1
|
Barrère-Lemaire S, Vincent A, Jorgensen C, Piot C, Nargeot J, Djouad F. Mesenchymal stromal cells for improvement of cardiac function following acute myocardial infarction: a matter of timing. Physiol Rev 2024; 104:659-725. [PMID: 37589393 DOI: 10.1152/physrev.00009.2023] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 07/05/2023] [Accepted: 08/16/2023] [Indexed: 08/18/2023] Open
Abstract
Acute myocardial infarction (AMI) is the leading cause of cardiovascular death and remains the most common cause of heart failure. Reopening of the occluded artery, i.e., reperfusion, is the only way to save the myocardium. However, the expected benefits of reducing infarct size are disappointing due to the reperfusion paradox, which also induces specific cell death. These ischemia-reperfusion (I/R) lesions can account for up to 50% of final infarct size, a major determinant for both mortality and the risk of heart failure (morbidity). In this review, we provide a detailed description of the cell death and inflammation mechanisms as features of I/R injury and cardioprotective strategies such as ischemic postconditioning as well as their underlying mechanisms. Due to their biological properties, the use of mesenchymal stromal/stem cells (MSCs) has been considered a potential therapeutic approach in AMI. Despite promising results and evidence of safety in preclinical studies using MSCs, the effects reported in clinical trials are not conclusive and even inconsistent. These discrepancies were attributed to many parameters such as donor age, in vitro culture, and storage time as well as injection time window after AMI, which alter MSC therapeutic properties. In the context of AMI, future directions will be to generate MSCs with enhanced properties to limit cell death in myocardial tissue and thereby reduce infarct size and improve the healing phase to increase postinfarct myocardial performance.
Collapse
Affiliation(s)
- Stéphanie Barrère-Lemaire
- Institut de Génomique Fonctionnelle, Université de Montpellier, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Montpellier, France
- LabEx Ion Channel Science and Therapeutics, Université de Nice, Nice, France
| | - Anne Vincent
- Institut de Génomique Fonctionnelle, Université de Montpellier, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Montpellier, France
- LabEx Ion Channel Science and Therapeutics, Université de Nice, Nice, France
| | - Christian Jorgensen
- Institute of Regenerative Medicine and Biotherapies, Université de Montpellier, Institut National de la Santé et de la Recherche Médicale, Montpellier, France
- Centre Hospitalier Universitaire Montpellier, Montpellier, France
| | - Christophe Piot
- Département de Cardiologie Interventionnelle, Clinique du Millénaire, Montpellier, France
| | - Joël Nargeot
- Institut de Génomique Fonctionnelle, Université de Montpellier, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Montpellier, France
- LabEx Ion Channel Science and Therapeutics, Université de Nice, Nice, France
| | - Farida Djouad
- Institute of Regenerative Medicine and Biotherapies, Université de Montpellier, Institut National de la Santé et de la Recherche Médicale, Montpellier, France
- Centre Hospitalier Universitaire Montpellier, Montpellier, France
| |
Collapse
|
2
|
Carmo HRP, Bonilha I, Barreto J, Tognolini M, Zanotti I, Sposito AC. High-Density Lipoproteins at the Interface between the NLRP3 Inflammasome and Myocardial Infarction. Int J Mol Sci 2024; 25:1290. [PMID: 38279290 PMCID: PMC10816227 DOI: 10.3390/ijms25021290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Revised: 01/10/2024] [Accepted: 01/18/2024] [Indexed: 01/28/2024] Open
Abstract
Despite significant therapeutic advancements, morbidity and mortality following myocardial infarction (MI) remain unacceptably high. This clinical challenge is primarily attributed to two significant factors: delayed reperfusion and the myocardial injury resulting from coronary reperfusion. Following reperfusion, there is a rapid intracellular pH shift, disruption of ionic balance, heightened oxidative stress, increased activity of proteolytic enzymes, initiation of inflammatory responses, and activation of several cell death pathways, encompassing apoptosis, necroptosis, and pyroptosis. The inflammatory cell death or pyroptosis encompasses the activation of the intracellular multiprotein complex known as the NLRP3 inflammasome. High-density lipoproteins (HDL) are endogenous particles whose components can either promote or mitigate the activation of the NLRP3 inflammasome. In this comprehensive review, we explore the role of inflammasome activation in the context of MI and provide a detailed analysis of how HDL can modulate this process.
Collapse
Affiliation(s)
- Helison R. P. Carmo
- Atherosclerosis and Vascular Biology Laboratory (Aterolab), Division of Cardiology, State University of Campinas (UNICAMP), Campinas 13084-971, SP, Brazil; (H.R.P.C.); (I.B.); (J.B.); (A.C.S.)
| | - Isabella Bonilha
- Atherosclerosis and Vascular Biology Laboratory (Aterolab), Division of Cardiology, State University of Campinas (UNICAMP), Campinas 13084-971, SP, Brazil; (H.R.P.C.); (I.B.); (J.B.); (A.C.S.)
| | - Joaquim Barreto
- Atherosclerosis and Vascular Biology Laboratory (Aterolab), Division of Cardiology, State University of Campinas (UNICAMP), Campinas 13084-971, SP, Brazil; (H.R.P.C.); (I.B.); (J.B.); (A.C.S.)
| | | | - Ilaria Zanotti
- Department of Food and Drug, University of Parma, 43124 Parma, Italy;
| | - Andrei C. Sposito
- Atherosclerosis and Vascular Biology Laboratory (Aterolab), Division of Cardiology, State University of Campinas (UNICAMP), Campinas 13084-971, SP, Brazil; (H.R.P.C.); (I.B.); (J.B.); (A.C.S.)
| |
Collapse
|
3
|
Schoch L, Alcover S, Padró T, Ben-Aicha S, Mendieta G, Badimon L, Vilahur G. Update of HDL in atherosclerotic cardiovascular disease. CLINICA E INVESTIGACION EN ARTERIOSCLEROSIS : PUBLICACION OFICIAL DE LA SOCIEDAD ESPANOLA DE ARTERIOSCLEROSIS 2023; 35:297-314. [PMID: 37940388 DOI: 10.1016/j.arteri.2023.10.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Accepted: 10/13/2023] [Indexed: 11/10/2023]
Abstract
Epidemiologic evidence supported an inverse association between HDL (high-density lipoprotein) cholesterol (HDL-C) levels and atherosclerotic cardiovascular disease (ASCVD), identifying HDL-C as a major cardiovascular risk factor and postulating diverse HDL vascular- and cardioprotective functions beyond their ability to drive reverse cholesterol transport. However, the failure of several clinical trials aimed at increasing HDL-C in patients with overt cardiovascular disease brought into question whether increasing the cholesterol cargo of HDL was an effective strategy to enhance their protective properties. In parallel, substantial evidence supports that HDLs are complex and heterogeneous particles whose composition is essential for maintaining their protective functions, subsequently strengthening the "HDL quality over quantity" hypothesis. The following state-of-the-art review covers the latest understanding as per the roles of HDL in ASCVD, delves into recent advances in understanding the complexity of HDL particle composition, including proteins, lipids and other HDL-transported components and discusses on the clinical outcomes after the administration of HDL-C raising drugs with particular attention to CETP (cholesteryl ester transfer protein) inhibitors.
Collapse
Affiliation(s)
- Leonie Schoch
- Cardiovascular Program, Institut de Recerca, Hospital de la Santa Creu I Sant Pau, IIB Sant Pau, 08025 Barcelona, Spain; Faculty of Medicine, University of Barcelona (UB), 08036 Barcelona, Spain
| | - Sebastián Alcover
- Cardiovascular Program, Institut de Recerca, Hospital de la Santa Creu I Sant Pau, IIB Sant Pau, 08025 Barcelona, Spain
| | - Teresa Padró
- Cardiovascular Program, Institut de Recerca, Hospital de la Santa Creu I Sant Pau, IIB Sant Pau, 08025 Barcelona, Spain
| | | | - Guiomar Mendieta
- Cardiology Unit, Cardiovascular Clinical Institute, Hospital Clínic de Barcelona, Barcelona, Spain
| | - Lina Badimon
- Cardiovascular Program, Institut de Recerca, Hospital de la Santa Creu I Sant Pau, IIB Sant Pau, 08025 Barcelona, Spain; Cardiovascular Research Chair, UAB, 08025 Barcelona, Spain; CiberCV, Institute of Health Carlos III, Madrid, Spain
| | - Gemma Vilahur
- Cardiovascular Program, Institut de Recerca, Hospital de la Santa Creu I Sant Pau, IIB Sant Pau, 08025 Barcelona, Spain; CiberCV, Institute of Health Carlos III, Madrid, Spain.
| |
Collapse
|
4
|
Cipolla MJ. Therapeutic Induction of Collateral Flow. Transl Stroke Res 2023; 14:53-65. [PMID: 35416577 PMCID: PMC10155807 DOI: 10.1007/s12975-022-01019-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 03/31/2022] [Accepted: 04/01/2022] [Indexed: 01/31/2023]
Abstract
Therapeutic induction of collateral flow as a means to salvage tissue and improve outcome from acute ischemic stroke is a promising approach in the era in which endovascular therapy is no longer time-dependent but collateral-dependent. The importance of collateral flow enhancement as a therapeutic for acute ischemic stroke extends beyond those patients with large amounts of salvageable tissue. It also has the potential to extend the time window for reperfusion therapies in patients who are ineligible for endovascular thrombectomy. In addition, collateral enhancement may be an important adjuvant to neuroprotective agents by providing a more robust vascular route for which treatments can gain access to at risk tissue. However, our understanding of collateral hemodynamics, including under comorbid conditions that are highly prevalent in the stroke population, has hindered the efficacy of collateral flow augmentation for improving stroke outcome in the clinical setting. This review will discuss our current understanding of pial collateral function and hemodynamics, including vasoactivity that is critical for enhancing penumbral perfusion. In addition, mechanisms by which collateral flow can be increased during acute ischemic stroke to limit ischemic injury, that may be different depending on the state of the brain and vasculature prior to stroke, will also be reviewed.
Collapse
Affiliation(s)
- Marilyn J Cipolla
- Department of Neurological Sciences, University of Vermont Robert Larner College of Medicine, 149 Beaumont Ave, HSRF 416A, Burlington, VT, USA.
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of Vermont Larner College of Medicine, Burlington, VT, USA.
- Department of Pharmacology, University of Vermont Larner College of Medicine, Burlington, VT, USA.
| |
Collapse
|
5
|
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
|
6
|
Bonaca MP, Morrow DA, Bergmark BA, Berg DD, Lima JAC, Hoffmann U, Kato Y, Lu MT, Kuder J, Murphy SA, Spinar J, Oude Ophuis T, Kiss RG, Lopez-Sendon J, Averkov O, Wheatcroft SB, Kubica J, Carlos Nicolau J, Furtado RHM, Abuhatzira L, Hirshberg B, Omar SA, Vavere AL, Chang YT, George RT, Sabatine MS. Randomized, Placebo-Controlled Phase 2b Study to Evaluate the Safety and Efficacy of Recombinant Human Lecithin Cholesterol Acyltransferase in Acute ST-Segment-Elevation Myocardial Infarction: Results of REAL-TIMI 63B. Circulation 2022; 146:907-916. [PMID: 36039762 DOI: 10.1161/circulationaha.122.059325] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
BACKGROUND High-density lipoprotein plays a key role in reverse cholesterol transport. In addition, high-density lipoprotein particles may be cardioprotective and reduce infarct size in the setting of myocardial injury. Lecithin-cholesterol acyltransferase is a rate-limiting enzyme in reverse cholesterol transport. MEDI6012 is a recombinant human lecithin-cholesterol acyltransferase that increases high-density lipoprotein cholesterol. Administration of lecithin-cholesterol acyltransferase has the potential to reduce infarct size and regress coronary plaque in acute ST-segment-elevation myocardial infarction. METHODS REAL-TIMI 63B (A Randomized, Placebo‑controlled Phase 2b Study to Evaluate the Safety and Efficacy of MEDI6012 in Acute ST Elevation Myocardial Infarction) was a phase 2B multinational, placebo-controlled, randomized trial. Patients with ST-segment-elevation myocardial infarction within 6 hours of symptom onset and planned for percutaneous intervention were randomly assigned 2:1 to MEDI6012 (2- or 6-dose regimen) or placebo and followed for 12 weeks. The primary outcome was infarct size as a percentage of left ventricular mass by cardiac MRI at 10 to 12 weeks, with the primary analysis in patients with TIMI Flow Grade 0 to 1 before percutaneous intervention who received at least 2 doses of MEDI6012. The secondary outcome was change in noncalcified plaque volume on coronary computed tomographic angiography from baseline to 10 to 12 weeks with the primary analysis in patients who received all 6 doses of MEDI6012. RESULTS A total of 593 patients were randomly assigned. Patients were a median of 62 years old, 77.9% male, and 95.8% statin naive. Median time from symptom onset to randomization was 146 (interquartile range [IQR], 103-221) minutes and from hospitalization to randomization was 12.7 (IQR, 6.6-24.0) minutes, and the first dose of drug was administered a median of 8 (IQR, 3-13) minutes before percutaneous intervention. The index myocardial infarction was anterior in 69.6% and TIMI Flow Grade 0 to 1 in 65.1% of patients. At 12 weeks, infarct size did not differ between treatment groups (MEDI6012: 9.71%, IQR 4.79-16.38; placebo: 10.48%, [IQR, 4.92-16.61], 1-sided P=0.79. There was also no difference in noncalcified plaque volume (geometric mean ratio, 0.96 [95% CI, NA-1.10], 1-sided P=0.30). There was no significant difference in treatment emergent serious adverse events. CONCLUSIONS Administration of MEDI6012 in patients with acute ST-segment-elevation myocardial infarction did not result in a significant reduction in infarct size or noncalcified plaque volume at 12 weeks. MEDI6012 was well tolerated with no excess in overall serious adverse events. REGISTRATION URL: https://www. CLINICALTRIALS gov; Unique identifier: NCT03578809.
