101
|
Wang P, Cong M, Liu T, Xu H, Wang L, Sun G, Yang A, Zhang D, Huang J, Sun Y, Zhao W, Ma H, Jia J, You H. Inhibitory effects of HNF4α on migration/maltransformation of hepatic progenitors: HNF4α-overexpressing hepatic progenitors for liver repopulation. Stem Cell Res Ther 2017; 8:183. [PMID: 28807057 PMCID: PMC5557474 DOI: 10.1186/s13287-017-0629-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Revised: 06/22/2017] [Accepted: 07/10/2017] [Indexed: 12/29/2022] Open
Abstract
Background Although they are expandable in vitro, hepatic progenitors are immature cells and share many immunomarkers with hepatocellular carcinoma, raising potential concerns regarding maltransformation after transplantation. This study investigated the effects of hepatic nuclear factor (HNF) 4α on the proliferation, migration, and maltransformation of hepatic progenitors and determined the feasibility of using these manipulated cells for transplantation. Methods The effects of HNF4α on rat hepatic progenitors (i.e. hepatic oval cells) were analyzed by HNF4α overexpression and HNF4α shRNA. Nonobese diabetic/severe combined immunodeficiency (NOD/SCID) mice injured by carbon chloride (CCl4) were then transplanted with control, HNF4α-overexpressing or HNF4α-suppressing hepatic oval cells. Finally, the engraftment of these cells in the recipient liver was analyzed. Results Rat hepatic progenitors (i.e. hepatic oval cells) expressed HNF4α, although less than that in hepatocytes. When HNF4α was overexpressed in these cells, the proliferation and migration of hepatic oval cells were reduced; but when HNF4α was suppressed by shRNA, the proliferation and migration, and even anchorage-independent growth, of these cells were accelerated. RNA microarray and gene functional analysis revealed that suppressing HNF4α not only impaired many biosynthesis and metabolism pathways of hepatocytes but also increased pathways for cancer. When transplanted into CCl4-injured NOD/SCID mice, few HNF4α-suppressing hepatic oval cells localized into the liver, while control cells and HNF4α-overexpressing cells engrafted into the liver and differentiated into albumin-positive hepatocytes. Interestingly, the hepatocytes derived from HNF4α-overexpressing cells were less migrative and expressed less c-Myc than the cells derived from control cells. Conclusion HNF4α constrains proliferation, migration, and maltransformation of hepatic progenitors, and HNF4α-overexpressing hepatic progenitors serve as an optimal candidate for cell transplantation. Electronic supplementary material The online version of this article (doi:10.1186/s13287-017-0629-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Ping Wang
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing Key Laboratory of Translational Medicine on Liver Cirrhosis & National Clinical Research Center of Digestive Diseases, Beijing, 100050, China.,Municipal Laboratory for Liver Protection and Regulation of Regeneration, Capital Medical University, Beijing, 100069, China
| | - Min Cong
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing Key Laboratory of Translational Medicine on Liver Cirrhosis & National Clinical Research Center of Digestive Diseases, Beijing, 100050, China
| | - Tianhui Liu
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing Key Laboratory of Translational Medicine on Liver Cirrhosis & National Clinical Research Center of Digestive Diseases, Beijing, 100050, China
| | - Hufeng Xu
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing Key Laboratory of Translational Medicine on Liver Cirrhosis & National Clinical Research Center of Digestive Diseases, Beijing, 100050, China
| | - Lin Wang
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing Key Laboratory of Translational Medicine on Liver Cirrhosis & National Clinical Research Center of Digestive Diseases, Beijing, 100050, China
| | - Guangyong Sun
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing Key Laboratory of Translational Medicine on Liver Cirrhosis & National Clinical Research Center of Digestive Diseases, Beijing, 100050, China
| | - Aiting Yang
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing Key Laboratory of Translational Medicine on Liver Cirrhosis & National Clinical Research Center of Digestive Diseases, Beijing, 100050, China
| | - Dong Zhang
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing Key Laboratory of Translational Medicine on Liver Cirrhosis & National Clinical Research Center of Digestive Diseases, Beijing, 100050, China
| | - Jian Huang
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing Key Laboratory of Translational Medicine on Liver Cirrhosis & National Clinical Research Center of Digestive Diseases, Beijing, 100050, China
| | - Yameng Sun
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing Key Laboratory of Translational Medicine on Liver Cirrhosis & National Clinical Research Center of Digestive Diseases, Beijing, 100050, China
| | - Wenshan Zhao
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing Key Laboratory of Translational Medicine on Liver Cirrhosis & National Clinical Research Center of Digestive Diseases, Beijing, 100050, China
| | - Hong Ma
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing Key Laboratory of Translational Medicine on Liver Cirrhosis & National Clinical Research Center of Digestive Diseases, Beijing, 100050, China
| | - Jidong Jia
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing Key Laboratory of Translational Medicine on Liver Cirrhosis & National Clinical Research Center of Digestive Diseases, Beijing, 100050, China. .,Municipal Laboratory for Liver Protection and Regulation of Regeneration, Capital Medical University, Beijing, 100069, China.
| | - Hong You
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing Key Laboratory of Translational Medicine on Liver Cirrhosis & National Clinical Research Center of Digestive Diseases, Beijing, 100050, China. .,Municipal Laboratory for Liver Protection and Regulation of Regeneration, Capital Medical University, Beijing, 100069, China.
| |
Collapse
|
102
|
Abstract
Liver regeneration is a fascinating and complex process with many medical implications. An important component of this regenerative process is the hepatic progenitor cell (HPC). These appealing cells are able to participate in the renewal of hepatocytes and cholangiocytes when the normal homeostatic regeneration is exhausted. Moreover, the HPC niche is of vital importance toward the activation, differentiation, and proliferation of the HPC. This niche provides a rich microenvironment for the regulation of the HPC, thanks to the intercellular secretion of molecules. New findings indicate that the regenerative possibilities in the liver could provide a diverse basis for therapeutic targets.
Collapse
Affiliation(s)
- Matthias Van Haele
- Liver Research Unit, Department of Imaging and Pathology, KU Leuven and University Hospitals Leuven, Minderbroederstraat 12, 3000 Leuven, Belgium
| | - Tania Roskams
- Liver Research Unit, Department of Imaging and Pathology, KU Leuven and University Hospitals Leuven, Minderbroederstraat 12, 3000 Leuven, Belgium.
| |
Collapse
|
103
|
Ji H, Lu Y, Shi Y. Seeds in the liver. Acta Histochem 2017; 119:349-356. [PMID: 28389020 DOI: 10.1016/j.acthis.2017.03.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Revised: 02/28/2017] [Accepted: 03/21/2017] [Indexed: 02/05/2023]
Abstract
The liver is a crucial organ for homeostasis and has a tremendous self-renewal and regenerative capacity. It has long been believed that the self-renewal and repair of the liver within a given physiological condition or its repopulation in chronic liver diseases, when hepatocyte proliferation is impaired, will primarily be conducted by the proliferating duct cells, termed "oval cells" or hepatic progenitor cells (HPCs). In addition, numerous studies have revealed that HPCs are the initial tumor cells of liver cancer under certain micro-environments. However, benefit from the extensive application of lineage tracing strategies using the Cre/LoxP system, researchers have redefined the fate of these bipotential cells, raising obvious controversies regarding the capacity of liver cells to control their own biology and differentiation. Here, we review the relevant articles, focusing on cell-lineage tracing to better understanding seed cells and their distinct fate in the liver.
Collapse
|
104
|
Wang Y, Huang X, He L, Pu W, Li Y, Liu Q, Li Y, Zhang L, Yu W, Zhao H, Zhou Y, Zhou B. Genetic tracing of hepatocytes in liver homeostasis, injury, and regeneration. J Biol Chem 2017; 292:8594-8604. [PMID: 28377509 DOI: 10.1074/jbc.m117.782029] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Revised: 03/27/2017] [Indexed: 12/20/2022] Open
Abstract
The liver possesses a remarkable capacity to regenerate after damage. There is a heated debate on the origin of new hepatocytes after injuries in adult liver. Hepatic stem/progenitor cells have been proposed to produce functional hepatocytes after injury. Recent studies have argued against this model and suggested that pre-existing hepatocytes, rather than stem cells, contribute new hepatocytes. This hepatocyte-to-hepatocyte model is mainly based on labeling of hepatocytes with Cre-recombinase delivered by the adeno-associated virus. However, the impact of virus infection on cell fate determination, consistency of infection efficiency, and duration of Cre-virus in hepatocytes remain confounding factors that interfere with the data interpretation. Here, we generated a new genetic tool Alb-DreER to label almost all hepatocytes (>99.5%) and track their contribution to different cell lineages in the liver. By "pulse-and-chase" strategy, we found that pre-existing hepatocytes labeled by Alb-DreER contribute to almost all hepatocytes during normal homeostasis and after liver injury. Virtually all hepatocytes in the injured liver are descendants of pre-existing hepatocytes through self-expansion. We concluded that stem cell differentiation is unlikely to be responsible for the generation of a substantial number of new hepatocytes in adult liver. Our study also provides a new mouse tool for more precise in vivo genetic study of hepatocytes in the field.
Collapse
Affiliation(s)
- Yue Wang
- From The 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.,the Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - XiuZhen Huang
- From The 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.,the Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Lingjuan He
- From The 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.,the Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Wenjuan Pu
- From The 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.,the Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Yan Li
- From The 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.,the Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Qiaozhen Liu
- From The 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.,the Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Yi Li
- From The 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.,the Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Libo Zhang
- From The 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.,the Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Wei Yu
- From The 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.,the Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Huan Zhao
- From The 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.,the Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Yingqun Zhou
- the Department of Gastroenterology, Shanghai 10th People's Hospital, Tongji University School of Medicine, Shanghai 200072, China,
| | - Bin Zhou
- From The 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, .,the Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China.,the Key Laboratory of Regenerative Medicine of Ministry of Education, Institute of Aging and Regenerative Medicine, Jinan University, Guangzhou 510632, China, and.,the School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| |
Collapse
|
105
|
Lukacs-Kornek V, Lammert F. The progenitor cell dilemma: Cellular and functional heterogeneity in assistance or escalation of liver injury. J Hepatol 2017; 66:619-630. [PMID: 27826058 DOI: 10.1016/j.jhep.2016.10.033] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Revised: 10/18/2016] [Accepted: 10/31/2016] [Indexed: 12/16/2022]
Abstract
Liver progenitor cells (LPCs) are quiescent cells that are activated during liver injury and thought to give rise to hepatocytes and cholangiocytes in order to support liver regeneration and tissue restitution. While hepatocytes are capable of self-renewal, during most chronic injuries the proliferative capacity of hepatocytes is inhibited, thus LPCs provide main source for regeneration. Despite extensive lineage tracing studies, their role and involvement in these processes are often controversial. Additionally, increasing evidence suggests that the LPC compartment consists of heterogeneous cell populations that are actively involved in cellular interactions with myeloid and lymphoid cells during regeneration. On the other hand, LPC expansion has been associated with an increased fibrogenic response, raising concerns about the therapeutic use of these cells. This review aims to summarize the current understanding of the identity, the cellular interactions and the key pathways affecting the biology of LPCs. Understanding the regulatory circuits and the specific role of LPCs is especially important as it could provide novel therapeutic platforms for the treatment of liver inflammation, fibrosis and regeneration.
Collapse
Affiliation(s)
- Veronika Lukacs-Kornek
- Department of Medicine II, Saarland University Medical Center, Saarland University, Homburg, Germany.
| | - Frank Lammert
- Department of Medicine II, Saarland University Medical Center, Saarland University, Homburg, Germany
| |
Collapse
|
106
|
Abstract
Despite decades of basic research, biliary diseases remain prevalent, highly morbid, and notoriously difficult to treat. We have, however, dramatically increased our understanding of biliary developmental biology, cholangiocyte pathophysiology, and the endogenous mechanisms of biliary regeneration and repair. All of this complex and rapidly evolving knowledge coincides with an explosion of new technological advances in the area of regenerative medicine. New breakthroughs such as induced pluripotent stem cells and organoid culture are increasingly being applied to the biliary system; it is only a matter of time until new regenerative therapeutics for the cholangiopathies are unveiled. In this review, the authors integrate what is known about biliary development, regeneration, and repair, and link these conceptual advances to the technological breakthroughs that are collectively driving the emergence of a new global field in biliary regenerative medicine.
Collapse
Affiliation(s)
- Thiago M. De Assuncao
- Division of Gastroenterology and Hepatology, Mayo Clinic and Foundation, Rochester, MN,Gastroenterology Research Unit, Mayo Clinic and Foundation, Rochester, MN
| | - Nidhi Jalan-Sakrikar
- Division of Gastroenterology and Hepatology, Mayo Clinic and Foundation, Rochester, MN,Gastroenterology Research Unit, Mayo Clinic and Foundation, Rochester, MN
| | - Robert C. Huebert
- Division of Gastroenterology and Hepatology, Mayo Clinic and Foundation, Rochester, MN,Gastroenterology Research Unit, Mayo Clinic and Foundation, Rochester, MN,Center for Cell Signaling in Gastroenterology; Mayo Clinic and Foundation, Rochester, MN
| |
Collapse
|
107
|
Li XY, Yang X, Zhao QD, Han ZP, Liang L, Pan XR, Zhu JN, Li R, Wu MC, Wei LX. Lipopolysaccharide promotes tumorigenicity of hepatic progenitor cells by promoting proliferation and blocking normal differentiation. Cancer Lett 2017; 386:35-46. [DOI: 10.1016/j.canlet.2016.10.044] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2016] [Revised: 10/27/2016] [Accepted: 10/28/2016] [Indexed: 12/19/2022]
|
108
|
Cellular Mechanisms of Liver Regeneration and Cell-Based Therapies of Liver Diseases. BIOMED RESEARCH INTERNATIONAL 2017; 2017:8910821. [PMID: 28210629 PMCID: PMC5292184 DOI: 10.1155/2017/8910821] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/13/2016] [Revised: 11/29/2016] [Accepted: 12/27/2016] [Indexed: 12/14/2022]
Abstract
The emerging field of regenerative medicine offers innovative methods of cell therapy and tissue/organ engineering as a novel approach to liver disease treatment. The ultimate scientific foundation of both cell therapy of liver diseases and liver tissue and organ engineering is delivered by the in-depth studies of the cellular and molecular mechanisms of liver regeneration. The cellular mechanisms of the homeostatic and injury-induced liver regeneration are unique. Restoration of the mass of liver parenchyma is achieved by compensatory hypertrophy and hyperplasia of the differentiated parenchymal cells, hepatocytes, while expansion and differentiation of the resident stem/progenitor cells play a minor or negligible role. Participation of blood-borne cells of the bone marrow origin in liver parenchyma regeneration has been proven but does not exceed 1-2% of newly formed hepatocytes. Liver regeneration is activated spontaneously after injury and can be further stimulated by cell therapy with hepatocytes, hematopoietic stem cells, or mesenchymal stem cells. Further studies aimed at improving the outcomes of cell therapy of liver diseases are underway. In case of liver failure, transplantation of engineered liver can become the best option in the foreseeable future. Engineering of a transplantable liver or its major part is an enormous challenge, but rapid progress in induced pluripotency, tissue engineering, and bioprinting research shows that it may be doable.
