1
|
Pagliaro P, Weber NC, Femminò S, Alloatti G, Penna C. Gasotransmitters and noble gases in cardioprotection: unraveling molecular pathways for future therapeutic strategies. Basic Res Cardiol 2024; 119:509-544. [PMID: 38878210 PMCID: PMC11319428 DOI: 10.1007/s00395-024-01061-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2024] [Revised: 05/28/2024] [Accepted: 05/29/2024] [Indexed: 08/13/2024]
Abstract
Despite recent progress, ischemic heart disease poses a persistent global challenge, driving significant morbidity and mortality. The pursuit of therapeutic solutions has led to the emergence of strategies such as ischemic preconditioning, postconditioning, and remote conditioning to shield the heart from myocardial ischemia/reperfusion injury (MIRI). These ischemic conditioning approaches, applied before, after, or at a distance from the affected organ, inspire future therapeutic strategies, including pharmacological conditioning. Gasotransmitters, comprising nitric oxide, hydrogen sulfide, sulfur dioxide, and carbon monoxide, play pivotal roles in physiological and pathological processes, exhibiting shared features such as smooth muscle relaxation, antiapoptotic effects, and anti-inflammatory properties. Despite potential risks at high concentrations, physiological levels of gasotransmitters induce vasorelaxation and promote cardioprotective effects. Noble gases, notably argon, helium, and xenon, exhibit organ-protective properties by reducing cell death, minimizing infarct size, and enhancing functional recovery in post-ischemic organs. The protective role of noble gases appears to hinge on their modulation of molecular pathways governing cell survival, leading to both pro- and antiapoptotic effects. Among noble gases, helium and xenon emerge as particularly promising in the field of cardioprotection. This overview synthesizes our current understanding of the roles played by gasotransmitters and noble gases in the context of MIRI and cardioprotection. In addition, we underscore potential future developments involving the utilization of noble gases and gasotransmitter donor molecules in advancing cardioprotective strategies.
Collapse
Affiliation(s)
- Pasquale Pagliaro
- Department of Clinical and Biological Sciences, University of Turin, Regione Gonzole 10, 10043, Orbassano, TO), Italy.
- National Institute for Cardiovascular Research (INRC), 40126, Bologna, Italy.
| | - Nina C Weber
- Department of Anesthesiology, Laboratory of Experimental Intensive Care and Anesthesiology-L.E.I.C.A, Amsterdam University Medical Centers, Amsterdam Cardiovascular Science (ACS), Amsterdam, The Netherlands
| | - Saveria Femminò
- Department of Clinical and Biological Sciences, University of Turin, Regione Gonzole 10, 10043, Orbassano, TO), Italy
| | | | - Claudia Penna
- Department of Clinical and Biological Sciences, University of Turin, Regione Gonzole 10, 10043, Orbassano, TO), Italy
- National Institute for Cardiovascular Research (INRC), 40126, Bologna, Italy
| |
Collapse
|
2
|
Zhang H, Hu H, Zhai C, Jing L, Tian H. Cardioprotective Strategies After Ischemia-Reperfusion Injury. Am J Cardiovasc Drugs 2024; 24:5-18. [PMID: 37815758 PMCID: PMC10806044 DOI: 10.1007/s40256-023-00614-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/28/2023] [Indexed: 10/11/2023]
Abstract
Acute myocardial infarction (AMI) is associated with high morbidity and mortality worldwide. Although early reperfusion is the most effective strategy to salvage ischemic myocardium, reperfusion injury can develop with the restoration of blood flow. Therefore, it is important to identify protection mechanisms and strategies for the heart after myocardial infarction. Recent studies have shown that multiple intracellular molecules and signaling pathways are involved in cardioprotection. Meanwhile, device-based cardioprotective modalities such as cardiac left ventricular unloading, hypothermia, coronary sinus intervention, supersaturated oxygen (SSO2), and remote ischemic conditioning (RIC) have become important areas of research. Herein, we review the molecular mechanisms of cardioprotection and cardioprotective modalities after ischemia-reperfusion injury (IRI) to identify potential approaches to reduce mortality and improve prognosis in patients with AMI.
Collapse
Affiliation(s)
- Honghong Zhang
- Department of Cardiology, Affiliated Hospital of Jiaxing University: First Hospital of Jiaxing, No. 1882 Zhonghuan South Road, Jiaxing, 314000, Zhejiang, People's Republic of China
| | - Huilin Hu
- Department of Cardiology, Affiliated Hospital of Jiaxing University: First Hospital of Jiaxing, No. 1882 Zhonghuan South Road, Jiaxing, 314000, Zhejiang, People's Republic of China.
| | - Changlin Zhai
- Department of Cardiology, Affiliated Hospital of Jiaxing University: First Hospital of Jiaxing, No. 1882 Zhonghuan South Road, Jiaxing, 314000, Zhejiang, People's Republic of China
| | - Lele Jing
- Department of Cardiology, Affiliated Hospital of Jiaxing University: First Hospital of Jiaxing, No. 1882 Zhonghuan South Road, Jiaxing, 314000, Zhejiang, People's Republic of China
| | - Hongen Tian
- Department of Cardiology, Affiliated Hospital of Jiaxing University: First Hospital of Jiaxing, No. 1882 Zhonghuan South Road, Jiaxing, 314000, Zhejiang, People's Republic of China
| |
Collapse
|
3
|
Pathways for Cardioprotection in Perspective: Focus on Remote Conditioning and Extracellular Vesicles. BIOLOGY 2023; 12:biology12020308. [PMID: 36829584 PMCID: PMC9953525 DOI: 10.3390/biology12020308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 02/08/2023] [Accepted: 02/10/2023] [Indexed: 02/17/2023]
Abstract
Despite the development of cutting-edge treatments, coronary artery disease (CAD) morbidity and mortality rates remain present at high levels. Therefore, new cardioprotective approaches are crucial to improve the health of patients. To date, experimental investigations of acute ischemia-reperfusion injury (IRI) have generally demonstrated the efficacy of local ischemic preconditioning and postconditioning cardioprotection techniques as well as of remote conditioning. However, application in clinical settings is still highly controversial and debated. Currently, remote ischemic conditioning (RIC) seems to be the most promising method for heart repair. Protective factors are released into the bloodstream, and protection can be transferred within and across species. For a long time, the cross-function and cross-transmission mechanisms of cardioprotection were largely unknown. Recently, it has been shown that small, anuclear, bilayered lipid membrane particles, known as extracellular vesicles (EVs), are the drivers of signal transduction in cardiac IRI and RIC. EVs are related to the pathophysiological processes of cardiovascular diseases (CVDs), according to compelling evidence. In this review, we will first review the current state of knowledge on myocardial IRI and cardioprotective strategies explored over the past 37 years. Second, we will briefly discuss the role of EVs in CVD and the most recent improvements on EVs as prognostic biomarkers, diagnostic, and therapeutic agents. We will discuss how EVs can be used as a new drug delivery mechanism and how they can be employed in cardiac treatment, also from a perspective of overcoming the impasse that results from neglecting confounding factors.
Collapse
|
4
|
Paz-Artigas L, Montero-Calle P, Iglesias-García O, Mazo MM, Ochoa I, Ciriza J. Current approaches for the recreation of cardiac ischaemic environment in vitro. Int J Pharm 2023; 632:122589. [PMID: 36623742 DOI: 10.1016/j.ijpharm.2023.122589] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 12/14/2022] [Accepted: 01/04/2023] [Indexed: 01/09/2023]
Abstract
Myocardial ischaemia is one of the leading dead causes worldwide. Although animal experiments have historically provided a wealth of information, animal models are time and money consuming, and they usually miss typical human patient's characteristics associated with ischemia prevalence, including aging and comorbidities. Generating reliable in vitro models that recapitulate the human cardiac microenvironment during an ischaemic event can boost the development of new drugs and therapeutic strategies, as well as our understanding of the underlying cellular and molecular events, helping the optimization of therapeutic approaches prior to animal and clinical testing. Although several culture systems have emerged for the recreation of cardiac physiology, mimicking the features of an ischaemic heart tissue in vitro is challenging and certain aspects of the disease process remain poorly addressed. Here, current in vitro cardiac culture systems used for modelling cardiac ischaemia, from self-aggregated organoids to scaffold-based constructs and heart-on-chip platforms are described. The advantages of these models to recreate ischaemic hallmarks such as oxygen gradients, pathological alterations of mechanical strength or fibrotic responses are highlighted. The new models represent a step forward to be considered, but unfortunately, we are far away from recapitulating all complexity of the clinical situations.
Collapse
Affiliation(s)
- Laura Paz-Artigas
- Tissue Microenvironment (TME) Lab, Aragón Institute of Engineering Research (I3A), University of Zaragoza, 50018 Zaragoza, Spain; Institute for Health Research Aragón (IIS Aragón), 50009 Zaragoza, Spain
| | - Pilar Montero-Calle
- Regenerative Medicine Program, Cima Universidad de Navarra, and Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Olalla Iglesias-García
- Regenerative Medicine Program, Cima Universidad de Navarra, and Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Manuel M Mazo
- Regenerative Medicine Program, Cima Universidad de Navarra, and Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain; Hematology and Cell Therapy, Clínica Universidad de Navarra, and Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Ignacio Ochoa
- Tissue Microenvironment (TME) Lab, Aragón Institute of Engineering Research (I3A), University of Zaragoza, 50018 Zaragoza, Spain; Institute for Health Research Aragón (IIS Aragón), 50009 Zaragoza, Spain; CIBER-BBN, ISCIII, Zaragoza, Spain.
| | - Jesús Ciriza
- Tissue Microenvironment (TME) Lab, Aragón Institute of Engineering Research (I3A), University of Zaragoza, 50018 Zaragoza, Spain; Institute for Health Research Aragón (IIS Aragón), 50009 Zaragoza, Spain; CIBER-BBN, ISCIII, Zaragoza, Spain.
| |
Collapse
|
5
|
Insights into research on myocardial ischemia/reperfusion injury from 2012 to 2021: a bibliometric analysis. Eur J Med Res 2023; 28:17. [PMID: 36624514 PMCID: PMC9827672 DOI: 10.1186/s40001-022-00967-7] [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: 12/11/2022] [Accepted: 12/20/2022] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND Numerous studies on myocardial ischemia/reperfusion (MI/R) injury have been undertaken in recent years. Hotspots and developmental trends in MI/R research are being rapidly updated. However, there has been no bibliometric analysis that systematically evaluates existing literature on MI/R injury. Our study explores developments in MI/R research over the past decade, and provides a reference for future research. MATERIALS AND METHODS Both experimental and clinical publications on MI/R injury from 2012 to 2021 were retrieved from the Web of Science Core Collection database. The CiteSpace and VOSviewer tools were used to perform a bibliometric analysis. RESULTS A total of 8419 papers were analyzed. The number of annual publications demonstrated an overall upward trend, rising from 629 publications in 2012 to 1024 publications in 2021. China, the USA, Germany, England, and Italy were the top five contributors to MI/R studies. The Fourth Military Medical University in China contributed the most publications (188, 2.23%), while the University College London in England cooperated the most with relevant research institutions. Derek J Hausenloy (University College London), Derek M Yellon (University College London), and Gerd Heusch (University of Essen Medical School) were the top three most active and influential scholars according to the H-index. Among the top 10 journals with the most publications, Basic Research in Cardiology had the highest impact factors. The top three co-cited journals were Circulation, Circulation Research, and Cardiovascular Research. According to a co-cited reference analysis, MI/R research can be divided across 10 major subfields of mitophagy, cardioprotection, inflammation, remote ischemic preconditioning, long non-coding RNA, melatonin, postconditioning, mitochondria, microvascular obstruction, and ferroptosis. After 2018, the keywords with strongest citation bursts included extracellular vesicles, long non-coding RNA, cell proliferation, microRNA, mitochondrial quality control, mitophagy, biomarker, and mitochondrial biogenesis. CONCLUSIONS The present study reveals the influential authors, cooperating institutions, and main research foci in the field of MI/R injury in the past decade. The latest hotspots are a more in-depth insight into the molecular mechanisms underlying MI/R injury, such as mitochondrial quality control, non-coding RNAs, cell proliferation, and extracellular vesicles.
Collapse
|
6
|
Tarannum A, Arif Z, Mustafa M, Abul Qais F, Habib S, Uddin M, Alam K. Studies on the synergistic action of methylglyoxal and peroxynitrite on structure and function of human serum albumin. J Biomol Struct Dyn 2023; 41:67-80. [PMID: 34842044 DOI: 10.1080/07391102.2021.2003865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Albumin, an important serum protein, is continuously exposed to various oxidizing/nitrating and glycating agents. Depending upon the nature/concentration of reactive species present, the protein may be glycated, oxidized/nitroxidized or glyco-nitro-oxidized. Peroxynitrite is a powerful nitroxidant and has been reported to damage a wide array of macromolecules. On the other hand, methylglyoxal is a very strong reactive dicarbonyl and a potent precursor for the formation of advanced glycation end products under pathological conditions. In certain pathological conditions albumin may be modified by peroxynitrite and methylglyoxal simultaneously. There is dearth of literature suggests that structural/conformational and functional alteration in albumin upon glycation and oxidation/nitroxidation, however the alterations produced by glyco-nitro-oxidation has not yet been explored. Therefore, in this study, simultaneous effect of glycation and nitroxidation on the structure and conformation, vis-a-vis function of albumin was explored. Glyco-nitro-oxidized albumin showed decreased free amino acid content together with decreased affinity of albumin towards cobalt. Molecular docking model and molecular dynamic simulations showed close interaction and formation of stable complexes between methylglyoxal, peroxynitrite and albumin. Formation of carboxymethyl lysine and 3-nitrotyrosine in glyco-nitro-oxidized albumin were confirmed by MALDI-TOF MS and UP-LC MS. Aggregate formation in glyco-nitro-oxidized albumin was visualized by transmission electron microscopy. On the basis of these results, it may be speculated that, albumin modified with endogenously generated methylglyoxal and peroxynitrite might be a driving factor in the progression of heightened inflammatory autoimmune responses. The work presents a ground to study the role of glyco-nitro-oxidized albumin in the pathogenesis and progression of various autoimmune diseases including rheumatoid arthritis. Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Akhlas Tarannum
- Department of Biochemistry, Faculty of Medicine, Jawaharlal Nehru Medical College, Aligarh Muslim University, Aligarh, Uttar Pradesh, India
| | - Zarina Arif
- Department of Biochemistry, Faculty of Medicine, Jawaharlal Nehru Medical College, Aligarh Muslim University, Aligarh, Uttar Pradesh, India
| | - Mohd Mustafa
- Department of Biochemistry, Faculty of Medicine, Jawaharlal Nehru Medical College, Aligarh Muslim University, Aligarh, Uttar Pradesh, India
| | - Faizan Abul Qais
- Dept of Agricultural Microbiology, Faculty of Agricultural Sciences, Aligarh Muslim University, Aligarh, Uttar Pradesh, India
| | - Safia Habib
- Department of Biochemistry, Faculty of Medicine, Jawaharlal Nehru Medical College, Aligarh Muslim University, Aligarh, Uttar Pradesh, India
| | - Moin Uddin
- Department of Biochemistry, Faculty of Medicine, Jawaharlal Nehru Medical College, Aligarh Muslim University, Aligarh, Uttar Pradesh, India
| | - Khursheed Alam
- Department of Biochemistry, Faculty of Medicine, Jawaharlal Nehru Medical College, Aligarh Muslim University, Aligarh, Uttar Pradesh, India
| |
Collapse
|
7
|
Chen L, Weng Y, Qing A, Li J, Yang P, Ye L, Zhu T. Protective Effect of Remote Ischemic Preconditioning against Myocardial Ischemia-Reperfusion Injury in Rats and Mice: A Systematic Review and Meta-Analysis. Rev Cardiovasc Med 2022; 23:413. [PMID: 39076668 PMCID: PMC11270448 DOI: 10.31083/j.rcm2312413] [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: 09/20/2022] [Revised: 11/12/2022] [Accepted: 11/29/2022] [Indexed: 07/31/2024] Open
Abstract
Background Remote ischemic preconditioning (RIPC) has cardioprotective effects. This study was designed to evaluate the effectiveness and potential influencing factors of RIPC for myocardial ischemia-reperfusion injury (MIRI) in rats and mice. Methods The PubMed, Web of Science, Embase, and Cochrane Library databases were searched to identify animal model studies that explored the effect of RIPC on MIRI. The primary outcome was myocardial infarct size, and secondary outcomes included serum cardiac markers, vital signs, hemodynamic parameters, and TUNEL-positive cells. Quality was assessed using SYRCLE's Risk of Bias Tool. Results This systematic review and meta-analysis included 713 male animals from 37 studies. RIPC significantly protected against MIRI in small animal models by reducing infarct size, decreasing serum myocardial marker levels and cell death, and improving cardiac function. Subgroup analysis indicated that RIPC duration and sites influence the protective effect of RIPC on MIRI. Meta-regression suggested that study type and staining method might be sources of heterogeneity. The funnel plot, Egger's test, and Begg's test suggested the existence of publication bias, but results of the sensitivity analysis and nonparametric trim-and-fill method showed that the overall effect of RIPC on MIRI infarct size was robust. Conclusions RIPC significantly protected against MIRI in small animal models by reducing infarct size, decreasing serum myocardial markers and limiting cell death, and improving cardiac function. RIPC duration and site influence the protective effect of RIPC on MIRI, which contributes in reducing confounding factors and determines the best approach for human studies.
