1
|
Savić R, Yang J, Koplev S, An MC, Patel PL, O'Brien RN, Dubose BN, Dodatko T, Rogatsky E, Sukhavasi K, Ermel R, Ruusalepp A, Houten SM, Kovacic JC, Stewart AF, Yohn CB, Schadt EE, Laberge RM, Björkegren JLM, Tu Z, Argmann C. Integration of transcriptomes of senescent cell models with multi-tissue patient samples reveals reduced COL6A3 as an inducer of senescence. Cell Rep 2023; 42:113371. [PMID: 37938972 PMCID: PMC10955802 DOI: 10.1016/j.celrep.2023.113371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 05/23/2023] [Accepted: 10/17/2023] [Indexed: 11/10/2023] Open
Abstract
Senescent cells are a major contributor to age-dependent cardiovascular tissue dysfunction, but knowledge of their in vivo cell markers and tissue context is lacking. To reveal tissue-relevant senescence biology, we integrate the transcriptomes of 10 experimental senescence cell models with a 224 multi-tissue gene co-expression network based on RNA-seq data of seven tissues biopsies from ∼600 coronary artery disease (CAD) patients. We identify 56 senescence-associated modules, many enriched in CAD GWAS genes and correlated with cardiometabolic traits-which supports universality of senescence gene programs across tissues and in CAD. Cross-tissue network analyses reveal 86 candidate senescence-associated secretory phenotype (SASP) factors, including COL6A3. Experimental knockdown of COL6A3 induces transcriptional changes that overlap the majority of the experimental senescence models, with cell-cycle arrest linked to modulation of DREAM complex-targeted genes. We provide a transcriptomic resource for cellular senescence and identify candidate biomarkers, SASP factors, and potential drivers of senescence in human tissues.
Collapse
Affiliation(s)
- Radoslav Savić
- Department of Genetics & Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029-6574, USA
| | - Jialiang Yang
- Department of Genetics & Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029-6574, USA
| | - Simon Koplev
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, UK
| | - Mahru C An
- UNITY Biotechnology, South San Francisco, CA 94080, USA
| | | | | | | | - Tetyana Dodatko
- Department of Genetics & Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029-6574, USA
| | - Eduard Rogatsky
- Department of Genetics & Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029-6574, USA
| | - Katyayani Sukhavasi
- Department of Cardiac Surgery and The Heart Clinic, Tartu University Hospital, Tartu, Estonia
| | - Raili Ermel
- Department of Cardiac Surgery and The Heart Clinic, Tartu University Hospital, Tartu, Estonia
| | - Arno Ruusalepp
- Department of Cardiac Surgery and The Heart Clinic, Tartu University Hospital, Tartu, Estonia; Clinical Gene Networks AB, Stockholm, Sweden
| | - Sander M Houten
- Department of Genetics & Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029-6574, USA
| | - Jason C Kovacic
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029-6574, USA; Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia; St. Vincent's Clinical School, University of New South Wales, Sydney, NSW, Australia
| | - Andrew F Stewart
- Diabetes Obesity Metabolism Institute, The Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | | | - Eric E Schadt
- Department of Genetics & Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029-6574, USA
| | | | - Johan L M Björkegren
- Department of Genetics & Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029-6574, USA; Clinical Gene Networks AB, Stockholm, Sweden; Department of Medicine, Karolinska Institutet, Karolinska Universitetssjukhuset, Huddinge, Sweden
| | - Zhidong Tu
- Department of Genetics & Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029-6574, USA
| | - Carmen Argmann
- Department of Genetics & Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029-6574, USA.
