1
|
Dorris ER, Phelan DE, Russell J, Murphy M. Bone morphogenetic protein-3 is a negative regulator of transforming growth factor beta and fibrosis. Biochem Biophys Res Commun 2024; 738:150497. [PMID: 39151293 DOI: 10.1016/j.bbrc.2024.150497] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 07/29/2024] [Accepted: 08/01/2024] [Indexed: 08/19/2024]
Abstract
Fibrosis results in one-third of all deaths globally and is a major healthcare challenge. Fibrosis is scarring caused by the excess deposition of extracellular matrix proteins by fibroblasts. Inhibition of pathways downstream of transforming growth factor β (TGF-β) a pluripotent growth factor, has potent antifibrotic effects in different organs. Here we show that loss of bone morphogenetic protein (BMP-3) is a feature of kidney fibrosis, independent of the initiating injury, suggesting loss of this cytokine is a core fibrotic mechanism. TGF-β decreased BMP3 expression in human fibroblasts is possibly a feed-forward loop that contributes to increased and sustained TGF-β activity. Recombinant human BMP-3 reduced TGF-β induced fibroblast contraction, migration and invasion, pathways that lead to scarring and tissue stiffening. BMP-3 reduced TGF-β stimulated collagen cross-linking, and Ox-LDL receptor 1, a regulator of collagen deposition. BMP-3 inhibited TGF-β stimulated lysyl oxidase activity. Lysyl oxidase mediated collagen cross-linking is a critical process in TGF-β induced fibrosis. We propose that BMP-3 alters fibroblast responses to TGF-β, shifting the balance from fibrosis to repair. Recombinant human BMP-3 shows promise for development as a novel therapeutic for fibrosis.
Collapse
Affiliation(s)
- Emma R Dorris
- School of Medicine, University College Dublin, Dublin, Ireland; National Children's Research Centre, Children's Health Ireland (Crumlin), Dublin, Ireland
| | - David E Phelan
- School of Medicine, University College Dublin, Dublin, Ireland; National Children's Research Centre, Children's Health Ireland (Crumlin), Dublin, Ireland
| | - John Russell
- National Children's Research Centre, Children's Health Ireland (Crumlin), Dublin, Ireland; Department of Pediatric Otorhinolaryngology, Children's Health Ireland (Crumlin), Dublin, Ireland
| | - Madeline Murphy
- School of Medicine, University College Dublin, Dublin, Ireland; National Children's Research Centre, Children's Health Ireland (Crumlin), Dublin, Ireland.
| |
Collapse
|
2
|
Hertig V, Villeneuve L, Calderone A. Nestin identifies a subpopulation of rat ventricular fibroblasts and participates in cell migration. Am J Physiol Cell Physiol 2023; 325:C496-C508. [PMID: 37458435 DOI: 10.1152/ajpcell.00161.2023] [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: 04/25/2023] [Revised: 07/03/2023] [Accepted: 07/03/2023] [Indexed: 08/08/2023]
Abstract
Fibroblast progenitor cells migrate to the endocardial region during cardiogenesis, and the migration of ventricular fibroblasts to the ischemically damaged region of the infarcted adult heart is a seminal event of reparative fibrosis. The intermediate filament protein nestin is implicated in cell migration and expression identified in a subpopulation of scar-derived myofibroblasts. The present study tested the hypothesis that fibroblast progenitor cells express nestin, and the intermediate filament protein drives the migratory phenotype of ventricular fibroblasts. Transcription factor 21 (Tcf21)- and Wilms tumor 1 (WT1)-fibroblast progenitor cells identified in the epicardial/endocardial regions of the E12.5- to E13.5-day embryonic mouse heart predominantly expressed nestin. Nuclear Tcf21/WT1 staining was identified in neonatal rat ventricular fibroblasts (NNVFbs), and a subpopulation coexpressed nestin. Nuclear Tcf21/WT1 expression persisted in adult rat ventricular fibroblasts, whereas nestin protein levels were downregulated. Nestin-expressing NNVFbs exhibited a unique phenotype as the subpopulation was refractory to cell cycle reentry in response to selective stimuli. Nestin(-)- and nestin(+)-scar-derived rat myofibroblasts plated in Matrigel unmasked a migratory phenotype characterized by the de novo formation of lumen-like structures. The elongated membrane projections emanating from scar myofibroblasts delineating the boundary of lumen-like structures expressed nestin. Lentiviral short-hairpin RNA (shRNA)-mediated nestin depletion inhibited the in vitro migratory response of NNVFbs as the wound radius was significantly larger compared with NNVFbs infected with the empty lentivirus. Thus, nestin represents a marker of embryonic Tcf21/WT1(+)-fibroblast progenitor cells. The neonatal rat heart contains a distinct subpopulation of nestin-immunoreactive Tcf21/WT1(+) fibroblasts refractory to cell cycle reentry, and the intermediate filament protein may preferentially facilitate ventricular fibroblast migration during physiological/pathological remodeling.NEW & NOTEWORTHY Tcf21/WT1(+)-fibroblast progenitor cells of the embryonic mouse heart predominantly express the intermediate filament protein nestin. A subpopulation of Tcf21/WT1(+)-neonatal rat ventricular fibroblasts express nestin and are refractory to selective stimuli influencing cell cycle reentry. Scar-derived myofibroblasts plated in Matrigel elicit the formation of lumen-like structures characterized by the appearance of nestin(+)-membrane projections. Lentiviral shRNA-mediated nestin depletion in a subpopulation of neonatal rat ventricular fibroblasts suppressed the migratory response following the in vitro scratch assay.
Collapse
Affiliation(s)
- Vanessa Hertig
- Research Center, Montreal Heart Institute, Montréal, Québec, Canada
| | - Louis Villeneuve
- Research Center, Montreal Heart Institute, Montréal, Québec, Canada
| | - Angelino Calderone
- Research Center, Montreal Heart Institute, Montréal, Québec, Canada
- Département de Pharmacologie et Physiologie, Université de Montréal, Montréal, Québec, Canada
| |
Collapse
|
3
|
Gutsol AA, Hale TM, Thibodeau JF, Holterman CE, Nasrallah R, Correa JWN, Touyz RM, Kennedy CRJ, Burger D, Hébert RL, Burns KD. Comparative Analysis of Hypertensive Tubulopathy in Animal Models of Hypertension and Its Relevance to Human Pathology. Toxicol Pathol 2023; 51:160-175. [PMID: 37632371 DOI: 10.1177/01926233231191128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/28/2023]
Abstract
Assessment of hypertensive tubulopathy for more than fifty animal models of hypertension in experimental pathology employs criteria that do not correspond to lesional descriptors for tubular lesions in clinical pathology. We provide a critical appraisal of experimental hypertension with the same approach used to estimate hypertensive renal tubulopathy in humans. Four models with different pathogenesis of hypertension were analyzed-chronic angiotensin (Ang) II-infused and renin-overexpressing (TTRhRen) mice, spontaneously hypertensive (SHR), and Goldblatt two-kidney one-clip (2K1C) rats. Mouse models, SHR, and the nonclipped kidney in 2K1C rats had no regular signs of hypertensive tubulopathy. Histopathology in animals was mild and limited to variations in the volume density of tubular lumen and epithelium, interstitial space, and interstitial collagen. Affected kidneys in animals demonstrated lesion values that are significantly different compared with healthy controls but correspond to mild damage if compared with hypertensive humans. The most substantial human-like hypertensive tubulopathy was detected in the clipped kidney of 2K1C rats. For the first time, our study demonstrated the regular presence of chronic progressive nephropathy (CPN) in relatively young mice and rats with induced hypertension. Because CPN may confound the assessment of rodent models of hypertension, proliferative markers should be used to verify nonhypertensive tubulopathy.
Collapse
Affiliation(s)
- Alex A Gutsol
- Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - Taben M Hale
- The University of Arizona, Phoenix, Arizona, USA
| | | | | | | | | | | | - Chris R J Kennedy
- Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
- University of Ottawa, Ottawa, Ontario, Canada
| | - Dylan Burger
- Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
- University of Ottawa, Ottawa, Ontario, Canada
| | - Richard L Hébert
- Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
- University of Ottawa, Ottawa, Ontario, Canada
| | - Kevin D Burns
- Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
- University of Ottawa, Ottawa, Ontario, Canada
| |
Collapse
|
4
|
Li Y, Li C, Liu Q, Wang L, Bao AX, Jung JP, Dodlapati S, Sun J, Gao P, Zhang X, Francis J, Molkentin JD, Fu X. Loss of Acta2 in cardiac fibroblasts does not prevent the myofibroblast differentiation or affect the cardiac repair after myocardial infarction. J Mol Cell Cardiol 2022; 171:117-132. [PMID: 36007455 PMCID: PMC10478266 DOI: 10.1016/j.yjmcc.2022.08.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 08/04/2022] [Accepted: 08/10/2022] [Indexed: 11/24/2022]
Abstract
In response to myocardial infarction (MI), quiescent cardiac fibroblasts differentiate into myofibroblasts mediating tissue repair. One of the most widely accepted markers of myofibroblast differentiation is the expression of Acta2 which encodes smooth muscle alpha-actin (SMαA) that is assembled into stress fibers. However, the requirement of Acta2/SMαA in the myofibroblast differentiation of cardiac fibroblasts and its role in post-MI cardiac repair remained unknown. To answer these questions, we generated a tamoxifen-inducible cardiac fibroblast-specific Acta2 knockout mouse line. Surprisingly, mice that lacked Acta2 in cardiac fibroblasts had a normal post-MI survival rate. Moreover, Acta2 deletion did not affect the function or histology of infarcted hearts. No difference was detected in the proliferation, migration, or contractility between WT and Acta2-null cardiac myofibroblasts. Acta2-null cardiac myofibroblasts had a normal total filamentous actin level and total actin level. Acta2 deletion caused a significant compensatory increase in the transcription level of non-Acta2 actin isoforms, especially Actg2 and Acta1. Moreover, in myofibroblasts, the transcription levels of cytoplasmic actin isoforms were significantly higher than those of muscle actin isoforms. In addition, we found that myocardin-related transcription factor-A is critical for myofibroblast differentiation but is not required for the compensatory effects of non-Acta2 isoforms. In conclusion, the Acta2 deletion does not prevent the myofibroblast differentiation of cardiac fibroblasts or affect the post-MI cardiac repair, and the increased expression and stress fiber formation of non-SMαA actin isoforms and the functional redundancy between actin isoforms are able to compensate for the loss of Acta2 in cardiac myofibroblasts.
Collapse
Affiliation(s)
- Yuxia Li
- School of Animal Sciences, AgCenter, Louisiana State University, Baton Rouge, LA, USA
| | - Chaoyang Li
- School of Animal Sciences, AgCenter, Louisiana State University, Baton Rouge, LA, USA
| | - Qianglin Liu
- School of Animal Sciences, AgCenter, Louisiana State University, Baton Rouge, LA, USA
| | - Leshan Wang
- School of Animal Sciences, AgCenter, Louisiana State University, Baton Rouge, LA, USA
| | - Adam X Bao
- Department of Biological Engineering, Louisiana State University, Baton Rouge, LA, USA
| | - Jangwook P Jung
- Department of Biological Engineering, Louisiana State University, Baton Rouge, LA, USA
| | - Sanjeev Dodlapati
- Department of Computer Science, Old Dominion University, Norfolk, VA, USA
| | - Jiangwen Sun
- Department of Computer Science, Old Dominion University, Norfolk, VA, USA
| | - Peidong Gao
- School of Animal Sciences, AgCenter, Louisiana State University, Baton Rouge, LA, USA
| | - Xujia Zhang
- School of Animal Sciences, AgCenter, Louisiana State University, Baton Rouge, LA, USA
| | - Joseph Francis
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, USA
| | - Jeffery D Molkentin
- Cincinnati Children's Hospital Medical Center, Department of Pediatrics, University of Cincinnati, Cincinnati, OH, USA
| | - Xing Fu
- School of Animal Sciences, AgCenter, Louisiana State University, Baton Rouge, LA, USA.
| |
Collapse
|
5
|
Cancer type-specific alterations in actin genes: Worth a closer look? INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2021; 360:133-184. [PMID: 33962749 DOI: 10.1016/bs.ircmb.2021.02.017] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Actins form a strongly conserved family of proteins that are central to the functioning of the actin cytoskeleton partaking in natural processes such as cell division, adhesion, contraction and migration. These processes, however, also occur during the various phases of cancer progression. Yet, surprisingly, alterations in the six human actin genes in cancer studies have received little attention and the focus was mostly on deregulated expression levels of actins and even more so of actin-binding or regulatory proteins. Starting from the early mutation work in the 1980s, we propose based on reviewing literature and data from patient cancer genomes that alterations in actin genes are different in distinct cancer subtypes, suggesting some specificity. These actin gene alterations include (missense) mutations, gene fusions and copy number alterations (deletions and amplifications) and we illustrate their occurrence for a limited number of examples including actin mutations in lymphoid cancers and nonmelanoma skin cancer and actin gene copy number alterations for breast, prostate and liver cancers. A challenge in the future will be to further sort out the specificity per actin gene, alteration type and cancer subtype. Even more challenging is (experimentally) distinguishing between cause and consequence: which alterations are passengers and which are involved in tumor progression of particular cancer subtypes?
