201
|
Antifibrotics in liver disease: are we getting closer to clinical use? Hepatol Int 2018; 13:25-39. [PMID: 30302735 DOI: 10.1007/s12072-018-9897-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Accepted: 09/14/2018] [Indexed: 12/14/2022]
Abstract
The process of wound healing in response to chronic liver injury leads to the development of liver fibrosis. Regardless of etiology, the profound impact of the degree of liver fibrosis on the prognosis of chronic liver diseases has been well demonstrated. While disease-specific therapy, such as treatments for viral hepatitis, has been shown to reverse liver fibrosis and cirrhosis in both clinical trials and real-life practice, subsets of patients do not demonstrate fibrosis regression. Moreover, where disease-specific therapies are not available, the need for antifibrotics exists. Increased understanding into the pathogenesis of liver fibrosis sets the stage to focus on antifibrotic therapies attempting to: (1) Minimize liver injury and inflammation; (2) Inhibit liver fibrogenesis by enhancing or inhibiting target receptor-ligand interactions or intracellular signaling pathways; and (3) Promote fibrosis resolution. While no antifibrotic therapies are currently available, a number are now being evaluated in clinical trials, and their use is becoming closer to reality for select subsets of patients.
Collapse
|
202
|
Tee JK, Peng F, Tan YL, Yu B, Ho HK. Magnesium Isoglycyrrhizinate Ameliorates Fibrosis and Disrupts TGF-β-Mediated SMAD Pathway in Activated Hepatic Stellate Cell Line LX2. Front Pharmacol 2018; 9:1018. [PMID: 30319402 PMCID: PMC6167412 DOI: 10.3389/fphar.2018.01018] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2018] [Accepted: 08/22/2018] [Indexed: 12/21/2022] Open
Abstract
Liver fibrosis is a histological change often attributed to the activation of hepatic stellate cells (HSCs) and the excessive formation of scar tissues in the liver. Advanced stages of the disease frequently lead to cirrhosis. Magnesium isoglycyrrhizinate (MgIG) has been accepted as a hepatoprotective drug with the potential of alleviating inflammatory conditions and thus promote liver recovery from viral- or drug-induced injury. While MgIG has been empirically integrated into the clinics to treat some liver diseases, its anti-fibrotic effect and the associated mechanisms remain poorly characterized. Herein, we demonstrated that 1 mg/ml MgIG attenuated the production of αSMA and collagen-1 in activated HSCs using TGF-β1-induced human HSCs LX2 as the fibrotic cell model. We found that MgIG exerts an inhibitory effect on the TGF-β-SMAD signaling pathway by arresting the binding of downstream transcription factors SMAD2/3 and SMAD4. Furthermore, MgIG was shown to suppress proliferation and induce senescence of activated LX2 cells. Protein expression of p27 and enzymatic activity of senescence-associated β-galactosidase were elevated upon exposure to MgIG. In addition, we observed that exposure of activated LX2 cells to MgIG reduces TGF-β-induced apoptosis. Interestingly, a lower toxicity profile was observed when human fetal hepatocytes LO2 were exposed to the same concentration and duration of the drug, suggesting the specificity of MgIG effect toward activated HSCs. Overall, hepatoprotective concentrations of MgIG is shown to exert a direct effect on liver fibrosis through inhibiting TGF-β-signaling, in which SMAD2/3 pathway could be one of the mechanisms responsible for the fibrotic response, thereby restoring the surviving cells toward a more quiescent phenotype. This provides critical mechanistic insights to support an otherwise empirical therapy.
Collapse
Affiliation(s)
- Jie Kai Tee
- NUS Graduate School for Integrative Sciences and Engineering, Centre for Life Sciences, National University of Singapore, Singapore, Singapore
- Department of Pharmacy, Faculty of Science, National University of Singapore, Singapore, Singapore
| | - Fei Peng
- Department of Pharmacy, Faculty of Science, National University of Singapore, Singapore, Singapore
| | - Yeong Lan Tan
- NUS Graduate School for Integrative Sciences and Engineering, Centre for Life Sciences, National University of Singapore, Singapore, Singapore
- Department of Pharmacy, Faculty of Science, National University of Singapore, Singapore, Singapore
| | - Bo Yu
- Department of Pharmacy, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Han Kiat Ho
- NUS Graduate School for Integrative Sciences and Engineering, Centre for Life Sciences, National University of Singapore, Singapore, Singapore
- Department of Pharmacy, Faculty of Science, National University of Singapore, Singapore, Singapore
| |
Collapse
|
203
|
Strowitzki MJ, Kirchberg J, Tuffs C, Schiedeck M, Ritter AS, Biller M, Harnoss JM, Lasitschka F, Schmidt T, Radhakrishnan P, Ulrich A, Schneider M. Loss of Prolyl-Hydroxylase 1 Protects against Biliary Fibrosis via Attenuated Activation of Hepatic Stellate Cells. THE AMERICAN JOURNAL OF PATHOLOGY 2018; 188:2826-2838. [PMID: 30248340 DOI: 10.1016/j.ajpath.2018.08.003] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/15/2017] [Revised: 08/10/2018] [Accepted: 08/14/2018] [Indexed: 12/12/2022]
Abstract
Liver fibrosis, eventually progressing to cirrhosis necessitating liver transplantation, poses a significant clinical problem. Oxygen shortage (hypoxia) and hypoxia-inducible transcription factors (HIFs) have been acknowledged as important drivers of liver fibrosis. The significance of oxygen-sensing HIF prolyl-hydroxylase (PHD) enzymes in this context has, however, remained elusive. In this study, we demonstrate that loss of PHD1 (PHD1-/-) attenuates the development of liver fibrosis in mice subjected to chronic bile duct injury, induced by 3,5-diethoxycarbonyl-1,4-dihydrocollidine. This effect was accompanied with reduced recruitment of inflammatory leukocytes and attenuated occurrence of profibrotic myofibroblasts in PHD1-/- livers. Further analyses focused on the significance of PHD1 in the activation of hepatic stellate cells (HSCs), which represent the driving force in liver fibrosis. Primary HSCs isolated from PHD1-/- mice displayed significantly attenuated myofibroblast differentiation and profibrogenic properties compared with HSCs isolated from wild-type mice. Consistently, the expression of various profibrogenic and promitogenic factors was reduced in PHD1-/- HSCs, without alterations in HIF-1α protein levels. Of importance, PHD1 protein was expressed in HSCs within human livers, and PHD1 transcript expression was significantly increased with disease severity in hepatic tissue from patients with liver fibrosis. Collectively, these findings indicate that PHD1 deficiency protects against liver fibrosis and that these effects are partly due to attenuated activation of HSCs. PHD1 may represent a therapeutic target to alleviate liver fibrosis.
Collapse
Affiliation(s)
- Moritz J Strowitzki
- Department of General, Visceral and Transplantation Surgery, University Hospital Heidelberg, Heidelberg, Germany
| | - Johanna Kirchberg
- Department of General, Visceral and Transplantation Surgery, University Hospital Heidelberg, Heidelberg, Germany
| | - Christopher Tuffs
- Department of General, Visceral and Transplantation Surgery, University Hospital Heidelberg, Heidelberg, Germany
| | - Maximilian Schiedeck
- Department of General, Visceral and Transplantation Surgery, University Hospital Heidelberg, Heidelberg, Germany
| | - Alina S Ritter
- Department of General, Visceral and Transplantation Surgery, University Hospital Heidelberg, Heidelberg, Germany
| | - Marvin Biller
- Department of General, Visceral and Transplantation Surgery, University Hospital Heidelberg, Heidelberg, Germany
| | - Jonathan M Harnoss
- Department of General, Visceral and Transplantation Surgery, University Hospital Heidelberg, Heidelberg, Germany
| | - Felix Lasitschka
- Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
| | - Thomas Schmidt
- Department of General, Visceral and Transplantation Surgery, University Hospital Heidelberg, Heidelberg, Germany
| | - Praveen Radhakrishnan
- Department of General, Visceral and Transplantation Surgery, University Hospital Heidelberg, Heidelberg, Germany
| | - Alexis Ulrich
- Department of General, Visceral and Transplantation Surgery, University Hospital Heidelberg, Heidelberg, Germany
| | - Martin Schneider
- Department of General, Visceral and Transplantation Surgery, University Hospital Heidelberg, Heidelberg, Germany.
| |
Collapse
|
204
|
Bobowski-Gerard M, Zummo FP, Staels B, Lefebvre P, Eeckhoute J. Retinoids Issued from Hepatic Stellate Cell Lipid Droplet Loss as Potential Signaling Molecules Orchestrating a Multicellular Liver Injury Response. Cells 2018; 7:cells7090137. [PMID: 30217095 PMCID: PMC6162435 DOI: 10.3390/cells7090137] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Revised: 09/11/2018] [Accepted: 09/13/2018] [Indexed: 02/08/2023] Open
Abstract
Hepatic stellate cells (HSCs) serve as the main body storage compartment for vitamin A through retinyl ester (RE)-filled lipid droplets (LDs). Upon liver injury, HSCs adopt a myofibroblastic phenotype characterized by an elevated expression of extracellular matrix proteins and a concomitant loss of LDs. On the one hand, LD breakdown has been suggested to provide the energy required for HSC activation into myofibroblast-like cells. On the other hand, this process could mitigate HSC activation following the transformation of released REs into retinoic acids (RAs), ligands for nuclear receptors exerting antifibrotic transcriptional regulatory activities in HSCs. Importantly, RAs may also constitute a means for HSCs to orchestrate the liver response to injury by triggering transcriptional effects in multiple additional surrounding liver cell populations. We envision that new approaches, such as single-cell technologies, will allow to better define how RAs are issued from LD loss in HSCs exert a multicellular control of the liver (patho)physiology.
Collapse
Affiliation(s)
- Marie Bobowski-Gerard
- Institut Pasteur de Lille, The University of Lille, Inserm, CHU Lille, U1011-EGID, F-59000 Lille, France.
| | - Francesco Paolo Zummo
- Institut Pasteur de Lille, The University of Lille, Inserm, CHU Lille, U1011-EGID, F-59000 Lille, France.
| | - Bart Staels
- Institut Pasteur de Lille, The University of Lille, Inserm, CHU Lille, U1011-EGID, F-59000 Lille, France.
| | - Philippe Lefebvre
- Institut Pasteur de Lille, The University of Lille, Inserm, CHU Lille, U1011-EGID, F-59000 Lille, France.
| | - Jérôme Eeckhoute
- Institut Pasteur de Lille, The University of Lille, Inserm, CHU Lille, U1011-EGID, F-59000 Lille, France.
| |
Collapse
|
205
|
Liver fibrosis: Pathophysiology, pathogenetic targets and clinical issues. Mol Aspects Med 2018; 65:37-55. [PMID: 30213667 DOI: 10.1016/j.mam.2018.09.002] [Citation(s) in RCA: 746] [Impact Index Per Article: 106.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Revised: 09/05/2018] [Accepted: 09/07/2018] [Indexed: 02/06/2023]
Abstract
The progression of chronic liver diseases (CLD), irrespective of etiology, involves chronic parenchymal injury, persistent activation of inflammatory response as well as sustained activation of liver fibrogenesis and wound healing response. Liver fibrogenesis, is a dynamic, highly integrated molecular, cellular and tissue process responsible for driving the excess accumulation of extracellular matrix (ECM) components (i.e., liver fibrosis) sustained by an eterogeneous population of hepatic myofibroblasts (MFs). The process of liver fibrogenesis recognizes a number of common and etiology-independent mechanisms and events but it is also significantly influenced by the specific etiology, as also reflected by peculiar morphological patterns of liver fibrosis development. In this review we will analyze the most relevant established and/or emerging pathophysiological issues underlying CLD progression with a focus on the role of critical hepatic cell populations, mechanisms and signaling pathways involved, as they represent potential therapeutic targets, to finally analyze selected and relevant clinical issues.
Collapse
|
206
|
Qu C, Zheng D, Li S, Liu Y, Lidofsky A, Holmes JA, Chen J, He L, Wei L, Liao Y, Yuan H, Jin Q, Lin Z, Hu Q, Jiang Y, Tu M, Chen X, Li W, Lin W, Fuchs BC, Chung RT, Hong A. Tyrosine kinase SYK is a potential therapeutic target for liver fibrosis. Hepatology 2018; 68:1125-1139. [PMID: 29537660 PMCID: PMC6138581 DOI: 10.1002/hep.29881] [Citation(s) in RCA: 75] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Revised: 02/28/2018] [Accepted: 03/09/2018] [Indexed: 12/15/2022]
Abstract
Spleen tyrosine kinase (SYK) plays a critical role in immune cell signaling pathways and has been reported as a biomarker for human hepatocellular carcinoma (HCC). We sought to investigate the mechanism by which SYK promotes liver fibrosis and to evaluate SYK as a therapeutic target for liver fibrosis. We evaluated the cellular localization of SYK and the association between SYK expression and liver fibrogenesis in normal, hepatitis B virus (HBV)-infected, hepatitis C virus (HCV)-infected and non-alcoholic steatohepatitis (NASH) liver tissue (n=36, 127, 22 and 30, respectively). A polymerase chain reaction (PCR) array was used to detect the changes in transcription factor (TF) expression in hepatic stellate cells (HSCs) with SYK knockdown. The effects of SYK antagonism on liver fibrogenesis were studied in LX-2 cells, TWNT-4 cells, primary human HSCs, and three progressive fibrosis/cirrhosis animal models, including a CCL4 mouse model, and diethylnitrosamine (DEN) and bile duct ligation (BDL) rat models. We found that SYK protein in HSCs and hepatocytes correlated positively with liver fibrosis stage in human liver tissue. HBV or HCV infection significantly increased SYK and cytokine expression in hepatocytes. Increasing cytokine production further induced SYK expression and fibrosis-related gene transcription in HSCs. Up-regulated SYK in HSCs promoted HSC activation by increasing the expression of specific TFs related to activation of HSCs. SYK antagonism effectively suppressed liver fibrosis via inhibition of HSC activation, and decreased obstructive jaundice and reduced HCC development in animal models. Conclusion: SYK promotes liver fibrosis via activation of HSCs and is an attractive potential therapeutic target for liver fibrosis and prevention of HCC development. (Hepatology 2018).
