1
|
Baek EB, Eun HS, Song JY, Hong EJ, Park SH, Kumbukgahadeniya P, Park SM, Kim SH, Kim SO, Kim HN, Cho YE, Won YS, Kwon HJ. Vitamin D supplementation ameliorates ductular reaction, liver inflammation and fibrosis in mice by upregulating TXNIP in ductular cells. Nat Commun 2025; 16:4420. [PMID: 40360509 PMCID: PMC12075793 DOI: 10.1038/s41467-025-59724-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Accepted: 05/02/2025] [Indexed: 05/15/2025] Open
Abstract
Ductular reaction is associated with liver disease progression, but there are no drugs targeting ductular reaction. Vitamin D deficiency is common in chronic liver diseases and related to disease progression, but the underlying mechanisms by which vitamin D regulates liver diseases progression remain unclear. Here, we show that vitamin D plasma levels are negatively correlated with the degree of ductular reaction in patients with chronic liver diseases. 1,25(OH)2D3, the active form of vitamin D, reduces 3,5-diethoxycarbonyl-1,4-dihydrocollidin (DDC)-induced ductular reaction, liver inflammation, and fibrosis in female mice and upregulates the vitamin D target gene, TXNIP (encoding thioredoxin-interacting protein), in ductular cells. Cholangiocyte-specific Txnip-knockout female mice are more susceptible to DDC-induced ductular reaction, inflammation, and fibrosis. Deletion of Txnip in cholangiocytes promotes proliferation and suppressed death. Furthermore, Txnip deficiency increases TNF-α and TGF-β secretion by cholangiocytes to stimulate Kupffer cells and hepatic stellate cells, consequently leading to inflammation and collagen deposition. Biliary Txnip deficiency abolishes the protective effects of vitamin D, and TXNIP overexpression attenuates DDC-induced ductular reaction and inflammation and fibrosis. Collectively, our findings identify new mechanism how vitamin D ameliorates liver diseases and suggest that the vitamin D/TXNIP axis is a therapeutic target for addressing ductular reaction and liver diseases.
Collapse
Affiliation(s)
- Eun Bok Baek
- College of Veterinary Medicine, Chungnam National University, Daejeon, Republic of Korea
- Department of Physiology, College of Medicine, Jeju National University, Jeju, Republic of Korea
| | - Hyuk Soo Eun
- Department of Internal Medicine, School of Medicine, Chungnam National University, Daejeon, Republic of Korea
- Department of Internal Medicine, Chungnam National University Hospital, Daejeon, Republic of Korea
| | - Jun-Yeop Song
- College of Veterinary Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Eun-Ju Hong
- College of Veterinary Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Se-Hee Park
- Laboratory Animal Resource Center, Korea Research Institute of Bioscience and Biotechnology, Chungbuk, Republic of Korea
| | | | - Sang-Min Park
- College of Pharmacy, Chungnam National University, Daejeon, Republic of Korea
| | - Seok-Hwan Kim
- Department of Surgery, College of Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Soon Ok Kim
- Department of Medical Sciences, Chungnam National University, Daejeon, Republic of Korea
| | - Ha Neul Kim
- Department of Medical Sciences, Chungnam National University, Daejeon, Republic of Korea
| | - Young-Eun Cho
- Department of Food and Nutrition, Andong National University, Andong, Republic of Korea
| | - Young-Suk Won
- Laboratory Animal Resource Center, Korea Research Institute of Bioscience and Biotechnology, Chungbuk, Republic of Korea.
| | - Hyo-Jung Kwon
- College of Veterinary Medicine, Chungnam National University, Daejeon, Republic of Korea.
| |
Collapse
|
2
|
Cheon I, Kim M, Kim KH, Ko S. Hepatic Nuclear Receptors in Cholestasis-to-Cholangiocarcinoma Pathology. THE AMERICAN JOURNAL OF PATHOLOGY 2025; 195:409-421. [PMID: 39326734 PMCID: PMC11983697 DOI: 10.1016/j.ajpath.2024.07.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 07/22/2024] [Accepted: 07/26/2024] [Indexed: 09/28/2024]
Abstract
Cholestasis, characterized by impaired bile flow, is associated with an increased risk of cholangiocarcinoma (CCA), a malignancy originating from the biliary epithelium and hepatocytes. Hepatic nuclear receptors (NRs) are pivotal in regulating bile acid and metabolic homeostasis, and their dysregulation is implicated in cholestatic liver diseases and the progression of liver cancer. This review elucidates the role of various hepatic NRs in the pathogenesis of cholestasis-to-CCA progression. It explores their impact on bile acid metabolism as well as their interactions with other signaling pathways implicated in CCA development. Additionally, it introduces available murine models of cholestasis/primary sclerosing cholangitis leading to CCA and discusses the clinical potential of targeting hepatic NRs as a promising approach for the prevention and treatment of cholestatic liver diseases and CCA. Understanding the complex interplay between hepatic NRs and cholestasis-to-CCA pathology holds promise for the development of novel preventive and therapeutic strategies for this devastating disease.
Collapse
Affiliation(s)
- Inyoung Cheon
- Department of Anesthesiology, Critical Care, and Pain Medicine and Center for Perioperative Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, Texas; Department of Molecular Medicine and Inflammation-Cancer Microenvironment Research Center, College of Medicine, Ewha Womans University, Seoul, Republic of Korea
| | - Minwook Kim
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania; Pittsburgh Liver Research Center, University of Pittsburgh Medical Center and University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Kang Ho Kim
- Department of Anesthesiology, Critical Care, and Pain Medicine and Center for Perioperative Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, Texas.
| | - Sungjin Ko
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania; Pittsburgh Liver Research Center, University of Pittsburgh Medical Center and University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania.
| |
Collapse
|
3
|
Liu N, Zhao P, Cao P, Hui J, Pan Y, Cheng J. Vitamin D3/VDR alleviates double-stranded RNA virus -induced biliary epithelial cell damage by inhibiting autophagy. BMC Gastroenterol 2025; 25:44. [PMID: 39881269 PMCID: PMC11780797 DOI: 10.1186/s12876-025-03640-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Accepted: 01/21/2025] [Indexed: 01/31/2025] Open
Abstract
BACKGROUND The increased apoptosis of bile duct epithelial cells (BECs) due to some damage factors is considered the initiating factor in the occurrence and progression of biliary atresia (BA). Vitamin D receptor (VDR) is thought to play a crucial role in maintaining the intrinsic immune balance and integrity of bile duct epithelial cells (BECs). To investigate the role of VDRs in the pathogenesis and progression of BA using in vitro and in vivo models. MATERIALS AND METHODS The VDR expression levels in intrahepatic bile duct epithelial cells (IBDECs) in pediatric patients with BA were examined using immunohistochemical analysis. The correlation of the VDR levels with the incidence of refractory cholangitis after Kasai portoenterostomy (KPE) and the autologous liver survival time was analyzed. The levels of genes and proteins involved in related pathways were examined using quantitative real-time polymerase chain reaction and western blotting, respectively. The secretory levels of inflammatory factors were analyzed using enzyme-linked immunosorbent assay. A BA mouse model was established through the intraperitoneal sequential injection of rhesus rotavirus (RRV). The role of VDR in the pathogenesis and progression of BA was examined using in vitro and in vivo models. Retrospective analysis of patients with BA to examine the therapeutic efficacy of VDR activators on BA. RESULTS 15 pediatric BA patients exhibiting VDR downregulation in IBDECs showed a higher incidence of refractory cholangitis after Kasai portoenterostomy (p = 0.037) and a lower native liver survival time compare to 23 BA patients without VDR downregulation (p = 0.032). 1,25-VD3 inhibited the degree of autophagy induction in HIBECs by poly(I: C) (p < 0.05), mitigated poly(I: C)-induced BEC damage and apoptosis by inhibiting autophagy (p < 0.05). 1,25-VD3 significantly suppressed the poly(I: C)-induced downregulation of SRC (p < 0.05) and ERK1/2 phosphorylation (p < 0.05). 1,25-VD3 exert a protective effect against RRV-induced BEC damage by inhibiting autophagy in BA mouse model. The incidence of cholangitis and the native liver survival time after surgery in the calcitriol-treated group was significantly lower than that in the control group. (p = 0.033, p = 0.035, respectively). CONCLUSIONS VDR activator mitigated dsRNA-induced BEC damage and apoptosis by inhibiting autophagy in vitro and in vivo. The 1,25-VD3/VDR/Src axis alleviated poly(I: C)-induced HIBEC damage and apoptosis through the PLA2/PKC/ERK pathway.
Collapse
Affiliation(s)
- Na Liu
- Department of Ultrasound, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, 710004, China
| | - Pu Zhao
- Department of Neonatology, The Third Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, 710068, China
| | - Ping Cao
- Department of Pediatric Surgery, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, 710004, China
| | - JunPeng Hui
- Department of Pathology, Xi'an Children's Hospital, Xi'an, Shaanxi Province, 710003, China
| | - YongKang Pan
- Department of Neonatal Surgery, Xi'an Children's Hospital, Xi'an, Shaanxi Province, 710003, China
| | - Jiwen Cheng
- Department of Pediatrics, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, 710061, China.
- Department of Pediatric Surgery, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, 710004, China.
| |
Collapse
|
4
|
Zhang W, Wang Z, Sun R, Zeng Y, Chen Y, Hu Q, Chen L, Ma X, Guo Y, Zhao Y. Exploration of the Combined Mechanism of Direct and Indirect Effects of Paeoniflorin in the Treatment of Cholestasis. Inflammation 2025:10.1007/s10753-025-02245-0. [PMID: 39869299 DOI: 10.1007/s10753-025-02245-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Revised: 12/16/2024] [Accepted: 01/12/2025] [Indexed: 01/28/2025]
Abstract
Cholestasis is a multifactorial hepatobiliary disorder, characterized by obstruction of bile flow and accumulation of bile, which in turn causes damage to liver cells and other tissues. In severe cases, it can result in the development of life-threatening conditions, including cirrhosis and liver cancer. Paeoniflorin (PF) has been demonstrated to possess favourable therapeutic potential for the treatment of cholestasis. The objective of this research was to examine the molecular mechanism of PF in the treatment of ANIT-induced cholestasis and to propose novel avenues for further research on the pharmacological effects of PF. In vivo and in vitro models of cholestasis were developed. The histopathological changes in the bile ducts and liver were evaluated through the use of hematoxylin and eosin (HE) staining. The extent of apoptosis was evaluated through the use of immunofluorescence (IF), immunoblotting (WB), and electron microscopy. The JNK signalling pathway was identified as the direct mechanism of action of PF through the utilisation of HuProt™ 20 K chips and other technologies. The present study demonstrated that PF markedly alleviated liver injury in an ANIT-induced cholestasis model. Specifically, PF was observed to attenuate cholestasis-induced liver injury by reducing the abnormal elevation of liver function indices and suppressing the expression of inflammatory mediators. Furthermore, PF exhibited anti-apoptotic properties in both in vivo and in vitro experiments, thereby mitigating cholestasis-induced hepatocyte apoptosis. These protective effects are attributable to the fact that PF exerts its action through direct interaction with the JNK pathway. It has been demonstrated that PF is capable of binding directly to MAPK8 (JNK1) and MAPK9 (JNK2), thereby inhibiting JNK activation and reducing apoptosis. With regard to the protection of bile ducts, PF may indirectly inhibit hepatocyte apoptosis by maintaining the structural integrity and tight junctions of bile duct cells. PF improved cholestasis by inhibiting hepatocyte apoptosis directly by targeting the JNK signaling pathway and indirectly inhibited hepatocyte apoptosis by improving the tight junctions of bile duct cells to regulate the bile duct microenvironment.
Collapse
Affiliation(s)
- Wenwen Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Zexin Wang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Rong Sun
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yi Zeng
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yuan Chen
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Qichao Hu
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Lisheng Chen
- Department of Pharmacy, Chinese PLA General Hospital, Beijing, China
| | - Xiao Ma
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| | - Yaoguang Guo
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| | - Yanling Zhao
- Department of Pharmacy, Chinese PLA General Hospital, Beijing, China.
| |
Collapse
|
5
|
Wen T, Xie J, Ma L, Hao Z, Zhang W, Wu T, Li L. Vitamin D Receptor Activation Reduces Hepatic Inflammation via Enhancing Macrophage Autophagy in Cholestatic Mice. THE AMERICAN JOURNAL OF PATHOLOGY 2024; 194:369-383. [PMID: 38104651 DOI: 10.1016/j.ajpath.2023.11.016] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 11/09/2023] [Accepted: 11/28/2023] [Indexed: 12/19/2023]
Abstract
Macrophage autophagy dysfunction aggravates liver injury by activating inflammasomes, which can cleave pro-IL-1β to its active, secreted form. We investigated whether the vitamin D/vitamin D receptor (VDR) axis could up-regulate macrophage autophagy function to inhibit the activation of inflammasome-dependent IL-1β during cholestasis. Paricalcitol (PAL; VDR agonist) was intraperitoneally injected into bile duct-ligated mice for 5 days. Up-regulation of VDR expression by PAL reduced liver injury by reducing the oxidative stress-induced inflammatory reaction in macrophages. Moreover, PAL inhibited inflammasome-dependent IL-1β generation. Mechanistically, the knockdown of VDR increased IL-1β generation, whereas VDR overexpression exerted the opposite effect following tert-butyl hydroperoxide treatment. The inflammasome antagonist glyburide, the caspase-1-specific inhibitor YVAD, and the reactive oxygen species (ROS) scavenger N-acetyl-l-cysteine (NAC) blocked the increase in Vdr shRNA-induced IL-1β production. Interestingly, up-regulation of VDR also enhanced macrophage autophagy. Autophagy reduction impaired the up-regulation of VDR-inhibited macrophage inflammasome-generated IL-1β, whereas autophagy induction showed a synergistic effect with VDR overexpression through ROS-p38 mitogen-activated protein kinase (MAPK) pathway. This result was confirmed by p38 MAPK inhibitor, MAPK activator, and ROS inhibitor NAC. Collectively, PAL triggered macrophage autophagy by suppressing activation of the ROS-p38 MAPK pathway, which, in turn, suppressed inflammasome-generated cleaved, active forms of IL-1β, eventually leading to reduced inflammation. Thus, triggering the VDR may be a potential target for the anti-inflammatory treatment of cholestatic liver disease.