| |
Collapse
|
2
|
De Angelis Rigotti F, Wiedmann L, Hubert MO, Vacca M, Hasan SS, Moll I, Carvajal S, Jiménez W, Starostecka M, Billeter AT, Müller-Stich B, Wolff G, Ekim-Üstünel B, Herzig S, Fandos-Ramo C, Krätzner R, Reich M, Keitel-Anselmino V, Heikenwälder M, Mogler C, Fischer A, Rodriguez-Vita J. Semaphorin 3C exacerbates liver fibrosis. Hepatology 2023; 78:1092-1105. [PMID: 37055018 DOI: 10.1097/hep.0000000000000407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Accepted: 03/28/2023] [Indexed: 04/15/2023]
Abstract
BACKGROUND AND AIMS Chronic liver disease is a growing epidemic, leading to fibrosis and cirrhosis. TGF-β is the pivotal profibrogenic cytokine that activates HSC, yet other molecules can modulate TGF-β signaling during liver fibrosis. Expression of the axon guidance molecules semaphorins (SEMAs), which signal through plexins and neuropilins (NRPs), have been associated with liver fibrosis in HBV-induced chronic hepatitis. This study aims at determining their function in the regulation of HSCs. APPROACH AND RESULTS We analyzed publicly available patient databases and liver biopsies. We used transgenic mice, in which genes are deleted only in activated HSCs to perform ex vivo analysis and animal models. SEMA3C is the most enriched member of the semaphorin family in liver samples from patients with cirrhosis. Higher expression of SEMA3C in patients with NASH, alcoholic hepatitis, or HBV-induced hepatitis discriminates those with a more profibrotic transcriptomic profile. SEMA3C expression is also elevated in different mouse models of liver fibrosis and in isolated HSCs on activation. In keeping with this, deletion of SEMA3C in activated HSCs reduces myofibroblast marker expression. Conversely, SEMA3C overexpression exacerbates TGF-β-mediated myofibroblast activation, as shown by increased SMAD2 phosphorylation and target gene expression. Among SEMA3C receptors, only NRP2 expression is maintained on activation of isolated HSCs. Interestingly, lack of NRP2 in those cells reduces myofibroblast marker expression. Finally, deletion of either SEMA3C or NRP2, specifically in activated HSCs, reduces liver fibrosis in mice. CONCLUSION SEMA3C is a novel marker for activated HSCs that plays a fundamental role in the acquisition of the myofibroblastic phenotype and liver fibrosis.
Collapse
Affiliation(s)
- Francesca De Angelis Rigotti
- Vascular Signaling and Cancer Division, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Tumor-Stroma Communication Laboratory, Centro de Investigación Príncipe Felipe, Valencia, Spain
| | - Lena Wiedmann
- Vascular Signaling and Cancer Division, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Faculty of Biosciences, University of Heidelberg, Heidelberg, Germany
| | - Max Ole Hubert
- Vascular Signaling and Cancer Division, German Cancer Research Center (DKFZ), Heidelberg, Germany
- European Center for Angioscience (ECAS), Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Margherita Vacca
- Vascular Signaling and Cancer Division, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Sana S Hasan
- Vascular Signaling and Cancer Division, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Iris Moll
- Vascular Signaling and Cancer Division, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Silvia Carvajal
- Service of Biochemistry and Molecular Genetics, Hospital Clinic Universitari, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Wladimiro Jiménez
- Service of Biochemistry and Molecular Genetics, Hospital Clinic Universitari, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Department of Biomedicine, Medical and Health Sciences School, University of Barcelona, Barcelona, Spain
| | - Maja Starostecka
- Vascular Signaling and Cancer Division, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Faculty of Biosciences, University of Heidelberg, Heidelberg, Germany
- European Molecular Biology Laboratory (EMBL), Genome Biology Unit, Heidelberg, Germany
| | - Adrian T Billeter
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg Hospital, Heidelberg, Germany
| | - Beat Müller-Stich
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg Hospital, Heidelberg, Germany
| | - Gretchen Wolff
- Institute for Diabetes and Cancer (IDC), Helmholtz Diabetes Center, Helmholtz Centre Munich, Neuherberg, Germany
- Joint Heidelberg-IDC Translational Diabetes Program, Department of Internal Medicine 1, Heidelberg University Hospital, Heidelberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany, and Chair Molecular Metabolic Control, Technical University Munich, Munich, Germany