Collapse
|
109
|
A label-retaining but unipotent cell population resides in biliary compartment of mammalian liver. Sci Rep 2017; 7:40322. [PMID: 28084309 PMCID: PMC5234023 DOI: 10.1038/srep40322] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Accepted: 12/05/2016] [Indexed: 12/17/2022] Open
Abstract
Cells with slow proliferation kinetics that retain the nuclear label over long time periods-the label-retaining cells (LRCs)-represent multipotent stem cells in a number of adult tissues. Since the identity of liver LRCs (LLRCs) had remained elusive we utilized a genetic approach to reveal LLRCs in normal non-injured livers and characterized their regenerative properties in vivo and in culture. We found that LLRCs were located in biliary vessels and participated in the regeneration of biliary but not hepatocyte injury. In culture experiments the sorted LLRCs displayed an enhanced self-renewal capacity but a unipotent biliary differentiation potential. Transcriptome analysis revealed a unique set of tumorigenesis- and nervous system-related genes upregulated in LLRCs when compared to non-LRC cholangiocytes. We conclude that the LLRCs established during the normal morphogenesis of the liver do not represent a multipotent primitive somatic stem cell population but act as unipotent biliary progenitor cells.
Collapse
|
110
|
de Jonge J, Olthoff KM. Liver regeneration. BLUMGART'S SURGERY OF THE LIVER, BILIARY TRACT AND PANCREAS, 2-VOLUME SET 2017:93-109.e7. [DOI: 10.1016/b978-0-323-34062-5.00006-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
111
|
Infliximab and Dexamethasone Attenuate the Ductular Reaction in Mice. Sci Rep 2016; 6:36586. [PMID: 27824131 PMCID: PMC5100545 DOI: 10.1038/srep36586] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Accepted: 10/17/2016] [Indexed: 12/16/2022] Open
Abstract
Chronic hepatic injury is accompanied by a ductular response that is strongly correlated with disease severity and progression of fibrosis. To investigate whether anti-inflammatory drugs can modulate the ductular response, we treated mice suffering from a steatotic or cholestatic injury with anti-TNF-α antibodies (Infliximab) or glucocorticoids (Dexamethasone). We discovered that Dexamethasone and Infliximab can both modulate the adaptive remodeling of the biliary architecture that occurs upon liver injury and limit extracellular matrix deposition. Infliximab treatment, at least in these steatotic and cholestatic mouse models, is the safer approach since it does not increase liver injury, allows inflammation to take place but inhibits efficiently the ductular response and extracellular matrix deposition. Infliximab-based therapy could, thus, still be of importance in multiple chronic liver disorders that display a ductular response such as alcoholic liver disease or sclerosing cholangitis.
Collapse
|
112
|
Kitade M, Kaji K, Yoshiji H. Relationship between hepatic progenitor cell-mediated liver regeneration and non-parenchymal cells. Hepatol Res 2016; 46:1187-1193. [PMID: 26895456 DOI: 10.1111/hepr.12682] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2016] [Accepted: 02/12/2016] [Indexed: 12/21/2022]
Abstract
Hepatic progenitor cells (HPCs) are thought to reside in the canals of Hering and can be activated and contribute to liver regeneration in response to liver injury by proliferating and differentiating towards both hepatocytes and biliary epithelial cells. In this setting, several cytokines, chemokines, and growth factors related to liver inflammation and other liver cells comprising the HPC niche, namely hepatic stellate cells (HSCs), play crucial roles in HPC activation and differentiation. In response to several types of liver injury, tumor necrosis factor-like weak inducer of apoptosis (TWEAK) is secreted by several inflammatory cells, including monocytes, T lymphocytes, and macrophages, and acts as an initiator of the HPC niche and HSC activation. Following TWEAK-induced activation of the HPC niche, fibroblast growth factor 7 and hepatocyte growth factor released from activated HSC play central roles in maintaining HPC proliferation. In contrast, HGF-MET and Wnt3a-β-catenin signals are the predominant mediators of the hepatocyte differentiation of HPC, whereas epidermal growth factor receptor-NOTCH signaling controls HPC differentiation towards biliary epithelial cells. These signals are maintained exclusively by activated HSC and inflammatory cells surrounding HPC. Together, HSC and inflammatory cells surrounding HPC are responsible for the precise control of HPC proliferation and differentiation fate. In this review, we discuss recent progress in understanding of interactions between HPC and other liver cells in HPC-mediated liver regeneration in the setting of liver inflammation.
Collapse
|
113
|
LPS-TLR4 Pathway Mediates Ductular Cell Expansion in Alcoholic Hepatitis. Sci Rep 2016; 6:35610. [PMID: 27752144 PMCID: PMC5067590 DOI: 10.1038/srep35610] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Accepted: 09/29/2016] [Indexed: 02/06/2023] Open
Abstract
Alcoholic hepatitis (AH) is the most severe form of alcoholic liver disease for which there are no effective therapies. Patients with AH show impaired hepatocyte proliferation, expansion of inefficient ductular cells and high lipopolysaccharide (LPS) levels. It is unknown whether LPS mediates ductular cell expansion. We performed transcriptome studies and identified keratin 23 (KRT23) as a new ductular cell marker. KRT23 expression correlated with mortality and LPS serum levels. LPS-TLR4 pathway role in ductular cell expansion was assessed in human and mouse progenitor cells, liver slices and liver injured TLR4 KO mice. In AH patients, ductular cell expansion correlated with portal hypertension and collagen expression. Functional studies in ductular cells showed that KRT23 regulates collagen expression. These results support a role for LPS-TLR4 pathway in promoting ductular reaction in AH. Maneuvers aimed at decreasing LPS serum levels in AH patients could have beneficial effects by preventing ductular reaction development.
Collapse
|
114
|
Passman AM, Low J, London R, Tirnitz-Parker JEE, Miyajima A, Tanaka M, Strick-Marchand H, Darlington GJ, Finch-Edmondson M, Ochsner S, Zhu C, Whelan J, Callus BA, Yeoh GCT. A Transcriptomic Signature of Mouse Liver Progenitor Cells. Stem Cells Int 2016; 2016:5702873. [PMID: 27777588 PMCID: PMC5061959 DOI: 10.1155/2016/5702873] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2016] [Revised: 08/04/2016] [Accepted: 08/14/2016] [Indexed: 01/07/2023] Open
Abstract
Liver progenitor cells (LPCs) can proliferate extensively, are able to differentiate into hepatocytes and cholangiocytes, and contribute to liver regeneration. The presence of LPCs, however, often accompanies liver disease and hepatocellular carcinoma (HCC), indicating that they may be a cancer stem cell. Understanding LPC biology and establishing a sensitive, rapid, and reliable method to detect their presence in the liver will assist diagnosis and facilitate monitoring of treatment outcomes in patients with liver pathologies. A transcriptomic meta-analysis of over 400 microarrays was undertaken to compare LPC lines against datasets of muscle and embryonic stem cell lines, embryonic and developed liver (DL), and HCC. Three gene clusters distinguishing LPCs from other liver cell types were identified. Pathways overrepresented in these clusters denote the proliferative nature of LPCs and their association with HCC. Our analysis also revealed 26 novel markers, LPC markers, including Mcm2 and Ltbp3, and eight known LPC markers, including M2pk and Ncam. These markers specified the presence of LPCs in pathological liver tissue by qPCR and correlated with LPC abundance determined using immunohistochemistry. These results showcase the value of global transcript profiling to identify pathways and markers that may be used to detect LPCs in injured or diseased liver.
Collapse
Affiliation(s)
- Adam M. Passman
- School of Chemistry and Biochemistry, The University of Western Australia, Crawley, WA 6009, Australia
- The Centre for Medical Research, Harry Perkins Institute of Medical Research, Nedlands, WA 6009, Australia
| | - Jasmine Low
- School of Chemistry and Biochemistry, The University of Western Australia, Crawley, WA 6009, Australia
- ARC Centre of Excellence in Plant Energy Biology, The University of Western Australia, Crawley, WA 6009, Australia
| | - Roslyn London
- School of Chemistry and Biochemistry, The University of Western Australia, Crawley, WA 6009, Australia
| | - Janina E. E. Tirnitz-Parker
- School of Biomedical Sciences, Curtin Health Innovation Research Institute, Curtin University, Bentley, WA 6102, Australia
- School of Medicine and Pharmacology, The University of Western Australia, Fremantle, WA 6160, Australia
| | - Atsushi Miyajima
- Institute of Molecular and Cellular Biosciences, The University of Tokyo, Tokyo 113-8654, Japan
| | - Minoru Tanaka
- Institute of Molecular and Cellular Biosciences, The University of Tokyo, Tokyo 113-8654, Japan
| | | | | | - Megan Finch-Edmondson
- Department of Physiology, NUS Yong Loo Lin School of Medicine, Singapore 117411
- Mechanobiology Institute (MBI), National University of Singapore, Singapore 117411
| | - Scott Ochsner
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Cornelia Zhu
- School of Chemistry and Biochemistry, The University of Western Australia, Crawley, WA 6009, Australia
- The Centre for Medical Research, Harry Perkins Institute of Medical Research, Nedlands, WA 6009, Australia
| | - James Whelan
- ARC Centre of Excellence in Plant Energy Biology, The University of Western Australia, Crawley, WA 6009, Australia
- Department of Animal, Plant and Soil Sciences, La Trobe University, Melbourne, VIC 3086, Australia
| | - Bernard A. Callus
- School of Chemistry and Biochemistry, The University of Western Australia, Crawley, WA 6009, Australia
- The Centre for Medical Research, Harry Perkins Institute of Medical Research, Nedlands, WA 6009, Australia
- School of Health Sciences, The University of Notre Dame Australia, Fremantle, WA 6959, Australia
| | - George C. T. Yeoh
- School of Chemistry and Biochemistry, The University of Western Australia, Crawley, WA 6009, Australia
- The Centre for Medical Research, Harry Perkins Institute of Medical Research, Nedlands, WA 6009, Australia
| |
Collapse
|
115
|
Aravinthan AD, Alexander GJM. Senescence in chronic liver disease: Is the future in aging? J Hepatol 2016; 65:825-834. [PMID: 27245432 DOI: 10.1016/j.jhep.2016.05.030] [Citation(s) in RCA: 100] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Revised: 04/04/2016] [Accepted: 05/23/2016] [Indexed: 12/25/2022]
Abstract
Cellular senescence is a fundamental, complex mechanism with an important protective role present from embryogenesis to late life across all species. It limits the proliferative potential of damaged cells thus protecting against malignant change, but at the expense of substantial alterations to the microenvironment and tissue homeostasis, driving inflammation, fibrosis and paradoxically, malignant disease if the process is sustained. Cellular senescence has attracted considerable recent interest with recognition of pathways linking aging, malignancy and insulin resistance and the current focus on therapeutic interventions to extend health-span. There are major implications for hepatology in the field of fibrosis and cancer, where cellular senescence of hepatocytes, cholangiocytes, stellate cells and immune cells has been implicated in chronic liver disease progression. This review focuses on cellular senescence in chronic liver disease and explores therapeutic opportunities.
Collapse
Affiliation(s)
- Aloysious D Aravinthan
- Department of Medicine, University of Toronto, Toronto, Canada; National Institute for Health Research (NIHR) Nottingham Digestive Diseases Biomedical Research Unit, University of Nottingham and Nottingham University Hospitals NHS Trust, Nottingham, UK
| | - Graeme J M Alexander
- UCL Institute for Liver and Digestive Health, The Royal Free Trust, London, UK; Department of Medicine, University of Cambridge, Cambridge, UK.
| |
Collapse
|
116
|
Boens S, Verbinnen I, Verhulst S, Szekér K, Ferreira M, Gevaert T, Baes M, Roskams T, van Grunsven LA, Van Eynde A, Bollen M. Brief Report: The Deletion of the Phosphatase Regulator NIPP1 Causes Progenitor Cell Expansion in the Adult Liver. Stem Cells 2016; 34:2256-62. [PMID: 27068806 DOI: 10.1002/stem.2375] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2015] [Revised: 02/17/2016] [Accepted: 03/14/2016] [Indexed: 01/10/2023]
Abstract
The Ppp1r8 gene encodes NIPP1, a nuclear interactor of protein phosphatase PP1. The deletion of NIPP1 is embryonic lethal at the gastrulation stage, which has hampered its functional characterization in adult tissues. Here, we describe the effects of a conditional deletion of NIPP1 in mouse liver epithelial cells. Ppp1r8(-/-) livers developed a ductular reaction, that is, bile-duct hyperplasia with associated fibrosis. The increased proliferation of biliary epithelial cells was at least partially due to an expansion of the progenitor cell compartment that was independent of liver injury. Gene-expression analysis confirmed an upregulation of progenitor cell markers in the liver knockout livers but showed no effect on the expression of liver-injury associated regulators of cholangiocyte differentiation markers. Consistent with an inhibitory effect of NIPP1 on progenitor cell proliferation, Ppp1r8(-/-) livers displayed an increased sensitivity to diet-supplemented 3,5-diethoxycarbonyl-1,4-dihydrocollidine, which also causes bile-duct hyperplasia through progenitor cell expansion. In contrast, the liver knockouts responded normally to injuries (partial hepatectomy, single CCl4 administration) that are restored through proliferation of differentiated parenchymal cells. Our data indicate that NIPP1 does not regulate the proliferation of hepatocytes but is a suppressor of biliary epithelial cell proliferation, including progenitor cells, in the adult liver. Stem Cells 2016;34:2256-2262.
