1
|
Li Y. Novel Therapeutic Strategies Targeting Fibroblasts to Improve Heart Disease. J Cell Physiol 2025; 240:e31504. [PMID: 39690827 DOI: 10.1002/jcp.31504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 11/09/2024] [Accepted: 11/22/2024] [Indexed: 12/19/2024]
Abstract
Cardiac fibrosis represents the terminal pathological manifestation of various heart diseases, with the formation of fibroblasts playing a pivotal role in this process. Consequently, targeting the formation and function of fibroblasts holds significant potential for improving outcomes in heart disease. Recent research reveals the considerable potential of fibroblasts in ameliorating cardiac conditions, demonstrating different functional characteristics at various time points and spatial locations. Therefore, precise modulation of fibroblast activity may offer an effective approach for treating cardiac fibrosis and achieving targeted therapeutic outcomes. In this review, we focus on the fate and inhibition of fibroblasts, analyze their dynamic changes in cardiac diseases, and propose a framework for identifying markers of fibroblast activation mechanisms and selecting optimal time windows for therapeutic intervention. By synthesizing research findings in these areas, we aim to provide new strategies and directions for the precise treatment of fibroblasts in cardiac diseases.
Collapse
Affiliation(s)
- Yujuan Li
- Medical College of Optometry and Ophthalmology, Shandong University of Traditional Chinese Medicine, Jinan, China
- Shandong Provincial Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Therapy of Ocular Diseases, Shandong Academy of Eye Disease Prevention and Therapy, Affiliated Eye Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| |
Collapse
|
2
|
FitzGerald G. Metabolomic Response to Non-Steroidal Anti-Inflammatory Drugs. RESEARCH SQUARE 2024:rs.3.rs-5530702. [PMID: 39711561 PMCID: PMC11661377 DOI: 10.21203/rs.3.rs-5530702/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2024]
Abstract
Non-steroidal anti-inflammatory drugs (NSAIDs) are popular choices for the mitigation of pain and inflammation; however, they are accompanied by side effects in the gastrointestinal and cardiovascular systems. We compared the effects of naproxen, a traditional NSAID, and celecoxib, a cyclooxygenase - 2 (Cox-2) inhibitor, in humans. Our findings showed a decrease in tryptophan and kynurenine levels in plasma of volunteers treated with naproxen. We further validated this result in mice. Additionally, we find that the depression of tryptophan was independent of both Cox-1 and Cox-2 inhibition, but rather was due to the displacement of bound tryptophan by naproxen. Supplementation of tryptophan in naproxen-treated mice rescued fecal blood loss and inflammatory gene expression driven by IL-1β in the heart.
Collapse
|
3
|
Ghosh S, Lahens N, Barekat K, Tang SY, Theken KN, Ricciotti E, Sengupta A, Joshi R, Bushman FD, Weljie A, Grosser T, FitzGerald GA. Metabolomic Response to Non-Steroidal Anti-Inflammatory Drugs. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.26.625478. [PMID: 39677795 PMCID: PMC11642787 DOI: 10.1101/2024.11.26.625478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/17/2024]
Abstract
Non-steroidal anti-inflammatory drugs (NSAIDs) are popular choices for the mitigation of pain and inflammation; however, they are accompanied by side effects in the gastrointestinal and cardiovascular systems. We compared the effects of naproxen, a traditional NSAID, and celecoxib, a cyclooxygenase -2 (Cox-2) inhibitor, in humans. Our findings showed a decrease in tryptophan and kynurenine levels in plasma of volunteers treated with naproxen. We further validated this result in mice. Additionally, we find that the depression of tryptophan was independent of both Cox-1 and Cox-2 inhibition, but rather was due to the displacement of bound tryptophan by naproxen. Supplementation of tryptophan in naproxen-treated mice rescued fecal blood loss and inflammatory gene expression driven by IL-1β in the heart.
Collapse
Affiliation(s)
- Soumita Ghosh
- Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Nick Lahens
- Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Kayla Barekat
- Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Soon-Yew Tang
- Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Katherine N Theken
- Department of Oral and Maxillofacial Surgery and Pharmacology, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Emanuela Ricciotti
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, PA, United States
| | - Arjun Sengupta
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, PA, United States
| | - Robin Joshi
- Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Frederic D Bushman
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Aalim Weljie
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, PA, United States
| | - Tilo Grosser
- Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Department of Translational Pharmacology, EWL School of Medicine, Bielefeld University, Bielefeld, Germany
| | - Garret A FitzGerald
- Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Department of Medicine, School of Medicine, University of Pennsylvania, Philadelphia, PA
| |
Collapse
|
4
|
Yu J, Long B, Li Z, Tian X, Li D, Long J, Wang Y, Chen Y, Zhang F, Liu H, Qian C, Shan J. Central memory CD4+ T cells play a protective role against immune checkpoint inhibitor-associated myocarditis. Cardiovasc Res 2024; 120:1442-1455. [PMID: 38850163 DOI: 10.1093/cvr/cvae133] [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: 08/22/2023] [Revised: 03/04/2024] [Accepted: 05/11/2024] [Indexed: 06/10/2024] Open
Abstract
AIMS The widespread use of immune checkpoint inhibitors (ICIs) has demonstrated significant survival benefits for cancer patients and also carries the risk of immune-related adverse events. ICI-associated myocarditis is a rare and serious adverse event with a high mortality rate. Here, we explored the mechanism underlying ICI-associated myocarditis. METHODS AND RESULTS Using the peripheral blood of patients with ICI therapy and of ICI-treated mice with transplanted tumours, we dissect the immune cell subsets and inflammatory factors associated with myocarditis. Compared to the control group, patients with myocarditis after ICI therapy showed an increase in NK cells and myeloid cells in the peripheral blood, while T cells significantly decreased. Among T cells, there was an imbalance of CD4/CD8 ratio in the peripheral blood of myocarditis patients, with a significant decrease in central memory CD4+ T (CD4+ TCM) cells. RNA sequencing revealed that CD4+ TCM cells in myocarditis patients were immunosuppressive cell subsets, which highly express the immunosuppressive factor IL-4I1. To elucidate the potential mechanism of the decrease in CD4+ TCM cells, protein array was performed and revealed that several inflammatory factors gradually increased with the severity of myocarditis in the myocarditis group, such as IL-1B/CXCL13/CXCL9, while the myocardial protective factor IL-15 decreased. Correlation analysis indicated a positive correlation between IL-15 and CD4+ TCM cells, with high expression of IL-15 receptor IL15RA. Furthermore, in vivo studies using an anti-PDL1 antibody in a mouse tumour model indicated a reduction in CD4+ TCM cells and an increase in effector memory-expressing CD45RA CD8+ T (TEMRA) cells, alongside evidence of cardiac fibrosis. Conversely, combining anti-PDL1 antibody treatment with IL-15 led to a resurgence of CD4+ TCM cells, a reduction in CD8+ TEMRA cells, and a mitigated risk of cardiac fibrosis. CONCLUSION Our data highlight CD4+ TCM cells' crucial role in cardiac protection during ICI therapy. IL-15, IL-4I1, and CD4+ TCM cells can serve as therapeutic targets to reduce ICI-associated myocarditis in cancer patients.
Collapse
Affiliation(s)
- Jiajun Yu
- School of Medicine, Chongqing University, Chongqing 400030, China
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, No. 181, Hanyulu, Shapingba, Chongqing 400030, China
| | - Bo Long
- Department of Cardio-Oncology, Chongqing University Cancer Hospital, No. 181, Hanyulu, Shapingba, Chongqing 400030, China
| | - Ziyong Li
- School of Medicine, Chongqing University, Chongqing 400030, China
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, No. 181, Hanyulu, Shapingba, Chongqing 400030, China
| | - Xiaolong Tian
- School of Medicine, Chongqing University, Chongqing 400030, China
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, No. 181, Hanyulu, Shapingba, Chongqing 400030, China
| | - Dairong Li
- Department of Medical Oncology, Chongqing University Cancer Hospital, No. 181, Hanyulu, Shapingba, Chongqing 400030, China
| | - Jianling Long
- Department of Cardio-Oncology, Chongqing University Cancer Hospital, No. 181, Hanyulu, Shapingba, Chongqing 400030, China
| | - Yujue Wang
- Department of Cardio-Oncology, Chongqing University Cancer Hospital, No. 181, Hanyulu, Shapingba, Chongqing 400030, China
| | - Yue Chen
- Department of Cardio-Oncology, Chongqing University Cancer Hospital, No. 181, Hanyulu, Shapingba, Chongqing 400030, China
| | - Fang Zhang
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, No. 181, Hanyulu, Shapingba, Chongqing 400030, China
| | - Haixia Liu
- Department of Cardio-Oncology, Chongqing University Cancer Hospital, No. 181, Hanyulu, Shapingba, Chongqing 400030, China
| | - Cheng Qian
- School of Medicine, Chongqing University, Chongqing 400030, China
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, No. 181, Hanyulu, Shapingba, Chongqing 400030, China
| | - Juanjuan Shan
- School of Medicine, Chongqing University, Chongqing 400030, China
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, No. 181, Hanyulu, Shapingba, Chongqing 400030, China
| |
Collapse
|
5
|
Barisione C, Verzola D, Garibaldi S, Altieri P, Furfaro AL, Nitti M, Pratesi G, Palombo D, Ameri P. Indoxyl sulphate-initiated activation of cardiac fibroblasts is modulated by aryl hydrocarbon receptor and nuclear factor-erythroid-2-related factor 2. J Cell Mol Med 2024; 28:e18192. [PMID: 38506079 PMCID: PMC10951876 DOI: 10.1111/jcmm.18192] [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: 10/23/2023] [Revised: 01/15/2024] [Accepted: 02/09/2024] [Indexed: 03/21/2024] Open
Abstract
In the last decade, extensive attention has been paid to the uremic toxin indoxyl sulphate (IS) as an inducer of cardiac fibroblast (cFib) activation and cardiac fibrosis in chronic kidney disease. At cellular level, IS engages aryl hydrocarbon receptor (AhR) and regulates many biological functions. We analysed how AhR inhibition by CH-223191 (CH) and overexpression of non-functional (dominant negative, DN) nuclear factor-erythroid-2-related factor 2 (NRF2), a transcription factor recruited by AhR, modulate the response of neonatal mouse (nm) cFib to IS. We also evaluated nm-cardiomyocytes after incubation with the conditioned medium (CM) of IS±CH-treated nm-cFib. IS induced activation, collagen synthesis, TLR4 and-downstream-MCP-1, and the genes encoding angiotensinogen, angiotensin-converting enzyme, angiotensin type 1 receptor (AT1r) and neprilysin (Nepr) in nm-cFib. CH antagonized IS-initiated nm-cFib activation, but did not affect or even magnified the other features. IS promoted NRF2 nuclear translocation and expression the NRF2 target Nqo1. Both pre-incubation with CH and transfection of DN-NRF2 resulted in loss of NRF2 nuclear localization. Moreover, DN-NRF2 overexpression led to greater TLR4 and MCP-1 levels following exposure to IS. The CM of IS-primed nm-cFib and to a larger extent the CM of IS+CH-treated nm-cFib upregulated AT1r, Nepr and TNFα and myostatin genes in nm-cardiomyocytes. Hence, IS triggers pro-inflammatory activation of nm-cFib partly via AhR, and AhR-NRF2 counteract it. Strategies other than AhR inhibition are needed to target IS detrimental actions on cardiac cells.
Collapse
Affiliation(s)
- Chiara Barisione
- Department of Surgical and Integrated Diagnostic SciencesUniversity of GenovaGenovaItaly
- Cardiac, Thoracic and Vascular DepartmentIRCCS Ospedale Policlinico San MartinoGenovaItaly
| | - Daniela Verzola
- Department of Internal MedicineUniversity of GenovaGenovaItaly
| | | | - Paola Altieri
- Department of Internal MedicineUniversity of GenovaGenovaItaly
| | | | - Mariapaola Nitti
- Department of Experimental MedicineUniversity of GenovaGenovaItaly
| | - Giovanni Pratesi
- Department of Surgical and Integrated Diagnostic SciencesUniversity of GenovaGenovaItaly
- Cardiac, Thoracic and Vascular DepartmentIRCCS Ospedale Policlinico San MartinoGenovaItaly
| | - Domenico Palombo
- Department of Surgical and Integrated Diagnostic SciencesUniversity of GenovaGenovaItaly
| | - Pietro Ameri
- Cardiac, Thoracic and Vascular DepartmentIRCCS Ospedale Policlinico San MartinoGenovaItaly
- Department of Internal MedicineUniversity of GenovaGenovaItaly
| |
Collapse
|
6
|
Babaei G, Sadraei S, Yarahmadi M, Omidvari S, Aarabi A, Rajabibazl M. STAT protein family and cardiovascular diseases: overview of pathological mechanisms and therapeutic implications. Mol Biol Rep 2024; 51:440. [PMID: 38520542 DOI: 10.1007/s11033-024-09371-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Accepted: 02/21/2024] [Indexed: 03/25/2024]
Abstract
Globally, cardiovascular diseases (CVD) are one of the significant causes of death and are considered a major concern of human society. One of the most crucial objectives of scientists is to reveal the mechanisms associated with the pathogenesis of CVD, which has attracted the attention of many scientists. Accumulating evidence showed that the signal transducer and activator of transcription (STAT) signaling pathway is involved in various physiological and pathological processes. According to research on the molecular mechanisms of CVDs, the STAT family of proteins is one of the most crucial players in these diseases. Numerous studies have demonstrated the undeniable relevance of STAT family proteins in various CVDs. The aim of this review is to shed light on how STAT signaling pathways are related to CVD and the potential for using these signaling pathways as therapeutic targets.
Collapse
Affiliation(s)
- Ghader Babaei
- Department of Clinical Biochemistry, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Samin Sadraei
- Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Maral Yarahmadi
- Department of Clinical Biochemistry, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Samareh Omidvari
- Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Aryan Aarabi
- Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Masoumeh Rajabibazl
- Department of Clinical Biochemistry, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
7
|
Agoston-Coldea L, Negru A. Myocardial fibrosis in right heart dysfunction. Adv Clin Chem 2024; 119:71-116. [PMID: 38514212 DOI: 10.1016/bs.acc.2024.02.005] [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: 03/23/2024]
Abstract
Cardiac fibrosis, associated with right heart dysfunction, results in significant morbidity and mortality. Stimulated by various cellular and humoral stimuli, cardiac fibroblasts, macrophages, CD4+ and CD8+ T cells, mast and endothelial cells promote fibrogenesis directly and indirectly by synthesizing numerous profibrotic factors. Several systems, including the transforming growth factor-beta and the renin-angiotensin system, produce type I and III collagen, fibronectin and α-smooth muscle actin, thus modifying the extracellular matrix. Although magnetic resonance imaging with gadolinium enhancement remains the gold standard, the use of circulating biomarkers represents an inexpensive and attractive means to facilitate detection and monitor cardiovascular fibrosis. This review explores the use of protein and nucleic acid (miRNAs) markers to better understand underlying pathophysiology as well as their role in the development of therapeutics to inhibit and potentially reverse cardiac fibrosis.
Collapse
Affiliation(s)
- Lucia Agoston-Coldea
- Department of Internal Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania.
| | - Andra Negru
- Department of Internal Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| |
Collapse
|
8
|
Martin KR, Gamell C, Tai TY, Bonelli R, Hansen J, Tatoulis J, Alhamdoosh M, Wilson N, Wicks I. Whole blood transcriptomics reveals granulocyte colony-stimulating factor as a mediator of cardiopulmonary bypass-induced systemic inflammatory response syndrome. Clin Transl Immunology 2024; 13:e1490. [PMID: 38375330 PMCID: PMC10875393 DOI: 10.1002/cti2.1490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 12/20/2023] [Accepted: 01/23/2024] [Indexed: 02/21/2024] Open
Abstract
Objectives Systemic inflammatory response syndrome (SIRS) is a frequent complication of cardiopulmonary bypass (CPB). SIRS is associated with significant morbidity and mortality, but its pathogenesis remains incompletely understood, and as a result, biomarkers are lacking and treatment remains expectant and supportive. This study aimed to understand the pathophysiological mechanisms driving SIRS induced by CPB and identify novel therapeutic targets that might reduce systemic inflammation and improve patient outcomes. Methods Twenty-one patients undergoing cardiac surgery and CPB were recruited, and blood was sampled before, during and after surgery. SIRS was defined using the American College of Chest Physicians/Society of Critical Care Medicine criteria. We performed immune cell profiling and whole blood transcriptomics and measured individual mediators in plasma/serum to characterise SIRS induced by CPB. Results Nineteen patients fulfilled criteria for SIRS, with a mean duration of 2.7 days. Neutrophil numbers rose rapidly with CPB and remained elevated for at least 48 h afterwards. Transcriptional signatures associated with neutrophil activation and degranulation were enriched during CPB. We identified a network of cytokines governing these transcriptional changes, including granulocyte colony-stimulating factor (G-CSF), a regulator of neutrophil production and function. Conclusions We identified neutrophils and G-CSF as major regulators of CPB-induced systemic inflammation. Short-term targeting of G-CSF could provide a novel therapeutic strategy to limit neutrophil-mediated inflammation and tissue damage in SIRS induced by CPB.
