1
|
Cai H, Shen J, Peng W, Zhang X, Wen T. Identification of SOX9-related prognostic DEGs and a prediction model for hepatitis C-induced early-stage fibrosis. Gene 2025; 937:149133. [PMID: 39622395 DOI: 10.1016/j.gene.2024.149133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 11/24/2024] [Accepted: 11/27/2024] [Indexed: 12/06/2024]
Abstract
BACKGROUND Hepatitis C virus (HCV) infection induces liver inflammation, activating hepatic stellate cells (HSC) and advancing fibrosis. Studies have indicated that SOX9 overexpression is closely linked to HSC activation. The study aims to identify genes associated with SOX9 and search for potential targets for detecting and treating liver fibrosis. METHOD The dataset GSE15654, containing 216 biopsy samples from HCV-induced early-stage cirrhosis patients, was obtained from the GEO database. Prognostic genes were identified through differential gene analysis, LASSO, and Cox regression analyses. CIBERSORT analysis quantified infiltration levels across 22 immune cell types. Constructing a prognostic prediction model using screened genes and conducting preliminary validation using qRT PCR and RNA sequencing techniques. RESULTS Elevated SOX9 expression correlates with unfavorable outcomes in patients with early-stage liver fibrosis induced by HCV. We identified nine SOX9-related prognostic DEGs in our study. ADAMTS2, ARHGEF5, CCT8, ERG, LBH, FRMD6, INMT, and RASGRF2 were considered risk factors in the disease progression, while DHRS4 was considered a protective factor. SOX9 expression showed a positive correlation with mast cell infiltration, whereas ARHGEF5 and FRMD6 expressions were positively associated with M0 macrophage infiltration. Our combined model surpasses the commonly used APRI and FIB4 indicators in predicting patient prognosis. The testing of clinical samples also preliminarily validated our research results. CONCLUSION The prognostic model based on nine SOX9-related DEGs provides an effective tool for forecasting the progression and outcomes of liver fibrosis. This study introduces a new strategy for advancing liver fibrosis prediction and treatment.
Collapse
Affiliation(s)
- Haozheng Cai
- Division of Liver Surgery, Department of General Surgery, West China Hospital, Si Chuan University, Chengdu, China
| | - Junyi Shen
- Division of Liver Surgery, Department of General Surgery, West China Hospital, Si Chuan University, Chengdu, China
| | - Wei Peng
- Division of Liver Surgery, Department of General Surgery, West China Hospital, Si Chuan University, Chengdu, China
| | - Xiaoyun Zhang
- Division of Liver Surgery, Department of General Surgery, West China Hospital, Si Chuan University, Chengdu, China
| | - Tianfu Wen
- Division of Liver Surgery, Department of General Surgery, West China Hospital, Si Chuan University, Chengdu, China.
| |
Collapse
|
2
|
Tsuchiya J, Miyoshi M, Kakinuma S, Kawai-Kitahata F, Kamiya A, Shimizu T, Sato A, Watakabe K, Mochida T, Inada K, Kamimae R, Kaneko S, Murakawa M, Nitta S, Nakagawa M, Watanabe M, Asahina Y, Okamoto R. Hepatitis B Virus-KMT2B Integration Drives Hepatic Oncogenic Processes in a Human Gene-edited Induced Pluripotent Stem Cells-derived Model. Cell Mol Gastroenterol Hepatol 2024; 19:101422. [PMID: 39419394 PMCID: PMC11664014 DOI: 10.1016/j.jcmgh.2024.101422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 10/07/2024] [Accepted: 10/08/2024] [Indexed: 10/19/2024]
Abstract
BACKGROUND & AIMS Hepatitis B virus (HBV)-DNA integration into the host genome contributes to hepatocellular carcinoma (HCC) development. KMT2B is the second most frequent locus of HBV-DNA integration in HCC; however, its role and function remain unclear. We aimed to clarify the impact of HBV-KMT2B integration in HCC development using a human genome-edited induced pluripotent stem cell (iPSCs) model. METHODS Based on the genetic information on HBV-KMT2B integration in HCC, we determined its complete DNA sequence and transcript variants. To exclude the effect of other oncogenic mutations, we reproduced HBV integration in healthy donor iPSCs with an intact genome and analyzed its effects using iPSC-derived hepatic progenitor cells (HPCs) and hepatocytes (iPS-Heps). RESULTS The reproduced HBV-KMT2B integration significantly upregulated the proliferation of hepatic cells. Comprehensive transcriptional and epigenetic analyses revealed enhanced expression of cell cycle-related genes in hepatic cells with HBV-KMT2B integration based on perturbation of histone 3 lysine 4 tri-methylation (H3K4me3), mimicking that in the original HCC sample. Long-read RNA-sequence detected the common KMT2B transcript variants in the HCC sample and HPCs. Overexpression of the truncated variant significantly enhanced proliferation of hepatic cells, whereas HBV-KMT2B fusion transcripts did not enhance proliferation. HBV-KMT2B-integrated HPCs exhibited replication stress and DNA damage, indicating that our model initiated the process of hepatocarcinogenesis due to abnormally promoted KMT2B function. CONCLUSIONS Our disease model using genetically engineered iPSCs provides the first insight into both the KMT2B function in HCC development and the oncogenic processes by HBV-KMT2B integration. We clarified the novel oncogenic mechanism in HBV-related HCC due to aberrant KMT2B function.
Collapse
Affiliation(s)
- Jun Tsuchiya
- Department of Gastroenterology and Hepatology, Institute of Science Tokyo (Science Tokyo), Tokyo, Japan
| | - Masato Miyoshi
- Department of Gastroenterology and Hepatology, Institute of Science Tokyo (Science Tokyo), Tokyo, Japan
| | - Sei Kakinuma
- Department of Gastroenterology and Hepatology, Institute of Science Tokyo (Science Tokyo), Tokyo, Japan; Department of Clinical and Diagnostic Laboratory Science, Institute of Science Tokyo (Science Tokyo), Tokyo, Japan.
| | - Fukiko Kawai-Kitahata
- Department of Gastroenterology and Hepatology, Institute of Science Tokyo (Science Tokyo), Tokyo, Japan
| | - Akihide Kamiya
- Department of Molecular Life Sciences, School of Medicine, Tokai University, Kanagawa, Japan
| | - Taro Shimizu
- Department of Gastroenterology and Hepatology, Institute of Science Tokyo (Science Tokyo), Tokyo, Japan
| | - Ayako Sato
- Department of Gastroenterology and Hepatology, Institute of Science Tokyo (Science Tokyo), Tokyo, Japan
| | - Keiya Watakabe
- Department of Gastroenterology and Hepatology, Institute of Science Tokyo (Science Tokyo), Tokyo, Japan
| | - Tomohiro Mochida
- Department of Gastroenterology and Hepatology, Institute of Science Tokyo (Science Tokyo), Tokyo, Japan
| | - Kento Inada
- Department of Gastroenterology and Hepatology, Institute of Science Tokyo (Science Tokyo), Tokyo, Japan
| | - Rion Kamimae
- Department of Clinical and Diagnostic Laboratory Science, Institute of Science Tokyo (Science Tokyo), Tokyo, Japan
| | - Shun Kaneko
- Department of Gastroenterology and Hepatology, Institute of Science Tokyo (Science Tokyo), Tokyo, Japan
| | - Miyako Murakawa
- Department of Gastroenterology and Hepatology, Institute of Science Tokyo (Science Tokyo), Tokyo, Japan
| | - Sayuri Nitta
- Department of Gastroenterology and Hepatology, Institute of Science Tokyo (Science Tokyo), Tokyo, Japan
| | - Mina Nakagawa
- Department of Gastroenterology and Hepatology, Institute of Science Tokyo (Science Tokyo), Tokyo, Japan
| | - Mamoru Watanabe
- Department of Gastroenterology and Hepatology, Institute of Science Tokyo (Science Tokyo), Tokyo, Japan; Faculty of Medicine, Juntendo University, Tokyo, Japan
| | - Yasuhiro Asahina
- Department of Gastroenterology and Hepatology, Institute of Science Tokyo (Science Tokyo), Tokyo, Japan; Division of Hepatic Medical Science, Institute of Science Tokyo (Science Tokyo), Tokyo, Japan.
| | - Ryuichi Okamoto
- Department of Gastroenterology and Hepatology, Institute of Science Tokyo (Science Tokyo), Tokyo, Japan.
| |
Collapse
|
3
|
Fujisawa H, Ota N, Shiojiri N. Inversin-deficient (inv) mice do not establish a polarized duct system in the liver and pancreas. Anat Rec (Hoboken) 2024; 307:2197-2212. [PMID: 37921502 DOI: 10.1002/ar.25346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2023] [Revised: 10/16/2023] [Accepted: 10/16/2023] [Indexed: 11/04/2023]
Abstract
Inversin-deficient (inv) mice have anomalies in liver and pancreatic development in addition to an inverted left-right axis of the body. The present study was undertaken to unveil mechanisms of bile and pancreatic duct development from immunohistochemical analyses of anomalies in inv mice. Intrahepatic bile ducts having proximodistal polarity in size and the height of their epithelia, and ductules were formed in livers of wild-type neonates. By contrast, in inv mice, ductal plates, precursor structures of intrahepatic bile ducts and ductules, persisted without the proximodistal polarity. Their epithelial cells did not acquire planar cell polarity (PCP) in terms of expression of tight junction proteins although they expressed bile duct markers, HNF1β and SOX9. They had an apicobasal polarity from expression of basal laminar components. Enlargement of the hepatic artery and poor connective tissue development, including the abnormal deposition of the extracellular matrices, were also noted in inv mice, suggesting that bile duct development was coupled to that of the hepatic artery and portal vein. In pancreata of inv neonates, neither the main pancreatic duct was formed, nor dilated duct-like structures had the morphological polarity from the connecting point with the common bile duct. Lumina of acini was dilated, and centroacinar cells changed their position in the acini to their neck region. Immunohistochemical analyses of tight junction proteins suggested that epithelial cells of the duct-like structures did not have a PCP. Thus, Invs may be required for the establishment of the PCP of the whole duct system in the liver and pancreas.
