1
|
Rao SV, Lerner JBA. Inflammation in the Peri-ACS Period: Ready for Prime Time? Curr Atheroscler Rep 2024; 27:20. [PMID: 39714732 DOI: 10.1007/s11883-024-01263-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/02/2024] [Indexed: 12/24/2024]
Abstract
PURPOSE Inflammation has been demonstrated to negatively impact patients in the peri-ACS period. This narrative review outlines the inflammatory response in ACS, highlighting the role of the NLRP3 inflammasome pathway following acute plaque rupture and coronary intervention and its potential as a pharmacologic target. RECENT: nvestigators have leveraged medications targeting the NLRP3 inflammasome currently used for other inflammatory pathologies, including colchicine, tocilizumab and anakinra. Investigation into these drugs in the peri-ACS period has yielded varying results, with the most encouraging findings in ACS patients treated with tocilizumab. More conflicting data exists for the role of colchicine and anakinra, with many studies limited in their power to detect clinical outcomes and heterogeneity in their patient populations and endpoints. Despite conflicting data, the NLRP3 remains an attractive therapeutic target in the peri-ACS period. Further investigation is required to prove benefit and safety with large clinical trials adequately powered for clinical outcomes.
Collapse
Affiliation(s)
- Sunil V Rao
- Department of Medicine, Division of Cardiology, New York University Langone Medical Center, NYU Langone Health System, 550 1st Ave, New York, NY, 10010, USA.
| | - Johanna Ben-Ami Lerner
- Department of Medicine, Division of Cardiology, New York University Langone Medical Center, NYU Langone Health System, 550 1st Ave, New York, NY, 10010, USA
| |
Collapse
|
2
|
Juhasz V, Charlier FT, Zhao TX, Tsiantoulas D. Targeting the adaptive immune continuum in atherosclerosis and post-MI injury. Atherosclerosis 2024; 399:118616. [PMID: 39546915 DOI: 10.1016/j.atherosclerosis.2024.118616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 09/04/2024] [Accepted: 09/24/2024] [Indexed: 11/17/2024]
Abstract
Atherosclerotic disease is a cholesterol-rich lipoprotein particle-driven disease resulting in the formation of atherosclerotic plaques in large and medium size arteries. Rupture or erosion of atherosclerotic plaques can trigger the formation of a thrombus causing the obstruction of the blood flow in the coronary artery and thereby leading to myocardial infarction (MI). Inflammation is a crucial pillar of the mechanisms leading to atherosclerosis and governing the cardiac repair post-MI. Dissecting the complex and sophisticated networks of the immune responses underlying the formation of atherosclerotic plaques and affecting the healing of the heart after MI will allow the designing of highly precise immunomodulatory therapies for these settings. Notably, MI also accelerates atherosclerosis via modulating the response of the immune system. Therefore, for the identification of effective and safe therapeutic targets, it is critical to consider the inflammatory continuum that interconnects the two pathologies and identify immunomodulatory strategies that confer a protective effect in both settings or at least, affect each pathology independently. Adaptive immunity, which consists of B and T lymphocytes, is a major regulator of atherosclerosis and post-MI cardiac repair. Here, we review and discuss the effect of potential adaptive immunity-targeting therapies, such as cell-depleting therapies, in atherosclerosis and post-MI cardiac injury.
Collapse
Affiliation(s)
- Viktoria Juhasz
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Fiona T Charlier
- Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Tian X Zhao
- Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, Cambridge, United Kingdom; Department of Cardiology, Royal Papworth Hospital NHS Trust, Cambridge, United Kingdom
| | | |
Collapse
|
3
|
Gutiérrez-Aguilar M, Klutho PJ, Aguayo-Ortiz R, Song L, Baines CP. Endogenous complement 1q binding protein (C1qbp) regulates mitochondrial permeability transition and post-myocardial infarction remodeling and dysfunction. J Mol Cell Cardiol 2024; 196:1-11. [PMID: 39209214 PMCID: PMC11534557 DOI: 10.1016/j.yjmcc.2024.08.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 08/25/2024] [Accepted: 08/27/2024] [Indexed: 09/04/2024]
Abstract
The mitochondrial permeability transition (MPT) pore regulates necrotic cell death following diverse cardiac insults. While the componentry of the pore itself remains controversial, Cyclophilin D (CypD) has been well-established as a positive regulator of pore opening. We have previously identified Complement 1q-binding protein (C1qbp) as a novel CypD-interacting molecule and a negative regulator of MPT-dependent cell death in vitro. However, its effects on the MPT pore and sensitivity to cell death in the heart remain untested. We therefore hypothesized that C1qbp would inhibit MPT in cardiac mitochondria and protect cardiac myocytes against cell death in vivo. To investigate the effects of C1qbp in the myocardium we generated gain- and loss-of-function mice. Transgenic C1qbp overexpression resulted in decreased complex protein expression and reduced mitochondrial respiration and ATP production but MPT was unaffected. In contrast, while C1qbp+/- mice did not exhibit any changes in mitochondrial protein expression, respiration, or ATP, the MPT pore was markedly sensitized to Ca2+ in these animals. Neither overexpression nor depletion of C1qbp significantly affected baseline heart morphology or function at 3 months of age. When subjected to myocardial infarction, C1qbp transgenic mice exhibited similar infarct sizes and cardiac remodeling to non-transgenic mice, consistent with the lack of an effect on MPT. In contrast, cardiac scar formation and dysfunction were significantly increased in the C1qbp+/- mice compared to C1qbp+/+ controls. Our results suggest that C1qbp is required for normal regulation of the MPT pore and mitochondrial function, and influences cardiac remodeling following MI, the latter more likely being independent of C1qbp effects on the MPT pore.
Collapse
Affiliation(s)
- Manuel Gutiérrez-Aguilar
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO 65211, USA; Departamento de Bioquímica, Facultad de Química, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico
| | - Paula J Klutho
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO 65211, USA
| | - Rodrigo Aguayo-Ortiz
- Departamento de Farmacia, Facultad de Química, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico
| | - Lihui Song
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO 65211, USA
| | - Christopher P Baines
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO 65211, USA; Department of Biomedical Sciences, University of Missouri, Columbia, MO 65211, USA; Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, MO 65211, USA.
| |
Collapse
|
4
|
Li Z, Kang L, Jiang K. Analysis of the differences in immune-related genes and immune cell subtypes in acute myocardial infarction. Braz J Med Biol Res 2024; 57:e14345. [PMID: 39417451 PMCID: PMC11484353 DOI: 10.1590/1414-431x2024e14345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Accepted: 08/29/2024] [Indexed: 10/19/2024] Open
Abstract
Acute myocardial infarction (AMI) continues to be a leading cause of death globally, with distinct immune cell dynamics in ST-segment elevation myocardial infarction (STEMI) and non-ST-segment elevation myocardial infarction (NSTEMI) playing a critical role in disease progression and patient outcomes. Sample data for STEMI and NSTEMI were downloaded from the Sequence Read Archive (SRA) database (https://www.ncbi.nlm.nih.gov/sra). Differences and correlations of immune infiltrating cells were assessed by CIBERSORT. Differentially expressed genes (DEGs) were identified between STEMI and NSTEMI, followed by functional analysis. Immune-related DEGs were further identified. Some immune-related DEGs were selected to perform expression verification using real-time PCR. There was a significant difference in immune cells between STEMI and NSTEMI, including activated dendritic cells, memory CD4 T cells, mast cells, and CD8 T cells. A total of 229 DEGs were identified, with functions related to inflammatory regulation and drug metabolism. A total of 21 immune-related DEGs, which may play important roles in STEMI and NSTEMI, were identified. Among the 21 immune-related DEGs, genes like CCL18, NRP2, CXCR2, CXCL9, KIR2DL4, BPIFB1, and IL33 were significantly correlated with immune cells and had a tendency for differential expression between STEMI and NSTEMI patients. Our study reveals differences in the distribution of immune cell subsets between STEMI and NSTEMI, highlighting key immune-related genes and their association with immune cells, which may provide new insights into the treatment of AMI.
Collapse
Affiliation(s)
- Zhengmei Li
- School of Radiology, Shandong First Medical University & Shandong Academy of Medical Sciences, Tai'an, Shandong Province, China
| | - Ling Kang
- Department of Cardiology, The Second Affiliated Hospital of Shandong First Medical University, Tai'an, Shandong Province, China
| | - Ke Jiang
- Department of Cardiology, The Second Affiliated Hospital of Shandong First Medical University, Tai'an, Shandong Province, China
| |
Collapse
|
5
|
Lin Z, Jiwani Z, Serpooshan V, Aghaverdi H, Yang PC, Aguirre A, Wu JC, Mahmoudi M. Sex Influences the Safety and Therapeutic Efficacy of Cardiac Nanomedicine Technologies. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2305940. [PMID: 37803920 PMCID: PMC10997742 DOI: 10.1002/smll.202305940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 09/18/2023] [Indexed: 10/08/2023]
Abstract
Nanomedicine technologies are being developed for the prevention, diagnosis, and treatment of cardiovascular disease (CVD), which is the leading cause of death worldwide. Before delving into the nuances of cardiac nanomedicine, it is essential to comprehend the fundamental sex-specific differences in cardiovascular health. Traditionally, CVDs have been more prevalent in males, but it is increasingly evident that females also face significant risks, albeit with distinct characteristics. Females tend to develop CVDs at a later age, exhibit different clinical symptoms, and often experience worse outcomes compared to males. These differences indicate the need for sex-specific approaches in cardiac nanomedicine. This Perspective discusses the importance of considering sex in the safety and therapeutic efficacy of nanomedicine approaches for the prevention, diagnosis, and treatment of CVD.
Collapse
Affiliation(s)
- Zijin Lin
- Department of Radiology and Precision Health Program, Michigan State University, East Lansing, MI 48824 USA
| | - Zahra Jiwani
- Department of Radiology and Precision Health Program, Michigan State University, East Lansing, MI 48824 USA
| | - Vahid Serpooshan
- Department of Biomedical Engineering, Emory University School of Medicine and Georgia Institute of Technology, Atlanta, GA 30322, USA
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA 30322, USA
- Children’s Healthcare of Atlanta, Atlanta, GA 30322, USA
| | - Haniyeh Aghaverdi
- Department of Radiology and Precision Health Program, Michigan State University, East Lansing, MI 48824 USA
| | - Phillip C Yang
- Department of Medicine, Cardiovascular Medicine and Cardiovascular Institute, Stanford University, Stanford, CA 94309
| | - Aitor Aguirre
- Regenerative Biology and cell Reprogramming Laboratory, Institute for Quantitative Health Sciences and Engineering, Michigan State University, East Lansing, MI 48823, USA
- Department of Biomedical Engineering, Michigan State University, East Lansing, MI 48823, USA
| | - Joseph C. Wu
- Department of Medicine, Cardiovascular Medicine and Cardiovascular Institute, Stanford University, Stanford, CA 94309
- Department of Medicine, Division of Cardiology, Stanford University, Stanford, CA 94305, USA
| | - Morteza Mahmoudi
- Department of Radiology and Precision Health Program, Michigan State University, East Lansing, MI 48824 USA
- Connors Center for Women’s Health & Gender Biology, Brigham & Women’s Hospital, Harvard Medical School, Boston, MA 02115 USA
| |
Collapse
|
6
|
Cimerman M, Druzhaeva N, Nemec Svete A, Hajdinjak M, Pohar K, Ihan A, Domanjko Petrič A. Inflammatory and immune variables as predictors of survival in dogs with myxomatous mitral valve disease. BMC Vet Res 2024; 20:431. [PMID: 39342325 PMCID: PMC11438168 DOI: 10.1186/s12917-024-04266-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Accepted: 09/03/2024] [Indexed: 10/01/2024] Open
Abstract
BACKGROUND We aimed to investigate the association between selected inflammatory and immune variables and survival of dogs with myxomatous mitral valve disease (MMVD). We evaluated data of 62 client-owned dogs with MMVD, grouped into preclinical, stable congestive heart failure (CHF) and unstable CHF. Univariate Cox proportional hazards regression analysis was used to quantify the association of white blood cell count, concentrations and percentages of T lymphocytes and their subtypes (T helper lymphocytes, cytotoxic T lymphocytes, double positive T lymphocytes, double negative T lymphocytes) and B lymphocytes with survival. P values < 0.1 in individual groups and P values < 0.05 in the group of all patients were considered significant. Spearman correlation coefficients between significant covariates were calculated to assess the relationships among variables and with survival. RESULTS In the preclinical group, percentage of double positive T lymphocytes was negatively associated with survival (hazard ratio (HR) = 2.328; P = 0.051). In the unstable CHF, T lymphocyte (HR = 1.613; P = 0.085), cytotoxic T lymphocyte (HR = 1.562; P = 0.048), double positive (HR = 1.751; P = 0.042), and double negative T lymphocyte (HR = 1.613; P = 0.096) concentrations were negatively associated with survival, as well as cytotoxic T lymphocyte (HR = 1.502; P = 0.007) concentration in the group of all patients. The percentage of T helper lymphocytes was positively associated with survival in the unstable CHF (HR = 0.604; P = 0.053) and in the group of all patients (HR = 0.733; P = 0.044). The concentration of cytotoxic T lymphocytes positively correlated with left atrial to aortic ratio (LA/Ao) (rho = 0.259, P = 0.037), and peak velocity of early diastolic mitral flow (rho = 0.259, P = 0.039), whereas the percentage of T helper lymphocytes negatively correlated with left atrial to aortic ratio (LA/Ao) (rho = -0.212, P = 0.090) and early to late mitral flow ratio (rho = -0.232, P = 0.072). CONCLUSIONS Cytotoxic T lymphocytes, T helper lymphocytes, double positive and double negative T lymphocytes as well as biomarkers cardiac troponin I, N-terminal pro-B-type natriuretic peptide, C-reactive protein are implicated in the progression of MMVD.
Collapse
Affiliation(s)
- Martina Cimerman
- Veterinary Faculty, Small Animal Clinic, University of Ljubljana, Ljubljana, Slovenia
| | - Natalia Druzhaeva
- Veterinary Faculty, Small Animal Clinic, University of Ljubljana, Ljubljana, Slovenia
| | - Alenka Nemec Svete
- Veterinary Faculty, Small Animal Clinic, University of Ljubljana, Ljubljana, Slovenia
| | - Melita Hajdinjak
- Faculty of Electrical Engineering, Laboratory of Applied Mathematics and Statistics, University of Ljubljana, Ljubljana, Slovenia
| | - Katka Pohar
- Faculty of Medicine, Institute of Microbiology and Immunology, University of Ljubljana, Ljubljana, Slovenia
| | - Alojz Ihan
- Faculty of Medicine, Institute of Microbiology and Immunology, University of Ljubljana, Ljubljana, Slovenia
| | | |
Collapse
|
7
|
Duni A, Kitsos A, Bechlioulis A, Lakkas L, Markopoulos G, Tatsis V, Koutlas V, Tzalavra E, Baxevanos G, Vartholomatos G, Mitsis M, Naka KK, Dounousi E. Identification of Novel Independent Correlations between Cellular Components of the Immune System and Strain-Related Indices of Myocardial Dysfunction in CKD Patients and Kidney Transplant Recipients without Established Cardiovascular Disease. Int J Mol Sci 2024; 25:9162. [PMID: 39273110 PMCID: PMC11395156 DOI: 10.3390/ijms25179162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 08/15/2024] [Accepted: 08/21/2024] [Indexed: 09/15/2024] Open
Abstract
The role of immune system components in the development of myocardial remodeling in chronic kidney disease (CKD) and kidney transplantation remains an open question. Our aim was to investigate the associations between immune cell subpopulations in the circulation of CKD patients and kidney transplant recipients (KTRs) with subclinical indices of myocardial performance. We enrolled 44 CKD patients and 38 KTRs without established cardiovascular disease. A selected panel of immune cells was measured by flow cytometry. Classical and novel strain-related indices of ventricular function were measured by speckle-tracking echocardiography at baseline and following dipyridamole infusion. In CKD patients, the left ventricular (LV) relative wall thickness correlated with the CD14++CD16- monocytes (β = 0.447, p = 0.004), while the CD14++CD16+ monocytes were independent correlates of the global radial strain (β = 0.351, p = 0.04). In KTRs, dipyridamole induced changes in global longitudinal strain correlated with CD14++CD16+ monocytes (β = 0.423, p = 0.009) and CD4+ T-cells (β = 0.403, p = 0.01). LV twist and untwist were independently correlated with the CD8+ T-cells (β = 0.405, p = 0.02 and β = -0.367, p = 0.03, respectively) in CKD patients, whereas the CD14++CD16+ monocytes were independent correlates of LV twist and untwist in KTRs (β = 0.405, p = 0.02 and β = -0.367, p = 0.03, respectively). Immune cell subsets independently correlate with left ventricular strain and torsion-related indices in CKD patients and KTRs without established CVD.