Collapse
Affiliation(s)
- Katherine R Martin
- WEHIParkvilleVICAustralia
- Department of Medical BiologyUniversity of MelbourneParkvilleVICAustralia
| | | | - Tsin Yee Tai
- WEHIParkvilleVICAustralia
- CSL Innovation, Bio21 InstituteParkvilleVICAustralia
| | - Roberto Bonelli
- WEHIParkvilleVICAustralia
- Department of Medical BiologyUniversity of MelbourneParkvilleVICAustralia
- CSL Innovation, Bio21 InstituteParkvilleVICAustralia
| | | | - James Tatoulis
- Cardiothoracic SurgeryRoyal Melbourne HospitalParkvilleVICAustralia
- Department of SurgeryUniversity of MelbourneParkvilleVICAustralia
| | | | | | - Ian Wicks
- WEHIParkvilleVICAustralia
- Department of Medical BiologyUniversity of MelbourneParkvilleVICAustralia
- Department of RheumatologyRoyal Melbourne HospitalParkvilleVICAustralia
| |
Collapse
|
9
|
Ter Mors B, Spieler V, Merino Asumendi E, Gantert B, Lühmann T, Meinel L. Bioresponsive Cytokine Delivery Responding to Matrix Metalloproteinases. ACS Biomater Sci Eng 2024; 10:29-37. [PMID: 37102329 DOI: 10.1021/acsbiomaterials.2c01320] [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: 04/28/2023]
Abstract
Cytokines are regulated in acute and chronic inflammation, including rheumatoid arthritis (RA) and myocardial infarction (MI). However, the dynamic windows within which cytokine activity/inhibition is desirable in RA and MI change timely and locally during the disease. Therefore, traditional, static delivery regimens are unlikely to meet the idiosyncrasy of these highly dynamic pathophysiological and individual processes. Responsive delivery systems and biomaterials, sensing surrogate markers of inflammation (i.e., matrix metalloproteinases - MMPs) and answering with drug release, may present drug activity at the right time, manner, and place. This article discusses MMPs as surrogate markers for disease activity in RA and MI to clock drug discharge to MMP concentration profiles from MMP-responsive drug delivery systems and biomaterials.
Collapse
Affiliation(s)
- Björn Ter Mors
- Institute of Pharmacy and Food Chemistry, University of Würzburg, Am Hubland, 97074 Würzburg, Germany
| | - Valerie Spieler
- Institute of Pharmacy and Food Chemistry, University of Würzburg, Am Hubland, 97074 Würzburg, Germany
| | - Eduardo Merino Asumendi
- Institute of Pharmacy and Food Chemistry, University of Würzburg, Am Hubland, 97074 Würzburg, Germany
| | - Benedikt Gantert
- Institute of Pharmacy and Food Chemistry, University of Würzburg, Am Hubland, 97074 Würzburg, Germany
| | - Tessa Lühmann
- Institute of Pharmacy and Food Chemistry, University of Würzburg, Am Hubland, 97074 Würzburg, Germany
| | - Lorenz Meinel
- Institute of Pharmacy and Food Chemistry, University of Würzburg, Am Hubland, 97074 Würzburg, Germany
- Helmholtz Institute for RNA-Based Infection Research (HIRI), Helmholtz Center for Infection Research (HZI), 97080 Würzburg, Germany
| |
Collapse
|
10
|
Pyvovar SM, Rudyk I, Scherban TD. The associations of cytokines and gens polymorphisms of β-adrenoceptors in patients with heart failure and some thyroid pathology (literature review and own observations). WIADOMOSCI LEKARSKIE (WARSAW, POLAND : 1960) 2024; 77:105-113. [PMID: 38431814 DOI: 10.36740/wlek202401113] [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: 03/05/2024]
Abstract
OBJECTIVE Aim: To analyze the role of cytokines in the progression of heart failure (HF) in patients with concomitant pathology of the thyroid gland. PATIENTS AND METHODS Materials and Methods: The systematization of literature data on the role of cytokines in the progression of HF in patients with concomitant thyroid pathology (TP) was carried out. The results of our own research were presented. CONCLUSION Conclusions: The final chapter in the history of the role of cytokines in the progression of HF has not yet been written. Further studies, including genetic ones, are necessary. The patients with HF have higher levels of TNFβ and IL-6, and a lower concentration of IL-4, compared to the control group. Patients with a fatal outcome of the disease, in contrast to those who survived for two years, have an increased level of TNFβ. In patients with concomitant TP, who had repeated hospitalization, a lower level was registered, compared to that under conditions of a more favorable course of heart failure. Concentrations of cytokines in the blood of patients with HF are associated with gene polymorphisms of the β-adrenoreceptor system: the C-allele of the Gly389A polymorphism of the β1-adrenoceptor gene leads to a decrease in the risk of increasing TNFα; IL-1α increases in the presence of the A-allele of the Ser49Gly polymorphism of this gene. In patients with HF and concomitant thyroid pathology, the risk of IL-6 growth increases in homozygous (C) patients for the Ser275 polymorphism of the β3 subunit of the G-protein.
Collapse
Affiliation(s)
- Sergiy M Pyvovar
- L.T.MALAYA THERAPY NATIONAL INSTITUTE OF THE NATIONAL ACADEMY OF MEDICAL SCIENCES OF UKRAINE, KHARKIV, UKRAINE
| | - Iurii Rudyk
- L.T.MALAYA THERAPY NATIONAL INSTITUTE OF THE NATIONAL ACADEMY OF MEDICAL SCIENCES OF UKRAINE, KHARKIV, UKRAINE
| | - Tetiana D Scherban
- L.T.MALAYA THERAPY NATIONAL INSTITUTE OF THE NATIONAL ACADEMY OF MEDICAL SCIENCES OF UKRAINE, KHARKIV, UKRAINE
| |
Collapse
|
11
|
Tarcău BM, Vicaș LG, Filip L, Maghiar F, Șandor M, Pallag A, Jurca T, Mureșan ME, Marian E. Emerging Perspectives on the Set of Conditions That Lead to the Emergence of Metabolic Syndrome. J Pers Med 2023; 14:32. [PMID: 38248733 PMCID: PMC10820431 DOI: 10.3390/jpm14010032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 12/18/2023] [Accepted: 12/21/2023] [Indexed: 01/23/2024] Open
Abstract
Metabolic syndrome, as a medical condition, presents multifactorial complexity that is characterized by the resulting damage from genetic, environmental, and lifestyle factors (presence or absence of physical activity, food choices). Thus, metabolic syndrome qualifies unequivocally as a medical condition in which there are, simultaneously, several independent metabolic risk factors, namely, abdominal obesity, high triglyceride level, low HDL cholesterol level, arterial hypertension, and high glycemic level. Although age, sex, socio-economic status, and the precise definition of metabolic syndrome all influence the prevalence and risk of developing the condition, clinical and epidemiological studies clearly show that central obesity, as measured by an increased abdominal circumference, is the main risk factor. Thus, due to the growing global incidence of obesity, there has been an increase in the incidence of metabolic syndrome. Starting with obesity, all other metabolic risk factors are influenced: for example, as a result of insulin resistance with hyperglycemia, diabetes is linked to an increased risk of cardiovascular disease due to increased abdominal circumference. Through this review, we aimed to highlight the latest research studies and dietary nutritional interventions useful in the prevention of this disease but also implementation strategies for primary prevention among the healthy population.
Collapse
Affiliation(s)
- Bogdan M. Tarcău
- Doctoral School of Biomedical Science, University of Oradea, 1 University Street, 410087 Oradea, Romania;
| | - Laura G. Vicaș
- Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, 29 Nicolae Jiga Street, 410028 Oradea, Romania; (A.P.); (T.J.); (E.M.)
| | - Lorena Filip
- Department of Bromatology, Hygiene, Nutrition, Faculty of Pharmacy, Iuliu Hațieganu University of Medicine and Pharmacy, 400349 Cluj-Napoca, Romania;
| | - Florin Maghiar
- Medical Department, Faculty of Medicine and Pharmacy, University of Oradea, 10 1st December Square, 410073 Oradea, Romania;
| | - Mircea Șandor
- Department of Surgical Disciplines, Faculty of Medicine and Pharmacy, University of Oradea, 10 1st December Square, 410073 Oradea, Romania;
| | - Annamaria Pallag
- Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, 29 Nicolae Jiga Street, 410028 Oradea, Romania; (A.P.); (T.J.); (E.M.)
| | - Tunde Jurca
- Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, 29 Nicolae Jiga Street, 410028 Oradea, Romania; (A.P.); (T.J.); (E.M.)
| | - Mariana Eugenia Mureșan
- Department of Preclinical Discipline, Faculty of Medicine and Pharmacy, University of Oradea, 10 1st December Square, 410073 Oradea, Romania;
| | - Eleonora Marian
- Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, 29 Nicolae Jiga Street, 410028 Oradea, Romania; (A.P.); (T.J.); (E.M.)
| |
Collapse
|
12
|
Schuermans A, Pournamdari AB, Lee J, Bhukar R, Ganesh S, Darosa N, Small AM, Yu Z, Hornsby W, Koyama S, Januzzi JL, Honigberg MC, Natarajan P. Integrative proteomic analyses across common cardiac diseases yield new mechanistic insights and enhanced prediction. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.12.19.23300218. [PMID: 38196601 PMCID: PMC10775327 DOI: 10.1101/2023.12.19.23300218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2024]
Abstract
Cardiac diseases represent common highly morbid conditions for which underlying molecular mechanisms remain incompletely understood. Here, we leveraged 1,459 protein measurements in 44,313 UK Biobank participants to characterize the circulating proteome associated with incident coronary artery disease, heart failure, atrial fibrillation, and aortic stenosis. Multivariable-adjusted Cox regression identified 820 protein-disease associations-including 441 proteins-at Bonferroni-adjusted P <8.6×10 -6 . Cis -Mendelian randomization suggested causal roles that aligned with epidemiological findings for 6% of proteins identified in primary analyses, prioritizing novel therapeutic targets for different cardiac diseases (e.g., interleukin-4 receptor for heart failure and spondin-1 for atrial fibrillation). Interaction analyses identified seven protein-disease associations that differed Bonferroni-significantly by sex. Models incorporating proteomic data (vs. clinical risk factors alone) improved prediction for coronary artery disease, heart failure, and atrial fibrillation. These results lay a foundation for future investigations to uncover novel disease mechanisms and assess the clinical utility of protein-based prevention strategies for cardiac diseases.
Collapse
|
13
|
Scott TE, Lewis CV, Zhu M, Wang C, Samuel CS, Drummond GR, Kemp-Harper BK. IL-4 and IL-13 induce equivalent expression of traditional M2 markers and modulation of reactive oxygen species in human macrophages. Sci Rep 2023; 13:19589. [PMID: 37949903 PMCID: PMC10638413 DOI: 10.1038/s41598-023-46237-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Accepted: 10/30/2023] [Indexed: 11/12/2023] Open
Abstract
In cardiovascular disease, pathological and protective roles are reported for the Th2 cytokines IL-4 and IL-13, respectively. We hypothesised that differential effects on macrophage function are responsible. Type I and II receptor subunit (IL-2Rγ, IL-4Rα and IL-13Rα1) and M2 marker (MRC-1, CCL18, CCL22) expression was assessed via RT-qPCR in IL-4- and IL-13-treated human primary macrophages. Downstream signalling was evaluated via STAT1, STAT3 and STAT6 inhibitors, and IL-4- and IL-13-induced reactive oxygen species (ROS) generation assessed. IL-4 and IL-13 exhibited equivalent potency and efficacy for M2 marker induction, which was attenuated by STAT3 inhibition. Both cytokines enhanced PDBu-stimulated superoxide generation however this effect was 17% greater with IL-4 treatment. Type I IL-4 receptor expression was increased on M1-like macrophages but did not lead to a differing ability of these cytokines to modulate M1-like macrophage superoxide production. Overall, this study did not identify major differences in the ability of IL-4 and IL-13 to modulate macrophage function, suggesting that the opposing roles of these cytokines in cardiovascular disease are likely to be via actions on other cell types. Future studies should directly compare IL-4 and IL-13 in vivo to more thoroughly investigate the therapeutic validity of selective targeting of these cytokines.
Collapse
Affiliation(s)
- Tara E Scott
- Cardiovascular Disease Program, Department of Pharmacology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Caitlin V Lewis
- Cardiovascular Disease Program, Department of Pharmacology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Mingyu Zhu
- Cardiovascular Disease Program, Department of Pharmacology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Chao Wang
- Cardiovascular Disease Program, Department of Pharmacology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Chrishan S Samuel
- Cardiovascular Disease Program, Department of Pharmacology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Grant R Drummond
- Department of Physiology, Anatomy and Microbiology, School of Life Sciences, La Trobe University, Melbourne, VIC, Australia
| | - Barbara K Kemp-Harper
- Cardiovascular Disease Program, Department of Pharmacology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia.
| |
Collapse
|
14
|
Song P, Li P, Huang Z, Yuan Y, Wei M, Wang C, Zhang G, Ji M, Guan H. Pro-Fibrotic Role of Interleukin-4 in Influencing Idiopathic Epiretinal Membrane in Cataract Patients: Analysis From Clinical-Experimental Approaches. Transl Vis Sci Technol 2023; 12:23. [PMID: 37982769 PMCID: PMC10668627 DOI: 10.1167/tvst.12.11.23] [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: 04/26/2023] [Accepted: 10/09/2023] [Indexed: 11/21/2023] Open
Abstract
Purpose To evaluate the role of interleukin-4 in influencing idiopathic epiretinal membrane (iERM) formation and early progression post cataract surgery (PCS) from clinical and experimental perspectives. Methods We quantified levels of IL-4 in aqueous humor (AH) samples from 22 iERM patients and 31 control subjects collected before and 20 hours after cataract surgeries using ELISA. After a 3-month follow-up, the association between IL-4 levels and iERM progression measurements was identified. In addition, in vitro studies were conducted to investigate the effects of IL-4 on primary rat retinal Müller glia proliferation, migration, and glial-mesenchymal transition (GMT). Results Concentrations of IL-4 were significantly higher in preoperative AH samples from iERM patients versus controls (P = 0.006). Postoperatively, although IL-4 levels were elevated in both groups compared to their respective preoperative levels, they were even more obviously so in the iERM group (P < 0.001). Multivariate linear regression analyses revealed that, postoperatively, IL-4 level elevation was positively associated with macular volume and thickness increase (both P < 0.05) in iERM patients. However, no correlations were observed between IL-4 level (changes) and clinical characters in the controls. In vitro studies demonstrated that IL-4 promoted Müller glia proliferation and migration and increased the expression of GMT-related markers in a manner independent of transforming growth factor-β1 (TGF-β1). Conclusions IL-4 plays a crucial pro-fibrotic role in iERM formation and early progression 3 months PCS possibly by stimulating Müller glia proliferation, migration, and GMT in a TGF-β1-independent manner. Translational Relevance The current study suggests the potential of IL-4 as a novel therapeutic target for iERM.
