1
|
Fatehi Hassanabad A, Zarzycki AN, Fedak PWM. Cellular and molecular mechanisms driving cardiac tissue fibrosis: On the precipice of personalized and precision medicine. Cardiovasc Pathol 2024; 71:107635. [PMID: 38508436 DOI: 10.1016/j.carpath.2024.107635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 03/13/2024] [Accepted: 03/15/2024] [Indexed: 03/22/2024] Open
Abstract
Cardiac fibrosis is a significant contributor to heart failure, a condition that continues to affect a growing number of patients worldwide. Various cardiovascular comorbidities can exacerbate cardiac fibrosis. While fibroblasts are believed to be the primary cell type underlying fibrosis, recent and emerging data suggest that other cell types can also potentiate or expedite fibrotic processes. Over the past few decades, clinicians have developed therapeutics that can blunt the development and progression of cardiac fibrosis. While these strategies have yielded positive results, overall clinical outcomes for patients suffering from heart failure continue to be dire. Herein, we overview the molecular and cellular mechanisms underlying cardiac tissue fibrosis. To do so, we establish the known mechanisms that drive fibrosis in the heart, outline the diagnostic tools available, and summarize the treatment options used in contemporary clinical practice. Finally, we underscore the critical role the immune microenvironment plays in the pathogenesis of cardiac fibrosis.
Collapse
Affiliation(s)
- Ali Fatehi Hassanabad
- Section of Cardiac Surgery, Department of Cardiac Science, Libin Cardiovascular Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Anna N Zarzycki
- Section of Cardiac Surgery, Department of Cardiac Science, Libin Cardiovascular Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Paul W M Fedak
- Section of Cardiac Surgery, Department of Cardiac Science, Libin Cardiovascular Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.
| |
Collapse
|
2
|
Guo Z, Liu FY, Yang D, Wang MY, Li CF, Tang N, Ma SQ, An P, Yang Z, Tang QZ. Salidroside ameliorates pathological cardiac hypertrophy via TLR4-TAK1-dependent signaling. Phytother Res 2023; 37:1839-1849. [PMID: 36512326 DOI: 10.1002/ptr.7701] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 11/09/2022] [Accepted: 11/19/2022] [Indexed: 12/14/2022]
Abstract
Salidroside, a prominent active ingredient in traditional Chinese medicines, is garnering increased attention because of its unique pharmacological effects against ischemic heart disease via MAPK signaling, which plays a critical role in regulating the evolution of ventricular hypertrophy. However, the function of Salidroside on myocardial hypertrophy has not yet been elucidated. C57BL/6 mice were subjected to transverse aortic constriction (TAC), and treated with Salidroside (100 mg kg-1 day-1 ) by oral gavage for 3 weeks starting 1 week after surgery. Four weeks after TAC surgery, the mice were subjected to echocardiography and then sacrificed to harvest the hearts for analysis. For in vitro study, neonatal rat cardiomyocytes were used to validate the protective effects of Salidroside in response to Angiotensin II (Ang II, 1 μM) stimulation. Here, we proved that Salidroside dramatically inhibited hypertrophic reactions generated by pressure overload and isoproterenol (ISO) injection. Salidroside prevented the activation of the TAK1-JNK/p38 axis. Salidroside pretreatment of TAK1-inhibited cardiomyocytes shows no additional attenuation of Ang II-induced cardiomyocytes hypertrophy and signaling pathway activation. The overexpression of constitutively active TAK1 removed the protective effects of Salidroside on myocardial hypertrophy. TAC-induced increase of TLR4 protein expression was reduced considerably in the Salidroside treated mice. Transient transfection of small interfering RNA targeting TLR4 (siTLR4) in cardiomyocytes did not further decrease the activation of the TAK1/JNK-p38 axis. In conclusion, Salidroside functioned as a TLR4 inhibitor and displayed anti-hypertrophic action via the TAK1/JNK-p38 pathway.
Collapse
Affiliation(s)
- Zhen Guo
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, People's Republic of China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, People's Republic of China
| | - Fang-Yuan Liu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, People's Republic of China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, People's Republic of China
| | - Dan Yang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, People's Republic of China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, People's Republic of China
| | - Ming-Yu Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, People's Republic of China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, People's Republic of China
| | - Chen-Fei Li
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, People's Republic of China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, People's Republic of China
| | - Nan Tang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, People's Republic of China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, People's Republic of China
| | - Shu-Qing Ma
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, People's Republic of China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, People's Republic of China
| | - Peng An
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, People's Republic of China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, People's Republic of China
| | - Zheng Yang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, People's Republic of China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, People's Republic of China
| | - Qi-Zhu Tang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, People's Republic of China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, People's Republic of China
| |
Collapse
|
3
|
Pronio A, Covotta F, Pallotta L, Palma R, Badiali D, Sacchi MC, Lamazza A, Severi C. Eosinophilic Esophagitis: Cytokines Expression and Fibrotic Markers in Comparison to Celiac Disease. Diagnostics (Basel) 2022; 12:diagnostics12092092. [PMID: 36140492 PMCID: PMC9497632 DOI: 10.3390/diagnostics12092092] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 08/24/2022] [Accepted: 08/24/2022] [Indexed: 11/16/2022] Open
Abstract
Introduction: Eosinophilic esophagitis (EoE) is now recognized as the main inflammatory condition that leads to fibrosis, unlike other chronic inflammatory gastrointestinal diseases, such as celiac disease. The aim of our study is to characterize the collagen deposition and cytokine expression involved in the fibrogenic response in patients affected by EoE in comparison to celiac disease. Materials and Methods: Consecutive patients with a clinical suspicion of untreated EoE or active celiac disease were enrolled. In the control group, patients with negative upper endoscopy were included. Total RNA was isolated from biopsy specimens using a commercial kit (SV Total RNA Isolation System, Promega Italia Srl). Quantitative real-time PCR (qRT-PCR) was performed in triplicate using a StepOne™ Real-Time PCR instrument (Thermo Fisher Scientific, Monza, Italy). mRNA encoding for inflammatory molecules: interleukin 4 (IL-4), interleukin 5 (IL-5), interleukin 13 (IL-13), and fibrotic markers: transforming growth factor beta 1 (TGF-β), mitogen-activated protein kinase kinase kinase 7 (MAP3K7), serpin family E member 1 (SERPINE1), were quantified using TaqMan Gene Expression Assays (Applied Biosystems). RESULTS. In EoE, the qPCR analysis showed an increase in all the inflammatory cytokines. Both IL-5 and Il-3 mRNA expression resulted in a statistically significant increase in oesophageal mucosa with respect to the celiac duodenum, while no differences were present in IL-4 expression. TGF-β expression was similar to the controls in the mid esophagus but reduced in the distal EoE esophagus (RQ: 0.46 ± 0.1). MAP3K7 expression was reduced in the mid esophagus (RQ: 0.59 ± 0.3) and increased in the distal esophagus (RQ: 1.75 ± 0.6). In turn, the expression of SERPINE1 was increased in both segments and was higher in the mid than in the distal esophagus (RQ: 5.25 ± 3.9, 1.92 ± 0.9, respectively). Collagen deposition was greater in the distal esophagus compared to the mid esophagus [18.1% ± 8 vs. 1.3% ± 1; p = 0.008]. Conclusions: The present study confirms the esophageal fibrotic involution involving the distal esophagus and shows that the inflammatory pathway in EoE is peculiar to this disease and different from other chronic inflammatory gastrointestinal disorders such as celiac disease.
Collapse
Affiliation(s)
- Annamaria Pronio
- Department of General Surgery and Surgical Specialties ‘Paride Stefanini’, Sapienza University of Rome, 00185 Rome, Italy
| | - Francesco Covotta
- Department of General Surgery and Surgical Specialties ‘Paride Stefanini’, Sapienza University of Rome, 00185 Rome, Italy
| | - Lucia Pallotta
- Department of Translational and Precision Medicine, Sapienza University of Rome, 00185 Rome, Italy
| | - Rossella Palma
- Department of General Surgery and Surgical Specialties ‘Paride Stefanini’, Sapienza University of Rome, 00185 Rome, Italy
- Correspondence: ; Tel.: +39-33-4309-1174
| | - Danilo Badiali
- Department of Translational and Precision Medicine, Sapienza University of Rome, 00185 Rome, Italy
| | - Maria Carlotta Sacchi
- Department of Translational and Precision Medicine, Sapienza University of Rome, 00185 Rome, Italy
| | - Antonietta Lamazza
- Department of Surgery Pietro Valdoni, Sapienza University of Rome, 00185 Rome, Italy
| | - Carola Severi
- Department of Translational and Precision Medicine, Sapienza University of Rome, 00185 Rome, Italy
| |
Collapse
|
4
|
Targeting Myocardial Fibrosis—A Magic Pill in Cardiovascular Medicine? Pharmaceutics 2022; 14:pharmaceutics14081599. [PMID: 36015225 PMCID: PMC9414721 DOI: 10.3390/pharmaceutics14081599] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 07/27/2022] [Accepted: 07/28/2022] [Indexed: 11/16/2022] Open
Abstract
Fibrosis, characterized by an excessive accumulation of extracellular matrix, has long been seen as an adaptive process that contributes to tissue healing and regeneration. More recently, however, cardiac fibrosis has been shown to be a central element in many cardiovascular diseases (CVDs), contributing to the alteration of cardiac electrical and mechanical functions in a wide range of clinical settings. This paper aims to provide a comprehensive review of cardiac fibrosis, with a focus on the main pathophysiological pathways involved in its onset and progression, its role in various cardiovascular conditions, and on the potential of currently available and emerging therapeutic strategies to counteract the development and/or progression of fibrosis in CVDs. We also emphasize a number of questions that remain to be answered, and we identify hotspots for future research.
Collapse
|
5
|
Katwa LC, Mendoza C, Clements M. CVD and COVID-19: Emerging Roles of Cardiac Fibroblasts and Myofibroblasts. Cells 2022; 11:cells11081316. [PMID: 35455995 PMCID: PMC9031661 DOI: 10.3390/cells11081316] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 04/07/2022] [Accepted: 04/12/2022] [Indexed: 02/07/2023] Open
Abstract
Cardiovascular disease (CVD) is the leading cause of death worldwide. Current data suggest that patients with cardiovascular diseases experience more serious complications with coronavirus disease-19 (COVID-19) than those without CVD. In addition, severe COVID-19 appears to cause acute cardiac injury, as well as long-term adverse remodeling of heart tissue. Cardiac fibroblasts and myofibroblasts, being crucial in response to injury, may play a pivotal role in both contributing to and healing COVID-19-induced cardiac injury. The role of cardiac myofibroblasts in cardiac fibrosis has been well-established in the literature for decades. However, with the emergence of the novel coronavirus SARS-CoV-2, new cardiac complications are arising. Bursts of inflammatory cytokines and upregulation of TGF-β1 and angiotensin (AngII) are common in severe COVID-19 patients. Cytokines, TGF-β1, and Ang II can induce cardiac fibroblast differentiation, potentially leading to fibrosis. This review details the key information concerning the role of cardiac myofibroblasts in CVD and COVID-19 complications. Additionally, new factors including controlling ACE2 expression and microRNA regulation are explored as promising treatments for both COVID-19 and CVD. Further understanding of this topic may provide insight into the long-term cardiac manifestations of the COVID-19 pandemic and ways to mitigate its negative effects.
Collapse
|
6
|
Travers JG, Tharp CA, Rubino M, McKinsey TA. Therapeutic targets for cardiac fibrosis: from old school to next-gen. J Clin Invest 2022; 132:148554. [PMID: 35229727 PMCID: PMC8884906 DOI: 10.1172/jci148554] [Citation(s) in RCA: 63] [Impact Index Per Article: 31.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Cardiovascular diseases remain the leading cause of death worldwide, with pathological fibrotic remodeling mediated by activated cardiac myofibroblasts representing a unifying theme across etiologies. Despite the profound contributions of myocardial fibrosis to cardiac dysfunction and heart failure, there currently exist limited clinical interventions that effectively target the cardiac fibroblast and its role in fibrotic tissue deposition. Exploration of novel strategies designed to mitigate or reverse myofibroblast activation and cardiac fibrosis will likely yield powerful therapeutic approaches for the treatment of multiple diseases of the heart, including heart failure with preserved or reduced ejection fraction, acute coronary syndrome, and cardiovascular disease linked to type 2 diabetes. In this Review, we provide an overview of classical regulators of cardiac fibrosis and highlight emerging, next-generation epigenetic regulatory targets that have the potential to revolutionize treatment of the expanding cardiovascular disease patient population.
Collapse
|
7
|
Kang H, Seo E, Oh YS, Jun HS. TGF-β activates NLRP3 inflammasome by an autocrine production of TGF-β in LX-2 human hepatic stellate cells. Mol Cell Biochem 2022; 477:1329-1338. [PMID: 35138513 PMCID: PMC8989865 DOI: 10.1007/s11010-022-04369-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Accepted: 01/20/2022] [Indexed: 12/12/2022]
Abstract
Inflammation contributes to the pathogenesis of liver disease, and inflammasome activation has been identified as a major contributor to the amplification of liver inflammation. Transforming growth factor-beta (TGF-β) is a key regulator of liver physiology, contributing to all stages of liver disease. We investigated whether TGF-β is involved in inflammasome-mediated fibrosis in hepatic stellate cells. Treatment with TGF-β increased priming of NLRP3 inflammasome signaling by increasing NLRP3 levels and activating TAK1-NF-kB signaling. Moreover, TGF-β increased the expression of p-Smad2/3-NOX4 in LX-2 cells and consequently increased ROS content, which is a trigger for NLRP3 inflammasome activation. Elevated expression of NEK7 and active caspase-1 was also shown in TGF-β-induced LX-2 cells, and this level was reduced by (5Z)-oxozeaenol, a TAK inhibitor. Finally, TGF-β-treated cells significantly increased TGF-β secretion levels, and their production was inhibited by IL-1β receptor antagonist treatment. In conclusion, TGF-β may represent an endogenous danger signal to the active NLRP3 inflammasome, by which IL-1β mediates TGF-β expression in an autocrine manner. Therefore, targeting the NLRP3 inflammasome may be a promising approach for the development of therapies for TGF-β-induced liver fibrosis.
Collapse
Affiliation(s)
- Hwansu Kang
- College of Pharmacy and Gachon Institute of Pharmaceutical Science, Gachon University, Incheon, Korea
- Lee Gil Ya Cancer and Diabetes Institute, College of Pharmacy and Gachon Institute of Pharmaceutical Science, 155 Gaetbeol-ro, Yeonsu-ku, Incheon, 21999, Korea
| | - Eunhui Seo
- College of Pharmacy and Gachon Institute of Pharmaceutical Science, Gachon University, Incheon, Korea
- Lee Gil Ya Cancer and Diabetes Institute, College of Pharmacy and Gachon Institute of Pharmaceutical Science, 155 Gaetbeol-ro, Yeonsu-ku, Incheon, 21999, Korea
| | - Yoon Sin Oh
- Department of Food and Nutrition, Eulji University, 553 Sanseong-daero, Sujeong-gu, Seongnam, 13135, Korea.
| | - Hee-Sook Jun
- College of Pharmacy and Gachon Institute of Pharmaceutical Science, Gachon University, Incheon, Korea.
- Lee Gil Ya Cancer and Diabetes Institute, College of Pharmacy and Gachon Institute of Pharmaceutical Science, 155 Gaetbeol-ro, Yeonsu-ku, Incheon, 21999, Korea.