Collapse
Affiliation(s)
- Anila Duni
- Department of Nephrology, Faculty of Medicine, School of Health Sciences, University Hospital of Ioannina, University of Ioannina, 455 00 Ioannina, Greece
- Kidney Transplant Unit, Department of Surgery, Faculty of Medicine, School of Health Sciences, University Hospital of Ioannina, University of Ioannina, 455 00 Ioannina, Greece
| | - Athanasios Kitsos
- Department of Nephrology, Faculty of Medicine, School of Health Sciences, University Hospital of Ioannina, University of Ioannina, 455 00 Ioannina, Greece
- Kidney Transplant Unit, Department of Surgery, Faculty of Medicine, School of Health Sciences, University Hospital of Ioannina, University of Ioannina, 455 00 Ioannina, Greece
| | - Aris Bechlioulis
- Second Department of Cardiology, Faculty of Medicine, School of Health Sciences, University Hospital of Ioannina, University of Ioannina, 455 00 Ioannina, Greece
| | - Lampros Lakkas
- Second Department of Cardiology, Faculty of Medicine, School of Health Sciences, University Hospital of Ioannina, University of Ioannina, 455 00 Ioannina, Greece
| | - Georgios Markopoulos
- Laboratory of Haematology-Unit of Molecular Biology and Translational Flow Cytometry, University Hospital of Ioannina, 455 00 Ioannina, Greece
| | - Vasileios Tatsis
- Kidney Transplant Unit, Department of Surgery, Faculty of Medicine, School of Health Sciences, University Hospital of Ioannina, University of Ioannina, 455 00 Ioannina, Greece
| | - Vasileios Koutlas
- Kidney Transplant Unit, Department of Surgery, Faculty of Medicine, School of Health Sciences, University Hospital of Ioannina, University of Ioannina, 455 00 Ioannina, Greece
| | - Eirini Tzalavra
- Kidney Transplant Unit, Department of Surgery, Faculty of Medicine, School of Health Sciences, University Hospital of Ioannina, University of Ioannina, 455 00 Ioannina, Greece
| | - Gerasimos Baxevanos
- Laboratory of Haematology-Unit of Molecular Biology and Translational Flow Cytometry, University Hospital of Ioannina, 455 00 Ioannina, Greece
- Department of Internal Medicine, General Hospital of Ioannina, G. Chatzikosta, 454 45 Ioannina, Greece
| | - Georgios Vartholomatos
- Laboratory of Haematology-Unit of Molecular Biology and Translational Flow Cytometry, University Hospital of Ioannina, 455 00 Ioannina, Greece
| | - Michail Mitsis
- Kidney Transplant Unit, Department of Surgery, Faculty of Medicine, School of Health Sciences, University Hospital of Ioannina, University of Ioannina, 455 00 Ioannina, Greece
| | - Katerina K Naka
- Second Department of Cardiology, Faculty of Medicine, School of Health Sciences, University Hospital of Ioannina, University of Ioannina, 455 00 Ioannina, Greece
| | - Evangelia Dounousi
- Department of Nephrology, Faculty of Medicine, School of Health Sciences, University Hospital of Ioannina, University of Ioannina, 455 00 Ioannina, Greece
- Kidney Transplant Unit, Department of Surgery, Faculty of Medicine, School of Health Sciences, University Hospital of Ioannina, University of Ioannina, 455 00 Ioannina, Greece
| |
Collapse
|
8
|
Alshoubaki YK, Nayer B, Lu YZ, Salimova E, Lau SN, Tan JL, Amann-Zalcenstein D, Hickey PF, Del Monte-Nieto G, Vasanthakumar A, Martino MM. Tregs delivered post-myocardial infarction adopt an injury-specific phenotype promoting cardiac repair via macrophages in mice. Nat Commun 2024; 15:6480. [PMID: 39090108 PMCID: PMC11294480 DOI: 10.1038/s41467-024-50806-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Accepted: 07/22/2024] [Indexed: 08/04/2024] Open
Abstract
Regulatory T cells (Tregs) are key immune regulators that have shown promise in enhancing cardiac repair post-MI, although the mechanisms remain elusive. Here, we show that rapidly increasing Treg number in the circulation post-MI via systemic administration of exogenous Tregs improves cardiac function in male mice, by limiting cardiomyocyte death and reducing fibrosis. Mechanistically, exogenous Tregs quickly home to the infarcted heart and adopt an injury-specific transcriptome that mediates repair by modulating monocytes/macrophages. Specially, Tregs lead to a reduction in pro-inflammatory Ly6CHi CCR2+ monocytes/macrophages accompanied by a rapid shift of macrophages towards a pro-repair phenotype. Additionally, exogenous Treg-derived factors, including nidogen-1 and IL-10, along with a decrease in cardiac CD8+ T cell number, mediate the reduction of the pro-inflammatory monocyte/macrophage subset in the heart. Supporting the pivotal role of IL-10, exogenous Tregs knocked out for IL-10 lose their pro-repair capabilities. Together, this study highlights the beneficial use of a Treg-based therapeutic approach for cardiac repair with important mechanistic insights that could facilitate the development of novel immunotherapies for MI.
Collapse
Affiliation(s)
- Yasmin K Alshoubaki
- European Molecular Biology Laboratory Australia, Australian Regenerative Medicine Institute, Monash University, Melbourne, Australia
| | - Bhavana Nayer
- European Molecular Biology Laboratory Australia, Australian Regenerative Medicine Institute, Monash University, Melbourne, Australia
| | - Yen-Zhen Lu
- European Molecular Biology Laboratory Australia, Australian Regenerative Medicine Institute, Monash University, Melbourne, Australia
| | | | - Sin Nee Lau
- European Molecular Biology Laboratory Australia, Australian Regenerative Medicine Institute, Monash University, Melbourne, Australia
| | - Jean L Tan
- European Molecular Biology Laboratory Australia, Australian Regenerative Medicine Institute, Monash University, Melbourne, Australia
| | - Daniela Amann-Zalcenstein
- Advanced Genomics Facility, Advanced Technology and Biology Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, Australia
| | - Peter F Hickey
- Advanced Genomics Facility, Advanced Technology and Biology Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, Australia
| | - Gonzalo Del Monte-Nieto
- European Molecular Biology Laboratory Australia, Australian Regenerative Medicine Institute, Monash University, Melbourne, Australia
- Victorian Heart Institute, Monash University, Victorian Heart Hospital, Melbourne, Australia
| | - Ajithkumar Vasanthakumar
- Olivia Newton-John Cancer Research Institute, Heidelberg, Australia
- La Trobe University, Bundoora, Australia
- Department of Microbiology and Immunology, University of Melbourne, Melbourne, Australia
| | - Mikaël M Martino
- European Molecular Biology Laboratory Australia, Australian Regenerative Medicine Institute, Monash University, Melbourne, Australia.
- Victorian Heart Institute, Monash University, Victorian Heart Hospital, Melbourne, Australia.
| |
Collapse
|
9
|
Ramos-Regalado L, Alcover S, Badimon L, Vilahur G. The Influence of Metabolic Risk Factors on the Inflammatory Response Triggered by Myocardial Infarction: Bridging Pathophysiology to Treatment. Cells 2024; 13:1125. [PMID: 38994977 PMCID: PMC11240659 DOI: 10.3390/cells13131125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 06/19/2024] [Accepted: 06/27/2024] [Indexed: 07/13/2024] Open
Abstract
Myocardial infarction (MI) sets off a complex inflammatory cascade that is crucial for effective cardiac healing and scar formation. Yet, if this response becomes excessive or uncontrolled, it can lead to cardiovascular complications. This review aims to provide a comprehensive overview of the tightly regulated local inflammatory response triggered in the early post-MI phase involving cardiomyocytes, (myo)fibroblasts, endothelial cells, and infiltrating immune cells. Next, we explore how the bone marrow and extramedullary hematopoiesis (such as in the spleen) contribute to sustaining immune cell supply at a cardiac level. Lastly, we discuss recent findings on how metabolic cardiovascular risk factors, including hypercholesterolemia, hypertriglyceridemia, diabetes, and hypertension, disrupt this immunological response and explore the potential modulatory effects of lifestyle habits and pharmacological interventions. Understanding how different metabolic risk factors influence the inflammatory response triggered by MI and unraveling the underlying molecular and cellular mechanisms may pave the way for developing personalized therapeutic approaches based on the patient's metabolic profile. Similarly, delving deeper into the impact of lifestyle modifications on the inflammatory response post-MI is crucial. These insights may enable the adoption of more effective strategies to manage post-MI inflammation and improve cardiovascular health outcomes in a holistic manner.
Collapse
Affiliation(s)
- Lisaidy Ramos-Regalado
- Research Institute, Hospital de la Santa Creu i Sant Pau, IIB-Sant Pau, 08025 Barcelona, Spain (S.A.)
- Faculty of Biology, Universitat de Barcelona, 08028 Barcelona, Spain
| | - Sebastià Alcover
- Research Institute, Hospital de la Santa Creu i Sant Pau, IIB-Sant Pau, 08025 Barcelona, Spain (S.A.)
- Faculty of Biology, Universitat de Barcelona, 08028 Barcelona, Spain
| | - Lina Badimon
- Research Institute, Hospital de la Santa Creu i Sant Pau, IIB-Sant Pau, 08025 Barcelona, Spain (S.A.)
- Ciber CV, Institute Carlos III, 28029 Madrid, Spain
- Cardiovascular Research Chair, Universitat Autònoma de Barcelona (UAB), 08193 Barcelona, Spain
| | - Gemma Vilahur
- Research Institute, Hospital de la Santa Creu i Sant Pau, IIB-Sant Pau, 08025 Barcelona, Spain (S.A.)
- Ciber CV, Institute Carlos III, 28029 Madrid, Spain
| |
Collapse
|
10
|
Wang Y, Gao Y, Shi H, Gao R, Yang J, Qu Y, Hu S, Zhang J, Wang J, Cao J, Zhang F, Ge J. CCL11 released by GSDMD-mediated macrophage pyroptosis regulates angiogenesis after hindlimb ischemia. Cell Death Discov 2024; 10:294. [PMID: 38906863 PMCID: PMC11192718 DOI: 10.1038/s41420-023-01764-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2023] [Revised: 12/02/2023] [Accepted: 12/06/2023] [Indexed: 06/23/2024] Open
Abstract
Peripheral vascular disease (PVD) is an emerging public health burden with a high rate of disability and mortality. Gasdermin D (GSDMD) has been reported to exert pyroptosis and play a critical role in the pathophysiology of many cardiovascular diseases. We ought to determine the role of GSDMD in the regulation of perfusion recovery after hindlimb ischemia (HLI). Our study revealed that GSDMD-mediated pyroptosis occurred in HLI. GSDMD deletion aggravated perfusion recovery and angiogenesis in vitro and in vivo. However, how GSDMD regulates angiogenesis after ischemic injury remains unclear. We then found that GSDMD-mediated pyroptosis exerted the angiogenic capacity in macrophages rather than endothelial cells after HLI. GSDMD deletion led to a lower level of CCL11 in mice serum. GSDMD knockdown in macrophages downregulated the expression and decreased the releasing level of CCL11. Furthermore, recombinant CCL11 improved endothelial functions and angiogenesis, which was attenuated by CCL11 antibody. Taken together, these results demonstrate that GSDMD promotes angiogenesis by releasing CCL11, thereby improving blood flow perfusion recovery after hindlimb ischemic injury. Therefore, CCL11 may be a novel target for prevention and treatment of vascular ischemic diseases.
Collapse
Affiliation(s)
- Yiwen Wang
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, 200032, Shanghai, China
| | - Yang Gao
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, 200032, Shanghai, China
| | - Huairui Shi
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, 200032, Shanghai, China
| | - Rifeng Gao
- Department of Cardiology, Shanghai Fifth People's Hospital, Fudan University, 200240, Shanghai, China
| | - Ji'e Yang
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, 200032, Shanghai, China
| | - Ya'nan Qu
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, 200032, Shanghai, China
| | - Shiyu Hu
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, 200032, Shanghai, China
| | - Jian Zhang
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, 200032, Shanghai, China
| | - Jingpu Wang
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, 200032, Shanghai, China
| | - Jiatian Cao
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, 200032, Shanghai, China
| | - Feng Zhang
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, 200032, Shanghai, China.
| | - Junbo Ge
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, 200032, Shanghai, China.
- Key Laboratory of Viral Heart Diseases, National Health Commission, 200032, Shanghai, China.
- Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, 200032, Shanghai, China.
- National Clinical Research Center for Interventional Medicine, 200032, Shanghai, China.
- Institutes of Biomedical Sciences, Fudan University, 200032, Shanghai, China.
| |
Collapse
|
11
|
Zhou T, Pan J, Yan C, Yuan J, Song H, Han Y. Unveiling shared biomarkers and therapeutic targets between systemic lupus erythematosus and heart failure through bioinformatics analysis. Front Med (Lausanne) 2024; 11:1402010. [PMID: 38912340 PMCID: PMC11190381 DOI: 10.3389/fmed.2024.1402010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2024] [Accepted: 05/21/2024] [Indexed: 06/25/2024] Open
Abstract
Background Systemic lupus erythematosus (SLE) is frequently accompanied by various complications, with cardiovascular diseases being particularly concerning due to their high mortality rate. Although there is clinical evidence suggesting a potential correlation between SLE and heart failure (HF), the underlying shared mechanism is not fully understood. Therefore, it is imperative to explore the potential mechanisms and shared therapeutic targets between SLE and HF. Methods The SLE and HF datasets were downloaded from the NCBI Gene Expression Omnibus database. Differentially expressed genes (DEGs) in both SLE and HF were performed using "limma" R package. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genes (KEGG) analyses were conducted to analyze the enriched functions and pathways of DEGs in both SLE and HF datasets. Protein-Protein Interaction network (PPI) and the molecular complex detection (MCODE) plugins in the Cytoscape software were performed to identify the shared hub genes between SLE and HF datasets. R package "limma" was utilized to validate the expression of hub genes based on SLE (GSE122459) and HF (GSE196656) datasets. CIBERSORT algorithm was utilized to analyze the immune cell infiltration of SLE and HF samples based on SLE (GSE112087) and HF (GSE116250) datasets. A weighted gene co-expression network analysis (WGCNA) network was established to further validate the hub genes based on HF dataset (GSE116250). Molecular biology techniques were conducted to validate the hub genes. Results 999 shared DGEs were identified between SLE and HF datasets, which were mainly enriched in pathways related to Th17 cell differentiation. 5 shared hub genes among the common DGEs between SLE and HF datasets were screened and validated, including HSP90AB1, NEDD8, RPLP0, UBB, and UBC. Additionally, 5 hub genes were identified in the central part of the MEbrown module, showing the strongest correlation with dilated cardiomyopathy. HSP90AB1 and UBC were upregulated in failing hearts compared to non-failing hearts, while UBB, NEDD8, and RPLP0 did not show significant changes. Conclusion HSP90AB1 and UBC are closely related to the co-pathogenesis of SLE and HF mediated by immune cell infiltration. They serve as promising molecular markers and potential therapeutic targets for the treatment of SLE combined with HF.
Collapse
Affiliation(s)
- Ting Zhou
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- State Key Laboratory of Frigid Zone Cardiovascular Disease, Cardiovascular Research Institute and Department of Cardiology, General Hospital of Northern Theater Command, Shenyang, China
| | - Jing Pan
- State Key Laboratory of Frigid Zone Cardiovascular Disease, Cardiovascular Research Institute and Department of Cardiology, General Hospital of Northern Theater Command, Shenyang, China
| | - Chenghui Yan
- State Key Laboratory of Frigid Zone Cardiovascular Disease, Cardiovascular Research Institute and Department of Cardiology, General Hospital of Northern Theater Command, Shenyang, China
| | - Jing Yuan
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Haixu Song
- State Key Laboratory of Frigid Zone Cardiovascular Disease, Cardiovascular Research Institute and Department of Cardiology, General Hospital of Northern Theater Command, Shenyang, China
| | - Yaling Han
- State Key Laboratory of Frigid Zone Cardiovascular Disease, Cardiovascular Research Institute and Department of Cardiology, General Hospital of Northern Theater Command, Shenyang, China
| |
Collapse
|
12
|
Hilgendorf I, Frantz S, Frangogiannis NG. Repair of the Infarcted Heart: Cellular Effectors, Molecular Mechanisms and Therapeutic Opportunities. Circ Res 2024; 134:1718-1751. [PMID: 38843294 PMCID: PMC11164543 DOI: 10.1161/circresaha.124.323658] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2024] [Accepted: 05/08/2024] [Indexed: 06/12/2024]
Abstract
The adult mammalian heart has limited endogenous regenerative capacity and heals through the activation of inflammatory and fibrogenic cascades that ultimately result in the formation of a scar. After infarction, massive cardiomyocyte death releases a broad range of damage-associated molecular patterns that initiate both myocardial and systemic inflammatory responses. TLRs (toll-like receptors) and NLRs (NOD-like receptors) recognize damage-associated molecular patterns (DAMPs) and transduce downstream proinflammatory signals, leading to upregulation of cytokines (such as interleukin-1, TNF-α [tumor necrosis factor-α], and interleukin-6) and chemokines (such as CCL2 [CC chemokine ligand 2]) and recruitment of neutrophils, monocytes, and lymphocytes. Expansion and diversification of cardiac macrophages in the infarcted heart play a major role in the clearance of the infarct from dead cells and the subsequent stimulation of reparative pathways. Efferocytosis triggers the induction and release of anti-inflammatory mediators that restrain the inflammatory reaction and set the stage for the activation of reparative fibroblasts and vascular cells. Growth factor-mediated pathways, neurohumoral cascades, and matricellular proteins deposited in the provisional matrix stimulate fibroblast activation and proliferation and myofibroblast conversion. Deposition of a well-organized collagen-based extracellular matrix network protects the heart from catastrophic rupture and attenuates ventricular dilation. Scar maturation requires stimulation of endogenous signals that inhibit fibroblast activity and prevent excessive fibrosis. Moreover, in the mature scar, infarct neovessels acquire a mural cell coat that contributes to the stabilization of the microvascular network. Excessive, prolonged, or dysregulated inflammatory or fibrogenic cascades accentuate adverse remodeling and dysfunction. Moreover, inflammatory leukocytes and fibroblasts can contribute to arrhythmogenesis. Inflammatory and fibrogenic pathways may be promising therapeutic targets to attenuate heart failure progression and inhibit arrhythmia generation in patients surviving myocardial infarction.
Collapse
Affiliation(s)
- Ingo Hilgendorf
- Department of Cardiology and Angiology, University Heart Center Freiburg-Bad Krozingen and Faculty of Medicine at the University of Freiburg, Freiburg, Germany
| | - Stefan Frantz
- Medizinische Klinik und Poliklinik I, Universitätsklinikum Würzburg, Würzburg, Germany
| | - Nikolaos G Frangogiannis
- The Wilf Family Cardiovascular Research Institute, Department of Medicine (Cardiology), Albert Einstein College of Medicine, Bronx NY
| |
Collapse
|
13
|
Rani D, Kaur S, Shahjahan, Dey JK, Dey SK. Engineering immune response to regulate cardiovascular disease and cancer. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2024; 140:381-417. [PMID: 38762276 DOI: 10.1016/bs.apcsb.2023.12.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2024]
Abstract
Cardiovascular disease (CVD) and cancer are major contributors to global morbidity and mortality. This book chapter delves into the intricate relationship between the immune system and the pathogenesis of both cardiovascular and cancer diseases, exploring the roles of innate and adaptive immunities, immune regulation, and immunotherapy in these complex conditions. The innate immune system acts as the first line of defense against tissue damage and infection, with a significant impact on the initiation and progression of CVD and cancer. Endothelial dysfunction, a hallmark in CVD, shares commonalities with the tumor microenvironment in cancer, emphasizing the parallel involvement of the immune system in both conditions. The adaptive immune system, particularly T cells, contributes to prolonged inflammation in both CVD and cancer. Regulatory T cells and the intricate balance between different T cell subtypes influence disease progression, wound healing, and the outcomes of ischemic injury and cancer immunosurveillance. Dysregulation of immune homeostasis can lead to chronic inflammation, contributing to the development and progression of both CVD and cancer. Thus, immunotherapy emerged as a promising avenue for preventing and managing these diseases, with strategies targeting immune cell modulation, cytokine manipulation, immune checkpoint blockade, and tolerance induction. The impact of gut microbiota on CVD and cancer too is explored in this chapter, highlighting the role of gut leakiness, microbial metabolites, and the potential for microbiome-based interventions in cardiovascular and cancer immunotherapies. In conclusion, immunomodulatory strategies and immunotherapy hold promise in reshaping the landscape of cardiovascular and cancer health. Additionally, harnessing the gut microbiota for immune modulation presents a novel approach to prevent and manage these complex diseases, emphasizing the importance of personalized and precision medicine in healthcare. Ongoing research and clinical trials are expected to further elucidate the complex immunological underpinnings of CVD and cancer thereby refining these innovative approaches.
