1
|
Wang Z, Ren Y, Zhang D, She G, Wang Y, Li G, Sun X, Zheng D, Wang Z, Deng XL, Zhao Y, Zhao L. Elevated K Ca3.1 expression by angiotensin II via the ERK/NF-κB pathway contributes to atrial fibrosis. J Mol Cell Cardiol 2025; 202:133-143. [PMID: 40122157 DOI: 10.1016/j.yjmcc.2025.03.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2024] [Revised: 03/12/2025] [Accepted: 03/19/2025] [Indexed: 03/25/2025]
Abstract
Atrial fibrillation (AF) is a prevalent cardiac arrhythmia characterized by atrial fibrosis which involves excessed proliferation and increased activity of fibroblast and myofibroblast, as well as alterations in the extracellular matrix (ECM). The specific mechanism driving fibrosis in atrial fibroblasts and myofibroblsats remains incompletely understood. This study investigates the role of the intermediate-conductance Ca2+-activated K+ channel (KCa3.1) in Angiotensin II (Ang II)-induced atrial fibrosis and elucidates the underlying mechanisms. Primary rat atrial fibroblasts/myofibroblasts were treated with Ang II to evaluate KCa3.1 expression, cells proliferation and ECM production. The involvement of ERK/NF-κB signaling pathway was assessed using specific inhibitors. Ang II treatment increased KCa3.1 expression, stimulated the proliferation of fibroblasts/myofibroblasts, and enhanced ECM production, effects that were attenuated by the Ang II receptor antagonist Losartan and the KCa3.1 inhibitor TRAM-34. Knockdown of KCa3.1 using siRNA significantly reduced Ang II-induced collagen synthesis, confirming its critical role in fibrosis. The ERK/NF-κB pathway was found to mediate Ang II-induced upregulation of KCa3.1, as evidenced by inhibition with specific inhibitors. In vivo, Ang II infusion in rats increased KCa3.1 expression and atrial fibrosis, with atria showing greater susceptibility to fibrosis compared to ventricle. These effects were mitigated by losartan and TRAM-34. In conclusion, our findings demonstrate that Ang II-induced upregulation of KCa3.1 through ERK/NF-κB pathway activation in atrial fibroblasts/myofibroblasts promotes cellular proliferation and collagen deposition, ultimately contributing to atrial fibrosis. KCa3.1 represents a promising therapeutic target for the treatment of atrial fibrosis in AF.
Collapse
Affiliation(s)
- Zujuan Wang
- Department of Pathology and Pathophysiology, Suzhou Medical College of Soochow University, 199 Ren-ai Road, Suzhou 215123, Jiangsu, China
| | - Yujie Ren
- Department of Pathology, Xi'an People's Hospital (Xian Fourth Hospital), 21 Jiefang Road, Xi'an 710004, Shaanxi, China
| | - Dongmei Zhang
- Department of Pathology and Pathophysiology, Suzhou Medical College of Soochow University, 199 Ren-ai Road, Suzhou 215123, Jiangsu, China
| | - Gang She
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 76 West Yanta Road, Xi'an 710061, Shaanxi, China
| | - Yan Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 76 West Yanta Road, Xi'an 710061, Shaanxi, China
| | - Guangyao Li
- Department of Pathology and Pathophysiology, Suzhou Medical College of Soochow University, 199 Ren-ai Road, Suzhou 215123, Jiangsu, China
| | - Xiaodong Sun
- Department of Pathology and Pathophysiology, Suzhou Medical College of Soochow University, 199 Ren-ai Road, Suzhou 215123, Jiangsu, China
| | - Dong Zheng
- Department of Pathology and Pathophysiology, Suzhou Medical College of Soochow University, 199 Ren-ai Road, Suzhou 215123, Jiangsu, China
| | - Zhongjuan Wang
- Institute of Biology and Medical Sciences, Soochow University, 199 Ren-ai Road, Suzhou 215123, China
| | - Xiu-Ling Deng
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 76 West Yanta Road, Xi'an 710061, Shaanxi, China
| | - Ying Zhao
- Department of Pathology and Pathophysiology, Suzhou Medical College of Soochow University, 199 Ren-ai Road, Suzhou 215123, Jiangsu, China.
| | - Limei Zhao
- Department of Pathology and Pathophysiology, Suzhou Medical College of Soochow University, 199 Ren-ai Road, Suzhou 215123, Jiangsu, China.
| |
Collapse
|
2
|
Moccia F, Totaro A, Guerra G, Testa G. Ca 2+ Signaling in Cardiac Fibroblasts: An Emerging Signaling Pathway Driving Fibrotic Remodeling in Cardiac Disorders. Biomedicines 2025; 13:734. [PMID: 40149710 PMCID: PMC11940070 DOI: 10.3390/biomedicines13030734] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2025] [Revised: 03/13/2025] [Accepted: 03/15/2025] [Indexed: 03/29/2025] Open
Abstract
Cardiac fibrosis is a scarring event that occurs in the myocardium in response to multiple cardiovascular disorders, such as acute myocardial infarction (AMI), ischemic cardiomyopathy, dilated cardiomyopathy, hypertensive heart disease, inflammatory heart disease, diabetic cardiomyopathy, and aortic stenosis. Fibrotic remodeling is mainly sustained by the differentiation of fibroblasts into myofibroblasts, which synthesize and secrete most of the extracellular matrix (ECM) proteins. An increase in the intracellular Ca2+ concentration ([Ca2+]i) in cardiac fibroblasts is emerging as a critical mediator of the fibrogenic signaling cascade. Herein, we review the mechanisms that may shape intracellular Ca2+ signals involved in fibroblast transdifferentiation into myofibroblasts. We focus our attention on the functional interplay between inositol-1,4,5-trisphosphate (InsP3) receptors (InsP3Rs) and store-operated Ca2+ entry (SOCE). In accordance with this, InsP3Rs and SOCE drive the Ca2+ response elicited by Gq-protein coupled receptors (GqPCRs) that promote fibrotic remodeling. Then, we describe the additional mechanisms that sustain extracellular Ca2+ entry, including receptor-operated Ca2+ entry (ROCE), P2X receptors, Transient Receptor Potential (TRP) channels, and Piezo1 channels. In parallel, we discuss the pharmacological manipulation of the Ca2+ handling machinery as a promising approach to mitigate or reverse fibrotic remodeling in cardiac disorders.
Collapse
Affiliation(s)
- Francesco Moccia
- Department of Medicine and Health Sciences “V. Tiberio”, University of Molise, 86100 Campobasso, Italy; (A.T.); (G.G.); (G.T.)
