201
|
Kutryb-Zajac B, Mierzejewska P, Slominska EM, Smolenski RT. Therapeutic Perspectives of Adenosine Deaminase Inhibition in Cardiovascular Diseases. Molecules 2020; 25:molecules25204652. [PMID: 33053898 PMCID: PMC7587364 DOI: 10.3390/molecules25204652] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 10/02/2020] [Accepted: 10/07/2020] [Indexed: 02/06/2023] Open
Abstract
Adenosine deaminase (ADA) is an enzyme of purine metabolism that irreversibly converts adenosine to inosine or 2'deoxyadenosine to 2'deoxyinosine. ADA is active both inside the cell and on the cell surface where it was found to interact with membrane proteins, such as CD26 and adenosine receptors, forming ecto-ADA (eADA). In addition to adenosine uptake, the activity of eADA is an essential mechanism that terminates adenosine signaling. This is particularly important in cardiovascular system, where adenosine protects against endothelial dysfunction, vascular inflammation, or thrombosis. Besides enzymatic function, ADA protein mediates cell-to-cell interactions involved in lymphocyte co-stimulation or endothelial activation. Furthermore, alteration in ADA activity was demonstrated in many cardiovascular pathologies such as atherosclerosis, myocardial ischemia-reperfusion injury, hypertension, thrombosis, or diabetes. Modulation of ADA activity could be an important therapeutic target. This work provides a systematic review of ADA activity and anchoring inhibitors as well as summarizes the perspectives of their therapeutic use in cardiovascular pathologies associated with increased activity of ADA.
Collapse
Affiliation(s)
- Barbara Kutryb-Zajac
- Correspondence: (B.K.-Z); (R.T.S.); Tel.: +48-58-349-14-64 (B.K.-Z.); +48-58-349-14-60 (R.T.S.)
| | | | | | - Ryszard T. Smolenski
- Correspondence: (B.K.-Z); (R.T.S.); Tel.: +48-58-349-14-64 (B.K.-Z.); +48-58-349-14-60 (R.T.S.)
| |
Collapse
|
202
|
Al-Darraji A, Donahue RR, Tripathi H, Peng H, Levitan BM, Chelvarajan L, Haydar D, Gao E, Henson D, Gensel JC, Feola DJ, Venditto VJ, Abdel-Latif A. Liposomal delivery of azithromycin enhances its immunotherapeutic efficacy and reduces toxicity in myocardial infarction. Sci Rep 2020; 10:16596. [PMID: 33024189 PMCID: PMC7538891 DOI: 10.1038/s41598-020-73593-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2020] [Accepted: 09/18/2020] [Indexed: 12/20/2022] Open
Abstract
A growing body of evidence shows that altering the inflammatory response by alternative macrophage polarization is protective against complications related to acute myocardial infarction (MI). We have previously shown that oral azithromycin (AZM), initiated prior to MI, reduces inflammation and its negative sequelae on the myocardium. Here, we investigated the immunomodulatory role of a liposomal AZM formulation (L-AZM) in a clinically relevant model to enhance its therapeutic potency and avoid off-target effects. L-AZM (40 or 10 mg/kg, IV) was administered immediately post-MI and compared to free AZM (F-AZM). L-AZM reduced cardiac toxicity and associated mortality by 50% in mice. We observed a significant shift favoring reparatory/anti-inflammatory macrophages with L-AZM formulation. L-AZM use resulted in a remarkable decrease in cardiac inflammatory neutrophils and the infiltration of inflammatory monocytes. Immune cell modulation was associated with the downregulation of pro-inflammatory genes and the upregulation of anti-inflammatory genes. The immunomodulatory effects of L-AZM were associated with a reduction in cardiac cell death and scar size as well as enhanced angiogenesis. Overall, L-AZM use enhanced cardiac recovery and survival after MI. Importantly, L-AZM was protective from F-AZM cardiac off-target effects. We demonstrate that the liposomal formulation of AZM enhances the drug’s efficacy and safety in an animal model of acute myocardial injury. This is the first study to establish the immunomodulatory properties of liposomal AZM formulations. Our findings strongly support clinical trials using L-AZM as a novel and clinically relevant therapeutic target to improve cardiac recovery and reduce heart failure post-MI in humans.
Collapse
Affiliation(s)
- Ahmed Al-Darraji
- Gill Heart and Vascular Institute and Division of Cardiovascular Medicine, University of Kentucky, Lexington, KY, USA
| | - Renée R Donahue
- Gill Heart and Vascular Institute and Division of Cardiovascular Medicine, University of Kentucky, Lexington, KY, USA
| | - Himi Tripathi
- Gill Heart and Vascular Institute and Division of Cardiovascular Medicine, University of Kentucky, Lexington, KY, USA
| | - Hsuan Peng
- Gill Heart and Vascular Institute and Division of Cardiovascular Medicine, University of Kentucky, Lexington, KY, USA
| | - Bryana M Levitan
- Gill Heart and Vascular Institute and Division of Cardiovascular Medicine, University of Kentucky, Lexington, KY, USA
| | - Lakshman Chelvarajan
- Gill Heart and Vascular Institute and Division of Cardiovascular Medicine, University of Kentucky, Lexington, KY, USA
| | - Dalia Haydar
- College of Pharmacy, University of Kentucky, Lexington, KY, USA
| | - Erhe Gao
- The Center for Translational Medicine, Temple University School of Medicine, Philadelphia, PA, USA
| | - David Henson
- College of Pharmacy, University of Kentucky, Lexington, KY, USA
| | - John C Gensel
- Spinal Cord and Brain Injury Research Center, Department of Physiology, College of Medicine University of Kentucky, Lexington, USA
| | - David J Feola
- College of Pharmacy, University of Kentucky, Lexington, KY, USA
| | | | - Ahmed Abdel-Latif
- Gill Heart and Vascular Institute and Division of Cardiovascular Medicine, University of Kentucky, Lexington, KY, USA. .,Division of Cardiology, University of Kentucky and the Lexington VAMC, 741 S. Limestone Street, BBSRB, Room 349, Lexington, KY, 40536-0509, USA.
| |
Collapse
|
203
|
Xu W, Wong G, Hwang YY, Larbi A. The untwining of immunosenescence and aging. Semin Immunopathol 2020; 42:559-572. [PMID: 33165716 PMCID: PMC7665974 DOI: 10.1007/s00281-020-00824-x] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2020] [Accepted: 10/19/2020] [Indexed: 02/08/2023]
Abstract
From a holistic point of view, aging results from the cumulative erosion of the various systems. Among these, the immune system is interconnected to the rest as immune cells are present in all organs and recirculate through bloodstream. Immunosenescence is the term used to define the remodelling of immune changes during aging. Because immune cells-and particularly lymphocytes-can further differentiate after their maturation in response to pathogen recognition, it is therefore unclear when senescence is induced in these cells. Additionally, it is also unclear which signals triggers senescence in immune cells (i) aging per se, (ii) specific response to pathogens, (iii) underlying conditions, or (iv) inflammaging. In this review, we will cover the current knowledge and concepts linked to immunosenescence and we focus this review on lymphocytes and T cells, which represent the typical model for replicative senescence. With the evidence presented, we propose to disentangle the senescence of immune cells from chronological aging.
Collapse
Affiliation(s)
- Weili Xu
- Singapore Immunology Network (SIgN), Agency for Science Technology and Research (A*STAR), Immunos, Singapore, Singapore
| | - Glenn Wong
- Singapore Immunology Network (SIgN), Agency for Science Technology and Research (A*STAR), Immunos, Singapore, Singapore
| | - You Yi Hwang
- Singapore Immunology Network (SIgN), Agency for Science Technology and Research (A*STAR), Immunos, Singapore, Singapore
| | - Anis Larbi
- Singapore Immunology Network (SIgN), Agency for Science Technology and Research (A*STAR), Immunos, Singapore, Singapore.
- Department of Geriatrics, Faculty of Medicine, University of Sherbrooke, Sherbrooke, QC, J1K 2R1, Canada.
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore.
| |
Collapse
|
204
|
Mouton AJ, Hall JE. Novel roles of immunometabolism and nonmyocyte metabolism in cardiac remodeling and injury. Am J Physiol Regul Integr Comp Physiol 2020; 319:R476-R484. [PMID: 32877243 DOI: 10.1152/ajpregu.00188.2020] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Changes in cardiomyocyte metabolism have been heavily implicated in cardiac injury and heart failure (HF). However, there is emerging evidence that metabolism in nonmyocyte populations, including cardiac fibroblasts, immune cells, and endothelial cells, plays an important role in cardiac remodeling and adaptation to injury. Here, we discuss recent advances and insights into nonmyocyte metabolism in the healthy and injured heart. Metabolic switching from mitochondrial oxidative phosphorylation to glycolysis is critical for immune cell (macrophage and T lymphocyte) and fibroblast phenotypic switching in the inflamed and fibrotic heart. On the other hand, cardiac endothelial cells are heavily reliant on glycolytic metabolism, and thus impairments in glycolytic metabolism underlie endothelial cell dysfunction. Finally, we review current and ongoing metabolic therapies for HF and the potential implications for nonmyocyte metabolism.
Collapse
Affiliation(s)
- Alan J Mouton
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, Mississippi.,Mississippi Center for Obesity Research, University of Mississippi Medical Center, Jackson, Mississippi
| | - John E Hall
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, Mississippi.,Mississippi Center for Obesity Research, University of Mississippi Medical Center, Jackson, Mississippi
| |
Collapse
|
205
|
Hume RD, Chong JJH. The Cardiac Injury Immune Response as a Target for Regenerative and Cellular Therapies. Clin Ther 2020; 42:1923-1943. [PMID: 33010930 DOI: 10.1016/j.clinthera.2020.09.006] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 08/20/2020] [Accepted: 09/09/2020] [Indexed: 12/11/2022]
Abstract
PURPOSE Despite modern reperfusion and pharmacologic therapies, myocardial infarction (MI) remains a leading cause of morbidity and mortality worldwide. Therefore, the development of further therapeutics affecting post-MI recovery poses significant benefits. This review focuses on the post-MI immune response and immunomodulatory therapeutics that could improve the wound-healing response. METHODS This narrative review used OVID versions of MEDLINE and EMBASE searching for clinical therapeutics targeting the immune system during MI. Preclinical models and clinical trials were included. Additional studies were sourced from the reference lists of relevant articles and other personal files. FINDINGS After MI, cardiomyocytes are starved of oxygen and undergo cell death via coagulative necrosis. This process activates the immune system and a multifaceted wound-healing response, comprising a number of complex and overlapping phases. Overactivation or persistence of one or more of these phases can have potentially lethal implications. This review describes the immune response post-MI and any adverse events that can occur during these different phases. Second, we describe immunomodulatory therapies that attempt to target these immune cell aberrations by mitigating or diminishing their effects on the wound-healing response. Also discussed are adult stem/progenitor cell therapies, exosomes, and regulatory T cells, and their immunomodulatory effects in the post-MI setting. IMPLICATIONS An updated understanding into the importance of various inflammatory cell phenotypes, coupled with new technologies, may hold promise for a new era of immunomodulatory therapeutics. The implications of such therapies could dramatically improve patients' quality of life post-MI and reduce the incidence of progressive heart failure.
Collapse
Affiliation(s)
- Robert D Hume
- Centre for Heart Research, Westmead Institute for Medical Research, The University of Sydney, 176 Hawkesbury Rd, Westmead, NSW 2145, Australia
| | - James J H Chong
- Centre for Heart Research, Westmead Institute for Medical Research, The University of Sydney, 176 Hawkesbury Rd, Westmead, NSW 2145, Australia; Department of Cardiology, Westmead Hospital, Hawkesbury Rd, Westmead, NSW 2145, Australia.
| |
Collapse
|
206
|
LncRNA NRON alleviates atrial fibrosis through suppression of M1 macrophages activated by atrial myocytes. Biosci Rep 2020; 39:220726. [PMID: 31693733 PMCID: PMC6879354 DOI: 10.1042/bsr20192215] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Revised: 09/09/2019] [Accepted: 09/25/2019] [Indexed: 01/09/2023] Open
Abstract
The aim of the present study was to explore the role of long non-coding RNA (lncRNA) non-coding repressor of NFAT (NRON) in the atrial fibrosis and to explore whether its underlying mechanism was associated with macrophage polarization. Enzyme-linked immunosorbent assay (ELISA) analysis of pro-inflammatory cytokines revealed that NRON overexpression suppressed, whereas NRON silencing facilitated the angiotensin II (Ang II)-induced inflammatory response in primary cultured atrial myocytes. The chromatin immunoprecipitation (ChIP) results showed that nuclear factor of activated T cell 3 (NFATc3) was recruited to the promoter region of interleukin (IL) 12 (IL-12) in atrial myocytes. Further data showed that NRON overexpression suppressed, whereas NRON silencing further promoted the Ang II-induced NFATc3 nuclear transport and IL-12 expression in atrial myocytes. Moreover, RAW264.7 macrophages were incubated with the conditioned medium from the Ang II-treated atrial myocytes transfected with NRON and IL-12 overexpression vectors. IL-12 overexpression abrogated the NRON overexpression-mediated inhibition of RAW264.7 macrophage polarization to the M1-like phenotype. Additionally, mouse atrial fibroblasts were incubated with the culture medium from RAW264.7 macrophages treated as described above. IL-12 overexpression rescued the NRON overexpression-inhibited protein levels of fibrosis markers Collagen I/III in mouse atrial fibroblasts. Collectively, our data indicate that lncRNA NRON alleviates atrial fibrosis through suppression of M1 macrophages activated by atrial myocytes.
Collapse
|
207
|
Zhou K, Guo T, Xu Y, Guo R. Correlation Between Plasma Matrix Metalloproteinase-28 Levels and Severity of Calcific Aortic Valve Stenosis. Med Sci Monit 2020; 26:e925260. [PMID: 32950995 PMCID: PMC7526340 DOI: 10.12659/msm.925260] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2020] [Accepted: 06/22/2020] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND Calcific aortic valve disease is a common cardiovascular disorder worldwide. This study aimed to investigate the correlation between plasma matrix metalloproteinase-28 (MMP-28) levels and the severity of calcific aortic valve stenosis. MATERIAL AND METHODS Calcific aortic valve stenosis patients who were admitted to the heart center of our hospital between January 2016 and January 2019 to undergo surgery were successively enrolled in this study (55 males and 24 females with an average age of 58.5±9.6). Information on echocardiography, plasma MMP-28 levels, and other clinical data of the patients was retrospectively collected. RESULTS The average plasma MMP-28 level was 2.43±2.22 ng/mL (range, 0.22-8.27 ng/mL). Plasma MMP-28 levels in patients with mild (n=24), moderate (n=31), or severe (n=24) aortic valve stenosis were 0.74 (0.25-2.23), 1.46 (0.50-3.22), and 4.13 (1.54-6.18) ng/mL, respectively, indicating that the patients with severe aortic valve stenosis had significantly higher MMP-28 levels than the patients with moderate or mild aortic valve stenosis (both P<0.01). Regression analysis using the general linear model further revealed that plasma MMP-28 level was correlated with the peak blood flow velocity and mean pressure gradient of the transaortic valve, and the correlations were statistically significant (both P<0.01). CONCLUSIONS MMP-28 level is significantly elevated in severe cases of calcific aortic valve stenosis. Moreover, plasma MMP-28 levels are positively correlated with the mean pressure gradients and peak blood flow velocity of the transaortic valve.