Collapse
Affiliation(s)
- Diksha Rani
- Laboratory for Structural Biology of Membrane Proteins, Dr. B.R. Ambedkar Center for Biomedical Research, University of Delhi, New Delhi, Delhi, India
| | - Smaranjot Kaur
- Laboratory for Structural Biology of Membrane Proteins, Dr. B.R. Ambedkar Center for Biomedical Research, University of Delhi, New Delhi, Delhi, India
| | - Shahjahan
- Laboratory for Structural Biology of Membrane Proteins, Dr. B.R. Ambedkar Center for Biomedical Research, University of Delhi, New Delhi, Delhi, India
| | - Joy Kumar Dey
- Central Council for Research in Homoeopathy, Ministry of Ayush, Govt. of India, New Delhi, Delhi, India
| | - Sanjay Kumar Dey
- Laboratory for Structural Biology of Membrane Proteins, Dr. B.R. Ambedkar Center for Biomedical Research, University of Delhi, New Delhi, Delhi, India.
| |
Collapse
|
14
|
Xu H, Wei Z, Chen B, Wang J, Weng H, Li J, Yang X, Zhao S. Granzyme B PET imaging inflammation and remodeling in myocardial infarction. Eur J Nucl Med Mol Imaging 2024; 51:991-1001. [PMID: 37991527 DOI: 10.1007/s00259-023-06521-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Accepted: 11/13/2023] [Indexed: 11/23/2023]
Abstract
PURPOSE This study aimed to evaluate whether granzyme B (GzmB)-targeted positron emission tomography (PET) imaging agent (68 Ga-grazytracer) can characterize cardiac inflammation and remodeling in myocardial infarction (MI). METHODS Rats with MI were subjected to GzmB-targeted PET/CT on post-operative days 1, 3, 6, 14, and 28. Autoradiography, Masson staining, immunohistochemistry, and ELISA were performed to verify the inflammatory response and remodeling after MI in vitro. Rats were treated with GzmB inhibitor Z-IETD-FMK to improve cardiac remodeling. Cardiac function tests were performed by echocardiography at 6 weeks after MI. RESULTS The highest uptake of 68 Ga-grazytracer was observed on day 3 after MI compared with the values obtained on the other days (0.294 ± 0.03% ID/g at 3 days vs. 0.122 ± 0.01% ID/g in the sham group, P < 0.001). Immunohistochemistry showed significantly high expression of GzmB and CD8, in line with the PET/CT imaging results. Autoradiography revealed 68 Ga-grazytracer accumulation in the infarcted myocardium. The 68 Ga-grazytracer uptake of treated rats was significantly reduced compared with that in the MI groups (0.184 ± 0.03%ID/g vs. 0.286 ± 0.03%ID/g; P < 0.001). Echocardiography showed that the left ventricular ejection fraction was lower in the MI groups than in the ischemia reperfusion group. GzmB inhibitor treatment was shown to be effective in improving cardiac function without significantly shortening infarct size. CONCLUSIONS This study demonstrated the potential of 68 Ga-grazytracer imaging to delineate adverse inflammatory responses and pathological cardiac remodeling, which can help predict heart function. PET/CT imaging-guided therapy may reduce myocardial injury and improve heart function in MI.
Collapse
Affiliation(s)
- Hongchuang Xu
- Department of Nuclear Medicine, Peking University First Hospital, Xishiku Rd 8, Xicheng District, Beijing, 100034, China
| | - Zhuxin Wei
- Department of MRI, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beilishi Rd 167, Xicheng District, Beijing, 100037, China
| | - Bixi Chen
- Department of Nuclear Medicine, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China
| | - Jiaxin Wang
- Department of MRI, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beilishi Rd 167, Xicheng District, Beijing, 100037, China
| | - Haoyu Weng
- Department of Cardiology, Peking University First Hospital, Xishiku Rd 8, Xicheng District, Beijing, 100034, China
| | - Jianping Li
- Department of Cardiology, Peking University First Hospital, Xishiku Rd 8, Xicheng District, Beijing, 100034, China.
| | - Xing Yang
- Department of Nuclear Medicine, Peking University First Hospital, Xishiku Rd 8, Xicheng District, Beijing, 100034, China.
- Department of Central Laboratory, Peking University First Hospital, Beijing, 100034, China.
| | - Shihua Zhao
- Department of MRI, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beilishi Rd 167, Xicheng District, Beijing, 100037, China.
| |
Collapse
|
15
|
Abdalla AME, Miao Y, Ahmed AIM, Meng N, Ouyang C. CAR-T cell therapeutic avenue for fighting cardiac fibrosis: Roadblocks and perspectives. Cell Biochem Funct 2024; 42:e3955. [PMID: 38379220 DOI: 10.1002/cbf.3955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 02/06/2024] [Accepted: 02/09/2024] [Indexed: 02/22/2024]
Abstract
Heart diseases remain the primary cause of human mortality in the world. Although conventional therapeutic opportunities fail to halt or recover cardiac fibrosis, the promising clinical results and therapeutic efficacy of engineered chimeric antigen receptor (CAR) T cell therapy show several advancements. However, the current models of CAR-T cells need further improvement since the T cells are associated with the triggering of excessive inflammatory cytokines that directly affect cardiac functions. Thus, the current study highlights the critical function of heart immune cells in tissue fibrosis and repair. The study also confirms CAR-T cell as an emerging therapeutic for treating cardiac fibrosis, explores the current roadblocks to CAR-T cell therapy, and considers future outlooks for research development.
Collapse
Affiliation(s)
- Ahmed M E Abdalla
- School of Biological Sciences and Technology, University of Jinan, Jinan, China
- Department of Biochemistry, College of Applied Science, University of Bahri, Khartoum, Sudan
| | - Yu Miao
- NHC Key Laboratory of Diagnosis and Therapy of Gastrointestinal Tumor, Gansu Provincial Hospital, Lanzhou, China
- Key Laboratory of Molecular Diagnostics and Precision Medicine for Surgical Oncology in Gansu Province, Gansu Provincial Hospital, Lanzhou, Gansu, China
| | - Ahmed I M Ahmed
- Department of Biochemistry, College of Applied Science, University of Bahri, Khartoum, Sudan
| | - Ning Meng
- School of Biological Sciences and Technology, University of Jinan, Jinan, China
| | - Chenxi Ouyang
- Department of Vascular Surgery, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
16
|
Gonzalez AL, Dungan MM, Smart CD, Madhur MS, Doran AC. Inflammation Resolution in the Cardiovascular System: Arterial Hypertension, Atherosclerosis, and Ischemic Heart Disease. Antioxid Redox Signal 2024; 40:292-316. [PMID: 37125445 PMCID: PMC11071112 DOI: 10.1089/ars.2023.0284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 04/12/2023] [Indexed: 05/02/2023]
Abstract
Significance: Chronic inflammation has emerged as a major underlying cause of many prevalent conditions in the Western world, including cardiovascular diseases. Although targeting inflammation has emerged as a promising avenue by which to treat cardiovascular disease, it is also associated with increased risk of infection. Recent Advances: Though previously assumed to be passive, resolution has now been identified as an active process, mediated by unique immunoresolving mediators and mechanisms designed to terminate acute inflammation and promote tissue repair. Recent work has determined that failures of resolution contribute to chronic inflammation and the progression of human disease. Specifically, failure to produce pro-resolving mediators and the impaired clearance of dead cells from inflamed tissue have been identified as major mechanisms by which resolution fails in disease. Critical Issues: Drawing from a rapidly expanding body of experimental and clinical studies, we review here what is known about the role of inflammation resolution in arterial hypertension, atherosclerosis, myocardial infarction, and ischemic heart disease. For each, we discuss the involvement of specialized pro-resolving mediators and pro-reparative cell types, including T regulatory cells, myeloid-derived suppressor cells, and macrophages. Future Directions: Pro-resolving therapies offer the promise of limiting chronic inflammation without impairing host defense. Therefore, it is imperative to better understand the mechanisms underlying resolution to identify therapeutic targets. Antioxid. Redox Signal. 40, 292-316.
Collapse
Affiliation(s)
- Azuah L. Gonzalez
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Matthew M. Dungan
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - C. Duncan Smart
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Meena S. Madhur
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Amanda C. Doran
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| |
Collapse
|
17
|
Matter MA, Paneni F, Libby P, Frantz S, Stähli BE, Templin C, Mengozzi A, Wang YJ, Kündig TM, Räber L, Ruschitzka F, Matter CM. Inflammation in acute myocardial infarction: the good, the bad and the ugly. Eur Heart J 2024; 45:89-103. [PMID: 37587550 PMCID: PMC10771378 DOI: 10.1093/eurheartj/ehad486] [Citation(s) in RCA: 45] [Impact Index Per Article: 45.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 06/30/2023] [Accepted: 07/18/2023] [Indexed: 08/18/2023] Open
Abstract
Convergent experimental and clinical evidence have established the pathophysiological importance of pro-inflammatory pathways in coronary artery disease. Notably, the interest in treating inflammation in patients suffering acute myocardial infarction (AMI) is now expanding from its chronic aspects to the acute setting. Few large outcome trials have proven the benefits of anti-inflammatory therapies on cardiovascular outcomes by targeting the residual inflammatory risk (RIR), i.e. the smouldering ember of low-grade inflammation persisting in the late phase after AMI. However, these studies have also taught us about potential risks of anti-inflammatory therapy after AMI, particularly related to impaired host defence. Recently, numerous smaller-scale trials have addressed the concept of targeting a deleterious flare of excessive inflammation in the early phase after AMI. Targeting different pathways and implementing various treatment regimens, those trials have met with varied degrees of success. Promising results have come from those studies intervening early on the interleukin-1 and -6 pathways. Taking lessons from such past research may inform an optimized approach to target post-AMI inflammation, tailored to spare 'The Good' (repair and defence) while treating 'The Bad' (smouldering RIR) and capturing 'The Ugly' (flaming early burst of excess inflammation in the acute phase). Key constituents of such a strategy may read as follows: select patients with large pro-inflammatory burden (i.e. large AMI); initiate treatment early (e.g. ≤12 h post-AMI); implement a precisely targeted anti-inflammatory agent; follow through with a tapering treatment regimen. This approach warrants testing in rigorous clinical trials.
Collapse
Affiliation(s)
- Michael A Matter
- Department of Cardiology, University Heart Center, University Hospital Zurich, Rämistrasse 100, 8091 Zurich, Switzerland
- Center for Translational and Experimental Cardiology (CTEC), Department of Cardiology, Zurich University Hospital and University of Zurich, Rämistrasse 100, 8091 Zurich, Switzerland
| | - Francesco Paneni
- Department of Cardiology, University Heart Center, University Hospital Zurich, Rämistrasse 100, 8091 Zurich, Switzerland
- Center for Translational and Experimental Cardiology (CTEC), Department of Cardiology, Zurich University Hospital and University of Zurich, Rämistrasse 100, 8091 Zurich, Switzerland
| | - Peter Libby
- Division of Cardiovascular Medicine, Brigham and Women’s Hospital, 75 Francis Street, Boston, MA 02115, USA
- Harvard Medical School, 25 Shattuck Street, Boston, MA 02115, USA
| | - Stefan Frantz
- Department of Internal Medicine I, University Hospital Würzburg, Oberdürrbacher Strasse 6, 97080 Würzburg, Germany
| | - Barbara E Stähli
- Department of Cardiology, University Heart Center, University Hospital Zurich, Rämistrasse 100, 8091 Zurich, Switzerland
| | - Christian Templin
- Department of Cardiology, University Heart Center, University Hospital Zurich, Rämistrasse 100, 8091 Zurich, Switzerland
| | - Alessandro Mengozzi
- Center for Translational and Experimental Cardiology (CTEC), Department of Cardiology, Zurich University Hospital and University of Zurich, Rämistrasse 100, 8091 Zurich, Switzerland
- Department of Clinical and Experimental Medicine, University of Pisa, Via Roma 67, 56126 Pisa, Italy
| | - Yu-Jen Wang
- Center for Translational and Experimental Cardiology (CTEC), Department of Cardiology, Zurich University Hospital and University of Zurich, Rämistrasse 100, 8091 Zurich, Switzerland
| | - Thomas M Kündig
- Department of Dermatology, University Hospital Zurich, Rämistrasse 100, 8091 Zurich, Switzerland
| | - Lorenz Räber
- Department of Cardiology, Bern University Hospital, Inselspital, Freiburgstrasse 18, 3010 Bern, Switzerland
| | - Frank Ruschitzka
- Department of Cardiology, University Heart Center, University Hospital Zurich, Rämistrasse 100, 8091 Zurich, Switzerland
- Center for Translational and Experimental Cardiology (CTEC), Department of Cardiology, Zurich University Hospital and University of Zurich, Rämistrasse 100, 8091 Zurich, Switzerland
| | - Christian M Matter
- Department of Cardiology, University Heart Center, University Hospital Zurich, Rämistrasse 100, 8091 Zurich, Switzerland
- Center for Translational and Experimental Cardiology (CTEC), Department of Cardiology, Zurich University Hospital and University of Zurich, Rämistrasse 100, 8091 Zurich, Switzerland
| |
Collapse
|
18
|
He YC, Yuan GD, Li N, Ren MF, Qian-Zhang, Deng KN, Wang LC, Xiao WL, Ma N, Stamm C, Felthaus O, Prantl L, Nie J, Wang G. Recent advances in mesenchymal stem cell therapy for myocardial infarction. Clin Hemorheol Microcirc 2024; 87:383-398. [PMID: 38578884 DOI: 10.3233/ch-249101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/07/2024]
Abstract
Myocardial infarction refers to the ischemic necrosis of myocardium, characterized by a sharp reduction or interruption of blood flow in the coronary arteries due to the coronary artery occlusion, resulting in severe and prolonged ischemia in the corresponding myocardium and ultimately leading to ischemic necrosis of the myocardium. Given its high risk, it is considered as one of the most serious health threats today. In current clinical practice, multiple approaches have been explored to diminish myocardial oxygen consumption and alleviate symptoms, but notable success remains elusive. Accumulated clinical evidence has showed that the implantation of mesenchymal stem cell for treating myocardial infarction is both effective and safe. Nevertheless, there persists controversy and variability regarding the standardizing MSC transplantation protocols, optimizing dosage, and determining the most effective routes of administration. Addressing these remaining issues will pave the way of integration of MSCs as a feasible mainstream cardiac treatment.
Collapse
Affiliation(s)
- Yu-Chuan He
- Graduate School, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, China
| | - Guo-Dong Yuan
- Hebei Provincial Hospital of Chinese Medicine, Shijiazhuang, Hebei, China
| | - Nan Li
- Shijiazhuang Obstetrics and Gynecology Hospital, Shijiazhuang, Hebei, China
| | - Mei-Fang Ren
- Hebei Provincial Hospital of Chinese Medicine, Shijiazhuang, Hebei, China
| | - Qian-Zhang
- Hebei Provincial Hospital of Chinese Medicine, Shijiazhuang, Hebei, China
| | - Kai-Ning Deng
- Graduate School, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, China
| | - Le-Chuan Wang
- Graduate School, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, China
| | - Wei-Ling Xiao
- Graduate School, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, China
| | - Nan Ma
- Helmholtz-Zentrum Hereon, Institute of Active Polymers, Teltow, Germany
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany
| | | | - Oliver Felthaus
- Department of Plastic, Hand and Reconstructive Surgery, University Hospital Regensburg, Regensburg, Germany
| | - Lukas Prantl
- Department of Plastic, Hand and Reconstructive Surgery, University Hospital Regensburg, Regensburg, Germany
| | - Jia Nie
- Hebei Provincial Hospital of Chinese Medicine, Shijiazhuang, Hebei, China
| | - Gang Wang
- Hebei Provincial Hospital of Chinese Medicine, Shijiazhuang, Hebei, China
| |
Collapse
|
19
|
Liu Z, Sammani S, Barber CJ, Kempf CL, Li F, Yang Z, Bermudez RT, Camp SM, Herndon VR, Furenlid LR, Martin DR, Garcia JGN. An eNAMPT-neutralizing mAb reduces post-infarct myocardial fibrosis and left ventricular dysfunction. Biomed Pharmacother 2024; 170:116103. [PMID: 38160623 PMCID: PMC10872269 DOI: 10.1016/j.biopha.2023.116103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 12/21/2023] [Accepted: 12/26/2023] [Indexed: 01/03/2024] Open
Abstract
Myocardial infarction (MI) triggers adverse ventricular remodeling (VR), cardiac fibrosis, and subsequent heart failure. Extracellular nicotinamide phosphoribosyltransferase (eNAMPT) is postulated to play a significant role in VR processing via activation of the TLR4 inflammatory pathway. We hypothesized that an eNAMPT specific monoclonal antibody (mAb) could target and neutralize overexpressed eNAMPT post-MI and attenuate chronic cardiac inflammation and fibrosis. We investigated humanized ALT-100 and ALT-300 mAb with high eNAMPT-neutralizing capacity in an infarct rat model to test our hypothesis. ALT-300 was 99mTc-labeled to generate 99mTc-ALT-300 for imaging myocardial eNAMPT expression at 2 hours, 1 week, and 4 weeks post-IRI. The eNAMPT-neutralizing ALT-100 mAb (0.4 mg/kg) or saline was administered intraperitoneally at 1 hour and 24 hours post-reperfusion and twice a week for 4 weeks. Cardiac function changes were determined by echocardiography at 3 days and 4 weeks post-IRI. 99mTc-ALT-300 uptake was initially localized to the ischemic area at risk (IAR) of the left ventricle (LV) and subsequently extended to adjacent non-ischemic areas 2 hours to 4 weeks post-IRI. Radioactive uptake (%ID/g) of 99mTc-ALT-300 in the IAR increased from 1 week to 4 weeks (0.54 ± 0.16 vs. 0.78 ± 0.13, P < 0.01). Rats receiving ALT-100 mAb exhibited significantly improved myocardial histopathology and cardiac function at 4 weeks, with a significant reduction in the collagen volume fraction (%LV) compared to controls (21.5 ± 6.1% vs. 29.5 ± 9.9%, P < 0.05). Neutralization of the eNAMPT/TLR4 inflammatory cascade is a promising therapeutic strategy for MI by reducing chronic inflammation, fibrosis, and preserving cardiac function.
