1
|
Liu YJ, Sulc J, Auwerx J. Mitochondrial genetics, signalling and stress responses. Nat Cell Biol 2025; 27:393-407. [PMID: 40065146 DOI: 10.1038/s41556-025-01625-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Accepted: 01/22/2025] [Indexed: 03/15/2025]
Abstract
Mitochondria are multifaceted organelles with crucial roles in energy generation, cellular signalling and a range of synthesis pathways. The study of mitochondrial biology is complicated by its own small genome, which is matrilineally inherited and not subject to recombination, and present in multiple, possibly different, copies. Recent methodological developments have enabled the analysis of mitochondrial DNA (mtDNA) in large-scale cohorts and highlight the far-reaching impact of mitochondrial genetic variation. Genome-editing techniques have been adapted to target mtDNA, further propelling the functional analysis of mitochondrial genes. Mitochondria are finely tuned signalling hubs, a concept that has been expanded by advances in methodologies for studying the function of mitochondrial proteins and protein complexes. Mitochondrial respiratory complexes are of dual genetic origin, requiring close coordination between mitochondrial and nuclear gene-expression systems (transcription and translation) for proper assembly and function, and recent findings highlight the importance of the mitochondria in this bidirectional signalling.
Collapse
Affiliation(s)
- Yasmine J Liu
- Laboratory of Integrative Systems Physiology, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Jonathan Sulc
- Laboratory of Integrative Systems Physiology, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Johan Auwerx
- Laboratory of Integrative Systems Physiology, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland.
| |
Collapse
|
2
|
Hill AP, Iqbal SB, Case BC, Shankar AA, Merdler I. Acute Coronary Syndrome and Rheumatic Disease. J Clin Med 2025; 14:1490. [PMID: 40094966 PMCID: PMC11900616 DOI: 10.3390/jcm14051490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Revised: 02/13/2025] [Accepted: 02/19/2025] [Indexed: 03/19/2025] Open
Abstract
Patients with rheumatic disease and immune disorders have been noted to show an earlier development of atherosclerosis and to present with acute coronary syndrome. These diseases disproportionately affect women, and patients frequently have a higher number of comorbidities and other risk factors. Inflammation has long been known to play a role in the development of coronary artery disease. Early studies have shown some benefit in treatment targeting inflammation. While this has not been realized for all populations, there remains potential in treatment with targeted and individualized therapies. Especially since these diseases are associated with a worse prognosis, management benefits from the multidisciplinary expertise of cardiologists, rheumatologists, and other providers. However, the prevention and treatment of underlying rheumatic disease remains essential. This review will seek to highlight prior studies and future directions in the treatment of acute coronary syndrome in patients with rheumatologic disease.
Collapse
Affiliation(s)
- Andrew P. Hill
- Section of Cardiology, MedStar Washington Hospital Center and Georgetown University, Washington, DC 20007, USA;
| | - Shaikh B. Iqbal
- Section of Internal Medicine, MedStar Union Memorial Hospital and Georgetown University, Washington, DC 20007, USA;
| | - Brian C. Case
- Section of Interventional Cardiology, MedStar Washington Hospital Center, Washington, DC 20010, USA;
| | | | | |
Collapse
|
3
|
Potere N, Bonaventura A, Abbate A. Novel Therapeutics and Upcoming Clinical Trials Targeting Inflammation in Cardiovascular Diseases. Arterioscler Thromb Vasc Biol 2024; 44:2371-2395. [PMID: 39387118 PMCID: PMC11602387 DOI: 10.1161/atvbaha.124.319980] [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] [Indexed: 10/12/2024]
Abstract
Cardiovascular disease (CVD) remains a major health burden despite significant therapeutic advances accomplished over the last decades. It is widely and increasingly recognized that systemic inflammation not only represents a major cardiovascular risk and prognostic factor but also plays key pathogenic roles in CVD development and progression. Despite compelling preclinical evidence suggesting large potential of anti-inflammatory pharmacological interventions across numerous CVDs, clinical translation remains incomplete, mainly due to (1) yet undefined molecular signaling; (2) challenges of safety and efficacy profile of anti-inflammatory drugs; and (3) difficulties in identifying optimal patient candidates and responders to anti-inflammatory therapeutics, as well as optimal therapeutic windows. Randomized controlled trials demonstrated the safety/efficacy of canakinumab and colchicine in secondary cardiovascular prevention, providing confirmation for the involvement of a specific inflammatory pathway (NLRP3 [NACHT, LRR, and PYD domain-containing protein 3] inflammasome/IL [interleukin]-1β) in atherosclerotic CVD. Colchicine was recently approved by the US Food and Drug Administration for this indication. Diverse anti-inflammatory drugs targeting distinct inflammatory pathways are widely used for the management of other CVDs including myocarditis and pericarditis. Ongoing research efforts are directed to implementing anti-inflammatory therapeutic strategies across a growing number of CVDs, through repurposing of available anti-inflammatory drugs and development of novel anti-inflammatory compounds, which are herein concisely discussed. This review also summarizes the main characteristics and findings of completed and upcoming randomized controlled trials directly targeting inflammation in CVDs, and discusses major challenges and future perspectives in the exciting and constantly expanding landscape of cardioimmunology.
Collapse
Affiliation(s)
- Nicola Potere
- Department of Medicine and Ageing Sciences, “G. d’Annunzio” University of Chieti-Pescara, Chieti, Italy
| | - Aldo Bonaventura
- Medical Center, S.C. Medicina Generale 1, Ospedale di Circolo and Fondazione Macchi, Department of Internal Medicine, ASST Sette Laghi Varese, Italy
| | - Antonio Abbate
- Berne Cardiovascular Research Center and Division of Cardiology, University of Virginia, Charlottesville, VA, USA
| |
Collapse
|
4
|
Rodrigues KE, Pontes MHB, Cantão MBS, Prado AF. The role of matrix metalloproteinase-9 in cardiac remodeling and dysfunction and as a possible blood biomarker in heart failure. Pharmacol Res 2024; 206:107285. [PMID: 38942342 DOI: 10.1016/j.phrs.2024.107285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 06/15/2024] [Accepted: 06/23/2024] [Indexed: 06/30/2024]
Abstract
Heart failure (HF) is the leading cause of morbidity and mortality in cardiovascular diseases, being responsible for many hospitalizations annually. HF is considered a public health problem with significant economic and social impact, which makes searches essential for strategies that improve the ability to predict and diagnose HF. In this way, biomarkers can help in risk stratification for a more personalized approach to patients with HF. Preclinical and clinical evidence shows the participation of matrix metalloproteinase 9 (MMP-9) in the HF process. In this review, we will demonstrate the critical role that MMP-9 plays in cardiac remodeling and dysfunction. We will also show its importance as a blood biomarker in acute and chronic HF patients.
Collapse
Affiliation(s)
- Keuri Eleutério Rodrigues
- Biodiversity and Biotechnology Post Graduate Program - BIONORTE, Institute of Biological Sciences, Federal University of Para, Belem, Brazil; Cardiovascular System Pharmacology and Toxicology Laboratory, Institute of Biological Sciences, Federal University of Para, Belem, Brazil
| | - Maria Helena Barbosa Pontes
- Cardiovascular System Pharmacology and Toxicology Laboratory, Institute of Biological Sciences, Federal University of Para, Belem, Brazil; Pharmacology and Biochemistry Post Graduate Program - FARMABIO, Institute of Biological Sciences, Federal University of Para, Belem, Brazil
| | - Manoel Benedito Sousa Cantão
- Cardiovascular System Pharmacology and Toxicology Laboratory, Institute of Biological Sciences, Federal University of Para, Belem, Brazil; Pharmacology and Biochemistry Post Graduate Program - FARMABIO, Institute of Biological Sciences, Federal University of Para, Belem, Brazil
| | - Alejandro Ferraz Prado
- Biodiversity and Biotechnology Post Graduate Program - BIONORTE, Institute of Biological Sciences, Federal University of Para, Belem, Brazil; Cardiovascular System Pharmacology and Toxicology Laboratory, Institute of Biological Sciences, Federal University of Para, Belem, Brazil; Pharmacology and Biochemistry Post Graduate Program - FARMABIO, Institute of Biological Sciences, Federal University of Para, Belem, Brazil.
| |
Collapse
|
5
|
Wu P, Xu L, Wang Q, Ma X, Wang X, Wang H, He S, Ru H, Zhao Y, Xiao Y, Zhang J, Wang X, An S, Hacker M, Li X, Zhang X, Wang Y, Yang M, Wu Z, Li S. Left Ventricular Remodelling Associated with the Transient Elevated [ 68Ga]Ga-Pentixafor Activity in the Remote Myocardium Following Acute Myocardial Infarction. Mol Imaging Biol 2024; 26:693-703. [PMID: 38641708 DOI: 10.1007/s11307-024-01912-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 02/17/2024] [Accepted: 03/14/2024] [Indexed: 04/21/2024]
Abstract
BACKGROUND Previous studies have initially reported accompanying elevated 2-deoxy-2[18F]fluoro-D-glucose ([18F]F-FDG) inflammatory activity in the remote area and its prognostic value after acute myocardial infarction (AMI). Non-invasive characterization of the accompanying inflammation in the remote myocardium may be of potency in guiding future targeted theranostics. [68Ga]Ga-Pentixafor targeting chemokine receptor 4 (CXCR4) on the surface of inflammatory cells is currently one of the promising inflammatory imaging agents. In this study, we sought to focus on the longitudinal evolution of [68Ga]Ga-Pentixafor activities in the remote myocardium following AMI and its association with cardiac function. METHODS Twelve AMI rats and six Sham rats serially underwent [68Ga]Ga-Pentixafor imaging at pre-operation, and 5, 7, 14 days post-operation. Maximum and mean standard uptake value (SUV) and target-to-background ratio (TBR) were assessed to indicate the uptake intensity. Gated [18F]F-FDG imaging and immunofluorescent staining were performed to obtain cardiac function and responses of pro-inflammatory and reparative macrophages, respectively. RESULTS The uptake of [68Ga]Ga-Pentixafor in the infarcted myocardium peaked at day 5 (all P = 0.003), retained at day 7 (all P = 0.011), and recovered at day 14 after AMI (P > 0.05), paralleling with the rise-fall pro-inflammatory M1 macrophages (P < 0.05). Correlated with the peak activity in the infarct territory, [68Ga]Ga-Pentixafor uptake in the remote myocardium on day 5 early after AMI significantly increased (AMI vs. Sham: SUVmean, SUVmax, and TBRmean: all P < 0.05), and strongly correlated with contemporaneous EDV and/or ESV (SUVmean and TBRmean: both P < 0.05). The transitory remote activity recovered as of day 7 post-AMI (AMI vs. Sham: P > 0.05). CONCLUSIONS Corresponding with the peaked [68Ga]Ga-Pentixafor activity in the infarcted myocardium, the activity in the remote region elevated accordingly and led to contemporaneous left ventricular remodelling early after AMI. Further studies are warranted to clarify its clinical application potential.
Collapse
Affiliation(s)
- Ping Wu
- Department of Nuclear Medicine, First Hospital of Shanxi Medical University, No. 85 Jiefang South Road, Taiyuan, 030001, Shanxi, China
- Collaborative Innovation Center for Molecular Imaging of Precision Medicine, Shanxi Medical University, Taiyuan, China
| | - Li Xu
- Department of Nuclear Medicine, First Hospital of Shanxi Medical University, No. 85 Jiefang South Road, Taiyuan, 030001, Shanxi, China
- Collaborative Innovation Center for Molecular Imaging of Precision Medicine, Shanxi Medical University, Taiyuan, China
| | - Qi Wang
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Shanxi Medical University, Taiyuan, China
| | - Xiaofang Ma
- Department of Nuclear Medicine, First Hospital of Shanxi Medical University, No. 85 Jiefang South Road, Taiyuan, 030001, Shanxi, China
| | - Xinzhu Wang
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Shanxi Medical University, Taiyuan, China
| | - Hongliang Wang
- Department of Nuclear Medicine, First Hospital of Shanxi Medical University, No. 85 Jiefang South Road, Taiyuan, 030001, Shanxi, China
- Collaborative Innovation Center for Molecular Imaging of Precision Medicine, Shanxi Medical University, Taiyuan, China
| | - Sheng He
- Department of Radiology, First Hospital of Shanxi Medical University, Taiyuan, China
| | - Huibin Ru
- Department of Nuclear Medicine, First Hospital of Shanxi Medical University, No. 85 Jiefang South Road, Taiyuan, 030001, Shanxi, China
- Collaborative Innovation Center for Molecular Imaging of Precision Medicine, Shanxi Medical University, Taiyuan, China
| | - Yuting Zhao
- Department of Nuclear Medicine, First Hospital of Shanxi Medical University, No. 85 Jiefang South Road, Taiyuan, 030001, Shanxi, China
| | - Yuxin Xiao
- Department of Nuclear Medicine, First Hospital of Shanxi Medical University, No. 85 Jiefang South Road, Taiyuan, 030001, Shanxi, China
| | - Jingying Zhang
- Department of Nuclear Medicine, First Hospital of Shanxi Medical University, No. 85 Jiefang South Road, Taiyuan, 030001, Shanxi, China
| | - Xinchao Wang
- Collaborative Innovation Center for Molecular Imaging of Precision Medicine, Shanxi Medical University, Taiyuan, China
| | - Shaohui An
- Collaborative Innovation Center for Molecular Imaging of Precision Medicine, Shanxi Medical University, Taiyuan, China
- Shanghai United Imaging Healthcare Co., Ltd., Shanghai, China
| | - Marcus Hacker
- Division of Nuclear Medicine, Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Vienna, Austria
| | - Xiang Li
- Division of Nuclear Medicine, Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Vienna, Austria
| | - Xiaoli Zhang
- Laboratory for Molecular Imaging, Department of Nuclear Medicine, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Yuetao Wang
- Department of Nuclear Medicine, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
| | - Minfu Yang
- Department of Nuclear Medicine, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Zhifang Wu
- Department of Nuclear Medicine, First Hospital of Shanxi Medical University, No. 85 Jiefang South Road, Taiyuan, 030001, Shanxi, China
- Collaborative Innovation Center for Molecular Imaging of Precision Medicine, Shanxi Medical University, Taiyuan, China
| | - Sijin Li
- Department of Nuclear Medicine, First Hospital of Shanxi Medical University, No. 85 Jiefang South Road, Taiyuan, 030001, Shanxi, China.
- Collaborative Innovation Center for Molecular Imaging of Precision Medicine, Shanxi Medical University, Taiyuan, China.
| |
Collapse
|
6
|
Matta A, Ohlmann P, Nader V, Moussallem N, Carrié D, Roncalli J. A review of therapeutic approaches for post-infarction left ventricular remodeling. Curr Probl Cardiol 2024; 49:102562. [PMID: 38599556 DOI: 10.1016/j.cpcardiol.2024.102562] [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: 03/31/2024] [Accepted: 04/07/2024] [Indexed: 04/12/2024]
Abstract
Left ventricular remodeling is an adaptive process initially developed in response to acute myocardial infarction (AMI), but it ends up with negative adverse outcomes such as infarcted wall thinning, ventricular dilation, and cardiac dysfunction. A prolonged excessive inflammatory reaction to cardiomyocytes death and necrosis plays the crucial role in the pathophysiological mechanisms. The pharmacological treatment includes nitroglycerine, β-blockers, ACEi/ARBs, SGLT2i, mineralocorticoid receptor antagonists, and some miscellaneous aspects. Stem cells therapy, CD34+ cells transplantation and gene therapy constitute the promissing therapeutic approaches for post AMI cardiac remodeling, thereby enhancing angiogenesis, cardiomyocytes differenciation and left ventricular function on top of inhibiting apoptosis, inflammation, and collagen deposition. All these lead to reduce infarct size, scar formation and myocardial fibrosis.
Collapse
Affiliation(s)
- Anthony Matta
- Department of Cardiology, Civilian Hospitals of Colmar, Colmar, France; School of Medicine and Medical Sciences, Holy Spirit University of Kaslik, P.O.Box 446, Jounieh, Lebanon.
| | - Patrick Ohlmann
- Department of Cardiology, Strasbourg University Hospital, Strasbourg, France
| | - Vanessa Nader
- Department of Cardiology, Civilian Hospitals of Colmar, Colmar, France
| | - Nicolas Moussallem
- School of Medicine and Medical Sciences, Holy Spirit University of Kaslik, P.O.Box 446, Jounieh, Lebanon
| | - Didier Carrié
- Department of Cardiology, Toulouse University Hospital, Toulouse, France
| | - Jerome Roncalli
- Department of Cardiology, Toulouse University Hospital, Toulouse, France
| |
Collapse
|
7
|
Zulfaj E, Nejat A, Haamid A, Elmahdy A, Espinosa A, Redfors B, Omerovic E. Animal models of Takotsubo syndrome: bridging the gap to the human condition. Front Cardiovasc Med 2024; 11:1351587. [PMID: 38841261 PMCID: PMC11152046 DOI: 10.3389/fcvm.2024.1351587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Accepted: 03/18/2024] [Indexed: 06/07/2024] Open
Abstract
Modelling human diseases serves as a crucial tool to unveil underlying mechanisms and pathophysiology. Takotsubo syndrome (TS), an acute form of heart failure resembling myocardial infarction, manifests with reversible regional wall motion abnormalities (RWMA) of the ventricles. Despite its mortality and clinical similarity to myocardial infarction, TS aetiology remains elusive, with stress and catecholamines playing central roles. This review delves into current animal models of TS, aiming to assess their ability to replicate key clinical traits and identifying limitations. An in-depth evaluation of published animal models reveals a variation in the definition of TS among studies. We notice a substantial prevalence of catecholamine-induced models, particularly in rodents. While these models shed light on TS, there remains potential for refinement. Translational success in TS research hinges on models that align with human TS features and exhibit the key features, including transient RWMA. Animal models should be comprehensively evaluated regarding the various systemic changes of the applied trigger(s) for a proper interpretation. This review acts as a guide for researchers, advocating for stringent TS model standards and enhancing translational validity.
