51
|
Kim M, Delgado E, Ko S. DNA methylation in cell plasticity and malignant transformation in liver diseases. Pharmacol Ther 2023; 241:108334. [PMID: 36535346 PMCID: PMC9841769 DOI: 10.1016/j.pharmthera.2022.108334] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 12/09/2022] [Accepted: 12/14/2022] [Indexed: 12/23/2022]
Abstract
The liver possesses extraordinary regenerative capacity mainly attributable to the ability of hepatocytes (HCs) and biliary epithelial cells (BECs) to self-replicate. This ability is left over from their bipotent parent cell, the hepatoblast, during development. When this innate regeneration is compromised due to the absence of proliferative parenchymal cells, such as during cirrhosis, HCs and BEC can transdifferentiate; thus, adding another layer of complexity to the process of liver repair. In addition, dysregulated lineage maintenance in these two cell populations has been shown to promote malignant growth in experimental conditions. Here, malignant transformation, driven in part by insufficient maintenance of lineage reprogramming, contributes to end-stage liver disease. Epigenetic changes are key drivers for cell fate decisions as well as transformation by finetuning overall transcription and gene expression. In this review, we address how altered DNA methylation contributes to the initiation and progression of hepatic cell fate conversion and cancer formation. We also discussed the diagnostic and therapeutic potential of targeting DNA methylation in liver cancer, its current limitations, and what future research is necessary to facilitate its contribution to clinical translation.
Collapse
Affiliation(s)
- Minwook Kim
- Department of Developmental Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States of America
| | - Evan Delgado
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States of America; Pittsburgh Liver Research Center, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States of America
| | - Sungjin Ko
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States of America; Pittsburgh Liver Research Center, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States of America.
| |
Collapse
|
52
|
Chen T, Dalton G, Oh SH, Maeso-Diaz R, Du K, Meyers RA, Guy C, Abdelmalek MF, Henao R, Guarnieri P, Pullen SS, Gregory S, Locker J, Brown JM, Diehl AM. Hepatocyte Smoothened Activity Controls Susceptibility to Insulin Resistance and Nonalcoholic Fatty Liver Disease. Cell Mol Gastroenterol Hepatol 2022; 15:949-970. [PMID: 36535507 PMCID: PMC9957752 DOI: 10.1016/j.jcmgh.2022.12.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 12/09/2022] [Accepted: 12/09/2022] [Indexed: 01/07/2023]
Abstract
BACKGROUND & AIMS Nonalcoholic steatohepatitis (NASH), a leading cause of cirrhosis, strongly associates with the metabolic syndrome, an insulin-resistant proinflammatory state that disrupts energy balance and promotes progressive liver degeneration. We aimed to define the role of Smoothened (Smo), an obligatory component of the Hedgehog signaling pathway, in controlling hepatocyte metabolic homeostasis and, thereby, susceptibility to NASH. METHODS We conditionally deleted Smo in hepatocytes of healthy chow-fed mice and performed metabolic phenotyping, coupled with single-cell RNA sequencing (RNA-seq), to characterize the role of hepatocyte Smo in regulating basal hepatic and systemic metabolic homeostasis. Liver RNA-seq datasets from 2 large human cohorts were also analyzed to define the relationship between Smo and NASH susceptibility in people. RESULTS Hepatocyte Smo deletion inhibited the Hedgehog pathway and promoted fatty liver, hyperinsulinemia, and insulin resistance. We identified a plausible mechanism whereby inactivation of Smo stimulated the mTORC1-SREBP1c signaling axis, which promoted lipogenesis while inhibiting the hepatic insulin cascade. Transcriptomics of bulk and single Smo-deficient hepatocytes supported suppression of insulin signaling and also revealed molecular abnormalities associated with oxidative stress and mitochondrial dysfunction. Analysis of human bulk RNA-seq data revealed that Smo expression was (1) highest in healthy livers, (2) lower in livers with NASH than in those with simple steatosis, (3) negatively correlated with markers of insulin resistance and liver injury, and (4) declined progressively as fibrosis severity worsened. CONCLUSIONS The Hedgehog pathway controls insulin sensitivity and energy homeostasis in adult livers. Loss of hepatocyte Hedgehog activity induces hepatic and systemic metabolic stress and enhances susceptibility to NASH by promoting hepatic lipoxicity and insulin resistance.
Collapse
Affiliation(s)
- Tianyi Chen
- Department of Medicine, Duke University, Durham, North Carolina
| | - George Dalton
- Department of Medicine, Duke University, Durham, North Carolina
| | - Seh-Hoon Oh
- Department of Medicine, Duke University, Durham, North Carolina
| | | | - Kuo Du
- Department of Medicine, Duke University, Durham, North Carolina
| | - Rachel A Meyers
- Department of Medicine, Duke University, Durham, North Carolina
| | - Cynthia Guy
- Department of Medicine, Duke University, Durham, North Carolina
| | | | - Ricardo Henao
- Department of Medicine, Duke University, Durham, North Carolina
| | - Paolo Guarnieri
- Boehringer Ingelheim Pharmaceuticals Inc, Ridgefield, Connecticut
| | - Steven S Pullen
- Boehringer Ingelheim Pharmaceuticals Inc, Ridgefield, Connecticut
| | - Simon Gregory
- Department of Medicine, Duke University, Durham, North Carolina
| | - Joseph Locker
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - J Mark Brown
- Department of Cardiovascular and Metabolic Sciences, Cleveland Clinic Lerner Research Institute, Cleveland, Ohio
| | - Anna Mae Diehl
- Department of Medicine, Duke University, Durham, North Carolina.
| |
Collapse
|
53
|
Zhao C, Matalonga J, Lancman JJ, Liu L, Xiao C, Kumar S, Gates KP, He J, Graves A, Huisken J, Azuma M, Lu Z, Chen C, Ding BS, Dong PDS. Regenerative failure of intrahepatic biliary cells in Alagille syndrome rescued by elevated Jagged/Notch/Sox9 signaling. Proc Natl Acad Sci U S A 2022; 119:e2201097119. [PMID: 36469766 PMCID: PMC9897440 DOI: 10.1073/pnas.2201097119] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Accepted: 10/21/2022] [Indexed: 12/08/2022] Open
Abstract
Despite the robust healing capacity of the liver, regenerative failure underlies numerous hepatic diseases, including the JAG1 haploinsufficient disorder, Alagille syndrome (ALGS). Cholestasis due to intrahepatic duct (IHD) paucity resolves in certain ALGS cases but fails in most with no clear mechanisms or therapeutic interventions. We find that modulating jag1b and jag2b allele dosage is sufficient to stratify these distinct outcomes, which can be either exacerbated or rescued with genetic manipulation of Notch signaling, demonstrating that perturbations of Jag/Notch signaling may be causal for the spectrum of ALGS liver severities. Although regenerating IHD cells proliferate, they remain clustered in mutants that fail to recover due to a blunted elevation of Notch signaling in the distal-most IHD cells. Increased Notch signaling is required for regenerating IHD cells to branch and segregate into the peripheral region of the growing liver, where biliary paucity is commonly observed in ALGS. Mosaic loss- and-gain-of-function analysis reveals Sox9b to be a key Notch transcriptional effector required cell autonomously to regulate these cellular dynamics during IHD regeneration. Treatment with a small-molecule putative Notch agonist stimulates Sox9 expression in ALGS patient fibroblasts and enhances hepatic sox9b expression, rescues IHD paucity and cholestasis, and increases survival in zebrafish mutants, thereby providing a proof-of-concept therapeutic avenue for this disorder.
Collapse
Affiliation(s)
- Chengjian Zhao
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Sichuan, 610041People’s Republic of China
- Human Genetics Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA92037
| | - Jonathan Matalonga
- Human Genetics Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA92037
| | - Joseph J. Lancman
- Human Genetics Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA92037
| | - Lu Liu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Sichuan, 610041People’s Republic of China
| | - Chaoxin Xiao
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Sichuan, 610041People’s Republic of China
| | - Shiv Kumar
- Human Genetics Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA92037
| | - Keith P. Gates
- Human Genetics Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA92037
| | - Jiaye He
- Morgridge Institute for Research, Madison, WI53715
| | | | - Jan Huisken
- Morgridge Institute for Research, Madison, WI53715
- Department of Integrative Biology, University of Wisconsin-Madison, Madison, WI53706
| | - Mizuki Azuma
- Department of Molecular Biosciences, University of Kansas, Lawrence, KS66045
| | - Zhenghao Lu
- Chengdu Organoidmed Medical Laboratory Ltd., Sichuan, 610041People’s Republic of China
| | - Chong Chen
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Sichuan, 610041People’s Republic of China
| | - Bi-Sen Ding
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Sichuan, 610041People’s Republic of China
| | - P. Duc Si Dong
- Human Genetics Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA92037
- Graduate School of Biomedical Sciences, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA92037
| |
Collapse
|
54
|
Rosenberg N, Van Haele M, Lanton T, Brashi N, Bromberg Z, Adler H, Giladi H, Peled A, Goldenberg DS, Axelrod JH, Simerzin A, Chai C, Paldor M, Markezana A, Yaish D, Shemulian Z, Gross D, Barnoy S, Gefen M, Amran O, Claerhout S, Fernández-Vaquero M, García-Beccaria M, Heide D, Shoshkes-Carmel M, Schmidt Arras D, Elgavish S, Nevo Y, Benyamini H, Tirnitz-Parker JEE, Sanchez A, Herrera B, Safadi R, Kaestner KH, Rose-John S, Roskams T, Heikenwalder M, Galun E. Combined hepatocellular-cholangiocarcinoma derives from liver progenitor cells and depends on senescence and IL-6 trans-signaling. J Hepatol 2022; 77:1631-1641. [PMID: 35988690 DOI: 10.1016/j.jhep.2022.07.029] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 07/07/2022] [Accepted: 07/19/2022] [Indexed: 12/27/2022]
Abstract
BACKGROUND & AIMS Primary liver cancers include hepatocellular carcinoma (HCC), intrahepatic cholangiocarcinoma (CCA) and combined HCC-CCA tumors (cHCC-CCA). It has been suggested, but not unequivocally proven, that hepatic progenitor cells (HPCs) can contribute to hepatocarcinogenesis. We aimed to determine whether HPCs contribute to HCC, cHCC-CCA or both types of tumors. METHODS To trace progenitor cells during hepatocarcinogenesis, we generated Mdr2-KO mice that harbor a yellow fluorescent protein (YFP) reporter gene driven by the Foxl1 promoter which is expressed specifically in progenitor cells. These mice (Mdr2-KOFoxl1-CRE;RosaYFP) develop chronic inflammation and HCCs by the age of 14-16 months, followed by cHCC-CCA tumors at the age of 18 months. RESULTS In this Mdr2-KOFoxl1-CRE;RosaYFP mouse model, liver progenitor cells are the source of cHCC-CCA tumors, but not the source of HCC. Ablating the progenitors, caused reduction of cHCC-CCA tumors but did not affect HCCs. RNA-sequencing revealed enrichment of the IL-6 signaling pathway in cHCC-CCA tumors compared to HCC tumors. Single-cell RNA-sequencing (scRNA-seq) analysis revealed that IL-6 is expressed by immune and parenchymal cells during senescence, and that IL-6 is part of the senescence-associated secretory phenotype. Administration of an anti-IL-6 antibody to Mdr2-KOFoxl1-CRE;RosaYFP mice inhibited the development of cHCC-CCA tumors. Blocking IL-6 trans-signaling led to a decrease in the number and size of cHCC-CCA tumors, indicating their dependence on this pathway. Furthermore, the administration of a senolytic agent inhibited IL-6 and the development of cHCC-CCA tumors. CONCLUSION Our results demonstrate that cHCC-CCA, but not HCC tumors, originate from HPCs, and that IL-6, which derives in part from cells in senescence, plays an important role in this process via IL-6 trans-signaling. These findings could be applied to develop new therapeutic approaches for cHCC-CCA tumors. LAY SUMMARY Combined hepatocellular carcinoma-cholangiocarcinoma is the third most prevalent type of primary liver cancer (i.e. a cancer that originates in the liver). Herein, we show that this type of cancer originates in stem cells in the liver and that it depends on inflammatory signaling. Specifically, we identify a cytokine called IL-6 that appears to be important in the development of these tumors. Our results could be used for the development of novel treatments for these aggressive tumors.
Collapse
Affiliation(s)
- Nofar Rosenberg
- Goldyne Savad Institute of Gene and Cell Therapy, Hadassah Hebrew University Hospital, Jerusalem, Israel
| | - Matthias Van Haele
- Department of Imaging and Pathology, Translational Cell and Tissue Research, KU Leuven and University Hospitals Leuven, Leuven, Belgium; Department of Pathology, GROW School for Oncology and Developmental Biology, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Tali Lanton
- Goldyne Savad Institute of Gene and Cell Therapy, Hadassah Hebrew University Hospital, Jerusalem, Israel
| | - Neta Brashi
- Goldyne Savad Institute of Gene and Cell Therapy, Hadassah Hebrew University Hospital, Jerusalem, Israel
| | - Zohar Bromberg
- Goldyne Savad Institute of Gene and Cell Therapy, Hadassah Hebrew University Hospital, Jerusalem, Israel
| | - Hanan Adler
- Goldyne Savad Institute of Gene and Cell Therapy, Hadassah Hebrew University Hospital, Jerusalem, Israel
| | - Hilla Giladi
- Goldyne Savad Institute of Gene and Cell Therapy, Hadassah Hebrew University Hospital, Jerusalem, Israel
| | - Amnon Peled
- Goldyne Savad Institute of Gene and Cell Therapy, Hadassah Hebrew University Hospital, Jerusalem, Israel
| | - Daniel S Goldenberg
- Goldyne Savad Institute of Gene and Cell Therapy, Hadassah Hebrew University Hospital, Jerusalem, Israel
| | - Jonathan H Axelrod
- Goldyne Savad Institute of Gene and Cell Therapy, Hadassah Hebrew University Hospital, Jerusalem, Israel
| | - Alina Simerzin
- Goldyne Savad Institute of Gene and Cell Therapy, Hadassah Hebrew University Hospital, Jerusalem, Israel
| | - Chofit Chai
- Goldyne Savad Institute of Gene and Cell Therapy, Hadassah Hebrew University Hospital, Jerusalem, Israel
| | - Mor Paldor
- Goldyne Savad Institute of Gene and Cell Therapy, Hadassah Hebrew University Hospital, Jerusalem, Israel
| | - Auerlia Markezana
- Goldyne Savad Institute of Gene and Cell Therapy, Hadassah Hebrew University Hospital, Jerusalem, Israel
| | - Dayana Yaish
- Goldyne Savad Institute of Gene and Cell Therapy, Hadassah Hebrew University Hospital, Jerusalem, Israel
| | - Zohar Shemulian
- Goldyne Savad Institute of Gene and Cell Therapy, Hadassah Hebrew University Hospital, Jerusalem, Israel
| | - Dvora Gross
- Goldyne Savad Institute of Gene and Cell Therapy, Hadassah Hebrew University Hospital, Jerusalem, Israel
| | - Shanny Barnoy
- Goldyne Savad Institute of Gene and Cell Therapy, Hadassah Hebrew University Hospital, Jerusalem, Israel
| | - Maytal Gefen
- Goldyne Savad Institute of Gene and Cell Therapy, Hadassah Hebrew University Hospital, Jerusalem, Israel
| | - Osher Amran
- Goldyne Savad Institute of Gene and Cell Therapy, Hadassah Hebrew University Hospital, Jerusalem, Israel
| | - Sofie Claerhout
- Department of Imaging and Pathology, Translational Cell and Tissue Research, KU Leuven and University Hospitals Leuven, Leuven, Belgium
| | - Mirian Fernández-Vaquero
- Division of Chronic Inflammation and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - María García-Beccaria
- Division of Chronic Inflammation and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Danijela Heide
- Division of Chronic Inflammation and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Michal Shoshkes-Carmel
- Department of Genetics and Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Center for Translational Research, Philadelphia, USA
| | - Dirk Schmidt Arras
- Institut für Biochemie, Christian-Albrechts-Universität zu Kiel, Kiel, Germany; Department of Biosciences, University of Salzburg, Salzburg, Austria
| | - Sharona Elgavish
- Bioinformatics Unit, The Institute for Medical Research Israel-Canada, Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Yuval Nevo
- Bioinformatics Unit, The Institute for Medical Research Israel-Canada, Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Hadar Benyamini
- Bioinformatics Unit, The Institute for Medical Research Israel-Canada, Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Janina E E Tirnitz-Parker
- Centre for Medical Research, University of Western Australia & Harry Perkins Institute of Medical Research, Crawley, Australia
| | - Aranzazu Sanchez
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Complutense University of Madrid, Spain
| | - Blanca Herrera
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Complutense University of Madrid, Spain
| | - Rifaat Safadi
- The Liver Institute, Hadassah Hebrew University Hospital, Jerusalem, Israel
| | - Klaus H Kaestner
- Department of Genetics and Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Center for Translational Research, Philadelphia, USA
| | - Stefan Rose-John
- Institut für Biochemie, Christian-Albrechts-Universität zu Kiel, Kiel, Germany
| | - Tania Roskams
- Department of Imaging and Pathology, Translational Cell and Tissue Research, KU Leuven and University Hospitals Leuven, Leuven, Belgium
| | - Mathias Heikenwalder
- Division of Chronic Inflammation and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany; The M3 Research Institute, Rosenauer Weg 30, Medical Faculty Tuebingen (MFT), 72076 Tuebingen, Germany.