Collapse
Affiliation(s)
- Tianfu Wen
- Department of General Surgery, The Affiliated Wenling First People's Hospital, Taizhou University, Taizhou, China
| | - Jing Xie
- Department of Cell Biology, School of Medicine, Taizhou University, Taizhou, China
| | - Liman Ma
- Department of Cell Biology, School of Medicine, Taizhou University, Taizhou, China
| | - Zhiqing Hao
- Department of Pathophysiology, School of Basic Medicine, Shenyang Medical College, Shenyang, China
| | - Weiwei Zhang
- Department of Pathophysiology, School of Basic Medicine, Shenyang Medical College, Shenyang, China
| | - Tingyao Wu
- Department of Hematology, First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| | - Lihua Li
- Department of General Surgery, The Affiliated Wenling First People's Hospital, Taizhou University, Taizhou, China.
| |
Collapse
|
6
|
Zeng J, Fan J, Zhou H. Bile acid-mediated signaling in cholestatic liver diseases. Cell Biosci 2023; 13:77. [PMID: 37120573 PMCID: PMC10149012 DOI: 10.1186/s13578-023-01035-1] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Accepted: 04/18/2023] [Indexed: 05/01/2023] Open
Abstract
Chronic cholestatic liver diseases, such as primary biliary cholangitis (PBC) and primary sclerosing cholangitis (PSC), are associated with bile stasis and gradually progress to fibrosis, cirrhosis, and liver failure, which requires liver transplantation. Although ursodeoxycholic acid is effective in slowing the disease progression of PBC, it has limited efficacy in PSC patients. It is challenging to develop effective therapeutic agents due to the limited understanding of disease pathogenesis. During the last decade, numerous studies have demonstrated that disruption of bile acid (BA) metabolism and intrahepatic circulation promotes the progression of cholestatic liver diseases. BAs not only play an essential role in nutrition absorption as detergents but also play an important role in regulating hepatic metabolism and modulating immune responses as key signaling molecules. Several excellent papers have recently reviewed the role of BAs in metabolic liver diseases. This review focuses on BA-mediated signaling in cholestatic liver disease.
Collapse
Affiliation(s)
- Jing Zeng
- Department of Microbiology and Immunology, Medical College of Virginia and Richmond VA Medical Center, Central Virginia Veterans Healthcare System, Virginia Commonwealth University, 1220 East Broad Street, MMRB-5044, Richmond, VA, 23298-0678, USA
- Department of Gastroenterology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Jiangao Fan
- Department of Gastroenterology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Huiping Zhou
- Department of Microbiology and Immunology, Medical College of Virginia and Richmond VA Medical Center, Central Virginia Veterans Healthcare System, Virginia Commonwealth University, 1220 East Broad Street, MMRB-5044, Richmond, VA, 23298-0678, USA.
| |
Collapse
|
7
|
Aseem SO, Hylemon PB, Zhou H. Bile Acids and Biliary Fibrosis. Cells 2023; 12:cells12050792. [PMID: 36899928 PMCID: PMC10001305 DOI: 10.3390/cells12050792] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 02/22/2023] [Accepted: 02/24/2023] [Indexed: 03/06/2023] Open
Abstract
Biliary fibrosis is the driving pathological process in cholangiopathies such as primary biliary cholangitis (PBC) and primary sclerosing cholangitis (PSC). Cholangiopathies are also associated with cholestasis, which is the retention of biliary components, including bile acids, in the liver and blood. Cholestasis may worsen with biliary fibrosis. Furthermore, bile acid levels, composition and homeostasis are dysregulated in PBC and PSC. In fact, mounting data from animal models and human cholangiopathies suggest that bile acids play a crucial role in the pathogenesis and progression of biliary fibrosis. The identification of bile acid receptors has advanced our understanding of various signaling pathways involved in regulating cholangiocyte functions and the potential impact on biliary fibrosis. We will also briefly review recent findings linking these receptors with epigenetic regulatory mechanisms. Further detailed understanding of bile acid signaling in the pathogenesis of biliary fibrosis will uncover additional therapeutic avenues for cholangiopathies.
Collapse
Affiliation(s)
- Sayed Obaidullah Aseem
- Stravitz-Sanyal Institute for Liver Disease & Metabolic Health, School of Medicine, Virginia Commonwealth University, Richmond, VA 23298, USA
- Division of Gastroenterology, Hepatology and Nutrition, Department of Internal Medicine, Medical College of Virginia, Virginia Commonwealth University, Richmond, VA 23298, USA
- Correspondence:
| | - Phillip B. Hylemon
- Department of Microbiology and Immunology, Virginia Commonwealth University, Richmond, VA 23298, USA
- Central Virginia Veterans Healthcare System, Richmond, VA 23249, USA
| | - Huiping Zhou
- Department of Microbiology and Immunology, Virginia Commonwealth University, Richmond, VA 23298, USA
- Central Virginia Veterans Healthcare System, Richmond, VA 23249, USA
| |
Collapse
|
8
|
Zheng Z, Xie J, Ma L, Hao Z, Zhang W, Li L. Vitamin D Receptor Activation Targets ROS-Mediated Crosstalk Between Autophagy and Apoptosis in Hepatocytes in Cholestasic Mice. Cell Mol Gastroenterol Hepatol 2023; 15:887-901. [PMID: 36280140 PMCID: PMC9972562 DOI: 10.1016/j.jcmgh.2022.10.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 10/17/2022] [Accepted: 10/17/2022] [Indexed: 12/10/2022]
Abstract
BACKGROUND & AIMS Observational epidemiologic studies have associated vitamin D deficiency with cholestasis. We reported previously that activation of the vitamin D/vitamin D receptor (VDR) axis in cholangiocytes mitigates cholestatic liver injury by remodeling the damaged bile duct. However, the function of VDR in hepatocytes during cholestasis remains unclear. METHODS Paricalcitol (VDR agonist, 200 ng/kg) was injected intraperitoneally into bile duct-ligated mice every other day for 5 days. Primary hepatocytes and HepG2 hepatoma cells were transfected with Vdr short hairpin RNA, control short hairpin RNA, Vdr plasmid, control vector, Atg5 small interfering RNA (siRNA), and control siRNA. Liver histology, cell proliferation, and autophagy were evaluated. RESULTS Treatment with the VDR agonist paricalcitol improved liver injury in bile duct-ligated mice by up-regulating VDR expression in hepatocytes, which in turn reduced hepatocyte apoptosis by inhibiting reactive oxygen species (ROS) generation via suppressing the Ras-related C3 botulinum toxin substrate 1/reduced nicotinamide adenine dinucleotide phosphate oxidase 1 pathway. Mechanistically, upon exposure to an ROS-inducing compound, Vdr siRNA contributed to apoptosis, whereas the Vdr overexpression caused resistance to apoptosis. Interestingly, up-regulated VDR expression also increased the generation of autophagosomes and macroautophagic/autophagic flux, which was the underlying mechanism for reduced apoptosis following VDR activation. Autophagy depletion impaired the positive effects of VDR overexpression, whereas autophagy induction was synergystic with VDR overexpression. Importantly, up-regulation of VDR promoted autophagy activation by suppressing the activation of the extracellular signal-regulated kinase (ERK)/p38 mitogen-activated protein kinase (p38MAPK) pathway. Thus, a p38MAPK inhibitor abrogated the Vdr siRNA-induced decrease in autophagy and the Vdr siRNA-induced increase in apoptosis. In contrast, a Mitogen-activated protein kinase kinase (MEK)/ERK activator prevented the enhancement of autophagy and decreased apoptosis following Vdr overexpression. Moreover, the ROS inhibitor N-acetylcystein (NAC) blocked Vdr siRNA-enhanced activation of the ERK/p38MAPK pathway. CONCLUSIONS VDR activation mitigated liver cholestatic injury by reducing autophagy-dependent hepatocyte apoptosis and suppressing the activation of the ROS-dependent ERK/p38MAPK pathway. Thus, VDR activation may be a potential target for the treatment of cholestatic liver disease.
Collapse
Affiliation(s)
- Zhijian Zheng
- Department of General Surgery, Affiliated Wenling First People's Hospital, Taizhou University, Taizhou, Zhejiang Province, P R China
| | - Jing Xie
- Department of Cell Biology, School of Medicine, Taizhou University, Taizhou, Zhejiang Province, P R China
| | - Liman Ma
- Department of Cell Biology, School of Medicine, Taizhou University, Taizhou, Zhejiang Province, P R China
| | - Zhiqing Hao
- Department of Pathophysiology, School of Basic Medicine, Shenyang Medical College, Shenyang, Liaoning Province, PR China
| | - Weiwei Zhang
- Department of Pathophysiology, School of Basic Medicine, Shenyang Medical College, Shenyang, Liaoning Province, PR China
| | - Lihua Li
- Department of General Surgery, Affiliated Wenling First People's Hospital, Taizhou University, Taizhou, Zhejiang Province, P R China.
| |
Collapse
|
9
|
Yu D, Shao Z, Fu Y, Tang X, Chen Q, Deng Z. Metabolomics- and systems toxicology-based hepatotoxicity mechanism of Sophorae Tonkinensis Radix et Rhizoma in rats. Front Pharmacol 2022; 13:1015008. [DOI: 10.3389/fphar.2022.1015008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Accepted: 10/17/2022] [Indexed: 11/19/2022] Open
Abstract
Drug-induced liver injury (DILI) is a major challenge to the development and clinical application of drugs, especially limits the global application of Chinese herbal medicines, because the material basis and mechanisms of some Chinese herbal medicines are not well clear. In this study, a comprehensive method integrating metabolomics and systems toxicology (SysT) was used to investigate how the main substances in Sophorae TonkinensisRadix et Rhizoma (STRER) influence the metabolic pathways and molecular mechanisms of hepatotoxicity. Through a 28-day continuous oral administration toxicity study combined with serum metabolomics analyses, the aqueous, ethanol-precipitation and dichloromethane extracts of STRER exhibited significant hepatotoxic effects. In addition, 19 differential metabolites with a time-dose-effect relationship were identified in rats. The primary bile acid biosynthesis pathway was significantly altered, which was consistent with the findings of the SysT analysis. Furthermore, through the quantification of bile acids in serum, 16 differential bile acids were identified as being significantly changed; moreover, 21 relevant targets which intersected with the hepatotoxic targets of STRER were identified. Molecular docking was used to confirm the validation of bindings between targets and corresponding compounds, and finally, six important compounds and 14 potential targets were identified to be involved in STRER-induced liver injury in relation to bile acid metabolism.
Collapse
|
10
|
Markotić A, Kelava T, Markotić H, Silovski H, Mrzljak A. Vitamin D in liver cancer: novel insights and future perspectives. Croat Med J 2022; 63:187-196. [PMID: 35505652 PMCID: PMC9086812 DOI: 10.3325/cmj.2022.63.187] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Accepted: 02/09/2022] [Indexed: 11/17/2022] Open
Abstract
Vitamin D has been a focus of attention in liver cancer due to its direct and indirect antineoplastic effects. This review critically evaluates data from recently published basic and clinical studies investigating the role of vitamin D in liver cancer. Basic studies indicate that vitamin D plays an important role in liver cancer development by suppressing the activity of hepatic stellate cells and Kupffer cells. Furthermore, vitamin D has a direct anti-proliferative, anti-angiogenic, proapoptotic, and prodifferentiative effect on liver cancer cells. Recent investigation suggested several interesting mechanisms of these actions, such as inactivation of Notch signaling, p27 accumulation, and tyrosine-protein kinase Met/extracellular signal-regulated kinases inhibition. On the other hand, data from clinical observational studies, although promising, are still inconclusive. Unfortunately, studies on the effect of vitamin D supplementation were generally focused on short-term outcomes of chronic liver diseases (liver enzyme levels or elastographic finding); therefore, there are still no reliable data on the effect of vitamin D supplementation on liver cancer occurrence or survival.
