1
|
Tran DT, Batchu SN, Advani A. Interferons and interferon-related pathways in heart disease. Front Cardiovasc Med 2024; 11:1357343. [PMID: 38665231 PMCID: PMC11043610 DOI: 10.3389/fcvm.2024.1357343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2023] [Accepted: 04/01/2024] [Indexed: 04/28/2024] Open
Abstract
Interferons (IFNs) and IFN-related pathways play key roles in the defence against microbial infection. However, these processes may also be activated during the pathogenesis of non-infectious diseases, where they may contribute to organ injury, or function in a compensatory manner. In this review, we explore the roles of IFNs and IFN-related pathways in heart disease. We consider the cardiac effects of type I IFNs and IFN-stimulated genes (ISGs); the emerging role of the cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) pathway; the seemingly paradoxical effects of the type II IFN, IFN-γ; and the varied actions of the interferon regulatory factor (IRF) family of transcription factors. Recombinant IFNs and small molecule inhibitors of mediators of IFN receptor signaling are already employed in the clinic for the treatment of some autoimmune diseases, infections, and cancers. There has also been renewed interest in IFNs and IFN-related pathways because of their involvement in SARS-CoV-2 infection, and because of the relatively recent emergence of cGAS-STING as a pattern recognition receptor-activated pathway. Whether these advances will ultimately result in improvements in the care of those experiencing heart disease remains to be determined.
Collapse
Affiliation(s)
| | | | - Andrew Advani
- Keenan Research Centre for Biomedical Science and Li Ka Shing Knowledge Institute, St. Michael’s Hospital, Toronto, ON, Canada
| |
Collapse
|
2
|
Jiao H, James SJ, Png CW, Cui C, Li H, Li L, Chia WN, Min N, Li W, Claser C, Rénia L, Wang H, Chen MIC, Chu JJH, Tan KSW, Deng Y, Zhang Y. DUSP4 modulates RIG-I- and STING-mediated IRF3-type I IFN response. Cell Death Differ 2024; 31:280-291. [PMID: 38383887 PMCID: PMC10923883 DOI: 10.1038/s41418-024-01269-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 02/05/2024] [Accepted: 02/07/2024] [Indexed: 02/23/2024] Open
Abstract
Detection of cytosolic nucleic acids by pattern recognition receptors, including STING and RIG-I, leads to the activation of multiple signalling pathways that culminate in the production of type I interferons (IFNs) which are vital for host survival during virus infection. In addition to protective immune modulatory functions, type I IFNs are also associated with autoimmune diseases. Hence, it is important to elucidate the mechanisms that govern their expression. In this study, we identified a critical regulatory function of the DUSP4 phosphatase in innate immune signalling. We found that DUSP4 regulates the activation of TBK1 and ERK1/2 in a signalling complex containing DUSP4, TBK1, ERK1/2 and IRF3 to regulate the production of type I IFNs. Mice deficient in DUSP4 were more resistant to infections by both RNA and DNA viruses but more susceptible to malaria parasites. Therefore, our study establishes DUSP4 as a regulator of nucleic acid sensor signalling and sheds light on an important facet of the type I IFN regulatory system.
Collapse
Affiliation(s)
- Huipeng Jiao
- Zhejiang Provincial Key Laboratory of Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang University, Hangzhou, Zhejiang, 310058, China
- Department of Microbiology and Immunology, TRP Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
- Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore, 117597, Singapore
| | - Sharmy J James
- Department of Microbiology and Immunology, TRP Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
- Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore, 117597, Singapore
| | - Chin Wen Png
- Department of Microbiology and Immunology, TRP Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
- Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore, 117597, Singapore
| | - Chaoyu Cui
- Department of Microbiology and Immunology, TRP Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, 518100, China
| | - Heng Li
- Department of Microbiology and Immunology, TRP Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
- Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore, 117597, Singapore
| | - Liang Li
- Department of Pharmacology, School of Medicine, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Wan Ni Chia
- Department of Microbiology and Immunology, TRP Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
| | - Nyo Min
- Department of Microbiology and Immunology, TRP Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
| | - Weiyun Li
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Innovation Center for Cell Signaling Network, Shanghai, 200031, China
| | - Carla Claser
- Singapore Immunology Network, Agency for Science, Technology and Research (A*STAR), Singapore, 138668, Singapore
| | - Laurent Rénia
- Singapore Immunology Network, Agency for Science, Technology and Research (A*STAR), Singapore, 138668, Singapore
| | - Hongyan Wang
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Innovation Center for Cell Signaling Network, Shanghai, 200031, China
| | - Mark I-Cheng Chen
- Saw Swee Hock School of Public Health, National University of Singapore, Singapore, 117597, Singapore
| | - Justin Jang Hann Chu
- Department of Microbiology and Immunology, TRP Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
| | - Kevin Shyong Wei Tan
- Department of Microbiology and Immunology, TRP Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
| | - Yinyue Deng
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, 518100, China.
| | - Yongliang Zhang
- Department of Microbiology and Immunology, TRP Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore.
- Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore, 117597, Singapore.
| |
Collapse
|
3
|
Parichatikanond W, Duangrat R, Mangmool S. G αq protein-biased ligand of angiotensin II type 1 receptor mediates myofibroblast differentiation through TGF-β1/ERK axis in human cardiac fibroblasts. Eur J Pharmacol 2023; 951:175780. [PMID: 37209939 DOI: 10.1016/j.ejphar.2023.175780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 04/28/2023] [Accepted: 05/10/2023] [Indexed: 05/22/2023]
Abstract
Angiotensin II receptors are members of G protein-coupled receptor superfamily that manifest biased signals toward G protein- and β-arrestin-dependent pathways. However, the role of angiotensin II receptor-biased ligands and the mechanisms underlying myofibroblast differentiation in human cardiac fibroblasts have not been fully elucidated. Our results demonstrated that antagonism of angiotensin II type 1 receptor (AT1 receptor) and blockade of Gαq protein suppressed angiotensin II (Ang II)-induced fibroblast proliferation, overexpression of collagen I and α-smooth muscle actin (α-SMA), and stress fibre formation, indicating the AT1 receptor/Gαq axis is necessary for fibrogenic effects of Ang II. Stimulation of AT1 receptors by their Gαq-biased ligand (TRV120055), but not β-arrestin-biased ligand (TRV120027), substantially exerted fibrogenic effects at a level similar to that of Ang II, suggesting that AT1 receptor induced cardiac fibrosis in a Gαq-dependent and β-arrestin-independent manner. Valsartan prevents TRV120055-mediated fibroblast activation. TRV120055 mediated the upregulation of transforming growth factor-beta1 (TGF-β1) through the AT1 receptor/Gαq cascade. In addition, Gαq protein and TGF-β1 were necessary for ERK1/2 activation induced by Ang II and TRV120055. Collectively, TGF-β1 and ERK1/2 are downstream effectors of the Gαq-biased ligand of AT1 receptor for the induction of cardiac fibrosis.
Collapse
Affiliation(s)
- Warisara Parichatikanond
- Department of Pharmacology, Faculty of Pharmacy, Mahidol University, Bangkok, 10400, Thailand; Centre of Biopharmaceutical Science for Healthy Ageing (BSHA), Faculty of Pharmacy, Mahidol University, Bangkok, 10400, Thailand.
| | - Ratchanee Duangrat
- Department of Pharmacology, Faculty of Science, Mahidol University, Bangkok, 10400, Thailand; Molecular Medicine Graduate Program, Faculty of Science, Mahidol University, Bangkok, 10400, Thailand.
| | - Supachoke Mangmool
- Department of Pharmacology, Faculty of Science, Mahidol University, Bangkok, 10400, Thailand.
| |
Collapse
|
4
|
Duangrat R, Parichatikanond W, Morales NP, Pinthong D, Mangmool S. Sustained AT1R stimulation induces upregulation of growth factors in human cardiac fibroblasts via Gαq/TGF-β/ERK signaling that influences myocyte hypertrophy. Eur J Pharmacol 2022; 937:175384. [DOI: 10.1016/j.ejphar.2022.175384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 11/03/2022] [Accepted: 11/04/2022] [Indexed: 11/13/2022]
|
5
|
Pothen L, Verdoy R, De Mulder D, Esfahani H, Farah C, Michel LYM, Dei Zotti F, Bearzatto B, Ambroise J, Bouzin C, Dessy C, Balligand JL. Sustained Downregulation of Vascular Smooth Muscle Acta2 After Transient Angiotensin II Infusion: A New Model of "Vascular Memory". Front Cardiovasc Med 2022; 9:854361. [PMID: 35360022 PMCID: PMC8964264 DOI: 10.3389/fcvm.2022.854361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Accepted: 02/14/2022] [Indexed: 11/19/2022] Open
Abstract
Background Activation of the renin-angiotensin-aldosterone system (RAAS) plays a critical role in the development of hypertension. Published evidence on a putative "memory effect" of AngII on the vascular components is however scarce. Aim To evaluate the long-term effects of transient exposure to AngII on the mouse heart and the arterial tissue. Methods Blood pressure, cardiovascular tissue damage and remodeling, and systemic oxidative stress were evaluated in C57/B6/J mice at the end of a 2-week AngII infusion (AngII); 2 and 3 weeks after the interruption of a 2-week AngII treatment (AngII+2W and AngII +3W; so-called "memory" conditions) and control littermate (CTRL). RNAseq profiling of aortic tissues was used to identify potential key regulated genes accounting for legacy effects on the vascular phenotype. RNAseq results were validated by RT-qPCR and immunohistochemistry in a reproduction cohort of mice. Key findings were reproduced in a homotypic cell culture model. Results The 2 weeks AngII infusion induced cardiac hypertrophy and aortic damage that persisted beyond AngII interruption and despite blood pressure normalization, with a sustained vascular expression of ICAM1, infiltration by CD45+ cells, and cell proliferation associated with systemic oxidative stress. RNAseq profiling in aortic tissue identified robust Acta2 downregulation at transcript and protein levels (α-smooth muscle actin) that was maintained beyond interruption of AngII treatment. Among regulators of Acta2 expression, the transcription factor Myocardin (Myocd), exhibited a similar expression pattern. The sustained downregulation of Acta2 and Myocd was associated with an increase in H3K27me3 in nuclei of aortic sections from mice in the "memory" conditions. A sustained downregulation of ACTA2 and MYOCD was reproduced in the cultured human aortic vascular smooth muscle cells upon transient exposure to Ang II. Conclusion A transient exposure to Ang II produces prolonged vascular remodeling with robust ACTA2 downregulation, associated with epigenetic imprinting supporting a "memory" effect despite stimulus withdrawal.
