101
|
Roth M, Gaceb A, Enström A, Padel T, Genové G, Özen I, Paul G. Regulator of G-protein signaling 5 regulates the shift from perivascular to parenchymal pericytes in the chronic phase after stroke. FASEB J 2019; 33:8990-8998. [PMID: 31039042 PMCID: PMC6662981 DOI: 10.1096/fj.201900153r] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Poststroke recovery requires multiple repair mechanisms, including vascular remodeling and blood-brain barrier (BBB) restoration. Brain pericytes are essential for BBB repair and angiogenesis after stroke, but they also give rise to scar-forming platelet-derived growth factor receptor β (PDGFR-β)–expressing cells. However, many of the molecular mechanisms underlying this pericyte response after stroke still remain unknown. Regulator of G-protein signaling 5 (RGS5) has been associated with pericyte detachment from the vascular wall, but whether it regulates pericyte function and vascular stabilization in the chronic phase of stroke is not known. Using RGS5–knockout (KO) mice, we study how loss of RGS5 affects the pericyte response and vascular remodeling in a stroke model at 7 d after ischemia. Loss of RGS5 leads to a shift toward an increase in the number of perivascular pericytes and reduction in the density of parenchymal PDGFR-β–expressing cells associated with normalized PDGFR-β activation after stroke. The redistribution of pericytes resulted in higher pericyte coverage, increased vascular density, preservation of vessel lengths, and a significant reduction in vascular leakage in RGS5-KO mice compared with controls. Our study demonstrates RGS5 in pericytes as an important target to enhance vascular remodeling.—Roth, M., Gaceb, A., Enström, A., Padel, T., Genové, G., Özen, I., Paul, G. Regulator of G-protein signaling 5 regulates the shift from perivascular to parenchymal pericytes in the chronic phase after stroke.
Collapse
Affiliation(s)
- Michaela Roth
- Translational Neurology Group, Department of Clinical Science, Lund University, Lund, Sweden
| | - Abderahim Gaceb
- Translational Neurology Group, Department of Clinical Science, Lund University, Lund, Sweden
| | - Andreas Enström
- Translational Neurology Group, Department of Clinical Science, Lund University, Lund, Sweden
| | - Thomas Padel
- Translational Neurology Group, Department of Clinical Science, Lund University, Lund, Sweden
| | - Guillem Genové
- Department of Medicine, Integrated Cardio Metabolic Centre, Karolinska Institute, Huddinge, Sweden
| | - Ilknur Özen
- Translational Neurology Group, Department of Clinical Science, Lund University, Lund, Sweden
| | - Gesine Paul
- Translational Neurology Group, Department of Clinical Science, Lund University, Lund, Sweden.,Department of Neurology, Scania University Hospital, Lund, Sweden.,Wallenberg Centre for Molecular Medicine, Lund University, Lund, Sweden
| |
Collapse
|
102
|
Lambrecht J, Verhulst S, Mannaerts I, Sowa JP, Best J, Canbay A, Reynaert H, van Grunsven LA. A PDGFRβ-based score predicts significant liver fibrosis in patients with chronic alcohol abuse, NAFLD and viral liver disease. EBioMedicine 2019; 43:501-512. [PMID: 31036530 PMCID: PMC6558023 DOI: 10.1016/j.ebiom.2019.04.036] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Revised: 04/18/2019] [Accepted: 04/18/2019] [Indexed: 12/15/2022] Open
Abstract
Background Platelet Derived Growth Factor Receptor beta (PDGFRβ) has been associated to hepatic stellate cell activation and has been the target of multiple therapeutic studies. However, little is known concerning its use as a diagnostic agent. Methods Circulating PDGFRβ levels were analysed in a cohort of patients with liver fibrosis/cirrhosis due to chronic alcohol abuse, viral hepatitis, or non-alcoholic fatty liver disease (NAFLD). The diagnostic performance of PDGFRβ as individual blood parameter, or in combination with other metabolic factors was evaluated. Findings sPDGFRβ levels are progressively increased with increasing fibrosis stage and the largest difference was observed in patients with significant fibrosis, compared to no or mild fibrosis. The accuracy of sPDGFRβ-levels predicting fibrosis could be increased by combining it with albumin levels and platelet counts into a novel diagnostic algorithm, the PRTA-score, generating a predictive value superior to Fib-4, APRI, and AST/ALT. The sPDGFRβ levels and the PRTA-score are independent of liver disease aetiology, thus overcoming one of the major weaknesses of current non-invasive clinical and experimental scores. Finally, we confirmed the diagnostic value of sPDGFRβ levels and the PRTA-score in an independent patient cohort with NAFLD which was staged for fibrosis by liver biopsy. Interpretation The PRTA-score is an accurate tool for detecting significant liver fibrosis in a broad range of liver disease aetiologies. Fund Vrije Universiteit Brussel, the Institute for the Promotion of Innovation through Science and Technology in Flanders (IWT-Flanders) (HILIM-3D; SBO140045), and the Fund of Scientific Research Flanders (FWO).
Collapse
Affiliation(s)
- Joeri Lambrecht
- Department of Basic (Bio-)medical Sciences, Liver Cell Biology Research Group, Vrije Universiteit Brussel, Brussels, Belgium
| | - Stefaan Verhulst
- Department of Basic (Bio-)medical Sciences, Liver Cell Biology Research Group, Vrije Universiteit Brussel, Brussels, Belgium
| | - Inge Mannaerts
- Department of Basic (Bio-)medical Sciences, Liver Cell Biology Research Group, Vrije Universiteit Brussel, Brussels, Belgium
| | - Jan-Peter Sowa
- Department of Gastroenterology, Hepatology and Infectious Diseases, University Hospital Magdeburg, Magdeburg, Germany
| | - Jan Best
- Department of Gastroenterology, Hepatology and Infectious Diseases, University Hospital Magdeburg, Magdeburg, Germany
| | - Ali Canbay
- Department of Gastroenterology, Hepatology and Infectious Diseases, University Hospital Magdeburg, Magdeburg, Germany
| | - Hendrik Reynaert
- Department of Basic (Bio-)medical Sciences, Liver Cell Biology Research Group, Vrije Universiteit Brussel, Brussels, Belgium; Department of Gastroenterology and Hepatology, University Hospital Brussels (UZBrussel), Brussels, Belgium
| | - Leo A van Grunsven
- Department of Basic (Bio-)medical Sciences, Liver Cell Biology Research Group, Vrije Universiteit Brussel, Brussels, Belgium.
| |
Collapse
|
103
|
Zhang R, Kikuchi AT, Nakao T, Russell JO, Preziosi ME, Poddar M, Singh S, Bell AW, England SG, Monga SP. Elimination of Wnt Secretion From Stellate Cells Is Dispensable for Zonation and Development of Liver Fibrosis Following Hepatobiliary Injury. Gene Expr 2019; 19:121-136. [PMID: 30236172 PMCID: PMC6466178 DOI: 10.3727/105221618x15373858350141] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Alterations in the Wnt signaling pathway including those impacting hepatic stellate cells (HSCs) have been implicated in liver fibrosis. In the current study, we first examined the expression of Wnt genes in human HSC (HHSCs) after treatment with a profibrogenic factor TGF-β1. Next, we generated HSC-specific Wntless (Wls) knockout (KO) using the Lrat-cre and Wls-floxed mice. KO and littermate controls (CON) were characterized for any basal phenotype and subjected to two liver fibrosis protocols. In vitro, TGF-β1 induced expression of Wnt2, 5a and 9a while decreasing Wnt2b, 3a, 4, and 11 in HHSC. In vivo, KO and CON mice were born at normal Mendelian ratio and lacked any overt phenotype. Loss of Wnt secretion from HSCs had no effect on liver weight and did not impact β-catenin activation in the pericentral hepatocytes. After 7 days of bile duct ligation (BDL), KO and CON showed comparable levels of serum alkaline phosphatase, alanine aminotransferase, aspartate aminotransferase, total and direct bilirubin. Comparable histology, Sirius red staining, and immunohistochemistry for α-SMA, desmin, Ki-67, F4/80, and CD45 indicated similar proliferation, inflammation, and portal fibrosis in both groups. Biweekly administration of carbon tetrachloride for 4 or 8 weeks also led to comparable serum biochemistry, inflammation, and fibrosis in KO and CON. Specific Wnt genes were altered in HHSCs in response to TGF-β1; however, eliminating Wnt secretion from HSC did not impact basal β-catenin activation in normal liver nor did it alter the injury-repair response during development of liver fibrosis.
Collapse
Affiliation(s)
- Rong Zhang
- Department of Pathology, University of Pittsburgh, School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Alexander T Kikuchi
- Department of Pathology, University of Pittsburgh, School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Toshimasa Nakao
- Department of Organ Transplantation and General Surgery, Graduate School of Medical Sciences, Kyoto Prefectural University of Medical School, Hirokoji, Kawaramachi, Kamikyo-ku, Kyoto City, Kyoto, Japan
| | - Jacquelyn O Russell
- Department of Pathology, University of Pittsburgh, School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Morgan E Preziosi
- Department of Pathology, University of Pittsburgh, School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Minakshi Poddar
- Department of Pathology, University of Pittsburgh, School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Sucha Singh
- Department of Pathology, University of Pittsburgh, School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Aaron W Bell
- Department of Pathology, University of Pittsburgh, School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Steven G England
- Future Therapeutics and Technologies, Abbvie, North Chicago, IL, USA
| | - Satdarshan P Monga
- Department of Pathology, University of Pittsburgh, School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| |
Collapse
|
104
|
Lai SS, Fu X, Cheng Q, Yu ZH, Jiang EZ, Zhao DD, Yu DC, Qiu YD, Gao X, Ju HX, Wang W, Jiang Q, Zhu MS, Li CJ, Xue B. HSC-specific knockdown of GGPPS alleviated CCl 4-induced chronic liver fibrosis through mediating RhoA/Rock pathway. Am J Transl Res 2019; 11:2382-2392. [PMID: 31105844 PMCID: PMC6511779] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Accepted: 02/07/2019] [Indexed: 06/09/2023]
Abstract
Hepatic stellate cells (HSCs) play a critical role in the pathogenesis and reversal of liver fibrosis. Targeting HSCs is of great significance in the treatment of hepatic fibrosis, and has attracted wide attention of scholars. Here we demonstrated that expression of geranylgeranyldiphosphate synthase (GGPPS) predominantly increased in HSCs in murine fibrotic liver. HSC-specific knockdown of GGPPS using vitamin A-coupled liposome carrying siRNA-ggpps decreased activation of HSCs and alleviated fiber accumulation in vivo. Furthermore, our in vitro studies showed that GGPPS was up-regulated during HSCs activation in TGF-β1-dependent manner. Inhibition of GGPPS suppressed TGF-β1 induced F-actin reorganization and HSCs activation in LX-2 cells. Further, we found that GGPPS regulated HSCs activation and liver fibrosis possibly by enhancing RhoA/Rock kinase signaling. So its concluded that GGPPS promotes liver fibrosis by activating HSCs, which may represent a potential target for anti-fibrosis therapies.