Collapse
Affiliation(s)
- Chen Qu
- Department of Abdominal Surgery, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong 510315, China
- Cancer Center, Southern Medical University, Guangzhou, Guangdong 510315, China
| | - Dandan Zheng
- Department of Abdominal Surgery, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong 510315, China
- Cancer Center, Southern Medical University, Guangzhou, Guangdong 510315, China
| | - Sai Li
- Department of Pharmacy, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong 510315, China
| | - Yingjun Liu
- Department of General Surgery, Affiliated Tumor Hospital of Zhengzhou University, Zhengzhou, Henan 450008, China
| | - Anna Lidofsky
- Gastrointestinal Unit, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Jacinta A. Holmes
- Gastrointestinal Unit, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Jianning Chen
- Department of Pathology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510630, China
| | - Lu He
- Department of Abdominal Surgery, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong 510315, China
- Cancer Center, Southern Medical University, Guangzhou, Guangdong 510315, China
| | - Lan Wei
- Division of Surgical Oncology, Massachusetts General Hospital Cancer Center and Harvard Medical School, Boston, MA
| | - Yadi Liao
- Department of Abdominal Surgery, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong 510315, China
- Cancer Center, Southern Medical University, Guangzhou, Guangdong 510315, China
| | - Hui Yuan
- Department of Abdominal Surgery, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong 510315, China
- Cancer Center, Southern Medical University, Guangzhou, Guangdong 510315, China
| | - Qimeng Jin
- Department of Abdominal Surgery, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong 510315, China
- Cancer Center, Southern Medical University, Guangzhou, Guangdong 510315, China
| | - Zelong Lin
- Department of Abdominal Surgery, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong 510315, China
- Cancer Center, Southern Medical University, Guangzhou, Guangdong 510315, China
| | - Qiaoting Hu
- Department of Abdominal Surgery, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong 510315, China
- Cancer Center, Southern Medical University, Guangzhou, Guangdong 510315, China
| | - Yuchuan Jiang
- Department of Abdominal Surgery, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong 510315, China
- Cancer Center, Southern Medical University, Guangzhou, Guangdong 510315, China
| | - Mengxian Tu
- Department of Abdominal Surgery, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong 510315, China
- Cancer Center, Southern Medical University, Guangzhou, Guangdong 510315, China
| | - Xijun Chen
- Department of Abdominal Surgery, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong 510315, China
- Cancer Center, Southern Medical University, Guangzhou, Guangdong 510315, China
| | - Weiming Li
- Department of Abdominal Surgery, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong 510315, China
- Cancer Center, Southern Medical University, Guangzhou, Guangdong 510315, China
| | - Wenyu Lin
- Gastrointestinal Unit, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Bryan C. Fuchs
- Division of Surgical Oncology, Massachusetts General Hospital Cancer Center and Harvard Medical School, Boston, MA
| | - Raymond T. Chung
- Gastrointestinal Unit, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - andJian Hong
- Department of Abdominal Surgery, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong 510315, China
- Cancer Center, Southern Medical University, Guangzhou, Guangdong 510315, China
- Gastrointestinal Unit, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| |
Collapse
|
207
|
Targeting Endothelial Erk1/2-Akt Axis as a Regeneration Strategy to Bypass Fibrosis during Chronic Liver Injury in Mice. Mol Ther 2018; 26:2779-2797. [PMID: 30266653 DOI: 10.1016/j.ymthe.2018.08.016] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Revised: 08/13/2018] [Accepted: 08/19/2018] [Indexed: 02/08/2023] Open
Abstract
Liver sinusoidal endothelial cells (LSECs) have great capacity for liver regeneration, and this capacity can easily switch to profibrotic phenotype, which is still poorly understood. In this study, we elucidated a potential target in LSECs for regenerative treatment that can bypass fibrosis during chronic liver injury. Proregenerative LSECs can be transformed to profibrotic phenotype after 4 weeks of carbon tetrachloride administration or 10 days of bile duct ligation. This phenotypic alternation of LSECs was mediated by extracellular regulated protein kinases 1 and 2 (Erk1/2)-Akt axis switch in LSECs during chronic liver injury; Erk1/2 was normally associated with maintenance of the LSEC proregenerative phenotype, inhibiting hepatic stellate cell (HSC) activation and promoting tissue repair by enhancing nitric oxide (NO)/reactive oxygen species (ROS) ratio and increasing expression of hepatic growth factor (HGF) and Wingless-type MMTV integration site family member 2 (Wnt2). Alternatively, Akt induced LSEC profibrotic phenotype, which mainly stimulated HSC activation and concomitant senescence by reducing NO/ROS ratio and decreasing HGF/Wnt2 expression. LSEC-targeted adenovirus or drug particle to promote Erk1/2 activity can alleviate liver fibrosis, accelerate fibrosis resolution, and enhance liver regeneration. This study demonstrated that the Erk1/2-Akt axis acted as a switch to regulate the proregenerative and profibrotic phenotypes of LSECs, and targeted therapy promoted liver regeneration while bypassing fibrosis, providing clues for a more effective treatment of liver diseases.
Collapse
|
208
|
Pakshir P, Hinz B. The big five in fibrosis: Macrophages, myofibroblasts, matrix, mechanics, and miscommunication. Matrix Biol 2018; 68-69:81-93. [DOI: 10.1016/j.matbio.2018.01.019] [Citation(s) in RCA: 162] [Impact Index Per Article: 23.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2018] [Revised: 01/25/2018] [Accepted: 01/28/2018] [Indexed: 02/07/2023]
|
209
|
Abstract
Stellate cells are resident lipid-storing cells of the pancreas and liver that transdifferentiate to a myofibroblastic state in the context of tissue injury. Beyond having roles in tissue homeostasis, stellate cells are increasingly implicated in pathological fibrogenic and inflammatory programs that contribute to tissue fibrosis and that constitute a growth-permissive tumor microenvironment. Although the capacity of stellate cells for extracellular matrix production and remodeling has long been appreciated, recent research efforts have demonstrated diverse roles for stellate cells in regulation of epithelial cell fate, immune modulation, and tissue health. Our present understanding of stellate cell biology in health and disease is discussed here, as are emerging means to target these multifaceted cells for therapeutic benefit.
Collapse
Affiliation(s)
- Mara H Sherman
- Department of Cell, Developmental & Cancer Biology, Oregon Health & Science University, Portland, Oregon 97201, USA;
| |
Collapse
|
210
|
Athwal VS, Pritchett J, Llewellyn J, Martin K, Camacho E, Raza SM, Phythian-Adams A, Birchall LJ, Mullan AF, Su K, Pearmain L, Dolman G, Zaitoun AM, Friedman SL, MacDonald A, Irving WL, Guha IN, Hanley NA, Piper Hanley K. SOX9 predicts progression toward cirrhosis in patients while its loss protects against liver fibrosis. EMBO Mol Med 2018; 9:1696-1710. [PMID: 29109128 PMCID: PMC5709769 DOI: 10.15252/emmm.201707860] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Fibrosis and organ failure is a common endpoint for many chronic liver diseases. Much is known about the upstream inflammatory mechanisms provoking fibrosis and downstream potential for tissue remodeling. However, less is known about the transcriptional regulation in vivo governing fibrotic matrix deposition by liver myofibroblasts. This gap in understanding has hampered molecular predictions of disease severity and clinical progression and restricted targets for antifibrotic drug development. In this study, we show the prevalence of SOX9 in biopsies from patients with chronic liver disease correlated with fibrosis severity and accurately predicted disease progression toward cirrhosis. Inactivation of Sox9 in mice protected against both parenchymal and biliary fibrosis, and improved liver function and ameliorated chronic inflammation. SOX9 was downstream of mechanosignaling factor, YAP1. These data demonstrate a role for SOX9 in liver fibrosis and open the way for the transcription factor and its dependent pathways as new diagnostic, prognostic, and therapeutic targets in patients with liver fibrosis.
Collapse
Affiliation(s)
- Varinder S Athwal
- Division of Diabetes, Endocrinology and Gastroenterology, Faculty of Biology, Medicine & Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK.,Research & Innovation Division, Central Manchester University Hospitals NHS Foundation Trust, Manchester, UK
| | - James Pritchett
- School of Healthcare Science, Manchester Metropolitan University, Manchester, UK
| | - Jessica Llewellyn
- Division of Diabetes, Endocrinology and Gastroenterology, Faculty of Biology, Medicine & Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK
| | - Katherine Martin
- Division of Diabetes, Endocrinology and Gastroenterology, Faculty of Biology, Medicine & Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK.,Research & Innovation Division, Central Manchester University Hospitals NHS Foundation Trust, Manchester, UK
| | - Elizabeth Camacho
- Centre for Health Economics, Institute of Population Health, Faculty of Medical & Human Sciences, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK
| | - Sayyid Ma Raza
- Division of Diabetes, Endocrinology and Gastroenterology, Faculty of Biology, Medicine & Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK.,Research & Innovation Division, Central Manchester University Hospitals NHS Foundation Trust, Manchester, UK
| | - Alexander Phythian-Adams
- Manchester Centre for Collaborative Inflammation Research, Faculty of Life Sciences, University of Manchester, Manchester, UK
| | - Lindsay J Birchall
- Division of Diabetes, Endocrinology and Gastroenterology, Faculty of Biology, Medicine & Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK.,Research & Innovation Division, Central Manchester University Hospitals NHS Foundation Trust, Manchester, UK
| | - Aoibheann F Mullan
- Division of Diabetes, Endocrinology and Gastroenterology, Faculty of Biology, Medicine & Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK.,Research & Innovation Division, Central Manchester University Hospitals NHS Foundation Trust, Manchester, UK
| | - Kim Su
- Division of Diabetes, Endocrinology and Gastroenterology, Faculty of Biology, Medicine & Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK.,Research & Innovation Division, Central Manchester University Hospitals NHS Foundation Trust, Manchester, UK
| | - Laurence Pearmain
- Division of Diabetes, Endocrinology and Gastroenterology, Faculty of Biology, Medicine & Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK.,Research & Innovation Division, Central Manchester University Hospitals NHS Foundation Trust, Manchester, UK
| | - Grace Dolman
- Nottingham Digestive Diseases Centre and National Institute for Health Research (NIHR) Nottingham Biomedical Research Centre, Nottingham University Hospitals NHS Trust and University of Nottingham, Nottingham, UK
| | - Abed M Zaitoun
- Department of Cellular Pathology, Nottingham Digestive Diseases Centre and National Institute of Health Research Biomedical Research Unit in Gastroenterology and Liver Disease, University of Nottingham and Nottingham University Hospitals NHS Trust, Nottingham, UK
| | - Scott L Friedman
- Division of Liver Diseases, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Andrew MacDonald
- Manchester Centre for Collaborative Inflammation Research, Faculty of Life Sciences, University of Manchester, Manchester, UK
| | - William L Irving
- Nottingham Digestive Diseases Centre and National Institute for Health Research (NIHR) Nottingham Biomedical Research Centre, Nottingham University Hospitals NHS Trust and University of Nottingham, Nottingham, UK.,School of Life Sciences, Nottingham Digestive Diseases Centre and National Institute of Health Research Biomedical Research Unit in Gastroenterology and Liver Disease, University of Nottingham and Nottingham University Hospitals NHS Trust, Nottingham, UK
| | - Indra N Guha
- Nottingham Digestive Diseases Centre and National Institute for Health Research (NIHR) Nottingham Biomedical Research Centre, Nottingham University Hospitals NHS Trust and University of Nottingham, Nottingham, UK
| | - Neil A Hanley
- Division of Diabetes, Endocrinology and Gastroenterology, Faculty of Biology, Medicine & Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK.,Research & Innovation Division, Central Manchester University Hospitals NHS Foundation Trust, Manchester, UK
| | - Karen Piper Hanley
- Division of Diabetes, Endocrinology and Gastroenterology, Faculty of Biology, Medicine & Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK .,Research & Innovation Division, Central Manchester University Hospitals NHS Foundation Trust, Manchester, UK
| |
Collapse
|
211
|
Weiskirchen R, Weiskirchen S, Tacke F. Organ and tissue fibrosis: Molecular signals, cellular mechanisms and translational implications. Mol Aspects Med 2018; 65:2-15. [PMID: 29958900 DOI: 10.1016/j.mam.2018.06.003] [Citation(s) in RCA: 385] [Impact Index Per Article: 55.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Accepted: 06/25/2018] [Indexed: 02/08/2023]
Abstract
Fibrosis denotes excessive scarring, which exceeds the normal wound healing response to injury in many tissues. Although the extracellular matrix deposition appears unstructured disrupting the normal tissue architecture and subsequently impairing proper organ function, fibrogenesis is a highly orchestrated process determined by defined sequences of molecular signals and cellular response mechanisms. Persistent injury and parenchymal cell death provokes tissue inflammation, macrophage activation and immune cell infiltration. The release of biologically highly active soluble mediators (alarmins, cytokines, chemokines) lead to the local activation of collagen producing mesenchymal cells such as pericytes, myofibroblasts or Gli1 positive mesenchymal stem cell-like cells, to a transition of various cell types into myofibroblasts as well as to the recruitment of fibroblast precursors. Clinical observations and experimental models highlighted that fibrosis is not a one-way road. Specific mechanistic principles of fibrosis regression involve the resolution of chronic tissue injury, the shift of inflammatory processes towards recovery, deactivation of myofibroblasts and finally fibrolysis of excess matrix scaffold. The thorough understanding of common principles of fibrogenic molecular signals and cellular mechanisms in various organs - such as liver, kidney, lung, heart or skin - is the basis for developing improved diagnostics including biomarkers or imaging techniques and novel antifibrotic therapeutics.
