1
|
Varzideh F, Wang B, Qin Y, Kansakar U, Santulli G, Jankauskas SS. Mechanistic role of mesencephalic astrocyte-derived neurotrophic factor in myocardial ischemia/reperfusion injury. Mol Med 2024; 30:188. [PMID: 39462320 PMCID: PMC11512500 DOI: 10.1186/s10020-024-00927-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Accepted: 09/05/2024] [Indexed: 10/29/2024] Open
Abstract
Mesencephalic astrocyte-derived neurotrophic factor (MANF) is a protein crucial for cellular stress response and survival, particularly in the nervous and cardiovascular systems. Unlike traditional neurotrophic factors, MANF primarily regulates endoplasmic reticulum (ER) stress and protects cells by reducing ER stress-induced apoptosis. MANF operates both inside and outside cells, influencing key pathways like JAK/STAT and NF-κB to enhance cell survival during stress. Beyond its neuroprotective role, MANF is also vital in cardiovascular protection, mitigating damage by reducing inflammation and maintaining cellular function. Elevated MANF levels have been observed in patients experiencing myocardial infarction and murine models of ischemia-reperfusion (I/R) injury, highlighting its importance in these conditions. Overexpression of MANF in cardiomyocytes reduces ER-stress-induced cell death, while its depletion worsens this effect. Treatment with recombinant MANF (rMANF) has been shown to improve cardiac function in mice with I/R injury by decreasing infarct size and inflammation. Research also indicates that alterations in the α1-helix region of MANF can impact its structure, expression, secretion, and overall function. Given its protective effects and involvement in critical signaling pathways, MANF is being explored as a potential therapeutic target for ER stress-related diseases, including neurodegenerative disorders and cardiovascular conditions like myocardial I/R injury.
Collapse
Affiliation(s)
- Fahimeh Varzideh
- Department of Medicine, Division of Cardiology, Wilf Family Cardiovascular Research Institute, Einstein Institute for Neuroimmunology and Inflammation (INI), Albert Einstein College of Medicine, Montefiore University Hospital, 1300 Morris Park Avenue, New York, NY, 10461, USA
| | - Brandon Wang
- Department of Medicine, Division of Cardiology, Wilf Family Cardiovascular Research Institute, Einstein Institute for Neuroimmunology and Inflammation (INI), Albert Einstein College of Medicine, Montefiore University Hospital, 1300 Morris Park Avenue, New York, NY, 10461, USA
| | - Yifei Qin
- Department of Molecular Pharmacology, Einstein-Mount Sinai Diabetes Research Center (ES-DRC), Fleischer Institute for Diabetes and Metabolism (FIDAM), Einstein Institute for Aging Research, Albert Einstein College of Medicine, Montefiore University Hospital, New York, NY, 10461, USA
| | - Urna Kansakar
- Department of Molecular Pharmacology, Einstein-Mount Sinai Diabetes Research Center (ES-DRC), Fleischer Institute for Diabetes and Metabolism (FIDAM), Einstein Institute for Aging Research, Albert Einstein College of Medicine, Montefiore University Hospital, New York, NY, 10461, USA
| | - Gaetano Santulli
- Department of Medicine, Division of Cardiology, Wilf Family Cardiovascular Research Institute, Einstein Institute for Neuroimmunology and Inflammation (INI), Albert Einstein College of Medicine, Montefiore University Hospital, 1300 Morris Park Avenue, New York, NY, 10461, USA.
- Department of Molecular Pharmacology, Einstein-Mount Sinai Diabetes Research Center (ES-DRC), Fleischer Institute for Diabetes and Metabolism (FIDAM), Einstein Institute for Aging Research, Albert Einstein College of Medicine, Montefiore University Hospital, New York, NY, 10461, USA.
| | - Stanislovas S Jankauskas
- Department of Medicine, Division of Cardiology, Wilf Family Cardiovascular Research Institute, Einstein Institute for Neuroimmunology and Inflammation (INI), Albert Einstein College of Medicine, Montefiore University Hospital, 1300 Morris Park Avenue, New York, NY, 10461, USA
| |
Collapse
|
2
|
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
|
3
|
Jiang H, Yang J, Li T, Wang X, Fan Z, Ye Q, Du Y. JAK/STAT3 signaling in cardiac fibrosis: a promising therapeutic target. Front Pharmacol 2024; 15:1336102. [PMID: 38495094 PMCID: PMC10940489 DOI: 10.3389/fphar.2024.1336102] [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: 11/10/2023] [Accepted: 01/18/2024] [Indexed: 03/19/2024] Open
Abstract
Cardiac fibrosis is a serious health problem because it is a common pathological change in almost all forms of cardiovascular diseases. Cardiac fibrosis is characterized by the transdifferentiation of cardiac fibroblasts (CFs) into cardiac myofibroblasts and the excessive deposition of extracellular matrix (ECM) components produced by activated myofibroblasts, which leads to fibrotic scar formation and subsequent cardiac dysfunction. However, there are currently few effective therapeutic strategies protecting against fibrogenesis. This lack is largely because the molecular mechanisms of cardiac fibrosis remain unclear despite extensive research. The Janus kinase/signal transducer and activator of transcription (JAK/STAT) signaling cascade is an extensively present intracellular signal transduction pathway and can regulate a wide range of biological processes, including cell proliferation, migration, differentiation, apoptosis, and immune response. Various upstream mediators such as cytokines, growth factors and hormones can initiate signal transmission via this pathway and play corresponding regulatory roles. STAT3 is a crucial player of the JAK/STAT pathway and its activation is related to inflammation, malignant tumors and autoimmune illnesses. Recently, the JAK/STAT3 signaling has been in the spotlight for its role in the occurrence and development of cardiac fibrosis and its activation can promote the proliferation and activation of CFs and the production of ECM proteins, thus leading to cardiac fibrosis. In this manuscript, we discuss the structure, transactivation and regulation of the JAK/STAT3 signaling pathway and review recent progress on the role of this pathway in cardiac fibrosis. Moreover, we summarize the current challenges and opportunities of targeting the JAK/STAT3 signaling for the treatment of fibrosis. In summary, the information presented in this article is critical for comprehending the role of the JAK/STAT3 pathway in cardiac fibrosis, and will also contribute to future research aimed at the development of effective anti-fibrotic therapeutic strategies targeting the JAK/STAT3 signaling.
Collapse
Affiliation(s)
- Heng Jiang
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Junjie Yang
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Tao Li
- Key Laboratory of Medical Electrophysiology, Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
| | - Xinyu Wang
- Key Laboratory of Medical Electrophysiology, Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
| | - Zhongcai Fan
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Qiang Ye
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Yanfei Du
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Key Laboratory of Medical Electrophysiology, Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
| |
Collapse
|
4
|
Ahn BY, Zhang Y, Wei S, Jeong Y, Park DH, Lee SJ, Leem YE, Kang JS. Prmt7 regulates the JAK/STAT/Socs3 signaling pathway in postmenopausal cardiomyopathy. Exp Mol Med 2024; 56:711-720. [PMID: 38486105 PMCID: PMC10985114 DOI: 10.1038/s12276-024-01193-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 12/07/2023] [Accepted: 01/02/2024] [Indexed: 04/04/2024] Open
Abstract
Protein arginine methyltransferases (PRMTs) modulate diverse cellular processes, including stress responses. The present study explored the role of Prmt7 in protecting against menopause-associated cardiomyopathy. Mice with cardiac-specific Prmt7 ablation (cKO) exhibited sex-specific cardiomyopathy. Male cKO mice exhibited impaired cardiac function, myocardial hypertrophy, and interstitial fibrosis associated with increased oxidative stress. Interestingly, female cKO mice predominantly exhibited comparable phenotypes only after menopause or ovariectomy (OVX). Prmt7 inhibition in cardiomyocytes exacerbated doxorubicin (DOX)-induced oxidative stress and DNA double-strand breaks, along with apoptosis-related protein expression. Treatment with 17β-estradiol (E2) attenuated the DOX-induced decrease in Prmt7 expression in cardiomyocytes, and Prmt7 depletion abrogated the protective effect of E2 against DOX-induced cardiotoxicity. Transcriptome analysis of ovariectomized wild-type (WT) or cKO hearts and mechanical analysis of Prmt7-deficient cardiomyocytes demonstrated that Prmt7 is required for the control of the JAK/STAT signaling pathway by regulating the expression of suppressor of cytokine signaling 3 (Socs3), which is a negative feedback inhibitor of the JAK/STAT signaling pathway. These data indicate that Prmt7 has a sex-specific cardioprotective effect by regulating the JAK/STAT signaling pathway and, ultimately, may be a potential therapeutic tool for heart failure treatment depending on sex.
Collapse
Affiliation(s)
- Byeong-Yun Ahn
- Department of Molecular Cell Biology, Sungkyunkwan University, School of Medicine, Suwon, Republic of Korea
| | - Yan Zhang
- Department of Molecular Cell Biology, Sungkyunkwan University, School of Medicine, Suwon, Republic of Korea
| | - Shibo Wei
- Department of Molecular Cell Biology, Sungkyunkwan University, School of Medicine, Suwon, Republic of Korea
| | - Yideul Jeong
- Research Institute of Aging-Related Diseases, AniMusCure, Inc, Suwon, Republic of Korea
| | - Dong-Hyun Park
- Department of Molecular Cell Biology, Sungkyunkwan University, School of Medicine, Suwon, Republic of Korea
| | - Sang-Jin Lee
- Research Institute of Aging-Related Diseases, AniMusCure, Inc, Suwon, Republic of Korea
| | - Young-Eun Leem
- Department of Molecular Cell Biology, Sungkyunkwan University, School of Medicine, Suwon, Republic of Korea.
| | - Jong-Sun Kang
- Department of Molecular Cell Biology, Sungkyunkwan University, School of Medicine, Suwon, Republic of Korea.
| |
Collapse
|
5
|
Casella C, Kiles F, Urquhart C, Michaud DS, Kirwa K, Corlin L. Methylomic, Proteomic, and Metabolomic Correlates of Traffic-Related Air Pollution in the Context of Cardiorespiratory Health: A Systematic Review, Pathway Analysis, and Network Analysis. TOXICS 2023; 11:1014. [PMID: 38133415 PMCID: PMC10748071 DOI: 10.3390/toxics11121014] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 11/18/2023] [Accepted: 12/06/2023] [Indexed: 12/23/2023]
Abstract
A growing body of literature has attempted to characterize how traffic-related air pollution (TRAP) affects molecular and subclinical biological processes in ways that could lead to cardiorespiratory disease. To provide a streamlined synthesis of what is known about the multiple mechanisms through which TRAP could lead to cardiorespiratory pathology, we conducted a systematic review of the epidemiological literature relating TRAP exposure to methylomic, proteomic, and metabolomic biomarkers in adult populations. Using the 139 papers that met our inclusion criteria, we identified the omic biomarkers significantly associated with short- or long-term TRAP and used these biomarkers to conduct pathway and network analyses. We considered the evidence for TRAP-related associations with biological pathways involving lipid metabolism, cellular energy production, amino acid metabolism, inflammation and immunity, coagulation, endothelial function, and oxidative stress. Our analysis suggests that an integrated multi-omics approach may provide critical new insights into the ways TRAP could lead to adverse clinical outcomes. We advocate for efforts to build a more unified approach for characterizing the dynamic and complex biological processes linking TRAP exposure and subclinical and clinical disease and highlight contemporary challenges and opportunities associated with such efforts.
Collapse
Affiliation(s)
- Cameron Casella
- Department of Public Health and Community Medicine, Tufts University School of Medicine, Boston, MA 02111, USA; (C.C.); (F.K.); (C.U.); (D.S.M.); (K.K.)
| | - Frances Kiles
- Department of Public Health and Community Medicine, Tufts University School of Medicine, Boston, MA 02111, USA; (C.C.); (F.K.); (C.U.); (D.S.M.); (K.K.)
| | - Catherine Urquhart
- Department of Public Health and Community Medicine, Tufts University School of Medicine, Boston, MA 02111, USA; (C.C.); (F.K.); (C.U.); (D.S.M.); (K.K.)
| | - Dominique S. Michaud
- Department of Public Health and Community Medicine, Tufts University School of Medicine, Boston, MA 02111, USA; (C.C.); (F.K.); (C.U.); (D.S.M.); (K.K.)
| | - Kipruto Kirwa
- Department of Public Health and Community Medicine, Tufts University School of Medicine, Boston, MA 02111, USA; (C.C.); (F.K.); (C.U.); (D.S.M.); (K.K.)
- Department of Environmental Health, Boston University School of Public Health, Boston, MA 02118, USA
| | - Laura Corlin
- Department of Public Health and Community Medicine, Tufts University School of Medicine, Boston, MA 02111, USA; (C.C.); (F.K.); (C.U.); (D.S.M.); (K.K.)
