1
|
Shao Q, Wang H, Li S, Zeng M, Zhang S, Yan X. IRF5 Mediates Artery Inflammation in Salt-Sensitive Hypertension by Regulating STAT1 and STAT2 Phosphorylation to Increase ESM1 Transcription: Insights from Bioinformatics and Mechanistic Analysis. Int J Mol Sci 2025; 26:3722. [PMID: 40332339 PMCID: PMC12027925 DOI: 10.3390/ijms26083722] [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: 02/15/2025] [Revised: 04/04/2025] [Accepted: 04/06/2025] [Indexed: 05/08/2025] Open
Abstract
Salt-sensitive hypertension (SSH) is closely associated with arterial inflammation, yet its molecular mechanisms remain unclear. In this study, we utilized deoxycorticosterone acetate (DOCA)-salt-induced hypertensive mice, which exhibited elevated blood pressure and significant arterial inflammation. Single-cell RNA sequencing (scRNA-seq) identified interferon regulatory factor 5 (IRF5) and its downstream targets, signal transducer and activator of transcription (STAT), as key regulators of these inflammatory changes. In vivo, IRF5 levels were significantly elevated in the DOCA group, while STAT1 and STAT2 protein levels were comparable to those in the normal salt group. However, nuclear levels of phosphorylated STAT1 (pSTAT1) and phosphorylated STAT2 (pSTAT2) were markedly higher in the DOCA group. Furthermore, scRNA-seq analysis showed increased IRF5 expression in endothelial cells (ECs) in both human and mouse aorta samples. In vitro, IRF5 knockdown in artery ECs led to a reduction in nuclear pSTAT1 and pSTAT2 expression. These results suggest that IRF5 promotes STAT1 and STAT2 phosphorylation, enabling their nuclear translocation. Additionally, RNA sequencing indicated a positive correlation between endothelial cell-specific molecule 1 (ESM1) and STAT1/STAT2. Using the UCSC and JASPAR databases, we identified multiple binding sites for the STAT1::STAT2 dimer on the ESM1 promoter. Luciferase reporter assays revealed enhanced ESM1 transcription following pSTAT1::pSTAT2 binding, and pinpoint potential binding sites. Chromatin Immunoprecipitation Quantitative PCR (ChIP-qPCR) further confirmed the specific binding sites between the pSTAT1::pSTAT2 dimer and the ESM1 promoter. These findings highlight the critical role of the IRF5-pSTAT1::pSTAT2-ESM1 pathway in the pathogenesis of SSH and suggest potential therapeutic targets.
Collapse
Affiliation(s)
- Qiaoyu Shao
- Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China; (Q.S.); (H.W.); (S.L.); (M.Z.)
- Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Hao Wang
- Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China; (Q.S.); (H.W.); (S.L.); (M.Z.)
| | - Shicheng Li
- Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China; (Q.S.); (H.W.); (S.L.); (M.Z.)
| | - Mengying Zeng
- Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China; (Q.S.); (H.W.); (S.L.); (M.Z.)
| | - Shuyang Zhang
- Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China; (Q.S.); (H.W.); (S.L.); (M.Z.)
- Medical Research Center, State Key Laboratory of Complex Severe and Rare Diseases, Chinese Academy of Medical Sciences & Peking Union Medical College Hospital, Beijing 100730, China
| | - Xiaowei Yan
- Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China; (Q.S.); (H.W.); (S.L.); (M.Z.)
| |
Collapse
|
2
|
Li Y, Liao Y, Miao Y, Yu C, Long J, Wu J, Zuo J, Zhang Z, Dou W, Wang X, Zhang B, Yu C, Yang J, Wang S. Interleukin-35 mRNA therapy for influenza virus-induced pneumonia in mice. Eur J Pharmacol 2025; 993:177366. [PMID: 39947345 DOI: 10.1016/j.ejphar.2025.177366] [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: 11/09/2024] [Revised: 01/27/2025] [Accepted: 02/10/2025] [Indexed: 02/18/2025]
Abstract
Influenza virus-induced pneumonia is a common complication caused by influenza A virus infection and causes severe lung inflammation. After infection, the body induces an active immune response that can produce cytokine storm, leading to increased expression of pro-inflammatory factors and tissue damage. Interleukin-35 (IL-35) is a recently identified cytokine associated with viral infection. IL-35 may inhibit the inflammation caused by viral infection and therefore may be developed into an antiviral treatment. Compared with traditional drugs, mRNA drugs have the advantages of simple production process, short development cycle, strong target specificity, high safety, and long-lasting action. In this study,we prepared IL-35 mRNA and IL-35 mRNA/Lipid Nanoparticle (IL-35 mRNA/LNP). To investigate the role of IL-35 mRNA in the host defense against post-influenza pneumonia, a mouse model of pneumonia caused by influenza infection was established. After influenza infection, the mice produced a large number of inflammatory factors that caused lung tissue damage, while administration of IL-35 mRNA/LNP effectively reduced the inflammatory response and improved the survival rate of mice. In addition, mice injected with IL-35 mRNA/LNP (125 μg/kg) directly via tail vein did not show significant inflammatory responses or tissue damage. These data suggest that IL-35 mRNA attenuates the inflammatory response caused by influenza virus infection and shows potential for development as a new drug for the treatment of influenza virus-induced pneumonia.
Collapse
Affiliation(s)
- Yanyan Li
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, 421001, PR China; Bioinformatics Center of AMMS, Beijing, 100850, PR China
| | - Yuqin Liao
- Bioinformatics Center of AMMS, Beijing, 100850, PR China
| | - Yiqi Miao
- Bioinformatics Center of AMMS, Beijing, 100850, PR China
| | - Changxiao Yu
- Bioinformatics Center of AMMS, Beijing, 100850, PR China
| | - Jinrong Long
- Bioinformatics Center of AMMS, Beijing, 100850, PR China
| | - Jiayu Wu
- Bioinformatics Center of AMMS, Beijing, 100850, PR China; Department of Pharmacy, Peking Union Medical College Hospital (Dongdan Campus), Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, PR China
| | - Jun Zuo
- Bioinformatics Center of AMMS, Beijing, 100850, PR China
| | - Zhen Zhang
- Bioinformatics Center of AMMS, Beijing, 100850, PR China
| | - Wei Dou
- Bioinformatics Center of AMMS, Beijing, 100850, PR China; Department of Pharmacy, Peking Union Medical College Hospital (Dongdan Campus), Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, PR China
| | - Xin Wang
- Bioinformatics Center of AMMS, Beijing, 100850, PR China
| | - Bo Zhang
- Department of Pharmacy, Peking Union Medical College Hospital (Dongdan Campus), Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, PR China
| | - Cuiyun Yu
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, 421001, PR China.
| | - Jing Yang
- Bioinformatics Center of AMMS, Beijing, 100850, PR China.
| | - Shengqi Wang
- Bioinformatics Center of AMMS, Beijing, 100850, PR China.
| |
Collapse
|
3
|
Zhou M, Xia Y, Wang S. IL35 modulates HBV-related HCC progression via IL6-STAT3 signaling. Sci Rep 2025; 15:6293. [PMID: 39984567 PMCID: PMC11845586 DOI: 10.1038/s41598-025-89015-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2024] [Accepted: 02/03/2025] [Indexed: 02/23/2025] Open
Abstract
Hepatocellular carcinoma (HCC) is a common malignant tumor with incidences reported worldwide along with high mortality rates. It is a significant public health concern as hepatitis B virus (HBV) infection is the leading cause of HCC. IL35, a novel heterodimeric cytokine belonging to the IL-12 family, comprises two subunits, namely IL-12p35 and Epstein-Barr virus-induced gene 3 (EBI3). They are crucial in regulating immune responses to tumors and infectious diseases. However, their function in HBV-related HCC is unclear. The objective of this study was to identify the regulatory role of IL35 in the occurrence of HBV-related HCC and its underlying molecular mechanisms. The expression of IL35 was enhanced in human HBV-related HCC tissues. HBV induction, particularly HBx, enhanced the expression of IL-35 in hepatoma cell lines. Silencing IL-35 promoted apoptosis and suppressed proliferation, cell cycle progression, migration, and invasion of HBx-induced hepatoma cells. Mechanistically, silencing IL-35 effectively inhibited the activation of the IL-6-STAT3 signaling pathway by suppressing the expression of IL-6 and nuclear import and phosphorylation of STAT3 in HBx-induced hepatoma cells. Therefore, inhibiting the IL6-STAT3 signaling pathway by silencing IL35 effectively alleviated the progression of HBV-related HCC. IL35 is a potential target for treating HBV-related HCC.
Collapse
Affiliation(s)
- Mingran Zhou
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230002, China
- Anhui Public Health Clinical Center, Hefei, 230002, China
| | - Yunhong Xia
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230002, China
- Anhui Public Health Clinical Center, Hefei, 230002, China
| | - Shuomin Wang
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230002, China.
- Anhui Public Health Clinical Center, Hefei, 230002, China.
| |
Collapse
|
4
|
Zhang J, Ding W, Yin Z, Liu S, Zhao M, Xu Y, Liu J, Pan W, Peng S, Wei C, Zheng Z, Qin JJ, Wan J, Wang M. Interleukin-12p40 deficiency attenuates myocardial ferroptosis in doxorubicin-induced chronic cardiomyopathy by inhibiting Th17 differentiation and interleukin-17A production. Cardiovasc Res 2024; 120:2117-2133. [PMID: 39298642 DOI: 10.1093/cvr/cvae208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 05/07/2024] [Accepted: 08/06/2024] [Indexed: 09/22/2024] Open
Abstract
AIMS Interleukin (IL)-12p40 is a common subunit of the bioactive cytokines IL-12 and IL-23, and it also has its own intrinsic functional activity. However, its role in doxorubicin-induced chronic cardiomyopathy (DICCM) as well as the underlying mechanisms are still unknown. METHODS AND RESULTS In this study, we used IL-12p40-knockout mice, IL-23p19-knockout mice, Rag1-knockout mice, a ferroptosis inhibitor, recombinant IL-12 (rIL-12), rIL-23, rIL-12p40, rIL-12p80, and anti-IL17A to investigate the effects of IL-12p40 on DICCM and elucidate the underlying mechanisms. We found that myocardial ferroptosis were increased in DICCM and that the inhibition of ferroptosis protected against DICCM. The expression of IL-12p40 was upregulated, and IL-12p40 was predominantly expressed by CD4+ T cells in the hearts of mice with DICCM. IL-12p40 knockout attenuated cardiac dysfunction, fibrosis and ferroptosis in DICCM, and similar results were observed in the context of CD4+ T cell IL-12p40 deficiency in Rag1-/- mice. Treatment with rIL-23, but not rIL-12, rIL-12p40 monomer or rIL-12p80, abolished the protective effects of IL-12p40 knockout. Moreover, rIL-23 treatment and IL-23p19 knockout exacerbated and ameliorated DICCM, respectively. IL-12p40 knockout might protect against DICCM by inhibiting Th17 differentiation and IL-17A production but not Th1, Th2 and Treg differentiation. Neutralizing IL-17A with an antibody also attenuated cardiac dysfunction, fibrosis, and ferroptosis. The IL-12p40/Th17/IL-17A axis might promote cardiomyocyte ferroptosis by activating TNF receptor-associated factor 6 (TRAF6)/mitogen-activated protein kinase (MAPK)/P53 signalling in DICCM. CONCLUSION Interleukin-12p40 deficiency protects against DICCM by inhibiting Th17 differentiation and the production of IL-17A, which plays critical roles in cardiomyocyte ferroptosis in DICCM via activating TRAF6/MAPK/P53 signalling. Our study may provide novel insights for the identification of therapeutic targets for treating DICCM in the clinic.
Collapse
Affiliation(s)
- Jishou Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan 430060, China
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Wen Ding
- Department of Cardiology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan 430060, China
- Department of Radiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Wuhan, China
| | - Zheng Yin
- Department of Cardiology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan 430060, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Siqi Liu
- Department of Cardiology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan 430060, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Mengmeng Zhao
- Department of Cardiology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan 430060, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Yao Xu
- Department of Cardiology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan 430060, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Jianfang Liu
- Department of Cardiology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan 430060, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Wei Pan
- Department of Cardiology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan 430060, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Shanshan Peng
- Department of Cardiology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan 430060, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Cheng Wei
- Department of Cardiology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan 430060, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Zihui Zheng
- Department of Cardiology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan 430060, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Juan-Juan Qin
- Department of Cardiology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan 430060, China
- Department of Geriatrics, Zhongnan Hospital of Wuhan University, Wuhan, China
- Center for Healthy Aging, Wuhan University School of Nursing, Wuhan, China
| | - Jun Wan
- Department of Cardiology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan 430060, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Menglong Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan 430060, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| |
Collapse
|
5
|
Kopaliani I, Elsaid B, Speier S, Deussen A. Immune and Metabolic Mechanisms of Endothelial Dysfunction. Int J Mol Sci 2024; 25:13337. [PMID: 39769104 PMCID: PMC11728141 DOI: 10.3390/ijms252413337] [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: 08/30/2024] [Revised: 12/01/2024] [Accepted: 12/06/2024] [Indexed: 01/16/2025] Open
Abstract
Endothelial dysfunction is a strong prognostic factor in predicting the development of cardiovascular diseases. Dysfunctional endothelium loses its homeostatic ability to regulate vascular tone and prevent overactivation of inflammation, leading to vascular dysfunction. These functions are critical for vascular homeostasis and arterial pressure control, the disruption of which may lead to hypertension. Hypertension itself can also cause endothelial dysfunction, as endothelial cells are susceptible to haemodynamic changes. Although it is unclear which of those factors appear first, they create a vicious circle further damaging multiple organs, including the heart and vessels. There are also sex-specific differences in homeostatic functions of the endothelium regarding vessel tone regulation, which may contribute to differences in arterial blood pressure between men and women. Even more importantly, there are sex-differences in the development of endothelial dysfunction and vessel remodelling. Hence, an understanding of the mechanisms of endothelial dysfunction and its contribution to pathological vascular remodelling during hypertension is of critical importance. This review addresses immunological and metabolic aspects in mechanisms of endothelial dysfunction and the resulting mechanisms in vascular remodelling with respect to arterial hypertension, including the potential role of sex-specific differences.
Collapse
Affiliation(s)
- Irakli Kopaliani
- Institute of Physiology, Medical Faculty Carl Gustav Carus, Technische Universität Dresden, 01037 Dresden, Germany; (B.E.); (S.S.); (A.D.)
| | - Basant Elsaid
- Institute of Physiology, Medical Faculty Carl Gustav Carus, Technische Universität Dresden, 01037 Dresden, Germany; (B.E.); (S.S.); (A.D.)
- Department of Physiology, Faculty of Medicine, Ain Shams University, Cairo 1181, Egypt
| | - Stephan Speier
- Institute of Physiology, Medical Faculty Carl Gustav Carus, Technische Universität Dresden, 01037 Dresden, Germany; (B.E.); (S.S.); (A.D.)
- Paul Langerhans Institute Dresden of the Helmholtz Zentrum München at the University Clinic Carl Gustav Carus of Technische Universität Dresden, Helmholtz Zentrum München, 85764 Neuherberg, Germany
| | - Andreas Deussen
- Institute of Physiology, Medical Faculty Carl Gustav Carus, Technische Universität Dresden, 01037 Dresden, Germany; (B.E.); (S.S.); (A.D.)
| |
Collapse
|
6
|
Zhang J, Liu S, Ding W, Wan J, Qin JJ, Wang M. Resolution of inflammation, an active process to restore the immune microenvironment balance: A novel drug target for treating arterial hypertension. Ageing Res Rev 2024; 99:102352. [PMID: 38857706 DOI: 10.1016/j.arr.2024.102352] [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: 11/24/2023] [Revised: 05/11/2024] [Accepted: 05/27/2024] [Indexed: 06/12/2024]
Abstract
The resolution of inflammation, the other side of the inflammatory response, is defined as an active and highly coordinated process that promotes the restoration of immune microenvironment balance and tissue repair. Inflammation resolution involves several key processes, including dampening proinflammatory signaling, specialized proresolving lipid mediator (SPM) production, nonlipid proresolving mediator production, efferocytosis and regulatory T-cell (Treg) induction. In recent years, increasing attention has been given to the effects of inflammation resolution on hypertension. Furthermore, our previous studies reported the antihypertensive effects of SPMs. Therefore, in this review, we aim to summarize and discuss the detailed association between arterial hypertension and inflammation resolution. Additional, the association between gut microbe-mediated immune and hypertension is discussed. This findings suggested that accelerating the resolution of inflammation can have beneficial effects on hypertension and its related organ damage. Exploring novel drug targets by focusing on various pathways involved in accelerating inflammation resolution will contribute to the treatment and control of hypertensive diseases in the future.