Collapse
|
6
|
Shi Z, Ren M, Rockey DC. Myocardin and myocardin-related transcription factor-A synergistically mediate actin cytoskeletal-dependent inhibition of liver fibrogenesis. Am J Physiol Gastrointest Liver Physiol 2020; 318:G504-G517. [PMID: 31928221 PMCID: PMC7099496 DOI: 10.1152/ajpgi.00302.2019] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Activation of hepatic stellate cells (HSCs), characterized by development of a robust actin cytoskeleton and expression of abundant extracellular matrix (ECM) proteins, such as type 1 collagen (COL.1), is a central cellular and molecular event in liver fibrosis. It has been demonstrated that HSCs express both myocardin and myocardin-related transcription factor-A (MRTF-A). However, the biological effects of myocardin and MRTF-A on HSC activation and liver fibrosis, as well as the molecular mechanism under the process, remain unclear. Here, we report that myocardin and MRTF-A's expression and nuclear accumulation are prominently increased during the HSC activation process, accompanied by robust activation of actin cytoskeleton dynamics. Targeting myocardin and MRTF-A binding and function with a novel small molecule, CCG-203971, led to dose-dependent inhibition of HSC actin cytoskeleton dynamics and abrogated multiple functional features of HSC activation (i.e., HSC contraction, migration and proliferation) and decreased COL.1 expression in vitro and liver fibrosis in vivo. Mechanistically, blocking the myocardin and MRTF-A nuclear translocation pathway with CCG-203971 directly inhibited myocardin/MRTF-A-mediated serum response factor (SRF), and Smad2/3 activation in the COL.1α2 promoter and indirectly abrogated actin cytoskeleton-dependent regulation of Smad2/3 and Erk1/2 phosphorylation and their nuclear accumulation. Finally, there was no effect of CCG-203971 on markers of inflammation, suggesting a direct effect of the compound on HSCs and liver fibrosis. These data reveal that myocardin and MRTF-A are two important cotranscriptional factors in HSCs and represent entirely novel therapeutic pathways that might be targeted to treat liver fibrosis.NEW & NOTEWORTHY Myocardin and myocardin-related transcription factor-A (MRTF-A) are upregulated in activated hepatic stellate cells (HSCs) in vitro and in vivo, closely associated with robustly increased actin cytoskeleton remodeling. Targeting myocardin and MRTF-A by CCG-203971 leads to actin cytoskeleton-dependent inhibition of HSC activation, reduced cell contractility, impeded cell migration and proliferation, and decreased COL.1 expression in vitro and in vivo. Dual expression of myocardin and MRTF-A in HSCs may represent novel therapeutic targets in liver fibrosis.
Collapse
Affiliation(s)
- Zengdun Shi
- Department of Internal Medicine, Medical University of South Carolina, Charleston, South Carolina
| | - Mudan Ren
- Department of Internal Medicine, Medical University of South Carolina, Charleston, South Carolina
| | - Don C. Rockey
- Department of Internal Medicine, Medical University of South Carolina, Charleston, South Carolina
| |
Collapse
|
7
|
Sacco A, Bruno A, Contursi A, Dovizio M, Tacconelli S, Ricciotti E, Guillem-Llobat P, Salvatore T, Di Francesco L, Fullone R, Ballerini P, Arena V, Alberti S, Liu G, Gong Y, Sgambato A, Patrono C, FitzGerald GA, Yu Y, Patrignani P. Platelet-Specific Deletion of Cyclooxygenase-1 Ameliorates Dextran Sulfate Sodium-Induced Colitis in Mice. J Pharmacol Exp Ther 2019; 370:416-426. [PMID: 31248980 DOI: 10.1124/jpet.119.259382] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Accepted: 06/24/2019] [Indexed: 01/10/2023] Open
Abstract
Inflammatory bowel disease (IBD) is associated with an increased risk for thromboembolism, platelet activation, and abnormalities in platelet number and size. In colitis, platelets can extravasate into the colonic interstitium. We generated a mouse with a specific deletion of cyclooxygenase (COX)-1 in megakaryocytes/platelets [(COX-1 conditional knockout (cKO)] to clarify the role of platelet activation in the development of inflammation and fibrosis in dextran sodium sulfate (DSS)-induced colitis. The disease activity index was assessed, and colonic specimens were evaluated for histologic features of epithelial barrier damage, inflammation, and fibrosis. Cocultures of platelets and myofibroblasts were performed. We found that the specific deletion of COX-1 in platelets, which recapitulated the human pharmacodynamics of low-dose aspirin, that is, suppression of platelet thromboxane (TX)A2 production associated with substantial sparing of the systemic production of prostacyclin, resulted in milder symptoms of colitis, in the acute phase, and almost complete recovery from the disease after DSS withdrawal. Reduced colonic accumulation of macrophages and myofibroblasts and collagen deposition was found. Platelet-derived TXA2 enhanced the ability of myofibroblasts to proliferate and migrate in vitro, and these effects were prevented by platelet COX-1 inhibition or antagonism of the TXA2 receptor. Our findings allow a significant advance in the knowledge of the role of platelet-derived TXA2 in the development of colitis and fibrosis in response to intestinal damage and provide the rationale to investigate the potential efficacy of the antiplatelet agent low-dose aspirin in limiting the inflammatory response and fibrosis associated with IBD. SIGNIFICANCE STATEMENT: Inflammatory bowel disease (IBD) is characterized by the development of a chronic inflammatory response, which can lead to intestinal fibrosis for which currently there is no medical treatment. Through the generation of a mouse with specific deletion of cyclooxygenase-1 in megakaryocytes/platelets, which recapitulates the human pharmacodynamics of low-dose aspirin, we demonstrate the important role of platelet-derived thromboxane A2 in the development of experimental colitis and fibrosis, thus providing the rationale to investigate the potential efficacy of low-dose aspirin in limiting the inflammation and tissue damage associated with IBD.
Collapse
Affiliation(s)
- Angela Sacco
- Department of Neuroscience, Imaging, and Clinical Sciences and Center for Research on Aging and Translational Medicine, "G. d'Annunzio" University School of Medicine, Chieti, Italy (A.Sa., A.B., A.C., M.D., S.T., P.G.-L., T.S., L.D.F., R.F., P.B., S.A., P.P.); Department of Systems Pharmacology and Translational Therapeutics and Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania (E.R., G.A.F.); Departments of General Pathology (V.A., A.Sg.) and Pharmacology (C.P.), Catholic University School of Medicine, Rome, Italy; Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Graduate School of the Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China (G.L., Y.G.); and Department of Pharmacology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China (Y.Y.)
| | - Annalisa Bruno
- Department of Neuroscience, Imaging, and Clinical Sciences and Center for Research on Aging and Translational Medicine, "G. d'Annunzio" University School of Medicine, Chieti, Italy (A.Sa., A.B., A.C., M.D., S.T., P.G.-L., T.S., L.D.F., R.F., P.B., S.A., P.P.); Department of Systems Pharmacology and Translational Therapeutics and Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania (E.R., G.A.F.); Departments of General Pathology (V.A., A.Sg.) and Pharmacology (C.P.), Catholic University School of Medicine, Rome, Italy; Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Graduate School of the Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China (G.L., Y.G.); and Department of Pharmacology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China (Y.Y.)
| | - Annalisa Contursi
- Department of Neuroscience, Imaging, and Clinical Sciences and Center for Research on Aging and Translational Medicine, "G. d'Annunzio" University School of Medicine, Chieti, Italy (A.Sa., A.B., A.C., M.D., S.T., P.G.-L., T.S., L.D.F., R.F., P.B., S.A., P.P.); Department of Systems Pharmacology and Translational Therapeutics and Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania (E.R., G.A.F.); Departments of General Pathology (V.A., A.Sg.) and Pharmacology (C.P.), Catholic University School of Medicine, Rome, Italy; Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Graduate School of the Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China (G.L., Y.G.); and Department of Pharmacology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China (Y.Y.)
| | - Melania Dovizio
- Department of Neuroscience, Imaging, and Clinical Sciences and Center for Research on Aging and Translational Medicine, "G. d'Annunzio" University School of Medicine, Chieti, Italy (A.Sa., A.B., A.C., M.D., S.T., P.G.-L., T.S., L.D.F., R.F., P.B., S.A., P.P.); Department of Systems Pharmacology and Translational Therapeutics and Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania (E.R., G.A.F.); Departments of General Pathology (V.A., A.Sg.) and Pharmacology (C.P.), Catholic University School of Medicine, Rome, Italy; Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Graduate School of the Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China (G.L., Y.G.); and Department of Pharmacology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China (Y.Y.)
| | - Stefania Tacconelli
- Department of Neuroscience, Imaging, and Clinical Sciences and Center for Research on Aging and Translational Medicine, "G. d'Annunzio" University School of Medicine, Chieti, Italy (A.Sa., A.B., A.C., M.D., S.T., P.G.-L., T.S., L.D.F., R.F., P.B., S.A., P.P.); Department of Systems Pharmacology and Translational Therapeutics and Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania (E.R., G.A.F.); Departments of General Pathology (V.A., A.Sg.) and Pharmacology (C.P.), Catholic University School of Medicine, Rome, Italy; Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Graduate School of the Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China (G.L., Y.G.); and Department of Pharmacology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China (Y.Y.)
| | - Emanuela Ricciotti
- Department of Neuroscience, Imaging, and Clinical Sciences and Center for Research on Aging and Translational Medicine, "G. d'Annunzio" University School of Medicine, Chieti, Italy (A.Sa., A.B., A.C., M.D., S.T., P.G.-L., T.S., L.D.F., R.F., P.B., S.A., P.P.); Department of Systems Pharmacology and Translational Therapeutics and Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania (E.R., G.A.F.); Departments of General Pathology (V.A., A.Sg.) and Pharmacology (C.P.), Catholic University School of Medicine, Rome, Italy; Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Graduate School of the Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China (G.L., Y.G.); and Department of Pharmacology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China (Y.Y.)
| | - Paloma Guillem-Llobat
- Department of Neuroscience, Imaging, and Clinical Sciences and Center for Research on Aging and Translational Medicine, "G. d'Annunzio" University School of Medicine, Chieti, Italy (A.Sa., A.B., A.C., M.D., S.T., P.G.-L., T.S., L.D.F., R.F., P.B., S.A., P.P.); Department of Systems Pharmacology and Translational Therapeutics and Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania (E.R., G.A.F.); Departments of General Pathology (V.A., A.Sg.) and Pharmacology (C.P.), Catholic University School of Medicine, Rome, Italy; Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Graduate School of the Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China (G.L., Y.G.); and Department of Pharmacology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China (Y.Y.)
| | - Tania Salvatore
- Department of Neuroscience, Imaging, and Clinical Sciences and Center for Research on Aging and Translational Medicine, "G. d'Annunzio" University School of Medicine, Chieti, Italy (A.Sa., A.B., A.C., M.D., S.T., P.G.-L., T.S., L.D.F., R.F., P.B., S.A., P.P.); Department of Systems Pharmacology and Translational Therapeutics and Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania (E.R., G.A.F.); Departments of General Pathology (V.A., A.Sg.) and Pharmacology (C.P.), Catholic University School of Medicine, Rome, Italy; Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Graduate School of the Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China (G.L., Y.G.); and Department of Pharmacology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China (Y.Y.)
| | - Luigia Di Francesco
- Department of Neuroscience, Imaging, and Clinical Sciences and Center for Research on Aging and Translational Medicine, "G. d'Annunzio" University School of Medicine, Chieti, Italy (A.Sa., A.B., A.C., M.D., S.T., P.G.-L., T.S., L.D.F., R.F., P.B., S.A., P.P.); Department of Systems Pharmacology and Translational Therapeutics and Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania (E.R., G.A.F.); Departments of General Pathology (V.A., A.Sg.) and Pharmacology (C.P.), Catholic University School of Medicine, Rome, Italy; Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Graduate School of the Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China (G.L., Y.G.); and Department of Pharmacology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China (Y.Y.)
| | - Rosa Fullone
- Department of Neuroscience, Imaging, and Clinical Sciences and Center for Research on Aging and Translational Medicine, "G. d'Annunzio" University School of Medicine, Chieti, Italy (A.Sa., A.B., A.C., M.D., S.T., P.G.-L., T.S., L.D.F., R.F., P.B., S.A., P.P.); Department of Systems Pharmacology and Translational Therapeutics and Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania (E.R., G.A.F.); Departments of General Pathology (V.A., A.Sg.) and Pharmacology (C.P.), Catholic University School of Medicine, Rome, Italy; Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Graduate School of the Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China (G.L., Y.G.); and Department of Pharmacology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China (Y.Y.)
| | - Patrizia Ballerini
- Department of Neuroscience, Imaging, and Clinical Sciences and Center for Research on Aging and Translational Medicine, "G. d'Annunzio" University School of Medicine, Chieti, Italy (A.Sa., A.B., A.C., M.D., S.T., P.G.-L., T.S., L.D.F., R.F., P.B., S.A., P.P.); Department of Systems Pharmacology and Translational Therapeutics and Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania (E.R., G.A.F.); Departments of General Pathology (V.A., A.Sg.) and Pharmacology (C.P.), Catholic University School of Medicine, Rome, Italy; Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Graduate School of the Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China (G.L., Y.G.); and Department of Pharmacology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China (Y.Y.)
| | - Vincenzo Arena
- Department of Neuroscience, Imaging, and Clinical Sciences and Center for Research on Aging and Translational Medicine, "G. d'Annunzio" University School of Medicine, Chieti, Italy (A.Sa., A.B., A.C., M.D., S.T., P.G.-L., T.S., L.D.F., R.F., P.B., S.A., P.P.); Department of Systems Pharmacology and Translational Therapeutics and Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania (E.R., G.A.F.); Departments of General Pathology (V.A., A.Sg.) and Pharmacology (C.P.), Catholic University School of Medicine, Rome, Italy; Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Graduate School of the Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China (G.L., Y.G.); and Department of Pharmacology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China (Y.Y.)
| | - Sara Alberti
- Department of Neuroscience, Imaging, and Clinical Sciences and Center for Research on Aging and Translational Medicine, "G. d'Annunzio" University School of Medicine, Chieti, Italy (A.Sa., A.B., A.C., M.D., S.T., P.G.-L., T.S., L.D.F., R.F., P.B., S.A., P.P.); Department of Systems Pharmacology and Translational Therapeutics and Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania (E.R., G.A.F.); Departments of General Pathology (V.A., A.Sg.) and Pharmacology (C.P.), Catholic University School of Medicine, Rome, Italy; Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Graduate School of the Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China (G.L., Y.G.); and Department of Pharmacology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China (Y.Y.)
