101
|
Fn14 hepatic progenitor cells are associated with liver fibrosis in biliary atresia. Pediatr Surg Int 2017; 33:593-599. [PMID: 28180936 DOI: 10.1007/s00383-017-4068-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/18/2017] [Indexed: 12/19/2022]
Abstract
PURPOSE The liver in biliary atresia (BA) is characterized by progressing fibrosis which is promoted by unclear reasons. We aimed to understand the factors influencing liver fibrosis. This study hypothesized that HPCs (hepatic progenitor cells) are activated and associated with liver fibrosis in biliary atresia. METHODS Liver samples from biliary atresia patients are as BA group, and the normal liver derived from hepatoblastoma infants during operation are control group. The extent of fibrosis in liver samples was blindly evaluated by two experienced pathologists depending on Ishak system. The BA liver samples were divided into mild liver fibrosis group (grade I-IV, BAa) and severe liver fibrosis group (grade V-VI, BAb) to detect Fn14 protein expression. RESULTS In mRNA level, Fn14 expression was 21.23 ± 8.3 vs. 1.00 ± 0.17, p = 0.023 < 0.05 and CD133 expression was 6.02 ± 2.16 vs. 1.14 ± 0.75, p = 0.008 < 0.01 between BA group and control group. Fn14 cells co-expressed the progenitor marker CD133 in liver, and activated in BA. Fn14 andα-SMA were co-location in fibrous area in liver. Compared to the control group, Fn14, CD133, and α-SMA protein expression were 2.10 ± 0.53 vs. 0.97 ± 0.2, p = 0.001, 2.23 ± 0.57 vs. 1.00 ± 0.03, p = 0.000, 4.96 ± 2.4 vs. 1.00 ± 0.22, p = 0.001. The Fn14 protein expression was 2.60 ± 0.35 vs. 1.86 ± 0.42, p = 0.012, between BAb and BAa group. CONCLUSION Fn14 cells, which co-express the progenitor marker CD133 in liver, are HPCs and activated in BA. Fn14 + HPCs are associated with liver fibrosis in BA.
Collapse
|
102
|
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: 36] [Impact Index Per Article: 4.5] [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
|
103
|
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
|
104
|
Involvement of prolyl isomerase PIN1 in the cell cycle progression and proliferation of hepatic oval cells. Pathol Res Pract 2017; 213:373-380. [PMID: 28214206 DOI: 10.1016/j.prp.2017.01.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Revised: 01/06/2017] [Accepted: 01/06/2017] [Indexed: 12/29/2022]
Abstract
Liver regenerates remarkably after toxic injury or surgical resection. In the case of failure of resident hepatocytes to restore loss, repopulation is carried out by induction, proliferation, and differentiation of the progenitor cell. Although, some signaling pathways have been verified to contribute oval cell-mediated liver regeneration, role of Peptidyl-prolyl cis-trans isomerase NIMA-interacting 1(Pin1) in the oval cells proliferation is unknown. In the present study, we evaluate the role of Pin1 in oval cells proliferation. In our study, the expression of Pin1 in the mice liver increased after three weeks feeding of 3, 5-diethoxycarbonyl-1, 4-dihydrocollidine (DDC) diet along with the proliferation of oval cells. The expression of Pin1 was higher in oval cells compared to the hepatocytes.Pin1 inhibition by Juglone reduced oval cell proliferation, which was restored to normal when oval cells were treated with IGF-1. Consistent with increased cell growth, expression of Pin1, β-catenin and PCNA were increased in IGF-1 treated cells in a time dependent manner. In FACS analysis, siRNA-mediated knockdown of the Pin1 protein in the oval cells significantly increased the numbers of cells in G0/G1 phase. Furthermore, hepatocyte when treated with TGF-β showed marked reduction in cell proliferation and expression of Pin1 whereas this effect was not seen in the oval cells treated with TGF-β. In conclusion, Pin1 plays important role in the cell cycle progression and increase oval cells proliferation which may be crucial in chronic liver injury.
Collapse
|
105
|
Haideri SS, McKinnon AC, Taylor AH, Kirkwood P, Starkey Lewis PJ, O’Duibhir E, Vernay B, Forbes S, Forrester LM. Injection of embryonic stem cell derived macrophages ameliorates fibrosis in a murine model of liver injury. NPJ Regen Med 2017; 2:14. [PMID: 29302350 PMCID: PMC5677947 DOI: 10.1038/s41536-017-0017-0] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Revised: 02/27/2017] [Accepted: 04/02/2017] [Indexed: 12/18/2022] Open
Abstract
Chronic liver injury can be caused by viral hepatitis, alcohol, obesity, and metabolic disorders resulting in fibrosis, hepatic scarring, and cirrhosis. Novel therapies are urgently required and previous work has demonstrated that treatment with bone marrow derived macrophages can improve liver regeneration and reduce fibrosis in a murine model of hepatic injury and fibrosis. Here, we describe a protocol whereby pure populations of therapeutic macrophages can be produced in vitro from murine embryonic stem cells on a large scale. Embryonic stem cell derived macrophages display comparable morphology and cell surface markers to bone marrow derived macrophages but our novel imaging technique revealed that their phagocytic index was significantly lower. Differences were also observed in their response to classical induction protocols with embryonic stem cell derived macrophages having a reduced response to lipopolysaccharide and interferon gamma and an enhanced response to IL4 compared to bone marrow derived macrophages. When their therapeutic potential was assessed in a murine, carbon tetrachloride-induced injury and fibrosis model, embryonic stem cell derived macrophages significantly reduced the amount of hepatic fibrosis to 50% of controls, down-regulated the number of fibrogenic myofibroblasts and activated liver progenitor cells. To our knowledge, this is the first study that demonstrates a therapeutic effect of macrophages derived in vitro from pluripotent stem cells in a model of liver injury. We also found that embryonic stem cell derived macrophages repopulated the Kupffer cell compartment of clodronate-treated mice more efficiently than bone marrow derived macrophages, and expressed comparatively lower levels of Myb and Ccr2, indicating that their phenotype is more comparable to tissue-resident rather than monocyte-derived macrophages.
Collapse
Affiliation(s)
- Sharmin S. Haideri
- 0000 0004 1936 7988grid.4305.2Centre for Regenerative Medicine, University of Edinburgh, 5 Little France Drive,, Edinburgh, EH16 4UU UK
| | - Alison C. McKinnon
- 0000 0004 1936 7988grid.4305.2Centre for Regenerative Medicine, University of Edinburgh, 5 Little France Drive,, Edinburgh, EH16 4UU UK
| | - A. Helen Taylor
- 0000 0004 1936 7988grid.4305.2Centre for Regenerative Medicine, University of Edinburgh, 5 Little France Drive,, Edinburgh, EH16 4UU UK
| | - Phoebe Kirkwood
- 0000 0004 1936 7988grid.4305.2Centre for Regenerative Medicine, University of Edinburgh, 5 Little France Drive,, Edinburgh, EH16 4UU UK
| | - Philip J. Starkey Lewis
- 0000 0004 1936 7988grid.4305.2Centre for Regenerative Medicine, University of Edinburgh, 5 Little France Drive,, Edinburgh, EH16 4UU UK
| | - Eoghan O’Duibhir
- 0000 0004 1936 7988grid.4305.2Centre for Regenerative Medicine, University of Edinburgh, 5 Little France Drive,, Edinburgh, EH16 4UU UK
| | - Bertrand Vernay
- 0000 0004 1936 7988grid.4305.2Centre for Regenerative Medicine, University of Edinburgh, 5 Little France Drive,, Edinburgh, EH16 4UU UK
| | - Stuart Forbes
- 0000 0004 1936 7988grid.4305.2Centre for Regenerative Medicine, University of Edinburgh, 5 Little France Drive,, Edinburgh, EH16 4UU UK
| | - Lesley M. Forrester
- 0000 0004 1936 7988grid.4305.2Centre for Regenerative Medicine, University of Edinburgh, 5 Little France Drive,, Edinburgh, EH16 4UU UK
| |
Collapse
|
106
|
Zhang F, Zhang M, Wang A, Xu M, Wang C, Xu G, Zhang B, Zou X, Zhuge Y. TWEAK increases SIRT1 expression and promotes p53 deacetylation affecting human hepatic stellate cell senescence. Cell Biol Int 2016; 41:147-154. [PMID: 27888541 DOI: 10.1002/cbin.10706] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Accepted: 11/19/2016] [Indexed: 01/03/2023]
Abstract
To detect the effects of tumor necrosis factor-like weak inducer of apoptosis (TWEAK) on SIRT1 expression and p53 deacetylation, involving cell senescence, in activated human hepatic stellate cell (HSC) in vitro, human HSC LX-2 was cultured with TWEAK for 24 h. The result showed that the expression of membrane receptor Fn14 was remarkably increased by TWEAK, which upregulated SIRT1 in LX-2 cells, detected by Western blotting and real-time PCR. The expression of p53 was not significantly altered; however, the ac-p53 was decreased. Furthermore, the viability of LX-2 cells was significantly enhanced by TWEAK. The activity of SA-β-Gal was notably inhibited, showing a suppressing effect of TWEAK on the senescence of activated HSC. Primary cultured HSC on days 7 and 11 was used to examine the expression of TWEAK, Fn14, SIRT1, and the activity of SA-β-Gal. The result indicated that the mRNA of TWEAK, SIRT1, and Fn14 was all decreased on day 11 compared to that on day 7, and the activity of SA-β-Gal was higher on day 11 than that on day 7. The present study suggested that TWEAK enhanced the expression of SIRT1 and decreased the acetylation of p53, probably inhibiting the senescence of activated HSC in vitro, which provides a molecular basis for TWEAK as a potential target in the therapy of liver fibrosis.
Collapse
Affiliation(s)
- Feng Zhang
- Department of Gastroenterology, Affiliated Drum Tower Hospital of Nanjing University Medical School, 321# Zhongshan Road, Nanjing 210008, Jiangsu, China
| | - Ming Zhang
- Department of Gastroenterology, Affiliated Drum Tower Hospital of Nanjing University Medical School, 321# Zhongshan Road, Nanjing 210008, Jiangsu, China
| | - Aixiu Wang
- Department of Gastroenterology, Affiliated Drum Tower Hospital of Nanjing University Medical School, 321# Zhongshan Road, Nanjing 210008, Jiangsu, China
| | - Mingcui Xu
- Department of Gastroenterology, Affiliated Drum Tower Hospital of Nanjing University Medical School, 321# Zhongshan Road, Nanjing 210008, Jiangsu, China
| | - Chen Wang
- Department of Gastroenterology, Affiliated Drum Tower Hospital of Nanjing University Medical School, 321# Zhongshan Road, Nanjing 210008, Jiangsu, China
| | - Guifang Xu
- Department of Gastroenterology, Affiliated Drum Tower Hospital of Nanjing University Medical School, 321# Zhongshan Road, Nanjing 210008, Jiangsu, China
| | - Bin Zhang
- Department of Gastroenterology, Affiliated Drum Tower Hospital of Nanjing University Medical School, 321# Zhongshan Road, Nanjing 210008, Jiangsu, China
| | - Xiaoping Zou
- Department of Gastroenterology, Affiliated Drum Tower Hospital of Nanjing University Medical School, 321# Zhongshan Road, Nanjing 210008, Jiangsu, China
| | - Yuzheng Zhuge
- Department of Gastroenterology, Affiliated Drum Tower Hospital of Nanjing University Medical School, 321# Zhongshan Road, Nanjing 210008, Jiangsu, China
| |
Collapse
|
107
|
Magro-Checa C, Zirkzee EJ, Huizinga TW, Steup-Beekman GM. Management of Neuropsychiatric Systemic Lupus Erythematosus: Current Approaches and Future Perspectives. Drugs 2016; 76:459-83. [PMID: 26809245 PMCID: PMC4791452 DOI: 10.1007/s40265-015-0534-3] [Citation(s) in RCA: 126] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Neuropsychiatric systemic lupus erythematosus (NPSLE) is a generic definition referring to a series of neurological and psychiatric symptoms directly related to systemic lupus erythematosus (SLE). NPSLE includes heterogeneous and rare neuropsychiatric (NP) manifestations involving both the central and peripheral nervous system. Due to the lack of a gold standard, the attribution of NP symptoms to SLE represents a clinical challenge that obligates the strict exclusion of any other potential cause. In the acute setting, management of these patients does not differ from other non-SLE subjects presenting with the same NP manifestation. Afterwards, an individualized therapeutic strategy, depending on the presenting manifestation and severity of symptoms, must be started. Clinical trials in NPSLE are scarce and most of the data are extracted from case series and case reports. High-dose glucocorticoids and intravenous cyclophosphamide remain the cornerstone for patients with severe symptoms that are thought to reflect inflammation or an underlying autoimmune process. Rituximab, intravenous immunoglobulins, or plasmapheresis may be used if response is not achieved. When patients present with mild to moderate NP manifestations, or when maintenance therapy is warranted, azathioprine and mycophenolate may be considered. When symptoms are thought to reflect a thrombotic underlying process, anticoagulation and antiplatelet agents are the mainstay of therapy, especially if antiphospholipid antibodies or antiphospholipid syndrome are present. Recent trials on SLE using new biologicals, based on newly understood SLE mechanisms, have shown promising results. Based on what we currently know about its pathogenesis, it is tempting to speculate how these new therapies may affect the management of NPSLE patients. This article provides a comprehensive and critical review of the literature on the epidemiology, pathophysiology, diagnosis, and management of NPSLE. We describe the most common pharmacological treatments used in NPSLE, based on both a literature search and our expert opinion. The extent to which new drugs in the advanced development of SLE, or the blockade of new targets, may impact future treatment of NPSLE will also be discussed.
