1
|
Wang Y, Piao C, Liu T, Lu X, Ma Y, Zhang J, Ma H, Wang H. Exosomes Derived from Adipose Mesenchymal Stem Cells Promote Regeneration of Injured Liver in Minipigs. Int J Mol Sci 2024; 25:6604. [PMID: 38928308 PMCID: PMC11203699 DOI: 10.3390/ijms25126604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Revised: 06/04/2024] [Accepted: 06/11/2024] [Indexed: 06/28/2024] Open
Abstract
Hepatic ischemia/reperfusion injury (IRI) is an important factor affecting liver regeneration and functional recovery postoperatively. Many studies have suggested that mesenchymal stem cells (MSCs) contribute to hepatic tissue repair and functional recovery through paracrine mechanisms mediated by exosomes. Minipigs exhibit much more similar characteristics of the liver to those of humans than rodents. This study aimed to explore whether exosomes from adipose-derived MSCs (ADSCs-exo) could actively promote liver regeneration after hepatectomy combined with HIRI in minipigs and the role they play in the cell proliferation process. This study also compared the effects and differences in the role of ADSCs and ADSCs-exo in the inflammatory response and liver regeneration. The results showed that ADSCs-exo suppressed histopathological changes and reduced inflammatory infiltration in the liver; significantly decreased levels of ALT, TBIL, HA, and the pro-inflammatory cytokines TNF-α, IL-6, and CRP; increased levels of the anti-inflammatory cytokine IL-10 and the pro-regeneration factors Ki67, PCNA, CyclinD1, HGF, STAT3, VEGF, ANG1, ANG2; and decreased levels of the anti-regeneration factors SOCS3 and TGF-β. These indicators above showed similar changes with the ADSCs intervention group. Indicating that ADSCs-exo can exert the same role as ADSCs in regulating inflammatory responses and promoting liver regeneration. Our findings provide experimental evidence for the possibility that ADSCs-exo could be considered a safe and effective cell-free therapy to promote regeneration of injured livers.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Hongbin Wang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China; (Y.W.); (C.P.); (T.L.); (X.L.); (Y.M.); (J.Z.); (H.M.)
| |
Collapse
|
2
|
Oliva-Vilarnau N, Beusch CM, Sabatier P, Sakaraki E, Tjaden A, Graetz L, Büttner FA, Dorotea D, Nguyen M, Bergqvist F, Sundström Y, Müller S, Zubarev RA, Schulte G, Tredup C, Gramignoli R, Tietge UJ, Lauschke VM. Wnt/β-catenin and NFκB signaling synergize to trigger growth factor-free regeneration of adult primary human hepatocytes. Hepatology 2024; 79:1337-1351. [PMID: 37870288 PMCID: PMC11095891 DOI: 10.1097/hep.0000000000000648] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Accepted: 09/18/2023] [Indexed: 10/24/2023]
Abstract
BACKGROUND AND AIMS The liver has a remarkable capacity to regenerate, which is sustained by the ability of hepatocytes to act as facultative stem cells that, while normally quiescent, re-enter the cell cycle after injury. Growth factor signaling is indispensable in rodents, whereas Wnt/β-catenin is not required for effective tissue repair. However, the molecular networks that control human liver regeneration remain unclear. METHODS Organotypic 3D spheroid cultures of primary human or murine hepatocytes were used to identify the signaling network underlying cell cycle re-entry. Furthermore, we performed chemogenomic screening of a library enriched for epigenetic regulators and modulators of immune function to determine the importance of epigenomic control for human hepatocyte regeneration. RESULTS Our results showed that, unlike in rodents, activation of Wnt/β-catenin signaling is the major mitogenic cue for adult primary human hepatocytes. Furthermore, we identified TGFβ inhibition and inflammatory signaling through NF-κB as essential steps for the quiescent-to-regenerative switch that allows Wnt/β-catenin-induced proliferation of human cells. In contrast, growth factors, but not Wnt/β-catenin signaling, triggered hyperplasia in murine hepatocytes. High-throughput screening in a human model confirmed the relevance of NFκB and revealed the critical roles of polycomb repressive complex 2, as well as of the bromodomain families I, II, and IV. CONCLUSIONS This study revealed a network of NFκB, TGFβ, and Wnt/β-catenin that controls human hepatocyte regeneration in the absence of exogenous growth factors, identified novel regulators of hepatocyte proliferation, and highlighted the potential of organotypic culture systems for chemogenomic interrogation of complex physiological processes.
Collapse
Affiliation(s)
- Nuria Oliva-Vilarnau
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Christian M. Beusch
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Pierre Sabatier
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Eirini Sakaraki
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Amelie Tjaden
- Institute of Pharmaceutical Chemistry, Johann Wolfgang Goethe University, Frankfurt am Main, Germany
- Buchmann Institute for Molecular Life Sciences and Structural Genomics Consortium (SGC), Frankfurt am Main, Germany
| | - Lukas Graetz
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Florian A. Büttner
- Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany
- University of Tübingen, Tübingen, Germany
| | - Debra Dorotea
- Department of Laboratory Medicine, Division of Clinical Chemistry, Karolinska Institutet, Stockholm, Sweden
| | - My Nguyen
- Department of Laboratory Medicine, Division of Clinical Chemistry, Karolinska Institutet, Stockholm, Sweden
| | - Filip Bergqvist
- Department of Medicine, Karolinska Institutet, and Karolinska University Hospital, Stockholm, Sweden
- The Structural Genomics Consortium (SGC), Karolinska Institutet, Stockholm, Sweden
| | - Yvonne Sundström
- Department of Medicine, Karolinska Institutet, and Karolinska University Hospital, Stockholm, Sweden
- The Structural Genomics Consortium (SGC), Karolinska Institutet, Stockholm, Sweden
| | - Susanne Müller
- Institute of Pharmaceutical Chemistry, Johann Wolfgang Goethe University, Frankfurt am Main, Germany
- Buchmann Institute for Molecular Life Sciences and Structural Genomics Consortium (SGC), Frankfurt am Main, Germany
| | - Roman A. Zubarev
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Gunnar Schulte
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Claudia Tredup
- Institute of Pharmaceutical Chemistry, Johann Wolfgang Goethe University, Frankfurt am Main, Germany
- Buchmann Institute for Molecular Life Sciences and Structural Genomics Consortium (SGC), Frankfurt am Main, Germany
| | - Roberto Gramignoli
- Department of Laboratory Medicine, Division of Pathology, Karolinska Institutet, Stockholm, Sweden
- Clinical Pathology and Cancer Diagnosis Unit, Karolinska University Hospital, Stockholm, Sweden
| | - Uwe J.F. Tietge
- Department of Laboratory Medicine, Division of Clinical Chemistry, Karolinska Institutet, Stockholm, Sweden
- Clinical Chemistry, Karolinska University Laboratory, Karolinska University Hospital, Stockholm, Sweden
| | - Volker M. Lauschke
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
- Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany
- University of Tübingen, Tübingen, Germany
| |
Collapse
|
3
|
Karima G, Kim HD. Unlocking the regenerative key: Targeting stem cell factors for bone renewal. J Tissue Eng 2024; 15:20417314241287491. [PMID: 39479284 PMCID: PMC11523181 DOI: 10.1177/20417314241287491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2024] [Accepted: 09/12/2024] [Indexed: 11/02/2024] Open
Abstract
Stem cell factors (SCFs) are pivotal factors existing in both soluble and membrane-bound forms, expressed by endothelial cells (ECs) and fibroblasts throughout the body. These factors enhance cell growth, viability, and migration in multipotent cell lineages. The preferential expression of SCF by arteriolar ECs indicates that arterioles create a unique microenvironment tailored to hematopoietic stem cells (HSCs). Insufficiency of SCF within bone marrow (BM)-derived adipose tissue results in decreased their overall cellularity, affecting HSCs and their immediate progenitors critical for generating diverse blood cells and maintaining the hematopoietic microenvironment. SCF deficiency disrupts BM function, impacting the production and differentiation of HSCs. Additionally, deleting SCF from adipocytes reduces lipogenesis, highlighting the crucial role of SCF/c-kit signaling in controlling lipid accumulation. This review elucidates the sources, roles, mechanisms, and molecular strategies of SCF in bone renewal, offering a comprehensive overview of recent advancements, challenges, and future directions for leveraging SCF as a key agent in regenerative medicine.
Collapse
Affiliation(s)
- Gul Karima
- Department of Polymer Science and Engineering, Korea National University of Transportation, Chungju, Republic of Korea
| | - Hwan D. Kim
- Department of Polymer Science and Engineering, Korea National University of Transportation, Chungju, Republic of Korea
- Department of IT Convergence (Brain Korea Plus 21), Korea National University of Transportation, Chungju, Republic of Korea
- Department of Biomedical Engineering, Korea National University of Transportation, Chungju, Republic of Korea
| |
Collapse
|
4
|
Cai YL, Nan F, Tang GT, Ma Y, Ren Y, Xiong XZ, Zhou RX, Li FY, Cheng NS, Jiang X. Fabrication of 3D printed PCL/PEG artificial bile ducts as supportive scaffolds to promote regeneration of extrahepatic bile ducts in a canine biliary defect model. J Mater Chem B 2023; 11:9443-9458. [PMID: 37727116 DOI: 10.1039/d3tb01250f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/21/2023]
Abstract
In this study, a 3D porous poly(ε-caprolactone)/polyethylene glycol (PCL/PEG) composite artificial tubular bile duct was fabricated for extrahepatic bile duct regeneration. PCL/PEG composite scaffolds were fabricated by 3D printing, and the molecular structure, mechanical properties, thermal properties, morphology, and in vitro biocompatibility were characterized for further application as artificial bile ducts. A bile duct defect model was established in beagle dogs for in vivo implantation. The results demonstrated that the implanted PE1 ABD, serving as a supportive scaffold, effectively stimulated the regeneration of a new bile duct comprising CK19-positive and CK7-positive epithelial cells within 30 days. Remarkably, after 8 months, the newly formed bile duct exhibited an epithelial layer resembling the normal structure. Furthermore, the study revealed collagen deposition, biliary muscular formation, and the involvement of microvessels and fibroblasts in the regenerative process. In contrast, the anastomotic area without ABD implantation displayed only partial restoration of the epithelial layer, accompanied by fibroblast proliferation and subsequent bile duct fibrosis. These findings underscore the limited inherent repair capacity of the bile duct and underscore the beneficial role of the PE1 ABD artificial tubular bile duct in promoting biliary regeneration.
Collapse
Affiliation(s)
- Yu-Long Cai
- Division of Biliary Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China
- Research Center for Biliary Diseases, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China
| | - Fang Nan
- Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China.
| | - Guo-Tao Tang
- Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China.
| | - Yuan Ma
- Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China.
| | - Yi Ren
- Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China.
| | - Xian-Ze Xiong
- Division of Biliary Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China
- Research Center for Biliary Diseases, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China
| | - Rong-Xing Zhou
- Division of Biliary Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China
- Research Center for Biliary Diseases, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China
| | - Fu-Yu Li
- Division of Biliary Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China
- Research Center for Biliary Diseases, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China
| | - Nan-Sheng Cheng
- Division of Biliary Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China
- Research Center for Biliary Diseases, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China
| | - Xia Jiang
- Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China.
| |
Collapse
|
5
|
Wolf SD, Ehlting C, Müller-Dott S, Poschmann G, Petzsch P, Lautwein T, Wang S, Helm B, Schilling M, Saez-Rodriguez J, Vucur M, Stühler K, Köhrer K, Tacke F, Dooley S, Klingmüller U, Luedde T, Bode JG. Hepatocytes reprogram liver macrophages involving control of TGF-β activation, influencing liver regeneration and injury. Hepatol Commun 2023; 7:e0208. [PMID: 37486964 PMCID: PMC10368377 DOI: 10.1097/hc9.0000000000000208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Accepted: 05/12/2023] [Indexed: 07/26/2023] Open
Abstract
BACKGROUND Macrophages play an important role in maintaining liver homeostasis and regeneration. However, it is not clear to what extent the different macrophage populations of the liver differ in terms of their activation state and which other liver cell populations may play a role in regulating the same. METHODS Reverse transcription PCR, flow cytometry, transcriptome, proteome, secretome, single cell analysis, and immunohistochemical methods were used to study changes in gene expression as well as the activation state of macrophages in vitro and in vivo under homeostatic conditions and after partial hepatectomy. RESULTS We show that F4/80+/CD11bhi/CD14hi macrophages of the liver are recruited in a C-C motif chemokine receptor (CCR2)-dependent manner and exhibit an activation state that differs substantially from that of the other liver macrophage populations, which can be distinguished on the basis of CD11b and CD14 expressions. Thereby, primary hepatocytes are capable of creating an environment in vitro that elicits the same specific activation state in bone marrow-derived macrophages as observed in F4/80+/CD11bhi/CD14hi liver macrophages in vivo. Subsequent analyses, including studies in mice with a myeloid cell-specific deletion of the TGF-β type II receptor, suggest that the availability of activated TGF-β and its downregulation by a hepatocyte-conditioned milieu are critical. Reduction of TGF-βRII-mediated signal transduction in myeloid cells leads to upregulation of IL-6, IL-10, and SIGLEC1 expression, a hallmark of the activation state of F4/80+/CD11bhi/CD14hi macrophages, and enhances liver regeneration. CONCLUSIONS The availability of activated TGF-β determines the activation state of specific macrophage populations in the liver, and the observed rapid transient activation of TGF-β may represent an important regulatory mechanism in the early phase of liver regeneration in this context.
Collapse
Affiliation(s)
- Stephanie D Wolf
- Department of Gastroenterology, Hepatology and Infectious Disease, Faculty of Medicine & Düsseldorf University Hospital, Heinrich Heine University, Düsseldorf, Germany
| | - Christian Ehlting
- Department of Gastroenterology, Hepatology and Infectious Disease, Faculty of Medicine & Düsseldorf University Hospital, Heinrich Heine University, Düsseldorf, Germany
| | - Sophia Müller-Dott
- Institute for Computational Biomedicine, Faculty of Medicine & Heidelberg University Hospital, Heidelberg University, Heidelberg, Germany
| | - Gereon Poschmann
- Molecular Proteomics Laboratory, BMFZ, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Patrick Petzsch
- Genomics & Transcriptomics Laboratory, BMFZ, Heinrich Heine University, Düsseldorf, Germany
| | - Tobias Lautwein
- Genomics & Transcriptomics Laboratory, BMFZ, Heinrich Heine University, Düsseldorf, Germany
| | - Sai Wang
- Molecular Hepatology Section, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Barbara Helm
- Division of Systems Biology of Signal Transduction, German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance, Heidelberg, Germany
| | - Marcel Schilling
- Division of Systems Biology of Signal Transduction, German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance, Heidelberg, Germany
| | - Julio Saez-Rodriguez
- Institute for Computational Biomedicine, Faculty of Medicine & Heidelberg University Hospital, Heidelberg University, Heidelberg, Germany
| | - Mihael Vucur
- Department of Gastroenterology, Hepatology and Infectious Disease, Faculty of Medicine & Düsseldorf University Hospital, Heinrich Heine University, Düsseldorf, Germany
| | - Kai Stühler
- Molecular Proteomics Laboratory, BMFZ, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- Institute of Molecular Medicine, Proteome Research, Medical Faculty and University Hospital, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Karl Köhrer
- Genomics & Transcriptomics Laboratory, BMFZ, Heinrich Heine University, Düsseldorf, Germany
| | - Frank Tacke
- Department of Hepatology & Gastroenterology, Charité Universitätsmedizin Berlin, Campus Virchow-Klinikum (CVK) and Campus Charité Mitte (CCM), Berlin, Germany
| | - Steven Dooley
- Molecular Hepatology Section, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Ursula Klingmüller
- Division of Systems Biology of Signal Transduction, German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance, Heidelberg, Germany
| | - Tom Luedde
- Department of Gastroenterology, Hepatology and Infectious Disease, Faculty of Medicine & Düsseldorf University Hospital, Heinrich Heine University, Düsseldorf, Germany
| | - Johannes G Bode
- Department of Gastroenterology, Hepatology and Infectious Disease, Faculty of Medicine & Düsseldorf University Hospital, Heinrich Heine University, Düsseldorf, Germany
| |
Collapse
|
6
|
Paluschinski M, Kordes C, Vucur M, Buettner V, Roderburg C, Xu HC, Shinte PV, Lang PA, Luedde T, Castoldi M. Differential Modulation of miR-122 Transcription by TGFβ1/BMP6: Implications for Nonresolving Inflammation and Hepatocarcinogenesis. Cells 2023; 12:1955. [PMID: 37566034 PMCID: PMC10416984 DOI: 10.3390/cells12151955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 07/24/2023] [Accepted: 07/26/2023] [Indexed: 08/12/2023] Open
Abstract
Chronic inflammation is widely recognized as a significant factor that promotes and worsens the development of malignancies, including hepatocellular carcinoma. This study aimed to explore the potential role of microRNAs in inflammation-associated nonresolving hepatocarcinogenesis. By conducting a comprehensive analysis of altered microRNAs in animal models with liver cancer of various etiologies, we identified miR-122 as the most significantly downregulated microRNA in the liver of animals with inflammation-associated liver cancer. Although previous research has indicated the importance of miR-122 in maintaining hepatocyte function, its specific role as either the trigger or the consequence of underlying diseases remains unclear. Through extensive analysis of animals and in vitro models, we have successfully demonstrated that miR-122 transcription is differentially regulated by the immunoregulatory cytokines, by the transforming growth factor-beta 1 (TGFβ1), and the bone morphogenetic protein-6 (BMP6). Furthermore, we presented convincing evidence directly linking reduced miR-122 transcription to inflammation and in chronic liver diseases. The results of this study strongly suggest that prolonged activation of pro-inflammatory signaling pathways, leading to disruption of cytokine-mediated regulation of miR-122, may significantly contribute to the onset and exacerbation of chronic liver disease.