Collapse
Affiliation(s)
- Jishou Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University, Department of Geriatrics, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China; Department of Cardiology, Renmin Hospital of Wuhan University; Cardiovascular Research Institute, Wuhan University; Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Siqi Liu
- Department of Cardiology, Renmin Hospital of Wuhan University, Department of Geriatrics, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China; Department of Cardiology, Renmin Hospital of Wuhan University; Cardiovascular Research Institute, Wuhan University; Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Wen Ding
- Department of Cardiology, Renmin Hospital of Wuhan University; Cardiovascular Research Institute, Wuhan University; Hubei Key Laboratory of Cardiology, Wuhan, China; Department of Radiology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jun Wan
- Department of Cardiology, Renmin Hospital of Wuhan University, Department of Geriatrics, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China; Department of Cardiology, Renmin Hospital of Wuhan University; Cardiovascular Research Institute, Wuhan University; Hubei Key Laboratory of Cardiology, Wuhan, China.
| | - Juan-Juan Qin
- Department of Cardiology, Renmin Hospital of Wuhan University, Department of Geriatrics, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China; Center for Healthy Aging, Wuhan University School of Nursing, Wuhan, China.
| | - Menglong Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Department of Geriatrics, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China; Department of Cardiology, Renmin Hospital of Wuhan University; Cardiovascular Research Institute, Wuhan University; Hubei Key Laboratory of Cardiology, Wuhan, China.
| |
Collapse
|
7
|
Pan H, Ji Q, Zhao M, Zheng Z, Lu X, Feng Y, Gan L, Ye J, Wan J, Ye D. IL-12p40 deletion reduces M1 macrophage polarization and alleviates cardiac remodeling via regulating Th17 cells differentiation, but not γδT 17 cells, in TAC mice. Eur J Pharmacol 2024; 974:176602. [PMID: 38677538 DOI: 10.1016/j.ejphar.2024.176602] [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: 11/03/2023] [Revised: 04/16/2024] [Accepted: 04/18/2024] [Indexed: 04/29/2024]
Abstract
BACKGROUND The interleukin (IL) -12 p40 subunit is the common subunit of IL-12 and IL-23. It affects the immune inflammatory response, which may be closely related to cardiac remodeling. In this study, the regulatory effect of IL-12p40 knockout (KO) on cardiac remodeling was investigated, and the underlying mechanism was explored. METHODS AND RESULTS Mice were subjected to transverse aortic constriction (TAC) to establish a model of cardiac remodeling. First, IL-12p40 was deleted to observe its effects on cardiac remodeling and cardiac inflammation, and the results showed that IL-12p40 deletion reduced both T helper 17 (Th17) and γδT17 cell differentiation, decreased proinflammatory macrophage differentiation, alleviated cardiac remodeling, and relieved cardiac dysfunction in TAC mice. Next, we explored whether IL-17 regulated TAC-induced cardiac remodeling, and the results showed that IL-17 neutralization alleviated proinflammatory macrophage differentiation and cardiac remodeling in IL-12p40 knockout mice and WT mice. Neutralization with cluster of differentiation 4 receptor (CD4) and γδ T-cell receptor (γδTCR) antibodies inhibited pro-inflammatory macrophage polarization and improved cardiac remodeling, and CD4 neutralizing antibody (NAb) had more significant effects. Finally, adoptive transfer of Th17 cells aggravated proinflammatory macrophage differentiation and cardiac remodeling in TAC-treated CD4 KO mice, while neutralization with the IL-12p40 antibody alleviated these pathological changes. CONCLUSION Mainly Th17 cells but not γδT17 cells secrete IL-17, which mediates IL-12p40, promotes the polarization of proinflammatory macrophages, and exacerbates cardiac remodeling in TAC mice. IL-12p40 may be a potential target for the prevention and treatment of cardiac remodeling.
Collapse
Affiliation(s)
- Heng Pan
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, China; Cardiovascular Research Institute, Wuhan University, Wuhan, 430060, China; Hubei Key Laboratory of Cardiology, Wuhan, 430060, China
| | - Qingwei Ji
- People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, 530000, China; Institute of Cardiovascular Diseases, Guangxi Academy of Medical Sciences, Nanning, 530000, China
| | - Mengmeng Zhao
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, China; Cardiovascular Research Institute, Wuhan University, Wuhan, 430060, China; Hubei Key Laboratory of Cardiology, Wuhan, 430060, China
| | - Zihui Zheng
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, China; Cardiovascular Research Institute, Wuhan University, Wuhan, 430060, China; Hubei Key Laboratory of Cardiology, Wuhan, 430060, China
| | - Xiyi Lu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, China; Cardiovascular Research Institute, Wuhan University, Wuhan, 430060, China; Hubei Key Laboratory of Cardiology, Wuhan, 430060, China
| | - Yongqi Feng
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, China; Cardiovascular Research Institute, Wuhan University, Wuhan, 430060, China; Hubei Key Laboratory of Cardiology, Wuhan, 430060, China
| | - Liren Gan
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, China; Cardiovascular Research Institute, Wuhan University, Wuhan, 430060, China; Hubei Key Laboratory of Cardiology, Wuhan, 430060, China
| | - Jing Ye
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, China; Cardiovascular Research Institute, Wuhan University, Wuhan, 430060, China; Hubei Key Laboratory of Cardiology, Wuhan, 430060, China.
| | - Jun Wan
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, China; Cardiovascular Research Institute, Wuhan University, Wuhan, 430060, China; Hubei Key Laboratory of Cardiology, Wuhan, 430060, China.
| | - Di Ye
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, China; Cardiovascular Research Institute, Wuhan University, Wuhan, 430060, China; Hubei Key Laboratory of Cardiology, Wuhan, 430060, China.
| |
Collapse
|
8
|
Shen C, Wu N, Chen X, Peng J, Feng M, Wang J, Yu Y. Interleukin-5 alleviates cardiac remodelling via the STAT3 pathway in angiotensin II-infused mice. J Cell Mol Med 2024; 28:e18493. [PMID: 38963241 PMCID: PMC11223166 DOI: 10.1111/jcmm.18493] [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: 08/14/2023] [Revised: 06/02/2024] [Accepted: 06/05/2024] [Indexed: 07/05/2024] Open
Abstract
Interleukin-5 (IL-5) has been reported to be involved in cardiovascular diseases, such as atherosclerosis and cardiac injury. This study aimed to investigate the effects of IL-5 on cardiac remodelling. Mice were infused with angiotensin II (Ang II), and the expression and source of cardiac IL-5 were analysed. The results showed that cardiac IL-5 expression was time- and dose-dependently decreased after Ang II infusion, and was mainly derived from cardiac macrophages. Additionally, IL-5-knockout (IL-5-/-) mice were used to observe the effects of IL-5 knockout on Ang II-induced cardiac remodelling. We found knockout of IL-5 significantly increased the expression of cardiac hypertrophy markers, elevated myocardial cell cross-sectional areas and worsened cardiac dysfunction in Ang II-infused mice. IL-5 deletion also promoted M2 macrophage differentiation and exacerbated cardiac fibrosis. Furthermore, the effects of IL-5 deletion on cardiac remodelling was detected after the STAT3 pathway was inhibited by S31-201. The effects of IL-5 on cardiac remodelling and M2 macrophage differentiation were reversed by S31-201. Finally, the effects of IL-5 on macrophage differentiation and macrophage-related cardiac hypertrophy and fibrosis were analysed in vitro. IL-5 knockout significantly increased the Ang II-induced mRNA expression of cardiac hypertrophy markers in myocardial cells that were co-cultured with macrophages, and this effect was reversed by S31-201. Similar trends in the mRNA levels of fibrosis markers were observed when cardiac fibroblasts and macrophages were co-cultured. In conclusions, IL-5 deficiency promote the differentiation of M2 macrophages by activating the STAT3 pathway, thereby exacerbating cardiac remodelling in Ang II-infused mice. IL-5 may be a potential target for the clinical prevention of cardiac remodelling.
Collapse
Affiliation(s)
- Caijie Shen
- Department of Cardiovascular MedicineThe First Affiliated Hospital of Ningbo UniversityNingboChina
| | - Nan Wu
- Department of Cardiovascular MedicineThe First Affiliated Hospital of Ningbo UniversityNingboChina
| | - Xiaomin Chen
- Department of Cardiovascular MedicineThe First Affiliated Hospital of Ningbo UniversityNingboChina
| | - Jianye Peng
- Cardiovascular MedicineThe Second Affiliated Hospital of University of South ChinaHengyangChina
| | - Mingjun Feng
- Department of Cardiovascular MedicineThe First Affiliated Hospital of Ningbo UniversityNingboChina
| | - Jian Wang
- Department of Cardiovascular MedicineThe First Affiliated Hospital of Ningbo UniversityNingboChina
| | - Yibo Yu
- Department of Cardiovascular MedicineThe First Affiliated Hospital of Ningbo UniversityNingboChina
| |
Collapse
|
9
|
Okunishi K, Kochi Y, Zhao M, Wang H, Nakagome K, Izumi T. Munc13-4 regulates asthma and obesity in mice by controlling functions of CD11c + antigen-presenting cells. Allergy 2024; 79:1992-1995. [PMID: 38426389 DOI: 10.1111/all.16087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 01/30/2024] [Accepted: 02/20/2024] [Indexed: 03/02/2024]
Affiliation(s)
- Katsuhide Okunishi
- Laboratory of Molecular Endocrinology and Metabolism, Department of Molecular Medicine, Institute for Molecular and Cellular Regulation, Gunma University, Maebashi, Japan
- Laboratory of Endocrine and Metabolic System Regulation, Department of Molecular Medicine, Institute for Molecular and Cellular Regulation, Gunma University, Maebashi, Japan
| | - Yuta Kochi
- Genomic Function and Diversity, Medical Research Institute, Tokyo Medical and Dental University, Tokyo, Japan
| | - Min Zhao
- Laboratory of Molecular Endocrinology and Metabolism, Department of Molecular Medicine, Institute for Molecular and Cellular Regulation, Gunma University, Maebashi, Japan
| | - Hao Wang
- Laboratory of Molecular Endocrinology and Metabolism, Department of Molecular Medicine, Institute for Molecular and Cellular Regulation, Gunma University, Maebashi, Japan
| | - Kazuyuki Nakagome
- Department of Respiratory Medicine, Saitama Medical University, Saitama, Japan
| | - Tetsuro Izumi
- Laboratory of Molecular Endocrinology and Metabolism, Department of Molecular Medicine, Institute for Molecular and Cellular Regulation, Gunma University, Maebashi, Japan
| |
Collapse
|
10
|
Lu X, Ji Q, Pan H, Feng Y, Ye D, Gan L, Wan J, Ye J. IL-23p19 deficiency reduces M1 macrophage polarization and improves stress-induced cardiac remodeling by alleviating macrophage ferroptosis in mice. Biochem Pharmacol 2024; 222:116072. [PMID: 38387530 DOI: 10.1016/j.bcp.2024.116072] [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: 11/01/2023] [Revised: 01/24/2024] [Accepted: 02/19/2024] [Indexed: 02/24/2024]
Abstract
BACKGROUND Interleukin-23p19 (IL-23p19) has been demonstrated to be involved in the occurrence and development of cardiovascular diseases such as myocardial infarction and atherosclerosis. This study aimed to examine whether IL-23p19 regulates cardiac remodeling processes and explore its possible mechanisms. METHODS AND RESULTS Transverse aortic constriction was performed to construct a mouse cardiac remodeling model, and sham surgery was used as a control. The results showed that IL-23p19 expression was increased in the heart after surgery and may be mainly produced by cardiac macrophages. Knockout of IL-23p19 attenuated M1 macrophage polarization, reduced ferroptosis, improved the process of cardiac remodeling and alleviated cardiac dysfunction in TAC mice. Cell culture experiments found that macrophages were the main cause of ferroptosis when phenylephrine (PE) was added, and blocking ferroptosis with ferrostatin-1 (Fer-1), a ferroptosis inhibitor, significantly inhibited M1 macrophage polarization. Treatment with Fer-1 also improved cardiac remodeling and alleviated cardiac dysfunction in IL-23p19-/- mice subjected to TAC surgery. Finally, TAC IL-23p19-/- mice that were administered macrophages isolated from WT mice exhibited an increased proportion of M1 macrophages and aggravated cardiac remodeling, and these effects were reversed when Fer-1 was administered. CONCLUSION Knockout of IL-23p19 may attenuate M1 macrophage polarization to improve the cardiac remodeling process by reducing macrophage ferroptosis, and IL-23p19 may be a potential target for the prevention and treatment of cardiac remodeling.
Collapse
Affiliation(s)
- Xiyi Lu
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute, Wuhan University, Hubei Key Laboratory of Cardiology, Wuhan 430060, China
| | - Qingwei Ji
- Department of Cardiology, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China; Institute of Cardiovascular Diseases, Guangxi Academy of Medical Sciences, Nanning, China
| | - Heng Pan
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute, Wuhan University, Hubei Key Laboratory of Cardiology, Wuhan 430060, China
| | - Yongqi Feng
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute, Wuhan University, Hubei Key Laboratory of Cardiology, Wuhan 430060, China
| | - Di Ye
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute, Wuhan University, Hubei Key Laboratory of Cardiology, Wuhan 430060, China
| | - Liren Gan
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute, Wuhan University, Hubei Key Laboratory of Cardiology, Wuhan 430060, China
| | - Jun Wan
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute, Wuhan University, Hubei Key Laboratory of Cardiology, Wuhan 430060, China.
| | - Jing Ye
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute, Wuhan University, Hubei Key Laboratory of Cardiology, Wuhan 430060, China.
| |
Collapse
|
11
|
Xu Y, Zheng Z, Pan H, Zhao M, Zhang J, Peng S, Liu J, Pan W, Yin Z, Xu S, Wei C, Qin JJ, Lin Y, Wan J, Wang M. Kielin/chordin-like protein deficiency aggravates pressure overload-induced cardiac dysfunction and remodeling via P53/P21/CCNB1 signaling in mice. FASEB J 2024; 38:e23513. [PMID: 38421300 DOI: 10.1096/fj.202301841r] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Revised: 01/07/2024] [Accepted: 02/09/2024] [Indexed: 03/02/2024]
Abstract
Targeting cardiac remodeling is regarded as a key therapeutic strategy for heart failure. Kielin/chordin-like protein (KCP) is a secretory protein with 18 cysteine-rich domains and associated with kidney and liver fibrosis. However, the relationship between KCP and cardiac remodeling remains unclear. Here, we aimed to investigate the role of KCP in cardiac remodeling induced by pressure overload and explore its potential mechanisms. Left ventricular (LV) KCP expression was measured with real-time quantitative PCR, western blotting, and immunofluorescence staining in pressure overload-induced cardiac remodeling in mice. Cardiac function and remodeling were evaluated in wide-type (WT) mice and KCP knockout (KO) mice by echocardiography, which were further confirmed by histological analysis with hematoxylin and eosin and Masson staining. RNA sequence was performed with LV tissue from WT and KO mice to identify differentially expressed genes and related signaling pathways. Primary cardiac fibroblasts (CFs) were used to validate the regulatory role and potential mechanisms of KCP during fibrosis. KCP was down-regulated in the progression of cardiac remodeling induced by pressure overload, and was mainly expressed in fibroblasts. KCP deficiency significantly aggravated pressure overload-induced cardiac dysfunction and remodeling. RNA sequence revealed that the role of KCP deficiency in cardiac remodeling was associated with cell division, cell cycle, and P53 signaling pathway, while cyclin B1 (CCNB1) was the most significantly up-regulated gene. Further investigation in vivo and in vitro suggested that KCP deficiency promoted the proliferation of CFs via P53/P21/CCNB1 pathway. Taken together, these results suggested that KCP deficiency aggravates cardiac dysfunction and remodeling induced by pressure overload via P53/P21/CCNB1 signaling in mice.
Collapse
Affiliation(s)
- Yao Xu
- Department of Cardiology, Renmin Hospital of Wuhan University, Department of Geriatrics, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute, Wuhan University, Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Zihui Zheng
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute, Wuhan University, Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Heng Pan
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute, Wuhan University, Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Mengmeng Zhao
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute, Wuhan University, Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Jishou Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute, Wuhan University, Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Shanshan Peng
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute, Wuhan University, Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Jianfang Liu
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute, Wuhan University, Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Wei Pan
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute, Wuhan University, Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Zheng Yin
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute, Wuhan University, Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Shuwan Xu
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute, Wuhan University, Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Cheng Wei
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute, Wuhan University, Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Juan-Juan Qin
- Department of Cardiology, Renmin Hospital of Wuhan University, Department of Geriatrics, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China
- Center for Healthy Aging, Wuhan University School of Nursing, Wuhan, China
| | - Yingzhong Lin
- Department of Cardiology, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Jun Wan
- Department of Cardiology, Renmin Hospital of Wuhan University, Department of Geriatrics, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute, Wuhan University, Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Menglong Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Department of Geriatrics, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute, Wuhan University, Hubei Key Laboratory of Cardiology, Wuhan, China
| |
Collapse
|
12
|
Yin Z, Zhang J, Zhao M, Peng S, Ye J, Liu J, Xu Y, Xu S, Pan W, Wei C, Qin J, Wan J, Wang M. Maresin-1 ameliorates hypertensive vascular remodeling through its receptor LGR6. MedComm (Beijing) 2024; 5:e491. [PMID: 38463394 PMCID: PMC10924638 DOI: 10.1002/mco2.491] [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: 04/12/2023] [Revised: 12/30/2023] [Accepted: 01/11/2024] [Indexed: 03/12/2024] Open
Abstract
Hypertensive vascular remodeling is defined as the changes in vascular function and structure induced by persistent hypertension. Maresin-1 (MaR1), one of metabolites from Omega-3 fatty acids, has been reported to promote inflammation resolution in several inflammatory diseases. This study aims to investigate the effect of MaR1 on hypertensive vascular remodeling. Here, we found serum MaR1 levels were reduced in hypertensive patients and was negatively correlated with systolic blood pressure (SBP). The treatment of MaR1 reduced the elevation of blood pressure and alleviated vascular remodeling in the angiotensin II (AngII)-infused mouse model. In addition, MaR1-treated vascular smooth muscle cells (VSMCs) exhibited reduced excessive proliferation, migration, and phenotype switching, as well as impaired pyroptosis. However, the knockout of the receptor of MaR1, leucine-rich repeat-containing G protein-coupled receptor 6 (LGR6), was seen to aggravate pathological vascular remodeling, which could not be reversed by additional MaR1 treatment. The mechanisms by which MaR1 regulates vascular remodeling through LGR6 involves the Ca2+/calmodulin-dependent protein kinase II/nuclear factor erythroid 2-related factor 2/heme oxygenase-1 signaling pathway. Overall, supplementing MaR1 may be a novel therapeutic strategy for the prevention and treatment of hypertension.