Collapse
Affiliation(s)
- Shan-Shan Lai
- Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Sciences, Nanjing Normal UniversityNanjing 210023, China
- Jiangsu Key Laboratory of Molecular Medicine and School of Medicine, Nanjing UniversityNanjing 210093, China
| | - Xiao Fu
- Jiangsu Key Laboratory of Molecular Medicine and School of Medicine, Nanjing UniversityNanjing 210093, China
- Department of Hepatobiliary Surgery, The Affiliated Drum Tower Hospital, Medical School of Nanjing UniversityNanjing 210093, China
| | - Qi Cheng
- Jiangsu Key Laboratory of Molecular Medicine and School of Medicine, Nanjing UniversityNanjing 210093, China
| | - Zi-Han Yu
- Jiangsu Key Laboratory of Molecular Medicine and School of Medicine, Nanjing UniversityNanjing 210093, China
| | - En-Ze Jiang
- Jiangsu Key Laboratory of Molecular Medicine and School of Medicine, Nanjing UniversityNanjing 210093, China
| | - Dan-Dan Zhao
- Jiangsu Key Laboratory of Molecular Medicine and School of Medicine, Nanjing UniversityNanjing 210093, China
- Research Center, Shanghai Chest Hospital, Shanghai Jiao Tong UniversityShanghai 200030, China
| | - De-Cai Yu
- Department of Hepatobiliary Surgery, The Affiliated Drum Tower Hospital, Medical School of Nanjing UniversityNanjing 210093, China
| | - Yu-Dong Qiu
- Department of Hepatobiliary Surgery, The Affiliated Drum Tower Hospital, Medical School of Nanjing UniversityNanjing 210093, China
| | - Xiang Gao
- Key Laboratory of Model Animal for Disease Study of Ministry of Education, Model Animal Research Center, Nanjing UniversityNanjing 210093, China
| | - Huang-Xian Ju
- MOE Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing UniversityNanjing 210093, China
| | - Wei Wang
- National Laboratory of Solid State Microstructures, Department of Physics, Nanjing UniversityNanjing 210093, China
| | - Qing Jiang
- Department of Sports Medicine and Adult Reconstructive Surgery, Drum Tower Hospital, School of Medicine, Nanjing UniversityNanjing 210008, China
- Joint Research Center for Bone and Joint Disease, Model Animal Research Center (MARC), Nanjing UniversityNanjing 210093, China
| | - Min-Sheng Zhu
- Key Laboratory of Model Animal for Disease Study of Ministry of Education, Model Animal Research Center, Nanjing UniversityNanjing 210093, China
| | - Chao-Jun Li
- Jiangsu Key Laboratory of Molecular Medicine and School of Medicine, Nanjing UniversityNanjing 210093, China
| | - Bin Xue
- Core Laboratory, Sir Run Run Hospital, Nanjing Medical UniversityNanjing 211166, China
- Jiangsu Key Laboratory of Molecular Medicine and School of Medicine, Nanjing UniversityNanjing 210093, China
- State Key Laboratory of Pharmaceutical Biotechnology, Jiangsu Key Laboratory of Molecular Medicine and School of Medicine, Nanjing UniversityNanjing 210093, China
- State Key Laboratory of Natural Medicines, China Pharmaceutical UniversityNanjing 210009, China
| |
Collapse
|
105
|
Pingitore P, Sasidharan K, Ekstrand M, Prill S, Lindén D, Romeo S. Human Multilineage 3D Spheroids as a Model of Liver Steatosis and Fibrosis. Int J Mol Sci 2019; 20:ijms20071629. [PMID: 30986904 PMCID: PMC6480107 DOI: 10.3390/ijms20071629] [Citation(s) in RCA: 79] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Revised: 03/29/2019] [Accepted: 03/30/2019] [Indexed: 02/06/2023] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is the most common liver disorder in western countries. Despite the high prevalence of NAFLD, the underlying biology of the disease progression is not clear, and there are no approved drugs to treat non-alcoholic steatohepatitis (NASH), the most advanced form of the disease. Thus, there is an urgent need for developing advanced in vitro human cellular systems to study disease mechanisms and drug responses. We attempted to create an organoid system genetically predisposed to NAFLD and to induce steatosis and fibrosis in it by adding free fatty acids. We used multilineage 3D spheroids composed by hepatocytes (HepG2) and hepatic stellate cells (LX-2) with a physiological ratio (24:1). HepG2 and LX-2 cells are homozygotes for the PNPLA3 I148M sequence variant, the strongest genetic determinant of NAFLD. We demonstrate that hepatic stellate cells facilitate the compactness of 3D spheroids. Then, we show that the spheroids develop accumulations of fat and collagen upon exposure to free fatty acids. Finally, this accumulation was rescued by incubating spheroids with liraglutide or elafibranor, drugs that are in clinical trials for the treatment of NASH. In conclusion, we have established a simple, easy to handle, in vitro model of genetically induced NAFLD consisting of multilineage 3D spheroids. This tool may be used to understand molecular mechanisms involved in the early stages of fibrogenesis induced by lipid accumulation. Moreover, it may be used to identify new compounds to treat NASH using high-throughput drug screening.
Collapse
Affiliation(s)
- Piero Pingitore
- Department of Molecular and Clinical Medicine, University of Gothenburg, SE-413 45 Gothenburg, Sweden.
| | - Kavitha Sasidharan
- Department of Molecular and Clinical Medicine, University of Gothenburg, SE-413 45 Gothenburg, Sweden.
| | - Matias Ekstrand
- Department of Molecular and Clinical Medicine, University of Gothenburg, SE-413 45 Gothenburg, Sweden.
| | - Sebastian Prill
- Cardiovascular, Renal and Metabolism, IMED Biotech Unit, AstraZeneca, SE-431 83 Gothenburg, Sweden.
| | - Daniel Lindén
- Cardiovascular, Renal and Metabolism, IMED Biotech Unit, AstraZeneca, SE-431 83 Gothenburg, Sweden.
- Division of Endocrinology, Department of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, SE-405 30 Gothenburg, Sweden.
| | - Stefano Romeo
- Department of Molecular and Clinical Medicine, University of Gothenburg, SE-413 45 Gothenburg, Sweden.
- Clinical Nutrition Unit, Department of Medical and Surgical Sciences, Magna Graecia University, 88100 Catanzaro, Italy.
- Cardiology Department, Sahlgrenska University Hospital, SE-413 45 Gothenburg, Sweden.
| |
Collapse
|
106
|
Rho-kinase inhibitor coupled to peptide-modified albumin carrier reduces portal pressure and increases renal perfusion in cirrhotic rats. Sci Rep 2019; 9:2256. [PMID: 30783172 PMCID: PMC6381202 DOI: 10.1038/s41598-019-38678-5] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Accepted: 01/07/2019] [Indexed: 02/07/2023] Open
Abstract
Rho-kinase (ROCK) activation in hepatic stellate cells (HSC) is a key mechanism promoting liver fibrosis and portal hypertension (PTH). Specific delivery of ROCK-inhibitor Y-27632 (Y27) to HSC targeting mannose-6-phosphate-receptors reduces portal pressure and fibrogenesis. In decompensated cirrhosis, presence of ascites is associated with reduced renal perfusion. Since in cirrhosis, platelet-derived growth factor receptor beta (PDGFRβ) is upregulated in the liver as well as the kidney, this study coupled Y27 to human serum albumin (HSA) substituted with PDGFRβ-recognizing peptides (pPB), and investigated its effect on PTH in cirrhotic rats. In vitro collagen contraction assays tested biological activity on LX2 cells. Hemodynamics were analyzed in BDL and CCl4 cirrhotic rats 3 h, 6 h and 24 h after i.v. administration of Y27pPBHSA (0.5/1 mg/kg b.w). Phosphorylation of moesin and myosin light chain (MLC) assessed ROCK activity in liver, femoral muscle, mesenteric artery, kidney and heart. Three Y27 molecules were coupled to pPBHSA as confirmed by HPLC/MS, which was sufficient to relax LX2 cells. In vivo, Y27pPBHSA-treated rats exhibited lower portal pressure, hepatic vascular resistance without effect on systemic vascular resistance, but a tendency towards lower cardiac output compared to non-treated cirrhotic rats. Y27pPBHSA reduced intrahepatic resistance by reduction of phosphorylation of moesin and MLC in Y27pPBHSA-treated cirrhotic rats. Y27pPBHSA was found in the liver of rats up to 6 hours after its injection, in the HSC demonstrated by double-immunostainings. Interestingly, Y27pPBHSA increased renal arterial flow over time combined with an antifibrotic effect as shown by decreased renal acta2 and col1a1 mRNA expression. Therefore, targeting the ROCK inhibitor Y27 to PDGFRβ decreases portal pressure with potential beneficial effects in the kidney. This unique approach should be tested in human cirrhosis.
Collapse
|
107
|
Li S, Sun X, Chen M, Ying Z, Wan Y, Pi L, Ren B, Cao Q. Liver Fibrosis Conventional and Molecular Imaging Diagnosis Update. JOURNAL OF LIVER 2019; 8:236. [PMID: 31341723 PMCID: PMC6653681] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Liver fibrosis is a serious, life-threatening disease with high morbidity and mortality that result from diverse causes. Liver biopsy, considered the "gold standard" to diagnose, grade, and stage liver fibrosis, has limitations in terms of invasiveness, cost, sampling variability, inter-observer variability, and the dynamic process of fibrosis. Compelling evidence has demonstrated that all stages of fibrosis are reversible if the injury is removed. There is a clear need for safe, effective, and reliable non-invasive assessment modalities to determine liver fibrosis in order to manage it precisely in personalized medicine. However, conventional imaging methods used to assess morphological and structural changes related to liver fibrosis, including ultrasound, computed tomography (CT), and magnetic resonance imaging (MRI), are only useful in assessing advanced liver disease, including cirrhosis. Functional imaging techniques, including MR elastography (MRE), US elastography, and CT perfusion are useful for assessing moderate to advanced liver fibrosis. MRE is considered the most accurate noninvasive imaging technique, and US elastography is currently the most widely used noninvasive means. However, these modalities are less accurate in early-stage liver fibrosis and some factors affect the accuracy of these techniques. Molecular imaging is a target-specific imaging mechanism that has the potential to accurately diagnose early-stage liver fibrosis. We provide an overview of recent advances in molecular imaging for the diagnosis and staging of liver fibrosis which will enable clinicians to monitor the progression of disease and potentially reverse liver fibrosis. We compare the promising technologies with conventional and functional imaging and assess the utility of molecular imaging in precision and personalized clinical medicine in the early stages of liver fibrosis.