Collapse
Affiliation(s)
- Ralf Weiskirchen
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry, RWTH University Hospital Aachen, Germany
| | - Sabine Weiskirchen
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry, RWTH University Hospital Aachen, Germany
| | - Frank Tacke
- Dept. of Medicine III, University Hospital Aachen, Germany.
| |
Collapse
|
212
|
Talal AH, Venuto CS, Younis I. Assessment of Hepatic Impairment and Implications for Pharmacokinetics of Substance Use Treatment. Clin Pharmacol Drug Dev 2018; 6:206-212. [PMID: 28263464 DOI: 10.1002/cpdd.336] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Accepted: 12/30/2016] [Indexed: 12/12/2022]
Abstract
Although the liver is the primary site of metabolism and biliary excretion for many medications, data are limited on the liver's pharmacokinetic abilities in cirrhosis. Cirrhosis develops through collagen deposition, eventually culminating in end-stage liver disease that compromises hepatic drug metabolism. Consequently, the US Food and Drug Administration (FDA) recommends evaluating the pharmacokinetics of medications in subjects with hepatic impairment if hepatic metabolism constitutes more than 20% of their elimination or if they have a narrow therapeutic range. A variety of noninvasive indices and radiologic procedures can be employed to assess hepatic drug metabolism and excretion. The Child-Pugh score is the most commonly used scale for assessing hepatic impairment among drugs submitted for US FDA approval. The score, originally developed to guide operative mortality in patients undergoing hepatic resection, has not been modified since its inception 5 decades ago. Furthermore, the score was not originally intended to be a guide for potential dose modification in patients with hepatic impairment. These reasons, in combination with the availability of a variety of new imaging modalities and an enhanced understanding of hepatic biology, should foster the development of novel methods to assess the effect of hepatic impairment on liver drug metabolism.
Collapse
Affiliation(s)
- Andrew H Talal
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, University at Buffalo, Buffalo, NY, USA
| | - Charles S Venuto
- Center for Human Experimental Therapeutics, University of Rochester, Rochester, NY, USA.,AIDS Clinical Trials Group Pharmacology Specialty Laboratory, New York State Center of Excellence in Bioinformatics and Life Sciences; School of Pharmacy and Pharmaceutical Sciences, University at Buffalo, Buffalo, NY, USA
| | - Islam Younis
- Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, MD, USA
| |
Collapse
|
213
|
Pan XY, Yang Y, Meng HW, Li HD, Chen X, Huang HM, Bu FT, Yu HX, Wang Q, Huang C, Meng XM, Li J. DNA Methylation of PTGIS Enhances Hepatic Stellate Cells Activation and Liver Fibrogenesis. Front Pharmacol 2018; 9:553. [PMID: 29892223 PMCID: PMC5985735 DOI: 10.3389/fphar.2018.00553] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Accepted: 05/08/2018] [Indexed: 12/20/2022] Open
Abstract
The activation of hepatic stellate cells (HSCs) is a central event in the progression of liver fibrosis. Multiple studies proved that DNA methylation might accelerate HSCs activation. However, the specific pathogenesis of liver fibrosis remains not fully addressed. Our laboratory performed Genome methylation screening to find out the methylated gene in mice with liver fibrosis. The pilot experiments showed that the promoter of prostacyclin synthase (PTGIS) gene was hypermethylated in CCl4-induced liver fibrosis mouse model. Moreover, the down-regulated PTGIS expression can be restored by DNMTs-RNAi and 5-aza-2-deoxycytidine (5-azadC), an inhibitor of DNA methyltransferase (DNMTs). Methylation-specific PCR (MSP) showed that the methylation status of PTGIS in HSC-T6 cells cultures with TGF-β1 (10 ng/mL) was elevated compared with control group. Chromatin immunoprecipitation (ChIP) assay indicated that PTGIS methylation was mainly induced by DNMT1 and DNMT3b. We further investigated the function of PTGIS in liver fibrosis by Recombinant Hepatic-adeno-associated virus (rAAV8)-PTGIS overexpression. The data indicated that overexpression of PTGIS in mouse liver accompanied by elevated apoptosis-related proteins expression in primary HSCs. Conversely, PTGIS silencing mediated by RNAi enhanced the expression of α-SMA and COL1a1 in vitro. Those results illustrated that adding PTGIS expression inhibits the activation of HSCs and alleviates liver fibrosis. Therefore, our study unveils the role of PTGIS in HSCs activation, which may provide a possible explanation for CCl4-mediated liver fibrosis.
Collapse
Affiliation(s)
- Xue-Yin Pan
- The Key Laboratory of Major Autoimmune Diseases, Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China.,The Key Laboratory of Anti-inflammatory of Immune Medicine, Ministry of Education, Hefei, China.,Institute for Liver Diseases of Anhui Medical University, Hefei, China
| | - Yang Yang
- The Key Laboratory of Major Autoimmune Diseases, Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China.,The Key Laboratory of Anti-inflammatory of Immune Medicine, Ministry of Education, Hefei, China.,Institute for Liver Diseases of Anhui Medical University, Hefei, China
| | - Hong-Wu Meng
- The Key Laboratory of Major Autoimmune Diseases, Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China.,The Key Laboratory of Anti-inflammatory of Immune Medicine, Ministry of Education, Hefei, China.,Institute for Liver Diseases of Anhui Medical University, Hefei, China
| | - Hai-di Li
- The Key Laboratory of Major Autoimmune Diseases, Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China.,The Key Laboratory of Anti-inflammatory of Immune Medicine, Ministry of Education, Hefei, China.,Institute for Liver Diseases of Anhui Medical University, Hefei, China
| | - Xin Chen
- The Key Laboratory of Major Autoimmune Diseases, Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China.,The Key Laboratory of Anti-inflammatory of Immune Medicine, Ministry of Education, Hefei, China.,Institute for Liver Diseases of Anhui Medical University, Hefei, China
| | - Hui-Min Huang
- The Key Laboratory of Major Autoimmune Diseases, Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China.,The Key Laboratory of Anti-inflammatory of Immune Medicine, Ministry of Education, Hefei, China.,Institute for Liver Diseases of Anhui Medical University, Hefei, China
| | - Fang-Tian Bu
- The Key Laboratory of Major Autoimmune Diseases, Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China.,The Key Laboratory of Anti-inflammatory of Immune Medicine, Ministry of Education, Hefei, China.,Institute for Liver Diseases of Anhui Medical University, Hefei, China
| | - Hai-Xia Yu
- The Key Laboratory of Major Autoimmune Diseases, Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China.,The Key Laboratory of Anti-inflammatory of Immune Medicine, Ministry of Education, Hefei, China.,Institute for Liver Diseases of Anhui Medical University, Hefei, China
| | - Qin Wang
- The Key Laboratory of Major Autoimmune Diseases, Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China.,The Key Laboratory of Anti-inflammatory of Immune Medicine, Ministry of Education, Hefei, China.,Institute for Liver Diseases of Anhui Medical University, Hefei, China
| | - Cheng Huang
- The Key Laboratory of Major Autoimmune Diseases, Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China.,The Key Laboratory of Anti-inflammatory of Immune Medicine, Ministry of Education, Hefei, China.,Institute for Liver Diseases of Anhui Medical University, Hefei, China
| | - Xiao-Ming Meng
- The Key Laboratory of Major Autoimmune Diseases, Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China.,The Key Laboratory of Anti-inflammatory of Immune Medicine, Ministry of Education, Hefei, China.,Institute for Liver Diseases of Anhui Medical University, Hefei, China
| | - Jun Li
- The Key Laboratory of Major Autoimmune Diseases, Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China.,The Key Laboratory of Anti-inflammatory of Immune Medicine, Ministry of Education, Hefei, China.,Institute for Liver Diseases of Anhui Medical University, Hefei, China
| |
Collapse
|
214
|
Abdulkhaleq LA, Assi MA, Abdullah R, Zamri-Saad M, Taufiq-Yap YH, Hezmee MNM. The crucial roles of inflammatory mediators in inflammation: A review. Vet World 2018; 11:627-635. [PMID: 29915501 PMCID: PMC5993766 DOI: 10.14202/vetworld.2018.627-635] [Citation(s) in RCA: 370] [Impact Index Per Article: 52.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Accepted: 04/10/2018] [Indexed: 12/12/2022] Open
Abstract
The inflammatory response is a crucial aspect of the tissues' responses to deleterious inflammogens. This complex response involves leukocytes cells such as macrophages, neutrophils, and lymphocytes, also known as inflammatory cells. In response to the inflammatory process, these cells release specialized substances which include vasoactive amines and peptides, eicosanoids, proinflammatory cytokines, and acute-phase proteins, which mediate the inflammatory process by preventing further tissue damage and ultimately resulting in healing and restoration of tissue function. This review discusses the role of the inflammatory cells as well as their by-products in the mediation of inflammatory process. A brief insight into the role of natural anti-inflammatory agents is also discussed. The significance of this study is to explore further and understand the potential mechanism of inflammatory processes to take full advantage of vast and advanced anti-inflammatory therapies. This review aimed to reemphasize the importance on the knowledge of inflammatory processes with the addition of newest and current issues pertaining to this phenomenon.
Collapse
Affiliation(s)
- L. A. Abdulkhaleq
- Department of Pathology and Poultry Diseases, Faculty of Veterinary Medicine, Baghdad University, Baghdad, Iraq
- Department of Veterinary Pathology and Microbiology, Faculty of Veterinary Medicine, Universiti Putra Malaysia, Malaysia
| | - M. A. Assi
- Department of Community Health, College of Health and Medical Techniques, Al-Furat Al-Awsat Technical University, Iraq
- Department of Veterinary Preclinical Sciences, Faculty of Veterinary Medicine, Universiti Putra Malaysia, Malaysia
| | - Rasedee Abdullah
- Department of Veterinary Laboratory Diagnostics, Faculty of Veterinary Medicine, Universiti Putra Malaysia, Malaysia
| | - M. Zamri-Saad
- Department of Veterinary Laboratory Diagnostics, Faculty of Veterinary Medicine, Universiti Putra Malaysia, Malaysia
| | - Y. H. Taufiq-Yap
- Department of Chemistry, Faculty of Sains, Universiti Putra Malaysia, Malaysia
| | - M. N. M. Hezmee
- Department of Veterinary Preclinical Sciences, Faculty of Veterinary Medicine, Universiti Putra Malaysia, Malaysia
| |
Collapse
|
215
|
El-Lakkany NM, El-Maadawy WH, Seif El-Din SH, Saleh S, Safar MM, Ezzat SM, Mohamed SH, Botros SS, Demerdash Z, Hammam OA. Antifibrotic effects of gallic acid on hepatic stellate cells: In vitro and in vivo mechanistic study. J Tradit Complement Med 2018; 9:45-53. [PMID: 30671365 PMCID: PMC6335492 DOI: 10.1016/j.jtcme.2018.01.010] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Revised: 01/12/2018] [Accepted: 01/14/2018] [Indexed: 02/07/2023] Open
Abstract
Few studies reported the antifibrotic effects of gallic acid (GA) despite its known hepatoprotective and antioxidant activities. Accordingly, this study investigated the antifibrotic effects of GA through clarifying its mechanisms on hepatic stellate cells' (HSCs) activation, proliferation and/or apoptosis. In vitro effects of GA on HSC-T6 activation/proliferation, morphology and safety on hepatocytes were assessed. In vivo, hepatic fibrosis was induced via chronic thioacetamide (TAA)-intoxication. TAA-intoxicated rats were treated with silyamrin or GA. At end of experiment, liver functions, hepatic MDA, GSH, PDGF-BB, TGF-β1, TIMP-1 and hydroxyproline were determined. Histological analysis and Sirius red staining of hepatic sections, expressions of alpha-smooth muscle actin (α-SMA), proliferating cellular nuclear antigen (PCNA) and caspase-3 were examined. In vitro, GA resulted in a concentration and time-dependent inhibition in HSCs activation, proliferation (IC50= 45 and 19 μg/mL at 24 and 48 h respectively); restored the quiescent morphology of some activated HSCs plus its safety on hepatocytes. In vivo, GA reduced ALT, AST, MDA, PDGF-BB levels, collagen deposition and fibrosis score (S1 vs S4); increased caspase-3 expression and restored GSH stores, TGF-β1 level, α-SMA and PCNA expressions. In conclusion, GA counteracted the progression of hepatic fibrosis through reduction of HSCs proliferation/activation mutually with their apoptosis induction.