- Department of Civil and Environmental Engineering, Tufts University School of Engineering, Medford, MA 02155, USA
| |
Collapse
|
6
|
Kaesler N, Cheng M, Nagai J, O’Sullivan J, Peisker F, Bindels EM, Babler A, Moellmann J, Droste P, Franciosa G, Dugourd A, Saez-Rodriguez J, Neuss S, Lehrke M, Boor P, Goettsch C, Olsen JV, Speer T, Lu TS, Lim K, Floege J, Denby L, Costa I, Kramann R. Mapping cardiac remodeling in chronic kidney disease. SCIENCE ADVANCES 2023; 9:eadj4846. [PMID: 38000021 PMCID: PMC10672229 DOI: 10.1126/sciadv.adj4846] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Accepted: 10/24/2023] [Indexed: 11/26/2023]
Abstract
Patients with advanced chronic kidney disease (CKD) mostly die from sudden cardiac death and recurrent heart failure. The mechanisms of cardiac remodeling are largely unclear. To dissect molecular and cellular mechanisms of cardiac remodeling in CKD in an unbiased fashion, we performed left ventricular single-nuclear RNA sequencing in two mouse models of CKD. Our data showed a hypertrophic response trajectory of cardiomyocytes with stress signaling and metabolic changes driven by soluble uremia-related factors. We mapped fibroblast to myofibroblast differentiation in this process and identified notable changes in the cardiac vasculature, suggesting inflammation and dysfunction. An integrated analysis of cardiac cellular responses to uremic toxins pointed toward endothelin-1 and methylglyoxal being involved in capillary dysfunction and TNFα driving cardiomyocyte hypertrophy in CKD, which was validated in vitro and in vivo. TNFα inhibition in vivo ameliorated the cardiac phenotype in CKD. Thus, interventional approaches directed against uremic toxins, such as TNFα, hold promise to ameliorate cardiac remodeling in CKD.
Collapse
Affiliation(s)
- Nadine Kaesler
- Clinic for Renal and Hypertensive Disorders, Rheumatological and Immunological Disease, University Hospital of the RWTH Aachen, Aachen, Germany
- Institute of Experimental Medicine and Systems Biology, University Hospital of the RWTH Aachen, Aachen, Germany
| | - Mingbo Cheng
- Institute for Computational Genomics, University Hospital of the RWTH Aachen, Aachen, Germany
| | - James Nagai
- Institute for Computational Genomics, University Hospital of the RWTH Aachen, Aachen, Germany
| | - James O’Sullivan
- Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
| | - Fabian Peisker
- Institute of Experimental Medicine and Systems Biology, University Hospital of the RWTH Aachen, Aachen, Germany
| | - Eric M. J. Bindels
- Department of Hematology, Erasmus Medical Center, Rotterdam, Netherlands
| | - Anne Babler
- Institute of Experimental Medicine and Systems Biology, University Hospital of the RWTH Aachen, Aachen, Germany
| | - Julia Moellmann
- Department of Internal Medicine I, University Hospital of the RWTH Aachen, Aachen, Germany
| | - Patrick Droste
- Clinic for Renal and Hypertensive Disorders, Rheumatological and Immunological Disease, University Hospital of the RWTH Aachen, Aachen, Germany
- Institute of Pathology, University Hospital of the RWTH Aachen, Aachen, Germany
| | - Giulia Franciosa
- Novo Nordisk Foundation Center for Protein Research, University of Copenhagen, Copenhagen, Denmark
| | - Aurelien Dugourd
- Heidelberg University, Faculty of Medicine, and Heidelberg University Hospital, Institute for Computational Biomedicine, Bioquant, Heidelberg, Germany
| | - Julio Saez-Rodriguez
- Heidelberg University, Faculty of Medicine, and Heidelberg University Hospital, Institute for Computational Biomedicine, Bioquant, Heidelberg, Germany
| | - Sabine Neuss
- Institute of Pathology, University Hospital of the RWTH Aachen, Aachen, Germany
- Helmholtz Institute for Biomedical Engineering, Biointerface Laboratory, RWTH Aachen University, Aachen, Germany
| | - Michael Lehrke
- Department of Internal Medicine I, University Hospital of the RWTH Aachen, Aachen, Germany
| | - Peter Boor
- Clinic for Renal and Hypertensive Disorders, Rheumatological and Immunological Disease, University Hospital of the RWTH Aachen, Aachen, Germany
- Institute of Pathology, University Hospital of the RWTH Aachen, Aachen, Germany
| | - Claudia Goettsch
- Department of Internal Medicine I, University Hospital of the RWTH Aachen, Aachen, Germany
| | - Jesper V. Olsen
- Novo Nordisk Foundation Center for Protein Research, University of Copenhagen, Copenhagen, Denmark
| | - Thimoteus Speer
- Department of Medicine (Nephrology), Goethe University Frankfurt, Frankfurt, Germany
| | - Tzong-Shi Lu
- Brigham and Women’s Hospital, Renal Division, Boston, MA, USA
| | - Kenneth Lim
- Division of Nephrology and Hypertension, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Jürgen Floege
- Clinic for Renal and Hypertensive Disorders, Rheumatological and Immunological Disease, University Hospital of the RWTH Aachen, Aachen, Germany
| | - Laura Denby
- Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
| | - Ivan Costa
- Institute for Computational Genomics, University Hospital of the RWTH Aachen, Aachen, Germany
| | - Rafael Kramann
- Clinic for Renal and Hypertensive Disorders, Rheumatological and Immunological Disease, University Hospital of the RWTH Aachen, Aachen, Germany
- Institute of Experimental Medicine and Systems Biology, University Hospital of the RWTH Aachen, Aachen, Germany
- Department of Internal Medicine, Nephrology and Transplantation, Erasmus Medical Center, Rotterdam, Netherlands
| |
Collapse
|
7
|
Yang V, Kragstrup TW, McMaster C, Reid P, Singh N, Haysen SR, Robinson PC, Liew DFL. Managing Cardiovascular and Cancer Risk Associated with JAK Inhibitors. Drug Saf 2023; 46:1049-1071. [PMID: 37490213 PMCID: PMC10632271 DOI: 10.1007/s40264-023-01333-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/20/2023] [Indexed: 07/26/2023]
Abstract
Janus kinase inhibitors (JAKi) have enormous appeal as immune-modulating therapies across many chronic inflammatory diseases, but recently this promise has been overshadowed by questions regarding associated cardiovascular and cancer risk emerging from the ORAL Surveillance phase 3b/4 post-marketing requirement randomized controlled trial. In that study of patients with rheumatoid arthritis with existing cardiovascular risk, tofacitinib, the first JAKi registered for chronic inflammatory disease, failed to meet non-inferiority thresholds when compared with tumor necrosis factor inhibitors for both incident major adverse cardiovascular events and incident cancer. While this result was unexpected by many, subsequently published observational data have also supported this finding. Notably, however, such a risk has largely not yet been demonstrated in patients outside the specific clinical situation examined in the trial, even in the face of many studies examining this. Nevertheless, this signal has practically re-aligned approaches to both tofacitinib and other JAKi to varying extents, in other patient populations and contexts: within rheumatoid arthritis, but also in psoriatic arthritis, axial spondyloarthritis, inflammatory bowel disease, atopic dermatitis, and beyond. Application to individual patients can be more challenging but remains important to harness the substantive potential of JAKi to the maximum extent safely possible. This review not only explores the evolution of the regulatory response to the signal, its informing data, biological plausibility, and its impact on guidelines, but also the many factors that clinicians must consider in navigating cardiovascular and cancer risk for their patients considering JAKi as immune-modulating therapy.
Collapse
Affiliation(s)
- Victor Yang
- Department of Rheumatology, Level 1, North Wing, Heidelberg Repatriation Hospital, Austin Health, 300 Waterdale Road, PO Box 5444, Heidelberg West, VIC, 3081, Australia
| | - Tue W Kragstrup
- Department of Rheumatology, Aarhus University Hospital, Aarhus, Denmark
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- Sector for Rheumatology, Diagnostic Center, Silkeborg Regional Hospital, Silkeborg, Denmark
| | - Christopher McMaster
- Department of Rheumatology, Level 1, North Wing, Heidelberg Repatriation Hospital, Austin Health, 300 Waterdale Road, PO Box 5444, Heidelberg West, VIC, 3081, Australia
- Department of Clinical Pharmacology and Therapeutics, Austin Health, Melbourne, VIC, Australia
- Centre for Digital Transformation of Health, University of Melbourne, Melbourne, VIC, Australia
| | - Pankti Reid
- Division of Rheumatology and Committee on Clinical Pharmacology and Pharmacogenomics, Department of Medicine, University of Chicago Biological Sciences Division, Chicago, IL, USA
| | - Namrata Singh
- Division of Rheumatology, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Stine R Haysen
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Philip C Robinson
- Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia
- Royal Brisbane and Women's Hospital, Metro North Hospital and Health Service, Herston, QLD, Australia
| | - David F L Liew
- Department of Rheumatology, Level 1, North Wing, Heidelberg Repatriation Hospital, Austin Health, 300 Waterdale Road, PO Box 5444, Heidelberg West, VIC, 3081, Australia.
- Department of Clinical Pharmacology and Therapeutics, Austin Health, Melbourne, VIC, Australia.
- Department of Medicine, University of Melbourne, Melbourne, VIC, Australia.
| |
Collapse
|
8
|
Casella C, Kiles F, Urquhart C, Michaud DS, Kirwa K, Corlin L. Methylomic, proteomic, and metabolomic correlates of traffic-related air pollution: A systematic review, pathway analysis, and network analysis relating traffic-related air pollution to subclinical and clinical cardiorespiratory outcomes. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.09.30.23296386. [PMID: 37873294 PMCID: PMC10592990 DOI: 10.1101/2023.09.30.23296386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2023]
Abstract
A growing body of literature has attempted to characterize how traffic-related air pollution (TRAP) affects molecular and subclinical biological processes in ways that could lead to cardiorespiratory disease. To provide a streamlined synthesis of what is known about the multiple mechanisms through which TRAP could lead cardiorespiratory pathology, we conducted a systematic review of the epidemiological literature relating TRAP exposure to methylomic, proteomic, and metabolomic biomarkers in adult populations. Using the 139 papers that met our inclusion criteria, we identified the omic biomarkers significantly associated with short- or long-term TRAP and used these biomarkers to conduct pathway and network analyses. We considered the evidence for TRAP-related associations with biological pathways involving lipid metabolism, cellular energy production, amino acid metabolism, inflammation and immunity, coagulation, endothelial function, and oxidative stress. Our analysis suggests that an integrated multi-omics approach may provide critical new insights into the ways TRAP could lead to adverse clinical outcomes. We advocate for efforts to build a more unified approach for characterizing the dynamic and complex biological processes linking TRAP exposure and subclinical and clinical disease, and highlight contemporary challenges and opportunities associated with such efforts.
Collapse
Affiliation(s)
- Cameron Casella
- Department of Public Health and Community Medicine, Tufts University School of Medicine, Boston, MA 02111, USA
| | - Frances Kiles
- Department of Public Health and Community Medicine, Tufts University School of Medicine, Boston, MA 02111, USA
| | - Catherine Urquhart
- Department of Public Health and Community Medicine, Tufts University School of Medicine, Boston, MA 02111, USA
| | - Dominique S. Michaud
- Department of Public Health and Community Medicine, Tufts University School of Medicine, Boston, MA 02111, USA
| | - Kipruto Kirwa
- Department of Public Health and Community Medicine, Tufts University School of Medicine, Boston, MA 02111, USA
- Department of Environmental Health, Boston University School of Public Health, Boston, MA, 02118, USA
| | - Laura Corlin
- Department of Public Health and Community Medicine, Tufts University School of Medicine, Boston, MA 02111, USA
- Department of Civil and Environmental Engineering, Tufts University School of Engineering, Medford, MA 02155, USA
| |
Collapse
|
9
|
Kleinbongard P. Perspective: mitochondrial STAT3 in cardioprotection. Basic Res Cardiol 2023; 118:32. [PMID: 37620559 PMCID: PMC10449977 DOI: 10.1007/s00395-023-01003-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 08/07/2023] [Accepted: 08/09/2023] [Indexed: 08/26/2023]
Abstract
Activation of signal transducer and activator of transcription 3 (STAT3) has been identified as a key cardioprotective signal not only in animal studies but also in humans-in animals, STAT3 is causally involved in cardioprotection. In response to late ischemic conditioning, canonical function of STAT3 activation upregulates the expression of cardioprotective and anti-apoptotic proteins. In its non-canonical function, STAT3 is activated during ischemic conditioning and is part of the cardioprotective cytosolic survival activating factor enhancement pathway. Activated STAT3 is imported and localized to the mitochondria. Mitochondrial STAT3 stimulates the activity of mitochondrial electron transport chain complex I, reduces mitochondrial reactive oxygen species production and mitochondrial permeability transition pore opening. Finally, two novel aspects of STAT activation in cardioprotection are discussed: a genetic variance of the STAT encoding region as a potential primordial confounding variable for cardioprotection, and the cardioprotective potential of sodium-glucose cotransporter 2 inhibitors through STAT3 activation.
