the redox state of HMGB1 is a critical determinant of the priming properties of HMGB1 such that the disulfide form of HMGB1 induces a primed immunophenotype in the CNS, which may result in an exacerbated neuroinflammatory response upon exposure to a subsequent pro-inflammatory stimulus. Disulfides 149-158 high mobility group box 1 Homo sapiens 129-134 26482581-19 2016 Taken together, the present results suggest that the redox state of HMGB1 is a critical determinant of the priming properties of HMGB1 such that the disulfide form of HMGB1 induces a primed immunophenotype in the CNS, which may result in an exacerbated neuroinflammatory response upon exposure to a subsequent pro-inflammatory stimulus. Disulfides 149-158 high mobility group box 1 Homo sapiens 129-134 26896489-4 2016 High mobility group box 1 (HMGB1) protein is considered to be involved in the removal of the lesions as it binds with high affinity to cisplatin/DNA adducts. Cisplatin 135-144 high mobility group box 1 Homo sapiens 0-25 26896489-4 2016 High mobility group box 1 (HMGB1) protein is considered to be involved in the removal of the lesions as it binds with high affinity to cisplatin/DNA adducts. Cisplatin 135-144 high mobility group box 1 Homo sapiens 27-32 26896489-8 2016 H1299 cells overexpressing HMGB1 were significantly sensitized to treatment with cisplatin demonstrating the close relation between the role of HMGB1 in repair of cis-platinated DNA and the efficiency of the anticancer drug, the process being modulated by the C-terminus. Cisplatin 81-90 high mobility group box 1 Homo sapiens 27-32 26896489-8 2016 H1299 cells overexpressing HMGB1 were significantly sensitized to treatment with cisplatin demonstrating the close relation between the role of HMGB1 in repair of cis-platinated DNA and the efficiency of the anticancer drug, the process being modulated by the C-terminus. Cisplatin 81-90 high mobility group box 1 Homo sapiens 144-149 26896489-9 2016 In A549 cells with functional p53, the repair of cisplatin/DNA adducts is determined by a complex action of HMGB1 and p53 as an increase of DNA repair capacity was registered only after silencing of both proteins. Cisplatin 49-58 high mobility group box 1 Homo sapiens 108-113 27174706-5 2016 In addition, we evaluated the expression of the high mobility group box-1 protein (HMGB1), a chromatin remodelling protein involved in DSBs repair and obesity. dsbs 135-139 high mobility group box 1 Homo sapiens 83-88 27174706-7 2016 Concordantly, 1) HMGB1 levels of obese individuals increased and decreased at 2h- or 4 h-post mutagen treatment, respectively, and 2) the opposite occurred for the normal weight adolescents where the protein was down-expressed at 2h and over-expressed at 4h. Deuterium 78-80 high mobility group box 1 Homo sapiens 17-22 26952810-4 2016 MIF-induced ROS led to ERK phosphorylation, which facilitated HMGB1 release from the nucleus to the cytoplasm. ros 12-15 high mobility group box 1 Homo sapiens 62-67 27446084-9 2016 Interestingly, exposure of the cells to HMGB1 in inflammatory conditions increased synergistically Amigo2 expression and significantly reduced Cd-mediated cellular toxicity. Cadmium 143-145 high mobility group box 1 Homo sapiens 40-45 26733616-0 2016 HMGB1 and Its Hyperacetylated Isoform are Sensitive and Specific Serum Biomarkers to Detect Asbestos Exposure and to Identify Mesothelioma Patients. Asbestos 92-100 high mobility group box 1 Homo sapiens 0-5 26733616-1 2016 PURPOSE: To determine whether serum levels of high mobility group box protein 1 (HMGB1) could differentiate malignant mesothelioma patients, asbestos-exposed individuals, and unexposed controls. Asbestos 141-149 high mobility group box 1 Homo sapiens 81-86 26733616-4 2016 RESULTS: HMGB1 serum levels reliably distinguished malignant mesothelioma patients, asbestos-exposed individuals, and unexposed controls. Asbestos 84-92 high mobility group box 1 Homo sapiens 9-14 26733616-5 2016 Total HMGB1 was significantly higher in malignant mesothelioma patients and asbestos-exposed individuals compared with healthy controls. Asbestos 76-84 high mobility group box 1 Homo sapiens 6-11 26733616-6 2016 Hyperacetylated HMGB1 was significantly higher in malignant mesothelioma patients compared with asbestos-exposed individuals and healthy controls, and did not vary with tumor stage. Asbestos 96-104 high mobility group box 1 Homo sapiens 16-21 26733616-7 2016 At the cut-off value of 2.00 ng/mL, the sensitivity and specificity of serum hyperacetylated HMGB1 in differentiating malignant mesothelioma patients from asbestos-exposed individuals and healthy controls was 100%, outperforming other previously proposed biomarkers. Asbestos 155-163 high mobility group box 1 Homo sapiens 93-98 26733616-9 2016 CONCLUSIONS: Our results are significant and clinically relevant as they provide the first biomarker of asbestos exposure and indicate that hyperacetylated HMGB1 is an accurate biomarker to differentiate malignant mesothelioma patients from individuals occupationally exposed to asbestos and unexposed controls. Asbestos 104-112 high mobility group box 1 Homo sapiens 156-161 26733616-9 2016 CONCLUSIONS: Our results are significant and clinically relevant as they provide the first biomarker of asbestos exposure and indicate that hyperacetylated HMGB1 is an accurate biomarker to differentiate malignant mesothelioma patients from individuals occupationally exposed to asbestos and unexposed controls. Asbestos 279-287 high mobility group box 1 Homo sapiens 156-161 26921471-0 2016 In-vivo evidence that high mobility group box 1 exerts deleterious effects in the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine model and Parkinson"s disease which can be attenuated by glycyrrhizin. 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine 82-126 high mobility group box 1 Homo sapiens 22-47 26921471-0 2016 In-vivo evidence that high mobility group box 1 exerts deleterious effects in the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine model and Parkinson"s disease which can be attenuated by glycyrrhizin. Glycyrrhizic Acid 184-196 high mobility group box 1 Homo sapiens 22-47 27348219-5 2016 Our results showed that HMGB1 levels are increased in patients with CCHFV, DOBV or PUUV infection. dobv 75-79 high mobility group box 1 Homo sapiens 24-29 27348219-7 2016 Our results indicate that HMGB1 could be a useful prognostic biomarker for disease severity in PUUV and CCHFV infection, where the difference between the mild and severe patients group was highly significant. puuv 95-99 high mobility group box 1 Homo sapiens 26-31 27348219-8 2016 Even in patients with severe DOBV infection concentrations of HMGB1 were 2.8-times higher than in the mild group, but the difference was not statistically significant. dobv 29-33 high mobility group box 1 Homo sapiens 62-67 26873936-5 2016 Aspirin consumption significantly blocked thrombin- and collagen-induced increases in exosome cargo levels of chemokines and HMGB1, without altering total exosome secretion or GPVI cargo. Aspirin 0-7 high mobility group box 1 Homo sapiens 125-130 26923673-0 2016 Structural aspects of the interaction of anticancer drug Actinomycin-D to the GC rich region of hmgb1 gene. Dactinomycin 57-70 high mobility group box 1 Homo sapiens 96-101 26923673-5 2016 The molecular interaction of actinomycin (ACT) with the regulatory region of hmgb1 gene was characterized by spectroscopic, calorimetric and molecular docking studies. Dactinomycin 29-40 high mobility group box 1 Homo sapiens 77-82 26923673-6 2016 The hypochromic and bathochromic shift in the absorption spectrum, stabilization of 25RY duplex against thermal denaturation, perturbation of CD spectrum of duplex and enhancement of fluorescence intensity of actinomycin indicate strong binding of actinomycin to the hmgb1 promoter region (25RY).The energetics was characterized to be endothermic and entropy driven. Dactinomycin 209-220 high mobility group box 1 Homo sapiens 267-272 26923673-6 2016 The hypochromic and bathochromic shift in the absorption spectrum, stabilization of 25RY duplex against thermal denaturation, perturbation of CD spectrum of duplex and enhancement of fluorescence intensity of actinomycin indicate strong binding of actinomycin to the hmgb1 promoter region (25RY).The energetics was characterized to be endothermic and entropy driven. Dactinomycin 248-259 high mobility group box 1 Homo sapiens 267-272 26956470-0 2016 Propofol Protects Rats and Human Alveolar Epithelial Cells Against Lipopolysaccharide-Induced Acute Lung Injury via Inhibiting HMGB1 Expression. Propofol 0-8 high mobility group box 1 Homo sapiens 127-132 26956470-10 2016 Propofol protects rats and human alveolar epithelial cells against HMGB1 expression in a rat model of LPS-induced ALI. Propofol 0-8 high mobility group box 1 Homo sapiens 67-72 27247778-7 2016 Plasma levels of HMGB-1 on day 1 correlated with prognostic parameters (estimated interval to return of spontaneous circulation, lactate, and NH3), tissue damage, systemic inflammation, and disease severity. Lactic Acid 129-136 high mobility group box 1 Homo sapiens 17-23 27247709-2 2016 We show that HMGB1 alters the structure and stability of the canonical nucleosome (N) in a nonenzymatic, adenosine triphosphate-independent manner. Adenosine Triphosphate 105-127 high mobility group box 1 Homo sapiens 13-18 27009165-7 2016 In particular, doxorubicin triggers an HMGB1 autocrine/paracrine loop with its receptor, TLR4, which is also upregulated in DX-LCLs and is responsible for NF-kappaB activation and a delayed apoptosis that allows a prolonged stimulation of EBV-specific T-cell precursors. Doxorubicin 15-26 high mobility group box 1 Homo sapiens 39-44 26873936-7 2016 The plasma platelet-derived exosome number is lower and its chemokine and HMGB1 levels higher after age 65 yr. Aspirin consumption significantly suppressed cargo protein levels of plasma platelet-derived exosomes without altering total levels of exosomes. Aspirin 111-118 high mobility group box 1 Homo sapiens 74-79 27035254-7 2016 HMGB1 increased FITC-dextran permeability, but suppressed epithelial resistance in a dose- and time-dependent manner. fluorescein isothiocyanate dextran 16-28 high mobility group box 1 Homo sapiens 0-5 27035254-11 2016 Anti-RAGE antibody and the ERK1/2 inhibitor, U0126, attenuated the HMGB1-mediated changes in barrier permeability, restored the expression levels of occludin and claudin-1 and pevented the redistribution of E-cadherin and beta-catenin. U 0126 45-50 high mobility group box 1 Homo sapiens 67-72 25787746-5 2016 RESULTS: Studies from our laboratory employing reverse transcription polymerase chain reaction (RT-PCR) to assess mRNA, immunohistochemistry and Western blot analysis to assess protein expression, and others suggest that ethanol increases brain neuroimmune gene and protein expression through two distinct mechanisms involving (1) systemic induction of innate immune molecules that are transported from blood to the brain and (2) the direct release of high-mobility group box 1 (HMGB1) from neurons in the brain. Ethanol 221-228 high mobility group box 1 Homo sapiens 452-477 25787746-5 2016 RESULTS: Studies from our laboratory employing reverse transcription polymerase chain reaction (RT-PCR) to assess mRNA, immunohistochemistry and Western blot analysis to assess protein expression, and others suggest that ethanol increases brain neuroimmune gene and protein expression through two distinct mechanisms involving (1) systemic induction of innate immune molecules that are transported from blood to the brain and (2) the direct release of high-mobility group box 1 (HMGB1) from neurons in the brain. Ethanol 221-228 high mobility group box 1 Homo sapiens 479-484 25787746-8 2016 Expression of HMGB1, TLRs, and other ISMs is increased several-fold in the human orbital frontal cortex, and expression of these molecules is highly correlated with each other as well as lifetime alcohol consumption and age of drinking onset. Alcohols 196-203 high mobility group box 1 Homo sapiens 14-19 25787746-9 2016 CONCLUSIONS: The persistent and cumulative nature of alcohol on HMGB1 and TLR gene induction support their involvement in alcohol-induced long-term changes in brain function and neurodegeneration. Alcohols 53-60 high mobility group box 1 Homo sapiens 64-69 25787746-9 2016 CONCLUSIONS: The persistent and cumulative nature of alcohol on HMGB1 and TLR gene induction support their involvement in alcohol-induced long-term changes in brain function and neurodegeneration. Alcohols 122-129 high mobility group box 1 Homo sapiens 64-69 28773427-4 2016 In this study, Vascular Endothelial Growth Factor (VEGF) and High Mobility Group Box 1 (HMGB1) were used for incorporation into poly-epsilon-caprolactone (PCL)-coated porous titanium implants. polycaprolactone 128-153 high mobility group box 1 Homo sapiens 61-86 28773427-4 2016 In this study, Vascular Endothelial Growth Factor (VEGF) and High Mobility Group Box 1 (HMGB1) were used for incorporation into poly-epsilon-caprolactone (PCL)-coated porous titanium implants. polycaprolactone 128-153 high mobility group box 1 Homo sapiens 88-93 28773427-4 2016 In this study, Vascular Endothelial Growth Factor (VEGF) and High Mobility Group Box 1 (HMGB1) were used for incorporation into poly-epsilon-caprolactone (PCL)-coated porous titanium implants. polycaprolactone 155-158 high mobility group box 1 Homo sapiens 61-86 28773427-4 2016 In this study, Vascular Endothelial Growth Factor (VEGF) and High Mobility Group Box 1 (HMGB1) were used for incorporation into poly-epsilon-caprolactone (PCL)-coated porous titanium implants. polycaprolactone 155-158 high mobility group box 1 Homo sapiens 88-93 26592934-0 2016 Anti-septic effects of dabrafenib on HMGB1-mediated inflammatory responses. dabrafenib 23-33 high mobility group box 1 Homo sapiens 37-42 26481429-3 2016 The extracellular function of HMGB1 is dependent upon redox modification of cysteine residues that control chemoattractant and cytokine-inducing properties. Cysteine 76-84 high mobility group box 1 Homo sapiens 30-35 26481429-5 2016 RECENT ADVANCES: HMGB1 isoforms have been shown to be more sensitive biomarkers than ALT for predicting DILI development and the requirement for liver transplant following acetaminophen (APAP) overdose. Acetaminophen 172-185 high mobility group box 1 Homo sapiens 17-22 26481429-5 2016 RECENT ADVANCES: HMGB1 isoforms have been shown to be more sensitive biomarkers than ALT for predicting DILI development and the requirement for liver transplant following acetaminophen (APAP) overdose. Acetaminophen 187-191 high mobility group box 1 Homo sapiens 17-22 26715031-6 2016 The redox states of the cysteines in positions 23, 45, and 106 determine the biological activity of the extracellular HMGB1. Cysteine 24-33 high mobility group box 1 Homo sapiens 118-123 25532033-7 2016 Monomethylation of lysine 43 (K43), a proposed neutrophil-specific PTM, was strongly associated with high HMGB1 levels, implying that neutrophils are a source of released HMGB1. Lysine 19-25 high mobility group box 1 Homo sapiens 106-111 25532033-7 2016 Monomethylation of lysine 43 (K43), a proposed neutrophil-specific PTM, was strongly associated with high HMGB1 levels, implying that neutrophils are a source of released HMGB1. Lysine 19-25 high mobility group box 1 Homo sapiens 171-176 26592934-4 2016 Here, we examined the effects of dabrafenib (DAB) on the modulation of HMGB1-mediated septic responses. dabrafenib 33-43 high mobility group box 1 Homo sapiens 71-76 26592934-4 2016 Here, we examined the effects of dabrafenib (DAB) on the modulation of HMGB1-mediated septic responses. dabrafenib 45-48 high mobility group box 1 Homo sapiens 71-76 26592934-5 2016 DAB inhibited the release of HMGB1 and downregulated HMGB1-dependent inflammatory responses by enhancing the expressions of cell adhesion molecules (CAMs) in human endothelial cells. dabrafenib 0-3 high mobility group box 1 Homo sapiens 29-34 26592934-5 2016 DAB inhibited the release of HMGB1 and downregulated HMGB1-dependent inflammatory responses by enhancing the expressions of cell adhesion molecules (CAMs) in human endothelial cells. dabrafenib 0-3 high mobility group box 1 Homo sapiens 53-58 26592934-6 2016 In addition, treatment with DAB inhibited the HMGB1 secretion by CLP and sepsis-related mortality and pulmonary injury. dabrafenib 28-31 high mobility group box 1 Homo sapiens 46-51 26592934-7 2016 This study demonstrated that DAB could be alternative therapeutic options for sepsis or septic shock via the inhibition of the HMGB1 signaling pathway. dabrafenib 29-32 high mobility group box 1 Homo sapiens 127-132 27026438-11 2016 CNT-L tended to induce stronger cytotoxicity and 8-nitroG formation than CNT-S. Endocytosis inhibitors, TLR9 siRNA and antibodies against HMGB1 and RAGE largely reduced MWCNT-induced 8-nitroG formation. 8-nitroguanine 183-191 high mobility group box 1 Homo sapiens 138-143 27026438-0 2016 Multi-walled carbon nanotube induces nitrative DNA damage in human lung epithelial cells via HMGB1-RAGE interaction and Toll-like receptor 9 activation. Carbon 13-19 high mobility group box 1 Homo sapiens 93-98 27026438-8 2016 The release of HMGB1 and double-stranded DNA (dsDNA) into the culture supernatant from MWCNT-treated cells was examined by ELISA and fluorometric analysis, respectively. mwcnt 87-92 high mobility group box 1 Homo sapiens 15-20 27026438-16 2016 The HMGB1-DNA complex binds to RAGE on neighboring cells and then CpG DNA is recognized by TLR9 in lysosomes, leading to generation of nitric oxide and 8-nitroG formation. Nitric Oxide 135-147 high mobility group box 1 Homo sapiens 4-9 27026438-16 2016 The HMGB1-DNA complex binds to RAGE on neighboring cells and then CpG DNA is recognized by TLR9 in lysosomes, leading to generation of nitric oxide and 8-nitroG formation. 8-nitroguanine 152-160 high mobility group box 1 Homo sapiens 4-9 26962684-5 2016 When injected into established cancers, LTX-315 caused a transiently hemorrhagic focal necrosis that was accompanied by massive release of HMGB1 (from close-to-all cancer cells), as well as caspase-3 activation in a fraction of the cells. LTX-315 40-47 high mobility group box 1 Homo sapiens 139-144 27033788-7 2016 According to the results of MTT, the growth of HEC-1A cells was obviously inhibited after transfection of HEC-1A HMGB1 shRNA (P<0.05). monooxyethylene trimethylolpropane tristearate 28-31 high mobility group box 1 Homo sapiens 113-118 26346170-11 2016 In summary, our results demonstrate that 14-3-3sigma induces cisplatin resistance in ESCC cells and that 14-3-3sigma-mediated cisplatin resistance involves DNA repair molecules HMGB1 and XPA. Cisplatin 126-135 high mobility group box 1 Homo sapiens 177-182 26948869-3 2016 HMGB1 allosterically inhibits the tetrameric pyruvate kinase isoform M2, thus blocking glucose-driven aerobic respiration. Glucose 87-94 high mobility group box 1 Homo sapiens 0-5 26760176-2 2016 OBJECTIVES: To verify high-mobility group box 1 protein (HMGB1) levels in CF patients according to glucose tolerance state, and analyze relationships with insulin secretion and resistance. Glucose 99-106 high mobility group box 1 Homo sapiens 22-47 26760176-2 2016 OBJECTIVES: To verify high-mobility group box 1 protein (HMGB1) levels in CF patients according to glucose tolerance state, and analyze relationships with insulin secretion and resistance. Glucose 99-106 high mobility group box 1 Homo sapiens 57-62 26899177-0 2016 EGCG in Green Tea Induces Aggregation of HMGB1 Protein through Large Conformational Changes with Polarized Charge Redistribution. epigallocatechin gallate 0-4 high mobility group box 1 Homo sapiens 41-46 26899177-2 2016 Recent experimental evidences indicate that EGCG can induce the aggregation of HMGB1 protein, a late mediator of inflammation, which subsequently stimulates the autophagic degradation and thus provides protection from lethal endotoxemia and sepsis. epigallocatechin gallate 44-48 high mobility group box 1 Homo sapiens 79-84 26899177-3 2016 In this study, we use molecular dynamics (MD) simulations to explore the underlying molecular mechanism of this aggregation of HMGB1 facilitated by EGCG. epigallocatechin gallate 148-152 high mobility group box 1 Homo sapiens 127-132 26899177-4 2016 Our simulation results reveal that EGCG firmly binds to HMGB1 near Cys106, which supports previous preliminary experimental evidence. epigallocatechin gallate 35-39 high mobility group box 1 Homo sapiens 56-61 26899177-5 2016 A large HMGB1 conformational change is observed, where Box A and Box B, two homogenous domains of HMGB1, are repositioned and packed together by EGCG. epigallocatechin gallate 145-149 high mobility group box 1 Homo sapiens 8-13 26899177-5 2016 A large HMGB1 conformational change is observed, where Box A and Box B, two homogenous domains of HMGB1, are repositioned and packed together by EGCG. epigallocatechin gallate 145-149 high mobility group box 1 Homo sapiens 98-103 26899177-7 2016 We suggest that the highly polarized charge distribution leads to the aggregation of HMGB1, which differs from the previous hypothesis that two HMGB1 monomers are linked by the dimer of EGCG. epigallocatechin gallate 186-190 high mobility group box 1 Homo sapiens 85-90 26578599-4 2016 Here, we demonstrate that HMGB1 recognizes TFO-ICLs in human cells, and its depletion increases ICL-induced mutagenesis in human cells without altering the mutation spectra. tfo 43-46 high mobility group box 1 Homo sapiens 26-31 26453003-0 2016 Ketamine attenuates high mobility group box-1-induced inflammatory responses in endothelial cells. Ketamine 0-8 high mobility group box 1 Homo sapiens 20-45 26453003-3 2016 However, the effects of ketamine on HMGB1-mediated proinflammatory responses have not been fully investigated. Ketamine 24-32 high mobility group box 1 Homo sapiens 36-41 26453003-4 2016 In the present study, we investigated the effects of ketamine on HMGB1-activated endothelial cells and explored the underlying mechanisms. Ketamine 53-61 high mobility group box 1 Homo sapiens 65-70 26453003-6 2016 The anti-inflammatory activities of ketamine were determined by measuring solute flux, leukocyte adhesion and migration, and activation of proinflammatory proteins in HMGB1-activated endothelial cells. Ketamine 36-44 high mobility group box 1 Homo sapiens 167-172 26872033-6 2016 In preceding immunohistochemistry experiments using paraffin-block sections from endometriosis (N = 33) and control (N = 27) group, retrospectively, HMGB-1 expression was shown in both epithelial and stromal cell. Paraffin 52-60 high mobility group box 1 Homo sapiens 149-155 26453003-8 2016 RESULTS: We found that ketamine inhibited the HMGB1-mediated barrier disruption, neutrophil adhesion and migration, and expression of cell adhesion molecules in a dose-dependent manner. Ketamine 23-31 high mobility group box 1 Homo sapiens 46-51 26453003-9 2016 Furthermore, ketamine downregulated the TLR-2 and -4, expression in HMGB1-activated endothelial cells. Ketamine 13-21 high mobility group box 1 Homo sapiens 68-73 26453003-12 2016 CONCLUSIONS: Our study has demonstrated that ketamine exerts anti-inflammatory effects in HMGB1-mediated proinflammatory responses in a dose-dependent manner. Ketamine 45-53 high mobility group box 1 Homo sapiens 90-95 26346170-9 2016 14-3-3sigma, HMGB1, and XPA were preferentially expressed in cisplatin-resistant SLMT-1 cells when compared to those more sensitive to cisplatin. Cisplatin 61-70 high mobility group box 1 Homo sapiens 13-18 26346170-10 2016 In ESCC patients with poor response to cisplatin-based chemoradiation, their pre-treatment tumors expressed higher expression of HMGB1 than those with response to such treatment. Cisplatin 39-48 high mobility group box 1 Homo sapiens 129-134 26739898-8 2016 Treatment with PI3K inhibitor (LY294006) and beta-catenin shRNA reversed HMGB1-induced EMT. ly294006 31-39 high mobility group box 1 Homo sapiens 73-78 26848281-1 2016 BACKGROUND: Conditioning regimens including total body irradiation (TBI) or cyclophosphamide can mobilize high-mobility group box 1 (HMGB1) to peripheral blood. Cyclophosphamide 76-92 high mobility group box 1 Homo sapiens 106-131 26848281-1 2016 BACKGROUND: Conditioning regimens including total body irradiation (TBI) or cyclophosphamide can mobilize high-mobility group box 1 (HMGB1) to peripheral blood. Cyclophosphamide 76-92 high mobility group box 1 Homo sapiens 133-138 26848281-7 2016 Additionally, levels of HMGB1 at day 0 were significantly increased in TBI+ patients and cyclophosphamide/TBI patients. Cyclophosphamide 89-105 high mobility group box 1 Homo sapiens 24-29 26382001-7 2016 In addition, the MTT assay, growth curve, and the colony forming assay confirmed the promotion by HMGB1 to the proliferation of CNE-2 cells, depending on RAGE. monooxyethylene trimethylolpropane tristearate 17-20 high mobility group box 1 Homo sapiens 98-103 27627916-0 2016 Inhibitory Effect of Three Diketopiperazines from Marine-Derived Bacteria on HMGB1-Induced Septic Responses in Vitro and in Vivo. Diketopiperazines 27-44 high mobility group box 1 Homo sapiens 77-82 27627916-5 2016 Here, three diketopiperazines (1-3) isolated from two strains of marine-derived bacteria were investigated for their potential activities against HMGB1-mediated septic responses. Diketopiperazines 12-29 high mobility group box 1 Homo sapiens 146-151 27826978-6 2016 RESULTS: Our data showed a clear band of ~ 684 bp and ~ 981 bp related to HMGB1 and E7-HMGB1 genes in agarose gel, respectively. Sepharose 102-109 high mobility group box 1 Homo sapiens 74-79 27826978-6 2016 RESULTS: Our data showed a clear band of ~ 684 bp and ~ 981 bp related to HMGB1 and E7-HMGB1 genes in agarose gel, respectively. Sepharose 102-109 high mobility group box 1 Homo sapiens 87-92 27910767-4 2016 Emerging evidence indicates that melphalan can foster an immunogenic microenvironment by inducing immunogenic cell death (ICD) as characterized by membrane translocation of endoplasmic reticulum protein calreticulin (CRT) and by release of chromatin-binding protein high-mobility group box 1 (HMGB1). Melphalan 33-42 high mobility group box 1 Homo sapiens 266-291 27184952-9 2016 High-mobility group box 1 (HMGB1) was rapidly released and associated with TLR4 after TNF-alpha stimulation with a peak at 5 min, which was prevented by N-acetylcysteine, an antioxidant. Acetylcysteine 153-169 high mobility group box 1 Homo sapiens 0-25 27184952-9 2016 High-mobility group box 1 (HMGB1) was rapidly released and associated with TLR4 after TNF-alpha stimulation with a peak at 5 min, which was prevented by N-acetylcysteine, an antioxidant. Acetylcysteine 153-169 high mobility group box 1 Homo sapiens 27-32 27184952-10 2016 Glycyrrhizin (HMGB1 inhibitor), HMGB1-neutralizing antibody and siRNA depletion of HMGB1 all suppressed TNF-alpha-induced Syk activation and ET-1 production. Glycyrrhizic Acid 0-12 high mobility group box 1 Homo sapiens 14-19 27184952-11 2016 CONCLUSION: Upon TNF-alpha stimulation, TLR4 is activated by HMGB1 that is immediately released after the generation of reactive oxygen species, and plays a crucial role in the signal transduction. Reactive Oxygen Species 120-143 high mobility group box 1 Homo sapiens 61-66 27910767-4 2016 Emerging evidence indicates that melphalan can foster an immunogenic microenvironment by inducing immunogenic cell death (ICD) as characterized by membrane translocation of endoplasmic reticulum protein calreticulin (CRT) and by release of chromatin-binding protein high-mobility group box 1 (HMGB1). Melphalan 33-42 high mobility group box 1 Homo sapiens 293-298 26567221-0 2016 N-linked glycosylation plays a crucial role in the secretion of HMGB1. Nitrogen 0-1 high mobility group box 1 Homo sapiens 64-69 25139532-7 2016 Furthermore, the response to NACRT was found to be significantly associated with the preoperative serum HMGB-1 levels. nacrt 29-34 high mobility group box 1 Homo sapiens 104-110 25139532-8 2016 The use of NACRT contributes to preoperative serum HMGB-1 elevation, and these were risk factors for the occurrence of severe postoperative pulmonary complications in patients with esophageal cancer after thoracic esophagectomy. nacrt 11-16 high mobility group box 1 Homo sapiens 51-57 26567221-3 2016 Here, we identified the role of N-glycosylation of HMGB1 in extracellular secretion. Nitrogen 32-33 high mobility group box 1 Homo sapiens 51-56 26567221-4 2016 We found two consensus (N37 and N134) and one non-consensus (N135) residues that were N-glycosylated in HMGB1 by performing liquid chromatography tandem mass spectrometry (LC-MS/MS) and analyzing for N-glycan composition and structure. Nitrogen 24-25 high mobility group box 1 Homo sapiens 104-109 26567221-4 2016 We found two consensus (N37 and N134) and one non-consensus (N135) residues that were N-glycosylated in HMGB1 by performing liquid chromatography tandem mass spectrometry (LC-MS/MS) and analyzing for N-glycan composition and structure. n-glycan 200-208 high mobility group box 1 Homo sapiens 104-109 26567221-5 2016 Inhibition of N-glycosylation with tunicamycin resulted in a molecular shift of HMGB1 as assessed by gel electrophoresis. Nitrogen 14-15 high mobility group box 1 Homo sapiens 80-85 26567221-5 2016 Inhibition of N-glycosylation with tunicamycin resulted in a molecular shift of HMGB1 as assessed by gel electrophoresis. Tunicamycin 35-46 high mobility group box 1 Homo sapiens 80-85 26567221-8 2016 Taken together, we propose that HMGB1 is N-glycosylated, and that this is important for its DNA interaction and is a prerequisite for its nucleocytoplasmic transport and extracellular secretion. Nitrogen 41-42 high mobility group box 1 Homo sapiens 32-37 26334381-0 2015 Simvastatin protects the heart against ischemia reperfusion injury via inhibiting HMGB1 expression through PI3K/Akt signal pathways. Simvastatin 0-11 high mobility group box 1 Homo sapiens 82-87 26966912-6 2016 Glycyrrhetic acid inhibits the chemotactic and mitogenic function of HMGB1, binding to the hydrophobic residues that delimit the pockets in box A and B. Glycyrrhetinic Acid 0-17 high mobility group box 1 Homo sapiens 69-74 26702616-10 2015 MTX upregulated autophagy through its ability to enhance the expression of HMGB1 and Beclin-1 rather than through the Akt/mTOR pathway. Methotrexate 0-3 high mobility group box 1 Homo sapiens 75-80 26515875-11 2016 In addition, blockade of JNK MAPK and NF-kappaB phosphorylation prohibited HMGB1-induced production of pro-osteogenic factors, and mineralization of VICs. vics 149-153 high mobility group box 1 Homo sapiens 75-80 25307746-8 2015 Moreover, PGZ inhibited proliferative activity and invasive potential, and induced apoptosis and cell cycle arrest in HCC cells resulting in increased expression of PPARgamma and decreased expression of RAGE, NF-kappaB, HMGB1, p38MAPK, Ki-67, MMP-2, and CyclinD1. Pioglitazone 10-13 high mobility group box 1 Homo sapiens 220-225 27337797-5 2015 After treating the schizophrenics with the neuroleptic risperidone for 6 months, the serum levels of HMGB1, IL-1beta, TNF-alpha and IL-6 were decreased. Risperidone 55-66 high mobility group box 1 Homo sapiens 101-106 27337797-8 2015 HMGB1 may play a proinflammatory role in schizophrenia and the decrease of HMGB1 after neuroleptic risperidone treatment may be a marker of mental symptoms remission. Risperidone 99-110 high mobility group box 1 Homo sapiens 75-80 26293782-4 2015 In vitro, EGCG divergently affected HMGB1-mediated production of several chemokines: reducing CXCL15 and RANTES/CCL5, but elevating G-CSF and MIP-1alpha/CCL3 production by peritoneal macrophages. epigallocatechin gallate 10-14 high mobility group box 1 Homo sapiens 36-41 26769828-7 2015 Short-term (5 days) riboflavin deprivation resulted in the pathological macrophages activation, manifested especially in a reduction of cell viability and excess release of tumor necrosis factor-alpha (TNF-alpha) and high-mobility group box 1 (HMGB1) protein. Riboflavin 20-30 high mobility group box 1 Homo sapiens 217-242 26769828-7 2015 Short-term (5 days) riboflavin deprivation resulted in the pathological macrophages activation, manifested especially in a reduction of cell viability and excess release of tumor necrosis factor-alpha (TNF-alpha) and high-mobility group box 1 (HMGB1) protein. Riboflavin 20-30 high mobility group box 1 Homo sapiens 244-249 26884867-5 2015 HMGB1 siRNAs were designed and chemically synthesized, and then transfected into the breast cancer cell line MCF-7 with lipofectamine 2000. Lipofectamine 120-138 high mobility group box 1 Homo sapiens 0-5 26349060-0 2015 The HIV Protease Inhibitor Saquinavir Inhibits HMGB1-Driven Inflammation by Targeting the Interaction of Cathepsin V with TLR4/MyD88. Saquinavir 27-37 high mobility group box 1 Homo sapiens 47-52 26397184-5 2015 Anticancer agents including doxorubicin, cisplatin and etoposide each induced HMGB1 upregulation, promoted cytosolic HMGB1 translocation and the elevation of autophagic activity in human NB cells. Doxorubicin 28-39 high mobility group box 1 Homo sapiens 78-83 26349060-1 2015 Extracellular high-mobility group box 1 (HMGB1) (disulfide form), via activation of toll-like receptor 4 (TLR4)-dependent signaling, is a strong driver of pathologic inflammation in both acute and chronic conditions. Disulfides 49-58 high mobility group box 1 Homo sapiens 14-39 26349060-1 2015 Extracellular high-mobility group box 1 (HMGB1) (disulfide form), via activation of toll-like receptor 4 (TLR4)-dependent signaling, is a strong driver of pathologic inflammation in both acute and chronic conditions. Disulfides 49-58 high mobility group box 1 Homo sapiens 41-46 26349060-4 2015 Here we report that the first-generation HIV-PI saquinavir (SQV), as well as a newly identified mammalian protease inhibitor STO33438 (334), potently block disulfide HMGB1-induced TLR4 activation, as assayed by the production of TNF-alpha by human monocyte-derived macrophages (THP-1). sto33438 125-133 high mobility group box 1 Homo sapiens 166-171 26349060-4 2015 Here we report that the first-generation HIV-PI saquinavir (SQV), as well as a newly identified mammalian protease inhibitor STO33438 (334), potently block disulfide HMGB1-induced TLR4 activation, as assayed by the production of TNF-alpha by human monocyte-derived macrophages (THP-1). Disulfides 156-165 high mobility group box 1 Homo sapiens 166-171 26349060-10 2015 Disulfide HMGB1 drives pathologic inflammation in many models by activating signaling through TLR4. Disulfides 0-9 high mobility group box 1 Homo sapiens 10-15 26349060-11 2015 Cell-based screening identified the mammalian protease cathepsin V as a novel therapeutic target to inhibit TLR4-mediated inflammation induced by extracellular HMGB1 (disulfide form). Disulfides 167-176 high mobility group box 1 Homo sapiens 160-165 26349060-12 2015 We identified two protease inhibitors (PIs) that block cathepsin V and thereby inhibit disulfide HMGB1-induced TLR4 activation: saquinavir (SQV), a first-generation PI targeting viral HIV protease and STO33438 (334), targeting mammalian proteases. Disulfides 87-96 high mobility group box 1 Homo sapiens 97-102 26349060-12 2015 We identified two protease inhibitors (PIs) that block cathepsin V and thereby inhibit disulfide HMGB1-induced TLR4 activation: saquinavir (SQV), a first-generation PI targeting viral HIV protease and STO33438 (334), targeting mammalian proteases. Saquinavir 128-138 high mobility group box 1 Homo sapiens 97-102 26397184-5 2015 Anticancer agents including doxorubicin, cisplatin and etoposide each induced HMGB1 upregulation, promoted cytosolic HMGB1 translocation and the elevation of autophagic activity in human NB cells. Doxorubicin 28-39 high mobility group box 1 Homo sapiens 117-122 26397184-5 2015 Anticancer agents including doxorubicin, cisplatin and etoposide each induced HMGB1 upregulation, promoted cytosolic HMGB1 translocation and the elevation of autophagic activity in human NB cells. Cisplatin 41-50 high mobility group box 1 Homo sapiens 78-83 26397184-5 2015 Anticancer agents including doxorubicin, cisplatin and etoposide each induced HMGB1 upregulation, promoted cytosolic HMGB1 translocation and the elevation of autophagic activity in human NB cells. Cisplatin 41-50 high mobility group box 1 Homo sapiens 117-122 26397184-5 2015 Anticancer agents including doxorubicin, cisplatin and etoposide each induced HMGB1 upregulation, promoted cytosolic HMGB1 translocation and the elevation of autophagic activity in human NB cells. Etoposide 55-64 high mobility group box 1 Homo sapiens 78-83 26397184-5 2015 Anticancer agents including doxorubicin, cisplatin and etoposide each induced HMGB1 upregulation, promoted cytosolic HMGB1 translocation and the elevation of autophagic activity in human NB cells. Etoposide 55-64 high mobility group box 1 Homo sapiens 117-122 26397184-9 2015 In addition, we found that HMGB1 facilitated autophagic progression and reduced oxidative stress induced by doxorubicin. Doxorubicin 108-119 high mobility group box 1 Homo sapiens 27-32 26596890-6 2015 Anti-HMGB1 antibody levels were analysed in patient and control (n = 112) sera by an in-house ELISA using recombinant histidine-tagged HMGB1. Histidine 118-127 high mobility group box 1 Homo sapiens 135-140 26597985-4 2016 In this study, we showed that SBP1, -2, and -3 individually bind different basic amino acids with exquisite specificity. Amino Acids, Basic 75-92 high mobility group box 1 Homo sapiens 30-34 26548953-6 2015 Induced asymmetry involving tilting of the nonamer-binding domain dimer of RAG1 upon binding of HMGB1-bent 12-RSS or 23-RSS underlies the molecular mechanism for the 12/23 rule. 12-rss 107-113 high mobility group box 1 Homo sapiens 96-101 26548953-6 2015 Induced asymmetry involving tilting of the nonamer-binding domain dimer of RAG1 upon binding of HMGB1-bent 12-RSS or 23-RSS underlies the molecular mechanism for the 12/23 rule. 23-rss 117-123 high mobility group box 1 Homo sapiens 96-101 26557438-0 2015 Suberoylanilide hydroxamic acid suppresses hepatic stellate cells activation by HMGB1 dependent reduction of NF-kappaB1. Vorinostat 0-31 high mobility group box 1 Homo sapiens 80-85 26525891-6 2015 In HepG2 cells, palmitic acid (PA)-induced pro-inflammatory cytokines release were blocked by HMGB1 small interfering RNA (siRNA) transfection. Palmitic Acid 16-29 high mobility group box 1 Homo sapiens 94-99 26525891-6 2015 In HepG2 cells, palmitic acid (PA)-induced pro-inflammatory cytokines release were blocked by HMGB1 small interfering RNA (siRNA) transfection. Palmitic Acid 31-33 high mobility group box 1 Homo sapiens 94-99 26557438-7 2015 Bioinformatic analyses of these altered genes highlighted the high mobility group box 1 (HMGB1) pathway was one of the most relevant pathways that contributed to SAHA induced suppression of HSCs activation. Vorinostat 162-166 high mobility group box 1 Homo sapiens 62-87 26557438-7 2015 Bioinformatic analyses of these altered genes highlighted the high mobility group box 1 (HMGB1) pathway was one of the most relevant pathways that contributed to SAHA induced suppression of HSCs activation. Vorinostat 162-166 high mobility group box 1 Homo sapiens 89-94 26557438-8 2015 Further studies demonstrated the increased acetylation of intracellular HMGB1 in SAHA treated HSCs, and this increasing is most likely to be responsible for SAHA induced down-regulation of nuclear factor kappa B1 (NF-kappaB1) and is one of the main underlying mechanisms for the therapeutic effect of SAHA for liver fibrosis. Vorinostat 81-85 high mobility group box 1 Homo sapiens 72-77 26557438-8 2015 Further studies demonstrated the increased acetylation of intracellular HMGB1 in SAHA treated HSCs, and this increasing is most likely to be responsible for SAHA induced down-regulation of nuclear factor kappa B1 (NF-kappaB1) and is one of the main underlying mechanisms for the therapeutic effect of SAHA for liver fibrosis. Vorinostat 157-161 high mobility group box 1 Homo sapiens 72-77 26557438-8 2015 Further studies demonstrated the increased acetylation of intracellular HMGB1 in SAHA treated HSCs, and this increasing is most likely to be responsible for SAHA induced down-regulation of nuclear factor kappa B1 (NF-kappaB1) and is one of the main underlying mechanisms for the therapeutic effect of SAHA for liver fibrosis. Vorinostat 157-161 high mobility group box 1 Homo sapiens 72-77 26040768-10 2015 The results demonstrated that the expression level of HMGB1 in A549/DDP cells was much higher than that of A549 cells; moreover, transfection with HMGB1 shRNA in A549/DDP cells decreased the IC50 value of cisplatin in A549/DDP cells. Cisplatin 205-214 high mobility group box 1 Homo sapiens 54-59 26040768-10 2015 The results demonstrated that the expression level of HMGB1 in A549/DDP cells was much higher than that of A549 cells; moreover, transfection with HMGB1 shRNA in A549/DDP cells decreased the IC50 value of cisplatin in A549/DDP cells. Cisplatin 205-214 high mobility group box 1 Homo sapiens 147-152 26040768-5 2015 Firstly, we detected the expression levels of HMGB1 by by real-time PCR and western blotting analysis, and the results demonstrated that HMGB1 was much higher expressed in non-small cell lung cancer cell lines, including A549, SPC-1-1, NCI-2170, SK-MES-1, and NCI-H1299, compared with that of WI-38. sk-mes 246-252 high mobility group box 1 Homo sapiens 137-142 26875270-11 2015 The expression of HMGB1 protein in Pgenesil-1/HMGB1 siRNA group (0.134 +- 0.048) was significantly lower than that of Pgenesil-1 group (0.581+- 0.032) and untransfected group (0.514 +- 0.069) (P < 0.05). pgenesil-1 35-45 high mobility group box 1 Homo sapiens 18-23 26875270-11 2015 The expression of HMGB1 protein in Pgenesil-1/HMGB1 siRNA group (0.134 +- 0.048) was significantly lower than that of Pgenesil-1 group (0.581+- 0.032) and untransfected group (0.514 +- 0.069) (P < 0.05). pgenesil-1 35-45 high mobility group box 1 Homo sapiens 46-51 26875270-12 2015 ELISA results showed that IL-1beta and TNF-alpha in supernatants of Pgenesil-1/HMGB1 siRNA group were significantly lower than those of Pgenesil-1 group and untransfected group (P < 0.05). pgenesil-1 68-78 high mobility group box 1 Homo sapiens 79-84 26875270-12 2015 ELISA results showed that IL-1beta and TNF-alpha in supernatants of Pgenesil-1/HMGB1 siRNA group were significantly lower than those of Pgenesil-1 group and untransfected group (P < 0.05). pgenesil 68-76 high mobility group box 1 Homo sapiens 79-84 26454330-3 2015 This study explored HMGB-1 as a therapeutic target for DR treatment through observing its role in retinal ganglion cells (GRCs) in a high glucose environment. Glucose 138-145 high mobility group box 1 Homo sapiens 20-26 26770371-0 2015 Effect of high mobility group box 1 on the human retinal pigment epithelial cell in high-glucose condition. Glucose 89-96 high mobility group box 1 Homo sapiens 10-35 26770371-6 2015 In the advanced in-vitro study, we detect the protective effect of HMGB1 on the RPE cells in high glucose condition. Glucose 98-105 high mobility group box 1 Homo sapiens 67-72 26770371-10 2015 Besides, HMGB1 treatment would up-regulate the expression of VEGFA in the RPE cells in high glucose condition. Glucose 92-99 high mobility group box 1 Homo sapiens 9-14 26537405-3 2015 We found that hairpin polyamide 1 triggers the release of the damage-associated molecular patterns calreticulin, ATP and HMGB1 in a slow necrotic-type cell death. Nylons 22-31 high mobility group box 1 Homo sapiens 121-126 26469759-5 2015 Supernatants from DTX-treated DU145 tumor cells, which were shown to contain HMGB1, effectively induced sCLU from newly-plated DU145 tumor cells and protected them from DTX toxicity. Docetaxel 18-21 high mobility group box 1 Homo sapiens 77-82 26469759-5 2015 Supernatants from DTX-treated DU145 tumor cells, which were shown to contain HMGB1, effectively induced sCLU from newly-plated DU145 tumor cells and protected them from DTX toxicity. Docetaxel 169-172 high mobility group box 1 Homo sapiens 77-82 26454330-9 2015 RESULTS: HMGB-1 mRNA was up-regulated (P=0.015) and protein secretion increased (P=0.022) in the high glucose environment. Glucose 102-109 high mobility group box 1 Homo sapiens 9-15 26248639-3 2015 It is revealed that HMGB1 (high-mobility group protein B1) was the most commonly found protein that recognizes all Pt(II)-DNA probes and prefers cisplatin-DNA probes more than the others. Cisplatin 145-154 high mobility group box 1 Homo sapiens 20-25 26248639-3 2015 It is revealed that HMGB1 (high-mobility group protein B1) was the most commonly found protein that recognizes all Pt(II)-DNA probes and prefers cisplatin-DNA probes more than the others. Cisplatin 145-154 high mobility group box 1 Homo sapiens 27-57 26813430-11 2015 MTT assay showed that the cell proliferation in the HMGB1-siRNA group was significantly inhibited when compared with that in the HMGB1-siRNA-Neg group and control group (P<0.05). monooxyethylene trimethylolpropane tristearate 0-3 high mobility group box 1 Homo sapiens 52-57 25943534-9 2015 The antitumor activity of anti-PD-L1 was enhanced by combination with oxaliplatin, which resulted in increased release of HMGB1 within CT26 tumors. Oxaliplatin 70-81 high mobility group box 1 Homo sapiens 122-127 26273067-12 2015 CONCLUSIONS: We conclude that altered OPN expression in CAVS affects cellular HMGB1 function inducing cytoplasmic translocation and secretion of HMGB1 in endothelial cells and VICs, thus indicating a regulatory role for OPN in the progression of CAVS through alteration of HMGB1 function. vics 176-180 high mobility group box 1 Homo sapiens 78-83 26206236-0 2015 Inhibitory effects of polyozellin from Polyozellus multiplex on HMGB1-mediated septic responses. polyozellin 22-33 high mobility group box 1 Homo sapiens 64-69 26206236-4 2015 METHODS: The anti-inflammatory activities of polyozellin were determined by measuring permeability, human neutrophil adhesion and migration, and activation of proinflammatory proteins in HMGB1-activated HUVECs and mice. polyozellin 45-56 high mobility group box 1 Homo sapiens 187-192 26206236-5 2015 RESULTS: According to the results, polyozellin effectively inhibited lipopolysaccharide (LPS)-induced release of HMGB1, and suppressed HMGB1-mediated septic responses, such as hyperpermeability, adhesion and migration of leukocytes, and expression of cell adhesion molecules. polyozellin 35-46 high mobility group box 1 Homo sapiens 113-118 26206236-5 2015 RESULTS: According to the results, polyozellin effectively inhibited lipopolysaccharide (LPS)-induced release of HMGB1, and suppressed HMGB1-mediated septic responses, such as hyperpermeability, adhesion and migration of leukocytes, and expression of cell adhesion molecules. polyozellin 35-46 high mobility group box 1 Homo sapiens 135-140 26206236-6 2015 In addition, polyozellin suppressed the production of tumor necrosis factor-alpha and interleukin (IL)-6, and the activation of nuclear factor-kappaB and extracellular signal-regulated kinases 1/2 by HMGB1. polyozellin 13-24 high mobility group box 1 Homo sapiens 200-205 26206236-7 2015 CONCLUSION: Collectively, these results indicate that P. multiplex containing polyozellin could be commercialized as functional food for preventing and treatment of various severe vascular inflammatory diseases via inhibition of the HMGB1 signaling pathway. polyozellin 78-89 high mobility group box 1 Homo sapiens 233-238 25956731-6 2015 The equipotent P2Y2R agonists ATP and UTP enhanced proliferation, RAGE expression and HMGB1 secretion in IL-1beta-pretreated VSMCs. Adenosine Triphosphate 30-33 high mobility group box 1 Homo sapiens 86-91 25956731-6 2015 The equipotent P2Y2R agonists ATP and UTP enhanced proliferation, RAGE expression and HMGB1 secretion in IL-1beta-pretreated VSMCs. Uridine Triphosphate 38-41 high mobility group box 1 Homo sapiens 86-91 25956731-6 2015 The equipotent P2Y2R agonists ATP and UTP enhanced proliferation, RAGE expression and HMGB1 secretion in IL-1beta-pretreated VSMCs. vsmcs 125-130 high mobility group box 1 Homo sapiens 86-91 25956731-9 2015 The ERK, AKT, PKC, Rac-1 and ROCK2 pathways were involved in UTP-induced cell proliferation and migration, MMP-2 and HMGB1 secretion and RAGE expression in the IL-1beta-pretreated VSMCs. Uridine Triphosphate 61-64 high mobility group box 1 Homo sapiens 117-122 26406975-4 2015 Many of the intra- and extracellular functions of HMGB1 depend on redox-sensitive cysteine residues of the protein. Cysteine 82-90 high mobility group box 1 Homo sapiens 50-55 26337661-0 2015 Role of high-mobility group box 1 in methamphetamine-induced activation and migration of astrocytes. Methamphetamine 37-52 high mobility group box 1 Homo sapiens 8-33 26337661-8 2015 RESULTS: Exposure of C6 cells to methamphetamine increased the expression of HMGB1 via the activation of sigma-1R, Src, ERK mitogen-activated protein kinase, and downstream NF-kappaB p65 pathways. Methamphetamine 33-48 high mobility group box 1 Homo sapiens 77-82 26337661-10 2015 Knockdown of HMGB1 in astrocytes transfected with HMGB1 siRNA attenuated the increased cell activation and migration induced by methamphetamine, thereby implicating the role of HMGB1 in the activation and migration of C6 cells and primary human astrocytes. Methamphetamine 128-143 high mobility group box 1 Homo sapiens 13-18 26337661-10 2015 Knockdown of HMGB1 in astrocytes transfected with HMGB1 siRNA attenuated the increased cell activation and migration induced by methamphetamine, thereby implicating the role of HMGB1 in the activation and migration of C6 cells and primary human astrocytes. Methamphetamine 128-143 high mobility group box 1 Homo sapiens 50-55 26337661-10 2015 Knockdown of HMGB1 in astrocytes transfected with HMGB1 siRNA attenuated the increased cell activation and migration induced by methamphetamine, thereby implicating the role of HMGB1 in the activation and migration of C6 cells and primary human astrocytes. Methamphetamine 128-143 high mobility group box 1 Homo sapiens 50-55 26337661-11 2015 CONCLUSIONS: This study demonstrated that methamphetamine-mediated activation and migration of astrocytes involved HMGB1 up-regulation through an autocrine mechanism. Methamphetamine 42-57 high mobility group box 1 Homo sapiens 115-120 26337661-12 2015 Targeting HMGB1 could provide insights into the development of a potential therapeutic approach for alleviation of cell activation and migration of astrocytes induced by methamphetamine. Methamphetamine 170-185 high mobility group box 1 Homo sapiens 10-15 26408615-1 2015 OBJECTIVE: To determine the effect of p38 MAPK inhibitor (SB203580) on TNF-alpha -induced high mobility group protein B1 (HMGB1) expression in microglial cells. SB 203580 58-66 high mobility group box 1 Homo sapiens 90-120 26408615-1 2015 OBJECTIVE: To determine the effect of p38 MAPK inhibitor (SB203580) on TNF-alpha -induced high mobility group protein B1 (HMGB1) expression in microglial cells. SB 203580 58-66 high mobility group box 1 Homo sapiens 122-127 26408615-5 2015 The TNF-alpha-induced HMGB1 protein and mRNA expression was suppressed by SB203580. SB 203580 74-82 high mobility group box 1 Homo sapiens 22-27 26813430-11 2015 MTT assay showed that the cell proliferation in the HMGB1-siRNA group was significantly inhibited when compared with that in the HMGB1-siRNA-Neg group and control group (P<0.05). monooxyethylene trimethylolpropane tristearate 0-3 high mobility group box 1 Homo sapiens 129-134 27057465-2 2016 This effect is in part due to the capacity of anthracyclines to induce immunogenic cell death (ICD), a cell death modality that is preceded by autophagy and followed by HMGB1 release. Anthracyclines 46-60 high mobility group box 1 Homo sapiens 169-174 26079697-0 2015 Ethanol directly induced HMGB1 release through NOX2/NLRP1 inflammasome in neuronal cells. Ethanol 0-7 high mobility group box 1 Homo sapiens 25-30 26079697-3 2015 The present study was designed to test whether ethanol directly induced HMGB1 release and to explore the cellular and molecular mechanism mediating its action. Ethanol 47-54 high mobility group box 1 Homo sapiens 72-77 26245198-0 2015 HMGB1: a novel protein that induced platelets active and aggregation via Toll-like receptor-4, NF-kappaB and cGMP dependent mechanisms. Cyclic GMP 109-113 high mobility group box 1 Homo sapiens 0-5 26245198-11 2015 iv) We proposed that extracellular HMGB1 seems to promote the phosphorylation of GPIIb/IIIa and subsequent platelet aggregation via TLR4/NF-kappaB and cGMP pathway. Cyclic GMP 151-155 high mobility group box 1 Homo sapiens 35-40 25816800-2 2015 HMGB1 has been implicated in the pathophysiology of several neuroinflammatory conditions including ischemia, traumatic brain injury, seizure and chronic ethanol use. Ethanol 153-160 high mobility group box 1 Homo sapiens 0-5 26079697-4 2015 It was found that ethanol increased significant HMGB1 release from SH-SY5Y cells and from cultured primary cortical neurons as detected by ELISA assay. Ethanol 18-25 high mobility group box 1 Homo sapiens 48-53 26079697-8 2015 In addition, z-YVAD-fmk, an inhibitor of caspase-1, decreased the ethanol-induced HMGB1 release. benzyloxycarbonyltyrosyl-valyl-alanyl-aspartic acid fluoromethyl ketone 13-23 high mobility group box 1 Homo sapiens 82-87 26079697-8 2015 In addition, z-YVAD-fmk, an inhibitor of caspase-1, decreased the ethanol-induced HMGB1 release. Ethanol 66-73 high mobility group box 1 Homo sapiens 82-87 26079697-9 2015 It is concluded that ethanol-induced HMGB1 release is associated with NOX2/NLRP1 inflammasome signaling, which represents a novel mechanism of ethanol-associated neuron injury. Ethanol 21-28 high mobility group box 1 Homo sapiens 37-42 26079697-9 2015 It is concluded that ethanol-induced HMGB1 release is associated with NOX2/NLRP1 inflammasome signaling, which represents a novel mechanism of ethanol-associated neuron injury. Ethanol 143-150 high mobility group box 1 Homo sapiens 37-42 25743565-3 2015 We accessed this question by monitoring the effects of posttreatment carbazole derivatives on LPS- and CLP-mediated release of HMGB1 and HMGB1-mediated regulation of proinflammatory responses in human umbilical vein endothelial cells (HUVECs) and septic mice. carbazole 69-78 high mobility group box 1 Homo sapiens 127-132 26140987-2 2015 In order to down-regulate HMGB1 expression in airway epithelial cells, siRNA directed against HMGB1 was delivered through nanocomplexes based on a cationic copolymer of poly(N-2-hydroxyethyl)-d,l-aspartamide (PHEA) by using H441 cells. copolymer 156-165 high mobility group box 1 Homo sapiens 94-99 26140987-2 2015 In order to down-regulate HMGB1 expression in airway epithelial cells, siRNA directed against HMGB1 was delivered through nanocomplexes based on a cationic copolymer of poly(N-2-hydroxyethyl)-d,l-aspartamide (PHEA) by using H441 cells. poly(n-2-hydroxyethyl)-d,l-aspartamide 169-207 high mobility group box 1 Homo sapiens 94-99 26140987-2 2015 In order to down-regulate HMGB1 expression in airway epithelial cells, siRNA directed against HMGB1 was delivered through nanocomplexes based on a cationic copolymer of poly(N-2-hydroxyethyl)-d,l-aspartamide (PHEA) by using H441 cells. Phenethylamines 209-213 high mobility group box 1 Homo sapiens 94-99 26140987-10 2015 PEGylated PHEA-Spm/siRNA nanocomplexes were able to down-regulate HMGB1 mRNA levels up to 61% of control cells. Phenethylamines 10-14 high mobility group box 1 Homo sapiens 66-71 26140987-12 2015 In conclusion, we have found optimal conditions for down-regulation of HMGB1 by siRNA delivery mediated by polyaminoacidic polymers in airway epithelial cells in the absence of cytotoxicity. polyaminoacidic polymers 107-131 high mobility group box 1 Homo sapiens 71-76 25743565-3 2015 We accessed this question by monitoring the effects of posttreatment carbazole derivatives on LPS- and CLP-mediated release of HMGB1 and HMGB1-mediated regulation of proinflammatory responses in human umbilical vein endothelial cells (HUVECs) and septic mice. carbazole 69-78 high mobility group box 1 Homo sapiens 137-142 25743565-4 2015 The new 3"-N-substituted carbazole derivatives 1-5 inhibited the release of HMGB1 and downregulated HMGB1-dependent inflammatory responses in human endothelial cells. 3"-n-substituted carbazole 8-34 high mobility group box 1 Homo sapiens 76-81 25743565-4 2015 The new 3"-N-substituted carbazole derivatives 1-5 inhibited the release of HMGB1 and downregulated HMGB1-dependent inflammatory responses in human endothelial cells. 3"-n-substituted carbazole 8-34 high mobility group box 1 Homo sapiens 100-105 26124331-0 2015 High-mobility Group Box 1 and Mitogen-activated Protein Kinase activated Protein Kinase-2 Are Up-regulated in Gemcitabine-resistant Pancreatic Cancer Cells. gemcitabine 110-121 high mobility group box 1 Homo sapiens 0-25 26143914-5 2015 Two box A domains of HMGB1 collaborate in an unusual configuration in which the Phe37 residues of both domains stack together and intercalate the same CG base pair, generating highly kinked DNA. cysteinylglycine 151-153 high mobility group box 1 Homo sapiens 21-26 26124331-9 2015 CONCLUSION: The increase of both HMGB1 and MAPKAPK2 in KLM1-R cells compared to KLM1 suggest the possibility of the activation of the pathway of HSP27 by HMGB1 and MAPKAPK2 in gemcitabine-resistant KLM1-R cells. gemcitabine 176-187 high mobility group box 1 Homo sapiens 33-38 26124331-9 2015 CONCLUSION: The increase of both HMGB1 and MAPKAPK2 in KLM1-R cells compared to KLM1 suggest the possibility of the activation of the pathway of HSP27 by HMGB1 and MAPKAPK2 in gemcitabine-resistant KLM1-R cells. gemcitabine 176-187 high mobility group box 1 Homo sapiens 154-159 26101955-7 2015 SA suppresses both the chemoattractant activity of fully reduced HMGB1 and the increased expression of proinflammatory cytokine genes and cyclooxygenase 2 (COX-2) induced by disulfide HMGB1. Disulfides 174-183 high mobility group box 1 Homo sapiens 184-189 25861801-18 2015 In contrast to TLR4- and TLR2-binding lipopolysaccharide used as a positive control, disulfide-HMGB1 did not stimulate proinflammatory cytokines. Disulfides 85-94 high mobility group box 1 Homo sapiens 95-100 26109393-13 2015 CG + GG genotype was associated with high HMGB1 levels compared with the genotype CC at 4 h (p = 0.023), and 24 h (p = 0.015) after CPB. cysteinylglycine 0-2 high mobility group box 1 Homo sapiens 42-47 26101955-0 2015 Aspirin"s Active Metabolite Salicylic Acid Targets High Mobility Group Box 1 to Modulate Inflammatory Responses. Aspirin 0-7 high mobility group box 1 Homo sapiens 51-76 26101955-8 2015 Natural and synthetic SA derivatives with greater potency for inhibition of HMGB1 were identified, providing proof-of-concept that new molecules with high efficacy against sterile inflammation are attainable. Salicylic Acid 22-24 high mobility group box 1 Homo sapiens 76-81 26101955-0 2015 Aspirin"s Active Metabolite Salicylic Acid Targets High Mobility Group Box 1 to Modulate Inflammatory Responses. Salicylic Acid 28-42 high mobility group box 1 Homo sapiens 51-76 26101955-5 2015 SA-binding sites on HMGB1 were identified in the HMG-box domains by nuclear magnetic resonance (NMR) spectroscopic studies and confirmed by mutational analysis. Salicylic Acid 0-2 high mobility group box 1 Homo sapiens 20-25 26101955-9 2015 An HMGB1 protein mutated in one of the SA-binding sites identified by NMR chemical shift perturbation studies retained chemoattractant activity, but lost binding of and inhibition by SA and its derivatives, thereby firmly establishing that SA binding to HMGB1 directly suppresses its proinflammatory activities. Salicylic Acid 39-41 high mobility group box 1 Homo sapiens 3-8 26101955-7 2015 SA suppresses both the chemoattractant activity of fully reduced HMGB1 and the increased expression of proinflammatory cytokine genes and cyclooxygenase 2 (COX-2) induced by disulfide HMGB1. Salicylic Acid 0-2 high mobility group box 1 Homo sapiens 65-70 26101955-9 2015 An HMGB1 protein mutated in one of the SA-binding sites identified by NMR chemical shift perturbation studies retained chemoattractant activity, but lost binding of and inhibition by SA and its derivatives, thereby firmly establishing that SA binding to HMGB1 directly suppresses its proinflammatory activities. Salicylic Acid 39-41 high mobility group box 1 Homo sapiens 254-259 26101955-7 2015 SA suppresses both the chemoattractant activity of fully reduced HMGB1 and the increased expression of proinflammatory cytokine genes and cyclooxygenase 2 (COX-2) induced by disulfide HMGB1. Salicylic Acid 0-2 high mobility group box 1 Homo sapiens 184-189 26101955-9 2015 An HMGB1 protein mutated in one of the SA-binding sites identified by NMR chemical shift perturbation studies retained chemoattractant activity, but lost binding of and inhibition by SA and its derivatives, thereby firmly establishing that SA binding to HMGB1 directly suppresses its proinflammatory activities. Salicylic Acid 183-185 high mobility group box 1 Homo sapiens 3-8 26101955-9 2015 An HMGB1 protein mutated in one of the SA-binding sites identified by NMR chemical shift perturbation studies retained chemoattractant activity, but lost binding of and inhibition by SA and its derivatives, thereby firmly establishing that SA binding to HMGB1 directly suppresses its proinflammatory activities. Salicylic Acid 183-185 high mobility group box 1 Homo sapiens 3-8 26101955-10 2015 Identification of HMGB1 as a pharmacological target of SA/aspirin provides new insights into the mechanisms of action of one of the world"s longest and most used natural and synthetic drugs. Salicylic Acid 55-57 high mobility group box 1 Homo sapiens 18-23 26101955-10 2015 Identification of HMGB1 as a pharmacological target of SA/aspirin provides new insights into the mechanisms of action of one of the world"s longest and most used natural and synthetic drugs. Aspirin 58-65 high mobility group box 1 Homo sapiens 18-23 26068794-0 2015 Aspirin delays mesothelioma growth by inhibiting HMGB1-mediated tumor progression. Aspirin 0-7 high mobility group box 1 Homo sapiens 49-54 26068794-3 2015 We hypothesized that ASA may exert anticancer properties in MM by abrogating the carcinogenic effects of HMGB1. Aspirin 21-24 high mobility group box 1 Homo sapiens 105-110 26068794-4 2015 Using HMGB1-secreting and -non-secreting human MM cell lines, we determined whether aspirin inhibited the hallmarks of HMGB1-induced MM cell growth in vitro and in vivo. Aspirin 84-91 high mobility group box 1 Homo sapiens 119-124 26068794-5 2015 Our data demonstrated that ASA and its metabolite, salicylic acid (SA), inhibit motility, migration, invasion and anchorage-independent colony formation of MM cells via a novel HMGB1-mediated mechanism. Aspirin 27-30 high mobility group box 1 Homo sapiens 177-182 26068794-5 2015 Our data demonstrated that ASA and its metabolite, salicylic acid (SA), inhibit motility, migration, invasion and anchorage-independent colony formation of MM cells via a novel HMGB1-mediated mechanism. Salicylic Acid 51-65 high mobility group box 1 Homo sapiens 177-182 26068794-5 2015 Our data demonstrated that ASA and its metabolite, salicylic acid (SA), inhibit motility, migration, invasion and anchorage-independent colony formation of MM cells via a novel HMGB1-mediated mechanism. Salicylic Acid 28-30 high mobility group box 1 Homo sapiens 177-182 26068794-6 2015 ASA/SA, at serum concentrations comparable to those achieved in humans taking therapeutic doses of aspirin, and BoxA, a specific inhibitor of HMGB1, markedly reduced MM growth in xenograft mice and significantly improved survival of treated animals. Salicylic Acid 1-3 high mobility group box 1 Homo sapiens 142-147 26068794-7 2015 The effects of ASA and BoxA were cyclooxygenase-2 independent and were not additive, consistent with both acting via inhibition of HMGB1 activity. Aspirin 15-18 high mobility group box 1 Homo sapiens 131-136 26068794-8 2015 Our findings provide a rationale for the well documented, yet poorly understood antitumorigenic activity of aspirin, which we show proceeds via HMGB1 inhibition. Aspirin 108-115 high mobility group box 1 Homo sapiens 144-149 26261543-0 2015 MiR-218 inhibits HMGB1-mediated autophagy in endometrial carcinoma cells during chemotherapy. mir-218 0-7 high mobility group box 1 Homo sapiens 17-22 25856291-8 2015 Xenon treatment enhanced HIF-1alpha, which attenuated HMGB-1 translocation and NF-kappaB activation in A549 cells with oxidative and inflammatory stress. Xenon 0-5 high mobility group box 1 Homo sapiens 54-60 25856291-11 2015 CONCLUSION: Xenon treatment confers protection against distant lung injury triggered by renal graft IRI, which is likely through the activation of mTOR-HIF-1alpha pathway and suppression of the HMGB-1 translocation from nuclei to cytoplasm. Xenon 12-17 high mobility group box 1 Homo sapiens 194-200 26045910-0 2015 Increase in the Level of Proinflammatory Cytokine HMGB1 in Nasal Fluids of Patients With Rhinitis and its Sequestration by Glycyrrhizin Induces Eosinophil Cell Death. Glycyrrhizic Acid 123-135 high mobility group box 1 Homo sapiens 50-55 26045910-6 2015 We also found that a formulation containing the HMGB1-binding compound glycyrrhizin (GLT) reduced the HMGB1 content in nasal fluids of rhinitis patients to an extent similar to that with nasal budesonide treatment. Glycyrrhizic Acid 71-83 high mobility group box 1 Homo sapiens 48-53 26045910-6 2015 We also found that a formulation containing the HMGB1-binding compound glycyrrhizin (GLT) reduced the HMGB1 content in nasal fluids of rhinitis patients to an extent similar to that with nasal budesonide treatment. Glycyrrhizic Acid 71-83 high mobility group box 1 Homo sapiens 102-107 26157321-15 2015 miR-218 can negatively regulate the HMGB1 protein expression and inhibit the proliferation and invasion of pancreatic cancer cells. mir-218 0-7 high mobility group box 1 Homo sapiens 36-41 26157323-0 2015 microRNA-218 promotes gemcitabine sensitivity in human pancreatic cancer cells by regulating HMGB1 expression. gemcitabine 22-33 high mobility group box 1 Homo sapiens 93-98 26157323-11 2015 miR-218 promoted the sensitivity of PANC-1 cells to GEM, which was achieved mainly through regulating the expression of HMGB1 in PANC-1 cells. mir-218 0-7 high mobility group box 1 Homo sapiens 120-125 26261543-6 2015 HMGB1 was upregulated in paclitaxel resistant EC cells, it mediated autophagy and contributed to chemotherapy resistance in endometrial carcinoma in vitro. Paclitaxel 25-35 high mobility group box 1 Homo sapiens 0-5 26261550-0 2015 HMGB1 is activated in type 2 diabetes mellitus patients and in mesangial cells in response to high glucose. Glucose 99-106 high mobility group box 1 Homo sapiens 0-5 26261550-4 2015 In the present study, we examined the promotion by high glucose to High-mobility group box-1 (HMGB1) in patients with type 2 diabetes mellitus or in renal mesangial SV40 MES 13 cells. Glucose 56-63 high mobility group box 1 Homo sapiens 67-92 26261550-4 2015 In the present study, we examined the promotion by high glucose to High-mobility group box-1 (HMGB1) in patients with type 2 diabetes mellitus or in renal mesangial SV40 MES 13 cells. Glucose 56-63 high mobility group box 1 Homo sapiens 94-99 26261550-4 2015 In the present study, we examined the promotion by high glucose to High-mobility group box-1 (HMGB1) in patients with type 2 diabetes mellitus or in renal mesangial SV40 MES 13 cells. 