Collapse
Affiliation(s)
- Ermir Zulfaj
- Department of Molecular and Clinical Medicine, Institute of Medicine, Gothenburg University, Gothenburg, Sweden
| | - AmirAli Nejat
- Department of Molecular and Clinical Medicine, Institute of Medicine, Gothenburg University, Gothenburg, Sweden
| | - Abdulhussain Haamid
- Department of Cardiology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Ahmed Elmahdy
- Department of Molecular and Clinical Medicine, Institute of Medicine, Gothenburg University, Gothenburg, Sweden
| | - Aaron Espinosa
- Department of Molecular and Clinical Medicine, Institute of Medicine, Gothenburg University, Gothenburg, Sweden
| | - Björn Redfors
- Department of Molecular and Clinical Medicine, Institute of Medicine, Gothenburg University, Gothenburg, Sweden
- Core Facilities - Experimental Biomedicine, Sahlgrenska Academy, Gothenburg, Sweden
| | - Elmir Omerovic
- Department of Molecular and Clinical Medicine, Institute of Medicine, Gothenburg University, Gothenburg, Sweden
- Core Facilities - Experimental Biomedicine, Sahlgrenska Academy, Gothenburg, Sweden
| |
Collapse
|
8
|
Sordi R, Bojko L, Oliveira FRMB, Rosales TO, Souza CF, Moreno LW, Ferreira Alves G, Vellosa JCR, Fernandes D, Gomes JR. Doxycycline reduces liver and kidney injuries in a rat hemorrhagic shock model. Intensive Care Med Exp 2024; 12:2. [PMID: 38194181 PMCID: PMC10776514 DOI: 10.1186/s40635-023-00586-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Accepted: 12/18/2023] [Indexed: 01/10/2024] Open
Abstract
BACKGROUND Hemorrhagic shock (HS), which causes insufficient tissue perfusion, can result in multiple organ failure (MOF) and death. This study aimed to evaluate whether doxycycline (DOX) protects cardiovascular, kidney, and liver tissue from damage in a rat model of HS. Immediately before the resuscitation, DOX (10 mg/kg; i.v.) was administered, and its protective effects were assessed 24 h later. Mean arterial pressure, renal blood flow, heart rate, vasoactive drug response, and blood markers such as urea, creatinine, AST, ALT, CPK, CPR, and NOx levels were determined. RESULTS We showed that DOX has a significant effect on renal blood flow and on urea, creatinine, AST, ALT, CPK, and NOx. Morphologically, DOX reduced the inflammatory process in the liver tissue. CONCLUSIONS We conclude that DOX protects the liver and kidney against injury and dysfunction in a HS model and could be a strategy to reduce organ damage associated with ischemia-and-reperfusion injury.
Collapse
Affiliation(s)
- Regina Sordi
- Department of Pharmacology, Graduate Program in Pharmacology, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil
- Department of Structural Biology, Molecular and Genetics, Post Graduation Program in Biomedical Science, Universidade Estadual de Ponta Grossa, Avenida Carlos Cavalcanti, 4748, Ponta Grossa, PR, 84030-900, Brazil
| | - Luana Bojko
- Department of Structural Biology, Molecular and Genetics, Universidade Estadual de Ponta Grossa, Ponta Grossa, PR, Brazil
| | - Filipe R M B Oliveira
- Department of Pharmacology, Graduate Program in Pharmacology, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil
| | - Thiele Osvaldt Rosales
- Department of Pharmacology, Graduate Program in Pharmacology, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil
| | - Camila Fernandes Souza
- Department of Pharmacology, Graduate Program in Pharmacology, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil
| | - Lucas Wenceslau Moreno
- Department of Structural Biology, Molecular and Genetics, Universidade Estadual de Ponta Grossa, Ponta Grossa, PR, Brazil
| | - Gustavo Ferreira Alves
- Department of Pharmacology, Graduate Program in Pharmacology, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil
| | - José Carlos Rebuglio Vellosa
- Department of Clinical and Toxicological Analysis, Universidade Estadual de Ponta Grossa, Ponta Grossa, PR, Brazil
| | - Daniel Fernandes
- Department of Pharmacology, Graduate Program in Pharmacology, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil
| | - Jose Rosa Gomes
- Department of Structural Biology, Molecular and Genetics, Post Graduation Program in Biomedical Science, Universidade Estadual de Ponta Grossa, Avenida Carlos Cavalcanti, 4748, Ponta Grossa, PR, 84030-900, Brazil.
| |
Collapse
|
9
|
Hartley B, Bassiouni W, Schulz R, Julien O. The roles of intracellular proteolysis in cardiac ischemia-reperfusion injury. Basic Res Cardiol 2023; 118:38. [PMID: 37768438 DOI: 10.1007/s00395-023-01007-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 08/29/2023] [Accepted: 08/30/2023] [Indexed: 09/29/2023]
Abstract
Ischemic heart disease remains a leading cause of human mortality worldwide. One form of ischemic heart disease is ischemia-reperfusion injury caused by the reintroduction of blood supply to ischemic cardiac muscle. The short and long-term damage that occurs due to ischemia-reperfusion injury is partly due to the proteolysis of diverse protein substrates inside and outside of cardiomyocytes. Ischemia-reperfusion activates several diverse intracellular proteases, including, but not limited to, matrix metalloproteinases, calpains, cathepsins, and caspases. This review will focus on the biological roles, intracellular localization, proteolytic targets, and inhibitors of these proteases in cardiomyocytes following ischemia-reperfusion injury. Recognition of the intracellular function of each of these proteases includes defining their activation, proteolytic targets, and their inhibitors during myocardial ischemia-reperfusion injury. This review is a step toward a better understanding of protease activation and involvement in ischemic heart disease and developing new therapeutic strategies for its treatment.
Collapse
Affiliation(s)
- Bridgette Hartley
- Department of Biochemistry, University of Alberta, Edmonton, AB, Canada
| | - Wesam Bassiouni
- Department of Pharmacology, University of Alberta, Edmonton, AB, Canada
| | - Richard Schulz
- Department of Pharmacology, University of Alberta, Edmonton, AB, Canada.
- Department of Pediatrics, University of Alberta, Edmonton, AB, Canada.
- Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, AB, Canada.
- Women and Children's Health Research Institute, University of Alberta, Edmonton, AB, Canada.
| | - Olivier Julien
- Department of Biochemistry, University of Alberta, Edmonton, AB, Canada.
| |
Collapse
|
10
|
Bräuninger H, Krüger S, Bacmeister L, Nyström A, Eyerich K, Westermann D, Lindner D. Matrix metalloproteinases in coronary artery disease and myocardial infarction. Basic Res Cardiol 2023; 118:18. [PMID: 37160529 PMCID: PMC10169894 DOI: 10.1007/s00395-023-00987-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 04/20/2023] [Accepted: 04/21/2023] [Indexed: 05/11/2023]
Abstract
Cardiovascular diseases (CVDs) remain the leading cause of death worldwide. Most cardiovascular deaths are caused by ischaemic heart diseases such as myocardial infarction (MI). Hereby atherosclerosis in the coronary arteries often precedes disease manifestation. Since tissue remodelling plays an important role in the development and progression of atherosclerosis as well as in outcome after MI, regulation of matrix metalloproteinases (MMPs) as the major ECM-degrading enzymes with diverse other functions is crucial. Here, we provide an overview of the expression profiles of MMPs in coronary artery and left ventricular tissue using publicly available data from whole tissue to single-cell resolution. To approach an association between MMP expression and the development and outcome of CVDs, we further review studies investigating polymorphisms in MMP genes since polymorphisms are known to have an impact on gene expression. This review therefore aims to shed light on the role of MMPs in atherosclerosis and MI by summarizing current knowledge from publically available datasets, human studies, and analyses of polymorphisms up to preclinical and clinical trials of pharmacological MMP inhibition.
Collapse
Affiliation(s)
- Hanna Bräuninger
- Department of Cardiology and Angiology, University Heart Center Freiburg-Bad Krozingen, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Hugstetter Str. 55, 79106, Freiburg, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Side Hamburg/Kiel/Lübeck, Hamburg, Germany
| | - Saskia Krüger
- Clinic for Cardiology, University Heart and Vascular Centre Hamburg, Hamburg, Germany
| | - Lucas Bacmeister
- Department of Cardiology and Angiology, University Heart Center Freiburg-Bad Krozingen, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Hugstetter Str. 55, 79106, Freiburg, Germany
| | - Alexander Nyström
- Department of Dermatology, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Kilian Eyerich
- Department of Dermatology, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Dirk Westermann
- Department of Cardiology and Angiology, University Heart Center Freiburg-Bad Krozingen, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Hugstetter Str. 55, 79106, Freiburg, Germany
| | - Diana Lindner
- Department of Cardiology and Angiology, University Heart Center Freiburg-Bad Krozingen, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Hugstetter Str. 55, 79106, Freiburg, Germany.
- German Centre for Cardiovascular Research (DZHK), Partner Side Hamburg/Kiel/Lübeck, Hamburg, Germany.
| |
Collapse
|
11
|
Xu H, Yang F, Bao Z. Gut microbiota and myocardial fibrosis. Eur J Pharmacol 2023; 940:175355. [PMID: 36309048 DOI: 10.1016/j.ejphar.2022.175355] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 10/19/2022] [Accepted: 10/21/2022] [Indexed: 01/18/2023]
Abstract
Myocardial fibrosis (MF) is a pathophysiological condition that accompanies various myocardial diseases and comprises a damaged myocardial matrix repair process. Although fibrosis plays a vital role in repair, it ultimately alters cardiac systolic and diastolic functions. The gut microbiota is a complex and dynamic ecosystem with billions of microorganisms that produce bioactive compounds that influence host health and disease progression. Intestinal microbiota has been shown to correlate with cardiovascular disease, and dysbiosis of the intestinal microbiota is involved in the development of MF. In this review, we discuss the role of intestinal microbiota in the process of MF, including alterations in microbiota composition and the effects of metabolites. We also discuss how diet and medicines can affect cardiac fibrosis by influencing the gut microbiota, and potential future therapies targeting the gut-heart axis. A healthy gut microbiota can prevent disease, but dysbiosis can lead to various symptoms, including the induction of heart disease. In this review, we discuss the relevance of the gut-heart axis and the multiple pathways by which gut microbiota may affect cardiac fibrosis, including inflammatory factors, immune cells, and gut microbiota metabolites, such as trimethylamine-N-oxide (TMAO) and short-chain fatty acids (SCFAs). Finally, we discuss the involvement of gut microbiota in the treatment of cardiac fibrosis, including drugs, fecal microbiota transplantation, and oral probiotics or prebiotics. With future studies on the relationship between the heart and gut microbiota, we hope to find better ways to improve MF through the gut-heart axis.
Collapse
Affiliation(s)
- Han Xu
- Department of Gerontology, Huadong Hospital Affiliated to Fudan University, Shanghai, China; Shanghai Key Laboratory of Clinical Geriatric Medicine, Shanghai, China; Research Center on Aging and Medicine, Fudan University, Shanghai, China
| | - Fan Yang
- Department of Gerontology, Huadong Hospital Affiliated to Fudan University, Shanghai, China; Shanghai Key Laboratory of Clinical Geriatric Medicine, Shanghai, China; Research Center on Aging and Medicine, Fudan University, Shanghai, China.
| | - Zhijun Bao
- Department of Gerontology, Huadong Hospital Affiliated to Fudan University, Shanghai, China; Shanghai Key Laboratory of Clinical Geriatric Medicine, Shanghai, China; Research Center on Aging and Medicine, Fudan University, Shanghai, China.
| |
Collapse
|
12
|
Welcome MO, Dogo D, Nikos E Mastorakis. Cellular mechanisms and molecular pathways linking bitter taste receptor signalling to cardiac inflammation, oxidative stress, arrhythmia and contractile dysfunction in heart diseases. Inflammopharmacology 2023; 31:89-117. [PMID: 36471190 PMCID: PMC9734786 DOI: 10.1007/s10787-022-01086-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Accepted: 10/11/2022] [Indexed: 12/12/2022]
Abstract
Heart diseases and related complications constitute a leading cause of death and socioeconomic threat worldwide. Despite intense efforts and research on the pathogenetic mechanisms of these diseases, the underlying cellular and molecular mechanisms are yet to be completely understood. Several lines of evidence indicate a critical role of inflammatory and oxidative stress responses in the development and progression of heart diseases. Nevertheless, the molecular machinery that drives cardiac inflammation and oxidative stress is not completely known. Recent data suggest an important role of cardiac bitter taste receptors (TAS2Rs) in the pathogenetic mechanism of heart diseases. Independent groups of researchers have demonstrated a central role of TAS2Rs in mediating inflammatory, oxidative stress responses, autophagy, impulse generation/propagation and contractile activities in the heart, suggesting that dysfunctional TAS2R signalling may predispose to cardiac inflammatory and oxidative stress disorders, characterised by contractile dysfunction and arrhythmia. Moreover, cardiac TAS2Rs act as gateway surveillance units that monitor and detect toxigenic or pathogenic molecules, including microbial components, and initiate responses that ultimately culminate in protection of the host against the aggression. Unfortunately, however, the molecular mechanisms that link TAS2R sensing of the cardiac milieu to inflammatory and oxidative stress responses are not clearly known. Therefore, we sought to review the possible role of TAS2R signalling in the pathophysiology of cardiac inflammation, oxidative stress, arrhythmia and contractile dysfunction in heart diseases. Potential therapeutic significance of targeting TAS2R or its downstream signalling molecules in cardiac inflammation, oxidative stress, arrhythmia and contractile dysfunction is also discussed.
Collapse
Affiliation(s)
- Menizibeya O Welcome
- Department of Physiology, Faculty of Basic Medical Sciences, College of Health Sciences, Nile University of Nigeria, Plot 681 Cadastral Zone, C-00 Research and Institution Area, Jabi Airport Road Bypass, FCT, Abuja, Nigeria.
| | - Dilli Dogo
- Department of Surgery, Faculty of Clinical Sciences, College of Health Sciences, Nile University of Nigeria, Abuja, Nigeria
| | - Nikos E Mastorakis
- Technical University of Sofia, Klement Ohridksi 8, Sofia, 1000, Bulgaria
| |
Collapse
|
13
|
The Carthamus tinctorius L. and Lepidium apetalum Willd. Drug Pair Inhibits EndMT through the TGF β1/Snail Signaling Pathway in the Treatment of Myocardial Fibrosis. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2023; 2023:6018375. [PMID: 36686974 PMCID: PMC9851799 DOI: 10.1155/2023/6018375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 12/22/2022] [Accepted: 12/28/2022] [Indexed: 01/13/2023]
Abstract
Background Myocardial fibrosis (MF) is an essential pathological factor for heart failure. Previous studies have shown that the combination of Carthamus tinctorius L. and Lepidium apetalum Willd. (C-L), two types of Chinese herbal medicine, can ameliorate MF after myocardial infarction (MI) in rats and inhibit the activation of myocardial fibroblasts. However, the mechanism of C-L in the treatment of MF remains unclear. Methods A rat model of MF with left anterior descending coronary ligation-induced MI was first established. Then, the effects of C-L on cardiac function, MF, and endothelial-to-mesenchymal transition (EndMT) were evaluated by the left ventricular ejection fraction (LVEF), serum N-terminal pro-brain natriuretic peptide (NT-proBNP) levels, Masson's trichrome staining, and immunohistochemical and immunofluorescence staining. Next, a hypoxia-induced cardiac microvascular endothelial cell (CMEC) model was established to observe the effects of C-L on EndMT. The supernatant of CMECs was collected and used to culture cardiac fibroblasts (CFs) and observe the effects of CMEC paracrine factors on CFs. Results Animal experiments indicated that C-L improves the cardiac function of rats after MI, inhibits the progression of EndMT and MF, and downregulates TGFβ1, Snail, and CTGF expression. Cell experiments showed that drug-loaded serum containing C-L inhibits the EndMT of CMECs under hypoxic conditions. The culture supernatant of CMECs grown under hypoxic conditions significantly activated CFs. After treatment with C-L, the activating factor for CFs in hypoxic CMEC culture supernatant was substantially downregulated, and the effect of the culture supernatant on CF activation was also reduced. However, TGFβ1 agonists inhibited the effects of C-L on CMECs and CFs. Conclusion Our data demonstrated that by regulating the TGFβ1/Snail pathway, C-L inhibits EndMT of CMECs and reduces the release of CF-activating factors in cells undergoing EndMT.