Collapse
Affiliation(s)
- Pei Song
- Eye Institute, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
- Department of Ophthalmology, Shanghai Fourth People's Hospital Affiliated to Tongji University, Shanghai, China
| | - Pengfei Li
- Eye Institute, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
- Medical School of Nantong University, Nantong, Jiangsu, China
| | - Zeyu Huang
- Eye Institute, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
| | - Yurong Yuan
- Eye Institute, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
| | - Miao Wei
- Eye Institute, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
| | - Congyu Wang
- Eye Institute, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
- Medical School of Nantong University, Nantong, Jiangsu, China
| | - Guowei Zhang
- Eye Institute, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
| | - Min Ji
- Eye Institute, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
| | - Huaijin Guan
- Eye Institute, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
| |
Collapse
|
15
|
Wu Y, Zhan S, Chen L, Sun M, Li M, Mou X, Zhang Z, Xu L, Xu Y. TNFSF14/LIGHT promotes cardiac fibrosis and atrial fibrillation vulnerability via PI3Kγ/SGK1 pathway-dependent M2 macrophage polarisation. J Transl Med 2023; 21:544. [PMID: 37580750 PMCID: PMC10424430 DOI: 10.1186/s12967-023-04381-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 07/21/2023] [Indexed: 08/16/2023] Open
Abstract
BACKGROUND Tumour necrosis factor superfamily protein 14 (TNFSF14), also called LIGHT, is an important regulator of immunological and fibrosis diseases. However, its specific involvement in cardiac fibrosis and atrial fibrillation (AF) has not been fully elucidated. The objective of this study is to examine the influence of LIGHT on the development of myocardial fibrosis and AF. METHODS PCR arrays of peripheral blood mononuclear cells (PBMCs) from patients with AF and sinus rhythm was used to identify the dominant differentially expressed genes, followed by ELISA to evaluate its serum protein levels. Morphological, functional, and electrophysiological changes in the heart were detected in vivo after the tail intravenous injection of recombinant LIGHT (rLIGHT) in mice for 4 weeks. rLIGHT was used to stimulate bone marrow-derived macrophages (BMDMs) to prepare a macrophage-conditioned medium (MCM) in vitro. Then, the MCM was used to culture mouse cardiac fibroblasts (CFs). The expression of relevant proteins and genes was determined using qRT-PCR, western blotting, and immunostaining. RESULTS The mRNA levels of LIGHT and TNFRSF14 were higher in the PBMCs of patients with AF than in those of the healthy controls. Additionally, the serum protein levels of LIGHT were higher in patients with AF than those in the healthy controls and were correlated with left atrial reverse remodelling. Furthermore, we demonstrated that rLIGHT injection promoted macrophage infiltration and M2 polarisation in the heart, in addition to promoting atrial fibrosis and AF inducibility in vivo, as detected with MASSON staining and atrial burst pacing respectively. RNA sequencing of heart samples revealed that the PI3Kγ/SGK1 pathway may participate in these pathological processes. Therefore, we confirmed the hypothesis that rLIGHT promotes BMDM M2 polarisation and TGB-β1 secretion, and that this process can be inhibited by PI3Kγ and SGK1 inhibitors in vitro. Meanwhile, increased collagen synthesis and myofibroblast transition were observed in LIGHT-stimulated MCM-cultured CFs and were ameliorated in the groups treated with PI3Kγ and SGK1 inhibitors. CONCLUSION LIGHT protein levels in peripheral blood can be used as a prognostic marker for AF and to evaluate its severity. LIGHT promotes cardiac fibrosis and AF inducibility by promoting macrophage M2 polarisation, wherein PI3Kγ and SGK1 activation is indispensable.
Collapse
Affiliation(s)
- Yirong Wu
- Department of Cardiology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Zhejiang, 310006, China
| | - Siyao Zhan
- Department of Cardiology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Zhejiang, 310006, China
| | - Lian Chen
- Department of Cardiology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Zhejiang, 310006, China
| | - Mingrui Sun
- Department of Cardiology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Zhejiang, 310006, China
| | - Miaofu Li
- Department of Cardiology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Zhejiang, 310006, China
| | - Xuanting Mou
- Department of Cardiology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Zhejiang, 310006, China
| | - Zhen Zhang
- Department of Orthopedics, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Zhejiang, 310006, China
| | - Linhao Xu
- Department of Cardiology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Zhejiang, 310006, China.
- Translational Medicine Research Center, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Zhejiang, 310006, China.
| | - Yizhou Xu
- Department of Cardiology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Zhejiang, 310006, China.
| |
Collapse
|
16
|
Abstract
PURPOSE OF REVIEW The intricate interplay between inflammatory and reparative responses in the context of heart injury is central to the pathogenesis of heart failure. Recent clinical studies have shown the therapeutic benefits of anti-inflammatory strategies in the treatment of cardiovascular diseases. This review provides a comprehensive overview of the cross-talk between immune cells and fibroblasts in the diseased heart. RECENT FINDINGS The role of inflammatory cells in fibroblast activation after cardiac injury is well-documented, but recent single-cell transcriptomics studies have identified putative pro-inflammatory fibroblasts in the infarcted heart, suggesting that fibroblasts, in turn, can modify inflammatory cell behavior. Furthermore, anti-inflammatory immune cells and fibroblasts have been described. The use of spatial and temporal-omics analyses may provide additional insights toward a better understanding of disease-specific microenvironments, where activated fibroblasts and inflammatory cells are in proximity. Recent studies focused on the interplay between fibroblasts and immune cells have brought us closer to the identification of cell type-specific targets for intervention. Further exploration of these intercellular communications will provide deeper insights toward the development of novel therapeutics.
Collapse
Affiliation(s)
- Akitoshi Hara
- Center for Cardiovascular Research, University of Hawaii at Manoa, Honolulu, HI, 96825, USA.
| | - Michelle D Tallquist
- Center for Cardiovascular Research, University of Hawaii at Manoa, Honolulu, HI, 96825, USA
| |
Collapse
|
17
|
Liu Y, Zhang D, Yin D. Pathophysiological Effects of Various Interleukins on Primary Cell Types in Common Heart Disease. Int J Mol Sci 2023; 24:ijms24076497. [PMID: 37047468 PMCID: PMC10095356 DOI: 10.3390/ijms24076497] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 03/22/2023] [Accepted: 03/25/2023] [Indexed: 03/31/2023] Open
Abstract
Myocardial infarction (MI), heart failure, cardiomyopathy, myocarditis, and myocardial ischemia-reperfusion injury (I/R) are the most common heart diseases, yet there is currently no effective therapy due to their complex pathogenesis. Cardiomyocytes (CMs), fibroblasts (FBs), endothelial cells (ECs), and immune cells are the primary cell types involved in heart disorders, and, thus, targeting a specific cell type for the treatment of heart disease may be more effective. The same interleukin may have various effects on different kinds of cell types in heart disease, yet the exact role of interleukins and their pathophysiological pathways on primary cell types remain largely unexplored. This review will focus on the pathophysiological effects of various interleukins including the IL-1 family (IL-1, IL-18, IL-33, IL-37), IL-2, IL-4, the IL-6 family (IL-6 and IL-11), IL-8, IL-10, IL-17 on primary cell types in common heart disease, which may contribute to the more precise and effective treatment of heart disease.
Collapse
Affiliation(s)
- Yong Liu
- State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Science, Hubei University, Wuhan 430062, China
- Hubei Province Key Laboratory of Biotechnology of Chinese Traditional Medicine, National & Local Joint Engineering Research Center of High-Throughput Drug Screening Technology, Hubei University, Wuhan 430062, China
| | - Donghui Zhang
- State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Science, Hubei University, Wuhan 430062, China
- Hubei Province Key Laboratory of Biotechnology of Chinese Traditional Medicine, National & Local Joint Engineering Research Center of High-Throughput Drug Screening Technology, Hubei University, Wuhan 430062, China
- Correspondence: (D.Z.); (D.Y.)
| | - Dan Yin
- State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Science, Hubei University, Wuhan 430062, China
- Hubei Province Key Laboratory of Biotechnology of Chinese Traditional Medicine, National & Local Joint Engineering Research Center of High-Throughput Drug Screening Technology, Hubei University, Wuhan 430062, China
- Correspondence: (D.Z.); (D.Y.)
| |
Collapse
|
18
|
Yang C, Li J, Deng Z, Luo S, Liu J, Fang W, Liu F, Liu T, Zhang X, Zhang Y, Meng Z, Zhang S, Luo J, Liu C, Yang D, Liu L, Sukhova GK, Sadybekov A, Katritch V, Libby P, Wang J, Guo J, Shi GP. Eosinophils protect pressure overload- and β-adrenoreceptor agonist-induced cardiac hypertrophy. Cardiovasc Res 2023; 119:195-212. [PMID: 35394031 PMCID: PMC10022866 DOI: 10.1093/cvr/cvac060] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2021] [Revised: 02/01/2022] [Accepted: 03/23/2022] [Indexed: 11/12/2022] Open
Abstract
AIMS Blood eosinophil (EOS) counts and EOS cationic protein (ECP) levels associate positively with major cardiovascular disease (CVD) risk factors and prevalence. This study investigates the role of EOS in cardiac hypertrophy. METHODS AND RESULTS A retrospective cross-section study of 644 consecutive inpatients with hypertension examined the association between blood EOS counts and cardiac hypertrophy. Pressure overload- and β-adrenoreceptor agonist isoproterenol-induced cardiac hypertrophy was produced in EOS-deficient ΔdblGATA mice. This study revealed positive correlations between blood EOS counts and left ventricular (LV) mass and mass index in humans. ΔdblGATA mice showed exacerbated cardiac hypertrophy and dysfunction, with increased LV wall thickness, reduced LV internal diameter, and increased myocardial cell size, death, and fibrosis. Repopulation of EOS from wild-type (WT) mice, but not those from IL4-deficient mice ameliorated cardiac hypertrophy and cardiac dysfunctions. In ΔdblGATA and WT mice, administration of ECP mEar1 improved cardiac hypertrophy and function. Mechanistic studies demonstrated that EOS expression of IL4, IL13, and mEar1 was essential to control mouse cardiomyocyte hypertrophy and death and cardiac fibroblast TGF-β signalling and fibrotic protein synthesis. The use of human cardiac cells yielded the same results. Human ECP, EOS-derived neurotoxin, human EOS, or murine recombinant mEar1 reduced human cardiomyocyte death and hypertrophy and human cardiac fibroblast TGF-β signalling. CONCLUSION Although blood EOS counts correlated positively with LV mass or LV mass index in humans, this study established a cardioprotective role for EOS IL4 and cationic proteins in cardiac hypertrophy and tested a therapeutic possibility of ECPs in this human CVD.
Collapse
Affiliation(s)
| | | | | | | | | | - Wenqian Fang
- Department of Medicine, Cardiovascular Medicine, Brigham and Women’s Hospital and Harvard Medical School, 77 Avenue Louis Pasteur, NRB-7, Boston, MA 02115, USA
| | - Feng Liu
- Department of Geriatrics, National Key Clinical Specialty, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou 510000, China
| | - Tianxiao Liu
- Department of Medicine, Cardiovascular Medicine, Brigham and Women’s Hospital and Harvard Medical School, 77 Avenue Louis Pasteur, NRB-7, Boston, MA 02115, USA
| | - Xian Zhang
- Department of Medicine, Cardiovascular Medicine, Brigham and Women’s Hospital and Harvard Medical School, 77 Avenue Louis Pasteur, NRB-7, Boston, MA 02115, USA
| | - Yuanyuan Zhang
- Department of Medicine, Cardiovascular Medicine, Brigham and Women’s Hospital and Harvard Medical School, 77 Avenue Louis Pasteur, NRB-7, Boston, MA 02115, USA
- Hainan Provincial Key Laboratory for Tropical Cardiovascular Diseases Research & Key Laboratory of Emergency and Trauma of Ministry of Education, Institute of Cardiovascular Research of the First Affiliated Hospital, Hainan Medical University, Haikou 571199, China
| | - Zhaojie Meng
- Department of Medicine, Cardiovascular Medicine, Brigham and Women’s Hospital and Harvard Medical School, 77 Avenue Louis Pasteur, NRB-7, Boston, MA 02115, USA
| | - Shuya Zhang
- Department of Medicine, Cardiovascular Medicine, Brigham and Women’s Hospital and Harvard Medical School, 77 Avenue Louis Pasteur, NRB-7, Boston, MA 02115, USA
- Hainan Provincial Key Laboratory for Tropical Cardiovascular Diseases Research & Key Laboratory of Emergency and Trauma of Ministry of Education, Institute of Cardiovascular Research of the First Affiliated Hospital, Hainan Medical University, Haikou 571199, China
| | - Jianfang Luo
- Department of Cardiology, Vascular Center, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangzhou 510000, China
| | - Conglin Liu
- Department of Medicine, Cardiovascular Medicine, Brigham and Women’s Hospital and Harvard Medical School, 77 Avenue Louis Pasteur, NRB-7, Boston, MA 02115, USA
| | - Dafeng Yang
- Department of Medicine, Cardiovascular Medicine, Brigham and Women’s Hospital and Harvard Medical School, 77 Avenue Louis Pasteur, NRB-7, Boston, MA 02115, USA
| | - Lijun Liu
- Department of Biochemistry and Cancer Biology, College of Medicine and Life Sciences, University of Toledo, Toledo, OH 43614, USA
| | - Galina K Sukhova
- Department of Medicine, Cardiovascular Medicine, Brigham and Women’s Hospital and Harvard Medical School, 77 Avenue Louis Pasteur, NRB-7, Boston, MA 02115, USA
| | - Anastasiia Sadybekov
- Department of Chemistry, Bridge Institute, USC Michelson Center for Convergent Biosciences, University of Southern California, Los Angeles, CA 90089, USA
- Department of Biological Sciences, Bridge Institute, USC Michelson Center for Convergent Biosciences, University of Southern California, Los Angeles, CA 90089, USA
| | - Vsevolod Katritch
- Department of Chemistry, Bridge Institute, USC Michelson Center for Convergent Biosciences, University of Southern California, Los Angeles, CA 90089, USA
- Department of Biological Sciences, Bridge Institute, USC Michelson Center for Convergent Biosciences, University of Southern California, Los Angeles, CA 90089, USA
| | - Peter Libby
- Department of Medicine, Cardiovascular Medicine, Brigham and Women’s Hospital and Harvard Medical School, 77 Avenue Louis Pasteur, NRB-7, Boston, MA 02115, USA
| | - Jing Wang
- Corresponding authors. Tel: +1 617 525 4358, E-mail: (G.-P.S.); Tel: +86 10 6915 6477, E-mail: (J.W.); Tel: +86 1868983 5101, E-mail: (J.G.)
| | - Junli Guo
- Corresponding authors. Tel: +1 617 525 4358, E-mail: (G.-P.S.); Tel: +86 10 6915 6477, E-mail: (J.W.); Tel: +86 1868983 5101, E-mail: (J.G.)
| | - Guo-Ping Shi
- Corresponding authors. Tel: +1 617 525 4358, E-mail: (G.-P.S.); Tel: +86 10 6915 6477, E-mail: (J.W.); Tel: +86 1868983 5101, E-mail: (J.G.)
| |
Collapse
|
19
|
Singla A, Reuter S, Taube C, Peters M, Peters K. The molecular mechanisms of remodeling in asthma, COPD and IPF with a special emphasis on the complex role of Wnt5A. Inflamm Res 2023; 72:577-588. [PMID: 36658268 PMCID: PMC10023767 DOI: 10.1007/s00011-023-01692-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 11/28/2022] [Accepted: 01/09/2023] [Indexed: 01/20/2023] Open
Abstract
INTRODUCTION Chronic inflammatory lung diseases are a common cause of suffering and death. Chronic obstructive pulmonary disease (COPD) is the reason for 6% of all deaths worldwide. A total of 262 million people are affected by asthma and 461,000 people died in 2019. Idiopathic pulmonary fibrosis (IPF) is diagnosed in 3 million people worldwide, with an onset over the age of 50 with a mean survival of only 24-30 months. These three diseases have in common that remodeling of the lung tissue takes place, which is responsible for an irreversible decline of lung function. Pathological lung remodeling is mediated by a complex interaction of different, often misguided, repair processes regulated by a variety of mediators. One group of these, as has recently become known, are the Wnt ligands. In addition to their well-characterized role in embryogenesis, this group of glycoproteins is also involved in immunological and structural repair processes. Depending on the combination of the Wnt ligand with its receptors and co-receptors, canonical and noncanonical signaling cascades can be induced. Wnt5A is a mediator that is described mainly in noncanonical Wnt signaling and has been shown to play an important role in different inflammatory diseases and malignancies. OBJECTIVES In this review, we summarize the literature available regarding the role of Wnt5A as an immune modulator and its role in the development of asthma, COPD and IPF. We will focus specifically on what is known about Wnt5A concerning its role in the remodeling processes involved in the chronification of the diseases. CONCLUSION Wnt5A has been shown to be involved in all three inflammatory lung diseases. Since the ligand affects both structural and immunological processes, it is an interesting target for the treatment of lung diseases whose pathology involves a restructuring of the lung tissue triggered in part by an inflammatory immune response.