Collapse
Affiliation(s)
- Ke Zhou
- Department of Cardiology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, P.R. China
| | - Ting Guo
- Department of Anesthesiology, Huashan Hospital, Fudan University, Shanghai, P.R. China
| | - Yawei Xu
- Department of Cardiology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, P.R. China
| | - Rong Guo
- Department of Cardiology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, P.R. China
| |
Collapse
|
208
|
Imanaka-Yoshida K, Tawara I, Yoshida T. Tenascin-C in cardiac disease: a sophisticated controller of inflammation, repair, and fibrosis. Am J Physiol Cell Physiol 2020; 319:C781-C796. [PMID: 32845719 DOI: 10.1152/ajpcell.00353.2020] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Tenascin-C (TNC) is a large extracellular matrix glycoprotein classified as a matricellular protein that is generally upregulated at high levels during physiological and pathological tissue remodeling and is involved in important biological signaling pathways. In the heart, TNC is transiently expressed at several important steps during embryonic development and is sparsely detected in normal adult heart but is re-expressed in a spatiotemporally restricted manner under pathological conditions associated with inflammation, such as myocardial infarction, hypertensive cardiac fibrosis, myocarditis, dilated cardiomyopathy, and Kawasaki disease. Despite its characteristic and spatiotemporally restricted expression, TNC knockout mice develop a grossly normal phenotype. However, various disease models using TNC null mice combined with in vitro experiments have revealed many important functions for TNC and multiple molecular cascades that control cellular responses in inflammation, tissue repair, and even myocardial regeneration. TNC has context-dependent diverse functions and, thus, may exert both harmful and beneficial effects in damaged hearts. However, TNC appears to deteriorate adverse ventricular remodeling by proinflammatory and profibrotic effects in most cases. Its specific expression also makes TNC a feasible diagnostic biomarker and target for molecular imaging to assess inflammation in the heart. Several preclinical studies have shown the utility of TNC as a biomarker for assessing the prognosis of patients and selecting appropriate therapy, particularly for inflammatory heart diseases.
Collapse
Affiliation(s)
- Kyoko Imanaka-Yoshida
- Department of Pathology and Matrix Biology, Mie University Graduate School of Medicine, Tsu, Japan.,Mie University Research Center for Matrix Biology, Tsu, Japan
| | - Isao Tawara
- Department of Hematology and Oncology, Mie University Graduate School of Medicine, Tsu, Japan.,Mie University Research Center for Matrix Biology, Tsu, Japan
| | - Toshimichi Yoshida
- Department of Pathology and Matrix Biology, Mie University Graduate School of Medicine, Tsu, Japan.,Mie University Research Center for Matrix Biology, Tsu, Japan
| |
Collapse
|
209
|
Liao Y, Li H, Cao H, Dong Y, Gao L, Liu Z, Ge J, Zhu H. Therapeutic silencing miR-146b-5p improves cardiac remodeling in a porcine model of myocardial infarction by modulating the wound reparative phenotype. Protein Cell 2020; 12:194-212. [PMID: 32845445 PMCID: PMC7895884 DOI: 10.1007/s13238-020-00750-6] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Accepted: 05/29/2020] [Indexed: 12/18/2022] Open
Abstract
Fibrotic remodeling is an adverse consequence of immune response-driven phenotypic modulation of cardiac cells following myocardial infarction (MI). MicroRNA-146b (miR-146b) is an active regulator of immunomodulation, but its function in the cardiac inflammatory cascade and its clinical implication in fibrotic remodeling following MI remain largely unknown. Herein, miR-146b-5p was found to be upregulated in the infarcted myocardium of mice and the serum of myocardial ischemia patients. Gain- and loss-of-function experiments demonstrated that miR-146b-5p was a hypoxia-induced regulator that governed the pro-fibrotic phenotype transition of cardiac cells. Overexpression of miR-146b-5p activated fibroblast proliferation, migration, and fibroblast-to-myofibroblast transition, impaired endothelial cell function and stress survival, and disturbed macrophage paracrine signaling. Interestingly, the opposite effects were observed when miR-146b-5p expression was inhibited. Luciferase assays and rescue studies demonstrated that the miR-146b-5p target genes mediating the above phenotypic modulations included interleukin 1 receptor associated kinase 1 (IRAK1) and carcinoembryonic antigen related cell adhesion molecule 1 (CEACAM1). Local delivery of a miR-146b-5p antagomir significantly reduced fibrosis and cell death, and upregulated capillary and reparative macrophages in the infarcted myocardium to restore cardiac remodeling and function in both mouse and porcine MI models. Local inhibition of miR-146b-5p may represent a novel therapeutic approach to treat cardiac fibrotic remodeling and dysfunction following MI.
Collapse
Affiliation(s)
- Yiteng Liao
- Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China.,Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China
| | - Hao Li
- Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China.,Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China
| | - Hao Cao
- Department of Cardiovascular Surgery, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| | - Yun Dong
- Department of Ultrasound in Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| | - Lei Gao
- Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| | - Zhongmin Liu
- Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China. .,Department of Cardiovascular Surgery, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China.
| | - Junbo Ge
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital of Fudan University, Shanghai, 200032, China.
| | - Hongming Zhu
- Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China.
| |
Collapse
|
210
|
Zhang S, Chen R, Chakrabarti S, Su Z. Resident macrophages as potential therapeutic targets for cardiac ageing and injury. Clin Transl Immunology 2020; 9:e1167. [PMID: 32874584 PMCID: PMC7450172 DOI: 10.1002/cti2.1167] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 07/21/2020] [Accepted: 08/01/2020] [Indexed: 12/12/2022] Open
Abstract
Cardiac‐resident macrophages (CRMs) play critical roles in maintaining cardiac homoeostasis and removing senescent and dying cells. Recent preclinical data have re‐energised the area of cardioimmunology and provided improved understanding of the modulation of compositional and functional phenotypes of CRMs. These data can aid in achieving improved cardiac regeneration, repair and functional remodelling following cardiac injury. In this review, we discuss the composition and renewal of various subsets of CRMs. Specific attention has been given to delineate the roles of various CRM subsets with respect to (1) facilitation of cardiac development and maintenance of physiological function such as electrical conduction and rhythm; (2) promotion of cardiac regeneration, inflammation resolution and functional remodelling following a cardiac injury; and (3) therapeutic potential. We have also highlighted the relationship between CRM replenishment and cardiomyocyte senescence as well as cardiovascular diseases development. Finally, we have addressed future perspectives and directions in basic research and potentially clinical applications of CRMs.
Collapse
Affiliation(s)
- Shiqing Zhang
- International Genome Center Jiangsu University Zhenjiang China.,Department of Immunology Jiangsu University Zhenjiang China
| | - Rong Chen
- International Genome Center Jiangsu University Zhenjiang China.,Department of Immunology Jiangsu University Zhenjiang China
| | | | - Zhaoliang Su
- International Genome Center Jiangsu University Zhenjiang China.,Department of Immunology Jiangsu University Zhenjiang China.,Laboratory Center The Fourth Affiliated Hospital of Jiangsu University Zhenjiang China
| |
Collapse
|
211
|
Tobin SW, Alibhai FJ, Weisel RD, Li RK. Considering Cause and Effect of Immune Cell Aging on Cardiac Repair after Myocardial Infarction. Cells 2020; 9:E1894. [PMID: 32823583 PMCID: PMC7465938 DOI: 10.3390/cells9081894] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 08/11/2020] [Accepted: 08/12/2020] [Indexed: 12/16/2022] Open
Abstract
The importance of the immune system for cardiac repair following myocardial infarction is undeniable; however, the complex nature of immune cell behavior has limited the ability to develop effective therapeutics. This limitation highlights the need for a better understanding of the function of each immune cell population during the inflammatory and resolution phases of cardiac repair. The development of reliable therapies is further complicated by aging, which is associated with a decline in cell and organ function and the onset of cardiovascular and immunological diseases. Aging of the immune system has important consequences on heart function as both chronic cardiac inflammation and an impaired immune response to cardiac injury are observed in older individuals. Several studies have suggested that rejuvenating the aged immune system may be a valid therapeutic candidate to prevent or treat heart disease. Here, we review the basic patterns of immune cell behavior after myocardial infarction and discuss the autonomous and nonautonomous manners of hematopoietic stem cell and immune cell aging. Lastly, we identify prospective therapies that may rejuvenate the aged immune system to improve heart function such as anti-inflammatory and senolytic therapies, bone marrow transplant, niche remodeling and regulation of immune cell differentiation.
Collapse
Affiliation(s)
- Stephanie W. Tobin
- Division of Cardiovascular Surgery, Toronto General Hospital Research Institute, University Health Network, Toronto, ON M5T 1P5, Canada; (S.W.T.); (F.J.A.); (R.D.W.)
| | - Faisal J. Alibhai
- Division of Cardiovascular Surgery, Toronto General Hospital Research Institute, University Health Network, Toronto, ON M5T 1P5, Canada; (S.W.T.); (F.J.A.); (R.D.W.)
| | - Richard D. Weisel
- Division of Cardiovascular Surgery, Toronto General Hospital Research Institute, University Health Network, Toronto, ON M5T 1P5, Canada; (S.W.T.); (F.J.A.); (R.D.W.)
- Division of Cardiac Surgery, Peter Munk Cardiac Centre, Toronto General Hospital and University of Toronto, Toronto, ON M5G 2N2, Canada
| | - Ren-Ke Li
- Division of Cardiovascular Surgery, Toronto General Hospital Research Institute, University Health Network, Toronto, ON M5T 1P5, Canada; (S.W.T.); (F.J.A.); (R.D.W.)
- Division of Cardiac Surgery, Peter Munk Cardiac Centre, Toronto General Hospital and University of Toronto, Toronto, ON M5G 2N2, Canada
| |
Collapse
|
212
|
Qi Z, Wu D, Li M, Yan Z, Yang X, Ji N, Wang Y, Zhang J. The pluripotent role of exosomes in mediating non-coding RNA in ventricular remodeling after myocardial infarction. Life Sci 2020; 254:117761. [PMID: 32413403 DOI: 10.1016/j.lfs.2020.117761] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Revised: 05/03/2020] [Accepted: 05/05/2020] [Indexed: 12/21/2022]
Abstract
With the increase of an aging population and the rising incidence of cardiovascular diseases, heart failure (HF) patients are on the rise every year. Myocardial infarction (MI) is the leading cause of HF in patients among cardiovascular diseases. In clinic, patients with MI are often assessed by biochemical indicators, electrocardiography, brain natriuretic peptide levels, myocardial enzymology, echocardiography and other means to predict the occurrence of HF and ventricular remodeling (VR). But there is still a lack of more accurate evaluation. VR is the basic mechanism of HF. In recent years, the molecular mechanism of VR has been studied mainly from the aspects of myocardial hypertrophy, myocardial fibrosis, inflammation, myocardial energy disorder, apoptosis, autophagy and pyroptosis. Exosomes are considered as the main mediators of intercellular information transmission. In addition, exosomes can promote the migration and transformation of intercellular RNAs, which are highly conserved non-coding RNAs. They can mediate the process of cell proliferation and differentiation of the target cell membrane. Exosomes have protective effects on VR after MI by inhibiting fibrosis, promoting angiogenesis and inhibiting inflammation and pyroptosis. We reviewed the specific protective mechanisms of exosomes for VR after MI. In addition, we discussed the formation of targeted exosomes and the role of non-coding RNAs in VR.
Collapse
Affiliation(s)
- Zhongwen Qi
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300183, China
| | - Dan Wu
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China
| | - Meng Li
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300183, China
| | - Zhipeng Yan
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China
| | - Xiaoya Yang
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China
| | - Nan Ji
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China
| | - Yueyao Wang
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China
| | - Junping Zhang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300183, China.
| |
Collapse
|
213
|
Daseke MJ, Tenkorang-Impraim MAA, Ma Y, Chalise U, Konfrst SR, Garrett MR, DeLeon-Pennell KY, Lindsey ML. Exogenous IL-4 shuts off pro-inflammation in neutrophils while stimulating anti-inflammation in macrophages to induce neutrophil phagocytosis following myocardial infarction. J Mol Cell Cardiol 2020; 145:112-121. [PMID: 32574573 PMCID: PMC7483959 DOI: 10.1016/j.yjmcc.2020.06.006] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Revised: 06/08/2020] [Accepted: 06/11/2020] [Indexed: 12/26/2022]
Abstract
INTRODUCTION Macrophages and neutrophils are primary leukocytes involved in the inflammatory response to myocardial infarction (MI). While interleukin (IL)-4 is an in vitro anti-inflammatory stimulus, the MI myocardium does not express a considerable amount of IL-4 but does express IL4 receptors. We hypothesized that continuous exogenous IL-4 infusion starting 24 h after MI would promote a polarization switch in inflammatory cells towards a reparative phenotype. METHODS C57BL/6J male mice (3-6 months of age) were subcutaneously infused with either saline (n = 17) or IL-4 (20 ng/g/day; n = 17) beginning 24 h after MI and evaluated at MI day 3. RESULTS Macrophages and neutrophils were isolated ex vivo from the infarct region and examined. Exogenous IL-4 decreased pro-inflammatory Ccl3, Il12a, Tnfa, and Tgfb1 in neutrophils and increased anti-inflammatory Arg1 and Ym1 in macrophages (all p < .05). Tissue clearance by IL-4 treated neutrophils was not different, while selective phagocytosis of neutrophils doubled in IL-4 treated macrophages (p < .05). Of 24,339 genes examined by RNA-sequencing, 2042 genes were differentially expressed in macrophages from IL-4 stimulated infarct (all FDR p < .05). Pdgfc gene expression was ranked first, increasing 3-fold in macrophages stimulated with IL-4 (p = 1 × 10-9). Importantly, changes in macrophage physiology and transcriptome occurred in the absence of global LV effects. Bone marrow derived monocytes stimulated with mouse recombinant PDGF-CC protein (10 μg/ml) or PDGF-CC blocking antibody (200 ng/ml) did not change Arg1 or Ym1 expression, indicating the in vivo effect of IL-4 to stimulate macrophage anti-inflammatory gene expression was independent of PDGF-CC. CONCLUSIONS Our results indicate that exogenous IL-4 promotes inflammation resolution by turning off pro-inflammation in neutrophils while stimulating anti-inflammation in macrophages to mediate removal of apoptotic neutrophils.
Collapse
Affiliation(s)
- Michael J Daseke
- Department of Cellular and Integrative Physiology, Center for Heart and Vascular Research, University of Nebraska Medical Center, Omaha, NE, USA; Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS, USA; Research Service, Nebraska-Western Iowa Health Care System, Omaha, NE, USA
| | - Mavis A A Tenkorang-Impraim
- Department of Cellular and Integrative Physiology, Center for Heart and Vascular Research, University of Nebraska Medical Center, Omaha, NE, USA
| | - Yonggang Ma
- Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, FL, USA
| | - Upendra Chalise
- Department of Cellular and Integrative Physiology, Center for Heart and Vascular Research, University of Nebraska Medical Center, Omaha, NE, USA; Research Service, Nebraska-Western Iowa Health Care System, Omaha, NE, USA
| | - Shelby R Konfrst
- Department of Cellular and Integrative Physiology, Center for Heart and Vascular Research, University of Nebraska Medical Center, Omaha, NE, USA; Research Service, Nebraska-Western Iowa Health Care System, Omaha, NE, USA
| | - Michael R Garrett
- Department of Pharmacology, University of Mississippi Medical Center, Jackson, MS, USA
| | - Kristine Y DeLeon-Pennell
- Ralph H. Johnson Veterans Affairs Medical Center, Charleston, SC, USA; Division of Cardiology, Department of Medicine, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Merry L Lindsey
- Department of Cellular and Integrative Physiology, Center for Heart and Vascular Research, University of Nebraska Medical Center, Omaha, NE, USA; Research Service, Nebraska-Western Iowa Health Care System, Omaha, NE, USA.
| |
Collapse
|
214
|
Jin L, Zhang Y, Liang W, Lu X, Piri N, Wang W, Kaplan HJ, Dean DC, Zhang L, Liu Y. Zeb1 promotes corneal neovascularization by regulation of vascular endothelial cell proliferation. Commun Biol 2020; 3:349. [PMID: 32620870 PMCID: PMC7335040 DOI: 10.1038/s42003-020-1069-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Accepted: 06/08/2020] [Indexed: 12/12/2022] Open
Abstract
Angiogenesis is required for tissue repair; but abnormal angiogenesis or neovascularization (NV) causes diseases in the eye. The avascular status in the cornea is a prerequisite for corneal clarity and thought to be maintained by the equilibrium between proangiogenic and antiangiogenic factors that controls proliferation and migration of vascular endothelial cells (ECs) sprouting from the pericorneal plexus. VEGF is the most important intrinsic factor for angiogenesis; anti-VEGF therapies are available for treating ocular NV. However, the effectiveness of the therapies is limited because of VEGF-independent mechanism(s). We show that Zeb1 is an important factor promoting vascular EC proliferation and corneal NV; and a couple of small molecule inhibitors can evict Ctbp from the Zeb1-Ctbp complex, thereby reducing EC Zeb1 expression, proliferation, and corneal NV. We conclude that Zeb1-regulation of angiogenesis is independent of Vegf and that the ZEB1-CtBP inhibitors can be of potential therapeutic significance in treating corneal NV.