Collapse
Affiliation(s)
- Shujing Li
- Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland School of Medicine, Baltimore, Maryland, USA,Department of Radiology, The first affiliated Hospital of Hebei Medical University, Shijiazhuang, Hebei province, P.R.China
| | - Xicui Sun
- Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Minjie Chen
- Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Zhekang Ying
- Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Yamin Wan
- Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland School of Medicine, Baltimore, Maryland, USA,Department of Radiology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan province, P.R.China
| | - Liya Pi
- Department of Pediatrics in the College of Medicine, University of Florida, Gainesville, Florida, USA
| | - Bin Ren
- Department of Surgery, University of Alabama at Birmingham School of Medicine, Alabama, USA
| | - Qi Cao
- Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland School of Medicine, Baltimore, Maryland, USA,Corresponding author: Qi Cao, MD. Ph.D, Department of Diagnostic Radiology and Nuclear Medicine University of Maryland School of Medicine, West Baltimore, Street Baltimore, Maryland, USA, Tel: +1 410-706-6432;,
| |
Collapse
|
108
|
|
109
|
Lim BJ, Lee WK, Lee HW, Lee KS, Kim JK, Chang HY, Lee JI. Selective deletion of hepatocyte platelet-derived growth factor receptor α and development of liver fibrosis in mice. Cell Commun Signal 2018; 16:93. [PMID: 30509307 PMCID: PMC6276164 DOI: 10.1186/s12964-018-0306-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Accepted: 11/21/2018] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND Platelet-derived growth factor receptor α (PDGFRα) expression is increased in activated hepatic stellate cells (HSCs) in cirrhotic liver, while normal hepatocytes express PDGFRα at a negligible level. However, cancerous hepatocytes may show upregulation of PDGFRα, and hepatocellular carcinoma is preceded by chronic liver injury. The role of PDGFRα in non-cancerous hepatocytes and liver fibrosis is unclear. We hypothesized that upon liver injury, PDGFRα in insulted hepatocytes contributes to liver fibrosis by facilitating intercellular crosstalk between hepatocytes and HSCs. METHODS Hepatocytes were isolated from normal and thioacetamide (TAA)-induced cirrhotic livers for assessment of PDGFRα expression. Conditional knock-out (KO) C57BL/6 mice, in which PDGFRα was selectively deleted in hepatocytes, were generated. Liver fibrosis was induced by injecting TAA for 8 weeks. Hep3B cells were transfected with a small interfering RNA (siRNA) (PDGFRα or control) and co-cultured with LX2 cells. RESULTS PDGFRα expression was increased in hepatocytes from fibrotic livers compared to normal livers. Conditional PDGFRα KO mice had attenuated TAA-induced liver fibrosis with decreased HSC activation and proliferation. Immunoblot analyses revealed decreased expression of phospho-p44/42 MAPK in TAA-treated KO mice; these mice also showed almost complete suppression of the upregulation of mouse double minute 2. Although KO mice exhibited increased expression of transforming growth factor (TGF)-β and Smad2/3, this was compensated for by increased expression of inhibitory Smad7. LX2 cells co-cultured with PDGFRα siRNA-infected Hep3B cells showed decreased PDGFRα, α smooth muscle actin, collagen α1(I), TGFβ, and Smad2/3 expression. LX2/PDGFRα-deleted hepatocyte co-culture medium showed decreased PDGF-BB and PDGF-CC levels. CONCLUSIONS Deletion of PDGFRα in hepatocytes attenuated the upregulation of PDGFRα in HSCs after TAA treatment, resulting in decreased liver fibrosis and HSC activation. This suggests that in the event of chronic liver injury, PDGFRα in hepatocytes plays an important role in liver fibrosis by affecting PDGFRα expression in HSCs.
Collapse
Affiliation(s)
- Beom Jin Lim
- Department of Pathology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Woon-Kyu Lee
- Laboratory of Developmental Genetics, Department of Biomedical Sciences, Inha University College of Medicine, Incheon, Republic of Korea
| | - Hyun Woong Lee
- Department of Internal Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Kwan Sik Lee
- Department of Internal Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Ja Kyung Kim
- Department of Internal Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Hye Young Chang
- Medical Research Center, Gangnam Severance Hospital, Seoul, South Korea
| | - Jung Il Lee
- Department of Internal Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
110
|
Jiang Y, Zhao Y, He F, Wang H. Artificial MicroRNA-Mediated Tgfbr2 and Pdgfrb Co-Silencing Ameliorates Carbon Tetrachloride-Induced Hepatic Fibrosis in Mice. Hum Gene Ther 2018; 30:179-196. [PMID: 30024280 DOI: 10.1089/hum.2018.047] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Hepatic stellate cells (HSCs) are the primary cell type responsible for liver fibrogenesis. Transforming growth factor beta 1 (TGF-β1) and platelet-derived growth factor (PDGF) are key profibrotic cytokines that regulate HSC activation and proliferation with functional convergence. Dual RNA interference against their receptors may achieve therapeutic effects. A novel RNAi strategy based on HSC-specific GFAP promoter-driven and lentiviral-expressed artificial microRNAs (amiRNAs) was devised that consists of an microRNA-30a backbone and effective shRNAs against mouse Pdgfrβ and Tgfbr2. Then, its antifibrotic efficacy was tested in primary and cultured HSCs and in mice affected with carbon tetrachloride-induced hepatic fibrosis. The study shows that amiRNA-mediated Pdgfrβ and Tgfbr2 co-silencing inhibits HSC activation and proliferation. After recombinant lentiviral particles were delivered into the liver via tail-vein injection, therapeutic amiRNAs were preferentially expressed in HSCs and efficiently co-knocked down in situ Tgfbr2 and Pdgfrβ expression, which correlates with downregulated expression of target or effector genes of their signaling, which include Pai-1, P70S6K, and D-cyclins. amiRNA-based HSC-specific co-silencing of Tgfbr2 and Pdgfrβ significantly suppressed hepatic expression of fibrotic markers α-Sma and Col1a1, extracellular matrix regulators Mmps and Timp1, and phenotypically ameliorated liver fibrosis, as indicated by reductions in serum alanine aminotransferase activity, collagen deposition, and α-Sma-positive staining. The findings provide proof of concept for the use of amiRNA-mediated co-silencing of two profibrogenic pathways in liver fibrosis treatment and highlight the therapeutic potential of concatenated amiRNAs for gene therapy.
Collapse
Affiliation(s)
- Yan Jiang
- 1 The Fifth People's Hospital of Shanghai, State Key Laboratory of Genetic Engineering and Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Institutes of Biomedical Sciences of Shanghai Medical College, Fudan University, Shanghai, P.R. China
| | - Yuanyuan Zhao
- 1 The Fifth People's Hospital of Shanghai, State Key Laboratory of Genetic Engineering and Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Institutes of Biomedical Sciences of Shanghai Medical College, Fudan University, Shanghai, P.R. China
| | - Fuchu He
- 1 The Fifth People's Hospital of Shanghai, State Key Laboratory of Genetic Engineering and Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Institutes of Biomedical Sciences of Shanghai Medical College, Fudan University, Shanghai, P.R. China.,2 State Key Laboratory of Proteomics, Beijing Institute of Radiation Medicine, Beijing, P.R. China
| | - Haijian Wang
- 1 The Fifth People's Hospital of Shanghai, State Key Laboratory of Genetic Engineering and Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Institutes of Biomedical Sciences of Shanghai Medical College, Fudan University, Shanghai, P.R. China
| |
Collapse
|
111
|
Ai WL, Dong LY, Wang J, Li ZW, Wang X, Gao J, Wu Y, An W. Deficiency in augmenter of liver regeneration accelerates liver fibrosis by promoting migration of hepatic stellate cell. Biochim Biophys Acta Mol Basis Dis 2018; 1864:3780-3791. [DOI: 10.1016/j.bbadis.2018.09.011] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2018] [Revised: 08/19/2018] [Accepted: 09/09/2018] [Indexed: 12/14/2022]
|
112
|
Zhu D, Yang C, Shen P, Chen L, Chen J, Sun X, Duan L, Zhang L, Zhu J, Duan Y. rSjP40 suppresses hepatic stellate cell activation by promoting microRNA-155 expression and inhibiting STAT5 and FOXO3a expression. J Cell Mol Med 2018; 22:5486-5493. [PMID: 30091834 PMCID: PMC6201359 DOI: 10.1111/jcmm.13819] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2018] [Revised: 06/12/2018] [Accepted: 06/29/2018] [Indexed: 01/09/2023] Open
Abstract
Activation of hepatic stellate cells (HSCs) is the central event of the evolution of hepatic fibrosis. Schistosomiasis is one of the pathogenic factors which could induce hepatic fibrosis. Previous studies have shown that recombinant Schistosoma japonicum egg antigen P40 (rSjP40) can inhibit the activation and proliferation of HSCs. MicroRNA‐155 is one of the multifunctional noncoding RNA, which is involved in a series of important biological processes including cell development, proliferation, differentiation and apoptosis. Here, we try to observe the role of microRNA‐155 in rSjP40‐inhibited HSC activation and explore its potential mechanisms. We found that microRNA‐155 was raised in rSjP40‐treated HSCs, and further studies have shown that rSjP40 enhanced microRNA‐155 expression by inhibiting STAT5 transcription. Up‐regulated microRNA‐155 can down‐regulate the expression of FOXO3a and then participate in rSjP40‐inhibited expression of α‐smooth muscle actin (α‐SMA) and collagen I. Furthermore, we observed microRNA‐155 inhibitor could partially restore the down‐regulation of FOXO3a, α‐SMA and collagen I expression in LX‐2 cells induced by rSjP40. Therefore, our research provides further insight into the mechanism by which rSjP40 could inhibit HSC activation via miR‐155.