Collapse
Affiliation(s)
- Petra Kleinbongard
- Institute for Pathophysiology, West German Heart and Vascular Center, University of Essen Medical School, Hufelandstr. 55, 45122, Essen, Germany.
| |
Collapse
|
10
|
Rani A, Marsche G. A Current Update on the Role of HDL-Based Nanomedicine in Targeting Macrophages in Cardiovascular Disease. Pharmaceutics 2023; 15:1504. [PMID: 37242746 PMCID: PMC10221824 DOI: 10.3390/pharmaceutics15051504] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 05/10/2023] [Accepted: 05/12/2023] [Indexed: 05/28/2023] Open
Abstract
High-density lipoproteins (HDL) are complex endogenous nanoparticles involved in important functions such as reverse cholesterol transport and immunomodulatory activities, ensuring metabolic homeostasis and vascular health. The ability of HDL to interact with a plethora of immune cells and structural cells places it in the center of numerous disease pathophysiologies. However, inflammatory dysregulation can lead to pathogenic remodeling and post-translational modification of HDL, rendering HDL dysfunctional or even pro-inflammatory. Monocytes and macrophages play a critical role in mediating vascular inflammation, such as in coronary artery disease (CAD). The fact that HDL nanoparticles have potent anti-inflammatory effects on mononuclear phagocytes has opened new avenues for the development of nanotherapeutics to restore vascular integrity. HDL infusion therapies are being developed to improve the physiological functions of HDL and to quantitatively restore or increase the native HDL pool. The components and design of HDL-based nanoparticles have evolved significantly since their initial introduction with highly anticipated results in an ongoing phase III clinical trial in subjects with acute coronary syndrome. The understanding of mechanisms involved in HDL-based synthetic nanotherapeutics is critical to their design, therapeutic potential and effectiveness. In this review, we provide a current update on HDL-ApoA-I mimetic nanotherapeutics, highlighting the scope of treating vascular diseases by targeting monocytes and macrophages.
Collapse
Affiliation(s)
- Alankrita Rani
- Division of Pharmacology, Otto Loewi Research Center, Medical University of Graz, Neue Stiftingtalstrasse 6, 8010 Graz, Austria;
- BioTechMed-Graz, Mozartgasse 12/II, 8010 Graz, Austria
| | - Gunther Marsche
- Division of Pharmacology, Otto Loewi Research Center, Medical University of Graz, Neue Stiftingtalstrasse 6, 8010 Graz, Austria;
- BioTechMed-Graz, Mozartgasse 12/II, 8010 Graz, Austria
| |
Collapse
|
11
|
Effect of Selenium Nanoparticles and/or Bee Venom against STZ-Induced Diabetic Cardiomyopathy and Nephropathy. Metabolites 2023; 13:metabo13030400. [PMID: 36984840 PMCID: PMC10057804 DOI: 10.3390/metabo13030400] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 12/03/2022] [Accepted: 02/22/2023] [Indexed: 03/11/2023] Open
Abstract
The main purpose of our study was to examine the role of selenium nanoparticles (SeNPs) and/or bee venom (BV) in ameliorating diabetic cardiomyopathy (DCM) and nephropathy (DN) at the biochemical, histopathological and molecular levels. Fifty male albino rats were used in this experiment, divided into five groups: control, Streptozocin (STZ) diabetic, STZ-diabetic treated with SeNPs, STZ-diabetic treated with BV, and STZ-diabetic treated with SeNPs and BV. Biochemically, STZ injection resulted in a significant increase in serum glucose, BUN, creatinine, CRP, CK-MB, AST, LDH and cardiac troponins with a significant decrease in the serum insulin and albumin concentrations. Histopathologically, STZ injection resulted in diabetes, as revealed by glomerulonephritis, perivascular hemorrhage, inflammatory cell infiltrations and fibrosis, with widening of interstitial spaces of cardiomyocytes, loss of muscle cells continuity and some hyaline degeneration. At the molecular levels, the expression levels of miRNA 328, miRNA-21, TGFβ1, TGFβ1R, JAK1, STST-3, SMAD-1 and NFκβ genes were significantly up-regulated, whereas the expression levels of SMAD-7 were significantly down-regulated. It is concluded that SeNPs and/or BV administration ameliorates the deleterious effects resulting from STZ administration through improving the biochemical, histopathological and molecular effects, suggesting their protective role against the long-term diabetic complications of DCM and DN.
Collapse
|
12
|
Lozano J, Rai A, Lees JG, Fang H, Claridge B, Lim SY, Greening DW. Scalable Generation of Nanovesicles from Human-Induced Pluripotent Stem Cells for Cardiac Repair. Int J Mol Sci 2022; 23:14334. [PMID: 36430812 PMCID: PMC9696585 DOI: 10.3390/ijms232214334] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 11/03/2022] [Accepted: 11/15/2022] [Indexed: 11/22/2022] Open
Abstract
Extracellular vesicles (EVs) from stem cells have shown significant therapeutic potential to repair injured cardiac tissues and regulate pathological fibrosis. However, scalable generation of stem cells and derived EVs for clinical utility remains a huge technical challenge. Here, we report a rapid size-based extrusion strategy to generate EV-like membranous nanovesicles (NVs) from easily sourced human iPSCs in large quantities (yield 900× natural EVs). NVs isolated using density-gradient separation (buoyant density 1.13 g/mL) are spherical in shape and morphologically intact and readily internalised by human cardiomyocytes, primary cardiac fibroblasts, and endothelial cells. NVs captured the dynamic proteome of parental cells and include pluripotency markers (LIN28A, OCT4) and regulators of cardiac repair processes, including tissue repair (GJA1, HSP20/27/70, HMGB1), wound healing (FLNA, MYH9, ACTC1, ILK), stress response/translation initiation (eIF2S1/S2/S3/B4), hypoxia response (HMOX2, HSP90, GNB1), and extracellular matrix organization (ITGA6, MFGE8, ITGB1). Functionally, NVs significantly promoted tubule formation of endothelial cells (angiogenesis) (p < 0.05) and survival of cardiomyocytes exposed to low oxygen conditions (hypoxia) (p < 0.0001), as well as attenuated TGF-β mediated activation of cardiac fibroblasts (p < 0.0001). Quantitative proteome profiling of target cell proteome following NV treatments revealed upregulation of angiogenic proteins (MFGE8, MYH10, VDAC2) in endothelial cells and pro-survival proteins (CNN2, THBS1, IGF2R) in cardiomyocytes. In contrast, NVs attenuated TGF-β-driven extracellular matrix remodelling capacity in cardiac fibroblasts (ACTN1, COL1A1/2/4A2/12A1, ITGA1/11, THBS1). This study presents a scalable approach to generating functional NVs for cardiac repair.
Collapse
Affiliation(s)
- Jonathan Lozano
- Baker Heart and Diabetes Institute, Melbourne, VIC 3004, Australia
- Baker Department of Cardiovascular Research Translation and Implementation, La Trobe University, Melbourne, VIC 3086, Australia
- Department of Microbiology, Anatomy, Physiology and Pharmacology, School of Agriculture, Biomedicine and Environment, La Trobe University, Melbourne, VIC 3086, Australia
| | - Alin Rai
- Baker Heart and Diabetes Institute, Melbourne, VIC 3004, Australia
- Baker Department of Cardiovascular Research Translation and Implementation, La Trobe University, Melbourne, VIC 3086, Australia
- Baker Department of Cardiometabolic Health, University of Melbourne, Melbourne, VIC 3010, Australia
- Central Clinical School, Monash University, Melbourne, VIC 3004, Australia
| | - Jarmon G. Lees
- O’Brien Institute Department, St Vincent’s Institute of Medical Research, Melbourne, VIC 3065, Australia
- Department of Surgery and Medicine, University of Melbourne, Melbourne, VIC 3010, Australia
| | - Haoyun Fang
- Baker Heart and Diabetes Institute, Melbourne, VIC 3004, Australia
- Baker Department of Cardiometabolic Health, University of Melbourne, Melbourne, VIC 3010, Australia
| | - Bethany Claridge
- Baker Heart and Diabetes Institute, Melbourne, VIC 3004, Australia
- Department of Biochemistry and Chemistry, School of Agriculture, Biomedicine and Environment, La Trobe University, Melbourne, VIC 3086, Australia
| | - Shiang Y. Lim
- O’Brien Institute Department, St Vincent’s Institute of Medical Research, Melbourne, VIC 3065, Australia
- Department of Surgery and Medicine, University of Melbourne, Melbourne, VIC 3010, Australia
- National Heart Research Institute Singapore, National Heart Centre, Singapore 169609, Singapore
- Drug Discovery Biology, Faculty of Pharmacy and Pharmaceutical Sciences, Monash University, Melbourne, VIC 3800, Australia
| | - David W. Greening
- Baker Heart and Diabetes Institute, Melbourne, VIC 3004, Australia
- Baker Department of Cardiovascular Research Translation and Implementation, La Trobe University, Melbourne, VIC 3086, Australia
- Baker Department of Cardiometabolic Health, University of Melbourne, Melbourne, VIC 3010, Australia
- Central Clinical School, Monash University, Melbourne, VIC 3004, Australia
- Department of Biochemistry and Chemistry, School of Agriculture, Biomedicine and Environment, La Trobe University, Melbourne, VIC 3086, Australia
| |
Collapse
|
13
|
Sandenon Seteyen AL, Girard-Valenciennes E, Septembre-Malaterre A, Gasque P, Guiraud P, Sélambarom J. Anti-Alphaviral Alkaloids: Focus on Some Isoquinolines, Indoles and Quinolizidines. Molecules 2022; 27:molecules27165080. [PMID: 36014321 PMCID: PMC9416297 DOI: 10.3390/molecules27165080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 07/21/2022] [Accepted: 07/21/2022] [Indexed: 11/16/2022] Open
Abstract
The discovery and the development of safe and efficient therapeutics against arthritogenic alphaviruses (e.g., chikungunya virus) remain a continuous challenge. Alkaloids are structurally diverse and naturally occurring compounds in plants, with a wide range of biological activities including beneficial effects against prominent pathogenic viruses and inflammation. In this short review, we discuss the effects of some alkaloids of three biologically relevant structural classes (isoquinolines, indoles and quinolizidines). Based on various experimental models (viral infections and chronic diseases), we highlight the immunomodulatory effects of these alkaloids. The data established the capacity of these alkaloids to interfere in host antiviral and inflammatory responses through key components (antiviral interferon response, ROS production, inflammatory signaling pathways and pro- and anti-inflammatory cytokines production) also involved in alphavirus infection and resulting inflammation. Thus, these data may provide a convincing perspective of research for the use of alkaloids as immunomodulators against arthritogenic alphavirus infection and induced inflammation.
Collapse
Affiliation(s)
- Anne-Laure Sandenon Seteyen
- Unité de Recherche Etudes Pharmaco-Immunologiques (UR-EPI), Université de La Réunion, 97400 Saint-Denis, France
| | - Emmanuelle Girard-Valenciennes
- Laboratoire de Chimie et de Biotechnologie des Produits Naturels (CHEMBIOPRO), Université de La Réunion, 97400 Saint-Denis, France
| | - Axelle Septembre-Malaterre
- Centre Hospitalier Universitaire de La Réunion, Laboratoire d’Immunologie Clinique et Expérimentale de la Zone Océan Indien (LICE-OI), Pôle de Biologie, 97400 Saint-Denis, France
| | - Philippe Gasque
- Unité de Recherche Etudes Pharmaco-Immunologiques (UR-EPI), Université de La Réunion, 97400 Saint-Denis, France
- Centre Hospitalier Universitaire de La Réunion, Laboratoire d’Immunologie Clinique et Expérimentale de la Zone Océan Indien (LICE-OI), Pôle de Biologie, 97400 Saint-Denis, France
| | - Pascale Guiraud
- Unité de Recherche Etudes Pharmaco-Immunologiques (UR-EPI), Université de La Réunion, 97400 Saint-Denis, France
| | - Jimmy Sélambarom
- Unité de Recherche Etudes Pharmaco-Immunologiques (UR-EPI), Université de La Réunion, 97400 Saint-Denis, France
- Correspondence:
| |
Collapse
|
14
|
Beneficial Effects of O-GlcNAc Stimulation in a Young Rat Model of Sepsis: Beyond Modulation of Gene Expression. Int J Mol Sci 2022; 23:ijms23126430. [PMID: 35742875 PMCID: PMC9224386 DOI: 10.3390/ijms23126430] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 06/05/2022] [Accepted: 06/06/2022] [Indexed: 11/21/2022] Open
Abstract
The young population, which is particularly at risk of sepsis, is, paradoxically, rarely studied. Acute stimulation of O-GlcNAcylation, a post-translational modification involved in metabolic regulation, cell survival and stress response, is beneficial in young rats with sepsis. Considering that sepsis impacts the gene expression profile and that O-GlcNAcylation is a regulator of transcription, the aims of this study are to (i) unveil beneficial mechanisms of O-GlcNAcylation and (ii) decipher the relationship between O-GlcNAcylation and transcription during sepsis. Endotoxemic challenge was induced in 28-day-old male rats using a lipopolysaccharide injection (E. coli O111:B4, 20 mg·kg−1) and compared to control rats (NaCl 0.9%). One hour after, rats were assigned to no therapy or fluidotherapy (NaCl 0.9%, 10 mL.kg−1) ± NButGT (10 mg·kg−1) to stimulate O-GlcNAc levels. Cardiac O-GlcNAcylation levels were evaluated via Western blot and gene transcription using 3′ SRP analysis. Lipopolysaccharide injection favorizes inflammatory state with the overexpression of genes involved in the NF-κB, JAK/STAT and MAPK pathways. NButGT treatment increased cardiac O-GlcNAcylation levels (p < 0.05). Yet, the mRNA expression was not impacted two hours after fluidotherapy or NButGT treatment. In conclusion, O-GlcNAc stimulation-induced beneficial effects are not dependent on the gene expression profile at the early phase of sepsis.
