PMID-sentid Pub_year Sent_text comp_official_name comp_offset protein_name organism prot_offset 32713345-15 2020 As a result, circPACRGL promoted CRC cell proliferation, migration and invasion, as well as differentiation of N1 to N2 neutrophils via miR-142-3p/miR-506-3p-TGF-beta1 axis. circpacrgl 13-23 microRNA 506 Homo sapiens 147-154 34987126-3 2021 Stimulation of human intestinal epithelial cells (Caco-2) with 1000 muM of 5-ASA suppressed the levels of miR-125b, miR-150, miR-155, miR-346 and miR-506, which are known to be involved in the regulation of colitis and/or colorectal cancer in patients with inflammatory bowel disease. Mesalamine 75-80 microRNA 506 Homo sapiens 146-153 35392987-0 2022 Knockdown of circ_0003928 ameliorates high glucose-induced dysfunction of human tubular epithelial cells through the miR-506-3p/HDAC4 pathway in diabetic nephropathy. Glucose 43-50 microRNA 506 Homo sapiens 117-124 33666835-0 2021 Alginic acid inhibits non-small cell lung cancer-induced angiogenesis via activating miR-506 expression. Alginic Acid 0-12 microRNA 506 Homo sapiens 85-92 33980925-8 2021 A functional in vitro analysis in Caco-2 cells showed that the induction of miR-506 activity by miR-506 mimic or GDCDA bile acid suppressed, whereas inhibition of miR-506 by miR-506 inhibitor or lipopolysaccharide (LPS) upregulated the expression of the examined target genes. gdcda bile acid 113-128 microRNA 506 Homo sapiens 76-83 33980925-10 2021 In ascending colons with PSC + UC, upregulation of miR-506 may result in failure of bicarbonate secretion and inhibition of p53, which predisposes to pro-tumorigenic transformation. Bicarbonates 84-95 microRNA 506 Homo sapiens 51-58 32833189-5 2021 The relationship between LINC01541 and miR-506-5p was verified in 17beta-E2-stimulated ESCs. 17beta-e2 66-75 microRNA 506 Homo sapiens 39-46 32833189-10 2021 Furthermore, silencing of miR-506-5p promoted apoptosis and suppressed the proliferation of 17beta-E2-treated ESCs. 17beta-e2 92-101 microRNA 506 Homo sapiens 26-33 34062009-10 2021 Moreover, we indicated that the ERK/Fos signaling pathway was intensively inactivated after overexpression of miR-506 and PENK both in vitro and in vivo, which was further validated by the ERK1/2-specific inhibitor SCH772984. SCH772984 215-224 microRNA 506 Homo sapiens 110-117 33000204-0 2020 KCNQ1OT1 contributes to sorafenib resistance and programmed death-ligand-1-mediated immune escape via sponging miR-506 in hepatocellular carcinoma cells. Sorafenib 24-33 microRNA 506 Homo sapiens 111-118 33000204-8 2020 KCNQ1OT1 and PD-L1 were found to be upregulated and miR-506 was downregulated in sorafenib-resistant HCC tissues and cells. Sorafenib 81-90 microRNA 506 Homo sapiens 52-59 33000204-11 2020 In addition, it was demonstrated that KCNQ1OT1 functioned as a competing endogenous RNA of miR-506 and increased PD-L1 expression in sorafenib-resistant HCC cells. Sorafenib 133-142 microRNA 506 Homo sapiens 91-98 33000204-12 2020 miR-506 inhibition abolished the effects of KCNQ1OT1 knockdown on sorafenib sensitivity, tumor growth, the tumor microenvironment and T-cell apoptosis. Sorafenib 66-75 microRNA 506 Homo sapiens 0-7 33000204-13 2020 In conclusion, KCNQ1OT1 knockdown inhibited sorafenib resistance and PD-L1-mediated immune escape by sponging miR-506 in sorafenib-resistant HCC cells. Sorafenib 121-130 microRNA 506 Homo sapiens 110-117 32944040-6 2020 Next, miR-506 was transfected with concentrations of 50 and 100 nM with Lipofectamine 2000. Lipofectamine 72-90 microRNA 506 Homo sapiens 6-13 31087496-7 2019 Moreover, a further miRNAs screen by qRT-PCR indicated that miR-124-3p and miR-506-3p (miR-124/506) were remarkably reduced in sorafenib insensitive patients. Sorafenib 127-136 microRNA 506 Homo sapiens 87-98 32099459-0 2019 circPCNX and Pecanex Promote Hepatocellular Carcinoma Cell Viability by Inhibiting miR-506. circpcnx 0-8 microRNA 506 Homo sapiens 83-90 31515847-12 2019 TRPV1 channel stimulation and ROS generation, which was related to osteoclastogenesis, were