Collapse
Affiliation(s)
- Lu Chen
- Department of Anesthesiology, West China Hospital, Sichuan University,
610041 Chengdu, Sichuan, China
| | - Yan Weng
- Department of Anesthesiology, The People's Hospital of Jianyang, 641400
Jianyang, Sichuan, China
| | - Ailing Qing
- Department of Anesthesiology, West China School of Public Health and West
China Fourth Hospital, Sichuan University, 610041 Chengdu, Sichuan, China
| | - Jun Li
- Department of Pain Management, West China Hospital, Sichuan University,
610041 Chengdu, Sichuan, China
| | - Pingliang Yang
- Department of Anesthesiology, The First Affiliated Hospital of Chengdu
Medical College, 610500 Chengdu, Sichuan, China
| | - Ling Ye
- Department of Pain Management, West China Hospital, Sichuan University,
610041 Chengdu, Sichuan, China
| | - Tao Zhu
- Department of Anesthesiology, West China Hospital, Sichuan University,
610041 Chengdu, Sichuan, China
| |
Collapse
|
8
|
Mancardi D, Ottolenghi S, Attanasio U, Tocchetti CG, Paroni R, Pagliaro P, Samaja M. Janus, or the Inevitable Battle Between Too Much and Too Little Oxygen. Antioxid Redox Signal 2022; 37:972-989. [PMID: 35412859 DOI: 10.1089/ars.2021.0232] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Significance: Oxygen levels are key regulators of virtually every living mammalian cell, under both physiological and pathological conditions. Starting from embryonic and fetal development, through the growth, onset, and progression of diseases, oxygen is a subtle, although pivotal, mediator of key processes such as differentiation, proliferation, autophagy, necrosis, and apoptosis. Hypoxia-driven modifications of cellular physiology are investigated in depth or for their clinical and translational relevance, especially in the ischemic scenario. Recent Advances: The mild or severe lack of oxygen is, undoubtedly, related to cell death, although abundant evidence points at oscillating oxygen levels, instead of permanent low pO2, as the most detrimental factor. Different cell types can consume oxygen at different rates and, most interestingly, some cells can shift from low to high consumption according to the metabolic demand. Hence, we can assume that, in the intracellular compartment, oxygen tension varies from low to high levels depending on both supply and consumption. Critical Issues: The positive balance between supply and consumption leads to a pro-oxidative environment, with some cell types facing hypoxia/hyperoxia cycles, whereas some others are under fairly constant oxygen tension. Future Directions: Within this frame, the alterations of oxygen levels (dysoxia) are critical in two paradigmatic organs, the heart and brain, under physiological and pathological conditions and the interactions of oxygen with other physiologically relevant gases, such as nitric oxide, can alternatively contribute to the worsening or protection of ischemic organs. Further, the effects of dysoxia are of pivotal importance for iron metabolism. Antioxid. Redox Signal. 37, 972-989.
Collapse
Affiliation(s)
- Daniele Mancardi
- Department of Clinical and Biological Sciences, University of Torino, Turin, Italy
| | - Sara Ottolenghi
- Department of Health Sciences, University of Milano, Milan, Italy
- School of Medicine and Surgery, University of Milano Bicocca, Milan, Italy
| | - Umberto Attanasio
- Cardio-Oncology Unit, Department of Translational Medical Sciences, Federico II University, Naples, Italy
| | - Carlo Gabriele Tocchetti
- Cardio-Oncology Unit, Department of Translational Medical Sciences, Federico II University, Naples, Italy
- Interdepartmental Center for Clinical and Translational Research (CIRCET), Federico II University, Naples, Italy
- Interdepartmental Hypertension Research Center (CIRIAPA), Federico II University, Naples, Italy
- Center for Basic and Clinical Immunology Research (CISI), Federico II University, Naples, Italy
| | - Rita Paroni
- Department of Health Sciences, University of Milano, Milan, Italy
| | - Pasquale Pagliaro
- Department of Clinical and Biological Sciences, University of Torino, Turin, Italy
| | - Michele Samaja
- Department of Health Sciences, University of Milano, Milan, Italy
- MAGI GROUP, San Felice del Benaco, Italy
| |
Collapse
|
9
|
Effects of Lycopene Attenuating Injuries in Ischemia and Reperfusion. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:9309327. [PMID: 36246396 PMCID: PMC9568330 DOI: 10.1155/2022/9309327] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 07/25/2022] [Accepted: 09/15/2022] [Indexed: 11/18/2022]
Abstract
Tissue and organ ischemia can lead to cell trauma, tissue necrosis, irreversible damage, and death. While intended to reverse ischemia, reperfusion can further aggravate an ischemic injury (ischemia-reperfusion injury, I/R injury) through a range of pathologic processes. An I/R injury to one organ can also harm other organs, leading to systemic multiorgan failure. A type of carotenoid, lycopene, has been shown to treat and prevent many diseases (e.g., rheumatoid arthritis, cancer, diabetes, osteoporosis, male infertility, neurodegenerative diseases, and cardiovascular disease), making it a hot research topic in health care. Some recent researches have suggested that lycopene can evidently ameliorate ischemic and I/R injuries to many organs, but few clinical studies are available. Therefore, it is essential to review the effects of lycopene on ischemic and I/R injuries to different organs, which may help further research into its potential clinical applications.
Collapse
|
10
|
Andelova N, Waczulikova I, Kunstek L, Talian I, Ravingerova T, Jasova M, Suty S, Ferko M. Dichloroacetate as a metabolic modulator of heart mitochondrial proteome under conditions of reduced oxygen utilization. Sci Rep 2022; 12:16348. [PMID: 36175475 PMCID: PMC9522880 DOI: 10.1038/s41598-022-20696-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Accepted: 09/16/2022] [Indexed: 11/08/2022] Open
Abstract
Myocardial compensatory mechanisms stimulated by reduced oxygen utilization caused by streptozotocin-induced diabetes mellitus (DM) and treated with dichloroacetate (DCA) are presumably associated with the regulation of mitochondria. We aimed to promote the understanding of key signaling pathways and identify effectors involved in signal transduction. Proteomic analysis and fluorescence spectroscopy measurements revealed significantly decreased membrane potential and upregulated protein amine oxidase [flavin-containing] A (AOFA) in DM mitochondria, indicative of oxidative damage. DCA in diabetic animals (DM + DCA) downregulated AOFA, increased membrane potential, and stimulated thioredoxin-dependent peroxide reductase, a protein with antioxidant function. Furthermore, the DM condition was associated with mitochondrial resistance to calcium overload through mitochondrial permeability transition pores (mPTPs) regulation, despite an increased protein level of voltage-dependent anion-selective protein (VDAC1). In contrast, DM + DCA influenced ROS levels and downregulated VDAC1 and VDAC3 when compared to DM alone. The diabetic myocardium showed an identical pattern of mPTP protein interactions as in the control group, but the interactions were attenuated. Characterization of the combined effect of DM + DCA is a novel finding showing that DCA acted as an effector of VDAC protein interactions, calcium uptake regulation, and ROS production. Overall, DM and DCA did not exhibit an additive effect, but an individual cardioprotective pathway.
Collapse
Affiliation(s)
- Natalia Andelova
- Centre of Experimental Medicine, Institute for Heart Research, Slovak Academy of Sciences, 84104, Bratislava, Slovakia
| | - Iveta Waczulikova
- Division of Biomedical Physics, Department of Nuclear Physics and Biophysics, Faculty of Mathematics, Physics and Informatics, Comenius University, 84248, Bratislava, Slovakia
| | - Lukas Kunstek
- Centre of Experimental Medicine, Institute for Heart Research, Slovak Academy of Sciences, 84104, Bratislava, Slovakia
| | - Ivan Talian
- Department of Medical and Clinical Biophysics, Faculty of Medicine, P. J. Safarik University, 04011, Kosice, Slovakia
| | - Tanya Ravingerova
- Centre of Experimental Medicine, Institute for Heart Research, Slovak Academy of Sciences, 84104, Bratislava, Slovakia
| | - Magdalena Jasova
- Centre of Experimental Medicine, Institute for Heart Research, Slovak Academy of Sciences, 84104, Bratislava, Slovakia
| | - Simon Suty
- Division of Biomedical Physics, Department of Nuclear Physics and Biophysics, Faculty of Mathematics, Physics and Informatics, Comenius University, 84248, Bratislava, Slovakia
| | - Miroslav Ferko
- Centre of Experimental Medicine, Institute for Heart Research, Slovak Academy of Sciences, 84104, Bratislava, Slovakia.
| |
Collapse
|
11
|
Role of Oxidative Stress in Cardiac Dysfunction and Subcellular Defects Due to Ischemia-Reperfusion Injury. Biomedicines 2022; 10:biomedicines10071473. [PMID: 35884777 PMCID: PMC9313001 DOI: 10.3390/biomedicines10071473] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 06/15/2022] [Accepted: 06/16/2022] [Indexed: 11/17/2022] Open
Abstract
Ischemia-reperfusion (I/R) injury is well-known to be associated with impaired cardiac function, massive arrhythmias, marked alterations in cardiac metabolism and irreversible ultrastructural changes in the heart. Two major mechanisms namely oxidative stress and intracellular Ca2+-overload are considered to explain I/R-induced injury to the heart. However, it is becoming apparent that oxidative stress is the most critical pathogenic factor because it produces myocardial abnormalities directly or indirectly for the occurrence of cardiac damage. Furthermore, I/R injury has been shown to generate oxidative stress by promoting the formation of different reactive oxygen species due to defects in mitochondrial function and depressions in both endogenous antioxidant levels as well as regulatory antioxidative defense systems. It has also been demonstrated to adversely affect a wide variety of metabolic pathways and targets in cardiomyocytes, various resident structures in myocardial interstitium, as well as circulating neutrophils and leukocytes. These I/R-induced alterations in addition to myocardial inflammation may cause cell death, fibrosis, inflammation, Ca2+-handling abnormalities, activation of proteases and phospholipases, as well as subcellular remodeling and depletion of energy stores in the heart. Analysis of results from isolated hearts perfused with or without some antioxidant treatments before subjecting to I/R injury has indicated that cardiac dysfunction is associated with the development of oxidative stress, intracellular Ca2+-overload and protease activation. In addition, changes in the sarcolemma and sarcoplasmic reticulum Ca2+-handling, mitochondrial oxidative phosphorylation as well as myofibrillar Ca2+-ATPase activities in I/R hearts were attenuated by pretreatment with antioxidants. The I/R-induced alterations in cardiac function were simulated upon perfusing the hearts with oxyradical generating system or oxidant. These observations support the view that oxidative stress may be intimately involved in inducing intracellular Ca2+-overload, protease activation, subcellular remodeling, and cardiac dysfunction as a consequence of I/R injury to the heart.
Collapse
|
12
|
Alloatti G, Penna C, Comità S, Tullio F, Aragno M, Biasi F, Pagliaro P. Aging, sex and NLRP3 inflammasome in cardiac ischaemic disease. Vascul Pharmacol 2022; 145:107001. [PMID: 35623548 DOI: 10.1016/j.vph.2022.107001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 05/01/2022] [Accepted: 05/20/2022] [Indexed: 10/18/2022]
Abstract
Experimentally, many strong cardioprotective treatments have been identified in different animal models of acute ischaemia/reperfusion injury (IRI) and coronary artery disease (CAD). However, the translation of these cardioprotective therapies for the benefit of the patients into the clinical scenario has been very disappointing. The reasons for this lack are certainly multiple. Indeed, many confounding factors we must deal in clinical reality, such as aging, sex and inflammatory processes are neglected in many experiments. Due to the pivotal role of aging, sex and inflammation in determining cardiac ischaemic disease, in this review, we take into account age as a modifier of tolerance to IRI in the two sexes, dissecting aging and myocardial reperfusion injury mechanisms and the sex differences in tolerance to IRI. Then we focus on the role of the gut microbiota and the NLRP3 inflammasome in myocardial IRI and on the possibility to consider NLRP3 inflammasome as a potential target in the treatment of CAD in relationship with age and sex. Finally, we consider the cardioprotective mechanisms and cardioprotective treatments during aging in the two sexes.
Collapse
Affiliation(s)
| | - Claudia Penna
- Department of Clinical and Biological Sciences, University of Turin, Regione Gonzole 10, Orbassano, 10043 Torino, TO, Italy; National Institute for Cardiovascular Research (INRC), Bologna, Italy
| | - Stefano Comità
- Department of Clinical and Biological Sciences, University of Turin, Regione Gonzole 10, Orbassano, 10043 Torino, TO, Italy
| | - Francesca Tullio
- Department of Clinical and Biological Sciences, University of Turin, Regione Gonzole 10, Orbassano, 10043 Torino, TO, Italy
| | - Manuela Aragno
- Department of Clinical and Biological Sciences, University of Turin, Regione Gonzole 10, Orbassano, 10043 Torino, TO, Italy
| | - Fiorella Biasi
- Department of Clinical and Biological Sciences, University of Turin, Regione Gonzole 10, Orbassano, 10043 Torino, TO, Italy
| | - Pasquale Pagliaro
- Department of Clinical and Biological Sciences, University of Turin, Regione Gonzole 10, Orbassano, 10043 Torino, TO, Italy; National Institute for Cardiovascular Research (INRC), Bologna, Italy.
| |
Collapse
|
13
|
Signaling Pathways Involved in Myocardial Ischemia-Reperfusion Injury and Cardioprotection: A Systematic Review of Transcriptomic Studies in Sus scrofa. J Cardiovasc Dev Dis 2022; 9:jcdd9050132. [PMID: 35621843 PMCID: PMC9145716 DOI: 10.3390/jcdd9050132] [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: 03/30/2022] [Revised: 04/20/2022] [Accepted: 04/21/2022] [Indexed: 11/17/2022] Open
Abstract
Myocardial damage in acute myocardial infarctions (AMI) is primarily the result of ischemia−reperfusion injury (IRI). Recognizing the timing of transcriptional events and their modulation by cardioprotective strategies is critical to address the pathophysiology of myocardial IRI. Despite the relevance of pigs for translational studies of AMI, only a few have identified how transcriptomic changes shape cellular signaling pathways in response to injury. We systematically reviewed transcriptomic studies of myocardial IRI and cardioprotection in Sus scrofa. Gene expression datasets were analyzed for significantly enriched terms using the Enrichr analysis tool, and statistically significant results (adjusted p-values of <0.05) for Signaling Pathways, Transcription Factors, Molecular Functions, and Biological Processes were compared between eligible studies to describe how these dynamic changes transform the myocardium from an injured and inflamed tissue into a scar. Then, we address how cardioprotective interventions distinctly modulate the myocardial transcriptome and discuss the implications of uncovering gene regulatory networks for cardiovascular pathologies and translational applications.
Collapse
|
14
|
Morelli MB, Bongiovanni C, Da Pra S, Miano C, Sacchi F, Lauriola M, D’Uva G. Cardiotoxicity of Anticancer Drugs: Molecular Mechanisms and Strategies for Cardioprotection. Front Cardiovasc Med 2022; 9:847012. [PMID: 35497981 PMCID: PMC9051244 DOI: 10.3389/fcvm.2022.847012] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Accepted: 03/03/2022] [Indexed: 12/13/2022] Open
Abstract
Chemotherapy and targeted therapies have significantly improved the prognosis of oncology patients. However, these antineoplastic treatments may also induce adverse cardiovascular effects, which may lead to acute or delayed onset of cardiac dysfunction. These common cardiovascular complications, commonly referred to as cardiotoxicity, not only may require the modification, suspension, or withdrawal of life-saving antineoplastic therapies, with the risk of reducing their efficacy, but can also strongly impact the quality of life and overall survival, regardless of the oncological prognosis. The onset of cardiotoxicity may depend on the class, dose, route, and duration of administration of anticancer drugs, as well as on individual risk factors. Importantly, the cardiotoxic side effects may be reversible, if cardiac function is restored upon discontinuation of the therapy, or irreversible, characterized by injury and loss of cardiac muscle cells. Subclinical myocardial dysfunction induced by anticancer therapies may also subsequently evolve in symptomatic congestive heart failure. Hence, there is an urgent need for cardioprotective therapies to reduce the clinical and subclinical cardiotoxicity onset and progression and to limit the acute or chronic manifestation of cardiac damages. In this review, we summarize the knowledge regarding the cellular and molecular mechanisms contributing to the onset of cardiotoxicity associated with common classes of chemotherapy and targeted therapy drugs. Furthermore, we describe and discuss current and potential strategies to cope with the cardiotoxic side effects as well as cardioprotective preventive approaches that may be useful to flank anticancer therapies.