Collapse
Affiliation(s)
- Dandan Zhu
- Department of Pathogen Biology, School of Medicine, Nantong University, Nantong, China
| | - Chunzhao Yang
- Department of Pathogen Biology, School of Medicine, Nantong University, Nantong, China
| | - Pei Shen
- Laboratory Medicine Center, Affiliated Hospital of Nantong University, Nantong, China
| | - Liuting Chen
- Department of Pathogen Biology, School of Medicine, Nantong University, Nantong, China
| | - Jinling Chen
- Department of Pathogen Biology, School of Medicine, Nantong University, Nantong, China
| | - Xiaolei Sun
- Department of Pathogen Biology, School of Medicine, Nantong University, Nantong, China
| | - Lian Duan
- Department of Medical Informatics, School of Medicine, Nantong University, Nantong, China
| | - Li Zhang
- Department of Pathogen Biology, School of Medicine, Nantong University, Nantong, China
| | - Jinhua Zhu
- Department of Pathogen Biology, School of Medicine, Nantong University, Nantong, China
| | - Yinong Duan
- Department of Pathogen Biology, School of Medicine, Nantong University, Nantong, China
| |
Collapse
|
113
|
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
|
114
|
Abderrahmani A, Yengo L, Caiazzo R, Canouil M, Cauchi S, Raverdy V, Plaisance V, Pawlowski V, Lobbens S, Maillet J, Rolland L, Boutry R, Queniat G, Kwapich M, Tenenbaum M, Bricambert J, Saussenthaler S, Anthony E, Jha P, Derop J, Sand O, Rabearivelo I, Leloire A, Pigeyre M, Daujat-Chavanieu M, Gerbal-Chaloin S, Dayeh T, Lassailly G, Mathurin P, Staels B, Auwerx J, Schürmann A, Postic C, Schafmayer C, Hampe J, Bonnefond A, Pattou F, Froguel P. Increased Hepatic PDGF-AA Signaling Mediates Liver Insulin Resistance in Obesity-Associated Type 2 Diabetes. Diabetes 2018; 67:1310-1321. [PMID: 29728363 DOI: 10.2337/db17-1539] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Accepted: 04/26/2018] [Indexed: 12/17/2022]
Abstract
In type 2 diabetes (T2D), hepatic insulin resistance is strongly associated with nonalcoholic fatty liver disease (NAFLD). In this study, we hypothesized that the DNA methylome of livers from patients with T2D compared with livers of individuals with normal plasma glucose levels can unveil some mechanism of hepatic insulin resistance that could link to NAFLD. Using DNA methylome and transcriptome analyses of livers from obese individuals, we found that hypomethylation at a CpG site in PDGFA (encoding platelet-derived growth factor α) and PDGFA overexpression are both associated with increased T2D risk, hyperinsulinemia, increased insulin resistance, and increased steatohepatitis risk. Genetic risk score studies and human cell modeling pointed to a causative effect of high insulin levels on PDGFA CpG site hypomethylation, PDGFA overexpression, and increased PDGF-AA secretion from the liver. We found that PDGF-AA secretion further stimulates its own expression through protein kinase C activity and contributes to insulin resistance through decreased expression of insulin receptor substrate 1 and of insulin receptor. Importantly, hepatocyte insulin sensitivity can be restored by PDGF-AA-blocking antibodies, PDGF receptor inhibitors, and by metformin, opening therapeutic avenues. Therefore, in the liver of obese patients with T2D, the increased PDGF-AA signaling contributes to insulin resistance, opening new therapeutic avenues against T2D and possibly NAFLD.
Collapse
Affiliation(s)
- Amar Abderrahmani
- University Lille, Centre National de la Recherche Scientifique, Institut Pasteur de Lille, UMR 8199 - European Genomic Institute for Diabetes, Lille, France
- Section of Genomics of Common Disease, Department of Medicine, Imperial College London, London, U.K
| | - Loïc Yengo
- University Lille, Centre National de la Recherche Scientifique, Institut Pasteur de Lille, UMR 8199 - European Genomic Institute for Diabetes, Lille, France
| | - Robert Caiazzo
- University Lille, INSERM, CHU Lille, U1190 - European Genomic Institute for Diabetes, Lille, France
| | - Mickaël Canouil
- University Lille, Centre National de la Recherche Scientifique, Institut Pasteur de Lille, UMR 8199 - European Genomic Institute for Diabetes, Lille, France
| | - Stéphane Cauchi
- University Lille, Centre National de la Recherche Scientifique, Institut Pasteur de Lille, UMR 8199 - European Genomic Institute for Diabetes, Lille, France
| | - Violeta Raverdy
- University Lille, INSERM, CHU Lille, U1190 - European Genomic Institute for Diabetes, Lille, France
| | - Valérie Plaisance
- University Lille, Centre National de la Recherche Scientifique, Institut Pasteur de Lille, UMR 8199 - European Genomic Institute for Diabetes, Lille, France
| | - Valérie Pawlowski
- University Lille, Centre National de la Recherche Scientifique, Institut Pasteur de Lille, UMR 8199 - European Genomic Institute for Diabetes, Lille, France
| | - Stéphane Lobbens
- University Lille, Centre National de la Recherche Scientifique, Institut Pasteur de Lille, UMR 8199 - European Genomic Institute for Diabetes, Lille, France
| | - Julie Maillet
- University Lille, Centre National de la Recherche Scientifique, Institut Pasteur de Lille, UMR 8199 - European Genomic Institute for Diabetes, Lille, France
| | - Laure Rolland
- University Lille, Centre National de la Recherche Scientifique, Institut Pasteur de Lille, UMR 8199 - European Genomic Institute for Diabetes, Lille, France
| | - Raphael Boutry
- University Lille, Centre National de la Recherche Scientifique, Institut Pasteur de Lille, UMR 8199 - European Genomic Institute for Diabetes, Lille, France
| | - Gurvan Queniat
- University Lille, Centre National de la Recherche Scientifique, Institut Pasteur de Lille, UMR 8199 - European Genomic Institute for Diabetes, Lille, France
| | - Maxime Kwapich
- University Lille, Centre National de la Recherche Scientifique, Institut Pasteur de Lille, UMR 8199 - European Genomic Institute for Diabetes, Lille, France
| | - Mathie Tenenbaum
- University Lille, Centre National de la Recherche Scientifique, Institut Pasteur de Lille, UMR 8199 - European Genomic Institute for Diabetes, Lille, France
| | - Julien Bricambert
- University Lille, Centre National de la Recherche Scientifique, Institut Pasteur de Lille, UMR 8199 - European Genomic Institute for Diabetes, Lille, France
| | - Sophie Saussenthaler
- Department of Experimental Diabetology, German Institute of Human Nutrition Potsdam-Rehbrücke, Nuthetal, and German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| | - Elodie Anthony
- Inserm U1016, Institut Cochin, Centre National de la Recherche Scientifique UMR 8104, Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Pooja Jha
- Laboratory of Integrative Systems Physiology, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Julien Derop
- University Lille, Centre National de la Recherche Scientifique, Institut Pasteur de Lille, UMR 8199 - European Genomic Institute for Diabetes, Lille, France
| | - Olivier Sand
- University Lille, Centre National de la Recherche Scientifique, Institut Pasteur de Lille, UMR 8199 - European Genomic Institute for Diabetes, Lille, France
| | - Iandry Rabearivelo
- University Lille, Centre National de la Recherche Scientifique, Institut Pasteur de Lille, UMR 8199 - European Genomic Institute for Diabetes, Lille, France
| | - Audrey Leloire
- University Lille, Centre National de la Recherche Scientifique, Institut Pasteur de Lille, UMR 8199 - European Genomic Institute for Diabetes, Lille, France
| | - Marie Pigeyre
- University Lille, INSERM, CHU Lille, U1190 - European Genomic Institute for Diabetes, Lille, France
| | - Martine Daujat-Chavanieu
- INSERM U1183, University Montpellier, Institute for Regenerative Medicine and Biotherapy, CHU Montpellier, France
| | - Sabine Gerbal-Chaloin
- INSERM U1183, University Montpellier, Institute for Regenerative Medicine and Biotherapy, CHU Montpellier, France
| | - Tasnim Dayeh
- Department of Clinical Science, Skane University Hospital Malmö, Malmö, Sweden
| | - Guillaume Lassailly
- University Lille, INSERM, CHU Lille, U995 - Lille Inflammation Research International Center, Lille, France
| | - Philippe Mathurin
- University Lille, INSERM, CHU Lille, U995 - Lille Inflammation Research International Center, Lille, France
| | - Bart Staels
- University Lille, INSERM, CHU Lille, Institut Pasteur de Lille, U1011- European Genomic Institute for Diabetes, Lille, France
| | - Johan Auwerx
- Laboratory of Integrative Systems Physiology, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Annette Schürmann
- Department of Experimental Diabetology, German Institute of Human Nutrition Potsdam-Rehbrücke, Nuthetal, and German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| | - Catherine Postic
- Inserm U1016, Institut Cochin, Centre National de la Recherche Scientifique UMR 8104, Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Clemens Schafmayer
- Department of Visceral and Thoracic Surgery, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Jochen Hampe
- Medical Department 1, Technische Universität Dresden, Dresden, Germany
| | - Amélie Bonnefond
- University Lille, Centre National de la Recherche Scientifique, Institut Pasteur de Lille, UMR 8199 - European Genomic Institute for Diabetes, Lille, France
- Section of Genomics of Common Disease, Department of Medicine, Imperial College London, London, U.K
| | - François Pattou
- University Lille, INSERM, CHU Lille, U1190 - European Genomic Institute for Diabetes, Lille, France
| | - Philippe Froguel
- University Lille, Centre National de la Recherche Scientifique, Institut Pasteur de Lille, UMR 8199 - European Genomic Institute for Diabetes, Lille, France
- Section of Genomics of Common Disease, Department of Medicine, Imperial College London, London, U.K
| |
Collapse
|
115
|
Ding Q, Li Z, Liu B, Ling L, Tian X, Zhang C. Propranolol prevents liver cirrhosis by inhibiting hepatic stellate cell activation mediated by the PDGFR/Akt pathway. Hum Pathol 2018. [PMID: 29514109 DOI: 10.1016/j.humpath.2018.02.018] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Propranolol is known to reduce portal pressure by decreasing blood flow to the splanchnic circulation and the liver. However, it is unknown if propranolol improves fibrogenesis and sinusoidal remodeling in the cirrhotic liver. The aim of this study was to investigate the therapeutic effects of propranolol on carbon tetrachloride (CCl4)-induced liver fibrosis in a mouse model and the intrinsic mechanisms underlying those effects. In this study, a hepatic cirrhosis mouse model was induced by CCl4 administration for 6 weeks. Propranolol was simultaneously administered orally in the experimental group. Liver tissue and blood samples were collected for histological and molecular analyses. LX-2 cells induced by platelet-derived growth factor-BB (PDGF-BB) were used to evaluate the anti-fibrogenic effect of propranolol in vitro. The results showed that treatment of mice with CCl4 induced hepatic fibrosis, as evidenced by inflammatory cell infiltration, collagen deposition and abnormal vascular formation in the liver tissue. All these changes were significantly attenuated by propranolol treatment. Furthermore, we also found that propranolol inhibited PDGF-BB-induced hepatic stellate cell migration, fibrogenesis, and PDGFR/Akt phosphorylation. Taken together, propranolol might prevent CCl4-induced liver injury and fibrosis at least partially through inhibiting the PDGF-BB-induced PDGFR/Akt pathway. The anti-fibrogenic effect of propranolol may support its status as a first-line treatment in patients with chronic liver disease.