Collapse
Affiliation(s)
- Shannah Boens
- Department of Cellular and Molecular Medicine, Laboratory of Biosignaling & Therapeutics, KU Leuven, Belgium
| | - Iris Verbinnen
- Department of Cellular and Molecular Medicine, Laboratory of Biosignaling & Therapeutics, KU Leuven, Belgium
| | - Stefaan Verhulst
- Liver Cell Biology Lab, Vrije Universiteit Brussel, Brussel, Belgium
| | - Kathelijne Szekér
- Department of Cellular and Molecular Medicine, Laboratory of Biosignaling & Therapeutics, KU Leuven, Belgium
| | - Monica Ferreira
- Department of Cellular and Molecular Medicine, Laboratory of Biosignaling & Therapeutics, KU Leuven, Belgium
| | - Thomas Gevaert
- Department of Development and Regeneration, Organ Systems, KU Leuven, Belgium
| | - Myriam Baes
- Department of Pharmaceutical & Pharmacological Sciences, Laboratory for Cell Metabolism, KU Leuven, Belgium
| | - Tania Roskams
- Department of Imaging & Pathology, Laboratory of Translational Cell & Tissue Research, KU Leuven, Belgium
| | | | - Aleyde Van Eynde
- Department of Cellular and Molecular Medicine, Laboratory of Biosignaling & Therapeutics, KU Leuven, Belgium
| | - Mathieu Bollen
- Department of Cellular and Molecular Medicine, Laboratory of Biosignaling & Therapeutics, KU Leuven, Belgium
| |
Collapse
|
117
|
Multi-OMICs and Genome Editing Perspectives on Liver Cancer Signaling Networks. BIOMED RESEARCH INTERNATIONAL 2016; 2016:6186281. [PMID: 27403431 PMCID: PMC4923561 DOI: 10.1155/2016/6186281] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/01/2016] [Revised: 04/23/2016] [Accepted: 05/08/2016] [Indexed: 12/26/2022]
Abstract
The advent of the human genome sequence and the resulting ~20,000 genes provide a crucial framework for a transition from traditional biology to an integrative “OMICs” arena (Lander et al., 2001; Venter et al., 2001; Kitano, 2002). This brings in a revolution for cancer research, which now enters a big data era. In the past decade, with the facilitation by next-generation sequencing, there have been a huge number of large-scale sequencing efforts, such as The Cancer Genome Atlas (TCGA), the HapMap, and the 1000 genomes project. As a result, a deluge of genomic information becomes available from patients stricken by a variety of cancer types. The list of cancer-associated genes is ever expanding. New discoveries are made on how frequent and highly penetrant mutations, such as those in the telomerase reverse transcriptase (TERT) and TP53, function in cancer initiation, progression, and metastasis. Most genes with relatively frequent but weakly penetrant cancer mutations still remain to be characterized. In addition, genes that harbor rare but highly penetrant cancer-associated mutations continue to emerge. Here, we review recent advances related to cancer genomics, proteomics, and systems biology and suggest new perspectives in targeted therapy and precision medicine.
Collapse
|
118
|
Environmental Ligands of the Aryl Hydrocarbon Receptor and Their Effects in Models of Adult Liver Progenitor Cells. Stem Cells Int 2016; 2016:4326194. [PMID: 27274734 PMCID: PMC4870370 DOI: 10.1155/2016/4326194] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Accepted: 04/07/2016] [Indexed: 12/20/2022] Open
Abstract
The toxicity of environmental and dietary ligands of the aryl hydrocarbon receptor (AhR) in mature liver parenchymal cells is well appreciated, while considerably less attention has been paid to their impact on cell populations exhibiting phenotypic features of liver progenitor cells. Here, we discuss the results suggesting that the consequences of the AhR activation in the cellular models derived from bipotent liver progenitors could markedly differ from those in hepatocytes. In contact-inhibited liver progenitor cells, the AhR agonists induce a range of effects potentially linked with tumor promotion. They can stimulate cell cycle progression/proliferation and deregulate cell-to-cell communication, which is associated with downregulation of proteins forming gap junctions, adherens junctions, and desmosomes (such as connexin 43, E-cadherin, β-catenin, and plakoglobin), as well as with reduced cell adhesion and inhibition of intercellular communication. At the same time, toxic AhR ligands may affect the activity of the signaling pathways contributing to regulation of liver progenitor cell activation and/or differentiation, such as downregulation of Wnt/β-catenin and TGF-β signaling, or upregulation of transcriptional targets of YAP/TAZ, the effectors of Hippo signaling pathway. These data illustrate the need to better understand the potential role of liver progenitors in the AhR-mediated liver carcinogenesis and tumor promotion.
Collapse
|
119
|
The potential role of liver stem cells in initiation of primary liver cancer. Hepatol Int 2016; 10:893-901. [PMID: 27139191 DOI: 10.1007/s12072-016-9730-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2015] [Accepted: 04/01/2016] [Indexed: 02/06/2023]
Abstract
Identification of the cellular origin of primary liver cancer remains challenging. Some data point toward liver stem cells (LSCs) or liver progenitor cells (LPCs) not only as propagators of liver regeneration, but also as initiators of liver cancer. LSCs exhibit a long lifespan and strong duplicative potential upon activation and are inclined to accumulate more mutations that can be passed down to the next generations. Recent evidence shows that dysregulation of signaling pathways associated with self-renewal of LSCs can drive their aberrant proliferation and even malignant transformation. If LSCs could be proved to be an initiator of liver carcinogenesis, they would be promising for ultra-early diagnosis and targeting therapy of liver cancer. This review mainly summarizes the potential role of LSCs in the carcinogenesis of primary liver cancer.
Collapse
|
120
|
El'chaninov AV, Fatkhudinov TK, Kananykhina EY, Usman NY, Arutyunyan IV, Makarov AV, Bykov AV, Bolshakova GV, Sukhikh GT. Role of Progenitor Cells in Liver Regeneration after Subtotal Resection. Bull Exp Biol Med 2016; 161:155-61. [PMID: 27265142 DOI: 10.1007/s10517-016-3367-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2015] [Indexed: 02/07/2023]
Abstract
In the liver of rats subjected to subtotal liver resection (80% organ weight), the expression of sox9 gene and SOX9 protein content increased and cells with hepatocyte morphology expressing SOX9 appeared; the proportion of cells expressing cytokeratin-19 also increased. Based on these data, we cannot completely exclude the involvement of resident progenitor cells and hepatocyte reprogramming in liver regeneration after subtotal resection, however, the contribution of these processes seems to be insignificant. The leading mechanism of liver mass recovery after subtotal resection is proliferation of hepatocytes.
Collapse
Affiliation(s)
- A V El'chaninov
- V. I. Kulakov Research Center of Obstetrics, Gynecology, and Perinatology, Ministry of Health of the Russian Federation, Moscow, Russia
- Institute of Human Morphology, Moscow, Russia
- N. I. Pirogov Russian State Medical University, Ministry of Health of the Russian Federation, Moscow, Russia
| | - T Kh Fatkhudinov
- V. I. Kulakov Research Center of Obstetrics, Gynecology, and Perinatology, Ministry of Health of the Russian Federation, Moscow, Russia.
- Institute of Human Morphology, Moscow, Russia.
- N. I. Pirogov Russian State Medical University, Ministry of Health of the Russian Federation, Moscow, Russia.
| | - E Yu Kananykhina
- V. I. Kulakov Research Center of Obstetrics, Gynecology, and Perinatology, Ministry of Health of the Russian Federation, Moscow, Russia
- Institute of Human Morphology, Moscow, Russia
| | - N Yu Usman
- V. I. Kulakov Research Center of Obstetrics, Gynecology, and Perinatology, Ministry of Health of the Russian Federation, Moscow, Russia
- Institute of Human Morphology, Moscow, Russia
| | - I V Arutyunyan
- V. I. Kulakov Research Center of Obstetrics, Gynecology, and Perinatology, Ministry of Health of the Russian Federation, Moscow, Russia
- Institute of Human Morphology, Moscow, Russia
| | - A V Makarov
- V. I. Kulakov Research Center of Obstetrics, Gynecology, and Perinatology, Ministry of Health of the Russian Federation, Moscow, Russia
- Institute of Human Morphology, Moscow, Russia
- N. I. Pirogov Russian State Medical University, Ministry of Health of the Russian Federation, Moscow, Russia
| | - A V Bykov
- N. I. Pirogov Russian State Medical University, Ministry of Health of the Russian Federation, Moscow, Russia
| | | | - G T Sukhikh
- V. I. Kulakov Research Center of Obstetrics, Gynecology, and Perinatology, Ministry of Health of the Russian Federation, Moscow, Russia
| |
Collapse
|
121
|
Font-Burgada J, Shalapour S, Ramaswamy S, Hsueh B, Rossell D, Umemura A, Taniguchi K, Nakagawa H, Valasek MA, Ye L, Kopp JL, Sander M, Carter H, Deisseroth K, Verma IM, Karin M. Hybrid Periportal Hepatocytes Regenerate the Injured Liver without Giving Rise to Cancer. Cell 2016; 162:766-79. [PMID: 26276631 DOI: 10.1016/j.cell.2015.07.026] [Citation(s) in RCA: 368] [Impact Index Per Article: 40.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2014] [Revised: 03/25/2015] [Accepted: 06/26/2015] [Indexed: 12/24/2022]
Abstract
Compensatory proliferation triggered by hepatocyte loss is required for liver regeneration and maintenance but also promotes development of hepatocellular carcinoma (HCC). Despite extensive investigation, the cells responsible for hepatocyte restoration or HCC development remain poorly characterized. We used genetic lineage tracing to identify cells responsible for hepatocyte replenishment following chronic liver injury and queried their roles in three distinct HCC models. We found that a pre-existing population of periportal hepatocytes, located in the portal triads of healthy livers and expressing low amounts of Sox9 and other bile-duct-enriched genes, undergo extensive proliferation and replenish liver mass after chronic hepatocyte-depleting injuries. Despite their high regenerative potential, these so-called hybrid hepatocytes do not give rise to HCC in chronically injured livers and thus represent a unique way to restore tissue function and avoid tumorigenesis. This specialized set of pre-existing differentiated cells may be highly suitable for cell-based therapy of chronic hepatocyte-depleting disorders.
Collapse
Affiliation(s)
- Joan Font-Burgada
- Laboratory of Gene Regulation and Signal Transduction, Department of Pharmacology, School of Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA.
| | - Shabnam Shalapour
- Laboratory of Gene Regulation and Signal Transduction, Department of Pharmacology, School of Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Suvasini Ramaswamy
- Laboratory of Genetics, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Brian Hsueh
- Departments of Bioengineering, Psychiatry, and Behavioral Sciences, Neurosciences Program, Howard Hughes Medical Institute, Stanford University, 318 Campus Drive West, Clark Center W080, Stanford, CA 94305, USA
| | - David Rossell
- Department of Statistics, University of Warwick, Gibbet Hill Road, Coventry CV4 7AL, UK
| | - Atsushi Umemura
- Laboratory of Gene Regulation and Signal Transduction, Department of Pharmacology, School of Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Koji Taniguchi
- Laboratory of Gene Regulation and Signal Transduction, Department of Pharmacology, School of Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Hayato Nakagawa
- Laboratory of Gene Regulation and Signal Transduction, Department of Pharmacology, School of Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA; Department of Gastroenterology, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655
| | - Mark A Valasek
- Department of Pathology, School of Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Li Ye
- Departments of Bioengineering, Psychiatry, and Behavioral Sciences, Neurosciences Program, Howard Hughes Medical Institute, Stanford University, 318 Campus Drive West, Clark Center W080, Stanford, CA 94305, USA
| | - Janel L Kopp
- Department of Pediatrics and Department of Cellular & Molecular Medicine, School of Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA; Department of Cellular & Physiological Sciences, University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC V6T 1Z3, Canada
| | - Maike Sander
- Department of Pediatrics and Department of Cellular & Molecular Medicine, School of Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Hannah Carter
- Department of Medicine, School of Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Karl Deisseroth
- Departments of Bioengineering, Psychiatry, and Behavioral Sciences, Neurosciences Program, Howard Hughes Medical Institute, Stanford University, 318 Campus Drive West, Clark Center W080, Stanford, CA 94305, USA
| | - Inder M Verma
- Laboratory of Genetics, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Michael Karin
- Laboratory of Gene Regulation and Signal Transduction, Department of Pharmacology, School of Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA; Department of Pathology, School of Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA.
| |
Collapse
|
122
|
Michelotti GA, Tucker A, Swiderska-Syn M, Machado MV, Choi SS, Kruger L, Soderblom E, Thompson JW, Mayer-Salman M, Himburg HA, Moylan CA, Guy CD, Garman KS, Premont RT, Chute JP, Diehl AM. Pleiotrophin regulates the ductular reaction by controlling the migration of cells in liver progenitor niches. Gut 2016; 65:683-92. [PMID: 25596181 PMCID: PMC4504836 DOI: 10.1136/gutjnl-2014-308176] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2014] [Accepted: 12/22/2014] [Indexed: 12/12/2022]
Abstract
OBJECTIVE The ductular reaction (DR) involves mobilisation of reactive-appearing duct-like cells (RDC) along canals of Hering, and myofibroblastic (MF) differentiation of hepatic stellate cells (HSC) in the space of Disse. Perivascular cells in stem cell niches produce pleiotrophin (PTN) to inactivate the PTN receptor, protein tyrosine phosphatase receptor zeta-1 (PTPRZ1), thereby augmenting phosphoprotein-dependent signalling. We hypothesised that the DR is regulated by PTN/PTPRZ1 signalling. DESIGN PTN-GFP, PTN-knockout (KO), PTPRZ1-KO, and wild type (WT) mice were examined before and after bile duct ligation (BDL) for PTN, PTPRZ1 and the DR. RDC and HSC from WT, PTN-KO, and PTPRZ1-KO mice were also treated with PTN to determine effects on downstream signaling phosphoproteins, gene expression, growth, and migration. Liver biopsies from patients with DRs were also interrogated. RESULTS Although quiescent HSC and RDC lines expressed PTN and PTPRZ1 mRNAs, neither PTN nor PTPRZ1 protein was demonstrated in healthy liver. BDL induced PTN in MF-HSC and increased PTPRZ1 in MF-HSC and RDC. In WT mice, BDL triggered a DR characterised by periportal accumulation of collagen, RDC and MF-HSC. All aspects of this DR were increased in PTN-KO mice and suppressed in PTPRZ1-KO mice. In vitro studies revealed PTN-dependent accumulation of phosphoproteins that control cell-cell adhesion and migration, with resultant inhibition of cell migration. PTPRZ1-positive cells were prominent in the DRs of patients with ductal plate defects and adult cholestatic diseases. CONCLUSIONS PTN, and its receptor, PTPRZ1, regulate the DR to liver injury by controlling the migration of resident cells in adult liver progenitor niches.