- Gachon Medical Research Institute, Gil Hospital, Incheon, Korea.
| |
Collapse
|
8
|
Micale L, Morlino S, Carbone A, Carissimo A, Nardella G, Fusco C, Palumbo O, Schirizzi A, Russo F, Mazzoccoli G, Breckpot J, De Luca C, Ferraris A, Giunta C, Grammatico P, Haanpää MK, Mancano G, Forzano G, Cacchiarelli D, Van Esch H, Callewaert B, Rohrbach M, Castori M. Loss-of-function variants in exon 4 of TAB2 cause a recognizable multisystem disorder with cardiovascular, facial, cutaneous, and musculoskeletal involvement. Genet Med 2021; 24:439-453. [PMID: 34906501 DOI: 10.1016/j.gim.2021.10.009] [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: 07/23/2021] [Revised: 09/02/2021] [Accepted: 10/15/2021] [Indexed: 11/19/2022] Open
Abstract
PURPOSE This study aimed to describe a multisystemic disorder featuring cardiovascular, facial, musculoskeletal, and cutaneous anomalies caused by heterozygous loss-of-function variants in TAB2. METHODS Affected individuals were analyzed by next-generation technologies and genomic array. The presumed loss-of-function effect of identified variants was assessed by luciferase assay in cells transiently expressing TAB2 deleterious alleles. In available patients' fibroblasts, variant pathogenicity was further explored by immunoblot and osteoblast differentiation assays. The transcriptomic profile of fibroblasts was investigated by RNA sequencing. RESULTS A total of 11 individuals from 8 families were heterozygotes for a novel TAB2 variant. In total, 7 variants were predicted to be null alleles and 1 was a missense change. An additional subject was heterozygous for a 52 kb microdeletion involving TAB2 exons 1 to 3. Luciferase assay indicated a decreased transcriptional activation mediated by NF-κB signaling for all point variants. Immunoblot analysis showed a reduction of TAK1 phosphorylation while osteoblast differentiation was impaired. Transcriptomic analysis identified deregulation of multiple pleiotropic pathways, such as TGFβ-, Ras-MAPK-, and Wnt-signaling networks. CONCLUSION Our data defined a novel disorder associated with loss-of-function or, more rarely, hypomorphic alleles in a restricted linker region of TAB2. The pleiotropic manifestations in this disorder partly recapitulate the 6q25.1 (TAB2) microdeletion syndrome and deserve the definition of cardio-facial-cutaneous-articular syndrome.
Collapse
Affiliation(s)
- Lucia Micale
- Division of Medical Genetics, Fondazione IRCCS-Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Silvia Morlino
- Division of Medical Genetics, Fondazione IRCCS-Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Annalucia Carbone
- Unit of Chronobiology, Division of Internal Medicine, Fondazione IRCCS-Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Annamaria Carissimo
- Institute for Applied Mathematics "Mauro Picone" National Research Council, Naples, Italy
| | - Grazia Nardella
- Division of Medical Genetics, Fondazione IRCCS-Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Carmela Fusco
- Division of Medical Genetics, Fondazione IRCCS-Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Orazio Palumbo
- Division of Medical Genetics, Fondazione IRCCS-Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Annalisa Schirizzi
- Division of Medical Genetics, Fondazione IRCCS-Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Federica Russo
- Division of Medical Genetics, Fondazione IRCCS-Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Gianluigi Mazzoccoli
- Unit of Chronobiology, Division of Internal Medicine, Fondazione IRCCS-Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Jeroen Breckpot
- Center for Human Genetics, University Hospital Leuven, Leuven, Belgium
| | - Chiara De Luca
- Center for Human Genetics, University Hospital Leuven, Leuven, Belgium
| | - Alessandro Ferraris
- Laboratory of Medical Genetics, Department of Molecular Medicine, Sapienza University, San Camillo-Forlanini Hospital, Rome, Italy
| | - Cecilia Giunta
- Division of Metabolism and Children's Research Center, University Children's Hospital Zurich and University of Zurich, Zurich, Switzerland
| | - Paola Grammatico
- Laboratory of Medical Genetics, Department of Molecular Medicine, Sapienza University, San Camillo-Forlanini Hospital, Rome, Italy
| | - Maria K Haanpää
- Department of Clinical Genetics and Genomics, Turku University Hospital and University of Turku, Turku, Finland
| | - Giorgia Mancano
- Medical Genetics Unit, Meyer Children's University Hospital, Florence, Italy
| | - Giulia Forzano
- Medical Genetics Unit, University of Florence, Florence, Italy
| | - Davide Cacchiarelli
- Telethon Institute of Genetics and Medicine (TIGEM), Naples, Italy; Department of Translational Medicine, University of Naples "Federico II", Naples, Italy
| | - Hilde Van Esch
- Center for Human Genetics, University Hospital Leuven, Leuven, Belgium
| | - Bert Callewaert
- Center for Medical Genetics and Department of Biomolecular Medicine, Ghent University Hospital, Ghent, Belgium
| | - Marianne Rohrbach
- Division of Metabolism and Children's Research Center, University Children's Hospital Zurich and University of Zurich, Zurich, Switzerland
| | - Marco Castori
- Division of Medical Genetics, Fondazione IRCCS-Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy.
| |
Collapse
|
9
|
Exogenous extracellular matrix proteins decrease cardiac fibroblast activation in stiffening microenvironment through CAPG. J Mol Cell Cardiol 2021; 159:105-119. [PMID: 34118218 PMCID: PMC10066715 DOI: 10.1016/j.yjmcc.2021.06.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 06/04/2021] [Accepted: 06/07/2021] [Indexed: 11/20/2022]
Abstract
Controlling fibrosis is an essential part of regenerating the post-ischemic heart. In the post-ischemic heart, fibroblasts differentiate to myofibroblasts that produce collagen-rich matrix to physically stabilize the infarct area. Infarct models in adult mice result in permanent scarring unlike newborn animals which fully regenerate. Decellularized extracellular matrix (dECM) hydrogels derived from early-aged hearts have been shown to be a transplantable therapy that preserves heart function and stimulates cardiomyocyte proliferation and vascularization. In this study, we investigate the anti-fibrotic effects of injectable dECM hydrogels in a cardiac explant model in the context of age-associated tissue compliance. Treatments with adult and fetal dECM hydrogels were tested for molecular effects on cardiac fibroblast activation and fibrosis. Altered sensitivity of fibroblasts to the mechanosignaling of the remodeling microenvironment was evaluated by manipulating the native extracellular matrix in explants and also with elastomeric substrates in the presence of dECM hydrogels. The injectable fetal dECM hydrogel treatment decreases fibroblast activation and contractility and lowers the stiffness-mediated increases in fibroblast activation observed in stiffened explants. The anti-fibrotic effect of dECM hydrogel is most observable at highest stiffness. Experiments with primary cells on elastomeric substrates with dECM treatment support this phenomenon. Transcriptome analysis indicated that dECM hydrogels affect cytoskeleton related signaling including Macrophage capping protein (CAPG) and Leupaxin (LPXN). CAPG was down-regulated by the fetal dECM hydrogel. LPXN expression was decreased by stiffening the explants; however, this effect was reversed by dECM hydrogel treatment. Pharmacological disruption of cytoskeleton polymerization lowered fibroblast activation and CAPG levels. Knocking down CAPG expression with siRNA inhibited fibroblast activation and collagen deposition. Collectively, fibroblast activation is dependent on cooperative action of extracellular molecular signals and mechanosignaling by cytoskeletal integrity.
Collapse
|
10
|
Li X, Li L, Lei W, Chua HZ, Li Z, Huang X, Wang Q, Li N, Zhang H. Traditional Chinese medicine as a therapeutic option for cardiac fibrosis: Pharmacology and mechanisms. Biomed Pharmacother 2021; 142:111979. [PMID: 34358754 DOI: 10.1016/j.biopha.2021.111979] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 07/05/2021] [Accepted: 07/26/2021] [Indexed: 02/06/2023] Open
Abstract
Cardiovascular diseases are one of the leading causes of death worldwide and cardiac fibrosis is a common pathological process for cardiac remodeling in cardiovascular diseases. Cardiac fibrosis not only accelerates the deterioration progress of diseases but also becomes a pivotal contributor for futile treatment in clinical cardiovascular trials. Although cardiac fibrosis is common and prevalent, effective medicines to provide sufficient clinical intervention for cardiac fibrosis are still unavailable. Traditional Chinese medicine (TCM) is the natural essence experienced boiling, fry, and other processing methods, including active ingredients, extracts, and herbal formulas, which have been applied to treat human diseases for a long history. Recently, research has increasingly focused on the great potential of TCM for the prevention and treatment of cardiac fibrosis. Here, we aim to clarify the identified pro-fibrotic mechanisms and intensively summarize the application of TCM in improving cardiac fibrosis by working on these mechanisms. Through comprehensively analyzing, TCM mainly regulates the following pathways during ameliorating cardiac fibrosis: attenuation of inflammation and oxidative stress, inhibition of cardiac fibroblasts activation, reduction of extracellular matrix accumulation, modulation of the renin-angiotensin-aldosterone system, modulation of autophagy, regulation of metabolic-dependent mechanisms, and targeting microRNAs. We also discussed the deficiencies and the development direction of anti-fibrotic therapies on cardiac fibrosis. The data reviewed here demonstrates that TCM shows a robust effect on alleviating cardiac fibrosis, which provides us a rich source of new drugs or drug candidates. Besides, we also hope this review may give some enlightenment for treating cardiac fibrosis in clinical practice.
Collapse
Affiliation(s)
- Xiao Li
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Innovation Team of Research on Compound Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| | - Lin Li
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Innovation Team of Research on Compound Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| | - Wei Lei
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Innovation Team of Research on Compound Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| | - Hui Zi Chua
- Evidence-Based Medicine Center, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| | - Zining Li
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Innovation Team of Research on Compound Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| | - Xianglong Huang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300381, China.
| | - Qilong Wang
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Innovation Team of Research on Compound Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| | - Nan Li
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Innovation Team of Research on Compound Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| | - Han Zhang
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Innovation Team of Research on Compound Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| |
Collapse
|
11
|
Aashaq S, Batool A, Mir SA, Beigh MA, Andrabi KI, Shah ZA. TGF-β signaling: A recap of SMAD-independent and SMAD-dependent pathways. J Cell Physiol 2021; 237:59-85. [PMID: 34286853 DOI: 10.1002/jcp.30529] [Citation(s) in RCA: 67] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Revised: 06/06/2021] [Accepted: 07/06/2021] [Indexed: 12/20/2022]
Abstract
Transforming growth factor-β (TGF-β) is a proinflammatory cytokine known to control a diverse array of pathological and physiological conditions during normal development and tumorigenesis. TGF-β-mediated physiological effects are heterogeneous and vary among different types of cells and environmental conditions. TGF-β serves as an antiproliferative agent and inhibits tumor development during primary stages of tumor progression; however, during the later stages, it encourages tumor development and mediates metastatic progression and chemoresistance. The fundamental elements of TGF-β signaling have been divulged more than a decade ago; however, the process by which the signals are relayed from cell surface to nucleus is very complex with additional layers added in tumor cell niches. Although the intricate understanding of TGF-β-mediated signaling pathways and their regulation are still evolving, we tried to make an attempt to summarize the TGF-β-mediated SMAD-dependent andSMAD-independent pathways. This manuscript emphasizes the functions of TGF-β as a metastatic promoter and tumor suppressor during the later and initial phases of tumor progression respectively.
Collapse
Affiliation(s)
- Sabreena Aashaq
- Department of Immunology and Molecular Medicine, Sher-i-Kashmir Institute of Medical Sciences, Soura, Srinagar, JK, India
| | - Asiya Batool
- Division of Cancer Pharmacology, Indian Institute of Integrative Medicine, Srinagar, JK, India
| | | | | | | | - Zaffar Amin Shah
- Department of Immunology and Molecular Medicine, Sher-i-Kashmir Institute of Medical Sciences, Soura, Srinagar, JK, India
| |
Collapse
|
12
|
Theret M, Low M, Rempel L, Li FF, Tung LW, Contreras O, Chang CK, Wu A, Soliman H, Rossi FMV. In vitro assessment of anti-fibrotic drug activity does not predict in vivo efficacy in murine models of Duchenne muscular dystrophy. Life Sci 2021; 279:119482. [PMID: 33891939 DOI: 10.1016/j.lfs.2021.119482] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 03/22/2021] [Accepted: 04/02/2021] [Indexed: 02/09/2023]
Abstract
AIM Fibrosis is the most common complication from chronic diseases, and yet no therapy capable of mitigating its effects is available. Our goal is to unveil specific signaling regulating the fibrogenic process and to identify potential small molecule candidates that block fibrogenic differentiation of fibro/adipogenic progenitors. METHOD We performed a large-scale drug screen using muscle-resident fibro/adipogenic progenitors from a mouse model expressing EGFP under the Collagen1a1 promotor. We first confirmed that the EGFP was expressed in response to TGFβ1 stimulation in vitro. Then we treated cells with TGFβ1 alone or with drugs from two libraries of known compounds. The drugs ability to block the fibrogenic differentiation was quantified by imaging and flow cytometry. From a two-rounds screening, positive hits were tested in vivo in the mice model for the Duchenne Muscular Dystrophy (mdx mice). The histopathology of the muscles was assessed with picrosirius red (fibrosis) and laminin staining (myofiber size). KEY FINDINGS From the in vitro drug screening, we identified 21 drugs and tested 3 in vivo on the mdx mice. None of the three drugs significantly improved muscle histopathology. SIGNIFICANCE The in vitro drug screen identified various efficient compounds, none of them strongly inhibited fibrosis in skeletal muscle of mdx mice. To explain these observations, we hypothesize that in Duchenne Muscular Dystrophy, in which fibrosis is a secondary event due to chronic degeneration and inflammation, the drugs tested could have adverse effect on regeneration or inflammation, balancing off any positive effects and leading to the absence of significant results.
Collapse
Affiliation(s)
- Marine Theret
- School of Biomedical Engineering and the Biomedical Research Centre, Department of Medical Genetics, 2222 Health Sciences Mall, Vancouver, BC V6T 1Z3, Canada.
| | - Marcela Low
- School of Biomedical Engineering and the Biomedical Research Centre, Department of Medical Genetics, 2222 Health Sciences Mall, Vancouver, BC V6T 1Z3, Canada
| | - Lucas Rempel
- School of Biomedical Engineering and the Biomedical Research Centre, Department of Medical Genetics, 2222 Health Sciences Mall, Vancouver, BC V6T 1Z3, Canada
| | - Fang Fang Li
- School of Biomedical Engineering and the Biomedical Research Centre, Department of Medical Genetics, 2222 Health Sciences Mall, Vancouver, BC V6T 1Z3, Canada
| | - Lin Wei Tung
- School of Biomedical Engineering and the Biomedical Research Centre, Department of Medical Genetics, 2222 Health Sciences Mall, Vancouver, BC V6T 1Z3, Canada
| | - Osvaldo Contreras
- Developmental and Stem Cell Biology Division, Victor Chang Cardiac Research Institute, Darlinghurst, NSW 2010, Australia; Departamento de Biología Celular y Molecular and Center for Aging and Regeneration (CARE-ChileUC), Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, 8331150 Santiago, Chile
| | - Chih-Kai Chang
- School of Biomedical Engineering and the Biomedical Research Centre, Department of Medical Genetics, 2222 Health Sciences Mall, Vancouver, BC V6T 1Z3, Canada
| | - Andrew Wu
- School of Biomedical Engineering and the Biomedical Research Centre, Department of Medical Genetics, 2222 Health Sciences Mall, Vancouver, BC V6T 1Z3, Canada
| | - Hesham Soliman
- School of Biomedical Engineering and the Biomedical Research Centre, Department of Medical Genetics, 2222 Health Sciences Mall, Vancouver, BC V6T 1Z3, Canada; Department of Pharmacology and Toxicology, Faculty of Pharmaceutical Sciences, Minia University, Minia, Egypt
| | - Fabio M V Rossi
- School of Biomedical Engineering and the Biomedical Research Centre, Department of Medical Genetics, 2222 Health Sciences Mall, Vancouver, BC V6T 1Z3, Canada
| |
Collapse
|
13
|
Sun X, Kang Y, Xue S, Zou J, Xu J, Tang D, Qin H. In vivo therapeutic success of MicroRNA-155 antagomir in a mouse model of pulmonary fibrosis induced by bleomycin. Korean J Intern Med 2021; 36:S160-S169. [PMID: 32506869 PMCID: PMC8009162 DOI: 10.3904/kjim.2019.098] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Accepted: 10/07/2019] [Indexed: 11/27/2022] Open
Abstract
BACKGROUND/AIMS MicroRNAs (miRNAs) play critical regulatory roles in the pathogenesis of pulmonary fibrosis. The aim of this study was to explore whether miRNA antagomirs could serve as potential therapeutic agents in interstitial lung diseases. METHODS A mouse model of pulmonary fibrosis was established by intratracheal injection of bleomycin (BLM). Using microarray analysis, up-regulated miRNAs were identified during the development of pulmonary fibrosis. miR-155 was chosen as the candidate miRNA. Fifteen mice were then randomized into the following three groups: BLM + antagomiR-155 group, treated with BLM plus intravenously injected with antagomiR-155; BLM group, treated with intratracheal BLM plus phosphate-buffered saline (PBS); and a control group, treated with PBS only. Lung tissues were collected for histopathological analysis, hydroxyproline measurement, and Western blotting. Enzyme-linked immunosorbent assays were used for the measurement of cytokines associated with pulmonary fibrosis. RESULTS Histological changes and hydroxyproline levels induced by BLM were significantly inhibited by antagomiR-155. The levels of interleukin 4 (IL-4) and transforming growth factor-β (TGF-β) expression were increased after BLM treatment. However, miR-155 silencing decreased the expression of IL-4, TGF-β, and interferon-γ. TGF-β-activated kinase 1/mitogen-activated protein kinase kinase kinase 7 (MAP3K7)-binding protein 2 (TAB2) of the mitogen-activated protein kinase (MAPK) signaling pathway, was activated by BLM and inhibited by in vivo silencing of miR-155 via antagomiR-155. CONCLUSION In vivo treatment with antagomiR-155 alleviated the pathological changes induced by BLM and may be a promising therapeutic strategy for pulmonary fibrosis.