Collapse
Affiliation(s)
- Abhinav Singla
- Department of Pulmonary Medicine, University Medical Center Essen-Ruhrlandklinik, Essen, Germany
- Department of Molecular Immunology, Ruhr-University Bochum, Universitätsstraße 150, 44801, Bochum, Germany
| | - Sebastian Reuter
- Department of Pulmonary Medicine, University Medical Center Essen-Ruhrlandklinik, Essen, Germany
| | - Christian Taube
- Department of Pulmonary Medicine, University Medical Center Essen-Ruhrlandklinik, Essen, Germany
| | - Marcus Peters
- Department of Molecular Immunology, Ruhr-University Bochum, Universitätsstraße 150, 44801, Bochum, Germany.
| | - Karin Peters
- Department of Molecular Immunology, Ruhr-University Bochum, Universitätsstraße 150, 44801, Bochum, Germany
| |
Collapse
|
20
|
Frąk W, Wojtasińska A, Lisińska W, Młynarska E, Franczyk B, Rysz J. Pathophysiology of Cardiovascular Diseases: New Insights into Molecular Mechanisms of Atherosclerosis, Arterial Hypertension, and Coronary Artery Disease. Biomedicines 2022; 10:biomedicines10081938. [PMID: 36009488 PMCID: PMC9405799 DOI: 10.3390/biomedicines10081938] [Citation(s) in RCA: 77] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 08/04/2022] [Accepted: 08/06/2022] [Indexed: 12/12/2022] Open
Abstract
Cardiovascular diseases (CVDs) are disorders associated with the heart and circulatory system. Atherosclerosis is its major underlying cause. CVDs are chronic and can remain hidden for a long time. Moreover, CVDs are the leading cause of global morbidity and mortality, thus creating a major public health concern. This review summarizes the available information on the pathophysiological implications of CVDs, focusing on coronary artery disease along with atherosclerosis as its major cause and arterial hypertension. We discuss the endothelium dysfunction, inflammatory factors, and oxidation associated with atherosclerosis. Mechanisms such as dysfunction of the endothelium and inflammation, which have been identified as critical pathways for development of coronary artery disease, have become easier to diagnose in recent years. Relatively recently, evidence has been found indicating that interactions of the molecular and cellular elements such as matrix metalloproteinases, elements of the immune system, and oxidative stress are involved in the pathophysiology of arterial hypertension. Many studies have revealed several important inflammatory and genetic risk factors associated with CVDs. However, further investigation is crucial to improve our knowledge of CVDs progression and, more importantly, accelerate basic research to improve our understanding of the mechanism of pathophysiology.
Collapse
|
21
|
Zhang Y, Ye Y, Tang X, Wang H, Tanaka T, Tian R, Yang X, Wang L, Xiao Y, Hu X, Jin Y, Pang H, Du T, Liu H, Sun L, Xiao S, Dong R, Ferrucci L, Tian Z, Zhang S. CCL17 acts as a novel therapeutic target in pathological cardiac hypertrophy and heart failure. J Exp Med 2022; 219:213274. [PMID: 35687056 PMCID: PMC9194836 DOI: 10.1084/jem.20200418] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 04/22/2022] [Accepted: 06/01/2022] [Indexed: 11/18/2022] Open
Abstract
Circulating proteomic signatures of age are closely associated with aging and age-related diseases; however, the utility of changes in secreted proteins in identifying therapeutic targets for diseases remains unclear. Serum proteomic profiling of an age-stratified healthy population and further community-based cohort together with heart failure patients study demonstrated that circulating C-C motif chemokine ligand 17 (CCL17) level increased with age and correlated with cardiac dysfunction. Subsequent animal experiments further revealed that Ccll7-KO significantly repressed aging and angiotensin II (Ang II)–induced cardiac hypertrophy and fibrosis, accompanied by the plasticity and differentiation of T cell subsets. Furthermore, the therapeutic administration of an anti-CCL17 neutralizing antibody inhibited Ang II–induced pathological cardiac remodeling. Our findings reveal that chemokine CCL17 is identifiable as a novel therapeutic target in age-related and Ang II–induced pathological cardiac hypertrophy and heart failure.
Collapse
Affiliation(s)
- Yang Zhang
- Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yicong Ye
- Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiaoqiang Tang
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of Ministry of Education, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Hui Wang
- Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Toshiko Tanaka
- Longitudinal Studies Section, Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, MD
| | - Ran Tian
- Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xufei Yang
- Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Lun Wang
- Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ying Xiao
- Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiaomin Hu
- Department of Central Research Laboratory, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ye Jin
- Department of Central Research Laboratory, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Haiyu Pang
- Department of Central Research Laboratory, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Tian Du
- Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Honghong Liu
- Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Lihong Sun
- State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Shuo Xiao
- Thermo Fisher Scientific (China) Co., Ltd, Changning, Shanghai, China
| | - Ruijia Dong
- Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Luigi Ferrucci
- Longitudinal Studies Section, Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, MD
| | - Zhuang Tian
- Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Shuyang Zhang
- Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,Department of Central Research Laboratory, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
22
|
Cai S, Hu Z, Chen Y, Zhong J, Dong L. Potential roles of non-lymphocytic cells in the pathogenesis of IgG4-related disease. Front Immunol 2022; 13:940581. [PMID: 35967331 PMCID: PMC9366038 DOI: 10.3389/fimmu.2022.940581] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Accepted: 07/04/2022] [Indexed: 11/18/2022] Open
Abstract
Studies have confirmed the involvement of a variety of lymphocyte subsets, including type 2 helper T lymphocytes (Th2) and IgG4+ B lymphocytes, in the pathogenesis of IgG4-related disease (IgG4-RD). Those lymphocytes contribute to the major pathogenetic features of IgG4-RD. However, they are not the only cellular components in the immunoinflammatory environment of this mysterious disease entity. Recent studies have suggested that various non-lymphocytic components, including macrophages and fibroblasts, may also play an important role in the pathogenetic process of IgG4-RD in terms of contributing to the chronic and complex progress of the disease. Therefore, the potential role of non-lymphocyte in the pathogenesis of IgG4-RD is worth discussing.
Collapse
Affiliation(s)
| | | | - Yu Chen
- *Correspondence: Lingli Dong, ; Jixin Zhong, ; Yu Chen,
| | - Jixin Zhong
- *Correspondence: Lingli Dong, ; Jixin Zhong, ; Yu Chen,
| | - Lingli Dong
- *Correspondence: Lingli Dong, ; Jixin Zhong, ; Yu Chen,
| |
Collapse
|
23
|
Hershberger RE, Zareba KM. Dilated Cardiomyopathy: New Distinct Phenotypes or Temporal Phases of Disease? J Am Coll Cardiol 2022; 79:2233-2235. [PMID: 35654494 PMCID: PMC11026084 DOI: 10.1016/j.jacc.2022.04.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 04/13/2022] [Accepted: 04/13/2022] [Indexed: 10/18/2022]
Affiliation(s)
- Ray E Hershberger
- Division of Human Genetics, Department of Internal Medicine, Ohio State University Wexner Medical Center, Columbus, Ohio, USA; Division of Cardiovascular Medicine, Department of Internal Medicine, Ohio State University Wexner Medical Center, Columbus, Ohio, USA; Dorothy M. Davis Heart and Lung Research Institute, Ohio State University Wexner Medical Center, Columbus, Ohio, USA.
| | - Karolina M Zareba
- Division of Cardiovascular Medicine, Department of Internal Medicine, Ohio State University Wexner Medical Center, Columbus, Ohio, USA; Dorothy M. Davis Heart and Lung Research Institute, Ohio State University Wexner Medical Center, Columbus, Ohio, USA. https://twitter.com/ZarebaCMR
| |
Collapse
|
24
|
Geng C, Feng Y, Yang Y, Yang H, Li Z, Tang Y, Wang J, Zhao H. Allergic asthma aggravates angiotensin Ⅱ-induced cardiac remodeling in mice. Transl Res 2022; 244:88-100. [PMID: 35108660 DOI: 10.1016/j.trsl.2022.01.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 01/13/2022] [Accepted: 01/24/2022] [Indexed: 10/19/2022]
Abstract
Cardiovascular disease remains the leading cause of death globally, and heart failure (HF) represents its terminal stage. Asthma, one of the most common chronic diseases, has been reported to be associated with an increased risk of cardiovascular disease. However, the link between asthma and HF has rarely been studied, and the possible mechanisms by which asthma affects HF are unclear. This study aimed to explore the influence of asthma on HF and the possible mechanisms. We analyzed data from the National Health and Nutrition Examination Survey and found a higher prevalence of HF among asthmatic individuals, and identified an independent association between HF and asthma. Subsequently, we produced mice with concurrent ovalbumin (OVA) sensitization-induced allergic asthma and angiotensin Ⅱ infusion-induced cardiac remodeling to explore the effect of asthma on cardiac remodeling in vivo. The results showed that OVA-induced asthma impaired heart function and aggravated cardiac remodeling in mice. We also found that OVA sensitization increased the expression levels of immunoglobulin E (IgE) in serum and IgE receptor (FcεR1) in the heart, and enhanced the activation of downstream signaling molecules of IgE-FcεR1 in the heart. Importantly, blockage of IgE-FcεR1 using FcεR1-deficient mice or an anti-IgE antibody prevented asthma-induced decline of cardiac function, and alleviated cardiac remodeling. These findings demonstrate the adverse effects of allergic asthma on the heart, and suggest the potential application of anti-IgE therapy in the treatment of asthma complicated with heart conditions.
Collapse
Key Words
- AKT, protein kinase B
- ANP, natriuretic peptide type A
- Ang Ⅱ, angiotensin Ⅱ
- BALF, bronchioalveolar lavage fluid
- BMI, body mass index
- BNP, natriuretic peptide type B
- BW, body weight
- CAD, coronary heart disease
- COPD, chronic obstructive pulmonary disease
- CVD, cardiovascular disease
- EF, ejection fraction
- FS, fraction shortening
- HF, heart failure
- HW, heart weight
- IgE, immunoglobulin E
- LVAW, left ventricular anterior wall
- LVID, left ventricular internal dimension
- LVPW, left ventricular posterior wall
- NHANES, National Health and Nutrition Examination Survey
- OVA, ovalbumin
- TC, total cholesterol
- TG, triglyceride
- WGA, wheat germ agglutinin
- WT, wild type
- pSmad2/3, phosphorylated small mothers against decapentaplegic 2 and 3
- α-SMA, α-smooth muscle actin
- β-MHC, β-myosin heavy chain
Collapse
Affiliation(s)
- Chi Geng
- Department of Pathophysiology, State Key Laboratory of Medical Molecular Biology Institute of Basic Medicine, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China
| | - Yufan Feng
- Department of Pathophysiology, State Key Laboratory of Medical Molecular Biology Institute of Basic Medicine, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China
| | - Yang Yang
- Department of Pathophysiology, State Key Laboratory of Medical Molecular Biology Institute of Basic Medicine, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China
| | - Hongqin Yang
- Department of Pathophysiology, State Key Laboratory of Medical Molecular Biology Institute of Basic Medicine, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China
| | - Zhiwei Li
- Department of Pathophysiology, State Key Laboratory of Medical Molecular Biology Institute of Basic Medicine, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China
| | - Yaqin Tang
- Department of Pathophysiology, State Key Laboratory of Medical Molecular Biology Institute of Basic Medicine, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China
| | - Jing Wang
- Department of Pathophysiology, State Key Laboratory of Medical Molecular Biology Institute of Basic Medicine, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China.
| | - Hongmei Zhao
- Department of Pathophysiology, State Key Laboratory of Medical Molecular Biology Institute of Basic Medicine, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China.
| |
Collapse
|
25
|
Besse S, Nadaud S, Balse E, Pavoine C. Early Protective Role of Inflammation in Cardiac Remodeling and Heart Failure: Focus on TNFα and Resident Macrophages. Cells 2022; 11:1249. [PMID: 35406812 PMCID: PMC8998130 DOI: 10.3390/cells11071249] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 02/24/2022] [Accepted: 04/01/2022] [Indexed: 12/13/2022] Open
Abstract
Cardiac hypertrophy, initiated by a variety of physiological or pathological stimuli (hemodynamic or hormonal stimulation or infarction), is a critical early adaptive compensatory response of the heart. The structural basis of the progression from compensated hypertrophy to pathological hypertrophy and heart failure is still largely unknown. In most cases, early activation of an inflammatory program reflects a reparative or protective response to other primary injurious processes. Later on, regardless of the underlying etiology, heart failure is always associated with both local and systemic activation of inflammatory signaling cascades. Cardiac macrophages are nodal regulators of inflammation. Resident macrophages mostly attenuate cardiac injury by secreting cytoprotective factors (cytokines, chemokines, and growth factors), scavenging damaged cells or mitochondrial debris, and regulating cardiac conduction, angiogenesis, lymphangiogenesis, and fibrosis. In contrast, excessive recruitment of monocyte-derived inflammatory macrophages largely contributes to the transition to heart failure. The current review examines the ambivalent role of inflammation (mainly TNFα-related) and cardiac macrophages (Mφ) in pathophysiologies from non-infarction origin, focusing on the protective signaling processes. Our objective is to illustrate how harnessing this knowledge could pave the way for innovative therapeutics in patients with heart failure.
Collapse
Affiliation(s)
| | | | | | - Catherine Pavoine
- INSERM, Institute of Cardiometabolism and Nutrition (ICAN), Sorbonne Université, UMR_S1166, F-75013 Paris, France; (S.B.); (S.N.); (E.B.)
| |
Collapse
|
26
|
Bowers SL, Meng Q, Molkentin JD. Fibroblasts orchestrate cellular crosstalk in the heart through the ECM. NATURE CARDIOVASCULAR RESEARCH 2022; 1:312-321. [PMID: 38765890 PMCID: PMC11101212 DOI: 10.1038/s44161-022-00043-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 03/02/2022] [Indexed: 05/22/2024]
Abstract
Cell communication is needed for organ function and stress responses, especially in the heart. Cardiac fibroblasts, cardiomyocytes, immune cells, and endothelial cells comprise the major cell types in ventricular myocardium that together coordinate all functional processes. Critical to this cellular network is the non-cellular extracellular matrix (ECM) that provides structure and harbors growth factors and other signaling proteins that affect cell behavior. The ECM is not only produced and modified by cells within the myocardium, largely cardiac fibroblasts, it also acts as an avenue for communication among all myocardial cells. In this Review, we discuss how the development of therapeutics to combat cardiac diseases, specifically fibrosis, relies on a deeper understanding of how the cardiac ECM is intertwined with signaling processes that underlie cellular activation and behavior.
Collapse
Affiliation(s)
| | | | - Jeffery D. Molkentin
- Cincinnati Children’s Hospital, Division of Molecular Cardiovascular Biology; University of Cincinnati, Department of Pediatrics, Cincinnati, OH
| |
Collapse
|
27
|
Melamud MM, Ermakov EA, Boiko AS, Parshukova DA, Sizikov AE, Ivanova SA, Nevinsky GA, Buneva VN. Serum cytokine levels of systemic lupus erythematosus patients in the presence of concomitant cardiovascular diseases. Endocr Metab Immune Disord Drug Targets 2022; 22:852-861. [PMID: 35249511 DOI: 10.2174/1871530322666220304214512] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 12/20/2021] [Accepted: 01/07/2022] [Indexed: 11/22/2022]
Abstract
BACKGROUND Systemic lupus erythematosus (SLE) is known to be associated with an increased risk of cardiovascular diseases (CVD). SLE patients suffer from CVD 3.5 times more often than healthy people. Cytokine-mediated inflammation is actively involved in the development of cardiovascular pathology. OBJECTIVE Here, we analyzed serum levels of nine cytokines of steroids treated SLE patients depending on the presence of concomitant CVD. METHODS The levels of interleukin (IL)-1β, IL-2, IL-4, IL-6, IL-10, IL-21, tumor necrosis factor α (TNFα), B-cell activating factor (BAFF), and a proliferation-inducing ligand (APRIL) were analyzed using multiplex immunoassay. RESULTS In the total group of SLE patients (n=29), the concentrations of IL-6 and IL-10 were higher, and the APRIL level decreased compared with healthy donors (n=39, p<0.05). The same changes were observed in the group of patients without CVD (n=15): the levels of IL-6 and IL-10 increased, and the level of APRIL was lower than in healthy individuals (p<0.05). In the group of SLE patients with CVD (n=14), the concentrations of IL-4, IL-6, IL-10, and TNFα increased (p<0.05). Interestingly, the levels of TNFα and BAFF in SLE patients with CVD were higher than in patients without cardiovascular pathology. Thus, TNFα and BAFF levels were significantly altered in SLE with concomitant CVD compared to SLE without CVD. CONCLUSION These findings suggest that cytokine profiles in SLE with concomitant CVD and SLE without CVD are different, which should be considered in further research with large samples.