| | - Guizhu Liu
- Department of Neuroscience, Imaging, and Clinical Sciences and Center for Research on Aging and Translational Medicine, "G. d'Annunzio" University School of Medicine, Chieti, Italy (A.Sa., A.B., A.C., M.D., S.T., P.G.-L., T.S., L.D.F., R.F., P.B., S.A., P.P.); Department of Systems Pharmacology and Translational Therapeutics and Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania (E.R., G.A.F.); Departments of General Pathology (V.A., A.Sg.) and Pharmacology (C.P.), Catholic University School of Medicine, Rome, Italy; Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Graduate School of the Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China (G.L., Y.G.); and Department of Pharmacology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China (Y.Y.)
| | - Yanjun Gong
- Department of Neuroscience, Imaging, and Clinical Sciences and Center for Research on Aging and Translational Medicine, "G. d'Annunzio" University School of Medicine, Chieti, Italy (A.Sa., A.B., A.C., M.D., S.T., P.G.-L., T.S., L.D.F., R.F., P.B., S.A., P.P.); Department of Systems Pharmacology and Translational Therapeutics and Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania (E.R., G.A.F.); Departments of General Pathology (V.A., A.Sg.) and Pharmacology (C.P.), Catholic University School of Medicine, Rome, Italy; Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Graduate School of the Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China (G.L., Y.G.); and Department of Pharmacology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China (Y.Y.)
| | - Alessandro Sgambato
- Department of Neuroscience, Imaging, and Clinical Sciences and Center for Research on Aging and Translational Medicine, "G. d'Annunzio" University School of Medicine, Chieti, Italy (A.Sa., A.B., A.C., M.D., S.T., P.G.-L., T.S., L.D.F., R.F., P.B., S.A., P.P.); Department of Systems Pharmacology and Translational Therapeutics and Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania (E.R., G.A.F.); Departments of General Pathology (V.A., A.Sg.) and Pharmacology (C.P.), Catholic University School of Medicine, Rome, Italy; Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Graduate School of the Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China (G.L., Y.G.); and Department of Pharmacology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China (Y.Y.)
| | - Carlo Patrono
- Department of Neuroscience, Imaging, and Clinical Sciences and Center for Research on Aging and Translational Medicine, "G. d'Annunzio" University School of Medicine, Chieti, Italy (A.Sa., A.B., A.C., M.D., S.T., P.G.-L., T.S., L.D.F., R.F., P.B., S.A., P.P.); Department of Systems Pharmacology and Translational Therapeutics and Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania (E.R., G.A.F.); Departments of General Pathology (V.A., A.Sg.) and Pharmacology (C.P.), Catholic University School of Medicine, Rome, Italy; Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Graduate School of the Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China (G.L., Y.G.); and Department of Pharmacology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China (Y.Y.)
| | - Garret A FitzGerald
- Department of Neuroscience, Imaging, and Clinical Sciences and Center for Research on Aging and Translational Medicine, "G. d'Annunzio" University School of Medicine, Chieti, Italy (A.Sa., A.B., A.C., M.D., S.T., P.G.-L., T.S., L.D.F., R.F., P.B., S.A., P.P.); Department of Systems Pharmacology and Translational Therapeutics and Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania (E.R., G.A.F.); Departments of General Pathology (V.A., A.Sg.) and Pharmacology (C.P.), Catholic University School of Medicine, Rome, Italy; Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Graduate School of the Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China (G.L., Y.G.); and Department of Pharmacology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China (Y.Y.)
| | - Ying Yu
- Department of Neuroscience, Imaging, and Clinical Sciences and Center for Research on Aging and Translational Medicine, "G. d'Annunzio" University School of Medicine, Chieti, Italy (A.Sa., A.B., A.C., M.D., S.T., P.G.-L., T.S., L.D.F., R.F., P.B., S.A., P.P.); Department of Systems Pharmacology and Translational Therapeutics and Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania (E.R., G.A.F.); Departments of General Pathology (V.A., A.Sg.) and Pharmacology (C.P.), Catholic University School of Medicine, Rome, Italy; Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Graduate School of the Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China (G.L., Y.G.); and Department of Pharmacology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China (Y.Y.)
| | - Paola Patrignani
- Department of Neuroscience, Imaging, and Clinical Sciences and Center for Research on Aging and Translational Medicine, "G. d'Annunzio" University School of Medicine, Chieti, Italy (A.Sa., A.B., A.C., M.D., S.T., P.G.-L., T.S., L.D.F., R.F., P.B., S.A., P.P.); Department of Systems Pharmacology and Translational Therapeutics and Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania (E.R., G.A.F.); Departments of General Pathology (V.A., A.Sg.) and Pharmacology (C.P.), Catholic University School of Medicine, Rome, Italy; Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Graduate School of the Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China (G.L., Y.G.); and Department of Pharmacology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China (Y.Y.)
| |
Collapse
|
8
|
Rockey DC, Du Q, Weymouth ND, Shi Z. Smooth Muscle α-Actin Deficiency Leads to Decreased Liver Fibrosis via Impaired Cytoskeletal Signaling in Hepatic Stellate Cells. THE AMERICAN JOURNAL OF PATHOLOGY 2019; 189:2209-2220. [PMID: 31476284 DOI: 10.1016/j.ajpath.2019.07.019] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/17/2019] [Revised: 07/03/2019] [Accepted: 07/29/2019] [Indexed: 01/18/2023]
Abstract
In the liver, smooth muscle α-actin (SM α-actin) is up-regulated in hepatic stellate cells (HSCs) as they transition to myofibroblasts during liver injury and the wound healing response. Whether SM α-actin has specific functional effects on cellular effectors of fibrosis such as HSC is controversial. Here, the relationship between SM α-actin and type 1 collagen expression (COL1A1), a major extracellular matrix protein important in liver fibrosis, is investigated with the results demonstrating that knockout of SM α-actin leads to reduced liver fibrosis and COL1 expression. The mechanism for the reduction in fibrogenesis in vivo is multifactorial, including not only a reduction in the number of HSCs, but also an HSC-specific reduction in COL1 expression in Acta2-deficient HSCs. Despite a compensatory increase in expression of cytoplasmic β-actin and γ-actin isoforms in Acta2-/- HSCs, defects were identified in each transforming growth factor beta/Smad2/3 and ET-1/Erk1/2 signaling in Acta2-/- HSCs. These data not only suggest a molecular link between the SM α-actin cytoskeleton and classic fibrogenic signaling cascades, but also emphasize the relationship between SM α-actin and fibrogenesis in hepatic myofibroblasts in vivo.
Collapse
Affiliation(s)
- Don C Rockey
- Department of Internal Medicine, Medical University of South Carolina, Charleston, South Carolina.
| | - Qinghong Du
- Department of Internal Medicine, Medical University of South Carolina, Charleston, South Carolina
| | | | - Zengdun Shi
- Department of Internal Medicine, Medical University of South Carolina, Charleston, South Carolina.
| |
Collapse
|
9
|
Moonen L, Geryl H, D'Haese PC, Vervaet BA. Short-term dexamethasone treatment transiently, but not permanently, attenuates fibrosis after acute-to-chronic kidney injury. BMC Nephrol 2018; 19:343. [PMID: 30509215 PMCID: PMC6276259 DOI: 10.1186/s12882-018-1151-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Accepted: 11/23/2018] [Indexed: 01/30/2023] Open
Abstract
Background Acute kidney injury (AKI) is an underestimated, yet important, risk factor for the development of chronic kidney disease (CKD). Persistence of inflammation after a renal ischemic injury has been observed, both in experimental models and patients, and is thought to be an important mechanisms underlying progression of acute-to-chronic renal injury. Temporary suppression of inflammation immediately after AKI might therefore be a good first-line therapeutic strategy towards a better long term outcome. Methods Male C57Bl/6 J mice (Charles River, 10–12 weeks of age) underwent warm (36 °C body temperature) unilateral ischemia-reperfusion of the kidney for 21 min, after which treatment with intraperitoneal injection of the corticosteroid dexamethasone (10 mg/kg) was initiated for 3 weeks. Both at that time point and after an additional 3 week post-treatment follow up period, fibrosis was quantified by collagen I gene expression and immunostaining, as well as gene expression analysis of fibrosis-related genes Tgfβ, Ccn2 (Ctgf), Pai-1 and Ccn3. Furthermore, inflammation was evaluated by Tnfα gene expression and protein expression of the F4/80 macrophage marker and the α-SMA fibroblast marker. Lastly, renal histopathology was quantified by a morphometric analysis of the tubulointerstitial area. Results Treatment with dexamethasone attenuated development of fibrosis, as evidenced by reduced collagen I gene expression and immunostaining, in combination with reduced gene expression of the pro-fibrotic Ccn2 and increased expression of the anti-fibrotic Ccn3. The effects of dexamethasone on renal fibrosis persisted during the 3 week follow up period, as evidenced by stagnation of collagen I deposition in the ischemic kidney, in contrast to vehicle-treatment, where progression of fibrosis was observed. However, expression levels of the pro-fibrotic genes re-approached those of vehicle-treated injured kidneys suggesting that the effects of dexamethasone on fibrosis beyond the treatment period are temporary. Conclusion A short term anti-inflammatory therapy with dexamethasone only transiently attenuates ischemia induced fibrosis. Prolonged or persistent anti-inflammatory treatment seems warranted to achieve long term benefit. Electronic supplementary material The online version of this article (10.1186/s12882-018-1151-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Lies Moonen
- Laboratory of Pathophysiology, University of Antwerp, 2160, Antwerpen, Belgium
| | - Hilde Geryl
- Laboratory of Pathophysiology, University of Antwerp, 2160, Antwerpen, Belgium
| | - Patrick C D'Haese
- Laboratory of Pathophysiology, University of Antwerp, 2160, Antwerpen, Belgium
| | - Benjamin A Vervaet
- Laboratory of Pathophysiology, University of Antwerp, 2160, Antwerpen, Belgium.
| |
Collapse
|
10
|
Upregulation of the actin cytoskeleton via myocardin leads to increased expression of type 1 collagen. J Transl Med 2017; 97:1412-1426. [PMID: 29035375 PMCID: PMC6437559 DOI: 10.1038/labinvest.2017.96] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Revised: 06/19/2017] [Accepted: 07/05/2017] [Indexed: 12/28/2022] Open
Abstract
Liver fibrosis, a model wound healing system, is characterized by excessive deposition of extracellular matrix (ECM) in the liver. Although many fibrogenic cell types may express ECM, the hepatic stellate cell (HSC) is currently considered to be the major effector. HSCs transform into myofibroblast-like cells, also known as hepatic myofibroblasts in a process known as activation; this process is characterized in particular by de novo expression of smooth muscle alpha actin (SM α-actin) and type 1 collagen. The family of actins, which form the cell's cytoskeleton, are essential in many cellular processes. β-actin and cytoplasmic γ-actin (γ-actin) are ubiquitously expressed, whereas SM α-actin defines smooth muscle cell and myofibroblast phenotypes. Thus, SM α-actin is tightly associated with multiple functional properties. However, the regulatory mechanisms by which actin isoforms might regulate type 1 collagen remain unclear. In primary HSCs from normal and fibrotic rat liver, we demonstrate that myocardin, a canonical SRF cofactor, is upregulated in hepatic myofibroblasts and differentially regulates SM α-actin, γ-actin, and β-actins through activation of an ATTA box in the SM α-actin and a CCAAT box in γ-actin and β-actin promoters, respectively; moreover, myocardin differentially activated serum response factor (SRF) in CArG boxes of actin promoters. In addition, myocardin-stimulated Smad2 phosphorylation and RhoA expression, leading to increased expression of type 1 collagen in an actin cytoskeleton-dependent manner. Myocardin also directly enhanced SRF expression and stimulated collagen 1α1 and 1α2 promoter activities. In addition, overexpression of myocardin in vivo during carbon tetrachloride-induced liver injury led to increased HSC activation and fibrogenesis. In summary, our data suggest that myocardin plays a critical role in actin cytoskeletal dynamics during HSC activation, in turn, specifically regulating type I collagen expression in hepatic myofibroblasts.
Collapse
|
11
|
Higgins DF, Ewart LM, Masterson E, Tennant S, Grebnev G, Prunotto M, Pomposiello S, Conde-Knape K, Martin FM, Godson C. BMP7-induced-Pten inhibits Akt and prevents renal fibrosis. Biochim Biophys Acta Mol Basis Dis 2017; 1863:3095-3104. [PMID: 28923783 DOI: 10.1016/j.bbadis.2017.09.011] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Revised: 09/12/2017] [Accepted: 09/14/2017] [Indexed: 12/18/2022]
Abstract
Bone morphogenetic protein-7 (BMP-7) counteracts pro-fibrotic effects of TGFβ1 in cultured renal cells and protects from fibrosis in acute and chronic renal injury models. Using the unilateral ureteral obstruction (UUO) model of chronic renal fibrosis, we investigated the effect of exogenous-rhBMP-7 on pro-fibrotic signaling pathways mediated by TGFβ1 and hypoxia. Mice undergoing UUO were treated with vehicle or rhBMP-7 (300μg/kg i.p.) every other day for eight days and kidneys analysed for markers of fibrosis and SMAD, MAPK, and PI3K signaling. In the kidney, collecting duct and tubular epithelial cells respond to BMP-7 via activation of SMAD1/5/8. Phosphorylation of SMAD1/5/8 was reduced in UUO kidneys from vehicle-treated animals yet maintained in UUO kidneys from BMP-7-treated animals, confirming renal bioactivity of exogenous rhBMP-7. BMP-7 inhibited Collagen Iα1 and Collagen IIIα1 gene expression and Collagen I protein accumulation, while increasing expression of Collagen IVα1 in UUO kidneys. Activation of SMAD2, SMAD3, ERK, p38 and PI3K/Akt signaling occurred during fibrogenesis and BMP-7 significantly attenuated SMAD3 and Akt signaling in vivo. Analysis of renal collecting duct (mIMCD) and tubular epithelial (HK-2) cells stimulated with TGFβ1 or hypoxia (1% oxygen) to activate Akt provided further evidence that BMP-7 specifically inhibited PI3K/Akt signaling. PTEN is a negative regulator of PI3K and BMP-7 increased PTEN expression in vivo and in vitro. These data demonstrate an important mechanism by which BMP-7 orchestrates renal protection through Akt inhibition and highlights Akt inhibitors as anti-fibrotic therapeutics.
