PMID-sentid Pub_year Sent_text compound_name comp_offset prot_official_name organism prot_offset 30886645-5 2019 According to the results from imaging analyses and bioanalytical experiments, acetaminophen (APAP) caused cytotoxicity in the zone-3 region of the 3D hepatic zonal channel, and the levels of nonphosphorylated beta-catenin, CYP2E, and apoptotic proteins were remarkably increased in the zone-3-like region. 4-amino-N-acetyl-N-methylaniline 93-97 cytochrome P450 family 2 subfamily E member 1 Homo sapiens 223-228 31029706-2 2019 Mitochondrial SH3BP5 (also called SAB) and phosphorylation of c-Jun N-terminal kinase (JNK) mediate the hepatotoxic effects of APAP. 4-amino-N-acetyl-N-methylaniline 127-131 SH3-domain binding protein 5 (BTK-associated) Mus musculus 14-20 31029706-2 2019 Mitochondrial SH3BP5 (also called SAB) and phosphorylation of c-Jun N-terminal kinase (JNK) mediate the hepatotoxic effects of APAP. 4-amino-N-acetyl-N-methylaniline 127-131 mitogen-activated protein kinase 8 Mus musculus 62-85 31029706-2 2019 Mitochondrial SH3BP5 (also called SAB) and phosphorylation of c-Jun N-terminal kinase (JNK) mediate the hepatotoxic effects of APAP. 4-amino-N-acetyl-N-methylaniline 127-131 mitogen-activated protein kinase 8 Mus musculus 87-90 31029706-15 2019 CONCLUSIONS: In studies of mice, we found that upregulation of STARD1 following ER stress mediates APAP hepatoxicity via SH3BP5 and phosphorylation of JNK1 and JNK2. 4-amino-N-acetyl-N-methylaniline 99-103 steroidogenic acute regulatory protein Mus musculus 63-69 31029706-15 2019 CONCLUSIONS: In studies of mice, we found that upregulation of STARD1 following ER stress mediates APAP hepatoxicity via SH3BP5 and phosphorylation of JNK1 and JNK2. 4-amino-N-acetyl-N-methylaniline 99-103 SH3-domain binding protein 5 (BTK-associated) Mus musculus 121-127 30886645-5 2019 According to the results from imaging analyses and bioanalytical experiments, acetaminophen (APAP) caused cytotoxicity in the zone-3 region of the 3D hepatic zonal channel, and the levels of nonphosphorylated beta-catenin, CYP2E, and apoptotic proteins were remarkably increased in the zone-3-like region. 4-amino-N-acetyl-N-methylaniline 93-97 catenin beta 1 Homo sapiens 209-221 31029706-15 2019 CONCLUSIONS: In studies of mice, we found that upregulation of STARD1 following ER stress mediates APAP hepatoxicity via SH3BP5 and phosphorylation of JNK1 and JNK2. 4-amino-N-acetyl-N-methylaniline 99-103 mitogen-activated protein kinase 8 Mus musculus 151-155 31029706-15 2019 CONCLUSIONS: In studies of mice, we found that upregulation of STARD1 following ER stress mediates APAP hepatoxicity via SH3BP5 and phosphorylation of JNK1 and JNK2. 4-amino-N-acetyl-N-methylaniline 99-103 mitogen-activated protein kinase 9 Mus musculus 160-164 29374630-8 2019 Mice lacking TREM-2 exhibited heightened liver damage and inflammation during acute and repetitive carbon tetrachloride and acetaminophen (APAP) intoxication, the latter of which TREM-2 deficiency was remarkably associated with worsened survival. 4-amino-N-acetyl-N-methylaniline 139-143 triggering receptor expressed on myeloid cells 2 Mus musculus 13-19 29374630-9 2019 Liver damage in Trem-2-/- mice following chronic injury and APAP challenge was associated with elevated hepatic lipid peroxidation and macrophage content. 4-amino-N-acetyl-N-methylaniline 60-64 triggering receptor expressed on myeloid cells 2 Mus musculus 16-22 30425454-8 2018 In vivo, the vitamins and their combination effectively reduced APAP-induced serum liver enzymes levels, namely ALT, AST, and ALP, and also attenuated oxidative stress and lipids peroxidation confirmed by the results of glutathione, superoxide dismutase, and maloondialdehyde. 4-amino-N-acetyl-N-methylaniline 64-68 PDZ and LIM domain 3 Rattus norvegicus 126-129 31933721-10 2019 Nrf2-silenced LOS cells were sensitive to APAP-induced injury, while Hype did not exhibit any further effects on LO2 cells, which demonstrate the critical role of Nrf2 in this process. 4-amino-N-acetyl-N-methylaniline 42-46 NFE2 like bZIP transcription factor 2 Homo sapiens 0-4 31933721-11 2019 Taken together, our results demonstrated the ability of Hype to inhibit APAP-induced acute hepatic injury and its potential use in the treatment of Nrf2-associated diseases. 4-amino-N-acetyl-N-methylaniline 72-76 NFE2 like bZIP transcription factor 2 Homo sapiens 148-152 30546306-0 2018 Isoorientin Ameliorates APAP-Induced Hepatotoxicity via Activation Nrf2 Antioxidative Pathway: The Involvement of AMPK/Akt/GSK3beta. 4-amino-N-acetyl-N-methylaniline 24-28 nuclear factor, erythroid derived 2, like 2 Mus musculus 67-71 30546306-0 2018 Isoorientin Ameliorates APAP-Induced Hepatotoxicity via Activation Nrf2 Antioxidative Pathway: The Involvement of AMPK/Akt/GSK3beta. 4-amino-N-acetyl-N-methylaniline 24-28 thymoma viral proto-oncogene 1 Mus musculus 119-122 30546306-0 2018 Isoorientin Ameliorates APAP-Induced Hepatotoxicity via Activation Nrf2 Antioxidative Pathway: The Involvement of AMPK/Akt/GSK3beta. 4-amino-N-acetyl-N-methylaniline 24-28 glycogen synthase kinase 3 beta Mus musculus 123-131 30546306-6 2018 We found that Iso treatment significantly reduced APAP-induced hepatotoxicity by reducing the lethality, histopathological liver changes, and alanine transaminase (ALT) and aspartate aminotransferase (AST) levels in serum. 4-amino-N-acetyl-N-methylaniline 50-54 glutamic pyruvic transaminase, soluble Mus musculus 142-162 30546306-12 2018 In summary, Iso ameliorated APAP-induced hepatotoxicity by activating Nrf2 via the AMPK/Akt/GSK3beta pathway. 4-amino-N-acetyl-N-methylaniline 28-32 nuclear factor, erythroid derived 2, like 2 Mus musculus 70-74 30546306-12 2018 In summary, Iso ameliorated APAP-induced hepatotoxicity by activating Nrf2 via the AMPK/Akt/GSK3beta pathway. 4-amino-N-acetyl-N-methylaniline 28-32 thymoma viral proto-oncogene 1 Mus musculus 88-91 30546306-12 2018 In summary, Iso ameliorated APAP-induced hepatotoxicity by activating Nrf2 via the AMPK/Akt/GSK3beta pathway. 4-amino-N-acetyl-N-methylaniline 28-32 glycogen synthase kinase 3 beta Mus musculus 92-100 30006487-4 2018 We also provide novel evidence that APAP-induced glutathionylation of carnitine O-palmitoyltransferase 1 (CPT1) and voltage-dependent anion-selective channel protein 1 are respectively involved in inhibition of fatty acid beta-oxidation and opening of the mitochondrial permeability transition pore. 4-amino-N-acetyl-N-methylaniline 36-40 carnitine palmitoyltransferase 1A Homo sapiens 70-104 30006487-4 2018 We also provide novel evidence that APAP-induced glutathionylation of carnitine O-palmitoyltransferase 1 (CPT1) and voltage-dependent anion-selective channel protein 1 are respectively involved in inhibition of fatty acid beta-oxidation and opening of the mitochondrial permeability transition pore. 4-amino-N-acetyl-N-methylaniline 36-40 carnitine palmitoyltransferase 1A Homo sapiens 106-110 30006487-4 2018 We also provide novel evidence that APAP-induced glutathionylation of carnitine O-palmitoyltransferase 1 (CPT1) and voltage-dependent anion-selective channel protein 1 are respectively involved in inhibition of fatty acid beta-oxidation and opening of the mitochondrial permeability transition pore. 4-amino-N-acetyl-N-methylaniline 36-40 voltage dependent anion channel 1 Homo sapiens 116-167 28442342-1 2017 Thioredoxin reductase 1 (TXNRD1) is associated with susceptibility to acetaminophen (APAP)-induced liver damage. 4-amino-N-acetyl-N-methylaniline 85-89 thioredoxin reductase 1 Homo sapiens 0-23 28442342-9 2017 In addition, APAP treatment significantly increased TXNRD1 expression in MsrA-/- hepatocytes, while no significant change was observed in MsrA+/+ cells. 4-amino-N-acetyl-N-methylaniline 13-17 thioredoxin reductase 1 Homo sapiens 52-58 28442342-9 2017 In addition, APAP treatment significantly increased TXNRD1 expression in MsrA-/- hepatocytes, while no significant change was observed in MsrA+/+ cells. 4-amino-N-acetyl-N-methylaniline 13-17 methionine sulfoxide reductase A Homo sapiens 73-77 30309416-1 2018 OBJECTIVE: To investigate whether GLT25D2 gene regulates autophagy in acetaminophen (APAP)-induced hepatotoxicity injury. 