Collapse
Affiliation(s)
- Martha Paluschinski
- Department of Gastroenterology, Hepatology and Infectious Diseases, Medical Faculty, University Hospital, Heinrich Heine University Dusseldorf, 40225 Dusseldorf, Germany; (M.P.); (C.K.); (M.V.); (V.B.); (C.R.); (T.L.)
| | - Claus Kordes
- Department of Gastroenterology, Hepatology and Infectious Diseases, Medical Faculty, University Hospital, Heinrich Heine University Dusseldorf, 40225 Dusseldorf, Germany; (M.P.); (C.K.); (M.V.); (V.B.); (C.R.); (T.L.)
| | - Mihael Vucur
- Department of Gastroenterology, Hepatology and Infectious Diseases, Medical Faculty, University Hospital, Heinrich Heine University Dusseldorf, 40225 Dusseldorf, Germany; (M.P.); (C.K.); (M.V.); (V.B.); (C.R.); (T.L.)
| | - Veronika Buettner
- Department of Gastroenterology, Hepatology and Infectious Diseases, Medical Faculty, University Hospital, Heinrich Heine University Dusseldorf, 40225 Dusseldorf, Germany; (M.P.); (C.K.); (M.V.); (V.B.); (C.R.); (T.L.)
| | - Christoph Roderburg
- Department of Gastroenterology, Hepatology and Infectious Diseases, Medical Faculty, University Hospital, Heinrich Heine University Dusseldorf, 40225 Dusseldorf, Germany; (M.P.); (C.K.); (M.V.); (V.B.); (C.R.); (T.L.)
| | - Haifeng C. Xu
- Institute for Molecular Medicine II, Medical Faculty, Heinrich-Heine University Hospital, 40225 Dusseldorf, Germany; (H.C.X.); (P.V.S.); (P.A.L.)
| | - Prashant V. Shinte
- Institute for Molecular Medicine II, Medical Faculty, Heinrich-Heine University Hospital, 40225 Dusseldorf, Germany; (H.C.X.); (P.V.S.); (P.A.L.)
| | - Philipp A. Lang
- Institute for Molecular Medicine II, Medical Faculty, Heinrich-Heine University Hospital, 40225 Dusseldorf, Germany; (H.C.X.); (P.V.S.); (P.A.L.)
| | - Tom Luedde
- Department of Gastroenterology, Hepatology and Infectious Diseases, Medical Faculty, University Hospital, Heinrich Heine University Dusseldorf, 40225 Dusseldorf, Germany; (M.P.); (C.K.); (M.V.); (V.B.); (C.R.); (T.L.)
| | - Mirco Castoldi
- Department of Gastroenterology, Hepatology and Infectious Diseases, Medical Faculty, University Hospital, Heinrich Heine University Dusseldorf, 40225 Dusseldorf, Germany; (M.P.); (C.K.); (M.V.); (V.B.); (C.R.); (T.L.)
| |
Collapse
|
7
|
Zhang X, Li S, Ren X, Xiang P, Zhang Y, Wang T, Qin Q, Sun F, Liu J, Gao L, Ma C, Yue X, Yang X, Han S, Liang X. TIPE1 promotes liver regeneration by enhancing ROS-FoxO1 axis mediated autophagy. FEBS J 2023; 290:1117-1133. [PMID: 36111440 DOI: 10.1111/febs.16629] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 07/15/2022] [Accepted: 08/22/2022] [Indexed: 11/30/2022]
Abstract
The strong regenerative ability of the liver safeguards the crucial hepatic functions. The balance between hepatocyte proliferation and death is critical for restoring liver size and physiology. Tumour necrosis factor (TNF) alpha-induced protein 8-like 1 (TIPE1) is highly expressed in liver and has been identified as a candidate regulator for cell proliferation and death, being involved in a variety of biological processes and diseases. However, the role of TIPE1 in liver regeneration remains unexplored. In the present study, we found that TIPE1 expression was elevated in the regenerating liver induced by either partial hepatectomy or 10% carbon tetrachloride administration. Mice with hepatocyte conditional Tipe1 knockout presented significantly impaired liver regeneration. Mechanistically, hepatic Tipe1 deficiency decreased the level of reactive oxygen species in hepatocytes, which in turn led to the inhibition of Forkhead box O1 acetylation and microtubule-associated protein 1 light chain 3 I to microtubule-associated protein 1 light chain 3 II conversion, and the accumulation of sequestosome 1. By contrast, forced expression of TIPE1 in hepatocyte significantly promoted liver regeneration following 70% partial hepatectomy and enhanced hepatocyte reactive oxygen species/acetylated-Forkhead box O1 level and autophagy. These findings indicate that TIPE1 plays a crucial role in liver regeneration by finely regulating the oxidative stress and autophagy and is a potential target for medical intervention of liver regeneration.
Collapse
Affiliation(s)
- Xiaodong Zhang
- Depertment of Central Laboratory and Mitochondrial Medicine Laboratory, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, China
| | - Shuangjie Li
- Key Laboratory for Experimental Teratology of Ministry of Education and Department of Immunology, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Xiaolei Ren
- Key Laboratory for Experimental Teratology of Ministry of Education and Department of Immunology, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Peng Xiang
- Key Laboratory for Experimental Teratology of Ministry of Education and Department of Immunology, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Yankun Zhang
- Key Laboratory for Experimental Teratology of Ministry of Education and Department of Immunology, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Tixiao Wang
- Key Laboratory for Experimental Teratology of Ministry of Education and Department of Immunology, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Qinghua Qin
- Key Laboratory for Experimental Teratology of Ministry of Education and Department of Cell Biology, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Fengkai Sun
- Department of Gastroenterology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Jingkang Liu
- Department of Gynecology, Qilu Hospital of Shandong University, Jinan, China
| | - Lifen Gao
- Key Laboratory for Experimental Teratology of Ministry of Education and Department of Immunology, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Chunhong Ma
- Key Laboratory for Experimental Teratology of Ministry of Education and Department of Immunology, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Xuetian Yue
- Key Laboratory for Experimental Teratology of Ministry of Education and Department of Cell Biology, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Xiaoyun Yang
- Department of Gastroenterology, Qilu Hospital, Shandong University, Jinan, China
| | - Shuang Han
- Department of Gastroenterology, Honghui Hospital, Xi'an Jiaotong University, China
| | - Xiaohong Liang
- Key Laboratory for Experimental Teratology of Ministry of Education and Department of Immunology, Cheeloo College of Medicine, Shandong University, Jinan, China
| |
Collapse
|
8
|
Loss of YB-1 alleviates liver fibrosis by suppressing epithelial-mesenchymal transition in hepatic progenitor cells. Biochim Biophys Acta Mol Basis Dis 2022; 1868:166510. [DOI: 10.1016/j.bbadis.2022.166510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 07/26/2022] [Accepted: 07/27/2022] [Indexed: 11/19/2022]
|
9
|
Jiao Z, Ma Y, Zhang Q, Wang Y, Liu T, Liu X, Piao C, Liu B, Wang H. The adipose-derived mesenchymal stem cell secretome promotes hepatic regeneration in miniature pigs after liver ischaemia-reperfusion combined with partial resection. Stem Cell Res Ther 2021; 12:218. [PMID: 33781342 PMCID: PMC8008619 DOI: 10.1186/s13287-021-02284-y] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Accepted: 03/11/2021] [Indexed: 02/06/2023] Open
Abstract
Background Hepatic ischaemia-reperfusion injury (HIRI) is inevitable in complicated liver surgery and is a major factor leading to postoperative complications and liver dysfunction. Studies have shown that the paracrine mechanisms of stem cell may be essential to tissue repair and functional improvement after transplantation. However, the role of the adipose-derived mesenchymal stem cell secretome (ASC-secretome) in liver regeneration in large animals remains to be determined. Methods Twenty-four miniature pigs were subjected to laparoscopic liver ischaemia-reperfusion combined with partial hepatectomy and divided into the following four groups: the saline group, the DMEM group, the ASC group and the ASC-secretome group. Serum and liver tissue samples were collected before the operation and at 1, 3 and 7 days after the operation, and changes in tissue pathology, serum inflammation, liver function, angiogenesis-related factors and liver tissue regeneration-related genes and proteins were evaluated. Results Detailed histological analysis showed that ASCs and the ASC-secretome changed pathological damage to liver tissue after liver ischaemia-reperfusion combined with partial hepatectomy (1 and 3 days: p < 0.01). Compared with the saline and DMEM control groups, the ASC-secretome group had significantly reduced expression levels of ALP (1 and 3 days: p < 0.05), ALT (1 day: p < 0.01; 3 days: p < 0.05) and AST (1 and 3 days: p < 0.01), which promoted the recovery of liver function. Moreover, detection of the expression levels of TNF-α and IL-1β (1 day: p < 0.01; 3 days: p < 0.05), IL-6 (1 and 3 days: p < 0.05) and IL-10 (1 and 3 days: p < 0.01) in serum confirmed that the ASC-secretome had obvious anti-inflammatory effects. In addition, the ASC-secretome increased the expression levels of ANG-1 (3 days: p < 0.01), ANG-2 (3 and 7 days: p < 0.01) and VEGF (1 and 7 days: p < 0.05; 3 days: p < 0.01) and promoted angiogenesis during liver regeneration. Moreover, it promoted the mRNA expression of HGF and Cyclin D1 (1 and 3 days: p < 0.01); increased the levels of p-STAT3 (1 and 3 days: p < 0.01), PCNA and Ki67 (1 and 3 days: p < 0.01; 7 days: p < 0.05); inhibited the negative feedback of SOCS3 (1 and 3 days: p < 0.01); and decreased the mRNA expression of TGF-β (3 days: p < 0.01). The cytokines and growth factors detected in the ASC-secretome included TNF-α, IL-6, IL-1β, ANG-1, ANG-2, VEGF and b-FGF. Conclusion The ASC-secretome alleviates the inflammatory response induced by ischaemia-reperfusion combined with partial hepatectomy in miniature pigs and promotes liver regeneration.
Collapse
Affiliation(s)
- Zhihui Jiao
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, People's Republic of China.,College of Wildlife and Protected Area, Northeast Forestry University, Harbin, People's Republic of China
| | - Yajun Ma
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, People's Republic of China
| | - Qianzhen Zhang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, People's Republic of China
| | - Yue Wang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, People's Republic of China
| | - Tao Liu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, People's Republic of China
| | - Xiaoning Liu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, People's Republic of China
| | - Chenxi Piao
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, People's Republic of China
| | - Boyang Liu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, People's Republic of China.,College of Wildlife and Protected Area, Northeast Forestry University, Harbin, People's Republic of China
| | - Hongbin Wang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, People's Republic of China.
| |
Collapse
|
10
|
Abarca-Buis RF, Mandujano-Tinoco EA, Cabrera-Wrooman A, Krötzsch E. The complexity of TGFβ/activin signaling in regeneration. J Cell Commun Signal 2021; 15:7-23. [PMID: 33481173 DOI: 10.1007/s12079-021-00605-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Accepted: 01/05/2021] [Indexed: 12/11/2022] Open
Abstract
The role of transforming growth factor β TGFβ/activin signaling in wound repair and regeneration is highly conserved in the animal kingdom. Various studies have shown that TGF-β/activin signaling can either promote or inhibit different aspects of the regeneration process (i.e., proliferation, differentiation, and re-epithelialization). It has been demonstrated in several biological systems that some of the different cellular responses promoted by TGFβ/activin signaling depend on the activation of Smad-dependent or Smad-independent signal transduction pathways. In the context of regeneration and wound healing, it has been shown that the type of R-Smad stimulated determines the different effects that can be obtained. However, neither the possible roles of Smad-independent pathways nor the interaction of the TGFβ/activin pathway with other complex signaling networks involved in the regenerative process has been studied extensively. Here, we review the important aspects concerning the TGFβ/activin signaling pathway in the regeneration process. We discuss data regarding the role of TGF-β/activin in the most common animal regenerative models to demonstrate how this signaling promotes or inhibits regeneration, depending on the cellular context.
Collapse
Affiliation(s)
- René Fernando Abarca-Buis
- Laboratory of Connective Tissue, Centro Nacional de Investigación y Atención de Quemados, Instituto Nacional de Rehabilitación "Luís Guillermo Ibarra Ibarra", Calzada México-Xochimilco No. 289, Col. Arenal de Guadalupe, Tlalpan, 14389, Mexico City, Mexico.
| | - Edna Ayerim Mandujano-Tinoco
- Laboratory of Connective Tissue, Centro Nacional de Investigación y Atención de Quemados, Instituto Nacional de Rehabilitación "Luís Guillermo Ibarra Ibarra", Calzada México-Xochimilco No. 289, Col. Arenal de Guadalupe, Tlalpan, 14389, Mexico City, Mexico
| | - Alejandro Cabrera-Wrooman
- Laboratory of Connective Tissue, Centro Nacional de Investigación y Atención de Quemados, Instituto Nacional de Rehabilitación "Luís Guillermo Ibarra Ibarra", Calzada México-Xochimilco No. 289, Col. Arenal de Guadalupe, Tlalpan, 14389, Mexico City, Mexico
| | - Edgar Krötzsch
- Laboratory of Connective Tissue, Centro Nacional de Investigación y Atención de Quemados, Instituto Nacional de Rehabilitación "Luís Guillermo Ibarra Ibarra", Calzada México-Xochimilco No. 289, Col. Arenal de Guadalupe, Tlalpan, 14389, Mexico City, Mexico
| |
Collapse
|
11
|
So J, Kim A, Lee SH, Shin D. Liver progenitor cell-driven liver regeneration. Exp Mol Med 2020; 52:1230-1238. [PMID: 32796957 PMCID: PMC8080804 DOI: 10.1038/s12276-020-0483-0] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Revised: 06/08/2020] [Accepted: 06/17/2020] [Indexed: 12/28/2022] Open
Abstract
The liver is a highly regenerative organ, but its regenerative capacity is compromised in severe liver diseases. Hepatocyte-driven liver regeneration that involves the proliferation of preexisting hepatocytes is a primary regeneration mode. On the other hand, liver progenitor cell (LPC)-driven liver regeneration that involves dedifferentiation of biliary epithelial cells or hepatocytes into LPCs, LPC proliferation, and subsequent differentiation of LPCs into hepatocytes is a secondary mode. This secondary mode plays a significant role in liver regeneration when the primary mode does not effectively work, as observed in severe liver injury settings. Thus, promoting LPC-driven liver regeneration may be clinically beneficial to patients with severe liver diseases. In this review, we describe the current understanding of LPC-driven liver regeneration by exploring current knowledge on the activation, origin, and roles of LPCs during regeneration. We also describe animal models used to study LPC-driven liver regeneration, given their potential to further deepen our understanding of the regeneration process. This understanding will eventually contribute to developing strategies to promote LPC-driven liver regeneration in patients with severe liver diseases.