Collapse
|
14
|
Delbaere Q, Chapet N, Huet F, Delmas C, Mewton N, Prunier F, Angoulvant D, Roubille F. Anti-Inflammatory Drug Candidates for Prevention and Treatment of Cardiovascular Diseases. Pharmaceuticals (Basel) 2023; 16:78. [PMID: 36678575 PMCID: PMC9865197 DOI: 10.3390/ph16010078] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Revised: 12/26/2022] [Accepted: 12/28/2022] [Indexed: 01/07/2023] Open
Abstract
Incidence and mortality rates for cardiovascular disease are declining, but it still remains a major cause of morbidity and mortality. Drug treatments to slow the progression of atherosclerosis focus on reducing cholesterol levels. The paradigm shift to consider atherosclerosis an inflammatory disease by itself has led to the development of new treatments. In this article, we discuss the pathophysiology of inflammation and focus attention on therapeutics targeting different inflammatory pathways of atherosclerosis and myocardial infarction. In atherosclerosis, colchicine is included in new recommendations, and eight randomized clinical trials are testing new drugs in different inflammatory pathways. After a myocardial infarction, no drug has shown a significant benefit, but we present four randomized clinical trials with new treatments targeting inflammation.
Collapse
Affiliation(s)
- Quentin Delbaere
- Department of Cardiology, Arnaud de Villeneuve University Hospital, 34295 Montpellier, France
| | - Nicolas Chapet
- Department of Cardiology, Arnaud de Villeneuve University Hospital, 34295 Montpellier, France
| | - Fabien Huet
- Department of Cardiology, Arnaud de Villeneuve University Hospital, 34295 Montpellier, France
- Department of Cardiology, Bretagne Atlantique General Hospital, 56000 Vannes, France
| | - Clément Delmas
- Department of Cardiology, Arnaud de Villeneuve University Hospital, 34295 Montpellier, France
| | - Nathan Mewton
- Hôpital Cardiovasculaire Louis Pradel, 69002 Lyon, France
| | - Fabrice Prunier
- Department of Cardiology, CHU Angers, Université d’Angers, 49100 Angers, France
| | - Denis Angoulvant
- Cardiology Department, CHRU de Tours, 37044 Tours, France
- EA 4245 T2I, Université de Tours, 37044 Tours, France
| | - François Roubille
- Department of Cardiology, Arnaud de Villeneuve University Hospital, 34295 Montpellier, France
| |
Collapse
|
15
|
Calabretta R, Castello A, Giglioli C, Cecchi E, Cerisano G, Hacker M, Sciagrà R. Prognostic value of divergent pattern detection by 99mTc-sestamibi gated SPECT in patients with anterior acute myocardial infarction. J Nucl Cardiol 2022; 29:3115-3122. [PMID: 34914082 DOI: 10.1007/s12350-021-02874-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2021] [Accepted: 10/27/2021] [Indexed: 01/18/2023]
Abstract
PURPOSE In gated myocardial perfusion SPECT, apical remodeling may be identified by the presence of a divergent pattern (DP) of the left ventricle (LV). METHODS AND RESULTS We examined 150 anterior ST-elevation myocardial infarction (STEMI) patients, all successfully treated with primary percutaneous coronary interventions (PCI). Perfusion gated-SPECT to measure infarct size, LV end-diastolic (ED) and end-systolic (ES) volumes and ejection fraction (EF) was acquired before hospital discharge and repeated at 6-month follow-up. DP was observed in 26 patients, who had larger infarct size (28 ± 19% vs. 15.7 ± 17%, P < 0.02), and lower EF (33 ± 7% vs. 41 ± 10%, P < 0.001) than patients without DP. At follow-up, DP patients had significantly larger EDV (156 ± 54 vs. 107 ± 44 mL, P < 0.0001), ESV (104 ± 47 vs. 59 ± 36 mL, P < 0.0001) and lower EF (35 ± 12% vs. 48 ± 13%, P < 0.0001). 54% of DP patients developed remodeling at follow-up vs. 12% of those without DP (P < 0.001). During follow up, 7 events in the DP group (27%) and 11 events in patients without DP (9%; P < 0.02) occurred. Kaplan-Meier survival curves showed a worse prognosis for DP patients. CONCLUSION In patients with anterior AMI, early DP detection is related to subsequent LV dysfunction, larger infarct size, and worse severity. It is helpful for predicting LV remodeling at short-term follow-up and has prognostic implications.
Collapse
Affiliation(s)
- R Calabretta
- Nuclear Medicine Unit, Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Florence, Italy.
- Division of Nuclear Medicine, Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Währinger Gürtel 18-20, 1090, Vienna, Austria.
| | - A Castello
- Nuclear Medicine Unit, Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Florence, Italy
- Division of Nuclear Medicine, Fondazione IRCCS Ca´ Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - C Giglioli
- Intensive Cardiac Coronary Unit, Heart and Vessel Department, Azienda Ospedaliero-Universitaria Careggi, Florence, Italy
| | - E Cecchi
- Intensive Cardiac Coronary Unit, Heart and Vessel Department, Azienda Ospedaliero-Universitaria Careggi, Florence, Italy
| | - G Cerisano
- Division of Cardiology, Azienda Ospedaliero-Universitaria Careggi, Florence, Italy
| | - M Hacker
- Division of Nuclear Medicine, Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Währinger Gürtel 18-20, 1090, Vienna, Austria
| | - R Sciagrà
- Nuclear Medicine Unit, Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Florence, Italy
| |
Collapse
|
16
|
Cianci R, Franza L, Borriello R, Pagliari D, Gasbarrini A, Gambassi G. The Role of Gut Microbiota in Heart Failure: When Friends Become Enemies. Biomedicines 2022; 10:2712. [PMID: 36359233 PMCID: PMC9687270 DOI: 10.3390/biomedicines10112712] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2022] [Revised: 10/20/2022] [Accepted: 10/22/2022] [Indexed: 10/29/2023] Open
Abstract
Heart failure is a complex health issue, with important consequences on the overall wellbeing of patients. It can occur both in acute and chronic forms and, in the latter, the immune system appears to play an important role in the pathogenesis of the disease. In particular, in the forms with preserved ejection fraction or with only mildly reduced ejection fraction, some specific associations with chronic inflammatory diseases have been observed. Another interesting aspect that is worth considering is the role of microbiota modulation, in this context: given the importance of microbiota in the modulation of immune responses, it is possible that changes in its composition may somewhat influence the progression and even the pathogenesis of heart failure. In this narrative review, we aim to examine the relationship between immunity and heart failure, with a special focus on the role of microbiota in this pathological condition.
Collapse
Affiliation(s)
- Rossella Cianci
- Department of Translational Medicine and Surgery, Catholic University of Rome, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy
| | - Laura Franza
- Emergency Medicine Unit, Catholic University of Rome, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy
| | - Raffaele Borriello
- Department of Translational Medicine and Surgery, Catholic University of Rome, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy
| | - Danilo Pagliari
- Medical Officer of the Carabinieri Corps, Health Service of the Carabinieri General Headquarters, 00197 Rome, Italy
| | - Antonio Gasbarrini
- Department of Translational Medicine and Surgery, Catholic University of Rome, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy
| | - Giovanni Gambassi
- Department of Translational Medicine and Surgery, Catholic University of Rome, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy
| |
Collapse
|
17
|
Healing the Broken Hearts: A Glimpse on Next Generation Therapeutics. HEARTS 2022. [DOI: 10.3390/hearts3040013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Cardiovascular diseases are the leading cause of death worldwide, accounting for 32% of deaths globally and thus representing almost 18 million people according to WHO. Myocardial infarction, the most prevalent adult cardiovascular pathology, affects over half a million people in the USA according to the last records of the AHA. However, not only adult cardiovascular diseases are the most frequent diseases in adulthood, but congenital heart diseases also affect 0.8–1.2% of all births, accounting for mild developmental defects such as atrial septal defects to life-threatening pathologies such as tetralogy of Fallot or permanent common trunk that, if not surgically corrected in early postnatal days, they are incompatible with life. Therefore, both congenital and adult cardiovascular diseases represent an enormous social and economic burden that invariably demands continuous efforts to understand the causes of such cardiovascular defects and develop innovative strategies to correct and/or palliate them. In the next paragraphs, we aim to briefly account for our current understanding of the cellular bases of both congenital and adult cardiovascular diseases, providing a perspective of the plausible lines of action that might eventually result in increasing our understanding of cardiovascular diseases. This analysis will come out with the building blocks for designing novel and innovative therapeutic approaches to healing the broken hearts.
Collapse
|
18
|
Ong GJ, Nguyen TH, Surikow SY, Horowitz JD. Risk factors for a broken heart: understanding drug-induced causes for Takotsubo syndrome and pharmacological treatment options. Expert Rev Clin Pharmacol 2022; 15:1017-1025. [DOI: 10.1080/17512433.2022.2121701] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Affiliation(s)
- Gao Jing Ong
- Cardiology Unit, Central Adelaide Local Health Network, Adelaide, Australia
- Cardiovascular Pathophysiology and Therapeutics Group, Basil Hetzel Institute, University of Adelaide, Woodville, Australia
| | - Thanh Ha Nguyen
- Cardiovascular Pathophysiology and Therapeutics Group, Basil Hetzel Institute, University of Adelaide, Woodville, Australia
- Northern Adelaide Local Health Network, Elizabeth Vale, Australia
| | - Sven Y Surikow
- Cardiovascular Pathophysiology and Therapeutics Group, Basil Hetzel Institute, University of Adelaide, Woodville, Australia
- Northern Adelaide Local Health Network, Elizabeth Vale, Australia
| | - John D Horowitz
- Cardiovascular Pathophysiology and Therapeutics Group, Basil Hetzel Institute, University of Adelaide, Woodville, Australia
| |
Collapse
|
19
|
Doxycycline Attenuates Doxorubicin-Induced Cardiotoxicity by Improving Myocardial Energy Metabolism in Rats. J Cardiovasc Dev Dis 2022; 9:jcdd9080254. [PMID: 36005418 PMCID: PMC9410319 DOI: 10.3390/jcdd9080254] [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: 05/27/2022] [Revised: 08/01/2022] [Accepted: 08/02/2022] [Indexed: 12/06/2022] Open
Abstract
Aim: Evaluate the influence of doxycycline, an anti-inflammatory and matrix metalloproteinase (MMP) inhibitor, on the attenuation of chronic doxorubicin-induced cardiotoxicity in rats. Methods: We allocated male Wistar rats into four groups: control (C), doxorubicin (D), doxycycline (inhibitor of MMP, IM), and Dox + doxycycline (DIM). Groups IM and DIM received doxycycline (5 mg/kg, IP) once a week for 4 weeks. In addition, 48 h after every doxycycline injection, groups D and DIM received Dox (5 mg/kg, IP). We performed echocardiogram and evaluated TIMP-4 and collagen I protein expression, MMP-2 activity, and oxidative stress and myocardial metabolism. Results: Doxorubicin promotes left atrium (LA) and left ventricle (LV) dilatation and decreases in LV fractional shortening, which was improved by doxycycline. Moreover, doxycycline attenuated the LV cardiomyocyte hypertrophy and collagen type I expression. Doxorubicin increased phosphofructokinase and decreased beta-hydroxyacyl Co-A dehydrogenase, pyruvate dehydrogenase, citrate synthase, and ATP synthase activity, which was partially attenuated by doxycycline. Lastly, doxycycline improved antioxidant enzyme activity in the DIM group. Conclusion: Doxorubicin increases oxidative stress and promotes changes in myocardial energy metabolism, accompanied by structural and functional changes. Doxycycline attenuated the doxorubicin-induced cardiotoxicity, at least in part, through changes in myocardial energy metabolism.
Collapse
|
20
|
Gonçalves PR, Nascimento LD, Gerlach RF, Rodrigues KE, Prado AF. Matrix Metalloproteinase 2 as a Pharmacological Target in Heart Failure. Pharmaceuticals (Basel) 2022; 15:ph15080920. [PMID: 35893744 PMCID: PMC9331741 DOI: 10.3390/ph15080920] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 07/11/2022] [Accepted: 07/11/2022] [Indexed: 12/18/2022] Open
Abstract
Heart failure (HF) is an acute or chronic clinical syndrome that results in a decrease in cardiac output and an increase in intracardiac pressure at rest or upon exertion. The pathophysiology of HF is heterogeneous and results from an initial harmful event in the heart that promotes neurohormonal changes such as autonomic dysfunction and activation of the renin-angiotensin-aldosterone system, endothelial dysfunction, and inflammation. Cardiac remodeling occurs, which is associated with degradation and disorganized synthesis of extracellular matrix (ECM) components that are controlled by ECM metalloproteinases (MMPs). MMP-2 is part of this group of proteases, which are classified as gelatinases and are constituents of the heart. MMP-2 is considered a biomarker of patients with HF with reduced ejection fraction (HFrEF) or preserved ejection fraction (HFpEF). The role of MMP-2 in the development of cardiac injury and dysfunction has clearly been demonstrated in animal models of cardiac ischemia, transgenic models that overexpress MMP-2, and knockout models for this protease. New research to minimize cardiac structural and functional alterations using non-selective and selective inhibitors for MMP-2 demonstrates that this protease could be used as a possible pharmacological target in the treatment of HF.
Collapse
Affiliation(s)
- Pricila Rodrigues Gonçalves
- Cardiovascular System Pharmacology and Toxicology Laboratory, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, PA, Brazil; (P.R.G.); (L.D.N.); (K.E.R.)
| | - Lisandra Duarte Nascimento
- Cardiovascular System Pharmacology and Toxicology Laboratory, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, PA, Brazil; (P.R.G.); (L.D.N.); (K.E.R.)
| | - Raquel Fernanda Gerlach
- Department of Basic and Oral Biology, Faculty of Dentistry of Ribeirao Preto, University of Sao Paulo (FORP/USP), Ribeirao Preto 14040-904, SP, Brazil;
| | - Keuri Eleutério Rodrigues
- Cardiovascular System Pharmacology and Toxicology Laboratory, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, PA, Brazil; (P.R.G.); (L.D.N.); (K.E.R.)
| | - Alejandro Ferraz Prado
- Cardiovascular System Pharmacology and Toxicology Laboratory, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, PA, Brazil; (P.R.G.); (L.D.N.); (K.E.R.)
- Correspondence:
| |
Collapse
|
21
|
Sia CH, Tan SH, Chan SP, Marchesseau S, Sim HW, Carvalho L, Chen R, Amin NHM, Fong AYY, Richards AM, Yip C, Chan MY. Enhanced Thrombin Generation Is Associated with Worse Left Ventricular Scarring after ST-Segment Elevation Myocardial Infarction: A Cohort Study. Pharmaceuticals (Basel) 2022; 15:718. [PMID: 35745638 PMCID: PMC9231218 DOI: 10.3390/ph15060718] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 05/29/2022] [Accepted: 05/31/2022] [Indexed: 12/29/2022] Open
Abstract
Acute myocardial infarction (AMI) is associated with heightened thrombin generation. There are limited data relating to thrombin generation and left ventricular (LV) scarring and LV dilatation in post-MI LV remodeling. We studied 113 patients with ST-segment elevation myocardial infarction (STEMI) who had undergone primary percutaneous coronary intervention (PPCI) (n = 76) or pharmaco-invasive management (thrombolysis followed by early PCI, n = 37). Endogenous thrombin potential (ETP) was measured at baseline, 1 month and 6 months. Cardiovascular magnetic resonance imaging was performed at baseline and 6 months post-MI. Outcomes studied were an increase in scar change, which was defined as an increase in left ventricular infarct size of any magnitude detected by late gadolinium enhancement, adverse LV remodeling, defined as dilatation (increase) of left ventricular end-diastolic volume (LVEDV) by more than 20% and an increase in left ventricular ejection fraction (LVEF). The mean age was 55.19 ± 8.25 years and 91.2% were men. The baseline ETP was similar in the PPCI and pharmaco-invasive groups (1400.3 nM.min vs. 1334.1 nM.min, p = 0.473). Each 10-unit increase in baseline ETP was associated with a larger scar size (adjusted OR 1.020, 95% CI 1.002-1.037, p = 0.027). Baseline ETP was not associated with adverse LV remodeling or an increase in LVEF. There was no difference in scar size or adverse LV remodeling among patients undergoing PPCI vs. pharmaco-invasive management or patients receiving ticagrelor vs. clopidogrel. Enhanced thrombin generation after STEMI is associated with a subsequent increase in myocardial scarring but not LV dilatation or an increase in LVEF at 6 months post-MI.