| | | | | | | |
Collapse
|
3
|
She G, Hai XX, Jia LY, Zhang YJ, Ren YJ, Pang ZD, Wu LH, Han MZ, Zhang Y, Li JJ, Bai RY, Lai BC, Yang YY, Sadoshima J, Du XJ, Deng XL, Zhang Y. Hippo pathway activation mediates cardiomyocyte ferroptosis to promote dilated cardiomyopathy through downregulating NFS1. Redox Biol 2025; 82:103597. [PMID: 40107016 DOI: 10.1016/j.redox.2025.103597] [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: 02/02/2025] [Revised: 03/13/2025] [Accepted: 03/13/2025] [Indexed: 03/22/2025] Open
Abstract
Cardiomyocyte loss by regulated death modes, like apoptosis and ferroptosis, has been implicated in the development of dilated cardiomyopathy (DCM). It remains unclear whether cardiomyocyte ferroptosis occurs as a consequence of Hippo pathway activation. Using a mouse model of DCM by overexpression of Mst1 transgene (Mst1-TG) leading to Hippo pathway activation, we showed that cardiomyocyte ferroptosis was evident by transcriptomic profiles, elevated mitochondrial Fe2+ content, increased levels of lipid peroxidation and obvious mitochondrial damage. Transcriptome revealed significant alterations of genes participating in iron metabolism and lipid peroxidation. Treatment of Mst1-TG mice with the ferroptosis inhibitor ferrostatin-1 reduced cardiomyocyte ferroptosis and improved cardiac function. Using heart samples from human patients with DCM, we also found significant cardiomyocyte loss and lipid peroxidation. In cultured cardiomyocytes, ferroptosis was induced by treatment with erastin or YAP inhibitor verteporfin, and cell ferroptosis under these conditions was largely prevented by either iron chelation or Mst1 gene knockdown. In a strain of transgenic mice with cardiomyocyte inactivation of Mst1 (dnMst1-TG), erastin-induced ferroptosis and cardiac dysfunction, seen in control mice, were mitigated. Mechanistically, nuclear YAP and YY1 were shown to interact and bind to the Nfs1 promoter, thus mediating downregulation of Nfs1 (encoding cysteine desulfurase). Subsequent inhibition of iron-sulfur cluster (ISC) biosynthesis promoted cardiomyocyte ferroptosis and DCM phenotype. Restoration of Nfs1 expression was achieved by treatment of Mst1-TG mice with AAV9-Nfs1 virus, which alleviated ferroptosis, mitochondrial damage and DCM phenotype. In conclusion, in the DCM model with Hippo pathway activation, our findings unravel that NFS1 downregulation occurs and leads to insufficient ISC biosynthesis and cardiomyocyte ferroptosis. Our findings implicate that restoration of cardiomyocyte NFS1 level may represent a new therapeutic strategy for DCM.
Collapse
Affiliation(s)
- Gang She
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 76 West Yanta Road, Xi'an, 710061, Shaanxi, China; Department of Cardiology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Xia-Xia Hai
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 76 West Yanta Road, Xi'an, 710061, Shaanxi, China
| | - Li-Ye Jia
- School of Nursing and Rehabilitation, Xi'an Medical University, 1 Xinwang Road, Xi'an, 710021, Shaanxi, China
| | - Yong-Jian Zhang
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, 88 Zhuque Street, Xi'an, 710061, Shaanxi, China
| | - Yu-Jie Ren
- Department of Pathology, Xi'an People's Hospital (Xian Fourth Hospital), Affiliated to Xi'an Jiaotong University Health Science Center, 21 Jiefang Road, Xi'an, 710005, Shaanxi, China
| | - Zheng-Da Pang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 76 West Yanta Road, Xi'an, 710061, Shaanxi, China
| | - Lin-Hong Wu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 76 West Yanta Road, Xi'an, 710061, Shaanxi, China
| | - Meng-Zhuan Han
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 76 West Yanta Road, Xi'an, 710061, Shaanxi, China
| | - Yu Zhang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 76 West Yanta Road, Xi'an, 710061, Shaanxi, China
| | - Jing-Jing Li
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 76 West Yanta Road, Xi'an, 710061, Shaanxi, China
| | - Ru-Yue Bai
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 76 West Yanta Road, Xi'an, 710061, Shaanxi, China
| | - Bao-Chang Lai
- Cardiovascular Research Centre, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 76 West Yanta Road, Xi'an, 710061, Shaanxi, China
| | - Yi-Yi Yang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 76 West Yanta Road, Xi'an, 710061, Shaanxi, China
| | - Junichi Sadoshima
- Department of Cell Biology and Molecular Medicine, Rutgers New Jersey Medical School, New Jersey, United States of America
| | - Xiao-Jun Du
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 76 West Yanta Road, Xi'an, 710061, Shaanxi, China; Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, Victoria, 3004, Australia
| | - Xiu-Ling Deng
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 76 West Yanta Road, Xi'an, 710061, Shaanxi, China.
| | - Yi Zhang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 76 West Yanta Road, Xi'an, 710061, Shaanxi, China.
| |
Collapse
|
4
|
Zhang D, Li G, Liu X, Wang Y, Wu J, Ren Y, She G, Zheng D, Zhao Y, Deng XL, Li M, Zhao L. K Ca3.1 upregulation mediated by Ang II-induced JNK/AP-1 activation contributes to atrial fibrosis. Cell Signal 2025; 131:111731. [PMID: 40064281 DOI: 10.1016/j.cellsig.2025.111731] [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: 11/29/2024] [Revised: 02/27/2025] [Accepted: 03/07/2025] [Indexed: 03/15/2025]
Abstract
Atrial fibrillation is strongly associated with an increased risk of embolism, stroke, and heart failure. Current therapeutic approaches often have limited efficacy, and controlling atrial fibrosis remains a critical objective for upstream therapies. The specific mechanisms driving atrial fibrosis remain incompletely understood. The intermediate-conductance calcium-activated potassium channel KCa3.1 has been implicated in promoting fibroblast activation in various fibrotic diseases. This study investigates the role of angiotensin II (Ang II) in regulating KCa3.1, as well as its involvement in the pathogenesis of atrial fibrosis and the underlying signaling mechanisms. In a rat model, chronic Ang II infusion for 4 weeks induced atrial fibrosis, which was significantly attenuated by TRAM-34, a specific KCa3.1 channel blocker. In cultured rat atrial fibroblasts, Ang II treatment promoted fibroblast differentiation, proliferation, migration and collagen production, effects that were suppressed by TRAM-34 and KCa3.1 knockdown. Overexpression of KCa3.1 in fibroblasts further confirmed its pro-fibrotic role. Mechanistically, Ang II upregulated KCa3.1 expression and current density by activating the JNK/AP-1 signaling pathway. This involved phosphorylation of JNK, c-Jun, and c-Fos, leading to the formation of c-Jun/c-Fos heterodimers that directly bound to the KCa3.1 promoter to enhance its transcription. Together, these findings demonstrate that KCa3.1 mediates fibroblast activation and atrial fibrosis through the JNK/AP-1 pathway.
Collapse
Affiliation(s)
- Dongmei Zhang
- Department of Pathology and Pathophysiology, Suzhou Medical College of Soochow University, 199 Ren-ai Road, Suzhou 215123, Jiangsu, China
| | - Guangyao Li
- Department of Pathology and Pathophysiology, Suzhou Medical College of Soochow University, 199 Ren-ai Road, Suzhou 215123, Jiangsu, China
| | - Xiang Liu
- Department of Pathology and Pathophysiology, Suzhou Medical College of Soochow University, 199 Ren-ai Road, Suzhou 215123, Jiangsu, China
| | - Yan Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 76 West Yanta Road, Xi'an 710061, Shaanxi, China
| | - Jie Wu
- Department of Pathology and Pathophysiology, Suzhou Medical College of Soochow University, 199 Ren-ai Road, Suzhou 215123, Jiangsu, China
| | - Yujie Ren
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 76 West Yanta Road, Xi'an 710061, Shaanxi, China
| | - Gang She
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 76 West Yanta Road, Xi'an 710061, Shaanxi, China
| | - Dong Zheng
- Department of Pathology and Pathophysiology, Suzhou Medical College of Soochow University, 199 Ren-ai Road, Suzhou 215123, Jiangsu, China
| | - Yinxia Zhao
- Central Laboratory, Shanghai Xuhui Central Hospital, 366 North Longchuan Road, Shanghai 200031, China
| | - Xiu-Ling Deng
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 76 West Yanta Road, Xi'an 710061, Shaanxi, China
| | - Min Li
- Institute of Biology and Medical Sciences, Soochow University, 199 Ren-ai Road, Suzhou 215123, Jiangsu, China.