Collapse
Affiliation(s)
- Juhoon So
- Department of Developmental Biology, McGowan Institute for Regenerative Medicine, Pittsburgh Liver Research Center, University of Pittsburgh, Pittsburgh, PA, 15260, USA.
| | - Angie Kim
- Department of Developmental Biology, McGowan Institute for Regenerative Medicine, Pittsburgh Liver Research Center, University of Pittsburgh, Pittsburgh, PA, 15260, USA
| | - Seung-Hoon Lee
- Department of Developmental Biology, McGowan Institute for Regenerative Medicine, Pittsburgh Liver Research Center, University of Pittsburgh, Pittsburgh, PA, 15260, USA
| | - Donghun Shin
- Department of Developmental Biology, McGowan Institute for Regenerative Medicine, Pittsburgh Liver Research Center, University of Pittsburgh, Pittsburgh, PA, 15260, USA.
| |
Collapse
|
12
|
Ohshiro K, Chen J, Srivastav J, Mishra L, Mishra B. Alterations in TGF-β signaling leads to high HMGA2 levels potentially through modulation of PJA1/SMAD3 in HCC cells. Genes Cancer 2020; 11:43-52. [PMID: 32577156 PMCID: PMC7289907 DOI: 10.18632/genesandcancer.199] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Recently, we observed that the TGF-β pathway is altered in 39% of HCCs. The alterations are correlated with a raised HMGA2 level. Therefore, we compared genetic alterations of HMGA2 and 43 TGF-β pathway core genes in HCC patients from TCGA database. Genetic alterations of 15 genes, including INHBE, INHBC, GDF11, ACVRL and TGFB2 out of 43 core genes, highly-moderately matched that of HMGA2. Co-occurrences of mutation amplification, gains, deletions and high/low mRNA of HMGA2 with those of the core genes were highly significant in INHBE, INHBC, ACVR1B, ACVRL and GDF11. Mass spectrometry studies revealed that HMGA2 interacted with an E3 ligase, PJA1, and that this interaction is enhanced by TGF-β treatment in the nuclear of HCC cells. Co-localization of nuclear PJA1 and HMGA2 in HCC cells increased upon TGF-β treatment. Raised HMGA2 levels that occur with alterations in the TGF-β signaling pathway may reflect an altered activity of E3 ligases, such as PJA1, and potentially contribute to the tumor-promoting roles of TGF-β signaling. Here, we report that the co-occurrence of genetic alterations in HMGA2 and TGF-β pathway core genes is implicated in HCC progression, and propose that HMGA2 and PJA1 may be potential novel targets in dysfunctional TGF-β signaling in HCC.
Collapse
Affiliation(s)
- Kazufumi Ohshiro
- Department of Surgery, Center for Translational Medicine, George Washington University, Washington DC, USA
| | - Jian Chen
- Department of Gastroenterology, Hepatology, and Nutrition, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | - Lopa Mishra
- Department of Surgery, Center for Translational Medicine, George Washington University, Washington DC, USA.,Department of Gastroenterology and Hepatology, VA Medical Center, Washington DC, USA
| | - Bibhuti Mishra
- Department of Surgery, Center for Translational Medicine, George Washington University, Washington DC, USA
| |
Collapse
|
13
|
Tang N, Zhang J, Fu X, Xie W, Qiu Y. PP2Acα inhibits PFKFB2-induced glycolysis to promote termination of liver regeneration. Biochem Biophys Res Commun 2020; 526:1-7. [PMID: 32192773 DOI: 10.1016/j.bbrc.2020.03.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Accepted: 03/03/2020] [Indexed: 01/17/2023]
Abstract
The mechanisms underlying the initiation and proliferation of liver regeneration (LR) has been extensively studied using the partial hepatectomy (PHx) model, while little is known about the termination of LR. PP2Acα (protein phosphatase 2 A catalytic subunit α isoform) is the catalytic subunit of protein phosphatase 2 A (PP2A), accounting for most of intracellular serine/threonine phosphatase activity. We have previously observed that termination of LR delayed in PP2Acα liver-specific knockout (LKO) mice after PHx. In our study, we used phospho explorer antibody array analysis to screen the potential phosphorylation targets of PP2Acα, and PP2Acα had a great influence on the hepatic phosphoproteomic signaling in the termination of LR after PHx. We then tested the phosphorylation changes and metabolic function of 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase-2 (PFKFB2), an isoform of the key glycolytic enzyme PFKFB, which was significantly regulated by PP2Acα knockout. PP2Acα knockout enhanced glycolysis in vivo and in vitro, while adenoviral-mediated RNAi of PFKFB2 reversed the extension of postoperative liver regeneration in KO mice along with the downregulation of glycolysis. Therefore, we demonstrated that PP2Acα liver-specific knockout regulated the hepatocytes glycolysis via activating PFKFB2, thus enhancing liver regeneration during the termination stage.
Collapse
Affiliation(s)
- Neng Tang
- The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Jingzi Zhang
- Medical School and Model Animal Research Center of Nanjing University, Nanjing, China
| | - Xiao Fu
- The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Weiqi Xie
- The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Yudong Qiu
- The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China.
| |
Collapse
|
14
|
Chen J, Mitra A, Li S, Song S, Nguyen BN, Chen JS, Shin JH, Gough NR, Lin P, Obias V, He AR, Yao Z, Malta TM, Noushmehr H, Latham PS, Su X, Rashid A, Mishra B, Wu RC, Mishra L. Targeting the E3 Ubiquitin Ligase PJA1 Enhances Tumor-Suppressing TGFβ Signaling. Cancer Res 2020; 80:1819-1832. [PMID: 32127355 DOI: 10.1158/0008-5472.can-19-3116] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Revised: 01/17/2020] [Accepted: 02/24/2020] [Indexed: 12/22/2022]
Abstract
RING-finger E3 ligases are instrumental in the regulation of inflammatory cascades, apoptosis, and cancer. However, their roles are relatively unknown in TGFβ/SMAD signaling. SMAD3 and its adaptors, such as β2SP, are important mediators of TGFβ signaling and regulate gene expression to suppress stem cell-like phenotypes in diverse cancers, including hepatocellular carcinoma (HCC). Here, PJA1, an E3 ligase, promoted ubiquitination and degradation of phosphorylated SMAD3 and impaired a SMAD3/β2SP-dependent tumor-suppressing pathway in multiple HCC cell lines. In mice deficient for SMAD3 (Smad3 +/-), PJA1 overexpression promoted the transformation of liver stem cells. Analysis of genes regulated by PJA1 knockdown and TGFβ1 signaling revealed 1,584 co-upregulated genes and 1,280 co-downregulated genes, including many implicated in cancer. The E3 ligase inhibitor RTA405 enhanced SMAD3-regulated gene expression and reduced growth of HCC cells in culture and xenografts of HCC tumors, suggesting that inhibition of PJA1 may be beneficial in treating HCC or preventing HCC development in at-risk patients.Significance: These findings provide a novel mechanism regulating the tumor suppressor function of TGFβ in liver carcinogenesis.
Collapse
Affiliation(s)
- Jian Chen
- Department of Gastroenterology, Hepatology, and Nutrition, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| | - Abhisek Mitra
- Pfizer Inc. Integrative Biotechnology Group, Pearl River, New York
| | - Shulin Li
- Department of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Shumei Song
- Department of GI Medical Oncology-Research, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Bao-Ngoc Nguyen
- Center for Translational Medicine, Department of Surgery, George Washington University, Washington, D.C
| | - Jiun-Sheng Chen
- Department of Gastroenterology, Hepatology, and Nutrition, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Ji-Hyun Shin
- Department of Gastroenterology, Hepatology, and Nutrition, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Nancy R Gough
- Center for Translational Medicine, Department of Surgery, George Washington University, Washington, D.C.,BioSerendipity, LLC, Elkridge, Maryland
| | - Paul Lin
- Department of Surgery, George Washington University, Washington, D.C
| | - Vincent Obias
- Department of Surgery, George Washington University, Washington, D.C
| | - Aiwu Ruth He
- Department of Medicine and Oncology, Georgetown University, Lombardi Comprehensive Cancer Center Washington, D.C
| | - Zhixing Yao
- Department of Biochemistry and Molecular Biology, Howard University, Washington, D.C
| | - Tathiane M Malta
- Department of Neurosurgery, Henry Ford Hospital, Detroit, Michigan
| | - Houtan Noushmehr
- Department of Neurosurgery, Henry Ford Hospital, Detroit, Michigan
| | - Patricia S Latham
- Department of Pathology, George Washington University, Washington, D.C
| | - Xiaoping Su
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Asif Rashid
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Bibhuti Mishra
- Center for Translational Medicine, Department of Surgery, George Washington University, Washington, D.C
| | - Ray-Chang Wu
- Department of Biochemistry & Molecular Biology, George Washington University, Washington, D.C
| | - Lopa Mishra
- Center for Translational Medicine, Department of Surgery, George Washington University, Washington, D.C. .,Gastroenterology, Hepatology, and Nutrition Section, VA Medical Center, Washington, D.C
| |
Collapse
|
15
|
Hyslip J, Martins PN. Liver Repair and Regeneration in Transplant: State of the Art. CURRENT TRANSPLANTATION REPORTS 2020. [DOI: 10.1007/s40472-020-00269-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
|
16
|
Bangru S, Kalsotra A. Cellular and molecular basis of liver regeneration. Semin Cell Dev Biol 2020; 100:74-87. [PMID: 31980376 DOI: 10.1016/j.semcdb.2019.12.004] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2019] [Revised: 11/29/2019] [Accepted: 12/03/2019] [Indexed: 12/13/2022]
Abstract
Recent advances in genetics and genomics have reinvigorated the field of liver regeneration. It is now possible to combine lineage-tracing with genome-wide studies to genetically mark individual liver cells and their progenies and detect precise changes in their genome, transcriptome, and proteome under normal versus regenerative settings. The recent use of single-cell RNA sequencing methodologies in model organisms has, in some ways, transformed our understanding of the cellular and molecular biology of liver regeneration. Here, we review the latest strides in our knowledge of general principles that coordinate regeneration of the liver and reflect on some conflicting evidence and controversies surrounding this topic. We consider the prominent mechanisms that stimulate homeostasis-related vis-à-vis injury-driven regenerative responses, highlight the likely cellular sources/depots that reconstitute the liver following various injuries and discuss the extrinsic and intrinsic signals that direct liver cells to proliferate, de-differentiate, or trans-differentiate while the tissue recovers from acute or chronic damage.
Collapse
Affiliation(s)
- Sushant Bangru
- Departments of Biochemistry and Pathology, University of Illinois, Urbana-Champaign, IL, USA; Cancer Center@ Illinois, University of Illinois, Urbana-Champaign, IL, USA
| | - Auinash Kalsotra
- Departments of Biochemistry and Pathology, University of Illinois, Urbana-Champaign, IL, USA; Cancer Center@ Illinois, University of Illinois, Urbana-Champaign, IL, USA; Carl R. Woese Institute for Genomic Biology, University of Illinois, Urbana-Champaign, IL, USA.
| |
Collapse
|
17
|
Mohseni R, Arab Sadeghabadi Z, Karimi J, Gholami H, Ghasemi H, Ghadimipour HR, Kheiripour N. Chlorella vulgaris supplementation attenuates the progression of liver fibrosis through targeting TGF-β-signaling pathway in the CCl4-induced liver fibrosis in rats. TOXIN REV 2019. [DOI: 10.1080/15569543.2019.1700525] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Affiliation(s)
- Roohollah Mohseni
- Student research committee, Hamadan University of Medical Sciences, Hamadan, Iran
- Clinical Biochemistry Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Zahra Arab Sadeghabadi
- Department of Clinical Biochemistry, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Jamshid Karimi
- Department of Clinical Biochemistry, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Hamid Gholami
- Department of Clinical Biochemistry, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Hadi Ghasemi
- Department of Clinical Biochemistry, Hamadan University of Medical Sciences, Hamadan, Iran
| | | | - Nejat Kheiripour
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Kashan University of Medical Sciences, Kashan, Iran
| |
Collapse
|
18
|
Chen Z, Wan L, Jin X, Wang W, Li D. Transforming growth factor-β signaling confers hepatic stellate cells progenitor features after partial hepatectomy. J Cell Physiol 2019; 235:2655-2667. [PMID: 31584200 DOI: 10.1002/jcp.29169] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Accepted: 08/26/2019] [Indexed: 12/29/2022]
Abstract
Liver regeneration involves not only hepatocyte replication but progenitor aggregation and scarring. Partial hepatectomy (PH), an established model for liver regeneration, reactivates transforming growth factor-β (TGF-β) signaling. Hepatic stellate cells (HSCs) are primarily responding cells for TGF-β and resident in stem cell niche. In the current study, PH mice were treated with SB-431542, an inhibitor of TGF-β Type I receptor, aiming to address the role of TGF-β signaling on the fate determination of HSCs during liver regeneration. After PH, control mice exhibited HSCs activation, progenitor cells accumulation, and a fraction of HSCs acquired the phenotype of hepatocyte or cholangiocyte. Blocking TGF-β signaling delayed proliferation, impaired progenitor response, and scarring repair. In SB-431542 group, merely no HSCs were found coexpressed progenitor makers, such as SOX9 and AFP. Inhibition of TGF-β pathway disturbed the epithelial-mesenchymal transitions and diminished the nuclear accumulation of β-catenin as well as the expression of cytochrome P450 2E1 in HSC during liver regeneration. We identify a key role of TGF-β signaling on promoting HSC transition, which subsequently becomes progenitor for generating liver epithelial cells after PH. This process might interact with an acknowledged stem cell function signaling, Wnt/β-catenin.
Collapse
Affiliation(s)
- Zixin Chen
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Li Wan
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xin Jin
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Wei Wang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Dewei Li
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
19
|
Comparative study on protective effect of hydrogen rich saline and adipose-derived stem cells on hepatic ischemia-reperfusion and hepatectomy injury in swine. Biomed Pharmacother 2019; 120:109453. [PMID: 31561069 DOI: 10.1016/j.biopha.2019.109453] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Revised: 09/08/2019] [Accepted: 09/09/2019] [Indexed: 01/25/2023] Open
Abstract
AIM To compare and evaluate the hepatoprotective effect of liver parenchyma injection of ADSCs and portal vein injection of HRS in laparoscopic hepatic ischemia reperfusion combined with hepatectomy injury in miniature pigs. METHODS Eighteen miniature pigs were randomly assigned to IRI group, HRS group and ADSCs group. HRS was injected through the portal vein 10 min before reperfusion, 1 d, 2 d, and 3 d after surgery. ADSCs were injected into liver parenchyma after hepatectomy. The serum and liver tissue samples were collected at different time points (preoperative, and postoperative at 1 d, 3 d and 7 d). RESULTS Compared with the IRI group, both ADSCs and HRS groups can promote liver function recovery, reduce oxidative stress, reduce inflammation, and promote liver regeneration. Compared with HRS, ALT and TBIL in ADSCs group were significantly decreased at 3 d, and AST was significantly reduced at 1 d. The activities of SOD and GSH-Px in ADSCs group were significantly higher than that in HRS group, but the MDA level in HRS group was markedly lower than that in ADSCs group at 1 d. IL-1β was significantly lower in the ADSCs group than in the HRS group at 1 day after operation. The expressions of HGF and PCNA were significantly higher than that in the HRS group at 3 day after surgery. CONCLUSION Our study has demonstrated that HRS and ADSCs have significant hepatoprotective effects in miniature pigs after HIRI and hepatectomy injury. However, liver parenchyma injection of ADSCs is more beneficial to the recovery of liver function than portal vein injection of HRS.
Collapse
|
20
|
Meta-Analysis of Human and Mouse Biliary Epithelial Cell Gene Profiles. Cells 2019; 8:cells8101117. [PMID: 31547151 PMCID: PMC6829476 DOI: 10.3390/cells8101117] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Revised: 09/03/2019] [Accepted: 09/18/2019] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Chronic liver diseases are frequently accompanied with activation of biliary epithelial cells (BECs) that can differentiate into hepatocytes and cholangiocytes, providing an endogenous back-up system. Functional studies on BECs often rely on isolations of an BEC cell population from healthy and/or injured livers. However, a consensus on the characterization of these cells has not yet been reached. The aim of this study was to compare the publicly available transcriptome profiles of human and mouse BECs and to establish gene signatures that can identify quiescent and activated human and mouse BECs. METHODS We used publicly available transcriptome data sets of human and mouse BECs, compared their profiles and analyzed co-expressed genes and pathways. By merging both human and mouse BEC-enriched genes, we obtained a quiescent and activation gene signature and tested them on BEC-like cells and different liver diseases using gene set enrichment analysis. In addition, we identified several genes from both gene signatures to identify BECs in a scRNA sequencing data set. RESULTS Comparison of mouse BEC transcriptome data sets showed that the isolation method and array platform strongly influences their general profile, still most populations are highly enriched in most genes currently associated with BECs. Pathway analysis on human and mouse BECs revealed the KRAS signaling as a new potential pathway in BEC activation. We established a quiescent and activated BEC gene signature that can be used to identify BEC-like cells and detect BEC enrichment in alcoholic hepatitis, non-alcoholic steatohepatitis (NASH) and peribiliary sclerotic livers. Finally, we identified a gene set that can distinguish BECs from other liver cells in mouse and human scRNAseq data. CONCLUSIONS Through a meta-analysis of human and mouse BEC gene profiles we identified new potential pathways in BEC activation and created unique gene signatures for quiescent and activated BECs. These signatures and pathways will help in the further characterization of this progenitor cell type in mouse and human liver development and disease.
