1
|
Kim JY, Kang YC, Kim MJ, Kim SU, Kang HR, Yeo JS, Kim Y, Yu SH, Song B, Hwang JW, Lee YS, Byun JW, Yoo DH, Kim HS, Han K, Kim CH, Lee EY. Mitochondrial transplantation as a novel therapeutic approach in idiopathic inflammatory myopathy. Ann Rheum Dis 2025; 84:609-619. [PMID: 39893098 DOI: 10.1016/j.ard.2024.11.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 11/04/2024] [Accepted: 11/21/2024] [Indexed: 02/04/2025]
Abstract
OBJECTIVES This study aimed to investigate the efficacy of mitochondrial transplantation as a therapeutic intervention for idiopathic inflammatory myopathy (IIM). This study used a comprehensive approach, incorporating both in vitro and in vivo IIM models, and conducted a first-in-human clinical trial to assess the effectiveness and safety of mitochondria isolated from human umbilical cord mesenchymal stem cells (PN-101). METHODS Mitochondria isolated from umbilical cord mesenchymal stem cells were designated as PN-101. The efficacy of PN-101 was assessed using myoblasts derived from patients with IIM and C2C12 mouse perforin/granzyme B-treated myoblasts as an in vitro IIM model. PN-101's effect on IIM was examined using C protein-induced myositis (CIM) mice as an in vivo model. The efficacy and safety of PN-101 were evaluated in a phase 1/2a clinical trial involving 9 adult patients with refractory polymyositis or dermatomyositis. RESULTS The myoblasts derived from patients with IIM exhibited defects in mitochondrial function and myogenesis. PN-101 transplantation enhances muscle differentiation and mitochondrial function in IIM myoblasts. PN-101 also enhanced intracellular adenosine triphosphate content, cell viability, and myogenesis in C2C12 perforin/granzyme B-treated myoblasts. In an in vivo model, PN-101 reduced myositis severity by exhibiting anti-inflammatory effects and restoring the CIM-induced metabolic shift. In a phase 1/2a prospective clinical trial involving adult patients with refractory IIM, PN-101 demonstrated no severe adverse drug reactions and showed at least minimal improvement in the International Myositis Assessment and Clinical Studies Group (IMACS)-Total Improvement Scores (TISs) compared with baseline. CONCLUSIONS PN-101 transplantation could serve as a novel treatment for IIM by enhancing mitochondrial repair and reducing inflammation in muscle tissues.
Collapse
Affiliation(s)
- Jeong Yeon Kim
- Division of Rheumatology, Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
| | | | - Min Jung Kim
- Division of Rheumatology, Department of Internal Medicine, Seoul Metropolitan Government-Seoul National University Hospital Boramae Medical Center, Seoul, Korea
| | - Seon Uk Kim
- Division of Rheumatology, Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Hae Rim Kang
- Division of Rheumatology, Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
| | | | - Yujin Kim
- Paean Biotechnology Inc, Seoul, Korea
| | | | | | | | - Yun-Sang Lee
- Department of Nuclear Medicine, Seoul National University Hospital, Seoul, Korea
| | - Jung Woo Byun
- Department of Nuclear Medicine, Seoul National University Hospital, Seoul, Korea; Department of Biomedical Sciences, Seoul National University, Seoul, Korea
| | - Dae Hyun Yoo
- Department of Rheumatology, College of Medicine, Hanyang University Hospital for Rheumatic Diseases, Hanyang University, Seoul, Korea
| | - Hyun Sook Kim
- Division of Rheumatology, Department of Internal Medicine, Soonchunhyang University Seoul Hospital, Seoul, Korea
| | | | | | - Eun Young Lee
- Division of Rheumatology, Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea.
| |
Collapse
|
2
|
Hijazi N, Shi Z, Rockey DC. Paxillin regulates liver fibrosis via actin polymerization and ERK activation in hepatic stellate cells. J Cell Sci 2023; 136:jcs261122. [PMID: 37667902 PMCID: PMC10560551 DOI: 10.1242/jcs.261122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 08/15/2023] [Indexed: 09/06/2023] Open
Abstract
Liver injury leads to fibrosis and cirrhosis. The primary mechanism underlying the fibrogenic response is the activation of hepatic stellate cells (HSCs), which are 'quiescent' in normal liver but become 'activated' after injury by transdifferentiating into extracellular matrix (ECM)-secreting myofibroblasts. Given that integrins are important in HSC activation and fibrogenesis, we hypothesized that paxillin, a key downstream effector in integrin signaling, might be critical in the fibrosis pathway. Using a cell-culture-based model of HSC activation and in vivo models of liver injury, we found that paxillin is upregulated in activated HSCs and fibrotic livers. Overexpression of paxillin (both in vitro and in vivo) led to increased ECM protein expression, and depletion of paxillin in a novel conditional mouse injury model reduced fibrosis. The mechanism by which paxillin mediated this effect appeared to be through the actin cytoskeleton, which signals to the ERK pathway and induces ECM protein production. These data highlight a novel role for paxillin in HSC biology and fibrosis.
Collapse
Affiliation(s)
- Nour Hijazi
- Digestive Disease Research Center Core, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Zengdun Shi
- Digestive Disease Research Center Core, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Don C. Rockey
- Digestive Disease Research Center Core, Medical University of South Carolina, Charleston, SC 29425, USA
| |
Collapse
|
3
|
Hijazi N, Shi Z, Rockey DC. Characterization of focal adhesion proteins in rodent hepatic stellate cells. Histochem Cell Biol 2022; 158:325-334. [PMID: 35960334 PMCID: PMC10824234 DOI: 10.1007/s00418-022-02123-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/01/2022] [Indexed: 11/04/2022]
Abstract
Ongoing liver injury leads to fibrosis and ultimately cirrhosis, a leading cause of death worldwide. The primary mechanism underlying the fibrogenic response is the activation of cells known as hepatic stellate cells (HSCs) which are "quiescent" in the normal liver but become "activated" after injury by transdifferentiating into extracellular matrix-secreting myofibroblasts. Since integrins (extracellular matrix binding receptors) are important mediators of HSC activation and fibrogenesis, we hypothesized that focal adhesion (FA) proteins, which link integrins to the intracellular protein machinery, may be important in the activation process. Therefore, using both an in vitro model of activation in primary rat HSCs and an in vivo model of liver injury, we examined three FA proteins: vinculin, FAK, and talin. All three proteins were significantly upregulated during HSC activation at both the messenger RNA (mRNA) and protein levels. Confocal microscopy demonstrated that the proteins had a widespread expression throughout HSCs with prominent localization at the end of actin filaments. Finally, we stimulated HSCs with the profibrotic ligands endothelin-1 (ET-1) and transforming growth factor beta (TGF-β) and observed an increase in the size of vinculin-containing FAs and the cell area occupied by them. The data indicate that HSCs possess a broad array of FA proteins, and given their upregulation during activation, this raises the possibility that they play a role in the fibrogenic response to injury.