Collapse
Affiliation(s)
- Zhonglin Liu
- Translational Imaging Center, Houston Methodist Research Institute, Houston Methodist Hospital, Houston, TX, United States; Department of Medical Imaging, University of Arizona Health Sciences, Tucson, AZ, United States.
| | - Saad Sammani
- Department of Medicine, University of Arizona Health Sciences, Tucson, AZ, United States
| | - Christy J Barber
- Department of Medical Imaging, University of Arizona Health Sciences, Tucson, AZ, United States
| | - Carrie L Kempf
- University of Florida UF Scripps Research Institute, Jupiter, FL, United States
| | - Feng Li
- Translational Imaging Center, Houston Methodist Research Institute, Houston Methodist Hospital, Houston, TX, United States
| | - Zhen Yang
- Translational Imaging Center, Houston Methodist Research Institute, Houston Methodist Hospital, Houston, TX, United States
| | - Rosendo T Bermudez
- Department of Medicine, University of Arizona Health Sciences, Tucson, AZ, United States
| | - Sara M Camp
- University of Florida UF Scripps Research Institute, Jupiter, FL, United States
| | - Vivian Reyes Herndon
- Department of Medicine, University of Arizona Health Sciences, Tucson, AZ, United States
| | - Lars R Furenlid
- Department of Medical Imaging, University of Arizona Health Sciences, Tucson, AZ, United States
| | - Diego R Martin
- Translational Imaging Center, Houston Methodist Research Institute, Houston Methodist Hospital, Houston, TX, United States.
| | - Joe G N Garcia
- University of Florida UF Scripps Research Institute, Jupiter, FL, United States
| |
Collapse
|
20
|
Liu P, Yu X, Wang J, Wang L, Ding Y, Liu J. Establishment of pelvic inflammatory disease model induced by vaginal injection of Ureaplasma urealyticum liquids combined with fatigue and hunger. Anim Reprod 2023; 20:e20220106. [PMID: 38025994 PMCID: PMC10681126 DOI: 10.1590/1984-3143-ar2022-0106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2022] [Accepted: 09/12/2023] [Indexed: 12/01/2023] Open
Abstract
Pelvic inflammatory disease (PID) is an inflammation of the upper genital tract. PID is the leading cause of some severe sequelae in the absence of timely and accurate diagnosis and treatment. An appropriate animal model is needed to explore the underlying mechanism of PID sequelae. This study introduced an animal model of PID by vaginal injection of liquid Ureaplasma urealyticum combined with fatigue and hunger (UVF). This study was designed to test the feasibility of a rat model. A rat model was established using UVF irradiation. Levels of some inflammatory cytokines in the serum and the homogenates of the fallopian tubes were measured by ELISA, RT-PCR, and flow cytometry and compared with another rat model of Ureaplasma urealyticum liquids injected into the two uterus horns during laparotomy. Inflammatory alterations and adhesions were observed after hematoxylin and eosin (H&E) staining and detected using the Blauer scoring system. The results showed that the combined UVF and rat model caused apparent obstruction, edema, and adhesion in the fallopian tubes and connective tissues. The rat model showed upregulated CD4, CD8, and CD4/CD8 in peripheral blood mononuclear cells (PBMCs) and significantly increased levels of IL-4, IL-6, IL-10, and IL-17. UVF also enhanced the expression of tumor necrosis factor (TNF)-α, transforming growth factor (TGF)-β, vascular endothelial growth factor (VEGF) β, and matrix metalloproteinase (MMP)-2 (P<0.05). The UVF rat model can induce inflammatory alterations in the fallopian tubes and connective tissues, and can be used as a model of PID.
Collapse
Affiliation(s)
- Pengfei Liu
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xiao Yu
- Department of Gynecology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Jinjin Wang
- Department of Gynecology, The Second Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Li Wang
- Department of Gynecology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yi Ding
- Department of Gynecology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Jinxing Liu
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| |
Collapse
|
21
|
Shook PL, Singh M, Singh K. Macrophages in the Inflammatory Phase following Myocardial Infarction: Role of Exogenous Ubiquitin. BIOLOGY 2023; 12:1258. [PMID: 37759657 PMCID: PMC10526096 DOI: 10.3390/biology12091258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 09/19/2023] [Accepted: 09/19/2023] [Indexed: 09/29/2023]
Abstract
Cardiovascular disease (CVD) is one of the leading causes of death worldwide. One of the most common implications of CVD is myocardial infarction (MI). Following MI, the repair of the infarcted heart occurs through three distinct, yet overlapping phases of inflammation, proliferation, and maturation. Macrophages are essential to the resolution of the inflammatory phase due to their role in phagocytosis and efferocytosis. However, excessive and long-term macrophage accumulation at the area of injury and dysregulated function can induce adverse cardiac remodeling post-MI. Ubiquitin (UB) is a highly evolutionarily conserved small protein and is a normal constituent of plasma. Levels of UB are increased in the plasma during a variety of pathological conditions, including ischemic heart disease. Treatment of mice with UB associates with decreased inflammatory response and improved heart function following ischemia/reperfusion injury. This review summarizes the role of macrophages in the infarct healing process of the heart post-MI, and discusses the role of exogenous UB in myocardial remodeling post-MI and in the modulation of macrophage phenotype and function.
Collapse
Affiliation(s)
- Paige L. Shook
- Department of Biomedical Sciences, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, USA; (P.L.S.); (M.S.)
| | - Mahipal Singh
- Department of Biomedical Sciences, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, USA; (P.L.S.); (M.S.)
| | - Krishna Singh
- Department of Biomedical Sciences, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, USA; (P.L.S.); (M.S.)
- Center of Excellence in Inflammation, Infectious Disease and Immunity, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, USA
- James H. Quillen Veterans Affairs Medical Center, Mountain Home, TN 37684, USA
| |
Collapse
|
22
|
Learmonth M, Corker A, Dasgupta S, DeLeon-Pennell KY. Regulation of cardiac fibroblasts by lymphocytes after a myocardial infarction: playing in the major league. Am J Physiol Heart Circ Physiol 2023; 325:H553-H561. [PMID: 37450290 PMCID: PMC10538980 DOI: 10.1152/ajpheart.00250.2023] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Revised: 07/10/2023] [Accepted: 07/11/2023] [Indexed: 07/18/2023]
Abstract
Cardiac fibrosis is a pathological condition characterized by excessive accumulation of extracellular matrix components within the myocardium, which can lead to impaired cardiac function and heart failure. Studies have shown that lymphocytes including B and T cells play important roles in the development and progression of cardiac fibrosis after a myocardial infarction. In this review, we focus on the regulation of cardiac fibrosis by lymphocyte subsets, with a particular emphasis on CD4+ and CD8+ T cells and their effects on fibroblasts and cardiac remodeling. We also highlight areas for further exploration of the interactions between T cells and fibroblasts necessary for understanding and treating cardiac fibrosis and heart failure.
Collapse
Affiliation(s)
- Maya Learmonth
- College of Graduate Studies, Medical University of South Carolina, Charleston, South Carolina, United States
- Division of Cardiology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina, United States
| | - Alexa Corker
- College of Graduate Studies, Medical University of South Carolina, Charleston, South Carolina, United States
- Division of Cardiology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina, United States
| | - Shaoni Dasgupta
- College of Graduate Studies, Medical University of South Carolina, Charleston, South Carolina, United States
- Division of Cardiology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina, United States
| | - Kristine Y DeLeon-Pennell
- Division of Cardiology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina, United States
- Research Service, Ralph H. Johnson Veterans Affairs Medical Center, Charleston, South Carolina, United States
| |
Collapse
|
23
|
Sopova K, Tual-Chalot S, Mueller-Hennessen M, Vlachogiannis NI, Georgiopoulos G, Biener M, Sachse M, Turchinovich A, Polycarpou-Schwarz M, Spray L, Maneta E, Bennaceur K, Mohammad A, Richardson GD, Gatsiou A, Langer HF, Frey N, Stamatelopoulos K, Heineke J, Duerschmied D, Giannitsis E, Spyridopoulos I, Stellos K. Effector T cell chemokine IP-10 predicts cardiac recovery and clinical outcomes post-myocardial infarction. Front Immunol 2023; 14:1177467. [PMID: 37426649 PMCID: PMC10326041 DOI: 10.3389/fimmu.2023.1177467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Accepted: 05/23/2023] [Indexed: 07/11/2023] Open
Abstract
Background and aims Preclinical data suggest that activation of the adaptive immune system is critical for myocardial repair processes in acute myocardial infarction. The aim of the present study was to determine the clinical value of baseline effector T cell chemokine IP-10 blood levels in the acute phase of ST-segment elevation myocardial infarction (STEMI) for the prediction of the left ventricular function changes and cardiovascular outcomes after STEMI. Methods Serum IP-10 levels were retrospectively quantified in two independent cohorts of STEMI patients undergoing primary percutaneous coronary intervention. Results We report a biphasic response of the effector T cell trafficking chemokine IP-10 characterized by an initial increase of its serum levels in the acute phase of STEMI followed by a rapid reduction at 90min post reperfusion. Patients at the highest IP-10 tertile presented also with more CD4 effector memory T cells (CD4 TEM cells), but not other T cell subtypes, in blood. In the Newcastle cohort (n=47), patients in the highest IP-10 tertile or CD4 TEM cells at admission exhibited an improved cardiac systolic function 12 weeks after STEMI compared to patients in the lowest IP-10 tertile. In the Heidelberg cohort (n=331), STEMI patients were followed for a median of 540 days for major adverse cardiovascular events (MACE). Patients presenting with higher serum IP-10 levels at admission had a lower risk for MACE after adjustment for traditional risk factors, CRP and high-sensitivity troponin-T levels (highest vs. rest quarters: HR [95% CI]=0.420 [0.218-0.808]). Conclusion Increased serum levels of IP-10 in the acute phase of STEMI predict a better recovery in cardiac systolic function and less adverse events in patients after STEMI.
Collapse
Affiliation(s)
- Kateryna Sopova
- Translational and Clinical Research Institute, Vascular Biology and Medicine Theme, Faculty of Medical Sciences, Newcastle University, Newcastle Upon Tyne, United Kingdom
- Department of Cardiology, Angiology, Haemostaseology and Medical Intensive Care, University Medical Centre Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- Department of Cardiology, Royal Victoria Infirmary (RVI) and Freeman Hospitals, Newcastle Upon Tyne Hospitals National Health Service (NHS) Foundation Trust, Newcastle Upon Tyne, United Kingdom
- German Centre for Cardiovascular Research (DZHK), partner site Heidelberg/Mannheim, Mannheim, Germany
- Department of Cardiovascular Research, European Center for Angioscience (ECAS), Heidelberg University, Heidelberg/Mannheim, Germany
| | - Simon Tual-Chalot
- Biosciences Institute, Vascular Biology and Medicine Theme, Faculty of Medical Sciences, Newcastle University, Newcastle Upon Tyne, United Kingdom
| | - Matthias Mueller-Hennessen
- German Centre for Cardiovascular Research (DZHK), partner site Heidelberg/Mannheim, Mannheim, Germany
- Department of Internal Medicine III, Cardiology, University Hospital Heidelberg, Heidelberg, Germany
| | - Nikolaos I. Vlachogiannis
- Biosciences Institute, Vascular Biology and Medicine Theme, Faculty of Medical Sciences, Newcastle University, Newcastle Upon Tyne, United Kingdom
| | - Georgios Georgiopoulos
- Department of Clinical Therapeutics, Alexandra Hospital, National and Kapodistrian University of Athens School of Medicine, Athens, Greece
| | - Moritz Biener
- German Centre for Cardiovascular Research (DZHK), partner site Heidelberg/Mannheim, Mannheim, Germany
- Department of Internal Medicine III, Cardiology, University Hospital Heidelberg, Heidelberg, Germany
| | - Marco Sachse
- Department of Cardiovascular Research, European Center for Angioscience (ECAS), Heidelberg University, Heidelberg/Mannheim, Germany
| | - Andrey Turchinovich
- Department of Cardiovascular Research, European Center for Angioscience (ECAS), Heidelberg University, Heidelberg/Mannheim, Germany
| | - Maria Polycarpou-Schwarz
- Department of Cardiovascular Research, European Center for Angioscience (ECAS), Heidelberg University, Heidelberg/Mannheim, Germany
| | - Luke Spray
- Department of Cardiology, Royal Victoria Infirmary (RVI) and Freeman Hospitals, Newcastle Upon Tyne Hospitals National Health Service (NHS) Foundation Trust, Newcastle Upon Tyne, United Kingdom
- Biosciences Institute, Vascular Biology and Medicine Theme, Faculty of Medical Sciences, Newcastle University, Newcastle Upon Tyne, United Kingdom
| | - Eleni Maneta
- Department of Clinical Therapeutics, Alexandra Hospital, National and Kapodistrian University of Athens School of Medicine, Athens, Greece
| | - Karim Bennaceur
- Biosciences Institute, Vascular Biology and Medicine Theme, Faculty of Medical Sciences, Newcastle University, Newcastle Upon Tyne, United Kingdom
| | - Ashfaq Mohammad
- Department of Cardiology, Royal Victoria Infirmary (RVI) and Freeman Hospitals, Newcastle Upon Tyne Hospitals National Health Service (NHS) Foundation Trust, Newcastle Upon Tyne, United Kingdom
- Biosciences Institute, Vascular Biology and Medicine Theme, Faculty of Medical Sciences, Newcastle University, Newcastle Upon Tyne, United Kingdom
| | - Gavin David Richardson
- Biosciences Institute, Vascular Biology and Medicine Theme, Faculty of Medical Sciences, Newcastle University, Newcastle Upon Tyne, United Kingdom
| | - Aikaterini Gatsiou
- Biosciences Institute, Vascular Biology and Medicine Theme, Faculty of Medical Sciences, Newcastle University, Newcastle Upon Tyne, United Kingdom
| | - Harald F. Langer
- Department of Cardiology, Angiology, Haemostaseology and Medical Intensive Care, University Medical Centre Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- German Centre for Cardiovascular Research (DZHK), partner site Heidelberg/Mannheim, Mannheim, Germany
| | - Norbert Frey
- German Centre for Cardiovascular Research (DZHK), partner site Heidelberg/Mannheim, Mannheim, Germany
- Department of Internal Medicine III, Cardiology, University Hospital Heidelberg, Heidelberg, Germany
| | - Kimon Stamatelopoulos
- Biosciences Institute, Vascular Biology and Medicine Theme, Faculty of Medical Sciences, Newcastle University, Newcastle Upon Tyne, United Kingdom
- Department of Clinical Therapeutics, Alexandra Hospital, National and Kapodistrian University of Athens School of Medicine, Athens, Greece
| | - Joerg Heineke
- German Centre for Cardiovascular Research (DZHK), partner site Heidelberg/Mannheim, Mannheim, Germany
- Department of Cardiovascular Physiology, European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Daniel Duerschmied
- Department of Cardiology, Angiology, Haemostaseology and Medical Intensive Care, University Medical Centre Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- German Centre for Cardiovascular Research (DZHK), partner site Heidelberg/Mannheim, Mannheim, Germany
| | - Evangelos Giannitsis
- Department of Internal Medicine III, Cardiology, University Hospital Heidelberg, Heidelberg, Germany
| | - Ioakim Spyridopoulos
- Translational and Clinical Research Institute, Vascular Biology and Medicine Theme, Faculty of Medical Sciences, Newcastle University, Newcastle Upon Tyne, United Kingdom
- Department of Cardiology, Royal Victoria Infirmary (RVI) and Freeman Hospitals, Newcastle Upon Tyne Hospitals National Health Service (NHS) Foundation Trust, Newcastle Upon Tyne, United Kingdom
| | - Konstantinos Stellos
- Department of Cardiology, Angiology, Haemostaseology and Medical Intensive Care, University Medical Centre Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- German Centre for Cardiovascular Research (DZHK), partner site Heidelberg/Mannheim, Mannheim, Germany
- Department of Cardiovascular Research, European Center for Angioscience (ECAS), Heidelberg University, Heidelberg/Mannheim, Germany
- Biosciences Institute, Vascular Biology and Medicine Theme, Faculty of Medical Sciences, Newcastle University, Newcastle Upon Tyne, United Kingdom
- Department of Internal Medicine III, Cardiology, University Hospital Heidelberg, Heidelberg, Germany
- Department of Cardiovascular Physiology, European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| |
Collapse
|
24
|
Yap J, Irei J, Lozano-Gerona J, Vanapruks S, Bishop T, Boisvert WA. Macrophages in cardiac remodelling after myocardial infarction. Nat Rev Cardiol 2023; 20:373-385. [PMID: 36627513 DOI: 10.1038/s41569-022-00823-5] [Citation(s) in RCA: 75] [Impact Index Per Article: 37.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/25/2022] [Indexed: 01/12/2023]
Abstract
Myocardial infarction (MI), as a result of thrombosis or vascular occlusion, is the most prevalent cause of morbidity and mortality among all cardiovascular diseases. The devastating consequences of MI are compounded by the complexities of cellular functions involved in the initiation and resolution of early-onset inflammation and the longer-term effects related to scar formation. The resultant tissue damage can occur as early as 1 h after MI and activates inflammatory signalling pathways to elicit an immune response. Macrophages are one of the most active cell types during all stages after MI, including the cardioprotective, inflammatory and tissue repair phases. In this Review, we describe the phenotypes of cardiac macrophage involved in MI and their cardioprotective functions. A specific subset of macrophages called resident cardiac macrophages (RCMs) are derived from yolk sac progenitor cells and are maintained as a self-renewing population, although their numbers decrease with age. We explore sophisticated sequencing techniques that demonstrate the cardioprotective properties of this cardiac macrophage phenotype. Furthermore, we discuss the interactions between cardiac macrophages and other important cell types involved in the pathology and resolution of inflammation after MI. We summarize new and promising therapeutic approaches that target macrophage-mediated inflammation and the cardioprotective properties of RCMs after MI. Finally, we discuss future directions for the study of RCMs in MI and cardiovascular health in general.