Collapse
Affiliation(s)
| | - Chiara Bongiovanni
- National Laboratory of Molecular Biology and Stem Cell Engineering, National Institute of Biostructures and Biosystems (INBB), Bologna, Italy
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Bologna, Italy
| | - Silvia Da Pra
- National Laboratory of Molecular Biology and Stem Cell Engineering, National Institute of Biostructures and Biosystems (INBB), Bologna, Italy
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Bologna, Italy
| | - Carmen Miano
- National Laboratory of Molecular Biology and Stem Cell Engineering, National Institute of Biostructures and Biosystems (INBB), Bologna, Italy
| | - Francesca Sacchi
- National Laboratory of Molecular Biology and Stem Cell Engineering, National Institute of Biostructures and Biosystems (INBB), Bologna, Italy
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Bologna, Italy
| | - Mattia Lauriola
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Bologna, Italy
| | - Gabriele D’Uva
- National Laboratory of Molecular Biology and Stem Cell Engineering, National Institute of Biostructures and Biosystems (INBB), Bologna, Italy
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Bologna, Italy
- *Correspondence: Gabriele D’Uva,
| |
Collapse
|
15
|
Penna C, Comità S, Tullio F, Alloatti G, Pagliaro P. Challenges facing the clinical translation of cardioprotection: 35 years after the discovery of ischemic preconditioning. Vascul Pharmacol 2022; 144:106995. [DOI: 10.1016/j.vph.2022.106995] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 03/17/2022] [Accepted: 04/16/2022] [Indexed: 12/19/2022]
|
16
|
Xin Y, Zhang X, Li J, Gao H, Li J, Li J, Hu W, Li H. New Insights Into the Role of Mitochondria Quality Control in Ischemic Heart Disease. Front Cardiovasc Med 2021; 8:774619. [PMID: 34901234 PMCID: PMC8661033 DOI: 10.3389/fcvm.2021.774619] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2021] [Accepted: 11/09/2021] [Indexed: 02/05/2023] Open
Abstract
IHD is a significant cause of mortality and morbidity worldwide. In the acute phase, it's demonstrated as myocardial infarction and ischemia-reperfusion injury, while in the chronic stage, the ischemic heart is mainly characterised by adverse myocardial remodelling. Although interventions such as thrombolysis and percutaneous coronary intervention could reduce the death risk of these patients, the underlying cellular and molecular mechanisms need more exploration. Mitochondria are crucial to maintain the physiological function of the heart. During IHD, mitochondrial dysfunction results in the pathogenesis of ischemic heart disease. Ischemia drives mitochondrial damage not only due to energy deprivation, but also to other aspects such as mitochondrial dynamics, mitochondria-related inflammation, etc. Given the critical roles of mitochondrial quality control in the pathological process of ischemic heart disease, in this review, we will summarise the efforts in targeting mitochondria (such as mitophagy, mtROS, and mitochondria-related inflammation) on IHD. In addition, we will briefly revisit the emerging therapeutic targets in this field.
Collapse
Affiliation(s)
- Yanguo Xin
- Department of Cardiology, Cardiovascular Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Xiaodong Zhang
- General Surgery Department, Beijing Friendship Hospital, Capital Medical University, Beijing, China.,National Clinical Research Center for Digestive Diseases, Beijing, China
| | - Jingye Li
- Department of Cardiology, Cardiovascular Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Hui Gao
- Department of Cardiology, Cardiovascular Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Jiayu Li
- Department of Cardiology, Cardiovascular Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Junli Li
- Laboratory of Heart Valve Disease, West China Hospital, Sichuan University, Chengdu, China
| | - Wenyu Hu
- Department of Cardiology, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Hongwei Li
- Department of Cardiology, Cardiovascular Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Metabolic Disorder Related Cardiovascular Disease, Beijing, China.,Department of Geriatrics, Cardiovascular Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
17
|
Wu D, Kampmann E, Qian G. Novel Insights Into the Role of Mitochondria-Derived Peptides in Myocardial Infarction. Front Physiol 2021; 12:750177. [PMID: 34777013 PMCID: PMC8582487 DOI: 10.3389/fphys.2021.750177] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 09/28/2021] [Indexed: 01/02/2023] Open
Abstract
Mitochondria-derived peptides (MDPs) are a new class of bioactive peptides encoded by small open reading frames (sORFs) within known mitochondrial DNA (mtDNA) genes. MDPs may affect the expression of nuclear genes and play cytoprotective roles against chronic and age-related diseases by maintaining mitochondrial function and cell viability in the face of metabolic stress and cytotoxic insults. In this review, we summarize clinical and experimental findings indicating that MDPs act as local and systemic regulators of glucose homeostasis, immune and inflammatory responses, mitochondrial function, and adaptive stress responses, and focus on evidence supporting the protective effects of MDPs against myocardial infarction. These insights into MDPs actions suggest their potential in the treatment of cardiovascular diseases and should encourage further research in this field.
Collapse
Affiliation(s)
- Dan Wu
- Department of Cardiology, The First Medical Center, Chinese People's Liberation Army Hospital, Medical School of Chinese People's Liberation Army, Beijing, China
| | - Enny Kampmann
- School of Life Sciences, City College of San Francisco, San Francisco, CA, United States
| | - Geng Qian
- Department of Cardiology, The First Medical Center, Chinese People's Liberation Army Hospital, Medical School of Chinese People's Liberation Army, Beijing, China
| |
Collapse
|
18
|
Chen L, Shi D, Guo M. The roles of PKC-δ and PKC-ε in myocardial ischemia/reperfusion injury. Pharmacol Res 2021; 170:105716. [PMID: 34102229 DOI: 10.1016/j.phrs.2021.105716] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 05/01/2021] [Accepted: 06/03/2021] [Indexed: 01/14/2023]
Abstract
Ischemia and reperfusion (I/R) cause a reduction in arterial blood supply to tissues, followed by the restoration of perfusion and consequent reoxygenation. The reestablishment of blood flow triggers further damage to ischemic tissue through reactive oxygen species (ROS) accumulation, interference with cellular ion homeostasis, opening of mitochondrial permeability transition pores (mPTPs) and promotion of cell death (apoptosis or necrosis). PKC-δ and PKC-ε, belonging to a family of serine/threonine kinases, have been demonstrated to play important roles during I/R injury in cardiovascular diseases. However, the cardioprotective mechanisms of PKC-δ and PKC-ε in I/R injury have not been elaborated until now. This article discusses the roles of PKC-δ and PKC-ε during myocardial I/R in redox regulation (redox signaling and oxidative stress), cell death (apoptosis and necrosis), Ca2+ overload, and mitochondrial dysfunction.
Collapse
Affiliation(s)
- Li Chen
- Peking University Traditional Chinese Medicine Clinical Medical School (Xi yuan), Beijing, China; National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Dazhuo Shi
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China.
| | - Ming Guo
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China.
| |
Collapse
|
19
|
Attanasio U, Pirozzi F, Poto R, Cuomo A, Carannante A, Russo M, Ghigo A, Hirsch E, Tocchetti CG, Varricchi G, Mercurio V. Oxidative stress in anticancer therapies-related cardiac dysfunction. Free Radic Biol Med 2021; 169:410-415. [PMID: 33930514 DOI: 10.1016/j.freeradbiomed.2021.04.021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Revised: 03/30/2021] [Accepted: 04/08/2021] [Indexed: 02/06/2023]
Abstract
Redox abnormalities are at the crossroad of cardiovascular diseases, cancer and cardiotoxicity from anticancer treatments. Indeed, disturbances of the redox equilibrium are common drivers of these conditions. Not only is an increase in oxidative stress a fundamental mechanism of action of anthracyclines (which have historically been the most studied anticancer treatments) but also this is at the basis of the toxic cardiovascular effects of antineoplastic targeted drugs and radiotherapy. Here we examine the oxidative mechanisms involved in the different cardiotoxicities induced by the main redox-based antineoplastic treatments, and discuss novel approaches for the treatment of such toxicities.
Collapse
Affiliation(s)
- Umberto Attanasio
- Department of Translational Medical Sciences, Federico II University, Naples, Italy
| | - Flora Pirozzi
- Department of Translational Medical Sciences, Federico II University, Naples, Italy
| | - Remo Poto
- Department of Translational Medical Sciences, Federico II University, Naples, Italy
| | - Alessandra Cuomo
- Department of Translational Medical Sciences, Federico II University, Naples, Italy
| | - Antonio Carannante
- Department of Translational Medical Sciences, Federico II University, Naples, Italy
| | - Michele Russo
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Torino, Italy
| | - Alessandra Ghigo
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Torino, Italy
| | - Emilio Hirsch
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Torino, Italy
| | - Carlo Gabriele Tocchetti
- Department of Translational Medical Sciences, Federico II University, Naples, Italy; Interdepartmental Center of Clinical and Translational Research (CIRCET), Federico II University, Naples, Italy; Interdepartmental Hypertension Research Center (CIRIAPA), Federico II University, Naples, Italy; Center for Basic and Clinical Immunology Research (CISI), Federico II University, Naples, Italy.
| | - Gilda Varricchi
- Department of Translational Medical Sciences, Federico II University, Naples, Italy; Center for Basic and Clinical Immunology Research (CISI), Federico II University, Naples, Italy; WAO Center of Excellence, Naples, Italy; Institute of Experimental Endocrinology and Oncology "G. Salvatore" (IEOS), National Research Council (CNR), Naples, Italy
| | - Valentina Mercurio
- Department of Translational Medical Sciences, Federico II University, Naples, Italy
| |
Collapse
|
20
|
Querio G, Geddo F, Antoniotti S, Gallo MP, Penna C. Sex and Response to Cardioprotective Conditioning Maneuvers. Front Physiol 2021; 12:667961. [PMID: 34054579 PMCID: PMC8160310 DOI: 10.3389/fphys.2021.667961] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Accepted: 03/30/2021] [Indexed: 12/02/2022] Open
Abstract
Ischemic heart disease (IHD) is a multifactorial pathological condition strictly related to genetic, dietary, and lifestyle factors. Its morbidity and mortality rate represent one of the most important pathological issues that today involve younger people in a stronger way than in the past. IHD clinical outcomes are difficult to treat and have a high economic impact on health care. So prevention of this pathological condition through cardioprotective maneuvers represents the first line of intervention, as already underlined by several animal and human studies. Even if the time of intervention is important to prevent severe outcomes, many studies highlight that sex-dependent responses are crucial for the result of cardioprotective procedures. In this scenario sexual hormones have revealed an important role in cardioprotective approach, as women seem to be more protected toward cardiac insults when compared to male counterparts. The aim of this mini review is to show the molecular pathways involved in cardioprotective protocols and to elucidate how sexual hormones can contribute in ameliorating or worsening the physiological responses to IHD.
Collapse
Affiliation(s)
- Giulia Querio
- Department of Life Sciences and Systems Biology, University of Turin, Turin, Italy
| | - Federica Geddo
- Department of Life Sciences and Systems Biology, University of Turin, Turin, Italy
| | - Susanna Antoniotti
- Department of Life Sciences and Systems Biology, University of Turin, Turin, Italy
| | - Maria Pia Gallo
- Department of Life Sciences and Systems Biology, University of Turin, Turin, Italy
| | - Claudia Penna
- Department of Clinical and Biological Sciences, University of Turin, Turin, Italy
| |
Collapse
|
21
|
Penna C, Femminò S, Caldera F, Rubin Pedrazzo A, Cecone C, Alfì E, Comità S, Higashiyama T, Trotta F, Pagliaro P, Cavalli R. Cyclic Nigerosyl-Nigerose as Oxygen Nanocarrier to Protect Cellular Models from Hypoxia/Reoxygenation Injury: Implications from an In Vitro Model. Int J Mol Sci 2021; 22:ijms22084208. [PMID: 33921614 PMCID: PMC8073687 DOI: 10.3390/ijms22084208] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 03/30/2021] [Accepted: 04/14/2021] [Indexed: 01/30/2023] Open
Abstract
Heart failure (HF) prevalence is increasing among the aging population, and the mortality rate remains unacceptably high despite improvements in therapy. Myocardial ischemia (MI) and, consequently, ischemia/reperfusion injury (IRI), are frequently the basis of HF development. Therefore, cardioprotective strategies to limit IRI are mandatory. Nanocarriers have been proposed as alternative therapy for cardiovascular disease. Controlled reoxygenation may be a promising strategy. Novel nanocarriers, such as cyclic nigerosyl-nigerose (CNN), can be innovative tools for oxygen delivery in a controlled manner. In this study we analyzed new CNN-based formulations as oxygen nanocarriers (O2-CNN), and compared them with nitrogen CNN (N2-CNN). These different CNN-based formulations were tested using two cellular models, namely, cardiomyoblasts (H9c2), and endothelial (HMEC) cell lines, at different concentrations. The effects on the growth curve during normoxia (21% O2, 5% CO2 and 74% N2) and their protective effects during hypoxia (1% O2, 5% CO2 and 94% N2) and reoxygenation (21% O2, 5% CO2 and 74% N2) were studied. Neither O2-CNN nor N2-CNN has any effect on the growth curve during normoxia. However, O2-CNN applied before hypoxia induces a 15–30% reduction in cell mortality after hypoxia/re-oxygenation when compared to N2-CNN. O2-CNN showed a marked efficacy in controlled oxygenation, which suggests an interesting potential for the future medical application of soluble nanocarrier systems for MI treatment.
Collapse
Affiliation(s)
- Claudia Penna
- Department of Clinical and Biological Sciences, University of Turin, 10043 Turin, Italy; (C.P.); (S.F.); (E.A.); (S.C.)
| | - Saveria Femminò
- Department of Clinical and Biological Sciences, University of Turin, 10043 Turin, Italy; (C.P.); (S.F.); (E.A.); (S.C.)
- Department of Medical Sciences, University of Turin, 10126 Turin, Italy
| | - Fabrizio Caldera
- Department of Chemistry, University of Turin, 10125 Turin, Italy; (F.C.); (A.R.P.); (C.C.)
| | - Alberto Rubin Pedrazzo
- Department of Chemistry, University of Turin, 10125 Turin, Italy; (F.C.); (A.R.P.); (C.C.)
| | - Claudio Cecone
- Department of Chemistry, University of Turin, 10125 Turin, Italy; (F.C.); (A.R.P.); (C.C.)
| | - Edoardo Alfì
- Department of Clinical and Biological Sciences, University of Turin, 10043 Turin, Italy; (C.P.); (S.F.); (E.A.); (S.C.)
| | - Stefano Comità
- Department of Clinical and Biological Sciences, University of Turin, 10043 Turin, Italy; (C.P.); (S.F.); (E.A.); (S.C.)
| | - Takanobu Higashiyama
- Hayashibara CO., LTD./Nagase Group 675-1 Fujisaki, Naka-ku, Okayama 702-8006, Japan;
| | - Francesco Trotta
- Department of Chemistry, University of Turin, 10125 Turin, Italy; (F.C.); (A.R.P.); (C.C.)
- Correspondence: (F.T.); (P.P.); (R.C.)
| | - Pasquale Pagliaro
- Department of Clinical and Biological Sciences, University of Turin, 10043 Turin, Italy; (C.P.); (S.F.); (E.A.); (S.C.)
- Correspondence: (F.T.); (P.P.); (R.C.)
| | - Roberta Cavalli
- Department of Drug Science and Technology, University of Turin, 10125 Turin, Italy
- Correspondence: (F.T.); (P.P.); (R.C.)
| |
Collapse
|
22
|
Andreadou I, Daiber A, Baxter GF, Brizzi MF, Di Lisa F, Kaludercic N, Lazou A, Varga ZV, Zuurbier CJ, Schulz R, Ferdinandy P. Influence of cardiometabolic comorbidities on myocardial function, infarction, and cardioprotection: Role of cardiac redox signaling. Free Radic Biol Med 2021; 166:33-52. [PMID: 33588049 DOI: 10.1016/j.freeradbiomed.2021.02.012] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 02/03/2021] [Accepted: 02/06/2021] [Indexed: 02/06/2023]
Abstract
The morbidity and mortality from cardiovascular diseases (CVD) remain high. Metabolic diseases such as obesity, hyperlipidemia, diabetes mellitus (DM), non-alcoholic fatty liver disease (NAFLD) and non-alcoholic steatohepatitis (NASH) as well as hypertension are the most common comorbidities in patients with CVD. These comorbidities result in increased myocardial oxidative stress, mainly from increased activity of nicotinamide adenine dinucleotide phosphate oxidases, uncoupled endothelial nitric oxide synthase, mitochondria as well as downregulation of antioxidant defense systems. Oxidative and nitrosative stress play an important role in ischemia/reperfusion injury and may account for increased susceptibility of the myocardium to infarction and myocardial dysfunction in the presence of the comorbidities. Thus, while early reperfusion represents the most favorable therapeutic strategy to prevent ischemia/reperfusion injury, redox therapeutic strategies may provide additive benefits, especially in patients with heart failure. While oxidative and nitrosative stress are harmful, controlled release of reactive oxygen species is however important for cardioprotective signaling. In this review we summarize the current data on the effect of hypertension and major cardiometabolic comorbidities such as obesity, hyperlipidemia, DM, NAFLD/NASH on cardiac redox homeostasis as well as on ischemia/reperfusion injury and cardioprotection. We also review and discuss the therapeutic interventions that may restore the redox imbalance in the diseased myocardium in the presence of these comorbidities.
