51
|
Liu H, Pan X, Cao H, Shu X, Sun H, Lu J, Liang J, Zhang K, Zhu F, Li G, Zhang Q. IL-32γ promotes integrin αvβ6 expression through the activation of NF-κB in HSCs. Exp Ther Med 2017; 14:3880-3886. [PMID: 29042996 DOI: 10.3892/etm.2017.4956] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2015] [Accepted: 02/01/2017] [Indexed: 12/11/2022] Open
Abstract
Hepatic stellate cell (HSC) activation is important in the pathogenesis of liver fibrosis. However, the molecular mechanism of HSC activation is not completely understood. In the present study, it was demonstrated that interleukin-32γ (IL-32γ) is capable of enhancing intefgrin αvβ6 expression by inducing integrin αvβ6 promoter activity in a dose-dependent manner in HSCs. Furthermore, it was determined that nuclear factor κB (NF-κB) activation is required for IL-32γ-induced integrin αvβ6 expression. Increased integrin αvβ6 expression is then able to activate HSCs. These results indicate that NF-κB activation is required for IL-32γ to induce integrin αvβ6 expression and consequently promote HSC activation. Therefore, IL-32γ activates HSCs and therefore may be associated with hepatic fibrogenesis. These results may enable the development of novel effective strategies to treat hepatic fibrosis.
Collapse
Affiliation(s)
- Hongcan Liu
- Department of Clinical Laboratory, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510630, P.R. China.,Department of Clinical Laboratory, The Second Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510120, P.R. China
| | - Xingfei Pan
- Department of Infectious Diseases, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong 510150, P.R. China
| | - Hong Cao
- Department of Infectious Diseases, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510630, P.R. China
| | - Xin Shu
- Department of Infectious Diseases, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510630, P.R. China
| | - Haixia Sun
- Department of Infectious Diseases, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510630, P.R. China
| | - Jianxi Lu
- Vaccine Research Institute, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510630, P.R. China
| | - Jiayin Liang
- Department of Clinical Laboratory, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510630, P.R. China
| | - Ka Zhang
- Department of Infectious Diseases, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510630, P.R. China
| | - Fengqin Zhu
- Department of Infectious Diseases, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510630, P.R. China
| | - Gang Li
- Department of Infectious Diseases, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510630, P.R. China
| | - Qi Zhang
- Vaccine Research Institute, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510630, P.R. China
| |
Collapse
|
52
|
Fujii M, Yoneda A, Takei N, Sakai-Sawada K, Kosaka M, Minomi K, Yokoyama A, Tamura Y. Endoplasmic reticulum oxidase 1α is critical for collagen secretion from and membrane type 1-matrix metalloproteinase levels in hepatic stellate cells. J Biol Chem 2017; 292:15649-15660. [PMID: 28774960 DOI: 10.1074/jbc.m117.783126] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2017] [Revised: 07/21/2017] [Indexed: 01/31/2023] Open
Abstract
Upon liver injury, excessive deposition of collagen from activated hepatic stellate cells (HSCs) is a leading cause of liver fibrosis. An understanding of the mechanism by which collagen biosynthesis is regulated in HSCs will provide important clues for practical anti-fibrotic therapy. Endoplasmic reticulum oxidase 1α (ERO1α) functions as an oxidative enzyme of protein disulfide isomerase, which forms intramolecular disulfide bonds of membrane and secreted proteins. However, the role of ERO1α in HSCs remains unclear. Here, we show that ERO1α is expressed and mainly localized in the endoplasmic reticulum in human HSCs. When HSCs were transfected with ERO1α siRNA or an ERO1α shRNA-expressing plasmid, expression of ERO1α was completely silenced. Silencing of ERO1α expression in HSCs markedly suppressed their proliferation but did not induce apoptosis, which was accompanied by impaired secretion of collagen type 1. Silencing of ERO1α expression induced impaired disulfide bond formation and inhibited autophagy via activation of the Akt/mammalian target of rapamycin signaling pathway, resulting in intracellular accumulation of collagen type 1 in HSCs. Furthermore, silencing of ERO1α expression also promoted proteasome-dependent degradation of membrane type 1-matrix metalloproteinase (MT1-MMP), which stimulates cell proliferation through cleavage of secreted collagens. The inhibition of HSC proliferation was reversed by treatment with MT1-MMP-cleaved collagen type 1. The results suggest that ERO1α plays a crucial role in HSC proliferation via posttranslational modification of collagen and MT1-MMP and, therefore, may be a suitable therapeutic target for managing liver fibrosis.
Collapse
Affiliation(s)
- Mizuki Fujii
- From the Department of Oral Functional Prosthodontics, Division of Oral Functional Science, Graduate School of Dental Medicine, Hokkaido University, Nishi-7, Kita-13, Kita-ku, Sapporo 060-8486, Japan.,the Department of Molecular Therapeutics, Center for Food and Medical Innovation, Institute for the Promotion of Business-Regional Collaboration, Hokkaido University, Nishi-11, Kita-21, Kita-ku, Sapporo 001-0021, Japan, and
| | - Akihiro Yoneda
- the Department of Molecular Therapeutics, Center for Food and Medical Innovation, Institute for the Promotion of Business-Regional Collaboration, Hokkaido University, Nishi-11, Kita-21, Kita-ku, Sapporo 001-0021, Japan, and
| | - Norio Takei
- the Department of Molecular Therapeutics, Center for Food and Medical Innovation, Institute for the Promotion of Business-Regional Collaboration, Hokkaido University, Nishi-11, Kita-21, Kita-ku, Sapporo 001-0021, Japan, and
| | - Kaori Sakai-Sawada
- the Department of Molecular Therapeutics, Center for Food and Medical Innovation, Institute for the Promotion of Business-Regional Collaboration, Hokkaido University, Nishi-11, Kita-21, Kita-ku, Sapporo 001-0021, Japan, and
| | - Marina Kosaka
- the Department of Molecular Therapeutics, Center for Food and Medical Innovation, Institute for the Promotion of Business-Regional Collaboration, Hokkaido University, Nishi-11, Kita-21, Kita-ku, Sapporo 001-0021, Japan, and.,the Research and Development Department, Nucleic Acid Medicine Business Division, Nitto Denko Corporation, Nishi-11, Kita-21, Kita-ku, Sapporo 001-0021, Japan
| | - Kenjiro Minomi
- the Department of Molecular Therapeutics, Center for Food and Medical Innovation, Institute for the Promotion of Business-Regional Collaboration, Hokkaido University, Nishi-11, Kita-21, Kita-ku, Sapporo 001-0021, Japan, and.,the Research and Development Department, Nucleic Acid Medicine Business Division, Nitto Denko Corporation, Nishi-11, Kita-21, Kita-ku, Sapporo 001-0021, Japan
| | - Atsuro Yokoyama
- From the Department of Oral Functional Prosthodontics, Division of Oral Functional Science, Graduate School of Dental Medicine, Hokkaido University, Nishi-7, Kita-13, Kita-ku, Sapporo 060-8486, Japan
| | - Yasuaki Tamura
- the Department of Molecular Therapeutics, Center for Food and Medical Innovation, Institute for the Promotion of Business-Regional Collaboration, Hokkaido University, Nishi-11, Kita-21, Kita-ku, Sapporo 001-0021, Japan, and
| |
Collapse
|
53
|
Zhang X, Guo Q, Shi Y, Xu W, Yu S, Yang Z, Cao L, Liu C, Zhao Z, Xin J. 99m Tc-3PRGD2 scintigraphy to stage liver fibrosis and evaluate reversal after fibrotic stimulus withdrawn. Nucl Med Biol 2017; 49:44-49. [DOI: 10.1016/j.nucmedbio.2017.02.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2016] [Revised: 02/01/2017] [Accepted: 02/26/2017] [Indexed: 12/14/2022]
|
54
|
Ansa-Addo EA, Thaxton J, Hong F, Wu BX, Zhang Y, Fugle CW, Metelli A, Riesenberg B, Williams K, Gewirth DT, Chiosis G, Liu B, Li Z. Clients and Oncogenic Roles of Molecular Chaperone gp96/grp94. Curr Top Med Chem 2017; 16:2765-78. [PMID: 27072698 DOI: 10.2174/1568026616666160413141613] [Citation(s) in RCA: 75] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2015] [Revised: 09/07/2015] [Accepted: 01/17/2016] [Indexed: 12/18/2022]
Abstract
As an endoplasmic reticulum heat shock protein (HSP) 90 paralogue, glycoprotein (gp) 96 possesses immunological properties by chaperoning antigenic peptides for activation of T cells. Genetic studies in the last decade have unveiled that gp96 is also an essential master chaperone for multiple receptors and secreting proteins including Toll-like receptors (TLRs), integrins, the Wnt coreceptor, Low Density Lipoprotein Receptor-Related Protein 6 (LRP6), the latent TGFβ docking receptor, Glycoprotein A Repetitions Predominant (GARP), Glycoprotein (GP) Ib and insulin-like growth factors (IGF). Clinically, elevated expression of gp96 in a variety of cancers correlates with the advanced stage and poor survival of cancer patients. Recent preclinical studies have also uncovered that gp96 expression is closely linked to cancer progression in multiple myeloma, hepatocellular carcinoma, breast cancer and inflammation-associated colon cancer. Thus, gp96 is an attractive therapeutic target for cancer treatment. The chaperone function of gp96 depends on its ATPase domain, which is structurally distinct from other HSP90 members, and thus favors the design of highly selective gp96-targeted inhibitors against cancer. We herein discuss the strategically important oncogenic clients of gp96 and their underlying biology. The roles of cell-intrinsic gp96 in T cell biology are also discussed, in part because it offers another opportunity of cancer therapy by manipulating levels of gp96 in T cells to enhance host immune defense.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | - Zihai Li
- Department of Microbiology and Immunology, Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29466, USA.
| |
Collapse
|
55
|
Chen L, Brigstock DR. Integrins and heparan sulfate proteoglycans on hepatic stellate cells (HSC) are novel receptors for HSC-derived exosomes. FEBS Lett 2016; 590:4263-4274. [PMID: 27714787 PMCID: PMC5154766 DOI: 10.1002/1873-3468.12448] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Revised: 09/22/2016] [Accepted: 09/22/2016] [Indexed: 12/15/2022]
Abstract
Exosomes mediate intercellular microRNA delivery between hepatic stellate cells (HSC), the principal fibrosis-producing cells in the liver. The purpose of this study was to identify receptors on HSC for HSC-derived exosomes, which bind to HSC rather than to hepatocytes. Our findings indicate that exosome binding to HSC is blocked by treating HSC with RGD, EDTA, integrin αv or β1 siRNAs, integrin αvβ3 or α5β1 neutralizing antibodies, heparin, or sodium chlorate. Furthermore, exosome cargo delivery and exosome-regulated functions in HSC, including expression of fibrosis- or activation-associated genes and/or miR-214 target gene regulation, are dependent on cellular integrin αvβ3, integrin α5β1, or heparan sulfate proteolgycans (HSPG). Thus, integrins and HSPG mediate the binding of HSC-derived exosomes to HSC as well as the delivery and intracellular action of the exosomal payload.
Collapse
Affiliation(s)
- Li Chen
- The Research Institute at Nationwide Children’s Hospital, 700 Children’s Drive, Columbus OH 43205 USA
| | - David R Brigstock
- The Research Institute at Nationwide Children’s Hospital, 700 Children’s Drive, Columbus OH 43205 USA
- Department of Surgery, Wexner Medical Center, The Ohio State University, Columbus, OH 43212 USA
| |
Collapse
|
56
|
Ramachandran P, Henderson NC. Antifibrotics in chronic liver disease: tractable targets and translational challenges. Lancet Gastroenterol Hepatol 2016; 1:328-340. [PMID: 28404203 DOI: 10.1016/s2468-1253(16)30110-8] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Revised: 07/22/2016] [Accepted: 07/26/2016] [Indexed: 12/30/2022]
Abstract
Chronic liver disease prevalence is increasing globally. Iterative liver damage, secondary to any cause of liver injury, results in progressive fibrosis, disrupted hepatic architecture, and aberrant regeneration, which are defining characteristics of liver cirrhosis. Liver transplantation is an effective treatment for end-stage liver disease; however, demand greatly outweighs donor organ supply, and in many parts of the world liver transplantation is unavailable. Hence, effective antifibrotic therapies are urgently required. In the past decade, rapid progress has been made in our understanding of the pathophysiology of liver fibrosis and a large number of potential cellular and molecular antifibrotic targets have been identified. This has led to numerous clinical trials of antifibrotic agents in patients with chronic liver disease. However, none of these have resulted in a robust and reproducible effect on fibrosis. It is therefore imperative that the ongoing translational challenges are addressed, to convert scientific discoveries into potent antifibrotics and enable bridging of the translational gap between putative therapeutic targets and effective treatments for patients with chronic liver disease.
Collapse
Affiliation(s)
- Prakash Ramachandran
- Medical Research Council (MRC) Centre for Inflammation Research, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Neil C Henderson
- Medical Research Council (MRC) Centre for Inflammation Research, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK.
| |
Collapse
|
57
|
Sugiyama A, Kanno K, Nishimichi N, Ohta S, Ono J, Conway SJ, Izuhara K, Yokosaki Y, Tazuma S. Periostin promotes hepatic fibrosis in mice by modulating hepatic stellate cell activation via α v integrin interaction. J Gastroenterol 2016; 51:1161-1174. [PMID: 27039906 DOI: 10.1007/s00535-016-1206-0] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2016] [Accepted: 03/26/2016] [Indexed: 02/04/2023]
Abstract
BACKGROUND Periostin is a matricellular protein that serves as a ligand for integrins and is required for tissue remodeling and fibrosis. We investigated the role of periostin in hepatic fibrosis and the mechanisms involved. METHODS Primary hepatic stellate cells (HSCs) and the HSC-immortalized cell line LX2 were used to study the profibrotic property of periostin and the interaction of periostin with integrins. Wild-type and periostin-deficient (periostin-/-) mice were subjected to two distinct models of liver fibrosis induced by hepatotoxic (carbon tetrachloride or thioacetamide) or cholestatic (3.5-diethoxycarbonyl-1.4-dihydrocollidine) injury. RESULTS Periostin expression in HSCs and LX2 cells increased in association with their activation. Gene silencing of periostin resulted in a significant reduction in the levels of profibrotic markers. In addition to enhanced cell migration in response to periostin, LX2 cells incubated on periostin showed significant induction of α-smooth muscle actin and collagen, indicating a profibrotic property. An antibody targeting αvβ5 and αvβ3 integrins suppressed cell attachment to periostin by 60 and 30 % respectively, whereas anti-α5β1 antibody had no effect. Consistently, αv integrin-silenced LX2 cells exhibited decreased attachment to periostin, with a significant reduction in the levels of profibrotic markers. Moreover, these profibrotic effects of periostin were observed in the mouse models. In contrast to extensive collagen deposition in wild-type mice, periostin-/- mice developed less noticeable hepatic fibrosis induced by hepatotoxic and cholestatic liver injury. Accordingly, the profibrotic markers were significantly reduced in periostin-/- mice. CONCLUSION Periostin exerts potent profibrotic activity mediated by αv integrin, suggesting the periostin-αv integrin axis as a novel therapeutic target for hepatic fibrosis.