| | - Limei Zhao
- Department of Pathology and Pathophysiology, Suzhou Medical College of Soochow University, 199 Ren-ai Road, Suzhou 215123, Jiangsu, China; MOE Key Laboratory of Geriatric Diseases and Immunology, School of Basic Medical Sciences, Suzhou Medical College of Soochow University, Suzhou 215123, Jiangsu, China.
| |
Collapse
|
5
|
Du XJ, She G, Wu W, Deng XL. Coupling of β-adrenergic and Hippo pathway signaling: Implications for heart failure pathophysiology and metabolic therapy. Mitochondrion 2024; 78:101941. [PMID: 39122227 DOI: 10.1016/j.mito.2024.101941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 07/17/2024] [Accepted: 08/03/2024] [Indexed: 08/12/2024]
Abstract
Activation of the sympatho-β-adrenergic receptor (βAR) system is the hallmark of heart disease with adverse consequences that facilitate the onset and progression of heart failure (HF). Use of β-blocking drugs has become the front-line therapy for HF. Last decade has witnessed progress in research demonstrating a pivotal role of Hippo pathway in cardiomyopathy and HF. Clinical studies have revealed myocardial Hippo pathway activation/YAP-TEAD1 inactivation in several types of human cardiomyopathy. Experimental activation of cardiac Hippo signaling or inhibition of YAP-TEAD1 have been shown to leads dilated cardiomyopathy with severe mitochondrial dysfunction and metabolic reprogramming. Studies have also convincingly shown that stimulation of βAR activates cardiac Hippo pathway with inactivation of the down-stream effector molecules YAP/TAZ. There is strong evidence for the adverse consequences of the βAR-Hippo signaling leading to HF. In addition to promoting cardiomyocyte death and fibrosis, recent progress is the demonstration of mitochondrial dysfunction and metabolic reprogramming mediated by βAR-Hippo pathway signaling. Activation of cardiac βAR-Hippo signaling is potent in downregulating a range of mitochondrial and metabolic genes, whereas expression of pro-inflammatory and pro-fibrotic factors are upregulated. Coupling of βAR-Hippo pathway signaling is mediated by several kinases, mechanotransduction and/or Ca2+ signaling, and can be blocked by β-antagonists. Demonstration of the converge of βAR signaling and Hippo pathway bears implications for a better understanding on the role of enhanced sympathetic nervous activity, efficacy of β-antagonists, and metabolic therapy targeting this pathway in HF. In this review we summarize the progress and discuss future research directions in this field.
Collapse
Affiliation(s)
- Xiao-Jun Du
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, and Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an Jiaotong University Health Science Center, 76 West Yanta Road, Xi'an, Shaanxi 710061, China; Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, Victoria 3004, Australia,.
| | - Gang She
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, and Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an Jiaotong University Health Science Center, 76 West Yanta Road, Xi'an, Shaanxi 710061, China
| | - Wei Wu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, and Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an Jiaotong University Health Science Center, 76 West Yanta Road, Xi'an, Shaanxi 710061, China; Department of Cardiology, Shaanxi Provincial Hospital and the Third Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Xiu-Ling Deng
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, and Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an Jiaotong University Health Science Center, 76 West Yanta Road, Xi'an, Shaanxi 710061, China
| |
Collapse
|
6
|
Xiao H, Wang T, Gao B, Liu J, Li S, Ma J. The effects of a galectin-3 inhibitor on bladder pain syndrome in mice with cyclophosphamide-induced cystitis. Neurourol Urodyn 2024; 43:754-766. [PMID: 38356381 DOI: 10.1002/nau.25415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 12/13/2023] [Accepted: 01/09/2024] [Indexed: 02/16/2024]
Abstract
AIMS To explore the effect of blocking galectin-3 in the bladder pain syndrome associated with interstitial cystitis. METHODS A galectin-3 inhibitor was used to treat mice with cyclophosphamide-induced cystitis. The expression of galectin-3 in bladder tissues and urine was examined by immunohistochemistry and enzyme-linked immunosorbent assay (ELISA), respectively. Suprapubic-pelvic pain, bladder voiding, bladder pain-like nociceptive behavior, and referred hyperalgesia were assessed. The weights of the bladders were also measured, and inflammatory cell infiltration and inflammatory cytokine levels were examined by histopathological evaluation. The inflammatory cytokines interleukin 1β (IL-1β), nerve growth factor (NGF), IL-6, and tumor necrosis factor α (TNF-α) were measured by ELISA. RESULTS Increases in galectin-3 levels, inflammation, bladder weight, and bladder pain-related symptoms were observed in bladders with cyclophosphamide-induced cystitis. Administration of the galectin-3 inhibitor significantly mitigated bladder pain-related symptoms and inflammatory response. In response to the 500 μM dose of the galectin-3 inhibitor, nociceptive behaviors, nociceptive score, and bladder-to-body weight ratios were reduced by 65.1%, 65.3%, and 40.3%, respectively, while 500 μM Gal-3 inhibitor increased pelvic pain threshold by 86.7%. Moreover, galectin-3 inhibitor treatment inhibited the inflammation. Compared to untreated CYP-induced mice, there were significant changes in the levels of IL-1β (41.72 ± 2.05 vs. 18.91 ± 2.26 pg/mg tissues), NGF (9.64 ± 0.38 vs. 1.88 ± 0.05 pg/mg tissues), IL-6 (42.67 + 1.51 vs. 21.26 + 2.78 pg/mg tissues, and TNF-α (22.02 ± 1.08 vs. 10.70 ± 0.80 pg/mg tissues) in response to the highest dose of the Gal-3 inhibitor subgroup (500 μM), and 500 μM Gal-3 inhibitor reduced mast cell infiltration ratios by 71.8%. CONCLUSIONS The galectin-3 inhibitor relieved pelvic pain, urinary symptoms, and bladder inflammation in mice with cyclophosphamide-induced cystitis. Thus, galectin-3 inhibitors may be novel agents in interstitial cystitis treatment.
Collapse
Affiliation(s)
- Helong Xiao
- Department of Surgery, Hebei Medical University, Shijiazhuang, Hebei, China
- Department of Urology, Hebei General Hospital, Shijiazhuang, Hebei, China
| | - Ting Wang
- Department of Cardiology, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Bo Gao
- Department of Urology, Hebei General Hospital, Shijiazhuang, Hebei, China
| | - Junjiang Liu
- Department of Urology, Hebei General Hospital, Shijiazhuang, Hebei, China
| | - Shoubin Li
- Department of Urology, Hebei General Hospital, Shijiazhuang, Hebei, China
| | - Jianguo Ma
- Department of Urology, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| |
Collapse
|
7
|
Qian JF, Liang SQ, Wang QY, Xu JC, Luo W, Huang WJ, Wu GJ, Liang G. Isoproterenol induces MD2 activation by β-AR-cAMP-PKA-ROS signalling axis in cardiomyocytes and macrophages drives inflammatory heart failure. Acta Pharmacol Sin 2024; 45:531-544. [PMID: 37919475 PMCID: PMC10834947 DOI: 10.1038/s41401-023-01179-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Accepted: 09/29/2023] [Indexed: 11/04/2023]
Abstract
Cardiac inflammation contributes to heart failure (HF) induced by isoproterenol (ISO) through activating β-adrenergic receptors (β-AR). Recent evidence shows that myeloid differentiation factor 2 (MD2), a key protein in endotoxin-induced inflammation, mediates inflammatory heart diseases. In this study, we investigated the role of MD2 in ISO-β-AR-induced heart injuries and HF. Mice were infused with ISO (30 mg·kg-1·d-1) via osmotic mini-pumps for 2 weeks. We showed that MD2 in cardiomyocytes and cardiac macrophages was significantly increased and activated in the heart tissues of ISO-challenged mice. Either MD2 knockout or administration of MD2 inhibitor L6H21 (10 mg/kg every 2 days, i.g.) could prevent mouse hearts from ISO-induced inflammation, remodelling and dysfunction. Bone marrow transplantation study revealed that both cardiomyocyte MD2 and bone marrow-derived macrophage MD2 contributed to ISO-induced cardiac inflammation and injuries. In ISO-treated H9c2 cardiomyocyte-like cells, neonatal rat primary cardiomyocytes and primary mouse peritoneal macrophages, MD2 knockout or pre-treatment with L6H21 (10 μM) alleviated ISO-induced inflammatory responses, and the conditioned medium from ISO-challenged macrophages promoted the hypertrophy and fibrosis in cardiomyocytes and fibroblasts. We demonstrated that ISO induced MD2 activation in cardiomyocytes via β1-AR-cAMP-PKA-ROS signalling axis, and induced inflammatory responses in macrophages via β2-AR-cAMP-PKA-ROS axis. This study identifies MD2 as a key inflammatory mediator and a promising therapeutic target for ISO-induced heart failure.