Collapse
Affiliation(s)
| | - Tomislav Kelava
- Tomislav Kelava, Department of Physiology, University of Zagreb School of Medicine, Šalata 3, 10000 Zagreb, Croatia,
| | | | | | | |
Collapse
|
11
|
Triantos C, Aggeletopoulou I, Thomopoulos K, Mouzaki A. Vitamin D-Liver Disease Association: Biological Basis and Mechanisms of Action. Hepatology 2021; 74:1065-1073. [PMID: 33405236 DOI: 10.1002/hep.31699] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 11/30/2020] [Accepted: 12/19/2020] [Indexed: 12/11/2022]
Affiliation(s)
- Christos Triantos
- Division of GastroenterologyDepartment of Internal MedicineUniversity Hospital of PatrasPatrasGreece
| | - Ioanna Aggeletopoulou
- Division of GastroenterologyDepartment of Internal MedicineUniversity Hospital of PatrasPatrasGreece
- Division of HematologyDepartment of Internal MedicineMedical SchoolUniversity of PatrasPatrasGreece
| | - Konstantinos Thomopoulos
- Division of GastroenterologyDepartment of Internal MedicineUniversity Hospital of PatrasPatrasGreece
| | - Athanasia Mouzaki
- Division of HematologyDepartment of Internal MedicineMedical SchoolUniversity of PatrasPatrasGreece
| |
Collapse
|
12
|
Xie J, Fan Y, Jia R, Yang F, Ma L, Li L. Yes-associated protein regulates the hepatoprotective effect of vitamin D receptor activation through promoting adaptive bile duct remodeling in cholestatic mice. J Pathol 2021; 255:95-106. [PMID: 34156701 DOI: 10.1002/path.5750] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 05/13/2021] [Accepted: 06/19/2021] [Indexed: 12/31/2022]
Abstract
Mounting clinical evidence has revealed that the vitamin D receptor (VDR) is associated with cholestatic liver injury, although the functions of VDR in this condition remain largely unexplored. Here, we investigated the effects of VDR activation on bile duct ligation (BDL) mice, and the underlying mechanisms were further investigated. A low-calcemic VDR agonist, paricalcitol (PAL, 200 ng/kg), was intraperitoneally injected into BDL mice every other day for 5 days or 28 days. Liver histology, liver function indicators, cholangiocyte proliferation, fibrosis scores, and inflammation were evaluated. Mice treated with PAL were rescued from the decreased survival rate induced by BDL and liver damage was reduced. Mechanistically, PAL promoted cholangiocyte proliferation, which was likely conducive to proliferating bile duct maturation and increased branching of bile ducts. PAL treatment also increased the expression of Yes-associated protein (YAP) and its target protein epithelial cell adhesion molecule (EpCam) and decreased the level of inactive cytoplasmic phosphorylated YAP. YAP knockdown abrogated PAL-induced primary bile duct epithelial cell proliferation, confirmed with YAP inhibitor administration. In addition, BDL-induced liver fibrosis and inflammatory cell infiltration were reduced by PAL treatment at both day 5 and day 28 post-BDL. In conclusion, VDR activation mitigates cholestatic liver injury by promoting adaptive bile duct remodeling through cholangiocytic YAP upregulation. Because PAL is an approved clinical drug, it may be useful for treatment of cholestatic liver disease. © 2021 The Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Jing Xie
- Department of Cell Biology, School of Medicine, Taizhou University, Taizhou, PR China
| | - Yonggang Fan
- Institute of Health Sciences, Key Laboratory of Medical Cell Biology of the Ministry of Education, China Medical University, Shenyang, PR China
| | - Rongjun Jia
- Department of Cell Biology, Jinzhou Medical University, Jinzhou, PR China
| | - Fan Yang
- Department of Cell Biology, Jinzhou Medical University, Jinzhou, PR China
| | - Liman Ma
- Department of Cell Biology, School of Medicine, Taizhou University, Taizhou, PR China
| | - Lihua Li
- Department of Cell Biology, School of Medicine, Taizhou University, Taizhou, PR China
| |
Collapse
|
13
|
Jia R, Yang F, Yan P, Ma L, Yang L, Li L. Paricalcitol inhibits oxidative stress-induced cell senescence of the bile duct epithelium dependent on modulating Sirt1 pathway in cholestatic mice. Free Radic Biol Med 2021; 169:158-168. [PMID: 33872698 DOI: 10.1016/j.freeradbiomed.2021.04.019] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 03/30/2021] [Accepted: 04/13/2021] [Indexed: 12/25/2022]
Abstract
BACKGROUND Clinical studies indicate that vitamin D receptor (VDR) expression is reduced in primary biliary cirrhosis patient livers. However, the mechanism by which activated VDR effect cholestatic liver injury remains unclear. METHODS Mice were injected intraperitoneally with the VDR agonist paricalcitol or a vehicle 3 days prior to bile duct ligation (BDL) and for 5 or 28 days after surgery. The analyses of liver morphology and necrotic areas were based on H&E staining. Serum biochemical indicators of liver damage were analyzed by commercial kits. The mechanisms of paricalcitol on cholestatic liver injury were determined by Western blot analysis. RESULTS Paricalcitol ameliorated the BDL-induced liver damage in mice. Paricalcitol increased the proliferation of BECs to promote the repair of the bile duct. Paricalcitol also reduced the BDL-induced oxidative stress level in the mice. Mechanistic analysis revealed that paricalcitol decreased the number of SA-β-gal-positive cells and downregulated the expression of p53, p21 and p16 proteins which was associated with reducing oxidative stress. Additionally, paricalcitol exerted the inhibitory effect of cell senescence was through reducing DNA damage and promoting DNA repair. Interesting, we found that paricalcitol prevented the downregulation of oxidative stress-induced Sirt1 expression in the BDL mice and t-BHP-induced BECs models. Moreover, paricalcitol suppressed cell senescence through a Sirt1-dependent pathway. These results were confirmed by antioxidant ALCAR and the Sirt1 inhibitor EX-527. CONCLUSION Paricalcitol alleviated cholestatic liver injury through promoting the repair of damaged bile ducts and reducing oxidative stress-induced cell senescence of the bile duct via modulating Sirt1 pathway.
Collapse
Affiliation(s)
- Rongjun Jia
- Department of Cell Biology, School of Medicine, Taizhou University, Taizhou, PR China; Department of Cell Biology, Jinzhou Medical University, Jinzhou, PR China.
| | - Fan Yang
- Department of Cell Biology, Jinzhou Medical University, Jinzhou, PR China.
| | - Pengfei Yan
- Department of Cell Biology, Jinzhou Medical University, Jinzhou, PR China.
| | - Liman Ma
- Department of Cell Biology, School of Medicine, Taizhou University, Taizhou, PR China.
| | - Longfei Yang
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetics, The Second Hospital of Jilin University, Changchun, PR China.
| | - Lihua Li
- Department of Cell Biology, School of Medicine, Taizhou University, Taizhou, PR China.
| |
Collapse
|
14
|
Gonzalez-Sanchez E, El Mourabit H, Jager M, Clavel M, Moog S, Vaquero J, Ledent T, Cadoret A, Gautheron J, Fouassier L, Wendum D, Chignard N, Housset C. Cholangiopathy aggravation is caused by VDR ablation and alleviated by VDR-independent vitamin D signaling in ABCB4 knockout mice. Biochim Biophys Acta Mol Basis Dis 2021; 1867:166067. [PMID: 33418034 DOI: 10.1016/j.bbadis.2020.166067] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 12/19/2020] [Accepted: 12/29/2020] [Indexed: 12/12/2022]
Abstract
BACKGROUND & AIMS Cholangiopathies are chronic liver diseases in which damaged cholangiocytes trigger a proinflammatory and profibrotic reaction. The nuclear vitamin D receptor (VDR) is highly expressed in cholangiocytes and exerts immune-regulatory functions in these cells. In the present study, we examined the protective function of VDR and other vitamin D signaling pathways in chronic cholangiopathy and cholangiocytes. METHODS Vdr was invalidated in Abcb4 knockout mice, a widely used animal model of chronic cholangiopathy. The impact of vitamin D signaling on cholangiopathy features was examined in vivo and in cholangiocytes (primary and cell lines). RESULTS Cholangiopathy features (i.e, cholestasis, ductular reaction and fibrosis) were aggravated in Vdr;Abcb4 double knockout mice compared to the Abcb4 simple knockout, and associated with an overexpression of proinflammatory factors. The proinflammatory phenotype of cholangiocytes was also exacerbated following VDR silencing in vitro. The expression of proinflammatory factors and the severity of cholangiopathy were reduced in the double knockout mice treated with the vitamin D analog calcipotriol or with vitamin D. In vitro, the inflammatory response to TNFα was significantly reduced by calcipotriol in biliary cells silenced for VDR, and this effect was abolished by co-silencing the plasma membrane receptor of vitamin D, protein disulfide-isomerase A3 (PDIA3). CONCLUSIONS Our results demonstrate an anti-inflammatory role of VDR signaling in cholangiocytes and cholangiopathy. They also provide evidence for PDIA3-mediated anti-inflammatory effects of vitamin D and vitamin D analog in these settings.
Collapse
Affiliation(s)
- Ester Gonzalez-Sanchez
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine (CRSA), Paris, France; Inovarion, Paris, France; Oncology Program, CIBEREHD, National Biomedical Research Institute on Liver and Gastrointestinal Diseases, Instituto de Salud Carlos III, Madrid, Spain; TGF-β and Cancer Group, Oncobell Program, Bellvitge Biomedical Research Institute (IDIBELL), Barcelona, Spain.
| | - Haquima El Mourabit
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine (CRSA), Paris, France.
| | - Marion Jager
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine (CRSA), Paris, France
| | - Marie Clavel
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine (CRSA), Paris, France; Inovarion, Paris, France
| | - Sophie Moog
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine (CRSA), Paris, France; Inovarion, Paris, France.
| | - Javier Vaquero
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine (CRSA), Paris, France; Oncology Program, CIBEREHD, National Biomedical Research Institute on Liver and Gastrointestinal Diseases, Instituto de Salud Carlos III, Madrid, Spain; TGF-β and Cancer Group, Oncobell Program, Bellvitge Biomedical Research Institute (IDIBELL), Barcelona, Spain; LPP (Laboratoire de Physique des Plasmas, UMR 7648), Sorbonne Université, Centre National de la Recherche Scientifique (CNRS), Ecole Polytechnique, 75005 Paris, France.
| | - Tatiana Ledent
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine (CRSA), Paris, France.
| | - Axelle Cadoret
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine (CRSA), Paris, France.
| | - Jérémie Gautheron
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine (CRSA), Paris, France.
| | - Laura Fouassier
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine (CRSA), Paris, France.
| | - Dominique Wendum
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine (CRSA), Paris, France; Assistance Publique-Hôpitaux de Paris (AP-HP) Sorbonne Université, Hôpital St Antoine, Paris, France.
| | | | - Chantal Housset
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine (CRSA), Paris, France; AP-HP, Reference Center for Inflammatory Biliary Diseases and Autoimmune Hepatitis (CRMR, MIVB-H), Department of Hepatology, Saint-Antoine Hospital, Paris, France.
| |
Collapse
|
15
|
Li L, Yang F, Jia R, Yan P, Ma L. Velvet antler polypeptide prevents the disruption of hepatic tight junctions via inhibiting oxidative stress in cholestatic mice and liver cell lines. Food Funct 2020; 11:9752-9763. [PMID: 33073799 DOI: 10.1039/d0fo01899f] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
The present study aims to examine the protective effects and mechanism of a velvet antler polypeptide (VAP) against lithocholic acid (LCA)-induced cholestatic liver injury in mice. A 7.0 kDa VAP was orally administered at doses of 10 and 20 mg kg-1 day-1. Hematoxylin and eosin (H&E) staining of the liver showed that VAP7.0 reduced LCA-induced infiltration of inflammatory cells and areas of necrotic hepatocytes. In addition, VAP7.0 greatly reduced the levels of alanine aminotransferase (ALT), total bile acid (TBA) and total bilirubin (TBIL) in LCA mouse serum and prolonged the survival time of mice with LCA. VAP7.0 reduced the production of reactive oxygen species (ROS), decreased malondialdehyde (MDA) and increased the superoxide dismutase (SOD) levels in LCA mice. VAP7.0 also reduced OGG1 expression, which is a biochemical indicator of oxidative stress. Mechanistic analysis revealed that VAP7.0 significantly inhibited LCA-induced disruption of tight junction integrity, as determined by observing the morphology of the bile canaliculus, and this finding was confirmed by observation of the bile canalicular structure and tight junction proteins Occludin and ZO-1 expression. Moreover, we also found that VAP7.0 maintained the stability of hepatic paracellular permeability, as determined by Evans blue dye assays and horseradish peroxidase (HRP) tracer distribution through inhibiting the activation of the PI3K pathway in LCA mouse livers. In addition, VAP7.0 ameliorated H2O2-induced barrier dysfunction and tight junction disruption via inhibiting the PI3K activity in human HepG2 and SMMC7721 cells, which was confirmed by the PI3K activator 740Y-P. H2O2 disturbed the localization of the tight junction proteins ZO-1 and Occludin, resulting in the transfer of these proteins from the membrane to the cytoplasm of cells, whereas pretreatment of cells with VAP7.0 prevented the disruption of the localization of these proteins, as determined by immunofluorescence staining and western blot analysis. These results demonstrate that VAP7.0 reduces liver injury by inhibiting oxidative stress and maintains the stability of hepatic tight junctions via suppressing the activation of the intracellular signaling molecule PI3K in LCA mice and hepatocellular carcinoma cells.
Collapse
Affiliation(s)
- Lihua Li
- Department of Cell Biology, Taizhou University, Taizhou, PR China.
| | | | | | | | | |
Collapse
|
16
|
Wang X, Wang G, Qu J, Yuan Z, Pan R, Li K. Calcipotriol Inhibits NLRP3 Signal Through YAP1 Activation to Alleviate Cholestatic Liver Injury and Fibrosis. Front Pharmacol 2020; 11:200. [PMID: 32296329 PMCID: PMC7136474 DOI: 10.3389/fphar.2020.00200] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2019] [Accepted: 02/14/2020] [Indexed: 12/13/2022] Open
Abstract
Cholestasis is common in multiple clinical circumstances. The NOD-like receptor protein 3 (NLRP3) inflammasome pathway has been demonstrated to play an important role in liver injury and fibrosis induced by cholestasis. We previously proved that MCC950, a selective NLRP3 inhibitor, alleviates liver fibrosis and injury in experimental liver cholestasis induced by bile-duct ligation (BDL) in mice. Herein, we investigate the role of calcipotriol, a potent vitamin D receptor agonist, in experimental liver cholestasis, test its therapeutic efficacy, and explore its potential protective mechanism. C57BL/6 mice were made to undergo BDL or fed the 0.1% 3,5-diethoxycarbonyl-1,4-dihydrocollidine (DDC) diet to establish two classic cholestatic models. Calcipotriol was administered intraperitoneally to these mice daily. Serum makers of liver damage and integrity, liver histological changes, levels of liver pro-fibrotic markers, bile acid synthetases and transporters were measured in vivo. The underlying mechanism by which calcipotriol alleviates cholestatic liver injury and fibrosis was further investigated. The results of the current study demonstrated that calcipotriol supplement significantly alleviate cholestatic liver injury and fibrosis. Moreover, calcipotriol supplement markedly inhibited NLRP3 inflammasome pathway activation to alleviate liver injury and fibrosis in vivo and inhibit hepatic stellate cell (HSC) activation in vitro. In addition, VDR agonist calcipotriol exert inhibitory effect on NLRP3 inflammasome activation through activating yes-associated protein 1 (YAP1). In conclusion, our findings proved that calcipotriol suppressed the NLRP3 signal by activating YAP1 to alleviate liver injury and retard fibrogenesis in cholestasis.
Collapse
Affiliation(s)
- Xiaopeng Wang
- Department of Biliary-Pancreatic Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Guiyang Wang
- The First Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Second Military Medical University, Shanghai, China
| | - Junwen Qu
- Department of Biliary-Pancreatic Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Zhiqing Yuan
- Department of Biliary-Pancreatic Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Ruogu Pan
- Department of Biliary-Pancreatic Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Kewei Li
- Department of Biliary-Pancreatic Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
17
|
Bellan M, Andreoli L, Mele C, Sainaghi PP, Rigamonti C, Piantoni S, De Benedittis C, Aimaretti G, Pirisi M, Marzullo P. Pathophysiological Role and Therapeutic Implications of Vitamin D in Autoimmunity: Focus on Chronic Autoimmune Diseases. Nutrients 2020; 12:E789. [PMID: 32192175 PMCID: PMC7146294 DOI: 10.3390/nu12030789] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Accepted: 03/13/2020] [Indexed: 02/07/2023] Open
Abstract
Vitamin D is a pleiotropic secosteroid yielding multiple actions in human physiology. Besides the canonical regulatory activity on bone metabolism, several non-classical actions have been described and the ability of vitamin D to partake in the regulation of the immune system is particularly interesting, though far stronger and convincing evidence has been collected in in vitro as compared to in vivo studies. Whether vitamin D is able to regulate at physiological concentrations the human immune system remains unproven to date. Consequently, it is not established if vitamin D status is a factor involved in the pathogenesis of immune-mediated diseases and if cholecalciferol supplementation acts as an adjuvant for autoimmune diseases. The development of autoimmunity is a heterogeneous process, which may involve different organs and systems with a wide range of clinical implications. In the present paper, we reviewed the current evidences regarding vitamin D role in the pathogenesis and management of different autoimmune diseases.