Collapse
Affiliation(s)
- Nour Hijazi
- Digestive Disease Research Core Center, Medical University of South Carolina, 96 Jonathan Lucas Street, Clinical Sciences Building, Suite 912, Charleston, SC 29425, USA
| | - Zengdun Shi
- Digestive Disease Research Core Center, Medical University of South Carolina, 96 Jonathan Lucas Street, Clinical Sciences Building, Suite 912, Charleston, SC 29425, USA
| | - Don C. Rockey
- Digestive Disease Research Core Center, Medical University of South Carolina, 96 Jonathan Lucas Street, Clinical Sciences Building, Suite 912, Charleston, SC 29425, USA
| |
Collapse
|
4
|
Chae SA, Pyo MC, Yoo HJ, Lee KW. Ochratoxin a induces hepatic fibrosis through TGF-β receptor I/Smad2/3 signaling pathway. ENVIRONMENTAL TOXICOLOGY 2022; 37:2084-2095. [PMID: 35543154 DOI: 10.1002/tox.23552] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Revised: 04/20/2022] [Accepted: 04/23/2022] [Indexed: 06/14/2023]
Abstract
Ochratoxin A (OTA) is a mycotoxin generated by Penicillium and Aspergillus species. It is often found in cereals. We hypothesized that OTA exposure induces epithelial-mesenchymal transition (EMT), leading to liver fibrosis. In this research, we explored whether the TGF-β receptor I (TGF-β RI)/Smad2/3 signaling pathway is related to EMT-induced hepatic fibrosis. In vitro and in vivo experiments, mRNA and protein expression of liver fibrosis-related markers such as fibronectin, α-smooth muscle actin (α-SMA) and E-cadherin were assessed. The levels of alkaline phosphatase, alanine transaminase, aspartate aminotransferase, and total bilirubin, which are used to assess damage, increased. We also confirmed the increase in mRNA and protein expression of TGF-β RI, Smad2, and Smad3. The expression of liver fibrosis-related markers was decreased by siRNA-mediated silencing of Smad2/3, as well as TGF-RI suppression. Liver cells exposed to OTA showed enhanced TGF-β RI expression on the cell membrane. These results demonstrated that OTA induces hepatic fibrosis through TGF-β RI and Smad2/3 pathways in vitro and in vivo.
Collapse
Affiliation(s)
- Seung A Chae
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, Republic of Korea
| | - Min Cheol Pyo
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, Republic of Korea
| | - Hee Joon Yoo
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, Republic of Korea
| | - Kwang-Won Lee
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, Republic of Korea
- Department of Food Bioscience and Technology, College of Life Sciences and Biotechnology, Korea University, Seoul, Republic of Korea
| |
Collapse
|
5
|
Liu X, Taylor SA, Gromer KD, Zhang D, Hubchak SC, LeCuyer BE, Iwawaki T, Shi Z, Rockey DC, Green RM. Mechanisms of liver injury in high fat sugar diet fed mice that lack hepatocyte X-box binding protein 1. PLoS One 2022; 17:e0261789. [PMID: 35030194 PMCID: PMC8759640 DOI: 10.1371/journal.pone.0261789] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Accepted: 12/09/2021] [Indexed: 11/26/2022] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is one of the most common causes of liver diseases in the United States and can progress to cirrhosis, end-stage liver disease and need for liver transplantation. There are limited therapies for NAFLD, in part, due to incomplete understanding of the disease pathogenesis, which involves different cell populations in the liver. Endoplasmic reticulum stress and its adaptative unfolded protein response (UPR) signaling pathway have been implicated in the progression from simple hepatic steatosis to nonalcoholic steatohepatitis (NASH). We have previously shown that mice lacking the UPR protein X-box binding protein 1 (XBP1) in the liver demonstrated enhanced liver injury and fibrosis in a high fat sugar (HFS) dietary model of NAFLD. In this study, to better understand the role of liver XBP1 in the pathobiology of NAFLD, we fed hepatocyte XBP1 deficient mice a HFS diet or chow and investigated UPR and other cell signaling pathways in hepatocytes, hepatic stellate cells and immune cells. We demonstrate that loss of XBP1 in hepatocytes increased inflammatory pathway expression and altered expression of the UPR signaling in hepatocytes and was associated with enhanced hepatic stellate cell activation after HFS feeding. We believe that a better understanding of liver cell-specific signaling in the pathogenesis of NASH may allow us to identify new therapeutic targets.