Collapse
Affiliation(s)
- César Magro-Checa
- Department of Rheumatology, Leiden University Medical Center, PO Box 9600, 2300 RC, Leiden, The Netherlands
| | - Elisabeth J Zirkzee
- Department of Rheumatology, Leiden University Medical Center, PO Box 9600, 2300 RC, Leiden, The Netherlands.,Department of Rheumatology, Maasstad Hospital, Rotterdam, The Netherlands
| | - Tom W Huizinga
- Department of Rheumatology, Leiden University Medical Center, PO Box 9600, 2300 RC, Leiden, The Netherlands
| | - Gerda M Steup-Beekman
- Department of Rheumatology, Leiden University Medical Center, PO Box 9600, 2300 RC, Leiden, The Netherlands.
| |
Collapse
|
108
|
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
|
109
|
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
|
110
|
Suzuki Y, Katagiri H, Wang T, Kakisaka K, Kume K, Nishizuka SS, Takikawa Y. Ductular reactions in the liver regeneration process with local inflammation after physical partial hepatectomy. J Transl Med 2016; 96:1211-1222. [PMID: 27617400 DOI: 10.1038/labinvest.2016.97] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2016] [Revised: 07/27/2016] [Accepted: 08/09/2016] [Indexed: 01/06/2023] Open
Abstract
Partial hepatectomy models in mice have been widely used for liver regeneration studies. A typical procedure removes ~2/3 of the liver by lobular ligation without tissue dissection. However, hepatectomy in humans involves physical damage (ie, physical partial hepatectomy, PPHx). Therefore, the liver regeneration process after PPHx should involve reactions to acute local injury followed by systematic remodeling. To clarify the liver regeneration process after PPHx, we used a murine liver injury model that mimics the actual human surgical procedure. A 20-30% PPHx was performed by transection of the left lobe of the liver using an ultrasonically activated scalpel in mice. Gene expression and morphological characteristics were analyzed during the liver regeneration process. Liver weight continuously increased by hypertrophic reaction of hepatocytes, whereas Ki67 staining showed hepatocyte proliferation. At the transected border, emergence of ductular reactions, a representative process of hepatic tissue remodeling that contain liver stem/progenitor cells, were observed. Gene expression of the transected border and non-damaged lobes revealed that inflammatory cytokine- and extracellular matrix-associated genes were significantly upregulated at the transected border. Our PPHx model triggered local extracellular matrix remodeling that resulted in ductular reactions. These processes occurred during the tissue repair process in local inflammatory responses as well as compensatory hepatocyte hypertrophy of the entire liver. These findings may provide insight for elucidating the mechanism of tissue repair and regeneration of the liver after PPHx.
Collapse
Affiliation(s)
- Yuji Suzuki
- Molecular Therapeutics Laboratory, Department of Surgery, School of Medicine, Iwate Medical University, Morioka, Japan.,Division of Hepatology, Department of Internal Medicine, School of Medicine, Iwate Medical University, Morioka, Japan
| | - Hirokatsu Katagiri
- Molecular Therapeutics Laboratory, Department of Surgery, School of Medicine, Iwate Medical University, Morioka, Japan
| | - Ting Wang
- Division of Hepatology, Department of Internal Medicine, School of Medicine, Iwate Medical University, Morioka, Japan
| | - Keisuke Kakisaka
- Division of Hepatology, Department of Internal Medicine, School of Medicine, Iwate Medical University, Morioka, Japan
| | - Kohei Kume
- Molecular Therapeutics Laboratory, Department of Surgery, School of Medicine, Iwate Medical University, Morioka, Japan.,Institute of Biomedical Sciences, Iwate Medical University, Yahaba, Japan
| | - Satoshi S Nishizuka
- Molecular Therapeutics Laboratory, Department of Surgery, School of Medicine, Iwate Medical University, Morioka, Japan.,Institute of Biomedical Sciences, Iwate Medical University, Yahaba, Japan
| | - Yasuhiro Takikawa
- Division of Hepatology, Department of Internal Medicine, School of Medicine, Iwate Medical University, Morioka, Japan
| |
Collapse
|
111
|
Tanaka M, Miyajima A. Liver regeneration and fibrosis after inflammation. Inflamm Regen 2016; 36:19. [PMID: 29259692 PMCID: PMC5725806 DOI: 10.1186/s41232-016-0025-2] [Citation(s) in RCA: 73] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2016] [Accepted: 08/05/2016] [Indexed: 02/08/2023] Open
Abstract
The liver is a unique organ with an extraordinary capacity to regenerate upon various injuries. In acute and transient liver injury by insults such as chemical hepatotoxins, the liver in rodents returns to the original architecture by proliferation and remodeling of the remaining cells within a week. In contrast, chronic liver inflammation due to various etiologies, e.g., virus infection and metabolic and immune disorders, results in liver fibrosis, often leading to cirrhosis and carcinogenesis. In both acute and chronic inflammation, a variety of immune and non-immune cells in the liver is involved in the processes resulting in either regeneration or fibrosis. In addition, chronic hepatitis often accompanies proliferation of atypical biliary cells, also known as liver progenitor cells or oval cells. Although the origin of liver progenitor cells and its contribution to hepatic repair is still under intense debate, recent studies have revealed a regulatory role for immune cells in progenitor proliferation and differentiation. In this review, we summarize recent studies on liver regeneration and fibrosis in the viewpoint of inflammation.
Collapse
Affiliation(s)
- Minoru Tanaka
- Department of Regenerative Medicine, Research Institute, National Center for Global Health and Medicine, Tokyo, Japan
| | - Atsushi Miyajima
- Institute of Molecular and Cellular Biosciences, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
112
|
Macrophage Depletion Attenuates Extracellular Matrix Deposition and Ductular Reaction in a Mouse Model of Chronic Cholangiopathies. PLoS One 2016; 11:e0162286. [PMID: 27618307 PMCID: PMC5019458 DOI: 10.1371/journal.pone.0162286] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Accepted: 08/19/2016] [Indexed: 02/08/2023] Open
Abstract
Chronic cholangiopathies, such as primary and secondary sclerosing cholangitis, are progressive disease entities, associated with periportal accumulation of inflammatory cells, encompassing monocytes and macrophages, peribiliary extracellular matrix (ECM) deposition and ductular reaction (DR). This study aimed to elucidate the relevance of macrophages in the progression of chronic cholangiopathies through macrophage depletion in a 3,5-diethoxycarbonyl-1,4-dihydrocollidine (DDC) mouse model. One group of mice received a single i.p. injection of Clodronate encapsulated liposomes (CLOLipo) at day 7 of a 14 day DDC treatment, while control animals were co-treated with PBSLipo instead. Mice were sacrificed after 7 or respectively 14 days of treatment for immunohistochemical assessment of macrophage recruitment (F4/80), ECM deposition (Sirius Red, Laminin) and DR (CK19). Macrophage depletion during a 14 day DDC treatment resulted in a significant inhibition of ECM deposition. Porto-lobular migration patterns of laminin-rich ECM and ductular structures were significantly attenuated and a progression of DR was effectively inhibited by macrophage depletion. CLOLipo co-treatment resulted in a confined DR to portal regions without amorphous cell clusters. This study suggests that therapeutic options selectively directed towards macrophages might represent a feasible treatment for chronic cholestatic liver diseases.
Collapse
|
113
|
Abstract
The mechanisms underlying hepatic inflammation and fibrogenesis in chronic hepatitis B (CHB) are complex and several cytokines are involved. Tumor necrosis factor-like weak inducer of apoptosis (TWEAK) is a member of the tumor necrosis factor superfamily which also acts as a cytokine. This study was conducted to evaluate serum soluble TWEAK (sTWEAK) levels in noncirrhotic CHB patients.Fifty-two treatment naive CHB patients and 30 healthy controls were included in the study and serum sTWEAK concentrations were measured using commercially available ELISA kits.Mean serum sTWEAK concentration was significantly lower in CHB group than healthy controls (189.6 ± 63.3 pg/mL in CHB group and 297.6 ± 61.5 pg/mL in control group, P < 0.001). According to the degree of necroinflammation in liver biopsies mean sTWEAK concentrations were found to be 168.14 ± 51.51, 206.96 ± 58.51, and 223.62 ± 78.88 pg/mL in patients with mild, moderate, and severe inflammation, respectively, and the difference between groups was statistically significant (P = 0.022). sTWEAK concentration was also found to be significantly higher in patients with advanced fibrosis in liver biopsy samples (169.59 ± 52.02 and 211.17 ± 68.22 pg/mL in patients with mild and advanced fibrosis, respectively, P = 0.016). Receiver operating characteristic (ROC) curves were obtained in CHB group to differentiate patients with advanced fibrosis from patients with mild fibrosis. Area under curve (AUC) was 0.676 (95% Cl; 0.526-0.825) for sTWEAK and for the specified cut-off value of 213.67 pg/mL sensitivity and specificity were 60% and 81.4%, respectively. ROC curve for sTWEAK to differentiate patients with severe inflammation revealed an AUC of 0.664 (95% Cl; 0.450-0.878). A cut-off value of 243.27 pg/mL yielded 54.5% sensitivity and 82.9% specificity.Serum sTWEAK concentration is decreased in treatment naive CHB patients. Further studies with simultaneous determination of circulating sTWEAK concentrations and TWEAK and factor-inducible 14 (Fn14) expressions in the liver biopsy samples would clarify the exact association of TWEAK/Fn14 pathway in the pathogenesis of CHB.
Collapse
Affiliation(s)
- Mehmet Asil
- Division of Gastroenterology, Department of Internal Medicine, Meram School of Medicine, Necmettin Erbakan University, Meram, Konya, Turkey
- Correspondence: Mehmet Asil, Necmettin Erbakan Üniversitesi, Meram Tıp Fakültesi, İç Hastalıkları Anabilim, Dalı, Gastroenteroloji Kliniği, 42090 Meram, Konya, Turkey (e-mail: )
| | | |
Collapse
|
114
|
Guldiken N, Kobazi Ensari G, Lahiri P, Couchy G, Preisinger C, Liedtke C, Zimmermann HW, Ziol M, Boor P, Zucman-Rossi J, Trautwein C, Strnad P. Keratin 23 is a stress-inducible marker of mouse and human ductular reaction in liver disease. J Hepatol 2016; 65:552-9. [PMID: 27151178 DOI: 10.1016/j.jhep.2016.04.024] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2015] [Revised: 04/06/2016] [Accepted: 04/23/2016] [Indexed: 01/09/2023]
Abstract
BACKGROUND & AIMS Keratins (K) constitute the epithelial intermediate filaments. Among them, K7/K19 are widely used markers of the regenerative liver response termed ductular reaction (DR) that consists of activated biliary epithelial cells (BECs) and hepatic progenitor cells (HPCs) and correlates with liver disease severity. In the present study we aimed to characterize K23 in the liver. METHODS We analyzed the expression and localization of K23 in the digestive system under basal conditions as well as in various human and mouse liver diseases/stress models. Cell culture studies were used to study factors regulating K23 expression. RESULTS In untreated mice, K23 was restricted to biliary epithelia. It was (together with K7/K19) markedly upregulated in three different DR/cholestatic injury models, i.e., multidrug resistance protein 2 (Mdr2) knockouts, animals treated with 3,5-diethoxycarbonyl-1,4-dihydrocollidine or subjected to bile duct ligation. K23 levels correlated with the DR marker Fn14 and immunofluorescence staining showed a distinct co-localization with K7/K19. In chronic human liver disease, K23 expression increased in patients with a more prominent inflammation/fibrosis. A dramatic upregulation (>200times) was observed in patients with acute liver failure (ALF) and end-stage primary biliary cholangitis (PBC). Patients with alcoholic liver cirrhosis displayed increased K23 serum levels. In primary hepatocytes as well as hepatobiliary cell lines, treatment with TNF-related weak inducer of apoptosis (TWEAK), and the type I acute phase inducer interleukin (IL)-1β but not the type II inducer IL-6 elevated K23 expression. CONCLUSIONS K23 represents a specific, stress-inducible DR marker, whose levels correlate with liver disease severity. K23 may represent a useful non-invasive DR marker. LAY SUMMARY Ductular reaction represents a basic response to liver injury and correlates with liver disease severity. Our study identifies K23 as a novel ductular reaction marker in mice and humans.