Collapse
Affiliation(s)
- Lei Jin
- Department of Ophthalmology and Visual Sciences, University of Louisville School of Medicine, Louisville, KY, 40202, USA
- Department of Ophthalmology, The Third People's Hospital of Dalian, Dalian Medical University, Dalian, 116033, China
| | - Yingnan Zhang
- Department of Ophthalmology and Visual Sciences, University of Louisville School of Medicine, Louisville, KY, 40202, USA
- Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing Ophthalmology & Visual Science Key Lab, Beijing, 100730, China
| | - Wei Liang
- Department of Ophthalmology and Visual Sciences, University of Louisville School of Medicine, Louisville, KY, 40202, USA
- Department of Ophthalmology, The Third People's Hospital of Dalian, Dalian Medical University, Dalian, 116033, China
| | - Xiaoqin Lu
- Department of Ophthalmology and Visual Sciences, University of Louisville School of Medicine, Louisville, KY, 40202, USA
| | - Niloofar Piri
- Department of Ophthalmology and Visual Sciences, University of Louisville School of Medicine, Louisville, KY, 40202, USA
| | - Wei Wang
- Department of Ophthalmology and Visual Sciences, University of Louisville School of Medicine, Louisville, KY, 40202, USA
| | - Henry J Kaplan
- Department of Ophthalmology and Visual Sciences, University of Louisville School of Medicine, Louisville, KY, 40202, USA
| | - Douglas C Dean
- Department of Ophthalmology and Visual Sciences, University of Louisville School of Medicine, Louisville, KY, 40202, USA.
- Birth Defects Center, University of Louisville School of Dentistry, Louisville, KY, 40202, USA.
- James Brown Cancer Center, University of Louisville School of Medicine, Louisville, KY, 40202, USA.
| | - Lijun Zhang
- Department of Ophthalmology, The Third People's Hospital of Dalian, Dalian Medical University, Dalian, 116033, China.
| | - Yongqing Liu
- Department of Ophthalmology and Visual Sciences, University of Louisville School of Medicine, Louisville, KY, 40202, USA.
- Birth Defects Center, University of Louisville School of Dentistry, Louisville, KY, 40202, USA.
- James Brown Cancer Center, University of Louisville School of Medicine, Louisville, KY, 40202, USA.
| |
Collapse
|
215
|
Yang D, Liu HQ, Liu FY, Tang N, Guo Z, Ma SQ, An P, Wang MY, Wu HM, Yang Z, Fan D, Tang QZ. The Roles of Noncardiomyocytes in Cardiac Remodeling. Int J Biol Sci 2020; 16:2414-2429. [PMID: 32760209 PMCID: PMC7378633 DOI: 10.7150/ijbs.47180] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2020] [Accepted: 06/16/2020] [Indexed: 02/07/2023] Open
Abstract
Cardiac remodeling is a common characteristic of almost all forms of heart disease, including cardiac infarction, valvular diseases, hypertension, arrhythmia, dilated cardiomyopathy and other conditions. It is not merely a simple outcome induced by an increase in the workload of cardiomyocytes (CMs). The remodeling process is accompanied by abnormalities of cardiac structure as well as disturbance of cardiac function, and emerging evidence suggests that a wide range of cells in the heart participate in the initiation and development of cardiac remodeling. Other than CMs, there are numerous noncardiomyocytes (non-CMs) that regulate the process of cardiac remodeling, such as cardiac fibroblasts and immune cells (including macrophages, lymphocytes, neutrophils, and mast cells). In this review, we summarize recent knowledge regarding the definition and significant effects of various non-CMs in the pathogenesis of cardiac remodeling, with a particular emphasis on the involved signaling mechanisms. In addition, we discuss the properties of non-CMs, which serve as targets of many cardiovascular drugs that reduce adverse cardiac remodeling.
Collapse
Affiliation(s)
- Dan Yang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, RP China
- Cardiovascular Research Institute of Wuhan University, Wuhan 430060, RP China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan 430060, RP China
| | - Han-Qing Liu
- Department of Thyroid and Breast, Renmin Hospital of Wuhan University, Wuhan 430060, RP China
| | - Fang-Yuan Liu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, RP China
- Cardiovascular Research Institute of Wuhan University, Wuhan 430060, RP China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan 430060, RP China
| | - Nan Tang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, RP China
- Cardiovascular Research Institute of Wuhan University, Wuhan 430060, RP China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan 430060, RP China
| | - Zhen Guo
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, RP China
- Cardiovascular Research Institute of Wuhan University, Wuhan 430060, RP China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan 430060, RP China
| | - Shu-Qing Ma
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, RP China
- Cardiovascular Research Institute of Wuhan University, Wuhan 430060, RP China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan 430060, RP China
| | - Peng An
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, RP China
- Cardiovascular Research Institute of Wuhan University, Wuhan 430060, RP China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan 430060, RP China
| | - Ming-Yu Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, RP China
- Cardiovascular Research Institute of Wuhan University, Wuhan 430060, RP China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan 430060, RP China
| | - Hai-Ming Wu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, RP China
- Cardiovascular Research Institute of Wuhan University, Wuhan 430060, RP China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan 430060, RP China
| | - Zheng Yang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, RP China
- Cardiovascular Research Institute of Wuhan University, Wuhan 430060, RP China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan 430060, RP China
| | - Di Fan
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, RP China
- Cardiovascular Research Institute of Wuhan University, Wuhan 430060, RP China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan 430060, RP China
| | - Qi-Zhu Tang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, RP China
- Cardiovascular Research Institute of Wuhan University, Wuhan 430060, RP China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan 430060, RP China
| |
Collapse
|
216
|
Zhao M, Wang DDH, Liu X, Tian R. Metabolic Modulation of Macrophage Function Post Myocardial Infarction. Front Physiol 2020; 11:674. [PMID: 32695016 PMCID: PMC7338762 DOI: 10.3389/fphys.2020.00674] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Accepted: 05/26/2020] [Indexed: 02/05/2023] Open
Abstract
Macrophages are key components of innate immunity, and they play critical roles in heart health and diseases. Following acute myocardial infarction (MI), infiltrating macrophages undergo drastic phenotypic transition from pro-inflammatory in the early stage to pro-healing in the late stage. Transcriptome analyses of macrophage in the infarct zone show a time-dependent reprogramming of mitochondrial and metabolic functions, which parallels the changes of macrophage function. These observations suggest that mitochondrial and metabolic targets could be exploited for therapeutic opportunities. In this article, we reviewed the recent work on immunometabolic features of macrophage over the MI time continuum. In addition, we summarized currently proposed mitochondrial pathways involved in the functional polarization of macrophage and discussed their potential relevance to the outcome of MI. We expect that these findings will stimulate further investigations in metabolic modulation of innate immunity in the post-MI setting, which could ultimately lead to new strategies for therapy.
Collapse
Affiliation(s)
- Mingyue Zhao
- Laboratory of Cardiovascular Diseases, Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, China
| | - Dennis Ding-Hwa Wang
- Mitochondria and Metabolism Center, Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, WA, United States.,Division of Cardiology, Department of Medicine, University of Washington, Seattle, WA, United States
| | - Xiaojing Liu
- Laboratory of Cardiovascular Diseases, Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, China.,Department of Cardiology, West China Hospital, Sichuan University, Chengdu, China
| | - Rong Tian
- Mitochondria and Metabolism Center, Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, WA, United States
| |
Collapse
|
217
|
O'Brien M, Baicu CF, Van Laer AO, Zhang Y, McDonald LT, LaRue AC, Zile MR, Bradshaw AD. Pressure overload generates a cardiac-specific profile of inflammatory mediators. Am J Physiol Heart Circ Physiol 2020; 319:H331-H340. [PMID: 32589444 DOI: 10.1152/ajpheart.00274.2020] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Mechanisms that contribute to myocardial fibrosis, particularly in response to left ventricular pressure overload (LVPO), remain poorly defined. To test the hypothesis that a myocardial-specific profile of secreted factors is produced in response to PO, levels of 44 factors implicated in immune cell recruitment and function were assessed in a murine model of cardiac hypertrophy and compared with levels produced in a model of pulmonary fibrosis (PF). Mice subjected to PO were assessed at 1 and 4 wk. Protein from plasma, LV, lungs, and kidneys were analyzed by specific protein array analysis in parallel with protein from mice subjected to silica-instilled PF. Of the 44 factors assessed, 13 proteins were elevated in 1-wk PO myocardium, whereas 18 proteins were found increased in fibrotic lung. Eight of those increased in 1-wk LVPO were not found to be increased in fibrotic lungs (CCL-11, CCL-12, CCL-17, CCL-19, CCL-21, CCL-22, IL-16, and VEGF). Additionally, six factors were increased in plasma of 1-wk LVPO in the absence of increases in myocardial levels. In contrast, in mice with PF, no factors were found increased in plasma that were not elevated in lung tissue. Of those factors increased at 1 wk, only TIMP-1 remained elevated at 4 wk of LVPO. Immunohistochemistry of myocardial vasculature at 1 and 4 wk revealed similar amounts of total vasculature; however, evidence of activated endothelium was observed at 1 wk and, to a lesser extent, at 4 wk LVPO. In conclusion, PO myocardium generated a unique signature of cytokine expression versus that of fibrotic lung.NEW & NOTEWORTHY Myocardial fibrosis and the resultant increases in myocardial stiffness represent pivotal consequences of chronic pressure overload (PO). In this study, cytokine profiles produced in a murine model of cardiac fibrosis induced by PO were compared with those produced in response to silica-induced lung fibrosis. A unique profile of cardiac tissue-specific and plasma-derived factors generated in response to PO are reported.
Collapse
Affiliation(s)
- Matthew O'Brien
- Division of Cardiology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina
| | - Catalin F Baicu
- Division of Cardiology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina
| | - An O Van Laer
- Division of Cardiology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina
| | - Yuhua Zhang
- Division of Cardiology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina
| | - Lindsey T McDonald
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, South Carolina.,Ralph H. Johnson Veterans Affairs Medical Center, Charleston, South Carolina
| | - Amanda C LaRue
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, South Carolina.,Ralph H. Johnson Veterans Affairs Medical Center, Charleston, South Carolina
| | - Michael R Zile
- Division of Cardiology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina.,Ralph H. Johnson Veterans Affairs Medical Center, Charleston, South Carolina
| | - Amy D Bradshaw
- Division of Cardiology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina.,Ralph H. Johnson Veterans Affairs Medical Center, Charleston, South Carolina
| |
Collapse
|
218
|
Pagliaro P, Penna C. ACE/ACE2 Ratio: A Key Also in 2019 Coronavirus Disease (Covid-19)? Front Med (Lausanne) 2020; 7:335. [PMID: 32626721 PMCID: PMC7314898 DOI: 10.3389/fmed.2020.00335] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Accepted: 06/05/2020] [Indexed: 01/04/2023] Open
Affiliation(s)
- Pasquale Pagliaro
- Department of Clinical and Biological Sciences, University of Torino, Turin, Italy
| | | |
Collapse
|
219
|
Elder AM, Stoller AR, Black SA, Lyons TR. Macphatics and PoEMs in Postpartum Mammary Development and Tumor Progression. J Mammary Gland Biol Neoplasia 2020; 25:103-113. [PMID: 32535810 PMCID: PMC7395889 DOI: 10.1007/s10911-020-09451-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Accepted: 05/29/2020] [Indexed: 12/13/2022] Open
Abstract
Postpartum mammary gland involution is a mammalian tissue remodeling event that occurs after pregnancy and lactation to return the gland to the pre-pregnant state. This event is characterized by apoptosis and lysosomal-mediated cell death of the majority of the lactational mammary epithelium, followed by remodeling of the extracellular matrix, influx of immune cell populations (in particular, T helper cells, monocytes, and macrophages), and neo-lymphangiogenesis. This postpartum environment has been shown to be promotional for tumor growth and metastases and may partially account for why women diagnosed with breast cancer during the postpartum period or within 5 years of last childbirth have an increased risk of developing metastases when compared to their nulliparous counterparts. The lymphatics and macrophages present during mammary gland involution have been implicated in promoting the observed growth and metastasis. Of importance are the macrophages, which are of the "M2" phenotype and are known to create a pro-tumor microenvironment. In this report, we describe a subset of postpartum macrophages that express lymphatic proteins (PoEMs) and directly interact with lymphatic vessels to form chimeric vessels or "macphatics". Additionally, these PoEMs are very similar to tumor-associated macrophages that also express lymphatic proteins and are present at the sites of lymphatic vessels where tumors escape the tissue and enter the lymphatic vasculature. Further characterizing these PoEMs may offer insight in preventing lymphatic metastasis of breast cancer, as well as provide information for how developmental programming of lymphatic endothelial cells and macrophages can contribute to different disease progression.
Collapse
Affiliation(s)
- Alan M Elder
- Young Women's Breast Cancer Translational Program, Division of Medical Oncology, University of Colorado Cancer Center, 12801 E 17th Ave, RC1 South, Mailstop 8117, Aurora, CO, 80045, USA
- Division of Medical Oncology, Anschutz Medical Center, University of Colorado, Aurora, CO, USA
- Graduate Program in Cancer Biology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Alexander R Stoller
- Young Women's Breast Cancer Translational Program, Division of Medical Oncology, University of Colorado Cancer Center, 12801 E 17th Ave, RC1 South, Mailstop 8117, Aurora, CO, 80045, USA
- Division of Medical Oncology, Anschutz Medical Center, University of Colorado, Aurora, CO, USA
| | - Sarah A Black
- University of Colorado School of Medicine, Aurora, CO, USA
| | - Traci R Lyons
- Young Women's Breast Cancer Translational Program, Division of Medical Oncology, University of Colorado Cancer Center, 12801 E 17th Ave, RC1 South, Mailstop 8117, Aurora, CO, 80045, USA.
- Division of Medical Oncology, Anschutz Medical Center, University of Colorado, Aurora, CO, USA.