Collapse
Affiliation(s)
- Xiaoying Liu
- Division of Gastroenterology and Hepatology, Department of Medicine, Northwestern University, Chicago, Illinois, United States of America
| | - Sarah A. Taylor
- Division of Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, Ann & Robert H. Lurie Children’s Hospital of Chicago, Chicago, Illinois, United States of America
| | - Kyle D. Gromer
- Division of Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, Ann & Robert H. Lurie Children’s Hospital of Chicago, Chicago, Illinois, United States of America
| | - Danny Zhang
- Division of Gastroenterology and Hepatology, Department of Medicine, Northwestern University, Chicago, Illinois, United States of America
| | - Susan C. Hubchak
- Division of Gastroenterology and Hepatology, Department of Medicine, Northwestern University, Chicago, Illinois, United States of America
| | - Brian E. LeCuyer
- Division of Gastroenterology and Hepatology, Department of Medicine, Northwestern University, Chicago, Illinois, United States of America
| | - Takao Iwawaki
- Division of Cell Medicine, Department of Life Science, Medical Research Institute, Kanazawa Medical University, Kahoku, Ishikawa, Japan
| | - Zengdun Shi
- Digestive Disease Research Center, Medical University of South Carolina, Charleston, South Carolina, United States of America
| | - Don C. Rockey
- Digestive Disease Research Center, Medical University of South Carolina, Charleston, South Carolina, United States of America
| | - Richard M. Green
- Division of Gastroenterology and Hepatology, Department of Medicine, Northwestern University, Chicago, Illinois, United States of America
| |
Collapse
|
6
|
Medley SC, Rathnakar BH, Georgescu C, Wren JD, Olson LE. Fibroblast-specific Stat1 deletion enhances the myofibroblast phenotype during tissue repair. Wound Repair Regen 2020; 28:448-459. [PMID: 32175700 DOI: 10.1111/wrr.12807] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Revised: 02/12/2020] [Accepted: 03/10/2020] [Indexed: 12/17/2022]
Abstract
Signal transducer and activator of transcription 1 (Stat1) is a ubiquitously expressed latent transcription factor that is activated by many cytokines and growth factors. Global Stat1 knockout mice are prone to chemical-induced lung and liver fibrosis, suggesting roles for Stat1 in tissue repair. However, the importance of Stat1 in fibroblast-mediated and vascular smooth muscle cell (VSMC)-mediated injury response has not been directly evaluated in vivo. Here, we focused on two models of tissue repair in conditional Stat1 knockout mice: excisional skin wounding in mice with Stat1 deletion in dermal fibroblasts, and carotid artery ligation in mice with global Stat1 deletion or deletion specific to VSMCs. In the skin model, dermal wounds closed at a similar rate in mice with fibroblast Stat1 deletion and controls, but collagen and α-smooth muscle actin (αSMA) expression were increased in the mutant granulation tissue. Cultured Stat1 -/- and Stat1 +/- dermal fibroblasts exhibited similar αSMA+ stress fiber assembly, collagen gel contraction, proliferation, migration, and growth factor-induced gene expression. In the artery ligation model, there was a significant increase in fibroblast-driven perivascular fibrosis when Stat1 was deleted globally. However, VSMC-driven remodeling and neointima formation were unchanged when Stat1 was deleted specifically in VSMCs. These results suggest an in vivo role for Stat1 as a suppressor of fibroblast mediated, but not VSMC mediated, injury responses, and a suppressor of the myofibroblast phenotype.
Collapse
Affiliation(s)
- Shayna C Medley
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, USA.,Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| | - Bharath H Rathnakar
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, USA
| | - Constantin Georgescu
- Genes and Human Disease Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, USA
| | - Jonathan D Wren
- Genes and Human Disease Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, USA
| | - Lorin E Olson
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, USA.,Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| |
Collapse
|
7
|
Shi Z, Ren M, Rockey DC. Myocardin and myocardin-related transcription factor-A synergistically mediate actin cytoskeletal-dependent inhibition of liver fibrogenesis. Am J Physiol Gastrointest Liver Physiol 2020; 318:G504-G517. [PMID: 31928221 PMCID: PMC7099496 DOI: 10.1152/ajpgi.00302.2019] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Activation of hepatic stellate cells (HSCs), characterized by development of a robust actin cytoskeleton and expression of abundant extracellular matrix (ECM) proteins, such as type 1 collagen (COL.1), is a central cellular and molecular event in liver fibrosis. It has been demonstrated that HSCs express both myocardin and myocardin-related transcription factor-A (MRTF-A). However, the biological effects of myocardin and MRTF-A on HSC activation and liver fibrosis, as well as the molecular mechanism under the process, remain unclear. Here, we report that myocardin and MRTF-A's expression and nuclear accumulation are prominently increased during the HSC activation process, accompanied by robust activation of actin cytoskeleton dynamics. Targeting myocardin and MRTF-A binding and function with a novel small molecule, CCG-203971, led to dose-dependent inhibition of HSC actin cytoskeleton dynamics and abrogated multiple functional features of HSC activation (i.e., HSC contraction, migration and proliferation) and decreased COL.1 expression in vitro and liver fibrosis in vivo. Mechanistically, blocking the myocardin and MRTF-A nuclear translocation pathway with CCG-203971 directly inhibited myocardin/MRTF-A-mediated serum response factor (SRF), and Smad2/3 activation in the COL.1α2 promoter and indirectly abrogated actin cytoskeleton-dependent regulation of Smad2/3 and Erk1/2 phosphorylation and their nuclear accumulation. Finally, there was no effect of CCG-203971 on markers of inflammation, suggesting a direct effect of the compound on HSCs and liver fibrosis. These data reveal that myocardin and MRTF-A are two important cotranscriptional factors in HSCs and represent entirely novel therapeutic pathways that might be targeted to treat liver fibrosis.NEW & NOTEWORTHY Myocardin and myocardin-related transcription factor-A (MRTF-A) are upregulated in activated hepatic stellate cells (HSCs) in vitro and in vivo, closely associated with robustly increased actin cytoskeleton remodeling. Targeting myocardin and MRTF-A by CCG-203971 leads to actin cytoskeleton-dependent inhibition of HSC activation, reduced cell contractility, impeded cell migration and proliferation, and decreased COL.1 expression in vitro and in vivo. Dual expression of myocardin and MRTF-A in HSCs may represent novel therapeutic targets in liver fibrosis.
Collapse
Affiliation(s)
- Zengdun Shi
- Department of Internal Medicine, Medical University of South Carolina, Charleston, South Carolina
| | - Mudan Ren
- Department of Internal Medicine, Medical University of South Carolina, Charleston, South Carolina
| | - Don C. Rockey
- Department of Internal Medicine, Medical University of South Carolina, Charleston, South Carolina
| |
Collapse
|
8
|
Rockey DC, Du Q, Weymouth ND, Shi Z. Smooth Muscle α-Actin Deficiency Leads to Decreased Liver Fibrosis via Impaired Cytoskeletal Signaling in Hepatic Stellate Cells. THE AMERICAN JOURNAL OF PATHOLOGY 2019; 189:2209-2220. [PMID: 31476284 DOI: 10.1016/j.ajpath.2019.07.019] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/17/2019] [Revised: 07/03/2019] [Accepted: 07/29/2019] [Indexed: 01/18/2023]
Abstract
In the liver, smooth muscle α-actin (SM α-actin) is up-regulated in hepatic stellate cells (HSCs) as they transition to myofibroblasts during liver injury and the wound healing response. Whether SM α-actin has specific functional effects on cellular effectors of fibrosis such as HSC is controversial. Here, the relationship between SM α-actin and type 1 collagen expression (COL1A1), a major extracellular matrix protein important in liver fibrosis, is investigated with the results demonstrating that knockout of SM α-actin leads to reduced liver fibrosis and COL1 expression. The mechanism for the reduction in fibrogenesis in vivo is multifactorial, including not only a reduction in the number of HSCs, but also an HSC-specific reduction in COL1 expression in Acta2-deficient HSCs. Despite a compensatory increase in expression of cytoplasmic β-actin and γ-actin isoforms in Acta2-/- HSCs, defects were identified in each transforming growth factor beta/Smad2/3 and ET-1/Erk1/2 signaling in Acta2-/- HSCs. These data not only suggest a molecular link between the SM α-actin cytoskeleton and classic fibrogenic signaling cascades, but also emphasize the relationship between SM α-actin and fibrogenesis in hepatic myofibroblasts in vivo.