Collapse
|
15
|
Iampanichakul M, Poasakate A, Potue P, Rattanakanokchai S, Maneesai P, Prachaney P, Settheetham-Ishida W, Pakdeechote P. Nobiletin resolves left ventricular and renal changes in 2K-1C hypertensive rats. Sci Rep 2022; 12:9289. [PMID: 35662276 PMCID: PMC9166784 DOI: 10.1038/s41598-022-13513-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2022] [Accepted: 05/25/2022] [Indexed: 01/09/2023] Open
Abstract
This study investigated the effects of nobiletin on cardiorenal changes and the underlying mechanisms involved in two-kidney, one-clip (2K-1C) hypertension. 2K-1C rats were treated with nobiletin (15 or 30 mg/kg/day) or losartan (10 mg/kg/day) for 4 weeks (n = 8/group). Nobiletin (30 mg/kg) reduced high levels of blood pressure and circulating angiotensin II and angiotensin-converting enzyme activity in 2K-1C rats. Left ventricular (LV) dysfunction and remodelling in 2K-1C rats were alleviated in the nobiletin-treated group (P < 0.05). Nobiletin reduced the upregulation of Ang II type I receptor (AT1R)/JAK (Janus kinase)/STAT (signal transducer and activator of transcription) protein expression in cardiac tissue of 2K-1C rats (P < 0.05). The reduction in kidney function, and accumulation of renal fibrosis in 2K-1C rats were alleviated by nobiletin (P < 0.05). Overexpression of AT1R and NADPH oxidase 4 (Nox4) protein in nonclipped kidney tissue was suppressed in the nobiletin-treated group (P < 0.05). The elevations in oxidative stress parameters and the reductions in antioxidant enzymes were attenuated in 2K-1C rats treated with nobiletin (P < 0.05). In summary, nobiletin had renin-angiotensin system inhibitory and antioxidant effects and attenuated LV dysfunction and remodelling via restoration of the AT1R/JAK/STAT pathway. Nobiletin also resolved renal damage that was related to modulation of the AT1R/Nox4 cascade in 2K-1C hypertension.
Collapse
Affiliation(s)
- Metee Iampanichakul
- grid.9786.00000 0004 0470 0856Department of Physiology, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40002 Thailand
| | - Anuson Poasakate
- grid.9786.00000 0004 0470 0856Department of Physiology, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40002 Thailand
| | - Prapassorn Potue
- grid.9786.00000 0004 0470 0856Department of Physiology, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40002 Thailand
| | - Siwayu Rattanakanokchai
- grid.9786.00000 0004 0470 0856Faculty of Veterinary Medicine, Khon Kaen University, Khon Kaen, 40002 Thailand
| | - Putcharawipa Maneesai
- grid.9786.00000 0004 0470 0856Department of Physiology, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40002 Thailand ,grid.9786.00000 0004 0470 0856Research Institute for Human High Performance and Health Promotion, Khon Kaen University, Khon Kaen, 40002 Thailand
| | - Parichat Prachaney
- grid.9786.00000 0004 0470 0856Department of Anatomy, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40002 Thailand
| | - Wannapa Settheetham-Ishida
- grid.9786.00000 0004 0470 0856Department of Physiology, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40002 Thailand
| | - Poungrat Pakdeechote
- grid.9786.00000 0004 0470 0856Department of Physiology, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40002 Thailand ,grid.9786.00000 0004 0470 0856Research Institute for Human High Performance and Health Promotion, Khon Kaen University, Khon Kaen, 40002 Thailand
| |
Collapse
|
16
|
La Manna S, Leone M, Mercurio FA, Florio D, Marasco D. Structure-Activity Relationship Investigations of Novel Constrained Chimeric Peptidomimetics of SOCS3 Protein Targeting JAK2. Pharmaceuticals (Basel) 2022; 15:ph15040458. [PMID: 35455455 PMCID: PMC9031227 DOI: 10.3390/ph15040458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 03/31/2022] [Accepted: 04/07/2022] [Indexed: 12/03/2022] Open
Abstract
SOCS3 (suppressor of cytokine signaling 3) protein suppresses cytokine-induced inflammation and its deletion in neurons or immune cells increases the pathological growth of blood vessels. Recently, we designed several SOCS3 peptidomimetics by assuming as template structures the interfacing regions of the ternary complex constituted by SOCS3, JAK2 (Janus Kinase 2) and gp130 (glycoprotein 130) proteins. A chimeric peptide named KIRCONG chim, including non-contiguous regions demonstrated able to bind to JAK2 and anti-inflammatory and antioxidant properties in VSMCs (vascular smooth muscle cells). With the aim to improve drug-like features of KIRCONG, herein we reported novel cyclic analogues bearing different linkages. In detail, in two of them hydrocarbon cycles of different lengths were inserted at positions i/i+5 and i/i+7 to improve helical conformations of mimetics. Structural features of cyclic compounds were investigated by CD (Circular Dichroism) and NMR (Nuclear Magnetic Resonance) spectroscopies while their ability to bind to catalytic domain of JAK2 was assessed through MST (MicroScale Thermophoresis) assay as well as their stability in biological serum. Overall data indicate a crucial role exerted by the length and the position of the cycle within the chimeric structure and could pave the way to the miniaturization of SOCS3 protein for therapeutic aims.
Collapse
Affiliation(s)
- Sara La Manna
- Department of Pharmacy, Research Center on Bioactive Peptides (CIRPEB), University of Naples “Federico II”, 80131 Naples, Italy; (S.L.M.); (D.F.)
| | - Marilisa Leone
- Institute of Biostructures and Bioimaging (CNR), 80145 Naples, Italy; (M.L.); (F.A.M.)
| | - Flavia Anna Mercurio
- Institute of Biostructures and Bioimaging (CNR), 80145 Naples, Italy; (M.L.); (F.A.M.)
| | - Daniele Florio
- Department of Pharmacy, Research Center on Bioactive Peptides (CIRPEB), University of Naples “Federico II”, 80131 Naples, Italy; (S.L.M.); (D.F.)
| | - Daniela Marasco
- Department of Pharmacy, Research Center on Bioactive Peptides (CIRPEB), University of Naples “Federico II”, 80131 Naples, Italy; (S.L.M.); (D.F.)
- Correspondence: ; Tel.: +39-0812534607
| |
Collapse
|
17
|
Khalifa AA, El Sokkary NH, Elblehi SS, Diab MA, Ali MA. Potential cardioprotective effect of octreotide via NOXs mitigation, mitochondrial biogenesis and MAPK/Erk1/2/STAT3/NF-kβ pathway attenuation in isoproterenol-induced myocardial infarction in rats. Eur J Pharmacol 2022; 925:174978. [DOI: 10.1016/j.ejphar.2022.174978] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Revised: 04/13/2022] [Accepted: 04/22/2022] [Indexed: 11/03/2022]
|
18
|
Moscoso I, Cebro-Márquez M, Martínez-Gómez Á, Abou-Jokh C, Martínez-Monzonís MA, Martínez-Sande JL, González-Melchor L, García-Seara J, Fernández-López XA, Moraña-Fernández S, González-Juanatey JR, Rodríguez-Mañero M, Lage R. Circulating miR-499a and miR-125b as Potential Predictors of Left Ventricular Ejection Fraction Improvement after Cardiac Resynchronization Therapy. Cells 2022; 11:cells11020271. [PMID: 35053387 PMCID: PMC8773679 DOI: 10.3390/cells11020271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 12/24/2021] [Accepted: 01/06/2022] [Indexed: 11/30/2022] Open
Abstract
Cardiac resynchronization therapy represents a therapeutic option for heart failure drug-refractory patients. However, due to the lack of success in 30% of the cases, there is a demand for an in-depth analysis of individual heterogeneity. In this study, we aimed to evaluate the prognostic value of circulating miRNA differences. Responder patients were defined by a composite endpoint of the presence of left ventricular reverse remodelling (a reduction ≥15% in telesystolic volume and an increment ≥10% in left ventricular ejection fraction). Circulating miRNAs signature was analysed at the time of the procedure and at a 6-month follow-up. An expression analysis showed, both at baseline and at follow-up, differences between responders and non-responders. Responders presented lower baseline expressions of miR-499, and at follow-up, downregulation of miR-125b-5p, both associated with a significant improvement in left ventricular ejection fraction. The miRNA profile differences showed a marked sensitivity to distinguish between responders and non-responders. Our data suggest that miRNA differences might contribute to prognostic stratification of patients undergoing cardiac resynchronization therapy and suggest that preimplant cardiac context as well as remodelling response are key to therapeutic success.
Collapse
Affiliation(s)
- Isabel Moscoso
- Cardiology Group, Centre for Research in Molecular Medicine and Chronic Diseases (CIMUS), Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain; (I.M.); (M.C.-M.); (J.R.G.-J.)
- Department of Cardiology and Coronary Unit and Cellular and Molecular Cardiology Research Unit, Institute of Biomedical Research (IDIS-SERGAS), University Clinical Hospital, 15706 Santiago de Compostela, Spain; (Á.M.-G.); (C.A.-J.); (M.A.M.-M.); (J.L.M.-S.); (L.G.-M.); (J.G.-S.); (X.A.F.-L.); (S.M.-F.); (M.R.-M.)
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain
| | - María Cebro-Márquez
- Cardiology Group, Centre for Research in Molecular Medicine and Chronic Diseases (CIMUS), Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain; (I.M.); (M.C.-M.); (J.R.G.-J.)
- Department of Cardiology and Coronary Unit and Cellular and Molecular Cardiology Research Unit, Institute of Biomedical Research (IDIS-SERGAS), University Clinical Hospital, 15706 Santiago de Compostela, Spain; (Á.M.-G.); (C.A.-J.); (M.A.M.-M.); (J.L.M.-S.); (L.G.-M.); (J.G.-S.); (X.A.F.-L.); (S.M.-F.); (M.R.-M.)
| | - Álvaro Martínez-Gómez
- Department of Cardiology and Coronary Unit and Cellular and Molecular Cardiology Research Unit, Institute of Biomedical Research (IDIS-SERGAS), University Clinical Hospital, 15706 Santiago de Compostela, Spain; (Á.M.-G.); (C.A.-J.); (M.A.M.-M.); (J.L.M.-S.); (L.G.-M.); (J.G.-S.); (X.A.F.-L.); (S.M.-F.); (M.R.-M.)
| | - Charigan Abou-Jokh
- Department of Cardiology and Coronary Unit and Cellular and Molecular Cardiology Research Unit, Institute of Biomedical Research (IDIS-SERGAS), University Clinical Hospital, 15706 Santiago de Compostela, Spain; (Á.M.-G.); (C.A.-J.); (M.A.M.-M.); (J.L.M.-S.); (L.G.-M.); (J.G.-S.); (X.A.F.-L.); (S.M.-F.); (M.R.-M.)
| | - María Amparo Martínez-Monzonís
- Department of Cardiology and Coronary Unit and Cellular and Molecular Cardiology Research Unit, Institute of Biomedical Research (IDIS-SERGAS), University Clinical Hospital, 15706 Santiago de Compostela, Spain; (Á.M.-G.); (C.A.-J.); (M.A.M.-M.); (J.L.M.-S.); (L.G.-M.); (J.G.-S.); (X.A.F.-L.); (S.M.-F.); (M.R.-M.)
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain
| | - José Luis Martínez-Sande
- Department of Cardiology and Coronary Unit and Cellular and Molecular Cardiology Research Unit, Institute of Biomedical Research (IDIS-SERGAS), University Clinical Hospital, 15706 Santiago de Compostela, Spain; (Á.M.-G.); (C.A.-J.); (M.A.M.-M.); (J.L.M.-S.); (L.G.-M.); (J.G.-S.); (X.A.F.-L.); (S.M.-F.); (M.R.-M.)
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain
| | - Laila González-Melchor
- Department of Cardiology and Coronary Unit and Cellular and Molecular Cardiology Research Unit, Institute of Biomedical Research (IDIS-SERGAS), University Clinical Hospital, 15706 Santiago de Compostela, Spain; (Á.M.-G.); (C.A.-J.); (M.A.M.-M.); (J.L.M.-S.); (L.G.-M.); (J.G.-S.); (X.A.F.-L.); (S.M.-F.); (M.R.-M.)
| | - Javier García-Seara
- Department of Cardiology and Coronary Unit and Cellular and Molecular Cardiology Research Unit, Institute of Biomedical Research (IDIS-SERGAS), University Clinical Hospital, 15706 Santiago de Compostela, Spain; (Á.M.-G.); (C.A.-J.); (M.A.M.-M.); (J.L.M.-S.); (L.G.-M.); (J.G.-S.); (X.A.F.-L.); (S.M.-F.); (M.R.-M.)