Collapse
Affiliation(s)
- Akiko Sugiyama
- Department of General Internal Medicine, Hiroshima University Hospital, 1-2-3, Kasumi, Minami-ku, Hiroshima, 734-8551, Japan
| | - Keishi Kanno
- Department of General Internal Medicine, Hiroshima University Hospital, 1-2-3, Kasumi, Minami-ku, Hiroshima, 734-8551, Japan.
| | - Norihisa Nishimichi
- Cell-Matrix Frontier Laboratory, Biomedical Research Unit, Hiroshima University, 1-2-3, Kasumi, Minami-ku, Hiroshima, 734-8551, Japan
| | - Shoichiro Ohta
- Division of Medical Biochemistry, Department of Laboratory Medicine, Saga Medical School, 5-1-1, Nabeshima, Saga, 849-8501, Japan
| | - Junya Ono
- Central Institute, Shino-Test Corporation, 2-29-14, Oonodai Minami-ku, Sagamihara, Kanagawa, 252-0331, Japan
| | - Simon J Conway
- Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Kenji Izuhara
- Division of Medical Biochemistry, Department of Biomolecular Sciences, Saga Medical School, 5-1-1, Nabeshima, Saga, 849-8501, Japan
| | - Yasuyuki Yokosaki
- Cell-Matrix Frontier Laboratory, Biomedical Research Unit, Hiroshima University, 1-2-3, Kasumi, Minami-ku, Hiroshima, 734-8551, Japan
| | - Susumu Tazuma
- Department of General Internal Medicine, Hiroshima University Hospital, 1-2-3, Kasumi, Minami-ku, Hiroshima, 734-8551, Japan
| |
Collapse
|
58
|
Haldar D, Henderson NC, Hirschfield G, Newsome PN. Mesenchymal stromal cells and liver fibrosis: a complicated relationship. FASEB J 2016; 30:3905-3928. [DOI: 10.1096/fj.201600433r] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Accepted: 08/15/2016] [Indexed: 12/17/2022]
Affiliation(s)
- Debashis Haldar
- National Institute for Health ResearchBirmingham Liver Biomedical Research Unit and Centre for Liver Research University of Birmingham Birmingham United Kingdom
- Liver UnitUniversity Hospital Birmingham National Health Service (NHS) Foundation Trust Birmingham United Kingdom
| | - Neil C. Henderson
- Medical Research Council (MRC) Centre for Inflammation ResearchQueens Medical Research Institute University of Edinburgh Edinburgh United Kingdom
| | - Gideon Hirschfield
- National Institute for Health ResearchBirmingham Liver Biomedical Research Unit and Centre for Liver Research University of Birmingham Birmingham United Kingdom
- Liver UnitUniversity Hospital Birmingham National Health Service (NHS) Foundation Trust Birmingham United Kingdom
| | - Philip N. Newsome
- National Institute for Health ResearchBirmingham Liver Biomedical Research Unit and Centre for Liver Research University of Birmingham Birmingham United Kingdom
- Liver UnitUniversity Hospital Birmingham National Health Service (NHS) Foundation Trust Birmingham United Kingdom
| |
Collapse
|
59
|
Li F, Yan H, Wang J, Li C, Wu J, Wu S, Rao S, Gao X, Jin Q. Non-invasively differentiating extent of liver fibrosis by visualizing hepatic integrin αvβ3 expression with an MRI modality in mice. Biomaterials 2016; 102:162-74. [DOI: 10.1016/j.biomaterials.2016.06.026] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2016] [Revised: 05/20/2016] [Accepted: 06/13/2016] [Indexed: 12/11/2022]
|
60
|
Dzobo K, Vogelsang M, Parker MI. Wnt/β-Catenin and MEK-ERK Signaling are Required for Fibroblast-Derived Extracellular Matrix-Mediated Endoderm Differentiation of Embryonic Stem Cells. Stem Cell Rev Rep 2016; 11:761-73. [PMID: 26022506 DOI: 10.1007/s12015-015-9598-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Human embryonic stem cells (hESCs) have the potential to differentiate into all cells of the three germ layers, thus making them an attractive source of cells for use in regenerative medicine. The greatest challenge lies in regulating the differentiation of hESCs into specific cell lineages by both intrinsic and extrinsic factors. In this study we determined the effect of a fibroblast-derived extracellular matrix (fd-ECM) on hESCs differentiation. We demonstrate that growth of hESCs on fd-ECM results in hESCs losing their stemness and proliferation potential. As the stem cells differentiate they attain gene expression profiles similar to the primitive streak of the in vivo embryo. The activation of both the MEK-ERK and Wnt/β-catenin signaling pathways is required for the fd-ECM-mediated differentiation of hESCs towards the endoderm and involves integrins α1, α2, α3 and β1. This study illustrates the importance of the cellular microenvironment in directing stem cell fate and that the nature and composition of the extracellular matrix is a crucial determining factor.
Collapse
Affiliation(s)
- Kevin Dzobo
- Cape Town Component, Wernher and Beit Building (South), UCT Campus, International Centre for Genetic Engineering and Biotechnology (ICGEB), Anzio Road, Observatory, 7925, Cape Town, South Africa
| | | | | |
Collapse
|
61
|
Maintaining human fetal pancreatic stellate cell function and proliferation require β1 integrin and collagen I matrix interactions. Oncotarget 2016; 6:14045-59. [PMID: 26062655 PMCID: PMC4546450 DOI: 10.18632/oncotarget.4338] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2015] [Accepted: 05/22/2015] [Indexed: 12/29/2022] Open
Abstract
Pancreatic stellate cells (PaSCs) are cells that are located around the acinar, ductal, and vasculature tissue of the rodent and human pancreas, and are responsible for regulating extracellular matrix (ECM) turnover and maintaining the architecture of pancreatic tissue. This study examines the contributions of integrin receptor signaling in human PaSC function and survival. Human PaSCs were isolated from pancreata collected during the 2nd trimester of pregnancy and identified by expression of stellate cell markers, ECM proteins and associated growth factors. Multiple integrins are found in isolated human PaSCs, with high levels of β1, α3 and α5. Cell adhesion and migration assays demonstrated that human PaSCs favour collagen I matrix, which enhanced PaSC proliferation and increased TGFβ1, CTGF and α3β1 integrin. Significant activation of FAK/ERK and AKT signaling pathways, and up-regulation of cyclin D1 protein levels, were observed within PaSCs cultured on collagen I matrix. Blocking β1 integrin significantly decreased PaSC adhesion, migration and proliferation, further complementing the aforementioned findings. This study demonstrates that interaction of β1 integrin with collagen I is required for the proliferation and function of human fetal PaSCs, which may contribute to the biomedical engineering of the ECM microenvironment needed for the efficient regulation of pancreatic development.
Collapse
|
62
|
Conroy KP, Kitto LJ, Henderson NC. αv integrins: key regulators of tissue fibrosis. Cell Tissue Res 2016; 365:511-9. [PMID: 27139180 PMCID: PMC5010580 DOI: 10.1007/s00441-016-2407-9] [Citation(s) in RCA: 108] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2016] [Accepted: 04/08/2016] [Indexed: 01/09/2023]
Abstract
Chronic tissue injury with fibrosis results in the disruption of tissue architecture, organ dysfunction and eventual organ failure. Therefore, the development of effective anti-fibrotic therapies is urgently required. During fibrogenesis, complex interplay occurs between cellular and extracellular matrix components of the wound healing response. Integrins, a family of transmembrane cell adhesion molecules, play a key role in mediating intercellular and cell-matrix interactions. Thus, integrins provide a major node of communication between the extracellular matrix, inflammatory cells, fibroblasts and parenchymal cells and, as such, are intimately involved in the initiation, maintenance and resolution of tissue fibrosis. Modulation of members of the αv integrin family has exhibited profound effects on fibrosis in multiple organs and disease states. In this review, we discuss the current knowledge of the mechanisms of αv-integrin-mediated regulation of fibrogenesis and show that the therapeutic targeting of specific αv integrins represents a promising avenue to treat patients with a broad range of fibrotic diseases.
Collapse
Affiliation(s)
- Kylie P Conroy
- MRC Centre for Inflammation Research, The Queen's Medical Research Institute, University of Edinburgh, 47 Little France Crescent, Edinburgh, EH16 4TJ, UK
| | - Laura J Kitto
- MRC Centre for Inflammation Research, The Queen's Medical Research Institute, University of Edinburgh, 47 Little France Crescent, Edinburgh, EH16 4TJ, UK
| | - Neil C Henderson
- MRC Centre for Inflammation Research, The Queen's Medical Research Institute, University of Edinburgh, 47 Little France Crescent, Edinburgh, EH16 4TJ, UK.
| |
Collapse
|
63
|
Rieder F, Bettenworth D, Imai J, Inagaki Y. Intestinal Fibrosis and Liver Fibrosis: Consequences of Chronic Inflammation or Independent Pathophysiology? Inflamm Intest Dis 2016; 1:41-49. [PMID: 29922656 DOI: 10.1159/000445135] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Accepted: 03/02/2016] [Indexed: 02/06/2023] Open
Abstract
Background Intestinal fibrosis and liver fibrosis represent a significant burden for our patients and health-care systems. Despite the severe clinical problem and the observation that fibrosis is reversible, no specific antifibrotic therapies exist. Summary In this review, using an 'East-West' scientific collaboration, we summarize the current knowledge on principal mechanisms shared by intestinal fibrosis and liver fibrosis. We furthermore discuss inflammation as the cause of fibrogenesis in both entities, depict unique features of intestinal and hepatic fibrosis, and provide a future outlook on the development of antifibrotic therapies. Key Messages A collaborative effort in the field of fibrosis, covering multiple organ systems, will have the highest chance of leading to the development of a successful antifibrotic intervention.
Collapse
Affiliation(s)
- Florian Rieder
- Department of Gastroenterology and Hepatology, Digestive Disease Institute, Cleveland, Ohio, USA.,Department of Pathobiology, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, Ohio, USA
| | | | - Jin Imai
- Center for Matrix Biology and Medicine, Graduate School of Medicine, Tokai University, Isehara, Japan.,Department of Gastroenterology, Tokai University School of Medicine, Isehara, Japan
| | - Yutaka Inagaki
- Center for Matrix Biology and Medicine, Graduate School of Medicine, Tokai University, Isehara, Japan.,Department of Regenerative Medicine, Tokai University School of Medicine, Isehara, Japan
| |
Collapse
|
64
|
Zhang C, Liu H, Cui Y, Li X, Zhang Z, Zhang Y, Wang D. Molecular magnetic resonance imaging of activated hepatic stellate cells with ultrasmall superparamagnetic iron oxide targeting integrin αvβ₃ for staging liver fibrosis in rat model. Int J Nanomedicine 2016; 11:1097-108. [PMID: 27051285 PMCID: PMC4807947 DOI: 10.2147/ijn.s101366] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Purpose To evaluate the expression level of integrin αvβ3 on activated hepatic stellate cells (HSCs) at different stages of liver fibrosis induced by carbon tetrachloride (CCl4) in rat model and the feasibility to stage liver fibrosis by using molecular magnetic resonance imaging (MRI) with arginine-glycine-aspartic acid (RGD) peptide modified ultrasmall superparamagnetic iron oxide nanoparticle (USPIO) specifically targeting integrin αvβ3. Materials and methods All experiments received approval from our Institutional Animal Care and Use Committee. Thirty-six rats were randomly divided into three groups of 12 subjects each, and intraperitoneally injected with CCl4 for either 3, 6, or 9 weeks. Controls (n=10) received pure olive oil. The change in T2* relaxation rate (ΔR2*) pre- and postintravenous administration of RGD-USPIO or naked USPIO was measured by 3.0T clinical MRI and compared by one-way analysis of variance or the Student’s t-test. The relationship between expression level of integrin αvβ3 and liver fibrotic degree was evaluated by Spearman’s ranked correlation. Results Activated HSCs were confirmed to be the main cell types expressing integrin αvβ3 during liver fibrogenesis. The protein level of integrin αv and β3 subunit expressed on activated HSCs was upregulated and correlated well with the progression of liver fibrosis (r=0.954, P<0.001; r=0.931, P<0.001, respectively). After injection of RGD-USPIO, there is significant difference in ΔR2* among rats treated with 0, 3, 6, and 9 weeks of CCl4 (P<0.001). The accumulation of iron particles in fibrotic liver specimen is significantly greater for RGD-USPIO than naked USPIO after being injected with equal dose of iron. Conclusion Molecular MRI of integrin αvβ3 expressed on activated HSCs by using RGD-USPIO may distinguish different liver fibrotic stages in CCl4 rat model and shows promising to noninvasively monitor the progression of the liver fibrosis and therapeutic response to antifibrotic treatment.
Collapse
Affiliation(s)
- Caiyuan Zhang
- Department of Radiology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Huanhuan Liu
- Department of Radiology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Yanfen Cui
- Department of Radiology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Xiaoming Li
- Department of Radiology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Zhongyang Zhang
- Department of Radiology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Yong Zhang
- MR Advanced Application and Research Center, GE Healthcare China, Shanghai, People's Republic of China
| | - Dengbin Wang
- Department of Radiology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| |
Collapse
|
65
|
Liu J, Quan J, Feng J, Zhang Q, Xu Y, Liu J, Huang W, Liu J, Tian L. High glucose regulates LN expression in human liver sinusoidal endothelial cells through ROS/integrin αvβ3 pathway. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2016; 42:231-236. [PMID: 26896612 DOI: 10.1016/j.etap.2016.01.021] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/11/2015] [Revised: 01/24/2016] [Accepted: 01/25/2016] [Indexed: 06/05/2023]
Abstract
Diabetes mellitus can cause a wide variety of vascular complications and is one of the major risk factors for Non Alcoholic Fatty Liver Disease (NAFLD). The present study was designed investigate the expression of laminin (LN) in human liver sinusoidal endothelial cells (HLSECs) induced by high glucose and the role of reactive oxygen species (ROS) and integrin αvβ3 in the regulation of LN expression. HLSECs were cultured and treated with media containing 25 mM glucose in the presence or absence of N-acetylcysteine (NAC) or clone LM609. The level of intracellular ROS of HLSECs was measured with 2',7' dichloro-fluorescein diacetate (DCFH-DA) probe. Expression of integrin αvβ3 was measured using RT-PCR and Western blot. Expression of LN was testified by immunofluorescence assay. Compared with that in control group, ROS level and the expression of integrin αvβ3 and LN increased in high glucose group. Compared with that in high glucose group, antioxidant NAC inhibited the expression of integrin αvβ3, NAC and the anti-body for blocking integrin αvβ3 (clone LM609) down-regulated the expression of LN. However, the above parameters did not differ between control and mannitol groups. High glucose up-regulates expression of LN in HLSECs through ROS/integrin αvβ3 pathway.