4-amino-N-acetyl-N-methylaniline 85-89 collagen beta(1-O)galactosyltransferase 2 Mus musculus 34-41 30309416-14 2018 RESULTS: (1) In vivo experiment: compared with the corresponding PBS control group, the expressions of the positive-associated proteins ATG5, ATG7 and LC3-II in liver tissue of the APAP intervention group were down-regulated in the wild-type and GLT25D2-/- mice, while the expression of the negative correlation protein P62 was up-regulated, indicating that the overall level of autophagy decreased after treatment with APAP. 4-amino-N-acetyl-N-methylaniline 181-185 autophagy related 5 Mus musculus 136-140 30309416-14 2018 RESULTS: (1) In vivo experiment: compared with the corresponding PBS control group, the expressions of the positive-associated proteins ATG5, ATG7 and LC3-II in liver tissue of the APAP intervention group were down-regulated in the wild-type and GLT25D2-/- mice, while the expression of the negative correlation protein P62 was up-regulated, indicating that the overall level of autophagy decreased after treatment with APAP. 4-amino-N-acetyl-N-methylaniline 181-185 autophagy related 7 Mus musculus 142-146 30309416-14 2018 RESULTS: (1) In vivo experiment: compared with the corresponding PBS control group, the expressions of the positive-associated proteins ATG5, ATG7 and LC3-II in liver tissue of the APAP intervention group were down-regulated in the wild-type and GLT25D2-/- mice, while the expression of the negative correlation protein P62 was up-regulated, indicating that the overall level of autophagy decreased after treatment with APAP. 4-amino-N-acetyl-N-methylaniline 181-185 microtubule-associated protein 1 light chain 3 alpha Mus musculus 151-154 30309416-14 2018 RESULTS: (1) In vivo experiment: compared with the corresponding PBS control group, the expressions of the positive-associated proteins ATG5, ATG7 and LC3-II in liver tissue of the APAP intervention group were down-regulated in the wild-type and GLT25D2-/- mice, while the expression of the negative correlation protein P62 was up-regulated, indicating that the overall level of autophagy decreased after treatment with APAP. 4-amino-N-acetyl-N-methylaniline 181-185 collagen beta(1-O)galactosyltransferase 2 Mus musculus 246-253 30309416-14 2018 RESULTS: (1) In vivo experiment: compared with the corresponding PBS control group, the expressions of the positive-associated proteins ATG5, ATG7 and LC3-II in liver tissue of the APAP intervention group were down-regulated in the wild-type and GLT25D2-/- mice, while the expression of the negative correlation protein P62 was up-regulated, indicating that the overall level of autophagy decreased after treatment with APAP. 4-amino-N-acetyl-N-methylaniline 181-185 nucleoporin 62 Mus musculus 320-323 30309416-21 2018 Fluorescence microscopy showed that GFP-LC3 positive cells in both wild-type and GLT25D2-/- mice hepatocytes were significantly increased after APAP intervention as compared with those of PBS control group, and the proportion of GFP-LC3 positive cells in GLT25D2-/- mice was significantly higher than that in wild-type mice (0.64+-0.08 vs. 0.36+-0.05, P > 0.05). 4-amino-N-acetyl-N-methylaniline 144-148 microtubule-associated protein 1 light chain 3 alpha Mus musculus 40-43 30309416-21 2018 Fluorescence microscopy showed that GFP-LC3 positive cells in both wild-type and GLT25D2-/- mice hepatocytes were significantly increased after APAP intervention as compared with those of PBS control group, and the proportion of GFP-LC3 positive cells in GLT25D2-/- mice was significantly higher than that in wild-type mice (0.64+-0.08 vs. 0.36+-0.05, P > 0.05). 4-amino-N-acetyl-N-methylaniline 144-148 collagen beta(1-O)galactosyltransferase 2 Mus musculus 81-88 30309416-21 2018 Fluorescence microscopy showed that GFP-LC3 positive cells in both wild-type and GLT25D2-/- mice hepatocytes were significantly increased after APAP intervention as compared with those of PBS control group, and the proportion of GFP-LC3 positive cells in GLT25D2-/- mice was significantly higher than that in wild-type mice (0.64+-0.08 vs. 0.36+-0.05, P > 0.05). 4-amino-N-acetyl-N-methylaniline 144-148 microtubule-associated protein 1 light chain 3 alpha Mus musculus 233-236 30309416-21 2018 Fluorescence microscopy showed that GFP-LC3 positive cells in both wild-type and GLT25D2-/- mice hepatocytes were significantly increased after APAP intervention as compared with those of PBS control group, and the proportion of GFP-LC3 positive cells in GLT25D2-/- mice was significantly higher than that in wild-type mice (0.64+-0.08 vs. 0.36+-0.05, P > 0.05). 4-amino-N-acetyl-N-methylaniline 144-148 collagen beta(1-O)galactosyltransferase 2 Mus musculus 255-262 29192844-12 2018 However, in the APAP group, iNOS immunostaining was most evident in pericentral hepatocytes. 4-amino-N-acetyl-N-methylaniline 16-20 nitric oxide synthase 2 Rattus norvegicus 28-32 29192844-13 2018 In the same areas in APAP+RSV group, intensity of iNOS immunostaining decreased. 4-amino-N-acetyl-N-methylaniline 21-25 nitric oxide synthase 2 Rattus norvegicus 50-54 29192844-14 2018 A significant increase in MDA and decreases in GSH level, CAT, and SOD activity indicated that APAP-induced hepatotoxicity was mediated through oxidative stress. 4-amino-N-acetyl-N-methylaniline 95-99 catalase Rattus norvegicus 58-61 29042982-4 2017 GSTA1 protein content was 3.8, 1.3 and 2.6 times lower in the CCl4, APAP and ethanol model groups, respectively. 4-amino-N-acetyl-N-methylaniline 68-72 glutathione S-transferase, alpha 1 (Ya) Mus musculus 0-5 29042982-5 2017 Furthermore, GSTA1 mRNA expression levels decreased by 4.9, 2.1 and 3.7 times in the CCl4, APAP and ethanol model groups, respectively. 4-amino-N-acetyl-N-methylaniline 91-95 glutathione S-transferase, alpha 1 (Ya) Mus musculus 13-18 28442342-10 2017 Overexpression of MsrA reduced APAP-induced cytotoxicity and TXNRD1 expression levels in APAP-treated MsrA-/- hepatocytes. 4-amino-N-acetyl-N-methylaniline 31-35 methionine sulfoxide reductase A Homo sapiens 18-22 28442342-10 2017 Overexpression of MsrA reduced APAP-induced cytotoxicity and TXNRD1 expression levels in APAP-treated MsrA-/- hepatocytes. 4-amino-N-acetyl-N-methylaniline 89-93 methionine sulfoxide reductase A Homo sapiens 18-22 28442342-10 2017 Overexpression of MsrA reduced APAP-induced cytotoxicity and TXNRD1 expression levels in APAP-treated MsrA-/- hepatocytes. 4-amino-N-acetyl-N-methylaniline 89-93 thioredoxin reductase 1 Homo sapiens 61-67 28442342-10 2017 Overexpression of MsrA reduced APAP-induced cytotoxicity and TXNRD1 expression levels in APAP-treated MsrA-/- hepatocytes. 4-amino-N-acetyl-N-methylaniline 89-93 methionine sulfoxide reductase A Homo sapiens 102-106 28442342-1 2017 Thioredoxin reductase 1 (TXNRD1) is associated with susceptibility to acetaminophen (APAP)-induced liver damage. 4-amino-N-acetyl-N-methylaniline 85-89 thioredoxin reductase 1 Homo sapiens 25-31 28442342-4 2017 In this study, we used primary hepatocytes to investigate the underlying mechanism of the protective effect of MsrA against APAP-induced hepatotoxicity. 4-amino-N-acetyl-N-methylaniline 124-128 methionine sulfoxide reductase A Homo sapiens 111-115 28442342-5 2017 MsrA gene-deleted (MsrA-/-) hepatocytes showed higher susceptibility to APAP-induced cytotoxicity than wild-type (MsrA+/+) cells, consistent with our previous in vivo results. 4-amino-N-acetyl-N-methylaniline 72-76 methionine sulfoxide reductase A Homo sapiens 0-4 28442342-5 2017 MsrA gene-deleted (MsrA-/-) hepatocytes showed higher susceptibility to APAP-induced cytotoxicity than wild-type (MsrA+/+) cells, consistent with our previous in vivo results. 4-amino-N-acetyl-N-methylaniline 72-76 methionine sulfoxide reductase A Homo sapiens 19-23 28442342-5 2017 MsrA gene-deleted (MsrA-/-) hepatocytes showed higher susceptibility to APAP-induced cytotoxicity than wild-type (MsrA+/+) cells, consistent with our previous in vivo results. 4-amino-N-acetyl-N-methylaniline 72-76 methionine sulfoxide reductase A Homo sapiens 19-23 28442342-7 2017 APAP treatment increased Nrf2 activation more profoundly in MsrA-/- than in MsrA+/+ hepatocytes. 4-amino-N-acetyl-N-methylaniline 0-4 NFE2 like bZIP transcription factor 2 Homo sapiens 25-29 28442342-7 2017 APAP treatment increased Nrf2 activation more profoundly in MsrA-/- than in MsrA+/+ hepatocytes. 4-amino-N-acetyl-N-methylaniline 0-4 methionine sulfoxide reductase A Homo sapiens 60-64 28442342-7 2017 APAP treatment increased Nrf2 activation more profoundly in MsrA-/- than in MsrA+/+ hepatocytes. 4-amino-N-acetyl-N-methylaniline 0-4 methionine sulfoxide reductase A Homo sapiens 76-80 28330995-7 2017 As such, global or liver-specific deletion of FcRn results in resistance to APAP-induced liver injury through increased albumin loss into the bile and increased intracellular albumin scavenging of reactive oxygen species. 4-amino-N-acetyl-N-methylaniline 76-80 Fc fragment of IgG receptor and transporter Mus musculus 46-50 26304691-3 2015 Our in vitro experiments indicated that the formation of these novel APAP metabolites is, at least in part, attributable to the interaction of CysSSnSH produced by CSE and GSH persulfide with APAP-derived NAPQI. 4-amino-N-acetyl-N-methylaniline 69-73 cystathionase (cystathionine gamma-lyase) Mus musculus 164-167 27151180-7 2016 APAP-AD displayed a punctate pattern and were colocalized with GFP-LC3 positive autophagosomes and Lamp1 positive lysosomes in APAP-treated primary hepatocytes. 4-amino-N-acetyl-N-methylaniline 0-4 lysosomal-associated membrane protein 1 Mus musculus 99-104 26454172-2 2015 Structures of AAA were determined in its native form and in complex with the analgesic acetanilide, p-acetaminophenol, at 1.70 A and 1.73 A resolutions, respectively. 4-amino-N-acetyl-N-methylaniline 100-117 AAA1 Homo sapiens 14-17 26304691-1 2015 While N-acetyl-p-benzoquinoneimine (NAPQI), an electrophilic metabolite of acetaminophen (APAP), has been found to undergo GSH conjugation associated with its detoxification, interaction of NAPQI with nucleophilic per- and polysulfides produced by cystathionine gamma-lyase (CSE), cystathionine beta-synthase, and/or other enzymes is not known. 4-amino-N-acetyl-N-methylaniline 90-94 cystathionase (cystathionine gamma-lyase) Mus musculus 248-273 26304691-1 2015 While N-acetyl-p-benzoquinoneimine (NAPQI), an electrophilic metabolite of acetaminophen (APAP), has been found to undergo GSH conjugation associated with its detoxification, interaction of NAPQI with nucleophilic per- and polysulfides produced by cystathionine gamma-lyase (CSE), cystathionine beta-synthase, and/or other enzymes is not known. 