Collapse
Affiliation(s)
- Lucie Pothen
- Institute of Experimental and Clinical Research (IREC), Pole of Pharmacology and Therapeutics (FATH), Cliniques Universitaires St-Luc and Université Catholique de Louvain (UCLouvain), Brussels, Belgium
| | - Roxane Verdoy
- Institute of Experimental and Clinical Research (IREC), Pole of Pharmacology and Therapeutics (FATH), Cliniques Universitaires St-Luc and Université Catholique de Louvain (UCLouvain), Brussels, Belgium
| | - Delphine De Mulder
- Institute of Experimental and Clinical Research (IREC), Pole of Pharmacology and Therapeutics (FATH), Cliniques Universitaires St-Luc and Université Catholique de Louvain (UCLouvain), Brussels, Belgium
| | - Hrag Esfahani
- Institute of Experimental and Clinical Research (IREC), Pole of Pharmacology and Therapeutics (FATH), Cliniques Universitaires St-Luc and Université Catholique de Louvain (UCLouvain), Brussels, Belgium
| | - Charlotte Farah
- Institute of Experimental and Clinical Research (IREC), Pole of Pharmacology and Therapeutics (FATH), Cliniques Universitaires St-Luc and Université Catholique de Louvain (UCLouvain), Brussels, Belgium
| | - Lauriane Y. M. Michel
- Institute of Experimental and Clinical Research (IREC), Pole of Pharmacology and Therapeutics (FATH), Cliniques Universitaires St-Luc and Université Catholique de Louvain (UCLouvain), Brussels, Belgium
| | - Flavia Dei Zotti
- Institute of Experimental and Clinical Research (IREC), Pole of Pharmacology and Therapeutics (FATH), Cliniques Universitaires St-Luc and Université Catholique de Louvain (UCLouvain), Brussels, Belgium
| | - Bertrand Bearzatto
- Institute of Experimental and Clinical Research (IREC), Centre des Technologies Moléculaires Appliquées (CTMA), Cliniques Universitaires St-Luc and Université Catholique de Louvain (UCLouvain), Brussels, Belgium
| | - Jerome Ambroise
- Institute of Experimental and Clinical Research (IREC), Centre des Technologies Moléculaires Appliquées (CTMA), Cliniques Universitaires St-Luc and Université Catholique de Louvain (UCLouvain), Brussels, Belgium
| | - Caroline Bouzin
- Institute of Experimental and Clinical Research (IREC), Imaging Platform (2IP), Cliniques Universitaires St-Luc and Université Catholique de Louvain (UCLouvain), Brussels, Belgium
| | - Chantal Dessy
- Institute of Experimental and Clinical Research (IREC), Pole of Pharmacology and Therapeutics (FATH), Cliniques Universitaires St-Luc and Université Catholique de Louvain (UCLouvain), Brussels, Belgium
| | - Jean-Luc Balligand
- Institute of Experimental and Clinical Research (IREC), Pole of Pharmacology and Therapeutics (FATH), Cliniques Universitaires St-Luc and Université Catholique de Louvain (UCLouvain), Brussels, Belgium
| |
Collapse
|
6
|
Abstract
Cells respond to stress by activating a variety of defense signaling pathways, including cell survival and cell death pathways. Although cell survival signaling helps the cell to recover from acute insults, cell death or senescence pathways induced by chronic insults can lead to unresolved pathologies. Arterial hypertension results from chronic physiological maladaptation against various stressors represented by abnormal circulating or local neurohormonal factors, mechanical stress, intracellular accumulation of toxic molecules, and dysfunctional organelles. Hypertension and aging share common mechanisms that mediate or prolong chronic cell stress, such as endoplasmic reticulum stress and accumulation of protein aggregates, oxidative stress, metabolic mitochondrial stress, DNA damage, stress-induced senescence, and proinflammatory processes. This review discusses common adaptive signaling mechanisms against these stresses including unfolded protein responses, antioxidant response element signaling, autophagy, mitophagy, and mitochondrial fission/fusion, STING (signaling effector stimulator of interferon genes)-mediated responses, and activation of pattern recognition receptors. The main molecular mechanisms by which the vasculature copes with hypertensive and aging stressors are presented and recent advancements in stress-adaptive signaling mechanisms as well as potential therapeutic targets are discussed.
Collapse
Affiliation(s)
- Stephanie M. Cicalese
- These authors contributed equally and are considered co-first authors
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania, USA
| | - Josiane Fernandes da Silva
- These authors contributed equally and are considered co-first authors
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, SP, Brazil
| | - Fernanda Priviero
- These authors contributed equally and are considered co-first authors
- Cardiovascular Translational Research Center and Department of Cell Biology and Anatomy, University of South Carolina, Columbia, South Carolina, USA
| | - R. Clinton Webb
- Cardiovascular Translational Research Center and Department of Cell Biology and Anatomy, University of South Carolina, Columbia, South Carolina, USA
| | - Satoru Eguchi
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania, USA
| | - Rita C. Tostes
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, SP, Brazil
| |
Collapse
|
7
|
Angiotensin II-induced cardiomyocyte hypertrophy: A complex response dependent on intertwined pathways. REVISTA PORTUGUESA DE CARDIOLOGIA (ENGLISH EDITION) 2021. [DOI: 10.1016/j.repce.2021.03.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|
8
|
Consegal M, Valls-Lacalle L, Rodríguez-Sinovas A. Angiotensin II-induced cardiomyocyte hypertrophy: A complex response dependent on intertwined pathways. Rev Port Cardiol 2021; 40:201-203. [PMID: 33478847 DOI: 10.1016/j.repc.2020.12.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
Affiliation(s)
- Marta Consegal
- Cardiovascular Diseases Research Group, Department of Cardiology, Vall d'Hebron University Hospital and Research Institute, Universitat Autònoma de Barcelona, Departament de Medicina, Pg. Vall d'Hebron 119-129, 08035 Barcelona, Spain; Centro de Investigación Biomédica en Red sobre Enfermedades Cardiovasculares (CIBERCV), Spain
| | - Laura Valls-Lacalle
- Biomaterials for Regenerative Therapies Group, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology, 08028 Barcelona, Spain
| | - Antonio Rodríguez-Sinovas
- Cardiovascular Diseases Research Group, Department of Cardiology, Vall d'Hebron University Hospital and Research Institute, Universitat Autònoma de Barcelona, Departament de Medicina, Pg. Vall d'Hebron 119-129, 08035 Barcelona, Spain; Centro de Investigación Biomédica en Red sobre Enfermedades Cardiovasculares (CIBERCV), Spain.
| |
Collapse
|
9
|
Current potential therapeutic strategies targeting the TGF-β/Smad signaling pathway to attenuate keloid and hypertrophic scar formation. Biomed Pharmacother 2020; 129:110287. [PMID: 32540643 DOI: 10.1016/j.biopha.2020.110287] [Citation(s) in RCA: 227] [Impact Index Per Article: 45.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Revised: 05/08/2020] [Accepted: 05/16/2020] [Indexed: 12/12/2022] Open
Abstract
Aberrant scar formation, which includes keloid and hypertrophic scars, is associated with a pathological disorganized wound healing process with chronic inflammation. The TGF-β/Smad signaling pathway is the most canonical pathway through which the formation of collagen in the fibroblasts and myofibroblasts is regulated. Sustained activation of the TGF-β/Smad signaling pathway results in the long-term overactivation of fibroblasts and myofibroblasts, which is necessary for the excessive collagen formation in aberrant scars. There are two categories of therapeutic strategies that aim to target the TGF-β/Smad signaling pathway in fibroblasts and myofibroblasts to interfere with their cellular functions and reduce cell proliferation. The first therapeutic strategy includes medications, and the second strategy is composed of genetic and cellular therapeutics. Therefore, the focus of this review is to critically evaluate these two main therapeutic strategies that target the TGF-β/Smad pathway to attenuate abnormal skin scar formation.
Collapse
|
10
|
Uchida L, Tanaka T, Saito H, Sugahara M, Wakashima T, Fukui K, Nangaku M. Effects of a prolyl hydroxylase inhibitor on kidney and cardiovascular complications in a rat model of chronic kidney disease. Am J Physiol Renal Physiol 2019; 318:F388-F401. [PMID: 31841388 DOI: 10.1152/ajprenal.00419.2019] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Cardiovascular disease (CVD) is the main cause of death in patients with kidney disease. Hypoxia plays a crucial role in the progression of chronic kidney disease (CKD) and cardiovascular disease, which is associated with fibrosis, inflammation, and oxidative injury. Previous studies have indicated that prolyl hydroxylase (PHD) inhibitors, stabilizers of hypoxia-inducible factors (HIFs), can be used to treat acute organ injuries such as renal ischemia-reperfusion, myocardial infarction, and, in some contexts, CKD. However, the effects of PHD inhibitors on cardiovascular complications in CKD remain unknown. In the present study, we investigated whether HIF activation has a beneficial effect on kidney and cardiovascular outcomes in the remnant kidney model. We used the 5/6 nephrectomy model with the nitric oxide synthase inhibitor Nω-nitro-l-arginine (20 mg/L in the drinking water). Rats received diet with 0.005% enarodustat (PHD inhibitor) or vehicle for 8 wk starting 2 wk before 5/6 nephrectomy. Activation of HIF by the PHD inhibitor reduced cardiac hypertrophy and ameliorated myocardial fibrosis in association with restored capillary density and improvement in mitochondrial morphology. With regard to kidneys, enarodustat ameliorated fibrosis in association with reduced proinflammatory cytokine expression, reduced apoptosis, and restored capillary density, even though renal endpoints such as proteinuria and serum creatinine levels were not significantly affected by enarodustat, except for blood urea nitrogen levels at 4 wk. In addition, cardiac hypertrophy marker genes, including atrial natriuretic peptide, were suppressed in P19CL6 cells treated with enarodustat. These findings suggest that PHD inhibitors might show beneficial effects in cardiovascular complications caused by CKD.
Collapse
Affiliation(s)
- Lisa Uchida
- Division of Nephrology and Endocrinology, the University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | - Tetsuhiro Tanaka
- Division of Nephrology and Endocrinology, the University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | - Hisako Saito
- Division of Nephrology and Endocrinology, the University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | - Mai Sugahara
- Division of Nephrology and Endocrinology, the University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | - Takeshi Wakashima
- Division of Nephrology and Endocrinology, the University of Tokyo Graduate School of Medicine, Tokyo, Japan.,Biological and Pharmacological Research Laboratories, Central Pharmaceutical Research Institute, Japan Tobacco Incorporated, Takatsuki, Japan
| | - Kenji Fukui
- Division of Nephrology and Endocrinology, the University of Tokyo Graduate School of Medicine, Tokyo, Japan.,Biological and Pharmacological Research Laboratories, Central Pharmaceutical Research Institute, Japan Tobacco Incorporated, Takatsuki, Japan
| | - Masaomi Nangaku
- Division of Nephrology and Endocrinology, the University of Tokyo Graduate School of Medicine, Tokyo, Japan
| |
Collapse
|
11
|
Opposite Effects of Moderate and Extreme Cx43 Deficiency in Conditional Cx43-Deficient Mice on Angiotensin II-Induced Cardiac Fibrosis. Cells 2019; 8:cells8101299. [PMID: 31652649 PMCID: PMC6830333 DOI: 10.3390/cells8101299] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 10/18/2019] [Accepted: 10/21/2019] [Indexed: 01/27/2023] Open
Abstract
Connexin 43 (Cx43) is essential for cardiac electrical coupling, but its effects on myocardial fibrosis is controversial. Here, we analyzed the role of Cx43 in myocardial fibrosis caused by angiotensin II (AngII) using Cx43fl/fl and Cx43Cre-ER(T)/fl inducible knock-out (Cx43 content: 50%) mice treated with vehicle or 4-hydroxytamoxifen (4-OHT) to induce a Cre-ER(T)-mediated global deletion of the Cx43 floxed allele. Myocardial collagen content was enhanced by AngII in all groups (n = 8–10/group, p < 0.05). However, animals with partial Cx43 deficiency (vehicle-treated Cx43Cre-ER(T)/fl) had a significantly higher AngII-induced collagen accumulation that reverted when treated with 4-OHT, which abolished Cx43 expression. The exaggerated fibrotic response to AngII in partially deficient Cx43Cre-ER(T)/fl mice was associated with enhanced p38 MAPK activation and was not evident in Cx43 heterozygous (Cx43+/-) mice. In contrast, normalization of interstitial collagen in 4-OHT-treated Cx43Cre-ER(T)/fl animals correlated with enhanced MMP-9 activity, IL-6 and NOX2 mRNA expression, and macrophage content, and with reduced α-SMA and SM22α in isolated fibroblasts. In conclusion, our data demonstrates an exaggerated, p38 MAPK-dependent, fibrotic response to AngII in partially deficient Cx43Cre-ER(T)/fl mice, and a paradoxical normalization of collagen deposition in animals with an almost complete Cx43 ablation, an effect associated with increased MMP-9 activity and inflammatory response and reduced fibroblasts differentiation.