Collapse
Affiliation(s)
- Mattia Bellan
- Department of Translational Medicine, Università del Piemonte Orientale UPO, 28100 Novara, Italy; (M.B.); (C.M.); (P.P.S.); (C.R.); (C.D.B.); (G.A.); (M.P.)
- Division of Internal Medicine, “AOU Maggiore della Carità”, 28100 Novara, Italy
- CAAD, Centre for Autoimmune and Allergic Diseases, 28100 Novara, Italy
| | - Laura Andreoli
- Rheumatology and Clinical Immunology Unit and Department of Clinical and Experimental Sciences, Spedali Civili and University of Brescia, 25128 Brescia, Italy; (L.A.); (S.P.)
| | - Chiara Mele
- Department of Translational Medicine, Università del Piemonte Orientale UPO, 28100 Novara, Italy; (M.B.); (C.M.); (P.P.S.); (C.R.); (C.D.B.); (G.A.); (M.P.)
| | - Pier Paolo Sainaghi
- Department of Translational Medicine, Università del Piemonte Orientale UPO, 28100 Novara, Italy; (M.B.); (C.M.); (P.P.S.); (C.R.); (C.D.B.); (G.A.); (M.P.)
- Division of Internal Medicine, “AOU Maggiore della Carità”, 28100 Novara, Italy
- CAAD, Centre for Autoimmune and Allergic Diseases, 28100 Novara, Italy
| | - Cristina Rigamonti
- Department of Translational Medicine, Università del Piemonte Orientale UPO, 28100 Novara, Italy; (M.B.); (C.M.); (P.P.S.); (C.R.); (C.D.B.); (G.A.); (M.P.)
- Division of Internal Medicine, “AOU Maggiore della Carità”, 28100 Novara, Italy
- CAAD, Centre for Autoimmune and Allergic Diseases, 28100 Novara, Italy
| | - Silvia Piantoni
- Rheumatology and Clinical Immunology Unit and Department of Clinical and Experimental Sciences, Spedali Civili and University of Brescia, 25128 Brescia, Italy; (L.A.); (S.P.)
| | - Carla De Benedittis
- Department of Translational Medicine, Università del Piemonte Orientale UPO, 28100 Novara, Italy; (M.B.); (C.M.); (P.P.S.); (C.R.); (C.D.B.); (G.A.); (M.P.)
- Division of Internal Medicine, “AOU Maggiore della Carità”, 28100 Novara, Italy
- CAAD, Centre for Autoimmune and Allergic Diseases, 28100 Novara, Italy
| | - Gianluca Aimaretti
- Department of Translational Medicine, Università del Piemonte Orientale UPO, 28100 Novara, Italy; (M.B.); (C.M.); (P.P.S.); (C.R.); (C.D.B.); (G.A.); (M.P.)
| | - Mario Pirisi
- Department of Translational Medicine, Università del Piemonte Orientale UPO, 28100 Novara, Italy; (M.B.); (C.M.); (P.P.S.); (C.R.); (C.D.B.); (G.A.); (M.P.)
- Division of Internal Medicine, “AOU Maggiore della Carità”, 28100 Novara, Italy
- CAAD, Centre for Autoimmune and Allergic Diseases, 28100 Novara, Italy
| | - Paolo Marzullo
- Department of Translational Medicine, Università del Piemonte Orientale UPO, 28100 Novara, Italy; (M.B.); (C.M.); (P.P.S.); (C.R.); (C.D.B.); (G.A.); (M.P.)
- Division of General Medicine, Ospedale S. Giuseppe, I.R.C.C.S. Istituto Auxologico Italiano, 28921 Verbania, Italy
| |
Collapse
|
18
|
Li SR, Tan ZX, Chen YH, Hu B, Zhang C, Wang H, Zhao H, Xu DX. Vitamin D deficiency exacerbates bleomycin-induced pulmonary fibrosis partially through aggravating TGF-β/Smad2/3-mediated epithelial-mesenchymal transition. Respir Res 2019; 20:266. [PMID: 31775746 PMCID: PMC6882226 DOI: 10.1186/s12931-019-1232-6] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Accepted: 11/05/2019] [Indexed: 12/19/2022] Open
Abstract
Background Our earlier report indicated that active vitamin D3 inhibited epithelial-mesenchymal transition (EMT) in bleomycin (BLM)-induced pulmonary fibrosis. The objective of this study was to further investigate whether vitamin D deficiency exacerbates BLM-induced pulmonary fibrosis. Methods This study consists of two independent experiments. Experiment 1, male mice were fed with vitamin D deficient (VDD) fodder. Experiment 2, Cyp27b1+/+, Cyp27b1+/− and Cyp27b1−/− mice were fed with standard diet. For pulmonary fibrosis, mice were intratracheally instilled with a single dose of BLM (1.5 mg/kg). Serum 25(OH) D level was measured. Pulmonary collagen deposition was assessed by Sirius red staining. EMT was measured and transforming growth factor-beta (TGF-β)/Smad3 signaling was evaluated in the lungs of BLM-treated mice. Results The relative weight of lungs was elevated in BLM-treated mice. Col1α1 and Col1α2, two collagen protein genes, were upregulated, and collagen deposition, as determined by Sirius red staining, was observed in the lungs of BLM-treated mice. E-cadherin, an epithelial marker, was downregulated. By contrast, vimentin and α-SMA, two EMT markers, were upregulated in the lungs of BLM-treated mice. Pulmonary TGF-β/Smad3 signaling was activated in BLM-induced lung fibrosis. Further analysis showed that feeding VDD diet, leading to vitamin D deficiency, aggravated elevation of BLM-induced relative lung weight. Moreover, feeding VDD diet aggravated BLM-induced TGF-β/Smad3 activation and subsequent EMT in the lungs. In addition, feeding VDD diet exacerbated BLM-induced pulmonary fibrosis. Additional experiment showed that Cyp27b1 gene knockout, leading to active vitamin D3 deficiency, exacerbated BLM-induced pulmonary fibrosis. Moreover, Cyp27b1 gene knockout aggravated pulmonary TGF-β/Smad2/3 activation and subsequent EMT in BLM-induced lung fibrosis. Conclusion Vitamin D deficiency exacerbates BLM-induced pulmonary fibrosis partially through aggravating TGF-β/Smad2/3-mediated EMT in the lungs.
Collapse
Affiliation(s)
- Se-Ruo Li
- Second Affiliated Hospital, Anhui Medical University, Hefei, 230032, China
| | - Zhu-Xia Tan
- Second Affiliated Hospital, Anhui Medical University, Hefei, 230032, China
| | - Yuan-Hua Chen
- Department of Toxicology, Anhui Medical University, 81 Meishan Road, Hefei, 230032, China
| | - Biao Hu
- Second Affiliated Hospital, Anhui Medical University, Hefei, 230032, China
| | - Cheng Zhang
- Department of Toxicology, Anhui Medical University, 81 Meishan Road, Hefei, 230032, China
| | - Hua Wang
- Department of Toxicology, Anhui Medical University, 81 Meishan Road, Hefei, 230032, China
| | - Hui Zhao
- Second Affiliated Hospital, Anhui Medical University, Hefei, 230032, China.
| | - De-Xiang Xu
- Department of Toxicology, Anhui Medical University, 81 Meishan Road, Hefei, 230032, China.
| |
Collapse
|
19
|
Wang PF, Yao DH, Hu YY, Li Y. Vitamin D Improves Intestinal Barrier Function in Cirrhosis Rats by Upregulating Heme Oxygenase-1 Expression. Biomol Ther (Seoul) 2019; 27:222-230. [PMID: 30173501 PMCID: PMC6430230 DOI: 10.4062/biomolther.2018.052] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Revised: 06/04/2018] [Accepted: 07/17/2018] [Indexed: 12/21/2022] Open
Abstract
Intestinal barrier dysfunction always accompanies cirrhosis in patients with advanced liver disease and is an important contributor facilitating bacterial translocation (BT), which has been involved in the pathogenesis of cirrhosis and its complications. Several studies have demonstrated the protective effect of Vitamin D on intestinal barrier function. However, severe cholestasis leads to vitamin D depletion. This study was designed to test whether vitamin D therapy improves intestinal dysfunction in cirrhosis. Rats were subcutaneously injected with 50% sterile CCl4 (a mixture of pure CCl4 and olive oil, 0.3 mL/100 g) twice a week for 6 weeks. Next, 1,25(OH)2D3 (0.5 µg/100 g) and the vehicle were administered simultaneously with CCl4 to compare the extent of intestinal histologic damage, tight junction protein expression, intestinal barrier function, BT, intestinal proliferation, apoptosis, and enterocyte turnover. Intestinal heme oxygenase-1 (HO-1) expression and oxidative stress were also assessed. We found that vitamin D could maintain intestinal epithelial proliferation and turnover, inhibit intestinal epithelial apoptosis, alleviate structural damage, and prevent BT and intestinal barrier dysfunction. These were achieved partly through restoration of HO-1 and inhibition of oxidative stress. Taken together, our results suggest that vitamin D ameliorated intestinal epithelial turnover and improved the integrity and function of intestinal barrier in CCl4-induced liver cirrhotic rats. HO-1 signaling activation was involved in these above beneficial effects.
Collapse
Affiliation(s)
- Peng-Fei Wang
- Department of General Surgery, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200011, China
| | - Dan-Hua Yao
- Department of General Surgery, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200011, China
| | - Yue-Yu Hu
- Department of Neurology, The Fourth Affiliated Hospital of Tongji University, Shanghai 200081, China
| | - Yousheng Li
- Department of General Surgery, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200011, China
| |
Collapse
|
20
|
Udomsinprasert W, Jittikoon J. Vitamin D and liver fibrosis: Molecular mechanisms and clinical studies. Biomed Pharmacother 2018; 109:1351-1360. [PMID: 30551386 DOI: 10.1016/j.biopha.2018.10.140] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Revised: 10/22/2018] [Accepted: 10/24/2018] [Indexed: 12/21/2022] Open
Abstract
Vitamin D plays a primary role in regulation of bone metabolism and calcium homeostasis. Interestingly, emerging evidence suggests protective effects of vitamin D against liver fibrogenesis. However, the precise mechanisms of this action remain mysterious. Herein, this review aimed to summarize the role of vitamin D in liver fibrosis pathology and to update the current comprehensive knowledge regarding the clinical utility of vitamin D-based treatment in liver fibrosis. In regard to its effect on liver fibrosis, vitamin D possesses an anti-fibrotic effect on hepatic stellate cells via vitamin D receptor-mediated specific signal transduction pathways, which in turn inhibit expression of pro-fibrogenic genes. Furthermore, several studies demonstrated a significant association between low vitamin D levels and an increased risk of liver fibrosis. Additionally, high prevalence of vitamin D deficiency was noted in patients with liver fibrosis, suggesting the use of vitamin D status as a biochemical marker reflecting the progression of liver fibrosis. It is therefore reasonable to postulate that vitamin D supplementation being a cost effective and relative simple procedure may benefit to liver fibrosis. Nevertheless, further research is needed to fully elucidate its regulatory role in inhibiting liver fibrogenesis and to estimate the safety and efficiency of vitamin D supplementation as a relatively inexpensive treatment for liver fibrosis in patients with chronic liver diseases.
Collapse
Affiliation(s)
- Wanvisa Udomsinprasert
- Department of Biochemistry, Faculty of Pharmacy, Mahidol University, Bangkok 10400, Thailand.
| | - Jiraphun Jittikoon
- Department of Biochemistry, Faculty of Pharmacy, Mahidol University, Bangkok 10400, Thailand.
| |
Collapse
|
21
|
Zhao G, Elhafiz M, Jiang J, Das D, Li Z, Zhou W, Fan S, Wang C, Yuan Z, Xu D, Jiang Z, Zhang L, Wang T. Adaptive homeostasis of the vitamin D-vitamin D nuclear receptor axis in 8-methoxypsoralen-induced hepatotoxicity. Toxicol Appl Pharmacol 2018; 362:150-158. [PMID: 30419252 DOI: 10.1016/j.taap.2018.11.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Revised: 10/31/2018] [Accepted: 11/02/2018] [Indexed: 10/27/2022]
Abstract
8-methoxypsoralen (8-MOP) with ultraviolet A radiation therapy (PUVA) is the standard therapy for patients with psoriasis, despite the reported potential risks of 8-MOP-induced cholestatic liver injury in both humans and animals. Usually, patients with chronic cholestasis exhibit lower serum 25-hydroxy vitamin D (25(OH)D) levels. But those patients receiving PUVA for psoriasis showed an increase in serum 25(OH)D levels, probably highlighting that the vitamin D-vitamin D nuclear receptor (VD-VDR) axis play a protective role in 8-MOP-induced hepatotoxicity. The present study confirmed 8-MOP could increase serum 25(OH)D levels in conventional lighting and diet (CLD) and vitamin D deficient (VDD) Sprague-Dawley rats. Potential liver risks were also found in CLD and VDD rats after 8-MOP treatment. We proved that 8-MOP could be a potent ligand for VDR using molecular docking and luciferase report assay. Effect of 8-MOP on VDR subcellular distribution was determined using human liver cell line L02. We found 8-MOP could increase VDR protein expression in the nuclear and cytosol extracts and also total cell extracts in L02. siRNAs for VDR were used to determine the role of VDR in protecting 8-MOP-induced cholestasis and potential cellular mechanisms. The results showed 8-MOP could affect the CYP7A1, SHP and MRP3 expression via VDR, and such effects could be reversed by knockdown of VDR expression, suggesting a vital role of VDR involved in 8-MOP-regulated bile acid synthesis and transportation. In conclusion, these results revealed activation of VD-VDR axis may play a beneficial role in 8-MOP-mediated regulation of bile acid synthesis and transportation.