4-amino-N-acetyl-N-methylaniline 90-94 cystathionase (cystathionine gamma-lyase) Mus musculus 275-278 27965156-12 2017 Induction of Mmp12, was dramatically reduced in APAP-treated Fibgamma390-396A mice. 4-amino-N-acetyl-N-methylaniline 48-52 matrix metallopeptidase 12 Mus musculus 13-18 27965156-13 2017 Mice lacking Mmp12 displayed exacerbated APAP-induced liver injury, resembling Fibgamma390-396A mice. 4-amino-N-acetyl-N-methylaniline 41-45 matrix metallopeptidase 12 Mus musculus 13-18 27965156-16 2017 CONCLUSIONS: These studies highlight a novel pathway of liver repair after APAP overdose, mediated by fibrin(ogen)-alphaMbeta2 integrin engagement, and demonstrate a protective role of Mmp12 expression after APAP overdose. 4-amino-N-acetyl-N-methylaniline 208-212 matrix metallopeptidase 12 Mus musculus 185-190 27866976-2 2017 The aim of the present study was to investigate the effects of rosiglitazone (RSG), a synthetic PPAR-gamma agonist, on acetaminophen (APAP)-induced acute liver injury. 4-amino-N-acetyl-N-methylaniline 134-138 peroxisome proliferator activated receptor gamma Mus musculus 96-106 27866976-8 2017 In addition, RSG pretreatment suppressed activation of hepatic nuclear factor kappa B (NF-kappaB) and extracellular signal-related kinase (ERK)/mitogen-activated protein kinase (MAPK) signaling during APAP-induced acute liver injury. 4-amino-N-acetyl-N-methylaniline 201-205 nuclear factor of kappa light polypeptide gene enhancer in B cells 1, p105 Mus musculus 87-96 27866976-8 2017 In addition, RSG pretreatment suppressed activation of hepatic nuclear factor kappa B (NF-kappaB) and extracellular signal-related kinase (ERK)/mitogen-activated protein kinase (MAPK) signaling during APAP-induced acute liver injury. 4-amino-N-acetyl-N-methylaniline 201-205 mitogen-activated protein kinase 1 Mus musculus 102-137 27866976-8 2017 In addition, RSG pretreatment suppressed activation of hepatic nuclear factor kappa B (NF-kappaB) and extracellular signal-related kinase (ERK)/mitogen-activated protein kinase (MAPK) signaling during APAP-induced acute liver injury. 4-amino-N-acetyl-N-methylaniline 201-205 mitogen-activated protein kinase 1 Mus musculus 139-142 27866976-8 2017 In addition, RSG pretreatment suppressed activation of hepatic nuclear factor kappa B (NF-kappaB) and extracellular signal-related kinase (ERK)/mitogen-activated protein kinase (MAPK) signaling during APAP-induced acute liver injury. 4-amino-N-acetyl-N-methylaniline 201-205 mitogen-activated protein kinase 1 Mus musculus 178-182 27866976-10 2017 The present results suggest that synthetic PPAR-gamma agonists might be effective agents for preventing the progression of APAP-induced acute liver injury. 4-amino-N-acetyl-N-methylaniline 123-127 peroxisome proliferator activated receptor gamma Mus musculus 43-53 27830004-6 2016 These data suggested that FA could effectively protect against APAP-induced liver injury by down-regulated expression of CYP 2E1 and the suppression of TLR4-mediated inflammatory responses. 4-amino-N-acetyl-N-methylaniline 63-67 cytochrome P450, family 2, subfamily e, polypeptide 1 Mus musculus 121-128 27830004-6 2016 These data suggested that FA could effectively protect against APAP-induced liver injury by down-regulated expression of CYP 2E1 and the suppression of TLR4-mediated inflammatory responses. 4-amino-N-acetyl-N-methylaniline 63-67 toll-like receptor 4 Mus musculus 152-156 26806302-11 2016 CONCLUSION: SolB exhibits a remarkable protective effect against APAP-induced hepatotoxicity, partially via activation of the NRF2/ARE pathway and regulation of NRF2 target genes, which induce detoxification and increase antioxidant capacity. 4-amino-N-acetyl-N-methylaniline 65-69 nuclear factor, erythroid derived 2, like 2 Mus musculus 126-130 26806302-11 2016 CONCLUSION: SolB exhibits a remarkable protective effect against APAP-induced hepatotoxicity, partially via activation of the NRF2/ARE pathway and regulation of NRF2 target genes, which induce detoxification and increase antioxidant capacity. 4-amino-N-acetyl-N-methylaniline 65-69 nuclear factor, erythroid derived 2, like 2 Mus musculus 161-165 26304691-1 2015 While N-acetyl-p-benzoquinoneimine (NAPQI), an electrophilic metabolite of acetaminophen (APAP), has been found to undergo GSH conjugation associated with its detoxification, interaction of NAPQI with nucleophilic per- and polysulfides produced by cystathionine gamma-lyase (CSE), cystathionine beta-synthase, and/or other enzymes is not known. 4-amino-N-acetyl-N-methylaniline 90-94 cystathionine beta-synthase Mus musculus 281-308 26304691-3 2015 Our in vitro experiments indicated that the formation of these novel APAP metabolites is, at least in part, attributable to the interaction of CysSSnSH produced by CSE and GSH persulfide with APAP-derived NAPQI. 4-amino-N-acetyl-N-methylaniline 192-196 cystathionase (cystathionine gamma-lyase) Mus musculus 164-167 25681557-10 2015 Notably, subtoxic doses of APAP caused cell death and sustained JNK phosphorylation in Gadd45beta-deficient primary hepatocytes. 4-amino-N-acetyl-N-methylaniline 27-31 mitogen-activated protein kinase 8 Mus musculus 64-67 25681557-10 2015 Notably, subtoxic doses of APAP caused cell death and sustained JNK phosphorylation in Gadd45beta-deficient primary hepatocytes. 4-amino-N-acetyl-N-methylaniline 27-31 growth arrest and DNA-damage-inducible 45 beta Mus musculus 87-97 25681557-7 2015 RESULTS: Metformin pretreatment protected against APAP toxicity with decreased liver damage, and inhibited APAP-induced prolonged hepatic JNK phosphorylation in WT mice. 4-amino-N-acetyl-N-methylaniline 107-111 mitogen-activated protein kinase 8 Mus musculus 138-141 25681557-13 2015 CONCLUSIONS: This study is the first to demonstrate that metformin shows protective and therapeutic effects against APAP overdose-evoked hepatotoxicity via Gadd45beta-dependent JNK regulation. 4-amino-N-acetyl-N-methylaniline 116-120 growth arrest and DNA-damage-inducible 45 beta Mus musculus 156-166 25681557-13 2015 CONCLUSIONS: This study is the first to demonstrate that metformin shows protective and therapeutic effects against APAP overdose-evoked hepatotoxicity via Gadd45beta-dependent JNK regulation. 4-amino-N-acetyl-N-methylaniline 116-120 mitogen-activated protein kinase 8 Mus musculus 177-180 25681557-9 2015 The effects of metformin on APAP-induced liver injury and JNK phosphorylation were abolished in Gadd45beta KO mice. 4-amino-N-acetyl-N-methylaniline 28-32 growth arrest and DNA-damage-inducible 45 beta Mus musculus 96-106 25645193-1 2015 OBJECTIVES: This study aimed to investigate the protective effects of Danshen (Salvia miltiorrhiza) water extract (DSE) and its major phenolic acid components against CYP2E1-mediated paracetamol (APAP)-induced hepatic toxicity. 4-amino-N-acetyl-N-methylaniline 196-200 cytochrome P450, family 2, subfamily e, polypeptide 1 Rattus norvegicus 167-173 25752611-8 2015 In contrast to chronic deletion of Parkin, acute knockdown of Parkin in mouse livers using adenovirus shRNA reduced mitophagy and Mcl-1 expression but increased JNK activation after APAP administration, which exacerbated APAP-induced liver injury. 4-amino-N-acetyl-N-methylaniline 182-186 mitogen-activated protein kinase 8 Mus musculus 161-164 25752611-8 2015 In contrast to chronic deletion of Parkin, acute knockdown of Parkin in mouse livers using adenovirus shRNA reduced mitophagy and Mcl-1 expression but increased JNK activation after APAP administration, which exacerbated APAP-induced liver injury. 4-amino-N-acetyl-N-methylaniline 221-225 mitogen-activated protein kinase 8 Mus musculus 161-164 25667042-0 2015 Role of nuclear factor-erythroid 2-related factor 2 (Nrf2) in the transcriptional regulation of brain ABC transporters during acute acetaminophen (APAP) intoxication in mice. 4-amino-N-acetyl-N-methylaniline 147-151 nuclear factor, erythroid derived 2, like 2 Mus musculus 8-51 25667042-0 2015 Role of nuclear factor-erythroid 2-related factor 2 (Nrf2) in the transcriptional regulation of brain ABC transporters during acute acetaminophen (APAP) intoxication in mice. 4-amino-N-acetyl-N-methylaniline 147-151 nuclear factor, erythroid derived 2, like 2 Mus musculus 53-57 25667042-9 2015 Furthermore, APAP treated Nrf2 knockout mice did not increase mRNA or protein expression of Mrp2 and Mrp4 as observed in wildtypes. 4-amino-N-acetyl-N-methylaniline 13-17 nuclear factor, erythroid derived 2, like 2 Mus musculus 26-30 25667042-10 2015 In contrast, P-gp induction by APAP was observed in both genotypes. 4-amino-N-acetyl-N-methylaniline 31-35 phosphoglycolate phosphatase Mus musculus 13-17 25667042-12 2015 This study also suggests that brain changes in Mrp2 and Mrp4 expression may be due to in situ Nrf2 activation by APAP, while P-gp induction is independent of Nrf2 function. 4-amino-N-acetyl-N-methylaniline 113-117 prolactin family 2, subfamily c, member 3 Mus musculus 47-51 25667042-12 2015 This study also suggests that brain changes in Mrp2 and Mrp4 expression may be due to in situ Nrf2 activation by APAP, while P-gp induction is independent of Nrf2 function. 4-amino-N-acetyl-N-methylaniline 113-117 prolactin family 2, subfamily c, member 5 Mus musculus 56-60 25667042-12 2015 This study also suggests that brain changes in Mrp2 and Mrp4 expression may be due to in situ Nrf2 activation by APAP, while P-gp induction is independent of Nrf2 function. 