Collapse
Affiliation(s)
- Debra F Higgins
- UCD Diabetes Complications Research Centre, School of Medicine, Conway Institute, University College Dublin, Belfield, Dublin 4, Ireland.
| | - Leah M Ewart
- UCD Diabetes Complications Research Centre, School of Medicine, Conway Institute, University College Dublin, Belfield, Dublin 4, Ireland
| | - Enda Masterson
- UCD Diabetes Complications Research Centre, School of Medicine, Conway Institute, University College Dublin, Belfield, Dublin 4, Ireland
| | - Sadhbh Tennant
- UCD Diabetes Complications Research Centre, School of Medicine, Conway Institute, University College Dublin, Belfield, Dublin 4, Ireland
| | - Gleb Grebnev
- UCD Diabetes Complications Research Centre, School of Medicine, Conway Institute, University College Dublin, Belfield, Dublin 4, Ireland
| | - Marco Prunotto
- F. Hoffmann-La Roche, Dept of Metabolic and Vascular Disease, CH4070 Basel, Switzerland
| | - Sylvia Pomposiello
- F. Hoffmann-La Roche, Dept of Metabolic and Vascular Disease, CH4070 Basel, Switzerland
| | - Karin Conde-Knape
- F. Hoffmann-La Roche, Dept of Metabolic and Vascular Disease, CH4070 Basel, Switzerland
| | - Finian M Martin
- UCD Diabetes Complications Research Centre, School of Medicine, Conway Institute, University College Dublin, Belfield, Dublin 4, Ireland
| | - Catherine Godson
- UCD Diabetes Complications Research Centre, School of Medicine, Conway Institute, University College Dublin, Belfield, Dublin 4, Ireland
| |
Collapse
|
12
|
Yung S, Yap DYH, Chan TM. Recent advances in the understanding of renal inflammation and fibrosis in lupus nephritis. F1000Res 2017; 6:874. [PMID: 28663794 PMCID: PMC5473406 DOI: 10.12688/f1000research.10445.1] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/12/2017] [Indexed: 01/08/2023] Open
Abstract
Lupus nephritis is a potentially reversible cause of severe acute kidney injury and is an important cause of end-stage renal failure in Asians and patients of African or Hispanic descent. It is characterized by aberrant exaggerated innate and adaptive immune responses, autoantibody production and their deposition in the kidney parenchyma, triggering complement activation, activation and proliferation of resident renal cells, and expression of pro-inflammatory and chemotactic molecules leading to the influx of inflammatory cells, all of which culminate in destruction of normal nephrons and their replacement by fibrous tissue. Anti-double-stranded DNA (anti-dsDNA) antibody level correlates with disease activity in most patients. There is evidence that apart from mediating pathogenic processes through the formation of immune complexes, pathogenic anti-dsDNA antibodies can bind to resident renal cells and induce downstream pro-apoptotic, pro-inflammatory, or pro-fibrotic processes or a combination of these. Recent data also highlight the critical role of macrophages in acute and chronic kidney injury. Though clinically effective, current treatments for lupus nephritis encompass non-specific immunosuppression and the anti-inflammatory action of high-dose corticosteroids. The clinical and histological impact of novel biologics targeting pro-inflammatory molecules remains to be investigated. Insight into the underlying mechanisms that induce inflammatory and fibrotic processes in the kidney of lupus nephritis could present opportunities for more specific novel treatment options to improve clinical outcomes while minimizing off-target untoward effects. This review discusses recent advances in the understanding of pathogenic mechanisms leading to inflammation and fibrosis of the kidney in lupus nephritis in the context of established standard-of-care and emerging therapies.
Collapse
Affiliation(s)
- Susan Yung
- Department of Medicine, University of Hong Kong, Hong Kong, Hong Kong
| | - Desmond YH Yap
- Department of Medicine, University of Hong Kong, Hong Kong, Hong Kong
| | - Tak Mao Chan
- Department of Medicine, University of Hong Kong, Hong Kong, Hong Kong
| |
Collapse
|
13
|
Hertig V, Tardif K, Meus MA, Duquette N, Villeneuve L, Toussaint F, Ledoux J, Calderone A. Nestin expression is upregulated in the fibrotic rat heart and is localized in collagen-expressing mesenchymal cells and interstitial CD31(+)- cells. PLoS One 2017; 12:e0176147. [PMID: 28448522 PMCID: PMC5407835 DOI: 10.1371/journal.pone.0176147] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Accepted: 04/05/2017] [Indexed: 11/19/2022] Open
Abstract
Renal and lung fibrosis was characterized by the accumulation of collagen-immunoreactive mesenchymal cells expressing the intermediate filament protein nestin. The present study tested the hypothesis that nestin expression was increased in the hypertrophied/fibrotic left ventricle of suprarenal abdominal aorta constricted adult male Sprague-Dawley rats and induced in ventricular fibroblasts by pro-fibrotic peptide growth factors. Nestin protein levels were upregulated in the pressure-overloaded left ventricle and expression positively correlated with the rise of mean arterial pressure. In sham and pressure-overloaded hearts, nestin immunoreactivity was detected in collagen type I(+)-and CD31(+)-cells identified in the interstitium and perivascular region whereas staining was absent in smooth muscle α-actin(+)-cells. A significantly greater number of collagen type I(+)-cells co-expressing nestin was identified in the left ventricle of pressure-overloaded rats. Moreover, an accumulation of nestin(+)-cells lacking collagen, CD31 and smooth muscle α-actin staining was selectively observed at the adventitial region of predominantly large calibre blood vessels in the hypertrophied/fibrotic left ventricle. Angiotensin II and TGF-β1 stimulation of ventricular fibroblasts increased nestin protein levels via phosphatidylinositol 3-kinase- and protein kinase C/SMAD3-dependent pathways, respectively. CD31/eNOS(+)-rat cardiac microvascular endothelial cells synthesized/secreted collagen type I, expressed prolyl 4-hydroxylase and TGF-β1 induced nestin expression. The selective accumulation of adventitial nestin(+)-cells highlighted a novel feature of large vessel remodelling in the pressure-overloaded heart and increased appearance of collagen type I/nestin(+)-cells may reflect an activated phenotype of ventricular fibroblasts. CD31/collagen/nestin(+)-interstitial cells could represent displaced endothelial cells displaying an unmasked mesenchymal phenotype, albeit contribution to the reactive fibrotic response of the pressure-overloaded heart remains unknown.
Collapse
Affiliation(s)
- Vanessa Hertig
- Research Center, Montreal Heart Institute and Université de Montréal, Montréal, Québec, Canada
| | - Kim Tardif
- Research Center, Montreal Heart Institute and Université de Montréal, Montréal, Québec, Canada
| | - Marc Andre Meus
- Research Center, Montreal Heart Institute and Université de Montréal, Montréal, Québec, Canada
| | - Natacha Duquette
- Research Center, Montreal Heart Institute and Université de Montréal, Montréal, Québec, Canada
| | - Louis Villeneuve
- Research Center, Montreal Heart Institute and Université de Montréal, Montréal, Québec, Canada
| | - Fanny Toussaint
- Research Center, Montreal Heart Institute and Université de Montréal, Montréal, Québec, Canada
- Department of Pharmacology & Physiology, Université de Montréal, Québec, Montréal, Canada
| | - Jonathan Ledoux
- Research Center, Montreal Heart Institute and Université de Montréal, Montréal, Québec, Canada
- Department of Medicine, Université de Montréal, Québec, Montréal, Canada
| | - Angelino Calderone
- Research Center, Montreal Heart Institute and Université de Montréal, Montréal, Québec, Canada
- Department of Pharmacology & Physiology, Université de Montréal, Québec, Montréal, Canada
- * E-mail:
| |
Collapse
|
14
|
Peters M, Köhler-Bachmann S, Lenz-Habijan T, Bufe A. Influence of an Allergen-Specific Th17 Response on Remodeling of the Airways. Am J Respir Cell Mol Biol 2016. [PMID: 26222011 DOI: 10.1165/rcmb.2014-0429oc] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
We showed previously that sensitization of mice with dendritic cells (DCs) via the airways depends on activation of these cells with LPS. Allergen-pulsed DCs that were stimulated with low doses of LPS induce a strong Th2 response in vivo. Our objective was to investigate whether airway sensitization of mice by the application of DCs with a phenotype that is able to induce Th17 cells results in increased remodeling of the airways. We generated DCs from the bone marrow of mice and pulsed them with LPS-free ovalbumin. Subsequently, cells were activated with LPS with or without ATP for inflammasome activation. The activated cells were used to sensitize mice via the airways. Intranasal instillation of DCs that were activated with 0.1 ng/ml LPS induced a Th2 response with airway eosinophilia. High doses of LPS, particularly when given in combination with ATP, led to induction of a mixed Th2/Th17 response. Interestingly, we found a correlation between IL-17A production and the remodeling of the airways. Stimulation of mouse fibroblasts with purified IL-17A protein in vitro resulted in transforming growth factor-β1 secretion and collagen transcription. Interestingly, we found enhanced secretion of transforming growth factor-β1 by fibroblasts after costimulation with IL-17A and the profibrotic factor wingless-type MMTV integration site family, member 5A (Wnt5a). We showed that an allergen-specific Th17 response in the airway is accompanied by increased airway remodeling. Furthermore, we revealed that increased remodeling is not only based on neutrophilic inflammation, but also on the direct impact of IL-17A on airway structural cells.
Collapse
Affiliation(s)
- Marcus Peters
- Department of Experimental Pneumology, Ruhr University Bochum, Bochum, Germany
| | | | - Tim Lenz-Habijan
- Department of Experimental Pneumology, Ruhr University Bochum, Bochum, Germany
| | - Albrecht Bufe
- Department of Experimental Pneumology, Ruhr University Bochum, Bochum, Germany
| |
Collapse
|
15
|
Mattoli S. Involvement of fibrocytes in asthma and clinical implications. Clin Exp Allergy 2016; 45:1497-509. [PMID: 25752439 DOI: 10.1111/cea.12525] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Bloodborne fibrocytes are bone marrow-derived cells that participate in immune responses and exhibit pro-inflammatory and matrix remodelling properties. In patients with asthma receiving an adequate treatment, the blood fibrocyte count is very low and comparable to that obtained in healthy individuals. In these patients, a transient increase in fibrocyte numbers in the peripheral blood and in the airways occurs in concomitance with increased bronchial inflammation and reflects disease worsening and the need for more intensive treatment. Persistently elevated numbers of fibrocytes in the peripheral blood and in the bronchial mucosa are observed in chronically undertreated or corticosteroid-resistant asthma and are associated with persistent airway inflammation and ongoing remodelling of the bronchial wall. The asthmatic bronchial epithelium is the main source of fibrocyte chemoattractants in asthma and contributes with T helper type 2 lymphocytes and eosinophils to promote the proliferation and pro-remodelling function of recruited fibrocytes. The presence of elevated numbers of fibrocytes in the bronchial mucosa of allergic patients with undertreated or treatment-resistant asthma may also increase the risk of acute exacerbations because these cells can amplify T helper type 2 lymphocyte-driven inflammation on every exposure to the clinically relevant allergen and can promote further inflammation on rhinovirus infections by allowing viral replication and releasing additional pro-inflammatory factors. Improved methods for the isolation and functional analysis of pure populations of viable circulating fibrocytes have allowed a better understanding of the effector role of these cells. A reliable and clinically applicable assay has been developed to measure blood fibrocyte counts as outcome measure in future clinical trials. New therapeutic agents are needed to block both persistent inflammation and fibrocytosis in corticosteroid-resistant asthma.