| | - Bilgen Ekim-Üstünel
- Institute for Diabetes and Cancer (IDC), Helmholtz Diabetes Center, Helmholtz Centre Munich, Neuherberg, Germany
- Joint Heidelberg-IDC Translational Diabetes Program, Department of Internal Medicine 1, Heidelberg University Hospital, Heidelberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany, and Chair Molecular Metabolic Control, Technical University Munich, Munich, Germany
| | - Stephan Herzig
- Institute for Diabetes and Cancer (IDC), Helmholtz Diabetes Center, Helmholtz Centre Munich, Neuherberg, Germany
- Joint Heidelberg-IDC Translational Diabetes Program, Department of Internal Medicine 1, Heidelberg University Hospital, Heidelberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany, and Chair Molecular Metabolic Control, Technical University Munich, Munich, Germany
| | - Cristina Fandos-Ramo
- Tumor-Stroma Communication Laboratory, Centro de Investigación Príncipe Felipe, Valencia, Spain
| | - Ralph Krätzner
- Department of Pediatrics and Adolescent Medicine, University Medical Center Göttingen, Göttingen, Germany
| | - Maria Reich
- Department of Gastroenterology, Hepatology and Infectious Diseases, Otto-von-Guericke University Hospital Magdeburg, Magdeburg, Germany
- Chronic Inflammation and Cancer Division, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Verena Keitel-Anselmino
- Department of Gastroenterology, Hepatology and Infectious Diseases, Otto-von-Guericke University Hospital Magdeburg, Magdeburg, Germany
| | - Mathias Heikenwälder
- Chronic Inflammation and Cancer Division, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Carolin Mogler
- Institute of Pathology, Technical University of Munich, Munich, Germany
| | - Andreas Fischer
- Vascular Signaling and Cancer Division, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Institute for Clinical Chemistry, University Medical Center Göttingen, Göttingen, Germany
| | - Juan Rodriguez-Vita
- Vascular Signaling and Cancer Division, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Tumor-Stroma Communication Laboratory, Centro de Investigación Príncipe Felipe, Valencia, Spain
| |
Collapse
|
3
|
Luo J, Li L, Chang B, Zhu Z, Deng F, Hu M, Yu Y, Lu X, Chen Z, Zuo D, Zhou J. Mannan-Binding Lectin via Interaction With Cell Surface Calreticulin Promotes Senescence of Activated Hepatic Stellate Cells to Limit Liver Fibrosis Progression. Cell Mol Gastroenterol Hepatol 2022; 14:75-99. [PMID: 35381393 PMCID: PMC9117817 DOI: 10.1016/j.jcmgh.2022.03.011] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2021] [Revised: 03/28/2022] [Accepted: 03/28/2022] [Indexed: 12/18/2022]
Abstract
BACKGROUND & AIMS Liver fibrosis represents a hallmark of most chronic liver diseases (CLD) triggered by recurrent liver injury and subsequent myofibroblast transdifferentiations of resident hepatic stellate cells (HSCs). Mannan-binding lectin (MBL) is potentially involved in hepatic fibrosis in CLD through unclear mechanisms. Therefore, we investigated the crosstalk between MBL and HSCs, and the consequent effects on fibrosis progression. METHODS Samples from patients with liver cirrhosis were collected. MBL deficiency (MBL-/-) and wild-type (WT) C57BL/6J mice were used to construct a CCl4-induced liver fibrosis model. Administration of MBL-expressing, liver-specific, adeno-associated virus was performed to restore hepatic MBL expression in MBL-/- mice. The human HSC line LX-2 was used for in vitro experiments. RESULTS MBL levels in patients with liver cirrhosis were correlated with disease severity. In the CCl4-induced liver fibrosis model, MBL-/- mice showed severer liver fibrosis accompanied by reduced senescent activated HSCs in liver tissue compared with WT mice, which could be inhibited by administering MBL-expressing, liver-specific, adeno-associated virus. Moreover, depleting senescent cells with senolytic treatment could abrogate these differences owing to MBL absence. Furthermore, MBL could interact directly with calreticulin associated with low-density lipoprotein receptor-related protein 1 on the cell surface of HSCs, which further promotes senescence in HSCs by up-regulating the mammalian target of rapamycin/p53/p21 signaling pathway. CONCLUSIONS MBL as a newfound senescence-promoting modulator and its crosstalk with HSCs in the liver microenvironment is essential for the control of hepatic fibrosis progression, suggesting its potential therapeutic use in treating CLD associated with liver fibrosis.