4-amino-N-acetyl-N-methylaniline 113-117 nuclear factor, erythroid derived 2, like 2 Mus musculus 94-98 25666961-5 2015 Administration of APAP, significantly increased, lactate dehydrogenase (LDH) and catalase (CAT) activity in liver tissue and pretreatment with TSBE returned these parameters to control group, moreover TSBE reduces APAP-induced hepatic Glutathione (GSH) depletion. 4-amino-N-acetyl-N-methylaniline 18-22 catalase Rattus norvegicus 81-89 25666961-5 2015 Administration of APAP, significantly increased, lactate dehydrogenase (LDH) and catalase (CAT) activity in liver tissue and pretreatment with TSBE returned these parameters to control group, moreover TSBE reduces APAP-induced hepatic Glutathione (GSH) depletion. 4-amino-N-acetyl-N-methylaniline 18-22 catalase Rattus norvegicus 91-94 24779639-5 2014 The contents of 4-acetamidophenol, 6beta-hydroxyltestosterone and 4-hydroxydiclofenac, the metabolites of phenacetin, testosterone and diclofenac, which were selected as specific probe drugs of CYP1A2, CYP2C9 and CYP3A4, respectively, were analyzed by high-performance liquid chromatography. 4-amino-N-acetyl-N-methylaniline 16-33 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 194-200 24882054-4 2014 METHODS: Adiponectin knockout (ADN KO) and C57 wild type mice were treated with an overdose of APAP, followed by histological and biochemical evaluation of liver injury and activation of autophagy. 4-amino-N-acetyl-N-methylaniline 95-99 adiponectin, C1Q and collagen domain containing Mus musculus 9-20 24882054-5 2014 The mechanism of adiponectin in APAP-induced hepatocytic toxicity was also explored in primary cultured hepatocytes. 4-amino-N-acetyl-N-methylaniline 32-36 adiponectin, C1Q and collagen domain containing Mus musculus 17-28 24882054-6 2014 RESULTS: APAP overdose triggers a marked accumulation of adiponectin in injured liver tissues. 4-amino-N-acetyl-N-methylaniline 9-13 adiponectin, C1Q and collagen domain containing Mus musculus 57-68 24882054-7 2014 ADN KO mice exhibit severely exacerbated mitochondrial dysfunction and damage, oxidative stress and necrosis and much higher mortality in response to APAP overdose, whereas these changes are reversed by a single injection of adiponectin. 4-amino-N-acetyl-N-methylaniline 150-154 adiponectin, C1Q and collagen domain containing Mus musculus 225-236 24882054-10 2014 CONCLUSIONS: Our findings suggest that the APAP-induced accumulation of adiponectin in liver tissues serves as an adaptive mechanism to ameliorate hepatotoxicity by promoting autophagy-mediated clearance of damaged mitochondria. 4-amino-N-acetyl-N-methylaniline 43-47 adiponectin, C1Q and collagen domain containing Mus musculus 72-83 25139304-9 2014 CONCLUSIONS: Neostigmine is an acetylcholinesterase inhibitor that ameliorates the effects of APAP-induced acute liver failure in the mouse and therefore may provide new treatment options for affected patients. 4-amino-N-acetyl-N-methylaniline 94-98 acetylcholinesterase Mus musculus 31-51 21654829-8 2011 Consequently, TRAIL- or Bim-deficient mice were substantially protected from APAP-induced liver damage. 4-amino-N-acetyl-N-methylaniline 77-81 BCL2-like 11 (apoptosis facilitator) Mus musculus 24-27 23661004-5 2013 We have assessed PTP1B expression in APAP-induced liver failure in humans and its role in the molecular mechanisms that regulate the balance between cell death and survival in human and mouse hepatocytes, as well as in a mouse model of APAP-induced hepatotoxicity. 4-amino-N-acetyl-N-methylaniline 37-41 protein tyrosine phosphatase non-receptor type 1 Homo sapiens 17-22 23661004-10 2013 APAP-treated PTP1B(-/-) hepatocytes showed enhanced antioxidant defense through the glycogen synthase kinase 3 (GSK3)beta/Src kinase family (SKF) axis, delaying tyrosine phosphorylation of the transcription factor nuclear factor-erythroid 2-related factor (Nrf2) and its nuclear exclusion, ubiquitination and degradation. 4-amino-N-acetyl-N-methylaniline 0-4 protein tyrosine phosphatase, non-receptor type 1 Mus musculus 13-18 23661004-10 2013 APAP-treated PTP1B(-/-) hepatocytes showed enhanced antioxidant defense through the glycogen synthase kinase 3 (GSK3)beta/Src kinase family (SKF) axis, delaying tyrosine phosphorylation of the transcription factor nuclear factor-erythroid 2-related factor (Nrf2) and its nuclear exclusion, ubiquitination and degradation. 4-amino-N-acetyl-N-methylaniline 0-4 glycogen synthase kinase 3 beta Mus musculus 112-121 23661004-10 2013 APAP-treated PTP1B(-/-) hepatocytes showed enhanced antioxidant defense through the glycogen synthase kinase 3 (GSK3)beta/Src kinase family (SKF) axis, delaying tyrosine phosphorylation of the transcription factor nuclear factor-erythroid 2-related factor (Nrf2) and its nuclear exclusion, ubiquitination and degradation. 4-amino-N-acetyl-N-methylaniline 0-4 nuclear factor, erythroid derived 2, like 2 Mus musculus 214-255 23661004-10 2013 APAP-treated PTP1B(-/-) hepatocytes showed enhanced antioxidant defense through the glycogen synthase kinase 3 (GSK3)beta/Src kinase family (SKF) axis, delaying tyrosine phosphorylation of the transcription factor nuclear factor-erythroid 2-related factor (Nrf2) and its nuclear exclusion, ubiquitination and degradation. 4-amino-N-acetyl-N-methylaniline 0-4 nuclear factor, erythroid derived 2, like 2 Mus musculus 257-261 23661004-11 2013 Insulin-like growth factor-I receptor-mediated signaling decreased in APAP-treated wild-type hepatocytes, but was maintained in PTP1B(-/-) cells or in wild-type hepatocytes with reduced PTP1B levels by RNA interference. 4-amino-N-acetyl-N-methylaniline 70-74 insulin-like growth factor I receptor Mus musculus 0-37 22107450-1 2012 In a recent study, we reported that interleukin (IL)-4 had a protective role against acetaminophen (APAP)-induced liver injury (AILI), although the mechanism of protection was unclear. 4-amino-N-acetyl-N-methylaniline 100-104 interleukin 4 Mus musculus 36-54 22107450-6 2012 We also report that the prolonged reduction of GSH in APAP-treated IL-4(-/-) mice appeared to contribute toward increased liver injury by causing a sustained activation of c-Jun-N-terminal kinase (JNK) since levels of phosphorylated JNK remained significantly higher in the IL-4(-/-) mice up to 24 h after APAP treatment. 4-amino-N-acetyl-N-methylaniline 54-58 interleukin 4 Mus musculus 67-71 22107450-6 2012 We also report that the prolonged reduction of GSH in APAP-treated IL-4(-/-) mice appeared to contribute toward increased liver injury by causing a sustained activation of c-Jun-N-terminal kinase (JNK) since levels of phosphorylated JNK remained significantly higher in the IL-4(-/-) mice up to 24 h after APAP treatment. 4-amino-N-acetyl-N-methylaniline 54-58 mitogen-activated protein kinase 8 Mus musculus 172-195 22107450-6 2012 We also report that the prolonged reduction of GSH in APAP-treated IL-4(-/-) mice appeared to contribute toward increased liver injury by causing a sustained activation of c-Jun-N-terminal kinase (JNK) since levels of phosphorylated JNK remained significantly higher in the IL-4(-/-) mice up to 24 h after APAP treatment. 4-amino-N-acetyl-N-methylaniline 54-58 mitogen-activated protein kinase 8 Mus musculus 197-200 22107450-6 2012 We also report that the prolonged reduction of GSH in APAP-treated IL-4(-/-) mice appeared to contribute toward increased liver injury by causing a sustained activation of c-Jun-N-terminal kinase (JNK) since levels of phosphorylated JNK remained significantly higher in the IL-4(-/-) mice up to 24 h after APAP treatment. 4-amino-N-acetyl-N-methylaniline 54-58 mitogen-activated protein kinase 8 Mus musculus 233-236 22107450-6 2012 We also report that the prolonged reduction of GSH in APAP-treated IL-4(-/-) mice appeared to contribute toward increased liver injury by causing a sustained activation of c-Jun-N-terminal kinase (JNK) since levels of phosphorylated JNK remained significantly higher in the IL-4(-/-) mice up to 24 h after APAP treatment. 4-amino-N-acetyl-N-methylaniline 54-58 interleukin 4 Mus musculus 274-278 22107450-6 2012 We also report that the prolonged reduction of GSH in APAP-treated IL-4(-/-) mice appeared to contribute toward increased liver injury by causing a sustained activation of c-Jun-N-terminal kinase (JNK) since levels of phosphorylated JNK remained significantly higher in the IL-4(-/-) mice up to 24 h after APAP treatment. 4-amino-N-acetyl-N-methylaniline 306-310 interleukin 4 Mus musculus 67-71 22107450-6 2012 We also report that the prolonged reduction of GSH in APAP-treated IL-4(-/-) mice appeared to contribute toward increased liver injury by causing a sustained activation of c-Jun-N-terminal kinase (JNK) since levels of phosphorylated JNK remained significantly higher in the IL-4(-/-) mice up to 24 h after APAP treatment. 4-amino-N-acetyl-N-methylaniline 306-310 mitogen-activated protein kinase 8 Mus musculus 172-195 22107450-6 2012 We also report that the prolonged reduction of GSH in APAP-treated IL-4(-/-) mice appeared to contribute toward increased liver injury by causing a sustained activation of c-Jun-N-terminal kinase (JNK) since levels of phosphorylated JNK remained significantly higher in the IL-4(-/-) mice up to 24 h after APAP treatment. 