reduced via miR-506 depletion. Reactive Oxygen Species 30-33 microRNA 506 Homo sapiens 104-111 31515847-13 2019 miR-506 modulated osteoclastogenesis by targeting SIRT1 expression in part through modulation of the TRPV1 channel, ROS production, and TNF-alpha. Reactive Oxygen Species 116-119 microRNA 506 Homo sapiens 0-7 31087496-9 2019 Interestingly, we finally proved that the sorafenib resistant cells regained sensitivity for sorafenib by EZH2 intervention with miR-124/506 overexpression or EZH2 inhibitor treatment in vitro and in vivo, which will lead to the decreased tri-methylation at lysine 27 of histone H3 (H3K27me3) and increased acetylated lysine 27 of histone H3 (H3K27ac) levels. Sorafenib 42-51 microRNA 506 Homo sapiens 129-140 31087496-9 2019 Interestingly, we finally proved that the sorafenib resistant cells regained sensitivity for sorafenib by EZH2 intervention with miR-124/506 overexpression or EZH2 inhibitor treatment in vitro and in vivo, which will lead to the decreased tri-methylation at lysine 27 of histone H3 (H3K27me3) and increased acetylated lysine 27 of histone H3 (H3K27ac) levels. Sorafenib 93-102 microRNA 506 Homo sapiens 129-140 31087496-9 2019 Interestingly, we finally proved that the sorafenib resistant cells regained sensitivity for sorafenib by EZH2 intervention with miR-124/506 overexpression or EZH2 inhibitor treatment in vitro and in vivo, which will lead to the decreased tri-methylation at lysine 27 of histone H3 (H3K27me3) and increased acetylated lysine 27 of histone H3 (H3K27ac) levels. Lysine 258-264 microRNA 506 Homo sapiens 129-140 31087496-9 2019 Interestingly, we finally proved that the sorafenib resistant cells regained sensitivity for sorafenib by EZH2 intervention with miR-124/506 overexpression or EZH2 inhibitor treatment in vitro and in vivo, which will lead to the decreased tri-methylation at lysine 27 of histone H3 (H3K27me3) and increased acetylated lysine 27 of histone H3 (H3K27ac) levels. Lysine 318-324 microRNA 506 Homo sapiens 129-140 30105876-6 2018 Moreover, DQ786243 reversed the inhibitory effect of miR-506 on the growth of ovarian cancer cells, which might be involved in the derepression of cAMP responsive element binding protein 1 (CREB1) expression. dq786243 10-18 microRNA 506 Homo sapiens 53-60 30669957-8 2019 The alterations induced by miR-506-3p mimics were partly reversed by SPHK1 overexpression or treatment of rapamycin. Sirolimus 106-115 microRNA 506 Homo sapiens 27-34 30105876-9 2018 Taken together, our study showed for the first time that DQ786243 interacted with miR-506 and promoted progression of ovarian cancer via targeting CREB1 in ovarian cancer. dq786243 57-65 microRNA 506 Homo sapiens 82-89 28849090-9 2017 H2O2 markedly upregulated the expression levels of KLF4 and KLF5, and downregulated the expression levels of miR-506 and miR-124 in the HCMs. Hydrogen Peroxide 0-4 microRNA 506 Homo sapiens 109-116 28922472-8 2018 In cholangiocytes, miR-506 (1) induced dedifferentiation with down-regulation of biliary and epithelial markers together with up-regulation of mesenchymal, proinflammatory, and profibrotic markers; (2) impaired cell proliferation and adhesion; (3) increased oxidative and endoplasmic reticulum stress; (4) caused DNA damage; and (5) sensitized to caspase-3-dependent apoptosis induced by cytotoxic bile acids. Bile Acids and Salts 398-408 microRNA 506 Homo sapiens 19-26 28962898-4 2018 Recently, it has been shown that microRNA-506 (miR-506) on chromosome X is up-regulated in PBC cholangiocytes and suppresses AE2 expression, which sensitizes cholangiocytes to bile salt-induced apoptosis by activating soluble adenylyl cyclase (sAC), an evolutionarily conserved bicarbonate sensor. Bile Acids and Salts 176-185 microRNA 506 Homo sapiens 33-45 28962898-4 2018 Recently, it has been shown that microRNA-506 (miR-506) on chromosome X is up-regulated in PBC cholangiocytes and suppresses AE2 expression, which sensitizes cholangiocytes to bile salt-induced apoptosis by activating soluble adenylyl