Collapse
Affiliation(s)
- Don C Rockey
- Department of Internal Medicine, Medical University of South Carolina, Charleston, South Carolina.
| | - Qinghong Du
- Department of Internal Medicine, Medical University of South Carolina, Charleston, South Carolina
| | | | - Zengdun Shi
- Department of Internal Medicine, Medical University of South Carolina, Charleston, South Carolina.
| |
Collapse
|
9
|
Haldar R, Shaashua L, Lavon H, Lyons YA, Zmora O, Sharon E, Birnbaum Y, Allweis T, Sood AK, Barshack I, Cole S, Ben-Eliyahu S. Perioperative inhibition of β-adrenergic and COX2 signaling in a clinical trial in breast cancer patients improves tumor Ki-67 expression, serum cytokine levels, and PBMCs transcriptome. Brain Behav Immun 2018; 73:294-309. [PMID: 29800703 DOI: 10.1016/j.bbi.2018.05.014] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Revised: 05/10/2018] [Accepted: 05/18/2018] [Indexed: 12/18/2022] Open
Abstract
Catecholamines and prostaglandins are secreted abundantly during the perioperative period in response to stress and surgery, and were shown by translational studies to promote tumor metastasis. Here, in a phase-II biomarker clinical trial in breast cancer patients (n = 38), we tested the combined perioperative use of the β-blocker, propranolol, and the COX2-inhibitor, etodolac, scheduled for 11 consecutive perioperative days, starting 5 days before surgery. Blood samples were taken before treatment (T1), on the mornings before and after surgery (T2&T3), and after treatment cessation (T4). Drugs were well tolerated. Results based on a-priori hypotheses indicated that already before surgery (T2), serum levels of pro-inflammatory IL-6, CRP, and IFNγ, and anti-inflammatory, cortisol and IL-10, increased. At T2 and/or T3, drug treatment reduced serum levels of the above pro-inflammatory cytokines and of TRAIL, as well as activity of multiple inflammation-related transcription factors (including NFκB, STAT3, ISRE), but not serum levels of cortisol, IL-10, IL-18, IL-8, VEGF and TNFα. In the excised tumor, treatment reduced the expression of the proliferation marker Ki-67, and positively affected its transcription factors SP1 and AhR. Exploratory analyses of transcriptome modulation in PBMCs revealed treatment-induced improvement at T2/T3 in several transcription factors that in primary tumors indicate poor prognosis (CUX1, THRa, EVI1, RORa, PBX1, and T3R), angiogenesis (YY1), EMT (GATA1 and deltaEF1/ZEB1), proliferation (GATA2), and glucocorticoids response (GRE), while increasing the activity of the oncogenes c-MYB and N-MYC. Overall, the drug treatment may benefit breast cancer patients through reducing systemic inflammation and pro-metastatic/pro-growth biomarkers in the excised tumor and PBMCs.
Collapse
Affiliation(s)
- Rita Haldar
- Sagol School of Neuroscience and School of Psychological Sciences, Tel Aviv University, Israel
| | - Lee Shaashua
- Sagol School of Neuroscience and School of Psychological Sciences, Tel Aviv University, Israel
| | - Hagar Lavon
- Sagol School of Neuroscience and School of Psychological Sciences, Tel Aviv University, Israel
| | - Yasmin A Lyons
- Department of Gynecologic Oncology and Reproductive Medicine, Division of Surgery, M.D. Anderson Cancer Center at University of Texas, Huston, TX, USA
| | - Oded Zmora
- Department of Surgery and Transplantation, Sheba Medical Center, Ramat Gan, Israel
| | - Eran Sharon
- Department of Surgery, Rabin Medical Center, Beilinson Hospital, Petach-Tikva, Israel
| | - Yehudit Birnbaum
- Department of Surgery, Rabin Medical Center, Beilinson Hospital, Petach-Tikva, Israel
| | - Tanir Allweis
- Department of Surgery, Kaplan Medical Center, Rehovot, Israel
| | - Anil K Sood
- Department of Gynecologic Oncology and Reproductive Medicine, Division of Surgery, M.D. Anderson Cancer Center at University of Texas, Huston, TX, USA
| | - Iris Barshack
- Department of Pathology, Sheba Medical Center, Ramat Gan, Israel
| | - Steve Cole
- Department of Medicine, Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Shamgar Ben-Eliyahu
- Sagol School of Neuroscience and School of Psychological Sciences, Tel Aviv University, Israel.
| |
Collapse
|
10
|
Zhou F, Wang A, Li D, Wang Y, Lin L. Pinocembrin from Penthorum chinense Pursh suppresses hepatic stellate cells activation through a unified SIRT3-TGF-β-Smad signaling pathway. Toxicol Appl Pharmacol 2018; 341:38-50. [DOI: 10.1016/j.taap.2018.01.009] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Revised: 01/03/2018] [Accepted: 01/13/2018] [Indexed: 01/18/2023]
|
11
|
Upregulation of the actin cytoskeleton via myocardin leads to increased expression of type 1 collagen. J Transl Med 2017; 97:1412-1426. [PMID: 29035375 PMCID: PMC6437559 DOI: 10.1038/labinvest.2017.96] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Revised: 06/19/2017] [Accepted: 07/05/2017] [Indexed: 12/28/2022] Open
Abstract
Liver fibrosis, a model wound healing system, is characterized by excessive deposition of extracellular matrix (ECM) in the liver. Although many fibrogenic cell types may express ECM, the hepatic stellate cell (HSC) is currently considered to be the major effector. HSCs transform into myofibroblast-like cells, also known as hepatic myofibroblasts in a process known as activation; this process is characterized in particular by de novo expression of smooth muscle alpha actin (SM α-actin) and type 1 collagen. The family of actins, which form the cell's cytoskeleton, are essential in many cellular processes. β-actin and cytoplasmic γ-actin (γ-actin) are ubiquitously expressed, whereas SM α-actin defines smooth muscle cell and myofibroblast phenotypes. Thus, SM α-actin is tightly associated with multiple functional properties. However, the regulatory mechanisms by which actin isoforms might regulate type 1 collagen remain unclear. In primary HSCs from normal and fibrotic rat liver, we demonstrate that myocardin, a canonical SRF cofactor, is upregulated in hepatic myofibroblasts and differentially regulates SM α-actin, γ-actin, and β-actins through activation of an ATTA box in the SM α-actin and a CCAAT box in γ-actin and β-actin promoters, respectively; moreover, myocardin differentially activated serum response factor (SRF) in CArG boxes of actin promoters. In addition, myocardin-stimulated Smad2 phosphorylation and RhoA expression, leading to increased expression of type 1 collagen in an actin cytoskeleton-dependent manner. Myocardin also directly enhanced SRF expression and stimulated collagen 1α1 and 1α2 promoter activities. In addition, overexpression of myocardin in vivo during carbon tetrachloride-induced liver injury led to increased HSC activation and fibrogenesis. In summary, our data suggest that myocardin plays a critical role in actin cytoskeletal dynamics during HSC activation, in turn, specifically regulating type I collagen expression in hepatic myofibroblasts.
