1
|
Jia X, Li L, Wang T, Ma X, Li C, Liu M, Tong H, Wang S. Puerarin inhibits macrophage M1 polarization by combining STAT1 to reduce myocardial damage in EAM model mice. Biochem Biophys Res Commun 2024; 733:150702. [PMID: 39298917 DOI: 10.1016/j.bbrc.2024.150702] [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: 06/06/2024] [Revised: 09/07/2024] [Accepted: 09/13/2024] [Indexed: 09/22/2024]
Abstract
Myocarditis is an inflammatory lesion of the myocardium that is caused by a variety of factors. At present, treatment of symptoms remains the main clinical intervention, but it cannot reduce the myocarditis damage caused by inflammation. M1 macrophages are thought to contribute significantly to the occurrence and development of inflammation by secreting a large number of proinflammatory factors. Puerarin is an isoflavone derivative isolated from pueraria that can be used as a dietary supplement and exerts wide range of anti-inflammatory and antioxidant effects. However, the mechanism underlying its anti-inflammatory effects needs to be further studied. The objective of this study was to investigate whether puerarin inhibited M1 polarization by affecting the JAK-STAT signaling pathway in a mouse model of autoimmune myocarditis, thus inhibiting the occurrence of inflammation in experimental autoimmune myocarditis (EAM) model mice. The results showed that EAM model mice treated with puerarin showed milder clinical symptoms and inflammatory infiltration than EAM model mice. Puerarin suppressed the in vivo and in vitro JAK1/2-STAT1 signal transduction in macrophages, thus inhibiting M1 polarization, reducing the secretion of proinflammatory factors, and ultimately decreasing IFN-γ and TNF-α levels in vivo, which led to myocardial apoptosis. Thus, puerarin could alleviate myocardial damage caused by inflammation. The conclusion of this study was that puerarin reduced myocardial damage in EAM model mice by regulating the polarization of macrophages toward M1, and this inhibitory effect may be achieved by inhibiting JAK1/2-STAT1 signaling.
Collapse
Affiliation(s)
- Xihui Jia
- Department of Biochemistry and Molecular Biology, School of Basic Medical, Qingdao University, Qingdao, China
| | - Ling Li
- School of Basic Medical, Qingdao University, Qingdao, China
| | - Tiantian Wang
- School of Basic Medical, Qingdao University, Qingdao, China
| | - Xiaoran Ma
- Department of Special Medicine, School of Basic Medical, Qingdao University, Qingdao, China
| | - Chenglin Li
- School of Basic Medical, Qingdao University, Qingdao, China
| | - Meng Liu
- Department of Biochemistry and Molecular Biology, School of Basic Medical, Qingdao University, Qingdao, China
| | - Huimin Tong
- School of Basic Medical, Qingdao University, Qingdao, China
| | - Shuang Wang
- Department of Biochemistry and Molecular Biology, School of Basic Medical, Qingdao University, Qingdao, China.
| |
Collapse
|
2
|
Chen Z, Huo X, Huang Y, Cheng Z, Xu X, Li Z. Elevated plasma solMER concentrations link ambient PM 2.5 and PAHs to myocardial injury and reduced left ventricular systolic function in children. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2024; 355:124151. [PMID: 38740242 DOI: 10.1016/j.envpol.2024.124151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/15/2023] [Revised: 05/09/2024] [Accepted: 05/10/2024] [Indexed: 05/16/2024]
Abstract
Exposure to fine particulate matter (PM2.5) and polycyclic aromatic hydrocarbons (PAHs) is known to be associated with the polarization of pro-inflammatory macrophages and the development of various cardiovascular diseases. The pro-inflammatory polarization of resident cardiac macrophages (cMacs) enhances the cleavage of membrane-bound myeloid-epithelial-reproductive receptor tyrosine kinase (MerTK) and promotes the formation of soluble MerTK (solMER). This process influences the involvement of cMacs in cardiac repair, thus leading to an imbalance in cardiac homeostasis, myocardial injury, and reduced cardiac function. However, the relative impacts of PM2.5 and PAHs on human cMacs have yet to be elucidated. In this study, we aimed to investigate the effects of PM2.5 and PAH exposure on solMER in terms of myocardial injury and left ventricular (LV) systolic function in healthy children. A total of 258 children (aged three to six years) were recruited from Guiyu (an area exposed to e-waste) and Haojiang (a reference area). Mean daily PM2.5 concentration data were collected to calculate the individual chronic daily intake (CDI) of PM2.5. We determined concentrations of solMER and creatine kinase MB (CKMB) in plasma, and hydroxylated PAHs (OH-PAHs) in urine. LV systolic function was evaluated by stroke volume (SV). Higher CDI values and OH-PAH concentrations were detected in the exposed group. Plasma solMER and CKMB were higher in the exposed group and were associated with a reduced SV. Elevated CDI and 1-hydroxynaphthalene (1-OHNa) were associated with a higher solMER. Furthermore, increased solMER concentrations were associated with a lower SV and higher CKMB. CDI and 1-OHNa were positively associated with CKMB and mediated by solMER. In conclusion, exposure to PM2.5 and PAHs may lead to the pro-inflammatory polarization of cMacs and increase the risk of myocardial injury and systolic function impairment in children. Furthermore, the pro-inflammatory polarization of cMacs may mediate cardiotoxicity caused by PM2.5 and PAHs.