2-(N-morpholino)ethanesulfonic acid 170-173 high mobility group box 1 Homo sapiens 94-99 26261550-7 2015 The in vitro results indicated that HMGB1 mediated the D-glucose-induced pro-inflammatory cytokines in mesangial cells. Glucose 55-64 high mobility group box 1 Homo sapiens 36-41 26261550-9 2015 In summary, the present study indicated that HMGB1 was significantly promoted by the glucose in vivo or in vitro, in an association with an upregulation of pro-inflammatory cytokines, via activating NF-kappaB signaling pathway. Glucose 85-92 high mobility group box 1 Homo sapiens 45-50 26261550-10 2015 And the strategy of HMGB1 inhibition reduced the upregulation of pro-inflammatory cytokines in response to high glucose, via inhibiting the NF-kappaB signaling pathway. Glucose 112-119 high mobility group box 1 Homo sapiens 20-25 25192219-3 2015 Therefore, we hypothesized that CS promotes neutrophil necrosis with subsequent release of damage-associated molecular patterns (DAMPs), including high mobility group box 1 (HMGB1), alarming the innate immune system. Cesium 32-34 high mobility group box 1 Homo sapiens 147-172 25940319-0 2015 Retraction Note to: Nafamostat mesilate inhibits the expression of HMGB1 in lipopolysaccharide-induced acute lung injury. nafamostat 20-39 high mobility group box 1 Homo sapiens 67-72 25873535-5 2015 A Pt(II) N-heterocyclic carbene complex was found to display the characteristic hallmarks of a type II ICD inducer, namely focused oxidative endoplasmic reticulum (ER) stress, calreticulin exposure, and both HMGB1 and ATP release, and thus identified as the first small-molecule immuno-chemotherapeutic agent. carbene 24-31 high mobility group box 1 Homo sapiens 208-213 25966225-9 2015 Furthermore, HMGB1 expression level was positively correlated with 8-hydroxyguanosine, vascular cell adhesion molecule 1, nuclear factor-kB, CD3, and CD68 expression. 8-hydroxyguanosine 67-85 high mobility group box 1 Homo sapiens 13-18 25651569-10 2015 Direct activation of proximal tubular cells by uric acid resulted in (C-X-C motif) ligand 12 and high mobility group box-1 release and accelerated macrophage recruitment and the M1 phenotype. Uric Acid 47-56 high mobility group box 1 Homo sapiens 97-122 25192219-3 2015 Therefore, we hypothesized that CS promotes neutrophil necrosis with subsequent release of damage-associated molecular patterns (DAMPs), including high mobility group box 1 (HMGB1), alarming the innate immune system. Cesium 32-34 high mobility group box 1 Homo sapiens 174-179 25561283-0 2015 HMGB1 Silencing Potentiates the Anti-inflammatory Effects of Sodium Ferulate in ox-LDL-Stimulated Vascular Smooth Muscle Cells. ferulic acid 61-76 high mobility group box 1 Homo sapiens 0-5 25561283-3 2015 In addition, we also sought to determine whether HMGB1 knockdown could potentiate the anti-inflammatory effects of sodium ferulate. ferulic acid 115-130 high mobility group box 1 Homo sapiens 49-54 25817178-0 2015 Irbesartan attenuates production of high-mobility group box 1 in response to lipopolysaccharide via downregulation of interferon-beta production. Irbesartan 0-10 high mobility group box 1 Homo sapiens 36-61 25983116-0 2015 Quercetin protects necrotic insult and promotes apoptosis by attenuating the expression of RAGE and its ligand HMGB1 in human breast adenocarcinoma cells. Quercetin 0-9 high mobility group box 1 Homo sapiens 111-116 25983116-6 2015 This study was aimed to examine the biological potential of quercetin on the regulation of RAGE- and HMGB1-mediated activation of NF-kappaB and induction of apoptotic cell death in MCF-7 cells. Quercetin 60-69 high mobility group box 1 Homo sapiens 101-106 25983116-7 2015 Our findings demonstrate that quercetin inhibits the expression of RAGE and HMGB1 in MCF-7 cells. Quercetin 30-39 high mobility group box 1 Homo sapiens 76-81 25983116-9 2015 Taken together, these results suggest that quercetin plays an important role in modulating RAGE and HMGB1 signaling and induces apoptotic cell death in MCF-7 cells. Quercetin 43-52 high mobility group box 1 Homo sapiens 100-105 25726846-10 2015 Silencing of c-Jun ablated the HMGB1-induced proliferation in PASMC. pasmc 62-67 high mobility group box 1 Homo sapiens 31-36 25755254-0 2015 Carbon-ion beams induce production of an immune mediator protein, high mobility group box 1, at levels comparable with X-ray irradiation. Carbon 0-6 high mobility group box 1 Homo sapiens 66-91 25578401-5 2015 In the present study, we explored the mechanisms whereby HMGB1 regulates tumor cell autophagy during nutrient depletion (the cells were cultured in Hank"s balanced salt solution, HBSS). hank"s balanced salt solution 148-177 high mobility group box 1 Homo sapiens 57-62 25578401-5 2015 In the present study, we explored the mechanisms whereby HMGB1 regulates tumor cell autophagy during nutrient depletion (the cells were cultured in Hank"s balanced salt solution, HBSS). hbss 179-183 high mobility group box 1 Homo sapiens 57-62 25755254-2 2015 Here, we examined whether carbon-ion beams, as well as X-rays, can induce HMGB1 release from human cancer cell lines. Carbon 26-32 high mobility group box 1 Homo sapiens 74-79 25755254-8 2015 X-rays and carbon-ion beams significantly increased HMGB1 levels in the culture supernatants of A549, NCI-H460 and WiDr cells at 72 h post-irradiation with a D10 dose. Carbon 11-17 high mobility group box 1 Homo sapiens 52-57 25755254-9 2015 Furthermore, irradiation with X-rays or carbon-ion beams significantly increased HMGB1 levels in the culture supernatants of all five cell lines at 96 h post-irradiation. Carbon 40-46 high mobility group box 1 Homo sapiens 81-86 26131056-2 2015 This study was to investigate the relationship between average blood glucose level and HMGB1 level in CAD with T2DM patients. Glucose 69-76 high mobility group box 1 Homo sapiens 87-92 25769801-7 2015 In addition, synergistic NO accumulation along with direct HMGB1-LPS binding was also observed when primary microglial cultures were treated with LPS (5 ng/ml) and HMGB1 accumulated in NMDA-conditioned medium (NCM). N-Methylaspartate 185-189 high mobility group box 1 Homo sapiens 59-64 25769801-7 2015 In addition, synergistic NO accumulation along with direct HMGB1-LPS binding was also observed when primary microglial cultures were treated with LPS (5 ng/ml) and HMGB1 accumulated in NMDA-conditioned medium (NCM). N-Methylaspartate 185-189 high mobility group box 1 Homo sapiens 164-169 25769801-10 2015 Together these results show HBHP has anti-inflammatory effects, and that it inhibits synergism caused by the binding of HMGB1 and LPS. hbhp 28-32 high mobility group box 1 Homo sapiens 120-125 26131056-10 2015 Increased blood glucose levels may contribute to the increased HMGB1 levels. Glucose 16-23 high mobility group box 1 Homo sapiens 63-68 25612780-7 2015 CONCLUSIONS: Collectively, this study suggested that the HMGB1-mediated Mcl-1 transcription upregulation is a key mechanism by which autophagy protects oral cancer cells against vincristine-induced apoptosis. Vincristine 178-189 high mobility group box 1 Homo sapiens 57-62 25831509-3 2015 We have developed single-molecule assays to examine RSS binding by RAG1/2 and their cofactor high-mobility group-box protein 1 (HMGB1) as they proceed through the steps of this reaction. RSS 52-55 high mobility group box 1 Homo sapiens 128-133 25807407-0 2015 Ketamine reduces LPS-induced HMGB1 via activation of the Nrf2/HO-1 pathway and NF-kappaB suppression. Ketamine 0-8 high mobility group box 1 Homo sapiens 29-34 25612780-3 2015 RESEARCH DESIGN AND METHODS: In this study, we determined that HMGB1 released by oral cancer cells protected the cells against apoptosis caused by vincristine by upregulating the transcription of Mcl-1. Vincristine 147-158 high mobility group box 1 Homo sapiens 63-68 25612780-6 2015 HMGB1 expression inhibited blocked the Mcl-1 transcription increase and reduced Mcl-1 expression, demonstrate that HMGB1 is required for the upregulation of Mcl-1 transcriptional, and thereby maintaining Mcl-1 protein expression levels is required for the survival of oral cancer cells by vincristine. Vincristine 289-300 high mobility group box 1 Homo sapiens 0-5 25612780-6 2015 HMGB1 expression inhibited blocked the Mcl-1 transcription increase and reduced Mcl-1 expression, demonstrate that HMGB1 is required for the upregulation of Mcl-1 transcriptional, and thereby maintaining Mcl-1 protein expression levels is required for the survival of oral cancer cells by vincristine. Vincristine 289-300 high mobility group box 1 Homo sapiens 115-120 25807407-7 2015 RESULTS: Ketamine treatment dose-dependently attenuated the increased levels of proinflammatory mediators (HMGB1, nitric oxide, tumor necrosis factor alpha, and interleukin 1beta) and increased the HO-1 protein expression in LPS-activated macrophages. Ketamine 9-17 high mobility group box 1 Homo sapiens 107-112 25807407-2 2015 In the present study, we investigated whether ketamine inhibits release of high mobility group box 1 (HMGB1), a late-phase cytokine of sepsis, in lipopolysaccharide (LPS)-stimulated macrophages through heme oxygenase-1 (HO-1) induction. Ketamine 46-54 high mobility group box 1 Homo sapiens 75-100 25807407-11 2015 CONCLUSION: Ketamine inhibits the release of HMGB1 in LPS-stimulated macrophages, and this effect is at least partly mediated by the activation of the Nrf2/HO-1 pathway and NF-kappaB suppression. Ketamine 12-20 high mobility group box 1 Homo sapiens 45-50 25807407-2 2015 In the present study, we investigated whether ketamine inhibits release of high mobility group box 1 (HMGB1), a late-phase cytokine of sepsis, in lipopolysaccharide (LPS)-stimulated macrophages through heme oxygenase-1 (HO-1) induction. Ketamine 46-54 high mobility group box 1 Homo sapiens 102-107 25715249-10 2015 By contrast, the secretion of the nuclear-associated danger signal HMGB1 is independent of cysteine oxidation, as shown by experiments with a cysteine-free HMGB1 mutant. Cysteine 91-99 high mobility group box 1 Homo sapiens 67-72 25652880-5 2015 Western blot analysis and ELISA were used to assess the effects of gefitinib, an epidermal growth factor receptor inhibitor, on autophagy and HMGB1 release in BGC-823 cells. Gefitinib 67-76 high mobility group box 1 Homo sapiens 142-147 25652880-10 2015 Gefitinib increased autophagy and cytoplasmic HMGB1 release from the BGC-823 cells. Gefitinib 0-9 high mobility group box 1 Homo sapiens 46-51 25652880-11 2015 Extracellular HMGB1 in autophagic cell supernatant promoted proliferation that was abolished by glycyrrhizic acid, an HMGB1 inhibitor. Glycyrrhizic Acid 96-113 high mobility group box 1 Homo sapiens 14-19 25652880-11 2015 Extracellular HMGB1 in autophagic cell supernatant promoted proliferation that was abolished by glycyrrhizic acid, an HMGB1 inhibitor. Glycyrrhizic Acid 96-113 high mobility group box 1 Homo sapiens 118-123 25385285-3 2015 The cell proliferation, invasion, apoptosis and the gene and protein expression of HMGB1 and NF-kappaB were detected using a tetrazolium assay, Transwell cell invasion assays, a caspase-3 activity assay, western blot analysis and reverse transcription quantitative polymerase chain reaction, respectively. Tetrazolium Salts 125-136 high mobility group box 1 Homo sapiens 83-88 25582705-9 2015 Collectively, our results demonstrate that PA prevents experimental pulmonary fibrosis by modulating HMGB1/RAGE pathway. protocatechualdehyde 43-45 high mobility group box 1 Homo sapiens 101-106 25879749-8 2015 The newly generated mCRP induced platelets activation on ANCA-induced netting neutrophils, enhanced D-dimer formation, and enhanced high mobility group box 1 secretion by platelets. mcrp 20-24 high mobility group box 1 Homo sapiens 132-157 29449921-0 2015 HMGB1 bound to cisplatin-DNA adducts undergoes extensive acetylation and phosphorylation in vivo. Cisplatin 15-24 high mobility group box 1 Homo sapiens 0-5 29449921-2 2015 For many years, HMGB1 has been known to be a recognition protein for cisplatin-DNA lesions. Cisplatin 69-78 high mobility group box 1 Homo sapiens 16-21 29449921-5 2015 Interestingly, at least 4 subforms of HMGB1 bind to cisplatin-DNA adducts. Cisplatin 52-61 high mobility group box 1 Homo sapiens 38-43 25582705-0 2015 Protocatechuic aldehyde ameliorates experimental pulmonary fibrosis by modulating HMGB1/RAGE pathway. Aldehydes 15-23 high mobility group box 1 Homo sapiens 82-87 25715249-10 2015 By contrast, the secretion of the nuclear-associated danger signal HMGB1 is independent of cysteine oxidation, as shown by experiments with a cysteine-free HMGB1 mutant. Cysteine 142-150 high mobility group box 1 Homo sapiens 67-72 25452436-2 2015 SUMMARY ANSWER: HMGB1 appears to be involved in regulating inflammatory reactions in testes, as HMGB1 is translocated from testicular cells during the course of EAO and blocking its action by ethyl pyruvate (EP) reduces disease progression and spermatogenic damage. ethyl pyruvate 192-206 high mobility group box 1 Homo sapiens 16-21 25543984-0 2015 Adsorption properties of an activated carbon for 18 cytokines and HMGB1 from inflammatory model plasma. Carbon 38-44 high mobility group box 1 Homo sapiens 66-71 25543984-1 2015 The ability of an activated carbon (AC) to adsorb 18 different cytokines with molecular weights ranging from 8 kDa to 70 kDa and high mobility group box-1 (HMGB1) from inflammatory model plasma at 310 K and the mechanisms of adsorption were examined. Carbon 28-34 high mobility group box 1 Homo sapiens 129-154 25543984-1 2015 The ability of an activated carbon (AC) to adsorb 18 different cytokines with molecular weights ranging from 8 kDa to 70 kDa and high mobility group box-1 (HMGB1) from inflammatory model plasma at 310 K and the mechanisms of adsorption were examined. Carbon 28-34 high mobility group box 1 Homo sapiens 156-161 25452436-2 2015 SUMMARY ANSWER: HMGB1 appears to be involved in regulating inflammatory reactions in testes, as HMGB1 is translocated from testicular cells during the course of EAO and blocking its action by ethyl pyruvate (EP) reduces disease progression and spermatogenic damage. ethyl pyruvate 192-206 high mobility group box 1 Homo sapiens 96-101 25452436-19 2015 In support, HMGB1 triggered extracellular signal regulated kinase (ERK)1/2 and cyclic adenosine monophosphate (cAMP) response element-binding protein (CREB) activation in SC and PTC, while TM responded to HMGB1 stimulation with p38 mitogen-activated protein kinase (MAPK) and p65 nuclear factor Kappa B (NF-kB) phosphorylation followed by increased tumor necrosis factor alpha (TNF-alpha) and interleukin 6 (IL-6) mRNA levels. Cyclic AMP 79-109 high mobility group box 1 Homo sapiens 12-17 25452436-19 2015 In support, HMGB1 triggered extracellular signal regulated kinase (ERK)1/2 and cyclic adenosine monophosphate (cAMP) response element-binding protein (CREB) activation in SC and PTC, while TM responded to HMGB1 stimulation with p38 mitogen-activated protein kinase (MAPK) and p65 nuclear factor Kappa B (NF-kB) phosphorylation followed by increased tumor necrosis factor alpha (TNF-alpha) and interleukin 6 (IL-6) mRNA levels. Cyclic AMP 111-115 high mobility group box 1 Homo sapiens 12-17 25973018-12 2015 BCG induced HMGB1, IL-6, IL-10 and TNF-alpha production effectively in PMA-treated THP-1 cells. Tetradecanoylphorbol Acetate 71-74 high mobility group box 1 Homo sapiens 12-17 25434832-7 2015 Moreover, treatment of CRC cells with TRAIL or ABT-737 induces a release of endogenous HMGB1 into the extracellular space, and preincubation with glycyrrhizin, an HMGB1 inhibitor, significantly inhibits induction of cell death by TRAIL and ABT-737, suggesting that HMGB1 functionally contributes to the execution of cell death triggered by pro-apoptotic agents. ABT-737 47-54 high mobility group box 1 Homo sapiens 163-168 25434832-7 2015 Moreover, treatment of CRC cells with TRAIL or ABT-737 induces a release of endogenous HMGB1 into the extracellular space, and preincubation with glycyrrhizin, an HMGB1 inhibitor, significantly inhibits induction of cell death by TRAIL and ABT-737, suggesting that HMGB1 functionally contributes to the execution of cell death triggered by pro-apoptotic agents. Glycyrrhizic Acid 146-158 high mobility group box 1 Homo sapiens 87-92 25434832-7 2015 Moreover, treatment of CRC cells with TRAIL or ABT-737 induces a release of endogenous HMGB1 into the extracellular space, and preincubation with glycyrrhizin, an HMGB1 inhibitor, significantly inhibits induction of cell death by TRAIL and ABT-737, suggesting that HMGB1 functionally contributes to the execution of cell death triggered by pro-apoptotic agents. Glycyrrhizic Acid 146-158 high mobility group box 1 Homo sapiens 163-168 25434832-7 2015 Moreover, treatment of CRC cells with TRAIL or ABT-737 induces a release of endogenous HMGB1 into the extracellular space, and preincubation with glycyrrhizin, an HMGB1 inhibitor, significantly inhibits induction of cell death by TRAIL and ABT-737, suggesting that HMGB1 functionally contributes to the execution of cell death triggered by pro-apoptotic agents. Glycyrrhizic Acid 146-158 high mobility group box 1 Homo sapiens 163-168 25434832-5 2015 HMGB1-triggered cell death is associated with intracellular ROS release, and overexpression of Bcl-2 blocks both the increase of ROS as well as HMGB1-dependent cell death. ros 60-63 high mobility group box 1 Homo sapiens 0-5 25434832-5 2015 HMGB1-triggered cell death is associated with intracellular ROS release, and overexpression of Bcl-2 blocks both the increase of ROS as well as HMGB1-dependent cell death. ros 129-132 high mobility group box 1 Homo sapiens 0-5 25434832-6 2015 Importantly, treatment with recombinant HMGB1 or overexpression of endogenous HMGB1 strongly sensitizes CRC cells to the cytotoxic activity of the pro-apoptotic death ligand TRAIL as well as the small molecule Bcl-2 family inhibitor ABT-737. ABT-737 233-240 high mobility group box 1 Homo sapiens 40-45 25247290-9 2015 Disease control coincided with supplementary etoposide therapy initiated to boost apoptotic cell death, when systemic HMGB1 levels drastically declined and the molecule emerged mainly in its oxidized, noninflammatory isoform. Etoposide 45-54 high mobility group box 1 Homo sapiens 118-123 25434832-6 2015 Importantly, treatment with recombinant HMGB1 or overexpression of endogenous HMGB1 strongly sensitizes CRC cells to the cytotoxic activity of the pro-apoptotic death ligand TRAIL as well as the small molecule Bcl-2 family inhibitor ABT-737. ABT-737 233-240 high mobility group box 1 Homo sapiens 78-83 25434832-7 2015 Moreover, treatment of CRC cells with TRAIL or ABT-737 induces a release of endogenous HMGB1 into the extracellular space, and preincubation with glycyrrhizin, an HMGB1 inhibitor, significantly inhibits induction of cell death by TRAIL and ABT-737, suggesting that HMGB1 functionally contributes to the execution of cell death triggered by pro-apoptotic agents. ABT-737 47-54 high mobility group box 1 Homo sapiens 87-92 25434832-7 2015 Moreover, treatment of CRC cells with TRAIL or ABT-737 induces a release of endogenous HMGB1 into the extracellular space, and preincubation with glycyrrhizin, an HMGB1 inhibitor, significantly inhibits induction of cell death by TRAIL and ABT-737, suggesting that HMGB1 functionally contributes to the execution of cell death triggered by pro-apoptotic agents. ABT-737 47-54 high mobility group box 1 Homo sapiens 163-168 25377911-6 2015 Moreover, a glycyrrhizic acid derivative, a blocker of HMGB1 release, reduced UUO-mediated kidney injury and ameliorated UUO-induced renal fibrosis. Glycyrrhizic Acid 12-29 high mobility group box 1 Homo sapiens 55-60 25973309-0 2015 Reactive oxygen species regulate the differentiation of acute promyelocytic leukemia cells through HMGB1-mediated autophagy. Reactive Oxygen Species 0-23 high mobility group box 1 Homo sapiens 99-104 25973309-5 2015 HMGB1 was vital for the differentiation of ROS-mediated NB4 cells and its up-regulation promoted ATRA-induced autophagy and the degradation of PML-RARalpha. Reactive Oxygen Species 43-46 high mobility group box 1 Homo sapiens 0-5 25973309-6 2015 Furthermore, ATRA treatment elevated the levels of ROS, enhanced autophagic flux and thereby promoted cytosolic translocation of HMGB1. Tretinoin 13-17 high mobility group box 1 Homo sapiens 129-134 25973309-7 2015 HMGB1 regulated the interactions between ubiquitin-binding adaptor protein p62/SQSTM and PML-RARalpha so as to affect the degradation of PML-RARalpha during ATRA-induced autophagy. Tretinoin 157-161 high mobility group box 1 Homo sapiens 0-5 25973309-9 2015 The overall results suggested that HMGB1 is an essential regulator of ROS-induced cell differentiation. Reactive Oxygen Species 70-73 high mobility group box 1 Homo sapiens 35-40 26695754-2 2015 Ethanol exposure activates signaling pathways featuring high-mobility group box 1 and Toll-like receptor 4 (TLR4), resulting in induction of the transcription factor nuclear factor kappa-light-chain-enhancer of activated B cells, which regulates expression of several cytokine genes involved in innate immunity, and its target genes. Ethanol 0-7 high mobility group box 1 Homo sapiens 56-81 25607248-0 2015 Poly-ADP-ribosylation of HMGB1 regulates TNFSF10/TRAIL resistance through autophagy. poly-adp 0-8 high mobility group box 1 Homo sapiens 25-30 25778491-0 2015 HMGB1-mediated autophagy modulates sensitivity of colorectal cancer cells to oxaliplatin via MEK/ERK signaling pathway. Oxaliplatin 77-88 high mobility group box 1 Homo sapiens 0-5 25778491-3 2015 Administration of oxaliplatin to human CRC cells significantly enhanced the expression of HMGB1, which regulated the autophagy response and negatively regulate the cell apoptosis. Oxaliplatin 18-29 high mobility group box 1 Homo sapiens 90-95 25778491-4 2015 Moreover, a decreased oxaliplatin -induced autophagy response and an increased apoptosis level were detected in stable CRC cells harboring HMGB1 shRNA. Oxaliplatin 22-33 high mobility group box 1 Homo sapiens 139-144 25778491-6 2015 Taken together, these data suggest that HMGB1-mediated autophagy modulates sensitivity of colorectal cancer cells to oxaliplatin via MEK/ERK signaling pathway. Oxaliplatin 117-128 high mobility group box 1 Homo sapiens 40-45 25896065-10 2015 An HMGB-1 inhibitor glycyrrhizin suppressed high glucose-induced Syk activation. Glycyrrhizic Acid 20-32 high mobility group box 1 Homo sapiens 3-9 25896065-10 2015 An HMGB-1 inhibitor glycyrrhizin suppressed high glucose-induced Syk activation. Glucose 49-56 high mobility group box 1 Homo sapiens 3-9 25896065-9 2015 High-mobility group box-1 (HMGB-1), an endogenous activator of TLR4, rapidly increased in TLR4 immunoprecipitates upon high glucose stimulation, and this association was reduced by N-acetylcysteine, an antioxidant. Glucose 124-131 high mobility group box 1 Homo sapiens 0-25 25896065-12 2015 High glucose induces an immediate, reactive oxygen species-dependent, extracellular release of HMGB-1 which binds to TLR4 and activates it, leading to Syk activation. Glucose 5-12 high mobility group box 1 Homo sapiens 95-101 25896065-9 2015 High-mobility group box-1 (HMGB-1), an endogenous activator of TLR4, rapidly increased in TLR4 immunoprecipitates upon high glucose stimulation, and this association was reduced by N-acetylcysteine, an antioxidant. Glucose 124-131 high mobility group box 1 Homo sapiens 27-33 25896065-12 2015 High glucose induces an immediate, reactive oxygen species-dependent, extracellular release of HMGB-1 which binds to TLR4 and activates it, leading to Syk activation. Reactive Oxygen Species 35-58 high mobility group box 1 Homo sapiens 95-101 25896065-9 2015 High-mobility group box-1 (HMGB-1), an endogenous activator of TLR4, rapidly increased in TLR4 immunoprecipitates upon high glucose stimulation, and this association was reduced by N-acetylcysteine, an antioxidant. Acetylcysteine 181-197 high mobility group box 1 Homo sapiens 0-25 26299081-13 2015 In OA patients, HMGB-1 in the KL2/3 group was higher than in the KL4 group. adenosine-2',3'-vanadate 30-33 high mobility group box 1 Homo sapiens 16-22 25896065-9 2015 High-mobility group box-1 (HMGB-1), an endogenous activator of TLR4, rapidly increased in TLR4 immunoprecipitates upon high glucose stimulation, and this association was reduced by N-acetylcysteine, an antioxidant. Acetylcysteine 181-197 high mobility group box 1 Homo sapiens 27-33 26299081-13 2015 In OA patients, HMGB-1 in the KL2/3 group was higher than in the KL4 group. sinapultide 65-68 high mobility group box 1 Homo sapiens 16-22 26260962-7 2015 Moreover, recent studies have described models of epilepsy induced by the administration of bicuculline and kainic acid that highlight the nature of HMGB1-TLR4 interactions, their intracellular signaling pathway as well as their role in ictiogenesis and epileptic recurrence.The aim of our review is to focus on different branches of innate immunity and their role in epilepsy, emphasizing the role of immune related molecules in epileptogenesis and highlighting the research implications for novel therapeutic strategies. Bicuculline 92-103 high mobility group box 1 Homo sapiens 149-154 26260962-7 2015 Moreover, recent studies have described models of epilepsy induced by the administration of bicuculline and kainic acid that highlight the nature of HMGB1-TLR4 interactions, their intracellular signaling pathway as well as their role in ictiogenesis and epileptic recurrence.The aim of our review is to focus on different branches of innate immunity and their role in epilepsy, emphasizing the role of immune related molecules in epileptogenesis and highlighting the research implications for novel therapeutic strategies. Kainic Acid 108-119 high mobility group box 1 Homo sapiens 149-154 27163120-5 2014 Combining CMT-3 with TSN-SS led to enhanced accumulation of endogenous LC3-II, but reduced HMGB1 cytoplasmic translocation. tanshinone II A sodium sulfonate 21-27 high mobility group box 1 Homo sapiens 91-96 25311659-0 2015 Interaction of adriamycin with a regulatory element of hmgb1: spectroscopic and calorimetric approach. Doxorubicin 15-25 high mobility group box 1 Homo sapiens 55-60 25311659-3 2015 Because of the clear association between HMGB1 and cancer, we studied the binding of adriamycin (ADM), a well-known anticancer drug with the promoter region (-165 to -183) of hmgb1 by using a variety of spectroscopic, calorimetric techniques, and in-silico molecular modeling. Doxorubicin 85-95 high mobility group box 1 Homo sapiens 175-180 25311659-3 2015 Because of the clear association between HMGB1 and cancer, we studied the binding of adriamycin (ADM), a well-known anticancer drug with the promoter region (-165 to -183) of hmgb1 by using a variety of spectroscopic, calorimetric techniques, and in-silico molecular modeling. Doxorubicin 97-100 high mobility group box 1 Homo sapiens 175-180 24940804-4 2015 Proteomic analysis showed that SIRT1 deacetylates HMGB1 at four lysine residues (55, 88, 90, and 177) within the proinflammatory and nuclear localization signal domains of HMGB1. Lysine 64-70 high mobility group box 1 Homo sapiens 50-55 24940804-4 2015 Proteomic analysis showed that SIRT1 deacetylates HMGB1 at four lysine residues (55, 88, 90, and 177) within the proinflammatory and nuclear localization signal domains of HMGB1. Lysine 64-70 high mobility group box 1 Homo sapiens 172-177 24940804-7 2015 Conversely, resveratrol pretreatment led to decreased HMGB1 acetylation, its nuclear retention, decreased systemic release, and reduced tubular damage. Resveratrol 12-23 high mobility group box 1 Homo sapiens 54-59 25512109-8 2014 The effect of an HMGB1 neutralizing antibody on Dox resistance induced by extracellular HMGB1 from non-viable Dox-treated cancer cells or recombinant HMGB1 was also investigated. Doxorubicin 48-51 high mobility group box 1 Homo sapiens 17-22 25512109-8 2014 The effect of an HMGB1 neutralizing antibody on Dox resistance induced by extracellular HMGB1 from non-viable Dox-treated cancer cells or recombinant HMGB1 was also investigated. Doxorubicin 48-51 high mobility group box 1 Homo sapiens 88-93 25512109-8 2014 The effect of an HMGB1 neutralizing antibody on Dox resistance induced by extracellular HMGB1 from non-viable Dox-treated cancer cells or recombinant HMGB1 was also investigated. Doxorubicin 48-51 high mobility group box 1 Homo sapiens 88-93 25512109-11 2014 Intracellular HMGB1 expression was induced in BCF-CM-treated breast cancer cells and also in Dox-treated cells. Doxorubicin 93-96 high mobility group box 1 Homo sapiens 14-19 25512109-12 2014 Extracellular HMGB1 was strongly expressed in the CM after Dox-induced MDA-MB-231 cell death and was higher in cells pre-treated with BCF-CM than NTF-CM. Doxorubicin 59-62 high mobility group box 1 Homo sapiens 14-19 25512109-14 2014 Recombinant HMGB1 was shown to increase Dox resistance and this was associated with evidence of autophagy. Doxorubicin 40-43 high mobility group box 1 Homo sapiens 12-17 25512109-15 2014 Anti-HMGB1 neutralizing antibody significantly reduced the effect of extracellular HMGB1 released from dying cancer cells or of recombinant HMGB1 on Dox resistance. Doxorubicin 149-152 high mobility group box 1 Homo sapiens 5-10 25512109-15 2014 Anti-HMGB1 neutralizing antibody significantly reduced the effect of extracellular HMGB1 released from dying cancer cells or of recombinant HMGB1 on Dox resistance. Doxorubicin 149-152 high mobility group box 1 Homo sapiens 83-88 25512109-15 2014 Anti-HMGB1 neutralizing antibody significantly reduced the effect of extracellular HMGB1 released from dying cancer cells or of recombinant HMGB1 on Dox resistance. Doxorubicin 149-152 high mobility group box 1 Homo sapiens 83-88 25499383-8 2014 In the overall comparison, reduction of both SBP and DBP with eplerenone was greater than other antihypertensive agents (WMD for SBP -1.50 mm Hg, p < 0.0001; WMD for DBP -0.54 mm Hg, p < 0.00001); this was essentially driven by a greater anti-hypertensive action vs enalapril and losartan for SBP and vs losartan for DBP. Eplerenone 62-72 high mobility group box 1 Homo sapiens 129-135 25864748-4 2015 This study was undertaken to analyze whether disrupting the microtubule cytoskeleton by colchicine modulates transcriptional levels of MEFV, NF-kappaB p65, NLRP3, HMGB1, and caspase-3 in neutrophils from patients with familial Mediterranean fever (FMF) and healthy subjects. Colchicine 88-98 high mobility group box 1 Homo sapiens 163-168 26226834-5 2015 BQ123 and FAK inhibitor or shRNA, but not BQ788, completely abolished the promoting effect of ET-1 on the expression of HMGB1. cyclo(Trp-Asp-Pro-Val-Leu) 0-5 high mobility group box 1 Homo sapiens 120-125 26226834-7 2015 In the presence of the transcription inhibitor actinomycin D, the HMGB1 mRNA level markedly decreased over time, and ET-1 dose-dependently rescued the HMGB1 mRNA level. Dactinomycin 47-60 high mobility group box 1 Homo sapiens 66-71 26226834-7 2015 In the presence of the transcription inhibitor actinomycin D, the HMGB1 mRNA level markedly decreased over time, and ET-1 dose-dependently rescued the HMGB1 mRNA level. Dactinomycin 47-60 high mobility group box 1 Homo sapiens 151-156 25549101-7 2014 In starvation-stressed TBP/Q36 and TBP/Q79 cells, increased reactive oxygen species generation accelerated the cytoplasmic translocation of HMGB1, which accompanied autophagy activation. Oxygen 69-75 high mobility group box 1 Homo sapiens 140-145 25926862-9 2014 The HMGB1 levels were higher in patients with infection during the clinical course, the HMGB1 levels in non-survivors were higher than those in survivors, and positively correlated with DAO activity, L/M ratio, the concentration of endotoxin (R = 0.484, P <0.01). lauric acid 186-189 high mobility group box 1 Homo sapiens 88-93 27163120-7 2014 As an ovel candidate for cancer therapy, CMT-3 may be used in combination with TSN-SS, which possibly facilitates the execution of a death signal (e.g. autophagy) and prevents the survival of an inducer (e.g. HMGB1 cytoplasmic translocation), thus improving its therapeutic effect. tanshinone II A sodium sulfonate 79-85 high mobility group box 1 Homo sapiens 209-214 24780928-9 2014 We found that different intracellular sources of ROS are triggered by contact allergens, and an important role for HMGB1 in chemical allergen-induced IL-18 production was demonstrated. Reactive Oxygen Species 49-52 high mobility group box 1 Homo sapiens 115-120 25014009-3 2014 The degree to which HMGB1 activates a receptor is thought to be dependent upon the oxidative state of the ligand, resulting in the functional isoforms of all-thiol HMGB1 (at-HMGB1) acting through RAGE, and disufide HMGB1 (ds-HMGB1) interacting with TLR4. Sulfhydryl Compounds 158-163 high mobility group box 1 Homo sapiens 20-25 25014009-3 2014 The degree to which HMGB1 activates a receptor is thought to be dependent upon the oxidative state of the ligand, resulting in the functional isoforms of all-thiol HMGB1 (at-HMGB1) acting through RAGE, and disufide HMGB1 (ds-HMGB1) interacting with TLR4. Sulfhydryl Compounds 158-163 high mobility group box 1 Homo sapiens 164-169 25014009-3 2014 The degree to which HMGB1 activates a receptor is thought to be dependent upon the oxidative state of the ligand, resulting in the functional isoforms of all-thiol HMGB1 (at-HMGB1) acting through RAGE, and disufide HMGB1 (ds-HMGB1) interacting with TLR4. Sulfhydryl Compounds 158-163 high mobility group box 1 Homo sapiens 164-169 25014009-3 2014 The degree to which HMGB1 activates a receptor is thought to be dependent upon the oxidative state of the ligand, resulting in the functional isoforms of all-thiol HMGB1 (at-HMGB1) acting through RAGE, and disufide HMGB1 (ds-HMGB1) interacting with TLR4. Sulfhydryl Compounds 158-163 high mobility group box 1 Homo sapiens 164-169 25014009-3 2014 The degree to which HMGB1 activates a receptor is thought to be dependent upon the oxidative state of the ligand, resulting in the functional isoforms of all-thiol HMGB1 (at-HMGB1) acting through RAGE, and disufide HMGB1 (ds-HMGB1) interacting with TLR4. Sulfhydryl Compounds 158-163 high mobility group box 1 Homo sapiens 164-169 25014009-3 2014 The degree to which HMGB1 activates a receptor is thought to be dependent upon the oxidative state of the ligand, resulting in the functional isoforms of all-thiol HMGB1 (at-HMGB1) acting through RAGE, and disufide HMGB1 (ds-HMGB1) interacting with TLR4. disufide 206-214 high mobility group box 1 Homo sapiens 20-25 25014009-3 2014 The degree to which HMGB1 activates a receptor is thought to be dependent upon the oxidative state of the ligand, resulting in the functional isoforms of all-thiol HMGB1 (at-HMGB1) acting through RAGE, and disufide HMGB1 (ds-HMGB1) interacting with TLR4. disufide 206-214 high mobility group box 1 Homo sapiens 164-169 25014009-3 2014 The degree to which HMGB1 activates a receptor is thought to be dependent upon the oxidative state of the ligand, resulting in the functional isoforms of all-thiol HMGB1 (at-HMGB1) acting through RAGE, and disufide HMGB1 (ds-HMGB1) interacting with TLR4. disufide 206-214 high mobility group box 1 Homo sapiens 164-169 25014009-3 2014 The degree to which HMGB1 activates a receptor is thought to be dependent upon the oxidative state of the ligand, resulting in the functional isoforms of all-thiol HMGB1 (at-HMGB1) acting through RAGE, and disufide HMGB1 (ds-HMGB1) interacting with TLR4. disufide 206-214 high mobility group box 1 Homo sapiens 164-169 25014009-3 2014 The degree to which HMGB1 activates a receptor is thought to be dependent upon the oxidative state of the ligand, resulting in the functional isoforms of all-thiol HMGB1 (at-HMGB1) acting through RAGE, and disufide HMGB1 (ds-HMGB1) interacting with TLR4. disufide 206-214 high mobility group box 1 Homo sapiens 164-169 25052363-11 2014 A positive correlation was found between urinary HMGB1/creatinine ratio and CD4(+) T cells/creatinine ratio (P = 0 028) and effector memory T cells/creatinine ratio (P = 0 039) in urine. Creatinine 55-65 high mobility group box 1 Homo sapiens 49-54 25052363-11 2014 A positive correlation was found between urinary HMGB1/creatinine ratio and CD4(+) T cells/creatinine ratio (P = 0 028) and effector memory T cells/creatinine ratio (P = 0 039) in urine. Creatinine 91-101 high mobility group box 1 Homo sapiens 49-54 25052363-11 2014 A positive correlation was found between urinary HMGB1/creatinine ratio and CD4(+) T cells/creatinine ratio (P = 0 028) and effector memory T cells/creatinine ratio (P = 0 039) in urine. Creatinine 91-101 high mobility group box 1 Homo sapiens 49-54 25419374-0 2014 Intraoperative systemic lidocaine inhibits the expression of HMGB1 in patients undergoing radical hysterectomy. Lidocaine 24-33 high mobility group box 1 Homo sapiens 61-66 25004063-0 2014 Novel role of resveratrol: suppression of high-mobility group protein box 1 nucleocytoplasmic translocation by the upregulation of sirtuin 1 in sepsis-induced liver injury. Resveratrol 14-25 high mobility group box 1 Homo sapiens 42-75 25004063-2 2014 We hypothesized that resveratrol (RESV) administration would inhibit nuclear-cytoplasmic HMGB1 translocation in hepatocytes, which is associated with sirtuin 1 (SIRT1) upregulation. Resveratrol 21-32 high mobility group box 1 Homo sapiens 89-94 25004063-2 2014 We hypothesized that resveratrol (RESV) administration would inhibit nuclear-cytoplasmic HMGB1 translocation in hepatocytes, which is associated with sirtuin 1 (SIRT1) upregulation. Resveratrol 34-38 high mobility group box 1 Homo sapiens 89-94 25004063-9 2014 Resveratrol inhibited HMGB1 cytoplasmic translocation. Resveratrol 0-11 high mobility group box 1 Homo sapiens 22-27 25004063-10 2014 Nicotinamide, an SIRT1 inhibitor, reduced the SIRT1-mediated suppression of HMGB1 translocation and aggravated cecal ligation and puncture-induced liver damage. Niacinamide 0-12 high mobility group box 1 Homo sapiens 76-81 25004063-13 2014 CONCLUSIONS: Resveratrol protects against sepsis-induced liver injury through the SIRT1-mediated HMGB1 nucleocytoplasmic translocation pathway, a new potential therapeutic target in sepsis-induced liver injury. Resveratrol 13-24 high mobility group box 1 Homo sapiens 97-102 25419374-4 2014 The present study sought to determine whether intraoperative systemic lidocaine could attenuate the level of HMGB1 by inhibiting it expression in PBMC from patients underwent radical hysterectomy. Lidocaine 70-79 high mobility group box 1 Homo sapiens 109-114 25419374-10 2014 The result showed that lidocaine not only attenuated the level of HMGB1 protein in serum and supernatant, but inhibited the transcription of HMGB1 mRAN in PBMC. Lidocaine 23-32 high mobility group box 1 Homo sapiens 66-71 25419374-10 2014 The result showed that lidocaine not only attenuated the level of HMGB1 protein in serum and supernatant, but inhibited the transcription of HMGB1 mRAN in PBMC. Lidocaine 23-32 high mobility group box 1 Homo sapiens 141-146 25419374-11 2014 The present study of us demonstrated that intraoperative systemic lidocaine can attenuate the level of HMGB1 and inhibit its expression in PBMC from patients underwent radical hysterectomy. Lidocaine 66-75 high mobility group box 1 Homo sapiens 103-108 25419374-12 2014 Therefore, lidocaine may play an important role in many other clinical diseases by inhibiting HMGB1. Lidocaine 11-20 high mobility group box 1 Homo sapiens 94-99 25126737-2 2014 Increasing evidence demonstrates that HMGB1-dependent autophagy promotes chemotherapy resistance, sustains tumor metabolism requirements and T cell survival, prevents polyglutamine aggregates and excitotoxicity, and protects against endotoxemia, bacterial infection, and ischemia-reperfusion injury in vitro or in vivo. polyglutamine 167-180 high mobility group box 1 Homo sapiens 38-43 25303153-6 2014 HMGB1 was increased in the supernatants of cells exposed to 30 mM and 11.2 mM glucose compared to control. Glucose 78-85 high mobility group box 1 Homo sapiens 0-5 25572734-6 2014 Our results indicated that the positive expression of HMGB1 was significantly increased in the nucleus of gastric cancer tissues compared with the adjacent non-cancerous tissues (ANCT) (64.0&#x0025; vs 44.0&#x0025;, P=0.025), but was not linked to the clinicopathologic features, including the TNM stage (P=0.533) and metastatic lymph node (P=0.771), in patients with gastric cancer. Adenosine Monophosphate 191-194 high mobility group box 1 Homo sapiens 54-59 25014664-7 2014 We also show that HMGB1 is a direct target of miR-let-7f-1 and forced expression of HMGB1 cDNA enhanced cisplatin sensitivity in SPARC expressed cells. Cisplatin 104-113 high mobility group box 1 Homo sapiens 18-23 25014664-7 2014 We also show that HMGB1 is a direct target of miR-let-7f-1 and forced expression of HMGB1 cDNA enhanced cisplatin sensitivity in SPARC expressed cells. Cisplatin 104-113 high mobility group box 1 Homo sapiens 84-89 25014664-8 2014 In summary, our results suggest that SPARC modulates cisplatin resistance by modulating the Let-7f-1 miRNA/HMGB1 axis in medulloblastoma cells. Cisplatin 53-62 high mobility group box 1 Homo sapiens 107-112 24706001-5 2014 First, we monitored the effects of post-treatment fisetin on lipopolysaccharide (LPS) and cecal ligation and puncture (CLP)-mediated release of HMGB1 and HMGB1-mediated regulation of pro-inflammatory responses in human umbilical vein endothelial cells (HUVECs) and septic mice. fisetin 50-57 high mobility group box 1 Homo sapiens 144-149 24706001-5 2014 First, we monitored the effects of post-treatment fisetin on lipopolysaccharide (LPS) and cecal ligation and puncture (CLP)-mediated release of HMGB1 and HMGB1-mediated regulation of pro-inflammatory responses in human umbilical vein endothelial cells (HUVECs) and septic mice. fisetin 50-57 high mobility group box 1 Homo sapiens 154-159 25572734-6 2014 Our results indicated that the positive expression of HMGB1 was significantly increased in the nucleus of gastric cancer tissues compared with the adjacent non-cancerous tissues (ANCT) (64.0&#x0025; vs 44.0&#x0025;, P=0.025), but was not linked to the clinicopathologic features, including the TNM stage (P=0.533) and metastatic lymph node (P=0.771), in patients with gastric cancer. Adenosine Monophosphate 211-214 high mobility group box 1 Homo sapiens 54-59 24970310-6 2014 To explore the possible mechanisms, the HMGB1 receptor for advanced glycation endproducts (RAGE) in the NS/PCs was blocked with anti-RAGE antibody, and c-Jun N-terminal protein kinase (JNK) in the NS/PCs was inhibited using the potent JNK inhibitor, SP600125. pyrazolanthrone 250-258 high mobility group box 1 Homo sapiens 40-45 25062563-0 2014 Heparin attenuates HMGB1 expression in arterial tissue subjected to limb ischemia/reperfusion. Heparin 0-7 high mobility group box 1 Homo sapiens 19-24 25188497-11 2014 We found significantly increased serum concentrations of sRAGE, esRAGE and HMGB1 in CTEPH. cteph 84-89 high mobility group box 1 Homo sapiens 75-80 24942766-7 2014 During Period III, uptitrated patients showed further BP reductions, which were largest in those on olmesartan/amlodipine 40/10 mg. SI values increased in uptitrated patients and were highest with olmesartan/amlodipine 40/10 mg (SBP 1.62/DBP 1.41). olmesartan 197-207 high mobility group box 1 Homo sapiens 229-234 25060178-10 2014 Therefore, the combination of a HMGB1+ and HMGB2- expression potentially predicts a poor prognosis in patients with PDAC, and these new biomarkers may be useful parameters for clinical management in the early postoperative phase. pdac 116-120 high mobility group box 1 Homo sapiens 32-37 25126750-4 2014 Tube formations by human microvascular endothelial cells (HMVECs) were significantly increased by direct exposure to HMGB1 or to conditioned medium derived from HMGB1-stimulated RA fibroblasts, and these increases were attenuated by cilostazol, the latter of which was blocked by sirtinol. Cilostazol 233-243 high mobility group box 1 Homo sapiens 117-122 24477600-8 2014 Moreover, adenosine inhibited thrombin-induced elevated expression of proinflammatory cytokines, IL-6 and HMGB-1; and chemokines, MCP-1, CXCL-1, and CXCL-3. Adenosine 10-19 high mobility group box 1 Homo sapiens 106-112 25140900-4 2014 The goal of the present work was to investigate if Rose Bengal Acetate (RBAc), a powerful PS able to trigger apoptosis and autophagy, enables photosensitized HeLa cells to expose and/or release pivotal DAMPs, i.e. ATP, HSP70, HSP90, HMGB1, and calreticulin (CRT), that characterize ICD. Rose Bengal acetate 51-70 high mobility group box 1 Homo sapiens 233-238 25140900-4 2014 The goal of the present work was to investigate if Rose Bengal Acetate (RBAc), a powerful PS able to trigger apoptosis and autophagy, enables photosensitized HeLa cells to expose and/or release pivotal DAMPs, i.e. ATP, HSP70, HSP90, HMGB1, and calreticulin (CRT), that characterize ICD. Rose Bengal acetate 72-76 high mobility group box 1 Homo sapiens 233-238 25140900-4 2014 The goal of the present work was to investigate if Rose Bengal Acetate (RBAc), a powerful PS able to trigger apoptosis and autophagy, enables photosensitized HeLa cells to expose and/or release pivotal DAMPs, i.e. ATP, HSP70, HSP90, HMGB1, and calreticulin (CRT), that characterize ICD. Adenosine Triphosphate 214-217 high mobility group box 1 Homo sapiens 233-238 24928512-12 2014 In serum from ethanol-fed mice and from patients with ALD, there was disulfide-bonded hyperacetylated HMGB1, disulfide-bonded non-acetylated HMGB1, and HMGB1 phosphorylated in serine 35. Ethanol 14-21 high mobility group box 1 Homo sapiens 102-107 24928512-12 2014 In serum from ethanol-fed mice and from patients with ALD, there was disulfide-bonded hyperacetylated HMGB1, disulfide-bonded non-acetylated HMGB1, and HMGB1 phosphorylated in serine 35. Ethanol 14-21 high mobility group box 1 Homo sapiens 141-146 24928512-12 2014 In serum from ethanol-fed mice and from patients with ALD, there was disulfide-bonded hyperacetylated HMGB1, disulfide-bonded non-acetylated HMGB1, and HMGB1 phosphorylated in serine 35. Ethanol 14-21 high mobility group box 1 Homo sapiens 141-146 24928512-12 2014 In serum from ethanol-fed mice and from patients with ALD, there was disulfide-bonded hyperacetylated HMGB1, disulfide-bonded non-acetylated HMGB1, and HMGB1 phosphorylated in serine 35. Disulfides 69-78 high mobility group box 1 Homo sapiens 102-107 25126750-4 2014 Tube formations by human microvascular endothelial cells (HMVECs) were significantly increased by direct exposure to HMGB1 or to conditioned medium derived from HMGB1-stimulated RA fibroblasts, and these increases were attenuated by cilostazol, the latter of which was blocked by sirtinol. Cilostazol 233-243 high mobility group box 1 Homo sapiens 161-166 25126750-4 2014 Tube formations by human microvascular endothelial cells (HMVECs) were significantly increased by direct exposure to HMGB1 or to conditioned medium derived from HMGB1-stimulated RA fibroblasts, and these increases were attenuated by cilostazol, the latter of which was blocked by sirtinol. sirtinol 280-288 high mobility group box 1 Homo sapiens 161-166 24996221-0 2014 HMGB1-mediated autophagy promotes docetaxel resistance in human lung adenocarcinoma. Docetaxel 34-43 high mobility group box 1 Homo sapiens 0-5 24905701-0 2014 Genomic analysis and differential expression of HMG and S100A family in human arthritis: upregulated expression of chemokines, IL-8 and nitric oxide by HMGB1. Nitric Oxide 136-148 high mobility group box 1 Homo sapiens 152-157 24905701-11 2014 The recombinant HMGB1 utilized in this study shows properties that are similar to disulfide-HMGB1. Disulfides 82-91 high mobility group box 1 Homo sapiens 16-21 24905701-11 2014 The recombinant HMGB1 utilized in this study shows properties that are similar to disulfide-HMGB1. Disulfides 82-91 high mobility group box 1 Homo sapiens 92-97 24863048-0 2014 Evaluation of in vitro properties of predicted kinases that phosphorylate serine residues within nuclear localization signal 1 of high mobility group box 1. Serine 74-80 high mobility group box 1 Homo sapiens 130-155 24863048-5 2014 Among the predicted kinases, protein kinase C phosphorylated Ser(46) of HMGB1-derived peptides, and a mutagenesis experiment confirmed that phosphorylation at this site could induce the translocation of the N-terminal region of NLS1-containing HMGB1 into the cytosol. Serine 61-64 high mobility group box 1 Homo sapiens 72-77 24863048-5 2014 Among the predicted kinases, protein kinase C phosphorylated Ser(46) of HMGB1-derived peptides, and a mutagenesis experiment confirmed that phosphorylation at this site could induce the translocation of the N-terminal region of NLS1-containing HMGB1 into the cytosol. Serine 61-64 high mobility group box 1 Homo sapiens 244-249 24983978-7 2014 RESULTS: Fecal HMGB1 increased by 5-, 11-, 18-, and 24-folds with dextran sodium sulphate doses of 0.25%, 0.50%, 1%, and 4%, respectively, showing that the protein detected a high-grade and a subclinical inflammation. dextran sodium sulphate 66-89 high mobility group box 1 Homo sapiens 15-20 24838617-7 2014 Using the potent PI3K/Akt inhibitor, LY294002, we explored the likely mechanism of HMGB1-induced NS/PC proliferation. 2-(4-morpholinyl)-8-phenyl-4H-1-benzopyran-4-one 37-45 high mobility group box 1 Homo sapiens 83-88 24838617-10 2014 LY294002 effectively reduced phospho-Akt levels and reduced NS/PC proliferation induced by HMGB1 in vitro. 2-(4-morpholinyl)-8-phenyl-4H-1-benzopyran-4-one 0-8 high mobility group box 1 Homo sapiens 91-96 24996221-8 2014 RESULTS: HMGB1 translocated from the nucleus to the cytoplasm in LAD cells exposed to docetaxel and acted as a positive regulator of autophagy, which inhibited apoptosis and increased drug resistance. Docetaxel 86-95 high mobility group box 1 Homo sapiens 9-14 24996221-9 2014 Suppression of HMGB1 restored the sensitivity of LAD cells to docetaxel both in vivo and in vitro. Docetaxel 62-71 high mobility group box 1 Homo sapiens 15-20 24996221-11 2014 CONCLUSIONS: Our results demonstrated that HMGB1-regulated autophagy is a significant contributor to docetaxel resistance in LAD cells. Docetaxel 101-110 high mobility group box 1 Homo sapiens 43-48 24996221-12 2014 Suppression of HMGB1 or limiting HMGB1 cytosolic translocation diminished autophagic protection in response to docetaxel in LAD cells. Docetaxel 111-120 high mobility group box 1 Homo sapiens 15-20 24996221-12 2014 Suppression of HMGB1 or limiting HMGB1 cytosolic translocation diminished autophagic protection in response to docetaxel in LAD cells. Docetaxel 111-120 high mobility group box 1 Homo sapiens 33-38 25182828-0 2014 [Serum levels and clinical significance of high mobility group protein B1 in patients with delayed encephalopathy after acute carbon monoxide poisoning]. Carbon Monoxide 126-141 high mobility group box 1 Homo sapiens 43-73 24773263-4 2014 The mean (SD) levels of the nuclear protein plasma high-mobility group box 1 increased significantly from 5.1 (2.2) ng ml(-1) during (16.6 (7.3) ng ml(-1) ) and after (14.3 (8.2) ng ml(-1) ) cardiopulmonary bypass in the saline group. Sodium Chloride 221-227 high mobility group box 1 Homo sapiens 51-76 24359220-3 2014 RECENT ADVANCES: Low levels of ROS modify Atg4 and high mobility group box 1 (HMGB1) proteins, activate AMP-activated protein kinase (AMPK) and apoptosis signal-regulating kinase/c-Jun N-terminal kinase (JNK) pathways, or transactivate various proteins that could upregulate autophagy, leading to reductions in apoptosis. Reactive Oxygen Species 31-34 high mobility group box 1 Homo sapiens 51-76 24359220-3 2014 RECENT ADVANCES: Low levels of ROS modify Atg4 and high mobility group box 1 (HMGB1) proteins, activate AMP-activated protein kinase (AMPK) and apoptosis signal-regulating kinase/c-Jun N-terminal kinase (JNK) pathways, or transactivate various proteins that could upregulate autophagy, leading to reductions in apoptosis. Reactive Oxygen Species 31-34 high mobility group box 1 Homo sapiens 78-83 25080914-8 2014 The MTT assay demonstrated that HMGB1 knockdown significantly reduced the cell proliferation. monooxyethylene trimethylolpropane tristearate 4-7 high mobility group box 1 Homo sapiens 32-37 25182828-1 2014 OBJECTIVE: To investigate the changes in serum level of high mobility group protein B1 (HMGB1) in patients with delayed encephalopathy after acute carbon monoxide poisoning and the clinical significance of these changes. Carbon Monoxide 147-162 high mobility group box 1 Homo sapiens 56-86 25182828-1 2014 OBJECTIVE: To investigate the changes in serum level of high mobility group protein B1 (HMGB1) in patients with delayed encephalopathy after acute carbon monoxide poisoning and the clinical significance of these changes. Carbon Monoxide 147-162 high mobility group box 1 Homo sapiens 88-93 25182828-5 2014 RESULTS: In the acute stage of carbon monoxide poisoning, the serum HMGB1 level of delayed encephalopathy group was significantly higher than those of the carbon monoxide poisoning group and the control group (P < 0.01). Carbon Monoxide 31-46 high mobility group box 1 Homo sapiens 68-73 25182828-5 2014 RESULTS: In the acute stage of carbon monoxide poisoning, the serum HMGB1 level of delayed encephalopathy group was significantly higher than those of the carbon monoxide poisoning group and the control group (P < 0.01). Carbon Monoxide 155-170 high mobility group box 1 Homo sapiens 68-73 25182828-8 2014 CONCLUSION: HMGB1, as an important late mediator of inflammation, is involved in the inflammatory reaction in delayed encephalopathy, and is positively correlated with HDS and ADL scores, indicating that it can be used as one of the major indicators in monitoring carbon monoxide poisoning. Carbon Monoxide 264-279 high mobility group box 1 Homo sapiens 12-17 24684392-1 2014 The aim of the present study was to investigate the association of genetic polymorphisms in high mobility group box 1 and 2 (HMGB1 and HMGB2, respectively) with platinum-based chemotherapy responses in Chinese lung cancer patients. Platinum 161-169 high mobility group box 1 Homo sapiens 92-123 24684392-1 2014 The aim of the present study was to investigate the association of genetic polymorphisms in high mobility group box 1 and 2 (HMGB1 and HMGB2, respectively) with platinum-based chemotherapy responses in Chinese lung cancer patients. Platinum 161-169 high mobility group box 1 Homo sapiens 125-130 24684392-5 2014 We found that rs1412125 and rs2249825 of HMGB1 were significantly associated with the platinum-based chemotherapy response in both recessive and genotypic models. Platinum 86-94 high mobility group box 1 Homo sapiens 41-46 24684392-8 2014 The HMGB1 SNPs (rs1412125 and rs2249825) were associated with platinum-based chemotherapy responses in Chinese lung cancer patients. Platinum 62-70 high mobility group box 1 Homo sapiens 4-9 24684392-9 2014 In conclusion, HMGB1 SNPs may serve as potential biomarkers for predicting the efficacy of platinum-based chemotherapy. Platinum 91-99 high mobility group box 1 Homo sapiens 15-20 24581270-0 2014 Andrographolide inhibits HMGB1-induced inflammatory responses in human umbilical vein endothelial cells and in murine polymicrobial sepsis. andrographolide 0-15 high mobility group box 1 Homo sapiens 25-30 24467264-9 2014 In cultured endothelial cells, human kallistatin via its heparin-binding site inhibited HMGB1-induced nuclear factor-kappaB activation and inflammatory gene expression. Heparin 57-64 high mobility group box 1 Homo sapiens 88-93 24965297-1 2014 Intraperitoneal glucose degradation products (GDP) load influences systemic advanced glycation end products (AGEs) but the effects on soluble receptor for AGEs (s-RAGE) and its proinflammatory ligands: extracellular newly identified receptor for advanced glycation end-products binding protein(EN-RAGE) and high mobility group box-1 protein (HMGB-1) are unknown. Glucose 16-23 high mobility group box 1 Homo sapiens 307-332 24965297-1 2014 Intraperitoneal glucose degradation products (GDP) load influences systemic advanced glycation end products (AGEs) but the effects on soluble receptor for AGEs (s-RAGE) and its proinflammatory ligands: extracellular newly identified receptor for advanced glycation end-products binding protein(EN-RAGE) and high mobility group box-1 protein (HMGB-1) are unknown. Glucose 16-23 high mobility group box 1 Homo sapiens 342-348 24965297-9 2014 A lower intraperitoneal GDP load is associated with decreased plasma levels of EN-RAGE and HMGB-1. Guanosine Diphosphate 24-27 high mobility group box 1 Homo sapiens 91-97 24574398-7 2014 That and the pattern of viral determinants maintained in gemcitabine-treated cells suggested the activation of an inflammasome/caspase 1 (CASP1) platform alongside DNA detachment and/or nuclear exclusion of HMGB1 during early stages of the viral life cycle. gemcitabine 57-68 high mobility group box 1 Homo sapiens 207-212 24468889-4 2014 METHODS: We investigated the effects of PL on HMGB1-mediated inflammatory response by monitoring the effects of PL on lipopolysaccharide or cecal ligation and puncture (CLP)-mediated release of HMGB1, as well as on the modulation of HMGB1-mediated inflammatory responses. piperlonguminine 40-42 high mobility group box 1 Homo sapiens 46-51 24468889-7 2014 In addition, treatment with PL reduced the CLP-induced release of HMGB1 and sepsis-related mortality. piperlonguminine 28-30 high mobility group box 1 Homo sapiens 66-71 24468889-8 2014 CONCLUSION: These results indicate that PL could be a candidate therapeutic agent for various severe vascular inflammatory diseases via inhibition of the HMGB1 signaling pathway. piperlonguminine 40-42 high mobility group box 1 Homo sapiens 154-159 24776932-3 2014 In vitro the effect of atorvastatin on the production of HMGB1 by lipopolysaccharide (LPS)-stimulated human umbilical vein endothelial cells (HUVEC) was assessed. Atorvastatin 23-35 high mobility group box 1 Homo sapiens 57-62 24776932-6 2014 HMGB1 levels were reduced in GPA patients on statins and prednisolone. Prednisolone 57-69 high mobility group box 1 Homo sapiens 0-5 24776932-7 2014 In vitro, atorvastatin reduced HMGB1 levels in supernatants of activated HUVEC. Atorvastatin 10-22 high mobility group box 1 Homo sapiens 31-36 24776932-9 2014 Statins and prednisolone are associated with reduced serum HMGB1 levels and atorvastatin decreases HMGB1 release by activated HUVEC in vitro, indicating an additional anti-inflammatory effect of statins. Prednisolone 12-24 high mobility group box 1 Homo sapiens 59-64 24776932-9 2014 Statins and prednisolone are associated with reduced serum HMGB1 levels and atorvastatin decreases HMGB1 release by activated HUVEC in vitro, indicating an additional anti-inflammatory effect of statins. Atorvastatin 76-88 high mobility group box 1 Homo sapiens 99-104 24412753-3 2014 BTB, as a negative regulator of cell cycle progression, was identified as a HMGB1 interacting partner. btb 0-3 high mobility group box 1 Homo sapiens 76-81 24412753-4 2014 The ectopic expression of HMGB1 activates cell growth by suppressing BTB-induced cell death, decreasing Bax and p53 expression, while enhancing Bcl-xL, Bcl-2, cyclin D1, and NF-kappaB expression. btb 69-72 high mobility group box 1 Homo sapiens 26-31 24412753-5 2014 HMGB1 activates the FAK/PI3K/mTOR signaling cascade, and BTB prominently inhibits HMGB1-induced oncogenesis. btb 57-60 high mobility group box 1 Homo sapiens 82-87 24549588-6 2014 MTT and apoptosis assays were also performed and it was demonstrated that extracellular HMGB1 subsequently enhanced resistance to the P-gp-related drugs, adriamycin and vincristine. monooxyethylene trimethylolpropane tristearate 0-3 high mobility group box 1 Homo sapiens 88-93 24077682-0 2014 Anti-septic effects of pellitorine in HMGB1-induced inflammatory responses in vitro and in vivo. pellitorine 23-34 high mobility group box 1 Homo sapiens 38-43 24077682-3 2014 In this study, we investigated the anti-septic effects of PT against pro-inflammatory responses in human umbilical vein endothelial cells (HUVECs) induced by HMGB1 and the associated signaling pathways. pellitorine 58-60 high mobility group box 1 Homo sapiens 158-163 24549588-7 2014 In brief, this study demonstrated that extracellular HMGB1 may promote drug resistance to adriamycin and vincristine by upregulating P-gp in human gastric adenocarcinoma cells. Doxorubicin 90-100 high mobility group box 1 Homo sapiens 53-58 24549588-7 2014 In brief, this study demonstrated that extracellular HMGB1 may promote drug resistance to adriamycin and vincristine by upregulating P-gp in human gastric adenocarcinoma cells. Vincristine 105-116 high mobility group box 1 Homo sapiens 53-58 24549588-6 2014 MTT and apoptosis assays were also performed and it was demonstrated that extracellular HMGB1 subsequently enhanced resistance to the P-gp-related drugs, adriamycin and vincristine. Doxorubicin 154-164 high mobility group box 1 Homo sapiens 88-93 24549588-6 2014 MTT and apoptosis assays were also performed and it was demonstrated that extracellular HMGB1 