1
|
Liu H, Wang S, Wang J, Guo X, Song Y, Fu K, Gao Z, Liu D, He W, Yang LL. Energy metabolism in health and diseases. Signal Transduct Target Ther 2025; 10:69. [PMID: 39966374 PMCID: PMC11836267 DOI: 10.1038/s41392-025-02141-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 11/08/2024] [Accepted: 12/25/2024] [Indexed: 02/20/2025] Open
Abstract
Energy metabolism is indispensable for sustaining physiological functions in living organisms and assumes a pivotal role across physiological and pathological conditions. This review provides an extensive overview of advancements in energy metabolism research, elucidating critical pathways such as glycolysis, oxidative phosphorylation, fatty acid metabolism, and amino acid metabolism, along with their intricate regulatory mechanisms. The homeostatic balance of these processes is crucial; however, in pathological states such as neurodegenerative diseases, autoimmune disorders, and cancer, extensive metabolic reprogramming occurs, resulting in impaired glucose metabolism and mitochondrial dysfunction, which accelerate disease progression. Recent investigations into key regulatory pathways, including mechanistic target of rapamycin, sirtuins, and adenosine monophosphate-activated protein kinase, have considerably deepened our understanding of metabolic dysregulation and opened new avenues for therapeutic innovation. Emerging technologies, such as fluorescent probes, nano-biomaterials, and metabolomic analyses, promise substantial improvements in diagnostic precision. This review critically examines recent advancements and ongoing challenges in metabolism research, emphasizing its potential for precision diagnostics and personalized therapeutic interventions. Future studies should prioritize unraveling the regulatory mechanisms of energy metabolism and the dynamics of intercellular energy interactions. Integrating cutting-edge gene-editing technologies and multi-omics approaches, the development of multi-target pharmaceuticals in synergy with existing therapies such as immunotherapy and dietary interventions could enhance therapeutic efficacy. Personalized metabolic analysis is indispensable for crafting tailored treatment protocols, ultimately providing more accurate medical solutions for patients. This review aims to deepen the understanding and improve the application of energy metabolism to drive innovative diagnostic and therapeutic strategies.
Collapse
Affiliation(s)
- Hui Liu
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Shuo Wang
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jianhua Wang
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xin Guo
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yujing Song
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Kun Fu
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhenjie Gao
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Danfeng Liu
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| | - Wei He
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| | - Lei-Lei Yang
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| |
Collapse
|
2
|
Nikolaou PE, Konijnenberg LS, Kostopoulos IV, Miliotis M, Mylonas N, Georgoulis A, Pavlidis G, Kuster CT, van Reijmersdal VP, Luiken TT, Agapaki A, Roverts R, Orologas N, Grigoriadis D, Pallot G, Boucher P, Kostomitsopoulos N, Pieper MP, Germain S, Loukas Y, Dotsikas Y, Ikonomidis I, Hatzigeorgiou AG, Tsitsilonis O, Zuurbier CJ, Nijveldt R, van Royen N, Andreadou I. Empagliflozin in Acute Myocardial Infarction Reduces No-Reflow and Preserves Cardiac Function by Preventing Endothelial Damage. JACC Basic Transl Sci 2025; 10:43-61. [PMID: 39958474 PMCID: PMC11830260 DOI: 10.1016/j.jacbts.2024.08.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 08/20/2024] [Accepted: 08/20/2024] [Indexed: 02/18/2025]
Abstract
Empagliflozin treatment before acute myocardial infarction mainly targets the endothelial cell transcriptome. Empagliflozin treatment before and after myocardial infarction decreased no reflow and microvascular injury, leading to reduced infiltration of inflammatory cells, reduced infarct size, and improved cardiac function in mice. In diabetic patients receiving empagliflozin after myocardial infarction, perfused boundary region, flow-mediated dilation, and global longitudinal strain were improved.