Collapse
Affiliation(s)
- Marc P Bonaca
- CPC Clinical Research, Department of Medicine, University of Colorado Anschutz School of Medicine, Aurora (M.P.B.)
| | - David A Morrow
- TIMI Study Group, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (D.A.M., B.A.B., D.D.B., J. Kuder, S.A.M., M.S.S.)
| | - Brian A Bergmark
- TIMI Study Group, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (D.A.M., B.A.B., D.D.B., J. Kuder, S.A.M., M.S.S.)
| | - David D Berg
- TIMI Study Group, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (D.A.M., B.A.B., D.D.B., J. Kuder, S.A.M., M.S.S.)
| | - Joao A C Lima
- Division of Cardiology, Johns Hopkins University, Baltimore, MD (J.A.C.L., Y.K.)
| | - Udo Hoffmann
- Internal Cardioangiology Department, St. Ann University Hospital and Masaryk University, Brno, Czechia (J.S.)
| | - Yoko Kato
- Division of Cardiology, Johns Hopkins University, Baltimore, MD (J.A.C.L., Y.K.)
| | - Michael T Lu
- Cardiovascular Imaging Research Center, Massachusetts General Hospital and Harvard Medical School, Boston (U.H., M.T.L.)
| | - Julia Kuder
- TIMI Study Group, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (D.A.M., B.A.B., D.D.B., J. Kuder, S.A.M., M.S.S.)
| | - Sabina A Murphy
- TIMI Study Group, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (D.A.M., B.A.B., D.D.B., J. Kuder, S.A.M., M.S.S.)
| | - Jindrich Spinar
- Internal Cardioangiology Department, St. Ann University Hospital and Masaryk University, Brno, Czechia (J.S.)
| | - Ton Oude Ophuis
- Canisius Wilhelmina Ziekenhuis, Nijmegen, The Netherlands (T.O.O.)
| | - Róbert G Kiss
- Department of Cardiology, Military Hospital, Budapest, Hungary (R.G.K.)
| | - Jose Lopez-Sendon
- IdiPaz Research Institute, Hospital Universitario La Paz, UAM, Madrid, Spain (J.L.-S.)
| | - Oleg Averkov
- Pirogov Russian National Research Medical University, Moscow (O.A.)
| | - Stephen B Wheatcroft
- Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, UK (S.B.W.)
| | - Jacek Kubica
- Department of Cardiology and Internal Medicine, Collegium Medicum, Nicolaus Copernicus University, Bydgoszcz, Poland (J. Kubica)
| | - Jose Carlos Nicolau
- Instituto do Coracao, Hospital das Clinicas, Faculdade de Medicina, Universidade de Sao Paulo, Brazil (J.C.N., R.H.M.F.)
| | - Remo H M Furtado
- Instituto do Coracao, Hospital das Clinicas, Faculdade de Medicina, Universidade de Sao Paulo, Brazil (J.C.N., R.H.M.F.).,Academic Research Organization, Hospital Israelita Albert Einstein, Sao Paulo, Brazil (R.H.M.G.)
| | | | | | - Sami A Omar
- Early Clinical Development, Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD (S.A.O., A.L.V., Y.-T.C., R.T.G.)
| | - Andrea L Vavere
- Early Clinical Development, Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD (S.A.O., A.L.V., Y.-T.C., R.T.G.)
| | - Yi-Ting Chang
- Early Clinical Development, Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD (S.A.O., A.L.V., Y.-T.C., R.T.G.)
| | - Richard T George
- Early Clinical Development, Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD (S.A.O., A.L.V., Y.-T.C., R.T.G.)
| | - Marc S Sabatine
- TIMI Study Group, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (D.A.M., B.A.B., D.D.B., J. Kuder, S.A.M., M.S.S.)
| |
Collapse
|
7
|
de Sousa NF, Scotti L, de Moura ÉP, dos Santos Maia M, Soares Rodrigues GC, de Medeiros HIR, Lopes SM, Scotti MT. Computer Aided Drug Design Methodologies with Natural Products in the Drug Research Against Alzheimer's Disease. Curr Neuropharmacol 2022; 20:857-885. [PMID: 34636299 PMCID: PMC9881095 DOI: 10.2174/1570159x19666211005145952] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 08/19/2021] [Accepted: 08/26/2021] [Indexed: 11/22/2022] Open
Abstract
Natural products are compounds isolated from plants that provide a variety of lead structures for the development of new drugs by the pharmaceutical industry. The interest in these substances increases because of their beneficial effects on human health. Alzheimer's disease (AD) affects occur in about 80% of individuals aged 65 years. AD, the most common cause of dementia in elderly people, is characterized by progressive neurodegenerative alterations, as decrease of cholinergic impulse, increased toxic effects caused by reactive oxygen species and the inflammatory process that the amyloid plaque participates. In silico studies is relevant in the process of drug discovery; through technological advances in the areas of structural characterization of molecules, computational science and molecular biology have contributed to the planning of new drugs used against neurodegenerative diseases. Considering the social impairment caused by an increased incidence of disease and that there is no chemotherapy treatment effective against AD; several compounds are studied. In the researches for effective neuroprotectants as potential treatments for Alzheimer's disease, natural products have been extensively studied in various AD models. This study aims to carry out a literature review with articles that address the in silico studies of natural products aimed at potential drugs against Alzheimer's disease (AD) in the period from 2015 to 2021.
Collapse
Affiliation(s)
- Natália Ferreira de Sousa
- Postgraduate Program of Natural and Synthetic Bioactive Products (PgPNSB), Health Sciences Center, Federal University of Paraíba, João Pessoa-PB, Brazil
| | - Luciana Scotti
- Postgraduate Program of Natural and Synthetic Bioactive Products (PgPNSB), Health Sciences Center, Federal University of Paraíba, João Pessoa-PB, Brazil;,Lauro Wanderley University Hospital (HULW), Health Sciences Center, Federal University of Paraíba, João Pessoa-PB, Brazil,Address correspondence to this author at the Health Sciences Center, Chemioinformatic Laboratory, Federal University of Paraíba, Paraíba, Brazil; E-mail:
| | - Érika Paiva de Moura
- Postgraduate Program of Natural and Synthetic Bioactive Products (PgPNSB), Health Sciences Center, Federal University of Paraíba, João Pessoa-PB, Brazil
| | - Mayara dos Santos Maia
- Postgraduate Program of Natural and Synthetic Bioactive Products (PgPNSB), Health Sciences Center, Federal University of Paraíba, João Pessoa-PB, Brazil
| | - Gabriela Cristina Soares Rodrigues
- Postgraduate Program of Natural and Synthetic Bioactive Products (PgPNSB), Health Sciences Center, Federal University of Paraíba, João Pessoa-PB, Brazil
| | - Herbert Igor Rodrigues de Medeiros
- Postgraduate Program of Natural and Synthetic Bioactive Products (PgPNSB), Health Sciences Center, Federal University of Paraíba, João Pessoa-PB, Brazil
| | - Simone Mendes Lopes
- Postgraduate Program of Natural and Synthetic Bioactive Products (PgPNSB), Health Sciences Center, Federal University of Paraíba, João Pessoa-PB, Brazil
| | - Marcus Tullius Scotti
- Lauro Wanderley University Hospital (HULW), Health Sciences Center, Federal University of Paraíba, João Pessoa-PB, Brazil
| |
Collapse
|
8
|
Abbasi-Habashi S, Jickling GC, Winship IR. Immune Modulation as a Key Mechanism for the Protective Effects of Remote Ischemic Conditioning After Stroke. Front Neurol 2021; 12:746486. [PMID: 34956045 PMCID: PMC8695500 DOI: 10.3389/fneur.2021.746486] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Accepted: 11/09/2021] [Indexed: 12/12/2022] Open
Abstract
Remote ischemic conditioning (RIC), which involves a series of short cycles of ischemia in an organ remote to the brain (typically the limbs), has been shown to protect the ischemic penumbra after stroke and reduce ischemia/reperfusion (IR) injury. Although the exact mechanism by which this protective signal is transferred from the remote site to the brain remains unclear, preclinical studies suggest that the mechanisms of RIC involve a combination of circulating humoral factors and neuronal signals. An improved understanding of these mechanisms will facilitate translation to more effective treatment strategies in clinical settings. In this review, we will discuss potential protective mechanisms in the brain and cerebral vasculature associated with RIC. We will discuss a putative role of the immune system and circulating mediators of inflammation in these protective processes, including the expression of pro-and anti-inflammatory genes in peripheral immune cells that may influence the outcome. We will also review the potential role of extracellular vesicles (EVs), biological vectors capable of delivering cell-specific cargo such as proteins and miRNAs to cells, in modulating the protective effects of RIC in the brain and vasculature.
Collapse
Affiliation(s)
- Sima Abbasi-Habashi
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada
| | - Glen C Jickling
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada
- Division of Neurology, Faculty of Medicine, University of Alberta, Edmonton, AB, Canada
| | - Ian R Winship
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada
- Neurochemical Research Unit, Department of Psychiatry, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
9
|
Berg PC, Hansson ÅML, Røsand Ø, Marwarha G, Høydal MA. Overexpression of Neuron-Derived Orphan Receptor 1 (NOR-1) Rescues Cardiomyocytes from Cell Death and Improves Viability after Doxorubicin Induced Stress. Biomedicines 2021; 9:1233. [PMID: 34572418 PMCID: PMC8471245 DOI: 10.3390/biomedicines9091233] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 09/07/2021] [Accepted: 09/12/2021] [Indexed: 12/16/2022] Open
Abstract
Following myocardial infarction, reperfusion injury (RI) is commonly observed due to the excessive formation of, e.g., reactive oxygen species (ROS). Doxorubicin (DOX), a widely used anti-cancer drug, is also known to cause cardiotoxicity due to excessive ROS production. Exercise training has been shown to protect the heart against both RI- and DOX-induced cardiotoxicity, but the exact mechanism is still unknown. Neuron-derived orphan receptor 1 (NOR-1) is an important exercise-responsive protein in the skeletal muscle which has also been reported to facilitate cellular survival during hypoxia. Therefore, we hypothesized that NOR-1 could protect cardiomyocytes (CMs) against cellular stress induced by DOX. We also hypothesized that NOR-1 is involved in preparing the CMs against a stress situation during nonstimulated conditions by increasing cell viability. To determine the protective effect of NOR-1 in CMs stressed with DOX challenge, we overexpressed NOR-1 in AC16 human CMs treated with 5 µM DOX for 12 h or the respective vehicle control, followed by performing Lactate dehydrogenase (LDH) activity, 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT), and caspase-3 activity assays to measure cell death, cell viability, and apoptosis, respectively. In addition, Western blotting analysis was performed to determine the expression of key proteins involved in cardioprotection. We demonstrated that NOR-1 overexpression decreased cell death (p < 0.105) and apoptosis (p < 0.01) while increasing cell viability (p < 0.05) in DOX-treated CMs. We also observed that NOR-1 overexpression increased phosphorylation of extracellular signal-regulated kinase (ERK) (p < 0.01) and protein expression levels of B cell lymphoma extra-large (Bcl-xL) (p < 0.01). We did not detect any significant changes in phosphorylation of protein kinase B (Akt), glycogen synthase kinase-3β (GSK-3β) and signal transducer and activator of transcription 3 (STAT3) or expression levels of superoxide dismutase 2 (SOD2) and cyclin D1. Furthermore, we demonstrated that NOR-1 overexpression increased the cell viability (p < 0.0001) of CMs during nonstimulated conditions without affecting cell death or apoptosis. Our findings indicate that NOR-1 could serve as a potential cardioprotective protein in response to Doxorubicin-induced cellular stress.