Collapse
Affiliation(s)
- S Mattoli
- Avail Biomedical Research Institute, Scientific Direction and Project Management Centre, Basel, Switzerland
| |
Collapse
|
16
|
Chabot A, Hertig V, Boscher E, Nguyen QT, Boivin B, Chebli J, Bissonnette E, Villeneuve L, Brochiero E, Dupuis J, Calderone A. Endothelial and Epithelial Cell Transition to a Mesenchymal Phenotype Was Delineated by Nestin Expression. J Cell Physiol 2015; 231:1601-10. [DOI: 10.1002/jcp.25257] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2015] [Accepted: 11/16/2015] [Indexed: 12/21/2022]
Affiliation(s)
- Andréanne Chabot
- Montreal Heart Institute; Université de Montréal; Montréal Québec Canada
- Département de Physiologie Moléculaire et Intégrative; Université de Montréal; Montréal Quebéc Canada
| | - Vanessa Hertig
- Montreal Heart Institute; Université de Montréal; Montréal Québec Canada
- Département de Physiologie Moléculaire et Intégrative; Université de Montréal; Montréal Quebéc Canada
| | - Elena Boscher
- Montreal Heart Institute; Université de Montréal; Montréal Québec Canada
| | - Quang Trinh Nguyen
- Montreal Heart Institute; Université de Montréal; Montréal Québec Canada
| | - Benoît Boivin
- Montreal Heart Institute; Université de Montréal; Montréal Québec Canada
- Département de Biochimie et; Montréal Québec Canada
- Médecine; Université de Montréal; Montréal Québec Canada
| | | | - Elyse Bissonnette
- Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec; Département de Médicine; Université Laval; Québec Canada
| | - Louis Villeneuve
- Montreal Heart Institute; Université de Montréal; Montréal Québec Canada
| | | | - Jocelyn Dupuis
- Montreal Heart Institute; Université de Montréal; Montréal Québec Canada
- Médecine; Université de Montréal; Montréal Québec Canada
| | - Angelino Calderone
- Montreal Heart Institute; Université de Montréal; Montréal Québec Canada
- Département de Physiologie Moléculaire et Intégrative; Université de Montréal; Montréal Quebéc Canada
| |
Collapse
|
17
|
Mia MM, Bank RA. The pro-fibrotic properties of transforming growth factor on human fibroblasts are counteracted by caffeic acid by inhibiting myofibroblast formation and collagen synthesis. Cell Tissue Res 2015; 363:775-89. [PMID: 26453399 PMCID: PMC4761014 DOI: 10.1007/s00441-015-2285-6] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Accepted: 08/26/2015] [Indexed: 01/18/2023]
Abstract
Fibrosis is a chronic disorder affecting many organs. A universal process in fibrosis is the formation of myofibroblasts and the subsequent collagen deposition by these cells. Transforming growth factor beta1 (TGFβ1) plays a major role in the formation of myofibroblasts, e.g. by activating fibroblasts. Currently, no treatments are available to circumvent fibrosis. Caffeic acid phenethyl ester (CAPE) shows a broad spectrum of biological activities, including anti-fibrotic properties in vivo in mice and rats. However, little is known about the direct effects of CAPE on fibroblasts. We have tested whether CAPE is able to suppress myofibroblast formation and collagen formation of human dermal and lung fibroblasts exposed to TGFβ1, and found that this was indeed the case. In fact, the formation of myofibroblasts by TGFβ1 and subsequent collagen formation was completely abolished by CAPE. The same was observed for fibronectin and tenascin C. The lack of myofibroblast formation is likely due to the suppression of GLI1 and GLI2 expression by CAPE because of diminished nuclear SMAD2/3 levels. Post-treatment with CAPE after myofibroblast formation even resulted in a partial reversal of myofibroblasts into fibroblasts and/or reduction in collagen formation. Major discrepancies were seen between mRNA levels of collagen type I and cells stained positive for collagen, underlining the need for protein data in fibrosis studies to make reliable conclusions.
Collapse
Affiliation(s)
- Masum M Mia
- Department of Pathology & Medical Biology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ, Groningen, The Netherlands
| | - Ruud A Bank
- Department of Pathology & Medical Biology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ, Groningen, The Netherlands.
| |
Collapse
|
18
|
Mia MM, Bank RA. The IκB kinase inhibitor ACHP strongly attenuates TGFβ1-induced myofibroblast formation and collagen synthesis. J Cell Mol Med 2015; 19:2780-92. [PMID: 26337045 PMCID: PMC4687706 DOI: 10.1111/jcmm.12661] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2015] [Accepted: 06/23/2015] [Indexed: 01/07/2023] Open
Abstract
Excessive accumulation of a collagen‐rich extracellular matrix (ECM) by myofibroblasts is a characteristic feature of fibrosis, a pathological state leading to serious organ dysfunction. Transforming growth factor beta1 (TGFβ1) is a strong inducer of myofibroblast formation and subsequent collagen production. Currently, there are no remedies for the treatment of fibrosis. Activation of the nuclear factor kappa B (NF‐κB) pathway by phosphorylating IκB with the enzyme IκB kinase (IKK) plays a major role in the induction of fibrosis. ACHP {2‐Amino‐6‐[2‐(cyclopropylmethoxy)‐6‐hydroxyphenyl]‐4‐(4‐piperidinyl)‐3 pyridinecarbonitrile}, a selective inhibitor of IKK, prohibits the activation of the NF‐κB pathway. It is not known whether ACHP has potential anti‐fibrotic properties. Using adult human dermal and lung fibroblasts we have investigated whether ACHP has the ability to inhibit the TGFβ1‐induced transition of fibroblasts into myofibroblasts and its excessive synthesis of ECM. The presence of ACHP strongly suppressed the induction of the myofibroblast markers alpha‐smooth muscle actin (αSMA) and SM22α, as well as the deposition of the ECM components collagen type I and fibronectin. Furthermore, post‐treatment with ACHP partly reversed the expression of αSMA and collagen type I production. Finally, ACHP suppressed the expression of the three collagen‐modifying enzymes lysyl hydroxylase (PLOD1,PLOD2 and PLOD3) in dermal fibroblasts, but did not do so in lung fibroblasts. We conclude that the IKK inhibitor ACHP has potent antifibrotic properties, and that the NF‐κB pathway plays an important role in myofibroblast biology.
Collapse
Affiliation(s)
- Masum M Mia
- Department of Pathology and Medical Biology, Division of Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Ruud A Bank
- Department of Pathology and Medical Biology, Division of Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| |
Collapse
|
19
|
Luzina IG, Todd NW, Sundararajan S, Atamas SP. The cytokines of pulmonary fibrosis: Much learned, much more to learn. Cytokine 2015; 74:88-100. [DOI: 10.1016/j.cyto.2014.11.008] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2014] [Revised: 11/09/2014] [Accepted: 11/10/2014] [Indexed: 02/07/2023]
|
20
|
Boersema M, van den Born J, van Ark J, Harms G, Seelen M, van Dijk M, van Goor H, Navis G, Popa E, Hillebrands J. CD16+ monocytes with smooth muscle cell characteristics are reduced in human renal chronic transplant dysfunction. Immunobiology 2015; 220:673-83. [DOI: 10.1016/j.imbio.2014.11.011] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2014] [Revised: 11/07/2014] [Accepted: 11/13/2014] [Indexed: 11/17/2022]
|
21
|
|
22
|
Berardis S, Sattwika PD, Najimi M, Sokal EM. Use of mesenchymal stem cells to treat liver fibrosis: Current situation and future prospects. World J Gastroenterol 2015; 21:742-758. [PMID: 25624709 PMCID: PMC4299328 DOI: 10.3748/wjg.v21.i3.742] [Citation(s) in RCA: 99] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2014] [Revised: 09/05/2014] [Accepted: 11/19/2014] [Indexed: 02/06/2023] Open
Abstract
Progressive liver fibrosis is a major health issue for which no effective treatment is available, leading to cirrhosis and orthotopic liver transplantation. However, organ shortage is a reality. Hence, there is an urgent need to find alternative therapeutic strategies. Cell-based therapy using mesenchymal stem cells (MSCs) may represent an attractive therapeutic option, based on their immunomodulatory properties, their potential to differentiate into hepatocytes, allowing the replacement of damaged hepatocytes, their potential to promote residual hepatocytes regeneration and their capacity to inhibit hepatic stellate cell activation or induce their apoptosis, particularly via paracrine mechanisms. The current review will highlight recent findings regarding the input of MSC-based therapy for the treatment of liver fibrosis, from in vitro studies to pre-clinical and clinical trials. Several studies have shown the ability of MSCs to reduce liver fibrosis and improve liver function. However, despite these promising results, some limitations need to be considered. Future prospects will also be discussed in this review.
Collapse
|
23
|
Abstract
The common pathogenetic pathway of progressive injury in patients with chronic kidney disease (CKD) is epitomized as normal kidney parenchymal destruction due to scarring (fibrosis). Understanding the fundamental pathways that lead to renal fibrosis is essential in order to develop better therapeutic options for human CKD. Although complex, four cellular responses are pivotal. (1) An interstitial inflammatory response that has multiple consequences—some harmful and others healing. (2) The appearance of a unique interstitial cell population of myofibroblasts, primarily derived from kidney stromal cells (fibroblasts and pericytes), that are the primary source of the various extracellular matrix proteins that form interstitial scars. (3) Tubular epithelial cells that have variable and time-dependent roles as early responders to injury and later as victims of fibrosis due to the loss of their regenerative abilities. (4) Loss of interstitial capillary integrity that compromises oxygen delivery and leads to a vicious cascade of hypoxia–oxidant stress that accentuates injury and fibrosis. In the absence of adequate angiogenic responses, a healthy interstitial capillary network is not maintained. The fibrotic ‘scar' that typifies CKD is an interesting consortium of multifunctional macromolecules that not only change in composition and structure over time, but can be degraded via extracellular and intracellular proteases. Although transforming growth factor beta appears to be the primary driver of kidney fibrosis, a vast array of additional molecules may have modulating roles. The importance of genetic and epigenetic factors is increasingly appreciated. An intriguing but incompletely understood cardiorenal syndrome underlies the high morbidity and mortality rates that develop in association with progressive kidney fibrosis.
Collapse
|
24
|
Sargin AK, Can B, Turan B. Comparative investigation of kidney mesangial cells from increased oxidative stress-induced diabetic rats by using different microscopy techniques. Mol Cell Biochem 2013; 390:41-9. [PMID: 24374793 DOI: 10.1007/s11010-013-1953-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2013] [Accepted: 12/19/2013] [Indexed: 11/30/2022]
Abstract
High glucose and increased oxidative stress levels are the known important mediators of diabetic nephropathy. However, the effects of these mediators on tissue damage basically due to extracellular matrix expansion in mesangial cells have yet to be fully examined within the context of early stage diabetic nephropathy. In this study, we attempted to characterize changes in mesangial cells of streptozotocin-induced diabetic rats with a comparative investigation of kidney tissue by using different microscopy techniques. The serum levels of urea and creatinine of diabetic rats, as biomarkers of kidney degeneration, decreased significantly compared to those of age-matched controls. In diabetic rats, there are increased malondialdehyde and oxidized-glutathione levels as well as reduced-glutathione and glutathione-peroxidase activity levels in renal tissue compared to those of the controls. By using light and electron microscopies, we showed that there were marked thickening in Bowman's membrane and glomerular capillary wall, increased amount of extracellular matrix often occupying Bowman's space, degenerations in tubules, an increased number of mesangial cells in the network of glomerular capillary walls, and increased amount of lipid accumulation in proximal tubules in the renal tissue of diabetic rats. Our confocal microscopy data confirmed also the presence of irregularity and widened in glomerular capillaries, their attachment to the Bowman's capsule, degenerated heterochromatin, thickening in foci of glomerular basement membrane, and marked increase in mesangial cells. These results suggest that a detailed structural investigation of kidney tissue provides further information on the important role of mesangial cells in pathogenesis of diabetic nephropathy.
Collapse
Affiliation(s)
- Ayse Kose Sargin
- Department of Histology-Embriology, Faculty of Medicine, Ankara University, Ankara, Turkey
| | | | | |
Collapse
|
25
|
Cieslik KA, Trial J, Crawford JR, Taffet GE, Entman ML. Adverse fibrosis in the aging heart depends on signaling between myeloid and mesenchymal cells; role of inflammatory fibroblasts. J Mol Cell Cardiol 2013; 70:56-63. [PMID: 24184998 DOI: 10.1016/j.yjmcc.2013.10.017] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2013] [Revised: 10/02/2013] [Accepted: 10/22/2013] [Indexed: 11/18/2022]
Abstract
Aging has been associated with adverse fibrosis. Here we formulate a new hypothesis and present new evidence that unresponsiveness of mesenchymal stem cells (MSC) and fibroblasts to transforming growth factor beta (TGF-β), due to reduced expression of TGF-β receptor I (TβRI), provides a foundation for cardiac fibrosis in the aging heart via two mechanisms. 1) TGF-β promotes expression of Nanog, a transcription factor that retains MSC in a primitive state. In MSC derived from the aging heart, Nanog expression is reduced and therefore MSC gradually differentiate and the number of mesenchymal fibroblasts expressing collagen increases. 2) As TGF-β signaling pathway components negatively regulate transcription of monocyte chemoattractant protein-1 (MCP-1), a reduced expression of TβRI prevents aging mesenchymal cells from shutting down their own MCP-1 expression. Elevated MCP-1 levels that originated from MSC attract transendothelial migration of mononuclear leukocytes from blood to the tissue. MCP-1 expressed by mesenchymal fibroblasts promotes further migration of monocytes and T lymphocytes away from the endothelial barrier and supports the monocyte transition into macrophages and finally into myeloid fibroblasts. Both myeloid and mesenchymal fibroblasts contribute to fibrosis in the aging heart via collagen synthesis. This article is part of a Special Issue entitled "Myocyte-Fibroblast Signalling in Myocardium ".
Collapse
Affiliation(s)
- Katarzyna A Cieslik
- Division of Cardiovascular Sciences and the DeBakey Heart Center, Department of Medicine, Baylor College of Medicine, and Houston Methodist, Houston, TX 77030, USA.
| | - JoAnn Trial
- Division of Cardiovascular Sciences and the DeBakey Heart Center, Department of Medicine, Baylor College of Medicine, and Houston Methodist, Houston, TX 77030, USA
| | - Jeffrey R Crawford
- Division of Cardiovascular Sciences and the DeBakey Heart Center, Department of Medicine, Baylor College of Medicine, and Houston Methodist, Houston, TX 77030, USA
| | - George E Taffet
- Division of Cardiovascular Sciences and the DeBakey Heart Center, Department of Medicine, Baylor College of Medicine, and Houston Methodist, Houston, TX 77030, USA
| | - Mark L Entman
- Division of Cardiovascular Sciences and the DeBakey Heart Center, Department of Medicine, Baylor College of Medicine, and Houston Methodist, Houston, TX 77030, USA.
| |
Collapse
|
26
|
|
27
|
Usuki J, Matsuda K, Azuma A, Kudoh S, Gemma A. Sequential analysis of myofibroblast differentiation and transforming growth factor-β1/Smad pathway activation in murine pulmonary fibrosis. J NIPPON MED SCH 2013; 79:46-59. [PMID: 22398790 DOI: 10.1272/jnms.79.46] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Myofibroblasts play a critical role in tissue fibrosis. However, the intracellular signaling pathways in myofibroblast differentiation are poorly understood. Here, we studied the relationship between transforming growth factor-β (TGF-β)/Smad pathway activation and myofibroblast differentiation in both in vivo and in vitro experiments. In murine bleomycin-induced pulmonary fibrosis, nuclear localization of phosphorylated Smad2/3 (p-Smad2/3) was observed in pulmonary fibrotic lesions 7 days after bleomycin injection, whereas α-smooth muscle actin (ASMA)-positive myofibroblasts appeared in the lesions at 14 days, when the cytoplasmic localization of p-Smad2/3 was observed. We also compared the effects of TGF-β1 on myofibroblast differentiation and on type I collagen expression in a murine lung fibroblast cell line (MLg2908). TGF-β1 induced rapid expression of p-Smad2/3 in nuclei, after which ASMA organization in the cytoplasm of fibroblasts was observed. However, TGF-β1 produced no effect on the quantity of ASMA, either in mRNA levels or protein levels, even after the phosphorylation of Smad2/3. In contrast, TGF-β1 upregulated the expression of type I collagen mRNA. These findings suggest that in pulmonary fibrosis the molecular mechanism of myofibroblast differentiation is complex and that the difference between ASMA expression and type I collagen expression is mediated by the TGF-β/Smad pathway.