Collapse
Affiliation(s)
- Jing Liu
- Department of Endocrinology, Gansu Provincial Hospital, Lanzhou, Gansu 730000, China
| | - Jinxing Quan
- Department of Endocrinology, Gansu Provincial Hospital, Lanzhou, Gansu 730000, China
| | - Jing Feng
- Department of Endocrinology, Gansu Provincial Hospital, Lanzhou, Gansu 730000, China
| | - Qi Zhang
- Department of Endocrinology, Gansu Provincial Hospital, Lanzhou, Gansu 730000, China
| | - Yanjia Xu
- Department of Endocrinology, Gansu Provincial Hospital, Lanzhou, Gansu 730000, China
| | - Jia Liu
- Department of Endocrinology, Gansu Provincial Hospital, Lanzhou, Gansu 730000, China
| | - Wenhui Huang
- Department of Endocrinology, Gansu Provincial Hospital, Lanzhou, Gansu 730000, China
| | - Juxiang Liu
- Department of Endocrinology, Gansu Provincial Hospital, Lanzhou, Gansu 730000, China
| | - Limin Tian
- Department of Endocrinology, Gansu Provincial Hospital, Lanzhou, Gansu 730000, China.
| |
Collapse
|
66
|
Versican: a novel modulator of hepatic fibrosis. J Transl Med 2016; 96:361-74. [PMID: 26752747 DOI: 10.1038/labinvest.2015.152] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Revised: 10/29/2015] [Accepted: 11/02/2015] [Indexed: 01/17/2023] Open
Abstract
Little is known about the deposition and turnover of proteoglycans in liver fibrosis, despite their abundance in the extracellular matrix. Versican plays diverse roles in modulating cell behavior in other fibroproliferative diseases, but remains poorly described in the liver. Hepatic fibrosis was induced by carbon tetrachloride treatment of C57BL/6 mice over 4 weeks followed by recovery over a 28-day period. Primary mouse hepatic stellate cells (HSCs) were activated in culture and versican was transiently knocked down in human (LX2) and mouse HSCs. Expression of versican, A Disintegrin-like and Metalloproteinase with Thrombospondin-1 motifs (ADAMTS)-1, -4, -5, -8, -9, -15, and -20, and markers of fibrogenesis were studied using immunohistochemistry, real-time quantitative PCR, and western blotting. Immunohistochemistry showed increased expression of versican in cirrhotic human livers and the mouse model of fibrosis. Carbon tetrachloride treatment led to significant increases in versican expression and the proteoglycanases ADAMTS-5, -9, -15, and -20, alongside TNF-α, α-smooth muscle actin (α-SMA), collagen-1, and TGF-β expression. During recovery, expression of many of these genes returned to control levels. However, expression of ADAMTS-5, -8, -9, and -15 showed delayed increases in expression at 28 days of recovery, which corresponded with decreases in versican V0 and V1 cleavage products (G1-DPEAAE(1401) and G1-DPEAAE(441)). Activation of primary HSCs in vitro significantly increased versican, α-SMA, and collagen-1 expression. Transient knockdown of versican in HSCs led to decreases in markers of fibrogenesis and reduced cell proliferation, without inducing apoptosis. Versican expression increases during HSC activation and liver fibrosis, and proteolytic processing occurs during the resolution of fibrosis. Knockdown studies in vitro suggest a possible role of versican in modulating hepatic fibrogenesis.
Collapse
|
67
|
Yoneda A, Sakai-Sawada K, Niitsu Y, Tamura Y. Vitamin A and insulin are required for the maintenance of hepatic stellate cell quiescence. Exp Cell Res 2016; 341:8-17. [PMID: 26812497 DOI: 10.1016/j.yexcr.2016.01.012] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2015] [Revised: 01/22/2016] [Accepted: 01/22/2016] [Indexed: 01/26/2023]
Abstract
Transdifferentiation of vitamin A-storing hepatic stellate cells (HSCs) to vitamin A-depleted myofibroblastic cells leads to liver fibrosis. Vitamin A regulates lipid accumulation and gene transcription, suggesting that vitamin A is involved in the maintenance of HSC quiescence under a physiological condition. However, the precise mechanism remains elusive because there is no appropriate in vitro culture system for quiescent HSCs. Here, we show that treatment of quiescent HSCs with vitamin A partially maintained the accumulation of lipid droplets and expression of quiescent HSC markers (glial fibrillary acidic protein, peroxisome proliferator-activator receptor-γ and CCAAT/enhancer-binding protein-α) and also the expression of myofibroblastic markers (α-smooth muscle actin, heat shock protein 47 and collagen type I). On the other hand, combined treatment with vitamin A and insulin sustained the characteristic of HSC quiescence and completely suppressed the expression of myofibroblastic markers through activation of the JAK2/STAT5 signaling pathway and increased expression of sterol regulatory element binding protein-1. These treated HSCs transdifferentiated to myofibroblastic cells under a culture condition with fetal bovine serum. The results suggest an important role of vitamin A and insulin in the maintenance of HSC quiescence under a physiological condition.
Collapse
Affiliation(s)
- Akihiro Yoneda
- Department of Molecular Therapeutics, Center for Food & Medical Innovation, Institute for the Promotion of Business-Regional Collaboration, Hokkaido University, West-11, North-21, Kita-ku, Sapporo 001-0021, Hokkaido, Japan.
| | - Kaori Sakai-Sawada
- Department of Molecular Therapeutics, Center for Food & Medical Innovation, Institute for the Promotion of Business-Regional Collaboration, Hokkaido University, West-11, North-21, Kita-ku, Sapporo 001-0021, Hokkaido, Japan
| | - Yoshiro Niitsu
- Department of Molecular Target Exploration, School of Medicine, Sapporo Medical University, Japan
| | - Yasuaki Tamura
- Department of Molecular Therapeutics, Center for Food & Medical Innovation, Institute for the Promotion of Business-Regional Collaboration, Hokkaido University, West-11, North-21, Kita-ku, Sapporo 001-0021, Hokkaido, Japan
| |
Collapse
|
68
|
Abstract
Intestinal fibrosis is a common feature of Crohn's disease and may appear as a stricture, stenosis, or intestinal obstruction. Fibrostenosing Crohn's disease leads to a significantly impaired quality of life in affected patients and constitutes a challenging treatment situation. In the absence of specific medical antifibrotic treatment options, endoscopic or surgical therapy approaches with their potential harmful side effects are frequently used. However, our understanding of mechanisms of fibrogenesis in general and specifically intestinal fibrosis has emerged. Progression of fibrosis in the liver, lung, or skin can be halted or even reversed, and possible treatment targets have been identified. In face of this observation and given the fact that fibrotic alterations in various organs of the human body share distinct core characteristics, this article aims to address whether reversibility of intestinal fibrosis may be conceivable and to highlight promising research avenues and therapies.
Collapse
Affiliation(s)
| | - Florian Rieder
- Department of Gastroenterology, Hepatology & Nutrition, Digestive Disease Institute, Cleveland Clinic Foundation, Cleveland, Ohio, USA
- Department of Pathobiology, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, Ohio, USA
| |
Collapse
|
69
|
Benitez PL, Mascharak S, Proctor AC, Heilshorn SC. Use of protein-engineered fabrics to identify design rules for integrin ligand clustering in biomaterials. Integr Biol (Camb) 2015; 8:50-61. [PMID: 26692238 DOI: 10.1039/c5ib00258c] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
While ligand clustering is known to enhance integrin activation, this insight has been difficult to apply to the design of implantable biomaterials because the local and global ligand densities that enable clustering-enhanced integrin signaling were unpredictable. Here, two general design principles for biomaterial ligand clustering are elucidated. First, clustering ligands enhances integrin-dependent signals when the global ligand density, i.e., the ligand density across the cellular length scale, is near the ligand's effective dissociation constant (KD,eff). Second, clustering ligands enhances integrin activation when the local ligand density, i.e., the ligand density across the length scale of individual focal adhesions, is less than an overcrowding threshold. To identify these principles, we fabricated a series of elastin-like, electrospun fabrics with independent control over the local (0 to 122 000 ligands μm(-2)) and global (0 to 71 000 ligand μm(-2)) densities of an arginine-glycine-aspartate (RGD) ligand. Antibody blocking studies confirmed that human umbilical vein endothelial cell adhesion to these protein-engineered biomaterials was primarily due to αVβ3 integrin binding. Clustering ligands enhanced cell proliferation, focal adhesion number, and focal adhesion kinase expression near the ligand's KD,eff of 12 000 RGD μm(-2). Near this global ligand density, cells on ligand-clustered fabrics behaved similarly to cells grown on fabrics with significantly larger global ligand densities but without clustering. However, this enhanced ligand-clustering effect was not observed above a threshold cut-off concentration. At a local ligand density of 122 000 RGD μm(-2), cell division, focal adhesion number, and focal adhesion kinase expression were significantly reduced relative to fabrics with identical global ligand density and lesser local ligand densities. Thus, when clustering results in overcrowding of ligands, integrin receptors are no longer able to effectively engage with their target ligands. Together, these two insights into the cellular responses to ligand clustering at the cell-matrix interface may serve as design principles when developing future generations of implantable biomaterials.
Collapse
Affiliation(s)
- Patrick L Benitez
- Department of Bioengineering, Stanford University, 476 Lomita Mall, Stanford, CA 94305,
| | - Shamik Mascharak
- Department of Bioengineering, Stanford University, 476 Lomita Mall, Stanford, CA 94305,
| | - Amy C Proctor
- Department of Chemical Engineering, Stanford University, 476 Lomita Mall, Stanford, CA 94305,
| | - Sarah C Heilshorn
- Department of Materials Science and Engineering, Stanford University, 476 Lomita Mall, Stanford, CA 94305
| |
Collapse
|
70
|
Yu X, Wu Y, Liu H, Gao L, Sun X, Zhang C, Shi J, Zhao H, Jia B, Liu Z, Wang F. Small-Animal SPECT/CT of the Progression and Recovery of Rat Liver Fibrosis by Using an Integrin αvβ3-targeting Radiotracer. Radiology 2015; 279:502-12. [PMID: 26669696 DOI: 10.1148/radiol.2015150090] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
PURPOSE To assess the potential utility of an integrin αvβ3-targeting radiotracer, technetium 99m-PEG4-E[PEG4-cyclo(arginine-glycine-aspartic acid-D-phenylalanine-lysine)]2 ((99m)Tc-3PRGD2), for single photon emission computed tomography (SPECT)/computed tomography (CT) for monitoring of the progression and prognosis of liver fibrosis in a rat model. MATERIALS AND METHODS All animal experiments were performed by following the protocol approved by the institutional animal care and use committee. (99m)Tc-3PRGD2 was prepared and longitudinal SPECT/CT was performed to monitor the progression (n = 8) and recovery (n = 5) of liver fibrosis induced in a rat model by means of thioacetamide (TAA) administration. The mean liver-to-background radioactivity per unit volume ratio was analyzed for comparisons between the TAA and control (saline) groups at different stages of liver fibrosis. Data were compared by using Student t and Mann-Whitney tests. Results:of SPECT/CT were compared with those of ex vivo biodistribution analysis (n = 5). RESULTS Accumulation of (99m)Tc-3PRGD2 in the liver increased in proportion to the progression of fibrosis and TAA exposure time; accumulation levels were significantly different between the TAA and control groups as early as week 4 of TAA administration (liver-to-background ratio: 32.30 ± 3.39 vs 19.01 ± 3.31; P = .0002). Results of ex vivo immunofluorescence staining demonstrated the positive expression of integrin αvβ3 on the activated hepatic stellate cells, and the integrin αvβ3 levels in the liver corresponded to the results of SPECT/CT (R(2) = 0.75, P < .0001). (99m)Tc-3PRGD2 uptake in the fibrotic liver decreased after antifibrotic therapy with interferon α2b compared with that in the control group (relative liver-to-background ratio: 0.45 ± 0.05 vs 1.01 ± 0.05; P < .0001) or spontaneous recovery (relative liver-to-background ratio: 0.56 ± 0.06 vs 1.01 ± 0.05; P < .0001). CONCLUSION (99m)Tc-3PRGD2 SPECT/CT was successfully used to monitor the progression and recovery of liver fibrosis and shows potential applications for noninvasive diagnosis of early stage liver fibrosis.