Collapse
Affiliation(s)
| | | | | | | | - Morten Andre Høydal
- Group of Molecular and Cellular Cardiology, Department of Circulation and Medical Imaging, Faculty of Medicine and Health, Norwegian University of Technology and Science (NTNU), 7030 Trondheim, Norway; (P.-C.B.); (Å.M.L.H.); (Ø.R.); (G.M.)
| |
Collapse
|
10
|
George RT, Abuhatzira L, Stoughton SM, Karathanasis SK, She D, Jin C, Buss NAPS, Bakker‐Arkema R, Ongstad EL, Koren M, Hirshberg B. MEDI6012: Recombinant Human Lecithin Cholesterol Acyltransferase, High-Density Lipoprotein, and Low-Density Lipoprotein Receptor-Mediated Reverse Cholesterol Transport. J Am Heart Assoc 2021; 10:e014572. [PMID: 34121413 PMCID: PMC8403308 DOI: 10.1161/jaha.119.014572] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Accepted: 04/12/2021] [Indexed: 02/07/2023]
Abstract
Background MEDI6012 is recombinant human lecithin cholesterol acyltransferase, the rate-limiting enzyme in reverse cholesterol transport. Infusions of lecithin cholesterol acyltransferase have the potential to enhance reverse cholesterol transport and benefit patients with coronary heart disease. The purpose of this study was to test the safety, pharmacokinetic, and pharmacodynamic profile of MEDI6012. Methods and Results This phase 2a double-blind study randomized 48 subjects with stable coronary heart disease on a statin to a single dose of MEDI6012 or placebo (6:2) (NCT02601560) with ascending doses administered intravenously (24, 80, 240, and 800 mg) and subcutaneously (80 and 600 mg). MEDI6012 demonstrated rates of treatment-emergent adverse events that were similar to those of placebo. Dose-dependent increases in high-density lipoprotein cholesterol were observed with area under the concentration-time curves from 0 to 96 hours of 728, 1640, 3035, and 5318 should be: mg·h/mL in the intravenous dose groups and 422 and 2845 mg·h/mL in the subcutaneous dose groups. Peak mean high-density lipoprotein cholesterol percent change was 31.4%, 71.4%, 125%, and 177.8% in the intravenous dose groups and 18.3% and 111.2% in the subcutaneous dose groups, and was accompanied by increases in endogenous apoA1 (apolipoprotein A1) and non-ATP-binding cassette transporter A1 cholesterol efflux capacity. Decreases in apoB (apolipoprotein B) were observed across all dose levels and decreases in atherogenic small low-density lipoprotein particles by 41%, 88%, and 79% at the 80-, 240-, and 800-mg IV doses, respectively. Conclusions MEDI6012 demonstrated an acceptable safety profile and increased high-density lipoprotein cholesterol, endogenous apoA1, and non-ATP-binding cassette transporter A1 cholesterol efflux capacity while reducing the number of atherogenic low-density lipoprotein particles. These findings are supportive of enhanced reverse cholesterol transport and a functional high-density lipoprotein phenotype. Registration URL: https://www.clinicaltrials.gov; Unique identifier: NCT02601560.
Collapse
Affiliation(s)
- Richard T. George
- Early Clinical DevelopmentResearch and Early DevelopmentCardiovascular, Renal and MetabolismBioPharmaceuticals R&DAstraZenecaGaithersburgMD
| | - Liron Abuhatzira
- Early Clinical DevelopmentResearch and Early DevelopmentCardiovascular, Renal and MetabolismBioPharmaceuticals R&DAstraZenecaGaithersburgMD
| | - Susan M. Stoughton
- Early Clinical DevelopmentResearch and Early DevelopmentCardiovascular, Renal and MetabolismBioPharmaceuticals R&DAstraZenecaGaithersburgMD
| | - Sotirios K. Karathanasis
- BioscienceResearch and Early DevelopmentCardiovascular, Renal and MetabolismBioPharmaceuticals R&DAstraZenecaGaithersburgMD
| | - Dewei She
- Early CVRM BiometricsResearch and Early DevelopmentCardiovascular, Renal and MetabolismBioPharmaceuticals R&DAstraZenecaGaithersburgMD
| | - ChaoYu Jin
- Integrated BioanalysisClinical Pharmacology and Quantitative PharmacologyClinical Pharmacology & Safety SciencesR&DAstraZenecaSouth San FranciscoCA
| | - Nicholas A. P. S. Buss
- Cardiovascular, Renal and Metabolism SafetyClinical Pharmacology & Safety SciencesR&DAstraZenecaGaithersburgMD
| | | | - Emily L. Ongstad
- BioscienceResearch and Early DevelopmentCardiovascular, Renal and MetabolismBioPharmaceuticals R&DAstraZenecaGaithersburgMD
| | - Michael Koren
- Jacksonville Center for Clinical ResearchJacksonvilleFL
| | - Boaz Hirshberg
- Early Clinical DevelopmentResearch and Early DevelopmentCardiovascular, Renal and MetabolismBioPharmaceuticals R&DAstraZenecaGaithersburgMD
| |
Collapse
|
11
|
Wang H, Pang W, Xu X, You B, Zhang C, Li D. Cryptotanshinone Attenuates Ischemia/Reperfusion-induced Apoptosis in Myocardium by Upregulating MAPK3. J Cardiovasc Pharmacol 2021; 77:370-377. [PMID: 33662979 DOI: 10.1097/fjc.0000000000000971] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Accepted: 11/26/2020] [Indexed: 01/17/2023]
Abstract
ABSTRACT Chinese people have used the root of Salvia miltiorrhiza Bunge (called "Danshen" in Chinese) for centuries as an anticancer agent, anti-inflammatory agent, antioxidant, and cardiovascular disease drug. In addition, Danshen is considered to be a drug that can improve ischemia/reperfusion (I/R)-induced myocardium injury in traditional Chinese medicine. However, Danshen is a mixture that includes various bioactive substances. In this study, we aimed to identify the protective component and mechanism of Danshen on myocardium through network pharmacology and molecular simulation methods. First, cryptotanshinone (CTS) was identified as a potential active compound from Danshen that was associated with apoptosis by a network pharmacology approach. Subsequently, biological experiments validated that CTS inhibited ischemia/reperfusion-induced cardiomyocyte apoptosis in vivo and in vitro. Molecular docking techniques were used to screen key target information. Based on the simulative results, MAPKs were verified as well-connected molecules of CTS. Western blotting assays also demonstrated that CTS could enhance MAPK expression. Furthermore, we demonstrated that inhibition of the MAPK pathway reversed the CTS-mediated effect on cardiomyocyte apoptosis. Altogether, our work screened out CTS from Danshen and demonstrated that it protected cardiomyocytes from apoptosis.
Collapse
Affiliation(s)
- Hefeng Wang
- Department of Cardiology, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, Shandong, China
| | - Wenhui Pang
- Department of Otolaryngology Head and Neck Surgery, the Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Xingsheng Xu
- Department of Cardiology, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, Shandong, China
| | - Beian You
- Department of Cardiology, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, Shandong, China
| | - Cuijuan Zhang
- Department of Cardiology, the Affiliated Hospital of Qingdao University, Qingdao, Shandong, China; and
| | - Dan Li
- Department of Cardiology, the Affiliated Hospital of Qingdao University, Qingdao, Shandong, China; and
| |
Collapse
|
12
|
Bonaca MP, George RT, Morrow DA, Bergmark BA, Park JG, Abuhatzira L, Vavere AL, Karathanasis SK, Jin C, She D, Hirshberg B, Hsia J, Sabatine MS. Recombinant human Lecithin-Cholesterol acyltransferase in patients with atherosclerosis: Phase 2a primary results and phase 2b design. EUROPEAN HEART JOURNAL. CARDIOVASCULAR PHARMACOTHERAPY 2021; 8:243-252. [PMID: 33493256 DOI: 10.1093/ehjcvp/pvab001] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 12/09/2020] [Accepted: 01/05/2021] [Indexed: 01/30/2023]
Abstract
BACKGROUND Reverse cholesterol transport (RCT) removes cholesterol and stabilizes vulnerable plaques. In addition, high-density lipoprotein (HDL) may be cardioprotective in acute MI. Lecithin-cholesterol acyltransferase (LCAT) may enhance RCT. The objective of this study was to investigate the pharmacokinetics, pharmacodynamics, and safety of multiple ascending doses of recombinant human LCAT (MEDI6012) to inform a Phase 2 b program. METHODS This was a randomized, blinded, placebo-controlled, dose-escalation Phase 2a study of MEDI6012. Patients were randomized into 1 of 4 cohorts (40, 120, 300 mg IV weekly x3 doses, or 300 mg IV-push, 150 mg at 48-hours and 100 mg at 7 days). All cohorts were planned to randomize 6:2 (MEDI6012 vs placebo). The primary endpoints were baseline-adjusted AUC from 0-96 hours post dose-3 (AUC0-96hr) for HDL-C, HDL cholesteryl ester (HDL-CE), and total cholesteryl ester (CE). The primary safety endpoints were treatment-emergent adverse events (AEs). RESULTS A total of 32 patients were randomized. MEDI6012 significantly increased AUC0-96hr for HDL-C, HDL-CE and CE in a graded fashion with increasing doses. Relative to placebo, MEDI6012 increased HDL-C at Day 19 by 66% (95%CI 33-99, p = 0.014) with 120 mg and 144% (95%CI 108-181, p < 0.001) with 300 mg. An IV-push increased HDL-C by 40.8% at 30 minutes. Overall AEs were similar between groups with no severe, life-threatening/fatal AEs or neutralizing antibodies. CONCLUSIONS Multiple ascending doses of MEDI6012 were safe and well tolerated and significantly increased HDL-C, HDL-CE and CE in a dose-related manner. These data support the ongoing Phase 2 b program investigating MEDI6012 in ST-elevation MI.
Collapse
Affiliation(s)
- Marc P Bonaca
- CPC Clinical Research, Department of Medicine, University of Colorado Anschutz School of Medicine
| | - Richard T George
- Early Clinical Development, Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | - David A Morrow
- TIMI Study Group, Brigham and Women's Hospital, Harvard Medical School
| | - Brian A Bergmark
- TIMI Study Group, Brigham and Women's Hospital, Harvard Medical School
| | - Jeong-Gun Park
- TIMI Study Group, Brigham and Women's Hospital, Harvard Medical School
| | - Liron Abuhatzira
- Early Clinical Development, Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Andrea L Vavere
- Early Clinical Development, Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Sotirios K Karathanasis
- Bioscience, Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | - ChaoYu Jin
- Integrated Bioanalysis, Clinical Pharmacology and Quantitative Pharmacology, Clinical Pharmacology & Safety Sciences, R&D, AstraZeneca, 121 Oyster Point Boulevard, South San Francisco, CA 94080
| | - Dewei She
- Early CVRM Biometrics, Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Boaz Hirshberg
- Early Clinical Development, Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Judy Hsia
- Early Clinical Development, Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Marc S Sabatine
- TIMI Study Group, Brigham and Women's Hospital, Harvard Medical School
| |
Collapse
|
13
|
Soppert J, Lehrke M, Marx N, Jankowski J, Noels H. Lipoproteins and lipids in cardiovascular disease: from mechanistic insights to therapeutic targeting. Adv Drug Deliv Rev 2020; 159:4-33. [PMID: 32730849 DOI: 10.1016/j.addr.2020.07.019] [Citation(s) in RCA: 141] [Impact Index Per Article: 35.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 07/20/2020] [Accepted: 07/22/2020] [Indexed: 12/12/2022]
Abstract
With cardiovascular disease being the leading cause of morbidity and mortality worldwide, effective and cost-efficient therapies to reduce cardiovascular risk are highly needed. Lipids and lipoprotein particles crucially contribute to atherosclerosis as underlying pathology of cardiovascular disease and influence inflammatory processes as well as function of leukocytes, vascular and cardiac cells, thereby impacting on vessels and heart. Statins form the first-line therapy with the aim to block cholesterol synthesis, but additional lipid-lowering drugs are sometimes needed to achieve low-density lipoprotein (LDL) cholesterol target values. Furthermore, beyond LDL cholesterol, also other lipid mediators contribute to cardiovascular risk. This review comprehensively discusses low- and high-density lipoprotein cholesterol, lipoprotein (a), triglycerides as well as fatty acids and derivatives in the context of cardiovascular disease, providing mechanistic insights into their role in pathological processes impacting on cardiovascular disease. Also, an overview of applied as well as emerging therapeutic strategies to reduce lipid-induced cardiovascular burden is provided.