Collapse
Affiliation(s)
- Guolin Zhao
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing 210009, China
| | - Muhanad Elhafiz
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing 210009, China
| | - Jingwei Jiang
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing 210009, China
| | - Debanjan Das
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing 210009, China
| | - Zhijian Li
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing 210009, China; Department of Pharmacology and Toxicology Laboratory, Xinjiang Institute of Traditional Uighur Medicine, Urumqi, Xinjiang 830049, China
| | - Wang Zhou
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing 210009, China
| | - Sisi Fan
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing 210009, China
| | - Changling Wang
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing 210009, China
| | - Ziqiao Yuan
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing 210009, China
| | - Dengqiu Xu
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing 210009, China
| | - Zhenzhou Jiang
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing 210009, China; Key Laboratory of Drug Quality Control and Pharmacovigilance, China Pharmaceutical University, Nanjing 210009, China
| | - Luyong Zhang
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing 210009, China; Center for Drug Screening and Pharmacodynamics Evaluation, School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China; Jiangsu Center for Pharmacodynamics Research and Evaluation, China Pharmaceutical University, Nanjing 210009, China; State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China.
| | - Tao Wang
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing 210009, China; Jiangsu Center for Pharmacodynamics Research and Evaluation, China Pharmaceutical University, Nanjing 210009, China.
| |
Collapse
|
22
|
Hua H, Dai M, Luo Y, Lin H, Xu G, Hu X, Xu L, Zhang H, Tang Z, Chang L, Liu A, Yang J. Basal PPARα inhibits bile acid metabolism adaptation in chronic cholestatic model induced by α-naphthylisothiocyanate. Toxicol Lett 2018; 300:31-39. [PMID: 30352267 DOI: 10.1016/j.toxlet.2018.10.015] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Revised: 09/26/2018] [Accepted: 10/12/2018] [Indexed: 12/12/2022]
Abstract
Cholestasis is one of the most challenging diseases to be treated in current hepatology. However little is known about the adaptation difference and the underlying mechanism between acute and chronic cholestasis. In this study, wild-type and Pparα-null mice were orally administered diet containing 0.05% ANIT to induce chronic cholestasis. Biochemistry, histopathology and serum metabolome analysis exhibited the similar toxic phenotype between wild-type and Pparα-null mice. Bile acid metabolism was strongly adapted in Pparα-null mice but not in wild-type mice. The Shp and Fxr mRNA was found to be doubled in cholestatic Pparα-null mice compared with the control group. Western blot confirmed the up-regulated expression of FXR in Pparα-null mice treated with ANIT. Inflammation was found to be stronger in Pparα-null mice than those in wild-type mice in chronic cholestasis. These data chain indicated that bile acid metabolism and inflammation signaling were different between wild-type and Pparα-null mice developing chronic cholestasis, although their toxic phenotypes could not be discriminated. So basal PPARα cross-talked with FXR and inhibited bile acid metabolism adaptation in chronic cholestasis.
Collapse
Affiliation(s)
- Huiying Hua
- Medical School of Ningbo University, Ningbo 315211, China
| | - Manyun Dai
- Medical School of Ningbo University, Ningbo 315211, China; State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, 650201, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yishuang Luo
- Medical School of Ningbo University, Ningbo 315211, China
| | - Hante Lin
- Medical School of Ningbo University, Ningbo 315211, China
| | - Gangming Xu
- Medical School of Ningbo University, Ningbo 315211, China
| | - Xiaowei Hu
- Medical School of Ningbo University, Ningbo 315211, China
| | - Liping Xu
- Medical School of Ningbo University, Ningbo 315211, China
| | - Haoyue Zhang
- Medical School of Ningbo University, Ningbo 315211, China
| | - Zhiyuan Tang
- Medical School of Ningbo University, Ningbo 315211, China
| | - Liming Chang
- Medical School of Ningbo University, Ningbo 315211, China
| | - Aiming Liu
- Medical School of Ningbo University, Ningbo 315211, China.
| | - Julin Yang
- Ningbo College of Health Sciences, Ningbo 315100, China.
| |
Collapse
|
23
|
Gorman S, Buckley AG, Ling KM, Berry LJ, Fear VS, Stick SM, Larcombe AN, Kicic A, Hart PH. Vitamin D supplementation of initially vitamin D-deficient mice diminishes lung inflammation with limited effects on pulmonary epithelial integrity. Physiol Rep 2018; 5:5/15/e13371. [PMID: 28774952 PMCID: PMC5555896 DOI: 10.14814/phy2.13371] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2017] [Revised: 07/03/2017] [Accepted: 07/07/2017] [Indexed: 12/18/2022] Open
Abstract
In disease settings, vitamin D may be important for maintaining optimal lung epithelial integrity and suppressing inflammation, but less is known of its effects prior to disease onset. Female BALB/c dams were fed a vitamin D3‐supplemented (2280 IU/kg, VitD+) or nonsupplemented (0 IU/kg, VitD−) diet from 3 weeks of age, and mated at 8 weeks of age. Male offspring were fed the same diet as their mother. Some offspring initially fed the VitD− diet were switched to a VitD+ diet from 8 weeks of age (VitD−/+). At 12 weeks of age, signs of low‐level inflammation were observed in the bronchoalveolar lavage fluid (BALF) of VitD− mice (more macrophages and neutrophils), which were suppressed by subsequent supplementation with vitamin D3. There was no difference in the level of expression of the tight junction proteins occludin or claudin‐1 in lung epithelial cells of VitD+ mice compared to VitD− mice; however, claudin‐1 levels were reduced when initially vitamin D‐deficient mice were fed the vitamin D3‐containing diet (VitD−/+). Reduced total IgM levels were detected in BALF and serum of VitD−/+ mice compared to VitD+ mice. Lung mRNA levels of the vitamin D receptor (VDR) were greatest in VitD−/+ mice. Total IgG levels in BALF were greater in mice fed the vitamin D3‐containing diet, which may be explained by increased activation of B cells in airway‐draining lymph nodes. These findings suggest that supplementation of initially vitamin D‐deficient mice with vitamin D3 suppresses signs of lung inflammation but has limited effects on the epithelial integrity of the lungs.
Collapse
Affiliation(s)
- Shelley Gorman
- Telethon Kids Institute University of Western Australia, Subiaco, Western Australia, Australia
| | - Alysia G Buckley
- Centre of Microscopy, Characterisation and Analysis The University of Western Australia, Nedlands, Western Australia, Australia
| | - Kak-Ming Ling
- Telethon Kids Institute University of Western Australia, Subiaco, Western Australia, Australia
| | - Luke J Berry
- Telethon Kids Institute University of Western Australia, Subiaco, Western Australia, Australia
| | - Vanessa S Fear
- Telethon Kids Institute University of Western Australia, Subiaco, Western Australia, Australia
| | - Stephen M Stick
- Telethon Kids Institute University of Western Australia, Subiaco, Western Australia, Australia.,Department of Respiratory Medicine, Princess Margaret Hospital for Children, Perth, Western Australia, Australia.,School of Paediatrics and Child Health The University of Western Australia, Nedlands, Western Australia, Australia.,Centre for Cell Therapy and Regenerative Medicine, School of Medicine and Pharmacology The University of Western Australia and Harry Perkins Institute of Medical Research, Nedlands, Western Australia, Australia
| | - Alexander N Larcombe
- Telethon Kids Institute University of Western Australia, Subiaco, Western Australia, Australia.,Occupation and Environment School of Public Health Curtin University, Perth, Western Australia, Australia
| | - Anthony Kicic
- Telethon Kids Institute University of Western Australia, Subiaco, Western Australia, Australia.,Department of Respiratory Medicine, Princess Margaret Hospital for Children, Perth, Western Australia, Australia.,School of Paediatrics and Child Health The University of Western Australia, Nedlands, Western Australia, Australia.,Centre for Cell Therapy and Regenerative Medicine, School of Medicine and Pharmacology The University of Western Australia and Harry Perkins Institute of Medical Research, Nedlands, Western Australia, Australia.,Occupation and Environment School of Public Health Curtin University, Perth, Western Australia, Australia
| | - Prue H Hart
- Telethon Kids Institute University of Western Australia, Subiaco, Western Australia, Australia
| |
Collapse
|
24
|
Keane JT, Elangovan H, Stokes RA, Gunton JE. Vitamin D and the Liver-Correlation or Cause? Nutrients 2018; 10:nu10040496. [PMID: 29659559 PMCID: PMC5946281 DOI: 10.3390/nu10040496] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Revised: 04/09/2018] [Accepted: 04/11/2018] [Indexed: 02/06/2023] Open
Abstract
Vitamin D is becoming increasingly accepted as an important physiological regulator outside of its classical role in skeletal homeostasis. A growing body of evidence connects vitamin D with hepatic disease. This review summarises the role of vitamin D in liver homeostasis and disease and discusses the therapeutic potential of vitamin D-based treatments to protect against hepatic disease progression and to improve response to treatment. While pre-clinical experimental data is promising, clinical trials around liver diseases have mostly been under-powered, and further studies will be required to clarify whether vitamin D or vitamin D analogues have beneficial effects on liver disease.
Collapse
Affiliation(s)
- Jeremy T Keane
- Centre for Diabetes, Obesity & Endocrinology, The Westmead Institute for Medical Research (WIMR), Westmead, Sydney, NSW 2145, Australia.
| | - Harendran Elangovan
- The Garvan Institute of Medical Research, The University of New South Wales (UNSW), Darlinghurst, Sydney, NSW 2010, Australia.
| | - Rebecca A Stokes
- Centre for Diabetes, Obesity & Endocrinology, The Westmead Institute for Medical Research (WIMR), Westmead, Sydney, NSW 2145, Australia.
- The Garvan Institute of Medical Research, The University of New South Wales (UNSW), Darlinghurst, Sydney, NSW 2010, Australia.
| | - Jenny E Gunton
- Centre for Diabetes, Obesity & Endocrinology, The Westmead Institute for Medical Research (WIMR), Westmead, Sydney, NSW 2145, Australia.
- The Garvan Institute of Medical Research, The University of New South Wales (UNSW), Darlinghurst, Sydney, NSW 2010, Australia.
- Faculty of Medicine and Health, The University of Sydney, Westmead Hospital, Westmead, Sydney, NSW 2145, Australia.
| |
Collapse
|
25
|
Pellat A, Vaquero J, Fouassier L. Role of ErbB/HER family of receptor tyrosine kinases in cholangiocyte biology. Hepatology 2018; 67:762-773. [PMID: 28671339 DOI: 10.1002/hep.29350] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2017] [Revised: 05/18/2017] [Accepted: 06/28/2017] [Indexed: 12/17/2022]
Abstract
The ErbB/HER family comprises four distinct tyrosine kinase receptors, EGFR/ErbB1/HER1, ErbB2/HER2, ErbB3/HER3, and ErbB4/HER4, which trigger intracellular signals at the origin of essential cellular functions, including differentiation, proliferation, survival, and migration. Epithelial cells, named cholangiocytes, that line intrahepatic and extrahepatic bile ducts, contribute substantially to biliary secretory functions and bile transport. Although ErbB receptors have been widely studied in cholangiocarcinoma (CCA), a malignancy of the biliary tract, knowledge of these receptors in biliary epithelium physiology and in non-malignant cholangiopathies is far from complete. Current knowledge suggests a role for epidermal growth factor receptor (EGFR) in cholangiocyte specification and proliferation, and in hepatocyte transdifferentiation into cholangiocytes during liver regeneration to restore biliary epithelium integrity. High expression and activation of EGFR and/or ErbB2 were recently demonstrated in biliary lithiasis and primary sclerosing cholangitis, two cholangiopathies regarded as risk factors for CCA. In CCA, ErbB receptors are frequently overexpressed, leading to tumor progression and low prognosis. Anti-ErbB therapies were efficient only in preclinical trials and have suggested the existence of resistance mechanisms with the need to identify predictive factors of therapy response. This review aims to compile the current knowledge on the functions of ErbB receptors in physiology and physiopathology of the biliary epithelium. (Hepatology 2018;67:762-773).
Collapse
Affiliation(s)
- Anna Pellat
- Sorbonne Universités, UPMC Université Paris 06, INSERM, Centre de Recherche Saint-Antoine (CRSA), Paris, France
| | - Javier Vaquero
- Sorbonne Universités, UPMC Université Paris 06, INSERM, Centre de Recherche Saint-Antoine (CRSA), Paris, France.,FONDATION ARC, Villejuif, France
| | - Laura Fouassier
- Sorbonne Universités, UPMC Université Paris 06, INSERM, Centre de Recherche Saint-Antoine (CRSA), Paris, France
| |
Collapse
|
26
|
Gonzalez-Sanchez E, Firrincieli D, Housset C, Chignard N. Expression patterns of nuclear receptors in parenchymal and non-parenchymal mouse liver cells and their modulation in cholestasis. Biochim Biophys Acta Mol Basis Dis 2017; 1863:1699-1708. [PMID: 28390947 DOI: 10.1016/j.bbadis.2017.04.004] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2016] [Revised: 03/02/2017] [Accepted: 04/04/2017] [Indexed: 12/19/2022]
Abstract
Nuclear receptors (NR), the largest family of transcription factors, control many physiological and pathological processes. To gain insight into hepatic NR and their potential as therapeutic targets in cholestatis, we determined their expression in individual cell types of the mouse liver in normal and cholestatic conditions. Hepatocytes, cholangiocytes, hepatic stellate cells (HSC), sinusoidal endothelial cells (SEC) and Kupffer cells (KC) were isolated from the liver of mice with acute or chronic cholestasis (i.e. bile duct-ligated or Abcb4-/- mice, respectively) and healthy controls. The expression of 43 out of the 49 NR was evidenced by RT-qPCR in one or several liver cell types. Expression of four NR was restricted to non-parenchymal liver cells. In normal conditions, NR were expressed at higher levels in individual cell types when compared to total liver. Half of the NR expressed in the liver had maximal expression in non-parenchymal cells. After bile duct ligation, NR mRNA changes occurred mostly in non-parenchymal cells and mainly consisted in down-regulations. In Abcb4-/- mice, NR mRNA changes were equally frequent in hepatocytes and non-parenchymal cells. Essentially down-regulations were found in hepatocytes, HSC and cholangiocytes, as opposed to up-regulations in SEC and KC. While undetectable in total liver, Vdr expression was up-regulated in all non-parenchymal cells in Abcb4-/- mice. In conclusion, non-parenchymal liver cells are a major site of NR expression. During cholestasis, NR expression is markedly altered mainly by down-regulations, suggesting major changes in metabolic activity. Thus, non-parenchymal cells are important new targets to consider in NR-directed therapies.