Collapse
Affiliation(s)
- Jiro Usuki
- Division of Pulmonary Medicine, Infection Diseases and Oncology, Graduate School of Medicine, Nippon Medical School, Tokyo, Japan.
| | | | | | | | | |
Collapse
|
28
|
Papke CL, Cao J, Kwartler CS, Villamizar C, Byanova KL, Lim SM, Sreenivasappa H, Fischer G, Pham J, Rees M, Wang M, Chaponnier C, Gabbiani G, Khakoo AY, Chandra J, Trache A, Zimmer W, Milewicz DM. Smooth muscle hyperplasia due to loss of smooth muscle α-actin is driven by activation of focal adhesion kinase, altered p53 localization and increased levels of platelet-derived growth factor receptor-β. Hum Mol Genet 2013; 22:3123-37. [PMID: 23591991 DOI: 10.1093/hmg/ddt167] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Mutations in ACTA2, encoding the smooth muscle cell (SMC)-specific isoform of α-actin (α-SMA), cause thoracic aortic aneurysms and dissections and occlusive vascular diseases, including early onset coronary artery disease and stroke. We have shown that occlusive arterial lesions in patients with heterozygous ACTA2 missense mutations show increased numbers of medial or neointimal SMCs. The contribution of SMC hyperplasia to these vascular diseases and the pathways responsible for linking disruption of α-SMA filaments to hyperplasia are unknown. Here, we show that the loss of Acta2 in mice recapitulates the SMC hyperplasia observed in ACTA2 mutant SMCs and determine the cellular pathways responsible for SMC hyperplasia. Acta2(-/-) mice showed increased neointimal formation following vascular injury in vivo, and SMCs explanted from these mice demonstrated increased proliferation and migration. Loss of α-SMA induced hyperplasia through focal adhesion (FA) rearrangement, FA kinase activation, re-localization of p53 from the nucleus to the cytoplasm and increased expression and ligand-independent activation of platelet-derived growth factor receptor beta (Pdgfr-β). Disruption of α-SMA in wild-type SMCs also induced similar cellular changes. Imatinib mesylate inhibited Pdgfr-β activation and Acta2(-/-) SMC proliferation in vitro and neointimal formation with vascular injury in vivo. Loss of α-SMA leads to SMC hyperplasia in vivo and in vitro through a mechanism involving FAK, p53 and Pdgfr-β, supporting the hypothesis that SMC hyperplasia contributes to occlusive lesions in patients with ACTA2 missense mutations.
Collapse
Affiliation(s)
- Christina L Papke
- Department of Internal Medicine, University of Texas Health Science Center at Houston, 6431 Fannin, MSB 6.100, Houston, TX 77030, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Cieslik KA, Trial J, Carlson S, Taffet GE, Entman ML. Aberrant differentiation of fibroblast progenitors contributes to fibrosis in the aged murine heart: role of elevated circulating insulin levels. FASEB J 2013; 27:1761-71. [PMID: 23303205 DOI: 10.1096/fj.12-220145] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
With age, the collagen content of the heart increases, leading to interstitial fibrosis. We have shown that CD44(pos) fibroblasts derived from aged murine hearts display reduced responsiveness to TGF-β but, paradoxically, have increased collagen expression in vivo and in vitro. We postulated that this phenomenon was due to the defect in mesenchymal stem cell (MSC) differentiation in a setting of elevated circulating insulin levels and production that we observed in aging mice. We discovered that cultured fibroblasts derived from aged but not young cardiac MSCs of nonhematopoietic lineage displayed increased basal and insulin-induced (1 nM) collagen expression (2-fold), accompanied by increased farnesyltransferase (FTase) and Erk activities. In a quest for a possible mechanism, we found that a chronic pathophysiologic insulin concentration (1 nM) caused abnormal fibroblast differentiation of MSCs isolated from young hearts. Fibroblasts derived from these MSCs responded to insulin by elevating collagen expression as seen in untreated aged fibroblast cultures, suggesting a causal link between increased insulin levels and defective MSC responses. Here we report an insulin-dependent pathway that specifically targets collagen type I transcriptional activation leading to a unique mechanism of fibrosis that is TGF-β and inflammation-independent in the aged heart.
Collapse
Affiliation(s)
- Katarzyna A Cieslik
- Baylor College of Medicine, Department of Medicine, Division of Cardiovascular Sciences, One Baylor Plaza, Mail Station BCM620, Houston, Texas 77030, USA.
| | | | | | | | | |
Collapse
|
30
|
Lin J, Wang C, Redies C. Expression of delta-protocadherins in the spinal cord of the chicken embryo. J Comp Neurol 2012; 520:1509-31. [PMID: 22102158 DOI: 10.1002/cne.22808] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Protocadherins constitute the largest subfamily of cadherin genes and are widely expressed in the nervous system. In the present study, we cloned eight members of the delta-protocadherin subfamily of cadherins (Pcdh1, Pcdh7, Pcdh8, Pcdh9, Pcdh10, Pcdh17, Pcdh18, and Pcdh19) from the chicken, and investigated their expression in the developing chicken spinal cord by in situ hybridization. Our results showed that each of the investigated delta-protocadherins exhibits a spatially restricted and temporally regulated pattern of expression. Pcdh1, Pcdh8, Pcdh18, and Pcdh19 are expressed in restricted dorsoventral domains of the neuroepithelial layer at early developmental stages (E2.5–E4). In the differentiating mantle layer, specific expression profiles are observed for all eight delta-protocadherins along the dorsoventral, mediolateral, and rostrocaudal dimensions at intermediate stages of development (E6–E10). Expression profiles are especially diverse in the motor column, where different pools of motor neurons exhibit signal for subsets of delta-protocadherins. In the dorsal root ganglion, subpopulations of cells express combinations of Pcdh1, Pcdh7, Pcdh8, Pcdh9, Pcdh10, and Pcdh17. The ventral boundary cap cells are positive for Pcdh7, Pcdh9, and Pcdh10. Signals for Pcdh8, Pcdh18, and Pcdh19 are found in the meninges. Surrounding tissues, such as the notochord, dermomyotome, and sclerotome also exhibit differential expression patterns. The highly regulated spatiotemporal expression patterns of delta-protocadherins suggest that they have multiple and diverse functions during development of the spinal cord and its surrounding tissues.
Collapse
Affiliation(s)
- Juntang Lin
- Institute of Anatomy I, University of Jena School of Medicine, Jena University Hospital, D-07743 Jena, Germany
| | | | | |
Collapse
|
31
|
Grabias BM, Konstantopoulos K. Epithelial-mesenchymal transition and fibrosis are mutually exclusive reponses in shear-activated proximal tubular epithelial cells. FASEB J 2012; 26:4131-41. [PMID: 22744866 DOI: 10.1096/fj.12-207324] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Renal fibrosis (RF) is thought to be a direct consequence of dedifferentiation of resident epithelial cells via an epithelial-mesenchymal transition (EMT). Increased glomerular flow is a critical initiator of fibrogenesis. Yet, the responses of proximal tubular epithelial cells (PTECs) to fluid flow remain uncharacterized. Here, we investigate the effects of pathological shear stresses on the development of fibrosis in PTECs. Our data reveal that type I collagen accumulation in shear-activated PTECs is accompanied by a ∼40-60% decrease in cell motility, thus excluding EMT as a relevant pathological process. In contrast, static incubation of PTECs with TGFβ1 increases cell motility by ∼50%, and induces stable expression of key mesenchymal markers, including Snail1, N-cadherin, and vimentin. Ectopic expression of TGFβ1 in shear-activated PTECs fails to induce EMT-associated changes but abrogates collagen accumulation via SMAD2-dependent mechanisms. Shear-mediated inhibition of EMT occurs via cyclic oscillations in both ERK2 activity and downstream expression of EMT genes. A constitutive ERK2 mutant induces stable expression of Snail1, N-cadherin, and vimentin, and increases cell motility in shear-activated PTECs by 250% without concomitant collagen deposition. Collectively, our data reveal that RF not only occurs without EMT but also that these two responses represent mutually exclusive cell fates.
Collapse
Affiliation(s)
- Bryan M Grabias
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, New Engineering Bldg. 114, 3400 N. Charles St., Baltimore, MD 21218, USA
| | | |
Collapse
|
32
|
Pechkovsky DV, Prêle CM, Wong J, Hogaboam CM, McAnulty RJ, Laurent GJ, Zhang SSM, Selman M, Mutsaers SE, Knight DA. STAT3-Mediated Signaling Dysregulates Lung Fibroblast-Myofibroblast Activation and Differentiation in UIP/IPF. THE AMERICAN JOURNAL OF PATHOLOGY 2012; 180:1398-412. [DOI: 10.1016/j.ajpath.2011.12.022] [Citation(s) in RCA: 88] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/15/2011] [Revised: 11/28/2011] [Accepted: 12/06/2011] [Indexed: 12/20/2022]
|
33
|
Shardonofsky FR, Moore J, Schwartz RJ, Boriek AM. Airways in smooth muscle α-actin null mice experience a compensatory mechanism that modulates their contractile response. J Appl Physiol (1985) 2011; 112:898-903. [PMID: 22134689 DOI: 10.1152/japplphysiol.00417.2011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
We hypothesized that ablation of smooth muscle α-actin (SM α-A), a contractile-cytoskeletal protein expressed in airway smooth muscle (ASM) cells, abolishes ASM shortening capacity and decreases lung stiffness. In both SM α-A knockout and wild-type (WT) mice, airway resistance (Raw) determined by the forced oscillation technique rose in response to intravenous methacholine (Mch). However, the slope of Raw (cmH(2)O·ml(-1)·s) vs. log(2) Mch dose (μg·kg(-1)·min(-1)) was lower (P = 0.007) in mutant (0.54 ± 0.14) than in WT mice (1.23 ± 0.19). RT-PCR analysis performed on lung tissues confirmed that mutant mice lacked SM α-A mRNA and showed that these mice had robust expressions of both SM γ-A mRNA and skeletal muscle (SKM) α-A mRNA, which were not expressed in WT mice, and an enhanced SM22 mRNA expression relative to that in WT mice. Compared with corresponding spontaneously breathing mice, mechanical ventilation-induced lung mechanical strain increased the expression of SM α-A mRNA in WT lungs; in mutant mice, it augmented the expressions of SM γ-A mRNA and SM22 mRNA and did not alter that of SKM α-A mRNA. In mutant mice, the expression of SM γ-A mRNA in the lung during spontaneous breathing and its enhanced expression following mechanical ventilation are consistent with the likely possibility that in the absence of SM α-A, SM γ-A underwent polymerization and interacted with smooth muscle myosin to produce ASM shortening during cholinergic stimulation. Thus our data are consistent with ASM in mutant mice experiencing compensatory mechanisms that modulated its contractile muscle capacity.
Collapse
Affiliation(s)
- Felix R Shardonofsky
- F. R. Shardonofsky, Scott & White Children’s Hospital, 2401 31st St., MS 27-134, Temple, TX 76508, USA.
| | | | | | | |
Collapse
|
34
|
Abstract
Renal fibrosis, particularly tubulointerstitial fibrosis, is the common final outcome of almost all progressive chronic kidney diseases. Renal fibrosis is also a reliable predictor of prognosis and a major determinant of renal insufficiency. Irrespective of the initial causes, renal fibrogenesis is a dynamic and converging process that consists of four overlapping phases: priming, activation, execution and progression. Nonresolving inflammation after a sustained injury sets up the fibrogenic stage (priming) and triggers the activation and expansion of matrix-producing cells from multiple sources through diverse mechanisms, including activation of interstitial fibroblasts and pericytes, phenotypic conversion of tubular epithelial and endothelial cells and recruitment of circulating fibrocytes. Upon activation, matrix-producing cells assemble a multicomponent, integrin-associated protein complex that integrates input from various fibrogenic signals and orchestrates the production of matrix components and their extracellular assembly. Multiple cellular and molecular events, such as tubular atrophy, microvascular rarefaction and tissue hypoxia, promote scar formation and ensure a vicious progression to end-stage kidney failure. This Review outlines our current understanding of the cellular and molecular mechanisms of renal fibrosis, which could offer novel insights into the development of new therapeutic strategies.