Collapse
|
12
|
ROCKEY DONC. The Molecular Basis of Portal Hypertension. TRANSACTIONS OF THE AMERICAN CLINICAL AND CLIMATOLOGICAL ASSOCIATION 2017; 128:330-345. [PMID: 28790516 PMCID: PMC5525430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Cirrhosis leads to portal hypertension and vascular abnormalities in multiple vascular beds. There is intense vasoconstriction in the liver and the kidneys, but also vasodilation in the other vascular beds, including the periphery, lungs, brain, and mesentery. The derangement in each of these beds leads to specific clinical disease. The vasoconstrictive phenotype in the liver ultimately leads to clinical portal hypertension, and is caused by an imbalance of vasoconstrictive and vasorelaxing molecules, which will be the focus of this review.
Collapse
Affiliation(s)
- DON C. ROCKEY
- Correspondence and reprint requests: Don C. Rockey, MD, Department of Internal Medicine, Medical University of South Carolina,
96 Jonathan Lucas Street, Suite 803, MSC 623, Charleston, South Carolina 29425843-792-2914
| |
Collapse
|
13
|
Jansson D, Scotter EL, Rustenhoven J, Coppieters N, Smyth LCD, Oldfield RL, Bergin PS, Mee EW, Graham ES, Faull RLM, Dragunow M. Interferon-γ blocks signalling through PDGFRβ in human brain pericytes. J Neuroinflammation 2016; 13:249. [PMID: 27654972 PMCID: PMC5031293 DOI: 10.1186/s12974-016-0722-4] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Accepted: 09/13/2016] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND Neuroinflammation and blood-brain barrier (BBB) disruption are common features of many brain disorders, including Alzheimer's disease, epilepsy, and motor neuron disease. Inflammation is thought to be a driver of BBB breakdown, but the underlying mechanisms for this are unclear. Brain pericytes are critical cells for maintaining the BBB and are immunologically active. We sought to test the hypothesis that inflammation regulates the BBB by altering pericyte biology. METHODS We exposed primary adult human brain pericytes to chronic interferon-gamma (IFNγ) for 4 days and measured associated functional aspects of pericyte biology. Specifically, we examined the influence of inflammation on platelet-derived growth factor receptor-beta (PDGFRβ) expression and signalling, as well as pericyte proliferation and migration by qRT-PCR, immunocytochemistry, flow cytometry, and western blotting. RESULTS Chronic IFNγ treatment had marked effects on pericyte biology most notably through the PDGFRβ, by enhancing agonist (PDGF-BB)-induced receptor phosphorylation, internalization, and subsequent degradation. Functionally, chronic IFNγ prevented PDGF-BB-mediated pericyte proliferation and migration. CONCLUSIONS Because PDGFRβ is critical for pericyte function and its removal leads to BBB leakage, our results pinpoint a mechanism linking chronic brain inflammation to BBB dysfunction.
Collapse
Affiliation(s)
- Deidre Jansson
- Department of Pharmacology and Clinical Pharmacology, The University of Auckland, 1023, Auckland, New Zealand.,Gravida National Centre for Growth and Development, The University of Auckland, 1023, Auckland, New Zealand.,Centre for Brain Research, The University of Auckland, 1023, Auckland, New Zealand
| | - Emma L Scotter
- Department of Pharmacology and Clinical Pharmacology, The University of Auckland, 1023, Auckland, New Zealand.,Centre for Brain Research, The University of Auckland, 1023, Auckland, New Zealand
| | - Justin Rustenhoven
- Department of Pharmacology and Clinical Pharmacology, The University of Auckland, 1023, Auckland, New Zealand.,Centre for Brain Research, The University of Auckland, 1023, Auckland, New Zealand
| | - Natacha Coppieters
- Department of Anatomy and Medical Imaging, The University of Auckland, 1023, Auckland, New Zealand.,Centre for Brain Research, The University of Auckland, 1023, Auckland, New Zealand
| | - Leon C D Smyth
- Department of Pharmacology and Clinical Pharmacology, The University of Auckland, 1023, Auckland, New Zealand.,Centre for Brain Research, The University of Auckland, 1023, Auckland, New Zealand
| | | | - Peter S Bergin
- Centre for Brain Research, The University of Auckland, 1023, Auckland, New Zealand.,Auckland City Hospital, 1023, Auckland, New Zealand
| | - Edward W Mee
- Centre for Brain Research, The University of Auckland, 1023, Auckland, New Zealand.,Auckland City Hospital, 1023, Auckland, New Zealand
| | - E Scott Graham
- Department of Pharmacology and Clinical Pharmacology, The University of Auckland, 1023, Auckland, New Zealand.,Centre for Brain Research, The University of Auckland, 1023, Auckland, New Zealand
| | - Richard L M Faull
- Department of Anatomy and Medical Imaging, The University of Auckland, 1023, Auckland, New Zealand.,Centre for Brain Research, The University of Auckland, 1023, Auckland, New Zealand
| | - Mike Dragunow
- Department of Pharmacology and Clinical Pharmacology, The University of Auckland, 1023, Auckland, New Zealand. .,Gravida National Centre for Growth and Development, The University of Auckland, 1023, Auckland, New Zealand. .,Centre for Brain Research, The University of Auckland, 1023, Auckland, New Zealand. .,Department of Pharmacology and Clinical Pharmacology, The University of Auckland, Private Bag 92019, 1142, Auckland, New Zealand.