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain
| | - Xesús Alberte Fernández-López
- Department of Cardiology and Coronary Unit and Cellular and Molecular Cardiology Research Unit, Institute of Biomedical Research (IDIS-SERGAS), University Clinical Hospital, 15706 Santiago de Compostela, Spain; (Á.M.-G.); (C.A.-J.); (M.A.M.-M.); (J.L.M.-S.); (L.G.-M.); (J.G.-S.); (X.A.F.-L.); (S.M.-F.); (M.R.-M.)
| | - Sandra Moraña-Fernández
- Department of Cardiology and Coronary Unit and Cellular and Molecular Cardiology Research Unit, Institute of Biomedical Research (IDIS-SERGAS), University Clinical Hospital, 15706 Santiago de Compostela, Spain; (Á.M.-G.); (C.A.-J.); (M.A.M.-M.); (J.L.M.-S.); (L.G.-M.); (J.G.-S.); (X.A.F.-L.); (S.M.-F.); (M.R.-M.)
| | - José R. González-Juanatey
- Cardiology Group, Centre for Research in Molecular Medicine and Chronic Diseases (CIMUS), Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain; (I.M.); (M.C.-M.); (J.R.G.-J.)
- Department of Cardiology and Coronary Unit and Cellular and Molecular Cardiology Research Unit, Institute of Biomedical Research (IDIS-SERGAS), University Clinical Hospital, 15706 Santiago de Compostela, Spain; (Á.M.-G.); (C.A.-J.); (M.A.M.-M.); (J.L.M.-S.); (L.G.-M.); (J.G.-S.); (X.A.F.-L.); (S.M.-F.); (M.R.-M.)
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain
| | - Moisés Rodríguez-Mañero
- Department of Cardiology and Coronary Unit and Cellular and Molecular Cardiology Research Unit, Institute of Biomedical Research (IDIS-SERGAS), University Clinical Hospital, 15706 Santiago de Compostela, Spain; (Á.M.-G.); (C.A.-J.); (M.A.M.-M.); (J.L.M.-S.); (L.G.-M.); (J.G.-S.); (X.A.F.-L.); (S.M.-F.); (M.R.-M.)
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain
| | - Ricardo Lage
- Cardiology Group, Centre for Research in Molecular Medicine and Chronic Diseases (CIMUS), Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain; (I.M.); (M.C.-M.); (J.R.G.-J.)
- Department of Cardiology and Coronary Unit and Cellular and Molecular Cardiology Research Unit, Institute of Biomedical Research (IDIS-SERGAS), University Clinical Hospital, 15706 Santiago de Compostela, Spain; (Á.M.-G.); (C.A.-J.); (M.A.M.-M.); (J.L.M.-S.); (L.G.-M.); (J.G.-S.); (X.A.F.-L.); (S.M.-F.); (M.R.-M.)
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain
- Correspondence:
| |
Collapse
|
19
|
Singh L, Bhatti R. Cellular and molecular mechanisms involved in metabolic disorders. DRUG DELIVERY SYSTEMS FOR METABOLIC DISORDERS 2022:21-29. [DOI: 10.1016/b978-0-323-99616-7.00015-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
20
|
Varghese R, George Priya Doss C, Kumar RS, Almansour AI, Arumugam N, Efferth T, Ramamoorthy S. Cardioprotective effects of phytopigments via multiple signaling pathways. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2022; 95:153859. [PMID: 34856476 DOI: 10.1016/j.phymed.2021.153859] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 11/08/2021] [Accepted: 11/14/2021] [Indexed: 06/13/2023]
Abstract
BACKGROUND Cardiovascular diseases (CVDs) are among the deadliest non-communicable diseases, and millions of dollars are spent every year to combat CVDs. Unfortunately, the multifactorial etiology of CVDs complicates the development of efficient therapeutics. Interestingly, phytopigments show significant pleiotropic cardioprotective effects both in vitro and in vivo. PURPOSE This review gives an overview of the cardioprotective effects of phytopigments based on in vitro and in vivo studies as well as clinical trials. METHODS A literature-based survey was performed to collect the available data on cardioprotective activities of phytopigments via electronic search engines such as PubMed, Google Scholar, and Scopus. RESULTS Different classes of phytopigments such as carotenoids, xanthophylls, flavonoids, anthocyanins, anthraquinones alleviate major CVDs (e.g., cardiac hypertrophy, atherosclerosis, hypertension, cardiotoxicities) via acting on signaling pathways related to AMPK, NF-κB, NRF2, PPARs, AKT, TLRs, MAPK, JAK/STAT, NLRP3, TNF-α, and RA. CONCLUSION Phytopigments represent promising candidates to develop novel and effective CVD therapeutics. More randomized, placebo-controlled clinical studies are recommended to establish the clinical efficacy of phytopigments.
Collapse
Affiliation(s)
- Ressin Varghese
- School of Bio Sciences and Technology, VIT University, Vellore 632014, Tamil Nadu, India
| | - C George Priya Doss
- School of Bio Sciences and Technology, VIT University, Vellore 632014, Tamil Nadu, India
| | - Raju Suresh Kumar
- Department of Chemistry, College of Science, King Saud University, P. O. Box 2455, Riyadh 11451, Saudi Arabia
| | - Abdulrahman I Almansour
- Department of Chemistry, College of Science, King Saud University, P. O. Box 2455, Riyadh 11451, Saudi Arabia
| | - Natarajan Arumugam
- Department of Chemistry, College of Science, King Saud University, P. O. Box 2455, Riyadh 11451, Saudi Arabia
| | - Thomas Efferth
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University, Mainz, Germany
| | - Siva Ramamoorthy
- School of Bio Sciences and Technology, VIT University, Vellore 632014, Tamil Nadu, India.
| |
Collapse
|
21
|
Polyakova EA, Mikhaylov EN, Galagudza MM, Shlyakhto EV. Hyperleptinemia results in systemic inflammation and the exacerbation of ischemia-reperfusion myocardial injury. Heliyon 2021; 7:e08491. [PMID: 34901513 PMCID: PMC8640453 DOI: 10.1016/j.heliyon.2021.e08491] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2021] [Revised: 09/09/2021] [Accepted: 11/24/2021] [Indexed: 12/01/2022] Open
Abstract
Aim Hyperleptinemia potentiates the effects of many atherogenic factors, such as inflammation, platelet aggregation, migration, hypertrophy, proliferation of vascular smooth muscle cells, and endothelial cell dysfunction. The present study analysed the effects of long-term hyperleptinemia in an in vivo myocardial ischemia-reperfusion model to demonstrate whether the in vivo deleterious effect also affects cardiac structure and function. Main methods Rats were subcutaneously administered leptin for 8 days to estimate the involvement of the JAK/STAT pathway. Data from 58 male Wistar rats were included in the final analysis. Myocardial infarction (MI) was modelled by the 30-minute ligation of the main left coronary artery followed by 120-minute reperfusion. Hemodynamic measurements, electrocardiography monitoring, echocardiography, myocardial infarct size and area at risk, blood biochemical parameters, leptin, IL-6, TNF-alpha, FGF-21, and cardiomyocyte morphology were measured. The expression of JAK2, p-JAK2, STAT3, p-STAT3 was assessed by Western Blot analysis. Statistical analyses were performed using IBM SPSS Statistics v.26. Key findings Eight-day hyperleptinemia in rats leads to an increase in blood pressure and heart rate, myocardial hypertrophy, impaired LV function, the frequency of ischemic arrhythmias, dyslipidemia, systemic inflammation, and the size of induced myocardial infarction. Significance: The blockade of the JAK/STAT signalling pathway effectively reverses the negative effects of leptin, including increased blood pressure and total cholesterol.
Collapse
Affiliation(s)
- Ekaterina A Polyakova
- Almazov National Medical Research Centre, Institute of Experimental Medicine, Saint-Petersburg, Russian Federation
| | - Evgeny N Mikhaylov
- Almazov National Medical Research Centre, Institute of Experimental Medicine, Saint-Petersburg, Russian Federation
| | - Michael M Galagudza
- Almazov National Medical Research Centre, Institute of Experimental Medicine, Saint-Petersburg, Russian Federation
| | - Evgeny V Shlyakhto
- Almazov National Medical Research Centre, Institute of Experimental Medicine, Saint-Petersburg, Russian Federation
| |
Collapse
|
22
|
El-Sayed N, Mostafa YM, AboGresha NM, Ahmed AAM, Mahmoud IZ, El-Sayed NM. Dapagliflozin attenuates diabetic cardiomyopathy through erythropoietin up-regulation of AKT/JAK/MAPK pathways in streptozotocin-induced diabetic rats. Chem Biol Interact 2021; 347:109617. [PMID: 34391751 DOI: 10.1016/j.cbi.2021.109617] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Revised: 07/13/2021] [Accepted: 08/04/2021] [Indexed: 12/12/2022]
Abstract
PURPOSE This study was designed to investigate the mechanism of Dapagliflozin (Dapa) cardioprotection against diabetic cardiomyopathy (DCM). Structural and functional changes in the heart as well as decrease of erythropoietin (EPO) levels were reported in DCM. EPO simultaneously activates three pathways: the Janus-activated kinase-signal transducer and activator of transcription (JAK2/STAT5), phosphatidylinositol-3-kinase-Akt (PI3K/Akt), and extracellular signal-related kinase (ERK/MAPK) cascades, that result in proliferation and differentiation of cardiac cells. METHODS AND RESULTS DCM was induced by a high fat diet for 10 weeks followed by administration of streptozotocin. After confirmation of diabetes, rats were divided randomly to 5 groups: Group 1; normal control group, Group 2; untreated diabetic group and Groups (3-5); diabetic groups received Dapa daily (0.75 mg, 1.5 or 3 mg/Kg, p.o) respectively for a month. At the end of the experiment, full anaesthesia was induced in all rats using ether inhalation and ECG was recorded. Blood samples were collected then rats were sacrificed and their heart were dissected out and processed for biochemical and histopathological studies. Untreated diabetic rats showed abnormal ECG pattern, elevation of serum cardiac enzymes, decrease EPO levels, downregulation of P-Akt, P-JAK2 and pMAPK pathways, abnormal histological structure of the heart and increase immunostaining intensity of P53 and TNF α in the cardiomyocytes. Dapa in a dose dependent manner attenuated the alterations in the previously mentioned parameters. CONCLUSION The cardioprotective effect of Dapa could be mediated by increasing EPO levels and activation of P-Akt, P-JAK2 and pMAPK signalling cascades which in turn decrease apoptosis.
Collapse
Affiliation(s)
- Nora El-Sayed
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Sinai University, Kantra Branch, Ismailia, Egypt
| | - Yasser M Mostafa
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Suez Canal University, Ismailia, 41522, Egypt; Department of Pharmacology & Toxicology, Faculty of Pharmacy, Badr University, Badr, Egypt
| | - Noha M AboGresha
- Department of Physiology, Faculty of Medicine, Suez Canal University, Ismailia, Egypt
| | - Amal A M Ahmed
- Department of Cytology & Histology, Faculty of Veterinary Medicine, Suez Canal University, Ismailia, Egypt
| | - Islam Z Mahmoud
- Department of Cardiovascular Medicine, Faculty of Medicine, Suez Canal University, Ismailia, Egypt
| | - Norhan M El-Sayed
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Suez Canal University, Ismailia, 41522, Egypt.
| |
Collapse
|
23
|
Mahjoubin-Tehran M, Atkin SL, Bezsonov EE, Jamialahmadi T, Sahebkar A. Harnessing the Therapeutic Potential of Decoys in Non-Atherosclerotic Cardiovascular Diseases: State of the Art. J Cardiovasc Dev Dis 2021; 8:jcdd8090103. [PMID: 34564121 PMCID: PMC8467637 DOI: 10.3390/jcdd8090103] [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: 06/29/2021] [Revised: 08/06/2021] [Accepted: 08/20/2021] [Indexed: 01/30/2023] Open
Abstract
Cardiovascular disease (CVD) is the main cause of global death, highlighting the fact that conventional therapeutic approaches for the treatment of CVD patients are insufficient, and there is a need to develop new therapeutic approaches. In recent years, decoy technology, decoy oligodeoxynucleotides (ODN), and decoy peptides show promising results for the future treatment of CVDs. Decoy ODN inhibits transcription by binding to the transcriptional factor, while decoy peptide neutralizes receptors by binding to the ligands. This review focused on studies that have investigated the effects of decoy ODN and decoy peptides on non-atherosclerotic CVD.