Collapse
Affiliation(s)
- Naglaa M El-Lakkany
- Department of Pharmacology, Theodor Bilharz Research Institute, Warak El-Hadar, Imbaba P.O. Box 30, Giza 12411, Egypt
| | - Walaa H El-Maadawy
- Department of Pharmacology, Theodor Bilharz Research Institute, Warak El-Hadar, Imbaba P.O. Box 30, Giza 12411, Egypt
| | - Sayed H Seif El-Din
- Department of Pharmacology, Theodor Bilharz Research Institute, Warak El-Hadar, Imbaba P.O. Box 30, Giza 12411, Egypt
| | - Samira Saleh
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo 11562, Egypt
| | - Marwa M Safar
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo 11562, Egypt.,Department of Pharmacology and Biochemistry, Faculty of Pharmacy, The British University in Egypt, Suez Desert Road, P.O. Box 43, ElSherouk City, Cairo 11837, Egypt
| | - Shahira M Ezzat
- Department of Pharmacognosy, Faculty of Pharmacy, Cairo University, Cairo 11562, Egypt.,Department of Pharmacognosy, Faculty of Pharmacy, October University for Modern Sciences and Arts (MSA), 6th of October, Giza 12566, Egypt
| | - Salwa H Mohamed
- Department of Immunology, Theodor Bilharz Research Institute, Warak El-Hadar, Imbaba P.O. Box 30, Giza 12411, Egypt
| | - Sanaa S Botros
- Department of Pharmacology, Theodor Bilharz Research Institute, Warak El-Hadar, Imbaba P.O. Box 30, Giza 12411, Egypt
| | - Zeinab Demerdash
- Department of Immunology, Theodor Bilharz Research Institute, Warak El-Hadar, Imbaba P.O. Box 30, Giza 12411, Egypt
| | - Olfat A Hammam
- Department of Pathology, Theodor Bilharz Research Institute, Warak El-Hadar, Imbaba P.O. Box 30, Giza 12411, Egypt
| |
Collapse
|
216
|
Altered AKAP12 expression in portal fibroblasts and liver sinusoids mediates transition from hepatic fibrogenesis to fibrosis resolution. Exp Mol Med 2018; 50:1-13. [PMID: 29700280 PMCID: PMC5938025 DOI: 10.1038/s12276-018-0074-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Revised: 02/20/2018] [Accepted: 02/22/2018] [Indexed: 12/11/2022] Open
Abstract
Liver fibrosis can be reversed by removing its causative injuries; however, the molecular mechanisms mediating the resolution of liver fibrogenesis are poorly understood. We investigate the role of a scaffold protein, A-Kinase Anchoring Protein 12 (AKAP12), during liver fibrosis onset, and resolution. Biliary fibrogenesis and fibrosis resolution was induced in wild-type (WT) or AKAP12-deficient C57BL/6 mice through different feeding regimens with 0.1% 3,5-diethoxycarbonyl-1,4-dihydrocollidine (DDC)-containing chow. AKAP12 expression in portal fibroblasts (PFs) and liver sinusoidal endothelial cells (LSECs) gradually decreased as fibrosis progressed but was restored after cessation of the fibrotic challenge. Histological analysis of human liver specimens with varying degrees of fibrosis of different etiologies revealed that AKAP12 expression diminishes in hepatic fibrosis from its early stages onward. AKAP12 KO mice displayed reduced fibrosis resolution in a DDC-induced biliary fibrosis model, which was accompanied by impaired normalization of myofibroblasts and capillarized sinusoids. RNA sequencing of the liver transcriptome revealed that genes related to ECM accumulation and vascular remodeling were mostly elevated in AKAP12 KO samples. Gene ontology (GO) and bioinformatic pathway analyses identified that the differentially expressed genes were significantly enriched in GO categories and pathways, such as the adenosine 3′,5′-cyclic monophosphate (cAMP) pathway. Knockdown of the AKAP12 gene in cultured primary PFs revealed that AKAP12 inhibited PF activation in association with the adenosine 3′,5′-cyclic monophosphate (cAMP) pathway. Moreover, AKAP12 knockdown in LSECs led to enhanced angiogenesis, endothelin-1 expression and alterations in laminin composition. Collectively, this study demonstrates that AKAP12-mediated regulation of PFs and LSECs has a central role in resolving hepatic fibrosis. A scaffolding protein that modulates cell signaling pathways contributes to reverse liver scarring. Liver fibrosis is caused by a build-up of scar tissue that interferes with liver function. However, the damage is reversed when the cause of injury is removed. Kyu-Won Kim at Seoul National University, South Korea, and colleagues examined the levels of A-Kinase Anchoring Protein 12 (AKAP12), a scaffolding protein that regulates the subcellular location of signaling proteins, in mouse and human livers. Levels of AKAP12 were reduced in fibrotic livers but restored when fibrosis was reversed. Mice lacking AKAP12 were unable to effectively repair the damage caused by fibrosis. Genetic analyses suggest that AKAP12 stimulates signaling through the adenosine 3′,5′-cyclic monophosphate (cAMP) pathway, which can inhibit fibrosis. These findings highlight a key role for AKAP12 in accelerating liver recovery.
Collapse
|
217
|
Anuja GI, Shine VJ, Latha PG, Suja SR. Protective effect of ethyl acetate fraction of Drynaria quercifolia against CCl 4 induced rat liver fibrosis via Nrf2/ARE and NFκB signalling pathway. JOURNAL OF ETHNOPHARMACOLOGY 2018; 216:79-88. [PMID: 29174446 DOI: 10.1016/j.jep.2017.11.015] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/08/2017] [Revised: 10/30/2017] [Accepted: 11/11/2017] [Indexed: 06/07/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Drynaria quercifolia rhizome is traditionally used as hepatoprotective drug especially in chronic jaundice. AIM OF THE STUDY The present study was undertaken to scientifically evaluate the efficacy of D. quercifolia rhizome against liver fibrosis. MATERIALS AND METHODS D. quercifolia rhizome crude extract (DQ) and its fractions of hexane (HDQ), ethyl acetate (EDQ), butanol (BDQ) were evaluated in vitro using primary hepatocytes and RAW 264.7 cells. In vivo anti-liver fibrotic activity of EDQ was assessed using CCl4 induced liver fibrosis in Wistar rats and serum biochemical parameters (AST, ALT, ALP, SB, cholesterol), MDA, PT, INR, GSH, SOD, CAT, liver glycogen, serum albumin levels were monitored. qRT-PCR analysis of TNF-α, COX-2, iNOS were performed. ELISA method was used to estimate TNF-α, COX-1 & 2. Histopathological studies like H & E, Masson's trichrome, immunohistochemistry staining for α-SMA, TIMP-1, Nrf2 were conducted. LC-Q-TOF-MS analysis of EDQ was conducted. RESULTS In vitro activity guided fractionation of D. quercifolia revealed EDQ as active fraction when compared to other extracts. EDQ treatment significantly inhibited the expression of α-SMA, TIMP-1, COX-2, TNF-α, iNOS and increased the levels of Nrf2 in rat liver fibrosis. LC-Q-TOF-MS analysis of EDQ confirmed the presence of naringin and naringenin. CONCLUSION The anti-liver fibrotic activity of EDQ is via inhibition of NFκB signalling pathway, antioxidant response through Nrf2 activation and further inhibition of HSC activation.
Collapse
Affiliation(s)
- G I Anuja
- Ethnomedicine and Ethnopharmacology Division, Jawaharlal Nehru Tropical Botanic Garden and Research Institute, Palode, Thiruvananthapuram 695562, Kerela, India.
| | - V J Shine
- Ethnomedicine and Ethnopharmacology Division, Jawaharlal Nehru Tropical Botanic Garden and Research Institute, Palode, Thiruvananthapuram 695562, Kerela, India
| | - P G Latha
- Ethnomedicine and Ethnopharmacology Division, Jawaharlal Nehru Tropical Botanic Garden and Research Institute, Palode, Thiruvananthapuram 695562, Kerela, India
| | - S R Suja
- Ethnomedicine and Ethnopharmacology Division, Jawaharlal Nehru Tropical Botanic Garden and Research Institute, Palode, Thiruvananthapuram 695562, Kerela, India
| |
Collapse
|
218
|
Shi WP, Ju D, Li H, Yuan L, Cui J, Luo D, Chen ZN, Bian H. CD147 Promotes CXCL1 Expression and Modulates Liver Fibrogenesis. Int J Mol Sci 2018; 19:ijms19041145. [PMID: 29642635 PMCID: PMC5979418 DOI: 10.3390/ijms19041145] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2018] [Revised: 03/18/2018] [Accepted: 04/03/2018] [Indexed: 12/30/2022] Open
Abstract
Activated hepatic stellate cells (HSCs) release pro-inflammatory and pro-fibrogenic factors. CXC chemokine-ligand-1 (CXCL1) is expressed on HSCs. We previously found that the CD147 is overexpressed in activated HSCs. In this study, we showed an important role of CD147 in promoting liver fibrosis by activating HSCs and upregulating expression of chemokines. Specifically, we found that CD147 specific deletion in HSCs mice alleviated CCl4-induced liver fibrosis and inhibited HSCs activation. Overexpression of CD147 upregulated the secretion of CXCL1. Meanwhile, CXCL1 promoted HSCs activation through autocrine. Treating with PI3K/AKT inhibitor could effectively suppress CD147-induced CXCL1 expression. Taken together, these findings suggest that CD147 regulates CXCL1 release in HSCs by PI3K/AKT signaling. Inhibition of CD147 attenuates CCl4-induced liver fibrosis and inflammation. Therefore, administration of targeting CD147 could be a promising therapeutic strategy in liver fibrosis.
Collapse
Affiliation(s)
- Wen-Pu Shi
- Department of Cell Biology, National Translational Science Center for Molecular Medicine, State Key Laboratory of Cancer Biology, Fourth Military Medical University, Xi'an 710032, China.
| | - Di Ju
- Department of Physiology, Basic Medical College, Shaanxi University of Chinese Medicine, Xianyang 712046, China.
| | - Hao Li
- Department of Cell Biology, National Translational Science Center for Molecular Medicine, State Key Laboratory of Cancer Biology, Fourth Military Medical University, Xi'an 710032, China.
| | - Lin Yuan
- Clinical Laboratory, No. 457 Hospital of PLA, Wuhan 430000, China.
| | - Jian Cui
- Department of Cell Biology, National Translational Science Center for Molecular Medicine, State Key Laboratory of Cancer Biology, Fourth Military Medical University, Xi'an 710032, China.
| | - Dan Luo
- Department of Cell Biology, National Translational Science Center for Molecular Medicine, State Key Laboratory of Cancer Biology, Fourth Military Medical University, Xi'an 710032, China.
| | - Zhi-Nan Chen
- Department of Cell Biology, National Translational Science Center for Molecular Medicine, State Key Laboratory of Cancer Biology, Fourth Military Medical University, Xi'an 710032, China.
| | - Huijie Bian
- Department of Cell Biology, National Translational Science Center for Molecular Medicine, State Key Laboratory of Cancer Biology, Fourth Military Medical University, Xi'an 710032, China.
| |
Collapse
|
219
|
Ramzy MM, Abdelghany HM, Zenhom NM, El-Tahawy NF. Effect of histone deacetylase inhibitor on epithelial-mesenchymal transition of liver fibrosis. IUBMB Life 2018; 70:511-518. [DOI: 10.1002/iub.1742] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Revised: 03/02/2018] [Accepted: 03/05/2018] [Indexed: 01/04/2023]
Affiliation(s)
- Maggie M. Ramzy
- Department of Biochemistry, Faculty of Medicine; Minia University; Egypt
| | - Hend M Abdelghany
- Department of Biochemistry, Faculty of Medicine; Minia University; Egypt
| | - Nagwa M. Zenhom
- Department of Biochemistry, Faculty of Medicine; Minia University; Egypt
| | - Nashwa F. El-Tahawy
- Department of Histology and Cell Biology, Faculty of Medicine; Minia University; Egypt
| |
Collapse
|
220
|
Ding D, Chen LL, Zhai YZ, Hou CJ, Tao LL, Lu SH, Wu J, Liu XP. Trichostatin A inhibits the activation of Hepatic stellate cells by Increasing C/EBP-α Acetylation in vivo and in vitro. Sci Rep 2018. [PMID: 29535398 PMCID: PMC5849734 DOI: 10.1038/s41598-018-22662-6] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Reversal of activated hepatic stellate cells (HSCs) to a quiescent state and apoptosis of activated HSCs are key elements in the reversion of hepatic fibrosis. CCAAT/enhancer binding protein α (C/EBP-α) has been shown to inhibit HSC activation and promote its apoptosis. This study aims to investigate how C/EBP-α acetylation affects the fate of activated HSCs. Effects of a histone deacetylation inhibitor trichostatin A (TSA) on HSC activation were evaluated in a mouse model of liver fibrosis caused by carbon tetrachloride (CCl4) intoxication. TSA was found to ameliorate CCl4-induced hepatic fibrosis and improve liver function through increasing the protein level and enhancing C/EBP-α acetylation in the mouse liver. C/EBP-α acetylation was determined in HSC lines in the presence or absence of TSA, and the lysine residue K276 was identified as a main acetylation site in C/EBP-α protein. C/EBP-α acetylation increased its stability and protein level, and inhibited HSC activation. The present study demonstrated that C/EBP-α acetylation increases the protein level by inhibiting its ubiquitination-mediated degradation, and may be involved in the fate of activated HSCs. Use of TSA may confer an option in minimizing hepatic fibrosis by suppressing HSC activation, a key process in the initiation and progression of hepatic fibrosis.
Collapse
Affiliation(s)
- Di Ding
- Department of Pathology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Lin-Lin Chen
- Department of Pathology, The Fifth People's Hospital, Fudan University, Shanghai, 200040, China
| | - Ying-Zhen Zhai
- Department of Pathology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Chen-Jian Hou
- Department of Pathology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Li-Li Tao
- Department of Pathology, Peking University, Shenzhen Hospital, Shenzhen, 518036, China
| | - Shu-Han Lu
- Department of Nutrition, University of California at Davis, Davis, California, USA
| | - Jian Wu
- Department of Medical Microbiology, Key Laboratory of Molecular Virology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China. .,Shanghai Institute of Liver Disease, Fudan University, Shanghai, 200032, China.
| | - Xiu-Ping Liu
- Department of Pathology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China. .,Department of Pathology, The Fifth People's Hospital, Fudan University, Shanghai, 200040, China.
| |
Collapse
|
221
|
Zhou Y, Horowitz JC, Naba A, Ambalavanan N, Atabai K, Balestrini J, Bitterman PB, Corley RA, Ding BS, Engler AJ, Hansen KC, Hagood JS, Kheradmand F, Lin QS, Neptune E, Niklason L, Ortiz LA, Parks WC, Tschumperlin DJ, White ES, Chapman HA, Thannickal VJ. Extracellular matrix in lung development, homeostasis and disease. Matrix Biol 2018. [PMID: 29524630 DOI: 10.1016/j.matbio.2018.03.005] [Citation(s) in RCA: 194] [Impact Index Per Article: 27.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The lung's unique extracellular matrix (ECM), while providing structural support for cells, is critical in the regulation of developmental organogenesis, homeostasis and injury-repair responses. The ECM, via biochemical or biomechanical cues, regulates diverse cell functions, fate and phenotype. The composition and function of lung ECM become markedly deranged in pathological tissue remodeling. ECM-based therapeutics and bioengineering approaches represent promising novel strategies for regeneration/repair of the lung and treatment of chronic lung diseases. In this review, we assess the current state of lung ECM biology, including fundamental advances in ECM composition, dynamics, topography, and biomechanics; the role of the ECM in normal and aberrant lung development, adult lung diseases and autoimmunity; and ECM in the regulation of the stem cell niche. We identify opportunities to advance the field of lung ECM biology and provide a set recommendations for research priorities to advance knowledge that would inform novel approaches to the pathogenesis, diagnosis, and treatment of chronic lung diseases.