Collapse
Affiliation(s)
- Qian Ding
- Department of Gastroenterology, Shandong Provincial Hospital affiliated to Shandong University, Jinan 250021, China
| | - Zhen Li
- Department of Gastroenterology, Shandong Provincial Hospital affiliated to Shandong University, Jinan 250021, China; Shandong Provincial Engineering and Technological Research Center for Liver Diseases Prevention and Control, Jinan, China
| | - Bin Liu
- Department of Neurosurgery, Kyoto University Graduate School of Medicine, Kyoto 606-8507 Japan
| | - Liping Ling
- Department of Gastroenterology, Shandong Provincial Hospital affiliated to Shandong University, Jinan 250021, China; Shandong Provincial Engineering and Technological Research Center for Liver Diseases Prevention and Control, Jinan, China
| | - Xiangguo Tian
- Department of Gastroenterology, Shandong Provincial Hospital affiliated to Shandong University, Jinan 250021, China
| | - Chunqing Zhang
- Department of Gastroenterology, Shandong Provincial Hospital affiliated to Shandong University, Jinan 250021, China.
| |
Collapse
|
116
|
Abstract
Fibrosis is part of a tissue repair response to injury, defined as increased deposition of extracellular matrix. In some instances, fibrosis is beneficial; however, in the majority of diseases fibrosis is detrimental. Virtually all chronic progressive diseases are associated with fibrosis, representing a huge number of patients worldwide. Fibrosis occurs in all organs and tissues, becomes irreversible with time and further drives loss of tissue function. Various cells types initiate and perpetuate pathological fibrosis by paracrine activation of the principal cellular executors of fibrosis, i.e. stromal mesenchymal cells like fibroblasts, pericytes and myofibroblasts. Multiple pathways are involved in fibrosis, platelet-derived growth factor (PDGF)-signaling being one of the central mediators. Stromal mesenchymal cells express both PDGF receptors (PDGFR) α and β, activation of which drives proliferation, migration and production of extracellular matrix, i.e. the principal processes of fibrosis. Here, we review the role of PDGF signaling in organ fibrosis, with particular focus on the more recently described ligands PDGF-C and -D. We discuss the potential challenges, opportunities and open questions in using PDGF as a potential target for anti-fibrotic therapies.
Collapse
Affiliation(s)
| | - Jürgen Floege
- Division of Nephrology, RWTH University of Aachen, Germany
| | - Peter Boor
- Institute of Pathology, RWTH University of Aachen, Germany; Division of Nephrology, RWTH University of Aachen, Germany.
| |
Collapse
|
117
|
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: 906] [Impact Index Per Article: 129.4] [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
|
118
|
Lv X, Fang C, Yin R, Qiao B, Shang R, Wang J, Song W, He Y, Chen Y. Agrin para-secreted by PDGF-activated human hepatic stellate cells promotes hepatocarcinogenesis in vitro and in vivo. Oncotarget 2017; 8:105340-105355. [PMID: 29285255 PMCID: PMC5739642 DOI: 10.18632/oncotarget.22186] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Accepted: 07/25/2017] [Indexed: 12/22/2022] Open
Abstract
Evaluating the process and mechanism of fibrogenesis is essential in hepatocellular carcinoma (HCC), especially in hepatocyte transformation and oncogenic signaling. We evaluated the oncogenic role of agrin secreted by platelet-derived growth factor (PDGF)-induced hepatic stellate cell (HSC) in HCC. Cells were co-cultured to investigate the effect of activated HSC on hepatocytes. Liquid chromatography and protein profiling analysis were used to search the distinct proteins secreted in HSC supernatant. Sprague Dawley rats with Diethylnitrosamine (DEN)-induced HCC were used to simulate human liver cancer and sorafenib was administered to investigate its effect on hepatocarcinogenesis. A paired "two-tailed" Student t-test and chi-square tests was used for statistical analysis. PDGF acted as an activator of the HSC and sorafenib inhibits the activation by blocking the combination of PDGF and PDGF receptor. The supernatant of activated HSCs promoted the proliferation, metastasis, and invasion of HL-7702 and SMMC-7721, as well as epithelial-mesenchymal transition (EMT). Agrin found in the HSC supernatant showed the same effect on SMMC-7721 as to the supernatant of activated LX-2. Furthermore, downregulation of agrin by siRNA could decrease the proliferation, metastasis, and invasion of SMMC-7721, and promote MET. Sorafenib prevented DEN-induced hepatocarcinogenesis and could alleviate the liver inflammation and fibrosis. Sorafenib could improve the liver function of Sprague Dawley rats by decreasing the serum levels of ALT and AST. These results demonstrate thatPDGF is an effective activator of HSC and sorafenib could inhibit the activation. In vivo experiment suggested sorafenib could alleviate the hepatocarcinogenesis mediated through agrin secretion and could be potential candidate for treatment of cirrhosis.
Collapse
Affiliation(s)
- Xing Lv
- Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, P.R. China
| | - Cheng Fang
- Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, P.R. China
| | - Ruozhe Yin
- Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, P.R. China
| | - Bowei Qiao
- Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, P.R. China
| | - Runze Shang
- Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, P.R. China
| | - Jianlin Wang
- Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, P.R. China
| | - Wenjie Song
- Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, P.R. China
| | - Yong He
- Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, P.R. China
| | - Yong Chen
- Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, P.R. China
| |
Collapse
|
119
|
Ying HZ, Chen Q, Zhang WY, Zhang HH, Ma Y, Zhang SZ, Fang J, Yu CH. PDGF signaling pathway in hepatic fibrosis pathogenesis and therapeutics (Review). Mol Med Rep 2017; 16:7879-7889. [PMID: 28983598 PMCID: PMC5779870 DOI: 10.3892/mmr.2017.7641] [Citation(s) in RCA: 140] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Accepted: 07/20/2017] [Indexed: 02/06/2023] Open
Abstract
The platelet‑derived growth factor (PDFG) signaling pathway exerts persistent activation in response to a variety of stimuli and facilitates the progression of hepatic fibrosis. Since this pathway modulates a broad spectrum of cellular processes, including cell growth, differentiation, inflammation and carcinogenesis, it has emerged as a therapeutic target for hepatic fibrosis and liver‑associated disorders. The present review exhibits the current knowledge of the role of the PDGF signaling pathway and its pathological profiles in hepatic fibrosis, and assesses the potential of inhibitors which have been investigated in the experimental hepatic fibrosis model, in addition to the clinical challenges associated with these inhibitors.
Collapse
Affiliation(s)
- Hua-Zhong Ying
- Key Laboratory of Experimental Animal and Safety Evaluation, Zhejiang Academy of Medical Sciences, Hangzhou, Zhejiang 310013, P.R. China
| | - Qin Chen
- Department of Clinical Laboratory Medicine, Second Affiliated Hospital, Zhejiang University College of Medicine, Hangzhou, Zhejiang 310009, P.R. China
| | - Wen-You Zhang
- Key Laboratory of Experimental Animal and Safety Evaluation, Zhejiang Academy of Medical Sciences, Hangzhou, Zhejiang 310013, P.R. China
| | - Huan-Huan Zhang
- Key Laboratory of Experimental Animal and Safety Evaluation, Zhejiang Academy of Medical Sciences, Hangzhou, Zhejiang 310013, P.R. China
| | - Yue Ma
- Key Laboratory of Experimental Animal and Safety Evaluation, Zhejiang Academy of Medical Sciences, Hangzhou, Zhejiang 310013, P.R. China
| | - Song-Zhao Zhang
- Department of Clinical Laboratory Medicine, Second Affiliated Hospital, Zhejiang University College of Medicine, Hangzhou, Zhejiang 310009, P.R. China
| | - Jie Fang
- Key Laboratory of Experimental Animal and Safety Evaluation, Zhejiang Academy of Medical Sciences, Hangzhou, Zhejiang 310013, P.R. China
| | - Chen-Huan Yu
- Key Laboratory of Experimental Animal and Safety Evaluation, Zhejiang Academy of Medical Sciences, Hangzhou, Zhejiang 310013, P.R. China
| |
Collapse
|
120
|
Y-box Protein-1 Regulates the Expression of Collagen I in Hepatic Progenitor Cells via PDGFR- β/ERK/p90RSK Signalling. Stem Cells Int 2017; 2017:6193106. [PMID: 28928774 PMCID: PMC5591999 DOI: 10.1155/2017/6193106] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2017] [Accepted: 04/06/2017] [Indexed: 12/20/2022] Open
Abstract
Y-box protein-1 (YB-1) is a highly conserved transcription factor that is involved in multiple biological processes via transcriptional regulation of several genes, including p53, cyclin D1, and EGFR. YB-1 has been reported to be overexpressed in injured livers. This study aims to explore the functions of YB-1 in hepatic progenitor cells (HPCs). Herein, chromatin immunoprecipitation sequencing (ChIP-sequencing) and RNA-sequencing assays identified that YB-1 participated in the biological adhesion process and ECM-receptor interactions in HPCs. Further study demonstrated that YB-1 modulated the expression of extracellular matrix components in HPCs. ChIP-sequencing assays established that PDGFR-β was a target gene of YB-1, and luciferase reporter assays confirmed that YB-1 negatively regulated PDGFR-β promoter activity in HPCs. In addition, PDGFR-β can regulate the expression of collagen I through ERK/p90RSK signalling, and disruption of the signalling pathway with a PDGFR-β inhibitor or ERK1/2 inhibitor abolished the regulatory effect of PDGFR-β on collagen I expression in HPCs. Conclusively, YB-1 can modulate the expression of collagen I in HPCs via direct binding to the PDGFR-β promoter, negatively regulating its expression. In addition, the ERK/p90RSK axis serves as the downstream signalling pathway of PDGFR-β.