subsequently enhanced resistance to the P-gp-related drugs, adriamycin and vincristine. Vincristine 169-180 high mobility group box 1 Homo sapiens 88-93 24531895-2 2014 Recently, it was shown that different redox states of the three cysteines of HMGB1 endow it with mutually exclusive activities, such as inducing chemotaxis or the transcription of cytokines and chemokines, via the interaction with different receptors. Cysteine 64-73 high mobility group box 1 Homo sapiens 77-82 24262631-2 2014 Given that no evidence for roles of HMGB1 in visceral pain signaling is available, we asked if HMGB1 participates in bladder pain accompanying cystitis caused by cyclophosphamide in mice, using the anti-HMGB1 neutralizing antibody and recombinant human soluble thrombomodulin (rhsTM) that sequesters HMGB1 and promotes its degradation by thrombin. Cyclophosphamide 162-178 high mobility group box 1 Homo sapiens 95-100 24070090-7 2014 Leukocyte activation abated in the presence of inhibitors of HMGB1 or of catalase, which catalyzes the dismutation of hydrogen peroxide into water and molecular oxygen. Hydrogen Peroxide 118-135 high mobility group box 1 Homo sapiens 61-66 24070090-7 2014 Leukocyte activation abated in the presence of inhibitors of HMGB1 or of catalase, which catalyzes the dismutation of hydrogen peroxide into water and molecular oxygen. Water 141-146 high mobility group box 1 Homo sapiens 61-66 23373897-5 2014 By damaging cells, superoxide and peroxynitrite promote leakage of HMGB1. Superoxides 19-29 high mobility group box 1 Homo sapiens 67-72 24070090-7 2014 Leukocyte activation abated in the presence of inhibitors of HMGB1 or of catalase, which catalyzes the dismutation of hydrogen peroxide into water and molecular oxygen. Oxygen 161-167 high mobility group box 1 Homo sapiens 61-66 23373897-5 2014 By damaging cells, superoxide and peroxynitrite promote leakage of HMGB1. Peroxynitrous Acid 34-47 high mobility group box 1 Homo sapiens 67-72 24070090-10 2014 INNOVATION AND CONCLUSION: ROS dramatically increase the ability of extracellular HMGB1 to activate blood leukocytes. Reactive Oxygen Species 27-30 high mobility group box 1 Homo sapiens 82-87 23373897-6 2014 RECENT ADVANCES: The activity of HMGB1 strongly depends on its redox state: Inflammatory-active HMGB1 requires an intramolecular disulfide bond (Cys23 and Cys45) and a reduced Cys106. Disulfides 129-138 high mobility group box 1 Homo sapiens 33-38 24420848-0 2014 Metformin protects against hyperglycemia-induced cardiomyocytes injury by inhibiting the expressions of receptor for advanced glycation end products and high mobility group box 1 protein. Metformin 0-9 high mobility group box 1 Homo sapiens 153-178 23373897-6 2014 RECENT ADVANCES: The activity of HMGB1 strongly depends on its redox state: Inflammatory-active HMGB1 requires an intramolecular disulfide bond (Cys23 and Cys45) and a reduced Cys106. Disulfides 129-138 high mobility group box 1 Homo sapiens 96-101 24431405-5 2014 Using in vitro models with mouse and human malignant mesothelioma cells, curcumin is shown to induce pyroptosis through activation of caspase-1 and increased release of high-mobility group box 1 (HMGB1) without processing of IL-1beta and IL-18. Curcumin 73-81 high mobility group box 1 Homo sapiens 169-194 24431405-5 2014 Using in vitro models with mouse and human malignant mesothelioma cells, curcumin is shown to induce pyroptosis through activation of caspase-1 and increased release of high-mobility group box 1 (HMGB1) without processing of IL-1beta and IL-18. Curcumin 73-81 high mobility group box 1 Homo sapiens 196-201 24362470-4 2014 Here, we found that colorectal cancer (CRC) cells treated with oxaliplatin (OXA) and/or 5-fluorouracil (5-Fu) released high levels of high-mobility group box 1 (HMGB1) and heat shock protein 70 (HSP70). Fluorouracil 104-108 high mobility group box 1 Homo sapiens 134-159 24362470-4 2014 Here, we found that colorectal cancer (CRC) cells treated with oxaliplatin (OXA) and/or 5-fluorouracil (5-Fu) released high levels of high-mobility group box 1 (HMGB1) and heat shock protein 70 (HSP70). Fluorouracil 104-108 high mobility group box 1 Homo sapiens 161-166 24362470-5 2014 After OXA/5-Fu therapy, the sera of CRC patients also exhibited increased levels of HMGB1 and HSP70, both of which are well-known DAMPs. Oxaliplatin 6-9 high mobility group box 1 Homo sapiens 84-89 24362470-5 2014 After OXA/5-Fu therapy, the sera of CRC patients also exhibited increased levels of HMGB1 and HSP70, both of which are well-known DAMPs. Fluorouracil 10-14 high mobility group box 1 Homo sapiens 84-89 24370952-5 2014 We determined that high amplitude cyclic stretch (18% CS) increased HMGB1 expression (2-4-fold) via a signaling pathway with critical involvement of the transcription factor, STAT3. Cesium 54-56 high mobility group box 1 Homo sapiens 68-73 24370952-6 2014 Concomitant exposure to 18% CS and oxidative stress (H2O2) augmented HMGB1 expression (~13 fold increase) whereas lipopolysaccharide (LPS) challenge increased HMGB1 expression in static EC, but not in 18% CS-challenged EC. Cesium 28-30 high mobility group box 1 Homo sapiens 69-74 24370952-6 2014 Concomitant exposure to 18% CS and oxidative stress (H2O2) augmented HMGB1 expression (~13 fold increase) whereas lipopolysaccharide (LPS) challenge increased HMGB1 expression in static EC, but not in 18% CS-challenged EC. Hydrogen Peroxide 53-57 high mobility group box 1 Homo sapiens 69-74 24370952-7 2014 In contrast, physiologic, low amplitude cyclic stretch (5% CS) attenuated both oxidative H2O2- and LPS-induced increases in HMGB1 expression, suggesting that physiologic mechanical stress is protective. Cesium 59-61 high mobility group box 1 Homo sapiens 124-129 24370952-7 2014 In contrast, physiologic, low amplitude cyclic stretch (5% CS) attenuated both oxidative H2O2- and LPS-induced increases in HMGB1 expression, suggesting that physiologic mechanical stress is protective. Hydrogen Peroxide 89-93 high mobility group box 1 Homo sapiens 124-129 24362470-4 2014 Here, we found that colorectal cancer (CRC) cells treated with oxaliplatin (OXA) and/or 5-fluorouracil (5-Fu) released high levels of high-mobility group box 1 (HMGB1) and heat shock protein 70 (HSP70). Oxaliplatin 63-74 high mobility group box 1 Homo sapiens 134-159 24362470-4 2014 Here, we found that colorectal cancer (CRC) cells treated with oxaliplatin (OXA) and/or 5-fluorouracil (5-Fu) released high levels of high-mobility group box 1 (HMGB1) and heat shock protein 70 (HSP70). Oxaliplatin 63-74 high mobility group box 1 Homo sapiens 161-166 24362470-4 2014 Here, we found that colorectal cancer (CRC) cells treated with oxaliplatin (OXA) and/or 5-fluorouracil (5-Fu) released high levels of high-mobility group box 1 (HMGB1) and heat shock protein 70 (HSP70). Oxaliplatin 76-79 high mobility group box 1 Homo sapiens 134-159 24362470-4 2014 Here, we found that colorectal cancer (CRC) cells treated with oxaliplatin (OXA) and/or 5-fluorouracil (5-Fu) released high levels of high-mobility group box 1 (HMGB1) and heat shock protein 70 (HSP70). Oxaliplatin 76-79 high mobility group box 1 Homo sapiens 161-166 24362470-4 2014 Here, we found that colorectal cancer (CRC) cells treated with oxaliplatin (OXA) and/or 5-fluorouracil (5-Fu) released high levels of high-mobility group box 1 (HMGB1) and heat shock protein 70 (HSP70). Fluorouracil 88-102 high mobility group box 1 Homo sapiens 134-159 24362470-4 2014 Here, we found that colorectal cancer (CRC) cells treated with oxaliplatin (OXA) and/or 5-fluorouracil (5-Fu) released high levels of high-mobility group box 1 (HMGB1) and heat shock protein 70 (HSP70). Fluorouracil 88-102 high mobility group box 1 Homo sapiens 161-166 24551219-2 2014 Recent reports from several laboratories provided evidence that a number of both the intracellular and extracellular functions of HMGB1 may depend on redox-sensitive cysteine residues of the protein. Cysteine 166-174 high mobility group box 1 Homo sapiens 130-135 24524683-1 2014 An MS-based proteomic strategy combined with chemically functionalized gold nanoparticles as affinity probes was developed and validated by successful identification and quantification of HMGB1, which is well characterized to interact selectively with 1,2-cross-linked DNA by cisplatin, from whole cell lysates. Cisplatin 276-285 high mobility group box 1 Homo sapiens 188-193 24551070-4 2014 We previously found increased HMGB1 in post-mortem alcoholic human brain as well as in ethanol treated mice and rat brain slice cultures. Ethanol 87-94 high mobility group box 1 Homo sapiens 30-35 25004875-0 2014 Baicalein inhibits HMGB1 release and MMP-2/-9 expression in lipopolysaccharide-induced cardiac hypertrophy. baicalein 0-9 high mobility group box 1 Homo sapiens 19-24 23318458-5 2014 RAGE and HMGB1 coordinately enhanced tumor cell mitochondrial complex I activity, ATP production, tumor cell proliferation and migration. Adenosine Triphosphate 82-85 high mobility group box 1 Homo sapiens 9-14 23318458-6 2014 Lack of RAGE or inhibition of HMGB1 release diminished ATP production and slowed tumor growth in vitro and in vivo. Adenosine Triphosphate 55-58 high mobility group box 1 Homo sapiens 30-35 24361892-0 2014 Ethyl pyruvate inhibits proliferation and induces apoptosis of hepatocellular carcinoma via regulation of the HMGB1-RAGE and AKT pathways. ethyl pyruvate 0-14 high mobility group box 1 Homo sapiens 110-115 24361892-7 2014 Real-time PCR, Western blotting and immunofluorescence demonstrated ethyl pyruvate reduced the HMGB1-RAGE and AKT pathways. ethyl pyruvate 68-82 high mobility group box 1 Homo sapiens 95-100 25826983-4 2014 The article presents the proposed role of dipeptide vilon (Lys-Glu) and tetrapeptide epitalon (Ala-Glu-Asp-Gly) in CCL11 and HMGB1 genes regulation as activators of their expression. Dipeptides 42-51 high mobility group box 1 Homo sapiens 125-130 25826983-4 2014 The article presents the proposed role of dipeptide vilon (Lys-Glu) and tetrapeptide epitalon (Ala-Glu-Asp-Gly) in CCL11 and HMGB1 genes regulation as activators of their expression. lysylglutamic acid 59-66 high mobility group box 1 Homo sapiens 125-130 25826983-4 2014 The article presents the proposed role of dipeptide vilon (Lys-Glu) and tetrapeptide epitalon (Ala-Glu-Asp-Gly) in CCL11 and HMGB1 genes regulation as activators of their expression. alanyl-glutamyl-aspartyl-glycine 95-110 high mobility group box 1 Homo sapiens 125-130 25004875-4 2014 In addition, pretreatment with baicalein inhibited LPS-induced early (e.g., tumor necrosis factor-alpha (TNF-alpha) and interleukin-6) and late (e.g., high mobility group box 1 (HMGB1) pro-inflammatory cytokine release, inducible nitric oxide synthase (iNOS) expression and NO production. baicalein 31-40 high mobility group box 1 Homo sapiens 151-176 25004875-4 2014 In addition, pretreatment with baicalein inhibited LPS-induced early (e.g., tumor necrosis factor-alpha (TNF-alpha) and interleukin-6) and late (e.g., high mobility group box 1 (HMGB1) pro-inflammatory cytokine release, inducible nitric oxide synthase (iNOS) expression and NO production. baicalein 31-40 high mobility group box 1 Homo sapiens 178-183 26629935-8 2014 We then investigated the effect of HMGB1 knockdown/out on the sensitivity of BUC cells treated with the anticancer drug cisplatin. Cisplatin 120-129 high mobility group box 1 Homo sapiens 35-40 23811849-4 2014 The absence of HMGB1 expression by dying tumor cells exposed to anthracyclines or oxaliplatin compromises DC-dependent T-cell priming by tumor-associated antigens. Anthracyclines 64-78 high mobility group box 1 Homo sapiens 15-20 23811849-5 2014 Here, we show that transplantable tumors exhibiting weak expression of nuclear HMGB1 respond to chemotherapy more effectively if the treatment is combined with the local or systemic administration of a highly purified and physiochemically defined and standardized lipopolysaccharide solution, which acts as a high-potency and exclusive TLR4 agonist, called Dendrophilin (DEN). dendrophilin 357-369 high mobility group box 1 Homo sapiens 79-84 23811849-5 2014 Here, we show that transplantable tumors exhibiting weak expression of nuclear HMGB1 respond to chemotherapy more effectively if the treatment is combined with the local or systemic administration of a highly purified and physiochemically defined and standardized lipopolysaccharide solution, which acts as a high-potency and exclusive TLR4 agonist, called Dendrophilin (DEN). Diethylnitrosamine 371-374 high mobility group box 1 Homo sapiens 79-84 23811849-8 2014 Moreover, DEN could restore the immunogenicity of dying tumor cells from which HMGB1 had been depleted by RNA interference. Diethylnitrosamine 10-13 high mobility group box 1 Homo sapiens 79-84 23832118-6 2014 In this review, we provide a brief outline of the well-characterized DAMPs such as calreticulin (CRT) exposure, high-mobility group protein B1 (HMGB1), and adenosine triphosphate (ATP) release, which are induced by the morphologically distinct types of cell death. Adenosine Triphosphate 156-178 high mobility group box 1 Homo sapiens 144-149 24184598-0 2014 Anti-septic effects of glyceollins in HMGB1-induced inflammatory responses in vitro and in vivo. glyceollin 23-34 high mobility group box 1 Homo sapiens 38-43 24178442-0 2014 HMGB1 is involved in autophagy inhibition caused by SNCA/alpha-synuclein overexpression: a process modulated by the natural autophagy inducer corynoxine B. corynoxine B 142-154 high mobility group box 1 Homo sapiens 0-5 24178442-8 2014 Corynoxine B, a natural autophagy inducer, restores the deficient cytosolic translocation of HMGB1 and autophagy in cells overexpressing SNCA, which may be attributed to its ability to block SNCA-HMGB1 interaction. corynoxine B 0-12 high mobility group box 1 Homo sapiens 93-98 24178442-8 2014 Corynoxine B, a natural autophagy inducer, restores the deficient cytosolic translocation of HMGB1 and autophagy in cells overexpressing SNCA, which may be attributed to its ability to block SNCA-HMGB1 interaction. corynoxine B 0-12 high mobility group box 1 Homo sapiens 196-201 25243124-0 2014 Adsorption of the inflammatory mediator high-mobility group box 1 by polymers with different charge and porosity. Polymers 69-77 high mobility group box 1 Homo sapiens 40-65 25243124-4 2014 Here, we studied the adsorption of HMGB1 to anionic methacrylate-based polymers as well as to neutral polystyrene-divinylbenzene copolymers. methacrylate-based polymers 52-79 high mobility group box 1 Homo sapiens 35-40 23852373-3 2014 The release of ATP from dying cells constitutes one of the three major hallmarks of ICD and occurs independently of the two others, namely, the pre-apoptotic exposure of calreticulin on the cell surface and the postmortem release of high-mobility group box 1 (HMBG1) into the extracellular space. Adenosine Triphosphate 15-18 high mobility group box 1 Homo sapiens 233-258 23852373-3 2014 The release of ATP from dying cells constitutes one of the three major hallmarks of ICD and occurs independently of the two others, namely, the pre-apoptotic exposure of calreticulin on the cell surface and the postmortem release of high-mobility group box 1 (HMBG1) into the extracellular space. Adenosine Triphosphate 15-18 high mobility group box 1 Homo sapiens 260-265 25175868-4 2014 In addition, HMGB1-TLR4 and innate immune NF-kappaB target genes are increased leading to persistent and sensitized neuroimmune responses to ethanol and other agents that release HMGB1 or directly stimulate TLR receptors and/or NMDA receptors. Ethanol 141-148 high mobility group box 1 Homo sapiens 13-18 25175868-4 2014 In addition, HMGB1-TLR4 and innate immune NF-kappaB target genes are increased leading to persistent and sensitized neuroimmune responses to ethanol and other agents that release HMGB1 or directly stimulate TLR receptors and/or NMDA receptors. Ethanol 141-148 high mobility group box 1 Homo sapiens 179-184 26629935-9 2014 Knockdown or knockout of HMGB1 rendered BUC cells more sensitive to cisplatin. Cisplatin 68-77 high mobility group box 1 Homo sapiens 25-30 23895427-0 2013 The cytokine mRNA increase induced by withdrawal from chronic ethanol in the sterile environment of brain is mediated by CRF and HMGB1 release. Ethanol 62-69 high mobility group box 1 Homo sapiens 129-134 24011512-2 2014 RAGE transduces the signals of advanced glycation end-products (AGEs), proinflammatory S100/calgranulins, and high mobility group box 1 (HMGB1), and is a one of a family of receptors for lysophosphatidic acid (LPA). lysophosphatidic acid 187-208 high mobility group box 1 Homo sapiens 110-135 24011512-2 2014 RAGE transduces the signals of advanced glycation end-products (AGEs), proinflammatory S100/calgranulins, and high mobility group box 1 (HMGB1), and is a one of a family of receptors for lysophosphatidic acid (LPA). lysophosphatidic acid 187-208 high mobility group box 1 Homo sapiens 137-142 24011512-2 2014 RAGE transduces the signals of advanced glycation end-products (AGEs), proinflammatory S100/calgranulins, and high mobility group box 1 (HMGB1), and is a one of a family of receptors for lysophosphatidic acid (LPA). lysophosphatidic acid 210-213 high mobility group box 1 Homo sapiens 137-142 23895427-15 2013 The CRF1RA and the HMGB1 antagonist, ethyl pyruvate, also reduced the HMGB1 mRNA increase that followed CE withdrawal. ethyl pyruvate 37-51 high mobility group box 1 Homo sapiens 19-24 23895427-15 2013 The CRF1RA and the HMGB1 antagonist, ethyl pyruvate, also reduced the HMGB1 mRNA increase that followed CE withdrawal. ethyl pyruvate 37-51 high mobility group box 1 Homo sapiens 70-75 24094823-1 2013 In this study, pH-sensitive biomaterials coated polymer/DNA nanocomplexes containing a high mobility group box 1 (HMGB1) were developed as an efficient non-viral gene delivery system. Polymers 48-55 high mobility group box 1 Homo sapiens 87-112 24091596-0 2013 HMGB1 recruits hepatic stellate cells and liver endothelial cells to sites of ethanol-induced parenchymal cell injury. Ethanol 78-85 high mobility group box 1 Homo sapiens 0-5 24091596-3 2013 We hypothesized that HMGB1 may be released from ethanol-stimulated liver parenchymal cells and contribute to HSC and LEC recruitment. Ethanol 48-55 high mobility group box 1 Homo sapiens 21-26 24091596-4 2013 Ethanol stimulation of rat hepatocytes and HepG2 cells resulted in translocation of HMGB1 from the nucleus as assessed by Western blot. Ethanol 0-7 high mobility group box 1 Homo sapiens 84-89 24091596-5 2013 HMGB1 protein levels were increased in the supernatant of ethanol-treated hepatocytes compared with vehicle-treated cells. Ethanol 58-65 high mobility group box 1 Homo sapiens 0-5 24091596-7 2013 However, the effect of CMEtOH on migration was almost entirely reversed by treatment with HMGB1-neutralizing antibody or when HepG2 cells were pretransfected with HMGB1-siRNA compared with control siRNA-transfected HepG2 cells (P < 0.05). cmetoh 23-29 high mobility group box 1 Homo sapiens 90-95 24091596-7 2013 However, the effect of CMEtOH on migration was almost entirely reversed by treatment with HMGB1-neutralizing antibody or when HepG2 cells were pretransfected with HMGB1-siRNA compared with control siRNA-transfected HepG2 cells (P < 0.05). cmetoh 23-29 high mobility group box 1 Homo sapiens 163-168 24091596-9 2013 HMGB1 stimulation of HSC increased the phosphorylation of Src and Erk and HMGB1-induced HSC migration was blocked by the Src inhibitor PP2 and the Erk inhibitor U0126. U 0126 161-166 high mobility group box 1 Homo sapiens 0-5 24091596-9 2013 HMGB1 stimulation of HSC increased the phosphorylation of Src and Erk and HMGB1-induced HSC migration was blocked by the Src inhibitor PP2 and the Erk inhibitor U0126. U 0126 161-166 high mobility group box 1 Homo sapiens 74-79 24091596-10 2013 Hepatocytes release HMGB1 in response to ethanol with subsequent recruitment of HSC and LEC. Ethanol 41-48 high mobility group box 1 Homo sapiens 20-25 24094823-1 2013 In this study, pH-sensitive biomaterials coated polymer/DNA nanocomplexes containing a high mobility group box 1 (HMGB1) were developed as an efficient non-viral gene delivery system. Polymers 48-55 high mobility group box 1 Homo sapiens 114-119 24160326-0 2013 Continuous exposure to chrysotile asbestos can cause transformation of human mesothelial cells via HMGB1 and TNF-alpha signaling. Asbestos 34-42 high mobility group box 1 Homo sapiens 99-104 23928875-0 2013 High mobility group box 1 (HMGB1) mediates high-glucose-induced calcification in vascular smooth muscle cells of saphenous veins. Glucose 48-55 high mobility group box 1 Homo sapiens 0-25 23928875-0 2013 High mobility group box 1 (HMGB1) mediates high-glucose-induced calcification in vascular smooth muscle cells of saphenous veins. Glucose 48-55 high mobility group box 1 Homo sapiens 27-32 23928875-4 2013 Therefore, the role of their common ligand, high mobility group box 1 (HMGB1), in high-glucose-induced calcification in VSMC of saphenous vein was investigated. Glucose 87-94 high mobility group box 1 Homo sapiens 44-69 23928875-4 2013 Therefore, the role of their common ligand, high mobility group box 1 (HMGB1), in high-glucose-induced calcification in VSMC of saphenous vein was investigated. Glucose 87-94 high mobility group box 1 Homo sapiens 71-76 23928875-6 2013 We first demonstrated high-glucose-induced HMGB1 translocation from nucleus to cytosol, and this translocation was induced through a NADPH oxidase and PKC-dependent pathway. Glucose 27-34 high mobility group box 1 Homo sapiens 43-48 23928875-8 2013 Then, we revealed downregulating HMGB1 expression abolished high-glucose-induced calcification accompanied by NFkappaB inactivation and low expression of bone morphogenetic protein-2 (BMP-2). Glucose 65-72 high mobility group box 1 Homo sapiens 33-38 23928875-11 2013 Our findings thus suggest HMGB1 plays an important role in mediating the calcification process induced by high glucose through NFkappaB activation and BMP-2 expression in VSMC of saphenous vein. Glucose 111-118 high mobility group box 1 Homo sapiens 26-31 24255708-2 2013 The human HMGB1 is composed of two binding motifs, known as Boxes A and B, are L-shaped alpha-helix structures, followed by a random-coil acidic tail that consists of 30 Asp and Glu residues. Aspartic Acid 170-173 high mobility group box 1 Homo sapiens 10-15 24255708-2 2013 The human HMGB1 is composed of two binding motifs, known as Boxes A and B, are L-shaped alpha-helix structures, followed by a random-coil acidic tail that consists of 30 Asp and Glu residues. Glutamic Acid 178-181 high mobility group box 1 Homo sapiens 10-15 24133029-8 2013 RESULTS: HMGB1 signalling pathway components including receptors for HMGB1 as well as NF-kappaB and TNFalpha/VEGF were significantly upregulated in type 2 diabetic retinas and in high glucose treated ARPE-19 cells, compared to their respective counterparts. Glucose 184-191 high mobility group box 1 Homo sapiens 9-14 24133029-9 2013 HMGB1 blockage significantly alleviated NF-kappaB activity and VEGF secretion in ARPE-19 cells cultured with high glucose. Glucose 114-121 high mobility group box 1 Homo sapiens 0-5 24465145-2 2013 In the postischemic brain, HMGB1 is massively released during NMDA-induced acute damage and triggers inflammatory processes. N-Methylaspartate 62-66 high mobility group box 1 Homo sapiens 27-32 24465145-8 2013 A pull-down assay using biotin-labeled HBHP showed that HBHP binds directly to HMGB1 (more specifically to HMGB1 A box) in LCM. Biotin 24-30 high mobility group box 1 Homo sapiens 79-84 24465145-8 2013 A pull-down assay using biotin-labeled HBHP showed that HBHP binds directly to HMGB1 (more specifically to HMGB1 A box) in LCM. Biotin 24-30 high mobility group box 1 Homo sapiens 107-112 24427810-0 2013 Redox-sensitive structural change in the A-domain of HMGB1 and its implication for the binding to cisplatin modified DNA. Cisplatin 98-107 high mobility group box 1 Homo sapiens 53-58 24427810-3 2013 Two cysteines, Cys23 and Cys45, in the A-domain of HMGB1 form a disulfide bond under oxidative conditions. Cysteine 4-13 high mobility group box 1 Homo sapiens 51-56 24427810-3 2013 Two cysteines, Cys23 and Cys45, in the A-domain of HMGB1 form a disulfide bond under oxidative conditions. Disulfides 64-73 high mobility group box 1 Homo sapiens 51-56 24427810-8 2013 The reorientation of the Phe38 ring by the disulfide bond in the A-domain may explain the reduced HMGB1 binding affinity towards cisplatinated DNA. Disulfides 43-52 high mobility group box 1 Homo sapiens 98-103 24160326-10 2013 Similarly, HMGB1 release in vivo progressively increased for 10 or more weeks after crocidolite exposure, but returned to background levels within 8 weeks after chrysotile exposure. Asbestos, Crocidolite 84-95 high mobility group box 1 Homo sapiens 11-16 24591411-8 2013 Correlation analysis showed that plasma levels of HMGB-1 correlated with initial serum creatinine (r = 0.275, P = 0.018), estimated glomerular filtration rate (r = -0.277, P = 0.017), the Birmingham Vasculitis Activity Score (r = 0.308, P = 0.008), and C-reactive protein level (r = 0.309, P = 0.008). Creatinine 87-97 high mobility group box 1 Homo sapiens 50-56 24204700-7 2013 One hour after injection, ELISA showed that circulating extracellular HMGB1 levels were elevated after BMC transplantation compared to the PBS injection. bmc 103-106 high mobility group box 1 Homo sapiens 70-75 23867237-11 2013 Adolescent intermittent ethanol exposure also increased TLR4 and HMGB1 expression at P56 that persisted into young adulthood (P80). Ethanol 24-31 high mobility group box 1 Homo sapiens 65-70 24204700-5 2013 We thus studied the role of extracellular HMGB1 in the effect of BMC transplantation for heart failure. bmc 65-68 high mobility group box 1 Homo sapiens 42-47 23398207-6 2013 Furthermore, (-)- gossypol treatment increased the translocation of high mobility group box 1 (HMGB1) from nuclei to cytoplasm, which can be suppressed by NAC pretreatment. Gossypol 13-26 high mobility group box 1 Homo sapiens 68-93 24012904-0 2013 Histamine inhibits high mobility group box 1-induced adhesion molecule expression on human monocytes. Histamine 0-9 high mobility group box 1 Homo sapiens 19-44 24012904-5 2013 This study was designed to study the inhibitory effect of histamine on HMGB1 activity. Histamine 58-67 high mobility group box 1 Homo sapiens 71-76 24012904-6 2013 We examined the effect of histamine on HMGB1-induced expression of ICAM-1, B7.1, B7.2 and CD40 on monocytes, production of IFN-gamma and TNF-alpha and lymphocyte proliferation in PBMCs. Histamine 26-35 high mobility group box 1 Homo sapiens 39-44 24012904-7 2013 Histamine inhibited HMGB1 activity in a concentration-dependent manner. Histamine 0-9 high mobility group box 1 Homo sapiens 20-25 24012904-11 2013 These results together indicated that histamine inhibited HMGB1 activity. Histamine 38-47 high mobility group box 1 Homo sapiens 58-63 23398207-0 2013 Natural Bcl-2 inhibitor (-)- gossypol induces protective autophagy via reactive oxygen species-high mobility group box 1 pathway in Burkitt lymphoma. Gossypol 24-37 high mobility group box 1 Homo sapiens 95-120 23398207-0 2013 Natural Bcl-2 inhibitor (-)- gossypol induces protective autophagy via reactive oxygen species-high mobility group box 1 pathway in Burkitt lymphoma. Reactive Oxygen Species 71-94 high mobility group box 1 Homo sapiens 95-120 24204700-9 2013 Echocardiography and catheterization showed enhanced cardiac function after BMC transplantation compared to PBS injection at day 28, while this effect was abolished by antibody-neutralization of HMGB1. bmc 76-79 high mobility group box 1 Homo sapiens 195-200 24204700-10 2013 BMC transplantation reduced post-infarction fibrosis, improved neovascularization, and increased proliferation, while all these effects in repairing the failing myocardium were eliminated by HMGB1-inhibition. bmc 0-3 high mobility group box 1 Homo sapiens 191-196 24204700-11 2013 Furthermore, BMC transplantation drove the macrophage polarization towards alternatively-activated, anti-inflammatory M2 macrophages in the heart at day 3, while this was abolished by HMGB1-inhibition. bmc 13-16 high mobility group box 1 Homo sapiens 184-189 24204700-14 2013 These data suggest that extracellular HMGB1 contributes to the effect of BMC transplantation to recover the damaged myocardium by favorably modulating innate immunity in heart failure. bmc 73-76 high mobility group box 1 Homo sapiens 38-43 24255785-6 2013 In pulmonary system, binding of HMGB1 to its receptor for advanced glycation end-products (RAGE) triggers the production of pro-inflammatory cytokines, chemokines, adhesion molecules and reactive oxygen species, promoting the development of ALI/ARDS. Reactive Oxygen Species 187-210 high mobility group box 1 Homo sapiens 32-37 23398207-6 2013 Furthermore, (-)- gossypol treatment increased the translocation of high mobility group box 1 (HMGB1) from nuclei to cytoplasm, which can be suppressed by NAC pretreatment. Gossypol 13-26 high mobility group box 1 Homo sapiens 95-100 23398207-6 2013 Furthermore, (-)- gossypol treatment increased the translocation of high mobility group box 1 (HMGB1) from nuclei to cytoplasm, which can be suppressed by NAC pretreatment. Acetylcysteine 155-158 high mobility group box 1 Homo sapiens 68-93 23398207-6 2013 Furthermore, (-)- gossypol treatment increased the translocation of high mobility group box 1 (HMGB1) from nuclei to cytoplasm, which can be suppressed by NAC pretreatment. Acetylcysteine 155-158 high mobility group box 1 Homo sapiens 95-100 23398207-8 2013 These results indicate that (-)- gossypol induces a protective autophagy in Burkitt lymphoma cells, partly due to ROS induction and cytosolic translocation of HMGB1. Gossypol 28-41 high mobility group box 1 Homo sapiens 159-164 23932051-0 2013 Glycyrrhizic acid confers neuroprotection after subarachnoid hemorrhage via inhibition of high mobility group box-1 protein: a hypothesis for novel therapy of subarachnoid hemorrhage. Glycyrrhizic Acid 0-17 high mobility group box 1 Homo sapiens 90-115 24065095-0 2013 Potential of the angiotensin receptor blockers (ARBs) telmisartan, irbesartan, and candesartan for inhibiting the HMGB1/RAGE axis in prevention and acute treatment of stroke. Telmisartan 54-65 high mobility group box 1 Homo sapiens 114-119 23932051-11 2013 Glycyrrhizic acid (GA) which was extracted from liquorice and confirmed as a nature inhibitor of HMGB1 with little side-effects could inhibit extracellular HMGB1 cytokine activities and reduce the level of inflammatory response, thus alleviating early brain injury and cerebrovasospasm. Glycyrrhizic Acid 0-17 high mobility group box 1 Homo sapiens 97-102 23932051-11 2013 Glycyrrhizic acid (GA) which was extracted from liquorice and confirmed as a nature inhibitor of HMGB1 with little side-effects could inhibit extracellular HMGB1 cytokine activities and reduce the level of inflammatory response, thus alleviating early brain injury and cerebrovasospasm. Glycyrrhizic Acid 0-17 high mobility group box 1 Homo sapiens 156-161 23932051-11 2013 Glycyrrhizic acid (GA) which was extracted from liquorice and confirmed as a nature inhibitor of HMGB1 with little side-effects could inhibit extracellular HMGB1 cytokine activities and reduce the level of inflammatory response, thus alleviating early brain injury and cerebrovasospasm. Glycyrrhizic Acid 19-21 high mobility group box 1 Homo sapiens 97-102 23932051-11 2013 Glycyrrhizic acid (GA) which was extracted from liquorice and confirmed as a nature inhibitor of HMGB1 with little side-effects could inhibit extracellular HMGB1 cytokine activities and reduce the level of inflammatory response, thus alleviating early brain injury and cerebrovasospasm. Glycyrrhizic Acid 19-21 high mobility group box 1 Homo sapiens 156-161 24417167-8 2013 The level of HMGB1 mRNA expression in Hep-2 cells increased after the cells were induced by hypoxia for 6h PD98059 and SP600125 with 20 micromol/ L and PDTC with 50 mg/L partly inhibited extracellular release of HMGB1 in hypoxia-cultured Hcp-2 cells. pyrazolanthrone 119-127 high mobility group box 1 Homo sapiens 13-18 24417167-8 2013 The level of HMGB1 mRNA expression in Hep-2 cells increased after the cells were induced by hypoxia for 6h PD98059 and SP600125 with 20 micromol/ L and PDTC with 50 mg/L partly inhibited extracellular release of HMGB1 in hypoxia-cultured Hcp-2 cells. pyrazolanthrone 119-127 high mobility group box 1 Homo sapiens 212-217 24065095-0 2013 Potential of the angiotensin receptor blockers (ARBs) telmisartan, irbesartan, and candesartan for inhibiting the HMGB1/RAGE axis in prevention and acute treatment of stroke. Irbesartan 67-77 high mobility group box 1 Homo sapiens 114-119 24065095-0 2013 Potential of the angiotensin receptor blockers (ARBs) telmisartan, irbesartan, and candesartan for inhibiting the HMGB1/RAGE axis in prevention and acute treatment of stroke. candesartan 83-94 high mobility group box 1 Homo sapiens 114-119 23244202-4 2013 To investigate these processes, the extracellular release of HMGB1 during necrosis was characterized in vitro using Jurkat T cell leukemia cells treated to induce necrosis by freeze-thaw, heat, hydrogen peroxide or ethanol. Hydrogen Peroxide 194-211 high mobility group box 1 Homo sapiens 61-66 23911608-8 2013 HMGB1-induced increases in ICAM-1 and P-selectin expression were also inhibited by a specific eIF2alpha inhibitor (salubrinal) and a specific JNK inhibitor (SP600125). salubrinal 115-125 high mobility group box 1 Homo sapiens 0-5 23911608-8 2013 HMGB1-induced increases in ICAM-1 and P-selectin expression were also inhibited by a specific eIF2alpha inhibitor (salubrinal) and a specific JNK inhibitor (SP600125). pyrazolanthrone 157-165 high mobility group box 1 Homo sapiens 0-5 23834721-0 2013 Supporting cells regulate the remodelling of aminoglycoside-injured organ of Corti, through the release of high mobility group box 1. Aminoglycosides 45-59 high mobility group box 1 Homo sapiens 107-132 23685846-7 2013 RESULT: We demonstrated that the patients with DMPM had significantly higher serum levels of HMGB1 compared with the population who had been exposed to asbestos but did not develop DMPM. dmpm 47-51 high mobility group box 1 Homo sapiens 93-98 23685846-8 2013 CONCLUSION: Our data suggest that serum HMGB1 concentration is a useful serum marker for DMPM. dmpm 89-93 high mobility group box 1 Homo sapiens 40-45 23148306-9 2013 In order to bind and signal via TLR4, HMGB1 must have a reduced cysteine 106 and a disulphide linkage between cysteine 23 and 45. Cysteine 64-72 high mobility group box 1 Homo sapiens 38-43 23148306-9 2013 In order to bind and signal via TLR4, HMGB1 must have a reduced cysteine 106 and a disulphide linkage between cysteine 23 and 45. disulphide 83-93 high mobility group box 1 Homo sapiens 38-43 23148306-9 2013 In order to bind and signal via TLR4, HMGB1 must have a reduced cysteine 106 and a disulphide linkage between cysteine 23 and 45. Cysteine 110-118 high mobility group box 1 Homo sapiens 38-43 23769884-8 2013 However, the pretreatment with CM1 could significantly decrease AGE or high-mobility group box-1 (HMGB1, a ligand of RAGE)-induced cytotoxicity, apoptosis and reactive oxygen species (ROS) in human umbilical vein endothelial cells (HUVECs). Reactive Oxygen Species 159-182 high mobility group box 1 Homo sapiens 71-96 23769884-8 2013 However, the pretreatment with CM1 could significantly decrease AGE or high-mobility group box-1 (HMGB1, a ligand of RAGE)-induced cytotoxicity, apoptosis and reactive oxygen species (ROS) in human umbilical vein endothelial cells (HUVECs). Reactive Oxygen Species 184-187 high mobility group box 1 Homo sapiens 71-96 23697559-4 2013 The phosphorylation of both extracellular signal-regulated kinase (ERK)1/2 and p38 mitogen-activated protein kinase (MAPK) was markedly upregulated by treating with high amount of HMGB1, while pretreatment with ERK1/2 and p38 MAPK-specific inhibitors (U0126 and SB203580) could attenuate suppression of T cell immune function and nuclear factor of activated T cell (NFAT) activation induced by HMGB1, respectively. U 0126 252-257 high mobility group box 1 Homo sapiens 180-185 23697559-4 2013 The phosphorylation of both extracellular signal-regulated kinase (ERK)1/2 and p38 mitogen-activated protein kinase (MAPK) was markedly upregulated by treating with high amount of HMGB1, while pretreatment with ERK1/2 and p38 MAPK-specific inhibitors (U0126 and SB203580) could attenuate suppression of T cell immune function and nuclear factor of activated T cell (NFAT) activation induced by HMGB1, respectively. U 0126 252-257 high mobility group box 1 Homo sapiens 394-399 23697559-4 2013 The phosphorylation of both extracellular signal-regulated kinase (ERK)1/2 and p38 mitogen-activated protein kinase (MAPK) was markedly upregulated by treating with high amount of HMGB1, while pretreatment with ERK1/2 and p38 MAPK-specific inhibitors (U0126 and SB203580) could attenuate suppression of T cell immune function and nuclear factor of activated T cell (NFAT) activation induced by HMGB1, respectively. SB 203580 262-270 high mobility group box 1 Homo sapiens 180-185 23697559-4 2013 The phosphorylation of both extracellular signal-regulated kinase (ERK)1/2 and p38 mitogen-activated protein kinase (MAPK) was markedly upregulated by treating with high amount of HMGB1, while pretreatment with ERK1/2 and p38 MAPK-specific inhibitors (U0126 and SB203580) could attenuate suppression of T cell immune function and nuclear factor of activated T cell (NFAT) activation induced by HMGB1, respectively. SB 203580 262-270 high mobility group box 1 Homo sapiens 394-399 23697559-5 2013 HMGB1-induced activity of ERK1/2 and p38 was not fully inhibited in the presence of U0126 or SB203580. U 0126 84-89 high mobility group box 1 Homo sapiens 0-5 23697559-5 2013 HMGB1-induced activity of ERK1/2 and p38 was not fully inhibited in the presence of U0126 or SB203580. SB 203580 93-101 high mobility group box 1 Homo sapiens 0-5 23244202-4 2013 To investigate these processes, the extracellular release of HMGB1 during necrosis was characterized in vitro using Jurkat T cell leukemia cells treated to induce necrosis by freeze-thaw, heat, hydrogen peroxide or ethanol. Ethanol 215-222 high mobility group box 1 Homo sapiens 61-66 23207101-5 2013 Notably, only one of the redox forms of HMGB1, the one where all cysteines are reduced (all-thiol), can bind CXCL12. Cysteine 65-74 high mobility group box 1 Homo sapiens 40-45 23647088-6 2013 After exposure to H2O2, a marked decrease in cell viability (<50%) was demonstrated, and this was significantly ameliorated upon culture at 32 C. Significantly intracellular damage was found to affect the 24-hour H2O2-exposed cells in 37 C (P < .05), including an increase in apoptosis and necrosis, intracellular ROS, caspase-3 activity, HMGB1 protein expression, and some alterations to the cell cycle. Hydrogen Peroxide 18-22 high mobility group box 1 Homo sapiens 345-350 23647088-6 2013 After exposure to H2O2, a marked decrease in cell viability (<50%) was demonstrated, and this was significantly ameliorated upon culture at 32 C. Significantly intracellular damage was found to affect the 24-hour H2O2-exposed cells in 37 C (P < .05), including an increase in apoptosis and necrosis, intracellular ROS, caspase-3 activity, HMGB1 protein expression, and some alterations to the cell cycle. Hydrogen Peroxide 216-220 high mobility group box 1 Homo sapiens 345-350 23390034-8 2013 CONCLUSION: Elevations in plasma miR-122, HMGB1, and necrosis K18 identified subsequent ALI development in patients on admission to the hospital, soon after acetaminophen overdose, and in patients with ALTs in the normal range. ali 88-91 high mobility group box 1 Homo sapiens 42-47 23712703-4 2013 However, the receptor for advanced glycation end products (RAGE) blocker RAGE-Ab (5 mug/ml) or 10 muM c-Jun N-terminal kinases (JNK) inhibitor SP600125 could inhibit HMGB1-induced the release of inflammation cytokines including TNF-alpha, IL-8, IL-10, and MCP-1 in a dose-dependent manner. pyrazolanthrone 143-151 high mobility group box 1 Homo sapiens 166-171 23712703-5 2013 Furthermore, HMGB1-induced RAGE protein expression, JNK and NF-kappaB activation were attenuated by the pretreatment with RAGE-Ab or JNK inhibitor SP600125 in Western blot analysis. pyrazolanthrone 147-155 high mobility group box 1 Homo sapiens 13-18 23737485-6 2013 CatmAb promoted tumor cell death associated with ATP release and strongly synergized with oxaliplatin for the exposure of the three hallmarks of immunogenic cell death (calreticulin, HMGB1, and ATP). Oxaliplatin 90-101 high mobility group box 1 Homo sapiens 183-188 23207101-5 2013 Notably, only one of the redox forms of HMGB1, the one where all cysteines are reduced (all-thiol), can bind CXCL12. Sulfhydryl Compounds 92-97 high mobility group box 1 Homo sapiens 40-45 23207101-6 2013 Both HMGB1 containing a disulfide bond between C23 and C45, which induces chemokine and cytokine release by activating TLR4, and HMGB1 terminally oxidized to contain a cysteine sulfonate are inactive in recruiting leukocytes. Disulfides 24-33 high mobility group box 1 Homo sapiens 5-10 23207101-6 2013 Both HMGB1 containing a disulfide bond between C23 and C45, which induces chemokine and cytokine release by activating TLR4, and HMGB1 terminally oxidized to contain a cysteine sulfonate are inactive in recruiting leukocytes. cysteine sulfonate 168-186 high mobility group box 1 Homo sapiens 129-134 23835906-0 2013 Carbenoxolone blocks endotoxin-induced protein kinase R (PKR) activation and high mobility group box 1 (HMGB1) release. Carbenoxolone 0-13 high mobility group box 1 Homo sapiens 77-102 23835906-7 2013 Collectively, these results suggested that CBX blocks LPS-induced HMGB1 release possibly through impairing PKR activation, supporting the involvement of PKR in the regulation of HMGB1 release. Carbenoxolone 43-46 high mobility group box 1 Homo sapiens 66-71 23835906-0 2013 Carbenoxolone blocks endotoxin-induced protein kinase R (PKR) activation and high mobility group box 1 (HMGB1) release. Carbenoxolone 0-13 high mobility group box 1 Homo sapiens 104-109 23835906-7 2013 Collectively, these results suggested that CBX blocks LPS-induced HMGB1 release possibly through impairing PKR activation, supporting the involvement of PKR in the regulation of HMGB1 release. Carbenoxolone 43-46 high mobility group box 1 Homo sapiens 178-183 23835906-1 2013 The pathogen- and damage-associated molecular patterns (for example, bacterial endotoxin and adenosine 5"-triphosphate [ATP]) activate the double-stranded RNA-activated protein kinase R (PKR) to trigger the inflammasome-dependent high mobility group box 1 (HMGB1) release. Adenosine Triphosphate 93-118 high mobility group box 1 Homo sapiens 230-255 23835906-1 2013 The pathogen- and damage-associated molecular patterns (for example, bacterial endotoxin and adenosine 5"-triphosphate [ATP]) activate the double-stranded RNA-activated protein kinase R (PKR) to trigger the inflammasome-dependent high mobility group box 1 (HMGB1) release. Adenosine Triphosphate 93-118 high mobility group box 1 Homo sapiens 257-262 23835906-1 2013 The pathogen- and damage-associated molecular patterns (for example, bacterial endotoxin and adenosine 5"-triphosphate [ATP]) activate the double-stranded RNA-activated protein kinase R (PKR) to trigger the inflammasome-dependent high mobility group box 1 (HMGB1) release. Adenosine Triphosphate 120-123 high mobility group box 1 Homo sapiens 230-255 23835906-1 2013 The pathogen- and damage-associated molecular patterns (for example, bacterial endotoxin and adenosine 5"-triphosphate [ATP]) activate the double-stranded RNA-activated protein kinase R (PKR) to trigger the inflammasome-dependent high mobility group box 1 (HMGB1) release. Adenosine Triphosphate 120-123 high mobility group box 1 Homo sapiens 257-262 23711070-5 2013 There was a positive relationship between the BEBSS and HMGB1 levels (r = 0.54, P = 0.004). bebss 46-51 high mobility group box 1 Homo sapiens 56-61 23576640-6 2013 Glucose (11.2 mM) maximally increased TLR2 and 4 expression, HMGB1 release, and NF-kappaB activation with increased expression of cytokines. Glucose 0-7 high mobility group box 1 Homo sapiens 61-66 23576640-10 2013 Therefore, short-term moderate increases in glucose in vitro increase HMGB1, which mediates NF-kappaB activation through both TLR2 and 4. Glucose 44-51 high mobility group box 1 Homo sapiens 70-75 23421467-7 2013 The LMWH significantly inhibited HMGB1-induced NFkappaB activation through RAGE using an NFkappaB-dependent luciferase reporter assay and the HT1080 cell lines. Heparin, Low-Molecular-Weight 4-8 high mobility group box 1 Homo sapiens 33-38 23446148-3 2013 HMGB1 contains three conserved redox-sensitive cysteines (C23, C45, and C106); modification of these cysteines determines the bioactivity of extracellular HMGB1. Cysteine 47-56 high mobility group box 1 Homo sapiens 0-5 23446148-3 2013 HMGB1 contains three conserved redox-sensitive cysteines (C23, C45, and C106); modification of these cysteines determines the bioactivity of extracellular HMGB1. Cysteine 47-56 high mobility group box 1 Homo sapiens 155-160 23446148-3 2013 HMGB1 contains three conserved redox-sensitive cysteines (C23, C45, and C106); modification of these cysteines determines the bioactivity of extracellular HMGB1. Cysteine 101-110 high mobility group box 1 Homo sapiens 0-5 23446148-3 2013 HMGB1 contains three conserved redox-sensitive cysteines (C23, C45, and C106); modification of these cysteines determines the bioactivity of extracellular HMGB1. Cysteine 101-110 high mobility group box 1 Homo sapiens 155-160 23446148-4 2013 Firstly, the cytokine-stimulating activity of HMGB1 requires C23 and C45 to be in a disulfide linkage, at the same time that C106 must remain in its reduced form as a thiol. Disulfides 84-93 high mobility group box 1 Homo sapiens 46-51 23446148-4 2013 Firstly, the cytokine-stimulating activity of HMGB1 requires C23 and C45 to be in a disulfide linkage, at the same time that C106 must remain in its reduced form as a thiol. Sulfhydryl Compounds 167-172 high mobility group box 1 Homo sapiens 46-51 23446148-6 2013 Secondly, for HMGB1 to act as a chemotactic mediator, all three cysteines must be in the reduced form. Cysteine 64-73 high mobility group box 1 Homo sapiens 14-19 23446148-7 2013 This all-thiol HMGB1 exerts its chemotactic activity to initiate inflammation by forming a heterocomplex with CXCL12; that complex binds exclusively to CXCR4 to initiate chemotaxis. Sulfhydryl Compounds 9-14 high mobility group box 1 Homo sapiens 15-20 23446148-8 2013 Thirdly, binding of the HMGB1 to CXCR4 or to TLR4 is completely prevented by all-cysteine oxidation. all-cysteine 77-89 high mobility group box 1 Homo sapiens 24-29 23446148-10 2013 Lastly, post-translational acetylation of key lysine residues within NLSs of HMGB1 affects HMGB1 to promote inflammation; hyperacetylation of HMGB1 shifts its equilibrium from a predominant nuclear location toward a cytosolic and subsequent extracellular presence. Lysine 46-52 high mobility group box 1 Homo sapiens 77-82 23446148-10 2013 Lastly, post-translational acetylation of key lysine residues within NLSs of HMGB1 affects HMGB1 to promote inflammation; hyperacetylation of HMGB1 shifts its equilibrium from a predominant nuclear location toward a cytosolic and subsequent extracellular presence. Lysine 46-52 high mobility group box 1 Homo sapiens 91-96 23446148-10 2013 Lastly, post-translational acetylation of key lysine residues within NLSs of HMGB1 affects HMGB1 to promote inflammation; hyperacetylation of HMGB1 shifts its equilibrium from a predominant nuclear location toward a cytosolic and subsequent extracellular presence. Lysine 46-52 high mobility group box 1 Homo sapiens 91-96 23042518-0 2013 Barrier protective effects of rosmarinic acid on HMGB1-induced inflammatory responses in vitro and in vivo. rosmarinic acid 30-45 high mobility group box 1 Homo sapiens 49-54 23508573-1 2013 High mobility group box 1 (HMGB1) is a DNA-binding protein that possesses cytokinelike, proinflammatory properties when released extracellularly in the C23-C45 disulfide form. Disulfides 160-169 high mobility group box 1 Homo sapiens 0-25 23508573-1 2013 High mobility group box 1 (HMGB1) is a DNA-binding protein that possesses cytokinelike, proinflammatory properties when released extracellularly in the C23-C45 disulfide form. Disulfides 160-169 high mobility group box 1 Homo sapiens 27-32 23508573-6 2013 TLR4 and MD2 are recruited into CD14-containing lipid rafts of RAW264.7 macrophages after stimulation with HMGB1, and TLR4 interacts closely with the lipid raft protein GM1. G(M1) Ganglioside 169-172 high mobility group box 1 Homo sapiens 107-112 23628898-7 2013 One of the mechanisms underlying these activating effects of ICs on human endothelial cells involves cell signaling by high-mobility group box 1 protein (HMGB1)-RAGE axis, as these effects can be partially blocked by HMGB1 A-box, soluble RAGE (sRAGE), SB203580, PD98059, Bay 117082 (P<0.05) and co-treatment with these agents (P<0.05). SB 203580 252-260 high mobility group box 1 Homo sapiens 119-144 23628898-7 2013 One of the mechanisms underlying these activating effects of ICs on human endothelial cells involves cell signaling by high-mobility group box 1 protein (HMGB1)-RAGE axis, as these effects can be partially blocked by HMGB1 A-box, soluble RAGE (sRAGE), SB203580, PD98059, Bay 117082 (P<0.05) and co-treatment with these agents (P<0.05). SB 203580 252-260 high mobility group box 1 Homo sapiens 154-159 23628898-7 2013 One of the mechanisms underlying these activating effects of ICs on human endothelial cells involves cell signaling by high-mobility group box 1 protein (HMGB1)-RAGE axis, as these effects can be partially blocked by HMGB1 A-box, soluble RAGE (sRAGE), SB203580, PD98059, Bay 117082 (P<0.05) and co-treatment with these agents (P<0.05). 2-(2-amino-3-methoxyphenyl)-4H-1-benzopyran-4-one 262-269 high mobility group box 1 Homo sapiens 119-144 23628898-7 2013 One of the mechanisms underlying these activating effects of ICs on human endothelial cells involves cell signaling by high-mobility group box 1 protein (HMGB1)-RAGE axis, as these effects can be partially blocked by HMGB1 A-box, soluble RAGE (sRAGE), SB203580, PD98059, Bay 117082 (P<0.05) and co-treatment with these agents (P<0.05). 2-(2-amino-3-methoxyphenyl)-4H-1-benzopyran-4-one 262-269 high mobility group box 1 Homo sapiens 154-159 23628898-7 2013 One of the mechanisms underlying these activating effects of ICs on human endothelial cells involves cell signaling by high-mobility group box 1 protein (HMGB1)-RAGE axis, as these effects can be partially blocked by HMGB1 A-box, soluble RAGE (sRAGE), SB203580, PD98059, Bay 117082 (P<0.05) and co-treatment with these agents (P<0.05). 3-(4-methylphenylsulfonyl)-2-propenenitrile 271-281 high mobility group box 1 Homo sapiens 119-144 23628898-7 2013 One of the mechanisms underlying these activating effects of ICs on human endothelial cells involves cell signaling by high-mobility group box 1 protein (HMGB1)-RAGE axis, as these effects can be partially blocked by HMGB1 A-box, soluble RAGE (sRAGE), SB203580, PD98059, Bay 117082 (P<0.05) and co-treatment with these agents (P<0.05). 3-(4-methylphenylsulfonyl)-2-propenenitrile 271-281 high mobility group box 1 Homo sapiens 154-159 23042518-2 2013 Rosmarinic acid (RA) is an important component of the leaves of Perilla frutescens and has neuroprotective, anti-microbial, anti-oxidant, and anti-cancer effects but little is known of its effects on HMGB1-mediated inflammatory response. rosmarinic acid 17-19 high mobility group box 1 Homo sapiens 200-205 23042518-3 2013 Here, we investigated this issue by monitoring the effects of RA on the lipopolysaccharide (LPS) or cecal ligation and puncture (CLP)-mediated release of HMGB1 and HMGB1-mediated modulation of inflammatory responses. rosmarinic acid 62-64 high mobility group box 1 Homo sapiens 154-159 23042518-4 2013 RA potently inhibited the release of HMGB1 and down-regulated HMGB1-dependent inflammatory responses in human endothelial cells. rosmarinic acid 0-2 high mobility group box 1 Homo sapiens 37-42 23042518-4 2013 RA potently inhibited the release of HMGB1 and down-regulated HMGB1-dependent inflammatory responses in human endothelial cells. rosmarinic acid 0-2 high mobility group box 1 Homo sapiens 62-67 23042518-6 2013 Furthermore, RA reduced CLP-induced HMGB1 release and sepsis-related mortality. rosmarinic acid 13-15 high mobility group box 1 Homo sapiens 36-41 23042518-7 2013 Given these results, RA should be viewed as a candidate therapeutic agent for the treatment of various inflammatory diseases via inhibition of the HMGB1 signaling pathway. rosmarinic acid 21-23 high mobility group box 1 Homo sapiens 147-152 22939552-0 2013 Dexmedetomidine inhibits the secretion of high mobility group box 1 from lipopolysaccharide-activated macrophages in vitro. Dexmedetomidine 0-15 high mobility group box 1 Homo sapiens 42-67 22939552-4 2013 In this study, we explored the effect of dexmedetomidine on the expression and secretion of HMGB1 from lipopolysaccharide (LPS)-activated macrophages. Dexmedetomidine 41-56 high mobility group box 1 Homo sapiens 92-97 22939552-10 2013 RESULTS: Dexmedetomidine inhibited the translocation of HMGB1 from the nucleus to the cytoplasm and its extracellular secretion in LPS-activated macrophages while suppressing the expression of HMGB1 mRNA. Dexmedetomidine 9-24 high mobility group box 1 Homo sapiens 56-61 22939552-10 2013 RESULTS: Dexmedetomidine inhibited the translocation of HMGB1 from the nucleus to the cytoplasm and its extracellular secretion in LPS-activated macrophages while suppressing the expression of HMGB1 mRNA. Dexmedetomidine 9-24 high mobility group box 1 Homo sapiens 193-198 22939552-13 2013 CONCLUSIONS: Our study demonstrates that dexmedetomidine inhibits the translocation of HMGB1 from the nucleus to the cytoplasm and the expression of HMGB1 mRNA at clinically relevant dosages. Dexmedetomidine 41-56 high mobility group box 1 Homo sapiens 87-92 22939552-13 2013 CONCLUSIONS: Our study demonstrates that dexmedetomidine inhibits the translocation of HMGB1 from the nucleus to the cytoplasm and the expression of HMGB1 mRNA at clinically relevant dosages. Dexmedetomidine 41-56 high mobility group box 1 Homo sapiens 149-154 23488692-0 2013 IL-1beta/HMGB1 complexes promote The PGE2 biosynthesis pathway in synovial fibroblasts. Dinoprostone 37-41 high mobility group box 1 Homo sapiens 9-14 23488692-6 2013 We aimed to investigate the effects of IL-1beta/HMGB1 complexes on mPGES-1 and other enzymes of the PGE2 pathway in synovial fibroblasts (SFs) from patients with arthritis. Dinoprostone 100-104 high mobility group box 1 Homo sapiens 48-53 23488692-8 2013 Stimulation of SFs with HMGB1 in complex with suboptimal amounts of IL-1beta significantly increased mPGES-1 and COX-2 expressions as well as PGE2 production, as compared to treatment with HMGB1 or IL-1beta alone. SFS 15-18 high mobility group box 1 Homo sapiens 24-29 23488692-8 2013 Stimulation of SFs with HMGB1 in complex with suboptimal amounts of IL-1beta significantly increased mPGES-1 and COX-2 expressions as well as PGE2 production, as compared to treatment with HMGB1 or IL-1beta alone. SFS 15-18 high mobility group box 1 Homo sapiens 189-194 23488692-8 2013 Stimulation of SFs with HMGB1 in complex with suboptimal amounts of IL-1beta significantly increased mPGES-1 and COX-2 expressions as well as PGE2 production, as compared to treatment with HMGB1 or IL-1beta alone. Dinoprostone 142-146 high mobility group box 1 Homo sapiens 24-29 23488692-9 2013 Furthermore, NS-398 reduced the production of IL-6 and IL-8, thus indicating that IL-1beta/HMGB1 complexes modulate cytokine production in part through prostanoid synthesis. N-(2-cyclohexyloxy-4-nitrophenyl)methanesulfonamide 13-19 high mobility group box 1 Homo sapiens 91-96 23488692-9 2013 Furthermore, NS-398 reduced the production of IL-6 and IL-8, thus indicating that IL-1beta/HMGB1 complexes modulate cytokine production in part through prostanoid synthesis. Prostaglandins 152-162 high mobility group box 1 Homo sapiens 91-96 23488692-11 2013 IL-1beta/HMGB1 complexes promote the induction of mPGES-1, COX-2 and PGE2 in SF. Dinoprostone 69-73 high mobility group box 1 Homo sapiens 9-14 23488692-12 2013 The amplification of the PGE2 biosynthesis pathway by HMGB1 might constitute an important pathogenic mechanism perpetuating inflammatory and destructive activities in rheumatoid arthritis. Dinoprostone 25-29 high mobility group box 1 Homo sapiens 54-59 23985250-2 2013 METHODS: HMGB1 gene targeting siRNA was designed and synthesized, and HMGB1 siRNA oligonucleotides were transfected into the MGC-803 cells with Lipofectamine 2000. Oligonucleotides 82-98 high mobility group box 1 Homo sapiens 70-75 23447529-4 2013 Using a unique NMR-based approach, we have investigated the kinetics of HMGB1 oxidation and the half-lives of all-thiol and disulfide HMGB1 species in serum, saliva, and cell culture medium. Disulfides 124-133 high mobility group box 1 Homo sapiens 134-139 23447529-5 2013 In this approach, salt-free lyophilized (15)N-labeled all-thiol HMGB1 was dissolved in actual extracellular fluids, and the oxidation and clearance kinetics were monitored in situ by recording a series of heteronuclear (1)H-(15)N correlation spectra. Salts 18-22 high mobility group box 1 Homo sapiens 64-69 23447529-5 2013 In this approach, salt-free lyophilized (15)N-labeled all-thiol HMGB1 was dissolved in actual extracellular fluids, and the oxidation and clearance kinetics were monitored in situ by recording a series of heteronuclear (1)H-(15)N correlation spectra. Sulfhydryl Compounds 58-63 high mobility group box 1 Homo sapiens 64-69 23447529-7 2013 For example, the half-life of all-thiol HMGB1 ranged from ~17 min (in human serum and saliva) to 3 h (in prostate cancer cell culture medium). Sulfhydryl Compounds 34-39 high mobility group box 1 Homo sapiens 40-45 23447529-8 2013 Furthermore, the binding of ligands (glycyrrhizin and heparin) to HMGB1 significantly modulated the oxidation kinetics. Glycyrrhizic Acid 37-49 high mobility group box 1 Homo sapiens 66-71 23447529-8 2013 Furthermore, the binding of ligands (glycyrrhizin and heparin) to HMGB1 significantly modulated the oxidation kinetics. Heparin 54-61 high mobility group box 1 Homo sapiens 66-71 23447529-9 2013 Thus, the balance between the roles of all-thiol and disulfide HMGB1 proteins depends significantly on the extracellular environment and can also be artificially modulated by ligands. Disulfides 53-62 high mobility group box 1 Homo sapiens 63-68 22911316-4 2013 Persicarin potently inhibited the release of HMGB1 and down-regulated HMGB1-dependent inflammatory responses in human endothelial cells, and inhibited HMGB1-mediated hyperpermeability and leukocyte migration in mice. persicarin 0-10 high mobility group box 1 Homo sapiens 45-50 22911316-4 2013 Persicarin potently inhibited the release of HMGB1 and down-regulated HMGB1-dependent inflammatory responses in human endothelial cells, and inhibited HMGB1-mediated hyperpermeability and leukocyte migration in mice. persicarin 0-10 high mobility group box 1 Homo sapiens 70-75 22911316-4 2013 Persicarin potently inhibited the release of HMGB1 and down-regulated HMGB1-dependent inflammatory responses in human endothelial cells, and inhibited HMGB1-mediated hyperpermeability and leukocyte migration in mice. persicarin 0-10 high mobility group box 1 Homo sapiens 70-75 23617783-9 2013 We demonstrated that patients with MPM had significantly higher serum levels of HMGB1 than the population who had been exposed to asbestos but had not developed MPM. Asbestos 130-138 high mobility group box 1 Homo sapiens 80-85 23403945-0 2013 Insulin infusion suppresses while glucose infusion induces Toll-like receptors and high-mobility group-B1 protein expression in mononuclear cells of type 1 diabetes patients. Glucose 34-41 high mobility group box 1 Homo sapiens 83-105 23403945-4 2013 Glucose infusion led to an increase in plasma glucose concentration from 115 (fasting) to 215 (at 4 and 6 h) mg/dl and to an increase in ROS generation, the expression of TLR-4, TLR-2, TLR-1, HMGB1, p38 MAP kinase, and JNK-1, and plasma concentrations of HMGB1. Glucose 0-7 high mobility group box 1 Homo sapiens 192-197 23403945-4 2013 Glucose infusion led to an increase in plasma glucose concentration from 115 (fasting) to 215 (at 4 and 6 h) mg/dl and to an increase in ROS generation, the expression of TLR-4, TLR-2, TLR-1, HMGB1, p38 MAP kinase, and JNK-1, and plasma concentrations of HMGB1. Glucose 0-7 high mobility group box 1 Homo sapiens 255-260 23206318-5 2013 Postmortem human alcoholic brain also showed increased HMGB1, TLR2, TLR3, and TLR4 +IR cells that correlated with lifetime alcohol consumption, as well as each other. Alcohols 17-24 high mobility group box 1 Homo sapiens 55-60 24250621-9 2013 5-FU also induced HMGB1 secretion, Cas3 and PARP activity and these effects were stronger in cells expressing WT TLR4 than the other cells. Fluorouracil 0-4 high mobility group box 1 Homo sapiens 18-23 23352503-0 2013 Inhibitory effects of epi-sesamin on HMGB1-induced vascular barrier disruptive responses in vitro and in vivo. sesamin 22-33 high mobility group box 1 Homo sapiens 37-42 23384711-9 2013 At multivariate analysis, HMGB1 was found to be independently correlated with BMI, IL23, IL6, free triiodothyronine, HDL, and HOMA-IR. Triiodothyronine 99-115 high mobility group box 1 Homo sapiens 26-31 23410002-0 2013 Plasma HMGB-1 after the initial dose of epirubicin/docetaxel in cancer. Epirubicin 40-50 high mobility group box 1 Homo sapiens 7-13 23410002-6 2013 RESULTS: Treatment of HCC1143 cells with epirubicin/docetaxel resulted in a significant HMGB-1 release in vitro. Epirubicin 41-51 high mobility group box 1 Homo sapiens 88-94 23410002-0 2013 Plasma HMGB-1 after the initial dose of epirubicin/docetaxel in cancer. Docetaxel 51-60 high mobility group box 1 Homo sapiens 7-13 23410002-6 2013 RESULTS: Treatment of HCC1143 cells with epirubicin/docetaxel resulted in a significant HMGB-1 release in vitro. Docetaxel 52-61 high mobility group box 1 Homo sapiens 88-94 23410002-3 2013 We hypothesized that the initial dose of NCT with epirubicin/docetaxel induces changes in plasma HMGB-1 which could allow for an early prediction of response to therapy. Epirubicin 50-60 high mobility group box 1 Homo sapiens 97-103 23410002-3 2013 We hypothesized that the initial dose of NCT with epirubicin/docetaxel induces changes in plasma HMGB-1 which could allow for an early prediction of response to therapy. Docetaxel 61-70 high mobility group box 1 Homo sapiens 97-103 23410002-4 2013 MATERIALS AND METHODS: First, we analysed whether epirubicin/docetaxel releases HMGB-1 from HCC1143 breast cancer cells in vitro. Epirubicin 50-60 high mobility group box 1 Homo sapiens 80-86 23410002-4 2013 MATERIALS AND METHODS: First, we analysed whether epirubicin/docetaxel releases HMGB-1 from HCC1143 breast cancer cells in vitro. Docetaxel 61-70 high mobility group box 1 Homo sapiens 80-86 23222484-7 2013 Importantly, priming through surface TLRs but not endosomal TLRs during pyroptosis leads to the release of a new TLR4-agonist cysteine redox isoform of HMGB1. Cysteine 126-134 high mobility group box 1 Homo sapiens 152-157 23808169-0 2013 [The structure of the complexes of DNA with nonhistone chromosomal protein HMGB1 and the histone H1 in the presence of manganese ions. Manganese 119-128 high mobility group box 1 Homo sapiens 75-80 23808169-3 2013 Complexes of DNA with nonhistone chromosomal protein HMGB1 and histone H1 in the presence of manganese ions were studied using absorption and circular dichroism spectroscopy in infrared region. Manganese 93-102 high mobility group box 1 Homo sapiens 53-58 23808169-5 2013 It was also shown that the manganese ions are able to coordinate to the chemical groups of DNA as well as to the carboxylic amino acid residues of the protein HMGB1. Manganese 27-36 high mobility group box 1 Homo sapiens 159-164 23808169-5 2013 It was also shown that the manganese ions are able to coordinate to