Collapse
Affiliation(s)
- Zheng Yin
- Department of Cardiology, Renmin Hospital of Wuhan University, Department of Geriatrics, Zhongnan Hospital of Wuhan UniversityWuhan UniversityWuhanChina
- Cardiovascular Research InstituteWuhan UniversityWuhanChina
- Hubei Key Laboratory of CardiologyWuhanChina
| | - Jishou Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University, Department of Geriatrics, Zhongnan Hospital of Wuhan UniversityWuhan UniversityWuhanChina
- Cardiovascular Research InstituteWuhan UniversityWuhanChina
- Hubei Key Laboratory of CardiologyWuhanChina
| | - Mengmeng Zhao
- Department of Cardiology, Renmin Hospital of Wuhan University, Department of Geriatrics, Zhongnan Hospital of Wuhan UniversityWuhan UniversityWuhanChina
- Cardiovascular Research InstituteWuhan UniversityWuhanChina
- Hubei Key Laboratory of CardiologyWuhanChina
| | - Shanshan Peng
- Department of Cardiology, Renmin Hospital of Wuhan University, Department of Geriatrics, Zhongnan Hospital of Wuhan UniversityWuhan UniversityWuhanChina
- Cardiovascular Research InstituteWuhan UniversityWuhanChina
- Hubei Key Laboratory of CardiologyWuhanChina
| | - Jing Ye
- Department of Cardiology, Renmin Hospital of Wuhan University, Department of Geriatrics, Zhongnan Hospital of Wuhan UniversityWuhan UniversityWuhanChina
- Cardiovascular Research InstituteWuhan UniversityWuhanChina
- Hubei Key Laboratory of CardiologyWuhanChina
| | - Jianfang Liu
- Department of Cardiology, Renmin Hospital of Wuhan University, Department of Geriatrics, Zhongnan Hospital of Wuhan UniversityWuhan UniversityWuhanChina
- Cardiovascular Research InstituteWuhan UniversityWuhanChina
- Hubei Key Laboratory of CardiologyWuhanChina
| | - Yao Xu
- Department of Cardiology, Renmin Hospital of Wuhan University, Department of Geriatrics, Zhongnan Hospital of Wuhan UniversityWuhan UniversityWuhanChina
- Cardiovascular Research InstituteWuhan UniversityWuhanChina
- Hubei Key Laboratory of CardiologyWuhanChina
| | - Shuwan Xu
- Department of Cardiology, Renmin Hospital of Wuhan University, Department of Geriatrics, Zhongnan Hospital of Wuhan UniversityWuhan UniversityWuhanChina
- Cardiovascular Research InstituteWuhan UniversityWuhanChina
- Hubei Key Laboratory of CardiologyWuhanChina
| | - Wei Pan
- Department of Cardiology, Renmin Hospital of Wuhan University, Department of Geriatrics, Zhongnan Hospital of Wuhan UniversityWuhan UniversityWuhanChina
- Cardiovascular Research InstituteWuhan UniversityWuhanChina
- Hubei Key Laboratory of CardiologyWuhanChina
| | - Cheng Wei
- Department of Cardiology, Renmin Hospital of Wuhan University, Department of Geriatrics, Zhongnan Hospital of Wuhan UniversityWuhan UniversityWuhanChina
- Cardiovascular Research InstituteWuhan UniversityWuhanChina
- Hubei Key Laboratory of CardiologyWuhanChina
| | - Juan‐Juan Qin
- Department of Cardiology, Renmin Hospital of Wuhan University, Department of Geriatrics, Zhongnan Hospital of Wuhan UniversityWuhan UniversityWuhanChina
- Center for Healthy AgingWuhan University School of NursingWuhanChina
| | - Jun Wan
- Department of Cardiology, Renmin Hospital of Wuhan University, Department of Geriatrics, Zhongnan Hospital of Wuhan UniversityWuhan UniversityWuhanChina
- Cardiovascular Research InstituteWuhan UniversityWuhanChina
- Hubei Key Laboratory of CardiologyWuhanChina
| | - Menglong Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Department of Geriatrics, Zhongnan Hospital of Wuhan UniversityWuhan UniversityWuhanChina
- Cardiovascular Research InstituteWuhan UniversityWuhanChina
- Hubei Key Laboratory of CardiologyWuhanChina
| |
Collapse
|
13
|
Zhang J, Yin Z, Xu Y, Wei C, Peng S, Zhao M, Liu J, Xu S, Pan W, Zheng Z, Liu S, Ye J, Qin JJ, Wan J, Wang M. Resolvin E1/ChemR23 Protects Against Hypertension and Vascular Remodeling in Angiotensin II-Induced Hypertensive Mice. Hypertension 2023; 80:2650-2664. [PMID: 37800344 DOI: 10.1161/hypertensionaha.123.21348] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2023] [Accepted: 09/18/2023] [Indexed: 10/07/2023]
Abstract
BACKGROUND Inflammation plays a critical role in the development of hypertension and vascular remodeling. Resolvin E1 (RvE1), as one of the specialized proresolving lipid mediators, promotes inflammation resolution by binding with a G protein-coupled receptor, ChemR23 (chemerin receptor 23). However, whether RvE1/ChemR23 regulates hypertension and vascular remodeling is unknown. METHODS Hypertension in mice was induced by Ang II (angiotensin II) infusion (750 ng/kg per minute), and RvE1 (2 µg/kg per day) was administered through intraperitoneal injection. Loss of ChemR23 was achieved by mice receiving intravenous injection of adeno-associated virus 9-encoding shRNA against ChemR23. RESULTS Aortic ChemR23 expression was increased in Ang II-induced hypertensive mice and that ChemR23 was mainly expressed on vascular smooth muscle cells (VSMCs). RvE1 lowered blood pressure, reduced aortic media thickness, attenuated aortic fibrosis, and mitigated VSMC phenotypic transformation and proliferation in hypertensive mice, which were all reversed by the knockdown of ChemR23. Moreover, RvE1 reduced the aortic infiltration of macrophages and T cells, which was also reversed by ChemR23 knockdown. RvE1 inhibited Ccl5 expression in VSMCs via the AMPKα (AMP-activated protein kinase α)/Nrf2 (nuclear factor E2-related factor 2)/canonical NF-κB (nuclear factor κB) pathway, thereby reducing the infiltration of macrophages and T cells. The AMPKα/Nrf2 pathway also mediated the effects of RvE1 on VSMC phenotypic transformation and proliferation. In patients with hypertension, the serum levels of RvE1 and other eicosapentaenoic acid-derived metabolites were significantly decreased. CONCLUSIONS RvE1/ChemR23 ameliorated hypertension and vascular remodeling by activating AMPKα/Nrf2 signaling, which mediated immune cell infiltration by inhibiting the canonical NF-κB/Ccl5 pathway, and regulated VSMC proliferation and phenotypic transformation. RvE1/ChemR23 may be a potential therapeutic target for hypertension.
Collapse
Affiliation(s)
- Jishou Zhang
- Department of Cardiology, Renmin Hospital, Department of Geriatrics, Zhongnan Hospital, Wuhan University, China (J.Z., Z.Y., Y.X., C.W., S.P., M.Z., J.L., S.X., W.P., Z.Z., S.L., J.Y., J.-J.Q., J.W., M.W.)
- Cardiovascular Research Institute, Wuhan University, China (J.Z., Z.Y., Y.X., C.W., S.P., M.Z., J.L., S.X., W.P., Z.Z., S.L., J.Y., J.W., M.W.)
- Hubei Key Laboratory of Cardiology, Wuhan, China (J.Z., Z.Y., Y.X., C.W., S.P., M.Z., J.L., S.X., W.P., Z.Z., S.L., J.Y., J.W., M.W.)
| | - Zheng Yin
- Department of Cardiology, Renmin Hospital, Department of Geriatrics, Zhongnan Hospital, Wuhan University, China (J.Z., Z.Y., Y.X., C.W., S.P., M.Z., J.L., S.X., W.P., Z.Z., S.L., J.Y., J.-J.Q., J.W., M.W.)
- Cardiovascular Research Institute, Wuhan University, China (J.Z., Z.Y., Y.X., C.W., S.P., M.Z., J.L., S.X., W.P., Z.Z., S.L., J.Y., J.W., M.W.)
- Hubei Key Laboratory of Cardiology, Wuhan, China (J.Z., Z.Y., Y.X., C.W., S.P., M.Z., J.L., S.X., W.P., Z.Z., S.L., J.Y., J.W., M.W.)
| | - Yao Xu
- Department of Cardiology, Renmin Hospital, Department of Geriatrics, Zhongnan Hospital, Wuhan University, China (J.Z., Z.Y., Y.X., C.W., S.P., M.Z., J.L., S.X., W.P., Z.Z., S.L., J.Y., J.-J.Q., J.W., M.W.)
- Cardiovascular Research Institute, Wuhan University, China (J.Z., Z.Y., Y.X., C.W., S.P., M.Z., J.L., S.X., W.P., Z.Z., S.L., J.Y., J.W., M.W.)
- Hubei Key Laboratory of Cardiology, Wuhan, China (J.Z., Z.Y., Y.X., C.W., S.P., M.Z., J.L., S.X., W.P., Z.Z., S.L., J.Y., J.W., M.W.)
| | - Cheng Wei
- Department of Cardiology, Renmin Hospital, Department of Geriatrics, Zhongnan Hospital, Wuhan University, China (J.Z., Z.Y., Y.X., C.W., S.P., M.Z., J.L., S.X., W.P., Z.Z., S.L., J.Y., J.-J.Q., J.W., M.W.)
- Cardiovascular Research Institute, Wuhan University, China (J.Z., Z.Y., Y.X., C.W., S.P., M.Z., J.L., S.X., W.P., Z.Z., S.L., J.Y., J.W., M.W.)
- Hubei Key Laboratory of Cardiology, Wuhan, China (J.Z., Z.Y., Y.X., C.W., S.P., M.Z., J.L., S.X., W.P., Z.Z., S.L., J.Y., J.W., M.W.)
| | - Shanshan Peng
- Department of Cardiology, Renmin Hospital, Department of Geriatrics, Zhongnan Hospital, Wuhan University, China (J.Z., Z.Y., Y.X., C.W., S.P., M.Z., J.L., S.X., W.P., Z.Z., S.L., J.Y., J.-J.Q., J.W., M.W.)
- Cardiovascular Research Institute, Wuhan University, China (J.Z., Z.Y., Y.X., C.W., S.P., M.Z., J.L., S.X., W.P., Z.Z., S.L., J.Y., J.W., M.W.)
- Hubei Key Laboratory of Cardiology, Wuhan, China (J.Z., Z.Y., Y.X., C.W., S.P., M.Z., J.L., S.X., W.P., Z.Z., S.L., J.Y., J.W., M.W.)
| | - Mengmeng Zhao
- Department of Cardiology, Renmin Hospital, Department of Geriatrics, Zhongnan Hospital, Wuhan University, China (J.Z., Z.Y., Y.X., C.W., S.P., M.Z., J.L., S.X., W.P., Z.Z., S.L., J.Y., J.-J.Q., J.W., M.W.)
- Cardiovascular Research Institute, Wuhan University, China (J.Z., Z.Y., Y.X., C.W., S.P., M.Z., J.L., S.X., W.P., Z.Z., S.L., J.Y., J.W., M.W.)
- Hubei Key Laboratory of Cardiology, Wuhan, China (J.Z., Z.Y., Y.X., C.W., S.P., M.Z., J.L., S.X., W.P., Z.Z., S.L., J.Y., J.W., M.W.)
| | - Jianfang Liu
- Department of Cardiology, Renmin Hospital, Department of Geriatrics, Zhongnan Hospital, Wuhan University, China (J.Z., Z.Y., Y.X., C.W., S.P., M.Z., J.L., S.X., W.P., Z.Z., S.L., J.Y., J.-J.Q., J.W., M.W.)
- Cardiovascular Research Institute, Wuhan University, China (J.Z., Z.Y., Y.X., C.W., S.P., M.Z., J.L., S.X., W.P., Z.Z., S.L., J.Y., J.W., M.W.)
- Hubei Key Laboratory of Cardiology, Wuhan, China (J.Z., Z.Y., Y.X., C.W., S.P., M.Z., J.L., S.X., W.P., Z.Z., S.L., J.Y., J.W., M.W.)
| | - Shuwan Xu
- Department of Cardiology, Renmin Hospital, Department of Geriatrics, Zhongnan Hospital, Wuhan University, China (J.Z., Z.Y., Y.X., C.W., S.P., M.Z., J.L., S.X., W.P., Z.Z., S.L., J.Y., J.-J.Q., J.W., M.W.)
- Cardiovascular Research Institute, Wuhan University, China (J.Z., Z.Y., Y.X., C.W., S.P., M.Z., J.L., S.X., W.P., Z.Z., S.L., J.Y., J.W., M.W.)
- Hubei Key Laboratory of Cardiology, Wuhan, China (J.Z., Z.Y., Y.X., C.W., S.P., M.Z., J.L., S.X., W.P., Z.Z., S.L., J.Y., J.W., M.W.)
| | - Wei Pan
- Department of Cardiology, Renmin Hospital, Department of Geriatrics, Zhongnan Hospital, Wuhan University, China (J.Z., Z.Y., Y.X., C.W., S.P., M.Z., J.L., S.X., W.P., Z.Z., S.L., J.Y., J.-J.Q., J.W., M.W.)
- Cardiovascular Research Institute, Wuhan University, China (J.Z., Z.Y., Y.X., C.W., S.P., M.Z., J.L., S.X., W.P., Z.Z., S.L., J.Y., J.W., M.W.)
- Hubei Key Laboratory of Cardiology, Wuhan, China (J.Z., Z.Y., Y.X., C.W., S.P., M.Z., J.L., S.X., W.P., Z.Z., S.L., J.Y., J.W., M.W.)
| | - Zihui Zheng
- Department of Cardiology, Renmin Hospital, Department of Geriatrics, Zhongnan Hospital, Wuhan University, China (J.Z., Z.Y., Y.X., C.W., S.P., M.Z., J.L., S.X., W.P., Z.Z., S.L., J.Y., J.-J.Q., J.W., M.W.)
- Cardiovascular Research Institute, Wuhan University, China (J.Z., Z.Y., Y.X., C.W., S.P., M.Z., J.L., S.X., W.P., Z.Z., S.L., J.Y., J.W., M.W.)
- Hubei Key Laboratory of Cardiology, Wuhan, China (J.Z., Z.Y., Y.X., C.W., S.P., M.Z., J.L., S.X., W.P., Z.Z., S.L., J.Y., J.W., M.W.)
| | - Siqi Liu
- Department of Cardiology, Renmin Hospital, Department of Geriatrics, Zhongnan Hospital, Wuhan University, China (J.Z., Z.Y., Y.X., C.W., S.P., M.Z., J.L., S.X., W.P., Z.Z., S.L., J.Y., J.-J.Q., J.W., M.W.)
- Cardiovascular Research Institute, Wuhan University, China (J.Z., Z.Y., Y.X., C.W., S.P., M.Z., J.L., S.X., W.P., Z.Z., S.L., J.Y., J.W., M.W.)
- Hubei Key Laboratory of Cardiology, Wuhan, China (J.Z., Z.Y., Y.X., C.W., S.P., M.Z., J.L., S.X., W.P., Z.Z., S.L., J.Y., J.W., M.W.)
| | - Jing Ye
- Department of Cardiology, Renmin Hospital, Department of Geriatrics, Zhongnan Hospital, Wuhan University, China (J.Z., Z.Y., Y.X., C.W., S.P., M.Z., J.L., S.X., W.P., Z.Z., S.L., J.Y., J.-J.Q., J.W., M.W.)
- Cardiovascular Research Institute, Wuhan University, China (J.Z., Z.Y., Y.X., C.W., S.P., M.Z., J.L., S.X., W.P., Z.Z., S.L., J.Y., J.W., M.W.)
- Hubei Key Laboratory of Cardiology, Wuhan, China (J.Z., Z.Y., Y.X., C.W., S.P., M.Z., J.L., S.X., W.P., Z.Z., S.L., J.Y., J.W., M.W.)
| | - Juan-Juan Qin
- Department of Cardiology, Renmin Hospital, Department of Geriatrics, Zhongnan Hospital, Wuhan University, China (J.Z., Z.Y., Y.X., C.W., S.P., M.Z., J.L., S.X., W.P., Z.Z., S.L., J.Y., J.-J.Q., J.W., M.W.)
- Center for Healthy Aging, Wuhan University School of Nursing, China (J.-J.Q.)
| | - Jun Wan
- Department of Cardiology, Renmin Hospital, Department of Geriatrics, Zhongnan Hospital, Wuhan University, China (J.Z., Z.Y., Y.X., C.W., S.P., M.Z., J.L., S.X., W.P., Z.Z., S.L., J.Y., J.-J.Q., J.W., M.W.)
- Cardiovascular Research Institute, Wuhan University, China (J.Z., Z.Y., Y.X., C.W., S.P., M.Z., J.L., S.X., W.P., Z.Z., S.L., J.Y., J.W., M.W.)
- Hubei Key Laboratory of Cardiology, Wuhan, China (J.Z., Z.Y., Y.X., C.W., S.P., M.Z., J.L., S.X., W.P., Z.Z., S.L., J.Y., J.W., M.W.)
| | - Menglong Wang
- Department of Cardiology, Renmin Hospital, Department of Geriatrics, Zhongnan Hospital, Wuhan University, China (J.Z., Z.Y., Y.X., C.W., S.P., M.Z., J.L., S.X., W.P., Z.Z., S.L., J.Y., J.-J.Q., J.W., M.W.)
- Cardiovascular Research Institute, Wuhan University, China (J.Z., Z.Y., Y.X., C.W., S.P., M.Z., J.L., S.X., W.P., Z.Z., S.L., J.Y., J.W., M.W.)