Collapse
|
12
|
Wang J, Duan Y, Sluijter JPG, Xiao J. Lymphocytic subsets play distinct roles in heart diseases. Am J Cancer Res 2019; 9:4030-4046. [PMID: 31281530 PMCID: PMC6592175 DOI: 10.7150/thno.33112] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Accepted: 05/06/2019] [Indexed: 12/24/2022] Open
Abstract
Heart diseases are one of the leading causes of death for humans in the world. Increasing evidence has shown that myocardial injury induced innate and adaptive immune responses upon early cellular damage but also during chronic phases post-injury. The immune cells can not only aggravate the injury but also play an essential role in the induction of wound healing responses, which means they play a complex role throughout the acute inflammatory response and reparative response after cardiac injury. This review will summarize the current experimental and clinical evidence of lymphocytes, one of the major types of immune cells, participate in heart diseases and try to explain the possible role of these immune cells following cardiac injury.
Collapse
|
13
|
Forrester SJ, Booz GW, Sigmund CD, Coffman TM, Kawai T, Rizzo V, Scalia R, Eguchi S. Angiotensin II Signal Transduction: An Update on Mechanisms of Physiology and Pathophysiology. Physiol Rev 2018; 98:1627-1738. [PMID: 29873596 DOI: 10.1152/physrev.00038.2017] [Citation(s) in RCA: 727] [Impact Index Per Article: 103.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The renin-angiotensin-aldosterone system plays crucial roles in cardiovascular physiology and pathophysiology. However, many of the signaling mechanisms have been unclear. The angiotensin II (ANG II) type 1 receptor (AT1R) is believed to mediate most functions of ANG II in the system. AT1R utilizes various signal transduction cascades causing hypertension, cardiovascular remodeling, and end organ damage. Moreover, functional cross-talk between AT1R signaling pathways and other signaling pathways have been recognized. Accumulating evidence reveals the complexity of ANG II signal transduction in pathophysiology of the vasculature, heart, kidney, and brain, as well as several pathophysiological features, including inflammation, metabolic dysfunction, and aging. In this review, we provide a comprehensive update of the ANG II receptor signaling events and their functional significances for potential translation into therapeutic strategies. AT1R remains central to the system in mediating physiological and pathophysiological functions of ANG II, and participation of specific signaling pathways becomes much clearer. There are still certain limitations and many controversies, and several noteworthy new concepts require further support. However, it is expected that rigorous translational research of the ANG II signaling pathways including those in large animals and humans will contribute to establishing effective new therapies against various diseases.
Collapse
Affiliation(s)
- Steven J Forrester
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University , Philadelphia, Pennsylvania ; Department of Pharmacology and Toxicology, School of Medicine, University of Mississippi Medical Center , Jackson, Mississippi ; Department of Pharmacology, Center for Hypertension Research, Roy J. and Lucille A. Carver College of Medicine, University of Iowa , Iowa City, Iowa ; and Duke-NUS, Singapore and Department of Medicine, Duke University Medical Center , Durham, North Carolina
| | - George W Booz
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University , Philadelphia, Pennsylvania ; Department of Pharmacology and Toxicology, School of Medicine, University of Mississippi Medical Center , Jackson, Mississippi ; Department of Pharmacology, Center for Hypertension Research, Roy J. and Lucille A. Carver College of Medicine, University of Iowa , Iowa City, Iowa ; and Duke-NUS, Singapore and Department of Medicine, Duke University Medical Center , Durham, North Carolina
| | - Curt D Sigmund
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University , Philadelphia, Pennsylvania ; Department of Pharmacology and Toxicology, School of Medicine, University of Mississippi Medical Center , Jackson, Mississippi ; Department of Pharmacology, Center for Hypertension Research, Roy J. and Lucille A. Carver College of Medicine, University of Iowa , Iowa City, Iowa ; and Duke-NUS, Singapore and Department of Medicine, Duke University Medical Center , Durham, North Carolina
| | - Thomas M Coffman
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University , Philadelphia, Pennsylvania ; Department of Pharmacology and Toxicology, School of Medicine, University of Mississippi Medical Center , Jackson, Mississippi ; Department of Pharmacology, Center for Hypertension Research, Roy J. and Lucille A. Carver College of Medicine, University of Iowa , Iowa City, Iowa ; and Duke-NUS, Singapore and Department of Medicine, Duke University Medical Center , Durham, North Carolina
| | - Tatsuo Kawai
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University , Philadelphia, Pennsylvania ; Department of Pharmacology and Toxicology, School of Medicine, University of Mississippi Medical Center , Jackson, Mississippi ; Department of Pharmacology, Center for Hypertension Research, Roy J. and Lucille A. Carver College of Medicine, University of Iowa , Iowa City, Iowa ; and Duke-NUS, Singapore and Department of Medicine, Duke University Medical Center , Durham, North Carolina
| | - Victor Rizzo
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University , Philadelphia, Pennsylvania ; Department of Pharmacology and Toxicology, School of Medicine, University of Mississippi Medical Center , Jackson, Mississippi ; Department of Pharmacology, Center for Hypertension Research, Roy J. and Lucille A. Carver College of Medicine, University of Iowa , Iowa City, Iowa ; and Duke-NUS, Singapore and Department of Medicine, Duke University Medical Center , Durham, North Carolina
| | - Rosario Scalia
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University , Philadelphia, Pennsylvania ; Department of Pharmacology and Toxicology, School of Medicine, University of Mississippi Medical Center , Jackson, Mississippi ; Department of Pharmacology, Center for Hypertension Research, Roy J. and Lucille A. Carver College of Medicine, University of Iowa , Iowa City, Iowa ; and Duke-NUS, Singapore and Department of Medicine, Duke University Medical Center , Durham, North Carolina
| | - Satoru Eguchi
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University , Philadelphia, Pennsylvania ; Department of Pharmacology and Toxicology, School of Medicine, University of Mississippi Medical Center , Jackson, Mississippi ; Department of Pharmacology, Center for Hypertension Research, Roy J. and Lucille A. Carver College of Medicine, University of Iowa , Iowa City, Iowa ; and Duke-NUS, Singapore and Department of Medicine, Duke University Medical Center , Durham, North Carolina
| |
Collapse
|
14
|
Angiotensin II requires an intact cardiac thyrotropin-releasing hormone (TRH) system to induce cardiac hypertrophy in mouse. J Mol Cell Cardiol 2018; 124:1-11. [DOI: 10.1016/j.yjmcc.2018.09.009] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Revised: 08/13/2018] [Accepted: 09/25/2018] [Indexed: 11/18/2022]
|
15
|
Rysä J, Tokola H, Ruskoaho H. Mechanical stretch induced transcriptomic profiles in cardiac myocytes. Sci Rep 2018; 8:4733. [PMID: 29549296 PMCID: PMC5856749 DOI: 10.1038/s41598-018-23042-w] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Accepted: 03/06/2018] [Indexed: 12/15/2022] Open
Abstract
Mechanical forces are able to activate hypertrophic growth of cardiomyocytes in the overloaded myocardium. However, the transcriptional profiles triggered by mechanical stretch in cardiac myocytes are not fully understood. Here, we performed the first genome-wide time series study of gene expression changes in stretched cultured neonatal rat ventricular myocytes (NRVM)s, resulting in 205, 579, 737, 621, and 1542 differentially expressed (>2-fold, P < 0.05) genes in response to 1, 4, 12, 24, and 48 hours of cyclic mechanical stretch. We used Ingenuity Pathway Analysis to predict functional pathways and upstream regulators of differentially expressed genes in order to identify regulatory networks that may lead to mechanical stretch induced hypertrophic growth of cardiomyocytes. We also performed micro (miRNA) expression profiling of stretched NRVMs, and identified that a total of 8 and 87 miRNAs were significantly (P < 0.05) altered by 1-12 and 24-48 hours of mechanical stretch, respectively. Finally, through integration of miRNA and mRNA data, we predicted the miRNAs that regulate mRNAs potentially leading to the hypertrophic growth induced by mechanical stretch. These analyses predicted nuclear factor-like 2 (Nrf2) and interferon regulatory transcription factors as well as the let-7 family of miRNAs as playing roles in the regulation of stretch-regulated genes in cardiomyocytes.
Collapse
Affiliation(s)
- Jaana Rysä
- School of Pharmacy, University of Eastern Finland, Kuopio, Finland.
- Research Unit of Biomedicine, Pharmacology and Toxicology, University of Oulu, Oulu, Finland.
| | - Heikki Tokola
- Research Unit of Biomedicine, Pharmacology and Toxicology, University of Oulu, Oulu, Finland
- Department of Pathology, Cancer Research and Translational Medicine Research Unit, University of Oulu and Oulu University Hospital, Oulu, Finland
| | - Heikki Ruskoaho
- Research Unit of Biomedicine, Pharmacology and Toxicology, University of Oulu, Oulu, Finland
- Drug Research Program, Division of Pharmacology and Pharmacotherapy, University of Helsinki, Helsinki, Finland
| |
Collapse
|
16
|
Zhang Y, Huang Z, Li H. Insights into innate immune signalling in controlling cardiac remodelling. Cardiovasc Res 2017; 113:1538-1550. [PMID: 29088374 DOI: 10.1093/cvr/cvx130] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Accepted: 06/29/2017] [Indexed: 01/03/2025] Open
Abstract
Canonical innate immune signalling involves complex cascades: multiple germline-encoded pattern recognition receptors rapidly recognize pathogen-associated or damage-associated molecular patterns to induce the production of cytokines, which bind to their corresponding receptors to orchestrate subsequent host defense phases. Inflammation is a healthy response to pathogenic signals, which are typically rapid and specific, and they terminate once the threat has passed. However, excessive activation or suppression of innate immune or inflammatory responses can lead to considerable human suffering, such as cardiac remodelling. Interestingly, recent studies have revealed that innate immune molecules in the parenchymal cells of the heart influence cardiac homeostasis not only by directly regulating innate immune responses but also through reprogrammed signalling pathways, which are independent of conventional innate immune signalling. Elucidating 'innate immune signalling reprogramming' events will help us better understand the functions of innate immune molecules and, moreover, the pathogenesis of cardiac diseases.