| | - Eithan Galun
- Goldyne Savad Institute of Gene and Cell Therapy, Hadassah Hebrew University Hospital, Jerusalem, Israel.
| |
Collapse
|
55
|
Liu HL, Yang AY, Xiong QF, Zhong YD, Liu DX, Huang P, Feng XN, Zhang Y, Yang YF. Aberrant cytokeratin 7 expression by hepatocytes can predict the ductopenia grade in primary biliary cholangitis. BMC Gastroenterol 2022; 22:443. [PMID: 36324070 PMCID: PMC9628093 DOI: 10.1186/s12876-022-02538-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Accepted: 10/12/2022] [Indexed: 11/07/2022] Open
Abstract
Background Aberrant cytokeratin 7 expression by hepatocytes (CK7+Hs) is the hallmark characteristic of cholestasis diseases, especially in ductopenia diseases such as primary biliary cholangitis (PBC). This study attempted to evaluate the differences and relationships between the clinical and histological features of aberrant cytokeratin 7 (CK7) expression by hepatocytes in PBC patients. Methods The clinicopathological data of patients diagnosed with PBC at the Second Hospital of Nanjing between January 2016 and September 2018 were analysed with SPSS 20.0. Results Eighty-nine PBC patients who underwent liver biopsy were enrolled in this study, and 15, 29 and 45 patients had aberrant CK7 expression by hepatocytes (CK7+Hs (2 +), CK7+Hs (1 +), and CK7−Hs, respectively). There were significant differences in TB, DB, ALP, TA, IgM, interface activity, and ductopenia grade between patients with CK7−Hs and CK7+Hs (2 +) (P < 0.05). The ductopenia grade was also significantly different between patients with CK7+Hs (2 +) and CK7+Hs (1 +) according to sex (P < 0.05). Upon merging the data of CK7+Hs (2 +) and CK7+Hs (1 +) into CK7+Hs, we found significant differences in AMA, AMA-M2, anti-gp210, TB, DB, ALP, TA, IgM, fibrosis, and ductopenia grade between CK7+Hs and CK7−Hs (P < 0.05). The odds ratios (ORs) (and 95% confidence intervals (CIs)) of CK7+Hs according to anti-gp210, ductopenia grade, and interface activity were 6.413 (95% CI 1.363–30.162), 4.145 (95% CI 1.898–9.052) and 3.247 (95% CI 1.556–6.775), respectively (P < 0.05). Spearman's rank correlation according to interface activity and ductopenia grade in patients with CK7+Hs (2 + , 1 + , 0) was r = 0.359 (P = 0.001) and r = 0.396 (P < 0.001), respectively. Conclusion CK7+Hs serves as a cholestasis index of PBC and are associated with the ductopenia grade and interface activity. Aberrant cytokeratin 7 expression by hepatocytes can predict the ductopenia grade in primary biliary cholangitis.
Collapse
Affiliation(s)
- Hong-Li Liu
- Southeast University School of Medicine, No87 Dingjiaqiao Road, Gulou District, Nanjing, 210009, China.,The Second Hospital of Nanjing, Teaching Hospital of Southeast University, No.1 Zhongfu Road, Gulou District, Nanjing, 210003, China
| | - An-Yin Yang
- Department of Liver Diseases, The Second Hospital of Nanjing, Nanjing University of Chinese Medicine, No.1 Zhongfu Road, Gulou District, Nanjing, 210003, China
| | - Qing-Fang Xiong
- Department of Liver Diseases, The Second Hospital of Nanjing, Nanjing University of Chinese Medicine, No.1 Zhongfu Road, Gulou District, Nanjing, 210003, China
| | - Yan-Dan Zhong
- Department of Liver Diseases, The Second Hospital of Nanjing, Nanjing University of Chinese Medicine, No.1 Zhongfu Road, Gulou District, Nanjing, 210003, China
| | - Du-Xian Liu
- Department of Pathology, The Second Hospital of Nanjing, Nanjing University of Chinese Medicine, No.1 Zhongfu Road, Gulou District, Nanjing, 210003, China
| | - Ping Huang
- Department of Liver Diseases, The Second Hospital of Nanjing, Nanjing University of Chinese Medicine, No.1 Zhongfu Road, Gulou District, Nanjing, 210003, China
| | - Xiao-Ning Feng
- Department of Liver Diseases, The Second Hospital of Nanjing, Nanjing University of Chinese Medicine, No.1 Zhongfu Road, Gulou District, Nanjing, 210003, China
| | - Yu Zhang
- Southeast University School of Medicine, No87 Dingjiaqiao Road, Gulou District, Nanjing, 210009, China
| | - Yong-Feng Yang
- The Second Hospital of Nanjing, Teaching Hospital of Southeast University, No.1 Zhongfu Road, Gulou District, Nanjing, 210003, China. .,Department of Liver Diseases, The Second Hospital of Nanjing, Nanjing University of Chinese Medicine, No.1 Zhongfu Road, Gulou District, Nanjing, 210003, China.
| |
Collapse
|
56
|
Peng J, Li F, Wang J, Wang C, Jiang Y, Liu B, He J, Yuan K, Pan C, Lin M, Zhou B, Chen L, Gao D, Zhao Y. Identification of a rare Gli1 + progenitor cell population contributing to liver regeneration during chronic injury. Cell Discov 2022; 8:118. [PMID: 36316325 PMCID: PMC9622734 DOI: 10.1038/s41421-022-00474-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 09/21/2022] [Indexed: 11/06/2022] Open
Abstract
In adults, hepatocytes are mainly replenished from the existing progenitor pools of hepatocytes and cholangiocytes during chronic liver injury. However, it is unclear whether other cell types in addition to classical hepatocytes and cholangiocytes contribute to hepatocyte regeneration after chronic liver injuries. Here, we identified a new biphenotypic cell population that contributes to hepatocyte regeneration during chronic liver injuries. We found that a cell population expressed Gli1 and EpCAM (EpCAM+Gli1+), which was further characterized with both epithelial and mesenchymal identities by single-cell RNA sequencing. Genetic lineage tracing using dual recombinases revealed that Gli1+ nonhepatocyte cell population could generate hepatocytes after chronic liver injury. EpCAM+Gli1+ cells exhibited a greater capacity for organoid formation with functional hepatocytes in vitro and liver regeneration upon transplantation in vivo. Collectively, these findings demonstrate that EpCAM+Gli1+ cells can serve as a new source of liver progenitor cells and contribute to liver repair and regeneration.
Collapse
Affiliation(s)
- Jiayin Peng
- grid.9227.e0000000119573309State Key Laboratory of Molecular Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China
| | - Fei Li
- grid.9227.e0000000119573309State Key Laboratory of Molecular Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China
| | - Jia Wang
- grid.9227.e0000000119573309State Key Laboratory of Molecular Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China ,grid.410726.60000 0004 1797 8419University of Chinese Academy of Sciences, Beijing, China
| | - Chaoxiong Wang
- grid.9227.e0000000119573309State Key Laboratory of Molecular Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China ,grid.410726.60000 0004 1797 8419University of Chinese Academy of Sciences, Beijing, China
| | - Yiao Jiang
- grid.9227.e0000000119573309State Key Laboratory of Molecular Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China ,grid.410726.60000 0004 1797 8419University of Chinese Academy of Sciences, Beijing, China
| | - Biao Liu
- grid.9227.e0000000119573309State Key Laboratory of Molecular Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China ,grid.410726.60000 0004 1797 8419University of Chinese Academy of Sciences, Beijing, China
| | - Juan He
- grid.9227.e0000000119573309State Key Laboratory of Molecular Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China ,grid.410726.60000 0004 1797 8419University of Chinese Academy of Sciences, Beijing, China
| | - Kai Yuan
- grid.9227.e0000000119573309State Key Laboratory of Molecular Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China
| | - Chenyu Pan
- grid.24516.340000000123704535Department of General Surgery, Yangpu Hospital, Tongji University School of Medicine, Shanghai, China
| | - Moubin Lin
- grid.24516.340000000123704535Department of General Surgery, Yangpu Hospital, Tongji University School of Medicine, Shanghai, China
| | - Bin Zhou
- grid.9227.e0000000119573309State Key Laboratory of Molecular Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China
| | - Luonan Chen
- grid.9227.e0000000119573309State Key Laboratory of Molecular Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China
| | - Dong Gao
- grid.9227.e0000000119573309State Key Laboratory of Molecular Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China ,grid.9227.e0000000119573309Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
| | - Yun Zhao
- grid.9227.e0000000119573309State Key Laboratory of Molecular Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China ,grid.440637.20000 0004 4657 8879School of Life Science and Technology, ShanghaiTech University, Shanghai, China ,grid.410726.60000 0004 1797 8419Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, Zhejiang China
| |
Collapse
|
57
|
Jain I, Berg IC, Acharya A, Blaauw M, Gosstola N, Perez-Pinera P, Underhill GH. Delineating cooperative effects of Notch and biomechanical signals on patterned liver differentiation. Commun Biol 2022; 5:1073. [PMID: 36207581 PMCID: PMC9546876 DOI: 10.1038/s42003-022-03840-9] [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: 03/10/2022] [Accepted: 08/12/2022] [Indexed: 11/22/2022] Open
Abstract
Controlled in vitro multicellular culture systems with defined biophysical microenvironment have been used to elucidate the role of Notch signaling in the spatiotemporal regulation of stem and progenitor cell differentiation. In addition, computational models incorporating features of Notch ligand-receptor interactions have provided important insights into Notch pathway signaling dynamics. However, the mechanistic relationship between Notch-mediated intercellular signaling and cooperative microenvironmental cues is less clear. Here, liver progenitor cell differentiation patterning was used as a model to systematically evaluate the complex interplay of cellular mechanics and Notch signaling along with identifying combinatorial mechanisms guiding progenitor fate. We present an integrated approach that pairs a computational intercellular signaling model with defined microscale culture configurations provided within a cell microarray platform. Specifically, the cell microarray-based experiments were used to validate and optimize parameters of the intercellular Notch signaling model. This model incorporated the experimentally established multicellular dimensions of the cellular microarray domains, mechanical stress-related activation parameters, and distinct Notch receptor-ligand interactions based on the roles of the Notch ligands Jagged-1 and Delta-like-1. Overall, these studies demonstrate the spatial control of mechanotransduction-associated components, key growth factor and Notch signaling interactions, and point towards a possible role of E-Cadherin in translating intercellular mechanical gradients to downstream Notch signaling.
Collapse
Affiliation(s)
- Ishita Jain
- Department of Bioengineering, University of Illinois at Urbana Champaign, Urbana, USA
| | - Ian C Berg
- Department of Bioengineering, University of Illinois at Urbana Champaign, Urbana, USA
| | - Ayusha Acharya
- Department of Bioengineering, University of Illinois at Urbana Champaign, Urbana, USA
| | - Maddie Blaauw
- Department of Bioengineering, University of Illinois at Urbana Champaign, Urbana, USA
| | - Nicholas Gosstola
- Department of Bioengineering, University of Illinois at Urbana Champaign, Urbana, USA
| | - Pablo Perez-Pinera
- Department of Bioengineering, University of Illinois at Urbana Champaign, Urbana, USA
| | - Gregory H Underhill
- Department of Bioengineering, University of Illinois at Urbana Champaign, Urbana, USA.
| |
Collapse
|
58
|
Xu J, Kausalya PJ, Ong AGM, Goh CMF, Mohamed Ali S, Hunziker W. ZO-2/Tjp2 suppresses Yap and Wwtr1/Taz-mediated hepatocyte to cholangiocyte transdifferentiation in the mouse liver. NPJ Regen Med 2022; 7:55. [PMID: 36151109 PMCID: PMC9508083 DOI: 10.1038/s41536-022-00251-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Accepted: 09/02/2022] [Indexed: 01/11/2023] Open
Abstract
TJP2/ZO-2-inactivating mutations in humans cause progressive cholestatic liver disease. Liver-specific deletion of Tjp2 in the mouse (Tjp2 cKO mice) leads to mild progressive cholestasis without an overt degradation of the bile-blood barrier (BBB). These mice are more susceptible to cholic acid (CA) induced liver injury. Interestingly, while initially also more susceptible, Tjp2 cKO mice develop tolerance to a DDC-supplemented diet. The DDC diet induces an exacerbated ductular reaction in Tjp2 cKO mice, which arises from the transdifferentiation of hepatocytes to cholangiocytes. Consequently, this transdifferentiation is only observed if Tjp2 is inactivated in hepatocytes, but not if deleted in cholangiocytes. The DDC-diet-induced hepatocyte transdifferentiation in Tjp2 cKO mice requires Yap and Wwtr1/Taz, whose protein expression is upregulated in hepatocytes lacking Tjp2, but is independent of Notch2. Although inactivating Tjp2 is sufficient for the upregulation of Yap and Wwtr1/Taz protein, efficient transdifferentiation requires the DDC-diet insult. Thus, Tjp2 negatively regulates Yap/Taz-mediated transdifferentiation of hepatocytes to cholangiocytes in response to DDC-diet-induced liver injury. Furthermore, transdifferentiation is regulated at multiple levels and the type of injury inflicted on the Tjp2 deficient liver plays an important role in the resulting pathophysiology.