Collapse
Affiliation(s)
- Ching-Hui Sia
- Department of Cardiology, National University Heart Centre Singapore, 1E Kent Ridge Road, NUHS Tower Block Level 9, Singapore 119228, Singapore; (C.-H.S.); (H.-W.S.); (L.C.)
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, 1E Kent Ridge Road, NUHS Tower Block Level 10, Singapore 119228, Singapore;
| | - Sock-Hwee Tan
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, 1E Kent Ridge Road, NUHS Tower Block Level 10, Singapore 119228, Singapore;
| | - Siew-Pang Chan
- National University Heart Centre Singapore, 1E Kent Ridge Road, NUHS Tower Block Level 9, Singapore 119228, Singapore;
- Yong Loo Lin School of Medicine, National University of Singapore, 10 Medical Drive, Singapore 117597, Singapore
| | | | - Hui-Wen Sim
- Department of Cardiology, National University Heart Centre Singapore, 1E Kent Ridge Road, NUHS Tower Block Level 9, Singapore 119228, Singapore; (C.-H.S.); (H.-W.S.); (L.C.)
| | - Leonardo Carvalho
- Department of Cardiology, National University Heart Centre Singapore, 1E Kent Ridge Road, NUHS Tower Block Level 9, Singapore 119228, Singapore; (C.-H.S.); (H.-W.S.); (L.C.)
- Laboratory of Genetics and Molecular Cardiology, Heart Institute (InCor-HCFMUSP), Sao Paulo 05403-904, Brazil
- Cardiac Department, Ferderal University of Sao Paulo State (UNIFESP), Sao Paulo 05303-000, Brazil
| | - Ruth Chen
- Department of Cardiology, Woodlands Health Campus, Tower E, Level 5, Yishun Community Hospital, 2 Yishun Central 2, Singapore 768024, Singapore;
| | - Nor Hanim Mohd Amin
- Clinical Research Centre, Sarawak General Hospital, Jalan Hospital, Kuching 93586, Malaysia; (N.H.M.A.); (A.Y.-Y.F.)
- Department of Cardiology, Sarawak Heart Centre, Samarahan Expressway, Kota Samarahan 94300, Malaysia
| | - Alan Yean-Yip Fong
- Clinical Research Centre, Sarawak General Hospital, Jalan Hospital, Kuching 93586, Malaysia; (N.H.M.A.); (A.Y.-Y.F.)
- Department of Cardiology, Sarawak Heart Centre, Samarahan Expressway, Kota Samarahan 94300, Malaysia
| | - Arthur Mark Richards
- Cardiovascular Research Institute, National University of Singapore, Singapore 119228, Singapore;
- Christchurch Heart Institute, Department of Medicine, University of Otago, P.O. Box 4345, Christchurch 8140, New Zealand
| | - Christina Yip
- Department of Laboratory Medicine, Main Building, Level 3, National University Hospital, 5 Lower Kent Ridge Road, Singapore 119074, Singapore;
| | - Mark Y. Chan
- Department of Cardiology, National University Heart Centre Singapore, 1E Kent Ridge Road, NUHS Tower Block Level 9, Singapore 119228, Singapore; (C.-H.S.); (H.-W.S.); (L.C.)
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, 1E Kent Ridge Road, NUHS Tower Block Level 10, Singapore 119228, Singapore;
| |
Collapse
|
22
|
Nasiri A, Shafiee A, Hosseinsabet A, Talasaz AH, Jalali A, Salarifar M. Effect of minocycline on the left ventricular function following ST-elevation myocardial infarction treated by primary percutaneous coronary intervention. Trials 2022; 23:112. [PMID: 35120566 PMCID: PMC8815113 DOI: 10.1186/s13063-021-05921-2] [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: 04/26/2021] [Accepted: 12/07/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Cardiac remodeling following myocardial infarction is a pathological process. We aimed to examine the effect of early short-term minocycline on the left ventricular function following ST-elevation myocardial infarction treated by the primary percutaneous coronary intervention. METHODS In this double-blind, randomized controlled trial, data of 73 patients STEMI patients who were candidates for primary PCI were enrolled. Patients were then randomized to receive minocycline 50 mg orally, followed by 50 mg once a day for 5 days or a placebo with the same schedule. Measurement of serum matrix metalloproteinase-9 (MMP-9) and 2-dimensional speckle tracking echocardiography was performed at baseline and between 4 and 6 months after discharge. Then the demographic, clinical, echocardiographic, and angiographic data, as well as the levels of MMP-9, were compared between the study groups. RESULTS There was no statistically significant difference between the study groups regarding the baseline characteristics. Serum levels of MMP-9 did not change following the intervention within each group and were not significantly different between the groups after follow-up. In the follow-up echocardiography, we also did not observe any difference between the two groups CONCLUSION: In this study, we did not observe any effect of minocycline on cardiac remodeling based on 2-dimensional speckle tracking echocardiography and MMP-9 levels. TRIAL REGISTRATION Iranian Registry of Clinical Trials IRCT201411188698N15 . Registered on 22 June 2015, prospectively.
Collapse
Affiliation(s)
- Alireza Nasiri
- Tehran Heart Center, Cardiovascular Diseases Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Akbar Shafiee
- Tehran Heart Center, Cardiovascular Diseases Research Institute, Tehran University of Medical Sciences, Tehran, Iran.
| | - Ali Hosseinsabet
- Tehran Heart Center, Cardiovascular Diseases Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Azita Hajhosein Talasaz
- Tehran Heart Center, Cardiovascular Diseases Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Arash Jalali
- Tehran Heart Center, Cardiovascular Diseases Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Mojtaba Salarifar
- Tehran Heart Center, Cardiovascular Diseases Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
23
|
Zheng Y, Reinhardt JD, Li J, Hu D, Lin S, Wang L, Dai R, Fan Z, Ding R, Chen L, Yuan L, Xu Z, Cheng Y, Yan C, Zhang X, Wang L, Zhang X, Teng M, Yu Q, Yin A, Lu X. Can Clinical and Functional Outcomes Be Improved with an Intelligent "Internet Plus"-Based Full Disease Cycle Remote Ischemic Conditioning Program in Acute ST-elevation Myocardial Infarction Patients Undergoing Percutaneous Coronary Intervention? Rationale and Design of the i-RIC Trial. Cardiovasc Drugs Ther 2022; 36:45-57. [PMID: 32607820 DOI: 10.1007/s10557-020-07022-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/04/2020] [Indexed: 12/12/2022]
Abstract
BACKGROUND Acute ST-elevation myocardial infarction (STEMI) is associated with a high incidence of complications as well as a considerable hospitalization rate and economic burden. Preliminary evidence suggests that remote ischemic conditioning (RIC) is a promising non-invasive intervention that may effectively and safely reduce myocardial infarct size, subsequent cardiac events and complications, and mortality. However, RIC's cardio-protective effect remains under debate, especially for single timepoint RIC programs. Adequately powered large-scale randomized controlled trials investigating clinical outcomes are thus needed to clarify the role of full disease cycle RIC programs. METHODS The intelligent "Internet Plus"-based full disease cycle remote ischemic conditioning (i-RIC) trial is a pragmatic, multicenter, randomized controlled, parallel group, clinical trial. The term, intelligent "Internet Plus"-based full disease cycle, refers to smart devices aided automatic and real-time monitoring of remote ischemic pre-, per- or post-conditioning intervention for patients with STEMI undergoing percutaneous coronary intervention (PCI). Based on this perspective, 4700 STEMI patients from five hospitals in China will be randomized to a control and an intervention group. The control group will receive PCI and usual care, including pharmacotherapy, before and after PCI. The intervention group will receive pre-, per-, and post-operative RIC combined with long-term i-RIC over a one-month period in addition. A smartphone application, an automated cuff inflation/deflation device and "Internet Plus"-based administration will be used in the long-term phase. The primary outcome is the combined cardiac death or hospitalization for heart failure rate. Secondary outcomes include clinical and functional outcomes: major adverse cardiac and cerebrovascular events rate, all-cause mortality, myocardial reinfarction rate, readmission rate for heart failure and ischemic stroke rate, unplanned revascularization rate, plasma concentration of myocardial infarction-related key biomarkers, infarct size, cardiac function, cardiopulmonary endurance, health-related quality of life, total hospital length of stay, total medical cost, and compliance with treatment regime. DISCUSSION The i-RIC trial is designed to test the hypothesis that clinical and functional outcomes can be improved with the i-RIC program in STEMI patients undergoing PCI. The concept of RIC is expected to be enhanced with this intelligent "Internet Plus"-based program focusing on the full disease cycle. If the i-RIC program results in superior improvement in primary and secondary outcomes, it will offer an innovative treatment option for STEMI patients and form the basis of future recommendations. CLINICAL TRIAL REGISTRATION Chinese Clinical Trial Registry ( http://www.chictr.org.cn ): ChiCTR2000031550, 04 April 2020.
Collapse
Affiliation(s)
- Yu Zheng
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Nanjing Medical University, No.300 Guangzhou Road, Nanjing, 210029, China
| | - Jan D Reinhardt
- Institute for Disaster Management and Reconstruction of Sichuan University and Hongkong Polytechnic University, Chengdu, 610207, China
- Swiss Paraplegic Research, 6207, Nottwil, Switzerland
- Department of Health Sciences and Medicine, University of Lucerne, 6000, Lucerne, Switzerland
| | - Jianan Li
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Nanjing Medical University, No.300 Guangzhou Road, Nanjing, 210029, China
| | - Dayi Hu
- Heart Centre, Peking University People's Hospital, Beijing, 100000, China
| | - Song Lin
- Department of Cardiology, the Affiliated Nanjing First Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Liansheng Wang
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Ruozhu Dai
- Department of Cardiology, Quanzhou First Hospital Affiliated to Fujian Medical University, Quanzhou, 362000, China
| | - Zhiqing Fan
- Department of Cardiology, Daqing Oilfield General Hospital, Daqing, 163001, China
| | - Rongjing Ding
- Heart Centre, Peking University People's Hospital, Beijing, 100000, China
| | - Leilei Chen
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Liang Yuan
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Zhihui Xu
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Yihui Cheng
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Nanjing Medical University, No.300 Guangzhou Road, Nanjing, 210029, China
| | - Chengjie Yan
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Nanjing Medical University, No.300 Guangzhou Road, Nanjing, 210029, China
- Department of Neurorehabilitation, Kunshan Rehabilitation Hospital, Kunshan, 215300, China
| | - Xintong Zhang
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Nanjing Medical University, No.300 Guangzhou Road, Nanjing, 210029, China
| | - Lu Wang
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Nanjing Medical University, No.300 Guangzhou Road, Nanjing, 210029, China
| | - Xiu Zhang
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Nanjing Medical University, No.300 Guangzhou Road, Nanjing, 210029, China
| | - Meiling Teng
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Nanjing Medical University, No.300 Guangzhou Road, Nanjing, 210029, China
| | - Qiuyu Yu
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Nanjing Medical University, No.300 Guangzhou Road, Nanjing, 210029, China
| | - Aimei Yin
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Nanjing Medical University, No.300 Guangzhou Road, Nanjing, 210029, China
| | - Xiao Lu
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Nanjing Medical University, No.300 Guangzhou Road, Nanjing, 210029, China.
| |
Collapse
|
24
|
Haley KE, Almas T, Shoar S, Shaikh S, Azhar M, Cheema FH, Hameed A. The role of anti-inflammatory drugs and nanoparticle-based drug delivery models in the management of ischemia-induced heart failure. Biomed Pharmacother 2021; 142:112014. [PMID: 34391184 DOI: 10.1016/j.biopha.2021.112014] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 07/27/2021] [Accepted: 08/03/2021] [Indexed: 12/20/2022] Open
Abstract
Ongoing advancements in the treatment of acute myocardial infarction (MI) have significantly decreased MI related mortality. Consequently, the number of patients experiencing post-MI heart failure (HF) has continued to rise. Infarction size and the extent of left ventricular (LV) remodeling are largely determined by the extent of ischemia at the time of myocardial injury. In the setting of MI or acute phase of post-MI LV remodeling, anti-inflammatory drugs including intravenous immunoglobulin (IVIG) and Pentoxifylline have shown potential efficacy in preventing post-MI remodeling in-vitro and in some clinical trials. However, systemic administration of anti-inflammatory drugs are not without their off-target side effects. Herein, we explore the clinical feasibility of targeted myocardial delivery of anti-inflammatory drugs via biodegradable polymers, liposomes, hydrogels, and nano-particle based drug delivery models (NDDM) based on existing pre-clinical and clinical models. We summarize the barriers to clinical application of targeted anti-inflammatory delivery post-MI, including challenges in achieving sufficient retention and distribution, as well as the potential need for multiple dosing. Collectively, we suggest that localized delivery of anti-inflammatory agents to the myocardium using NDDM is a promising approach for successful treatment of ischemic HF. Future studies will be instrumental in determining the most effective target and delivery modalities for orchestrating NDDM-mediated treatment of HF.
Collapse
Affiliation(s)
- Kathryn E Haley
- Graduate Entry Medicine, RCSI University of Medicine and Health Sciences, Dublin 2 Dublin, Ireland; Tissue Engineering Research Group (TERG), Department of Anatomy and Regenerative Medicine, RCSI University of Medicine and Health Sciences, Dublin 2 Dublin, Ireland
| | - Talal Almas
- Tissue Engineering Research Group (TERG), Department of Anatomy and Regenerative Medicine, RCSI University of Medicine and Health Sciences, Dublin 2 Dublin, Ireland; School of Medicine, RCSI University of Medicine and Health Sciences, Dublin 2 Dublin, Ireland
| | - Saeed Shoar
- HCA Healthcare Gulf Coast Division, Houston, TX, USA
| | - Shan Shaikh
- HCA Healthcare Gulf Coast Division, Houston, TX, USA
| | - Maimoona Azhar
- Graduate Entry Medicine, RCSI University of Medicine and Health Sciences, Dublin 2 Dublin, Ireland; Department of Surgery, St. Vincent's University Hospital, Dublin 4 Dublin, Ireland
| | - Faisal Habib Cheema
- HCA Healthcare Gulf Coast Division, Houston, TX, USA; University of Houston, College of Medicine, Houston, TX, USA
| | - Aamir Hameed
- Tissue Engineering Research Group (TERG), Department of Anatomy and Regenerative Medicine, RCSI University of Medicine and Health Sciences, Dublin 2 Dublin, Ireland; Trinity Centre for Biomedical Engineering (TCBE), Trinity College Dublin (TCD), Dublin, Ireland.
| |
Collapse
|
25
|
Maleki Dizaji N, Garjani A, Mousavi S, Mohammadi M, Vaez H. Time-dependent influence of infliximab on hemodynamic responses and cardiac injuries of isoproterenol-induced myocardial infarction in rats. Eur J Pharmacol 2021; 903:174122. [PMID: 33932452 DOI: 10.1016/j.ejphar.2021.174122] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Revised: 04/04/2021] [Accepted: 04/21/2021] [Indexed: 10/21/2022]
Abstract
Immune-induced inflammation plays an important role both in aggravating and healing of post myocardial infarction (MI) injuries. Potent anti-inflammatory and local immunomodulatory activity of infliximab has been suggested to have modulating effects on immune responses after MI. The aim of the present study was to evaluate the efficacy of infliximab on hemodynamic responses and myocardial injuries following isoproterenol-induced myocardial infarction. Male Wistar rats, weighting 260 ± 20 g were assigned into ten groups (n = 6) of saline (normal saline), infliximab (7 mg/kg), isoproterenol (100 mg/kg for two consecutive days), and isoproterenol plus infliximab (30 min after the second injection of isoproterenol). The heart tissues and serums were analyzed 24, 48, 72, and 96 h post-MI and hemodynamic parameters, histopathological changes, malondialdehyde (MDA), Total antioxidant capacity (TAC), lactate dehydrogenase (LDH), and lactate levels were assessed in the respective groups. Infliximab partially improved hemodynamic depression in the first days after MI, but the heart became more suppressed later. A similar result also obtained at the MDA tissue levels but not serum levels. Anti-inflammatory effects of Infliximab may improve cardiac function and prevent heart tissue injury early after MI; however, it can worsen the condition later by inhibiting compensatory reactions such as cardiac remodeling and tissue repair.