Collapse
Affiliation(s)
- Youhua Liu
- Department of Pathology, University of Pittsburgh, S-405 Biomedical Science Tower, 200 Lothrop Street, Pittsburgh, PA 15261, USA.
| |
Collapse
|
35
|
Li J, Liu P, Zhang R, Cao L, Qian H, Liao J, Xu W, Wu M, Yin Z. Icaritin induces cell death in activated hepatic stellate cells through mitochondrial activated apoptosis and ameliorates the development of liver fibrosis in rats. JOURNAL OF ETHNOPHARMACOLOGY 2011; 137:714-23. [PMID: 21726622 DOI: 10.1016/j.jep.2011.06.030] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/26/2011] [Revised: 06/15/2011] [Accepted: 06/20/2011] [Indexed: 05/17/2023]
Abstract
AIM OF THE STUDY Icaritin is an active ingredient extracted from the plant Herba Epimedium Sagittatum (Sieb. et Zucc.) Maxim. The purpose of this study is to investigate the effects and mechanisms of icaritin-induced cell death in activated hepatic stellate cells (HSCs) and ameliorating the development of liver fibrosis in rats. MATERIALS AND METHODS : In vitro, icaritin-induced cell death rates in HSC-T6 (rat) and LX-2 (human) HSC lines as well as normal hepatocyte cell lines HL-7702 (L02) and WRL-68 were assayed by MTT method, and the apoptotic ratios were detected by both flow cytometry and the Annexin-V-FITC Apoptosis Detection Kit. A Whole Rat Genome Microarray Kit was used to identify expression of interest genes through fold-change screening. In vivo study, experimental liver fibrosis models were built by carbon tetrachloride (CCl(4)) or common bile duct ligation (CBDL) in Wistar rats. Icaritin (1mg/kg/day, three days a week) was administered by gastric gavage for four weeks (n=6 per group). At the end of the study, serum levels of aspartate aminotransferase (AST), alanine aminotransferase (ALT) as well as the contents of hydroxyproline and collagen I in liver tissues were measured. Histopathological changes and the distribution of activated HSCs were observed in the liver tissues using hematoxyline-eosin (HE) staining and immunohistochemical staining for α-smooth muscle actin (α-SMA). RESULTS Icaritin induced apoptosis in HSC-T6 and LX-2 in a concentration- and time-dependent manner with little toxicity to normal hepatocyte cell lines. The IC(50) of icaritin in HSC-T6 was 12.83 μM at 48 h. Apoptotic ratio of HSC-T6 treated with 24 μM icaritin was 20.19%, and the G2 phase of the cell cycle did not occur (P<0.05). Gene analysis showed that icaritin up-regulated Bak-1, Bmf and Bax expression while significantly down-regulated Bcl-2 expression (vs. control group, P<0.01). These results suggested that mitochondrial pathway played an important role in icaritin-induced apoptosis in activated HSCs. In vivo results showed that icaritin reduced the number of activated HSCs, and brought the elevated levels of AST, ALT, hydroxyproline and collagen I to normal or near normal values (vs. model group, P<0.05). CONCLUSIONS Icaritin can induce cell death in activated HSCs through mitochondria-mediated apoptosis, ameliorate the progression of hepatic fibrosis in rats, and could be a promising drug for treating liver fibrosis.
Collapse
Affiliation(s)
- Jing Li
- Molecular Oncology Laboratory, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Changhai Rd. 225, Shanghai 200438, PR China
| | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Dynamics of Sept4 expression in fibrotic livers of mice infected with Schistosoma japonicum. Parasitology 2011; 138:1003-10. [DOI: 10.1017/s0031182011000667] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
SUMMARYIn order to investigate the dynamics of Septin4 (Sept4) expression and its function in the formation of fibrotic livers in mice infected with Schistosoma japonicum, we constructed the mouse model of S. japonicum egg-induced liver fibrosis for 24 weeks. Immunohistochemical staining, qRT-PCR and Western blot were used to detect the expression of Sept4 and α-smooth muscle actin (α-SMA). We found Sept4 localized in the perisinusoidal space where hepatic stellate cells (HSCs) distribute in the periphery of circumoval granulomas and the portal venule. The expression of Sept4 and α-SMA had a similar significant tendency of an up-regulation to a peak at 12 weeks post-infection (p.i.) followed by a down-regulation. At 24 weeks p.i. both were at a low level. These results suggest that Sept4 and α-SMA may interact together in HSCs. Based on this evidence, we hypothesize that Sept4 seems to be involved in the formation of inflammatory granulomata and subsequent liver fibrosis by regulating HSCs activation.
Collapse
|
37
|
Abstract
Stromal fibroblasts are the primary cells of the kidney that produce fibrotic matrix. CD248 is a stromal marker expressed on fibroblasts and pericytes within the human kidney. Here, we tested whether CD248 expression in the kidney colocalizes with fibrosis and if it is associated with known determinants of chronic kidney disease (CKD). CD248 expression was located and quantified in situ by immunohistochemistry in kidney biopsies from 93 patients with IgA nephropathy and compared with 22 archived biopsies encompassing normal kidney tissue as control. In normal kidney tissue, CD248 was expressed by resident pericytes, stromal fibroblasts, and was upregulated in human CKD. The expression was linked to known determinants of renal progression. This relationship was maintained in a multivariate analysis with CD248 expression linked to renal survival. CD248 was expressed by a population of α-smooth muscle actin (SMA)(+) myofibroblasts and α-SMA(-) stromal cells but not expressed on CD45(+) leukocytes. Thus, CD248 defines a subset of stromal cells, including but not limited to some myofibroblasts, linked to albuminuria and tubulointerstitial damage during tissue remodeling in CKD.
Collapse
|
38
|
Abstract
Progression of fibrosis involves interstitial hypercellularity, matrix accumulation, and atrophy of epithelial structures, resulting in loss of normal function and ultimately organ failure. There is common agreement that the fibroblast/myofibroblast is the cell type most responsible for interstitial matrix accumulation and consequent structural deformations associated with fibrosis. During wound healing and progressive fibrotic events, fibroblasts transform into myofibroblasts acquiring smooth muscle features, most notably the expression of alpha-smooth muscle actin and synthesis of mesenchymal cell-related matrix proteins. In renal disease, glomerular mesangial cells also acquire a myofibroblast phenotype and synthesize the same matrix proteins. The origin of interstitial myofibroblasts during fibrosis is a matter of debate, where the cells are proposed to derive from resident fibroblasts, pericytes, perivascular adventitial, epithelial, and/or endothelial sources. Regardless of the origin of the cells, transforming growth factor-beta1 (TGF-β1) is the principal growth factor responsible for myofibroblast differentiation to a profibrotic phenotype and exerts its effects via Smad signaling pathways involving mitogen-activated protein kinase and Akt/protein kinase B. Additionally, reactive oxygen species (ROS) have important roles in progression of fibrosis. ROS are derived from a variety of enzyme sources, of which the nicotinamide adenine dinucleotide phosphate (NAD(P)H) oxidase family has been identified as a major source of superoxide and hydrogen peroxide generation in the cardiovasculature and kidney during health and disease. Recent evidence indicates that the NAD(P)H oxidase homolog Nox4 is most accountable for ROS-induced fibroblast and mesangial cell activation, where it has an essential role in TGF-β1 signaling of fibroblast activation and differentiation into a profibrotic myofibroblast phenotype and matrix production. Information on the role of ROS in mesangial cell and fibroblast signaling is incomplete, and further research on myofibroblast differentiation during fibrosis is warranted.
Collapse
|
39
|
Borensztajn K, Bresser P, van der Loos C, Bot I, van den Blink B, den Bakker MA, Daalhuisen J, Groot AP, Peppelenbosch MP, von der Thüsen JH, Spek CA. Protease-activated receptor-2 induces myofibroblast differentiation and tissue factor up-regulation during bleomycin-induced lung injury: potential role in pulmonary fibrosis. THE AMERICAN JOURNAL OF PATHOLOGY 2010; 177:2753-64. [PMID: 20971733 DOI: 10.2353/ajpath.2010.091107] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Idiopathic pulmonary fibrosis constitutes the most devastating form of fibrotic lung disorders and remains refractory to current therapies. The coagulation cascade is frequently activated during pulmonary fibrosis, but this observation has so far resisted a mechanistic explanation. Recent data suggest that protease-activated receptor (PAR)-2, a receptor activated by (among others) coagulation factor (F)Xa, plays a key role in fibrotic disease; consequently, we assessed the role of PAR-2 in the development of pulmonary fibrosis in this study. We show that PAR-2 is up-regulated in the lungs of patients with idiopathic pulmonary fibrosis and that bronchoalveolar lavage fluid from these patients displays increased procoagulant activity that triggers fibroblast survival. Using a bleomycin model of pulmonary fibrosis, we show that bleomycin induces PAR-2 expression, as well as both myofibroblast differentiation and collagen synthesis. In PAR-2-/- mice, both the extent and severity of fibrotic lesions are reduced, whereas myofibroblast differentiation is diminished and collagen expression is decreased. Moreover, fibrin deposition in the lungs of fibrotic PAR-2-/- mice is reduced compared with wild-type mice due to differential tissue factor expression in response to bleomycin. Taken together, these results suggest an important role for PAR-2 in the development of pulmonary fibrosis, and the inhibition of the PAR-2-coagulation axis may provide a novel therapeutic approach to treat this devastating disease.
Collapse
Affiliation(s)
- Keren Borensztajn
- Center for Experimental and Molecular Medicine, Academic Medical Center, Meibergdreef 9, NL-1105 AZ, Amsterdam, The Netherlands.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Abstract
The pathologic paradigm for renal progression is advancing tubulointerstitial fibrosis. Whereas mechanisms underlying fibrogenesis have grown in scope and understanding in recent decades, effective human treatment to directly halt or even reverse fibrosis remains elusive. Here, we examine key features mediating the molecular and cellular basis of tubulointerstitial fibrosis and highlight new insights that may lead to novel therapies. How to prevent chronic kidney disease from progressing to renal failure awaits even deeper biochemical understanding.
Collapse
Affiliation(s)
- Michael Zeisberg
- Division of Matrix Biology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA.
| | | |
Collapse
|
41
|
Boor P, Ostendorf T, Floege J. Renal fibrosis: novel insights into mechanisms and therapeutic targets. Nat Rev Nephrol 2010; 6:643-56. [PMID: 20838416 DOI: 10.1038/nrneph.2010.120] [Citation(s) in RCA: 481] [Impact Index Per Article: 32.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Renal fibrosis is the common end point of virtually all progressive kidney diseases. Renal fibrosis should not be viewed as a simple and uniform 'scar', but rather as a dynamic system that involves extracellular matrix components and many, if not all, renal and infiltrating cell types. The involved cells exhibit enormous plasticity or phenotypic variability-a fact that we are only beginning to appreciate. Only a detailed understanding of the underlying mechanisms of renal fibrosis can facilitate the development of effective treatments. In this Review, we discuss the most recent advances in renal, or more specifically, tubulointerstitial fibrosis. Novel mechanisms as well as potential treatment targets based on different cell types are described. Problems that continue to plague the field are also discussed, including specific therapeutic targeting of the kidney, the development of improved diagnostic methods to assess renal fibrosis and the shortcomings of available animal models.
Collapse
Affiliation(s)
- Peter Boor
- Department of Nephrology, RWTH University of Aachen, Pauwelsstrasse 30, 52074 Aachen, Germany
| | | | | |
Collapse
|
42
|
Genetic variants promoting smooth muscle cell proliferation can result in diffuse and diverse vascular diseases: evidence for a hyperplastic vasculomyopathy. Genet Med 2010; 12:196-203. [PMID: 20130469 DOI: 10.1097/gim.0b013e3181cdd687] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Genetic predisposition to early onset of occlusive vascular diseases, including coronary artery disease, ischemic stroke, and Moyamoya disease, may represent varying presentations of a common underlying dysregulation of vascular smooth muscle cell proliferation. We discuss mutations in two genes, NF1 and ACTA2, which predispose affected individuals to diffuse and diverse vascular diseases. These patients show evidence of diffuse occlusive disease in multiple arterial beds or even develop seemingly diverse arterial pathologies, ranging from occlusions to arterial aneurysms. We also present the current evidence that both NF1 and ACTA2 mutations promote increased smooth muscle cell proliferation in vitro and in vivo, which leads us to propose that these diffuse and diverse vascular diseases are the outward signs of a more fundamental disease: a hyperplastic vasculomyopathy. We suggest that the concept of a hyperplastic vasculomyopathy offers a new approach not only to identifying mutated genes that lead to vascular diseases but also to counseling and possibly treating patients harboring such mutations. In other words, this framework may offer the opportunity to therapeutically target the inappropriate smooth muscle cell behavior that predisposes to a variety of vascular diseases throughout the arterial system.
Collapse
|
43
|
Lin A, Hokugo A, Nishimura I. Wound closure and wound management: A new therapeutic molecular target. Cell Adh Migr 2010; 4:396-9. [PMID: 20448469 DOI: 10.4161/cam.4.3.11917] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Wound closure and infection control are the primary goal of wound management. A variety of disinfectants and antimicrobial agents are widely available today and routinely achieve infection control. On the contrary, wound closure still remains a challenging goal. Cell adhesion, migration and contraction play significant roles in creating contractile force of patent wound margins and in contributing to wound closure. Modulations of these cellular behaviors have been investigated in the context of wound contraction; however, therapeutic strategy to achieve wound closure has not been established. Recently, we have reported that a previously unknown cytoskeleton molecule, wound inducible transcript-3.0 (wit3.0) also known as fibroblast growth factor receptor 1 oncogene partner 2 (FGFR1OP2), can significantly modulate fibroblast-driven wound closure in vitro and in vivo. The dynamic role of cytoskeleton in different experimental models may provide a novel platform for designing the therapeutic target of wound management.