Collapse
Affiliation(s)
- Jianliang Xu
- Epithelial Polarity in Disease and Tissue Regeneration Laboratory, Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive Proteos, Singapore, 138673, Singapore.
| | - P Jaya Kausalya
- Epithelial Polarity in Disease and Tissue Regeneration Laboratory, Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive Proteos, Singapore, 138673, Singapore.,M Diagnostics Pte. Ltd. (MiRXES), 30 Biopolis Road, #09-05/06 Matrix, Singapore, 138671, Singapore
| | - Alicia Ghia Min Ong
- Epithelial Polarity in Disease and Tissue Regeneration Laboratory, Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive Proteos, Singapore, 138673, Singapore
| | - Christine Meng Fan Goh
- Epithelial Polarity in Disease and Tissue Regeneration Laboratory, Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive Proteos, Singapore, 138673, Singapore
| | - Safiah Mohamed Ali
- Epithelial Polarity in Disease and Tissue Regeneration Laboratory, Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive Proteos, Singapore, 138673, Singapore
| | - Walter Hunziker
- Epithelial Polarity in Disease and Tissue Regeneration Laboratory, Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive Proteos, Singapore, 138673, Singapore. .,Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, 2 Medical Drive MD9, Singapore, 117593, Singapore.
| |
Collapse
|
59
|
Dynamics of hepatocyte-cholangiocyte cell-fate decisions during liver development and regeneration. iScience 2022; 25:104955. [PMID: 36060070 PMCID: PMC9437857 DOI: 10.1016/j.isci.2022.104955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 05/17/2022] [Accepted: 08/12/2022] [Indexed: 11/25/2022] Open
Abstract
The immense regenerative potential of the liver is attributed to the ability of its two key cell types – hepatocytes and cholangiocytes – to trans-differentiate to one another either directly or through intermediate progenitor states. However, the dynamic features of decision-making between these cell-fates during liver development and regeneration remains elusive. Here, we identify a core gene regulatory network comprising c/EBPα, TGFBR2, and SOX9 which is multistable in nature, enabling three distinct cell states – hepatocytes, cholangiocytes, and liver progenitor cells (hepatoblasts/oval cells) – and stochastic switching among them. Predicted expression signature for these three states are validated through multiple bulk and single-cell transcriptomic datasets collected across developmental stages and injury-induced liver repair. This network can also explain the experimentally observed spatial organization of phenotypes in liver parenchyma and predict strategies for efficient cellular reprogramming. Our analysis elucidates how the emergent dynamics of underlying regulatory networks drive diverse cell-fate decisions in liver development and regeneration. Identified minimal regulatory network to model liver development and regeneration Changes in phenotypic landscapes by in-silico perturbations of regulatory networks Ability to explain physiological spatial patterning of liver cell types Decoded strategies for efficient reprogramming among liver cell phenotypes
Collapse
|
60
|
Annunziato S, Sun T, Tchorz JS. The RSPO-LGR4/5-ZNRF3/RNF43 module in liver homeostasis, regeneration, and disease. Hepatology 2022; 76:888-899. [PMID: 35006616 DOI: 10.1002/hep.32328] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 12/02/2021] [Accepted: 01/06/2022] [Indexed: 01/05/2023]
Abstract
WNT/β-catenin signaling plays pivotal roles during liver development, homeostasis, and regeneration. Likewise, its deregulation disturbs metabolic liver zonation and is responsible for the development of a large number of hepatic tumors. Liver fibrosis, which has become a major health burden for society and a hallmark of NASH, can also be promoted by WNT/β-catenin signaling. Upstream regulatory mechanisms controlling hepatic WNT/β-catenin activity may constitute targets for the development of novel therapies addressing these life-threatening conditions. The R-spondin (RSPO)-leucine-rich repeat-containing G protein-coupled receptor (LGR) 4/5-zinc and ring finger (ZNRF) 3/ring finger 43 (RNF43) module is fine-tuning WNT/β-catenin signaling in several tissues and is essential for hepatic WNT/β-catenin activity. In this review article, we recapitulate the role of the RSPO-LGR4/5-ZNRF3/RNF43 module during liver development, homeostasis, metabolic zonation, regeneration, and disease. We further discuss the controversy around LGR5 as a liver stem cell marker.
Collapse
Affiliation(s)
- Stefano Annunziato
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, Basel, Switzerland
| | - Tianliang Sun
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, Basel, Switzerland
| | - Jan S Tchorz
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, Basel, Switzerland
| |
Collapse
|
61
|
Laemmle A, Poms M, Hsu B, Borsuk M, Rüfenacht V, Robinson J, Sadowski MC, Nuoffer J, Häberle J, Willenbring H. Aquaporin 9 induction in human iPSC-derived hepatocytes facilitates modeling of ornithine transcarbamylase deficiency. Hepatology 2022; 76:646-659. [PMID: 34786702 PMCID: PMC9295321 DOI: 10.1002/hep.32247] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 10/30/2021] [Accepted: 11/14/2021] [Indexed: 12/16/2022]
Abstract
BACKGROUND AND AIMS Patient-derived human-induced pluripotent stem cells (hiPSCs) differentiated into hepatocytes (hiPSC-Heps) have facilitated the study of rare genetic liver diseases. Here, we aimed to establish an in vitro liver disease model of the urea cycle disorder ornithine transcarbamylase deficiency (OTCD) using patient-derived hiPSC-Heps. APPROACH AND RESULTS Before modeling OTCD, we addressed the question of why hiPSC-Heps generally secrete less urea than adult primary human hepatocytes (PHHs). Because hiPSC-Heps are not completely differentiated and maintain some characteristics of fetal PHHs, we compared gene-expression levels in human fetal and adult liver tissue to identify genes responsible for reduced urea secretion in hiPSC-Heps. We found lack of aquaporin 9 (AQP9) expression in fetal liver tissue as well as in hiPSC-Heps, and showed that forced expression of AQP9 in hiPSC-Heps restores urea secretion and normalizes the response to ammonia challenge by increasing ureagenesis. Furthermore, we proved functional ureagenesis by challenging AQP9-expressing hiPSC-Heps with ammonium chloride labeled with the stable isotope [15 N] (15 NH4 Cl) and by assessing enrichment of [15 N]-labeled urea. Finally, using hiPSC-Heps derived from patients with OTCD, we generated a liver disease model that recapitulates the hepatic manifestation of the human disease. Restoring OTC expression-together with AQP9-was effective in fully correcting OTC activity and normalizing ureagenesis as assessed by 15 NH4 Cl stable-isotope challenge. CONCLUSION Our results identify a critical role for AQP9 in functional urea metabolism and establish the feasibility of in vitro modeling of OTCD with hiPSC-Heps. By facilitating studies of OTCD genotype/phenotype correlation and drug screens, our model has potential for improving the therapy of OTCD.
Collapse
Affiliation(s)
- Alexander Laemmle
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell ResearchUniversity of California San FranciscoSan FranciscoCaliforniaUSA,Department of PediatricsUniversity Children's HospitalBernSwitzerland,University Institute of Clinical ChemistryUniversity of BernBernSwitzerland
| | - Martin Poms
- Division of Clinical Chemistry and BiochemistryUniversity Children’s Hospital ZurichZurichSwitzerland
| | - Bernadette Hsu
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell ResearchUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| | - Mariia Borsuk
- University Institute of Clinical ChemistryUniversity of BernBernSwitzerland
| | - Véronique Rüfenacht
- Division of Metabolism and Children`s Research CenterUniversity Children’s HospitalZurichSwitzerland
| | - Joshua Robinson
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell ResearchUniversity of California San FranciscoSan FranciscoCaliforniaUSA,Center for Reproductive SciencesUniversity of California San FranciscoSan FranciscoCaliforniaUSA,Department of Obstetrics, Gynecology, and Reproductive SciencesUniversity of California San FranciscoSan FranciscoCaliforniaUSA,Department of PediatricsMedical GeneticsUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| | | | - Jean‐Marc Nuoffer
- Department of PediatricsUniversity Children's HospitalBernSwitzerland,University Institute of Clinical ChemistryUniversity of BernBernSwitzerland
| | - Johannes Häberle
- Division of Metabolism and Children`s Research CenterUniversity Children’s HospitalZurichSwitzerland,Zurich Center for Integrative Human PhysiologyUniversity of ZurichZurichSwitzerland
| | - Holger Willenbring
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell ResearchUniversity of California San FranciscoSan FranciscoCaliforniaUSA,Department of SurgeryDivision of Transplant SurgeryUniversity of California San FranciscoSan FranciscoCaliforniaUSA,Liver CenterUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| |
Collapse
|
62
|
Liu G, Wei J, Li X, Tian M, Wang Z, Shen C, Sun W, Li C, Li X, Lv E, Tian S, Wang J, Xu S, Zhao B. Near-Infrared-Responded High Sensitivity Nanoprobe for Steady and Visualized Detection of Albumin in Hepatic Organoids and Mouse Liver. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2202505. [PMID: 35853243 PMCID: PMC9475548 DOI: 10.1002/advs.202202505] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 05/27/2022] [Indexed: 05/28/2023]
Abstract
Exploring the advanced techniques for protein detection facilitates cell fate investigation. However, it remains challenging to quantify and visualize the protein with one single probe. Here, a luminescent approach to detect hepatic cell fate marker albumin in vitro and living cell labeling with upconversion nanoparticles (UCNPs), which are conjugated with antibody (Ab) and rose bengal hexanoic acid (RBHA) is reported. To guarantee the detection quality and accuracy, an "OFF-ON" strategy is adopted: in the presence of albumin, the luminescence of nanoparticles remains suppressed owing to energy transfer to the quencher. Upon albumin binding to the antibody, the luminescence is recovered under near-infrared light. In various bio-samples, the UCNPs-Ab-RBHA (UCAR) nanoprobe can sense albumin with a broad detection range (5-315 ng mL-1 ). When applied to liver ductal organoid culture medium, the UCAR can monitor hepatocyte differentiation in real time by sensing the secreted albumin. Further, UCAR enables live imaging of cellular albumin in cells, organoids, and tissues. In a CCl4 -induced liver injury model, UCAR detects reduced albumin in liver tissue and serum. Thus, a biocompatible nanoprobe for both quantification and imaging of protein in complex biological environment with superior stability and high sensitivity is provided.
Collapse
Affiliation(s)
- Guofeng Liu
- Shandong Key Laboratory of BiophysicsInstitute of BiophysicsCollege of Physics and Electronic InformationDezhou UniversityDezhou253023China
| | - Jinsong Wei
- State Key Laboratory of Genetic EngineeringSchool of Life SciencesZhongshan HospitalFudan UniversityShanghai200438China
- Greater Bay Area Institute of Precision Medicine (Guangzhou)Fudan UniversityNansha DistrictGuangzhou511458China
| | - Xiaoyu Li
- State Key Laboratory of Genetic EngineeringSchool of Life SciencesZhongshan HospitalFudan UniversityShanghai200438China
| | - Meng Tian
- Shandong Key Laboratory of BiophysicsInstitute of BiophysicsCollege of Physics and Electronic InformationDezhou UniversityDezhou253023China
| | - Zhenxing Wang
- Shandong Key Laboratory of BiophysicsInstitute of BiophysicsCollege of Physics and Electronic InformationDezhou UniversityDezhou253023China
| | - Congcong Shen
- Shandong Key Laboratory of BiophysicsInstitute of BiophysicsCollege of Physics and Electronic InformationDezhou UniversityDezhou253023China
| | - Wan Sun
- Shandong Key Laboratory of BiophysicsInstitute of BiophysicsCollege of Physics and Electronic InformationDezhou UniversityDezhou253023China
| | - Chonghui Li
- Shandong Key Laboratory of BiophysicsInstitute of BiophysicsCollege of Physics and Electronic InformationDezhou UniversityDezhou253023China
| | - Xuewen Li
- State Key Laboratory of Genetic EngineeringSchool of Life SciencesZhongshan HospitalFudan UniversityShanghai200438China
| | - Enguang Lv
- Shandong Key Laboratory of BiophysicsInstitute of BiophysicsCollege of Physics and Electronic InformationDezhou UniversityDezhou253023China
| | - Shizheng Tian
- State Key Laboratory of Genetic EngineeringSchool of Life SciencesZhongshan HospitalFudan UniversityShanghai200438China
| | - Jihua Wang
- Shandong Key Laboratory of BiophysicsInstitute of BiophysicsCollege of Physics and Electronic InformationDezhou UniversityDezhou253023China
| | - Shicai Xu
- Shandong Key Laboratory of BiophysicsInstitute of BiophysicsCollege of Physics and Electronic InformationDezhou UniversityDezhou253023China
| | - Bing Zhao
- Shandong Key Laboratory of BiophysicsInstitute of BiophysicsCollege of Physics and Electronic InformationDezhou UniversityDezhou253023China
- State Key Laboratory of Genetic EngineeringSchool of Life SciencesZhongshan HospitalFudan UniversityShanghai200438China
| |
Collapse
|
63
|
Guo B, Friedland SC, Alexander W, Myers JA, Wang W, O'Dell MR, Getman M, Whitney-Miller CL, Agostini-Vulaj D, Huber AR, Mello SS, Vertino PM, Land HK, Steiner LA, Hezel AF. Arid1a mutation suppresses TGF-β signaling and induces cholangiocarcinoma. Cell Rep 2022; 40:111253. [PMID: 36044839 PMCID: PMC9808599 DOI: 10.1016/j.celrep.2022.111253] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 06/02/2022] [Accepted: 08/01/2022] [Indexed: 01/07/2023] Open
Abstract
Activating KRAS mutations and functional loss of members of the SWI/SNF complex, including ARID1A, are found together in the primary liver tumor cholangiocarcinoma (CC). How these mutations cooperate to promote CC has not been established. Using murine models of hepatocyte and biliary-specific lineage tracing, we show that Kras and Arid1a mutations drive the formation of CC and tumor precursors from the biliary compartment, which are accelerated by liver inflammation. Using cultured cells, we find that Arid1a loss causes cellular proliferation, escape from cell-cycle control, senescence, and widespread changes in chromatin structure. Notably, we show that the biliary proliferative response elicited by Kras/Arid1a cooperation and tissue injury in CC is caused by failed engagement of the TGF-β-Smad4 tumor suppressor pathway. We thus identify an ARID1A-TGF-β-Smad4 axis as essential in limiting the biliary epithelial response to oncogenic insults, while its loss leads to biliary pre-neoplasia and CC.
Collapse
Affiliation(s)
- Bing Guo
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY 14642, USA; Division of Hematology and Oncology, Department of Medicine, Wilmot Cancer Institute, University of Rochester Medical Center, 300 Elmwood Avenue, Rochester, NY 14642, USA
| | - Scott C Friedland
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - William Alexander
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Jacquelyn A Myers
- Genomics Research Center, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Wenjia Wang
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY 14642, USA; Division of Hematology and Oncology, Department of Medicine, Wilmot Cancer Institute, University of Rochester Medical Center, 300 Elmwood Avenue, Rochester, NY 14642, USA
| | - Michael R O'Dell
- Division of Hematology and Oncology, Department of Medicine, Wilmot Cancer Institute, University of Rochester Medical Center, 300 Elmwood Avenue, Rochester, NY 14642, USA
| | - Michael Getman
- Department of Pediatrics, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Christa L Whitney-Miller
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Diana Agostini-Vulaj
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Aaron R Huber
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Stephano S Mello
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Paula M Vertino
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Hartmut K Land
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY 14642, USA; Division of Hematology and Oncology, Department of Medicine, Wilmot Cancer Institute, University of Rochester Medical Center, 300 Elmwood Avenue, Rochester, NY 14642, USA
| | - Laurie A Steiner
- Department of Pediatrics, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Aram F Hezel
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY 14642, USA; Division of Hematology and Oncology, Department of Medicine, Wilmot Cancer Institute, University of Rochester Medical Center, 300 Elmwood Avenue, Rochester, NY 14642, USA.
| |
Collapse
|
64
|
Shu W, Yang M, Yang J, Lin S, Wei X, Xu X. Cellular crosstalk during liver regeneration: unity in diversity. Cell Commun Signal 2022; 20:117. [PMID: 35941604 PMCID: PMC9358812 DOI: 10.1186/s12964-022-00918-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 06/08/2022] [Indexed: 11/27/2022] Open
Abstract
The liver is unique in its ability to regenerate from a wide range of injuries and diseases. Liver regeneration centers around hepatocyte proliferation and requires the coordinated actions of nonparenchymal cells, including biliary epithelial cells, liver sinusoidal endothelial cells, hepatic stellate cells and kupffer cells. Interactions among various hepatocyte and nonparenchymal cells populations constitute a sophisticated regulatory network that restores liver mass and function. In addition, there are two different ways of liver regeneration, self-replication of liver epithelial cells and transdifferentiation between liver epithelial cells. The interactions among cell populations and regenerative microenvironment in the two modes are distinct. Herein, we first review recent advances in the interactions between hepatocytes and surrounding cells and among nonparenchymal cells in the context of liver epithelial cell self-replication. Next, we discuss the crosstalk of several cell types in the context of liver epithelial transdifferentiation, which is also crucial for liver regeneration. Video abstract
Collapse
Affiliation(s)
- Wenzhi Shu
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China.,Institute of Organ Transplantation, Zhejiang University, Hangzhou, 310003, China.,NHC Key Laboratory of Combined Multi-Organ Transplantation, Hangzhou, 310003, China.,Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, 310024, China.,Program in Clinical Medicine, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
| | - Mengfan Yang
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China.,Institute of Organ Transplantation, Zhejiang University, Hangzhou, 310003, China.,NHC Key Laboratory of Combined Multi-Organ Transplantation, Hangzhou, 310003, China.,Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, 310024, China
| | - Jiayin Yang
- Department of Liver Surgery and Liver Transplantation Center, West China Hospital of Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Shengda Lin
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China.,Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang University, Hangzhou, 310058, Zhejiang, China
| | - Xuyong Wei
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China. .,Institute of Organ Transplantation, Zhejiang University, Hangzhou, 310003, China. .,NHC Key Laboratory of Combined Multi-Organ Transplantation, Hangzhou, 310003, China. .,Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, 310024, China.