Collapse
Affiliation(s)
- Xinhe Yu
- From the Medical Isotopes Research Center and Department of Radiation Medicine, School of Basic Medical Sciences, Peking University Health Science Center, 38 Xueyuan Rd, Beijing 100191, China (X.Y., Y.W., H.L., L.G., S.X., C.Z., J.S., H.Z., B.J., Z.L., F.W.); Interdisciplinary Laboratory, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China (J.S., F.W.); and State Key Laboratory of Natural and Biomimetic Drugs, Center for Molecular and Translational Medicine, Peking University, Beijing, China (F.W.)
| | - Yue Wu
- From the Medical Isotopes Research Center and Department of Radiation Medicine, School of Basic Medical Sciences, Peking University Health Science Center, 38 Xueyuan Rd, Beijing 100191, China (X.Y., Y.W., H.L., L.G., S.X., C.Z., J.S., H.Z., B.J., Z.L., F.W.); Interdisciplinary Laboratory, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China (J.S., F.W.); and State Key Laboratory of Natural and Biomimetic Drugs, Center for Molecular and Translational Medicine, Peking University, Beijing, China (F.W.)
| | - Hao Liu
- From the Medical Isotopes Research Center and Department of Radiation Medicine, School of Basic Medical Sciences, Peking University Health Science Center, 38 Xueyuan Rd, Beijing 100191, China (X.Y., Y.W., H.L., L.G., S.X., C.Z., J.S., H.Z., B.J., Z.L., F.W.); Interdisciplinary Laboratory, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China (J.S., F.W.); and State Key Laboratory of Natural and Biomimetic Drugs, Center for Molecular and Translational Medicine, Peking University, Beijing, China (F.W.)
| | - Liquan Gao
- From the Medical Isotopes Research Center and Department of Radiation Medicine, School of Basic Medical Sciences, Peking University Health Science Center, 38 Xueyuan Rd, Beijing 100191, China (X.Y., Y.W., H.L., L.G., S.X., C.Z., J.S., H.Z., B.J., Z.L., F.W.); Interdisciplinary Laboratory, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China (J.S., F.W.); and State Key Laboratory of Natural and Biomimetic Drugs, Center for Molecular and Translational Medicine, Peking University, Beijing, China (F.W.)
| | - Xianlei Sun
- From the Medical Isotopes Research Center and Department of Radiation Medicine, School of Basic Medical Sciences, Peking University Health Science Center, 38 Xueyuan Rd, Beijing 100191, China (X.Y., Y.W., H.L., L.G., S.X., C.Z., J.S., H.Z., B.J., Z.L., F.W.); Interdisciplinary Laboratory, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China (J.S., F.W.); and State Key Laboratory of Natural and Biomimetic Drugs, Center for Molecular and Translational Medicine, Peking University, Beijing, China (F.W.)
| | - Chenran Zhang
- From the Medical Isotopes Research Center and Department of Radiation Medicine, School of Basic Medical Sciences, Peking University Health Science Center, 38 Xueyuan Rd, Beijing 100191, China (X.Y., Y.W., H.L., L.G., S.X., C.Z., J.S., H.Z., B.J., Z.L., F.W.); Interdisciplinary Laboratory, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China (J.S., F.W.); and State Key Laboratory of Natural and Biomimetic Drugs, Center for Molecular and Translational Medicine, Peking University, Beijing, China (F.W.)
| | - Jiyun Shi
- From the Medical Isotopes Research Center and Department of Radiation Medicine, School of Basic Medical Sciences, Peking University Health Science Center, 38 Xueyuan Rd, Beijing 100191, China (X.Y., Y.W., H.L., L.G., S.X., C.Z., J.S., H.Z., B.J., Z.L., F.W.); Interdisciplinary Laboratory, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China (J.S., F.W.); and State Key Laboratory of Natural and Biomimetic Drugs, Center for Molecular and Translational Medicine, Peking University, Beijing, China (F.W.)
| | - Huiyun Zhao
- From the Medical Isotopes Research Center and Department of Radiation Medicine, School of Basic Medical Sciences, Peking University Health Science Center, 38 Xueyuan Rd, Beijing 100191, China (X.Y., Y.W., H.L., L.G., S.X., C.Z., J.S., H.Z., B.J., Z.L., F.W.); Interdisciplinary Laboratory, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China (J.S., F.W.); and State Key Laboratory of Natural and Biomimetic Drugs, Center for Molecular and Translational Medicine, Peking University, Beijing, China (F.W.)
| | - Bing Jia
- From the Medical Isotopes Research Center and Department of Radiation Medicine, School of Basic Medical Sciences, Peking University Health Science Center, 38 Xueyuan Rd, Beijing 100191, China (X.Y., Y.W., H.L., L.G., S.X., C.Z., J.S., H.Z., B.J., Z.L., F.W.); Interdisciplinary Laboratory, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China (J.S., F.W.); and State Key Laboratory of Natural and Biomimetic Drugs, Center for Molecular and Translational Medicine, Peking University, Beijing, China (F.W.)
| | - Zhaofei Liu
- From the Medical Isotopes Research Center and Department of Radiation Medicine, School of Basic Medical Sciences, Peking University Health Science Center, 38 Xueyuan Rd, Beijing 100191, China (X.Y., Y.W., H.L., L.G., S.X., C.Z., J.S., H.Z., B.J., Z.L., F.W.); Interdisciplinary Laboratory, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China (J.S., F.W.); and State Key Laboratory of Natural and Biomimetic Drugs, Center for Molecular and Translational Medicine, Peking University, Beijing, China (F.W.)
| | - Fan Wang
- From the Medical Isotopes Research Center and Department of Radiation Medicine, School of Basic Medical Sciences, Peking University Health Science Center, 38 Xueyuan Rd, Beijing 100191, China (X.Y., Y.W., H.L., L.G., S.X., C.Z., J.S., H.Z., B.J., Z.L., F.W.); Interdisciplinary Laboratory, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China (J.S., F.W.); and State Key Laboratory of Natural and Biomimetic Drugs, Center for Molecular and Translational Medicine, Peking University, Beijing, China (F.W.)
| |
Collapse
|
71
|
Li D, He L, Guo H, Chen H, Shan H. Targeting activated hepatic stellate cells (aHSCs) for liver fibrosis imaging. EJNMMI Res 2015; 5:71. [PMID: 26650603 PMCID: PMC4674461 DOI: 10.1186/s13550-015-0151-x] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2015] [Accepted: 11/27/2015] [Indexed: 12/12/2022] Open
Abstract
Following injurious stimuli, quiescent hepatic stellate cells (qHSCs) transdifferentiate into activated HSCs (aHSCs). aHSCs play pivotal roles in the onset and progression of liver fibrosis. Therefore, molecular imaging of aHSCs in liver fibrosis will facilitate early diagnosis, prognosis prediction, and instruction and evaluation of aHSC-targeted treatment. To date, several receptors, such as integrin αvβ3, mannose 6-phosphate/insulin-like growth factor II receptor (M6P/IGF-IIR), collagen type VI receptor (CVIR), platelet-derived growth factor receptor-β (PDGFR-β), vimentin, and desmin, have been identified as biomarkers of aHSCs. Corresponding ligands to these receptors have also been developed. This review will discuss strategies for developing aHSC-targeted imaging in liver fibrosis.
Collapse
Affiliation(s)
- Dan Li
- Department of Radiology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, China.,Guangdong Provincial Engineering Research Center of Molecular Imaging, Guangzhou, 510630, China
| | - Li He
- Department of Radiology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, China
| | - Huizhuang Guo
- Department of Radiology, Guangzhou Panyu Central Hospital, Guangzhou, 511400, China
| | - Hanwei Chen
- Department of Radiology, Guangzhou Panyu Central Hospital, Guangzhou, 511400, China.
| | - Hong Shan
- Department of Radiology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, China. .,Guangdong Provincial Engineering Research Center of Molecular Imaging, Guangzhou, 510630, China. .,Interventional Radiology Institute of Sun Yat-sen University, Guangzhou, 510630, China.
| |
Collapse
|
72
|
Chen C, Li R, Ross RS, Manso AM. Integrins and integrin-related proteins in cardiac fibrosis. J Mol Cell Cardiol 2015; 93:162-74. [PMID: 26562414 DOI: 10.1016/j.yjmcc.2015.11.010] [Citation(s) in RCA: 113] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2015] [Revised: 11/07/2015] [Accepted: 11/07/2015] [Indexed: 12/21/2022]
Abstract
Cardiac fibrosis is one of the major components of the healing mechanism following any injury of the heart and as such may contribute to both systolic and diastolic dysfunction in a range of pathophysiologic conditions. Canonically, it can occur as part of the remodeling process that occurs following myocardial infarction or that follows as a response to pressure overload. Integrins are cell surface receptors which act in both cellular adhesion and signaling. Most importantly, in the context of the continuously contracting myocardium, they are recognized as mechanotransducers. They have been implicated in the development of fibrosis in several organs, including the heart. This review will focus on the involvement of integrins and integrin-related proteins, in cardiac fibrosis, outlining the roles of these proteins in the fibrotic responses in specific cardiac pathologies, discuss some of the common end effectors (angiotensin II, transforming growth factor beta 1 and mechanical stress) through which integrins function and finally discuss how manipulation of this set of proteins may lead to new treatments which could prove useful to alter the deleterious effects of cardiac fibrosis.
Collapse
Affiliation(s)
- Chao Chen
- Department of Medicine, Cardiology, UCSD School of Medicine, La Jolla, CA 92093-0613, USA; Veterans Administration San Diego Healthcare System, San Diego, CA 92161, USA.
| | - Ruixia Li
- Department of Medicine, Cardiology, UCSD School of Medicine, La Jolla, CA 92093-0613, USA; Veterans Administration San Diego Healthcare System, San Diego, CA 92161, USA.
| | - Robert S Ross
- Department of Medicine, Cardiology, UCSD School of Medicine, La Jolla, CA 92093-0613, USA; Veterans Administration San Diego Healthcare System, San Diego, CA 92161, USA.
| | - Ana Maria Manso
- Department of Medicine, Cardiology, UCSD School of Medicine, La Jolla, CA 92093-0613, USA; Veterans Administration San Diego Healthcare System, San Diego, CA 92161, USA.
| |
Collapse
|
73
|
Abstract
Significant progress has been made in understanding the principles underlying the development of liver fibrosis. This includes appreciating its dynamic nature, the importance of active fibrolysis in fibrosis regression, and the plasticity of cell populations endowing them with fibrogenic or fibrolytic properties. This is complemented by an increasing array of therapeutic targets with known roles in the progression or regression of fibrosis. With a key role for fibrosis in determining clinical outcomes and encouraging data from recently Food and Drug Administration-approved antifibrotics for pulmonary fibrosis, the development and validation of antifibrotic therapies has taken center stage in translational hepatology. In addition to summarizing the recent progress in antifibrotic therapies, the authors discuss some of the challenges ahead, such as achieving a better understanding of the interindividual heterogeneity of the fibrotic response, how to match interventions with the ideal patient population, and the development of better noninvasive methods to assess the dynamics of fibrogenesis and fibrolysis. Together, these advances will permit a better targeting and dose titration of individualized therapies. Finally, the authors discuss combination therapy with different antifibrotics as possibly the most potent approach for treating fibrosis in the liver.
Collapse
Affiliation(s)
- W. Z. Mehal
- Section of Digestive Diseases, Yale University, New Haven, Connecticut,West Haven Veterans Medical Center, West Haven, Connecticut
| | - D. Schuppan
- Department of Medicine, Institute of Translational Immunology and Research Center for Immunotherapy, University of Mainz Medical Center, Mainz, Germany,Division of Gastroenterology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
74
|
Hintermann E, Bayer M, Pfeilschifter JM, Deák F, Kiss I, Paulsson M, Christen U. Upregulation of matrilin-2 expression in murine hepatic stellate cells during liver injury has no effect on fibrosis formation and resolution. Liver Int 2015; 35:1265-73. [PMID: 24905825 DOI: 10.1111/liv.12604] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2014] [Accepted: 05/31/2014] [Indexed: 12/16/2022]
Abstract
BACKGROUND & AIMS Matrilins are a family of four oligomeric adaptor proteins whose functions in extracellular matrix assembly during pathophysiological events still need to be explored in more detail. Matrilin-2 is the largest family member and the only matrilin expressed in the naive liver. Several studies demonstrate that matrilin-2 interacts with collagen I, fibronectin or laminin-111-nidogen-1 complexes. All these matrix components get upregulated during hepatic scar tissue formation. Therefore, we tested whether matrilin-2 has an influence on the formation and/or the resolution of fibrotic tissue in the mouse liver. METHODS Fibrosis was induced by infection with an adenovirus encoding cytochrome P450 2D6 (autoimmune liver damage) or by exposure to the hepatotoxin carbon tetrachloride. Fibrosis severity and matrilin-2 expression were assessed by immunohistochemistry. Hepatic stellate cells (HSCs) were isolated and analysed by immunocytochemistry and Transwell migration assays. RESULTS Both autoimmune as well as chemically induced liver damage led to simultaneous upregulation of matrilin-2 and collagen I expression. Discontinuation of carbon tetrachloride exposure resulted in concomitant dissolution of both proteins. Activated HSCs were the source of de novo matrilin-2 expression. Comparing wild type and matrilin-2-deficient mice, no differences were detected in fibronectin and collagen I upregulation and resolution kinetics as well as amount or location of fibronectin and collagen I production and degradation. CONCLUSIONS Our findings suggest that the absence of matrilin-2 has no effect on HSC activation and regression kinetics, synthetic activity, proliferative capacity, motility, or HSC apoptosis.
Collapse
Affiliation(s)
- Edith Hintermann
- Pharmazentrum Frankfurt / ZAFES, Johann Wolfgang Goethe University, Frankfurt am Main, Germany
| | | | | | | | | | | | | |
Collapse
|
75
|
Recent advances in molecular magnetic resonance imaging of liver fibrosis. BIOMED RESEARCH INTERNATIONAL 2015; 2015:595467. [PMID: 25874221 PMCID: PMC4385649 DOI: 10.1155/2015/595467] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/30/2014] [Accepted: 09/03/2014] [Indexed: 12/19/2022]
Abstract
Liver fibrosis is a life-threatening disease with high morbidity and mortality owing to its diverse causes. Liver biopsy, as the current gold standard for diagnosing and staging liver fibrosis, has a number of limitations, including sample variability, relatively high cost, an invasive nature, and the potential of complications. Most importantly, in clinical practice, patients often reject additional liver biopsies after initiating treatment despite their being necessary for long-term follow-up. To resolve these problems, a number of different noninvasive imaging-based methods have been developed for accurate diagnosis of liver fibrosis. However, these techniques only reflect morphological or perfusion-related alterations in the liver, and thus they are generally only useful for the diagnosis of late-stage liver fibrosis (liver cirrhosis), which is already characterized by "irreversible" anatomic and hemodynamic changes. Thus, it is essential that new approaches are developed for accurately diagnosing early-stage liver fibrosis as at this stage the disease may be "reversed" by active treatment. The development of molecular MR imaging technology has potential in this regard, as it facilitates noninvasive, target-specific imaging of liver fibrosis. We provide an overview of recent advances in molecular MR imaging for the diagnosis and staging of liver fibrosis and we compare novel technologies with conventional MR imaging techniques.