Collapse
Affiliation(s)
- Jialiang Luo
- Department of Immunology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China; Department of Medical Laboratory, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, Guangdong, China
| | - Lei Li
- Department of Immunology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China; Department of Medical Laboratory, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, Guangdong, China
| | - Bo Chang
- Department of Immunology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Zhengyumeng Zhu
- Department of Immunology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China; Department of Medical Laboratory, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, Guangdong, China
| | - Fan Deng
- Department of Immunology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China; Department of Medical Laboratory, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, Guangdong, China
| | - Mengyao Hu
- Department of Immunology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Yu Yu
- Department of Immunology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Xiao Lu
- Department of Immunology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Zhengliang Chen
- Department of Immunology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Daming Zuo
- Department of Medical Laboratory, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, Guangdong, China.
| | - Jia Zhou
- Department of Immunology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China.
| |
Collapse
|
4
|
Xie T, Xia Z, Wang W, Zhou X, Xu C. BMPER Ameliorates Renal Fibrosis by Inhibiting Tubular Dedifferentiation and Fibroblast Activation. Front Cell Dev Biol 2021; 9:608396. [PMID: 33644047 PMCID: PMC7905093 DOI: 10.3389/fcell.2021.608396] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2020] [Accepted: 01/08/2021] [Indexed: 12/02/2022] Open
Abstract
Tubulointerstitial fibrosis is both a pathological manifestation of chronic kidney disease and a driving force for the progression of kidney disease. A previous study has shown that bone morphogenetic protein-binding endothelial cell precursor-derived regulator (BMPER) is involved in lung fibrogenesis. However, the role of BMPER in renal fibrosis remains unknown. In the present study, the expression of BMPER was examined by real-time PCR, Western blot and immunohistochemical staining. The in vitro effects of BMPER on tubular dedifferentiation and fibroblast activation were analyzed in cultured HK-2 and NRK-49F cells. The in vivo effects of BMPER were dissected in unilateral ureteral obstruction (UUO) mice by delivery of BMPER gene via systemic administration of plasmid vector. We reported that the expression of BMPER decreased in the kidneys of UUO mice and HK-2 cells. TGF-β1 increased inhibitor of differentiation-1 (Id-1) and induced epithelial mesenchymal transition in HK-2 cells, and knockdown of BMPER aggravated Id-1 up-regulation, E-cadherin loss, and tubular dedifferentiation. On the contrary, exogenous BMPER inhibited Id-1 up-regulation, prevented E-cadherin loss and tubular dedifferentiation after TGF-β1 exposure. In addition, exogenous BMPER suppressed fibroblast activation by hindering Erk1/2 phosphorylation. Knockdown of low-density lipoprotein receptor-related protein 1 abolished the inhibitory effect of BMPER on Erk1/2 phosphorylation and fibroblast activation. Moreover, delivery of BMPER gene improved renal tubular damage and interstitial fibrosis in UUO mice. Therefore, BMPER inhibits TGF-β1-induced tubular dedifferentiation and fibroblast activation and may hold therapeutic potential for tubulointerstitial fibrosis.
Collapse
Affiliation(s)
- Ting Xie
- Department of Woman's Health Care, Maternal and Child Health Hospital of Hubei Province, Wuhan, China
| | - Zunen Xia
- Department of Clinical Laboratory, Renmin Hospital of Wuhan University, Wuhan, China
| | - Wei Wang
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, China.,Institute of Urology, Anhui Medical University, Hefei, China
| | - Xiangjun Zhou
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Changgeng Xu
- Department of Urology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
5
|
Schnieder J, Mamazhakypov A, Birnhuber A, Wilhelm J, Kwapiszewska G, Ruppert C, Markart P, Wujak L, Rubio K, Barreto G, Schaefer L, Wygrecka M. Loss of LRP1 promotes acquisition of contractile-myofibroblast phenotype and release of active TGF-β1 from ECM stores. Matrix Biol 2019; 88:69-88. [PMID: 31841706 DOI: 10.1016/j.matbio.2019.12.001] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Revised: 12/06/2019] [Accepted: 12/07/2019] [Indexed: 12/16/2022]
Abstract
In healing tissue, fibroblasts differentiate to α-smooth muscle actin (SMA)-expressing contractile-myofibroblasts, which pull the wound edges together ensuring proper tissue repair. Uncontrolled expansion of the myofibroblast population may, however, lead to excessive tissue scarring and finally to organ dysfunction. Here, we demonstrate that the loss of low-density lipoprotein receptor-related protein (LRP) 1 overactivates the JNK1/2-c-Jun-Fra-2 signaling pathway leading to the induction of α-SMA and periostin expression in human lung fibroblasts (hLF). These changes are accompanied by increased contractility of the cells and the integrin- and protease-dependent release of active transforming growth factor (TGF)-β1 from the extracellular matrix (ECM) stores. Liberation of active TGF-β1 from the ECM further enhances α-SMA and periostin expression thus accelerating the phenotypic switch of hLF. Global gene expression profiling of LRP1-depleted hLF revealed that the loss of LRP1 affects cytoskeleton reorganization, cell-ECM contacts, and ECM production. In line with these findings, fibrotic changes in the skin and lung of Fra-2 transgenic mice were associated with LRP1 depletion and c-Jun overexpression. Altogether, our results suggest that dysregulation of LRP1 expression in fibroblasts in healing tissue may lead to the unrestrained expansion of contractile myofibroblasts and thereby to fibrosis development. Further studies identifying molecules, which regulate LRP1 expression, may provide new therapeutic options for largely untreatable human fibrotic diseases.