Collapse
Affiliation(s)
- Panagiota Efstathia Nikolaou
- Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece
- Department of Cardiology, Radboud University Medical Center, Nijmegen, the Netherlands
| | | | - Ioannis V. Kostopoulos
- Section of Animal and Human Physiology, Department of Biology, National and Kapodistrian University of Athens, Athens, Greece
| | - Marios Miliotis
- DIANA-Lab, Department of Computer Science and Biomedical Informatics, University of Thessaly, Lamia, Greece
| | - Nikolaos Mylonas
- Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece
| | - Anastasios Georgoulis
- Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece
| | - George Pavlidis
- Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Carolien T.A. Kuster
- Department of Cardiology, Radboud University Medical Center, Nijmegen, the Netherlands
| | | | - Tom T.J. Luiken
- Department of Medical BioSciences, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Anna Agapaki
- Center of Basic Research, Biomedical Research Foundation, Academy of Athens, Athens, Greece
| | - Rona Roverts
- Electron Microscopy Center, Radboud UMC Technology Center, Radboud Institute of Molecular Life Sciences, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Nikolaos Orologas
- Section of Animal and Human Physiology, Department of Biology, National and Kapodistrian University of Athens, Athens, Greece
| | - Dimitris Grigoriadis
- DIANA-Lab, Department of Computer Science and Biomedical Informatics, University of Thessaly, Lamia, Greece
| | - Gaëtan Pallot
- Center for Interdisciplinary Research in Biology, College de France, CNRS, INSERM, PSL Research University, Paris, France
| | - Pierre Boucher
- Center for Interdisciplinary Research in Biology, College de France, CNRS, INSERM, PSL Research University, Paris, France
| | - Nikolaos Kostomitsopoulos
- Laboratory Animal Facilities, Center of Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | | | - Stéphane Germain
- Center for Interdisciplinary Research in Biology, College de France, CNRS, INSERM, PSL Research University, Paris, France
| | - Yannis Loukas
- Laboratory of Pharmaceutical Analysis, Department of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece
| | - Yannis Dotsikas
- Laboratory of Pharmaceutical Analysis, Department of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece
| | - Ignatios Ikonomidis
- Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Artemis G. Hatzigeorgiou
- DIANA-Lab, Department of Computer Science and Biomedical Informatics, University of Thessaly, Lamia, Greece
| | - Ourania Tsitsilonis
- Section of Animal and Human Physiology, Department of Biology, National and Kapodistrian University of Athens, Athens, Greece
| | - Coert J. Zuurbier
- Laboratory of Experimental Intensive Care and Anesthesiology, Department of Anesthesiology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, Amsterdam, the Netherlands
| | - Robin Nijveldt
- Department of Cardiology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Niels van Royen
- Department of Cardiology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Ioanna Andreadou
- Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
3
|
Cour M, Pedretti S, Nduhirabandi F, Hacking D, Frias MA, Hausenloy DJ, Lecour S. Interplay between the SAFE and the sphingolipid pathway for cardioprotection. Life Sci 2024; 358:123145. [PMID: 39401691 DOI: 10.1016/j.lfs.2024.123145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2024] [Revised: 10/07/2024] [Accepted: 10/10/2024] [Indexed: 10/19/2024]
Abstract
AIM Activation of both the Survivor Activating Factor Enhancement (SAFE) pathway (including Tumor Necrosis Factor-alpha (TNF-α) and Signal Transducer and Activator of Transcription-3 (STAT-3)) and the sphingolipid signalling pathway (including sphingosine kinase-1 (SK1) and sphingosine-1 phosphate (S1P)) play a key role in promoting cardioprotection against ischemia-reperfusion injury (IRI). We investigated whether the activation of the SAFE pathway by exogenous S1P is dependent on the activation of SK1 for cardioprotection. MATERIALS AND METHODS Isolated cardiomyocytes from TNF-α knockout (KO) mice, cardiomyocyte-specific STAT-3 KO mice and their wild-type (WT) littermates were exposed to simulated ischemia in the presence of a trigger of the SAFE pathway (S1P) and SK1 inhibitor (SK1-I). Similarly, isolated perfused hearts from adult TNF-α KO, STAT-3 KO and WT mice were subjected to IRI with S1P and/or SK1-I. Cell viability, infarct size (IS) and SK1 activity were assessed. KEY FINDINGS In isolated cardiomyocytes and in isolated hearts subjected to simulated ischemia/IRI, S1P pretreatment decreased cell death in WT mice, an effect that was abrogated in the presence of SK1-I. S1P failed to reduce cell death after simulated ischemia/IRI in both cardiomyocytes or hearts isolated from TNF-α KO and STAT-3 KO mice. Interestingly, S1P pretreatment increased SK1 activity in WT and STAT-3 KO mice, with no changes in TNF-α KO mice. SIGNIFICANCE Our data strongly suggest SK1 as a key component to activate STAT-3 downstream of TNF-α in the SAFE pathway, paving the way for the development of novel cardioprotective strategies that may target SK1 to modulate the SAFE pathway and increase cell survival following IRI.