- Graduate Program in Cancer Biology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
| |
Collapse
|
220
|
Wang X, Wang Q, Li W, Zhang Q, Jiang Y, Guo D, Sun X, Lu W, Li C, Wang Y. TFEB-NF-κB inflammatory signaling axis: a novel therapeutic pathway of Dihydrotanshinone I in doxorubicin-induced cardiotoxicity. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2020; 39:93. [PMID: 32448281 PMCID: PMC7245789 DOI: 10.1186/s13046-020-01595-x] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Accepted: 05/11/2020] [Indexed: 02/06/2023]
Abstract
BACKGROUND Doxorubicin is effective in a variety of solid and hematological malignancies. Unfortunately, clinical application of doxorubicin is limited due to a cumulative dose-dependent cardiotoxicity. Dihydrotanshinone I (DHT) is a natural product from Salvia miltiorrhiza Bunge with multiple anti-tumor activity and anti-inflammation effects. However, its anti-doxorubicin-induced cardiotoxicity (DIC) effect, either in vivo or in vitro, has not been elucidated yet. This study aims to explore the anti-inflammation effects of DHT against DIC, and to elucidate the potential regulatory mechanism. METHODS Effects of DHT on DIC were assessed in zebrafish, C57BL/6 mice and H9C2 cardiomyocytes. Echocardiography, histological examination, flow cytometry, immunochemistry and immunofluorescence were utilized to evaluate cardio-protective effects and anti-inflammation effects. mTOR agonist and lentivirus vector carrying GFP-TFEB were applied to explore the regulatory signaling pathway. RESULTS DHT improved cardiac function via inhibiting the activation of M1 macrophages and the excessive release of pro-inflammatory cytokines both in vivo and in vitro. The activation and nuclear localization of NF-κB were suppressed by DHT, and the effect was abolished by mTOR agonist with concomitant reduced expression of nuclear TFEB. Furthermore, reduced expression of nuclear TFEB is accompanied by up-regulated phosphorylation of IKKα/β and NF-κB, while TFEB overexpression reversed these changes. Intriguingly, DHT could upregulate nuclear expression of TFEB and reduce expressions of p-IKKα/β and p-NF-κB. CONCLUSIONS Our results demonstrated that DHT can be applied as a novel cardioprotective compound in the anti-inflammation management of DIC via mTOR-TFEB-NF-κB signaling pathway. The current study implicates TFEB-IKK-NF-κB signaling axis as a previously undescribed, druggable pathway for DIC.
Collapse
Affiliation(s)
- Xiaoping Wang
- grid.24695.3c0000 0001 1431 9176School of Life Science, Beijing University of Chinese Medicine, Beijing, 100029 China
| | - Qiyan Wang
- grid.24695.3c0000 0001 1431 9176School of Life Science, Beijing University of Chinese Medicine, Beijing, 100029 China
| | - Weili Li
- grid.24695.3c0000 0001 1431 9176School of Life Science, Beijing University of Chinese Medicine, Beijing, 100029 China
| | - Qian Zhang
- grid.24695.3c0000 0001 1431 9176School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029 China
| | - Yanyan Jiang
- grid.24695.3c0000 0001 1431 9176School of Life Science, Beijing University of Chinese Medicine, Beijing, 100029 China
| | - Dongqing Guo
- grid.24695.3c0000 0001 1431 9176School of Life Science, Beijing University of Chinese Medicine, Beijing, 100029 China
| | - Xiaoqian Sun
- grid.24695.3c0000 0001 1431 9176School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029 China
| | - Wenji Lu
- grid.24695.3c0000 0001 1431 9176School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029 China
| | - Chun Li
- grid.24695.3c0000 0001 1431 9176Modern Research Center for Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029 China
| | - Yong Wang
- grid.24695.3c0000 0001 1431 9176School of Life Science, Beijing University of Chinese Medicine, Beijing, 100029 China ,grid.24695.3c0000 0001 1431 9176School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029 China
| |
Collapse
|
221
|
Liu Y, Xu J, Wu M, Kang L, Xu B. The effector cells and cellular mediators of immune system involved in cardiac inflammation and fibrosis after myocardial infarction. J Cell Physiol 2020; 235:8996-9004. [PMID: 32352172 DOI: 10.1002/jcp.29732] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 04/11/2020] [Accepted: 04/15/2020] [Indexed: 01/05/2023]
Abstract
The cardiac repair after myocardial infarction (MI) involves two phases, namely, inflammatory response and proliferative response. The former is an inflammatory reaction, evoked by different kinds of pro-inflammatory leukocytes and molecules stimulated by myocardial necrosis, while the latter is a repair process, predominated by a magnitude of anti-inflammatory cells and cytokines, as well as fibroblasts. Cardiac remodeling post-MI is dependent on the balance of individualized intensity of the post-MI inflammation and subsequent cardiac fibrosis. During the past 30 years, enormous studies have focused on investigating immune cells and mediators involved in cardiac inflammation and fibrosis, which are two interacting processes of post-MI cardiac repair. These results contribute to revealing the mechanism of adverse cardiac remodeling after MI and alleviating the impairment of cardiac function. In this study, we will broadly discuss the role of immune cell subpopulation and the involved cytokines and chemokines during cardiac repair post-MI, particular in cardiac inflammation and fibrosis.
Collapse
Affiliation(s)
- Yihai Liu
- Department of Cardiology, Nanjing Drum Tower Hospital, Clinical college of Nanjing Medical University, Nanjing, China
| | - Jiamin Xu
- Department of Cardiology, Nanjing Drum Tower Hospital, Clinical college of Nanjing Medical University, Nanjing, China
| | - Mingyue Wu
- Department of Cardiology, Nanjing Drum Tower Hospital, Clinical college of Nanjing Medical University, Nanjing, China
| | - Lina Kang
- Department of Cardiology, Nanjing Drum Tower Hospital, Clinical college of Nanjing Medical University, Nanjing, China
| | - Biao Xu
- Department of Cardiology, Nanjing Drum Tower Hospital, Clinical college of Nanjing Medical University, Nanjing, China
| |
Collapse
|
222
|
Impact of BDNF Val66Met Polymorphism on Myocardial Infarction: Exploring the Macrophage Phenotype. Cells 2020; 9:cells9051084. [PMID: 32349267 PMCID: PMC7290372 DOI: 10.3390/cells9051084] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 04/22/2020] [Accepted: 04/24/2020] [Indexed: 12/13/2022] Open
Abstract
Brain-derived neurotrophic factor (BDNF) is a member of the neurotrophin growth factor family, well known for its role in the homeostasis of the cardiovascular system. Recently, the human BDNF Val66Met single nucleotide polymorphism has been associated with the increased propensity for arterial thrombosis related to acute myocardial infarction (AMI). Using cardiac magnetic resonance imaging and immunohistochemistry analyses, we showed that homozygous mice carrying the human BDNF Val66Met polymorphism (BDNFMet/Met) undergoing left anterior descending (LAD) coronary artery ligation display an adverse cardiac remodeling compared to wild-type (BDNFVal/Val). Interestingly, we observed a persistent presence of pro-inflammatory M1-like macrophages and a reduced accumulation of reparative-like phenotype macrophages (M2-like) in the infarcted heart of mutant mice. Further qPCR analyses showed that BDNFMet/Met peritoneal macrophages are more pro-inflammatory and have a higher migratory ability compared to BDNFVal/Val ones. Finally, macrophages differentiated from circulating monocytes isolated from BDNFMet/Met patients with coronary heart disease displayed the same pro-inflammatory characteristics of the murine ones. In conclusion, the BDNF Val66Met polymorphism predisposes to adverse cardiac remodeling after myocardial infarction in a mouse model and affects macrophage phenotype in both humans and mice. These results provide a new cellular mechanism by which this human BDNF genetic variant could influence cardiovascular disease.
Collapse
|
223
|
Wagner MJ, Khan M, Mohsin S. Healing the Broken Heart; The Immunomodulatory Effects of Stem Cell Therapy. Front Immunol 2020; 11:639. [PMID: 32328072 PMCID: PMC7160320 DOI: 10.3389/fimmu.2020.00639] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Accepted: 03/20/2020] [Indexed: 12/14/2022] Open
Abstract
Cardiovascular Disease (CVD) is a leading cause of mortality within the United States. Current treatments being administered to patients who suffered a myocardial infarction (MI) have increased patient survival, but do not facilitate the replacement of damaged myocardium. Recent studies demonstrate that stem cell-based therapies promote myocardial repair; however, the poor engraftment of the transferred stem cell populations within the infarcted myocardium is a major limitation, regardless of the cell type. One explanation for poor cell retention is attributed to the harsh inflammatory response mounted following MI. The inflammatory response coupled to cardiac repair processes is divided into two distinct phases. The first phase is initiated during ischemic injury when necrosed myocardium releases Danger Associated Molecular Patterns (DAMPs) and chemokines/cytokines to induce the activation and recruitment of neutrophils and pro-inflammatory M1 macrophages (MΦs); in turn, facilitating necrotic tissue clearance. During the second phase, a shift from the M1 inflammatory functional phenotype to the M2 anti-inflammatory and pro-reparative functional phenotype, permits the resolution of inflammation and the establishment of tissue repair. T-regulatory cells (Tregs) are also influential in mediating the establishment of the pro-reparative phase by directly regulating M1 to M2 MΦ differentiation. Current studies suggest CD4+ T-lymphocyte populations become activated when presented with autoantigens released from the injured myocardium. The identity of the cardiac autoantigens or paracrine signaling molecules released from the ischemic tissue that directly mediate the phenotypic plasticity of T-lymphocyte populations in the post-MI heart are just beginning to be elucidated. Stem cells are enriched centers that contain a diverse paracrine secretome that can directly regulate responses within neighboring cell populations. Previous studies identify that stem cell mediated paracrine signaling can influence the phenotype and function of immune cell populations in vitro, but how stem cells directly mediate the inflammatory microenvironment of the ischemic heart is poorly characterized and is a topic of extensive investigation. In this review, we summarize the complex literature that details the inflammatory microenvironment of the ischemic heart and provide novel insights regarding how paracrine mediated signaling produced by stem cell-based therapies can regulate immune cell subsets to facilitate pro-reparative myocardial wound healing.
Collapse
Affiliation(s)
- Marcus J Wagner
- Independence Blue Cross Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| | - Mohsin Khan
- Center for Metabolic Disease, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States.,Department of Physiology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| | - Sadia Mohsin
- Independence Blue Cross Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States.,Department of Pharmacology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| |
Collapse
|
224
|
Okyere AD, Tilley DG. Leukocyte-Dependent Regulation of Cardiac Fibrosis. Front Physiol 2020; 11:301. [PMID: 32322219 PMCID: PMC7156539 DOI: 10.3389/fphys.2020.00301] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Accepted: 03/17/2020] [Indexed: 12/24/2022] Open
Abstract
Cardiac fibrosis begins as an intrinsic response to injury or ageing that functions to preserve the tissue from further damage. Fibrosis results from activated cardiac myofibroblasts, which secrete extracellular matrix (ECM) proteins in an effort to replace damaged tissue; however, excessive ECM deposition leads to pathological fibrotic remodeling. At this extent, fibrosis gravely disturbs myocardial compliance, and ultimately leads to adverse outcomes like heart failure with heightened mortality. As such, understanding the complexity behind fibrotic remodeling has been a focal point of cardiac research in recent years. Resident cardiac fibroblasts and activated myofibroblasts have been proven integral to the fibrotic response; however, several findings point to additional cell types that may contribute to the development of pathological fibrosis. For one, leukocytes expand in number after injury and exhibit high plasticity, thus their distinct role(s) in cardiac fibrosis is an ongoing and controversial field of study. This review summarizes current findings, focusing on both direct and indirect leukocyte-mediated mechanisms of fibrosis, which may provide novel targeted strategies against fibrotic remodeling.
Collapse
Affiliation(s)
- Ama Dedo Okyere
- Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| | - Douglas G Tilley
- Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| |
Collapse
|
225
|
Bloise N, Rountree I, Polucha C, Montagna G, Visai L, Coulombe KLK, Munarin F. Engineering Immunomodulatory Biomaterials for Regenerating the Infarcted Myocardium. Front Bioeng Biotechnol 2020; 8:292. [PMID: 32318563 PMCID: PMC7154131 DOI: 10.3389/fbioe.2020.00292] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Accepted: 03/19/2020] [Indexed: 12/13/2022] Open
Abstract
Coronary artery disease is a severe ischemic condition characterized by the reduction of blood flow in the arteries of the heart that results in the dysfunction and death of cardiac tissue. Despite research over several decades on how to reduce long-term complications and promote angiogenesis in the infarct, the medical field has yet to define effective treatments for inducing revascularization in the ischemic tissue. With this work, we have developed functional biomaterials for the controlled release of immunomodulatory cytokines to direct immune cell fate for controlling wound healing in the ischemic myocardium. The reparative effects of colony-stimulating factor (CSF-1), and anti-inflammatory interleukins 4/6/13 (IL4/6/13) have been evaluated in vitro and in a predictive in vivo model of ischemia (the skin flap model) to optimize a new immunomodulatory biomaterial that we use for treating infarcted rat hearts. Alginate hydrogels have been produced by internal gelation with calcium carbonate (CaCO3) as carriers for the immunomodulatory cues, and their stability, degradation, rheological properties and release kinetics have been evaluated in vitro. CD14 positive human peripheral blood monocytes treated with the immunomodulatory biomaterials show polarization into pro-healing macrophage phenotypes. Unloaded and CSF-1/IL4 loaded alginate gel formulations have been implanted in skin flap ischemic wounds to test the safety and efficacy of the delivery system in vivo. Faster wound healing is observed with the new therapeutic treatment, compared to the wounds treated with the unloaded controls at day 14. The optimized therapy has been evaluated in a rat model of myocardial infarct (ischemia/reperfusion). Macrophage polarization toward healing phenotypes and global cardiac function measured with echocardiography and immunohistochemistry at 4 and 15 days demonstrate the therapeutic potential of the proposed immunomodulatory treatment in a clinically relevant infarct model.
Collapse
Affiliation(s)
- Nora Bloise
- Department of Molecular Medicine, Center for Health Technologies (CHT), INSTM UdR of Pavia, University of Pavia, Pavia, Italy.,Department of Occupational Medicine, Toxicology and Environmental Risks, ICS Maugeri, IRCCS, Pavia, Italy
| | - Isobel Rountree
- Center for Biomedical Engineering, School of Engineering, Brown University, Providence, RI, United States
| | - Collin Polucha
- Center for Biomedical Engineering, School of Engineering, Brown University, Providence, RI, United States
| | - Giulia Montagna
- Department of Molecular Medicine, Center for Health Technologies (CHT), INSTM UdR of Pavia, University of Pavia, Pavia, Italy.,Department of Electrical, Computer and Biomedical Engineering, Centre for Health Technologies (CHT), University of Pavia, Pavia, Italy
| | - Livia Visai
- Department of Molecular Medicine, Center for Health Technologies (CHT), INSTM UdR of Pavia, University of Pavia, Pavia, Italy.,Department of Occupational Medicine, Toxicology and Environmental Risks, ICS Maugeri, IRCCS, Pavia, Italy
| | - Kareen L K Coulombe
- Center for Biomedical Engineering, School of Engineering, Brown University, Providence, RI, United States
| | - Fabiola Munarin
- Center for Biomedical Engineering, School of Engineering, Brown University, Providence, RI, United States
| |
Collapse
|
226
|
Li N, Wang G, Lu H. WITHDRAWN: Development of Alzheimer's Disease after Myocardial Infarction. J Alzheimers Dis 2020:JAD200068. [PMID: 32280100 DOI: 10.3233/jad-200068] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Ahead of Print article withdrawn by publisher.