Collapse
Affiliation(s)
- Ester Gonzalez-Sanchez
- Sorbonne Universités, UPMC Univ Paris 06, INSERM, Centre de Recherche Saint-Antoine (CRSA), Institute of Cardiometabolism and Nutrition (ICAN), F-75012 Paris, France.
| | - Delphine Firrincieli
- Sorbonne Universités, UPMC Univ Paris 06, INSERM, Centre de Recherche Saint-Antoine (CRSA), Institute of Cardiometabolism and Nutrition (ICAN), F-75012 Paris, France.
| | - Chantal Housset
- Sorbonne Universités, UPMC Univ Paris 06, INSERM, Centre de Recherche Saint-Antoine (CRSA), Institute of Cardiometabolism and Nutrition (ICAN), F-75012 Paris, France; Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Saint-Antoine, Centre de Référence Maladies Rares des Maladies Inflammatoires des Voies Biliaires & Service d'Hépatologie, F-75012 Paris, France.
| | - Nicolas Chignard
- Sorbonne Universités, UPMC Univ Paris 06, INSERM, Centre de Recherche Saint-Antoine (CRSA), Institute of Cardiometabolism and Nutrition (ICAN), F-75012 Paris, France; Inovarion, F-75013 Paris, France.
| |
Collapse
|
27
|
SARNOVA L, GREGOR M. Biliary System Architecture: Experimental Models and Visualization Techniques. Physiol Res 2017; 66:383-390. [PMID: 28248543 DOI: 10.33549/physiolres.933499] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
The complex architecture of the liver biliary network represents a structural prerequisite for the formation and secretion of bile as well as excretion of toxic substances through bile ducts. Disorders of the biliary tract affect a significant portion of the worldwide population, often leading to cholestatic liver diseases. Cholestatic liver disease is a condition that results from an impairment of bile formation or bile flow to the gallbladder and duodenum. Cholestasis leads to dramatic changes in biliary tree architecture, worsening liver disease and systemic illness. Recent studies show that the prevalence of cholestatic liver diseases is increasing. The availability of well characterized animal models, as well as development of visualization approaches constitutes a critical asset to develop novel pathogenetic concepts and new treatment strategies.
Collapse
Affiliation(s)
| | - M. GREGOR
- Laboratory of Integrative Biology, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic
| |
Collapse
|
28
|
Pincikova T, Paquin-Proulx D, Sandberg JK, Flodström-Tullberg M, Hjelte L. Vitamin D treatment modulates immune activation in cystic fibrosis. Clin Exp Immunol 2017; 189:359-371. [PMID: 28470739 DOI: 10.1111/cei.12984] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/19/2017] [Indexed: 12/11/2022] Open
Abstract
Persistent inflammatory response in cystic fibrosis (CF) airways is believed to play a central role in the progression of lung damage. Anti-inflammatory treatment may slow lung disease progression, but adverse side effects have limited its use. Vitamin D has immunoregulatory properties. We randomized 16 CF patients to receive vitamin D2, vitamin D3 or to serve as controls, and investigated the effect of vitamin D supplementation on soluble immunological parameters, myeloid dendritic cells (mDCs) and T cell activation. Three months of vitamin D treatment were followed by two washout months. Vitamin D status at baseline was correlated negatively with haptoglobin, erythrocyte sedimentation rate and immunoglobulin A concentration. Total vitamin D dose per kg bodyweight correlated with the down-modulation of the co-stimulatory receptor CD86 on mDCs. Vitamin D treatment was associated with reduced CD279 (PD-1) expression on CD4+ and CD8+ T cells, as well as decreased frequency of CD8+ T cells co-expressing the activation markers CD38 and human leucocyte antigen D-related (HLA-DR) in a dose-dependent manner. There was a trend towards decreased mucosal-associated invariant T cells (MAIT) cell frequency in patients receiving vitamin D and free serum 25-hydroxyvitamin D (free-s25OHD) correlated positively with CD38 expression by these cells. At the end of intervention, the change in free-s25OHD was correlated negatively with the change in CD279 (PD-1) expression on MAIT cells. Collectively, these data indicate that vitamin D has robust pleiotropic immunomodulatory effects in CF. Larger studies are needed to explore the immunomodulatory treatment potential of vitamin D in CF in more detail.
Collapse
Affiliation(s)
- T Pincikova
- Stockholm CF Center, Karolinska University Hospital Huddinge, Stockholm, Sweden.,Division of Pediatrics, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden.,Center for Infectious Medicine, Department of Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - D Paquin-Proulx
- Center for Infectious Medicine, Department of Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - J K Sandberg
- Center for Infectious Medicine, Department of Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - M Flodström-Tullberg
- Center for Infectious Medicine, Department of Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - L Hjelte
- Stockholm CF Center, Karolinska University Hospital Huddinge, Stockholm, Sweden.,Division of Pediatrics, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
29
|
Elangovan H, Chahal S, Gunton JE. Vitamin D in liver disease: Current evidence and potential directions. Biochim Biophys Acta Mol Basis Dis 2017; 1863:907-916. [PMID: 28064017 DOI: 10.1016/j.bbadis.2017.01.001] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2016] [Revised: 12/06/2016] [Accepted: 01/02/2017] [Indexed: 01/10/2023]
Abstract
Consistent with its multifaceted nature, growing evidence links vitamin D with hepatic disease. In this review, we summarise the roles of vitamin D in different liver pathologies and explore the clinical utility of vitamin D-based treatments in hepatology. We find that the small number of clinical trials coupled with the profound heterogeneity of study protocols limits the strength of evidence needed to ascribe definite clinical value to the hormone in liver disease. Nevertheless, the experimental data is promising and further bench and bedside studies will likely define a clearer role in hepatic therapeutics.
Collapse
Affiliation(s)
- Harendran Elangovan
- The Garvan Institute of Medical Research, The University of New South Wales (UNSW), Sydney, NSW, Australia
| | - Sarinder Chahal
- The Garvan Institute of Medical Research, The University of New South Wales (UNSW), Sydney, NSW, Australia
| | - Jenny E Gunton
- The Garvan Institute of Medical Research, The University of New South Wales (UNSW), Sydney, NSW, Australia; The Westmead Institute of Medical Research, The University of Sydney, NSW, Australia.
| |
Collapse
|
30
|
Bozic M, Guzmán C, Benet M, Sánchez-Campos S, García-Monzón C, Gari E, Gatius S, Valdivielso JM, Jover R. Hepatocyte vitamin D receptor regulates lipid metabolism and mediates experimental diet-induced steatosis. J Hepatol 2016; 65:748-757. [PMID: 27245430 DOI: 10.1016/j.jhep.2016.05.031] [Citation(s) in RCA: 75] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2015] [Revised: 05/03/2016] [Accepted: 05/23/2016] [Indexed: 01/10/2023]
Abstract
BACKGROUND & AIMS The pathogenesis and progression of non-alcoholic fatty liver disease (NAFLD) is still incompletely understood. Several nuclear receptors play a role in liver lipid metabolism and can promote hepatosteatosis, but the possible role of vitamin D receptor (VDR) in NAFLD has not been investigated. METHODS The expression of liver VDR was investigated in apolipoprotein E knockout (apoE(-/-)) mice on a high fat diet, in wild-type mice on methionine and choline deficient diet and in NAFLD patients with hepatosteatosis and non-alcoholic steatohepatitis. The relevance of VDR was assessed in apoE(-/-) mice by deletion of VDR or paricalcitol treatment and in human HepG2 cells by VDR transfection or silencing. The role of VDR in fibrosis was also determined in VDR knockout mice (VDR(-/-)) treated with thioacetamide. RESULTS Expression of liver VDR was markedly induced in two mouse models of NAFLD, as well as in patients with hepatosteatosis, but decreased in non-alcoholic steatohepatitis. VDR deletion in high fat diet-fed apoE(-/-) mice protected against fatty liver, dyslipidemia and insulin resistance, and caused a decrease in taurine-conjugated bile acids, but did not influence fibrosis by thioacetamide. apoE(-/-)VDR(-/-) mouse livers showed decreased gene expression of CD36, DGAT2, C/EBPα and FGF21, and increased expression of PNPLA2, LIPIN1 and PGC1α. Treatment of apoE(-/-) mice on high fat diet with paricalcitol had modest opposite effects on steatosis and gene expression. Finally, this set of genes showed concordant responses when VDR was overexpressed or silenced in HepG2 cells. CONCLUSIONS Induced hepatocyte VDR in NAFLD regulates key hepatic lipid metabolism genes and promotes high fat diet-associated liver steatosis. Therapeutic inhibition of liver VDR may reverse steatosis in early NAFLD. LAY SUMMARY The amount of vitamin D receptor is induced early in the livers of mice and humans when they develop non-alcoholic fatty liver disease. If the gene for the vitamin D receptor is deleted, hepatic lipid metabolism changes and mice do not accumulate fat in the liver. We conclude that the vitamin D receptor can contribute to the fatty liver disease promoted by a high fat diet.
Collapse
Affiliation(s)
- Milica Bozic
- Experimental Nephrology Laboratory, IRBLLEIDA, Lleida, Spain
| | - Carla Guzmán
- Experimental Hepatology Unit, IIS Hospital La Fe, Valencia, Spain
| | - Marta Benet
- Experimental Hepatology Unit, IIS Hospital La Fe, Valencia, Spain
| | - Sonia Sánchez-Campos
- Institute of Biomedicine (IBIOMED), University of León, Spain; CIBERehd, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas, Spain
| | - Carmelo García-Monzón
- Liver Research Unit, Hospital Santa Cristina, IIS Princesa, Madrid, Spain; CIBERehd, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas, Spain
| | - Eloi Gari
- Dep. Ciències Mèdiques Bàsiques, Universitat de Lleida, Spain
| | - Sonia Gatius
- Department of Pathology and Molecular Genetics, HUAV, IRBLLEIDA, Lleida, Spain
| | | | - Ramiro Jover
- Experimental Hepatology Unit, IIS Hospital La Fe, Valencia, Spain; CIBERehd, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas, Spain; Dep. Biochemistry and Molecular Biology, University of Valencia, Spain.
| |
Collapse
|
31
|
Delaunay JL, Durand-Schneider AM, Dossier C, Falguières T, Gautherot J, Davit-Spraul A, Aït-Slimane T, Housset C, Jacquemin E, Maurice M. A functional classification of ABCB4 variations causing progressive familial intrahepatic cholestasis type 3. Hepatology 2016; 63:1620-31. [PMID: 26474921 DOI: 10.1002/hep.28300] [Citation(s) in RCA: 75] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Revised: 09/24/2015] [Accepted: 10/15/2015] [Indexed: 12/12/2022]
Abstract
UNLABELLED Progressive familial intrahepatic cholestasis type 3 is caused by biallelic variations of ABCB4, most often (≥70%) missense. In this study, we examined the effects of 12 missense variations identified in progressive familial intrahepatic cholestasis type 3 patients. We classified these variations on the basis of the defects thus identified and explored potential rescue of trafficking-defective mutants by pharmacological means. Variations were reproduced in the ABCB4 complementary DNA and the mutants, thus obtained, expressed in HepG2 and HEK293 cells. Three mutants were either fully (I541F and L556R) or largely (Q855L) retained in the endoplasmic reticulum, in an immature form. Rescue of the defect, i.e., increase in the mature form at the bile canaliculi, was obtained by cell treatments with cyclosporin A or C and, to a lesser extent, B, D, or H. Five mutations with little or no effect on ABCB4 expression at the bile canaliculi caused a decrease (F357L, T775M, and G954S) or almost absence (S346I and P726L) of phosphatidylcholine secretion. Two mutants (T424A and N510S) were normally processed and expressed at the bile canaliculi, but their stability was reduced. We found no defect of the T175A mutant or of R652G, previously described as a polymorphism. In patients, the most severe phenotypes appreciated by the duration of transplant-free survival were caused by ABCB4 variants that were markedly retained in the endoplasmic reticulum and expressed in a homozygous status. CONCLUSION ABCB4 variations can be classified as follows: nonsense variations (I) and, on the basis of current findings, missense variations that primarily affect the maturation (II), activity (III), or stability (IV) of the protein or have no detectable effect (V); this classification provides a strong basis for the development of genotype-based therapies.