Collapse
Affiliation(s)
| | - Anikia Tucker
- Division of Gastroenterology, Duke University, Durham, North Carolina, USA
| | | | | | - Steve S Choi
- Division of Gastroenterology, Duke University, Durham, North Carolina, USA Section of Gastroenterology, Durham Veterans Affairs Medical Center, Durham, North Carolina, USA
| | - Leandi Kruger
- Division of Gastroenterology, Duke University, Durham, North Carolina, USA
| | - Erik Soderblom
- Proteomics Center, Duke University, Durham, North Carolina, USA
| | - J Will Thompson
- Proteomics Center, Duke University, Durham, North Carolina, USA
| | | | - Heather A Himburg
- Division of Hematology and Oncology, UCLA, Los Angeles, California, USA
| | - Cynthia A Moylan
- Division of Gastroenterology, Duke University, Durham, North Carolina, USA Section of Gastroenterology, Durham Veterans Affairs Medical Center, Durham, North Carolina, USA
| | - Cynthia D Guy
- Department of Pathology, Duke University, Durham, North Carolina, USA
| | - Katherine S Garman
- Division of Gastroenterology, Duke University, Durham, North Carolina, USA Section of Gastroenterology, Durham Veterans Affairs Medical Center, Durham, North Carolina, USA
| | - Richard T Premont
- Division of Gastroenterology, Duke University, Durham, North Carolina, USA
| | - John P Chute
- Division of Hematology and Oncology, UCLA, Los Angeles, California, USA
| | - Anna Mae Diehl
- Division of Gastroenterology, Duke University, Durham, North Carolina, USA
| |
Collapse
|
123
|
Ding ZY, Jin GN, Wang W, Sun YM, Chen WX, Chen L, Liang HF, Datta PK, Zhang MZ, Zhang B, Chen XP. Activin A-Smad Signaling Mediates Connective Tissue Growth Factor Synthesis in Liver Progenitor Cells. Int J Mol Sci 2016; 17:408. [PMID: 27011166 PMCID: PMC4813263 DOI: 10.3390/ijms17030408] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2015] [Revised: 03/02/2016] [Accepted: 03/02/2016] [Indexed: 01/19/2023] Open
Abstract
Liver progenitor cells (LPCs) are activated in chronic liver damage and may contribute to liver fibrosis. Our previous investigation reported that LPCs produced connective tissue growth factor (CTGF/CCN2), an inducer of liver fibrosis, yet the regulatory mechanism of the production of CTGF/CCN2 in LPCs remains elusive. In this study, we report that Activin A is an inducer of CTGF/CCN2 in LPCs. Here we show that expression of both Activin A and CTGF/CCN2 were upregulated in the cirrhotic liver, and the expression of Activin A positively correlates with that of CTGF/CCN2 in liver tissues. We go on to show that Activin A induced de novo synthesis of CTGF/CCN2 in LPC cell lines LE/6 and WB-F344. Furthermore, Activin A contributed to autonomous production of CTGF/CCN2 in liver progenitor cells (LPCs) via activation of the Smad signaling pathway. Smad2, 3 and 4 were all required for this induction. Collectively, these results provide evidence for the fibrotic role of LPCs in the liver and suggest that the Activin A-Smad-CTGF/CCN2 signaling in LPCs may be a therapeutic target of liver fibrosis.
Collapse
Affiliation(s)
- Ze-Yang Ding
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| | - Guan-Nan Jin
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| | - Wei Wang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| | - Yi-Min Sun
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| | - Wei-Xun Chen
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| | - Lin Chen
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| | - Hui-Fang Liang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| | - Pran K Datta
- Division of Hematology and Oncology, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA.
| | - Ming-Zhi Zhang
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University, Nashville, TN 37235, USA.
| | - Bixiang Zhang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| | - Xiao-Ping Chen
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| |
Collapse
|
124
|
Lu J, Zhou Y, Hu T, Zhang H, Shen M, Cheng P, Dai W, Wang F, Chen K, Zhang Y, Wang C, Li J, Zheng Y, Yang J, Zhu R, Wang J, Lu W, Zhang H, Wang J, Xia Y, De Assuncao TM, Jalan-Sakrikar N, Huebert RC, Bin Zhou, Guo C. Notch Signaling Coordinates Progenitor Cell-Mediated Biliary Regeneration Following Partial Hepatectomy. Sci Rep 2016; 6:22754. [PMID: 26951801 PMCID: PMC4782135 DOI: 10.1038/srep22754] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2015] [Accepted: 02/19/2016] [Indexed: 02/08/2023] Open
Abstract
Aberrant transcriptional regulation contributes to the pathogenesis of both congenital and adult forms of liver disease. Although the transcription factor RBPJ is essential for liver morphogenesis and biliary development, its specific function in the differentiation of hepatic progenitor cells (HPC) has not been investigated, and little is known about its role in adult liver regeneration. HPCs are bipotent liver stem cells that can self-replicate and differentiate into hepatocytes or cholangiocytes in vitro. HPCs are thought to play an important role in liver regeneration and repair responses. While the coordinated repopulation of both hepatocyte and cholangiocyte compartment is pivotal to the structure and function of the liver after regeneration, the mechanisms coordinating biliary regeneration remain vastly understudied. Here, we utilized complex genetic manipulations to drive liver-specific deletion of the Rbpj gene in conjunction with lineage tracing techniques to delineate the precise functions of RBPJ during biliary development and HPC-associated biliary regeneration after hepatectomy. Furthermore, we demonstrate that RBPJ promotes HPC differentiation toward cholangiocytes in vitro and blocks hepatocyte differentiation through mechanisms involving Hippo-Notch crosstalk. Overall, this study demonstrates that the Notch-RBPJ signaling axis critically regulates biliary regeneration by coordinating the fate decision of HPC and clarifies the molecular mechanisms involved.
Collapse
Affiliation(s)
- Jie Lu
- Department of Gastroenterology, Shanghai 10th People's Hospital, Tongji University School of Medicine, Shanghai, People's Republic of China
| | - Yingqun Zhou
- Department of Gastroenterology, Shanghai 10th People's Hospital, Tongji University School of Medicine, Shanghai, People's Republic of China
| | - Tianyuan Hu
- Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, People's Republic of China
| | - Hui Zhang
- Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, People's Republic of China
| | - Miao Shen
- Department of Gastroenterology, Shanghai 10th People's Hospital, Tongji University School of Medicine, Shanghai, People's Republic of China
| | - Ping Cheng
- Department of Gastroenterology, Shanghai 10th People's Hospital, Tongji University School of Medicine, Shanghai, People's Republic of China
| | - Weiqi Dai
- Department of Gastroenterology, Shanghai 10th People's Hospital, Tongji University School of Medicine, Shanghai, People's Republic of China
| | - Fan Wang
- Department of Gastroenterology, Shanghai 10th People's Hospital, Tongji University School of Medicine, Shanghai, People's Republic of China
| | - Kan Chen
- Department of Gastroenterology, Shanghai 10th People's Hospital, Tongji University School of Medicine, Shanghai, People's Republic of China
| | - Yan Zhang
- Department of Gastroenterology, Shanghai 10th People's Hospital, Tongji University School of Medicine, Shanghai, People's Republic of China
| | - Chengfeng Wang
- Department of Gastroenterology, Shanghai 10th People's Hospital, Tongji University School of Medicine, Shanghai, People's Republic of China
| | - Jingjing Li
- Department of Gastroenterology, Shanghai 10th People's Hospital, Tongji University School of Medicine, Shanghai, People's Republic of China
| | - Yuanyuan Zheng
- Department of Gastroenterology, Shanghai 10th People's Hospital, Tongji University School of Medicine, Shanghai, People's Republic of China
| | - Jing Yang
- Department of Gastroenterology, Shanghai 10th People's Hospital, Tongji University School of Medicine, Shanghai, People's Republic of China
| | - Rong Zhu
- Department of Gastroenterology, Shanghai 10th People's Hospital, Tongji University School of Medicine, Shanghai, People's Republic of China
| | - Jianrong Wang
- Department of Gastroenterology, Shanghai 10th People's Hospital, Tongji University School of Medicine, Shanghai, People's Republic of China
| | - Wenxia Lu
- Department of Gastroenterology, Shanghai 10th People's Hospital, Tongji University School of Medicine, Shanghai, People's Republic of China
| | - Huawei Zhang
- Department of Gastroenterology, Shanghai 10th People's Hospital, Tongji University School of Medicine, Shanghai, People's Republic of China
| | - Junshan Wang
- Department of Gastroenterology, Shanghai 10th People's Hospital, Tongji University School of Medicine, Shanghai, People's Republic of China
| | - Yujing Xia
- Department of Gastroenterology, Shanghai 10th People's Hospital, Tongji University School of Medicine, Shanghai, People's Republic of China
| | - Thiago M De Assuncao
- Division of Gastroenterology and Hepatology; Mayo Clinic and Foundation, Rochester, MN, USA
| | - Nidhi Jalan-Sakrikar
- Division of Gastroenterology and Hepatology; Mayo Clinic and Foundation, Rochester, MN, USA
| | - Robert C Huebert
- Division of Gastroenterology and Hepatology; Mayo Clinic and Foundation, Rochester, MN, USA
| | - Bin Zhou
- Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, People's Republic of China
| | - Chuanyong Guo
- Department of Gastroenterology, Shanghai 10th People's Hospital, Tongji University School of Medicine, Shanghai, People's Republic of China
| |
Collapse
|
125
|
Zhang H, Siegel CT, Shuai L, Lai J, Zeng L, Zhang Y, Lai X, Bie P, Bai L. Repair of liver mediated by adult mouse liver neuro-glia antigen 2-positive progenitor cell transplantation in a mouse model of cirrhosis. Sci Rep 2016; 6:21783. [PMID: 26905303 PMCID: PMC4764864 DOI: 10.1038/srep21783] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2015] [Accepted: 02/01/2016] [Indexed: 02/07/2023] Open
Abstract
NG2-expressing cells are a population of periportal vascular stem/progenitors (MLpvNG2(+) cells) that were isolated from healthy adult mouse liver by using a "Percoll-Plate-Wait" procedure. We demonstrated that isolated cells are able to restore liver function after transplantation into a cirrhotic liver, and co-localized with the pericyte marker (immunohistochemistry: PDGFR-β) and CK19. Cells were positive for: stem cell (Sca-1, CD133, Dlk) and liver stem cell markers (EpCAM, CD14, CD24, CD49f); and negative for: hematopoietic (CD34, CD45) and endothelial markers (CD31, vWf, von Willebrand factor). Cells were transplanted (1 × 10(6) cells) in mice with diethylnitrosamine-induced cirrhosis at week 6. Cells showed increased hepatic associated gene expression of alpha-fetoprotein (AFP), Albumin (Alb), Glucose-6-phosphatase (G6Pc), SRY (sex determining region Y)-box 9 (Sox9), hepatic nuclear factors (HNF1a, HNF1β, HNF3β, HNF4α, HNF6, Epithelial cell adhesion molecule (EpCAM), Leucine-rich repeated-containing G-protein coupled receptor 5-positive (Lgr5) and Tyrosine aminotransferase (TAT). Cells showed decreased fibrogenesis, hepatic stellate cell infiltration, Kupffer cells and inflammatory cytokines. Liver function markers improved. In a cirrhotic liver environment, cells could differentiate into hepatic lineages. In addition, grafted MLpvNG2(+) cells could mobilize endogenous stem/progenitors to participate in liver repair. These results suggest that MLpvNG2(+) cells may be novel adult liver progenitors that participate in liver regeneration.
Collapse
Affiliation(s)
- Hongyu Zhang
- Hepatobiliary Institute, Southwestern Hospital, No. 30 Gaotanyan, ShapingBa Distract, Chongqing 400038, China
| | - Christopher T. Siegel
- Department of Surgery, Division of Hepatobiliary and Abdominal Organ Transplantation, Case Western Reserve University Hospital, Cleveland OH 44106, USA
| | - Ling Shuai
- Hepatobiliary Institute, Southwestern Hospital, No. 30 Gaotanyan, ShapingBa Distract, Chongqing 400038, China
| | - Jiejuan Lai
- Hepatobiliary Institute, Southwestern Hospital, No. 30 Gaotanyan, ShapingBa Distract, Chongqing 400038, China
| | - Linli Zeng
- Hepatobiliary Institute, Southwestern Hospital, No. 30 Gaotanyan, ShapingBa Distract, Chongqing 400038, China
| | - Yujun Zhang
- Hepatobiliary Institute, Southwestern Hospital, No. 30 Gaotanyan, ShapingBa Distract, Chongqing 400038, China
| | - Xiangdong Lai
- Hepatobiliary Institute, Southwestern Hospital, No. 30 Gaotanyan, ShapingBa Distract, Chongqing 400038, China
| | - Ping Bie
- Hepatobiliary Institute, Southwestern Hospital, No. 30 Gaotanyan, ShapingBa Distract, Chongqing 400038, China
| | - Lianhua Bai
- Hepatobiliary Institute, Southwestern Hospital, No. 30 Gaotanyan, ShapingBa Distract, Chongqing 400038, China
| |
Collapse
|
126
|
Köhn-Gaone J, Gogoi-Tiwari J, Ramm GA, Olynyk JK, Tirnitz-Parker JEE. The role of liver progenitor cells during liver regeneration, fibrogenesis, and carcinogenesis. Am J Physiol Gastrointest Liver Physiol 2016; 310:G143-54. [PMID: 26608186 DOI: 10.1152/ajpgi.00215.2015] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Accepted: 11/19/2015] [Indexed: 01/31/2023]
Abstract
The growing worldwide challenge of cirrhosis and hepatocellular carcinoma due to increasing prevalence of excessive alcohol consumption, viral hepatitis, obesity, and the metabolic syndrome has sparked interest in stem cell-like liver progenitor cells (LPCs) as potential candidates for cell therapy and tissue engineering, as an alternative approach to whole organ transplantation. However, LPCs always proliferate in chronic liver diseases with a predisposition to cancer; they have been suggested to play major roles in driving fibrosis, disease progression, and may even represent tumor-initiating cells. Hence, a greater understanding of the factors that govern their activation, communication with other hepatic cell types, and bipotential differentiation as opposed to their potential transformation is needed before their therapeutic potential can be harnessed.