Collapse
Affiliation(s)
- Nurdan Guldiken
- Department of Internal Medicine III, RWTH University Hospital Aachen, Germany; Interdisciplinary Center for Clinical Research (IZKF), RWTH University Hospital Aachen, Germany
| | - Gokce Kobazi Ensari
- Department of Internal Medicine III, RWTH University Hospital Aachen, Germany
| | - Pooja Lahiri
- Institute of Pathology, Medical University of Graz, Graz, Austria
| | - Gabrielle Couchy
- Inserm, UMR-1162, Génomique fonctionnelle des Tumeurs solides, Equipe Labellisée Ligue Contre le Cancer, Paris F-75010, France; Université Paris Descartes, Labex Immuno-Oncology, Sorbonne Paris Cité, F-75010 Paris, France; Université Paris 13, Sorbonne Paris Cité, UFR SMBH, F-93000 Bobigny, France; Université Paris Diderot, IUH, F-75010 Paris, France
| | - Christian Preisinger
- Interdisciplinary Center for Clinical Research (IZKF), RWTH University Hospital Aachen, Germany
| | - Christian Liedtke
- Department of Internal Medicine III, RWTH University Hospital Aachen, Germany
| | | | - Marianne Ziol
- Université Paris 13, Sorbonne Paris Cité, UFR SMBH, F-93000 Bobigny, France; Pathology Department, GH Paris-Seine-Saint-Denis, APHP, Bondy, France; Centre de ressources biologiques BB-0033-00027 - Hôpital Jean Verdier, GH Paris-Seine-Saint-Denis, APHP, Bondy, France
| | - Peter Boor
- Department of Nephrology and Institute of Pathology, RWTH University Hospital Aachen, Germany
| | - Jessica Zucman-Rossi
- Inserm, UMR-1162, Génomique fonctionnelle des Tumeurs solides, Equipe Labellisée Ligue Contre le Cancer, Paris F-75010, France; Université Paris Descartes, Labex Immuno-Oncology, Sorbonne Paris Cité, F-75010 Paris, France; Université Paris 13, Sorbonne Paris Cité, UFR SMBH, F-93000 Bobigny, France; Université Paris Diderot, IUH, F-75010 Paris, France
| | - Christian Trautwein
- Department of Internal Medicine III, RWTH University Hospital Aachen, Germany
| | - Pavel Strnad
- Department of Internal Medicine III, RWTH University Hospital Aachen, Germany; Interdisciplinary Center for Clinical Research (IZKF), RWTH University Hospital Aachen, Germany.
| |
Collapse
|
115
|
Hepatic stellate cells: fibrogenic, regenerative or both? Heterogeneity and context are key. Hepatol Int 2016; 10:902-908. [PMID: 27578210 DOI: 10.1007/s12072-016-9758-x] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Accepted: 07/29/2016] [Indexed: 12/24/2022]
Abstract
Since their original identification, our understanding of the role of hepatic stellate cells in both health and disease continues to grow. Numerous studies have delineated the role of stellate cell activation in contributing to the pool of myofibroblasts responsible for liver fibrosis, and these have resulted in the development of a number of anti-fibrotic strategies targeting this cell. However, their potential role in liver regeneration, both initiation and termination, is also emerging and needs to be contemplated when considering targeted therapy. Perhaps what is most striking is the increasing recognition that this is not just one cell, but rather, a heterogenous population made up of a number of different subsets of cells, each with differentiated and specific functions. The tools are emerging for this dissection and are greatly needed to truly develop targeted therapies that will inhibit fibrosis while promoting liver regeneration and repair.
Collapse
|
116
|
Mekonnen GA, Ijzer J, Nederbragt H. Tenascin-C in Chronic Canine Hepatitis: Immunohistochemical Localization and Correlation with Necro-Inflammatory Activity, Fibrotic Stage, and Expression of Alpha-Smooth Muscle Actin, Cytokeratin 7, and CD3+ Cells. Vet Pathol 2016; 44:803-13. [DOI: 10.1354/vp.44-6-803] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
During fibrosis, the extracellular matrix (ECM) is continuously remodeled and increases in volume due to the production of various proteins. We studied the distribution of tenascin-C (TN-C) and the correlation of TN-C with the necro-inflammatory activity and expression of alpha–smooth muscle actin (α-SMA), cytokeratin 7 (CK7), and CD3+ T-lymphocytes in canine chronic hepatitis. This was analyzed using immunohistochemistry and semiquantitative scoring. We used 3 groups ( n = 19) of dogs: group 1 ( n = 5) with neonatal hepatitis/lobular dissecting hepatitis (NH/LDH), group 2 ( n = 8) with chronic hepatitis/cirrhosis (CH/CIRR), and group 3 ( n = 6) consisting of healthy animals. In normal livers, TN-C was localized in Disse's space and around bile ducts and blood vessels. In CH/CIRR livers, TN-C was localized at the periphery of the regenerating nodules and was conspicuous in the bridging fibrous bands. In NH/LDH, TN-C was diffusely distributed along the reticular fibers that dissected between single cells or groups of hepatocytes. α-SMA in the normal hepatic parenchyma showed an irregular distribution along the perisinusoidal linings. In other groups, α-SMA was increased in fibrotic septa and perisinusoidal linings. In normal livers, CK7 was positive in bile ducts. In other groups, CK7-expressing cells were conspicuous in the portal-parenchymal interface, the periphery of the regenerative nodules, and the degenerated parenchyma. The pattern of CD3+ lymphocytes was inversely proportional to that of TN-C. These results also showed that TN-C is strongly correlated with increased fibrotic stage, inflammatory activity, and expression of CK7 and α-SMA. TN-C, CK7, and CD3 expression did not differ between diagnostic groups.
Collapse
Affiliation(s)
- G. A. Mekonnen
- National Animal Health Research Center, Sebeta, Ethiopia
- Utrecht University, Faculty of Veterinary Medicine, Department of Pathobiology, Division of Pathology, Utrecht, the Netherlands
| | - J. Ijzer
- Utrecht University, Faculty of Veterinary Medicine, Department of Pathobiology, Division of Pathology, Utrecht, the Netherlands
| | - H. Nederbragt
- Utrecht University, Faculty of Veterinary Medicine, Department of Pathobiology, Division of Pathology, Utrecht, the Netherlands
| |
Collapse
|
117
|
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
|
118
|
Liu WH, Ren LN, Wang T, Navarro-Alvarez N, Tang LJ. The Involving Roles of Intrahepatic and Extrahepatic Stem/Progenitor Cells (SPCs) to Liver Regeneration. Int J Biol Sci 2016; 12:954-63. [PMID: 27489499 PMCID: PMC4971734 DOI: 10.7150/ijbs.15715] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Accepted: 05/09/2016] [Indexed: 12/17/2022] Open
Abstract
Liver regeneration is usually attributed to mature hepatocytes, which possess a remarkable potential to proliferate under mild to moderate injury. However, when the liver is severely damaged or hepatocyte proliferation is greatly inhibited, liver stem/progenitor cells (LSPCs) will contribute to the liver regeneration process. LSPCs in the developing liver have been extensively characterized, however, their contributing role to liver regeneration has not been completely understood. In addition to the restoration of the liver parenchymal tissue by hepatocytes or/and LSPCs, or in some cases bone marrow (BM) derived cells, such as hematopoietic stem cells (HSCs) and mesenchymal stem cells (MSCs), the wound healing after injury in terms of angiopoiesis by liver sinusoidal endothelial cells (LSECs) or/and sinusoidal endothelial progenitor cells (SEPCs) is another important aspect taking place during regeneration. To conclude, liver regeneration can be mainly divided into three distinct restoring levels according to the cause and severity of injury: hepatocyte dominant regeneration, LSPCs mediated regeneration, extrahepatic stem cells participative regeneration. In this review, we focus on the recent findings of liver regeneration, especially on those related to stem/progenitor cells (SPCs)-mediated regeneration and their potential clinical applications and challenges.
Collapse
Affiliation(s)
- Wei-Hui Liu
- 1. General Surgery Center, Chengdu Military General Hospital; Chengdu, Sichuan Province, 610083
| | - Li-Na Ren
- 1. General Surgery Center, Chengdu Military General Hospital; Chengdu, Sichuan Province, 610083
| | - Tao Wang
- 1. General Surgery Center, Chengdu Military General Hospital; Chengdu, Sichuan Province, 610083
| | - Nalu Navarro-Alvarez
- 2. Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Li-Jun Tang
- 1. General Surgery Center, Chengdu Military General Hospital; Chengdu, Sichuan Province, 610083
| |
Collapse
|
119
|
DJ-1 deficiency attenuates expansion of liver progenitor cells through modulating the inflammatory and fibrogenic niches. Cell Death Dis 2016; 7:e2257. [PMID: 27277679 PMCID: PMC5143389 DOI: 10.1038/cddis.2016.161] [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: 11/27/2015] [Revised: 05/08/2016] [Accepted: 05/10/2016] [Indexed: 12/27/2022]
Abstract
Our previous study suggested that DJ-1 has a critical role in initiating an inflammatory response, but its role in the liver progenitor cell (LPC) expansion, a process highly dependent on the inflammatory niche, remains elusive. The objective of this study is to determine the role of DJ-1 in LPC expansion. The correlation of DJ-1 expression with LPC markers was examined in the liver of patients with hepatitis B or hepatitis C virus (HBV and HCV, respectively) infection, primary biliary cirrhosis (PBC), primary sclerosing cholangitis (PSC), nonalcoholic fatty liver disease (NAFLD), cirrhosis or hepatocellular carcinoma (HCC), respectively. The role of DJ-1 in LPC expansion and the formation of LPC-associated fibrosis and inflammation was examined in a 3,5-diethoxycarbonyl-1,4-dihydrocollidine (DDC) diet-induced liver injury murine model. We also determined the ability of hepatic stellate cells (HSCs) in recruiting macrophages in DJ-1 knockout (KO) mice. The expression levels of DJ-1 were upregulated in the liver of HBV, HCV, PBC and PSC patients and DDC-fed mice. Additionally, DJ-1 expression was positively correlated with LPC proliferation in patients with liver injury and mice with DDC exposure. DJ-1 has no direct effect on LPC proliferation. Reduced activation of HSCs and collagen deposition were observed in DJ-1 KO mice. Furthermore, infiltrated CD11b+Gr-1low macrophages and pro-inflammatory factors (IL-6, TNF-α) were attenuated in DJ-1 KO mice. Mechanistically, we found that HSCs isolated from DJ-1 KO mice had decreased secretion of macrophage-mobilizing chemokines, such as CCL2 and CX3CL1, resulting in impaired macrophage infiltration. DJ-1 positively correlates with LPC expansion during liver injury. DJ-1 deficiency negatively regulates LPC proliferation by impairing the formation of LPC-associated fibrosis and inflammatory niches.
Collapse
|
120
|
Wilhelm A, Shepherd EL, Amatucci A, Munir M, Reynolds G, Humphreys E, Resheq Y, Adams DH, Hübscher S, Burkly LC, Weston CJ, Afford SC. Interaction of TWEAK with Fn14 leads to the progression of fibrotic liver disease by directly modulating hepatic stellate cell proliferation. J Pathol 2016; 239:109-21. [PMID: 26924336 PMCID: PMC4949530 DOI: 10.1002/path.4707] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Revised: 01/31/2016] [Accepted: 02/17/2016] [Indexed: 12/21/2022]
Abstract
Tumour necrosis factor‐like weak inducer of apoptosis (TWEAK) and its receptor fibroblast growth factor‐inducible 14 (Fn14) have been associated with liver regeneration in vivo. To further investigate the role of this pathway we examined their expression in human fibrotic liver disease and the effect of pathway deficiency in a murine model of liver fibrosis. The expression of Fn14 and TWEAK in normal and diseased human and mouse liver tissue and primary human hepatic stellate cells (HSCs) were investigated by qPCR, western blotting and immunohistochemistry. In addition, the levels of Fn14 in HSCs following pro‐fibrogenic and pro‐inflammatory stimuli were assessed and the effects of exogenous TWEAK on HSCs proliferation and activation were studied in vitro. Carbon tetrachloride (CCl4) was used to induce acute and chronic liver injury in TWEAK KO mice. Elevated expression of both Fn14 and TWEAK were detected in acute and chronic human liver injury, and co‐localized with markers of activated HSCs. Fn14 levels were low in quiescent HSCs but were significantly induced in activated HSCs, which could be further enhanced with the profibrogenic cytokine TGFβin vitro. Stimulation with recombinant TWEAK induced proliferation but not further HSCs activation. Fn14 gene expression was also significantly up‐regulated in CCl4 models of hepatic injury whereas TWEAK KO mice showed reduced levels of liver fibrosis following chronic CCl4 injury. In conclusion, TWEAK/Fn14 interaction leads to the progression of fibrotic liver disease via direct modulation of HSCs proliferation, making it a potential therapeutic target for liver fibrosis. © 2016 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Annika Wilhelm
- Centre for Liver Research and National Institute for Health Research (NIHR) Birmingham Liver Biomedical Research Unit, University of Birmingham, UK
| | - Emma L Shepherd
- Centre for Liver Research and National Institute for Health Research (NIHR) Birmingham Liver Biomedical Research Unit, University of Birmingham, UK
| | - Aldo Amatucci
- Department of Immunology, Biogen, Cambridge, MA, USA
| | - Mamoona Munir
- Centre for Liver Research and National Institute for Health Research (NIHR) Birmingham Liver Biomedical Research Unit, University of Birmingham, UK
| | - Gary Reynolds
- Centre for Liver Research and National Institute for Health Research (NIHR) Birmingham Liver Biomedical Research Unit, University of Birmingham, UK
| | - Elizabeth Humphreys
- Centre for Liver Research and National Institute for Health Research (NIHR) Birmingham Liver Biomedical Research Unit, University of Birmingham, UK
| | - Yazid Resheq
- Centre for Liver Research and National Institute for Health Research (NIHR) Birmingham Liver Biomedical Research Unit, University of Birmingham, UK.,Medizinische Klinik 5/Department of Internal Medicine 5, Universitätsklinikum Erlangen/University Medical Centre Erlangen, Germany
| | - David H Adams
- Centre for Liver Research and National Institute for Health Research (NIHR) Birmingham Liver Biomedical Research Unit, University of Birmingham, UK
| | - Stefan Hübscher
- Centre for Liver Research and National Institute for Health Research (NIHR) Birmingham Liver Biomedical Research Unit, University of Birmingham, UK.,Department of Cellular Pathology, University Hospital Birmingham NHS Foundation Trust, Birmingham, UK
| | | | - Christopher J Weston
- Centre for Liver Research and National Institute for Health Research (NIHR) Birmingham Liver Biomedical Research Unit, University of Birmingham, UK
| | - Simon C Afford
- Centre for Liver Research and National Institute for Health Research (NIHR) Birmingham Liver Biomedical Research Unit, University of Birmingham, UK
| |
Collapse
|
121
|
Reich M, Deutschmann K, Sommerfeld A, Klindt C, Kluge S, Kubitz R, Ullmer C, Knoefel WT, Herebian D, Mayatepek E, Häussinger D, Keitel V. TGR5 is essential for bile acid-dependent cholangiocyte proliferation in vivo and in vitro. Gut 2016; 65:487-501. [PMID: 26420419 DOI: 10.1136/gutjnl-2015-309458] [Citation(s) in RCA: 144] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2015] [Accepted: 09/01/2015] [Indexed: 12/13/2022]
Abstract
OBJECTIVE Cholestatic liver diseases in humans as well as bile acid (BA)-feeding and common bile duct ligation (CBDL) in rodents trigger hyperplasia of cholangiocytes within the portal fields. Furthermore, elevation of BA levels enhances proliferation and invasiveness of cholangiocarcinoma (CCA) cells in animal models, thus promoting tumour progression. TGR5 is a G-protein coupled BA receptor, which is highly expressed in cholangiocytes and postulated to mediate the proliferative effects of BA. DESIGN BA-dependent cholangiocyte proliferation was examined in TGR5-knockout and wild type mice following cholic acid (CA)-feeding and CBDL. TGR5-dependent proliferation and protection from apoptosis was studied in isolated cholangiocytes and CCA cell lines following stimulation with TGR5 ligands and kinase inhibitors. TGR5 expression was analysed in human CCA tissue. RESULTS Cholangiocyte proliferation was significantly reduced in TGR5-knockout mice in response to CA-feeding and CBDL. Taurolithocholic acid and TGR5-selective agonists induced cholangiocyte proliferation through elevation of reactive oxygen species and cSrc mediated epidermal growth factor receptor transactivation and subsequent Erk1/2 phosphorylation only in wild type but not in TGR5-knockout-derived cells. In human CCA tissue TGR5 was overexpressed and the pathway of TGR5-dependent proliferation via epidermal growth factor receptor and extracellular signal-regulated kinase (ERK)1/2 activation also translated to CCA cell lines. Furthermore, apoptosis was inhibited by TGR5-dependent CD95 receptor serine phosphorylation. CONCLUSIONS TGR5 is an important mediator of BA-induced cholangiocyte proliferation in vivo and in vitro. Furthermore, TGR5 protects cholangiocytes from death receptor-mediated apoptosis. These mechanisms may protect cholangiocytes from BA toxicity under cholestatic conditions, however, they may trigger proliferation and apoptosis resistance in malignantly transformed cholangiocytes, thus promoting CCA progression.