Collapse
Affiliation(s)
- Yaxing Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University, Jiefang Road 238, Wuchang District, Wuhan 430060, People's Republic of China
- Institute of Model Animal of Wuhan University, Donghu Road 115, Wuchang District, Wuhan 430071, People's Republic of China
- Medical Research Institute, School of Medicine, Wuhan University, Donghu Road 115, Wuchang District, Wuhan 430071, People's Republic of China
| | - Zan Huang
- Department of Cardiology, Renmin Hospital of Wuhan University, Jiefang Road 238, Wuchang District, Wuhan 430060, People's Republic of China
- Institute of Model Animal of Wuhan University, Donghu Road 115, Wuchang District, Wuhan 430071, People's Republic of China
- Medical Research Institute, School of Medicine, Wuhan University, Donghu Road 115, Wuchang District, Wuhan 430071, People's Republic of China
- College of Life Sciences, Wuhan University, Wuhan 430072, People's Republic of China
| | - Hongliang Li
- Department of Cardiology, Renmin Hospital of Wuhan University, Jiefang Road 238, Wuchang District, Wuhan 430060, People's Republic of China
- Institute of Model Animal of Wuhan University, Donghu Road 115, Wuchang District, Wuhan 430071, People's Republic of China
- Medical Research Institute, School of Medicine, Wuhan University, Donghu Road 115, Wuchang District, Wuhan 430071, People's Republic of China
| |
Collapse
|
17
|
Chami B, Jeong G, Varda A, Maw AM, Kim HB, Fong G, Simone M, Rayner B, Wang XS, Dennis J, Witting P. The nitroxide 4-methoxy TEMPO inhibits neutrophil-stimulated kinase activation in H9c2 cardiomyocytes. Arch Biochem Biophys 2017; 629:19-35. [DOI: 10.1016/j.abb.2017.07.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Revised: 06/23/2017] [Accepted: 07/03/2017] [Indexed: 12/12/2022]
|
18
|
Zhang Y, Li H. Reprogramming Interferon Regulatory Factor Signaling in Cardiometabolic Diseases. Physiology (Bethesda) 2017; 32:210-223. [PMID: 28404737 DOI: 10.1152/physiol.00038.2016] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2016] [Revised: 02/06/2017] [Accepted: 02/09/2017] [Indexed: 01/12/2023] Open
Abstract
Interferon regulatory factors (IRFs) are evolutionarily conserved proteins expressed not only in immune cells but also in other tissues and organs outside the immune system. In this review, we discuss mechanisms responsible for IRF-mediated innate immune responses and the function and mechanism of IRFs in cardiometabolic diseases. We focus on the role of IRFs in innate immunity and cardiometabolic homeostasis, and highlight reprogrammed IRF signaling.
Collapse
Affiliation(s)
- Yaxing Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, People's Republic of China
- Institute of Model Animal, Wuhan University, Wuhan, People's Republic of China; and
- Medical Research Institute, School of Medicine, Wuhan University, Wuhan, People's Republic of China
| | - Hongliang Li
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, People's Republic of China
- Institute of Model Animal, Wuhan University, Wuhan, People's Republic of China; and
- Medical Research Institute, School of Medicine, Wuhan University, Wuhan, People's Republic of China
| |
Collapse
|
19
|
Zhang Y, Zhang L, Lin XH, Li ZM, Zhang QY. Knockdown of IRF3 inhibits extracellular matrix expression in keloid fibroblasts. Biomed Pharmacother 2017; 88:1064-1068. [DOI: 10.1016/j.biopha.2017.01.142] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Revised: 01/23/2017] [Accepted: 01/25/2017] [Indexed: 12/31/2022] Open
|
20
|
Zhang Y, Zhang XJ, Wang PX, Zhang P, Li H. Reprogramming Innate Immune Signaling in Cardiometabolic Disease. Hypertension 2017; 69:747-760. [PMID: 28320852 DOI: 10.1161/hypertensionaha.116.08192] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Affiliation(s)
- Yaxing Zhang
- From the Department of Cardiology, Renmin Hospital (Y.Z., X.-J.Z., P.-X.W., P.Z., H.L.), School of Basic Medical Sciences (Y.Z., X.-J.Z., P.-X.W., P.Z., H.L.), Institute of Model Animal (Y.Z., X.-J.Z., P.-X.W., P.Z., H.L.), and Medical Research Institute, School of Medicine (Y.Z., X.-J.Z., P.-X.W., P.Z., H.L.), Wuhan University, P.R. China
| | - Xiao-Jing Zhang
- From the Department of Cardiology, Renmin Hospital (Y.Z., X.-J.Z., P.-X.W., P.Z., H.L.), School of Basic Medical Sciences (Y.Z., X.-J.Z., P.-X.W., P.Z., H.L.), Institute of Model Animal (Y.Z., X.-J.Z., P.-X.W., P.Z., H.L.), and Medical Research Institute, School of Medicine (Y.Z., X.-J.Z., P.-X.W., P.Z., H.L.), Wuhan University, P.R. China
| | - Pi-Xiao Wang
- From the Department of Cardiology, Renmin Hospital (Y.Z., X.-J.Z., P.-X.W., P.Z., H.L.), School of Basic Medical Sciences (Y.Z., X.-J.Z., P.-X.W., P.Z., H.L.), Institute of Model Animal (Y.Z., X.-J.Z., P.-X.W., P.Z., H.L.), and Medical Research Institute, School of Medicine (Y.Z., X.-J.Z., P.-X.W., P.Z., H.L.), Wuhan University, P.R. China
| | - Peng Zhang
- From the Department of Cardiology, Renmin Hospital (Y.Z., X.-J.Z., P.-X.W., P.Z., H.L.), School of Basic Medical Sciences (Y.Z., X.-J.Z., P.-X.W., P.Z., H.L.), Institute of Model Animal (Y.Z., X.-J.Z., P.-X.W., P.Z., H.L.), and Medical Research Institute, School of Medicine (Y.Z., X.-J.Z., P.-X.W., P.Z., H.L.), Wuhan University, P.R. China
| | - Hongliang Li
- From the Department of Cardiology, Renmin Hospital (Y.Z., X.-J.Z., P.-X.W., P.Z., H.L.), School of Basic Medical Sciences (Y.Z., X.-J.Z., P.-X.W., P.Z., H.L.), Institute of Model Animal (Y.Z., X.-J.Z., P.-X.W., P.Z., H.L.), and Medical Research Institute, School of Medicine (Y.Z., X.-J.Z., P.-X.W., P.Z., H.L.), Wuhan University, P.R. China.
| |
Collapse
|
21
|
Siedlinski M, Nosalski R, Szczepaniak P, Ludwig-Gałęzowska AH, Mikołajczyk T, Filip M, Osmenda G, Wilk G, Nowak M, Wołkow P, Guzik TJ. Vascular transcriptome profiling identifies Sphingosine kinase 1 as a modulator of angiotensin II-induced vascular dysfunction. Sci Rep 2017; 7:44131. [PMID: 28276483 PMCID: PMC5343497 DOI: 10.1038/srep44131] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Accepted: 02/03/2017] [Indexed: 12/22/2022] Open
Abstract
Vascular dysfunction is an important phenomenon in hypertension. We hypothesized that angiotensin II (AngII) affects transcriptome in the vasculature in a region-specific manner, which may help to identify genes related to vascular dysfunction in AngII-induced hypertension. Mesenteric artery and aortic transcriptome was profiled using Illumina WG-6v2.0 chip in control and AngII infused (490 ng/kg/min) hypertensive mice. Gene set enrichment and leading edge analyses identified Sphingosine kinase 1 (Sphk1) in the highest number of pathways affected by AngII. Sphk1 mRNA, protein and activity were up-regulated in the hypertensive vasculature. Chronic sphingosine-1-phosphate (S1P) infusion resulted in a development of significantly increased vasoconstriction and endothelial dysfunction. AngII-induced hypertension was blunted in Sphk1-/- mice (systolic BP 167 ± 4.2 vs. 180 ± 3.3 mmHg, p < 0.05), which was associated with decreased aortic and mesenteric vasoconstriction in hypertensive Sphk1-/- mice. Pharmacological inhibition of S1P synthesis reduced vasoconstriction of mesenteric arteries. While Sphk1 is important in mediating vasoconstriction in hypertension, Sphk1-/- mice were characterized by enhanced endothelial dysfunction, suggesting a local protective role of Sphk1 in the endothelium. S1P serum level in humans was correlated with endothelial function (arterial tonometry). Thus, vascular transcriptome analysis shows that S1P pathway is critical in the regulation of vascular function in AngII-induced hypertension, although Sphk1 may have opposing roles in the regulation of vasoconstriction and endothelium-dependent vasorelaxation.
Collapse
Affiliation(s)
- Mateusz Siedlinski
- Department of Internal and Agricultural Medicine, Faculty of Medicine, Jagiellonian University Medical College, Kraków, Poland
| | - Ryszard Nosalski
- Department of Internal and Agricultural Medicine, Faculty of Medicine, Jagiellonian University Medical College, Kraków, Poland.,British Heart Foundation Centre for Excellence, Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, Scotland, UK
| | - Piotr Szczepaniak
- Department of Internal and Agricultural Medicine, Faculty of Medicine, Jagiellonian University Medical College, Kraków, Poland
| | | | - Tomasz Mikołajczyk
- Department of Internal and Agricultural Medicine, Faculty of Medicine, Jagiellonian University Medical College, Kraków, Poland.,British Heart Foundation Centre for Excellence, Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, Scotland, UK
| | - Magdalena Filip
- Department of Internal and Agricultural Medicine, Faculty of Medicine, Jagiellonian University Medical College, Kraków, Poland
| | - Grzegorz Osmenda
- Department of Internal and Agricultural Medicine, Faculty of Medicine, Jagiellonian University Medical College, Kraków, Poland
| | - Grzegorz Wilk
- Department of Internal and Agricultural Medicine, Faculty of Medicine, Jagiellonian University Medical College, Kraków, Poland
| | - Michał Nowak
- Department of Internal and Agricultural Medicine, Faculty of Medicine, Jagiellonian University Medical College, Kraków, Poland
| | - Paweł Wołkow
- Centre for Medical Genomics-OMICRON, Jagiellonian University Medical College, Kraków, Poland
| | - Tomasz J Guzik
- Department of Internal and Agricultural Medicine, Faculty of Medicine, Jagiellonian University Medical College, Kraków, Poland.,British Heart Foundation Centre for Excellence, Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, Scotland, UK
| |
Collapse
|
22
|
Liu H, Cheng WL, Jiang X, Wang PX, Fang C, Zhu XY, Huang Z, She ZG, Li H. Ablation of Interferon Regulatory Factor 3 Protects Against Atherosclerosis in Apolipoprotein E-Deficient Mice. Hypertension 2017; 69:510-520. [PMID: 28115514 DOI: 10.1161/hypertensionaha.116.08395] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2016] [Revised: 09/12/2016] [Accepted: 12/08/2016] [Indexed: 01/13/2023]
Abstract
The secretion of adhesion molecules by endothelial cells, as well as the subsequent infiltration of macrophages, determines the initiation and progression of atherosclerosis. Accumulating evidence suggests that IRF3 (interferon regulatory factor 3) is required for the induction of proinflammatory cytokines and for endothelial cell proliferation. However, the effect and underlying mechanism of IRF3 on atherogenesis remain unknown. Our results demonstrated a moderate-to-strong immunoreactivity effect associated with IRF3 in the endothelium and macrophages of the atherosclerotic plaques in patients with coronary heart disease and in hyperlipidemic mice. IRF3-/-ApoE-/- mice showed significantly decreased atherosclerotic lesions in the whole aorta, aortic sinus, and brachiocephalic arteries. The bone marrow transplantation further suggested that the amelioration of atherosclerosis might be attributed to the effects of IRF3 deficiency mainly in endothelial cells, as well as in macrophages. The enhanced stability of atherosclerotic plaques in IRF3-/-ApoE-/- mice was characterized by the reduction of necrotic core size, macrophage infiltration, and lipids, which was accompanied by increased collagen and smooth muscle cell content. Furthermore, multiple proinflammatory cytokines showed a marked decrease in IRF3-/-ApoE-/- mice. Mechanistically, IRF3 deficiency suppresses the secretion of VCAM-1 (vascular cell adhesion molecule 1) and the expression of ICAM-1 (intercellular adhesion molecule 1) by directly binding to the ICAM-1 promoter, which subsequently attenuates macrophage infiltration. Thus, our study suggests that IRF3 might be a potential target for the treatment of atherosclerosis development.