the chemical groups of DNA as well as to the carboxylic amino acid residues of the protein HMGB1. carboxylic amino acid 113-134 high mobility group box 1 Homo sapiens 159-164 23325855-2 2013 HMGB1/2 play a critical, although poorly understood, role in vitro in the assembly of functional RAG-RSS complexes, into which HMGB1/2 stably incorporate. rag-rss 97-104 high mobility group box 1 Homo sapiens 0-5 23325855-2 2013 HMGB1/2 play a critical, although poorly understood, role in vitro in the assembly of functional RAG-RSS complexes, into which HMGB1/2 stably incorporate. rag-rss 97-104 high mobility group box 1 Homo sapiens 127-132 23325855-4 2013 Here, we report data demonstrating only a weak HMGB1-RAG1 interaction in the absence of DNA in several assays, including fluorescence anisotropy experiments using a novel Alexa488-labeled HMGB1 protein. alexa488 171-179 high mobility group box 1 Homo sapiens 188-193 23333393-0 2013 Inhibitory effects of berberine on lipopolysaccharide-induced inducible nitric oxide synthase and the high-mobility group box 1 release in macrophages. Berberine 22-31 high mobility group box 1 Homo sapiens 102-127 23333393-1 2013 We investigated the molecular mechanism by which berberine reduces nitric oxide (NO) expression and high-mobility group box 1 (HMGB1) release in lipopolysaccharide (LPS)-induced macrophages. Berberine 49-58 high mobility group box 1 Homo sapiens 100-125 23333393-1 2013 We investigated the molecular mechanism by which berberine reduces nitric oxide (NO) expression and high-mobility group box 1 (HMGB1) release in lipopolysaccharide (LPS)-induced macrophages. Berberine 49-58 high mobility group box 1 Homo sapiens 127-132 23333393-2 2013 Berberine significantly inhibited the LPS-stimulated NO production and HMGB1 release in macrophages. Berberine 0-9 high mobility group box 1 Homo sapiens 71-76 23333393-4 2013 The inhibitory effect of berberine on LPS-stimulated NO and HMGB1 release was reversed by siRNA-Nrf2, SB203580 (p38 MAPK inhibitor) and zinc protoporphyrin (ZnPP; HO-1 inhibitor) within macrophages. Berberine 25-34 high mobility group box 1 Homo sapiens 60-65 23333393-4 2013 The inhibitory effect of berberine on LPS-stimulated NO and HMGB1 release was reversed by siRNA-Nrf2, SB203580 (p38 MAPK inhibitor) and zinc protoporphyrin (ZnPP; HO-1 inhibitor) within macrophages. SB 203580 102-110 high mobility group box 1 Homo sapiens 60-65 23333393-4 2013 The inhibitory effect of berberine on LPS-stimulated NO and HMGB1 release was reversed by siRNA-Nrf2, SB203580 (p38 MAPK inhibitor) and zinc protoporphyrin (ZnPP; HO-1 inhibitor) within macrophages. zinc protoporphyrin 136-155 high mobility group box 1 Homo sapiens 60-65 23333393-4 2013 The inhibitory effect of berberine on LPS-stimulated NO and HMGB1 release was reversed by siRNA-Nrf2, SB203580 (p38 MAPK inhibitor) and zinc protoporphyrin (ZnPP; HO-1 inhibitor) within macrophages. zinc protoporphyrin 157-161 high mobility group box 1 Homo sapiens 60-65 23303669-5 2013 HMGB1 directly interacted with polyQ on immunofluorescence and coimmunoprecipitation assay, whereas the overexpression of HMGB1 or exogenous administration of recombinant HMGB1 protein remarkably reduced polyQ aggregates in SHSY5Y cells and hmgb1(-/-) mouse embryonic fibroblasts upon filter trap and immunofluorescence assay. polyglutamine 31-36 high mobility group box 1 Homo sapiens 0-5 23303669-5 2013 HMGB1 directly interacted with polyQ on immunofluorescence and coimmunoprecipitation assay, whereas the overexpression of HMGB1 or exogenous administration of recombinant HMGB1 protein remarkably reduced polyQ aggregates in SHSY5Y cells and hmgb1(-/-) mouse embryonic fibroblasts upon filter trap and immunofluorescence assay. polyglutamine 204-209 high mobility group box 1 Homo sapiens 0-5 23303669-5 2013 HMGB1 directly interacted with polyQ on immunofluorescence and coimmunoprecipitation assay, whereas the overexpression of HMGB1 or exogenous administration of recombinant HMGB1 protein remarkably reduced polyQ aggregates in SHSY5Y cells and hmgb1(-/-) mouse embryonic fibroblasts upon filter trap and immunofluorescence assay. polyglutamine 204-209 high mobility group box 1 Homo sapiens 122-127 23303669-5 2013 HMGB1 directly interacted with polyQ on immunofluorescence and coimmunoprecipitation assay, whereas the overexpression of HMGB1 or exogenous administration of recombinant HMGB1 protein remarkably reduced polyQ aggregates in SHSY5Y cells and hmgb1(-/-) mouse embryonic fibroblasts upon filter trap and immunofluorescence assay. polyglutamine 204-209 high mobility group box 1 Homo sapiens 122-127 23241947-6 2013 Our results suggest that PEF suppresses LPC-induced inflammatory factor production through inhibition of the HMGB1-RAGE/TLR-2/TLR-4-NF-kappaB pathway. Lysophosphatidylcholines 40-43 high mobility group box 1 Homo sapiens 109-114 23414442-7 2013 RA 50 mg/kg attenuated histopathological damage, decreased brain edema, inhibited NF-kappaB activation and reduced HMGB1 expression. rosmarinic acid 0-2 high mobility group box 1 Homo sapiens 115-120 23414442-8 2013 CONCLUSION: These data show that RA protects the brain against I/R injury with a favorable therapeutic time-window by alleviating diabetic cerebral I/R injury and attenuating blood-brain barrier (BBB) breakdown, and its protective effects may involve HMGB1 and the NF-kappaB signaling pathway. rosmarinic acid 33-35 high mobility group box 1 Homo sapiens 251-256 24148794-9 2013 Moreover, H2 treatment dose-dependently attenuated the increased levels of pro-inflammatory cytokines (TNF-alpha, IL-1beta, HMGB1), but further increased the level of anti-inflammatory cytokine IL-10 at 3 h, 6 h, 12 h and 24 h after LPS stimulation. Hydrogen 10-12 high mobility group box 1 Homo sapiens 124-129 24289579-10 2013 Methylation of HMGB1 at lysine 112 in ccRCC was detected by MS. Bioinformatics analysis showed that post-translational modification might affect the binding ability to DNA and mediate its translocation. Lysine 24-30 high mobility group box 1 Homo sapiens 15-20 24289579-12 2013 Methylation of HMGB1 at lysine 112 might the redistribution of this cofactor protein. Lysine 24-30 high mobility group box 1 Homo sapiens 15-20 24191125-6 2013 The UA-HMGB1 level in the BHT (-) group did not differ significantly from reference values, but was significantly increased 24 hours after birth. Butylated Hydroxytoluene 26-29 high mobility group box 1 Homo sapiens 7-12 23036589-0 2013 Evidence for the binding affinity of daunomycin to HMGB1 protein in chromatin and in solution. Daunorubicin 37-47 high mobility group box 1 Homo sapiens 51-56 23036589-1 2013 In this study the interaction of daunomycin with HMGB1 nonhistone chromatin protein in the chromatin context using hydroxyapatite (HAP) column chromatography and free in solution was investigated employing fluorescence, circular dichroism spectroscopy and thermal denaturation techniques. Daunorubicin 33-43 high mobility group box 1 Homo sapiens 49-54 23036589-1 2013 In this study the interaction of daunomycin with HMGB1 nonhistone chromatin protein in the chromatin context using hydroxyapatite (HAP) column chromatography and free in solution was investigated employing fluorescence, circular dichroism spectroscopy and thermal denaturation techniques. Durapatite 115-129 high mobility group box 1 Homo sapiens 49-54 23036589-2 2013 The results demonstrate that HMGB1 fraction eluted from HAP column contained the most amount of daunomycin. Daunorubicin 96-106 high mobility group box 1 Homo sapiens 29-34 23036589-3 2013 Upon addition of daunomycin to HMGB1 solution, fluorescence emission intensity was dependent on the drug concentration used whereas the ellipticity in CD spectra was decreased at both 205 and 220 nm extremes implying that quenching of the drug with the HMGB1 chromospheres alters secondary structure of the protein. Daunorubicin 17-27 high mobility group box 1 Homo sapiens 31-36 23036589-3 2013 Upon addition of daunomycin to HMGB1 solution, fluorescence emission intensity was dependent on the drug concentration used whereas the ellipticity in CD spectra was decreased at both 205 and 220 nm extremes implying that quenching of the drug with the HMGB1 chromospheres alters secondary structure of the protein. Daunorubicin 17-27 high mobility group box 1 Homo sapiens 253-258 23036589-3 2013 Upon addition of daunomycin to HMGB1 solution, fluorescence emission intensity was dependent on the drug concentration used whereas the ellipticity in CD spectra was decreased at both 205 and 220 nm extremes implying that quenching of the drug with the HMGB1 chromospheres alters secondary structure of the protein. Cadmium 151-153 high mobility group box 1 Homo sapiens 253-258 23036589-4 2013 Although daunomycin slightly increased the melting point of HMGB1, but exhibited a significant hyperchromicity at low concentrations and hypochromicity at higher concentrations of daunomycin. Daunorubicin 9-19 high mobility group box 1 Homo sapiens 60-65 23036589-5 2013 The results suggest that daunomycin binds to HMGB1 protein which may influence its interaction with DNA in nucleosomes and other cellular processes. Daunorubicin 25-35 high mobility group box 1 Homo sapiens 45-50 23073660-4 2012 Reduced all-thiol-HMGB1 has sole chemokine activity, whereas disulfide-HMGB1 has only cytokine activity, and oxidized, denatured HMGB1 has neither. Sulfhydryl Compounds 12-17 high mobility group box 1 Homo sapiens 18-23 23042506-11 2013 Knockdown of HMGB1 downregulated the expression of p-AKT, Ki-67 and MMP-2, inhibited the proliferative activities and metastatic potential of SMMC-7721 cells, induced cell cycle arrest and apoptosis, and slowed the growth of xenograft tumors. smmc 166-170 high mobility group box 1 Homo sapiens 13-18 22487382-0 2013 Magnesium sulfate inhibits the secretion of high mobility group box 1 from lipopolysaccharide-activated RAW264.7 macrophages in vitro. Magnesium Sulfate 0-17 high mobility group box 1 Homo sapiens 44-69 22487382-4 2013 In the present study, we explored the effect of magnesium sulfate on the expression and release of HMGB1 in lipopolysaccharide (LPS)-activated macrophages. Magnesium Sulfate 48-65 high mobility group box 1 Homo sapiens 99-104 22487382-11 2013 RESULTS: We found that magnesium sulfate inhibited translocation of HMGB1 from the nucleus to the cytoplasm and its extracellular release in LPS-activated macrophages and also suppressed the expression of HMGB1 mRNA. Magnesium Sulfate 23-40 high mobility group box 1 Homo sapiens 68-73 22487382-11 2013 RESULTS: We found that magnesium sulfate inhibited translocation of HMGB1 from the nucleus to the cytoplasm and its extracellular release in LPS-activated macrophages and also suppressed the expression of HMGB1 mRNA. Magnesium Sulfate 23-40 high mobility group box 1 Homo sapiens 205-210 22487382-13 2013 CONCLUSIONS: Our study has demonstrated that magnesium sulfate inhibits the translocation of HMGB1 from the nucleus to the cytoplasm and the expression of HMGB1 mRNA in a dose-dependent manner. Magnesium Sulfate 45-62 high mobility group box 1 Homo sapiens 93-98 22487382-13 2013 CONCLUSIONS: Our study has demonstrated that magnesium sulfate inhibits the translocation of HMGB1 from the nucleus to the cytoplasm and the expression of HMGB1 mRNA in a dose-dependent manner. Magnesium Sulfate 45-62 high mobility group box 1 Homo sapiens 155-160 24302816-7 2013 Furthermore, both NF- kappa B inhibitor Bay11-7085 and p38 inhibitor SB203580 significantly suppressed the enhanced production of IL-6 and MMPs induced by HMGB1-LPS. BAY 11-7085 40-50 high mobility group box 1 Homo sapiens 155-160 24302816-7 2013 Furthermore, both NF- kappa B inhibitor Bay11-7085 and p38 inhibitor SB203580 significantly suppressed the enhanced production of IL-6 and MMPs induced by HMGB1-LPS. SB 203580 69-77 high mobility group box 1 Homo sapiens 155-160 23073660-4 2012 Reduced all-thiol-HMGB1 has sole chemokine activity, whereas disulfide-HMGB1 has only cytokine activity, and oxidized, denatured HMGB1 has neither. Disulfides 61-70 high mobility group box 1 Homo sapiens 71-76 23073660-4 2012 Reduced all-thiol-HMGB1 has sole chemokine activity, whereas disulfide-HMGB1 has only cytokine activity, and oxidized, denatured HMGB1 has neither. Disulfides 61-70 high mobility group box 1 Homo sapiens 71-76 23226786-0 2012 Atorvastatin reduces serum HMGB1 levels in patients with hyperlipidemia. Atorvastatin 0-12 high mobility group box 1 Homo sapiens 27-32 23022229-0 2012 Tanshinone IIA sodium sulfonate facilitates endocytic HMGB1 uptake. tanshinone II A sodium sulfonate 0-31 high mobility group box 1 Homo sapiens 54-59 23022229-2 2012 We previously reported that a major Danshen ingredient, tanshinone IIA sodium sulfonate (TSN-SS), selectively inhibited endotoxin-induced HMGB1 release and conferred protection against lethal endotoxemia and sepsis. tanshinone II A sodium sulfonate 56-87 high mobility group box 1 Homo sapiens 138-143 23022229-2 2012 We previously reported that a major Danshen ingredient, tanshinone IIA sodium sulfonate (TSN-SS), selectively inhibited endotoxin-induced HMGB1 release and conferred protection against lethal endotoxemia and sepsis. tanshinone II A sodium sulfonate 89-95 high mobility group box 1 Homo sapiens 138-143 23022229-3 2012 To investigate the underlying mechanisms by which TSN-SS effectively inhibits HMGB1 release, we examined whether TSN-SS stimulates HMGB1 uptake by macrophages and whether genetic depletion of HMGB1 receptors [e.g., toll-like receptors (TLR)2, TLR4, or the receptor for advanced glycation end product (RAGE)] or pharmacological inhibition of endocytosis impairs TSN-SS-facilitated HMGB1 cellular uptake. tanshinone II A sodium sulfonate 50-56 high mobility group box 1 Homo sapiens 78-83 23022229-4 2012 TSN-SS stimulated internalization of exogenous HMGB1 protein into macrophage cytoplasmic vesicles that subsequently co-localized with microtubule-associated protein light chain 3 (LC3)-positive punctate structures (likely amphisomes). trichostatin A 0-3 high mobility group box 1 Homo sapiens 47-52 23022229-8 2012 Taken together, these findings suggest that TSN-SS may facilitate HMGB1 endocytic uptake, and subsequently delivered it to LC3-positive vacuoles (possibly amphisomes) for degradation via a lysosome-dependent pathway. tanshinone II A sodium sulfonate 44-50 high mobility group box 1 Homo sapiens 66-71 23226786-2 2012 This study investigated the effect of atorvastatin on serum HMGB1 levels in patients with hyperlipidemia. Atorvastatin 38-50 high mobility group box 1 Homo sapiens 60-65 23226786-9 2012 Serum HMGB1 levels are increased in patients with hyperlipidemia which could be reduced by atorvastatin. Atorvastatin 91-103 high mobility group box 1 Homo sapiens 6-11 22862965-5 2012 The number of CD34-positive cells was negatively correlated with the serum levels of glucose and hemoglobin A1c, whereas the HMGB-1-positive area was positively correlated with the levels of serum glucose. Glucose 197-204 high mobility group box 1 Homo sapiens 125-131 22941653-2 2012 We show that HMGB1 alters the structure and stability of the canonical nucleosome (N) in a nonenzymatic, ATP-independent manner. Adenosine Triphosphate 105-108 high mobility group box 1 Homo sapiens 13-18 23298486-0 2012 Ethyl pyruvate administration suppresses growth and invasion of gallbladder cancer cells via downregulation of HMGB1-RAGE axis. ethyl pyruvate 0-14 high mobility group box 1 Homo sapiens 111-116 23298486-2 2012 Some studies have confirmed that Ethyl pyruvate (EP), a potent inhibitor of HMGB1, exerts the therapeutic effects on metastatic live tumor from gastric cancer. ethyl pyruvate 33-47 high mobility group box 1 Homo sapiens 76-81 23298486-2 2012 Some studies have confirmed that Ethyl pyruvate (EP), a potent inhibitor of HMGB1, exerts the therapeutic effects on metastatic live tumor from gastric cancer. ethyl pyruvate 49-51 high mobility group box 1 Homo sapiens 76-81 23019417-0 2012 Apicidin and docetaxel combination treatment drives CTCFL expression and HMGB1 release acting as potential antitumor immune response inducers in metastatic breast cancer cells. apicidin 0-8 high mobility group box 1 Homo sapiens 73-78 22759395-5 2012 Ethyl pyruvate (EP) treatment blocked H(2)O(2) (oxidative stress)-induced HMGB1 release from human umbilical vein endothelial cells in vitro, and in vivo resulted in nuclear retention and significant blunting of HMGB1 release into the circulation after IRI. ethyl pyruvate 0-14 high mobility group box 1 Homo sapiens 74-79 22759395-5 2012 Ethyl pyruvate (EP) treatment blocked H(2)O(2) (oxidative stress)-induced HMGB1 release from human umbilical vein endothelial cells in vitro, and in vivo resulted in nuclear retention and significant blunting of HMGB1 release into the circulation after IRI. ethyl pyruvate 0-14 high mobility group box 1 Homo sapiens 212-217 22759395-5 2012 Ethyl pyruvate (EP) treatment blocked H(2)O(2) (oxidative stress)-induced HMGB1 release from human umbilical vein endothelial cells in vitro, and in vivo resulted in nuclear retention and significant blunting of HMGB1 release into the circulation after IRI. ethyl pyruvate 16-18 high mobility group box 1 Homo sapiens 74-79 22759395-5 2012 Ethyl pyruvate (EP) treatment blocked H(2)O(2) (oxidative stress)-induced HMGB1 release from human umbilical vein endothelial cells in vitro, and in vivo resulted in nuclear retention and significant blunting of HMGB1 release into the circulation after IRI. ethyl pyruvate 16-18 high mobility group box 1 Homo sapiens 212-217 22759395-5 2012 Ethyl pyruvate (EP) treatment blocked H(2)O(2) (oxidative stress)-induced HMGB1 release from human umbilical vein endothelial cells in vitro, and in vivo resulted in nuclear retention and significant blunting of HMGB1 release into the circulation after IRI. Water 38-43 high mobility group box 1 Homo sapiens 74-79 22759395-5 2012 Ethyl pyruvate (EP) treatment blocked H(2)O(2) (oxidative stress)-induced HMGB1 release from human umbilical vein endothelial cells in vitro, and in vivo resulted in nuclear retention and significant blunting of HMGB1 release into the circulation after IRI. Water 38-43 high mobility group box 1 Homo sapiens 212-217 22782142-0 2012 IGF-1 alleviates ox-LDL-induced inflammation via reducing HMGB1 release in HAECs. ox-ldl 17-23 high mobility group box 1 Homo sapiens 58-63 22782142-13 2012 In conclusion, IGF-1 can alleviate ox-LDL-induced inflammation by reducing HMGB1 release, suggesting an unexpected beneficial role of IGF-1 in inflammatory disease. ox-ldl 35-41 high mobility group box 1 Homo sapiens 75-80 23019417-0 2012 Apicidin and docetaxel combination treatment drives CTCFL expression and HMGB1 release acting as potential antitumor immune response inducers in metastatic breast cancer cells. Docetaxel 13-22 high mobility group box 1 Homo sapiens 73-78 22931605-0 2012 [Hydrogen peroxide induces high mobility group box 1 release in human bronchial epithelial cells]. Hydrogen Peroxide 1-18 high mobility group box 1 Homo sapiens 27-60 22901013-4 2012 The binding affinities of the two individual DNA-binding domains of HMGB4 to DNA carrying a cisplatin 1,2-intrastrand d(GpG) cross-link are weaker than those of the DNA-binding domains of HMGB1. Cisplatin 92-101 high mobility group box 1 Homo sapiens 188-193 22901013-6 2012 The residue Phe37 plays a critical role in stabilizing the binding complex of HMGB4 with the cisplatin-modified DNA, as it does for HMGB1. Cisplatin 93-102 high mobility group box 1 Homo sapiens 132-137 22869893-5 2012 A nonoxidizable HMGB1 mutant in which serines replace all cysteines (3S-HMGB1) does not promote cytokine production, but is more effective than wild-type HMGB1 in recruiting leukocytes in vivo. Cysteine 58-67 high mobility group box 1 Homo sapiens 72-77 22869893-5 2012 A nonoxidizable HMGB1 mutant in which serines replace all cysteines (3S-HMGB1) does not promote cytokine production, but is more effective than wild-type HMGB1 in recruiting leukocytes in vivo. 3s 69-71 high mobility group box 1 Homo sapiens 16-21 22869893-5 2012 A nonoxidizable HMGB1 mutant in which serines replace all cysteines (3S-HMGB1) does not promote cytokine production, but is more effective than wild-type HMGB1 in recruiting leukocytes in vivo. 3s 69-71 high mobility group box 1 Homo sapiens 72-77 22869893-5 2012 A nonoxidizable HMGB1 mutant in which serines replace all cysteines (3S-HMGB1) does not promote cytokine production, but is more effective than wild-type HMGB1 in recruiting leukocytes in vivo. 3s 69-71 high mobility group box 1 Homo sapiens 72-77 22869893-3 2012 Reduced cysteines make HMGB1 a chemoattractant, whereas a disulfide bond makes it a proinflammatory cytokine and further cysteine oxidation to sulfonates by reactive oxygen species abrogates both activities. Cysteine 8-17 high mobility group box 1 Homo sapiens 23-28 22869893-3 2012 Reduced cysteines make HMGB1 a chemoattractant, whereas a disulfide bond makes it a proinflammatory cytokine and further cysteine oxidation to sulfonates by reactive oxygen species abrogates both activities. Cysteine 8-16 high mobility group box 1 Homo sapiens 23-28 22869893-5 2012 A nonoxidizable HMGB1 mutant in which serines replace all cysteines (3S-HMGB1) does not promote cytokine production, but is more effective than wild-type HMGB1 in recruiting leukocytes in vivo. Serine 38-45 high mobility group box 1 Homo sapiens 16-21 22869893-5 2012 A nonoxidizable HMGB1 mutant in which serines replace all cysteines (3S-HMGB1) does not promote cytokine production, but is more effective than wild-type HMGB1 in recruiting leukocytes in vivo. Serine 38-45 high mobility group box 1 Homo sapiens 72-77 22869893-5 2012 A nonoxidizable HMGB1 mutant in which serines replace all cysteines (3S-HMGB1) does not promote cytokine production, but is more effective than wild-type HMGB1 in recruiting leukocytes in vivo. Serine 38-45 high mobility group box 1 Homo sapiens 72-77 22869893-5 2012 A nonoxidizable HMGB1 mutant in which serines replace all cysteines (3S-HMGB1) does not promote cytokine production, but is more effective than wild-type HMGB1 in recruiting leukocytes in vivo. Cysteine 58-67 high mobility group box 1 Homo sapiens 16-21 22869893-5 2012 A nonoxidizable HMGB1 mutant in which serines replace all cysteines (3S-HMGB1) does not promote cytokine production, but is more effective than wild-type HMGB1 in recruiting leukocytes in vivo. Cysteine 58-67 high mobility group box 1 Homo sapiens 72-77 22931605-1 2012 OBJECTIVE: To investigate the effect of hydrogen dioxide (H(2)O(2)) on the release and translocation of high mobility group box 1 release (HMGB1) from normal human bronchiolar epithelial cells (HBE). Hydrogen Peroxide 40-56 high mobility group box 1 Homo sapiens 104-137 22931605-1 2012 OBJECTIVE: To investigate the effect of hydrogen dioxide (H(2)O(2)) on the release and translocation of high mobility group box 1 release (HMGB1) from normal human bronchiolar epithelial cells (HBE). Hydrogen Peroxide 40-56 high mobility group box 1 Homo sapiens 139-144 22931605-9 2012 CONCLUSION: H(2)O(2) can induce HMGB1 translocation and release in human bronchial epithelial cells, suggesting the involvement of HMGB1 in airway oxidative stress in chronic inflammatory diseases such as asthma and COPD. Hydrogen Peroxide 12-20 high mobility group box 1 Homo sapiens 32-37 22931605-9 2012 CONCLUSION: H(2)O(2) can induce HMGB1 translocation and release in human bronchial epithelial cells, suggesting the involvement of HMGB1 in airway oxidative stress in chronic inflammatory diseases such as asthma and COPD. Hydrogen Peroxide 12-20 high mobility group box 1 Homo sapiens 131-136 22549180-6 2012 Up-regulation of Mfn2 attenuated the suppressive effect of HMGB1 on CD4(+) T lymphocytes, which was associated with profound elevation of intracellular calcium concentration ([Ca(2+)](i)) and nuclear factor of activated T cells (NFAT) activity. Calcium 152-159 high mobility group box 1 Homo sapiens 59-64 22750245-7 2012 The serine residues at S39, S53 and S181 of HMGB1 were related to enhancing HMGB1 secretion. Serine 4-10 high mobility group box 1 Homo sapiens 44-49 22750245-7 2012 The serine residues at S39, S53 and S181 of HMGB1 were related to enhancing HMGB1 secretion. Serine 4-10 high mobility group box 1 Homo sapiens 76-81 22750245-9 2012 Our findings suggest that PKC-zeta is involved in the phosphorylation of HMGB1, and the phosphorylation of specific serine residues in the nuclear localization signal regions is related to enhanced HMGB1 secretion in colon cancer cells. Serine 116-122 high mobility group box 1 Homo sapiens 198-203 22634181-4 2012 In wild-type mice, recombinant human HMGB1 aggravated indomethacin-induced small intestinal damage; enhanced the mRNA expression levels of tumor necrosis factor alpha (TNF-alpha), monocyte chemotactic protein 1, and KC; activated nuclear factor kappa B; and stimulated phosphorylation of the mitogen-activated protein kinases p38, extracellular signal-regulated kinase (ERK), and c-Jun N-terminal kinase (JNK). Indomethacin 54-66 high mobility group box 1 Homo sapiens 37-42 22617774-0 2012 HMGB1 was a pivotal synergistic effecor for CpG oligonucleotide to enhance the progression of human lung cancer cells. effecor 32-39 high mobility group box 1 Homo sapiens 0-5 22617774-0 2012 HMGB1 was a pivotal synergistic effecor for CpG oligonucleotide to enhance the progression of human lung cancer cells. Oligonucleotides 48-63 high mobility group box 1 Homo sapiens 0-5 22617774-5 2012 We further showed that blockade of extracellular HMGB1 using A box peptide and ethyl pyruvate significantly abrogated the CpG ODN enhanced progression of 95D cells. ethyl pyruvate 79-93 high mobility group box 1 Homo sapiens 49-54 22504532-0 2012 Vascular barrier protective effects of phlorotannins on HMGB1-mediated proinflammatory responses in vitro and in vivo. phlorotannins 39-52 high mobility group box 1 Homo sapiens 56-61 22655796-5 2012 Enforced expression of HMGB1-3 or HMGB4 by adenoviral-vector-mediated gene transfer resulted in significant inhibition of breast cancer cell proliferation through an LXCXE- or LXCXD-dependent mechanism and an increased radiosensitivity through an LXCXE- or LXCXD-independent mechanism. lxcxe 166-171 high mobility group box 1 Homo sapiens 23-28 22655796-5 2012 Enforced expression of HMGB1-3 or HMGB4 by adenoviral-vector-mediated gene transfer resulted in significant inhibition of breast cancer cell proliferation through an LXCXE- or LXCXD-dependent mechanism and an increased radiosensitivity through an LXCXE- or LXCXD-independent mechanism. lxcxd 176-181 high mobility group box 1 Homo sapiens 23-28 22655796-5 2012 Enforced expression of HMGB1-3 or HMGB4 by adenoviral-vector-mediated gene transfer resulted in significant inhibition of breast cancer cell proliferation through an LXCXE- or LXCXD-dependent mechanism and an increased radiosensitivity through an LXCXE- or LXCXD-independent mechanism. lxcxe 247-252 high mobility group box 1 Homo sapiens 23-28 22655796-5 2012 Enforced expression of HMGB1-3 or HMGB4 by adenoviral-vector-mediated gene transfer resulted in significant inhibition of breast cancer cell proliferation through an LXCXE- or LXCXD-dependent mechanism and an increased radiosensitivity through an LXCXE- or LXCXD-independent mechanism. lxcxd 257-262 high mobility group box 1 Homo sapiens 23-28 22429818-0 2012 Inhibitory effects of lycopene on HMGB1-mediated pro-inflammatory responses in both cellular and animal models. Lycopene 22-30 high mobility group box 1 Homo sapiens 34-39 22429818-3 2012 The potential anti-inflammatory roles of lycopene in HMGB1-mediated proinflammatory responses in both primary human umbilical vein endothelial cells (HUVECs) and animal were investigated. Lycopene 41-49 high mobility group box 1 Homo sapiens 53-58 22429818-6 2012 Lycopene inhibited lipopolysaccharide (LPS)-mediated release of HMGB1, expression of HMGB1-mediated tumor necrosis factor (TNF)-secretory phospholipase A2 (sPLA2)-IIA, and HMGB1-mediated pro-inflammatory signaling responses in endothelial cells. Lycopene 0-8 high mobility group box 1 Homo sapiens 64-69 22429818-6 2012 Lycopene inhibited lipopolysaccharide (LPS)-mediated release of HMGB1, expression of HMGB1-mediated tumor necrosis factor (TNF)-secretory phospholipase A2 (sPLA2)-IIA, and HMGB1-mediated pro-inflammatory signaling responses in endothelial cells. Lycopene 0-8 high mobility group box 1 Homo sapiens 85-90 22429818-6 2012 Lycopene inhibited lipopolysaccharide (LPS)-mediated release of HMGB1, expression of HMGB1-mediated tumor necrosis factor (TNF)-secretory phospholipase A2 (sPLA2)-IIA, and HMGB1-mediated pro-inflammatory signaling responses in endothelial cells. Lycopene 0-8 high mobility group box 1 Homo sapiens 85-90 22429818-8 2012 These findings suggest that lycopene promotes barrier integrity, inhibits monocyte adhesion and migration to HMGB1 activating HUVECs by blocking activation of proinflammatory cytokines and expression of CAMs and HMGB1 receptors, thereby showing its usefulness as a therapy for vascular inflammatory diseases. Lycopene 28-36 high mobility group box 1 Homo sapiens 109-114 22561332-0 2012 Barrier protective effects of withaferin A in HMGB1-induced inflammatory responses in both cellular and animal models. withaferin A 30-42 high mobility group box 1 Homo sapiens 46-51 22561332-4 2012 We found that WFA inhibited lipopolysaccharide (LPS)-induced HMGB1 release and HMGB1-mediated barrier disruption, expression of cell adhesion molecules (CAMs) and adhesion/transendothelial migration of leukocytes to human endothelial cells. withaferin A 14-17 high mobility group box 1 Homo sapiens 61-66 22561332-4 2012 We found that WFA inhibited lipopolysaccharide (LPS)-induced HMGB1 release and HMGB1-mediated barrier disruption, expression of cell adhesion molecules (CAMs) and adhesion/transendothelial migration of leukocytes to human endothelial cells. withaferin A 14-17 high mobility group box 1 Homo sapiens 79-84 22561332-6 2012 Further studies revealed that WFA suppressed the production of interleukin 6, tumor necrosis factor-alpha (TNF-alpha) and activation of nuclear factor-kappaB (NF-kappaB) by HMGB1. withaferin A 30-33 high mobility group box 1 Homo sapiens 173-178 22467309-8 2012 In activated HAEC, OSS amplified HMGB1-induced VCAM-1 (3-fold) and RAGE (1.6-fold) expression and proportionally enhanced monocyte adhesion to HAEC in a RAGE-dependent manner, while HSS mitigated these increases to the level of TNF-alpha alone. OSS 19-22 high mobility group box 1 Homo sapiens 33-38 22467309-8 2012 In activated HAEC, OSS amplified HMGB1-induced VCAM-1 (3-fold) and RAGE (1.6-fold) expression and proportionally enhanced monocyte adhesion to HAEC in a RAGE-dependent manner, while HSS mitigated these increases to the level of TNF-alpha alone. hepatic stimulator substance 182-185 high mobility group box 1 Homo sapiens 33-38 22429818-8 2012 These findings suggest that lycopene promotes barrier integrity, inhibits monocyte adhesion and migration to HMGB1 activating HUVECs by blocking activation of proinflammatory cytokines and expression of CAMs and HMGB1 receptors, thereby showing its usefulness as a therapy for vascular inflammatory diseases. Lycopene 28-36 high mobility group box 1 Homo sapiens 212-217 22504532-3 2012 The protective activities of the phlorotannins were determined by measuring permeability, leukocyte adhesion and migration, and the activations of pro-inflammatory proteins in HMGB1-activated HUVECs. phlorotannins 33-46 high mobility group box 1 Homo sapiens 176-181 22504532-4 2012 We found that the phlorotannins inhibited; lipopolysaccharide (LPS)-induced HMGB1 release, HMGB1-mediated barrier disruption, the expressions of cell adhesion molecules (CAMs), and the adhesion/transendothelial migration of leukocytes to human endothelial cells. phlorotannins 18-31 high mobility group box 1 Homo sapiens 76-81 22504532-4 2012 We found that the phlorotannins inhibited; lipopolysaccharide (LPS)-induced HMGB1 release, HMGB1-mediated barrier disruption, the expressions of cell adhesion molecules (CAMs), and the adhesion/transendothelial migration of leukocytes to human endothelial cells. phlorotannins 18-31 high mobility group box 1 Homo sapiens 91-96 22475723-8 2012 Further bioinformatic analysis shows that proteins modulated by curcumin are implicated in protein folding (such as heat-shock protein PPP2R1A; RNA splicing proteins RBM17, DDX39; cell death proteins HMGB1 and NPM1; proteins involved in androgen receptor signaling, NPM1 and FKBP4/FKBP52), and that this compound could have an impact on miR-141, miR-152, and miR-183 expression. Curcumin 64-72 high mobility group box 1 Homo sapiens 200-205 21932058-2 2012 The study was tested in a fecal peritonitis-induced septic shock model, we observed that levosimendan combined with arginine vasopressin supplemented with norepinephrine therapy resulted in lower mean pulmonary artery pressure, lactate concentrations, arterial total nitrate/nitrite, and high-mobility group box 1 levels; decreased lung wet/dry ratio, and pulmonary levels of interleukin-6, total histological scores, and improved pulmonary gas exchange when compared with norepinephrine group. Simendan 89-101 high mobility group box 1 Homo sapiens 288-313 21932058-2 2012 The study was tested in a fecal peritonitis-induced septic shock model, we observed that levosimendan combined with arginine vasopressin supplemented with norepinephrine therapy resulted in lower mean pulmonary artery pressure, lactate concentrations, arterial total nitrate/nitrite, and high-mobility group box 1 levels; decreased lung wet/dry ratio, and pulmonary levels of interleukin-6, total histological scores, and improved pulmonary gas exchange when compared with norepinephrine group. Norepinephrine 155-169 high mobility group box 1 Homo sapiens 288-313 22394598-4 2012 In vitro and in vivo, HMGB-1 inhibition achieved by glycyrrhizin treatment led to a drastic reduction in ALK(+) cell invasiveness. Glycyrrhizic Acid 52-64 high mobility group box 1 Homo sapiens 22-28 22521432-5 2012 Furthermore, HMGB1 increased the production of intracellular reactive oxygen species (ROS). Reactive Oxygen Species 61-84 high mobility group box 1 Homo sapiens 13-18 22521432-5 2012 Furthermore, HMGB1 increased the production of intracellular reactive oxygen species (ROS). Reactive Oxygen Species 86-89 high mobility group box 1 Homo sapiens 13-18 22521432-7 2012 Taken together, our results suggest that HMGB1 induces MUC8 expression in a JNK and PI3K/Akt signaling pathway-dependent manner but that HMGB1 acts in an ROS-independent manner. Reactive Oxygen Species 154-157 high mobility group box 1 Homo sapiens 137-142 22593488-6 2012 HMGB1 levels were found to be considerably lower in EDTA plasma as compared to serum samples. Edetic Acid 52-56 high mobility group box 1 Homo sapiens 0-5 22204001-0 2012 Poly(ADP-ribosyl)ation of high mobility group box 1 (HMGB1) protein enhances inhibition of efferocytosis. poly(adp-ribosyl) 0-17 high mobility group box 1 Homo sapiens 26-51 22204001-0 2012 Poly(ADP-ribosyl)ation of high mobility group box 1 (HMGB1) protein enhances inhibition of efferocytosis. poly(adp-ribosyl) 0-17 high mobility group box 1 Homo sapiens 53-58 22386814-0 2012 Anti-inflammatory activities of oleanolic acid on HMGB1 activated HUVECs. Oleanolic Acid 32-46 high mobility group box 1 Homo sapiens 50-55 22386814-3 2012 Oleanolic acid (OA), a triterpenoid known for its anti-inflammatory and anti-cancer properties, is commonly present in several medicinal plants but the effects of OA on HMGB1-mediated pro-inflammatory responses of human endothelial cells is not well-studied. Oleanolic Acid 0-14 high mobility group box 1 Homo sapiens 169-174 22266604-0 2012 Molecular forms of HMGB1 and keratin-18 as mechanistic biomarkers for mode of cell death and prognosis during clinical acetaminophen hepatotoxicity. Acetaminophen 119-132 high mobility group box 1 Homo sapiens 19-24 22266604-4 2012 METHODS: HMGB1 (total, acetylated) and K18 (apoptotic, necrotic) were identified and quantified by novel LC-MS/MS assays in APAP overdose patients (n=78). Acetaminophen 124-128 high mobility group box 1 Homo sapiens 9-14 22366983-10 2012 The present study showed that patients who received sivelestat had reduced release of HMGB1, and that IL-6 levels decreased more rapidly in patients treated with sivelestat than in those who received the placebo. sivelestat 52-62 high mobility group box 1 Homo sapiens 86-91 22246223-0 2012 Inhibitory effects of ethyl pyruvate administration on human gastric cancer growth via regulation of the HMGB1-RAGE and Akt pathways in vitro and in vivo. ethyl pyruvate 22-36 high mobility group box 1 Homo sapiens 117-122 22246223-2 2012 Ethyl pyruvate (EP), a potent inhibitor of HMGB1 release, can exert antitumor effects on the growth of gastric cancer. ethyl pyruvate 0-14 high mobility group box 1 Homo sapiens 55-60 22246223-2 2012 Ethyl pyruvate (EP), a potent inhibitor of HMGB1 release, can exert antitumor effects on the growth of gastric cancer. ethyl pyruvate 16-18 high mobility group box 1 Homo sapiens 55-60 22246807-0 2012 Heat shock transcription factor 1 inhibits H2O2-induced cardiomyocyte death through suppression of high-mobility group box 1. Hydrogen Peroxide 43-47 high mobility group box 1 Homo sapiens 99-124 22248244-8 2012 CONCLUSION: Serum HMGB1 levels are increased in pSS patients and more specifically in patients with SSA autoantibodies. pss 48-51 high mobility group box 1 Homo sapiens 18-23 22178603-0 2012 Inhibitory effects of kaempferol-3-O-sophoroside on HMGB1-mediated proinflammatory responses. kaempferol 3-O-sophoroside 22-48 high mobility group box 1 Homo sapiens 52-57 22189943-2 2012 UA, which is significantly increased in the circulation following renal ischemia-reperfusion injury (IRI), was used both in vitro and in vivo as an early response-signaling molecule to determine its ability to induce the secretion of HMGB1 from endothelial cells. Uric Acid 0-2 high mobility group box 1 Homo sapiens 234-239 22189943-4 2012 Treatment of HUVEC with UA and a calcium mobilization inhibitor (TMB-8) or a MEK/Erk pathway inhibitor (U0126) prevented translocation of HMGB1 from the nucleus, resulting in reduced cytoplasmic and circulating levels of HMGB1. Uric Acid 24-26 high mobility group box 1 Homo sapiens 138-143 22189943-4 2012 Treatment of HUVEC with UA and a calcium mobilization inhibitor (TMB-8) or a MEK/Erk pathway inhibitor (U0126) prevented translocation of HMGB1 from the nucleus, resulting in reduced cytoplasmic and circulating levels of HMGB1. Uric Acid 24-26 high mobility group box 1 Homo sapiens 221-226 22189943-4 2012 Treatment of HUVEC with UA and a calcium mobilization inhibitor (TMB-8) or a MEK/Erk pathway inhibitor (U0126) prevented translocation of HMGB1 from the nucleus, resulting in reduced cytoplasmic and circulating levels of HMGB1. Calcium 33-40 high mobility group box 1 Homo sapiens 138-143 22189943-4 2012 Treatment of HUVEC with UA and a calcium mobilization inhibitor (TMB-8) or a MEK/Erk pathway inhibitor (U0126) prevented translocation of HMGB1 from the nucleus, resulting in reduced cytoplasmic and circulating levels of HMGB1. 8-(N,N-diethylamino)octyl-3,4,5-trimethoxybenzoate 65-70 high mobility group box 1 Homo sapiens 138-143 22189943-4 2012 Treatment of HUVEC with UA and a calcium mobilization inhibitor (TMB-8) or a MEK/Erk pathway inhibitor (U0126) prevented translocation of HMGB1 from the nucleus, resulting in reduced cytoplasmic and circulating levels of HMGB1. 8-(N,N-diethylamino)octyl-3,4,5-trimethoxybenzoate 65-70 high mobility group box 1 Homo sapiens 221-226 22189943-4 2012 Treatment of HUVEC with UA and a calcium mobilization inhibitor (TMB-8) or a MEK/Erk pathway inhibitor (U0126) prevented translocation of HMGB1 from the nucleus, resulting in reduced cytoplasmic and circulating levels of HMGB1. U 0126 104-109 high mobility group box 1 Homo sapiens 138-143 22189943-4 2012 Treatment of HUVEC with UA and a calcium mobilization inhibitor (TMB-8) or a MEK/Erk pathway inhibitor (U0126) prevented translocation of HMGB1 from the nucleus, resulting in reduced cytoplasmic and circulating levels of HMGB1. U 0126 104-109 high mobility group box 1 Homo sapiens 221-226 22189943-7 2012 In summary, UA after IRI mediates the acetylation and release of HMGB1 from endothelial cells by mechanisms that involve calcium mobilization, the MEK/Erk pathway, and activation of TLR4. Calcium 121-128 high mobility group box 1 Homo sapiens 65-70 21743489-0 2012 NAC1 modulates sensitivity of ovarian cancer cells to cisplatin by altering the HMGB1-mediated autophagic response. Cisplatin 54-63 high mobility group box 1 Homo sapiens 80-85 22047946-5 2012 Peripheral whole blood samples obtained immediately after admission were determined for HMGB1 production in response to ex vivo lipopolysaccharide (LPS) stimulation. ex vivo lipopolysaccharide 120-146 high mobility group box 1 Homo sapiens 88-93 22065079-8 2012 Asbestos induces cell necrosis, causing the release of HMGB1, which in turn may activate Nalp3 inflammasome, a process that is enhanced by asbestos-induced production of reactive oxygen species. Asbestos 0-8 high mobility group box 1 Homo sapiens 55-60 22363140-0 2012 Glycyrrhizin attenuates HMGB1-induced hepatocyte apoptosis by inhibiting the p38-dependent mitochondrial pathway. Glycyrrhizic Acid 0-12 high mobility group box 1 Homo sapiens 24-29 22363140-5 2012 It is also tried to identify hepatocyte apoptosis affected by the HMGB1 inhibitor, GL. Glycyrrhizic Acid 83-85 high mobility group box 1 Homo sapiens 66-71 22363140-9 2012 HMGB1-induced caspase 3 activation was significantly attenuated by the p38 inhibitor SB203580. SB 203580 85-93 high mobility group box 1 Homo sapiens 0-5 22363140-10 2012 GL significantly attenuated HMGB1-induced hepatocyte apoptosis. Glycyrrhizic Acid 0-2 high mobility group box 1 Homo sapiens 28-33 22363140-11 2012 GL also prevented HMGB1-induced cytochrome c release and p38 activation in Huh-BAT cells. Glycyrrhizic Acid 0-2 high mobility group box 1 Homo sapiens 18-23 22363140-13 2012 In addition, GL had an anti-apoptotic effect on HMGB1-treated hepatocytes. Glycyrrhizic Acid 13-15 high mobility group box 1 Homo sapiens 48-53 22065079-8 2012 Asbestos induces cell necrosis, causing the release of HMGB1, which in turn may activate Nalp3 inflammasome, a process that is enhanced by asbestos-induced production of reactive oxygen species. Asbestos 139-147 high mobility group box 1 Homo sapiens 55-60 22065079-8 2012 Asbestos induces cell necrosis, causing the release of HMGB1, which in turn may activate Nalp3 inflammasome, a process that is enhanced by asbestos-induced production of reactive oxygen species. Reactive Oxygen Species 170-193 high mobility group box 1 Homo sapiens 55-60 22293756-3 2012 The activity of HMGB1 varies with the redox states of the cysteine residues, which are required for binding to TLR4. Cysteine 58-66 high mobility group box 1 Homo sapiens 16-21 21864037-5 2012 While the use of autophagy inhibitor, 3-methyladenine (3-MA), blocked the autophagic reaction and increased leukemia cell sensitivity to chemotherapy, enhancing HMGB1 expression decreased this sensitivity. 3-methyladenine 38-53 high mobility group box 1 Homo sapiens 161-166 21494799-4 2012 We found that HMGB1 increased human umbilical vein endothelial cell permeability to FITC-dextran 40 kDa in a time- and concentration-dependent manner. fluorescein isothiocyanate dextran 84-96 high mobility group box 1 Homo sapiens 14-19 22085682-0 2012 Tricin 7-glucoside protects against experimental cerebral ischemia by reduction of NF-kappaB and HMGB1 expression. Tricin 7-glucoside 0-18 high mobility group box 1 Homo sapiens 97-102 22277195-9 2012 Treatment with DT attenuated the expression of TLR2, TLR4, IRAK1 and Hmgb1, all genes involved in the TLR-mediated signaling pathway associated with classical microglia activation. 3,6'-dithiothalidomide 15-17 high mobility group box 1 Homo sapiens 69-74 22085682-5 2012 For in vivo experiment, rats were subjected to middle cerebral artery occlusion (MACO) for 1h, then followed by reperfusion for 23 h. Treatment of SH-SY5Y cells with Tricin 7-glucoside reduced the OGD-induced apoptosis and cytotoxicity, blocked TNF-alpha-induced NF-kappaB and IkappaB-alpha phosphorylation, and decreased HMGB1 expression. Tricin 7-glucoside 166-184 high mobility group box 1 Homo sapiens 322-327 22085682-8 2012 Tricin 7-glucoside 100mg/kg attenuated histopathological damage, decreased brain edema, inhibited NF-kappaB activation and reduced HMGB1 expression. Tricin 7-glucoside 0-18 high mobility group box 1 Homo sapiens 131-136 22085682-9 2012 These data show that Tricin 7-glucoside protects brain against I/R injury with a favorable therapeutic time-window by alleviating cerebral I/R injury and attenuating blood-brain barrier (BBB) breakdown, and its protective effects may involve HMGB1 and NF-kappaB signaling pathway. Tricin 7-glucoside 21-39 high mobility group box 1 Homo sapiens 242-247 21861984-8 2012 IL-1beta promotes HMGB1 production in human gingival epithelial cells in a nitric oxide-dependent manner. Nitric Oxide 75-87 high mobility group box 1 Homo sapiens 18-23 22068710-6 2012 The effect of sodium reduction in: (i) Normotensives: Caucasians: systolic BP (SBP) -1.27 mm Hg (95% confidence interval (CI): -1.88, -0.66; P = 0.0001), diastolic BP (DBP) -0.05 mm Hg (95% CI: -0.51, 0.42; P = 0.85). Sodium 14-20 high mobility group box 1 Homo sapiens 66-86 21760473-9 2012 However, the decreases in UAE, serum HMGB1 and serum IL-6 were significantly greater in the telmisartan group than in the enalapril group at 6 months (P < 0.05, P < 0.01 and P < 0.01, respectively) and 12 months (all, P < 0.05). Telmisartan 92-103 high mobility group box 1 Homo sapiens 37-42 22108005-0 2012 Autophagy-mediated HMGB1 release antagonizes apoptosis of gastric cancer cells induced by vincristine via transcriptional regulation of Mcl-1. Vincristine 90-101 high mobility group box 1 Homo sapiens 19-24 22799333-4 2012 HMGB1 was also found significantly enhanced the activity of ATP to induce IL-1beta and IL-18 by the induction of increased expression of pro-IL-1beta and pro-IL-18. Adenosine Triphosphate 60-63 high mobility group box 1 Homo sapiens 0-5 22108005-3 2012 We show in this study that HMGB1 released into the extracellular space protects gastric cancer cells from apoptosis induced by the microtubule-targeting drug vincristine through transcriptional upregulation of Mcl-1. Vincristine 158-169 high mobility group box 1 Homo sapiens 27-32 22108005-9 2012 Taken together, these results identify HMGB1-mediated upregulation of Mcl-1 transcription as an important mechanism by which autophagy protects gastric cancer cells from apoptosis induced by vincristine. Vincristine 191-202 high mobility group box 1 Homo sapiens 39-44 22038506-3 2012 METHODS: We synthesized three specific small interfering RNAs of HMGB1 (HMGB1-siRNAs) and transfected these into HCCLM3 cells by use of Lipofectamine 2000. Lipofectamine 136-154 high mobility group box 1 Homo sapiens 65-70 23275711-0 2012 Structural insight into the ligand-receptor interaction between glycyrrhetinic acid (GA) and the high-mobility group protein B1 (HMGB1)-DNA complex. Glycyrrhetinic Acid 64-83 high mobility group box 1 Homo sapiens 97-127 23275711-0 2012 Structural insight into the ligand-receptor interaction between glycyrrhetinic acid (GA) and the high-mobility group protein B1 (HMGB1)-DNA complex. Glycyrrhetinic Acid 64-83 high mobility group box 1 Homo sapiens 129-134 23275711-0 2012 Structural insight into the ligand-receptor interaction between glycyrrhetinic acid (GA) and the high-mobility group protein B1 (HMGB1)-DNA complex. Glycyrrhetinic Acid 85-87 high mobility group box 1 Homo sapiens 97-127 23275711-0 2012 Structural insight into the ligand-receptor interaction between glycyrrhetinic acid (GA) and the high-mobility group protein B1 (HMGB1)-DNA complex. Glycyrrhetinic Acid 85-87 high mobility group box 1 Homo sapiens 129-134 23275711-1 2012 UNLABELLED: Structural analysis of the high-mobility group protein B1 (HMGB1)-DNA complex and a docking simulation between glycyrrhetinic acid (GA) and the HMGB1-DNA complex were performed with a software package the Molecular Operating Environment (MOE). Glycyrrhetinic Acid 123-142 high mobility group box 1 Homo sapiens 156-161 23275711-1 2012 UNLABELLED: Structural analysis of the high-mobility group protein B1 (HMGB1)-DNA complex and a docking simulation between glycyrrhetinic acid (GA) and the HMGB1-DNA complex were performed with a software package the Molecular Operating Environment (MOE). Glycyrrhetinic Acid 144-146 high mobility group box 1 Homo sapiens 156-161 23275711-4 2012 The docking simulation revealed that GA possibly inhibits functions of HMGB1 interfering with Lys(90), Arg(91), Ser(101), Tyr(149), C(230) and C(231) in the HMGB1-DNA complex. Glycyrrhetinic Acid 37-39 high mobility group box 1 Homo sapiens 71-76 23275711-4 2012 The docking simulation revealed that GA possibly inhibits functions of HMGB1 interfering with Lys(90), Arg(91), Ser(101), Tyr(149), C(230) and C(231) in the HMGB1-DNA complex. Glycyrrhetinic Acid 37-39 high mobility group box 1 Homo sapiens 157-162 23275711-4 2012 The docking simulation revealed that GA possibly inhibits functions of HMGB1 interfering with Lys(90), Arg(91), Ser(101), Tyr(149), C(230) and C(231) in the HMGB1-DNA complex. Lysine 94-97 high mobility group box 1 Homo sapiens 71-76 23275711-4 2012 The docking simulation revealed that GA possibly inhibits functions of HMGB1 interfering with Lys(90), Arg(91), Ser(101), Tyr(149), C(230) and C(231) in the HMGB1-DNA complex. Lysine 94-97 high mobility group box 1 Homo sapiens 157-162 23275711-4 2012 The docking simulation revealed that GA possibly inhibits functions of HMGB1 interfering with Lys(90), Arg(91), Ser(101), Tyr(149), C(230) and C(231) in the HMGB1-DNA complex. Arginine 103-106 high mobility group box 1 Homo sapiens 71-76 23275711-4 2012 The docking simulation revealed that GA possibly inhibits functions of HMGB1 interfering with Lys(90), Arg(91), Ser(101), Tyr(149), C(230) and C(231) in the HMGB1-DNA complex. Arginine 103-106 high mobility group box 1 Homo sapiens 157-162 23275711-4 2012 The docking simulation revealed that GA possibly inhibits functions of HMGB1 interfering with Lys(90), Arg(91), Ser(101), Tyr(149), C(230) and C(231) in the HMGB1-DNA complex. Serine 112-115 high mobility group box 1 Homo sapiens 71-76 23275711-4 2012 The docking simulation revealed that GA possibly inhibits functions of HMGB1 interfering with Lys(90), Arg(91), Ser(101), Tyr(149), C(230) and C(231) in the HMGB1-DNA complex. Serine 112-115 high mobility group box 1 Homo sapiens 157-162 23275711-4 2012 The docking simulation revealed that GA possibly inhibits functions of HMGB1 interfering with Lys(90), Arg(91), Ser(101), Tyr(149), C(230) and C(231) in the HMGB1-DNA complex. Tyrosine 122-125 high mobility group box 1 Homo sapiens 71-76 23275711-4 2012 The docking simulation revealed that GA possibly inhibits functions of HMGB1 interfering with Lys(90), Arg(91), Ser(101), Tyr(149), C(230) and C(231) in the HMGB1-DNA complex. Tyrosine 122-125 high mobility group box 1 Homo sapiens 157-162 22102692-4 2012 Anticancer agents including doxorubicin, cisplatin, and methotrexate each induced HMGB1 upregulation in human osteosarcoma cells, and RNA interference-mediated knockdown of HMGB1 restored the chemosensitivity of osteosarcoma cells in vivo and in vitro. Doxorubicin 28-39 high mobility group box 1 Homo sapiens 82-87 22102692-4 2012 Anticancer agents including doxorubicin, cisplatin, and methotrexate each induced HMGB1 upregulation in human osteosarcoma cells, and RNA interference-mediated knockdown of HMGB1 restored the chemosensitivity of osteosarcoma cells in vivo and in vitro. Doxorubicin 28-39 high mobility group box 1 Homo sapiens 173-178 22102692-4 2012 Anticancer agents including doxorubicin, cisplatin, and methotrexate each induced HMGB1 upregulation in human osteosarcoma cells, and RNA interference-mediated knockdown of HMGB1 restored the chemosensitivity of osteosarcoma cells in vivo and in vitro. Cisplatin 41-50 high mobility group box 1 Homo sapiens 82-87 22102692-4 2012 Anticancer agents including doxorubicin, cisplatin, and methotrexate each induced HMGB1 upregulation in human osteosarcoma cells, and RNA interference-mediated knockdown of HMGB1 restored the chemosensitivity of osteosarcoma cells in vivo and in vitro. Cisplatin 41-50 high mobility group box 1 Homo sapiens 173-178 22102692-4 2012 Anticancer agents including doxorubicin, cisplatin, and methotrexate each induced HMGB1 upregulation in human osteosarcoma cells, and RNA interference-mediated knockdown of HMGB1 restored the chemosensitivity of osteosarcoma cells in vivo and in vitro. Methotrexate 56-68 high mobility group box 1 Homo sapiens 82-87 22102692-4 2012 Anticancer agents including doxorubicin, cisplatin, and methotrexate each induced HMGB1 upregulation in human osteosarcoma cells, and RNA interference-mediated knockdown of HMGB1 restored the chemosensitivity of osteosarcoma cells in vivo and in vitro. Methotrexate 56-68 high mobility group box 1 Homo sapiens 173-178 21967802-7 2012 Inhibition of HMGB1 with glycyrrhizin increased the pro-inflammatory response, increasing cell death and up regulating chemokine and TLR4 mRNA expression. Glycyrrhizic Acid 25-37 high mobility group box 1 Homo sapiens 14-19 21953494-9 2012 Furthermore, HMGB1 silencing sensitized cells to apoptosis that was induced by oxaliplatin and mediated by the caspase-3 pathway. Oxaliplatin 79-90 high mobility group box 1 Homo sapiens 13-18 22038506-9 2012 MTT assay demonstrated that the growth of cells transfected with HMGB1-siRNA-1 was significantly lower than that of control cells (P < 0.01). monooxyethylene trimethylolpropane tristearate 0-3 high mobility group box 1 Homo sapiens 65-70 22038506-11 2012 FCM revealed that apoptosis was significantly increased in HMGB1-siRNA-1-transfected cells compared with control cells (P < 0.01). Fosfomycin 0-3 high mobility group box 1 Homo sapiens 59-64 21944908-13 2012 Inhibition of autophagy with 3MA or chloroquine abrogated tube formation, whereas its induction with rapamycin enhanced tubing and promoted HMGB1 translocation. Sirolimus 101-110 high mobility group box 1 Homo sapiens 140-145 22082188-2 2012 In this study, we stimulated primary keratinocytes with the TLR3-ligand polyI:C. This induced a toxic effect shown by up-regulation of the alarmin high-mobility group protein B1 and reduced responses in a MTT-assay. Poly I 72-77 high mobility group box 1 Homo sapiens 147-177 21947243-6 2012 (2)HMGB1 incubation induced ERK1/2 activation in a time- and dose-dependent manner, which could be completely blocked by U0126 (MEK1/2 inhibitor) and partially reversed by RAGE knockdown. U 0126 121-126 high mobility group box 1 Homo sapiens 3-8 22514737-4 2012 We hypothesized that oxidation of HMGB1 may reduce its pro-inflammatory potential and could take place during prolonged ischemia and upon reperfusion.Liver grafts were cold preserved for 24 h and flushed with saline in hourly intervals to collect the effluent. Sodium Chloride 209-215 high mobility group box 1 Homo sapiens 34-39 25215057-7 2012 Anti-HMGB1 treatment, such as anti-HMGB1 polyclonal or monoclonal antibodies, inhibitors (e.g., ethyl pyruvate) and antagonists (e.g., A box), can protect against sepsis lethality and give a wider window for the treatment opportunity. ethyl pyruvate 96-110 high mobility group box 1 Homo sapiens 5-10 22607100-7 2012 After 10-h treatment with CPFA, the MAP and oxygen index (PaO(2)/FiO(2)) were significantly higher than those at 0 h. Furthermore, the serum levels of HMGB-1, ICAM-1, and TNF-alpha decreased after 10 h of CPFA (p < 0.05), while the serum levels of HMGB-1 declined at 5 h of HVHF, but rebounded at 10 h, and the serum levels of TNF-alpha and ICAM-1 were no significant change after treatment with HVHF. cpfa 26-30 high mobility group box 1 Homo sapiens 251-257 22607100-7 2012 After 10-h treatment with CPFA, the MAP and oxygen index (PaO(2)/FiO(2)) were significantly higher than those at 0 h. Furthermore, the serum levels of HMGB-1, ICAM-1, and TNF-alpha decreased after 10 h of CPFA (p < 0.05), while the serum levels of HMGB-1 declined at 5 h of HVHF, but rebounded at 10 h, and the serum levels of TNF-alpha and ICAM-1 were no significant change after treatment with HVHF. cpfa 205-209 high mobility group box 1 Homo sapiens 151-157 22607100-7 2012 After 10-h treatment with CPFA, the MAP and oxygen index (PaO(2)/FiO(2)) were significantly higher than those at 0 h. Furthermore, the serum levels of HMGB-1, ICAM-1, and TNF-alpha decreased after 10 h of CPFA (p < 0.05), while the serum levels of HMGB-1 declined at 5 h of HVHF, but rebounded at 10 h, and the serum levels of TNF-alpha and ICAM-1 were no significant change after treatment with HVHF. hvhf 277-281 high mobility group box 1 Homo sapiens 151-157 22607100-7 2012 After 10-h treatment with CPFA, the MAP and oxygen index (PaO(2)/FiO(2)) were significantly higher than those at 0 h. Furthermore, the serum levels of HMGB-1, ICAM-1, and TNF-alpha decreased after 10 h of CPFA (p < 0.05), while the serum levels of HMGB-1 declined at 5 h of HVHF, but rebounded at 10 h, and the serum levels of TNF-alpha and ICAM-1 were no significant change after treatment with HVHF. hvhf 399-403 high mobility group box 1 Homo sapiens 151-157 22607100-7 2012 After 10-h treatment with CPFA, the MAP and oxygen index (PaO(2)/FiO(2)) were significantly higher than those at 0 h. Furthermore, the serum levels of HMGB-1, ICAM-1, and TNF-alpha decreased after 10 h of CPFA (p < 0.05), while the serum levels of HMGB-1 declined at 5 h of HVHF, but rebounded at 10 h, and the serum levels of TNF-alpha and ICAM-1 were no significant change after treatment with HVHF. cpfa 26-30 high mobility group box 1 Homo sapiens 151-157 25215057-7 2012 Anti-HMGB1 treatment, such as anti-HMGB1 polyclonal or monoclonal antibodies, inhibitors (e.g., ethyl pyruvate) and antagonists (e.g., A box), can protect against sepsis lethality and give a wider window for the treatment opportunity. ethyl pyruvate 96-110 high mobility group box 1 Homo sapiens 35-40 22113336-4 2011 The data revealed that, in the atherosclerotic animal model, the protein HMGB1 and its gene expression were increased and that fluvastatin treatment significantly reduced the release of HMGB1 into the extracellular space. Fluvastatin 127-138 high mobility group box 1 Homo sapiens 186-191 21922289-0 2011 Vascular anti-inflammatory effects of curcumin on HMGB1-mediated responses in vitro. Curcumin 38-46 high mobility group box 1 Homo sapiens 50-55 22014880-6 2011 MAIN OUTCOME MEASURE(S): TLR4 in the immortalized progesterone receptor-expressing human endometrial epithelial cell line, EM-PR, was activated with lipopolysaccharide and high-mobility group box 1 (LPS/HMGB1) in the presence or absence of the synthetic progestin dienogest or endogenous progesterone. dienogest 264-273 high mobility group box 1 Homo sapiens 172-197 22014880-6 2011 MAIN OUTCOME MEASURE(S): TLR4 in the immortalized progesterone receptor-expressing human endometrial epithelial cell line, EM-PR, was activated with lipopolysaccharide and high-mobility group box 1 (LPS/HMGB1) in the presence or absence of the synthetic progestin dienogest or endogenous progesterone. Progesterone 50-62 high mobility group box 1 Homo sapiens 172-197 22014880-6 2011 MAIN OUTCOME MEASURE(S): TLR4 in the immortalized progesterone receptor-expressing human endometrial epithelial cell line, EM-PR, was activated with lipopolysaccharide and high-mobility group box 1 (LPS/HMGB1) in the presence or absence of the synthetic progestin dienogest or endogenous progesterone. Progesterone 50-62 high mobility group box 1 Homo sapiens 203-208 21922289-4 2011 However, the effects of curcumin on HMGB1-mediated proinflammatory responses have not been studied. Curcumin 24-32 high mobility group box 1 Homo sapiens 36-41 21922289-5 2011 METHODS: The anti-inflammatory activities of curcumin were determined by measuring solute flux, leukocyte adhesion and migration and activation of proinflammatory proteins in HMGB1-activated human umbilical vein endothelial cells. Curcumin 45-53 high mobility group box 1 Homo sapiens 175-180 21922289-6 2011 RESULTS: Curcumin inhibited the release of HMGB1 by lipopolysaccharide (LPS)- and HMGB1-mediated barrier disruption, neutrophil adhesion and migration, and expression of cell adhesion molecules. Curcumin 9-17 high mobility group box 1 Homo sapiens 43-48 21922289-6 2011 RESULTS: Curcumin inhibited the release of HMGB1 by lipopolysaccharide (LPS)- and HMGB1-mediated barrier disruption, neutrophil adhesion and migration, and expression of cell adhesion molecules. Curcumin 9-17 high mobility group box 1 Homo sapiens 82-87 21922289-7 2011 Further studies revealed that curcumin down-regulated the cell surface receptor of HMGB1 in human endothelial cells. Curcumin 30-38 high mobility group box 1 Homo sapiens 83-88 21922289-8 2011 CONCLUSION: These findings suggest that curcumin exerts anti-inflammatory effects in HMGB1-mediated proinflammatory responses, endorsing its usefulness as therapy for vascular inflammatory diseases. Curcumin 40-48 high mobility group box 1 Homo sapiens 85-90 21273029-3 2011 We investigated here the effects of endotoxin removal by polymyxin B-immobilized polystyrene fiber (PMX-F) treatment on circulating levels of HMGB1, soluble RAGE (sRAGE), and interleukin-6 (IL-6) in septic shock patients. Polystyrenes 81-92 high mobility group box 1 Homo sapiens 142-147 21273029-3 2011 We investigated here the effects of endotoxin removal by polymyxin B-immobilized polystyrene fiber (PMX-F) treatment on circulating levels of HMGB1, soluble RAGE (sRAGE), and interleukin-6 (IL-6) in septic shock patients. pmx-f 100-105 high mobility group box 1 Homo sapiens 142-147 21273029-9 2011 The changes in endotoxin obtained by PMX-F treatment were correlated with those in HMGB1, sRAGE, and IL-6. pmx-f 37-42 high mobility group box 1 Homo sapiens 83-88 22071811-6 2011 MAIN RESULTS: A total of 167 studies were included in this 2011 update.The effect of sodium reduction in normotensive Caucasians was SBP -1.27 mmHg (95% CI: -1.88, -0.66; p=0.0001), DBP -0.05 mmHg (95% CI: -0.51, 0.42; p=0.85). Sodium 85-91 high mobility group box 1 Homo sapiens 133-139 21273029-11 2011 CONCLUSIONS: Our present study suggests that PMX-F treatment could block the HMGB1-RAGE axis in patients with septic shock via removal of endotoxin-induced inflammatory reactions. pmx-f 45-50 high mobility group box 1 Homo sapiens 77-82 22169720-4 2011 The molecular weight and identity of HMGB1 was confirmed by SDS-PAGE and Western blot. Sodium Dodecyl Sulfate 60-63 high mobility group box 1 Homo sapiens 37-42 22169720-5 2011 RESULTS: A sharp stained protein band with a molecular weight of about 26 kD was obtained by SDS-PAGE analysis and shown to be HMGB1 confirmed by Western blot. Sodium Dodecyl Sulfate 93-96 high mobility group box 1 Homo sapiens 127-132 21294652-8 2011 HMGB1, a protein thiol, weakly expressed in healthy muscles, increases during regeneration in parallel with the antioxidant response in both fibers and leukocytes. Sulfhydryl Compounds 17-22 high mobility group box 1 Homo sapiens 0-5 21922361-9 2011 Serum levels of tumor necrosis factor-alpha, interleukin-6, and high-mobility group box 1 were higher in the CT group than in the IIT group. ct 109-111 high mobility group box 1 Homo sapiens 64-89 21691146-8 2011 Loss of either HMGB1 or HSPB1 results in a phenotypically similar deficiency in mitophagy typified by mitochondrial fragmentation with decreased aerobic respiration and adenosine triphosphate (ATP) production. Adenosine Triphosphate 169-191 high mobility group box 1 Homo sapiens 15-20 21691146-8 2011 Loss of either HMGB1 or HSPB1 results in a phenotypically similar deficiency in mitophagy typified by mitochondrial fragmentation with decreased aerobic respiration and adenosine triphosphate (ATP) production. Adenosine Triphosphate 193-196 high mobility group box 1 Homo sapiens 15-20 21617575-0 2011 Peroxisome proliferator-activated receptor gamma agonist troglitazone inhibits high mobility group box 1 expression in endothelial cells via suppressing transcriptional activity of nuclear factor kappaB and activator protein 1. Troglitazone 57-69 high mobility group box 1 Homo sapiens 79-104 21756977-8 2011 Curculigoside A 20 mg/kg attenuated histopathological damage, decreased cerebral Evans Blue extravasation, inhibited NF-kappaB activation and reduced HMGB1 expression. curculigoside 0-15 high mobility group box 1 Homo sapiens 150-155 21756977-9 2011 These data show that Curculigoside A protects brain against I/R injury with a favorable therapeutic time-window by alleviating cerebral I/R injury and attenuating blood-brain barrier (BBB) breakdown, and its protective effects may involve HMGB1 and NF-kappaB signaling pathway. curculigoside 21-36 high mobility group box 1 Homo sapiens 239-244 21457762-0 2011 CKD712, (S)-1-(alpha-naphthylmethyl)-6,7-dihydroxy-1,2,3,4-tetrahydroisoquinoline, inhibits the lipopolysaccharide-stimulated secretion of HMGB1 by inhibiting PI3K and classical protein kinase C. CKD712, (S)-1-(alpha-naphthylmethyl)-6,7-dihydroxy-1,2,3,4-tetrahydroisoquinoline, was considered as a new effective drug candidate to sepsis, based on its anti-inflammatory activity. YS 49 8-81 high mobility group box 1 Homo sapiens 139-144 21457762-3 2011 CKD712 can reduce Gram-negative lipopolysaccharide (LPS)- and Gram-positive lipoteichoic acid (LTA)-stimulated HMGB1 secretion in RAW264.7 and human peripheral blood monocytes (PBMo), and also reduce LPS-induced nucleocytoplasmic translocation of HMGB1 1h before or after LPS treatment. positive 67-75 high mobility group box 1 Homo sapiens 111-116 21457762-3 2011 CKD712 can reduce Gram-negative lipopolysaccharide (LPS)- and Gram-positive lipoteichoic acid (LTA)-stimulated HMGB1 secretion in RAW264.7 and human peripheral blood monocytes (PBMo), and also reduce LPS-induced nucleocytoplasmic translocation of HMGB1 1h before or after LPS treatment. lipoteichoic acid 76-93 high mobility group box 1 Homo sapiens 111-116 21457762-3 2011 CKD712 can reduce Gram-negative lipopolysaccharide (LPS)- and Gram-positive lipoteichoic acid (LTA)-stimulated HMGB1 secretion in RAW264.7 and human peripheral blood monocytes (PBMo), and also reduce LPS-induced nucleocytoplasmic translocation of HMGB1 1h before or after LPS treatment. lipoteichoic acid 95-98 high mobility group box 1 Homo sapiens 111-116 21457762-3 2011 CKD712 can reduce Gram-negative lipopolysaccharide (LPS)- and Gram-positive lipoteichoic acid (LTA)-stimulated HMGB1 secretion in RAW264.7 and human peripheral blood monocytes (PBMo), and also reduce LPS-induced nucleocytoplasmic translocation of HMGB1 1h before or after LPS treatment. lipoteichoic acid 95-98 high mobility group box 1 Homo sapiens 247-252 21696758-7 2011 Furthermore, there was a negative correlation between the serum HMGB-1 levels and PaO2/FiO2 ratio on POD3. pao2 82-86 high mobility group box 1 Homo sapiens 64-70 21696758-7 2011 Furthermore, there was a negative correlation between the serum HMGB-1 levels and PaO2/FiO2 ratio on POD3. fio2 87-91 high mobility group box 1 Homo sapiens 64-70 21617575-4 2011 Herein, we provide evidence that PPARgamma agonist troglitazone, a member of the TZD class, modulates HMGB1 expression in the endothelial cell line EA.hy926 and propose a potential mechanism for that. Troglitazone 51-63 high mobility group box 1 Homo sapiens 102-107 21617575-4 2011 Herein, we provide evidence that PPARgamma agonist troglitazone, a member of the TZD class, modulates HMGB1 expression in the endothelial cell line EA.hy926 and propose a potential mechanism for that. 2,4-thiazolidinedione 81-84 high mobility group box 1 Homo sapiens 102-107 21617575-6 2011 Troglitazone inhibited the basal and LPS-stimulated HMGB1 expression at the mRNA level and protein level. Troglitazone 0-12 high mobility group box 1 Homo sapiens 52-57 21617575-7 2011 A luciferase reporter assay showed that troglitazone inhibited not only the transcriptional activation of the HMGB1 promoter but also activities of heterologous promoters driven by nuclear factor kappaB (NF-kappaB) or activator protein 1 (AP-1) response elements. Troglitazone 40-52 high mobility group box 1 Homo sapiens 110-115 21617575-8 2011 Altogether, these data suggest that NF-kappaB and AP-1 may participate in the inhibitory effect on HMGB1 transcription induced by troglitazone. Troglitazone 130-142 high mobility group box 1 Homo sapiens 99-104 21617575-9 2011 Activation of PPARgamma by troglitazone is effective for HMGB1 inhibition via suppressing NF-kappaB and AP-1 transcriptional activity in endothelial cells, which provides a new potential strategy to suppress excessive HMGB1 in inflammatory diseases. Troglitazone 27-39 high mobility group box 1 Homo sapiens 57-62 21617575-9 2011 Activation of PPARgamma by troglitazone is effective for HMGB1 inhibition via suppressing NF-kappaB and AP-1 transcriptional activity in endothelial cells, which provides a new potential strategy to suppress excessive HMGB1 in inflammatory diseases. Troglitazone 27-39 high mobility group box 1 Homo sapiens 218-223 21617575-3 2011 Because many proinflammatory cytokines expression were suppressed by thiazolidinediones (TZDs), agonists for nuclear receptor peroxisome proliferator-activated receptor gamma (PPARgamma), whether TZDs can inhibit HMGB1 expression and function is of great interest, however, it remains unknown. Thiazolidinediones 196-200 high mobility group box 1 Homo sapiens 213-218 21116767-0 2011 Extracellular HMGB1 released by NMDA treatment confers neuronal apoptosis via RAGE-p38 MAPK/ERK signaling pathway. N-Methylaspartate 32-36 high mobility group box 1 Homo sapiens 14-19 21487246-5 2011 Hydrogen peroxide (H 2O 2) and loss of superoxide dismutase 1 (SOD1)-mediated oxidative stress promotes cytosolic HMGB1 expression and extracellular release. Hydrogen Peroxide 0-17 high mobility group box 1 Homo sapiens 114-119 21487246-5 2011 Hydrogen peroxide (H 2O 2) and loss of superoxide dismutase 1 (SOD1)-mediated oxidative stress promotes cytosolic HMGB1 expression and extracellular release. Water 19-23 high mobility group box 1 Homo sapiens 114-119 21740486-0 2011 Synergistic requirement of orphan nonamer-like elements and DNA bending enhanced by HMGB1 for RAG-mediated nicking at cryptic 12-RSS but not authentic 12-RSS. 12-rss 126-132 high mobility group box 1 Homo sapiens 84-89 21740486-5 2011 The cryptic nicking site within the TEL fragment was cleaved by RAG proteins essentially depending on a 12-RSS framework, and the nicking activity was enhanced synergistically by both HMGB1 and orphan nonamer-like (NL) sequences, which do not possess counterpart heptamers. RSS 107-110 high mobility group box 1 Homo sapiens 184-189 21740486-7 2011 Collectively, we would propose the mechanism of HMGB1-dependent enhancement of RAG-mediated nicking at a cryptic RSS through enhanced DNA bending. RSS 113-116 high mobility group box 1 Homo sapiens 48-53 21520052-6 2011 Pretreatment with c-Src inhibitor (PP2), Akt inhibitor and NF-kappaB inhibitor (pyrrolidine dithiocarbamate and L-1-tosylamido-2-phenylenylethyl chloromethyl ketone) also inhibited the potentiating action of HMGB-1. l-1-tosylamido-2-phenylenylethyl chloromethyl ketone 112-164 high mobility group box 1 Homo sapiens 208-214 21116767-8 2011 A series of RAGE and HMGB1 co-immunoprecipitation experiments in the presence of SB203580 and PD98059 (p38 MAPK and ERK inhibitors, respectively) demonstrated that RAGE-p38 MAPK and RAGE-ERK pathway might underlie extracellular HMGB1-mediated neuronal apoptosis. SB 203580 81-89 high mobility group box 1 Homo sapiens 21-26 21116767-8 2011 A series of RAGE and HMGB1 co-immunoprecipitation experiments in the presence of SB203580 and PD98059 (p38 MAPK and ERK inhibitors, respectively) demonstrated that RAGE-p38 MAPK and RAGE-ERK pathway might underlie extracellular HMGB1-mediated neuronal apoptosis. 