Collapse
Affiliation(s)
- Ioanna Andreadou
- Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece.
| | - Andreas Daiber
- Department of Cardiology 1, Molecular Cardiology, University Medical Center, Langenbeckstr. 1, 55131, Mainz, Germany; Partner Site Rhine-Main, German Center for Cardiovascular Research (DZHK), Langenbeckstr, Germany.
| | - Gary F Baxter
- Division of Pharmacology, School of Pharmacy and Pharmaceutical Sciences, Cardiff University, United Kingdom
| | | | - Fabio Di Lisa
- Department of Biomedical Sciences, University of Padova, Italy; Neuroscience Institute, National Research Council of Italy (CNR), Padova, Italy
| | - Nina Kaludercic
- Neuroscience Institute, National Research Council of Italy (CNR), Padova, Italy
| | - Antigone Lazou
- Laboratory of Animal Physiology, School of Biology, Aristotle University of Thessaloniki, Thessaloniki, 54124, Greece
| | - Zoltán V Varga
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary; HCEMM-SU Cardiometabolic Immunology Research Group, Budapest, Hungary
| | - Coert J Zuurbier
- Laboratory of Experimental Intensive Care Anesthesiology, Department Anesthesiology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Rainer Schulz
- Institute of Physiology, Justus Liebig University Giessen, Giessen, Germany.
| | - Péter Ferdinandy
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary; Pharmahungary Group, Szeged, Hungary
| |
Collapse
|
23
|
Tsoumani M, Georgoulis A, Nikolaou PE, Kostopoulos IV, Dermintzoglou T, Papatheodorou I, Zoga A, Efentakis P, Konstantinou M, Gikas E, Kostomitsopoulos N, Papapetropoulos A, Lazou A, Skaltsounis AL, Hausenloy DJ, Tsitsilonis O, Tseti I, Di Lisa F, Iliodromitis EK, Andreadou I. Acute administration of the olive constituent, oleuropein, combined with ischemic postconditioning increases myocardial protection by modulating oxidative defense. Free Radic Biol Med 2021; 166:18-32. [PMID: 33582227 DOI: 10.1016/j.freeradbiomed.2021.02.011] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 01/22/2021] [Accepted: 02/06/2021] [Indexed: 12/13/2022]
Abstract
Oleuropein, one of the main polyphenolic constituents of olive, is cardioprotective against ischemia reperfusion injury (IRI). We aimed to assess the cardioprotection afforded by acute administration of oleuropein and to evaluate the underlying mechanism. Importantly, since antioxidant therapies have yielded inconclusive results in attenuating IRI-induced damage on top of conditioning strategies, we investigated whether oleuropein could enhance or imbed the cardioprotective manifestation of ischemic postconditioning (PostC). Oleuropein, given during ischemia as a single intravenous bolus dose reduced the infarct size compared to the control group both in rabbits and mice subjected to myocardial IRI. None of the inhibitors of the cardioprotective pathways, l-NAME, wortmannin and AG490, influence its infarct size limiting effects. Combined oleuropein and PostC cause further limitation of infarct size in comparison with PostC alone in both animal models. Oleuropein did not inhibit the calcium induced mitochondrial permeability transition pore opening in isolated mitochondria and did not increase cGMP production. To provide further insights to the different cardioprotective mechanism of oleuropein, we sought to characterize its anti-inflammatory potential in vivo. Oleuropein, PostC and their combination reduce inflammatory monocytes infiltration into the heart and the circulating monocyte cell population. Oleuropein's mechanism of action involves a direct protective effect on cardiomyocytes since it significantly increased their viability following simulated IRI as compared to non-treated cells. Οleuropein confers additive cardioprotection on top of PostC, via increasing the expression of the transcription factor Nrf-2 and its downstream targets in vivo. In conclusion, acute oleuropein administration during ischemia in combination with PostC provides robust and synergistic cardioprotection in experimental models of IRI by inducing antioxidant defense genes through Nrf-2 axis and independently of the classic cardioprotective signaling pathways (RISK, cGMP/PKG, SAFE).
Collapse
Affiliation(s)
- Maria Tsoumani
- Laboratory of Pharmacology, School of Pharmacy, National and Kapodistrian University of Athens, 15771, Athens, Greece
| | - Anastasios Georgoulis
- Laboratory of Pharmacology, School of Pharmacy, National and Kapodistrian University of Athens, 15771, Athens, Greece
| | - Panagiota-Efstathia Nikolaou
- Laboratory of Pharmacology, School of Pharmacy, National and Kapodistrian University of Athens, 15771, Athens, Greece
| | - Ioannis V Kostopoulos
- Department of Animal and Human Physiology, Faculty of Biology, National and Kapodistrian University of Athens, 15784, Athens, Greece
| | - Theano Dermintzoglou
- Laboratory of Pharmacology, School of Pharmacy, National and Kapodistrian University of Athens, 15771, Athens, Greece
| | - Ioanna Papatheodorou
- Laboratory of Animal Physiology, School of Biology, Aristotle University of Thessaloniki, Thessaloniki, 54124, Greece
| | - Anastasia Zoga
- 2nd Department of Cardiology, Attikon Hospital, Medical School, National and Kapodistrian University of Athens, 12462, Athens, Greece
| | - Panagiotis Efentakis
- Laboratory of Pharmacology, School of Pharmacy, National and Kapodistrian University of Athens, 15771, Athens, Greece
| | - Maria Konstantinou
- Laboratory of Pharmaceutical Analysis, Department of Pharmacy, School of Health Sciences, National and Kapodistrian University of Athens, 15771, Athens, Greece
| | - Evangelos Gikas
- Laboratory of Analytical Chemistry, Department of Chemistry, School of Science, National and Kapodistrian University of Athens, 15771, Athens, Greece
| | - Nikolaos Kostomitsopoulos
- Centre of Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, 11527, Αthens Greece
| | - Andreas Papapetropoulos
- Laboratory of Pharmacology, School of Pharmacy, National and Kapodistrian University of Athens, 15771, Athens, Greece; Centre of Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, 11527, Αthens Greece
| | - Antigone Lazou
- Laboratory of Animal Physiology, School of Biology, Aristotle University of Thessaloniki, Thessaloniki, 54124, Greece
| | - Alexios-Leandros Skaltsounis
- Division of Pharmacognosy and Natural Products Chemistry, School of Pharmacy, National and Kapodistrian University of Athens, 15771, Athens, Greece
| | - Derek J Hausenloy
- Cardiovascular & Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore; National Heart Research Institute Singapore, National Heart Centre, Singapore; Yong Loo Lin School of Medicine, National University Singapore, Singapore; The Hatter Cardiovascular Institute, University College London, London, UK; Cardiovascular Research Center, College of Medical and Health Sciences, Asia University, Taiwan
| | - Ourania Tsitsilonis
- Department of Animal and Human Physiology, Faculty of Biology, National and Kapodistrian University of Athens, 15784, Athens, Greece
| | | | - Fabio Di Lisa
- Department of Biomedical Sciences, Università Degli Studi di Padova, Padova, Italy
| | - Efstathios K Iliodromitis
- 2nd Department of Cardiology, Attikon Hospital, Medical School, National and Kapodistrian University of Athens, 12462, Athens, Greece
| | - Ioanna Andreadou
- Laboratory of Pharmacology, School of Pharmacy, National and Kapodistrian University of Athens, 15771, Athens, Greece.
| |
Collapse
|
24
|
He Y, Lu X, Chen T, Yang Y, Zheng J, Chen C, Zhang Y, Lei W. Resveratrol protects against myocardial ischemic injury via the inhibition of NF‑κB‑dependent inflammation and the enhancement of antioxidant defenses. Int J Mol Med 2021; 47:29. [PMID: 33537801 PMCID: PMC7895514 DOI: 10.3892/ijmm.2021.4862] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Accepted: 12/21/2020] [Indexed: 12/23/2022] Open
Abstract
Resveratrol (RES) is a natural phenol which possesses multiple pharmacological actions. The present study aimed to determine whether RES protects against myocardial ischemic injury in association with the inhibition of NF‑κB‑dependent inflammation and the enhancement of antioxidant defenses in mice following acute myocardial infarction (AMI). Male C57/BL mice were randomly assigned to 3 groups as follows: The sham‑operated (sham) group, AMI + vehicle group and AMI + RES group. Rat H9C2 cells were also used to examine the effects of RES on hypoxia‑induced oxidative injury in vitro. Redox homeostasis in the mouse myocardium and rat H9C2 cells was determined post‑treatment. The mRNA and protein levels of phosphorylated (p‑)IκB kinase (p‑IKK), p‑nuclear factor (NF)‑κB p65, interleukin (IL)‑1β, IL‑6, nerve growth factor (NGF) and insulin‑like growth factor‑1 (IGF‑1) were measured by RT‑qPCR and western blot analysis. It was found that RES slightly protected the myocardium against ischemic injury in mice, while it prevented the hypoxia‑induced apoptosis of H9C2 cells. RES decreased the production of reactive oxygen species (ROS) and enhanced the activities of superoxide dismutase (SOD), glutathione (GSH) and glutathione peroxidase (GPx). RES also downregulated the protein and/or mRNA levels of p‑IKK, p‑NF‑κB p65, IL‑1β, IL‑6, NGF and IGF‑1 at 7 and 28 days after infarction. On the whole, these data indicate that RES protects the myocardium against ischemic injury in association with the inhibition of oxidative stress and inflammatory responses. Thus, RES has the potential to be used as an adjunctive therapeutic drug for heart diseases.
Collapse
Affiliation(s)
- Yuan He
- Laboratory of Cardiovascular Diseases
| | | | | | - Yu Yang
- Gerontology Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524001, P.R. China
| | - Jing Zheng
- Department of Obstetrics and Gynecology, University of Wisconsin-Madison, Madison, WI 53715, USA
| | | | - Yuanqi Zhang
- Department of Vascular, Thyroid and Breast Surgery, Affiliated Hospital of Guangdong Medical College, Zhanjiang, Guangdong 524001, P.R. China
| | - Wei Lei
- Laboratory of Cardiovascular Diseases
- Cardiovascular Medicine Center
| |
Collapse
|
25
|
Cadeddu Dessalvi C, Deidda M, Noto A, Madeddu C, Cugusi L, Santoro C, López-Fernández T, Galderisi M, Mercuro G. Antioxidant Approach as a Cardioprotective Strategy in Chemotherapy-Induced Cardiotoxicity. Antioxid Redox Signal 2021; 34:572-588. [PMID: 32151144 DOI: 10.1089/ars.2020.8055] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Significance: Chemotherapy-induced cardiotoxicity (CTX) has been associated with redox signaling imbalance. In fact, redox reactions are crucial for normal heart physiology, whereas excessive oxidative stress can cause cardiomyocyte structural damage. Recent Advances: An antioxidant approach as a cardioprotective strategy in this setting has shown encouraging results in preventing anticancer drug-induced CTX. Critical Issues: In fact, traditional heart failure drugs as well as many other compounds and nonpharmacological strategies, with a partial effect in reducing oxidative stress, have been shown to counterbalance chemotherapy-induced CTX in this setting to some extent. Future Directions: Given the various pathways of toxicity involved in different chemotherapeutic schemes, interactions with redox balance need to be fine-tuned and a personalized cardioprotective approach seems to be required.
Collapse
Affiliation(s)
| | - Martino Deidda
- Department of Medical Sciences and Public Health, University of Cagliari, Cagliari, Italy
| | - Antonio Noto
- Department of Medical Sciences and Public Health, University of Cagliari, Cagliari, Italy
| | - Clelia Madeddu
- Department of Medical Sciences and Public Health, University of Cagliari, Cagliari, Italy
| | - Lucia Cugusi
- Department of Medical Sciences and Public Health, University of Cagliari, Cagliari, Italy
| | - Ciro Santoro
- Department of Advanced Biomedical Sciences, Federico II University, Naples, Italy
| | - Teresa López-Fernández
- Cardiology Service, Cardio-Oncology Unit, La Paz University Hospital, IdiPAz Research Institute, Ciber CV, Madrid, Spain
| | - Maurizio Galderisi
- Department of Advanced Biomedical Sciences, Federico II University, Naples, Italy
| | - Giuseppe Mercuro
- Department of Medical Sciences and Public Health, University of Cagliari, Cagliari, Italy
| |
Collapse
|
26
|
Daiber A, Andreadou I, Oelze M, Davidson SM, Hausenloy DJ. Discovery of new therapeutic redox targets for cardioprotection against ischemia/reperfusion injury and heart failure. Free Radic Biol Med 2021; 163:325-343. [PMID: 33359685 DOI: 10.1016/j.freeradbiomed.2020.12.026] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2020] [Revised: 12/10/2020] [Accepted: 12/16/2020] [Indexed: 02/06/2023]
Abstract
Global epidemiological studies reported a shift from maternal/infectious communicable diseases to chronic non-communicable diseases and a major part is attributable to atherosclerosis and metabolic disorders. Accordingly, ischemic heart disease was identified as a leading risk factor for global mortality and morbidity with a prevalence of 128 million people. Almost 9 million premature deaths can be attributed to ischemic heart disease and subsequent acute myocardial infarction and heart failure, also representing a substantial socioeconomic burden. As evidenced by typical oxidative stress markers such as lipid peroxidation products or oxidized DNA/RNA bases, the formation of reactive oxygen species by various sources (NADPH oxidases, xanthine oxidase and mitochondrial resperatory chain) plays a central role for the severity of ischemia/reperfusion damage. The underlying mechanisms comprise direct oxidative damage but also adverse redox-regulation of kinase and calcium signaling, inflammation and cardiac remodeling among others. These processes and the role of reactive oxygen species are discussed in the present review. We also present and discuss potential targets for redox-based therapies that are either already established in the clinics (e.g. guanylyl cyclase activators and stimulators) or at least successfully tested in preclinical models of myocardial infarction and heart failure (mitochondria-targeted antioxidants). However, reactive oxygen species have not only detrimental effects but are also involved in essential cellular signaling and may even act protective as seen by ischemic pre- and post-conditioning or eustress - which makes redox therapy quite challenging.
Collapse
Affiliation(s)
- Andreas Daiber
- Department of Cardiology 1, Molecular Cardiology, University Medical Center, Langenbeckstr. 1, 55131, Mainz, Germany; Partner Site Rhine-Main, German Center for Cardiovascular Research (DZHK), Langenbeckstr. 1, 55131, Mainz, Germany.
| | - Ioanna Andreadou
- Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, 15771, Athens, Greece
| | - Matthias Oelze
- Department of Cardiology 1, Molecular Cardiology, University Medical Center, Langenbeckstr. 1, 55131, Mainz, Germany
| | - Sean M Davidson
- The Hatter Cardiovascular Institute, 67 Chenies Mews, London, WC1E 6HX, United Kingdom
| | - Derek J Hausenloy
- The Hatter Cardiovascular Institute, 67 Chenies Mews, London, WC1E 6HX, United Kingdom; Cardiovascular & Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore; National Heart Research Institute Singapore, National Heart Centre, Singapore; Yong Loo Lin School of Medicine, National University Singapore, Singapore; Cardiovascular Research Center, College of Medical and Health Sciences, Asia University, Taiwan.
| |
Collapse
|
27
|
Li YQ, Jiao Y, Liu YN, Fu JY, Sun LK, Su J. PGC-1α protects from myocardial ischaemia-reperfusion injury by regulating mitonuclear communication. J Cell Mol Med 2021; 26:593-600. [PMID: 33470050 PMCID: PMC8817131 DOI: 10.1111/jcmm.16236] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 11/25/2020] [Accepted: 12/15/2020] [Indexed: 12/17/2022] Open
Abstract
The recovery of blood supply after a period of myocardial ischaemia does not restore the heart function and instead results in a serious dysfunction called myocardial ischaemia‐reperfusion injury (IRI), which involves several complex pathophysiological processes. Mitochondria have a wide range of functions in maintaining the cellular energy supply, cell signalling and programmed cell death. When mitochondrial function is insufficient or disordered, it may have adverse effects on myocardial ischaemia‐reperfusion and therefore mitochondrial dysfunction caused by oxidative stress a core molecular mechanism of IRI. Peroxisome proliferator‐activated receptor gamma co‐activator 1α (PGC‐1α) is an important antioxidant molecule found in mitochondria. However, its role in IRI has not yet been systematically summarized. In this review, we speculate the role of PGC‐1α as a key regulator of mitonuclear communication, which may interacts with nuclear factor, erythroid 2 like ‐1 and ‐2 (NRF‐1/2) to inhibit mitochondrial oxidative stress, promote the clearance of damaged mitochondria, enhance mitochondrial biogenesis, and reduce the burden of IRI.