| | - Xiao Xu
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China. .,Institute of Organ Transplantation, Zhejiang University, Hangzhou, 310003, China. .,NHC Key Laboratory of Combined Multi-Organ Transplantation, Hangzhou, 310003, China. .,Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, 310024, China.
| |
Collapse
|
65
|
Kim J, Tomida K, Matsumoto T, Adachi T. Spheroid culture for chondrocytes triggers early stage of endochondral ossification. Biotechnol Bioeng 2022; 119:3311-3318. [PMID: 35923099 DOI: 10.1002/bit.28203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Revised: 07/07/2022] [Accepted: 07/30/2022] [Indexed: 11/11/2022]
Abstract
Endochondral ossification is the process of bone formation derived from growing cartilage during the development of the skeletal system. In previous studies, we have attempted to evoke the osteocyte differentiation of osteoblast precursor cells under a three-dimensional (3D) culture model. In order to recapitulate the endochondral ossification, the present study utilized the self-organized scaffold-free spheroid model reconstructed by pre-chondrocyte cells. Within 2-day cultivation in the absence of the chemically induced chondrogenesis supplements, the chondrocyte marker was greatly expressed in the inner region of the spheroid, whereas the hypertrophic chondrocyte marker was strongly detected in the surface region of the spheroid. Notably, we found out that the gene expression levels of osteocyte markers were also greatly up-regulated compared to the conventional 2D monolayer. Moreover, there was a hypertrophied morphologic change in the pre-chondrocyte spheroid from 4-day to 28-day cultivation. In this study, we highlighted the potentials of the 3D culture method to acquire the hypertrophic chondrocyte differentiation of the pre-chondrocyte cells to recapitulate the early stage of the endochondral ossification. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Jeonghyun Kim
- Department of Mechanical Systems Engineering, Nagoya University, Nagoya, 464-8603, Japan
| | - Kosei Tomida
- Department of Mechanical Systems Engineering, Nagoya University, Nagoya, 464-8603, Japan
| | - Takeo Matsumoto
- Department of Mechanical Systems Engineering, Nagoya University, Nagoya, 464-8603, Japan
| | - Taiji Adachi
- Institute for Life and Medical Sciences, Kyoto University, Kyoto, 606-8507, Japan
| |
Collapse
|
66
|
Wang W, Chen D, Wang J, Wen L. Cellular Homeostasis and Repair in the Biliary Tree. Semin Liver Dis 2022; 42:271-282. [PMID: 35672015 DOI: 10.1055/a-1869-7714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
During biliary tree homeostasis, BECs are largely in a quiescent state and their turnover is slow for maintaining normal tissue homeostasis. BTSCs continually replenish new BECs in the luminal surface of EHBDs. In response to various types of biliary injuries, distinct cellular sources, including HPCs, BTSCs, hepatocytes, and BECs, repair or regenerate the injured bile duct. BEC, biliary epithelial cell; BTSC, biliary tree stem/progenitor cell; EHBD, extrahepatic bile ducts; HPC, hepatic progenitor cell.The biliary tree comprises intrahepatic bile ducts and extrahepatic bile ducts lined with epithelial cells known as biliary epithelial cells (BECs). BECs are a common target of various cholangiopathies for which there is an unmet therapeutic need in clinical hepatology. The repair and regeneration of biliary tissue may potentially restore the normal architecture and function of the biliary tree. Hence, the repair and regeneration process in detail, including the replication of existing BECs, expansion and differentiation of the hepatic progenitor cells and biliary tree stem/progenitor cells, and transdifferentiation of the hepatocytes, should be understood. In this paper, we review biliary tree homeostasis, repair, and regeneration and discuss the feasibility of regenerative therapy strategies for cholangiopathy treatment.
Collapse
Affiliation(s)
- Wei Wang
- Department of Gastroenterology, Daping Hospital, Army Medical University, Chongqing, China
| | - Dongfeng Chen
- Department of Gastroenterology, Daping Hospital, Army Medical University, Chongqing, China
| | - Jun Wang
- Department of Gastroenterology, Daping Hospital, Army Medical University, Chongqing, China
| | - Liangzhi Wen
- Department of Gastroenterology, Daping Hospital, Army Medical University, Chongqing, China
| |
Collapse
|
67
|
Athavale D, Song Z, Desert R, Han H, Das S, Ge X, Komakula SSB, Chen W, Gao S, Lantvit D, Guzman G, Nieto N. Ablation of high-mobility group box-1 in the liver reduces hepatocellular carcinoma but causes hyperbilirubinemia in Hippo signaling-deficient mice. Hepatol Commun 2022; 6:2155-2169. [PMID: 35344292 PMCID: PMC9315122 DOI: 10.1002/hep4.1943] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 02/15/2022] [Accepted: 03/03/2022] [Indexed: 12/24/2022] Open
Abstract
Silencing the Hippo kinases mammalian sterile 20-like 1 and 2 (MST1/2) activates the transcriptional coactivator yes-associated protein (YAP) in human hepatocellular carcinoma (HCC). Hepatocyte-derived high-mobility group box-1 (HMGB1) regulates YAP expression; however, its contribution to HCC in the context of deregulated Hippo signaling is unknown. Here, we hypothesized that HMGB1 is required for hepatocarcinogenesis by activating YAP in Hippo signaling-deficient (Mst1/2ΔHep ) mice. Mst1/2ΔHep mice developed HCC within 3.5 months of age and had increased hepatic expression of HMGB1 and elevated YAP activity compared to controls. To understand the contribution of HMGB1, we generated Mst1/2&Hmgb1ΔHep mice. They exhibited decreased YAP activity, cell proliferation, inflammation, fibrosis, atypical ductal cell expansion, and HCC burden at 3.5 months compared to Mst1/2∆Hep mice. However, Mst1/2&Hmgb1ΔHep mice were smaller, developed hyperbilirubinemia, had more liver injury with intrahepatic biliary defects, and had reduced hemoglobin compared to Mst1/2ΔHep mice. Conclusion: Hepatic HMGB1 promotes hepatocarcinogenesis by regulation of YAP activity; nevertheless, it maintains intrahepatic bile duct physiology under Hippo signaling deficiency.
Collapse
Affiliation(s)
- Dipti Athavale
- Department of PathologyUniversity of Illinois at ChicagoChicagoIllinoisUSA
| | - Zhuolun Song
- Department of PathologyUniversity of Illinois at ChicagoChicagoIllinoisUSA
| | - Romain Desert
- Department of PathologyUniversity of Illinois at ChicagoChicagoIllinoisUSA
| | - Hui Han
- Department of PathologyUniversity of Illinois at ChicagoChicagoIllinoisUSA
| | - Sukanta Das
- Department of PathologyUniversity of Illinois at ChicagoChicagoIllinoisUSA
| | - Xiaodong Ge
- Department of PathologyUniversity of Illinois at ChicagoChicagoIllinoisUSA
| | | | - Wei Chen
- Department of PathologyUniversity of Illinois at ChicagoChicagoIllinoisUSA
| | - Shenglan Gao
- Department of PathologyUniversity of Illinois at ChicagoChicagoIllinoisUSA
| | - Daniel Lantvit
- Department of PathologyUniversity of Illinois at ChicagoChicagoIllinoisUSA
| | - Grace Guzman
- Department of PathologyUniversity of Illinois at ChicagoChicagoIllinoisUSA
| | - Natalia Nieto
- Department of PathologyUniversity of Illinois at ChicagoChicagoIllinoisUSA
- Division of Gastroenterology and HepatologyDepartment of MedicineUniversity of Illinois at ChicagoChicagoIllinoisUSA
- Research Biologist, Research & Development Service, Jesse Brown Veterans Affairs Medical CenterChicagoIllinoisUSA
| |
Collapse
|
68
|
Florentino RM, Li Q, Coard MC, Haep N, Motomura T, Diaz-Aragon R, Faccioli LAP, Amirneni S, Kocas-Kilicarslan ZN, Ostrowska A, Squires JE, Feranchak AP, Soto-Gutierrez A. Transmembrane channel activity in human hepatocytes and cholangiocytes derived from induced pluripotent stem cells. Hepatol Commun 2022; 6:1561-1573. [PMID: 35289126 PMCID: PMC9234678 DOI: 10.1002/hep4.1920] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 01/06/2022] [Accepted: 01/22/2022] [Indexed: 11/10/2022] Open
Abstract
The initial creation of human-induced pluripotent stem cells (iPSCs) set the foundation for the future of regenerative medicine. Human iPSCs can be differentiated into a variety of cell types in order to study normal and pathological molecular mechanisms. Currently, there are well-defined protocols for the differentiation, characterization, and establishment of functionality in human iPSC-derived hepatocytes (iHep) and iPSC-derived cholangiocytes (iCho). Electrophysiological study on chloride ion efflux channel activity in iHep and iCho cells has not been previously reported. We generated iHep and iCho cells and characterized them based on hepatocyte-specific and cholangiocyte-specific markers. The relevant transmembrane channels were selected: cystic fibrosis transmembrane conductance regulator, leucine rich repeat-containing 8 subunit A, and transmembrane member 16 subunit A. To measure the activity in these channels, we used whole-cell patch-clamp techniques with a standard intracellular and extracellular solution. Our iHep and iCho cells demonstrated definitive activity in the selected transmembrane channels, and this approach may become an important tool for investigating human liver biology of cholestatic diseases.
Collapse
Affiliation(s)
- Rodrigo M Florentino
- Department of PathologyUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA.,Pittsburgh Liver Research CenterUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Qin Li
- Department of PediatricsUniversity of Pittsburgh Medical CenterPittsburghPennsylvaniaUSA
| | - Michael C Coard
- Department of PathologyUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
| | - Nils Haep
- Department of PathologyUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
| | - Takashi Motomura
- Department of PathologyUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
| | - Ricardo Diaz-Aragon
- Department of PathologyUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
| | - Lanuza A P Faccioli
- Department of PathologyUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
| | - Sriram Amirneni
- Department of PathologyUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
| | | | - Alina Ostrowska
- Department of PathologyUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA.,Pittsburgh Liver Research CenterUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - James E Squires
- Pittsburgh Liver Research CenterUniversity of PittsburghPittsburghPennsylvaniaUSA.,Division of Gastroenterology, Hepatology, and NutritionUniversity of Pittsburgh Medical CenterPittsburghPennsylvaniaUSA
| | - Andrew P Feranchak
- Pittsburgh Liver Research CenterUniversity of PittsburghPittsburghPennsylvaniaUSA.,Department of PediatricsUniversity of Pittsburgh Medical CenterPittsburghPennsylvaniaUSA
| | - Alejandro Soto-Gutierrez
- Department of PathologyUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA.,Pittsburgh Liver Research CenterUniversity of PittsburghPittsburghPennsylvaniaUSA.,McGowan Institute for Regenerative MedicinePittsburghPennsylvaniaUSA
| |
Collapse
|
69
|
Huang R, Zhang X, Gracia-Sancho J, Xie WF. Liver regeneration: Cellular origin and molecular mechanisms. Liver Int 2022; 42:1486-1495. [PMID: 35107210 DOI: 10.1111/liv.15174] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 12/16/2021] [Accepted: 01/12/2022] [Indexed: 01/11/2023]
Abstract
The liver is known as an organ with high proliferation potential. Clarifying the cellular origin and deepening the understanding of liver regeneration mechanisms will help provide new directions for the treatment of liver disease. With the development and application of lineage tracing technology, the specific distribution and dynamic changes of hepatocyte subpopulations in homeostasis and liver injury have been illustrated. Self-replication of hepatocytes is responsible for the maintenance of liver function and mass under homeostasis. The compensatory proliferation of remaining hepatocytes is the main mechanism of liver regeneration following acute and chronic liver injury. Transdifferentiation between hepatocytes and cholangiocytes has been recognized upon severe chronic liver injury. Wnt/β-catenin, Hippo/YAP and Notch signalling play essential roles in the maintenance of homeostatic liver and hepatocyte-to-cholangiocyte conversion under liver injury. In this review, we summarized the recent studies on cell origin of newly generated hepatocytes and the underlying mechanisms of liver regeneration in homeostasis and liver injury.
Collapse
Affiliation(s)
- Ru Huang
- Department of Gastroenterology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Xin Zhang
- Department of Gastroenterology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Jordi Gracia-Sancho
- Liver Vascular Biology Research Group, Barcelona Hepatic Hemodynamic Unit, IDIBAPS, CIBEREHD, Barcelona, Spain
| | - Wei-Fen Xie
- Department of Gastroenterology, Changzheng Hospital, Naval Medical University, Shanghai, China
| |
Collapse
|
70
|
Pan Y, Gu Z, Lyu Y, Yang Y, Chung M, Pan X, Cai S. Link between senescence and cell fate: Senescence-associated secretory phenotype (SASP) and its effects on stem cell fate transition. Rejuvenation Res 2022; 25:160-172. [PMID: 35658548 DOI: 10.1089/rej.2022.0021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Senescence is a form of durable cell cycle arrest elicited in response to a wide range of stimuli. Senescent cells remain metabolically active and secrete a variety of factors collectively termed senescence-associated secretory phenotype (SASP). SASP is highly pleiotropic and can impact numerous biological processes in which it has both beneficial and deleterious roles. The underlying mechanisms by which SASP exerts its pleiotropic influence remain largely unknown. SASP serves as an environmental factor, which regulates stem cell differentiation and alters its routine. The latter can potentially be accomplished through dedifferentiation, transdifferentiation, or reprogramming. Behavioral changes that cells undergo when exposed to SASP are involved in several senescence-associated physiological and pathological phenomena. These findings provide clues for identifying possible interventions to reduce the deleterious effects without interfering in the beneficial outcomes. Here, we discuss the multifaced effects of SASP and the changes occurring in cellular states upon exposure to SASP factors.
Collapse
Affiliation(s)
- Yu Pan
- Shenzhen University, 47890, Shenzhen, Guangdong, China;
| | - Zhenzhen Gu
- Shenzhen University, 47890, Shenzhen, Guangdong, China;
| | - Yansi Lyu
- Shenzhen University, 47890, Shenzhen, Guangdong, China;
| | - Yi Yang
- Shenzhen University, 47890, Shenzhen, Guangdong, China;
| | - Manhon Chung
- Shanghai Jiao Tong University School of Medicine, 56694, Shanghai, China;
| | - Xiaohua Pan
- Shenzhen University, 47890, Shenzhen, Guangdong, China;
| | - Sa Cai
- Shenzhen University, 47890, 3688 Nanhai Avenue, Nanshan District, Shenzhen, Shenzhen, China, 518060;
| |
Collapse
|
71
|
Yuan RH, Hsu CL, Jhuang YL, Liu YR, Hsieh TH, Jeng YM. Tumor-matrix interaction induces phenotypic switching in liver cancer cells. Hepatol Int 2022; 16:562-576. [PMID: 35525880 DOI: 10.1007/s12072-022-10315-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2021] [Accepted: 02/13/2022] [Indexed: 01/16/2023]
Abstract
BACKGROUND Intrahepatic cholangiocarcinoma (ICC) is characterized by fibrous stroma and clinical behavior more aggressive than that of hepatocellular carcinoma (HCC). Scirrhous HCC is a subtype of HCC with fibrous stroma, frequently has partial cholangiocytic differentiation, and is more likely to have an aggressive behavior. This study explored the interaction of liver cancer cells with the extracellular matrix. METHODS AND RESULTS Liver cancer cells grown on collagen 1-coated plates showed upregulation of cholangiocytic marker expression but downregulation of hepatocytic marker expression. Three-dimensional sphere culture and Boyden chamber assay showed enhanced invasion and migration ability in collagen 1-conditioned liver cancer cells. Interaction with collagen 1 reduced liver cancer cell proliferation. RNA sequencing showed that in the liver cancer cells, collagen 1 upregulated cell cycle inhibitor expression and cell-matrix interaction, tumor migration, and angiogenesis pathways, but downregulated liver metabolic function pathways. Cholangiocytic differentiation and invasiveness induced by collagen 1 was mediated by the mitogen-activated protein kinase (MAPK) pathway, which was regulated by cell-matrix interaction-induced Src activation. Analysis of the Cancer Genome Atlas cohort showed that collagen 1 induced and suppressed genes were highly enriched in ICC and HCC, respectively. In HCC samples, collagen 1-regulated genes were strongly coexpressed and correlated with COL1A1 expression. CONCLUSIONS Liver cancer cell-matrix interaction induces cholangiocytic differentiation and switches liver cancer cells from a proliferative to an invasive phenotype through the Src/MAPK pathway, which may partly explain the differences in the behaviors of HCC and ICC.