Collapse
Affiliation(s)
- Zihan Chen
- Department of Cardiology, the First Affiliated Hospital of Shantou University Medical College, Shantou, 515000, Guangdong, China; Shantou University Medical College, Shantou, 15041, Guangdong, China
| | - Xia Huo
- Laboratory of Environmental Medicine and Developmental Toxicology, Guangdong Key Laboratory of Environmental Pollution and Health, School of Environment, Jinan University, Guangzhou, 511443, Guangdong, China
| | - Yu Huang
- Laboratory of Environmental Medicine and Developmental Toxicology, Shantou University Medical College, Shantou, 515041, Guangdong, China
| | - Zhiheng Cheng
- Laboratory of Environmental Medicine and Developmental Toxicology, Shantou University Medical College, Shantou, 515041, Guangdong, China; Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713, GZ, Groningen, the Netherlands
| | - Xijin Xu
- Laboratory of Environmental Medicine and Developmental Toxicology, Shantou University Medical College, Shantou, 515041, Guangdong, China; Department of Cell Biology and Genetics, Shantou University Medical College, Shantou, 515041, Guangdong, China
| | - Zhi Li
- Department of Cardiology, the First Affiliated Hospital of Shantou University Medical College, Shantou, 515000, Guangdong, China.
| |
Collapse
|
3
|
Zheng K, Hao Y, Xia C, Cheng S, Yu J, Chen Z, Li Y, Niu Y, Ran S, Wang S, Ye W, Luo Z, Li X, Zhao J, Li R, Zong J, Zhang H, Lai L, Huang P, Zhou C, Xia J, Zhang X, Wu J. Effects and mechanisms of the myocardial microenvironment on cardiomyocyte proliferation and regeneration. Front Cell Dev Biol 2024; 12:1429020. [PMID: 39050889 PMCID: PMC11266095 DOI: 10.3389/fcell.2024.1429020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Accepted: 06/20/2024] [Indexed: 07/27/2024] Open
Abstract
The adult mammalian cardiomyocyte has a limited capacity for self-renewal, which leads to the irreversible heart dysfunction and poses a significant threat to myocardial infarction patients. In the past decades, research efforts have been predominantly concentrated on the cardiomyocyte proliferation and heart regeneration. However, the heart is a complex organ that comprises not only cardiomyocytes but also numerous noncardiomyocyte cells, all playing integral roles in maintaining cardiac function. In addition, cardiomyocytes are exposed to a dynamically changing physical environment that includes oxygen saturation and mechanical forces. Recently, a growing number of studies on myocardial microenvironment in cardiomyocyte proliferation and heart regeneration is ongoing. In this review, we provide an overview of recent advances in myocardial microenvironment, which plays an important role in cardiomyocyte proliferation and heart regeneration.
Collapse
Affiliation(s)
- Kexiao Zheng
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yanglin Hao
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chenkun Xia
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shaoxian Cheng
- Jingshan Union Hospital, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jizhang Yu
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhang Chen
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuan Li
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuqing Niu
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shuan Ran
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Song Wang
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Weicong Ye
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zilong Luo
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaohan Li
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jiulu Zhao
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ran Li
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Junjie Zong
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Han Zhang
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Longyong Lai
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Pinyan Huang
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Cheng Zhou
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jiahong Xia
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xi Zhang
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jie Wu
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
4
|
Cai W, Lian L, Li A, Zhang Q, Li M, Zhang J, Xie Y. Cardiac resident macrophages: The core of cardiac immune homeostasis. Cell Signal 2024; 119:111169. [PMID: 38599440 DOI: 10.1016/j.cellsig.2024.111169] [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: 01/31/2024] [Revised: 03/24/2024] [Accepted: 04/07/2024] [Indexed: 04/12/2024]
Abstract
Cardiac resident macrophages (CRMs) are essential in maintaining the balance of the immune homeostasis in the heart. One of the main factors in the progression of cardiovascular diseases, such as myocarditis, myocardial infarction(MI), and heart failure(HF), is the imbalance in the regulatory mechanisms of CRMs. Recent studies have reported novel heterogeneity and spatiotemporal complexity of CRMs, and their role in maintaining cardiac immune homeostasis and treating cardiovascular diseases. In this review, we focus on the functions of CRMs, including immune surveillance, immune phagocytosis, and immune metabolism, and explore the impact of CRM's homeostasis imbalance on cardiac injury and cardiac repair. We also discuss the therapeutic approaches linked to CRMs. The immunomodulatory strategies targeting CRMs may be a therapeutic approach for the treatment of cardiovascular disease.
Collapse
Affiliation(s)
- Wenhui Cai
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300183, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300193, China; Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China
| | - Lu Lian
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300183, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300193, China; Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China
| | - Aolin Li
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300183, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300193, China; Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China
| | - Qianqian Zhang
- Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China
| | - Mengmeng Li
- Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China
| | - Junping Zhang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300183, China.
| | - YingYu Xie
- College of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China.
| |
Collapse
|
5
|
Jin J, Wang Y, Liu Y, Chakrabarti S, Su Z. Cardiac resident macrophages: Spatiotemporal distribution, development, physiological functions, and their translational potential on cardiac diseases. Acta Pharm Sin B 2024; 14:1483-1493. [PMID: 38572111 PMCID: PMC10985034 DOI: 10.1016/j.apsb.2023.12.018] [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: 10/10/2023] [Revised: 11/27/2023] [Accepted: 12/29/2023] [Indexed: 04/05/2024] Open
Abstract
Cardiac resident macrophages (CRMs) are the main population of cardiac immune cells. The role of these cells in regeneration, functional remodeling, and repair after cardiac injury is always the focus of research. However, in recent years, their dynamic changes and contributions in physiological states have a significant attention. CRMs have specific phenotypes and functions in different cardiac chambers or locations of the heart and at different stages. They further show specific differentiation and development processes. The present review will summarize the new progress about the spatiotemporal distribution, potential developmental regulation, and their roles in cardiac development and aging as well as the translational potential of CRMs on cardiac diseases. Of course, the research tools for CRMs, their respective advantages and disadvantages, and key issues on CRMs will further be discussed.