Collapse
|
76
|
Matsue Y, Tsutsumi M, Hayashi N, Saito T, Tsuchishima M, Toshikuni N, Arisawa T, George J. Serum osteopontin predicts degree of hepatic fibrosis and serves as a biomarker in patients with hepatitis C virus infection. PLoS One 2015; 10:e0118744. [PMID: 25760884 PMCID: PMC4356538 DOI: 10.1371/journal.pone.0118744] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2014] [Accepted: 01/12/2015] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND & AIMS Osteopontin (OPN) is a matricellular protein that upregulates during pathogenesis of hepatic fibrosis. The present study was aimed to evaluate whether serum OPN could be used as a biomarker to assess the degree of hepatic fibrosis in patients with hepatitis C virus (HCV) infection. METHODS Needle biopsy was performed on HCV patients and scored as zero fibrosis (F0), mild fibrosis (F1), moderate fibrosis (F2), severe fibrosis (F3) and liver cirrhosis (F4) based on Masson's trichrome and α-smooth muscle actin (α-SMA) staining. Serum OPN levels were measured using ELISA and correlated with the degree of fibrosis. Furthermore, the OPN values were correlated and evaluated with platelets count, serum hyaluronic acid (HA), and collagen type IV and subjected to receiver operating characteristic (ROC) curve analysis. RESULTS Serum OPN levels were remarkably increased from F0 through F4 in a progressive manner and the differences were significant (P < 0.001) between each group. The data were highly correlated with the degree of hepatic fibrosis. The ROC curve analysis depicted that serum OPN is an independent risk factor and an excellent biomarker and a prognostic index in HCV patients. CONCLUSIONS The results of the present study indicate that serum OPN levels reflect the degree of hepatic fibrosis and could be used as a biomarker to assess the stage of fibrosis in HCV patients which would help to reduce the number of liver biopsies. Furthermore, serum OPN serves as a prognostic index towards the progression of hepatic fibrosis to cirrhosis and hepatocellular carcinoma.
Collapse
Affiliation(s)
- Yasuhiro Matsue
- Department of Hepatology, Kanazawa Medical University, Uchinada, Ishikawa, Japan
| | - Mikihiro Tsutsumi
- Department of Hepatology, Kanazawa Medical University, Uchinada, Ishikawa, Japan
| | - Nobuhiko Hayashi
- Department of Hepatology, Kanazawa Medical University, Uchinada, Ishikawa, Japan
| | - Takashi Saito
- Department of Hepatology, Kanazawa Medical University, Uchinada, Ishikawa, Japan
| | - Mutsumi Tsuchishima
- Department of Hepatology, Kanazawa Medical University, Uchinada, Ishikawa, Japan
| | - Nobuyuki Toshikuni
- Department of Hepatology, Kanazawa Medical University, Uchinada, Ishikawa, Japan
| | - Tomiyasu Arisawa
- Department of Gastroenterology, Kanazawa Medical University, Uchinada, Ishikawa, Japan
| | - Joseph George
- Department of Hepatology, Kanazawa Medical University, Uchinada, Ishikawa, Japan
- * E-mail:
| |
Collapse
|
77
|
Dash A, Chakraborty S, Pillai MRA, Knapp FFR. Peptide receptor radionuclide therapy: an overview. Cancer Biother Radiopharm 2015; 30:47-71. [PMID: 25710506 DOI: 10.1089/cbr.2014.1741] [Citation(s) in RCA: 77] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Peptide receptor radionuclide therapy (PRRT) is a site-directed targeted therapeutic strategy that specifically uses radiolabeled peptides as biological targeting vectors designed to deliver cytotoxic levels of radiation dose to cancer cells, which overexpress specific receptors. Interest in PRRT has steadily grown because of the advantages of targeting cellular receptors in vivo with high sensitivity as well as specificity and treatment at the molecular level. Recent advances in molecular biology have not only stimulated advances in PRRT in a sustainable manner but have also pushed the field significantly forward to several unexplored possibilities. Recent decades have witnessed unprecedented endeavors for developing radiolabeled receptor-binding somatostatin analogs for the treatment of neuroendocrine tumors, which have played an important role in the evolution of PRRT and paved the way for the development of other receptor-targeting peptides. Several peptides targeting a variety of receptors have been identified, demonstrating their potential to catalyze breakthroughs in PRRT. In this review, the authors discuss several of these peptides and their analogs with regard to their applications and potential in radionuclide therapy. The advancement in the availability of combinatorial peptide libraries for peptide designing and screening provides the capability of regulating immunogenicity and chemical manipulability. Moreover, the availability of a wide range of bifunctional chelating agents opens up the scope of convenient radiolabeling. For these reasons, it would be possible to envision a future where the scope of PRRT can be tailored for patient-specific application. While PRRT lies at the interface between many disciplines, this technology is inextricably linked to the availability of the therapeutic radionuclides of required quality and activity levels and hence their production is also reviewed.
Collapse
Affiliation(s)
- Ashutosh Dash
- 1 Isotope Production and Applications Division, Bhabha Atomic Research Centre , Mumbai, India
| | | | | | | |
Collapse
|
78
|
Mòdol T, Brice N, Ruiz de Galarreta M, García Garzón A, Iraburu MJ, Martínez-Irujo JJ, López-Zabalza MJ. Fibronectin peptides as potential regulators of hepatic fibrosis through apoptosis of hepatic stellate cells. J Cell Physiol 2015; 230:546-53. [PMID: 24976518 DOI: 10.1002/jcp.24714] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2014] [Revised: 05/26/2014] [Accepted: 06/24/2014] [Indexed: 12/23/2022]
Abstract
The turnover of extracellular matrix (ECM) components can generate signals that regulate several cellular functions such as proliferation, differentiation, and apoptosis. During liver injury, matrix metalloproteases (MMPs) production is enhanced and increased levels of peptides derived from extracellular matrix proteins can be generated. Synthetic peptides with sequences present in extracellular matrix proteins were previously found to induce both stimulating and apoptotic effects on several cell types including the inflammatory cells monocytes/macrophages. Therefore, in inflammatory liver diseases, locally accumulated peptides could be also important in regulating hepatic fibrosis by inducing apoptosis of hepatic stellate cells (HSC), the primary cellular source of extracellular matrix components. Here, we describe the apoptotic effect of fibronectin peptides on the cell line of human hepatic stellate cells LX-2 based on oligonucleosomal DNA fragmentation, caspase-3 and -9 activation, Bcl-2 depletion, and accumulation of Bax protein. We also found that these peptides trigger the activation of Src kinase, which in turn mediated the increase of JNK and p38 activities. By the use of specific inhibitors we demonstrated the involvement of Src, JNK, and p38 in apoptosis induced by fibronectin peptides on HSC. Moreover, fibronectin peptides increased iNOS expression in human HSC, and specific inhibition of iNOS significantly reduced the sustained activity of JNK and the programmed cell death caused by these peptides. Finally, the possible regulatory effect of fibronectin peptides in liver fibrosis was further supported by the ability of these peptides to induce metalloprotease-9 (MMP-9) expression in human monocytes.
Collapse
Affiliation(s)
- Teresa Mòdol
- Departamento de Bioquímica y Genética, Universidad de Navarra, Pamplona, Spain
| | | | | | | | | | | | | |
Collapse
|
79
|
Duarte S, Baber J, Fujii T, Coito AJ. Matrix metalloproteinases in liver injury, repair and fibrosis. Matrix Biol 2015; 44-46:147-56. [PMID: 25599939 PMCID: PMC4495728 DOI: 10.1016/j.matbio.2015.01.004] [Citation(s) in RCA: 324] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2014] [Revised: 01/09/2015] [Accepted: 01/10/2015] [Indexed: 01/18/2023]
Abstract
The liver is a large highly vascularized organ with a central function in metabolic homeostasis, detoxification, and immunity. Due to its roles, the liver is frequently exposed to various insults which can cause cell death and hepatic dysfunction. Alternatively, the liver has a remarkable ability to self-repair and regenerate after injury. Liver injury and regeneration have both been linked to complex extracellular matrix (ECM) related pathways. While normal degradation of ECM components is an important feature of tissue repair and remodeling, irregular ECM turnover contributes to a variety of liver diseases. Matrix metalloproteinases (MMPs) are the main enzymes implicated in ECM degradation. MMPs not only remodel the ECM, but also regulate immune responses. In this review, we highlight some of the MMP-attributed roles in acute and chronic liver injury and emphasize the need for further experimentation to better understand their functions during hepatic physiological conditions and disease progression.
Collapse
Affiliation(s)
- Sergio Duarte
- The Dumont-UCLA Transplant Center, Division of Liver and Pancreas Transplantation, Department of Surgery, David Geffen School of Medicine at UCLA, Los Angeles, CA, United States
| | - John Baber
- The Dumont-UCLA Transplant Center, Division of Liver and Pancreas Transplantation, Department of Surgery, David Geffen School of Medicine at UCLA, Los Angeles, CA, United States
| | - Takehiro Fujii
- The Dumont-UCLA Transplant Center, Division of Liver and Pancreas Transplantation, Department of Surgery, David Geffen School of Medicine at UCLA, Los Angeles, CA, United States
| | - Ana J Coito
- The Dumont-UCLA Transplant Center, Division of Liver and Pancreas Transplantation, Department of Surgery, David Geffen School of Medicine at UCLA, Los Angeles, CA, United States.
| |
Collapse
|
80
|
Ramezani-Moghadam M, Wang J, Ho V, Iseli TJ, Alzahrani B, Xu A, Van der Poorten D, Qiao L, George J, Hebbard L. Adiponectin reduces hepatic stellate cell migration by promoting tissue inhibitor of metalloproteinase-1 (TIMP-1) secretion. J Biol Chem 2015; 290:5533-42. [PMID: 25575598 DOI: 10.1074/jbc.m114.598011] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Hepatic stellate cells (HSC) are central players in liver fibrosis that when activated, proliferate, migrate to sites of liver injury, and secrete extracellular matrix. Obesity, a known risk factor for liver fibrosis is associated with reduced levels of adiponectin, a protein that inhibits liver fibrosis in vivo and limits HSC proliferation and migration in vitro. Adiponectin-mediated activation of adenosine monophosphate-activated kinase (AMPK) inhibits HSC proliferation, but the mechanism by which it limits HSC migration to sites of injury is unknown. Here we sought to elucidate how adiponectin regulates HSC motility. Primary rat HSCs were isolated and treated with adiponectin in migration assays. The in vivo actions of adiponectin were examined by treating mice with carbon tetrachloride for 12 weeks and then injecting them with adiponectin. Cell and tissue samples were collected and analyzed for gene expression, signaling, and histology. Serum from patients with liver fibrosis was examined for adiponectin and tissue inhibitor of metalloproteinase-1 (TIMP-1) protein. Adiponectin administration into mice increased TIMP-1 gene and protein expression. In cultured HSCs, adiponectin promoted TIMP-1 expression and through binding of TIMP-1 to the CD63/β1-integrin complex reduced phosphorylation of focal adhesion kinase to limit HSC migration. In mice with liver fibrosis, adiponectin had similar effects and limited focal adhesion kinase phosphorylation. Finally, in patients with advanced fibrosis, there was a positive correlation between serum adiponectin and TIMP-1 levels. In sum, these data show that adiponectin stimulates TIMP-1 secretion by HSCs to retard their migration and contributes to the anti-fibrotic effects of adiponectin.
Collapse
Affiliation(s)
- Mehdi Ramezani-Moghadam
- From the Storr Liver Centre, Westmead Millennium Institute, University of Sydney at Westmead Hospital, Westmead, New South Wales 2145, Australia and
| | - Jianhua Wang
- From the Storr Liver Centre, Westmead Millennium Institute, University of Sydney at Westmead Hospital, Westmead, New South Wales 2145, Australia and
| | - Vikki Ho
- From the Storr Liver Centre, Westmead Millennium Institute, University of Sydney at Westmead Hospital, Westmead, New South Wales 2145, Australia and
| | - Tristan J Iseli
- From the Storr Liver Centre, Westmead Millennium Institute, University of Sydney at Westmead Hospital, Westmead, New South Wales 2145, Australia and
| | - Badr Alzahrani
- From the Storr Liver Centre, Westmead Millennium Institute, University of Sydney at Westmead Hospital, Westmead, New South Wales 2145, Australia and
| | - Aimin Xu
- the Department of Medicine, the University of Hong Kong, Hong Kong, China
| | - David Van der Poorten
- From the Storr Liver Centre, Westmead Millennium Institute, University of Sydney at Westmead Hospital, Westmead, New South Wales 2145, Australia and
| | - Liang Qiao
- From the Storr Liver Centre, Westmead Millennium Institute, University of Sydney at Westmead Hospital, Westmead, New South Wales 2145, Australia and
| | - Jacob George
- From the Storr Liver Centre, Westmead Millennium Institute, University of Sydney at Westmead Hospital, Westmead, New South Wales 2145, Australia and
| | - Lionel Hebbard
- From the Storr Liver Centre, Westmead Millennium Institute, University of Sydney at Westmead Hospital, Westmead, New South Wales 2145, Australia and
| |
Collapse
|
81
|
Bartneck M, Warzecha KT, Tacke F. Therapeutic targeting of liver inflammation and fibrosis by nanomedicine. Hepatobiliary Surg Nutr 2015; 3:364-76. [PMID: 25568860 DOI: 10.3978/j.issn.2304-3881.2014.11.02] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2014] [Accepted: 10/17/2014] [Indexed: 12/25/2022]
Abstract
Nanomedicine constitutes the emerging field of medical applications for nanotechnology such as nanomaterial-based drug delivery systems. This technology may hold exceptional potential for novel therapeutic approaches to liver diseases. The specific and unspecific targeting of macrophages, hepatic stellate cells (HSC), hepatocytes, and liver sinusoidal endothelial cells (LSEC) using nanomedicine has been developed and tested in preclinical settings. These four major cell types in the liver are crucially involved in the complex sequence of events that occurs during the initiation and maintenance of liver inflammation and fibrosis. Targeting different cell types can be based on their capacity to ingest surrounding material, endocytosis, and specificity for a single cell type can be achieved by targeting characteristic structures such as receptors, sugar moieties or peptide sequences. Macrophages and especially the liver-resident Kupffer cells are in the focus of nanomedicine due to their highly efficient and unspecific uptake of most nanomaterials as well as due to their critical pathogenic functions during inflammation and fibrogenesis. The mannose receptor enables targeting macrophages in liver disease, but macrophages can also become activated by certain nanomaterials, such as peptide-modified gold nanorods (AuNRs) that render them proinflammatory. HSC, the main collagen-producing cells during fibrosis, are currently targeted using nanoconstructs that recognize the mannose 6-phosphate and insulin-like growth factor II, peroxisome proliferator activated receptor 1, platelet-derived growth factor (PDGF) receptor β, or integrins. Targeting of the major liver parenchymal cell, the hepatocyte, has only recently been achieved with high specificity by mimicking apolipoproteins, naturally occurring nanoparticles of the body. LSEC were found to be targeted most efficiently using carboxy-modified micelles and their integrin receptors. This review will summarize important functions of these cell types in healthy and diseased livers and discuss current strategies of cell-specific targeting for liver diseases by nanomedicine.