Collapse
Affiliation(s)
- Julia Köhn-Gaone
- Curtin Health Innovation Research Institute, Curtin University, Perth Western Australia, Australia
| | - Jully Gogoi-Tiwari
- Curtin Health Innovation Research Institute, Curtin University, Perth Western Australia, Australia
| | - Grant A Ramm
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia; Faculty of Medicine and Biomedical Sciences, The University of Queensland, Brisbane, Queensland, Australia
| | - John K Olynyk
- Curtin Health Innovation Research Institute, Curtin University, Perth Western Australia, Australia; Fiona Stanley and Fremantle Hospitals, Western Australia, Australia; School of Veterinary and Life Sciences, Murdoch University, Murdoch, Western Australia, Australia; and
| | - Janina E E Tirnitz-Parker
- Curtin Health Innovation Research Institute, Curtin University, Perth Western Australia, Australia; School of Medicine and Pharmacology, University of Western Australia, Fremantle Western Australia, Australia
| |
Collapse
|
127
|
Stem/Progenitor Cell Niches Involved in Hepatic and Biliary Regeneration. Stem Cells Int 2016; 2016:3658013. [PMID: 26880956 PMCID: PMC4737003 DOI: 10.1155/2016/3658013] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2015] [Revised: 11/16/2015] [Accepted: 11/23/2015] [Indexed: 12/28/2022] Open
Abstract
Niches containing stem/progenitor cells are present in different anatomical locations along the human biliary tree and within liver acini. The most primitive stem/progenitors, biliary tree stem/progenitor cells (BTSCs), reside within peribiliary glands located throughout large extrahepatic and intrahepatic bile ducts. BTSCs are multipotent and can differentiate towards hepatic and pancreatic cell fates. These niches' matrix chemistry and other characteristics are undefined. Canals of Hering (bile ductules) are found periportally and contain hepatic stem/progenitor cells (HpSCs), participating in the renewal of small intrahepatic bile ducts and being precursors to hepatocytes and cholangiocytes. The niches also contain precursors to hepatic stellate cells and endothelia, macrophages, and have a matrix chemistry rich in hyaluronans, minimally sulfated proteoglycans, fetal collagens, and laminin. The microenvironment furnishes key signals driving HpSC activation and differentiation. Newly discovered third niches are pericentral within hepatic acini, contain Axin2+ unipotent hepatocytic progenitors linked on their lateral borders to endothelia forming the central vein, and contribute to normal turnover of mature hepatocytes. Their relationship to the other stem/progenitors is undefined. Stem/progenitor niches have important implications in regenerative medicine for the liver and biliary tree and in pathogenic processes leading to diseases of these tissues.
Collapse
|
128
|
Liver progenitor cells-mediated liver regeneration in liver cirrhosis. Hepatol Int 2016; 10:440-7. [PMID: 26742763 DOI: 10.1007/s12072-015-9693-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2015] [Accepted: 12/07/2015] [Indexed: 02/08/2023]
Abstract
Cirrhosis is defined as the histological development of regenerative nodules surrounded by fibrous bands in response to chronic liver injury. In cirrhotic liver where hepatocytes proliferation is compromised, liver progenitor cells (LPCs) are activated and then differentiated into hepatocytes and cholangiocytes, leading to the generation of regenerative nodules and functional restoration. Here, we summarize and discuss recent findings on the mechanisms underlying LPCs-mediated regeneration in liver cirrhosis. Firstly, we provide recent research on the mechanism underlying LPCs activation in severe or chronic liver injury. Secondly, we present new and exciting data on exploring the origin of LPCs, which reveal that the hepatocytes give rise to duct-like progenitors that then differentiate back into hepatocytes in chronic liver injury or liver cirrhosis. Finally, we highlight recent findings from the literature exploring the role of LPCs niche in directing the behavior and fate of LPCs. This remarkable insight into the cellular and molecular mechanisms of LPCs-mediated regeneration in liver cirrhosis will provide a basis for translating this knowledge into clinical application.
Collapse
|
129
|
Melino M, Gadd VL, Alexander KA, Beattie L, Lineburg KE, Martinez M, Teal B, Le Texier L, Irvine KM, Miller GC, Boyle GM, Hill GR, Clouston AD, Powell EE, MacDonald KPA. Spatiotemporal Characterization of the Cellular and Molecular Contributors to Liver Fibrosis in a Murine Hepatotoxic-Injury Model. THE AMERICAN JOURNAL OF PATHOLOGY 2016; 186:524-38. [PMID: 26762581 DOI: 10.1016/j.ajpath.2015.10.029] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/10/2015] [Revised: 09/17/2015] [Accepted: 10/27/2015] [Indexed: 12/15/2022]
Abstract
The interplay between the inflammatory infiltrate and tissue resident cell populations invokes fibrogenesis. However, the temporal and mechanistic contributions of these cells to fibrosis are obscure. To address this issue, liver inflammation, ductular reaction (DR), and fibrosis were induced in C57BL/6 mice by thioacetamide administration for up to 12 weeks. Thioacetamide treatment induced two phases of liver fibrosis. A rapid pericentral inflammatory infiltrate enriched in F4/80(+) monocytes co-localized with SMA(+) myofibroblasts resulted in early collagen deposition, marking the start of an initial fibrotic phase (1 to 6 weeks). An expansion of bone marrow-derived macrophages preceded a second phase, characterized by accelerated progression of fibrosis (>6 weeks) after DR migration from the portal tracts to the centrilobular site of injury, in association with an increase in DR/macrophage interactions. Although chemokine (C-C motif) ligand 2 (CCL2) mRNA was induced rapidly in response to thioacetamide, CCL2 deficiency only partially abrogated fibrosis. In contrast, colony-stimulating factor 1 receptor blockade diminished C-C chemokine receptor type 2 [CCR2(neg) (Ly6C(lo))] monocytes, attenuated the DR, and significantly reduced fibrosis, illustrating the critical role of colony-stimulating factor 1-dependent monocyte/macrophage differentiation and linking the two phases of injury. In response to liver injury, colony-stimulating factor 1 drives early monocyte-mediated myofibroblast activation and collagen deposition, subsequent macrophage differentiation, and their association with the advancing DR, the formation of fibrotic septa, and the progression of liver fibrosis to cirrhosis.
Collapse
Affiliation(s)
- Michelle Melino
- Department of Immunology, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Victoria L Gadd
- Centre for Liver Disease Research, The University of Queensland, Brisbane, Queensland, Australia
| | - Kylie A Alexander
- Department of Immunology, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Lynette Beattie
- Department of Immunology, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Katie E Lineburg
- Department of Immunology, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Michelle Martinez
- Department of Immunology, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Bianca Teal
- Department of Immunology, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Laetitia Le Texier
- Department of Immunology, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Katharine M Irvine
- Centre for Liver Disease Research, The University of Queensland, Brisbane, Queensland, Australia
| | | | - Glen M Boyle
- Department of Cell and Molecular Biology, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Geoffrey R Hill
- Department of Immunology, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Andrew D Clouston
- Centre for Liver Disease Research, The University of Queensland, Brisbane, Queensland, Australia; Department of Cell and Molecular Biology, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia; Envoi Specialist Pathologists, Brisbane, Queensland, Australia
| | - Elizabeth E Powell
- Envoi Specialist Pathologists, Brisbane, Queensland, Australia; Department of Gastroenterology and Hepatology, Princess Alexandra Hospital, Brisbane, Queensland, Australia
| | - Kelli P A MacDonald
- Department of Immunology, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia.
| |
Collapse
|
130
|
Elchaninov A, Fatkhudinov T, Usman N, Kananykhina E, Arutyunyan I, Makarov A, Bolshakova G, Goldshtein D, Sukhikh G. Molecular Survey of Cell Source Usage during Subtotal Hepatectomy-Induced Liver Regeneration in Rats. PLoS One 2016; 11:e0162613. [PMID: 27631110 PMCID: PMC5025203 DOI: 10.1371/journal.pone.0162613] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Accepted: 08/11/2016] [Indexed: 02/07/2023] Open
Abstract
Proliferation of hepatocytes is known to be the main process in the hepatectomy-induced liver regrowth; however, in cases of extensive loss it may be insufficient for complete recovery unless supported by some additional sources e.g. mobilization of undifferentiated progenitors. The study was conducted on rat model of 80% subtotal hepatectomy; the objective was to evaluate contributions of hepatocytes and resident progenitor cells to the hepatic tissue recovery via monitoring specific mRNA and/or protein expression levels for a panel of genes implicated in growth, cell differentiation, angiogenesis, and inflammation. Some of the genes showed distinctive temporal expression patterns, which were loosely associated with two waves of hepatocyte proliferation observed at 2 and 7 days after the surgery. Focusing on genes implicated in regulation of the progenitor cell activity, we came across slight increases in expression levels for Sox9 and two genes encoding tumor necrosis factor-like cytokine TWEAK (Tnfsf12) and its receptor Fn14 (Tnfrsf12a). At the same time, no increase in numbers of cytokeratin 19-positive (CK19+) cells was observed in periportal areas, and no CK19+ cells were found in hepatic plates. Since CK19 is thought to be a specific marker of both cholangiocytes and the hepatic progenitor cells, the data indicate a lack of activation of the resident progenitor cells during recovery of hepatic tissue after 80% subtotal hepatectomy. Thus, proliferation of hepatocytes invariably makes the major contribution to the hepatic tissue recovery, although in the cases of subtotal loss this contribution is distinctively modulated. In particular, induction of Sox9 and TWEAK/Fn14 regulatory pathways, conventionally attributed to progenitor cell activation, may incidentally stimulate mitotic activity of hepatocytes.
Collapse
Affiliation(s)
- Andrey Elchaninov
- Research Center for Obstetrics, Gynecology and Perinatology of Ministry of Healthcare of the Russian Federation, 4 Oparina Street, Moscow 117997, Russia
- Scientific Research Institute of Human Morphology, 3 Tsurupa Street, Moscow 117418, Russia
- Pirogov Russian National Research Medical University, Ministry of Healthcare of the Russian Federation, 1 Ostrovitianov Street, Moscow 117997, Russia
| | - Timur Fatkhudinov
- Research Center for Obstetrics, Gynecology and Perinatology of Ministry of Healthcare of the Russian Federation, 4 Oparina Street, Moscow 117997, Russia
- Scientific Research Institute of Human Morphology, 3 Tsurupa Street, Moscow 117418, Russia
- * E-mail:
| | - Natalia Usman
- Research Center for Obstetrics, Gynecology and Perinatology of Ministry of Healthcare of the Russian Federation, 4 Oparina Street, Moscow 117997, Russia
- Scientific Research Institute of Human Morphology, 3 Tsurupa Street, Moscow 117418, Russia
- Pirogov Russian National Research Medical University, Ministry of Healthcare of the Russian Federation, 1 Ostrovitianov Street, Moscow 117997, Russia
| | - Evgeniya Kananykhina
- Research Center for Obstetrics, Gynecology and Perinatology of Ministry of Healthcare of the Russian Federation, 4 Oparina Street, Moscow 117997, Russia
- Scientific Research Institute of Human Morphology, 3 Tsurupa Street, Moscow 117418, Russia
| | - Irina Arutyunyan
- Research Center for Obstetrics, Gynecology and Perinatology of Ministry of Healthcare of the Russian Federation, 4 Oparina Street, Moscow 117997, Russia
- Scientific Research Institute of Human Morphology, 3 Tsurupa Street, Moscow 117418, Russia
| | - Andrey Makarov
- Research Center for Obstetrics, Gynecology and Perinatology of Ministry of Healthcare of the Russian Federation, 4 Oparina Street, Moscow 117997, Russia
- Scientific Research Institute of Human Morphology, 3 Tsurupa Street, Moscow 117418, Russia
- Pirogov Russian National Research Medical University, Ministry of Healthcare of the Russian Federation, 1 Ostrovitianov Street, Moscow 117997, Russia
| | - Galina Bolshakova
- Scientific Research Institute of Human Morphology, 3 Tsurupa Street, Moscow 117418, Russia
| | - Dmitry Goldshtein
- Research Centre of Medical Genetics, 1 Moskvorechie Street, Moscow 115478, Russia
| | - Gennady Sukhikh
- Research Center for Obstetrics, Gynecology and Perinatology of Ministry of Healthcare of the Russian Federation, 4 Oparina Street, Moscow 117997, Russia
| |
Collapse
|
131
|
Passman AM, Strauss RP, McSpadden SB, Finch-Edmondson ML, Woo KH, Diepeveen LA, London R, Callus BA, Yeoh GC. A modified choline-deficient, ethionine-supplemented diet reduces morbidity and retains a liver progenitor cell response in mice. Dis Model Mech 2015; 8:1635-41. [PMID: 26496771 PMCID: PMC4728320 DOI: 10.1242/dmm.022020] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2015] [Accepted: 10/19/2015] [Indexed: 12/25/2022] Open
Abstract
The choline-deficient, ethionine-supplemented (CDE) dietary model induces chronic liver damage, and stimulates liver progenitor cell (LPC)-mediated repair. Long-term CDE administration leads to hepatocellular carcinoma in rodents and lineage-tracing studies show that LPCs differentiate into functional hepatocytes in this model. The CDE diet was first modified for mice by our laboratory by separately administering choline-deficient chow and ethionine in the drinking water (CD+E diet). Although this CD+E diet is widely used, concerns with variability in weight loss, morbidity, mortality and LPC response have been raised by researchers who have adopted this model. We propose that these inconsistencies are due to differential consumption of chow and ethionine in the drinking water, and that incorporating ethionine in the choline-deficient chow, and altering the strength, will achieve better outcomes. Therefore, C57Bl/6 mice, 5 and 6 weeks of age, were fed an all-inclusive CDE diet of various strengths (67% to 100%) for 3 weeks. The LPC response was quantitated and cell lines were derived. We found that animal survival, LPC response and liver damage are correlated with CDE diet strength. The 67% and 75% CDE diet administered to mice older than 5 weeks and greater than 18 g provides a consistent and acceptable level of animal welfare and induces a substantial LPC response, permitting their isolation and establishment of cell lines. This study shows that an all-inclusive CDE diet for mice reproducibly induces an LPC response conducive to in vivo studies and isolation, whilst minimizing morbidity and mortality. Summary: This modified choline-deficient, ethionine-supplemented model induces liver injury in mice and reproducibly minimizes morbidity and mortality, whilst maintaining a liver-progenitor-cell response sufficient for cell-line establishment.