4-amino-N-acetyl-N-methylaniline 306-310 mitogen-activated protein kinase 8 Mus musculus 197-200 21654829-7 2011 Furthermore, we found that APAP strongly induces the expression of Bim in a c-JNK-dependent manner. 4-amino-N-acetyl-N-methylaniline 27-31 BCL2-like 11 (apoptosis facilitator) Mus musculus 67-70 21654829-7 2011 Furthermore, we found that APAP strongly induces the expression of Bim in a c-JNK-dependent manner. 4-amino-N-acetyl-N-methylaniline 27-31 mitogen-activated protein kinase 8 Mus musculus 78-81 21654829-8 2011 Consequently, TRAIL- or Bim-deficient mice were substantially protected from APAP-induced liver damage. 4-amino-N-acetyl-N-methylaniline 77-81 tumor necrosis factor (ligand) superfamily, member 10 Mus musculus 14-19 22902588-3 2012 Since hypoxia inducible factor-one alpha (HIF-1alpha) is induced very early in APAP toxicity, a role for oxidative stress in the induction has been postulated. 4-amino-N-acetyl-N-methylaniline 79-83 hypoxia inducible factor 1, alpha subunit Mus musculus 42-52 22902588-12 2012 In summary, the MPT inhibitor TFP delayed the onset of toxicity and lowered HIF-1alpha induction in APAP treated mice. 4-amino-N-acetyl-N-methylaniline 100-104 hypoxia inducible factor 1, alpha subunit Mus musculus 76-86 21654829-9 2011 This study identifies the TRAIL-JNK-Bim axis as a novel target in the treatment of APAP-induced liver damage and substantiates its general role in hepatocyte death. 4-amino-N-acetyl-N-methylaniline 83-87 tumor necrosis factor (ligand) superfamily, member 10 Mus musculus 26-31 21654829-9 2011 This study identifies the TRAIL-JNK-Bim axis as a novel target in the treatment of APAP-induced liver damage and substantiates its general role in hepatocyte death. 4-amino-N-acetyl-N-methylaniline 83-87 mitogen-activated protein kinase 8 Mus musculus 32-35 21654829-9 2011 This study identifies the TRAIL-JNK-Bim axis as a novel target in the treatment of APAP-induced liver damage and substantiates its general role in hepatocyte death. 4-amino-N-acetyl-N-methylaniline 83-87 BCL2-like 11 (apoptosis facilitator) Mus musculus 36-39 21316383-5 2011 We previously reported the induction of HIF-1alpha in mice with APAP toxicity and have shown that VEGF is important in hepatocyte regeneration following APAP toxicity. 4-amino-N-acetyl-N-methylaniline 64-68 hypoxia inducible factor 1, alpha subunit Mus musculus 40-50 21316383-9 2011 Subsequent studies with the MPT inhibitor cyclosporine A showed that cyclosporine A (CYC; 10mg/kg) reduced HIF-1alpha induction in APAP treated mice at 1 and 4h and did not inhibit the metabolism of APAP (depletion of hepatic non-protein sulfhydryls and hepatic protein adduct levels). 4-amino-N-acetyl-N-methylaniline 131-135 hypoxia inducible factor 1, alpha subunit Mus musculus 107-117 20207720-3 2010 The amount of SAA needed for the metabolism of two doses of APAP is equivalent to 62% of the recommended dietary allowance (RDA) for SAA in humans. 4-amino-N-acetyl-N-methylaniline 60-64 serum amyloid A1 cluster Homo sapiens 14-17 21135413-2 2011 The objective of the present study was to examine the role of VEGF receptor 1 (VEGFR1) signaling in hepatic tissue repair after acetaminophen (N-acetyl-para-aminophenol) (APAP)-induced liver injury. 4-amino-N-acetyl-N-methylaniline 143-168 FMS-like tyrosine kinase 1 Mus musculus 62-77 21135413-2 2011 The objective of the present study was to examine the role of VEGF receptor 1 (VEGFR1) signaling in hepatic tissue repair after acetaminophen (N-acetyl-para-aminophenol) (APAP)-induced liver injury. 4-amino-N-acetyl-N-methylaniline 143-168 FMS-like tyrosine kinase 1 Mus musculus 79-85 21205919-15 2011 The data indicate that MnSOD inactivation by nitration is an early event in APAP-induced hepatic toxicity. 4-amino-N-acetyl-N-methylaniline 76-80 superoxide dismutase 2, mitochondrial Mus musculus 23-28 20207720-3 2010 The amount of SAA needed for the metabolism of two doses of APAP is equivalent to 62% of the recommended dietary allowance (RDA) for SAA in humans. 4-amino-N-acetyl-N-methylaniline 60-64 serum amyloid A1 cluster Homo sapiens 133-136 19969075-0 2010 Arjunolic acid, a triterpenoid saponin, prevents acetaminophen (APAP)-induced liver and hepatocyte injury via the inhibition of APAP bioactivation and JNK-mediated mitochondrial protection. 4-amino-N-acetyl-N-methylaniline 64-68 mitogen-activated protein kinase 8 Rattus norvegicus 151-154 20067817-4 2010 APAP exposure for 24h significantly increased plasma level of blood urea nitrogen (BUN), creatinine, uric acid, TNF-alpha, NO production, urinary gamma-glutamyl transpeptidase (gamma-GT) activity, total urinary protein and urinary glucose level accompanied by a decrease in Na(+)-K(+)-ATPase activity. 4-amino-N-acetyl-N-methylaniline 0-4 tumor necrosis factor Mus musculus 112-121 20067817-7 2010 Besides, APAP exposure significantly reduced mitochondrial membrane potential and induced up-regulation of CYP2E1 in renal tissues although JNK did not play any significant role in this APAP-induced renal pathophysiology. 4-amino-N-acetyl-N-methylaniline 9-13 cytochrome P450, family 2, subfamily e, polypeptide 1 Mus musculus 107-113 20024105-6 2009 In contrast, the efficacy of ApAP as an inhibitor of lipid hydroperoxide biosynthesis by soybean LOX-1 (sLOX-1) increased upon incorporation of nitrogen into the ring, suggesting a different mechanism of inhibition dependent on the acidity of the phenolic O-H which may involve chelation of the catalytic non-heme iron atom. 4-amino-N-acetyl-N-methylaniline 29-33 oxidized low density lipoprotein receptor 1 Homo sapiens 104-110 19117117-11 2009 A significant increase in MDA and decreases in GSH level and GSH-Px, CAT, and SOD activity indicated that APAP-induced renal damage was mediated through oxidative stress. 4-amino-N-acetyl-N-methylaniline 106-110 catalase Rattus norvegicus 69-72 16552707-8 2006 An HO-1 inhibitor, tin-protoporphyrin-IX, significantly increased APAP-induced mortality, implicating HO-1 as a protective molecule for APAP-induced liver injury. 4-amino-N-acetyl-N-methylaniline 66-70 heme oxygenase 1 Mus musculus 3-7 17584759-10 2007 Additionally, male Gclm wild-type mice demonstrated greater APAP-induced hepatotoxicity than female wild-type mice. 4-amino-N-acetyl-N-methylaniline 60-64 glutamate-cysteine ligase, modifier subunit Mus musculus 19-23 17584759-11 2007 Cotreatment with either NAC or GSHee mitigated the effects of APAP in Gclm wild-type and heterozygous mice, but not in Gclm null mice. 4-amino-N-acetyl-N-methylaniline 62-66 glutamate-cysteine ligase, modifier subunit Mus musculus 70-74 16552707-4 2006 APAP injection markedly augmented intrahepatic gene expression of inducible nitric oxide synthase (iNOS) and heme oxygenase (HO)-1. 4-amino-N-acetyl-N-methylaniline 0-4 nitric oxide synthase 2, inducible Mus musculus 66-97 16552707-4 2006 APAP injection markedly augmented intrahepatic gene expression of inducible nitric oxide synthase (iNOS) and heme oxygenase (HO)-1. 4-amino-N-acetyl-N-methylaniline 0-4 nitric oxide synthase 2, inducible Mus musculus 99-103 16552707-5 2006 Moreover, neutrophils expressed iNOS, which is presumed to be an aggravating molecule for APAP-induced liver injury, while HO-1 was mainly expressed by macrophages. 4-amino-N-acetyl-N-methylaniline 90-94 nitric oxide synthase 2, inducible Mus musculus 32-36 18700144-11 2008 Primary hepatocyte culture also revealed that ASK1 and JNK, but not p38, contributed to direct APAP-induced cellular damage. 4-amino-N-acetyl-N-methylaniline 95-99 mitogen-activated protein kinase kinase kinase 5 Mus musculus 46-50 18700144-11 2008 Primary hepatocyte culture also revealed that ASK1 and JNK, but not p38, contributed to direct APAP-induced cellular damage. 4-amino-N-acetyl-N-methylaniline 95-99 mitogen-activated protein kinase 8 Mus musculus 55-58 17935745-2 2008 Mice deficient in Nrf2 (Nrf2-null) are highly susceptible to acetaminophen (APAP) hepatotoxicity and exhibit lower basal and inducible expression of cytoprotective genes, including NADPH quinone oxidoreductase 1 (Nqo1) and glutamate cysteine ligase (catalytic subunit, or Gclc). 4-amino-N-acetyl-N-methylaniline 76-80 nuclear factor, erythroid derived 2, like 2 Mus musculus 18-22 17935745-2 2008 Mice deficient in Nrf2 (Nrf2-null) are highly susceptible to acetaminophen (APAP) hepatotoxicity and exhibit lower basal and inducible expression of cytoprotective genes, including NADPH quinone oxidoreductase 1 (Nqo1) and glutamate cysteine ligase (catalytic subunit, or Gclc). 4-amino-N-acetyl-N-methylaniline 76-80 nuclear factor, erythroid derived 2, like 2 Mus musculus 24-28 17935745-4 2008 It was hypothesized that induction of hepatic Mrp1-4 expression following APAP is Nrf2 dependent. 4-amino-N-acetyl-N-methylaniline 74-78 S100 calcium binding protein A9 (calgranulin B) Mus musculus 46-52 17935745-4 2008 It was hypothesized that induction of hepatic Mrp1-4 expression following APAP is Nrf2 dependent. 4-amino-N-acetyl-N-methylaniline 74-78 nuclear factor, erythroid derived 2, like 2 Mus musculus 82-86 17935745-9 2008 Similarly, APAP treatment increased hepatic Mrp3 and Mrp4 mRNA and protein in WT, but not Nrf2-null mice. 4-amino-N-acetyl-N-methylaniline 11-15 prolactin family 2, subfamily c, member 4 Mus musculus 44-48 17935745-9 2008 Similarly, APAP treatment increased hepatic Mrp3 and Mrp4 mRNA and protein in WT, but not Nrf2-null mice. 4-amino-N-acetyl-N-methylaniline 11-15 prolactin family 2, subfamily c, member 5 Mus musculus 53-57 17935745-12 2008 These results show that Nrf2 mediates induction of Mrp3 and Mrp4 after APAP but does not affect Mrp1 or Mrp2. 