Collapse
Affiliation(s)
- Xiaoyuan Sun
- Department of Respiratory, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Yu Kang
- Department of Cardiology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Shan Xue
- Department of Respiratory, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Jing Zou
- Department of Respiratory, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Jiabo Xu
- Department of Respiratory, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Daoqiang Tang
- Department of Pathology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Hui Qin
- Department of Respiratory, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
- Correspondence to Hui Qin, M.D. Department of Respiratory, Renji Hospital, School of Medicine, Shanghai Jiaotong University, No 160, Pujian Road, Shanghai 200127, China Tel: +86-68383101 Fax: +86-2168383101 E-mail:
| |
Collapse
|
14
|
Di Gregorio J, Robuffo I, Spalletta S, Giambuzzi G, De Iuliis V, Toniato E, Martinotti S, Conti P, Flati V. The Epithelial-to-Mesenchymal Transition as a Possible Therapeutic Target in Fibrotic Disorders. Front Cell Dev Biol 2020; 8:607483. [PMID: 33409282 PMCID: PMC7779530 DOI: 10.3389/fcell.2020.607483] [Citation(s) in RCA: 81] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Accepted: 11/25/2020] [Indexed: 12/11/2022] Open
Abstract
Fibrosis is a chronic and progressive disorder characterized by excessive deposition of extracellular matrix, which leads to scarring and loss of function of the affected organ or tissue. Indeed, the fibrotic process affects a variety of organs and tissues, with specific molecular background. However, two common hallmarks are shared: the crucial role of the transforming growth factor-beta (TGF-β) and the involvement of the inflammation process, that is essential for initiating the fibrotic degeneration. TGF-β in particular but also other cytokines regulate the most common molecular mechanism at the basis of fibrosis, the Epithelial-to-Mesenchymal Transition (EMT). EMT has been extensively studied, but not yet fully explored as a possible therapeutic target for fibrosis. A deeper understanding of the crosstalk between fibrosis and EMT may represent an opportunity for the development of a broadly effective anti-fibrotic therapy. Here we report the evidences of the relationship between EMT and multi-organ fibrosis, and the possible therapeutic approaches that may be developed by exploiting this relationship.
Collapse
Affiliation(s)
- Jacopo Di Gregorio
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL, United States
| | - Iole Robuffo
- Institute of Molecular Genetics, National Research Council, Section of Chieti, Chieti, Italy
| | - Sonia Spalletta
- Department of Clinical Pathology, E. Profili Hospital, Fabriano, Ancona, Italy
| | - Giulia Giambuzzi
- Department of Medical and Oral Sciences and Biotechnologies, University “G. d’Annunzio”, Chieti, Italy
| | - Vincenzo De Iuliis
- Department of Medical and Oral Sciences and Biotechnologies, University “G. d’Annunzio”, Chieti, Italy
| | - Elena Toniato
- Department of Medical and Oral Sciences and Biotechnologies, University “G. d’Annunzio”, Chieti, Italy
| | - Stefano Martinotti
- Department of Medical and Oral Sciences and Biotechnologies, University “G. d’Annunzio”, Chieti, Italy
| | - Pio Conti
- Postgraduate Medical School, University of Chieti-Pescara, Chieti, Italy
| | - Vincenzo Flati
- Department of Biotechnological and Applied Clinical Sciences, University of L’Aquila, L’Aquila, Italy
| |
Collapse
|
15
|
Xu D, Zhao L, Jiang J, Li S, Sun Z, Huang X, Li C, Wang T, Sun L, Li X, Jiang Z, Zhang L. A potential therapeutic effect of catalpol in Duchenne muscular dystrophy revealed by binding with TAK1. J Cachexia Sarcopenia Muscle 2020; 11:1306-1320. [PMID: 32869445 PMCID: PMC7567147 DOI: 10.1002/jcsm.12581] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Revised: 02/29/2020] [Accepted: 04/07/2020] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Duchenne muscular dystrophy (DMD) is a progressive muscle disease caused by the loss of dystrophin, which results in inflammation, fibrosis, and the inhibition of myoblast differentiation in skeletal muscle. Catalpol, an iridoid glycoside, improves skeletal muscle function by enhancing myogenesis; it has potential to treat DMD. We demonstrate the positive effects of catalpol in dystrophic skeletal muscle. METHODS mdx (loss of dystrophin) mice (n = 18 per group) were treated with catalpol (200 mg/kg) for six consecutive weeks. Serum analysis, skeletal muscle performance and histology, muscle contractile function, and gene and protein expression were performed. Molecular docking and ligand-target interactions, RNA interference, immunofluorescence, and plasmids transfection were utilized to explore the protective mechanism in DMD by which catalpol binding with transforming growth factor-β-activated kinase 1 (TAK1) in skeletal muscle. RESULTS Six weeks of catalpol treatment improved whole-body muscle health in mdx mice, which was characterized by reduced plasma creatine kinase (n = 18, -35.1%, P < 0.05) and lactic dehydrogenase (n = 18, -10.3%, P < 0.05) activity. These effects were accompanied by enhanced grip strength (n = 18, +25.4%, P < 0.05) and reduced fibrosis (n = 18, -29.0% for hydroxyproline content, P < 0.05). Moreover, catalpol treatment protected against muscle fatigue and promoted muscle recovery in the tibialis anterior (TA) and diaphragm (DIA) muscles (n = 6, +69.8%, P < 0.05 and + 74.8%, P < 0.001, respectively), which was accompanied by enhanced differentiation in primary myoblasts from DMD patients (n = 6, male, mean age: 4.7 ± 1.9 years) and mdx mice. In addition, catalpol eliminated p-TAK1 overexpression in mdx mice (n = 12, -21.3%, P < 0.05) and primary myoblasts. The catalpol-induced reduction in fibrosis and increased myoblast differentiation resulted from the inhibition of TAK1 phosphorylation, leading to reduced myoblast trans-differentiation into myofibroblasts. Catalpol inhibited the phosphorylation of TAK1 by binding to TAK1, possibly at Asp-206, Thr-208, Asn-211, Glu-297, Lys-294, and Tyr-293. CONCLUSIONS Our findings show that catalpol and TAK1 inhibitors substantially improve whole-body muscle health and the function of dystrophic skeletal muscles and may provide a novel therapy for DMD.
Collapse
Affiliation(s)
- Dengqiu Xu
- Jiangsu Key Laboratory of Drug ScreeningChina Pharmaceutical UniversityNanjingChina
| | - Lei Zhao
- Department of NeurologyChildren's Hospital of Fudan UniversityShanghaiChina
| | - Jingwei Jiang
- Jiangsu Key Laboratory of Drug ScreeningChina Pharmaceutical UniversityNanjingChina
| | - Sijia Li
- Jiangsu Key Laboratory of Drug ScreeningChina Pharmaceutical UniversityNanjingChina
| | - Zeren Sun
- Jiangsu Key Laboratory of Drug ScreeningChina Pharmaceutical UniversityNanjingChina
| | - Xiaofei Huang
- Jiangsu Key Laboratory of Drug ScreeningChina Pharmaceutical UniversityNanjingChina
| | - Chunjie Li
- Jiangsu Key Laboratory of Drug ScreeningChina Pharmaceutical UniversityNanjingChina
| | - Tao Wang
- Jiangsu Key Laboratory of Drug ScreeningChina Pharmaceutical UniversityNanjingChina
- Jiangsu Center for Pharmacodynamics Research and EvaluationChina Pharmaceutical UniversityNanjingChina
| | - Lixin Sun
- Jiangsu Key Laboratory of Drug ScreeningChina Pharmaceutical UniversityNanjingChina
| | - Xihua Li
- Department of NeurologyChildren's Hospital of Fudan UniversityShanghaiChina
| | - Zhenzhou Jiang
- Jiangsu Key Laboratory of Drug ScreeningChina Pharmaceutical UniversityNanjingChina
- Key Laboratory of Drug Quality Control and PharmacovigilanceChina Pharmaceutical UniversityNanjingChina
| | - Luyong Zhang
- Jiangsu Key Laboratory of Drug ScreeningChina Pharmaceutical UniversityNanjingChina
- Center for Drug Research and DevelopmentGuangdong Pharmaceutical UniversityGuangzhouChina
- Key Laboratory of Drug Quality Control and PharmacovigilanceChina Pharmaceutical UniversityNanjingChina
| |
Collapse
|
16
|
Ran D, Ma Y, Liu W, Luo T, Zheng J, Wang D, Song R, Zhao H, Zou H, Gu J, Yuan Y, Bian J, Liu Z. TGF-β-activated kinase 1 (TAK1) mediates cadmium-induced autophagy in osteoblasts via the AMPK / mTORC1 / ULK1 pathway. Toxicology 2020; 442:152538. [PMID: 32693121 DOI: 10.1016/j.tox.2020.152538] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 06/23/2020] [Accepted: 07/13/2020] [Indexed: 01/28/2023]
Abstract
Cadmium (Cd) is one of worldwide environmental pollutants that causes bone homeostasis, which depends on the resorption of bones by osteoclasts and formation of bones by the osteoblasts (OB). However, the Cd toxicity on OB and its mechanism are unclear. Autophagy is an evolutionarily conserved degradation process in which domestic intracellular components are selectively digested for the recycling of nutrients and energy. This process is indispensable for cell homeostasis maintenance and stress responses. Dysregulation at the level of autophagic activity consequently disturbs the balance between bone formation and bone resorption and mediates the onset and progression of multiple bone diseases, including osteoporosis. TAK1 has been recently emerged as an activator of AMPK and hence an autophagy inducer. AMPK is a key molecule that induces autophagy and regulates cellular metabolism to maintain energy homeostasis. Conversely, autophagy is inhibited by mTORC1. In this study, we found that Cd treatment caused the formation of autophagosomes, LC3-II lipidation and p62 downregulation, and the increased autophagic flux, indicating that Cd treatment induced autophagy in OBs. Cd treatment induced TAK1 activation mediated AMPK phosphorylation, which promoted autophagy via phosphorylation of ULK1 at S317. Meanwhile, Cd treatment dramatically decreased mTORC1, S6K1, 4E-BP1, S6, ULK1S555 and ULK1S757 phosphorylation, suggesting that mTORC1 activity was inhibited and inactive mTORC1 prevents ULK1 activation by phosphorylating ULK1 at SerS555 and Ser757. Our data strongly suggest that TAK1 mediates AMPK activation, which activates ULK1 by phosphorylating ULK1S317 and suppressing mTORC1-mediated ULK1S555 and ULK1S757 phosphorylation. Our study has revealed a signaling mechanism for TAK1 in Cd-induced autophagy in OBs.
Collapse
Affiliation(s)
- Di Ran
- College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu, 225009, PR China; Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, Jiangsu, 225009, PR China; Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education of China, Yangzhou University, PR China
| | - Yonggang Ma
- College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu, 225009, PR China; Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, Jiangsu, 225009, PR China; Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education of China, Yangzhou University, PR China
| | - Wei Liu
- College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu, 225009, PR China; Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, Jiangsu, 225009, PR China; Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education of China, Yangzhou University, PR China
| | - Tongwang Luo
- College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu, 225009, PR China; Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, Jiangsu, 225009, PR China; Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education of China, Yangzhou University, PR China
| | - Jiaming Zheng
- College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu, 225009, PR China; Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, Jiangsu, 225009, PR China; Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education of China, Yangzhou University, PR China
| | - Dedong Wang
- College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu, 225009, PR China; Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, Jiangsu, 225009, PR China; Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education of China, Yangzhou University, PR China
| | - Ruilong Song
- College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu, 225009, PR China; Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, Jiangsu, 225009, PR China; Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education of China, Yangzhou University, PR China
| | - Hongyan Zhao
- College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu, 225009, PR China; Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, Jiangsu, 225009, PR China; Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education of China, Yangzhou University, PR China
| | - Hui Zou
- College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu, 225009, PR China; Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, Jiangsu, 225009, PR China; Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education of China, Yangzhou University, PR China
| | - Jianhong Gu
- College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu, 225009, PR China; Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, Jiangsu, 225009, PR China; Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education of China, Yangzhou University, PR China
| | - Yan Yuan
- College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu, 225009, PR China; Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, Jiangsu, 225009, PR China; Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education of China, Yangzhou University, PR China
| | - Jianchun Bian
- College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu, 225009, PR China; Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, Jiangsu, 225009, PR China; Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education of China, Yangzhou University, PR China
| | - Zongping Liu
- College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu, 225009, PR China; Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, Jiangsu, 225009, PR China; Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education of China, Yangzhou University, PR China.
| |
Collapse
|
17
|
Micale L, Morlino S, Biagini T, Carbone A, Fusco C, Ritelli M, Giambra V, Zoppi N, Nardella G, Notarangelo A, Schirizzi A, Mazzoccoli G, Grammatico P, Wade EM, Mazza T, Colombi M, Castori M. Insights into the molecular pathogenesis of cardiospondylocarpofacial syndrome: MAP3K7 c.737-7A > G variant alters the TGFβ-mediated α-SMA cytoskeleton assembly and autophagy. Biochim Biophys Acta Mol Basis Dis 2020; 1866:165742. [PMID: 32105826 DOI: 10.1016/j.bbadis.2020.165742] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Revised: 02/20/2020] [Accepted: 02/21/2020] [Indexed: 12/31/2022]
Abstract
Transforming growth factor beta-activated kinase 1 (TAK1) is a highly conserved kinase protein encoded by MAP3K7, and activated by multiple extracellular stimuli, growth factors and cytokines. Heterozygous variants in MAP3K7 cause the cardiospondylocarpofacial syndrome (CSCFS) which is characterized by short stature, dysmorphic facial features, cardiac septal defects with valve dysplasia, and skeletal anomalies. CSCFS has been described in seven patients to date and its molecular pathogenesis is only partially understood. Here, the functional effects of the MAP3K7 c.737-7A > G variant, previously identified in a girl with CSCFS and additional soft connective tissue features, were explored. This splice variant generates an in-frame insertion of 2 amino acid residues in the kinase domain of TAK1. Computational analysis revealed that this in-frame insertion alters protein dynamics in the kinase activation loop responsible for TAK1 autophosphorylation after binding with its interactor TAB1. Co-immunoprecipitation studies demonstrate that the ectopic expression of TAK1-mutated protein impairs its ability to physically bind TAB1. In patient's fibroblasts, MAP3K7 c.737-7A > G variant results in reduced TAK1 autophosphorylation and dysregulation of the downstream TAK1-dependent signaling pathway. TAK1 loss-of-function is associated with an impaired TGFβ-mediated α-SMA cytoskeleton assembly and cell migration, and defective autophagy process. These findings contribute to our understanding of the molecular pathogenesis of CSCFS and might offer the rationale for the design of novel therapeutic targets.