Collapse
Affiliation(s)
- Naiyan Li
- Department of Cardiac Surgery, People's Hospital of Linyi City, Linyi, China
| | - Gang Wang
- Department of Cardiac Surgery, People's Hospital of Linyi City, Linyi, China
| | - Huarong Lu
- Department of Digestion Medicine, People's Hospital of Linyi City, Linyi, China
| |
Collapse
|
227
|
Susca MG, Hodas R, Benedek T, Benedek I, Chitu M, Opincariu D, Chiotoroiu A, Rezus C. Impact of cardiac rehabilitation programs on left ventricular remodeling after acute myocardial infarction: Study Protocol Clinical Trial (SPIRIT Compliant). Medicine (Baltimore) 2020; 99:e19759. [PMID: 32311978 PMCID: PMC7220465 DOI: 10.1097/md.0000000000019759] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Accepted: 03/05/2020] [Indexed: 11/25/2022] Open
Abstract
INTRODUCTION While the role of early mobilization in the immediate postinfarction period has been well demonstrated, little is known in present about the link between early mobilization and reduction of systemic inflammation. At the same time, the impact of early mobilization on regression of left ventricular remodeling has not been elucidated so far. MATERIAL AND METHODS Here we present the study protocol of the REHAB trial, a clinical descriptive, prospective study, conducted in a single-center, with the purpose to analyze the impact of early mobilization in reducing left ventricular remodeling, the complication rates and mortality in patients who had suffered a recent acute myocardial infarction (AMI). At the same time, the study aims to demonstrate the contribution of early mobilization to reduction of systemic inflammation, thus reducing the inflammation-mediated ventricular remodeling. 100 patients with AMI in the last 12 hours, and successful revascularization of the culprit artery within the first 12 hours after the onset of symptoms in ST-segment elevation acute myocardial infarction or within first 48 hours in non ST-segment elevation AMI will be enrolled in the study. Based on the moment of mobilization after AMI patients will be distributed in 2 groups: group 1 - patients with early mobilization (<2 days after the onset of symptoms) and; group 2 - subjects with delayed mobilization after AMI (>2 days after the onset of symptoms). Study outcomes will consist in the impact of early mobilization after AMI on the ventricular remodeling in the post-infarction period, as assessed by cardiac magnetic resonance imaging, the rate of in-hospital mortality, the rate of repeated revascularization or MACE and the effect of early mobilization on systemic inflammation in the immediate postinfarction phase. CONCLUSION In conclusion, REHAB will be the first trial that will elucidate the impact of early mobilization in the first period after AMI, as a first step of a complex cardiac rehabilitation program, to reduce systemic inflammation and prevent deleterious ventricular remodeling in patients who suffered a recent AMI.
Collapse
Affiliation(s)
| | | | - Theodora Benedek
- Clinic of Cardiology, University of Medicine, Pharmacy, Sciences and Technology of Targu Mures
- Department of Advanced Research in Multimodality Cardiovascular Imaging, Cardio Med Medical Center, Targu Mures
| | - Imre Benedek
- Clinic of Cardiology, University of Medicine, Pharmacy, Sciences and Technology of Targu Mures
| | - Monica Chitu
- Clinic of Cardiology, University of Medicine, Pharmacy, Sciences and Technology of Targu Mures
| | - Diana Opincariu
- Clinic of Cardiology, University of Medicine, Pharmacy, Sciences and Technology of Targu Mures
- Department of Advanced Research in Multimodality Cardiovascular Imaging, Cardio Med Medical Center, Targu Mures
| | - Andreea Chiotoroiu
- University of Medicine, Pharmacy, Sciences and Technology of Targu Mures
| | - Ciprian Rezus
- University of Medicine and Pharmacy ‘Gr.T.Popa’, Iasi, Romania
| |
Collapse
|
228
|
Kimura T, Tajiri K, Sato A, Sakai S, Wang Z, Yoshida T, Uede T, Hiroe M, Aonuma K, Ieda M, Imanaka-Yoshida K. Tenascin-C accelerates adverse ventricular remodelling after myocardial infarction by modulating macrophage polarization. Cardiovasc Res 2020; 115:614-624. [PMID: 30295707 DOI: 10.1093/cvr/cvy244] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2018] [Revised: 09/03/2018] [Accepted: 10/05/2018] [Indexed: 12/13/2022] Open
Abstract
AIMS Tenascin-C (TN-C) is an extracellular matrix protein undetected in the normal adult heart, but expressed in several heart diseases associated with inflammation. We previously reported that serum TN-C levels of myocardial infarction (MI) patients were elevated during the acute stage, and that patients with high peak TN-C levels were at high risk of left ventricular (LV) remodelling and poor outcome, suggesting that TN-C could play a significant role in the progression of ventricular remodelling. However, the detailed molecular mechanisms associated with this process remain unknown. We aimed to elucidate the role and underlying mechanisms associated with TN-C in adverse remodelling after MI. METHODS AND RESULTS MI was induced by permanent ligation of the coronary artery of TN-C knockout (TN-C-KO) and wild type (WT) mice. In WT mice, TN-C was expressed at the borders between intact and necrotic areas, with a peak at 3 days post-MI and observed in the immediate vicinity of infiltrating macrophages. TN-C-KO mice were protected from ventricular adverse remodelling as evidenced by a higher LV ejection fraction as compared with WT mice (19.0 ± 6.3% vs. 10.6 ± 4.4%; P < 0.001) at 3 months post-MI. During the acute phase, flow-cytometric analyses showed a decrease in F4/80+CD206lowCD45+ M1 macrophages and an increase in F4/80+CD206highCD45+ M2 macrophages in the TN-C-KO heart. To clarify the role of TN-C on macrophage polarization, we examined the direct effect of TN-C on bone marrow-derived macrophages in culture, observing that TN-C promoted macrophage shifting into an M1 phenotype via Toll-like receptor 4 (TLR4). Under M2-skewing conditions, TN-C suppressed the expression of interferon regulatory factor 4, a key transcription factor that controls M2-macrophage polarization, via TLR4, thereby inhibiting M2 polarization. CONCLUSION These results suggested that TN-C accelerates LV remodelling after MI, at least in part, by modulating M1/M2-macrophage polarization.
Collapse
Affiliation(s)
- Taizo Kimura
- Department of Cardiology, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, Japan
| | - Kazuko Tajiri
- Department of Cardiology, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, Japan
| | - Akira Sato
- Department of Cardiology, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, Japan
| | - Satoshi Sakai
- Department of Cardiology, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, Japan
| | - Zheng Wang
- Department of Cardiology, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, Japan
| | - Toshimichi Yoshida
- Department of Pathology and Matrix Biology, Mie University Graduate School of Medicine, Tsu, Japan.,Mie University Research Center for Matrix Biology, Tsu, Japan
| | - Toshimitsu Uede
- Department of Matrix Medicine, Institute for Genetic Medicine, Hokkaido University, Sapporo, Japan
| | - Michiaki Hiroe
- Mie University Research Center for Matrix Biology, Tsu, Japan.,National Center of Global Health and Medicine, Tokyo, Japan
| | - Kazutaka Aonuma
- Department of Cardiology, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, Japan
| | - Masaki Ieda
- Department of Cardiology, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, Japan
| | - Kyoko Imanaka-Yoshida
- Department of Pathology and Matrix Biology, Mie University Graduate School of Medicine, Tsu, Japan.,Mie University Research Center for Matrix Biology, Tsu, Japan
| |
Collapse
|
229
|
Protease-activated receptor 2 deficiency in hematopoietic lineage protects against myocardial infarction through attenuated inflammatory response and fibrosis. Biochem Biophys Res Commun 2020; 526:253-260. [PMID: 32204916 DOI: 10.1016/j.bbrc.2020.03.077] [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: 02/21/2020] [Accepted: 03/12/2020] [Indexed: 11/20/2022]
Abstract
Ischaemic heart disease is one of the leading causes of death. Protease-activated receptor 2 (PAR2) is widely expressed within the cardiovascular system and is known to mediate inflammatory processes in various immunocytes, such as macrophages, mastocytes and neutrophils. Here, we investigated whether activating macrophage PAR2 modulates cardiac remodelling in a murine model of myocardial infarction. Myocardial infarction was produced by the permanent ligation of the left anterior descending coronary artery (LAD) in C57BL/6J background wild-type (WT) mice transplanted with bone marrow from WT or PAR2 knockout (PAR2 KO) mice. Hematopoietic deficiency of PAR2 had improvement of left ventricular systolic dysfunction and dilatation and decreased fibrosis deposition in remote zone at 1 week after LAD ligation. Inactivation of PAR2 also led to less recruitment of macrophages in myocardium, which was accompanied by decreased expression of pro-inflammatory cytokines. Furthermore, cultured cardiac fibroblasts (CFs) were activated and showed a fibrotic phenotype after being co-cultured in medium containing PAR2-activating macrophage, which enhances interferon-beta (INF-β) expression. The beneficial effects of macrophages with INF-β neutralisation or PAR2-deletion ameliorates the JAK/STAT3 pathway in CFs, which might be attributed to CF activation. These data suggest that macrophage-derived IFN-β plays a crucial role in adverse cardiac remodelling after myocardial infarction, at least in part, through a PAR2-dependent mechanism.
Collapse
|
230
|
Nasser MI, Zhu S, Huang H, Zhao M, Wang B, Ping H, Geng Q, Zhu P. Macrophages: First guards in the prevention of cardiovascular diseases. Life Sci 2020; 250:117559. [PMID: 32198051 DOI: 10.1016/j.lfs.2020.117559] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 03/12/2020] [Accepted: 03/13/2020] [Indexed: 12/19/2022]
Abstract
Cardiovascular diseases (CVD) remain one of the leading causes of mortality worldwide, especially in developing countries. It is widely known that severe inflammation can lead to atherosclerosis, which can cause various downstream pathologies, including myocardial injury and viral myocarditis. To date, several strategies have been proposed to prevent and cure CVD. The use of targeting macrophages has emerged as one of the most effective therapeutic approaches. Macrophages play a crucial role in eliminating senescent and dead cells while maintaining myocardial electrical activity and repairing myocardial injury. They also contribute to tissue repair and remodeling and plaque stabilization. Targeting macrophage pathways can, therefore, be advantageous in CVD care since it can lead to decreased aggregation of mononuclear cells at the injured site in the heart. Furthermore, it inhibits the development of pro-inflammatory factors, facilitates cholesterol outflow, and reduces the lipid concentration. More in-depth studies are still needed to formulate a comprehensive classification of phenotypes for different macrophages and determine their roles in the pathogenesis of CVD. In this review, we summarize the recent advances in the understanding of the role of macrophages in the prevention and cure of CVD.
Collapse
Affiliation(s)
- M I Nasser
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, 106 ZhongshanEr Road, Guangzhou, Guangdong 510100, China
| | - Shuoji Zhu
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, 106 ZhongshanEr Road, Guangzhou, Guangdong 510100, China
| | - Huanlei Huang
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, 106 ZhongshanEr Road, Guangzhou, Guangdong 510100, China
| | - Mingyi Zhao
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, 106 ZhongshanEr Road, Guangzhou, Guangdong 510100, China
| | - Bo Wang
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, 106 ZhongshanEr Road, Guangzhou, Guangdong 510100, China
| | - Huang Ping
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, 106 ZhongshanEr Road, Guangzhou, Guangdong 510100, China
| | - Qingshan Geng
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, 106 ZhongshanEr Road, Guangzhou, Guangdong 510100, China.
| | - Ping Zhu
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, 106 ZhongshanEr Road, Guangzhou, Guangdong 510100, China.
| |
Collapse
|
231
|
Mouton AJ, Li X, Hall ME, Hall JE. Obesity, Hypertension, and Cardiac Dysfunction: Novel Roles of Immunometabolism in Macrophage Activation and Inflammation. Circ Res 2020; 126:789-806. [PMID: 32163341 PMCID: PMC7255054 DOI: 10.1161/circresaha.119.312321] [Citation(s) in RCA: 352] [Impact Index Per Article: 70.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Obesity and hypertension, which often coexist, are major risk factors for heart failure and are characterized by chronic, low-grade inflammation, which promotes adverse cardiac remodeling. While macrophages play a key role in cardiac remodeling, dysregulation of macrophage polarization between the proinflammatory M1 and anti-inflammatory M2 phenotypes promotes excessive inflammation and cardiac injury. Metabolic shifting between glycolysis and mitochondrial oxidative phosphorylation has been implicated in macrophage polarization. M1 macrophages primarily rely on glycolysis, whereas M2 macrophages rely on the tricarboxylic acid cycle and oxidative phosphorylation; thus, factors that affect macrophage metabolism may disrupt M1/M2 homeostasis and exacerbate inflammation. The mechanisms by which obesity and hypertension may synergistically induce macrophage metabolic dysfunction, particularly during cardiac remodeling, are not fully understood. We propose that obesity and hypertension induce M1 macrophage polarization via mechanisms that directly target macrophage metabolism, including changes in circulating glucose and fatty acid substrates, lipotoxicity, and tissue hypoxia. We discuss canonical and novel proinflammatory roles of macrophages during obesity-hypertension-induced cardiac injury, including diastolic dysfunction and impaired calcium handling. Finally, we discuss the current status of potential therapies to target macrophage metabolism during heart failure, including antidiabetic therapies, anti-inflammatory therapies, and novel immunometabolic agents.
Collapse
Affiliation(s)
- Alan J. Mouton
- Department of Physiology and Biophysics, University of Mississippi Medical Center, 2500 North State Street; Jackson, MS, 39216-4505
- Mississippi Center for Obesity Research, University of Mississippi Medical Center, 2500 North State Street; Jackson, MS, 39216-4505
| | - Xuan Li
- Department of Physiology and Biophysics, University of Mississippi Medical Center, 2500 North State Street; Jackson, MS, 39216-4505
- Mississippi Center for Obesity Research, University of Mississippi Medical Center, 2500 North State Street; Jackson, MS, 39216-4505
| | - Michael E. Hall
- Department of Physiology and Biophysics, University of Mississippi Medical Center, 2500 North State Street; Jackson, MS, 39216-4505
- Department of Medicine, University of Mississippi Medical Center, 2500 North State Street; Jackson, MS, 39216-4505
- Mississippi Center for Obesity Research, University of Mississippi Medical Center, 2500 North State Street; Jackson, MS, 39216-4505
| | - John E. Hall
- Department of Physiology and Biophysics, University of Mississippi Medical Center, 2500 North State Street; Jackson, MS, 39216-4505
- Mississippi Center for Obesity Research, University of Mississippi Medical Center, 2500 North State Street; Jackson, MS, 39216-4505
| |
Collapse
|
232
|
Lindsey ML, Jung M, Yabluchanskiy A, Cannon PL, Iyer RP, Flynn ER, DeLeon-Pennell KY, Valerio FM, Harrison CL, Ripplinger CM, Hall ME, Ma Y. Exogenous CXCL4 infusion inhibits macrophage phagocytosis by limiting CD36 signalling to enhance post-myocardial infarction cardiac dilation and mortality. Cardiovasc Res 2020; 115:395-408. [PMID: 30169632 DOI: 10.1093/cvr/cvy211] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2018] [Accepted: 08/27/2018] [Indexed: 12/20/2022] Open
Abstract
Aims Macrophage phagocytosis of dead cells is a prerequisite for inflammation resolution. Because CXCL4 induces macrophage phagocytosis in vitro, we examined the impact of exogenous CXCL4 infusion on cardiac wound healing and macrophage phagocytosis following myocardial infarction (MI). Methods and results CXCL4 expression significantly increased in the infarct region beginning at Day 3 post-MI, and macrophages were the predominant source. Adult male C57BL/6J mice were subjected to coronary artery occlusion, and MI mice were randomly infused with recombinant mouse CXCL4 or saline beginning at 24 h post-MI by mini-pump infusion. Compared with saline controls, CXCL4 infusion dramatically reduced 7 day post-MI survival [10% (3/30) for CXCL4 vs. 47% (7/15) for saline, P < 0.05] as a result of acute congestive heart failure. By echocardiography, CXCL4 significantly increased left ventricular (LV) volumes and dimensions at Day 5 post-MI (all P < 0.05), despite similar infarct areas compared with saline controls. While macrophage numbers were similar at Day 5 post-MI, CXCL4 infusion increased Ccr4 and Itgb4 and decreased Adamts8 gene levels in the infarct region, all of which linked to CXCL4-mediated cardiac dilation. Isolated Day 5 post-MI macrophages exhibited comparable levels of M1 and M4 markers between saline and CXCL4 groups. Interestingly, by both ex vivo and in vitro phagocytosis assays, CXCL4 reduced macrophage phagocytic capacity, which was connected to decreased levels of the phagocytosis receptor CD36. In vitro, a CD36 neutralizing antibody (CD36Ab) significantly inhibited macrophage phagocytic capacity. The combination of CXCL4 and CD36Ab did not have an additive effect, indicating that CXCL4 regulated phagocytosis through CD36 signalling. CXCL4 infusion significantly elevated infarct matrix metalloproteinase (MMP)-9 levels at Day 5 post-MI, and MMP-9 can cleave CD36 as a down-regulation mechanism. Conclusion CXCL4 infusion impaired macrophage phagocytic capacity by reducing CD36 levels through MMP-9 dependent and independent signalling, leading to higher mortality and LV dilation.