Collapse
|
21
|
Tascher G, Burban A, Camus S, Plumel M, Chanon S, Le Guevel R, Shevchenko V, Van Dorsselaer A, Lefai E, Guguen-Guillouzo C, Bertile F. In-Depth Proteome Analysis Highlights HepaRG Cells as a Versatile Cell System Surrogate for Primary Human Hepatocytes. Cells 2019; 8:E192. [PMID: 30795634 PMCID: PMC6406872 DOI: 10.3390/cells8020192] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Revised: 02/16/2019] [Accepted: 02/18/2019] [Indexed: 12/12/2022] Open
Abstract
Of the hepatic cell lines developed for in vitro studies of hepatic functions as alternatives to primary human hepatocytes, many have lost major liver-like functions, but not HepaRG cells. The increasing use of the latter worldwide raises the need for establishing the reference functional status of early biobanked HepaRG cells. Using deep proteome and secretome analyses, the levels of master regulators of the hepatic phenotype and of the structural elements ensuring biliary polarity were found to be close to those in primary hepatocytes. HepaRG cells proved to be highly differentiated, with functional mitochondria, hepatokine secretion abilities, and an adequate response to insulin. Among differences between primary human hepatocytes and HepaRG cells, the factors that possibly support HepaRG transdifferentiation properties are discussed. The HepaRG cell system thus appears as a robust surrogate for primary hepatocytes, which is versatile enough to study not only xenobiotic detoxification, but also the control of hepatic energy metabolism, secretory function and disease-related mechanisms.
Collapse
Affiliation(s)
- Georg Tascher
- Laboratoire de Spectrométrie de Masse BioOrganique, CNRS, IPHC UMR 7178, Université de Strasbourg, F-67087 Strasbourg, France.
- Institute of Biochemistry II, Goethe University Hospital, D-60590 Frankfurt am Main, Germany.
| | - Audrey Burban
- INSERM U1241 NuMeCan, Université de Rennes 1, F-35033 Rennes, France.
| | - Sandrine Camus
- Biopredic International, Parc d'Affaires de la Bretêche, F-35760 St Grégoire, France.
| | - Marine Plumel
- Laboratoire de Spectrométrie de Masse BioOrganique, CNRS, IPHC UMR 7178, Université de Strasbourg, F-67087 Strasbourg, France.
| | - Stéphanie Chanon
- CarMeN Laboratory, INSERM, INRA, University of Lyon, F-69310 Pierre-Bénite, France.
| | - Remy Le Guevel
- ImPACcell platform, Biosit, Université de Rennes 1, F-35043 Rennes, France.
| | - Valery Shevchenko
- Biopredic International, Parc d'Affaires de la Bretêche, F-35760 St Grégoire, France.
| | - Alain Van Dorsselaer
- Laboratoire de Spectrométrie de Masse BioOrganique, CNRS, IPHC UMR 7178, Université de Strasbourg, F-67087 Strasbourg, France.
| | - Etienne Lefai
- CarMeN Laboratory, INSERM, INRA, University of Lyon, F-69310 Pierre-Bénite, France.
| | - Christiane Guguen-Guillouzo
- INSERM U1241 NuMeCan, Université de Rennes 1, F-35033 Rennes, France.
- Biopredic International, Parc d'Affaires de la Bretêche, F-35760 St Grégoire, France.
| | - Fabrice Bertile
- Laboratoire de Spectrométrie de Masse BioOrganique, CNRS, IPHC UMR 7178, Université de Strasbourg, F-67087 Strasbourg, France.
| |
Collapse
|
22
|
Ge YS, Zhang QZ, Li H, Bai G, Jiao ZH, Wang HB. Hydrogen-rich saline protects against hepatic injury induced by ischemia-reperfusion and laparoscopic hepatectomy in swine. Hepatobiliary Pancreat Dis Int 2019; 18:48-61. [PMID: 30573299 DOI: 10.1016/j.hbpd.2018.12.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Accepted: 11/28/2018] [Indexed: 02/08/2023]
Abstract
BACKGROUND Hydrogen-rich saline (HRS) has antioxidative, anti-inflammatory and anti-apoptotic properties. We investigated the effects of hydrogen on hepatic ischemia-reperfusion (I/R) and laparoscopic hepatectomy in swine. METHODS Twenty-one healthy Bama miniature pigs were randomly divided into the sham group, ischemia-reperfusion injury (IRI) group, HRS-5 (5 mL/kg) group, and HRS-10 (10 mL/kg) group. HRS was injected through the portal vein 10 min before reperfusion and at postoperative day 1, 2 and 3. The roles of HRS on oxidative stress, inflammatory response and liver regeneration were studied. RESULTS Compared with the IRI group, HRS treatment attenuated oxidative stress by increasing catalase activity and reducing myeloperoxidase. White blood cells in the HRS-10 group were reduced compared with the IRI group (P < 0.01). In the HRS-10 group, interleukin-1 beta, interleukin-6 and tumor necrosis factor alpha, C-reactive protein and cortisol were downregulated, whereas interleukin-10 was upregulated. In addition, HRS attenuated endothelial cell injury and promoted the secretion of angiogenic cytokines, including vascular endothelial growth factor, angiopoietin-1 and angiopoietin-2. HRS elevated the levels of hepatocyte growth factor, Cyclin D1, proliferating cell nuclear antigen, Ki-67 and reduced the secretion of transforming growth factor-beta. CONCLUSIONS HRS treatment may exert a protective effect against I/R and hepatectomy-induced hepatic damage by reducing oxidative stress, suppressing the inflammatory response and promoting liver regeneration.
Collapse
Affiliation(s)
- Yan-Song Ge
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| | - Qian-Zhen Zhang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| | - Hui Li
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| | - Ge Bai
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| | - Zhi-Hui Jiao
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| | - Hong-Bin Wang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China.
| |
Collapse
|
23
|
Gaitantzi H, Meyer C, Rakoczy P, Thomas M, Wahl K, Wandrer F, Bantel H, Alborzinia H, Wölfl S, Ehnert S, Nüssler A, Bergheim I, Ciuclan L, Ebert M, Breitkopf-Heinlein K, Dooley S. Ethanol sensitizes hepatocytes for TGF-β-triggered apoptosis. Cell Death Dis 2018; 9:51. [PMID: 29352207 PMCID: PMC5833779 DOI: 10.1038/s41419-017-0071-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Revised: 09/19/2017] [Accepted: 10/09/2017] [Indexed: 12/14/2022]
Abstract
UNLABELLED Alcohol abuse is a global health problem causing a substantial fraction of chronic liver diseases. Abundant TGF-β-a potent pro-fibrogenic cytokine-leads to disease progression. Our aim was to elucidate the crosstalk of TGF-β and alcohol on hepatocytes. Primary murine hepatocytes were challenged with ethanol and TGF-β and cell fate was determined. Fluidigm RNA analyses revealed transcriptional effects that regulate survival and apoptosis. Mechanistic insights were derived from enzyme/pathway inhibition experiments and modulation of oxidative stress levels. To substantiate findings, animal model specimens and human liver tissue cultures were investigated. RESULTS On its own, ethanol had no effect on hepatocyte apoptosis, whereas TGF-β increased cell death. Combined treatment led to massive hepatocyte apoptosis, which could also be recapitulated in human HCC liver tissue treated ex vivo. Alcohol boosted the TGF-β pro-apoptotic gene signature. The underlying mechanism of pathway crosstalk involves SMAD and non-SMAD/AKT signaling. Blunting CYP2E1 and ADH activities did not prevent this effect, implying that it was not a consequence of alcohol metabolism. In line with this, the ethanol metabolite acetaldehyde did not mimic the effect and glutathione supplementation did not prevent the super-induction of cell death. In contrast, blocking GSK-3β activity, a downstream mediator of AKT signaling, rescued the strong apoptotic response triggered by ethanol and TGF-β. This study provides novel information on the crosstalk between ethanol and TGF-β. We give evidence that ethanol directly leads to a boost of TGF-β's pro-apoptotic function in hepatocytes, which may have implications for patients with chronic alcoholic liver disease.
Collapse
Affiliation(s)
- Haristi Gaitantzi
- Department of Medicine II, Section Molecular Hepatology, Medical Faculty Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany
| | - Christoph Meyer
- Department of Medicine II, Section Molecular Hepatology, Medical Faculty Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany
| | - Pia Rakoczy
- Department of Medicine II, Section Molecular Hepatology, Medical Faculty Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany
- Miltenyi Biotec GmbH, Friedrich-Ebert-Straße 68, 51429, Bergisch Gladbach, Germany
| | - Maria Thomas
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Auerbachstr. 112, 70376, Stuttgart, Germany
| | - Kristin Wahl
- Department of Gastroenterology, Hepatology, and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Franziska Wandrer
- Department of Gastroenterology, Hepatology, and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Heike Bantel
- Department of Gastroenterology, Hepatology, and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Hamed Alborzinia
- Institute of Pharmacy and Molecular Biotechnology, Heidelberg University, Heidelberg, Germany
| | - Stefan Wölfl
- Institute of Pharmacy and Molecular Biotechnology, Heidelberg University, Heidelberg, Germany
| | - Sabrina Ehnert
- Eberhard-Karls University Tübingen, BG Trauma Center, SWI, Schnarrenbergstraße 95, 72076, Tübingen, Germany
| | - Andreas Nüssler
- Eberhard-Karls University Tübingen, BG Trauma Center, SWI, Schnarrenbergstraße 95, 72076, Tübingen, Germany
| | - Ina Bergheim
- University of Vienna, Department of Nutritional Sciences, Molecular Nutritional Science, Althanstr. 14, UZA II, A-1090, Wien, Austria
| | - Loredana Ciuclan
- Roche Products Limited, 6 Falcon Way, Shire Park, Welwyn Garden City, AL7 1TW, UK
| | - Matthias Ebert
- Department of Medicine II, Section Molecular Hepatology, Medical Faculty Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany
| | - Katja Breitkopf-Heinlein
- Department of Medicine II, Section Molecular Hepatology, Medical Faculty Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany
| | - Steven Dooley
- Department of Medicine II, Section Molecular Hepatology, Medical Faculty Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany.
| |
Collapse
|
24
|
Dogra D, Ahuja S, Kim HT, Rasouli SJ, Stainier DYR, Reischauer S. Opposite effects of Activin type 2 receptor ligands on cardiomyocyte proliferation during development and repair. Nat Commun 2017; 8:1902. [PMID: 29196619 PMCID: PMC5711791 DOI: 10.1038/s41467-017-01950-1] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Accepted: 10/27/2017] [Indexed: 01/14/2023] Open
Abstract
Zebrafish regenerate damaged myocardial tissue very effectively. Hence, insights into the molecular networks underlying zebrafish heart regeneration might help develop alternative strategies to restore human cardiac performance. While TGF-β signaling has been implicated in zebrafish cardiac regeneration, the role of its individual ligands remains unclear. Here, we report the opposing expression response during zebrafish heart regeneration of two genes, mstnb and inhbaa, which encode TGF-β family ligands. Using gain-of-function (GOF) and loss-of-function (LOF) approaches, we show that these ligands mediate inverse effects on cardiac regeneration and specifically on cardiomyocyte (CM) proliferation. Notably, we find that Inhbaa functions as a CM mitogen and that its overexpression leads to accelerated cardiac recovery and scar clearance after injury. In contrast, mstnb GOF and inhbaa LOF both lead to unresolved scarring after cardiac injury. We further show that Mstnb and Inhbaa inversely control Smad2 and Smad3 transcription factor activities through alternate Activin type 2 receptors.
Collapse
Affiliation(s)
- Deepika Dogra
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, 61231, Bad Nauheim, Germany
| | - Suchit Ahuja
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, 61231, Bad Nauheim, Germany
| | - Hyun-Taek Kim
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, 61231, Bad Nauheim, Germany
| | - S Javad Rasouli
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, 61231, Bad Nauheim, Germany
| | - Didier Y R Stainier
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, 61231, Bad Nauheim, Germany
- Department of Biochemistry and Biophysics, University of California San Francisco, San Francisco, 94158, CA, USA
| | - Sven Reischauer
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, 61231, Bad Nauheim, Germany.
- Department of Biochemistry and Biophysics, University of California San Francisco, San Francisco, 94158, CA, USA.
| |
Collapse
|
25
|
Li L, Li H, Zhang Z, Zheng J, Shi Y, Liu J, Cao Y, Yuan X, Chu Y. Recombinant truncated TGF‑β receptor II attenuates carbon tetrachloride‑induced epithelial‑mesenchymal transition and liver fibrosis in rats. Mol Med Rep 2017; 17:315-321. [PMID: 29115426 DOI: 10.3892/mmr.2017.7845] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Accepted: 08/03/2017] [Indexed: 11/06/2022] Open
Abstract
Liver fibrosis is a pathological process of chronic liver diseases. In particular, epithelial‑mesenchymal transition (EMT) is a major source of myofibroblast structure in liver fibrosis. The present study investigated the effects of recombinant truncated transforming growth factor‑ß receptor II (rtTGFβRII) on EMT and liver fibrosis in a carbon tetrachloride (CCl4)‑induced rat model. A total of 24 rats were randomly separated into three groups: Normal control (NC), model (CCl4) and treatment (CCl4 + rtTGFβRII) groups. Histological methods, including hematoxylin and eosin, Masson's trichrome and Sirius red staining were conducted. The activities of serum alanine aminotransferase (ALT) and aspartate aminotransferase (AST) were measured using an automatic biochemical analyzer. The mRNA expression levels of fibroblast specific protein‑1 (FSP‑1), α‑smooth muscle actin (α‑SMA), fibronectin, collagen I, vimentin and E‑cadherin were detected using reverse transcription‑quantitative polymerase chain reaction analysis. The protein levels of fibronectin, collagen I, E‑cadherin, Smad2/3 and phosphorylated (p)‑Smad2/3 were detected using western blot analysis. The expression of α‑SMA, fibronectin, vimentin and E‑cadherin in the liver tissue was detected using immunofluorescence staining. The results demonstrated that in vivo, rtTGFβRII significantly reduced the degree of liver injury, serum ALT and AST activities and liver fibrosis. These factors were associated with reduced expression of FSP‑1, α‑SMA, fibronectin, collagen I, vimentin and p‑Smad2/3, and increased expression of E‑cadherin. The results of the present study suggest that rtTGFβRII may inhibit EMT processes in CCl4‑induced liver fibrosis in rats and alter the expression of epithelial and myofibroblast markers. Therefore, rtTGFβRII may be considered a possible treatment for preventing liver fibrosis via EMT processes.