Collapse
Affiliation(s)
- Jin-Fu Qian
- Department of Cardiology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, China
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Shi-Qi Liang
- Department of Cardiology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, China
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Qin-Yan Wang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Jia-Chen Xu
- Department of Cardiology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, China
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Wu Luo
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China
- Medical Research Center, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou, 325035, China
| | - Wei-Jian Huang
- Department of Cardiology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, China
| | - Gao-Jun Wu
- Department of Cardiology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, China.
| | - Guang Liang
- Department of Cardiology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, China.
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China.
| |
Collapse
|
8
|
Jiang D, Guo R, Dai R, Knoedler S, Tao J, Machens HG, Rinkevich Y. The Multifaceted Functions of TRPV4 and Calcium Oscillations in Tissue Repair. Int J Mol Sci 2024; 25:1179. [PMID: 38256251 PMCID: PMC10816018 DOI: 10.3390/ijms25021179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 01/09/2024] [Accepted: 01/16/2024] [Indexed: 01/24/2024] Open
Abstract
The transient receptor potential vanilloid 4 (TRPV4) specifically functions as a mechanosensitive ion channel and is responsible for conveying changes in physical stimuli such as mechanical stress, osmotic pressure, and temperature. TRPV4 enables the entry of cation ions, particularly calcium ions, into the cell. Activation of TRPV4 channels initiates calcium oscillations, which trigger intracellular signaling pathways involved in a plethora of cellular processes, including tissue repair. Widely expressed throughout the body, TRPV4 can be activated by a wide array of physicochemical stimuli, thus contributing to sensory and physiological functions in multiple organs. This review focuses on how TRPV4 senses environmental cues and thereby initiates and maintains calcium oscillations, critical for responses to organ injury, tissue repair, and fibrosis. We provide a summary of TRPV4-induced calcium oscillations in distinct organ systems, along with the upstream and downstream signaling pathways involved. In addition, we delineate current animal and disease models supporting TRPV4 research and shed light on potential therapeutic targets for modulating TRPV4-induced calcium oscillation to promote tissue repair while reducing tissue fibrosis.
Collapse
Affiliation(s)
- Dongsheng Jiang
- Institute of Regenerative Biology and Medicine, Helmholtz Center Munich, 81377 Munich, Germany; (R.G.); (R.D.); (S.K.)
| | - Ruiji Guo
- Institute of Regenerative Biology and Medicine, Helmholtz Center Munich, 81377 Munich, Germany; (R.G.); (R.D.); (S.K.)
- Department of Plastic and Hand Surgery, Klinikum Rechts der Isar, School of Medicine, Technical University of Munich, 81675 Munich, Germany;
| | - Ruoxuan Dai
- Institute of Regenerative Biology and Medicine, Helmholtz Center Munich, 81377 Munich, Germany; (R.G.); (R.D.); (S.K.)
| | - Samuel Knoedler
- Institute of Regenerative Biology and Medicine, Helmholtz Center Munich, 81377 Munich, Germany; (R.G.); (R.D.); (S.K.)
- Department of Plastic and Hand Surgery, Klinikum Rechts der Isar, School of Medicine, Technical University of Munich, 81675 Munich, Germany;
- Division of Plastic Surgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02152, USA
| | - Jin Tao
- Department of Physiology and Neurobiology and Centre for Ion Channelopathy, Medical College of Soochow University, Suzhou 215123, China;
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, Soochow University, Suzhou 215123, China
| | - Hans-Günther Machens
- Department of Plastic and Hand Surgery, Klinikum Rechts der Isar, School of Medicine, Technical University of Munich, 81675 Munich, Germany;
| | - Yuval Rinkevich
- Institute of Regenerative Biology and Medicine, Helmholtz Center Munich, 81377 Munich, Germany; (R.G.); (R.D.); (S.K.)
| |
Collapse
|
9
|
Kim JS, Ru F, Meeker S, Undem BJ. Direct activation of airway sensory C-fibers by SARS-CoV-2 S1 spike protein. Physiol Rep 2023; 11:e15900. [PMID: 38123162 PMCID: PMC10733116 DOI: 10.14814/phy2.15900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Revised: 12/02/2023] [Accepted: 12/04/2023] [Indexed: 12/23/2023] Open
Abstract
Respiratory viral infection can lead to activation of sensory afferent nerves as indicated by the consequential sore throat, sneezing, coughing, and reflex secretions. In addition to causing troubling symptoms, sensory nerve activation likely accelerates viral spreading. The mechanism how viruses activate sensory nerve terminals during infection is unknown. In this study, we investigate whether coronavirus spike protein activates sensory nerves terminating in the airways. We used isolated vagally-innervated mouse trachea-lung preparation for two-photon microscopy and extracellular electrophysiological recordings. Using two-photon Ca2+ imaging, we evaluated a total number of 786 vagal bronchopulmonary nerves in six experiments. Approximately 49% of the sensory fibers were activated by S1 protein (4 μg/mL intratracheally). Extracellular nerve recording showed the S1 protein evoked action potential discharge in sensory C-fibers; of 39 airway C-fibers (one fiber per mouse), 17 were activated. Additionally, Fura-2 Ca2+ imaging was performed on neurons dissociated from vagal sensory ganglia (n = 254 from 22 mice). The result showed that 63% of neurons responded to S1 protein. SARS-CoV-2 S1 protein can lead to direct activation of sensory C-fiber nerve terminals in the bronchopulmonary tract. Direct activation of C-fibers may contribute to coronavirus symptoms, and amplify viral spreading in a population.