Collapse
Affiliation(s)
- Jonathan Yap
- Center for Cardiovascular Research, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI, USA
| | - Jason Irei
- Center for Cardiovascular Research, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI, USA
| | - Javier Lozano-Gerona
- Center for Cardiovascular Research, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI, USA
| | - Selena Vanapruks
- Center for Cardiovascular Research, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI, USA
| | - Tianmai Bishop
- Center for Cardiovascular Research, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI, USA
| | - William A Boisvert
- Center for Cardiovascular Research, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI, USA.
| |
Collapse
|
25
|
Leancă SA, Afrăsânie I, Crișu D, Matei IT, Duca ȘT, Costache AD, Onofrei V, Tudorancea I, Mitu O, Bădescu MC, Șerban LI, Costache II. Cardiac Reverse Remodeling in Ischemic Heart Disease with Novel Therapies for Heart Failure with Reduced Ejection Fraction. Life (Basel) 2023; 13:1000. [PMID: 37109529 PMCID: PMC10143569 DOI: 10.3390/life13041000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 04/03/2023] [Accepted: 04/11/2023] [Indexed: 04/29/2023] Open
Abstract
Despite the improvements in the treatment of coronary artery disease (CAD) and acute myocardial infarction (MI) over the past 20 years, ischemic heart disease (IHD) continues to be the most common cause of heart failure (HF). In clinical trials, over 70% of patients diagnosed with HF had IHD as the underlying cause. Furthermore, IHD predicts a worse outcome for patients with HF, leading to a substantial increase in late morbidity, mortality, and healthcare costs. In recent years, new pharmacological therapies have emerged for the treatment of HF, such as sodium-glucose cotransporter-2 inhibitors, angiotensin receptor-neprilysin inhibitors, selective cardiac myosin activators, and oral soluble guanylate cyclase stimulators, demonstrating clear or potential benefits in patients with HF with reduced ejection fraction. Interventional strategies such as cardiac resynchronization therapy, cardiac contractility modulation, or baroreflex activation therapy might provide additional therapeutic benefits by improving symptoms and promoting reverse remodeling. Furthermore, cardiac regenerative therapies such as stem cell transplantation could become a new therapeutic resource in the management of HF. By analyzing the existing data from the literature, this review aims to evaluate the impact of new HF therapies in patients with IHD in order to gain further insight into the best form of therapeutic management for this large proportion of HF patients.
Collapse
Affiliation(s)
- Sabina Andreea Leancă
- Cardiology Clinic, “St. Spiridon” County Clinical Emergency Hospital, 700111 Iași, Romania
- Department of Internal Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iași, Romania
| | - Irina Afrăsânie
- Cardiology Clinic, “St. Spiridon” County Clinical Emergency Hospital, 700111 Iași, Romania
- Department of Internal Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iași, Romania
| | - Daniela Crișu
- Cardiology Clinic, “St. Spiridon” County Clinical Emergency Hospital, 700111 Iași, Romania
| | - Iulian Theodor Matei
- Cardiology Clinic, “St. Spiridon” County Clinical Emergency Hospital, 700111 Iași, Romania
- Department of Internal Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iași, Romania
| | - Ștefania Teodora Duca
- Cardiology Clinic, “St. Spiridon” County Clinical Emergency Hospital, 700111 Iași, Romania
- Department of Internal Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iași, Romania
| | - Alexandru Dan Costache
- Department of Internal Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iași, Romania
- Department of Cardiovascular Rehabilitation, Clinical Rehabilitation Hospital, 700661 Iași, Romania
| | - Viviana Onofrei
- Cardiology Clinic, “St. Spiridon” County Clinical Emergency Hospital, 700111 Iași, Romania
- Department of Internal Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iași, Romania
| | - Ionuţ Tudorancea
- Cardiology Clinic, “St. Spiridon” County Clinical Emergency Hospital, 700111 Iași, Romania
- Department of Physiology, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iași, Romania
| | - Ovidiu Mitu
- Cardiology Clinic, “St. Spiridon” County Clinical Emergency Hospital, 700111 Iași, Romania
- Department of Internal Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iași, Romania
| | - Minerva Codruța Bădescu
- Department of Internal Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iași, Romania
- Internal Medicine Clinic, “St. Spiridon” County Clinical Emergency Hospital, 700111 Iași, Romania
| | - Lăcrămioara Ionela Șerban
- Department of Physiology, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iași, Romania
| | - Irina Iuliana Costache
- Cardiology Clinic, “St. Spiridon” County Clinical Emergency Hospital, 700111 Iași, Romania
- Department of Internal Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iași, Romania
| |
Collapse
|
26
|
Huang B, Lin H, Zhang Q, Luo Y, Zhou B, Zhuo Z, Sha W, Wei J, Luo L, Zhang H, Chen K. Identification of shared fatty acid metabolism related signatures in dilated cardiomyopathy and myocardial infarction. Future Sci OA 2023; 9:FSO847. [PMID: 37056578 PMCID: PMC10088053 DOI: 10.2144/fsoa-2023-0008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Accepted: 03/13/2023] [Indexed: 03/30/2023] Open
Abstract
Aim: It is to be elucidated the risk-predictive role of differentially expressed fatty acid metabolism related genes (DE-FRGs) in dilated cardiomyopathy (DCM) and myocardial infarction. Materials & methods: Four gene enrichment analyses defined DE-FRGs’ biological functions and pathways. Three strategies were applied to identify risk biomarkers and construct a nomogram. The 4-DE-FRG correlation with immune cell infiltration, drugs, and ceRNA was explored. Results: DE-FRGs were enriched in lipid metabolism. A risk nomogram was established by ACSL1, ALDH2, CYP27A1 and PPARA, demonstrating a good ability for DCM and myocardial infarction prediction. PPARA was positively correlated with adaptive immunocytes. Thirty-five drugs are candidate therapeutic targets. Conclusion: A nomogram and new biological targets for early diagnosis and treatment of DCM and myocardial infarction were provided.
Collapse
|
27
|
Li R, Xiang C, Li Y, Nie Y. Targeting immunoregulation for cardiac regeneration. J Mol Cell Cardiol 2023; 177:1-8. [PMID: 36801268 DOI: 10.1016/j.yjmcc.2023.02.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 02/06/2023] [Accepted: 02/14/2023] [Indexed: 02/18/2023]
Abstract
Inducing endogenous cardiomyocyte proliferation and heart regeneration is a promising strategy to treat ischemic heart failure. The immune response has recently been considered critical in cardiac regeneration. Thus, targeting the immune response is a potent strategy to improve cardiac regeneration and repair after myocardial infarction. Here we reviewed the characteristics of the relationship between the postinjury immune response and heart regenerative capacity and summarized the latest studies focusing on inflammation and heart regeneration to identify potent targets of the immune response and strategies in the immune response to promote cardiac regeneration.
Collapse
Affiliation(s)
- Ruopu Li
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China
| | - Chenying Xiang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China
| | - Yixun Li
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China
| | - Yu Nie
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China; National Health Commission Key Laboratory of Cardiovascular Regenerative Medicine, Fuwai Central-China Hospital, Central China Branch of National Center for Cardiovascular Diseases, Zhengzhou 450046, China.
| |
Collapse
|
28
|
Li T, Yan Z, Fan Y, Fan X, Li A, Qi Z, Zhang J. Cardiac repair after myocardial infarction: A two-sided role of inflammation-mediated. Front Cardiovasc Med 2023; 9:1077290. [PMID: 36698953 PMCID: PMC9868426 DOI: 10.3389/fcvm.2022.1077290] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2022] [Accepted: 12/19/2022] [Indexed: 01/11/2023] Open
Abstract
Myocardial infarction is the leading cause of death and disability worldwide, and the development of new treatments can help reduce the size of myocardial infarction and prevent adverse cardiovascular events. Cardiac repair after myocardial infarction can effectively remove necrotic tissue, induce neovascularization, and ultimately replace granulation tissue. Cardiac inflammation is the primary determinant of whether beneficial cardiac repair occurs after myocardial infarction. Immune cells mediate inflammatory responses and play a dual role in injury and protection during cardiac repair. After myocardial infarction, genetic ablation or blocking of anti-inflammatory pathways is often harmful. However, enhancing endogenous anti-inflammatory pathways or blocking endogenous pro-inflammatory pathways may improve cardiac repair after myocardial infarction. A deficiency of neutrophils or monocytes does not improve overall cardiac function after myocardial infarction but worsens it and aggravates cardiac fibrosis. Several factors are critical in regulating inflammatory genes and immune cells' phenotypes, including DNA methylation, histone modifications, and non-coding RNAs. Therefore, strict control and timely suppression of the inflammatory response, finding a balance between inflammatory cells, preventing excessive tissue degradation, and avoiding infarct expansion can effectively reduce the occurrence of adverse cardiovascular events after myocardial infarction. This article reviews the involvement of neutrophils, monocytes, macrophages, and regulatory T cells in cardiac repair after myocardial infarction. After myocardial infarction, neutrophils are the first to be recruited to the damaged site to engulf necrotic cell debris and secrete chemokines that enhance monocyte recruitment. Monocytes then infiltrate the infarct site and differentiate into macrophages and they release proteases and cytokines that are harmful to surviving myocardial cells in the pre-infarct period. As time progresses, apoptotic neutrophils are cleared, the recruitment of anti-inflammatory monocyte subsets, the polarization of macrophages toward the repair phenotype, and infiltration of regulatory T cells, which secrete anti-inflammatory factors that stimulate angiogenesis and granulation tissue formation for cardiac repair. We also explored how epigenetic modifications regulate the phenotype of inflammatory genes and immune cells to promote cardiac repair after myocardial infarction. This paper also elucidates the roles of alarmin S100A8/A9, secreted frizzled-related protein 1, and podoplanin in the inflammatory response and cardiac repair after myocardial infarction.
Collapse
Affiliation(s)
- Tingting Li
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Zhipeng Yan
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yajie Fan
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Xinbiao Fan
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Aolin Li
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Zhongwen Qi
- Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China,*Correspondence: Zhongwen Qi,
| | - Junping Zhang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China,Junping Zhang,
| |
Collapse
|
29
|
Nian W, Huang Z, Fu C. Immune cells drive new immunomodulatory therapies for myocardial infarction: From basic to clinical translation. Front Immunol 2023; 14:1097295. [PMID: 36761726 PMCID: PMC9903069 DOI: 10.3389/fimmu.2023.1097295] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Accepted: 01/10/2023] [Indexed: 01/26/2023] Open
Abstract
The high incidence of heart failure secondary to myocardial infarction (MI) has been difficult to effectively address. MI causes strong aseptic inflammation, and infiltration of different immune cells and changes in the local inflammatory microenvironment play a key regulatory role in ventricular remodeling. Therefore, the possibility of improving the prognosis of MI through targeted immunity has been of interest and importance in MI. However, previously developed immune-targeted therapies have not achieved significant success in clinical trials. Here, we propose that the search for therapeutic targets from different immune cells may be more precise and lead to better clinical translation. Specifically, this review summarizes the role and potential therapeutic targets of various immune cells in ventricular remodeling after MI, especially monocytes/macrophages and neutrophils, as a way to demonstrate the importance and potential of immunomodulatory therapies for MI. In addition, we analyze the reasons for the failure of previous immunomodulatory therapies and the issues that need to be addressed, as well as the prospects and targeting strategies of using immune cells to drive novel immunomodulatory therapies, hoping to advance the development of immunomodulatory therapies by providing evidence and new ideas.
Collapse
Affiliation(s)
- Wenjian Nian
- Department of Clinical Medicine, Wannan Medical College, Wuhu, China
| | - Zijian Huang
- Department of Cardiology, Yi Ji Shan Hospital affiliated to Wannan Medical College, Wuhu, China.,Anesthesia Laboratory and Training Center, Wannan Medical College, Wuhu, China.,Key Laboratory of Non-coding RNA Transformation Research of Anhui Higher Education Institution, Wannan Medical College, Wuhu, China
| | - Cong Fu
- Department of Cardiology, Yi Ji Shan Hospital affiliated to Wannan Medical College, Wuhu, China.,Anesthesia Laboratory and Training Center, Wannan Medical College, Wuhu, China.,Key Laboratory of Non-coding RNA Transformation Research of Anhui Higher Education Institution, Wannan Medical College, Wuhu, China
| |
Collapse
|
30
|
Chalise U, Becirovic‐Agic M, Lindsey ML. The cardiac wound healing response to myocardial infarction. WIREs Mech Dis 2023; 15:e1584. [PMID: 36634913 PMCID: PMC10077990 DOI: 10.1002/wsbm.1584] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 03/31/2022] [Accepted: 05/18/2022] [Indexed: 01/14/2023]
Abstract
Myocardial infarction (MI) is defined as evidence of myocardial necrosis consistent with prolonged ischemia. In response to MI, the myocardium undergoes a series of wound healing events that initiate inflammation and shift to anti-inflammation before transitioning to tissue repair that culminates in scar formation to replace the region of the necrotic myocardium. The overall response to MI is determined by two major steps, the first of which is the secretion of proteases by infiltrating leukocytes to breakdown extracellular matrix (ECM) components, a necessary step to remove necrotic cardiomyocytes. The second step is the generation of new ECM that comprises the scar; and this step is governed by the cardiac fibroblasts as the major source of new ECM synthesis. The leukocyte component resides in the middle of the two-step process, contributing to both sides as the leukocytes transition from pro-inflammatory to anti-inflammatory and reparative cell phenotypes. The balance between the two steps determines the final quantity and quality of scar formed, which in turn contributes to chronic outcomes following MI, including the progression to heart failure. This review will summarize our current knowledge regarding the cardiac wound healing response to MI, primarily focused on experimental models of MI in mice. This article is categorized under: Cardiovascular Diseases > Molecular and Cellular Physiology Immune System Diseases > Molecular and Cellular Physiology.
Collapse
Affiliation(s)
- Upendra Chalise
- Department of Cellular and Integrative Physiology, Center for Heart and Vascular ResearchUniversity of Nebraska Medical CenterOmahaNebraskaUSA
- Research ServiceNebraska‐Western Iowa Health Care SystemOmahaNebraskaUSA
| | - Mediha Becirovic‐Agic
- Department of Cellular and Integrative Physiology, Center for Heart and Vascular ResearchUniversity of Nebraska Medical CenterOmahaNebraskaUSA
- Research ServiceNebraska‐Western Iowa Health Care SystemOmahaNebraskaUSA
| | - Merry L. Lindsey
- Department of Cellular and Integrative Physiology, Center for Heart and Vascular ResearchUniversity of Nebraska Medical CenterOmahaNebraskaUSA
- Research ServiceNebraska‐Western Iowa Health Care SystemOmahaNebraskaUSA
| |
Collapse
|
31
|
Wienecke LM, Leid JM, Leuschner F, Lavine KJ. Imaging Targets to Visualize the Cardiac Immune Landscape in Heart Failure. Circ Cardiovasc Imaging 2023; 16:e014071. [PMID: 36649453 PMCID: PMC9858350 DOI: 10.1161/circimaging.122.014071] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Heart failure involves a complex interplay between diverse populations of immune cells that dynamically shift across the natural history of disease. Within this context, the character of the immune response is a key determinant of clinical outcomes. Recent technological advances in single-cell transcriptomic, spatial, and proteomic technologies have fueled an explosion of new and clinically relevant insights into distinct immune cell populations that reside within the diseased heart including potential targets for molecular imaging and therapy. In this review, we will discuss the immune cell types and their respective functions with respect to myocardial infarction remodeling, dilated cardiomyopathy, and heart failure with preserved ejection fraction. In addition, we give a brief overview regarding myocarditis and cardiac sarcoidosis as inflammatory heart failure etiologies. We will highlight markers and cell populations as targets for molecular imaging to visualize inflammation and tissue healing and discuss clinical implications including the development and implementation of precision medicine approaches.
Collapse
Affiliation(s)
- Laura M. Wienecke
- Department of Cardiology, University Hospital Heidelberg, Heidelberg, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Heidelberg, Heidelberg, Germany
| | - Jamison M. Leid
- Cardiovascular Division, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Florian Leuschner
- Department of Cardiology, University Hospital Heidelberg, Heidelberg, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Heidelberg, Heidelberg, Germany
| | - Kory J. Lavine
- Cardiovascular Division, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, Missouri, USA
- Department of Developmental Biology, Washington University School of Medicine, Saint Louis, Missouri, USA
- Center for Regenerative Medicine, Washington University School of Medicine, Saint Louis, Missouri, USA
| |
Collapse
|
32
|
Inflammation in myocardial infarction: roles of mesenchymal stem cells and their secretome. Cell Death Dis 2022; 8:452. [DOI: 10.1038/s41420-022-01235-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 09/25/2022] [Accepted: 10/21/2022] [Indexed: 11/11/2022]
Abstract
AbstractInflammation plays crucial roles in the regulation of pathophysiological processes involved in injury, repair and remodeling of the infarcted heart; hence, it has become a promising target to improve the prognosis of myocardial infarction (MI). Mesenchymal stem cells (MSCs) serve as an effective and innovative treatment option for cardiac repair owing to their paracrine effects and immunomodulatory functions. In fact, transplanted MSCs have been shown to accumulate at injury sites of heart, exerting multiple effects including immunomodulation, regulating macrophages polarization, modulating the activation of T cells, NK cells and dendritic cells and alleviating pyroptosis of non-immune cells. Many studies also proved that preconditioning of MSCs can enhance their inflammation-regulatory effects. In this review, we provide an overview on the current understanding of the mechanisms on MSCs and their secretome regulating inflammation and immune cells after myocardial infarction and shed light on the applications of MSCs in the treatment of cardiac infarction.