Collapse
Affiliation(s)
- Maria Reich
- Clinic for Gastroenterology, Hepatology and Infectious Diseases, Heinrich-Heine-University, Düsseldorf, Germany
| | - Kathleen Deutschmann
- Clinic for Gastroenterology, Hepatology and Infectious Diseases, Heinrich-Heine-University, Düsseldorf, Germany
| | - Annika Sommerfeld
- Clinic for Gastroenterology, Hepatology and Infectious Diseases, Heinrich-Heine-University, Düsseldorf, Germany
| | - Caroline Klindt
- Clinic for Gastroenterology, Hepatology and Infectious Diseases, Heinrich-Heine-University, Düsseldorf, Germany
| | - Stefanie Kluge
- Clinic for Gastroenterology, Hepatology and Infectious Diseases, Heinrich-Heine-University, Düsseldorf, Germany
| | - Ralf Kubitz
- Clinic for Gastroenterology, Hepatology and Infectious Diseases, Heinrich-Heine-University, Düsseldorf, Germany
| | - Christoph Ullmer
- Roche Pharmaceutical Research and Early Development, Roche Innovation Center Basel, Basel, Switzerland
| | - Wolfram T Knoefel
- Department of General, Visceral, and Pediatric Surgery, Heinrich-Heine-University, Düsseldorf, Germany
| | - Diran Herebian
- Department of General Pediatrics, Neonatalogy and Pediatric Cardiology, Heinrich-Heine-University, Düsseldorf, Germany
| | - Ertan Mayatepek
- Department of General Pediatrics, Neonatalogy and Pediatric Cardiology, Heinrich-Heine-University, Düsseldorf, Germany
| | - Dieter Häussinger
- Clinic for Gastroenterology, Hepatology and Infectious Diseases, Heinrich-Heine-University, Düsseldorf, Germany
| | - Verena Keitel
- Clinic for Gastroenterology, Hepatology and Infectious Diseases, Heinrich-Heine-University, Düsseldorf, Germany
| |
Collapse
|
122
|
Soluble TNF-Like Weak Inducer of Apoptosis as a New Marker in Preeclampsia: A Pilot Clinical Study. DISEASE MARKERS 2016; 2016:5930589. [PMID: 26989294 PMCID: PMC4773523 DOI: 10.1155/2016/5930589] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 10/31/2015] [Accepted: 01/17/2016] [Indexed: 01/13/2023]
Abstract
Introduction. All findings of preeclampsia appear as the clinical consequences of diffuse endothelial dysfunction. Soluble tumor necrosis factor-like weak inducer of apoptosis (sTWEAK) was recently introduced as a TNF related cytokine in various inflammatory and noninflammatory disorders. sTWEAK was found to be related to endothelial dysfunction in patients with chronic kidney disease. In our study we aimed to compare sTWEAK levels in women with preeclampsia to corresponding levels in a healthy pregnant control group. Materials and Methods. The study was undertaken with 33 patients with preeclampsia and 33 normal pregnant women. The concentration of sTWEAK in serum was calculated with an enzyme linked immunosorbent assay (ELISA) kit. Results. Serum creatinine, uric acid, LDH levels, and uPCR were significantly higher in the patient group compared to the control group. sTWEAK levels were significantly lower in preeclamptic patients (332 ± 144 pg/mL) than in control subjects (412 ± 166 pg/mL) (p = 0.04). Discussion. Our study demonstrates that sTWEAK is decreased in patients with preeclampsia compared to healthy pregnant women. There is a need for further studies to identify the role of sTWEAK in the pathogenesis of preeclampsia and to determine whether it can be regarded as a predictor of the development of preeclampsia.
Collapse
|
123
|
Magro-Checa C, Zirkzee EJ, Huizinga TW, Steup-Beekman GM. Management of Neuropsychiatric Systemic Lupus Erythematosus: Current Approaches and Future Perspectives. Drugs 2016. [PMID: 26809245 DOI: 10.1007/s40265-015-0534-3"] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Neuropsychiatric systemic lupus erythematosus (NPSLE) is a generic definition referring to a series of neurological and psychiatric symptoms directly related to systemic lupus erythematosus (SLE). NPSLE includes heterogeneous and rare neuropsychiatric (NP) manifestations involving both the central and peripheral nervous system. Due to the lack of a gold standard, the attribution of NP symptoms to SLE represents a clinical challenge that obligates the strict exclusion of any other potential cause. In the acute setting, management of these patients does not differ from other non-SLE subjects presenting with the same NP manifestation. Afterwards, an individualized therapeutic strategy, depending on the presenting manifestation and severity of symptoms, must be started. Clinical trials in NPSLE are scarce and most of the data are extracted from case series and case reports. High-dose glucocorticoids and intravenous cyclophosphamide remain the cornerstone for patients with severe symptoms that are thought to reflect inflammation or an underlying autoimmune process. Rituximab, intravenous immunoglobulins, or plasmapheresis may be used if response is not achieved. When patients present with mild to moderate NP manifestations, or when maintenance therapy is warranted, azathioprine and mycophenolate may be considered. When symptoms are thought to reflect a thrombotic underlying process, anticoagulation and antiplatelet agents are the mainstay of therapy, especially if antiphospholipid antibodies or antiphospholipid syndrome are present. Recent trials on SLE using new biologicals, based on newly understood SLE mechanisms, have shown promising results. Based on what we currently know about its pathogenesis, it is tempting to speculate how these new therapies may affect the management of NPSLE patients. This article provides a comprehensive and critical review of the literature on the epidemiology, pathophysiology, diagnosis, and management of NPSLE. We describe the most common pharmacological treatments used in NPSLE, based on both a literature search and our expert opinion. The extent to which new drugs in the advanced development of SLE, or the blockade of new targets, may impact future treatment of NPSLE will also be discussed.
Collapse
Affiliation(s)
- César Magro-Checa
- Department of Rheumatology, Leiden University Medical Center, PO Box 9600, 2300 RC, Leiden, The Netherlands
| | - Elisabeth J Zirkzee
- Department of Rheumatology, Leiden University Medical Center, PO Box 9600, 2300 RC, Leiden, The Netherlands.,Department of Rheumatology, Maasstad Hospital, Rotterdam, The Netherlands
| | - Tom W Huizinga
- Department of Rheumatology, Leiden University Medical Center, PO Box 9600, 2300 RC, Leiden, The Netherlands
| | - Gerda M Steup-Beekman
- Department of Rheumatology, Leiden University Medical Center, PO Box 9600, 2300 RC, Leiden, The Netherlands.
| |
Collapse
|
124
|
Mustafa S, Martin HL, Burkly L, Costa A, Martins ML, Schwaninger M, Teismann P. The role of TWEAK/Fn14 signaling in the MPTP-model of Parkinson's disease. Neuroscience 2016; 319:116-22. [PMID: 26808775 PMCID: PMC4771015 DOI: 10.1016/j.neuroscience.2016.01.034] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2015] [Revised: 12/23/2015] [Accepted: 01/13/2016] [Indexed: 02/06/2023]
Abstract
The tumor necrosis factor like weak inducer of apoptosis (TWEAK) and its receptor, fibroblast growth factor-inducible 14 (Fn14), mediate inflammation and neuronal apoptosis in cerebral edema, ischemic stroke and multiple sclerosis. The downstream effectors and pathways linked to TWEAK-Fn14 signaling are strongly implicated in the pathology of Parkinson's disease (PD), thus indicating a putative role for TWEAK/Fn14 signaling in PD neurodegeneration. Using the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) mouse model, we aimed to determine whether genetic ablation or pharmacologic mitigation of the TWEAK protein and its Fn14 receptor affected substantia nigra and striatum Parkinsonian pathology. Changes in endogenous TWEAK protein expression were also quantified in tissue from both MPTP-treated mice and PD human samples. TWEAK protein expression was transiently increased in the striatal tissue but remained unaltered in substantia nigra tissue of MPTP-treated mice. There was also no change of TWEAK protein levels in the substantia nigra or the striatum of human PD patients as compared to matched control subjects. Mitigating the effects of endogenous TWEAK protein using neutralizing antibody did affect MPTP-mediated neurotoxicity in the substantia nigra using the sub-acute model of MPTP (30mg/kg i.p. over five consecutive days). Neither TWEAK nor Fn14 genetic ablation led to attenuation of MPTP-toxicity in the acute model. These findings suggest that TWEAK signaling might be an aspect of MPTP-mediated neuropathology and be involved in the overall neurodegenerative pathology of PD.
Collapse
Affiliation(s)
- S Mustafa
- School of Medical Sciences, University of Aberdeen, Aberdeen, United Kingdom
| | - H L Martin
- School of Medical Sciences, University of Aberdeen, Aberdeen, United Kingdom
| | - L Burkly
- Department of Immunology, Biogen Idec, Inc., Cambridge, MA, United States
| | - A Costa
- Cell Death Regulation Laboratory, MRC Toxicology Unit, Leicester, United Kingdom
| | - M L Martins
- Cell Death Regulation Laboratory, MRC Toxicology Unit, Leicester, United Kingdom
| | - M Schwaninger
- Institute of Experimental and Clinical Pharmacology and Toxicology, University of Lübeck, Lübeck, Germany; Department of Pharmacology, University of Heidelberg, Germany
| | - P Teismann
- School of Medical Sciences, University of Aberdeen, Aberdeen, United Kingdom.
| |
Collapse
|
125
|
Golberg A, Bruinsma BG, Jaramillo M, Yarmush ML, Uygun BE. Rat liver regeneration following ablation with irreversible electroporation. PeerJ 2016; 4:e1571. [PMID: 26819842 PMCID: PMC4727979 DOI: 10.7717/peerj.1571] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2015] [Accepted: 12/14/2015] [Indexed: 12/18/2022] Open
Abstract
During the past decade, irreversible electroporation (IRE) ablation has emerged as a promising tool for the treatment of multiple diseases including hepatic cancer. However, the mechanisms behind the tissue regeneration following IRE ablation have not been investigated. Our results indicate that IRE treatment immediately kills the cells at the treatment site preserving the extracellular architecture, in effect causing in vivo decellularization. Over the course of 4 weeks, progenitor cell differentiation, through YAP and notch pathways, together with hepatocyte expansion led to almost complete regeneration of the ablated liver leading to the formation of hepatocyte like cells at the ablated zone. We did not observe significant scarring or tumor formation at the regenerated areas 6 months post IRE. Our study suggests a new model to study the regeneration of liver when the naïve extracellular matrix is decellularized in vivo with completely preserved extracellular architecture.