Collapse
Affiliation(s)
- Luxin Li
- Heilongjiang Key Laboratory of Anti‑Fibrosis Biotherapy, Mudanjiang Medical University, Mudanjiang, Heilongjiang 157011, P.R. China
| | - Hongzhi Li
- Heilongjiang Key Laboratory of Anti‑Fibrosis Biotherapy, Mudanjiang Medical University, Mudanjiang, Heilongjiang 157011, P.R. China
| | - Zhen Zhang
- Heilongjiang Key Laboratory of Anti‑Fibrosis Biotherapy, Mudanjiang Medical University, Mudanjiang, Heilongjiang 157011, P.R. China
| | - Junya Zheng
- Heilongjiang Key Laboratory of Anti‑Fibrosis Biotherapy, Mudanjiang Medical University, Mudanjiang, Heilongjiang 157011, P.R. China
| | - Yongping Shi
- Heilongjiang Key Laboratory of Anti‑Fibrosis Biotherapy, Mudanjiang Medical University, Mudanjiang, Heilongjiang 157011, P.R. China
| | - Jieting Liu
- Heilongjiang Key Laboratory of Anti‑Fibrosis Biotherapy, Mudanjiang Medical University, Mudanjiang, Heilongjiang 157011, P.R. China
| | - Yanan Cao
- Heilongjiang Key Laboratory of Anti‑Fibrosis Biotherapy, Mudanjiang Medical University, Mudanjiang, Heilongjiang 157011, P.R. China
| | - Xiaohuan Yuan
- Heilongjiang Key Laboratory of Anti‑Fibrosis Biotherapy, Mudanjiang Medical University, Mudanjiang, Heilongjiang 157011, P.R. China
| | - Yanhui Chu
- Heilongjiang Key Laboratory of Anti‑Fibrosis Biotherapy, Mudanjiang Medical University, Mudanjiang, Heilongjiang 157011, P.R. China
| |
Collapse
|
26
|
Rao S, Zaidi S, Banerjee J, Jogunoori W, Sebastian R, Mishra B, Nguyen BN, Wu RC, White J, Deng C, Amdur R, Li S, Mishra L. Transforming growth factor-β in liver cancer stem cells and regeneration. Hepatol Commun 2017; 1:477-493. [PMID: 29404474 PMCID: PMC5678904 DOI: 10.1002/hep4.1062] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Revised: 04/27/2017] [Accepted: 06/01/2017] [Indexed: 12/11/2022] Open
Abstract
Cancer stem cells have established mechanisms that contribute to tumor heterogeneity as well as resistance to therapy. Over 40% of hepatocellular carcinomas (HCCs) are considered to be clonal and arise from a stem-like/cancer stem cell. Moreover, HCC is the second leading cause of cancer death worldwide, and an improved understanding of cancer stem cells and targeting these in this cancer are urgently needed. Multiple studies have revealed etiological patterns and multiple genes/pathways signifying initiation and progression of HCC; however, unlike the transforming growth factor β (TGF-β) pathway, loss of p53 and/or activation of β-catenin do not spontaneously drive HCC in animal models. Despite many advances in cancer genetics that include identifying the dominant role of TGF-β signaling in gastrointestinal cancers, we have not reached an integrated view of genetic mutations, copy number changes, driver pathways, and animal models that support effective targeted therapies for these common and lethal cancers. Moreover, pathways involved in stem cell transformation into gastrointestinal cancers remain largely undefined. Identifying the key mechanisms and developing models that reflect the human disease can lead to effective new treatment strategies. In this review, we dissect the evidence obtained from mouse and human liver regeneration, and mouse genetics, to provide insight into the role of TGF-β in regulating the cancer stem cell niche. (Hepatology Communications 2017;1:477-493).
Collapse
Affiliation(s)
- Shuyun Rao
- Center for Translational Medicine Department of Surgery, George Washington University Washington DC
| | - Sobia Zaidi
- Center for Translational Medicine Department of Surgery, George Washington University Washington DC
| | - Jaideep Banerjee
- Center for Translational Medicine Department of Surgery, George Washington University Washington DC
| | - Wilma Jogunoori
- Center for Translational Medicine Department of Surgery, George Washington University Washington DC
| | - Raul Sebastian
- Center for Translational Medicine Department of Surgery, George Washington University Washington DC
| | - Bibhuti Mishra
- Center for Translational Medicine Department of Surgery, George Washington University Washington DC.,Institute for Clinical Research, Veterans Affairs Medical Center Washington DC
| | - Bao-Ngoc Nguyen
- Center for Translational Medicine Department of Surgery, George Washington University Washington DC
| | - Ray-Chang Wu
- Department of Biochemistry and Molecular Medicine George Washington University Washington DC
| | - Jon White
- Institute for Clinical Research, Veterans Affairs Medical Center Washington DC
| | - Chuxia Deng
- Center for Translational Medicine Department of Surgery, George Washington University Washington DC.,Health Sciences University of Macau Taipa Macau China
| | - Richard Amdur
- Center for Translational Medicine Department of Surgery, George Washington University Washington DC
| | - Shulin Li
- Department of Pediatrics The University of Texas MD Anderson Cancer Center Houston TX
| | - Lopa Mishra
- Center for Translational Medicine Department of Surgery, George Washington University Washington DC.,Institute for Clinical Research, Veterans Affairs Medical Center Washington DC
| |
Collapse
|
27
|
Zhang QZ, Liu YL, Wang YR, Fu LN, Zhang J, Wang XR, Wang BM. Effects of telmisartan on improving leptin resistance and inhibiting hepatic fibrosis in rats with non-alcoholic fatty liver disease. Exp Ther Med 2017; 14:2689-2694. [PMID: 28962213 DOI: 10.3892/etm.2017.4809] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2016] [Accepted: 04/21/2017] [Indexed: 02/06/2023] Open
Abstract
The present study aimed to investigate the impacts of telmisartan (TEL) on hepatic fibrosis, serum leptin, leptin protein in liver tissue and its mRNA expression level in rats with non-alcoholic fatty liver disease (NAFLD). Male Sprague Dawley rats were randomly divided into the control (N), model (M), polyene phosphatidylcholine (P) and TEL (T) groups. Group M and the intervention groups were given a high-fat diet for 12 weeks to induce NAFLD, followed by 4 weeks of intragastric administration of normal saline (1.0 ml/kg/day), polyene phosphatidylcholine (PPC; 123.1 mg/kg/day) and TEL (8 mg/kg/day). The liver tissue was then assessed for the NAFLD activity score and fibrosis score (FS), and serum biochemistry and leptin levels were determined. Additionally, leptin protein expression levels were examined by western blotting and the expression of leptin mRNA was investigated by reverse transcription-polymerase chain reaction. TEL significantly improved FS in rats (P<0.01) and was more effective than PPC. TEL significantly reduced the expression of serum leptin, as well as the expression levels of leptin protein and its mRNA in liver tissue (P<0.01); however, the effects of PPC were not significant (P>0.05). TEL reduced serum leptin, leptin protein and its mRNA in the liver tissue of NAFLD rats, and improved the pathological indicators of liver fibrosis.
Collapse
Affiliation(s)
- Qiu-Zan Zhang
- Department of Gastroenterology, Tianjin Medical University, The Fourth Central Clinical College, Tianjin 300140, P.R. China
| | - Ying-Li Liu
- Department of Gastroenterology, Tianjin Medical University, The Fourth Central Clinical College, Tianjin 300140, P.R. China
| | - Yan-Rong Wang
- Department of Gastroenterology, Tianjin Medical University, The Fourth Central Clinical College, Tianjin 300140, P.R. China
| | - Li-Na Fu
- Department of Gastroenterology, Tianjin Medical University, The Fourth Central Clinical College, Tianjin 300140, P.R. China
| | - Jing Zhang
- Department of Gastroenterology, Tianjin Medical University, The Fourth Central Clinical College, Tianjin 300140, P.R. China
| | - Xiu-Ru Wang
- Department of Gastroenterology, Tianjin Medical University, The Fourth Central Clinical College, Tianjin 300140, P.R. China
| | - Bang-Mao Wang
- Department of Gastroenterology, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| |
Collapse
|
28
|
Mitra A, Yan J, Xia X, Zhou S, Chen J, Mishra L, Li S. IL6-mediated inflammatory loop reprograms normal to epithelial-mesenchymal transition + metastatic cancer stem cells in preneoplastic liver of transforming growth factor beta-deficient β2-spectrin +/- mice. Hepatology 2017; 65:1222-1236. [PMID: 27863449 PMCID: PMC5360560 DOI: 10.1002/hep.28951] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2016] [Revised: 11/04/2016] [Accepted: 11/09/2016] [Indexed: 12/26/2022]
Abstract
UNLABELLED Hepatocellular carcinoma (HCC) is the second-leading cause of cancer-related deaths worldwide with a poor survival rate. As many as 40% of HCCs are clonal, with alteration of key tumor-suppressor pathways in stem cells as the primary cause of HCC initiation. However, mechanisms that generate metastatic stem cells in preneoplastic liver tissue are not well understood. We hypothesized that chronic inflammation is a major driver of the transformation of genetically defective liver stem cells (LSCs) into highly metastatic liver cancer cells in premalignant liver tissue. We developed models of chronic inflammation in wild-type (WT) and β2-spectrin (β2SP)+/- (SPTBN1) mice. CD133+ LSCs derived from preneoplastic livers of β2SP+/- mice treated with interleukin-6 (pIL6; IL6 β2SP+/- LSCs) were highly tumorigenic and metastatic, whereas those derived from WT mice treated with pIL6 (IL6 WT LSCs) had significantly less proliferation and no tumorigenic properties. IL6 β2SP+/- LSCs not only exhibited nuclear localization of Twist and Slug, markers of epithelial-mesenchymal transition (EMT), but also constitutive activation of nuclear factor kappa B (NFκB; RELA). Knockdown of NFκB decreased the EMT phenotypes and metastatic capacity of these cells. NFκB in IL6 β2SP+/- LSCs was activated by transforming growth factor β (TGFβ)-activated kinase 1 (TAK1; MAP3K7), which is associated with poor survival in HCC and interleukin-6 (IL6) expression. The amount of constitutively activated NFκB increased dramatically from normal to cirrhotic to HCC tissues from human patients. CONCLUSION IL6-mediated inflammation programs constitutive activation of the TAK1-NFκB signaling cascade in CD133+ LSCs, and this program interacts with deficient TGFβ signaling, thereby accelerating the transformation of normal LSCs to metastatic cancer stem cells (mCSCs). Indeed, this study delineates the development of EMT-positive mCSCs in HCC-free liver tissue upon chronic inflammation. (Hepatology 2017;65:1222-1236).
Collapse
Affiliation(s)
- Abhisek Mitra
- Department of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jun Yan
- Department of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Xueqing Xia
- Department of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Shouhao Zhou
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jian Chen
- Department of Gastroenterology, Hepatology and Nutrition, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Lopa Mishra
- Department of Surgery and Katzen Research Cancer Center, George Washington University, Washington, D.C. 20052, USA,Co-corresponding author: Lopa Mishra, M.D., Director, The Center for Translational Research, Department of Surgery and Katzen Research Cancer CenterGeorge Washington University and VA Medical Center2150 Pennsylvania Avenue, NW, Suite 1-200, Washington, D.C. USA, , Phone: 202-745-8000, Fax: 202-462-2006
| | - Shulin Li
- Department of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA,Corresponding author: Shulin Li, PhD, Department of Pediatrics, Unit 0853, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX 77030 USA, , Phone: 713-563-9608, Fax: 713-563-9607
| |
Collapse
|
29
|
Mesenchymal Stem Cells Transplantation following Partial Hepatectomy: A New Concept to Promote Liver Regeneration-Systematic Review of the Literature Focused on Experimental Studies in Rodent Models. Stem Cells Int 2017; 2017:7567958. [PMID: 28386285 PMCID: PMC5366767 DOI: 10.1155/2017/7567958] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2016] [Revised: 02/09/2017] [Accepted: 02/14/2017] [Indexed: 02/08/2023] Open
Abstract
Mesenchymal stem cells (MSCs) are an attractive source for regenerative medicine because they are easily accessible through minimally invasive methods and have the potential to enhance liver regeneration (LG) and improve liver function, following partial hepatectomy (PH) and acute or chronic liver injury. A systematic review of the literature was conducted for articles published up to September 1st, 2016, using the MEDLINE database. The keywords that were used in various combinations were as follows: “Mesenchymal stem cells”, “transplantation”, “stem cells”, “adipose tissue derived stem cells”, “bone marrow-derived stem cells”, “partial hepatectomy”, “acute liver failure”, “chronic liver failure”, “liver fibrosis”, “liver cirrhosis”, “rats”, “mice”, and “liver regeneration”. All introduced keywords were searched for separately in MeSH Database to control relevance and terminological accuracy and validity. A total of 41 articles were identified for potential inclusion and reviewed in detail. After a strict selection process, a total of 28 articles were excluded, leaving 13 articles to form the basis of this systematic review. MSCs transplantation promoted LG and improved liver function. Furthermore, MSCs had the ability to differentiate in hepatocyte-like cells, increase survival, and protect hepatocytes by paracrine mechanisms. MSCs transplantation may provide beneficial effects in the process of LG after PH and acute or chronic liver injury. They may represent a new therapeutic option to treat posthepatectomy acute liver failure.
Collapse
|
30
|
Lukacs-Kornek V, Lammert F. The progenitor cell dilemma: Cellular and functional heterogeneity in assistance or escalation of liver injury. J Hepatol 2017; 66:619-630. [PMID: 27826058 DOI: 10.1016/j.jhep.2016.10.033] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Revised: 10/18/2016] [Accepted: 10/31/2016] [Indexed: 12/16/2022]
Abstract
Liver progenitor cells (LPCs) are quiescent cells that are activated during liver injury and thought to give rise to hepatocytes and cholangiocytes in order to support liver regeneration and tissue restitution. While hepatocytes are capable of self-renewal, during most chronic injuries the proliferative capacity of hepatocytes is inhibited, thus LPCs provide main source for regeneration. Despite extensive lineage tracing studies, their role and involvement in these processes are often controversial. Additionally, increasing evidence suggests that the LPC compartment consists of heterogeneous cell populations that are actively involved in cellular interactions with myeloid and lymphoid cells during regeneration. On the other hand, LPC expansion has been associated with an increased fibrogenic response, raising concerns about the therapeutic use of these cells. This review aims to summarize the current understanding of the identity, the cellular interactions and the key pathways affecting the biology of LPCs. Understanding the regulatory circuits and the specific role of LPCs is especially important as it could provide novel therapeutic platforms for the treatment of liver inflammation, fibrosis and regeneration.
Collapse
Affiliation(s)
- Veronika Lukacs-Kornek
- Department of Medicine II, Saarland University Medical Center, Saarland University, Homburg, Germany.
| | - Frank Lammert
- Department of Medicine II, Saarland University Medical Center, Saarland University, Homburg, Germany
| |
Collapse
|
31
|
Fukuda T, Takayama K, Hirata M, Liu YJ, Yanagihara K, Suga M, Mizuguchi H, Furue MK. Isolation and expansion of human pluripotent stem cell-derived hepatic progenitor cells by growth factor defined serum-free culture conditions. Exp Cell Res 2017; 352:333-345. [PMID: 28215634 DOI: 10.1016/j.yexcr.2017.02.022] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Revised: 02/14/2017] [Accepted: 02/15/2017] [Indexed: 12/30/2022]
Abstract
Limited growth potential, narrow ranges of sources, and difference in variability and functions from batch to batch of primary hepatocytes cause a problem for predicting drug-induced hepatotoxicity during drug development. Human pluripotent stem cell (hPSC)-derived hepatocyte-like cells in vitro are expected as a tool for predicting drug-induced hepatotoxicity. Several studies have already reported efficient methods for differentiating hPSCs into hepatocyte-like cells, however its differentiation process is time-consuming, labor-intensive, cost-intensive, and unstable. In order to solve this problem, expansion culture for hPSC-derived hepatic progenitor cells, including hepatic stem cells and hepatoblasts which can self-renewal and differentiate into hepatocytes should be valuable as a source of hepatocytes. However, the mechanisms of the expansion of hPSC-derived hepatic progenitor cells are not yet fully understood. In this study, to isolate hPSC-derived hepatic progenitor cells, we tried to develop serum-free growth factor defined culture conditions using defined components. Our culture conditions were able to isolate and grow hPSC-derived hepatic progenitor cells which could differentiate into hepatocyte-like cells through hepatoblast-like cells. We have confirmed that the hepatocyte-like cells prepared by our methods were able to increase gene expression of cytochrome P450 enzymes upon encountering rifampicin, phenobarbital, or omeprazole. The isolation and expansion of hPSC-derived hepatic progenitor cells in defined culture conditions should have advantages in terms of detecting accurate effects of exogenous factors on hepatic lineage differentiation, understanding mechanisms underlying self-renewal ability of hepatic progenitor cells, and stably supplying functional hepatic cells.