Collapse
Affiliation(s)
- Yong Zhou
- Division of Pulmonary, Allergy and Critical Care Medicine, University of Alabama at Birmingham, United States.
| | - Jeffrey C Horowitz
- Division of Pulmonary and Critical Care Medicine, University of Michigan, United States.
| | - Alexandra Naba
- Department of Physiology & Biophysics, University of Illinois at Chicago, United States.
| | | | - Kamran Atabai
- Lung Biology Center, University of California, San Francisco, United States.
| | | | | | - Richard A Corley
- Systems Toxicology & Exposure Science, Pacific Northwest National Laboratory, United States.
| | - Bi-Sen Ding
- Weill Cornell Medical College, United States.
| | - Adam J Engler
- Sanford Consortium for Regenerative Medicine, University of California, San Diego, United States.
| | - Kirk C Hansen
- Biochemistry & Molecular Genetics, University of Colorado Denver, United States.
| | - James S Hagood
- Pediatric Respiratory Medicine, University of California San Diego, United States.
| | - Farrah Kheradmand
- Division of Pulmonary and Critical Care, Baylor College of Medicine, United States.
| | - Qing S Lin
- Division of Lung Diseases, National Heart, Lung, and Blood Institute, United States.
| | - Enid Neptune
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins School of Medicine, United States.
| | - Laura Niklason
- Department of Anesthesiology, Yale University, United States.
| | - Luis A Ortiz
- Division of Environmental and Occupational Health, University of Pittsburgh, United States.
| | - William C Parks
- Department of Medicine, Cedars-Sinai Medical Center, United States.
| | - Daniel J Tschumperlin
- Department of Physiology & Biomedical Engineering, Mayo Clinic College of Medicine, United States.
| | - Eric S White
- Division of Pulmonary and Critical Care Medicine, University of Michigan, United States.
| | - Harold A Chapman
- Division of Pulmonary and Critical Care Medicine, University of California, San Francisco, United States.
| | - Victor J Thannickal
- Division of Pulmonary, Allergy and Critical Care Medicine, University of Alabama at Birmingham, United States.
| |
Collapse
|
222
|
Gajendiran P, Vega LI, Itoh K, Sesaki H, Vakili MR, Lavasanifar A, Hong K, Mezey E, Ganapathy-Kanniappan S. Elevated mitochondrial activity distinguishes fibrogenic hepatic stellate cells and sensitizes for selective inhibition by mitotropic doxorubicin. J Cell Mol Med 2018; 22:2210-2219. [PMID: 29397578 PMCID: PMC5867155 DOI: 10.1111/jcmm.13501] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Accepted: 11/18/2017] [Indexed: 12/17/2022] Open
Abstract
Activation of hepatic stellate cells (HSCs) is an integral component of the wound‐healing process in liver injury/inflammation. However, uncontrolled activation of HSCs leads to constant secretion of collagen‐rich extracellular matrix (ECM) proteins, resulting in liver fibrosis. The enhanced ECM synthesis/secretion demands an uninterrupted supply of intracellular energy; however, there is a paucity of data on the bioenergetics, particularly the mitochondrial (mito) metabolism of fibrogenic HSCs. Here, using human and rat HSCs in vitro, we show that the mito‐respiration, mito‐membrane potential (Δψm) and cellular ‘bioenergetic signature’ distinguish fibrogenic HSCs from normal, less‐active HSCs. Ex vivo, HSCs from mouse and rat models of liver fibrosis further confirmed the altered ‘bioenergetic signature’ of fibrogenic HSCs. Importantly, the distinctive elevation in mito‐Δψm sensitized fibrogenic HSCs for selective inhibition by mitotropic doxorubicin while normal, less‐active HSCs and healthy human primary hepatocytes remained minimally affected if not, unaffected. Thus, the increased mito‐Δψm may provide an opportunity to selectively target fibrogenic HSCs in liver fibrosis.
Collapse
Affiliation(s)
- Priya Gajendiran
- Division of Interventional Radiology, Russell H. Morgan Department of Radiology & Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Leonel Iglesias Vega
- Division of Interventional Radiology, Russell H. Morgan Department of Radiology & Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Kie Itoh
- Department of Cell Biology, School of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Hiromi Sesaki
- Department of Cell Biology, School of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Mohammad Reza Vakili
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Canada
| | - Afsaneh Lavasanifar
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Canada
| | - Kelvin Hong
- Division of Interventional Radiology, Russell H. Morgan Department of Radiology & Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Esteban Mezey
- Division of Gastroenterology and Hepatology, Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Shanmugasundaram Ganapathy-Kanniappan
- Division of Interventional Radiology, Russell H. Morgan Department of Radiology & Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
223
|
Schuppan D, Surabattula R, Wang XY. Determinants of fibrosis progression and regression in NASH. J Hepatol 2018; 68:238-250. [PMID: 29154966 DOI: 10.1016/j.jhep.2017.11.012] [Citation(s) in RCA: 360] [Impact Index Per Article: 51.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Revised: 11/02/2017] [Accepted: 11/09/2017] [Indexed: 02/06/2023]
Abstract
Cirrhosis has become the major liver-related clinical endpoint in non-alcoholic steatohepatitis (NASH). However, progression to cirrhosis is less predictable in NASH than in other chronic liver diseases. This is due to the complex and multifactorial aetiology of NASH, which is determined by lifestyle and nutrition, multiple genetic and epigenetic factors, and a prominent role of hepatic and extrahepatic comorbidities. Thus, modest changes in these cofactors can also induce fibrosis regression, at least in patients with precirrhotic liver disease. Fibrogenesis in NASH correlates with, but is indirectly coupled to, classical inflammation, since fibrosis progression is driven by repetitive periods of repair. While hepatocyte lipoapoptosis is a key driving force of fibrosis progression, activated hepatic stellate cells, myofibroblasts, cholangiocytes, macrophages and components of the pathological extracellular matrix are major fibrogenic effectors and thus pharmacological targets for therapies aimed at inhibition of fibrosis progression or induction of fibrosis reversal. The advent of novel, highly sensitive and specific serum biomarkers and imaging methods to assess the dynamics of liver fibrosis in NASH will improve detection, stratification and follow-up of patients with progressive NASH . These non-invasive tools will also promote the clinical development of antifibrotic drugs, by permitting the design of lean proof-of-concept studies, and enabling development of a personalised antifibrotic therapy for patients with rapid fibrosis progression or advanced disease.
Collapse
Affiliation(s)
- Detlef Schuppan
- Institute of Translational Immunology and Research Center for Immunotherapy, University of Mainz Medical Center, Mainz, Germany; Division of Gastroenterology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, USA.
| | - Rambabu Surabattula
- Institute of Translational Immunology and Research Center for Immunotherapy, University of Mainz Medical Center, Mainz, Germany
| | - Xiao Yu Wang
- Institute of Translational Immunology and Research Center for Immunotherapy, University of Mainz Medical Center, Mainz, Germany
| |
Collapse
|
224
|
Zhou F, Wang A, Li D, Wang Y, Lin L. Pinocembrin from Penthorum chinense Pursh suppresses hepatic stellate cells activation through a unified SIRT3-TGF-β-Smad signaling pathway. Toxicol Appl Pharmacol 2018; 341:38-50. [DOI: 10.1016/j.taap.2018.01.009] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Revised: 01/03/2018] [Accepted: 01/13/2018] [Indexed: 01/18/2023]
|
225
|
BH3 mimetics as anti-fibrotic therapy: Unleashing the mitochondrial pathway of apoptosis in myofibroblasts. Matrix Biol 2018; 68-69:94-105. [PMID: 29408011 DOI: 10.1016/j.matbio.2018.01.020] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Revised: 01/26/2018] [Accepted: 01/28/2018] [Indexed: 12/22/2022]
Abstract
Organs and tissues in mammals can undergo self-repair following injury. However, chronic or severe tissue injury leads to the development of dense scar tissue or fibrosis at the expense of regeneration. The identification of novel therapeutic strategies aiming at reversing fibrosis is therefore a major clinical unmet need in regenerative medicine. Persistent activation of scar-forming myofibroblasts distinguishes non-resolving pathological fibrosis from self-limited physiological wound healing. Thus, therapeutic strategies selectively inducing myofibroblast apoptosis could prevent progression and potentially reverse established fibrosis in fibrotic diseases. In this Review, we discuss recent findings that have demonstrated that activated myofibroblasts, traditionally viewed as apoptosis-resistant cells, are actually "primed for death". In this state, mitochondria of activated myofibroblasts are loaded with proapoptotic BH3 proteins, which creates a cellular "addiction" to individual antiapoptotic proteins to block prodeath signaling and ensure survival. This creates a novel therapeutic opportunity to treat organ fibrosis by inducing myofibroblast apoptosis with the so-called BH3 mimetic drugs, which have recently shown potent antifibrotic activities in experimental models. Finally, we discuss the potential use of BH3 profiling as a functional tool to diagnose myofibroblast addiction to individual antiapoptotic proteins, which may serve to guide and assign the most effective BH3 mimetic drug for patients with fibrotic disease.
Collapse
|
226
|
Zhang Y, Luo G, Zhang Y, Zhang M, Zhou J, Gao W, Xuan X, Yang X, Yang D, Tian Z, Ni B, Tang J. Critical effects of long non-coding RNA on fibrosis diseases. Exp Mol Med 2018; 50:e428. [PMID: 29350677 PMCID: PMC5799794 DOI: 10.1038/emm.2017.223] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Revised: 07/03/2017] [Accepted: 07/05/2017] [Indexed: 02/07/2023] Open
Abstract
The expression or dysfunction of long non-coding RNAs (lncRNAs) is closely related to various hereditary diseases, autoimmune diseases, metabolic diseases and tumors. LncRNAs were also recently recognized as functional regulators of fibrosis, which is a secondary process in many of these diseases and a primary pathology in fibrosis diseases. We review the latest findings on lncRNAs in fibrosis diseases of the liver, myocardium, kidney, lung and peritoneum. We also discuss the potential of disease-related lncRNAs as therapeutic targets for the clinical treatment of human fibrosis diseases.
Collapse
Affiliation(s)
- Yue Zhang
- Department of Dermatology, 105th Hospital of PLA, Hefei, China.,Department of Pathophysiology and High Altitude Pathology, Third Military Medical University, Chongqing, China.,Graduate School, Bengbu Medical College, Bengbu, China
| | - Gang Luo
- Department of Pathophysiology and High Altitude Pathology, Third Military Medical University, Chongqing, China
| | - Yi Zhang
- Department of Clinical Laboratory, 150th Hospital of PLA, Luoyang, China
| | - Mengjie Zhang
- Department of Pathophysiology and High Altitude Pathology, Third Military Medical University, Chongqing, China
| | - Jian Zhou
- Department of Pathophysiology and High Altitude Pathology, Third Military Medical University, Chongqing, China
| | - Weiwu Gao
- Department of Pathophysiology and High Altitude Pathology, Third Military Medical University, Chongqing, China
| | - Xiuyun Xuan
- Department of Microbiology and Immunology, Shanxi Medical University, Taiyuan, China
| | - Xia Yang
- Institute of Immunology, PLA, Third Military Medical University, Chongqing, China
| | - Di Yang
- Institute of Immunology, PLA, Third Military Medical University, Chongqing, China
| | - Zhiqiang Tian
- Institute of Immunology, PLA, Third Military Medical University, Chongqing, China
| | - Bing Ni
- Department of Pathophysiology and High Altitude Pathology, Third Military Medical University, Chongqing, China
| | - Jun Tang
- Department of Dermatology, 105th Hospital of PLA, Hefei, China.,Graduate School, Bengbu Medical College, Bengbu, China
| |
Collapse
|
227
|
Cordero-Espinoza L, Huch M. The balancing act of the liver: tissue regeneration versus fibrosis. J Clin Invest 2018; 128:85-96. [PMID: 29293095 PMCID: PMC5749503 DOI: 10.1172/jci93562] [Citation(s) in RCA: 156] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Epithelial cell loss alters a tissue's optimal function and awakens evolutionarily adapted healing mechanisms to reestablish homeostasis. Although adult mammalian organs have a limited regeneration potential, the liver stands out as one remarkable exception. Following injury, the liver mounts a dynamic multicellular response wherein stromal cells are activated in situ and/or recruited from the bloodstream, the extracellular matrix (ECM) is remodeled, and epithelial cells expand to replenish their lost numbers. Chronic damage makes this response persistent instead of transient, tipping the system into an abnormal steady state known as fibrosis, in which ECM accumulates excessively and tissue function degenerates. Here we explore the cellular and molecular switches that balance hepatic regeneration and fibrosis, with a focus on uncovering avenues of disease modeling and therapeutic intervention.
Collapse
|
228
|
Abstract
Fibrosis is the excessive accumulation of extracellular matrix that often occurs as a wound healing response to repeated or chronic tissue injury, and may lead to the disruption of organ architecture and loss of function. Although fibrosis was previously thought to be irreversible, recent evidence indicates that certain circumstances permit the resolution of fibrosis when the underlying causes of injury are eradicated. The mechanism of fibrosis resolution encompasses degradation of the fibrotic extracellular matrix as well as elimination of fibrogenic myofibroblasts through their adaptation of various cell fates, including apoptosis, senescence, dedifferentiation, and reprogramming. In this Review, we discuss the present knowledge and gaps in our understanding of how matrix degradation is regulated and how myofibroblast cell fates can be manipulated, areas that may identify potential therapeutic approaches for fibrosis.
Collapse
|
229
|
Crawford JM, Bioulac-Sage P, Hytiroglou P. Structure, Function, and Responses to Injury. MACSWEEN'S PATHOLOGY OF THE LIVER 2018:1-87. [DOI: 10.1016/b978-0-7020-6697-9.00001-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
230
|
Abstract
Fibrosis of the liver is an inherent wound healing response to chronic liver injury. Regeneration of liver epithelium and restoration of normal liver structure were generally involved in this process. Although the liver has a striking capacity to adapt to damage through tissue repair, excessive accumulation of extracellular matrix during this process often leads to scar tissue formation and subsequent fibrosis. Epithelial to mesenchymal transition (EMT) enables a polarized epithelial cell to undergo multiple changes biochemically and to bear a mesenchymal cell phenotype. EMT plays a critical role in tissue and organ development and embryogenesis. In the liver, it is proposed that epithelial cells can acquire fibroblastic phonotype via EMT and contribute to fibrogenesis. This made EMT a potential target for antifibrotic strategies. Following an original passion, many investigators devote themselves to exploring this mechanism in liver fibrosis. However, as research continues, this hypothesis became highly controversial. The exact contribution of EMT to fibrogenesis was challenged due to the contradictory results from related studies. In this review, we summarized the recent advances regarding EMT in hepatic fibrosis and discussed the potentially involved liver cell types and pathways in order to reach rational and helpful conclusions.