Collapse
Affiliation(s)
- Martin Cour
- Hatter Institute/Cape Heart Institute, Department of Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Sarah Pedretti
- Hatter Institute/Cape Heart Institute, Department of Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Frederic Nduhirabandi
- Hatter Institute/Cape Heart Institute, Department of Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Damian Hacking
- Hatter Institute/Cape Heart Institute, Department of Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Miguel A Frias
- Division of Laboratory Medicine, Diagnostic Department, Geneva University Hospitals, Geneva, Switzerland
| | - Derek J Hausenloy
- The Hatter Cardiovascular Institute, University College London, London, UK
| | - Sandrine Lecour
- Hatter Institute/Cape Heart Institute, Department of Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa.
| |
Collapse
|
4
|
de Assis JL, Grelle GMRS, Fernandes AM, da Silva Aniceto B, Pompeu P, de Mello FV, Garrett R, Valverde RHF, Einicker-Lamas M. Sphingosine 1-phosphate protective effect on human proximal tubule cells submitted to an in vitro ischemia model: the role of JAK2/STAT3. J Physiol Biochem 2024; 80:831-843. [PMID: 39155330 DOI: 10.1007/s13105-024-01038-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Accepted: 07/22/2024] [Indexed: 08/20/2024]
Abstract
Acute kidney injury is a serious public health problem worldwide, being ischemia and reperfusion (I/R) the main lesion-aggravating factor that contributes to the evolution towards chronic kidney disease. Nonetheless, intervention approaches currently available are just considered palliative options. In order to offer an alternative treatment, it is important to understand key factors involved in the development of the disease including the rescue of the affected cells and/or the release of paracrine factors that are crucial for tissue repair. Bioactive lipids such as sphingosine 1-phosphate (S1P) have significant effects on the modulation of signaling pathways involved in tissue regeneration, such as cell survival, proliferation, differentiation, and migration. The main objective of this work was to explore the protective effect of S1P using human kidney proximal tubule cells submitted to a mimetic I/R lesion, via ATP depletion. We observed that the S1P pre-treatment increases cell survival by 50% and preserves the cell proliferation capacity of injured cells. We showed the presence of different bioactive lipids notably related to tissue repair but, more importantly, we noted that the pre-treatment with S1P attenuated the ischemia-induced effects in response to the injury, resulting in higher endogenous S1P production. All receptors but S1PR3 are present in these cells and the protective and proliferative effect of S1P/S1P receptors axis occur, at least in part, through the activation of the SAFE pathway. To our knowledge, this is the first time that S1PR4 and S1PR5 are referred in these cells and also the first indication of JAK2/STAT3 pathway involvement in S1P-mediated protection in an I/R renal model.
Collapse
Affiliation(s)
- Juliane Lopes de Assis
- Laboratório de Biomembranas, Instituto de Biofísica Carlos Chagas Filho - Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Gloria Maria Ramalho Soares Grelle
- Laboratório de Biomembranas, Instituto de Biofísica Carlos Chagas Filho - Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Laboratório de Metabolômica, LADETEC, Instituto de Química - Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Aline Marie Fernandes
- Laboratório de Biomembranas, Instituto de Biofísica Carlos Chagas Filho - Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Bárbara da Silva Aniceto
- Laboratório de Biomembranas, Instituto de Biofísica Carlos Chagas Filho - Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Pedro Pompeu
- Laboratório de Biomembranas, Instituto de Biofísica Carlos Chagas Filho - Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Fabiana Vieira de Mello
- Serviço de Citometria do Instituto de Pediatria e Puericultura Martagão Gesteira (IPPMG) - Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Rafael Garrett
- Laboratório de Metabolômica, LADETEC, Instituto de Química - Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Rafael Hospodar Felippe Valverde
- Laboratório de Biomembranas, Instituto de Biofísica Carlos Chagas Filho - Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Marcelo Einicker-Lamas
- Laboratório de Biomembranas, Instituto de Biofísica Carlos Chagas Filho - Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.