Collapse
Affiliation(s)
- Takayuki Fukuda
- Laboratory of Stem Cell Cultures, National Institutes of Biomedical Innovation, Health and Nutrition, 7-6-8, Saito-Asagi, Ibaraki, Osaka 567-0085, Japan
| | - Kazuo Takayama
- Laboratory of Biochemistry and Molecular Biology, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka 565-0871, Japan; Laboratory of Hepatocyte Regulation, National Institutes of Biomedical Innovation, Health and Nutrition, 7-6-8, Saito-Asagi, Ibaraki, Osaka 567-0085, Japan; PRESTO, Japan Science and Technology Agency, 4-1-8 Honcho, Kawaguchi, Saitama 332-0012, Japan; K-CONNEX, Kyoto University, Yoshida Honmachi, Sakyo-ku, Kyoto 606-8501, Japan
| | - Mitsuhi Hirata
- Laboratory of Stem Cell Cultures, National Institutes of Biomedical Innovation, Health and Nutrition, 7-6-8, Saito-Asagi, Ibaraki, Osaka 567-0085, Japan
| | - Yu-Jung Liu
- Laboratory of Stem Cell Cultures, National Institutes of Biomedical Innovation, Health and Nutrition, 7-6-8, Saito-Asagi, Ibaraki, Osaka 567-0085, Japan
| | - Kana Yanagihara
- Laboratory of Stem Cell Cultures, National Institutes of Biomedical Innovation, Health and Nutrition, 7-6-8, Saito-Asagi, Ibaraki, Osaka 567-0085, Japan
| | - Mika Suga
- Laboratory of Stem Cell Cultures, National Institutes of Biomedical Innovation, Health and Nutrition, 7-6-8, Saito-Asagi, Ibaraki, Osaka 567-0085, Japan
| | - Hiroyuki Mizuguchi
- Laboratory of Biochemistry and Molecular Biology, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka 565-0871, Japan; Laboratory of Hepatocyte Regulation, National Institutes of Biomedical Innovation, Health and Nutrition, 7-6-8, Saito-Asagi, Ibaraki, Osaka 567-0085, Japan; iPS Cell-based Research Project on Hepatic Toxicity and Metabolism, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka 565-0871, Japan; Global Center for Medical Engineering and Informatics, Osaka University, 1-6 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Miho K Furue
- Laboratory of Stem Cell Cultures, National Institutes of Biomedical Innovation, Health and Nutrition, 7-6-8, Saito-Asagi, Ibaraki, Osaka 567-0085, Japan.
| |
Collapse
|
32
|
Müller M, Wetzel S, Köhn-Gaone J, Chalupsky K, Lüllmann-Rauch R, Barikbin R, Bergmann J, Wöhner B, Zbodakova O, Leuschner I, Martin G, Tiegs G, Rose-John S, Sedlacek R, Tirnitz-Parker JEE, Saftig P, Schmidt-Arras D. A disintegrin and metalloprotease 10 (ADAM10) is a central regulator of murine liver tissue homeostasis. Oncotarget 2017; 7:17431-41. [PMID: 26942887 PMCID: PMC4951223 DOI: 10.18632/oncotarget.7836] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Accepted: 02/17/2016] [Indexed: 01/28/2023] Open
Abstract
UNLABELLED A Disintegrin And Metalloprotease (ADAM) 10 exerts essential roles during organ development and tissue integrity in different organs, mainly through activation of the Notch pathway. However, only little is known about its implication in liver tissue physiology. Here we show that in contrast to its role in other tissues, ADAM10 is dispensable for the Notch2-dependent biliary tree formation. However, we demonstrate that expression of bile acid transporters is dependent on ADAM10. Consequently, mice deficient for Adam10 in hepatocytes, cholangiocytes and liver progenitor cells develop spontaneous hepatocyte necrosis and concomitant liver fibrosis. We furthermore observed a strongly augmented ductular reaction in 15-week old ADAM10(Δhep/Δch) mice and demonstrate that c-Met dependent liver progenitor cell activation is enhanced. Additionally, liver progenitor cells are primed to hepatocyte differentiation in the absence of ADAM10. These findings show that ADAM10 is a novel central node controlling liver tissue homeostasis. HIGHLIGHTS Loss of ADAM10 in murine liver results in hepatocyte necrosis and concomitant liver fibrosis. ADAM10 directly regulates expression of bile acid transporters but is dispensable for Notch2-dependent formation of the biliary system. Activation of liver progenitor cells is enhanced through increased c-Met signalling, in the absence of ADAM10. Differentiation of liver progenitor cells to hepatocytes is augmented in the absence of ADAM10.
Collapse
Affiliation(s)
- Miryam Müller
- Institute of Biochemistry, Christian-Albrechts-University, Kiel, Germany
| | - Sebastian Wetzel
- Institute of Biochemistry, Christian-Albrechts-University, Kiel, Germany
| | - Julia Köhn-Gaone
- School of Biomedical Sciences, Curtin Health Innovation Research Institute, Faculty of Health Sciences, Curtin University, Bentley, Australia
| | - Karel Chalupsky
- Laboratory of Transgenic Models of Diseases, Institute of Molecular Genetics of the ASCR, Prague, Czech Republic
| | | | - Roja Barikbin
- Institute of Experimental Immunology and Hepatology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Juri Bergmann
- Institute of Biochemistry, Christian-Albrechts-University, Kiel, Germany.,Department of General and Thoracic Surgery, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Birte Wöhner
- Institute of Biochemistry, Christian-Albrechts-University, Kiel, Germany
| | - Olga Zbodakova
- Laboratory of Transgenic Models of Diseases, Institute of Molecular Genetics of the ASCR, Prague, Czech Republic
| | - Ivo Leuschner
- Institute of Pathology, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Gregor Martin
- Laboratory of Transgenic Models of Diseases, Institute of Molecular Genetics of the ASCR, Prague, Czech Republic.,Laboratory of Integrative Biology, Institute of Molecular Genetics of the ASCR, Prague, Czech Republic
| | - Gisa Tiegs
- Institute of Experimental Immunology and Hepatology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Stefan Rose-John
- Institute of Biochemistry, Christian-Albrechts-University, Kiel, Germany
| | - Radislav Sedlacek
- Laboratory of Transgenic Models of Diseases, Institute of Molecular Genetics of the ASCR, Prague, Czech Republic
| | - Janina E E Tirnitz-Parker
- School of Biomedical Sciences, Curtin Health Innovation Research Institute, Faculty of Health Sciences, Curtin University, Bentley, Australia.,School of Medicine and Pharmacology, University of Western Australia, Fremantle, Australia
| | - Paul Saftig
- Institute of Biochemistry, Christian-Albrechts-University, Kiel, Germany
| | - Dirk Schmidt-Arras
- Institute of Biochemistry, Christian-Albrechts-University, Kiel, Germany
| |
Collapse
|
33
|
Transforming growth factor-β-induced plasticity causes a migratory stemness phenotype in hepatocellular carcinoma. Cancer Lett 2017; 392:39-50. [PMID: 28161507 DOI: 10.1016/j.canlet.2017.01.037] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2016] [Revised: 01/24/2017] [Accepted: 01/25/2017] [Indexed: 02/07/2023]
Abstract
As part of its potential pro-tumorigenic actions, Transforming Growth Factor-(TGF)-β induces epithelial-mesenchymal transition (EMT) in hepatocellular carcinoma (HCC) cells. Whether EMT induces changes in tumor cell plasticity has not been fully explored yet. Here, we analyze the effects of TGF-β on the EMT and stem-related properties of HCC cells and the potential correlation among those processes. The translational aim of the study was to propose a TGF-β/EMT/stem gene signature that would help in recognizing HCC patients as good candidates for anti-TGF-β therapy. Results indicate that when TGF-β induces EMT in HCC cells, a switch in the expression of stem genes is observed and their stemness potential and migratory/invasive capacity are enhanced. However, TGF-β may induce a partial EMT in some epithelial HCC cells, increasing the expression of mesenchymal genes and CD44, but maintaining epithelial gene expression. Epithelial cells show higher stemness potential than the mesenchymal ones, but respond to TGF-β increasing their migratory and invasive capacity. In HCC patient samples, TGFB1 expression most frequently correlates with a partial EMT, increase in mesenchymal genes and CD44 expression, as well as maintenance or over-expression of epithelial-related genes.
Collapse
|
34
|
2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD) increases necroinflammation and hepatic stellate cell activation but does not exacerbate experimental liver fibrosis in mice. Toxicol Appl Pharmacol 2016; 311:42-51. [PMID: 27693115 DOI: 10.1016/j.taap.2016.09.025] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2016] [Revised: 09/21/2016] [Accepted: 09/26/2016] [Indexed: 12/12/2022]
Abstract
2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD) is a persistent environmental contaminant and high-affinity ligand for the aryl hydrocarbon receptor (AhR). Increasing evidence indicates that AhR signaling contributes to wound healing, which involves the coordinated deposition and remodeling of the extracellular matrix. In the liver, wound healing is attributed to the activation of hepatic stellate cells (HSCs), which mediate fibrogenesis through the production of soluble mediators and collagen type I. We recently reported that TCDD treatment increases the activation of human HSCs in vitro. The goal of this study was to determine how TCDD impacts HSC activation in vivo using a mouse model of experimental liver fibrosis. To elicit fibrosis, C57BL6/male mice were treated twice weekly for 8weeks with 0.5ml/kg carbon tetrachloride (CCl4). TCDD (20μg/kg) or peanut oil (vehicle) was administered once a week during the last 2weeks. Results indicate that TCDD increased liver-body-weight ratios, serum alanine aminotransferase activity, and hepatic necroinflammation in CCl4-treated mice. Likewise, TCDD treatment increased mRNA expression of HSC activation and fibrogenesis genes, namely α-smooth muscle actin, desmin, delta-like homolog-1, TGF-β1, and collagen type I. However, TCDD treatment did not exacerbate fibrosis, nor did it increase the collagen content of the liver. Instead, TCDD increased hepatic collagenase activity and increased expression of matrix metalloproteinase (MMP)-13 and the matrix regulatory proteins, TIMP-1 and PAI-1. These results support the conclusion that TCDD increases CCl4-induced liver damage and exacerbates HSC activation, yet collagen deposition and the development of fibrosis may be limited by TCDD-mediated changes in extracellular matrix remodeling.
Collapse
|
35
|
Dynamic Change of Total Bilirubin after Portal Vein Embolization is Predictive of Major Complications and Posthepatectomy Mortality in Patients with Hilar Cholangiocarcinoma. J Gastrointest Surg 2016; 20:960-9. [PMID: 26831059 DOI: 10.1007/s11605-016-3086-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2015] [Accepted: 01/14/2016] [Indexed: 02/07/2023]
Abstract
OBJECTIVES This study aims to evaluate the role of dynamic change in total bilirubin after portal vein embolization (PVE) in predicting major complications and 30-day mortality in patients with hilar cholangiocarcinoma (HCCA). METHODS Retrospective analysis of prospectively maintained data of 64 HCCA patients who underwent PVE before hepatectomy in our institution was used. Total bilirubin and other parameters were measured daily in peri-PVE period. The difference between them and the baseline value from days 0-5 to day -1 (∆D1) and days 5-14 to day -1 (∆D2) were calculated. The relationship between ∆D1 and ∆D2 of total bilirubin and major complications as well as 30-day mortality was analyzed. RESULTS Out of 64 patients, 10 developed major complications (15.6 %) and 6 patients (9.3 %) had died within 30 days after surgery. The ∆D2 of total bilirubin after PVE was most significantly associated with major complications (P < 0.001) and 30-day mortality (P = 0.002). In addition, it was found to be an independent predictor of major complications after PVE (odds ratio (OR) = 1.050; 95 % CI 1.017-1.084). ASA >3 (OR = 12.048; 95 % CI 1.019-143.321), ∆D2 of total bilirubin (OR = 1.058; 95 % CI 1.007-1.112), and ∆D2 of prealbumin (OR = 0.975; 95 % CI 0.952-0.999) were associated with higher risk of 30-day mortality after PVE. Receiver operating characteristic curves showed that ∆D2 of total bilirubin were better predictors than ∆D1 for major complications (AUC (∆D2) 0.817; P = 0.002 vs. AUC (∆D1) 0.769; P = 0.007) and 30-day mortality (ACU(∆D2) 0.868; P = 0.003 vs. AUC(∆D1) 0.721;P = 0.076). CONCLUSION Patients with increased total bilirubin in 5-14 days after PVE may indicate a higher risk of major complications and 30-day mortality if the major hepatectomy were performed.
Collapse
|
36
|
Shang H, Wang Z, Song Y. Liver progenitor cells-mediated liver regeneration in liver cirrhosis. Hepatol Int 2016; 10:440-447. [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] [MESH Headings] [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
Affiliation(s)
- Haitao Shang
- Department of Gastroenterology, Binzhou Medical University Hospital, Binzhou, 256603, People's Republic of China
| | - Zhijun Wang
- Department of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People's Republic of China
| | - Yuhu Song
- Department of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People's Republic of China.
| |
Collapse
|
37
|
Chuah C, Jones MK, McManus DP, Nawaratna SK, Burke ML, Owen HC, Ramm GA, Gobert GN. Characterising granuloma regression and liver recovery in a murine model of schistosomiasis japonica. Int J Parasitol 2016; 46:239-52. [PMID: 26812024 DOI: 10.1016/j.ijpara.2015.12.004] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2015] [Revised: 11/30/2015] [Accepted: 12/07/2015] [Indexed: 02/07/2023]
Abstract
For hepatic schistosomiasis the egg-induced granulomatous response and the development of extensive fibrosis are the main pathologies. We used a Schistosoma japonicum-infected mouse model to characterise the multi-cellular pathways associated with the recovery from hepatic fibrosis following clearance of the infection with the anti-schistosomal drug, praziquantel. In the recovering liver splenomegaly, granuloma density and liver fibrosis were all reduced. Inflammatory cell infiltration into the liver was evident, and the numbers of neutrophils, eosinophils and macrophages were significantly decreased. Transcriptomic analysis revealed the up-regulation of fatty acid metabolism genes and the identification of Peroxisome proliferator activated receptor alpha as the upstream regulator of liver recovery. The aryl hydrocarbon receptor signalling pathway which regulates xenobiotic metabolism was also differentially up-regulated. These findings provide a better understanding of the mechanisms associated with the regression of hepatic schistosomiasis.
Collapse
Affiliation(s)
- Candy Chuah
- QIMR Berghofer Medical Research Institute, Brisbane, Qld 4006, Australia; School of Veterinary Sciences, The University of Queensland, Gatton, Qld 4343, Australia; School of Medical Sciences, Universiti Sains Malaysia, 16150 Kelantan, Malaysia
| | - Malcolm K Jones
- School of Veterinary Sciences, The University of Queensland, Gatton, Qld 4343, Australia
| | - Donald P McManus
- QIMR Berghofer Medical Research Institute, Brisbane, Qld 4006, Australia
| | | | - Melissa L Burke
- QIMR Berghofer Medical Research Institute, Brisbane, Qld 4006, Australia
| | - Helen C Owen
- School of Veterinary Sciences, The University of Queensland, Gatton, Qld 4343, Australia
| | - Grant A Ramm
- QIMR Berghofer Medical Research Institute, Brisbane, Qld 4006, Australia
| | - Geoffrey N Gobert
- QIMR Berghofer Medical Research Institute, Brisbane, Qld 4006, Australia.
| |
Collapse
|
38
|
Abstract
Liver regeneration after partial hepatectomy is an extremely complicated pathophysiologic process, which involves the up-regulation of many proliferation associated proteins and genes. The molecular mechanisms responsible for initiating, maintaining, and terminating this process are still under active investigation and remain one of the research focuses in the field of regenerative medicine. Studies of the mechanism of liver regeneration can provide a theoretical foundation for regeneration promotion and hepatic failure prevention, which is extremely important in clinical practice. This review aims to elucidate the molecular mechanism responsible for the initiation, proliferation and termination of liver regeneration.