Collapse
|
121
|
Kikuchi A, Pradhan-Sundd T, Singh S, Nagarajan S, Loizos N, Monga SP. Platelet-Derived Growth Factor Receptor α Contributes to Human Hepatic Stellate Cell Proliferation and Migration. THE AMERICAN JOURNAL OF PATHOLOGY 2017; 187:2273-2287. [PMID: 28734947 DOI: 10.1016/j.ajpath.2017.06.009] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Revised: 05/19/2017] [Accepted: 06/08/2017] [Indexed: 02/06/2023]
Abstract
Platelet-derived growth factor receptor α (PDGFRα), a tyrosine kinase receptor, is up-regulated in hepatic stellate cells (HSCs) during chronic liver injury. HSCs mediate hepatic fibrosis through their activation from a quiescent state partially in response to profibrotic growth factors. HSC activation entails enhanced expression of profibrotic genes, increase in proliferation, and increase in motility, which facilitates migration within the hepatic lobule. We show colocalization of PDGFRα in murine carbon tetrachloride, bile duct ligation, and 0.1% 3,5-diethoxycarbonyl-1,4-dihydrocollidine models of chronic liver injury, and investigate the role of PDGFRα on proliferation, profibrotic gene expression, and migration in primary human HSCs (HHSteCs) using the PDGFRα-specific inhibitory monoclonal antibody olaratumab. Although lacking any effects on HHSteC transdifferentiation assessed by gene expression of ACTA2, TGFB1, COL1A1, SYP1, and FN1, olaratumab specifically reduced HHSteC proliferation (AlamarBlue assay) and cell migration (transwell migration assays). Using phospho-specific antibodies, we show that olaratumab attenuates PDGFRα activation in response to PDGF-BB, and reduced phosphorylation of extracellular signal-regulated kinase 1 and 2, Elk-1, p38, Akt, focal adhesion kinase, mechanistic target of rapamycin, C10 regulator of kinase II, and C10 regulator of kinase-like, suggesting that PDGFRα contributes to mitogenesis and actin reorganization through diverse downstream effectors. Our findings support a distinct contribution of PDGFRα signaling to HSC proliferation and migration and provide evidence that inhibition of PDGFRα signaling could alter the pathogenesis of hepatic fibrosis.
Collapse
Affiliation(s)
- Alexander Kikuchi
- Department of Pathology and Medicine and Pittsburgh Liver Research Center, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Tirthadipa Pradhan-Sundd
- Department of Pathology and Medicine and Pittsburgh Liver Research Center, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Sucha Singh
- Department of Pathology and Medicine and Pittsburgh Liver Research Center, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Shanmugam Nagarajan
- Department of Pathology and Medicine and Pittsburgh Liver Research Center, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Nick Loizos
- Department of Immunology, Eli Lilly and Company, New York, New York
| | - Satdarshan P Monga
- Department of Pathology and Medicine and Pittsburgh Liver Research Center, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania.
| |
Collapse
|
122
|
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
|
123
|
Abstract
Understanding the underlying molecular mechanisms of liver fibrosis is important to develop effective therapy. Herein, we show that focal-adhesion-kinse (FAK) plays a key role in promoting hepatic stellate cells (HSCs) activation in vitro and liver fibrosis progression in vivo. FAK activation is associated with increased expression of α-smooth muscle actin (α-SMA) and collagen in fibrotic live tissues. Transforming growth factor beta-1 (TGF-β1) induces FAK activation in a time and dose dependent manner. FAK activation precedes the α-SMA expression in HSCs. Inhibition of FAK activation blocks the α-SMA and collagen expression, and inhibits the formation of stress fibers in TGF-β1 treated HSCs. Furthermore, inhibition of FAK activation significantly reduces HSC migration and small GTPase activation, and induces apoptotic signaling in TGF-β1 treated HSCs. Importantly, FAK inhibitor attenuates liver fibrosis in vivo and significantly reduces collagen and α-SMA expression in an animal model of liver fibrosis. These data demonstrate that FAK plays an essential role in HSC activation and liver fibrosis progression, and FAK signaling pathway could be a potential target for liver fibrosis.
Collapse
|
124
|
Hall C, Sato K, Wu N, Zhou T, Kyritsi K, Meng F, Glaser S, Alpini G. Regulators of Cholangiocyte Proliferation. Gene Expr 2017; 17:155-171. [PMID: 27412505 PMCID: PMC5494439 DOI: 10.3727/105221616x692568] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Cholangiocytes, a small population of cells within the normal liver, have been the focus of a significant amount of research over the past two decades because of their involvement in cholangiopathies such as primary sclerosing cholangitis and primary biliary cholangitis. This article summarizes landmark studies in the field of cholangiocyte physiology and aims to provide an updated review of biliary pathogenesis. The historical approach of rodent extrahepatic bile duct ligation and the relatively recent utilization of transgenic mice have led to significant discoveries in cholangiocyte pathophysiology. Cholangiocyte physiology is a complex system based on heterogeneity within the biliary tree and a number of signaling pathways that serve to regulate bile composition. Studies have expanded the list of neuropeptides, neurotransmitters, and hormones that have been shown to be key regulators of proliferation and biliary damage. The peptide histamine and hormones, such as melatonin and angiotensin, angiotensin, as well as numerous sex hormones, have been implicated in cholangiocyte proliferation during cholestasis. Numerous pathways promote cholangiocyte proliferation during cholestasis, and there is growing evidence to suggest that cholangiocyte proliferation may promote hepatic fibrosis. These pathways may represent significant therapeutic potential for a subset of cholestatic liver diseases that currently lack effective therapies.
Collapse
Affiliation(s)
- Chad Hall
- *Research, Central Texas Veterans Health Care System, Temple, TX, USA
- †Baylor Scott & White Digestive Disease Research Center, Temple, TX, USA
- ‡Department of Surgery, Baylor Scott & White and Texas A&M Health Science Center, Temple, TX, USA
| | - Keisaku Sato
- §Operational Funds, Baylor Scott & White, Temple, TX, USA
| | - Nan Wu
- §Operational Funds, Baylor Scott & White, Temple, TX, USA
| | - Tianhao Zhou
- §Operational Funds, Baylor Scott & White, Temple, TX, USA
| | | | - Fanyin Meng
- *Research, Central Texas Veterans Health Care System, Temple, TX, USA
- §Operational Funds, Baylor Scott & White, Temple, TX, USA
- ¶Department of Medicine, Baylor Scott & White and Texas A&M Health Science Center, Temple, TX, USA
| | - Shannon Glaser
- *Research, Central Texas Veterans Health Care System, Temple, TX, USA
- §Operational Funds, Baylor Scott & White, Temple, TX, USA
- ¶Department of Medicine, Baylor Scott & White and Texas A&M Health Science Center, Temple, TX, USA
| | - Gianfranco Alpini
- ‡Department of Surgery, Baylor Scott & White and Texas A&M Health Science Center, Temple, TX, USA
- §Operational Funds, Baylor Scott & White, Temple, TX, USA
- ¶Department of Medicine, Baylor Scott & White and Texas A&M Health Science Center, Temple, TX, USA
| |
Collapse
|
125
|
Expression of muscarinic acetylcholine receptors in hepatocytes from rat fibrotic liver. ACTA ACUST UNITED AC 2017; 69:73-81. [DOI: 10.1016/j.etp.2016.11.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Revised: 10/07/2016] [Accepted: 11/21/2016] [Indexed: 01/11/2023]
|
126
|
Rókusz A, Bugyik E, Szabó V, Szücs A, Paku S, Nagy P, Dezső K. Imatinib accelerates progenitor cell-mediated liver regeneration in choline-deficient ethionine-supplemented diet-fed mice. Int J Exp Pathol 2016; 97:389-396. [PMID: 27918111 DOI: 10.1111/iep.12209] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2016] [Accepted: 09/10/2016] [Indexed: 12/12/2022] Open
Abstract
Severe chronic hepatic injury can induce complex reparative processes. Ductular reaction and the appearance of small hepatocytes are standard components of this response, which is thought to have both adverse (e.g. fibrosis, carcinogenesis) and beneficial (regeneration) consequences. This complex tissue reaction is regulated by orchestrated cytokine action. We have investigated the influence of the tyrosine kinase inhibitor imatinib on a regenerative process. Ductular reaction was induced in mice by the widely used choline-deficient ethionine-supplemented diet (CDE). Test animals were treated daily with imatinib. After 6 weeks of treatment, imatinib successfully reduced the extent of ductular reaction and fibrosis in the CDE model. Furthermore, the number of small hepatocytes increased, and these cells had high proliferative activity, were positive for hepatocyte nuclear factor 4 and expressed high levels of albumin and peroxisome proliferator-activated receptor alpha. The overall functional zonality of the hepatic parenchyma (cytochrome P450 2E1 and glucose 6 phosphatase activity; endogenous biotin content) was maintained. The expression of platelet-derived growth factor receptor beta, which is the major target of imatinib, was downregulated. The anti-fibrotic activity of imatinib has already been reported in several experimental models. Additionally, in the CDE model imatinib was able to enhance regeneration and preserve the functional arrangement of hepatic lobules. These results suggest that imatinib might promote the recovery of the liver following parenchymal injury through the inhibition of platelet-derived growth factor receptor beta.
Collapse
Affiliation(s)
- András Rókusz
- First Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - Edina Bugyik
- First Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - Vanessza Szabó
- First Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - Armanda Szücs
- First Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - Sándor Paku
- First Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary.,Tumor Progression Research Group, Joint Research Organization of the Hungarian Academy of Sciences and Semmelweis University, Budapest, Hungary
| | - Péter Nagy
- First Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - Katalin Dezső
- First Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| |
Collapse
|
127
|
Akihara R, Homma T, Lee J, Yamada KI, Miyata S, Fujii J. Ablation of aldehyde reductase aggravates carbon tetrachloride-induced acute hepatic injury involving oxidative stress and endoplasmic reticulum stress. Biochem Biophys Res Commun 2016; 478:765-71. [DOI: 10.1016/j.bbrc.2016.08.022] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Accepted: 08/04/2016] [Indexed: 11/16/2022]
|
128
|
Kim KH, Lee JM, Zhou Y, Harpavat S, Moore DD. Glucocorticoids Have Opposing Effects on Liver Fibrosis in Hepatic Stellate and Immune Cells. Mol Endocrinol 2016; 30:905-16. [PMID: 27355192 DOI: 10.1210/me.2016-1029] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Liver fibrosis is a reversible wound-healing process that is protective in the short term, but prolonged fibrotic responses lead to excessive accumulation of extracellular matrix components that suppresses hepatocyte regeneration, resulting in permanent liver damage. Upon liver damage, nonparenchymal cells including immune cells and hepatic stellate cells (HSCs) have crucial roles in the progression and regression of liver fibrosis. Here, we report differential roles of the glucocorticoid receptor (GR), acting in immune cells and HSCs, in liver fibrosis. In the carbon tetrachloride hepatotoxin-induced fibrosis model, both steroidal and nonsteroidal GR ligands suppressed expression of fibrotic genes and decreased extracellular matrix deposition but also inhibited immune cell infiltration and exacerbated liver injury. These counteracting effects of GR ligands were dissociated in mice with conditional GR knockout in immune cells (GR(LysM)) or HSC (GR(hGFAP)): the impacts of dexamethasone on immune cell infiltration and liver injury were totally blunted in GR(LysM) mice, whereas the suppression of fibrotic gene expression was diminished in GR(hGFAP) mice. The effect of GR activation in HSC was further confirmed in the LX-2 HSC cell line, in which antifibrotic effects were mediated by GR ligand inhibition of Sma and mad-related protein 3 (SMAD3) expression. We conclude that GR has differential roles in immune cells and HSCs to modulate liver injury and liver fibrosis. Specific activation of HSC-GR without alteration of GR activity in immune cells provides a potential therapeutic approach to treatment of hepatic fibrosis.