2-(2-amino-3-methoxyphenyl)-4H-1-benzopyran-4-one 94-101 high mobility group box 1 Homo sapiens 21-26 21116767-4 2011 HMGB1 was found to be accumulated in NMDA-treated primary cortical culture media, and media collected from these cultures were able to induce neuronal cell death when added to fresh primary cortical cultures. N-Methylaspartate 37-41 high mobility group box 1 Homo sapiens 0-5 21116767-5 2011 However, HMGB1-depleted NMDA-conditioned media produced by HMGB1 siRNA transfection or by preincubation with anti-HMGB1 antibody or with HMGB1 A box failed to induce neuronal cell death. N-Methylaspartate 24-28 high mobility group box 1 Homo sapiens 9-14 21116767-5 2011 However, HMGB1-depleted NMDA-conditioned media produced by HMGB1 siRNA transfection or by preincubation with anti-HMGB1 antibody or with HMGB1 A box failed to induce neuronal cell death. N-Methylaspartate 24-28 high mobility group box 1 Homo sapiens 59-64 21116767-5 2011 However, HMGB1-depleted NMDA-conditioned media produced by HMGB1 siRNA transfection or by preincubation with anti-HMGB1 antibody or with HMGB1 A box failed to induce neuronal cell death. N-Methylaspartate 24-28 high mobility group box 1 Homo sapiens 59-64 21116767-5 2011 However, HMGB1-depleted NMDA-conditioned media produced by HMGB1 siRNA transfection or by preincubation with anti-HMGB1 antibody or with HMGB1 A box failed to induce neuronal cell death. N-Methylaspartate 24-28 high mobility group box 1 Homo sapiens 59-64 21116767-6 2011 Furthermore, siRNA-mediated HMGB1 knockdown substantially suppressed NMDA- or Zn(2+)-induced cell death. N-Methylaspartate 69-73 high mobility group box 1 Homo sapiens 28-33 21116767-6 2011 Furthermore, siRNA-mediated HMGB1 knockdown substantially suppressed NMDA- or Zn(2+)-induced cell death. Zinc 78-84 high mobility group box 1 Homo sapiens 28-33 21426233-0 2011 18beta-glycyrrhetic acid inhibits immune activation triggered by HMGB1, a pro-inflammatory protein found in the tear fluid during conjunctivitis and blepharitis. Glycyrrhetinic Acid 0-24 high mobility group box 1 Homo sapiens 65-70 21884474-6 2011 Among the four membranes, surface-treated polyacrylonitrile (AN69ST) showed the highest capacity to adsorb HMGB1; it adsorbed nearly 100 microg of HMGB1 in the initial 60 min and showed a markedly high clearance rate (60.8 +- 5.0 mL/min) at 15 min. polyacrylonitrile 42-59 high mobility group box 1 Homo sapiens 107-112 21884474-6 2011 Among the four membranes, surface-treated polyacrylonitrile (AN69ST) showed the highest capacity to adsorb HMGB1; it adsorbed nearly 100 microg of HMGB1 in the initial 60 min and showed a markedly high clearance rate (60.8 +- 5.0 mL/min) at 15 min. polyacrylonitrile 42-59 high mobility group box 1 Homo sapiens 147-152 21884474-8 2011 Although the highest sieving coefficient for HMGB1 was obtained with the high cut-off polyarylethersulfone membrane, which correlated with a constant filtrate clearance rate, albumin loss was observed. polyarylethersulfone 86-106 high mobility group box 1 Homo sapiens 45-50 21602432-5 2011 We found that only anthracyclines induced a rapid translocation of calreticulin, HSP70, and HSP90 to the cell surface and the release of HMGB1 12 hours after the treatment. Anthracyclines 19-33 high mobility group box 1 Homo sapiens 137-142 21642542-5 2011 Short hairpin RNA-mediated knockdown of HMGB1 (HMGB1 KD) in tumor cells did not affect tumor cell growth but uncovered naturally acquired long-lasting tumor-specific IFN-gamma- or TNF-alpha-producing CD8 T cell responses and attenuated their ability to induce Treg, leading to naturally acquired CD8 T cell- or IFN-gamma-dependent tumor rejection. treg 260-264 high mobility group box 1 Homo sapiens 40-45 21642542-5 2011 Short hairpin RNA-mediated knockdown of HMGB1 (HMGB1 KD) in tumor cells did not affect tumor cell growth but uncovered naturally acquired long-lasting tumor-specific IFN-gamma- or TNF-alpha-producing CD8 T cell responses and attenuated their ability to induce Treg, leading to naturally acquired CD8 T cell- or IFN-gamma-dependent tumor rejection. treg 260-264 high mobility group box 1 Homo sapiens 47-52 21610098-6 2011 In addition, HMGB-1 prevents the pro-neurotoxic effect of tPA, by blocking its interaction with N-methyl-D-aspartate (NMDA) receptors and the attendant potentiation of NMDA-induced neuronal Ca2+ influx. N-Methylaspartate 118-122 high mobility group box 1 Homo sapiens 13-19 21565414-0 2011 Fluvastatin reduces the high mobility group box 1 protein expression in hyperlipidemia. Fluvastatin 0-11 high mobility group box 1 Homo sapiens 24-49 21426233-6 2011 The authors also report that 18beta-glycyrrhetic acid impairs antibody recognition of HMGB1, suggesting direct binding to the protein. Glycyrrhetinic Acid 29-53 high mobility group box 1 Homo sapiens 86-91 21426233-7 2011 Accordingly, 18beta-glycyrrhetic acid prevented HMGB1-dependent COX2 expression and cluster formation in primary cultures of human macrophages. Glycyrrhetinic Acid 13-37 high mobility group box 1 Homo sapiens 48-53 21355578-1 2011 HMGB1, one of the most abundant nuclear proteins, has a strong binding affinity for cisplatin-modified DNA. Cisplatin 84-93 high mobility group box 1 Homo sapiens 0-5 21294273-0 2011 HMGB1-carbenoxolone interactions: dynamics insights from combined nuclear magnetic resonance and molecular dynamics. Carbenoxolone 6-19 high mobility group box 1 Homo sapiens 0-5 21294273-3 2011 Herein, we have combined NMR relaxation experiments and residual dipolar coupling (RDC) measurements with molecular dynamics (MD) simulations to study the effects of the anti-inflammatory drug carbenoxolone (CBNX) on the conformational properties and on the internal dynamics of a subdomain (box A) of high-mobility group B protein (HMGB1). Carbenoxolone 193-206 high mobility group box 1 Homo sapiens 333-338 21294273-3 2011 Herein, we have combined NMR relaxation experiments and residual dipolar coupling (RDC) measurements with molecular dynamics (MD) simulations to study the effects of the anti-inflammatory drug carbenoxolone (CBNX) on the conformational properties and on the internal dynamics of a subdomain (box A) of high-mobility group B protein (HMGB1). Carbenoxolone 208-212 high mobility group box 1 Homo sapiens 333-338 21443191-3 2011 NMR structures of two bent DNA oligomers, only one of which binds strongly to HMGB1, revealed that the presence of a pocket structure on the minor groove is crucial for strong binding through penetration of a phenylalanine residue. Phenylalanine 209-222 high mobility group box 1 Homo sapiens 78-83 21355578-2 2011 It has been proposed that HMGB1 enhances the anticancer efficacy of cisplatin by shielding platinated DNA lesions from repair. Cisplatin 68-77 high mobility group box 1 Homo sapiens 26-31 21355578-3 2011 Two cysteine residues in HMGB1 domain A form a reversible disulfide bond under mildly oxidizing conditions. Cysteine 4-12 high mobility group box 1 Homo sapiens 25-30 21355578-3 2011 Two cysteine residues in HMGB1 domain A form a reversible disulfide bond under mildly oxidizing conditions. Disulfides 58-67 high mobility group box 1 Homo sapiens 25-30 21355578-5 2011 The binding affinities of singly and doubly mutated HMGB1 domain A, respectively deficient in one or both cysteine residues that form the disulfide bond, are unaffected by changes in external redox conditions. Cysteine 106-114 high mobility group box 1 Homo sapiens 52-57 21355578-5 2011 The binding affinities of singly and doubly mutated HMGB1 domain A, respectively deficient in one or both cysteine residues that form the disulfide bond, are unaffected by changes in external redox conditions. Disulfides 138-147 high mobility group box 1 Homo sapiens 52-57 21355578-6 2011 The redox-dependent nature of the binding of HMGB1 domain A to cisplatin-modified DNA suggests that formation of the intradomain disulfide bond induces a conformational change that disfavors binding to cisplatin-modified DNA. Cisplatin 63-72 high mobility group box 1 Homo sapiens 45-50 21355578-6 2011 The redox-dependent nature of the binding of HMGB1 domain A to cisplatin-modified DNA suggests that formation of the intradomain disulfide bond induces a conformational change that disfavors binding to cisplatin-modified DNA. Disulfides 129-138 high mobility group box 1 Homo sapiens 45-50 21355578-6 2011 The redox-dependent nature of the binding of HMGB1 domain A to cisplatin-modified DNA suggests that formation of the intradomain disulfide bond induces a conformational change that disfavors binding to cisplatin-modified DNA. Cisplatin 202-211 high mobility group box 1 Homo sapiens 45-50 21355578-8 2011 The results of this study reveal that the cellular redox environment can influence the interaction of HMGB1 with the platinated DNA and suggest that the redox state of the A domain is a potential factor in regulating the role of the protein in modulating the activity of cisplatin as an anticancer drug. Cisplatin 271-280 high mobility group box 1 Homo sapiens 102-107 21634123-6 2011 Lysine-rich histone H1 facilitates the binding of the HMGB1 with DNA by screening the negatively charged groups of the sugar-phosphate backbone of DNA and dicarboxylic amino-acid residues in the C-terminal domain of the HMGB1 protein. Lysine 0-6 high mobility group box 1 Homo sapiens 54-59 20969478-6 2011 HMGB1 causes activation of nicotinamide adenine dinucleotide phosphate oxidase and increased reactive oxygen species production in neutrophils. Reactive Oxygen Species 93-116 high mobility group box 1 Homo sapiens 0-5 20969478-8 2011 Antioxidants such as ethyl pyruvate, quercetin, green tea, N-acetylcysteine, and curcumin are protective in the setting of experimental infection/sepsis and injury including ischemia-reperfusion, partly through attenuating HMGB1 release and systemic accumulation. Acetylcysteine 59-75 high mobility group box 1 Homo sapiens 223-228 20969478-8 2011 Antioxidants such as ethyl pyruvate, quercetin, green tea, N-acetylcysteine, and curcumin are protective in the setting of experimental infection/sepsis and injury including ischemia-reperfusion, partly through attenuating HMGB1 release and systemic accumulation. Curcumin 81-89 high mobility group box 1 Homo sapiens 223-228 21186276-0 2011 Endogenous HMGB1 contributes to ischemia-reperfusion-induced myocardial apoptosis by potentiating the effect of TNF-&alpha;/JNK. Adenosine Monophosphate 117-120 high mobility group box 1 Homo sapiens 11-16 21634123-0 2011 [The structure of the complexes of DNA with chromosomal protein HMGB1 and histone H1 in the presence of manganese ions. Manganese 104-113 high mobility group box 1 Homo sapiens 64-69 21186276-13 2011 Furthermore, inhibition of JNK (SP-600125) prevented the myocyte apoptosis induced by a TNF-alpha/HMGB1 cocktail. pyrazolanthrone 32-41 high mobility group box 1 Homo sapiens 98-103 21634123-6 2011 Lysine-rich histone H1 facilitates the binding of the HMGB1 with DNA by screening the negatively charged groups of the sugar-phosphate backbone of DNA and dicarboxylic amino-acid residues in the C-terminal domain of the HMGB1 protein. Lysine 0-6 high mobility group box 1 Homo sapiens 220-225 21634123-6 2011 Lysine-rich histone H1 facilitates the binding of the HMGB1 with DNA by screening the negatively charged groups of the sugar-phosphate backbone of DNA and dicarboxylic amino-acid residues in the C-terminal domain of the HMGB1 protein. Sugar Phosphates 119-134 high mobility group box 1 Homo sapiens 54-59 21634123-6 2011 Lysine-rich histone H1 facilitates the binding of the HMGB1 with DNA by screening the negatively charged groups of the sugar-phosphate backbone of DNA and dicarboxylic amino-acid residues in the C-terminal domain of the HMGB1 protein. Sugar Phosphates 119-134 high mobility group box 1 Homo sapiens 220-225 21634123-9 2011 Manganese ions significantly modify the character of interactions between the components in the triple DNA-HMGB1-H1 complex. Manganese 0-9 high mobility group box 1 Homo sapiens 107-112 21248133-11 2011 The treatment of microglia with HMGB1 led to membrane translocation of p47(phox) (a cytosolic subunit of NADPH oxidase) and consequent superoxide release, which required the presence of Mac1. Superoxides 135-145 high mobility group box 1 Homo sapiens 32-37 21149855-5 2011 Butyric acid, an extracellular metabolite from periodontopathic bacteria populating the periodontal pocket, induced the passive release of HMGB1 as a result of eliciting necrosis in the human gingival epithelial cell line. Butyric Acid 0-12 high mobility group box 1 Homo sapiens 139-144 20739958-0 2011 Synergistic anti-tumor effects between oncolytic vaccinia virus and paclitaxel are mediated by the IFN response and HMGB1. Paclitaxel 68-78 high mobility group box 1 Homo sapiens 116-121 21300299-7 2011 In trained patients, decreased HMGB1 levels were significantly associated with the improvement in peak oxygen consumption (beta = -3.879, P = .003) and heart rate recovery (beta = -2.492, P = .002), and with reduced left ventricular end-diastolic volume (beta = 1.412, P = .001) and wall motion score index (beta = 1.138, P = .002). Oxygen 103-109 high mobility group box 1 Homo sapiens 31-36 21647302-0 2011 The DNA binding and bending activities of truncated tail-less HMGB1 protein are differentially affected by Lys-2 and Lys-81 residues and their acetylation. Lysine 107-110 high mobility group box 1 Homo sapiens 62-67 21283827-9 2011 These effects were specifically linked to HMGB1 since they were blocked by glycyrrhizin or by a neutralizing anti-HMGB1 antibody, and were mediated through TLR2 and the receptor for Advanced Glycation End-products (RAGE). Glycyrrhizic Acid 75-87 high mobility group box 1 Homo sapiens 42-47 21167721-8 2011 Expectedly, DR396, which is the most potent and specific inhibitor of DNase gamma, was found to almost completely inhibit both HMGB1 release and internucleosomal DNA cleavage in HeLa S3 cells transfected with DNase gamma expression vector and stably expressing DNase gamma (HeLa S3/gamma cells). 4-(4,6-dichloro-(1,3,5)-triazin-2-ylamino)-2-(6-hydroxy-3-oxo-3H-xanthen-9-yl)benzoic acid 12-17 high mobility group box 1 Homo sapiens 127-132 21647302-1 2011 The role of lysines 2 and 81 as target sites for acetylation in full-length HMGB1 and truncated tail-less protein, respectively, has been studied by mutation analysis for the abilities of these proteins to bind and bend DNA. Lysine 12-19 high mobility group box 1 Homo sapiens 76-81 21647302-2 2011 The DNA bending ability of truncated tail-less HMGB1 containing Lys-2 mutated to alanine does not differ from that of the wild-type protein, while the same mutation of Lys-81 reduced the bending capacity of the mutant protein. Lysine 64-67 high mobility group box 1 Homo sapiens 47-52 21647302-3 2011 These data demonstrate that Lys-81 is critical for the DNA bending ability of truncated HMGB1. Lysine 28-31 high mobility group box 1 Homo sapiens 88-93 21647302-5 2011 On the contrary, the acetylation of Lys-81 in the mutant K2/A2 enhanced the bending potential of HMGB1 C. Regarding the ability of HMGB1 to specifically bind bent DNA, the individual mutations of either K2 or K81 as well as the double mutation of both residues to alanine were found to completely abolish binding of truncated tail-less HMGB1 to cisplatin-modified DNA. Lysine 36-39 high mobility group box 1 Homo sapiens 97-102 21647302-5 2011 On the contrary, the acetylation of Lys-81 in the mutant K2/A2 enhanced the bending potential of HMGB1 C. Regarding the ability of HMGB1 to specifically bind bent DNA, the individual mutations of either K2 or K81 as well as the double mutation of both residues to alanine were found to completely abolish binding of truncated tail-less HMGB1 to cisplatin-modified DNA. Lysine 36-39 high mobility group box 1 Homo sapiens 131-136 21647302-5 2011 On the contrary, the acetylation of Lys-81 in the mutant K2/A2 enhanced the bending potential of HMGB1 C. Regarding the ability of HMGB1 to specifically bind bent DNA, the individual mutations of either K2 or K81 as well as the double mutation of both residues to alanine were found to completely abolish binding of truncated tail-less HMGB1 to cisplatin-modified DNA. Lysine 36-39 high mobility group box 1 Homo sapiens 131-136 21647302-5 2011 On the contrary, the acetylation of Lys-81 in the mutant K2/A2 enhanced the bending potential of HMGB1 C. Regarding the ability of HMGB1 to specifically bind bent DNA, the individual mutations of either K2 or K81 as well as the double mutation of both residues to alanine were found to completely abolish binding of truncated tail-less HMGB1 to cisplatin-modified DNA. Alanine 264-271 high mobility group box 1 Homo sapiens 131-136 21647302-5 2011 On the contrary, the acetylation of Lys-81 in the mutant K2/A2 enhanced the bending potential of HMGB1 C. Regarding the ability of HMGB1 to specifically bind bent DNA, the individual mutations of either K2 or K81 as well as the double mutation of both residues to alanine were found to completely abolish binding of truncated tail-less HMGB1 to cisplatin-modified DNA. Alanine 264-271 high mobility group box 1 Homo sapiens 131-136 21647302-5 2011 On the contrary, the acetylation of Lys-81 in the mutant K2/A2 enhanced the bending potential of HMGB1 C. Regarding the ability of HMGB1 to specifically bind bent DNA, the individual mutations of either K2 or K81 as well as the double mutation of both residues to alanine were found to completely abolish binding of truncated tail-less HMGB1 to cisplatin-modified DNA. Cisplatin 345-354 high mobility group box 1 Homo sapiens 131-136 21647302-5 2011 On the contrary, the acetylation of Lys-81 in the mutant K2/A2 enhanced the bending potential of HMGB1 C. Regarding the ability of HMGB1 to specifically bind bent DNA, the individual mutations of either K2 or K81 as well as the double mutation of both residues to alanine were found to completely abolish binding of truncated tail-less HMGB1 to cisplatin-modified DNA. Cisplatin 345-354 high mobility group box 1 Homo sapiens 131-136 21647302-6 2011 We conclude that unlike the case with the bending ability of truncated HMGB1, where Lys-81 has a primary function, Lys-2 and Lys-81 are both critical for the protein"s binding to cisplatin-modified DNA. Lysine 84-87 high mobility group box 1 Homo sapiens 71-76 21647302-6 2011 We conclude that unlike the case with the bending ability of truncated HMGB1, where Lys-81 has a primary function, Lys-2 and Lys-81 are both critical for the protein"s binding to cisplatin-modified DNA. Lysine 115-118 high mobility group box 1 Homo sapiens 71-76 21647302-6 2011 We conclude that unlike the case with the bending ability of truncated HMGB1, where Lys-81 has a primary function, Lys-2 and Lys-81 are both critical for the protein"s binding to cisplatin-modified DNA. Cisplatin 179-188 high mobility group box 1 Homo sapiens 71-76 21647302-8 2011 Any further substitutions at the acetylable lysines in the truncated form of HMGB1 do not have an additional effect. Lysine 44-51 high mobility group box 1 Homo sapiens 77-82 20803525-8 2010 Inhibition of SIRT1 by sirtinol or SIRT1 siRNA blocked the HMGB1-stimulated expression of RANKL and cytokines. sirtinol 23-31 high mobility group box 1 Homo sapiens 59-64 20845025-8 2010 ETR-P1/fl administration resulted in higher MAP at 6 and 9 h after CLP, less negative BE, lower mean pulmonary arterial pressure (mPAP)/MAP ratio at 9 h after CLP, and lower TNF-alpha, NOx, and HMGB-1 compared to the CLP group. etr-p1 0-6 high mobility group box 1 Homo sapiens 194-200 20845025-8 2010 ETR-P1/fl administration resulted in higher MAP at 6 and 9 h after CLP, less negative BE, lower mean pulmonary arterial pressure (mPAP)/MAP ratio at 9 h after CLP, and lower TNF-alpha, NOx, and HMGB-1 compared to the CLP group. fl 7-9 high mobility group box 1 Homo sapiens 194-200 20845025-11 2010 CONCLUSIONS: ETR-P1/fl treatment significantly attenuated the elevation of NOx, TNF-alpha, and HMGB-1, which improved the systemic hypotension, pulmonary hypertension, and blood gases, thereby causing improvement of survival time in a progressive neonatal sepsis CLP model. etr 13-16 high mobility group box 1 Homo sapiens 95-101 21602594-0 2011 Immunosuppressant cytoprotection correlates with HMGB1 suppression in primary astrocyte cultures exposed to combined oxygen-glucose deprivation. oxygen-glucose 117-131 high mobility group box 1 Homo sapiens 49-54 21602594-2 2011 Because high-mobility group box 1 (HMGB1) protein has recently been reported to be involved in ischemic brain injury, the purpose of the present study was to determine whether treatment with immunosuppressants could decrease the expression and release of HMGB1 in astrocytes exposed to simulated ischemic conditions (combined oxygen-glucose deprivation, OGD). oxygen-glucose 326-340 high mobility group box 1 Homo sapiens 35-40 21602594-2 2011 Because high-mobility group box 1 (HMGB1) protein has recently been reported to be involved in ischemic brain injury, the purpose of the present study was to determine whether treatment with immunosuppressants could decrease the expression and release of HMGB1 in astrocytes exposed to simulated ischemic conditions (combined oxygen-glucose deprivation, OGD). oxygen-glucose 326-340 high mobility group box 1 Homo sapiens 255-260 21602594-7 2011 Our data suggest that HMGB1 release may be partly an active process triggered by oxidative stress because the antioxidant N-acetylcysteine (NAC) clearly attenuated HMGB1 expression and release. Acetylcysteine 122-138 high mobility group box 1 Homo sapiens 22-27 21602594-7 2011 Our data suggest that HMGB1 release may be partly an active process triggered by oxidative stress because the antioxidant N-acetylcysteine (NAC) clearly attenuated HMGB1 expression and release. Acetylcysteine 122-138 high mobility group box 1 Homo sapiens 164-169 21602594-7 2011 Our data suggest that HMGB1 release may be partly an active process triggered by oxidative stress because the antioxidant N-acetylcysteine (NAC) clearly attenuated HMGB1 expression and release. Acetylcysteine 140-143 high mobility group box 1 Homo sapiens 22-27 21602594-7 2011 Our data suggest that HMGB1 release may be partly an active process triggered by oxidative stress because the antioxidant N-acetylcysteine (NAC) clearly attenuated HMGB1 expression and release. Acetylcysteine 140-143 high mobility group box 1 Homo sapiens 164-169 20163887-7 2010 Additionally, the HMGB1-induced activation of HUVECs was significantly inhibited by anti-RAGE monoclonal antibody and Ethyl pyruvate (EP) that had been shown to be an effective anti-inflammatory agent. ethyl pyruvate 118-132 high mobility group box 1 Homo sapiens 18-23 20568967-7 2010 One of the cellular cofactors known to play a key role during SB-mediated transposition is the high-mobility group DNA-binding protein-1 (HMGB1). Antimony 62-64 high mobility group box 1 Homo sapiens 95-136 20714791-8 2010 These findings indicate that atorvastatin attenuates HMGB1-induced vascular endothelial activation. Atorvastatin 29-41 high mobility group box 1 Homo sapiens 53-58 20714791-3 2010 The aim of our study was to investigate whether atorvastatin inhibits HMGB1-induced vascular endothelial activation, and elucidate the underlying molecular mechanism. Atorvastatin 48-60 high mobility group box 1 Homo sapiens 70-75 20714791-4 2010 In this study, we found that atorvastatin, at concentrations ranging from 0.1 to 10 muM, effectively and in a dose-dependent manner inhibited HMGB1-induced endothelial cells (ECs) activation. Atorvastatin 29-41 high mobility group box 1 Homo sapiens 142-147 20714791-5 2010 Incubation of ECs with 10 muM atorvastatin reduced adhesion molecules (ICAM-1 and E-selectin) expression concomitant with a significant inhibition in HMGB1-stimulated leukocyte-endothelial adhesion. Atorvastatin 30-42 high mobility group box 1 Homo sapiens 150-155 20714791-6 2010 Further experiments showed that atorvastatin markedly suppressed HMGB1-induced Toll like receptor 4 (TLR4) expression, Nuclear factor kappaB (NF-kappaB) nuclear translocation and DNA binding activity in ECs. Atorvastatin 32-44 high mobility group box 1 Homo sapiens 65-70 20935509-7 2010 Moreover, the intramolecular disulfide bridge (C23/45) of HMGB1 is required for binding to Beclin 1 and sustaining autophagy. Disulfides 29-38 high mobility group box 1 Homo sapiens 58-63 20568967-7 2010 One of the cellular cofactors known to play a key role during SB-mediated transposition is the high-mobility group DNA-binding protein-1 (HMGB1). Antimony 62-64 high mobility group box 1 Homo sapiens 138-143 20568967-10 2010 As a result, HSV/SB amplicon virions arrive prearmed with HMGB1 protein at levels sufficient for facilitating SB-mediated transposition in the transduced mammalian cell. Antimony 17-19 high mobility group box 1 Homo sapiens 58-63 20664360-0 2010 Preconditioning with high mobility group box 1 (HMGB1) induces lipoteichoic acid (LTA) tolerance. lipoteichoic acid 63-80 high mobility group box 1 Homo sapiens 21-46 20622903-5 2010 Diminished HMGB1 by short hairpin RNA transfection or inhibition of HMGB1 release by ethyl pyruvate or other small molecules led predominantly to apoptosis and decreased autophagy in stressed cancer cells. ethyl pyruvate 85-99 high mobility group box 1 Homo sapiens 68-73 20622903-6 2010 In this setting, reducible HMGB1 binds to the receptor for advanced glycation end products (RAGEs), but not to Toll-like receptor 4, induces Beclin1-dependent autophagy and promotes tumor resistance to alkylators (melphalan), tubulin disrupting agents (paclitaxel), DNA crosslinkers (ultraviolet light) and DNA intercalators (oxaliplatin or adriamycin). Paclitaxel 253-263 high mobility group box 1 Homo sapiens 27-32 20622903-6 2010 In this setting, reducible HMGB1 binds to the receptor for advanced glycation end products (RAGEs), but not to Toll-like receptor 4, induces Beclin1-dependent autophagy and promotes tumor resistance to alkylators (melphalan), tubulin disrupting agents (paclitaxel), DNA crosslinkers (ultraviolet light) and DNA intercalators (oxaliplatin or adriamycin). Oxaliplatin 326-337 high mobility group box 1 Homo sapiens 27-32 20622903-6 2010 In this setting, reducible HMGB1 binds to the receptor for advanced glycation end products (RAGEs), but not to Toll-like receptor 4, induces Beclin1-dependent autophagy and promotes tumor resistance to alkylators (melphalan), tubulin disrupting agents (paclitaxel), DNA crosslinkers (ultraviolet light) and DNA intercalators (oxaliplatin or adriamycin). Doxorubicin 341-351 high mobility group box 1 Homo sapiens 27-32 20819940-3 2010 Stimuli that enhance reactive oxygen species promote cytosolic translocation of HMGB1 and thereby enhance autophagic flux. Reactive Oxygen Species 21-44 high mobility group box 1 Homo sapiens 80-85 20819940-5 2010 Mutation of cysteine 106 (C106), but not the vicinal C23 and C45, of HMGB1 promotes cytosolic localization and sustained autophagy. Cysteine 12-20 high mobility group box 1 Homo sapiens 69-74 20819940-6 2010 Pharmacological inhibition of HMGB1 cytoplasmic translocation by agents such as ethyl pyruvate limits starvation-induced autophagy. ethyl pyruvate 80-94 high mobility group box 1 Homo sapiens 30-35 20819940-7 2010 Moreover, the intramolecular disulfide bridge (C23/45) of HMGB1 is required for binding to Beclin1 and sustaining autophagy. Disulfides 29-38 high mobility group box 1 Homo sapiens 58-63 20664360-0 2010 Preconditioning with high mobility group box 1 (HMGB1) induces lipoteichoic acid (LTA) tolerance. lipoteichoic acid 63-80 high mobility group box 1 Homo sapiens 48-53 20664360-0 2010 Preconditioning with high mobility group box 1 (HMGB1) induces lipoteichoic acid (LTA) tolerance. lipoteichoic acid 82-85 high mobility group box 1 Homo sapiens 21-46 20664360-0 2010 Preconditioning with high mobility group box 1 (HMGB1) induces lipoteichoic acid (LTA) tolerance. lipoteichoic acid 82-85 high mobility group box 1 Homo sapiens 48-53 20616036-0 2010 Programmed necrosis induced by asbestos in human mesothelial cells causes high-mobility group box 1 protein release and resultant inflammation. Asbestos 31-39 high mobility group box 1 Homo sapiens 74-99 20664360-3 2010 We tested the hypothesis that extracellular HMGB1 can induce tolerance to the bacterial product, lipoteichoic acid (LTA). lipoteichoic acid 97-114 high mobility group box 1 Homo sapiens 44-49 20664360-3 2010 We tested the hypothesis that extracellular HMGB1 can induce tolerance to the bacterial product, lipoteichoic acid (LTA). lipoteichoic acid 116-119 high mobility group box 1 Homo sapiens 44-49 20664360-5 2010 Denaturation of HMGB1 with boiling water or coincubation with anti-HMGB1 antibody abrogated the induction of tolerance to LTA. Water 35-40 high mobility group box 1 Homo sapiens 16-21 20664360-7 2010 These findings support the view that the induction of LTA tolerance by HMGB1 was not due to lipopolysaccharide contamination. lipoteichoic acid 54-57 high mobility group box 1 Homo sapiens 71-76 20182437-0 2010 High-mobility group box-1 protein in tracheal aspirates from premature infants: relationship with bronchopulmonary dysplasia and steroid therapy. Steroids 129-136 high mobility group box 1 Homo sapiens 0-25 20182437-3 2010 We studied the association between HMGB1 levels in tracheal aspirates (TAs) and adverse outcomes (BPD/death) in ventilated premature infants (VPIs) and modulation of HMGB1 levels with dexamethasone (Dex) use. Dexamethasone 184-197 high mobility group box 1 Homo sapiens 166-171 20182437-3 2010 We studied the association between HMGB1 levels in tracheal aspirates (TAs) and adverse outcomes (BPD/death) in ventilated premature infants (VPIs) and modulation of HMGB1 levels with dexamethasone (Dex) use. Dexamethasone 199-202 high mobility group box 1 Homo sapiens 166-171 20386869-5 2010 Pharmacologically relevant doses of dexamethasone, gold sodium thiomalate and chloroquine inhibited the extracellular release of HMGB1 in a dose-dependent mode. Dexamethasone 36-49 high mobility group box 1 Homo sapiens 129-134 20386869-5 2010 Pharmacologically relevant doses of dexamethasone, gold sodium thiomalate and chloroquine inhibited the extracellular release of HMGB1 in a dose-dependent mode. Gold Sodium Thiomalate 51-73 high mobility group box 1 Homo sapiens 129-134 20386869-5 2010 Pharmacologically relevant doses of dexamethasone, gold sodium thiomalate and chloroquine inhibited the extracellular release of HMGB1 in a dose-dependent mode. Chloroquine 78-89 high mobility group box 1 Homo sapiens 129-134 20386869-6 2010 Immunostaining demonstrated that dexamethasone caused intracellular HMGB1 retention. Dexamethasone 33-46 high mobility group box 1 Homo sapiens 68-73 20386869-9 2010 We conclude that dexamethasone, gold sodium thiomalate and chloroquine share a capacity to inhibit HMGB1 release from activated monocytes. Dexamethasone 17-30 high mobility group box 1 Homo sapiens 99-104 20386869-9 2010 We conclude that dexamethasone, gold sodium thiomalate and chloroquine share a capacity to inhibit HMGB1 release from activated monocytes. Gold Sodium Thiomalate 32-54 high mobility group box 1 Homo sapiens 99-104 20386869-9 2010 We conclude that dexamethasone, gold sodium thiomalate and chloroquine share a capacity to inhibit HMGB1 release from activated monocytes. Chloroquine 59-70 high mobility group box 1 Homo sapiens 99-104 20647757-4 2010 Combining genetic and cell death assays, we demonstrate that elevated FFA concentrations lead to necrotic cell death, as evidenced by loss of membrane integrity and release of nuclear HMGB1. Fatty Acids, Nonesterified 70-73 high mobility group box 1 Homo sapiens 184-189 20483361-5 2010 HMGB1 treatment for 24h significantly inhibited the current densities of heterologously expressed Kv4.3 and Kv4.2 in COS-7 cells and native I(to) in neonatal rat ventricular myocytes (NRVMs) in a dose-dependent manner. 9-(3-hydroxy-2-phosphonomethoxypropyl)guanine 20-23 high mobility group box 1 Homo sapiens 0-5 20483361-5 2010 HMGB1 treatment for 24h significantly inhibited the current densities of heterologously expressed Kv4.3 and Kv4.2 in COS-7 cells and native I(to) in neonatal rat ventricular myocytes (NRVMs) in a dose-dependent manner. carbonyl sulfide 117-120 high mobility group box 1 Homo sapiens 0-5 20616036-4 2010 Asbestos-exposed HM activate poly(ADP-ribose) polymerase, secrete H(2)O(2), deplete ATP, and translocate high-mobility group box 1 protein (HMGB1) from the nucleus to the cytoplasm, and into the extracellular space. Adenosine Triphosphate 84-87 high mobility group box 1 Homo sapiens 140-145 20616036-8 2010 HMGB1 levels in asbestos-exposed individuals were significantly higher than in nonexposed controls (P < 0.0001). Asbestos 16-24 high mobility group box 1 Homo sapiens 0-5 20616036-10 2010 Chemopreventive approaches aimed at inhibiting the chronic inflammatory response, and especially blocking HMGB1, may decrease the risk of malignant mesothelioma among asbestos-exposed cohorts. Asbestos 167-175 high mobility group box 1 Homo sapiens 106-111 20616036-4 2010 Asbestos-exposed HM activate poly(ADP-ribose) polymerase, secrete H(2)O(2), deplete ATP, and translocate high-mobility group box 1 protein (HMGB1) from the nucleus to the cytoplasm, and into the extracellular space. Asbestos 0-8 high mobility group box 1 Homo sapiens 105-130 20616036-4 2010 Asbestos-exposed HM activate poly(ADP-ribose) polymerase, secrete H(2)O(2), deplete ATP, and translocate high-mobility group box 1 protein (HMGB1) from the nucleus to the cytoplasm, and into the extracellular space. Asbestos 0-8 high mobility group box 1 Homo sapiens 140-145 20514401-7 2010 These results demonstrate that in metabolically stressed cancer cells, ROS induce a specific set of cellular proteins to form insoluble aggregates that are highly toxic to cells and trigger the necrosis-associated membrane rupture and HMGB1 release to promote tumour progression. Reactive Oxygen Species 71-74 high mobility group box 1 Homo sapiens 235-240 22993577-13 2010 Here, we present Western blotting and immunofluorescence microscopy data indicating that MK615 inhibited the expression of RAGE in SK-MEL28 cells, and suppressed the release of HMGB1 by SK-MEL28 cells. mk615 89-94 high mobility group box 1 Homo sapiens 177-182 20514411-4 2010 In solid tumours, ROS is produced by metabolic stress due to insufficient oxygen and glucose supply and induces necrosis that is known to promote tumour progression by releasing the proinflammatory cytokine HMGB1. Reactive Oxygen Species 18-21 high mobility group box 1 Homo sapiens 207-212 20512925-3 2010 We earlier showed that carboxylated glycans on the V-domain of RAGE promote the binding of HMGB1 and S100A8/A9. Polysaccharides 36-43 high mobility group box 1 Homo sapiens 91-96 20514411-5 2010 We observed that CuZnSOD and MnSOD overexpression prevents metabolic stress-induced necrosis and HMGB1 release by inhibiting mitochondrial ROS and intracellular O2- production in response to glucose depletion in two dimensional cell culture. Glucose 191-198 high mobility group box 1 Homo sapiens 97-102 20547845-0 2010 A critical cysteine is required for HMGB1 binding to Toll-like receptor 4 and activation of macrophage cytokine release. Cysteine 11-19 high mobility group box 1 Homo sapiens 36-41 20547845-8 2010 Surface plasmon resonance studies indicate that HMGB1 binds specifically to TLR4, and that this binding requires a cysteine in position 106. Cysteine 115-123 high mobility group box 1 Homo sapiens 48-53 20547845-10 2010 Inhibition of TLR4 binding with neutralizing anti-HMGB1 mAb or by mutating cysteine 106 prevents HMGB1 activation of cytokine release. Cysteine 75-83 high mobility group box 1 Homo sapiens 97-102 20928981-4 2010 In addition to advanced glycation end products (AGE), RAGE has other important ligands such as: high mobility group box 1 protein (HMGB1, also termed amphoterin), the group of calcium binding cellular factors S100 (also termed calgranulin), amiloid beta peptides and Mac-1, a beta-2 integrin (CD1lb/CD18). Calcium 176-183 high mobility group box 1 Homo sapiens 131-136 20928981-4 2010 In addition to advanced glycation end products (AGE), RAGE has other important ligands such as: high mobility group box 1 protein (HMGB1, also termed amphoterin), the group of calcium binding cellular factors S100 (also termed calgranulin), amiloid beta peptides and Mac-1, a beta-2 integrin (CD1lb/CD18). Calcium 176-183 high mobility group box 1 Homo sapiens 150-160 20618458-6 2010 Extracellular HMGB-1 levels markedly increased after hydrogen peroxide stimulation, but did not change after STS stimulation. Hydrogen Peroxide 53-70 high mobility group box 1 Homo sapiens 14-20 19906009-3 2010 A growing number of HMGB1 inhibitors ranging from neutralizing antibodies, endogenous hormones, to medicinal herb-derived small molecule HMGB1 inhibitors (such as nicotine, glycyrrhizin, tanshinones, and EGCG) are proven protective against lethal infection and ischemic injury. Nicotine 163-171 high mobility group box 1 Homo sapiens 20-25 19898867-4 2010 METHODS: In present study, we investigated HMGB1 expression and its prognostic significance in CRC by performing immunohistochemical analysis, using a total of 192 paraffin-embedded archival CRC samples. Paraffin 164-172 high mobility group box 1 Homo sapiens 43-48 20368109-7 2010 The recombinant protein of human HMGB1 A box was purificated by Ni(2+)-NTA chromatography and the inhibit effection of the purpose protern to the activation of monocyte stimulated by immunocomplex was identified by RT-PCR. nickel nitrilotriacetic acid 64-74 high mobility group box 1 Homo sapiens 33-38 20368109-9 2010 Western blot showed recombinant protein can specificly reacted with anti-human HMGB1 polyclonal antibody and anti-His-Tag polyclonal antibody.The purpose protein was found more than 90% after purified, and can effectively inhibit the production of BAFF, IFN-gamma and TNF-alpha in monocyte which were induced by IC. Histidine 114-117 high mobility group box 1 Homo sapiens 79-84 20302902-1 2010 BACKGROUND: High mobility group box 1 (HMGB1) is a highly conserved, ubiquitous protein that functions as an activator for inducing the immune response and can be released from neurons after glutamate excitotoxicity. Glutamic Acid 191-200 high mobility group box 1 Homo sapiens 12-37 20302902-1 2010 BACKGROUND: High mobility group box 1 (HMGB1) is a highly conserved, ubiquitous protein that functions as an activator for inducing the immune response and can be released from neurons after glutamate excitotoxicity. Glutamic Acid 191-200 high mobility group box 1 Homo sapiens 39-44 20133931-8 2010 BAL HMGB1 correlated positively with IL-1beta (r(s) = 0.438; P = 0.0006) and negatively with FEV(1) (r(s) = -0.570; P < 0.0001) and transfer factor of the lung for carbon monoxide (r(s) = -0.382; P = 0.0026). Carbon Monoxide 167-182 high mobility group box 1 Homo sapiens 4-9 20335795-3 2010 It is well known that the cationic portion of HMGB1 binds to heparin, which has abundant sulfates in its structure. Heparin 61-68 high mobility group box 1 Homo sapiens 46-51 20335795-3 2010 It is well known that the cationic portion of HMGB1 binds to heparin, which has abundant sulfates in its structure. Sulfates 89-97 high mobility group box 1 Homo sapiens 46-51 20335795-4 2010 In this study, we determined whether spherical sulfated cellulose (SC) efficiently adsorbed HMGB1, as well as other inflammatory mediators, in vitro. Cellulose 56-65 high mobility group box 1 Homo sapiens 92-97 20510002-2 2010 METHODS: Three specific siRNAs of HMGB1 were designed and synthesized, and were transiently transfected into HepG2 cells by Lipofectamine 2000. Lipofectamine 124-142 high mobility group box 1 Homo sapiens 34-39 20510002-6 2010 RESULTS: All of these specific HMGB1-siRNAs (1, 2, 3) efficiently and specifically inhibited the expression of the HMGB1 gene, and the levels of HMGB1 mRNA were 1.147+/-0.024, 1.014+/-0.042, 0.435+/-0.055, respectively, in HMGB1-siRNAs transfection group, which were significantly lower than that in Lipofectamine 2000 alone group (1.411+/-0.065, P < 0.01). Lipofectamine 300-318 high mobility group box 1 Homo sapiens 31-36 20510002-6 2010 RESULTS: All of these specific HMGB1-siRNAs (1, 2, 3) efficiently and specifically inhibited the expression of the HMGB1 gene, and the levels of HMGB1 mRNA were 1.147+/-0.024, 1.014+/-0.042, 0.435+/-0.055, respectively, in HMGB1-siRNAs transfection group, which were significantly lower than that in Lipofectamine 2000 alone group (1.411+/-0.065, P < 0.01). Lipofectamine 300-318 high mobility group box 1 Homo sapiens 115-120 20510002-6 2010 RESULTS: All of these specific HMGB1-siRNAs (1, 2, 3) efficiently and specifically inhibited the expression of the HMGB1 gene, and the levels of HMGB1 mRNA were 1.147+/-0.024, 1.014+/-0.042, 0.435+/-0.055, respectively, in HMGB1-siRNAs transfection group, which were significantly lower than that in Lipofectamine 2000 alone group (1.411+/-0.065, P < 0.01). Lipofectamine 300-318 high mobility group box 1 Homo sapiens 115-120 20510002-6 2010 RESULTS: All of these specific HMGB1-siRNAs (1, 2, 3) efficiently and specifically inhibited the expression of the HMGB1 gene, and the levels of HMGB1 mRNA were 1.147+/-0.024, 1.014+/-0.042, 0.435+/-0.055, respectively, in HMGB1-siRNAs transfection group, which were significantly lower than that in Lipofectamine 2000 alone group (1.411+/-0.065, P < 0.01). Lipofectamine 300-318 high mobility group box 1 Homo sapiens 115-120 20332323-2 2010 Because selenium-binding protein 1 (SELENBP1, SBP1, hSP56) has been shown to bind selenium covalently and selenium deficiency has been associated with esophageal adenocarcinoma (EAC), we examined its role in EAC development and its potential effect on chemosensitivity in the presence of selenium. Selenium 8-16 high mobility group box 1 Homo sapiens 46-50 20332323-2 2010 Because selenium-binding protein 1 (SELENBP1, SBP1, hSP56) has been shown to bind selenium covalently and selenium deficiency has been associated with esophageal adenocarcinoma (EAC), we examined its role in EAC development and its potential effect on chemosensitivity in the presence of selenium. Selenium 82-90 high mobility group box 1 Homo sapiens 46-50 20332323-2 2010 Because selenium-binding protein 1 (SELENBP1, SBP1, hSP56) has been shown to bind selenium covalently and selenium deficiency has been associated with esophageal adenocarcinoma (EAC), we examined its role in EAC development and its potential effect on chemosensitivity in the presence of selenium. Selenium 82-90 high mobility group box 1 Homo sapiens 46-50 20348922-5 2010 The proconvulsant effects of HMGB1, like those of interleukin-1beta (IL-1beta), are partly mediated by ifenprodil-sensitive N-methyl-d-aspartate (NMDA) receptors. ifenprodil 103-113 high mobility group box 1 Homo sapiens 29-34 19906009-3 2010 A growing number of HMGB1 inhibitors ranging from neutralizing antibodies, endogenous hormones, to medicinal herb-derived small molecule HMGB1 inhibitors (such as nicotine, glycyrrhizin, tanshinones, and EGCG) are proven protective against lethal infection and ischemic injury. Nicotine 163-171 high mobility group box 1 Homo sapiens 137-142 19906009-3 2010 A growing number of HMGB1 inhibitors ranging from neutralizing antibodies, endogenous hormones, to medicinal herb-derived small molecule HMGB1 inhibitors (such as nicotine, glycyrrhizin, tanshinones, and EGCG) are proven protective against lethal infection and ischemic injury. epigallocatechin gallate 204-208 high mobility group box 1 Homo sapiens 20-25 19906009-3 2010 A growing number of HMGB1 inhibitors ranging from neutralizing antibodies, endogenous hormones, to medicinal herb-derived small molecule HMGB1 inhibitors (such as nicotine, glycyrrhizin, tanshinones, and EGCG) are proven protective against lethal infection and ischemic injury. epigallocatechin gallate 204-208 high mobility group box 1 Homo sapiens 137-142 20009166-8 2010 The percentage of GFP-transfected cells and VEGF protein expression level induced by HMGB1/PEG-PEI were 2.6-4.9-fold and 1.4-2.8-fold higher, respectively, than that of a common cationic polymer PEI 25 kDa. Polymers 187-194 high mobility group box 1 Homo sapiens 85-90 19914413-5 2010 In addition to RAGE, proteoglycans and sulfated carbohydrate epitopes of glycolipids and glycoproteins may play a role as cell surface binding sites of HMGB1, affecting migratory behaviour of cells. Carbohydrates 48-60 high mobility group box 1 Homo sapiens 152-157 19651408-6 2010 RESULTS: HMGB1 levels were inversely correlated with peak oxygen consumption (VO(2peak)) (r=-0.449, P<0.001), with left ventricular ejection fraction (LVEF) (r=-0.360, P=0.003), and with HRR (r=-0.387, P<0.001). Oxygen 58-64 high mobility group box 1 Homo sapiens 9-14 19651408-7 2010 In a linear regression analysis adjusted for multiple confounders, we found a significant inverse association between HMGB1 levels and HRR independent of age, gender, body mass index, VO(2peak), slope of increase in ventilation over carbon dioxide output (VE/VCO(2slope)), and presence of diabetes (beta=-0.377, P=0.034). Carbon Dioxide 233-247 high mobility group box 1 Homo sapiens 118-123 19657355-6 2010 Release of High Mobility Group Box-1 protein by MBEH-treated melanocytes and ultrastructural features further confirmed a necrotic death pathway mediated by MBEH. hydroquinone 48-52 high mobility group box 1 Homo sapiens 11-36 20123074-4 2010 Directing of base lesion repair to the long-patch sub-pathway can result in trinucleotide repeat instability suggesting an important role of HMGB1 in modulating genome stability. trinucleotide 76-89 high mobility group box 1 Homo sapiens 141-146 19956840-2 2010 We have previously shown that metabolic stress due to glucose depletion (GD) induces necrosis and HMGB1 release through mitochondrial ROS production in A549 lung adenocarcinoma cells. Glucose 54-61 high mobility group box 1 Homo sapiens 98-103 19956840-2 2010 We have previously shown that metabolic stress due to glucose depletion (GD) induces necrosis and HMGB1 release through mitochondrial ROS production in A549 lung adenocarcinoma cells. ros 134-137 high mobility group box 1 Homo sapiens 98-103 19956840-3 2010 In this study, we examined the effects of hypoxia on GD-induced necrosis and show that hypoxia prevented GD-induced mitochondrial ROS production, HMGB1 release, and necrosis and switched the cell death mode to apoptosis that is dependent on caspase-3 and -9. Gadolinium 105-107 high mobility group box 1 Homo sapiens 146-151 19657355-6 2010 Release of High Mobility Group Box-1 protein by MBEH-treated melanocytes and ultrastructural features further confirmed a necrotic death pathway mediated by MBEH. hydroquinone 157-161 high mobility group box 1 Homo sapiens 11-36 20936104-9 2010 Meanwhile, heme could stimulate cultured microglia to release large amounts of HMGB1 whereas Fe(2+/3+) ions failed to stimulate HMGB1 production from microglia. Heme 11-15 high mobility group box 1 Homo sapiens 79-84 19588230-7 2009 The HMGb1-induced increases of cell proliferation, cell migration, and wound closure were abolished by the MEK inhibitor PD98059. 2-(2-amino-3-methoxyphenyl)-4H-1-benzopyran-4-one 121-128 high mobility group box 1 Homo sapiens 4-9 19943199-7 2009 Targeting HMGB1 using platinum-containing compounds, ethyl pyruvate or glycyrrhizin has also been used to limit autophagy. Platinum 22-30 high mobility group box 1 Homo sapiens 10-15 19943199-7 2009 Targeting HMGB1 using platinum-containing compounds, ethyl pyruvate or glycyrrhizin has also been used to limit autophagy. Glycyrrhizic Acid 71-83 high mobility group box 1 Homo sapiens 10-15 19148928-3 2009 The phorbol myristate acetate-dependent effects of the material chemistry on cell activation and degradation were evaluated by adding reactive oxygen species scavengers, catalase plus superoxide dismutase to MDM and assaying possible markers of MDM activation: esterase activity, acid phosphatase activity, and high molecular weight group box 1 protein (HMGB1). Tetradecanoylphorbol Acetate 4-29 high mobility group box 1 Homo sapiens 354-359 19148928-6 2009 Secretion of HMGB1 from MDM on HDI431 was higher than MDI; however only secretion from MDM on HDI was inhibited by phorbol myristate acetate. Tetradecanoylphorbol Acetate 115-140 high mobility group box 1 Homo sapiens 13-18 19148928-7 2009 In MDM on HDI, catalase plus superoxide dismutase reduced intracellular HMGB1 levels +/- phorbol myristate acetate; whereas, catalase, superoxide dismutase plus phorbol myristate acetate increased intracellular HMGB1 in MDM on MDI, suggesting that esterase and HMGB1 are more specific markers of activation than acid phosphatase. Tetradecanoylphorbol Acetate 161-186 high mobility group box 1 Homo sapiens 211-216 19148928-7 2009 In MDM on HDI, catalase plus superoxide dismutase reduced intracellular HMGB1 levels +/- phorbol myristate acetate; whereas, catalase, superoxide dismutase plus phorbol myristate acetate increased intracellular HMGB1 in MDM on MDI, suggesting that esterase and HMGB1 are more specific markers of activation than acid phosphatase. Tetradecanoylphorbol Acetate 161-186 high mobility group box 1 Homo sapiens 211-216 19265175-0 2009 Quercetin prevents LPS-induced high-mobility group box 1 release and proinflammatory function. Quercetin 0-9 high mobility group box 1 Homo sapiens 31-56 19265175-5 2009 Quercetin treatment reduced circulating levels of HMGB1 in animals with established endotoxemia. Quercetin 0-9 high mobility group box 1 Homo sapiens 50-55 19265175-6 2009 In macrophage cultures, quercetin inhibited release as well as the cytokine activities of HMGB1, including limiting the activation of mitogen-activated protein kinase and NF-kappaB, two signaling pathways that are critical for HMGB1-induced subsequent cytokine release. Quercetin 24-33 high mobility group box 1 Homo sapiens 90-95 19265175-6 2009 In macrophage cultures, quercetin inhibited release as well as the cytokine activities of HMGB1, including limiting the activation of mitogen-activated protein kinase and NF-kappaB, two signaling pathways that are critical for HMGB1-induced subsequent cytokine release. Quercetin 24-33 high mobility group box 1 Homo sapiens 227-232 19265175-7 2009 Quercetin and autophagic inhibitor, wortmannin, inhibited LPS-induced type-II microtubule-associated protein 1A/1B-light chain 3 production and aggregation, as well as HMGB1 translocation and release, suggesting a potential association between autophagy and HMGB1 release. Quercetin 0-9 high mobility group box 1 Homo sapiens 168-173 19265175-7 2009 Quercetin and autophagic inhibitor, wortmannin, inhibited LPS-induced type-II microtubule-associated protein 1A/1B-light chain 3 production and aggregation, as well as HMGB1 translocation and release, suggesting a potential association between autophagy and HMGB1 release. Quercetin 0-9 high mobility group box 1 Homo sapiens 258-263 19265175-7 2009 Quercetin and autophagic inhibitor, wortmannin, inhibited LPS-induced type-II microtubule-associated protein 1A/1B-light chain 3 production and aggregation, as well as HMGB1 translocation and release, suggesting a potential association between autophagy and HMGB1 release. Wortmannin 36-46 high mobility group box 1 Homo sapiens 168-173 19265175-7 2009 Quercetin and autophagic inhibitor, wortmannin, inhibited LPS-induced type-II microtubule-associated protein 1A/1B-light chain 3 production and aggregation, as well as HMGB1 translocation and release, suggesting a potential association between autophagy and HMGB1 release. Wortmannin 36-46 high mobility group box 1 Homo sapiens 258-263 19265175-8 2009 Quercetin delivery, a strategy to pharmacologically inhibit HMGB1 release that is effective at clinically achievable concentrations, now warrants further evaluation in sepsis and other systemic inflammatory disorders. Quercetin 0-9 high mobility group box 1 Homo sapiens 60-65 19295481-0 2009 Edaravone, a novel free radical scavenger, reduces high-mobility group box 1 and prolongs survival in a neonatal sepsis model. Edaravone 0-9 high mobility group box 1 Homo sapiens 51-76 19295481-8 2009 In conclusion, edaravone suppressed free radicals, delayed the TNF-alpha surge, and prevented HMGB1 elevation, thereby maintaining MAP and prolonging survival time in a neonatal sepsis CLP model. Edaravone 15-24 high mobility group box 1 Homo sapiens 94-99 20193282-13 2009 MTT indicated the growth of HMGB1-siRNA group was significantly inhibited than other two groups. monooxyethylene trimethylolpropane tristearate 0-3 high mobility group box 1 Homo sapiens 28-33 19786828-3 2009 In addition to demonstrating that cisplatin prevents liver damage associated with liver I/R by sequestering HMGB1 inside the nucleus of ischemic cells, cisplatin also alters cell survival signaling through autophagy. Cisplatin 34-43 high mobility group box 1 Homo sapiens 108-113 19786011-0 2009 Histidine-rich glycoprotein inhibited high mobility group box 1 in complex with heparin-induced angiogenesis in matrigel plug assay. Heparin 80-87 high mobility group box 1 Homo sapiens 38-63 19786011-3 2009 Additionally, we reported that high mobility group box 1 (HMGB1) in complex with heparin induces angiogenesis; therefore, we examined the effect of HRG on heparin-dependent HMGB1-induced angiogenesis in the present study. Heparin 81-88 high mobility group box 1 Homo sapiens 58-63 19786011-3 2009 Additionally, we reported that high mobility group box 1 (HMGB1) in complex with heparin induces angiogenesis; therefore, we examined the effect of HRG on heparin-dependent HMGB1-induced angiogenesis in the present study. Heparin 155-162 high mobility group box 1 Homo sapiens 58-63 19786011-3 2009 Additionally, we reported that high mobility group box 1 (HMGB1) in complex with heparin induces angiogenesis; therefore, we examined the effect of HRG on heparin-dependent HMGB1-induced angiogenesis in the present study. Heparin 155-162 high mobility group box 1 Homo sapiens 173-178 19786011-4 2009 HRG completely inhibited angiogenesis induced by HMGB1 in complex with heparin. Heparin 71-78 high mobility group box 1 Homo sapiens 49-54 19786011-5 2009 HRG inhibited the diffusion of a complex of HMGB1 with heparin from matrigel into surrounding tissue. Heparin 55-62 high mobility group box 1 Homo sapiens 44-49 19786011-6 2009 HRG also competed with HMGB1 for heparin binding in vitro. Heparin 33-40 high mobility group box 1 Homo sapiens 23-28 19604520-0 2009 Positive association of serum levels of advanced glycation end products and high mobility group box-1 with asymmetric dimethylarginine in nondiabetic chronic kidney disease patients. dimethylarginine 118-134 high mobility group box 1 Homo sapiens 76-101 20101991-0 2009 [Analysis of the impact of heparin on the affinity of high mobility group box-1 protein and the receptor of advanced glycation end products by surface plasmon resonance technology]. Heparin 27-34 high mobility group box 1 Homo sapiens 54-79 20101991-1 2009 To investigate the affinity constants of heparin with high mobility group protein 1(HMGB1) and HMGB1 with the receptor of advanced glycation end products (RAGE) and to analyze the impact of heparin on the affinity of HMGB1 and RAGE, the standard BIAcore amine coupling chemistry protocol using EDC and NHS was employed for immobilizing. Heparin 41-48 high mobility group box 1 Homo sapiens 84-89 20101991-3 2009 Binding analysis was used to investigate the impact of heparin on the affinity of HMGB1 and RAGE. Heparin 55-62 high mobility group box 1 Homo sapiens 82-87 20101991-7 2009 The highest concentration of 10 000 u x L(-1) of heparin in this experiment did not reach the saturation with HMGB1. Heparin 49-56 high mobility group box 1 Homo sapiens 110-115 20101991-8 2009 After 50 mg x L(-1) of HMGB1 was mixed with heparin of 50, 100, 1 000, 10 000 u x L(-1), the combining amount of HMGB1 and RAGE declined from 100 to 50 RU. Heparin 44-51 high mobility group box 1 Homo sapiens 23-28 20101991-8 2009 After 50 mg x L(-1) of HMGB1 was mixed with heparin of 50, 100, 1 000, 10 000 u x L(-1), the combining amount of HMGB1 and RAGE declined from 100 to 50 RU. Heparin 44-51 high mobility group box 1 Homo sapiens 113-118 20101991-10 2009 It was concluded that heparin can combine with HMGB1 and affect the affinity of HMGB1/RAGE. Heparin 22-29 high mobility group box 1 Homo sapiens 47-52 20101991-10 2009 It was concluded that heparin can combine with HMGB1 and affect the affinity of HMGB1/RAGE. Heparin 22-29 high mobility group box 1 Homo sapiens 80-85 19604520-11 2009 The present study demonstrated that AGE and sRAGE levels were correlated with each other and that AGEs and HMGB-1 were independently associated with ADMA in nondiabetic CKD patients. N,N-dimethylarginine 149-153 high mobility group box 1 Homo sapiens 107-113 19751561-2 2009 METHODS: HepG2 cells were cultured, and purified HMGB1 protein was prepared by chromatography on Ni(2+)-NTA Sepharose column under natural conditions with recombinant expression plasmid pET14b-HMGB1. nickel nitrilotriacetic acid 97-107 high mobility group box 1 Homo sapiens 49-54 19751561-2 2009 METHODS: HepG2 cells were cultured, and purified HMGB1 protein was prepared by chromatography on Ni(2+)-NTA Sepharose column under natural conditions with recombinant expression plasmid pET14b-HMGB1. Sepharose 108-117 high mobility group box 1 Homo sapiens 49-54 19545550-5 2009 RESULTS: HMGB1 level in USAP group was higher than that in control and SAP group. usap 24-28 high mobility group box 1 Homo sapiens 9-14 19727205-9 2009 These results suggested that heparin"s anti-inflammatory effects can be partly explained by its blocking of the interaction between HMGB1 or S100A12 and RAGE. Heparin 29-36 high mobility group box 1 Homo sapiens 132-137 19387923-0 2009 Carbon dioxide, hypoxia and low pH lead to overexpression of c-myc and HMGB-1 oncogenes in neuroblastoma cells. Carbon Dioxide 0-14 high mobility group box 1 Homo sapiens 71-77 19387923-3 2009 We studied whether exposure to CO (2), hypoxia or acidosis affects the expression of C-MYC and HMGB-1 in neuroblastoma cells. Carbon Dioxide 31-37 high mobility group box 1 Homo sapiens 95-101 19387923-10 2009 CONCLUSIONS: Exposures mimicking conditions of CO (2) pneumoperitoneum lead to significant overexpression of C-MYC and HMGB-1 in neuroblastoma cells with decreased apoptosis. Carbon Dioxide 47-53 high mobility group box 1 Homo sapiens 119-125 19506549-3 2009 To simulate the activated state of HMGB1, we mutated serine residues of nuclear localization signals (NLSs) to glutamic acid and performed transfection assays. Serine 53-59 high mobility group box 1 Homo sapiens 35-40 19506549-6 2009 Concurrent mutations at six serine residues (35, 39, 42, 46, 53, and 181) to glutamic acid induced the nuclear to cytoplasmic transport of HMGB1, which was detected in the culture medium. Serine 28-34 high mobility group box 1 Homo sapiens 139-144 19506549-6 2009 Concurrent mutations at six serine residues (35, 39, 42, 46, 53, and 181) to glutamic acid induced the nuclear to cytoplasmic transport of HMGB1, which was detected in the culture medium. Glutamic Acid 77-90 high mobility group box 1 Homo sapiens 139-144 19379716-7 2009 In light of recent observations as well as the good safety profile of minocycline in humans, we propose that minocycline might play a potent neuroprotective role through the inhibition of HMGB1-induced neuronal cell death in cerebral infarction. Minocycline 109-120 high mobility group box 1 Homo sapiens 188-193 19726280-8 2009 This synergistic effect was significantly inhibited by pretreatment with MAPK signaling kinases inhibitors, especially the p38 MAP kinase inhibitor SB203580, and the cocktail of MAP kinase inhibitors almost totally blocked the expression of these chemokines in HUVECs treated with HMGB1 and LPS. SB 203580 148-156 high mobility group box 1 Homo sapiens 281-286 19506468-11 2009 These data suggest that HMGB1 and oxidative stress play a role in the pathogenesis of ARDS and that PMX-F treatment may ameliorate increased blood HMGB1 and urinary 8-OHdG levels in patients with ARDS. pmx-f 100-105 high mobility group box 1 Homo sapiens 147-152 19380828-10 2009 HMGB1 secretion was also induced by the calcium ionophore A23187 and inhibited by the Ca(2+) chelators BAPTA-AM and EGTA. Calcium 40-47 high mobility group box 1 Homo sapiens 0-5 19401149-0 2009 Native HMGB1 protein inhibits repair of cisplatin-damaged nucleosomes in vitro. Cisplatin 40-49 high mobility group box 1 Homo sapiens 7-12 19401149-1 2009 The high mobility group box (HMGB) 1 protein, one of the most abundant nuclear non-histone proteins has been known for its inhibitory effect on repair of DNA damaged by the antitumor drug cisplatin. Cisplatin 188-197 high mobility group box 1 Homo sapiens 4-36 19401149-2 2009 Here, we report the first results that link HMGB1 to repair of cisplatin-treated DNA at nucleosome level. Cisplatin 63-72 high mobility group box 1 Homo sapiens 44-49 19401149-10 2009 We show that both the repair of cisplatin-damaged nucleosomes and its inhibition by HMGB1 are nucleosome position-dependent events which are accomplished via the acidic tail and modulated by acetylation. Cisplatin 32-41 high mobility group box 1 Homo sapiens 84-89 18670905-2 2009 Here, we have investigated how the post-synthetic acetylation of HMGB1 affects its interaction with negatively supercoiled DNA by employing monoacetylated at Lys2 protein, isolated from butyrate-treated cells. Butyrates 186-194 high mobility group box 1 Homo sapiens 65-70 19442462-0 2009 High mobility group box 1 protein enhances polyethylenimine mediated gene delivery in vitro. Polyethyleneimine 43-59 high mobility group box 1 Homo sapiens 0-25 19442462-7 2009 In MTT assay the results of cell viability suggested lower cytotoxicity for HMGB1/PEI combined carriers. monooxyethylene trimethylolpropane tristearate 3-6 high mobility group box 1 Homo sapiens 76-81 19380828-0 2009 HMGB1 is phosphorylated by classical protein kinase C and is secreted by a calcium-dependent mechanism. Calcium 75-82 high mobility group box 1 Homo sapiens 0-5 19380828-4 2009 In this study, we show that HMGB1 is phosphorylated by the classical protein kinase C (cPKC) and is secreted by a calcium-dependent mechanism. Calcium 114-121 high mobility group box 1 Homo sapiens 28-33 19570774-8 2009 We also showed the killing efficacy of Ad-TK+GCV in human GBM cell lines and GBM primary cultures, which also elicited release of HMGB1. Ganciclovir 45-48 high mobility group box 1 Homo sapiens 130-135 19811284-8 2009 Moreover, tagging of oxidized cysteine residues by a maleimide moiety linked to polyethylene glycol showed that HMGB1 passively released from primary and secondary necrotic cells was predominantly oxidized. Cysteine 30-38 high mobility group box 1 Homo sapiens 112-117 19811284-8 2009 Moreover, tagging of oxidized cysteine residues by a maleimide moiety linked to polyethylene glycol showed that HMGB1 passively released from primary and secondary necrotic cells was predominantly oxidized. maleimide 53-62 high mobility group box 1 Homo sapiens 112-117 19811284-8 2009 Moreover, tagging of oxidized cysteine residues by a maleimide moiety linked to polyethylene glycol showed that HMGB1 passively released from primary and secondary necrotic cells was predominantly oxidized. Polyethylene Glycols 80-99 high mobility group box 1 Homo sapiens 112-117 19811284-10 2009 In conclusion, HMGB1 undergoes reversible oxidative modifications at cysteine residues during cell death, which may modulate its biological properties. Cysteine 69-77 high mobility group box 1 Homo sapiens 15-20 19380828-5 2009 For this study, RAW264.7 cells and human peripheral blood monocytes were treated with PI3K inhibitors wortmannin, LY294002, and ZSTK474, resulting in inhibition of LPS-stimulated HMGB1 secretion, whereas inhibitors of NF-kappaB and MAPKs p38 and ERK showed no inhibition. Wortmannin 102-112 high mobility group box 1 Homo sapiens 179-184 19380828-5 2009 For this study, RAW264.7 cells and human peripheral blood monocytes were treated with PI3K inhibitors wortmannin, LY294002, and ZSTK474, resulting in inhibition of LPS-stimulated HMGB1 secretion, whereas inhibitors of NF-kappaB and MAPKs p38 and ERK showed no inhibition. 