Collapse
Affiliation(s)
- Jing Jin
- International Genome Center, Jiangsu University, Zhenjiang 212013, China
- Institute for Medical Immunology, Jiangsu University, Zhenjiang 212013, China
| | - Yurou Wang
- International Genome Center, Jiangsu University, Zhenjiang 212013, China
- Institute for Medical Immunology, Jiangsu University, Zhenjiang 212013, China
| | - Yueqin Liu
- Center Laboratory, the Fourth People's Hospital of Zhenjiang, Zhenjiang 212008, China
| | - Subrata Chakrabarti
- Department of Pathology and Laboratory Medicine, Western University, London, Ontario N6A 5C1, Canada
| | - Zhaoliang Su
- International Genome Center, Jiangsu University, Zhenjiang 212013, China
- Institute for Medical Immunology, Jiangsu University, Zhenjiang 212013, China
| |
Collapse
|
6
|
Li L, Cao J, Li S, Cui T, Ni J, Zhang H, Zhu Y, Mao J, Gao X, Midgley AC, Zhu M, Fan G. M2 Macrophage-Derived sEV Regulate Pro-Inflammatory CCR2 + Macrophage Subpopulations to Favor Post-AMI Cardiac Repair. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2202964. [PMID: 36950739 PMCID: PMC10190454 DOI: 10.1002/advs.202202964] [Citation(s) in RCA: 19] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 02/21/2023] [Indexed: 05/18/2023]
Abstract
Tissue-resident cardiac macrophage subsets mediate cardiac tissue inflammation and repair after acute myocardial infarction (AMI). CC chemokine receptor 2 (CCR2)-expressing macrophages have phenotypical similarities to M1-polarized macrophages, are pro-inflammatory, and recruit CCR2+ circulating monocytes to infarcted myocardium. Small extracellular vesicles (sEV) from CCR2̶ macrophages, which phenotypically resemble M2-polarized macrophages, promote anti-inflammatory activity and cardiac repair. Here, the authors harvested M2 macrophage-derived sEV (M2EV ) from M2-polarized bone-marrow-derived macrophages for intramyocardial injection and recapitulation of sEV-mediated anti-inflammatory activity in ischemic-reperfusion (I/R) injured hearts. Rats and pigs received sham surgery; I/R without treatment; or I/R with autologous M2EV treatment. M2EV rescued cardiac function and attenuated injury markers, infarct size, and scar size. M2EV inhibited CCR2+ macrophage numbers, reduced monocyte-derived CCR2+ macrophage recruitment to infarct sites, induced M1-to-M2 macrophage switching and promoted neovascularization. Analysis of M2EV microRNA content revealed abundant miR-181b-5p, which regulated macrophage glucose uptake, glycolysis, and mitigated mitochondrial reactive oxygen species generation. Functional blockade of miR-181b-5p is detrimental to beneficial M2EV actions and resulted in failure to inhibit CCR2+ macrophage numbers and infarct size. Taken together, this investigation showed that M2EV rescued myocardial function, improved myocardial repair, and regulated CCR2+ macrophages via miR-181b-5p-dependent mechanisms, indicating an option for cell-free therapy for AMI.
Collapse
Affiliation(s)
- Lan Li
- State Key Laboratory of Modern Chinese MedicineKey Laboratory of Pharmacology of Traditional Chinese Medical Formulae for the Ministry of EducationTianjin University of Traditional Chinese MedicineTianjin301617China
- National Clinical Research Center for Chinese Medicine Acupuncture and MoxibustionState Key Laboratory of Component‐based Chinese MedicineFirst Teaching Hospital of Tianjin University of Traditional Chinese MedicineTianjin300193China
| | - Jiasong Cao
- Tianjin Key Laboratory of Human Development and Reproductive RegulationTianjin Central Hospital of Gynecology ObstetricsTianjin300052China
| | - Sheng Li
- State Key Laboratory of Modern Chinese MedicineKey Laboratory of Pharmacology of Traditional Chinese Medical Formulae for the Ministry of EducationTianjin University of Traditional Chinese MedicineTianjin301617China
| | - Tianyi Cui
- State Key Laboratory of Modern Chinese MedicineKey Laboratory of Pharmacology of Traditional Chinese Medical Formulae for the Ministry of EducationTianjin University of Traditional Chinese MedicineTianjin301617China
| | - Jingyu Ni
- State Key Laboratory of Modern Chinese MedicineKey Laboratory of Pharmacology of Traditional Chinese Medical Formulae for the Ministry of EducationTianjin University of Traditional Chinese MedicineTianjin301617China
- National Clinical Research Center for Chinese Medicine Acupuncture and MoxibustionState Key Laboratory of Component‐based Chinese MedicineFirst Teaching Hospital of Tianjin University of Traditional Chinese MedicineTianjin300193China
| | - Han Zhang
- State Key Laboratory of Modern Chinese MedicineKey Laboratory of Pharmacology of Traditional Chinese Medical Formulae for the Ministry of EducationTianjin University of Traditional Chinese MedicineTianjin301617China
| | - Yan Zhu
- State Key Laboratory of Modern Chinese MedicineKey Laboratory of Pharmacology of Traditional Chinese Medical Formulae for the Ministry of EducationTianjin University of Traditional Chinese MedicineTianjin301617China
| | - Jingyuan Mao
- National Clinical Research Center for Chinese Medicine Acupuncture and MoxibustionState Key Laboratory of Component‐based Chinese MedicineFirst Teaching Hospital of Tianjin University of Traditional Chinese MedicineTianjin300193China
| | - Xiumei Gao
- State Key Laboratory of Modern Chinese MedicineKey Laboratory of Pharmacology of Traditional Chinese Medical Formulae for the Ministry of EducationTianjin University of Traditional Chinese MedicineTianjin301617China
| | - Adam C. Midgley
- Key Laboratory of Bioactive Materials for the Ministry of EducationCollege of Life SciencesNankai UniversityTianjin300071China
| | - Meifeng Zhu
- Key Laboratory of Bioactive Materials for the Ministry of EducationCollege of Life SciencesNankai UniversityTianjin300071China
| | - Guanwei Fan
- State Key Laboratory of Modern Chinese MedicineKey Laboratory of Pharmacology of Traditional Chinese Medical Formulae for the Ministry of EducationTianjin University of Traditional Chinese MedicineTianjin301617China
- National Clinical Research Center for Chinese Medicine Acupuncture and MoxibustionState Key Laboratory of Component‐based Chinese MedicineFirst Teaching Hospital of Tianjin University of Traditional Chinese MedicineTianjin300193China
| |
Collapse
|
7
|
Wang K, Sun X, Sun Y, Jiao B, Yao J, Hu Y, Deng Q, Dong J, Wang W, Wang Y, Li C. Transcriptional regulation of macrophages in heart failure. Front Cardiovasc Med 2023; 10:1148041. [PMID: 37063966 PMCID: PMC10097991 DOI: 10.3389/fcvm.2023.1148041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Accepted: 03/13/2023] [Indexed: 04/03/2023] Open
Abstract
Adverse cardiac remodeling after acute myocardial infarction is the most important pathological mechanism of heart failure and remains a major problem in clinical practice. Cardiac macrophages, derived from tissue resident macrophages and circulating monocyte, undergo significant phenotypic and functional changes following cardiac injury and play crucial roles in inflammatory response and tissue repair response. Currently, numerous studies indicate that epigenetic regulatory factors and transcription factors can regulate the transcription of inflammatory and reparative genes and timely conversion of inflammatory macrophages into reparative macrophages and then alleviate cardiac remodeling. Accordingly, targeting transcriptional regulation of macrophages may be a promising option for heart failure treatment. In this review, we not only summarize the origin and function of cardiac macrophages, but more importantly, describe the transcriptional regulation of macrophages in heart failure, aiming to provide a potential therapeutic target for heart failure.