| |
Collapse
|
14
|
Rehman H, Liu Q, Krishnasamy Y, Shi Z, Ramshesh VK, Haque K, Schnellmann RG, Murphy MP, Lemasters JJ, Rockey DC, Zhong Z. The mitochondria-targeted antioxidant MitoQ attenuates liver fibrosis in mice. INTERNATIONAL JOURNAL OF PHYSIOLOGY, PATHOPHYSIOLOGY AND PHARMACOLOGY 2016; 8:14-27. [PMID: 27186319 PMCID: PMC4859875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Accepted: 03/15/2016] [Indexed: 06/05/2023]
Abstract
Oxidative stress plays an essential role in liver fibrosis. This study investigated whether MitoQ, an orally active mitochondrial antioxidant, decreases liver fibrosis. Mice were injected with corn oil or carbon tetrachloride (CCl4, 1:3 dilution in corn oil; 1 µl/g, ip) once every 3 days for up to 6 weeks. 4-Hydroxynonenal adducts increased markedly after CCl4 treatment, indicating oxidative stress. MitoQ attenuated oxidative stress after CCl4. Collagen 1α1 mRNA and hydroxyproline increased markedly after CCl4 treatment, indicating increased collagen formation and deposition. CCl4 caused overt pericentral fibrosis as revealed by both the sirius red staining and second harmonic generation microscopy. MitoQ blunted fibrosis after CCl4. Profibrotic transforming growth factor-β1 (TGF-β1) mRNA and expression of smooth muscle α-actin, an indicator of hepatic stellate cell (HSC) activation, increased markedly after CCl4 treatment. Smad 2/3, the major mediator of TGF-β fibrogenic effects, was also activated after CCl4 treatment. MitoQ blunted HSC activation, TGF-β expression, and Smad2/3 activation after CCl4 treatment. MitoQ also decreased necrosis, apoptosis and inflammation after CCl4 treatment. In cultured HSCs, MitoQ decreased oxidative stress, inhibited HSC activation, TGF-β1 expression, Smad2/3 activation, and extracellular signal-regulated protein kinase activation. Taken together, these data indicate that mitochondrial reactive oxygen species play an important role in liver fibrosis and that mitochondria-targeted antioxidants are promising potential therapies for prevention and treatment of liver fibrosis.
Collapse
Affiliation(s)
- Hasibur Rehman
- Department of Drug Discovery & Biomedical Sciences, Medical University of South CarolinaCharleston, SC 29425, USA
- Department of Biology, Faculty of Sciences, University of TabukSaudi Arabia
| | - Qinlong Liu
- Department of Drug Discovery & Biomedical Sciences, Medical University of South CarolinaCharleston, SC 29425, USA
- The Second Affiliated Hospital of Dalian Medical UniversityDalian, Liaoning Province, China
| | - Yasodha Krishnasamy
- Department of Drug Discovery & Biomedical Sciences, Medical University of South CarolinaCharleston, SC 29425, USA
| | - Zengdun Shi
- Department of Medicine, Medical University of South CarolinaCharleston, SC 29425, USA
| | - Venkat K Ramshesh
- Department of Drug Discovery & Biomedical Sciences, Medical University of South CarolinaCharleston, SC 29425, USA
| | - Khujista Haque
- Department of Drug Discovery & Biomedical Sciences, Medical University of South CarolinaCharleston, SC 29425, USA
| | - Rick G Schnellmann
- Department of Drug Discovery & Biomedical Sciences, Medical University of South CarolinaCharleston, SC 29425, USA
- Ralph H. Johnson VA Medical CenterCharleston, SC 29403, USA
| | - Michael P Murphy
- MRC Mitochondrial Biology Unit, Wellcome Trust/MRC BuildingCambridge CB2 0XY, U.K.
| | - John J Lemasters
- Department of Drug Discovery & Biomedical Sciences, Medical University of South CarolinaCharleston, SC 29425, USA
- Department of Biochemistry & Molecular Biology, Medical University of South CarolinaCharleston, SC 29425, USA
- Institute of Theoretical & Experimental Biophysics, Russian Academy of SciencesPushchino, Russian Federation
| | - Don C Rockey
- Department of Medicine, Medical University of South CarolinaCharleston, SC 29425, USA
| | - Zhi Zhong
- Department of Drug Discovery & Biomedical Sciences, Medical University of South CarolinaCharleston, SC 29425, USA
| |
Collapse
|
15
|
Singh S, Liu S, Rockey DC. Caveolin-1 is upregulated in hepatic stellate cells but not sinusoidal endothelial cells after liver injury. Tissue Cell 2016; 48:126-32. [PMID: 26847875 DOI: 10.1016/j.tice.2015.12.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Revised: 12/30/2015] [Accepted: 12/30/2015] [Indexed: 01/03/2023]
Abstract
Sinusoidal endothelial cells (SEC) and hepatic stellate cells (HSC) are closely associated specialized vascular cells residing in the hepatic sinusoid. These cells have been shown to play important roles in many different pathophysiologic processes, in particular in liver fibrosis/cirrhosis and portal hypertension. Caveolin-1 functions as a scaffolding protein, and has a variety of functions including in many disease states, such as liver cirrhosis. Although previous studies have shown that in the injured rat liver, caveolin-1 is upregulated, the precise cells in which remains unclear. Therefore, the purpose of this study was to clarify the cell type (or types) in which caveolin-1 is expressed in normal and injured rat liver. We have utilized both detailed immunohistochemical labeling with cell specific markers as well as cell isolation techniques (isolating sinusoidal endothelial cells, HSCs, and hepatocytes) in normal and injured (bile duct ligation) rat liver. We show here that in the normal liver caveolin-1 is expressed predominantly in HSCs and SECs but after liver injury there is upregulation of caveolin-1 in HSCs, but not in SECs. These data have functional implications for the cells in which caveolin-1 is regulated.