Collapse
Affiliation(s)
- Kangkang Yu
- Department of Infectious Diseases, Huashan Hospital, Fudan University, Shanghai, People's Republic of China
| | - Qian Li
- Department of General Surgery, Qingdao Municipal Hospital, Qingdao, People's Republic of China
| | - Guangfeng Shi
- Department of Infectious Diseases, Huashan Hospital, Fudan University, Shanghai, People's Republic of China
| | - Ning Li
- Department of Infectious Diseases, Huashan Hospital, Fudan University, Shanghai, People's Republic of China,Address for correspondence: Dr. Ning Li, Department of Infectious Diseases, Huashan Hospital, Fudan University, Shanghai – 200040, People's Republic of China. E-mail:
| |
Collapse
|
231
|
Cong M, Jia J, Kisseleva T, Brenner DA. The Liver's Response to Injury. ZAKIM AND BOYER'S HEPATOLOGY 2018:77-83.e5. [DOI: 10.1016/b978-0-323-37591-7.00005-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
232
|
Paish HL, Kalson NS, Smith GR, Del Carpio Pons A, Baldock TE, Smith N, Swist-Szulik K, Weir DJ, Bardgett M, Deehan DJ, Mann DA, Borthwick LA. Fibroblasts Promote Inflammation and Pain via IL-1α Induction of the Monocyte Chemoattractant Chemokine (C-C Motif) Ligand 2. THE AMERICAN JOURNAL OF PATHOLOGY 2017; 188:696-714. [PMID: 29248462 PMCID: PMC5842035 DOI: 10.1016/j.ajpath.2017.11.007] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Revised: 10/24/2017] [Accepted: 11/09/2017] [Indexed: 01/06/2023]
Abstract
Fibroblasts persist within fibrotic scar tissue and exhibit considerable phenotypic and functional plasticity. Herein, we hypothesized that scar-associated fibroblasts may be a source of stress-induced inflammatory exacerbations and pain. To test this idea, we used a human model of surgery-induced fibrosis, total knee arthroplasty (TKA). Using a combination of tissue protein expression profiling and bioinformatics, we discovered that many months after TKA, the fibrotic joint exists in a state of unresolved chronic inflammation. Moreover, the infrapatellar fat pad, a soft tissue that becomes highly fibrotic in the post-TKA joint, expresses multiple inflammatory mediators, including the monocyte chemoattractant, chemokine (C-C motif) ligand (CCL) 2, and the innate immune trigger, IL-1α. Fibroblasts isolated from the post-TKA fibrotic infrapatellar fat pad express the IL-1 receptor and on exposure to IL-1α polarize to a highly inflammatory state that enables them to stimulate the recruitment of monocytes. Blockade of fibroblast CCL2 or its transcriptional regulator NF-κB prevented IL-1α-induced monocyte recruitment. Clinical investigations discovered that levels of patient-reported pain in the post-TKA joint correlated with concentrations of CCL2 in the joint tissue, such that the chemokine is effectively a pain biomarker in the TKA patient. We propose that an IL-1α-NF-κB-CCL2 signaling pathway, operating within scar-associated fibroblasts, may be therapeutically manipulated for alleviating inflammation and pain in fibrotic joints and other tissues.
Collapse
Affiliation(s)
- Hannah L Paish
- Fibrosis Research Group, Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Nicholas S Kalson
- Fibrosis Research Group, Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom; Musculoskeletal Unit, Freeman Hospital, Newcastle Hospitals, NHS Trust, Newcastle upon Tyne, United Kingdom
| | - Graham R Smith
- Fibrosis Research Group, Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom; Bioinformatics Support Unit, Institute of Neuroscience, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Alicia Del Carpio Pons
- Fibrosis Research Group, Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Thomas E Baldock
- Musculoskeletal Unit, Freeman Hospital, Newcastle Hospitals, NHS Trust, Newcastle upon Tyne, United Kingdom
| | - Nicholas Smith
- Fibrosis Research Group, Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Katarzyna Swist-Szulik
- Wellcome Centre for Mitochondrial Research, Institute of Neuroscience, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - David J Weir
- Musculoskeletal Unit, Freeman Hospital, Newcastle Hospitals, NHS Trust, Newcastle upon Tyne, United Kingdom
| | - Michelle Bardgett
- Musculoskeletal Unit, Freeman Hospital, Newcastle Hospitals, NHS Trust, Newcastle upon Tyne, United Kingdom
| | - David J Deehan
- Fibrosis Research Group, Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom; Musculoskeletal Unit, Freeman Hospital, Newcastle Hospitals, NHS Trust, Newcastle upon Tyne, United Kingdom
| | - Derek A Mann
- Fibrosis Research Group, Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Lee A Borthwick
- Fibrosis Research Group, Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom.
| |
Collapse
|
233
|
Lagares D, Santos A, Grasberger PE, Liu F, Probst CK, Rahimi RA, Sakai N, Kuehl T, Ryan J, Bhola P, Montero J, Kapoor M, Baron M, Varelas X, Tschumperlin DJ, Letai A, Tager AM. Targeted apoptosis of myofibroblasts with the BH3 mimetic ABT-263 reverses established fibrosis. Sci Transl Med 2017; 9:eaal3765. [PMID: 29237758 PMCID: PMC8520471 DOI: 10.1126/scitranslmed.aal3765] [Citation(s) in RCA: 170] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Revised: 07/28/2017] [Accepted: 10/30/2017] [Indexed: 08/26/2023]
Abstract
Persistent myofibroblast activation distinguishes pathological fibrosis from physiological wound healing, suggesting that therapies selectively inducing myofibroblast apoptosis could prevent progression and potentially reverse established fibrosis in diseases such as scleroderma, a heterogeneous autoimmune disease characterized by multiorgan fibrosis. We demonstrate that fibroblast-to-myofibroblast differentiation driven by matrix stiffness increases the mitochondrial priming (proximity to the apoptotic threshold) of these activated cells. Mitochondria in activated myofibroblasts, but not quiescent fibroblasts, are primed by death signals such as the proapoptotic BH3-only protein BIM, which creates a requirement for tonic expression of the antiapoptotic protein BCL-XL to sequester BIM and ensure myofibroblast survival. Myofibroblasts become particularly susceptible to apoptosis induced by "BH3 mimetic" drugs inhibiting BCL-XL such as ABT-263. ABT-263 displaces BCL-XL binding to BIM, allowing BIM to activate apoptosis on stiffness-primed myofibroblasts. Therapeutic blockade of BCL-XL with ABT-263 (navitoclax) effectively treats established fibrosis in a mouse model of scleroderma dermal fibrosis by inducing myofibroblast apoptosis. Using a BH3 profiling assay to assess mitochondrial priming in dermal fibroblasts derived from patients with scleroderma, we demonstrate that the extent of apoptosis induced by BH3 mimetic drugs correlates with the extent of their mitochondrial priming, indicating that BH3 profiling could predict apoptotic responses of fibroblasts to BH3 mimetic drugs in patients with scleroderma. Together, our findings elucidate the potential efficacy of targeting myofibroblast antiapoptotic proteins with BH3 mimetic drugs in scleroderma and other fibrotic diseases.
Collapse
Affiliation(s)
- David Lagares
- Fibrosis Research Center and Center for Immunology and Inflammatory Diseases, Division of Pulmonary and Critical Care Medicine, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA.
| | - Alba Santos
- Fibrosis Research Center and Center for Immunology and Inflammatory Diseases, Division of Pulmonary and Critical Care Medicine, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Paula E Grasberger
- Fibrosis Research Center and Center for Immunology and Inflammatory Diseases, Division of Pulmonary and Critical Care Medicine, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Fei Liu
- Molecular and Integrative Physiological Sciences Program, Harvard T. H. Chan School of Public Health, Boston, MA 02115, USA
| | - Clemens K Probst
- Fibrosis Research Center and Center for Immunology and Inflammatory Diseases, Division of Pulmonary and Critical Care Medicine, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Rod A Rahimi
- Fibrosis Research Center and Center for Immunology and Inflammatory Diseases, Division of Pulmonary and Critical Care Medicine, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Norihiko Sakai
- Fibrosis Research Center and Center for Immunology and Inflammatory Diseases, Division of Pulmonary and Critical Care Medicine, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
- Division of Nephrology and Division of Blood Purification, Kanazawa University Hospital, Kanazawa, Japan
| | - Tobias Kuehl
- Fibrosis Research Center and Center for Immunology and Inflammatory Diseases, Division of Pulmonary and Critical Care Medicine, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Jeremy Ryan
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Patrick Bhola
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Joan Montero
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Mohit Kapoor
- Krembil Research Institute, University Health Network and Department of Surgery and Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Murray Baron
- Jewish General Hospital, McGill University, Montreal, Quebec, Canada
| | - Xaralabos Varelas
- Department of Biochemistry, Boston University School of Medicine, Boston, MA 02118, USA
| | - Daniel J Tschumperlin
- Molecular and Integrative Physiological Sciences Program, Harvard T. H. Chan School of Public Health, Boston, MA 02115, USA
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA
| | - Anthony Letai
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Andrew M Tager
- Fibrosis Research Center and Center for Immunology and Inflammatory Diseases, Division of Pulmonary and Critical Care Medicine, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| |
Collapse
|
234
|
Wen Z, Ji X, Tang J, Lin G, Xiao L, Liang C, Wang M, Su F, Ferrandon D, Li Z. Positive Feedback Regulation between Transglutaminase 2 and Toll-Like Receptor 4 Signaling in Hepatic Stellate Cells Correlates with Liver Fibrosis Post Schistosoma japonicum Infection. Front Immunol 2017; 8:1808. [PMID: 29321784 PMCID: PMC5733538 DOI: 10.3389/fimmu.2017.01808] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2017] [Accepted: 11/30/2017] [Indexed: 12/12/2022] Open
Abstract
Liver fibrosis induced by Schistosoma japonicum (Sj) infection is characterized by the accumulation of extracellular matrix (ECM). The activated and differentiated hepatic stellate cells (HSCs) are the predominant ECM-producing cell type in the liver. Toll-like receptor (TLR) 4 pathway activation plays a key role in mice liver fibrosis models induced by alcohol, biliary ligation, and carbon tetrachloride 4. In this work, we found that TLR4 pathway activation correlated with the severity of liver fibrosis post Sj infection. The TLR4 receptor inhibitor TAK242 reduced the extent of liver fibrosis. The increased expression of TLR4, α-smooth muscle actin (α-SMA), and cytoglobin was observed in the HSCs of mouse liver after Sj infection. In response to stimulation with either lipopolysaccharide or Sj's soluble egg antigen (SEA), high levels of TLR4 and α-SMA were induced in HSCs and were inhibited by TAK242 treatment. In previous work, we had reported that a high level of transglutaminase 2 (TGM2) is crucial for liver fibrosis post Sj infection. Herein, we found that TLR4 signaling also controlled Tgm2 expression. Inhibition of TGM2 activity by cystamine (CTM) in Sj-infected mice or in HSCs induced with all-trans-retinoic acid (ATRA) stimulation led to a lowered activation of TLR4 signaling and a reduced α-SMA expression. These results were confirmed by downregulating the Tgm2 gene by specific siRNA. These observations implied the presence of a positive feedback regulation between TGM2 and TLR4 signaling in HSCs that correlated with liver fibrosis post Sj infection. This novel connection between TGM2 and TLR4 pathway activation in liver fibrosis induced by Sj infection enhances our understanding of liver diseases.
Collapse
Affiliation(s)
- Zhencheng Wen
- Sino-French Hoffmann Institute, Guangzhou Medical University, Guangzhou, China
| | - Xiaofang Ji
- Sino-French Hoffmann Institute, Guangzhou Medical University, Guangzhou, China
| | - Juanjuan Tang
- Sino-French Hoffmann Institute, Guangzhou Medical University, Guangzhou, China
| | - Guiying Lin
- Sino-French Hoffmann Institute, Guangzhou Medical University, Guangzhou, China
| | - Linzhuo Xiao
- Sino-French Hoffmann Institute, Guangzhou Medical University, Guangzhou, China
| | - Cuiying Liang
- Sino-French Hoffmann Institute, Guangzhou Medical University, Guangzhou, China
| | - Manni Wang
- Sino-French Hoffmann Institute, Guangzhou Medical University, Guangzhou, China
| | - Fang Su
- Sino-French Hoffmann Institute, Guangzhou Medical University, Guangzhou, China
| | - Dominique Ferrandon
- Sino-French Hoffmann Institute, Guangzhou Medical University, Guangzhou, China.,Université de Strasbourg, RIDI UPR9022 du CNRS, Strasbourg, France
| | - Zi Li
- Sino-French Hoffmann Institute, Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
235
|
Delire B, Lebrun V, Selvais C, Henriet P, Bertrand A, Horsmans Y, Leclercq IA. Aging enhances liver fibrotic response in mice through hampering extracellular matrix remodeling. Aging (Albany NY) 2017; 9:98-113. [PMID: 27941216 PMCID: PMC5310658 DOI: 10.18632/aging.101124] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Accepted: 11/24/2016] [Indexed: 02/06/2023]
Abstract
Clinical data identify age as a factor for severe liver fibrosis. We evaluate whether and how aging modulates the fibrotic response in a mouse model. Liver fibrosis was induced by CCl4 injections (thrice weekly for 2 weeks) in 7 weeks- and 15 months-old mice (young and old, respectively). Livers were analyzed for fibrosis, inflammation and remodeling 48 and 96 hours after the last injection. Old mice developed more severe fibrosis compared to young ones as evaluated by sirius red morphometry. Expression of pro-fibrogenic genes was equally induced in the two age-groups but enhanced fibrolysis in young mice was demonstrated by a significantly higher Mmp13 induction and collagenase activity. While fibrosis resolution occurred in young mice within 96 hours, no significant fibrosis attenuation was observed in old mice. Although recruitment of monocytes-derived macrophages was similar in young and old livers, young macrophages had globally a remodeling phenotype while old ones, a pro-fibrogenic phenotype. Moreover, we observed a higher proportion of thick fibers and enhanced expression of enzymes involved in collagen maturation in old mice. CONCLUSION Impaired fibrolysis of a matrix less prone to remodeling associated with a pro-inflammatory phenotype of infiltrated macrophages contribute to a more severe fibrosis in old mice.