Collapse
Affiliation(s)
- Yan-Qing Li
- Department of Pathophysiology, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Yan Jiao
- Department of Hepatobiliary and Pancreatic Surgery, The First Hospital of Jilin University, Changchun, China
| | - Ya-Nan Liu
- Department of Pathophysiology, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Jia-Ying Fu
- Department of Pathophysiology, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Lian-Kun Sun
- Department of Pathophysiology, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Jing Su
- Department of Pathophysiology, College of Basic Medical Sciences, Jilin University, Changchun, China
| |
Collapse
|
28
|
The Calcilytic Drug Calhex-231 Ameliorates Vascular Hyporesponsiveness in Traumatic Hemorrhagic Shock by Inhibiting Oxidative Stress and miR-208a-Mediated Mitochondrial Fission. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:4132785. [PMID: 33343806 PMCID: PMC7732383 DOI: 10.1155/2020/4132785] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 10/27/2020] [Accepted: 11/26/2020] [Indexed: 12/29/2022]
Abstract
Background The calcium-sensing receptor (CaSR) plays a fundamental role in extracellular calcium homeostasis in humans. Surprisingly, CaSR is also expressed in nonhomeostatic tissues and is involved in regulating diverse cellular functions. The objective of this study was to determine if Calhex-231 (Cal), a negative modulator of CaSR, may be beneficial in the treatment of traumatic hemorrhagic shock (THS) by improving cardiovascular function and investigated the mechanisms. Methods Rats that had been subjected to THS and hypoxia-treated vascular smooth muscle cells (VSMCs) were used in this study. The effects of Cal on cardiovascular function, animal survival, hemodynamics, and vital organ function in THS rats and the relationship to oxidative stress, mitochondrial fusion-fission, and microRNA (miR-208a) were investigated. Results Cal significantly improved hemodynamics, elevated blood pressure, increased vital organ blood perfusion and local oxygen supply, and markedly improved the survival outcomes of THS rats. Furthermore, Cal significantly improved vascular reactivity after THS in vivo and in vitro. Cal also restored the THS-induced decrease in myosin light chain (MLC) phosphorylation (the key element for VSMC contraction). Inhibition of MLC phosphorylation antagonized the Cal-induced restoration of vascular reactivity following THS. Cal suppressed oxidative stress in THS rats and hypoxic-VSMCs. Meanwhile, THS induced expression of mitochondrial fission proteins Drp1 and Fis1 and decreased expression of mitochondrial fusion protein Mfn1 in vascular tissues. Cal reduced expression of Drp1 and Fis1. In hypoxic-VSMCs, Cal inhibited mitochondrial fragmentation and preserved mitochondrial morphology. In addition, miR-208a mimic decreased Fis1 expression, and miR-208a inhibitor prevented Cal-induced Fis1 downregulation in hypoxic-VSMCs. Conclusion Calhex-231 exhibits outstanding potential for effective therapy of traumatic hemorrhagic shock, and the beneficial effects result from its protection of vascular function via inhibition of oxidative stress and miR-208a-mediated mitochondrial fission.
Collapse
|
29
|
Penna C, Andreadou I, Aragno M, Beauloye C, Bertrand L, Lazou A, Falcão‐Pires I, Bell R, Zuurbier CJ, Pagliaro P, Hausenloy DJ. Effect of hyperglycaemia and diabetes on acute myocardial ischaemia-reperfusion injury and cardioprotection by ischaemic conditioning protocols. Br J Pharmacol 2020; 177:5312-5335. [PMID: 31985828 PMCID: PMC7680002 DOI: 10.1111/bph.14993] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2019] [Revised: 11/19/2019] [Accepted: 01/09/2020] [Indexed: 12/12/2022] Open
Abstract
Diabetic patients are at increased risk of developing coronary artery disease and experience worse clinical outcomes following acute myocardial infarction. Novel therapeutic strategies are required to protect the myocardium against the effects of acute ischaemia-reperfusion injury (IRI). These include one or more brief cycles of non-lethal ischaemia and reperfusion prior to the ischaemic event (ischaemic preconditioning [IPC]) or at the onset of reperfusion (ischaemic postconditioning [IPost]) either to the heart or to extracardiac organs (remote ischaemic conditioning [RIC]). Studies suggest that the diabetic heart is resistant to cardioprotective strategies, although clinical evidence is lacking. We overview the available animal models of diabetes, investigating acute myocardial IRI and cardioprotection, experiments investigating the effects of hyperglycaemia on susceptibility to acute myocardial IRI, the response of the diabetic heart to cardioprotective strategies e.g. IPC, IPost and RIC. Finally we highlight the effects of anti-hyperglycaemic agents on susceptibility to acute myocardial IRI and cardioprotection. LINKED ARTICLES: This article is part of a themed issue on Risk factors, comorbidities, and comedications in cardioprotection. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v177.23/issuetoc.
Collapse
Affiliation(s)
- Claudia Penna
- Department of Clinical and Biological SciencesUniversity of TurinTurinItaly
| | - Ioanna Andreadou
- Laboratory of Pharmacology, Faculty of PharmacyNational and Kapodistrian University of AthensAthensGreece
| | - Manuela Aragno
- Department of Clinical and Biological SciencesUniversity of TurinTurinItaly
| | | | - Luc Bertrand
- Division of CardiologyCliniques Universitaires Saint‐LucBrusselsBelgium
- Pole of Cardiovascular Research, Institut de Recherche Experimetnale et CliniqueUCLouvainBrusselsBelgium
| | - Antigone Lazou
- School of BiologyAristotle University of ThessalonikiThessalonikiGreece
| | - Ines Falcão‐Pires
- Unidade de Investigação Cardiovascular, Departamento de Cirurgia e Fisiologia, Faculdade de MedicinaUniversidade do PortoPortoPortugal
| | - Robert Bell
- The Hatter Cardiovascular InstituteUniversity College LondonLondonUK
| | - Coert J. Zuurbier
- Laboratory of Experimental Intensive Care and Anesthesiology (L.E.I.C.A.), Department of Anesthesiology, Amsterdam UMCUniversity of Amsterdam, Cardiovascular SciencesAmsterdamThe Netherlands
| | - Pasquale Pagliaro
- Department of Clinical and Biological SciencesUniversity of TurinTurinItaly
| | - Derek J. Hausenloy
- The Hatter Cardiovascular InstituteUniversity College LondonLondonUK
- Cardiovascular and Metabolic Disorders ProgramDuke–NUS Medical SchoolSingapore
- National Heart Research Institute SingaporeNational Heart Centre SingaporeSingapore
- Yong Loo Lin School of MedicineNational University of SingaporeSingapore
- Cardiovascular Research Center, College of Medical and Health SciencesAsia UniversityTaiwan
| |
Collapse
|
30
|
Gouda E, Babiker F. Micronized flavonoid fraction Daflon 500 protects heart against ischemia–reperfusion injury: an old medicine for a new target. ALL LIFE 2020. [DOI: 10.1080/26895293.2020.1832921] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
Affiliation(s)
- Eman Gouda
- Department of Physiology, Faculty of Medicine, Health Science Center, Kuwait University, Kuwait, Kuwait
| | - Fawzi Babiker
- Department of Physiology, Faculty of Medicine, Health Science Center, Kuwait University, Kuwait, Kuwait
| |
Collapse
|
31
|
Mercurio V, Cuomo A, Cadeddu Dessalvi C, Deidda M, Di Lisi D, Novo G, Manganaro R, Zito C, Santoro C, Ameri P, Spallarossa P, Arboscello E, Tocchetti CG, Penna C. Redox Imbalances in Ageing and Metabolic Alterations: Implications in Cancer and Cardiac Diseases. An Overview from the Working Group of Cardiotoxicity and Cardioprotection of the Italian Society of Cardiology (SIC). Antioxidants (Basel) 2020; 9:E641. [PMID: 32708201 PMCID: PMC7402085 DOI: 10.3390/antiox9070641] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 07/14/2020] [Accepted: 07/16/2020] [Indexed: 12/14/2022] Open
Abstract
Metabolic syndrome (MetS) is a well established risk factor for cardiovascular (CV) diseases. In addition, several studies indicate that MetS correlates with the increased risk of cancer in adults. The mechanisms linking MetS and cancer are not fully understood. Several risk factors involved in MetS are also cancer risk factors, such as the consumption of high calorie-food or high fat intake, low fibre intake, and sedentary lifestyle. Other common aspects of both cancer and MetS are oxidative stress and inflammation. In addition, some anticancer treatments can induce cardiotoxicity, including, for instance, left ventricular (LV) dysfunction and heart failure (HF), endothelial dysfunction and hypertension. In this review, we analyse several aspects of MetS, cancer and cardiotoxicity from anticancer drugs. In particular, we focus on oxidative stress in ageing, cancer and CV diseases, and we analyse the connections among CV risk factors, cancer and cardiotoxicity from anticancer drugs.
Collapse
Affiliation(s)
- Valentina Mercurio
- Department of Translational Medical Sciences, Federico II University, 80131 Naples, Italy; (V.M.); (A.C.)
| | - Alessandra Cuomo
- Department of Translational Medical Sciences, Federico II University, 80131 Naples, Italy; (V.M.); (A.C.)
| | - Christian Cadeddu Dessalvi
- Department of Medical Sciences and Public Health, University of Cagliari, 09042 Cagliari, Italy; (C.C.D.); (M.D.)
| | - Martino Deidda
- Department of Medical Sciences and Public Health, University of Cagliari, 09042 Cagliari, Italy; (C.C.D.); (M.D.)
| | - Daniela Di Lisi
- Cardiology Unit AUOP Policlinico, Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, University of Palermo, 90127 Palermo, Italy; (D.D.L.); (G.N.)
| | - Giuseppina Novo
- Cardiology Unit AUOP Policlinico, Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, University of Palermo, 90127 Palermo, Italy; (D.D.L.); (G.N.)
| | - Roberta Manganaro
- Cardiology with Coronary Intensive Care Unit, Department of Clinical and Experimental Medicine, University Hospital Policlinico “G. Martino”, University of Messina, 98124 Messina, Italy; (R.M.); (C.Z.)
| | - Concetta Zito
- Cardiology with Coronary Intensive Care Unit, Department of Clinical and Experimental Medicine, University Hospital Policlinico “G. Martino”, University of Messina, 98124 Messina, Italy; (R.M.); (C.Z.)
| | - Ciro Santoro
- Department of Advanced Biomedical Sciences, Federico II University, 80131 Naples, Italy;
| | - Pietro Ameri
- Cardiovascular Disease Unit, IRCCS Ospedale Policlinico San Martino, Genova, Italy—IRCCS Italian Cardiovascular Network & Department of Internal Medicine, University of Genova, 16121 Genova, Italy; (P.A.); (P.S.); (E.A.)
| | - Paolo Spallarossa
- Cardiovascular Disease Unit, IRCCS Ospedale Policlinico San Martino, Genova, Italy—IRCCS Italian Cardiovascular Network & Department of Internal Medicine, University of Genova, 16121 Genova, Italy; (P.A.); (P.S.); (E.A.)
| | - Eleonora Arboscello
- Cardiovascular Disease Unit, IRCCS Ospedale Policlinico San Martino, Genova, Italy—IRCCS Italian Cardiovascular Network & Department of Internal Medicine, University of Genova, 16121 Genova, Italy; (P.A.); (P.S.); (E.A.)
| | - Carlo Gabriele Tocchetti
- Department of Translational Medical Sciences, Federico II University, 80131 Naples, Italy; (V.M.); (A.C.)
- Interdepartmental Center of Clinical and Translational Sciences, Federico II University, 80131 Naples, Italy
| | - Claudia Penna
- Department of Clinical and Biological Sciences, University of Turin, 10043 Torino, Italy
| |
Collapse
|
32
|
Selvaraju V, Thirunavukkarasu M, Joshi M, Oriowo B, Shaikh IA, Rishi MT, Tapias L, Coca-Soliz V, Saad I, Campbell J, Pradeep SR, Swaminathan S, Yee SP, McFadden DW, Alexander Palesty J, Maulik N. Deletion of newly described pro-survival molecule Pellino-1 increases oxidative stress, downregulates cIAP2/NF-κB cell survival pathway, reduces angiogenic response, and thereby aggravates tissue function in mouse ischemic models. Basic Res Cardiol 2020; 115:45. [DOI: 10.1007/s00395-020-0804-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Accepted: 06/03/2020] [Indexed: 12/16/2022]
|
33
|
Diabetic Cardiomyopathy and Ischemic Heart Disease: Prevention and Therapy by Exercise and Conditioning. Int J Mol Sci 2020; 21:ijms21082896. [PMID: 32326182 PMCID: PMC7215312 DOI: 10.3390/ijms21082896] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Revised: 04/14/2020] [Accepted: 04/18/2020] [Indexed: 02/06/2023] Open
Abstract
Metabolic syndrome, diabetes, and ischemic heart disease are among the leading causes of death and disability in Western countries. Diabetic cardiomyopathy is responsible for the most severe signs and symptoms. An important strategy for reducing the incidence of cardiovascular disease is regular exercise. Remote ischemic conditioning has some similarity with exercise and can be induced by short periods of ischemia and reperfusion of a limb, and it can be performed in people who cannot exercise. There is abundant evidence that exercise is beneficial in diabetes and ischemic heart disease, but there is a need to elucidate the specific cardiovascular effects of emerging and unconventional forms of exercise in people with diabetes. In addition, remote ischemic conditioning may be considered among the options to induce beneficial effects in these patients. The characteristics and interactions of diabetes and ischemic heart disease, and the known effects of exercise and remote ischemic conditioning in the presence of metabolic syndrome and diabetes, are analyzed in this brief review.
Collapse
|
34
|
Boardman NT, Falck AT, Lund T, Chu X, Martin-Armas M, Norvik JV, Jenssen TG, Ytrehus K. Human concentrations of uric acid scavenges adaptive and maladaptive reactive oxygen species in isolated rat hearts subjected to ischemic stress. Can J Physiol Pharmacol 2019; 98:139-146. [PMID: 31518503 DOI: 10.1139/cjpp-2019-0024] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Uric acid is a purine degradation product but also an important antioxidant and reactive oxygen species (ROS) scavenger. Experimental settings that mimic myocardial ischemia-reperfusion have not included uric acid despite that it is always present in human extracellular fluid and plasma. We hypothesized that uric acid has an important role in myocardial ROS scavenging. Here, we tested the cardiac response to uric acid on infarct size following ischemia-reperfusion with and without exacerbated oxidative stress due to acute pressure overload and during preconditioning. We also examined mitochondrial respiration and ROS-induced mitochondrial permeability transition pore opening. Under exacerbated ROS stress induced by high-pressure perfusion, uric acid lowered oxidative stress and reduced infarct size. In contrast, uric acid blocked cardioprotection induced by ischemic preconditioning. However, this effect was reversed by probenecid, an inhibitor of cellular uptake of uric acid. In accordance, in intact cardiomyocytes, extracellular uric acid reduced the susceptibility of mitochondria towards opening of the permeability transition pore, suggesting that uric acid may prevent ischemia-reperfusion injury due to scavenging of maladaptive ROS. Moreover, as uric acid also scavenges adaptive ROS, this may interfere with preconditioning. Altogether, uric acid might be a confounder when translating preclinical experimental results into clinical treatment.