Collapse
Affiliation(s)
- Nasrin Maleki Dizaji
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Alireza Garjani
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Samin Mousavi
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mahdieh Mohammadi
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Haleh Vaez
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
26
|
Therapies to prevent post-infarction remodelling: From repair to regeneration. Biomaterials 2021; 275:120906. [PMID: 34139506 DOI: 10.1016/j.biomaterials.2021.120906] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2020] [Revised: 05/02/2021] [Accepted: 05/20/2021] [Indexed: 12/15/2022]
Abstract
Myocardial infarction is the first cause of worldwide mortality, with an increasing incidence also reported in developing countries. Over the past decades, preclinical research and clinical trials continually tested the efficacy of cellular and acellular-based treatments. However, none of them resulted in a drug or device currently used in combination with either percutaneous coronary intervention or coronary artery bypass graft. Inflammatory, proliferation and remodelling phases follow the ischaemic event in the myocardial tissue. Only recently, single-cell sequencing analyses provided insights into the specific cell populations which determine the final fibrotic deposition in the affected region. In this review, ischaemia, inflammation, fibrosis, angiogenesis, cellular stress and fundamental cellular and molecular components are evaluated as therapeutic targets. Given the emerging evidence of biomaterial-based systems, the increasing use of injectable hydrogels/scaffolds and epicardial patches is reported both as acellular and cellularised/functionalised treatments. Since several variables influence the outcome of any experimented treatment, we return to the pathological basis with an unbiased view towards any specific process or cellular component. Thus, by evaluating the benefits and limitations of the approaches based on these targets, the reader can weigh the rationale of each of the strategies that reached the clinical trials stage. As recent studies focused on the relevance of the extracellular matrix in modulating ischaemic remodelling and enhancing myocardial regeneration, we aim to portray current trends in the field with this review. Finally, approaches towards feasible translational studies that are as yet unexplored are also suggested.
Collapse
|
27
|
Ministrini S, Carbone F, Montecucco F. Updating concepts on atherosclerotic inflammation: From pathophysiology to treatment. Eur J Clin Invest 2021; 51:e13467. [PMID: 33259635 DOI: 10.1111/eci.13467] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 11/16/2020] [Accepted: 11/28/2020] [Indexed: 12/14/2022]
Abstract
BACKGROUND Atherosclerosis is recognized as a systemic low-grade inflammatory disease. Furthermore, the dysregulation of the inflammatory response and its timely resolution is a pivotal process in determining the clinical manifestations of cardiac and cerebral acute ischaemia following atherothrombosis. METHODS This narrative review is based on the material searched on PubMed up to October 2020. The search terms we used were as follows: "atherosclerosis, inflammation, acute myocardial infarction and ischemic stroke" in combination with "biomarker, inflammatory cells and molecules, treatment." RESULTS The expected goal of addressing inflammation for the treatment of atherosclerosis and its acute ischaemic complications is reducing mortality and morbidity related to atherosclerotic cardiovascular disease, which are currently the first cause of death and disability worldwide. In this narrative review, we summarize the evidence about the main cellular and molecular mechanisms of inflammation in atherogenesis, atherothrombosis and acute ischaemic complications, with particular focus on the potential molecular targets for novel pharmacological treatments. CONCLUSION Although a large amount of evidence from animal models of atherothrombotic disease, and promising results of clinical trials, anti-inflammatory treatments against atherosclerosis are not yet recommended. A deepest understanding of pathophysiological mechanisms underlying the mechanisms driving resolution of the acute inflammation will probably allow to identify the optimal molecular target.
Collapse
Affiliation(s)
- Stefano Ministrini
- Department of Medicine, Internal Medicine, Università degli Studi di Perugia, Perugia, Italy
- Department of Internal Medicine, First Clinic of Internal Medicine, University of Genoa, Genoa, Italy
| | - Federico Carbone
- Department of Internal Medicine, First Clinic of Internal Medicine, University of Genoa, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino, Genoa - Italian Cardiovascular Network, Genoa, Italy
| | - Fabrizio Montecucco
- Department of Internal Medicine, First Clinic of Internal Medicine, University of Genoa, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino, Genoa - Italian Cardiovascular Network, Genoa, Italy
| |
Collapse
|
28
|
Gresele P, Falcinelli E, Momi S, Petito E, Sebastiano M. Platelets and Matrix Metalloproteinases: A Bidirectional Interaction with Multiple Pathophysiologic Implications. Hamostaseologie 2021; 41:136-145. [PMID: 33860521 DOI: 10.1055/a-1393-8339] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Platelets contain and release several matrix metalloproteinases (MMPs), a highly conserved protein family with multiple functions in organism defense and repair. Platelet-released MMPs as well as MMPs generated by other cells within the cardiovascular system modulate platelet function in health and disease. In particular, a normal hemostatic platelet response to vessel wall injury may be transformed into pathological thrombus formation by platelet-released and/or by locally generated MMPs. However, it is becoming increasingly clear that platelets play a role not only in hemostasis but also in immune response, inflammation and allergy, atherosclerosis, and cancer development, and MMPs seem to contribute importantly to this role. A deeper understanding of these mechanisms may open the way to novel therapeutic approaches to the inhibition of their pathogenic effects and lead to significant advances in the treatment of cardiovascular, inflammatory, and neoplastic disorders.
Collapse
Affiliation(s)
- P Gresele
- Department of Medicine and Surgery, Section of Internal and Cardiovascular Medicine, University of Perugia, Perugia, Italy
| | - E Falcinelli
- Department of Medicine and Surgery, Section of Internal and Cardiovascular Medicine, University of Perugia, Perugia, Italy
| | - S Momi
- Department of Medicine and Surgery, Section of Internal and Cardiovascular Medicine, University of Perugia, Perugia, Italy
| | - E Petito
- Department of Medicine and Surgery, Section of Internal and Cardiovascular Medicine, University of Perugia, Perugia, Italy
| | - M Sebastiano
- Department of Medicine and Surgery, Section of Internal and Cardiovascular Medicine, University of Perugia, Perugia, Italy
| |
Collapse
|
29
|
Sarker H, Haimour A, Toor R, Fernandez-Patron C. The Emerging Role of Epigenetic Mechanisms in the Causation of Aberrant MMP Activity during Human Pathologies and the Use of Medicinal Drugs. Biomolecules 2021; 11:biom11040578. [PMID: 33920915 PMCID: PMC8071227 DOI: 10.3390/biom11040578] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2021] [Revised: 04/12/2021] [Accepted: 04/14/2021] [Indexed: 12/15/2022] Open
Abstract
Matrix metalloproteinases (MMPs) cleave extracellular matrix proteins, growth factors, cytokines, and receptors to influence organ development, architecture, function, and the systemic and cell-specific responses to diseases and pharmacological drugs. Conversely, many diseases (such as atherosclerosis, arthritis, bacterial infections (tuberculosis), viral infections (COVID-19), and cancer), cholesterol-lowering drugs (such as statins), and tetracycline-class antibiotics (such as doxycycline) alter MMP activity through transcriptional, translational, and post-translational mechanisms. In this review, we summarize evidence that the aforementioned diseases and drugs exert significant epigenetic pressure on genes encoding MMPs, tissue inhibitors of MMPs, and factors that transcriptionally regulate the expression of MMPs. Our understanding of human pathologies associated with alterations in the proteolytic activity of MMPs must consider that these pathologies and their medicinal treatments may impose epigenetic pressure on the expression of MMP genes. Whether the epigenetic mechanisms affecting the activity of MMPs can be therapeutically targeted warrants further research.
Collapse
|
30
|
Wüst RCI, Coolen BF, Held NM, Daal MRR, Alizadeh Tazehkandi V, Baks-te Bulte L, Wiersma M, Kuster DWD, Brundel BJJM, van Weeghel M, Strijkers GJ, Houtkooper RH. The Antibiotic Doxycycline Impairs Cardiac Mitochondrial and Contractile Function. Int J Mol Sci 2021; 22:4100. [PMID: 33921053 PMCID: PMC8071362 DOI: 10.3390/ijms22084100] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 04/08/2021] [Accepted: 04/08/2021] [Indexed: 12/22/2022] Open
Abstract
Tetracycline antibiotics act by inhibiting bacterial protein translation. Given the bacterial ancestry of mitochondria, we tested the hypothesis that doxycycline-which belongs to the tetracycline class-reduces mitochondrial function, and results in cardiac contractile dysfunction in cultured H9C2 cardiomyoblasts, adult rat cardiomyocytes, in Drosophila and in mice. Ampicillin and carbenicillin were used as control antibiotics since these do not interfere with mitochondrial translation. In line with its specific inhibitory effect on mitochondrial translation, doxycycline caused a mitonuclear protein imbalance in doxycycline-treated H9C2 cells, reduced maximal mitochondrial respiration, particularly with complex I substrates, and mitochondria appeared fragmented. Flux measurements using stable isotope tracers showed a shift away from OXPHOS towards glycolysis after doxycycline exposure. Cardiac contractility measurements in adult cardiomyocytes and Drosophila melanogaster hearts showed an increased diastolic calcium concentration, and a higher arrhythmicity index. Systolic and diastolic dysfunction were observed after exposure to doxycycline. Mice treated with doxycycline showed mitochondrial complex I dysfunction, reduced OXPHOS capacity and impaired diastolic function. Doxycycline exacerbated diastolic dysfunction and reduced ejection fraction in a diabetes mouse model vulnerable for metabolic derangements. We therefore conclude that doxycycline impairs mitochondrial function and causes cardiac dysfunction.
Collapse
Affiliation(s)
- Rob C. I. Wüst
- Laboratory Genetic Metabolic Diseases, Amsterdam Gastroenterology, Endocrinology, and Metabolism, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands; (N.M.H.); (V.A.T.); (M.v.W.)
- Biomedical Engineering and Physics, Amsterdam Cardiovascular Sciences, University of Amsterdam, Amsterdam UMC, 1105 AZ Amsterdam, The Netherlands; (B.F.C.); (M.R.R.D.); (G.J.S.)
- Laboratory for Myology, Department of Human Movement Sciences, Faculty of Behavioural and Movement Sciences, Amsterdam Movement Sciences, Vrije University Amsterdam, 1081 BT Amsterdam, The Netherlands
| | - Bram F. Coolen
- Biomedical Engineering and Physics, Amsterdam Cardiovascular Sciences, University of Amsterdam, Amsterdam UMC, 1105 AZ Amsterdam, The Netherlands; (B.F.C.); (M.R.R.D.); (G.J.S.)
| | - Ntsiki M. Held
- Laboratory Genetic Metabolic Diseases, Amsterdam Gastroenterology, Endocrinology, and Metabolism, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands; (N.M.H.); (V.A.T.); (M.v.W.)
| | - Mariah R. R. Daal
- Biomedical Engineering and Physics, Amsterdam Cardiovascular Sciences, University of Amsterdam, Amsterdam UMC, 1105 AZ Amsterdam, The Netherlands; (B.F.C.); (M.R.R.D.); (G.J.S.)
| | - Vida Alizadeh Tazehkandi
- Laboratory Genetic Metabolic Diseases, Amsterdam Gastroenterology, Endocrinology, and Metabolism, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands; (N.M.H.); (V.A.T.); (M.v.W.)
| | - Luciënne Baks-te Bulte
- Department of Physiology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, VU University Medical Center, 1081 HZ Amsterdam, The Netherlands; (L.B.-t.B.); (M.W.); (D.W.D.K.); (B.J.J.M.B.)
| | - Marit Wiersma
- Department of Physiology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, VU University Medical Center, 1081 HZ Amsterdam, The Netherlands; (L.B.-t.B.); (M.W.); (D.W.D.K.); (B.J.J.M.B.)
| | - Diederik W. D. Kuster
- Department of Physiology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, VU University Medical Center, 1081 HZ Amsterdam, The Netherlands; (L.B.-t.B.); (M.W.); (D.W.D.K.); (B.J.J.M.B.)
| | - Bianca J. J. M. Brundel
- Department of Physiology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, VU University Medical Center, 1081 HZ Amsterdam, The Netherlands; (L.B.-t.B.); (M.W.); (D.W.D.K.); (B.J.J.M.B.)
| | - Michel van Weeghel
- Laboratory Genetic Metabolic Diseases, Amsterdam Gastroenterology, Endocrinology, and Metabolism, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands; (N.M.H.); (V.A.T.); (M.v.W.)
| | - Gustav J. Strijkers
- Biomedical Engineering and Physics, Amsterdam Cardiovascular Sciences, University of Amsterdam, Amsterdam UMC, 1105 AZ Amsterdam, The Netherlands; (B.F.C.); (M.R.R.D.); (G.J.S.)
- Department of Radiology, Translational and Molecular Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Riekelt H. Houtkooper
- Laboratory Genetic Metabolic Diseases, Amsterdam Gastroenterology, Endocrinology, and Metabolism, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands; (N.M.H.); (V.A.T.); (M.v.W.)
| |
Collapse
|
31
|
Hadzic S, Wu CY, Gredic M, Kojonazarov B, Pak O, Kraut S, Sommer N, Kosanovic D, Grimminger F, Schermuly RT, Seeger W, Bellusci S, Weissmann N. The effect of long-term doxycycline treatment in a mouse model of cigarette smoke-induced emphysema and pulmonary hypertension. Am J Physiol Lung Cell Mol Physiol 2021; 320:L903-L915. [PMID: 33760647 DOI: 10.1152/ajplung.00048.2021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is a major cause of death and a still incurable disease, comprising emphysema and chronic bronchitis. In addition to airflow limitation, patients with COPD can suffer from pulmonary hypertension (PH). Doxycycline, an antibiotic from the tetracycline family, in addition to its pronounced antimicrobial activity, acts as a matrix metalloproteinase (MMP) inhibitor and has anti-inflammatory properties. Furthermore, doxycycline treatment exhibited a beneficial effect in several preclinical cardiovascular disease models. In preclinical research, doxycycline is frequently employed for gene expression modulation in Tet-On/Tet-Off transgenic animal models. Therefore, it is crucial to know whether doxycycline treatment in Tet-On/Tet-Off systems has effects independent of gene expression modulation by such systems. Against this background, we assessed the possible curative effects of long-term doxycycline administration in a mouse model of chronic CS exposure. Animals were exposed to cigarette smoke (CS) for 8 mo and then subsequently treated with doxycycline for additional 3 mo in room air conditions. Doxycycline decreased the expression of MMPs and general pro-inflammatory markers in the lungs from CS-exposed mice. This downregulation was, however, insufficient to ameliorate CS-induced emphysema or PH. Tet-On/Tet-Off induction by doxycycline in such models is a feasible genetic approach to study curative effects at least in established CS-induced emphysema and PH. However, we report several parameters that are influenced by doxycycline and use of a Tet-On/Tet-Off system when evaluating those parameters should be interpreted with caution.