Collapse
|
39
|
Baek HJ, Lee YM, Kim TH, Kim JY, Park EJ, Iwabuchi K, Mishra L, Kim SS. Caspase-3/7-mediated Cleavage of β2-spectrin is Required for Acetaminophen-induced Liver Damage. Int J Biol Sci 2016; 12:172-83. [PMID: 26884715 PMCID: PMC4737674 DOI: 10.7150/ijbs.13420] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2015] [Accepted: 11/21/2015] [Indexed: 02/05/2023] Open
Abstract
UNLABELLED The ubiquitously expressed β2-spectrin (β2SP, SPTBN1) is the most common non-erythrocytic member of the β-spectrin gene family. Loss of β2-spectrin leads to defects in liver development, and its haploinsufficiency spontaneously leads to chronic liver disease and the eventual development of hepatocellular cancer. However, the specific role of β2-spectrin in liver homeostasis remains to be elucidated. Here, we reported that β2-spectrin was cleaved by caspase-3/7 upon treatment with acetaminophen which is the main cause of acute liver injury. Blockage of β2-spectrin cleavage robustly attenuated β2-spectrin-specific functions, including regulation of the cell cycle, apoptosis, and transcription. Cleaved fragments of β2-spectrin were physiologically active, and the N- and C-terminal fragments retained discrete interaction partners and activity in transcriptional regulation and apoptosis, respectively. Cleavage of β2-spectrin facilitated the redistribution of the resulting fragments under conditions of liver damage induced by acetaminophen. In contrast, downregulation of β2-spectrin led to resistance to acetaminophen-induced cytotoxicity, and its insufficiency in the liver promoted suppression of acetaminophen-induced liver damage and enhancement of liver regeneration. CONCLUSIONS β2-Spectrin, a TGF-β mediator and signaling molecule, is cleaved and activated by caspase-3/7, consequently enhancing apoptosis and transcriptional control to determine cell fate upon liver damage. These findings have extended our knowledge on the spectrum of β2-spectrin functions from a scaffolding protein to a target and transmitter of TGF-β in liver damage.
Collapse
Affiliation(s)
| | | | | | | | - Eun Jung Park
- 2. Cancer Immunology Branch, National Cancer Center, Goyang, Gyeonggi, 410-769, Korea
| | - Kuniyoshi Iwabuchi
- 3. Department of Biochemistry I, School of Medicine, Kanazawa Medical University, Kahoku-gun, Ishikawa, 920-0293, Japan
| | - Lopa Mishra
- 4. Department of Gastroenterology, Hepatology, and Nutrition, MD Anderson Cancer Center, Houston, Texas, 77030, USA
| | | |
Collapse
|
40
|
TGF-β1 Induces the Dual Regulation of Hepatic Progenitor Cells with Both Anti- and Proliver Fibrosis. Stem Cells Int 2015; 2016:1492694. [PMID: 26839553 PMCID: PMC4709730 DOI: 10.1155/2016/1492694] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2015] [Accepted: 09/17/2015] [Indexed: 02/06/2023] Open
Abstract
Transforming growth factor-beta 1 (TGF-β1) plays a central role in hepatic progenitor cells- (HPCs-) mediated liver repair and fibrosis. However, different effects of TGF-β1 on progenitor cells have not been described. In this study, both in vitro (HPCs cocultured with hepatic stellate cells (HSCs) in transwells) and in vivo (CCl4-injured liver fibrosis rat) systems were used to evaluate the impacts. We found that HPCs pretreated with TGF-β1 for 12 hours inhibited the activation of HSCs, while sensitization for 48 hours increased the activation of HSCs. Consistent with these in vitro results, the in vivo fibrosis rat model showed the same time-dependent dual effect of TGF-β1. Regression of liver fibrosis as well as normalization of serum aminotransferase and albumin levels was detected in the rats transplanted with HPCs pretreated with TGF-β1 for 12 hours. In contrast, severe liver fibrosis and elevated collagen-1 levels were detected in the rats transplanted with HPCs pretreated with TGF-β1 for 48 hours. Furthermore, the TGF-β1-pretreated HPCs were shown to deactivate HSCs via enhancing SERPINE1 expression. Inhibition of SERPINE1 reversed the deactivation response in a dose-dependent manner.
Collapse
|
41
|
Katselis C, Apostolou K, Feretis T, Papanikolaou IG, Zografos GC, Toutouzas K, Papalois A. Role of Stem Cells Transplantation in Tissue Regeneration After Acute or Chronic Acetaminophen Induced Liver Injury. J INVEST SURG 2015; 29:112-20. [PMID: 26650889 DOI: 10.3109/08941939.2015.1086040] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
PURPOSE Acetaminophen-induced liver injury (APAP) is recognized as a frequent etiologic factor responsible for hepatic damage in the developed world. Management remains still elusive as treatment options are limited and their results are inconclusive. Consequently new strategies are explored at the experimental level. Mesenchymal stem cells (MSCs) present a promising modality as they can promote liver regeneration (LG) and compensate acute liver injury (ALI). MATERIALS AND METHODS Our research was focused on articles related to drug-induced liver injury, mechanisms of liver regeneration (LG) after Acute Liver Injury (ALI) and recent experimental protocols of Mesenchymal Stem Cells (MSCs) transplantation after chemical insult. All these studies are cited on Pubmed and MedLine. RESULTS This review has three distinct sections. First recent developments in ALI pathogenesis are presented. The second section covers cellular pathways and histological findings relevant to liver regeneration. The final chapter analyzes MSCs transplantation protocols after ALI and interrelation between liver regeneration and hepatic differentiation of MSCs. CONCLUSION Adipose tissue stem cells (ADSCs) and (MSCs) transplantation represents a promising modality in severe ALI management although many aspects remain to be clarified.
Collapse
Affiliation(s)
- Charalampos Katselis
- a Department of Propaedeutic Surgery , University of Athens, School of Medicine, General Hospital of Athens "Hippocration" , Athens , Greece.,b Experimental - Research Center , ELPEN Pharmaceuticals , Pikermi , Attica
| | - Konstantinos Apostolou
- a Department of Propaedeutic Surgery , University of Athens, School of Medicine, General Hospital of Athens "Hippocration" , Athens , Greece.,b Experimental - Research Center , ELPEN Pharmaceuticals , Pikermi , Attica
| | - Themistoklis Feretis
- a Department of Propaedeutic Surgery , University of Athens, School of Medicine, General Hospital of Athens "Hippocration" , Athens , Greece.,b Experimental - Research Center , ELPEN Pharmaceuticals , Pikermi , Attica
| | - Ioannis G Papanikolaou
- a Department of Propaedeutic Surgery , University of Athens, School of Medicine, General Hospital of Athens "Hippocration" , Athens , Greece.,b Experimental - Research Center , ELPEN Pharmaceuticals , Pikermi , Attica
| | - George C Zografos
- a Department of Propaedeutic Surgery , University of Athens, School of Medicine, General Hospital of Athens "Hippocration" , Athens , Greece
| | - Konstantinos Toutouzas
- a Department of Propaedeutic Surgery , University of Athens, School of Medicine, General Hospital of Athens "Hippocration" , Athens , Greece
| | - Apostolos Papalois
- a Department of Propaedeutic Surgery , University of Athens, School of Medicine, General Hospital of Athens "Hippocration" , Athens , Greece.,b Experimental - Research Center , ELPEN Pharmaceuticals , Pikermi , Attica
| |
Collapse
|
42
|
García-Pérez R, Revilla-Nuin B, Martínez CM, Bernabé-García A, Baroja Mazo A, Parrilla Paricio P. Associated Liver Partition and Portal Vein Ligation (ALPPS) vs Selective Portal Vein Ligation (PVL) for Staged Hepatectomy in a Rat Model. Similar Regenerative Response? PLoS One 2015; 10:e0144096. [PMID: 26630386 PMCID: PMC4668031 DOI: 10.1371/journal.pone.0144096] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Accepted: 11/12/2015] [Indexed: 12/14/2022] Open
Abstract
Associated liver partition and portal vein ligation for staged hepatectomy (ALPPS) is a two-stage hepatectomy technique which can be associated with a hypertrophic stimulus on the future liver remnant (FLR) stronger than other techniques–such as portal vein ligation (PVL). However, the reason of such hypertrophy is still unclear, but it is suggested that liver transection combined with portal vein ligation (ALPPS) during the first stage of this technique may play a key role. The aim of this study is to compare the hypertrophic stimulus on the FLR and the clinical changes associated with both ALPPS and PVL in a rat surgical model. For this purpose, three groups of SD rats were used, namely ALPPS (n = 30), PVL (n = 30) and sham-treated (n = 30). The second stage of ALPPS (hepatectomy of the atrophic lobes), was performed at day 8. Blood and FLR samples were collected at 1, 24, 48 hours, 8 days and 12 weeks after the surgeries. ALPPS provoked a greater degree of hypertrophy of the FLR than the PVL at 48 hours and 8 days (p<0.05). The molecular pattern was also different, with the highest expression of IL-1β at 24h, IL-6 at 8 days, and HGF and TNF-α at 48 hours and 8 days (p<0.05). ALPPS also brought about a mild proliferative stimulus at 12 weeks, with a higher expression of HGF and TGF-β (p<0.05) than PVL. Clinically, ALPPS caused a significant liver damage during the first 48 hours, with a recovery of liver function at day 8. In conclusion, ALPPS seems to induce higher functional hypertrophy on the FLR than PVL at day 8. Such regenerative response seems to be leaded by a complex interaction between pro-mitogenic (IL-6, HGF, TNF-α) and antiproliferative (IL1-β and TGF-β) cytokines.
Collapse
Affiliation(s)
- Rocío García-Pérez
- Department of Surgery, Experimental Surgery Unit, IMIB-LAIB Research Center, El Palmar (Murcia), Spain
| | - Beatriz Revilla-Nuin
- Department of Surgery, Experimental Surgery Unit, IMIB-LAIB Research Center, El Palmar (Murcia), Spain
- CIBERehd, Barcelona, Spain
| | - Carlos M. Martínez
- Department of Surgery, Experimental Surgery Unit, IMIB-LAIB Research Center, El Palmar (Murcia), Spain
- CIBERehd, Barcelona, Spain
- * E-mail:
| | - Angel Bernabé-García
- Department of Surgery, Experimental Surgery Unit, IMIB-LAIB Research Center, El Palmar (Murcia), Spain
| | - Alberto Baroja Mazo
- Department of Surgery, Experimental Surgery Unit, IMIB-LAIB Research Center, El Palmar (Murcia), Spain
| | - Pascual Parrilla Paricio
- Department of Surgery, Experimental Surgery Unit, IMIB-LAIB Research Center, El Palmar (Murcia), Spain
- CIBERehd, Barcelona, Spain
| |
Collapse
|
43
|
Wang SS, Jiang J, Liang XH, Tang YL. Links between cancer stem cells and epithelial-mesenchymal transition. Onco Targets Ther 2015; 8:2973-80. [PMID: 26527883 PMCID: PMC4621173 DOI: 10.2147/ott.s91863] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
The epithelial-mesenchymal transition (EMT) has been reported to be an important program that is often activated during the process of cancer invasion and metastasis. Cancer stem cells (CSCs) that can initiate and maintain cancer are also involved in invasion and metastasis of cancer. Recently, insights into the molecular mechanisms and functional features of mesenchymal cells have been greatly colored by findings that some of them have been endowed with the self-renewal trait associated with normal tissue stem cells and CSCs. Among cancer cells experiencing EMT, only some of the most competent CSCs will succeed in planting in another organ. In this paper, we review the molecular mechanism behind the link of EMT and CSCs in cancer progression.
Collapse
Affiliation(s)
- Sha-Sha Wang
- State Key Laboratory of Oral Diseases, Sichuan University, Chengdu, People's Republic of China ; Department of Oral and Maxillofacial Surgery, Sichuan University, Chengdu, People's Republic of China
| | - Jian Jiang
- State Key Laboratory of Oral Diseases, Sichuan University, Chengdu, People's Republic of China ; Department of Oral and Maxillofacial Surgery, Sichuan University, Chengdu, People's Republic of China
| | - Xin-Hua Liang
- State Key Laboratory of Oral Diseases, Sichuan University, Chengdu, People's Republic of China ; Department of Oral and Maxillofacial Surgery, Sichuan University, Chengdu, People's Republic of China
| | - Ya-Ling Tang
- State Key Laboratory of Oral Diseases, Sichuan University, Chengdu, People's Republic of China ; Department of Oral Pathology, West China Hospital of Stomatology, Sichuan University, Chengdu, People's Republic of China
| |
Collapse
|
44
|
Motawi TK, El-Boghdady NA, El-Sayed AM, Helmy HS. Comparative study of the effects of PEGylated interferon-α2a versus 5-fluorouracil on cancer stem cells in a rat model of hepatocellular carcinoma. Tumour Biol 2015; 37:1617-25. [PMID: 26304505 DOI: 10.1007/s13277-015-3920-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2015] [Accepted: 08/11/2015] [Indexed: 12/20/2022] Open
Abstract
Cancer stem cells (CSCs) in hepatocellular carcinoma (HCC) possess tumor-initiating, metastatic, and drug resistance properties. This study was conducted to evaluate the effects of PEGylated interferon-α2a (PEG-IFN-α2a) and 5-fluorouracil (5-FU) on the expression of CSC markers and on specific pathways that contribute to the propagation of CSCs in HCC. HCC was initiated in rats using a single intraperitoneal dose of diethylnitrosamine (DENA) (200 mg/kg) and promoted by weekly subcutaneous injections of carbon tetrachloride (CCl4) for 6 weeks. After the appearance of dysplastic nodules, the animals received PEG-IFN-α2a or 5-FU for 8 weeks. CSC markers (OV6, CD90) and molecules related to transforming growth factor β (TGF-β) and other signaling pathways were assessed in hepatic tissues. The PEG-IFN-α2a treatment effectively suppressed the hepatic expression of OV6 and CD90, ameliorated the diminished hepatic expression of TGF-β receptor II (TGF-βRII) and β2-spectrin (β2SP), and significantly reduced the elevated hepatic expression of TGF-β1, interleukin6 (IL6), signal transducer and activator of transcription3 (STAT3), and vascular endothelial growth factor (VEGF). In contrast, the 5-FU treatment failed to reduce the overexpression of CSC markers and barely affected the disrupted TGF-β signaling. Furthermore, it had no effect on angiogenesis or nitrosative stress. PEG-IFN-α2a, but not 5-FU, could reduce the propagation of CSCs during the progression of HCC by upregulating the disrupted TGF-β signaling, suppressing the IL6/STAT3 pathway and reducing angiogenesis.
Collapse
Affiliation(s)
- Tarek Kamal Motawi
- Department of Biochemistry, Faculty of Pharmacy, Cairo University, Cairo, 11562, Egypt
| | | | - Abeer Mostafa El-Sayed
- Department of Pathology, National Cancer Institute, Cairo University, Cairo, 11796, Egypt
| | - Hebatullah Samy Helmy
- Department of Biochemistry, Faculty of Pharmacy, Cairo University, Cairo, 11562, Egypt.
| |
Collapse
|
45
|
Coombes J, Swiderska-Syn M, Dollé L, Reid D, Eksteen B, Claridge L, Briones-Orta MA, Shetty S, Oo YH, Riva A, Chokshi S, Papa S, Mi Z, Kuo PC, Williams R, Canbay A, Adams DH, Diehl AM, van Grunsven LA, Choi SS, Syn WK. Osteopontin neutralisation abrogates the liver progenitor cell response and fibrogenesis in mice. Gut 2015; 64:1120-31. [PMID: 24902765 PMCID: PMC4487727 DOI: 10.1136/gutjnl-2013-306484] [Citation(s) in RCA: 73] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2013] [Accepted: 05/22/2014] [Indexed: 12/29/2022]
Abstract
BACKGROUND Chronic liver injury triggers a progenitor cell repair response, and liver fibrosis occurs when repair becomes deregulated. Previously, we reported that reactivation of the hedgehog pathway promotes fibrogenic liver repair. Osteopontin (OPN) is a hedgehog-target, and a cytokine that is highly upregulated in fibrotic tissues, and regulates stem-cell fate. Thus, we hypothesised that OPN may modulate liver progenitor cell response, and thereby, modulate fibrotic outcomes. We further evaluated the impact of OPN-neutralisation on murine liver fibrosis. METHODS Liver progenitors (603B and bipotential mouse oval liver) were treated with OPN-neutralising aptamers in the presence or absence of transforming growth factor (TGF)-β, to determine if (and how) OPN modulates liver progenitor function. Effects of OPN-neutralisation (using OPN-aptamers or OPN-neutralising antibodies) on liver progenitor cell response and fibrogenesis were assessed in three models of liver fibrosis (carbon tetrachloride, methionine-choline deficient diet, 3,5,-diethoxycarbonyl-1,4-dihydrocollidine diet) by quantitative real time (qRT) PCR, Sirius-Red staining, hydroxyproline assay, and semiquantitative double-immunohistochemistry. Finally, OPN expression and liver progenitor response were corroborated in liver tissues obtained from patients with chronic liver disease. RESULTS OPN is overexpressed by liver progenitors in humans and mice. In cultured progenitors, OPN enhances viability and wound healing by modulating TGF-β signalling. In vivo, OPN-neutralisation attenuates the liver progenitor cell response, reverses epithelial-mesenchymal-transition in Sox9+ cells, and abrogates liver fibrogenesis. CONCLUSIONS OPN upregulation during liver injury is a conserved repair response, and influences liver progenitor cell function. OPN-neutralisation abrogates the liver progenitor cell response and fibrogenesis in mouse models of liver fibrosis.