Collapse
Affiliation(s)
- Matthias Bartneck
- Department of Medicine III, University Hospital Aachen, 52074 Aachen, Germany
| | | | - Frank Tacke
- Department of Medicine III, University Hospital Aachen, 52074 Aachen, Germany
| |
Collapse
|
82
|
Montaldo C, Mattei S, Baiocchini A, Rotiroti N, Del Nonno F, Pucillo LP, Cozzolino AM, Battistelli C, Amicone L, Ippolito G, van Noort V, Conigliaro A, Alonzi T, Tripodi M, Mancone C. Spike-in SILAC proteomic approach reveals the vitronectin as an early molecular signature of liver fibrosis in hepatitis C infections with hepatic iron overload. Proteomics 2014; 14:1107-15. [PMID: 24616218 DOI: 10.1002/pmic.201300422] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2013] [Revised: 01/08/2014] [Accepted: 01/09/2014] [Indexed: 01/06/2023]
Abstract
Hepatitis C virus (HCV)-induced iron overload has been shown to promote liver fibrosis, steatosis, and hepatocellular carcinoma. The zonal-restricted histological distribution of pathological iron deposits has hampered the attempt to perform large-scale in vivo molecular investigations on the comorbidity between iron and HCV. Diagnostic and prognostic markers are not yet available to assess iron overload-induced liver fibrogenesis and progression in HCV infections. Here, by means of Spike-in SILAC proteomic approach, we first unveiled a specific membrane protein expression signature of HCV cell cultures in the presence of iron overload. Computational analysis of proteomic dataset highlighted the hepatocytic vitronectin expression as the most promising specific biomarker for iron-associated fibrogenesis in HCV infections. Next, the robustness of our in vitro findings was challenged in human liver biopsies by immunohistochemistry and yielded two major results: (i) hepatocytic vitronectin expression is associated to liver fibrogenesis in HCV-infected patients with iron overload; (ii) hepatic vitronectin expression was found to discriminate also the transition between mild to moderate fibrosis in HCV-infected patients without iron overload.
Collapse
Affiliation(s)
- Claudia Montaldo
- Department of Cellular Biotechnologies and Haematology, Istituto Pasteur-Fondazione Cenci Bolognetti, Sapienza University of Rome, Italy; "L. Spallanzani" National Institute for Infectious Diseases, IRCCS, Rome, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
83
|
Zhang X, Xin J, Shi Y, Xu W, Yu S, Yang Z, Liu C, Cao L, Guo Q. Assessing activation of hepatic stellate cells by (99m)Tc-3PRGD2 scintigraphy targeting integrin αvβ3: a feasibility study. Nucl Med Biol 2014; 42:250-5. [PMID: 25530210 DOI: 10.1016/j.nucmedbio.2014.11.007] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2014] [Accepted: 11/19/2014] [Indexed: 11/25/2022]
Abstract
OBJECTIVE Hepatic stellate cell (HSC) activation, which is accompanied by increased expression of integrin αvβ3, is an important factor in liver fibrogenesis. Molecular imaging targeting the integrin αvβ3 could provide a non-invasive method for evaluating the expression and the function of the integrin αvβ3 on the activated HSCs (aHSCs) in the injured liver, and then provide important prognostic information. (99m)Tc-3PRGD2 is such a radiotracer specific for integrin αvβ3. In this study, we aimed to compare the differences in liver uptake and retention of the (99m)Tc-3PRGD2 between normal liver and injured liver to evaluate the feasibility of (99m)Tc-3PRGD2 scintigraphy for this purpose. METHODS We used planar scintigraphy to assess changes in integrin αvβ3 binding of intravenously-administered (99m)Tc-3PRGD2 in the livers of rats with thioacetamide (TAA)-induced liver fibrosis compared with the controls. We co-injected cold c(RGDyK) with (99m)Tc-3PRGD2 to assess the specific binding of the radiotracer. We performed Sirius red staining to assess liver fibrosis, immunofluorescent colocalization to identify the location of integrin αvβ3 expressed in the fibrotic liver, and we measured protein and messenger RNA expression of integrin αvβ3 and alpha smooth muscle actin (α-SMA) in the control and fibrotic livers. RESULTS The fibrotic livers showed enhanced (99m)Tc-3PRGD2 uptake and retention. The radiotracer was demonstrated to bind specifically with the integrin αvβ3 mainly expressed on the aHSCs. The liver-to-heart ratio at 30 min post-injection was higher in the fibrotic livers than in the control livers (TAA, 1.98±0.08 vs. control, 1.50±0.12, p<0.01). The liver t1/2 was longer than in the controls (TAA, 27.07±10.69 min vs. control, 12.67±4.10 min, p<0.01). The difference of heart t1/2 between the two groups was not statistically significant (TAA, 3.13±0.63 min vs. control, 3.41±0.77 min, p=0.94). CONCLUSIONS (99m)Tc-3PRGD2 molecular imaging can provide a non-invasive method for assessing activation of HSCs.
Collapse
Affiliation(s)
- Xin Zhang
- Department of Radiology and Nuclear Medicine, Shengjing Hospital of China Medical University, Shenyang, China.
| | - Jun Xin
- Department of Radiology and Nuclear Medicine, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yu Shi
- Department of Radiology and Nuclear Medicine, Shengjing Hospital of China Medical University, Shenyang, China
| | - Weina Xu
- Department of Radiology and Nuclear Medicine, Shengjing Hospital of China Medical University, Shenyang, China
| | - Shupeng Yu
- Department of Radiology and Nuclear Medicine, Shengjing Hospital of China Medical University, Shenyang, China
| | - Zhiguang Yang
- Department of Radiology and Nuclear Medicine, Shengjing Hospital of China Medical University, Shenyang, China
| | - Changping Liu
- Department of Radiology and Nuclear Medicine, Shengjing Hospital of China Medical University, Shenyang, China
| | - Li Cao
- Department of Radiology and Nuclear Medicine, Shengjing Hospital of China Medical University, Shenyang, China
| | - Qiyong Guo
- Department of Radiology and Nuclear Medicine, Shengjing Hospital of China Medical University, Shenyang, China.
| |
Collapse
|
84
|
Davis PJ, Lin HY, Sudha T, Yalcin M, Tang HY, Hercbergs A, Leith JT, Luidens MK, Ashur-Fabian O, Incerpi S, Mousa SA. Nanotetrac targets integrin αvβ3 on tumor cells to disorder cell defense pathways and block angiogenesis. Onco Targets Ther 2014; 7:1619-24. [PMID: 25258542 PMCID: PMC4172128 DOI: 10.2147/ott.s67393] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
The extracellular domain of integrin αvβ3 contains a receptor for thyroid hormone and hormone analogs. The integrin is amply expressed by tumor cells and dividing blood vessel cells. The proangiogenic properties of thyroid hormone and the capacity of the hormone to promote cancer cell proliferation are functions regulated nongenomically by the hormone receptor on αvβ3. An L-thyroxine (T4) analog, tetraiodothyroacetic acid (tetrac), blocks binding of T4 and 3,5,3'-triiodo-L-thyronine (T3) by αvβ3 and inhibits angiogenic activity of thyroid hormone. Covalently bound to a 200 nm nanoparticle that limits its activity to the cell exterior, tetrac reformulated as Nanotetrac has additional effects mediated by αvβ3 beyond the inhibition of binding of T4 and T3 to the integrin. These actions of Nanotetrac include disruption of transcription of cell survival pathway genes, promotion of apoptosis by multiple mechanisms, and interruption of repair of double-strand deoxyribonucleic acid breaks caused by irradiation of cells. Among the genes whose expression is suppressed by Nanotetrac are EGFR, VEGFA, multiple cyclins, catenins, and multiple cytokines. Nanotetrac has been effective as a chemotherapeutic agent in preclinical studies of human cancer xenografts. The low concentrations of αvβ3 on the surface of quiescent nonmalignant cells have minimized toxicity of the agent in animal studies.
Collapse
Affiliation(s)
- Paul J Davis
- Department of Medicine, Albany Medical College, Albany, NY, USA ; Pharmaceutical Research Institute, Albany College of Pharmacy and Health Sciences, Albany, NY, USA
| | - Hung-Yun Lin
- Pharmaceutical Research Institute, Albany College of Pharmacy and Health Sciences, Albany, NY, USA ; PhD Program for Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Thangirala Sudha
- Pharmaceutical Research Institute, Albany College of Pharmacy and Health Sciences, Albany, NY, USA
| | - Murat Yalcin
- Pharmaceutical Research Institute, Albany College of Pharmacy and Health Sciences, Albany, NY, USA ; Department of Physiology, Veterinary Medicine Faculty, Uludag University, Gorukle, Bursa, Turkey
| | - Heng-Yuan Tang
- Pharmaceutical Research Institute, Albany College of Pharmacy and Health Sciences, Albany, NY, USA
| | | | - John T Leith
- Rhode Island Nuclear Science Center, Narragansett, RI, USA
| | - Mary K Luidens
- Department of Medicine, Albany Medical College, Albany, NY, USA
| | - Osnat Ashur-Fabian
- Translational Hemato-oncology Laboratory, Hematology Institute and Blood Bank, Meir Medical Center, Kfar-Saba, Israel ; Department of Human Molecular Genetics and Biochemistry, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Sandra Incerpi
- Department of Sciences, University of Roma Tre, Rome, Italy
| | - Shaker A Mousa
- Pharmaceutical Research Institute, Albany College of Pharmacy and Health Sciences, Albany, NY, USA
| |
Collapse
|
85
|
Campana L, Iredale JP. Extracellular Matrix Metabolism and Fibrotic Disease. CURRENT PATHOBIOLOGY REPORTS 2014. [DOI: 10.1007/s40139-014-0058-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
86
|
Bi Y, Mukhopadhyay D, Drinane M, Ji B, Li X, Cao S, Shah VH. Endocytosis of collagen by hepatic stellate cells regulates extracellular matrix dynamics. Am J Physiol Cell Physiol 2014; 307:C622-33. [PMID: 25080486 DOI: 10.1152/ajpcell.00086.2014] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Hepatic stellate cells (HSCs) generate matrix, which in turn may also regulate HSCs function during liver fibrosis. We hypothesized that HSCs may endocytose matrix proteins to sense and respond to changes in microenvironment. Primary human HSCs, LX2, or mouse embryonic fibroblasts (MEFs) [wild-type; c-abl(-/-); or Yes, Src, and Fyn knockout mice (YSF(-/-))] were incubated with fluorescent-labeled collagen or gelatin. Fluorescence-activated cell sorting analysis and confocal microscopy were used for measuring cellular internalization of matrix proteins. Targeted PCR array and quantitative real-time PCR were used to evaluate gene expression changes. HSCs and LX2 cells endocytose collagens in a concentration- and time-dependent manner. Endocytosed collagen colocalized with Dextran 10K, a marker of macropinocytosis, and 5-ethylisopropyl amiloride, an inhibitor of macropinocytosis, reduced collagen internalization by 46%. Cytochalasin D and ML7 blocked collagen internalization by 47% and 45%, respectively, indicating that actin and myosin are critical for collagen endocytosis. Wortmannin and AKT inhibitor blocked collagen internalization by 70% and 89%, respectively, indicating that matrix macropinocytosis requires phosphoinositide-3-kinase (PI3K)/AKT signaling. Overexpression of dominant-negative dynamin-2 K44A blocked matrix internalization by 77%, indicating a role for dynamin-2 in matrix macropinocytosis. Whereas c-abl(-/-) MEF showed impaired matrix endocytosis, YSF(-/-) MEF surprisingly showed increased matrix endocytosis. It was also associated with complex gene regulations that related with matrix dynamics, including increased matrix metalloproteinase 9 (MMP-9) mRNA levels and zymographic activity. HSCs endocytose matrix proteins through macropinocytosis that requires a signaling network composed of PI3K/AKT, dynamin-2, and c-abl. Interaction with extracellular matrix regulates matrix dynamics through modulating multiple gene expressions including MMP-9.
Collapse
Affiliation(s)
- Yan Bi
- GI Research Unit, Department of Gastroenterology and Hepatology
| | | | - Mary Drinane
- GI Research Unit, Department of Gastroenterology and Hepatology
| | - Baoan Ji
- Department of Biochemistry and Molecular Biology
| | - Xing Li
- Department of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, Minneosota
| | - Sheng Cao
- GI Research Unit, Department of Gastroenterology and Hepatology
| | - Vijay H Shah
- GI Research Unit, Department of Gastroenterology and Hepatology,
| |
Collapse
|
87
|
Giannitrapani L, Soresi M, Bondì ML, Montalto G, Cervello M. Nanotechnology applications for the therapy of liver fibrosis. World J Gastroenterol 2014; 20:7242-7251. [PMID: 24966595 PMCID: PMC4064070 DOI: 10.3748/wjg.v20.i23.7242] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2013] [Accepted: 03/06/2014] [Indexed: 02/06/2023] Open
Abstract
Chronic liver diseases represent a major global health problem both for their high prevalence worldwide and, in the more advanced stages, for the limited available curative treatment options. In fact, when lesions of different etiologies chronically affect the liver, triggering the fibrogenesis mechanisms, damage has already occurred and the progression of fibrosis will have a major clinical impact entailing severe complications, expensive treatments and death in end-stage liver disease. Despite significant advances in the understanding of the mechanisms of liver fibrinogenesis, the drugs used in liver fibrosis treatment still have a limited therapeutic effect. Many drugs showing potent antifibrotic activities in vitro often exhibit only minor effects in vivo because insufficient concentrations accumulate around the target cell and adverse effects result as other non-target cells are affected. Hepatic stellate cells play a critical role in liver fibrogenesis , thus they are the target cells of antifibrotic therapy. The application of nanoparticles has emerged as a rapidly evolving area for the safe delivery of various therapeutic agents (including drugs and nucleic acid) in the treatment of various pathologies, including liver disease. In this review, we give an overview of the various nanotechnology approaches used in the treatment of liver fibrosis.