Collapse
Affiliation(s)
- Jennifer Schnieder
- Departments of Biochemistry and Internal Medicine, Universities of Giessen and Marburg Lung Center, Giessen, Germany
| | - Argen Mamazhakypov
- Departments of Biochemistry and Internal Medicine, Universities of Giessen and Marburg Lung Center, Giessen, Germany
| | - Anna Birnhuber
- Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria
| | - Jochen Wilhelm
- Departments of Internal Medicine, Universities of Giessen and Marburg Lung Center, Giessen, Germany
| | | | - Clemens Ruppert
- Departments of Internal Medicine, Universities of Giessen and Marburg Lung Center, Giessen, Germany
| | - Philipp Markart
- Department of Pulmonary Medicine, Fulda Hospital, University Medicine Marburg, Campus Fulda, Fulda, Germany
| | - Lukasz Wujak
- Departments of Biochemistry and Internal Medicine, Universities of Giessen and Marburg Lung Center, Giessen, Germany
| | - Karla Rubio
- Lung Cancer Epigenetic, Max-Planck-Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Guillermo Barreto
- Lung Cancer Epigenetic, Max-Planck-Institute for Heart and Lung Research, Bad Nauheim, Germany; Brain and Lung Epigenetics, Laboratoire Croissance, Réparation et Régénération Tissulaires (CRRET), Université Paris Est Créteil (UPEC), Créteil, France
| | - Liliana Schaefer
- Institute of Pharmacology and Toxicology, Goethe University, Frankfurt Am Main, Germany
| | - Malgorzata Wygrecka
- Departments of Biochemistry and Internal Medicine, Universities of Giessen and Marburg Lung Center, Giessen, Germany.
| |
Collapse
|
6
|
Murphy-Ullrich JE. Thrombospondin 1 and Its Diverse Roles as a Regulator of Extracellular Matrix in Fibrotic Disease. J Histochem Cytochem 2019; 67:683-699. [PMID: 31116066 PMCID: PMC6713974 DOI: 10.1369/0022155419851103] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Accepted: 04/26/2019] [Indexed: 01/06/2023] Open
Abstract
Thrombospondin 1 (TSP1) is a matricellular extracellular matrix protein that has diverse roles in regulating cellular processes important for the pathogenesis of fibrotic diseases. We will present evidence for the importance of TSP1 control of latent transforming growth factor beta activation in renal fibrosis with an emphasis on diabetic nephropathy. Other functions of TSP1 that affect renal fibrosis, including regulation of inflammation and capillary density, will be addressed. Emerging roles for TSP1 N-terminal domain regulation of collagen matrix assembly, direct effects of TSP1-collagen binding, and intracellular functions of TSP1 in mediating endoplasmic reticulum stress responses in extracellular matrix remodeling and fibrosis, which could potentially affect renal fibrogenesis, will also be discussed. Finally, we will address possible strategies for targeting TSP1 functions to treat fibrotic renal disease.