Collapse
Affiliation(s)
- Alexander Golberg
- Center for Engineering in Medicine, Massachusetts General Hospital, Harvard Medical School, and Shriners Hospitals for Children in Boston, Boston, MA, United States; Porter School of Environmental Studies, Tel Aviv University, Tel Aviv, Israel
| | - Bote G Bruinsma
- Center for Engineering in Medicine, Massachusetts General Hospital, Harvard Medical School, and Shriners Hospitals for Children in Boston, Boston, MA, United States; Department of Surgery (Surgical Laboratory), Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Maria Jaramillo
- Center for Engineering in Medicine, Massachusetts General Hospital, Harvard Medical School, and Shriners Hospitals for Children in Boston , Boston, MA , United States
| | - Martin L Yarmush
- Center for Engineering in Medicine, Massachusetts General Hospital, Harvard Medical School, and Shriners Hospitals for Children in Boston, Boston, MA, United States; Department of Biomedical Engineering, Rutgers University, Piscataway, New Jersey, United States
| | - Basak E Uygun
- Center for Engineering in Medicine, Massachusetts General Hospital, Harvard Medical School, and Shriners Hospitals for Children in Boston , Boston, MA , United States
| |
Collapse
|
126
|
Yu YR, Ni XQ, Huang J, Zhu YH, Qi YF. Taurine drinking ameliorates hepatic granuloma and fibrosis in mice infected with Schistosoma japonicum. INTERNATIONAL JOURNAL FOR PARASITOLOGY-DRUGS AND DRUG RESISTANCE 2016; 6:35-43. [PMID: 27054062 PMCID: PMC4805782 DOI: 10.1016/j.ijpddr.2016.01.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/17/2015] [Revised: 01/12/2016] [Accepted: 01/12/2016] [Indexed: 02/08/2023]
Abstract
In schistosomiasis, egg-induced hepatic granuloma formation is a cytokine-mediated, predominantly CD4+ Th2 immune response that can give rise to hepatic fibrosis. Hepatic fibrosis is the main cause of increased morbidity and mortality in humans with schistosome infection. Taurine has various physiological functions and hepatoprotective properties as well as anti-inflammatory and immunomodulatory activity. However, little is known about the role of taurine in schistosome egg-induced granuloma formation and fibrosis. We aimed to evaluate the therapeutic potential of taurine as preventative treatment for Schistosoma japonicum infection. Mice infected with S. japonicum cercariae were supplied with taurine drinking water (1% w/v) for 4 weeks starting at 4 weeks post-infection. Taurine supplementation significantly improved the liver pathologic findings, reduced the serum levels of aminotransferases and area of hepatic granuloma, and prevented fibrosis progression. In addition, taurine decreased the expression of the granulomatous and fibrogenic mediators transforming growth factor β1, tumor necrosis factor α, monocyte chemotactic protein 1α and macrophage inflammatory protein 1α as well as the endoplasmic reticulum stress marker glucose-regulated protein 78. Thus, taurine can significantly attenuate S. japonicum egg-induced hepatic granuloma and fibrosis, which may depend in part on the downregulation of some relevant cytokine/chemokines and reducing the endoplasmic reticulum stress response. Taurine has potential as preventative & therapeutic treatment for schistosomiasis. Taurine reduced the development of liver pathology caused by S. japonicum infection. Taurine attenuated S. japonicum egg-induced hepatic granuloma and fibrosis.
Collapse
Affiliation(s)
- Yan-Rong Yu
- Department of Pathogen Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China.
| | - Xian-Qiang Ni
- Department of Pathogen Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Jie Huang
- Department of Pathogen Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Yong-Hong Zhu
- Department of Pathogen Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Yong-Fen Qi
- Department of Pathogen Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China; Laboratory of Cardiovascular Bioactive Molecule, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China; Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing 100191, China.
| |
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
|
Peng ZW, Ikenaga N, Liu SB, Sverdlov DY, Vaid KA, Dixit R, Weinreb PH, Violette S, Sheppard D, Schuppan D, Popov Y. Integrin αvβ6 critically regulates hepatic progenitor cell function and promotes ductular reaction, fibrosis, and tumorigenesis. Hepatology 2016; 63:217-32. [PMID: 26448099 PMCID: PMC5312042 DOI: 10.1002/hep.28274] [Citation(s) in RCA: 96] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2015] [Revised: 09/22/2015] [Accepted: 10/05/2015] [Indexed: 12/12/2022]
Abstract
UNLABELLED Integrin αvβ6 is rapidly up-regulated on cells of epithelial lineage during tissue injury, where one of its primary functions is activation of latent transforming growth factor beta 1 (TGFβ1). In human liver cirrhosis, αvβ6 is overexpressed by cells comprising the ductular reaction, and its inhibition suppresses experimental biliary fibrosis in rodents. Here, we show that αvβ6 is expressed on the actively proliferating subset of hepatic progenitor cells and is required for their progenitor function in vivo and in vitro through integrin αvβ6-dependent TGFβ1 activation. Freshly isolated αvβ6(+) liver cells demonstrate clonogenic potential and differentiate into cholangiocytes and functional hepatocytes in vitro, whereas colony formation by epithelial cell adhesion molecule-positive progenitor cells is blocked by αvβ6-neutralizing antibody and in integrin beta 6-deficient cells. Inhibition of progenitors by anti-αvβ6 antibody is recapitulated by TGFβ1 neutralization and rescued by addition of bioactive TGFβ1. Genetic disruption or selective targeting of αvβ6 with 3G9 antibody potently inhibits progenitor cell responses in mouse models of chronic biliary injury and protects from liver fibrosis and tumorigenesis, two conditions clinically associated with exacerbated ductular reaction. CONCLUSION These results suggest that αvβ6 is a promising target for chronic fibrotic liver diseases and associated cancers.
Collapse
Affiliation(s)
- Zhen-Wei Peng
- Department of Oncology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China,Division of Gastroenterology and Hepatology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA
| | - Naoki Ikenaga
- Division of Gastroenterology and Hepatology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA
| | - Susan B. Liu
- Division of Gastroenterology and Hepatology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA
| | - Deanna Y. Sverdlov
- Division of Gastroenterology and Hepatology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA
| | - Kahini A. Vaid
- Division of Gastroenterology and Hepatology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA
| | - Richa Dixit
- Division of Gastroenterology and Hepatology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA
| | | | | | - Dean Sheppard
- Lung Biology Center, University of California, San Francisco School of Medicine, San Francisco, CA
| | - Detlef Schuppan
- Institute of Translational Immunology and Research Center for Immune Therapy, University Medical Center, Mainz, Germany
| | - Yury Popov
- Division of Gastroenterology and Hepatology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA
| |
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
|
Fukuda T, Fukuchi T, Yagi S, Shiojiri N. Immunohistochemical analyses of cell cycle progression and gene expression of biliary epithelial cells during liver regeneration after partial hepatectomy of the mouse. Exp Anim 2015; 65:135-46. [PMID: 26633692 PMCID: PMC4873482 DOI: 10.1538/expanim.15-0082] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The liver has a remarkable regeneration capacity, and, after surgical removal of its
mass, the remaining tissue undergoes rapid regeneration through compensatory growth of its
constituent cells. Although hepatocytes synchronously proliferate under the control of
various signaling molecules from neighboring cells, there have been few detailed analyses
on how biliary cells regenerate for their cell population after liver resection. The
present study was undertaken to clarify how biliary cells regenerate after partial
hepatectomy of mice through extensive analyses of their cell cycle progression and gene
expression using immunohistochemical and RT-PCR techniques. When expression of PCNA, Ki67
antigen, topoisomerase IIα and phosphorylated histone H3, which are cell cycle markers,
was immunohistochemically examined during liver regeneration, hepatocytes had a peak of
the S phase and M phase at 48–72 h after resection. By contrast, biliary epithelial cells
had much lower proliferative activity than that of hepatocytes, and their peak of the S
phase was delayed. Mitotic figures were rarely detectable in biliary cells. RT-PCR
analyses of gene expression of biliary markers such as Spp1
(osteopontin), Epcam and Hnf1b demonstrated that they
were upregulated during liver regeneration. Periportal hepatocytes expressed some of
biliary markers, including Spp1 mRNA and protein. Some periportal
hepatocytes had downregulated expression of HNF4α and HNF1α. Gene expression of Notch
signaling molecules responsible for cell fate decision of hepatoblasts to biliary cells
during development was upregulated during liver regeneration. Notch signaling may be
involved in biliary regeneration.
Collapse
Affiliation(s)
- Tatsuya Fukuda
- Department of Biology, Faculty of Science, Shizuoka University, Oya 836, Suruga-ku, Shizuoka city, Shizuoka 422-8529, Japan
| | | | | | | |
Collapse
|
132
|
Dubuquoy L, Louvet A, Lassailly G, Truant S, Boleslawski E, Artru F, Maggiotto F, Gantier E, Buob D, Leteurtre E, Cannesson A, Dharancy S, Moreno C, Pruvot FR, Bataller R, Mathurin P. Progenitor cell expansion and impaired hepatocyte regeneration in explanted livers from alcoholic hepatitis. Gut 2015; 64:1949-60. [PMID: 25731872 PMCID: PMC4558407 DOI: 10.1136/gutjnl-2014-308410] [Citation(s) in RCA: 117] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2014] [Accepted: 01/30/2015] [Indexed: 12/20/2022]
Abstract
OBJECTIVE In alcoholic hepatitis (AH), development of targeted therapies is crucial and requires improved knowledge of cellular and molecular drivers in liver dysfunction. The unique opportunity of using explanted livers from patients with AH having undergone salvage liver transplantation allowed to perform more in-depth molecular translational studies. DESIGN We studied liver explants from patients with AH submitted to salvage transplantation (n=16), from patients with alcoholic cirrhosis without AH (n=12) and fragments of normal livers (n=16). Hepatic cytokine content was quantified. Hepatocyte function and proliferation and the presence of hepatic progenitor cells (HPCs) were evaluated by immunohistochemistry, western blot or quantitative PCR. Mitochondrial morphology was evaluated by electron microscopy. RESULTS Livers from patients with AH showed decreased cytokine levels involved in liver regeneration (tumour necrosis factor α and interleukin-6), as well as a virtual absence of markers of hepatocyte proliferation compared with alcoholic cirrhosis and normal livers. Electron microscopy revealed obvious mitochondrial abnormalities in AH hepatocytes. Importantly, livers from patients with AH showed substantial accumulation of HPCs that, unexpectedly, differentiate only into biliary cells. AH livers predominantly express laminin (extracellular matrix protein favouring cholangiocyte differentiation); consequently, HPC expansion is inefficient at yielding mature hepatocytes. CONCLUSIONS AH not responding to medical therapy is associated with lack of expression of cytokines involved in liver regeneration and profound mitochondrial damage along with lack of proliferative hepatocytes. Expansion of HPCs is inefficient to yield mature hepatocytes. Manoeuvres aimed at promoting differentiation of HPCs into mature hepatocytes should be tested in AH.
Collapse
Affiliation(s)
- Laurent Dubuquoy
- Inserm, UMR995 – LIRIC, Lille, France,Univ Lille, UMR995 – LIRIC, Lille, France,CHRU Lille, Service des Maladies de l’Appareil Digestif et de la Nutrition, Hôpital Huriez, Lille, France
| | - Alexandre Louvet
- Inserm, UMR995 – LIRIC, Lille, France,Univ Lille, UMR995 – LIRIC, Lille, France,CHRU Lille, Service des Maladies de l’Appareil Digestif et de la Nutrition, Hôpital Huriez, Lille, France
| | - Guillaume Lassailly
- Inserm, UMR995 – LIRIC, Lille, France,Univ Lille, UMR995 – LIRIC, Lille, France,CHRU Lille, Service des Maladies de l’Appareil Digestif et de la Nutrition, Hôpital Huriez, Lille, France
| | - Stéphanie Truant
- CHRU Lille, Service de Chirurgie Digestive et Transplantation, Hôpital Huriez, Lille, France
| | - Emmanuel Boleslawski
- CHRU Lille, Service de Chirurgie Digestive et Transplantation, Hôpital Huriez, Lille, France
| | - Florent Artru
- Inserm, UMR995 – LIRIC, Lille, France,Univ Lille, UMR995 – LIRIC, Lille, France,CHRU Lille, Service des Maladies de l’Appareil Digestif et de la Nutrition, Hôpital Huriez, Lille, France
| | - François Maggiotto
- Inserm, UMR995 – LIRIC, Lille, France,Univ Lille, UMR995 – LIRIC, Lille, France,CHRU Lille, Service des Maladies de l’Appareil Digestif et de la Nutrition, Hôpital Huriez, Lille, France
| | - Emilie Gantier
- Inserm, UMR995 – LIRIC, Lille, France,Univ Lille, UMR995 – LIRIC, Lille, France,CHRU Lille, Service des Maladies de l’Appareil Digestif et de la Nutrition, Hôpital Huriez, Lille, France
| | - David Buob
- CHRU Lille, Service d’Anatomo-Pathologie, Lille, France
| | | | - Amélie Cannesson
- Inserm, UMR995 – LIRIC, Lille, France,Univ Lille, UMR995 – LIRIC, Lille, France,CHRU Lille, Service des Maladies de l’Appareil Digestif et de la Nutrition, Hôpital Huriez, Lille, France
| | - Sébastien Dharancy
- Inserm, UMR995 – LIRIC, Lille, France,Univ Lille, UMR995 – LIRIC, Lille, France,CHRU Lille, Service des Maladies de l’Appareil Digestif et de la Nutrition, Hôpital Huriez, Lille, France
| | - Christophe Moreno
- Department of Gastroenterology and Hepato-pancreatology, Erasme Hospital, Université Libre de Bruxelles, Brussels, Belgium,Laboratory of Experimental Gastroenterology, Université Libre de Bruxelles, Brussels, Belgium
| | - François-René Pruvot
- CHRU Lille, Service de Chirurgie Digestive et Transplantation, Hôpital Huriez, Lille, France
| | - Ramon Bataller
- Department of Medicine, Division of Gastroenterology and Hepatology, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Philippe Mathurin
- Inserm, UMR995 – LIRIC, Lille, France,Univ Lille, UMR995 – LIRIC, Lille, France,CHRU Lille, Service des Maladies de l’Appareil Digestif et de la Nutrition, Hôpital Huriez, Lille, France
| |
Collapse
|
133
|
Marquardt JU, Andersen JB, Thorgeirsson SS. Functional and genetic deconstruction of the cellular origin in liver cancer. Nat Rev Cancer 2015; 15:653-67. [PMID: 26493646 DOI: 10.1038/nrc4017] [Citation(s) in RCA: 224] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
During the past decade, research on primary liver cancers has particularly highlighted the uncommon plasticity of differentiated parenchymal liver cells (that is, hepatocytes and cholangiocytes (also known as biliary epithelial cells)), the role of liver progenitor cells in malignant transformation, the importance of the tumour microenvironment and the molecular complexity of liver tumours. Whereas other reviews have focused on the landscape of genetic alterations that promote development and progression of primary liver cancers and the role of the tumour microenvironment, the crucial importance of the cellular origin of liver cancer has been much less explored. Therefore, in this Review, we emphasize the importance and complexity of the cellular origin in tumour initiation and progression, and attempt to integrate this aspect with recent discoveries in tumour genomics and the contribution of the disrupted hepatic microenvironment to liver carcinogenesis.