| |
Collapse
|
5
|
Babaei G, Sadraei S, Yarahmadi M, Omidvari S, Aarabi A, Rajabibazl M. STAT protein family and cardiovascular diseases: overview of pathological mechanisms and therapeutic implications. Mol Biol Rep 2024; 51:440. [PMID: 38520542 DOI: 10.1007/s11033-024-09371-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Accepted: 02/21/2024] [Indexed: 03/25/2024]
Abstract
Globally, cardiovascular diseases (CVD) are one of the significant causes of death and are considered a major concern of human society. One of the most crucial objectives of scientists is to reveal the mechanisms associated with the pathogenesis of CVD, which has attracted the attention of many scientists. Accumulating evidence showed that the signal transducer and activator of transcription (STAT) signaling pathway is involved in various physiological and pathological processes. According to research on the molecular mechanisms of CVDs, the STAT family of proteins is one of the most crucial players in these diseases. Numerous studies have demonstrated the undeniable relevance of STAT family proteins in various CVDs. The aim of this review is to shed light on how STAT signaling pathways are related to CVD and the potential for using these signaling pathways as therapeutic targets.
Collapse
Affiliation(s)
- Ghader Babaei
- Department of Clinical Biochemistry, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Samin Sadraei
- Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Maral Yarahmadi
- Department of Clinical Biochemistry, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Samareh Omidvari
- Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Aryan Aarabi
- Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Masoumeh Rajabibazl
- Department of Clinical Biochemistry, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
6
|
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
|
7
|
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: 1.0] [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
|
8
|
Muñoz JP, Sànchez-Fernàndez-de-Landa P, Diarte-Añazco EMG, Zorzano A, Blanco-Vaca F, Julve J. FTY720-P, a Biased S1PR Ligand, Increases Mitochondrial Function through STAT3 Activation in Cardiac Cells. Int J Mol Sci 2023; 24:ijms24087374. [PMID: 37108539 PMCID: PMC10139230 DOI: 10.3390/ijms24087374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 04/14/2023] [Accepted: 04/15/2023] [Indexed: 04/29/2023] Open
Abstract
FTY720 is an FDA-approved sphingosine derivative drug for the treatment of multiple sclerosis. This compound blocks lymphocyte egress from lymphoid organs and autoimmunity through sphingosine 1-phosphate (S1P) receptor blockage. Drug repurposing of FTY720 has revealed improvements in glucose metabolism and metabolic diseases. Studies also demonstrate that preconditioning with this compound preserves the ATP levels during cardiac ischemia in rats. The molecular mechanisms by which FTY720 promotes metabolism are not well understood. Here, we demonstrate that nanomolar concentrations of the phosphorylated form of FTY720 (FTY720-P), the active ligand of S1P receptor (S1PR), activates mitochondrial respiration and the mitochondrial ATP production rate in AC16 human cardiomyocyte cells. Additionally, FTY720-P increases the number of mitochondrial nucleoids, promotes mitochondrial morphology alterations, and induces activation of STAT3, a transcription factor that promotes mitochondrial function. Notably, the effect of FTY720-P on mitochondrial function was suppressed in the presence of a STAT3 inhibitor. In summary, our results suggest that FTY720 promotes the activation of mitochondrial function, in part, through a STAT3 action.