Collapse
Affiliation(s)
- Stefan Hadzic
- Cardiopulmonary Institute (CPI), University of Giessen and Marburg Lung Center (UGMLC), German Center for Lung Research (DZL), Justus-Liebig-University, Giessen, Germany
| | - Cheng-Yu Wu
- Cardiopulmonary Institute (CPI), University of Giessen and Marburg Lung Center (UGMLC), German Center for Lung Research (DZL), Justus-Liebig-University, Giessen, Germany
| | - Marija Gredic
- Cardiopulmonary Institute (CPI), University of Giessen and Marburg Lung Center (UGMLC), German Center for Lung Research (DZL), Justus-Liebig-University, Giessen, Germany
| | - Baktybek Kojonazarov
- Cardiopulmonary Institute (CPI), University of Giessen and Marburg Lung Center (UGMLC), German Center for Lung Research (DZL), Justus-Liebig-University, Giessen, Germany.,Institute for Lung Health (ILH), Justus-Liebig-University, Giessen, Germany
| | - Oleg Pak
- Cardiopulmonary Institute (CPI), University of Giessen and Marburg Lung Center (UGMLC), German Center for Lung Research (DZL), Justus-Liebig-University, Giessen, Germany
| | - Simone Kraut
- Cardiopulmonary Institute (CPI), University of Giessen and Marburg Lung Center (UGMLC), German Center for Lung Research (DZL), Justus-Liebig-University, Giessen, Germany
| | - Natascha Sommer
- Cardiopulmonary Institute (CPI), University of Giessen and Marburg Lung Center (UGMLC), German Center for Lung Research (DZL), Justus-Liebig-University, Giessen, Germany
| | - Djuro Kosanovic
- Cardiopulmonary Institute (CPI), University of Giessen and Marburg Lung Center (UGMLC), German Center for Lung Research (DZL), Justus-Liebig-University, Giessen, Germany.,Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| | - Friedrich Grimminger
- Cardiopulmonary Institute (CPI), University of Giessen and Marburg Lung Center (UGMLC), German Center for Lung Research (DZL), Justus-Liebig-University, Giessen, Germany
| | - Ralph T Schermuly
- Cardiopulmonary Institute (CPI), University of Giessen and Marburg Lung Center (UGMLC), German Center for Lung Research (DZL), Justus-Liebig-University, Giessen, Germany
| | - Werner Seeger
- Cardiopulmonary Institute (CPI), University of Giessen and Marburg Lung Center (UGMLC), German Center for Lung Research (DZL), Justus-Liebig-University, Giessen, Germany.,Institute for Lung Health (ILH), Justus-Liebig-University, Giessen, Germany.,Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Saverio Bellusci
- Cardiopulmonary Institute (CPI), University of Giessen and Marburg Lung Center (UGMLC), German Center for Lung Research (DZL), Justus-Liebig-University, Giessen, Germany
| | - Norbert Weissmann
- Cardiopulmonary Institute (CPI), University of Giessen and Marburg Lung Center (UGMLC), German Center for Lung Research (DZL), Justus-Liebig-University, Giessen, Germany
| |
Collapse
|
32
|
Sethi NJ, Safi S, Korang SK, Hróbjartsson A, Skoog M, Gluud C, Jakobsen JC. Antibiotics for secondary prevention of coronary heart disease. Cochrane Database Syst Rev 2021; 2:CD003610. [PMID: 33704780 PMCID: PMC8094925 DOI: 10.1002/14651858.cd003610.pub4] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
BACKGROUND Coronary heart disease is the leading cause of mortality worldwide with approximately 7.4 million deaths each year. People with established coronary heart disease have a high risk of subsequent cardiovascular events including myocardial infarction, stroke, and cardiovascular death. Antibiotics might prevent such outcomes due to their antibacterial, antiinflammatory, and antioxidative effects. However, a randomised clinical trial and several observational studies have suggested that antibiotics may increase the risk of cardiovascular events and mortality. Furthermore, several non-Cochrane Reviews, that are now outdated, have assessed the effects of antibiotics for coronary heart disease and have shown conflicting results. No previous systematic review using Cochrane methodology has assessed the effects of antibiotics for coronary heart disease. OBJECTIVES We assessed the benefits and harms of antibiotics compared with placebo or no intervention for the secondary prevention of coronary heart disease. SEARCH METHODS We searched CENTRAL, MEDLINE, Embase, LILACS, SCI-EXPANDED, and BIOSIS in December 2019 in order to identify relevant trials. Additionally, we searched TRIP, Google Scholar, and nine trial registries in December 2019. We also contacted 11 pharmaceutical companies and searched the reference lists of included trials, previous systematic reviews, and other types of reviews. SELECTION CRITERIA Randomised clinical trials assessing the effects of antibiotics versus placebo or no intervention for secondary prevention of coronary heart disease in adult participants (≥18 years). Trials were included irrespective of setting, blinding, publication status, publication year, language, and reporting of our outcomes. DATA COLLECTION AND ANALYSIS Three review authors independently extracted data. Our primary outcomes were all-cause mortality, serious adverse event according to the International Conference on Harmonization - Good Clinical Practice (ICH-GCP), and quality of life. Our secondary outcomes were cardiovascular mortality, myocardial infarction, stroke, and sudden cardiac death. Our primary time point of interest was at maximum follow-up. Additionally, we extracted outcome data at 24±6 months follow-up. We assessed the risks of systematic errors using Cochrane 'Rosk of bias' tool. We calculated risk ratios (RRs) with 95% confidence intervals (CIs) for dichotomous outcomes. We calculated absolute risk reduction (ARR) or increase (ARI) and number needed to treat for an additional beneficial outcome (NNTB) or for an additional harmful outcome (NNTH) if the outcome result showed a beneficial or harmful effect, respectively. The certainty of the body of evidence was assessed by GRADE. MAIN RESULTS We included 38 trials randomising a total of 26,638 participants (mean age 61.6 years), with 23/38 trials reporting data on 26,078 participants that could be meta-analysed. Three trials were at low risk of bias and the 35 remaining trials were at high risk of bias. Trials assessing the effects of macrolides (28 trials; 22,059 participants) and quinolones (two trials; 4162 participants) contributed with the vast majority of the data. Meta-analyses at maximum follow-up showed that antibiotics versus placebo or no intervention seemed to increase the risk of all-cause mortality (RR 1.06; 95% CI 0.99 to 1.13; P = 0.07; I2 = 0%; ARI 0.48%; NNTH 208; 25,774 participants; 20 trials; high certainty of evidence), stroke (RR 1.14; 95% CI 1.00 to 1.29; P = 0.04; I2 = 0%; ARI 0.73%; NNTH 138; 14,774 participants; 9 trials; high certainty of evidence), and probably also cardiovascular mortality (RR 1.11; 95% CI 0.98 to 1.25; P = 0.11; I2= 0%; 4674 participants; 2 trials; moderate certainty of evidence). Little to no difference was observed when assessing the risk of myocardial infarction (RR 0.95; 95% CI 0.88 to 1.03; P = 0.23; I2 = 0%; 25,523 participants; 17 trials; high certainty of evidence). No evidence of a difference was observed when assessing sudden cardiac death (RR 1.08; 95% CI 0.90 to 1.31; P = 0.41; I2 = 0%; 4520 participants; 2 trials; moderate certainty of evidence). Meta-analyses at 24±6 months follow-up showed that antibiotics versus placebo or no intervention increased the risk of all-cause mortality (RR 1.25; 95% CI 1.06 to 1.48; P = 0.007; I2 = 0%; ARI 1.26%; NNTH 79 (95% CI 335 to 42); 9517 participants; 6 trials; high certainty of evidence), cardiovascular mortality (RR 1.50; 95% CI 1.17 to 1.91; P = 0.001; I2 = 0%; ARI 1.12%; NNTH 89 (95% CI 261 to 49); 9044 participants; 5 trials; high certainty of evidence), and probably also sudden cardiac death (RR 1.77; 95% CI 1.28 to 2.44; P = 0.0005; I2 = 0%; ARI 1.9%; NNTH 53 (95% CI 145 to 28); 4520 participants; 2 trials; moderate certainty of evidence). No evidence of a difference was observed when assessing the risk of myocardial infarction (RR 0.95; 95% CI 0.82 to 1.11; P = 0.53; I2 = 43%; 9457 participants; 5 trials; moderate certainty of evidence) and stroke (RR 1.17; 95% CI 0.90 to 1.52; P = 0.24; I2 = 0%; 9457 participants; 5 trials; high certainty of evidence). Meta-analyses of trials at low risk of bias differed from the overall analyses when assessing cardiovascular mortality at maximum follow-up. For all other outcomes, meta-analyses of trials at low risk of bias did not differ from the overall analyses. None of the trials specifically assessed serious adverse event according to ICH-GCP. No data were found on quality of life. AUTHORS' CONCLUSIONS Our present review indicates that antibiotics (macrolides or quinolones) for secondary prevention of coronary heart disease seem harmful when assessing the risk of all-cause mortality, cardiovascular mortality, and stroke at maximum follow-up and all-cause mortality, cardiovascular mortality, and sudden cardiac death at 24±6 months follow-up. Current evidence does, therefore, not support the clinical use of macrolides and quinolones for the secondary prevention of coronary heart disease. Future trials on the safety of macrolides or quinolones for the secondary prevention in patients with coronary heart disease do not seem ethical. In general, randomised clinical trials assessing the effects of antibiotics, especially macrolides and quinolones, need longer follow-up so that late-occurring adverse events can also be assessed.
Collapse
Affiliation(s)
- Naqash J Sethi
- Copenhagen Trial Unit, Centre for Clinical Intervention Research, The Capital Region of Denmark, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Sanam Safi
- Copenhagen Trial Unit, Centre for Clinical Intervention Research, The Capital Region of Denmark, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Steven Kwasi Korang
- Copenhagen Trial Unit, Centre for Clinical Intervention Research, The Capital Region of Denmark, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Asbjørn Hróbjartsson
- Centre for Evidence-Based Medicine Odense (CEBMO) and Cochrane Denmark, Department of Clinical Research, University of Southern Denmark, Odense, Denmark
- Open Patient data Explorative Network (OPEN), Odense University Hospital, Odense, Denmark
| | - Maria Skoog
- Copenhagen Trial Unit, Centre for Clinical Intervention Research, The Capital Region of Denmark, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
- Clinical Study Support, Clinical Studies Sweden - Forum South, Lund, Sweden
| | - Christian Gluud
- Copenhagen Trial Unit, Centre for Clinical Intervention Research, The Capital Region of Denmark, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
- Cochrane Hepato-Biliary Group, Copenhagen Trial Unit, Centre for Clinical Intervention Research, The Capital Region of Denmark, Rigshospitalet, Copenhagen, Denmark
- Department of Regional Health Research, The Faculty of Health Sciences, University of Southern Denmark, Odense, Denmark
| | - Janus C Jakobsen
- Copenhagen Trial Unit, Centre for Clinical Intervention Research, The Capital Region of Denmark, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
- Cochrane Hepato-Biliary Group, Copenhagen Trial Unit, Centre for Clinical Intervention Research, The Capital Region of Denmark, Rigshospitalet, Copenhagen, Denmark
- Department of Regional Health Research, The Faculty of Health Sciences, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
33
|
Watson C, Spiers JP, Waterstone M, Russell-Hallinan A, Gallagher J, McDonald K, Ryan C, Gilmer J, Ledwidge M. Investigation of association of genetic variant rs3918242 of matrix metalloproteinase-9 with hypertension, myocardial infarction and progression of ventricular dysfunction in Irish Caucasian patients with diabetes: a report from the STOP-HF follow-up programme. BMC Cardiovasc Disord 2021; 21:87. [PMID: 33579197 PMCID: PMC7879511 DOI: 10.1186/s12872-021-01860-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Accepted: 01/07/2021] [Indexed: 01/08/2023] Open
Abstract
Background Hypertension and/or myocardial infarction are common causes of heart failure in Type 2 diabetes. Progression to heart failure is usually preceded by ventricular dysfunction, linked to matrix metalloproteinase (MMP) mediated extracellular matrix changes. We hypothesise that the minor allele of genetic variant rs3918242 in the promoter region of the MMP-9 gene is associated with hypertension and/or myocardial infarction, with resultant progression of dysfunctional cardiac remodelling in patients with diabetes without symptomatic heart failure.
Methods We genotyped 498 diabetes patients participating in the St Vincent’s Screening TO Prevent Heart Failure (STOP-HF) follow-up programme for the rs3918242 single nucleotide polymorphism and investigated associations with the co-primary endpoints hypertension and/or myocardial infarction using a dominant model. We also evaluated resulting cardiometabolic phenotype and progression of ventricular dysfunction and cardiac structural abnormalities over a median follow-up period of 3.5 years. Results The CT/TT genotype comprised 28.1% of the cohort and was associated with a twofold higher risk of myocardial infarction (17.9% vs 8.4%), a reduction in ejection fraction and greater left ventricular systolic dysfunction progression [adjusted OR = 2.56 (1.09, 6.01), p = 0.026] over a median follow-up of 3.5 years [IQR 2.6, 4.9 years]. Conversely, rs3918242 was not associated with hypertension, blood pressure, pulse pressure or left ventricular mass index at baseline or over follow up. Conclusions Diabetes patients with the minor T allele of rs3918242 in the STOP-HF follow up programme have greater risk of myocardial infarction, lower ejection fraction and greater progression of left ventricular systolic abnormalities, a precursor to heart failure. These data may support further work on MMP-9 as a biomarker of ventricular dysfunction and the investigation of MMP-9 inhibitors for heart failure prevention in diabetes, particularly in the post-infarction setting. ClinicalTrials.gov Identifier: NCT00921960
Collapse
Affiliation(s)
- Chris Watson
- STOP-HF Unit, St. Vincent's University Healthcare Group, Dublin, Ireland.,Wellcome-Wolfson Institute for Experimental Medicine, Queen's University , Belfast, Northern Ireland.,School of Medicine, University College Dublin, Dublin, Ireland
| | - J Paul Spiers
- Department of Pharmacology and Therapeutics, Trinity College Dublin, Dublin, Ireland
| | - Max Waterstone
- Department of Pharmacology and Therapeutics, Trinity College Dublin, Dublin, Ireland
| | - Adam Russell-Hallinan
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University , Belfast, Northern Ireland
| | - Joseph Gallagher
- STOP-HF Unit, St. Vincent's University Healthcare Group, Dublin, Ireland.,School of Medicine, University College Dublin, Dublin, Ireland
| | - Kenneth McDonald
- STOP-HF Unit, St. Vincent's University Healthcare Group, Dublin, Ireland.,School of Medicine, University College Dublin, Dublin, Ireland
| | - Cristin Ryan
- School of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, Dublin, Ireland
| | - John Gilmer
- School of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, Dublin, Ireland
| | - Mark Ledwidge
- STOP-HF Unit, St. Vincent's University Healthcare Group, Dublin, Ireland. .,School of Medicine, University College Dublin, Dublin, Ireland.
| |
Collapse
|
34
|
Abstract
Diffuse myocardial fibrosis resulting from the excessive deposition of collagen fibres through the entire myocardium is encountered in a number of chronic cardiac diseases. This lesion results from alterations in the regulation of fibrillary collagen turnover by fibroblasts, facilitating the excessive deposition of type I and type III collagen fibres within the myocardial interstitium and around intramyocardial vessels. The available evidence suggests that, beyond the extent of fibrous deposits, collagen composition and the physicochemical properties of the fibres are also relevant in the detrimental effects of diffuse myocardial fibrosis on cardiac function and clinical outcomes in patients with heart failure. In this regard, findings from the past 20 years suggest that various clinicopathological phenotypes of diffuse myocardial fibrosis exist in patients with heart failure. In this Review, we summarize the current knowledge on the mechanisms and detrimental consequences of diffuse myocardial fibrosis in heart failure. Furthermore, we discuss the validity and usefulness of available imaging techniques and circulating biomarkers to assess the clinicopathological variation in this lesion and to track its clinical evolution. Finally, we highlight the currently available and potential future therapeutic strategies aimed at personalizing the prevention and reversal of diffuse myocardial fibrosis in patients with heart failure.
Collapse
|
35
|
Platt BN, Jacobs CA, Conley CEW, Stone AV. Tetracycline use in treating osteoarthritis: a systematic review. Inflamm Res 2021; 70:249-259. [PMID: 33512569 DOI: 10.1007/s00011-021-01435-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 12/11/2020] [Accepted: 01/11/2021] [Indexed: 11/27/2022] Open
Abstract
BACKGROUND AND AIMS The purpose of the review was to synthesize the current literature regarding tetracyclines in the treatment of osteoarthritis. METHODS Using multiple databases, a systematic review was performed with customized search terms crafted to identify studies examining doxycycline or minocycline in the treatment of osteoarthritis. Results were classified into basic science mechanistic studies, in vivo animal studies, and human clinical trials. A total of 1446 potentially relevant studies were reviewed, and after exclusion criteria were applied, 23 investigations were included in the final analysis. RESULTS From 12 basic science mechanistic studies, we report on three main mechanisms by which tetracyclines may exert benefit in osteoarthritis progression: matrix metalloproteinase inhibition, immunomodulation, and nitric oxide synthase inhibition. Seven animal studies showed generally encouraging results. Four articles reported human clinical studies, showing mixed results in the treatment of osteoarthritis, potentially related to the choice of patient population, primary outcomes, and timing of treatment. CONCLUSION Tetracyclines have the potential to benefit osteoarthritis patients via multiple mechanisms. Further study is warranted to examine the optimal dose and timing of tetracycline treatment in osteoarthritis.
Collapse
Affiliation(s)
- Brooks N Platt
- Division of Sports Medicine, Department of Orthopaedic Surgery and Sports Medicine, University of Kentucky, 740 S. Limestone, K403, Lexington, KY, 40536, USA
| | - Cale A Jacobs
- Division of Sports Medicine, Department of Orthopaedic Surgery and Sports Medicine, University of Kentucky, 740 S. Limestone, K403, Lexington, KY, 40536, USA
| | - Caitlin E W Conley
- Division of Sports Medicine, Department of Orthopaedic Surgery and Sports Medicine, University of Kentucky, 740 S. Limestone, K403, Lexington, KY, 40536, USA
| | - Austin V Stone
- Division of Sports Medicine, Department of Orthopaedic Surgery and Sports Medicine, University of Kentucky, 740 S. Limestone, K403, Lexington, KY, 40536, USA.
| |
Collapse
|
36
|
Chan BYH, Roczkowsky A, Cho WJ, Poirier M, Sergi C, Keschrumrus V, Churko JM, Granzier H, Schulz R. MMP inhibitors attenuate doxorubicin cardiotoxicity by preventing intracellular and extracellular matrix remodelling. Cardiovasc Res 2021; 117:188-200. [PMID: 31995179 PMCID: PMC7797218 DOI: 10.1093/cvr/cvaa017] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Revised: 12/18/2019] [Accepted: 01/21/2020] [Indexed: 12/20/2022] Open
Abstract
AIMS Heart failure is a major complication in cancer treatment due to the cardiotoxic effects of anticancer drugs, especially from the anthracyclines such as doxorubicin (DXR). DXR enhances oxidative stress and stimulates matrix metalloproteinase-2 (MMP-2) in cardiomyocytes. We investigated whether MMP inhibitors protect against DXR cardiotoxicity given the role of MMP-2 in proteolyzing sarcomeric proteins in the heart and remodelling the extracellular matrix. METHODS AND RESULTS Eight-week-old male C57BL/6J mice were treated with DXR weekly with or without MMP inhibitors doxycycline or ONO-4817 by daily oral gavage for 4 weeks. Echocardiography was used to determine cardiac function and left ventricular remodelling before and after treatment. MMP inhibitors ameliorated DXR-induced systolic and diastolic dysfunction by reducing the loss in left ventricular ejection fraction, fractional shortening, and E'/A'. MMP inhibitors attenuated adverse left ventricular remodelling, reduced cardiomyocyte dropout, and prevented myocardial fibrosis. DXR increased myocardial MMP-2 activity in part also by upregulating N-terminal truncated MMP-2. Immunogold transmission electron microscopy showed that DXR elevated MMP-2 levels within the sarcomere and mitochondria which were associated with myofilament lysis, mitochondrial degeneration, and T-tubule distention. DXR-induced myofilament lysis was associated with increased titin proteolysis in the heart which was prevented by ONO-4817. DXR also increased the level and activity of MMP-2 in human embryonic stem cell-derived cardiomyocytes, which was reduced by ONO-4817. CONCLUSIONS MMP-2 activation is an early event in DXR cardiotoxicity and contributes to myofilament lysis by proteolyzing cardiac titin. Two orally available MMP inhibitors ameliorated DXR cardiotoxicity by attenuating intracellular and extracellular matrix remodelling, suggesting their use may be a potential prophylactic strategy to prevent heart injury during chemotherapy.