cyclase (sAC), an evolutionarily conserved bicarbonate sensor. Bile Acids and Salts 176-185 microRNA 506 Homo sapiens 47-54 28962898-4 2018 Recently, it has been shown that microRNA-506 (miR-506) on chromosome X is up-regulated in PBC cholangiocytes and suppresses AE2 expression, which sensitizes cholangiocytes to bile salt-induced apoptosis by activating soluble adenylyl cyclase (sAC), an evolutionarily conserved bicarbonate sensor. Bicarbonates 278-289 microRNA 506 Homo sapiens 33-45 28962898-4 2018 Recently, it has been shown that microRNA-506 (miR-506) on chromosome X is up-regulated in PBC cholangiocytes and suppresses AE2 expression, which sensitizes cholangiocytes to bile salt-induced apoptosis by activating soluble adenylyl cyclase (sAC), an evolutionarily conserved bicarbonate sensor. Bicarbonates 278-289 microRNA 506 Homo sapiens 47-54 28849090-12 2017 The overexpression of miR-506 and miR-214 reversed the H2O2-induced apoptosis and increase of ROS in the HCMs. Reactive Oxygen Species 94-97 microRNA 506 Homo sapiens 22-29 28849090-13 2017 In conclusion, the overexpression of miR-506 and miR-214 were confirmed to have a protective effect against H2O2-induced HCM injury by suppressing the expression of KLF4 and KLF5. Hydrogen Peroxide 108-112 microRNA 506 Homo sapiens 37-44 28849090-11 2017 The overexpression of miR-506 and miR-124 inhibited the H2O2-induced upregulation of KLF4 and KLF5 in the HCMs. Hydrogen Peroxide 56-60 microRNA 506 Homo sapiens 22-29 28849090-12 2017 The overexpression of miR-506 and miR-214 reversed the H2O2-induced apoptosis and increase of ROS in the HCMs. Hydrogen Peroxide 55-59 microRNA 506 Homo sapiens 22-29 28217977-0 2017 miR-506 enhances the sensitivity of human colorectal cancer cells to oxaliplatin by suppressing MDR1/P-gp expression. Oxaliplatin 69-80 microRNA 506 Homo sapiens 0-7 28217977-2 2017 The aim of this study was to elucidate the mechanism by which miR-506 reverses oxaliplatin chemoresistance in CRC. Oxaliplatin 79-90 microRNA 506 Homo sapiens 62-69 28217977-12 2017 miR-506 overexpression inhibited cell growth and increased oxaliplatin-induced cell apoptosis in HCT116-OxR cells, as shown via FCM and apoptosis assay. Oxaliplatin 59-70 microRNA 506 Homo sapiens 0-7 28217977-15 2017 CONCLUSION: Taken together, the findings of our study demonstrate that miR-506 overexpression in HCT116-OxR cells enhances oxaliplatin sensitivity by inhibiting MDR1/P-gp expression via down-regulation of the Wnt/beta-catenin pathway and thus provide a rationale for the development of miRNA-based strategies to reverse oxaliplatin resistance in CRC cells. Oxaliplatin 123-134 microRNA 506 Homo sapiens 71-78 28217977-15 2017 CONCLUSION: Taken together, the findings of our study demonstrate that miR-506 overexpression in HCT116-OxR cells enhances oxaliplatin sensitivity by inhibiting MDR1/P-gp expression via down-regulation of the Wnt/beta-catenin pathway and thus provide a rationale for the development of miRNA-based strategies to reverse oxaliplatin resistance in CRC cells. Oxaliplatin 320-331 microRNA 506 Homo sapiens 71-78 27371108-8 2016 Moreover, miR-506 expression was negatively associated with pathologic T category (P = 0.004) and lymph node metastasis (P = 0.033), suggesting that miR-506 might inhibit the progression of PDAC. pdac 190-194 microRNA 506 Homo sapiens 10-17 28121485-10 2017 These findings expand the known mechanisms of MIR506-mediated tumor suppression to activation of autophagy-related cell death and suggest a strategy for using MIR506 as an anti-STAT3 approach to PDAC treatment. pdac 195-199 microRNA 506 Homo sapiens 46-52 28121485-10 2017 These findings expand the known mechanisms of MIR506-mediated tumor suppression to activation of autophagy-related cell death and suggest a strategy for using MIR506 as an anti-STAT3 approach to PDAC treatment. pdac 195-199 microRNA 506 Homo sapiens 159-165 27371108-8 2016 Moreover, miR-506 expression was negatively associated with pathologic T category (P = 0.004) and lymph node metastasis (P = 0.033), suggesting that miR-506 might inhibit the progression of PDAC. pdac 190-194 microRNA 506 Homo sapiens 149-156 27371108-9 2016 CONCLUSIONS: Our results suggest that miR-506 either plays a role as an oncogene in the tumorigenesis and a tumor suppressor in the progression or serves as a house-keeping, tumor-suppressing miRNA, whose expression can be activated by oncogenic signals in early development to hinder the progression of PDAC. pdac 304-308 microRNA 506 Homo sapiens 38-45 26398880-4 2015 Subsequently, gain-of-function and loss-of-function experiments were performed in vitro, and the results from MTT assay, Transwell-Matrigel invasion assay and cell adhesion assay revealed that miR-506 suppresses cell proliferation, invasion and adhesion of breast cancer cells. monooxyethylene trimethylolpropane tristearate 110-113 microRNA 506 Homo sapiens 193-200 27114802-0 2016 miR-506 inhibits cell proliferation and invasion by targeting TET family in colorectal cancer. tet 62-65 microRNA 506 Homo sapiens 0-7 27114802-8 2016 We showed that TET family member degradation by miR-506 inhibits cell proliferation and invasion in colorectal cancer. tet 15-18 microRNA 506 Homo sapiens 48-55 27114802-9 2016 CONCLUSION: Through this study, we advance our understanding of the expression levels TETs and miR-506 in CRC and further clarify the internal regulatory mechanism of miR-506 by targeting TET during CRC processes. tetramethylenedisulfotetramine 86-90 microRNA 506 Homo sapiens 167-174 27114802-9 2016 CONCLUSION: Through this study, we advance our understanding of the expression levels TETs and miR-506 in CRC and further clarify the internal regulatory mechanism of miR-506 by targeting TET during CRC processes. tet 86-89 microRNA 506 Homo sapiens 167-174 24469051-2 2015 Here we report that miR-506 upregulation occurs in 83% of lung cancer patients (156 cases), and its expression highly correlates with ROS. Reactive Oxygen Species 134-137 microRNA 506 Homo sapiens 20-27 26369335-4 2015 Further functional study demonstrated that miR-506 could augment the response to cisplatin and olaparib through targeting RAD51 and suppressing homologous recombination in a panel of ovarian cancer cell lines. Cisplatin 81-90 microRNA 506 Homo sapiens 43-50 26369335-4 2015 Further functional study demonstrated that miR-506 could augment the response to cisplatin and olaparib through targeting RAD51 and suppressing homologous recombination in a panel of ovarian cancer cell lines. olaparib 95-103 microRNA 506 Homo sapiens 43-50 26369335-5 2015 Systemic delivery of miR-506 in an orthotopic ovarian cancer mouse model significantly augmented the cisplatin response, thus recapitulating the clinical observation. Cisplatin 101-110 microRNA 506 Homo sapiens 21-28 24469051-3 2015 Ectopic expression of miR-506 inhibits NF-kappaB p65 expression, induces ROS accumulation and then activates p53 to suppress lung cancer cell viability, but not in normal cells. Reactive Oxygen Species 73-76 microRNA 506 Homo sapiens 22-29 24469051-4 2015 Interestingly, p53 promotes miR-506 expression level, indicating that miR-506 mediates cross talk between p53, NF-kappaB p65 and ROS. Reactive Oxygen Species 129-132 microRNA 506 Homo sapiens 28-35 24469051-4 2015 Interestingly, p53 promotes miR-506 expression level, indicating that miR-506 mediates cross talk between p53, NF-kappaB p65 and ROS. Reactive Oxygen Species 129-132 microRNA 506 Homo sapiens 70-77 21726609-0 2011 The role of miR-506 in transformed 16HBE cells induced by anti-benzo[a]pyrene-trans-7,8-dihydrodiol-9,10-epoxide. 16hbe 35-40 microRNA 506 Homo sapiens 12-19 25378392-10 2015 Histamine-induced, InsP3-mediated Ca(2+) signals were decreased by 50% in stable miR-506 cells compared with controls. Histamine 0-9 microRNA 506 Homo sapiens 81-88 21726609-0 2011 The role of miR-506 in transformed 16HBE cells induced by anti-benzo[a]pyrene-trans-7,8-dihydrodiol-9,10-epoxide. benzo[a]pyrene-trans-7,8-dihydrodiol-9,10-epoxide 63-112 microRNA 506 Homo sapiens 12-19