Collapse
Affiliation(s)
- Maryam Mahjoubin-Tehran
- Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran;
- Department of Medical Biotechnology and Nanotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Evgeny E. Bezsonov
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, 8 Baltiiskaya Street, 125315 Moscow, Russia;
- Laboratory of Cellular and Molecular Pathology of Cardiovascular System, Institute of Human Morphology, Moscow, Russia
- Department of Biology and General Genetics, I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| | - Tannaz Jamialahmadi
- Department of Food Science and Technology, Quchan Branch, Islamic Azad University, Quchan, Iran;
- Department of Nutrition, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- School of Medicine, The University of Western Australia, Perth, Australia
- School of Pharmacy, Mashhad University of Medical Sciences, Mashhad 9177948954, Iran
- Correspondence: or
| |
Collapse
|
24
|
The Role of HECT-Type E3 Ligase in the Development of Cardiac Disease. Int J Mol Sci 2021; 22:ijms22116065. [PMID: 34199773 PMCID: PMC8199989 DOI: 10.3390/ijms22116065] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 05/26/2021] [Accepted: 06/01/2021] [Indexed: 12/12/2022] Open
Abstract
Despite advances in medicine, cardiac disease remains an increasing health problem associated with a high mortality rate. Maladaptive cardiac remodeling, such as cardiac hypertrophy and fibrosis, is a risk factor for heart failure; therefore, it is critical to identify new therapeutic targets. Failing heart is reported to be associated with hyper-ubiquitylation and impairment of the ubiquitin–proteasome system, indicating an importance of ubiquitylation in the development of cardiac disease. Ubiquitylation is a post-translational modification that plays a pivotal role in protein function and degradation. In 1995, homologous to E6AP C-terminus (HECT) type E3 ligases were discovered. E3 ligases are key enzymes in ubiquitylation and are classified into three families: really interesting new genes (RING), HECT, and RING-between-RINGs (RBRs). Moreover, 28 HECT-type E3 ligases have been identified in human beings. It is well conserved in evolution and is characterized by the direct attachment of ubiquitin to substrates. HECT-type E3 ligase is reported to be involved in a wide range of human diseases and health. The role of HECT-type E3 ligases in the development of cardiac diseases has been uncovered in the last decade. There are only a few review articles summarizing recent advancements regarding HECT-type E3 ligase in the field of cardiac disease. This study focused on cardiac remodeling and described the role of HECT-type E3 ligases in the development of cardiac disease. Moreover, this study revealed that the current knowledge could be exploited for the development of new clinical therapies.
Collapse
|
25
|
Integrated transcriptomics and epigenomics reveal chamber-specific and species-specific characteristics of human and mouse hearts. PLoS Biol 2021; 19:e3001229. [PMID: 34003819 PMCID: PMC8130971 DOI: 10.1371/journal.pbio.3001229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Accepted: 04/12/2021] [Indexed: 12/02/2022] Open
Abstract
DNA methylation, chromatin accessibility, and gene expression represent different levels information in biological process, but a comprehensive multiomics analysis of the mammalian heart is lacking. Here, we applied nucleosome occupancy and methylome sequencing, which detected DNA methylation and chromatin accessibility simultaneously, as well as RNA-seq, for multiomics analysis of the 4 chambers of adult and fetal human hearts, and adult mouse hearts. Our results showed conserved region-specific patterns in the mammalian heart at transcriptome and DNA methylation level. Adult and fetal human hearts showed distinct features in DNA methylome, chromatin accessibility, and transcriptome. Novel long noncoding RNAs were identified in the human heart, and the gene expression profiles of major cardiovascular diseases associated genes were displayed. Furthermore, cross-species comparisons revealed human-specific and mouse-specific differentially expressed genes between the atria and ventricles. We also reported the relationship among multiomics and found there was a bell-shaped relationship between gene-body methylation and expression in the human heart. In general, our study provided comprehensive spatiotemporal and evolutionary insights into the regulation of gene expression in the heart. Multi-omic analyses of the four chambers of the human and mouse heart, including transcriptome, DNA methylation and chromatin accessibility, reveals characteristic patterns of gene regulation at the level of heart regions.
Collapse
|
26
|
Zhang X, van Rooij JGJ, Wakabayashi Y, Hwang SJ, Yang Y, Ghanbari M, Bos D, Levy D, Johnson AD, van Meurs JBJ, Kavousi M, Zhu J, O'Donnell CJ. Genome-wide transcriptome study using deep RNA sequencing for myocardial infarction and coronary artery calcification. BMC Med Genomics 2021; 14:45. [PMID: 33568140 PMCID: PMC7874462 DOI: 10.1186/s12920-020-00838-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Accepted: 11/29/2020] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Coronary artery calcification (CAC) is a noninvasive measure of coronary atherosclerosis, the proximal pathophysiology underlying most cases of myocardial infarction (MI). We sought to identify expression signatures of early MI and subclinical atherosclerosis in the Framingham Heart Study (FHS). In this study, we conducted paired-end RNA sequencing on whole blood collected from 198 FHS participants (55 with a history of early MI, 72 with high CAC without prior MI, and 71 controls free of elevated CAC levels or history of MI). We applied DESeq2 to identify coding-genes and long intergenic noncoding RNAs (lincRNAs) differentially expressed in MI and high CAC, respectively, compared with the control. RESULTS On average, 150 million paired-end reads were obtained for each sample. At the false discovery rate (FDR) < 0.1, we found 68 coding genes and 2 lincRNAs that were differentially expressed in early MI versus controls. Among them, 60 coding genes were detectable and thus tested in an independent RNA-Seq data of 807 individuals from the Rotterdam Study, and 8 genes were supported by p value and direction of the effect. Immune response, lipid metabolic process, and interferon regulatory factor were enriched in these 68 genes. By contrast, only 3 coding genes and 1 lincRNA were differentially expressed in high CAC versus controls. APOD, encoding a component of high-density lipoprotein, was significantly downregulated in both early MI (FDR = 0.007) and high CAC (FDR = 0.01) compared with controls. CONCLUSIONS We identified transcriptomic signatures of early MI that include differentially expressed protein-coding genes and lincRNAs, suggesting important roles for protein-coding genes and lincRNAs in the pathogenesis of MI.
Collapse
Affiliation(s)
- Xiaoling Zhang
- Division of Intramural Research, National Heart, Lung and Blood Institute, Bethesda, MD, USA.
- The National Heart, Lung and Blood Institute's Framingham Heart Study, Framingham, MA, USA.
- Department of Medicine (Biomedical Genetics), Boston University School of Medicine, 72 East Concord Street, Boston, MA, 02118-2526, USA.
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA.
| | - Jeroen G J van Rooij
- Department of Internal Medicine, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Yoshiyuki Wakabayashi
- DNA Sequencing and Genomics Core, National Heart, Lung and Blood Institute, Bethesda, MD, USA
| | - Shih-Jen Hwang
- Division of Intramural Research, National Heart, Lung and Blood Institute, Bethesda, MD, USA
- The National Heart, Lung and Blood Institute's Framingham Heart Study, Framingham, MA, USA
| | - Yanqin Yang
- DNA Sequencing and Genomics Core, National Heart, Lung and Blood Institute, Bethesda, MD, USA
| | - Mohsen Ghanbari
- Department of Internal Medicine, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Daniel Bos
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, the Netherlands
- Department of Radiology and Nuclear Medicine, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Daniel Levy
- Division of Intramural Research, National Heart, Lung and Blood Institute, Bethesda, MD, USA
- The National Heart, Lung and Blood Institute's Framingham Heart Study, Framingham, MA, USA
| | - Andrew D Johnson
- Division of Intramural Research, National Heart, Lung and Blood Institute, Bethesda, MD, USA
- The National Heart, Lung and Blood Institute's Framingham Heart Study, Framingham, MA, USA
| | - Joyce B J van Meurs
- Department of Internal Medicine, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Maryam Kavousi
- Department of Internal Medicine, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Jun Zhu
- DNA Sequencing and Genomics Core, National Heart, Lung and Blood Institute, Bethesda, MD, USA
| | - Christopher J O'Donnell
- Division of Intramural Research, National Heart, Lung and Blood Institute, Bethesda, MD, USA.
- The National Heart, Lung and Blood Institute's Framingham Heart Study, Framingham, MA, USA.
- Cardiology Section, Veteran's Administration Boston Healthcare System, Boston, USA.
| |
Collapse
|
27
|
In silico analysis of non-coding RNAs and putative target genes implicated in metabolic syndrome. Comput Biol Med 2021; 130:104229. [PMID: 33516961 DOI: 10.1016/j.compbiomed.2021.104229] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2020] [Revised: 01/14/2021] [Accepted: 01/15/2021] [Indexed: 01/12/2023]
Abstract
Regulation of gene expression is vital to maintain normal cellular functions and its dysregulation leads to molecular pathogenesis of many diseases and disorders. Non-coding RNAs regulate the expression of approximately 60% of protein-coding genes and their malfunction contribute to the development of numerous diseases. The involvement of variant forms of circulating non-coding RNAs in diseases has been established. However, their function as biomarkers or therapeutic targets in metabolic disorders are underexploited. The aim of this study was to predict therapeutic targets and construction of biomarker panel for early detection of metabolic syndrome (MS). Non-coding RNAs including circular RNAs (circRNAs), long chain non-coding RNAs (lncRNA) and micro RNAs (miRNAs) were extracted from intensive literature search and experimentally supported databases. Raw data of gene expression profiles of MS were obtained from the GEO dataset and analyzed to get differentially expressed genes (DEGs). Functional enrichment analysis, network illustration of non-coding RNAs and predicted target DEGs were performed. Furthermore, a few numbers of miRNAs targeted DEGs were subjected to homology study. The strong association of hsa-miR-548c-3p, hsa-miR-579-3p, hsa-miR-17-5p and hsa-miR-320a was observed with the pathogenesis of MS. It includes the regulation of genes in glucose and lipid homeostasis, MAPKK activity, regulation of inflammatory responses and many signaling pathways such as insulin resistance, JAK/STAT and mTOR. Finally, interactions of hsa-miR-17-5p:STAT3, hsa-miR-320:JAK2, hsa-miR-320:S6K and hsa-let-7:DVL hybrids were predicted. Results of this study suggest the designing of a biomarker panel to detect early onset and molecular approach for the management of MS.
Collapse
|
28
|
Patel NJ, Nassal DM, Gratz D, Hund TJ. Emerging therapeutic targets for cardiac arrhythmias: role of STAT3 in regulating cardiac fibroblast function. Expert Opin Ther Targets 2020; 25:63-73. [PMID: 33170045 DOI: 10.1080/14728222.2021.1849145] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Introduction : Cardiac fibrosis contributes to the development of cardiovascular disease (CVD) and arrhythmia. Cardiac fibroblasts (CFs) are collagen-producing cells that regulate extracellular matrix (ECM) homeostasis. A complex signaling network has been defined linking environmental stress to changes in CF function and fibrosis. Signal Transducer and Activator of Transcription 3 (STAT3) has emerged as a critical integrator of pro-fibrotic signals in CFs downstream of several established signaling networks. Areas covered : This article provides an overview of STAT3 function in CFs and its involvement in coordinating a vast web of intracellular pro-fibrotic signaling molecules and transcription factors. We highlight recent work elucidating a critical role for the fibroblast cytoskeleton in maintaining spatial and temporal control of STAT3-related signaling . Finally, we discuss potential opportunities and obstacles for therapeutic targeting of STAT3 to modulate cardiac fibrosis and arrhythmias. Relevant publications on the topic were identified through Pubmed. Expert opinion : Therapeutic targeting of STAT3 for CVD and arrhythmias presents unique challenges and opportunities. Thus, it is critical to consider the multimodal and dynamic nature of STAT3 signaling. Going forward, it will be beneficial to consider ways to maintain balanced STAT3 function, rather than large-scale perturbations in STAT3 function.
Collapse
Affiliation(s)
- Nehal J Patel
- The Frick Center for Heart Failure and Arrhythmia, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center , Columbus, OH, USA.,Department of Biomedical Engineering, College of Engineering, the Ohio State University , Columbus, OH, USA
| | - Drew M Nassal
- The Frick Center for Heart Failure and Arrhythmia, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center , Columbus, OH, USA.,Department of Biomedical Engineering, College of Engineering, the Ohio State University , Columbus, OH, USA
| | - Daniel Gratz
- The Frick Center for Heart Failure and Arrhythmia, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center , Columbus, OH, USA.,Department of Biomedical Engineering, College of Engineering, the Ohio State University , Columbus, OH, USA
| | - Thomas J Hund
- The Frick Center for Heart Failure and Arrhythmia, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center , Columbus, OH, USA.,Department of Biomedical Engineering, College of Engineering, the Ohio State University , Columbus, OH, USA.,Department of Internal Medicine, The Ohio State University Wexner Medical Center , Columbus, OH, USA
| |
Collapse
|
29
|
Zhao L, Feng S, Wang S, Fan M, Jin W, Li X, Wang C, Yang Y. Production of bioactive recombinant human myeloid-derived growth factor in Escherichia coli and its mechanism on vascular endothelial cell proliferation. J Cell Mol Med 2020; 24:1189-1199. [PMID: 31758636 PMCID: PMC6991672 DOI: 10.1111/jcmm.14602] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Revised: 07/25/2019] [Accepted: 07/26/2019] [Indexed: 12/22/2022] Open
Abstract
Myeloid-derived growth factor (MYDGF) is a novel protein secreted by bone marrow cells that features important physiological functions. In recent years, MYDGF has gained considerable interest due to their extensive beneficial effect on cardiac repair and protects cardiomyocytes from cell death. However, its precise molecular mechanisms have not been well elucidated. The purpose of this study was to produce sufficient amount of biologically active recombinant human (rh) MYDGF more economically and effectively by using in vitro molecular cloning techniques to study its clinical application. The prokaryotic expression system of Escherichia coli was established for the preparation of rhMYDGF. Finally, a large amount of high biologically active and purified form of recombinant protein was obtained. Moreover, we investigated the potential mechanism of rhMYDGF-mediated proliferation and survival in human coronary artery endothelial cells (HCAECs). Mechanistically, the results suggested that MAPK/STAT3 and the cyclin D1 signalling pathways are indispensable for rhMYDGF-mediated HCAEC proliferation and survival. Therefore, this study successfully established a preparation protocol for biologically active rhMYDGF and it may be a most economical way to produce high-quality active rhMYDGF for future clinical application.