Collapse
Affiliation(s)
- Neoma T Boardman
- Cardiovascular Research Group, Department of Medical Biology, UiT-Arctic University of Norway, Tromsø, Norway
| | - Aleksander Tank Falck
- Cardiovascular Research Group, Department of Medical Biology, UiT-Arctic University of Norway, Tromsø, Norway
| | - Trine Lund
- Cardiovascular Research Group, Department of Medical Biology, UiT-Arctic University of Norway, Tromsø, Norway
| | - Xi Chu
- Cardiovascular Research Group, Department of Medical Biology, UiT-Arctic University of Norway, Tromsø, Norway
| | - Montserrat Martin-Armas
- Cardiovascular Research Group, Department of Medical Biology, UiT-Arctic University of Norway, Tromsø, Norway
| | - Jon V Norvik
- Cardiovascular Research Group, Department of Medical Biology, UiT-Arctic University of Norway, Tromsø, Norway.,Metabolic and Renal Research Group, Department of Clinical Medicine, UiT-Arctic University of Norway, Tromsø, Norway
| | - Trond G Jenssen
- Metabolic and Renal Research Group, Department of Clinical Medicine, UiT-Arctic University of Norway, Tromsø, Norway.,Department of Organ Transplantation, Section of Nephrology, Oslo University Hospital, Oslo, Norway
| | - Kirsti Ytrehus
- Cardiovascular Research Group, Department of Medical Biology, UiT-Arctic University of Norway, Tromsø, Norway
| |
Collapse
|
35
|
Tocchetti CG, Cadeddu C, Di Lisi D, Femminò S, Madonna R, Mele D, Monte I, Novo G, Penna C, Pepe A, Spallarossa P, Varricchi G, Zito C, Pagliaro P, Mercuro G. From Molecular Mechanisms to Clinical Management of Antineoplastic Drug-Induced Cardiovascular Toxicity: A Translational Overview. Antioxid Redox Signal 2019; 30:2110-2153. [PMID: 28398124 PMCID: PMC6529857 DOI: 10.1089/ars.2016.6930] [Citation(s) in RCA: 83] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Significance: Antineoplastic therapies have significantly improved the prognosis of oncology patients. However, these treatments can bring to a higher incidence of side-effects, including the worrying cardiovascular toxicity (CTX). Recent Advances: Substantial evidence indicates multiple mechanisms of CTX, with redox mechanisms playing a key role. Recent data singled out mitochondria as key targets for antineoplastic drug-induced CTX; understanding the underlying mechanisms is, therefore, crucial for effective cardioprotection, without compromising the efficacy of anti-cancer treatments. Critical Issues: CTX can occur within a few days or many years after treatment. Type I CTX is associated with irreversible cardiac cell injury, and it is typically caused by anthracyclines and traditional chemotherapeutics. Type II CTX is generally caused by novel biologics and more targeted drugs, and it is associated with reversible myocardial dysfunction. Therefore, patients undergoing anti-cancer treatments should be closely monitored, and patients at risk of CTX should be identified before beginning treatment to reduce CTX-related morbidity. Future Directions: Genetic profiling of clinical risk factors and an integrated approach using molecular, imaging, and clinical data may allow the recognition of patients who are at a high risk of developing chemotherapy-related CTX, and it may suggest methodologies to limit damage in a wider range of patients. The involvement of redox mechanisms in cancer biology and anticancer treatments is a very active field of research. Further investigations will be necessary to uncover the hallmarks of cancer from a redox perspective and to develop more efficacious antineoplastic therapies that also spare the cardiovascular system.
Collapse
Affiliation(s)
| | - Christian Cadeddu
- 2 Department of Medical Sciences and Public Health, University of Cagliari, Cagliari, Italy
| | - Daniela Di Lisi
- 3 Biomedical Department of Internal Medicine, University of Palermo, Palermo, Italy
| | - Saveria Femminò
- 4 Department of Clinical and Biological Sciences, University of Turin, Turin, Italy
| | - Rosalinda Madonna
- 5 Center of Aging Sciences and Translational Medicine - CESI-MeT, "G. d'Annunzio" University, Chieti, Italy.,6 Department of Internal Medicine, The Texas Heart Institute and Center for Cardiovascular Biology and Atherosclerosis Research, The University of Texas Health Science Center at Houston, Houston, Texas
| | - Donato Mele
- 7 Cardiology Unit, Emergency Department, University Hospital of Ferrara, Ferrara, Italy
| | - Ines Monte
- 8 Department of General Surgery and Medical-Surgery Specialities, University of Catania, Catania, Italy
| | - Giuseppina Novo
- 3 Biomedical Department of Internal Medicine, University of Palermo, Palermo, Italy
| | - Claudia Penna
- 4 Department of Clinical and Biological Sciences, University of Turin, Turin, Italy
| | - Alessia Pepe
- 9 U.O.C. Magnetic Resonance Imaging, Fondazione Toscana G. Monasterio C.N.R., Pisa, Italy
| | - Paolo Spallarossa
- 10 Clinic of Cardiovascular Diseases, IRCCS San Martino IST, Genova, Italy
| | - Gilda Varricchi
- 1 Department of Translational Medical Sciences, Federico II University, Naples, Italy.,11 Center for Basic and Clinical Immunology Research (CISI) - Federico II University, Naples, Italy
| | - Concetta Zito
- 12 Division of Cardiology, Clinical and Experimental Department of Medicine and Pharmacology, Policlinico "G. Martino" University of Messina, Messina, Italy
| | - Pasquale Pagliaro
- 4 Department of Clinical and Biological Sciences, University of Turin, Turin, Italy
| | - Giuseppe Mercuro
- 2 Department of Medical Sciences and Public Health, University of Cagliari, Cagliari, Italy
| |
Collapse
|
36
|
Cristallini C, Vaccari G, Barbani N, Cibrario Rocchietti E, Barberis R, Falzone M, Cabiale K, Perona G, Bellotti E, Rastaldo R, Pascale S, Pagliaro P, Giachino C. Cardioprotection of PLGA/gelatine cardiac patches functionalised with adenosine in a large animal model of ischaemia and reperfusion injury: A feasibility study. J Tissue Eng Regen Med 2019; 13:1253-1264. [PMID: 31050859 PMCID: PMC6771506 DOI: 10.1002/term.2875] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Revised: 03/21/2019] [Accepted: 04/29/2019] [Indexed: 01/13/2023]
Abstract
The protection from ischaemia‐reperfusion‐associated myocardial infarction worsening remains a big challenge. We produced a bioartificial 3D cardiac patch with cardioinductive properties on stem cells. Its multilayer structure was functionalised with clinically relevant doses of adenosine. We report here the first study on the potential of these cardiac patches in the controlled delivery of adenosine into the in vivo ischaemic‐reperfused pig heart. A Fourier transform infrared chemical imaging approach allowed us to perform a characterisation, complementary to the histological and biochemical analyses on myocardial samples after in vivo patch implantation, increasing the number of investigations and results on the restricted number of pigs (n = 4) employed in this feasibility step. In vitro tests suggested that adenosine was completely released by a functionalised patch, a data that was confirmed in vivo after 24 hr from patch implantation. Moreover, the adenosine‐loaded patch enabled a targeted delivery of the drug to the ischaemic‐reperfused area of the heart, as highlighted by the activation of the pro‐survival signalling reperfusion injury salvage kinases pathway. At 3 months, though limited to one animal, the used methods provided a picture of a tissue in dynamic conditions, associated to the biosynthesis of new collagen and to a non‐fibrotic outcome of the healing process underway. The synergistic effect between the functionalised 3D cardiac patch and adenosine cardioprotection might represent a promising innovation in the treatment of reperfusion injury. As this is a feasibility study, the clinical implications of our findings will require further in vivo investigation on larger numbers of ischaemic‐reperfused pig hearts.
Collapse
Affiliation(s)
| | | | - Niccoletta Barbani
- Department of Civil and Industrial Engineering, University of Pisa, Pisa, Italy
| | | | | | | | | | - Giovanni Perona
- Department of Veterinary Science, University of Turin, Turin, Italy
| | - Elena Bellotti
- Department of Civil and Industrial Engineering, University of Pisa, Pisa, Italy
| | - Raffaella Rastaldo
- Department of Clinical and Biological Sciences, University of Turin, Turin, Italy
| | | | - Pasquale Pagliaro
- Department of Clinical and Biological Sciences, University of Turin, Turin, Italy
| | - Claudia Giachino
- Department of Clinical and Biological Sciences, University of Turin, Turin, Italy
| |
Collapse
|
37
|
Antioxidative Property and Molecular Mechanisms Underlying Geniposide-Mediated Therapeutic Effects in Diabetes Mellitus and Cardiovascular Disease. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:7480512. [PMID: 31089416 PMCID: PMC6476013 DOI: 10.1155/2019/7480512] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/23/2018] [Revised: 02/07/2019] [Accepted: 02/13/2019] [Indexed: 02/07/2023]
Abstract
Geniposide, an iridoid glucoside, is a major component in the fruit of Gardenia jasminoides Ellis (Gardenia fruits). Geniposide has been experimentally proved to possess multiple pharmacological actions involving antioxidative stress, anti-inflammatory, antiapoptosis, antiangiogenesis, antiendoplasmic reticulum stress (ERS), etc. In vitro and in vivo studies have further identified the value of geniposide in a spectrum of preclinical models of diabetes mellitus (DM) and cardiovascular disorders. The antioxidative property of geniposide should be attributed to the result of either the inhibition of numerous pathological processes or the activation of various proteins associated with cell survival or a combination of both. In this review, we will summarize the available knowledge on the antioxidative property and protective effects of geniposide in DM and cardiovascular disease in the literature and discuss antioxidant mechanisms as well as its potential applications in clinic.
Collapse
|
38
|
Meijles DN, Zoumpoulidou G, Markou T, Rostron KA, Patel R, Lay K, Handa BS, Wong B, Sugden PH, Clerk A. The cardiomyocyte "redox rheostat": Redox signalling via the AMPK-mTOR axis and regulation of gene and protein expression balancing survival and death. J Mol Cell Cardiol 2019; 129:118-129. [PMID: 30771309 PMCID: PMC6497135 DOI: 10.1016/j.yjmcc.2019.02.006] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Revised: 01/16/2019] [Accepted: 02/12/2019] [Indexed: 02/08/2023]
Abstract
Reactive oxygen species (ROS) play a key role in development of heart failure but, at a cellular level, their effects range from cytoprotection to induction of cell death. Understanding how this is regulated is crucial to develop novel strategies to ameliorate only the detrimental effects. Here, we revisited the fundamental hypothesis that the level of ROS per se is a key factor in the cellular response by applying different concentrations of H2O2 to cardiomyocytes. High concentrations rapidly reduced intracellular ATP and inhibited protein synthesis. This was associated with activation of AMPK which phosphorylated and inhibited Raptor, a crucial component of mTOR complex-1 that regulates protein synthesis. Inhibition of protein synthesis by high concentrations of H2O2 prevents synthesis of immediate early gene products required for downstream gene expression, and such mRNAs (many encoding proteins required to deal with oxidant stress) were only induced by lower concentrations. Lower concentrations of H2O2 promoted mTOR phosphorylation, associated with differential recruitment of some mRNAs to the polysomes for translation. Some of the upregulated genes induced by low H2O2 levels are cytoprotective. We identified p21Cip1/WAF1 as one such protein, and preventing its upregulation enhanced the rate of cardiomyocyte apoptosis. The data support the concept of a "redox rheostat" in which different degrees of ROS influence cell energetics and intracellular signalling pathways to regulate mRNA and protein expression. This sliding scale determines cell fate, modulating survival vs death.
Collapse
Affiliation(s)
- Daniel N Meijles
- Molecular and Clinical Sciences Research Institute, St George's University of London, London SW17 0RE, UK; School of Biological Sciences, University of Reading, Whiteknights, Reading RG6 6AS, UK.
| | - Georgia Zoumpoulidou
- National Heart and Lung Institute (Cardiovascular Sciences), Faculty of Medicine, Flowers Building, Imperial College, SW7 2AZ, UK and Dovehouse Street, London SW3 6LY, UK
| | - Thomais Markou
- School of Biological Sciences, University of Reading, Whiteknights, Reading RG6 6AS, UK; National Heart and Lung Institute (Cardiovascular Sciences), Faculty of Medicine, Flowers Building, Imperial College, SW7 2AZ, UK and Dovehouse Street, London SW3 6LY, UK
| | - Kerry A Rostron
- School of Biological Sciences, University of Reading, Whiteknights, Reading RG6 6AS, UK
| | - Rishi Patel
- National Heart and Lung Institute (Cardiovascular Sciences), Faculty of Medicine, Flowers Building, Imperial College, SW7 2AZ, UK and Dovehouse Street, London SW3 6LY, UK
| | - Kenneth Lay
- National Heart and Lung Institute (Cardiovascular Sciences), Faculty of Medicine, Flowers Building, Imperial College, SW7 2AZ, UK and Dovehouse Street, London SW3 6LY, UK
| | - Balvinder S Handa
- National Heart and Lung Institute (Cardiovascular Sciences), Faculty of Medicine, Flowers Building, Imperial College, SW7 2AZ, UK and Dovehouse Street, London SW3 6LY, UK
| | - Bethany Wong
- National Heart and Lung Institute (Cardiovascular Sciences), Faculty of Medicine, Flowers Building, Imperial College, SW7 2AZ, UK and Dovehouse Street, London SW3 6LY, UK
| | - Peter H Sugden
- School of Biological Sciences, University of Reading, Whiteknights, Reading RG6 6AS, UK; National Heart and Lung Institute (Cardiovascular Sciences), Faculty of Medicine, Flowers Building, Imperial College, SW7 2AZ, UK and Dovehouse Street, London SW3 6LY, UK
| | - Angela Clerk
- School of Biological Sciences, University of Reading, Whiteknights, Reading RG6 6AS, UK; National Heart and Lung Institute (Cardiovascular Sciences), Faculty of Medicine, Flowers Building, Imperial College, SW7 2AZ, UK and Dovehouse Street, London SW3 6LY, UK
| |
Collapse
|
39
|
Schröder K. NADPH oxidase-derived reactive oxygen species: Dosis facit venenum. Exp Physiol 2019; 104:447-452. [PMID: 30737851 PMCID: PMC6593456 DOI: 10.1113/ep087125] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Accepted: 02/07/2019] [Indexed: 01/24/2023]
Abstract
New Findings What is the topic of this review? Within this review, the role of reactive oxygen species in cellular homeostasis, physiology and pathophysiology is discussed.
What advances does it highlight? The review provides new concepts of how reactive oxygen species influence gene expression, energy consumption and other aspects of the life of a cell. Furthermore, a model is provided to illustrate how reactive oxygen species elicit specific oxidation of target molecules.
Abstract Reactive oxygen species (ROS) have a long history of bad reputation. They are needed and effective in host defense, but on the contrary may induce situations of oxidative stress. Besides that, within recent years several soft functions (functions that may occur and are not directly connected to an effect, but may influence signaling in an indirect manner) of NADPH oxidases have been discovered, which are slowly eroding the image of the solely dangerous ROS. NADPH oxidase‐derived ROS serve to ease or enable signal transduction and to maintain homeostasis. However, there is still an enormous lag in the knowledge concerning target proteins and how ROS can elicit specific signalling in different cells and tissues. The present review summarizes some important functions of Nox2 and Nox4. Furthermore, although highly speculative, a model is provided of how those NADPH oxidases might be able to oxidize target proteins in a specific way. Many concepts mentioned in this review represent my personal view and are supported only in part by published studies.
Collapse
Affiliation(s)
- Katrin Schröder
- Institute for Cardiovascular Physiology, Goethe-University, Frankfurt, Germany.,German Center for Cardiovascular Research (DZHK), Partner site RheinMain, Frankfurt, Germany
| |
Collapse
|
40
|
Russo I, Femminò S, Barale C, Tullio F, Geuna S, Cavalot F, Pagliaro P, Penna C. Cardioprotective Properties of Human Platelets Are Lost in Uncontrolled Diabetes Mellitus: A Study in Isolated Rat Hearts. Front Physiol 2018; 9:875. [PMID: 30042694 PMCID: PMC6048273 DOI: 10.3389/fphys.2018.00875] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Accepted: 06/19/2018] [Indexed: 12/13/2022] Open
Abstract
Platelets affect myocardial damage from ischemia/reperfusion. Redox-dependent sphingosine-1-phosphate production and release are altered in diabetic platelets. Sphingosine-1-phosphate is a double-edged sword for ischemia/reperfusion injury. Therefore, we aimed to verify whether: (1) human healthy- or diabetic-platelets are cardioprotective, (2) sphingosine-1-phosphate receptors and downstream kinases play a role in platelet-induced cardioprotection, and (3) a correlation between platelet redox status and myocardial ischemia/reperfusion injury exists. Isolated rat hearts were subjected to 30-min ischemia and 1-h reperfusion. Infarct size was studied in hearts pretreated with healthy- or diabetic-platelets. Healthy-platelets were co-infused with sphingosine-1-phosphate receptor blocker, ERK-1/2 inhibitor, PI3K antagonist or PKC inhibitor to ascertain the cardioprotective mechanisms. In platelets we assessed (i) aggregation response to ADP, collagen, and arachidonic-acid, (ii) cyclooxygenase-1 levels, and (iii) AKT and ERK-phosphorylation. Platelet sphingosine-1-phosphate production and platelet levels of reactive oxygen species (ROS) were quantified and correlated to infarct size. Infarct size was reduced by about 22% in healthy-platelets pretreated hearts only. This cardioprotective effect was abrogated by either sphingosine-1-phosphate receptors or ERK/PI3K/PKC pathway blockade. Cyclooxygenase-1 levels and aggregation indices were higher in diabetic-platelets than healthy-platelets. Diabetic-platelets released less sphingosine-1-phosphate than healthy-platelets when mechanical or chemically stimulated in vitro. Yet, ROS levels were higher in diabetic-platelets and correlated with infarct size. Cardioprotective effects of healthy-platelet depend on the platelet’s capacity to activate cardiac sphingosine-1-phosphate receptors and ERK/PI3K/PKC pathways. However, diabetic-platelets release less S1P and lose cardioprotective effects. Platelet ROS levels correlate with infarct size. Whether these redox alterations are responsible for sphingosine-1-phosphate dysfunction in diabetic-platelets remains to be ascertained.