Collapse
Affiliation(s)
- Juan Pablo Muñoz
- Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), 08041 Barcelona, Spain
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), 28029 Madrid, Spain
| | - Paula Sànchez-Fernàndez-de-Landa
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), 28029 Madrid, Spain
- Institute for Research in Biomedicine (IRB Barcelona), 08028 Barcelona, Spain
- Departament de Bioquímica i Biomedicina Molecular, Facultat de Biologia, Universitat de Barcelona, 08028 Barcelona, Spain
| | | | - Antonio Zorzano
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), 28029 Madrid, Spain
- Institute for Research in Biomedicine (IRB Barcelona), 08028 Barcelona, Spain
- Departament de Bioquímica i Biomedicina Molecular, Facultat de Biologia, Universitat de Barcelona, 08028 Barcelona, Spain
| | - Francisco Blanco-Vaca
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), 28029 Madrid, Spain
- Department of Clinical Biochemistry, Hospital de la Santa Creu i Sant Pau, 08041 Barcelona, Spain
- Department de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, 08041 Barcelona, Spain
| | - Josep Julve
- Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), 08041 Barcelona, Spain
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), 28029 Madrid, Spain
- Department of Endocrinology and Nutrition, Hospital de la Santa Creu i Sant Pau, 08041 Barcelona, Spain
| |
Collapse
|
9
|
Borodzicz-Jażdżyk S, Jażdżyk P, Łysik W, Cudnoch-Jȩdrzejewska A, Czarzasta K. Sphingolipid metabolism and signaling in cardiovascular diseases. Front Cardiovasc Med 2022; 9:915961. [PMID: 36119733 PMCID: PMC9471951 DOI: 10.3389/fcvm.2022.915961] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 08/05/2022] [Indexed: 01/10/2023] Open
Abstract
Sphingolipids are a structural component of the cell membrane, derived from sphingosine, an amino alcohol. Its sphingoid base undergoes various types of enzymatic transformations that lead to the formation of biologically active compounds, which play a crucial role in the essential pathways of cellular signaling, proliferation, maturation, and death. The constantly growing number of experimental and clinical studies emphasizes the pivotal role of sphingolipids in the pathophysiology of cardiovascular diseases, including, in particular, ischemic heart disease, hypertension, heart failure, and stroke. It has also been proven that altering the sphingolipid metabolism has cardioprotective properties in cardiac pathologies, including myocardial infarction. Recent studies suggest that selected sphingolipids may serve as valuable biomarkers useful in the prognosis of cardiovascular disorders in clinical practice. This review aims to provide an overview of the current knowledge of sphingolipid metabolism and signaling in cardiovascular diseases.
Collapse
Affiliation(s)
- Sonia Borodzicz-Jażdżyk
- Chair and Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, Warsaw, Poland
| | - Piotr Jażdżyk
- Chair and Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, Warsaw, Poland
- Second Department of Psychiatry, Institute of Psychiatry and Neurology in Warsaw, Warsaw, Poland
| | - Wojciech Łysik
- Chair and Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, Warsaw, Poland
| | - Agnieszka Cudnoch-Jȩdrzejewska
- Chair and Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, Warsaw, Poland
| | - Katarzyna Czarzasta
- Chair and Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, Warsaw, Poland
| |
Collapse
|
10
|
Assis JLD, Fernandes AM, Aniceto BS, Fernandes da Costa PP, Banchio C, Girardini J, Vieyra A, Valverde RRHF, Einicker‐Lamas M. Sphingosine 1‐Phosphate Prevents Human Embryonic Stem Cell Death Following Ischemic Injury. EUR J LIPID SCI TECH 2022. [DOI: 10.1002/ejlt.202200019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Affiliation(s)
- Juliane L. de Assis
- Laboratório de Biomembranas Instituto de Biofísica Carlos Chagas Filho–Universidade Federal do Rio de Janeiro Rio de Janeiro Brazil
| | - Aline M. Fernandes
- Laboratório de Biomembranas Instituto de Biofísica Carlos Chagas Filho–Universidade Federal do Rio de Janeiro Rio de Janeiro Brazil
| | - Bárbara S. Aniceto
- Laboratório de Biomembranas Instituto de Biofísica Carlos Chagas Filho–Universidade Federal do Rio de Janeiro Rio de Janeiro Brazil
| | - Pedro P. Fernandes da Costa
- Laboratório de Biomembranas Instituto de Biofísica Carlos Chagas Filho–Universidade Federal do Rio de Janeiro Rio de Janeiro Brazil
| | - Claudia Banchio
- Instituto de Biologia Molecular y Celular de Rosário Rosário Argentina
| | - Javier Girardini
- Instituto de Biologia Molecular y Celular de Rosário Rosário Argentina
| | - Adalberto Vieyra