Collapse
Affiliation(s)
- Jens U Marquardt
- Department of Medicine I, Johannes Gutenberg University, Langenbeckstrasse 1, 55131 Mainz, Germany
| | - Jesper B Andersen
- Biotech Research and Innovation Centre (BRIC), University of Copenhagen, Ole Maaløes Vej 5, 2200 Copenhagen N, Denmark
| | - Snorri S Thorgeirsson
- Laboratory of Experimental Carcinogenesis, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892, USA
| |
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
|
Cheng H, Xu M, Liu X, Zou X, Zhan N, Xia Y. TWEAK/Fn14 activation induces keratinocyte proliferation under psoriatic inflammation. Exp Dermatol 2015; 25:32-7. [PMID: 26264384 DOI: 10.1111/exd.12820] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/05/2015] [Indexed: 12/17/2022]
Affiliation(s)
- Hong Cheng
- Department of Medicine; The Second Affiliated Hospital; School of Medicine; Xi'an Jiaotong University; Xi'an China
- Department of Dermatology; The Second Affiliated Hospital; School of Medicine; Xi'an Jiaotong University; Xi'an China
| | - Meifeng Xu
- Department of Dermatology; The Second Affiliated Hospital; School of Medicine; Xi'an Jiaotong University; Xi'an China
| | - Xiaoming Liu
- Department of Dermatology; The Third Affiliated Hospital of Soochow University; Changzhou China
| | - Xiaoyan Zou
- Department of Dermatology; Hubei Maternity and Child Health Hospital; Wuhan China
| | - Na Zhan
- Department of Pathology; Renmin Hospital of Wuhan University; Wuhan China
| | - Yumin Xia
- Department of Dermatology; The Second Affiliated Hospital; School of Medicine; Xi'an Jiaotong University; Xi'an China
| |
Collapse
|
136
|
Abstract
In recent years, hepatic oval cells (HOC) have gradually become a research hotspot, and their participation in the reconstruction of liver structure and function has been preliminarily confirmed. This provides a new direction for the study of the pathogenesis and treatment of liver injury, hepatitis, liver fibrosis, cirrhosis, liver neoplasms and other liver diseases. This paper will discuss the relationship between hepatic oval cells and liver diseases.
Collapse
|
137
|
Zhang Z, Li Z, Zou C, Zhang J, Zhu Y, Miao Y. Angiogenesis and proliferation of bile duct enhances ischemic tolerance in rats with cirrhosis. Int J Clin Exp Med 2015; 8:12086-12095. [PMID: 26550120 PMCID: PMC4612805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2015] [Accepted: 08/02/2015] [Indexed: 06/05/2023]
Abstract
BACKGROUND/AIMS Primary biliary cirrhosis (PBC), an autoimmune disease of the liver, is marked by slow progressive destruction of bile ducts. These patients with PBC often undergo orthotopic liver transplantation (OLT). Ischemic bile duct lesion (IBDL) is a major source of morbidity and even mortality after OLT. Cirrhosis of the liver has a higher tolerance to ischemia than a normal liver, but the mechanism remains unknown. Angiogenesis and proliferation of bile duct often responses in bile duct ischemia, which may enhance ischemic tolerance in patients with cirrhosis. METHODOLOGY To test the hypothesis, a rat model with cirrhosis was established. Biochemical indexes of ischemic severity were measured including total bilirubin (TBIL) and direct bilirubin (DBIL). Immunohistochemical assay was performed for Ki67 (a biomarker for the proliferation of bile duct) and CD34 (a biomarker of angiogenesis). RESULTS The levels were lower for TBIL and DBIL in the bile duct from rat model with cirrhosis than that from a normal rat after ischemic surgery (P < 0.05). The levels were higher for Ki67 and CD34 from a rat model with cirrhosis than that from a normal rat after ischemic surgery (P < 0.05). CONCLUSIONS The results suggest that a liver with cirrhosis has a better ischemic tolerance than a normal liver. Angiogenesis and proliferation of bile duct enhances ischemic tolerance in rats with cirrhosis. More research on the pathogenesis of IBDLs is needed for developing more specific preventive or therapeutic strategies.
Collapse
Affiliation(s)
- Zhiqiang Zhang
- Department of General Surgery, First Affiliated Hospital of Nanjing Medical University Nanjing 210029, China
| | - Zhennan Li
- Department of General Surgery, First Affiliated Hospital of Nanjing Medical University Nanjing 210029, China
| | - Chen Zou
- Department of General Surgery, First Affiliated Hospital of Nanjing Medical University Nanjing 210029, China
| | - Jingjing Zhang
- Department of General Surgery, First Affiliated Hospital of Nanjing Medical University Nanjing 210029, China
| | - Yi Zhu
- Department of General Surgery, First Affiliated Hospital of Nanjing Medical University Nanjing 210029, China
| | - Yi Miao
- Department of General Surgery, First Affiliated Hospital of Nanjing Medical University Nanjing 210029, 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
|
Carotti S, Vespasiani-Gentilucci U, Perrone G, Picardi A, Morini S. Portal inflammation during NAFLD is frequent and associated with the early phases of putative hepatic progenitor cell activation. J Clin Pathol 2015; 68:883-90. [PMID: 26124313 DOI: 10.1136/jclinpath-2014-202717] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2014] [Accepted: 06/11/2015] [Indexed: 12/19/2022]
Abstract
AIMS We investigated whether portal tract inflammation observed in non-alcoholic fatty liver disease (NAFLD) is associated with hepatic progenitor cell compartment activation, as thoroughly evaluated with different markers of the staminal lineage. METHODS Fifty-two patients with NAFLD were studied. NAFLD activity score, fibrosis and portal inflammation were histologically evaluated. Putative hepatic progenitor cells, intermediate hepatobiliary cells and bile ductules/interlobular bile ducts were evaluated by immunohistochemistry for cytokeratin (CK)-7, CK-19 and epithelial cell adhesion molecule (EpCAM), and a hepatic progenitor cell compartment score was derived. Hepatic stellate cell and myofibroblast activity was determined by immunohistochemistry for α-smooth muscle actin. RESULTS Portal inflammation was absent in a minority of patients, mild in 40% of cases and more than mild in about half of patients, showing a strong correlation with fibrosis (r=0.76, p<0.001). Portal inflammation correlated with CK-7-counted putative hepatic progenitor cells (r=0.48, p<0.001), intermediate hepatobiliary cells (r=0.6, p<0.001) and bile ductules/interlobular bile ducts (r=0.6, p<0.001), and with the activity of myofibroblasts (r=0.5, p<0.001). Correlations were confirmed when elements were counted by immunostaining for CK-19 and EpCAM. Lobular inflammation, ballooning, myofibroblast activity and hepatic progenitor cell compartment activation were associated with portal inflammation by univariate analysis. In the multivariate model, the only variable independently associated with portal inflammation was hepatic progenitor cell compartment activation (OR 3.7, 95% CI 1.1 to 12.6). CONCLUSIONS Portal inflammation is frequent during NAFLD and strongly associated with activation of putative hepatic progenitor cells since the first steps of their differentiation, portal myofibroblast activity and fibrosis.
Collapse
Affiliation(s)
- Simone Carotti
- Laboratory of Microscopic and Ultrastructural Anatomy, CIR, University Campus Bio-Medico of Rome, Rome, Italy
| | | | - Giuseppe Perrone
- Department of Anatomical Pathology, University Campus Bio-Medico of Rome, Rome, Italy
| | - Antonio Picardi
- Clinical Medicine and Hepatology Unit, University Campus Bio-Medico of Rome, Rome, Italy
| | - Sergio Morini
- Laboratory of Microscopic and Ultrastructural Anatomy, CIR, University Campus Bio-Medico of Rome, Rome, Italy
| |
Collapse
|
140
|
Liu Y, Cao L, Chen R, Zhou X, Fan X, Liang Y, Jia R, Wang H, Liu G, Guo Y, Zhao J. Osteopontin Promotes Hepatic Progenitor Cell Expansion and Tumorigenicity via Activation of β-Catenin in Mice. Stem Cells 2015; 33:3569-80. [PMID: 26033745 DOI: 10.1002/stem.2072] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Revised: 04/12/2015] [Accepted: 04/27/2015] [Indexed: 12/20/2022]
Abstract
Upregulation of osteopontin (OPN) has been found in hepatic progenitor cells (HPCs) in several liver diseases with portal biliary proliferation. Here, we investigated the role of HPC-derived autocrine OPN in regulating HPC expansion, migration, and hepatocarcinogenesis in mice. Five-week-old, weighing between 18 and 20 g of either wild type (WT) or OPN gene knockout (OPN-KO) male mice were treated with modified choline-deficient, ethionine-supplemented diet (modified choline-deficient [MCDE]) for 2 weeks to induce HPC production, or 6-12 months to induce tumorigenesis. Epithelial cell adhesion molecule EpCAM(+) CD45(-) cells isolated from mouse liver and liver epithelial progenitor cells were used for in vitro study. OPN was blocked by specific antibody or RNAi-mediated silence to investigate the role of OPN. To evaluate correlation between OPN expression and β-catenin activity, expressions of OPN and β-catenin were assessed in human liver cancer specimens. We found autocrine OPN promotes HPC expansion and migration by decreasing membranous E-cadherin and increasing free cytoplasmic β-catenin via binding to αv integrin and activating Src activity. Depletion of OPN significantly attenuated MCDE-induced hepatocarcinogenesis. Clinical evidence revealed a strong correlation of high OPN expression with cytoplasmic/nuclear expression of β-catenin in 43 cases of human combined hepatocellular carcinoma and cholangiocarcinoma and mixed intrahepatic cholangiocarcinoma and 80 cases of hepatocellular carcinoma. Our results indicate that autocrine OPN plays a crucial role in HPC expansion, migration, and subsequent oncogenic transformation of HPCs, which may provide a new insight into hepatocarcinogenesis.