Collapse
Affiliation(s)
- Isabella Russo
- Department of Clinical and Biological Sciences, AOU San Luigi, University of Turin, Turin, Italy
| | - Saveria Femminò
- Department of Clinical and Biological Sciences, AOU San Luigi, University of Turin, Turin, Italy.,Istituto Nazionale Ricerche Cardiovascolari (INRC), Bologna, Italy
| | - Cristina Barale
- Department of Clinical and Biological Sciences, AOU San Luigi, University of Turin, Turin, Italy
| | - Francesca Tullio
- Department of Clinical and Biological Sciences, AOU San Luigi, University of Turin, Turin, Italy.,Istituto Nazionale Ricerche Cardiovascolari (INRC), Bologna, Italy
| | - Stefano Geuna
- Department of Clinical and Biological Sciences, AOU San Luigi, University of Turin, Turin, Italy
| | - Franco Cavalot
- Internal Medicine and Metabolic Disease Unit, AOU San Luigi, University of Turin, Turin, Italy.,Ospedale San Luigi Gonzaga, Orbassano, Italy
| | - Pasquale Pagliaro
- Department of Clinical and Biological Sciences, AOU San Luigi, University of Turin, Turin, Italy.,Istituto Nazionale Ricerche Cardiovascolari (INRC), Bologna, Italy
| | - Claudia Penna
- Department of Clinical and Biological Sciences, AOU San Luigi, University of Turin, Turin, Italy.,Istituto Nazionale Ricerche Cardiovascolari (INRC), Bologna, Italy
| |
Collapse
|
41
|
Jeremic N, Zivkovic V, Srejovic I, Jeremic J, Petkovic A, Bradic J, Jakovljevic V. Effects of Ischemic and Proton Pump Inhibitors Preconditioning on Oxidative Stress of Isolated Rat Heart. SERBIAN JOURNAL OF EXPERIMENTAL AND CLINICAL RESEARCH 2018. [DOI: 10.1515/sjecr-2017-0008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Abstract
Aim of present study was to determine the participation of various biomarkers of oxidative damage: nitrite (NO2
−), superoxide anion radicals (O2
−), index of lipid peroxidation (TBARS) and hydrogen peroxide (H2O2) in coronary circulation after application of the different models of preconditioning such as ischemic and preconditioning with proton pump inhibitors.
Examining a biochemical markers of oxidative damage we did not notice any increased production values of any parameter, according to that we can hypothesize that possible occurrence of reperfusion injury after ischemia and PPIs preconditioning is not mediated by this mechanism.
Due to the very difficult and controversial application of ischemic preconditioning in clinical practice, the results of this study suggest that in the future proton pump inhibitors can contribute to the prevention of myocardial damage following ischemia
Collapse
Affiliation(s)
- Nevena Jeremic
- Department of Pharmaceutical chemistry, Faculty of Medical Sciences , University of Kragujevac , Kragujevac , Serbia
| | - Vladimir Zivkovic
- Department of Physiology, Faculty of Medical Sciences , University of Kragujevac , Kragujevac , Serbia
| | - Ivan Srejovic
- Department of Physiology, Faculty of Medical Sciences , University of Kragujevac , Kragujevac , Serbia
| | - Jovana Jeremic
- Department of Pharmacy, Faculty of Medical Sciences , University of Kragujevac , Kragujevac , Serbia
| | - Anica Petkovic
- Department of Pharmacy, Faculty of Medical Sciences , University of Kragujevac , Kragujevac , Serbia
| | - Jovana Bradic
- Department of Pharmacy, Faculty of Medical Sciences , University of Kragujevac , Kragujevac , Serbia
| | - Vladimir Jakovljevic
- Department of Physiology, Faculty of Medical Sciences , University of Kragujevac , Kragujevac , Serbia
- Department of Human Pathology , 1st Moscow State Medical University IM Sechenov , Moscow , Russia
| |
Collapse
|
42
|
Sairam T, Patel AN, Subrahmanian M, Gopalan R, Pogwizd SM, Ramalingam S, Sankaran R, Rajasekaran NS. Evidence for a hyper-reductive redox in a sub-set of heart failure patients. J Transl Med 2018; 16:130. [PMID: 29776421 PMCID: PMC5960146 DOI: 10.1186/s12967-018-1503-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Accepted: 05/03/2018] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Oxidative stress has been linked to heart failure (HF) in humans. Antioxidant-based treatments are often ineffective. Therefore, we hypothesize that some of the HF patients might have a reductive stress (RS) condition. Investigating RS-related mechanisms will aid in personalized optimization of redox homeostasis for better outcomes among HF patients. METHODS Blood samples were collected from HF patients (n = 54) and healthy controls (n = 42) and serum was immediately preserved in - 80 °C for redox analysis. Malondialdehyde (MDA; lipid peroxidation) levels by HPLC, reduced glutathione (GSH) and its redox ratio (GSH/GSSG) using enzymatic-recycling assay in the serum of HF patients were measured. Further, the activities of key antioxidant enzymes were analyzed by UV-Vis spectrophotometry. Non-invasive echocardiography was used to relate circulating redox status with cardiac function and remodeling. RESULTS The circulatory redox state (GSH/MDA ratio) was used to stratify the HF patients into normal redox (NR), hyper-oxidative (HO), and hyper-reductive (HR) groups. While the majority of the HF patients exhibited the HO (42%), 41% of them had a normal redox (NR) state. Surprisingly, a subset of HF patients (17%) belonged to the hyper-reductive group, suggesting a strong implication for RS in the progression of HF. In all the groups of HF patients, SOD, GPx and catalase were significantly increased while GR activity was significantly reduced relative to healthy controls. Furthermore, echocardiography analyses revealed that 55% of HO patients had higher systolic dysfunction while 62.5% of the hyper-reductive patients had higher diastolic dysfunction. CONCLUSION These results suggest that RS may be associated with HF pathogenesis for a subset of cardiac patients. Thus, stratification of HF patients based on their circulating redox status may serve as a useful prognostic tool to guide clinicians designing personalized antioxidant therapies.
Collapse
Affiliation(s)
- Thiagarajan Sairam
- PSG Center for Molecular Medicine and Therapeutics, PSG Institute of Medical Sciences & Research (Affiliated to the Tamilnadu Dr MGR Medical University), Coimbatore, Tamil Nadu, India
| | - Amit N Patel
- Division of Cardiothoracic Surgery, University of Miami-Miller School of Medicine, Miami, FL, USA
| | - Meenu Subrahmanian
- PSG Center for Molecular Medicine and Therapeutics, PSG Institute of Medical Sciences & Research (Affiliated to the Tamilnadu Dr MGR Medical University), Coimbatore, Tamil Nadu, India
| | - Rajendiran Gopalan
- Department of Cardiology, PSG Institute of Medical Sciences & Research (Affiliated to the Tamilnadu Dr MGR Medical University), Coimbatore, Tamil Nadu, India
| | - Steven M Pogwizd
- Comprehensive Cardiovascular Center, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Sudha Ramalingam
- PSG Center for Molecular Medicine and Therapeutics, PSG Institute of Medical Sciences & Research (Affiliated to the Tamilnadu Dr MGR Medical University), Coimbatore, Tamil Nadu, India
| | - Ramalingam Sankaran
- PSG Center for Molecular Medicine and Therapeutics, PSG Institute of Medical Sciences & Research (Affiliated to the Tamilnadu Dr MGR Medical University), Coimbatore, Tamil Nadu, India.
| | - Namakkal Soorapan Rajasekaran
- Cardiac Aging & Redox Signaling Laboratory, Center for Free Radical Biology, Division of Molecular & Cellular Pathology, Department of Pathology, UAB
- The University of Alabama at Birmingham, BMR2 Room 533
- 901 19th Street South, Birmingham, AL, 35294-2180, USA. .,PSG Center for Molecular Medicine and Therapeutics, PSG Institute of Medical Sciences & Research (Affiliated to the Tamilnadu Dr MGR Medical University), Coimbatore, Tamil Nadu, India. .,Cardiovascular Medicine, University of Utah School of Medicine, Salt Lake City, UT, USA.
| |
Collapse
|
43
|
Jiménez-Altayó F, Meirelles T, Crosas-Molist E, Sorolla MA, Del Blanco DG, López-Luque J, Mas-Stachurska A, Siegert AM, Bonorino F, Barberà L, García C, Condom E, Sitges M, Rodríguez-Pascual F, Laurindo F, Schröder K, Ros J, Fabregat I, Egea G. Redox stress in Marfan syndrome: Dissecting the role of the NADPH oxidase NOX4 in aortic aneurysm. Free Radic Biol Med 2018; 118:44-58. [PMID: 29471108 DOI: 10.1016/j.freeradbiomed.2018.02.023] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Revised: 02/12/2018] [Accepted: 02/15/2018] [Indexed: 11/16/2022]
Abstract
Marfan syndrome (MFS) is characterized by the formation of ascending aortic aneurysms resulting from altered assembly of extracellular matrix fibrillin-containing microfibrils and dysfunction of TGF-β signaling. Here we identify the molecular targets of redox stress in aortic aneurysms from MFS patients, and investigate the role of NOX4, whose expression is strongly induced by TGF-β, in aneurysm formation and progression in a murine model of MFS. Working models included aortae and cultured vascular smooth muscle cells (VSMC) from MFS patients, and a NOX4-deficient Marfan mouse model (Fbn1C1039G/+-Nox4-/-). Increased tyrosine nitration and reactive oxygen species levels were found in the tunica media of human aortic aneurysms and in cultured VSMC. Proteomic analysis identified nitrated and carbonylated proteins, which included smooth muscle α-actin (αSMA) and annexin A2. NOX4 immunostaining increased in the tunica media of human Marfan aorta and was transcriptionally overexpressed in VSMC. Fbn1C1039G/+-Nox4-/- mice aortas showed a reduction of fragmented elastic fibers, which was accompanied by an amelioration in the Marfan-associated enlargement of the aortic root. Increase in the contractile phenotype marker calponin in the tunica media of MFS mice aortas was abrogated in Fbn1C1039G/+-Nox4-/- mice. Endothelial dysfunction evaluated by myography in the Marfan ascending aorta was prevented by the absence of Nox4 or catalase-induced H2O2 decomposition. We conclude that redox stress occurs in MFS, whose targets are actin-based cytoskeleton members and regulators of extracellular matrix homeostasis. Likewise, NOX4 have an impact in the progression of the aortic dilation in MFS and in the structural organization of the aortic tunica media, the VSMC phenotypic modulation, and endothelial function.
Collapse
Affiliation(s)
- Francesc Jiménez-Altayó
- Departament de Farmacologia, Terapèutica i Toxicologia, Institut de Neurociències, Facultat de Medicina, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Thayna Meirelles
- Department de Biomedicina, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, and IDIBAPS, Barcelona, Spain
| | - Eva Crosas-Molist
- TGF-β and Cancer Group, Oncobell Program, IDIBELL, L'Hospitalet de Llobregat, Barcelona, Spain
| | - M Alba Sorolla
- Institut de Recerca Biomèdica de Lleida (IRB Lleida), Lleida, Spain; Departament de Ciències Mèdiques Bàsiques, Universitat de Lleida, Lleida, Spain
| | - Darya Gorbenko Del Blanco
- Department de Biomedicina, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, and IDIBAPS, Barcelona, Spain
| | - Judit López-Luque
- TGF-β and Cancer Group, Oncobell Program, IDIBELL, L'Hospitalet de Llobregat, Barcelona, Spain
| | | | - Ana-Maria Siegert
- Department de Biomedicina, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, and IDIBAPS, Barcelona, Spain
| | - Fabio Bonorino
- Department de Biomedicina, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, and IDIBAPS, Barcelona, Spain
| | - Laura Barberà
- Department de Biomedicina, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, and IDIBAPS, Barcelona, Spain
| | - Carolina García
- Department of Pathology, Hospital de Bellvitge-IDIBELL, L'Hospitalet de Llobregat, Barcelona, and Department of Pathology and Experimental Therapeutics, University of Barcelona, Barcelona, Spain
| | - Enric Condom
- Department of Pathology, Hospital de Bellvitge-IDIBELL, L'Hospitalet de Llobregat, Barcelona, and Department of Pathology and Experimental Therapeutics, University of Barcelona, Barcelona, Spain
| | - Marta Sitges
- Cardiovascular Institute, Hospital Clinic, IDIBAPS-University of Barcelona, Barcelona, Spain
| | | | - Francisco Laurindo
- Heart Institute (InCor), University of São Paulo School of Medicine, São Paulo, Brazil
| | - Katrin Schröder
- German Center of Cardiovascular Research (DZHK), Partner site Rhein Main, Frankfurt am Main, Germany
| | - Joaquim Ros
- Institut de Recerca Biomèdica de Lleida (IRB Lleida), Lleida, Spain; Departament de Ciències Mèdiques Bàsiques, Universitat de Lleida, Lleida, Spain
| | - Isabel Fabregat
- TGF-β and Cancer Group, Oncobell Program, IDIBELL, L'Hospitalet de Llobregat, Barcelona, Spain; Department de Ciències Fisiològiques, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, Barcelona, Spain
| | - Gustavo Egea
- Department de Biomedicina, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, and IDIBAPS, Barcelona, Spain.
| |
Collapse
|
44
|
Bulmer AC, Bakrania B, Du Toit EF, Boon AC, Clark PJ, Powell LW, Wagner KH, Headrick JP. Bilirubin acts as a multipotent guardian of cardiovascular integrity: more than just a radical idea. Am J Physiol Heart Circ Physiol 2018; 315:H429-H447. [PMID: 29600900 DOI: 10.1152/ajpheart.00417.2017] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Bilirubin, a potentially toxic catabolite of heme and indicator of hepatobiliary insufficiency, exhibits potent cardiac and vascular protective properties. Individuals with Gilbert's syndrome (GS) may experience hyperbilirubinemia in response to stressors including reduced hepatic bilirubin excretion/increased red blood cell breakdown, with individuals usually informed by their clinician that their condition is of little consequence. However, GS appears to protect from all-cause mortality, with progressively elevated total bilirubin associated with protection from ischemic heart and chronic obstructive pulmonary diseases. Bilirubin may protect against these diseases and associated mortality by reducing circulating cholesterol, oxidative lipid/protein modifications, and blood pressure. In addition, bilirubin inhibits platelet activation and protects the heart from ischemia-reperfusion injury. These effects attenuate multiple stages of the atherosclerotic process in addition to protecting the heart during resultant ischemic stress, likely underpinning the profound reduction in cardiovascular mortality in hyperbilirubinemic GS. This review outlines our current knowledge of and uses for bilirubin in clinical medicine and summarizes recent progress in revealing the physiological importance of this poorly understood molecule. We believe that this review will be of significant interest to clinicians, medical researchers, and individuals who have GS.
Collapse
Affiliation(s)
- Andrew C Bulmer
- School of Medical Science and Menzies Health Institute Queensland, Griffith University , Gold Coast, Queensland , Australia
| | - Bhavisha Bakrania
- Department of Physiology and Biophysics, University of Mississippi Medical Centre , Jackson, Mississippi
| | - Eugene F Du Toit
- School of Medical Science and Menzies Health Institute Queensland, Griffith University , Gold Coast, Queensland , Australia
| | - Ai-Ching Boon
- School of Medical Science and Menzies Health Institute Queensland, Griffith University , Gold Coast, Queensland , Australia
| | - Paul J Clark
- QIMR-Berghofer Medical Research Institute, School of Medicine, University of Queensland and Princess Alexandra and Mater Hospitals , Brisbane, New South Wales , Australia
| | - Lawrie W Powell
- The Centre for the Advancement of Clinical Research, Royal Brisbane and Women's Hospital, The University of Queensland Centre for Clinical Research , Brisbane, Queensland , Australia
| | - Karl-Heinz Wagner
- Department of Nutritional Science, University of Vienna , Vienna , Austria
| | - John P Headrick
- School of Medical Science and Menzies Health Institute Queensland, Griffith University , Gold Coast, Queensland , Australia
| |
Collapse
|
45
|
Pagliaro P, Femminò S, Popara J, Penna C. Mitochondria in Cardiac Postconditioning. Front Physiol 2018; 9:287. [PMID: 29632499 PMCID: PMC5879113 DOI: 10.3389/fphys.2018.00287] [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: 01/24/2018] [Accepted: 03/12/2018] [Indexed: 12/11/2022] Open
Abstract
Mitochondria play a pivotal role in cardioprotection. Here we report some fundamental studies which considered the role of mitochondrial components (connexin 43, mitochondrial KATP channels and mitochondrial permeability transition pore) in postconditioning cardioprotection. We briefly discuss the role of mitochondria, reactive oxygen species and gaseous molecules in postconditioning. Also the effects of anesthetics-used as cardioprotective substances-is briefly considered in the context of postconditioning. The role of mitochondrial postconditioning signaling in determining the limitation of cell death is underpinned. Issues in clinical translation are briefly considered. The aim of the present mini-review is to discuss in a historical perspective the role of main mitochondria mechanisms in cardiac postconditioning.