Collapse
Affiliation(s)
- Shweta Singh
- Medical University of South Carolina, Department of Medicine, Charleston, SC 29425, United States
| | - Songling Liu
- Medical University of South Carolina, Department of Medicine, Charleston, SC 29425, United States
| | - Don C Rockey
- Medical University of South Carolina, Department of Medicine, Charleston, SC 29425, United States.
| |
Collapse
|
16
|
Senger S, Sapone A, Fiorentino MR, Mazzarella G, Lauwers GY, Fasano A. Celiac Disease Histopathology Recapitulates Hedgehog Downregulation, Consistent with Wound Healing Processes Activation. PLoS One 2015; 10:e0144634. [PMID: 26649570 PMCID: PMC4674131 DOI: 10.1371/journal.pone.0144634] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2015] [Accepted: 11/21/2015] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND In celiac disease (CD), intestinal epithelium damage occurs secondary to an immune insult and is characterized by blunting of the villi and crypt hyperplasia. Similarities between Hedgehog (Hh)/BMP4 downregulation, as reported in a mouse model, and CD histopathology, suggest mechanistic involvement of Hh/BMP4/WNT pathways in proliferation and differentiation of immature epithelial cells in the context of human intestinal homeostasis and regeneration after damage. Herein we examined the nature of intestinal crypt hyperplasia and involvement of Hh/BMP4 in CD histopathology. METHODS AND FINDINGS Immunohistochemistry, qPCR and in situ hybridization were used to study a cohort of 24 healthy controls (HC) and 24 patients with diagnosed acute celiac disease (A-CD) intestinal biopsies. In A-CD we observed an increase in cells positive for Leucin-rich repeat-containing G protein-coupled receptor 5 (LGR5), an epithelial stem cell specific marker and expansion of WNT responding compartment. Further, we observed alteration in number and distribution of mesenchymal cells, predicted to be part of the intestinal stem cells niche. At the molecular level we found downregulation of indian hedgehog (IHH) and other components of the Hh pathway, but we did not observe a concurrent downregulation of BMP4. However, we observed upregulation of BMPs antagonists, gremlin 1 and gremlin 2. CONCLUSIONS Our data suggest that acute CD histopathology partially recapitulates the phenotype reported in Hh knockdown models. Specifically, Hh/BMP4 paradigm appears to be decoupled in CD, as the expansion of the immature cell population does not occur consequent to downregulation of BMP4. Instead, we provide evidence that upregulation of BMP antagonists play a key role in intestinal crypt hyperplasia. This study sheds light on the molecular mechanisms underlying CD histopathology and the limitations in the use of mouse models for celiac disease.
Collapse
Affiliation(s)
- Stefania Senger
- Center for Mucosal Immunology and Biology Research, Massachusetts General Hospital for Children and Celiac Program at Harvard Medical School, Charlestown, Massachusetts, United States of America
| | - Anna Sapone
- Center for Mucosal Immunology and Biology Research, Massachusetts General Hospital for Children and Celiac Program at Harvard Medical School, Charlestown, Massachusetts, United States of America
- Celiac Center, Division of Gastroenterology, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, United States of America
| | - Maria Rosaria Fiorentino
- Center for Mucosal Immunology and Biology Research, Massachusetts General Hospital for Children and Celiac Program at Harvard Medical School, Charlestown, Massachusetts, United States of America
| | - Giuseppe Mazzarella
- Institute of Food Sciences, National Research Council (CNR), Avellino, 83100, Italy
| | - Gregory Y. Lauwers
- Department of Pathology, Massachusetts General Hospital, 55 Fruit Street, Boston, Massachusetts, United States of America
| | - Alessio Fasano
- Center for Mucosal Immunology and Biology Research, Massachusetts General Hospital for Children and Celiac Program at Harvard Medical School, Charlestown, Massachusetts, United States of America
| |
Collapse
|
17
|
|
18
|
Brennan-Laun SE, Li XL, Ezelle HJ, Venkataraman T, Blackshear PJ, Wilson GM, Hassel BA. RNase L attenuates mitogen-stimulated gene expression via transcriptional and post-transcriptional mechanisms to limit the proliferative response. J Biol Chem 2014; 289:33629-43. [PMID: 25301952 DOI: 10.1074/jbc.m114.589556] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The cellular response to mitogens is tightly regulated via transcriptional and post-transcriptional mechanisms to rapidly induce genes that promote proliferation and efficiently attenuate their expression to prevent malignant growth. RNase L is an endoribonuclease that mediates diverse antiproliferative activities, and tristetraprolin (TTP) is a mitogen-induced RNA-binding protein that directs the decay of proliferation-stimulatory mRNAs. In light of their roles as endogenous proliferative constraints, we examined the mechanisms and functional interactions of RNase L and TTP to attenuate a mitogenic response. Mitogen stimulation of RNase L-deficient cells significantly increased TTP transcription and the induction of other mitogen-induced mRNAs. This regulation corresponded with elevated expression of serum-response factor (SRF), a master regulator of mitogen-induced transcription. RNase L destabilized the SRF transcript and formed a complex with SRF mRNA in cells providing a mechanism by which RNase L down-regulates SRF-induced genes. TTP and RNase L proteins interacted in cells suggesting that RNase L is directed to cleave TTP-bound RNAs as a mechanism of substrate specificity. Consistent with their concerted function in RNA turnover, the absence of either RNase L or TTP stabilized SRF mRNA, and a subset of established TTP targets was also regulated by RNase L. RNase L deficiency enhanced mitogen-induced proliferation demonstrating its functional role in limiting the mitogenic response. Our findings support a model of feedback regulation in which RNase L and TTP target SRF mRNA and SRF-induced transcripts. Accordingly, meta-analysis revealed an enrichment of RNase L and TTP targets among SRF-regulated genes suggesting that the RNase L/TTP axis represents a viable target to inhibit SRF-driven proliferation in neoplastic diseases.