Collapse
Affiliation(s)
- Adam M Passman
- School of Chemistry and Biochemistry, The University of Western Australia, Crawley, Western Australia 6009, Australia Cancer and Cell Biology Division, The Harry Perkins Institute of Medical Research, Nedlands, Western Australia 6009, Australia
| | - Robyn P Strauss
- School of Chemistry and Biochemistry, The University of Western Australia, Crawley, Western Australia 6009, Australia Cancer and Cell Biology Division, The Harry Perkins Institute of Medical Research, Nedlands, Western Australia 6009, Australia
| | - Sarah B McSpadden
- School of Chemistry and Biochemistry, The University of Western Australia, Crawley, Western Australia 6009, Australia Cancer and Cell Biology Division, The Harry Perkins Institute of Medical Research, Nedlands, Western Australia 6009, Australia
| | - Megan L Finch-Edmondson
- School of Chemistry and Biochemistry, The University of Western Australia, Crawley, Western Australia 6009, Australia Cancer and Cell Biology Division, The Harry Perkins Institute of Medical Research, Nedlands, Western Australia 6009, Australia
| | - Ken H Woo
- School of Chemistry and Biochemistry, The University of Western Australia, Crawley, Western Australia 6009, Australia Cancer and Cell Biology Division, The Harry Perkins Institute of Medical Research, Nedlands, Western Australia 6009, Australia
| | - Luke A Diepeveen
- Cancer and Cell Biology Division, The Harry Perkins Institute of Medical Research, Nedlands, Western Australia 6009, Australia
| | - Roslyn London
- School of Chemistry and Biochemistry, The University of Western Australia, Crawley, Western Australia 6009, Australia Cancer and Cell Biology Division, The Harry Perkins Institute of Medical Research, Nedlands, Western Australia 6009, Australia
| | - Bernard A Callus
- School of Chemistry and Biochemistry, The University of Western Australia, Crawley, Western Australia 6009, Australia School of Health Sciences, The University of Notre Dame Australia, Fremantle, Western Australia 6959, Australia
| | - George C Yeoh
- School of Chemistry and Biochemistry, The University of Western Australia, Crawley, Western Australia 6009, Australia Cancer and Cell Biology Division, The Harry Perkins Institute of Medical Research, Nedlands, Western Australia 6009, Australia
| |
Collapse
|
132
|
Yu DD, Guo SW, Jing YY, Dong YL, Wei LX. A review on hepatocyte nuclear factor-1beta and tumor. Cell Biosci 2015; 5:58. [PMID: 26464794 PMCID: PMC4603907 DOI: 10.1186/s13578-015-0049-3] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2015] [Accepted: 10/01/2015] [Indexed: 01/06/2023] Open
Abstract
Hepatocyte nuclear factor-1beta (HNF1β) was initially identified as a liver-specific transcription factor. It is a homeobox transcription factor that functions as a homodimer or heterodimer with HNF1α. HNF1β plays an important role in organogenesis during embryonic stage, especially of the liver, kidney, and pancreas. Mutations in the HNF1β gene cause maturity-onset diabetes of the young type 5 (MODY5), renal cysts, genital malformations, and pancreas atrophy. Recently, it has been shown that the expression of HNF1β is associated with cancer risk in several tumors, including hepatocellular carcinoma, pancreatic carcinoma, renal cancer, ovarian cancer, endometrial cancer, and prostate cancer. HNF1β also regulates the expression of genes associated with stem/progenitor cells, which indicates that HNF1β may play an important role in stem cell regulation. In this review, we discuss some of the current developments about HNF1β and tumor, the relationship between HNF1β and stem/progenitor cells, and the potential pathogenesis of HNF1β in various tumors.
Collapse
Affiliation(s)
- Dan-Dan Yu
- Tumor Immunology and Gene Therapy Center, Eastern Hepatobiliary Surgery Hospital, The Second Military Medical University, 225 Changhai Road, 200438 Shanghai, China
| | - Shi-Wei Guo
- Tumor Immunology and Gene Therapy Center, Eastern Hepatobiliary Surgery Hospital, The Second Military Medical University, 225 Changhai Road, 200438 Shanghai, China
| | - Ying-Ying Jing
- Tumor Immunology and Gene Therapy Center, Eastern Hepatobiliary Surgery Hospital, The Second Military Medical University, 225 Changhai Road, 200438 Shanghai, China
| | - Yu-Long Dong
- Tumor Immunology and Gene Therapy Center, Eastern Hepatobiliary Surgery Hospital, The Second Military Medical University, 225 Changhai Road, 200438 Shanghai, China
| | - Li-Xin Wei
- Tumor Immunology and Gene Therapy Center, Eastern Hepatobiliary Surgery Hospital, The Second Military Medical University, 225 Changhai Road, 200438 Shanghai, China
| |
Collapse
|
133
|
Model of fibrolamellar hepatocellular carcinomas reveals striking enrichment in cancer stem cells. Nat Commun 2015; 6:8070. [PMID: 26437858 PMCID: PMC4600730 DOI: 10.1038/ncomms9070] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2015] [Accepted: 07/14/2015] [Indexed: 12/12/2022] Open
Abstract
The aetiology of human fibrolamellar hepatocellular carcinomas (hFL-HCCs), cancers occurring increasingly in children to young adults, is poorly understood. We present a transplantable tumour line, maintained in immune-compromised mice, and validate it as a bona fide model of hFL-HCCs by multiple methods. RNA-seq analysis confirms the presence of a fusion transcript (DNAJB1-PRKACA) characteristic of hFL-HCC tumours. The hFL-HCC tumour line is highly enriched for cancer stem cells as indicated by limited dilution tumourigenicity assays, spheroid formation and flow cytometry. Immunohistochemistry on the hFL-HCC model, with parallel studies on 27 primary hFL-HCC tumours, provides robust evidence for expression of endodermal stem cell traits. Transcriptomic analyses of the tumour line and of multiple, normal hepatic lineage stages reveal a gene signature for hFL-HCCs closely resembling that of biliary tree stem cells—newly discovered precursors for liver and pancreas. This model offers unprecedented opportunities to investigate mechanisms underlying hFL-HCCs pathogenesis and potential therapies. With no cell lines available, investigating the aetiology of human fibrolamellar hepatocellular carcinomas (hFL-HCCs) has proved problematic. Here, Oikawa et al. establish a model of hFL-HCCs as a transplantable tumour line maintained in immune-compromised mice, which proves rich in cancer stem cells.
Collapse
|
134
|
Michalopoulos GK, Khan Z. Liver Stem Cells: Experimental Findings and Implications for Human Liver Disease. Gastroenterology 2015; 149:876-882. [PMID: 26278502 PMCID: PMC4584191 DOI: 10.1053/j.gastro.2015.08.004] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2015] [Revised: 08/08/2015] [Accepted: 08/10/2015] [Indexed: 02/07/2023]
Abstract
Evidence from human histopathology and experimental studies with rodents and zebrafish has shown that hepatocytes and cholangiocytes may function as facultative stem cells for each other in conditions of impaired regeneration. The interpretation of the findings derived from these studies has generated considerable discussion and some controversies. This review examines the evidence obtained from the different experimental models and considers implications that these studies may have for human liver disease.
Collapse
Affiliation(s)
| | - Zahida Khan
- Department of Pediatric Gastroenterology University of Pittsburgh School of Medicine
| |
Collapse
|
135
|
Elsegood CL, Chan CW, Degli-Esposti MA, Wikstrom ME, Domenichini A, Lazarus K, van Rooijen N, Ganss R, Olynyk JK, Yeoh GCT. Kupffer cell-monocyte communication is essential for initiating murine liver progenitor cell-mediated liver regeneration. Hepatology 2015; 62:1272-84. [PMID: 26173184 DOI: 10.1002/hep.27977] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2014] [Revised: 06/10/2015] [Accepted: 07/04/2015] [Indexed: 12/27/2022]
Abstract
UNLABELLED Liver progenitor cells (LPCs) are necessary for repair in chronic liver disease because the remaining hepatocytes cannot replicate. However, LPC numbers also correlate with disease severity and hepatocellular carcinoma risk. Thus, the progenitor cell response in diseased liver may be regulated to optimize liver regeneration and minimize the likelihood of tumorigenesis. How this is achieved is currently unknown. Human and mouse diseased liver contain two subpopulations of macrophages with different ontogenetic origins: prenatal yolk sac-derived Kupffer cells and peripheral blood monocyte-derived macrophages. We examined the individual role(s) of Kupffer cells and monocyte-derived macrophages in the induction of LPC proliferation using clodronate liposome deletion of Kupffer cells and adoptive transfer of monocytes, respectively, in the choline-deficient, ethionine-supplemented diet model of liver injury and regeneration. Clodronate liposome treatment reduced initial liver monocyte numbers together with the induction of injury and LPC proliferation. Adoptive transfer of monocytes increased the induction of liver injury, LPC proliferation, and tumor necrosis factor-α production. CONCLUSION Kupffer cells control the initial accumulation of monocyte-derived macrophages. These infiltrating monocytes are in turn responsible for the induction of liver injury, the increase in tumor necrosis factor-α, and the subsequent proliferation of LPCs.
Collapse
Affiliation(s)
- Caryn L Elsegood
- School of Chemistry and Biochemistry, The University of Western Australia, Crawley, Western Australia, Australia.,School of Biomedical Sciences, CHIRI Biosciences Research Precinct, Curtin University, Bentley, Western Australia, Australia
| | - Chun Wei Chan
- School of Medicine and Pharmacology, The University of Western Australia, Fremantle, Western Australia, Australia.,School of Biological Sciences and Biotechnology, Murdoch University, Murdoch, Western Australia, Australia
| | - Mariapia A Degli-Esposti
- Immunology and Virology Program, Centre for Ophthalmology and Visual Science, The University of Western Australia, Nedlands, Western Australia, Australia.,Centre for Experimental Immunology, Lions Eye Institute, Nedlands, Western Australia, Australia
| | - Matthew E Wikstrom
- Immunology and Virology Program, Centre for Ophthalmology and Visual Science, The University of Western Australia, Nedlands, Western Australia, Australia.,Centre for Experimental Immunology, Lions Eye Institute, Nedlands, Western Australia, Australia
| | - Alice Domenichini
- School of Biomedical Sciences, CHIRI Biosciences Research Precinct, Curtin University, Bentley, Western Australia, Australia
| | - Kyren Lazarus
- School of Biomedical Sciences, CHIRI Biosciences Research Precinct, Curtin University, Bentley, Western Australia, Australia.,Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands and Centre for Medical Research, The University of Western Australia, Crawley, Western Australia, Australia
| | - Nico van Rooijen
- Department of Molecular Cell Biology, VU Medical Center, Amsterdam, The Netherlands
| | - Ruth Ganss
- Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands and Centre for Medical Research, The University of Western Australia, Crawley, Western Australia, Australia
| | - John K Olynyk
- School of Biomedical Sciences, CHIRI Biosciences Research Precinct, Curtin University, Bentley, Western Australia, Australia.,Department of Gastroenterology and Hepatology, Fiona Stanley and Fremantle Hospitals, South Metropolitan Health Service, Western Australia, Australia.,Institute for Immunology and Infectious Diseases, Murdoch University, Murdoch, Western Australia, Australia
| | - George C T Yeoh
- School of Chemistry and Biochemistry, The University of Western Australia, Crawley, Western Australia, Australia.,Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands and Centre for Medical Research, The University of Western Australia, Crawley, Western Australia, Australia
| |
Collapse
|
136
|
Abstract
The liver is a central regulator of metabolism, and liver failure thus constitutes a major health burden. Understanding how this complex organ develops during embryogenesis will yield insights into how liver regeneration can be promoted and how functional liver replacement tissue can be engineered. Recent studies of animal models have identified key signaling pathways and complex tissue interactions that progressively generate liver progenitor cells, differentiated lineages and functional tissues. In addition, progress in understanding how these cells interact, and how transcriptional and signaling programs precisely coordinate liver development, has begun to elucidate the molecular mechanisms underlying this complexity. Here, we review the lineage relationships, signaling pathways and transcriptional programs that orchestrate hepatogenesis.
Collapse
Affiliation(s)
- Miriam Gordillo
- Department of Surgery, Weill Cornell Medical College, New York, NY 10065, USA
| | - Todd Evans
- Department of Surgery, Weill Cornell Medical College, New York, NY 10065, USA
| | - Valerie Gouon-Evans
- Department of Developmental and Regenerative Biology, Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| |
Collapse
|
137
|
Yu DD, Jing YY, Guo SW, Ye F, Lu W, Li Q, Dong YL, Gao L, Yang YT, Yang Y, Wu MC, Wei LX. Overexpression Of Hepatocyte Nuclear Factor-1beta Predicting Poor Prognosis Is Associated With Biliary Phenotype In Patients With Hepatocellular Carcinoma. Sci Rep 2015; 5:13319. [PMID: 26311117 PMCID: PMC4550878 DOI: 10.1038/srep13319] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Accepted: 07/23/2015] [Indexed: 12/13/2022] Open
Abstract
Hepatocyte nuclear factor-1beta (HNF-1B) is involved in the hepatobiliary specification of hepatoblasts to cholangiocytes during liver development, and is strongly expressed throughout adult biliary epithelium. The aim of this study was to examine the expression of HNF-1B in different pathologic subtypes of primary liver cancer, including hepatocellular carcinoma (HCC) and cholangiocarcinoma (ICC), and the relationship between HNF-1B expression, clinicopathological features and prognosis. We retrospectively investigated 2 cohorts of patients, including 183 HCCs and 69 ICCs. The expression of HNF-1B was examined by immunohistochemistry. We found that HNF-1B expression was associated with pathological subtype of primary tumor, and HNF-1B expression in HCC tissue may be associated with the change of phenotype on recurrence. The HNF-1B expression was positively correlated with biliary/HPC (hepatic progenitor cell) markers expression. Further, multivariable analysis showed that HNF-1B expression was an independent prognostic factor for both overall survival and disease-free survival of HCC patients. However, no correlation between HNF-1B expression and survival was found in ICC patients. In summary, HCC with high HNF-1B expression displayed biliary phenotype and tended to show poorer prognosis. HNF-1B-positive malignant cells could be bipotential cells and give rise to both hepatocytic and cholangiocytic lineages during tumorigenesis.