Collapse
Affiliation(s)
- Joanne E Murphy-Ullrich
- Departments of Pathology, Cell Developmental and Integrative Biology, and Ophthalmology, The University of Alabama at Birmingham, Birmingham, AL
| |
Collapse
|
7
|
Pant A, Kopec AK, Baker KS, Cline-Fedewa H, Lawrence DA, Luyendyk JP. Plasminogen Activator Inhibitor-1 Reduces Tissue-Type Plasminogen Activator-Dependent Fibrinolysis and Intrahepatic Hemorrhage in Experimental Acetaminophen Overdose. THE AMERICAN JOURNAL OF PATHOLOGY 2018; 188:1204-1212. [PMID: 29454747 PMCID: PMC5911680 DOI: 10.1016/j.ajpath.2018.01.010] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Revised: 12/21/2017] [Accepted: 01/11/2018] [Indexed: 12/16/2022]
Abstract
Acetaminophen (APAP)-induced liver injury in mice is associated with activation of the coagulation cascade and deposition of fibrin in liver. Plasminogen activator inhibitor-1 (PAI-1) is an important physiological inhibitor of tissue-type plasminogen activator (tPA) and plays a critical role in fibrinolysis. PAI-1 expression is increased in both experimental APAP-induced liver injury and patients with acute liver failure. Prior studies have shown that PAI-1 prevents intrahepatic hemorrhage and mortality after APAP challenge, but the downstream mechanisms are not clear. We tested the hypothesis that PAI-1 limits liver-related morbidity after APAP challenge by reducing tPA-dependent fibrinolysis. Compared with APAP-challenged (300 mg/kg) wild-type mice, hepatic deposition of cross-linked fibrin was reduced, with intrahepatic congestion and hemorrhage increased in PAI-1-deficient mice 24 hours after APAP overdose. Administration of recombinant wild-type human PAI-1 reduced intrahepatic hemorrhage 24 hours after APAP challenge in PAI-1-/- mice, whereas a mutant PAI-1 lacking antiprotease function had no effect. Of interest, tPA deficiency alone did not affect APAP-induced liver damage. In contrast, fibrinolysis, intrahepatic congestion and hemorrhage, and mortality driven by PAI-1 deficiency were reduced in APAP-treated tPA-/-/PAI-1-/- double-knockout mice. The results identify PAI-1 as a critical regulator of intrahepatic fibrinolysis in experimental liver injury. Moreover, the results suggest that the balance between PAI-1 and tPA activity is an important determinant of liver pathology after APAP overdose.
Collapse
Affiliation(s)
- Asmita Pant
- Department of Pathobiology and Diagnostic Investigation, Michigan State University, East Lansing, Michigan; Institute for Integrative Toxicology, Michigan State University, East Lansing, Michigan
| | - Anna K Kopec
- Department of Pathobiology and Diagnostic Investigation, Michigan State University, East Lansing, Michigan; Institute for Integrative Toxicology, Michigan State University, East Lansing, Michigan
| | - Kevin S Baker
- Institute for Integrative Toxicology, Michigan State University, East Lansing, Michigan; Department of Pharmacology and Toxicology, Michigan State University, East Lansing, Michigan
| | - Holly Cline-Fedewa
- Department of Pathobiology and Diagnostic Investigation, Michigan State University, East Lansing, Michigan
| | - Daniel A Lawrence
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan
| | - James P Luyendyk
- Department of Pathobiology and Diagnostic Investigation, Michigan State University, East Lansing, Michigan; Institute for Integrative Toxicology, Michigan State University, East Lansing, Michigan; Department of Pharmacology and Toxicology, Michigan State University, East Lansing, Michigan.