Collapse
Affiliation(s)
- Mark M Melamud
- Laboratory of Repair Enzymes, Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - Evgeny A Ermakov
- Laboratory of Repair Enzymes, Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - Anastasiia S Boiko
- Laboratory of Molecular Genetics and Biochemistry, Mental Health Research Institute, Tomsk National Research Medical Center of the Russian Academy of Sciences, Tomsk, Russia
| | - Daria A Parshukova
- Laboratory of Molecular Genetics and Biochemistry, Mental Health Research Institute, Tomsk National Research Medical Center of the Russian Academy of Sciences, Tomsk, Russia
| | - Alexey E Sizikov
- Department of Rheumatology, Institute of Clinical Immunology, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - Svetlana A Ivanova
- Laboratory of Molecular Genetics and Biochemistry, Mental Health Research Institute, Tomsk National Research Medical Center of the Russian Academy of Sciences, Tomsk, Russia
| | - Georgy A Nevinsky
- Laboratory of Repair Enzymes, Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - Valentina N Buneva
- Laboratory of Repair Enzymes, Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| |
Collapse
|
28
|
Abdi A, AlOtaiby S, Badarin FA, Khraibi A, Hamdan H, Nader M. Interaction of SARS-CoV-2 with cardiomyocytes: Insight into the underlying molecular mechanisms of cardiac injury and pharmacotherapy. Biomed Pharmacother 2022; 146:112518. [PMID: 34906770 PMCID: PMC8654598 DOI: 10.1016/j.biopha.2021.112518] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 11/15/2021] [Accepted: 12/06/2021] [Indexed: 01/07/2023] Open
Abstract
SARS-CoV-2 causes respiratory illness with a spectrum of systemic complications. However, the mechanism for cardiac infection and cardiomyocyte injury in COVID-19 patients remains unclear. The current literature supports the notion that SARS-CoV-2 particles access the heart either by the circulating blood cells or by extracellular vesicles, originating from the inflamed lungs, and encapsulating the virus along with its receptor (ACE2). Both cardiomyocytes and pericytes (coronary arteries) express the necessary accessory proteins for access of SARS-CoV-2 particles (i.e. ACE2, NRP-1, TMPRSS2, CD147, integrin α5β1, and CTSB/L). These proteins facilitate the SARS-CoV-2 interaction and entry into the pericytes and cardiomyocytes thus leading to cardiac manifestations. Subsequently, various signaling pathways are altered in the infected cardiomyocytes (i.e. increased ROS production, reduced contraction, impaired calcium homeostasis), causing cardiac dysfunction. The currently adopted pharmacotherapy in severe COVID-19 subjects exhibited side effects on the heart, often manifested by electrical abnormalities. Nonetheless, cardiovascular adverse repercussions have been associated with the advent of some of the SARS-CoV-2 vaccines with no clear mechanisms underlining these complications. We provide herein an overview of the pathways involved with cardiomyocyte in COVID-19 subjects to help promoting pharmacotherapies that can protect against SARS-CoV-2-induced cardiac injuries.
Collapse
Affiliation(s)
- Abdulhamid Abdi
- Department of Physiology and Immunology, College of Medicine and Health Sciences, and Biotechnology Center, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
| | - Shahad AlOtaiby
- Research Center, King Fahad Medical City, Central Second Health Cluster, Ministry of Health, Riyadh, Saudi Arabia
| | - Firas Al Badarin
- Heart and Vascular Institute, Cleveland Clinic Abu Dhabi, Abu Dhabi, United Arab Emirates
| | - Ali Khraibi
- Department of Physiology and Immunology, College of Medicine and Health Sciences, and Biotechnology Center, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
| | - Hamdan Hamdan
- Department of Physiology and Immunology, College of Medicine and Health Sciences, and Biotechnology Center, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
| | - Moni Nader
- Department of Physiology and Immunology, College of Medicine and Health Sciences, and Biotechnology Center, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates.
| |
Collapse
|
29
|
Wade H, Duan Q, Su Q. Interaction between Sars-CoV-2 structural proteins and host cellular receptors: From basic mechanisms to clinical perspectives. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2022; 132:243-277. [PMID: 36088078 PMCID: PMC9182089 DOI: 10.1016/bs.apcsb.2022.05.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Severe acute respiratory syndrome coronavirus 2 (Sars-CoV-2) has caused a global pandemic that has affected the lives of billions of individuals. Sars-CoV-2 primarily infects human cells by binding of the viral spike protein to angiotensin-converting enzyme 2 (ACE2). In addition, novel means of viral entry are currently being investigated, including Neuropillin 1, toll-like receptors (TLRs), cluster of differentiation 147 (CD147), and integrin α5β1. Enriched expression of these proteins across metabolic regulatory organs/tissues, including the circulatory system, liver, pancreas, and intestine contributes to major clinical complications among COVID-19 patients, particularly the development of hypertension, myocardial injury, arrhythmia, acute coronary syndrome and increased coagulation in the circulatory system during and post-infection. Pre-existing metabolic disease, such as cardiovascular disease, obesity, diabetes, and non-alcoholic fatty liver disease, is associated with increased risk of hospitalization, persistent post-infection complications and worse outcomes in patients with COVID-19. This review overviews the biological features of Sars-CoV-2, highlights recent findings that delineate the pathological mechanisms of COVID-19 and the consequent clinical diseases.
Collapse
|
30
|
Paddock SJ, Swift SK, Alencar-Almeida V, Kenarsary A, Alvarez-Argote S, Flinn MA, Patterson M, O'Meara CC. IL4Rα signaling promotes neonatal cardiac regeneration and cardiomyocyte cell cycle activity. J Mol Cell Cardiol 2021; 161:62-74. [PMID: 34343540 PMCID: PMC8629844 DOI: 10.1016/j.yjmcc.2021.07.012] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 07/25/2021] [Accepted: 07/28/2021] [Indexed: 12/31/2022]
Abstract
Neonatal heart regeneration depends on proliferation of pre-existing cardiomyocytes, yet the mechanisms driving regeneration and cardiomyocyte proliferation are not comprehensively understood. We recently reported that the anti-inflammatory cytokine, interleukin 13 (IL13), promotes neonatal cardiac regeneration; however, the signaling pathway and cell types mediating this regenerative response remain unknown. Here, we hypothesized that expression of the type II heterodimer receptor for IL13, comprised of IL4Rα and IL13Rα1, expressed directly on cardiomyocytes mediates cardiomyocyte cell cycle and heart regeneration in neonatal mice. Our data demonstrate that indeed global deletion of one critical subunit of the type II receptor, IL4Rα (IL4Rα-/-), decreases cardiomyocyte proliferation during early postnatal development and significantly impairs cardiac regeneration following injury in neonatal mice. While multiple myocardial cell types express IL4Rα, we demonstrate that IL4Rα deletion specifically in cardiomyocytes mediates cell cycle activity and neonatal cardiac regeneration. This demonstrates for the first time a functional role for IL4Rα signaling directly on cardiomyocytes in vivo. Reciprocally, we examined the therapeutic benefit of activating the IL4Rα receptor in non-regenerative hearts via IL13 administration. Following myocardial infarction, administration of IL13 reduced scar size and promoted cardiomyocyte DNA synthesis and karyokinesis, but not complete cytokinesis, in 6-day old non-regenerative mice. Our data demonstrate a novel role for IL4Rα signaling directly on cardiomyocytes during heart regeneration and suggest the potential for type II receptor activation as one potential therapeutic target for promoting myocardial repair.
Collapse
Affiliation(s)
- Samantha J Paddock
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, United States of America; Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI, United States of America
| | - Samantha K Swift
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI, United States of America; Department of Cell Biology, Neurobiology, and Anatomy, Medical College of Wisconsin, Milwaukee, WI, United States of America
| | - Victor Alencar-Almeida
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, United States of America; Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI, United States of America
| | - Aria Kenarsary
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, United States of America; Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI, United States of America; Department of Cell Biology, Neurobiology, and Anatomy, Medical College of Wisconsin, Milwaukee, WI, United States of America
| | - Santiago Alvarez-Argote
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, United States of America; Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI, United States of America
| | - Michael A Flinn
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, United States of America; Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI, United States of America
| | - Michaela Patterson
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI, United States of America; Department of Cell Biology, Neurobiology, and Anatomy, Medical College of Wisconsin, Milwaukee, WI, United States of America
| | - Caitlin C O'Meara
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, United States of America; Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI, United States of America.
| |
Collapse
|
31
|
Hartmann C, Miggiolaro AFRDS, Motta JDS, Baena Carstens L, Busatta Vaz De Paula C, Fagundes Grobe S, Hermann de Souza Nunes L, Lenci Marques G, Libby P, Zytynski Moura L, de Noronha L, Pellegrino Baena C. The Pathogenesis of COVID-19 Myocardial Injury: An Immunohistochemical Study of Postmortem Biopsies. Front Immunol 2021; 12:748417. [PMID: 34804033 PMCID: PMC8602833 DOI: 10.3389/fimmu.2021.748417] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Accepted: 10/18/2021] [Indexed: 01/19/2023] Open
Abstract
Rationale Myocardial injury associates significantly and independently with mortality in COVID-19 patients. However, the pathogenesis of myocardial injury in COVID-19 remains unclear, and cardiac involvement by SARS-CoV-2 presents a major challenge worldwide. Objective This histological and immunohistochemical study sought to clarify the pathogenesis and propose a mechanism with pathways involved in COVID-19 myocardial injury. Methods and Results Postmortem minimally invasive autopsies were performed in six patients who died from COVID-19, and the myocardium samples were compared to a control group (n=11). Histological analysis was performed using hematoxylin-eosin and toluidine blue staining. Immunohistochemical (IHC) staining was performed using monoclonal antibodies against targets: caspase-1, caspase-9, gasdermin-d, ICAM-1, IL-1β, IL-4, IL-6, CD163, TNF-α, TGF-β, MMP-9, type 1 and type 3 collagen. The samples were also assessed for apoptotic cells by TUNEL. Histological analysis showed severe pericardiocyte interstitial edema and higher mast cells counts per high-power field in all COVID-19 myocardium samples. The IHC analysis showed increased expression of caspase-1, ICAM-1, IL-1β, IL-6, MMP-9, TNF-α, and other markers in the hearts of COVID-19 patients. Expression of caspase-9 did not differ from the controls, while gasdermin-d expression was less. The TUNEL assay was positive in all the COVID-19 samples supporting endothelial apoptosis. Conclusions The pathogenesis of COVID-19 myocardial injury does not seem to relate to primary myocardiocyte involvement but to local inflammation with associated interstitial edema. We found heightened TGF-β and interstitial collagen expression in COVID-affected hearts, a potential harbinger of chronic myocardial fibrosis. These results suggest a need for continued clinical surveillance of patients for myocardial dysfunction and arrythmias after recovery from the acute phase of COVID-19.
Collapse
Affiliation(s)
- Camila Hartmann
- School of Medicine, Pontifícia Universidade Católica do Paraná (PUCPR), Curitiba, Brazil.,Department of Medicine, Marcelino Champagnat Hospital, Curitiba, Brazil
| | | | | | - Lucas Baena Carstens
- School of Medicine, Pontifícia Universidade Católica do Paraná (PUCPR), Curitiba, Brazil
| | | | - Sarah Fagundes Grobe
- School of Medicine, Pontifícia Universidade Católica do Paraná (PUCPR), Curitiba, Brazil.,Department of Medicine, Marcelino Champagnat Hospital, Curitiba, Brazil
| | - Larissa Hermann de Souza Nunes
- School of Medicine, Pontifícia Universidade Católica do Paraná (PUCPR), Curitiba, Brazil.,Department of Medicine, Marcelino Champagnat Hospital, Curitiba, Brazil
| | - Gustavo Lenci Marques
- School of Medicine, Pontifícia Universidade Católica do Paraná (PUCPR), Curitiba, Brazil.,Department of Medicine, Marcelino Champagnat Hospital, Curitiba, Brazil
| | - Peter Libby
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
| | - Lidia Zytynski Moura
- School of Medicine, Pontifícia Universidade Católica do Paraná (PUCPR), Curitiba, Brazil.,Department of Medicine, Marcelino Champagnat Hospital, Curitiba, Brazil
| | - Lucia de Noronha
- School of Medicine, Pontifícia Universidade Católica do Paraná (PUCPR), Curitiba, Brazil
| | - Cristina Pellegrino Baena
- School of Medicine, Pontifícia Universidade Católica do Paraná (PUCPR), Curitiba, Brazil.,Department of Medicine, Marcelino Champagnat Hospital, Curitiba, Brazil
| |
Collapse
|
32
|
Bonnard B, Ibarrola J, Lima-Posada I, Fernández-Celis A, Durand M, Genty M, Lopez-Andreés N, Jaisser F. Neutrophil Gelatinase-Associated Lipocalin From Macrophages Plays a Critical Role in Renal Fibrosis Via the CCL5 (Chemokine Ligand 5)-Th2 Cells-IL4 (Interleukin 4) Pathway. Hypertension 2021; 79:352-364. [PMID: 34794340 DOI: 10.1161/hypertensionaha.121.17712] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
NGAL (neutrophil gelatinase-associated lipocalin; or lipocalin 2, Lcn2) is a novel mineralocorticoid target in the cardiovascular system. We showed that Lcn2 gene invalidation protects against proteinuria and renal injury upon mineralocorticoid excess and we hypothesized that NGAL produced from macrophages promotes the expression of chemoattractant molecules involved these renal lesions. The role of NGAL was analyzed using myeloid-specific (MΦ KO NGAL) Lcn2 knockout mice challenged with uni-nephrectomy, aldosterone, and salt (NAS) for 6 weeks. The role of the CCL5 (chemokine ligand 5) and IL4 (interleukin 4) in kidney fibrosis was studied by administration of the CCL5 receptor antagonist maraviroc or by injections of an anti-IL4 neutralizing antibody. In CTL mice, NAS increased the renal expression of extracellular matrix proteins, such as collagen I, αSMA, and fibronectin associated with interstitial fibrosis which were blunted in MΦ KO NGAL mice. The expression of CCL5 was blunted in sorted macrophages from MΦ KO NGAL mice challenged by NAS and in macrophages obtained from KO NGAL mice and challenged ex vivo with aldosterone and salt. The pharmacological blockade of the CCL5 receptor reduced renal fibrosis and the CD4+ Th cell infiltration induced by NAS. Neutralization of IL4 in NAS mice blunted kidney fibrosis and the overexpression of profibrotic proteins, such as collagen I, αSMA, and fibronectin. In conclusion, NGAL produced by macrophages plays a critical role in renal fibrosis and modulates the CCL5/IL4 pathway in mice exposed to mineralocorticoid excess.