Collapse
Affiliation(s)
- Longwei Zhao
- School of Life Science and TechnologyChina Pharmaceutical UniversityNanjingChina
- Center for New Drug Safety Evaluation and ResearchChina Pharmaceutical UniversityNanjingChina
| | - Shuang Feng
- Center for New Drug Safety Evaluation and ResearchChina Pharmaceutical UniversityNanjingChina
| | - Shen Wang
- School of Life Science and TechnologyChina Pharmaceutical UniversityNanjingChina
| | - Miaojuan Fan
- School of Pharmaceutical Sciences & Center for Structural BiologyWenzhou Medical UniversityWenzhouZhejiangChina
| | - Wei Jin
- School of Pharmaceutical Sciences & Center for Structural BiologyWenzhou Medical UniversityWenzhouZhejiangChina
| | - Xianjing Li
- School of Life Science and TechnologyChina Pharmaceutical UniversityNanjingChina
- Center for New Drug Safety Evaluation and ResearchChina Pharmaceutical UniversityNanjingChina
| | - Chen Wang
- School of Life Science and TechnologyChina Pharmaceutical UniversityNanjingChina
| | - Yong Yang
- School of Life Science and TechnologyChina Pharmaceutical UniversityNanjingChina
- Center for New Drug Safety Evaluation and ResearchChina Pharmaceutical UniversityNanjingChina
| |
Collapse
|
30
|
Coppini R, Santini L, Palandri C, Sartiani L, Cerbai E, Raimondi L. Pharmacological Inhibition of Serine Proteases to Reduce Cardiac Inflammation and Fibrosis in Atrial Fibrillation. Front Pharmacol 2019; 10:1420. [PMID: 31956307 PMCID: PMC6951407 DOI: 10.3389/fphar.2019.01420] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Accepted: 11/07/2019] [Indexed: 12/18/2022] Open
Abstract
Systemic inflammation correlates with an increased risk of atrial fibrillation (AF) and thrombogenesis. Systemic inflammation alters vessel permeability, allowing inflammatory and immune cell migration toward target organs, including the heart. Among inflammatory cells infiltrating the atria, macrophages and mast cell have recently attracted the interest of basic researchers due to the pathogenic mechanisms triggered by their activation. This chemotactic invasion is likely implicated in short- and long-term changes in cardiac cell-to-cell communication and in triggering fibrous tissue accumulation in the atrial myocardium and electrophysiological re-arrangements of atrial cardiomyocytes, thus favoring the onset and progression of AF. Serine proteases are a large and heterogeneous class of proteases involved in several processes that are important for cardiac function and are involved in cardiac diseases, such as (i) coagulation, (ii) fibrinolysis, (iii) extracellular matrix degradation, (iv) activation of receptors (i.e., protease-activated receptors [PPARs]), and (v) modulation of the activity of endogenous signals. The recognition of serine proteases substrates and their involvement in inflammatory/profibrotic mechanisms allowed the identification of novel cardio-protective mechanisms for commonly used drugs that inhibit serine proteases. The aim of this review is to summarize knowledge on the role of inflammation and fibrosis as determinants of AF. Moreover, we will recapitulate current findings on the role of serine proteases in the pathogenesis of AF and the possible beneficial effects of drugs inhibiting serine proteases in reducing the risk of AF through decrease of cardiac inflammation and fibrosis. These drugs include thrombin and factor Xa inhibitors (used as oral anticoagulants), dipeptidyl-peptidase 4 (DPP4) inhibitors, used for type-2 diabetes, as well as novel experimental inhibitors of mast cell chymases.
Collapse
Affiliation(s)
- Raffaele Coppini
- Section of Pharmacology, Department of Neurology, Psychology, Drug Sciences and Child Health, University of Florence, Florence, Italy
| | - Lorenzo Santini
- Section of Pharmacology, Department of Neurology, Psychology, Drug Sciences and Child Health, University of Florence, Florence, Italy
| | - Chiara Palandri
- Section of Pharmacology, Department of Neurology, Psychology, Drug Sciences and Child Health, University of Florence, Florence, Italy
| | - Laura Sartiani
- Section of Pharmacology, Department of Neurology, Psychology, Drug Sciences and Child Health, University of Florence, Florence, Italy
| | - Elisabetta Cerbai
- Section of Pharmacology, Department of Neurology, Psychology, Drug Sciences and Child Health, University of Florence, Florence, Italy
| | - Laura Raimondi
- Section of Pharmacology, Department of Neurology, Psychology, Drug Sciences and Child Health, University of Florence, Florence, Italy
| |
Collapse
|
31
|
Bucindolol Modulates Cardiac Remodeling by Attenuating Oxidative Stress in H9c2 Cardiac Cells Exposed to Norepinephrine. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:6325424. [PMID: 31360296 PMCID: PMC6652037 DOI: 10.1155/2019/6325424] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Revised: 05/12/2019] [Accepted: 06/15/2019] [Indexed: 11/18/2022]
Abstract
The increased circulation of norepinephrine, found in the diseased heart as a result of sympathetic nervous system overactivation, is responsible for its cardiotoxic effects including pathological hypertrophy, cell death, and oxidative stress. Bucindolol is a third generation adrenergic blocker, which acts on the β1 and β2 receptors, and has additional α1 antagonist activity. Thus, the aim of this study was to investigate the action of bucindolol on oxidative stress, hypertrophy, cell survival, and cell death signaling pathways in H9c2 cardiac cells exposed to norepinephrine. H9c2 cells were incubated with 10 μM norepinephrine for 24 h in the presence or absence of bucindolol (10 μM) treatment for 8 h. Western blot was used to determine the expression of proteins for hypertrophy/survival and death signaling pathways. Flow cytometry was used to assess cell death via caspase-3/7 activity and propidium iodide and reactive oxygen species via measuring the fluorescence of CM-H2DCFDA. Norepinephrine exposure resulted in an increase in oxidative stress as well as cell death. This was accompanied by an increased protein expression of LC3B-II/I. The protein kinase B/mammalian target of the rapamycin (Akt/mTOR) pathway which is involved in cardiac remodeling process was activated in response to norepinephrine and was mitigated by bucindolol. In conclusion, bucindolol was able to modulate cardiac remodeling which is mediated by oxidative stress.
Collapse
|
32
|
Abu-Halima M, Kahraman M, Henn D, Rädle-Hurst T, Keller A, Abdul-Khaliq H, Meese E. Deregulated microRNA and mRNA expression profiles in the peripheral blood of patients with Marfan syndrome. J Transl Med 2018. [PMID: 29530068 PMCID: PMC5848586 DOI: 10.1186/s12967-018-1429-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Background MicroRNAs (miRNAs) are small RNAs regulating gene expression post-transcriptionally. While acquired changes of miRNA and mRNA profiles in cancer have been extensively studied, little is known about expression changes of circulating miRNAs and messenger RNAs (mRNA) in monogenic constitutional anomalies affecting several organ systems, like Marfan syndrome (MFS). We performed integrated miRNA and mRNA expression profiling in blood samples of Marfan patients in order to investigate deregulated miRNA and mRNA networks in these patients which could serve as potential diagnostic and prognostic tools for MFS therapy. Methods MiRNA and mRNA expression profiles were determined in blood samples from MFS patients (n = 7) and from healthy volunteer controls (n = 7) by microarray analysis. Enrichment analyses of altered mRNA expression were identified using bioinformatic tools. Results A total of 28 miRNAs and 32 mRNAs were found to be significantly altered in MFS patients compared to controls (> 2.0-fold change, adjusted P < 0.05). The expression of 11 miRNA and 6 mRNA candidates was validated by RT-qPCR in an independent cohort of 26 MFS patients and 26 matched HV controls. Significant inverse correlations were evident between 8 miRNAs and 5 mRNAs involved in vascular pathology, inflammation and telomerase regulation. Significant positive correlations were present for 7 miRNAs with age, for 2 miRNAs with the MFS aortic root status (Z-score) and for 7 miRNAs with left ventricular end-diastolic diameter in MFS patients. In addition, miR-331-3p was significantly up-regulated in MFS patients without mitral valve prolapse (MVP) as compared with patients with MVP. Conclusions Our data show deregulated gene and miRNA expression profiles in the peripheral blood of MFS patients, demonstrating several candidates for prognostic biomarkers for cardiovascular manifestations in MFS as well as targets for novel therapeutic approaches. A deregulation of miRNA expression seems to play an important role in MFS, highlighting the plethora of effects on post-transcriptional regulation of miRNAs and mRNAs initiated by constitutional mutations in single genes. Trial registration Nr: EA2/131/10. Registered 28 December, 2010 Electronic supplementary material The online version of this article (10.1186/s12967-018-1429-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Masood Abu-Halima
- Institute of Human Genetics, Saarland University, 66421, Homburg/Saar, Germany.
| | - Mustafa Kahraman
- Chair for Clinical Bioinformatics, Saarland University, 66041, Saarbrücken, Germany
| | - Dominic Henn
- Department of Hand, Plastic and Reconstructive Surgery, BG Trauma Center Ludwigshafen, University of Heidelberg, 67071, Ludwigshafen, Germany
| | - Tanja Rädle-Hurst
- Department of Pediatric Cardiology, Saarland University Medical Center, 66421, Homburg/Saar, Germany
| | - Andreas Keller
- Chair for Clinical Bioinformatics, Saarland University, 66041, Saarbrücken, Germany
| | - Hashim Abdul-Khaliq
- Department of Pediatric Cardiology, Saarland University Medical Center, 66421, Homburg/Saar, Germany
| | - Eckart Meese
- Institute of Human Genetics, Saarland University, 66421, Homburg/Saar, Germany
| |
Collapse
|
33
|
Eid RA, Alkhateeb MA, Eleawa S, Al-Hashem FH, Al-Shraim M, El-Kott AF, Zaki MSA, Dallak MA, Aldera H. Cardioprotective effect of ghrelin against myocardial infarction-induced left ventricular injury via inhibition of SOCS3 and activation of JAK2/STAT3 signaling. Basic Res Cardiol 2018; 113:13. [PMID: 29392420 DOI: 10.1007/s00395-018-0671-4] [Citation(s) in RCA: 76] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Accepted: 01/22/2018] [Indexed: 11/27/2022]
Abstract
The molecular mechanisms through which ghrelin exerts its cardioprotective effects during cardiac remodeling post-myocardial infarction (MI) are poorly understood. The aim of this study was to investigate whether the cardioprotection mechanisms are mediated by modulation of JAK/STAT signaling and what triggers this modulation. Rats were divided into six groups (n = 12/group): control, sham, sham + ghrelin (100 µg/kg, s.c., daily, starting 1 day post-MI), MI, MI+ ghrelin, and MI+ ghrelin+ AG490, a potent JAK2 inhibitor (5 mg/kg, i.p., daily). All treatments were administered for 3 weeks. Administration of ghrelin to MI rats improved left ventricle (LV) architecture and restored cardiac contraction. In remote non-infarcted areas of MI rats, ghrelin reduced cardiac inflammation and lipid peroxidation and enhanced antioxidant enzymatic activity. In addition, independent of the growth factor/insulin growth factor-1 (GF/IGF-1) axis, ghrelin significantly increased the phosphorylation of JAK2 and Tyr702 and Ser727 residues of STAT3 and inhibited the phosphorylation of JAK1 and Tyr701 and Ser727 residues of STAT1, simultaneously increasing the expression of BCL-2 and decreasing in the expression of BAX, cleaved CASP3, and FAS. This effect coincided with decreased expression of SOCS3. All these beneficial effects of ghrelin, except its inhibitory action on IL-6 expression, were partially and significantly abolished by the co-administration of AG490. In conclusion, the cardioprotective effect of ghrelin against MI-induced LV injury is exerted via activation of JAK2/STAT3 signaling and inhibition of STAT1 signaling. These effects were independent of the GF/IGF-1 axis and could be partially mediated via inhibition of cardiac IL-6.