Collapse
Affiliation(s)
- Dan-Dan Yu
- Tumor Immunology and Gene Therapy Center, Eastern Hepatobiliary Surgery Hospital, The Second Military Medical University, Shanghai, China
| | - Ying-Ying Jing
- Tumor Immunology and Gene Therapy Center, Eastern Hepatobiliary Surgery Hospital, The Second Military Medical University, Shanghai, China
| | - Shi-Wei Guo
- Tumor Immunology and Gene Therapy Center, Eastern Hepatobiliary Surgery Hospital, The Second Military Medical University, Shanghai, China
| | - Fei Ye
- Tumor Immunology and Gene Therapy Center, Eastern Hepatobiliary Surgery Hospital, The Second Military Medical University, Shanghai, China
| | - Wen Lu
- Tumor Immunology and Gene Therapy Center, Eastern Hepatobiliary Surgery Hospital, The Second Military Medical University, Shanghai, China
| | - Quan Li
- Tumor Immunology and Gene Therapy Center, Eastern Hepatobiliary Surgery Hospital, The Second Military Medical University, Shanghai, China
| | - Yu-Long Dong
- Tumor Immunology and Gene Therapy Center, Eastern Hepatobiliary Surgery Hospital, The Second Military Medical University, Shanghai, China
| | - Lu Gao
- Tumor Immunology and Gene Therapy Center, Eastern Hepatobiliary Surgery Hospital, The Second Military Medical University, Shanghai, China
| | - Yu-Ting Yang
- Tumor Immunology and Gene Therapy Center, Eastern Hepatobiliary Surgery Hospital, The Second Military Medical University, Shanghai, China
| | - Yang Yang
- Tumor Immunology and Gene Therapy Center, Eastern Hepatobiliary Surgery Hospital, The Second Military Medical University, Shanghai, China
| | - Meng-Chao Wu
- Department of Comprehensive Treatment, Eastern Hepatobiliary Surgery Hospital, The Second Military Medical University, Shanghai, China
| | - Li-Xin Wei
- Tumor Immunology and Gene Therapy Center, Eastern Hepatobiliary Surgery Hospital, The Second Military Medical University, Shanghai, China
| |
Collapse
|
138
|
Lu WY, Bird TG, Boulter L, Tsuchiya A, Cole AM, Hay T, Guest RV, Wojtacha D, Man TY, Mackinnon A, Ridgway RA, Kendall T, Williams MJ, Jamieson T, Raven A, Hay DC, Iredale JP, Clarke AR, Sansom OJ, Forbes SJ. Hepatic progenitor cells of biliary origin with liver repopulation capacity. Nat Cell Biol 2015; 17:971-983. [PMID: 26192438 PMCID: PMC4612439 DOI: 10.1038/ncb3203] [Citation(s) in RCA: 361] [Impact Index Per Article: 36.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2015] [Accepted: 06/09/2015] [Indexed: 12/13/2022]
Abstract
Hepatocytes and cholangiocytes self-renew following liver injury. Following severe injury hepatocytes are increasingly senescent, but whether hepatic progenitor cells (HPCs) then contribute to liver regeneration is unclear. Here, we describe a mouse model where the E3 ubiquitin ligase Mdm2 is inducibly deleted in more than 98% of hepatocytes, causing apoptosis, necrosis and senescence with nearly all hepatocytes expressing p21. This results in florid HPC activation, which is necessary for survival, followed by complete, functional liver reconstitution. HPCs isolated from genetically normal mice, using cell surface markers, were highly expandable and phenotypically stable in vitro. These HPCs were transplanted into adult mouse livers where hepatocyte Mdm2 was repeatedly deleted, creating a non-competitive repopulation assay. Transplanted HPCs contributed significantly to restoration of liver parenchyma, regenerating hepatocytes and biliary epithelia, highlighting their in vivo lineage potency. HPCs are therefore a potential future alternative to hepatocyte or liver transplantation for liver disease.
Collapse
Affiliation(s)
- Wei-Yu Lu
- MRC Centre for Regenerative Medicine, 5 Little France Drive, Edinburgh, EH16 4UU
| | - Thomas G Bird
- MRC Centre for Regenerative Medicine, 5 Little France Drive, Edinburgh, EH16 4UU
| | - Luke Boulter
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, Edinburgh, EH4 2XU
| | - Atsunori Tsuchiya
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Science, Niigata University, Niigata, Japan
| | - Alicia M Cole
- The CRUK Beatson Institute for Cancer Research, Switchback Road, Bearsden, Glasgow, G61 1BD
| | - Trevor Hay
- European Cancer Stem Cell Research Institute, Cardiff School of Biosciences, CF24 4HQ
| | - Rachel V Guest
- MRC Centre for Regenerative Medicine, 5 Little France Drive, Edinburgh, EH16 4UU
| | - Davina Wojtacha
- MRC Centre for Regenerative Medicine, 5 Little France Drive, Edinburgh, EH16 4UU
| | - Tak Yung Man
- MRC Centre for Regenerative Medicine, 5 Little France Drive, Edinburgh, EH16 4UU
| | - Alison Mackinnon
- MRC Centre for Regenerative Medicine, 5 Little France Drive, Edinburgh, EH16 4UU
| | - Rachel A Ridgway
- The CRUK Beatson Institute for Cancer Research, Switchback Road, Bearsden, Glasgow, G61 1BD
| | - Timothy Kendall
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, Edinburgh, EH4 2XU
| | - Michael J Williams
- MRC Centre for Regenerative Medicine, 5 Little France Drive, Edinburgh, EH16 4UU
| | - Thomas Jamieson
- The CRUK Beatson Institute for Cancer Research, Switchback Road, Bearsden, Glasgow, G61 1BD
| | - Alex Raven
- MRC Centre for Regenerative Medicine, 5 Little France Drive, Edinburgh, EH16 4UU
| | - David C Hay
- MRC Centre for Regenerative Medicine, 5 Little France Drive, Edinburgh, EH16 4UU
| | - John P Iredale
- MRC Centre for Inflammation Research, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, Scotland, United Kingdom
| | - Alan R Clarke
- European Cancer Stem Cell Research Institute, Cardiff School of Biosciences, CF24 4HQ
| | - Owen J Sansom
- The CRUK Beatson Institute for Cancer Research, Switchback Road, Bearsden, Glasgow, G61 1BD
| | - Stuart J Forbes
- MRC Centre for Regenerative Medicine, 5 Little France Drive, Edinburgh, EH16 4UU
| |
Collapse
|
139
|
Gehart H, Clevers H. Repairing organs: lessons from intestine and liver. Trends Genet 2015; 31:344-51. [DOI: 10.1016/j.tig.2015.04.005] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2015] [Revised: 04/09/2015] [Accepted: 04/10/2015] [Indexed: 12/11/2022]
|
140
|
Lemaigre FP. Determining the fate of hepatic cells by lineage tracing: facts and pitfalls. Hepatology 2015; 61:2100-3. [PMID: 25503476 DOI: 10.1002/hep.27659] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2014] [Accepted: 12/10/2014] [Indexed: 12/19/2022]
Abstract
Slow renewal of the epithelial cells by proliferation ensures homeostasis of the liver, but extensive proliferation may occur upon injury. When proliferation is impaired, transdifferentiation of mature cells or differentiation of stem cells allows production of new hepatocytes and cholangiocytes. While lineage tracings using cyclization recombinase (Cre) recombinase-mediated cell labeling represent the gold standard for defining cell fate, there are more variables than was initially realized. This led to controversies about the capacity of liver cells to switch their fate. Here, I review how cells are traced in the liver and highlight the experimental pitfalls that may cause misinterpretations and controversies.
Collapse
|
141
|
Kaneko K, Kamimoto K, Miyajima A, Itoh T. Adaptive remodeling of the biliary architecture underlies liver homeostasis. Hepatology 2015; 61:2056-66. [PMID: 25572923 DOI: 10.1002/hep.27685] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2014] [Accepted: 12/25/2014] [Indexed: 12/18/2022]
Abstract
UNLABELLED Serving as the center for metabolism and detoxification, the liver is inherently susceptible to a wide variety of damage imposed by toxins or chemicals. Induction of cell populations with biliary epithelial phenotypes, which include progenitor-like cells and are referred to as liver progenitor cells, is often observed in histopathological examination of various liver diseases in both human patients and animal models and has been implicated in regeneration. However, the tissue dynamics underlying this phenomenon remains largely unclear. We have developed a simple imaging technique to reveal the global and fine-scale architecture of the biliary tract spreading in the mouse liver. Using this novel method, we show that the emergence and expansion of liver progenitor cells actually reflect structural transformation of the intrahepatic biliary tree in mouse liver injury models. The biliary branches expanded their area gradually and contiguously along with the course of chronic injury. Relevant regulatory signals known to be involved in liver progenitor cell regulation, including fibroblast growth factor 7 and tumor necrosis factor-like weak inducer of apoptosis, can modulate the dynamics of the biliary epithelium in different ways. Importantly, the structural transformations of the biliary tree were diverse and corresponded well with the parenchymal injury patterns. That is, when chronic hepatocyte damage was induced in the pericentral area, the biliary branches exhibited an extended structure from the periportal area with apparent tropism toward the distant injured area. CONCLUSION The hepatobiliary system possesses a unique and unprecedented structural flexibility and can remodel dynamically and adaptively in response to various injury conditions; this type of tissue plasticity should constitute an essential component to maintain liver homeostasis.
Collapse
Affiliation(s)
- Kota Kaneko
- Laboratory of Cell Growth and Differentiation, Institute of Molecular and Cellular Biosciences, The University of Tokyo, Tokyo, Japan
| | - Kenji Kamimoto
- Laboratory of Cell Growth and Differentiation, Institute of Molecular and Cellular Biosciences, The University of Tokyo, Tokyo, Japan
| | - Atsushi Miyajima
- Laboratory of Cell Growth and Differentiation, Institute of Molecular and Cellular Biosciences, The University of Tokyo, Tokyo, Japan
| | - Tohru Itoh
- Laboratory of Cell Growth and Differentiation, Institute of Molecular and Cellular Biosciences, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
142
|
Stueck AE, Wanless IR. Hepatocyte buds derived from progenitor cells repopulate regions of parenchymal extinction in human cirrhosis. Hepatology 2015; 61:1696-707. [PMID: 25644399 DOI: 10.1002/hep.27706] [Citation(s) in RCA: 74] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2014] [Accepted: 01/08/2015] [Indexed: 01/01/2023]
Abstract
UNLABELLED Repair of cirrhotic livers occurs, in part, by repopulation with hepatocytes through the stem/progenitor pathway. There remain many uncertainties regarding this pathway. Hepatocyte "buds" occurring in broad septa are hypothesized to be the anatomic manifestation of this pathway. Our purpose was to define a morphologic sequence of bud maturation to allow a quantitative measure of the importance of the stem/progenitor pathway in humans. Histologic sections from 37 liver resection specimens were stained with trichrome, epithelial cell adhesion molecule (EpCAM), K19, CD34, glutamine synthetase (GS), and Ki-67. Specimens were stratified by etiology (10 biliary, 22 nonbiliary, five controls) and stage. Buds were defined as clusters of hepatocytes within septa. Five levels of bud maturation (0-4) were defined by the progressive increase in hepatocyte progeny relative to cholangiocytes. Level 0 single-cell buds are K19(+) /GS(+) /EpCAM(+) /Heppar1(-) . In level 1, the progeny are morphologically hepatocytes (K19(-) /GS(+) /EpCAM(+) /Heppar1(+) ). In level 2-4 buds, hepatocytes increase and become progressively GS(-) and EpCAM(-) . Associated endothelium is CD34(+) in level 1-2 buds and becomes CD34(-) near hepatic veins in level 3-4 buds. Progeny of the bud sequence may represent up to 70% of hepatocytes (immaturity index of 70%). In biliary disease, bud number is reduced in association with duct loss and cholestatic destruction of nascent buds. CONCLUSIONS The stem/progenitor pathway, manifested anatomically by the bud sequence, is a major mechanism for repopulation of cirrhotic livers. The bud sequence reveals some critical features of hepatic morphogenesis, including that 1) the majority of distal cholangiocytes have stem-like properties, and 2) availability of bile ducts and/or venous drainage are limiting factors for regeneration.
Collapse
Affiliation(s)
- Ashley E Stueck
- Department of Pathology, Queen Elizabeth II Health Sciences Centre, Halifax, NS, Canada
| | | |
Collapse
|
143
|
Jörs S, Jeliazkova P, Ringelhan M, Thalhammer J, Dürl S, Ferrer J, Sander M, Heikenwalder M, Schmid RM, Siveke JT, Geisler F. Lineage fate of ductular reactions in liver injury and carcinogenesis. J Clin Invest 2015; 125:2445-57. [PMID: 25915586 DOI: 10.1172/jci78585] [Citation(s) in RCA: 133] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2014] [Accepted: 03/20/2015] [Indexed: 12/13/2022] Open
Abstract
Ductular reactions (DRs) are observed in virtually all forms of human liver disease; however, the histogenesis and function of DRs in liver injury are not entirely understood. It is widely believed that DRs contain bipotential liver progenitor cells (LPCs) that serve as an emergency cell pool to regenerate both cholangiocytes and hepatocytes and may eventually give rise to hepatocellular carcinoma (HCC). Here, we used a murine model that allows highly efficient and specific lineage labeling of the biliary compartment to analyze the histogenesis of DRs and their potential contribution to liver regeneration and carcinogenesis. In multiple experimental and genetic liver injury models, biliary cells were the predominant precursors of DRs but lacked substantial capacity to produce new hepatocytes, even when liver injuries were prolonged up to 12 months. Genetic modulation of NOTCH and/or WNT/β-catenin signaling within lineage-tagged DRs impaired DR expansion but failed to redirect DRs from biliary differentiation toward the hepatocyte lineage. Further, lineage-labeled DRs did not produce tumors in genetic and chemical HCC mouse models. In summary, we found no evidence in our system to support mouse biliary-derived DRs as an LPC pool to replenish hepatocytes in a quantitatively relevant way in injury or evidence that DRs give rise to HCCs.