Collapse
Affiliation(s)
- Yingying Liu
- International Joint Cancer Institute, The Second Military Medical University, Shanghai, People's Republic of China.,Changhai Hospital, The Second Military Medical University, Shanghai, People's Republic of China
| | - Lei Cao
- International Joint Cancer Institute, The Second Military Medical University, Shanghai, People's Republic of China.,Clinical Immunology Laboratory, The First Affiliated Hospital of Suzhou University, Suzhou, Jiangsu, People's Republic of China
| | - Rui Chen
- International Joint Cancer Institute, The Second Military Medical University, Shanghai, People's Republic of China
| | - Xuyu Zhou
- Changhai Hospital, The Second Military Medical University, Shanghai, People's Republic of China
| | - Xiaoyu Fan
- International Joint Cancer Institute, The Second Military Medical University, Shanghai, People's Republic of China
| | - Yingchao Liang
- International Joint Cancer Institute, The Second Military Medical University, Shanghai, People's Republic of China
| | - Rongjie Jia
- International Joint Cancer Institute, The Second Military Medical University, Shanghai, People's Republic of China
| | - Hao Wang
- International Joint Cancer Institute, The Second Military Medical University, Shanghai, People's Republic of China
| | - Guoke Liu
- International Joint Cancer Institute, The Second Military Medical University, Shanghai, People's Republic of China
| | - Yajun Guo
- International Joint Cancer Institute, The Second Military Medical University, Shanghai, People's Republic of China.,PLA General Hospital Cancer Center, PLA postgraduate School of Medicine, Beijing, People's Republic of China.,Beijing Key Laboratory of Cell Engineering and Antibody, Beijing, People's Republic of China
| | - Jian Zhao
- International Joint Cancer Institute, The Second Military Medical University, Shanghai, People's Republic of China.,PLA General Hospital Cancer Center, PLA postgraduate School of Medicine, Beijing, People's Republic of China.,Beijing Key Laboratory of Cell Engineering and Antibody, Beijing, People's Republic of China
| |
Collapse
|
141
|
Weng HL, Cai X, Yuan X, Liebe R, Dooley S, Li H, Wang TL. Two sides of one coin: massive hepatic necrosis and progenitor cell-mediated regeneration in acute liver failure. Front Physiol 2015; 6:178. [PMID: 26136687 PMCID: PMC4468385 DOI: 10.3389/fphys.2015.00178] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2015] [Accepted: 05/26/2015] [Indexed: 02/06/2023] Open
Abstract
Massive hepatic necrosis is a key event underlying acute liver failure, a serious clinical syndrome with high mortality. Massive hepatic necrosis in acute liver failure has unique pathophysiological characteristics including extremely rapid parenchymal cell death and removal. On the other hand, massive necrosis rapidly induces the activation of liver progenitor cells, the so-called "second pathway of liver regeneration." The final clinical outcome of acute liver failure depends on whether liver progenitor cell-mediated regeneration can efficiently restore parenchymal mass and function within a short time. This review summarizes the current knowledge regarding massive hepatic necrosis and liver progenitor cell-mediated regeneration in patients with acute liver failure, the two sides of one coin.
Collapse
Affiliation(s)
- Hong-Lei Weng
- Department of Medicine II, Section Molecular Hepatology, Medical Faculty Mannheim, Heidelberg UniversityMannheim, Germany
| | - Xiaobo Cai
- Department of Medicine II, Section Molecular Hepatology, Medical Faculty Mannheim, Heidelberg UniversityMannheim, Germany
| | - Xiaodong Yuan
- Department of Medicine II, Section Molecular Hepatology, Medical Faculty Mannheim, Heidelberg UniversityMannheim, Germany
| | - Roman Liebe
- Department of Medicine II, Section Molecular Hepatology, Medical Faculty Mannheim, Heidelberg UniversityMannheim, Germany
- Department of Medicine II, Saarland University HospitalHomburg, Germany
| | - Steven Dooley
- Department of Medicine II, Section Molecular Hepatology, Medical Faculty Mannheim, Heidelberg UniversityMannheim, Germany
| | - Hai Li
- Department of Gastroenterology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong UniversityShanghai, China
| | - Tai-Ling Wang
- Department of Pathology, Beijing China-Japan Friendship HospitalBeijing, China
| |
Collapse
|
142
|
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
|
143
|
Karsdal MA, Manon-Jensen T, Genovese F, Kristensen JH, Nielsen MJ, Sand JMB, Hansen NUB, Bay-Jensen AC, Bager CL, Krag A, Blanchard A, Krarup H, Leeming DJ, Schuppan D. Novel insights into the function and dynamics of extracellular matrix in liver fibrosis. Am J Physiol Gastrointest Liver Physiol 2015; 308:G807-30. [PMID: 25767261 PMCID: PMC4437019 DOI: 10.1152/ajpgi.00447.2014] [Citation(s) in RCA: 179] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Accepted: 03/04/2015] [Indexed: 02/06/2023]
Abstract
Emerging evidence suggests that altered components and posttranslational modifications of proteins in the extracellular matrix (ECM) may both initiate and drive disease progression. The ECM is a complex grid consisting of multiple proteins, most of which play a vital role in containing the essential information needed for maintenance of a sophisticated structure anchoring the cells and sustaining normal function of tissues. Therefore, the matrix itself may be considered as a paracrine/endocrine entity, with more complex functions than previously appreciated. The aims of this review are to 1) explore key structural and functional components of the ECM as exemplified by monogenetic disorders leading to severe pathologies, 2) discuss selected pathological posttranslational modifications of ECM proteins resulting in altered functional (signaling) properties from the original structural proteins, and 3) discuss how these findings support the novel concept that an increasing number of components of the ECM harbor signaling functions that can modulate fibrotic liver disease. The ECM entails functions in addition to anchoring cells and modulating their migratory behavior. Key ECM components and their posttranslational modifications often harbor multiple domains with different signaling potential, in particular when modified during inflammation or wound healing. This signaling by the ECM should be considered a paracrine/endocrine function, as it affects cell phenotype, function, fate, and finally tissue homeostasis. These properties should be exploited to establish novel biochemical markers and antifibrotic treatment strategies for liver fibrosis as well as other fibrotic diseases.
Collapse
Affiliation(s)
- Morten A. Karsdal
- 1Nordic Bioscience A/S, Herlev Hovedgade, Herlev, Denmark; ,2University of Southern Denmark, SDU, Odense, Denmark;
| | | | | | | | | | | | | | | | | | - Aleksander Krag
- 3Department of Gastroenterology and Hepatology, Odense University Hospital, University of Southern Denmark, Odense, Denmark;
| | - Andy Blanchard
- 4GlaxoSmithKline, Medicines Research Centre, Gunnels Wood Road, Stevenage, Hertfordshire, United Kingdom;
| | - Henrik Krarup
- 5Section of Molecular Biology, Clinical Biochemistry, Aalborg University Hospital, Aalborg, Denmark;
| | | | - Detlef Schuppan
- 6Institute of Translational Immunology and Research Center for Immunotherapy, University of Mainz Medical Center, Mainz, Germany; ,7Division of Gastroenterology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
144
|
Govaere O, Roskams T. Pathogenesis and prognosis of hepatocellular carcinoma at the cellular and molecular levels. Clin Liver Dis 2015; 19:261-76. [PMID: 25921662 DOI: 10.1016/j.cld.2015.01.002] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Different approaches predict the outcome for patients with hepatocellular carcinoma (HCC). The expression of biliary-hepatic progenitor cell markers generally correlates with poor prognosis. This article focuses on the pathogenesis of HCC, how differentiation or dedifferentiation leads to a phenotype switch, and heterogeneity in the same tumor. A tumor cell decides its fate based on a complex interplay of signaling pathways. Interaction with the microenvironment decides whether it will invade, proliferate, or enter survival mode. Several signaling pathways contribute to stemness features, reflecting a small chemoresistant subpopulation of the tumor that expresses biliary-hepatic progenitor cell markers.
Collapse
Affiliation(s)
- Olivier Govaere
- Translational Cell and Tissue Research, Department of Imaging and Pathology, KULeuven and University Hospitals Leuven, Minderbroedersstraat 12, Leuven B3000, Belgium.
| | - Tania Roskams
- Translational Cell and Tissue Research, Department of Imaging and Pathology, KULeuven and University Hospitals Leuven, Minderbroedersstraat 12, Leuven B3000, Belgium.
| |
Collapse
|
145
|
Tsuruya K, Chikada H, Ida K, Anzai K, Kagawa T, Inagaki Y, Mine T, Kamiya A. A Paracrine Mechanism Accelerating Expansion of Human Induced Pluripotent Stem Cell-Derived Hepatic Progenitor-Like Cells. Stem Cells Dev 2015; 24:1691-702. [PMID: 25808356 DOI: 10.1089/scd.2014.0479] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Hepatic stem/progenitor cells in liver development have a high proliferative potential and the ability to differentiate into both hepatocytes and cholangiocytes. In this study, we focused on the cell surface molecules of human induced pluripotent stem (iPS) cell-derived hepatic progenitor-like cells (HPCs) and analyzed how these molecules modulate expansion of these cells. Human iPS cells were differentiated into immature hepatic lineage cells by cytokines. In addition to hepatic progenitor markers (CD13 and CD133), the cells were coimmunostained for various cell surface markers (116 types). The cells were analyzed by flow cytometry and in vitro colony formation culture with feeder cells. Twenty types of cell surface molecules were highly expressed in CD13(+)CD133(+) cells derived from human iPS cells. Of these molecules, CD221 (insulin-like growth factor receptor), which was expressed in CD13(+)CD133(+) cells, was quickly downregulated after in vitro expansion. The proliferative ability was suppressed by a neutralizing antibody and specific inhibitor of CD221. Overexpression of CD221 increased colony-forming ability. We also found that inhibition of CD340 (erbB2) and CD266 (fibroblast growth factor-inducible 14) signals suppressed proliferation. In addition, both insulin-like growth factor (a ligand of CD221) and tumor necrosis factor-like weak inducer of apoptosis (a ligand of CD266) were provided by feeder cells in our culture system. This study revealed the expression profiles of cell surface molecules in human iPS cell-derived HPCs and that the paracrine interactions between HPCs and other cells through specific receptors are important for proliferation.
Collapse
Affiliation(s)
- Kota Tsuruya
- 1 Laboratory of Stem Cell Therapy, Institute of Innovative Science and Technology, Tokai University , Isehara, Japan .,2 Division of Gastroenterology, Department of Internal Medicine, School of Medicine, Tokai University , Isehara, Japan
| | - Hiromi Chikada
- 1 Laboratory of Stem Cell Therapy, Institute of Innovative Science and Technology, Tokai University , Isehara, Japan
| | - Kinuyo Ida
- 1 Laboratory of Stem Cell Therapy, Institute of Innovative Science and Technology, Tokai University , Isehara, Japan
| | - Kazuya Anzai
- 1 Laboratory of Stem Cell Therapy, Institute of Innovative Science and Technology, Tokai University , Isehara, Japan .,2 Division of Gastroenterology, Department of Internal Medicine, School of Medicine, Tokai University , Isehara, Japan
| | - Tatehiro Kagawa
- 2 Division of Gastroenterology, Department of Internal Medicine, School of Medicine, Tokai University , Isehara, Japan
| | - Yutaka Inagaki
- 3 Department of Regenerative Medicine, School of Medicine and Center for Matrix Biology and Medicine, Graduate School of Medicine, Tokai University , Isehara, Japan
| | - Tetsuya Mine
- 2 Division of Gastroenterology, Department of Internal Medicine, School of Medicine, Tokai University , Isehara, Japan
| | - Akihide Kamiya
- 1 Laboratory of Stem Cell Therapy, Institute of Innovative Science and Technology, Tokai University , Isehara, Japan
| |
Collapse
|
146
|
Pi L, Jorgensen M, Oh SH, Protopapadakis Y, Gjymishka A, Brown A, Robinson P, Liu C, Scott EW, Schultz GS, Petersen BE. A disintegrin and metalloprotease with thrombospondin type I motif 7: a new protease for connective tissue growth factor in hepatic progenitor/oval cell niche. THE AMERICAN JOURNAL OF PATHOLOGY 2015; 185:1552-63. [PMID: 25843683 DOI: 10.1016/j.ajpath.2015.02.008] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 11/10/2014] [Revised: 02/04/2015] [Accepted: 02/10/2015] [Indexed: 12/14/2022]
Abstract
Hepatic progenitor/oval cell (OC) activation occurs when hepatocyte proliferation is inhibited and is tightly associated with the fibrogenic response during severe liver damage. Connective tissue growth factor (CTGF) is important for OC activation and contributes to the pathogenesis of liver fibrosis. By using the Yeast Two-Hybrid approach, we identified a disintegrin and metalloproteinase with thrombospondin repeat 7 (ADAMTS7) as a CTGF binding protein. In vitro characterization demonstrated CTGF binding and processing by ADAMTS7. Moreover, Adamts7 mRNA was induced during OC activation, after the implantation of 2-acetylaminofluorene with partial hepatectomy in rats or on feeding a 3,5-diethoxycarbonyl-1,4-dihydrocollidine (DDC) diet in mice. X-Gal staining showed Adamts7 expression in hepatocyte nuclear factor 4α(+) hepatocytes and desmin(+) myofibroblasts surrounding reactive ducts in DDC-treated Adamts7(-/-) mice carrying a knocked-in LacZ gene. Adamts7 deficiency was associated with higher transcriptional levels of Ctgf and OC markers and enhanced OC proliferation compared to Adamts7(+/+) controls during DDC-induced liver injury. We also observed increased α-smooth muscle actin and procollagen type I mRNAs, large fibrotic areas in α-smooth muscle actin and Sirius red staining, and increased production of hepatic collagen by hydroxyproline measurement. These results suggest that ADAMTS7 is a new protease for CTGF protein and a novel regulator in the OC compartment, where its absence causes CTGF accumulation, leading to increased OC activation and biliary fibrosis.