- Hubei Key Laboratory of Cardiology, Wuhan, China (J.Z., Z.Y., Y.X., C.W., S.P., M.Z., J.L., S.X., W.P., Z.Z., S.L., J.Y., J.W., M.W.)
| |
Collapse
|
14
|
Zhang J, Xu Y, Wei C, Yin Z, Pan W, Zhao M, Ding W, Xu S, Liu J, Yu J, Ye J, Ye D, Qin JJ, Wan J, Wang M. Macrophage neogenin deficiency exacerbates myocardial remodeling and inflammation after acute myocardial infarction through JAK1-STAT1 signaling. Cell Mol Life Sci 2023; 80:324. [PMID: 37824022 PMCID: PMC11072237 DOI: 10.1007/s00018-023-04974-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 09/01/2023] [Accepted: 09/20/2023] [Indexed: 10/13/2023]
Abstract
Immune response plays a crucial role in post-myocardial infarction (MI) myocardial remodeling. Neogenin (Neo1), a multifunctional transmembrane receptor, plays a critical role in the immune response; however, whether Neo1 participates in pathological myocardial remodeling after MI is unclear. Our study found that Neo1 expression changed significantly after MI in vivo and after LPS + IFN-γ stimulation in bone marrow-derived macrophages (BMDMs) in vitro. Neo1 functional deficiency (using a neutralizing antibody) and macrophage-specific Neo1 deficiency (induced by Neo1flox/flox;Cx3cr1cre mice) increased infarction size, enhanced cardiac fibrosis and cardiomyocyte apoptosis, and exacerbated left ventricular dysfunction post-MI in mice. Mechanistically, Neo1 deficiency promoted macrophage infiltration into the ischemic myocardium and transformation to a proinflammatory phenotype, subsequently exacerbating the inflammatory response and impairing inflammation resolution post-MI. Neo1 deficiency regulated macrophage phenotype and function, possibly through the JAK1-STAT1 pathway, as confirmed in BMDMs in vitro. Blocking the JAK1-STAT1 pathway with fludarabine phosphate abolished the impact of Neo1 on macrophage phenotype and function, inflammatory response, inflammation resolution, cardiomyocyte apoptosis, cardiac fibrosis, infarction size and cardiac function. In conclusion, Neo1 deficiency aggravates inflammation and left ventricular remodeling post-MI by modulating macrophage phenotypes and functions via the JAK1-STAT1 signaling pathway. These findings highlight the anti-inflammatory potential of Neo1, offering new perspectives for therapeutic targets in MI treatment. Neo1 deficiency aggravated inflammation and left ventricular remodeling after MI by modulating macrophage phenotypes and functions via the JAK1-STAT1 signaling pathway.
Collapse
Affiliation(s)
- Jishou Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University, Department of Geriatrics, Zhongnan Hospital of Wuhan University, Wuhan University, 238 Jiefang Road, Wuhan, 430060, China
- Department of Cardiology, Renmin Hospital of Wuhan University, Hubei Key Laboratory of Cardiology, Cardiovascular Research Institute, Wuhan University, Wuhan, China
| | - Yao Xu
- Department of Cardiology, Renmin Hospital of Wuhan University, Hubei Key Laboratory of Cardiology, Cardiovascular Research Institute, Wuhan University, Wuhan, China
| | - Cheng Wei
- Department of Cardiology, Renmin Hospital of Wuhan University, Hubei Key Laboratory of Cardiology, Cardiovascular Research Institute, Wuhan University, Wuhan, China
| | - Zheng Yin
- Department of Cardiology, Renmin Hospital of Wuhan University, Hubei Key Laboratory of Cardiology, Cardiovascular Research Institute, Wuhan University, Wuhan, China
| | - Wei Pan
- Department of Cardiology, Renmin Hospital of Wuhan University, Hubei Key Laboratory of Cardiology, Cardiovascular Research Institute, Wuhan University, Wuhan, China
| | - Mengmeng Zhao
- Department of Cardiology, Renmin Hospital of Wuhan University, Hubei Key Laboratory of Cardiology, Cardiovascular Research Institute, Wuhan University, Wuhan, China
| | - Wen Ding
- Department of Cardiology, Renmin Hospital of Wuhan University, Hubei Key Laboratory of Cardiology, Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Department of Radiology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Shuwan Xu
- Department of Cardiology, Renmin Hospital of Wuhan University, Hubei Key Laboratory of Cardiology, Cardiovascular Research Institute, Wuhan University, Wuhan, China
| | - Jianfang Liu
- Department of Cardiology, Renmin Hospital of Wuhan University, Hubei Key Laboratory of Cardiology, Cardiovascular Research Institute, Wuhan University, Wuhan, China
| | - Junping Yu
- Department of Cardiology, Renmin Hospital of Wuhan University, Hubei Key Laboratory of Cardiology, Cardiovascular Research Institute, Wuhan University, Wuhan, China
| | - Jing Ye
- Department of Cardiology, Renmin Hospital of Wuhan University, Hubei Key Laboratory of Cardiology, Cardiovascular Research Institute, Wuhan University, Wuhan, China
| | - Di Ye
- Department of Cardiology, Renmin Hospital of Wuhan University, Hubei Key Laboratory of Cardiology, Cardiovascular Research Institute, Wuhan University, Wuhan, China
| | - Juan-Juan Qin
- Department of Cardiology, Renmin Hospital of Wuhan University, Department of Geriatrics, Zhongnan Hospital of Wuhan University, Wuhan University, 238 Jiefang Road, Wuhan, 430060, China.
- Center for Healthy Aging, Wuhan University School of Nursing, Wuhan, China.
| | - Jun Wan
- Department of Cardiology, Renmin Hospital of Wuhan University, Department of Geriatrics, Zhongnan Hospital of Wuhan University, Wuhan University, 238 Jiefang Road, Wuhan, 430060, China.
- Department of Cardiology, Renmin Hospital of Wuhan University, Hubei Key Laboratory of Cardiology, Cardiovascular Research Institute, Wuhan University, Wuhan, China.
| | - Menglong Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Department of Geriatrics, Zhongnan Hospital of Wuhan University, Wuhan University, 238 Jiefang Road, Wuhan, 430060, China.
- Department of Cardiology, Renmin Hospital of Wuhan University, Hubei Key Laboratory of Cardiology, Cardiovascular Research Institute, Wuhan University, Wuhan, China.
| |
Collapse
|
15
|
Shen C, Fang R, Wang J, Wu N, Wang S, Shu T, Dai J, Feng M, Chen X. Visfatin aggravates transverse aortic constriction-induced cardiac remodelling by enhancing macrophage-mediated oxidative stress in mice. J Cell Mol Med 2023; 27:2562-2571. [PMID: 37584247 PMCID: PMC10468652 DOI: 10.1111/jcmm.17854] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 06/27/2023] [Accepted: 07/10/2023] [Indexed: 08/17/2023] Open
Abstract
Previous studies have reported that visfatin can regulate macrophage polarisation, which has been demonstrated to participate in cardiac remodelling. The aims of this study were to investigate whether visfatin participates in transverse aortic constriction (TAC)-induced cardiac remodelling by regulating macrophage polarisation. First, TAC surgery and angiotensin II (Ang II) infusion were used to establish a mouse cardiac remodelling model, visfatin expression was measured, and the results showed that TAC surgery or Ang II infusion increased visfatin expression in the serum and heart in mice, and phenylephrine or hydrogen peroxide promoted the release of visfatin from macrophages in vitro. All these effects were dose-dependently reduced by superoxide dismutase. Second, visfatin was administered to TAC mice to observe the effects of visfatin on cardiac remodelling. We found that visfatin increased the cross-sectional area of cardiomyocytes, aggravated cardiac fibrosis, exacerbated cardiac dysfunction, further regulated macrophage polarisation and aggravated oxidative stress in TAC mice. Finally, macrophages were depleted in TAC mice to investigate whether macrophages mediate the regulatory effect of visfatin on cardiac remodelling, and the results showed that the aggravating effects of visfatin on oxidative stress and cardiac remodelling were abrogated. Our study suggests that visfatin enhances cardiac remodelling by promoting macrophage polarisation and enhancing oxidative stress. Visfatin may be a potential target for the prevention and treatment of clinical cardiac remodelling.
Collapse
Affiliation(s)
- Caijie Shen
- Department of Cardiovascular MedicineThe First Affliated Hospital of Ningbo University, Key Laboratory of Precision Medicine for Atherosclerotic Diseases of Zhejiang ProvinceNingboChina
| | - Renyuan Fang
- Department of Cardiovascular MedicineThe First Affliated Hospital of Ningbo University, Key Laboratory of Precision Medicine for Atherosclerotic Diseases of Zhejiang ProvinceNingboChina
| | - Jian Wang
- Department of Cardiovascular MedicineThe First Affliated Hospital of Ningbo University, Key Laboratory of Precision Medicine for Atherosclerotic Diseases of Zhejiang ProvinceNingboChina
| | - Nan Wu
- Department of Cardiovascular MedicineThe First Affliated Hospital of Ningbo University, Key Laboratory of Precision Medicine for Atherosclerotic Diseases of Zhejiang ProvinceNingboChina
| | - Shuangsuang Wang
- Department of CardiologyWenling First People's Hospital, The Affiliated Wenling Hospital of Wenzhou Medical UniversityWenzhouChina
| | - Tian Shu
- Zhejiang University School of MedicineHangzhouChina
| | - Jiating Dai
- Health Science Center, Ningbo UniversityNingboChina
| | - Mingjun Feng
- Department of Cardiovascular MedicineThe First Affliated Hospital of Ningbo University, Key Laboratory of Precision Medicine for Atherosclerotic Diseases of Zhejiang ProvinceNingboChina
| | - Xiaomin Chen
- Department of Cardiovascular MedicineThe First Affliated Hospital of Ningbo University, Key Laboratory of Precision Medicine for Atherosclerotic Diseases of Zhejiang ProvinceNingboChina
| |
Collapse
|
16
|
Yihui C, Yanfeng G. Inflammatory markers in patients with hypertension. Br J Hosp Med (Lond) 2023; 84:1-8. [PMID: 37235676 DOI: 10.12968/hmed.2022.0531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/28/2023]
Abstract
Hypertension is a chronic disease with high levels of morbidity and disability. Elevated blood pressure can lead to many complications and is the main risk factor for stroke, heart failure and nephropathy. Factors associated with hypertension and inflammatory response differ from those associated with vascular inflammation. The immune system plays a vital role in the pathophysiology of hypertension. Inflammation is particularly relevant in the progression of cardiovascular diseases, which has led to extensive research on inflammatory markers and indicators.
Collapse
Affiliation(s)
- Chen Yihui
- Department of General Practice, First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Gong Yanfeng
- Department of General Practice, First Affiliated Hospital of Nanchang University, Nanchang, China
| |
Collapse
|
17
|
Wu Z, Luo C, Zheng B. Progress of Research into the Interleukin-1 Family in Cardiovascular Disease. J Inflamm Res 2022; 15:6683-6694. [PMID: 36536642 PMCID: PMC9759010 DOI: 10.2147/jir.s390915] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Accepted: 11/30/2022] [Indexed: 09/01/2023] Open
Abstract
Inflammatory factors, such as the IL-1 family, are generally acknowledged to be involved in systemic diseases and IL-1α and IL-1β, in particular, have been linked to cardiovascular disease with IL-18, IL-33, IL-36, IL-37 and IL-38 yet to be explored. The current review aims to summarize mechanisms of IL-18, IL-33, IL-36, IL-37 and IL-38 in myocardial infarction, hypertension, arrhythmia, valvular disease and aneurysm and to explore the potential for cardiovascular disease treatment strategies and discuss future directions for prevention and treatment.
Collapse
Affiliation(s)
- Zimin Wu
- Department of Cardiovascular Surgery Ward, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, 530021, People’s Republic of China
| | - Cheng Luo
- Department of Cardiovascular Surgery Ward, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, 530021, People’s Republic of China
| | - Baoshi Zheng
- Department of Cardiovascular Surgery Ward, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, 530021, People’s Republic of China
| |
Collapse
|
18
|
Zhao M, Zheng Z, Li C, Wan J, Wang M. Developmental endothelial locus-1 in cardiovascular and metabolic diseases: A promising biomarker and therapeutic target. Front Immunol 2022; 13:1053175. [PMID: 36518760 PMCID: PMC9742254 DOI: 10.3389/fimmu.2022.1053175] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2022] [Accepted: 11/11/2022] [Indexed: 11/29/2022] Open
Abstract
Cardiovascular and metabolic diseases (CVMDs) are a leading cause of death worldwide and impose a major socioeconomic burden on individuals and healthcare systems, underscoring the urgent need to develop new drug therapies. Developmental endothelial locus-1 (DEL-1) is a secreted multifunctional domain protein that can bind to integrins and play an important role in the occurrence and development of various diseases. Recently, DEL-1 has attracted increased interest for its pharmacological role in the treatment and/or management of CVMDs. In this review, we present the current knowledge on the predictive and therapeutic role of DEL-1 in a variety of CVMDs, such as atherosclerosis, hypertension, cardiac remodeling, ischemic heart disease, obesity, and insulin resistance. Collectively, DEL-1 is a promising biomarker and therapeutic target for CVMDs.
Collapse
Affiliation(s)
- Mengmeng Zhao
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China,Cardiovascular Research Institute, Wuhan University, Wuhan, China,Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Zihui Zheng
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China,Cardiovascular Research Institute, Wuhan University, Wuhan, China,Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Chenfei Li
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China,Cardiovascular Research Institute, Wuhan University, Wuhan, China
| | - Jun Wan
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China,Cardiovascular Research Institute, Wuhan University, Wuhan, China,Hubei Key Laboratory of Cardiology, Wuhan, China,*Correspondence: Menglong Wang, ; Jun Wan,
| | - Menglong Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China,Cardiovascular Research Institute, Wuhan University, Wuhan, China,Hubei Key Laboratory of Cardiology, Wuhan, China,*Correspondence: Menglong Wang, ; Jun Wan,
| |
Collapse
|
19
|
Ngwa C, Al Mamun A, Qi S, Sharmeen R, Xu Y, Liu F. Regulation of microglial activation in stroke in aged mice: a translational study. Aging (Albany NY) 2022; 14:6047-6065. [PMID: 35963621 PMCID: PMC9417226 DOI: 10.18632/aging.204216] [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: 03/09/2022] [Accepted: 06/14/2022] [Indexed: 11/25/2022]
Abstract
Numerous neurochemical changes occur with aging and stroke mainly affects the elderly. Our previous study has found interferon regulatory factor 5 (IRF5) and 4 (IRF4) regulate neuroinflammation in young stroke mice. However, whether the IRF5-IRF4 regulatory axis has the same effect in aged brains is not known. In this study, aged (18-20-month-old), microglial IRF5 or IRF4 conditional knockout (CKO) mice were subjected to a 60-min middle cerebral artery occlusion (MCAO). Stroke outcomes were quantified at 3d after MCAO. Flow cytometry and ELISA were performed to evaluate microglial activation and immune responses. We found aged microglia express higher levels of IRF5 and lower levels of IRF4 than young microglia after stroke. IRF5 CKO aged mice had improved stroke outcomes; whereas worse outcomes were seen in IRF4 CKO vs. their flox controls. IRF5 CKO aged microglia had significantly lower levels of IL-1β and CD68 than controls; whereas significantly higher levels of IL-1β and TNF-α were seen in IRF4 CKO vs. control microglia. Plasma levels of TNF-α and MIP-1α were decreased in IRF5 CKO vs. flox aged mice, and IL-1β/IL-6 levels were increased in IRF4 CKO vs. controls. The anti-inflammatory cytokines (IL-4/IL-10) levels were higher in IRF5 CKO, and lower in IRF4 CKO aged mice vs. their flox controls. IRF5 and IRF4 signaling drives microglial pro- and anti-inflammatory response respectively; microglial IRF5 is detrimental and IRF4 beneficial for aged mice in stroke. IRF5-IRF4 axis is a promising target for developing new, effective therapeutic strategies for the cerebral ischemia.
Collapse
Affiliation(s)
- Conelius Ngwa
- Department of Neurology, The University of Texas Health Science Center at Houston, McGovern Medical School, Houston, TX 77030, USA
| | - Abdullah Al Mamun
- Department of Neurology, The University of Texas Health Science Center at Houston, McGovern Medical School, Houston, TX 77030, USA
| | - Shaohua Qi
- Department of Neurology, The University of Texas Health Science Center at Houston, McGovern Medical School, Houston, TX 77030, USA
| | - Romana Sharmeen
- Department of Neurology, The University of Texas Health Science Center at Houston, McGovern Medical School, Houston, TX 77030, USA
| | - Yan Xu
- Department of Neurology, The University of Texas Health Science Center at Houston, McGovern Medical School, Houston, TX 77030, USA
| | - Fudong Liu
- Department of Neurology, The University of Texas Health Science Center at Houston, McGovern Medical School, Houston, TX 77030, USA
| |
Collapse
|
20
|
Chen S, Xia J, Zhang Y, Zhan Q. IL35 attenuated LPS-induced acute lung injury by regulating macrophage polarization. Mol Biol Rep 2022; 49:5811-5820. [PMID: 35748972 PMCID: PMC9244303 DOI: 10.1007/s11033-022-07293-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 01/27/2022] [Accepted: 02/23/2022] [Indexed: 12/01/2022]
Abstract
BACKGROUND Interleukin 35 (IL35) has been reported to play a role in acute lung injury (ALI); however, the current results regarding the relationship between IL35 and ALI are inconsistent. Therefore, we aimed to further determine the function of IL35 in ALI in mice and the potential mechanism in this paper. MATERIALS AND METHODS Hematoxylin-eosin (HE) staining and Masson staining were used to evaluate lung injury in mice. Immunohistochemical staining was used to evaluate the expression of IL35 p35, TLR4 and MD2 and the Bax/Bcl2 and p-P65/P65 ratios. The expression levels of IL35 EBi3, CD68, CD206 and MPO were assessed by immunofluorescence staining. RT-PCR was used to examine the expression levels of IL1β and IL6. Terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling (TUNEL) staining was performed to detect apoptotic cells. RESULTS Overexpression of IL35 alleviated LPS-induced ALI in mice. IL35 overexpression decreased the expression of CD68 and increased the expression of CD206 in mice with ALI. Furthermore, upregulation of IL35 expression obviously reduced the expression of MPO, IL1β and IL6 in the lung tissues of mice with ALI. Mechanistically, IL35 suppressed the TLR4/NFκB-P65 pathway, leading to the promotion of the M1 to M2 macrophage transition and alleviation of inflammation in mice with ALI. CONCLUSIONS IL35 relieved LPS-induced inflammation and ALI in mice by regulating M1/M2 macrophage polarization and inhibiting the activation of the TLR4/NFκB-P65 pathway.