Collapse
Affiliation(s)
- Lucia Micale
- Division of Medical Genetics, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Foggia, Italy.
| | - Silvia Morlino
- Laboratory of Medical Genetics, Department of Molecular Medicine, Sapienza University, San Camillo-Forlanini Hospital, Rome, Italy
| | - Tommaso Biagini
- Unit of Bioinformatics, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Foggia, Italy
| | - Annalucia Carbone
- Division of Internal Medicine and Unit of Chronobiology, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Foggia, Italy
| | - Carmela Fusco
- Division of Medical Genetics, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Foggia, Italy
| | - Marco Ritelli
- Division of Biology and Genetics, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Vincenzo Giambra
- Institute for Stem Cell Biology, Regenerative Medicine and Innovative Therapies (ISBReMIT), Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Foggia, Italy
| | - Nicoletta Zoppi
- Division of Biology and Genetics, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Grazia Nardella
- Division of Medical Genetics, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Foggia, Italy; Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Angelantonio Notarangelo
- Division of Medical Genetics, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Foggia, Italy
| | - Annalisa Schirizzi
- Division of Medical Genetics, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Foggia, Italy; Dipartimento di Biologia, Università degli Studi di Bari, "Aldo Moro", Bari, Italy
| | - Gianluigi Mazzoccoli
- Division of Internal Medicine and Unit of Chronobiology, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Foggia, Italy
| | - Paola Grammatico
- Laboratory of Medical Genetics, Department of Molecular Medicine, Sapienza University, San Camillo-Forlanini Hospital, Rome, Italy
| | - Emma M Wade
- Department of Women's and Children's Health, Dunedin School of Medicine, University of Otago, Dunedin, New Zealand
| | - Tommaso Mazza
- Unit of Bioinformatics, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Foggia, Italy
| | - Marina Colombi
- Division of Biology and Genetics, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Marco Castori
- Division of Medical Genetics, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Foggia, Italy
| |
Collapse
|
18
|
Hu H, Lee SR, Bai H, Guo J, Hashimoto T, Isaji T, Guo X, Wang T, Wolf K, Liu S, Ono S, Yatsula B, Dardik A. TGFβ (Transforming Growth Factor-Beta)-Activated Kinase 1 Regulates Arteriovenous Fistula Maturation. Arterioscler Thromb Vasc Biol 2020; 40:e203-e213. [PMID: 32460580 DOI: 10.1161/atvbaha.119.313848] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
OBJECTIVE Arteriovenous fistulae (AVF) are the optimal conduit for hemodialysis access but have high rates of primary maturation failure. Successful AVF maturation requires wall thickening with deposition of ECM (extracellular matrix) including collagen and fibronectin, as well as lumen dilation. TAK1 (TGFβ [transforming growth factor-beta]-activated kinase 1) is a mediator of noncanonical TGFβ signaling and plays crucial roles in regulation of ECM production and deposition; therefore, we hypothesized that TAK1 regulates wall thickening and lumen dilation during AVF maturation. Approach and Results: In both human and mouse AVF, immunoreactivity of TAK1, JNK (c-Jun N-terminal kinase), p38, collagen 1, and fibronectin was significantly increased compared with control veins. Manipulation of TAK1 in vivo altered AVF wall thickening and luminal diameter; reduced TAK1 function was associated with reduced thickness and smaller diameter, whereas activation of TAK1 function was associated with increased thickness and larger diameter. Arterial magnitudes of laminar shear stress (20 dyne/cm2) activated noncanonical TGFβ signaling including TAK1 phosphorylation in mouse endothelial cells. CONCLUSIONS TAK1 is increased in AVF, and TAK1 manipulation in a mouse AVF model regulates AVF thickness and diameter. Targeting noncanonical TGFβ signaling such as TAK1 might be a novel therapeutic approach to improve AVF maturation.
Collapse
Affiliation(s)
- Haidi Hu
- From the Department of Vascular and Thyroid Surgery, The First Hospital of China Medical University, Shenyang (H.H.).,Department of Surgery (H.H., S.-R.L., H.B., J.G., T.H., T.I., X.G., T.W., K.W., S.L., S.O., B.Y., A.D.), Yale University School of Medicine, New Haven, CT.,Vascular Biology and Therapeutics Program (H.H., S.-R.L., H.B., J.G., T.H., T.I., X.G., T.W., K.W., S.L., S.O., B.Y., A.D.), Yale University School of Medicine, New Haven, CT
| | - Shin-Rong Lee
- Department of Surgery (H.H., S.-R.L., H.B., J.G., T.H., T.I., X.G., T.W., K.W., S.L., S.O., B.Y., A.D.), Yale University School of Medicine, New Haven, CT.,Vascular Biology and Therapeutics Program (H.H., S.-R.L., H.B., J.G., T.H., T.I., X.G., T.W., K.W., S.L., S.O., B.Y., A.D.), Yale University School of Medicine, New Haven, CT
| | - Hualong Bai
- Department of Surgery (H.H., S.-R.L., H.B., J.G., T.H., T.I., X.G., T.W., K.W., S.L., S.O., B.Y., A.D.), Yale University School of Medicine, New Haven, CT.,Vascular Biology and Therapeutics Program (H.H., S.-R.L., H.B., J.G., T.H., T.I., X.G., T.W., K.W., S.L., S.O., B.Y., A.D.), Yale University School of Medicine, New Haven, CT
| | - Jianming Guo
- Department of Surgery (H.H., S.-R.L., H.B., J.G., T.H., T.I., X.G., T.W., K.W., S.L., S.O., B.Y., A.D.), Yale University School of Medicine, New Haven, CT.,Vascular Biology and Therapeutics Program (H.H., S.-R.L., H.B., J.G., T.H., T.I., X.G., T.W., K.W., S.L., S.O., B.Y., A.D.), Yale University School of Medicine, New Haven, CT
| | - Takuya Hashimoto
- Department of Surgery (H.H., S.-R.L., H.B., J.G., T.H., T.I., X.G., T.W., K.W., S.L., S.O., B.Y., A.D.), Yale University School of Medicine, New Haven, CT.,Vascular Biology and Therapeutics Program (H.H., S.-R.L., H.B., J.G., T.H., T.I., X.G., T.W., K.W., S.L., S.O., B.Y., A.D.), Yale University School of Medicine, New Haven, CT
| | - Toshihiko Isaji
- Department of Surgery (H.H., S.-R.L., H.B., J.G., T.H., T.I., X.G., T.W., K.W., S.L., S.O., B.Y., A.D.), Yale University School of Medicine, New Haven, CT.,Vascular Biology and Therapeutics Program (H.H., S.-R.L., H.B., J.G., T.H., T.I., X.G., T.W., K.W., S.L., S.O., B.Y., A.D.), Yale University School of Medicine, New Haven, CT
| | - Xiangjiang Guo
- Department of Surgery (H.H., S.-R.L., H.B., J.G., T.H., T.I., X.G., T.W., K.W., S.L., S.O., B.Y., A.D.), Yale University School of Medicine, New Haven, CT.,Vascular Biology and Therapeutics Program (H.H., S.-R.L., H.B., J.G., T.H., T.I., X.G., T.W., K.W., S.L., S.O., B.Y., A.D.), Yale University School of Medicine, New Haven, CT
| | - Tun Wang
- Department of Surgery (H.H., S.-R.L., H.B., J.G., T.H., T.I., X.G., T.W., K.W., S.L., S.O., B.Y., A.D.), Yale University School of Medicine, New Haven, CT.,Vascular Biology and Therapeutics Program (H.H., S.-R.L., H.B., J.G., T.H., T.I., X.G., T.W., K.W., S.L., S.O., B.Y., A.D.), Yale University School of Medicine, New Haven, CT
| | - Katharine Wolf
- Department of Surgery (H.H., S.-R.L., H.B., J.G., T.H., T.I., X.G., T.W., K.W., S.L., S.O., B.Y., A.D.), Yale University School of Medicine, New Haven, CT.,Vascular Biology and Therapeutics Program (H.H., S.-R.L., H.B., J.G., T.H., T.I., X.G., T.W., K.W., S.L., S.O., B.Y., A.D.), Yale University School of Medicine, New Haven, CT
| | - Shirley Liu
- Department of Surgery (H.H., S.-R.L., H.B., J.G., T.H., T.I., X.G., T.W., K.W., S.L., S.O., B.Y., A.D.), Yale University School of Medicine, New Haven, CT.,Vascular Biology and Therapeutics Program (H.H., S.-R.L., H.B., J.G., T.H., T.I., X.G., T.W., K.W., S.L., S.O., B.Y., A.D.), Yale University School of Medicine, New Haven, CT
| | - Shun Ono
- Department of Surgery (H.H., S.-R.L., H.B., J.G., T.H., T.I., X.G., T.W., K.W., S.L., S.O., B.Y., A.D.), Yale University School of Medicine, New Haven, CT.,Vascular Biology and Therapeutics Program (H.H., S.-R.L., H.B., J.G., T.H., T.I., X.G., T.W., K.W., S.L., S.O., B.Y., A.D.), Yale University School of Medicine, New Haven, CT
| | - Bogdan Yatsula
- Department of Surgery (H.H., S.-R.L., H.B., J.G., T.H., T.I., X.G., T.W., K.W., S.L., S.O., B.Y., A.D.), Yale University School of Medicine, New Haven, CT.,Vascular Biology and Therapeutics Program (H.H., S.-R.L., H.B., J.G., T.H., T.I., X.G., T.W., K.W., S.L., S.O., B.Y., A.D.), Yale University School of Medicine, New Haven, CT
| | - Alan Dardik
- Department of Surgery (H.H., S.-R.L., H.B., J.G., T.H., T.I., X.G., T.W., K.W., S.L., S.O., B.Y., A.D.), Yale University School of Medicine, New Haven, CT.,Vascular Biology and Therapeutics Program (H.H., S.-R.L., H.B., J.G., T.H., T.I., X.G., T.W., K.W., S.L., S.O., B.Y., A.D.), Yale University School of Medicine, New Haven, CT.,Department of Surgery, VA Connecticut Healthcare Systems, West Haven, CT (A.D.)
| |
Collapse
|
19
|
Aashaq S, Batool A, Andrabi KI. TAK1 mediates convergence of cellular signals for death and survival. Apoptosis 2020; 24:3-20. [PMID: 30288639 DOI: 10.1007/s10495-018-1490-7] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
TGF-β activated kinase 1, a MAPK kinase kinase family serine threonine kinase has been implicated in regulating diverse range of cellular processes that include embryonic development, differentiation, autophagy, apoptosis and cell survival. TAK1 along with its binding partners TAB1, TAB2 and TAB3 displays a complex pattern of regulation that includes serious crosstalk with major signaling pathways including the C-Jun N-terminal kinase (JNK), p38 MAPK, and I-kappa B kinase complex (IKK) involved in establishing cellular commitments for death and survival. This review also highlights how TAK1 orchestrates regulation of energy homeostasis via AMPK and its emerging role in influencing mTORC1 pathway to regulate death or survival in tandem.
Collapse
Affiliation(s)
- Sabreena Aashaq
- Department of Biotechnology, University of Kashmir, Hazratbal, Srinagar, 190006, India.
| | - Asiya Batool
- Department of Biotechnology, University of Kashmir, Hazratbal, Srinagar, 190006, India
| | - Khurshid I Andrabi
- Department of Biotechnology, University of Kashmir, Hazratbal, Srinagar, 190006, India
| |
Collapse
|
20
|
Parichatikanond W, Luangmonkong T, Mangmool S, Kurose H. Therapeutic Targets for the Treatment of Cardiac Fibrosis and Cancer: Focusing on TGF-β Signaling. Front Cardiovasc Med 2020; 7:34. [PMID: 32211422 PMCID: PMC7075814 DOI: 10.3389/fcvm.2020.00034] [Citation(s) in RCA: 85] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2019] [Accepted: 02/24/2020] [Indexed: 12/22/2022] Open
Abstract
Transforming growth factor-β (TGF-β) is a common mediator of cancer progression and fibrosis. Fibrosis can be a significant pathology in multiple organs, including the heart. In this review, we explain how inhibitors of TGF-β signaling can work as antifibrotic therapy. After cardiac injury, profibrotic mediators such as TGF-β, angiotensin II, and endothelin-1 simultaneously activate cardiac fibroblasts, resulting in fibroblast proliferation and migration, deposition of extracellular matrix proteins, and myofibroblast differentiation, which ultimately lead to the development of cardiac fibrosis. The consequences of fibrosis include a wide range of cardiac disorders, including contractile dysfunction, distortion of the cardiac structure, cardiac remodeling, and heart failure. Among various molecular contributors, TGF-β and its signaling pathways which play a major role in carcinogenesis are considered master fibrotic mediators. In fact, recently the inhibition of TGF-β signaling pathways using small molecule inhibitors, antibodies, and gene deletion has shown that the progression of several cancer types was suppressed. Therefore, inhibitors of TGF-β signaling are promising targets for the treatment of tissue fibrosis and cancers. In this review, we discuss the molecular mechanisms of TGF-β in the pathogenesis of cardiac fibrosis and cancer. We will review recent in vitro and in vivo evidence regarding antifibrotic and anticancer actions of TGF-β inhibitors. In addition, we also present available clinical data on therapy based on inhibiting TGF-β signaling for the treatment of cancers and cardiac fibrosis.
Collapse
Affiliation(s)
| | - Theerut Luangmonkong
- Department of Pharmacology, Faculty of Pharmacy, Mahidol University, Bangkok, Thailand
| | - Supachoke Mangmool
- Department of Pharmacology, Faculty of Science, Mahidol University, Bangkok, Thailand
| | - Hitoshi Kurose
- Department of Pharmacology and Toxicology, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
| |
Collapse
|
21
|
Frangogiannis N. Transforming growth factor-β in tissue fibrosis. J Exp Med 2020; 217:e20190103. [PMID: 32997468 PMCID: PMC7062524 DOI: 10.1084/jem.20190103] [Citation(s) in RCA: 556] [Impact Index Per Article: 139.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Accepted: 12/24/2019] [Indexed: 12/21/2022] Open
Abstract
TGF-β is extensively implicated in the pathogenesis of fibrosis. In fibrotic lesions, spatially restricted generation of bioactive TGF-β from latent stores requires the cooperation of proteases, integrins, and specialized extracellular matrix molecules. Although fibroblasts are major targets of TGF-β, some fibrogenic actions may reflect activation of other cell types, including macrophages, epithelial cells, and vascular cells. TGF-β–driven fibrosis is mediated through Smad-dependent or non-Smad pathways and is modulated by coreceptors and by interacting networks. This review discusses the role of TGF-β in fibrosis, highlighting mechanisms of TGF-β activation and signaling, the cellular targets of TGF-β actions, and the challenges of therapeutic translation.
Collapse
Affiliation(s)
- Nikolaos Frangogiannis
- The Wilf Family Cardiovascular Research Institute, Department of Medicine (Cardiology), Albert Einstein College of Medicine, Bronx, NY
| |
Collapse
|
22
|
Abstract
Fibrosis is the abnormal deposition of extracellular matrix, which can lead to organ dysfunction, morbidity, and death. The disease burden caused by fibrosis is substantial, and there are currently no therapies that can prevent or reverse fibrosis. Metabolic alterations are increasingly recognized as an important pathogenic process that underlies fibrosis across many organ types. As a result, metabolically targeted therapies could become important strategies for fibrosis reduction. Indeed, some of the pathways targeted by antifibrotic drugs in development - such as the activation of transforming growth factor-β and the deposition of extracellular matrix - have metabolic implications. This Review summarizes the evidence to date and describes novel opportunities for the discovery and development of drugs for metabolic reprogramming, their associated challenges, and their utility in reducing fibrosis. Fibrotic therapies are potentially relevant to numerous common diseases such as cirrhosis, non-alcoholic steatohepatitis, chronic renal disease, heart failure, diabetes, idiopathic pulmonary fibrosis, and scleroderma.