Collapse
|
33
|
Broughton P, Troncoso M, Corker A, Williams A, Bolus D, Munoz G, McWhorter C, Roerden H, Huebsch P, DeLeon-Pennell KY. Riding the wave: a quantitative report of electrocardiogram utilization for myocardial infarction confirmation. Am J Physiol Heart Circ Physiol 2022; 323:H378-H387. [PMID: 35802516 PMCID: PMC9359650 DOI: 10.1152/ajpheart.00201.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 06/22/2022] [Accepted: 07/07/2022] [Indexed: 11/22/2022]
Abstract
The purpose of this study was to generate a quantitative profile of electrocardiograms (ECGs) for confirming surgical success of permanent coronary artery ligation. An ECG was recorded at baseline, and 0, 1, and 5 min after ligation and analyzed using iWorkx LabScribe software. Cohort 1 (C57Bl6/J, n = 8/sex) was enrolled to determine ECG characteristics that were confirmed in cohort 2 (C57Bl6/J, n = 6/sex; CD8-/-n = 6 males/4 females). Of the 16 mice in cohort 1, 12 (6/sex) had an infarct ≥35% and four mice (2/sex) had <35% based on 2,3,5-triphenyltetrazolium chloride staining. After ligation, the QRS complex and R-S amplitude were significantly different compared with baseline. No differences were observed in the R-S amplitude between mice with infarcts ≥35% versus <35% at any time point, whereas the QRS complex was significant 1 min after ligation. Receiver operating characteristic (ROC) curve linked changes in the QRS complex but not the R-S amplitude at 1 and 5 min with surgical success. Data were normalized to baseline values to calculate fold change. ROC analysis of the normalized QRS data indicated strong sensitivity and specificity for infarcts ≥35%; normalized R-S amplitude remained nonsignificant. With a cutoff generated by ROC analysis of cohort 1 (>80% sensitivity; >90% specificity), the non-normalized QRS complex of cohort 2 had an 86% success rate (2 false positives; 1 false negative). The normalized data had a 77% success rate (2 false positives; 3 false negatives). Neither sex nor genotype was associated with false predictions (P = 0.18). Our data indicate that the area under the QRS complex 1 min after ligation can improve reproducibility in MI surgeries.NEW & NOTEWORTHY Our study describes a quantitative method for using an electrocardiogram (ECG) to determine which animals have infarcts that reflect coronary artery ligation. Using a quantitative ECG, investigators will have the benefit of having real-time feedback during the procedure, which will ultimately decrease the amount of time investigators spend performing experiments. This overall increase in efficiency will help investigators decrease animal numbers used due to better surgical outcomes.
Collapse
Affiliation(s)
- Philip Broughton
- Division of Cardiology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina
| | - Miguel Troncoso
- Research Service, Ralph H. Johnson Veterans Affairs Medical Center, Charleston, South Carolina
| | - Alexa Corker
- Division of Cardiology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina
| | - Alexus Williams
- Division of Cardiology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina
| | - Dawson Bolus
- College of Medicine, Medical University of South Carolina, Charleston, South Carolina
| | - Gualberto Munoz
- College of Medicine, Medical University of South Carolina, Charleston, South Carolina
| | - Caroline McWhorter
- College of Medicine, Medical University of South Carolina, Charleston, South Carolina
| | - Hallie Roerden
- College of Medicine, Medical University of South Carolina, Charleston, South Carolina
| | - Penny Huebsch
- College of Medicine, Medical University of South Carolina, Charleston, South Carolina
| | - Kristine Y DeLeon-Pennell
- Division of Cardiology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina
- Research Service, Ralph H. Johnson Veterans Affairs Medical Center, Charleston, South Carolina
| |
Collapse
|
34
|
Sai X, Li Z, Deng G, Wang L, Xiaowu W, Nasser MI, Liu C, Zhu P. Immunomodulatory effects of icariin in a myocardial infarction mouse model. Bioengineered 2022; 13:12504-12515. [PMID: 35579292 PMCID: PMC9276034 DOI: 10.1080/21655979.2022.2076453] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Myocardial infarction (MI) is a prevalent cardiovascular disease defined by myocardial ischemia and hypoxic damage caused by plaque rupture, thrombosis, lumen stenosis, or blockage in the coronary artery. However, the development of emergency percutaneous coronary interventional therapy has enabled the rapid restoration of blood perfusion to ischemic myocardium and the rescue of dying myocardium cells. Some dying myocardium cells have caused irreversible damage and impaired cardiac function recovery in recent years. Icariin has been utilized to treat various ailments as a natural chemical extract. In this study, we employed a variety of approaches to observe MI, including western blotting, quantitative RT–PCR, immunohistochemistry, and flow cytometric analysis using icariin. As demonstrated by the research findings, icariin may prevent MI-induced cell apoptosis. This is accomplished by inhibiting proinflammatory factors via the Nrf2/HO-1 signaling pathways. These data imply that icariin may be an effective treatment for MI.
Collapse
Affiliation(s)
- Xiyalatu Sai
- Departement of Cardiology, The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China.,Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China.,Affiliated Hospital of Inner Mongolia University for the Nationalities, Tongliao City, People's Republic of China
| | - Zhetao Li
- Departement of Cardiology, The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China.,Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Gang Deng
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Lu Wang
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Wang Xiaowu
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Moussa Ide Nasser
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Chi Liu
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China.,Department of Nephrology, Sichuan Academy of Medical Science and Sichuan Provincial People's Hospital, Sichuan Renal Disease Clinical Research Center, University of Electronic Science and Technology of China, Chengdu, China.,Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu, China
| | - Ping Zhu
- Departement of Cardiology, The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China.,Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| |
Collapse
|
35
|
Abstract
The immune system is fundamental to tissue homeostasis and is the first line of defense following infection, injury or disease. In the damaged heart, large numbers of immune cells are recruited to the site of injury. These cells play an integral part in both repair by scar formation and the initiation of tissue regeneration. They initially assume inflammatory phenotypes, releasing pro-inflammatory cytokines and removing dead and dying tissue, before entering a reparative stage, replacing dead muscle tissue with a non-contractile scar. In this Review, we present an overview of the innate and adaptive immune response to heart injury. We explore the kinetics of immune cell mobilization following cardiac injury and how the different innate and adaptive immune cells interact with one another and with the damaged tissue. We draw on key findings from regenerative models, providing insight into how to support a robust immune response permissible for cardiac regeneration. Finally, we consider how the latest technological developments can offer opportunities for a deeper and unbiased functional understanding of the immune response to heart disease, highlighting the importance of such knowledge as the basis for promoting regeneration following cardiac injury in human patients.
Collapse
Affiliation(s)
- Filipa C. Simões
- MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford,Oxford, OxfordshireOX3 9DS, UK
- Institute of Developmental and Regenerative Medicine, Old Road Campus, Oxford, OxfordshireOX3 7DQ, UK
| | - Paul R. Riley
- Institute of Developmental and Regenerative Medicine, Old Road Campus, Oxford, OxfordshireOX3 7DQ, UK
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, OxfordshireOX1 3PT, UK
| |
Collapse
|
36
|
Ma Y, Yang X, Villalba N, Chatterjee V, Reynolds A, Spence S, Wu MH, Yuan SY. Circulating lymphocyte trafficking to the bone marrow contributes to lymphopenia in myocardial infarction. Am J Physiol Heart Circ Physiol 2022; 322:H622-H635. [PMID: 35179978 PMCID: PMC8934671 DOI: 10.1152/ajpheart.00003.2022] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 02/14/2022] [Accepted: 02/14/2022] [Indexed: 11/22/2022]
Abstract
Some patients with myocardial infarction (MI) exhibit lymphopenia, a reduction in blood lymphocyte count. Moreover, lymphopenia inversely correlates with patient prognosis. The objective of this study was to elucidate the underlying mechanisms that cause lymphopenia after MI. Multiparameter flow cytometric analysis demonstrated that MI induced profound B and T lymphopenia in a mouse model, peaking at day 1 post-MI. The finding that non-MI control and MI mice exhibited similar apoptotic rate for blood B and T lymphocytes argues against apoptosis being essential for MI-induced lymphopenia. Interestingly, the bone marrow in day 1 post-MI mice contained more B and T cells but showed less B- and T-cell proliferation compared with day 0 controls. This suggests that blood lymphocytes may travel to the bone marrow after MI. This was confirmed by adoptive transfer experiments demonstrating that MI caused the loss of transferred lymphocytes in the blood, but the accumulation of transferred lymphocytes in the bone marrow. To elucidate the underlying signaling pathways, β2-adrenergic receptor or sphingosine-1-phosphate receptor type 1 (S1PR1) was pharmacologically blocked, respectively. β2-receptor inhibition had no significant effect on blood lymphocyte count, whereas S1PR1 blockade aggravated lymphopenia in MI mice. Furthermore, we discovered that MI-induced glucocorticoid release triggered lymphopenia. This was supported by the findings that adrenalectomy (ADX) completely prevented mice from MI-induced lymphopenia, and supplementation with corticosterone in adrenalectomized MI mice reinduced lymphopenia. In conclusion, our study demonstrates that MI-associated lymphopenia involves lymphocyte redistribution from peripheral blood to the bone marrow, which is mediated by glucocorticoids.NEW & NOTEWORTHY Lymphopenia, a reduction in blood lymphocyte count, is known to inversely correlate with the prognosis for patients with myocardial infarction (MI). However, the underlying mechanisms by which cardiac ischemia induces lymphopenia remain elusive. This study provides the first evidence that MI activates the hypothalamic-pituitary-adrenal (HPA) axis to increase glucocorticoid secretion, and elevated circulating glucocorticoids induce blood lymphocytes trafficking to the bone marrow, leading to lymphopenia.
Collapse
Affiliation(s)
- Yonggang Ma
- Department of Molecular Pharmacology and Physiology, University of South Florida Morsani College of Medicine, Tampa, Florida
| | - Xiaoyuan Yang
- Department of Molecular Pharmacology and Physiology, University of South Florida Morsani College of Medicine, Tampa, Florida
| | - Nuria Villalba
- Department of Molecular Pharmacology and Physiology, University of South Florida Morsani College of Medicine, Tampa, Florida
| | - Victor Chatterjee
- Department of Molecular Pharmacology and Physiology, University of South Florida Morsani College of Medicine, Tampa, Florida
| | - Amanda Reynolds
- Department of Molecular Pharmacology and Physiology, University of South Florida Morsani College of Medicine, Tampa, Florida
| | - Sam Spence
- Department of Molecular Pharmacology and Physiology, University of South Florida Morsani College of Medicine, Tampa, Florida
| | - Mack H Wu
- Department of Surgery, University of South Florida Morsani College of Medicine, Tampa, Florida
| | - Sarah Y Yuan
- Department of Molecular Pharmacology and Physiology, University of South Florida Morsani College of Medicine, Tampa, Florida
- Department of Surgery, University of South Florida Morsani College of Medicine, Tampa, Florida
| |
Collapse
|
37
|
Peterson EA, Sun J, Wang J. Leukocyte-Mediated Cardiac Repair after Myocardial Infarction in Non-Regenerative vs. Regenerative Systems. J Cardiovasc Dev Dis 2022; 9:63. [PMID: 35200716 PMCID: PMC8877434 DOI: 10.3390/jcdd9020063] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 02/17/2022] [Accepted: 02/18/2022] [Indexed: 02/01/2023] Open
Abstract
Innate and adaptive leukocytes rapidly mobilize to ischemic tissues after myocardial infarction in response to damage signals released from necrotic cells. Leukocytes play important roles in cardiac repair and regeneration such as inflammation initiation and resolution; the removal of dead cells and debris; the deposition of the extracellular matrix and granulation tissue; supporting angiogenesis and cardiomyocyte proliferation; and fibrotic scar generation and resolution. By organizing and comparing the present knowledge of leukocyte recruitment and function after cardiac injury in non-regenerative to regenerative systems, we propose that the leukocyte response to cardiac injury differs in non-regenerative adult mammals such as humans and mice in comparison to cardiac regenerative models such as neonatal mice and adult zebrafish. Specifically, extensive neutrophil, macrophage, and T-cell persistence contributes to a lengthy inflammatory period in non-regenerative systems for adverse cardiac remodeling and heart failure development, whereas their quick removal supports inflammation resolution in regenerative systems for new contractile tissue formation and coronary revascularization. Surprisingly, other leukocytes have not been examined in regenerative model systems. With this review, we aim to encourage the development of improved immune cell markers and tools in cardiac regenerative models for the identification of new immune targets in non-regenerative systems to develop new therapies.
Collapse
Affiliation(s)
| | | | - Jinhu Wang
- Division of Cardiology, School of Medicine, Emory University, Atlanta, GA 30322, USA; (E.A.P.); (J.S.)
| |
Collapse
|
38
|
Corker A, Neff LS, Broughton P, Bradshaw AD, DeLeon-Pennell KY. Organized Chaos: Deciphering Immune Cell Heterogeneity's Role in Inflammation in the Heart. Biomolecules 2021; 12:11. [PMID: 35053159 PMCID: PMC8773626 DOI: 10.3390/biom12010011] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 12/10/2021] [Accepted: 12/18/2021] [Indexed: 12/24/2022] Open
Abstract
During homeostasis, immune cells perform daily housekeeping functions to maintain heart health by acting as sentinels for tissue damage and foreign particles. Resident immune cells compose 5% of the cellular population in healthy human ventricular tissue. In response to injury, there is an increase in inflammation within the heart due to the influx of immune cells. Some of the most common immune cells recruited to the heart are macrophages, dendritic cells, neutrophils, and T-cells. In this review, we will discuss what is known about cardiac immune cell heterogeneity during homeostasis, how these cell populations change in response to a pathology such as myocardial infarction or pressure overload, and what stimuli are regulating these processes. In addition, we will summarize technologies used to evaluate cell heterogeneity in models of cardiovascular disease.
Collapse
Affiliation(s)
- Alexa Corker
- Department of Medicine, Division of Cardiology, Medical University of South Carolina, Charleston, SC 29425, USA; (A.C.); (L.S.N.); (P.B.); (A.D.B.)
| | - Lily S. Neff
- Department of Medicine, Division of Cardiology, Medical University of South Carolina, Charleston, SC 29425, USA; (A.C.); (L.S.N.); (P.B.); (A.D.B.)
| | - Philip Broughton
- Department of Medicine, Division of Cardiology, Medical University of South Carolina, Charleston, SC 29425, USA; (A.C.); (L.S.N.); (P.B.); (A.D.B.)
| | - Amy D. Bradshaw
- Department of Medicine, Division of Cardiology, Medical University of South Carolina, Charleston, SC 29425, USA; (A.C.); (L.S.N.); (P.B.); (A.D.B.)
- Ralph H. Johnson Veterans Affairs Medical Center, Medical University of South Carolina, Charleston, SC 29401, USA
| | - Kristine Y. DeLeon-Pennell
- Department of Medicine, Division of Cardiology, Medical University of South Carolina, Charleston, SC 29425, USA; (A.C.); (L.S.N.); (P.B.); (A.D.B.)
- Ralph H. Johnson Veterans Affairs Medical Center, Medical University of South Carolina, Charleston, SC 29401, USA
| |
Collapse
|
39
|
Lindsey ML, Brunt KR, Kirk JA, Kleinbongard P, Calvert JW, de Castro Brás LE, DeLeon-Pennell KY, Del Re DP, Frangogiannis NG, Frantz S, Gumina RJ, Halade GV, Jones SP, Ritchie RH, Spinale FG, Thorp EB, Ripplinger CM, Kassiri Z. Guidelines for in vivo mouse models of myocardial infarction. Am J Physiol Heart Circ Physiol 2021; 321:H1056-H1073. [PMID: 34623181 PMCID: PMC8834230 DOI: 10.1152/ajpheart.00459.2021] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Revised: 10/05/2021] [Accepted: 10/05/2021] [Indexed: 12/11/2022]
Abstract
Despite significant improvements in reperfusion strategies, acute coronary syndromes all too often culminate in a myocardial infarction (MI). The consequent MI can, in turn, lead to remodeling of the left ventricle (LV), the development of LV dysfunction, and ultimately progression to heart failure (HF). Accordingly, an improved understanding of the underlying mechanisms of MI remodeling and progression to HF is necessary. One common approach to examine MI pathology is with murine models that recapitulate components of the clinical context of acute coronary syndrome and subsequent MI. We evaluated the different approaches used to produce MI in mouse models and identified opportunities to consolidate methods, recognizing that reperfused and nonreperfused MI yield different responses. The overall goal in compiling this consensus statement is to unify best practices regarding mouse MI models to improve interpretation and allow comparative examination across studies and laboratories. These guidelines will help to establish rigor and reproducibility and provide increased potential for clinical translation.