Collapse
Affiliation(s)
- Josefin Soppert
- Institute for Molecular Cardiovascular Research (IMCAR), University Hospital Aachen, Aachen, Germany
| | - Michael Lehrke
- Medical Clinic I, University Hospital Aachen, Aachen, Germany
| | - Nikolaus Marx
- Medical Clinic I, University Hospital Aachen, Aachen, Germany
| | - Joachim Jankowski
- Institute for Molecular Cardiovascular Research (IMCAR), University Hospital Aachen, Aachen, Germany; Department of Pathology, Cardiovascular Research Institute Maastricht, Maastricht University Medical Centre, Maastricht University, the Netherlands
| | - Heidi Noels
- Institute for Molecular Cardiovascular Research (IMCAR), University Hospital Aachen, Aachen, Germany; Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University, the Netherlands.
| |
Collapse
|
14
|
Sawashita Y, Hirata N, Yoshikawa Y, Terada H, Tokinaga Y, Yamakage M. Remote ischemic preconditioning reduces myocardial ischemia-reperfusion injury through unacylated ghrelin-induced activation of the JAK/STAT pathway. Basic Res Cardiol 2020; 115:50. [PMID: 32607622 DOI: 10.1007/s00395-020-0809-z] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Accepted: 06/24/2020] [Indexed: 01/06/2023]
Abstract
Remote ischemic preconditioning (RIPC) offers cardioprotection against myocardial ischemia-reperfusion injury. The humoral factors involved in RIPC that are released from parasympathetically innervated organs have not been identified. Previous studies showed that ghrelin, a hormone released from the stomach, is associated with cardioprotection. However, it is unknown whether or not ghrelin is involved in the mechanism of RIPC. This study aimed to determine whether ghrelin serves as one of the humoral factors in RIPC. RIPC group rats were subjected to three cycles of ischemia and reperfusion for 5 min in two limbs before left anterior descending (LAD) coronary artery ligation. Unacylated ghrelin (UAG) group rats were given 0.5 mcg/kg UAG intravenously 30 min before LAD ligation. Plasma levels of UAG in all groups were measured before and after RIPC procedures and UAG administration. Additionally, JAK2/STAT3 pathway inhibitor (AG490) was injected in RIPC and UAG groups to investigate abolishment of the cardioprotection of RIPC and UAG. Plasma levels of UAG, infarct size and phosphorylation of STAT3 were compared in all groups. Infarct size was significantly reduced in RIPC and UAG groups, compared to the other groups. Plasma levels of UAG in RIPC and UAG groups were significantly increased after RIPC and UAG administration, respectively. The cardioprotective effects of RIPC and UAG were accompanied by an increase in phosphorylation of STAT3 and abolished by AG490. This study indicated that RIPC reduces myocardial ischemia and reperfusion injury through UAG-induced activation of JAK/STAT pathway. UAG may be one of the humoral factors involved in the cardioprotective effects of RIPC.
Collapse
Affiliation(s)
- Yasuaki Sawashita
- Department of Anesthesiology, Sapporo Medical University School of Medicine, S-1, W-16, Chuo-ku, Sapporo, Hokkaido, 060-8543, Japan.
| | - Naoyuki Hirata
- Department of Anesthesiology, Sapporo Medical University School of Medicine, S-1, W-16, Chuo-ku, Sapporo, Hokkaido, 060-8543, Japan
| | - Yusuke Yoshikawa
- Department of Anesthesiology, Sapporo Medical University School of Medicine, S-1, W-16, Chuo-ku, Sapporo, Hokkaido, 060-8543, Japan
| | - Hirofumi Terada
- Department of Anesthesiology, Sapporo Medical University School of Medicine, S-1, W-16, Chuo-ku, Sapporo, Hokkaido, 060-8543, Japan
| | - Yasuyuki Tokinaga
- Department of Anesthesiology, Sapporo Medical University School of Medicine, S-1, W-16, Chuo-ku, Sapporo, Hokkaido, 060-8543, Japan
| | - Michiaki Yamakage
- Department of Anesthesiology, Sapporo Medical University School of Medicine, S-1, W-16, Chuo-ku, Sapporo, Hokkaido, 060-8543, Japan
| |
Collapse
|
15
|
Maciel L, de Oliveira DF, Monnerat G, Campos de Carvalho AC, Nascimento JHM. Exogenous 10 kDa-Heat Shock Protein Preserves Mitochondrial Function After Hypoxia/Reoxygenation. Front Pharmacol 2020; 11:545. [PMID: 32431608 PMCID: PMC7214810 DOI: 10.3389/fphar.2020.00545] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Accepted: 04/09/2020] [Indexed: 02/05/2023] Open
Abstract
Humoral factors released during ischemic preconditioning (IPC) protect the myocardium against ischemia/reperfusion (I/R) injury. We have recently identified 10 kDa-heat shock protein (HSP10) and a fraction of small 5-10 kDa peptides (5-10-sP) in the coronary effluent of IPC-treated hearts and demonstrated their cardioprotective potential. We here used our isolated mitochondria model to characterize the impact of exogenous HSP10 and 5-10-sP on mitochondria function from myocardium subjected to I/R injury. Isolated perfused rat hearts were submitted to 30-min global ischemia and 10-min reperfusion. Before ischemia, isolated hearts were infused with saline or 5-10-sP, with or without a mitochondrial ATP-sensitive-K+-channel blocker (5HD 10 μmol·L-1) or PKC inhibitor (chelerythrine 10 μmol·L-1), before I/R. HSP10 (1 µmol·L-1) was infused into isolated hearts before I/R without blockers. At 10-min reperfusion, the mitochondria were isolated and mitochondrial function was assessed. In a subset of experiments, freshly isolated mitochondria were directly incubated with HSP10 or 5-10-sP with or without 5HD or chelerythrine before in vitro hypoxia/reoxygenation. Infusion of 5-10-sP (n = 5) and HSP10 (n = 5) into isolated hearts before I/R improved mitochondrial ADP-stimulated respiration, ATP production and prevented mitochondrial ROS formation compared to the I/R group (n = 5); this effect was abrogated by 5HD and chelerythrine. In freshly isolated mitochondria with in vitro hypoxia/reoxygenation, HSP10 (n = 16) and 5-10-sP (n = 16) incubation prevented reductions of mitochondrial ADP-stimulated respiration (91.5 ± 5.1 nmol O2/min/mg PTN), ATP production (250.1 ± 9.3 μmol ATP/200μg PTN), and prevented mitochondrial ROS production (219.7 ± 9.0 nmol H2O2/200μg PTN) induced by hypoxia/reoxygenation (n = 12, 51.5 ± 5.0 nmol O2/min/mg PTN; 187 ± 21.7 μmol ATP/200 μg PTN; 339.0 ± 14.3 nmol H2O2/200 μg PTN, p < 0.001, respectively). 5HD reduced the ADP-stimulated respiration in the HSP10 group (65.84 ± 3.3 nmol O2/min/mg PTN), ATP production (193.7 ± 12.1 μmol ATP/200μg PTN) and increased ROS in the 5-10-sP group (274.4 ± 21.7 nmol H2O2/200 μg PTN). Mitochondria are a target of the cardioprotection induced by 5-10-sP and HSP10. This protection is dependent of PKC and mKATP activation. HSP10 can act directly on mitochondria and protects against hypoxia/reoxygenation injury by mKATP activation.
Collapse
Affiliation(s)
- Leonardo Maciel
- Laboratory of Cardiac Electrophysiology Antônio Paes de Carvalho, Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- *Correspondence: Leonardo Maciel,
| | - Dahienne Ferreira de Oliveira
- Laboratory of Cardiac Electrophysiology Antônio Paes de Carvalho, Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Gustavo Monnerat
- Laboratory of Cardiac Electrophysiology Antônio Paes de Carvalho, Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- National Institute of Cardiology, Rio de Janeiro, Brazil
| | - Antonio Carlos Campos de Carvalho
- Laboratory of Cardiac Electrophysiology Antônio Paes de Carvalho, Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- National Institute of Cardiology, Rio de Janeiro, Brazil
| | - Jose Hamilton Matheus Nascimento
- Laboratory of Cardiac Electrophysiology Antônio Paes de Carvalho, Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
16
|
Corydon KK, Matchkov V, Fais R, Abramochkin D, Hedegaard ER, Comerma-Steffensen S, Simonsen U. Effect of ischemic preconditioning and a Kv7 channel blocker on cardiac ischemia-reperfusion injury in rats. Eur J Pharmacol 2019; 866:172820. [PMID: 31760069 DOI: 10.1016/j.ejphar.2019.172820] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Revised: 11/19/2019] [Accepted: 11/20/2019] [Indexed: 12/30/2022]
Abstract
Recently, we found cardioprotective effects of ischemic preconditioning (IPC), and from a blocker of KCNQ voltage-gated K+ channels (KV7), XE991 (10,10-bis(4-pyridinylmethyl)-9(10H)-anthracenone), in isolated rat hearts. The purpose of the present study was to investigate the cardiovascular effects of IPC and XE991 and whether they are cardioprotective in intact rats. In conscious rats, we measured the effect of the KV7 channel blocker XE991 on heart rate and blood pressure by use of telemetry. In anesthetized rats, cardiac ischemia was induced by occluding the left coronary artery, and the animals received IPC (2 × 5 min of occlusion), XE991, or a combination. After a 2 h reperfusion period, the hearts were excised, and the area at risk and infarct size were determined. In both anesthetized and conscious rats, XE991 increased blood pressure, and the highest dose (7.5 mg/kg) of XE991 also increased heart rate, and 44% of conscious rats died. XE991 induced marked changes in the electrocardiogram (e.g., increased PR interval and prolonged QTC interval) without changing cardiac action potentials. The infarct size to area at risk ratio was reduced from 53 ± 2% (n = 8) in the vehicle compared to 36 ± 3% in the IPC group (P < 0.05, n = 9). XE991 (0.75 mg/kg) treatment alone or on top of IPC failed to reduce myocardial infarct size. Similar to the effect in isolated hearts, locally applied IPC was cardioprotective in intact animals exposed to ischemia-reperfusion. Systemic administration of XE991 failed to protect the heart against ischemia-reperfusion injury suggesting effects on the autonomic nervous system counteracting the cardioprotection in intact animals.
Collapse
Affiliation(s)
- Krestine Kjeldsen Corydon
- Department of Biomedicine, Pulmonary and Cardiovascular Pharmacology and Physiology, Aarhus University, Wilhelm Meyers Allé 4, 8000, Aarhus C, Denmark
| | - Vladimir Matchkov
- Department of Biomedicine, Pulmonary and Cardiovascular Pharmacology and Physiology, Aarhus University, Wilhelm Meyers Allé 4, 8000, Aarhus C, Denmark
| | - Rafael Fais
- Department of Biomedicine, Pulmonary and Cardiovascular Pharmacology and Physiology, Aarhus University, Wilhelm Meyers Allé 4, 8000, Aarhus C, Denmark; Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, São Paulo, Brazil
| | - Denis Abramochkin
- Department of Human and Animal Physiology, Biological Faculty, Lomonosov Moscow State University, Leninskiye Gory, 1, 12, Moscow, Russia; Ural Federal University, Mira 19, Ekaterinburg, Russia; Department of Physiology, Russian National Research Medical University, Ostrovityanova 1, Moscow, Russia
| | - Elise Røge Hedegaard
- Department of Biomedicine, Pulmonary and Cardiovascular Pharmacology and Physiology, Aarhus University, Wilhelm Meyers Allé 4, 8000, Aarhus C, Denmark
| | - Simon Comerma-Steffensen
- Department of Biomedicine, Pulmonary and Cardiovascular Pharmacology and Physiology, Aarhus University, Wilhelm Meyers Allé 4, 8000, Aarhus C, Denmark; Department of Biomedical Sciences/Animal Physiology, Veterinary Faculty, Central University of Venezuela, Maracay, Aragua, Venezuela
| | - Ulf Simonsen
- Department of Biomedicine, Pulmonary and Cardiovascular Pharmacology and Physiology, Aarhus University, Wilhelm Meyers Allé 4, 8000, Aarhus C, Denmark.
| |
Collapse
|
17
|
Sposito AC, de Lima-Junior JC, Moura FA, Barreto J, Bonilha I, Santana M, Virginio VW, Sun L, Carvalho LSF, Soares AA, Nadruz W, Feinstein SB, Nofer JR, Zanotti I, Kontush A, Remaley AT. Reciprocal Multifaceted Interaction Between HDL (High-Density Lipoprotein) and Myocardial Infarction. Arterioscler Thromb Vasc Biol 2019; 39:1550-1564. [DOI: 10.1161/atvbaha.119.312880] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Despite decades of therapeutic advances, myocardial infarction remains a leading cause of death worldwide. Recent studies have identified HDLs (high-density lipoproteins) as a potential candidate for mitigating coronary ischemia/reperfusion injury via a broad spectrum of signaling pathways. HDL ligands, such as S1P (sphingosine-1-phosphate), Apo (apolipoprotein) A-I, clusterin, and miRNA, may influence the opening of the mitochondrial channel, insulin sensitivity, and production of vascular autacoids, such as NO, prostacyclin, and endothelin-1. In parallel, antioxidant activity and sequestration of oxidized molecules provided by HDL can attenuate the oxidative stress that triggers ischemia/reperfusion. Nevertheless, during myocardial infarction, oxidation and the capture of oxidized and proinflammatory molecules generate large phenotypic and functional changes in HDL, potentially limiting its beneficial properties. In this review, new findings from cellular and animal models, as well as from clinical studies, will be discussed to describe the cardioprotective benefits of HDL on myocardial infarction. Furthermore, mechanisms by which HDL modulates cardiac function and potential strategies to mitigate postmyocardial infarction risk damage by HDL will be detailed throughout the review.