4-amino-N-acetyl-N-methylaniline 71-75 nuclear factor, erythroid derived 2, like 2 Mus musculus 24-28 17935745-12 2008 These results show that Nrf2 mediates induction of Mrp3 and Mrp4 after APAP but does not affect Mrp1 or Mrp2. 4-amino-N-acetyl-N-methylaniline 71-75 prolactin family 2, subfamily c, member 4 Mus musculus 51-55 17935745-12 2008 These results show that Nrf2 mediates induction of Mrp3 and Mrp4 after APAP but does not affect Mrp1 or Mrp2. 4-amino-N-acetyl-N-methylaniline 71-75 prolactin family 2, subfamily c, member 5 Mus musculus 60-64 17053928-3 2007 The purpose of this study was to assess the combination of autoadjusting CPAP (APAP) and C-Flex in terms of (1) treatment efficacy, and (2) patient preference when compared to standard CPAP. 4-amino-N-acetyl-N-methylaniline 79-83 centromere protein J Homo sapiens 73-77 16552707-6 2006 All anti-granulocyte antibody-treated neutropenic WT and most CXC chemokine receptor 2 (CXCR2)-deficient mice survived the same dose of APAP, with reduced neutrophil infiltration and iNOS expression, indicating the pathogenic roles of neutrophils in APAP-induced liver injury. 4-amino-N-acetyl-N-methylaniline 136-140 chemokine (C-X-C motif) receptor 2 Mus musculus 62-86 16552707-6 2006 All anti-granulocyte antibody-treated neutropenic WT and most CXC chemokine receptor 2 (CXCR2)-deficient mice survived the same dose of APAP, with reduced neutrophil infiltration and iNOS expression, indicating the pathogenic roles of neutrophils in APAP-induced liver injury. 4-amino-N-acetyl-N-methylaniline 136-140 chemokine (C-X-C motif) receptor 2 Mus musculus 88-93 16552707-8 2006 An HO-1 inhibitor, tin-protoporphyrin-IX, significantly increased APAP-induced mortality, implicating HO-1 as a protective molecule for APAP-induced liver injury. 4-amino-N-acetyl-N-methylaniline 66-70 heme oxygenase 1 Mus musculus 102-106 12143040-1 2002 Chronic alcohol consumption may potentiate acetaminophen (APAP) hepatotoxicity through enhanced formation of N-acetyl-p-benzoquinone imine (NAPQI) via induction of cytochrome P450 2E1 (CYP2E1). 4-amino-N-acetyl-N-methylaniline 58-62 cytochrome P450, family 2, subfamily e, polypeptide 1 Rattus norvegicus 164-183 16548897-6 2006 The more recent development of auto-adjusting CPAP (APAP) is a reflection of the understanding that the pressure required to prevent UA collapse fluctuates throughout the night and results in a lower mean pressure that may be more comfortable for some patients. 4-amino-N-acetyl-N-methylaniline 52-56 centromere protein J Homo sapiens 46-50 15205452-8 2004 CYP2E1 activity and hepatic glutathione (GSH) levels in untreated mice showed significant 24-h rhythms associated with APAP toxicity rhythm. 4-amino-N-acetyl-N-methylaniline 119-123 cytochrome P450, family 2, subfamily e, polypeptide 1 Mus musculus 0-6 16420515-2 2006 In the present study, the possible influence of PAF-R antagonist (BN52021) on the protection of liver injury after 4-hydroxyacetanilide, N-acetyl-p-aminophenol, paracetamol (APAP) intoxication was investigated. 4-amino-N-acetyl-N-methylaniline 174-178 platelet-activating factor receptor Rattus norvegicus 48-53 16420515-5 2006 RESULTS: APAP was found to cause an acute hepatic injury, evident by alterations of biochemical (serum enzymes: aspartate aminotransferase (AST), alanine aminotransferase (ALT) and alkaline phosphatase) and liver histopathological (degree of necrosis and apoptosis) indices, which was followed by liver regeneration, evident by three independent indices ([3H] thymidine incorporation into hepatic DNA, liver thymidine kinase activity and hepatocyte mitotic index). 4-amino-N-acetyl-N-methylaniline 9-13 glutamic-oxaloacetic transaminase 2 Rattus norvegicus 112-138 16420515-5 2006 RESULTS: APAP was found to cause an acute hepatic injury, evident by alterations of biochemical (serum enzymes: aspartate aminotransferase (AST), alanine aminotransferase (ALT) and alkaline phosphatase) and liver histopathological (degree of necrosis and apoptosis) indices, which was followed by liver regeneration, evident by three independent indices ([3H] thymidine incorporation into hepatic DNA, liver thymidine kinase activity and hepatocyte mitotic index). 4-amino-N-acetyl-N-methylaniline 9-13 glutamic-oxaloacetic transaminase 2 Rattus norvegicus 140-143 12143040-1 2002 Chronic alcohol consumption may potentiate acetaminophen (APAP) hepatotoxicity through enhanced formation of N-acetyl-p-benzoquinone imine (NAPQI) via induction of cytochrome P450 2E1 (CYP2E1). 4-amino-N-acetyl-N-methylaniline 58-62 cytochrome P450, family 2, subfamily e, polypeptide 1 Rattus norvegicus 185-191 12143040-8 2002 Partial inhibition of NAPQI formation by CYP2E1 inhibitor diethyldithiocarbamate to that of pair-fed controls abolished APAP toxicity in the 10-day ethanol group only. 4-amino-N-acetyl-N-methylaniline 120-124 cytochrome P450, family 2, subfamily e, polypeptide 1 Rattus norvegicus 41-47 11995823-8 2001 Non growing suspended cells of strain ST1 degraded 68, 96 and 76.8% of 4-aminophenol (1,000 ppm), phenol (500 ppm) and 4-acetamidophenol (1,000 ppm), respectively, in 72 hrs. 4-amino-N-acetyl-N-methylaniline 119-136 syndecan binding protein Homo sapiens 38-41 34289451-3 2021 RESULTS: In this present study, by employing an acute liver injury induced by APAP overdose mouse model, we demonstrated that DJ-1 knockout (DJ-1-/-) mice showed reduced liver injury and lower mortality. 4-amino-N-acetyl-N-methylaniline 78-82 Parkinson disease (autosomal recessive, early onset) 7 Mus musculus 126-130 10535745-1 1999 The role of NAD(P)H:quinone reductase (QR; EC 1.6.99.2) in the alcohol-derived protective effect against hepatotoxicity caused by acetaminophen (APAP) was studied. 4-amino-N-acetyl-N-methylaniline 145-149 crystallin, zeta Mus musculus 39-41 10535745-2 1999 In mice pretreated with dicoumarol (30 mg/kg), an inhibitor of QR, hepatic necrosis caused by APAP (400 mg/kg) was potentiated. 4-amino-N-acetyl-N-methylaniline 94-98 crystallin, zeta Mus musculus 63-65 10535745-9 1999 These results suggest that ethanol inhibited not only the microsomal (CYP2E1 mediated) formation of a toxic quinone metabolite from APAP, but also accelerated the conversion of the toxic quinone metabolite produced back to APAP by stimulating cytoplasmic QR activity. 4-amino-N-acetyl-N-methylaniline 132-136 cytochrome P450, family 2, subfamily e, polypeptide 1 Mus musculus 70-76 10535745-9 1999 These results suggest that ethanol inhibited not only the microsomal (CYP2E1 mediated) formation of a toxic quinone metabolite from APAP, but also accelerated the conversion of the toxic quinone metabolite produced back to APAP by stimulating cytoplasmic QR activity. 4-amino-N-acetyl-N-methylaniline 223-227 crystallin, zeta Mus musculus 255-257 9704905-3 1998 at 08:00, 14:00 or 20:00 h. APAP at this dose was markedly hepatotoxic to mice when administered at 20:00 h as determined by increases in serum alanine aminotransferase (ALT) and aspartate aminotransferase (AST) activities, and by decreases in hepatic glucose-6-phosphatase (G-6-Pase) activity. 4-amino-N-acetyl-N-methylaniline 28-32 glutamic pyruvic transaminase, soluble Mus musculus 144-168 9704905-3 1998 at 08:00, 14:00 or 20:00 h. APAP at this dose was markedly hepatotoxic to mice when administered at 20:00 h as determined by increases in serum alanine aminotransferase (ALT) and aspartate aminotransferase (AST) activities, and by decreases in hepatic glucose-6-phosphatase (G-6-Pase) activity. 4-amino-N-acetyl-N-methylaniline 28-32 glutamic pyruvic transaminase, soluble Mus musculus 170-173 9704905-3 1998 at 08:00, 14:00 or 20:00 h. APAP at this dose was markedly hepatotoxic to mice when administered at 20:00 h as determined by increases in serum alanine aminotransferase (ALT) and aspartate aminotransferase (AST) activities, and by decreases in hepatic glucose-6-phosphatase (G-6-Pase) activity. 4-amino-N-acetyl-N-methylaniline 28-32 solute carrier family 17 (anion/sugar transporter), member 5 Mus musculus 179-205 9704905-3 1998 at 08:00, 14:00 or 20:00 h. APAP at this dose was markedly hepatotoxic to mice when administered at 20:00 h as determined by increases in serum alanine aminotransferase (ALT) and aspartate aminotransferase (AST) activities, and by decreases in hepatic glucose-6-phosphatase (G-6-Pase) activity. 4-amino-N-acetyl-N-methylaniline 28-32 solute carrier family 17 (anion/sugar transporter), member 5 Mus musculus 207-210 9704905-3 1998 at 08:00, 14:00 or 20:00 h. APAP at this dose was markedly hepatotoxic to mice when administered at 20:00 h as determined by increases in serum alanine aminotransferase (ALT) and aspartate aminotransferase (AST) activities, and by decreases in hepatic glucose-6-phosphatase (G-6-Pase) activity. 4-amino-N-acetyl-N-methylaniline 28-32 glucose-6-phosphatase, catalytic Mus musculus 252-273 9704905-3 1998 at 08:00, 14:00 or 20:00 h. APAP at this dose was markedly hepatotoxic to mice when administered at 20:00 h as determined by increases in serum alanine aminotransferase (ALT) and aspartate aminotransferase (AST) activities, and by decreases in hepatic glucose-6-phosphatase (G-6-Pase) activity. 