Collapse
Affiliation(s)
- Shengsong Chen
- Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, No 2, East Yinghua Road, Chaoyang District, 100029, Beijing, P. R. China
- Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences, No 9, Dongdan Santao, Dongcheng District, 100730, Beijing, P. R. China
- National Center for Respiratory Medicine, No 2, East Yinghua Road, Chaoyang District, 100029, Beijing, P. R. China
- Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, No 2, East Yinghua Road, Chaoyang District, 100029, Beijing, P. R. China
- National Clinical Research Center for Respiratory Diseases, No 2, East Yinghua Road, Chaoyang District, 100029, Beijing, P. R. China
- WHO Collaborating Centre for Tobacco Cessation and Respiratory Diseases Prevention, No 2, East Yinghua Road, Chaoyang District, 100029, Beijing, P. R. China
| | - Jingen Xia
- Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, No 2, East Yinghua Road, Chaoyang District, 100029, Beijing, P. R. China
- National Center for Respiratory Medicine, No 2, East Yinghua Road, Chaoyang District, 100029, Beijing, P. R. China
- Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, No 2, East Yinghua Road, Chaoyang District, 100029, Beijing, P. R. China
- National Clinical Research Center for Respiratory Diseases, No 2, East Yinghua Road, Chaoyang District, 100029, Beijing, P. R. China
- WHO Collaborating Centre for Tobacco Cessation and Respiratory Diseases Prevention, No 2, East Yinghua Road, Chaoyang District, 100029, Beijing, P. R. China
| | - Yi Zhang
- Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, No 2, East Yinghua Road, Chaoyang District, 100029, Beijing, P. R. China.
- National Center for Respiratory Medicine, No 2, East Yinghua Road, Chaoyang District, 100029, Beijing, P. R. China.
- Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, No 2, East Yinghua Road, Chaoyang District, 100029, Beijing, P. R. China.
- National Clinical Research Center for Respiratory Diseases, No 2, East Yinghua Road, Chaoyang District, 100029, Beijing, P. R. China.
- WHO Collaborating Centre for Tobacco Cessation and Respiratory Diseases Prevention, No 2, East Yinghua Road, Chaoyang District, 100029, Beijing, P. R. China.
| | - Qingyuan Zhan
- Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, No 2, East Yinghua Road, Chaoyang District, 100029, Beijing, P. R. China.
- Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences, No 9, Dongdan Santao, Dongcheng District, 100730, Beijing, P. R. China.
- National Center for Respiratory Medicine, No 2, East Yinghua Road, Chaoyang District, 100029, Beijing, P. R. China.
- Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, No 2, East Yinghua Road, Chaoyang District, 100029, Beijing, P. R. China.
- National Clinical Research Center for Respiratory Diseases, No 2, East Yinghua Road, Chaoyang District, 100029, Beijing, P. R. China.
- WHO Collaborating Centre for Tobacco Cessation and Respiratory Diseases Prevention, No 2, East Yinghua Road, Chaoyang District, 100029, Beijing, P. R. China.
| |
Collapse
|
21
|
Liu S, Wang C, Guo J, Yang Y, Huang M, Li L, Wang Y, Qin Y, Zhang M. Serum Cytokines Predict the Severity of Coronary Artery Disease Without Acute Myocardial Infarction. Front Cardiovasc Med 2022; 9:896810. [PMID: 35651907 PMCID: PMC9149173 DOI: 10.3389/fcvm.2022.896810] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 04/25/2022] [Indexed: 12/05/2022] Open
Abstract
Introduction Various cytokines were involved in the process of atherosclerosis, and their serum levels were correlated with coronary artery disease (CAD) to varying degrees. However, there were limited reports about the correlation between serum cytokines and the severity of coronary atherosclerotic lesion in patients with non-acute myocardial infarction (AMI). The purpose of this study was to investigate the relationship between serum cytokines and the severity of CAD, and identify the predictors of severe CAD in patients suspected to have CAD but AMI had been ruled out. Methods A total of 502 patients who had suspected CAD and underwent coronary angiography were enrolled. The serum levels of IL-1β, IL-2, IL-4, IL-5, IL-6, IL-8, IL-10, IL-12p70, IL-17, TNF-α, IFN-α,and IFN-γ were determined by multiplexed particle-based flow cytometric assays technology. And the severity of CAD was evaluated by Gensini score (GS). Results The serum levels of IL-4, IL-12p70, IL-17, and IFN-α were significantly lower in the severe CAD group (GS≥30) than those in the non-severe CAD group (GS < 30). And IL-12p70 and IL-17 were negatively correlated with the severity of CAD. Multivariate logistic regression analyses demonstrated that two serum cytokines (IL-12p70 and IL-17), one clinical protective factor (HDL-C), and two clinical risk factors (gender and diabetes) were the independent predictors of severe CAD. ROC curve analysis showed that multivariate mode combined these predictors had a good performance in predicting severe CAD. Conclusion The combination of serum cytokines (IL-12p70 and IL-17) and clinical risk factors (HDL-C, gender, and diabetes) may help identify patients with more severe coronary artery lesions from those with suspected CAD but not AMI, and may contribute to guiding the risk stratification for patients with chest discomfort in health care facilities without sufficient medical resources (especially cardiac catheterization resources).
Collapse
Affiliation(s)
- Sheng Liu
- Center for Coronary Heart Disease, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Chenyang Wang
- Center for Coronary Heart Disease, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Jinzhu Guo
- Department of Cardiology, Baotou Jiuyuan District Hospital, Baotou, China
| | - Yunxiao Yang
- Surgical Center of Structural Heart Disease, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Mengling Huang
- Center for Coronary Heart Disease, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Li Li
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Yu Wang
- Key Laboratory of Upper Airway Dysfunction-related Cardiovascular Diseases, Beijing Institute of Heart, Lung and Blood Vessel Disease, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Yanwen Qin
- Key Laboratory of Upper Airway Dysfunction-related Cardiovascular Diseases, Beijing Institute of Heart, Lung and Blood Vessel Disease, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Ming Zhang
- Center for Coronary Heart Disease, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
22
|
Margraf A, Perretti M. Immune Cell Plasticity in Inflammation: Insights into Description and Regulation of Immune Cell Phenotypes. Cells 2022; 11:cells11111824. [PMID: 35681519 PMCID: PMC9180515 DOI: 10.3390/cells11111824] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 05/28/2022] [Accepted: 05/30/2022] [Indexed: 02/01/2023] Open
Abstract
Inflammation is a life-saving immune reaction occurring in response to invading pathogens. Nonetheless, inflammation can also occur in an uncontrolled, unrestricted manner, leading to chronic disease and organ damage. Mechanisms triggering an inflammatory response, hindering such a response, or leading to its resolution are well-studied but so far insufficiently elucidated with regard to precise therapeutic interventions. Notably, as an immune reaction evolves, requirements and environments for immune cells change, and thus cellular phenotypes adapt and shift, leading to the appearance of distinct cellular subpopulations with new functional features. In this article, we aim to highlight properties of, and overarching regulatory factors involved in, the occurrence of immune cell phenotypes with a special focus on neutrophils, macrophages and platelets. Additionally, we point out implications for both diagnostics and therapeutics in inflammation research.
Collapse
|
23
|
Wang Y, Li J, Xu Y, Liao S, Song J, Xu Z, Wei W, Zhu S. Interleukin-22 Deficiency Reduces Angiotensin II-Induced Aortic Dissection and Abdominal Aortic Aneurysm in ApoE-/- Mice. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:7555492. [PMID: 35340206 PMCID: PMC8956387 DOI: 10.1155/2022/7555492] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 02/02/2022] [Accepted: 02/23/2022] [Indexed: 11/18/2022]
Abstract
Background Our previous study showed that interleukin-22 (IL-22) levels were increased in patients with aortic dissection (AD). This study evaluated the effects of IL-22 on AD/abdominal aortic aneurysm (AAA) formation in angiotensin II (Ang II)-infused ApoE-/- mice. Methods ApoE-/- mice were treated with Ang II for 28 days, and IL-22 expression was examined. In addition, the effects of IL22 deficiency on AAA/AD formation induced by Ang II infusion in ApoE-/- mice were investigated. ApoE-/-IL-22-/- mice were transplanted with bone marrow cells isolated from ApoE-/- mice or ApoE-/-IL-22-/- mice, and AAA/AD formation was observed. Results IL-22 expression was increased in both the aortas and serum of ApoE-/- mice after Ang II infusion and was mainly derived from aortic CD4+ T lymphocytes (CD4+ TCs). IL-22 deficiency significantly reduced the AAA/AD formation as well as the maximal aortic diameter in Ang II-infused ApoE-/- mice. Decreased elastin fragmentation and reduced fibrosis were observed in the aortas of ApoE-/-IL-22-/- mice compared with ApoE-/- mice. The deletion of IL-22 also decreased aortic M1 macrophage differentiation, alleviated M1 macrophage-induced oxidative stress, and reduced aortic smooth muscle cell loss. Furthermore, M1 macrophage-induced oxidative stress was worsened and AAA/AD formation was promoted in ApoE-/-IL-22-/- mice that received transplanted bone marrow cells from ApoE-/- mice compared with those that were transplanted with bone marrow cells isolated from ApoE-/-IL-22-/- mice. Conclusions IL-22 deficiency inhibits AAA/AD formation by inhibiting M1 macrophage-induced oxidative stress. IL-22 potentially represents a promising new target for preventing the progression of AAA/AD.
Collapse
Affiliation(s)
- Yuan Wang
- Department of Thyroid Breast Surgery, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Juanjuan Li
- Department of Thyroid Breast Surgery, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Yulin Xu
- Department of Thyroid Breast Surgery, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Shichong Liao
- Department of Thyroid Breast Surgery, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Junlong Song
- Department of Thyroid Breast Surgery, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Zhiliang Xu
- Department of Thyroid Breast Surgery, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Wen Wei
- Department of Thyroid Breast Surgery, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Shan Zhu
- Department of Thyroid Breast Surgery, Renmin Hospital of Wuhan University, Wuhan 430060, China
| |
Collapse
|
24
|
Wang Z, Liu M, Ye D, Ye J, Wang M, Liu J, Xu Y, Zhang J, Zhao M, Feng Y, Xu S, Pan W, Luo Z, Li D, Wan J. Il12a Deletion Aggravates Sepsis-Induced Cardiac Dysfunction by Regulating Macrophage Polarization. Front Pharmacol 2021; 12:632912. [PMID: 34276358 PMCID: PMC8284189 DOI: 10.3389/fphar.2021.632912] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Accepted: 03/17/2021] [Indexed: 12/12/2022] Open
Abstract
Cardiac dysfunction is a well-recognized complication of sepsis and is associated with the outcome and prognosis of septic patients. Evidence suggests that Il12a participates in the regulation of various cardiovascular diseases, including heart failure, hypertension and acute myocardial infarction. However, the effects of Il12a in sepsis-induced cardiac dysfunction remain unknown. In our study, lipopolysaccharide (LPS) and cecal ligation and puncture (CLP) model were used to mimic sepsis, and cardiac Il12a expression was assessed. In addition, Il12a knockout mice were used to detect the role of Il12a in sepsis-related cardiac dysfunction. We observed for the first time that Il12a expression is upregulated in mice after LPS treatment and macrophages were the main sources of Il12a. In addition, our findings demonstrated that Il12a deletion aggravates LPS-induced cardiac dysfunction and injury, as evidenced by the increased serum and cardiac levels of lactate dehydrogenase (LDH) and cardiac creatine kinase-myocardial band (CK-MB). Moreover, Il12a deletion enhances LPS-induced macrophage accumulation and drives macrophages toward the M1 phenotype in LPS-treated mice. Il12a deletion also downregulated the activity of AMP-activated protein kinase (AMPK) but increased the phosphorylation levels of p65 (p-p65) and NF-κB inhibitor alpha (p-IκBα). In addition, Il12a deletion aggravates CLP-induced cardiac dysfunction and injury. Treatment with the AMPK activator AICAR abolishes the deterioration effect of Il12a deletion on LPS-induced cardiac dysfunction. In conclusion, Il12a deletion aggravated LPS-induced cardiac dysfunction and injury by exacerbating the imbalance of M1 and M2 macrophages. Our data provide evidence that Il12a may represent an attractive target for sepsis-induced cardiac dysfunction.
Collapse
Affiliation(s)
- Zhen Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Menglin Liu
- Department of Emergency, Renmin Hospital of Wuhan University, Wuhan, China
| | - Di Ye
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Jing Ye
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Menglong Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Jianfang Liu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Yao Xu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Jishou Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Mengmeng Zhao
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Yongqi Feng
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Shuwan Xu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Wei Pan
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Zhen Luo
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Dan Li
- Department of Pediatrics, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Jun Wan
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| |
Collapse
|
25
|
Liang Z, Pan F, Yang Z, Wang M, Hu C, Shi L, Ji Q, Liu L. Interleukin-9 deficiency affects lipopolysaccharide-induced macrophage-related oxidative stress and myocardial cell apoptosis via the Nrf2 pathway both in vivo and in vitro. Biofactors 2021; 47:674-685. [PMID: 33979459 DOI: 10.1002/biof.1754] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2021] [Accepted: 04/25/2021] [Indexed: 01/29/2023]
Abstract
Previous studies showed that interleukin-9 (IL-9) is involved in cardiovascular diseases, including hypertension and cardiac fibrosis. This study aimed to investigate the role of IL-9 in lipopolysaccharide (LPS)-induced myocardial cell (MC) apoptosis. Mice were treated with LPS, and IL-9 expression was measured and the results showed that compared with WT mice, LPS-treated mice exhibited increased cardiac Mø-derived IL-9. Additionally, the effects of IL-9 deficiency (IL-9-/-) on macrophage (Mø)-related oxidative stress and MC apoptosis were evaluated, the results showed that IL-9 knockout significantly exacerbated cardiac dysfunction, inhibited Nrf2 nuclear transfer, promoted an imbalance in M1 and M2 Møs, and exacerbated oxidative stress and MC apoptosis in LPS-treated mice. Treatment with ML385, a specific nuclear factor erythroid-2 related factor 2 (Nrf2) pathway inhibitor significantly alleviated the above effects in LPS-treated IL-9-/- mice. Bone marrow-derived Møs from wild-type (WT) mice and IL-9-/- mice were treated with LPS, and the differentiation and oxidative stress levels of Møs were measured. The effect of Mø differentiation on mouse MC apoptosis was also analyzed in vitro. The results showed that LPS-induced M1 Mø/M2 Mø imbalance and Mø-related oxidative stress were alleviated by IL-9 knockout but were exacerbated by ML385 treatment. The protective effects of IL-9 deficiency on the MC apoptosis mediated by LPS-treated Møs were reversed by ML-385. Our results suggest that deletion of IL-9 decreased the nuclear translocation of Nrf2 in Møs, which further aggravated Mø-related oxidative stress and MC apoptosis. IL-9 may be a target for the prevention of LPS-induced cardiac injury.
Collapse
MESH Headings
- Animals
- Antigens, CD/genetics
- Antigens, CD/immunology
- Apoptosis/genetics
- Apoptosis/immunology
- Cell Nucleus/metabolism
- Cytoplasm/metabolism
- Gene Expression Regulation
- Interleukin-9/deficiency
- Interleukin-9/genetics
- Interleukin-9/immunology
- Lipopolysaccharides/administration & dosage
- Macrophages/immunology
- Macrophages/pathology
- Male
- Mice
- Mice, Knockout
- Myocarditis/chemically induced
- Myocarditis/genetics
- Myocarditis/immunology
- Myocarditis/pathology
- Myocytes, Cardiac/immunology
- Myocytes, Cardiac/pathology
- NF-E2-Related Factor 2/antagonists & inhibitors
- NF-E2-Related Factor 2/genetics
- NF-E2-Related Factor 2/immunology
- Oxidative Stress
- Primary Cell Culture
- Protein Transport
- Proto-Oncogene Proteins c-bcl-2/genetics
- Proto-Oncogene Proteins c-bcl-2/immunology
- Receptors, Atrial Natriuretic Factor/genetics
- Receptors, Atrial Natriuretic Factor/immunology
- Receptors, Interleukin-9/genetics
- Receptors, Interleukin-9/immunology
- Signal Transduction
- Thiazoles/pharmacology
- Ventricular Function, Left/physiology
- bcl-2-Associated X Protein/genetics
- bcl-2-Associated X Protein/immunology
Collapse
Affiliation(s)
- Zhishan Liang
- Department of Cardiology, the People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Fuze Pan
- Department of Cardiology, the People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Zicong Yang
- Department of Cardiology, the People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Mengjie Wang
- Department of Cardiology, the People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Changxing Hu
- Department of Cardiology, the People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Lei Shi
- Department of Cardiology, the People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Qingwei Ji
- Department of Cardiology, the People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Ling Liu
- Department of Cardiology, the People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| |
Collapse
|
26
|
Anti-Interleukin-16-Neutralizing Antibody Attenuates Cardiac Inflammation and Protects against Cardiac Injury in Doxorubicin-Treated Mice. Mediators Inflamm 2021; 2021:6611085. [PMID: 33958974 PMCID: PMC8075660 DOI: 10.1155/2021/6611085] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 03/04/2021] [Accepted: 04/05/2021] [Indexed: 12/11/2022] Open
Abstract
Background Interleukin-16 (IL-16) is an important inflammatory regulator and has been shown to have a powerful effect on the regulation of the inflammatory response. Cardiac inflammation has been reported to be closely related to doxorubicin- (DOX-) induced cardiac injury. In this study, the role of IL-16 in DOX-induced cardiac injury and the possible mechanisms were examined. Methods Cardiac IL-16 levels were first measured in DOX- or saline-treated mice. Additionally, mice were pretreated with the anti-IL-16-neutralizing antibody (nAb) or isotype IgG for 1 day and further administered DOX or saline for 5 days. Then, cardiac injury, cardiac M1 macrophage levels, and cardiomyocyte apoptosis were analyzed. The effects of the anti-IL-16 nAb on macrophage differentiation and cardiomyocyte apoptosis were also investigated in vitro. Results DOX administration increased IL-16 expression in cardiac macrophages compared with that of saline treatment. The anti-IL-16 nAb significantly decreased serum levels of lactate dehydrogenase (LDH), myocardial-bound creatine kinase (CK-MB), and cardiac troponin T (cTnT) and elevated cardiac function in DOX-induced mice. Treatment with the anti-IL-16 nAb also reduced p65 pathway activation, decreased M1 macrophage-related marker and cytokine expression, and protected against cardiomyocyte apoptosis in DOX-induced mice. In cell studies, the anti-IL-16 nAb also reduced DOX-induced M1 macrophage differentiation and alleviated apoptosis in cardiomyocytes cocultured with macrophages. Conclusions The anti-IL-16 nAb protects against DOX-induced cardiac injury by reducing cardiac inflammation, and IL-16 may be a promising target to prevent DOX-related cardiac injury.