Collapse
Affiliation(s)
- Bénédicte Delire
- Laboratory of Hepato-Gastroenterology, Institut de Recherche Expérimentale et Clinique (IREC), Université catholique de Louvain (UCL), Brussels, Belgium
| | - Valérie Lebrun
- Laboratory of Hepato-Gastroenterology, Institut de Recherche Expérimentale et Clinique (IREC), Université catholique de Louvain (UCL), Brussels, Belgium
| | - Charlotte Selvais
- Laboratory of Hepato-Gastroenterology, Institut de Recherche Expérimentale et Clinique (IREC), Université catholique de Louvain (UCL), Brussels, Belgium
| | - Patrick Henriet
- Cell Biology Unit, de Duve Institute, Université catholique de Louvain, Brussels, Belgium
| | - Amélie Bertrand
- Laboratory of Hepato-Gastroenterology, Institut de Recherche Expérimentale et Clinique (IREC), Université catholique de Louvain (UCL), Brussels, Belgium
| | - Yves Horsmans
- Laboratory of Hepato-Gastroenterology, Institut de Recherche Expérimentale et Clinique (IREC), Université catholique de Louvain (UCL), Brussels, Belgium.,Department of Hepato-Gastroenterology, Cliniques Universitaires Saint-Luc and Institute of Clinical Research, Université catholique de Louvain, Brussels, Belgium
| | - Isabelle A Leclercq
- Laboratory of Hepato-Gastroenterology, Institut de Recherche Expérimentale et Clinique (IREC), Université catholique de Louvain (UCL), Brussels, Belgium
| |
Collapse
|
236
|
Hepatic stellate cells as key target in liver fibrosis. Adv Drug Deliv Rev 2017; 121:27-42. [PMID: 28506744 DOI: 10.1016/j.addr.2017.05.007] [Citation(s) in RCA: 1007] [Impact Index Per Article: 125.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Revised: 03/21/2017] [Accepted: 05/09/2017] [Indexed: 02/06/2023]
Abstract
Progressive liver fibrosis, induced by chronic viral and metabolic disorders, leads to more than one million deaths annually via development of cirrhosis, although no antifibrotic therapy has been approved to date. Transdifferentiation (or "activation") of hepatic stellate cells is the major cellular source of matrix protein-secreting myofibroblasts, the major driver of liver fibrogenesis. Paracrine signals from injured epithelial cells, fibrotic tissue microenvironment, immune and systemic metabolic dysregulation, enteric dysbiosis, and hepatitis viral products can directly or indirectly induce stellate cell activation. Dysregulated intracellular signaling, epigenetic changes, and cellular stress response represent candidate targets to deactivate stellate cells by inducing reversion to inactivated state, cellular senescence, apoptosis, and/or clearance by immune cells. Cell type- and target-specific pharmacological intervention to therapeutically induce the deactivation will enable more effective and less toxic precision antifibrotic therapies.
Collapse
|
237
|
3D in vitro models of liver fibrosis. Adv Drug Deliv Rev 2017; 121:133-146. [PMID: 28697953 DOI: 10.1016/j.addr.2017.07.004] [Citation(s) in RCA: 89] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Revised: 06/27/2017] [Accepted: 07/06/2017] [Indexed: 02/07/2023]
Abstract
Animal testing is still the most popular preclinical assessment model for liver fibrosis. To develop efficient anti-fibrotic therapies, robust and representative in vitro models are urgently needed. The most widely used in vitro fibrosis model is the culture-induced activation of primary rodent hepatic stellate cells. While these cultures have contributed greatly to the current understanding of hepatic stellate cell activation, they seem to be inadequate to cover the complexity of this regenerative response. This review summarizes recent progress towards the development of 3D culture models of liver fibrosis. Thus far, only a few hepatic culture systems have successfully implemented hepatic stellate cells (or other non-parenchymal cells) into hepatocyte cultures. Recent advances in bioprinting, spheroid- and precision-cut liver slice cultures and the use of microfluidic bioreactors will surely lead to valid 3D in vitro models of liver fibrosis in the near future.
Collapse
|
238
|
Mazza G, Al-Akkad W, Rombouts K. Engineering in vitro models of hepatofibrogenesis. Adv Drug Deliv Rev 2017; 121:147-157. [PMID: 28578016 DOI: 10.1016/j.addr.2017.05.018] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Revised: 05/17/2017] [Accepted: 05/26/2017] [Indexed: 02/07/2023]
Abstract
Chronic liver disease is a major cause of morbidity and mortality worldwide marked by chronic inflammation and fibrosis/scarring, resulting in end-stage liver disease and its complications. Hepatic stellate cells (HSCs) are a dominant contributor to liver fibrosis by producing excessive extracellular matrix (ECM), irrespective of the underlying disease aetiologies, and for many decades research has focused on the development of a number of anti-fibrotic strategies targeting this cell. Despite major improvements in two-dimensional systems (2D) by using a variety of cell culture models of different complexity, an efficient anti-fibrogenic therapy has yet to be developed. The development of well-defined three-dimensional (3D) in vitro models, which mimic ECM structures as found in vivo, have demonstrated the importance of cell-matrix bio-mechanics, the complex interactions between HSCs and hepatocytes and other non-parenchymal cells, and this to improve and promote liver cell-specific functions. Henceforth, refinement of these 3D in vitro models, which reproduce the liver microenvironment, will lead to new objectives and to a possible new era in the search for antifibrogenic compounds.
Collapse
|
239
|
Sato-Matsubara M, Matsubara T, Daikoku A, Okina Y, Longato L, Rombouts K, Thuy LTT, Adachi J, Tomonaga T, Ikeda K, Yoshizato K, Pinzani M, Kawada N. Fibroblast growth factor 2 (FGF2) regulates cytoglobin expression and activation of human hepatic stellate cells via JNK signaling. J Biol Chem 2017; 292:18961-18972. [PMID: 28916723 PMCID: PMC5706471 DOI: 10.1074/jbc.m117.793794] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Revised: 09/11/2017] [Indexed: 12/19/2022] Open
Abstract
Cytoglobin (CYGB) belongs to the mammalian globin family and is exclusively expressed in hepatic stellate cells (HSCs) in the liver. In addition to its gas-binding ability, CYGB is relevant to hepatic inflammation, fibrosis, and cancer because of its anti-oxidative properties; however, the regulation of CYGB gene expression remains unknown. Here, we sought to identify factors that induce CYGB expression in HSCs and to clarify the molecular mechanism involved. We used the human HSC cell line HHSteC and primary human HSCs isolated from intact human liver tissues. In HHSteC cells, treatment with a culture supplement solution that included fibroblast growth factor 2 (FGF2) increased CYGB expression with concomitant and time-dependent α-smooth muscle actin (αSMA) down-regulation. We found that FGF2 is a key factor in inducing the alteration in both CYGB and αSMA expression in HHSteCs and primary HSCs and that FGF2 triggered the rapid phosphorylation of both c-Jun N-terminal kinase (JNK) and c-JUN. Both the JNK inhibitor PS600125 and transfection of c-JUN-targeting siRNA abrogated FGF2-mediated CYGB induction, and conversely, c-JUN overexpression induced CYGB and reduced αSMA expression. Chromatin immunoprecipitation analyses revealed that upon FGF2 stimulation, phospho-c-JUN bound to its consensus motif (5'-TGA(C/G)TCA), located -218 to -222 bases from the transcription initiation site in the CYGB promoter. Of note, in bile duct-ligated mice, FGF2 administration ameliorated liver fibrosis and significantly reduced HSC activation. In conclusion, FGF2 triggers CYGB gene expression and deactivation of myofibroblastic human HSCs, indicating that FGF2 has therapeutic potential for managing liver fibrosis.
Collapse
Affiliation(s)
| | - Tsutomu Matsubara
- Department of Anatomy and Regenerative Biology, Graduate School of Medicine, Osaka City University, Osaka 545-8585, Japan
| | | | | | - Lisa Longato
- the Regenerative Medicine and Fibrosis Group, Institute for Liver and Digestive Health, University College London, Royal Free, London NW3 2PF, United Kingdom, and
| | - Krista Rombouts
- the Regenerative Medicine and Fibrosis Group, Institute for Liver and Digestive Health, University College London, Royal Free, London NW3 2PF, United Kingdom, and
| | | | - Jun Adachi
- the Laboratory of Proteome Research, Proteome Research Center, National Institute of Biomedical Innovation, Osaka 567-0085, Japan
| | - Takeshi Tomonaga
- the Laboratory of Proteome Research, Proteome Research Center, National Institute of Biomedical Innovation, Osaka 567-0085, Japan
| | - Kazuo Ikeda
- Department of Anatomy and Regenerative Biology, Graduate School of Medicine, Osaka City University, Osaka 545-8585, Japan
| | | | - Massimo Pinzani
- the Regenerative Medicine and Fibrosis Group, Institute for Liver and Digestive Health, University College London, Royal Free, London NW3 2PF, United Kingdom, and
| | | |
Collapse
|
240
|
吴 兰, 刘 文. 肝纤维化逆转机制的研究进展及治疗概况. Shijie Huaren Xiaohua Zazhi 2017; 25:2123-2132. [DOI: 10.11569/wcjd.v25.i23.2123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
肝纤维化是肝脏对慢性损伤的一种修复反应, 多是持续性肝脏损伤或纤维化刺激因子刺激产生的共有病理改变, 是一项严重的全球性健康难题. 近年来临床研究发现, 由病毒性肝炎造成肝纤维化或肝硬化的患者, 在成功接受病毒性肝炎治疗后, 其肝纤维化甚至肝硬化发生了逆转现象. 因此研究和了解肝纤维化逆转的机制有利于发现新的针对肝纤维化的治疗靶向. 本文就近年来有关肝纤维化逆转机制的研究以及治疗概况作一综述, 以期为肝纤维化的研究提供帮助.
Collapse
|
241
|
Ye J, Zhang Z, Zhu L, Lu M, Li Y, Zhou J, Lu X, Du Q. Polaprezinc inhibits liver fibrosis and proliferation in hepatocellular carcinoma. Mol Med Rep 2017; 16:5523-5528. [PMID: 28849143 DOI: 10.3892/mmr.2017.7262] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Accepted: 06/02/2017] [Indexed: 11/06/2022] Open
Abstract
Hepatic fibrosis is defined as a pathological process, and activation of hepatic stellate cells (HSCs) is believed to be the key event of liver fibrosis. Additionally, activated HSCs may participate in the formation of the tumor microenvironment. Polaprezinc, a protector of the gastric mucosa, has been recently demonstrated to be an inhibitor of liver fibrosis in a mouse model. Proliferation and colony formation assays were performed to determine the inhibitory effects of polaprezinc on the growth of LX‑2 and hepG2 cells. A migration assay was used to evaluate the change in mobility of LX‑2 cells and quantitative polymerase chain reaction was performed to detect the expression levels of key markers of fibrosis. Finally, a gene chip assay for polaprezinc‑treated hepG2 cells was performed to evaluate the effect of polaprezinc on the hepG2 gene expression profile. The proliferation assay indicated that polaprezinc may inhibit the LX‑2 cell proliferation and the migration assays confirmed the inhibition of mobility. The expression levels of fibrotic markers such as collagen I, fibronectin and α‑smooth muscle actin were downregulated following polaprezinc treatment. The proliferation activity of polaprezinc‑treated hepG2 cells was reduced and the gene chip assay indicated that series of gene expression changes associated with cancer migration, cell skeletal organization and proliferation had occurred. In conclusion, polaprezinc treatment mayinhibit the proliferation of hepatocellular carcinoma cells and reverse liver fibrosis by deactivating HSCs. The present findings suggest that polaprezinc provides a novel treatment for patients with gastritis complicated with cirrhosis.
Collapse
Affiliation(s)
- Jun Ye
- Department of Gastroenterology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310009, P.R. China
| | - Zhengsen Zhang
- Department of Gastroenterology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310009, P.R. China
| | - Liang Zhu
- Department of Laboratory Construction and Management, Hangzhou Normal University School of Medicine, Hangzhou, Zhejiang 310009, P.R. China
| | - Minfang Lu
- Department of Gastroenterology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310009, P.R. China
| | - Yan Li
- Department of Gastroenterology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310009, P.R. China
| | - Jingjing Zhou
- Department of Gastroenterology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310009, P.R. China
| | - Xinliang Lu
- Department of Gastroenterology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310009, P.R. China
| | - Qin Du
- Department of Gastroenterology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310009, P.R. China
| |
Collapse
|
242
|
NLRP3 inflammasome activation results in liver inflammation and fibrosis in mice infected with Schistosoma japonicum in a Syk-dependent manner. Sci Rep 2017; 7:8120. [PMID: 28808303 PMCID: PMC5556086 DOI: 10.1038/s41598-017-08689-1] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Accepted: 07/06/2017] [Indexed: 02/06/2023] Open
Abstract
Granulomatous and fibrosing inflammation in response to soluble egg antigen (SEA) from Schistosoma japonicum (S. japonicum) is the main pathological process of S. japonicum infection. Inflammasome activation has recently been implicated in the pathogenesis of liver disease. However, the role of inflammasome activation in schistosomiasis-associated liver fibrosis (SSLF) has not been extensively studied. In this study, it is demonstrated that the NLRP3 inflammasome is markedly activated in mouse HSCs both in vivo and in vitro during S. japonicum infection. Furthermore, it is demonstrated that inhibition of NLRP3 inflammasome significantly alleviates the liver inflammation and collagen deposition that are induced by infection with S. japonicum. The mechanism of SEA-induced NLRP3 inflammasome activation is studied in isolated, cultured mouse HSCs and it is shown that SEA-induced NLRP3 inflammasome activation in HSCs is dependent upon the activities of spleen tyrosine kinase (Syk), an enzyme usually associated with a pathogen recognition receptor for fungal pathogens. Moreover, it is demonstrated that Dectin-1 and JNK signaling are also involved in SEA-induced NLRP3 inflammasome activation in HSCs. These data shed new light on the mechanisms of NLRP3 inflammasome activation during an infection with S. japonicum, and further characterize its role in schistosomiasis-associated liver fibrosis (SSLF).