Collapse
|
88
|
Birukawa NK, Murase K, Sato Y, Kosaka A, Yoneda A, Nishita H, Fujita R, Nishimura M, Ninomiya T, Kajiwara K, Miyazaki M, Nakashima Y, Ota S, Murakami Y, Tanaka Y, Minomi K, Tamura Y, Niitsu Y. Activated hepatic stellate cells are dependent on self-collagen, cleaved by membrane type 1 matrix metalloproteinase for their growth. J Biol Chem 2014; 289:20209-21. [PMID: 24867951 DOI: 10.1074/jbc.m113.544494] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Stellate cells are distributed throughout organs, where, upon chronic damage, they become activated and proliferate to secrete collagen, which results in organ fibrosis. An intriguing property of hepatic stellate cells (HSCs) is that they undergo apoptosis when collagen is resolved by stopping tissue damage or by treatment, even though the mechanisms are unknown. Here we disclose the fact that HSCs, normal diploid cells, acquired dependence on collagen for their growth during the transition from quiescent to active states. The intramolecular RGD motifs of collagen were exposed by cleavage with their own membrane type 1 matrix metalloproteinase (MT1-MMP). The following evidence supports this conclusion. When rat activated HSCs (aHSCs) were transduced with siRNA against the collagen-specific chaperone gp46 to inhibit collagen secretion, the cells underwent autophagy followed by apoptosis. Concomitantly, the growth of aHSCs was suppressed, whereas that of quiescent HSCs was not. These in vitro results are compatible with the in vivo observation that apoptosis of aHSCs was induced in cirrhotic livers of rats treated with siRNAgp46. siRNA against MT1-MMP and addition of tissue inhibitor of metalloproteinase 2 (TIMP-2), which mainly inhibits MT1-MMP, also significantly suppressed the growth of aHSCs in vitro. The RGD inhibitors echistatin and GRGDS peptide and siRNA against the RGD receptor αVβ1 resulted in the inhibition of aHSCs growth. Transduction of siRNAs against gp46, αVβ1, and MT1-MMP to aHSCs inhibited the survival signal of PI3K/AKT/IκB. These results could provide novel antifibrosis strategies.
Collapse
Affiliation(s)
| | | | | | - Akemi Kosaka
- From the Department of Molecular Target Exploration
| | | | | | | | | | - Takafumi Ninomiya
- Department of Basic Medical Science Department of Anatomy (1), Sapporo Medical University School of Medicine, 060-8556 Sapporo, Japan
| | - Keiko Kajiwara
- From the Department of Molecular Target Exploration, the Translational Research Group, Hokkaido Laboratory, Molecular Therapeutics Department, Corporate Business Development Division, Nitto Denko Corporation, Hokkaido, 001-0021 Sapporo, Japan, and
| | - Miyono Miyazaki
- From the Department of Molecular Target Exploration, the Translational Research Group, Hokkaido Laboratory, Molecular Therapeutics Department, Corporate Business Development Division, Nitto Denko Corporation, Hokkaido, 001-0021 Sapporo, Japan, and
| | | | - Sigenori Ota
- From the Department of Molecular Target Exploration
| | | | - Yasunobu Tanaka
- From the Department of Molecular Target Exploration, the Translational Research Group, Hokkaido Laboratory, Molecular Therapeutics Department, Corporate Business Development Division, Nitto Denko Corporation, Hokkaido, 001-0021 Sapporo, Japan, and
| | - Kenjiro Minomi
- From the Department of Molecular Target Exploration, the Translational Research Group, Hokkaido Laboratory, Molecular Therapeutics Department, Corporate Business Development Division, Nitto Denko Corporation, Hokkaido, 001-0021 Sapporo, Japan, and
| | - Yasuaki Tamura
- the Faculty of Advanced Life Science, Hokkaido University, 001-0021 Sapporo, Japan
| | | |
Collapse
|
89
|
Bittnerová L, Jiroutová A, Rudolf E, Rezácová M, Kanta J. Effect of collagen I gel on apoptosis of rat hepatic stellate cells. ACTA MEDICA (HRADEC KRÁLOVÉ) 2013; 56:73-9. [PMID: 24069661 DOI: 10.14712/18059694.2014.27] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
Activated hepatic stellate cells (HSC) are a major source offibrous proteins in cirrhotic liver. Inducing or accelerating their apoptosis is a potential way of liver fibrosis treatment. Extracellular matrix (ECM) surrounding cells in tissue affects their differentiation, migration, proliferation and function. Type I collagen is the main ECM component in fibrotic liver. We have examined how this protein modifies apoptosis of normal rat HSC induced by gliotoxin, cycloheximide and cytochalasin D in vitro and spontaneous apoptosis of HSC isolated from CCl4-damaged liver. We have found that type I collagen gel enhances HSC apoptosis regardless of the agent triggering this process.
Collapse
Affiliation(s)
- Lenka Bittnerová
- Department of Medical Biochemistry, Charles University in Prague, Faculty of Medicine, Hradec Králové, Czech Republic
| | | | | | | | | |
Collapse
|
90
|
Henderson NC, Sheppard D. Integrin-mediated regulation of TGFβ in fibrosis. BIOCHIMICA ET BIOPHYSICA ACTA 2013; 1832:891-6. [PMID: 23046811 PMCID: PMC3573259 DOI: 10.1016/j.bbadis.2012.10.005] [Citation(s) in RCA: 121] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/09/2012] [Revised: 10/01/2012] [Accepted: 10/03/2012] [Indexed: 12/26/2022]
Abstract
Fibrosis is a major cause of morbidity and mortality worldwide. Currently, therapeutic options for tissue fibrosis are severely limited, and organ transplantation is the only effective treatment for end-stage fibrotic disease. However, demand for donor organs greatly outstrips supply, and so effective anti-fibrotic treatments are urgently required. In recent years, the integrin family of cell adhesion receptors has gained prominence as key regulators of chronic inflammation and fibrosis. Fibrosis models in multiple organs have demonstrated that integrins have profound effects on the fibrotic process. There is now abundant in vivo data demonstrating critical regulatory roles for integrins expressed on different cell types during tissue fibrogenesis. In this review, we will examine the ways in which integrins regulate these processes and discuss how the manipulation of integrins using function blocking antibodies and small molecule inhibitors may have clinical utility in the treatment of patients with a broad range of fibrotic diseases. This article is part of a Special Issue entitled: Fibrosis: Translation of basic research to human disease.
Collapse
Affiliation(s)
- Neil C Henderson
- MRC Centre for Inflammation Research, The Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Dean Sheppard
- Lung Biology Center, Department of Medicine, University of California, San Francisco, California, USA
| |
Collapse
|
91
|
Mazo DFC, de Oliveira MG, Pereira IVA, Cogliati B, Stefano JT, de Souza GFP, Rabelo F, Lima FR, Ferreira Alves VA, Carrilho FJ, de Oliveira CPMS. S-nitroso-N-acetylcysteine attenuates liver fibrosis in experimental nonalcoholic steatohepatitis. DRUG DESIGN DEVELOPMENT AND THERAPY 2013; 7:553-63. [PMID: 23843692 PMCID: PMC3702228 DOI: 10.2147/dddt.s43930] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
S-Nitroso-N-acetylcysteine (SNAC) is a water soluble primary S-nitrosothiol capable of transferring and releasing nitric oxide and inducing several biochemical activities, including modulation of hepatic stellate cell activation. In this study, we evaluated the antifibrotic activity of SNAC in an animal model of nonalcoholic steatohepatitis (NASH) induced in Sprague-Dawley rats fed with a choline-deficient, high trans fat diet and exposed to diethylnitrosamine for 8 weeks. The rats were divided into three groups: SNAC, which received oral SNAC solution daily; NASH, which received the vehicle; and control, which received standard diet and vehicle. Genes related to fibrosis (matrix metalloproteinases [MMP]-13, -9, and -2), transforming growth factor β-1 [TGFβ-1], collagen-1α, and tissue inhibitors of metalloproteinase [TIMP-1 and -2] and oxidative stress (heat-shock proteins [HSP]-60 and -90) were evaluated. SNAC led to a 34.4% reduction in the collagen occupied area associated with upregulation of MMP-13 and -9 and downregulation of HSP-60, TIMP-2, TGFβ-1, and collagen-1α. These results indicate that oral SNAC administration may represent a potential antifibrotic treatment for NASH.
Collapse
Affiliation(s)
- Daniel F C Mazo
- University of São Paulo School of Medicine, Department of Gastroenterology, Clinical Division, Hepatology Branch (LIM-07), Sao Paulo, Brazil
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
92
|
Abstract
Liver fibrosis is the generic response to chronic injury of varying aetiologies. A number of common mechanisms link this response to the pathogenesis of fibrosis in other organs. While long thought to be relentlessly progressive, there is now excellent evidence in both human liver disease and animal models that hepatic fibrosis is potentially reversible. The liver therefore provides an excellent bidirectional model for the study of fibrogenesis and fibrosis resolution. In this article, we will review the evidence for the reversibility of liver fibrosis. We will highlight some of the mechanisms responsible for fibrogenesis and fibrosis regression, focussing on the role of hepatic myofibroblast activation and apoptosis, the importance of matrix metalloproteinases and their tissue inhibitors and the central involvement of hepatic macrophages in orchestrating this process. Finally, we will briefly discuss what renders liver fibrosis irreversible and how this accumulating knowledge base could lead to badly needed anti-fibrotic therapies in the future.
Collapse
Affiliation(s)
- P Ramachandran
- University of Edinburgh, The Queen's Medical Research Institute, Edinburgh EH16 4TJ, UK
| | | |
Collapse
|
93
|
Rashid ST, Humphries JD, Byron A, Dhar A, Askari JA, Selley JN, Knight D, Goldin RD, Thursz M, Humphries MJ. Proteomic analysis of extracellular matrix from the hepatic stellate cell line LX-2 identifies CYR61 and Wnt-5a as novel constituents of fibrotic liver. J Proteome Res 2012; 11:4052-64. [PMID: 22694338 PMCID: PMC3411196 DOI: 10.1021/pr3000927] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
![]()
Activation of hepatic stellate cells (HSCs) and subsequent
uncontrolled
accumulation of altered extracellular matrix (ECM) underpin liver
fibrosis, a wound healing response to chronic injury, which can lead
to organ failure and death. We sought to catalogue the components
of fibrotic liver ECM to obtain insights into disease etiology and
aid identification of new biomarkers. Cell-derived ECM was isolated
from the HSC line LX-2, an in vitro model of liver
fibrosis, and compared to ECM from human foreskin fibroblasts (HFFs)
as a control. Mass spectrometry analyses of cell-derived ECMs identified,
with ≥99% confidence, 61 structural ECM or secreted proteins
(48 and 31 proteins for LX-2 and HFF, respectively). Gene ontology
enrichment analysis confirmed the enrichment of ECM proteins, and
hierarchical clustering coupled with protein–protein interaction
network analysis revealed a subset of proteins enriched to fibrotic
ECM, highlighting the existence of cell type-specific ECM niches.
Thirty-six proteins were enriched to LX-2 ECM as compared to HFF ECM,
of which Wnt-5a and CYR61 were validated by immunohistochemistry in
human and murine fibrotic liver tissue. Future studies will determine
if these and other components may play a role in the etiology of hepatic
fibrosis, serve as novel disease biomarkers, or open up new avenues
for drug discovery.
Collapse
Affiliation(s)
- S Tamir Rashid
- Wellcome Trust Centre for Cell-Matrix Research, Faculty of Life Sciences, and Department of Gastrointestinal Sciences, University of Manchester, Oxford Road, Manchester M13 9PT, UK
| | | | | | | | | | | | | | | | | | | |
Collapse
|
94
|
Pellicoro A, Ramachandran P, Iredale JP. Reversibility of liver fibrosis. FIBROGENESIS & TISSUE REPAIR 2012; 5:S26. [PMID: 23259590 PMCID: PMC3368758 DOI: 10.1186/1755-1536-5-s1-s26] [Citation(s) in RCA: 80] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Liver fibrosis, and its end stage cirrhosis are a major cause of morbidity and mortality and therapeutic options are limited. However, the traditional view of liver disease as an irreversible process is obsolete and it is now evident that the development of liver fibrosis is a dynamic and potentially bidirectional process. Spontaneous resolution of scarring is seen in animal models of liver fibrosis and in human trials in which the stimuli responsible for chronic or repeated hepatic inflammation is successfully removed. Key players in the process are hepatic stellate cells, macrophages, MMPs and their inhibitors Timps. It is also evident that in advanced fibrotic liver disease, specific histological features define what is currently described as "irreversible" fibrosis. This includes the development of paucicellular scars enriched in extensively cross-linked matrix components, such as fibrillar collagen and elastin. Our recent work has focused on the role of macrophage metalloelastase (MMP-12) in the turnover of elastin in reversible and irreversible models of fibrosis. We have shown that elastin turnover in liver injury and fibrosis is regulated by macrophages via Mmp-12 expression, activity and ratio to its inhibitor Timp-1. Failure of elastin degradation, together with increased deposition leads to accumulation of elastin in the fibrotic scars.
Collapse
Affiliation(s)
- Antonella Pellicoro
- Centre for Inflammation Research, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Prakash Ramachandran
- Centre for Inflammation Research, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - John P Iredale
- Centre for Inflammation Research, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
95
|
Derdak Z, Villegas KA, Wands JR. Early growth response-1 transcription factor promotes hepatic fibrosis and steatosis in long-term ethanol-fed Long-Evans rats. Liver Int 2012; 32:761-70. [PMID: 22292946 PMCID: PMC10026596 DOI: 10.1111/j.1478-3231.2012.02752.x] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2011] [Accepted: 12/28/2011] [Indexed: 12/30/2022]
Abstract
BACKGROUND Previous studies demonstrated that the Long-Evans (LE) rats exhibited liver injury and lipid metabolic abnormalities after 8 weeks of ethanol feeding. AIMS The goal of this study was to investigate if the LE rats develop more advanced hepatic abnormalities (e.g., fibrosis) after long-term feeding with an ethanol-containing Lieber-DeCarli diet. In addition, the contribution of early growth response-1 (EGR1) transcription factor to these pathological changes was assessed. METHODS Long-Evans rats were fed an ethanol-containing or isocaloric control liquid diet for 18 months. Livers were processed for histological analyses, studies of fibrosis-related gene expression, cell fractionation and triglyceride measurement. Serum alanine aminotransferase (ALT) levels were assessed. DNA binding activities of p53 and the sterol regulatory element-binding protein-1c (SREBP1c) were analysed. The abundance of EGR1 and enzymes involved in fatty acid synthesis were determined. Chromatin immunoprecipitation was employed to study EGR1 binding to the SREBP1c promoter region. RESULTS Ethanol feeding generated steatosis, chicken wire fibrosis and ALT elevations in the LE rats. Fibrosis was associated with the upregulation of EGR1 and its downstream target genes. EGR1 upregulation was associated with enhanced p53 activity and an increase in the cellular p66(shc) abundance. Steatosis was linked to the activation of SREBP1c. Importantly, EGR1 upregulation paralleled the expression and transcriptional activity of SREBP1c. Finally, EGR1 was shown to bind to the SREBP1c promoter region. CONCLUSIONS Long-term ethanol feeding promoted steatosis and fibrosis in LE rats via EGR1 activation. The highly abundant EGR1 bound to the SREBP1c promoter and contributed to the steatosis observed in the LE rat model.