Collapse
Affiliation(s)
- Jean-Louis Delaunay
- Sorbonne Universités, UPMC Université Paris 06, INSERM, UMR_S 938, Centre de Recherche Saint-Antoine, Paris, France
| | - Anne-Marie Durand-Schneider
- Sorbonne Universités, UPMC Université Paris 06, INSERM, UMR_S 938, Centre de Recherche Saint-Antoine, Paris, France
| | - Claire Dossier
- Sorbonne Universités, UPMC Université Paris 06, INSERM, UMR_S 938, Centre de Recherche Saint-Antoine, Paris, France
| | - Thomas Falguières
- Sorbonne Universités, UPMC Université Paris 06, INSERM, UMR_S 938, Centre de Recherche Saint-Antoine, Paris, France
| | - Julien Gautherot
- Sorbonne Universités, UPMC Université Paris 06, INSERM, UMR_S 938, Centre de Recherche Saint-Antoine, Paris, France
| | - Anne Davit-Spraul
- Assistance Publique-Hôpitaux de Paris, Hôpital de Bicêtre, Laboratoire de biochimie, Le Kremlin Bicêtre, France
| | - Tounsia Aït-Slimane
- Sorbonne Universités, UPMC Université Paris 06, INSERM, UMR_S 938, Centre de Recherche Saint-Antoine, Paris, France
| | - Chantal Housset
- Sorbonne Universités, UPMC Université Paris 06, INSERM, UMR_S 938, Centre de Recherche Saint-Antoine, Paris, France.,Assistance Publique-Hôpitaux de Paris, Hôpital Saint-Antoine, Centre de Référence Maladies Rares Maladies Inflammatoires des Voies Biliaires & Service d'Hépatologie, Paris, France
| | - Emmanuel Jacquemin
- Assistance Publique-Hôpitaux de Paris, Hôpital Bicêtre, Hépatologie Pédiatrique & Unité de Transplantation Hépatique, Centre de Référence Maladies Rares Atrésies des Voies Biliaires de l'Enfant, Le Kremlin Bicêtre, France.,Université Paris-Sud 11, INSERM, UMR_S 1174, Hepatinov, Orsay, France
| | - Michèle Maurice
- Sorbonne Universités, UPMC Université Paris 06, INSERM, UMR_S 938, Centre de Recherche Saint-Antoine, Paris, France
| |
Collapse
|
32
|
Ali AH, Tabibian JH, Carey EJ, Lindor KD. Emerging drugs for the treatment of Primary Biliary Cholangitis. Expert Opin Emerg Drugs 2016; 21:39-56. [PMID: 26901615 DOI: 10.1517/14728214.2016.1150999] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
INTRODUCTION Primary biliary cholangitis (PBC) is an autoimmune chronic disease of the liver that can progress to cirrhosis and hepatocellular carcinoma. It affects approximately 1 in 4,000 with a 10:1 female to male ratio. The diagnosis of PBC can be made based on serum antimitochondrial antibodies (AMA) in a patient with abnormally high serum alkaline phosphatase after ruling out other causes of cholestasis and biliary obstruction. Genome-wide association studies have revealed several human leukocyte antigen (HLA) and non-HLA risk loci in PBC, and complex environmental-host immunogenetic interactions are believed to underlie the etiopathogenesis of the disease. Fatigue and pruritus are the most common and often problematic symptoms; although often mild, these can be severe and life-alternating in a subset of patients. Ursodeoxycholic acid (UDCA) is the only drug approved by the United States Food and Drug Administration for the treatment of PBC. Clinical trials have shown that UDCA significantly improves transplant-free survival. However, nearly 40% of PBC patients do not respond adequately to PBC and are at higher risk for serious complications when compared to PBC patients with complete response to UDCA. AREAS COVERED Here we provide a detailed discussion regarding novel therapeutic agents and potential areas for further investigation in PBC-related research. EXPERT OPINION Results of ongoing clinical trials and emerging treatment paradigms for PBC will likely further improve medical management of this disorder in the near future.
Collapse
|
33
|
Pharmacology of bile acid receptors: Evolution of bile acids from simple detergents to complex signaling molecules. Pharmacol Res 2015; 104:9-21. [PMID: 26706784 DOI: 10.1016/j.phrs.2015.12.007] [Citation(s) in RCA: 175] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2015] [Accepted: 12/03/2015] [Indexed: 12/17/2022]
Abstract
For many years, bile acids were thought to only function as detergents which solubilize fats and facilitate the uptake of fat-soluble vitamins in the intestine. Many early observations; however, demonstrated that bile acids regulate more complex processes, such as bile acids synthesis and immune cell function through activation of signal transduction pathways. These studies were the first to suggest that receptors may exist for bile acids. Ultimately, seminal studies by many investigators led to the discovery of several bile acid-activated receptors including the farnesoid X receptor, the vitamin D receptor, the pregnane X receptor, TGR5, α5 β1 integrin, and sphingosine-1-phosphate receptor 2. Several of these receptors are expressed outside of the gastrointestinal system, indicating that bile acids may have diverse functions throughout the body. Characterization of the functions of these receptors over the last two decades has identified many important roles for these receptors in regulation of bile acid synthesis, transport, and detoxification; regulation of glucose utilization; regulation of fatty acid synthesis and oxidation; regulation of immune cell function; regulation of energy expenditure; and regulation of neural processes such as gastric motility. Through these many functions, bile acids regulate many aspects of digestion ranging from uptake of essential vitamins to proper utilization of nutrients. Accordingly, within a short time period, bile acids moved beyond simple detergents and into the realm of complex signaling molecules. Because of the important processes that bile acids regulate through activation of receptors, drugs that target these receptors are under development for the treatment of several diseases, including cholestatic liver disease and metabolic syndrome. In this review, we will describe the various bile acid receptors, the signal transduction pathways activated by these receptors, and briefly discuss the physiological processes that these receptors regulate.
Collapse
|
34
|
Guo GY, Shi YQ, Wang L, Ren X, Han ZY, Guo CC, Cui LN, Wang JB, Zhu J, Wang N, Zhang J, Cai Y, Han Y, Zhou XM, Fan DM. Serum vitamin D level is associated with disease severity and response to ursodeoxycholic acid in primary biliary cirrhosis. Aliment Pharmacol Ther 2015; 42:221-30. [PMID: 25982180 DOI: 10.1111/apt.13244] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2014] [Revised: 12/13/2014] [Accepted: 04/25/2015] [Indexed: 12/14/2022]
Abstract
BACKGROUND Serum vitamin D levels are associated with bone complications in patients with primary biliary cirrhosis (PBC). Increasing evidence suggests a nonskeletal role of vitamin D in various autoimmune and liver diseases. AIM To investigate the clinical relevance of vitamin D levels in PBC, especially their association with the therapeutic effects of ursodeoxycholic acid (UDCA). METHODS Consecutive PBC patients were retrospectively reviewed. 25-hydroxyvitamin D [25(OH)D] levels were determined in frozen serum samples collected before initiation of UDCA treatment. Response to UDCA was evaluated by Paris-I and Barcelona criteria. Logistic regressions were performed to identify the treatment response-associated parameters. RESULTS Among 98 patients, the mean serum 25(OH)D concentration was 17.9 ± 7.6 ng/mL. 25(OH)D levels decreased with increasing histological stage (P = 0.029) and were negatively correlated with bilirubin and alkaline phosphatase levels. After 1 year of UDCA therapy, 31 patients failed to achieve complete response according to Paris-I criteria. The baseline 25(OH)D level was significantly lower in nonresponders (14.8 ± 6.4 vs. 19.3 ± 7.6 ng/mL, P = 0.005). Vitamin D deficiency at baseline was associated with an increased risk of incomplete response independent of advanced stages (OR = 3.93, 95% CI = 1.02-15.19, P = 0.047). Similar results were obtained when biochemical response was evaluated by Barcelona criteria. Furthermore, 25(OH)D levels were lower in patients who subsequently suffered death or liver transplantation (12.1 ± 4.6 vs. 18.4 ± 7.6 ng/mL, P = 0.023). CONCLUSIONS 25(OH)D level is associated with biochemical and histological features in PBC. Pre-treatment vitamin D status is independently related to subsequent response to UDCA. Our results suggest that vitamin D status may have important clinical significance in PBC.
Collapse
Affiliation(s)
- G-Y Guo
- Division of Hepatology, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, China.,State Key Laboratory of Cancer Biology, Fourth Military Medical University, Xi'an, China
| | - Y-Q Shi
- Division of Hepatology, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, China.,State Key Laboratory of Cancer Biology, Fourth Military Medical University, Xi'an, China
| | - L Wang
- Division of Hepatology, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, China.,State Key Laboratory of Cancer Biology, Fourth Military Medical University, Xi'an, China
| | - X Ren
- Division of Hepatology, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, China.,State Key Laboratory of Cancer Biology, Fourth Military Medical University, Xi'an, China
| | - Z-Y Han
- Division of Hepatology, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, China.,State Key Laboratory of Cancer Biology, Fourth Military Medical University, Xi'an, China
| | - C-C Guo
- Division of Hepatology, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, China.,State Key Laboratory of Cancer Biology, Fourth Military Medical University, Xi'an, China
| | - L-N Cui
- Division of Hepatology, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, China
| | - J-B Wang
- Division of Hepatology, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, China
| | - J Zhu
- Division of Hepatology, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, China
| | - N Wang
- Division of Hepatology, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, China.,State Key Laboratory of Cancer Biology, Fourth Military Medical University, Xi'an, China
| | - J Zhang
- Division of Hepatology, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, China.,State Key Laboratory of Cancer Biology, Fourth Military Medical University, Xi'an, China
| | - Y Cai
- Division of Hepatology, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, China.,State Key Laboratory of Cancer Biology, Fourth Military Medical University, Xi'an, China
| | - Y Han
- Division of Hepatology, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, China.,State Key Laboratory of Cancer Biology, Fourth Military Medical University, Xi'an, China
| | - X-M Zhou
- Division of Hepatology, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, China.,State Key Laboratory of Cancer Biology, Fourth Military Medical University, Xi'an, China
| | - D-M Fan
- Division of Hepatology, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, China.,State Key Laboratory of Cancer Biology, Fourth Military Medical University, Xi'an, China
| |
Collapse
|
35
|
Zhang R, Zhao H, Dong H, Zou F, Cai S. 1α,25-dihydroxyvitamin D₃ counteracts the effects of cigarette smoke in airway epithelial cells. Cell Immunol 2015; 295:137-43. [PMID: 25880105 DOI: 10.1016/j.cellimm.2015.03.004] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2014] [Revised: 02/18/2015] [Accepted: 03/13/2015] [Indexed: 11/28/2022]
Abstract
Cigarette smoke extracts (CSE) alter calpain-1 expression via ERK signaling pathway in bronchial epithelial cells. 1α,25-dihydroxyvitamin D3 (1,25D3) inhibits cigarette smoke-induced epithelial barrier disruption. This study was aimed to explore whether the 1,25D3 counteracted the CSE effects in a human bronchial epithelial cell line (16HBE). In particular, transepithelial electrical resistance (TER) and permeability, expression and distribution of E-cadherin and β-catenin, calpain-1 expression, and ERK phosphorylation were assessed in the CSE-stimulated 16HBE cells. The CSE induced the ERK phosphorylation, improved the calpain-1 expression, increased the distribution anomalies and the cleaving of E-cadherin and β-catenin, and resulted in the TER reduction and the permeability increase. The 1,25D3 reduced these pathological changes. The 1,25D3 mediated effects were associated with a reduced ERK phosphorylation. In conclusion, the present study provides compelling evidences that the 1,25D3 may be considered a possible valid therapeutic option in controlling the cigarette smoke-induced epithelial barrier disruption.
Collapse
Affiliation(s)
- Ruhui Zhang
- Department of Respiratory, Chronic Airways Diseases Laboratory, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Haijin Zhao
- Department of Respiratory, Chronic Airways Diseases Laboratory, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Hangming Dong
- Department of Respiratory, Chronic Airways Diseases Laboratory, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Fei Zou
- School of Public Health and Tropical Medicine, Southern Medical University, Guangzhou 510515, China
| | - Shaoxi Cai
- Department of Respiratory, Chronic Airways Diseases Laboratory, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China.
| |
Collapse
|
36
|
Gonzalez-Sanchez E, Firrincieli D, Housset C, Chignard N. Nuclear receptors in acute and chronic cholestasis. Dig Dis 2015; 33:357-66. [PMID: 26045270 DOI: 10.1159/000371688] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
BACKGROUND Nuclear receptors (NRs) form a family of 48 members. NRs control hepatic processes such as bile acid homeostasis, lipid metabolism and mechanisms involved in fibrosis and inflammation. Due to their central role in the regulation of hepatoprotective mechanisms, NRs are promising therapeutic targets in cholestatic disorders. KEY MESSAGES NRs can be classified into five different physiological clusters. NRs from the 'bile acids and xenobiotic metabolism' and from the 'lipid metabolism and energy homeostasis' clusters are strongly expressed in the liver. Furthermore, NRs from these clusters, such as farnesoid X receptor α (FXRα), pregnane X receptor (PXR) and peroxisome proliferator-activated receptors (PPARs), have been associated with the pathogenesis and the progression of cholestasis. The latter observation is also true for vitamin D receptor (VDR), which is barely detectable in the whole liver, but has been linked to cholestatic diseases. Involvement of VDR in cholestasis is ascribed to a strong expression in nonparenchymal liver cells, such as biliary epithelial cells, Kupffer cells and hepatic stellate cells. Likewise, NRs from other physiological clusters with low hepatic expression, such as estrogen receptor α (ERα) or reverse-Erb α/β (REV-ERB α/β), may also control pathophysiological processes in cholestasis. CONCLUSIONS In this review, we will describe the impact of individual NRs on cholestasis. We will then discuss the potential role of these transcription factors as therapeutic targets.
Collapse
Affiliation(s)
- Ester Gonzalez-Sanchez
- INSERM UMR_S 938, Centre de Recherche Saint-Antoine, and Sorbonne Universités, UPMC Université Paris 06, Paris, France
| | | | | | | |
Collapse
|
37
|
Gonzalez-Sanchez E, Vaquero J, Fouassier L, Chignard N. E-cadherin, guardian of liver physiology. Clin Res Hepatol Gastroenterol 2015; 39:3-6. [PMID: 25459993 DOI: 10.1016/j.clinre.2014.09.008] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2014] [Revised: 09/26/2014] [Accepted: 09/30/2014] [Indexed: 02/04/2023]
Abstract
E-cadherin is a cell-to-cell adhesion molecule involved in epithelial cell behavior, tissue formation and cancer suppression. In the liver, E-cadherin is expressed by hepatocytes and biliary epithelial cells. However, the exact role of E-cadherin in hepatic pathophysiology remains largely unknown. Recently, specific loss of E-cadherin in liver epithelial cells has been shown to favor periportal fibrosis, periportal inflammation and liver cancer progression, suggesting that E-cadherin is a central liver protector.