Collapse
Affiliation(s)
- Keyan Wang
- College of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Xiaoqian Sun
- College of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Ying Sun
- College of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Boyang Jiao
- College of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Junkai Yao
- College of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Yueyao Hu
- College of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Qiong Deng
- College of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Jianteng Dong
- College of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Wei Wang
- College of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
- Beijing Key Laboratory of Traditional Chinese Medicine (TCM) Syndrome and Formula, Beijing University of Chinese Medicine, Beijing, China
- School of Chinese Materia Medica, Guangzhou University of Chinese Medicine, Guangzhou, China
- Correspondence: Wei Wang Yong Wang Chun Li
| | - Yong Wang
- College of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
- Beijing Key Laboratory of Traditional Chinese Medicine (TCM) Syndrome and Formula, Beijing University of Chinese Medicine, Beijing, China
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
- Correspondence: Wei Wang Yong Wang Chun Li
| | - Chun Li
- Beijing Key Laboratory of Traditional Chinese Medicine (TCM) Syndrome and Formula, Beijing University of Chinese Medicine, Beijing, China
- Modern Research Center for Traditional Chinese Medicine (TCM), Beijing University of Chinese Medicine, Beijing, China
- Correspondence: Wei Wang Yong Wang Chun Li
| |
Collapse
|
8
|
Sun W, Lu H, Cui S, Zhao S, Yu H, song H, Ruan Q, Zhang Y, Chu Y, Dong S. NEDD4 ameliorates myocardial reperfusion injury by preventing macrophages pyroptosis. Cell Commun Signal 2023; 21:29. [PMID: 36732831 PMCID: PMC9896697 DOI: 10.1186/s12964-022-01022-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Accepted: 12/17/2022] [Indexed: 02/04/2023] Open
Abstract
OBJECTIVES The inflammatory cascade and cell death post-myocardial ischemia reperfusion (MI/R) are very complex. Despite the understanding that macrophage inflammation has a pivotal role in the pathophysiology of MI/R, the contribution of macrophage inflammatory signals in tailoring the function of vascular endothelium remains unknown. MATERIALS AND METHODS In the present study, we analyzed the effects of NEDD4 on the NLRP3 inflammasome activation-mediated pyroptosis in vitro after an acute pro-inflammatory stimulus and in vivo in a MI/R mouse model. TTC and Evan's blue dye, Thioflavin S, immunohistochemistry staining, and ELISA were performed in wild-type and NEDD4 deficiency mice. THP-1 cells were transfected with si-NEDD4 or si-SF3A2. HEK293T cells were transfected with NEDD4 or SF3A2 overexpression plasmid. ELISA analyzed the inflammatory cytokines in the cell supernatant. The levels of NEDD4, SF3A2, and NLRP3/GSDMD pathway were determined by Western blot. Protein interactions were evaluated by immunoprecipitation. The protein colocalization in cells was monitored using a fluorescence microscope. RESULTS NEDD4 inhibited NLRP3 inflammasome activation and pyroptosis in THP-1 cells treated with lipopolysaccharide (LPS) and nigericin (Nig). Mechanistically, NEDD4 maintained the stability of NLRP3 through direct interaction with the SF3A2, whereas the latter association with NLRP3 indirectly interacted with NEDD4 promoting proteasomal degradation of NLRP3. Deletion of NLRP3 expression further inhibited the caspase cascade to induce pyroptosis. Interestingly, inhibiting NLRP3 inflammasome activation in THP-1 cells could prevent cardiac microvascular endothelial cells (CMECs) injury. In addition, NEDD4 deficiency decreased animal survival and increased myocardial infarct size, no-reflow area, and promoted macrophages infiltration post-MI/R. CONCLUSIONS NEDD4 could be a potential therapeutic target in microvascular injury following myocardial reperfusion. Video Abstract.
Collapse
Affiliation(s)
- Wenjing Sun
- grid.207374.50000 0001 2189 3846Department of Cardiology, Henan Provincial People’s Hospital, Zhengzhou University People’s Hospital, Zhengzhou, 450000 China ,grid.207374.50000 0001 2189 3846Department of Clinical Microbiology, Henan Provincial People’s Hospital, Zhengzhou University People’s Hospital, Zhengzhou, 450000 China
| | - Hongquan Lu
- grid.207374.50000 0001 2189 3846Department of Clinical Microbiology, Henan Provincial People’s Hospital, Zhengzhou University People’s Hospital, Zhengzhou, 450000 China
| | - Shihua Cui
- Department of Nuclear Medicine, Third People’s Hospital of Honghe State, Honghe, 661000 China
| | - Shenghui Zhao
- grid.207374.50000 0001 2189 3846Department of Cardiology, Henan Provincial People’s Hospital, Zhengzhou University People’s Hospital, Zhengzhou, 450000 China
| | - Haijia Yu
- grid.207374.50000 0001 2189 3846Department of Cardiology, Henan Provincial People’s Hospital, Zhengzhou University People’s Hospital, Zhengzhou, 450000 China
| | - Huihui song
- grid.207374.50000 0001 2189 3846Department of Cardiology, Henan Provincial People’s Hospital, Zhengzhou University People’s Hospital, Zhengzhou, 450000 China
| | - Qiuyue Ruan
- grid.411971.b0000 0000 9558 1426Department of Cardiology, Dalian Medical University, Dalian, 116000 China ,Department of Nephrology, First People’s Hospital of Honghe State, Honghe, 661000 China
| | - Yabin Zhang
- grid.207374.50000 0001 2189 3846Department of Clinical Microbiology, Henan Provincial People’s Hospital, Zhengzhou University People’s Hospital, Zhengzhou, 450000 China
| | - Yingjie Chu
- Department of Cardiology, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Zhengzhou, 450000, China.