Collapse
Affiliation(s)
- Benjamin Bonnard
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université de Paris, France (B.B., I.L.-P., M.D., M.G., F.J.)
| | - Jaime Ibarrola
- Cardiovascular Translational Research, Navarrabiomed (Miguel Servet Foundation), Instituto de Investigacioón Sanitaria de Navarra (IdiSNA), Pamplona, Spain (J.I., A.F.-C., N.L.-A.)
| | - Ixchel Lima-Posada
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université de Paris, France (B.B., I.L.-P., M.D., M.G., F.J.)
| | - Amaya Fernández-Celis
- Cardiovascular Translational Research, Navarrabiomed (Miguel Servet Foundation), Instituto de Investigacioón Sanitaria de Navarra (IdiSNA), Pamplona, Spain (J.I., A.F.-C., N.L.-A.)
| | - Manon Durand
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université de Paris, France (B.B., I.L.-P., M.D., M.G., F.J.)
| | - Marie Genty
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université de Paris, France (B.B., I.L.-P., M.D., M.G., F.J.)
| | - Natalia Lopez-Andreés
- Cardiovascular Translational Research, Navarrabiomed (Miguel Servet Foundation), Instituto de Investigacioón Sanitaria de Navarra (IdiSNA), Pamplona, Spain (J.I., A.F.-C., N.L.-A.)
| | - Frédéric Jaisser
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université de Paris, France (B.B., I.L.-P., M.D., M.G., F.J.).,INSERM, Clinical Investigation Centre 1433, French-Clinical Research Infrastructure Network (F-CRIN) INI-CRCT, Nancy, France (F.J.)
| |
Collapse
|
33
|
Gerbino A, Forleo C, Milano S, Piccapane F, Procino G, Pepe M, Piccolo M, Guida P, Resta N, Favale S, Svelto M, Carmosino M. Pro-inflammatory cytokines as emerging molecular determinants in cardiolaminopathies. J Cell Mol Med 2021; 25:10902-10915. [PMID: 34773379 PMCID: PMC8642682 DOI: 10.1111/jcmm.16975] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 09/16/2021] [Accepted: 09/23/2021] [Indexed: 12/12/2022] Open
Abstract
Mutations in Lamin A/C gene (lmna) cause a wide spectrum of cardiolaminopathies strictly associated with significant deterioration of the electrical and contractile function of the heart. Despite the continuous flow of biomedical evidence, linking cardiac inflammation to heart remodelling in patients harbouring lmna mutations is puzzling. Therefore, we profiled 30 serum cytokines/chemokines in patients belonging to four different families carrying pathogenic lmna mutations segregating with cardiac phenotypes at different stages of severity (n = 19) and in healthy subjects (n = 11). Regardless lmna mutation subtype, high levels of circulating granulocyte colony‐stimulating factor (G‐CSF) and interleukin 6 (IL‐6) were found in all affected patients’ sera. In addition, elevated levels of Interleukins (IL) IL‐1Ra, IL‐1β IL‐4, IL‐5 and IL‐8 and the granulocyte‐macrophage colony‐stimulating factor (GM‐CSF) were measured in a large subset of patients associated with more aggressive clinical manifestations. Finally, the expression of the pro‐inflammatory 70 kDa heat shock protein (Hsp70) was significantly increased in serum exosomes of patients harbouring the lmna mutation associated with the more severe phenotype. Overall, the identification of patient subsets with overactive or dysregulated myocardial inflammatory responses could represent an innovative diagnostic, prognostic and therapeutic tool against Lamin A/C cardiomyopathies.
Collapse
Affiliation(s)
- Andrea Gerbino
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, Bari, Italy
| | - Cinzia Forleo
- Department of Emergency and Organ Transplantation, Cardiology Unit, University of Bari Aldo Moro, Bari, Italy
| | - Serena Milano
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, Bari, Italy
| | - Francesca Piccapane
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, Bari, Italy
| | - Giuseppe Procino
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, Bari, Italy
| | - Martino Pepe
- Department of Emergency and Organ Transplantation, Cardiology Unit, University of Bari Aldo Moro, Bari, Italy
| | - Mara Piccolo
- Department of Emergency and Organ Transplantation, Cardiology Unit, University of Bari Aldo Moro, Bari, Italy
| | - Piero Guida
- Regional General Hospital "F. Miulli", Acquaviva delle Fonti, Italy
| | - Nicoletta Resta
- Division of Medical Genetics, Department of Biomedical Sciences and Human Oncology, University of Bari Aldo Moro, Bari, Italy
| | - Stefano Favale
- Department of Emergency and Organ Transplantation, Cardiology Unit, University of Bari Aldo Moro, Bari, Italy
| | - Maria Svelto
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, Bari, Italy
| | - Monica Carmosino
- Department of Sciences, University of Basilicata, Potenza, Italy
| |
Collapse
|
34
|
Peterson D, Wambier C, Dai F, Lampert R, Ahmad T, Yale KL, Mesinkovska NA, King B. Electrocardiogram Findings in Patients with Alopecia Areata. Dermatol Ther (Heidelb) 2021; 11:2217-2223. [PMID: 34564797 PMCID: PMC8611143 DOI: 10.1007/s13555-021-00614-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Accepted: 09/11/2021] [Indexed: 11/30/2022] Open
Abstract
Introduction While autoimmune comorbidities are common in alopecia areata, little is known about comorbid cardiovascular disease. The purpose of this study was to evaluate the incidence of bradyarrhythmia in patients with alopecia areata. Methods Retrospective review of electrocardiograms of 124 patients with \documentclass[12pt]{minimal}
\usepackage{amsmath}
\usepackage{wasysym}
\usepackage{amsfonts}
\usepackage{amssymb}
\usepackage{amsbsy}
\usepackage{mathrsfs}
\usepackage{upgreek}
\setlength{\oddsidemargin}{-69pt}
\begin{document}$$\ge$$\end{document}≥ 50% scalp hair loss (severe alopecia areata) was conducted and compared to National Health and Nutrition Examination Survey (NHANES) data. Results The prevalence of bradycardia in females with alopecia areata was 24.3% (95% CI, 14.5–34.1%) and in those age 40 years or older was 40.8% (95% CI, 22.2–53.5%) compared to 19.5% in the NHANES III population. The prevalence of bradycardia in males with alopecia areata was 36.0% (95% CI, 22.7–49.3%) and in those age 40 years or older was 50.0% (95% CI, 21.7–78.3%) compared to 26.9% in the NHANES III population. Conclusion The potential association between bradycardia and alopecia areata merits further investigation.
Collapse
Affiliation(s)
- Danielle Peterson
- Department of Dermatology, Yale School of Medicine, New Haven, CT, USA
| | - Carlos Wambier
- Department of Dermatology, Brown University, Providence, RI, USA
| | - Feng Dai
- Center for Analytical Sciences, Yale School of Medicine, New Haven, CT, USA
| | - Rachel Lampert
- Department of Internal Medicine, Section of Cardiovascular Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Tariq Ahmad
- Department of Internal Medicine, Section of Cardiovascular Medicine, Yale School of Medicine, New Haven, CT, USA.
| | - Katerina L Yale
- Department of Dermatology, University of California, Irvine, Irvine, CA, USA
| | | | - Brett King
- Department of Dermatology, Yale School of Medicine, New Haven, CT, USA.
| |
Collapse
|
35
|
Chen X, Huang W, Zhao L, Li Y, Wang L, Mo F, Guo W. Relationship Between the Eosinophil/Monocyte Ratio and Prognosis in Decompensated Heart Failure: A Retrospective Study. J Inflamm Res 2021; 14:4687-4696. [PMID: 34557013 PMCID: PMC8453176 DOI: 10.2147/jir.s325229] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Accepted: 08/23/2021] [Indexed: 12/28/2022] Open
Abstract
Purpose The aim of this study was to assess the value of the eosinophil/monocyte ratio (EMR) for predicting the prognosis of decompensated heart failure (HF). Patients and Methods This was a retrospective cohort study. We included adults (≥18 years old) diagnosed with decompensated HF for whom EMR data were available. The patients were divided into three groups according to EMR tertiles (T1 [EMR≤0.15], T2 [0.15<EMR≤0.32], and T3 [EMR>0.32]). The primary endpoint was the composite outcome of cardiovascular death or HF rehospitalization. Results Initially, the records of 2264 patients with decompensated HF were screened; 1883 of these patients had EMR data and were therefore included in the study. There were 627 patients in the T1 group, 628 in the T2 group, and 628 in the T3 group. The risk of cardiovascular death or HF rehospitalization was significantly different among the three groups (Log rank test, P=0.007). Compared with the T3 group, both the T1 group (hazard ratio [HR]: 1.50, 95% confidence interval [CI]: 1.16–1.94, P=0.002) and the T2 group (HR: 1.34, 95% CI: 1.03–1.74, P=0.030) had significantly higher rates of cardiovascular death or HF rehospitalization. A Cochran-Armitage test for trend showed a positive correlation between the EMR and the composite outcome of cardiovascular death or HF. There was a significant difference between the three groups in terms of cardiovascular death (Log rank test, P<0.001) and HF rehospitalization (Log rank test, P=0.03). Conclusion The EMR is positively correlated with the risk of cardiovascular death or HF rehospitalization in patients with decompensated HF. Specifically, the lower the EMR, the higher the risk of cardiovascular death or HF rehospitalization.
Collapse
Affiliation(s)
- Xiehui Chen
- Department of Cardiology, Shenzhen Longhua District Central Hospital, Shenzhen, People's Republic of China
| | - Weichao Huang
- Department of Cardiology, Fuwai Hospital Chinese Academy of Medical Sciences, Shenzhen, People's Republic of China
| | - Lingyue Zhao
- Department of Ambulatory Surgery, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen, People's Republic of China
| | - Yichong Li
- Department of Cardiology, Fuwai Hospital Chinese Academy of Medical Sciences, Shenzhen, People's Republic of China
| | - Lili Wang
- Department of Cardiology, Fuwai Hospital Chinese Academy of Medical Sciences, Shenzhen, People's Republic of China
| | - Fanrui Mo
- Department of Cardiology, The Fourth Affiliated Hospital of Guangxi Medical University, Liuzhou, People's Republic of China
| | - Wenqin Guo
- Department of Cardiology, Fuwai Hospital Chinese Academy of Medical Sciences, Shenzhen, People's Republic of China
| |
Collapse
|
36
|
Tuleta I, Frangogiannis NG. Fibrosis of the diabetic heart: Clinical significance, molecular mechanisms, and therapeutic opportunities. Adv Drug Deliv Rev 2021; 176:113904. [PMID: 34331987 PMCID: PMC8444077 DOI: 10.1016/j.addr.2021.113904] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 07/19/2021] [Accepted: 07/24/2021] [Indexed: 01/02/2023]
Abstract
In patients with diabetes, myocardial fibrosis may contribute to the pathogenesis of heart failure and arrhythmogenesis, increasing ventricular stiffness and delaying conduction. Diabetic myocardial fibrosis involves effects of hyperglycemia, lipotoxicity and insulin resistance on cardiac fibroblasts, directly resulting in increased matrix secretion, and activation of paracrine signaling in cardiomyocytes, immune and vascular cells, that release fibroblast-activating mediators. Neurohumoral pathways, cytokines, growth factors, oxidative stress, advanced glycation end-products (AGEs), and matricellular proteins have been implicated in diabetic fibrosis; however, the molecular links between the metabolic perturbations and activation of a fibrogenic program remain poorly understood. Although existing therapies using glucose- and lipid-lowering agents and neurohumoral inhibition may act in part by attenuating myocardial collagen deposition, specific therapies targeting the fibrotic response are lacking. This review manuscript discusses the clinical significance, molecular mechanisms and cell biology of diabetic cardiac fibrosis and proposes therapeutic targets that may attenuate the fibrotic response, preventing heart failure progression.
Collapse
Affiliation(s)
- Izabela Tuleta
- The Wilf Family Cardiovascular Research Institute, Department of Medicine (Cardiology), Albert Einstein College of Medicine, Bronx NY, USA
| | - Nikolaos G Frangogiannis
- The Wilf Family Cardiovascular Research Institute, Department of Medicine (Cardiology), Albert Einstein College of Medicine, Bronx NY, USA.
| |
Collapse
|
37
|
Abstract
Objective Arteriovenous fistulae (AVF) placed for hemodialysis have high flow rates that can stimulate left ventricular (LV) hypertrophy. LV hypertrophy generally portends poor cardiac outcomes, yet clinical studies point to superior cardiac-specific outcomes for patients with AVF when compared with other dialysis modalities. We hypothesize that AVF induce physiologic cardiac hypertrophy with cardioprotective features. Methods We treated 9- to 11-week-old C57Bl/6 male and female mice with sham laparotomy or an aortocaval fistula via a 25G needle. Cardiac chamber size and function were assessed with serial echocardiography, and cardiac computed tomography angiography. Hearts were harvested at 5 weeks postoperatively, and the collagen content was assessed with Masson's trichrome. Bulk messenger RNA sequencing was performed from LV of sham and AVF mice at 10 days. Differentially expressed genes were analyzed using Ingenuity Pathway Analysis (Qiagen) to identify affected pathways and predict downstream biological effects. Results Mice with AVF had similar body weight and wet lung mass, but increased cardiac mass compared with sham-operated mice. AVF increased cardiac output while preserving LV systolic and diastolic function, as well as indices of right heart function; all four cardiac chambers were enlarged, with a slight decrement in the relative LV wall thickness. Histology showed preserved collagen density within each of the four chambers without areas of fibrosis. RNA sequencing captured 19 384 genes, of which 857 were significantly differentially expressed, including transcripts from extracellular matrix-related genes, ion channels, metabolism, and cardiac fetal genes. The top upstream regulatory molecules predicted include activation of angiogenic (Vegf, Akt1), procardiomyocyte survival (Hgf, Foxm1, Erbb2, Lin9, Areg), and inflammation-related (CSF2, Tgfb1, TNF, Ifng, Ccr2, IL6) genes, as well as the inactivation of cardiomyocyte antiproliferative factors (Cdkn1a, FoxO3, α-catenin). The predicted downstream effects include a decrease in heart damage, and increased arrhythmia, angiogenesis, and cardiogenesis. There were no significant sex-dependent differences in the AVF-stimulated cardiac adaptation. Conclusions AVF stimulate adaptive cardiac hypertrophy in wild-type mice without heart failure or pathologic fibrosis. Transcriptional correlates suggest AVF-induced cardiac remodeling has some cardioprotective, although also arrhythmogenic features. (JVS–Vascular Science 2021;2:110-28.) Clinical Relevance Arteriovenous fistulae (AVF) are commonly used as access for hemodialysis in patients with end-stage renal disease. AVF induce a high-output state that is associated with long-term structural cardiac remodeling, including left ventricle hypertrophy, but this element has uncertain clinical significance. Although left ventricle hypertrophy has traditionally been associated with an increased risk of cardiovascular disease, clinical studies have suggested that cardiac-specific outcomes of patients with end-stage renal disease were better with AVF compared with other dialysis modalities. This study uses a mouse model of AVF to study the structural, functional, and molecular correlates of AVF-induced cardiac remodeling. It finds that AVF causes an adaptive cardiac hypertrophy without functional decline or fibrosis. Transcriptional correlates suggest an electrical remodeling and the upregulation of proangiogenic, procardiogenic, and prosurvival factors, implying that AVF-induced cardiac hypertrophy is potentially cardioprotective, but also arrhythmogenic.
Collapse
|
38
|
Passaro F, Tocchetti CG, Spinetti G, Paudice F, Ambrosone L, Costagliola C, Cacciatore F, Abete P, Testa G. Targeting fibrosis in the failing heart with nanoparticles. Adv Drug Deliv Rev 2021; 174:461-481. [PMID: 33984409 DOI: 10.1016/j.addr.2021.05.004] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 04/15/2021] [Accepted: 05/07/2021] [Indexed: 02/06/2023]
Abstract
Heart failure (HF) is a clinical syndrome characterized by typical symptoms and signs caused by a structural and/or functional cardiac abnormality, resulting in a reduced cardiac output and/or elevated intracardiac pressures at rest or during stress. Due to increasing incidence, prevalence and, most importantly mortality, HF is a healthcare burden worldwide, despite the improvement of treatment options and effectiveness. Acute and chronic cardiac injuries trigger the activation of neurohormonal, inflammatory, and mechanical pathways ultimately leading to fibrosis, which plays a key role in the development of cardiac dysfunction and HF. The use of nanoparticles for targeted drug delivery would greatly improve therapeutic options to identify, prevent and treat cardiac fibrosis. In this review we will highlight the mechanisms of cardiac fibrosis development to depict the pathophysiological features for passive and active targeting of acute and chronic cardiac fibrosis with nanoparticles. Then we will discuss how cardiomyocytes, immune and inflammatory cells, fibroblasts and extracellular matrix can be targeted with nanoparticles to prevent or restore cardiac dysfunction and to improve the molecular imaging of cardiac fibrosis.
Collapse
|
39
|
Song J, Frieler RA, Vigil TM, Ma J, Brombacher F, Goonewardena SN, Goldstein DR, Mortensen RM. Inactivation of Interleukin-4 Receptor α Signaling in Myeloid Cells Protects Mice From Angiotensin II/High Salt-Induced Cardiovascular Dysfunction Through Suppression of Fibrotic Remodeling. J Am Heart Assoc 2021; 10:e017329. [PMID: 34132103 PMCID: PMC8403318 DOI: 10.1161/jaha.120.017329] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Background Hypertension‐induced cardiovascular remodeling is characterized by chronic low‐grade inflammation. Interleukin‐4 receptor α (IL‐4Rα) signaling is importantly involved in cardiovascular remodeling, however, the target cell type(s) is unclear. Here, we investigated the role of myeloid‐specific IL‐4Rα signaling in cardiovascular remodeling induced by angiotensin II and high salt. Methods and Results Myeloid IL‐4Rα deficiency suppressed both the in vitro and in vivo expression of alternatively activated macrophage markers including Arg1 (arginase 1), Ym1 (chitinase 3‐like 3), and Relmα/Fizz1 (resistin‐like molecule α). After angiotensin II and high salt treatment, myeloid‐specific IL‐4Rα deficiency did not change hypertrophic remodeling within the heart and aorta. However, myeloid IL‐4Rα deficiency resulted in a substantial reduction in fibrosis through the suppression of profibrotic pathways and the enhancement of antifibrotic signaling. Decreased fibrosis was associated with significant preservation of myocardial function in MyIL4RαKO mice and was mediated by attenuated alternative macrophage activation. Conclusions Myeloid IL‐4Rα signaling is substantially involved in fibrotic cardiovascular remodeling by controlling alternative macrophage activation and regulating fibrosis‐related signaling. Inhibiting myeloid IL‐4Rα signaling may be a potential strategy to prevent hypertensive cardiovascular diseases.