Collapse
Affiliation(s)
- Andrei C. Sposito
- From the Atherosclerosis and Vascular Biology Laboratory, Cardiology Department, State University of Campinas, Brazil (A.C.S., J.C.d.L.-J., F.A.M., J.B., I.B., M.S., V.W.V., L.S.F.C., A.A.S.S., W.N.)
| | - José Carlos de Lima-Junior
- From the Atherosclerosis and Vascular Biology Laboratory, Cardiology Department, State University of Campinas, Brazil (A.C.S., J.C.d.L.-J., F.A.M., J.B., I.B., M.S., V.W.V., L.S.F.C., A.A.S.S., W.N.)
| | - Filipe A. Moura
- From the Atherosclerosis and Vascular Biology Laboratory, Cardiology Department, State University of Campinas, Brazil (A.C.S., J.C.d.L.-J., F.A.M., J.B., I.B., M.S., V.W.V., L.S.F.C., A.A.S.S., W.N.)
- Department of Medicine, Weill-Cornell Medical College, New York, NY (F.A.M.)
| | - Joaquim Barreto
- From the Atherosclerosis and Vascular Biology Laboratory, Cardiology Department, State University of Campinas, Brazil (A.C.S., J.C.d.L.-J., F.A.M., J.B., I.B., M.S., V.W.V., L.S.F.C., A.A.S.S., W.N.)
| | - Isabella Bonilha
- From the Atherosclerosis and Vascular Biology Laboratory, Cardiology Department, State University of Campinas, Brazil (A.C.S., J.C.d.L.-J., F.A.M., J.B., I.B., M.S., V.W.V., L.S.F.C., A.A.S.S., W.N.)
| | - Michele Santana
- From the Atherosclerosis and Vascular Biology Laboratory, Cardiology Department, State University of Campinas, Brazil (A.C.S., J.C.d.L.-J., F.A.M., J.B., I.B., M.S., V.W.V., L.S.F.C., A.A.S.S., W.N.)
| | - Vitor W. Virginio
- From the Atherosclerosis and Vascular Biology Laboratory, Cardiology Department, State University of Campinas, Brazil (A.C.S., J.C.d.L.-J., F.A.M., J.B., I.B., M.S., V.W.V., L.S.F.C., A.A.S.S., W.N.)
| | - Lufan Sun
- Lipoprotein Metabolism Section, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD (L.S., A.T.R.)
- Department of Cardiology, The First Hospital of China Medical University, Shenyang, Liaoning Province, China (L.S.)
| | - Luiz Sergio F. Carvalho
- From the Atherosclerosis and Vascular Biology Laboratory, Cardiology Department, State University of Campinas, Brazil (A.C.S., J.C.d.L.-J., F.A.M., J.B., I.B., M.S., V.W.V., L.S.F.C., A.A.S.S., W.N.)
| | - Alexandre A.S. Soares
- From the Atherosclerosis and Vascular Biology Laboratory, Cardiology Department, State University of Campinas, Brazil (A.C.S., J.C.d.L.-J., F.A.M., J.B., I.B., M.S., V.W.V., L.S.F.C., A.A.S.S., W.N.)
| | - Wilson Nadruz
- From the Atherosclerosis and Vascular Biology Laboratory, Cardiology Department, State University of Campinas, Brazil (A.C.S., J.C.d.L.-J., F.A.M., J.B., I.B., M.S., V.W.V., L.S.F.C., A.A.S.S., W.N.)
| | - Steve B. Feinstein
- Division of Cardiology, Rush University Medical Center, Chicago, IL (S.B.F.)
| | - Jerzy-Roch Nofer
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf, Germany (J.-R.N.)
| | - Ilaria Zanotti
- Department of Food and Drugs, University of Parma, Italy (I.Z.)
| | - Anatol Kontush
- UMR-ICAN 1166, National Institute for Health and Medical Research (INSERM), Sorbonne University, Paris, France (A.K.)
| | - Alan T. Remaley
- Lipoprotein Metabolism Section, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD (L.S., A.T.R.)
| |
Collapse
|
18
|
Leung CH, Caldarone CA, Guan R, Wen XY, Ailenberg M, Kapus A, Szaszi K, Rotstein OD. Nuclear Factor (Erythroid-Derived 2)-Like 2 Regulates the Hepatoprotective Effects of Remote Ischemic Conditioning in Hemorrhagic Shock. Antioxid Redox Signal 2019; 30:1760-1773. [PMID: 30403148 DOI: 10.1089/ars.2018.7541] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
AIMS Remote ischemic conditioning (RIC) protects against organ ischemia/reperfusion injury in experimental and clinical settings. We have demonstrated that RIC prevents liver and lung inflammation/injury after hemorrhagic shock/resuscitation (S/R). In this study, we used a murine model of S/R to investigate the role of nuclear factor (erythroid-derived 2)-like 2 (Nrf2) in mediating hepatoprotection. RESULTS The combination of RIC with S/R caused a synergistic rise in Nrf2 and its translocation to the nucleus in the liver. Increased activation of Nrf2 by RIC augmented heme oxygenase-1 (HO-1) and autophagy and exerted hepatoprotection, concurrent with reductions in S/R-induced TNF-α (tumor necrosis factor alpha) and IL-6 (interleukin-6). In Nrf2 knockout (KO) animals, RIC did not exert hepatoprotection, and it failed to upregulate HO-1 and autophagy. Further, resuscitating wildtype (WT) animals with blood from donor WT animals undergoing RIC was hepatoprotective, but not in Nrf2 KO recipient animals. Interestingly, RIC blood from Nrf2 KO donor animals was also not protective when used to resuscitate WT animals, suggesting a role for Nrf2 both in the afferent arm of RIC where protective factors are generated and also in the efferent arm where organ protection is exerted. Finally, RIC plasma prevented oxidant-induced zebrafish mortality, but not in Nrf2a morpholino knockdown fish. INNOVATION Activation of Nrf2 is an essential mechanism underlying the hepatoprotective effects of RIC. Nrf2 appears to play a role in the afferent limb of RIC protection, as its absence precludes the generation of the protective humoral factors induced by RIC. CONCLUSION Our studies demonstrate the critical role of Nrf2 in the ability of RIC to prevent organ injury after S/R.
Collapse
Affiliation(s)
- Chung Ho Leung
- 1 Department of Surgery, St. Michael's Hospital, Toronto, Ontario, Canada.,2 Department of Surgery, Hospital for Sick Children, Toronto, Ontario, Canada.,3 Department of Surgery, University of Toronto, Toronto, Ontario, Canada.,4 Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, Ontario, Canada
| | - Christopher A Caldarone
- 2 Department of Surgery, Hospital for Sick Children, Toronto, Ontario, Canada.,3 Department of Surgery, University of Toronto, Toronto, Ontario, Canada
| | - Rui Guan
- 1 Department of Surgery, St. Michael's Hospital, Toronto, Ontario, Canada.,4 Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, Ontario, Canada.,5 Zebrafish Centre for Advanced Drug Discovery, St. Michael's Hospital, Toronto, Ontario, Canada
| | - Xiao-Yan Wen
- 4 Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, Ontario, Canada.,5 Zebrafish Centre for Advanced Drug Discovery, St. Michael's Hospital, Toronto, Ontario, Canada
| | - Menachem Ailenberg
- 1 Department of Surgery, St. Michael's Hospital, Toronto, Ontario, Canada
| | - Andras Kapus
- 1 Department of Surgery, St. Michael's Hospital, Toronto, Ontario, Canada.,3 Department of Surgery, University of Toronto, Toronto, Ontario, Canada.,4 Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, Ontario, Canada
| | - Katalin Szaszi
- 1 Department of Surgery, St. Michael's Hospital, Toronto, Ontario, Canada.,3 Department of Surgery, University of Toronto, Toronto, Ontario, Canada.,4 Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, Ontario, Canada
| | - Ori D Rotstein
- 1 Department of Surgery, St. Michael's Hospital, Toronto, Ontario, Canada.,3 Department of Surgery, University of Toronto, Toronto, Ontario, Canada.,4 Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, Ontario, Canada.,5 Zebrafish Centre for Advanced Drug Discovery, St. Michael's Hospital, Toronto, Ontario, Canada
| |
Collapse
|
19
|
Tsibulnikov SY, Maslov LN, Gorbunov AS, Voronkov NS, Boshchenko AA, Popov SV, Prokudina ES, Singh N, Downey JM. A Review of Humoral Factors in Remote Preconditioning of the Heart. J Cardiovasc Pharmacol Ther 2019; 24:403-421. [PMID: 31035796 DOI: 10.1177/1074248419841632] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
A humoral mechanism of cardioprotection by remote ischemic preconditioning (RIP) has been clearly demonstrated in various models of ischemia-reperfusion including upper and lower extremities, liver, and the mesenteric and renal arteries. A wide range of humoral factors for RIP have been proposed including hydrophobic peptides, opioid peptides, adenosine, prostanoids, endovanilloids, endocannabinoids, calcitonin gene-related peptide, leukotrienes, noradrenaline, adrenomedullin, erythropoietin, apolipoprotein, A-I glucagon-like peptide-1, interleukin 10, stromal cell-derived factor 1, and microRNAs. Virtually, all of the components of ischemic preconditioning's signaling pathway such as nitric oxide synthase, protein kinase C, redox signaling, PI3-kinase/Akt, glycogen synthase kinase β, ERK1/2, mitoKATP channels, Connexin 43, and STAT were all found to play a role. The signaling pattern also depends on which remote vascular bed was subjected to ischemia and on the time between applying the rip and myocardial ischemia occurs. Because there is convincing evidence for many seemingly diverse humoral components in RIP, the most likely explanation is that the overall mechanism is complex like that seen in ischemic preconditioning where multiple components are both in series and in parallel and interact with each other. Inhibition of any single component in the right circumstance may block the resulting protective effect, and selectively activating that component may trigger the protection. Identifying the humoral factors responsible for RIP might be useful in developing drugs that confer RIP's protection in a more comfortable and reliable manner.
Collapse
Affiliation(s)
- Sergey Y Tsibulnikov
- 1 Cardiology Research Institute, Tomsk National Research Medical Center of the Russian Academy of Science, Tomsk, Russia
| | - Leonid N Maslov
- 1 Cardiology Research Institute, Tomsk National Research Medical Center of the Russian Academy of Science, Tomsk, Russia
| | - Alexander S Gorbunov
- 1 Cardiology Research Institute, Tomsk National Research Medical Center of the Russian Academy of Science, Tomsk, Russia
| | - Nikita S Voronkov
- 1 Cardiology Research Institute, Tomsk National Research Medical Center of the Russian Academy of Science, Tomsk, Russia
| | - Alla A Boshchenko
- 1 Cardiology Research Institute, Tomsk National Research Medical Center of the Russian Academy of Science, Tomsk, Russia
| | - Sergey V Popov
- 1 Cardiology Research Institute, Tomsk National Research Medical Center of the Russian Academy of Science, Tomsk, Russia
| | - Ekaterina S Prokudina
- 1 Cardiology Research Institute, Tomsk National Research Medical Center of the Russian Academy of Science, Tomsk, Russia
| | - Nirmal Singh
- 2 Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, India
| | - James M Downey
- 3 Department of Physiology and Cell Biology, College of Medicine, University of South Alabama, Mobile, AL, USA
| |
Collapse
|
20
|
Yang CF. Clinical manifestations and basic mechanisms of myocardial ischemia/reperfusion injury. Tzu Chi Med J 2018; 30:209-215. [PMID: 30305783 PMCID: PMC6172894 DOI: 10.4103/tcmj.tcmj_33_18] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Revised: 03/01/2018] [Accepted: 03/12/2018] [Indexed: 12/21/2022] Open
Abstract
Acute myocardial ischemia/reperfusion (I/R) injury is a significant, unsolved clinical puzzle. In the disease context of acute myocardial infarction, reperfusion remains the only effective strategy to salvage ischemic myocardium, but it also causes additional damage. Myocardial I/R injury is composed of four types of damage, and these events attenuate the benefits of reperfusion therapy. Thus, inventing new strategies to conquer I/R injury is an unmet clinical need. A variety of pathological processes and mediators, including changes in the pH, generation of reactive oxygen radicals, and intracellular calcium overload, are proposed to be crucial in I/R-related cell injury. Among the intracellular events that occur during I/R, we stress the importance of protein phosphorylation signaling and elaborate its regulation. A variety of protein kinase pathways could be activated in I/R, including reperfusion injury salvage kinase and survivor-activating factor enhancement pathways, which are critical to cardiomyocyte survival. In addition to serine/threonine phosphorylation signaling, protein tyrosine phosphorylation is also critical in multiple cell functions and survival. However, the roles of protein kinases and phosphatases in I/R have not been extensively studied yet. By better understanding the mechanisms of I/R injury, we may have a better chance to develop new strategies for I/R injury and apply them in the clinical patient care.