Collapse
Affiliation(s)
- Audrey Lin
- The Weintraub Center for Reconstructive Biotechnology, UCLA School of Dentistry, Los Angeles, CA, USA
| | | | | |
Collapse
|
44
|
Yamada M, Oda T, Higashi K, Kushiyama T, Yamakami K, Sakurai Y, Hirai Y, Yamamoto K, Hyodo T, Suzuki S, Miura S, Kumagai H. Involvement of epimorphin in the repair of experimental renal fibrosis in mice. J Transl Med 2010; 90:867-80. [PMID: 20195239 DOI: 10.1038/labinvest.2010.50] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Interaction between epithelial cells and mesenchymal cells is essential in normal organ morphogenesis and in tissue repair after injury. Epimorphin, a mesenchymal protein that regulates epithelial morphogenesis through epithelial-mesenchymal interactions, has recently attracted attention as an important modulator of tissue repair. In this study we analyzed the role of epimorphin in renal fibrosis. We first found a progressive increase in epimorphin expression corresponding to the progression of renal fibrosis in mice with unilateral ureteral obstruction (UUO). To determine whether this expression has a role in the repair or progression of renal fibrosis, we analyzed a model of renal fibrosis repair, the UUO-release (UUO-R) model. Epimorphin expression was increased at 3 and 7 days after the UUO-R rather than on the day of release, but was decreased at 21 days after the release. Inhibition of endogenous epimorphin with anti-epimorphin antibody (MC-1) significantly delayed the repair of fibrosis. When compared with normal-IgG-injected mice, MC-1-injected mice showed significantly decreased renal matrix metalloproteinase (MMP)-2 and MMP-9 expressions by western blotting and increased expression of TGF-beta and collagen-I mRNA by real-time RT-PCR. Recombinant epimorphin induced prominent increases in MMP-2 and MMP-9 activities in the culture media of renal interstitial fibroblasts in vitro. These findings indicate that epimorphin has a pivotal role in the repair of renal fibrosis by modulating both extracellular matrix (ECM) degradation and its production.
Collapse
Affiliation(s)
- Muneharu Yamada
- Division of Nephrology, Department of Internal Medicine, Saitama, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Lin A, Hokugo A, Choi J, Nishimura I. Small cytoskeleton-associated molecule, fibroblast growth factor receptor 1 oncogene partner 2/wound inducible transcript-3.0 (FGFR1OP2/wit3.0), facilitates fibroblast-driven wound closure. THE AMERICAN JOURNAL OF PATHOLOGY 2009; 176:108-21. [PMID: 19959814 DOI: 10.2353/ajpath.2010.090256] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Wounds created in the oral cavity heal rapidly and leave minimal scarring. We have examined a role of a previously isolated cDNA from oral wounds encoding wound inducible transcript-3.0 (wit3.0), also known as fibroblast growth factor receptor 1 oncogene partner 2 (FGFR1OP2). FGFR1OP2/wit3.0 was highly expressed in oral wound fibroblasts without noticeable up-regulation of alpha-smooth muscle actin. In silico analyses, denaturing and nondenaturing gel Western blot, and immunocytology together demonstrated that FGFR1OP2/wit3.0 were able to dimerize and oligomerize through coiled-coil structures and appeared to associate with cytoskeleton networks in oral wound fibroblasts. Overexpression of FGFR1OP2/wit3.0 increased the floating collagen gel contraction of naïve oral fibroblasts to the level of oral wound fibroblasts, which was in turn attenuated by small-interfering RNA knockdown. The FGFR1OP2/wit3.0 synthesis did not affect the expression of collagen I as well as procontractile peptides such as alpha-smooth muscle actin, and transforming growth factor-beta1 had no effect on FGFR1OP2/wit3.0 expression. Fibroblastic cells derived from embryonic stem cells carrying FGFR1OP2/wit3.0 (+/-) mutation showed significant retardation in cell migration. Thus, we postulate that FGFR1OP2/wit3.0 may regulate cell motility and stimulate wound closure. FGFR1OP2/wit3.0 was not up-regulated during skin wound healing; however, when treated with FGFR1OP2/wit3.0 -expression vector, the skin wound closure was significantly accelerated, resulting in the limited granulation tissue formation. Our data suggest that FGFR1OP2/wit3.0 may possess a therapeutic potential for wound management.
Collapse
Affiliation(s)
- Audrey Lin
- The Weintraub Center for Reconstructive Biotechnology, University of California, Los Angeles, School of Dentistry, Los Angeles, CA 90095-1668, USA
| | | | | | | |
Collapse
|
46
|
Yoo KH, Thornhill BA, Forbes MS, Chevalier RL. Inducible nitric oxide synthase modulates hydronephrosis following partial or complete unilateral ureteral obstruction in the neonatal mouse. Am J Physiol Renal Physiol 2009; 298:F62-71. [PMID: 19889956 DOI: 10.1152/ajprenal.00234.2009] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
To investigate the role of endogenous inducible nitric oxide synthase (iNOS) in the response of the developing kidney to unilateral ureteral obstruction (UUO), neonatal iNOS null mutant (-/-) and wild-type (WT) mice were subjected to partial or complete UUO. At 7 and 21 days of age, apoptosis, renin, vascular endothelial growth factor (VEGF), fibroblasts (anti-fibroblast-specific peptide 1), myofibroblasts (alpha-smooth muscle actin), macrophages (F4/80), and collagen were measured in kidney tissue. Compared with WT, renal parenchymal thickness was increased, with preservation of the papilla, in -/- mice with partial UUO, but decreased in -/- mice with complete UUO. Ureteral peristalsis increased with severity of pelvic dilatation in WT, and increased further in -/- mice with partial UUO. Apoptosis, fibroblasts, and macrophages were increased in -/- mice with complete UUO, but there was no effect of iNOS on other histological parameters following complete UUO. Renin was decreased in -/- mice with partial UUO. There was no effect of iNOS genotype on renal collagen accumulation at either 7 or 21 days of age. These results are consistent with an injurious role for endogenous iNOS following partial UUO by inhibiting ureteral peristalsis and increasing renal renin although renal fibrosis is not affected. In contrast, in mice with complete UUO, iNOS attenuates apoptosis and enhances renal parenchymal thickness. Alterations in the severity of ureteral obstruction may therefore influence the effect of iNOS on long-term renal injury.
Collapse
Affiliation(s)
- Kee Hwan Yoo
- Department of Pediatrics, University of Virginia, Charlottesville, Virginia 22908, USA
| | | | | | | |
Collapse
|
47
|
Cunnington RH, Nazari M, Dixon IM. c-Ski, Smurf2, and Arkadia as regulators of TGF-β signaling: new targets for managing myofibroblast function and cardiac fibrosisThis article is one of a selection of papers published in a special issue celebrating the 125th anniversary of the Faculty of Medicine at the University of Manitoba. Can J Physiol Pharmacol 2009; 87:764-72. [DOI: 10.1139/y09-076] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Recent studies demonstrate the critical role of the extracellular matrix in the organization of parenchymal cells in the heart. Thus, an understanding of the modes of regulation of matrix production by cardiac myofibroblasts is essential. Transforming growth factor β (TGF-β) signaling is transduced through the canonical Smad pathway, and the involvement of this pathway in matrix synthesis and other processes requires precise control. Inhibition of Smad signaling may be achieved at the receptor level through the targeting of the TGF-β type I receptors with an inhibitory Smad7 / Smurf2 complex, or at the transcriptional level through c-Ski / receptor-Smad / co-mediator Smad4 interactions. Conversely, Arkadia protein intensifies TGF-β-induced effects by marking c-Ski and inhibitory Smad7 for destruction. The study of these TGF-β mediators is essential for future treatment of fibrotic disease, and this review highlights recent relevant findings that may impact our understanding of cardiac fibrosis.
Collapse
Affiliation(s)
- Ryan H. Cunnington
- Institute of Cardiovascular Sciences, St. Boniface General Hospital Research Centre, Department of Physiology, University of Manitoba, 351 Tache Avenue, Winnipeg, MB R2H 2A6, Canada
| | - Mansoreh Nazari
- Institute of Cardiovascular Sciences, St. Boniface General Hospital Research Centre, Department of Physiology, University of Manitoba, 351 Tache Avenue, Winnipeg, MB R2H 2A6, Canada
| | - Ian M.C. Dixon
- Institute of Cardiovascular Sciences, St. Boniface General Hospital Research Centre, Department of Physiology, University of Manitoba, 351 Tache Avenue, Winnipeg, MB R2H 2A6, Canada
| |
Collapse
|
48
|
Guo DC, Papke CL, Tran-Fadulu V, Regalado ES, Avidan N, Johnson RJ, Kim DH, Pannu H, Willing MC, Sparks E, Pyeritz RE, Singh MN, Dalman RL, Grotta JC, Marian AJ, Boerwinkle EA, Frazier LQ, LeMaire SA, Coselli JS, Estrera AL, Safi HJ, Veeraraghavan S, Muzny DM, Wheeler DA, Willerson JT, Yu RK, Shete SS, Scherer SE, Raman C, Buja LM, Milewicz DM. Mutations in smooth muscle alpha-actin (ACTA2) cause coronary artery disease, stroke, and Moyamoya disease, along with thoracic aortic disease. Am J Hum Genet 2009; 84:617-27. [PMID: 19409525 DOI: 10.1016/j.ajhg.2009.04.007] [Citation(s) in RCA: 377] [Impact Index Per Article: 23.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2009] [Revised: 03/30/2009] [Accepted: 04/09/2009] [Indexed: 11/25/2022] Open
Abstract
The vascular smooth muscle cell (SMC)-specific isoform of alpha-actin (ACTA2) is a major component of the contractile apparatus in SMCs located throughout the arterial system. Heterozygous ACTA2 mutations cause familial thoracic aortic aneurysms and dissections (TAAD), but only half of mutation carriers have aortic disease. Linkage analysis and association studies of individuals in 20 families with ACTA2 mutations indicate that mutation carriers can have a diversity of vascular diseases, including premature onset of coronary artery disease (CAD) and premature ischemic strokes (including Moyamoya disease [MMD]), as well as previously defined TAAD. Sequencing of DNA from patients with nonfamilial TAAD and from premature-onset CAD patients independently identified ACTA2 mutations in these patients and premature onset strokes in family members with ACTA2 mutations. Vascular pathology and analysis of explanted SMCs and myofibroblasts from patients harboring ACTA2 suggested that increased proliferation of SMCs contributed to occlusive diseases. These results indicate that heterozygous ACTA2 mutations predispose patients to a variety of diffuse and diverse vascular diseases, including TAAD, premature CAD, ischemic strokes, and MMD. These data demonstrate that diffuse vascular diseases resulting from either occluded or enlarged arteries can be caused by mutations in a single gene and have direct implications for clinical management and research on familial vascular diseases.
Collapse
|
49
|
Hewitson TD. Renal tubulointerstitial fibrosis: common but never simple. Am J Physiol Renal Physiol 2009; 296:F1239-44. [PMID: 19144691 DOI: 10.1152/ajprenal.90521.2008] [Citation(s) in RCA: 187] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Regardless of etiology, all patients with chronic renal disease show a progressive decline in renal function with time. Fibrosis, so-called scarring, is a key cause of this pathophysiology. Fibrosis involves an excess accumulation of extracellular matrix (primarily composed of collagen) and usually results in loss of function when normal tissue is replaced with scar tissue. While recent major advances have led to a much better understanding of this process, many problems remain. We for instance know little about why some wounds heal and others scar and little about how many putative antifibrotic agents work. This review discusses recent advances in our understanding of the mechanisms of tubulointerstitial fibrosis, focusing on the regulation and role of the myofibroblast in this process, the role of recently recognized endogenous antifibrotic factors, controversy surrounding the effects of metalloproteinases, and the opportunities presented by new treatment strategies that abrogate and may even reverse fibrosis.
Collapse
Affiliation(s)
- Tim D Hewitson
- Department of Nephrology, The Royal Melbourne Hospital, Parkville Vic 3050, Australia.
| |
Collapse
|
50
|
Ding Q, Gladson CL, Wu H, Hayasaka H, Olman MA. Focal adhesion kinase (FAK)-related non-kinase inhibits myofibroblast differentiation through differential MAPK activation in a FAK-dependent manner. J Biol Chem 2008; 283:26839-49. [PMID: 18669633 PMCID: PMC2556008 DOI: 10.1074/jbc.m803645200] [Citation(s) in RCA: 71] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2008] [Revised: 07/24/2008] [Indexed: 11/06/2022] Open
Abstract
Transforming growth factor (TGF)-beta1 induces fibroblast transdifferentiation to myofibroblasts, a process that requires the involvement of integrin-mediated signaling and focal adhesion kinase (FAK). FAK-related non-kinase (FRNK) is known for its role in inhibiting integrin-mediated cell migration; however, its role in myofibroblast differentiation has not been defined. Here, we report that FRNK abrogates TGF-beta1-induced myofibroblast differentiation in vitro and in vivo. TGF-beta1 can induce alpha-smooth muscle actin (alpha-SMA) expression in the presence or absence of FAK; however, TGF-beta1-induced alpha-SMA expression is reduced (approximately 73%) in FAK-deficient fibroblasts. Although both ERK and p38 MAPK activation is required for maximal TGF-beta1-induced alpha-SMA expression, ERK is the major signaling intermediate in cells that express FAK. In contrast, p38 MAPK is the dominant mediator of TGF-beta1-induced alpha-SMA expression in FAK-deficient cells. FRNK overexpression blocks TGF-beta1-induced ERK or p38 MAPK activation in the presence, and surprisingly, in the absence of FAK. The loss of FRNK was tested in vivo during experimentally induced pulmonary fibrosis in mice. FRNK knock-out mice have a greater increase in alpha-SMA-expressing cells in response to a pulmonary fibrotic stimulus in vivo, as compared with congenic wild type mice. This is the first time that FRNK loss has been shown to modify the pathobiology in any animal disease model. Together, the data demonstrate that FRNK negatively regulates myofibroblast differentiation in vitro and in vivo. These data further suggest that modulation FRNK expression may be a novel avenue for therapeutic intervention in tissue fibrosis.
Collapse
Affiliation(s)
- Qiang Ding
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, University of Alabama at Birmingham, Birmingham, Alabama 35294, USA.
| | | | | | | | | |
Collapse
|