Collapse
|
27
|
Yu Y, Singh H, Kwon K, Tsitrin T, Petrini J, Nelson KE, Pieper R. Protein signatures from blood plasma and urine suggest changes in vascular function and IL-12 signaling in elderly with a history of chronic diseases compared with an age-matched healthy cohort. GeroScience 2021. [PMID: 32974878 DOI: 10.1007/s11357-020-00269-y/figures/10] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/26/2023] Open
Abstract
Key processes characterizing human aging are immunosenescence and inflammaging. The capacity of the immune system to adequately respond to external perturbations (e.g., pathogens, injuries, and biochemical irritants) and to repair somatic mutations that may cause cancers or cellular senescence declines. An important goal remains to identify genetic or biochemical, predictive biomarkers for healthy aging. We recruited two cohorts in the age range 70 to 82, one afflicted by chronic illnesses (non-healthy aging, NHA) and the other in good health (healthy aging, HA). NHA criteria included major cardiovascular, neurodegenerative, and chronic pulmonary diseases, diabetes, and cancers. Quantitative analysis of forty proinflammatory cytokines in blood plasma and more than 500 proteins in urine was performed to identify candidate biomarkers for and biological pathway implications of healthy aging. Nine cytokines revealed lower quantities in blood plasma for the NHA compared with the HA groups (fold change > 1.5; p value < 0.025) including IL-12p40 and IL-12p70. We note that, sampling at two timepoints, intra-individual cytokine abundance patterns clustered in 86% of all 60 cases, indicative of person-specific, highly controlled multi-cytokine signatures in blood plasma. Twenty-three urinary proteins were differentially abundant (HA versus NHA; fold change > 1.5; p value < 0.01). Among the proteins increased in abundance in the HA cohort were glycoprotein MUC18, ephrin type-B receptor 4, matrix remodeling-associated protein 8, angiopoietin-related protein 2, K-cadherin, and plasma protease C1 inhibitor. These proteins have been linked to the extracellular matrix, cell adhesion, and vascular remodeling and repair processes. In silico network analysis identified the regulation of coagulation, antimicrobial humoral immune responses, and the IL-12 signaling pathway as enriched GO terms. To validate links of these preliminary biomarkers and IL-12 signaling with healthy aging, clinical studies using larger cohorts and functional characterization of the genes/proteins in cellular models of aging need to be conducted.
Collapse
Affiliation(s)
- Yanbao Yu
- J. Craig Venter Institute, 9605 Medical Center Drive, Rockville, MD, 20850, USA
| | - Harinder Singh
- J. Craig Venter Institute, 9605 Medical Center Drive, Rockville, MD, 20850, USA
| | - Keehwan Kwon
- J. Craig Venter Institute, 9605 Medical Center Drive, Rockville, MD, 20850, USA
| | - Tamara Tsitrin
- J. Craig Venter Institute, 9605 Medical Center Drive, Rockville, MD, 20850, USA
| | - Joann Petrini
- Western Connecticut Health Network, 24 Hospital Avenue, Danbury, CT, 06810, USA
| | - Karen E Nelson
- J. Craig Venter Institute, 9605 Medical Center Drive, Rockville, MD, 20850, USA
- J. Craig Venter Institute, 4120 Capricorn Lane, La Jolla, CA, 92037, USA
| | - Rembert Pieper
- J. Craig Venter Institute, 9605 Medical Center Drive, Rockville, MD, 20850, USA.
| |
Collapse
|
28
|
Ye J, Wang Y, Wang Z, Lin Y, Liu L, Zhou Q, Wang M, Xu Y, Ye D, Zhang J, Wan J, Ji Q. Circulating IL-37 levels are elevated in patients with hypertension. Exp Ther Med 2021; 21:558. [PMID: 33850530 DOI: 10.3892/etm.2021.9990] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Accepted: 07/23/2020] [Indexed: 12/12/2022] Open
Abstract
Interleukin-37 (IL-37) has been reported to be closely linked to vascular diseases, including atherosclerosis and aortic calcification. The present study aimed to assess the expression levels of IL-37 in patients with hypertension. Blood samples were collected from control subjects (n=20) and patients with hypertension (n=45). Subsequently, macrophages, lymphocytes and dendritic cells were individually isolated and the mRNA expression of IL-37 was measured. In addition, the circulating IL-37 levels in control subjects (n=30) and patients with hypertension (n=334) were assessed. Furthermore, all patients who were subjected to detection of circulating IL-37 underwent ambulatory blood pressure monitoring. The results suggested that the mRNA levels of IL-37 in macrophages, but not in lymphocytes and dendritic cells, isolated from patients with hypertension were markedly elevated compared with those in cells isolated from control subjects. Circulating IL-37 levels were increased in patients with hypertension compared with those in control subjects and positively correlated with systolic and diastolic blood pressure in patients with hypertension. No differences were observed between patients with dipper hypertension and patients with non-dipper hypertension. In addition, patients with hypertension with a smoking habit, type 2 diabetes mellitus and carotid atherosclerotic plaque (CAP) exhibited higher IL-37 levels. IL-37 levels were positively correlated with creatinine, C-reactive protein and homocysteine levels. Furthermore, the results of a linear regression analysis suggested that IL-37 levels were independently associated with the presence of CAP. In conclusion, IL-37 levels are increased in patients with hypertension and may be associated with the onset of CAP.
Collapse
Affiliation(s)
- Jing Ye
- Department of Cardiology, People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi 530021, P.R. China.,Department of Cardiology, Renmin Hospital of Wuhan University, Hubei Key Laboratory of Cardiology, Cardiovascular Research Institute, Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Yuan Wang
- Department of Thyroid Breast Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Zhen Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Hubei Key Laboratory of Cardiology, Cardiovascular Research Institute, Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Yingzhong Lin
- Department of Cardiology, People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi 530021, P.R. China
| | - Ling Liu
- Department of Cardiology, People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi 530021, P.R. China
| | - Qi Zhou
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Key Laboratory of Remodeling-Related Cardiovascular Disease, Ministry of Education, Beijing 100029, P.R. China
| | - Menglong Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Hubei Key Laboratory of Cardiology, Cardiovascular Research Institute, Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Yao Xu
- Department of Cardiology, Renmin Hospital of Wuhan University, Hubei Key Laboratory of Cardiology, Cardiovascular Research Institute, Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Di Ye
- Department of Cardiology, Renmin Hospital of Wuhan University, Hubei Key Laboratory of Cardiology, Cardiovascular Research Institute, Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Jishou Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University, Hubei Key Laboratory of Cardiology, Cardiovascular Research Institute, Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Jun Wan
- Department of Cardiology, Renmin Hospital of Wuhan University, Hubei Key Laboratory of Cardiology, Cardiovascular Research Institute, Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Qingwei Ji
- Department of Cardiology, People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi 530021, P.R. China
| |
Collapse
|
29
|
Olivencia MA, Martínez-Casales M, Peraza DA, García-Redondo AB, Mondéjar-Parreño G, Hernanz R, Salaices M, Cogolludo A, Pennington MW, Valenzuela C, Briones AM. K V 1.3 channels are novel determinants of macrophage-dependent endothelial dysfunction in angiotensin II-induced hypertension in mice. Br J Pharmacol 2021; 178:1836-1854. [PMID: 33556997 DOI: 10.1111/bph.15407] [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: 07/16/2020] [Revised: 01/28/2021] [Accepted: 01/31/2021] [Indexed: 11/27/2022] Open
Abstract
BACKGROUND AND PURPOSE KV 1.3 channels are expressed in vascular smooth muscle cells (VSMCs), where they contribute to proliferation rather than contraction and participate in vascular remodelling. KV 1.3 channels are also expressed in macrophages, where they assemble with KV 1.5 channels (KV 1.3/KV 1.5), whose activation generates a KV current. In macrophages, the KV 1.3/KV 1.5 ratio is increased by classical activation (M1). Whether these channels are involved in angiotensin II (AngII)-induced vascular remodelling, and whether they can modulate the macrophage phenotype in hypertension, remains unknown. We characterized the role of KV 1.3 channels in vascular damage in hypertension. EXPERIMENTAL APPROACH We used AngII-infused mice treated with two selective KV 1.3 channel inhibitors (HsTX[R14A] and [EWSS]ShK). Vascular function and structure were measured using wire and pressure myography, respectively. VSMC and macrophage electrophysiology were studied using the patch-clamp technique; gene expression was analysed using RT-PCR. KEY RESULTS AngII increased KV 1.3 channel expression in mice aorta and peritoneal macrophages which was abolished by HsTX[R14A] treatment. KV 1.3 inhibition did not prevent hypertension, vascular remodelling, or stiffness but corrected AngII-induced macrophage infiltration and endothelial dysfunction in the small mesenteric arteries and/or aorta, via a mechanism independent of electrophysiological changes in VSMCs. AngII modified the electrophysiological properties of peritoneal macrophages, indicating an M1-like activated state, with enhanced expression of proinflammatory cytokines that induced endothelial dysfunction. These effects were prevented by KV 1.3 blockade. CONCLUSIONS AND IMPLICATIONS We unravelled a new role for KV 1.3 channels in the macrophage-dependent endothelial dysfunction induced by AngII in mice which might be due to modulation of macrophage phenotype.
Collapse
Affiliation(s)
- Miguel A Olivencia
- Departamento de Farmacología, Universidad Autónoma de Madrid, Instituto de Investigación Hospital La Paz, Madrid, Spain.,Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense de Madrid, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain.,Ciber de Enfermedades Respiratorias (CIBERES), Spain
| | - Marta Martínez-Casales
- Departamento de Farmacología, Universidad Autónoma de Madrid, Instituto de Investigación Hospital La Paz, Madrid, Spain.,Departamento de Ciencias Básicas de la Salud, Facultad de Ciencias de la Salud, Universidad Rey Juan Carlos, Alcorcón, Spain
| | - Diego A Peraza
- Instituto de Investigaciones Biomédicas Alberto Sols (CSIC-UAM), Madrid, Spain
| | - Ana B García-Redondo
- Departamento de Farmacología, Universidad Autónoma de Madrid, Instituto de Investigación Hospital La Paz, Madrid, Spain.,Ciber de Enfermedades Cardiovasculares (CIBERCV), Spain
| | - Gema Mondéjar-Parreño
- Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense de Madrid, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain.,Ciber de Enfermedades Respiratorias (CIBERES), Spain
| | - Raquel Hernanz
- Departamento de Ciencias Básicas de la Salud, Facultad de Ciencias de la Salud, Universidad Rey Juan Carlos, Alcorcón, Spain.,Ciber de Enfermedades Cardiovasculares (CIBERCV), Spain
| | - Mercedes Salaices
- Departamento de Farmacología, Universidad Autónoma de Madrid, Instituto de Investigación Hospital La Paz, Madrid, Spain.,Ciber de Enfermedades Cardiovasculares (CIBERCV), Spain
| | - Angel Cogolludo
- Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense de Madrid, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain.,Ciber de Enfermedades Respiratorias (CIBERES), Spain
| | | | - Carmen Valenzuela
- Instituto de Investigaciones Biomédicas Alberto Sols (CSIC-UAM), Madrid, Spain.,Ciber de Enfermedades Cardiovasculares (CIBERCV), Spain
| | - Ana M Briones
- Departamento de Farmacología, Universidad Autónoma de Madrid, Instituto de Investigación Hospital La Paz, Madrid, Spain.,Ciber de Enfermedades Cardiovasculares (CIBERCV), Spain
| |
Collapse
|
30
|
Zhou Q, Cheng W, Wang Z, Liu J, Han J, Wen S, Liu J. C1q/TNF-related protein-9 is elevated in hypertension and associated with the occurrence of hypertension-related atherogenesis. Cell Biol Int 2021; 45:989-1000. [PMID: 33377578 DOI: 10.1002/cbin.11542] [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: 07/29/2020] [Revised: 11/02/2020] [Accepted: 12/22/2020] [Indexed: 11/06/2022]
Abstract
C1q-tumor necrosis factor-related protein-9 (CTRP9) is an important adipocytokine that is closely associated with cardiovascular disease. This study aimed to detect CTRP9 expression in hypertensive patients and mice and to analyze its effects on hypertension-related atherogenesis. First, circulating CTRP9 levels were detected in both nonhypertensive subjects and hypertensive patients. The results showed that plasma CTRP9 levels were increased in hypertension patients compared with control subjects and gradually elevated in the Grade I, Grade II, and Grade III groups. While nondipper state did not affect CTRP9 expression in hypertension patients. Hypertension patients with carotid atherosclerotic plaque (CAP) exhibited higher CTRP9 levels and the high CTRP9 group exhibited significantly higher CAP morbidity, CTRP9 levels were positively correlated with the occurrence of CAP. Then, effects of CTRP9 on angiotensin II (Ang II)-induced endothelial dysfunction were analyzed in vitro, and the results exhibited that treatment with Ang II significantly increased CTRP9 mRNA expression in endothelial cells (ECs), and downregulation of CTRP9 expression aggravated Ang II-induced endothelial dysfunction in ECs. Mice were infused with Ang II, and CTRP9 was also increased in Ang II-infused mice and mainly secreted by ECs. In Ang II-infused ApoE-/- mice, treatment with recombinant CTRP9 significantly reduced atherosclerotic area and alleviated endothelial dysfunction. In conclusion, our results may found that CTRP9 delayed the progression of hypertension-related arteriosclerosis by alleviating endothelial dysfunction.
Collapse
Affiliation(s)
- Qi Zhou
- Department of Hypertension, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Wenli Cheng
- Department of Hypertension, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Zuoguang Wang
- Department of Hypertension Research, Beijing Anzhen Hospital, Capital Medical University and Beijing Institute of Heart Lung and Blood Vessel Disease, Beijing, China
| | - Jielin Liu
- Department of Hypertension Research, Beijing Anzhen Hospital, Capital Medical University and Beijing Institute of Heart Lung and Blood Vessel Disease, Beijing, China
| | - Jing Han
- Department of Hypertension Research, Beijing Anzhen Hospital, Capital Medical University and Beijing Institute of Heart Lung and Blood Vessel Disease, Beijing, China
| | - Shaojun Wen
- Department of Hypertension Research, Beijing Anzhen Hospital, Capital Medical University and Beijing Institute of Heart Lung and Blood Vessel Disease, Beijing, China
| | - Jinghua Liu
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
31
|
Crouch SH, Botha-Le Roux S, Delles C, Graham LA, Schutte AE. Inflammation and hypertension development: A longitudinal analysis of the African-PREDICT study. INTERNATIONAL JOURNAL CARDIOLOGY HYPERTENSION 2020; 7:100067. [PMID: 33392493 PMCID: PMC7768897 DOI: 10.1016/j.ijchy.2020.100067] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Revised: 10/31/2020] [Accepted: 11/06/2020] [Indexed: 12/20/2022]
Abstract
Background The role of inflammation in the development of hypertension remains incompletely understood. While single inflammatory mediators have been shown to associate with changes in blood pressure (ΔBP), the role of clusters of inflammatory mediators has been less comprehensively explored. We therefore determined whether individual or clusters of inflammatory mediators from a large biomarker panel were associated with ΔBP over 4.5 years, in young healthy adults. Methods We included 358 adults (white, n = 156; black, n = 202) with detailed information on ambulatory blood pressure (BP) at baseline and follow-up. Baseline blood samples were analysed for 22 inflammatory mediators using multiplexing technology. Principal component analysis was used to study associations between clusters of inflammatory mediators and ΔBP. Results In the total cohort in multivariable-adjusted regression analyses, percentage change in 24hr systolic BP associated positively with Factors 1 (Interferon-gamma, interleukin (IL)-4, IL-7, IL-10, IL-12, IL-17A, IL-21, IL-23, macrophage inflammatory protein (MIP)-1α, MIP-1β, TNF-α, granulocyte-macrophage colony-stimulating factor (GM-CSF)) and 2 (IL-5, IL-6, IL-8, IL-13). Change in daytime systolic BP associated positively with Factors 1, 2 and 3 (C-Reactive protein, IL-1β, IL-2, MIP-3α). Subgroup analysis found these findings were limited to white study participants. Numerous associations were present between individual inflammatory mediators (Interferon-gamma, GM-CSF, IL-4, IL-6, IL-7, IL-8, IL-10, IL-12, IL-13, IL-17A, IL-21, IL-23, MIP-1α and MIP-1β) and ΔBP in the white but not black subgroups. Conclusion We found independent relationships between numerous inflammatory mediators (individual and clusters) and ΔBP over 4.5 years. The relationship between inflammatory markers and ΔBP was only found in white participants. ClinicalTrials.gov (Identifier: NCT03292094)..