Collapse
Affiliation(s)
- Ester Gonzalez-Sanchez
- Inserm UMR_S 938, centre de recherche Saint-Antoine, 75012 Paris, France; Sorbonne universités, UPMC, université Paris 06, 75012 Paris, France
| | - Javier Vaquero
- Inserm UMR_S 938, centre de recherche Saint-Antoine, 75012 Paris, France; Sorbonne universités, UPMC, université Paris 06, 75012 Paris, France
| | - Laura Fouassier
- Inserm UMR_S 938, centre de recherche Saint-Antoine, 75012 Paris, France; Sorbonne universités, UPMC, université Paris 06, 75012 Paris, France
| | - Nicolas Chignard
- Inserm UMR_S 938, centre de recherche Saint-Antoine, 75012 Paris, France; Sorbonne universités, UPMC, université Paris 06, 75012 Paris, France.
| |
Collapse
|
38
|
Regnault C, Worms IAM, Oger-Desfeux C, MelodeLima C, Veyrenc S, Bayle ML, Combourieu B, Bonin A, Renaud J, Raveton M, Reynaud S. Impaired liver function in Xenopus tropicalis exposed to benzo[a]pyrene: transcriptomic and metabolic evidence. BMC Genomics 2014; 15:666. [PMID: 25103525 PMCID: PMC4141109 DOI: 10.1186/1471-2164-15-666] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2014] [Accepted: 07/30/2014] [Indexed: 12/20/2022] Open
Abstract
Background Despite numerous studies suggesting that amphibians are highly sensitive to cumulative anthropogenic stresses, the role pollutants play in the decline of amphibian populations remains unclear. Amongst the most common aquatic contaminants, polycyclic aromatic hydrocarbons (PAHs) have been shown to induce several adverse effects on amphibian species in the larval stages. Conversely, adults exposed to high concentrations of the ubiquitous PAH, benzo[a]pyrene (BaP), tolerate the compound thanks to their highly efficient hepatic detoxification mechanisms. Due to this apparent lack of toxic effect on adults, no studies have examined in depth the potential toxicological impact of PAH on the physiology of adult amphibian livers. This study sheds light on the hepatic responses of Xenopus tropicalis when exposed to high environmentally relevant concentrations of BaP, by combining a high throughput transcriptomic approach (mRNA deep sequencing) and a characterization of cellular and physiological modifications to the amphibian liver. Results Transcriptomic changes observed in BaP-exposed Xenopus were further characterized using a time-dependent enrichment analysis, which revealed the pollutant-dependent gene regulation of important biochemical pathways, such as cholesterol biosynthesis, insulin signaling, adipocytokines signaling, glycolysis/gluconeogenesis and MAPK signaling. These results were substantiated at the physiological level with the detection of a pronounced metabolic disorder resulting in a possible insulin resistance-like syndrome phenotype. Hepatotoxicity induced by lipid and cholesterol metabolism impairments was clearly identified in BaP-exposed individuals. Conclusions Our data suggested that BaP may disrupt overall liver physiology, and carbohydrate and cholesterol metabolism in particular, even after short-term exposure. These results are further discussed in terms of how this deregulation of liver physiology can lead to general metabolic impairment in amphibians chronically exposed to contaminants, thereby illustrating the role xenobiotics might play in the global decline in amphibian populations. Electronic supplementary material The online version of this article (doi:10.1186/1471-2164-15-666) contains supplementary material, which is available to authorized users.
Collapse
|
39
|
Dianat N, Dubois-Pot-Schneider H, Steichen C, Desterke C, Leclerc P, Raveux A, Combettes L, Weber A, Corlu A, Dubart-Kupperschmitt A. Generation of functional cholangiocyte-like cells from human pluripotent stem cells and HepaRG cells. Hepatology 2014; 60:700-14. [PMID: 24715669 PMCID: PMC4315871 DOI: 10.1002/hep.27165] [Citation(s) in RCA: 156] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2013] [Accepted: 04/07/2014] [Indexed: 12/11/2022]
Abstract
UNLABELLED Cholangiocytes are biliary epithelial cells, which, like hepatocytes, originate from hepatoblasts during embryonic development. In this study we investigated the potential of human embryonic stem cells (hESCs) to differentiate into cholangiocytes and we report a new approach, which drives differentiation of hESCs toward the cholangiocytic lineage using feeder-free and defined culture conditions. After differentiation into hepatic progenitors, hESCs were differentiated further into cholangiocytes using growth hormone, epidermal growth factor, interleukin-6, and then sodium taurocholate. These conditions also allowed us to generate cholangiocytes from HepaRG-derived hepatoblasts. hESC- and HepaRG-derived cholangiocyte-like cells expressed markers of cholangiocytes including cytokeratin 7 and osteopontin, and the transcription factors SOX9 and hepatocyte nuclear factor 6. The cells also displayed specific proteins important for cholangiocyte functions including cystic fibrosis transmembrane conductance regulator, secretin receptor, and nuclear receptors. They formed primary cilia and also responded to hormonal stimulation by increase of intracellular Ca(2+) . We demonstrated by integrative genomics that the expression of genes, which signed hESC- or HepaRG-cholangiocytes, separates hepatocytic lineage from cholangiocyte lineage. When grown in a 3D matrix, cholangiocytes developed epithelial/apicobasal polarity and formed functional cysts and biliary ducts. In addition, we showed that cholangiocyte-like cells could also be generated from human induced pluripotent stem cells, demonstrating the efficacy of our approach with stem/progenitor cells of diverse origins. CONCLUSION We have developed a robust and efficient method for differentiating pluripotent stem cells into cholangiocyte-like cells, which display structural and functional similarities to bile duct cells in normal liver. These cells will be useful for the in vitro study of the molecular mechanisms of bile duct development and have important potential for therapeutic strategies, including bioengineered liver approaches.
Collapse
Affiliation(s)
- Noushin Dianat
- INSERM, U972, Paul Brousse HospitalVillejuif, France,Université Paris Sud, UMR-S 972Villejuif, France,IFR 93, Bicêtre HospitalKremlin-Bicêtre, France,DHU Hepatinov, Paul Brousse HospitalVillejuif, France
| | | | - Clara Steichen
- INSERM, U972, Paul Brousse HospitalVillejuif, France,Université Paris Sud, UMR-S 972Villejuif, France,IFR 93, Bicêtre HospitalKremlin-Bicêtre, France,DHU Hepatinov, Paul Brousse HospitalVillejuif, France
| | - Christophe Desterke
- INSERM, U972, Paul Brousse HospitalVillejuif, France,Université Paris Sud, UMR-S 972Villejuif, France,DHU Hepatinov, Paul Brousse HospitalVillejuif, France
| | | | - Aurélien Raveux
- INSERM, U972, Paul Brousse HospitalVillejuif, France,Université Paris Sud, UMR-S 972Villejuif, France,IFR 93, Bicêtre HospitalKremlin-Bicêtre, France
| | - Laurent Combettes
- DHU Hepatinov, Paul Brousse HospitalVillejuif, France,INSERM UMR-S 757UPS-Orsay, Orsay, France
| | - Anne Weber
- INSERM, U972, Paul Brousse HospitalVillejuif, France,Université Paris Sud, UMR-S 972Villejuif, France,IFR 93, Bicêtre HospitalKremlin-Bicêtre, France,DHU Hepatinov, Paul Brousse HospitalVillejuif, France
| | - Anne Corlu
- INSERM, UMR-S 991, Pontchaillou HospitalRennes, France,University of Rennes 1Rennes, France,
Address reprint requests to: Anne Corlu, Ph.D., INSERM, UMR-S 991, Pontchaillou Hospital, Rennes F-35033, France. E-mail: ; or Anne Dubart-Kupperschmitt, M.D., INSERM, U972, Paul Brousse Hospital, Villejuif, F-94807, France. ; fax: +33 (0)1 47 26 03 19, +33 (0)2 99 54 01 37
| | - Anne Dubart-Kupperschmitt
- INSERM, U972, Paul Brousse HospitalVillejuif, France,Université Paris Sud, UMR-S 972Villejuif, France,IFR 93, Bicêtre HospitalKremlin-Bicêtre, France,DHU Hepatinov, Paul Brousse HospitalVillejuif, France
| |
Collapse
|
40
|
Gautherot J, Delautier D, Maubert MA, Aït-Slimane T, Bolbach G, Delaunay JL, Durand-Schneider AM, Firrincieli D, Barbu V, Chignard N, Housset C, Maurice M, Falguières T. Phosphorylation of ABCB4 impacts its function: insights from disease-causing mutations. Hepatology 2014; 60:610-21. [PMID: 24723470 DOI: 10.1002/hep.27170] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2013] [Revised: 04/07/2014] [Accepted: 04/08/2014] [Indexed: 12/18/2022]
Abstract
UNLABELLED The ABCB4 transporter mediates phosphatidylcholine (PC) secretion at the canalicular membrane of hepatocytes and its genetic defects cause biliary diseases. Whereas ABCB4 shares high sequence identity with the multidrug transporter, ABCB1, its N-terminal domain is poorly conserved, leading us to hypothesize a functional specificity of this domain. A database of ABCB4 genotyping in a large series of patients was screened for variations altering residues of the N-terminal domain. Identified variants were then expressed in cell models to investigate their biological consequences. Two missense variations, T34M and R47G, were identified in patients with low-phospholipid-associated cholelithiasis or intrahepatic cholestasis of pregnancy. The T34M and R47G mutated proteins showed no or minor defect, respectively, in maturation and targeting to the apical membrane, in polarized Madin-Darby Canine Kidney and HepG2 cells, whereas their stability was similar to that of wild-type (WT) ABCB4. By contrast, the PC secretion activity of both mutants was markedly decreased. In silico analysis indicated that the identified variants were likely to affect ABCB4 phosphorylation. Mass spectrometry analyses confirmed that the N-terminal domain of WT ABCB4 could undergo phosphorylation in vitro and revealed that the T34M and R47G mutations impaired such phosphorylation. ABCB4-mediated PC secretion was also increased by pharmacological activation of protein kinases A or C and decreased by inhibition of these kinases. Furthermore, secretion activity of the T34M and R47G mutants was less responsive than that of WT ABCB4 to protein kinase modulators. CONCLUSION We identified disease-associated variants of ABCB4 involved in the phosphorylation of its N-terminal domain and leading to decreased PC secretion. Our results also indicate that ABCB4 activity is regulated by phosphorylation, in particular, of N-terminal residues.
Collapse
Affiliation(s)
- Julien Gautherot
- INSERM, UMR_S 938, CDR Saint-Antoine, F-75012, Paris, France; Sorbonne Universités, UPMC Université Paris 06, UMR_S 938 and Institute of Cardiometabolism and Nutrition (ICAN), F-75005, Paris, France
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Hochrath K, Stokes CS, Geisel J, Pollheimer MJ, Fickert P, Dooley S, Lammert F. Vitamin D modulates biliary fibrosis in ABCB4-deficient mice. Hepatol Int 2014; 8:443-52. [PMID: 25191532 PMCID: PMC4148166 DOI: 10.1007/s12072-014-9548-2] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2014] [Accepted: 05/18/2014] [Indexed: 12/31/2022]
Abstract
Purpose Impaired vitamin D receptor signaling represents an aggravating factor during liver injury, and recent studies suggest that vitamin D might exert a protective role in chronic hepatobiliary diseases. We hypothesized that vitamin D supplementation would ameliorate liver fibrosis in ATP-binding cassette transporter B4 knockout (Abcb4−/−) mice as a preclinical model of sclerosing cholangitis. Methods Abcb4−/− and wild-type mice were fed a regular chow diet (600 IU vitamin D/kg food) or diets with lower (100 IU/kg) and higher (2,400 IU/kg) vitamin D concentrations for 12 weeks. Serum 25-hydroxyvitamin D concentrations were measured by chemiluminescence immunoassays. Liver injury and biliary fibrosis were assessed by liver enzyme activities, histopathology and hepatic collagen contents. Hepatic mRNA expression of markers for fibrosis, vitamin D and bile acid metabolism were analyzed by quantitative PCR. Results Different vitamin D concentrations were observed depending on genotype and diet group, with Abcb4−/− mice on the control diet showing lower vitamin D concentrations compared to wild-type mice. Abcb4−/− animals on the low vitamin D diet demonstrated the most advanced liver fibrosis and highest hepatic collagen contents. Feeding Abcb4−/− mice a high vitamin D diet enriched serum vitamin D levels, lowered liver enzyme activities, altered expression levels of profibrogenic genes and ameliorated, in part, liver injury. Conclusions This is the first report to demonstrate that fibrogenesis in the established Abcb4−/− model is influenced by vitamin D supplementation. Since vitamin D modulates sclerosing cholangitis in vivo, we speculate that sufficient vitamin D intake might improve liver damage and induce antifibrotic effects in chronic cholestasis in humans. Electronic supplementary material The online version of this article (doi:10.1007/s12072-014-9548-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Katrin Hochrath
- Department of Medicine II, Saarland University Medical Center, Kirrberger Str. 100, 66421 Homburg, Germany
| | - Caroline S. Stokes
- Department of Medicine II, Saarland University Medical Center, Kirrberger Str. 100, 66421 Homburg, Germany
| | - Jürgen Geisel
- Institute of Clinical Chemistry and Laboratory Medicine, Saarland University Medical Center, Homburg, Germany
| | | | - Peter Fickert
- Insititute of Pathology, Medical University Graz, Graz, Austria
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Steven Dooley
- Division of Molecular Hepatology-Alcohol Associated Diseases, Department of Medicine II, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Frank Lammert
- Department of Medicine II, Saarland University Medical Center, Kirrberger Str. 100, 66421 Homburg, Germany
| |
Collapse
|
42
|
Firrincieli D, Braescu T, Housset C, Chignard N. Illuminating liver fibrosis with vitamin D. Clin Res Hepatol Gastroenterol 2014; 38:5-8. [PMID: 24238723 DOI: 10.1016/j.clinre.2013.10.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2013] [Revised: 09/27/2013] [Accepted: 10/10/2013] [Indexed: 02/04/2023]
Abstract
Hepatic fibrosis results from the accumulation of extracellular matrix-producing myofibroblasts in the liver. The mechanisms leading to the activation of hepatic stellate cells (HSCs) into myofibroblasts have been well described. By contrast, few molecular pathways leading to myofibroblast deactivation have been documented. Recently, the vitamin D-VDR axis has been shown to modulate HSC activity through a complex mechanism involving epigenetic modifications induced by the SMAD pathway.
Collapse
Affiliation(s)
- D Firrincieli
- Inserm UMR_S 938, CdR Saint-Antoine, 75012 Paris, France; UPMC Univ Paris 06, 75012 Paris, France
| | - T Braescu
- Inserm UMR_S 938, CdR Saint-Antoine, 75012 Paris, France; UPMC Univ Paris 06, 75012 Paris, France
| | - C Housset
- Inserm UMR_S 938, CdR Saint-Antoine, 75012 Paris, France; UPMC Univ Paris 06, 75012 Paris, France; AP-HP, hôpital Saint-Antoine, service d'hépatologie, 75012 Paris, France
| | - N Chignard
- Inserm UMR_S 938, CdR Saint-Antoine, 75012 Paris, France; UPMC Univ Paris 06, 75012 Paris, France.
| |
Collapse
|