Collapse
Affiliation(s)
- Hui Liu
- From the Department of Cardiology, Renmin Hospital of Wuhan University, China (H. Liu, W.-L.C., X.J., P.-X.W., C.F., X.-Y.Z., Z.-G.S., H. Li); and The Institute of Model Animals (H. Liu, W.-L.C., X.J., P.-X.W., C.F., X.-Y.Z., Z.-G.S., H. Li), Medical Research Institute, School of Medicine (H. Liu, W.-L.C., X.J., P.-X.W., C.F., X.-Y.Z., Z.-G.S., H. Li), Collaborative Innovation Center of Model Animal (H. Liu, W.-L.C., X.J., P.-X.W., C.F., X.-Y.Z., Z.-G.S., H. Li), Cardiovascular Research Institute (H. Liu, W.-L.C., X.J., P.-X.W., C.F., X.-Y.Z., Z.-G.S., H. Li), and College of Life Science (Z.H.), Wuhan University, China
| | - Wen-Lin Cheng
- From the Department of Cardiology, Renmin Hospital of Wuhan University, China (H. Liu, W.-L.C., X.J., P.-X.W., C.F., X.-Y.Z., Z.-G.S., H. Li); and The Institute of Model Animals (H. Liu, W.-L.C., X.J., P.-X.W., C.F., X.-Y.Z., Z.-G.S., H. Li), Medical Research Institute, School of Medicine (H. Liu, W.-L.C., X.J., P.-X.W., C.F., X.-Y.Z., Z.-G.S., H. Li), Collaborative Innovation Center of Model Animal (H. Liu, W.-L.C., X.J., P.-X.W., C.F., X.-Y.Z., Z.-G.S., H. Li), Cardiovascular Research Institute (H. Liu, W.-L.C., X.J., P.-X.W., C.F., X.-Y.Z., Z.-G.S., H. Li), and College of Life Science (Z.H.), Wuhan University, China
| | - Xi Jiang
- From the Department of Cardiology, Renmin Hospital of Wuhan University, China (H. Liu, W.-L.C., X.J., P.-X.W., C.F., X.-Y.Z., Z.-G.S., H. Li); and The Institute of Model Animals (H. Liu, W.-L.C., X.J., P.-X.W., C.F., X.-Y.Z., Z.-G.S., H. Li), Medical Research Institute, School of Medicine (H. Liu, W.-L.C., X.J., P.-X.W., C.F., X.-Y.Z., Z.-G.S., H. Li), Collaborative Innovation Center of Model Animal (H. Liu, W.-L.C., X.J., P.-X.W., C.F., X.-Y.Z., Z.-G.S., H. Li), Cardiovascular Research Institute (H. Liu, W.-L.C., X.J., P.-X.W., C.F., X.-Y.Z., Z.-G.S., H. Li), and College of Life Science (Z.H.), Wuhan University, China
| | - Pi-Xiao Wang
- From the Department of Cardiology, Renmin Hospital of Wuhan University, China (H. Liu, W.-L.C., X.J., P.-X.W., C.F., X.-Y.Z., Z.-G.S., H. Li); and The Institute of Model Animals (H. Liu, W.-L.C., X.J., P.-X.W., C.F., X.-Y.Z., Z.-G.S., H. Li), Medical Research Institute, School of Medicine (H. Liu, W.-L.C., X.J., P.-X.W., C.F., X.-Y.Z., Z.-G.S., H. Li), Collaborative Innovation Center of Model Animal (H. Liu, W.-L.C., X.J., P.-X.W., C.F., X.-Y.Z., Z.-G.S., H. Li), Cardiovascular Research Institute (H. Liu, W.-L.C., X.J., P.-X.W., C.F., X.-Y.Z., Z.-G.S., H. Li), and College of Life Science (Z.H.), Wuhan University, China
| | - Chun Fang
- From the Department of Cardiology, Renmin Hospital of Wuhan University, China (H. Liu, W.-L.C., X.J., P.-X.W., C.F., X.-Y.Z., Z.-G.S., H. Li); and The Institute of Model Animals (H. Liu, W.-L.C., X.J., P.-X.W., C.F., X.-Y.Z., Z.-G.S., H. Li), Medical Research Institute, School of Medicine (H. Liu, W.-L.C., X.J., P.-X.W., C.F., X.-Y.Z., Z.-G.S., H. Li), Collaborative Innovation Center of Model Animal (H. Liu, W.-L.C., X.J., P.-X.W., C.F., X.-Y.Z., Z.-G.S., H. Li), Cardiovascular Research Institute (H. Liu, W.-L.C., X.J., P.-X.W., C.F., X.-Y.Z., Z.-G.S., H. Li), and College of Life Science (Z.H.), Wuhan University, China
| | - Xue-Yong Zhu
- From the Department of Cardiology, Renmin Hospital of Wuhan University, China (H. Liu, W.-L.C., X.J., P.-X.W., C.F., X.-Y.Z., Z.-G.S., H. Li); and The Institute of Model Animals (H. Liu, W.-L.C., X.J., P.-X.W., C.F., X.-Y.Z., Z.-G.S., H. Li), Medical Research Institute, School of Medicine (H. Liu, W.-L.C., X.J., P.-X.W., C.F., X.-Y.Z., Z.-G.S., H. Li), Collaborative Innovation Center of Model Animal (H. Liu, W.-L.C., X.J., P.-X.W., C.F., X.-Y.Z., Z.-G.S., H. Li), Cardiovascular Research Institute (H. Liu, W.-L.C., X.J., P.-X.W., C.F., X.-Y.Z., Z.-G.S., H. Li), and College of Life Science (Z.H.), Wuhan University, China
| | - Zan Huang
- From the Department of Cardiology, Renmin Hospital of Wuhan University, China (H. Liu, W.-L.C., X.J., P.-X.W., C.F., X.-Y.Z., Z.-G.S., H. Li); and The Institute of Model Animals (H. Liu, W.-L.C., X.J., P.-X.W., C.F., X.-Y.Z., Z.-G.S., H. Li), Medical Research Institute, School of Medicine (H. Liu, W.-L.C., X.J., P.-X.W., C.F., X.-Y.Z., Z.-G.S., H. Li), Collaborative Innovation Center of Model Animal (H. Liu, W.-L.C., X.J., P.-X.W., C.F., X.-Y.Z., Z.-G.S., H. Li), Cardiovascular Research Institute (H. Liu, W.-L.C., X.J., P.-X.W., C.F., X.-Y.Z., Z.-G.S., H. Li), and College of Life Science (Z.H.), Wuhan University, China
| | - Zhi-Gang She
- From the Department of Cardiology, Renmin Hospital of Wuhan University, China (H. Liu, W.-L.C., X.J., P.-X.W., C.F., X.-Y.Z., Z.-G.S., H. Li); and The Institute of Model Animals (H. Liu, W.-L.C., X.J., P.-X.W., C.F., X.-Y.Z., Z.-G.S., H. Li), Medical Research Institute, School of Medicine (H. Liu, W.-L.C., X.J., P.-X.W., C.F., X.-Y.Z., Z.-G.S., H. Li), Collaborative Innovation Center of Model Animal (H. Liu, W.-L.C., X.J., P.-X.W., C.F., X.-Y.Z., Z.-G.S., H. Li), Cardiovascular Research Institute (H. Liu, W.-L.C., X.J., P.-X.W., C.F., X.-Y.Z., Z.-G.S., H. Li), and College of Life Science (Z.H.), Wuhan University, China
| | - Hongliang Li
- From the Department of Cardiology, Renmin Hospital of Wuhan University, China (H. Liu, W.-L.C., X.J., P.-X.W., C.F., X.-Y.Z., Z.-G.S., H. Li); and The Institute of Model Animals (H. Liu, W.-L.C., X.J., P.-X.W., C.F., X.-Y.Z., Z.-G.S., H. Li), Medical Research Institute, School of Medicine (H. Liu, W.-L.C., X.J., P.-X.W., C.F., X.-Y.Z., Z.-G.S., H. Li), Collaborative Innovation Center of Model Animal (H. Liu, W.-L.C., X.J., P.-X.W., C.F., X.-Y.Z., Z.-G.S., H. Li), Cardiovascular Research Institute (H. Liu, W.-L.C., X.J., P.-X.W., C.F., X.-Y.Z., Z.-G.S., H. Li), and College of Life Science (Z.H.), Wuhan University, China.
| |
Collapse
|
23
|
Jeidane S, Scott-Boyer MP, Tremblay N, Cardin S, Picard S, Baril M, Lamarre D, Deschepper CF. Association of a Network of Interferon-Stimulated Genes with a Locus Encoding a Negative Regulator of Non-conventional IKK Kinases and IFNB1. Cell Rep 2016; 17:425-435. [PMID: 27705791 DOI: 10.1016/j.celrep.2016.09.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2015] [Revised: 08/11/2016] [Accepted: 09/02/2016] [Indexed: 11/25/2022] Open
Abstract
Functional genomic analysis of gene expression in mice allowed us to identify a quantitative trait locus (QTL) linked in trans to the expression of 190 gene transcripts and in cis to the expression of only two genes, one of which was Ypel5. Most of the trans-expression QTL genes were interferon-stimulated genes (ISGs), and their expression in mouse macrophage cell lines was stimulated in an IFNB1-dependent manner by Ypel5 silencing. In human HEK293T cells, YPEL5 silencing enhanced the induction of IFNB1 by pattern recognition receptors and phosphorylation of TBK1/IKBKE kinases, whereas co-immunoprecipitation experiments revealed that YPEL5 interacted physically with IKBKE. We thus found that the Ypel5 gene (contained in a locus linked to a network of ISGs in mice) is a negative regulator of IFNB1 production and innate immune responses that interacts functionally and physically with TBK1/IKBKE kinases.