Collapse
Affiliation(s)
- Merry L Lindsey
- Department of Physiology and Biophysics, Mississippi Center for Heart Research, University of Mississippi Medical Center, 2500 North State St., Jackson, MS, USA.,Research Service, G.V. (Sonny) Montgomery Veterans Affairs Medical Center, Jackson, MS, USA
| | - Mira Jung
- Department of Physiology and Biophysics, Mississippi Center for Heart Research, University of Mississippi Medical Center, 2500 North State St., Jackson, MS, USA
| | - Andriy Yabluchanskiy
- Department of Geriatric Medicine, Translational Geroscience Laboratory, Reynolds Oklahoma Center on Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Presley L Cannon
- Department of Physiology and Biophysics, Mississippi Center for Heart Research, University of Mississippi Medical Center, 2500 North State St., Jackson, MS, USA
| | - Rugmani Padmanabhan Iyer
- Department of Physiology and Biophysics, Mississippi Center for Heart Research, University of Mississippi Medical Center, 2500 North State St., Jackson, MS, USA
| | - Elizabeth R Flynn
- Department of Physiology and Biophysics, Mississippi Center for Heart Research, University of Mississippi Medical Center, 2500 North State St., Jackson, MS, USA
| | - Kristine Y DeLeon-Pennell
- Department of Physiology and Biophysics, Mississippi Center for Heart Research, University of Mississippi Medical Center, 2500 North State St., Jackson, MS, USA.,Research Service, G.V. (Sonny) Montgomery Veterans Affairs Medical Center, Jackson, MS, USA
| | - Fritz M Valerio
- Department of Physiology and Biophysics, Mississippi Center for Heart Research, University of Mississippi Medical Center, 2500 North State St., Jackson, MS, USA
| | - Courtney L Harrison
- Department of Physiology and Biophysics, Mississippi Center for Heart Research, University of Mississippi Medical Center, 2500 North State St., Jackson, MS, USA
| | - Crystal M Ripplinger
- Department of Pharmacology, School of Medicine, University of California, Davis, Davis, CA, USA
| | - Michael E Hall
- Department of Physiology and Biophysics, Mississippi Center for Heart Research, University of Mississippi Medical Center, 2500 North State St., Jackson, MS, USA.,Department of Medicine, Division of Cardiology, University of Mississippi Medical Center, Jackson, MS, USA
| | - Yonggang Ma
- Department of Physiology and Biophysics, Mississippi Center for Heart Research, University of Mississippi Medical Center, 2500 North State St., Jackson, MS, USA
| |
Collapse
|
233
|
Aimo A, Castiglione V, Borrelli C, Saccaro LF, Franzini M, Masi S, Emdin M, Giannoni A. Oxidative stress and inflammation in the evolution of heart failure: From pathophysiology to therapeutic strategies. Eur J Prev Cardiol 2020; 27:494-510. [DOI: 10.1177/2047487319870344] [Citation(s) in RCA: 181] [Impact Index Per Article: 36.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 08/30/2023]
Abstract
Both oxidative stress and inflammation are enhanced in chronic heart failure. Dysfunction of cardiac mitochondria is a hallmark of heart failure and a leading cause of oxidative stress, which in turn exerts detrimental effects on cellular components, including mitochondria themselves, thus generating a vicious circle. Oxidative stress also causes myocardial tissue damage and inflammation, contributing to heart failure progression. Furthermore, a subclinical inflammatory state may be caused by heart failure comorbidities such as obesity, diabetes mellitus or sleep apnoeas. Some markers of both oxidative stress and inflammation are enhanced in chronic heart failure and hold prognostic significance. For all these reasons, antioxidants or anti-inflammatory drugs may represent interesting additional therapies for subjects either at high risk or with established heart failure. Nonetheless, only a few clinical trials on antioxidants have been carried out so far, with several disappointing results except for vitamin C, elamipretide and coenzyme Q10. With regard to anti-inflammatory drugs, only preliminary data on the interleukin-1 antagonist anakinra are currently available. Therefore, a comprehensive, deep understanding of our current knowledge on oxidative stress and inflammation in chronic heart failure is key to providing some suggestions for future research on this topic.
Collapse
Affiliation(s)
- Alberto Aimo
- Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy
| | | | - Chiara Borrelli
- Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy
| | - Luigi F Saccaro
- Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy
| | | | | | - Michele Emdin
- Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy
- Fondazione Toscana Gabriele Monasterio, Pisa, Italy
| | - Alberto Giannoni
- Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy
- Fondazione Toscana Gabriele Monasterio, Pisa, Italy
| |
Collapse
|
234
|
Liu S, Chen J, Shi J, Zhou W, Wang L, Fang W, Zhong Y, Chen X, Chen Y, Sabri A, Liu S. M1-like macrophage-derived exosomes suppress angiogenesis and exacerbate cardiac dysfunction in a myocardial infarction microenvironment. Basic Res Cardiol 2020; 115:22. [PMID: 32112145 DOI: 10.1007/s00395-020-0781-7] [Citation(s) in RCA: 189] [Impact Index Per Article: 37.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Accepted: 02/13/2020] [Indexed: 12/21/2022]
Abstract
The roles and the underlying mechanisms of M1-type macrophages in angiogenesis and postmyocardial infarction (MI) cardiac repair have remained unclear. In this study, we investigated the role of M1-like macrophage-derived exosomes in a MI microenvironment. We found that the proinflammatory M1-like-type macrophages released an extensive array of proinflammatory exosomes (M1-Exos) after MI. M1-Exos exerted an anti-angiogenic effect and accelerated MI injury. They also exhibited highly expressed proinflammatory miRNAs, such as miR-155. miR-155 was transferred to endothelial cells (ECs), leading to the inhibition of angiogenesis and cardiac dysfunction by downregulating its novel target genes, including Rac family small GTPase 1 (RAC1), p21 (RAC1)-activated kinase 2 (PAK2), Sirtuin 1 (Sirt1), and protein kinase AMP-activated catalytic subunit alpha 2 (AMPKα2). M1-Exos depressed Sirt1/AMPKα2-endothelial nitric oxide synthase and RAC1-PAK2 signaling pathways by simultaneously targeting the five molecule nodes (genes), reduced the angiogenic ability of ECs, aggravated myocardial injury, and restrained cardiac healing. The elucidation of this mechanism provides novel insights into the functional significance of M1 macrophages and their derived exosomes on angiogenesis and cardiac repair. This mechanism can be used as a novel potential therapeutic approach for the prevention and treatment of MI.
Collapse
Affiliation(s)
- Shaojun Liu
- Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, State Key Laboratory of Respiratory Disease, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510260, Guangdong, People's Republic of China.
| | - Jing Chen
- Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, State Key Laboratory of Respiratory Disease, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510260, Guangdong, People's Republic of China
| | - Jian Shi
- Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, State Key Laboratory of Respiratory Disease, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510260, Guangdong, People's Republic of China
| | - Wenyi Zhou
- Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, State Key Laboratory of Respiratory Disease, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510260, Guangdong, People's Republic of China
| | - Li Wang
- Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, State Key Laboratory of Respiratory Disease, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510260, Guangdong, People's Republic of China.,Department of Cardiology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, 510260, Guangdong, People's Republic of China
| | - Weilun Fang
- Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, State Key Laboratory of Respiratory Disease, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510260, Guangdong, People's Republic of China
| | - Yun Zhong
- Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, State Key Laboratory of Respiratory Disease, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510260, Guangdong, People's Republic of China.,Department of Cardiology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, 510260, Guangdong, People's Republic of China
| | - Xiaohui Chen
- Department of Emergency, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, 510260, Guangdong, People's Republic of China
| | - Yanfang Chen
- Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, State Key Laboratory of Respiratory Disease, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510260, Guangdong, People's Republic of China.,Department of Pharmacology and Toxicology, Boonshoft School of Medicine, Wright State University, Dayton, OH, 45435, USA
| | - Abdelkarim Sabri
- Cardiovascular Research Center, Lewis Katz School of Medicine, Temple University, MERB 1045, 3500 N. Broad Street, Philadelphia, PA, 19140, USA
| | - Shiming Liu
- Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, State Key Laboratory of Respiratory Disease, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510260, Guangdong, People's Republic of China. .,Department of Cardiology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, 510260, Guangdong, People's Republic of China.
| |
Collapse
|
235
|
Ma S, Sun S, Geng L, Song M, Wang W, Ye Y, Ji Q, Zou Z, Wang S, He X, Li W, Esteban CR, Long X, Guo G, Chan P, Zhou Q, Belmonte JCI, Zhang W, Qu J, Liu GH. Caloric Restriction Reprograms the Single-Cell Transcriptional Landscape of Rattus Norvegicus Aging. Cell 2020; 180:984-1001.e22. [PMID: 32109414 DOI: 10.1016/j.cell.2020.02.008] [Citation(s) in RCA: 219] [Impact Index Per Article: 43.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Revised: 01/07/2020] [Accepted: 02/04/2020] [Indexed: 12/11/2022]
Abstract
Aging causes a functional decline in tissues throughout the body that may be delayed by caloric restriction (CR). However, the cellular profiles and signatures of aging, as well as those ameliorated by CR, remain unclear. Here, we built comprehensive single-cell and single-nucleus transcriptomic atlases across various rat tissues undergoing aging and CR. CR attenuated aging-related changes in cell type composition, gene expression, and core transcriptional regulatory networks. Immune cells were increased during aging, and CR favorably reversed the aging-disturbed immune ecosystem. Computational prediction revealed that the abnormal cell-cell communication patterns observed during aging, including the excessive proinflammatory ligand-receptor interplay, were reversed by CR. Our work provides multi-tissue single-cell transcriptional landscapes associated with aging and CR in a mammal, enhances our understanding of the robustness of CR as a geroprotective intervention, and uncovers how metabolic intervention can act upon the immune system to modify the process of aging.
Collapse
Affiliation(s)
- Shuai Ma
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Shuhui Sun
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Lingling Geng
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; Advanced Innovation Center for Human Brain Protection, and National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital Capital Medical University, Beijing 100053, China
| | - Moshi Song
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Wei Wang
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yanxia Ye
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China
| | - Qianzhao Ji
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zhiran Zou
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Si Wang
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; Advanced Innovation Center for Human Brain Protection, and National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital Capital Medical University, Beijing 100053, China; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China
| | - Xiaojuan He
- Advanced Innovation Center for Human Brain Protection, and National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital Capital Medical University, Beijing 100053, China
| | - Wei Li
- Advanced Innovation Center for Human Brain Protection, and National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital Capital Medical University, Beijing 100053, China
| | | | - Xiao Long
- Division of Plastic Surgery, Peking Union Medical College Hospital, Beijing 100032, China
| | - Guoji Guo
- Center for Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Piu Chan
- Advanced Innovation Center for Human Brain Protection, and National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital Capital Medical University, Beijing 100053, China
| | - Qi Zhou
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | | | - Weiqi Zhang
- Advanced Innovation Center for Human Brain Protection, and National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital Capital Medical University, Beijing 100053, China; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China; Disease Genomics and Individualized Medicine Laboratory, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China; China National Center for Bioinformation, Beijing 100101, China.
| | - Jing Qu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| | - Guang-Hui Liu
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China; Advanced Innovation Center for Human Brain Protection, and National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital Capital Medical University, Beijing 100053, China; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
236
|
Understanding the mechanisms that determine extracellular matrix remodeling in the infarcted myocardium. Biochem Soc Trans 2020; 47:1679-1687. [PMID: 31724697 DOI: 10.1042/bst20190113] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2019] [Revised: 10/22/2019] [Accepted: 10/24/2019] [Indexed: 02/06/2023]
Abstract
Myocardial Infarction (MI) initiates a series of wound healing events that begins with up-regulation of an inflammatory response and culminates in scar formation. The extracellular matrix (ECM) is intricately involved in all stages from initial break down of existing ECM to synthesis of new ECM to form the scar. This review will summarize our current knowledge on the processes involved in ECM remodeling after MI and identify the gaps that still need to be filled.
Collapse
|
237
|
Lu H, Chen R, Barnie PA, Tian Y, Zhang S, Xu H, Chakrabarti S, Su Z. Fibroblast transdifferentiation promotes conversion of M1 macrophages and replenishment of cardiac resident macrophages following cardiac injury in mice. Eur J Immunol 2020; 50:795-808. [PMID: 32068249 DOI: 10.1002/eji.201948414] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2019] [Revised: 02/13/2020] [Accepted: 02/17/2020] [Indexed: 12/24/2022]
Abstract
Resident cardiac macrophages play important roles in homeostasis, maintenance of cardiac function, and tissue repair. After cardiac injury, monocytes infiltrate the tissue, undergo phenotypic and functional changes, and are involved in inflammatory injury and functional remodelling. However, the fate of cardiac infiltrating/polarized macrophages and the relationship between these cells and resident cardiac macrophage replenishment following injury remain unclear. Our results showed that angiotensin II induces cardiac fibroblast transdifferentiation into cardiac myofibroblasts (MFBs). In cocultures with MFBs and murine macrophages, the MFBs promoted macrophage polarization to M1 phenotype, followed by selective apoptosis, which was associated with TNF/TNFR1 axis and independent of NO production. Surprisingly, after 36 h of coculture, the surviving macrophages were converted to M2 phenotype and settled in heart, which was dependent on leptin produced by MFBs or polarized macrophages via the PI3K or Akt pathway. CCR2+ CD45.2+ cells adoptively transferred into CD45.1+ mice with viral myocarditis, differentiated into CD45.2+ CCR2+ CX3CR1+ M2 cells during the resolution of inflammation and settled within the heart. Our data highlight a novel mechanism related to the renewal or replenishment of cardiac resident macrophages following cardiac injury; and suggest that transdifferentiation of cardiac fibroblasts may promote the resolution of inflammation.
Collapse
Affiliation(s)
- Hongxiang Lu
- International Genome Center, Jiangsu University, Zhenjiang, China.,Department of Immunology, Jiangsu University, Zhenjiang, China
| | - Rong Chen
- International Genome Center, Jiangsu University, Zhenjiang, China.,Department of Immunology, Jiangsu University, Zhenjiang, China
| | | | - Yu Tian
- International Genome Center, Jiangsu University, Zhenjiang, China.,Department of Immunology, Jiangsu University, Zhenjiang, China
| | - Shiqing Zhang
- International Genome Center, Jiangsu University, Zhenjiang, China.,Department of Immunology, Jiangsu University, Zhenjiang, China
| | - Huaxi Xu
- Department of Immunology, Jiangsu University, Zhenjiang, China
| | - Subrata Chakrabarti
- Pathology and Laboratory Medicine, Western University, London, Ontario, Canada
| | - Zhaoliang Su
- International Genome Center, Jiangsu University, Zhenjiang, China.,Department of Immunology, Jiangsu University, Zhenjiang, China.,Laboratory Center, The Fourth Affiliated Hospital of Jiangsu University, Zhenjiang, China
| |
Collapse
|
238
|
Connexin43 expression in bone marrow derived cells contributes to the electrophysiological properties of cardiac scar tissue. Sci Rep 2020; 10:2617. [PMID: 32054938 PMCID: PMC7018966 DOI: 10.1038/s41598-020-59449-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2019] [Accepted: 01/29/2020] [Indexed: 11/26/2022] Open
Abstract
Cardiac pathologies associated with arrhythmic activity are often accompanied by inflammation. The contribution of inflammatory cells to the electrophysiological properties of injured myocardium is unknown. Myocardial scar cell types and intercellular contacts were analyzed using a three-dimensional reconstruction from serial blockface scanning electron microscopy data. Three distinct cell populations were identified: inflammatory, fibroblastic and endocardial cells. While individual fibroblastic cells interface with a greater number of cells, inflammatory cells have the largest contact area suggesting a role in establishing intercellular electrical connections in scar tissue. Optical mapping was used to study the electrophysiological properties of scars in fetal liver chimeric mice generated using connexin43 knockout donors (bmpKO). Voltage changes were elicited in response to applied current pulses. Isopotential maps showed a steeper pattern of decay with distance from the electrode in scars compared with uninjured regions, suggesting reduced electrical coupling. The tissue decay constant, defined as the distance voltage reaches 37% of the amplitude at the edge of the scar, was 0.48 ± 0.04 mm (n = 11) in the scar of the bmpCTL group and decreased 37.5% in the bmpKO group (n = 10). Together these data demonstrate inflammatory cells significantly contribute to scar electrophysiology through coupling mediated at least partially by connexin43 expression.