4-amino-N-acetyl-N-methylaniline 28-32 glucose-6-phosphatase, catalytic Mus musculus 275-283 34558831-8 2021 Here, we identify hepatic NIK as a previously unrecognized mediator for acetaminophen (APAP)-induced acute liver failure. 4-amino-N-acetyl-N-methylaniline 87-91 mitogen-activated protein kinase kinase kinase 14 Mus musculus 26-29 34558831-9 2021 APAP treatment increased both NIK transcription and NIK protein stability in primary hepatocytes as well as in liver in mice. 4-amino-N-acetyl-N-methylaniline 0-4 mitogen-activated protein kinase kinase kinase 14 Mus musculus 30-33 34558831-9 2021 APAP treatment increased both NIK transcription and NIK protein stability in primary hepatocytes as well as in liver in mice. 4-amino-N-acetyl-N-methylaniline 0-4 mitogen-activated protein kinase kinase kinase 14 Mus musculus 52-55 34558831-10 2021 Hepatocyte-specific overexpression of NIK augmented APAP-induced liver oxidative stress in mice and increased hepatocyte death and mortality in a ROS-dependent manner. 4-amino-N-acetyl-N-methylaniline 52-56 mitogen-activated protein kinase kinase kinase 14 Mus musculus 38-41 34558831-12 2021 NIK increased lipid peroxidation and cell death in APAP-stimulated primary hepatocytes. 4-amino-N-acetyl-N-methylaniline 51-55 mitogen-activated protein kinase kinase kinase 14 Mus musculus 0-3 34558831-13 2021 Pretreatment with antioxidants or ferroptosis inhibitors blocked NIK/APAP-induced hepatocyte death. 4-amino-N-acetyl-N-methylaniline 69-73 mitogen-activated protein kinase kinase kinase 14 Mus musculus 65-68 34289451-3 2021 RESULTS: In this present study, by employing an acute liver injury induced by APAP overdose mouse model, we demonstrated that DJ-1 knockout (DJ-1-/-) mice showed reduced liver injury and lower mortality. 4-amino-N-acetyl-N-methylaniline 78-82 Parkinson disease (autosomal recessive, early onset) 7 Mus musculus 141-145 34289451-9 2021 CONCLUSION: our findings clearly defined that deletion of DJ-1 protects APAP-induced acute liver injury through decreasing inflammatory response, and suggest DJ-1 as a potential therapeutic and/or prophylactic target of APAP-induced acute liver injury. 4-amino-N-acetyl-N-methylaniline 72-76 Parkinson disease (autosomal recessive, early onset) 7 Mus musculus 58-62 34289451-9 2021 CONCLUSION: our findings clearly defined that deletion of DJ-1 protects APAP-induced acute liver injury through decreasing inflammatory response, and suggest DJ-1 as a potential therapeutic and/or prophylactic target of APAP-induced acute liver injury. 4-amino-N-acetyl-N-methylaniline 220-224 Parkinson disease (autosomal recessive, early onset) 7 Mus musculus 58-62 34289451-9 2021 CONCLUSION: our findings clearly defined that deletion of DJ-1 protects APAP-induced acute liver injury through decreasing inflammatory response, and suggest DJ-1 as a potential therapeutic and/or prophylactic target of APAP-induced acute liver injury. 4-amino-N-acetyl-N-methylaniline 220-224 Parkinson disease (autosomal recessive, early onset) 7 Mus musculus 158-162 32517496-8 2021 INNOVATION: This work demonstrates that both Trx and GSH systems are susceptible to APAP toxicity in vivo, and that the thiol-dependent redox environment is a key factor in determining the extent of APAP-induced hepatotoxicity. 4-amino-N-acetyl-N-methylaniline 84-88 thioredoxin 1 Mus musculus 45-48 3806412-1 1987 Pretreatment of mice with multiple doses of phenobarbital (PB) potentiates N-acetyl-para-aminophenol (APAP) hepatotoxicity through induction of cytochrome P-450, thus increasing the formation of APAP-reactive metabolites. 4-amino-N-acetyl-N-methylaniline 75-100 cytochrome P450, family 21, subfamily a, polypeptide 1 Mus musculus 144-160 3806412-1 1987 Pretreatment of mice with multiple doses of phenobarbital (PB) potentiates N-acetyl-para-aminophenol (APAP) hepatotoxicity through induction of cytochrome P-450, thus increasing the formation of APAP-reactive metabolites. 4-amino-N-acetyl-N-methylaniline 102-106 cytochrome P450, family 21, subfamily a, polypeptide 1 Mus musculus 144-160 3806412-1 1987 Pretreatment of mice with multiple doses of phenobarbital (PB) potentiates N-acetyl-para-aminophenol (APAP) hepatotoxicity through induction of cytochrome P-450, thus increasing the formation of APAP-reactive metabolites. 4-amino-N-acetyl-N-methylaniline 195-199 cytochrome P450, family 21, subfamily a, polypeptide 1 Mus musculus 144-160 3965129-8 1985 With livers of animals fed 4-acetamidophenol, a hepatotoxin chemically related to AAF, small decreases were noted in NADH:ferricyanide reductase, but not in xanthine dehydrogenase nor in DT-diaphorase. 4-amino-N-acetyl-N-methylaniline 27-44 xanthine dehydrogenase Rattus norvegicus 157-179 3965129-8 1985 With livers of animals fed 4-acetamidophenol, a hepatotoxin chemically related to AAF, small decreases were noted in NADH:ferricyanide reductase, but not in xanthine dehydrogenase nor in DT-diaphorase. 4-amino-N-acetyl-N-methylaniline 27-44 NAD(P)H quinone dehydrogenase 1 Rattus norvegicus 187-200 699902-3 1978 The conversion of [4-(3)H]acetanilide to 4-hydroxyacetanilide by rat liver microsomes (or purified cytochrome P-450) in the presence of either cumene hydroperoxide or NADPH is attended by comparable "NIH shifts". 4-amino-N-acetyl-N-methylaniline 41-61 cytochrome P450, family 2, subfamily g, polypeptide 1 Rattus norvegicus 99-115 34065411-7 2021 Moreover, HepG2 cells under spinning conditions exhibited intensive TGFbeta-induced epithelial-to-mesenchymal transition (EMT) and increased susceptibility to acetaminophen (APAP)-induced hepatotoxicity as well as hepatotoxicity prevention by administration of N-acetylcysteine (NAC). 4-amino-N-acetyl-N-methylaniline 174-178 transforming growth factor alpha Homo sapiens 68-75 34141985-4 2021 We found that both high-fructose diet feeding before APAP injection and fructose gavage after APAP injection reduced APAP-induced liver injury with a concomitant induction of the hepatic carbohydrate-response element-binding protein alpha (ChREBPalpha)-fibroblast growth factor 21 (FGF21) pathway. 4-amino-N-acetyl-N-methylaniline 53-57 fibroblast growth factor 21 Mus musculus 253-280 34141985-4 2021 We found that both high-fructose diet feeding before APAP injection and fructose gavage after APAP injection reduced APAP-induced liver injury with a concomitant induction of the hepatic carbohydrate-response element-binding protein alpha (ChREBPalpha)-fibroblast growth factor 21 (FGF21) pathway. 4-amino-N-acetyl-N-methylaniline 53-57 fibroblast growth factor 21 Mus musculus 282-287 34141985-4 2021 We found that both high-fructose diet feeding before APAP injection and fructose gavage after APAP injection reduced APAP-induced liver injury with a concomitant induction of the hepatic carbohydrate-response element-binding protein alpha (ChREBPalpha)-fibroblast growth factor 21 (FGF21) pathway. 4-amino-N-acetyl-N-methylaniline 94-98 fibroblast growth factor 21 Mus musculus 253-280 34141985-4 2021 We found that both high-fructose diet feeding before APAP injection and fructose gavage after APAP injection reduced APAP-induced liver injury with a concomitant induction of the hepatic carbohydrate-response element-binding protein alpha (ChREBPalpha)-fibroblast growth factor 21 (FGF21) pathway. 4-amino-N-acetyl-N-methylaniline 94-98 fibroblast growth factor 21 Mus musculus 282-287 34141985-4 2021 We found that both high-fructose diet feeding before APAP injection and fructose gavage after APAP injection reduced APAP-induced liver injury with a concomitant induction of the hepatic carbohydrate-response element-binding protein alpha (ChREBPalpha)-fibroblast growth factor 21 (FGF21) pathway. 4-amino-N-acetyl-N-methylaniline 117-121 fibroblast growth factor 21 Mus musculus 253-280 34141985-7 2021 Furthermore, overexpression of FGF21 in the liver was sufficient to reverse liver toxicity in APAP-injected Chrebpalpha-LKO mice. 4-amino-N-acetyl-N-methylaniline 94-98 fibroblast growth factor 21 Mus musculus 31-36 34141985-8 2021 Conclusion: Fructose protects against APAP-induced hepatotoxicity likely through its ability to activate the hepatocyte ChREBPalpha-FGF21 axis. 4-amino-N-acetyl-N-methylaniline 38-42 fibroblast growth factor 21 Mus musculus 132-137 35259261-4 2022 STUDY DESIGN, SIZE, DURATION: This study consists of (i) an in vivo human pharmaceutical APAP exposure experiment to understand to what degree APAP reaches the sperm cells in the seminal fluid; (ii) in vitro calcium imaging and functional experiments in freshly donated human sperm cells to investigate CatSper channel-dependent activation by APAP and its metabolites; and (iii) experiments to understand the in situ capabilities of human sperm cells to form APAP metabolite AM404. 4-amino-N-acetyl-N-methylaniline 143-147 cation channel sperm associated 1 Homo sapiens 303-310 32517496-3 2021 RESULTS: APAP treatment affected mouse liver selenoprotein thioredoxin reductase (TrxR) activity and glutathione level in a dose- and time-dependent manner. 4-amino-N-acetyl-N-methylaniline 9-13 peroxiredoxin 2 Mus musculus 59-80 32517496-3 2021 RESULTS: APAP treatment affected mouse liver selenoprotein thioredoxin reductase (TrxR) activity and glutathione level in a dose- and time-dependent manner. 4-amino-N-acetyl-N-methylaniline 9-13 peroxiredoxin 2 Mus musculus 82-86 32517496-5 2021 The decreases of ratio of GSH/GSSG, TrxR activity and the increase of protein S-glutathionylation were correlated with the APAP-induced hepatotoxicity. 