Collapse
Affiliation(s)
- Saloua Jeidane
- Cardiovascular Biology Research Unit, Institut de Recherches Cliniques de Montréal (IRCM), Montréal, QC H2W 1R7, Canada; Département de Médecine, Faculté de Médecine, Université de Montréal, Montréal, QC J2S 2M2, Canada
| | - Marie-Pier Scott-Boyer
- Cardiovascular Biology Research Unit, Institut de Recherches Cliniques de Montréal (IRCM), Montréal, QC H2W 1R7, Canada; Département de Médecine, Faculté de Médecine, Université de Montréal, Montréal, QC J2S 2M2, Canada
| | - Nicolas Tremblay
- Département de Médecine, Faculté de Médecine, Université de Montréal, Montréal, QC J2S 2M2, Canada; Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CR-CHUM), Montréal, QC H2X 3J4, Canada
| | - Sophie Cardin
- Cardiovascular Biology Research Unit, Institut de Recherches Cliniques de Montréal (IRCM), Montréal, QC H2W 1R7, Canada; Département de Médecine, Faculté de Médecine, Université de Montréal, Montréal, QC J2S 2M2, Canada
| | - Sylvie Picard
- Cardiovascular Biology Research Unit, Institut de Recherches Cliniques de Montréal (IRCM), Montréal, QC H2W 1R7, Canada
| | - Martin Baril
- Département de Médecine, Faculté de Médecine, Université de Montréal, Montréal, QC J2S 2M2, Canada; Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CR-CHUM), Montréal, QC H2X 3J4, Canada
| | - Daniel Lamarre
- Département de Médecine, Faculté de Médecine, Université de Montréal, Montréal, QC J2S 2M2, Canada; Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CR-CHUM), Montréal, QC H2X 3J4, Canada
| | - Christian F Deschepper
- Cardiovascular Biology Research Unit, Institut de Recherches Cliniques de Montréal (IRCM), Montréal, QC H2W 1R7, Canada; Département de Médecine, Faculté de Médecine, Université de Montréal, Montréal, QC J2S 2M2, Canada.
| |
Collapse
|
24
|
Ezzat SM, El Gaafary M, El Sayed AM, Sabry OM, Ali ZY, Hafner S, Schmiech M, Jin L, Syrovets T, Simmet T. The Cardenolide Glycoside Acovenoside A Affords Protective Activity in Doxorubicin-Induced Cardiotoxicity in Mice. J Pharmacol Exp Ther 2016; 358:262-70. [PMID: 27247000 DOI: 10.1124/jpet.116.232652] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2011] [Accepted: 11/15/2011] [Indexed: 03/08/2025] Open
Abstract
The current study aimed to investigate the protective effect of the cardenolide glycoside acovenoside A (AcoA) against doxorubicin-induced cardiotoxicity in mice. AcoA was isolated from the pericarps of Acokanthera oppositifolia to chemical homogeneity and characterized by means of one- and two-dimensional nuclear magnetic resonance spectroscopy. AcoA exhibited relatively low toxicity in mice (LD50 = 223.3 mg/kg bw). Repeated administration of doxorubicin induced cardiotoxicity manifested by reduced activity of myocardial membrane-bound ion pumps and elevated serum biomarkers of myocardial dysfunction, oxidative stress, and inflammation. Pretreatment of doxorubicin-exposed mice with AcoA (11.16 or 22.33 mg/kg bw, i.p.) for 2 weeks after 2 weeks of combined administration of AcoA and doxorubicin protected the animals dose dependently against doxorubicin-induced cardiotoxicity as indicated by normalization of the levels of different myocardial markers of oxidative stress (malondialdehyde, nitric oxide, total antioxidant capacity, and cardiac glutathione), serum myocardial diagnostic marker enzymes (serum cardiac troponin T, creatine kinase isoenzyme MB, aspartate aminotransferase, and lactate dehydrogenase), and inflammatory markers (c-reactive protein, tumor necrosis factor-α, and interleukin-6), as well as myocardial Na(+)/K(+)-ATPase activity. These effects were attributed to the negative impact of AcoA on transcription factors nuclear factor κB and interferon regulatory factor 3/7. Thus acovenoside A might act as a cardioprotective agent to prevent doxorubicin-induced cardiotoxicity.
Collapse
Affiliation(s)
- Shahira M Ezzat
- Department of Pharmacognosy, College of Pharmacy, Cairo University, Giza, Egypt (S.M.E., M.E.G., A.M.E.S., O.M.S.); Department of Biochemistry, National Organization for Drug Control and Research (NODCAR), Cairo, Egypt (Z.Y.A.); Institute of Pharmacology of Natural Products & Clinical Pharmacology, Ulm University, D-89081 Ulm, Germany (M.E.G., S.H., M.S., L.J., Ta.S., Th.S.)
| | - Menna El Gaafary
- Department of Pharmacognosy, College of Pharmacy, Cairo University, Giza, Egypt (S.M.E., M.E.G., A.M.E.S., O.M.S.); Department of Biochemistry, National Organization for Drug Control and Research (NODCAR), Cairo, Egypt (Z.Y.A.); Institute of Pharmacology of Natural Products & Clinical Pharmacology, Ulm University, D-89081 Ulm, Germany (M.E.G., S.H., M.S., L.J., Ta.S., Th.S.)
| | - Abeer M El Sayed
- Department of Pharmacognosy, College of Pharmacy, Cairo University, Giza, Egypt (S.M.E., M.E.G., A.M.E.S., O.M.S.); Department of Biochemistry, National Organization for Drug Control and Research (NODCAR), Cairo, Egypt (Z.Y.A.); Institute of Pharmacology of Natural Products & Clinical Pharmacology, Ulm University, D-89081 Ulm, Germany (M.E.G., S.H., M.S., L.J., Ta.S., Th.S.)
| | - Omar M Sabry
- Department of Pharmacognosy, College of Pharmacy, Cairo University, Giza, Egypt (S.M.E., M.E.G., A.M.E.S., O.M.S.); Department of Biochemistry, National Organization for Drug Control and Research (NODCAR), Cairo, Egypt (Z.Y.A.); Institute of Pharmacology of Natural Products & Clinical Pharmacology, Ulm University, D-89081 Ulm, Germany (M.E.G., S.H., M.S., L.J., Ta.S., Th.S.)
| | - Zeinab Y Ali
- Department of Pharmacognosy, College of Pharmacy, Cairo University, Giza, Egypt (S.M.E., M.E.G., A.M.E.S., O.M.S.); Department of Biochemistry, National Organization for Drug Control and Research (NODCAR), Cairo, Egypt (Z.Y.A.); Institute of Pharmacology of Natural Products & Clinical Pharmacology, Ulm University, D-89081 Ulm, Germany (M.E.G., S.H., M.S., L.J., Ta.S., Th.S.)
| | - Susanne Hafner
- Department of Pharmacognosy, College of Pharmacy, Cairo University, Giza, Egypt (S.M.E., M.E.G., A.M.E.S., O.M.S.); Department of Biochemistry, National Organization for Drug Control and Research (NODCAR), Cairo, Egypt (Z.Y.A.); Institute of Pharmacology of Natural Products & Clinical Pharmacology, Ulm University, D-89081 Ulm, Germany (M.E.G., S.H., M.S., L.J., Ta.S., Th.S.)
| | - Michael Schmiech
- Department of Pharmacognosy, College of Pharmacy, Cairo University, Giza, Egypt (S.M.E., M.E.G., A.M.E.S., O.M.S.); Department of Biochemistry, National Organization for Drug Control and Research (NODCAR), Cairo, Egypt (Z.Y.A.); Institute of Pharmacology of Natural Products & Clinical Pharmacology, Ulm University, D-89081 Ulm, Germany (M.E.G., S.H., M.S., L.J., Ta.S., Th.S.)
| | - Lu Jin
- Department of Pharmacognosy, College of Pharmacy, Cairo University, Giza, Egypt (S.M.E., M.E.G., A.M.E.S., O.M.S.); Department of Biochemistry, National Organization for Drug Control and Research (NODCAR), Cairo, Egypt (Z.Y.A.); Institute of Pharmacology of Natural Products & Clinical Pharmacology, Ulm University, D-89081 Ulm, Germany (M.E.G., S.H., M.S., L.J., Ta.S., Th.S.)
| | - Tatiana Syrovets
- Department of Pharmacognosy, College of Pharmacy, Cairo University, Giza, Egypt (S.M.E., M.E.G., A.M.E.S., O.M.S.); Department of Biochemistry, National Organization for Drug Control and Research (NODCAR), Cairo, Egypt (Z.Y.A.); Institute of Pharmacology of Natural Products & Clinical Pharmacology, Ulm University, D-89081 Ulm, Germany (M.E.G., S.H., M.S., L.J., Ta.S., Th.S.)
| | - Thomas Simmet
- Department of Pharmacognosy, College of Pharmacy, Cairo University, Giza, Egypt (S.M.E., M.E.G., A.M.E.S., O.M.S.); Department of Biochemistry, National Organization for Drug Control and Research (NODCAR), Cairo, Egypt (Z.Y.A.); Institute of Pharmacology of Natural Products & Clinical Pharmacology, Ulm University, D-89081 Ulm, Germany (M.E.G., S.H., M.S., L.J., Ta.S., Th.S.)
| |
Collapse
|
25
|
Gomez IG, Roach AM, Nakagawa N, Amatucci A, Johnson BG, Dunn K, Kelly MC, Karaca G, Zheng TS, Szak S, Peppiatt-Wildman CM, Burkly LC, Duffield JS. TWEAK-Fn14 Signaling Activates Myofibroblasts to Drive Progression of Fibrotic Kidney Disease. J Am Soc Nephrol 2016; 27:3639-3652. [PMID: 27026366 DOI: 10.1681/asn.2015111227] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2015] [Accepted: 02/16/2016] [Indexed: 01/15/2023] Open
Abstract
The identification of the cellular origins of myofibroblasts has led to the discovery of novel pathways that potentially drive myofibroblast perpetuation in disease. Here, we further investigated the role of innate immune signaling pathways in this process. In mice, renal injury-induced activation of pericytes, which are myofibroblast precursors attached to endothelial cells, led to upregulated expression of TNF receptor superfamily member 12a, also known as fibroblast growth factor-inducible 14 (Fn14), by these cells. In live rat kidney slices, administration of the Fn14 ligand, TNF-related weak inducer of apoptosis (TWEAK), promoted pericyte-dependent vasoconstriction followed by pericyte detachment from capillaries. In vitro, administration of TWEAK activated and differentiated pericytes into cytokine-producing myofibroblasts, and further activated established myofibroblasts in a manner requiring canonical and noncanonical NF-κB signaling pathways. Deficiency of Fn14 protected mouse kidneys from fibrogenesis, inflammation, and associated vascular instability after in vivo injury, and was associated with loss of NF-κB signaling. In a genetic model of spontaneous CKD, therapeutic delivery of anti-TWEAK blocking antibodies attenuated disease progression, preserved organ function, and increased survival. These results identify the TWEAK-Fn14 signaling pathway as an important factor in myofibroblast perpetuation, fibrogenesis, and chronic disease progression.