2-(4-morpholinyl)-8-phenyl-4H-1-benzopyran-4-one 114-122 high mobility group box 1 Homo sapiens 179-184 19380828-10 2009 HMGB1 secretion was also induced by the calcium ionophore A23187 and inhibited by the Ca(2+) chelators BAPTA-AM and EGTA. Calcimycin 58-64 high mobility group box 1 Homo sapiens 0-5 19380828-10 2009 HMGB1 secretion was also induced by the calcium ionophore A23187 and inhibited by the Ca(2+) chelators BAPTA-AM and EGTA. 1,2-bis(2-aminophenoxy)ethane N,N,N',N'-tetraacetic acid acetoxymethyl ester 103-111 high mobility group box 1 Homo sapiens 0-5 19380828-10 2009 HMGB1 secretion was also induced by the calcium ionophore A23187 and inhibited by the Ca(2+) chelators BAPTA-AM and EGTA. Egtazic Acid 116-120 high mobility group box 1 Homo sapiens 0-5 19317908-3 2009 The high mobility group proteins HMGB1 and HMGB2 (HMGB1/2) are highly abundant architectural DNA binding proteins known to promote RAG-mediated synapsis and cleavage of consensus recombination signals in vitro by facilitating RSS binding and bending by the RAG1/2 complex. RSS 226-229 high mobility group box 1 Homo sapiens 33-38 19372559-2 2009 High-mobility group protein 1 (HMGB1) is a DNA damage sensor responsive to the incorporation of nonnatural nucleosides into DNA; several nuclear and cytosolic proteins are functionally integrated with HMGB1 in the context of DNA damage response. Nucleosides 107-118 high mobility group box 1 Homo sapiens 31-36 19372559-2 2009 High-mobility group protein 1 (HMGB1) is a DNA damage sensor responsive to the incorporation of nonnatural nucleosides into DNA; several nuclear and cytosolic proteins are functionally integrated with HMGB1 in the context of DNA damage response. Nucleosides 107-118 high mobility group box 1 Homo sapiens 201-206 19372559-4 2009 Using the cell proliferation assay, we found that knockdown of HMGB1-associated proteins resulted in 8-fold to 50-fold decreased chemosensitivity of A549 cells to cytarabine. Cytarabine 163-173 high mobility group box 1 Homo sapiens 63-68 19372559-6 2009 Our results dissect the roles of HMGB1-associated proteins in DNA damage response: HMGB1 and HMGB2 facilitate p53 phosphorylation after exposure to genotoxic stress, and PDIA3 has been found essential for H2AX phosphorylation (no gamma-H2AX accumulated after 24-72 hours of incubation with 1 micromol/L of cytarabine in PDIA3 knockdown cells). Cytarabine 306-316 high mobility group box 1 Homo sapiens 33-38 18715145-4 2009 Forced cytoplasmic overexpression of APE1 profoundly attenuated the upregulation of HMGB1-mediated reactive oxygen species generation, cytokine secretion, and cyclooxygenase-2 expression by primary monocytes and macrophage-like THP-1 cell lines. Reactive Oxygen Species 99-122 high mobility group box 1 Homo sapiens 84-89 19166815-2 2009 We created mutants at the target sites (lysines 2 and 81) in the tailless HMGB1 modified by the histone acetyltransferase CBP. Lysine 40-47 high mobility group box 1 Homo sapiens 74-79 19166815-7 2009 In the case of HMGB1DeltaC modified at Lys 2 and Lys 81 prior to PKC treatment background phosphorylation is detected. Lysine 39-42 high mobility group box 1 Homo sapiens 15-20 19166815-7 2009 In the case of HMGB1DeltaC modified at Lys 2 and Lys 81 prior to PKC treatment background phosphorylation is detected. Lysine 49-52 high mobility group box 1 Homo sapiens 15-20 19109190-7 2009 We also show that beta-adrenergic receptor activation by catecholamine of macrophages mediates the HS/R-induced release of HMGB1. Catecholamines 57-70 high mobility group box 1 Homo sapiens 123-128 18515078-5 2008 Results showed that the HMGB1 Mut (102-105), one of the HMGB1 mutants, in which amino acids 102-105 (FFLF) were replaced with two Glys, significantly decreased the full-length HMGB1 protein induced TNF-alpha release in human monocyte cultures. glys 130-134 high mobility group box 1 Homo sapiens 24-29 18949384-0 2008 Induction of high mobility group box 1 release from serotonin-stimulated human umbilical vein endothelial cells. Serotonin 52-61 high mobility group box 1 Homo sapiens 13-38 18949384-5 2008 Therefore, we examined whether serotonin (5-HT), a key factor involved in the development of atherosclerosis, induced HMGB1 release in human umbilical vein endothelial cells (HUVECs). Serotonin 31-40 high mobility group box 1 Homo sapiens 118-123 18949384-7 2008 The p38MAPK inhibitor SB203580 and the 5-HT1B antagonist GR55526 markedly inhibited HMGB1 release from 5-HT-stimulated HUVECs. SB 203580 22-30 high mobility group box 1 Homo sapiens 84-89 18949384-7 2008 The p38MAPK inhibitor SB203580 and the 5-HT1B antagonist GR55526 markedly inhibited HMGB1 release from 5-HT-stimulated HUVECs. gr55526 57-64 high mobility group box 1 Homo sapiens 84-89 18949384-11 2008 The PI3-kinase inhibitor LY294002 significantly inhibited VEGF production in HMGB1-stimulated macrophages, while other kinase inhibitors did not. 2-(4-morpholinyl)-8-phenyl-4H-1-benzopyran-4-one 25-33 high mobility group box 1 Homo sapiens 77-82 18799030-0 2008 [Role of high mobility group box 1 in adriamycin-induced apoptosis in leukemia K562 cells]. Doxorubicin 38-48 high mobility group box 1 Homo sapiens 9-34 18799030-1 2008 BACKGROUND & OBJECTIVE: High mobility group box l (HMGB1), a nuclear DNA-binding protein, stabilizes the structure and function of chromatin, regulates gene transcription. Adenosine Monophosphate 12-15 high mobility group box 1 Homo sapiens 55-60 18799030-3 2008 This study was to explore the role of HMGB1 in adriamycin (ADM)-induced apoptosis in leukemia K562 cells. Doxorubicin 47-57 high mobility group box 1 Homo sapiens 38-43 18799030-3 2008 This study was to explore the role of HMGB1 in adriamycin (ADM)-induced apoptosis in leukemia K562 cells. Doxorubicin 59-62 high mobility group box 1 Homo sapiens 38-43 18996119-2 2008 We have carried out quantitative investigations of the redox chemistry involving Cys22 and Cys44 of the HMGB1 A-domain, which form an intramolecular disulfide bond. Disulfides 149-158 high mobility group box 1 Homo sapiens 104-109 19114373-0 2008 [Effect of progesterone on high mobility group Box-1 protein-induced interleukin-6 release by human umbilic vein endothelial cells]. Progesterone 11-23 high mobility group box 1 Homo sapiens 27-52 19114373-2 2008 METHOD: The recombinant expression plasmid pET14b-HMGB1 was constructed and transformed into competent E.coli BL21 cells to obtain HMGB1 protein, which was purified with chromatography on Ni-NTA Sepharose column. ni-nta 188-194 high mobility group box 1 Homo sapiens 131-136 19114373-2 2008 METHOD: The recombinant expression plasmid pET14b-HMGB1 was constructed and transformed into competent E.coli BL21 cells to obtain HMGB1 protein, which was purified with chromatography on Ni-NTA Sepharose column. Sepharose 195-204 high mobility group box 1 Homo sapiens 131-136 19114373-5 2008 RESULTS: The IL-6 levels in HUVEC culture medium was slightly decreased after treatment with low-concentration HMGB1 but increased obviously following treatment with high-concentration HMGB1, and these effects could be dose-dependently inhibited by progesterone. Progesterone 249-261 high mobility group box 1 Homo sapiens 111-116 19114373-5 2008 RESULTS: The IL-6 levels in HUVEC culture medium was slightly decreased after treatment with low-concentration HMGB1 but increased obviously following treatment with high-concentration HMGB1, and these effects could be dose-dependently inhibited by progesterone. Progesterone 249-261 high mobility group box 1 Homo sapiens 185-190 19114373-7 2008 CONCLUSIONS: Progesterone can dose-dependently inhibit HMGB1-induced IL-6 release from HUVECs, suggesting the protective role of progesterone in endotoxemia. Progesterone 129-141 high mobility group box 1 Homo sapiens 55-60 18515078-5 2008 Results showed that the HMGB1 Mut (102-105), one of the HMGB1 mutants, in which amino acids 102-105 (FFLF) were replaced with two Glys, significantly decreased the full-length HMGB1 protein induced TNF-alpha release in human monocyte cultures. glys 130-134 high mobility group box 1 Homo sapiens 56-61 18515078-5 2008 Results showed that the HMGB1 Mut (102-105), one of the HMGB1 mutants, in which amino acids 102-105 (FFLF) were replaced with two Glys, significantly decreased the full-length HMGB1 protein induced TNF-alpha release in human monocyte cultures. glys 130-134 high mobility group box 1 Homo sapiens 56-61 18682735-5 2008 DNA-protein complexes formed between HMGB1 and radiolabeled hcDNA are analyzed by electrophoresis on nondenaturing polyacrylamide gels using the band-shift assay method. polyacrylamide 115-129 high mobility group box 1 Homo sapiens 37-42 18682735-6 2008 In addition, using a simple and fast protocol to purify HMGB1 on the basis of its solubility in perchloric acid allowed us to increase the sensitivity by suppressing any nonspecific background. Perchloric Acid 96-111 high mobility group box 1 Homo sapiens 56-61 19112920-0 2008 [Enhancive effect of HMGB1 gene silence on adriamycin-induced apoptosis in K562/A02 drug resistance leukemia cells]. Doxorubicin 43-53 high mobility group box 1 Homo sapiens 21-26 18715162-5 2008 MSCs exposed to HMGB1 were negative for CD31, CD45, CD80, and HLA-DR, and displayed equal levels of CD73, CD166, and HLA-ABC compared with their counterparts, but HMGB1 profoundly suppressed MSC proliferation in a dose-dependent manner as evaluated by carboxyfluorescein diacetate succinmidyl ester dye dilution assay. carboxyfluorescein diacetate succinmidyl ester 252-298 high mobility group box 1 Homo sapiens 16-21 18953944-1 2008 OBJECTIVE: To investigate the release and intracellular localization of high mobility group box chromosomal protein 1 (HMGB1) in the peripheral blood monocytes of rheumatoid arthritis (RA) patients and the inhibitive effect of thalidomide. Thalidomide 227-238 high mobility group box 1 Homo sapiens 119-124 18631454-4 2008 ROS oxidized the potential danger signal high-mobility group box-1 protein (HMGB1) released from dying cells and thereby neutralized its stimulatory activity. Reactive Oxygen Species 0-3 high mobility group box 1 Homo sapiens 41-66 18631454-4 2008 ROS oxidized the potential danger signal high-mobility group box-1 protein (HMGB1) released from dying cells and thereby neutralized its stimulatory activity. Reactive Oxygen Species 0-3 high mobility group box 1 Homo sapiens 76-81 18425372-5 2008 While GD induced mostly necrotic death in A549 cells, salicylate suppresssed GD-induced necrosis and HMGB1 release. Salicylates 54-64 high mobility group box 1 Homo sapiens 101-106 18953944-10 2008 Thalidomide (40 microg/ml) could inhibit the release of HMGB1 in the monocytes from RA patients stimulated with TNFalpha (P < 0.05). Thalidomide 0-11 high mobility group box 1 Homo sapiens 56-61 18953944-12 2008 Treatment with TNFalpha (100 ng/ml) for 24 hours resulted in a cytoplasmic translocation of HMGB1, which was inhibited significantly by thalidomide. Thalidomide 136-147 high mobility group box 1 Homo sapiens 92-97 18483013-0 2008 Estradiol 17-beta and progesterone modulate inducible nitric oxide synthase and high mobility group box 1 expression in human endometrium. Estradiol 0-17 high mobility group box 1 Homo sapiens 80-105 18483013-0 2008 Estradiol 17-beta and progesterone modulate inducible nitric oxide synthase and high mobility group box 1 expression in human endometrium. Progesterone 22-34 high mobility group box 1 Homo sapiens 80-105 18483013-8 2008 HMGB1 is expressed by human endometrium, and its expression is increased by E2 and decreased by progesterone. Progesterone 96-108 high mobility group box 1 Homo sapiens 0-5 18483013-9 2008 Incubation with sodium nitroprusside results in a reduction in HMGB1 expression. Nitroprusside 16-36 high mobility group box 1 Homo sapiens 63-68 18483013-11 2008 HMGB1 is expressed in the human endometrium, and its expression is modulated by E2, progesterone, and nitric oxide. Progesterone 84-96 high mobility group box 1 Homo sapiens 0-5 18483013-11 2008 HMGB1 is expressed in the human endometrium, and its expression is modulated by E2, progesterone, and nitric oxide. Nitric Oxide 102-114 high mobility group box 1 Homo sapiens 0-5 18241198-0 2008 A critical role in structure-specific DNA binding for the acetylatable lysine residues in HMGB1. Lysine 71-77 high mobility group box 1 Homo sapiens 90-95 18241198-1 2008 The structure-specific DNA-binding protein HMGB1 (high-mobility group protein B1) which comprises two tandem HMG boxes (A and B) and an acidic C-terminal tail, is acetylated in vivo at Lys(2) and Lys(11) in the A box. Lysine 185-188 high mobility group box 1 Homo sapiens 43-48 18241198-1 2008 The structure-specific DNA-binding protein HMGB1 (high-mobility group protein B1) which comprises two tandem HMG boxes (A and B) and an acidic C-terminal tail, is acetylated in vivo at Lys(2) and Lys(11) in the A box. Lysine 185-188 high mobility group box 1 Homo sapiens 50-80 18241198-1 2008 The structure-specific DNA-binding protein HMGB1 (high-mobility group protein B1) which comprises two tandem HMG boxes (A and B) and an acidic C-terminal tail, is acetylated in vivo at Lys(2) and Lys(11) in the A box. Lysine 196-199 high mobility group box 1 Homo sapiens 43-48 18241198-1 2008 The structure-specific DNA-binding protein HMGB1 (high-mobility group protein B1) which comprises two tandem HMG boxes (A and B) and an acidic C-terminal tail, is acetylated in vivo at Lys(2) and Lys(11) in the A box. Lysine 196-199 high mobility group box 1 Homo sapiens 50-80 18241198-8 2008 We conclude that Lys(2) and Lys(11) are critical for binding of the isolated A domain and HMGB1 to distorted DNA substrates. Lysine 17-20 high mobility group box 1 Homo sapiens 90-95 18241198-8 2008 We conclude that Lys(2) and Lys(11) are critical for binding of the isolated A domain and HMGB1 to distorted DNA substrates. Lysine 28-31 high mobility group box 1 Homo sapiens 90-95 18953944-13 2008 CONCLUSION: TNFalpha induces the release and cytoplasmic translocation of HMGB1 in the peripheral blood monocytes of RA patients and thalidomide inhibits the release and translocation of HMGB1. Thalidomide 133-144 high mobility group box 1 Homo sapiens 74-79 18160415-4 2008 In vitro glutathione S-transferase pull-down and in vivo co-immunoprecipitation studies confirmed an interaction between HMGB1 and HNF1alpha. Glutathione 9-20 high mobility group box 1 Homo sapiens 121-126 18953944-13 2008 CONCLUSION: TNFalpha induces the release and cytoplasmic translocation of HMGB1 in the peripheral blood monocytes of RA patients and thalidomide inhibits the release and translocation of HMGB1. Thalidomide 133-144 high mobility group box 1 Homo sapiens 187-192 18844116-3 2008 The eukaryotic expression vector carrying human Pgenesil-1/HMGB1 siRNA was constructed and transfected into T3B cells by Lipofectamine 2000 and the positive clones was screened by G418. Lipofectamine 121-139 high mobility group box 1 Homo sapiens 59-64 18844116-10 2008 The HMGB1 expression of the T3B cells transfected with Pgenesil-1/HMGB1 siRNA was significantly lower than those of the other 2 groups (both P <0.05). pgenesil-1 55-65 high mobility group box 1 Homo sapiens 4-9 18844116-10 2008 The HMGB1 expression of the T3B cells transfected with Pgenesil-1/HMGB1 siRNA was significantly lower than those of the other 2 groups (both P <0.05). pgenesil-1 55-65 high mobility group box 1 Homo sapiens 66-71 18380906-5 2008 RESULTS: We show here that single HMG-box domains of HMGB1 stimulate RAG-mediated RSS cleavage in a concentration-dependent manner in the presence of Mn2+, but not Mg2+. RSS 82-85 high mobility group box 1 Homo sapiens 53-58 18353726-4 2008 It has recently been revealed that the translocation of calreticulin to the plasma membrane in tumor cells and the release of high-mobility-group box 1 (HMGB1) by tumor cells are two key post-transcriptional events required for the immunogenicity of anthracyclines. Anthracyclines 250-264 high mobility group box 1 Homo sapiens 126-151 18353726-4 2008 It has recently been revealed that the translocation of calreticulin to the plasma membrane in tumor cells and the release of high-mobility-group box 1 (HMGB1) by tumor cells are two key post-transcriptional events required for the immunogenicity of anthracyclines. Anthracyclines 250-264 high mobility group box 1 Homo sapiens 153-158 18223193-5 2008 The immediate negative inotropic effects of HMGB1 on cell contractility and calcium homeostasis were partially reversible upon washout of HMGB1. Calcium 76-83 high mobility group box 1 Homo sapiens 44-49 18223193-8 2008 These studies show for the first time that HMGB1 impairs sarcomere shortening by decreasing calcium availability in cardiac myocytes through modulating membrane calcium influx and suggest that HMGB1 maybe acts as a novel myocardial depressant factor during cardiac injury. Calcium 92-99 high mobility group box 1 Homo sapiens 43-48 18223193-8 2008 These studies show for the first time that HMGB1 impairs sarcomere shortening by decreasing calcium availability in cardiac myocytes through modulating membrane calcium influx and suggest that HMGB1 maybe acts as a novel myocardial depressant factor during cardiac injury. Calcium 92-99 high mobility group box 1 Homo sapiens 193-198 18223193-8 2008 These studies show for the first time that HMGB1 impairs sarcomere shortening by decreasing calcium availability in cardiac myocytes through modulating membrane calcium influx and suggest that HMGB1 maybe acts as a novel myocardial depressant factor during cardiac injury. Calcium 161-168 high mobility group box 1 Homo sapiens 43-48 18223193-8 2008 These studies show for the first time that HMGB1 impairs sarcomere shortening by decreasing calcium availability in cardiac myocytes through modulating membrane calcium influx and suggest that HMGB1 maybe acts as a novel myocardial depressant factor during cardiac injury. Calcium 161-168 high mobility group box 1 Homo sapiens 193-198 18380906-5 2008 RESULTS: We show here that single HMG-box domains of HMGB1 stimulate RAG-mediated RSS cleavage in a concentration-dependent manner in the presence of Mn2+, but not Mg2+. Manganese(2+) 150-154 high mobility group box 1 Homo sapiens 53-58 18380906-5 2008 RESULTS: We show here that single HMG-box domains of HMGB1 stimulate RAG-mediated RSS cleavage in a concentration-dependent manner in the presence of Mn2+, but not Mg2+. magnesium ion 164-168 high mobility group box 1 Homo sapiens 53-58 18380906-7 2008 Furthermore, we show that mutant forms of HMGB1 which otherwise fail to stimulate RAG-mediated RSS cleavage in Mg2+ can be substantially rescued when Mg2+ is replaced with Mn2+. RSS 95-98 high mobility group box 1 Homo sapiens 42-47 18380906-7 2008 Furthermore, we show that mutant forms of HMGB1 which otherwise fail to stimulate RAG-mediated RSS cleavage in Mg2+ can be substantially rescued when Mg2+ is replaced with Mn2+. magnesium ion 111-115 high mobility group box 1 Homo sapiens 42-47 18380906-7 2008 Furthermore, we show that mutant forms of HMGB1 which otherwise fail to stimulate RAG-mediated RSS cleavage in Mg2+ can be substantially rescued when Mg2+ is replaced with Mn2+. magnesium ion 150-154 high mobility group box 1 Homo sapiens 42-47 18380906-7 2008 Furthermore, we show that mutant forms of HMGB1 which otherwise fail to stimulate RAG-mediated RSS cleavage in Mg2+ can be substantially rescued when Mg2+ is replaced with Mn2+. Manganese(2+) 172-176 high mobility group box 1 Homo sapiens 42-47 18380906-9 2008 The observation that single HMG-box domains can promote RAG-mediated 23-RSS cleavage in Mg2+ in the presence, but not absence, of partner RSS suggests that synaptic complex assembly in vitro is associated with conformational changes that alter how the RAG and/or HMGB1 proteins bind and bend DNA in a manner that functionally replaces the role of one of the HMG-box domains in RAG-HMGB1 complexes assembled on a single RSS. magnesium ion 88-92 high mobility group box 1 Homo sapiens 263-268 18380906-9 2008 The observation that single HMG-box domains can promote RAG-mediated 23-RSS cleavage in Mg2+ in the presence, but not absence, of partner RSS suggests that synaptic complex assembly in vitro is associated with conformational changes that alter how the RAG and/or HMGB1 proteins bind and bend DNA in a manner that functionally replaces the role of one of the HMG-box domains in RAG-HMGB1 complexes assembled on a single RSS. magnesium ion 88-92 high mobility group box 1 Homo sapiens 381-386 18354232-4 2008 We used ELISA and surface plasmon resonance to show that HMGB1 binds LPS in a concentration-dependent manner and that the binding is stronger to lipid A moiety than to the polysaccharide moiety of LPS. Lipid A 145-152 high mobility group box 1 Homo sapiens 57-62 18354232-4 2008 We used ELISA and surface plasmon resonance to show that HMGB1 binds LPS in a concentration-dependent manner and that the binding is stronger to lipid A moiety than to the polysaccharide moiety of LPS. Polysaccharides 172-186 high mobility group box 1 Homo sapiens 57-62 18557992-4 2008 In the previous issue of Arthritis Research & Therapy, Sundberg and colleagues address, for the first time in a prospective cohort study, whether HMGB1 expression is dependent upon tumor necrosis factor activity in patients with RA. Adenosine Monophosphate 45-48 high mobility group box 1 Homo sapiens 150-155 18090368-10 2008 There was a significant correlation between CSF HMGB1 levels and CSF white blood cell counts and glucose levels in patients with bacterial meningitis. Glucose 97-104 high mobility group box 1 Homo sapiens 48-53 17979839-7 2007 A TLR4 polymorphism that affects the binding of HMGB1 to TLR4 predicts early relapse after anthracycline-based chemotherapy in breast cancer patients. Anthracyclines 91-104 high mobility group box 1 Homo sapiens 48-53 17913975-0 2008 Pivotal advance: inhibition of HMGB1 nuclear translocation as a mechanism for the anti-rheumatic effects of gold sodium thiomalate. Gold Sodium Thiomalate 108-130 high mobility group box 1 Homo sapiens 31-36 17913975-10 2008 Furthermore, gold chloride also inhibited release of HMGB1. gold chloride 13-26 high mobility group box 1 Homo sapiens 53-58 17667523-6 2007 Rather, we observed that tumor cells incubated with the platinating agent oxaliplatin, retained HMGB1 within the nucleus for significantly longer periods than other agents used at comparable cytotoxic concentrations or even with potent cytolytic cells. Oxaliplatin 74-85 high mobility group box 1 Homo sapiens 96-101 18049445-4 2007 The "cholinergic anti-inflammatory pathway" mediated by acetylcholine acts by inhibiting the production of tumor necrosis factor, interleukin-1, macrophage migration inhibitory factor, and high mobility group box-1 and suppresses the activation of nuclear factor-kappa B expression. Acetylcholine 56-69 high mobility group box 1 Homo sapiens 189-214 17984303-0 2007 HMGB1 release induced by liver ischemia involves Toll-like receptor 4 dependent reactive oxygen species production and calcium-mediated signaling. Reactive Oxygen Species 80-103 high mobility group box 1 Homo sapiens 0-5 17984303-0 2007 HMGB1 release induced by liver ischemia involves Toll-like receptor 4 dependent reactive oxygen species production and calcium-mediated signaling. Calcium 119-126 high mobility group box 1 Homo sapiens 0-5 17984303-4 2007 HMGB1 release from cultured hepatocytes was found to be an active process regulated by reactive oxygen species (ROS). Reactive Oxygen Species 87-110 high mobility group box 1 Homo sapiens 0-5 17984303-4 2007 HMGB1 release from cultured hepatocytes was found to be an active process regulated by reactive oxygen species (ROS). Reactive Oxygen Species 112-115 high mobility group box 1 Homo sapiens 0-5 17984303-7 2007 HMGB1 release induced by oxidative stress was markedly reduced by inhibition of calcium/calmodulin-dependent kinases (CaMKs), a family of proteins involved in a wide range of calcium-linked signaling events. Calcium 80-87 high mobility group box 1 Homo sapiens 0-5 17984303-9 2007 Collectively, these results demonstrate that hypoxia-induced HMGB1 release by hepatocytes is an active, regulated process that occurs through a mechanism promoted by TLR4-dependent ROS production and downstream CaMK-mediated signaling. Reactive Oxygen Species 181-184 high mobility group box 1 Homo sapiens 61-66 18073062-11 2007 There were positive correlations between HMGB1 and TBil and negative correlations between HMGB1 and PTA. tbil 51-55 high mobility group box 1 Homo sapiens 41-46 17885340-8 2007 We observed a relationship between hemodynamic improvement and increase of the serum HMGB-1 levels and between improvement of respiratory functions and increase of the serum 2-AG and PAI-1 levels in septic shock patients treated with DHP-PMX. dhp-pmx 234-241 high mobility group box 1 Homo sapiens 85-91 17525840-8 2007 A positive correlation was observed between HMGB1 and SOFA score and lactate, and procalcitonin concentrations. Lactic Acid 69-76 high mobility group box 1 Homo sapiens 44-49 17430886-7 2007 Both full-length HMGB1 and a truncated form of HMGB1 lacking the highly conserved glutamate-rich C-terminal tail can induce macrophage activation and tumor necrosis factor-alpha production. Glutamic Acid 82-91 high mobility group box 1 Homo sapiens 17-22 17611636-0 2007 Ethyl pyruvate induces necrosis-to-apoptosis switch and inhibits high mobility group box protein 1 release in A549 lung adenocarcinoma cells. ethyl pyruvate 0-14 high mobility group box 1 Homo sapiens 65-98 17611636-1 2007 Ethyl pyruvate (EP), a stable lipophilic pyruvate derivative, has been shown to exert anti-inflammatory activities through inhibiting the expression of various pro-inflammatory mediators as well as circulating levels of high mobility group box protein 1 (HMGB1) in a variety of in vitro and in vivo model systems. ethyl pyruvate 0-14 high mobility group box 1 Homo sapiens 220-253 17611636-1 2007 Ethyl pyruvate (EP), a stable lipophilic pyruvate derivative, has been shown to exert anti-inflammatory activities through inhibiting the expression of various pro-inflammatory mediators as well as circulating levels of high mobility group box protein 1 (HMGB1) in a variety of in vitro and in vivo model systems. ethyl pyruvate 0-14 high mobility group box 1 Homo sapiens 255-260 17611636-1 2007 Ethyl pyruvate (EP), a stable lipophilic pyruvate derivative, has been shown to exert anti-inflammatory activities through inhibiting the expression of various pro-inflammatory mediators as well as circulating levels of high mobility group box protein 1 (HMGB1) in a variety of in vitro and in vivo model systems. ethyl pyruvate 16-18 high mobility group box 1 Homo sapiens 220-253 17611636-1 2007 Ethyl pyruvate (EP), a stable lipophilic pyruvate derivative, has been shown to exert anti-inflammatory activities through inhibiting the expression of various pro-inflammatory mediators as well as circulating levels of high mobility group box protein 1 (HMGB1) in a variety of in vitro and in vivo model systems. ethyl pyruvate 16-18 high mobility group box 1 Homo sapiens 255-260 17611636-1 2007 Ethyl pyruvate (EP), a stable lipophilic pyruvate derivative, has been shown to exert anti-inflammatory activities through inhibiting the expression of various pro-inflammatory mediators as well as circulating levels of high mobility group box protein 1 (HMGB1) in a variety of in vitro and in vivo model systems. Pyruvic Acid 6-14 high mobility group box 1 Homo sapiens 220-253 17611636-1 2007 Ethyl pyruvate (EP), a stable lipophilic pyruvate derivative, has been shown to exert anti-inflammatory activities through inhibiting the expression of various pro-inflammatory mediators as well as circulating levels of high mobility group box protein 1 (HMGB1) in a variety of in vitro and in vivo model systems. Pyruvic Acid 6-14 high mobility group box 1 Homo sapiens 255-260 17611636-4 2007 Here we show that EP prevented GD-induced necrosis and HMGB1 release and switched the cell death mode to apoptosis through inhibiting GD-induced CuZn superoxide dismutase release and ROS production. Gadolinium 31-33 high mobility group box 1 Homo sapiens 55-60 17430886-7 2007 Both full-length HMGB1 and a truncated form of HMGB1 lacking the highly conserved glutamate-rich C-terminal tail can induce macrophage activation and tumor necrosis factor-alpha production. Glutamic Acid 82-91 high mobility group box 1 Homo sapiens 47-52 17430886-8 2007 However, displacement of HMGB1 from the nucleus following PARP activation requires the presence of the glutamate-rich C-terminal tail. Glutamic Acid 103-112 high mobility group box 1 Homo sapiens 25-30 17515723-3 2007 We investigated the high-mobility group box-1 (HMGB-1) level in septic shock patients treated with DHP-PMX. dhp-pmx 99-106 high mobility group box 1 Homo sapiens 20-45 17513788-0 2007 Nuclear heat shock protein 72 as a negative regulator of oxidative stress (hydrogen peroxide)-induced HMGB1 cytoplasmic translocation and release. Hydrogen Peroxide 75-92 high mobility group box 1 Homo sapiens 102-107 17515723-3 2007 We investigated the high-mobility group box-1 (HMGB-1) level in septic shock patients treated with DHP-PMX. dhp-pmx 99-106 high mobility group box 1 Homo sapiens 47-53 17515723-9 2007 In the E group, only the HMGB-1 levels improved significantly after DHP-PMX. dhp-pmx 68-75 high mobility group box 1 Homo sapiens 25-31 17515723-10 2007 Present data suggest that the circulation dynamics of septic shock patients can be improved by reducing HMGB-1 levels by using DHP-PMX. dhp-pmx 127-134 high mobility group box 1 Homo sapiens 104-110 17334246-14 2007 CONCLUSIONS: HMGB1 is elevated in almost all CAP subjects, and higher circulating HMGB1 is associated with mortality. cap 45-48 high mobility group box 1 Homo sapiens 13-18 17437420-7 2007 The serum HMGB1 level in CSS patients decreased after the steroid therapy, and showed significant positive correlations with several molecules, including soluble interleukin-2 receptor, soluble thrombomodulin, and eosinophil cationic protein in sera. Steroids 58-65 high mobility group box 1 Homo sapiens 10-15 17697615-13 2007 Inhibitors specific for the JNK (SP600125), MEK (PD98059), and p38 MAPK (SB203580), abrogated HMGB1-induced TNF-alpha secretion. pyrazolanthrone 33-41 high mobility group box 1 Homo sapiens 94-99 17697615-13 2007 Inhibitors specific for the JNK (SP600125), MEK (PD98059), and p38 MAPK (SB203580), abrogated HMGB1-induced TNF-alpha secretion. 2-(2-amino-3-methoxyphenyl)-4H-1-benzopyran-4-one 49-56 high mobility group box 1 Homo sapiens 94-99 17697615-13 2007 Inhibitors specific for the JNK (SP600125), MEK (PD98059), and p38 MAPK (SB203580), abrogated HMGB1-induced TNF-alpha secretion. SB 203580 73-81 high mobility group box 1 Homo sapiens 94-99 17462578-3 2007 We show that glycyrrhizin, a natural anti-inflammatory and antiviral triterpene in clinical use, inhibits HMGB1 chemoattractant and mitogenic activities, and has a weak inhibitory effect on its intranuclear DNA-binding function. Glycyrrhizic Acid 13-25 high mobility group box 1 Homo sapiens 106-111 17462578-4 2007 NMR and fluorescence studies indicate that glycyrrhizin binds directly to HMGB1 (K(d) approximately 150 microM), interacting with two shallow concave surfaces formed by the two arms of both HMG boxes. Glycyrrhizic Acid 43-55 high mobility group box 1 Homo sapiens 74-79 17235565-8 2007 The effect of HMGB-1 was abrogated by RAGE down-regulation by antisense S-oligodeoxynucleic acid. oligodeoxynucleic acid 74-96 high mobility group box 1 Homo sapiens 14-20 17339485-4 2007 However, several steroid-like pigments (tanshinone I, tanshinone IIA, and cryptotanshinone) of a popular Chinese herb, Danshen (Salvia miltiorrhiza), dose dependently attenuated endotoxin-induced HMGB1 release in macrophage/monocyte cultures. Steroids 17-24 high mobility group box 1 Homo sapiens 196-201 17339485-4 2007 However, several steroid-like pigments (tanshinone I, tanshinone IIA, and cryptotanshinone) of a popular Chinese herb, Danshen (Salvia miltiorrhiza), dose dependently attenuated endotoxin-induced HMGB1 release in macrophage/monocyte cultures. cryptotanshinone 74-90 high mobility group box 1 Homo sapiens 196-201 17339485-5 2007 A water-soluble tanshinone IIA sodium sulfonate derivative (TSNIIA-SS), which has been widely used as a Chinese medicine for patients with cardiovascular disorders, selectively abrogated endotoxin-induced HMGB1 cytoplasmic translocation and release in a glucocorticoid receptor-independent manner. Water 2-7 high mobility group box 1 Homo sapiens 205-210 17339485-5 2007 A water-soluble tanshinone IIA sodium sulfonate derivative (TSNIIA-SS), which has been widely used as a Chinese medicine for patients with cardiovascular disorders, selectively abrogated endotoxin-induced HMGB1 cytoplasmic translocation and release in a glucocorticoid receptor-independent manner. tanshinone II A sodium sulfonate 16-47 high mobility group box 1 Homo sapiens 205-210 17135572-0 2007 Hydrogen peroxide stimulates macrophages and monocytes to actively release HMGB1. Hydrogen Peroxide 0-17 high mobility group box 1 Homo sapiens 75-80 17135572-2 2007 Here, we demonstrated that a reactive oxygen species, hydrogen peroxide (H(2)O(2)), induces active and passive HMGB1 release from macrophage and monocyte cultures in a time- and dose-dependent manner. Reactive Oxygen Species 29-52 high mobility group box 1 Homo sapiens 111-116 17135572-2 2007 Here, we demonstrated that a reactive oxygen species, hydrogen peroxide (H(2)O(2)), induces active and passive HMGB1 release from macrophage and monocyte cultures in a time- and dose-dependent manner. Hydrogen Peroxide 54-71 high mobility group box 1 Homo sapiens 111-116 17135572-2 2007 Here, we demonstrated that a reactive oxygen species, hydrogen peroxide (H(2)O(2)), induces active and passive HMGB1 release from macrophage and monocyte cultures in a time- and dose-dependent manner. Hydrogen Peroxide 73-81 high mobility group box 1 Homo sapiens 111-116 17135572-3 2007 At nontoxic doses (e.g., 0.0125-0.125 mM), H(2)O(2) induced HMGB1 cytoplasmic translocation and active release within 3-24 h. At higher concentrations (e.g., 0.25 mM), however, H(2)O(2) exhibited cytotoxicity to macrophage and monocyte cell cultures and consequently, triggered active and passive HMGB1 release. Hydrogen Peroxide 43-51 high mobility group box 1 Homo sapiens 60-65 17135572-3 2007 At nontoxic doses (e.g., 0.0125-0.125 mM), H(2)O(2) induced HMGB1 cytoplasmic translocation and active release within 3-24 h. At higher concentrations (e.g., 0.25 mM), however, H(2)O(2) exhibited cytotoxicity to macrophage and monocyte cell cultures and consequently, triggered active and passive HMGB1 release. Hydrogen Peroxide 43-51 high mobility group box 1 Homo sapiens 297-302 17135572-4 2007 In addition, H(2)O(2) stimulated potential interaction of HMGB1 with a nuclear export factor, chromosome region maintenance (CRM1), in macrophage/monocyte cultures. Hydrogen Peroxide 13-21 high mobility group box 1 Homo sapiens 58-63 17135572-5 2007 Inhibitors specific for the JNK (SP600125) and MEK (PD98059), but not p38 MAPK (SB203580), abrogated H(2)O(2)-induced, active HMGB1 release. pyrazolanthrone 33-41 high mobility group box 1 Homo sapiens 126-131 17135572-5 2007 Inhibitors specific for the JNK (SP600125) and MEK (PD98059), but not p38 MAPK (SB203580), abrogated H(2)O(2)-induced, active HMGB1 release. 2-(2-amino-3-methoxyphenyl)-4H-1-benzopyran-4-one 52-59 high mobility group box 1 Homo sapiens 126-131 17135572-5 2007 Inhibitors specific for the JNK (SP600125) and MEK (PD98059), but not p38 MAPK (SB203580), abrogated H(2)O(2)-induced, active HMGB1 release. 2)o(2) 103-109 high mobility group box 1 Homo sapiens 126-131 17187938-0 2007 Inhibition of HMGB1 by deep ocean water attenuates endotoxin-induced sepsis. Water 34-39 high mobility group box 1 Homo sapiens 14-19 16968820-7 2007 TRAIL treatment, however, induced HMGB1 release, and it is interesting that this extrinsic pathway-mediated cell death was blocked with the pancaspase inhibitor N-benzyloxycarbonyl-Val-Ala-Asp-fluoromethylketone. Caspase Inhibitor VI 161-211 high mobility group box 1 Homo sapiens 34-39 16980512-3 2007 Mass analyses revealed that recombinant eukaryotic HMGB1 has an intrachain disulphide bond. disulphide 75-85 high mobility group box 1 Homo sapiens 51-56 16980512-4 2007 In mass analysis of tissue-derived HMGB1, two forms were detected: the carboxyl terminal glutamic acid residue lacking form and a full-length form. Glutamic Acid 89-102 high mobility group box 1 Homo sapiens 35-40 16980512-5 2007 Cell culture studies indicated that both eukaryotic and bacterial HMGB1 proteins induce TNF-alpha secretion and nitric oxide release from mononuclear cells. Nitric Oxide 112-124 high mobility group box 1 Homo sapiens 66-71 16980512-7 2007 A soluble proinflammatory substance was separated from the bacterial recombinant HMGB1 by chloroform-methanol treatment. Chloroform 90-100 high mobility group box 1 Homo sapiens 81-86 16980512-7 2007 A soluble proinflammatory substance was separated from the bacterial recombinant HMGB1 by chloroform-methanol treatment. Methanol 101-109 high mobility group box 1 Homo sapiens 81-86 17958722-9 2007 Serum NE activities and serum concentrations of IL-1beta, IL-6, and HMGB1 were significantly suppressed in the Sivelestat-treated group. sivelestat 111-121 high mobility group box 1 Homo sapiens 68-73 17678956-0 2007 The high-mobility group box 1 cytokine induces transporter-mediated release of glutamate from glial subcellular particles (gliosomes) prepared from in situ-matured astrocytes. Glutamic Acid 79-88 high mobility group box 1 Homo sapiens 4-29 17678956-8 2007 HMGB1-induced release of the stable glutamate analogue [(3)H]d-aspartate and endogenous glutamate form gliosomes, whereas nerve terminals were insensitive to the protein. Glutamic Acid 36-45 high mobility group box 1 Homo sapiens 0-5 17678956-8 2007 HMGB1-induced release of the stable glutamate analogue [(3)H]d-aspartate and endogenous glutamate form gliosomes, whereas nerve terminals were insensitive to the protein. Deuterium 8-9 high mobility group box 1 Homo sapiens 0-5 17678956-8 2007 HMGB1-induced release of the stable glutamate analogue [(3)H]d-aspartate and endogenous glutamate form gliosomes, whereas nerve terminals were insensitive to the protein. Aspartic Acid 63-72 high mobility group box 1 Homo sapiens 0-5 17678956-8 2007 HMGB1-induced release of the stable glutamate analogue [(3)H]d-aspartate and endogenous glutamate form gliosomes, whereas nerve terminals were insensitive to the protein. Glutamic Acid 88-97 high mobility group box 1 Homo sapiens 0-5 17678956-9 2007 The HMGB1-evoked release of glutamate was independent on modifications of cytosolic Ca(2+) concentration, but it was blocked by dl-threo-beta-benzyloxyaspartate, suggesting the involvement of transporter-mediated release mechanisms. Glutamic Acid 28-37 high mobility group box 1 Homo sapiens 4-9 17678956-9 2007 The HMGB1-evoked release of glutamate was independent on modifications of cytosolic Ca(2+) concentration, but it was blocked by dl-threo-beta-benzyloxyaspartate, suggesting the involvement of transporter-mediated release mechanisms. benzyloxyaspartate 128-160 high mobility group box 1 Homo sapiens 4-9 17678956-12 2007 Taken together, these results suggest that the HMGB1 cytokine could act as a modulator of glutamate homeostasis in adult mammalian brain. Glutamic Acid 90-99 high mobility group box 1 Homo sapiens 47-52 17636313-5 2007 Although HMGB1 stimulates ATP hydrolysis by topo IIalpha, the DNA cleavage is much more enhanced. Adenosine Triphosphate 26-29 high mobility group box 1 Homo sapiens 9-14 16842881-6 2006 The potential use of PAMAM-Arg was demonstrated by efficient gene knock-down by transfecting HMGB1 shRNA-expressing plasmid. pamam-arg 21-30 high mobility group box 1 Homo sapiens 93-98 18066803-0 2007 Bent oligonucleotide duplexes as HMGB1 inhibitors: a comparative study. Oligonucleotides 5-20 high mobility group box 1 Homo sapiens 33-38 18066803-1 2007 In this work we explore the ability of a chimeric LNA/DNA bent duplex, in which the kink is induced by 2 unpaired adenines in the middle of one strand, to bind HMGB1, a protein involved in many inflammatory processes. Adenine 114-122 high mobility group box 1 Homo sapiens 160-165 16855214-7 2006 Results of these experiments indicate that HMGB1 appears in the media of apoptotic Jurkat cells in a time-dependent manner and that this release can be reduced by Z-VAD-fmk. benzyloxycarbonylvalyl-alanyl-aspartyl fluoromethyl ketone 163-172 high mobility group box 1 Homo sapiens 43-48 17114460-5 2006 Phosphorylation of HMGB1 was detected by metabolic labeling and Western blot analysis after treatments with TNF-alpha and okadaic acid, a phosphatase inhibitor. Okadaic Acid 122-134 high mobility group box 1 Homo sapiens 19-24 17114460-8 2006 We mutated serine residues of either or both NLSs of HMGB1 to glutamic acid to simulate a phosphorylated state and examined the binding of HMGB1 to karyopherin-alpha1, which was identified as the nuclear import protein for HMGB1 in this study. Serine 11-17 high mobility group box 1 Homo sapiens 53-58 17114460-8 2006 We mutated serine residues of either or both NLSs of HMGB1 to glutamic acid to simulate a phosphorylated state and examined the binding of HMGB1 to karyopherin-alpha1, which was identified as the nuclear import protein for HMGB1 in this study. Glutamic Acid 62-75 high mobility group box 1 Homo sapiens 53-58 16911580-10 2006 These results suggest that the HMGB1 cytokine could act as a modulator of glutamate homeostasis in adult mammal brain. Glutamic Acid 74-83 high mobility group box 1 Homo sapiens 31-36 16732569-0 2006 The effect of manganese(II) on the structure of DNA/HMGB1/H1 complexes: electronic and vibrational circular dichroism studies. Manganese(2+) 14-27 high mobility group box 1 Homo sapiens 52-57 16732569-1 2006 The interactions were studied of DNA with the nonhistone chromatin protein HMGB1 and histone H1 in the presence of manganese(II) ions at different protein to DNA and manganese to DNA phosphate ratios by using absorption and optical activity spectroscopy in the electronic [ultraviolet (UV) and electronic circular dichroism ECD)] and vibrational [infrared (IR) and vibrational circular dichroism (VCD)] regions. Manganese(2+) 115-128 high mobility group box 1 Homo sapiens 75-80 16732569-3 2006 At the same time, the presence of HMGB1 and H1 also changed the mode of interaction of Mn2+ with DNA, which now takes place mostly in the major groove of DNA involving N7(G), whereas interactions between Mn2+ and DNA phosphate groups are weakened by histone molecules. Manganese(2+) 87-91 high mobility group box 1 Homo sapiens 34-39 16732569-3 2006 At the same time, the presence of HMGB1 and H1 also changed the mode of interaction of Mn2+ with DNA, which now takes place mostly in the major groove of DNA involving N7(G), whereas interactions between Mn2+ and DNA phosphate groups are weakened by histone molecules. Manganese(2+) 204-208 high mobility group box 1 Homo sapiens 34-39 16732569-3 2006 At the same time, the presence of HMGB1 and H1 also changed the mode of interaction of Mn2+ with DNA, which now takes place mostly in the major groove of DNA involving N7(G), whereas interactions between Mn2+ and DNA phosphate groups are weakened by histone molecules. Phosphates 217-226 high mobility group box 1 Homo sapiens 34-39 17060403-6 2006 However, cultured SMCs actively secrete HMGB1 after cholesterol loading. Cholesterol 52-63 high mobility group box 1 Homo sapiens 40-45 17079940-8 2006 Serum HMGB1 levels were significantly positively correlated with lactate dehydrogenase, C-reactive protein, and total bilirubin. Bilirubin 118-127 high mobility group box 1 Homo sapiens 6-11 16814517-3 2006 Ethyl pyruvate specifically inhibits tumor necrosis factor-alpha production and decreases circulating levels of high-mobility group box-1 and nuclear factor-kappaB signaling pathways by specifically targeting its p65 subunit in animals with established endotoxemia or sepsis and in macrophage cultures. ethyl pyruvate 0-14 high mobility group box 1 Homo sapiens 112-137 16842881-7 2006 The numbers of green fluorescent protein (GFP)-positive and HMGB1-negative cells indicated that PAMAM-Arg/shRNA-expressing plasmid complex suppressed target gene expression in over 40% of cells, which is the highest level achieved to date in primary cortical culture by any gene carrier. Poly(amidoamine) 96-101 high mobility group box 1 Homo sapiens 60-65 16842881-7 2006 The numbers of green fluorescent protein (GFP)-positive and HMGB1-negative cells indicated that PAMAM-Arg/shRNA-expressing plasmid complex suppressed target gene expression in over 40% of cells, which is the highest level achieved to date in primary cortical culture by any gene carrier. Arginine 102-105 high mobility group box 1 Homo sapiens 60-65 16807371-0 2006 Activation of sPLA2-IIA and PGE2 production by high mobility group protein B1 in vascular smooth muscle cells sensitized by IL-1beta. Dinoprostone 28-32 high mobility group box 1 Homo sapiens 47-77 16807371-7 2006 The present study investigates the role of HMGB1 in the activation of sPLA2-IIA expression and PGE2 production in VSMCs. Dinoprostone 95-99 high mobility group box 1 Homo sapiens 43-48 16807371-8 2006 Recombinant HMGB1 slightly activated the sPLA2-IIA, COX-2, and mPGES-1 genes but dramatically stimulated these genes in VSMCs that had been incubated with the proinflammatory cytokine IL-1beta for 24 h. This effect was accompanied by significantly increased PGE2 release. Dinoprostone 258-262 high mobility group box 1 Homo sapiens 12-17 16282196-0 2006 HMGB1 is secreted by immunostimulated enterocytes and contributes to cytomix-induced hyperpermeability of Caco-2 monolayers. caco-2 106-112 high mobility group box 1 Homo sapiens 0-5 16775128-6 2006 HMGB1 was found to accumulate in NMDA-treated primary cortical culture media, and supernatants collected from these cultures were found to trigger microglia activation, the hallmark of brain inflammation. N-Methylaspartate 33-37 high mobility group box 1 Homo sapiens 0-5 16641887-1 2006 Pyruvic acid, an intermediate metabolite of glucose, an effective scavenger of reactive oxygen species (ROS), inhibits tumor necrosis factor-alpha production and NF-kappaB signaling pathways, reduces circulating levels of HMGB1 (high mobility group B1), decreases COX-2 (cyclo-oxygenase-2), iNOS (inducible nitric oxide synthase), and IL-6 (interleukin-6) mRNA expression in liver, ileal mucosa, and colonic mucosa in animal models with endotoxemia. Pyruvic Acid 0-12 high mobility group box 1 Homo sapiens 222-227 16641887-1 2006 Pyruvic acid, an intermediate metabolite of glucose, an effective scavenger of reactive oxygen species (ROS), inhibits tumor necrosis factor-alpha production and NF-kappaB signaling pathways, reduces circulating levels of HMGB1 (high mobility group B1), decreases COX-2 (cyclo-oxygenase-2), iNOS (inducible nitric oxide synthase), and IL-6 (interleukin-6) mRNA expression in liver, ileal mucosa, and colonic mucosa in animal models with endotoxemia. Pyruvic Acid 0-12 high mobility group box 1 Homo sapiens 229-251 16641887-1 2006 Pyruvic acid, an intermediate metabolite of glucose, an effective scavenger of reactive oxygen species (ROS), inhibits tumor necrosis factor-alpha production and NF-kappaB signaling pathways, reduces circulating levels of HMGB1 (high mobility group B1), decreases COX-2 (cyclo-oxygenase-2), iNOS (inducible nitric oxide synthase), and IL-6 (interleukin-6) mRNA expression in liver, ileal mucosa, and colonic mucosa in animal models with endotoxemia. Glucose 44-51 high mobility group box 1 Homo sapiens 222-227 16641887-1 2006 Pyruvic acid, an intermediate metabolite of glucose, an effective scavenger of reactive oxygen species (ROS), inhibits tumor necrosis factor-alpha production and NF-kappaB signaling pathways, reduces circulating levels of HMGB1 (high mobility group B1), decreases COX-2 (cyclo-oxygenase-2), iNOS (inducible nitric oxide synthase), and IL-6 (interleukin-6) mRNA expression in liver, ileal mucosa, and colonic mucosa in animal models with endotoxemia. Glucose 44-51 high mobility group box 1 Homo sapiens 229-251 16354107-6 2006 In contrast, IL-1 stimulates the release of the immunological adjuvant high mobility group box 1 protein (HMGB1; a biochemical maker of necrosis) in a nitric oxide-dependent manner, while apoptosis inducers fail to stimulate HMGB1 release. Nitric Oxide 151-163 high mobility group box 1 Homo sapiens 71-96 16697213-5 2006 Dexamethasone, a known inhibitor of NF-kappaB signaling in U1 cells, inhibited HMGB1-induced stimulation of the viral production. Dexamethasone 0-13 high mobility group box 1 Homo sapiens 79-84 16403427-5 2006 Recombinant Sbp1p modified by Hmt1p in vivo were isolated by affinity chromatography followed by electrophoresis on a polyacrylamide gel and subjected to acid hydrolysis. polyacrylamide 118-132 high mobility group box 1 Homo sapiens 12-17 16354107-6 2006 In contrast, IL-1 stimulates the release of the immunological adjuvant high mobility group box 1 protein (HMGB1; a biochemical maker of necrosis) in a nitric oxide-dependent manner, while apoptosis inducers fail to stimulate HMGB1 release. Nitric Oxide 151-163 high mobility group box 1 Homo sapiens 106-111 15994314-5 2005 To identify determinants of HMGB1 required for stimulation of RAG-mediated RSS binding and cleavage, we prepared an extensive panel of mutant HMGB1 proteins and tested their ability to augment RAG-mediated RSS binding and cleavage activity. RSS 75-78 high mobility group box 1 Homo sapiens 28-33 15912505-6 2005 Histone H1 facilitated binding of HMGB1 to DNA by interacting with the negatively charged groups of the sugar-phosphate backbone and binding of aspartic and glutamic amino acid residues of HMGB1. Sugar Phosphates 104-119 high mobility group box 1 Homo sapiens 34-39 15912505-6 2005 Histone H1 facilitated binding of HMGB1 to DNA by interacting with the negatively charged groups of the sugar-phosphate backbone and binding of aspartic and glutamic amino acid residues of HMGB1. aspartic 144-152 high mobility group box 1 Homo sapiens 34-39 15912505-6 2005 Histone H1 facilitated binding of HMGB1 to DNA by interacting with the negatively charged groups of the sugar-phosphate backbone and binding of aspartic and glutamic amino acid residues of HMGB1. aspartic 144-152 high mobility group box 1 Homo sapiens 189-194 15912505-6 2005 Histone H1 facilitated binding of HMGB1 to DNA by interacting with the negatively charged groups of the sugar-phosphate backbone and binding of aspartic and glutamic amino acid residues of HMGB1. glutamic amino acid 157-176 high mobility group box 1 Homo sapiens 34-39 15912505-6 2005 Histone H1 facilitated binding of HMGB1 to DNA by interacting with the negatively charged groups of the sugar-phosphate backbone and binding of aspartic and glutamic amino acid residues of HMGB1. glutamic amino acid 157-176 high mobility group box 1 Homo sapiens 189-194 15922304-2 2005 The domain A of HMGB1 (HMGB1a protein) binds to the adduct formed by the R enantiomer at the CGGA sequence with a similar high affinity as it does to the adduct of antitumor cisplatin, and to the adduct formed by the S enantiomer with a slightly lower affinity. Cisplatin 174-183 high mobility group box 1 Homo sapiens 16-21 16040616-4 2005 In vitro glutathione S-transferase pull-down and in vivo coimmunoprecipitation studies confirmed an interaction between HMGB1 and both SREBP-1 and -2. Glutathione 9-20 high mobility group box 1 Homo sapiens 120-125 15823048-0 2005 The inhibitory effect of HMGB-1 protein on the repair of cisplatin-damaged DNA is accomplished through the acidic domain. Cisplatin 57-66 high mobility group box 1 Homo sapiens 25-31 15823048-5 2005 These findings strongly suggest that the HMGB-1-induced inhibition of cisplatin-DNA adduct repair is accomplished through the acidic domain. Cisplatin 70-79 high mobility group box 1 Homo sapiens 41-47 15687351-0 2005 Suppression of HMGB1 release by stearoyl lysophosphatidylcholine:an additional mechanism for its therapeutic effects in experimental sepsis. stearoyl alpha-lysolecithin 32-64 high mobility group box 1 Homo sapiens 15-20 15687351-2 2005 Here, we demonstrate that stearoyl LPC, but not caproyl LPC, significantly attenuates circulating high-mobility group box 1 (HMGB1) levels in endotoxemia and sepsis by suppressing endotoxin-induced HMGB1 release from macrophages/monocytes. stearoyl alpha-lysolecithin 26-38 high mobility group box 1 Homo sapiens 98-123 15687351-2 2005 Here, we demonstrate that stearoyl LPC, but not caproyl LPC, significantly attenuates circulating high-mobility group box 1 (HMGB1) levels in endotoxemia and sepsis by suppressing endotoxin-induced HMGB1 release from macrophages/monocytes. stearoyl alpha-lysolecithin 26-38 high mobility group box 1 Homo sapiens 125-130 15687351-2 2005 Here, we demonstrate that stearoyl LPC, but not caproyl LPC, significantly attenuates circulating high-mobility group box 1 (HMGB1) levels in endotoxemia and sepsis by suppressing endotoxin-induced HMGB1 release from macrophages/monocytes. stearoyl alpha-lysolecithin 26-38 high mobility group box 1 Homo sapiens 198-203 15687351-2 2005 Here, we demonstrate that stearoyl LPC, but not caproyl LPC, significantly attenuates circulating high-mobility group box 1 (HMGB1) levels in endotoxemia and sepsis by suppressing endotoxin-induced HMGB1 release from macrophages/monocytes. Lysophosphatidylcholines 35-38 high mobility group box 1 Homo sapiens 98-123 15687351-2 2005 Here, we demonstrate that stearoyl LPC, but not caproyl LPC, significantly attenuates circulating high-mobility group box 1 (HMGB1) levels in endotoxemia and sepsis by suppressing endotoxin-induced HMGB1 release from macrophages/monocytes. Lysophosphatidylcholines 35-38 high mobility group box 1 Homo sapiens 125-130 15687351-2 2005 Here, we demonstrate that stearoyl LPC, but not caproyl LPC, significantly attenuates circulating high-mobility group box 1 (HMGB1) levels in endotoxemia and sepsis by suppressing endotoxin-induced HMGB1 release from macrophages/monocytes. Lysophosphatidylcholines 35-38 high mobility group box 1 Homo sapiens 198-203 15687351-4 2005 Thus, stearoyl LPC confers protection against lethal experimental sepsis partly by facilitating the elimination of the invading pathogens and partly by inhibiting endotoxin-induced release of a late proinflammatory cytokine, HMGB1. stearoyl alpha-lysolecithin 6-18 high mobility group box 1 Homo sapiens 225-230 15502843-2 2004 Here we report that the neurotransmitter acetylcholine inhibits HMGB1 release from human macrophages by signaling through a nicotinic acetylcholine receptor. Acetylcholine 41-54 high mobility group box 1 Homo sapiens 64-69 15943034-3 2005 Production of IL-15 and/or TGF-alpha was also associated with amphoterin mRNA expression in colon cancer tissues with TAM depletion in both Dukes" B and C tumors (P = 0.0167 and P = 0.0062, respectively). tam 118-121 high mobility group box 1 Homo sapiens 62-72 15584967-5 2004 A nuclear relocation of HMGB1 to the cytoplasm was seen at 4 h. Subsequently, high amounts of HMGB1 could be seen in the supernatants from stimulated cells after 16 h. It was also observed that the pro-inflammatory activity of HMGB1 is sensitive to dexamethasone. Dexamethasone 249-262 high mobility group box 1 Homo sapiens 24-29 15584967-5 2004 A nuclear relocation of HMGB1 to the cytoplasm was seen at 4 h. Subsequently, high amounts of HMGB1 could be seen in the supernatants from stimulated cells after 16 h. It was also observed that the pro-inflammatory activity of HMGB1 is sensitive to dexamethasone. Dexamethasone 249-262 high mobility group box 1 Homo sapiens 94-99 15584967-5 2004 A nuclear relocation of HMGB1 to the cytoplasm was seen at 4 h. Subsequently, high amounts of HMGB1 could be seen in the supernatants from stimulated cells after 16 h. It was also observed that the pro-inflammatory activity of HMGB1 is sensitive to dexamethasone. Dexamethasone 249-262 high mobility group box 1 Homo sapiens 94-99 15584967-6 2004 Interestingly, the HMGB1-induced TNF-alpha release from monocytes could be inhibited by either the A-box of the protein or the p38 inhibitor CNI-1493, but neither had any inhibitory effects on the HMGB1-dependent upregulation of cell-adhesion molecules on HUVEC. semapimod 141-149 high mobility group box 1 Homo sapiens 19-24 15743787-5 2005 The