Collapse
Affiliation(s)
- Chiu-Fen Yang
- Department of Cardiology, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
- Doctoral Degree Program in Translation Medicine, Tzu Chi University and Academia Sinica, Hualien, Taiwan
- Institute of Biological Chemistry, Academia Sinica, Taipei, Taiwan
| |
Collapse
|
21
|
Zuo YH, Han QB, Dong GT, Yue RQ, Ren XC, Liu JX, Liu L, Luo P, Zhou H. Panax ginseng Polysaccharide Protected H9c2 Cardiomyocyte From Hypoxia/Reoxygenation Injury Through Regulating Mitochondrial Metabolism and RISK Pathway. Front Physiol 2018; 9:699. [PMID: 29962955 PMCID: PMC6013582 DOI: 10.3389/fphys.2018.00699] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Accepted: 05/18/2018] [Indexed: 12/13/2022] Open
Abstract
Background and Objective: Ischemic heart disease (IHD) has been the major issue of public health. Panax ginseng (ginseng) has been verified as an effective traditional Chinese medicines and exerted cardioprotective effect. This study aimed to investigate the polysaccharide fraction of ginseng on hypoxia/reoxygenation (H/R) injury in cardiomyocytes and the underlying mechanisms. Methods: Ginseng was extracted by ethanol and fractionated by high-speed counter current chromatography (HSCCC) and column separation. The cardioprotective effect was evaluated in H9c2 cardiomyocytes underwent H/R treatment. The cell viability, apoptosis and mitochondrial respiration were examined. Results: An acid polysaccharides fraction of ginseng (AP1) was identified the most effective fraction in protecting cardiomyocytes from H/R injury. AP1 restored the mitochondrial function by maintaining mitochondrial membrane potential (MMP), blocking the release of cytochrome C, and increasing the ATP generation and oxygen consumption rate (OCR) of cardiomyocytes. Meanwhile, AP1 induced the expression of glucocorticoid receptor (GR) and estrogen receptor (ER) which further activated reperfusion injury salvage kinase (RISK) pathway. Finally, AP1 increased nitric oxide (NO) production and regulated endothelial function by increasing endothelial NO synthase (eNOS) expression and decreasing inducible NOS (iNOS) expression in H/R injury. Conclusion: The results suggested that AP1 exerted a protective effect in myocardial H/R injury mainly through maintaining myocardial mitochondrial function, thereby inhibiting myocardial H/R caused apoptosis and increasing the expressions of GR and ER, which in turn mediated the activation of RISK pathway and eNOS-dependent mechanism to resist the reperfusion injury.
Collapse
Affiliation(s)
- Yi-Han Zuo
- State Key Laboratory of Quality Research in Chinese Medicine, Faculty of Chinese Medicine, Macau University of Science and Technology, Macau, China
| | - Quan-Bin Han
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China
| | - Geng-Ting Dong
- State Key Laboratory of Quality Research in Chinese Medicine, Faculty of Chinese Medicine, Macau University of Science and Technology, Macau, China
| | - Rui-Qi Yue
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China
| | - Xue-Cong Ren
- State Key Laboratory of Quality Research in Chinese Medicine, Faculty of Chinese Medicine, Macau University of Science and Technology, Macau, China
| | - Jian-Xin Liu
- State Key Laboratory of Quality Research in Chinese Medicine, Faculty of Chinese Medicine, Macau University of Science and Technology, Macau, China.,College of Pharmacy, Hunan University of Medicine, Huaihua, China
| | - Liang Liu
- State Key Laboratory of Quality Research in Chinese Medicine, Faculty of Chinese Medicine, Macau University of Science and Technology, Macau, China
| | - Pei Luo
- State Key Laboratory of Quality Research in Chinese Medicine, Faculty of Chinese Medicine, Macau University of Science and Technology, Macau, China
| | - Hua Zhou
- State Key Laboratory of Quality Research in Chinese Medicine, Faculty of Chinese Medicine, Macau University of Science and Technology, Macau, China.,International Institute of Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
22
|
Chen H, Jing XY, Shen YJ, Wang TL, Ou C, Lu SF, Cai Y, Li Q, Chen X, Ding YJ, Yu XC, Zhu BM. Stat5-dependent cardioprotection in late remote ischaemia preconditioning. Cardiovasc Res 2018; 114:679-689. [DOI: 10.1093/cvr/cvy014] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Accepted: 01/19/2018] [Indexed: 02/05/2023] Open
Affiliation(s)
- Hui Chen
- Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Xianlin Road 138, Qixia Street, Nanjing, Jiangsu 210023, China
| | - Xin-Yue Jing
- Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Xianlin Road 138, Qixia Street, Nanjing, Jiangsu 210023, China
| | - Yu-Jun Shen
- Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Xianlin Road 138, Qixia Street, Nanjing, Jiangsu 210023, China
| | - Tian-Lin Wang
- Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Xianlin Road 138, Qixia Street, Nanjing, Jiangsu 210023, China
| | - Chen Ou
- Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Xianlin Road 138, Qixia Street, Nanjing, Jiangsu 210023, China
| | - Sheng-Feng Lu
- Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Xianlin Road 138, Qixia Street, Nanjing, Jiangsu 210023, China
| | - Yun Cai
- Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Xianlin Road 138, Qixia Street, Nanjing, Jiangsu 210023, China
| | - Qian Li
- Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Xianlin Road 138, Qixia Street, Nanjing, Jiangsu 210023, China
| | - Xia Chen
- Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Xianlin Road 138, Qixia Street, Nanjing, Jiangsu 210023, China
| | - Ya-Juan Ding
- Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Xianlin Road 138, Qixia Street, Nanjing, Jiangsu 210023, China
| | - Xiao-Chun Yu
- Institute of Acupuncture and Moxibustion, China Academy of Chinese Medical Sciences, Nanxiaojie 16, Dongzhimennei, Beijing, 100700, China
| | - Bing-Mei Zhu
- Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Xianlin Road 138, Qixia Street, Nanjing, Jiangsu 210023, China
- Regenerative Medicine Research Center, West China Hospital, Sichuan University, Keyuan Road 4, Gaopeng Street, Chengdu, Sichuan 610041, PR China
| |
Collapse
|
23
|
The SAFE pathway for cardioprotection: is this a promising target? Basic Res Cardiol 2018; 113:9. [DOI: 10.1007/s00395-018-0670-5] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Revised: 12/21/2017] [Accepted: 01/11/2018] [Indexed: 02/08/2023]
|
24
|
In Silico Studies Applied to Natural Products with Potential Activity Against Alzheimer’s Disease. NEUROMETHODS 2018. [DOI: 10.1007/978-1-4939-7404-7_18] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/11/2023]
|
25
|
Fang R, Zhang LL, Zhang LZ, Li W, Li M, Wen K. Sphingosine 1-Phosphate Postconditioning Protects Against Myocardial Ischemia/reperfusion Injury in Rats via Mitochondrial Signaling and Akt-Gsk3β Phosphorylation. Arch Med Res 2017. [PMID: 28625317 DOI: 10.1016/j.arcmed.2017.03.013] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
BACKGROUND AND AIMS Although preconditioning of sphingosine 1-phosphate (S1P) has been shown to protect myocytes from hypoxia reoxgenation injury in vitro, the role of S1P postconditioning on myocardial ischemia reperfusion injury (MIRI) in vivo and its related mechanism are unknown. The aim of this study was to investigate the protective role of sphingosine 1-phosphate (S1P) postconditioning in MIRI via its effects on mitochondrial signaling and Akt/Gsk3β phosphorylation. METHODS Rats were subjected to MIRI, consisting of 30 min of ischemia followed by 120 min of reperfusion, with S1P administered at the beginning of the reperfusion. Myocardial infarct size and apoptotic index were measured by triphenyltetrazolium (TTC) and terminal deoxynucleotide transferase dUTP nick-end labeling (TUNEL) assays, respectively. Akt and Gsk3β phosphorylation, caspase-3 cleavage, and cytochrome c translocation were assessed by western blot. Mitochondrial permeability transition pore (MPTP) opening and mitochondrial membrane potential (MMP, ΔΨ) were also examined to determine overall mitochondrial function. RESULTS S1P postconditioning significantly decreased myocardial infarct size and apoptosis, as well as enhanced Akt and Gsk3β phosphorylation, attenuated caspase-3 cleavage and cytosolic cytochrome c translocation, and inhibited MPTP opening, which subsequently preserved Δψ. Electron microscopy also confirmed that S1P helped maintain myocardial mitochondria integrity. Moreover, the protective effects of S1P treatment were blocked by cotreatment with a PI3K inhibitor, LY294002. CONCLUSIONS These results suggest that S1P postconditioning protects against MIRI by regulating mitochondrial signaling and Akt/Gsk3β phosphorylation.
Collapse
Affiliation(s)
- Rui Fang
- Department of Pharmacology, College of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Lu-Lu Zhang
- Department of Pharmacology, College of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Li-Zhi Zhang
- Department of Obstetrics and Gynecology, Tianjin First Centre Hospital, Tianjin, China
| | - Wenchang Li
- Department of Pharmacology, College of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Mengmeng Li
- Department of Pharmacology, College of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Ke Wen
- Department of Pharmacology, College of Basic Medical Sciences, Tianjin Medical University, Tianjin, China.
| |
Collapse
|
26
|
Remote tissue conditioning - An emerging approach for inducing body-wide protection against diseases of ageing. Ageing Res Rev 2017; 37:69-78. [PMID: 28552720 DOI: 10.1016/j.arr.2017.05.005] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Revised: 05/05/2017] [Accepted: 05/18/2017] [Indexed: 12/13/2022]
Abstract
We have long accepted that exercise is 'good for us'; that - put more rigorously - moderate exercise is associated with not just aerobic fitness but also reduced morbidity and reduced mortality from cardiovascular disease and even malignancies. Caloric restriction (moderate hunger) and our exposure to dietary phytochemicals are also emerging as stresses which are 'good for us' in the same sense. This review focuses on an important extension of this concept: that stress localized within the body (e.g. in a limb) can induce resilience in tissues throughout the body. We describe evidence for the efficacy of two 'remote' protective interventions - remote ischemic conditioning and remote photobiomodulation - and discuss the mechanisms underlying their protective actions. While the biological phenomenon of remote tissue conditioning is only partially understood, it holds promise for protecting critical-to-life tissues while mitigating risks and practical barriers to direct conditioning of these tissues.
Collapse
|
27
|
Cardioprotection by the transfer of coronary effluent from ischaemic preconditioned rat hearts: identification of cardioprotective humoral factors. Basic Res Cardiol 2017; 112:52. [PMID: 28695353 DOI: 10.1007/s00395-017-0641-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Accepted: 07/06/2017] [Indexed: 01/24/2023]
Abstract
Ischaemic preconditioning (IPC) provides myocardial resistance to ischaemia/reperfusion (I/R) injuries. The protection afforded by IPC is not limited to the target tissue but extends to remote tissues, suggesting a mechanism mediated by humoral factors. The aim of the present study was to identify the humoral factors that are responsible for the cardioprotection induced by the coronary effluent transferred from IPC to naïve hearts. Isolated rat hearts were submitted to IPC (three cycles of 5 min I/R) before 30-min global ischaemia and 60-min reperfusion. The coronary effluent (Efl_IPC) collected during IPC was fractionated by ultrafiltration in different molecular weight ranges (<3, 3-5, 5-10, 10-30, 30-50, and >50 kDa) and evaluated for cardioprotective effects by perfusion before I/R in naïve hearts. Only the <3, 5-10 and <10 kDa fractions of hydrophobic eluate reduced I/R injuries. The cardioprotective effect of the 5-10 fraction was blocked by KATP channel blockers and a PKC inhibitor. An Efl_IPC proteomic analysis revealed 14 cytoprotection-related proteins in 4-12 kDa peptides. HSP10 perfusion protected the heart against I/R injuries. These data provide insights into the mechanisms of cardioprotection in humoral factors released by IPC. Cardioprotection is afforded by hydrophobic peptides in the 4-12 kDa size range, which activate pathways that are dependent on PKC and KATP. Fourteen 4-12 kDa peptides were identified, suggesting a potential therapeutic role for these molecules in ischaemic diseases. One of these, HSP10, identified by mass spectrometry, reduced I/R injuries and may be a potential candidate as a therapeutic target.