| | - Shujuan Dong
- Department of Cardiology, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Zhengzhou, 450000, China.
| |
Collapse
|
9
|
Nian W, Huang Z, Fu C. Immune cells drive new immunomodulatory therapies for myocardial infarction: From basic to clinical translation. Front Immunol 2023; 14:1097295. [PMID: 36761726 PMCID: PMC9903069 DOI: 10.3389/fimmu.2023.1097295] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Accepted: 01/10/2023] [Indexed: 01/26/2023] Open
Abstract
The high incidence of heart failure secondary to myocardial infarction (MI) has been difficult to effectively address. MI causes strong aseptic inflammation, and infiltration of different immune cells and changes in the local inflammatory microenvironment play a key regulatory role in ventricular remodeling. Therefore, the possibility of improving the prognosis of MI through targeted immunity has been of interest and importance in MI. However, previously developed immune-targeted therapies have not achieved significant success in clinical trials. Here, we propose that the search for therapeutic targets from different immune cells may be more precise and lead to better clinical translation. Specifically, this review summarizes the role and potential therapeutic targets of various immune cells in ventricular remodeling after MI, especially monocytes/macrophages and neutrophils, as a way to demonstrate the importance and potential of immunomodulatory therapies for MI. In addition, we analyze the reasons for the failure of previous immunomodulatory therapies and the issues that need to be addressed, as well as the prospects and targeting strategies of using immune cells to drive novel immunomodulatory therapies, hoping to advance the development of immunomodulatory therapies by providing evidence and new ideas.
Collapse
Affiliation(s)
- Wenjian Nian
- Department of Clinical Medicine, Wannan Medical College, Wuhu, China
| | - Zijian Huang
- Department of Cardiology, Yi Ji Shan Hospital affiliated to Wannan Medical College, Wuhu, China.,Anesthesia Laboratory and Training Center, Wannan Medical College, Wuhu, China.,Key Laboratory of Non-coding RNA Transformation Research of Anhui Higher Education Institution, Wannan Medical College, Wuhu, China
| | - Cong Fu
- Department of Cardiology, Yi Ji Shan Hospital affiliated to Wannan Medical College, Wuhu, China.,Anesthesia Laboratory and Training Center, Wannan Medical College, Wuhu, China.,Key Laboratory of Non-coding RNA Transformation Research of Anhui Higher Education Institution, Wannan Medical College, Wuhu, China
| |
Collapse
|
10
|
Novel insights into embryonic cardiac macrophages. Dev Biol 2022; 488:1-10. [DOI: 10.1016/j.ydbio.2022.05.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 04/07/2022] [Accepted: 05/02/2022] [Indexed: 11/19/2022]
|
11
|
Jin K, Gao S, Yang P, Guo R, Li D, Zhang Y, Lu X, Fan G, Fan X. Single-Cell RNA Sequencing Reveals the Temporal Diversity and Dynamics of Cardiac Immunity after Myocardial Infarction. SMALL METHODS 2022; 6:e2100752. [PMID: 35023642 DOI: 10.1002/smtd.202100752] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 12/19/2021] [Indexed: 06/14/2023]
Abstract
Myocardial infarction (MI) is strongly associated with the temporal regulation of cardiac immunity. However, a variety of current clinical trials have failed because of the lack of post-MI immunomodulating/anti-inflammatory targets. Single-cell RNA sequencing analysis of the cardiac Cd45+ immune cell at 0, 3, 7, and 14 d after injury in a mouse left anterior descending coronary artery ligation model is performed. Major immune cell populations, distinct subsets, and dynamic changes are identified. Macrophages (Mø) are most abundant, peaking at 3 d after infarction. Mø-5 and Mø-6 are the predominant infiltrated subsets at this time point, with strong expression of inflammatory factors. Further analysis demonstrates that suppressing these sets attenuated pathological MI progression by preventing subsequent leukocyte extravasation and adverse remodeling. Abundant apoptotic neutrophils and a profibrotic macrophage subset on days 7 and 14, respectively, are also detected. These results provide a basis for developing cell type- and time-specific interventions in MI.
Collapse
Affiliation(s)
- Kaiyu Jin
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Shan Gao
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin Key Laboratory of Translational Research of TCM Prescription and Syndrome, Tianjin, 300193, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300193, China
| | - Penghui Yang
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Rongfang Guo
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Dan Li
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin Key Laboratory of Translational Research of TCM Prescription and Syndrome, Tianjin, 300193, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300193, China
| | - Yunsha Zhang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China
| | - Xiaoyan Lu
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Guanwei Fan
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin Key Laboratory of Translational Research of TCM Prescription and Syndrome, Tianjin, 300193, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300193, China
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin, 301617, China
| | - Xiaohui Fan
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin, 301617, China
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, 310058, China
| |
Collapse
|
12
|
Ma X, Wang S, Cheng H, Ouyang H, Ma X. Melatonin Attenuates Ischemia/Reperfusion-Induced Oxidative Stress by Activating Mitochondrial Fusion in Cardiomyocytes. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:7105181. [PMID: 35047108 PMCID: PMC8763517 DOI: 10.1155/2022/7105181] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 09/20/2021] [Accepted: 10/01/2021] [Indexed: 12/31/2022]
Abstract
Myocardial ischemia/reperfusion (I/R) injury can stimulate mitochondrial reactive oxygen species production. Optic atrophy 1- (OPA1-) induced mitochondrial fusion is an endogenous antioxidative mechanism that preserves the mitochondrial function. In our study, we investigated whether melatonin augments OPA1-dependent mitochondrial fusion and thus maintains redox balance during myocardial I/R injury. In hypoxia/reoxygenation- (H/R-) treated H9C2 cardiomyocytes, melatonin treatment upregulated OPA1 mRNA and protein expression, thereby enhancing mitochondrial fusion. Melatonin also suppressed apoptosis in H/R-treated cardiomyocytes, as evidenced by increased cell viability, diminished caspase-3 activity, and reduced Troponin T secretion; however, silencing OPA1 abolished these effects. H/R treatment augmented mitochondrial ROS production and repressed antioxidative molecule levels, while melatonin reversed these changes in an OPA1-dependent manner. Melatonin also inhibited mitochondrial permeability transition pore opening and maintained the mitochondrial membrane potential, but OPA1 silencing prevented these outcomes. These results illustrate that melatonin administration alleviates cardiomyocyte I/R injury by activating OPA1-induced mitochondrial fusion and inhibiting mitochondrial oxidative stress.