number of PMA-U937 cells was markedly decreased by co-culture with WiDr cells exposed to HMGB1/amphoterin sense S-oligodeoxynucleotide (ODN) in spheroids or monolayers. Oligodeoxyribonucleotides 116-138 high mobility group box 1 Homo sapiens 99-109 15743787-5 2005 The number of PMA-U937 cells was markedly decreased by co-culture with WiDr cells exposed to HMGB1/amphoterin sense S-oligodeoxynucleotide (ODN) in spheroids or monolayers. Oligodeoxyribonucleotides 140-143 high mobility group box 1 Homo sapiens 99-109 15743787-8 2005 Recombinant human HMGB1/amphoterin induced growth inhibition in thioglycollate-induced rat peritoneal macrophages, PMA-U937 cells, and human alveolar macrophages, an effect that was abrogated by absorption with anti-HMGB1 antibody. Thioglycolates 64-78 high mobility group box 1 Homo sapiens 18-23 15743787-8 2005 Recombinant human HMGB1/amphoterin induced growth inhibition in thioglycollate-induced rat peritoneal macrophages, PMA-U937 cells, and human alveolar macrophages, an effect that was abrogated by absorption with anti-HMGB1 antibody. Thioglycolates 64-78 high mobility group box 1 Homo sapiens 24-34 15743787-8 2005 Recombinant human HMGB1/amphoterin induced growth inhibition in thioglycollate-induced rat peritoneal macrophages, PMA-U937 cells, and human alveolar macrophages, an effect that was abrogated by absorption with anti-HMGB1 antibody. Thioglycolates 64-78 high mobility group box 1 Homo sapiens 216-221 15672453-6 2005 Moreover, a group of downregulated genes, with corresponding disappearing proteins (HSP90, 14-3-3 protein, HMGB1) in two-dimensional polyacrylamide gel electrophoresis (2-D PAGE), are good candidates for use as biomarkers of uranium effects. polyacrylamide 133-147 high mobility group box 1 Homo sapiens 107-112 15612592-0 2004 [The structure of the complexes of DNA with non-histone chromosomal protein HMGB1 in the presence of manganese ions]. Manganese 101-110 high mobility group box 1 Homo sapiens 76-81 15612592-1 2004 The complexes of DNA - HMGB1 protein - manganese ions have been studied using circular dichroism (CD) technique. Manganese 39-48 high mobility group box 1 Homo sapiens 23-28 15612592-5 2004 The presence of Mn2+ ions prevents formation of the ordered supramolecular structures specific for the HMGB1-DNA complexes. Manganese(2+) 16-20 high mobility group box 1 Homo sapiens 103-108 15612592-9 2004 Moreover, the changes in local DNA structure induced by manganese ions promote the appearance of new HMGB1 binding sites on the DNA double helix. Manganese 56-65 high mobility group box 1 Homo sapiens 101-106 15612592-13 2004 Thus, structural changes of the DNA double helix in the three-component DNA-HMGB1-Mn2+ complexes come as a result of the combined and interdependent interactions of DNA with Mn2+ ions and the molecules of HMGB1. Manganese(2+) 82-86 high mobility group box 1 Homo sapiens 76-81 15612592-13 2004 Thus, structural changes of the DNA double helix in the three-component DNA-HMGB1-Mn2+ complexes come as a result of the combined and interdependent interactions of DNA with Mn2+ ions and the molecules of HMGB1. Manganese(2+) 82-86 high mobility group box 1 Homo sapiens 205-210 15612592-13 2004 Thus, structural changes of the DNA double helix in the three-component DNA-HMGB1-Mn2+ complexes come as a result of the combined and interdependent interactions of DNA with Mn2+ ions and the molecules of HMGB1. Manganese(2+) 174-178 high mobility group box 1 Homo sapiens 76-81 15612592-13 2004 Thus, structural changes of the DNA double helix in the three-component DNA-HMGB1-Mn2+ complexes come as a result of the combined and interdependent interactions of DNA with Mn2+ ions and the molecules of HMGB1. Manganese(2+) 174-178 high mobility group box 1 Homo sapiens 205-210 15502843-3 2004 Nicotine, a selective cholinergic agonist, is more efficient than acetylcholine and inhibits HMGB1 release induced by either endotoxin or tumor necrosis factor-alpha (TNF-alpha). Nicotine 0-8 high mobility group box 1 Homo sapiens 93-98 15502843-5 2004 In vivo, treatment with nicotine attenuates serum HMGB1 levels and improves survival in experimental models of sepsis, even when treatment is started after the onset of the disease. Nicotine 24-32 high mobility group box 1 Homo sapiens 50-55 15502843-6 2004 These results reveal acetylcholine as the first known physiological inhibitor of HMGB1 release from human macrophages and suggest that selective nicotinic agonists for the alpha7nAChR might have therapeutic potential for the treatment of sepsis. Acetylcholine 21-34 high mobility group box 1 Homo sapiens 81-86 15130941-6 2004 Amphoterin was secreted from phorbol ester and interferon-gamma (IFN-gamma)-activated macrophages, and the secretion was inhibited by blocking the adenosine 5"-triphosphate (ATP)-binding cassette transporter-1, a member of the multidrug resistance protein family. Phorbol Esters 29-42 high mobility group box 1 Homo sapiens 0-10 15469275-8 2004 Combining the (1)H-PRE data with the crystal structure of the HMGB-1 A-box/cisplatin-modified DNA complex allows one to obtain a semiquantitative estimate of the equilibrium populations at the various sites. Cisplatin 75-84 high mobility group box 1 Homo sapiens 62-68 15130941-6 2004 Amphoterin was secreted from phorbol ester and interferon-gamma (IFN-gamma)-activated macrophages, and the secretion was inhibited by blocking the adenosine 5"-triphosphate (ATP)-binding cassette transporter-1, a member of the multidrug resistance protein family. Adenosine Triphosphate 147-172 high mobility group box 1 Homo sapiens 0-10 15130941-6 2004 Amphoterin was secreted from phorbol ester and interferon-gamma (IFN-gamma)-activated macrophages, and the secretion was inhibited by blocking the adenosine 5"-triphosphate (ATP)-binding cassette transporter-1, a member of the multidrug resistance protein family. Adenosine Triphosphate 174-177 high mobility group box 1 Homo sapiens 0-10 15170359-1 2004 We have recently observed that chromatin architectural protein HMGB1 (previously reported to be involved in numerous biological processes such as DNA replication, recombination, repair, tumor growth, and metastasis) could bind with extremely high affinity (K(d) < 1 pM) to a novel DNA structure that forms a DNA loop maintained at its base by a hemicatenane (hcDNA). hemicatenane 348-360 high mobility group box 1 Homo sapiens 63-68 15274644-5 2004 Transcription stimulation assays using acidic C-tail deletion mutants showed that the five amino acid residues at the C-terminal end of HMGB1, a DDDDE sequence, are essential for the stimulation. dddde 145-150 high mobility group box 1 Homo sapiens 136-141 15274644-6 2004 The DDDDE sequence was also required for the preferential binding of HMGB1 to nucleosome linker DNA, which is a cognate HMGB1 binding site in chromatin. dddde 4-9 high mobility group box 1 Homo sapiens 69-74 15274644-6 2004 The DDDDE sequence was also required for the preferential binding of HMGB1 to nucleosome linker DNA, which is a cognate HMGB1 binding site in chromatin. dddde 4-9 high mobility group box 1 Homo sapiens 120-125 15274644-8 2004 These results strongly suggest that the interaction between the DDDDE sequence of HMGB1 and the H3 N-tail is a key factor for the transcription stimulation by HMGB1 as well as the preferential binding of HMGB1 to chromatin. dddde 64-69 high mobility group box 1 Homo sapiens 82-87 15274644-8 2004 These results strongly suggest that the interaction between the DDDDE sequence of HMGB1 and the H3 N-tail is a key factor for the transcription stimulation by HMGB1 as well as the preferential binding of HMGB1 to chromatin. dddde 64-69 high mobility group box 1 Homo sapiens 159-164 15274644-8 2004 These results strongly suggest that the interaction between the DDDDE sequence of HMGB1 and the H3 N-tail is a key factor for the transcription stimulation by HMGB1 as well as the preferential binding of HMGB1 to chromatin. dddde 64-69 high mobility group box 1 Homo sapiens 159-164 14550538-4 2003 We propose to classify HMGB1 into a new group of proteins unrelated structurally to chemokines but having chemokine-like functions, and to name this class CLF (chemokine-like functions). Chlorine fluoride 155-158 high mobility group box 1 Homo sapiens 23-28 15146429-9 2004 After treatment with high-dose prednisolone, cytoplasmic and extracellular HMGB-1 expression was significantly reduced, coinciding mainly with a decreased number of infiltrating inflammatory cells. Prednisolone 31-43 high mobility group box 1 Homo sapiens 75-81 15161015-1 2004 BACKGROUND: The human high mobility group protein B1 (HMGB1) has attracted considerable interest among oncologists because it sensitises cancer cells to the anticancer drug cisplatin by shielding cisplatin-DNA adducts from nucleotide excision repair. Cisplatin 173-182 high mobility group box 1 Homo sapiens 22-52 15161015-1 2004 BACKGROUND: The human high mobility group protein B1 (HMGB1) has attracted considerable interest among oncologists because it sensitises cancer cells to the anticancer drug cisplatin by shielding cisplatin-DNA adducts from nucleotide excision repair. Cisplatin 173-182 high mobility group box 1 Homo sapiens 54-59 15161015-1 2004 BACKGROUND: The human high mobility group protein B1 (HMGB1) has attracted considerable interest among oncologists because it sensitises cancer cells to the anticancer drug cisplatin by shielding cisplatin-DNA adducts from nucleotide excision repair. Cisplatin 196-205 high mobility group box 1 Homo sapiens 22-52 15161015-1 2004 BACKGROUND: The human high mobility group protein B1 (HMGB1) has attracted considerable interest among oncologists because it sensitises cancer cells to the anticancer drug cisplatin by shielding cisplatin-DNA adducts from nucleotide excision repair. Cisplatin 196-205 high mobility group box 1 Homo sapiens 54-59 14759813-8 2004 When combined with hyperactive transposons and transiently overexpressed HMGB1, a cellular cofactor of SB transposition, hyperactive transposases elevate transposition by almost an order of magnitude compared to the first-generation transposon system. Antimony 103-105 high mobility group box 1 Homo sapiens 73-78 14999020-5 2004 The multiprotein complex including HMGB1 showed ATP hydrolysis and ATP-dependent chromatin structural modulation activities that increased the susceptibility of chromatin to MNase digestion, while HMGB1 alone had no such activity. Adenosine Triphosphate 67-70 high mobility group box 1 Homo sapiens 35-40 15051925-9 2004 Infiltration of PMA-treated U937 monocytes through the KM12SM cell layer was increased by pretreatment of KM12SM cells with amphoterin antisense S-oligodeoxynucleotide exposure. Oligodeoxyribonucleotides 147-167 high mobility group box 1 Homo sapiens 124-134 14632258-4 2004 Multiple transposase binding sites within the terminal inverted repeats, a transpositional enhancer sequence, unequal affinity of the transposase to the binding sites and the activity of the cellular HMGB1 protein all contribute to a highly regulated assembly of SB synaptic complexes, which is likely a requirement for the subsequent catalytic steps. Antimony 263-265 high mobility group box 1 Homo sapiens 200-205 14999020-0 2004 ATP-dependent chromatin structural modulation by multiprotein complex including HMGB1. Adenosine Triphosphate 0-3 high mobility group box 1 Homo sapiens 80-85 14999020-5 2004 The multiprotein complex including HMGB1 showed ATP hydrolysis and ATP-dependent chromatin structural modulation activities that increased the susceptibility of chromatin to MNase digestion, while HMGB1 alone had no such activity. Adenosine Triphosphate 48-51 high mobility group box 1 Homo sapiens 35-40 14534709-7 2003 The in vitro invasive capacity of PC-3, a prostatic cancer cell line that co-expressed amphoterin and RAGE, was suppressed by treatment with amphoterin antisense S-oligodeoxynucleotide (ODN). Oligodeoxyribonucleotides 162-184 high mobility group box 1 Homo sapiens 87-97 14534709-7 2003 The in vitro invasive capacity of PC-3, a prostatic cancer cell line that co-expressed amphoterin and RAGE, was suppressed by treatment with amphoterin antisense S-oligodeoxynucleotide (ODN). Oligodeoxyribonucleotides 162-184 high mobility group box 1 Homo sapiens 141-151 14534709-7 2003 The in vitro invasive capacity of PC-3, a prostatic cancer cell line that co-expressed amphoterin and RAGE, was suppressed by treatment with amphoterin antisense S-oligodeoxynucleotide (ODN). Oligodeoxyribonucleotides 186-189 high mobility group box 1 Homo sapiens 87-97 14534709-7 2003 The in vitro invasive capacity of PC-3, a prostatic cancer cell line that co-expressed amphoterin and RAGE, was suppressed by treatment with amphoterin antisense S-oligodeoxynucleotide (ODN). Oligodeoxyribonucleotides 186-189 high mobility group box 1 Homo sapiens 141-151 14534709-9 2003 The conditioned medium of human prostatic stromal cells deprived of androgen recovered the in vitro invasive capacity of PC-3 cells suppressed by amphoterin antisense S-ODN. Oligodeoxyribonucleotides 167-172 high mobility group box 1 Homo sapiens 146-156 12646658-6 2003 AG490, a specific inhibitor for Janus kinase 2 of the IFN-gamma signaling pathway, dose-dependently attenuated IFN-gamma-induced HMGB1 release. alpha-cyano-(3,4-dihydroxy)-N-benzylcinnamide 0-5 high mobility group box 1 Homo sapiens 129-134 12887690-0 2003 Regulation of expression of sulfoglucuronyl carbohydrate (HNK-1), Amphoterin and RAGE in retinoic acid-differentiated P19 embryonal carcinoma cells. Tretinoin 89-102 high mobility group box 1 Homo sapiens 66-76 12887690-10 2003 RA also upregulated the synthesis of Amphoterin and RAGE in P19 cells. Tretinoin 0-2 high mobility group box 1 Homo sapiens 37-47 12759333-4 2003 In a functional yeast survival screen designed to select new anti-apoptotic mammalian genes, we have identified the chromosomal high-mobility group box-1 protein (HMGB1) as an inhibitor of yeast cell death induced by the pro-apoptotic Bcl-2 family member Bak. bakuchiol 255-258 high mobility group box 1 Homo sapiens 128-153 12759333-4 2003 In a functional yeast survival screen designed to select new anti-apoptotic mammalian genes, we have identified the chromosomal high-mobility group box-1 protein (HMGB1) as an inhibitor of yeast cell death induced by the pro-apoptotic Bcl-2 family member Bak. bakuchiol 255-258 high mobility group box 1 Homo sapiens 163-168 12614161-0 2003 Nature of full-length HMGB1 binding to cisplatin-modified DNA. Cisplatin 39-48 high mobility group box 1 Homo sapiens 22-27 12614161-1 2003 HMGB1, a highly conserved non-histone DNA-binding protein, interacts with specific DNA structural motifs such as those encountered at cisplatin damage, four-way junctions, and supercoils. Cisplatin 134-143 high mobility group box 1 Homo sapiens 0-5 12614161-2 2003 The interaction of full-length HMGB1, containing two tandem HMG box domains and a C-terminal acidic tail, with cisplatin-modified DNA was investigated by hydroxyl radical footprinting and electrophoretic gel mobility shift assays. Cisplatin 111-120 high mobility group box 1 Homo sapiens 31-36 12614161-4 2003 Site-directed mutagenesis of intercalating residues in both HMG domains A and B in full-length HMGB1 further supports the conclusion that only one HMG box domain is bound to the site of cisplatin damage. Cisplatin 186-195 high mobility group box 1 Homo sapiens 95-100 12614161-7 2003 These results illuminate the respective roles of the tandem HMG boxes and the C-terminal acidic tail of HMGB1 in binding to DNA and to the major DNA adducts formed by the anticancer drug cisplatin. Cisplatin 187-196 high mobility group box 1 Homo sapiens 104-109 12450412-7 2002 In the presence of protein HMGB1, shifted reassociation of the strands of DNA fragments containing a tract of the poly(CA).poly(TG) microsatellite leads to the formation of DNA loops maintained at their base by a hemicatenated junction located within the repetitive sequence. poly(tg) 123-131 high mobility group box 1 Homo sapiens 27-32 12534292-10 2003 In addition, the monofunctional adducts of trans-[PtCl(2)(NH(3))(thiazole)] are recognized by HMGB1 domain proteins and removed by the nucleotide excision repair system similarly as the 1,2-GG intrastrand CL of cisplatin. ptcl(2)(nh(3)) 50-64 high mobility group box 1 Homo sapiens 94-99 12534292-10 2003 In addition, the monofunctional adducts of trans-[PtCl(2)(NH(3))(thiazole)] are recognized by HMGB1 domain proteins and removed by the nucleotide excision repair system similarly as the 1,2-GG intrastrand CL of cisplatin. Thiazoles 65-73 high mobility group box 1 Homo sapiens 94-99 12534292-10 2003 In addition, the monofunctional adducts of trans-[PtCl(2)(NH(3))(thiazole)] are recognized by HMGB1 domain proteins and removed by the nucleotide excision repair system similarly as the 1,2-GG intrastrand CL of cisplatin. 1,2-gg 186-192 high mobility group box 1 Homo sapiens 94-99 12534292-10 2003 In addition, the monofunctional adducts of trans-[PtCl(2)(NH(3))(thiazole)] are recognized by HMGB1 domain proteins and removed by the nucleotide excision repair system similarly as the 1,2-GG intrastrand CL of cisplatin. Cisplatin 211-220 high mobility group box 1 Homo sapiens 94-99 12517784-0 2003 A nuclear protein complex containing high mobility group proteins B1 and B2, heat shock cognate protein 70, ERp60, and glyceraldehyde-3-phosphate dehydrogenase is involved in the cytotoxic response to DNA modified by incorporation of anticancer nucleoside analogues. Nucleosides 245-255 high mobility group box 1 Homo sapiens 37-106 12517784-6 2003 These findings indicate that the HMGB1-HMGB2-HSC70-ERp60-glyceraldehyde 3-phosphate dehydrogenase complex detects changes in DNA structure caused by incorporation of nonnatural nucleosides and is a determinant of cell sensitivity to such DNA modifying chemotherapy. Nucleosides 177-188 high mobility group box 1 Homo sapiens 33-38 12468716-1 2002 Using a cloned single domain of the high mobility group protein 1 (HMGB1), we evaluated the effect of introducing metal binding site(s) on protein stability and function. Metals 114-119 high mobility group box 1 Homo sapiens 67-72 12231511-5 2002 However, in keeping with their respective role of early and late inflammatory factors, IL-1beta and HMGB1 respond at different times to different stimuli: IL-1beta secretion is induced earlier by ATP, autocrinally released by monocytes soon after activation; HMGB1 secretion is triggered by lysophosphatidylcholine, generated later in the inflammation site. Adenosine Triphosphate 196-199 high mobility group box 1 Homo sapiens 100-105 12231511-5 2002 However, in keeping with their respective role of early and late inflammatory factors, IL-1beta and HMGB1 respond at different times to different stimuli: IL-1beta secretion is induced earlier by ATP, autocrinally released by monocytes soon after activation; HMGB1 secretion is triggered by lysophosphatidylcholine, generated later in the inflammation site. Lysophosphatidylcholines 291-314 high mobility group box 1 Homo sapiens 100-105 12006575-1 2002 Previously, we and others reported that the high mobility group proteins, HMGB-1/-2, enhance DNA binding in vitro and transactivation in situ by the steroid hormone subgroup of nuclear receptors but did not influence these functions of class II receptors. Steroids 149-164 high mobility group box 1 Homo sapiens 74-83 12358771-1 2002 Sulfoglucuronyl carbohydrate (SGC), reactive with antibody against human natural killer cell antigen, is expressed in several glycolipids, glycoproteins and proteoglycans of the nervous system and has been implicated in cell-cell recognition, neurite outgrowth and neuronal migration during development, through its interaction with SGC-binding protein (SBP) 1. sulfoglucuronyl carbohydrate 0-28 high mobility group box 1 Homo sapiens 333-360 12006575-2 2002 We show here that the DNA binding domain (DBD) is sufficient to account for the selective influence of HMGB-1/-2 on the steroid class of receptors. Steroids 120-127 high mobility group box 1 Homo sapiens 103-112 12006575-4 2002 HMGB-1/-2 interact directly with the DBDs of steroid but not class II receptors, and this interaction requires the CTE. 4,4'-dibenzamido-2,2'-stilbenedisulfonic acid 37-41 high mobility group box 1 Homo sapiens 0-9 12006575-4 2002 HMGB-1/-2 interact directly with the DBDs of steroid but not class II receptors, and this interaction requires the CTE. Steroids 45-52 high mobility group box 1 Homo sapiens 0-9 12006575-4 2002 HMGB-1/-2 interact directly with the DBDs of steroid but not class II receptors, and this interaction requires the CTE. 1,1,1-trifluoro-2-chloroethane 115-118 high mobility group box 1 Homo sapiens 0-9 12006575-5 2002 This in vitro interaction correlates with a requirement of the CTE for maximal HMGB-1/-2 enhancement of DNA binding in vitro and transcriptional activation in cells. 1,1,1-trifluoro-2-chloroethane 63-66 high mobility group box 1 Homo sapiens 79-88 12031669-2 2002 Electrophoretic mobility shift assays show that domains A and B of HMGB1 protein bind to (2R,3R)-diaminobutanedichloroplatinum(II)-generated crosslinks with a higher affinity than to those generated by (2S,3S)-diaminobutanedichloroplatinum(II). (2r,3r)-diaminobutanedichloroplatinum 89-126 high mobility group box 1 Homo sapiens 67-72 12031669-2 2002 Electrophoretic mobility shift assays show that domains A and B of HMGB1 protein bind to (2R,3R)-diaminobutanedichloroplatinum(II)-generated crosslinks with a higher affinity than to those generated by (2S,3S)-diaminobutanedichloroplatinum(II). (2s,3s)-diaminobutanedichloroplatinum 202-239 high mobility group box 1 Homo sapiens 67-72 10652254-4 2000 A role for amphoterin in extracellular matrix-dependent cell regulation is further suggested by the finding that specific decrease of amphoterin mRNA and protein, using antisense oligonucleotides transfected into cells, inhibits cell migration to laminin in a transfilter assay whereas the oligonucleotides in the culture medium have no effect. Oligonucleotides 179-195 high mobility group box 1 Homo sapiens 134-144 11953450-0 2002 N -Glycans on the receptor for advanced glycation end products influence amphoterin binding and neurite outgrowth. n -glycans 0-10 high mobility group box 1 Homo sapiens 73-83 11953450-1 2002 In this study we show that embryonic neurite growth-promoting protein amphoterin binds to carboxylated N -glycans previously identified on mammalian endothelial cells. n -glycans 103-113 high mobility group box 1 Homo sapiens 70-80 11953450-2 2002 Since amphoterin is a ligand for the receptor for advanced glycation end products (RAGE), and the ligand-binding V-domain of the receptor contains two potential N -glycosylation sites, we hypothesized that N -glycans on RAGE may mediate its interactions with amphoterin. Nitrogen 161-162 high mobility group box 1 Homo sapiens 6-16 11953450-2 2002 Since amphoterin is a ligand for the receptor for advanced glycation end products (RAGE), and the ligand-binding V-domain of the receptor contains two potential N -glycosylation sites, we hypothesized that N -glycans on RAGE may mediate its interactions with amphoterin. n -glycans 206-216 high mobility group box 1 Homo sapiens 6-16 11953450-2 2002 Since amphoterin is a ligand for the receptor for advanced glycation end products (RAGE), and the ligand-binding V-domain of the receptor contains two potential N -glycosylation sites, we hypothesized that N -glycans on RAGE may mediate its interactions with amphoterin. n -glycans 206-216 high mobility group box 1 Homo sapiens 259-269 11953450-4 2002 The binding potential of amphoterin to RAGE decreases significantly in presence of soluble carboxylated glycans or when the receptor is deglycosylated. Polysaccharides 104-111 high mobility group box 1 Homo sapiens 25-35 11953450-9 2002 These results indicate that carboxylated N -glycans on RAGE play an important functional role in amphoterin-RAGE-mediated signalling. n -glycans 41-51 high mobility group box 1 Homo sapiens 97-107 11514569-3 2001 Electrophoretic mobility shift assays show that both the A and B domains of HMGB1 as well as TBP discriminate between different platinum-DNA adducts. Platinum 128-136 high mobility group box 1 Homo sapiens 76-81 11514569-4 2001 HMGB1 domain A is the most sensitive to the nature of the spectator ligands on platinum. Platinum 79-87 high mobility group box 1 Homo sapiens 0-5 11514569-7 2001 The effects of HMGB1 overexpression in a BG-1 ovarian cancer cell line, induced by steroid hormones, on the sensitivity of cells treated with [Pt(1R,2R-diaminocyclohexane)Cl(2)] and cis-[Pt(NH(3))(cyclohexylamine)Cl(2)] were also examined. Steroids 83-99 high mobility group box 1 Homo sapiens 15-20 11514569-7 2001 The effects of HMGB1 overexpression in a BG-1 ovarian cancer cell line, induced by steroid hormones, on the sensitivity of cells treated with [Pt(1R,2R-diaminocyclohexane)Cl(2)] and cis-[Pt(NH(3))(cyclohexylamine)Cl(2)] were also examined. pt(1r,2r-diaminocyclohexane)cl(2) 143-176 high mobility group box 1 Homo sapiens 15-20 11514569-8 2001 The results suggest that HMGB1 protein levels influence the cellular processing of cis-[Pt(NH(3))- (cyclohexylamine)](2+), but not [Pt((1R,2R)-diaminocyclohexane)](2+), DNA lesions. pt(nh(3))- (cyclohexylamine) 88-116 high mobility group box 1 Homo sapiens 25-30 11154118-9 2000 Resting platelets treated with PGI2 and forskolin bound to immobilised recombinant amphoterin independently of divalent cations. Epoprostenol 31-35 high mobility group box 1 Homo sapiens 83-93 11154118-9 2000 Resting platelets treated with PGI2 and forskolin bound to immobilised recombinant amphoterin independently of divalent cations. Colforsin 40-49 high mobility group box 1 Homo sapiens 83-93 11154118-11 2000 Amphoterin-binding protein components on the platelet surface were not identified, but amphoterin bound to phosphatidylserine and sulfatide in lipid binding assays. Phosphatidylserines 107-125 high mobility group box 1 Homo sapiens 87-97 11154118-11 2000 Amphoterin-binding protein components on the platelet surface were not identified, but amphoterin bound to phosphatidylserine and sulfatide in lipid binding assays. Sulfoglycosphingolipids 130-139 high mobility group box 1 Homo sapiens 87-97 11154118-13 2000 Interaction of amphoterin with the platelet surface may be mediated by sulfoglycolipids and phospholipids. sulfoglycolipids 71-87 high mobility group box 1 Homo sapiens 15-25 11154118-13 2000 Interaction of amphoterin with the platelet surface may be mediated by sulfoglycolipids and phospholipids. Phospholipids 92-105 high mobility group box 1 Homo sapiens 15-25 11739380-4 2002 In this study we have investigated a possible role of RAGE and amphoterin in the retinoic acid-induced differentiation of neuroblastoma cells. Tretinoin 81-94 high mobility group box 1 Homo sapiens 63-73 11513603-2 2001 HMGB1 consists of 2 DNA binding domains, the HMG boxes A and B, followed by a short basic region and a continuous stretch of 30 glutamate or aspartate residues. Glutamic Acid 128-137 high mobility group box 1 Homo sapiens 0-5 11513603-2 2001 HMGB1 consists of 2 DNA binding domains, the HMG boxes A and B, followed by a short basic region and a continuous stretch of 30 glutamate or aspartate residues. Aspartic Acid 141-150 high mobility group box 1 Homo sapiens 0-5 10652254-4 2000 A role for amphoterin in extracellular matrix-dependent cell regulation is further suggested by the finding that specific decrease of amphoterin mRNA and protein, using antisense oligonucleotides transfected into cells, inhibits cell migration to laminin in a transfilter assay whereas the oligonucleotides in the culture medium have no effect. Oligonucleotides 290-306 high mobility group box 1 Homo sapiens 134-144 9507007-6 1998 The chondroitin sulfate chains on neurocan and phosphacan account for at least 80% of their binding to amphoterin and HB-GAM. Chondroitin Sulfates 4-23 high mobility group box 1 Homo sapiens 103-113 1909331-3 1991 Since the NH2-terminal part of amphoterin is exceptionally rich in lysine residues, we have studied its interactions with plasminogen and tissue plasminogen activator (t-PA). Lysine 67-73 high mobility group box 1 Homo sapiens 31-41 9398056-6 1997 RA stimulated the association of t-PA with the external cell membrane surface, which could be inhibited by heparin sulphate but not by mannose sugars or chelators of divalent cations, consistent with a role for amphoterin. Tretinoin 0-2 high mobility group box 1 Homo sapiens 211-221 9504913-0 1998 The identification of cDNAs that affect the mitosis-to-interphase transition in Schizosaccharomyces pombe, including sbp1, which encodes a spi1p-GTP-binding protein. Guanosine Triphosphate 145-148 high mobility group box 1 Homo sapiens 117-121 1909331-5 1991 In purified systems, both t-PA and plasminogen bound to immobilized amphoterin, and their binding was inhibited by the lysine analogue epsilon-aminocaproic acid. Lysine 119-125 high mobility group box 1 Homo sapiens 68-78 1909331-5 1991 In purified systems, both t-PA and plasminogen bound to immobilized amphoterin, and their binding was inhibited by the lysine analogue epsilon-aminocaproic acid. Aminocaproic Acid 135-160 high mobility group box 1 Homo sapiens 68-78 1909331-9 1991 The results indicate that t-PA and plasminogen form through their lysine-binding sites a complex with amphoterin, which results in acceleration of plasminogen activation and effective degradation of amphoterin. Lysine 66-72 high mobility group box 1 Homo sapiens 102-112 1909331-9 1991 The results indicate that t-PA and plasminogen form through their lysine-binding sites a complex with amphoterin, which results in acceleration of plasminogen activation and effective degradation of amphoterin. Lysine 66-72 high mobility group box 1 Homo sapiens 199-209 33799527-4 2021 In our study, we found that the novel topoisomerase I inhibitor lipotecan (TLC388) can elicit immunogenic cell death (ICD) to release damage-associated molecular patterns (DAMPs), including HMGB1, ANXA1, and CRT exposure. TLC 388 64-73 high mobility group box 1 Homo sapiens 190-195 33773396-2 2021 COVID-19 disease severity is correlated with heightened inflammatory responses, and HMGB1 is an important extracellular mediator in inflammation processes.In this study, we evaluated the effect of HMGB1 inhibitor Glycyrrhizin on the cellular perturbations in lung cells expressing SARS-CoV-2 viral proteins. Glycyrrhizic Acid 213-225 high mobility group box 1 Homo sapiens 197-202 33777021-10 2021 Pretreatment with necrostatin-1 significantly reduced the expression of RIP1, RIP3 and MLKL as well as PGAM5, DRP1 and HMGB1, which subsequently led to obvious attenuation of hepatic inflammation and damage. necrostatin-1 18-31 high mobility group box 1 Homo sapiens 119-124 33799527-4 2021 In our study, we found that the novel topoisomerase I inhibitor lipotecan (TLC388) can elicit immunogenic cell death (ICD) to release damage-associated molecular patterns (DAMPs), including HMGB1, ANXA1, and CRT exposure. TLC 388 75-81 high mobility group box 1 Homo sapiens 190-195 34808298-12 2022 It was found that ZJP has a down-regulating effect on inflammatory reaction and could inhibit the relative mRNA and protein expression of JMJD2B/COX-2/VEGF axis and HMGB1/NF-kappaB signaling pathway in vivo and in vitro, including JMJD2B, COX-2, VEGF, VEGFR1, and VEGFR2, which in turn reduced the damage of gastric mucosal cells. zjp 18-21 high mobility group box 1 Homo sapiens 165-170 33817244-6 2020 The binding relationship between miR-204-5p and LINC00704 or HMGB1 was predicted by LncBase Predicted v.2 or TargetScan, respectively, and then validated by dual luciferase reporter assay. mir-204-5p 33-43 high mobility group box 1 Homo sapiens 61-66 33817244-10 2020 Meanwhile, miR-204-5p overexpression repressed proliferation, migration, and invasion by targeting HMGB1. mir-204-5p 11-21 high mobility group box 1 Homo sapiens 99-104 33822516-8 2021 After cells were co-transfected with pcDNA-HMGB1 and pc-DNA-ctrl, we investigated apoptosis and autophagy in miR-451 overexpressed cells perturbed by exogenous HMGB1 through MTT, flow cytometry, and Western blot. monooxyethylene trimethylolpropane tristearate 174-177 high mobility group box 1 Homo sapiens 160-165 25489880-12 2015 Immunofluorescence showed enhanced reactive astrocytes, microglial activation, and HMGB1 expression localized to the injured cortex of TBI/alcohol as compared to TBI/air animals. Alcohols 139-146 high mobility group box 1 Homo sapiens 83-88 22895517-5 2012 Stimulation by hydrogen peroxide, or the TLR4 ligands recombinant human high-mobility group protein B1 or lipopolysaccharide, induced PTX3 expression in the Mile Sven 1 endothelial cell line and in primary renal endothelial cells, suggesting that endothelial PTX3 was induced by pathways involving TLR4 and reactive oxygen species. Reactive Oxygen Species 307-330 high mobility group box 1 Homo sapiens 72-102 34808298-13 2022 CONCLUSIONS: The results suggested that ZJP exerts therapeutic effects on H. pylori-induced CAG by inhibiting the JMJD2B/COX-2/VEGF axis and HMGB1/NF-kappaB signaling pathway. zjp 40-43 high mobility group box 1 Homo sapiens 141-146 34427852-5 2022 Our results showed that GA significantly attenuated LPS-induced ALI and decreased the production of inflammatory factors, including IL-1beta, MCP-1, COX2, HMGB1, and adhesion molecules, such as E-selectin, VCAM-1, and modulated expression of angiotensin-converting enzyme 2 (ACE2). Glycyrrhizic Acid 24-26 high mobility group box 1 Homo sapiens 155-160 34973275-0 2022 miR-142-3p simultaneously targets HMGA1, HMGA2, HMGB1, and HMGB3 and inhibits tumorigenic properties and in-vivo metastatic potential of human cervical cancer cells. mir-142-3p 0-10 high mobility group box 1 Homo sapiens 48-53 34363018-0 2022 Lactate promotes macrophage HMGB1 lactylation, acetylation, and exosomal release in polymicrobial sepsis. Lactic Acid 0-7 high mobility group box 1 Homo sapiens 28-33 34971702-5 2022 Interestingly, inhibition of HMGB1 secretion by ethyl pyruvate (EP) enhanced viral propagation while silencing of HMGB1 resulted in abrogated viral replication in DENV-2 infected A549 cells. ethyl pyruvate 48-62 high mobility group box 1 Homo sapiens 29-34 34971702-5 2022 Interestingly, inhibition of HMGB1 secretion by ethyl pyruvate (EP) enhanced viral propagation while silencing of HMGB1 resulted in abrogated viral replication in DENV-2 infected A549 cells. ethyl pyruvate 64-66 high mobility group box 1 Homo sapiens 29-34 34363018-2 2022 However, it is unclear whether lactate could promote HMGB1 release during sepsis. Lactic Acid 31-38 high mobility group box 1 Homo sapiens 53-58 34363018-3 2022 The present study demonstrated a novel role of lactate in HMGB1 lactylation and acetylation in macrophages during polymicrobial sepsis. Lactic Acid 47-54 high mobility group box 1 Homo sapiens 58-63 34363018-4 2022 We found that macrophages can uptake extracellular lactate via monocarboxylate transporters (MCTs) to promote HMGB1 lactylation via a p300/CBP-dependent mechanism. Lactic Acid 51-58 high mobility group box 1 Homo sapiens 110-115 34363018-5 2022 We also observed that lactate stimulates HMGB1 acetylation by Hippo/YAP-mediated suppression of deacetylase SIRT1 and beta-arrestin2-mediated recruitment of acetylases p300/CBP to the nucleus via G protein-coupled receptor 81 (GPR81). Lactic Acid 22-29 high mobility group box 1 Homo sapiens 41-46 34363018-7 2022 In vivo reduction of lactate production and/or inhibition of GPR81-mediated signaling decreases circulating exosomal HMGB1 levels and improves survival outcome in polymicrobial sepsis. Lactic Acid 21-28 high mobility group box 1 Homo sapiens 117-122 34696918-7 2022 Treatment with H2O2 + VD decreased gene/protein expression of NLRP1, NLRP3, caspase-1, HMGB1, IL-1beta, TNF-alpha and IL-18 in PE and NT explants with H2O2. Hydrogen Peroxide 15-19 high mobility group box 1 Homo sapiens 87-92 34696918-7 2022 Treatment with H2O2 + VD decreased gene/protein expression of NLRP1, NLRP3, caspase-1, HMGB1, IL-1beta, TNF-alpha and IL-18 in PE and NT explants with H2O2. Hydrogen Peroxide 151-155 high mobility group box 1 Homo sapiens 87-92 34696918-6 2022 Placental explants from NT cultured with H2O2 showed increased gene and protein expression of NLRP1, NLRP3, caspase-1, IL-1beta, TNF-alpha and HMGB1, while H2O2 was also able to increase TNF-alpha and caspase-1 gene expression in PE. Hydrogen Peroxide 41-45 high mobility group box 1 Homo sapiens 143-148 34934826-2 2021 Glycyrrhizin (GL), a traditional Chinese medicine for liver disease, binds to HMGB1, thereby inhibits tissue injury. Glycyrrhizic Acid 0-12 high mobility group box 1 Homo sapiens 78-83 34933679-3 2021 However, the mechanism how extracellular HMGB1 acts on AL cells and leads to chemoresistance remains elusive. Aluminum 55-57 high mobility group box 1 Homo sapiens 41-46 34916485-0 2021 An RNA-RNA crosstalk network involving HMGB1 and RICTOR facilitates hepatocellular carcinoma tumorigenesis by promoting glutamine metabolism and impedes immunotherapy by PD-L1+ exosomes activity. Glutamine 120-129 high mobility group box 1 Homo sapiens 39-44 34916485-8 2021 Mechanistically, the HMGB1-driven RNA-RNA crosstalk network facilitated HCC cell glutamine metabolism via dual mechanisms, activating a positive feedback loop involving mTORC2-AKT-C-MYC to upregulate glutamine synthetase (GS) expression, and inducing mTORC1 signaling to derepress SIRT4 on glutamate dehydrogenase (GDH). Glutamine 81-90 high mobility group box 1 Homo sapiens 21-26 34906140-11 2021 CONCLUSION: Wogonin alleviated the inflammation and apoptosis in LPS-induced A549 cells by SIRT1-mediated HMGB1 deacetylation, which might represent the identification of a novel mechanism by which wogonin exerts protective effects on ALI and provide ideas for the application of wogonin to ALI treatment. wogonin 12-19 high mobility group box 1 Homo sapiens 106-111 34906140-11 2021 CONCLUSION: Wogonin alleviated the inflammation and apoptosis in LPS-induced A549 cells by SIRT1-mediated HMGB1 deacetylation, which might represent the identification of a novel mechanism by which wogonin exerts protective effects on ALI and provide ideas for the application of wogonin to ALI treatment. wogonin 198-205 high mobility group box 1 Homo sapiens 106-111 34966671-4 2021 The High-Mobility Group Box 1 (HMGB1) is a multifunctional protein involved in intrinsic resistance to cisplatin. Cisplatin 103-112 high mobility group box 1 Homo sapiens 4-29 34966671-4 2021 The High-Mobility Group Box 1 (HMGB1) is a multifunctional protein involved in intrinsic resistance to cisplatin. Cisplatin 103-112 high mobility group box 1 Homo sapiens 31-36 34966671-5 2021 HMGB1 is released when cytotoxic agents, such as cisplatin, induce cell death. Cisplatin 49-58 high mobility group box 1 Homo sapiens 0-5 34966671-9 2021 Subcellular localization of HMGB1 was detected in the cell lines and in viable cells after a single exposure to cisplatin, which are designated as cisplatin-persistent cells. Cisplatin 112-121 high mobility group box 1 Homo sapiens 28-33 34966671-9 2021 Subcellular localization of HMGB1 was detected in the cell lines and in viable cells after a single exposure to cisplatin, which are designated as cisplatin-persistent cells. Cisplatin 147-156 high mobility group box 1 Homo sapiens 28-33 34966671-12 2021 Cisplatin-susceptible lung adenocarcinoma cell lines died by apoptosis or necrosis and released HMGB1. Cisplatin 0-9 high mobility group box 1 Homo sapiens 96-101 34966671-13 2021 In cisplatin-persistent cells, nuclear relocalization of HMGB1 and overexpression of HMGB1 and RAGE, but not TLR-2 or TLR-4, were observed. Cisplatin 3-12 high mobility group box 1 Homo sapiens 57-62 34966671-13 2021 In cisplatin-persistent cells, nuclear relocalization of HMGB1 and overexpression of HMGB1 and RAGE, but not TLR-2 or TLR-4, were observed. Cisplatin 3-12 high mobility group box 1 Homo sapiens 85-90 34966671-14 2021 In tumor cells, this HMGB1-RAGE interaction may be associated with the development of cisplatin resistance. Cisplatin 86-95 high mobility group box 1 Homo sapiens 21-26 34850958-0 2022 Corilagin induces apoptosis and inhibits autophagy of HL-60 cells by regulating miR-451/HMGB1 axis. corilagin 0-9 high mobility group box 1 Homo sapiens 88-93 34850958-11 2022 In conclusion, these results indicated that corilagin induced apoptosis and inhibited autophagy in HL-60 cells by regulating the miR-451/HMGB1 axis, and corilagin may be a novel therapeutic drug for the treatment of AML. corilagin 44-53 high mobility group box 1 Homo sapiens 137-142 34755645-0 2021 Indoprofen exerts a potent therapeutic effect against sepsis by alleviating high mobility group box 1-mediated inflammatory responses. Indoprofen 0-10 high mobility group box 1 Homo sapiens 76-101 34755645-2 2021 This study was carried out to examine the inhibitory effect of indoprofen on high mobility group box 1 (HMGB1)-mediated inflammatory responses in vivo and in vitro. Indoprofen 63-73 high mobility group box 1 Homo sapiens 77-102 34755645-2 2021 This study was carried out to examine the inhibitory effect of indoprofen on high mobility group box 1 (HMGB1)-mediated inflammatory responses in vivo and in vitro. Indoprofen 63-73 high mobility group box 1 Homo sapiens 104-109 34755645-5 2021 Mechanistically, indoprofen potently inhibited the release of HMGB1 following stimulation by lipopolysaccharide (LPS) or polyinosinic:polycytidylic acid (poly I:C), and suppressed recombinant human HMGB1(rhHMGB1)-induced inflammatory responses. Indoprofen 17-27 high mobility group box 1 Homo sapiens 62-67 34755645-5 2021 Mechanistically, indoprofen potently inhibited the release of HMGB1 following stimulation by lipopolysaccharide (LPS) or polyinosinic:polycytidylic acid (poly I:C), and suppressed recombinant human HMGB1(rhHMGB1)-induced inflammatory responses. Indoprofen 17-27 high mobility group box 1 Homo sapiens 198-203 34755645-5 2021 Mechanistically, indoprofen potently inhibited the release of HMGB1 following stimulation by lipopolysaccharide (LPS) or polyinosinic:polycytidylic acid (poly I:C), and suppressed recombinant human HMGB1(rhHMGB1)-induced inflammatory responses. polyinosinic 121-133 high mobility group box 1 Homo sapiens 62-67 34755645-5 2021 Mechanistically, indoprofen potently inhibited the release of HMGB1 following stimulation by lipopolysaccharide (LPS) or polyinosinic:polycytidylic acid (poly I:C), and suppressed recombinant human HMGB1(rhHMGB1)-induced inflammatory responses. Poly C 134-152 high mobility group box 1 Homo sapiens 62-67 34755645-5 2021 Mechanistically, indoprofen potently inhibited the release of HMGB1 following stimulation by lipopolysaccharide (LPS) or polyinosinic:polycytidylic acid (poly I:C), and suppressed recombinant human HMGB1(rhHMGB1)-induced inflammatory responses. Poly I-C 154-162 high mobility group box 1 Homo sapiens 62-67 34755645-6 2021 It was also found that indoprofen has both cyclooxygenase 2-dependent and -independent inhibitory effects on the proinflammatory effect of HMGB1 in THP-1 cells. Indoprofen 23-33 high mobility group box 1 Homo sapiens 139-144 34755645-8 2021 Collectively, these data demonstrated that the anti-inflammatory effect of indoprofen in sepsis was associated with HMGB1-mediated inflammatory responses, thus offering a favorable mechanistic basis to support the therapeutic potential of indoprofen for the treatment of lethal sepsis or other inflammatory diseases. Indoprofen 75-85 high mobility group box 1 Homo sapiens 116-121 34755645-8 2021 Collectively, these data demonstrated that the anti-inflammatory effect of indoprofen in sepsis was associated with HMGB1-mediated inflammatory responses, thus offering a favorable mechanistic basis to support the therapeutic potential of indoprofen for the treatment of lethal sepsis or other inflammatory diseases. Indoprofen 239-249 high mobility group box 1 Homo sapiens 116-121 34917229-0 2021 Folic Acid Protects Melanocytes from Oxidative Stress via Activation of Nrf2 and Inhibition of HMGB1. Folic Acid 0-10 high mobility group box 1 Homo sapiens 95-100 34934826-2 2021 Glycyrrhizin (GL), a traditional Chinese medicine for liver disease, binds to HMGB1, thereby inhibits tissue injury. Glycyrrhizic Acid 14-16 high mobility group box 1 Homo sapiens 78-83 34101714-13 2021 Furthermore, dexmedetomidine induced a significant reduction in peri-operative serum HMGB1 level from the baseline (222.5 +- 408.3 pg ml-1) to the first postoperative day (152.2 +- 280.0 pg ml-1) P = 0.0033. Dexmedetomidine 13-28 high mobility group box 1 Homo sapiens 85-90 34477479-7 2021 By analyzing the expression of High-Mobility Group Box 1 (HMGB1) and RAGE in HSC-4 cells, the result showed that EVO slightly reduced HMBG1 levels and dramatically decreased RAGE levels, while RAGE overexpression did have no marked influences on HMBG1 levels. evodiamine 113-116 high mobility group box 1 Homo sapiens 31-56 34477479-7 2021 By analyzing the expression of High-Mobility Group Box 1 (HMGB1) and RAGE in HSC-4 cells, the result showed that EVO slightly reduced HMBG1 levels and dramatically decreased RAGE levels, while RAGE overexpression did have no marked influences on HMBG1 levels. evodiamine 113-116 high mobility group box 1 Homo sapiens 58-63 34943830-0 2021 Post-Translational Modification of HMGB1 Disulfide Bonds in Stimulating and Inhibiting Inflammation. Disulfides 41-50 high mobility group box 1 Homo sapiens 35-40 34737812-11 2021 Mechanistically, the upregulation of HMGB1/RAGE expression induced by LPS was markedly blocked by anagliptin. anagliptin 98-108 high mobility group box 1 Homo sapiens 37-42 34737812-12 2021 In conclusion, anagliptin alleviated inflammation, apoptosis and endothelial dysfunction in LPS-induced HPMVECs via modulating HMGB1/RAGE expression. anagliptin 15-25 high mobility group box 1 Homo sapiens 127-132 34338955-9 2021 MIR3142HG impaired miR-450b-5p-mediated inhibition of HMGB1. mir-450b 19-27 high mobility group box 1 Homo sapiens 54-59 34338955-9 2021 MIR3142HG impaired miR-450b-5p-mediated inhibition of HMGB1. CHEMBL3740941 28-30 high mobility group box 1 Homo sapiens 54-59 34815344-0 2021 BAP1 forms a trimer with HMGB1 and HDAC1 that modulates gene x environment interaction with asbestos. Asbestos 92-100 high mobility group box 1 Homo sapiens 25-30 34943830-2 2021 As reviewed here, HMGB1 is an oxidation-reduction sensitive DAMP bearing three cysteines, and the post-translational modification of these residues establishes its proinflammatory and anti-inflammatory activities by binding to different extracellular cell surface receptors. Cysteine 79-88 high mobility group box 1 Homo sapiens 18-23 34867338-7 2021 Then, we used the HMGB1 inhibitor glycyrrhizin to reverse the effects of HMGB1 on C2C12 myotubes. Glycyrrhizic Acid 34-46 high mobility group box 1 Homo sapiens 18-23 34916197-0 2021 (Aloin inhibits lactate-induced proliferation and migration of gastric cancer cells by downregulating HMGB1 expression). alloin 1-6 high mobility group box 1 Homo sapiens 102-107 34916197-0 2021 (Aloin inhibits lactate-induced proliferation and migration of gastric cancer cells by downregulating HMGB1 expression). Lactic Acid 16-23 high mobility group box 1 Homo sapiens 102-107 34916197-5 2021 HMGB1 expression was knocked down in BGC-823 cells using RNA interference technique, and the effects of HMGB1 knockdown on proliferation and migration of the cells stimulated with lactate for 24 h were examined using EdU and wound healing assays. Lactic Acid 180-187 high mobility group box 1 Homo sapiens 104-109 34916197-7 2021 Treatment with lactate obviously up-regulated the expressions of cyclin D1, cyclin E1, PCNA, N-cadherin, MMP-2, MMP-9 and HMGB1, which were significantly inhibited by aloin; aloin significantly reversed inhibition of E-cadherin and blocked the release of HMGB1 in lactate-treated cells. Lactic Acid 15-22 high mobility group box 1 Homo sapiens 122-127 34916197-7 2021 Treatment with lactate obviously up-regulated the expressions of cyclin D1, cyclin E1, PCNA, N-cadherin, MMP-2, MMP-9 and HMGB1, which were significantly inhibited by aloin; aloin significantly reversed inhibition of E-cadherin and blocked the release of HMGB1 in lactate-treated cells. Lactic Acid 15-22 high mobility group box 1 Homo sapiens 255-260 34916197-7 2021 Treatment with lactate obviously up-regulated the expressions of cyclin D1, cyclin E1, PCNA, N-cadherin, MMP-2, MMP-9 and HMGB1, which were significantly inhibited by aloin; aloin significantly reversed inhibition of E-cadherin and blocked the release of HMGB1 in lactate-treated cells. alloin 167-172 high mobility group box 1 Homo sapiens 122-127 34916197-7 2021 Treatment with lactate obviously up-regulated the expressions of cyclin D1, cyclin E1, PCNA, N-cadherin, MMP-2, MMP-9 and HMGB1, which were significantly inhibited by aloin; aloin significantly reversed inhibition of E-cadherin and blocked the release of HMGB1 in lactate-treated cells. alloin 167-172 high mobility group box 1 Homo sapiens 255-260 34916197-7 2021 Treatment with lactate obviously up-regulated the expressions of cyclin D1, cyclin E1, PCNA, N-cadherin, MMP-2, MMP-9 and HMGB1, which were significantly inhibited by aloin; aloin significantly reversed inhibition of E-cadherin and blocked the release of HMGB1 in lactate-treated cells. alloin 174-179 high mobility group box 1 Homo sapiens 122-127 34916197-7 2021 Treatment with lactate obviously up-regulated the expressions of cyclin D1, cyclin E1, PCNA, N-cadherin, MMP-2, MMP-9 and HMGB1, which were significantly inhibited by aloin; aloin significantly reversed inhibition of E-cadherin and blocked the release of HMGB1 in lactate-treated cells. alloin 174-179 high mobility group box 1 Homo sapiens 255-260 34916197-7 2021 Treatment with lactate obviously up-regulated the expressions of cyclin D1, cyclin E1, PCNA, N-cadherin, MMP-2, MMP-9 and HMGB1, which were significantly inhibited by aloin; aloin significantly reversed inhibition of E-cadherin and blocked the release of HMGB1 in lactate-treated cells. Lactic Acid 264-271 high mobility group box 1 Homo sapiens 255-260 34916197-8 2021 BGC-823 cells with HMGB1 knockdown, as compared with the cells transfected with the negative control plasmid, showed significantly lowered proliferation and migration abilities following lactate treatment. Lactic Acid 187-194 high mobility group box 1 Homo sapiens 19-24 34916197-9 2021 CONCLUSION: Aloin inhibits lactate-induced proliferation and migration of gastric cancer cells by down-regulating the expression and release of HMGB1 and the expressions of proliferation- and migration-related proteins. alloin 12-17 high mobility group box 1 Homo sapiens 144-149 34916197-9 2021 CONCLUSION: Aloin inhibits lactate-induced proliferation and migration of gastric cancer cells by down-regulating the expression and release of HMGB1 and the expressions of proliferation- and migration-related proteins. Lactic Acid 27-34 high mobility group box 1 Homo sapiens 144-149 34867338-7 2021 Then, we used the HMGB1 inhibitor glycyrrhizin to reverse the effects of HMGB1 on C2C12 myotubes. Glycyrrhizic Acid 34-46 high mobility group box 1 Homo sapiens 73-78 34727314-7 2021 Both age (r = 0.210, p = 0.001), serum creatinine (r = 0.509, p < 0.001), eGFR (r = - 0.459, p < 0.001) and Hcy (r = 0.531, p < 0.001) were significantly correlated with HMGB1. Creatinine 39-49 high mobility group box 1 Homo sapiens 171-176 34403663-3 2021 This study aimed to address the effect of miR-141-3p on astrocyte activation and inflammatory response in BM through HMGB1. mir-141-3p 42-52 high mobility group box 1 Homo sapiens 117-122 34939761-9 2021 Sequential exposure of the cells to 5-aza and Alimta enhanced miR-34a expression and significantly downregulated HMGB1, HMGA2 and BCL-2 expressions. Azacitidine 36-41 high mobility group box 1 Homo sapiens 113-118 34939761-9 2021 Sequential exposure of the cells to 5-aza and Alimta enhanced miR-34a expression and significantly downregulated HMGB1, HMGA2 and BCL-2 expressions. Pemetrexed 46-52 high mobility group box 1 Homo sapiens 113-118 34939761-11 2021 Caspase-3 activation, HMGB1 release into extracellular space and staining of the cells with annexine V/PI suggested that 5-aza reduced late apoptotic/necrotic cell death induced by Alimta. Pemetrexed 181-187 high mobility group box 1 Homo sapiens 22-27 34543830-8 2021 The CSF HMGB1 concentrations were significantly higher in the DRE and NDE groups compared with the ONNDs group (p < 0.001). NDE 70-73 high mobility group box 1 Homo sapiens 8-13 34657898-7 2022 Correlation analysis indicated that the levels of HMGB1 and TLR4 were positively correlated with triglyceride (TG), fasting plasma glucose (FPG) levels and BMI, whereas a negative correlation between HMGB1 and high density lipoprotein (HDL) was noted. Triglycerides 111-113 high mobility group box 1 Homo sapiens 50-55 34164831-3 2021 The activation of HMGB1 secretion involves viral induction of reactive oxygen species and is mediated by the p38/STAT1 axis. Oxygen 71-77 high mobility group box 1 Homo sapiens 18-23 34619434-2 2021 Dociparstat sodium (DSTAT, CX-01) is a low anticoagulant heparin with multiple mechanisms of action, including inhibition of the CXCR4/CXCL12 axis, blocking HMGB1, and binding platelet factor 4 (PF-4). GM 1892 0-18 high mobility group box 1 Homo sapiens 157-162 34619434-2 2021 Dociparstat sodium (DSTAT, CX-01) is a low anticoagulant heparin with multiple mechanisms of action, including inhibition of the CXCR4/CXCL12 axis, blocking HMGB1, and binding platelet factor 4 (PF-4). GM 1892 20-25 high mobility group box 1 Homo sapiens 157-162 34351732-7 2021 We discovered that HSoligo 7 (HS oligosaccharide 7), a 3-O-sulfated octasaccharide, binds to high mobility group box 1 protein (HMGB1) and tau protein, both believed to be involved in the process of inflammation. Oligosaccharides 33-48 high mobility group box 1 Homo sapiens 93-126 34351732-7 2021 We discovered that HSoligo 7 (HS oligosaccharide 7), a 3-O-sulfated octasaccharide, binds to high mobility group box 1 protein (HMGB1) and tau protein, both believed to be involved in the process of inflammation. Oligosaccharides 33-48 high mobility group box 1 Homo sapiens 128-133 34351732-7 2021 We discovered that HSoligo 7 (HS oligosaccharide 7), a 3-O-sulfated octasaccharide, binds to high mobility group box 1 protein (HMGB1) and tau protein, both believed to be involved in the process of inflammation. Octasaccharide 68-82 high mobility group box 1 Homo sapiens 93-126 34351732-7 2021 We discovered that HSoligo 7 (HS oligosaccharide 7), a 3-O-sulfated octasaccharide, binds to high mobility group box 1 protein (HMGB1) and tau protein, both believed to be involved in the process of inflammation. Octasaccharide 68-82 high mobility group box 1 Homo sapiens 128-133 34778055-0 2021 HMGB1 Promotes Resistance to Doxorubicin in Human Hepatocellular Carcinoma Cells by Inducing Autophagy via the AMPK/mTOR Signaling Pathway. Doxorubicin 29-40 high mobility group box 1 Homo sapiens 0-5 34778055-4 2021 In the present study, we found that administration of doxorubicin (DOX) significantly promoted HMGB1 expression and induced HMGB1 cytoplasmic translocation in human HCC cell lines BEL7402 and SMMC7721, which enhanced autophagy that contributes to protecting HCC cells from apoptosis and increasing drug resistance. Doxorubicin 54-65 high mobility group box 1 Homo sapiens 95-100 34778055-4 2021 In the present study, we found that administration of doxorubicin (DOX) significantly promoted HMGB1 expression and induced HMGB1 cytoplasmic translocation in human HCC cell lines BEL7402 and SMMC7721, which enhanced autophagy that contributes to protecting HCC cells from apoptosis and increasing drug resistance. Doxorubicin 54-65 high mobility group box 1 Homo sapiens 124-129 34778055-4 2021 In the present study, we found that administration of doxorubicin (DOX) significantly promoted HMGB1 expression and induced HMGB1 cytoplasmic translocation in human HCC cell lines BEL7402 and SMMC7721, which enhanced autophagy that contributes to protecting HCC cells from apoptosis and increasing drug resistance. Doxorubicin 67-70 high mobility group box 1 Homo sapiens 95-100 34778055-4 2021 In the present study, we found that administration of doxorubicin (DOX) significantly promoted HMGB1 expression and induced HMGB1 cytoplasmic translocation in human HCC cell lines BEL7402 and SMMC7721, which enhanced autophagy that contributes to protecting HCC cells from apoptosis and increasing drug resistance. Doxorubicin 67-70 high mobility group box 1 Homo sapiens 124-129 34778055-5 2021 Moreover, we observed HMGB1 translocation and elevation of autophagy in DOX-resistant BEL7402 and SMMC7721 cells. Doxorubicin 72-75 high mobility group box 1 Homo sapiens 22-27 34778055-6 2021 Additionally, inhibition of HMGB1 and autophagy increased the sensitivities of BEL-7402 and SMMC-7721 cells to DOX and re-sensitized their DOX-resistant cells. Doxorubicin 111-114 high mobility group box 1 Homo sapiens 28-33 34778055-8 2021 In summary, our results indicate that HMGB1 promotes acquired DOX resistance in DOX-treated BEL7402 and SMMC7721 cells by enhancing autophagy through the AMPK/mTOR signaling pathway. Doxorubicin 62-65 high mobility group box 1 Homo sapiens 38-43 34778055-8 2021 In summary, our results indicate that HMGB1 promotes acquired DOX resistance in DOX-treated BEL7402 and SMMC7721 cells by enhancing autophagy through the AMPK/mTOR signaling pathway. Doxorubicin 80-83 high mobility group box 1 Homo sapiens 38-43 34914264-1 2021 Objective: To investigate the effect of silencing the high-mobility group box-1 protein (HMGB1) combined with docetaxel (DTX) on the proliferation and apoptosis of PCa cells and its possible action mechanism. Docetaxel 121-124 high mobility group box 1 Homo sapiens 54-79 34914264-10 2021 The expression of the HMGB1 protein was markedly lower in the si-HMGB1+DTX than in the DTX group (P < 0.05). Docetaxel 87-90 high mobility group box 1 Homo sapiens 22-27 34914264-11 2021 CONCLUSIONS: Silencing HMGB1 combined with DTX chemotherapy can inhibit the proliferation and promote the apoptosis of PCa cells, which may be attributed to its regulatory effect on the expressions of the Bcl-2 family-related proteins. Docetaxel 43-46 high mobility group box 1 Homo sapiens 23-28 34657898-7 2022 Correlation analysis indicated that the levels of HMGB1 and TLR4 were positively correlated with triglyceride (TG), fasting plasma glucose (FPG) levels and BMI, whereas a negative correlation between HMGB1 and high density lipoprotein (HDL) was noted. Triglycerides 97-109 high mobility group box 1 Homo sapiens 50-55 34657898-7 2022 Correlation analysis indicated that the levels of HMGB1 and TLR4 were positively correlated with triglyceride (TG), fasting plasma glucose (FPG) levels and BMI, whereas a negative correlation between HMGB1 and high density lipoprotein (HDL) was noted. Glucose 131-138 high mobility group box 1 Homo sapiens 50-55 34627452-2 2021 METHODS: Different concentrations of recombinant human HMGB1 protein (100, 200, 400, 800 and 1000 ng/ml) were incubated with MSC for 24, 48, 72 h and the proliferation of MSC were detected respectively by using the CCK-8 method and flow cytometry. Sincalide 215-220 high mobility group box 1 Homo sapiens 55-60 34546745-4 2021 Using synthetic protein chemistry, we generated site-specifically O-GlcNAc-modified HMGB1 at S100 and characterized biochemically the effect of the sugar modification on its DNA binding activity. Sugars 148-153 high mobility group box 1 Homo sapiens 84-89 34671276-2 2021 Salicylic acid can bind to human high mobility group box 1 (HMGB1) and interrupt its role in mediating immune responses. Salicylic Acid 0-14 high mobility group box 1 Homo sapiens 33-58 34671276-2 2021 Salicylic acid can bind to human high mobility group box 1 (HMGB1) and interrupt its role in mediating immune responses. Salicylic Acid 0-14 high mobility group box 1 Homo sapiens 60-65 34639171-8 2021 Cardiomyocytes exposed to 25mM glucose resulted in the downregulation of HSP60 and SIRT1 after 48 h. We further examined that hyperglycemia mediated the decrease in the gap junction protein CX43, as well as CXC chemokine receptor CXCR4 which may affect the physiological functions of the cardiomyocytes when exposed to high glucose for 24 and 48 h. Upregulated expression of DNA-binding nuclear protein HMGB1, along with changes in histone methylation marker H3K9me1 have demonstrated hyperglycemia-induced damage to cardiomyocyte at 24 h of exposure. Glucose 31-38 high mobility group box 1 Homo sapiens 403-408 34639171-8 2021 Cardiomyocytes exposed to 25mM glucose resulted in the downregulation of HSP60 and SIRT1 after 48 h. We further examined that hyperglycemia mediated the decrease in the gap junction protein CX43, as well as CXC chemokine receptor CXCR4 which may affect the physiological functions of the cardiomyocytes when exposed to high glucose for 24 and 48 h. Upregulated expression of DNA-binding nuclear protein HMGB1, along with changes in histone methylation marker H3K9me1 have demonstrated hyperglycemia-induced damage to cardiomyocyte at 24 h of exposure. Glucose 324-331 high mobility group box 1 Homo sapiens 403-408 34617194-11 2022 HMGB1 inhibitor, glycyrrhizin, abolishes all the observed proangiogenic phenotypes. Glycyrrhizic Acid 17-29 high mobility group box 1 Homo sapiens 0-5 34739475-0 2021 HMGB1-antagonism exerted by glycyrrhizin could be fruitful against COVID-19. Glycyrrhizic Acid 28-40 high mobility group box 1 Homo sapiens 0-5 34739475-5 2021 As HMGB1 is antagonized by glycyrrhizin, this substance could be potentially useful as ancillary treatment in COVID-19. Glycyrrhizic Acid 27-39 high mobility group box 1 Homo sapiens 3-8 34573077-0 2021 Verbascoside Protects Gingival Cells against High Glucose-Induced Oxidative Stress via PKC/HMGB1/RAGE/NFkappaB Pathway. acteoside 0-12 high mobility group box 1 Homo sapiens 91-96 34583662-0 2021 Overweight with HBV infection limited the efficacy of TACE in hepatocellular carcinoma by inhibiting the upregulated HMGB1. Chlorotrianisene 54-58 high mobility group box 1 Homo sapiens 117-122 34583662-10 2021 Irrespective of TACE, the average expression of HMGB1 in hepatitis or obesity patients was higher than that in normal individuals and did not show upregulation after TACE. Chlorotrianisene 16-20 high mobility group box 1 Homo sapiens 48-53 34583662-11 2021 Patients without overweight or HBV infection had a low expression of serum HMGB1 that was substantially upregulated after TACE. Chlorotrianisene 122-126 high mobility group box 1 Homo sapiens 75-80 34583662-13 2021 Thus, HBV and BMI maybe two factors affecting the efficacy of TACE via upregulated HMGB1. Chlorotrianisene 62-66 high mobility group box 1 Homo sapiens 83-88 34638242-4 2021 Anthracycline and taxane treatments cause ICD and immunogenic modulations, resulting in the activation of antitumor immunity through damage-associated molecular patterns (DAMPs), such as adenosine triphosphate, calreticulin, high mobility group box 1, heat shock proteins 70/90, and annexin A1. Anthracyclines 0-13 high mobility group box 1 Homo sapiens 225-250 34638242-4 2021 Anthracycline and taxane treatments cause ICD and immunogenic modulations, resulting in the activation of antitumor immunity through damage-associated molecular patterns (DAMPs), such as adenosine triphosphate, calreticulin, high mobility group box 1, heat shock proteins 70/90, and annexin A1. taxane 18-24 high mobility group box 1 Homo sapiens 225-250 34624942-0 2021 (Effect of PPAR-gamma agonist pioglitazone on the prolifeiration of malignant nesothelionma cells induced by HMGB1). Pioglitazone 30-42 high mobility group box 1 Homo sapiens 109-114 34624942-8 2021 There was a significant decrease in the mRNA expression level of HMGB1 in the PGZ-treated group (100 mumol/L) as compared to the control group in MSTO-211H (P<0.05) ; however, the expression level of HMGB1 in NCI-H2452 was an increase or no significant differences (P>0.05) . Pioglitazone 78-81 high mobility group box 1 Homo sapiens 65-70 34624942-9 2021 Western blotting and immunofluorescence results showed that the protein expression of HMGB1 was reduced in the PGZ-treated group compared with the control group in MSTO-211H (P<0.05) , but the protein expression of that in NCI-H2452 was no significant differences (P>0.05) . Pioglitazone 111-114 high mobility group box 1 Homo sapiens 86-91 34624942-11 2021 Conclusion: Pioglitazone suppresses the proliferation of MM cells through inhibition of HMGB1 by the activation of PPAR-gamma. Pioglitazone 12-24 high mobility group box 1 Homo sapiens 88-93 34538217-5 2022 In 2.5D culture, treatment with HMGB1 induced permeability of FITC-dextran into the lumen formed by human lung, nasal and intestinal epithelial cells. fluorescein isothiocyanate dextran 62-74 high mobility group box 1 Homo sapiens 32-37 34684184-6 2021 Success has also been obtained after other means to inhinit HMGB1 and include: use of HMGB1 Box A (one of three HMGB1 domains), anti-HMGB1 antibody blockage of HMGB1 and/or its receptors, Toll like receptor (TLR) 4, treatment with thrombomodulin (TM) or vasoactive intestinal peptide (VIP) and glycyrrhizin (GLY, a triterpenoid saponin) that directly binds to HMGB1. Glycyrrhizic Acid 294-306 high mobility group box 1 Homo sapiens 360-365 34684184-6 2021 Success has also been obtained after other means to inhinit HMGB1 and include: use of HMGB1 Box A (one of three HMGB1 domains), anti-HMGB1 antibody blockage of HMGB1 and/or its receptors, Toll like receptor (TLR) 4, treatment with thrombomodulin (TM) or vasoactive intestinal peptide (VIP) and glycyrrhizin (GLY, a triterpenoid saponin) that directly binds to HMGB1. Glycyrrhizic Acid 308-311 high mobility group box 1 Homo sapiens 360-365 34684184-6 2021 Success has also been obtained after other means to inhinit HMGB1 and include: use of HMGB1 Box A (one of three HMGB1 domains), anti-HMGB1 antibody blockage of HMGB1 and/or its receptors, Toll like receptor (TLR) 4, treatment with thrombomodulin (TM) or vasoactive intestinal peptide (VIP) and glycyrrhizin (GLY, a triterpenoid saponin) that directly binds to HMGB1. triterpenoid saponin 315-335 high mobility group box 1 Homo sapiens 360-365 34621188-7 2021 Immunohistochemical analysis of vagal sensory neurons collected from mice after intranasal infection with murine pneumovirus or influenza A virus (IAV), or after intratracheal administration with the viral mimetic PolyI:C, revealed a significant increase in nuclear-to-cytoplasm translocation of HMGB1 compared to mock-inoculated mice. Poly I-C 214-221 high mobility group box 1 Homo sapiens 296-301 34573077-7 2021 Likewise, we showed the inhibitory effect of verbascoside on oxidative stress was via repression of PKC/HMGB1/RAGE/NFkappaB activation. acteoside 45-57 high mobility group box 1 Homo sapiens 104-109 34573077-6 2021 Moreover, verbascoside upregulated the PGC1-alpha and NRF1 expression and promoted mitochondrial biogenesis, which was mediated by suppression of PKC/HMGB1/RAGE/NFkappaB signaling. acteoside 10-22 high mobility group box 1 Homo sapiens 150-155 34568076-0 2021 CD68+ Macrophage Infiltration Associates With Poor Outcome of HPV Negative Oral Squamous Carcinoma Patients Receiving Radiation: Poly(I:C) Enhances Radiosensitivity of CAL-27 Cells but Promotes Macrophage Recruitment Through HMGB1. Poly I-C 129-138 high mobility group box 1 Homo sapiens 225-230 34566641-6 2021 Acute liver injury triggered by carbon tetrachloride (CCl4) injection resulted in significant upregulation of the plasma kallikrein gene (Klkb1) and was highly associated with the high mobility group box 1 gene, the marker of cell death (r = 0.75, p < 0.0005, n = 7). Carbon Tetrachloride 32-52 high mobility group box 1 Homo sapiens 180-205 34375967-5 2021 In the presented study, model drug OP loaded poly(lactic-co-glycolic acid) (PLGA) nanoparticles (NPs) targeted with spike-binding peptide 1 (SBP1) of SARS-CoV-2 were designed to be used as an efficient and prolonged released antiviral drug delivery systems. Polylactic Acid-Polyglycolic Acid Copolymer 45-74 high mobility group box 1 Homo sapiens 141-145 34566641-6 2021 Acute liver injury triggered by carbon tetrachloride (CCl4) injection resulted in significant upregulation of the plasma kallikrein gene (Klkb1) and was highly associated with the high mobility group box 1 gene, the marker of cell death (r = 0.75, p < 0.0005, n = 7). Carbon Tetrachloride 54-58 high mobility group box 1 Homo sapiens 180-205 34287039-7 2021 These findings indicate that PCV2-induced activation of the PERK-ERO1alpha axis would lead to enhanced generation of ROS sufficient to decrease HMGB1 retention in the nuclei, thus derepressing viral DNA from HMGB1 sequestration. ros 117-120 high mobility group box 1 Homo sapiens 144-149 34287039-7 2021 These findings indicate that PCV2-induced activation of the PERK-ERO1alpha axis would lead to enhanced generation of ROS sufficient to decrease HMGB1 retention in the nuclei, thus derepressing viral DNA from HMGB1 sequestration. ros 117-120 high mobility group box 1 Homo sapiens 208-213 34287039-9 2021 Cysteine residues 107 and 305 of Rep or 108 of Cap played important roles in PCV2-induced PERK activation and distribution of HMGB1. Cysteine 0-8 high mobility group box 1 Homo sapiens 126-131 34287039-11 2021 Collectively, this study offers novel insight into the mechanism of enhanced viral replication in which PCV2 manipulates ER to perturb its redox homeostasis via the PERK-ERO1alpha axis and the ER-sourced ROS from oxidative folding is sufficient to reduce HMGB1 retention in the nuclei, hence the release of HMGB1-bound viral DNA for replication. ros 204-207 high mobility group box 1 Homo sapiens 255-260 34287039-11 2021 Collectively, this study offers novel insight into the mechanism of enhanced viral replication in which PCV2 manipulates ER to perturb its redox homeostasis via the PERK-ERO1alpha axis and the ER-sourced ROS from oxidative folding is sufficient to reduce HMGB1 retention in the nuclei, hence the release of HMGB1-bound viral DNA for replication. ros 204-207 high mobility group box 1 Homo sapiens 307-312 34287039-12 2021 IMPORTANCE Considering the fact that clinical PCVAD mostly results from activation of latent PCV2 infection by confounding factors such as co-infection or environmental stresses, we propose that such confounding factors might impose oxidative stress to the animals where PCV2 in infected cells might utilize the elevated ROS to promote HMGB1 migration out of nuclei in favor of its replication. pcvad 46-51 high mobility group box 1 Homo sapiens 336-341 34287039-12 2021 IMPORTANCE Considering the fact that clinical PCVAD mostly results from activation of latent PCV2 infection by confounding factors such as co-infection or environmental stresses, we propose that such confounding factors might impose oxidative stress to the animals where PCV2 in infected cells might utilize the elevated ROS to promote HMGB1 migration out of nuclei in favor of its replication. ros 321-324 high mobility group box 1 Homo sapiens 336-341 34479472-0 2021 Correction to: 2-O, 3-O desulfated heparin (ODSH) increases bacterial clearance and attenuates lung injury in cystic fibrosis by restoring HMGB1-compromised macrophage function. 