Collapse
Affiliation(s)
- Simone H. Crouch
- Hypertension in Africa Research Team (HART), North-West University, Potchefstroom, South Africa
| | - Shani Botha-Le Roux
- Hypertension in Africa Research Team (HART), North-West University, Potchefstroom, South Africa
- MRC Research Unit: Hypertension and Cardiovascular Disease, North-West University, Potchefstroom, South Africa
| | - Christian Delles
- The British Heart Foundation Centre of Excellence, Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary, and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Lesley A. Graham
- The British Heart Foundation Centre of Excellence, Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary, and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Aletta E. Schutte
- Hypertension in Africa Research Team (HART), North-West University, Potchefstroom, South Africa
- MRC Research Unit: Hypertension and Cardiovascular Disease, North-West University, Potchefstroom, South Africa
- School of Population Health, University of New South Wales; The George Institute for Global Health, Sydney, Australia
- Corresponding author. School of Population Health, UNSW Medicine, University of New South Wales, NSW 2052, Australia.
| |
Collapse
|
32
|
Liang W, Li J, Bai C, Chen Y, Li Y, Huang G, Wang X. Interleukin-5 deletion promotes sepsis-induced M1 macrophage differentiation, deteriorates cardiac dysfunction, and exacerbates cardiac injury via the NF-κB p65 pathway in mice. Biofactors 2020; 46:1006-1017. [PMID: 33043521 DOI: 10.1002/biof.1681] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Revised: 08/26/2020] [Accepted: 09/16/2020] [Indexed: 12/22/2022]
Abstract
Inflammation plays a crucial role in sepsis-induced cardiac injury. The purpose of this study was to determine whether interleukin-5 (IL-5) affected lipopolysaccharide (LPS)-induced cardiac injury by regulating the inflammatory response. First, the expression level and source of cardiac IL-5 were examined, and the results showed that LPS treatment and cecal ligation decreased cardiac IL-5 expression in macrophages. In addition, LPS was used to establish a mouse sepsis model, and the effects of IL-5 deletion on cardiac injury, M1 macrophage differentiation and myocardial cell apoptosis were analyzed. The results showed that IL-5 deficiency significantly increased cardiac injury marker expression, worsened cardiac dysfunction, promoted M1 macrophage differentiation and exacerbated myocardial cell apoptosis in LPS-induced septic mice. The nuclear factor-kappa B (NF-κB) p65 pathway was inhibited by JSH-23, and the results showed that treatment with JSH-23 inhibited M1 macrophage differentiation and alleviated cardiac injury in LPS-treated IL-5-knockout mice. Furthermore, the effects of IL-5 deficiency on M1 macrophage differentiation and myocardial cell apoptosis were measured in vitro. The IL-5-mediated promotion of M1 macrophage differentiation was also reversed by S31-201, and the pro-apoptotic effect of IL-5 knockout on macrophage-mediated myocardial cell apoptosis was also reversed by JSH-23. In conclusion, we found that IL-5 knockout may exacerbate sepsis-induced cardiac injury by promoting M1 macrophage differentiation in mice. IL-5 may be a potential target for the clinical prevention of sepsis-related cardiac injury.
Collapse
Affiliation(s)
- Wanqian Liang
- Department of Cardiology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, China
| | - Jianhua Li
- Department of Cardiology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, China
| | - Caiyan Bai
- Department of Cardiology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, China
| | - Yingen Chen
- Department of Cardiology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, China
| | - Yan Li
- Department of Cardiology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, China
| | - Guotao Huang
- Department of Cardiology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, China
| | - Xuehui Wang
- Department of Cardiology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, China
| |
Collapse
|
33
|
Wang Y, Zhu S, Wei W, Tu Y, Chen C, Song J, Li J, Wang C, Xu Z, Sun S. Interleukin-6 knockout reverses macrophage differentiation imbalance and alleviates cardiac dysfunction in aging mice. Aging (Albany NY) 2020; 12:20184-20197. [PMID: 33099539 PMCID: PMC7655174 DOI: 10.18632/aging.103749] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Accepted: 06/01/2020] [Indexed: 12/18/2022]
Abstract
Several interleukins (ILs) have been shown to be involved in aging, but the effects of IL-6 on aging-related cardiac dysfunction remain unknown. In this study, the expression and sources of cardiac IL-6 in aging hearts were investigated for the first time. The results showed that cardiac IL-6 expression in mice gradually increased with age, and the expression at 16 months, 20 months and 25 months was higher than that at 3 months. In addition, cardiac macrophages (Møs) were shown to be the main sources of IL-6 in aging mice. IL-6 knockout (KO) significantly alleviated cardiac dysfunction, increased M2 macrophage (Mø2) differentiation, reduced M1 macrophage (Mø1) differentiation and protected against cardiomyocyte apoptosis in aging mice. IL-6 KO also reversed the stimulatory effect of doxorubicin (DOX) treatment on Mø1s and the inhibitory effect of DOX treatment on Mø2s in vitro. Furthermore, the mRNA expression of both aging markers and apoptosis-related markers was markedly inhibited by IL-6 KO. Our results suggest that aging can be significantly reversed by IL-6 KO and that the mechanisms of this effect are related to alleviation of Mø1/Mø2 imbalance and protection against apoptosis in cardiomyocytes.
Collapse
Affiliation(s)
- Yuan Wang
- Department of Thyroid Breast Surgery, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Shan Zhu
- Department of Thyroid Breast Surgery, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Wen Wei
- Department of Thyroid Breast Surgery, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Yi Tu
- Department of Thyroid Breast Surgery, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Chuang Chen
- Department of Thyroid Breast Surgery, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Junlong Song
- Department of Thyroid Breast Surgery, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Juanjuan Li
- Department of Thyroid Breast Surgery, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Changhua Wang
- Basic Medical School of Wuhan University, Wuhan 430060, China
| | - Zhiliang Xu
- Department of Thyroid Breast Surgery, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Shengrong Sun
- Department of Thyroid Breast Surgery, Renmin Hospital of Wuhan University, Wuhan 430060, China
| |
Collapse
|
34
|
Qi L, Wu K, Shi S, Ji Q, Miao H, Bin Q. Thrombospondin-2 is upregulated in patients with aortic dissection and enhances angiotensin II-induced smooth muscle cell apoptosis. Exp Ther Med 2020; 20:150. [PMID: 33093888 PMCID: PMC7571314 DOI: 10.3892/etm.2020.9279] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Accepted: 04/01/2020] [Indexed: 12/16/2022] Open
Abstract
Thrombospondin-2 (TSP-2) is an important extracellular matrix protein that is involved in a variety of cardiovascular diseases, including viral myocarditis and abdominal aortic aneurysm. The present study aimed to investigate TSP-2 expression in patients with aortic dissection (AD). Aortas were obtained from patients with AD and healthy donors, and TSP-2 expression level in all samples was measured by western blotting and immunofluorescence assays. Blood samples were also collected from patients with AD and non-AD (NAD) subjects. Circulating TSP-2, tumor necrosis factor (TNF)-α and interleukin (IL)-6 levels in each sample were detected using ELISAs. In addition, the effect of TSP-2 on angiotensin II (Ang II)-induced smooth muscle cell (SMC) apoptosis was assessed in vitro. Compared with healthy donors, aortic TSP-2 expression level was significantly increased in patients with AD. Furthermore, TSP-2 was secreted primarily by SMCs, but also by endothelial cells. TSP-2 plasma expression level was also elevated in patients with AD compared with non-AD subjects. Furthermore, TSP-2 serum expression level was positively correlated with TNF-α and IL-6 expression levels in patients with AD. In addition, recombinant mouse TSP-2 treatment increased Bax mRNA expression and decreased Bcl2 mRNA expression in Ang II-treated SMCs; however, the effects were reversed following treatment with the NF-κB p65 signaling pathway inhibitor JSH-23 or with the anti-TNF-α and anti-IL-6 neutralizing antibodies. The present study demonstrated that TSP-2 expression was increased in the aortic tissues and plasma of patients with AD. These findings suggested that TSP-2 may participate in the progression of AD by activating the NF-κB p65 signaling pathway and amplifying the inflammatory response.
Collapse
Affiliation(s)
- Liping Qi
- Department of Cardiology, The Second Clinical Center, Chinese People's Liberation Army General Hospital, Beijing 100853, P.R. China
| | - Kui Wu
- Emergency and Critical Care Center, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung, and Blood Vessel Diseases, Beijing Lab for Cardiovascular Precision Medicine, Beijing 100029, P.R. China
| | - Shutian Shi
- Emergency and Critical Care Center, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung, and Blood Vessel Diseases, Beijing Lab for Cardiovascular Precision Medicine, Beijing 100029, P.R. China
| | - Qingwei Ji
- Emergency and Critical Care Center, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung, and Blood Vessel Diseases, Beijing Lab for Cardiovascular Precision Medicine, Beijing 100029, P.R. China
| | - Huangtai Miao
- Emergency and Critical Care Center, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung, and Blood Vessel Diseases, Beijing Lab for Cardiovascular Precision Medicine, Beijing 100029, P.R. China
| | - Que Bin
- Emergency and Critical Care Center, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung, and Blood Vessel Diseases, Beijing Lab for Cardiovascular Precision Medicine, Beijing 100029, P.R. China
| |
Collapse
|
35
|
Yu Y, Singh H, Kwon K, Tsitrin T, Petrini J, Nelson KE, Pieper R. Protein signatures from blood plasma and urine suggest changes in vascular function and IL-12 signaling in elderly with a history of chronic diseases compared with an age-matched healthy cohort. GeroScience 2020; 43:593-606. [PMID: 32974878 PMCID: PMC8110643 DOI: 10.1007/s11357-020-00269-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Accepted: 09/14/2020] [Indexed: 01/02/2023] Open
Abstract
Key processes characterizing human aging are immunosenescence and inflammaging. The capacity of the immune system to adequately respond to external perturbations (e.g., pathogens, injuries, and biochemical irritants) and to repair somatic mutations that may cause cancers or cellular senescence declines. An important goal remains to identify genetic or biochemical, predictive biomarkers for healthy aging. We recruited two cohorts in the age range 70 to 82, one afflicted by chronic illnesses (non-healthy aging, NHA) and the other in good health (healthy aging, HA). NHA criteria included major cardiovascular, neurodegenerative, and chronic pulmonary diseases, diabetes, and cancers. Quantitative analysis of forty proinflammatory cytokines in blood plasma and more than 500 proteins in urine was performed to identify candidate biomarkers for and biological pathway implications of healthy aging. Nine cytokines revealed lower quantities in blood plasma for the NHA compared with the HA groups (fold change > 1.5; p value < 0.025) including IL-12p40 and IL-12p70. We note that, sampling at two timepoints, intra-individual cytokine abundance patterns clustered in 86% of all 60 cases, indicative of person-specific, highly controlled multi-cytokine signatures in blood plasma. Twenty-three urinary proteins were differentially abundant (HA versus NHA; fold change > 1.5; p value < 0.01). Among the proteins increased in abundance in the HA cohort were glycoprotein MUC18, ephrin type-B receptor 4, matrix remodeling-associated protein 8, angiopoietin-related protein 2, K-cadherin, and plasma protease C1 inhibitor. These proteins have been linked to the extracellular matrix, cell adhesion, and vascular remodeling and repair processes. In silico network analysis identified the regulation of coagulation, antimicrobial humoral immune responses, and the IL-12 signaling pathway as enriched GO terms. To validate links of these preliminary biomarkers and IL-12 signaling with healthy aging, clinical studies using larger cohorts and functional characterization of the genes/proteins in cellular models of aging need to be conducted.
Collapse
Affiliation(s)
- Yanbao Yu
- J. Craig Venter Institute, 9605 Medical Center Drive, Rockville, MD, 20850, USA
| | - Harinder Singh
- J. Craig Venter Institute, 9605 Medical Center Drive, Rockville, MD, 20850, USA
| | - Keehwan Kwon
- J. Craig Venter Institute, 9605 Medical Center Drive, Rockville, MD, 20850, USA
| | - Tamara Tsitrin
- J. Craig Venter Institute, 9605 Medical Center Drive, Rockville, MD, 20850, USA
| | - Joann Petrini
- Western Connecticut Health Network, 24 Hospital Avenue, Danbury, CT, 06810, USA
| | - Karen E Nelson
- J. Craig Venter Institute, 9605 Medical Center Drive, Rockville, MD, 20850, USA.,J. Craig Venter Institute, 4120 Capricorn Lane, La Jolla, CA, 92037, USA
| | - Rembert Pieper
- J. Craig Venter Institute, 9605 Medical Center Drive, Rockville, MD, 20850, USA.
| |
Collapse
|
36
|
Ye J, Wang Y, Xu Y, Wang Z, Liu L, Wang M, Ye D, Zhang J, Yang Z, Lin Y, Ji Q, Wan J. Interleukin-22 deficiency alleviates doxorubicin-induced oxidative stress and cardiac injury via the p38 MAPK/macrophage/Fizz3 axis in mice. Redox Biol 2020; 36:101636. [PMID: 32863209 PMCID: PMC7371904 DOI: 10.1016/j.redox.2020.101636] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 06/26/2020] [Accepted: 07/03/2020] [Indexed: 02/07/2023] Open
Abstract
Several interleukin (IL) family members have been demonstrated to be involved in doxorubicin (DOX)-induced cardiac injury. This study aimed to investigate the role of IL-22 in DOX-induced cardiac injury and explore its possible mechanisms. In this study, mice were given DOX, and the cardiac expression and sources of IL-22 were determined. Then, IL-22 was knocked out to observe the effects on DOX-induced cardiac injury in mice. In addition, the p38 mitogen-activated protein kinase (MAPK) pathway was inhibited, macrophages were depleted and adoptively transferred, and Fizz3 was up-regulated in mice to explore the mechanisms. The results showed that cardiac IL-22 expression was significantly increased by DOX treatment and was mostly derived from cardiac macrophages. IL-22 knockout significantly reduced cardiac vacuolization and the expression of cardiomyocyte injury markers in both serum and left ventricular tissue and improved cardiac function in DOX-treated mice. In addition, IL-22 knockout reversed DOX-induced cardiac M1 macrophage/M2 macrophage imbalance, reduced oxidative stress and protected against cardiomyocyte apoptosis. p38 MAPK pathway inhibition with SB203580 and macrophage depletion further alleviated the above effects in DOX-treated IL-22-knockout mice. The effects were stronger IL-22-knockout mice with adoptive transfer of WT macrophages than in those with adoptive transfer of IL-22-knockout macrophages. Furthermore, increasing the expression of Fizz3 reduced cardiomyocyte apoptosis and alleviated cardiac dysfunction. Our results may suggest that IL-22 knockout alleviate DOX-induced oxidative stress and cardiac injury by inhibiting macrophage differentiation and thereby increasing the expression of Fizz3. Reductions in IL-22 expression may be beneficial for clinical chemotherapy in tumor patients.
Collapse
Affiliation(s)
- Jing Ye
- Department of Cardiology, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, 530021, China; Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute, Wuhan University, Hubei Key Laboratory of Cardiology, Wuhan, 430060, China
| | - Yuan Wang
- Department of Thyroid Breast Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Yao Xu
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute, Wuhan University, Hubei Key Laboratory of Cardiology, Wuhan, 430060, China
| | - Zhen Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute, Wuhan University, Hubei Key Laboratory of Cardiology, Wuhan, 430060, China
| | - Ling Liu
- Department of Cardiology, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, 530021, China
| | - Menglong Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute, Wuhan University, Hubei Key Laboratory of Cardiology, Wuhan, 430060, China
| | - Di Ye
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute, Wuhan University, Hubei Key Laboratory of Cardiology, Wuhan, 430060, China
| | - Jishou Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute, Wuhan University, Hubei Key Laboratory of Cardiology, Wuhan, 430060, China
| | - Zicong Yang
- Department of Cardiology, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, 530021, China
| | - Yingzhong Lin
- Department of Cardiology, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, 530021, China.
| | - Qingwei Ji
- Department of Cardiology, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, 530021, China.
| | - Jun Wan
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute, Wuhan University, Hubei Key Laboratory of Cardiology, Wuhan, 430060, China.
| |
Collapse
|
37
|
Ye J, Wang Y, Wang Z, Liu L, Yang Z, Wang M, Xu Y, Ye D, Zhang J, Zhou Q, Lin Y, Ji Q, Wan J. The Expression of IL-12 Family Members in Patients with Hypertension and Its Association with the Occurrence of Carotid Atherosclerosis. Mediators Inflamm 2020; 2020:2369279. [PMID: 32322161 PMCID: PMC7165332 DOI: 10.1155/2020/2369279] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 02/13/2020] [Accepted: 02/27/2020] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND The interleukin-12 (IL-12) family consists of four members, namely, IL-12, IL-23, IL-27, and IL-35. The aim of this study was to examine the expression of circulating IL-12, IL-23, IL-27, and IL-35 in hypertensive patients. METHODS Blood samples were collected from hypertensive patients and nonhypertensive (control) subjects, and protein multifactorial monitor kits were used to measure the plasma IL-12, IL-23, IL-27, and IL-35 levels in each sample. In addition, all enrolled subjects underwent ambulatory blood pressure monitoring (ABPM) and vascular stiffness. RESULTS Hypertensive patients exhibited higher IL-12, IL-23, and IL-27 levels and lower IL-35 levels than control subjects; IL-12, IL-23, and IL-27 levels were positively correlated with both systolic blood pressure (SBP) and diastolic blood pressure (DBP), while IL-35 levels were negatively correlated with SBP and DBP. IL-12, IL-23, and IL-27 levels gradually increased in patients with grade I, II, and III hypertension, while IL-35 levels gradually reduced. According to the ABPM results, hypertensive patients were divided into the dipper and nondipper hypertension groups; IL-12, IL-23, IL-27, and IL-35 levels showed no differences between the two groups, but IL-12, IL-23, and IL-27 levels in both groups increased compared with those in the control group, while IL-35 levels decreased. Additionally, the expression of these IL-12 family members was influenced by many clinical factors and was independently associated with the occurrence of carotid atherosclerotic plaques. CONCLUSIONS The changes in IL-12, IL-23, IL-27, and IL-35 levels were not associated with the presence of the nondipper type but were closely associated with the development of carotid atherosclerotic plaque in hypertensive patients.