| |
Collapse
|
8
|
Labeyrie PE, Goulay R, Martinez de Lizarrondo S, Hébert M, Gauberti M, Maubert E, Delaunay B, Gory B, Signorelli F, Turjman F, Touzé E, Courthéoux P, Vivien D, Orset C. Vascular Tissue-Type Plasminogen Activator Promotes Intracranial Aneurysm Formation. Stroke 2017; 48:2574-2582. [DOI: 10.1161/strokeaha.117.017305] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Revised: 06/08/2017] [Accepted: 07/05/2017] [Indexed: 12/14/2022]
Affiliation(s)
- Paul-Emile Labeyrie
- From the Department of Physiopathology and Imaging of Neurological Disorders, INSERM U1237, UNICAEN, GIP Cyceron, France (P.-E.L., R.G., S.M.d.L., M.H., M.G., E.M., B.D., E.T., P.C., D.V., C.O.); Department of Interventional Neuroradiology (P.-E.L., B.G., F.T.) and Department of Neurosurgery (F.S.), Hôpital Wertheimer, University Lyon 1, Bron, France; and Department of Neurology (E.T.), Department of Neuroradiology (P.C.), and Department of Clinical Research (D.V.), CHU Caen, University Caen
| | - Romain Goulay
- From the Department of Physiopathology and Imaging of Neurological Disorders, INSERM U1237, UNICAEN, GIP Cyceron, France (P.-E.L., R.G., S.M.d.L., M.H., M.G., E.M., B.D., E.T., P.C., D.V., C.O.); Department of Interventional Neuroradiology (P.-E.L., B.G., F.T.) and Department of Neurosurgery (F.S.), Hôpital Wertheimer, University Lyon 1, Bron, France; and Department of Neurology (E.T.), Department of Neuroradiology (P.C.), and Department of Clinical Research (D.V.), CHU Caen, University Caen
| | - Sara Martinez de Lizarrondo
- From the Department of Physiopathology and Imaging of Neurological Disorders, INSERM U1237, UNICAEN, GIP Cyceron, France (P.-E.L., R.G., S.M.d.L., M.H., M.G., E.M., B.D., E.T., P.C., D.V., C.O.); Department of Interventional Neuroradiology (P.-E.L., B.G., F.T.) and Department of Neurosurgery (F.S.), Hôpital Wertheimer, University Lyon 1, Bron, France; and Department of Neurology (E.T.), Department of Neuroradiology (P.C.), and Department of Clinical Research (D.V.), CHU Caen, University Caen
| | - Marie Hébert
- From the Department of Physiopathology and Imaging of Neurological Disorders, INSERM U1237, UNICAEN, GIP Cyceron, France (P.-E.L., R.G., S.M.d.L., M.H., M.G., E.M., B.D., E.T., P.C., D.V., C.O.); Department of Interventional Neuroradiology (P.-E.L., B.G., F.T.) and Department of Neurosurgery (F.S.), Hôpital Wertheimer, University Lyon 1, Bron, France; and Department of Neurology (E.T.), Department of Neuroradiology (P.C.), and Department of Clinical Research (D.V.), CHU Caen, University Caen
| | - Maxime Gauberti
- From the Department of Physiopathology and Imaging of Neurological Disorders, INSERM U1237, UNICAEN, GIP Cyceron, France (P.-E.L., R.G., S.M.d.L., M.H., M.G., E.M., B.D., E.T., P.C., D.V., C.O.); Department of Interventional Neuroradiology (P.-E.L., B.G., F.T.) and Department of Neurosurgery (F.S.), Hôpital Wertheimer, University Lyon 1, Bron, France; and Department of Neurology (E.T.), Department of Neuroradiology (P.C.), and Department of Clinical Research (D.V.), CHU Caen, University Caen
| | - Eric Maubert
- From the Department of Physiopathology and Imaging of Neurological Disorders, INSERM U1237, UNICAEN, GIP Cyceron, France (P.-E.L., R.G., S.M.d.L., M.H., M.G., E.M., B.D., E.T., P.C., D.V., C.O.); Department of Interventional Neuroradiology (P.-E.L., B.G., F.T.) and Department of Neurosurgery (F.S.), Hôpital Wertheimer, University Lyon 1, Bron, France; and Department of Neurology (E.T.), Department of Neuroradiology (P.C.), and Department of Clinical Research (D.V.), CHU Caen, University Caen
| | - Barbara Delaunay
- From the Department of Physiopathology and Imaging of Neurological Disorders, INSERM U1237, UNICAEN, GIP Cyceron, France (P.-E.L., R.G., S.M.d.L., M.H., M.G., E.M., B.D., E.T., P.C., D.V., C.O.); Department of Interventional Neuroradiology (P.-E.L., B.G., F.T.) and Department of Neurosurgery (F.S.), Hôpital Wertheimer, University Lyon 1, Bron, France; and Department of Neurology (E.T.), Department of Neuroradiology (P.C.), and Department of Clinical Research (D.V.), CHU Caen, University Caen