Collapse
Affiliation(s)
- Jianrui Song
- Department of Cell and Developmental Biology University of Michigan Medical School Ann Arbor MI.,Department of Molecular and Integrative Physiology University of Michigan Ann Arbor MI
| | - Ryan A Frieler
- Department of Molecular and Integrative Physiology University of Michigan Ann Arbor MI
| | - Thomas M Vigil
- Department of Molecular and Integrative Physiology University of Michigan Ann Arbor MI
| | - Jun Ma
- Department of Thoracic Surgery Shanxi Province People's Hospital Taiyuan P.R. China
| | - Frank Brombacher
- International Center for Genetic Engineering and Biotechnology University of Cape TownDivision of Immunology and South African Medical Research Council (SAMRC) Cape Town South Africa
| | - Sascha N Goonewardena
- Division of Cardiovascular Medicine Department of Internal Medicine University of Michigan Ann Arbor MI
| | - Daniel R Goldstein
- Division of Cardiovascular Medicine Department of Internal Medicine University of Michigan Ann Arbor MI.,Institute of Gerontology University of Michigan Ann Arbor MI.,Department of Microbiology and Immunology University of Michigan Ann Arbor MI
| | - Richard M Mortensen
- Department of Molecular and Integrative Physiology University of Michigan Ann Arbor MI.,Division of Metabolism, Endocrinology, and Diabetes Department of Internal Medicine University of Michigan Ann Arbor MI.,Department of Pharmacology University of Michigan Ann Arbor MI
| |
Collapse
|
40
|
Reddy SS, Agarwal H, Jaiswal A, Jagavelu K, Dikshit M, Barthwal MK. Macrophage p47 phox regulates pressure overload-induced left ventricular remodeling by modulating IL-4/STAT6/PPARγ signaling. Free Radic Biol Med 2021; 168:168-179. [PMID: 33736980 DOI: 10.1016/j.freeradbiomed.2021.03.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 03/02/2021] [Accepted: 03/05/2021] [Indexed: 11/19/2022]
Abstract
NADPH oxidase (Nox) mediates ROS production and contributes to cardiac remodeling. However, macrophage p47phox, a Nox subunit regulating cardiac remodeling, is unclear. We aimed to investigate the role of macrophage p47phox in hypertensive cardiac remodeling. Pressure-overload induced by Angiotensin II (AngII) for two weeks in young adult male p47phox deficient (KO) mice showed aggravated cardiac dysfunction and hypertrophy as indicated from echocardiographic and histological studies in comparison with wild-type littermates (WT). Additionally, LV of AngII-infused KO mice showed augmented interstitial fibrosis, collagen deposition and, myofibroblasts compared to AngII-infused WT mice. Moreover, these changes in AngII-infused KO mice correlated well with the gene analysis of hypertrophic and fibrotic markers. Similar results were also found in the transverse aortic constriction model. Further, AngII-infused KO mice showed elevated circulating immunokines and increased LV leukocytes infiltration and CD206+ macrophages compared to AngII-infused WT mice. Likewise, LV of AngII-infused KO mice showed upregulated mRNA expression of anti-inflammatory/pro-fibrotic M2 macrophage markers (Ym1, Arg-1) compared to AngII-infused WT mice. AngII and IL-4 treated bone marrow-derived macrophages (BMDMs) from KO mice showed upregulated M2 macrophage markers and STAT6 phosphorylation (Y641) compared to AngII and IL-4 treated WT BMDMs. These alterations were at least partly mediated by macrophage as bone marrow transplantation from KO mice into WT mice aggravated cardiac remodeling. Mechanistically, AngII-infused KO mice showed hyperactivated IL-4/STAT6/PPARγ signaling and downregulated SOCS3 expression compared to AngII-infused WT mice. Our studies show that macrophage p47phox limits anti-inflammatory signaling and extracellular matrix remodeling in response to pressure-overload.
Collapse
Affiliation(s)
- Sukka Santosh Reddy
- Pharmacology Division, CSIR-Central Drug Research Institute, Lucknow, 226031, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Heena Agarwal
- Pharmacology Division, CSIR-Central Drug Research Institute, Lucknow, 226031, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Anant Jaiswal
- Pharmacology Division, CSIR-Central Drug Research Institute, Lucknow, 226031, India
| | - Kumaravelu Jagavelu
- Pharmacology Division, CSIR-Central Drug Research Institute, Lucknow, 226031, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Madhu Dikshit
- Pharmacology Division, CSIR-Central Drug Research Institute, Lucknow, 226031, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Manoj Kumar Barthwal
- Pharmacology Division, CSIR-Central Drug Research Institute, Lucknow, 226031, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India.
| |
Collapse
|
41
|
Song J, Frieler RA, Whitesall SE, Chung Y, Vigil TM, Muir LA, Ma J, Brombacher F, Goonewardena SN, Lumeng CN, Goldstein DR, Mortensen RM. Myeloid interleukin-4 receptor α is essential in postmyocardial infarction healing by regulating inflammation and fibrotic remodeling. Am J Physiol Heart Circ Physiol 2021; 320:H323-H337. [PMID: 33164548 PMCID: PMC7847075 DOI: 10.1152/ajpheart.00251.2020] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 10/23/2020] [Accepted: 10/29/2020] [Indexed: 02/06/2023]
Abstract
Interleukin-4 receptor α (IL4Rα) signaling plays an important role in cardiac remodeling during myocardial infarction (MI). However, the target cell type(s) of IL4Rα signaling during this remodeling remains unclear. Here, we investigated the contribution of endogenous myeloid-specific IL4Rα signaling in cardiac remodeling post-MI. We established a murine myeloid-specific IL4Rα knockout (MyIL4RαKO) model with LysM promoter-driven Cre recombination. Macrophages from MyIL4RαKO mice showed significant downregulation of alternatively activated macrophage markers but an upregulation of classical activated macrophage markers both in vitro and in vivo, indicating the successful inactivation of IL4Rα signaling in macrophages. To examine the role of myeloid IL4Rα during MI, we subjected MyIL4RαKO and littermate floxed control (FC) mice to MI. We found that cardiac function was significantly impaired as a result of myeloid-specific IL4Rα deficiency. This deficiency resulted in a dysregulated inflammatory response consisting of decreased production of anti-inflammatory cytokines. Myeloid IL4Rα deficiency also led to reduced collagen 1 deposition and an imbalance of matrix metalloproteinases (MMPs)/tissue inhibitors of metalloproteinases (TIMPs), with upregulated MMPs and downregulated TIMPs, which resulted in insufficient fibrotic remodeling. In conclusion, this study identifies that myeloid-specific IL4Rα signaling regulates inflammation and fibrotic remodeling during MI. Therefore, myeloid-specific activation of IL4Rα signaling could offer protective benefits after MI.NEW & NOTEWORTHY This study showed, for the first time, the role of endogenous IL4Rα signaling in myeloid cells during cardiac remodeling and the underlying mechanisms. We identified myeloid cells are the critical target cell types of IL4Rα signaling during cardiac remodeling post-MI. Deficiency of myeloid IL4Rα signaling causes deteriorated cardiac function post-MI, due to dysregulated inflammation and insufficient fibrotic remodeling. This study sheds light on the potential of activating myeloid-specific IL4Rα signaling to modify remodeling post-MI. This brings hope to patients with MI and diminishes side effects by cell type-specific instead of whole body treatment.
Collapse
Affiliation(s)
- Jianrui Song
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, Michigan
| | - Ryan A Frieler
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan
| | - Steven E Whitesall
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan
| | - Yutein Chung
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan
| | - Thomas M Vigil
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan
| | - Lindsey A Muir
- Department of Pediatrics and Communicable Diseases, University of Michigan, Ann Arbor, Michigan
| | - Jun Ma
- Department of Thoracic Surgery, Shanxi Province People's Hospital, Taiyuan, People's Republic of China
| | - Frank Brombacher
- International Center for Genetic Engineering and Biotechnology, University of Cape Town, Cape Town, South Africa
| | - Sascha N Goonewardena
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
| | - Carey N Lumeng
- Department of Pediatrics and Communicable Diseases, University of Michigan, Ann Arbor, Michigan
| | - Daniel R Goldstein
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
- Institute of Gerontology, University of Michigan, Ann Arbor, Michigan
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, Michigan
| | - Richard M Mortensen
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan
- Division of Metabolism, Endocrinology, and Diabetes, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
- Department of Pharmacology, University of Michigan, Ann Arbor, Michigan
| |
Collapse
|
42
|
Monteonofrio L, Florio MC, AlGhatrif M, Lakatta EG, Capogrossi MC. Aging- and gender-related modulation of RAAS: potential implications in COVID-19 disease. VASCULAR BIOLOGY 2020; 3:R1-R14. [PMID: 33537555 PMCID: PMC7849461 DOI: 10.1530/vb-20-0014] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Accepted: 12/11/2020] [Indexed: 12/15/2022]
Abstract
Coronavirus disease 2019 (COVID-19) is a new infectious disease caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). COVID-19 is frequently characterized by a marked inflammatory response with severe pneumonia and respiratory failure associated with multiorgan involvement. Some risk factors predispose patients to develop a more severe infection and to an increased mortality; among them, advanced age and male gender have been identified as major and independent risk factors for COVID-19 poor outcome. The renin-angiotensin-aldosterone system (RAAS) is strictly involved in COVID-19 because angiotensin converting enzyme 2 (ACE2) is the host receptor for SARS-CoV-2 and also converts pro-inflammatory angiotensin (Ang) II into anti-inflammatory Ang(1–7). In this review, we have addressed the effect of aging and gender on RAAS with emphasis on ACE2, pro-inflammatory Ang II/Ang II receptor 1 axis and anti-inflammatory Ang(1–7)/Mas receptor axis.
Collapse
Affiliation(s)
- Laura Monteonofrio
- Laboratory of Cardiovascular Science, National Institute on Aging, National Institutes of Health, Baltimore, Maryland, USA
| | - Maria Cristina Florio
- Laboratory of Cardiovascular Science, National Institute on Aging, National Institutes of Health, Baltimore, Maryland, USA
| | - Majd AlGhatrif
- Laboratory of Cardiovascular Science, National Institute on Aging, National Institutes of Health, Baltimore, Maryland, USA.,Longitudinal Study Section, National Institute on Aging, National Institutes of Health, Baltimore, Maryland, USA.,Division of Cardiology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Edward G Lakatta
- Laboratory of Cardiovascular Science, National Institute on Aging, National Institutes of Health, Baltimore, Maryland, USA
| | - Maurizio C Capogrossi
- Laboratory of Cardiovascular Science, National Institute on Aging, National Institutes of Health, Baltimore, Maryland, USA.,Division of Cardiology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
43
|
Apaydin DC, Jaramillo PAM, Corradi L, Cosco F, Rathjen FG, Kammertoens T, Filosa A, Sawamiphak S. Early-Life Stress Regulates Cardiac Development through an IL-4-Glucocorticoid Signaling Balance. Cell Rep 2020; 33:108404. [PMID: 33207196 DOI: 10.1016/j.celrep.2020.108404] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 09/21/2020] [Accepted: 10/27/2020] [Indexed: 01/06/2023] Open
Abstract
Stressful experiences early in life can increase the risk of cardiovascular diseases. However, it remains largely unknown how stress influences susceptibility to the disease onset. Here, we show that exposure to brain-processed stress disrupts myocardial growth by reducing cardiomyocyte mitotic activity. Activation of the glucocorticoid receptor (GR), the primary stress response pathway, reduces cardiomyocyte numbers, disrupts trabecular formation, and leads to contractile dysfunction of the developing myocardium. However, a physiological level of GR signaling is required to prevent cardiomyocyte hyperproliferation. Mechanistically, we identify an antagonistic interaction between the GR and the cytokine interleukin-4 (IL-4) as a key player in cardiac development. IL-4 signals transcription of key regulators of cell-cycle progression in cardiomyocytes via signal transducer and activator of transcription 3 (Stat3). GR, on the contrary, inhibits this signaling system. Thus, our findings uncover an interplay between stress and immune signaling pathways critical to orchestrating physiological growth of the heart.
Collapse
Affiliation(s)
- Dilem C Apaydin
- Max Delbrück Center for Molecular Medicine, Robert-Rössle-Straße 10, 13092 Berlin, Germany
| | | | - Laura Corradi
- Max Delbrück Center for Molecular Medicine, Robert-Rössle-Straße 10, 13092 Berlin, Germany
| | - Francesca Cosco
- Max Delbrück Center for Molecular Medicine, Robert-Rössle-Straße 10, 13092 Berlin, Germany
| | - Fritz G Rathjen
- Max Delbrück Center for Molecular Medicine, Robert-Rössle-Straße 10, 13092 Berlin, Germany
| | - Thomas Kammertoens
- Max Delbrück Center for Molecular Medicine, Robert-Rössle-Straße 10, 13092 Berlin, Germany; Institute of Immunology, Charité Campus Berlin Buch, Lindenberger Weg 80, 13125 Berlin, Germany
| | - Alessandro Filosa
- Max Delbrück Center for Molecular Medicine, Robert-Rössle-Straße 10, 13092 Berlin, Germany
| | - Suphansa Sawamiphak
- Max Delbrück Center for Molecular Medicine, Robert-Rössle-Straße 10, 13092 Berlin, Germany; DZHK (German Center for Cardiovascular Research), Partner Site Berlin, Berlin, Germany.
| |
Collapse
|
44
|
Kariyawasam HH. Chronic rhinosinusitis with nasal polyps: mechanistic insights from targeting IL-4 and IL-13 via IL-4Rα inhibition with dupilumab. Expert Rev Clin Immunol 2020; 16:1115-1125. [PMID: 33148074 DOI: 10.1080/1744666x.2021.1847083] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Introduction: Chronic rhinosinusitis with nasal polyps (CRSwNP) is a complex immunological upper airway disease . CRSwNP, particularly in Caucasians, often has a more distinct T2 inflammatory endotype. IL-4 and IL-13 are key upstream cytokines that help establish and sustain T2 inflammation as well as strongly influencing tissue remodeling. They have a shared signaling receptor IL-4Rα. An attractive and novel therapeutic approach is by way of blocking IL-4 and IL-13 simultaneously via inhibiting IL-4Rα. Dupilumab is a murine derived fully human monoclonal inhibitory antibody directed against IL-4Rα which thereby prevents IL-4/IL-13 cell signaling. Following successful Phase 3 studies dupilumab has become the first licensed biologic for treating CRSwNP. Areas covered: This review covers the essential immunology of CRSwNP in the context of IL-4 and IL-13 signaling via IL-4Rα. The potential mechanisms by which therapeutic improvements occur with dupilumab are evaluated. IL-4, IL-13, dupilumab and rhinosinusitis were used as the search terms in PubMed and Google Scholar through to August 2020. Expert commentary: Dupilumab has the potential to transform the care for patients with CRSwNP. It is essential that further studies are conducted promptly to identify disease-specific biomarkers and clinical traits to guide clinicians on best patient selection thereby ensuring optimal dupilumab outcomes.