Collapse
MESH Headings
- Animals
- Apoptosis/drug effects
- Cardiovascular Agents/administration & dosage
- Disease Models, Animal
- Ghrelin/administration & dosage
- Heart Ventricles/drug effects
- Heart Ventricles/enzymology
- Heart Ventricles/pathology
- Heart Ventricles/physiopathology
- Interleukin-6/metabolism
- Janus Kinase 2/metabolism
- Male
- Myocardial Infarction/drug therapy
- Myocardial Infarction/enzymology
- Myocardial Infarction/pathology
- Myocardial Infarction/physiopathology
- Myocytes, Cardiac/drug effects
- Myocytes, Cardiac/enzymology
- Myocytes, Cardiac/pathology
- Oxidative Stress/drug effects
- Rats, Sprague-Dawley
- STAT1 Transcription Factor/metabolism
- STAT3 Transcription Factor/metabolism
- Signal Transduction/drug effects
- Suppressor of Cytokine Signaling 3 Protein/metabolism
- Ventricular Dysfunction, Left/enzymology
- Ventricular Dysfunction, Left/pathology
- Ventricular Dysfunction, Left/physiopathology
- Ventricular Dysfunction, Left/prevention & control
- Ventricular Function, Left/drug effects
- Ventricular Remodeling/drug effects
Collapse
Affiliation(s)
- Refaat A Eid
- Department of Pathology, College of Medicine, King Khalid University, Abha, 61421, Saudi Arabia.
| | - Mahmoud A Alkhateeb
- Department of Basic Medical Sciences, College of Medicine, King Saud bin Abdulaziz University for Health Sciences, Riyadh, 14611, Saudi Arabia
| | - Samy Eleawa
- College of Health Sciences, Applied Medical Sciences Department, PAAET, Shuwaikh, Kuwait
| | - Fahaid H Al-Hashem
- Department of Physiology, College of Medicine, King Khalid University, P.O. Box 641, Abha, 61421, Saudi Arabia
| | - Mubarak Al-Shraim
- Department of Pathology, College of Medicine, King Khalid University, Abha, 61421, Saudi Arabia
| | - Attalla Farag El-Kott
- Department of Biology, College of Science, King Khalid University, P.O. Box 641, Abha, 61421, Saudi Arabia
| | - Mohamed Samir Ahmed Zaki
- Department of Anatomy, College of Medicine, King Khalid University, P.O. Box 641, Abha, 61421, Saudi Arabia
| | - Mohammad A Dallak
- Department of Physiology, College of Medicine, King Khalid University, P.O. Box 641, Abha, 61421, Saudi Arabia
| | - Hussain Aldera
- Department of Basic Medical Sciences, College of Medicine, King Saud bin Abdulaziz University for Health Sciences, Riyadh, 14611, Saudi Arabia
| |
Collapse
|
34
|
Aboulhoda BE. Age-related remodeling of the JAK/STAT/SOCS signaling pathway and associated myocardial changes: From histological to molecular level. Ann Anat 2017; 214:21-30. [PMID: 28782583 DOI: 10.1016/j.aanat.2017.07.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2017] [Revised: 07/06/2017] [Accepted: 07/17/2017] [Indexed: 12/01/2022]
Abstract
BACKGROUND The cellular and molecular mechanisms implicated in age-associated changes in myocardial structure are of paramount importance since they cause profound alterations in the functional response and represent targets for alleviating age-related pathologies. One of these mechanisms is the JAK/STAT/SOCS signaling pathway. AIM OF THE STUDY The present study is designed to elucidate age-dependent changes of the myocardium to provide morphological basis displaying the pathogenesis of myocardial hypertrophy, fibrosis and inflammation with aging. MATERIAL AND METHODS Thirty male Sprague Dawley rats aged; 6, 30 and 36 months were used in this study. The animals were divided into three age groups, young adult, senile and very senile rats, respectively. The heart weight/body weight ratio was determined. The heart was subjected to gross morphologic examination, microscopic examination using H&E and Masson's trichrome stains and immunohistochemical examination for detection of JAK, pSTAT3, α-SMA, β-MHC and CD45. Western blotting was also carried out to detect SOCS genes. Real-time PCR was used to detect the inflammatory markers TNFα and IL1β and the hypertrophy marker α-SKA. Biochemical analysis of cardiac troponin I and creatine kinase-MB was done. Quantitative histomorphometric estimations included estimation of cardiac myocyte cross sectional area, estimation of the area percent of collagen fibers in Masson's trichrome stained sections and determination of optical density in immunostained sections. Electron microscopic examination was done to determine capillary density. RESULTS Jak and pSTAT3 were predominantly localized to the nuclei and exhibited progressive decline with aging, while SOCS3 activity displayed an age-related increase. The aged myocardium displayed profound age associated structural changes as well as myocardial hypertrophy, fibrosis and inflammation in senile and very senile rats. CONCLUSION The age-related modifications in the JAK/STAT/SOCS signaling as well as the age-associated pathological changes in myocardial structure are of particular interest as they provide further insight in age-associated heart pathologies and represent potential targets for cardioprotective and therapeutic approaches.
Collapse
Affiliation(s)
- Basma Emad Aboulhoda
- Department of Anatomy and Embryology, Faculty of Medicine, Cairo University, Egypt.
| |
Collapse
|
35
|
Chen Y, Surinkaew S, Naud P, Qi XY, Gillis MA, Shi YF, Tardif JC, Dobrev D, Nattel S. JAK-STAT signalling and the atrial fibrillation promoting fibrotic substrate. Cardiovasc Res 2017; 113:310-320. [PMID: 28158495 DOI: 10.1093/cvr/cvx004] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2016] [Accepted: 01/30/2017] [Indexed: 12/15/2022] Open
Abstract
Aims Left-atrial (LA) fibrosis is an important feature of many atrial fibrillation (AF) substrates. The JAK-STAT system contributes to cardiac remodelling, but its role in AF is unknown. Here we investigated JAK-STAT changes in an AF-model and their potential contributions to LA-fibrosis. Methods and results LA-remodelling was studied in dogs with heart failure (HF) induced by ventricular tachypacing (VTP, 240 bpm), and in mice with left-ventricular (LV) dysfunction due to myocardial infarction (MI). The selective STAT-3 inhibitor S3I-201 was administered to fibroblasts in vitro or mice in vivo (10 mg/kg/d, osmotic mini-pump). HF-dogs developed LA-selective fibrosis and AF-susceptibility at 1-week VTP. The mRNA-expression of platelet-derived growth factor (PDGF, a JAK-STAT activator) isoforms A, C and D, as well as JAK2, increased in LA fibroblasts from 1-week VTP. HF upregulated protein-expression of PDGF-receptor-β and phosphorylated (activated) signal transducer and activator of transcription 3 (STAT3) in LA. PDGF-AB stimulation of LA fibroblasts increased PDGFR-α, STAT3 and phosphorylated-STAT3 expression, as well as collagen-1 and fibronectin-1 protein secretion (by 1.6- to 20-fold), with smaller changes in LV fibroblasts. Phosphorylated-STAT3 and collagen upregulation were suppressed by the JAK2 inhibitor AG-490, PDGF receptor inhibitor AG1296 and STAT3-inhibitor SI3-201. In vivo S3I-201 treatment of MI-mice attenuated LA-fibrosis, LA-dilation and P-wave duration changes versus vehicle-control. Conclusions HF activates the LA JAK-STAT system and enhances PDGF-signalling. JAK-STAT inhibition reduces the profibrotic effects of PDGF stimulation on canine fibroblasts in vitro while attenuating in vivo LA-fibrosis and remodelling in post-MI mice, suggesting that the JAK/STAT pathway contributes to LA-fibrogenesis and might be a potential target for LA-fibrosis prevention.
Collapse
Affiliation(s)
- Yu Chen
- Department of Medicine and Research Center Montreal Heart Institute and Université de Montréal, 5000 Belanger St. E., Montreal, Quebec, H1T 1C8 Canada.,Department of Pharmacology and Therapeutics, McGill University, 3655 Promenade Sir William Osler, Montreal, Quebec, H3G 1Y6 Canada
| | - Sirirat Surinkaew
- Department of Medicine and Research Center Montreal Heart Institute and Université de Montréal, 5000 Belanger St. E., Montreal, Quebec, H1T 1C8 Canada
| | - Patrice Naud
- Department of Medicine and Research Center Montreal Heart Institute and Université de Montréal, 5000 Belanger St. E., Montreal, Quebec, H1T 1C8 Canada
| | - Xiao-Yan Qi
- Department of Medicine and Research Center Montreal Heart Institute and Université de Montréal, 5000 Belanger St. E., Montreal, Quebec, H1T 1C8 Canada
| | - Marc-Antoine Gillis
- Department of Medicine and Research Center Montreal Heart Institute and Université de Montréal, 5000 Belanger St. E., Montreal, Quebec, H1T 1C8 Canada
| | - Yan-Fen Shi
- Department of Medicine and Research Center Montreal Heart Institute and Université de Montréal, 5000 Belanger St. E., Montreal, Quebec, H1T 1C8 Canada
| | - Jean-Claude Tardif
- Department of Medicine and Research Center Montreal Heart Institute and Université de Montréal, 5000 Belanger St. E., Montreal, Quebec, H1T 1C8 Canada
| | - Dobromir Dobrev
- Faculty of Medicine, Institute of Pharmacology, University Duisburg-Essen, Hufelandstrasse 55, Essen 45122, Germany
| | - Stanley Nattel
- Department of Medicine and Research Center Montreal Heart Institute and Université de Montréal, 5000 Belanger St. E., Montreal, Quebec, H1T 1C8 Canada.,Department of Pharmacology and Therapeutics, McGill University, 3655 Promenade Sir William Osler, Montreal, Quebec, H3G 1Y6 Canada.,Faculty of Medicine, Institute of Pharmacology, University Duisburg-Essen, Hufelandstrasse 55, Essen 45122, Germany
| |
Collapse
|
36
|
Yang Y, Hu W, Di S, Ma Z, Fan C, Wang D, Jiang S, Li Y, Zhou Q, Li T, Luo E. Tackling myocardial ischemic injury: the signal transducer and activator of transcription 3 (STAT3) at a good site. Expert Opin Ther Targets 2016; 21:215-228. [PMID: 28001439 DOI: 10.1080/14728222.2017.1275566] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Yang Yang
- Department of Biomedical Engineering, The Fourth Military Medical University, Xi’an, China
- Department of Thoracic and Cardiovascular Surgery, Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Wei Hu
- Department of Biomedical Engineering, The Fourth Military Medical University, Xi’an, China
| | - Shouyin Di
- Department of Thoracic Surgery, Tangdu Hospital, The Fourth Military Medical University, Xi’an, China
| | - Zhiqiang Ma
- Department of Thoracic Surgery, Tangdu Hospital, The Fourth Military Medical University, Xi’an, China
| | - Chongxi Fan
- Department of Thoracic Surgery, Tangdu Hospital, The Fourth Military Medical University, Xi’an, China
| | - Dongjin Wang
- Department of Thoracic and Cardiovascular Surgery, Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Shuai Jiang
- Department of Aerospace Medicine, The Fourth Military Medical University, Xi’an, China
| | - Yue Li
- Department of Biomedical Engineering, The Fourth Military Medical University, Xi’an, China
| | - Qing Zhou
- Department of Thoracic and Cardiovascular Surgery, Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Tian Li
- Department of Biomedical Engineering, The Fourth Military Medical University, Xi’an, China
| | - Erping Luo
- Department of Biomedical Engineering, The Fourth Military Medical University, Xi’an, China
| |
Collapse
|
37
|
Cardiopulmonary Bypass Decreases Activation of the Signal Transducer and Activator of Transcription 3 (STAT3) Pathway in Diabetic Human Myocardium. Ann Thorac Surg 2015; 100:1636-45; discussion 1645. [PMID: 26228595 DOI: 10.1016/j.athoracsur.2015.05.013] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2015] [Revised: 05/01/2015] [Accepted: 05/05/2015] [Indexed: 01/03/2023]
Abstract
BACKGROUND Cardiopulmonary bypass (CPB) is associated with increased myocardial oxidative stress and apoptosis in diabetic patients. A mechanistic understanding of this relationship could have therapeutic value. To establish a possible mechanism, we compared the activation of the cardioprotective signal transducer and activator of transcription 3 (STAT3) pathway between patients with uncontrolled diabetes (UD) and nondiabetic (ND) patients. METHODS Right atrial tissue and serum were collected before and after CPB from 80 patients, 39 ND and 41 UD (HbA1c ≥ 6.5), undergoing cardiac operations. The samples were evaluated with Western blotting, immunohistochemistry, and microarray. RESULTS On Western blot, leptin levels were significantly increased in ND post-CPB (p < 0.05). Compared with ND, the expression of Janus kinase 2 and phosphorylation (p-) of STAT3 was significantly decreased in UD (p < 0.05). The apoptotic proteins p-Bc12/Bc12 and caspase 3 were significantly increased (p < 0.05), antiapoptotic proteins Mcl-1, Bcl-2, and p-Akt were significantly decreased (p < 0.05) in UD compared with ND. The microarray data suggested significantly increased expression of interleukin-6 R, proapoptotic p-STAT1, caspase 9, and decreased expression of Bc12 and protein inhibitor of activated STAT1 antiapoptotic genes (p = 0.05) in the UD patients. The oxidative stress marker nuclear factor-κB was significantly higher (p < 0.05) in UD patients post-CPB compared with the pre-CPB value, but was decreased, albeit insignificantly, in ND patients post-CPB. CONCLUSIONS Compared with ND, UD myocardium demonstrated attenuation of the cardioprotective STAT3 pathway. Identification of this mechanism offers a possible target for therapeutic modulation.
Collapse
|
38
|
Barry SP. JAK-STAT. JAKSTAT 2012; 1:90-1. [PMID: 24058757 PMCID: PMC3670300 DOI: 10.4161/jkst.20939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
|