Collapse
Affiliation(s)
- Jing Ye
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute, Wuhan University, Hubei Key Laboratory of Cardiology, Wuhan 430060, China
| | - Yuan Wang
- Department of Thyroid Breast Surgery, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Zhen Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute, Wuhan University, Hubei Key Laboratory of Cardiology, Wuhan 430060, China
| | - Ling Liu
- Department of Cardiology, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning 530021, China
| | - Zicong Yang
- Department of Cardiology, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning 530021, China
| | - Menglong Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute, Wuhan University, Hubei Key Laboratory of Cardiology, Wuhan 430060, China
| | - Yao Xu
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute, Wuhan University, Hubei Key Laboratory of Cardiology, Wuhan 430060, China
| | - Di Ye
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute, Wuhan University, Hubei Key Laboratory of Cardiology, Wuhan 430060, China
| | - Jishou Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute, Wuhan University, Hubei Key Laboratory of Cardiology, Wuhan 430060, China
| | - Qi Zhou
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung, and Blood Vessel Diseases, The Key Laboratory of Remodeling-Related Cardiovascular Disease, Ministry of Education, Beijing 100029, China
| | - Yingzhong Lin
- Department of Cardiology, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning 530021, China
| | - Qingwei Ji
- Department of Cardiology, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning 530021, China
| | - Jun Wan
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute, Wuhan University, Hubei Key Laboratory of Cardiology, Wuhan 430060, China
| |
Collapse
|
38
|
Xian D, Zhan Y, Yang Z, Fan C, Liu L, Lin Y. Anti-interleukin-5-neutralizing antibody attenuates caradiac injury and cadiac dysfunction by aggravating the inflammatory response in doxorubicin-treated mice. Cell Biol Int 2020; 44:1363-1372. [PMID: 32125042 DOI: 10.1002/cbin.11330] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2019] [Accepted: 03/01/2020] [Indexed: 12/20/2022]
Abstract
Previous studies have demonstrated that interleukins (ILs) are closely associated with doxorubicin (DOX)-induced cardiac injury. IL-5 is an important member of the IL family, and this study was performed to investigate whether IL-5 affects DOX-induced cardiac injury and its underlying mechanisms. The cardiac IL-5 expression was first detected and the results showed that cardiac IL-5 levels were significantly lower in DOX-treated mice, and IL-5 was mainly derived from cardiac macrophage (Mø). In addition, some DOX-treated mice received an injection of anti-IL-5-neutralizing antibody (nAb), and we found that treatment with a mouse anti-IL-5 nAb significantly upregulated the levels of myocardial injury markers, aggravated cardiac dysfunction, increased M1 macrophage (Mø1) and decreased M2 macrophage (Mø2) differentiation, and promoted apoptotic marker expression. Furthermore, the effect of mouse IL-5 nAb on DOX-induced Mø differentiation and its role on mouse cardiomyocyte (MCM) cells apoptosis were detected in vitro, and the results exhibited that mouse IL-5 nAb promoted Mø1 differentiation but inhibited Mø2 differentiation in vitro and alleviated apoptosis in MCM cells. Our results found a mouse anti-IL-5 nAb-aggravated DOX-induced cardiac injury and dysfunction by alleviating the inflammatory response and myocardial cell apoptosis.
Collapse
Affiliation(s)
- Dan Xian
- Xishu County People's Hospital affiliated to Hubei Univerisity of Science and Technology, Nanning, 530021, China
| | - Yingying Zhan
- Xishu County People's Hospital affiliated to Hubei Univerisity of Science and Technology, Nanning, 530021, China
| | - Zicong Yang
- Department of Cardiology, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, 530021, China
| | - Cao Fan
- Department of Cardiology, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, 530021, China
| | - Ling Liu
- Department of Cardiology, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, 530021, China
| | - Yingzhong Lin
- Department of Cardiology, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, 530021, China
| |
Collapse
|
39
|
Interleukin-9 Deletion Relieves Vascular Dysfunction and Decreases Blood Pressure via the STAT3 Pathway in Angiotensin II-Treated Mice. Mediators Inflamm 2020; 2020:5741047. [PMID: 32148441 PMCID: PMC7042522 DOI: 10.1155/2020/5741047] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Revised: 01/15/2020] [Accepted: 02/03/2020] [Indexed: 11/18/2022] Open
Abstract
Background Multiple interleukin (IL) family members were reported to be closely related to hypertension. We aimed to investigate whether IL-9 affects angiotensin II- (Ang II-) induced hypertension in mice. Methods Mice were treated with Ang II, and IL-9 expression was determined. In addition, effects of IL-9 knockout (KO) on blood pressure were observed in Ang II-infused mice. To determine whether the effects of IL-9 on blood pressure was mediated by the signal transducer and activator of the transcription 3 (STAT3) pathway, Ang II-treated mice were given S31-201. Furthermore, circulating IL-9 levels in patients with hypertension were measured. Results Ang II treatment increased serum and aortic IL-9 expression in a dose-dependent manner; IL-9 levels were the highest in the second week and continued to remain high into the fourth week after the treatment. IL-9 KO downregulated proinflammatory cytokine expression, whereas it upregulated anti-inflammatory cytokine levels, relieved vascular dysfunction, and decreased blood pressure in Ang II-infused mice. IL-9 also reduced smooth muscle 22α (SM22α (SM22 Conclusions IL-9 KO alleviates inflammatory response, prevents phenotypic transformation of smooth muscle, reduces vascular dysfunction, and lowers blood pressure via the STAT3 pathway in Ang II-infused mice. IL-9 might be a novel target for the treatment and prevention of clinical hypertension.
Collapse
|
40
|
Ye J, Wang Y, Wang Z, Liu L, Yang Z, Wang M, Xu Y, Ye D, Zhang J, Lin Y, Ji Q, Wan J. Roles and Mechanisms of Interleukin-12 Family Members in Cardiovascular Diseases: Opportunities and Challenges. Front Pharmacol 2020; 11:129. [PMID: 32194399 PMCID: PMC7064549 DOI: 10.3389/fphar.2020.00129] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Accepted: 01/30/2020] [Indexed: 12/19/2022] Open
Abstract
Cardiovascular diseases represent a complex group of clinical syndromes caused by a variety of interacting pathological factors. They include the most extensive disease population and rank first in all-cause mortality worldwide. Accumulating evidence demonstrates that cytokines play critical roles in the presence and development of cardiovascular diseases. Interleukin-12 family members, including IL-12, IL-23, IL-27 and IL-35, are a class of cytokines that regulate a variety of biological effects; they are closely related to the progression of various cardiovascular diseases, including atherosclerosis, hypertension, aortic dissection, cardiac hypertrophy, myocardial infarction, and acute cardiac injury. This paper mainly discusses the role of IL-12 family members in cardiovascular diseases, and the molecular and cellular mechanisms potentially involved in their action in order to identify possible intervention targets for the prevention and clinical treatment of cardiovascular diseases.
Collapse
Affiliation(s)
- Jing Ye
- Hubei Key Laboratory of Cardiology, Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute, Wuhan University, Wuhan, China
| | - Yuan Wang
- Department of Thyroid Breast Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Zhen Wang
- Hubei Key Laboratory of Cardiology, Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute, Wuhan University, Wuhan, China
| | - Ling Liu
- Department of Cardiology, the People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Zicong Yang
- Department of Cardiology, the People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Menglong Wang
- Hubei Key Laboratory of Cardiology, Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute, Wuhan University, Wuhan, China
| | - Yao Xu
- Hubei Key Laboratory of Cardiology, Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute, Wuhan University, Wuhan, China
| | - Di Ye
- Hubei Key Laboratory of Cardiology, Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute, Wuhan University, Wuhan, China
| | - Jishou Zhang
- Hubei Key Laboratory of Cardiology, Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute, Wuhan University, Wuhan, China
| | - Yingzhong Lin
- Department of Cardiology, the People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Qingwei Ji
- Department of Cardiology, the People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Jun Wan
- Hubei Key Laboratory of Cardiology, Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute, Wuhan University, Wuhan, China
| |
Collapse
|
41
|
Balasubbramanian D, Goodlett BL, Mitchell BM. Is IL-12 pro-inflammatory or anti-inflammatory? Depends on the blood pressure. Cardiovasc Res 2020; 115:998-999. [PMID: 30698673 DOI: 10.1093/cvr/cvz028] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Affiliation(s)
- Dakshnapriya Balasubbramanian
- Department of Medical Physiology, Texas A&M College of Medicine, 361 Reynolds Medical Building, College Station, TX, USA
| | - Bethany L Goodlett
- Department of Medical Physiology, Texas A&M College of Medicine, 361 Reynolds Medical Building, College Station, TX, USA
| | - Brett M Mitchell
- Department of Medical Physiology, Texas A&M College of Medicine, 361 Reynolds Medical Building, College Station, TX, USA
| |
Collapse
|
42
|
Yang Y, Xu C, Tang S, Xia Z. Interleukin-9 Aggravates Isoproterenol-Induced Heart Failure by Activating Signal Transducer and Activator of Transcription 3 Signalling. Can J Cardiol 2020; 36:1770-1781. [PMID: 32621886 DOI: 10.1016/j.cjca.2020.01.011] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2019] [Revised: 12/18/2019] [Accepted: 01/08/2020] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Previous studies have demonstrated that inflammation is closely related to the occurrence and development of heart failure (HF). As an inflammation-related cytokine, interleukin (IL)-9 has been reported to be involved in the development of cardiovascular diseases. However, the role of IL-9 in HF in response to isoproterenol (ISO) stimulation has barely been explored. Thus, this study aimed to investigate whether IL-9 participates in HF and the possible associated mechanisms. METHODS Chronic ISO infusion was used to establish an HF model, and the IL-9 levels in mice and isolated cardiomyocytes were measured. In addition, ISO-treated mice received an injection of recombinant mouse IL-9 (rIL-9) or an antimouse IL-9 neutralizing monoclonal antibody (mAb) to investigate the effects of IL-9 on cardiac function, hypertrophy, and fibrosis. RESULTS IL-9 levels were significantly increased in mice and isolated cardiomyocytes after ISO treatment. Treatment with rIL-9 resulted in aggravated cardiac dysfunction and amplified cardiac hypertrophy and fibrosis, whereas treatment with the anti-IL-9 neutralizing mAb ameliorated cardiac dysfunction and reduced cardiac hypertrophy and fibrosis in ISO-treated mice. In addition, ISO infusion-induced cardiac inflammation and cardiomyocyte apoptosis was aggravated by rIL-9 but prevented by the anti-IL-9 mAb. IL-9 did not activate signal transducer and activator of transcription (STAT)1 or STAT5 but induced STAT3 phosphorylation in ISO-induced HF. Moreover, S31-201, a specific STAT3 inhibitor, nearly abolished rIL-9-induced increases in cardiac dysfunction, hypertrophy, and fibrosis in response to ISO stimulation. CONCLUSIONS IL-9 aggravated cardiac dysfunction and amplified cardiac hypertrophy and fibrosis in the ISO-induced HF model by activating STAT3 signalling. These data indicate that blocking IL-9 may be an attractive pharmacotherapeutic strategy for the treatment of cardiac hypertrophy and fibrosis induced by chronic β-adrenergic receptor activation to limit the progression of HF.
Collapse
Affiliation(s)
- Yunzhao Yang
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Cheng Xu
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Shaoqun Tang
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Zhongyuan Xia
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China.
| |
Collapse
|
43
|
Ye J, Wang Y, Wang Z, Liu L, Yang Z, Ye D, Wang M, Xu Y, Zhang J, Zhao M, Liu J, Lin Y, Ji Q, Wan J. Interleukin-12p35 deficiency enhances mitochondrial dysfunction and aggravates cardiac remodeling in aging mice. Aging (Albany NY) 2020; 12:193-203. [PMID: 31901899 PMCID: PMC6977681 DOI: 10.18632/aging.102609] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Accepted: 12/05/2019] [Indexed: 12/14/2022]
Abstract
Our previous studies have demonstrated that interleukin-12p35 knockout (IL-12p35 KO) regulates the progression of various cardiovascular diseases, such as acute cardiac injury and hypertension. The aims of this study were to investigate whether IL-12p35 KO affects aging-related cardiac remodeling and to explore the possible mechanisms. First, the effects of IL-12p35 KO on heart structure and function were detected, and the results showed that IL-12p35 KO exacerbated cardiac remodeling and increased cardiac senescence-related protein levels in aged mice. In addition, whether IL-12p35 KO regulates cardiac senescence-related protein expression, cardiac mitochondrial dysfunction and cardiomyocyte apoptosis was also investigated. IL-12p35 KO increased mitochondrial calcium fluorescence intensity and ROS fluorescence intensity, while it reduced mitochondrial membrane potential. Furthermore, reduced mitochondrial complex (I-IV) activity and ATP levels and increased apoptosis-inducing factor (AIF)-related cardiomyocyte apoptosis were observed in aged IL-12p35 KO mice compared with wild-type mice. Our results demonstrate that aging is aggravated by IL-12p35 KO and that the mechanism may be related to exacerbation of mitochondrial dysfunction and AIF-related cardiomyocyte apoptosis.
Collapse
Affiliation(s)
- Jing Ye
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute, Wuhan University, Hubei Key Laboratory of Cardiology, Wuhan 430060, China
- Emergency and Critical Care Center, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung, and Blood Vessel Diseases, and Beijing Lab for Cardiovascular Precision Medicine, Beijing 100029, China
- Department of Cardiology, The People’s Hospital of Guangxi Zhuang Autonomous Region, Nanning 530021, China
| | - Yuan Wang
- Department of Thyroid Breast Surgery, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Zhen Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute, Wuhan University, Hubei Key Laboratory of Cardiology, Wuhan 430060, China
| | - Ling Liu
- Department of Cardiology, The People’s Hospital of Guangxi Zhuang Autonomous Region, Nanning 530021, China
| | - Zicong Yang
- Department of Cardiology, The People’s Hospital of Guangxi Zhuang Autonomous Region, Nanning 530021, China
| | - Di Ye
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute, Wuhan University, Hubei Key Laboratory of Cardiology, Wuhan 430060, China
| | - Menglong Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute, Wuhan University, Hubei Key Laboratory of Cardiology, Wuhan 430060, China
| | - Yao Xu
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute, Wuhan University, Hubei Key Laboratory of Cardiology, Wuhan 430060, China
| | - Jishou Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute, Wuhan University, Hubei Key Laboratory of Cardiology, Wuhan 430060, China
| | - Mengmeng Zhao
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute, Wuhan University, Hubei Key Laboratory of Cardiology, Wuhan 430060, China
| | - Jianfang Liu
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute, Wuhan University, Hubei Key Laboratory of Cardiology, Wuhan 430060, China
| | - Yingzhong Lin
- Department of Cardiology, The People’s Hospital of Guangxi Zhuang Autonomous Region, Nanning 530021, China
| | - Qingwei Ji
- Emergency and Critical Care Center, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung, and Blood Vessel Diseases, and Beijing Lab for Cardiovascular Precision Medicine, Beijing 100029, China
- Department of Cardiology, The People’s Hospital of Guangxi Zhuang Autonomous Region, Nanning 530021, China
| | - Jun Wan
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute, Wuhan University, Hubei Key Laboratory of Cardiology, Wuhan 430060, China
| |
Collapse
|
44
|
Abstract
PURPOSE OF REVIEW In recent years, a vast body of evidence has accumulated indicating the role of the immune system in the regulation of blood pressure and modulation of hypertensive pathology. Numerous cells of the immune system, both innate and adaptive immunity, have been indicated to play an important role in the development and maintenance of hypertension. The purpose of this review was to summarize the role of adaptive immunity in experimental models of hypertension (genetic, salt-sensitive, and Angiotensin (Ang) II induced) and in human studies. In particular, the role of T and B cells is discussed. RECENT FINDINGS In response to hypertensive stimuli such as Ang II and high salt, T cells become pro-inflammatory and they infiltrate the brain, blood vessel adventitia and periadventitial fat, heart, and the kidney. Pro-inflammatory T cell-derived cytokines such as IFN-γ and TNF-α (from CD8+ and CD4+Th1) and IL-17A (from the γδ-T cell and CD4+Th17) exacerbate hypertensive responses mediating both endothelial dysfunction and cardiac, renal, and neurodegenerative injury. The modulation of adaptive immune activation in hypertension has been attributed to target organ oxidative stress that leads to the generation of neoantigens, including isolevuglandin-modified proteins. The role of adaptive immunity is sex-specific with much more pronounced mechanisms in males than that in females. Hypertension is also associated with B cell activation and production of autoantibodies (anti-Hsp70, anti-Hsp65, anti-Hsp60, anti-AT1R, anti-α1AR, and anti-β1AR). The hypertensive responses can be inhibited by T regulatory lymphocytes (Tregs) and their anti-inflammatory IL-10. Adaptive immunity and its interface with innate mechanisms may represent valuable targets in the modulation of blood pressure, as well as hypertension-related residual risk.
Collapse
Affiliation(s)
- Tomasz P Mikolajczyk
- Department of Internal and Agricultural Medicine, Faculty of Medicine, Jagiellonian University Medical College, Krakow, Poland
| | - Tomasz J Guzik
- Department of Internal and Agricultural Medicine, Faculty of Medicine, Jagiellonian University Medical College, Krakow, Poland.
- BHF Centre for Excellence, Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK.
| |
Collapse
|