Collapse
Affiliation(s)
- Ray-Hwang Yuan
- Department of Surgery, National Taiwan University Hospital, Taipei, Taiwan
- Department of Surgery, Hsinchu Branch, National Taiwan University Hospital, Hsinchu, Taiwan
| | - Chia-Lang Hsu
- Department of Medical Research, National Taiwan University Hospital, Taipei, Taiwan
| | - Yu-Lin Jhuang
- Graduate Institute of Pathology, National Taiwan University, Taipei, Taiwan
- Department of Pathology, National Taiwan University Hospital, Taipei, Taiwan
| | - Yun-Ru Liu
- Joint Biobank Office of Human Research, Taipei Medical University, Taipei, Taiwan
| | - Tsung-Han Hsieh
- Joint Biobank Office of Human Research, Taipei Medical University, Taipei, Taiwan
| | - Yung-Ming Jeng
- Graduate Institute of Pathology, National Taiwan University, Taipei, Taiwan.
- Department of Pathology, National Taiwan University Hospital, Taipei, Taiwan.
| |
Collapse
|
72
|
He Q, Cui L, Yuan X, Wang M, Hui L. Cell identity conversion in liver regeneration after injury. Curr Opin Genet Dev 2022; 75:101921. [PMID: 35644120 DOI: 10.1016/j.gde.2022.101921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 04/21/2022] [Accepted: 04/24/2022] [Indexed: 11/03/2022]
Abstract
Cell identity conversion in liver injury is the process that mature cells, specifically hepatocytes or cholangiocytes, convert into cells with other identities, which is found to play a pivotal role in liver regeneration. A better characterization of cell identity conversion will not only facilitate the understanding of liver tissue repair but also the development of novel regenerative therapies. In this review, we discuss the latest advances in cell identity conversion during liver regeneration, including conversions between hepatocytes and cholangiocytes and hepatocyte reprogramming to liver progenitor-like cells. To develop a unified description of cellular states in injury-related liver regeneration, we further propose the quantitative approach to explore cell identity conversion based on the Waddington's landscape.
Collapse
Affiliation(s)
- Qiang He
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Lei Cui
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Xiang Yuan
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Mengyao Wang
- School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China
| | - Lijian Hui
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China; School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China; School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China.
| |
Collapse
|
73
|
Ring NAR, Valdivieso K, Grillari J, Redl H, Ogrodnik M. The role of senescence in cellular plasticity: Lessons from regeneration and development and implications for age-related diseases. Dev Cell 2022; 57:1083-1101. [PMID: 35472291 DOI: 10.1016/j.devcel.2022.04.005] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 02/15/2022] [Accepted: 04/01/2022] [Indexed: 12/14/2022]
Abstract
Senescence is a cellular state which involves cell cycle arrest and a proinflammatory phenotype, and it has traditionally been associated with cellular and organismal aging. However, increasing evidence suggests key roles in tissue growth and regrowth, especially during development and regeneration. Conversely, cellular plasticity-the capacity of cells to undergo identity change, including differentiation and dedifferentiation-is associated with development and regeneration but is now being investigated in the context of age-related diseases such as Alzheimer disease. Here, we discuss the paradox of the role for cellular senescence in cellular plasticity: senescence can act as a cell-autonomous barrier and a paracrine driver of plasticity. We provide a conceptual framework for integrating recent data and use the interplay between cellular senescence and plasticity to provide insight into age-related diseases. Finally, we argue that age-related diseases can be better deciphered when senescence is recognized as a core mechanism of regeneration and development.
Collapse
Affiliation(s)
- Nadja Anneliese Ruth Ring
- Ludwig Boltzmann Research Group Senescence and Healing of Wounds, Vienna, Austria; Ludwig Boltzmann Institute for Traumatology, The Research Center in Cooperation with AUVA, Vienna, Austria; Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Karla Valdivieso
- Ludwig Boltzmann Research Group Senescence and Healing of Wounds, Vienna, Austria; Ludwig Boltzmann Institute for Traumatology, The Research Center in Cooperation with AUVA, Vienna, Austria; Austrian Cluster for Tissue Regeneration, Vienna, Austria; Institute of Molecular Biotechnology, Department of Biotechnology, University of Natural Resources and Life Sciences, Vienna, Austria
| | - Johannes Grillari
- Ludwig Boltzmann Institute for Traumatology, The Research Center in Cooperation with AUVA, Vienna, Austria; Austrian Cluster for Tissue Regeneration, Vienna, Austria; Institute of Molecular Biotechnology, Department of Biotechnology, University of Natural Resources and Life Sciences, Vienna, Austria
| | - Heinz Redl
- Ludwig Boltzmann Research Group Senescence and Healing of Wounds, Vienna, Austria; Ludwig Boltzmann Institute for Traumatology, The Research Center in Cooperation with AUVA, Vienna, Austria; Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Mikolaj Ogrodnik
- Ludwig Boltzmann Research Group Senescence and Healing of Wounds, Vienna, Austria; Ludwig Boltzmann Institute for Traumatology, The Research Center in Cooperation with AUVA, Vienna, Austria; Austrian Cluster for Tissue Regeneration, Vienna, Austria.
| |
Collapse
|
74
|
Russell JO, Camargo FD. Hippo signalling in the liver: role in development, regeneration and disease. Nat Rev Gastroenterol Hepatol 2022; 19:297-312. [PMID: 35064256 PMCID: PMC9199961 DOI: 10.1038/s41575-021-00571-w] [Citation(s) in RCA: 84] [Impact Index Per Article: 42.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/14/2021] [Indexed: 02/07/2023]
Abstract
The Hippo signalling pathway has emerged as a major player in many aspects of liver biology, such as development, cell fate determination, homeostatic function and regeneration from injury. The regulation of Hippo signalling is complex, with activation of the pathway by diverse upstream inputs including signals from cellular adhesion, mechanotransduction and crosstalk with other signalling pathways. Pathological activation of the downstream transcriptional co-activators yes-associated protein 1 (YAP) and transcriptional co-activator with PDZ-binding motif (TAZ, encoded by WWTR1), which are negatively regulated by Hippo signalling, has been implicated in multiple aspects of chronic liver disease, such as the development of liver fibrosis and tumorigenesis. Thus, development of pharmacological inhibitors of YAP-TAZ signalling has been an area of great interest. In this Review, we summarize the diverse roles of Hippo signalling in liver biology and highlight areas where outstanding questions remain to be investigated. Greater understanding of the mechanisms of Hippo signalling in liver function should help facilitate the development of novel therapies for the treatment of liver disease.
Collapse
Affiliation(s)
- Jacquelyn O Russell
- Stem Cell Program, Boston Children's Hospital, Boston, MA, USA
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
| | - Fernando D Camargo
- Stem Cell Program, Boston Children's Hospital, Boston, MA, USA.
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA.
| |
Collapse
|
75
|
Lan T, Qian S, Tang C, Gao J. Role of Immune Cells in Biliary Repair. Front Immunol 2022; 13:866040. [PMID: 35432349 PMCID: PMC9005827 DOI: 10.3389/fimmu.2022.866040] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Accepted: 03/08/2022] [Indexed: 02/06/2023] Open
Abstract
The biliary system is comprised of cholangiocytes and plays an important role in maintaining liver function. Under normal conditions, cholangiocytes remain in the stationary phase and maintain a very low turnover rate. However, the robust biliary repair is initiated in disease conditions, and different repair mechanisms can be activated depending on the pathological changes. During biliary disease, immune cells including monocytes, lymphocytes, neutrophils, and mast cells are recruited to the liver. The cellular interactions between cholangiocytes and these recruited immune cells as well as hepatic resident immune cells, including Kupffer cells, determine disease outcomes. However, the role of immune cells in the initiation, regulation, and suspension of biliary repair remains elusive. The cellular processes of cholangiocyte proliferation, progenitor cell differentiation, and hepatocyte-cholangiocyte transdifferentiation during biliary diseases are reviewed to manifest the underlying mechanism of biliary repair. Furthermore, the potential role of immune cells in crucial biliary repair mechanisms is highlighted. The mechanisms of biliary repair in immune-mediated cholangiopathies, inherited cholangiopathies, obstructive cholangiopathies, and cholangiocarcinoma are also summarized. Additionally, novel techniques that could clarify the underlying mechanisms of biliary repair are displayed. Collectively, this review aims to deepen the understanding of the mechanisms of biliary repair and contributes potential novel therapeutic methods for treating biliary diseases.
Collapse
Affiliation(s)
- Tian Lan
- Lab of Gastroenterology and Hepatology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China.,Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu, China
| | - Shuaijie Qian
- Lab of Gastroenterology and Hepatology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China.,Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu, China
| | - Chengwei Tang
- Lab of Gastroenterology and Hepatology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China.,Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu, China
| | - Jinhang Gao
- Lab of Gastroenterology and Hepatology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China.,Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
76
|
Lin Y, Zhang F, Zhang L, Chen L, Zheng S. Characteristics of SOX9-positive progenitor-like cells during cholestatic liver regeneration in biliary atresia. Stem Cell Res Ther 2022; 13:114. [PMID: 35313986 PMCID: PMC8935712 DOI: 10.1186/s13287-022-02795-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Accepted: 03/02/2022] [Indexed: 11/16/2022] Open
Abstract
Background The progression of Biliary Atresia (BA) is associated with the number of reactive ductular cells (RDCs) whose heterogeneity in origin and evolution in humans remains unknown. SOX9-positive liver progenitor-like cells (LPLCs) have been shown to participate in RDCs and new hepatocyte formation during cholestatic liver regeneration in an animal model, which implies the possibility that hepatocyte-reprogrammed LPLCs could be a source of RDCs in BA. The present study aimed to elucidate the characteristics of SOX9-positive LPLCs in BA for exploring new possible therapeutic targets by manipulating the bi-differentiation process of LPLCs to prevent disease progression. Methods Twenty-eight patients, including 24 patients with BA and 4 patients with Congenital Choledochal Cyst as the control group, were retrospectively recruited. Liver biopsy samples were classified histologically using a 4-point scale based on fibrosis severity. LPLCs were detected by SOX9 and HNF4A double positive staining. Single immunohistochemistry, double immunohistochemistry, and multiple immunofluorescence staining were used to determine the different cell types and characteristics of LPLCs. Results The prognostic predictors of BA, namely total bile acid (TBA), RDCs, and fibrosis, were correlated to the emergence of LPLCs. SOX9 and HNF4A double-positive LPLCs co-stained rarely with relevant markers of portal hepatic progenitor cells (portal-HPCs), including CK19, CK7, EPCAM, PROM1 (CD133), TROP2, and AFP. Under cholestasis conditions, LPLCs acquired superior proliferation and anti-senescence ability among hepatocytes. Moreover, LPLCs arranged as a pseudo-rosette structure appeared from the periportal parenchyma to the portal region, which implied the differentiation from hepatocyte-reprogrammed LPLCs to RDCs with the progression of cholestasis. Conclusions LPLCs are associated with disease progression and prognostic factors of BA. The bipotent characteristics of LPLCs are different from those of portal-HPCs. As cholestasis progresses, LPLCs appear to gain superior proliferation and anti-senescence ability and continually differentiate to RDCs. Supplementary Information The online version contains supplementary material available at 10.1186/s13287-022-02795-2.
Collapse
Affiliation(s)
- Yuting Lin
- Department of Pediatric Surgery, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai Key Laboratory of Birth Defect, and Key Laboratory of Neonatal Disease, Ministry of Health, 399 Wan Yuan Road, Shanghai, 201102, China
| | - Fang Zhang
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China
| | - Ludi Zhang
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China
| | - Lian Chen
- Department of Pathology, Children's Hospital of Fudan University, National Children's Medical Center, 399 Wan Yuan Road, Shanghai, 201102, China
| | - Shan Zheng
- Department of Pediatric Surgery, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai Key Laboratory of Birth Defect, and Key Laboratory of Neonatal Disease, Ministry of Health, 399 Wan Yuan Road, Shanghai, 201102, China.
| |
Collapse
|
77
|
Hallett JM, Ferreira-Gonzalez S, Man TY, Kilpatrick AM, Esser H, Thirlwell K, Macmillan MT, Rodrigo-Torres D, Dwyer BJ, Gadd VL, Ashmore-Harris C, Lu WY, Thomson JP, Jansen MA, O'Duibhir E, Starkey Lewis PJ, Campana L, Aird RE, Bate TSR, Fraser AR, Campbell JDM, Oniscu GC, Hay DC, Callanan A, Forbes SJ. Human biliary epithelial cells from discarded donor livers rescue bile duct structure and function in a mouse model of biliary disease. Cell Stem Cell 2022; 29:355-371.e10. [PMID: 35245467 PMCID: PMC8900617 DOI: 10.1016/j.stem.2022.02.006] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 09/20/2021] [Accepted: 02/09/2022] [Indexed: 12/14/2022]
Abstract
Biliary diseases can cause inflammation, fibrosis, bile duct destruction, and eventually liver failure. There are no curative treatments for biliary disease except for liver transplantation. New therapies are urgently required. We have therefore purified human biliary epithelial cells (hBECs) from human livers that were not used for liver transplantation. hBECs were tested as a cell therapy in a mouse model of biliary disease in which the conditional deletion of Mdm2 in cholangiocytes causes senescence, biliary strictures, and fibrosis. hBECs are expandable and phenotypically stable and help restore biliary structure and function, highlighting their regenerative capacity and a potential alternative to liver transplantation for biliary disease.