- Laboratório de Físico‐Química Biológica Instituto de Biofísica Carlos Chagas Filho–Universidade Federal do Rio de Janeiro Rio de Janeiro Brazil
| | - Rafael R. H. F. Valverde
- Laboratório de Biomembranas Instituto de Biofísica Carlos Chagas Filho–Universidade Federal do Rio de Janeiro Rio de Janeiro Brazil
| | - Marcelo Einicker‐Lamas
- Laboratório de Biomembranas Instituto de Biofísica Carlos Chagas Filho–Universidade Federal do Rio de Janeiro Rio de Janeiro Brazil
| |
Collapse
|
11
|
Ma H, Hao J, Liu H, Yin J, Qiang M, Liu M, He S, Zeng D, Liu X, Lian C, Gao Y. Peoniflorin Preconditioning Protects Against Myocardial Ischemia/Reperfusion Injury Through Inhibiting Myocardial Apoptosis: RISK Pathway Involved. Appl Biochem Biotechnol 2022; 194:1149-1165. [PMID: 34596828 DOI: 10.1007/s12010-021-03680-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2021] [Accepted: 09/08/2021] [Indexed: 11/28/2022]
Abstract
Preconditioning with Peoniflorin, a component of traditional Chinese prescriptions, was proposed to be a potential strategy for cardioprotection against ischemia/reperfusion (I/R) injury. However, the cardioprotective effect of Peoniflorin preconditioning has not been thoroughly confirmed, and the underlying mechanism remains unclear. Here, we examined the cardioprotective effect and its mechanism of Peoniflorin preconditioning against myocardial I/R injury. Rats were subjected to 30 min of transient ischemia followed by 2 h of reperfusion with or without Peoniflorin (100 mg/kg) prior to reperfusion. Peoniflorin preconditioning significantly limited myocardial infarct size and reperfusion arrhythmias, as well as obviously attenuated the histomorphological and micromorphological damages induced by I/R injury. The reduced myocardial injury was also associated with the anti-apoptotic effect of Peoniflorin, as evidence by decreased TUNEL-positive cells, upregulation of BCL-2 expression, and downregulation of Bax and caspase-3 expression. In an effort to evaluate the mechanism responsible for the observed cardioprotective and anti-apoptotic effect, Western blot of phosphorylated protein was performed after 20 min of reperfusion. Results showed that Peoniflorin preconditioning activated both the Akt and ERK1/2 arm of the reperfusion injury salvage kinase (RISK) pathway. To further confirm this mechanism, the PI3K signaling inhibitor LY294002 and ERK1/2 signaling inhibitor PD98059 were administered in vivo. The cardioprotective and anti-apoptotic effects of Peoniflorin preconditioning were diminished but not abolished by pretreatment with LY294002 or PD98059. Taken together, these results indicate that Peoniflorin preconditioning protects the myocardial against I/R injury and inhibits myocardial apoptosis via the activation of the RISK pathway, highlighting the potential therapeutic effects of Peoniflorin on reducing myocardial I/R injury.
Collapse
Affiliation(s)
- Hongen Ma
- Department of Cardiology, Ninth Affiliated Hospital of Medical College of Xi'an Jiaotong University, No. 151 East Section of South Second Ring RoadBeilin District, Xi'an, 710054, Shaanxi, China
| | - Jiping Hao
- Department of Cardiology, Ninth Affiliated Hospital of Medical College of Xi'an Jiaotong University, No. 151 East Section of South Second Ring RoadBeilin District, Xi'an, 710054, Shaanxi, China
| | - Huihui Liu
- Medical College of Yan'an University, No. 38 Guanghua RoadBaota District, Yan'an, 716000, Shaanxi, China
| | - Jia Yin
- Medical College of Yan'an University, No. 38 Guanghua RoadBaota District, Yan'an, 716000, Shaanxi, China
| | - Mingmin Qiang
- Medical College of Yan'an University, No. 38 Guanghua RoadBaota District, Yan'an, 716000, Shaanxi, China
| | - Meilin Liu
- Department of Cardiology, Ninth Affiliated Hospital of Medical College of Xi'an Jiaotong University, No. 151 East Section of South Second Ring RoadBeilin District, Xi'an, 710054, Shaanxi, China
| | - Shaohui He
- Department of Cardiology, Ninth Affiliated Hospital of Medical College of Xi'an Jiaotong University, No. 151 East Section of South Second Ring RoadBeilin District, Xi'an, 710054, Shaanxi, China
| | - Di Zeng
- Department of Cardiology, Tangdu Hospital, Fourth Military Medical University, No. 1 Xinsi RoadBaqiao District, Xi'an, 710000, Shaanxi, China
| | - Xiongtao Liu
- Department of Cardiology, Tangdu Hospital, Fourth Military Medical University, No. 1 Xinsi RoadBaqiao District, Xi'an, 710000, Shaanxi, China
| | - Cheng Lian
- Department of Cardiology, Tangdu Hospital, Fourth Military Medical University, No. 1 Xinsi RoadBaqiao District, Xi'an, 710000, Shaanxi, China
| | - Yuqin Gao
- Department of Cardiology, Ninth Affiliated Hospital of Medical College of Xi'an Jiaotong University, No. 151 East Section of South Second Ring RoadBeilin District, Xi'an, 710054, Shaanxi, China.