4-amino-N-acetyl-N-methylaniline 123-127 peroxiredoxin 2 Mus musculus 36-40 32517496-7 2021 An increase in the oxidation state of the TrxR-mediated system, including cytosolic thioredoxin1 (Trx1) and peroxiredoxin1/2, and mitochondrial Trx2 and Prx3, were found in the livers from mice reared on selenium-deficient and excess selenium-supplemented diets upon APAP-treatment. 4-amino-N-acetyl-N-methylaniline 267-271 peroxiredoxin 2 Mus musculus 42-46 32517496-8 2021 INNOVATION: This work demonstrates that both Trx and GSH systems are susceptible to APAP toxicity in vivo, and that the thiol-dependent redox environment is a key factor in determining the extent of APAP-induced hepatotoxicity. 4-amino-N-acetyl-N-methylaniline 199-203 thioredoxin 1 Mus musculus 45-48 32517496-10 2021 CONCLUSION: APAP treatment in mice interrupts the function of the Trx and GSH system which are the main enzymatic antioxidant systems, in both cytosol and mitochondria. 4-amino-N-acetyl-N-methylaniline 12-16 thioredoxin 1 Mus musculus 66-69 32682928-4 2020 Baicalin alleviated APAP-induced hepatic parenchymal cells injury and enhanced the number of mitotic and proliferating cell nuclear antigen (PCNA)-positive hepatocytes in APAP-intoxicated mice. 4-amino-N-acetyl-N-methylaniline 171-175 proliferating cell nuclear antigen Mus musculus 105-139 33795997-10 2021 Moreover, the test chalcones (3a, 3f & 3 g) antagonized the effect of 4-acetaminophenol and thus, raised the catalase (CAT) and superoxide dismutase (SOD) while decreased the malondialdehyde (MDA) in experimental animals. 4-amino-N-acetyl-N-methylaniline 70-87 catalase Rattus norvegicus 109-117 33795997-10 2021 Moreover, the test chalcones (3a, 3f & 3 g) antagonized the effect of 4-acetaminophenol and thus, raised the catalase (CAT) and superoxide dismutase (SOD) while decreased the malondialdehyde (MDA) in experimental animals. 4-amino-N-acetyl-N-methylaniline 70-87 catalase Rattus norvegicus 119-122 33382198-2 2021 During the course of a study examining interactions between the common antipyretic acetaminophen (APAP; paracetamol) and interleukin-1beta (IL-1beta)-induced inflammation in neonatal mice we observed that subcutaneous (s.c.) injection of IL-1beta often leads to significantly shorter, blunt-tipped tails. 4-amino-N-acetyl-N-methylaniline 98-102 interleukin 1 alpha Mus musculus 238-246 32846216-7 2020 3, 293 T cells overexpressing Selenov gene (OE) were treated with APAP (0 to 4 mM) for 24 h or H2O2 (0 to 0.4 mM) for 12 h.Compared with the WT, the DQ- and APAP-injected KO mice had higher (P < 0.05) serum alanine aminotransferase activities and hepatic malondialdehyde (MDA), protein carbonyl, endoplasmic reticulum (ER) stress-related proteins (BIP and CHOP), apoptosis-related proteins (FAK and caspase-9), and 3-nitrotyrosine, along with lower total anti-oxidizing-capability (T-AOC) and severer hepatic necrosis. 4-amino-N-acetyl-N-methylaniline 66-70 selenoprotein V Homo sapiens 30-37 32682928-10 2020 Furthermore, the baicalin-provided NLRP3 inflammasome activation and promotion on liver regeneration after APAP-induced liver injury in wild-type mice were diminished in Nrf2 knockout mice. 4-amino-N-acetyl-N-methylaniline 107-111 NLR family, pyrin domain containing 3 Mus musculus 35-40 32682928-4 2020 Baicalin alleviated APAP-induced hepatic parenchymal cells injury and enhanced the number of mitotic and proliferating cell nuclear antigen (PCNA)-positive hepatocytes in APAP-intoxicated mice. 4-amino-N-acetyl-N-methylaniline 171-175 proliferating cell nuclear antigen Mus musculus 141-145 32682928-6 2020 Baicalin induced the activation of NOD-like receptor pyrin domain containing 3 (NLRP3) inflammasome, leading to the increased hepatic expression of interleukin-18 (IL-18) and IL-1beta in APAP-intoxicated mice. 4-amino-N-acetyl-N-methylaniline 187-191 NLR family, pyrin domain containing 3 Mus musculus 80-85 32682928-6 2020 Baicalin induced the activation of NOD-like receptor pyrin domain containing 3 (NLRP3) inflammasome, leading to the increased hepatic expression of interleukin-18 (IL-18) and IL-1beta in APAP-intoxicated mice. 4-amino-N-acetyl-N-methylaniline 187-191 interleukin 18 Mus musculus 148-162 32682928-6 2020 Baicalin induced the activation of NOD-like receptor pyrin domain containing 3 (NLRP3) inflammasome, leading to the increased hepatic expression of interleukin-18 (IL-18) and IL-1beta in APAP-intoxicated mice. 4-amino-N-acetyl-N-methylaniline 187-191 interleukin 18 Mus musculus 164-169 32682928-6 2020 Baicalin induced the activation of NOD-like receptor pyrin domain containing 3 (NLRP3) inflammasome, leading to the increased hepatic expression of interleukin-18 (IL-18) and IL-1beta in APAP-intoxicated mice. 4-amino-N-acetyl-N-methylaniline 187-191 interleukin 1 alpha Mus musculus 175-183 32682928-8 2020 Moreover, the baicalin-provided promotion on liver regeneration in APAP-intoxicated mice was diminished after the application of NLRP3 inhibitor MCC950 and the recombinant mouse IL-18 binding protein (rmIL-18BP). 4-amino-N-acetyl-N-methylaniline 67-71 NLR family, pyrin domain containing 3 Mus musculus 129-134 32682928-9 2020 Baicalin induced the cytosolic accumulation of nuclear factor erythroid 2-related factor 2 (Nrf2), and increased the interaction between Nrf2 with Nlrp3, ASC and pro-caspase-1 in livers from APAP-intoxicated mice. 4-amino-N-acetyl-N-methylaniline 191-195 nuclear factor, erythroid derived 2, like 2 Mus musculus 137-141 32793668-17 2020 Conclusions: These data indicate that DB alleviated hepatotoxicity caused by APAP at least in part via Neu1 inhibition, Akt/mTOR pathway is involved in the detoxification effect of DB on acetaminophen-induced hepatotoxicity. 4-amino-N-acetyl-N-methylaniline 77-81 neuraminidase 1 Mus musculus 103-107 32793668-17 2020 Conclusions: These data indicate that DB alleviated hepatotoxicity caused by APAP at least in part via Neu1 inhibition, Akt/mTOR pathway is involved in the detoxification effect of DB on acetaminophen-induced hepatotoxicity. 4-amino-N-acetyl-N-methylaniline 77-81 thymoma viral proto-oncogene 1 Mus musculus 120-123 32793668-17 2020 Conclusions: These data indicate that DB alleviated hepatotoxicity caused by APAP at least in part via Neu1 inhibition, Akt/mTOR pathway is involved in the detoxification effect of DB on acetaminophen-induced hepatotoxicity. 4-amino-N-acetyl-N-methylaniline 77-81 mechanistic target of rapamycin kinase Mus musculus 124-128 32484845-4 2020 We provide the means to challenge the ability of closely related model mechanisms to generate patterns of simulated hepatic injury and ALT release that scale (or not) to be quantitatively similar to the wet-lab validation targets, which are elevated plasma ALT values following acetaminophen (APAP) exposure in mice. 4-amino-N-acetyl-N-methylaniline 293-297 glutamic pyruvic transaminase, soluble Mus musculus 135-138 32213564-4 2020 In this report, using genetic and pharmacological approaches, we demonstrate that whereas Gpbar1 gene deletion worsens the severity of liver injury, its pharmacological activation by 6beta-ethyl-3a,7b-dihydroxy-5b-cholan-24-ol rescues mice from liver injury caused by APAP. 4-amino-N-acetyl-N-methylaniline 268-272 G protein-coupled bile acid receptor 1 Mus musculus 90-96 32029527-7 2020 Specifically, HNF1alpha-AS1 knockdown decreased APAP toxicity with increased cell viability and decreased LDH release, whereas HNF4alpha-AS1 knockdown exacerbated APAP toxicity, with opposite effects in the MTT and LDH assays. 4-amino-N-acetyl-N-methylaniline 48-52 HNF1 homeobox A Homo sapiens 14-23 32029527-7 2020 Specifically, HNF1alpha-AS1 knockdown decreased APAP toxicity with increased cell viability and decreased LDH release, whereas HNF4alpha-AS1 knockdown exacerbated APAP toxicity, with opposite effects in the MTT and LDH assays. 4-amino-N-acetyl-N-methylaniline 48-52 prostaglandin D2 receptor Homo sapiens 24-27 32029527-7 2020 Specifically, HNF1alpha-AS1 knockdown decreased APAP toxicity with increased cell viability and decreased LDH release, whereas HNF4alpha-AS1 knockdown exacerbated APAP toxicity, with opposite effects in the MTT and LDH assays. 4-amino-N-acetyl-N-methylaniline 163-167 hepatocyte nuclear factor 4 alpha Homo sapiens 127-136 32029527-7 2020 Specifically, HNF1alpha-AS1 knockdown decreased APAP toxicity with increased cell viability and decreased LDH release, whereas HNF4alpha-AS1 knockdown exacerbated APAP toxicity, with opposite effects in the MTT and LDH assays. 4-amino-N-acetyl-N-methylaniline 163-167 prostaglandin D2 receptor Homo sapiens 137-140 30669860-7 2019 SAG-Sh-SeNPs showed enhanced protection against APAP toxicity in comparison to Sh-SeNPs due to synergistic effect of SAG and Sh-SeNPs. 4-amino-N-acetyl-N-methylaniline 48-52 S-antigen visual arrestin Rattus norvegicus 0-3 30669860-7 2019 SAG-Sh-SeNPs showed enhanced protection against APAP toxicity in comparison to Sh-SeNPs due to synergistic effect of SAG and Sh-SeNPs. 4-amino-N-acetyl-N-methylaniline 48-52 S-antigen visual arrestin Rattus norvegicus 117-120 30669860-8 2019 SAG-Sh-SeNPs protected the liver and kidney against APAP toxicity through reducing oxidative stress, enhancing endogenous antioxidants and protecting mitochondrial functions. 4-amino-N-acetyl-N-methylaniline 52-56 S-antigen visual arrestin Rattus norvegicus 0-3