Collapse
|
23
|
Bao C, Yang Z, Cai Q, Li Q, Li H, Shu B. Incremental load training improves renal fibrosis by regulating the TGF‑β1/TAK1/MKK3/p38MAPK signaling pathway and inducing the activation of autophagy in aged mice. Int J Mol Med 2019; 44:1677-1686. [PMID: 31545406 PMCID: PMC6777683 DOI: 10.3892/ijmm.2019.4344] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Accepted: 07/26/2019] [Indexed: 02/07/2023] Open
Abstract
Recent studies have confirmed that kidney tissue fibrosis is closely linked to the natural aging of organs. One of its major characteristics is the reduction of autophagic activity. However, to date, few studies have assessed whether incremental load training is able to improve the occurrence of renal fibrosis caused by natural aging and the underlying mechanisms. In the present study involving male C57/BL mice, an elderly exercise group (OY group) was subjected to progressive load‑increasing rotary‑bar training (5 days/week, lasting for 6 weeks), with an elderly control group (OC group) and a young control group (YC group) used as controls. Renal fibrosis and autophagy‑associated indicators were assessed by Masson's staining, reverse transcription‑quantitative PCR analysis, western blotting, immunofluorescence and transmission electron microscopy. The results suggested that collagen deposition in the basal part of the renal tubular epithelium and glomeruli in the OY group was significantly lower than that in the OC group. In the OC group, the protein expression levels of E‑cadherin, Beclin 1 and light chain 3 were significantly decreased, and increases in α‑smooth muscle actin‑positive signals were observed in the glomerular matrix and renal capsule wall. Furthermore, the expression of transforming growth factor (TGF)‑β1 and its downstream signaling molecules TGF‑β‑activated kinase 1 (TAK1), mitogen‑activated protein kinase (MAPK) kinase (MKK3) and p38MAPK were downregulated following training. The present study confirmed that incremental load training may improve renal fibrosis in aged mice by regulating the TGF‑β1/TAK1/MMK3/p38MAPK signaling pathway and inducing the activation of autophagy to reduce the synthesis of extracellular matrix and delay the epithelial‑mesenchymal transition. The present study provides a novel experimental basis for the intervention of incremental load training to prevent senile renal fibrosis.
Collapse
Affiliation(s)
- Chuncha Bao
- Department of Rehabilitation Medicine, University Town Hospital, Chongqing Medical University, Chongqing 401331, P.R. China
| | - Zhong Yang
- Department of Clinical Blood Teaching and Research, Army Medical University, Chongqing 400038, P.R. China
| | - Qiyan Cai
- Department of Histology and Embryology, Army Medical University, Chongqing 400038, P.R. China
| | - Qian Li
- Department of Rehabilitation Medicine, University Town Hospital, Chongqing Medical University, Chongqing 401331, P.R. China
| | - Hongli Li
- Department of Histology and Embryology, Army Medical University, Chongqing 400038, P.R. China
| | - Bin Shu
- Department of Rehabilitation Medicine, University Town Hospital, Chongqing Medical University, Chongqing 401331, P.R. China
| |
Collapse
|
24
|
Morlino S, Carbone A, Ritelli M, Fusco C, Giambra V, Nardella G, Notarangelo A, Panelli P, Mazzoccoli G, Zoppi N, Grammatico P, Wade EM, Colombi M, Castori M, Micale L. TAB2 c.1398dup variant leads to haploinsufficiency and impairs extracellular matrix homeostasis. Hum Mutat 2019; 40:1886-1898. [PMID: 31250519 DOI: 10.1002/humu.23834] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Revised: 05/28/2019] [Accepted: 05/31/2019] [Indexed: 12/16/2022]
Abstract
Transforming growth factor β-activated kinase 1 (TAK1) mediates multiple biological processes through the nuclear factor κ-light-chain-enhancer of activated B cells (NF-κB) and the mitogen-activated protein kinase (MAPK) signaling pathways. TAK1 activation is tightly regulated by its binding partners (TABs). In particular, binding with TAB2 is crucial for cardiovascular development and extracellular matrix (ECM) homeostasis. In our previous work, we reported a novel multisystem disorder associated with the heterozygous TAB2 c.1398dup variant. Here, we dissect the functional effects of this variant in order to understand its molecular pathogenesis. We demonstrate that TAB2 c.1398dup considerably undergoes to nonsense-mediated messenger RNA decay and encodes a truncated protein that loses its ability to bind TAK1. We also show an alteration of the TAK1 autophosphorylation status and of selected downstream signaling pathways in patients' fibroblasts. Immunofluorescence analyses and ECM-related polymerase chain reaction-array panels highlight that patient fibroblasts display ECM disorganization and altered expression of selected ECM components and collagen-related pathways. In conclusion, we deeply dissect the molecular pathogenesis of the TAB2 c.1398dup variant and show that the resulting phenotype is well explained by TAB2 loss-of-function. Our data also offer initial insights on the ECM homeostasis impairment as a molecular mechanism probably underlying a multisystem disorder linked to TAB2.
Collapse
Affiliation(s)
- Silvia Morlino
- Laboratory of Medical Genetics, Department of Molecular Medicine, San Camillo-Forlanini Hospital, Sapienza University, Rome, Italy
| | - Annalucia Carbone
- Division of Internal Medicine and Unit of Chronobiology, Fondazione IRCCS Casa Sollievo della Sofferenza, Foggia, Italy
| | - Marco Ritelli
- Division of Biology and Genetics, Department of Molecular and Translational Medicine, Universityinflammatory disorders and cancer of Brescia, Brescia, Italy
| | - Carmela Fusco
- Division of Medical Genetics, Fondazione IRCCS Casa Sollievo della Sofferenza, Foggia, Italy
| | - Vincenzo Giambra
- Institute for Stem Cell Biology, Regenerative Medicine and Innovative Therapies (ISBReMIT), Fondazione IRCCS Casa Sollievo della Sofferenza, Foggia, Italy
| | - Grazia Nardella
- Division of Medical Genetics, Fondazione IRCCS Casa Sollievo della Sofferenza, Foggia, Italy.,Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | | | - Patrizio Panelli
- Institute for Stem Cell Biology, Regenerative Medicine and Innovative Therapies (ISBReMIT), Fondazione IRCCS Casa Sollievo della Sofferenza, Foggia, Italy
| | - Gianluigi Mazzoccoli
- Division of Internal Medicine and Unit of Chronobiology, Fondazione IRCCS Casa Sollievo della Sofferenza, Foggia, Italy
| | - Nicoletta Zoppi
- Division of Biology and Genetics, Department of Molecular and Translational Medicine, Universityinflammatory disorders and cancer of Brescia, Brescia, Italy
| | - Paola Grammatico
- Laboratory of Medical Genetics, Department of Molecular Medicine, San Camillo-Forlanini Hospital, Sapienza University, Rome, Italy
| | - Emma M Wade
- Department of Women's and Children's Health, Dunedin School of Medicine, University of Otago, Dunedin, New Zealand
| | - Marina Colombi
- Division of Biology and Genetics, Department of Molecular and Translational Medicine, Universityinflammatory disorders and cancer of Brescia, Brescia, Italy
| | - Marco Castori
- Division of Medical Genetics, Fondazione IRCCS Casa Sollievo della Sofferenza, Foggia, Italy
| | - Lucia Micale
- Division of Medical Genetics, Fondazione IRCCS Casa Sollievo della Sofferenza, Foggia, Italy
| |
Collapse
|
25
|
Formica C, Happé H, Veraar KA, Vortkamp A, Scharpfenecker M, McNeill H, Peters DJ. Four-jointed knock-out delays renal failure in an ADPKD model with kidney injury. J Pathol 2019; 249:114-125. [PMID: 31038742 PMCID: PMC6772084 DOI: 10.1002/path.5286] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Revised: 04/19/2019] [Accepted: 04/26/2019] [Indexed: 12/28/2022]
Abstract
Autosomal Dominant Polycystic Kidney Disease is characterised by the development of fluid‐filled cysts in the kidneys which lead to end‐stage renal disease (ESRD). In the majority of cases, the disease is caused by a mutation in the Pkd1 gene. In a previous study, we demonstrated that renal injury can accelerate cyst formation in Pkd1 knock‐out (KO) mice. In that study, we found that after injury four‐jointed (Fjx1), an upstream regulator of planar cell polarity and the Hippo pathway, was aberrantly expressed in Pkd1 KO mice compared to WT. Therefore, we hypothesised a role for Fjx1 in injury/repair and cyst formation. We generated single and double deletion mice for Pkd1 and Fjx1, and we induced toxic renal injury using the nephrotoxic compound 1,2‐dichlorovinyl‐cysteine. We confirmed that nephrotoxic injury can accelerate cyst formation in Pkd1 mutant mice. This caused Pkd1 KO mice to reach ESRD significantly faster; unexpectedly, double KO mice survived significantly longer. Cyst formation was comparable in both models, but we found significantly less fibrosis and macrophage infiltration in double KO mice. Taken together, these data suggest that Fjx1 disruption protects the cystic kidneys against kidney failure by reducing inflammation and fibrosis. Moreover, we describe, for the first time, an interesting (yet unidentified) mechanism that partially discriminates cyst growth from fibrogenesis. © 2019 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Chiara Formica
- Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | - Hester Happé
- Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | - Kimberley Am Veraar
- Department of Pathology, Leiden University Medical Center, Leiden, The Netherlands
| | - Andrea Vortkamp
- Department of Developmental Biology, Centre of Medical Biotechnology, Faculty of Biology, University of Duisburg-Essen, Essen, Germany
| | | | - Helen McNeill
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO, USA.,Department of Molecular Genetics, University of Toronto, Toronto, Canada.,Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, Canada
| | - Dorien Jm Peters
- Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
26
|
Wang P, Lei S, Wang X, Xu W, Hu P, Chen F, Zhang X, Yin C, Xie W. MicroRNA-134 deactivates hepatic stellate cells by targeting TGF-β activated kinase 1-binding protein 1. Biochem Cell Biol 2019; 97:505-512. [PMID: 30645141 DOI: 10.1139/bcb-2018-0211] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Aberrant expression of microRNAs is associated with liver fibrogenesis. We previously found that microRNA-134 (miR-134) expression was reduced in fibrosis-based hepatocarcinogenesis induced by diethylinitrosamine. Herein we investigate the role and mechanisms of miR-134 in hepatic fibrosis. Our data show that miR-134 expression is reduced in rat hepatic fibrogenesis induced by carbontetrachloride, bile duct ligation, and dimethylnitrosamine, as well as in activated hepatic stellate cells (HSCs). Moreover, miR-134 inhibited HSC proliferation, and decreased the expression of smooth muscle actin and collagen I in HSCs, whereas the miR-134 inhibitor increased HSC activation. MiR-134 also negatively regulated transforming growth factor-β-activated kinase 1-binding protein 1 (TAB1) expression in both human and rat HSCs by directly binding to its 3' untranslated region. Importantly, TAB1 expression was significantly elevated during liver fibrogenesis and HSC activation. Knockdown of TAB1 inhibited the proliferation and fibrogenic behavior of HSCs, and significantly reduced the effect of the miR-134 inhibitor on HSC proliferation. Collectively, these data suggest that miR-134 inhibits the activation of HSCs via directly targeting TAB1, and the restoration of miR-134 or targeting TAB1 is of clinical significance in the treatment of liver fibrosis.
Collapse
Affiliation(s)
- Peiqin Wang
- Department of Gastroenterology, Shanghai Changzheng Hospital, Second Military Medical University, Shanghai 200003, China
| | - Shujuan Lei
- Department of Gastroenterology, Shanghai Changzheng Hospital, Second Military Medical University, Shanghai 200003, China
| | - Xiaohang Wang
- Department of Gastroenterology, Shanghai Changzheng Hospital, Second Military Medical University, Shanghai 200003, China.,Department of Gastroenterology, Shanghai Changzheng Hospital, Second Military Medical University, Shanghai 200003, China
| | - Wenping Xu
- Department of Gastroenterology, Shanghai Changzheng Hospital, Second Military Medical University, Shanghai 200003, China.,Department of Gastroenterology, Shanghai Changzheng Hospital, Second Military Medical University, Shanghai 200003, China
| | - Pingfang Hu
- Department of Gastroenterology, Shanghai Changzheng Hospital, Second Military Medical University, Shanghai 200003, China.,Department of Gastroenterology, Shanghai Changzheng Hospital, Second Military Medical University, Shanghai 200003, China
| | - Fei Chen
- Department of Gastroenterology, Shanghai Changzheng Hospital, Second Military Medical University, Shanghai 200003, China.,Department of Gastroenterology, Shanghai Changzheng Hospital, Second Military Medical University, Shanghai 200003, China
| | - Xin Zhang
- Department of Gastroenterology, Shanghai Changzheng Hospital, Second Military Medical University, Shanghai 200003, China.,Department of Gastroenterology, Shanghai Changzheng Hospital, Second Military Medical University, Shanghai 200003, China
| | - Chuan Yin
- Department of Gastroenterology, Shanghai Changzheng Hospital, Second Military Medical University, Shanghai 200003, China.,Department of Gastroenterology, Shanghai Changzheng Hospital, Second Military Medical University, Shanghai 200003, China
| | - Weifen Xie
- Department of Gastroenterology, Shanghai Changzheng Hospital, Second Military Medical University, Shanghai 200003, China.,Department of Gastroenterology, Shanghai Changzheng Hospital, Second Military Medical University, Shanghai 200003, China
| |
Collapse
|
27
|
Nelson JW, Ferdaus MZ, McCormick JA, Minnier J, Kaul S, Ellison DH, Barnes AP. Endothelial transcriptomics reveals activation of fibrosis-related pathways in hypertension. Physiol Genomics 2018; 50:104-116. [PMID: 29212850 PMCID: PMC5867617 DOI: 10.1152/physiolgenomics.00111.2017] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Revised: 11/20/2017] [Accepted: 12/06/2017] [Indexed: 12/30/2022] Open
Abstract
Hypertension poses a significant challenge to vasculature homeostasis and stands as the most common cardiovascular disease in the world. Its effects are especially profound on endothelial cells that form the inner lining of the vasculature and are directly exposed to the effects of excess pressure. Here, we characterize the in vivo transcriptomic response of cardiac endothelial cells to hypertension by rapidly isolating these cells from the spontaneous hypertension mouse model BPH/2J and its normotensive BPN/3J control strain and performing and RNA sequencing on both. Comparison of transcriptional differences between these groups reveals statistically significant changes in cellular pathways consistent with cardiac fibrosis found in hypertensive animals. Importantly, many of the fibrosis-linked genes identified also differ significantly between juvenile prehypertensive and adult hypertensive BPH/2J mice, suggesting that these transcriptional differences are hypertension related. We examined the dynamic nature of these transcriptional changes by testing whether blood pressure normalization using either a calcium channel blocker (amlodipine) or a angiotensin II receptor blocker (losartan) is able to reverse these expression patterns associated with hypertension. We find that blood pressure reduction is capable of reversing some gene-expression patterns, while other transcripts are recalcitrant to therapeutic intervention. This illuminates the possibility that unmanaged hypertension may irreversibly alter some endothelial transcriptional patterns despite later intervention. This study quantifies how endothelial cells are remodeled at the molecular level in cardiovascular pathology and advances our understanding of the transcriptional events associated with endothelial response to hypertensive challenge.