Collapse
Affiliation(s)
- Joyce S. Kim
- Department of MedicineJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Fei Ru
- Department of MedicineJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Sonya Meeker
- Department of MedicineJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Bradley J. Undem
- Department of MedicineJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| |
Collapse
|
10
|
Frangogiannis NG. Targeting galectin-3 in myocardial infarction: a unique opportunity for biomarker-guided therapy. Cardiovasc Res 2023; 119:2495-2496. [PMID: 37772841 PMCID: PMC10676455 DOI: 10.1093/cvr/cvad156] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Accepted: 09/04/2023] [Indexed: 09/30/2023] Open
Affiliation(s)
- Nikolaos G Frangogiannis
- The Wilf Family Cardiovascular Research Institute, Department of Medicine (Cardiology), Department of Microbiology and Immunology, Albert Einstein College of Medicine, 1300 Morris Park Avenue Forchheimer G46B, Bronx NY 10461, USA
| |
Collapse
|
11
|
Zhang Y, Ma XZ, Zhao XY, Li JJ, Ma S, Pang ZD, Xu J, Du XJ, Deng XL, Wang JH. AGEs-RAGE-KCa3.1 pathway mediates palmitic acid-induced migration of PBMCs from patients with type 2 diabetes. Heliyon 2023; 9:e14823. [PMID: 37025887 PMCID: PMC10070889 DOI: 10.1016/j.heliyon.2023.e14823] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Revised: 03/08/2023] [Accepted: 03/17/2023] [Indexed: 03/29/2023] Open
Abstract
Type 2 diabetes mellitus (T2DM) is characterized by chronic low-grade systemic inflammation. Tissue infiltration by monocyte migration contributes to the pathogenesis of vascular complications in T2DM. We studied the role of intermediate-conductance Ca2+-activated K+ (KCa3.1) channels in the palmitic acid (PA)-induced migration of peripheral blood mononuclear cells (PBMCs) from T2DM patients and the influence of advanced glycation endproducts (AGEs). A total of 49 T2DM patients and 33 healthy subjects was recruited into this study. Using flow cytometry and Western blotting analysis as well as cell migration assay, we found that there was a significant decrease in frequency of T lymphocytes and monocytes in CD45+ leukocyte population. PA at 100 μM stimulated migration of PBMCs from T2DM individuals, which was inhibited by the specific KCa3.1 channel blocker TRAM-34 (1 μM). The PBMC migration was positively correlated with glycosylated hemoglobin A1 chain (HbA1c) level of T2DM patients, an indicator of AGEs, and PBMCs with higher level of HbA1c showed upregulated expression of toll-like receptor (TLR) 2/4 and KCa3.1 channels. In THP-1 cells, AGEs at 200 μg/ml increased protein expression of TLR 2/4 and KCa3.1 channels, and were synergistically involved in PA-induced migration through receptors of AGEs (RAGE)-mediated KCa3.1 upregulation. In conclusion, in PBMCs of T2DM patients, AGEs promotes PA-induced migration via upregulation of TLR2/4 and KCa3.1 channels.
Collapse
|
12
|
Bekedam FT, Goumans MJ, Bogaard HJ, de Man FS, Llucià-Valldeperas A. Molecular mechanisms and targets of right ventricular fibrosis in pulmonary hypertension. Pharmacol Ther 2023; 244:108389. [PMID: 36940790 DOI: 10.1016/j.pharmthera.2023.108389] [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: 11/29/2022] [Revised: 02/19/2023] [Accepted: 03/16/2023] [Indexed: 03/23/2023]
Abstract
Right ventricular fibrosis is a stress response, predominantly mediated by cardiac fibroblasts. This cell population is sensitive to increased levels of pro-inflammatory cytokines, pro-fibrotic growth factors and mechanical stimulation. Activation of fibroblasts results in the induction of various molecular signaling pathways, most notably the mitogen-activated protein kinase cassettes, leading to increased synthesis and remodeling of the extracellular matrix. While fibrosis confers structural protection in response to damage induced by ischemia or (pressure and volume) overload, it simultaneously contributes to increased myocardial stiffness and right ventricular dysfunction. Here, we review state-of-the-art knowledge of the development of right ventricular fibrosis in response to pressure overload and provide an overview of all published preclinical and clinical studies in which right ventricular fibrosis was targeted to improve cardiac function.
Collapse
Affiliation(s)
- F T Bekedam
- Amsterdam UMC location Vrije Universiteit Amsterdam, PHEniX laboratory, Department of Pulmonary Medicine, De Boelelaan 1117, Amsterdam, the Netherlands; Amsterdam Cardiovascular Sciences, Pulmonary Hypertension and Thrombosis, Amsterdam, the Netherlands
| | - M J Goumans
- Department of Cell and Chemical Biology, Leiden UMC, 2300 RC Leiden, the Netherlands
| | - H J Bogaard
- Amsterdam UMC location Vrije Universiteit Amsterdam, PHEniX laboratory, Department of Pulmonary Medicine, De Boelelaan 1117, Amsterdam, the Netherlands; Amsterdam Cardiovascular Sciences, Pulmonary Hypertension and Thrombosis, Amsterdam, the Netherlands
| | - F S de Man
- Amsterdam UMC location Vrije Universiteit Amsterdam, PHEniX laboratory, Department of Pulmonary Medicine, De Boelelaan 1117, Amsterdam, the Netherlands; Amsterdam Cardiovascular Sciences, Pulmonary Hypertension and Thrombosis, Amsterdam, the Netherlands.
| | - A Llucià-Valldeperas
- Amsterdam UMC location Vrije Universiteit Amsterdam, PHEniX laboratory, Department of Pulmonary Medicine, De Boelelaan 1117, Amsterdam, the Netherlands; Amsterdam Cardiovascular Sciences, Pulmonary Hypertension and Thrombosis, Amsterdam, the Netherlands.
| |
Collapse
|
13
|
Li H, Cao Z, Wang L, Li J, Cheng X, Tang Y, Xing M, Yao P. Chronic high-fat diet induces galectin-3 and TLR4 to activate NLRP3 inflammasome in NASH. J Nutr Biochem 2023; 112:109217. [PMID: 36402251 DOI: 10.1016/j.jnutbio.2022.109217] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2021] [Revised: 05/01/2022] [Accepted: 09/27/2022] [Indexed: 11/19/2022]
Abstract
NOD-like receptor protein 3 (NLRP3) inflammasome activation triggers inflammation progression in some metabolism disorders, frequently accompanying the up-regulation of galectin-3 (Gal-3). However, the precise mechanisms of Gal-3 activating NLRP3 inflammasome remain unclear in nonalcoholic steatohepatitis (NASH). Here, male C57BL/6J mice were fed by high-fat diet (HFD) for 32 weeks to induce NASH and then the hepatic damage, cytokines, Gal-3 and TLR4 expression, and NLRP3 inflammasome activation were examined. Such indicators were similarly determined when HepG2 cells were co-incubated with palmitic acid (PA, 200 μM), β-lactose, and TAK-242, or pre-transfected with TLR4. Immunofluorescence, immunohistochemistry, and co-immunoprecipitation were conducted to confirm the potential interaction between Gal-3 and TLR4. To further identify the inflammatory regulation roles of Gal-3 and its terminals in TLR4/NLRP3, HepG2 cells were transfected with Gal-3 and its variants. Chronic HFD induced sustained hepatic steatosis and inflammatory injury, with increased inflammatory cytokines, Gal-3 and TLR4 expression, and NLRP3 inflammasome activation. Similar changes were found in PA-dosed HepG2 cells, which were rescued by β-lactose but deteriorated with TLR4 overexpression. However, TAK-242 treatment decreased AST, ALT, cytokines, and normalized NLRP3, caspase-1, and ASC expression. Furthermore, TLR4 was pulled down when Gal-3 was enriched. Only full-length Gal-3 and its carbohydrate recognition domain (CRD) promoted cytokines, TLR4 expression, and NLRP3 inflammasome activation. Thus, gal-3 may induce chronic HFD-derived NASH progression by activating TLR4-mediating NLRP3 inflammasome via its CRD, which sheds new light on candidate target for the treatment and prevention of NASH inflammation despite further research for its precise roles in the future.