Collapse
Affiliation(s)
- Merry L Lindsey
- Department of Cellular and Integrative Physiology, Center for Heart and Vascular Research, University of Nebraska Medical Center, Omaha, Nebraska
- Research Service, Nebraska-Western Iowa Health Care System, Omaha, Nebraska
| | - Keith R Brunt
- Department of Pharmacology, Faculty of Medicine, Dalhousie University, Saint John, New Brunswick, Canada
| | - Jonathan A Kirk
- Department of Cell and Molecular Physiology, Loyola University Chicago Stritch School of Medicine, Chicago, Illinois
| | - Petra Kleinbongard
- Institute for Pathophysiology, West German Heart and Vascular Center, University of Essen Medical School, Essen, Germany
| | - John W Calvert
- Carlyle Fraser Heart Center of Emory University Hospital Midtown, Atlanta, Georgia
- Division of Cardiothoracic Surgery, Department of Surgery, Emory University School of Medicine, Atlanta, Georgia
| | - Lisandra E de Castro Brás
- Department of Physiology, The Brody School of Medicine, East Carolina University, Greenville, North Carolina
| | - Kristine Y DeLeon-Pennell
- Division of Cardiology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina
- Research Service, Ralph H. Johnson Veterans Affairs Medical Center, Charleston, South Carolina
| | - Dominic P Del Re
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark, New Jersey
| | - Nikolaos G Frangogiannis
- Division of Cardiology, Department of Medicine, The Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, Bronx, New York
| | - Stefan Frantz
- Department of Internal Medicine I, University Hospital Würzburg, Würzburg, Germany
| | - Richard J Gumina
- Division of Cardiovascular Medicine, Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, Ohio
- Department of Physiology and Cell Biology, The Ohio State University Wexner Medical Center, Columbus, Ohio
- The Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, Ohio
| | - Ganesh V Halade
- Division of Cardiovascular Sciences, Department of Medicine, University of South Florida, Tampa, Florida
| | - Steven P Jones
- Department of Medicine, Diabetes and Obesity Center, University of Louisville, Louisville, Kentucky
| | - Rebecca H Ritchie
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University (Parkville Campus), Victoria, Australia
| | - Francis G Spinale
- Cardiovascular Translational Research Center, University of South Carolina School of Medicine and the Columbia Veteran Affairs Medical Center, Columbia, South Carolina
| | - Edward B Thorp
- Department of Pathology and Feinberg Cardiovascular and Renal Research Institute, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Crystal M Ripplinger
- Department of Pharmacology, University of California Davis School of Medicine, Davis, California
| | - Zamaneh Kassiri
- Department of Physiology, Cardiovascular Research Center, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
40
|
Zaidi Y, Corker A, Vasileva VY, Oviedo K, Graham C, Wilson K, Martino J, Troncoso M, Broughton P, Ilatovskaya DV, Lindsey ML, DeLeon-Pennell KY. Chronic Porphyromonas gingivalis lipopolysaccharide induces adverse myocardial infarction wound healing through activation of CD8 + T cells. Am J Physiol Heart Circ Physiol 2021; 321:H948-H962. [PMID: 34597184 PMCID: PMC8616607 DOI: 10.1152/ajpheart.00082.2021] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 09/08/2021] [Accepted: 09/27/2021] [Indexed: 02/06/2023]
Abstract
Oral and gum health have long been associated with incidence and outcomes of cardiovascular disease. Periodontal disease increases myocardial infarction (MI) mortality by sevenfold through mechanisms that are not fully understood. The goal of this study was to evaluate whether lipopolysaccharide (LPS) from a periodontal pathogen accelerates inflammation after MI through memory T-cell activation. We compared four groups [no MI, chronic LPS, day 1 after MI, and day 1 after MI with chronic LPS (LPS + MI); n = 68 mice] using the mouse heart attack research tool 1.0 database and tissue bank coupled with new analyses and experiments. LPS + MI increased total CD8+ T cells in the left ventricle versus the other groups (P < 0.05 vs. all). Memory CD8+ T cells (CD44 + CD27+) were 10-fold greater in LPS + MI than in MI alone (P = 0.02). Interleukin (IL)-4 stimulated splenic CD8+ T cells away from an effector phenotype and toward a memory phenotype, inducing secretion of factors associated with the Wnt/β-catenin signaling that promoted monocyte migration and decreased viability. To dissect the effect of CD8+ T cells after MI, we administered a major histocompatibility complex-I-blocking antibody starting 7 days before MI, which prevented effector CD8+ T-cell activation without affecting the memory response. The reduction in effector cells diminished infarct wall thinning but had no effect on macrophage numbers or MertK expression. LPS + MI + IgG attenuated macrophages within the infarct without effecting CD8+ T cells, suggesting these two processes were independent. Overall, our data indicate that effector and memory CD8+ T cells at post-MI day 1 are amplified by chronic LPS to potentially promote infarct wall thinning.NEW & NOTEWORTHY Although there is a well-documented link between periodontal disease and heart health, the mechanisms are unclear. Our study indicates that in response to circulating periodontal endotoxins, memory CD8+ T cells are activated, resulting in an acceleration of macrophage-mediated inflammation after MI. Blocking activation of effector CD8+ T cells had no effect on the macrophage numbers or wall thinning at post-MI day 1, indicating that this response was likely due in part to memory CD8+ T cells.
Collapse
Affiliation(s)
- Yusra Zaidi
- Division of Cardiology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina
| | - Alexa Corker
- Division of Cardiology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina
| | - Valeriia Y Vasileva
- Division of Nephrology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina
| | - Kimberly Oviedo
- Division of Cardiology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina
| | - Connor Graham
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, South Carolina
| | - Kyrie Wilson
- Department of Ophthalmology, Medical University of South Carolina, Charleston, South Carolina
| | - John Martino
- Division of Cardiology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina
| | - Miguel Troncoso
- Division of Cardiology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina
| | - Philip Broughton
- Division of Cardiology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina
| | - Daria V Ilatovskaya
- Division of Nephrology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, Georgia
| | - Merry L Lindsey
- Department of Cellular and Integrative Physiology, Center for Heart and Vascular Research, University of Nebraska Medical Center, Omaha, Nebraska
- Research Service, Nebraska-Western Iowa Health Care System, Omaha, Nebraska
| | - Kristine Y DeLeon-Pennell
- Division of Cardiology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina
- Research Service, Ralph H. Johnson Veterans Affairs Medical Center, Charleston, South Carolina
| |
Collapse
|
41
|
Ravaud C, Ved N, Jackson DG, Vieira JM, Riley PR. Lymphatic Clearance of Immune Cells in Cardiovascular Disease. Cells 2021; 10:cells10102594. [PMID: 34685572 PMCID: PMC8533855 DOI: 10.3390/cells10102594] [Citation(s) in RCA: 7] [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: 08/25/2021] [Revised: 09/26/2021] [Accepted: 09/27/2021] [Indexed: 12/11/2022] Open
Abstract
Recent advances in our understanding of the lymphatic system, its function, development, and role in pathophysiology have changed our views on its importance. Historically thought to be solely involved in the transport of tissue fluid, lipids, and immune cells, the lymphatic system displays great heterogeneity and plasticity and is actively involved in immune cell regulation. Interference in any of these processes can be deleterious, both at the developmental and adult level. Preclinical studies into the cardiac lymphatic system have shown that invoking lymphangiogenesis and enhancing immune cell trafficking in ischaemic hearts can reduce myocardial oedema, reduce inflammation, and improve cardiac outcome. Understanding how immune cells and the lymphatic endothelium interact is also vital to understanding how the lymphatic vascular network can be manipulated to improve immune cell clearance. In this Review, we examine the different types of immune cells involved in fibrotic repair following myocardial infarction. We also discuss the development and function of the cardiac lymphatic vasculature and how some immune cells interact with the lymphatic endothelium in the heart. Finally, we establish how promoting lymphangiogenesis is now a prime therapeutic target for reducing immune cell persistence, inflammation, and oedema to restore heart function in ischaemic heart disease.
Collapse
Affiliation(s)
- Christophe Ravaud
- Burdon-Sanderson Cardiac Science Centre, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3PT, UK; (C.R.); (N.V.); (J.M.V.)
| | - Nikita Ved
- Burdon-Sanderson Cardiac Science Centre, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3PT, UK; (C.R.); (N.V.); (J.M.V.)
| | - David G. Jackson
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, University of Oxford, Oxford OX3 9DS, UK;
| | - Joaquim Miguel Vieira
- Burdon-Sanderson Cardiac Science Centre, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3PT, UK; (C.R.); (N.V.); (J.M.V.)
| | - Paul R. Riley
- Burdon-Sanderson Cardiac Science Centre, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3PT, UK; (C.R.); (N.V.); (J.M.V.)
- Correspondence:
| |
Collapse
|
42
|
Wang J, Xu T, Xu M. Roles and Mechanisms of TGR5 in the Modulation of CD4 + T Cell Functions in Myocardial Infarction. J Cardiovasc Transl Res 2021; 15:350-359. [PMID: 34402028 DOI: 10.1007/s12265-021-10164-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Accepted: 08/02/2021] [Indexed: 10/20/2022]
Abstract
Bile acid receptor TGR5 has been proved to play protective roles in the process of myocardial infarction (MI). Recently, we found spleen weight of Tgr5+/+ mice was increased at 7-day post-MI but not in Tgr5-/- mice. Since the spleen is one of the main resources of immune and inflammatory cells post-MI, we conducted flow cytometry analysis of multiple immune cells in the heart post-MI. It showed the recruitment of CD4+ T cells and CD8+ T cells was continuously more in the heart of Tgr5-/- mice post-MI until 7 days after MI. Furthermore, CD4-specific TGR5 depletion mice exhibited aggravated ischemic injury. The mRNA expressions of the markers of Th1 and Treg were upregulated in the heart of Tgr5-/- mice at 7-day post-MI. These results suggested TGR5 modulates CD4+ T cell functions and subsets distribution in the heart, and plays protective roles in myocardial infarction.
Collapse
Affiliation(s)
- Jiaxing Wang
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, Beijing, 100191, China.,NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptide, Peking University Third Hospital, Beijing, 100191, China.,Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Peking University Third Hospital, Beijing, 100191, China.,Beijing Key Laboratory of Cardiovascular Receptors Research, Peking University Third Hospital, Beijing, 100191, China
| | - Tan Xu
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, Beijing, 100191, China.,NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptide, Peking University Third Hospital, Beijing, 100191, China.,Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Peking University Third Hospital, Beijing, 100191, China.,Beijing Key Laboratory of Cardiovascular Receptors Research, Peking University Third Hospital, Beijing, 100191, China
| | - Ming Xu
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, Beijing, 100191, China. .,NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptide, Peking University Third Hospital, Beijing, 100191, China. .,Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Peking University Third Hospital, Beijing, 100191, China. .,Beijing Key Laboratory of Cardiovascular Receptors Research, Peking University Third Hospital, Beijing, 100191, China. .,State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China.
| |
Collapse
|
43
|
Wu CL, Yin R, Wang SN, Ying R. A Review of CXCL1 in Cardiac Fibrosis. Front Cardiovasc Med 2021; 8:674498. [PMID: 33996954 PMCID: PMC8113392 DOI: 10.3389/fcvm.2021.674498] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Accepted: 04/01/2021] [Indexed: 12/31/2022] Open
Abstract
Chemokine C-X-C motif ligand-1 (CXCL1), principally expressed in neutrophils, macrophages and epithelial cells, is a valid pro-inflammatory factor which performs an important role in mediating the infiltration of neutrophils and monocytes/macrophages. Elevated serum level of CXCL1 is considered a pro-inflammatory reaction by the organism. CXCL1 is also related to diverse organs fibrosis according to relevant studies. A growing body of evidence suggests that CXCL1 promotes the process of cardiac remodeling and fibrosis. Here, we review structure and physiological functions of CXCL1 and recent progress on the effects and mechanisms of CXCL1 in cardiac fibrosis. In addition, we explore the role of CXCL1 in the fibrosis of other organs. Besides, we probe the possibility that CXCL1 can be a therapeutic target for the treatment of cardiac fibrosis in cardiovascular diseases.
Collapse
Affiliation(s)
- Cheng-Long Wu
- Department of Cardiology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Ran Yin
- Department of Cardiology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Su-Nan Wang
- Department of Cardiology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Ru Ying
- Department of Cardiology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| |
Collapse
|
44
|
Kologrivova I, Shtatolkina M, Suslova T, Ryabov V. Cells of the Immune System in Cardiac Remodeling: Main Players in Resolution of Inflammation and Repair After Myocardial Infarction. Front Immunol 2021; 12:664457. [PMID: 33868315 PMCID: PMC8050340 DOI: 10.3389/fimmu.2021.664457] [Citation(s) in RCA: 149] [Impact Index Per Article: 37.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Accepted: 03/16/2021] [Indexed: 12/13/2022] Open
Abstract
The burden of heart failure (HF), developing after myocardial infarction MI, still represents a major issue in clinical practice. Failure of appropriate resolution of inflammation during post-myocardial injury is associated with unsuccessful left ventricular remodeling and underlies HF pathogenesis. Cells of the immune system have been shown to mediate both protective and damaging effects in heart remodeling. This ambiguity of the role of the immune system and inconsistent results of the recent clinical trials question the benefits of anti-inflammatory therapies during acute MI. The present review will summarize knowledge of the roles that different cells of the immune system play in the process of post-infarct cardiac healing. Data on the phenotype, active molecules and functions of the immune cells, based on the results of both experimental and clinical studies, will be provided. For some cellular subsets, such as macrophages, neutrophils, dendritic cells and lymphocytes, an anti-inflammatory activity has been attributed to the specific subpopulations. Activity of other cells, such as eosinophils, mast cells, natural killer (NK) cells and NKT cells has been shown to be highly dependent of the signals created by micro-environment. Also, new approaches for classification of cellular phenotypes based on the single-cell RNA sequencing allow better understanding of the phenotype of the cells involved in resolution of inflammation. Possible perspectives of immune-mediated therapy for AMI patients are discussed in the conclusion. We also outline unresolved questions that need to be solved in order to implement the current knowledge on the role of the immune cells in post-MI tissue repair into practice.
Collapse
Affiliation(s)
- Irina Kologrivova
- Department of Clinical Laboratory Diagnostics, Cardiology Research Institute, Tomsk National Research Medical Centre, Russian Academy of Sciences, Tomsk, Russia
| | - Marina Shtatolkina
- Department of Emergency Cardiology, Cardiology Research Institute, Tomsk National Research Medical Centre, Russian Academy of Sciences, Tomsk, Russia
| | - Tatiana Suslova
- Department of Clinical Laboratory Diagnostics, Cardiology Research Institute, Tomsk National Research Medical Centre, Russian Academy of Sciences, Tomsk, Russia
| | - Vyacheslav Ryabov
- Department of Emergency Cardiology, Cardiology Research Institute, Tomsk National Research Medical Centre, Russian Academy of Sciences, Tomsk, Russia.,Division of Cardiology, Department of Professional Development and Retraining, Siberian State Medical University, Tomsk, Russia
| |
Collapse
|
45
|
Infarct in the Heart: What's MMP-9 Got to Do with It? Biomolecules 2021; 11:biom11040491. [PMID: 33805901 PMCID: PMC8064345 DOI: 10.3390/biom11040491] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 03/19/2021] [Accepted: 03/21/2021] [Indexed: 12/12/2022] Open
Abstract
Over the past three decades, numerous studies have shown a strong connection between matrix metalloproteinase 9 (MMP-9) levels and myocardial infarction (MI) mortality and left ventricle remodeling and dysfunction. Despite this fact, clinical trials using MMP-9 inhibitors have been disappointing. This review focuses on the roles of MMP-9 in MI wound healing. Infiltrating leukocytes, cardiomyocytes, fibroblasts, and endothelial cells secrete MMP-9 during all phases of cardiac repair. MMP-9 both exacerbates the inflammatory response and aids in inflammation resolution by stimulating the pro-inflammatory to reparative cell transition. In addition, MMP-9 has a dual effect on neovascularization and prevents an overly stiff scar. Here, we review the complex role of MMP-9 in cardiac wound healing, and highlight the importance of targeting MMP-9 only for its detrimental actions. Therefore, delineating signaling pathways downstream of MMP-9 is critical.
Collapse
|
46
|
Santos-Zas I, Lemarié J, Zlatanova I, Cachanado M, Seghezzi JC, Benamer H, Goube P, Vandestienne M, Cohen R, Ezzo M, Duval V, Zhang Y, Su JB, Bizé A, Sambin L, Bonnin P, Branchereau M, Heymes C, Tanchot C, Vilar J, Delacroix C, Hulot JS, Cochain C, Bruneval P, Danchin N, Tedgui A, Mallat Z, Simon T, Ghaleh B, Silvestre JS, Ait-Oufella H. Cytotoxic CD8 + T cells promote granzyme B-dependent adverse post-ischemic cardiac remodeling. Nat Commun 2021; 12:1483. [PMID: 33674611 PMCID: PMC7935973 DOI: 10.1038/s41467-021-21737-9] [Citation(s) in RCA: 86] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Accepted: 02/09/2021] [Indexed: 12/21/2022] Open
Abstract
Acute myocardial infarction is a common condition responsible for heart failure and sudden death. Here, we show that following acute myocardial infarction in mice, CD8+ T lymphocytes are recruited and activated in the ischemic heart tissue and release Granzyme B, leading to cardiomyocyte apoptosis, adverse ventricular remodeling and deterioration of myocardial function. Depletion of CD8+ T lymphocytes decreases apoptosis within the ischemic myocardium, hampers inflammatory response, limits myocardial injury and improves heart function. These effects are recapitulated in mice with Granzyme B-deficient CD8+ T cells. The protective effect of CD8 depletion on heart function is confirmed by using a model of ischemia/reperfusion in pigs. Finally, we reveal that elevated circulating levels of GRANZYME B in patients with acute myocardial infarction predict increased risk of death at 1-year follow-up. Our work unravels a deleterious role of CD8+ T lymphocytes following acute ischemia, and suggests potential therapeutic strategies targeting pathogenic CD8+ T lymphocytes in the setting of acute myocardial infarction. Immune cells contribute to adverse remodeling following myocardial infarction. Here the authors show in mice and pigs that CD8+ lymphocytes release Granzyme B in the infarcted heart leading to cardiomyocyte death, enhanced inflammation and deterioration of cardiac function.