Collapse
|
239
|
TRPV2 channel as a possible drug target for the treatment of heart failure. J Transl Med 2020; 100:207-217. [PMID: 31857697 DOI: 10.1038/s41374-019-0349-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2019] [Revised: 11/09/2019] [Accepted: 11/13/2019] [Indexed: 02/07/2023] Open
Abstract
Heart transplantation is currently the only viable option available for the treatment of severe heart failure conditions such as dilated cardiomyopathy. Hence, novel drugs for treating such conditions need to be developed urgently. Recent studies suggest that Ca2+ overload is involved in the onset and progression of dilated cardiomyopathy, and thus heart failure. The expression and activation of the Ca2+ permeable channel, transient receptor potential vanilloid 2 (TRPV2) channel have been found to play an essential role in sustained intracellular Ca2+ concentration increase, leading to heart failure. However, since there have been no TRPV2-specific inhibitors available until recently, the effect of TRPV2 inhibition on the pathology has not been clearly elucidated. Recent reports show that inhibiting TRPV2 activity effectively improves cardiac function, suppressing myocardial fibrosis and ameliorating the prognosis in animal models of cardiomyopathy with heart failure. In addition to that, inflammation is reported to be involved in the development of heart failure. Here, we review the recent findings on TRPV2 in cardiomyocytes and immune cells involved in the development of heart failure and discuss the current progress of drug development for the treatment of heart failure via targeting TRPV2.
Collapse
|
240
|
Wang L, Yu P, Zhou B, Song J, Li Z, Zhang M, Guo G, Wang Y, Chen X, Han L, Hu S. Single-cell reconstruction of the adult human heart during heart failure and recovery reveals the cellular landscape underlying cardiac function. Nat Cell Biol 2020; 22:108-119. [PMID: 31915373 DOI: 10.1038/s41556-019-0446-7] [Citation(s) in RCA: 263] [Impact Index Per Article: 52.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Accepted: 11/28/2019] [Indexed: 01/04/2023]
Abstract
Owing to the prevalence and high mortality rates of cardiac diseases, a more detailed characterization of the human heart is necessary; however, this has been largely impeded by the cellular diversity of cardiac tissue and limited access to samples. Here, we show transcriptome profiling of 21,422 single cells-including cardiomyocytes (CMs) and non-CMs (NCMs)-from normal, failed and partially recovered (left ventricular assist device treatment) adult human hearts. Comparative analysis of atrial and ventricular cells revealed pronounced inter- and intracompartmental CM heterogeneity as well as compartment-specific utilization of NCM cell types as major cell-communication hubs. Systematic analysis of cellular compositions and cell-cell interaction networks showed that CM contractility and metabolism are the most prominent aspects that are correlated with changes in heart function. We also uncovered active engagement of NCMs in regulating the behaviour of CMs, exemplified by ACKR1+-endothelial cells, injection of which preserved cardiac function after injury. Beyond serving as a rich resource, our study provides insights into cell-type-targeted intervention of heart diseases.
Collapse
Affiliation(s)
- Li Wang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Peng Yu
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Bingying Zhou
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jiangping Song
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zheng Li
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Mingzhi Zhang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Guangran Guo
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yin Wang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiao Chen
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Leng Han
- Department of Biochemistry and Molecular Biology, The University of Texas Health Science Center at Houston McGovern Medical School, Houston, TX, USA
| | - Shengshou Hu
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| |
Collapse
|
241
|
Hilt ZT, Ture SK, Mohan A, Arne A, Morrell CN. Platelet-derived β2m regulates age related monocyte/macrophage functions. Aging (Albany NY) 2019; 11:11955-11974. [PMID: 31852838 PMCID: PMC6949047 DOI: 10.18632/aging.102520] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Accepted: 11/18/2019] [Indexed: 02/07/2023]
Abstract
Platelets have central roles in both immune responses and development. Stimulated platelets express leukocyte adhesion molecules and release numerous immune modulatory factors that recruit and activate leukocytes, both at the sites of activation and distantly. Monocytes are innate immune cells with dynamic immune modulatory functions that change during the aging process, a phenomenon termed “inflammaging”. We have previously shown that platelets are a major source of plasma beta-2 microglobulin (β2M) and that β2M induced a monocyte pro-inflammatory phenotype. Plasma β2M increases with age and is a pro-aging factor. We hypothesized that platelet derived β2M regulates monocyte phenotypes in the context of aging. Using wild-type (WT) and platelet specific β2M knockout mice (Plt-β2M-/-) mice, we found that plasma β2M increased with age and correlated with increased circulating Ly6CHi monocytes. However, aged Plt-β2M-/- mice had significantly fewer Ly6CHi monocytes compared to WT mice. Quantitative real-time PCR of circulating monocytes showed that WT mouse monocytes were more “pro-inflammatory” with age, while Plt-β2M-/- derived monocytes adopted a “pro-reparative” phenotype. Older Plt-β2M-/- mice had a significant decline in heart function compared to age matched WT mice, as well as increased cardiac fibrosis and pro-fibrotic markers. These data suggest that platelet-derived β2M regulates age associated monocyte polarization, and a loss of platelet derived β2M shifted monocytes and macrophages to a pro-reparative phenotype and increased pro-fibrotic cardiac responses. Platelet regulation of monocyte phenotypes via β2M may maintain a balance between inflammatory and reparative signals that affects age related physiologic outcomes.
Collapse
Affiliation(s)
- Zachary T Hilt
- Aab Cardiovascular Research Institute, University of Rochester School of Medicine, Box CVRI, Rochester, NY 14652, USA
| | - Sara K Ture
- Aab Cardiovascular Research Institute, University of Rochester School of Medicine, Box CVRI, Rochester, NY 14652, USA
| | - Amy Mohan
- Aab Cardiovascular Research Institute, University of Rochester School of Medicine, Box CVRI, Rochester, NY 14652, USA
| | - Allison Arne
- Aab Cardiovascular Research Institute, University of Rochester School of Medicine, Box CVRI, Rochester, NY 14652, USA
| | - Craig N Morrell
- Aab Cardiovascular Research Institute, University of Rochester School of Medicine, Box CVRI, Rochester, NY 14652, USA.,Department of Microbiology and Immunology, University of Rochester School of Medicine, Rochester, NY 14652, USA
| |
Collapse
|
242
|
Bajpai G, Bredemeyer A, Li W, Zaitsev K, Koenig AL, Lokshina I, Mohan J, Ivey B, Hsiao HM, Weinheimer C, Kovacs A, Epelman S, Artyomov M, Kreisel D, Lavine KJ. Tissue Resident CCR2- and CCR2+ Cardiac Macrophages Differentially Orchestrate Monocyte Recruitment and Fate Specification Following Myocardial Injury. Circ Res 2019; 124:263-278. [PMID: 30582448 DOI: 10.1161/circresaha.118.314028] [Citation(s) in RCA: 495] [Impact Index Per Article: 82.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
RATIONALE Recent advancements have brought to light the origins, complexity, and functions of tissue-resident macrophages. However, in the context of tissue injury or disease, large numbers of monocytes infiltrate the heart and are thought to contribute to adverse remodeling and heart failure pathogenesis. Little is understood about the diversity of monocytes and monocyte-derived macrophages recruited to the heart after myocardial injury, including the mechanisms that regulate monocyte recruitment and fate specification. OBJECTIVE We sought to test the hypothesis that distinct subsets of tissue-resident CCR2- (C-C chemokine receptor 2) and CCR2+ macrophages orchestrate monocyte recruitment and fate specification after myocardial injury. METHODS AND RESULTS We reveal that in numerous mouse models of cardiomyocyte cell death (permanent myocardial infarction, reperfused myocardial infarction, and diphtheria toxin cardiomyocyte ablation), there is a shift in macrophage ontogeny whereby tissue-resident macrophages are predominately replaced by infiltrating monocytes and monocyte-derived macrophages. Using syngeneic cardiac transplantation to model ischemia-reperfusion injury and distinguish tissue-resident from recruited cell populations in combination with intravital 2-photon microscopy, we demonstrate that monocyte recruitment is differentially orchestrated by distinct subsets of tissue-resident cardiac macrophages. Tissue-resident CCR2+ macrophages promote monocyte recruitment through an MYD88 (myeloid differentiation primary response 88)-dependent mechanism that results in release of MCPs (monocyte chemoattractant proteins) and monocyte mobilization. In contrast, tissue-resident CCR2- macrophages inhibit monocyte recruitment. Using CD (cluster of differentiation) 169-DTR (diphtheria toxin receptor) and CCR2-DTR mice, we further show that selective depletion of either tissue-resident CCR2- or CCR2+ macrophages before myocardial infarction results in divergent effects on left ventricular function, myocardial remodeling, and monocyte recruitment. Finally, using single-cell RNA sequencing, we show that tissue-resident cardiac macrophages differentially instruct monocyte fate specification. CONCLUSIONS Collectively, these observations establish the mechanistic basis by which monocytes are initially recruited to the injured heart and provide new insights into the heterogeneity of monocyte-derived macrophages.
Collapse
Affiliation(s)
- Geetika Bajpai
- From the Department of Medicine (G.B., A.B., A.L. Koenig, I.L., J.M., B.I., C.W., A. Kovacs, K.J.L.), Washington University School of Medicine, St. Louis, MO
| | - Andrea Bredemeyer
- From the Department of Medicine (G.B., A.B., A.L. Koenig, I.L., J.M., B.I., C.W., A. Kovacs, K.J.L.), Washington University School of Medicine, St. Louis, MO
| | - Wenjun Li
- Department of Surgery (W.L., H.-M.H., D.K., K.J.L.), Washington University School of Medicine, St. Louis, MO
| | - Konstantin Zaitsev
- Department of Immunology and Pathology (K.Z., M.A., D.K., K.J.L.), Washington University School of Medicine, St. Louis, MO
| | - Andrew L Koenig
- From the Department of Medicine (G.B., A.B., A.L. Koenig, I.L., J.M., B.I., C.W., A. Kovacs, K.J.L.), Washington University School of Medicine, St. Louis, MO
| | - Inessa Lokshina
- From the Department of Medicine (G.B., A.B., A.L. Koenig, I.L., J.M., B.I., C.W., A. Kovacs, K.J.L.), Washington University School of Medicine, St. Louis, MO
| | - Jayaram Mohan
- From the Department of Medicine (G.B., A.B., A.L. Koenig, I.L., J.M., B.I., C.W., A. Kovacs, K.J.L.), Washington University School of Medicine, St. Louis, MO
| | - Brooke Ivey
- From the Department of Medicine (G.B., A.B., A.L. Koenig, I.L., J.M., B.I., C.W., A. Kovacs, K.J.L.), Washington University School of Medicine, St. Louis, MO
| | - His-Min Hsiao
- Department of Surgery (W.L., H.-M.H., D.K., K.J.L.), Washington University School of Medicine, St. Louis, MO
| | - Carla Weinheimer
- From the Department of Medicine (G.B., A.B., A.L. Koenig, I.L., J.M., B.I., C.W., A. Kovacs, K.J.L.), Washington University School of Medicine, St. Louis, MO
| | - Attila Kovacs
- From the Department of Medicine (G.B., A.B., A.L. Koenig, I.L., J.M., B.I., C.W., A. Kovacs, K.J.L.), Washington University School of Medicine, St. Louis, MO
| | - Slava Epelman
- Toronto General Hospital Research Institute, Division of Cardiology, University Health Network, ON, Canada (S.E.)
| | - Maxim Artyomov
- Department of Immunology and Pathology (K.Z., M.A., D.K., K.J.L.), Washington University School of Medicine, St. Louis, MO
| | - Daniel Kreisel
- Department of Surgery (W.L., H.-M.H., D.K., K.J.L.), Washington University School of Medicine, St. Louis, MO.,Department of Immunology and Pathology (K.Z., M.A., D.K., K.J.L.), Washington University School of Medicine, St. Louis, MO
| | - Kory J Lavine
- From the Department of Medicine (G.B., A.B., A.L. Koenig, I.L., J.M., B.I., C.W., A. Kovacs, K.J.L.), Washington University School of Medicine, St. Louis, MO.,Department of Surgery (W.L., H.-M.H., D.K., K.J.L.), Washington University School of Medicine, St. Louis, MO.,Department of Immunology and Pathology (K.Z., M.A., D.K., K.J.L.), Washington University School of Medicine, St. Louis, MO.,Department of Developmental Biology (K.J.L.), Washington University School of Medicine, St. Louis, MO
| |
Collapse
|
243
|
Suetomi T, Miyamoto S, Brown JH. Inflammation in nonischemic heart disease: initiation by cardiomyocyte CaMKII and NLRP3 inflammasome signaling. Am J Physiol Heart Circ Physiol 2019; 317:H877-H890. [PMID: 31441689 PMCID: PMC6879920 DOI: 10.1152/ajpheart.00223.2019] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Revised: 08/09/2019] [Accepted: 08/21/2019] [Indexed: 12/14/2022]
Abstract
There is substantial evidence that chronic heart failure in humans and in animal models is associated with inflammation. Ischemic interventions such as myocardial infarction lead to necrotic cell death and release of damage associated molecular patterns, factors that signal cell damage and induce expression of proinflammatory chemokines and cytokines. It has recently become evident that nonischemic interventions are also associated with increases in inflammatory genes and immune cell accumulation in the heart and that these contribute to fibrosis and ventricular dysfunction. How proinflammatory responses are elicited in nonischemic heart disease which is not, at least initially, associated with cell death is a critical unanswered question. In this review we provide evidence supporting the hypothesis that cardiomyocytes are an initiating site of inflammatory gene expression in response to nonischemic stress. Furthermore we discuss the role of the multifunctional Ca2+/calmodulin-regulated kinase, CaMKIIδ, as a transducer of stress signals to nuclear factor-κB activation, expression of proinflammatory cytokines and chemokines, and priming and activation of the NOD-like pyrin domain-containing protein 3 (NLRP3) inflammasome in cardiomyocytes. We summarize recent evidence that subsequent macrophage recruitment, fibrosis and contractile dysfunction induced by angiotensin II infusion or transverse aortic constriction are ameliorated by blockade of CaMKII, of monocyte chemoattractant protein-1/C-C chemokine receptor type 2 signaling, or of NLRP3 inflammasome activation.