Collapse
Affiliation(s)
- Hiromu Fujisawa
- Department of Biology, Faculty of Science, Shizuoka University, Shizuoka, Japan
| | - Noriaki Ota
- Department of Biology, Faculty of Science, Shizuoka University, Shizuoka, Japan
| | - Nobuyoshi Shiojiri
- Department of Biology, Faculty of Science, Shizuoka University, Shizuoka, Japan
| |
Collapse
|
4
|
Long noncoding RNA SNHG1 silencing accelerates hepatocyte-like cell differentiation of bone marrow-derived mesenchymal stem cells to alleviate cirrhosis via the microRNA-15a/SMURF1/UVRAG axis. Cell Death Dis 2022; 8:77. [PMID: 35194023 PMCID: PMC8863836 DOI: 10.1038/s41420-022-00850-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2021] [Revised: 01/11/2022] [Accepted: 01/24/2022] [Indexed: 02/08/2023]
Abstract
Bone marrow-derived mesenchymal stem cells (BMSCs) can differentiate into hepatocyte-like cells (HLCs) to attenuate cirrhosis. Long noncoding RNA (lncRNA) SNHG1 has been demonstrated to orchestrate BMSC differentiation, whereas its role in cirrhosis remains elusive. Therefore, this study was performed to figure out whether lncRNA SNHG1 was involved in cirrhosis by affecting HLC differentiation of BMSCs. Mouse BMSCs were isolated, and the BMSC differentiation into HLCs was induced by hepatocyte growth factor (HGF). A cirrhotic mouse model was established using carbon tetrachloride and phenobarbital, followed by intravenous injection of BMSCs with manipulated expression of lncRNA SNHG1, microRNA (miR)-15a, and SMURF1. Subsequent to HGF induction, expression of hepatocyte-related genes, albumin secretion, and glycogen accumulation was increased in BMSCs, suggesting the differentiation of BMSCs into HLCs. Mechanistically, lncRNA SNHG1 bound to miR-15a that targeted SMURF1, and SMURF1 diminished ATG5 and Wnt5a expression by enhancing the ubiquitination of UVRAG. LncRNA SNHG1 or SMURF1 silencing or miR-15a overexpression promoted differentiation of BMSCs into HLCs and repressed cirrhosis of mice by upregulating ATG5 and Wnt5a via UVRAG. Conclusively, lncRNA SNHG1 silencing might facilitate HLC differentiation from mouse BMSCs and alleviate cirrhosis via the miR-15a/SMURF1/UVRAG/ATG5/Wnt5a axis.
Collapse
|
5
|
Airik M, Schüler M, McCourt B, Weiss AC, Herdman N, Lüdtke TH, Widmeier E, Stolz DB, Nejak-Bowen KN, Yimlamai D, Wu YL, Kispert A, Airik R, Hildebrandt F. Loss of Anks6 leads to YAP deficiency and liver abnormalities. Hum Mol Genet 2021; 29:3064-3080. [PMID: 32886109 PMCID: PMC7733532 DOI: 10.1093/hmg/ddaa197] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2020] [Revised: 07/03/2020] [Accepted: 08/27/2020] [Indexed: 12/17/2022] Open
Abstract
ANKS6 is a ciliary protein that localizes to the proximal compartment of the primary cilium, where it regulates signaling. Mutations in the ANKS6 gene cause multiorgan ciliopathies in humans, which include laterality defects of the visceral organs, renal cysts as part of nephronophthisis and congenital hepatic fibrosis (CHF) in the liver. Although CHF together with liver ductal plate malformations are common features of several human ciliopathy syndromes, including nephronophthisis-related ciliopathies, the mechanism by which mutations in ciliary genes lead to bile duct developmental abnormalities is not understood. Here, we generated a knockout mouse model of Anks6 and show that ANKS6 function is required for bile duct morphogenesis and cholangiocyte differentiation. The loss of Anks6 causes ciliary abnormalities, ductal plate remodeling defects and periportal fibrosis in the liver. Our expression studies and biochemical analyses show that biliary abnormalities in Anks6-deficient livers result from the dysregulation of YAP transcriptional activity in the bile duct-lining epithelial cells. Mechanistically, our studies suggest, that ANKS6 antagonizes Hippo signaling in the liver during bile duct development by binding to Hippo pathway effector proteins YAP1, TAZ and TEAD4 and promoting their transcriptional activity. Together, this study reveals a novel function for ANKS6 in regulating Hippo signaling during organogenesis and provides mechanistic insights into the regulatory network controlling bile duct differentiation and morphogenesis during liver development.
Collapse
Affiliation(s)
- Merlin Airik
- Division of Nephrology, Department of Pediatrics, University of Pittsburgh, Pittsburgh, PA, USA
| | - Markus Schüler
- Division of Nephrology and Internal Intensive Care Medicine, Charite University, Berlin, Germany.,Division of Nephrology, Boston Children's Hospital, Boston, MA, USA
| | - Blake McCourt
- Division of Nephrology, Department of Pediatrics, University of Pittsburgh, Pittsburgh, PA, USA
| | | | - Nathan Herdman
- Division of Nephrology, Department of Pediatrics, University of Pittsburgh, Pittsburgh, PA, USA
| | | | - Eugen Widmeier
- Division of Nephrology, Boston Children's Hospital, Boston, MA, USA.,Department of Medicine, Renal Division, Medical Center - University of Freiburg, Freiburg, Germany
| | - Donna B Stolz
- Department of Cell Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Kari N Nejak-Bowen
- Division of Experimental Pathology, Department of Pathology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Dean Yimlamai
- Division of Gastroenterology and Nutrition, Department of Pediatrics, University of Pittsburgh, Pittsburgh, PA, USA
| | - Yijen L Wu
- Department of Developmental Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | | | - Rannar Airik
- Division of Nephrology, Department of Pediatrics, University of Pittsburgh, Pittsburgh, PA, USA.,Department of Developmental Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | | |
Collapse
|
6
|
Sato A, Kakinuma S, Miyoshi M, Kamiya A, Tsunoda T, Kaneko S, Tsuchiya J, Shimizu T, Takeichi E, Nitta S, Kawai-Kitahata F, Murakawa M, Itsui Y, Nakagawa M, Azuma S, Koshikawa N, Seiki M, Nakauchi H, Asahina Y, Watanabe M. Vasoactive Intestinal Peptide Derived From Liver Mesenchymal Cells Mediates Tight Junction Assembly in Mouse Intrahepatic Bile Ducts. Hepatol Commun 2019; 4:235-254. [PMID: 32025608 PMCID: PMC6996346 DOI: 10.1002/hep4.1459] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Accepted: 11/23/2019] [Indexed: 01/20/2023] Open
Abstract
Formation of intrahepatic bile ducts (IHBDs) proceeds in accordance with their microenvironment. Particularly, mesenchymal cells around portal veins regulate the differentiation and ductular morphogenesis of cholangiocytes in the developing liver; however, further studies are needed to fully understand the arrangement of IHBDs into a continuous hierarchical network. This study aims to clarify the interaction between biliary and liver mesenchymal cells during IHBD formation. To identify candidate factors contributing to this cell–cell interaction, mesenchymal cells were isolated from embryonic day 16.5 matrix metalloproteinase 14 (MMP14)‐deficient (knockout [KO]) mice livers, in which IHBD formation is retarded, and compared with those of the wild type (WT). WT mesenchymal cells significantly facilitated the formation of luminal structures comprised of hepatoblast‐derived cholangiocytes (cholangiocytic cysts), whereas MMP14‐KO mesenchymal cells failed to promote cyst formation. Comprehensive analysis revealed that expression of vasoactive intestinal peptide (VIP) was significantly suppressed in MMP14‐KO mesenchymal cells. VIP and VIP receptor 1 (VIPR1) were mainly expressed in periportal mesenchymal cells and cholangiocytic progenitors during IHBD development, respectively, in vivo. VIP/VIPR1 signaling significantly encouraged cholangiocytic cyst formation and up‐regulated tight junction protein 1, cystic fibrosis transmembrane conductance regulator, and aquaporin 1, in vitro. VIP antagonist significantly suppressed the tight junction assembly and the up‐regulation of ion/water transporters during IHBD development in vivo. In a cholestatic injury model of adult mice, exogenous VIP administration promoted the restoration of damaged tight junctions in bile ducts and improved hyperbilirubinemia. Conclusion: VIP is produced by periportal mesenchymal cells during the perinatal stage. It supports bile duct development by establishing tight junctions and up‐regulating ion/water transporters in cholangiocytes. VIP contributes to prompt recovery from cholestatic damage through the establishment of tight junctions in the bile ducts.