Collapse
Affiliation(s)
- Jonathan W Nelson
- The Knight Cardiovascular Institute, Oregon Health & Science University , Portland, Oregon
| | - Mohammed Z Ferdaus
- Division of Nephrology and Hypertension, Oregon Health & Science University, Portland, Oregon
| | - James A McCormick
- Division of Nephrology and Hypertension, Oregon Health & Science University, Portland, Oregon
| | - Jessica Minnier
- The Knight Cardiovascular Institute, Oregon Health & Science University , Portland, Oregon
| | - Sanjiv Kaul
- The Knight Cardiovascular Institute, Oregon Health & Science University , Portland, Oregon
| | - David H Ellison
- Division of Nephrology and Hypertension, Oregon Health & Science University, Portland, Oregon
- Department of Medicine, Oregon Clinical and Translational Research Institute, Oregon Health & Science University , Portland, Oregon
| | - Anthony P Barnes
- The Knight Cardiovascular Institute, Oregon Health & Science University , Portland, Oregon
| |
Collapse
|
28
|
Fan Z, Xu X, Qi X, Wu Y. Role of TGF-β activated kinase-1 inhibitor on the interaction between macrophages and mesangial cells on the condition of high glucose. Immunol Invest 2018; 47:303-314. [PMID: 29373048 DOI: 10.1080/08820139.2018.1428199] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
OBJECTIVE To investigate the effect of TGF-β activated kinase-1(TAK1) inhibitor 5Z-7-oxozeaenol on the interaction between macrophages and mesangial cells exposed to high glucose. METHODS The macrophages and mesangial cells were cultured separately or co-cultured and divided into seven groups: inhibitor control group, mannitol control group, normal control group, high glucose group and inhibitor groups. The expression of p-TAK1, TAK1 binding protein (TAB1), transcription factor NF - κ B (NF-κB p65) of macrophages were analyzed by Western blotting. The intracellular localization of NF-κB p65 was analyzed by immunofluorescence. The levels of inflammation cytokines and extracellular matrix were determined by enzyme-linked immune sorbent assay. Migration of macrophages was observed by microscope. RESULTS Compared with control group, the expression of p-TAK1, TAB1, NF-κB p65 were significantly higher in high glucose group (P < 0.05). Both in co-culture group and single culture group, the levels of inflammation cytokines and extracellular matrix (P < 0.05) in high glucose group were higher than that in control group. Exposed to high glucose, the levels of inflammation cytokines and extracellular matrix in co-cultured group were higher than that in single culture group (P < 0.05). 5Z-7-oxozeaenol can decrease those cytokines secretion, comparing with high glucose group (P < 0.05). The number of macrophages migration were decreased by 5Z-7-oxozeaenol (P < 0.05). CONCLUSION Exposed to high glucose, macrophages and mesangial cells can interact with each other to promote the secretion of inflammation cytokines and extracellular matrix. TAK1 inhibitor can reduce the secretion of inflammation cytokines and extracellular matrix components by intervening NF-κB p65 nuclear transfer and inhibiting macrophage migration.
Collapse
Affiliation(s)
- Zhe Fan
- a Department of Nephrology , The First Affiliated Hospital, Anhui Medical University , Hefei , Anhui , PR China
| | - Xingxin Xu
- a Department of Nephrology , The First Affiliated Hospital, Anhui Medical University , Hefei , Anhui , PR China
| | - Xiangming Qi
- a Department of Nephrology , The First Affiliated Hospital, Anhui Medical University , Hefei , Anhui , PR China
| | - Yonggui Wu
- a Department of Nephrology , The First Affiliated Hospital, Anhui Medical University , Hefei , Anhui , PR China
| |
Collapse
|
29
|
Bansal T, Chatterjee E, Singh J, Ray A, Kundu B, Thankamani V, Sengupta S, Sarkar S. Arjunolic acid, a peroxisome proliferator-activated receptor α agonist, regresses cardiac fibrosis by inhibiting non-canonical TGF-β signaling. J Biol Chem 2017; 292:16440-16462. [PMID: 28821620 DOI: 10.1074/jbc.m117.788299] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2017] [Revised: 08/11/2017] [Indexed: 12/16/2022] Open
Abstract
Cardiac hypertrophy and associated heart fibrosis remain a major cause of death worldwide. Phytochemicals have gained attention as alternative therapeutics for managing cardiovascular diseases. These include the extract from the plant Terminalia arjuna, which is a popular cardioprotectant and may prevent or slow progression of pathological hypertrophy to heart failure. Here, we investigated the mode of action of a principal bioactive T. arjuna compound, arjunolic acid (AA), in ameliorating hemodynamic load-induced cardiac fibrosis and identified its intracellular target. Our data revealed that AA significantly represses collagen expression and improves cardiac function during hypertrophy. We found that AA binds to and stabilizes the ligand-binding domain of peroxisome proliferator-activated receptor α (PPARα) and increases its expression during cardiac hypertrophy. PPARα knockdown during AA treatment in hypertrophy samples, including angiotensin II-treated adult cardiac fibroblasts and renal artery-ligated rat heart, suggests that AA-driven cardioprotection primarily arises from PPARα agonism. Moreover, AA-induced PPARα up-regulation leads to repression of TGF-β signaling, specifically by inhibiting TGF-β-activated kinase1 (TAK1) phosphorylation. We observed that PPARα directly interacts with TAK1, predominantly via PPARα N-terminal transactivation domain (AF-1) thereby masking the TAK1 kinase domain. The AA-induced PPARα-bound TAK1 level thereby shows inverse correlation with the phosphorylation level of TAK1 and subsequent reduction in p38 MAPK and NF-κBp65 activation, ultimately culminating in amelioration of excess collagen synthesis in cardiac hypertrophy. In conclusion, our findings unravel the mechanism of AA action in regressing hypertrophy-associated cardiac fibrosis by assigning a role of AA as a PPARα agonist that inactivates non-canonical TGF-β signaling.
Collapse
Affiliation(s)
- Trisha Bansal
- From the Genetics and Molecular Cardiology Laboratory, Department of Zoology, University of Calcutta, 35 Ballygunge Circular Road, Kolkata 700019, West Bengal
| | - Emeli Chatterjee
- From the Genetics and Molecular Cardiology Laboratory, Department of Zoology, University of Calcutta, 35 Ballygunge Circular Road, Kolkata 700019, West Bengal
| | - Jasdeep Singh
- the Kusuma School of Biological Sciences, Indian Institute of Technology Delhi, Hauz Khas, New Delhi-110016
| | - Arjun Ray
- the Genomics and Molecular Medicine Unit, Council of Scientific and Industrial Research (CSIR)-Institute of Genomics and Integrative Biology, Sukhdev Vihar, Mathura Road, New Delhi 110020, and
| | - Bishwajit Kundu
- the Kusuma School of Biological Sciences, Indian Institute of Technology Delhi, Hauz Khas, New Delhi-110016
| | - V Thankamani
- the Department of Biotechnology, University of Kerala, Thiruvananthapuram 695014, Kerala, India
| | - Shantanu Sengupta
- the Genomics and Molecular Medicine Unit, Council of Scientific and Industrial Research (CSIR)-Institute of Genomics and Integrative Biology, Sukhdev Vihar, Mathura Road, New Delhi 110020, and
| | - Sagartirtha Sarkar
- From the Genetics and Molecular Cardiology Laboratory, Department of Zoology, University of Calcutta, 35 Ballygunge Circular Road, Kolkata 700019, West Bengal,
| |
Collapse
|
30
|
Abstract
Cardiac fibrosis is a significant global health problem that is closely associated with multiple forms of cardiovascular disease, including myocardial infarction, dilated cardiomyopathy, and diabetes. Fibrosis increases myocardial wall stiffness due to excessive extracellular matrix deposition, causing impaired systolic and diastolic function, and facilitating arrhythmogenesis. As a result, patient morbidity and mortality are often dramatically elevated compared with those with cardiovascular disease but without overt fibrosis, demonstrating that fibrosis itself is both a pathologic response to existing disease and a significant risk factor for exacerbation of the underlying condition. The lack of any specific treatment for cardiac fibrosis in patients suffering from cardiovascular disease is a critical gap in our ability to care for these individuals. Here we provide an overview of the development of cardiac fibrosis, and discuss new research directions that have recently emerged and that may lead to the creation of novel treatments for patients with cardiovascular diseases. Such treatments would, ideally, complement existing therapy by specifically focusing on amelioration of fibrosis.
Collapse
Affiliation(s)
- Danah Al Hattab
- a Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, 351 Tache Avenue, Winnipeg, MB R2H 2A6, Canada.,b Department of Physiology and Pathophysiology, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R2H 2A6, Canada
| | - Michael P Czubryt
- a Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, 351 Tache Avenue, Winnipeg, MB R2H 2A6, Canada.,b Department of Physiology and Pathophysiology, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R2H 2A6, Canada
| |
Collapse
|
31
|
Hirata Y, Takahashi M, Morishita T, Noguchi T, Matsuzawa A. Post-Translational Modifications of the TAK1-TAB Complex. Int J Mol Sci 2017; 18:ijms18010205. [PMID: 28106845 PMCID: PMC5297835 DOI: 10.3390/ijms18010205] [Citation(s) in RCA: 102] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Revised: 01/12/2017] [Accepted: 01/13/2017] [Indexed: 12/17/2022] Open
Abstract
Transforming growth factor-β (TGF-β)-activated kinase 1 (TAK1) is a member of the mitogen-activated protein kinase kinase kinase (MAPKKK) family that is activated by growth factors and cytokines such as TGF-β, IL-1β, and TNF-α, and mediates a wide range of biological processes through activation of the nuclear factor-κB (NF-κB) and the mitogen-activated protein (MAP) kinase signaling pathways. It is well established that activation status of TAK1 is tightly regulated by forming a complex with its binding partners, TAK1-binding proteins (TAB1, TAB2, and TAB3). Interestingly, recent evidence indicates the importance of post-translational modifications (PTMs) of TAK1 and TABs in the regulation of TAK1 activation. To date, a number of PTMs of TAK1 and TABs have been revealed, and these PTMs appear to fine-tune and coordinate TAK1 activities depending on the cellular context. This review therefore focuses on recent advances in the understanding of the PTMs of the TAK1-TAB complex.
Collapse
Affiliation(s)
- Yusuke Hirata
- Laboratory of Health Chemistry, Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3, Aoba, Aramaki, Aoba-ku, Sendai 980-8578, Japan.
| | - Miki Takahashi
- Laboratory of Health Chemistry, Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3, Aoba, Aramaki, Aoba-ku, Sendai 980-8578, Japan.
| | - Tohru Morishita
- Laboratory of Health Chemistry, Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3, Aoba, Aramaki, Aoba-ku, Sendai 980-8578, Japan.
| | - Takuya Noguchi
- Laboratory of Health Chemistry, Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3, Aoba, Aramaki, Aoba-ku, Sendai 980-8578, Japan.
| | - Atsushi Matsuzawa
- Laboratory of Health Chemistry, Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3, Aoba, Aramaki, Aoba-ku, Sendai 980-8578, Japan.
| |
Collapse
|
32
|
Shimizu I, Minamino T. Physiological and pathological cardiac hypertrophy. J Mol Cell Cardiol 2016; 97:245-62. [PMID: 27262674 DOI: 10.1016/j.yjmcc.2016.06.001] [Citation(s) in RCA: 616] [Impact Index Per Article: 77.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2015] [Revised: 05/10/2016] [Accepted: 06/01/2016] [Indexed: 12/24/2022]
Abstract
The heart must continuously pump blood to supply the body with oxygen and nutrients. To maintain the high energy consumption required by this role, the heart is equipped with multiple complex biological systems that allow adaptation to changes of systemic demand. The processes of growth (hypertrophy), angiogenesis, and metabolic plasticity are critically involved in maintenance of cardiac homeostasis. Cardiac hypertrophy is classified as physiological when it is associated with normal cardiac function or as pathological when associated with cardiac dysfunction. Physiological hypertrophy of the heart occurs in response to normal growth of children or during pregnancy, as well as in athletes. In contrast, pathological hypertrophy is induced by factors such as prolonged and abnormal hemodynamic stress, due to hypertension, myocardial infarction etc. Pathological hypertrophy is associated with fibrosis, capillary rarefaction, increased production of pro-inflammatory cytokines, and cellular dysfunction (impairment of signaling, suppression of autophagy, and abnormal cardiomyocyte/non-cardiomyocyte interactions), as well as undesirable epigenetic changes, with these complex responses leading to maladaptive cardiac remodeling and heart failure. This review describes the key molecules and cellular responses involved in physiological/pathological cardiac hypertrophy.
Collapse
Affiliation(s)
- Ippei Shimizu
- Department of Cardiovascular Biology and Medicine, Niigata University Graduate School of Medical and Dental Sciences, Niigata 951-8510, Japan; Division of Molecular Aging and Cell Biology, Niigata University Graduate School of Medical and Dental Sciences, Niigata 951-8510, Japan.
| | - Tohru Minamino
- Department of Cardiovascular Biology and Medicine, Niigata University Graduate School of Medical and Dental Sciences, Niigata 951-8510, Japan.
| |
Collapse
|
33
|
Abstract
Myocardial fibrosis is a significant global health problem associated with nearly all forms of heart disease. Cardiac fibroblasts comprise an essential cell type in the heart that is responsible for the homeostasis of the extracellular matrix; however, upon injury, these cells transform to a myofibroblast phenotype and contribute to cardiac fibrosis. This remodeling involves pathological changes that include chamber dilation, cardiomyocyte hypertrophy and apoptosis, and ultimately leads to the progression to heart failure. Despite the critical importance of fibrosis in cardiovascular disease, our limited understanding of the cardiac fibroblast impedes the development of potential therapies that effectively target this cell type and its pathological contribution to disease progression. This review summarizes current knowledge regarding the origins and roles of fibroblasts, mediators and signaling pathways known to influence fibroblast function after myocardial injury, as well as novel therapeutic strategies under investigation to attenuate cardiac fibrosis.
Collapse
Affiliation(s)
- Joshua G Travers
- From the Heart Institute, Division of Molecular Cardiovascular Biology, Cincinnati Children's Hospital Medical Center, OH
| | - Fadia A Kamal
- From the Heart Institute, Division of Molecular Cardiovascular Biology, Cincinnati Children's Hospital Medical Center, OH
| | - Jeffrey Robbins
- From the Heart Institute, Division of Molecular Cardiovascular Biology, Cincinnati Children's Hospital Medical Center, OH
| | - Katherine E Yutzey
- From the Heart Institute, Division of Molecular Cardiovascular Biology, Cincinnati Children's Hospital Medical Center, OH
| | - Burns C Blaxall
- From the Heart Institute, Division of Molecular Cardiovascular Biology, Cincinnati Children's Hospital Medical Center, OH.
| |
Collapse
|
34
|
Dubash AD, Kam CY, Aguado BA, Patel DM, Delmar M, Shea LD, Green KJ. Plakophilin-2 loss promotes TGF-β1/p38 MAPK-dependent fibrotic gene expression in cardiomyocytes. J Cell Biol 2016; 212:425-38. [PMID: 26858265 PMCID: PMC4754716 DOI: 10.1083/jcb.201507018] [Citation(s) in RCA: 74] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2015] [Accepted: 01/08/2016] [Indexed: 01/07/2023] Open
Abstract
Members of the desmosome protein family are integral components of the cardiac area composita, a mixed junctional complex responsible for electromechanical coupling between cardiomyocytes. In this study, we provide evidence that loss of the desmosomal armadillo protein Plakophilin-2 (PKP2) in cardiomyocytes elevates transforming growth factor β1 (TGF-β1) and p38 mitogen-activated protein kinase (MAPK) signaling, which together coordinate a transcriptional program that results in increased expression of profibrotic genes. Importantly, we demonstrate that expression of Desmoplakin (DP) is lost upon PKP2 knockdown and that restoration of DP expression rescues the activation of this TGF-β1/p38 MAPK transcriptional cascade. Tissues from PKP2 heterozygous and DP conditional knockout mouse models also exhibit elevated TGF-β1/p38 MAPK signaling and induction of fibrotic gene expression in vivo. These data therefore identify PKP2 and DP as central players in coordination of desmosome-dependent TGF-β1/p38 MAPK signaling in cardiomyocytes, pathways known to play a role in different types of cardiac disease, such as arrhythmogenic or hypertrophic cardiomyopathy.
Collapse
Affiliation(s)
- Adi D Dubash
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611 Department of Biology, Furman University, Greenville SC 29613
| | - Chen Y Kam
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
| | - Brian A Aguado
- Department of Biomedical Engineering, Northwestern University, Evanston IL 60208 Simpson Querrey Institute for BioNanotechnology, Northwestern University, Chicago IL 60611
| | - Dipal M Patel
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
| | - Mario Delmar
- New York University School of Medicine, New York, NY 10016
| | - Lonnie D Shea
- Department of Chemical and Biological Engineering, Northwestern University, Evanston, IL 60208 Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48105
| | - Kathleen J Green
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611 Department of Dermatology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
| |
Collapse
|
35
|
Sureshbabu A, Muhsin SA, Choi ME. TGF-β signaling in the kidney: profibrotic and protective effects. Am J Physiol Renal Physiol 2016; 310:F596-F606. [PMID: 26739888 DOI: 10.1152/ajprenal.00365.2015] [Citation(s) in RCA: 186] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2015] [Accepted: 01/04/2016] [Indexed: 12/31/2022] Open
Abstract
Transforming growth factor-β (TGF-β) is generally considered as a central mediator of fibrotic diseases. Indeed, much focus has been placed on inhibiting TGF-β and its downstream targets as ideal therapeutic strategies. However, pharmacological blockade of TGF-β has not yet translated into successful therapy for humans, which may be due to pleiotropic effects of TGF-β signaling. Equally, TGF-β signaling as a protective response in kidney injury has been relatively underexplored. An emerging body of evidence from experimental kidney disease models indicates multifunctionality of TGF-β capable of inducing profibrotic and protective effects. This review discusses recent advances highlighting the diverse roles of TGF-β in promoting not only renal fibrosis but also protective responses of TGF-β signaling. We review, in particular, growing evidence that supports protective effects of TGF-β by mechanisms which include inhibiting inflammation and induction of autophagy. Additional detailed studies are required to fully understand the diverse mechanisms of TGF-β actions in renal fibrosis and inflammation that will likely direct toward effective antifibrotic therapies.