Collapse
|
243
|
de Souza VCA, Pereira TA, Teixeira VW, Carvalho H, de Castro MCAB, D’assunção CG, de Barros AF, Carvalho CL, de Lorena VMB, Costa VMA, Teixeira ÁAC, Figueiredo RCBQ, de Oliveira SA. Bone marrow-derived monocyte infusion improves hepatic fibrosis by decreasing osteopontin, TGF-β1, IL-13 and oxidative stress. World J Gastroenterol 2017; 23:5146-5157. [PMID: 28811709 PMCID: PMC5537181 DOI: 10.3748/wjg.v23.i28.5146] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2016] [Revised: 02/25/2017] [Accepted: 04/12/2017] [Indexed: 02/06/2023] Open
Abstract
AIM To evaluate the therapeutic effects of bone marrow-derived CD11b+CD14+ monocytes in a murine model of chronic liver damage.
METHODS Chronic liver damage was induced in C57BL/6 mice by administration of carbon tetrachloride and ethanol for 6 mo. Bone marrow-derived monocytes isolated by immunomagnetic separation were used for therapy. The cell transplantation effects were evaluated by morphometry, biochemical assessment, immunohistochemistry and enzyme-linked immunosorbent assay.
RESULTS CD11b+CD14+ monocyte therapy significantly reduced liver fibrosis and increased hepatic glutathione levels. Levels of pro-inflammatory cytokines, including tumor necrosis factor-α, interleukin (IL)-6 and IL-1β, in addition to pro-fibrotic factors, such as IL-13, transforming growth factor-β1 and tissue inhibitor of metalloproteinase-1 also decreased, while IL-10 and matrix metalloproteinase-9 increased in the monocyte-treated group. CD11b+CD14+ monocyte transplantation caused significant changes in the hepatic expression of α-smooth muscle actin and osteopontin.
CONCLUSION Monocyte therapy is capable of bringing about improvement of liver fibrosis by reducing oxidative stress and inflammation, as well as increasing anti-fibrogenic factors.
Collapse
|
244
|
de Oliveira da Silva B, Ramos LF, Moraes KCM. Molecular interplays in hepatic stellate cells: apoptosis, senescence, and phenotype reversion as cellular connections that modulate liver fibrosis. Cell Biol Int 2017; 41:946-959. [PMID: 28498509 DOI: 10.1002/cbin.10790] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2016] [Accepted: 05/08/2017] [Indexed: 12/18/2022]
Abstract
Liver fibrosis is a pathophysiological process correlated with intense repair and cicatrization mechanisms in injured liver, and over the past few years, the characterization of the fine-tuning of molecular interconnections that support the development of liver fibrosis has been investigated. In this cellular process, the hepatic stellate cells (HSCs) support the organ fibrogenesis. The HSCs are found in two distinct morpho-physiological states: quiescent and activated. In normal liver, most HSCs are found in quiescent state, presenting a considerable amount of lipid droplets in the cytoplasm, while in injured liver, the activated phenotype of HSCs is a myofibroblast, that secrete extracellular matrix elements and contribute to the establishment of the fibrotic process. Studies on the molecular mechanisms by which HSCs try to restore their quiescent state have been performed; however, no effective treatment to reverse fibrosis has been so far prescribed. Therefore, the elucidation of the cellular and molecular mechanisms of apoptosis, senescence, and the cell reversion phenotype process from activate to quiescent state will certainly contribute to the development of effective therapies to treat hepatic fibrosis. In this context, this review aimed to address central elements of apoptosis, senescence, and reversal of HSC phenotype in the control of hepatic fibrogenesis, as a guide to future development of therapeutic strategies.
Collapse
Affiliation(s)
- Brenda de Oliveira da Silva
- Universidade Federal de Ouro Preto, Núcleo de Pesquisa em Ciências Biológicas, Programa de Pós-Graduação em Biotecnologia, Ouro Preto, Minas Gerais, Brazil.,Molecular Biology Laboratory, Departamento de Biologia, Instituto de Biociências, Universidade Estadual Paulista "Júlio de Mesquita Filho"-Campus Rio Claro, Rio Claro, São Paulo, Brazil
| | - Letícia Ferrreira Ramos
- Molecular Biology Laboratory, Departamento de Biologia, Instituto de Biociências, Universidade Estadual Paulista "Júlio de Mesquita Filho"-Campus Rio Claro, Rio Claro, São Paulo, Brazil
| | - Karen C M Moraes
- Molecular Biology Laboratory, Departamento de Biologia, Instituto de Biociências, Universidade Estadual Paulista "Júlio de Mesquita Filho"-Campus Rio Claro, Rio Claro, São Paulo, Brazil
| |
Collapse
|
245
|
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
|
246
|
Abstract
Hepatic fibrosis is a dynamic process characterized by the net accumulation of extracellular matrix resulting from chronic liver injury of any aetiology, including viral infection, alcoholic liver disease and NASH. Activation of hepatic stellate cells (HSCs) - transdifferentiation of quiescent, vitamin-A-storing cells into proliferative, fibrogenic myofibroblasts - is now well established as a central driver of fibrosis in experimental and human liver injury. Yet, the continued discovery of novel pathways and mediators, including autophagy, endoplasmic reticulum stress, oxidative stress, retinol and cholesterol metabolism, epigenetics and receptor-mediated signals, reveals the complexity of HSC activation. Extracellular signals from resident and inflammatory cells including macrophages, hepatocytes, liver sinusoidal endothelial cells, natural killer cells, natural killer T cells, platelets and B cells further modulate HSC activation. Finally, pathways of HSC clearance have been greatly clarified, and include apoptosis, senescence and reversion to an inactivated state. Collectively, these findings reinforce the remarkable complexity and plasticity of HSC activation, and underscore the value of clarifying its regulation in hopes of advancing the development of novel diagnostics and therapies for liver disease.
Collapse
Affiliation(s)
- Takuma Tsuchida
- Division of Liver Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, 1425 Madison Avenue, Box 1123, New York, New York 10029, USA.,Research Division, Mitsubishi Tanabe Pharma Corporation, 2-2-50, Kawagishi, Toda-shi, Saitama 335-8505, Japan
| | - Scott L Friedman
- Division of Liver Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, 1425 Madison Avenue, Box 1123, New York, New York 10029, USA
| |
Collapse
|
247
|
Abshagen K, Rotberg T, Genz B, Vollmar B. No significant impact of Foxf1 siRNA treatment in acute and chronic CCl 4 liver injury. Exp Biol Med (Maywood) 2017. [PMID: 28629226 DOI: 10.1177/1535370217716425] [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] [Indexed: 12/20/2022] Open
Abstract
Chronic liver injury of any etiology is the main trigger of fibrogenic responses and thought to be mediated by hepatic stellate cells. Herein, activating transcription factors like forkhead box f1 are described to stimulate pro-fibrogenic genes in hepatic stellate cells. By using a liver-specific siRNA delivery system (DBTC), we evaluated whether forkhead box f1 siRNA treatment exhibit beneficial effects in murine models of acute and chronic CCl4-induced liver injury. Systemic administration of DBTC-forkhead box f1 siRNA in mice was only sufficient to silence forkhead box f1 in acute CCl4 model, but was not able to attenuate liver injury as measured by liver enzymes and necrotic liver cell area. Therapeutic treatment of mice with DBTC-forkhead box f1 siRNA upon chronic CCl4 exposition failed to inhibit forkhead box f1 expression and hence lacked to diminish hepatic stellate cells activation or fibrosis development. As a conclusion, DBTC-forkhead box f1 siRNA reduced forkhead box f1 expression in a model of acute but not chronic toxic liver injury and showed no positive effects in either of these mice models. Impact statement As liver fibrosis is a worldwide health problem, antifibrotic therapeutic strategies are urgently needed. Therefore, further developments of new technologies including validation in different experimental models of liver disease are essential. Since activation of hepatic stellate cells is a key event upon liver injury, the activating transcription factor forkhead box f1 (Foxf1) represents a potential target gene. Previously, we evaluated Foxf1 silencing by a liver-specific siRNA delivery system (DBTC), exerting beneficial effects in cholestasis. The present study was designed to confirm the therapeutic potential of Foxf1 siRNA in models of acute and chronic CCl4-induced liver injury. DBTC-Foxf1 siRNA was only sufficient to silence Foxf1 in acute CCl4 model and did not ameliorate liver injury or fibrogenesis. This underlines the significance of the experimental model used. Each model displays specific characteristics in the pathogenic nature, time course and severity of fibrosis and the optimal time point for starting a therapy.
Collapse
Affiliation(s)
- Kerstin Abshagen
- 1 Institute for Experimental Surgery, Rostock University Medical Center, 18057 Rostock, Germany
| | - Tobias Rotberg
- 1 Institute for Experimental Surgery, Rostock University Medical Center, 18057 Rostock, Germany
| | - Berit Genz
- 1 Institute for Experimental Surgery, Rostock University Medical Center, 18057 Rostock, Germany.,2 QIMR Berghofer Medical Research Institute, Brisbane QLD 4006, Australia
| | - Brigitte Vollmar
- 1 Institute for Experimental Surgery, Rostock University Medical Center, 18057 Rostock, Germany
| |
Collapse
|
248
|
Long-term challenges and perspectives of pre-adolescent liver disease. Lancet Gastroenterol Hepatol 2017; 2:435-445. [DOI: 10.1016/s2468-1253(16)30160-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Revised: 09/13/2016] [Accepted: 09/22/2016] [Indexed: 12/11/2022]
|
249
|
Kim SU, Heo JY, Kim BK, Park JY, Kim DY, Han KH, Ahn SH, Kim HS. Wisteria floribunda agglutinin-positive human Mac-2 binding protein predicts the risk of HBV-related liver cancer development. Liver Int 2017; 37:879-887. [PMID: 27973711 DOI: 10.1111/liv.13341] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Accepted: 12/05/2016] [Indexed: 02/13/2023]
Abstract
BACKGROUND & AIMS Wisteria floribunda agglutinin-positive human Mac-2 binding protein (WFA+ -M2BP) can be used to assess the degree of liver fibrosis, but few studies have investigated its prognostic utility. We evaluated whether serum WFA+ -M2BP can predict the development of hepatocellular carcinoma (HCC) in chronic hepatitis B (CHB) patients. METHODS A total of 1323 CHB patients with WFA+ -M2BP test results between 2009 and 2011 were included in this retrospective analysis. RESULTS The mean age of patients (793 men) was 51.0 years. During the follow-up period (median 60.3 months), 52 (3.9%) patients developed HCC. Age, the proportion of male gender, the presence of diabetes and cirrhosis, and levels of aspartate aminotransferase, alpha-foetoprotein, and WFA+ -M2BP were significantly greater in patients with HCC than in those without HCC, whereas serum albumin levels and platelet counts were significantly lower in patients with HCC than in those without HCC (all P<.05). In multivariate analysis, WFA+ -M2BP level was an independent predictor of HCC development (adjusted hazard ratio 1.143, 95% CI: 1.139-1.829), along with male gender and diabetes (all P<.05). In patients without cirrhosis (n=1087), WFA+ -M2BP levels ≥1.8 were associated with a higher risk of HCC development (P<.001 by log-rank test), whereas WFA+ -M2BP levels ≥1.8 tended to be associated with a higher risk of HCC development in patients with cirrhosis (n=236) (P=.073 by log-rank test). CONCLUSIONS WFA+ -M2BP level can independently predict HCC development. Further studies should investigate whether WFA+ -M2BP level could be incorporated into surveillance strategies for CHB patients.
Collapse
Affiliation(s)
- Seung Up Kim
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, South Korea
- Institute of Gastroenterology, Yonsei University College of Medicine, Seoul, South Korea
| | - Ja Yoon Heo
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, South Korea
| | - Beom Kyung Kim
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, South Korea
- Institute of Gastroenterology, Yonsei University College of Medicine, Seoul, South Korea
| | - Jun Yong Park
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, South Korea
- Institute of Gastroenterology, Yonsei University College of Medicine, Seoul, South Korea
| | - Do Young Kim
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, South Korea
- Institute of Gastroenterology, Yonsei University College of Medicine, Seoul, South Korea
| | - Kwang-Hyub Han
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, South Korea
- Institute of Gastroenterology, Yonsei University College of Medicine, Seoul, South Korea
| | - Sang Hoon Ahn
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, South Korea
- Institute of Gastroenterology, Yonsei University College of Medicine, Seoul, South Korea
| | - Hyon-Suk Kim
- Department of Laboratory Medicine, Yonsei University College of Medicine, Seoul, South Korea
| |
Collapse
|
250
|
Saito Y, Morine Y, Shimada M. Mechanism of impairment on liver regeneration in elderly patients: Role of hepatic stellate cell function. Hepatol Res 2017; 47:505-513. [PMID: 28186674 DOI: 10.1111/hepr.12872] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Revised: 02/08/2017] [Accepted: 02/08/2017] [Indexed: 12/13/2022]
Abstract
Japan, along with most other countries in the world, is facing an increasingly aging population with a prolonged life expectancy. Concurrently, the need for medical intervention, including hepatectomy, has also increased for the elderly. Although surgical outcomes for older patients are reported to be comparable with those for younger patients, additional care in the selection of older patients for hepatectomy is considered necessary. Although the effect of aging on human liver regeneration is not fully understood, the regeneration of liver tissue after hepatectomy in elderly patients is shown to be generally worse than in younger patients and, to date, the mechanisms involved in the impairment of liver regeneration have not been fully clarified. Hepatic stellate cells (HSCs) are liver-specific mesenchymal cells that play critical roles in liver physiology and fibrogenesis. Recent studies in liver regeneration have increasingly focused on HSCs rather than on hepatocytes, Kupffer cells, endothelial cells, or infiltrating immune cells and suggest that HSCs might play a critical role in liver regeneration. In this review, we summarize the mechanisms involved in the impairment of liver regeneration in elderly patients, especially focusing on HSCs. We also discuss how HSCs contribute to the impairment of liver regeneration.
Collapse
Affiliation(s)
- Yu Saito
- Department of Surgery, Tokushima University, Tokushima, Japan
| | - Yuji Morine
- Department of Surgery, Tokushima University, Tokushima, Japan
| | - Mitsuo Shimada
- Department of Surgery, Tokushima University, Tokushima, Japan
| |
Collapse
|