Collapse
Affiliation(s)
- Ayako Sato
- Department of Gastroenterology and Hepatology Tokyo Medical and Dental University Tokyo Japan
| | - Sei Kakinuma
- Department of Gastroenterology and Hepatology Tokyo Medical and Dental University Tokyo Japan.,Department of Liver Disease Control Tokyo Medical and Dental University (TMDU) Tokyo Japan
| | - Masato Miyoshi
- Department of Gastroenterology and Hepatology Tokyo Medical and Dental University Tokyo Japan
| | - Akihide Kamiya
- Department of Molecular Life Sciences School of Medicine Tokai University Isehara Japan
| | - Tomoyuki Tsunoda
- Department of Gastroenterology and Hepatology Tokyo Medical and Dental University Tokyo Japan
| | - Shun Kaneko
- Department of Gastroenterology and Hepatology Tokyo Medical and Dental University Tokyo Japan
| | - Jun Tsuchiya
- Department of Gastroenterology and Hepatology Tokyo Medical and Dental University Tokyo Japan
| | - Taro Shimizu
- Department of Gastroenterology and Hepatology Tokyo Medical and Dental University Tokyo Japan
| | - Eiko Takeichi
- Department of Gastroenterology and Hepatology Tokyo Medical and Dental University Tokyo Japan
| | - Sayuri Nitta
- Department of Gastroenterology and Hepatology Tokyo Medical and Dental University Tokyo Japan
| | - Fukiko Kawai-Kitahata
- Department of Gastroenterology and Hepatology Tokyo Medical and Dental University Tokyo Japan
| | - Miyako Murakawa
- Department of Gastroenterology and Hepatology Tokyo Medical and Dental University Tokyo Japan
| | - Yasuhiro Itsui
- Department of Gastroenterology and Hepatology Tokyo Medical and Dental University Tokyo Japan
| | - Mina Nakagawa
- Department of Gastroenterology and Hepatology Tokyo Medical and Dental University Tokyo Japan
| | - Seishin Azuma
- Department of Gastroenterology and Hepatology Tokyo Medical and Dental University Tokyo Japan
| | - Naohiko Koshikawa
- Division of Cancer Cell Research Institute of Medical Science University of Tokyo Tokyo Japan
| | - Motoharu Seiki
- Division of Cancer Cell Research Institute of Medical Science University of Tokyo Tokyo Japan
| | - Hiromitsu Nakauchi
- Institute for Stem Cell Biology and Regenerative Medicine Stanford University School of Medicine Stanford CA.,Division of Stem Cell Therapy Institute of Medical Science University of Tokyo Tokyo Japan
| | - Yasuhiro Asahina
- Department of Gastroenterology and Hepatology Tokyo Medical and Dental University Tokyo Japan.,Department of Liver Disease Control Tokyo Medical and Dental University (TMDU) Tokyo Japan
| | - Mamoru Watanabe
- Department of Gastroenterology and Hepatology Tokyo Medical and Dental University Tokyo Japan.,Advanced Research Institute Tokyo Medical and Dental University (TMDU) Tokyo Japan
| |
Collapse
|
7
|
Lemaigre FP. Development of the Intrahepatic and Extrahepatic Biliary Tract: A Framework for Understanding Congenital Diseases. ANNUAL REVIEW OF PATHOLOGY-MECHANISMS OF DISEASE 2019; 15:1-22. [PMID: 31299162 DOI: 10.1146/annurev-pathmechdis-012418-013013] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The involvement of the biliary tract in the pathophysiology of liver diseases and the increased attention paid to bile ducts in the bioconstruction of liver tissue for regenerative therapy have fueled intense research into the fundamental mechanisms of biliary development. Here, I review the molecular, cellular and tissular mechanisms driving differentiation and morphogenesis of the intrahepatic and extrahepatic bile ducts. This review focuses on the dynamics of the transcriptional and signaling modules that promote biliary development in human and mouse liver and discusses studies in which the use of zebrafish uncovered unexplored processes in mammalian biliary development. The review concludes by providing a framework for interpreting the mechanisms that may help us understand the origin of congenital biliary diseases.
Collapse
Affiliation(s)
- Frédéric P Lemaigre
- de Duve Institute, Université Catholique de Louvain, 1200 Brussels, Belgium;
| |
Collapse
|
8
|
Yang L, Li LC, Wang X, Wang WH, Wang YC, Xu CR. The contributions of mesoderm-derived cells in liver development. Semin Cell Dev Biol 2018; 92:63-76. [PMID: 30193996 DOI: 10.1016/j.semcdb.2018.09.003] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Revised: 08/31/2018] [Accepted: 09/02/2018] [Indexed: 02/07/2023]
Abstract
The liver is an indispensable organ for metabolism and drug detoxification. The liver consists of endoderm-derived hepatobiliary lineages and various mesoderm-derived cells, and interacts with the surrounding tissues and organs through the ventral mesentery. Liver development, from hepatic specification to liver maturation, requires close interactions with mesoderm-derived cells, such as mesothelial cells, hepatic stellate cells, mesenchymal cells, liver sinusoidal endothelial cells and hematopoietic cells. These cells affect liver development through precise signaling events and even direct physical contact. Through the use of new techniques, emerging studies have recently led to a deeper understanding of liver development and its related mechanisms, especially the roles of mesodermal cells in liver development. Based on these developments, the current protocols for in vitro hepatocyte-like cell induction and liver-like tissue construction have been optimized and are of great importance for the treatment of liver diseases. Here, we review the roles of mesoderm-derived cells in the processes of liver development, hepatocyte-like cell induction and liver-like tissue construction.
Collapse
Affiliation(s)
- Li Yang
- Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, College of Life Sciences, Peking-Tsinghua Center for Life Sciences, China; Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, 100871, China
| | - Lin-Chen Li
- Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, College of Life Sciences, Peking-Tsinghua Center for Life Sciences, China; Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, 100871, China
| | - Xin Wang
- Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, College of Life Sciences, Peking-Tsinghua Center for Life Sciences, China
| | - Wei-Hua Wang
- Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, College of Life Sciences, Peking-Tsinghua Center for Life Sciences, China; Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, 100871, China
| | - Yan-Chun Wang
- Haidian Maternal & Child Health Hospital, Beijing, 100080, China
| | - Cheng-Ran Xu
- Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, College of Life Sciences, Peking-Tsinghua Center for Life Sciences, China.
| |
Collapse
|
9
|
So J, Khaliq M, Evason K, Ninov N, Martin BL, Stainier DY, Shin D. Wnt/β-catenin signaling controls intrahepatic biliary network formation in zebrafish by regulating notch activity. Hepatology 2018; 67:2352-2366. [PMID: 29266316 PMCID: PMC5991997 DOI: 10.1002/hep.29752] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Revised: 11/10/2017] [Accepted: 12/17/2017] [Indexed: 12/27/2022]
Abstract
UNLABELLED Malformations of the intrahepatic biliary structure cause cholestasis, a liver pathology that corresponds to poor bile flow, which leads to inflammation, fibrosis, and cirrhosis. Although the specification of biliary epithelial cells (BECs) that line the bile ducts is fairly well understood, the molecular mechanisms underlying intrahepatic biliary morphogenesis remain largely unknown. Wnt/β-catenin signaling plays multiple roles in liver biology; however, its role in intrahepatic biliary morphogenesis remains unclear. Using pharmacological and genetic tools that allow one to manipulate Wnt/β-catenin signaling, we show that in zebrafish both suppression and overactivation of Wnt/β-catenin signaling impaired intrahepatic biliary morphogenesis. Hepatocytes, but not BECs, exhibited Wnt/β-catenin activity; and the global suppression of Wnt/β-catenin signaling reduced Notch activity in BECs. Hepatocyte-specific suppression of Wnt/β-catenin signaling also reduced Notch activity in BECs, indicating a cell nonautonomous role for Wnt/β-catenin signaling in regulating hepatic Notch activity. Reducing Notch activity to the same level as that observed in Wnt-suppressed livers also impaired biliary morphogenesis. Intriguingly, expression of the Notch ligand genes jag1b and jag2b in hepatocytes was reduced in Wnt-suppressed livers and enhanced in Wnt-overactivated livers, revealing their regulation by Wnt/β-catenin signaling. Importantly, restoring Notch activity rescued the biliary defects observed in Wnt-suppressed livers. CONCLUSION Wnt/β-catenin signaling cell nonautonomously controls Notch activity in BECs by regulating the expression of Notch ligand genes in hepatocytes, thereby regulating biliary morphogenesis. (Hepatology 2018;67:2352-2366).
Collapse
Affiliation(s)
- Juhoon So
- Department of Developmental Biology, Pittsburgh Liver Research Center, McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Mehwish Khaliq
- Department of Developmental Biology, Pittsburgh Liver Research Center, McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Kimberley Evason
- Department of Biochemistry and Biophysics, Programs in Developmental and Stem Cell Biology, Genetics and Human Genetics, Diabetes Center, and Liver Center, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Nikolay Ninov
- Department of Biochemistry and Biophysics, Programs in Developmental and Stem Cell Biology, Genetics and Human Genetics, Diabetes Center, and Liver Center, University of California, San Francisco, San Francisco, CA 94158, USA,Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
| | - Benjamin L. Martin
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY 11794, USA
| | - Didier Y.R. Stainier
- Department of Biochemistry and Biophysics, Programs in Developmental and Stem Cell Biology, Genetics and Human Genetics, Diabetes Center, and Liver Center, University of California, San Francisco, San Francisco, CA 94158, USA,Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
| | - Donghun Shin
- Department of Developmental Biology, Pittsburgh Liver Research Center, McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15260, USA,Correspondence: Donghun Shin, 3501 5 Ave. #5063 Pittsburgh, PA 15260, 1-412-624-2144 (phone), 1-412-383-2211 (fax),
| |
Collapse
|
10
|
Ober EA, Lemaigre FP. Development of the liver: Insights into organ and tissue morphogenesis. J Hepatol 2018; 68:1049-1062. [PMID: 29339113 DOI: 10.1016/j.jhep.2018.01.005] [Citation(s) in RCA: 148] [Impact Index Per Article: 21.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Revised: 12/29/2017] [Accepted: 01/06/2018] [Indexed: 02/08/2023]
Abstract
Recent development of improved tools and methods to analyse tissues at the three-dimensional level has expanded our capacity to investigate morphogenesis of foetal liver. Here, we review the key morphogenetic steps during liver development, from the prehepatic endoderm stage to the postnatal period, and consider several model organisms while focussing on the mammalian liver. We first discuss how the liver buds out of the endoderm and gives rise to an asymmetric liver. We next outline the mechanisms driving liver and lobe growth, and review morphogenesis of the intra- and extrahepatic bile ducts; morphogenetic responses of the biliary tract to liver injury are discussed. Finally, we describe the mechanisms driving formation of the vasculature, namely venous and arterial vessels, as well as sinusoids.