Collapse
Affiliation(s)
- Pasquale Pagliaro
- Department of Clinical and Biological Sciences, University of Turin, Turin, Italy
| | - Saveria Femminò
- Department of Clinical and Biological Sciences, University of Turin, Turin, Italy
| | - Jasmin Popara
- Department of Clinical and Biological Sciences, University of Turin, Turin, Italy
| | - Claudia Penna
- Department of Clinical and Biological Sciences, University of Turin, Turin, Italy
| |
Collapse
|
46
|
Sorriento D, De Luca N, Trimarco B, Iaccarino G. The Antioxidant Therapy: New Insights in the Treatment of Hypertension. Front Physiol 2018; 9:258. [PMID: 29618986 PMCID: PMC5871811 DOI: 10.3389/fphys.2018.00258] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Accepted: 03/06/2018] [Indexed: 12/12/2022] Open
Abstract
Reactive oxygen species (ROS) and reactive nitrogen species (RNS) play a key role in the regulation of the physiological and pathological signaling within the vasculature. In physiological conditions, a delicate balance between oxidants and antioxidants protects cells from the detrimental effects of ROS/RNS. Indeed, the imbalance between ROS/RNS production and antioxidant defense mechanisms leads to oxidative and nitrosative stress within the cell. These processes promote the vascular damage observed in chronic conditions, such as hypertension. The strong implication of ROS/RNS in the etiology of hypertension suggest that antioxidants could be effective in the treatment of this pathology. Indeed, in animal models of hypertension, the overexpression of antioxidants and the genetic modulation of oxidant systems have provided an encouraging proof of concept. Nevertheless, the translation of these strategies to human disease did not reach the expected success. This could be due to the complexity of this condition, whose etiology depends on multiple factors (smoking, diet, life styles, genetics, family history, comorbidities). Indeed, 95% of reported high blood pressure cases are deemed "essential hypertension," and at the molecular level, oxidative stress seems to be a common feature of hypertensive states. In this scenario, new therapies are emerging that could be useful to reduce oxidative stress in hypertension. It is now ascertained the role of Vitamin D deficiency in the development of essential hypertension and it has been shown that an appropriate high dose of Vitamin D significantly reduces blood pressure in hypertensive cohorts with vitamin D deficiency. Moreover, new drugs are emerging which have both antihypertensive action and antioxidant properties, such as celiprolol, carvedilol, nebivolol. Indeed, besides adrenergic desensitization, these kind of drugs are able to interfere with ROS/RNS generation and/or signaling, and are therefore considered promising therapeutics in the management of hypertension. In the present review we have dealt with the effectiveness of the antioxidant therapy in the management of hypertension. In particular, we discuss about Vitamin D and anti-hypertensive drugs with antioxidant properties.
Collapse
Affiliation(s)
- Daniela Sorriento
- Dipartimento di Scienze Biomediche Avanzate, Università Federico II, Napoli, Italy
| | - Nicola De Luca
- Dipartimento di Scienze Biomediche Avanzate, Università Federico II, Napoli, Italy
| | - Bruno Trimarco
- Dipartimento di Scienze Biomediche Avanzate, Università Federico II, Napoli, Italy
| | - Guido Iaccarino
- Dipartimento di Medicina, Chirurgia e Odontoiatria, Università degli Studi di Salerno, Baronissi, Italy
| |
Collapse
|
47
|
Varricchi G, Ameri P, Cadeddu C, Ghigo A, Madonna R, Marone G, Mercurio V, Monte I, Novo G, Parrella P, Pirozzi F, Pecoraro A, Spallarossa P, Zito C, Mercuro G, Pagliaro P, Tocchetti CG. Antineoplastic Drug-Induced Cardiotoxicity: A Redox Perspective. Front Physiol 2018; 9:167. [PMID: 29563880 PMCID: PMC5846016 DOI: 10.3389/fphys.2018.00167] [Citation(s) in RCA: 100] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Accepted: 02/20/2018] [Indexed: 12/28/2022] Open
Abstract
Antineoplastic drugs can be associated with several side effects, including cardiovascular toxicity (CTX). Biochemical studies have identified multiple mechanisms of CTX. Chemoterapeutic agents can alter redox homeostasis by increasing the production of reactive oxygen species (ROS) and reactive nitrogen species RNS. Cellular sources of ROS/RNS are cardiomyocytes, endothelial cells, stromal and inflammatory cells in the heart. Mitochondria, peroxisomes and other subcellular components are central hubs that control redox homeostasis. Mitochondria are central targets for antineoplastic drug-induced CTX. Understanding the mechanisms of CTX is fundamental for effective cardioprotection, without compromising the efficacy of anticancer treatments. Type 1 CTX is associated with irreversible cardiac cell injury and is typically caused by anthracyclines and conventional chemotherapeutic agents. Type 2 CTX, associated with reversible myocardial dysfunction, is generally caused by biologicals and targeted drugs. Although oxidative/nitrosative reactions play a central role in CTX caused by different antineoplastic drugs, additional mechanisms involving directly and indirectly cardiomyocytes and inflammatory cells play a role in cardiovascular toxicities. Identification of cardiologic risk factors and an integrated approach using molecular, imaging, and clinical data may allow the selection of patients at risk of developing chemotherapy-related CTX. Although the last decade has witnessed intense research related to the molecular and biochemical mechanisms of CTX of antineoplastic drugs, experimental and clinical studies are urgently needed to balance safety and efficacy of novel cancer therapies.
Collapse
Affiliation(s)
- Gilda Varricchi
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy
- Department of Translational Medical Sciences, Center for Basic and Clinical Immunology Research, University of Naples Federico II, Naples, Italy
| | - Pietro Ameri
- Clinic of Cardiovascular Diseases, IRCCS San Martino IST, Genova, Italy
| | - Christian Cadeddu
- Department of Medical Sciences and Public Health, University of Cagliari, Cagliari, Italy
| | - Alessandra Ghigo
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Turin, Turin, Italy
| | - Rosalinda Madonna
- Institute of Cardiology, Center of Excellence on Aging, Università degli Studi “G. d'Annunzio” Chieti – Pescara, Chieti, Italy
- Department of Internal Medicine, Texas Heart Institute and Center for Cardiovascular Biology and Atherosclerosis Research, University of Texas Health Science Center, Houston, TX, United States
| | - Giancarlo Marone
- Section of Hygiene, Department of Public Health, University of Naples Federico II, Naples, Italy
- Monaldi Hospital Pharmacy, Naples, Italy
| | - Valentina Mercurio
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy
| | - Ines Monte
- Department of General Surgery and Medical-Surgery Specialities, University of Catania, Catania, Italy
| | - Giuseppina Novo
- U.O.C. Magnetic Resonance Imaging, Fondazione Toscana G. Monasterio C.N.R., Pisa, Italy
| | - Paolo Parrella
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy
| | - Flora Pirozzi
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy
| | - Antonio Pecoraro
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy
| | - Paolo Spallarossa
- Clinic of Cardiovascular Diseases, IRCCS San Martino IST, Genova, Italy
| | - Concetta Zito
- Division of Clinical and Experimental Cardiology, Department of Medicine and Pharmacology, Policlinico “G. Martino” University of Messina, Messina, Italy
| | - Giuseppe Mercuro
- Department of Medical Sciences and Public Health, University of Cagliari, Cagliari, Italy
| | - Pasquale Pagliaro
- Department of Clinical and Biological Sciences, University of Turin, Turin, Italy
| | - Carlo G. Tocchetti
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy
| |
Collapse
|
48
|
Cadenas S. ROS and redox signaling in myocardial ischemia-reperfusion injury and cardioprotection. Free Radic Biol Med 2018; 117:76-89. [PMID: 29373843 DOI: 10.1016/j.freeradbiomed.2018.01.024] [Citation(s) in RCA: 516] [Impact Index Per Article: 86.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Revised: 01/19/2018] [Accepted: 01/21/2018] [Indexed: 02/06/2023]
Abstract
Ischemia-reperfusion (IR) injury is central to the pathology of major cardiovascular diseases, such as stroke and myocardial infarction. IR injury is mediated by several factors including the elevated production of reactive oxygen species (ROS), which occurs particularly at reperfusion. The mitochondrial respiratory chain and NADPH oxidases of the NOX family are major sources of ROS in cardiomyocytes. The first part of this review discusses recent findings and controversies on the mechanisms of superoxide production by the mitochondrial electron transport chain during IR injury, as well as the contribution of the NOX isoforms expressed in cardiomyocytes, NOX1, NOX2 and NOX4, to this damage. It then focuses on the effects of ROS on the opening of the mitochondrial permeability transition pore (mPTP), an inner membrane non-selective pore that causes irreversible damage to the heart. The second part analyzes the redox mechanisms of cardiomyocyte mitochondrial protection; specifically, the activation of the hypoxia-inducible factor (HIF) pathway and the antioxidant transcription factor Nrf2, which are both regulated by the cellular redox state. Redox mechanisms involved in ischemic preconditioning, one of the most effective ways of protecting the heart against IR injury, are also reviewed. Interestingly, several of these protective pathways converge on the inhibition of mPTP opening during reperfusion. Finally, the clinical and translational implications of these cardioprotective mechanisms are discussed.
Collapse
Affiliation(s)
- Susana Cadenas
- Centro de Biología Molecular "Severo Ochoa" (CSIC-UAM) and Departamento de Biología Molecular, Universidad Autónoma de Madrid, Cantoblanco, 28049 Madrid, Spain; Instituto de Investigación Sanitaria Princesa (IIS-IP), 28006 Madrid, Spain.
| |
Collapse
|
49
|
Femminò S, Penna C, Bessone F, Caldera F, Dhakar N, Cau D, Pagliaro P, Cavalli R, Trotta F. α-Cyclodextrin and α-Cyclodextrin Polymers as Oxygen Nanocarriers to Limit Hypoxia/Reoxygenation Injury: Implications from an In Vitro Model. Polymers (Basel) 2018; 10:polym10020211. [PMID: 30966247 PMCID: PMC6414891 DOI: 10.3390/polym10020211] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Revised: 02/14/2018] [Accepted: 02/16/2018] [Indexed: 01/20/2023] Open
Abstract
The incidence of heart failure (HF) is increasing worldwide and myocardial infarction (MI), which follows ischemia and reperfusion (I/R), is often at the basis of HF development. Nanocarriers are interesting particles for their potential application in cardiovascular disease. Impaired drug delivery in ischemic disease is challenging. Cyclodextrin nanosponges (NS) can be considered innovative tools for improving oxygen delivery in a controlled manner. This study has developed new α-cyclodextrin-based formulations as oxygen nanocarriers such as native α-cyclodextrin (α-CD), branched α-cyclodextrin polymer (α-CD POLY), and α-cyclodextrin nanosponges (α-CD NS). The three different α-CD-based formulations were tested at 0.2, 2, and 20 µg/mL to ascertain their capability to reduce cell mortality during hypoxia and reoxygenation (H/R) in vitro protocols. H9c2, a cardiomyoblast cell line, was exposed to normoxia (20% oxygen) or hypoxia (5% CO2 and 95% N2). The different formulations, applied before hypoxia, induced a significant reduction in cell mortality (in a range of 15% to 30%) when compared to samples devoid of oxygen. Moreover, their application at the beginning of reoxygenation induced a considerable reduction in cell death (12% to 20%). α-CD NS showed a marked efficacy in controlled oxygenation, which suggests an interesting potential for future medical application of polymer systems for MI treatment.
Collapse
Affiliation(s)
- Saveria Femminò
- Department of Clinical and Biological Sciences, University of Turin, 10043 Orbassano, Italy.
| | - Claudia Penna
- Department of Clinical and Biological Sciences, University of Turin, 10043 Orbassano, Italy.
| | - Federica Bessone
- Department of Drug Science and Technology, University of Turin, 10125 Turin, Italy.
| | - Fabrizio Caldera
- Department of Chemistry, University of Turin, 10125 Turin, Italy.
| | - Nilesh Dhakar
- Department of Chemistry, University of Turin, 10125 Turin, Italy.
| | - Daniele Cau
- Department of Clinical and Biological Sciences, University of Turin, 10043 Orbassano, Italy.
| | - Pasquale Pagliaro
- Department of Clinical and Biological Sciences, University of Turin, 10043 Orbassano, Italy.
| | - Roberta Cavalli
- Department of Drug Science and Technology, University of Turin, 10125 Turin, Italy.
| | - Francesco Trotta
- Department of Chemistry, University of Turin, 10125 Turin, Italy.
| |
Collapse
|
50
|
Andreadou I, Efentakis P, Balafas E, Togliatto G, Davos CH, Varela A, Dimitriou CA, Nikolaou PE, Maratou E, Lambadiari V, Ikonomidis I, Kostomitsopoulos N, Brizzi MF, Dimitriadis G, Iliodromitis EK. Empagliflozin Limits Myocardial Infarction in Vivo and Cell Death in Vitro: Role of STAT3, Mitochondria, and Redox Aspects. Front Physiol 2017; 8:1077. [PMID: 29311992 PMCID: PMC5742117 DOI: 10.3389/fphys.2017.01077] [Citation(s) in RCA: 111] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Accepted: 12/06/2017] [Indexed: 12/11/2022] Open
Abstract
Empagliflozin (EMPA), a drug approved for type 2 diabetes management, reduced cardiovascular death but is unknown if it reduces myocardial infarction. We sought to investigate: (i) the effect of EMPA on myocardial function and infarct size after ischemia/reperfusion in mice fed with western diet (WD), (ii) the underlying signaling pathways, (iii) its effects on cell survival in rat embryonic-heart-derived cardiomyoblasts (H9C2) and endothelial cells (ECs). To facilitate the aforementioned aims, mice were initially randomized in Control and EMPA groups and were subjected to 30 min ischemia and 2 h reperfusion. EMPA reduced body weight, blood glucose levels, and mean arterial pressure. Cholesterol, triglyceride, and AGEs remained unchanged. Left ventricular fractional shortening was improved (43.97 ± 0.92 vs. 40.75 ± 0.61%) and infarct size reduced (33.2 ± 0.01 vs. 17.6 ± 0.02%). In a second series of experiments, mice were subjected to the above interventions up to the 10th min of reperfusion and myocardial biopsies were obtained for assessment of the signaling cascade. STAT3 was increased in parallel with reduced levels of malondialdehyde (MDA) and reduced expression of myocardial iNOS and interleukin-6. Cell viability and ATP content were increased in H9C2 and in ECs. While, STAT3 phosphorylation is known to bestow infarct sparing properties through interaction with mitochondria, we observed that EMPA did not directly alter the mitochondrial calcium retention capacity (CRC); therefore, its effect in reducing myocardial infarction is STAT3 dependent. In conclusion, EMPA improves myocardial function and reduces infarct size as well as improves redox regulation by decreasing iNOS expression and subsequently lipid peroxidation as shown by its surrogate marker MDA. The mechanisms of action implicate the activation of STAT3 anti-oxidant and anti-inflammatory properties.
Collapse
Affiliation(s)
- Ioanna Andreadou
- Laboratory of Pharmacology, Faculty of Pharmacy, School of Health Sciences, National and Kapodistrian University of Athens, Athens, Greece
| | - Panagiotis Efentakis
- Laboratory of Pharmacology, Faculty of Pharmacy, School of Health Sciences, National and Kapodistrian University of Athens, Athens, Greece
| | - Evangelos Balafas
- Academy of Athens Biomedical Research Foundation, Centre of Clinical Experimental Surgery and Translational Research, Athens, Greece
| | | | - Constantinos H Davos
- Cardiovascular Research Laboratory, Biomedical Research Foundation, Academy of Athens, Athens, Greece
| | - Aimilia Varela
- Cardiovascular Research Laboratory, Biomedical Research Foundation, Academy of Athens, Athens, Greece
| | - Constantinos A Dimitriou
- Cardiovascular Research Laboratory, Biomedical Research Foundation, Academy of Athens, Athens, Greece
| | - Panagiota-Efstathia Nikolaou
- Laboratory of Pharmacology, Faculty of Pharmacy, School of Health Sciences, National and Kapodistrian University of Athens, Athens, Greece
| | - Eirini Maratou
- Hellenic National Center for Research, Prevention and Treatment of Diabetes Mellitus and Its Complications, Athens, Greece
| | - Vaia Lambadiari
- 2nd Department of Internal Medicine, Research Institute and Diabetes Center, National and Kapodistrian University of Athens, "Attikon" University Hospital, Athens, Greece
| | - Ignatios Ikonomidis
- 2nd University Department of Cardiology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Nikolaos Kostomitsopoulos
- Academy of Athens Biomedical Research Foundation, Centre of Clinical Experimental Surgery and Translational Research, Athens, Greece
| | - Maria F Brizzi
- Department of Medical Sciences, University of Turin, Turin, Italy
| | - George Dimitriadis
- 2nd Department of Internal Medicine, Research Institute and Diabetes Center, National and Kapodistrian University of Athens, "Attikon" University Hospital, Athens, Greece
| | - Efstathios K Iliodromitis
- 2nd University Department of Cardiology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|