Collapse
Affiliation(s)
- Kang Ho Kim
- Department of Molecular and Cellular Biology (K.H.K., J.M.L., Y.Z., D.D.M.), Baylor College of Medicine, Houston, Texas 77030; Department of Biochemistry and Cell Biology (J.M.L.), School of Medicine, Kyungpook National University, Daegu, Republic of Korea 41944; Integrative Molecular and Biomedical Sciences Graduate Program (Y.Z., D.D.M.), Baylor College of Medicine, Houston, Texas 77030; and Department of Pediatrics (S.H.), Baylor College of Medicine and Texas Children's Hospital, Houston, Texas 77030
| | - Jae Man Lee
- Department of Molecular and Cellular Biology (K.H.K., J.M.L., Y.Z., D.D.M.), Baylor College of Medicine, Houston, Texas 77030; Department of Biochemistry and Cell Biology (J.M.L.), School of Medicine, Kyungpook National University, Daegu, Republic of Korea 41944; Integrative Molecular and Biomedical Sciences Graduate Program (Y.Z., D.D.M.), Baylor College of Medicine, Houston, Texas 77030; and Department of Pediatrics (S.H.), Baylor College of Medicine and Texas Children's Hospital, Houston, Texas 77030
| | - Ying Zhou
- Department of Molecular and Cellular Biology (K.H.K., J.M.L., Y.Z., D.D.M.), Baylor College of Medicine, Houston, Texas 77030; Department of Biochemistry and Cell Biology (J.M.L.), School of Medicine, Kyungpook National University, Daegu, Republic of Korea 41944; Integrative Molecular and Biomedical Sciences Graduate Program (Y.Z., D.D.M.), Baylor College of Medicine, Houston, Texas 77030; and Department of Pediatrics (S.H.), Baylor College of Medicine and Texas Children's Hospital, Houston, Texas 77030
| | - Sanjiv Harpavat
- Department of Molecular and Cellular Biology (K.H.K., J.M.L., Y.Z., D.D.M.), Baylor College of Medicine, Houston, Texas 77030; Department of Biochemistry and Cell Biology (J.M.L.), School of Medicine, Kyungpook National University, Daegu, Republic of Korea 41944; Integrative Molecular and Biomedical Sciences Graduate Program (Y.Z., D.D.M.), Baylor College of Medicine, Houston, Texas 77030; and Department of Pediatrics (S.H.), Baylor College of Medicine and Texas Children's Hospital, Houston, Texas 77030
| | - David D Moore
- Department of Molecular and Cellular Biology (K.H.K., J.M.L., Y.Z., D.D.M.), Baylor College of Medicine, Houston, Texas 77030; Department of Biochemistry and Cell Biology (J.M.L.), School of Medicine, Kyungpook National University, Daegu, Republic of Korea 41944; Integrative Molecular and Biomedical Sciences Graduate Program (Y.Z., D.D.M.), Baylor College of Medicine, Houston, Texas 77030; and Department of Pediatrics (S.H.), Baylor College of Medicine and Texas Children's Hospital, Houston, Texas 77030
| |
Collapse
|
129
|
Ebrahimi H, Naderian M, Sohrabpour AA. New Concepts on Pathogenesis and Diagnosis of Liver Fibrosis; A Review Article. Middle East J Dig Dis 2016; 8:166-178. [PMID: 27698966 PMCID: PMC5045669 DOI: 10.15171/mejdd.2016.29] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Liver fibrosis is a potentially reversible response to hepatic insults, triggered by different chronic diseases most importantly viral hepatitis, alcoholic, and nonalcoholic fatty liver disease. In the course of the chronic liver disease, hepatic fibrogenesis may develop, which is attributed to various types of cells, molecules, and pathways. Activated hepatic stellate cell (HSC), the primary source of extracellular matrix (ECM), is fundamental in pathophysiology of fibrogenesis, and thus is the most attractable target for reversing liver fibrosis. Although, liver biopsy has long been considered as the gold standard for diagnosis and staging of hepatic fibrosis, assessing progression and regression by biopsy is hampered by its limitations. We provide recent views on noninvasive approaches including serum biomarkers and radiologic techniques.
Collapse
Affiliation(s)
- Hedyeh Ebrahimi
- Liver and Pancreaticobiliary Diseases Research Center, Digestive Disease Research Institute, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran. Non-Communicable Diseases Research Center, Endocrinology and Metabolism Population Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammadreza Naderian
- Liver and Pancreaticobiliary Diseases Research Center, Digestive Disease Research Institute, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran. Non-Communicable Diseases Research Center, Endocrinology and Metabolism Population Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Amir Ali Sohrabpour
- Assistant Professor, Liver and Pancreaticobiliary Diseases Research Center, Digestive Disease Research Institute, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
130
|
Kocabayoglu P, Zhang DY, Kojima K, Hoshida Y, Friedman SL. Induction and contribution of beta platelet-derived growth factor signalling by hepatic stellate cells to liver regeneration after partial hepatectomy in mice. Liver Int 2016; 36:874-82. [PMID: 26256287 PMCID: PMC4749477 DOI: 10.1111/liv.12933] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Accepted: 08/04/2015] [Indexed: 12/19/2022]
Abstract
BACKGROUND & AIMS Hepatic stellate cells (HSCs) activate during injury to orchestrate the liver's inflammatory and fibrogenic responses. A critical feature of HSC activation is the rapid induction of beta platelet-derived growth factor (β-PDGFR), which drives cellular fibrogenesis and proliferation; in contrast, normal liver has minimal β-PDGFR expression. While the role of β-PDGFR is well established in liver injury, its expression and contribution during liver regeneration are unknown. The aim of this study was to determine whether β-PDGFR is induced during liver regeneration following partial hepatectomy (pHx), and to define its contribution to the regenerative response. METHODS Control mice or animals with HSC-specific β-PDGFR-depletion underwent two-thirds pHx followed by assessment of hepatocyte proliferation and expression of β-PDGFR. RNA-sequencing from whole liver tissue of both groups after pHx was used to uncover pathways regulated by β-PDGFR signalling in HSCs. RESULTS Beta platelet-derived growth factor expression on HSCs was up-regulated within 24 h following pHx in control mice, whereas absence of β-PDGFR blunted the expansion of HSCs. Mice lacking β-PDGFR displayed prolonged increases of transaminase levels within 72 h following pHx. Hepatocyte proliferation was impaired within the first 24 h based on Ki-67 and PCNA expression in β-PDGFR-deficient mice. This was associated with dysregulated growth in the β-PDGFR-deficient mice based on RNAseq with pathway analysis, and real-time quantitative PCR, which demonstrated reduced expression of Hgf, Igfbp1, Mapk and Il-6. CONCLUSIONS Beta platelet-derived growth factor is induced in HSCs following surgical pHx and its deletion in HSCs leads to prolonged liver injury. However, there is no significant difference in liver regeneration.
Collapse
Affiliation(s)
- Peri Kocabayoglu
- Division of Liver Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA,Department of General, Visceral and Transplant Surgery, University Hospital Essen, Germany
| | - David Y. Zhang
- Division of Liver Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Kensuke Kojima
- Division of Liver Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Yujin Hoshida
- Division of Liver Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA,Liver Cancer Program, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Scott L. Friedman
- Division of Liver Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA,Liver Cancer Program, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| |
Collapse
|
131
|
Wang X, Wu X, Zhang A, Wang S, Hu C, Chen W, Shen Y, Tan R, Sun Y, Xu Q. Targeting the PDGF-B/PDGFR-β Interface with Destruxin A5 to Selectively Block PDGF-BB/PDGFR-ββ Signaling and Attenuate Liver Fibrosis. EBioMedicine 2016; 7:146-56. [PMID: 27322468 PMCID: PMC4909612 DOI: 10.1016/j.ebiom.2016.03.042] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2015] [Revised: 03/12/2016] [Accepted: 03/29/2016] [Indexed: 12/22/2022] Open
Abstract
PDGF-BB/PDGFR-ββ signaling plays very crucial roles in the process of many diseases such as liver fibrosis. However, drug candidates with selective affinities for PDGF-B/PDGFR-β remain deficient. Here, we identified a natural cyclopeptide termed destruxin A5 that effectively inhibits PDGF-BB-induced PDGFR-β signaling. Interestingly and importantly, the inhibitory mechanism is distinct from the mechanism of tyrosine kinase inhibitors because destruxin A5 does not have the ability to bind to the ATP-binding pocket of PDGFR-β. Using Biacore T200 technology, thermal shift technology, microscale thermophoresis technology and computational analysis, we confirmed that destruxin A5 selectively targets the PDGF-B/PDGFR-β interaction interface to block this signaling. Additionally, the inhibitory effect of destruxin A5 on PDGF-BB/PDGFR-ββ signaling was verified using in vitro, ex vivo and in vivo models, in which the extent of liver fibrosis was effectively alleviated by destruxin A5. In summary, destruxin A5 may represent an efficacious and more selective inhibitor of PDGF-BB/PDGFR-ββ signaling.