Collapse
Affiliation(s)
- John M Hallett
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, University of Edinburgh, 5 Little France Drive, Edinburgh EH16 4UU, UK
| | - Sofia Ferreira-Gonzalez
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, University of Edinburgh, 5 Little France Drive, Edinburgh EH16 4UU, UK
| | - Tak Yung Man
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, University of Edinburgh, 5 Little France Drive, Edinburgh EH16 4UU, UK
| | - Alastair M Kilpatrick
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, University of Edinburgh, 5 Little France Drive, Edinburgh EH16 4UU, UK
| | - Hannah Esser
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, University of Edinburgh, 5 Little France Drive, Edinburgh EH16 4UU, UK
| | - Kayleigh Thirlwell
- Tissues, Cells and Advanced Therapeutics Scottish National Blood and Transfusion Service (SNBTS), Research Avenue North, Edinburgh EH14 4BE, UK
| | - Mark T Macmillan
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, University of Edinburgh, 5 Little France Drive, Edinburgh EH16 4UU, UK
| | - Daniel Rodrigo-Torres
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, University of Edinburgh, 5 Little France Drive, Edinburgh EH16 4UU, UK
| | - Benjamin J Dwyer
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, University of Edinburgh, 5 Little France Drive, Edinburgh EH16 4UU, UK; Curtin Medical School, Curtin Health Innovation Research Institute, Curtin University, Kent St., Bentley, Perth 6102, Australia
| | - Victoria L Gadd
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, University of Edinburgh, 5 Little France Drive, Edinburgh EH16 4UU, UK
| | - Candice Ashmore-Harris
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, University of Edinburgh, 5 Little France Drive, Edinburgh EH16 4UU, UK
| | - Wei-Yu Lu
- Centre for Inflammation Research (CIR), University of Edinburgh, The Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh EH16 4TJ, UK
| | - John P Thomson
- Cancer Research UK Edinburgh Centre, MRC Institute of Genetics and Cancer, University of Edinburgh, Crewe Road, Edinburgh EH4 2XU, UK
| | - Maurits A Jansen
- Centre for Cardiovascular Science, The Queen's Medical Research Institute, University of Edinburgh, 47 Little France Crescent, Edinburgh EH16 4TJ, UK
| | - Eoghan O'Duibhir
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, University of Edinburgh, 5 Little France Drive, Edinburgh EH16 4UU, UK
| | - Philip J Starkey Lewis
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, University of Edinburgh, 5 Little France Drive, Edinburgh EH16 4UU, UK
| | - Lara Campana
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, University of Edinburgh, 5 Little France Drive, Edinburgh EH16 4UU, UK
| | - Rhona E Aird
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, University of Edinburgh, 5 Little France Drive, Edinburgh EH16 4UU, UK
| | - Thomas S R Bate
- Institute or Bioengineering, School of Engineering, University of Edinburgh, Faraday Building Colin Maclaurin Road, Edinburgh EH9 3DW, UK
| | - Alasdair R Fraser
- Tissues, Cells and Advanced Therapeutics Scottish National Blood and Transfusion Service (SNBTS), Research Avenue North, Edinburgh EH14 4BE, UK
| | - John D M Campbell
- Tissues, Cells and Advanced Therapeutics Scottish National Blood and Transfusion Service (SNBTS), Research Avenue North, Edinburgh EH14 4BE, UK
| | - Gabriel C Oniscu
- Edinburgh Transplant Centre, Royal Infirmary of Edinburgh, 51 Little France Crescent, Edinburgh EH16 4SA, UK; University of Edinburgh, 51 Little France Crescent, Edinburgh EH16 4SA, UK
| | - David C Hay
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, University of Edinburgh, 5 Little France Drive, Edinburgh EH16 4UU, UK
| | - Anthony Callanan
- Institute or Bioengineering, School of Engineering, University of Edinburgh, Faraday Building Colin Maclaurin Road, Edinburgh EH9 3DW, UK
| | - Stuart J Forbes
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, University of Edinburgh, 5 Little France Drive, Edinburgh EH16 4UU, UK.
| |
Collapse
|
78
|
Human biliary epithelial cells for regenerative medicine. Cell Stem Cell 2022; 29:345-347. [PMID: 35245462 DOI: 10.1016/j.stem.2022.02.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
In this issue of Cell Stem Cell, Hallett et al. demonstrate the therapeutic potential of human biliary epithelial cells (hBECs) purified from discarded donor livers in a murine model of biliary disease. hBEC transplantation helped to reduce injury and repaired biliary architecture, suggesting its clinical potential for chronic liver diseases.
Collapse
|
79
|
Zhao C, Lancman JJ, Yang Y, Gates KP, Cao D, Barske L, Matalonga J, Pan X, He J, Graves A, Huisken J, Chen C, Dong PDS. Intrahepatic cholangiocyte regeneration from an Fgf-dependent extrahepatic progenitor niche in a zebrafish model of Alagille Syndrome. Hepatology 2022; 75:567-583. [PMID: 34569629 PMCID: PMC8844142 DOI: 10.1002/hep.32173] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2021] [Revised: 09/20/2021] [Accepted: 09/22/2021] [Indexed: 02/05/2023]
Abstract
BACKGROUND AND AIMS Alagille Syndrome (ALGS) is a congenital disorder caused by mutations in the Notch ligand gene JAGGED1, leading to neonatal loss of intrahepatic duct (IHD) cells and cholestasis. Cholestasis can resolve in certain patients with ALGS, suggesting regeneration of IHD cells. However, the mechanisms driving IHD cell regeneration following Jagged loss remains unclear. Here, we show that cholestasis due to developmental loss of IHD cells can be consistently phenocopied in zebrafish with compound jagged1b and jagged2b mutations or knockdown. APPROACH AND RESULTS Leveraging the transience of jagged knockdown in juvenile zebrafish, we find that resumption of Jagged expression leads to robust regeneration of IHD cells through a Notch-dependent mechanism. Combining multiple lineage tracing strategies with whole-liver three-dimensional imaging, we demonstrate that the extrahepatic duct (EHD) is the primary source of multipotent progenitors that contribute to the regeneration, but not to the development, of IHD cells. Hepatocyte-to-IHD cell transdifferentiation is possible but rarely detected. Progenitors in the EHD proliferate and migrate into the liver with Notch signaling loss and differentiate into IHD cells if Notch signaling increases. Tissue-specific mosaic analysis with an inducible dominant-negative Fgf receptor suggests that Fgf signaling from the surrounding mesenchymal cells maintains this extrahepatic niche by directly preventing premature differentiation and allocation of EHD progenitors to the liver. Indeed, transcriptional profiling and functional analysis of adult mouse EHD organoids uncover their distinct differentiation and proliferative potential relative to IHD organoids. CONCLUSIONS Our data show that IHD cells regenerate upon resumption of Jagged/Notch signaling, from multipotent progenitors originating from an Fgf-dependent extrahepatic stem cell niche. We posit that if Jagged/Notch signaling is augmented, through normal stochastic variation, gene therapy, or a Notch agonist, regeneration of IHD cells in patients with ALGS may be enhanced.
Collapse
Affiliation(s)
- Chengjian Zhao
- Human Genetics Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California, USA
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Sichuan, People's Republic of China
| | - Joseph J Lancman
- Human Genetics Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California, USA
| | - Yi Yang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Sichuan, People's Republic of China
| | - Keith P Gates
- Human Genetics Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California, USA
| | - Dan Cao
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Sichuan, People's Republic of China
| | - Lindsey Barske
- Department of Pediatrics, College of Medicine & Division of Human Genetics, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, Ohio, USA
| | - Jonathan Matalonga
- Human Genetics Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California, USA
| | - Xiangyu Pan
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Sichuan, People's Republic of China
| | - Jiaye He
- Morgridge Institute for Research, Madison, Wisconsin, USA
| | - Alyssa Graves
- Morgridge Institute for Research, Madison, Wisconsin, USA
| | - Jan Huisken
- Morgridge Institute for Research, Madison, Wisconsin, USA
- Department of Integrative Biology, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Chong Chen
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Sichuan, People's Republic of China
| | - P Duc Si Dong
- Human Genetics Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California, USA
- Graduate School of Biomedical Sciences, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California, USA
| |
Collapse
|
80
|
Andersson ER. Outside influence: The extrahepatic duct as a source for bile duct regeneration. Hepatology 2022; 75:505-507. [PMID: 35006621 PMCID: PMC9305096 DOI: 10.1002/hep.32334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Accepted: 01/06/2022] [Indexed: 12/08/2022]
Affiliation(s)
- Emma R. Andersson
- Department of Cell and Molecular BiologyKarolinska InstitutetStockholmSweden
| |
Collapse
|
81
|
Liu Y, Zhuo S, Zhou Y, Ma L, Sun Z, Wu X, Wang XW, Gao B, Yang Y. Yap-Sox9 signaling determines hepatocyte plasticity and lineage-specific hepatocarcinogenesis. J Hepatol 2022; 76:652-664. [PMID: 34793870 PMCID: PMC8858854 DOI: 10.1016/j.jhep.2021.11.010] [Citation(s) in RCA: 44] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 10/14/2021] [Accepted: 11/04/2021] [Indexed: 12/29/2022]
Abstract
BACKGROUND & AIMS Primary liver tumors comprise distinct subtypes. A subset of intrahepatic cholangiocarcinoma (iCCA) can arise from cell fate reprogramming of mature hepatocytes in mouse models. However, the underpinning of cell fate plasticity during hepatocarcinogenesis is still poorly understood, hampering therapeutic development for primary liver cancer. As YAP activation induces liver tumor formation and cell fate plasticity, we investigated the role of Sox9, a transcription factor downstream of Yap activation that is expressed in biliary epithelial cells (BECs), in Yap-induced cell fate plasticity during hepatocarcinogenesis. METHODS To evaluate the function of Sox9 in YAP-induced hepatocarcinogenesis in vivo, we used several genetic mouse models of inducible hepatocyte-specific YAP activation with simultaneous Sox9 removal. Cell fate reprogramming was determined by lineage tracing and immunohistochemistry. The molecular mechanism underlying Yap and Sox9 function in hepatocyte plasticity was investigated by transcription and transcriptomic analyses of mouse and human liver tumors. RESULTS Sox9, a marker of liver progenitor cells (LPCs) and BECs, is differentially required in YAP-induced stepwise hepatocyte programming. While Sox9 has a limited role in hepatocyte dedifferentiation to LPCs, it is required for BEC differentiation from LPCs. YAP activation in Sox9-deficient hepatocytes resulted in more aggressive HCC with enhanced Yap activity at the expense of iCCA-like tumors. Furthermore, we showed that 20% of primary human liver tumors were associated with a YAP activation signature, and tumor plasticity is highly correlated with YAP activation and SOX9 expression. CONCLUSION Our data demonstrated that Yap-Sox9 signaling determines hepatocyte plasticity and tumor heterogeneity in hepatocarcinogenesis in both mouse and human liver tumors. We identified Sox9 as a critical transcription factor required for Yap-induced hepatocyte cell fate reprogramming during hepatocarcinogenesis. LAY SUMMARY Sox9, a marker of liver progenitor cells and bile duct lining cells, is a downstream target of YAP protein activation. Herein, we found that YAP activation in hepatocytes leads to a transition from mature hepatocytes to liver progenitor cells and then to bile duct lining cells. Sox9 is required in the second step during mouse hepatocarcinogenesis. We also found that human YAP and SOX9 may play similar roles in liver cancers.
Collapse
Affiliation(s)
- Yuchen Liu
- Department of Developmental Biology, Harvard School of Dental Medicine, 188 Longwood Ave. Boston, MA 02115, USA
| | - Shu Zhuo
- Department of Developmental Biology, Harvard School of Dental Medicine, 188 Longwood Ave. Boston, MA 02115, USA
| | - Yaxing Zhou
- Department of Developmental Biology, Harvard School of Dental Medicine, 188 Longwood Ave. Boston, MA 02115, USA
| | - Lichun Ma
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA
| | - Zhonghe Sun
- Cancer Research Technology Program, Frederick National Laboratory for Cancer, Leidos Biomedical Research, Inc., Frederick, MD, 21702, USA
| | - Xiaolin Wu
- Cancer Research Technology Program, Frederick National Laboratory for Cancer, Leidos Biomedical Research, Inc., Frederick, MD, 21702, USA
| | - Xin Wei Wang
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA
| | - Bin Gao
- Laboratory of Liver Diseases, National Institute on Alcohol Abuse and Alcoholism; National Institutes of Health, 5625 Fishers Lane, Room 2S-33, Bethesda, MD 20892, USA
| | - Yingzi Yang
- Department of Developmental Biology, Harvard School of Dental Medicine, 188 Longwood Ave. Boston, MA 02115, USA; Harvard Stem Cell Institute, Dana-Farber/Harvard Cancer Center, 188 Longwood Ave. Boston, MA 02115, USA; Program in Gastrointestinal Malignancies, Dana-Farber/Harvard Cancer Center, 188 Longwood Ave. Boston, MA 02115, USA.
| |
Collapse
|
82
|
Barhouse PS, Andrade MJ, Smith Q. Home Away From Home: Bioengineering Advancements to Mimic the Developmental and Adult Stem Cell Niche. FRONTIERS IN CHEMICAL ENGINEERING 2022. [DOI: 10.3389/fceng.2022.832754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The inherent self-organizing capacity of pluripotent and adult stem cell populations has advanced our fundamental understanding of processes that drive human development, homeostasis, regeneration, and disease progression. Translating these principles into in vitro model systems has been achieved with the advent of organoid technology, driving innovation to harness patient-specific, cell-laden regenerative constructs that can be engineered to augment or replace diseased tissue. While developmental organization and regenerative adult stem cell niches are tightly regulated in vivo, in vitro analogs lack defined architecture and presentation of physicochemical cues, leading to the unhindered arrangement of mini-tissues that lack complete physiological mimicry. This review aims to highlight the recent integrative engineering approaches that elicit spatio-temporal control of the extracellular niche to direct the structural and functional maturation of pluripotent and adult stem cell derivatives. While the advances presented here leverage multi-pronged strategies ranging from synthetic biology to microfabrication technologies, the methods converge on recreating the biochemical and biophysical milieu of the native tissue to be modeled or regenerated.
Collapse
|
83
|
Abstract
Cellular identity is established through complex layers of genetic regulation, forged over a developmental lifetime. An expanding molecular toolbox is allowing us to manipulate these gene regulatory networks in specific cell types in vivo. In principle, if we found the right molecular tricks, we could rewrite cell identity and harness the rich repertoire of possible cellular functions and attributes. Recent work suggests that this rewriting of cell identity is not only possible, but that newly induced cells can mitigate disease phenotypes in animal models of major human diseases. So, is the sky the limit, or do we need to keep our feet on the ground? This Spotlight synthesises key concepts emerging from recent efforts to reprogramme cellular identity in vivo. We provide our perspectives on recent controversies in the field of glia-to-neuron reprogramming and identify important gaps in our understanding that present barriers to progress.
Collapse
Affiliation(s)
- Sydney Leaman
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London SE1 1UL, UK.,MRC Centre for Neurodevelopmental Disorders, King's College London, London SE1 1UL, UK
| | - Nicolás Marichal
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London SE1 1UL, UK
| | - Benedikt Berninger
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London SE1 1UL, UK.,MRC Centre for Neurodevelopmental Disorders, King's College London, London SE1 1UL, UK.,Institute of Physiological Chemistry, University Medical Center of the Johannes Gutenberg University Mainz, Mainz 55128, Germany.,The Francis Crick Institute, London NW1 1AT, UK
| |
Collapse
|
84
|
Tanimizu N. The neonatal liver: Normal development and response to injury and disease. Semin Fetal Neonatal Med 2022; 27:101229. [PMID: 33745829 DOI: 10.1016/j.siny.2021.101229] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
The liver emerges from the ventral foregut endoderm around 3 weeks in human and 1 week in mice after fertilization. The fetal liver works as a hematopoietic organ and then develops functions required for performing various metabolic reactions in late fetal and neonatal periods. In parallel with functional differentiation, the liver establishes three dimensional tissue structures. In particular, establishment of the bile excretion system consisting of bile canaliculi of hepatocytes and bile ducts of cholangiocytes is critical to maintain healthy tissue status. This is because hepatocytes produce bile as they functionally mature, and if allowed to remain within the liver tissue can lead to cytotoxicity. In this review, we focus on epithelial tissue morphogenesis in the perinatal period and cholestatic liver diseases caused by abnormal development of the biliary system.
Collapse
Affiliation(s)
- Naoki Tanimizu
- Department of Tissue Development and Regeneration, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, S-1, W-17, Chuo-ku, Sapporo, 060-8556, Japan.
| |
Collapse
|
85
|
Abstract
Yes-associated protein 1 (YAP1) is a transcriptional coactivator that activates transcriptional enhanced associate domain transcription factors upon inactivation of the Hippo signaling pathway, to regulate biological processes like proliferation, survival, and differentiation. YAP1 is most prominently expressed in biliary epithelial cells (BECs) in normal adult livers and during development. In the current review, we will discuss the multiple roles of YAP1 in the development and morphogenesis of bile ducts inside and outside the liver, as well as in orchestrating the cholangiocyte repair response to biliary injury. We will review how biliary repair can occur through the process of hepatocyte-to-BEC transdifferentiation and how YAP1 is pertinent to this process. We will also discuss the liver's capacity for metabolic reprogramming as an adaptive mechanism in extreme cholestasis, such as when intrahepatic bile ducts are absent due to YAP1 loss from hepatic progenitors. Finally, we will discuss the roles of YAP1 in the context of pediatric pathologies afflicting bile ducts, such as Alagille syndrome and biliary atresia. In conclusion, we will comprehensively discuss the spatiotemporal roles of YAP1 in biliary development and repair after biliary injury while describing key interactions with other well-known developmental pathways.