Collapse
Affiliation(s)
- Elke A Ober
- Novo Nordisk Center for Stem Cell Biology (DanStem), University of Copenhagen, Copenhagen, Denmark
| | | |
Collapse
|
11
|
Fabris L, Spirli C, Cadamuro M, Fiorotto R, Strazzabosco M. Emerging concepts in biliary repair and fibrosis. Am J Physiol Gastrointest Liver Physiol 2017; 313:G102-G116. [PMID: 28526690 PMCID: PMC5582882 DOI: 10.1152/ajpgi.00452.2016] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2016] [Revised: 04/20/2017] [Accepted: 05/11/2017] [Indexed: 01/31/2023]
Abstract
Chronic diseases of the biliary tree (cholangiopathies) represent one of the major unmet needs in clinical hepatology and a significant knowledge gap in liver pathophysiology. The common theme in cholangiopathies is that the target of the disease is the biliary tree. After damage to the biliary epithelium, inflammatory changes stimulate a reparative response with proliferation of cholangiocytes and restoration of the biliary architecture, owing to the reactivation of a variety of morphogenetic signals. Chronic damage and inflammation will ultimately result in pathological repair with generation of biliary fibrosis and clinical progression of the disease. The hallmark of pathological biliary repair is the appearance of reactive ductular cells, a population of cholangiocyte-like epithelial cells of unclear and likely mixed origin that are able to orchestrate a complex process that involves a number of different cell types, under joint control of inflammatory and morphogenetic signals. Several questions remain open concerning the histogenesis of reactive ductular cells, their role in liver repair, their mechanism of activation, and the signals exchanged with the other cellular elements cooperating in the reparative process. This review contributes to the current debate by highlighting a number of new concepts derived from the study of the pathophysiology of chronic cholangiopathies, such as congenital hepatic fibrosis, biliary atresia, and Alagille syndrome.
Collapse
Affiliation(s)
- Luca Fabris
- Department of Molecular Medicine, University of Padua School of Medicine, Padua, Italy; .,Liver Center, Section of Digestive Diseases, Yale University School of Medicine, New Haven, Connecticut.,International Center for Digestive Health, University of Milan-Bicocca School of Medicine, Milan, Italy; and
| | - Carlo Spirli
- 2Liver Center, Section of Digestive Diseases, Yale University School of Medicine, New Haven, Connecticut; ,3International Center for Digestive Health, University of Milan-Bicocca School of Medicine, Milan, Italy; and
| | - Massimiliano Cadamuro
- 3International Center for Digestive Health, University of Milan-Bicocca School of Medicine, Milan, Italy; and ,4Department of Medicine and Surgery, University of Milan-Bicocca School of Medicine, Milan, Italy
| | - Romina Fiorotto
- 2Liver Center, Section of Digestive Diseases, Yale University School of Medicine, New Haven, Connecticut; ,3International Center for Digestive Health, University of Milan-Bicocca School of Medicine, Milan, Italy; and
| | - Mario Strazzabosco
- 2Liver Center, Section of Digestive Diseases, Yale University School of Medicine, New Haven, Connecticut; ,3International Center for Digestive Health, University of Milan-Bicocca School of Medicine, Milan, Italy; and ,4Department of Medicine and Surgery, University of Milan-Bicocca School of Medicine, Milan, Italy
| |
Collapse
|
12
|
Tanimizu N, Mitaka T. Epithelial Morphogenesis during Liver Development. Cold Spring Harb Perspect Biol 2017; 9:cshperspect.a027862. [PMID: 28213465 DOI: 10.1101/cshperspect.a027862] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Tissue stem/progenitor cells supply multiple types of epithelial cells that eventually acquire specialized functions during organ development. In addition, three-dimensional (3D) tissue structures need to be established for organs to perform their physiological functions. The liver contains two types of epithelial cells, namely, hepatocytes and cholangiocytes, which are derived from hepatoblasts, fetal liver stem/progenitor cells (LPCs), in mid-gestation. Hepatocytes performing many metabolic reactions form cord-like structures, whereas cholangiocytes, biliary epithelial cells, form tubular structures called intrahepatic bile ducts. Analyses for human genetic diseases and mutant mice have identified crucial molecules for liver organogenesis. Functions of those molecules can be examined in in vitro culture systems where LPCs are induced to differentiate into hepatocytes or cholangiocytes. Recent technical advances have revealed 3D epithelial morphogenesis during liver organogenesis. Therefore, the liver is a good model to understand how tissue stem/progenitor cells differentiate and establish 3D tissue architectures during organ development.
Collapse
Affiliation(s)
- Naoki Tanimizu
- Department of Tissue Development and Regeneration, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Chuo-ku, Sapporo 060-8556, Japan
| | - Toshihiro Mitaka
- Department of Tissue Development and Regeneration, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Chuo-ku, Sapporo 060-8556, Japan
| |
Collapse
|
13
|
Goto F, Kakinuma S, Miyoshi M, Tsunoda T, Kaneko S, Sato A, Asano Y, Otani S, Azuma S, Nagata H, Kawai-Kitahata F, Murakawa M, Nitta S, Itsui Y, Nakagawa M, Asahina Y, Watanabe M. Bone morphogenetic protein-4 modulates proliferation and terminal differentiation of fetal hepatic stem/progenitor cells. Hepatol Res 2017; 47:941-952. [PMID: 27670640 DOI: 10.1111/hepr.12823] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2016] [Revised: 09/12/2016] [Accepted: 09/23/2016] [Indexed: 12/13/2022]
Abstract
UNLABELLED Fetal hepatic stem/progenitor cells, called hepatoblasts, play central roles in liver organogenesis; however, molecular mechanisms regulating proliferation and terminal differentiation of such cells have not been completely elucidated. Bone morphogenetic protein-4 (BMP-4) is essential for the development of stem cells in various tissues, but its function in regulating the phenotype of hepatoblasts after the mid-gestational fetal stage remains unclear. The aim of this study is to clarify a functional role for BMP-4 in proliferation and terminal differentiation of murine hepatoblasts in mid-gestational fetal livers. METHODS A functional role for BMP-4 in proliferation and terminal differentiation of murine hepatoblasts was validated by assay of colony formation, biliary luminal formation, and hepatic maturation using primary hepatoblasts in vitro. Molecular mechanisms regulating such effects of BMP-4 on primary hepatoblasts were also analyzed. RESULTS Stimulation of BMP-4 upregulated phosphorylation of Smad1/5 in hepatoblasts. Bone morphogenetic protein-4 significantly suppressed colony formation of primary hepatoblasts in a dose-dependent manner, significantly suppressed cholangiocytic luminal formation of hepatoblasts, and promoted hepatic maturation of primary hepatoblasts. Stimulation of BMP-4 regulated the activation of several mitogen-activated protein kinases, such as extracellular signal-regulated kinase, Akt, p38 mitogen-activated protein kinase, and calcium/calmodulin-dependent protein kinase IIα in primary hepatoblasts. Moreover, Wnt5a, a molecule regulating cholangiocytic luminal formation, and BMP-4 coordinately suppressed proliferation and cholangiocytic luminal formation of hepatoblasts. CONCLUSION This study shows that BMP-4-mediated signaling controls proliferation and terminal differentiation of fetal hepatic stem/progenitor cells.