2-o 15-18 high mobility group box 1 Homo sapiens 139-144 34488792-5 2021 RESULTS: Nano-DOX were first shown to stimulate the tumor cells and the TAMs to release the cytokine HMGB1 which, regardless of its source, acted through the RAGE/NF-kappaB pathway to induce PD-L1 in the tumor cells and PD-L1/PD-1 in the TAMs. Doxorubicin 14-17 high mobility group box 1 Homo sapiens 101-106 34488792-6 2021 Interestingly, Nano-DOX also induced NF-kappaB-dependent RAGE expression in the tumor cells and thus reinforced HMGB1"s action thereon. Doxorubicin 20-23 high mobility group box 1 Homo sapiens 112-117 34488792-11 2021 CONCLUSIONS: PD-L1/PD-1 upregulation mediated by autocrine and paracrine activation of the HMGB1/RAGE/NF-kappaB signaling is a key response of lung cancer cells and their TAMs to stress, which can be induced by Nano-DOX. Doxorubicin 216-219 high mobility group box 1 Homo sapiens 91-96 34181980-3 2021 Using nuclear magnetic resonance (NMR), fluorescence, and computational approaches, we investigated how the D/E repeats causes the autoinhibition of HMGB1 against its specific binding to cisplatin-modified DNA. Cisplatin 187-196 high mobility group box 1 Homo sapiens 149-154 34479472-0 2021 Correction to: 2-O, 3-O desulfated heparin (ODSH) increases bacterial clearance and attenuates lung injury in cystic fibrosis by restoring HMGB1-compromised macrophage function. 3-o 20-23 high mobility group box 1 Homo sapiens 139-144 34479472-0 2021 Correction to: 2-O, 3-O desulfated heparin (ODSH) increases bacterial clearance and attenuates lung injury in cystic fibrosis by restoring HMGB1-compromised macrophage function. Heparin 35-42 high mobility group box 1 Homo sapiens 139-144 34415075-11 2021 The induction of HMGB1-TLR-MyD88-NFkappaB proinflammatory signaling pathways correlates with neurodegeneration (i.e., Fluoro-Jade B), lifetime alcohol consumption, and age of drinking onset. Alcohols 143-150 high mobility group box 1 Homo sapiens 17-22 34118353-0 2021 Serum HMGB1 Associates with Liver Disease and Predicts Readmission and Mortality in Patients with Alcohol Use Disorder. Alcohols 98-105 high mobility group box 1 Homo sapiens 6-11 34237593-5 2021 Similarly, compound 48/80 increased levels of cardiac troponin I (cTnI) and tryptase, cardiomyocytes apoptosis, and expression of high-mobility group box 1 (HMGB1), toll-like receptor 4 (TLR4), and nuclear factor-kappa B p65 (NF-kappaB p65) in cardiac tissues induced by I/R injury, but it can be partially decreased by dexmedetomidine pretreatment. Dexmedetomidine 320-335 high mobility group box 1 Homo sapiens 130-155 34237593-5 2021 Similarly, compound 48/80 increased levels of cardiac troponin I (cTnI) and tryptase, cardiomyocytes apoptosis, and expression of high-mobility group box 1 (HMGB1), toll-like receptor 4 (TLR4), and nuclear factor-kappa B p65 (NF-kappaB p65) in cardiac tissues induced by I/R injury, but it can be partially decreased by dexmedetomidine pretreatment. Dexmedetomidine 320-335 high mobility group box 1 Homo sapiens 157-162 34237593-7 2021 Our data suggest that dexmedetomidine preconditioning alleviates the degranulation of mast cells and the apoptosis of cardiomyocytes caused by I/R injury, and inhibits the activation of inflammatory related factors HMGB1, TLR4, and NF-kappaB p65. Dexmedetomidine 22-37 high mobility group box 1 Homo sapiens 215-220 34384355-10 2021 Serum NLRP3 levels were correlated with headache duration and hospital stay, while headache response to paracetamol was negatively correlated with HMGB1 and positively associated with IL-10 levels. Acetaminophen 104-115 high mobility group box 1 Homo sapiens 147-152 34816916-8 2021 The correlation analysis showed that, in acute aortic dissection patients, the high-mobility group box 1 and receptor for advanced glycation end products levels were negatively correlated with partial pressure of oxygen/fraction of inspired oxygen, respectively (p<0.05). Oxygen 213-219 high mobility group box 1 Homo sapiens 79-117 34816916-8 2021 The correlation analysis showed that, in acute aortic dissection patients, the high-mobility group box 1 and receptor for advanced glycation end products levels were negatively correlated with partial pressure of oxygen/fraction of inspired oxygen, respectively (p<0.05). Oxygen 241-247 high mobility group box 1 Homo sapiens 79-117 34370950-0 2021 Molecular-Level Understanding of the Influence of Ions and Water on HMGB1 Adsorption Induced by Surface Hydroxylation of Titanium Implants. Water 59-64 high mobility group box 1 Homo sapiens 68-73 34370950-0 2021 Molecular-Level Understanding of the Influence of Ions and Water on HMGB1 Adsorption Induced by Surface Hydroxylation of Titanium Implants. Titanium 121-129 high mobility group box 1 Homo sapiens 68-73 34370950-4 2021 To develop different surface strategies that improve the clinical outcome of titanium-based implants by controlling their biological activity, a molecular-scale understanding of HMGB1-surface interactions is desired. Titanium 77-85 high mobility group box 1 Homo sapiens 178-183 34370950-5 2021 Here, we use molecular dynamics (MD) computer simulations to provide direct insight into the HMGB1 interactions and the possible molecular arrangements of HMGB1 on fully hydroxylated and nonhydroxylated rutile (110) TiO2 surfaces. titanium dioxide 216-220 high mobility group box 1 Homo sapiens 155-160 34370950-7 2021 The hydroxylated TiO2 surface shows a greater affinity for HMGB1 than the nonhydroxylated surface. titanium dioxide 17-21 high mobility group box 1 Homo sapiens 59-64 34457261-4 2021 Here we review the pathophysiological underpinnings of lidocaine"s role as an anti-nociceptive, anti-inflammatory mediated by toll-like receptor (TLR) and nuclear factor kappa-beta (NF-kbeta) signalling pathways and downstream cytokine effectors high mobility group box 1 (HMGB1) and tumour necrosis factor-alpha (TNF-alpha). Lidocaine 55-64 high mobility group box 1 Homo sapiens 246-271 34457261-4 2021 Here we review the pathophysiological underpinnings of lidocaine"s role as an anti-nociceptive, anti-inflammatory mediated by toll-like receptor (TLR) and nuclear factor kappa-beta (NF-kbeta) signalling pathways and downstream cytokine effectors high mobility group box 1 (HMGB1) and tumour necrosis factor-alpha (TNF-alpha). Lidocaine 55-64 high mobility group box 1 Homo sapiens 273-278 34153664-7 2021 Inhibition of HMGB1 decreased Nrf2 expression and ROS release, improved MMP level and reduced NLRP3 inflammasome activation. ros 50-53 high mobility group box 1 Homo sapiens 14-19 34294368-0 2021 Calunduloside E inhibits HepG2 cell proliferation and migration via p38/JNK-HMGB1 signalling axis. calunduloside e 0-15 high mobility group box 1 Homo sapiens 76-81 34374662-0 2021 lncRNA ANRIL aggravates the chemoresistance of pancreatic cancer cells to gemcitabine by targeting inhibition of miR-181a and targeting HMGB1-induced autophagy. gemcitabine 74-85 high mobility group box 1 Homo sapiens 136-141 34445093-9 2021 In 2.5D Matrigel culture, treatment with HMGB1 induced permeability of FITC-dextran (FD-4) into the lumen. fluorescein isothiocyanate dextran 71-83 high mobility group box 1 Homo sapiens 41-46 34445093-10 2021 Pretreatment with EW-7197 prevented the effects of HMGB1. vactosertib 18-25 high mobility group box 1 Homo sapiens 51-56 34128295-10 2021 The target gene of miR-141-5p was predicted with the TargetScan database, and the interaction between miR-141-5p and HMGB1/nuclear factor-kappaB (NF-kappaB) was further validated by dual-luciferase reporter assay, qRT-PCR, and Western blot analysis. mir-141-5p 19-29 high mobility group box 1 Homo sapiens 117-122 34128295-10 2021 The target gene of miR-141-5p was predicted with the TargetScan database, and the interaction between miR-141-5p and HMGB1/nuclear factor-kappaB (NF-kappaB) was further validated by dual-luciferase reporter assay, qRT-PCR, and Western blot analysis. mir-141-5p 102-112 high mobility group box 1 Homo sapiens 117-122 34366853-9 2021 As a result, LPS evidently promoted the expression of pro-inflammatory cytokines and HMGB1, the MyD88-NF-kappaB activation, and the NLRP3 inflammasome profile in THP-1 cells, while MAG obviously inhibited the "M1"" polarization of THP-1 cells. magnoflorine 181-184 high mobility group box 1 Homo sapiens 85-90 34366853-0 2021 Magnoflorine Alleviates "M1" Polarized Macrophage-Induced Intervertebral Disc Degeneration Through Repressing the HMGB1/Myd88/NF-kappaB Pathway and NLRP3 Inflammasome. magnoflorine 0-12 high mobility group box 1 Homo sapiens 114-119 34366853-12 2021 Furthermore, MAG dampened the HMGB1 expression and inactivated the MyD88/NF-kappaB pathway and NLRP3 inflammasome in NP cells. magnoflorine 13-16 high mobility group box 1 Homo sapiens 30-35 34366853-13 2021 In conclusion, this study confirmed that MAG alleviates "M1" polarized macrophage-mediated NP cell damage by inactivating the HMGB1-MyD88-NF-kappaB pathway and NLRP3 inflammasome, which provides a new reference for IDD treatment. magnoflorine 41-44 high mobility group box 1 Homo sapiens 126-131 34214538-8 2021 We hypothesized that mutating the hydrophobic residues (F28, F32, and F40) of SBP1 which do not directly interact with the spike protein to alanine would reduce peptide oligomerization without affecting its spike binding affinity. Alanine 140-147 high mobility group box 1 Homo sapiens 78-82 34350063-4 2021 Alternol triggered ICD in prostate cancer cells, as evidenced by the release of damage-associated molecular patterns (DAMPs) (i.e., calreticulin, CALR; high mobility group protein B1, HMGB1; and adenosine triphosphate, ATP) and pro-inflammatory cytokine (i.e., interleukin (IL)-1alpha, IL-1beta, IL-6, and IL-8) expression. Alternol 0-8 high mobility group box 1 Homo sapiens 152-182 34350063-4 2021 Alternol triggered ICD in prostate cancer cells, as evidenced by the release of damage-associated molecular patterns (DAMPs) (i.e., calreticulin, CALR; high mobility group protein B1, HMGB1; and adenosine triphosphate, ATP) and pro-inflammatory cytokine (i.e., interleukin (IL)-1alpha, IL-1beta, IL-6, and IL-8) expression. Alternol 0-8 high mobility group box 1 Homo sapiens 184-189 34289368-4 2021 TauO exposure triggers astrocyte senescence through high mobility group box 1 (HMGB1) release and inflammatory senescence-associated secretory phenotype (SASP), which mediates paracrine senescence in adjacent cells. tauo 0-4 high mobility group box 1 Homo sapiens 52-77 34289368-4 2021 TauO exposure triggers astrocyte senescence through high mobility group box 1 (HMGB1) release and inflammatory senescence-associated secretory phenotype (SASP), which mediates paracrine senescence in adjacent cells. tauo 0-4 high mobility group box 1 Homo sapiens 79-84 34289368-5 2021 HMGB1 release inhibition using ethyl pyruvate (EP) and glycyrrhizic acid (GA) prevents TauO-induced senescence through inhibition of p38-mitogen-activated protein kinase (MAPK) and nuclear factor kappaB (NF-kappaB)-the essential signaling pathways for SASP development. ethyl pyruvate 31-45 high mobility group box 1 Homo sapiens 0-5 34289368-5 2021 HMGB1 release inhibition using ethyl pyruvate (EP) and glycyrrhizic acid (GA) prevents TauO-induced senescence through inhibition of p38-mitogen-activated protein kinase (MAPK) and nuclear factor kappaB (NF-kappaB)-the essential signaling pathways for SASP development. ethyl pyruvate 47-49 high mobility group box 1 Homo sapiens 0-5 34289368-5 2021 HMGB1 release inhibition using ethyl pyruvate (EP) and glycyrrhizic acid (GA) prevents TauO-induced senescence through inhibition of p38-mitogen-activated protein kinase (MAPK) and nuclear factor kappaB (NF-kappaB)-the essential signaling pathways for SASP development. Glycyrrhizic Acid 55-72 high mobility group box 1 Homo sapiens 0-5 34289368-5 2021 HMGB1 release inhibition using ethyl pyruvate (EP) and glycyrrhizic acid (GA) prevents TauO-induced senescence through inhibition of p38-mitogen-activated protein kinase (MAPK) and nuclear factor kappaB (NF-kappaB)-the essential signaling pathways for SASP development. Glycyrrhizic Acid 74-76 high mobility group box 1 Homo sapiens 0-5 34289368-5 2021 HMGB1 release inhibition using ethyl pyruvate (EP) and glycyrrhizic acid (GA) prevents TauO-induced senescence through inhibition of p38-mitogen-activated protein kinase (MAPK) and nuclear factor kappaB (NF-kappaB)-the essential signaling pathways for SASP development. tauo 87-91 high mobility group box 1 Homo sapiens 0-5 34289368-7 2021 Collectively, TauO-induced HMGB1 release promotes cellular senescence and neuropathology, which could represent an important common pathomechanism in tauopathies including AD and FTD. tauo 14-18 high mobility group box 1 Homo sapiens 27-32 34412765-5 2021 This study focuses on the HMGB1 release progress, which connected with Janus kinases/signal transducer and activator of transcription (JAK/STAT), nuclear factor-kappaB (NF-kappaB), Notch, inflammasome, tumor necrosis factor (TNF), mitogen-activated protein kinase (MAPK), reactive oxygen species (ROS), peroxisome proliferator-activated receptor (PPAR) and other signaling or dependent pathways in ARDS. Reactive Oxygen Species 272-295 high mobility group box 1 Homo sapiens 26-31 33318439-0 2021 Role of N4-acetylcytidine for continuously activating NLRP3 inflammosome by HMGB1 pathway in microglia. N-acetylcytidine 8-25 high mobility group box 1 Homo sapiens 76-81 34412765-5 2021 This study focuses on the HMGB1 release progress, which connected with Janus kinases/signal transducer and activator of transcription (JAK/STAT), nuclear factor-kappaB (NF-kappaB), Notch, inflammasome, tumor necrosis factor (TNF), mitogen-activated protein kinase (MAPK), reactive oxygen species (ROS), peroxisome proliferator-activated receptor (PPAR) and other signaling or dependent pathways in ARDS. Reactive Oxygen Species 297-300 high mobility group box 1 Homo sapiens 26-31 34187428-11 2021 Moreover, the regression of tumors in mice that received PV-10 in combination with gemcitabine was associated with the depletion of splenic CD11b+Gr-1+ cells and increases in damage associated molecular patterns HMGB1, S100A8, and IL-1alpha. pv-10 57-62 high mobility group box 1 Homo sapiens 212-217 34187428-11 2021 Moreover, the regression of tumors in mice that received PV-10 in combination with gemcitabine was associated with the depletion of splenic CD11b+Gr-1+ cells and increases in damage associated molecular patterns HMGB1, S100A8, and IL-1alpha. gemcitabine 83-94 high mobility group box 1 Homo sapiens 212-217 34182538-0 2021 GPR30-mediated HMGB1 upregulation in CAFs induces autophagy and tamoxifen resistance in ERalpha-positive breast cancer cells. Tamoxifen 64-73 high mobility group box 1 Homo sapiens 15-20 34182538-3 2021 Herein, considering that GPR30 mediates transcriptional regulation in different cell backgrounds, a microarray strategy was applied in immortalized CAFs derived from primary breast cancer samples, resulting in the identification of 165 GPR30 target genes, among which HMGB1 was confirmed to be upregulated by 17-beta estradiol(E2)- and TAM-triggered GPR30 activation in CAFs. Estradiol 309-326 high mobility group box 1 Homo sapiens 268-273 34182538-5 2021 GPR30-induced HMGB1 upregulation triggered MEK/ERK signaling, leading to increased autophagic behavior to protect cancer cells from TAM-induced apoptosis, mimicking the recombinant HMGB1-mediated increase in cancer cell resistance potential to TAM. Tamoxifen 244-247 high mobility group box 1 Homo sapiens 181-186 34182538-7 2021 CAF-expressed GPR30 induced TAM resistance via HMGB1 in vivo. Tamoxifen 28-31 high mobility group box 1 Homo sapiens 47-52 34182538-3 2021 Herein, considering that GPR30 mediates transcriptional regulation in different cell backgrounds, a microarray strategy was applied in immortalized CAFs derived from primary breast cancer samples, resulting in the identification of 165 GPR30 target genes, among which HMGB1 was confirmed to be upregulated by 17-beta estradiol(E2)- and TAM-triggered GPR30 activation in CAFs. Estradiol 327-329 high mobility group box 1 Homo sapiens 268-273 34182538-8 2021 Overall, TAM upregulated HMGB1 expression and secretion in CAFs via GPR30/PI3K/AKT signaling, and the secreted HMGB1 induced autophagy to enhance TAM resistance in MCF-7 cells in an ERK-dependent manner. Tamoxifen 9-12 high mobility group box 1 Homo sapiens 25-30 34182538-8 2021 Overall, TAM upregulated HMGB1 expression and secretion in CAFs via GPR30/PI3K/AKT signaling, and the secreted HMGB1 induced autophagy to enhance TAM resistance in MCF-7 cells in an ERK-dependent manner. Tamoxifen 9-12 high mobility group box 1 Homo sapiens 111-116 34182538-8 2021 Overall, TAM upregulated HMGB1 expression and secretion in CAFs via GPR30/PI3K/AKT signaling, and the secreted HMGB1 induced autophagy to enhance TAM resistance in MCF-7 cells in an ERK-dependent manner. Tamoxifen 146-149 high mobility group box 1 Homo sapiens 111-116 34182538-3 2021 Herein, considering that GPR30 mediates transcriptional regulation in different cell backgrounds, a microarray strategy was applied in immortalized CAFs derived from primary breast cancer samples, resulting in the identification of 165 GPR30 target genes, among which HMGB1 was confirmed to be upregulated by 17-beta estradiol(E2)- and TAM-triggered GPR30 activation in CAFs. Tamoxifen 336-339 high mobility group box 1 Homo sapiens 268-273 34182538-4 2021 Activated GPR30 increased extracellular HMGB1 secretion by CAFs, which was reduced by blocking PI3K/AKT signaling using G15 or LY294002. 2-(4-morpholinyl)-8-phenyl-4H-1-benzopyran-4-one 127-135 high mobility group box 1 Homo sapiens 40-45 34182538-5 2021 GPR30-induced HMGB1 upregulation triggered MEK/ERK signaling, leading to increased autophagic behavior to protect cancer cells from TAM-induced apoptosis, mimicking the recombinant HMGB1-mediated increase in cancer cell resistance potential to TAM. Tamoxifen 132-135 high mobility group box 1 Homo sapiens 14-19 34163481-3 2021 The mechanisms by which metformin regulates the function of macrophages include AMPK, AMPK independent targets, NF-kappaB, ABCG5/8, Sirt1, FOXO1/FABP4 and HMGB1. Metformin 24-33 high mobility group box 1 Homo sapiens 155-160 34164408-6 2021 In contrast, the HMGB1-mediated expression of HLA-DR, CD40, and CD86 on dendritic cells and production of IL-1beta, IL-6, and TNF-alpha were reduced by rapamycin. Sirolimus 152-161 high mobility group box 1 Homo sapiens 17-22 34164408-7 2021 Rapamycin can inhibit HMGB1-induced activation of mDCs and secretion of pro-inflammatory cytokines. Sirolimus 0-9 high mobility group box 1 Homo sapiens 22-27 34214372-0 2021 Analysis of HMGB-1 level before and after providing atorvastatin standard therapy in coronary artery disease patients with type-2 diabetes mellitus compared to without type-2 diabetes mellitus. Atorvastatin 52-64 high mobility group box 1 Homo sapiens 12-18 34214372-2 2021 Atorvastatin has a pleiotropic effect as anti-inflammatory through one of the target levels of High Mobility Group Box-1 (HMGB-1). Atorvastatin 0-12 high mobility group box 1 Homo sapiens 95-120 34214372-2 2021 Atorvastatin has a pleiotropic effect as anti-inflammatory through one of the target levels of High Mobility Group Box-1 (HMGB-1). Atorvastatin 0-12 high mobility group box 1 Homo sapiens 122-128 34214372-3 2021 This prospective observational study aimed to analyze the effect of atorvastatin on serum HMGB-1 levels in CAD. Atorvastatin 68-80 high mobility group box 1 Homo sapiens 90-96 34214372-5 2021 Serum HMGB-1 levels were measured in patients with CAD who were given atorvastatin for CAD with type-2 diabetes mellitus compared without type-2 diabetes mellitus in a patient ward. Atorvastatin 70-82 high mobility group box 1 Homo sapiens 6-12 34214372-13 2021 CONCLUSIONS: HMGB-1 levels after providing atorvastatin in CAD with type-2 diabetes mellitus increased significantly, meanwhile, in CAD without type-2 diabetes mellitus did not decrease significantly. Atorvastatin 43-55 high mobility group box 1 Homo sapiens 13-19 34295655-7 2021 Among the various treatment modalities administered, platinum-based combination or single-agent chemotherapy tended to elicit robust increases in the concentrations of HMGB1 and CRT. Platinum 53-61 high mobility group box 1 Homo sapiens 168-173 34198762-7 2021 The Spearman"s rho revealed that HMGB1 SCs were positively correlated with the AQ attention to detail subscale (rs = 0.46, p = 0.045) and with the SQ total score (rs = 0.42, p = 0.04) in the ASD group. aq 79-81 high mobility group box 1 Homo sapiens 33-38 34127858-6 2021 Mechanistically, exercise-induced HMGB1 release enhances itaconate metabolism in the tricarboxylic acid cycle that impacts Kupffer cells in an NRF2-dependent manner. itaconic acid 57-66 high mobility group box 1 Homo sapiens 34-39 34127858-6 2021 Mechanistically, exercise-induced HMGB1 release enhances itaconate metabolism in the tricarboxylic acid cycle that impacts Kupffer cells in an NRF2-dependent manner. Tricarboxylic Acids 85-103 high mobility group box 1 Homo sapiens 34-39 34079539-0 2021 Prednisolone Suppresses the Extracellular Release of HMGB-1 and Associated Inflammatory Pathways in Kawasaki Disease. Prednisolone 0-12 high mobility group box 1 Homo sapiens 53-59 34094941-0 2021 lncRNA MIAT/HMGB1 Axis Is Involved in Cisplatin Resistance via Regulating IL6-Mediated Activation of the JAK2/STAT3 Pathway in Nasopharyngeal Carcinoma. Cisplatin 38-47 high mobility group box 1 Homo sapiens 12-17 34094941-7 2021 Furthermore, we determine that elevated lncRNA MIAT level upregulates HMGB1 expression, contributing to cisplatin resistance in NPC cells. Cisplatin 104-113 high mobility group box 1 Homo sapiens 70-75 34094941-8 2021 We find that the deficiency of the lncRNA MIAT/HMGB1 axis, inhibition of JAK2/STAT3, or neutralization of IL6 by antibodies significantly re-sensitizes resistant NPC cells to cisplatin in resistant NPC cells. Cisplatin 175-184 high mobility group box 1 Homo sapiens 47-52 34094941-9 2021 Moreover, we provide the in vivo evidence that the deficiency of HMGB1 reduces cisplatin-resistant tumor growth. Cisplatin 79-88 high mobility group box 1 Homo sapiens 65-70 34079539-10 2021 Our findings suggest that extracellular HMGB-1 plays an important role in mediating KD pathogenesis and that PSL treatment during the acute phase of KD may ameliorate HMGB-1-mediated inflammatory responses in KD vasculitis. Prednisolone 109-112 high mobility group box 1 Homo sapiens 40-46 34079539-10 2021 Our findings suggest that extracellular HMGB-1 plays an important role in mediating KD pathogenesis and that PSL treatment during the acute phase of KD may ameliorate HMGB-1-mediated inflammatory responses in KD vasculitis. Prednisolone 109-112 high mobility group box 1 Homo sapiens 167-173 34567165-0 2021 Inhibition of HMGB1 Might Enhance the Protective Effect of Taxifolin in Cardiomyocytes via PI3K/AKT Signaling Pathway. taxifolin 59-68 high mobility group box 1 Homo sapiens 14-19 34068442-8 2021 Treatment of gastric cancer cells with CML-HMGB1 enhanced cell proliferation and invasion, sphere formation, and protection from thapsigargin-induced apoptosis, and decreased 5-FU sensitivity in comparison to HMGB1. Thapsigargin 129-141 high mobility group box 1 Homo sapiens 43-48 34068442-8 2021 Treatment of gastric cancer cells with CML-HMGB1 enhanced cell proliferation and invasion, sphere formation, and protection from thapsigargin-induced apoptosis, and decreased 5-FU sensitivity in comparison to HMGB1. Fluorouracil 175-179 high mobility group box 1 Homo sapiens 43-48 34066647-0 2021 Ethyl Pyruvate Attenuates Microglial NLRP3 Inflammasome Activation via Inhibition of HMGB1/NF-kappaB/miR-223 Signaling. ethyl pyruvate 0-14 high mobility group box 1 Homo sapiens 85-90 34066647-6 2021 Furthermore, ethyl pyruvate reduced the formation of total and mitochondrial ROS and suppressed inflammasome-induced HMGB1 upregulation and nuclear NF-kappaB translocation and reversed the inflammasome activation-induced miRNA expression profile for miR-223 in N9 cells. ethyl pyruvate 13-27 high mobility group box 1 Homo sapiens 117-122 34066647-7 2021 Our study suggests that ethyl pyruvate effectively suppresses the NLRP3 inflammasome activation in microglial cells regulation by miR-223 and NF-kappaB/HMGB1 axis. ethyl pyruvate 24-38 high mobility group box 1 Homo sapiens 152-157 35471628-9 2022 Therefore, targeting of the HMGB1 pathway by anti-HMGB1 agents, such as heparin, resveratrol and metformin, may decrease COVID-19 severity. Heparin 72-79 high mobility group box 1 Homo sapiens 28-33 34163767-3 2021 Herein, we developed a dual-modal microscopy imaging strategy to investigate, in situ, the formation of ternary binding complexes of the transcription cofactor HMGB1 and transcription factor Smad3 with cisplatin crosslinked DNA in single cells. Cisplatin 202-211 high mobility group box 1 Homo sapiens 160-165 35617997-0 2022 Decreased HMGB1 expression contributed to cutaneous toxicity caused by lapatinib. Lapatinib 71-80 high mobility group box 1 Homo sapiens 10-15 35617997-7 2022 Therefore, restoring HMGB1 expression might be an effective remedy against lapatinib-induced cutaneous toxicity. Lapatinib 75-84 high mobility group box 1 Homo sapiens 21-26 35471628-9 2022 Therefore, targeting of the HMGB1 pathway by anti-HMGB1 agents, such as heparin, resveratrol and metformin, may decrease COVID-19 severity. Heparin 72-79 high mobility group box 1 Homo sapiens 50-55 35471628-9 2022 Therefore, targeting of the HMGB1 pathway by anti-HMGB1 agents, such as heparin, resveratrol and metformin, may decrease COVID-19 severity. Resveratrol 81-92 high mobility group box 1 Homo sapiens 28-33 35471628-9 2022 Therefore, targeting of the HMGB1 pathway by anti-HMGB1 agents, such as heparin, resveratrol and metformin, may decrease COVID-19 severity. Resveratrol 81-92 high mobility group box 1 Homo sapiens 50-55 35471628-9 2022 Therefore, targeting of the HMGB1 pathway by anti-HMGB1 agents, such as heparin, resveratrol and metformin, may decrease COVID-19 severity. Metformin 97-106 high mobility group box 1 Homo sapiens 28-33 35471628-9 2022 Therefore, targeting of the HMGB1 pathway by anti-HMGB1 agents, such as heparin, resveratrol and metformin, may decrease COVID-19 severity. Metformin 97-106 high mobility group box 1 Homo sapiens 50-55 35596723-0 2022 The HN1/HMGB1 axis promotes the proliferation and metastasis of hepatocellular carcinoma and attenuates the chemosensitivity to oxaliplatin. Oxaliplatin 128-139 high mobility group box 1 Homo sapiens 8-13 35605331-9 2022 Therefore, we further studied how hippocampal neurons affected microglia under PM2.5 exposure, Further investigation indicated that silencing HMGB1 could affect the activation of P2X7R and reduce the release of ATP from hippocampal neurons, thus protecting the interaction between microglia and hippocampal neurons. Adenosine Triphosphate 211-214 high mobility group box 1 Homo sapiens 142-147 35596723-8 2022 In the nucleus, downregulation of HMGB1 followed by HN1 knockdown resulted in increased DNA damage and cell death in the oxaliplatin-treated HCC cells. Oxaliplatin 121-132 high mobility group box 1 Homo sapiens 34-39 35596723-10 2022 Furthermore, HN1 knockdown in combination with HMGB1 overexpression restored the aggressive phenotypes of HCC cells and sensitivity of these cells to oxaliplatin. Oxaliplatin 150-161 high mobility group box 1 Homo sapiens 47-52 35543500-5 2022 Total internal reflection microscopy (TIRFM) and molecular dynamic (MD) simulation show that although HMGB1 is adsorbed more on plates with lower SAA, the exposure ratio of cysteine (CYS) residue in HMGB1 is significantly decreased in lower SAA group. saa 146-149 high mobility group box 1 Homo sapiens 102-107 35582883-11 2022 These results suggest that ketamine may reduce HMGB1 and RAGE accumulation in patients with depression, thereby providing a new therapeutic target for preventing and treating this disease. Ketamine 27-35 high mobility group box 1 Homo sapiens 47-52 35577872-0 2022 Doxorubicin impacts chromatin binding of HMGB1, Histone H1 and retinoic acid receptor. Doxorubicin 0-11 high mobility group box 1 Homo sapiens 41-46 35577872-2 2022 Here, we studied the effects of Dox on the chromatin binding of the architectural proteins high mobility group B1 (HMGB1) and the linker histone H1, and the transcription factor retinoic acid receptor (RARalpha) by fluorescence recovery after photobleaching (FRAP) and fluorescence correlation spectroscopy (FCS) in live cells. Doxorubicin 32-35 high mobility group box 1 Homo sapiens 91-113 35577872-2 2022 Here, we studied the effects of Dox on the chromatin binding of the architectural proteins high mobility group B1 (HMGB1) and the linker histone H1, and the transcription factor retinoic acid receptor (RARalpha) by fluorescence recovery after photobleaching (FRAP) and fluorescence correlation spectroscopy (FCS) in live cells. Doxorubicin 32-35 high mobility group box 1 Homo sapiens 115-120 35577872-3 2022 At lower doses, Dox increased the binding of HMGB1 to DNA while decreasing the binding of the linker histone H1. Doxorubicin 16-19 high mobility group box 1 Homo sapiens 45-50 35577872-4 2022 At higher doses that correspond to the peak plasma concentrations achieved during chemotherapy, Dox reduced the binding of HMGB1 as well. Doxorubicin 96-99 high mobility group box 1 Homo sapiens 123-128 35551614-15 2022 The suppression of ezrin by siRNA or the blockade of ROCK activation with Y-27632 reduced the production of TNF-alpha, IL-1beta, and HMGB1 in response to LPS. Y 27632 74-81 high mobility group box 1 Homo sapiens 133-138 35543500-5 2022 Total internal reflection microscopy (TIRFM) and molecular dynamic (MD) simulation show that although HMGB1 is adsorbed more on plates with lower SAA, the exposure ratio of cysteine (CYS) residue in HMGB1 is significantly decreased in lower SAA group. Cysteine 173-181 high mobility group box 1 Homo sapiens 199-204 35543500-5 2022 Total internal reflection microscopy (TIRFM) and molecular dynamic (MD) simulation show that although HMGB1 is adsorbed more on plates with lower SAA, the exposure ratio of cysteine (CYS) residue in HMGB1 is significantly decreased in lower SAA group. Cysteine 183-186 high mobility group box 1 Homo sapiens 199-204 35632744-0 2022 PRRSV Induces HMGB1 Phosphorylation at Threonine-51 Residue to Enhance Its Secretion. Threonine 39-48 high mobility group box 1 Homo sapiens 14-19 35526247-4 2022 This study was conducted to determine whether HMGB1 polymorphisms (rs1360485, rs2249825 and rs1060348) are associated with the incidence of differentiation syndrome in acute promyelocytic leukemia patients treated with all-trans retinoic acid and arsenic trioxide. Tretinoin 219-242 high mobility group box 1 Homo sapiens 46-51 35625834-5 2022 Among these peptides, Nal-P-113 demonstrated the best anticancer activity and caused cancer cells to release potent danger-associated molecular patterns (DAMPs), such as reactive oxygen species (ROS), cytochrome c, ATP, and high-mobility group box 1 (HMGB1). nal-p-113 22-31 high mobility group box 1 Homo sapiens 224-249 35625834-5 2022 Among these peptides, Nal-P-113 demonstrated the best anticancer activity and caused cancer cells to release potent danger-associated molecular patterns (DAMPs), such as reactive oxygen species (ROS), cytochrome c, ATP, and high-mobility group box 1 (HMGB1). nal-p-113 22-31 high mobility group box 1 Homo sapiens 251-256 35526247-4 2022 This study was conducted to determine whether HMGB1 polymorphisms (rs1360485, rs2249825 and rs1060348) are associated with the incidence of differentiation syndrome in acute promyelocytic leukemia patients treated with all-trans retinoic acid and arsenic trioxide. Arsenic Trioxide 247-263 high mobility group box 1 Homo sapiens 46-51 35632744-4 2022 Here, we discovered that the phosphorylation level of HMGB1 in threonine residues increased in PRRSV-infected cells. Threonine 63-72 high mobility group box 1 Homo sapiens 54-59 35632744-5 2022 A site-directed mutagenesis study showed that HMGB1 phosphorylation at threonine-51 was associated with HMGB1 secretion induced by PRRSV infection. Threonine 71-80 high mobility group box 1 Homo sapiens 46-51 35632744-5 2022 A site-directed mutagenesis study showed that HMGB1 phosphorylation at threonine-51 was associated with HMGB1 secretion induced by PRRSV infection. Threonine 71-80 high mobility group box 1 Homo sapiens 104-109 35632744-8 2022 Moreover, the phosphorylation of HMGB1 at threonine-51 was correlated with the interaction between HMGB1 and RPS3. Threonine 42-51 high mobility group box 1 Homo sapiens 33-38 35632744-8 2022 Moreover, the phosphorylation of HMGB1 at threonine-51 was correlated with the interaction between HMGB1 and RPS3. Threonine 42-51 high mobility group box 1 Homo sapiens 99-104 35632744-10 2022 These results demonstrate that PRRSV may induce HMGB1 phosphorylation at threonine-51 and increase its interaction with RPS3 to enhance HMGB1 secretion. Threonine 73-82 high mobility group box 1 Homo sapiens 48-53 35632744-10 2022 These results demonstrate that PRRSV may induce HMGB1 phosphorylation at threonine-51 and increase its interaction with RPS3 to enhance HMGB1 secretion. Threonine 73-82 high mobility group box 1 Homo sapiens 136-141 35490244-0 2022 Correction to: miR-495-3p depresses cell proliferation and migration by downregulating HMGB1 in colorectal cancer. mir-495-3p 15-25 high mobility group box 1 Homo sapiens 87-92 35255342-6 2022 Additionally, 7 at 0.1 and 1.0 microM synergistically enhanced the cytotoxicity of etoposide against SBC-3 cells; compound 7 induced the release of DAMPs; the release of HMGB1, the secretion of ATP, and the exposure of CALR in the SBC-3 cells. Etoposide 83-92 high mobility group box 1 Homo sapiens 170-175 34990082-0 2022 Perfluorooctane sulfonate aggravates CCl4-induced hepatic fibrosis via HMGB1/TLR4/Smad signaling. perfluorooctane sulfonic acid 0-25 high mobility group box 1 Homo sapiens 71-76 34990082-5 2022 Furthermore, PFOS exposure may promote the activation of high-mobility group box 1 (HMGB1)/toll-like receptor 4 (TLR4) signaling pathway through inducing the secretion of HMGB1 from hepatocytes. perfluorooctane sulfonic acid 13-17 high mobility group box 1 Homo sapiens 57-82 34990082-5 2022 Furthermore, PFOS exposure may promote the activation of high-mobility group box 1 (HMGB1)/toll-like receptor 4 (TLR4) signaling pathway through inducing the secretion of HMGB1 from hepatocytes. perfluorooctane sulfonic acid 13-17 high mobility group box 1 Homo sapiens 84-89 34990082-5 2022 Furthermore, PFOS exposure may promote the activation of high-mobility group box 1 (HMGB1)/toll-like receptor 4 (TLR4) signaling pathway through inducing the secretion of HMGB1 from hepatocytes. perfluorooctane sulfonic acid 13-17 high mobility group box 1 Homo sapiens 171-176 35147911-0 2022 All-Trans Retinoic Acid-Preconditioned Mesenchymal Stem Cells Improve Motor Function and Alleviate Tissue Damage After Spinal Cord Injury by Inhibition of HMGB1/NF-kappaB/NLRP3 Pathway Through Autophagy Activation. Tretinoin 0-23 high mobility group box 1 Homo sapiens 155-160 35477503-4 2022 METHODS: In this study, we developed oncolytic herpes simplex virus type 1 expressing HMGB1 protein (HSV-HMGB1) and investigated the cytotoxic effect of HSV-HMGB1 and its parental virus (HSV-ble) on three colorectal cancer cells (HCT116, SW480, and HT29) under normoxic (20% oxygen) and hypoxic (1% oxygen) conditions. Oxygen 299-305 high mobility group box 1 Homo sapiens 86-91 35366433-0 2022 HMGB1 is increased in patients with immune thrombocytopenia and negatively associates with Tregs. tregs 91-96 high mobility group box 1 Homo sapiens 0-5 35477503-4 2022 METHODS: In this study, we developed oncolytic herpes simplex virus type 1 expressing HMGB1 protein (HSV-HMGB1) and investigated the cytotoxic effect of HSV-HMGB1 and its parental virus (HSV-ble) on three colorectal cancer cells (HCT116, SW480, and HT29) under normoxic (20% oxygen) and hypoxic (1% oxygen) conditions. Oxygen 275-281 high mobility group box 1 Homo sapiens 86-91 35624958-9 2022 The knockout of MMP-9 rescues cognitive impairment and cisplatin-induced upregulation of HMGB1 in SHSY5Y cells. Cisplatin 55-64 high mobility group box 1 Homo sapiens 89-94 35573828-5 2022 Herein, we report that administration of recombinant HMGB1 into the spinal cord at the time of lysolecithin administration resulted in arrest of OPC differentiation in vivo, and a profound impairment of remyelination. Lysophosphatidylcholines 95-107 high mobility group box 1 Homo sapiens 53-58 35477503-4 2022 METHODS: In this study, we developed oncolytic herpes simplex virus type 1 expressing HMGB1 protein (HSV-HMGB1) and investigated the cytotoxic effect of HSV-HMGB1 and its parental virus (HSV-ble) on three colorectal cancer cells (HCT116, SW480, and HT29) under normoxic (20% oxygen) and hypoxic (1% oxygen) conditions. Oxygen 299-305 high mobility group box 1 Homo sapiens 105-110 35477503-4 2022 METHODS: In this study, we developed oncolytic herpes simplex virus type 1 expressing HMGB1 protein (HSV-HMGB1) and investigated the cytotoxic effect of HSV-HMGB1 and its parental virus (HSV-ble) on three colorectal cancer cells (HCT116, SW480, and HT29) under normoxic (20% oxygen) and hypoxic (1% oxygen) conditions. Oxygen 275-281 high mobility group box 1 Homo sapiens 105-110 35440409-6 2022 MEASUREMENTS AND MAIN RESULTS: Significant reductions in high mobility group box-1 protein (HMGB-1), calprotectin, serum amyloid A, and granulocyte colony-stimulating factor (G-CSF), and an increase in insulin-like growth factor (IGF-1) occurred 1 month after ivacaftor. ivacaftor 260-269 high mobility group box 1 Homo sapiens 92-98 35510176-9 2023 Additionally, photothermal therapy and CDT-mediated immunogenic cell death of tumor cells can further enhance anti-tumor immunity via exposure of CRT, HMGB1 and ATP. 1,5,9-cyclododecatriene 39-42 high mobility group box 1 Homo sapiens 151-156 35496500-4 2022 In tumor cell line models, AmB induced ICD with its typical hallmarks of calreticulin (CALR) expression and release of high mobility group box 1 (HMGB1) as well as Adenosine 5"-triphosphate (ATP). Amphotericin B 27-30 high mobility group box 1 Homo sapiens 119-144 35496500-4 2022 In tumor cell line models, AmB induced ICD with its typical hallmarks of calreticulin (CALR) expression and release of high mobility group box 1 (HMGB1) as well as Adenosine 5"-triphosphate (ATP). Amphotericin B 27-30 high mobility group box 1 Homo sapiens 146-151 35432719-5 2022 Herein, glycyrrhizin (GL), an HMGB1 inhibitor, was used to investigate the relationship between ferroptosis and HMGB1. Glycyrrhizic Acid 8-20 high mobility group box 1 Homo sapiens 30-35 35432719-5 2022 Herein, glycyrrhizin (GL), an HMGB1 inhibitor, was used to investigate the relationship between ferroptosis and HMGB1. Glycyrrhizic Acid 22-24 high mobility group box 1 Homo sapiens 30-35 35432719-7 2022 Western blot analysis revealed that GL markedly suppressed the expression of HMGB1 and increased the level of GPX4 in the context of HIBD. Glycyrrhizic Acid 36-38 high mobility group box 1 Homo sapiens 77-82 35432719-12 2022 More importantly, GL may improve oxidative stress imbalance and mitochondrial damage, alleviate the downstream production of inflammatory factors, and ultimately reduce ferroptosis and damage to cortical neurons following HIBD via the HMGB1/GPX4 pathway. Glycyrrhizic Acid 18-20 high mobility group box 1 Homo sapiens 235-240 35432719-13 2022 In conclusion, we showed for the first time that GL could suppress the occurrence of neuronal ferroptosis and reduce neuronal loss in HIBD via the HMGB1/GPX4 pathway. Glycyrrhizic Acid 49-51 high mobility group box 1 Homo sapiens 147-152 35432719-14 2022 These findings highlight the potential of HMGB1 signaling antagonists to treat neuronal damage by suppressing ferroptosis, provide new and unique insights into GL as a neuroprotective agent, and suggest new prevention and treatment strategies for HIBD. Glycyrrhizic Acid 160-162 high mobility group box 1 Homo sapiens 42-47 35278873-6 2022 Both HMGB1 neutralizing antibody and sRAGE inhibited Ang II-induced calcium deposition. Calcium 68-75 high mobility group box 1 Homo sapiens 5-10 35316522-5 2022 However, whether PQ exposure affects HMGB1, alpha-synuclein, and autophagy function have rarely been reported. pq 17-19 high mobility group box 1 Homo sapiens 37-42 35534452-0 2022 Erratum: Vitexin Inhibits Gastric Cancer Growth and Metastasis through HMGB1-mediated Inactivation of the PI3K/AKT/mTOR/HIF-1alpha Signaling Pathway. vitexin 9-16 high mobility group box 1 Homo sapiens 71-76 35392631-13 2022 Expression levels of other complement pathway-related proteins (HMGB1, S100A8, S100A9, CRP, C4) were decreased by DTMP, although not significantly affected by SNI. 2',3'-dideoxy-2',3'-didehydrothymidine monophosphate 114-118 high mobility group box 1 Homo sapiens 64-69 35415314-0 2023 ROS-responsive 18beta-glycyrrhetic acid-conjugated polymeric nanoparticles mediate neuroprotection in ischemic stroke through HMGB1 inhibition and microglia polarization regulation. Reactive Oxygen Species 0-3 high mobility group box 1 Homo sapiens 126-131 35415314-0 2023 ROS-responsive 18beta-glycyrrhetic acid-conjugated polymeric nanoparticles mediate neuroprotection in ischemic stroke through HMGB1 inhibition and microglia polarization regulation. Glycyrrhetinic Acid 15-39 high mobility group box 1 Homo sapiens 126-131 35415314-4 2023 18beta-glycyrrhetinic acid (GA) is an effective intracellular inhibitor of HMGB1, but of poor water solubility and dose-dependent toxicity. 18alpha-glycyrrhetinic acid 0-26 high mobility group box 1 Homo sapiens 75-80 35415314-4 2023 18beta-glycyrrhetinic acid (GA) is an effective intracellular inhibitor of HMGB1, but of poor water solubility and dose-dependent toxicity. 18alpha-glycyrrhetinic acid 28-30 high mobility group box 1 Homo sapiens 75-80 35415314-5 2023 To overcome the shortcomings of GA delivery and to improve the efficacy of cerebral ischemia therapy, herein, we designed reactive oxygen species (ROS) responsive polymer-drug conjugate nanoparticles (DGA) to manipulate microglia polarization by suppressing the translocation of nuclear HMGB1. Reactive Oxygen Species 122-145 high mobility group box 1 Homo sapiens 287-292 35415314-5 2023 To overcome the shortcomings of GA delivery and to improve the efficacy of cerebral ischemia therapy, herein, we designed reactive oxygen species (ROS) responsive polymer-drug conjugate nanoparticles (DGA) to manipulate microglia polarization by suppressing the translocation of nuclear HMGB1. Reactive Oxygen Species 147-150 high mobility group box 1 Homo sapiens 287-292 35316522-0 2022 Paraquat Inhibits Autophagy Via Intensifying the Interaction Between HMGB1 and alpha-Synuclein. Paraquat 0-8 high mobility group box 1 Homo sapiens 69-74 35316522-6 2022 In this study, we found that PQ exposure impaired autophagy function via disturbing the complex formation of HMGB1 and Beclin1. pq 29-31 high mobility group box 1 Homo sapiens 109-114 35316522-7 2022 Moreover, the expression of alpha-synuclein is modulated by HMGB1 and the interaction between HMGB1 and alpha-synuclein was intensified by PQ exposure. pq 139-141 high mobility group box 1 Homo sapiens 94-99 35354479-0 2022 miR-495-3p depresses cell proliferation and migration by downregulating HMGB1 in colorectal cancer. mir-495-3p 0-10 high mobility group box 1 Homo sapiens 72-77 35224793-0 2022 Lucidone inhibits autophagy and MDR1 via HMGB1/RAGE/PI3K/Akt signaling pathway in pancreatic cancer cells. lucidone 0-8 high mobility group box 1 Homo sapiens 41-46 35224793-7 2022 The data showed that lucidone significantly promoted apoptotic cell death and inhibited the expression of autophagic proteins (Atg5, Beclin-1, LC3-II, and Vps34) and MDR1 by inhibiting the HMGB1/RAGE/PI3K/Akt axis in both MIA Paca-2 cells and MIA Paca-2GEMR cells (GEM-resistant cells). lucidone 21-29 high mobility group box 1 Homo sapiens 189-194 35224793-10 2022 Collectively, this study provided the underlying mechanism by which lucidone treatment inhibited HMGB1/RAGE-initiated PI3K/Akt/MDR1 signaling and consequently enhanced chemosensitivity in PDAC. lucidone 68-76 high mobility group box 1 Homo sapiens 97-102 35354479-9 2022 We finally demonstrated that miR-495-3p inhibited CRC cell proliferation by targeting HMGB1 in vitro and in vivo. mir-495-3p 29-39 high mobility group box 1 Homo sapiens 86-91 35340325-0 2022 Doxorubicin induced immune abnormalities and inflammatory responses via HMGB1, HIF1-alpha and VEGF pathway in progressive of cardiovascular damage. Doxorubicin 0-11 high mobility group box 1 Homo sapiens 72-77 35462475-0 2022 HMGB1-mediated autophagy promotes gefitinib resistance in human non-small cell lung cancer. Gefitinib 34-43 high mobility group box 1 Homo sapiens 0-5 35462475-4 2022 In this study, we found that HMGB1 is highly expressed in drug-resistant cells and confers to gefitinib resistance in NSCLC cells via activating autophagy process. Gefitinib 94-103 high mobility group box 1 Homo sapiens 29-34 35462475-5 2022 Gefitinib upregulates HMGB1 expression in time-dependent and dose-dependent manners in human NSCLC cells. Gefitinib 0-9 high mobility group box 1 Homo sapiens 22-27 35462475-6 2022 RNA interference-mediated knockdown of HMGB1 reduces PC9GR cell viability, induces apoptosis, and partially restores gefitinib sensitivity. Gefitinib 117-126 high mobility group box 1 Homo sapiens 39-44 35462475-7 2022 Mechanistic analyses indicate that elevated HMGB1 expression contributes to gefitinib resistance by inducing autophagy. Gefitinib 76-85 high mobility group box 1 Homo sapiens 44-49 35462475-8 2022 Thus, our results suggest that HMGB1 is an autophagy regulator and plays a key role in gefitinib resistance of NSCLC. Gefitinib 87-96 high mobility group box 1 Homo sapiens 31-36 35408786-0 2022 A Dual Anti-Inflammatory and Anti-Proliferative 3-Styrylchromone Derivative Synergistically Enhances the Anti-Cancer Effects of DNA-Damaging Agents on Colon Cancer Cells by Targeting HMGB1-RAGE-ERK1/2 Signaling. 3-styrylchromone 48-64 high mobility group box 1 Homo sapiens 183-188 35340325-7 2022 Results: Several articles were revealed that molecular mechanisms of the DOX in cardiomyocyte damage and related to HMGB1, HIF-1alpha and VEGF and may potential treatment and prevention to cardiovascular disease in DOX intervention. Doxorubicin 73-76 high mobility group box 1 Homo sapiens 116-121 35340325-8 2022 Conclusion: HMGB1, HIF-1alpha and VEGF has a pivotal regulator in DOX-induce cardiomyocyte damage and predominantly acts through different pathways. Doxorubicin 66-69 high mobility group box 1 Homo sapiens 12-17 35340325-9 2022 The role of HMGB1 in DOX-induced myocardial damage suggests that HMGB1 is a mediator of DOX-induced damage. Doxorubicin 21-24 high mobility group box 1 Homo sapiens 12-17 35340325-9 2022 The role of HMGB1 in DOX-induced myocardial damage suggests that HMGB1 is a mediator of DOX-induced damage. Doxorubicin 21-24 high mobility group box 1 Homo sapiens 65-70 35340325-9 2022 The role of HMGB1 in DOX-induced myocardial damage suggests that HMGB1 is a mediator of DOX-induced damage. Doxorubicin 88-91 high mobility group box 1 Homo sapiens 65-70 35154909-6 2022 Indeed, in vitro experiments performed with a cell-permeable small molecule KSPi closely related to the active payload released from the TWEAKR-KSPi-ADCs revealed that KSPi was capable of stimulating several hallmarks of immunogenic cell death (ICD) on three different human cancer cell lines: cellular release of adenosine triphosphate (ATP) and high mobility group B1 protein (HMGB1), exposure of calreticulin on the cell surface as well as a transcriptional type-I interferon response. kspi 76-80 high mobility group box 1 Homo sapiens 347-377 35315432-3 2022 Mechanistically, Panx1 on endothelial cells acts as a conduit for ATP release that stimulates macrophage activation via P2X7 receptors and mitochondrial DNA release to increase IL-1beta and HMGB1 secretion. Adenosine Triphosphate 66-69 high mobility group box 1 Homo sapiens 190-195 35328729-6 2022 AGDP not only inhibited the expression of TNF-alpha and IL-8 but also appeared to suppress the extracellular release of high-mobility group box 1 (HMGB1) by inhibiting the output of nuclear HMGB1 in cells. agdp 0-4 high mobility group box 1 Homo sapiens 120-145 35328729-6 2022 AGDP not only inhibited the expression of TNF-alpha and IL-8 but also appeared to suppress the extracellular release of high-mobility group box 1 (HMGB1) by inhibiting the output of nuclear HMGB1 in cells. agdp 0-4 high mobility group box 1 Homo sapiens 147-152 35328729-6 2022 AGDP not only inhibited the expression of TNF-alpha and IL-8 but also appeared to suppress the extracellular release of high-mobility group box 1 (HMGB1) by inhibiting the output of nuclear HMGB1 in cells. agdp 0-4 high mobility group box 1 Homo sapiens 190-195 35045357-6 2022 After treatment with anagliptin, the state of oxidative stress in macrophages was alleviated, with the downregulation of TLR4, HMGB-1, iNOS, and the declined release of NO. anagliptin 21-31 high mobility group box 1 Homo sapiens 127-133 35246019-9 2022 However, miR-142-3p inhibitor reverses the effect of circHECTD1 on all the above-mentioned aspects, including HMGB1 expression. mir-142-3p 9-19 high mobility group box 1 Homo sapiens 110-115 35193644-0 2022 Autophagy-based unconventional secretion of HMGB1 in glioblastoma promotes chemosensitivity to temozolomide through macrophage M1-like polarization. Temozolomide 95-107 high mobility group box 1 Homo sapiens 44-49 35193644-5 2022 METHODS: Cancer genome databases and paired-GB patient samples with or without TMZ treatment were used to assess the relationship between HMGB1 mRNA levels and overall patient survival. Temozolomide 79-82 high mobility group box 1 Homo sapiens 138-143 35193644-6 2022 The relationship between HMGB1 protein level and TMZ sensitivity was measured by immunohistochemistry, ELISA, Western blot and qRT-PCR. Temozolomide 49-52 high mobility group box 1 Homo sapiens 25-30 35193644-13 2022 Clinically, the elevated level of HMGB1 in sera may serve as a beneficial therapeutic-predictor for GB patients under TMZ treatment. Temozolomide 118-121 high mobility group box 1 Homo sapiens 34-39 35193644-15 2022 HMGB1 acts as a key regulator in the crosstalk between GB cells and tumor-suppressive M1-like TAMs in GB microenvironment and may be considered as an adjuvant for the chemotherapeutic agent TMZ. tams 94-98 high mobility group box 1 Homo sapiens 0-5 35193644-15 2022 HMGB1 acts as a key regulator in the crosstalk between GB cells and tumor-suppressive M1-like TAMs in GB microenvironment and may be considered as an adjuvant for the chemotherapeutic agent TMZ. Temozolomide 190-193 high mobility group box 1 Homo sapiens 0-5 35080850-3 2022 Au10SG10 caused the loss of mitochondrial metabolic activity, increased lipid peroxidation and translocation of an alarmin molecule, high mobility group box 1 (HMGB1), from the nucleus to the cytosol. au10sg10 0-8 high mobility group box 1 Homo sapiens 133-158 35080850-3 2022 Au10SG10 caused the loss of mitochondrial metabolic activity, increased lipid peroxidation and translocation of an alarmin molecule, high mobility group box 1 (HMGB1), from the nucleus to the cytosol. au10sg10 0-8 high mobility group box 1 Homo sapiens 160-165 35080850-5 2022 We show that Au10SG10 can bind directly to the defined sites of reduced, oxidized, and acetylated HMGB1. au10sg10 13-21 high mobility group box 1 Homo sapiens 98-103 35154909-6 2022 Indeed, in vitro experiments performed with a cell-permeable small molecule KSPi closely related to the active payload released from the TWEAKR-KSPi-ADCs revealed that KSPi was capable of stimulating several hallmarks of immunogenic cell death (ICD) on three different human cancer cell lines: cellular release of adenosine triphosphate (ATP) and high mobility group B1 protein (HMGB1), exposure of calreticulin on the cell surface as well as a transcriptional type-I interferon response. kspi 76-80 high mobility group box 1 Homo sapiens 379-384 35154909-6 2022 Indeed, in vitro experiments performed with a cell-permeable small molecule KSPi closely related to the active payload released from the TWEAKR-KSPi-ADCs revealed that KSPi was capable of stimulating several hallmarks of immunogenic cell death (ICD) on three different human cancer cell lines: cellular release of adenosine triphosphate (ATP) and high mobility group B1 protein (HMGB1), exposure of calreticulin on the cell surface as well as a transcriptional type-I interferon response. kspi 168-172 high mobility group box 1 Homo sapiens 347-377 35154909-6 2022 Indeed, in vitro experiments performed with a cell-permeable small molecule KSPi closely related to the active payload released from the TWEAKR-KSPi-ADCs revealed that KSPi was capable of stimulating several hallmarks of immunogenic cell death (ICD) on three different human cancer cell lines: cellular release of adenosine triphosphate (ATP) and high mobility group B1 protein (HMGB1), exposure of calreticulin on the cell surface as well as a transcriptional type-I interferon response. kspi 168-172 high mobility group box 1 Homo sapiens 379-384 35096875-14 2021 Taken together, our data here suggest that upregulation of DUOX2 plays a crucial role in ROS production, thereafter, induce HMGB1 release and cell death, which triggers ocular surface inflammation in DED. Reactive Oxygen Species 89-92 high mobility group box 1 Homo sapiens 124-129 34997217-0 2022 Deubiquitylase USP12 induces pro-survival autophagy and bortezomib resistance in multiple myeloma by stabilizing HMGB1. Bortezomib 56-66 high mobility group box 1 Homo sapiens 113-118 34997217-8 2022 Furthermore, basal autophagy activity associated with USP12/HMGB1 was elevated in bortezomib (BTZ)-resistant MM cell lines. Bortezomib 82-92 high mobility group box 1 Homo sapiens 60-65 34997217-9 2022 USP12 depletion, concomitant with a reduced expression of HMGB1, suppressed autophagy and increased the sensitivity of resistant cells to BTZ. Bortezomib 138-141 high mobility group box 1 Homo sapiens 58-63 34997217-10 2022 Collectively, our findings have identified an important role of the deubiquitylase USP12 in pro-survival autophagy and resultant BTZ resistance in MM by stabilizing HMGB1, suggesting that the USP12/HMGB1 axis might be pursued as a potential diagnostic and therapeutic target in human MM. Bortezomib 129-132 high mobility group box 1 Homo sapiens 165-170 34997217-10 2022 Collectively, our findings have identified an important role of the deubiquitylase USP12 in pro-survival autophagy and resultant BTZ resistance in MM by stabilizing HMGB1, suggesting that the USP12/HMGB1 axis might be pursued as a potential diagnostic and therapeutic target in human MM. Bortezomib 129-132 high mobility group box 1 Homo sapiens 198-203 35058496-9 2022 Treatment of the ZIKV-infected cells with dexamethasone (150 microM) reduced HMGB1 extracellular release in a dose-dependent manner, with a maximum reduction of 71 +- 5.84% (P < 0.01). Dexamethasone 42-55 high mobility group box 1 Homo sapiens 77-82 35058496-12 2022 These results suggest that translocation of HMGB1 occurred during ZIKV infection and inhibition of the translocation by dexamethasone coincided with a reduction in ZIKV replication. Dexamethasone 120-133 high mobility group box 1 Homo sapiens 44-49 35145563-12 2022 Further studies showed that circ0036602 could bind to miR-34-5p and miR-431-5p to regulate the expression of the target gene HMGB1. circ0036602 28-39 high mobility group box 1 Homo sapiens 125-130 35006556-14 2022 Finally, miR-142-3p interacted with HMGB1 directly and inhibition of HMGB1 protein expression mediated by MALAT1 knockdown was reversed by miR-142-3p inhibitor. mir-142-3p 9-19 high mobility group box 1 Homo sapiens 36-41 35006556-14 2022 Finally, miR-142-3p interacted with HMGB1 directly and inhibition of HMGB1 protein expression mediated by MALAT1 knockdown was reversed by miR-142-3p inhibitor. mir-142-3p 9-19 high mobility group box 1 Homo sapiens 69-74 35006556-14 2022 Finally, miR-142-3p interacted with HMGB1 directly and inhibition of HMGB1 protein expression mediated by MALAT1 knockdown was reversed by miR-142-3p inhibitor. mir-142-3p 139-149 high mobility group box 1 Homo sapiens 36-41 35006556-14 2022 Finally, miR-142-3p interacted with HMGB1 directly and inhibition of HMGB1 protein expression mediated by MALAT1 knockdown was reversed by miR-142-3p inhibitor. mir-142-3p 139-149 high mobility group box 1 Homo sapiens 69-74 35006556-17 2022 MALAT1 exerted its effects on IH-treated HUVECs via miR-142-3p/HMGB1. mir-142 52-59 high mobility group box 1 Homo sapiens 63-68 35442113-11 2022 RESULTS: miR-200b-3p directly target HMGB1. mir-200b 9-17 high mobility group box 1 Homo sapiens 37-42 35442113-16 2022 CONCLUSION: Decreased miR-200b-3p may function as a biomarker for the diagnosis and survival prognosis of MODS patients, and miR-200b-3p may be involved in the progression of acute paraquat-induced MODS via regulating inflammatory responses by targeting HMGB1. Paraquat 181-189 high mobility group box 1 Homo sapiens 254-259