Collapse
Affiliation(s)
- Xiaoling Ma
- Department of Critical Care Medicine, Shijiazhuang People's Hospital, Shijiazhuang, Hebei 050000, China
| | - Shengchi Wang
- Department of Critical Care Medicine, Shijiazhuang People's Hospital, Shijiazhuang, Hebei 050000, China
| | - Hui Cheng
- Department of Critical Care Medicine, Shijiazhuang People's Hospital, Shijiazhuang, Hebei 050000, China
| | - Haichun Ouyang
- Department of Cardiology, The Seventh Affiliated Hospital, Southern Medical University, China
| | - Xiaoning Ma
- Department of Critical Care Medicine, Shijiazhuang People's Hospital, Shijiazhuang, Hebei 050000, China
| |
Collapse
|
13
|
FUNDC1 activates the mitochondrial unfolded protein response to preserve mitochondrial quality control in cardiac ischemia/reperfusion injury. Cell Signal 2022; 92:110249. [DOI: 10.1016/j.cellsig.2022.110249] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Revised: 12/28/2021] [Accepted: 01/12/2022] [Indexed: 12/13/2022]
|
14
|
Wu D, Kampmann E, Qian G. Novel Insights Into the Role of Mitochondria-Derived Peptides in Myocardial Infarction. Front Physiol 2021; 12:750177. [PMID: 34777013 PMCID: PMC8582487 DOI: 10.3389/fphys.2021.750177] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 09/28/2021] [Indexed: 01/02/2023] Open
Abstract
Mitochondria-derived peptides (MDPs) are a new class of bioactive peptides encoded by small open reading frames (sORFs) within known mitochondrial DNA (mtDNA) genes. MDPs may affect the expression of nuclear genes and play cytoprotective roles against chronic and age-related diseases by maintaining mitochondrial function and cell viability in the face of metabolic stress and cytotoxic insults. In this review, we summarize clinical and experimental findings indicating that MDPs act as local and systemic regulators of glucose homeostasis, immune and inflammatory responses, mitochondrial function, and adaptive stress responses, and focus on evidence supporting the protective effects of MDPs against myocardial infarction. These insights into MDPs actions suggest their potential in the treatment of cardiovascular diseases and should encourage further research in this field.
Collapse
Affiliation(s)
- Dan Wu
- Department of Cardiology, The First Medical Center, Chinese People's Liberation Army Hospital, Medical School of Chinese People's Liberation Army, Beijing, China
| | - Enny Kampmann
- School of Life Sciences, City College of San Francisco, San Francisco, CA, United States
| | - Geng Qian
- Department of Cardiology, The First Medical Center, Chinese People's Liberation Army Hospital, Medical School of Chinese People's Liberation Army, Beijing, China
| |
Collapse
|
15
|
Abstract
Macrophages are essential components of the immune system and play a role in the normal functioning of the cardiovascular system. Depending on their origin and phenotype, cardiac macrophages perform various functions. In a steady-state, these cells play a beneficial role in maintaining cardiac homeostasis by defending the body from pathogens and eliminating apoptotic cells, participating in electrical conduction, vessel patrolling, and arterial tone regulation. However, macrophages also take part in adverse cardiac remodeling that could lead to the development and progression of heart failure (HF) in such HF comorbidities as hypertension, obesity, diabetes, and myocardial infarction. Nevertheless, studies on detailed mechanisms of cardiac macrophage function are still in progress, and could enable potential therapeutic applications of these cells. This review aims to present the latest reports on the origin, heterogeneity, and functions of cardiac macrophages in the healthy heart and in cardiovascular diseases leading to HF. The potential therapeutic use of macrophages is also briefly discussed.
Collapse
|
16
|
Protective Effect of Optic Atrophy 1 on Cardiomyocyte Oxidative Stress: Roles of Mitophagy, Mitochondrial Fission, and MAPK/ERK Signaling. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:3726885. [PMID: 34211623 PMCID: PMC8205577 DOI: 10.1155/2021/3726885] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 05/08/2021] [Accepted: 05/21/2021] [Indexed: 12/14/2022]
Abstract
Myocardial infarction is associated with oxidative stress and mitochondrial damage. However, the regulatory mechanisms underlying cardiomyocyte oxidative stress during myocardial infarction are not fully understood. In the present study, we explored the cardioprotective action of optic atrophy 1- (Opa1-) mediated mitochondrial autophagy (mitophagy) in oxidative stress-challenged cardiomyocytes, with a focus on mitochondrial homeostasis and the MAPK/ERK pathway. Our results demonstrated that overexpression of Opa1 in cultured rat H9C2 cardiomyocytes, a procedure that stimulates mitophagy, attenuates oxidative stress and increases cellular antioxidant capacity. Activation of Opa1-mediated mitophagy suppressed cardiomyocyte apoptosis by downregulating Bax, caspase-9, and caspase-12 and upregulating Bcl-2 and c-IAP. Using mitochondrial tracker staining and a reactive oxygen species indicator, our assays showed that Opa1-mediated mitophagy attenuated mitochondrial fission and reduced ROS production in cardiomyocytes. In addition, we found that inhibition of the MAPK/ERK pathway abolished the antioxidant action of Opa1-mediated mitophagy in these cells. Taken together, our data demonstrate that Opa1-mediated mitophagy protects cardiomyocytes against oxidative stress damage through inhibition of mitochondrial fission and activation of MAPK/ERK signaling. These findings reveal a critical role for Opa1 in the modulation of cardiomyocyte redox balance and suggest a potential target for the treatment of myocardial infarction.