Collapse
Affiliation(s)
- Fumio Goto
- Department of Gastroenterology and Hepatology, Tokyo Medical and Dental University, Tokyo, Japan
| | - Sei Kakinuma
- Department of Gastroenterology and Hepatology, Tokyo Medical and Dental University, Tokyo, Japan.,Department for Liver Disease Control, Tokyo Medical and Dental University, Tokyo, Japan
| | - Masato Miyoshi
- Department of Gastroenterology and Hepatology, Tokyo Medical and Dental University, Tokyo, Japan
| | - Tomoyuki Tsunoda
- Department of Gastroenterology and Hepatology, Tokyo Medical and Dental University, Tokyo, Japan
| | - Shun Kaneko
- Department of Gastroenterology and Hepatology, Tokyo Medical and Dental University, Tokyo, Japan
| | - Ayako Sato
- Department of Gastroenterology and Hepatology, Tokyo Medical and Dental University, Tokyo, Japan
| | - Yu Asano
- Department of Gastroenterology and Hepatology, Tokyo Medical and Dental University, Tokyo, Japan
| | - Satoshi Otani
- Department of Gastroenterology and Hepatology, Tokyo Medical and Dental University, Tokyo, Japan
| | - Seishin Azuma
- Department of Gastroenterology and Hepatology, Tokyo Medical and Dental University, Tokyo, Japan
| | - Hiroko Nagata
- Department of Gastroenterology and Hepatology, Tokyo Medical and Dental University, Tokyo, Japan
| | - Fukiko Kawai-Kitahata
- Department of Gastroenterology and Hepatology, Tokyo Medical and Dental University, Tokyo, Japan
| | - Miyako Murakawa
- Department of Gastroenterology and Hepatology, Tokyo Medical and Dental University, Tokyo, Japan
| | - Sayuri Nitta
- Department of Gastroenterology and Hepatology, Tokyo Medical and Dental University, Tokyo, Japan
| | - Yasuhiro Itsui
- Department of Gastroenterology and Hepatology, Tokyo Medical and Dental University, Tokyo, Japan
| | - Mina Nakagawa
- Department of Gastroenterology and Hepatology, Tokyo Medical and Dental University, Tokyo, Japan
| | - Yasuhiro Asahina
- Department of Gastroenterology and Hepatology, Tokyo Medical and Dental University, Tokyo, Japan.,Department for Liver Disease Control, Tokyo Medical and Dental University, Tokyo, Japan
| | - Mamoru Watanabe
- Department of Gastroenterology and Hepatology, Tokyo Medical and Dental University, Tokyo, Japan
| |
Collapse
|
14
|
Zhang X, Xu Y, Chen JM, Liu C, Du GL, Zhang H, Chen GF, Jiang SL, Liu CH, Mu YP, Liu P. Huang Qi Decoction Prevents BDL-Induced Liver Fibrosis Through Inhibition of Notch Signaling Activation. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2017; 45:85-104. [PMID: 28081630 DOI: 10.1142/s0192415x17500070] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Notch signaling has been demonstrated to be involved in ductular reactions and fibrosis. Previous studies have shown that Huang Qi Decoction (HQD) can prevent the progression of cholestatic liver fibrosis (CLF). However, whether HQD affects the Notch signaling pathway is unclear. In this study, CLF was established by common bile duct ligation (BDL) in rats. At the end of the first week, the rats were randomly divided into a model group (i.e., BDL), an HQD group, and a sorafenib positive control group (SORA) and were treated for 3 weeks. Bile duct proliferation and liver fibrosis were determined by tissue staining. Activation of the Notch signaling pathway was evaluated by analyzing expressions of Notch-1, -2, -3, and -4, Jagged (JAG) 1, and Delta like (DLL)-1, -3, and -4. The results showed that HQD significantly reduced the deposition of collagen and the Hyp content of liver tissue and inhibited the activation of HSCs compared with the BDL group. In addition, HQD significantly decreased the protein and mRNA expressions of TGF-[Formula: see text]1 and [Formula: see text]-SMA. In contrast, HQD significantly enhanced expression of the Smad 7 protein. HQD also reduced biliary epithelial cell proliferation, and reduced the mRNA levels of CK7, CK8, CK18, SRY-related high mobility group-box gene (SOX) 9, epithelial cell adhesion molecule (EpCAM) and the positive areas of CK19 and OV6. In addition, the mRNA and protein expressions of Notch-3, -4, JAG1, and DLL-1, -3 were significantly reduced in the HQD compared to the BDL group. These results demonstrated that HQD may prevent biliary liver fibrosis through inhibition of the Notch signaling pathway, and it may be a potential treatment for cholestatic liver disease.
Collapse
Affiliation(s)
- Xiao Zhang
- * Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine (TCM), Institute of Liver Diseases, Key Laboratory of Liver and Kidney Diseases, China
- ‡ Shanghai Key Laboratory of TCM, Shanghai, P.R. China
- § Longhua Hospital Affiliated to Shanghai University of TCM, Shanghai, P.R. China
| | - Ying Xu
- * Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine (TCM), Institute of Liver Diseases, Key Laboratory of Liver and Kidney Diseases, China
- ‡ Shanghai Key Laboratory of TCM, Shanghai, P.R. China
| | - Jia-Mei Chen
- * Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine (TCM), Institute of Liver Diseases, Key Laboratory of Liver and Kidney Diseases, China
- ‡ Shanghai Key Laboratory of TCM, Shanghai, P.R. China
| | - Cheng Liu
- * Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine (TCM), Institute of Liver Diseases, Key Laboratory of Liver and Kidney Diseases, China
- ‡ Shanghai Key Laboratory of TCM, Shanghai, P.R. China
| | - Guang-Li Du
- † E-institute of Shanghai Municipal Education Commission, Shanghai University of TCM, Shanghai, P.R. China
| | - Hua Zhang
- * Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine (TCM), Institute of Liver Diseases, Key Laboratory of Liver and Kidney Diseases, China
- ‡ Shanghai Key Laboratory of TCM, Shanghai, P.R. China
| | - Gao-Feng Chen
- * Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine (TCM), Institute of Liver Diseases, Key Laboratory of Liver and Kidney Diseases, China
- ‡ Shanghai Key Laboratory of TCM, Shanghai, P.R. China
| | - Shi-Li Jiang
- * Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine (TCM), Institute of Liver Diseases, Key Laboratory of Liver and Kidney Diseases, China
- ‡ Shanghai Key Laboratory of TCM, Shanghai, P.R. China
| | - Cheng-Hai Liu
- * Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine (TCM), Institute of Liver Diseases, Key Laboratory of Liver and Kidney Diseases, China
- ‡ Shanghai Key Laboratory of TCM, Shanghai, P.R. China
| | - Yong-Ping Mu
- * Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine (TCM), Institute of Liver Diseases, Key Laboratory of Liver and Kidney Diseases, China
- ‡ Shanghai Key Laboratory of TCM, Shanghai, P.R. China
| | - Ping Liu
- * Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine (TCM), Institute of Liver Diseases, Key Laboratory of Liver and Kidney Diseases, China
- † E-institute of Shanghai Municipal Education Commission, Shanghai University of TCM, Shanghai, P.R. China
- ‡ Shanghai Key Laboratory of TCM, Shanghai, P.R. China
| |
Collapse
|
15
|
Gérard C, Tys J, Lemaigre FP. Gene regulatory networks in differentiation and direct reprogramming of hepatic cells. Semin Cell Dev Biol 2016; 66:43-50. [PMID: 27979774 DOI: 10.1016/j.semcdb.2016.12.003] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Accepted: 12/07/2016] [Indexed: 12/14/2022]
Abstract
Liver development proceeds by sequential steps during which gene regulatory networks (GRNs) determine differentiation and maturation of hepatic cells. Characterizing the architecture and dynamics of these networks is essential for understanding how cell fate decisions are made during development, and for recapitulating these processes during in vitro production of liver cells for toxicology studies, disease modelling and regenerative therapy. Here we review the GRNs that control key steps of liver development and lead to differentiation of hepatocytes and cholangiocytes in mammals. We focus on GRNs determining cell fate decisions and analyse subcircuitry motifs that may confer specific dynamic properties to the networks. Finally, we put our analysis in the perspective of recent attempts to directly reprogram cells to hepatocytes by forced expression of transcription factors.
Collapse
Affiliation(s)
- Claude Gérard
- Université catholique de Louvain, de Duve Institute, Avenue Hippocrate 75, 1200 Brussels, Belgium.
| | - Janne Tys
- Université catholique de Louvain, de Duve Institute, Avenue Hippocrate 75, 1200 Brussels, Belgium.
| | - Frédéric P Lemaigre
- Université catholique de Louvain, de Duve Institute, Avenue Hippocrate 75, 1200 Brussels, Belgium.
| |
Collapse
|
16
|
Comparison of the transcriptomic profile of hepatic human induced pluripotent stem like cells cultured in plates and in a 3D microscale dynamic environment. Genomics 2016; 109:16-26. [PMID: 27913249 DOI: 10.1016/j.ygeno.2016.11.008] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Revised: 11/14/2016] [Accepted: 11/28/2016] [Indexed: 12/12/2022]
Abstract
We have compared the transcriptomic profiles of human induced pluripotent stem cells after their differentiation in hepatocytes like cells in plates and microfluidic biochips. The biochips provided a 3D and dynamic support during the cell differentiation when compared to the 2D static cultures in plates. The microarray have demonstrated the up regulation of important pathway related to liver development and maturation during the culture in biochips. Furthermore, the results of the transcriptomic profile, coupled with immunostaining, and RTqPCR analysis have shown typical biomarkers illustrating the presence of responders of biliary like cells, hepatocytes like cells, and endothelial like cells. However, the overall tissue still presented characteristic of immature and foetal patterns. Nevertheless, the biochip culture provided a specific micro-environment in which a complex multicellular differentiation toward liver could be oriented.
Collapse
|
17
|
Ductular reaction-on-a-chip: Microfluidic co-cultures to study stem cell fate selection during liver injury. Sci Rep 2016; 6:36077. [PMID: 27796316 PMCID: PMC5086854 DOI: 10.1038/srep36077] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2016] [Accepted: 10/06/2016] [Indexed: 01/09/2023] Open
Abstract
Liver injury modulates local microenvironment, triggering production of signals that instruct stem cell fate choices. In this study, we employed a microfluidic co-culture system to recreate important interactions in the liver stem cell niche, those between adult hepatocytes and liver progenitor cells (LPCs). We demonstrate that pluripotent stem cell-derived LPCs choose hepatic fate when cultured next to healthy hepatocytes but begin biliary differentiation program when co-cultured with injured hepatocytes. We connect this fate selection to skewing in production of hepatocyte growth factor (HGF) and transforming growth factor (TGF)-β1 caused by injury. Significantly, biliary fate selection of LPCs was not observed in the absence of hepatocytes nor did it happen in the presence of TGF-β inhibitors. Our study demonstrates that microfluidic culture systems may offer an interesting new tool for dissecting cellular interactions leading to aberrant stem cell differentiation during injury.