Collapse
Affiliation(s)
- Brandon Y H Chan
- Department of Pediatrics and Pharmacology, Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, AB T6G 2S2, Canada
| | - Andrej Roczkowsky
- Department of Pediatrics and Pharmacology, Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, AB T6G 2S2, Canada
| | - Woo Jung Cho
- Faculty of Medicine and Dentistry Cell Imaging Centre, University of Alberta, Edmonton, AB, Canada
| | - Mathieu Poirier
- Department of Pediatrics and Pharmacology, Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, AB T6G 2S2, Canada
| | - Consolato Sergi
- Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, AB, Canada
| | - Vic Keschrumrus
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ, USA
| | - Jared M Churko
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ, USA
| | - Henk Granzier
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ, USA
| | - Richard Schulz
- Department of Pediatrics and Pharmacology, Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, AB T6G 2S2, Canada
| |
Collapse
|
37
|
Frangogiannis NG, Kovacic JC. Extracellular Matrix in Ischemic Heart Disease, Part 4/4: JACC Focus Seminar. J Am Coll Cardiol 2020; 75:2219-2235. [PMID: 32354387 DOI: 10.1016/j.jacc.2020.03.020] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2019] [Revised: 02/27/2020] [Accepted: 03/03/2020] [Indexed: 02/06/2023]
Abstract
Myocardial ischemia and infarction, both in the acute and chronic phases, are associated with cardiomyocyte loss and dramatic changes in the cardiac extracellular matrix (ECM). It has long been appreciated that these changes in the cardiac ECM result in altered mechanical properties of ischemic or infarcted myocardial segments. However, a growing body of evidence now clearly demonstrates that these alterations of the ECM not only affect the structural properties of the ischemic and post-infarct heart, but they also play a crucial and sometimes direct role in mediating a range of biological pathways, including the orchestration of inflammatory and reparative processes, as well as the pathogenesis of adverse remodeling. This final part of a 4-part JACC Focus Seminar reviews the evidence on the role of the ECM in relation to the ischemic and infarcted heart, as well as its contribution to cardiac dysfunction and adverse clinical outcomes.
Collapse
Affiliation(s)
- Nikolaos G Frangogiannis
- Wilf Family Cardiovascular Research Institute, Department of Medicine (Cardiology), Albert Einstein College of Medicine, Bronx, New York.
| | - Jason C Kovacic
- The Zena and Michael A. Wiener Cardiovascular Institute, Icahn School of Medicine at Mount Sinai, New York, New York; Victor Chang Cardiac Research Institute and St. Vincent's Clinical School, University of New South Wales, Darlinghurst, New South Wales, Australia.
| |
Collapse
|
38
|
Pharmacological Modulation of Cardiac Remodeling after Myocardial Infarction. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:8815349. [PMID: 33488934 PMCID: PMC7790555 DOI: 10.1155/2020/8815349] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 11/13/2020] [Accepted: 12/21/2020] [Indexed: 12/14/2022]
Abstract
Cardiac remodeling describes a series of structural and functional changes in the heart after myocardial infarction (MI). Adverse post-MI cardiac remodeling directly jeopardizes the recovery of cardiac functions and the survival rate in MI patients. Several classes of drugs are proven to be useful to reduce the mortality of MI patients. However, it is an ongoing challenge to prevent the adverse effects of cardiac remodeling. The present review aims to identify the pharmacological therapies from the existing clinical drugs for the treatment of adverse post-MI cardiac remodeling. Post-MI cardiac remodeling is a complex process involving ischemia/reperfusion, inflammation, cell death, and deposition of extracellular matrix (ECM). Thus, the present review included two parts: (1) to examine the basic pathophysiology in the cardiovascular system and the molecular basis of cardiac remodeling and (2) to identify the pathological aspects of cardiac remodeling and the potential of the existing pharmacotherapies. Ultimately, the present review highlights drug repositioning as a strategy to discover effective therapies from the existing drugs against post-MI cardiac remodeling.
Collapse
|
39
|
de Miranda DC, de Oliveira Faria G, Hermidorff MM, Dos Santos Silva FC, de Assis LVM, Isoldi MC. Pre- and Post-Conditioning of the Heart: An Overview of Cardioprotective Signaling Pathways. Curr Vasc Pharmacol 2020; 19:499-524. [PMID: 33222675 DOI: 10.2174/1570161119666201120160619] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 11/02/2020] [Accepted: 11/02/2020] [Indexed: 11/22/2022]
Abstract
Since the discovery of ischemic pre- and post-conditioning, more than 30 years ago, the knowledge about the mechanisms and signaling pathways involved in these processes has significantly increased. In clinical practice, on the other hand, such advancement has yet to be seen. This article provides an overview of ischemic pre-, post-, remote, and pharmacological conditioning related to the heart. In addition, we reviewed the cardioprotective signaling pathways and therapeutic agents involved in the above-mentioned processes, aiming to provide a comprehensive evaluation of the advancements in the field. The advancements made over the last decades cannot be ignored and with the exponential growth in techniques and applications. The future of pre- and post-conditioning is promising.
Collapse
Affiliation(s)
- Denise Coutinho de Miranda
- Laboratory of Cell Signaling, Research Center in Biological Science, Institute of Exact and Biological Sciences, Federal University of Ouro Preto, Ouro Preto, Brazil
| | - Gabriela de Oliveira Faria
- Laboratory of Cell Signaling, Research Center in Biological Science, Institute of Exact and Biological Sciences, Federal University of Ouro Preto, Ouro Preto, Brazil
| | - Milla Marques Hermidorff
- Laboratory of Cell Signaling, Research Center in Biological Science, Institute of Exact and Biological Sciences, Federal University of Ouro Preto, Ouro Preto, Brazil
| | - Fernanda Cacilda Dos Santos Silva
- Laboratory of Cardiovascular Physiology, Department of Biological Science, Institute of Exact and Biological Sciences, Federal University of Ouro Preto, Ouro Preto, Brazil
| | - Leonardo Vinícius Monteiro de Assis
- Laboratory of Comparative Physiology of Pigmentation, Department of Physiology, Institute of Biosciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Mauro César Isoldi
- Laboratory of Cell Signaling, Research Center in Biological Science, Institute of Exact and Biological Sciences, Federal University of Ouro Preto, Ouro Preto, Brazil
| |
Collapse
|
40
|
Alam MM, Mahmud S, Rahman MM, Simpson J, Aggarwal S, Ahmed Z. Clinical Outcomes of Early Treatment With Doxycycline for 89 High-Risk COVID-19 Patients in Long-Term Care Facilities in New York. Cureus 2020; 12:e9658. [PMID: 32802622 PMCID: PMC7419149 DOI: 10.7759/cureus.9658] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Accepted: 08/11/2020] [Indexed: 12/27/2022] Open
Abstract
Rationale Due to the cluster and associated comorbidities in residents of long-term care facilities (LTCFs), COVID-19-associated morbidity and mortality are significantly increased. Multiple therapeutic options, including hydroxychloroquine (HCQ) and azithromycin (AZI), were tried initially to treat moderate to severe COVID-19 and high-risk patients in LTCFs, but they were abandoned due to unfavorable reports. As a less toxic option, we initiated treatment with doxycycline (DOXY) very early in the course of illness. DOXY has antiviral, cardioprotective, immunomodulatory, and anti-inflammatory properties, but the efficacy of early intervention with DOXY in high-risk COVID-19 patients in LTCFs is unknown. Objective The goal of this retrospective study is to describe the clinical outcomes of high-risk COVID-19 patients with moderate to severe symptoms in LTCFs after early intervention with DOXY. Design Case-series analysis Setting LTCFs in New York Participants This observational study examines 89 patients who were diagnosed with COVID-19 from March 18 to May 13, 2020. Exposure All patients who were diagnosed with COVID-19 received DOXY and regular standard of care within 12 hours of the onset of symptoms. Additionally, four patients received meropenem, three patients received Zosyn, two patients received linezolid, and two patients received Bactrim DS. Four patients were on chronic ventilator support. No patients received any steroids or any other antiviral or immunomodulatory agents. The majority of the patients received zinc and calcium supplements as well. Main outcomes and measures Assessed measures were patients' characteristics, fever, shortness of breath (SOB), cough, oxygen saturation/pulse oximetry (POX), radiologic improvements, laboratory tests, DOXY side effects, hospital transfers, and death. Results Eighty-nine (89) high-risk patients, who developed a sudden onset of fever, cough, SOB, and hypoxia and were diagnosed with COVID-19, were treated with DOXY (100 mg PO or intravenous (IV) for seven days) and regular standard of care. Eighty-five percent (85%) of patients (n=76) demonstrated clinical recovery that is defined as resolution of fever (average 3.7 days, Coeff = -0.96, p = 0.0001), resolution of SOB (average 4.2 days), and improvement of POX: average 84% before treatment and average 95% after treatment (84.7 ± 7% vs. 95 ± 2.6%, p = 0.0001). Higher pre- and post-treatment POX is associated with lower mortality (oxygen saturation (Spo2) vs. Death, Coeff = -0.01, p = 0.023; post-Spo2 vs. Death, Coeff = -0.05, p = 0.0002). Within 10 days of symptom onset, 3% of patients (n=3) were transferred to hospital due to clinical deterioration and 11% of patients (n=10) died. The result was followed for 30 days from the onset of symptoms in each patient. Conclusion Early treatment with DOXY for high-risk patients with moderate to severe COVID-19 infections in non-hospital settings, such as LTCFs, is associated with early clinical recovery, decreased hospitalization, and decreased mortality.
Collapse
Affiliation(s)
- Mohammud M Alam
- Department of Medicine and Infectious Diseases, Northwell University Hospital, Plainview, USA
| | - Saborny Mahmud
- Department of Medicine, Johns Hopkins Bloomberg School of Public Health, Baltimore, USA
| | - Mohammad M Rahman
- Department of Medicine, New York University (NYU) School of Medicine, New York, USA
| | - JoAnn Simpson
- Department of Nursing, Stony Brook University, Stony Brook, USA
| | - Sandeep Aggarwal
- Department of Nephrology, University of Pennsylvania, Philadelphia, USA
| | - Ziauddin Ahmed
- Department of Medicine and Nephrology, Temple University, Philadelphia, USA
| |
Collapse
|
41
|
Mahtta D, Sudhakar D, Koneru S, Silva GV, Alam M, Virani SS, Jneid H. Targeting Inflammation After Myocardial Infarction. Curr Cardiol Rep 2020; 22:110. [PMID: 32770365 DOI: 10.1007/s11886-020-01358-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
PURPOSE OF REVIEW Inflammation plays a key role in clearing cellular debris and recovery after acute myocardial infarction (AMI). Dysregulation of or prolonged inflammation may result in adverse cardiac remodeling and major adverse clinical events (MACE). Several pre-clinical studies and moderate sized clinical trials have investigated the role of immunomodulation in improving clinical outcomes in patients with AMI. RECENT FINDINGS Clinical data from the Canakinumab Atherothrombosis Outcome (CANTOS) and Colchicine Cardiovascular Outcomes Trial (COLCOT) have provided encouraging results among patients with AMI. Several other clinical and pre-clinical trials have brought about the prospect of modulating inflammation at various junctures of the inflammatory cascade including inhibition of complement cascade, interleukins, and matrix metalloproteinases. In patients with AMI, modulation of residual inflammation via various inflammatory pathways and mediators may hold promise for further reducing MACE. Learning from current data and understanding the nuances of immunomodulation in AMI are key for future trials and before widespread dissemination of such therapies.
Collapse
Affiliation(s)
- Dhruv Mahtta
- Health Policy, Quality & Informatics Program,, Michael E. DeBakey VA Medical Center Health Services Research & Development Center for Innovations in Quality, Effectiveness, and Safety, Houston, TX, USA
- Division of Cardiovascular Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Deepthi Sudhakar
- Division of Cardiovascular Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Srikanth Koneru
- Division of Cardiovascular Medicine,, Texas Heart Institute and Baylor College of Medicine, Houston, TX, USA
| | - Guilherme Vianna Silva
- Division of Cardiovascular Medicine,, Texas Heart Institute and Baylor College of Medicine, Houston, TX, USA
| | - Mahboob Alam
- Division of Cardiovascular Medicine,, Texas Heart Institute and Baylor College of Medicine, Houston, TX, USA
| | - Salim S Virani
- Health Policy, Quality & Informatics Program,, Michael E. DeBakey VA Medical Center Health Services Research & Development Center for Innovations in Quality, Effectiveness, and Safety, Houston, TX, USA
- Division of Cardiovascular Medicine, Baylor College of Medicine, Houston, TX, USA
- Section of Cardiology, Michael E. DeBakey Veterans Affairs Medical Center, Houston, TX, 77030, USA
| | - Hani Jneid
- Division of Cardiovascular Medicine, Baylor College of Medicine, Houston, TX, USA.
- Section of Cardiology, Michael E. DeBakey Veterans Affairs Medical Center, Houston, TX, 77030, USA.
| |
Collapse
|
42
|
Matrix Metalloproteinases as Biomarkers of Atherosclerotic Plaque Instability. Int J Mol Sci 2020; 21:ijms21113946. [PMID: 32486345 PMCID: PMC7313469 DOI: 10.3390/ijms21113946] [Citation(s) in RCA: 116] [Impact Index Per Article: 23.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 05/27/2020] [Accepted: 05/29/2020] [Indexed: 02/07/2023] Open
Abstract
Matrix metalloproteinases (MMPs) are a family of zinc-dependent endopeptidases responsible for tissue remodeling and degradation of extracellular matrix (ECM) proteins. MMPs may modulate various cellular and signaling pathways in atherosclerosis responsible for progression and rupture of atherosclerotic plaques. The effect of MMPs polymorphisms and the expression of MMPs in both the atherosclerotic plaque and plasma was shown. They are independent predictors of atherosclerotic plaque instability in stable coronary heart disease (CHD) patients. Increased levels of MMPs in patients with advanced cardiovascular disease (CAD) and acute coronary syndrome (ACS) was associated with future risk of cardiovascular events. These data confirm that MMPs may be biomarkers in plaque instability as they target in potential drug therapies for atherosclerosis. They provide important prognostic information, independent of traditional risk factors, and may turn out to be useful in improving risk stratification.
Collapse
|
43
|
Affiliation(s)
- Amy McMillan
- Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute (A.M., S.L.H.).,Center for Microbiome & Human Health (A.M., S.L.H.)
| | - Stanley L Hazen
- Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute (A.M., S.L.H.).,Center for Microbiome & Human Health (A.M., S.L.H.).,Department of Cardiovascular Medicine, Cleveland Clinic, OH (S.L.H.)
| |
Collapse
|
44
|
Clemens DL, Duryee MJ, Hall JH, Thiele GM, Mikuls TR, Klassen LW, Zimmerman MC, Anderson DR. Relevance of the antioxidant properties of methotrexate and doxycycline to their treatment of cardiovascular disease. Pharmacol Ther 2019; 205:107413. [PMID: 31626869 DOI: 10.1016/j.pharmthera.2019.107413] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Accepted: 09/15/2019] [Indexed: 12/21/2022]
Abstract
Many medications exhibit clinical benefits that are unrelated to their primary therapeutic uses. In many cases, the mechanisms underpinning these pleotropic effects are unknown. Two commonly prescribed medications that exhibit pleotropic benefits in cardiovascular disease and other diseases associated with chronic inflammation are methotrexate (MTX) and doxycycline (DOX). The vast majority of cardiovascular disease is associated with atherosclerosis. Because atherosclerosis is a chronic inflammatory disease, possible mechanisms by which MTX and DOX reduce inflammation have been investigated. Interestingly, the primary structure of both of these medications contain aromatic phenolic rings, which resemble polyphenols that are known to possess antioxidant activity. Inflammation and oxidative stress are intimately related. Inflammation promotes oxidative stress, which in turn leads to further inflammation; in this way, oxidative stress and inflammation can establish a self-perpetuating cycle. It has been shown that MTX and DOX act as antioxidants and are capable of scavenging free radicals and the reactive oxygen species (ROS) superoxide (O2-). Furthermore, both MTX and DOX inhibit the formation of malondialdehyde acetaldehyde (MAA) adducts, products of oxidative stress and lipid peroxidation. Importantly, MAA-adducts are highly immunogenic and initiate inflammatory responses; thereby, fueling the cycle of inflammation and oxidative stress that results in chronic inflammation. Thus, reducing the formation of MAA-adducts may ameliorate inflammation that leads to ROS production and in this way, break the self-sustaining cycle of oxidative stress and inflammation. It is possible that the under-recognized antioxidant properties of these medications may be a mechanism by which they and other medications provide pleotropic benefit in the treatment of chronic inflammatory disease.