Collapse
Affiliation(s)
- Laura Molina
- Division of Experimental Pathology, Department of Pathology, University of Pittsburgh School of Medicine
| | - Kari Nejak-Bowen
- Division of Experimental Pathology, Department of Pathology, University of Pittsburgh School of Medicine,Pittsburgh Liver Research Center, University of Pittsburgh and University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Satdarshan P. Monga
- Division of Experimental Pathology, Department of Pathology, University of Pittsburgh School of Medicine,Pittsburgh Liver Research Center, University of Pittsburgh and University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania,Division of Gastroenterology, Hepatology, and Nutrition, University of Pittsburgh and UPMC, Pittsburgh, Pennsylvania
| |
Collapse
|
86
|
Choudhury S, Asthana S, Homer-Vanniasinkam S, Chatterjee K. Emerging Trends in Biliary Stents: A Materials and Manufacturing Perspective. Biomater Sci 2022; 10:3716-3729. [DOI: 10.1039/d2bm00234e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Biliary stent technology has come a long way since its inception. There have been significant advancements in materials used, designs, and deployment strategies. Options have expanded from thermoplastic and metallic...
Collapse
|
87
|
Abstract
Hepatocytes are liver parenchymal cells involved in performing various metabolic reactions. During the development of therapeutic drugs, toxicological assays are conducted using hepatocyte cultures before clinical trials. However, since primary hepatocytes cannot proliferate and rapidly lose their functions in vitro, many efforts have been put into modifying culture conditions to expand primary hepatocytes and induce hepatocyte functions in intrinsic and extrinsic stem/progenitor cells. In this chapter, we summarize recent advances in preparing hepatocyte cultures and induction of hepatocytes from various cellular sources.
Collapse
Affiliation(s)
- Ayumu Okumura
- Division of Regenerative Medicine, Center for Stem Cell Biology and Regenerative Medicine, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Naoki Tanimizu
- Division of Regenerative Medicine, Center for Stem Cell Biology and Regenerative Medicine, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan.
- Department of Tissue Development and Regeneration, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan.
| |
Collapse
|
88
|
Denk H, Pabst D, Abuja PM, Reihs R, Tessaro B, Zatloukal K, Lackner C. Senescence markers in focal nodular hyperplasia of the liver: pathogenic considerations on the basis of immunohistochemical results. Mod Pathol 2022; 35:87-95. [PMID: 34645984 DOI: 10.1038/s41379-021-00940-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 09/23/2021] [Accepted: 09/23/2021] [Indexed: 02/07/2023]
Abstract
Focal nodular hyperplasia (FNH) is a polyclonal tumour-like hepatic lesion characterised by parenchymal nodules, connective tissue septa without interlobular bile ducts, pronounced ductular reaction and inflammation. It may represent a response to local arterial hyperperfusion and hyperoxygenation resulting in oxidative stress. We aimed at obtaining closer insight into the pathogenesis of FNH with its characteristic morphologic features. Immunohistochemistry and immunofluorescence microscopy was performed on FNH specimens using antibodies against keratins (K) 7 and 19, neural cell adhesion molecule (NCAM), lamin B1, senescence markers (CDK inhibitor 1/p21Cip1, CDK inhibitor /p16Ink4a, senescence-associated (SA) β- galactosidase activity), proliferation markers (Ki-67, proliferating-cell nuclear antigen (PCNA)), and the abnormally phosphorylated histone γ-H2AX, indicating DNA double strand breaks; moreover SA β- galactosidase activity was determined histochemically. Ductular metaplasia of hepatocytes indicated by K7 expression in the absence of K19 plays a major role in the development of ductular reaction in FNH. Moreover, the expression of senescence markers (p21Cip1, p16Ink4a, γ-H2AX, SA β-galactosidase activity) in hepatocytes and cholangiocytes suggests that stress-induced cellular senescence contributes to fibrosis and inflammation via production of components of the senescence-associated secretory phenotype. Expression of proliferation markers (Ki-67, PCNA) was not enhanced in hepatocytes and biliary cells. Senescence and ductular metaplasia of hepatocytes may thus be involved in inflammation, fibrosis and apoptosis resistance. Hence, fibrosis, inflammation and reduced apoptotic cell death, rather than proliferation (hyperplasia) may be responsible for increased tissue mass and tumour-like appearance of FNH.
Collapse
Affiliation(s)
- Helmut Denk
- Diagnostic & Research Centre of Molecular Biomedicine, Institute of Pathology, Medical University of Graz, Graz, Austria.
| | - Daniela Pabst
- Diagnostic & Research Centre of Molecular Biomedicine, Institute of Pathology, Medical University of Graz, Graz, Austria
| | - Peter M Abuja
- Diagnostic & Research Centre of Molecular Biomedicine, Institute of Pathology, Medical University of Graz, Graz, Austria
| | - Robert Reihs
- Diagnostic & Research Centre of Molecular Biomedicine, Institute of Pathology, Medical University of Graz, Graz, Austria
| | - Brigitte Tessaro
- Diagnostic & Research Centre of Molecular Biomedicine, Institute of Pathology, Medical University of Graz, Graz, Austria
| | - Kurt Zatloukal
- Diagnostic & Research Centre of Molecular Biomedicine, Institute of Pathology, Medical University of Graz, Graz, Austria
| | - Carolin Lackner
- Diagnostic & Research Centre of Molecular Biomedicine, Institute of Pathology, Medical University of Graz, Graz, Austria
| |
Collapse
|
89
|
Wang YC, Wang ZJ, Zhang C, Ning BF. Cell reprogramming in liver with potential clinical correlations. J Dig Dis 2022; 23:13-21. [PMID: 34921720 DOI: 10.1111/1751-2980.13072] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 12/13/2021] [Accepted: 12/15/2021] [Indexed: 12/11/2022]
Abstract
The theory of cell reprogramming has developed rapidly during the past decades. Cell reprogramming has been widely used in the construction of experimental models and cytotherapy for certain diseases. Hepatocyte-like cells that are important for the treatment of end-stage liver disease can now be obtained with a variety of reprogramming techniques. However, improving the differentiation status and physiological function of these cells remains challenging. Hepatocytes can transdifferentiate into other types of cells directly, whereas other types of cells can also transdifferentiate into hepatocyte-like cells both in vitro and in vivo. Moreover, cell reprogramming is to some extent similar to malignant cell transformation. During the initiation and progression of liver cancer, cell reprogramming is always associated with cancer metastasis and chemoresistance. In this review, we summarized the research related to cell reprogramming in liver and highlighted the potential effects of cell reprogramming in the pathogenesis and treatment of liver diseases.
Collapse
Affiliation(s)
- Yi Chuan Wang
- Clinical Cancer Institute, Center for Translational Medicine, Second Military Medical University, Shanghai, China
| | - Zhi Jie Wang
- Clinical Cancer Institute, Center for Translational Medicine, Second Military Medical University, Shanghai, China
| | - Cheng Zhang
- Department of Gastroenterology, Bethune International Peace Hospital, Shijiazhuang, Hebei Province, China
| | - Bei Fang Ning
- Department of Gastroenterology, Changzheng Hospital, Second Military Medical University, Shanghai, China
| |
Collapse
|
90
|
Hadjittofi C, Feretis M, Martin J, Harper S, Huguet E. Liver regeneration biology: Implications for liver tumour therapies. World J Clin Oncol 2021; 12:1101-1156. [PMID: 35070734 PMCID: PMC8716989 DOI: 10.5306/wjco.v12.i12.1101] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 06/22/2021] [Accepted: 11/28/2021] [Indexed: 02/06/2023] Open
Abstract
The liver has remarkable regenerative potential, with the capacity to regenerate after 75% hepatectomy in humans and up to 90% hepatectomy in some rodent models, enabling it to meet the challenge of diverse injury types, including physical trauma, infection, inflammatory processes, direct toxicity, and immunological insults. Current understanding of liver regeneration is based largely on animal research, historically in large animals, and more recently in rodents and zebrafish, which provide powerful genetic manipulation experimental tools. Whilst immensely valuable, these models have limitations in extrapolation to the human situation. In vitro models have evolved from 2-dimensional culture to complex 3 dimensional organoids, but also have shortcomings in replicating the complex hepatic micro-anatomical and physiological milieu. The process of liver regeneration is only partially understood and characterized by layers of complexity. Liver regeneration is triggered and controlled by a multitude of mitogens acting in autocrine, paracrine, and endocrine ways, with much redundancy and cross-talk between biochemical pathways. The regenerative response is variable, involving both hypertrophy and true proliferative hyperplasia, which is itself variable, including both cellular phenotypic fidelity and cellular trans-differentiation, according to the type of injury. Complex interactions occur between parenchymal and non-parenchymal cells, and regeneration is affected by the status of the liver parenchyma, with differences between healthy and diseased liver. Finally, the process of termination of liver regeneration is even less well understood than its triggers. The complexity of liver regeneration biology combined with limited understanding has restricted specific clinical interventions to enhance liver regeneration. Moreover, manipulating the fundamental biochemical pathways involved would require cautious assessment, for fear of unintended consequences. Nevertheless, current knowledge provides guiding principles for strategies to optimise liver regeneration potential.
Collapse
Affiliation(s)
- Christopher Hadjittofi
- University Department of Surgery, Addenbrookes Hospital, NIHR Comprehensive Biomedical Research and Academic Health Sciences Center, Cambridge University Hospitals NHS Foundation Trust, Cambridge CB2 0QQ, United Kingdom
| | - Michael Feretis
- University Department of Surgery, Addenbrookes Hospital, NIHR Comprehensive Biomedical Research and Academic Health Sciences Center, Cambridge University Hospitals NHS Foundation Trust, Cambridge CB2 0QQ, United Kingdom
| | - Jack Martin
- University Department of Surgery, Addenbrookes Hospital, NIHR Comprehensive Biomedical Research and Academic Health Sciences Center, Cambridge University Hospitals NHS Foundation Trust, Cambridge CB2 0QQ, United Kingdom
| | - Simon Harper
- University Department of Surgery, Addenbrookes Hospital, NIHR Comprehensive Biomedical Research and Academic Health Sciences Center, Cambridge University Hospitals NHS Foundation Trust, Cambridge CB2 0QQ, United Kingdom
| | - Emmanuel Huguet
- University Department of Surgery, Addenbrookes Hospital, NIHR Comprehensive Biomedical Research and Academic Health Sciences Center, Cambridge University Hospitals NHS Foundation Trust, Cambridge CB2 0QQ, United Kingdom
| |
Collapse
|
91
|
Tian L, Wang Y, Jang YY. Wnt signaling in biliary development, proliferation, and fibrosis. Exp Biol Med (Maywood) 2021; 247:360-367. [PMID: 34861115 DOI: 10.1177/15353702211061376] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Biliary fibrosis is an important pathological indicator of hepatobiliary damage. Cholangiocyte is the key cell type involved in this process. To reveal the pathogenesis of biliary fibrosis, it is essential to understand the normal development as well as the aberrant generation and proliferation of cholangiocytes. Numerous reports suggest that the Wnt signaling pathway is implicated in the physiological and pathological processes of cholangiocyte development and ductular reaction. In this review, we summarize the effects of Wnt pathway in cholangiocyte development from embryonic stem cells, as well as the underlying mechanisms of cholangiocyte responses to adult ductal damage. Wnt signaling pathway is regulated in a step-wise manner during each of the liver differentiation stages from embryonic stem cells to functional mature cholangiocytes. With the modulation of Wnt pathway, cholangiocytes can also be generated from adult liver progenitor cells and mature hepatocytes to repair liver damage. Non-canonical Wnt signaling is triggered in the active ductal cells during biliary fibrosis. Targeted control of the Wnt signaling may hold the great potential to reduce and/or reverse the biliary fibrogenic process.
Collapse
Affiliation(s)
- Lipeng Tian
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Yichen Wang
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Yoon Young Jang
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.,Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| |
Collapse
|
92
|
Lendahl U, Lui VCH, Chung PHY, Tam PKH. Biliary Atresia - emerging diagnostic and therapy opportunities. EBioMedicine 2021; 74:103689. [PMID: 34781099 PMCID: PMC8604670 DOI: 10.1016/j.ebiom.2021.103689] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 10/15/2021] [Accepted: 10/28/2021] [Indexed: 02/06/2023] Open
Abstract
Biliary Atresia is a devastating pediatric cholangiopathy affecting the bile ducts of the liver. In this review, we describe recent progress in the understanding of liver development with a focus on cholangiocyte differentiation and how use of technical platforms, including rodent, zebrafish and organoid models, advances our understanding of Biliary Atresia. This is followed by a description of potential pathomechanisms, such as autoimmune responses, inflammation, disturbed apical-basal cell polarity, primary cilia dysfunction as well as beta-amyloid accumulation. Finally, we describe current and emerging diagnostic opportunities and recent translation breakthroughs for Biliary Atresia in the area of emerging therapy development, including immunomodulation and organoid-based systems for liver and bile duct repair.
Collapse
Affiliation(s)
- Urban Lendahl
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden; Dr. Li Dak-Sum Research Centre, the University of Hong Kong, Hong Kong.
| | - Vincent C H Lui
- Dr. Li Dak-Sum Research Centre, the University of Hong Kong, Hong Kong; Department of Surgery, Li Ka Shing Faculty of Medicine, the University of Hong Kong, Hong Kong
| | - Patrick H Y Chung
- Department of Surgery, Li Ka Shing Faculty of Medicine, the University of Hong Kong, Hong Kong; Department of Surgery, University of Hong Kong-Shenzhen Hospital, Shenzhen, Guangdong, China
| | - Paul K H Tam
- Dr. Li Dak-Sum Research Centre, the University of Hong Kong, Hong Kong; Department of Surgery, Li Ka Shing Faculty of Medicine, the University of Hong Kong, Hong Kong; Department of Surgery, University of Hong Kong-Shenzhen Hospital, Shenzhen, Guangdong, China.
| |
Collapse
|
93
|
Ogawa M, Jiang JX, Xia S, Yang D, Ding A, Laselva O, Hernandez M, Cui C, Higuchi Y, Suemizu H, Dorrell C, Grompe M, Bear CE, Ogawa S. Generation of functional ciliated cholangiocytes from human pluripotent stem cells. Nat Commun 2021; 12:6504. [PMID: 34764255 PMCID: PMC8586142 DOI: 10.1038/s41467-021-26764-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Accepted: 10/21/2021] [Indexed: 12/15/2022] Open
Abstract
The derivation of mature functional cholangiocytes from human pluripotent stem cells (hPSCs) provides a model for studying the pathogenesis of cholangiopathies and for developing therapies to treat them. Current differentiation protocols are not efficient and give rise to cholangiocytes that are not fully mature, limiting their therapeutic applications. Here, we generate functional hPSC-derived cholangiocytes that display many characteristics of mature bile duct cells including high levels of cystic fibrosis transmembrane conductance regulator (CFTR) and the presence of primary cilia capable of sensing flow. With this level of maturation, these cholangiocytes are amenable for testing the efficacy of cystic fibrosis drugs and for studying the role of cilia in cholangiocyte development and function. Transplantation studies show that the mature cholangiocytes generate ductal structures in the liver of immunocompromised mice indicating that it may be possible to develop cell-based therapies to restore bile duct function in patients with biliary disease.