Collapse
|
18
|
Loo CKC, Pereira TN, Ramsing M, Vogel I, Petersen OB, Ramm GA. Mechanism of pancreatic and liver malformations in human fetuses with short-rib polydactyly syndrome. ACTA ACUST UNITED AC 2016; 106:549-62. [PMID: 26970085 DOI: 10.1002/bdra.23495] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Revised: 02/09/2016] [Accepted: 02/11/2016] [Indexed: 12/29/2022]
Abstract
BACKGROUND The short-rib polydactyly (SRP) syndromes are rare skeletal dysplasias caused by abnormalities in primary cilia, sometimes associated with visceral malformations. METHODS The pathogenesis of ductal plate malformation (DPM) varies in different syndromes and has not been investigated in SRP. We have studied liver development in five SRP fetuses and pancreatic development in one SRP fetus, with genetically confirmed mutations in cilia related genes, with and without DPMs, using the immunoperoxidase technique, and compared these to other syndromes with DPM. RESULTS Acetylated tubulin expression was abnormal in DPM in SRP, Meckel syndrome, and autosomal recessive polycystic kidney disease (ARPKD), confirming ciliary anomalies. SDF-1 was abnormally expressed in SRP and two of three cases of autosomal dominant polycystic kidney disease (ADPKD) but not ARPKD or Meckel. Increased density of quiescent hepatic stellate cells was seen in SRP, Meckel, one of three cases of ARPKD, and two of three cases of ADPKD with aberrant hepatocyte expression of keratin 19 in SRP and ADPKD. Immunophenotypic abnormalities were present even in fetal liver without fully developed DPMs. The SRP case with DPM and pancreatic malformations showed abnormalities in the pancreatic head (influenced by mesenchyme from the septum transversum, similar to liver) but not pancreatic body (influenced by mesenchyme adjacent to the notochord). CONCLUSION In SRP, there are differentiation defects of hepatocytes, cholangiocytes, and liver mesenchyme and, in rare cases, pancreatic mesenchymal anomalies. The morphological changes were subtle in early gestation but immunophenotypic abnormalities were present. Mesenchymal-epithelial interactions may contribute to the malformations. Birth Defects Research (Part A) 106:549-562, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Christine K C Loo
- Department of Anatomical Pathology SEALS, Prince of Wales Hospital, Sydney, Australia (formerly: Department of Anatomical Pathology, Royal Brisbane and Women's Hospital, Brisbane, Australia.).,Hepatic Fibrosis Group, QIMR Berghofer Medical Research Institute, Brisbane, Australia.,Discipline of Pathology School of Medicine, University of Western Sydney, Australia
| | - Tamara N Pereira
- Hepatic Fibrosis Group, QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Mette Ramsing
- Department of Pathology, Aarhus University Hospital, Denmark
| | - Ida Vogel
- Department of Clinical Genetics, Aarhus University Hospital, Denmark
| | - Olav B Petersen
- Department of Obstetrics and Gynaecology, Aarhus University Hospital, Denmark
| | - Grant A Ramm
- Hepatic Fibrosis Group, QIMR Berghofer Medical Research Institute, Brisbane, Australia.,Faculty of Medicine and Biomedical Sciences, The University of Queensland, Brisbane, Australia
| |
Collapse
|
19
|
Cordi S, Godard C, Saandi T, Jacquemin P, Monga SP, Colnot S, Lemaigre FP. Role of β-catenin in development of bile ducts. Differentiation 2016; 91:42-9. [PMID: 26856660 DOI: 10.1016/j.diff.2016.02.001] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2015] [Revised: 01/15/2016] [Accepted: 02/01/2016] [Indexed: 01/13/2023]
Abstract
Beta-catenin is known to play stage- and cell-specific functions during liver development. However, its role in development of bile ducts has not yet been addressed. Here we used stage-specific in vivo gain- and loss-of-function approaches, as well as lineage tracing experiments in the mouse, to first demonstrate that β-catenin is dispensable for differentiation of liver precursor cells (hepatoblasts) to cholangiocyte precursors. Second, when β-catenin was depleted in the latter, maturation of cholangiocytes, bile duct morphogenesis and differentiation of periportal hepatocytes from cholangiocyte precursors was normal. In contrast, stabilization of β-catenin in cholangiocyte precursors perturbed duct development and cholangiocyte differentiation. We conclude that β-catenin is dispensable for biliary development but that its activity must be kept within tight limits. Our work is expected to significantly impact on in vitro differentiation of stem cells to cholangiocytes for toxicology studies and disease modeling.
Collapse
Affiliation(s)
- Sabine Cordi
- Université catholique de Louvain, de Duve Institute, Avenue Hippocrate 75/B1-7503, 1200 Brussels, Belgium
| | - Cécile Godard
- Institut Cochin, INSERM U1016; CNRS, UMR8104; Equipe labellisée Ligue Nationale Contre le Cancer; Université Paris Descartes, 24 rue du Faubourg Saint-Jacques, 75014 Paris, France
| | - Thoueiba Saandi
- Université catholique de Louvain, de Duve Institute, Avenue Hippocrate 75/B1-7503, 1200 Brussels, Belgium
| | - Patrick Jacquemin
- Université catholique de Louvain, de Duve Institute, Avenue Hippocrate 75/B1-7503, 1200 Brussels, Belgium
| | - Satdarshan P Monga
- Department of Pathology, Department of Medicine, University of Pittsburgh School of Medicine, 200 Lothrop St, Rm S-422 BST, Pittsburgh, PA 15261, USA
| | - Sabine Colnot
- Institut Cochin, INSERM U1016; CNRS, UMR8104; Equipe labellisée Ligue Nationale Contre le Cancer; Université Paris Descartes, 24 rue du Faubourg Saint-Jacques, 75014 Paris, France
| | - Frédéric P Lemaigre
- Université catholique de Louvain, de Duve Institute, Avenue Hippocrate 75/B1-7503, 1200 Brussels, Belgium.
| |
Collapse
|
20
|
Otani S, Kakinuma S, Kamiya A, Goto F, Kaneko S, Miyoshi M, Tsunoda T, Asano Y, Kawai-Kitahata F, Nitta S, Nakata T, Okamoto R, Itsui Y, Nakagawa M, Azuma S, Asahina Y, Yamaguchi T, Koshikawa N, Seiki M, Nakauchi H, Watanabe M. Matrix metalloproteinase-14 mediates formation of bile ducts and hepatic maturation of fetal hepatic progenitor cells. Biochem Biophys Res Commun 2016; 469:1062-8. [DOI: 10.1016/j.bbrc.2015.12.105] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2015] [Accepted: 12/22/2015] [Indexed: 01/29/2023]
|
21
|
Abstract
The liver is a central regulator of metabolism, and liver failure thus constitutes a major health burden. Understanding how this complex organ develops during embryogenesis will yield insights into how liver regeneration can be promoted and how functional liver replacement tissue can be engineered. Recent studies of animal models have identified key signaling pathways and complex tissue interactions that progressively generate liver progenitor cells, differentiated lineages and functional tissues. In addition, progress in understanding how these cells interact, and how transcriptional and signaling programs precisely coordinate liver development, has begun to elucidate the molecular mechanisms underlying this complexity. Here, we review the lineage relationships, signaling pathways and transcriptional programs that orchestrate hepatogenesis.
Collapse
Affiliation(s)
- Miriam Gordillo
- Department of Surgery, Weill Cornell Medical College, New York, NY 10065, USA
| | - Todd Evans
- Department of Surgery, Weill Cornell Medical College, New York, NY 10065, USA
| | - Valerie Gouon-Evans
- Department of Developmental and Regenerative Biology, Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| |
Collapse
|
22
|
Loo CKC, Pereira TN, Pozniak KN, Ramsing M, Vogel I, Ramm GA. The development of hepatic stellate cells in normal and abnormal human fetuses - an immunohistochemical study. Physiol Rep 2015; 3:3/8/e12504. [PMID: 26265759 PMCID: PMC4562587 DOI: 10.14814/phy2.12504] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
The precise embryological origin and development of hepatic stellate cells is not established. Animal studies and observations on human fetuses suggest that they derive from posterior mesodermal cells that migrate via the septum transversum and developing diaphragm to form submesothelial cells beneath the liver capsule, which give rise to mesenchymal cells including hepatic stellate cells. However, it is unclear if these are similar to hepatic stellate cells in adults or if this is the only source of stellate cells. We have studied hepatic stellate cells by immunohistochemistry, in developing human liver from autopsies of fetuses with and without malformations and growth restriction, using cellular Retinol Binding Protein-1 (cRBP-1), Glial Fibrillary Acidic Protein (GFAP), and α-Smooth Muscle Actin (αSMA) antibodies, to identify factors that influence their development. We found that hepatic stellate cells expressing cRBP-1 are present from the end of the first trimester of gestation and reduce in density throughout gestation. They appear abnormally formed and variably reduced in number in fetuses with abnormal mesothelial Wilms Tumor 1 (WT1) function, diaphragmatic hernia and in ectopic liver nodules without mesothelium. Stellate cells showed similarities to intravascular cells and their presence in a fetus with diaphragm agenesis suggests they may be derived from circulating stem cells. Our observations suggest circulating stem cells as well as mesothelium can give rise to hepatic stellate cells, and that they require normal mesothelial function for their development.