Collapse
Affiliation(s)
- Takeshi Suetomi
- Division of Cardiology, Department of Medicine and Clinical Science, Yamaguchi University Graduate School of Medicine, Ube, Yamaguchi, Japan
- Department of Pharmacology, University of California San Diego, La Jolla, California
| | - Shigeki Miyamoto
- Department of Pharmacology, University of California San Diego, La Jolla, California
| | - Joan Heller Brown
- Department of Pharmacology, University of California San Diego, La Jolla, California
| |
Collapse
|
244
|
Dissociation between hypertrophy and fibrosis in the left ventricle early after experimental kidney transplantation. J Hypertens 2019; 38:489-503. [PMID: 31652182 DOI: 10.1097/hjh.0000000000002285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVE Left ventricular (LV) hypertrophy is the most common cardiac alteration in patients with chronic kidney disease (CKD). Normalization of hypertension in CKD patients receiving a healthy kidney allograft often reverses LV hypertrophy, but effects on LV fibrosis remain unclear. To study causal interactions between graft and environment on LV hypertrophy, fibrosis and inflammation, we applied cross-kidney transplantation METHODS:: Orthotopic transplantation was performed after inducing CKD in rats by two-third bilateral ablation of kidney mass: Healthy kidney (K) donor to healthy heart (H) recipient (healthy-K→healthy-H); CKD-K→healthy-H; healthy-K→CKD-H; CKD-K→CKD-H; N= 6 per group. RESULTS At week 6 after transplantation, mean arterial pressure (MAP) and LV mass index (LVMI) increased in CKD-K versus healthy-K irrespective of recipient. Contrarily, LV fibrosis was more severe in CKD-H versus healthy-H recipients irrespective of graft. Indeed, MAP and plasma creatinine correlated with LVMI but not with LV fibrosis. Increased LVMI in CKD-K→CKD-H not accompanied by cardiomyocyte cross-sectional area gain is consistent with eccentric remodelling. Cardiac RNA sequencing found a strong transcriptional response associated with LV fibrosis but only sparse changes associated with LV hypertrophy. This response was, among others, characterized by changes in extracellular matrix (ECM) and inflammatory gene expression. CONCLUSION LVMI reversed and MAP and renal function were normalized early after transplantation of a healthy kidney. However, LV fibrosis persisted, dissociating LV hypertrophy from LV fibrosis within 6 weeks. Elucidating cardiac ECM dynamics in CKD patients, although challenging, appears promising.
Collapse
|
245
|
Harhous Z, Booz GW, Ovize M, Bidaux G, Kurdi M. An Update on the Multifaceted Roles of STAT3 in the Heart. Front Cardiovasc Med 2019; 6:150. [PMID: 31709266 PMCID: PMC6823716 DOI: 10.3389/fcvm.2019.00150] [Citation(s) in RCA: 94] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2019] [Accepted: 10/07/2019] [Indexed: 12/18/2022] Open
Abstract
Signal transducer and activator of transcription 3 (STAT3) is a signaling molecule and transcription factor that plays important protective roles in the heart. The protection mediated by STAT3 is attributed to its genomic actions as a transcription factor and other non-genomic roles targeting mitochondrial function and autophagy. As a transcription factor, STAT3 upregulates genes that are anti-oxidative, anti-apoptotic, and pro-angiogenic, but suppresses anti-inflammatory and anti-fibrotic genes. Its suppressive effects on gene expression are achieved through competing with other transcription factors or cofactors. STAT3 is also linked to the modification of mRNA expression profiles in cardiac cells by inhibiting or inducing miRNA. In addition to these genomic roles, STAT3 is suggested to function protectively in mitochondria, where it regulates ROS production, in part by regulating the activities of the electron transport chain complexes, although our recent evidence calls this role into question. Nonetheless, STAT3 is a key player known to be activated in the cardioprotective ischemic conditioning protocols. Through these varied roles, STAT3 participates in various mechanisms that contribute to cardioprotection against different heart pathologies, including myocardial infarction, hypertrophy, diabetic cardiomyopathy, and peripartum cardiomyopathy. Understanding how STAT3 is involved in the protective mechanisms against these different cardiac pathologies could lead to novel therapeutic strategies to treat them.
Collapse
Affiliation(s)
- Zeina Harhous
- Laboratory of Experimental and Clinical Pharmacology, Faculty of Sciences, Doctoral School of Sciences and Technology, Lebanese University, Beirut, Lebanon
- Univ-Lyon, CarMeN Laboratory, INSERM 1060, INRA 1397, University Claude Bernard Lyon1, INSA Lyon, Oullins, France
- IHU OPeRa, Groupement Hospitalier EST, Bron, France
| | - George W. Booz
- Department of Pharmacology and Toxicology, School of Medicine, The University of Mississippi Medical Center, Jackson, MS, United States
| | - Michel Ovize
- Univ-Lyon, CarMeN Laboratory, INSERM 1060, INRA 1397, University Claude Bernard Lyon1, INSA Lyon, Oullins, France
- IHU OPeRa, Groupement Hospitalier EST, Bron, France
| | - Gabriel Bidaux
- Univ-Lyon, CarMeN Laboratory, INSERM 1060, INRA 1397, University Claude Bernard Lyon1, INSA Lyon, Oullins, France
- IHU OPeRa, Groupement Hospitalier EST, Bron, France
| | - Mazen Kurdi
- Laboratory of Experimental and Clinical Pharmacology, Faculty of Sciences, Doctoral School of Sciences and Technology, Lebanese University, Beirut, Lebanon
| |
Collapse
|
246
|
Carotenuto F, Teodori L, Maccari AM, Delbono L, Orlando G, Di Nardo P. Turning regenerative technologies into treatment to repair myocardial injuries. J Cell Mol Med 2019; 24:2704-2716. [PMID: 31568640 PMCID: PMC7077550 DOI: 10.1111/jcmm.14630] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Revised: 06/28/2019] [Accepted: 07/23/2019] [Indexed: 02/06/2023] Open
Abstract
Regenerative therapies including stem cell treatments hold promise to allow curing patients affected by severe cardiac muscle diseases. However, the clinical efficacy of stem cell therapy remains elusive, so far. The two key roadblocks that still need to be overcome are the poor cell engraftment into the injured myocardium and the limited knowledge of the ideal mixture of bioactive factors to be locally delivered for restoring heart function. Thus, therapeutic strategies for cardiac repair are directed to increase the retention and functional integration of transplanted cells in the damaged myocardium or to enhance the endogenous repair mechanisms through cell‐free therapies. In this context, biomaterial‐based technologies and tissue engineering approaches have the potential to dramatically impact cardiac translational medicine. This review intends to offer some consideration on the cell‐based and cell‐free cardiac therapies, their limitations and the possible future developments.
Collapse
Affiliation(s)
- Felicia Carotenuto
- Centro Interdipartimentale di Medicina Rigenerativa, Università di Roma Tor Vergata, Rome, Italy.,Dipartimento di Scienze Cliniche e Medicina Traslazionale, Università di Roma Tor Vergata, Rome, Italy.,Diagnostics and Metrology (FSN-TECFIS-DIM), ENEA, C.R. Frascati, Rome, Italy
| | - Laura Teodori
- Diagnostics and Metrology (FSN-TECFIS-DIM), ENEA, C.R. Frascati, Rome, Italy
| | - Anna Maria Maccari
- Centro Interdipartimentale di Medicina Rigenerativa, Università di Roma Tor Vergata, Rome, Italy.,Dipartimento di Scienze Cliniche e Medicina Traslazionale, Università di Roma Tor Vergata, Rome, Italy
| | - Luciano Delbono
- Wake Forest University School of Medicine, Winston Salem, NC, USA
| | - Giuseppe Orlando
- Wake Forest University School of Medicine, Winston Salem, NC, USA.,Department of Surgery, Wake Forest University School of Medicine, Winston Salem, NC, USA
| | - Paolo Di Nardo
- Centro Interdipartimentale di Medicina Rigenerativa, Università di Roma Tor Vergata, Rome, Italy.,Dipartimento di Scienze Cliniche e Medicina Traslazionale, Università di Roma Tor Vergata, Rome, Italy.,I.M. Sechenov First Moscow State Medical University, Moscow, Russia
| |
Collapse
|
247
|
Schroer AK, Bersi MR, Clark CR, Zhang Q, Sanders LH, Hatzopoulos AK, Force TL, Majka SM, Lal H, Merryman WD. Cadherin-11 blockade reduces inflammation-driven fibrotic remodeling and improves outcomes after myocardial infarction. JCI Insight 2019; 4:131545. [PMID: 31534054 PMCID: PMC6795284 DOI: 10.1172/jci.insight.131545] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Accepted: 08/21/2019] [Indexed: 12/17/2022] Open
Abstract
Over one million Americans experience myocardial infarction (MI) annually, and the resulting scar and subsequent cardiac fibrosis gives rise to heart failure. A specialized cell-cell adhesion protein, cadherin-11 (CDH11), contributes to inflammation and fibrosis in rheumatoid arthritis, pulmonary fibrosis, and aortic valve calcification but has not been studied in myocardium after MI. MI was induced by ligation of the left anterior descending artery in mice with either heterozygous or homozygous knockout of CDH11, wild-type mice receiving bone marrow transplants from Cdh11-deficient animals, and wild-type mice treated with a functional blocking antibody against CDH11 (SYN0012). Flow cytometry revealed significant CDH11 expression in noncardiomyocyte cells after MI. Animals given SYN0012 had improved cardiac function, as measured by echocardiogram, reduced tissue remodeling, and altered transcription of inflammatory and proangiogenic genes. Targeting CDH11 reduced bone marrow-derived myeloid cells and increased proangiogenic cells in the heart 3 days after MI. Cardiac fibroblast and macrophage interactions increased IL-6 secretion in vitro. Our findings suggest that CDH11-expressing cells contribute to inflammation-driven fibrotic remodeling after MI and that targeting CDH11 with a blocking antibody improves outcomes by altering recruitment of bone marrow-derived cells, limiting the macrophage-induced expression of IL-6 by fibroblasts and promoting vascularization.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Susan M. Majka
- Department of Allergy, Pulmonary, and Critical Care Medicine, Vanderbilt University, Nashville, Tennessee, USA
| | - Hind Lal
- Department of Cardiovascular Medicine, and
| | | |
Collapse
|
248
|
Oishi Y, Manabe I. Macrophages in inflammation, repair and regeneration. Int Immunol 2019; 30:511-528. [PMID: 30165385 DOI: 10.1093/intimm/dxy054] [Citation(s) in RCA: 389] [Impact Index Per Article: 64.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Accepted: 08/22/2018] [Indexed: 12/12/2022] Open
Abstract
Tissue injury triggers a complex series of cellular responses, starting from inflammation activated by tissue and cell damage and proceeding to healing. By clearing cell debris, activating and resolving inflammation and promoting fibrosis, macrophages play key roles in most, if not all, phases of the response to injury. Recent studies of the mechanisms underlying the initial inflammation and later tissue regeneration and repair revealed that macrophages bridge these processes in part by supporting and activating stem/progenitor cells, clearing damaged tissue, remodeling extracellular matrix to prepare scaffolding for regeneration and promoting angiogenesis. However, macrophages also have a central role in the development of pathology induced by failed resolution (e.g. chronic inflammation) and excessive scarring. In this review, we summarize the activities of macrophages in inflammation and healing in response to acute injury in tissues with differing regenerative capacities. While macrophages lead similar processes in response to tissue injury in these tissues, their priorities and the consequences of their activities differ among tissues. Moreover, the magnitude, nature and duration of injury also greatly affect cellular responses and healing processes. In particular, continuous injury and/or failed resolution of inflammation leads to chronic ailments in which macrophage activities may become detrimental.
Collapse
Affiliation(s)
- Yumiko Oishi
- Department of Biochemistry & Molecular Biology, Nippon Medical School, Bunkyo-ku, Tokyo, Japan
| | - Ichiro Manabe
- Department of Disease Biology and Molecular Medicine, Chiba University Graduate School of Medicine, Chuo-ku, Chiba, Japan
| |
Collapse
|
249
|
Temporal dynamics of immune response following prolonged myocardial ischemia/reperfusion with and without cyclosporine A. Acta Pharmacol Sin 2019; 40:1168-1183. [PMID: 30858476 PMCID: PMC6786364 DOI: 10.1038/s41401-018-0197-1] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Accepted: 11/21/2018] [Indexed: 12/19/2022]
Abstract
Understanding the dynamics of the immune response following late myocardial reperfusion is critical for the development of immunomodulatory therapy for myocardial infarction (MI). Cyclosporine A (CSA) possesses multiple therapeutic applications for MI, but its effects on the inflammation caused by acute MI are not clear. This study aimed to determine the dynamics of the immune response following myocardial ischemia/reperfusion (I/R) and the effects of CSA in a mouse model of prolonged myocardial ischemia designated to represent the human condition of late reperfusion. Adult C57BL/6 mice were subjected to 90 min of closed-chest myocardial I/R, which induced severe myocardial injury and excessive inflammation in the heart. Multicomponent analysis of the immune response caused by prolonged I/R revealed that the peak of cytokines/chemokines in the systemic circulation was synchronized with the maximal influx of neutrophils and T-cells in the heart 1 day after MI. The peak of cytokine/chemokine secretion in the infarcted heart coincided with the maximal macrophage and natural killer cell infiltration on day 3 after MI. The cellular composition of the mediastinal lymph nodes changed similarly to that of the infarcted hearts. CSA (10 mg/kg/day) given after prolonged I/R impaired heart function, enlarged the resulting scar, and reduced heart vascularization. It did not change the content of immune cells in hearts exposed to prolonged I/R, but the levels of MCP-1 and MIP-1α (hearts) and IL-12 (hearts and serum) were significantly reduced in the CSA-treated group in comparison to the untreated group, indicating alterations in immune cell function. Our findings provide new knowledge necessary for the development of immunomodulatory therapy targeting the immune response after prolonged myocardial ischemia/reperfusion.
Collapse
|
250
|
Wang X, Iyer A, Lyons AB, Körner H, Wei W. Emerging Roles for G-protein Coupled Receptors in Development and Activation of Macrophages. Front Immunol 2019; 10:2031. [PMID: 31507616 PMCID: PMC6718513 DOI: 10.3389/fimmu.2019.02031] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Accepted: 08/12/2019] [Indexed: 12/12/2022] Open
Abstract
Macrophages have emerged as a key component of the innate immune system that emigrates to peripheral tissues during gestation and in the adult organism. Their complex pathway to maturity, their unique plasticity and their various roles as effector and regulatory cells during an immune response have been the focus of intense research. A class of surface molecules, the G-Protein coupled receptors (GPCRs) play important roles in many immune processes. They have drawn attention in regard to these functions and the potential for therapeutic targets that can modulate the response of immune cells in pathologies such as diabetes, atherosclerosis, and chronic inflammatory diseases. Of the more than 800 GPCRs identified, ~100 are currently targeted with drugs which have had their activity investigated in vivo. Macrophages express a number of GPCRs which have central roles during cell differentiation and in the regulation of their functions. While some macrophage GPCRs such as chemokine receptors have been studied in great detail, the roles of other receptors of this large family are still not well understood. This review summarizes new insights into macrophage biology, differences of human, and mouse macrophages and gives details of some of the GPCRs expressed by this cell type.
Collapse
Affiliation(s)
- Xinming Wang
- Key Laboratory of Anti-inflammatory and Immune Medicine, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Ministry of Education, Anhui Medical University, Hefei, China.,Department of Pharmacy, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Abishek Iyer
- Institute for Molecular Bioscience, University of Queensland, Brisbane, QLD, Australia
| | - A Bruce Lyons
- School of Medicine, University of Tasmania, Hobart, TAS, Australia
| | - Heinrich Körner
- Key Laboratory of Anti-inflammatory and Immune Medicine, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Ministry of Education, Anhui Medical University, Hefei, China.,Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS, Australia
| | - Wei Wei
- Key Laboratory of Anti-inflammatory and Immune Medicine, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Ministry of Education, Anhui Medical University, Hefei, China
| |
Collapse
|