Collapse
|
17
|
Lyu J, Huang J, Wu J, Yu T, Wei X, Lei Q. Lack of Macrophage Migration Inhibitory Factor Reduces Susceptibility to Ventricular Arrhythmias During the Acute Phase of Myocardial Infarction. J Inflamm Res 2021; 14:1297-1311. [PMID: 33854357 PMCID: PMC8039209 DOI: 10.2147/jir.s304553] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Accepted: 03/16/2021] [Indexed: 12/29/2022] Open
Abstract
Background Macrophages are involved in inflammatory responses and play a crucial role in aggravating ventricular arrhythmias (VAs) after myocardial infarction (MI). Macrophage migration inhibitory factor (MIF) participates in inflammatory responses during acute MI. In the present study, we hypothesized that knockout (KO) of MIF may prevent VAs during the acute phase of MI by inhibiting macrophage-derived pro-inflammatory mediators. Methods and Results We demonstrated that MIF-KO mice in a mouse model of MI exhibited a significant decrease in susceptibility to VAs both in vivo (84.6% vs 40.7%, P < 0.05) and ex vivo (86.7% vs 40.0%, P < 0.05) at day 3 after MI compared with that in wild-type (WT) mice. Both WT and MIF-KO mice presented similar left ventricular contractility, peri-infarct myocardial fibrosis and sympathetic reinnervation, and circulating and local norepinephrine levels during the acute phase of MI. Meanwhile, MIF-KO mice had inhibited macrophage aggregation, alleviated connexin 43 (Cx43) redistribution, and reduced level of pro-inflammatory mediators, including tumor necrosis factor-α and interleukin-1β (P < 0.05) at day 3 after MI. The differences in susceptibility to VAs, expression of pro-inflammatory mediators, and Cx43 redistribution after MI between WT and MIF-KO mice disappeared by macrophage depletion with clodronate liposomes in both groups. Furthermore, the pro-inflammatory activity of cultured peritoneal macrophages was inhibited by MIF deficiency and recovered with replenishment of exogenous MIF in vitro. Conclusion In conclusion, we found that lack of MIF reduced the susceptibility to VAs in mouse heart during the acute phase of MI by inhibiting pro-inflammatory activity of macrophages and improving gap-junction and electrical remodeling.
Collapse
Affiliation(s)
- Juanjuan Lyu
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, 610041, People's Republic of China
| | - Jia Huang
- Department of Anesthesiology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610072, People's Republic of China.,Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu, 610072, People's Republic of China
| | - Jin Wu
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, 610041, People's Republic of China
| | - Tao Yu
- Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu, 610072, People's Republic of China.,Department of Cardiac Surgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610072, People's Republic of China
| | - Xinchuan Wei
- Department of Anesthesiology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610072, People's Republic of China.,Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu, 610072, People's Republic of China
| | - Qian Lei
- Department of Anesthesiology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610072, People's Republic of China.,Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu, 610072, People's Republic of China
| |
Collapse
|
18
|
Niderla-Bielińska J, Ścieżyńska A, Moskalik A, Jankowska-Steifer E, Bartkowiak K, Bartkowiak M, Kiernozek E, Podgórska A, Ciszek B, Majchrzak B, Ratajska A. A Comprehensive miRNome Analysis of Macrophages Isolated from db/db Mice and Selected miRNAs Involved in Metabolic Syndrome-Associated Cardiac Remodeling. Int J Mol Sci 2021; 22:2197. [PMID: 33672153 PMCID: PMC7926522 DOI: 10.3390/ijms22042197] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 02/19/2021] [Accepted: 02/20/2021] [Indexed: 01/10/2023] Open
Abstract
Cardiac macrophages are known from various activities, therefore we presume that microRNAs (miRNAs) produced or released by macrophages in cardiac tissue have impact on myocardial remodeling in individuals with metabolic syndrome (MetS). We aim to assess the cardiac macrophage miRNA profile by selecting those miRNA molecules that potentially exhibit regulatory functions in MetS-related cardiac remodeling. Cardiac tissue macrophages from control and db/db mice (an animal model of MetS) were counted and sorted with flow cytometry, which yielded two populations: CD45+CD11b+CD64+Ly6Chi and CD45+CD11b+CD64+Ly6Clow. Total RNA was then isolated, and miRNA expression profiles were evaluated with Next Generation Sequencing. We successfully sequenced 1400 miRNAs in both macrophage populations: CD45+CD11b+CD64+Ly6Chi and CD45+CD11b+CD64+Ly6Clow. Among the 1400 miRNAs, about 150 showed different expression levels in control and db/db mice and between these two subpopulations. At least 15 miRNAs are possibly associated with MetS pathology in cardiac tissue due to direct or indirect regulation of the expression of miRNAs for proteins involved in angiogenesis, fibrosis, or inflammation. In this paper, for the first time we describe the miRNA transcription profile in two distinct macrophage populations in MetS-affected cardiac tissue. Although the results are preliminary, the presented data provide a foundation for further studies on intercellular cross-talk/molecular mechanism(s) involved in the regulation of MetS-related cardiac remodeling.
Collapse
Affiliation(s)
- Justyna Niderla-Bielińska
- Department of Histology and Embryology, Collegium Anatomicum, Medical University of Warsaw, 02-004 Warsaw, Poland; (J.N.-B.); (A.Ś.); (E.J.-S.)
| | - Aneta Ścieżyńska
- Department of Histology and Embryology, Collegium Anatomicum, Medical University of Warsaw, 02-004 Warsaw, Poland; (J.N.-B.); (A.Ś.); (E.J.-S.)
| | - Aneta Moskalik
- Postgraduate School of Molecular Medicine, Collegium Anatomicum, Medical University of Warsaw, 02-004 Warsaw, Poland;
| | - Ewa Jankowska-Steifer
- Department of Histology and Embryology, Collegium Anatomicum, Medical University of Warsaw, 02-004 Warsaw, Poland; (J.N.-B.); (A.Ś.); (E.J.-S.)
| | - Krzysztof Bartkowiak
- Student Scientific Group, Department of Histology and Embryology, Collegium Anatomicum, Medical University of Warsaw, 02-004 Warsaw, Poland; (K.B.); (M.B.)
| | - Mateusz Bartkowiak
- Student Scientific Group, Department of Histology and Embryology, Collegium Anatomicum, Medical University of Warsaw, 02-004 Warsaw, Poland; (K.B.); (M.B.)