Collapse
Affiliation(s)
- Hongxia Li
- Department of Nutrition and Food Hygiene, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Hubei Key Laboratory of Food Nutrition and Safety, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Ministry of Education Key Laboratory of Environment, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhiqiang Cao
- Department of Nutrition and Food Hygiene, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Hubei Key Laboratory of Food Nutrition and Safety, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Ministry of Education Key Laboratory of Environment, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lili Wang
- Department of Nutrition and Food Hygiene, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Hubei Key Laboratory of Food Nutrition and Safety, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Ministry of Education Key Laboratory of Environment, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Juan Li
- Key Laboratory of Environmental Health, Ministry of Education, Department of Toxicology, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Xueer Cheng
- Department of Nutrition and Food Hygiene, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Hubei Key Laboratory of Food Nutrition and Safety, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Ministry of Education Key Laboratory of Environment, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuhan Tang
- Department of Nutrition and Food Hygiene, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Hubei Key Laboratory of Food Nutrition and Safety, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Ministry of Education Key Laboratory of Environment, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Mingyou Xing
- Department of Infectious Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Ping Yao
- Department of Nutrition and Food Hygiene, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Hubei Key Laboratory of Food Nutrition and Safety, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Ministry of Education Key Laboratory of Environment, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
14
|
Brochet P, Ianni BM, Laugier L, Frade AF, Silva Nunes JP, Teixeira PC, Mady C, Ferreira LRP, Ferré Q, Santos RHB, Kuramoto A, Cabantous S, Steffen S, Stolf AN, Pomerantzeff P, Fiorelli AI, Bocchi EA, Pissetti CW, Saba B, Cândido DDS, Dias FC, Sampaio MF, Gaiotto FA, Marin-Neto JA, Fragata A, Zaniratto RCF, Siqueira S, Peixoto GDL, Rigaud VOC, Bacal F, Buck P, Almeida RR, Lin-Wang HT, Schmidt A, Martinelli M, Hirata MH, Donadi EA, Costa Pereira A, Rodrigues Junior V, Puthier D, Kalil J, Spinelli L, Cunha-Neto E, Chevillard C. Epigenetic regulation of transcription factor binding motifs promotes Th1 response in Chagas disease cardiomyopathy. Front Immunol 2022; 13:958200. [PMID: 36072583 PMCID: PMC9441916 DOI: 10.3389/fimmu.2022.958200] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 07/26/2022] [Indexed: 01/03/2023] Open
Abstract
Chagas disease, caused by the protozoan Trypanosoma cruzi, is an endemic parasitic disease of Latin America, affecting 7 million people. Although most patients are asymptomatic, 30% develop complications, including the often-fatal Chronic Chagasic Cardiomyopathy (CCC). Although previous studies have demonstrated some genetic deregulations associated with CCCs, the causes of their deregulations remain poorly described. Based on bulk RNA-seq and whole genome DNA methylation data, we investigated the genetic and epigenetic deregulations present in the moderate and severe stages of CCC. Analysis of heart tissue gene expression profile allowed us to identify 1407 differentially expressed transcripts (DEGs) specific from CCC patients. A tissue DNA methylation analysis done on the same tissue has permitted the identification of 92 regulatory Differentially Methylated Regions (DMR) localized in the promoter of DEGs. An in-depth study of the transcription factors binding sites (TFBS) in the DMRs corroborated the importance of TFBS’s DNA methylation for gene expression in CCC myocardium. TBX21, RUNX3 and EBF1 are the transcription factors whose binding motif appears to be affected by DNA methylation in the largest number of genes. By combining both transcriptomic and methylomic analysis on heart tissue, and methylomic analysis on blood, 4 biological processes affected by severe CCC have been identified, including immune response, ion transport, cardiac muscle processes and nervous system. An additional study on blood methylation of moderate CCC samples put forward the importance of ion transport and nervous system in the development of the disease.
Collapse
Affiliation(s)
- Pauline Brochet
- Institut National de la Santé Et de la Recherche Médicale (INSERM), Unité Mixte de Recherche (UMR)_1090, Aix Marseille Université, TAGC Theories and Approaches of Genomic Complexity, Institut MarMaRa, Marseille, France
| | - Barbara Maria Ianni
- Laboratory of Immunology, Heart Institute Instituto do Coração (InCor), University of São Paulo, School of Medicine, São Paulo, Brazil
| | - Laurie Laugier
- Aix Marseille Université, Génétique et Immunologie des Maladies Parasitaires, Inserm, UMR_906, Marseille, France
| | - Amanda Farage Frade
- Laboratory of Immunology, Heart Institute Instituto do Coração (InCor), University of São Paulo, School of Medicine, São Paulo, Brazil
- Division of Clinical Immunology and Allergy, University of São Paulo, School of Medicine, São Paulo, Brazil
- Instituto Nacional de Ciência e Tecnologia, INCT, III- Institute for Investigation in Immunology, São Paulo, Brazil
| | - João Paulo Silva Nunes
- Institut National de la Santé Et de la Recherche Médicale (INSERM), Unité Mixte de Recherche (UMR)_1090, Aix Marseille Université, TAGC Theories and Approaches of Genomic Complexity, Institut MarMaRa, Marseille, France
- Laboratory of Immunology, Heart Institute Instituto do Coração (InCor), University of São Paulo, School of Medicine, São Paulo, Brazil
- Division of Clinical Immunology and Allergy, University of São Paulo, School of Medicine, São Paulo, Brazil
- Instituto Nacional de Ciência e Tecnologia, INCT, III- Institute for Investigation in Immunology, São Paulo, Brazil
| | - Priscila Camillo Teixeira
- Laboratory of Immunology, Heart Institute Instituto do Coração (InCor), University of São Paulo, School of Medicine, São Paulo, Brazil
- Division of Clinical Immunology and Allergy, University of São Paulo, School of Medicine, São Paulo, Brazil
- Instituto Nacional de Ciência e Tecnologia, INCT, III- Institute for Investigation in Immunology, São Paulo, Brazil
| | - Charles Mady
- Myocardiopathies and Aortic Diseases Unit, Heart Institute Instituto do Coração (InCor), School of Medicine, University of São Paulo, São Paulo, Brazil
| | - Ludmila Rodrigues Pinto Ferreira
- RNA Systems Biology Laboratory (RSBL), Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Quentin Ferré
- Institut National de la Santé Et de la Recherche Médicale (INSERM), Unité Mixte de Recherche (UMR)_1090, Aix Marseille Université, TAGC Theories and Approaches of Genomic Complexity, Institut MarMaRa, Marseille, France
| | - Ronaldo Honorato Barros Santos
- Division of Transplantation, Heart Institute Instituto do Coração (InCor), University of São Paulo, School of Medicine, São Paulo, Brazil
| | - Andreia Kuramoto
- Laboratory of Immunology, Heart Institute Instituto do Coração (InCor), University of São Paulo, School of Medicine, São Paulo, Brazil
| | - Sandrine Cabantous
- Aix Marseille Université, Génétique et Immunologie des Maladies Parasitaires, Inserm, UMR_906, Marseille, France
| | - Samuel Steffen
- Division of Transplantation, Heart Institute Instituto do Coração (InCor), University of São Paulo, School of Medicine, São Paulo, Brazil
- Division of Surgery, Heart Institute Instituto do Coração (InCor), University of São Paulo, School of Medicine, São Paulo, Brazil
| | - Antonio Noedir Stolf
- Division of Surgery, Heart Institute Instituto do Coração (InCor), University of São Paulo, School of Medicine, São Paulo, Brazil
| | - Pablo Pomerantzeff
- Heart Institute (InCor), School of Medicine, University of São Paulo, São Paulo, São Paulo, Brazil
