File Download

There are no files associated with this item.

  Links for fulltext
     (May Require Subscription)
Supplementary

Article: Conjugated secondary 12α-hydroxylated bile acids promote liver fibrogenesis

TitleConjugated secondary 12α-hydroxylated bile acids promote liver fibrogenesis
Authors
Keywords12α-hydroxylated bile acids
ERK1/2
G protein-coupled bile acid receptor
Hepatic stellate cell
Liver fibrosis
p38MAPK
TGR5
Issue Date2021
Citation
EBioMedicine, 2021, v. 66, article no. 103290 How to Cite?
AbstractBackground: Significantly elevated serum and hepatic bile acid (BA) concentrations have been known to occur in patients with liver fibrosis. However, the roles of different BA species in liver fibrogenesis are not fully understood. Methods: We quantitatively measured blood BA concentrations in nonalcoholic steatohepatitis (NASH) patients with liver fibrosis and healthy controls. We characterized BA composition in three mouse models induced by carbon tetrachloride (CCl4), streptozotocin-high fat diet (STZ-HFD), and long term HFD, respectively. The molecular mechanisms underlying the fibrosis-promoting effects of BAs were investigated in cell line models, a 3D co-culture system, and a Tgr5 (HSC-specific) KO mouse model. Findings: We found that a group of conjugated 12α-hydroxylated (12α-OH) BAs, such as taurodeoxycholate (TDCA) and glycodeoxycholate (GDCA), significantly increased in NASH patients and liver fibrosis mouse models. 12α-OH BAs significantly increased HSC proliferation and protein expression of fibrosis-related markers. Administration of TDCA and GDCA directly activated HSCs and promoted liver fibrogenesis in mouse models. Blockade of BA binding to TGR5 or inhibition of ERK1/2 and p38 MAPK signaling both significantly attenuated the BA-induced fibrogenesis. Liver fibrosis was attenuated in mice with Tgr5 depletion. Interpretation: Increased hepatic concentrations of conjugated 12α-OH BAs significantly contributed to liver fibrosis via TGR5 mediated p38MAPK and ERK1/2 signaling. Strategies to antagonize TGR5 or inhibit ERK1/2 and p38 MAPK signaling may effectively prevent or reverse liver fibrosis. Fundings: This study was supported by the National Institutes of Health/National Cancer Institute Grant 1U01CA188387-01A1, the National Key Research and Development Program of China (2017YFC0906800); the State Key Program of National Natural Science Foundation (81430062); the National Natural Science Foundation of China (81974073, 81774196), China Postdoctoral Science Foundation funded project, China (2016T90381), and E-institutes of Shanghai Municipal Education Commission, China (E03008).
Persistent Identifierhttp://hdl.handle.net/10722/342619
ISI Accession Number ID

 

DC FieldValueLanguage
dc.contributor.authorXie, Guoxiang-
dc.contributor.authorJiang, Runqiu-
dc.contributor.authorWang, Xiaoning-
dc.contributor.authorLiu, Ping-
dc.contributor.authorZhao, Aihua-
dc.contributor.authorWu, Yiran-
dc.contributor.authorHuang, Fengjie-
dc.contributor.authorLiu, Zhipeng-
dc.contributor.authorRajani, Cynthia-
dc.contributor.authorZheng, Xiaojiao-
dc.contributor.authorQiu, Jiannan-
dc.contributor.authorZhang, Xiaoling-
dc.contributor.authorZhao, Suwen-
dc.contributor.authorBian, Hua-
dc.contributor.authorGao, Xin-
dc.contributor.authorSun, Beicheng-
dc.contributor.authorJia, Wei-
dc.date.accessioned2024-04-17T07:05:05Z-
dc.date.available2024-04-17T07:05:05Z-
dc.date.issued2021-
dc.identifier.citationEBioMedicine, 2021, v. 66, article no. 103290-
dc.identifier.urihttp://hdl.handle.net/10722/342619-
dc.description.abstractBackground: Significantly elevated serum and hepatic bile acid (BA) concentrations have been known to occur in patients with liver fibrosis. However, the roles of different BA species in liver fibrogenesis are not fully understood. Methods: We quantitatively measured blood BA concentrations in nonalcoholic steatohepatitis (NASH) patients with liver fibrosis and healthy controls. We characterized BA composition in three mouse models induced by carbon tetrachloride (CCl4), streptozotocin-high fat diet (STZ-HFD), and long term HFD, respectively. The molecular mechanisms underlying the fibrosis-promoting effects of BAs were investigated in cell line models, a 3D co-culture system, and a Tgr5 (HSC-specific) KO mouse model. Findings: We found that a group of conjugated 12α-hydroxylated (12α-OH) BAs, such as taurodeoxycholate (TDCA) and glycodeoxycholate (GDCA), significantly increased in NASH patients and liver fibrosis mouse models. 12α-OH BAs significantly increased HSC proliferation and protein expression of fibrosis-related markers. Administration of TDCA and GDCA directly activated HSCs and promoted liver fibrogenesis in mouse models. Blockade of BA binding to TGR5 or inhibition of ERK1/2 and p38 MAPK signaling both significantly attenuated the BA-induced fibrogenesis. Liver fibrosis was attenuated in mice with Tgr5 depletion. Interpretation: Increased hepatic concentrations of conjugated 12α-OH BAs significantly contributed to liver fibrosis via TGR5 mediated p38MAPK and ERK1/2 signaling. Strategies to antagonize TGR5 or inhibit ERK1/2 and p38 MAPK signaling may effectively prevent or reverse liver fibrosis. Fundings: This study was supported by the National Institutes of Health/National Cancer Institute Grant 1U01CA188387-01A1, the National Key Research and Development Program of China (2017YFC0906800); the State Key Program of National Natural Science Foundation (81430062); the National Natural Science Foundation of China (81974073, 81774196), China Postdoctoral Science Foundation funded project, China (2016T90381), and E-institutes of Shanghai Municipal Education Commission, China (E03008).-
dc.languageeng-
dc.relation.ispartofEBioMedicine-
dc.subject12α-hydroxylated bile acids-
dc.subjectERK1/2-
dc.subjectG protein-coupled bile acid receptor-
dc.subjectHepatic stellate cell-
dc.subjectLiver fibrosis-
dc.subjectp38MAPK-
dc.subjectTGR5-
dc.titleConjugated secondary 12α-hydroxylated bile acids promote liver fibrogenesis-
dc.typeArticle-
dc.description.naturelink_to_subscribed_fulltext-
dc.identifier.doi10.1016/j.ebiom.2021.103290-
dc.identifier.pmid33752128-
dc.identifier.scopuseid_2-s2.0-85102883530-
dc.identifier.volume66-
dc.identifier.spagearticle no. 103290-
dc.identifier.epagearticle no. 103290-
dc.identifier.eissn2352-3964-
dc.identifier.isiWOS:000647447600008-

Export via OAI-PMH Interface in XML Formats


OR


Export to Other Non-XML Formats