- Department of History of Medicine, Medical University of Warsaw, 00-575 Warsaw, Poland
| | - Ewelina Kiernozek
- Department of Immunology, Faculty of Biology, University of Warsaw, 02-096 Warsaw, Poland;
| | - Anna Podgórska
- Molecular Biology Laboratory, Department of Medical Biology, Cardinal Stefan Wyszyński Institute of Cardiology, 04-628 Warsaw, Poland;
| | - Bogdan Ciszek
- Department of Clinical Anatomy, Collegium Anatomicum, Medical University of Warsaw, 02-004 Warsaw, Poland;
| | - Barbara Majchrzak
- Department of Pathology, Collegium Anatomicum, Medical University of Warsaw, 02-004 Warsaw, Poland;
| | - Anna Ratajska
- Department of Pathology, Collegium Anatomicum, Medical University of Warsaw, 02-004 Warsaw, Poland;
| |
Collapse
|
19
|
Farache Trajano L, Smart N. Immunomodulation for optimal cardiac regeneration: insights from comparative analyses. NPJ Regen Med 2021; 6:8. [PMID: 33589632 PMCID: PMC7884783 DOI: 10.1038/s41536-021-00118-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Accepted: 01/14/2021] [Indexed: 02/06/2023] Open
Abstract
Despite decades of research, regeneration of the infarcted human heart remains an unmet ambition. A significant obstacle facing experimental regenerative therapies is the hostile immune response which arises following a myocardial infarction (MI). Upon cardiac damage, sterile inflammation commences via the release of pro-inflammatory meditators, leading to the migration of neutrophils, eosinophils and monocytes, as well as the activation of local vascular cells and fibroblasts. This response is amplified by components of the adaptive immune system. Moreover, the physical trauma of the infarction and immune-mediated tissue injury provides a supply of autoantigens, perpetuating a cycle of autoreactivity, which further contributes to adverse remodelling. A gradual shift towards an immune-resolving environment follows, culminating in the formation of a collagenous scar, which compromises cardiac function, ultimately driving the development of heart failure. Comparing the human heart with those of animal models that are capable of cardiac regeneration reveals key differences in the innate and adaptive immune responses to MI. By modulating key immune components to better resemble those of regenerative species, a cardiac environment may be established which would, either independently or via the synergistic application of emerging regenerative therapies, improve functional recovery post-MI.
Collapse
Affiliation(s)
- Luiza Farache Trajano
- British Heart Foundation Centre of Regenerative Medicine, Burdon Sanderson Cardiac Science centre, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | - Nicola Smart
- British Heart Foundation Centre of Regenerative Medicine, Burdon Sanderson Cardiac Science centre, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK.
| |
Collapse
|
20
|
Gula G, Rumiński S, Niderla-Bielińska J, Jasińska A, Kiernozek E, Jankowska-Steifer E, Flaht-Zabost A, Ratajska A. Potential functions of embryonic cardiac macrophages in angiogenesis, lymphangiogenesis and extracellular matrix remodeling. Histochem Cell Biol 2020; 155:117-132. [PMID: 33130914 PMCID: PMC7847984 DOI: 10.1007/s00418-020-01934-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/16/2020] [Indexed: 12/20/2022]
Abstract
The role of cardiac tissue macrophages (cTMs) during pre- and postnatal developmental stages remains in many aspects unknown. We aimed to characterize cTM populations and their potential functions based on surface markers. Our in situ studies of immunostained cardiac tissue specimens of murine fetuses (from E11to E17) revealed that a significant number of embryonic cTMs (phenotyped by CD45, CD68, CD64, F4/80, CD11b, CD206, Lyve-1) resided mostly in the subepicardial space, not in the entire myocardial wall, as observed in adult individuals. cTMs accompanied newly developed blood and lymphatic vessels adhering to vessel walls by cellular processes. A subpopulation of CD68-positive cells was found to form accumulations in areas of massive apoptosis during the outflow tract remodeling and shortening. Flow cytometry analysis at E14 and E17 stages revealed newly defined three subpopulations:CD64low, CD64highCD206-and CD64highCD206+. The levels of mRNA expression for genes related to regulation of angiogenesis (VEGFa, VEGFb, VEGFc, bFGF), lymphangiogenesis (VEGFc) and extracellular matrix (ECM) remodeling (MMP13, Arg1, Ym1/Chil3, Retlna/FIZZ1) differed among the selected populations and/or embryonic stages. Our results demonstrate a diversity of embryonic cTMs and their tissue-specific locations, suggesting their various potential roles in regulating angiogenesis, lymphangiogenesis and ECM remodeling.
Collapse
Affiliation(s)
- Grzegorz Gula
- Postgraduate School of Molecular Medicine, Medical University of Warsaw, Warsaw, Poland.,Department of Ophthalmology, Central Clinical Hospital of the MSWiA, Warsaw, Poland
| | - Sławomir Rumiński
- Department of Histology and Embryology, Medical University of Warsaw, Warsaw, Poland.,Centre for Preclinical Research and Technology, Warsaw, Poland
| | | | - Agnieszka Jasińska
- Department of Otorhinolaryngology, Head and Neck Surgery, Medical University of Warsaw, Student's Scientific Circle at Department of Pathology MUW, Warsaw, Poland
| | | | - Ewa Jankowska-Steifer
- Department of Histology and Embryology, Medical University of Warsaw, Warsaw, Poland
| | - Aleksandra Flaht-Zabost
- Department of Pathology, Medical University of Warsaw, Chałubińskiego 5, 02-004, Warsaw, Poland
| | - Anna Ratajska
- Department of Pathology, Medical University of Warsaw, Chałubińskiego 5, 02-004, Warsaw, Poland.
| |
Collapse
|
21
|
Lyu J, Wang M, Kang X, Xu H, Cao Z, Yu T, Huang K, Wu J, Wei X, Lei Q. Macrophage-mediated regulation of catecholamines in sympathetic neural remodeling after myocardial infarction. Basic Res Cardiol 2020; 115:56. [DOI: 10.1007/s00395-020-0813-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Accepted: 07/20/2020] [Indexed: 12/17/2022]
|