| | - Benjamin Gory
- From the Department of Physiopathology and Imaging of Neurological Disorders, INSERM U1237, UNICAEN, GIP Cyceron, France (P.-E.L., R.G., S.M.d.L., M.H., M.G., E.M., B.D., E.T., P.C., D.V., C.O.); Department of Interventional Neuroradiology (P.-E.L., B.G., F.T.) and Department of Neurosurgery (F.S.), Hôpital Wertheimer, University Lyon 1, Bron, France; and Department of Neurology (E.T.), Department of Neuroradiology (P.C.), and Department of Clinical Research (D.V.), CHU Caen, University Caen
| | - Francesco Signorelli
- From the Department of Physiopathology and Imaging of Neurological Disorders, INSERM U1237, UNICAEN, GIP Cyceron, France (P.-E.L., R.G., S.M.d.L., M.H., M.G., E.M., B.D., E.T., P.C., D.V., C.O.); Department of Interventional Neuroradiology (P.-E.L., B.G., F.T.) and Department of Neurosurgery (F.S.), Hôpital Wertheimer, University Lyon 1, Bron, France; and Department of Neurology (E.T.), Department of Neuroradiology (P.C.), and Department of Clinical Research (D.V.), CHU Caen, University Caen
| | - Francis Turjman
- From the Department of Physiopathology and Imaging of Neurological Disorders, INSERM U1237, UNICAEN, GIP Cyceron, France (P.-E.L., R.G., S.M.d.L., M.H., M.G., E.M., B.D., E.T., P.C., D.V., C.O.); Department of Interventional Neuroradiology (P.-E.L., B.G., F.T.) and Department of Neurosurgery (F.S.), Hôpital Wertheimer, University Lyon 1, Bron, France; and Department of Neurology (E.T.), Department of Neuroradiology (P.C.), and Department of Clinical Research (D.V.), CHU Caen, University Caen
| | - Emmanuel Touzé
- From the Department of Physiopathology and Imaging of Neurological Disorders, INSERM U1237, UNICAEN, GIP Cyceron, France (P.-E.L., R.G., S.M.d.L., M.H., M.G., E.M., B.D., E.T., P.C., D.V., C.O.); Department of Interventional Neuroradiology (P.-E.L., B.G., F.T.) and Department of Neurosurgery (F.S.), Hôpital Wertheimer, University Lyon 1, Bron, France; and Department of Neurology (E.T.), Department of Neuroradiology (P.C.), and Department of Clinical Research (D.V.), CHU Caen, University Caen
| | - Patrick Courthéoux
- From the Department of Physiopathology and Imaging of Neurological Disorders, INSERM U1237, UNICAEN, GIP Cyceron, France (P.-E.L., R.G., S.M.d.L., M.H., M.G., E.M., B.D., E.T., P.C., D.V., C.O.); Department of Interventional Neuroradiology (P.-E.L., B.G., F.T.) and Department of Neurosurgery (F.S.), Hôpital Wertheimer, University Lyon 1, Bron, France; and Department of Neurology (E.T.), Department of Neuroradiology (P.C.), and Department of Clinical Research (D.V.), CHU Caen, University Caen
| | - Denis Vivien
- From the Department of Physiopathology and Imaging of Neurological Disorders, INSERM U1237, UNICAEN, GIP Cyceron, France (P.-E.L., R.G., S.M.d.L., M.H., M.G., E.M., B.D., E.T., P.C., D.V., C.O.); Department of Interventional Neuroradiology (P.-E.L., B.G., F.T.) and Department of Neurosurgery (F.S.), Hôpital Wertheimer, University Lyon 1, Bron, France; and Department of Neurology (E.T.), Department of Neuroradiology (P.C.), and Department of Clinical Research (D.V.), CHU Caen, University Caen
| | - Cyrille Orset
- From the Department of Physiopathology and Imaging of Neurological Disorders, INSERM U1237, UNICAEN, GIP Cyceron, France (P.-E.L., R.G., S.M.d.L., M.H., M.G., E.M., B.D., E.T., P.C., D.V., C.O.); Department of Interventional Neuroradiology (P.-E.L., B.G., F.T.) and Department of Neurosurgery (F.S.), Hôpital Wertheimer, University Lyon 1, Bron, France; and Department of Neurology (E.T.), Department of Neuroradiology (P.C.), and Department of Clinical Research (D.V.), CHU Caen, University Caen
| |
Collapse
|