Collapse
Affiliation(s)
- Mina Ogawa
- grid.231844.80000 0004 0474 0428McEwen Stem Cell Institute, University Health Network, Toronto, ON Canada
| | - Jia-Xin Jiang
- grid.42327.300000 0004 0473 9646Programme in Molecular Medicine, Research Institute, Hospital for Sick Children, Toronto, ON Canada
| | - Sunny Xia
- grid.42327.300000 0004 0473 9646Programme in Molecular Medicine, Research Institute, Hospital for Sick Children, Toronto, ON Canada
| | - Donghe Yang
- grid.231844.80000 0004 0474 0428McEwen Stem Cell Institute, University Health Network, Toronto, ON Canada
| | - Avrilynn Ding
- grid.231844.80000 0004 0474 0428McEwen Stem Cell Institute, University Health Network, Toronto, ON Canada
| | - Onofrio Laselva
- grid.42327.300000 0004 0473 9646Programme in Molecular Medicine, Research Institute, Hospital for Sick Children, Toronto, ON Canada
| | - Marcela Hernandez
- grid.231844.80000 0004 0474 0428McEwen Stem Cell Institute, University Health Network, Toronto, ON Canada
| | - Changyi Cui
- grid.231844.80000 0004 0474 0428McEwen Stem Cell Institute, University Health Network, Toronto, ON Canada
| | - Yuichiro Higuchi
- grid.452212.20000 0004 0376 978XCentral Institute for Experimental Animals, Kawasaki, Kanagawa Japan
| | - Hiroshi Suemizu
- grid.452212.20000 0004 0376 978XCentral Institute for Experimental Animals, Kawasaki, Kanagawa Japan
| | - Craig Dorrell
- grid.5288.70000 0000 9758 5690Oregon Stem Cell Center, Oregon Health and Science University, Portland, OR USA
| | - Markus Grompe
- grid.5288.70000 0000 9758 5690Oregon Stem Cell Center, Oregon Health and Science University, Portland, OR USA
| | - Christine E. Bear
- grid.42327.300000 0004 0473 9646Programme in Molecular Medicine, Research Institute, Hospital for Sick Children, Toronto, ON Canada ,grid.17063.330000 0001 2157 2938Department of Physiology, University of Toronto, Toronto, ON Canada ,grid.17063.330000 0001 2157 2938Department of Biochemistry, University of Toronto, Toronto, ON Canada
| | - Shinichiro Ogawa
- McEwen Stem Cell Institute, University Health Network, Toronto, ON, Canada. .,Ajmera Transplant Centre, Toronto General Research Institute, University Health Network, Toronto, ON, Canada. .,Department of Surgery, Shinshu University School of Medicine, Matsumoto, Nagano, Japan. .,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
94
|
Liang R, Lin YH, Zhu H. Genetic and Cellular Contributions to Liver Regeneration. Cold Spring Harb Perspect Biol 2021; 14:a040832. [PMID: 34750173 PMCID: PMC9438780 DOI: 10.1101/cshperspect.a040832] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
The regenerative capabilities of the liver represent a paradigm for understanding tissue repair in solid organs. Regeneration after partial hepatectomy in rodent models is well understood, while regeneration in the context of clinically relevant chronic injuries is less studied. Given the growing incidence of fatty liver disease, cirrhosis, and liver cancer, interest in liver regeneration is increasing. Here, we will review the principles, genetics, and cell biology underlying liver regeneration, as well as new approaches being used to study heterogeneity in liver tissue maintenance and repair.
Collapse
Affiliation(s)
- Roger Liang
- Children's Research Institute, Departments of Pediatrics and Internal Medicine, Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| | - Yu-Hsuan Lin
- Children's Research Institute, Departments of Pediatrics and Internal Medicine, Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| | - Hao Zhu
- Children's Research Institute, Departments of Pediatrics and Internal Medicine, Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| |
Collapse
|
95
|
Sánchez PS, Rigual MDM, Djouder N. Inflammatory and Non-Inflammatory Mechanisms Controlling Cirrhosis Development. Cancers (Basel) 2021; 13:cancers13205045. [PMID: 34680192 PMCID: PMC8534267 DOI: 10.3390/cancers13205045] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Revised: 10/01/2021] [Accepted: 10/03/2021] [Indexed: 12/28/2022] Open
Abstract
Simple Summary The liver is continuously exposed to several harmful factors, subsequently activating sophisticated mechanisms set-up in order to repair and regenerate the damaged liver and hence to prevent its failure. When the injury becomes chronic, the regenerative response becomes perpetual and goes awry, leading to cirrhosis with a fatal liver dysfunction. Cirrhosis is a well-known risk factor for hepatocellular carcinoma (HCC), the most common, usually lethal, human primary liver neoplasm with very limited therapeutic options. Considering the pivotal role of immune factors in the development of cirrhosis, here we review and discuss the inflammatory pathways and components implicated in the development of cirrhosis. A better understanding of these circuits would help the design of novel strategies to prevent and treat cirrhosis and HCC, two lethal diseases. Abstract Because the liver is considered to be one of the most important metabolic organs in the body, it is continuously exposed to damaging environmental agents. Upon damage, several complex cellular and molecular mechanisms in charge of liver recovery and regeneration are activated to prevent the failure of the organ. When liver injury becomes chronic, the regenerative response goes awry and impairs the liver function, consequently leading to cirrhosis, a liver disorder that can cause patient death. Cirrhosis has a disrupted liver architecture and zonation, along with the presence of fibrosis and parenchymal nodules, known as regenerative nodules (RNs). Inflammatory cues contribute to the cirrhotic process in response to chronic damaging agents. Cirrhosis can progress to HCC, the most common and one of the most lethal liver cancers with unmet medical needs. Considering the essential role of inflammatory pathways in the development of cirrhosis, further understanding of the relationship between immune cells and the activation of RNs and fibrosis would guide the design of innovative therapeutic strategies to ameliorate the survival of cirrhotic and HCC patients. In this review, we will summarize the inflammatory mechanisms implicated in the development of cirrhosis.
Collapse
Affiliation(s)
| | | | - Nabil Djouder
- Correspondence: ; Tel.: +34-3-491-732-8000 (ext. 3830); Fax: +34-3-491-224-6914
| |
Collapse
|
96
|
Li B, Wang Y, Pelz C, Moss J, Shemer R, Dor Y, Akkari YK, Canady PS, Naugler WE, Orloff S, Grompe M. In vitro expansion of cirrhosis derived liver epithelial cells with defined small molecules. Stem Cell Res 2021; 56:102523. [PMID: 34601385 DOI: 10.1016/j.scr.2021.102523] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 07/30/2021] [Accepted: 08/24/2021] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND & AIMS Mature hepatocytes have limited expansion capability in culture and rapidly loose key functions. Recently however, tissue culture conditions have been developed that permit rodent hepatocytes to proliferate and transform into progenitor-like cells with ductal characteristics in vitro. Analogous cells expressing both hepatic and duct markers can be found in human cirrhotic liver in vivo and may represent an expandable population. METHODS An in vitro culture system to expand epithelial cells from human end stage liver disease organs was developed by inhibiting the canonical TGF-β, Hedgehog and BMP pathways. RESULTS Human cirrhotic liver epithelial cells became highly proliferative in vitro. Both gene expression and DNA methylation site analyses revealed that cirrhosis derived epithelial liver cells were intermediate between normal hepatocytes and cholangiocytes. Mouse hepatocytes could be expanded under the same conditions and retained the ability to re-differentiate into hepatocytes upon transplantation. In contrast, human cirrhotic liver derived cells had only low re-differentiation capacity. CONCLUSIONS Epithelial cells of intermediate ductal-hepatocytic phenotype can be isolated from human cirrhotic livers and expanded in vitro. Unlike their murine counterparts they have limited liver repopulation potential.
Collapse
Affiliation(s)
- Bin Li
- Oregon Stem Cell Center, USA; Department of Pediatrics, Papé Family Institute, Oregon Health & Science University, Portland, OR, USA
| | - Yuhan Wang
- Oregon Stem Cell Center, USA; Department of Pediatrics, Papé Family Institute, Oregon Health & Science University, Portland, OR, USA
| | - Carl Pelz
- Oregon Stem Cell Center, USA; Department of Pediatrics, Papé Family Institute, Oregon Health & Science University, Portland, OR, USA
| | - Josh Moss
- Department of Developmental Biology and Cancer Research, The Institute for Medical Research Israel-Canada, The Hebrew University-Hadassah Medical School, Israel
| | - Ruth Shemer
- Department of Developmental Biology and Cancer Research, The Institute for Medical Research Israel-Canada, The Hebrew University-Hadassah Medical School, Israel
| | - Yuval Dor
- Department of Developmental Biology and Cancer Research, The Institute for Medical Research Israel-Canada, The Hebrew University-Hadassah Medical School, Israel
| | - Yassmine K Akkari
- Cytogenetics Services and Molecular Pathology, Legacy Health, Portland, OR, USA
| | - Pamela S Canady
- Oregon Stem Cell Center, USA; Department of Pediatrics, Papé Family Institute, Oregon Health & Science University, Portland, OR, USA
| | - Willscott E Naugler
- Oregon Stem Cell Center, USA; School of Medicine, Division of Gastroenterology and Hepatology, Oregon Health & Science University, Portland, OR, USA
| | - Susan Orloff
- School of Medicine, Division of Gastroenterology and Hepatology, Oregon Health & Science University, Portland, OR, USA
| | - Markus Grompe
- Oregon Stem Cell Center, USA; Department of Pediatrics, Papé Family Institute, Oregon Health & Science University, Portland, OR, USA.
| |
Collapse
|
97
|
Genomic characterization of rare molecular subclasses of hepatocellular carcinoma. Commun Biol 2021; 4:1150. [PMID: 34608257 PMCID: PMC8490450 DOI: 10.1038/s42003-021-02674-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Accepted: 09/15/2021] [Indexed: 12/24/2022] Open
Abstract
Primary liver cancer, consisting of both cholangiocarcinoma (CCA) and hepatocellular carcinoma (HCC), is the second leading cause of cancer deaths worldwide. Our goal is to genomically characterize rare HCC subclasses to provide insight into disease biology. Leveraging The Cancer Genome Atlas (TCGA) to perform a combined analysis of CCA (n = 36) and HCC (n = 275), we integrated multiple genomic platforms, to assess transcriptional profiles, mutational signatures, and copy number patterns to uncover underlying etiology and linage specific patterns. We identified two molecular classes distinct from prototypical HCC tumors. The first, CCA-Like, although histologically indistinguishable from HCC, had enrichment of CCA mutations (IDH1, BAP1), mutational signatures, and transcriptional patterns (SOX9, KRT19). CCA-Like, however, retained a copy number landscape similar to HCC, suggesting a hepatocellular linage. The second, Blast-Like, is enriched in TP53 mutations, HBV infection, exposure related mutational signatures and transcriptionally similar to hepatoblasts. Although these subclasses are molecularly distinct, they both have a worse progression-free survival compared to classical HCC tumors, yet are clinically treated the same. The identification of and characterization of CCA-Like and Blast-Like subclasses advance our knowledge of HCC as well as represents an urgent need for the identification of class specific biomarkers and targeted therapy. Jeffrey Damrauer, Markia Smith et al. used existing datasets from cholangiocarcinoma (CCA) and hepatocellular carcinoma (HCC) to characterize two subsets of HCC distinct from prototypical HCC tumors, based on comprehensive analysis of molecular data. The two classes differed from HCC by their copy number, gene expression and mutational signature and exhibited worse progression free survival, highlighting the need to identify class-specific biomarkers and develop targeted therapies for these forms of cancer.
Collapse
|
98
|
McCarron S, Bathon B, Conlon DM, Abbey D, Rader DJ, Gawronski K, Brown CD, Olthoff KM, Shaked A, Raabe TD. Functional Characterization of Organoids Derived From Irreversibly Damaged Liver of Patients With NASH. Hepatology 2021; 74:1825-1844. [PMID: 33901295 DOI: 10.1002/hep.31857] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 03/01/2021] [Accepted: 03/21/2021] [Indexed: 12/11/2022]
Abstract
BACKGROUND AND AIMS NASH will soon become the leading cause of liver transplantation in the United States and is also associated with increased COVID-19 mortality. Currently, there are no Food and Drug Administration-approved drugs available that slow NASH progression or address NASH liver involvement in COVID-19. Because animal models cannot fully recapitulate human NASH, we hypothesized that stem cells isolated directly from end-stage liver from patients with NASH may address current knowledge gaps in human NASH pathology. APPROACH AND RESULTS We devised methods that allow the derivation, proliferation, hepatic differentiation, and extensive characterization of bipotent ductal organoids from irreversibly damaged liver from patients with NASH. The transcriptomes of organoids derived from NASH liver, but not healthy liver, show significant up-regulation of proinflammatory and cytochrome p450-related pathways, as well as of known liver fibrosis and tumor markers, with the degree of up-regulation being patient-specific. Functionally, NASH liver organoids exhibit reduced passaging/growth capacity and hallmarks of NASH liver, including decreased albumin production, increased free fatty acid-induced lipid accumulation, increased sensitivity to apoptotic stimuli, and increased cytochrome P450 metabolism. After hepatic differentiation, NASH liver organoids exhibit reduced ability to dedifferentiate back to the biliary state, consistent with the known reduced regenerative ability of NASH livers. Intriguingly, NASH liver organoids also show strongly increased permissiveness to severe acute respiratory syndrome-coronavirus 2 (SARS-CoV-2) vesicular stomatitis pseudovirus as well as up-regulation of ubiquitin D, a known inhibitor of the antiviral interferon host response. CONCLUSION Expansion of primary liver stem cells/organoids derived directly from irreversibly damaged liver from patients with NASH opens up experimental avenues for personalized disease modeling and drug development that has the potential to slow human NASH progression and to counteract NASH-related SARS-CoV-2 effects.
Collapse
Affiliation(s)
- Sarah McCarron
- Department of Medicine, Division of Translational Medicine and Human Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Brooke Bathon
- Department of Medicine, Division of Translational Medicine and Human Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Donna M Conlon
- Department of Medicine, Division of Translational Medicine and Human Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Deepti Abbey
- Department of Medicine, Division of Translational Medicine and Human Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Daniel J Rader
- Department of Medicine, Division of Translational Medicine and Human Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Katerina Gawronski
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Christopher D Brown
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Kim M Olthoff
- Department of Surgery, Division of Transplant Surgery, Hospital of the University of Pennsylvania, Philadelphia, PA
| | - Abraham Shaked
- Department of Surgery, Division of Transplant Surgery, Hospital of the University of Pennsylvania, Philadelphia, PA
| | - Tobias D Raabe
- Department of Medicine, Division of Translational Medicine and Human Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| |
Collapse
|
99
|
Peng WC, Kraaier LJ, Kluiver TA. Hepatocyte organoids and cell transplantation: What the future holds. Exp Mol Med 2021; 53:1512-1528. [PMID: 34663941 PMCID: PMC8568948 DOI: 10.1038/s12276-021-00579-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 01/11/2021] [Accepted: 01/14/2021] [Indexed: 12/29/2022] Open
Abstract
Historically, primary hepatocytes have been difficult to expand or maintain in vitro. In this review, we will focus on recent advances in establishing hepatocyte organoids and their potential applications in regenerative medicine. First, we provide a background on the renewal of hepatocytes in the homeostatic as well as the injured liver. Next, we describe strategies for establishing primary hepatocyte organoids derived from either adult or fetal liver based on insights from signaling pathways regulating hepatocyte renewal in vivo. The characteristics of these organoids will be described herein. Notably, hepatocyte organoids can adopt either a proliferative or a metabolic state, depending on the culture conditions. Furthermore, the metabolic gene expression profile can be modulated based on the principles that govern liver zonation. Finally, we discuss the suitability of cell replacement therapy to treat different types of liver diseases and the current state of cell transplantation of in vitro-expanded hepatocytes in mouse models. In addition, we provide insights into how the regenerative microenvironment in the injured host liver may facilitate donor hepatocyte repopulation. In summary, transplantation of in vitro-expanded hepatocytes holds great potential for large-scale clinical application to treat liver diseases.
Collapse
Affiliation(s)
- Weng Chuan Peng
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, 3584 CS, Utrecht, The Netherlands.
| | - Lianne J Kraaier
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, 3584 CS, Utrecht, The Netherlands
| | - Thomas A Kluiver
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, 3584 CS, Utrecht, The Netherlands
| |
Collapse
|
100
|
Martinez Lyons A, Boulter L. The developmental origins of Notch-driven intrahepatic bile duct disorders. Dis Model Mech 2021; 14:dmm048413. [PMID: 34549776 PMCID: PMC8480193 DOI: 10.1242/dmm.048413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
The Notch signaling pathway is an evolutionarily conserved mechanism of cell-cell communication that mediates cellular proliferation, cell fate specification, and maintenance of stem and progenitor cell populations. In the vertebrate liver, an absence of Notch signaling results in failure to form bile ducts, a complex tubular network that radiates throughout the liver, which, in healthy individuals, transports bile from the liver into the bowel. Loss of a functional biliary network through congenital malformations during development results in cholestasis and necessitates liver transplantation. Here, we examine to what extent Notch signaling is necessary throughout embryonic life to initiate the proliferation and specification of biliary cells and concentrate on the animal and human models that have been used to define how perturbations in this signaling pathway result in developmental liver disorders.
Collapse
Affiliation(s)
| | - Luke Boulter
- MRC Human Genetics Unit, Institute of Genetics and Cancer, Edinburgh EH4 2XU, UK
| |
Collapse
|