Collapse
Affiliation(s)
- Christine K C Loo
- Department of Anatomical Pathology, Prince of Wales Hospital, Randwick Sydney, NSW, Australia Hepatic Fibrosis Group, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia Discipline of Pathology, School of Medicine, University of Western Sydney, Sydney, NSW, Australia
| | - Tamara N Pereira
- Hepatic Fibrosis Group, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Katarzyna N Pozniak
- Hepatic Fibrosis Group, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Mette Ramsing
- Department of Pathology, Aarhus University Hospital, Aarhus, Denmark
| | - Ida Vogel
- Department of Clinical Genetics, Aarhus University Hospital, Aarhus, Denmark
| | - Grant A Ramm
- Hepatic Fibrosis Group, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia Faculty of Medicine and Biomedical Sciences, The University of Queensland, Brisbane, Queensland, Australia
| |
Collapse
|
23
|
Ye P, Wang T, Liu WH, Li XC, Tang LJ, Tian FZ. Enhancing HOTAIR/MiR-10b Drives Normal Liver Stem Cells Toward a Tendency to Malignant Transformation Through Inducing Epithelial- to-Mesenchymal Transition. Rejuvenation Res 2015; 18:332-340. [PMID: 25708830 DOI: 10.1089/rej.2014.1642] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Previously, other groups and our team consistently have demonstrated that the possible origination of liver cancer stem cells (LCSCs) is the malignant transformation from liver normal stem cells (LNSCs). However, this complex and multi-step process is far from clear due to the accumulation of various gene dysregulations. Because non-coding RNAs (ncRNAs) could regulate multiple genes, a family of genes, and even whole chromosomes, this study further investigated the effect of dysregulated short ncRNA microRNA-10b and long ncRNA HOX transcript antisense RNA (HOTAIR) between LNSCs and LCSCs on phenotype reversion. To clarify the role of ncRNA in malignant transformation of LNSCs, we used lentivirus transduction to enhance the miR-10b and HOTAIR expression levels in our previously isolated rat LNSCs. The malignant abilities of proliferation, invasiveness, and tumorigenesis were observed and compared in cells before and after ncRNAs enhancement. After microRNA-10b and HOTAIR were enhanced separately, several cancer stem cell (CSC)-like traits appeared in these LNSCs, including in vitro-enhanced proliferative capacity, expression of putative LCSC markers, progressive invasive ability, and even in vivo aggravation into and taking the place of normal liver tissue. Furthermore, strengthened expression of these ncRNAs partially degraded E-cadherin in LNSCs, which is one of the classic markers in epithelial-to-mesenchymal transition (EMT). HOTAIR or miR-10b enhanced in LNSCs may drive the LNSCs to a tendency toward malignant transformation. This study partially uncovers the mechanism by which miR-10b or HOTAIR promotes malignant transformation of LNSCs through down-regulating E-cadherin and inducing EMT.
Collapse
Affiliation(s)
- Ping Ye
- 1 Third Military Medical University , Chongqing, China
| | - Tao Wang
- 2 General Surgery Center, Chengdu Military General Hospital , Chengdu, Sichuan Province, China
| | - Wei-Hui Liu
- 2 General Surgery Center, Chengdu Military General Hospital , Chengdu, Sichuan Province, China
| | - Xiu-Chuan Li
- 3 Department of Cardiology, Chengdu Military General Hospital , Chengdu, Sichuan, Province, China
| | - Li-Jun Tang
- 2 General Surgery Center, Chengdu Military General Hospital , Chengdu, Sichuan Province, China
| | - Fu-Zhou Tian
- 2 General Surgery Center, Chengdu Military General Hospital , Chengdu, Sichuan Province, China
| |
Collapse
|
24
|
Yongping M, Zhang X, Xuewei L, Fan W, Chen J, Zhang H, Chen G, Liu C, Liu P. Astragaloside prevents BDL-induced liver fibrosis through inhibition of notch signaling activation. JOURNAL OF ETHNOPHARMACOLOGY 2015; 169:200-209. [PMID: 25917841 DOI: 10.1016/j.jep.2015.04.015] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2014] [Revised: 03/19/2015] [Accepted: 04/14/2015] [Indexed: 06/04/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Huangqi decoction was first described in Prescriptions of the Bureau of Taiping People׳s Welfare Pharmacy in the Song Dynasty (AD1078). It consists of Radix Astragali (Astragalus membranceus (Fisch.) Bge. Root, Huangqi) and Radix Glycyrrhizae (Glycyrrhiza uralensis Fisch., root and rhizome, Gancao), and it is an effective recipe that is usually used to treat consumptive disease and chronic liver diseases. Astragaloside (AS) is a main component of Radix Astragali had an effect similar to the Huangqi decoction on hepatic fibrosis. AIM OF THE STUDY Cholestasis is associated with a number of chronic liver diseases and Notch signaling has been demonstrated to be involved in ductular reaction. Previous studies have shown that AS can prevent the progression of cholestatic liver fibrosis, however, whether AS affects the Notch signaling pathway is unclear. MATERIALS AND METHODS Cholestatic liver fibrosis was established by common bile duct ligation (BDL) in rats. At first weekend, the rats were randomly divided into a model group (BDL), an AS group, and a Sorafenib positive control group (SORA) and treated for 3 weeks. Bile duct proliferation and liver fibrosis were determined by tissue staining. Activation of the Notch signaling pathway was evaluated by analyzing expressions of Notch-1, -2, -3, -4, Jagged 1 (JAG1), Delta-like (DLL)-1, -3, -4, Hes1, Numb and RBP-Jκ. Activation of the Wnt signaling pathway was evaluated by analyzing expressions of Wnt-4, -5a, -5b, Frizzled (Fzd)-2, -3, -6 and β-catenin. RESULTS (1) Compared with the BDL group, AS significantly reduced the deposition of collagen and the Hyp content of liver tissue and inhibited the activation of HSCs. In addition, AS significantly decreased the protein and mRNA expressions of TGF-β1 and α-SMA. In contrast, AS significantly enhanced expression of the Smad 7 protein. AS also reduced biliary epithelial cell proliferation, and reduced the mRNA and protein expressions of CK7, CK8, CK18, CK19, OV6, Sox9 and EpCAM. (2) The mRNA and protein expressions of Notch-2, -3, -4 and JAG1 were significantly reduced in the AS compared to the BDL group. In contrast, the mRNA and protein level of Numb was clearly enhanced after AS treatment. CONCLUSION AS may prevent biliary liver fibrosis via inhibition of the Notch signaling pathway, thereby inhibiting the abnormal proliferation of biliary epithelial cells. Results indicate that AS may be a potential therapeutic drug for cholestatic liver disease.
Collapse
Affiliation(s)
- Mu Yongping
- Shuguang Hospital affiliated to Shanghai University of Traditional Chinese Medicine, Institute of Liver Diseases, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xiao Zhang
- Shuguang Hospital affiliated to Shanghai University of Traditional Chinese Medicine, Institute of Liver Diseases, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Li Xuewei
- Shuguang Hospital affiliated to Shanghai University of Traditional Chinese Medicine, Institute of Liver Diseases, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Weiwei Fan
- Shuguang Hospital affiliated to Shanghai University of Traditional Chinese Medicine, Institute of Liver Diseases, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jiamei Chen
- Shuguang Hospital affiliated to Shanghai University of Traditional Chinese Medicine, Institute of Liver Diseases, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Hua Zhang
- Shuguang Hospital affiliated to Shanghai University of Traditional Chinese Medicine, Institute of Liver Diseases, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Gaofeng Chen
- Shuguang Hospital affiliated to Shanghai University of Traditional Chinese Medicine, Institute of Liver Diseases, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Chenghai Liu
- Shuguang Hospital affiliated to Shanghai University of Traditional Chinese Medicine, Institute of Liver Diseases, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Ping Liu
- Shuguang Hospital affiliated to Shanghai University of Traditional Chinese Medicine, Institute of Liver Diseases, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| |
Collapse
|
25
|
Tang WP, Akahoshi T, Piao JS, Narahara S, Murata M, Kawano T, Hamano N, Ikeda T, Hashizume M. Basic fibroblast growth factor-treated adipose tissue-derived mesenchymal stem cell infusion to ameliorate liver cirrhosis via paracrine hepatocyte growth factor. J Gastroenterol Hepatol 2015; 30:1065-74. [PMID: 25639333 DOI: 10.1111/jgh.12893] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/20/2014] [Indexed: 12/20/2022]
Abstract
BACKGROUND AND AIM Recent studies show that adipose tissue-derived mesenchymal stem cells have potential clinical applications. However, the mechanism has not been fully elucidated yet. Here, we investigated the effect of basic fibroblast growth factor-treated adipose tissue-derived mesenchymal stem cells infusion on a liver fibrosis rat model and elucidated the underlying mechanism. METHODS Adipose tissue-derived mesenchymal stem cells were infused into carbon tetrachloride-induced hepatic fibrosis rats through caudal vein. Liver functions and pathological changes were assessed. A co-culture model was used to clarify the potential mechanism. RESULTS Basic fibroblast growth factor treatment markedly improved the proliferation, differentiation, and hepatocyte growth factor expression ability of adipose tissue-derived mesenchymal stem cells. Although adipose tissue-derived mesenchymal stem cells infusion alone slightly ameliorated liver functions and suppressed fibrosis progression, basic fibroblast growth factor-treatment significantly enhanced the therapeutic effect in association with elevated hepatocyte growth factor expression. Moreover, double immunofluorescence staining confirmed that the infused cells located in fibrosis area. Furthermore, co-culture with adipose tissue-derived mesenchymal stem cell led to induction of hepatic stellate cell apoptosis and enhanced hepatocyte proliferation. However, these effects were significantly weakened by knockdown of hepatocyte growth factor. Mechanism investigation revealed that co-culture with adipose tissue-derived mesenchymal stem cells activated c-jun N-terminal kinase-p53 signaling in hepatic stellate cell and promoted apoptosis. CONCLUSIONS Basic fibroblast growth factor treatment enhanced the therapeutic effect of adipose tissue-derived mesenchymal stem cells, and secretion of hepatocyte growth factor from adipose tissue-derived mesenchymal stem cells plays a critical role in amelioration of liver injury and regression of fibrosis.