| | - Alfredo Inacio Fiorelli
- Division of Surgery, Heart Institute Instituto do Coração (InCor), University of São Paulo, School of Medicine, São Paulo, Brazil
| | - Edimar Alcides Bocchi
- Division of Surgery, Heart Institute Instituto do Coração (InCor), University of São Paulo, School of Medicine, São Paulo, Brazil
| | - Cristina Wide Pissetti
- Laboratory of Immunology, Universidade Federal Do Triângulo Mineiro (UFTM), Uberaba, Brazil
| | - Bruno Saba
- Laboratório de Investigação Molecular em Cardiologia, Instituto de Cardiologia Dante Pazzanese (IDPC), São Paulo, Brazil
| | - Darlan da Silva Cândido
- Laboratory of Immunology, Heart Institute Instituto do Coração (InCor), University of São Paulo, School of Medicine, São Paulo, Brazil
- Division of Clinical Immunology and Allergy, University of São Paulo, School of Medicine, São Paulo, Brazil
- Instituto Nacional de Ciência e Tecnologia, INCT, III- Institute for Investigation in Immunology, São Paulo, Brazil
| | - Fabrício C. Dias
- School of Medicine of Ribeirão Preto Faculdade de Medicina de Ribeirão Preto (FMRP), University of São Paulo, Ribeirão Preto, Brazil
| | - Marcelo Ferraz Sampaio
- Laboratório de Investigação Molecular em Cardiologia, Instituto de Cardiologia Dante Pazzanese (IDPC), São Paulo, Brazil
| | - Fabio Antônio Gaiotto
- Division of Transplantation, Heart Institute Instituto do Coração (InCor), University of São Paulo, School of Medicine, São Paulo, Brazil
- Division of Surgery, Heart Institute Instituto do Coração (InCor), University of São Paulo, School of Medicine, São Paulo, Brazil
| | - José Antonio Marin-Neto
- School of Medicine of Ribeirão Preto Faculdade de Medicina de Ribeirão Preto (FMRP), University of São Paulo, Ribeirão Preto, Brazil
| | - Abílio Fragata
- Laboratório de Investigação Molecular em Cardiologia, Instituto de Cardiologia Dante Pazzanese (IDPC), São Paulo, Brazil
| | - Ricardo Costa Fernandes Zaniratto
- Laboratory of Immunology, Heart Institute Instituto do Coração (InCor), University of São Paulo, School of Medicine, São Paulo, Brazil
| | - Sergio Siqueira
- Pacemaker Clinic, Heart Institute Instituto do Coração (InCor), School of Medicine, University of São Paulo, São Paulo, Brazil
| | - Giselle De Lima Peixoto
- Pacemaker Clinic, Heart Institute Instituto do Coração (InCor), School of Medicine, University of São Paulo, São Paulo, Brazil
| | - Vagner Oliveira-Carvalho Rigaud
- Laboratory of Immunology, Heart Institute Instituto do Coração (InCor), University of São Paulo, School of Medicine, São Paulo, Brazil
- Heart Failure Unit, Heart Institute Instituto do Coração (InCor) School of Medicine, University of Sao Paulo, Sao Paulo, Brazil
| | - Fernando Bacal
- Division of Transplantation, Heart Institute Instituto do Coração (InCor), University of São Paulo, School of Medicine, São Paulo, Brazil
| | - Paula Buck
- Heart Institute (InCor), School of Medicine, University of São Paulo, São Paulo, São Paulo, Brazil
| | - Rafael Ribeiro Almeida
- Laboratory of Immunology, Heart Institute Instituto do Coração (InCor), University of São Paulo, School of Medicine, São Paulo, Brazil
- Division of Clinical Immunology and Allergy, University of São Paulo, School of Medicine, São Paulo, Brazil
- Instituto Nacional de Ciência e Tecnologia, INCT, III- Institute for Investigation in Immunology, São Paulo, Brazil
| | - Hui Tzu Lin-Wang
- Laboratório de Investigação Molecular em Cardiologia, Instituto de Cardiologia Dante Pazzanese (IDPC), São Paulo, Brazil
| | - André Schmidt
- School of Medicine of Ribeirão Preto Faculdade de Medicina de Ribeirão Preto (FMRP), University of São Paulo, Ribeirão Preto, Brazil
| | - Martino Martinelli
- Pacemaker Clinic, Heart Institute Instituto do Coração (InCor), School of Medicine, University of São Paulo, São Paulo, Brazil
| | - Mario Hiroyuki Hirata
- Department of Clinical and Toxicological Analyses, Faculty of Pharmaceutical Sciences, University of São Paulo (USP), São Paulo, Brazil
| | - Eduardo Antonio Donadi
- School of Medicine of Ribeirão Preto Faculdade de Medicina de Ribeirão Preto (FMRP), University of São Paulo, Ribeirão Preto, Brazil
| | - Alexandre Costa Pereira
- Heart Institute (InCor), School of Medicine, University of São Paulo, São Paulo, São Paulo, Brazil
| | | | - Denis Puthier
- Institut National de la Santé Et de la Recherche Médicale (INSERM), Unité Mixte de Recherche (UMR)_1090, Aix Marseille Université, TAGC Theories and Approaches of Genomic Complexity, Institut MarMaRa, Marseille, France
| | - Jorge Kalil
- Laboratory of Immunology, Heart Institute Instituto do Coração (InCor), University of São Paulo, School of Medicine, São Paulo, Brazil
- Division of Clinical Immunology and Allergy, University of São Paulo, School of Medicine, São Paulo, Brazil
- Instituto Nacional de Ciência e Tecnologia, INCT, III- Institute for Investigation in Immunology, São Paulo, Brazil
| | - Lionel Spinelli
- Institut National de la Santé Et de la Recherche Médicale (INSERM), Unité Mixte de Recherche (UMR)_1090, Aix Marseille Université, TAGC Theories and Approaches of Genomic Complexity, Institut MarMaRa, Marseille, France
- Aix Marseille Université, CNRS, INSERM, Centre d'Immunologie de Marseille-Luminy, Marseille, France
- *Correspondence: Christophe Chevillard, ; Edecio Cunha-Neto, ; Lionel Spinelli,
| | - Edecio Cunha-Neto
- Laboratory of Immunology, Heart Institute Instituto do Coração (InCor), University of São Paulo, School of Medicine, São Paulo, Brazil
- Division of Clinical Immunology and Allergy, University of São Paulo, School of Medicine, São Paulo, Brazil
- Instituto Nacional de Ciência e Tecnologia, INCT, III- Institute for Investigation in Immunology, São Paulo, Brazil
- *Correspondence: Christophe Chevillard, ; Edecio Cunha-Neto, ; Lionel Spinelli,
| | - Christophe Chevillard
- Institut National de la Santé Et de la Recherche Médicale (INSERM), Unité Mixte de Recherche (UMR)_1090, Aix Marseille Université, TAGC Theories and Approaches of Genomic Complexity, Institut MarMaRa, Marseille, France
- *Correspondence: Christophe Chevillard, ; Edecio Cunha-Neto, ; Lionel Spinelli,
| |
Collapse
|
15
|
Transcriptomic and Lipidomic Mapping of Macrophages in the Hub of Chronic Beta-Adrenergic-Stimulation Unravels Hypertrophy-, Proliferation-, and Lipid Metabolism-Related Genes as Novel Potential Markers of Early Hypertrophy or Heart Failure. Biomedicines 2022; 10:biomedicines10020221. [PMID: 35203431 PMCID: PMC8869621 DOI: 10.3390/biomedicines10020221] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 01/13/2022] [Accepted: 01/18/2022] [Indexed: 02/05/2023] Open
Abstract
Sympathetic nervous system overdrive with chronic release of catecholamines is the most important neurohormonal mechanism activated to maintain cardiac output in response to heart stress. Beta-adrenergic signaling behaves first as a compensatory pathway improving cardiac contractility and maladaptive remodeling but becomes dysfunctional leading to pathological hypertrophy and heart failure (HF). Cardiac remodeling is a complex inflammatory syndrome where macrophages play a determinant role. This study aimed at characterizing the temporal transcriptomic evolution of cardiac macrophages in mice subjected to beta-adrenergic-stimulation using RNA sequencing. Owing to a comprehensive bibliographic analysis and complementary lipidomic experiments, this study deciphers typical gene profiles in early compensated hypertrophy (ECH) versus late dilated remodeling related to HF. We uncover cardiac hypertrophy- and proliferation-related transcription programs typical of ECH or HF macrophages and identify lipid metabolism-associated and Na+ or K+ channel-related genes as markers of ECH and HF macrophages, respectively. In addition, our results substantiate the key time-dependent role of inflammatory, metabolic, and functional gene regulation in macrophages during beta-adrenergic dependent remodeling. This study provides important and novel knowledge to better understand the prevalent key role of resident macrophages in response to chronically activated beta-adrenergic signaling, an effective diagnostic and therapeutic target in failing hearts.