Collapse
Affiliation(s)
| | | | | | - Marine Cachanado
- Assistance Publique-Hôpitaux de Paris, APHP.SU; Department of Clinical Pharmacology and Clinical Research Platform (URCEST-CRB-CRC-EST), Hôpital Saint Antoine, Paris, France
| | | | - Hakim Benamer
- Service de cardiologie, Institut Cardiovasculaire Paris Sud, Paris, France
| | - Pascal Goube
- Service de cardiologie, Centre Hospitalier de Corbeil, Corbeil, France
| | | | - Raphael Cohen
- Université de Paris, PARCC, INSERM, F-75015, Paris, France
| | - Maya Ezzo
- Université de Paris, PARCC, INSERM, F-75015, Paris, France
| | - Vincent Duval
- Université de Paris, PARCC, INSERM, F-75015, Paris, France
| | - Yujiao Zhang
- Université de Paris, PARCC, INSERM, F-75015, Paris, France
| | - Jin-Bo Su
- Inserm U955-IMRB, Equipe 03, UPEC, Ecole Nationale Vétérinaire d'Alfort, Maisons-Alfort, France
| | - Alain Bizé
- Inserm U955-IMRB, Equipe 03, UPEC, Ecole Nationale Vétérinaire d'Alfort, Maisons-Alfort, France
| | - Lucien Sambin
- Inserm U955-IMRB, Equipe 03, UPEC, Ecole Nationale Vétérinaire d'Alfort, Maisons-Alfort, France
| | - Philippe Bonnin
- Inserm U965, Department of Physiology, Assistance Publique Hôpitaux de Paris, Hôpital Lariboisière, France
| | - Maxime Branchereau
- Inserm U1048-Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), université Paul Sabatier, Toulouse, France
| | - Christophe Heymes
- Inserm U1048-Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), université Paul Sabatier, Toulouse, France
| | | | - José Vilar
- Université de Paris, PARCC, INSERM, F-75015, Paris, France
| | | | | | - Clement Cochain
- Institute of Experimental Biomedicine, University Hospital Würzburg, Würzburg, Germany
| | - Patrick Bruneval
- Université de Paris, PARCC, INSERM, F-75015, Paris, France.,Service d'anatomopathologie, Hôpital Europeen G. Pompidou, Assistance Publique, Hôpitaux de Paris, Paris, France
| | - Nicolas Danchin
- Service de cardiologie, Hôpital Europeen G. Pompidou, Assistance Publique, Hôpitaux de Paris, Paris, France
| | - Alain Tedgui
- Université de Paris, PARCC, INSERM, F-75015, Paris, France
| | - Ziad Mallat
- Université de Paris, PARCC, INSERM, F-75015, Paris, France.,Division of Cardiovascular Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge, CB2 2QQ, UK
| | - Tabassome Simon
- Assistance Publique-Hôpitaux de Paris, APHP.SU; Department of Clinical Pharmacology and Clinical Research Platform (URCEST-CRB-CRC-EST), Hôpital Saint Antoine, Paris, France.,Sorbonne Université, UPMC-site St Antoine, Service de Pharmacologie, Assistance Publique-Hôpitaux de Paris, APHP.SU; Department of Clinical Pharmacology and Clinical Research Platform (URCEST-CRB-CRC-EST), Hôpital Saint Antoine, Paris, France
| | - Bijan Ghaleh
- Inserm U955-IMRB, Equipe 03, UPEC, Ecole Nationale Vétérinaire d'Alfort, Maisons-Alfort, France
| | | | - Hafid Ait-Oufella
- Université de Paris, PARCC, INSERM, F-75015, Paris, France. .,Sorbonne Université, Service de médecine intensive-Réanimation, Assistance Publique, Hôpitaux de Paris, Paris, France.
| |
Collapse
|
47
|
Scalise RFM, De Sarro R, Caracciolo A, Lauro R, Squadrito F, Carerj S, Bitto A, Micari A, Bella GD, Costa F, Irrera N. Fibrosis after Myocardial Infarction: An Overview on Cellular Processes, Molecular Pathways, Clinical Evaluation and Prognostic Value. Med Sci (Basel) 2021; 9:medsci9010016. [PMID: 33804308 PMCID: PMC7931027 DOI: 10.3390/medsci9010016] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 02/17/2021] [Accepted: 02/22/2021] [Indexed: 12/16/2022] Open
Abstract
The ischemic injury caused by myocardial infarction activates a complex healing process wherein a powerful inflammatory response and a reparative phase follow and balance each other. An intricate network of mediators finely orchestrate a large variety of cellular subtypes throughout molecular signaling pathways that determine the intensity and duration of each phase. At the end of this process, the necrotic tissue is replaced with a fibrotic scar whose quality strictly depends on the delicate balance resulting from the interaction between multiple actors involved in fibrogenesis. An inflammatory or reparative dysregulation, both in term of excess and deficiency, may cause ventricular dysfunction and life-threatening arrhythmias that heavily affect clinical outcome. This review discusses cellular process and molecular signaling pathways that determine fibrosis and the imaging technique that can characterize the clinical impact of this process in-vivo.
Collapse
Affiliation(s)
- Renato Francesco Maria Scalise
- Department of Clinical and Experimental Medicine, Policlinic “G. Martino”, University of Messina, 98100 Messina, Italy; (R.F.M.S.); (R.D.S.); (A.C.); (S.C.); (G.D.B.); (N.I.)
| | - Rosalba De Sarro
- Department of Clinical and Experimental Medicine, Policlinic “G. Martino”, University of Messina, 98100 Messina, Italy; (R.F.M.S.); (R.D.S.); (A.C.); (S.C.); (G.D.B.); (N.I.)
| | - Alessandro Caracciolo
- Department of Clinical and Experimental Medicine, Policlinic “G. Martino”, University of Messina, 98100 Messina, Italy; (R.F.M.S.); (R.D.S.); (A.C.); (S.C.); (G.D.B.); (N.I.)
| | - Rita Lauro
- Section of Pharmacology, Department of Clinical and Experimental Medicine, University of Messina, 98100 Messina, Italy; (R.L.); (F.S.); (A.B.)
| | - Francesco Squadrito
- Section of Pharmacology, Department of Clinical and Experimental Medicine, University of Messina, 98100 Messina, Italy; (R.L.); (F.S.); (A.B.)
| | - Scipione Carerj
- Department of Clinical and Experimental Medicine, Policlinic “G. Martino”, University of Messina, 98100 Messina, Italy; (R.F.M.S.); (R.D.S.); (A.C.); (S.C.); (G.D.B.); (N.I.)
| | - Alessandra Bitto
- Section of Pharmacology, Department of Clinical and Experimental Medicine, University of Messina, 98100 Messina, Italy; (R.L.); (F.S.); (A.B.)
| | - Antonio Micari
- Department of Biomedical and Dental Sciences and Morphological and Functional Imaging, University of Messina, A.O.U. Policlinico “G. Martino”, 98100 Messina, Italy;
| | - Gianluca Di Bella
- Department of Clinical and Experimental Medicine, Policlinic “G. Martino”, University of Messina, 98100 Messina, Italy; (R.F.M.S.); (R.D.S.); (A.C.); (S.C.); (G.D.B.); (N.I.)
| | - Francesco Costa
- Department of Clinical and Experimental Medicine, Policlinic “G. Martino”, University of Messina, 98100 Messina, Italy; (R.F.M.S.); (R.D.S.); (A.C.); (S.C.); (G.D.B.); (N.I.)
- Correspondence: ; Tel.: +39-090-221-23-41; Fax: +39-090-221-23-81
| | - Natasha Irrera
- Department of Clinical and Experimental Medicine, Policlinic “G. Martino”, University of Messina, 98100 Messina, Italy; (R.F.M.S.); (R.D.S.); (A.C.); (S.C.); (G.D.B.); (N.I.)
| |
Collapse
|
48
|
Forte E, Perkins B, Sintou A, Kalkat HS, Papanikolaou A, Jenkins C, Alsubaie M, Chowdhury RA, Duffy TM, Skelly DA, Branca J, Bellahcene M, Schneider MD, Harding SE, Furtado MB, Ng FS, Hasham MG, Rosenthal N, Sattler S. Cross-Priming Dendritic Cells Exacerbate Immunopathology After Ischemic Tissue Damage in the Heart. Circulation 2021; 143:821-836. [PMID: 33297741 PMCID: PMC7899721 DOI: 10.1161/circulationaha.120.044581] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Accepted: 11/04/2020] [Indexed: 12/14/2022]
Abstract
BACKGROUND Ischemic heart disease is a leading cause of heart failure and despite advanced therapeutic options, morbidity and mortality rates remain high. Although acute inflammation in response to myocardial cell death has been extensively studied, subsequent adaptive immune activity and anti-heart autoimmunity may also contribute to the development of heart failure. After ischemic injury to the myocardium, dendritic cells (DC) respond to cardiomyocyte necrosis, present cardiac antigen to T cells, and potentially initiate a persistent autoimmune response against the heart. Cross-priming DC have the ability to activate both CD4+ helper and CD8+ cytotoxic T cells in response to necrotic cells and may thus be crucial players in exacerbating autoimmunity targeting the heart. This study investigates a role for cross-priming DC in post-myocardial infarction immunopathology through presentation of self-antigen from necrotic cardiac cells to cytotoxic CD8+ T cells. METHODS We induced type 2 myocardial infarction-like ischemic injury in the heart by treatment with a single high dose of the β-adrenergic agonist isoproterenol. We characterized the DC population in the heart and mediastinal lymph nodes and analyzed long-term cardiac immunopathology and functional decline in wild type and Clec9a-depleted mice lacking DC cross-priming function. RESULTS A diverse DC population, including cross-priming DC, is present in the heart and activated after ischemic injury. Clec9a-/- mice deficient in DC cross-priming are protected from persistent immune-mediated myocardial damage and decline of cardiac function, likely because of dampened activation of cytotoxic CD8+ T cells. CONCLUSION Activation of cytotoxic CD8+ T cells by cross-priming DC contributes to exacerbation of postischemic inflammatory damage of the myocardium and corresponding decline in cardiac function. Importantly, this provides novel therapeutic targets to prevent postischemic immunopathology and heart failure.
Collapse
Affiliation(s)
- Elvira Forte
- The Jackson Laboratory, Bar Harbor, ME (E.F., B.P., T.M.D., D.A.S., J.B., M.B.F., M.G.H., N.R.)
| | - Bryant Perkins
- The Jackson Laboratory, Bar Harbor, ME (E.F., B.P., T.M.D., D.A.S., J.B., M.B.F., M.G.H., N.R.)
| | - Amalia Sintou
- National Heart and Lung Institute, Imperial College London, UK (A.S., H.S.K., A.P., C.J., M.A., R.A.C., M.B., M.D.S., S.E.H., F.S.N., N.R., S.S.)
| | - Harkaran S. Kalkat
- National Heart and Lung Institute, Imperial College London, UK (A.S., H.S.K., A.P., C.J., M.A., R.A.C., M.B., M.D.S., S.E.H., F.S.N., N.R., S.S.)
| | - Angelos Papanikolaou
- National Heart and Lung Institute, Imperial College London, UK (A.S., H.S.K., A.P., C.J., M.A., R.A.C., M.B., M.D.S., S.E.H., F.S.N., N.R., S.S.)
| | - Catherine Jenkins
- National Heart and Lung Institute, Imperial College London, UK (A.S., H.S.K., A.P., C.J., M.A., R.A.C., M.B., M.D.S., S.E.H., F.S.N., N.R., S.S.)
| | - Mashael Alsubaie
- National Heart and Lung Institute, Imperial College London, UK (A.S., H.S.K., A.P., C.J., M.A., R.A.C., M.B., M.D.S., S.E.H., F.S.N., N.R., S.S.)
| | - Rasheda A. Chowdhury
- National Heart and Lung Institute, Imperial College London, UK (A.S., H.S.K., A.P., C.J., M.A., R.A.C., M.B., M.D.S., S.E.H., F.S.N., N.R., S.S.)
| | - Theodore M. Duffy
- The Jackson Laboratory, Bar Harbor, ME (E.F., B.P., T.M.D., D.A.S., J.B., M.B.F., M.G.H., N.R.)
| | - Daniel A. Skelly
- The Jackson Laboratory, Bar Harbor, ME (E.F., B.P., T.M.D., D.A.S., J.B., M.B.F., M.G.H., N.R.)
| | - Jane Branca
- The Jackson Laboratory, Bar Harbor, ME (E.F., B.P., T.M.D., D.A.S., J.B., M.B.F., M.G.H., N.R.)
| | - Mohamed Bellahcene
- National Heart and Lung Institute, Imperial College London, UK (A.S., H.S.K., A.P., C.J., M.A., R.A.C., M.B., M.D.S., S.E.H., F.S.N., N.R., S.S.)
| | - Michael D. Schneider
- National Heart and Lung Institute, Imperial College London, UK (A.S., H.S.K., A.P., C.J., M.A., R.A.C., M.B., M.D.S., S.E.H., F.S.N., N.R., S.S.)
| | - Sian E. Harding
- National Heart and Lung Institute, Imperial College London, UK (A.S., H.S.K., A.P., C.J., M.A., R.A.C., M.B., M.D.S., S.E.H., F.S.N., N.R., S.S.)
| | - Milena B. Furtado
- The Jackson Laboratory, Bar Harbor, ME (E.F., B.P., T.M.D., D.A.S., J.B., M.B.F., M.G.H., N.R.)
- Amgen Biotechnology, Thousand Oaks, CA (M.B.F.)
| | - Fu Siong Ng
- National Heart and Lung Institute, Imperial College London, UK (A.S., H.S.K., A.P., C.J., M.A., R.A.C., M.B., M.D.S., S.E.H., F.S.N., N.R., S.S.)
| | - Muneer G. Hasham
- The Jackson Laboratory, Bar Harbor, ME (E.F., B.P., T.M.D., D.A.S., J.B., M.B.F., M.G.H., N.R.)
| | - Nadia Rosenthal
- The Jackson Laboratory, Bar Harbor, ME (E.F., B.P., T.M.D., D.A.S., J.B., M.B.F., M.G.H., N.R.)
- National Heart and Lung Institute, Imperial College London, UK (A.S., H.S.K., A.P., C.J., M.A., R.A.C., M.B., M.D.S., S.E.H., F.S.N., N.R., S.S.)
| | - Susanne Sattler
- National Heart and Lung Institute, Imperial College London, UK (A.S., H.S.K., A.P., C.J., M.A., R.A.C., M.B., M.D.S., S.E.H., F.S.N., N.R., S.S.)
| |
Collapse
|
49
|
Li Y, Qin L, Bai Q, Zhang J, Chen R, Song K. CD100 modulates cytotoxicity of CD8 + T cells in patients with acute myocardial infarction. BMC Immunol 2021; 22:13. [PMID: 33593275 PMCID: PMC7888114 DOI: 10.1186/s12865-021-00406-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Accepted: 02/03/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND CD100 is an immune semaphorin family member that highly expressed on T cells, which take part in the development of acute myocardial infarction (AMI). Matrix metalloproteinases (MMPs) are important mediators for membrane-bound CD100 (mCD100) shedding from T cells to generate soluble CD100 (sCD100), which has immunoregulatory effect on T cells. The aim of this study was to investigate modulatory role of CD100 on CD8+ T cell activity in AMI patients. METHODS Peripheral sCD100 and MMP-2 level, as well as mCD100 level on T cells was assessed in patients with stable angina pectoris (SAP), unstable angina pectoris (UAP), and AMI. The regulatory function of MMP-2 on mCD100 shedding, sCD100 formation, and cytotoxicity of CD8+ T cells was analyzed in direct and indirect contact co-culture system. RESULTS AMI patients had higher peripheral sCD100 and lower mCD100 expression on CD8+ T cells in comparison with SAP, UAP, and controls. CD8+ T cells in AMI patients showed elevated direct cytotoxicity, enhanced cytokine production, and increased perforin/granzyme B secretion. Recombinant sCD100 stimulation promoted cytolytic function of CD8+ T cells in controls and AMI patients. Furthermore, AMI patients also had elevated circulating MMP-2 level. Recombinant MMP-2 stimulation induced mCD100 shedding from CD8+ T cells and sCD100 generation, resulting in enhancement of CD8+ T cell cytotoxicity in AMI patients. CONCLUSION Up-regulation of MMP-2 might contribute to elevation of mCD100 shedding and sCD100 formation, leading to increased cytotoxicity CD8+ T cells in AMI patients.
Collapse
Affiliation(s)
- Yan Li
- Department of Cardiovascular Medicine Ward II, Zhengzhou Central Hospital Affiliated to Zhengzhou University, 16 North Tongbai Road, Zhongyuan District, Zhengzhou, 450000, Henan Province, China
| | - Li Qin
- Department of Cardiovascular Medicine Ward II, Zhengzhou Central Hospital Affiliated to Zhengzhou University, 16 North Tongbai Road, Zhongyuan District, Zhengzhou, 450000, Henan Province, China
| | - Qijun Bai
- Department of Cardiovascular Medicine Ward II, Zhengzhou Central Hospital Affiliated to Zhengzhou University, 16 North Tongbai Road, Zhongyuan District, Zhengzhou, 450000, Henan Province, China
| | - Jingjing Zhang
- Department of Cardiovascular Medicine Ward II, Zhengzhou Central Hospital Affiliated to Zhengzhou University, 16 North Tongbai Road, Zhongyuan District, Zhengzhou, 450000, Henan Province, China
| | - Ruixue Chen
- Department of Cardiovascular Medicine Ward II, Zhengzhou Central Hospital Affiliated to Zhengzhou University, 16 North Tongbai Road, Zhongyuan District, Zhengzhou, 450000, Henan Province, China
| | - Kunpeng Song
- Department of Cardiovascular Medicine Ward II, Zhengzhou Central Hospital Affiliated to Zhengzhou University, 16 North Tongbai Road, Zhongyuan District, Zhengzhou, 450000, Henan Province, China.
| |
Collapse
|
50
|
Zaidi Y, Aguilar EG, Troncoso M, Ilatovskaya DV, DeLeon-Pennell KY. Immune regulation of cardiac fibrosis post myocardial infarction. Cell Signal 2021; 77:109837. [PMID: 33207261 PMCID: PMC7720290 DOI: 10.1016/j.cellsig.2020.109837] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 11/11/2020] [Accepted: 11/12/2020] [Indexed: 12/24/2022]
Abstract
Pathological changes resulting from myocardial infarction (MI) include extracellular matrix alterations of the left ventricle, which can lead to cardiac stiffness and impair systolic and diastolic function. The signals released from necrotic tissue initiate the immune cascade, triggering an extensive inflammatory response followed by reparative fibrosis of the infarct area. Immune cells such as neutrophils, monocytes, macrophages, mast cells, T-cells, and dendritic cells play distinct roles in orchestrating this complex pathological condition, and regulate the balance between pro-fibrotic and anti-fibrotic responses. This review discusses how molecular signals between fibroblasts and immune cells mutually regulate fibrosis post-MI, and outlines the emerging pharmacological targets and therapies for modulating inflammation and cardiac fibrosis associated with MI.
Collapse
Affiliation(s)
- Yusra Zaidi
- Department of Medicine, Division of Cardiology, Medical University of South Carolina, 30 Courtenay Drive, Charleston, SC 29425, USA
| | - Eslie G Aguilar
- Department of Medicine, Division of Cardiology, Medical University of South Carolina, 30 Courtenay Drive, Charleston, SC 29425, USA
| | - Miguel Troncoso
- Department of Medicine, Division of Cardiology, Medical University of South Carolina, 30 Courtenay Drive, Charleston, SC 29425, USA
| | - Daria V Ilatovskaya
- Division of Nephrology, Department of Medicine, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Kristine Y DeLeon-Pennell
- Department of Medicine, Division of Cardiology, Medical University of South Carolina, 30 Courtenay Drive, Charleston, SC 29425, USA; Ralph H. Johnson Veterans Affairs Medical Center, 109 Bee Street, Charleston, SC 29401, USA.
| |
Collapse
|