Collapse
Affiliation(s)
- Dahn L Clemens
- Department of Internal Medicine, University of Nebraska Medical Center, 982650 Nebraska Medical Center, Omaha, NE, 68198-2265, United States; Veterans Affairs (VA) Nebraska-Western Iowa Health Care System, 4101 Woolworth Ave., Omaha, NE, 68105, United States; Fred and Pamela Buffet Cancer Center, Nebraska Medical Center, Omaha, NE, 68114, United States
| | - Michael J Duryee
- Department of Internal Medicine, University of Nebraska Medical Center, 982650 Nebraska Medical Center, Omaha, NE, 68198-2265, United States; Veterans Affairs (VA) Nebraska-Western Iowa Health Care System, 4101 Woolworth Ave., Omaha, NE, 68105, United States
| | - Johnathan H Hall
- Department of Internal Medicine, University of Nebraska Medical Center, 982650 Nebraska Medical Center, Omaha, NE, 68198-2265, United States
| | - Geoffrey M Thiele
- Department of Internal Medicine, University of Nebraska Medical Center, 982650 Nebraska Medical Center, Omaha, NE, 68198-2265, United States; Veterans Affairs (VA) Nebraska-Western Iowa Health Care System, 4101 Woolworth Ave., Omaha, NE, 68105, United States
| | - Ted R Mikuls
- Department of Internal Medicine, University of Nebraska Medical Center, 982650 Nebraska Medical Center, Omaha, NE, 68198-2265, United States; Veterans Affairs (VA) Nebraska-Western Iowa Health Care System, 4101 Woolworth Ave., Omaha, NE, 68105, United States
| | - Lynell W Klassen
- Department of Internal Medicine, University of Nebraska Medical Center, 982650 Nebraska Medical Center, Omaha, NE, 68198-2265, United States
| | - Matthew C Zimmerman
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, 982650 Nebraska Medical Center, Omaha, NE, 68198-2265, United States
| | - Daniel R Anderson
- Department of Internal Medicine, University of Nebraska Medical Center, 982650 Nebraska Medical Center, Omaha, NE, 68198-2265, United States.
| |
Collapse
|
45
|
Chan BYH, Roczkowsky A, Cho WJ, Poirier M, Lee TYT, Mahmud Z, Schulz R. Junctophilin-2 is a target of matrix metalloproteinase-2 in myocardial ischemia-reperfusion injury. Basic Res Cardiol 2019; 114:42. [PMID: 31506724 DOI: 10.1007/s00395-019-0749-7] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Accepted: 09/02/2019] [Indexed: 11/30/2022]
Abstract
Junctophilin-2 is a structural membrane protein that tethers T-tubules to the sarcoplasmic reticulum to allow for coordinated calcium-induced calcium release in cardiomyocytes. Defective excitation-contraction coupling in myocardial ischemia-reperfusion (IR) injury is associated with junctophilin-2 proteolysis. However, it remains unclear whether preventing junctophilin-2 proteolysis improves the recovery of cardiac contractile dysfunction in IR injury. Matrix metalloproteinase-2 (MMP-2) is a zinc and calcium-dependent protease that is activated by oxidative stress in myocardial IR injury and cleaves both intracellular and extracellular substrates. To determine whether junctophilin-2 is targeted by MMP-2, isolated rat hearts were perfused in working mode aerobically or subjected to IR injury with the selective MMP inhibitor ARP-100. IR injury impaired the recovery of cardiac contractile function which was associated with increased degradation of junctophilin-2 and damaged cardiac dyads. In IR hearts, ARP-100 improved the recovery of cardiac contractile function, attenuated junctophilin-2 proteolysis, and prevented ultrastructural damage to the dyad. MMP-2 was co-localized with junctophilin-2 in aerobic and IR hearts by immunoprecipitation and immunohistochemistry. In situ zymography showed that MMP activity was localized to the Z-disc and sarcomere in aerobic hearts and accumulated at sites where the striated JPH-2 staining was disrupted in IR hearts. In vitro proteolysis assays determined that junctophilin-2 is susceptible to proteolysis by MMP-2 and in silico analysis predicted multiple MMP-2 cleavage sites between the membrane occupation and recognition nexus repeats and within the divergent region of junctophilin-2. Degradation of junctophilin-2 by MMP-2 is an early consequence of myocardial IR injury which may initiate a cascade of sequelae leading to impaired contractile function.
Collapse
Affiliation(s)
- Brandon Y H Chan
- Departments of Pediatrics and Pharmacology, Mazankowski Alberta Heart Institute, 462 Heritage Medical Research Centre, University of Alberta, Edmonton, AB, T6G 2S2, Canada
| | - Andrej Roczkowsky
- Departments of Pediatrics and Pharmacology, Mazankowski Alberta Heart Institute, 462 Heritage Medical Research Centre, University of Alberta, Edmonton, AB, T6G 2S2, Canada
| | - Woo Jung Cho
- Faculty of Medicine and Dentistry Cell Imaging Centre, University of Alberta, Edmonton, AB, Canada
| | - Mathieu Poirier
- Departments of Pediatrics and Pharmacology, Mazankowski Alberta Heart Institute, 462 Heritage Medical Research Centre, University of Alberta, Edmonton, AB, T6G 2S2, Canada
| | - Tim Y T Lee
- Departments of Pediatrics and Pharmacology, Mazankowski Alberta Heart Institute, 462 Heritage Medical Research Centre, University of Alberta, Edmonton, AB, T6G 2S2, Canada
| | - Zabed Mahmud
- Department of Biochemistry, University of Alberta, Edmonton, AB, Canada
| | - Richard Schulz
- Departments of Pediatrics and Pharmacology, Mazankowski Alberta Heart Institute, 462 Heritage Medical Research Centre, University of Alberta, Edmonton, AB, T6G 2S2, Canada.
| |
Collapse
|
46
|
Abstract
Matrix metalloproteinases (MMPs) and their endogenous inhibitors have been studied in the myocardium for the past 2 decades. An incomplete knowledge base and experimental design issues with inhibitors have hampered attempts at translation, but clinical interest remains high because of strong associations between MMPs and outcomes after myocardial infarction (MI) as well as mechanistic studies showing MMP involvement at multiple stages of the MI wound-healing process. This Review focuses on how our understanding of MMPs has evolved from a one-dimensional early focus on measuring MMP activity, monitoring MMP:inhibitor ratios, and evaluating one MMP-substrate pair to the current use of systems biology approaches to integrate the whole MMP repertoire of roles in the left ventricular response to MI. MMP9 is used as an example MMP to explain these concepts and to provide a template for examining MMPs as mechanistic mediators of cardiac remodelling.
Collapse
Affiliation(s)
- Merry L Lindsey
- Mississippi Center for Heart Research, Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS, USA. .,Research Service,, G.V. (Sonny) Montgomery Veterans Affairs Medical Center, Jackson, MS, USA.
| |
Collapse
|
47
|
Singh B, Ghosh N, Saha D, Sarkar S, Bhattacharyya P, Chaudhury K. Effect of doxycyline in chronic obstructive pulmonary disease - An exploratory study. Pulm Pharmacol Ther 2019; 58:101831. [PMID: 31349003 DOI: 10.1016/j.pupt.2019.101831] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Revised: 07/15/2019] [Accepted: 07/23/2019] [Indexed: 01/09/2023]
Abstract
PURPOSE Various mechanisms, including oxidative stress, inflammation, and protease-antiprotease imbalance are proposed for the progressive decline in lung function in chronic obstructive pulmonary disease (COPD). Doxycycline, a broad spectrum tetracycline antibiotic, is reported to have non-antimicrobial matrix metalloproteinases (MMP) inhibitory action in various inflammatory conditions. The effect of doxycycline in COPD is hereby assessed in the present randomized prospective study. PATIENTS AND METHODS The first group of COPD patients (n = 30; mild (n = 3), moderate (n = 6), severe (n = 7), very severe (n = 14) as per GOLD II & III criteria was prescribed the standard therapy, a combination of (i) short acting anti-muscarinic agent (SAMA) + short acting β2 agonist (SABA) inhaled and (ii) corticosteroid inhaled (ICS) + long acting β2 agonist (LABA) (iii) ICS + LABA + LAMA. Whereas doxycycline (100 mg), was used daily once or twice as per Body Mass Index (BMI), as an add-on to existing standard therapy for the second group of patients (n = 30; mild (n = 2), moderate (n = 7), severe (n = 8), very severe (n = 13). All recruited patients were followed-up after 3 months of treatment. Lung function index FEV1(%) predicted, FEV1/FVC (%), quality of life status including COPD Assessment Test (CAT), St. George's Respiratory Questionnaire (SGRQ) were assessed. Routine blood cell count also was performed. RESULTS Biochemical analysis included estimation of oxidative stress markers, inflammatory cytokines and proteases in plasma of both the groups. Reduction in oxidative stress is evidenced by a significant decrease in Lipid hydro peroxides (LPO), total oxidative stress (TOS) and increase in glutathione peroxidase (GSH-PX), reduced glutathione (GSH) and total anti-oxidant capacity (TAO) nitrite and nitrate (NOx) along with peroxynitrate following 3 months of add-on doxycycline treatment. Reduced levels of cytokines such as interleukin IL-6, TNF-α, IL-8 were also observed. Multivariate analysis identified TNF-α major effective discriminant among pre and post doxycycline treated COPD patients. The expression of TNF-α was inversely correlated with FEV1/FVC (%) changes. The levels of MMP-2 and MMP-9/tissue inhibitors of metalloproteinases (TIMP)-1 ratio (MMP-9/ TIMP-1), also decreased significantly and the decline could be associated with TOS. A significant increase in bilirubin and reduced glutathione (GSH) level was noticed in standard therapy group. CONCLUSION These data suggest that the improvement in lung function and quality of life in COPD patients may probably be attributed to the antioxidant, anti-inflammatory and anti-MMP activity of doxycycline. The potential therapeutic role of long-term doxycycline, in addition to its traditional antibiotic effect, definitely warrants further attention.
Collapse
Affiliation(s)
- Brajesh Singh
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, West Bengal, India
| | - Nilanjana Ghosh
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, West Bengal, India
| | | | - Surita Sarkar
- Department of Applied Physics, University of Calcutta, Kolkata, India
| | | | - Koel Chaudhury
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, West Bengal, India.
| |
Collapse
|
48
|
Mao S, Vincent M, Chen M, Zhang M, Hinek A. Exploration of Multiple Signaling Pathways Through Which Sodium Tanshinone IIA Sulfonate Attenuates Pathologic Remodeling Experimental Infarction. Front Pharmacol 2019; 10:779. [PMID: 31354493 PMCID: PMC6639725 DOI: 10.3389/fphar.2019.00779] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Accepted: 06/17/2019] [Indexed: 12/20/2022] Open
Abstract
The level of maladaptive myocardial remodeling consistently contributes to the poor prognosis of patients following a myocardial infarction (MI). In this study, we investigated whether and how sodium tanshinone IIA sulfonate (STS) would attenuate the post-infarct cardiac remodeling in mice model of MI developing after surgical ligation of the left coronary artery. All mice subjected to experimental MI or to the sham procedure were then treated for the following 4 weeks, either with STS or with a vehicle alone. Results of our studies indicated that STS treatment of MI mice prevented the left ventricular dilatation and improved their cardiac function. Results of further tests, aimed at mechanistic explanation of the beneficial effects of STS, indicated that treatment with this compound enhanced the autophagy and, at the same time, inhibited apoptosis of the cardiomyocytes. Meaningfully, we have also established that myocardium of STS-treated mice displayed significantly higher levels of adenosine monophosphate kinase than their untreated counterparts and that this effect additionally associated with the significantly diminished activities of apoptotic promoters: mammalian target of rapamycin and P70S6 kinase. Moreover, we also found that additional administration of the adenosine monophosphate kinase inhibitor (compound C) or autophagy inhibitor (chloroquine) practically eliminated the observed beneficial effects of STS. In conclusion, we suggest that the described multistage mechanism triggered by STS treatment enhanced autophagy, thereby attenuating pathologic remodeling of the post-infarct hearts.
Collapse
Affiliation(s)
- Shuai Mao
- Key Discipline of Integrated Traditional Chinese and Western Medicine, Second Clinical College, Guangzhou University of Chinese Medicine, Guangzhou, China
- Department of Critical Care Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
| | - Matthew Vincent
- Medical School, St. George’s, University of London, London, United Kingdom
| | - Maosheng Chen
- Key Discipline of Integrated Traditional Chinese and Western Medicine, Second Clinical College, Guangzhou University of Chinese Medicine, Guangzhou, China
- Department of Critical Care Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
| | - Minzhou Zhang
- Key Discipline of Integrated Traditional Chinese and Western Medicine, Second Clinical College, Guangzhou University of Chinese Medicine, Guangzhou, China
- Department of Critical Care Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
| | - Aleksander Hinek
- Physiology & Experimental Medicine, Hospital for Sick Children, Toronto, ON, Canada
| |
Collapse
|
49
|
Schroder J, Mygind ND, Frestad D, Michelsen M, Suhrs HE, Bove KB, Gustafsson I, Kastrup J, Prescott E. Pro-inflammatory biomarkers in women with non-obstructive angina pectoris and coronary microvascular dysfunction. IJC HEART & VASCULATURE 2019; 24:100370. [PMID: 31193994 PMCID: PMC6545380 DOI: 10.1016/j.ijcha.2019.100370] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Accepted: 04/30/2019] [Indexed: 12/13/2022]
Abstract
Background Studies that evaluate larger numbers of protein biomarkers in patients with coronary microvascular dysfunction (CMD) have not previously been performed, and very little is known concerning the pathogenetic mechanisms leading to CMD. Our objective was to analyze associations between a broad cardiovascular disease (CVD) protein biomarker assay and CMD, and further explore internal biomarker relations in order to identify possible targets for future treatment interventions. Methods In 174 women with angina pectoris and no significant obstructive coronary artery disease (<50% stenosis on invasive coronary angiography), CMD was assessed by transthoracic Doppler echocardiography measuring coronary flow velocity reserve (CFVR). Blood samples were analyzed with a CVD proteomic panel encompassing 92 biomarkers. The relation between biomarkers and CFVR was evaluated by regression analysis, and possible interrelations between significant biomarkers were investigated by principal component analysis (PCA). Results Median age (SD) was 64 years (9.8), median CFVR (IQR) was 2.3 (1.9–2.7), and 28% of patients had CFVR < 2.0. Eighteen biomarkers were significantly correlated with CFVR. In PCA, 8 of the biomarkers significantly related to CFVR showed high loadings on principal component 1 (PC1). The component scores of PC1 were significantly related to CFVR (p = 0.002). The majority of the 8 interrelated PC1 biomarkers were related to the pro-inflammatory TNF-α – IL-6 – CRP pathway. Conclusion Eighteen protein biomarkers were significantly associated with CMD. Eight biomarkers were interrelated in PCA, and share connection with pro-inflammatory pathways, highlighting a possible important role of inflammation in CMD.
Collapse
Affiliation(s)
- Jakob Schroder
- Department of Cardiology, Bispebjerg Hospital, University of Copenhagen, Copenhagen, Denmark
- Corresponding author.
| | - Naja Dam Mygind
- Department of Cardiology, Bispebjerg Hospital, University of Copenhagen, Copenhagen, Denmark
- Department of Cardiology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Daria Frestad
- Department of Cardiology, Bispebjerg Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Marie Michelsen
- Department of Cardiology, Bispebjerg Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Hannah Elena Suhrs
- Department of Cardiology, Bispebjerg Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Kira Bang Bove
- Department of Cardiology, Bispebjerg Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Ida Gustafsson
- Department of Cardiology, Bispebjerg Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Jens Kastrup
- Department of Cardiology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Eva Prescott
- Department of Cardiology, Bispebjerg Hospital, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
50
|
Cerisano G, Buonamici P, Migliorini A, Santini A, Marcucci R, Gori AM, Becatti M, Zocchi C, Berteotti M, Valenti R. Relation between post-myocardial infarct remodelling and gelatinase activity in patients enrolled in the TIPTOP trial. Eur J Heart Fail 2018; 21:127-129. [PMID: 30520536 DOI: 10.1002/ejhf.1361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2018] [Revised: 08/27/2018] [Accepted: 10/14/2018] [Indexed: 11/08/2022] Open
Affiliation(s)
| | | | | | - Alberto Santini
- Department of Heart and Vessels, Careggi Hospital, Florence, Italy
| | - Rossella Marcucci
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Anna Maria Gori
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Matteo Becatti
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Chiara Zocchi
- Department of Heart and Vessels, Careggi Hospital, Florence, Italy
| | | | - Renato Valenti
- Department of Heart and Vessels, Careggi Hospital, Florence, Italy
| |
Collapse
|