Collapse
Affiliation(s)
- Angara Sureshbabu
- Division of Nephrology and Hypertension, Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medical College, New York, New York; and
| | - Saif A Muhsin
- New York-Presbyterian Hospital-Weill Cornell Medical Center, New York, New York
| | - Mary E Choi
- Division of Nephrology and Hypertension, Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medical College, New York, New York; and .,New York-Presbyterian Hospital-Weill Cornell Medical Center, New York, New York
| |
Collapse
|
36
|
Identification and characterization of a TAB1 gene involved in innate immunity of amphioxus (Branchiostoma belcheri). Gene 2016; 575:294-302. [DOI: 10.1016/j.gene.2015.09.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Revised: 08/06/2015] [Accepted: 09/01/2015] [Indexed: 11/21/2022]
|
37
|
Grillo AR, Scarpa M, D'Incà R, Brun P, Scarpa M, Porzionato A, De Caro R, Martines D, Buda A, Angriman I, Palù G, Sturniolo GC, Castagliuolo I. TAK1 is a key modulator of the profibrogenic phenotype of human ileal myofibroblasts in Crohn's disease. Am J Physiol Gastrointest Liver Physiol 2015; 309:G443-54. [PMID: 26185333 DOI: 10.1152/ajpgi.00400.2014] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2014] [Accepted: 07/06/2015] [Indexed: 01/31/2023]
Abstract
Transforming growth factor (TGF)-β-activated kinase 1 (TAK1) signaling can mediate inflammatory responses as well as tissue remodeling. Intestinal mucosal myofibroblast (IMF) activation drives gut fibrosis in Crohn's disease (CD); however, the molecular pathways involved are largely unknown. Thus we investigated the yet-unknown expression and function of TAK1 in human CD-associated fibrosis. Ileal surgical specimens, ileal biopsies, and IMF isolated from controls and CD patients were analyzed for TAK1 and its active phosphorylated form (pTAK1) by Western blotting, immunohistochemistry, and real-time quantitative PCR. TAK1 pharmacological inhibition and silencing were used to assess its role in collagen and inflammatory cytokine synthesis in IMF. TAK1 and pTAK1 levels increased in ileum specimens from CD patients compared with controls and correlated to tissue fibrosis. Similarly, TAK1 mRNA in ileal biopsies of CD patients correlated with fibrogenic marker expression but not inflammatory cytokines. CD-derived IMF showed higher TAK1 and pTAK1 expression associated with increased collagen1(α)1 mRNA levels compared with control IMF. TGF-β1 promoted pTAK1 nuclear translocation and collagen synthesis. TAK1 inhibition or silencing significantly reduced TGF-β1-stimulated collagen production and normalized the profibrogenic phenotype of CD-derived IMF. Taken together, these data suggest that TAK1 activation and nuclear translocation induce and maintain a fibrogenic phenotype in the IMF. Thus the TAK1 signaling pathway may represent a suitable target to design new, antifibrotic therapies.
Collapse
Affiliation(s)
- Alessia Rosaria Grillo
- Department of Molecular Medicine, University of Padova, Padova, Italy; Department of Surgery Oncology and Gastroenterology, University of Padova, Padova, Italy; and
| | - Melania Scarpa
- Oncological Surgery Unit, Veneto Institute of Oncology IOV - IRCCS, Padova, Italy
| | - Renata D'Incà
- Department of Surgery Oncology and Gastroenterology, University of Padova, Padova, Italy; and
| | - Paola Brun
- Department of Molecular Medicine, University of Padova, Padova, Italy
| | - Marco Scarpa
- Oncological Surgery Unit, Veneto Institute of Oncology IOV - IRCCS, Padova, Italy
| | - Andrea Porzionato
- Department of Molecular Medicine, University of Padova, Padova, Italy
| | - Raffaele De Caro
- Department of Molecular Medicine, University of Padova, Padova, Italy
| | - Diego Martines
- Department of Surgery Oncology and Gastroenterology, University of Padova, Padova, Italy; and
| | - Andrea Buda
- Department of Surgery Oncology and Gastroenterology, University of Padova, Padova, Italy; and
| | - Imerio Angriman
- Department of Surgery Oncology and Gastroenterology, University of Padova, Padova, Italy; and
| | - Giorgio Palù
- Department of Molecular Medicine, University of Padova, Padova, Italy
| | - Giacomo Carlo Sturniolo
- Department of Surgery Oncology and Gastroenterology, University of Padova, Padova, Italy; and
| | | |
Collapse
|
38
|
Bao D, Lu D, Liu N, Dong W, Lu YD, Qin C, Zhang LF. Tomoregulin-1 prevents cardiac hypertrophy after pressure overload in mice by inhibiting TAK1-JNK pathways. Dis Model Mech 2015; 8:795-804. [PMID: 26092120 PMCID: PMC4527297 DOI: 10.1242/dmm.021303] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Accepted: 06/09/2015] [Indexed: 11/20/2022] Open
Abstract
Cardiac hypertrophy is associated with many forms of heart disease, and identifying important modifier genes involved in the pathogenesis of cardiac hypertrophy could lead to the development of new therapeutic strategies. Tomoregulin-1 is a growth factor that is primarily involved in embryonic development and adult central nervous system (CNS) function, and it is expressed abnormally in a variety of CNS pathologies. Tomoregulin-1 is also expressed in the myocardium. However, the effects of tomoregulin-1 on the heart, particularly on cardiac hypertrophy, remains unknown. The aim of the study is to examine whether and by what mechanism tomoregulin-1 regulates the development of cardiac hypertrophy induced by pressure overload. In this study, we found that tomoregulin-1 was significantly upregulated in two cardiac hypertrophy models: cTnT(R92Q) transgenic mice and thoracic aorta constriction (TAC)-induced cardiac hypertrophy mice. The transgenic overexpression of tomoregulin-1 increased the survival rate, improved the cardiac geometry and functional parameters of echocardiography, and decreased the degree of cardiac hypertrophy of the TAC mice, whereas knockdown of tomoregulin-1 expression resulted in an opposite phenotype and exacerbated phenotypes of cardiac hypertrophy induced by TAC. A possible mechanism by which tomoregulin-1 regulates the development of cardiac hypertrophy in TAC-induced cardiac hypertrophy is through inhibiting TGFβ non-canonical (TAK1-JNK) pathways in the myocardium. Tomoregulin-1 plays a protective role in the modulation of adverse cardiac remodeling from pressure overload in mice. Tomoregulin-1 could be a therapeutic target to control the development of cardiac hypertrophy.
Collapse
Affiliation(s)
- Dan Bao
- Key Laboratory of Human Disease Comparative Medicine, Ministry of Health, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences & Comparative Medical Center, Peking Union Medical College, Beijing 100021, China
| | - Dan Lu
- Key Laboratory of Human Disease Comparative Medicine, Ministry of Health, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences & Comparative Medical Center, Peking Union Medical College, Beijing 100021, China
| | - Ning Liu
- Key Laboratory of Human Disease Comparative Medicine, Ministry of Health, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences & Comparative Medical Center, Peking Union Medical College, Beijing 100021, China
| | - Wei Dong
- Key Laboratory of Human Disease Comparative Medicine, Ministry of Health, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences & Comparative Medical Center, Peking Union Medical College, Beijing 100021, China
| | - Ying-Dong Lu
- Key Laboratory of Human Disease Comparative Medicine, Ministry of Health, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences & Comparative Medical Center, Peking Union Medical College, Beijing 100021, China
| | - Chuan Qin
- Key Laboratory of Human Disease Animal Model, State Administration of Traditional Chinese Medicine, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences & Comparative Medical Center, Peking Union Medical College, Beijing 100021, China
| | - Lian-Feng Zhang
- Key Laboratory of Human Disease Comparative Medicine, Ministry of Health, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences & Comparative Medical Center, Peking Union Medical College, Beijing 100021, China
| |
Collapse
|
39
|
Abstract
Fibrotic diseases are a significant global burden for which there are limited treatment options. The effector cells of fibrosis are activated fibroblasts called myofibroblasts, a highly contractile cell type characterized by the appearance of α-smooth muscle actin stress fibers. The underlying mechanism behind myofibroblast differentiation and persistence has been under much investigation and is known to involve a complex signaling network involving transforming growth factor-β, endothelin-1, angiotensin II, CCN2 (connective tissue growth factor), and platelet-derived growth factor. This review addresses the contribution of these signaling molecules to cardiac fibrosis.
Collapse
Affiliation(s)
- Andrew Leask
- From the Departments of Dentistry and Physiology and Pharmacology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada.
| |
Collapse
|
40
|
Qin W, Du N, Zhang L, Wu X, Hu Y, Li X, Shen N, Li Y, Yang B, Xu C, Fang Z, Lu Y, Zhang Y, Du Z. Genistein alleviates pressure overload-induced cardiac dysfunction and interstitial fibrosis in mice. Br J Pharmacol 2015; 172:5559-72. [PMID: 25362897 DOI: 10.1111/bph.13002] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2014] [Revised: 10/23/2014] [Accepted: 10/28/2014] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND AND PURPOSE Pressure overload-induced cardiac interstitial fibrosis is viewed as a major cause of heart failure in patients with hypertension or aorta atherosclerosis. The purpose of this study was to investigate the effects and the underlying mechanisms of genistein, a natural phytoestrogen found in soy bean extract, on pressure overload-induced cardiac fibrosis. EXPERIMENTAL APPROACH Genisten was administered to mice with pressure overload induced by transverse aortic constriction. Eight weeks later, its effects on cardiac dysfunction, hypertrophy and fibrosis were determined. Its effects on proliferation, collagen production and myofibroblast transformation of cardiac fibroblasts (CFs) and the signalling pathways were also assessed in vitro. KEY RESULTS Pressure overload-induced cardiac dysfunction, hypertrophy and fibrosis were markedly attenuated by genistein. In cultured CFs, genistein inhibited TGFβ1-induced proliferation, collagen production and myofibroblast transformation. Genistein suppressed TGFβ-activated kinase 1 (TAK1) expression and produced anti-fibrotic effects by blocking the TAK1/MKK4/JNK pathway. Further analysis indicated that it up-regulated oestrogen-dependent expression of metastasis-associated gene 3 (MTA3), which was found to be a negative regulator of TAK1. Silencing MTA3 by siRNA, or inhibiting the activity of the MTA3-NuRD complex with trichostatin A, abolished genistein's anti-fibrotic effects. CONCLUSIONS AND IMPLICATIONS Genistein improved cardiac function and inhibited cardiac fibrosis in response to pressure overload. The underlying mechanism may involve regulation of the MTA3/TAK1/MKK4/JNK signalling pathway. Genistein may have potential as a novel agent for prevention and therapy of cardiac disorders associated with fibrosis.
Collapse
Affiliation(s)
- Wei Qin
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), Harbin Medical University, Harbin, Heilongjiang, China
| | - Ning Du
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), Harbin Medical University, Harbin, Heilongjiang, China
| | - Longyin Zhang
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), Harbin Medical University, Harbin, Heilongjiang, China
| | - Xianxian Wu
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), Harbin Medical University, Harbin, Heilongjiang, China
| | - Yingying Hu
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), Harbin Medical University, Harbin, Heilongjiang, China
| | - Xiaoguang Li
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), Harbin Medical University, Harbin, Heilongjiang, China
| | - Nannan Shen
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), Harbin Medical University, Harbin, Heilongjiang, China
| | - Yang Li
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), Harbin Medical University, Harbin, Heilongjiang, China
| | - Baofeng Yang
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), Harbin Medical University, Harbin, Heilongjiang, China.,Institute of Cardiovascular Research, Harbin Medical University, Harbin, Heilongjiang, China
| | - Chaoqian Xu
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), Harbin Medical University, Harbin, Heilongjiang, China
| | - Zhiwei Fang
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), Harbin Medical University, Harbin, Heilongjiang, China
| | - Yanjie Lu
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), Harbin Medical University, Harbin, Heilongjiang, China.,Institute of Cardiovascular Research, Harbin Medical University, Harbin, Heilongjiang, China
| | - Yong Zhang
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), Harbin Medical University, Harbin, Heilongjiang, China.,Institute of Cardiovascular Research, Harbin Medical University, Harbin, Heilongjiang, China
| | - Zhimin Du
- Institute of Clinical Pharmacy, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| |
Collapse
|
41
|
Abstract
Mitogen-activated protein kinases (MAPKs) mediate a wide variety of cellular behaviors in response to extracellular stimuli. One of the main subgroups, the p38 MAP kinases, has been implicated in a wide range of complex biologic processes, such as cell proliferation, cell differentiation, cell death, cell migration, and invasion. Dysregulation of p38 MAPK levels in patients are associated with advanced stages and short survival in cancer patients (e.g., prostate, breast, bladder, liver, and lung cancer). p38 MAPK plays a dual role as a regulator of cell death, and it can either mediate cell survival or cell death depending not only on the type of stimulus but also in a cell type specific manner. In addition to modulating cell survival, an essential role of p38 MAPK in modulation of cell migration and invasion offers a distinct opportunity to target this pathway with respect to tumor metastasis. The specific function of p38 MAPK appears to depend not only on the cell type but also on the stimuli and/or the isoform that is activated. p38 MAPK signaling pathway is activated in response to diverse stimuli and mediates its function by components downstream of p38. Extrapolation of the knowledge gained from laboratory findings is essential to address the clinical significance of p38 MAPK signaling pathways. The goal of this review is to provide an overview on recent progress made in defining the functions of p38 MAPK pathways with respect to solid tumor biology and generate testable hypothesis with respect to the role of p38 MAPK as an attractive target for intervention of solid tumors.
Collapse
Affiliation(s)
- Hari K Koul
- Department of Biochemistry & Molecular Biology, LSU Health Sciences Center, Shreveport, LA, USA ; Feist-Weiller Cancer Center, Shreveport, LA, USA ; Veterans Administration Medical Center, Shreveport, LA, USA
| | - Mantu Pal
- Department of Biochemistry & Molecular Biology, LSU Health Sciences Center, Shreveport, LA, USA ; Veterans Administration Medical Center, Shreveport, LA, USA
| | - Sweaty Koul
- Feist-Weiller Cancer Center, Shreveport, LA, USA ; Department of Urology, LSU Health Sciences Center, Shreveport, LA, USA
| |
Collapse
|
42
|
Abstract
Autophagy is a highly conserved homoeostatic mechanism for cell survival under conditions of stress, and is widely implicated as an important pathway in many biological processes and diseases. In progressive kidney diseases, fibrosis represents the common pathway to end-stage kidney failure. Transforming growth factor-β1 (TGF-β1) is a pleiotropic cytokine that has been established as a central mediator of kidney fibrosis. A recently emerging body of evidence from studies in renal cells in culture and experimental animal models suggests that TGF-β1 regulates autophagy and that autophagy regulates many critical aspects of normal and disease conditions associated with kidney fibrosis, such as tubulointerstitial fibrosis, glomerulosclerosis, and diabetic nephropathy. Here, we review the recent advances exploring the process of autophagy, its regulation by TGF-β1, and the implication in the pathogenesis of progressive kidney fibrosis and injury responses. Understanding the cellular and molecular bases of this process is crucial for identifying potential new diagnostic and therapeutic targets of kidney fibrosis.