Collapse
|
16
|
Lu Q, Zhang RC, Chen SP, Li T, Wang Y, Xue YB, Liu J, Han X, Su YD, Bai L, Du XJ, Ma AQ. The Diagnostic and Prognostic Value of Plasma Galectin 3 in HFrEF Related to the Etiology of Heart Failure. Front Cardiovasc Med 2022; 8:748875. [PMID: 35004876 PMCID: PMC8727364 DOI: 10.3389/fcvm.2021.748875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Accepted: 11/25/2021] [Indexed: 11/17/2022] Open
Abstract
Aim: The aim of present study is to evaluate the diagnostic and prognostic value of plasma galectin 3 (Gal-3) for HF originating from different causes. Methods: We investigated the plasma levels and expression of Gal-3 in cardiac tissues in two transgenic (TG) strains of mice with cardiomyocyte-restricted overexpression of either β2- adrenergic receptor (β2- AR TG) or Mammalian sterile 20-like kinase 1 (Mst1-TG) in the present study. Additionally, 166 patients suffering from heart failure with reduced ejection fraction (HFrEF) in two hospitals within the Shaanxi province were examined in this study. All these patients were treated according to the Chinese HF guidelines of 2014; subsequently, they were followed up for 50 months, and we analyzed the prediction value of baseline Gal-3 to endpoints in these patients. Results: Gal-3 was localized in the cytoplasm and nucleus of cardiomyocytes, often formed aggregates in Mst1-TG mice. Extracellular Gal-3 staining was uncommon in Mst1-TG hearts. However, in β2-AR TG mice, although Gal-3 was also expressed in myocardial cells, it was more highly expressed in interstitial cells (e.g., fibroblasts and macrophages). Plasma Gal-3 was comparable between nTG and Mst1-TG mice. However, plasma Gal-3 was higher in β2-AR TG mice than in nTG mice. In the cohort of HFrEF patients, the median plasma Gal-3 concentration was 158.42 pg/mL. All participants were divided into two groups according to Gal-3 levels. Patients with Gal-3 concentrations above the median were older, and had lower plasma hemoglobin, but higher plasma creatinine, tissue inhibitor of metalloproteinases 1 (TIMP-1), left ventricular end systolic diameter (LVESD), left ventricular end-systolic volumes (LVESV) and end-diastolic, as well as left ventricular end-diastolic volumes (LVEDV). Spearman correlation analysis revealed that Gal-3 was positively correlated with TIMP-1 (r = 0.396, P < 0.001), LVESV (r = 0.181, P = 0.020) and LVEDV (r = 0.190, P = 0.015). The 50-month clinical follow-up revealed 43 deaths, 97 unplanned re-hospitalizations, and 111 composite endpoint events. Cox analysis demonstrated that although Gal-3 did not provide any prognostic value in either total-HF subjects or coronary-heart-disease (CHD) patients, it did provide prognostic value in non-CHD patients. Conclusion: Although plasma Gal-3 is associated with TIMP-1 and echocardiographic parameters, the diagnostic and prognostic value of Gal-3 in HFrEF is determined by the etiology of HF.
Collapse
Affiliation(s)
- Qun Lu
- Department of Cardiovascular Medicine, First Affiliated Hospital, School of Medicine of Xi'an Jiaotong University, Xi'an, China
| | - Ruo-Chen Zhang
- Department of Cardiovascular Medicine, First Affiliated Hospital, School of Medicine of Xi'an Jiaotong University, Xi'an, China
| | - Shu-Ping Chen
- Department of Cardiovascular Medicine, First Affiliated Hospital, School of Medicine of Xi'an Jiaotong University, Xi'an, China
| | - Tao Li
- Department of Cardiovascular Medicine, First Affiliated Hospital, School of Medicine of Xi'an Jiaotong University, Xi'an, China.,Department of Cardiovascular Medicine, Xi'an Central Hospital, Xi'an, China
| | - Ya Wang
- Department of Cardiovascular Medicine, First Affiliated Hospital, School of Medicine of Xi'an Jiaotong University, Xi'an, China
| | - Yan-Bo Xue
- Department of Cardiovascular Medicine, First Affiliated Hospital, School of Medicine of Xi'an Jiaotong University, Xi'an, China
| | - Jing Liu
- Department of Cardiovascular Medicine, First Affiliated Hospital, School of Medicine of Xi'an Jiaotong University, Xi'an, China
| | - Xiu Han
- Department of Cardiovascular Medicine, First Affiliated Hospital, School of Medicine of Xi'an Jiaotong University, Xi'an, China
| | - Yi-Dan Su
- Experimental Cardiology Lab, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Ling Bai
- Department of Cardiovascular Medicine, First Affiliated Hospital, School of Medicine of Xi'an Jiaotong University, Xi'an, China
| | - Xiao-Jun Du
- Experimental Cardiology Lab, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia.,Department of Physiology and Pathophysiology, Medical College of Xi'an Jiaotong University, Xi'an, China
| | - Ai-Qun Ma
- Department of Cardiovascular Medicine, First Affiliated Hospital, School of Medicine of Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
17
|
Du X. Sympatho-adrenergic mechanisms in heart failure: new insights into pathophysiology. MEDICAL REVIEW (BERLIN, GERMANY) 2021; 1:47-77. [PMID: 37724075 PMCID: PMC10388789 DOI: 10.1515/mr-2021-0007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Accepted: 06/02/2021] [Indexed: 09/20/2023]
Abstract
The sympathetic nervous system is activated in the setting of heart failure (HF) to compensate for hemodynamic instability. However, acute sympathetic surge or sustained high neuronal firing rates activates β-adrenergic receptor (βAR) signaling contributing to myocardial remodeling, dysfunction and electrical instability. Thus, sympatho-βAR activation is regarded as a hallmark of HF and forms pathophysiological basis for β-blocking therapy. Building upon earlier research findings, studies conducted in the recent decades have significantly advanced our understanding on the sympatho-adrenergic mechanism in HF, which forms the focus of this article. This review notes recent research progress regarding the roles of cardiac β2AR or α1AR in the failing heart, significance of β1AR-autoantibodies, and βAR signaling through G-protein independent signaling pathways. Sympatho-βAR regulation of immune cells or fibroblasts is specifically discussed. On the neuronal aspects, knowledge is assembled on the remodeling of sympathetic nerves of the failing heart, regulation by presynaptic α2AR of NE release, and findings on device-based neuromodulation of the sympathetic nervous system. The review ends with highlighting areas where significant knowledge gaps exist but hold promise for new breakthroughs.
Collapse
Affiliation(s)
- Xiaojun Du
- Faculty of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, 76 West Yanta Road, Xi’an710061, Shaanxi, China
- Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC3004, Australia
| |
Collapse
|