Collapse
Affiliation(s)
- Liya Pi
- Department of Pediatrics, University of Florida, Gainesville, Florida.
| | - Marda Jorgensen
- Department of Pediatrics, University of Florida, Gainesville, Florida
| | - Seh-Hoon Oh
- Department of Pediatrics, University of Florida, Gainesville, Florida
| | | | - Altin Gjymishka
- Department of Pediatrics, University of Florida, Gainesville, Florida
| | - Alicia Brown
- Department of Pediatrics, University of Florida, Gainesville, Florida
| | - Paulette Robinson
- Department of Pediatrics, University of Florida, Gainesville, Florida
| | - Chuanju Liu
- Departments of Orthopaedic Surgery and Cell Biology, New York University School of Medicine, New York, New York
| | - Edward W Scott
- Department of Molecular Genetics and Microbiology, University of Florida, Gainesville, Florida
| | - Gregory S Schultz
- Department of Obstetrics and Gynecology, University of Florida, Gainesville, Florida
| | - Bryon E Petersen
- Department of Pediatrics, University of Florida, Gainesville, Florida
| |
Collapse
|
147
|
Doerner JL, Wen J, Xia Y, Paz KB, Schairer D, Wu L, Chalmers SA, Izmirly P, Michaelson JS, Burkly LC, Friedman AJ, Putterman C. TWEAK/Fn14 Signaling Involvement in the Pathogenesis of Cutaneous Disease in the MRL/lpr Model of Spontaneous Lupus. J Invest Dermatol 2015; 135:1986-1995. [PMID: 25826425 PMCID: PMC4504782 DOI: 10.1038/jid.2015.124] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2014] [Revised: 03/09/2015] [Accepted: 03/11/2015] [Indexed: 01/06/2023]
Abstract
Tumor necrosis factor (TNF)-like weak inducer of apoptosis (TWEAK, TNFSF12) and its sole receptor Fn14, belonging to the TNF ligand and receptor superfamilies respectively, are involved in cell survival and cytokine production. The role of TWEAK/Fn14 interactions in the pathogenesis of cutaneous lupus has not been explored. TWEAK treatment of murine PAM212 keratinocytes stimulated the secretion of RANTES via Fn14 and promoted apoptosis. Parthenolide, but not wortmanin or the MAPK inhibitor PD98059, significantly decreased production of RANTES, indicating that this effect of TWEAK is mediated via NF-κB signaling. UVB irradiation significantly upregulated the expression of Fn14 on keratinocytes in vitro and in vivo and increased RANTES production. MRL/lpr Fn14 knockout (KO) lupus mice were compared with MRL/lpr Fn14 wild-type (WT) mice to evaluate for any possible differences in the severity of cutaneous lesions and the presence of infiltrating immune cells. MRL/lpr Fn14 KO mice had markedly attenuated cutaneous disease as compared with their Fn14 WT littermates, as evidenced by the well-maintained architecture of the skin and significantly decreased skin infiltration of T cells and macrophages. Our data strongly implicate TWEAK/Fn14 signaling in the pathogenesis of the cutaneous manifestations in the MRL/lpr model of spontaneous lupus and suggest a possible target for therapeutic intervention.
Collapse
Affiliation(s)
- Jessica L Doerner
- The Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Jing Wen
- The Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Yumin Xia
- The Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Karin Blecher Paz
- The Division of Dermatology, Albert Einstein College of Medicine, Bronx, New York, USA
| | - David Schairer
- The Division of Dermatology, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Lan Wu
- Biogen Idec, Cambridge, Massachusetts, USA
| | - Samantha A Chalmers
- The Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Peter Izmirly
- Division of Rheumatology, NYU-Langone Medical Center, New York, USA
| | | | | | - Adam J Friedman
- The Division of Dermatology, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Chaim Putterman
- The Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York, USA; The Division of Rheumatology, Albert Einstein College of Medicine, Bronx, NY, USA.
| |
Collapse
|
148
|
Cheng Q, Inaba Y, Lu P, Xu M, He J, Zhao Y, Guo GL, Kuruba R, de la Vega R, Evans RW, Li S, Xie W. Chronic activation of FXR in transgenic mice caused perinatal toxicity and sensitized mice to cholesterol toxicity. Mol Endocrinol 2015; 29:571-82. [PMID: 25719402 DOI: 10.1210/me.2014-1337] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
The nuclear receptor farnesoid X receptor (FXR) (nuclear receptor subfamily 1, group H, member 4, or NR1H4) is highly expressed in the liver and intestine. Previous reports have suggested beneficial functions of FXR in the homeostasis of bile acids, lipids, and glucose, as well as in promoting liver regeneration and inhibiting carcinogenesis. To investigate the effect of chronic FXR activation in vivo, we generated transgenic mice that conditionally and tissue specifically express the activated form of FXR in the liver and intestine. Unexpectedly, the transgenic mice showed several intriguing phenotypes, including partial neonatal lethality, growth retardation, and spontaneous liver toxicity. The transgenic mice also displayed heightened sensitivity to a high-cholesterol diet-induced hepatotoxicity but resistance to the gallstone formation. The phenotypes were transgene specific, because they were abolished upon treatment with doxycycline to silence the transgene expression. The perinatal toxicity, which can be rescued by a maternal vitamin supplement, may have resulted from vitamin deficiency due to low biliary bile acid output as a consequence of inhibition of bile acid formation. Our results also suggested that the fibroblast growth factor-inducible immediate-early response protein 14 (Fn14), a member of the proinflammatory TNF family, is a FXR-responsive gene. However, the contribution of Fn14 induction in the perinatal toxic phenotype of the transgenic mice remains to be defined. Because FXR is being explored as a therapeutic target, our results suggested that a chronic activation of this nuclear receptor may have an unintended side effect especially during the perinatal stage.
Collapse
Affiliation(s)
- Qiuqiong Cheng
- Center for Pharmacogenetics and Department of Pharmaceutical Sciences (Q.C., Y.I., P.L., M.X., J.H., Y.Z., R.K., S.L., W.X.), and Departments of Epidemiology (R.D.L.V., R.W.E.) and Pharmacology and Chemical Biology (W.X.), University of Pittsburgh, Pittsburgh, Pennsylvania 15261; and Department of Pharmacology and Toxicology (G.L.G.), Ernest Mario School of Pharmacy, Rutgers University, Piscataway, New Jersey 08854
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
149
|
Best J, Manka P, Syn WK, Dollé L, van Grunsven LA, Canbay A. Role of liver progenitors in liver regeneration. Hepatobiliary Surg Nutr 2015; 4:48-58. [PMID: 25713804 DOI: 10.3978/j.issn.2304-3881.2015.01.16] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2014] [Accepted: 01/20/2015] [Indexed: 12/16/2022]
Abstract
During massive liver injury and hepatocyte loss, the intrinsic regenerative capacity of the liver by replication of resident hepatocytes is overwhelmed. Treatment of this condition depends on the cause of liver injury, though in many cases liver transplantation (LT) remains the only curative option. LT for end stage chronic and acute liver diseases is hampered by shortage of donor organs and requires immunosuppression. Hepatocyte transplantation is limited by yet unresolved technical difficulties. Since currently no treatment is available to facilitate liver regeneration directly, therapies involving the use of resident liver stem or progenitor cells (LPCs) or non-liver stem cells are coming to fore. LPCs are quiescent in the healthy liver, but may be activated under conditions where the regenerative capacity of mature hepatocytes is severely impaired. Non-liver stem cells include embryonic stem cells (ES cells) and mesenchymal stem cells (MSCs). In the first section, we aim to provide an overview of the role of putative cytokines, growth factors, mitogens and hormones in regulating LPC response and briefly discuss the prognostic value of the LPC response in clinical practice. In the latter section, we will highlight the role of other (non-liver) stem cells in transplantation and discuss advantages and disadvantages of ES cells, induced pluripotent stem cells (iPS), as well as MSCs.
Collapse
Affiliation(s)
- Jan Best
- 1 Department of Gastroenterology and Hepatology, University Hospital Essen, Essen, Germany ; 2 Liver Cell Biology Lab, Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel (VUB), Brussels, Belgium ; 3 Regeneration and Repair, The Institute of Hepatology, Foundation for Liver Research, London, UK ; 4 Liver Unit, Barts Health NHS Trust, London, UK ; 5 Department of Surgery, Loyola University Chicago, USA
| | - Paul Manka
- 1 Department of Gastroenterology and Hepatology, University Hospital Essen, Essen, Germany ; 2 Liver Cell Biology Lab, Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel (VUB), Brussels, Belgium ; 3 Regeneration and Repair, The Institute of Hepatology, Foundation for Liver Research, London, UK ; 4 Liver Unit, Barts Health NHS Trust, London, UK ; 5 Department of Surgery, Loyola University Chicago, USA
| | - Wing-Kin Syn
- 1 Department of Gastroenterology and Hepatology, University Hospital Essen, Essen, Germany ; 2 Liver Cell Biology Lab, Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel (VUB), Brussels, Belgium ; 3 Regeneration and Repair, The Institute of Hepatology, Foundation for Liver Research, London, UK ; 4 Liver Unit, Barts Health NHS Trust, London, UK ; 5 Department of Surgery, Loyola University Chicago, USA
| | - Laurent Dollé
- 1 Department of Gastroenterology and Hepatology, University Hospital Essen, Essen, Germany ; 2 Liver Cell Biology Lab, Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel (VUB), Brussels, Belgium ; 3 Regeneration and Repair, The Institute of Hepatology, Foundation for Liver Research, London, UK ; 4 Liver Unit, Barts Health NHS Trust, London, UK ; 5 Department of Surgery, Loyola University Chicago, USA
| | - Leo A van Grunsven
- 1 Department of Gastroenterology and Hepatology, University Hospital Essen, Essen, Germany ; 2 Liver Cell Biology Lab, Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel (VUB), Brussels, Belgium ; 3 Regeneration and Repair, The Institute of Hepatology, Foundation for Liver Research, London, UK ; 4 Liver Unit, Barts Health NHS Trust, London, UK ; 5 Department of Surgery, Loyola University Chicago, USA
| | - Ali Canbay
- 1 Department of Gastroenterology and Hepatology, University Hospital Essen, Essen, Germany ; 2 Liver Cell Biology Lab, Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel (VUB), Brussels, Belgium ; 3 Regeneration and Repair, The Institute of Hepatology, Foundation for Liver Research, London, UK ; 4 Liver Unit, Barts Health NHS Trust, London, UK ; 5 Department of Surgery, Loyola University Chicago, USA
| |
Collapse
|
150
|
Karaca G, Xie G, Moylan C, Swiderska-Syn M, Guy CD, Krüger L, Machado MV, Choi SS, Michelotti GA, Burkly LC, Diehl AM. Role of Fn14 in acute alcoholic steatohepatitis in mice. Am J Physiol Gastrointest Liver Physiol 2015; 308:G325-34. [PMID: 25524063 PMCID: PMC4329478 DOI: 10.1152/ajpgi.00429.2013] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
TNF-like weak inducer of apoptosis (TWEAK) is a growth factor for bipotent liver progenitors that express its receptor, fibroblast growth factor-inducible 14 (Fn14), a TNF receptor superfamily member. Accumulation of Fn14(+) progenitors occurs in severe acute alcoholic steatohepatitis (ASH) and correlates with acute mortality. In patients with severe ASH, inhibition of TNF-α increases acute mortality. The aim of this study was to determine whether deletion of Fn14 improves the outcome of liver injury in alcohol-consuming mice. Wild-type (WT) and Fn14 knockout (KO) mice were fed control high-fat Lieber deCarli diet or high-fat Lieber deCarli diet with 2% alcohol (ETOH) and injected intraperitoneally with CCl₄ for 2 wk to induce liver injury. Mice were euthanized 3 or 10 days after CCl₄ treatment. Survival was assessed. Liver tissues were analyzed for cell death, inflammation, proliferation, progenitor accumulation, and fibrosis by quantitative RT-PCR, immunoblot, hydroxyproline content, and quantitative immunohistochemistry. During liver injury, Fn14 expression, apoptosis, inflammation, hepatocyte replication, progenitor and myofibroblast accumulation, and fibrosis increased in WT mice fed either diet. Mice fed either diet expressed similar TWEAK/Fn14 levels, but ETOH-fed mice had higher TNF-α expression. The ETOH-fed group developed more apoptosis, inflammation, fibrosis, and regenerative responses. Fn14 deletion did not reduce hepatic TNF-α expression but improved all injury parameters in mice fed the control diet. In ETOH-fed mice, Fn14 deletion inhibited TNF-α induction and increased acute mortality, despite improvement in liver injury. Fn14 mediates wound-healing responses that are necessary to survive acute liver injury during alcohol exposure.
Collapse
Affiliation(s)
- Gamze Karaca
- 1Division of Gastroenterology, Department of Medicine, Duke University Medical Center, Durham, North Carolina;
| | - Guanhua Xie
- 1Division of Gastroenterology, Department of Medicine, Duke University Medical Center, Durham, North Carolina;
| | - Cynthia Moylan
- 1Division of Gastroenterology, Department of Medicine, Duke University Medical Center, Durham, North Carolina;
| | - Marzena Swiderska-Syn
- 1Division of Gastroenterology, Department of Medicine, Duke University Medical Center, Durham, North Carolina;
| | - Cynthia D. Guy
- 2Department of Pathology, Duke University Medical Center, Durham, North Carolina;
| | - Leandi Krüger
- 1Division of Gastroenterology, Department of Medicine, Duke University Medical Center, Durham, North Carolina;
| | - Mariana Verdelho Machado
- 1Division of Gastroenterology, Department of Medicine, Duke University Medical Center, Durham, North Carolina;
| | - Steve S. Choi
- 1Division of Gastroenterology, Department of Medicine, Duke University Medical Center, Durham, North Carolina; ,3Section of Gastroenterology, Durham Veterans Affairs Medical Center, Durham, North Carolina; and
| | - Gregory A. Michelotti
- 1Division of Gastroenterology, Department of Medicine, Duke University Medical Center, Durham, North Carolina;
| | - Linda C. Burkly
- 4Department of Immunology, Biogen Idec, Inc., Cambridge, Massachusetts
| | - Anna Mae Diehl
- Division of Gastroenterology, Department of Medicine, Duke University Medical Center, Durham, North Carolina;
| |
Collapse
|