Collapse
Affiliation(s)
- Sarah E Brennan-Laun
- From the Marlene and Stewart Greenebaum Cancer Center, Departments of Microbiology and Immunology and
| | - Xiao-Ling Li
- the Genetics Branch, NCI, National Institutes of Health, Bethesda, Maryland 20892
| | - Heather J Ezelle
- From the Marlene and Stewart Greenebaum Cancer Center, Departments of Microbiology and Immunology and the Research Services, Baltimore Veterans Affairs Medical Center, Baltimore, Maryland 21201, and
| | | | - Perry J Blackshear
- the Laboratory of Signal Transduction, NIEHS, National Institutes of Health, Research Triangle Park, North Carolina 27709
| | - Gerald M Wilson
- From the Marlene and Stewart Greenebaum Cancer Center, Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, Maryland 21201
| | - Bret A Hassel
- From the Marlene and Stewart Greenebaum Cancer Center, Departments of Microbiology and Immunology and the Research Services, Baltimore Veterans Affairs Medical Center, Baltimore, Maryland 21201, and
| |
Collapse
|
19
|
Iwakiri Y, Shah V, Rockey DC. Vascular pathobiology in chronic liver disease and cirrhosis - current status and future directions. J Hepatol 2014; 61:912-24. [PMID: 24911462 PMCID: PMC4346093 DOI: 10.1016/j.jhep.2014.05.047] [Citation(s) in RCA: 229] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2014] [Revised: 05/26/2014] [Accepted: 05/28/2014] [Indexed: 12/12/2022]
Abstract
Chronic liver disease is associated with remarkable alterations in the intra- and extrahepatic vasculature. Because of these changes, the fields of liver vasculature and portal hypertension have recently become closely integrated within the broader vascular biology discipline. As developments in vascular biology have evolved, a deeper understanding of vascular processes has led to a better understanding of the mechanisms of the dynamic vascular changes associated with portal hypertension and chronic liver disease. In this context, hepatic vascular cells, such as sinusoidal endothelial cells and pericyte-like hepatic stellate cells, are closely associated with one another, where they have paracrine and autocrine effects on each other and themselves. These cells play important roles in the pathogenesis of liver fibrosis/cirrhosis and portal hypertension. Further, a variety of signaling pathways have recently come to light. These include growth factor pathways involving cytokines such as transforming growth factor β, platelet derived growth factor, and others as well as a variety of vasoactive peptides and other molecules. An early and consistent feature of liver injury is the development of an increase in intra-hepatic resistance; this is associated with changes in hepatic vascular cells and their signaling pathway that cause portal hypertension. A critical concept is that this process aggregates signals to the extrahepatic circulation, causing derangement in this system's cells and signaling pathways, which ultimately leads to the collateral vessel formation and arterial vasodilation in the splanchnic and systemic circulation, which by virtue of the hydraulic derivation of Ohm's law (pressure = resistance × flow), worsens portal hypertension. This review provides a detailed review of the current status and future direction of the basic biology of portal hypertension with a focus on the physiology, pathophysiology, and signaling of cells within the liver, as well as those in the mesenteric vascular circulation. Translational implications of recent research and the future directions that it points to are also highlighted.
Collapse
Affiliation(s)
- Yasuko Iwakiri
- The Section of Digestive Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, United States
| | - Vijay Shah
- The Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, United States
| | - Don C Rockey
- The Department of Medicine, Medical University of South Carolina, Charleston, SC, United States.
| |
Collapse
|
20
|
Elpek G&O. Cellular and molecular mechanisms in the pathogenesis of liver fibrosis: An update. World J Gastroenterol 2014; 20:7260-7276. [PMID: 24966597 PMCID: PMC4064072 DOI: 10.3748/wjg.v20.i23.7260] [Citation(s) in RCA: 285] [Impact Index Per Article: 25.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2013] [Revised: 02/08/2014] [Accepted: 05/23/2014] [Indexed: 02/06/2023] Open
Abstract
There have been considerable recent advances towards a better understanding of the complex cellular and molecular network underlying liver fibrogenesis. Recent data indicate that the termination of fibrogenic processes and the restoration of deficient fibrolytic pathways may allow the reversal of advanced fibrosis and even cirrhosis. Therefore, efforts have been made to better clarify the cellular and molecular mechanisms that are involved in liver fibrosis. Activation of hepatic stellate cells (HSCs) remains a central event in fibrosis, complemented by other sources of matrix-producing cells, including portal fibroblasts, fibrocytes and bone marrow-derived myofibroblasts. These cells converge in a complex interaction with neighboring cells to provoke scarring in response to persistent injury. Defining the interaction of different cell types, revealing the effects of cytokines on these cells and characterizing the regulatory mechanisms that control gene expression in activated HSCs will enable the discovery of new therapeutic targets. Moreover, the characterization of different pathways associated with different etiologies aid in the development of disease-specific therapies. This article outlines recent advances regarding the cellular and molecular mechanisms involved in liver fibrosis that may be translated into future therapies. The pathogenesis of liver fibrosis associated with alcoholic liver disease, non-alcoholic fatty liver disease and viral hepatitis are also discussed to emphasize the various mechanisms involved in liver fibrosis.
Collapse
|
21
|
Li T, Shi Z, Rockey DC. Preproendothelin-1 expression is negatively regulated by IFNγ during hepatic stellate cell activation. Am J Physiol Gastrointest Liver Physiol 2012; 302:G948-57. [PMID: 22301113 PMCID: PMC3362071 DOI: 10.1152/ajpgi.00359.2011] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Endothelin-1 (ET-1), a powerful vasoconstrictor peptide, is produced by activated hepatic stellate cells (HSC) and promotes cell proliferation, fibrogenesis, and contraction, the latter of which has been thought to be mechanistically linked to portal hypertension in cirrhosis. Interferon-γ (IFNγ), a Th1 cytokine produced by T cells, inhibits stellate cell proliferation, fibrogenesis, and muscle-specific gene expression. Whether IFNγ-induced inhibitory effects are linked to regulation of ET-1 expression in activated stellate cells remains unknown. Here we examined IFNγ's effects on preproET-1 mRNA expression and the signaling pathways underlying this process. We demonstrated that preproET-1 mRNA expression in HSCs was prominently increased during cell culture-induced activation; IFNγ significantly inhibited both preproET-1 mRNA expression and ET-1 peptide production. Similar results were found in an in vivo model of liver injury and intraperitoneal administration of IFNγ. PreproET-1 promoter analysis revealed that IFNγ-induced inhibition of preproET-1 mRNA expression was closely linked to the AP-1 and Smad3 signaling pathways. Furthermore, IFNγ reduced JNK phosphorylation, which tightly was associated with decreased phosphorylation of downstream factors c-Jun and Smad3 and decreased binding activity of c-Jun and Smad3 in the preprpET-1 promoter. Importantly, IFNγ reduced both c-Jun mRNA and protein levels. Given the important role of ET-1 in wound healing, our results suggest a novel negative signaling network by which IFNγ inhibits preproET-1 expression, highlighting one potential molecular mechanism for IFNγ-induced host immunomodulation of liver fibrogenesis.
Collapse
Affiliation(s)
- Tianxia Li
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Zengdun Shi
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Don C. Rockey
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas
| |
Collapse
|