Collapse
Affiliation(s)
- Yan Ding
- Renal Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Division of Nephrology and Hypertension, Weill Cornell Medical College, New York, NY
| | - Mary E Choi
- Renal Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Division of Nephrology and Hypertension, Weill Cornell Medical College, New York, NY.
| |
Collapse
|
43
|
Transcriptional corepressors HIPK1 and HIPK2 control angiogenesis via TGF-β-TAK1-dependent mechanism. PLoS Biol 2013; 11:e1001527. [PMID: 23565059 PMCID: PMC3614511 DOI: 10.1371/journal.pbio.1001527] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2012] [Accepted: 02/15/2013] [Indexed: 01/10/2023] Open
Abstract
Several critical events dictate the successful establishment of nascent vasculature in yolk sac and in the developing embryos. These include aggregation of angioblasts to form the primitive vascular plexus, followed by the proliferation, differentiation, migration, and coalescence of endothelial cells. Although transforming growth factor-β (TGF-β) is known to regulate various aspects of vascular development, the signaling mechanism of TGF-β remains unclear. Here we show that homeodomain interacting protein kinases, HIPK1 and HIPK2, are transcriptional corepressors that regulate TGF-β-dependent angiogenesis during embryonic development. Loss of HIPK1 and HIPK2 leads to marked up-regulations of several potent angiogenic genes, including Mmp10 and Vegf, which result in excessive endothelial proliferation and poor adherens junction formation. This robust phenotype can be recapitulated by siRNA knockdown of Hipk1 and Hipk2 in human umbilical vein endothelial cells, as well as in endothelial cell-specific TGF-β type II receptor (TβRII) conditional mutants. The effects of HIPK proteins are mediated through its interaction with MEF2C, and this interaction can be further enhanced by TGF-β in a TAK1-dependent manner. Remarkably, TGF-β-TAK1 signaling activates HIPK2 by phosphorylating a highly conserved tyrosine residue Y-361 within the kinase domain. Point mutation in this tyrosine completely eliminates the effect of HIPK2 as a transcriptional corepressor in luciferase assays. Our results reveal a previously unrecognized role of HIPK proteins in connecting TGF-β signaling pathway with the transcriptional programs critical for angiogenesis in early embryonic development.
Collapse
|
44
|
Abstract
In progressive kidney diseases, fibrosis represents the common pathway to end-stage kidney failure. Transforming growth factor-β1 (TGF-β1) is a pleiotropic cytokine that has been established as a central mediator of kidney fibrosis. Emerging evidence shows a complex scheme of signaling networks that enable multifunctionality of TGF-β1 actions. Specific targeting of the TGF-β signaling pathway is seemingly critical and an attractive molecular therapeutic strategy. TGF-β1 signals through the interaction of type I and type II receptors to activate distinct intracellular pathways involving the Smad and the non-Smad. The Smad signaling axis is known as the canonical pathway induced by TGF-β1. Importantly, recent investigations have shown that TGF-β1 also induces various non-Smad signaling pathways. In this review, we focus on current insights into the mechanism and function of the Smad-independent signaling pathway via TGF-β-activated kinase 1 and its role in mediating the profibrotic effects of TGF-β1.
Collapse
Affiliation(s)
- Mary E Choi
- Renal Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA.
| | | | | |
Collapse
|
45
|
TGF-β-activated kinase-1: New insights into the mechanism of TGF-β signaling and kidney disease. Kidney Res Clin Pract 2012; 31:94-105. [PMID: 26889415 PMCID: PMC4715161 DOI: 10.1016/j.krcp.2012.04.322] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2012] [Revised: 04/13/2012] [Accepted: 04/18/2012] [Indexed: 01/06/2023] Open
Abstract
Transforming growth factor-β (TGF-β) is a multifunctional cytokine that regulates a wide variety of cellular functions, including cell growth, cellular differentiation, apoptosis, and wound healing. TGF-β1, the prototype member of the TGF-β superfamily, is well established as a central mediator of renal fibrosis. In chronic kidney disease, dysregulation of expression and activation of TGF-β1 results in the relentless synthesis and accumulation of extracellular matrix proteins that lead to the development of glomerulosclerosis and tubulointerstitial fibrosis, and ultimately to end-stage renal disease. Therefore, specific targeting of the TGF-β signaling pathway is seemingly an attractive molecular therapeutic strategy in chronic kidney disease. Accumulating evidence demonstrates that the multifunctionality of TGF-β1 is connected with the complexity of its cell signaling networks. TGF-β1 signals through the interaction of type I and type II receptors to activate distinct intracellular pathways. Although the Smad signaling pathway is known as a canonical pathway induced by TGF-β1, and has been the focus of many previous reviews, importantly TGF-β1 also induces various Smad-independent signaling pathways. In this review, we describe evidence that supports current insights into the mechanism and function of TGF-β-activated kinase 1 (TAK1), which has emerged as a critical signaling molecule in TGF-β-induced Smad-independent signaling pathways. We also discuss the functional role of TAK1 in mediating the profibrotic effects of TGF-β1.
Collapse
|
46
|
Inhibition of transforming growth factor-activated kinase 1 (TAK1) blocks and reverses epithelial to mesenchymal transition of mesothelial cells. PLoS One 2012; 7:e31492. [PMID: 22384029 PMCID: PMC3288041 DOI: 10.1371/journal.pone.0031492] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2011] [Accepted: 01/09/2012] [Indexed: 02/01/2023] Open
Abstract
Peritoneal fibrosis is a frequent complication of peritoneal dialysis following repeated low grade inflammatory and pro-fibrotic insults. This pathological process may lead to ultrafiltration failure and eventually to the discontinuing of the therapy. Fibrosis is linked to epithelial to mesenchymal transition (EMT) of the peritoneal mesothelial cells, which acquire invasive and fibrogenic abilities. Here, we analyzed the role of the transforming growth factor-activated kinase-1 (TAK1) in the EMT of primary mesothelial cells from human peritoneum. The inhibition of TAK1 in mesenchymal-like mesothelial cells from the effluents of patients undergoing peritoneal dialysis led to the reacquisition of the apical to basolateral polarity, to increased expression of epithelial and to down-regulation of mesenchymal markers. TAK1 inhibition also resulted in decreased migratory/invasive abilities of effluent-derived mesothelial cells. Simultaneous inhibition of ERK1/2 and TAK1 pathways did not lead to an additive effect in the reacquisition of the epithelial phenotype. Inhibition of TAK1 also blocked EMT in vitro and reduced the levels of PAI-1, which is involved in fibrosis and invasion. Analysis of signalling pathways downstream of TAK1 involved in EMT induction, showed that TAK1 inhibition reduced the transcriptional activity of NF-κB and Smad3, as well as the phosphorylation of c-jun, while enhancing Smad1–5–8 activity. These results demonstrate that TAK1 is a cross-point in a network including different pro-EMT transcription factors, such as NF-κB, Snail, AP-1 and Smads. The identification of TAK1 as a main biochemical mediator of EMT and fibrosis in mesothelial cells from human peritoneum and the study of signalling pathways induced by its activity may be relevant in the design of new therapies aimed to counteract peritoneal fibrosis.
Collapse
|
47
|
Criollo A, Niso-Santano M, Malik SA, Michaud M, Morselli E, Mariño G, Lachkar S, Arkhipenko AV, Harper F, Pierron G, Rain JC, Ninomiya-Tsuji J, Fuentes JM, Lavandero S, Galluzzi L, Maiuri MC, Kroemer G. Inhibition of autophagy by TAB2 and TAB3. EMBO J 2011; 30:4908-20. [PMID: 22081109 DOI: 10.1038/emboj.2011.413] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2011] [Accepted: 10/24/2011] [Indexed: 12/18/2022] Open
Abstract
Autophagic responses are coupled to the activation of the inhibitor of NF-κB kinase (IKK). Here, we report that the essential autophagy mediator Beclin 1 and TGFβ-activated kinase 1 (TAK1)-binding proteins 2 and 3 (TAB2 and TAB3), two upstream activators of the TAK1-IKK signalling axis, constitutively interact with each other via their coiled-coil domains (CCDs). Upon autophagy induction, TAB2 and TAB3 dissociate from Beclin 1 and bind TAK1. Moreover, overexpression of TAB2 and TAB3 suppresses, while their depletion triggers, autophagy. The expression of the C-terminal domain of TAB2 or TAB3 or that of the CCD of Beclin 1 competitively disrupts the interaction between endogenous Beclin 1, TAB2 and TAB3, hence stimulating autophagy through a pathway that requires endogenous Beclin 1, TAK1 and IKK to be optimally efficient. These results point to the existence of an autophagy-stimulatory 'switch' whereby TAB2 and TAB3 abandon inhibitory interactions with Beclin 1 to engage in a stimulatory liaison with TAK1.
Collapse
|
48
|
Ma FY, Tesch GH, Ozols E, Xie M, Schneider MD, Nikolic-Paterson DJ. TGF-β1-activated kinase-1 regulates inflammation and fibrosis in the obstructed kidney. Am J Physiol Renal Physiol 2011; 300:F1410-21. [PMID: 21367917 DOI: 10.1152/ajprenal.00018.2011] [Citation(s) in RCA: 91] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Activation of c-Jun amino kinase (JNK), p38 mitogen-activated protein kinase (MAPK), and the transcription factor nuclear factor-κB (NF-κB) drives renal inflammation and fibrosis. However, the upstream MAP kinase kinase kinase (MAP3K) enzyme(s) that activate these pathways in kidney disease are unknown. We determined the role of one candidate MAP3K enzyme, transforming growth factor-β1-activated kinase-1 (TAK1/ MAP3K7), in activation of JNK, p38, and NF-κB in the obstructed kidney using conditional gene deletion in adult mice, and assessed the potential protective effect of TAK1 deletion on renal pathology. TAK1 deletion in cultured tubular epithelial cells substantially inhibited IL-1 and TNF-α-induced JNK, p38, and NF-κB signaling and the proinflammatory response. Map3k7(f/f)Cre-ER(TM) mice (in which tamoxifen induces global TAK1 deletion) and control Map3k7(f/f) mice were given tamoxifen at the time of unilateral ureteric obstruction (UUO) and then killed 2, 4, or 5 days later. Tamoxifen-treated control Map3k7(f/f) mice showed the expected activation of JNK, p38, and NF-κB signaling on days 2, 4, and 5, with macrophage infiltration and upregulation of mRNA levels of proinflammatory molecules (IL-1α, TNF-α, NOS2, and CCL2). Control Map3k7(f/f) mice also showed interstitial myofibroblast accumulation and collagen deposition in the obstructed kidney. Tamoxifen treatment of Map3k7(f/f)Cre-ER(TM) mice caused a 60% reduction in renal TAK1 expression on day 4 and >80% on day 5 UUO. Coincident with TAK1 deletion, activation of JNK, p38, and NF-κB signaling was markedly suppressed on days 4 to 5 UUO, which halted renal macrophage accumulation and expression of proinflammatory molecules. TAK1 deletion also halted the development of renal fibrosis in terms of myofibroblast accumulation, collagen deposition, and expression of profibrotic molecules. In conclusion, these studies establish TAK1 as a major upstream activator of JNK, p38, and NF-κB signaling in the obstructed kidney, and they define a pathologic role for TAK1 in renal inflammation and fibrosis.
Collapse
Affiliation(s)
- Frank Y Ma
- Department of Nephrology, Monash Medical Centre, Clayton, Victoria, Australia
| | | | | | | | | | | |
Collapse
|
49
|
Borthwick LA, McIlroy EI, Gorowiec MR, Brodlie M, Johnson GE, Ward C, Lordan JL, Corris PA, Kirby JA, Fisher AJ. Inflammation and epithelial to mesenchymal transition in lung transplant recipients: role in dysregulated epithelial wound repair. Am J Transplant 2010; 10:498-509. [PMID: 20055810 DOI: 10.1111/j.1600-6143.2009.02953.x] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Epithelial to mesenchymal transition (EMT) has been implicated in the pathogenesis of obliterative bronchiolitis (OB) after lung transplant. Although TNF-alpha accentuates TGF-beta1 driven EMT in primary human bronchial epithelial cells (PBECs), we hypothesized that other acute pro-inflammatory cytokines elevated in the airways of patients with OB may also accentuate EMT and contribute to dysregulated epithelial wound repair. PBECs from lung transplant recipients were stimulated with TGF-beta1+/-IL-1beta, IL-8, TNF-alpha or activated macrophages in co-culture and EMT assessed. The quality and rate of wound closure in a standardized model of lung epithelial injury was assessed in response to above stimuli. Co-treatment with TGF-beta1+TNF-alpha or IL-1beta significantly accentuates phenotypic and some functional features of EMT compared to TGF-beta1 alone. Co-treatment with TGF-beta1+TNF-alpha or IL-1beta accelerates epithelial wound closure however the quality of repair is highly dysregulated. Co-treatment with TGF-beta1+IL-8 has no significant effect on EMT or the speed or quality of wound healing. Activated macrophages dramatically accentuate TGF-beta1-driven EMT and cause dysregulated wound repair. Crosstalk between macrophage-derived acute inflammation in the airway and elevated TGF-beta1 may favor dysregulated airway epithelial repair and fibrosis in the lung allograft via EMT.
Collapse
Affiliation(s)
- L A Borthwick
- Applied Immunobiology and Transplantation Research Group, Institute of Cellular Medicine, Newcastle University, Newcastle, UK
| | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Kim SI, Kwak JH, Na HJ, Kim JK, Ding Y, Choi ME. Transforming growth factor-beta (TGF-beta1) activates TAK1 via TAB1-mediated autophosphorylation, independent of TGF-beta receptor kinase activity in mesangial cells. J Biol Chem 2009; 284:22285-22296. [PMID: 19556242 DOI: 10.1074/jbc.m109.007146] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Transforming growth factor-beta1 (TGF-beta1) is a multifunctional cytokine that signals through the interaction of type I (TbetaRI) and type II (TbetaRII) receptors to activate distinct intracellular pathways. TAK1 is a serine/threonine kinase that is rapidly activated by TGF-beta1. However, the molecular mechanism of TAK1 activation is incompletely understood. Here, we propose a mechanism whereby TAK1 is activated by TGF-beta1 in primary mouse mesangial cells. Under unstimulated conditions, endogenous TAK1 is stably associated with TbetaRI. TGF-beta1 stimulation causes rapid dissociation from the receptor and induces TAK1 phosphorylation. Deletion mutant analysis indicates that the juxtamembrane region including the GS domain of TbetaRI is crucial for its interaction with TAK1. Both TbetaRI-mediated TAK1 phosphorylation and TGF-beta1-induced TAK1 phosphorylation do not require kinase activity of TbetaRI. Moreover, TbetaRI-mediated TAK1 phosphorylation correlates with the degree of its association with TbetaRI and requires kinase activity of TAK1. TAB1 does not interact with TGF-beta receptors, but TAB1 is indispensable for TGF-beta1-induced TAK1 activation. We also show that TRAF6 and TAB2 are required for the interaction of TAK1 with TbetaRI and TGF-beta1-induced TAK1 activation in mouse mesangial cells. Taken together, our data indicate that TGF-beta1-induced interaction of TbetaRI and TbetaRII triggers dissociation of TAK1 from TbetaRI, and subsequently TAK1 is phosphorylated through TAB1-mediated autophosphorylation and not by the receptor kinase activity of TbetaRI.
Collapse
Affiliation(s)
- Sung Il Kim
- Renal Division, Department of Medicine, Brigham and Womens Hospital, Harvard Medical School, Boston, Massachusetts 02115
| | - Joon Hyeok Kwak
- Renal Division, Department of Medicine, Brigham and Womens Hospital, Harvard Medical School, Boston, Massachusetts 02115
| | - Hee-Jun Na
- Renal Division, Department of Medicine, Brigham and Womens Hospital, Harvard Medical School, Boston, Massachusetts 02115
| | - Jin Kuk Kim
- Renal Division, Department of Medicine, Brigham and Womens Hospital, Harvard Medical School, Boston, Massachusetts 02115; Department of Internal Medicine, Bucheon Hospital, Soonchunhyang University, Bucheon, 420-767, Korea
| | - Yan Ding
- Renal Division, Department of Medicine, Brigham and Womens Hospital, Harvard Medical School, Boston, Massachusetts 02115
| | - Mary E Choi
- Renal Division, Department of Medicine, Brigham and Womens Hospital, Harvard Medical School, Boston, Massachusetts 02115
| |
Collapse
|