Collapse
Affiliation(s)
- Harsha H Kariyawasam
- Rhinology Section, Specialist Allergy and Clinical Immunology, Royal National ENT Hospital, London University College London Hospital NHS Foundation Trust, University College London , London, UK
| |
Collapse
|
45
|
Abstract
Myocardial fibrosis, the expansion of the cardiac interstitium through deposition of extracellular matrix proteins, is a common pathophysiologic companion of many different myocardial conditions. Fibrosis may reflect activation of reparative or maladaptive processes. Activated fibroblasts and myofibroblasts are the central cellular effectors in cardiac fibrosis, serving as the main source of matrix proteins. Immune cells, vascular cells and cardiomyocytes may also acquire a fibrogenic phenotype under conditions of stress, activating fibroblast populations. Fibrogenic growth factors (such as transforming growth factor-β and platelet-derived growth factors), cytokines [including tumour necrosis factor-α, interleukin (IL)-1, IL-6, IL-10, and IL-4], and neurohumoral pathways trigger fibrogenic signalling cascades through binding to surface receptors, and activation of downstream signalling cascades. In addition, matricellular macromolecules are deposited in the remodelling myocardium and regulate matrix assembly, while modulating signal transduction cascades and protease or growth factor activity. Cardiac fibroblasts can also sense mechanical stress through mechanosensitive receptors, ion channels and integrins, activating intracellular fibrogenic cascades that contribute to fibrosis in response to pressure overload. Although subpopulations of fibroblast-like cells may exert important protective actions in both reparative and interstitial/perivascular fibrosis, ultimately fibrotic changes perturb systolic and diastolic function, and may play an important role in the pathogenesis of arrhythmias. This review article discusses the molecular mechanisms involved in the pathogenesis of cardiac fibrosis in various myocardial diseases, including myocardial infarction, heart failure with reduced or preserved ejection fraction, genetic cardiomyopathies, and diabetic heart disease. Development of fibrosis-targeting therapies for patients with myocardial diseases will require not only understanding of the functional pluralism of cardiac fibroblasts and dissection of the molecular basis for fibrotic remodelling, but also appreciation of the pathophysiologic heterogeneity of fibrosis-associated myocardial disease.
Collapse
Affiliation(s)
- Nikolaos G Frangogiannis
- Department of Medicine (Cardiology), The Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, 1300 Morris Park Avenue Forchheimer G46B, Bronx, NY 10461, USA
| |
Collapse
|
46
|
Hammoud SH, Wehbe Z, Abdelhady S, Kobeissy F, Eid AH, El-Yazbi AF. Dysregulation of Angiotensin Converting Enzyme 2 Expression and Function in Comorbid Disease Conditions Possibly Contributes to Coronavirus Infectious Disease 2019 Complication Severity. Mol Pharmacol 2020; 99:17-28. [PMID: 33082267 DOI: 10.1124/molpharm.120.000119] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Accepted: 10/09/2020] [Indexed: 01/08/2023] Open
Abstract
ACE2 has emerged as a double agent in the COVID-19 ordeal, as it is both physiologically protective and virally conducive. The identification of ACE2 in as many as 72 tissues suggests that extrapulmonary invasion and damage is likely, which indeed has already been demonstrated by cardiovascular and gastrointestinal symptoms. On the other hand, identifying ACE2 dysregulation in patients with comorbidities may offer insight as to why COVID-19 symptoms are often more severe in these individuals. This may be attributed to a pre-existing proinflammatory state that is further propelled with the cytokine storm induced by SARS-CoV-2 infection or the loss of functional ACE2 expression as a result of viral internalization. Here, we aim to characterize the distribution and role of ACE2 in various organs to highlight the scope of damage that may arise upon SARS-CoV-2 invasion. Furthermore, by examining the disruption of ACE2 in several comorbid diseases, we offer insight into potential causes of increased severity of COVID-19 symptoms in certain individuals. SIGNIFICANCE STATEMENT: Cell surface expression of ACE2 determines the tissue susceptibility for coronavirus infectious disease 2019 infection. Comorbid disease conditions altering ACE2 expression could increase the patient's vulnerability for the disease and its complications, either directly, through modulation of viral infection, or indirectly, through alteration of inflammatory status.
Collapse
Affiliation(s)
- Safaa H Hammoud
- Department of Pharmacology and Therapeutics, Beirut Arab University, Beirut, Lebanon (S.H.); Departments of Biology (Z.W.), Biochemistry and Molecular Genetics (F.K.), and Pharmacology and Toxicology (A.H.E., A.F.E.-Y.), American University of Beirut, Beirut, Lebanon; Department of Pharmacology and Toxicology, Faculty of Pharmacy (A.F.E.-Y.) and Faculty of Medicine (S.A.), Alexandria University, Alexandria, Egypt; and Department of Basic Medical Sciences, College of Medicine, and Biomedical and Pharmaceutical Research Unit, QU Health, Qatar University, Doha, Qatar (A.H.E.)
| | - Zena Wehbe
- Department of Pharmacology and Therapeutics, Beirut Arab University, Beirut, Lebanon (S.H.); Departments of Biology (Z.W.), Biochemistry and Molecular Genetics (F.K.), and Pharmacology and Toxicology (A.H.E., A.F.E.-Y.), American University of Beirut, Beirut, Lebanon; Department of Pharmacology and Toxicology, Faculty of Pharmacy (A.F.E.-Y.) and Faculty of Medicine (S.A.), Alexandria University, Alexandria, Egypt; and Department of Basic Medical Sciences, College of Medicine, and Biomedical and Pharmaceutical Research Unit, QU Health, Qatar University, Doha, Qatar (A.H.E.)
| | - Samar Abdelhady
- Department of Pharmacology and Therapeutics, Beirut Arab University, Beirut, Lebanon (S.H.); Departments of Biology (Z.W.), Biochemistry and Molecular Genetics (F.K.), and Pharmacology and Toxicology (A.H.E., A.F.E.-Y.), American University of Beirut, Beirut, Lebanon; Department of Pharmacology and Toxicology, Faculty of Pharmacy (A.F.E.-Y.) and Faculty of Medicine (S.A.), Alexandria University, Alexandria, Egypt; and Department of Basic Medical Sciences, College of Medicine, and Biomedical and Pharmaceutical Research Unit, QU Health, Qatar University, Doha, Qatar (A.H.E.)
| | - Firas Kobeissy
- Department of Pharmacology and Therapeutics, Beirut Arab University, Beirut, Lebanon (S.H.); Departments of Biology (Z.W.), Biochemistry and Molecular Genetics (F.K.), and Pharmacology and Toxicology (A.H.E., A.F.E.-Y.), American University of Beirut, Beirut, Lebanon; Department of Pharmacology and Toxicology, Faculty of Pharmacy (A.F.E.-Y.) and Faculty of Medicine (S.A.), Alexandria University, Alexandria, Egypt; and Department of Basic Medical Sciences, College of Medicine, and Biomedical and Pharmaceutical Research Unit, QU Health, Qatar University, Doha, Qatar (A.H.E.)
| | - Ali H Eid
- Department of Pharmacology and Therapeutics, Beirut Arab University, Beirut, Lebanon (S.H.); Departments of Biology (Z.W.), Biochemistry and Molecular Genetics (F.K.), and Pharmacology and Toxicology (A.H.E., A.F.E.-Y.), American University of Beirut, Beirut, Lebanon; Department of Pharmacology and Toxicology, Faculty of Pharmacy (A.F.E.-Y.) and Faculty of Medicine (S.A.), Alexandria University, Alexandria, Egypt; and Department of Basic Medical Sciences, College of Medicine, and Biomedical and Pharmaceutical Research Unit, QU Health, Qatar University, Doha, Qatar (A.H.E.)
| | - Ahmed F El-Yazbi
- Department of Pharmacology and Therapeutics, Beirut Arab University, Beirut, Lebanon (S.H.); Departments of Biology (Z.W.), Biochemistry and Molecular Genetics (F.K.), and Pharmacology and Toxicology (A.H.E., A.F.E.-Y.), American University of Beirut, Beirut, Lebanon; Department of Pharmacology and Toxicology, Faculty of Pharmacy (A.F.E.-Y.) and Faculty of Medicine (S.A.), Alexandria University, Alexandria, Egypt; and Department of Basic Medical Sciences, College of Medicine, and Biomedical and Pharmaceutical Research Unit, QU Health, Qatar University, Doha, Qatar (A.H.E.)
| |
Collapse
|
47
|
Increased immunosuppression impairs tissue homeostasis with aging and age-related diseases. J Mol Med (Berl) 2020; 99:1-20. [PMID: 33025106 PMCID: PMC7782450 DOI: 10.1007/s00109-020-01988-7] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 09/21/2020] [Accepted: 09/29/2020] [Indexed: 01/10/2023]
Abstract
Abstract Chronic low-grade inflammation is a common hallmark of the aging process and many age-related diseases. There is substantial evidence that persistent inflammation is associated with a compensatory anti-inflammatory response which prevents excessive tissue damage. Interestingly, the inflammatory state encountered with aging, called inflammaging, is associated with the anti-inflammaging process. The age-related activation of immunosuppressive network includes an increase in the numbers of myeloid-derived suppressor cells (MDSC), regulatory T cells (Treg), and macrophages (Mreg/M2c). Immunosuppressive cells secrete several anti-inflammatory cytokines, e.g., TGF-β and IL-10, as well as reactive oxygen and nitrogen species (ROS/RNS). Moreover, immunosuppressive cells suppress the function of effector immune cells by catabolizing l-arginine and tryptophan through the activation of arginase 1 (ARG1) and indoleamine 2,3-dioxygenase (IDO), respectively. Unfortunately, the immunosuppressive armament also induces harmful bystander effects in neighboring cells by impairing host tissue homeostasis. For instance, TGF-β signaling can trigger many age-related degenerative changes, e.g., cellular senescence, fibrosis, osteoporosis, muscle atrophy, and the degeneration of the extracellular matrix. In addition, changes in the levels of ROS, RNS, and the metabolites of the kynurenine pathway can impair tissue homeostasis. This review will examine in detail the harmful effects of the immunosuppressive cells on host tissues. It seems that this age-related immunosuppression prevents inflammatory damage but promotes the tissue degeneration associated with aging and age-related diseases. Key messages • Low-grade inflammation is associated with the aging process and age-related diseases. • Persistent inflammation activates compensatory immunosuppression with aging. • The numbers of immunosuppressive cells increase with aging and age-related diseases. • Immunosuppressive mechanisms evoke harmful bystander effects in host tissues. • Immunosuppression promotes tissue degeneration with aging and age-related diseases.
Collapse
|
48
|
Thomas TP, Grisanti LA. The Dynamic Interplay Between Cardiac Inflammation and Fibrosis. Front Physiol 2020; 11:529075. [PMID: 33041853 PMCID: PMC7522448 DOI: 10.3389/fphys.2020.529075] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Accepted: 08/14/2020] [Indexed: 12/12/2022] Open
Abstract
Heart failure is a leading cause of death worldwide. While there are multiple etiologies contributing to the development of heart failure, all cause result in impairments in cardiac function that is characterized by changes in cardiac remodeling and compliance. Fibrosis is associated with nearly all forms of heart failure and is an important contributor to disease pathogenesis. Inflammation also plays a critical role in the heart and there is a large degree of interconnectedness between the inflammatory and fibrotic response. This review discusses the cellular and molecular mechanisms contributing to inflammation and fibrosis and the interplay between the two.
Collapse
Affiliation(s)
- Toby P Thomas
- Department of Biomedical Sciences, College of Veterinary Medicine, University of Missouri, Columbia, MO, United States
| | - Laurel A Grisanti
- Department of Biomedical Sciences, College of Veterinary Medicine, University of Missouri, Columbia, MO, United States
| |
Collapse
|
49
|
Li X, Zhang W, Cao Q, Wang Z, Zhao M, Xu L, Zhuang Q. Mitochondrial dysfunction in fibrotic diseases. Cell Death Discov 2020; 6:80. [PMID: 32963808 PMCID: PMC7474731 DOI: 10.1038/s41420-020-00316-9] [Citation(s) in RCA: 103] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 08/13/2020] [Accepted: 08/27/2020] [Indexed: 12/11/2022] Open
Abstract
Although fibrosis is a common pathological feature of most end-stage organ diseases, its pathogenesis remains unclear. There is growing evidence that mitochondrial dysfunction contributes to the development and progression of fibrosis. The heart, liver, kidney and lung are highly oxygen-consuming organs that are sensitive to mitochondrial dysfunction. Moreover, the fibrotic process of skin and islet is closely related to mitochondrial dysfunction as well. This review summarized emerging mechanisms related to mitochondrial dysfunction in different fibrotic organs and tissues above. First, it highlighted the important elucidation of mitochondria morphological changes, mitochondrial membrane potential and structural damage, mitochondrial DNA (mtDNA) damage and reactive oxidative species (ROS) production, etc. Second, it introduced the abnormality of mitophagy and mitochondrial transfer also contributed to the fibrotic process. Therefore, with gaining the increasing knowledge of mitochondrial structure, function, and origin, we could kindle a new era for the diagnostic and therapeutic strategies of many fibrotic diseases based on mitochondrial dysfunction.
Collapse
Affiliation(s)
- Xinyu Li
- Transplantation Center of the 3rd Xiangya Hospital, Central South University, 410013 Changsha, Hunan China
- Xiangya School of Medicine, Central South University, 410013 Changsha, Hunan China
| | - Wei Zhang
- Transplantation Center of the 3rd Xiangya Hospital, Central South University, 410013 Changsha, Hunan China
- Xiangya School of Medicine, Central South University, 410013 Changsha, Hunan China
| | - Qingtai Cao
- Hunan Normal University School of Medicine, 410013 Changsha, Hunan China
| | - Zeyu Wang
- Xiangya School of Medicine, Central South University, 410013 Changsha, Hunan China
| | - Mingyi Zhao
- Pediatric Department of the 3rd Xiangya Hospital, Central South University, 410013 Changsha, Hunan China
| | - Linyong Xu
- School of Life Science, Central South University, 410013 Changsha, Hunan China
| | - Quan Zhuang
- Transplantation Center of the 3rd Xiangya Hospital, Central South University, 410013 Changsha, Hunan China
- Research Center of National Health Ministry on Transplantation Medicine, 410013 Changsha, Hunan China
| |
Collapse
|
50
|
Coste MER, França CN, Izar MC, Teixeira D, Ishimura ME, Longo-Maugeri I, Bacchin AS, Bianco HT, Moreira FT, Pinto IM, Szarf G, Caixeta AM, Berwanger O, Gonçalves I, Fonseca FAH. Early Changes in Circulating Interleukins and Residual Inflammatory Risk After Acute Myocardial Infarction. Arq Bras Cardiol 2020; 115:1104-1111. [PMID: 32876202 PMCID: PMC8133737 DOI: 10.36660/abc.20190567] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Accepted: 12/27/2019] [Indexed: 02/05/2023] Open
Abstract
Fundamento Pacientes com infarto agudo do miocárdio podem apresentar uma grande área infartada e disfunção ventricular mesmo com trombólise e revascularização precoces. Objetivo Investigar o comportamento das citocinas circulantes em pacientes com infarto agudo do miocárdio com supradesnivelamento do segmento ST (IAMCSST) e a relação delas com a função ventricular. Métodos No estudo BATTLE-AMI (Avaliação dos Linfócitos Tipos B e T no Infarto Agudo do Miocárdio), os pacientes com IAMCSST foram tratados com uma estratégia farmacoinvasiva. Os níveis de citocinas (IL-1β, IL-4, IL-6, IL-10 e IL-18) no plasma foram testados através de ensaio de imunoadsorção enzimática (ELISA) no início do estudo e após 30 dias. A massa infartada e a fração de ejeção ventricular esquerda (FEVE) foram examinadas por ressonância magnética cardíaca 3-T. Valores de p menores que 0,05 foram considerados significativos. Resultados Na comparação com o início do estudo, níveis mais baixos foram detectados para IL-1β (p = 0,028) e IL-18 (p < 0,0001) após 30 dias do IAMCSST, enquanto níveis mais altos foram observados para IL-4 (p = 0,001) e IL-10 (p < 0,0001) no mesmo momento. Em contrapartida, nenhuma mudança foi detectada nos níveis de IL-6 (p = 0,63). Os níveis da proteína C-reativa de alta sensibilidade e de IL-6 se correlacionaram no início do estudo (rho = 0,45, p < 0,0001) e 30 dias após o IAMCSST (rho = 0,29, p = 0,009). No início do estudo, a correlação entre os níveis de IL-6 e FEVE também foi observada (rho = -0,50, p = 0,004). Conclusões Durante o primeiro mês pós-infarto agudo do miocárdio, observamos uma melhora significativa no balanço das citocinas pró e anti-inflamatórias, exceto da IL-6. Esses achados sugerem risco inflamatório residual. (Arq Bras Cardiol. 2020; [online].ahead print, PP.0-0)
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Gilberto Szarf
- Universidade Federal de São Paulo, São Paulo, SP - Brasil
| | | | | | - Iran Gonçalves
- Universidade Federal de São Paulo, São Paulo, SP - Brasil
| | | |
Collapse
|