Collapse
|
28
|
Reperfusion Therapy with Rapamycin Attenuates Myocardial Infarction through Activation of AKT and ERK. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2017; 2017:4619720. [PMID: 28373901 PMCID: PMC5360974 DOI: 10.1155/2017/4619720] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Revised: 01/26/2017] [Accepted: 02/14/2017] [Indexed: 01/04/2023]
Abstract
Prompt coronary reperfusion is the gold standard for minimizing injury following acute myocardial infarction. Rapamycin, mammalian target of Rapamycin (mTOR) inhibitor, exerts preconditioning-like cardioprotective effects against ischemia/reperfusion (I/R) injury. We hypothesized that Rapamycin, given at the onset of reperfusion, reduces myocardial infarct size through modulation of mTOR complexes. Adult C57 male mice were subjected to 30 min of myocardial ischemia followed by reperfusion for 1 hour/24 hours. Rapamycin (0.25 mg/kg) or DMSO (7.5%) was injected intracardially at the onset of reperfusion. Post-I/R survival (87%) and cardiac function (fractional shortening, FS: 28.63 ± 3.01%) were improved in Rapamycin-treated mice compared to DMSO (survival: 63%, FS: 17.4 ± 2.6%). Rapamycin caused significant reduction in myocardial infarct size (IS: 26.2 ± 2.2%) and apoptosis (2.87 ± 0.64%) as compared to DMSO-treated mice (IS: 47.0 ± 2.3%; apoptosis: 7.39 ± 0.81%). Rapamycin induced phosphorylation of AKT S473 (target of mTORC2) but abolished ribosomal protein S6 phosphorylation (target of mTORC1) after I/R. Rapamycin induced phosphorylation of ERK1/2 but inhibited p38 phosphorylation. Infarct-limiting effect of Rapamycin was abolished with ERK inhibitor, PD98059. Rapamycin also attenuated Bax and increased Bcl-2/Bax ratio. These results suggest that reperfusion therapy with Rapamycin protects the heart against I/R injury by selective activation of mTORC2 and ERK with concurrent inhibition of mTORC1 and p38.
Collapse
|
29
|
Kouassi Nzoughet J, Bocca C, Simard G, Prunier-Mirebeau D, Chao de la Barca JM, Bonneau D, Procaccio V, Prunier F, Lenaers G, Reynier P. A Nontargeted UHPLC-HRMS Metabolomics Pipeline for Metabolite Identification: Application to Cardiac Remote Ischemic Preconditioning. Anal Chem 2017; 89:2138-2146. [PMID: 27992159 DOI: 10.1021/acs.analchem.6b04912] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
In recent years, the number of investigations based on nontargeted metabolomics has increased, although often without a thorough assessment of analytical strategies applied to acquire data. Following published guidelines for metabolomics experiments, we report a validated nontargeted metabolomics strategy with pipeline for unequivocal identification of metabolites using the MSMLS molecule library. We achieved an in-house database containing accurate m/z values, retention times, isotopic patterns, full MS, and MS/MS spectra. A UHPLC-HRMS Q-Exactive method was developed, and experimental variations were determined within and between 3 experimental days. The extraction efficiency as well as the accuracy, precision, repeatability, and linearity of the method were assessed, the method demonstrating good performances. The methodology was further blindly applied to plasma from remote ischemic pre-conditioning (RIPC) rats. Samples, previously analyzed by targeted metabolomics using completely different protocol, analytical strategy, and platform, were submitted to our analytical pipeline. A combination of multivariate and univariate statistical analyses was employed. Selection of putative biomarkers from OPLS-DA model and S-plot was combined to jack-knife confidence intervals, metabolites' VIP values, and univariate statistics. Only variables with strong model contribution and highly statistical reliability were selected as discriminated metabolites. Three biomarkers identified by the previous targeted metabolomics study were found in the current work, in addition to three novel metabolites, emphasizing the efficiency of the current methodology and its ability to identify new biomarkers of clinical interest, in a single sequence. The biomarkers were identified to level 1 according to the metabolomics standard initiative and confirmed by both RPLC and HILIC-HRMS.
Collapse
Affiliation(s)
- Judith Kouassi Nzoughet
- PREMMi, Pôle de Recherche et d'Enseignement en Médecine Mitochondriale, Institut MITOVASC, CNRS 6214, INSERM U1083, Université d'Angers , 4 Rue Larrey, 49933 Angers CEDEX 9, France
| | - Cinzia Bocca
- PREMMi, Pôle de Recherche et d'Enseignement en Médecine Mitochondriale, Institut MITOVASC, CNRS 6214, INSERM U1083, Université d'Angers , 4 Rue Larrey, 49933 Angers CEDEX 9, France
| | - Gilles Simard
- PREMMi, Pôle de Recherche et d'Enseignement en Médecine Mitochondriale, Institut MITOVASC, CNRS 6214, INSERM U1083, Université d'Angers , 4 Rue Larrey, 49933 Angers CEDEX 9, France.,Département de Biochimie et Génétique, Centre Hospitalier Universitaire , 49933 Angers CEDEX 9, France
| | - Delphine Prunier-Mirebeau
- PREMMi, Pôle de Recherche et d'Enseignement en Médecine Mitochondriale, Institut MITOVASC, CNRS 6214, INSERM U1083, Université d'Angers , 4 Rue Larrey, 49933 Angers CEDEX 9, France.,Département de Biochimie et Génétique, Centre Hospitalier Universitaire , 49933 Angers CEDEX 9, France
| | - Juan Manuel Chao de la Barca
- PREMMi, Pôle de Recherche et d'Enseignement en Médecine Mitochondriale, Institut MITOVASC, CNRS 6214, INSERM U1083, Université d'Angers , 4 Rue Larrey, 49933 Angers CEDEX 9, France.,Département de Biochimie et Génétique, Centre Hospitalier Universitaire , 49933 Angers CEDEX 9, France
| | - Dominique Bonneau
- PREMMi, Pôle de Recherche et d'Enseignement en Médecine Mitochondriale, Institut MITOVASC, CNRS 6214, INSERM U1083, Université d'Angers , 4 Rue Larrey, 49933 Angers CEDEX 9, France.,Département de Biochimie et Génétique, Centre Hospitalier Universitaire , 49933 Angers CEDEX 9, France
| | - Vincent Procaccio
- PREMMi, Pôle de Recherche et d'Enseignement en Médecine Mitochondriale, Institut MITOVASC, CNRS 6214, INSERM U1083, Université d'Angers , 4 Rue Larrey, 49933 Angers CEDEX 9, France.,Département de Biochimie et Génétique, Centre Hospitalier Universitaire , 49933 Angers CEDEX 9, France
| | - Fabrice Prunier
- Institut MITOVASC, Laboratoire EA3860, Cardioprotection, Remodelage et Thrombose , Rue Haute de Reculée, FR-49045, Angers, France.,Département de Cardiologie, Centre Hospitalier Universitaire , 49933 Angers CEDEX 9, France
| | - Guy Lenaers
- PREMMi, Pôle de Recherche et d'Enseignement en Médecine Mitochondriale, Institut MITOVASC, CNRS 6214, INSERM U1083, Université d'Angers , 4 Rue Larrey, 49933 Angers CEDEX 9, France
| | - Pascal Reynier
- PREMMi, Pôle de Recherche et d'Enseignement en Médecine Mitochondriale, Institut MITOVASC, CNRS 6214, INSERM U1083, Université d'Angers , 4 Rue Larrey, 49933 Angers CEDEX 9, France.,Département de Biochimie et Génétique, Centre Hospitalier Universitaire , 49933 Angers CEDEX 9, France
| |
Collapse
|
30
|
White CR, Datta G, Giordano S. High-Density Lipoprotein Regulation of Mitochondrial Function. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 982:407-429. [PMID: 28551800 DOI: 10.1007/978-3-319-55330-6_22] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Lipoproteins play a key role in regulating plasma and tissue levels of cholesterol. Apolipoprotein B (apoB)-containing lipoproteins, including chylomicrons, very-low density lipoprotein (VLDL) and low-density lipoprotein (LDL), serve as carriers of triglycerides and cholesterol and deliver these metabolites to peripheral tissues. In contrast, high-density lipoprotein (HDL) mediates Reverse Cholesterol Transport (RCT), a process by which excess cholesterol is removed from the periphery and taken up by hepatocytes where it is metabolized and excreted. Anti-atherogenic properties of HDL have been largely ascribed to apoA-I, the major protein component of the lipoprotein particle. The inflammatory response associated with atherosclerosis and ischemia-reperfusion (I-R) injury has been linked to the development of mitochondrial dysfunction. Under these conditions, an increase in reactive oxygen species (ROS) formation induces damage to mitochondrial structural elements, leading to a reduction in ATP synthesis and initiation of the apoptotic program. Recent studies suggest that HDL-associated apoA-I and lysosphingolipids attenuate mitochondrial injury by multiple mechanisms, including the suppression of ROS formation and induction of autophagy. Other apolipoproteins, however, present in lower abundance in HDL particles may exert opposing effects on mitochondrial function. This chapter examines the role of HDL-associated apolipoproteins and lipids in the regulation of mitochondrial function and bioenergetics.
Collapse
Affiliation(s)
- C Roger White
- Department of Medicine, Division of Cardiovascular Disease, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Geeta Datta
- Department of Medicine, Division of Cardiovascular Disease, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Samantha Giordano
- Department of Medicine, Division of Cardiovascular Disease, University of Alabama at Birmingham, Birmingham, AL, USA.
| |
Collapse
|
31
|
Cardioprotection by remote ischemic conditioning and its signal transduction. Pflugers Arch 2016; 469:159-181. [DOI: 10.1007/s00424-016-1922-6] [Citation(s) in RCA: 107] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2016] [Accepted: 11/28/2016] [Indexed: 12/23/2022]
|
32
|
[Cardioprotection via the arm? : How a blood pressure cuff decreases infarct sizes]. Herz 2016; 42:565-572. [PMID: 27785525 DOI: 10.1007/s00059-016-4490-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Revised: 09/10/2016] [Accepted: 09/30/2016] [Indexed: 01/05/2023]
Abstract
Cardiovascular diseases and especially myocardial infarctions are responsible for a high morbidity and mortality throughout Europe. An essential aspect of myocardial infarction is ischemia/reperfusion injury which represents the necrosis of myocytes following reperfusion. One possible option to counteract ischemia/reperfusion injury is the much researched process of remote ischemic conditioning (RIC), whereby a certain tissue (e.g. skeletal muscle) is subjected to several cycles of short periods (e.g. 5 min) of ischemia and reperfusion and leads to the protection of another organ (e.g. the heart). Despite substantial efforts to elucidate the underlying mechanisms during the last decades, this phenomenon is not yet completely understood. Clinical studies mainly concentrated on laboratory and radiological parameters, which led to better understanding of RIC; however, large clinical studies evaluating the possible influence on mortality are still lacking. This review article provides an introduction to RIC and summarizes the current understanding of known pathomechanisms and the results of important clinical studies.
Collapse
|
33
|
Chao de la Barca JM, Bakhta O, Kalakech H, Simard G, Tamareille S, Catros V, Callebert J, Gadras C, Tessier L, Reynier P, Prunier F, Mirebeau-Prunier D. Metabolic Signature of Remote Ischemic Preconditioning Involving a Cocktail of Amino Acids and Biogenic Amines. J Am Heart Assoc 2016; 5:e003891. [PMID: 27664804 PMCID: PMC5079040 DOI: 10.1161/jaha.116.003891] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2016] [Accepted: 07/01/2016] [Indexed: 12/23/2022]
Abstract
BACKGROUND Remote ischemic preconditioning (RIPC) is an attractive therapeutic procedure for protecting the heart against ischemia/reperfusion injury. Despite evidence of humoral mediators transported through the circulation playing a critical role, their actual identities so far remain unknown. We sought to identify plasmatic RIPC-induced metabolites that may play a role. METHODS AND RESULTS Rat plasma samples from RIPC and control groups were analyzed using a targeted metabolomic approach aimed at measuring 188 metabolites. Principal component analysis and orthogonal partial least-squares discriminant analysis were used to identify the metabolites that discriminated between groups. Plasma samples from 50 patients subjected to RIPC were secondarily explored to confirm the results obtained in rats. Finally, a combination of the metabolites that were significantly increased in both rat and human plasma was injected prior to myocardial ischemia/reperfusion in rats. In the rat samples, 124 molecules were accurately quantified. Six metabolites (ornithine, glycine, kynurenine, spermine, carnosine, and serotonin) were the most significant variables for marked differentiation between the RIPC and control groups. In human plasma, analysis confirmed ornithine decrease and kynurenine and glycine increase following RIPC. Injection of the glycine and kynurenine alone or in combination replicated the protective effects of RIPC seen in rats. CONCLUSIONS We have hereby reported significant variations in a cocktail of amino acids and biogenic amines after remote ischemic preconditioning in both rat and human plasma. CLINICAL TRIAL REGISTRATION URL: http://www.clinicaltrials.gov. Unique identifier: NCT01390129.