Collapse
|
144
|
Kok CYY, Miyajima A, Itoh T. Adaptive remodeling of the biliary tree: the essence of liver progenitor cell expansion. JOURNAL OF HEPATO-BILIARY-PANCREATIC SCIENCES 2015; 22:546-50. [PMID: 25900773 DOI: 10.1002/jhbp.250] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/02/2015] [Accepted: 03/12/2015] [Indexed: 12/23/2022]
Abstract
The liver progenitor cell population has long been thought to exist within the liver. However, there are no standardized criteria for defining the liver progenitor cells, and there has been intense debate about the origin of these cells in the adult liver. The characteristics of such cells vary depending on the disease model used and also on the method of analysis. Visualization of three-dimensional biliary structures has revealed that the emergence of liver progenitor cells essentially reflects the adaptive remodeling of the hepatic biliary network in response to liver injury. We propose that the progenitor cell exists as a subpopulation in the biliary tree and show that the appearance of liver progenitor cells in injured parenchyma is reflective of extensive remodeling of the biliary structure.
Collapse
Affiliation(s)
- Cindy Yuet-Yin Kok
- Laboratory of Cell Growth and Differentiation, Institute of Molecular and Cellular Biosciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo, 113-0032, Japan
| | - Atsushi Miyajima
- Laboratory of Cell Growth and Differentiation, Institute of Molecular and Cellular Biosciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo, 113-0032, Japan
| | - Tohru Itoh
- Laboratory of Cell Growth and Differentiation, Institute of Molecular and Cellular Biosciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo, 113-0032, Japan.
| |
Collapse
|
145
|
Dollé L, Boulter L, Leclercq IA, van Grunsven LA. Next generation of ALDH substrates and their potential to study maturational lineage biology in stem and progenitor cells. Am J Physiol Gastrointest Liver Physiol 2015; 308:G573-8. [PMID: 25656041 PMCID: PMC4385895 DOI: 10.1152/ajpgi.00420.2014] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2014] [Accepted: 02/03/2015] [Indexed: 01/31/2023]
Abstract
High aldehyde dehydrogenase (ALDH) activity is a feature of stem cells from normal and cancerous tissues and a reliable universal marker used to isolate them. There are numerous ALDH isoforms with preferred substrate specificity variably expressed depending on tissue, cell type, and organelle and cell status. On the other hand, a given substrate may be metabolized by several enzyme isoforms. Currently ALDH activity is evidenced by using Aldefluor, a fluorescent substrate likely to be metabolized by numerous ALDH isoforms. Therefore, isolation techniques based on ALDH activity detection select a heterogeneous population of stem or progenitor cells. Despite active research in the field, the precise role(s) of different ALDH isoforms in stem cells remains enigmatic. Understanding the metabolic role of different ALDH isoform in the control of stem cell phenotype and cell fate during development, tissue homeostasis, or repair, as well as carcinogenesis, should open perspectives to significant discoveries in tissue biology. In this perspective, novel ALDH substrates are being developed. Here we describe how new substrates could be instrumental for better isolation of cell population with stemness potential and for defining hierarchy of cell populations in tissue. Finally, we speculate on other potential applications.
Collapse
Affiliation(s)
- Laurent Dollé
- Liver Cell Biology Lab, Vrije Universiteit Brussel (VUB), Brussels, Belgium;
| | - Luke Boulter
- 2MRC Human Genetics Unit, Institute for Genetics and Molecular Medicine, Edinburgh, United Kingdom; and
| | - Isabelle A. Leclercq
- 3Laboratory of Hepato-Gastroenterology, Institut de Recherche Expérimentale et Clinique (IREC), Université catholique de Louvain (UCL), Brussels, Belgium
| | - Leo A. van Grunsven
- 1Liver Cell Biology Lab, Vrije Universiteit Brussel (VUB), Brussels, Belgium;
| |
Collapse
|
146
|
Verhulst S, Best J, van Grunsven LA, Dollé L. Advances in hepatic stem/progenitor cell biology. EXCLI JOURNAL 2015; 14:33-47. [PMID: 26600740 PMCID: PMC4650945 DOI: 10.17179/excli2014-576] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/05/2014] [Accepted: 10/23/2014] [Indexed: 12/16/2022]
Abstract
The liver is famous for its strong regenerative capacity, employing different modes of regeneration according to type and extent of injury. Mature liver cells are able to proliferate in order to replace the damaged tissue allowing the recovery of the parenchymal function. In more severe scenarios hepatocytes are believed to arise also from a facultative liver progenitor cell compartment. In human, severe acute liver failure and liver cirrhosis are also both important clinical targets in which regeneration is impaired, where the role of this stem cell compartment seems more convincing. In animal models, the current state of ambiguity regarding the identity and role of liver progenitor cells in liver physiology dampens the enthusiasm for the potential use of these cells in regenerative medicine. The aim of this review is to give the basics of liver progenitor cell biology and discuss recent results vis-à-vis their identity and contribution to liver regeneration.
Collapse
Affiliation(s)
- Stefaan Verhulst
- Liver Cell Biology Laboratory, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Jan Best
- Liver Cell Biology Laboratory, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Leo A. van Grunsven
- Liver Cell Biology Laboratory, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Laurent Dollé
- Liver Cell Biology Laboratory, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| |
Collapse
|
147
|
Chen KT, Pernelle K, Tsai YH, Wu YH, Hsieh JY, Liao KH, Guguen-Guillouzo C, Wang HW. Liver X receptor α (LXRα/NR1H3) regulates differentiation of hepatocyte-like cells via reciprocal regulation of HNF4α. J Hepatol 2014; 61:1276-86. [PMID: 25073010 DOI: 10.1016/j.jhep.2014.07.025] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2013] [Revised: 07/10/2014] [Accepted: 07/14/2014] [Indexed: 12/12/2022]
Abstract
BACKGROUND & AIMS Hepatocyte-like cells, differentiated from different stem cell sources, are considered to have a range of possible therapeutic applications, including drug discovery, metabolic disease modelling, and cell transplantation. However, little is known about how stem cells differentiate into mature and functional hepatocytes. METHODS Using transcriptomic screening, a transcription factor, liver X receptor α (NR1H3), was identified as increased during HepaRG cell hepatogenesis; this protein was also upregulated during embryonic stem cell and induced pluripotent stem cell differentiation. RESULTS Overexpressing NR1H3 in human HepaRG cells promoted hepatic maturation; the hepatocyte-like cells exhibited various functions associated with mature hepatocytes, including cytochrome P450 (CYP) enzyme activity, secretion of urea and albumin, upregulation of hepatic-specific transcripts and an increase in glycogen storage. Importantly, the NR1H3-derived hepatocyte-like cells were able to rescue lethal fulminant hepatic failure using a non-obese diabetic/severe combined immunodeficient mouse model. CONCLUSIONS In this study, we found that NR1H3 accelerates hepatic differentiation through an HNF4α-dependent reciprocal network. This contributes to hepatogenesis and is therapeutically beneficial to liver disease.
Collapse
Affiliation(s)
- Kai-Ting Chen
- Taiwan International Graduate Program in Molecular Medicine, National Yang-Ming University and Academia Sinica, Taipei, Taiwan; Institute of Microbiology and Immunology, National Yang-Ming University, Taipei, Taiwan; Institute of Biochemistry and Molecular Biology, National Yang-Ming University, Taipei, Taiwan
| | - Kelig Pernelle
- Inserm UMR 991, Université de Rennes 1, Faculté de médecine, F-35043 Rennes cedex, France
| | - Yuan-Hau Tsai
- Institute of Microbiology and Immunology, National Yang-Ming University, Taipei, Taiwan
| | - Yu-Hsuan Wu
- Institute of Microbiology and Immunology, National Yang-Ming University, Taipei, Taiwan
| | - Jui-Yu Hsieh
- Institute of Microbiology and Immunology, National Yang-Ming University, Taipei, Taiwan
| | - Ko-Hsun Liao
- Institute of Microbiology and Immunology, National Yang-Ming University, Taipei, Taiwan
| | - Christiane Guguen-Guillouzo
- Inserm UMR 991, Université de Rennes 1, Faculté de médecine, F-35043 Rennes cedex, France; Biopredic international, Parc d'activité Bretèche batA4, 35760 Saint-Grégoire, France
| | - Hsei-Wei Wang
- Taiwan International Graduate Program in Molecular Medicine, National Yang-Ming University and Academia Sinica, Taipei, Taiwan; Institute of Microbiology and Immunology, National Yang-Ming University, Taipei, Taiwan; YM-VGH Genome Research Center, National Yang-Ming University, Taipei, Taiwan; Department of Education and Research, Taipei City Hospital, Taipei, Taiwan.
| |
Collapse
|
148
|
Xu LB, Liu C. Role of liver stem cells in hepatocarcinogenesis. World J Stem Cells 2014; 6:579-590. [PMID: 25426254 PMCID: PMC4178257 DOI: 10.4252/wjsc.v6.i5.579] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2014] [Revised: 08/24/2014] [Accepted: 09/01/2014] [Indexed: 02/06/2023] Open
Abstract
Liver cancer is an aggressive disease with a high mortality rate. Management of liver cancer is strongly dependent on the tumor stage and underlying liver disease. Unfortunately, most cases are discovered when the cancer is already advanced, missing the opportunity for surgical resection. Thus, an improved understanding of the mechanisms responsible for liver cancer initiation and progression will facilitate the detection of more reliable tumor markers and the development of new small molecules for targeted therapy of liver cancer. Recently, there is increasing evidence for the “cancer stem cell hypothesis”, which postulates that liver cancer originates from the malignant transformation of liver stem/progenitor cells (liver cancer stem cells). This cancer stem cell model has important significance for understanding the basic biology of liver cancer and has profound importance for the development of new strategies for cancer prevention and treatment. In this review, we highlight recent advances in the role of liver stem cells in hepatocarcinogenesis. Our review of the literature shows that identification of the cellular origin and the signaling pathways involved is challenging issues in liver cancer with pivotal implications in therapeutic perspectives. Although the dedifferentiation of mature hepatocytes/cholangiocytes in hepatocarcinogenesis cannot be excluded, neoplastic transformation of a stem cell subpopulation more easily explains hepatocarcinogenesis. Elimination of liver cancer stem cells in liver cancer could result in the degeneration of downstream cells, which makes them potential targets for liver cancer therapies. Therefore, liver stem cells could represent a new target for therapeutic approaches to liver cancer in the near future.
Collapse
|
149
|
Hindley CJ, Mastrogiovanni G, Huch M. The plastic liver: differentiated cells, stem cells, every cell? J Clin Invest 2014; 124:5099-102. [PMID: 25401467 DOI: 10.1172/jci78372] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The liver is capable of full regeneration following several types and rounds of injury, ranging from hepatectomy to toxin-mediated damage. The source of this regenerative capacity has long been a hotly debated topic. The damage response that occurs when hepatocyte proliferation is impaired is thought to be mediated by oval/dedifferentiated progenitor cells, which replenish the hepatocyte and ductal compartments of the liver. Recently, reports have questioned whether these oval/progenitor cells truly serve as the facultative stem cell of the liver following toxin-mediated damage. In this issue of the JCI, Kordes and colleagues use lineage tracing to follow transplanted rat hepatic stellate cells, a resident liver mesenchymal cell population, in hosts that have suffered liver damage. Transplanted stellate cells repopulated the damaged rat liver by contributing to the oval cell response. These data establish yet another cell type of mesenchymal origin as the progenitor for the oval/ductular response in the rat. The lack of uniformity between different damage models, the extent of the injury to the liver parenchyma, and potential species-specific differences might be at the core of the discrepancy between different studies. Taken together, these data imply a considerable degree of plasticity in the liver, whereby several cell types can contribute to regeneration.
Collapse
|
150
|
Tarlow BD, Pelz C, Naugler WE, Wakefield L, Wilson EM, Finegold MJ, Grompe M. Bipotential adult liver progenitors are derived from chronically injured mature hepatocytes. Cell Stem Cell 2014; 15:605-18. [PMID: 25312494 DOI: 10.1016/j.stem.2014.09.008] [Citation(s) in RCA: 409] [Impact Index Per Article: 37.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2014] [Revised: 08/14/2014] [Accepted: 09/17/2014] [Indexed: 02/07/2023]
Abstract
Adult liver progenitor cells are biliary-like epithelial cells that emerge only under injury conditions in the periportal region of the liver. They exhibit phenotypes of both hepatocytes and bile ducts. However, their origin and their significance to injury repair remain unclear. Here, we used a chimeric lineage tracing system to demonstrate that hepatocytes contribute to the progenitor pool. RNA-sequencing, ultrastructural analysis, and in vitro progenitor assays revealed that hepatocyte-derived progenitors were distinct from their biliary-derived counterparts. In vivo lineage tracing and serial transplantation assays showed that hepatocyte-derived proliferative ducts retained a memory of their origin and differentiated back into hepatocytes upon cessation of injury. Similarly, human hepatocytes in chimeric mice also gave rise to biliary progenitors in vivo. We conclude that human and mouse hepatocytes can undergo reversible ductal metaplasia in response to injury, expand as ducts, and subsequently contribute to restoration of the hepatocyte mass.
Collapse
Affiliation(s)
- Branden D Tarlow
- Department of Cell, Developmental, and Cancer Biology, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239, USA; Department of Pediatrics, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239, USA.
| | - Carl Pelz
- Department of Pediatrics, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239, USA
| | - Willscott E Naugler
- Department of Gastroenterology & Hepatology, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239, USA
| | - Leslie Wakefield
- Department of Pediatrics, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239, USA
| | | | - Milton J Finegold
- Department of Pathology, Baylor College of Medicine, 6621 Fannin Street, Houston, TX 77030, USA
| | - Markus Grompe
- Department of Pediatrics, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239, USA
| |
Collapse
|