Collapse
Affiliation(s)
- Wei-Ping Tang
- Department of Disaster and Emergency Medicine, Faculty of Medicine, Kyushu University, Fukuoka, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Abdelhamed ZA, Natarajan S, Wheway G, Inglehearn CF, Toomes C, Johnson CA, Jagger DJ. The Meckel-Gruber syndrome protein TMEM67 controls basal body positioning and epithelial branching morphogenesis in mice via the non-canonical Wnt pathway. Dis Model Mech 2015; 8:527-41. [PMID: 26035863 PMCID: PMC4457033 DOI: 10.1242/dmm.019083] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2014] [Accepted: 04/01/2015] [Indexed: 01/16/2023] Open
Abstract
Ciliopathies are a group of developmental disorders that manifest with multi-organ anomalies. Mutations in TMEM67 (MKS3) cause a range of human ciliopathies, including Meckel-Gruber and Joubert syndromes. In this study we describe multi-organ developmental abnormalities in the Tmem67tm1Dgen/H1 knockout mouse that closely resemble those seen in Wnt5a and Ror2 knockout mice. These include pulmonary hypoplasia, ventricular septal defects, shortening of the body longitudinal axis, limb abnormalities, and cochlear hair cell stereociliary bundle orientation and basal body/kinocilium positioning defects. The basal body/kinocilium complex was often uncoupled from the hair bundle, suggesting aberrant basal body migration, although planar cell polarity and apical planar asymmetry in the organ of Corti were normal. TMEM67 (meckelin) is essential for phosphorylation of the non-canonical Wnt receptor ROR2 (receptor-tyrosine-kinase-like orphan receptor 2) upon stimulation with Wnt5a-conditioned medium. ROR2 also colocalises and interacts with TMEM67 at the ciliary transition zone. Additionally, the extracellular N-terminal domain of TMEM67 preferentially binds to Wnt5a in an in vitro binding assay. Cultured lungs of Tmem67 mutant mice failed to respond to stimulation of epithelial branching morphogenesis by Wnt5a. Wnt5a also inhibited both the Shh and canonical Wnt/β-catenin signalling pathways in wild-type embryonic lung. Pulmonary hypoplasia phenotypes, including loss of correct epithelial branching morphogenesis and cell polarity, were rescued by stimulating the non-canonical Wnt pathway downstream of the Wnt5a-TMEM67-ROR2 axis by activating RhoA. We propose that TMEM67 is a receptor that has a main role in non-canonical Wnt signalling, mediated by Wnt5a and ROR2, and normally represses Shh signalling. Downstream therapeutic targeting of the Wnt5a-TMEM67-ROR2 axis might, therefore, reduce or prevent pulmonary hypoplasia in ciliopathies and other congenital conditions. Highlighted Article: TMEM67 is a receptor of non-canonical Wnt signalling, implicating the Wnt5a-TMEM67-ROR2 axis during developmental signalling and disruption in ciliopathy disease state.
Collapse
Affiliation(s)
- Zakia A Abdelhamed
- Ciliopathy Research Group, Section of Ophthalmology and Neurosciences, Leeds Institute of Molecular Medicine, University of Leeds, Leeds LS9 7TF, UK Department of Anatomy and Embryology, Faculty of Medicine, Al-Azhar University, Cairo 11844, Egypt
| | - Subaashini Natarajan
- Ciliopathy Research Group, Section of Ophthalmology and Neurosciences, Leeds Institute of Molecular Medicine, University of Leeds, Leeds LS9 7TF, UK
| | - Gabrielle Wheway
- Ciliopathy Research Group, Section of Ophthalmology and Neurosciences, Leeds Institute of Molecular Medicine, University of Leeds, Leeds LS9 7TF, UK
| | - Christopher F Inglehearn
- Ciliopathy Research Group, Section of Ophthalmology and Neurosciences, Leeds Institute of Molecular Medicine, University of Leeds, Leeds LS9 7TF, UK
| | - Carmel Toomes
- Ciliopathy Research Group, Section of Ophthalmology and Neurosciences, Leeds Institute of Molecular Medicine, University of Leeds, Leeds LS9 7TF, UK
| | - Colin A Johnson
- Ciliopathy Research Group, Section of Ophthalmology and Neurosciences, Leeds Institute of Molecular Medicine, University of Leeds, Leeds LS9 7TF, UK
| | - Daniel J Jagger
- UCL Ear Institute, University College London, 332 Gray's Inn Road, London WC1X 8EE, UK
| |
Collapse
|
27
|
Wills ES, Roepman R, Drenth JPH. Polycystic liver disease: ductal plate malformation and the primary cilium. Trends Mol Med 2014; 20:261-70. [PMID: 24506938 DOI: 10.1016/j.molmed.2014.01.003] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2013] [Revised: 01/03/2014] [Accepted: 01/07/2014] [Indexed: 02/06/2023]
Abstract
Polycystic livers are found in autosomal dominant polycystic kidney disease (ADPKD), caused by polycystic kidney disease (PKD)1 and PKD2 mutations in virtually all cases, and in isolated polycystic liver disease (PCLD), where 20% of cases are caused by mutations in Protein kinase C substrate 80K-H (PRKCSH) or SEC63. Loss of heterozygosity in single hepatoblasts leads to underlying cystogenic ductal plate malformations. Crucially, actual components driving this development remain elusive. Recent advances have unraveled the roles of transforming growth factor (TGF)-β, Notch and Wnt signaling, transcriptional regulators such as hepatocyte nuclear factor (HNF)6 and HNF1β, as well as cilium function in hepatobiliary organogenesis. In polycystic liver disease, mutation or defective co-translational processing of key elements required for primary cilium formation have been implicated. This review recapitulates liver patterning factors in hepatobiliary development and extracts molecular players in hepatic cystogenesis.
Collapse
Affiliation(s)
- Edgar S Wills
- Department of Medicine, Division of Gastroenterology and Hepatology, Radboud University Medical Centre, 6500 HB Nijmegen, The Netherlands; Department of Human Genetics, Radboud University Medical Centre, 6500 HB Nijmegen, The Netherlands
| | - Ronald Roepman
- Department of Human Genetics, Radboud University Medical Centre, 6500 HB Nijmegen, The Netherlands; Radboud Institute of Molecular Life Sciences, Radboud University Medical Centre, 6500 HB Nijmegen, The Netherlands
| | - Joost P H Drenth
- Department of Medicine, Division of Gastroenterology and Hepatology, Radboud University Medical Centre, 6500 HB Nijmegen, The Netherlands.
| |
Collapse
|
28
|
Abstract
Wnt/β-catenin signaling plays key roles not only during development but also in adult tissue homeostasis. This is also evident in liver biology where many temporal roles of β-catenin have been identified during hepatic development, where, in hepatic progenitors or hepatoblasts, it is a key determinant of proliferation and eventually differentiation to mature hepatocytes, while also playing an important role in bile duct homeostasis. β-Catenin signaling cascade is mostly quiescent in hepatocytes in an adult liver except in the centrizonal region of a hepatic lobule. This small rim of hepatocytes around the central vein show constitutive β-catenin activation that in turn regulates expression of genes whose products play an important role in ammonia and xenobiotic metabolism. Intriguingly, β-catenin can also undergo activation in hepatocytes after acute liver loss secondary to surgical or toxicant insult. Such activation of this progrowth protein is observed as nuclear translocation of β-catenin and formation of its complex with the T-cell factor (TCF) family of transcription factors. Expression of cyclin-D1, a key inducer of transition from the G1 to S phase of cell cycle, is regulated by β-catenin-TCF complex. Thus, β-catenin activation is absolutely critical in the normal regeneration process of the liver as shown by studies in several models across various species. In the current review, the temporal role and regulation of β-catenin in liver development, metabolic zonation in a basal adult liver, and during the liver regeneration process will be discussed. In addition, the probability of therapeutically regulating β-catenin activity as a possible future treatment strategy for liver insufficiency will also be discussed.
Collapse
|
29
|
Liu WH, Ren LN, Chen T, Liu LY, Tang LJ. Stages based molecular mechanisms for generating cholangiocytes from liver stem/progenitor cells. World J Gastroenterol 2013; 19:7032-7041. [PMID: 24222945 PMCID: PMC3819537 DOI: 10.3748/wjg.v19.i41.7032] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2013] [Revised: 09/01/2013] [Accepted: 09/15/2013] [Indexed: 02/06/2023] Open
Abstract
Except for the most organized mature hepatocytes, liver stem/progenitor cells (LSPCs) can differentiate into many other types of cells in the liver including cholangiocytes. In addition, LSPCs are demonstrated to be able to give birth to other kinds of extra-hepatic cell types such as insulin-producing cells. Even more, under some bad conditions, these LSPCs could generate liver cancer stem like cells (LCSCs) through malignant transformation. In this review, we mainly concentrate on the molecular mechanisms for controlling cell fates of LSPCs, especially differentiation of cholangiocytes, insulin-producing cells and LCSCs. First of all, to certificate the cell fates of LSPCs, the following three features need to be taken into account to perform accurate phenotyping: (1) morphological properties; (2) specific markers; and (3) functional assessment including in vivo transplantation. Secondly, to promote LSPCs differentiation, systematical attention should be paid to inductive materials (such as growth factors and chemical stimulators), progressive materials including intracellular and extracellular signaling pathways, and implementary materials (such as liver enriched transcriptive factors). Accordingly, some recommendations were proposed to standardize, optimize, and enrich the effective production of cholangiocyte-like cells out of LSPCs. At the end, the potential regulating mechanisms for generation of cholangiocytes by LSPCs were carefully analyzed. The differentiation of LSPCs is a gradually progressing process, which consists of three main steps: initiation, progression and accomplishment. It's the unbalanced distribution of affecting materials in each step decides the cell fates of LSPCs.
Collapse
|