Collapse
Affiliation(s)
- Xingqi Wang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, 22 Hankou Road, Nanjing 210093, China; Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University, Xuzhou 221116, China
| | - Xuefeng Wu
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, 22 Hankou Road, Nanjing 210093, China
| | - Aihua Zhang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, 22 Hankou Road, Nanjing 210093, China
| | - Shiyu Wang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, 22 Hankou Road, Nanjing 210093, China
| | - Chunhui Hu
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, 22 Hankou Road, Nanjing 210093, China
| | - Wei Chen
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, 22 Hankou Road, Nanjing 210093, China
| | - Yan Shen
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, 22 Hankou Road, Nanjing 210093, China
| | - Renxiang Tan
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, 22 Hankou Road, Nanjing 210093, China.
| | - Yang Sun
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, 22 Hankou Road, Nanjing 210093, China.
| | - Qiang Xu
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, 22 Hankou Road, Nanjing 210093, China.
| |
Collapse
|
132
|
Chen Q, Chen L, Wu X, Zhang F, Jin H, Lu C, Shao J, Kong D, Wu L, Zheng S. Dihydroartemisinin prevents liver fibrosis in bile duct ligated rats by inducing hepatic stellate cell apoptosis through modulating the PI3K/Akt pathway. IUBMB Life 2016; 68:220-31. [DOI: 10.1002/iub.1478] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Accepted: 01/05/2016] [Indexed: 12/17/2022]
Affiliation(s)
- Qin Chen
- Department of Pharmacology, School of Pharmacy; Nanjing University of Chinese Medicine; Nanjing Jiangsu Province China
| | - Lianyun Chen
- Department of Pharmacology, School of Pharmacy; Nanjing University of Chinese Medicine; Nanjing Jiangsu Province China
| | - Xiafei Wu
- Department of Pharmacology, School of Pharmacy; Nanjing University of Chinese Medicine; Nanjing Jiangsu Province China
| | - Feng Zhang
- Department of Pharmacology, School of Pharmacy; Nanjing University of Chinese Medicine; Nanjing Jiangsu Province China
- National First-Class Key Discipline for Traditional Chinese Medicine of Nanjing University of Chinese Medicine; Nanjing China
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Material Medical; Nanjing University of Chinese Medicine; Nanjing China
| | - Huanhuan Jin
- Department of Pharmacology, School of Pharmacy; Nanjing University of Chinese Medicine; Nanjing Jiangsu Province China
| | - Chunfeng Lu
- Department of Pharmacology, School of Pharmacy; Nanjing University of Chinese Medicine; Nanjing Jiangsu Province China
| | - Jiangjuan Shao
- Department of Pharmacology, School of Pharmacy; Nanjing University of Chinese Medicine; Nanjing Jiangsu Province China
| | - Desong Kong
- Department of Pharmacology, School of Pharmacy; Nanjing University of Chinese Medicine; Nanjing Jiangsu Province China
- Department of Science; Technology and Education, the Third Affiliated Hospital of Nanjing University of Chinese Medicine; Nanjing China
| | - Li Wu
- Department of Pharmacology, School of Pharmacy; Nanjing University of Chinese Medicine; Nanjing Jiangsu Province China
| | - Shizhong Zheng
- Department of Pharmacology, School of Pharmacy; Nanjing University of Chinese Medicine; Nanjing Jiangsu Province China
- National First-Class Key Discipline for Traditional Chinese Medicine of Nanjing University of Chinese Medicine; Nanjing China
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Material Medical; Nanjing University of Chinese Medicine; Nanjing China
| |
Collapse
|
133
|
Fagone P, Mangano K, Pesce A, Portale TR, Puleo S, Nicoletti F. Emerging therapeutic targets for the treatment of hepatic fibrosis. Drug Discov Today 2016; 21:369-75. [PMID: 26523773 DOI: 10.1016/j.drudis.2015.10.015] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2015] [Revised: 10/08/2015] [Accepted: 10/21/2015] [Indexed: 02/07/2023]
Abstract
Fibrosis represents a response to chronic injury, aimed at maintaining organ integrity. Hepatic fibrosis is mainly related to chronic viral hepatitis B or C (HBV or HCV), alcoholic and nonalcoholic steatohepatitis (NASH), and biliary diseases. A deep understanding of the cellular and molecular mechanisms underlying liver fibrosis has enabled the development of 'pathogenetic tailored' therapeutic interventions. However, effective drugs to prevent or revert hepatic fibrosis are still lacking. In this review, we discuss the cellular populations and the molecular pathways involved in liver fibrogenesis as well as the novel approaches currently being tested in clinical trials.
Collapse
Affiliation(s)
- Paolo Fagone
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Katia Mangano
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Antonio Pesce
- Department of Medical and Surgical Sciences and Advanced Technologies, G.F. Ingrassia, University of Catania, Catania, Italy
| | - Teresa Rosanna Portale
- Department of Medical and Surgical Sciences and Advanced Technologies, G.F. Ingrassia, University of Catania, Catania, Italy
| | - Stefano Puleo
- Department of Medical and Surgical Sciences and Advanced Technologies, G.F. Ingrassia, University of Catania, Catania, Italy
| | - Ferdinando Nicoletti
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy.
| |
Collapse
|
134
|
Rauff B, Douglas MW. Role of fibrogenic and inflammatory cytokines in HCV-induced fibrosis. Future Virol 2015. [DOI: 10.2217/fvl.15.102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
HCV is one of the main causative agents of liver fibrosis and hepatocellular carcinoma. Liver inflammation resulting from HCV infection triggers fibrosis. In HCV-related fibrosis, differentiated hepatic stellate cells (HSCs) known as myofibroblasts participate in the fibrogenic and inflammatory response. TGF-β1 and CTGF, released from these HSCs, have been implicated as master cytokines mediating HCV induced hepatic fibrosis. PDGF is another potent mitogen, which facilitates the progression of liver fibrosis by enhancing the proliferation and migration of HSCs. In addition to these major cytokines, the release of TNF-α, IL-6, IL-1b and IL-10 by immune cells also promotes the effect of HCV induced fibrosis. Targeting these cytokines may offer the potential for treatments to prevent or cure fibrosis.
Collapse
Affiliation(s)
- Bisma Rauff
- Storr Liver Centre, Westmead Millennium Institute, University of Sydney at Westmead Hospital, NSW, Australia
| | - Mark W Douglas
- Storr Liver Centre, Westmead Millennium Institute, University of Sydney at Westmead Hospital, NSW, Australia
- Centre for Infectious Diseases & Microbiology, Marie Bashir Institute for Infectious Diseases & Biosecurity, University of Sydney at Westmead Hospital, NSW, Australia
| |
Collapse
|
135
|
Wang Q, Wen R, Lin Q, Wang N, Lu P, Zhu X. Wogonoside Shows Antifibrotic Effects in an Experimental Regression Model of Hepatic Fibrosis. Dig Dis Sci 2015; 60:3329-39. [PMID: 26130019 DOI: 10.1007/s10620-015-3751-4] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2015] [Accepted: 06/06/2015] [Indexed: 01/18/2023]
Abstract
BACKGROUD Wogonoside (WO), a flavonoid extracted from Huangqin, plays multiple physiological roles. However, it has remained elusive how WO regulates hepatic fibrogenesis until now. AIM The purpose of the study was to investigate the potential protective effects of WO against liver fibrosis induced by carbon tetrachloride (CCl4). METHODS In this study, male rats were randomly allocated into four groups: a control group, the CCl4 group, the CCl4 and WO (4 mg/kg) group, and CCl4 and WO (8 mg/kg) group. Hepatic fibrosis was induced by subcutaneous injection of CCl4 twice a week for a continuous 6-week period. Then the rats were intragastrically administrated with WO daily for 4 weeks before being killed. RESULTS As expected, histopathological assessment, Masson trichrome staining, and Sirius red staining demonstrated that WO drastically ameliorated the hepatic fibrosis caused by CCl4. WO significantly attenuated the CCl4-induced upregulations of liver indices including alanine aminotransferase, aspartate aminotransferase, tumor necrosis factor-α, interleukin-1β, IL-6, hexadecenoic acid and laminin in serum, as well as hydroxyproline, malondialdehyde and phosphatidylinositol 3-kinase (PI3K)/protein Kinase B(Akt)/mechanistic target of rapamycin (mTOR)/nuclear factor-kappa B signalings in liver. Meanwhile, WO also effectively recovered the depletions of superoxide dismutase, glutathione and IL-10. Furthermore, we evaluated the effects of WO on the alpha smooth muscle actin, type I collagen expressions, and PI3K/Akt/ mTOR/ribosomal protein S6 kinase 70 kDa (p70S6K) signaling in transforming growth factor (TGF-β) stimulated hepatic stellate cell-T6 cells. CONCLUSIONS These results suggested that WO had significant protective effects against liver fibrosis induced by CCl4.
Collapse
Affiliation(s)
- Qichao Wang
- Translational Center for Stem Cell Research, Tongji Hospital, Department of Regenerative Medicine, Tongji University School of Medicine, Shanghai, 200065, China
| | - Rui Wen
- Department of Resources Science of Traditional Chinese Medicines, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, China
| | - Qinghua Lin
- Department of Natural Medicinal Chemistry, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, China
| | - Na Wang
- Translational Center for Stem Cell Research, Tongji Hospital, Department of Regenerative Medicine, Tongji University School of Medicine, Shanghai, 200065, China
| | - Ping Lu
- Translational Center for Stem Cell Research, Tongji Hospital, Department of Regenerative Medicine, Tongji University School of Medicine, Shanghai, 200065, China
| | - Xianmin Zhu
- Translational Center for Stem Cell Research, Tongji Hospital, Department of Regenerative Medicine, Tongji University School of Medicine, Shanghai, 200065, China.
| |
Collapse
|
136
|
Borkham-Kamphorst E, Weiskirchen R. The PDGF system and its antagonists in liver fibrosis. Cytokine Growth Factor Rev 2015; 28:53-61. [PMID: 26547628 DOI: 10.1016/j.cytogfr.2015.10.002] [Citation(s) in RCA: 114] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2015] [Accepted: 10/19/2015] [Indexed: 01/18/2023]
Abstract
Platelet derived growth factor (PDGF) signaling plays an important role in activated hepatic stellate cells and portal fibroblast proliferation, chemotaxis, migration and cell survival. PDGF receptors and ligands are upregulated in experimental liver fibrotic models as well as in human liver fibrotic diseases. Blocking of PDGF signaling ameliorates experimental liver fibrogenesis. The plurality of molecular and cellular activities of PDGF and its involvement in initiation, progression and resolution of hepatic fibrogenesis offers an infinite number of therapeutic possibilities. These include the application of therapeutic antibodies (e.g. AbyD3263, MOR8457) which specifically sequester individual PDGF isoforms or the inhibition of PDGF isoforms by synthetic aptamers. In particular, the isolation of innovative slow off-rate modified aptamers (e.g., SOMAmer SL1 and SL5) that carry functional groups absent in natural nucleic acids by the Systematic Evolution of Ligands by EXponential (SELEX) enrichment technique offers the possibility to design high affinity aptamers that target PDGF isoforms for clinical purposes. Dominant-negative soluble PDGF receptors are also effective in attenuation of hepatic stellate cell proliferation and hepatic fibrogenesis. Moreover, some multikinase inhibitors targeting PDGF signaling have been intensively tested during the last decade and are on the way into advanced preclinical studies and clinical trials. This narrative review aims to gauge the recent progression of research into PDGF systems and liver fibrosis.
Collapse
Affiliation(s)
- Erawan Borkham-Kamphorst
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry, RWTH University Hospital Aachen, Aachen, Germany.
| | - Ralf Weiskirchen
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry, RWTH University Hospital Aachen, Aachen, Germany.
| |
Collapse
|