Collapse
Affiliation(s)
- Juan Manuel Chao de la Barca
- University of Angers, France Department of Biochemistry and Genetics, University Hospital of Angers, France Laboratory of Neurovascular and Mitochondrial Integrated Biology, National Institute of Medical Research (INSERM) U771, National Centre of Scientific Research (CNRS) UMR 6214, Angers, France
| | - Oussama Bakhta
- University of Angers, France Laboratory of Cardioprotection, Remodeling, and Thrombosis, Université d'Angers, Angers, France
| | - Hussein Kalakech
- University of Angers, France Laboratory of Cardioprotection, Remodeling, and Thrombosis, Université d'Angers, Angers, France
| | - Gilles Simard
- University of Angers, France Department of Biochemistry and Genetics, University Hospital of Angers, France Laboratory of Oxidative Stress and Metabolic Pathologies (SOPAM), National Institute of Medical Research (INSERM) 1063, Angers, France
| | - Sophie Tamareille
- University of Angers, France Laboratory of Cardioprotection, Remodeling, and Thrombosis, Université d'Angers, Angers, France
| | - Véronique Catros
- Laboratory of Cytogenetic and Cellular Biology, University Hospital of Rennes and National Institute of Medical Research (INSERM) U991, Rennes, France
| | | | - Cédric Gadras
- Department of Biochemistry and Genetics, University Hospital of Angers, France
| | - Lydie Tessier
- Department of Biochemistry and Genetics, University Hospital of Angers, France
| | - Pascal Reynier
- University of Angers, France Department of Biochemistry and Genetics, University Hospital of Angers, France Laboratory of Neurovascular and Mitochondrial Integrated Biology, National Institute of Medical Research (INSERM) U771, National Centre of Scientific Research (CNRS) UMR 6214, Angers, France
| | - Fabrice Prunier
- University of Angers, France Department of Cardiology, University Hospital of Angers, France Laboratory of Cardioprotection, Remodeling, and Thrombosis, Université d'Angers, Angers, France
| | - Delphine Mirebeau-Prunier
- University of Angers, France Department of Biochemistry and Genetics, University Hospital of Angers, France Laboratory of Neurovascular and Mitochondrial Integrated Biology, National Institute of Medical Research (INSERM) U771, National Centre of Scientific Research (CNRS) UMR 6214, Angers, France
| |
Collapse
|
34
|
Epps JA, Smart NA. Remote ischaemic conditioning in the context of type 2 diabetes and neuropathy: the case for repeat application as a novel therapy for lower extremity ulceration. Cardiovasc Diabetol 2016; 15:130. [PMID: 27613524 PMCID: PMC5018170 DOI: 10.1186/s12933-016-0444-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2016] [Accepted: 08/19/2016] [Indexed: 02/07/2023] Open
Abstract
An emerging treatment modality for reducing damage caused by ischaemia–reperfusion injury is ischaemic conditioning. This technique induces short periods of ischaemia that have been found to protect against a more significant ischaemic insult. Remote ischaemic conditioning (RIC) can be administered more conveniently and safely, by inflation of a pneumatic blood pressure cuff to a suprasystolic pressure on a limb. Protection is then transferred to a remote organ via humoral and neural pathways. The diabetic state is particularly vulnerable to ischaemia–reperfusion injury, and ischaemia is a significant cause of many diabetic complications, including the diabetic foot. Despite this, studies utilising ischaemic conditioning and RIC in type 2 diabetes have often been disappointing. A newer strategy, repeat RIC, involves the repeated application of short periods of limb ischaemia over days or weeks. It has been demonstrated that this improves endothelial function, skin microcirculation, and modulates the systemic inflammatory response. Repeat RIC was recently shown to be beneficial for healing in lower extremity diabetic ulcers. This article summarises the mechanisms of RIC, and the impact that type 2 diabetes may have upon these, with the role of neural mechanisms in the context of diabetic neuropathy a focus. Repeat RIC may show more promise than RIC in type 2 diabetes, and its potential mechanisms and applications will also be explored. Considering the high costs, rates of chronicity and serious complications resulting from diabetic lower extremity ulceration, repeat RIC has the potential to be an effective novel advanced therapy for this condition.
Collapse
Affiliation(s)
- J A Epps
- School of Science and Technology, The University of New England, Armidale, NSW, 2351, Australia
| | - N A Smart
- School of Science and Technology, The University of New England, Armidale, NSW, 2351, Australia.
| |
Collapse
|
35
|
White CR, Giordano S, Anantharamaiah GM. High-density lipoprotein, mitochondrial dysfunction and cell survival mechanisms. Chem Phys Lipids 2016; 199:161-169. [PMID: 27150975 DOI: 10.1016/j.chemphyslip.2016.04.007] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2016] [Revised: 04/22/2016] [Accepted: 04/23/2016] [Indexed: 01/08/2023]
Abstract
Ischemic injury is associated with acute myocardial infarction, percutaneous coronary intervention, coronary artery bypass grafting and open heart surgery. The timely re-establishment of blood flow is critical in order to minimize cardiac complications. Reperfusion after a prolonged ischemic period, however, can induce severe cardiomyocyte dysfunction with mitochondria serving as a major target of ischemia/reperfusion (I/R) injury. An increase in the formation of reactive oxygen species (ROS) induces damage to mitochondrial respiratory complexes leading to uncoupling of oxidative phosphorylation. Mitochondrial membrane perturbations also contribute to calcium overload, opening of the mitochondrial permeability transition pore (mPTP) and the release of apoptotic mediators into the cytoplasm. Clinical and experimental studies show that ischemic preconditioning (ICPRE) and postconditioning (ICPOST) attenuate mitochondrial injury and improve cardiac function in the context of I/R injury. This is achieved by the activation of two principal cell survival cascades: 1) the Reperfusion Injury Salvage Kinase (RISK) pathway; and 2) the Survivor Activating Factor Enhancement (SAFE) pathway. Recent data suggest that high density lipoprotein (HDL) mimics the effects of conditioning protocols and attenuates myocardial I/R injury via activation of the RISK and SAFE signaling cascades. In this review, we discuss the roles of apolipoproteinA-I (apoA-I), the major protein constituent of HDL, and sphingosine 1-phosphate (S1P), a lysosphingolipid associated with small, dense HDL particles as mediators of cardiomyocyte survival. Both apoA-I and S1P exert an infarct-sparing effect by preventing ROS-dependent injury and inhibiting the opening of the mPTP.
Collapse
Affiliation(s)
- C Roger White
- Department of Medicine, Division of Cardiovascular Disease, University of Alabama at Birmingham, Birmingham, AL, USA.
| | - Samantha Giordano
- Department of Medicine, Division of Cardiovascular Disease, University of Alabama at Birmingham, Birmingham, AL, USA
| | - G M Anantharamaiah
- The Division of Gerontology, Geriatric Medicine and Palliative Care, University of Alabama at Birmingham, Birmingham, AL, USA; Biochemistry and Molecular Genetics, University of Alabama at Birmingham, Birmingham, AL, USA
| |
Collapse
|
36
|
|
37
|
Remote ischemic preconditioning in aortic valve surgery: Results of a randomized controlled study. J Cardiol 2016; 67:36-41. [DOI: 10.1016/j.jjcc.2015.06.007] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2015] [Revised: 05/13/2015] [Accepted: 06/01/2015] [Indexed: 02/06/2023]
|
38
|
Abstract
Reperfusion is mandatory to salvage ischemic myocardium from infarction, but reperfusion per se contributes to injury and ultimate infarct size. Therefore, cardioprotection beyond that by timely reperfusion is needed to reduce infarct size and improve the prognosis of patients with acute myocardial infarction. The conditioning phenomena provide such cardioprotection, insofar as brief episodes of coronary occlusion/reperfusion preceding (ischemic preconditioning) or following (ischemic postconditioning) sustained myocardial ischemia with reperfusion reduce infarct size. Even ischemia/reperfusion in organs remote from the heart provides cardioprotection (remote ischemic conditioning). The present review characterizes the signal transduction underlying the conditioning phenomena, including their physical and chemical triggers, intracellular signal transduction, and effector mechanisms, notably in the mitochondria. Cardioprotective signal transduction appears as a highly concerted spatiotemporal program. Although the translation of ischemic postconditioning and remote ischemic conditioning protocols to patients with acute myocardial infarction has been fairly successful, the pharmacological recruitment of cardioprotective signaling has been largely disappointing to date.
Collapse
Affiliation(s)
- Gerd Heusch
- From the Institute for Pathophysiology, West German Heart and Vascular Centre, University of Essen Medical School, Essen, Germany.
| |
Collapse
|
39
|
Wang M, Sun GB, Zhang JY, Luo Y, Yu YL, Xu XD, Meng XB, Zhang MD, Lin WB, Sun XB. Elatoside C protects the heart from ischaemia/reperfusion injury through the modulation of oxidative stress and intracellular Ca²⁺ homeostasis. Int J Cardiol 2015; 185:167-76. [PMID: 25796004 DOI: 10.1016/j.ijcard.2015.03.140] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2014] [Revised: 01/27/2015] [Accepted: 03/11/2015] [Indexed: 10/23/2022]
Abstract
BACKGROUND We have previously shown that Elatoside C reduces cardiomyocyte apoptosis during ischaemia/reperfusion (I/R). Here, we investigated whether Elatoside C improves heart function in isolated rat hearts subjected to I/R and elucidated the potential mechanisms involved in Elatoside C-induced protection. METHODS AND RESULTS Isolated rat hearts were subjected to global ischaemia followed by reperfusion in the absence or presence of Elatoside C. We found that Elatoside C significantly attenuated cardiac dysfunction and depressed oxidative stress induced by I/R. Consistently, Elatoside C prevented I/R-induced mitochondrial dysfunction, which was evident by the inhibition of mitochondrial ROS production, mitochondrial permeability transition pore (mPTP) opening, cytochrome c release from the mitochondria and Bax translocation. Moreover, Elatoside C improved abnormal calcium handling during I/R, including increasing sarcoplasmic reticulum Ca(2+) ATPase (SERCA2) activity, alleviating [Ca(2+)]ER depletion, and reducing the expression levels of ER stress protein markers. All of these protective effects of Elatoside C were partially abolished by the PI3K/Akt inhibitor LY294002, ERK1/2 inhibitor PD98059, and JAK2/STAT3 inhibitor AG490. Further assessment in isolated cardiomyocytes showed that Elatoside C maintained the Ca(2+) transients and cell shortening against I/R. CONCLUSIONS Elatoside C protects against cardiac injury during I/R by attenuating oxidative stress and [Ca(2+)]i overload through the activation of both the reperfusion injury salvage kinase (RISK) pathway (including PI3K/Akt and ERK1/2) and the survivor activating factor enhancement (SAFE) pathway (including JAK2/STAT3) and, subsequently, inhibiting the opening of mPTPs.
Collapse
Affiliation(s)
- Min Wang
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100193, PR China
| | - Gui-Bo Sun
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100193, PR China.
| | - Jing-Yi Zhang
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100193, PR China
| | - Yun Luo
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100193, PR China
| | - Ying-Li Yu
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100193, PR China
| | - Xu-Dong Xu
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100193, PR China
| | - Xiang-Bao Meng
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100193, PR China
| | - Miao-di Zhang
- Harbin University of Commerce, Harbin 150076, Heilongjiang, PR China
| | - Wen-Bin Lin
- Harbin University of Commerce, Harbin 150076, Heilongjiang, PR China
| | - Xiao-Bo Sun
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100193, PR China.
| |
Collapse
|
40
|
Plasma from human volunteers subjected to remote ischemic preconditioning protects human endothelial cells from hypoxia-induced cell damage. Basic Res Cardiol 2015; 110:17. [PMID: 25716080 PMCID: PMC4341024 DOI: 10.1007/s00395-015-0474-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2014] [Revised: 02/17/2015] [Accepted: 02/18/2015] [Indexed: 02/06/2023]
Abstract
Short repeated cycles of peripheral ischemia/reperfusion (I/R) can protect distant organs from subsequent prolonged I/R injury; a phenomenon known as remote ischemic preconditioning (RIPC). A RIPC-mediated release of humoral factors might play a key role in this protection and vascular endothelial cells are potential targets for these secreted factors. In the present study, RIPC-plasma obtained from healthy male volunteers was tested for its ability to protect human umbilical endothelial cells (HUVEC) from hypoxia–induced cell damage. 10 healthy male volunteers were subjected to a RIPC-protocol consisting of 4 × 5 min inflation/deflation of a blood pressure cuff located at the upper arm. Plasma was collected before (T0; control), directly after (T1) and 1 h after (T2) the RIPC procedure. HUVEC were subjected to 24 h hypoxia damage and simultaneously incubated with 5 % of the respective RIPC-plasma. Cell damage was evaluated by lactate dehydrogenase (LDH)-measurements. Western blot experiments of hypoxia inducible factor 1 alpha (HIF1alpha), phosphorylated signal transducer and activator of transcription 5 (STAT5), protein kinase B (AKT) and extracellular signal-related kinase 1/2 (ERK-1/2) were performed. Furthermore, the concentrations of hVEGF were evaluated in the RIPC-plasma by sandwich ELISA. Hypoxia–induced cell damage was significantly reduced by plasma T1 (p = 0.02 vs T0). The protective effect of plasma T1 was accompanied by an augmentation of the intracellular HIF1alpha (p = 0.01 vs T0) and increased phosphorylation of ERK-1/2 (p = 0.03 vs T0). Phosphorylation of AKT and STAT5 remained unchanged. Analysis of the protective RIPC-plasma T1 showed significantly reduced levels of hVEGF (p = 0.01 vs T0). RIPC plasma protects endothelial cells from hypoxia–induced cell damage and humoral mediators as well as intracellular HIF1alpha may be involved.
Collapse
|