Collapse
Affiliation(s)
- Ivan G Gomez
- Research & Development, Biogen, Cambridge, Massachusetts.,Division of Nephrology, Departments of Medicine & Pathology, and Institute of Stem Cell & Regenerative Medicine, University of Washington, Seattle, Washington; and
| | - Allie M Roach
- Research & Development, Biogen, Cambridge, Massachusetts.,Division of Nephrology, Departments of Medicine & Pathology, and Institute of Stem Cell & Regenerative Medicine, University of Washington, Seattle, Washington; and
| | - Naoki Nakagawa
- Division of Nephrology, Departments of Medicine & Pathology, and Institute of Stem Cell & Regenerative Medicine, University of Washington, Seattle, Washington; and
| | - Aldo Amatucci
- Research & Development, Biogen, Cambridge, Massachusetts
| | - Bryce G Johnson
- Research & Development, Biogen, Cambridge, Massachusetts.,Division of Nephrology, Departments of Medicine & Pathology, and Institute of Stem Cell & Regenerative Medicine, University of Washington, Seattle, Washington; and
| | - Kadeshia Dunn
- Medway School of Pharmacy, University of Kent, Chatham, Kent, United Kingdom
| | - Mark C Kelly
- Medway School of Pharmacy, University of Kent, Chatham, Kent, United Kingdom
| | - Gamze Karaca
- Research & Development, Biogen, Cambridge, Massachusetts
| | | | - Suzanne Szak
- Research & Development, Biogen, Cambridge, Massachusetts
| | | | - Linda C Burkly
- Research & Development, Biogen, Cambridge, Massachusetts;
| | - Jeremy S Duffield
- Research & Development, Biogen, Cambridge, Massachusetts; .,Division of Nephrology, Departments of Medicine & Pathology, and Institute of Stem Cell & Regenerative Medicine, University of Washington, Seattle, Washington; and
| |
Collapse
|
26
|
Intragraft transcriptional profiling of renal transplant patients with tubular dysfunction reveals mechanisms underlying graft injury and recovery. Hum Genomics 2016; 10:2. [PMID: 26742487 PMCID: PMC4705764 DOI: 10.1186/s40246-015-0059-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2015] [Accepted: 12/30/2015] [Indexed: 12/13/2022] Open
Abstract
Background Proximal tubular dysfunction (PTD) is associated with a decreased long-term graft survival in renal transplant patients and can be detected by the elevation of urinary tubular proteins. This study investigated transcriptional changes in biopsies from renal transplant patients with PTD to disclose molecular mechanisms underlying graft injury and functional recovery. Methods Thirty-three renal transplant patients with high urinary levels of retinol-binding protein, a biomarker of PTD, were enrolled in the study. The initial immunosuppressive scheme included azathioprine, cyclosporine, and steroids. After randomization, 18 patients (group 2) had their treatment modified by reducing cyclosporine dosage and substituting azathioprine for mycophenolate mofetil, while the other 15 patients (group 1) remained under the initial scheme. Patients were biopsied at enrollment and after 12 months of follow-up, and paired comparisons were performed between their intragraft gene expression profiles. The differential transcriptome profiles were analyzed by constructing gene co-expression networks and identifying enriched functions and central nodes in each network. Results Only the alternative immunosuppressive scheme used in group 2 ameliorated renal function and tubular proteinuria after 12 months of follow-up. Intragraft molecular changes observed in group 2 were linked to autophagy, extracellular matrix, and adaptive immunity. Conversely, gene expression changes in group 1 were related to fibrosis, endocytosis, ubiquitination, and endoplasmic reticulum stress. Conclusion These results suggest that molecular networks associated with the control of endocytosis, autophagy, protein overload, fibrosis, and adaptive immunity may be involved in improvement of graft function. Electronic supplementary material The online version of this article (doi:10.1186/s40246-015-0059-6) contains supplementary material, which is available to authorized users.
Collapse
|
27
|
Zhang X, Jiang D, Li H. The interferon regulatory factors as novel potential targets in the treatment of cardiovascular diseases. Br J Pharmacol 2015; 172:5457-5476. [PMID: 25131895 PMCID: PMC4667854 DOI: 10.1111/bph.12881] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2014] [Revised: 07/26/2014] [Accepted: 08/12/2014] [Indexed: 02/06/2023] Open
Abstract
The family of interferon regulatory factors (IRFs) consists of nine members (IRF1-IRF9) in mammals. They act as transcription factors for the interferons and thus exert essential regulatory functions in the immune system and in oncogenesis. Recent clinical and experimental studies have identified critically important roles of the IRFs in cardiovascular diseases, arising from their participation in divergent and overlapping molecular programmes beyond the immune response. Here we review the current knowledge of the regulatory effects and mechanisms of IRFs on the immune system. The role of IRFs and their potential molecular mechanisms as novel stress sensors and mediators of cardiovascular diseases are highlighted.
Collapse
Affiliation(s)
- Xiao‐Jing Zhang
- Department of Cardiology, Renmin HospitalWuhan UniversityWuhanChina
- Cardiovascular Research InstituteWuhan UniversityWuhanChina
- State Key Laboratory of Quality Research in Chinese MedicineInstitute of Chinese Medical SciencesUniversity of MacauMacaoChina
| | - Ding‐Sheng Jiang
- Department of Cardiology, Renmin HospitalWuhan UniversityWuhanChina
- Cardiovascular Research InstituteWuhan UniversityWuhanChina
| | - Hongliang Li
- Department of Cardiology, Renmin HospitalWuhan UniversityWuhanChina
- Cardiovascular Research InstituteWuhan UniversityWuhanChina
| |
Collapse
|
28
|
Ni MM, Xu T, Wang YR, He YH, Zhou Q, Huang C, Meng XM, Li J. Inhibition of IRF3 expression reduces TGF-β1-induced proliferation of hepatic stellate cells. J Physiol Biochem 2015; 72:9-23. [PMID: 26611114 DOI: 10.1007/s13105-015-0452-6] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2015] [Accepted: 11/20/2015] [Indexed: 02/07/2023]
Abstract
Therapeutic management of liver fibrosis remains an unresolved clinical problem. Activation of hepatic stellate cell (HSC) is a pivotal event in the progression of liver fibrosis. Recent reports have showed that inhibition of activated HSC proliferation contributes to the reversal of liver fibrosis. Interferon regulatory factor 3 (IRF3), one member of the interferon regulatory factor (IRF) family, is recently proven to be a critical modulator in cardiac fibrosis. And accumulating evidence demonstrated that IRF3 plays a crucial role in liver diseases, such as hepatic steatosis, liver inflammation, and alcoholic liver injury. However, the understanding of the function of IRF3 in liver fibrosis remains limited. Our results identified the role of IRF3 in regulating human HSC (LX-2 cell) cell proliferation and apoptosis. The present study indicated that the expression of IRF3 was significantly increased in HSCs in response to TGF-β1 stimulation. Moreover, a stable and unlimited source of human HSC, the LX-2 cell line, transfected with IRF3-siRNA significantly decreases the expression level of type I collagen (Col1a1) and α-smooth muscle actin (α-SMA) in activated LX-2 cells. On the contrary, overexpression of IRF3 gives rise to an upregulation of Col1a1 and α-SMA in LX-2 cells, and further promoted HSC proliferation. Moreover, the inhibition of IRF3 significantly suppressed TGF-β1-induced HSC proliferation and increased its apoptosis. Of note, the present study indicated IRF3 may regulate LX-2 cell proliferation by via AKT signaling pathway. In summary, these observations suggest IRF3 may function as a novel regulator to modulate TGF-β1-induced LX-2 proliferation, at least in part, via AKT signaling pathway.
Collapse
Affiliation(s)
- Ming-ming Ni
- School of Pharmacy, Anhui Key Laboratory of Bioactivity of Natural Products, Anhui Medical University, Hefei, 230032, China.,Institute for Liver Diseases of Anhui Medical University, Anhui Medical University, Hefei, 230032, China
| | - Tao Xu
- School of Pharmacy, Anhui Key Laboratory of Bioactivity of Natural Products, Anhui Medical University, Hefei, 230032, China.,Institute for Liver Diseases of Anhui Medical University, Anhui Medical University, Hefei, 230032, China
| | - Ya-rui Wang
- School of Pharmacy, Anhui Key Laboratory of Bioactivity of Natural Products, Anhui Medical University, Hefei, 230032, China.,Institute for Liver Diseases of Anhui Medical University, Anhui Medical University, Hefei, 230032, China
| | - Ying-hua He
- School of Pharmacy, Anhui Key Laboratory of Bioactivity of Natural Products, Anhui Medical University, Hefei, 230032, China.,Institute for Liver Diseases of Anhui Medical University, Anhui Medical University, Hefei, 230032, China
| | - Qun Zhou
- School of Pharmacy, Anhui Key Laboratory of Bioactivity of Natural Products, Anhui Medical University, Hefei, 230032, China.,Institute for Liver Diseases of Anhui Medical University, Anhui Medical University, Hefei, 230032, China
| | - Cheng Huang
- School of Pharmacy, Anhui Key Laboratory of Bioactivity of Natural Products, Anhui Medical University, Hefei, 230032, China.,Institute for Liver Diseases of Anhui Medical University, Anhui Medical University, Hefei, 230032, China
| | - Xiao-ming Meng
- School of Pharmacy, Anhui Key Laboratory of Bioactivity of Natural Products, Anhui Medical University, Hefei, 230032, China.,Institute for Liver Diseases of Anhui Medical University, Anhui Medical University, Hefei, 230032, China
| | - Jun Li
- School of Pharmacy, Anhui Key Laboratory of Bioactivity of Natural Products, Anhui Medical University, Hefei, 230032, China. .,Institute for Liver Diseases of Anhui Medical University, Anhui Medical University, Hefei, 230032, China. .,School of Pharmacy, Anhui Medical University, 81 Mei Shan Road, Hefei, Anhui Province, 230032, China.
| |
Collapse
|
29
|
Zewinger S, Schumann T, Fliser D, Speer T. Innate immunity in CKD-associated vascular diseases. Nephrol Dial Transplant 2015; 31:1813-1821. [DOI: 10.1093/ndt/gfv358] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2015] [Accepted: 09/09/2015] [Indexed: 12/11/2022] Open
|
30
|
Zhang XJ, Zhang P, Li H. Interferon regulatory factor signalings in cardiometabolic diseases. Hypertension 2015; 66:222-47. [PMID: 26077571 DOI: 10.1161/hypertensionaha.115.04898] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2015] [Accepted: 05/14/2015] [Indexed: 12/24/2022]
Affiliation(s)
- Xiao-Jing Zhang
- From the Department of Cardiology, Renmin Hospital (X.-J.Z., P.Z., H.L.) and Cardiovascular Research Institute (X.-J.Z., P.Z., H.L.), Wuhan University, Wuhan, China; and State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, PR China (X.-J.Z.)
| | - Peng Zhang
- From the Department of Cardiology, Renmin Hospital (X.-J.Z., P.Z., H.L.) and Cardiovascular Research Institute (X.-J.Z., P.Z., H.L.), Wuhan University, Wuhan, China; and State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, PR China (X.-J.Z.)
| | - Hongliang Li
- From the Department of Cardiology, Renmin Hospital (X.-J.Z., P.Z., H.L.) and Cardiovascular Research Institute (X.-J.Z., P.Z., H.L.), Wuhan University, Wuhan, China; and State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, PR China (X.-J.Z.).
| |
Collapse
|
31
|
Abstract
A powerful interaction between the autonomic and the immune systems plays a prominent role in the initiation and maintenance of hypertension and significantly contributes to cardiovascular pathology, end-organ damage and mortality. Studies have shown consistent association between hypertension, proinflammatory cytokines and the cells of the innate and adaptive immune systems. The sympathetic nervous system, a major determinant of hypertension, innervates the bone marrow, spleen and peripheral lymphatic system and is proinflammatory, whereas the parasympathetic nerve activity dampens the inflammatory response through α7-nicotinic acetylcholine receptors. The neuro-immune synapse is bidirectional as cytokines may enhance the sympathetic activity through their central nervous system action that in turn increases the mobilization, migration and infiltration of immune cells in the end organs. Kidneys may be infiltrated by immune cells and mesangial cells that may originate in the bone marrow and release inflammatory cytokines that cause renal damage. Hypertension is also accompanied by infiltration of the adventitia and perivascular adipose tissue by inflammatory immune cells including macrophages. Increased cytokine production induces myogenic and structural changes in the resistance vessels, causing elevated blood pressure. Cardiac hypertrophy in hypertension may result from the mechanical afterload and the inflammatory response to resident or migratory immune cells. Toll-like receptors on innate immune cells function as sterile injury detectors and initiate the inflammatory pathway. Finally, abnormalities of innate immune cells and the molecular determinants of their activation that include toll-like receptor, adrenergic, cholinergic and AT1 receptors can define the severity of inflammation in hypertension. These receptors are putative therapeutic targets.