Collapse
Affiliation(s)
- Zoltan Derdak
- Division of Gastroenterology & Liver Research Center, Warren Alpert Medical School of Brown University and Rhode Island Hospital, Providence, RI, USA.
| | | | | |
Collapse
|
96
|
Olsen AL, Sackey BK, Marcinkiewicz C, Boettiger D, Wells RG. Fibronectin extra domain-A promotes hepatic stellate cell motility but not differentiation into myofibroblasts. Gastroenterology 2012; 142:928-937.e3. [PMID: 22202457 PMCID: PMC3321084 DOI: 10.1053/j.gastro.2011.12.038] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2011] [Revised: 11/29/2011] [Accepted: 12/13/2011] [Indexed: 12/13/2022]
Abstract
BACKGROUND & AIMS Myofibroblasts are the primary cell type involved in physiologic wound healing and its pathologic counterpart, fibrosis. Cellular fibronectin that contains the alternatively spliced extra domain A (EIIIA) is up-regulated during these processes and is believed to promote myofibroblast differentiation. We sought to determine the requirement for EIIIA in fibrosis and differentiation of myofibroblasts in rodent livers. METHODS We used a mechanically tunable hydrogel cell culture system to study differentiation of primary hepatic stellate cells and portal fibroblasts from rats into myofibroblasts. Liver fibrosis was induced in mice by bile duct ligation or administration of thioacetamide. RESULTS EIIIA was not required for differentiation of rat hepatic stellate cells or portal fibroblasts into fibrogenic myofibroblasts. Instead, hepatic stellate cells cultured on EIIIA-containing cellular fibronectin formed increased numbers of lamellipodia; their random motility and chemotaxis also increased. These increases required the receptor for EIIIA, the integrin α(9)β(1). In contrast, the motility of portal fibroblasts did not increase on EIIIA, and these cells expressed little α(9)β(1). Male EIIIA(-/-) mice were modestly protected from thioacetamide-induced fibrosis, which requires motile hepatic stellate cells, but not from bile duct ligation-induced fibrosis, in which portal fibroblasts are more important. Notably, myofibroblasts developed during induction of fibrosis with either thioacetamide or bile duct ligation in EIIIA(-/-) mice. CONCLUSIONS EIIIA is dispensable for differentiation of hepatic stellate cells and portal fibroblasts to myofibroblasts, both in culture and in mouse models of fibrosis. Our findings, however, indicate a role for EIIIA in promoting stellate cell motility and parenchymal liver fibrosis.
Collapse
Affiliation(s)
- Abby L. Olsen
- Department of Medicine, University of Pennsylvania School of Medicine, Philadelphia, PA 19104,Cell and Molecular Biology Graduate Group, University of Pennsylvania School of Medicine, Philadelphia, PA 19104
| | - Bridget K. Sackey
- Department of Medicine, University of Pennsylvania School of Medicine, Philadelphia, PA 19104,Cell and Molecular Biology Graduate Group, University of Pennsylvania School of Medicine, Philadelphia, PA 19104
| | | | - David Boettiger
- Cell and Molecular Biology Graduate Group, University of Pennsylvania School of Medicine, Philadelphia, PA 19104,Department of Microbiology, University of Pennsylvania School of Medicine, Philadelphia, PA 19104,Department of Pharmacology, University of Pennsylvania School of Medicine, Philadelphia, PA 19104
| | - Rebecca G. Wells
- Department of Medicine, University of Pennsylvania School of Medicine, Philadelphia, PA 19104,Cell and Molecular Biology Graduate Group, University of Pennsylvania School of Medicine, Philadelphia, PA 19104,Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, PA 19104
| |
Collapse
|
97
|
Jiang JX, Chen X, Hsu DK, Baghy K, Serizawa N, Scott F, Takada Y, Takada Y, Fukada H, Chen J, Devaraj S, Adamson R, Liu FT, Török NJ. Galectin-3 modulates phagocytosis-induced stellate cell activation and liver fibrosis in vivo. Am J Physiol Gastrointest Liver Physiol 2012; 302:G439-46. [PMID: 22159281 PMCID: PMC3287392 DOI: 10.1152/ajpgi.00257.2011] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Hepatic stellate cells (HSC), the key fibrogenic cells of the liver, transdifferentiate into myofibroblasts upon phagocytosis of apoptotic hepatocytes. Galectin-3, a β-galactoside-binding lectin, is a regulator of the phagocytic process. In this study, our aim was to study the mechanism by which extracellular galectin-3 modulates HSC phagocytosis and activation. The role of galectin-3 in engulfment was evaluated by phagocytosis and integrin binding assays in primary HSC. Galectin-3 expression was studied by real-time PCR and enzyme-linked immunosorbent assay, and in vivo studies were done in wild-type and galectin-3(-/-) mice. We found that HSC from galectin-3(-/-) mice displayed decreased phagocytic activity, expression of transforming growth factor-β1, and procollagen α1(I). Recombinant galectin-3 reversed this defect, suggesting that extracellular galectin-3 is required for HSC activation. Galectin-3 facilitated the α(v)β(3) heterodimer-dependent binding, indicating that galectin-3 modulates HSC phagocytosis via cross-linking this integrin and enhancing the tethering of apoptotic cells. Blocking integrin α(v)β(3) resulted in decreased phagocytosis. Galectin-3 expression and release were induced in active HSC engulfing apoptotic cells, and this was mediated by the nuclear factor-κB signaling. The upregulation of galectin-3 in active HSC was further confirmed in vivo in bile duct-ligated (BDL) rats. Galectin-3(-/-) mice displayed significantly decreased fibrosis, with reduced expression of α-smooth muscle actin and procollagen α1(I) following BDL. In summary, extracellular galectin-3 plays a key role in liver fibrosis by mediating HSC phagocytosis, activation, and subsequent autocrine and paracrine signaling by a feedforward mechanism.
Collapse
Affiliation(s)
- Joy X. Jiang
- Department of Internal Medicine, 1Division of Gastroenterology and Hepatology,
| | - Xiangling Chen
- Department of Internal Medicine, 1Division of Gastroenterology and Hepatology,
| | - Daniel K. Hsu
- 4Department of Dermatology, University of California Davis Medical Center, Sacramento, California
| | - Kornelia Baghy
- Department of Internal Medicine, 1Division of Gastroenterology and Hepatology,
| | - Nobuko Serizawa
- Department of Internal Medicine, 1Division of Gastroenterology and Hepatology,
| | - Fiona Scott
- Department of Internal Medicine, 1Division of Gastroenterology and Hepatology,
| | - Yoshikazu Takada
- 4Department of Dermatology, University of California Davis Medical Center, Sacramento, California
| | - Yoko Takada
- 4Department of Dermatology, University of California Davis Medical Center, Sacramento, California
| | - Hiroo Fukada
- Department of Internal Medicine, 1Division of Gastroenterology and Hepatology,
| | - Jenny Chen
- Department of Internal Medicine, 1Division of Gastroenterology and Hepatology,
| | | | - Roger Adamson
- 3Department of Human Physiology, University of California Davis, Sacramento; and
| | - Fu-Tong Liu
- 4Department of Dermatology, University of California Davis Medical Center, Sacramento, California
| | - Natalie J. Török
- Department of Internal Medicine, 1Division of Gastroenterology and Hepatology,
| |
Collapse
|
98
|
Urtasun R, Lopategi A, George J, Leung TM, Lu Y, Wang X, Ge X, Fiel MI, Nieto N. Osteopontin, an oxidant stress sensitive cytokine, up-regulates collagen-I via integrin α(V)β(3) engagement and PI3K/pAkt/NFκB signaling. Hepatology 2012; 55:594-608. [PMID: 21953216 PMCID: PMC3561739 DOI: 10.1002/hep.24701] [Citation(s) in RCA: 188] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
UNLABELLED A key feature in the pathogenesis of liver fibrosis is fibrillar Collagen-I deposition; yet, mediators that could be key therapeutic targets remain elusive. We hypothesized that osteopontin (OPN), an extracellular matrix (ECM) cytokine expressed in hepatic stellate cells (HSCs), could drive fibrogenesis by modulating the HSC pro-fibrogenic phenotype and Collagen-I expression. Recombinant OPN (rOPN) up-regulated Collagen-I protein in primary HSCs in a transforming growth factor beta (TGFβ)-independent fashion, whereas it down-regulated matrix metalloprotease-13 (MMP13), thus favoring scarring. rOPN activated primary HSCs, confirmed by increased α-smooth muscle actin (αSMA) expression and enhanced their invasive and wound-healing potential. HSCs isolated from wild-type (WT) mice were more profibrogenic than those from OPN knockout (Opn(-/-)) mice and infection of primary HSCs with an Ad-OPN increased Collagen-I, indicating correlation between both proteins. OPN induction of Collagen-I occurred via integrin α(v)β(3) engagement and activation of the phosphoinositide 3-kinase/phosphorylated Akt/nuclear factor kappa B (PI3K/pAkt/NFκB)-signaling pathway, whereas cluster of differentiation 44 (CD44) binding and mammalian target of rapamycin/70-kDa ribosomal protein S6 kinase (mTOR/p70S6K) were not involved. Neutralization of integrin α(v) β(3) prevented the OPN-mediated activation of the PI3K/pAkt/NFκB-signaling cascade and Collagen-I up-regulation. Likewise, inhibition of PI3K and NFκB blocked the OPN-mediated Collagen-I increase. Hepatitis C Virus (HCV) cirrhotic patients showed coinduction of Collagen-I and cleaved OPN compared to healthy individuals. Acute and chronic liver injury by CCl(4) injection or thioacetamide (TAA) treatment elevated OPN expression. Reactive oxygen species up-regulated OPN in vitro and in vivo and antioxidants prevented this effect. Transgenic mice overexpressing OPN in hepatocytes (Opn(HEP) Tg) mice developed spontaneous liver fibrosis compared to WT mice. Last, chronic CCl(4) injection and TAA treatment caused more liver fibrosis to WT than to Opn(-/-) mice and the reverse occurred in Opn(HEP) Tg mice. CONCLUSION OPN emerges as a key cytokine within the ECM protein network driving the increase in Collagen-I protein contributing to scarring and liver fibrosis.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Maria Isabel Fiel
- Division of Liver Diseases, Departments of Medicine and Pathology, Mount Sinai School of Medicine, Box 1123, 1425 Madison Avenue, Room 11-76, New York, NY 10029, USA
| | | |
Collapse
|
99
|
Bae HB, Zmijewski JW, Deshane JS, Zhi D, Thompson LC, Peterson CB, Chaplin DD, Abraham E. Vitronectin inhibits neutrophil apoptosis through activation of integrin-associated signaling pathways. Am J Respir Cell Mol Biol 2012; 46:790-6. [PMID: 22281987 DOI: 10.1165/rcmb.2011-0187oc] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Vitronectin is present in large concentrations in serum and the extracellular matrix. Although vitronectin is known to modulate neutrophil adhesion and chemotaxis, and to contribute to neutrophil-associated proinflammatory processes, a role in apoptosis has not been demonstrated. In the present studies, we found that neutrophils demonstrated more rapid progression to spontaneous or TNF-related apoptosis-inducing ligand-induced apoptosis when incubated under vitronectin-free conditions than when vitronectin was present. The ability of native vitronectin to delay neutrophil apoptosis was not recapitulated by the vitronectin somatomedin B domain. In contrast, inclusion of the cyclo[Arg-Gly-Asp-D-Phe-Val] peptide in cultures containing vitronectin resulted in enhanced neutrophil apoptosis, showing that the vitronectin RGD motif (Arg-Gly-Asp motif) was responsible for the antiapoptotic effects of vitronectin. Addition of antibodies to β(1), β(3), or β(5), but not to β(2) or β(4) integrins, reversed the ability of vitronectin to diminish neutrophil apoptosis. The ability of vitronectin to enhance neutrophil viability was dependent on activation of phosphatidylinositol 3-kinase and extracellular signal-regulated kinase 1/2 kinases, but not on the p38 kinase. Increased numbers of apoptotic neutrophils were present in the lungs of LPS-treated transgenic vitronectin-deficient mice, as compared with control mice. These results demonstrate a novel antiapoptotic function for vitronectin.
Collapse
Affiliation(s)
- Hong-Beom Bae
- Department of Medicine, University of Alabama at Birmingham Division of Pulmonary, Allergy & Critical Care Medicine, 901 19th Street South, Birmingham, AL 35294, USA
| | | | | | | | | | | | | | | |
Collapse
|
100
|
Mechanisms and biomarkers of apoptosis in liver disease and fibrosis. Int J Hepatol 2012; 2012:648915. [PMID: 22567408 PMCID: PMC3332069 DOI: 10.1155/2012/648915] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2011] [Accepted: 01/24/2012] [Indexed: 02/07/2023] Open
Abstract
Liver fibrosis and cirrhosis are a major cause of morbidity and mortality worldwide. Development of the fibrotic scar is an outcome of chronic liver diseases of varying aetiologies including alcoholic liver disease (ALD) nonalcoholic liver disease (NAFLD) including non-alcoholic steatohepatitis (NASH) viral hepatitis B and C (HBV, HCV). The critical step in the development of scar is activation of hepatic stellate cells (HSCs), which become the primary source of extracellular matrix. Aberrant apoptosis is a feature of chronic liver diseases and is associated with worsening stages of fibrosis. However, apoptosis is also the main mechanism promoting the resolution of fibrosis, and spontaneous or targeted apoptosis of HSC is associated with regression of fibrosis in animal models and patients with chronic liver disease. Given the importance of apoptosis in disease progression and resolution, there is much interest in precisely delineating the mechanisms involved and also developing biomarkers that accurately reflect the underlying pathogenesis. Here, we review the mechanisms driving apoptosis in development of liver disease and use of apoptosis -related biomarkers to aid in clinical diagnosis. Finally, we will also examine the recent literature regarding new insights into mechanisms involved in apoptosis of activated HSCs as possible method of fibrosis regression.
Collapse
|