Collapse
Affiliation(s)
- J Coombes
- Regeneration and Repair Group, The Institute of Hepatology, Foundation for Liver Research, London, UK
| | - M Swiderska-Syn
- Division of Gastroenterology, Department of Medicine, Duke University, NC, USA
| | - L Dollé
- Liver Cell Biology Lab (LIVR), Department of Cell Biology (CYTO), Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel, Brussels, Belgium
| | - D Reid
- Snyder Institute for Chronic Diseases, Health Research and Innovation Centre (HRIC), University of Calgary, Canada
| | - B Eksteen
- Snyder Institute for Chronic Diseases, Health Research and Innovation Centre (HRIC), University of Calgary, Canada
| | - L Claridge
- Centre for Liver Research, NIHR Institute for Biomedical Research, University of Birmingham, UK
| | - MA Briones-Orta
- Regeneration and Repair Group, The Institute of Hepatology, Foundation for Liver Research, London, UK
| | - S Shetty
- Centre for Liver Research, NIHR Institute for Biomedical Research, University of Birmingham, UK
| | - YH Oo
- Centre for Liver Research, NIHR Institute for Biomedical Research, University of Birmingham, UK
| | - A Riva
- Viral Hepatitis Group, The Institute of Hepatology, Foundation for Liver Research, London, UK
| | - S Chokshi
- Viral Hepatitis Group, The Institute of Hepatology, Foundation for Liver Research, London, UK
| | - S Papa
- Cell Signaling Group, The Institute of Hepatology, Foundation for Liver Research, London, UK
| | - Z Mi
- Department of Surgery, Loyola University, Chicago, USA
| | - PC Kuo
- Department of Surgery, Loyola University, Chicago, USA
| | - R Williams
- Regeneration and Repair Group, The Institute of Hepatology, Foundation for Liver Research, London, UK
| | - A Canbay
- Department of Gastroenterology and Hepatology, Essen University Hospital, Essen, Germany
| | - DH Adams
- Centre for Liver Research, NIHR Institute for Biomedical Research, University of Birmingham, UK
| | - AM Diehl
- Division of Gastroenterology, Department of Medicine, Duke University, NC, USA
| | - LA van Grunsven
- Liver Cell Biology Lab (LIVR), Department of Cell Biology (CYTO), Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel, Brussels, Belgium
| | - SS Choi
- Division of Gastroenterology, Department of Medicine, Duke University, NC, USA,Section of Gastroenterology, Department of Medicine, Durham Veteran Affairs Medical Center, Durham, NC, USA
| | - WK Syn
- Regeneration and Repair Group, The Institute of Hepatology, Foundation for Liver Research, London, UK,Centre for Liver Research, NIHR Institute for Biomedical Research, University of Birmingham, UK,Department of Hepatology, Barts Health NHS Trust, London, UK,Senior and Corresponding Author: Dr Wing-Kin Syn, Head of Liver Regeneration and Repair, The Institute of Hepatology, Foundation for Liver Research, London WC1E 6HX, Tel: 44-20272559837,
| |
Collapse
|
46
|
Woods LT, Camden JM, El-Sayed FG, Khalafalla MG, Petris MJ, Erb L, Weisman GA. Increased Expression of TGF-β Signaling Components in a Mouse Model of Fibrosis Induced by Submandibular Gland Duct Ligation. PLoS One 2015; 10:e0123641. [PMID: 25955532 PMCID: PMC4425516 DOI: 10.1371/journal.pone.0123641] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2014] [Accepted: 02/21/2015] [Indexed: 02/06/2023] Open
Abstract
Transforming growth factor-β (TGF-β) is a multi-functional cytokine with a well-described role in the regulation of tissue fibrosis and regeneration in the liver, kidney and lung. Submandibular gland (SMG) duct ligation and subsequent deligation in rodents is a classical model for studying salivary gland damage and regeneration. While previous studies suggest that TGF-β may contribute to salivary gland fibrosis, the expression of TGF-β signaling components has not been investigated in relation to mouse SMG duct ligation-induced fibrosis and regeneration following ductal deligation. Following a 7 day SMG duct ligation, TGF-β1 and TGF-β3 were significantly upregulated in the SMG, as were TGF-β receptor 1 and downstream Smad family transcription factors in salivary acinar cells, but not in ductal cells. In acinar cells, duct ligation also led to upregulation of snail, a Smad-activated E-cadherin repressor and regulator of epithelial-mesenchymal transition, whereas in ductal cells upregulation of E-cadherin was observed while snail expression was unchanged. Upregulation of these TGF-β signaling components correlated with upregulation of fibrosis markers collagen 1 and fibronectin, responses that were inhibited by administration of the TGF-β receptor 1 inhibitors SB431542 or GW788388. After SMG regeneration following a 28 day duct deligation, TGF-β signaling components and epithelial-mesenchymal transition markers returned to levels similar to non-ligated controls. The results from this study indicate that increased TGF-β signaling contributes to duct ligation-induced changes in salivary epithelium that correlate with glandular fibrosis. Furthermore, the reversibility of enhanced TGF-β signaling in acinar cells of duct-ligated mouse SMG after deligation indicates that this is an ideal model for studying TGF-β signaling mechanisms in salivary epithelium as well as mechanisms of fibrosis initiation and their resolution.
Collapse
Affiliation(s)
- Lucas T. Woods
- Department of Biochemistry, University of Missouri, Columbia, Missouri, United States of America
- Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, Missouri, United States of America
| | - Jean M. Camden
- Department of Biochemistry, University of Missouri, Columbia, Missouri, United States of America
- Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, Missouri, United States of America
| | - Farid G. El-Sayed
- Department of Biochemistry, University of Missouri, Columbia, Missouri, United States of America
- Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, Missouri, United States of America
| | - Mahmoud G. Khalafalla
- Department of Biochemistry, University of Missouri, Columbia, Missouri, United States of America
- Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, Missouri, United States of America
| | - Michael J. Petris
- Department of Biochemistry, University of Missouri, Columbia, Missouri, United States of America
- Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, Missouri, United States of America
- Department of Nutritional Sciences and Exercise Physiology, University of Missouri, Columbia, Missouri, United States of America
| | - Laurie Erb
- Department of Biochemistry, University of Missouri, Columbia, Missouri, United States of America
- Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, Missouri, United States of America
| | - Gary A. Weisman
- Department of Biochemistry, University of Missouri, Columbia, Missouri, United States of America
- Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, Missouri, United States of America
- * E-mail:
| |
Collapse
|
47
|
Zhi X, Lin L, Yang S, Bhuvaneshwar K, Wang H, Gusev Y, Lee MH, Kallakury B, Shivapurkar N, Cahn K, Tian X, Marshall JL, Byers SW, He AR. βII-Spectrin (SPTBN1) suppresses progression of hepatocellular carcinoma and Wnt signaling by regulation of Wnt inhibitor kallistatin. Hepatology 2015; 61:598-612. [PMID: 25307947 PMCID: PMC4327990 DOI: 10.1002/hep.27558] [Citation(s) in RCA: 84] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2014] [Accepted: 10/07/2014] [Indexed: 12/27/2022]
Abstract
UNLABELLED βII-Spectrin (SPTBN1) is an adapter protein for Smad3/Smad4 complex formation during transforming growth factor beta (TGF-β) signal transduction. Forty percent of SPTBN1(+/-) mice spontaneously develop hepatocellular carcinoma (HCC), and most cases of human HCC have significant reductions in SPTBN1 expression. In this study, we investigated the possible mechanisms by which loss of SPTBN1 may contribute to tumorigenesis. Livers of SPTBN1(+/-) mice, compared to wild-type mouse livers, display a significant increase in epithelial cell adhesion molecule-positive (EpCAM(+)) cells and overall EpCAM expression. Inhibition of SPTBN1 in human HCC cell lines increased the expression of stem cell markers EpCAM, Claudin7, and Oct4, as well as decreased E-cadherin expression and increased expression of vimentin and c-Myc, suggesting reversion of these cells to a less differentiated state. HCC cells with decreased SPTBN1 also demonstrate increased sphere formation, xenograft tumor development, and invasion. Here we investigate possible mechanisms by which SPTBN1 may influence the stem cell traits and aggressive behavior of HCC cell lines. We found that HCC cells with decreased SPTBN1 express much less of the Wnt inhibitor kallistatin and exhibit decreased β-catenin phosphorylation and increased β-catenin nuclear localization, indicating Wnt signaling activation. Restoration of kallistatin expression in these cells reversed the observed Wnt activation. CONCLUSION SPTBN1 expression in human HCC tissues is positively correlated with E-cadherin and kallistatin levels, and decreased SPTBN1 and kallistatin gene expression is associated with decreased relapse-free survival. Our data suggest that loss of SPTBN1 activates Wnt signaling, which promotes acquisition of stem cell-like features, and ultimately contributes to malignant tumor progression.
Collapse
Affiliation(s)
- Xiuling Zhi
- Georgetown-Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, USA
- Laboratory of Medical Molecular Biology, Training Center of Medical Experiments, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Ling Lin
- Georgetown-Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, USA
| | - Shaoxian Yang
- Georgetown-Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, USA
| | - Krithika Bhuvaneshwar
- Georgetown-Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, USA
| | - Hongkun Wang
- Georgetown-Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, USA
| | - Yuriy Gusev
- Georgetown-Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, USA
| | - Mi-Hye Lee
- Georgetown-Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, USA
| | - Bhaskar Kallakury
- Georgetown-Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, USA
| | - Narayan Shivapurkar
- Georgetown-Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, USA
| | - Katherine Cahn
- Georgetown-Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, USA
| | - Xuefei Tian
- Georgetown-Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, USA
| | - John L. Marshall
- Georgetown-Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, USA
| | - Stephen W. Byers
- Georgetown-Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, USA
| | - Aiwu R. He
- Georgetown-Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, USA
- Corresponding author: Aiwu R. He, M.D. Ph.D., Departments of Medicine and Oncology, Georgetown-Lombardi Comprehensive Cancer Center, Georgetown University, 3800 Reservoir Road, NW, Washington, DC 20007, USA., Phone: 02-444-1259, Fax: 202-444-9429,
| |
Collapse
|
48
|
Paganelli M, Nyabi O, Sid B, Evraerts J, El Malmi I, Heremans Y, Dollé L, Benton C, Calderon PB, van Grunsven L, Heimberg H, Campard D, Sokal E, Najimi M. Downregulation of Sox9 Expression Associates with Hepatogenic Differentiation of Human Liver Mesenchymal Stem/Progenitor Cells. Stem Cells Dev 2014; 23:1377-91. [DOI: 10.1089/scd.2013.0169] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Affiliation(s)
- Massimiliano Paganelli
- Laboratory of Pediatric Hepatology and Cell Therapy, Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain, Brussels, Belgium
| | - Omar Nyabi
- Laboratory of Pediatric Hepatology and Cell Therapy, Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain, Brussels, Belgium
| | - Brice Sid
- Toxicology and Cancer Biology Research Group, PMNT Unit, Louvain Drug Research Institute, Université Catholique de Louvain, Brussels, Belgium
| | - Jonathan Evraerts
- Laboratory of Pediatric Hepatology and Cell Therapy, Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain, Brussels, Belgium
| | - Imane El Malmi
- Laboratory of Pediatric Hepatology and Cell Therapy, Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain, Brussels, Belgium
| | - Yves Heremans
- Diabetes Research Center, Vrije Universiteit Brussel, Brussels, Belgium
| | - Laurent Dollé
- Liver Cell Biology Lab, Vrije Universiteit Brussel, Brussels, Belgium
| | - Carley Benton
- Laboratory of Pediatric Hepatology and Cell Therapy, Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain, Brussels, Belgium
| | - Pedro-Buc Calderon
- Toxicology and Cancer Biology Research Group, PMNT Unit, Louvain Drug Research Institute, Université Catholique de Louvain, Brussels, Belgium
| | - Leo van Grunsven
- Liver Cell Biology Lab, Vrije Universiteit Brussel, Brussels, Belgium
| | - Harry Heimberg
- Diabetes Research Center, Vrije Universiteit Brussel, Brussels, Belgium
| | - David Campard
- Laboratory of Pediatric Hepatology and Cell Therapy, Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain, Brussels, Belgium
| | - Etienne Sokal
- Laboratory of Pediatric Hepatology and Cell Therapy, Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain, Brussels, Belgium
| | - Mustapha Najimi
- Laboratory of Pediatric Hepatology and Cell Therapy, Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain, Brussels, Belgium
| |
Collapse
|
49
|
Bogaerts E, Heindryckx F, Vandewynckel YP, Van Grunsven LA, Van Vlierberghe H. The roles of transforming growth factor-β, Wnt, Notch and hypoxia on liver progenitor cells in primary liver tumours (Review). Int J Oncol 2014; 44:1015-22. [PMID: 24504124 PMCID: PMC3977811 DOI: 10.3892/ijo.2014.2286] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2013] [Accepted: 11/28/2013] [Indexed: 12/11/2022] Open
Abstract
Primary liver tumours have a high incidence and mortality. The most important forms are hepatocellular carcinoma and intrahepatic cholangiocarcinoma, both can occur together in the mixed phenotype hepatocellular-cholangiocarcinoma. Liver progenitor cells (LPCs) are bipotential stem cells activated in case of severe liver damage and are capable of forming both cholangiocytes and hepatocytes. Possibly, alterations in Wnt, transforming growth factor-β, Notch and hypoxia pathways in these LPCs can cause them to give rise to cancer stem cells, capable of driving tumourigenesis. In this review, we summarize and discuss current knowledge on the role of these pathways in LPC activation and differentiation during hepatocarcinogenesis.
Collapse
Affiliation(s)
- Eliene Bogaerts
- Department of Gastroenterology and Hepatology, 1K12, Ghent University Hospital, 9000 Gent, Belgium
| | - Femke Heindryckx
- Department of Medical Biochemistry and Microbiology, Uppsala University, 751 23 Uppsala, Sweden
| | - Yves-Paul Vandewynckel
- Department of Gastroenterology and Hepatology, 1K12, Ghent University Hospital, 9000 Gent, Belgium
| | - Leo A Van Grunsven
- Department of Cell Biology, Liver Cell Biology Lab, Vrije Universiteit Brussel, 1090 Brussels, Belgium
| | - Hans Van Vlierberghe
- Department of Gastroenterology and Hepatology, 1K12, Ghent University Hospital, 9000 Gent, Belgium
| |
Collapse
|
50
|
Kim JB, Ann YH, Park SY, Jee HG, Kim HR, Lee JH, Yu SJ, Lee HS, Kim YJ. Side population in LX2 cells decreased by transforming growth factor-β. Hepatol Res 2014; 44:229-37. [PMID: 23578133 DOI: 10.1111/hepr.12106] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2012] [Revised: 02/18/2013] [Accepted: 02/25/2013] [Indexed: 12/11/2022]
Abstract
AIM Side population (SP) cells are known to be enriched in stem/progenitor-like cells. Transforming growth factor (TGF)-β signaling is associated with extracellular matrix (ECM) production in hepatic stellate cells. We hypothesized that the SP fraction in LX2 cells is associated with ECM deposition, which is regulated through TGF-β signaling. METHODS We investigated the relationship between SP cells and TGF-β signaling in the hepatic stellate cell line LX2. The effects of TGF-β and SB431542 on the SP fraction and expression of collagen type I and phospho-Smad2 was determined. RESULTS We identified 0.8-3% SP cells in LX2 cells. The growth rate of sorted SP and non-SP cells was similar to that of the original LX2 population, but population of the G0/G1 phase was increased in SP cells. Treatment of LX2 cells with TGF-β decreased the SP fraction in a dose-dependent manner and increased the production of collagen type I. Treatment of LX2 cells with SB431542 blocked the effect of TGF-β on the SP fraction and the expression of collagen type I. We cultured LX2 cells on collagen-coated dishes to observe the effect of ECM deposition on the SP fraction. The growth rate and cell cycle distribution was similar to that observed on normal tissue culture dishes, but the SP fraction was decreased when LX2 cells were cultured on collagen-coated plates. CONCLUSION These results show that LX2 cells contain an SP fraction and that TGF-β signaling is involved in the induction of ECM deposition as well as the number of SP cells.
Collapse
Affiliation(s)
- Jong Bin Kim
- Department of Internal Medicine and Liver Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | | | | | | | | | | | | | | | | |
Collapse
|