Collapse
|
32
|
Pioglitazone inhibits angiotensin II-induced atrial fibroblasts proliferation via NF-κB/TGF-β1/TRIF/TRAF6 pathway. Exp Cell Res 2014; 330:43-55. [PMID: 25152439 DOI: 10.1016/j.yexcr.2014.08.021] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2014] [Revised: 08/02/2014] [Accepted: 08/06/2014] [Indexed: 12/27/2022]
Abstract
The exact mechanisms underlying inhibitory effects of pioglitazone (Pio) on Angiotensin II (AngII)-induced atrial fibrosis are complex and remain largely unknown. In the present study, we examined the effect of Pio on AngII-induced mice atrial fibrosis in vivo and atrial fibroblasts proliferation in vitro. In vivo study showed that AngII infusion induced atrial fibrosis and increased expressions of Toll/IL-1 receptor domain-containing adaptor inducing IFN-β (TRIF) and tumor necrosis factor receptor associated factor 6 (TRAF6) in mice models. However, those effects could be attenuated by Pio (P<0.01). As for in vitro experiment, Pio suppressed AngII-induced atrial fibroblasts proliferation via nuclear factor-κB/transforming growth factor-β1/TRIF/TRAF6 signaling pathway in primary cultured mice atrial fibroblasts (P<0.01). In conclusion, suppression of Pio on AngII-induced atrial fibrosis might be related to its inhibitory effects on above signaling pathway.
Collapse
|
33
|
George PM, Oliver E, Dorfmuller P, Dubois OD, Reed DM, Kirkby NS, Mohamed NA, Perros F, Antigny F, Fadel E, Schreiber BE, Holmes AM, Southwood M, Hagan G, Wort SJ, Bartlett N, Morrell NW, Coghlan JG, Humbert M, Zhao L, Mitchell JA. Evidence for the involvement of type I interferon in pulmonary arterial hypertension. Circ Res 2013; 114:677-88. [PMID: 24334027 DOI: 10.1161/circresaha.114.302221] [Citation(s) in RCA: 118] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
RATIONALE Evidence is increasing of a link between interferon (IFN) and pulmonary arterial hypertension (PAH). Conditions with chronically elevated endogenous IFNs such as systemic sclerosis are strongly associated with PAH. Furthermore, therapeutic use of type I IFN is associated with PAH. This was recognized at the 2013 World Symposium on Pulmonary Hypertension where the urgent need for research into this was highlighted. OBJECTIVE To explore the role of type I IFN in PAH. METHODS AND RESULTS Cells were cultured using standard approaches. Cytokines were measured by ELISA. Gene and protein expression were measured using reverse transcriptase polymerase chain reaction, Western blotting, and immunohistochemistry. The role of type I IFN in PAH in vivo was determined using type I IFN receptor knockout (IFNAR1(-/-)) mice. Human lung cells responded to types I and II but not III IFN correlating with relevant receptor expression. Type I, II, and III IFN levels were elevated in serum of patients with systemic sclerosis associated PAH. Serum interferon γ inducible protein 10 (IP10; CXCL10) and endothelin 1 were raised and strongly correlated together. IP10 correlated positively with pulmonary hemodynamics and serum brain natriuretic peptide and negatively with 6-minute walk test and cardiac index. Endothelial cells grown out of the blood of PAH patients were more sensitive to the effects of type I IFN than cells from healthy donors. PAH lung demonstrated increased IFNAR1 protein levels. IFNAR1(-/-) mice were protected from the effects of hypoxia on the right heart, vascular remodeling, and raised serum endothelin 1 levels. CONCLUSIONS These data indicate that type I IFN, via an action of IFNAR1, mediates PAH.
Collapse
Affiliation(s)
- Peter M George
- From the Department of Cardiothoracic Pharmacology, National Heart and Lung Institute, Imperial College London, United Kingdom (P.M.G., D.M.R., N.S.K., N.A.M., S.J.W., J.A.M.); Centre for Pharmacology and Therapeutics, Experimental Medicine, Imperial College London, Hammersmith Hospital, London, United Kingdom (E.O., O.D.D., L.Z.); Service d'Anatomie Pathologique, Centre Chirurgical Marie Lannelongue, Le Plessis-Robinson, France (P.D.); Univ. Paris-Sud, Faculté de médecine, Kremlin-Bicêtre, France (P.D., F.P., F.A., M.H.); INSERM UMR-S 999, Labex LERMIT, Hypertension Artérielle Pulmonaire: Physiopathologie et Innovation Thérapeutique, Centre Chirurgical Marie Lannelongue, Le Plessis-Robinson, France (P.D., F.P., F.A., E.F., M.H.); AP-HP, DHU TORINO, Centre de Référence de l'Hypertension Pulmonaire Sévère, Service de Pneumologie et Réanimation Respiratoire, Hôpital Bicêtre, Le Kremlin-Bicêtre, France (F.P., F.A., M.H.); Pulmonary Hypertension service, Royal Free Hospital, London, United Kingdom (B.E.S., J.G.C.); Centre for Rheumatology and Connective Tissue Diseases, Department of Inflammation, University College London, Royal Free Campus, London, United Kingdom (A.M.H.); Pulmonary Vascular Diseases Unit, Papworth Hospital NHS Trust, Papworth Everard, Cambridge (M.S., G.H., N.W.M.); and National Heart & Lung Institute, MRC & Asthma UK Centre in Allergic Mechanisms of Asthma, Centre for Respiratory Infection, Imperial College London, United Kingdom (N.B.)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Beneficial innate signaling interference for antibacterial responses by a Toll-like receptor-mediated enhancement of the MKP-IRF3 axis. Proc Natl Acad Sci U S A 2013; 110:19884-9. [PMID: 24248350 DOI: 10.1073/pnas.1320145110] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
A major function of innate immune receptors is to recognize pathogen-associated molecular patterns and then evoke immune responses appropriate to the nature of the invading pathogen(s). Because innate immune cells express various types of these receptors, distinct combinations of signaling pathways are activated in response to a given pathogen. Although the conventional wisdom is that these signaling pathways cooperate with one another to ensure an effective host response, a more nuanced view recognizes antagonism between the individual pathways, where the attenuation of a signaling pathway(s) by others may shape the immune response. In this study, we show that, on Listeria monocytogenes infection, Toll-like receptor-triggered MyD88 signaling pathways suppress type I IFN gene induction, which is detrimental to macrophage bactericidal activity. These pathways target and suppress the IFN regulatory factor 3 (IRF3) transcription factor that is activated by the stimulator of IFN genes-TANK-binding kinase-1 kinase pathway. We also provide evidence for the involvement of the MAPK phosphatase family members, which renders IRF3 hypophosphorylated on Toll-like receptor signaling by enhancing the formation of an MAPK phosphatase-IRF3-TANK-binding kinase-1 ternary complex. This study, therefore, reveals a hitherto unrecognized and important contribution of a beneficial innate signaling interference against bacterial infections.
Collapse
|
35
|
Cordero-Reyes AM, Youker KA, Torre-Amione G. The role of B-cells in heart failure. Methodist Debakey Cardiovasc J 2013; 9:15-9. [PMID: 23519014 DOI: 10.14797/mdcj-9-1-15] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Heart failure is a complex disease that has great impact on morbidity and mortality in the general population. No recent therapies have proven to be effective; however, the discovery of new potential pathophysiological mechanisms involved in heart failure expression and progression could offer novel therapeutic strategies. A number of studies have shown that the immune system may be a central mediator in the development and progression of heart failure, and here we describe how the B-cell and B-cell-mediated pathways play specific roles in the heart failure state. Therapies aimed at B-cells, either blocking antibody production or inactivating B-cell function, may suggest potential new treatment strategies.
Collapse
|
36
|
Moore TC, Petro TM. IRF3 and ERK MAP-kinases control nitric oxide production from macrophages in response to poly-I:C. FEBS Lett 2013; 587:3014-20. [PMID: 23892079 DOI: 10.1016/j.febslet.2013.07.025] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2013] [Revised: 06/20/2013] [Accepted: 07/13/2013] [Indexed: 11/18/2022]
Abstract
Understanding nitric oxide (NO) in innate anti-viral immunity and immune-mediated pathology is hampered by incomplete details of its transcriptional and signaling factors. We found in macrophages that IRF3, ERK MAP-kinases, and PKR are essential to NO production in response to RNA-virus mimic, poly I:C, a TLR3 agonist. ERK's role in NO induction may be through phosphorylation of serine-171 of IRF3 and expression of NO-inducing cytokines, IL-6 and IFN-β. However, these cytokines induced less NO in IRF3 knockout or knockdown macrophages. These findings show that ERK and IRF3 coordinate induction of NO by macrophages in response to stimulation of TLR3.
Collapse
Affiliation(s)
- Tyler C Moore
- School of Biological Sciences, University of Nebraska-Lincoln, Lincoln, NE, USA
| | | |
Collapse
|
37
|
Lu J, Bian ZY, Zhang R, Zhang Y, Liu C, Yan L, Zhang SM, Jiang DS, Wei X, Zhu XH, Chen M, Wang AB, Chen Y, Yang Q, Liu PP, Li H. Interferon regulatory factor 3 is a negative regulator of pathological cardiac hypertrophy. Basic Res Cardiol 2013; 108:326. [PMID: 23307144 DOI: 10.1007/s00395-012-0326-9] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2012] [Revised: 12/13/2012] [Accepted: 12/20/2012] [Indexed: 11/25/2022]
Abstract
Interferon regulatory factor (IRF) 3, a member of the highly conserved IRF family transcription factors, plays a pivotal role in innate immune response, apoptosis, and oncogenesis. Recent studies have implicated IRF3 in a wide range of host defense. However, whether IRF3 induces defensive responses to hypertrophic stresses such as biomechanical stress and neurohumoral factors remains unclear. Herein, we employed an IRF3-deficient mouse model, cardiac-specific IRF3-overexpression mouse model and isolated cardiomyocytes to investigate the role of IRF3 in cardiac hypertrophy induced by aortic banding (AB) or isoproterenol (ISO). The extent of cardiac hypertrophy was quantitated by echocardiography as well as by pathological and molecular analysis. Our results demonstrate that IRF3 deficiency profoundly exacerbated cardiac hypertrophy, whereas overexpression of IRF3 in the heart significantly blunted pathological cardiac remodeling induced by pressure overload. Similar results were also observed in cultured cardiomyocytes upon the treatment with ISO. Mechanistically, we discovered that IRF3 interacted with ERK2 and thereby inhibited the ERK1/2 signaling. Furthermore, inactivation of ERK1/2 by U0126 offset the IRF3-deficient-mediated hypertrophic response induced by aortic banding. Altogether, these data demonstrate that IRF3 plays a protective role in AB-induced hypertrophic response by inactivating ERK1/2 in the heart. Therefore, IRF3 could be a new target for the prevention and therapy of cardiac hypertrophy and failure.
Collapse
Affiliation(s)
- Jing Lu
- Department of Cardiology, Renmin Hospital, Cardiovascular Research Institute, Wuhan University, Wuhan, People's Republic of China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Gu J, Liu X, Wang QX, Tan HW, Guo M, Jiang WF, Zhou L. Angiotensin II increases CTGF expression via MAPKs/TGF-β1/TRAF6 pathway in atrial fibroblasts. Exp Cell Res 2012; 318:2105-15. [DOI: 10.1016/j.yexcr.2012.06.015] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2012] [Revised: 06/10/2012] [Accepted: 06/18/2012] [Indexed: 01/09/2023]
|