| |
Collapse
|
12
|
Heart Failure after Cardiac Surgery: The Role of Halogenated Agents, Myocardial Conditioning and Oxidative Stress. Int J Mol Sci 2022; 23:ijms23031360. [PMID: 35163284 PMCID: PMC8836224 DOI: 10.3390/ijms23031360] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 01/21/2022] [Accepted: 01/22/2022] [Indexed: 12/07/2022] Open
Abstract
Heart disease requires a surgical approach sometimes. Cardiac-surgery patients develop heart failure associated with ischemia induced during extracorporeal circulation. This complication could be decreased with anesthetic drugs. The cardioprotective effects of halogenated agents are based on pre- and postconditioning (sevoflurane, desflurane, or isoflurane) compared to intravenous hypnotics (propofol). We tried to put light on the shadows walking through the line of the halogenated anesthetic drugs’ effects in several enzymatic routes and oxidative stress, waiting for the final results of the ACDHUVV-16 clinical trial regarding the genetic modulation of this kind of drugs.
Collapse
|
13
|
Maslov LN, Popov SV, Mukhomedzyanov AV, Derkachev IA, Ryabov VV, Boshchenko AA, Prasad NR, Sufianova GZ, Khlestkina MS, Gareev I. Pharmacological Approaches to Limit Ischemic and Reperfusion Injuries of the Heart. Analysis of Experimental and Clinical Data on P2Y 12 Receptor Antagonists. Korean Circ J 2022; 52:737-754. [PMID: 36217596 PMCID: PMC9551227 DOI: 10.4070/kcj.2022.0162] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 08/15/2022] [Accepted: 08/24/2022] [Indexed: 12/03/2022] Open
Abstract
High mortality among people with acute myocardial infarction is one of the most urgent problems of modern cardiology. And in recent years, much attention has been paid to the search for pharmacological approaches to prevent heart damage. In this review, we tried to analyze data on the effect of P2Y12 receptor antagonists on the ischemia/reperfusion tolerance of the heart. Ischemic and reperfusion injuries of the heart underlie the pathogenesis of acute myocardial infarction (AMI) and sudden cardiac death. The mortality rate is still high and is 5–7% in patients with ST-segment elevation myocardial infarction. The review is devoted to pharmacological approaches to limitation of ischemic and reperfusion injuries of the heart. The article analyzes experimental evidence and the clinical data on the effects of P2Y12 receptor antagonists on the heart’s tolerance to ischemia/reperfusion in animals with coronary artery occlusion and reperfusion and also in patients with AMI. Chronic administration of ticagrelor prevented adverse remodeling of the heart. There is evidence that sphingosine-1-phosphate is the molecule that mediates the infarct-reducing effect of P2Y12 receptor antagonists. It was discussed a role of adenosine in the cardioprotective effect of ticagrelor.
Collapse
Affiliation(s)
- Leonid N. Maslov
- Cardiology Research Institute, Tomsk National Research Medical Center of the RAS, Tomsk, Russia
| | - Sergey V. Popov
- Cardiology Research Institute, Tomsk National Research Medical Center of the RAS, Tomsk, Russia
| | | | - Ivan A. Derkachev
- Cardiology Research Institute, Tomsk National Research Medical Center of the RAS, Tomsk, Russia
| | - Vyacheslav V. Ryabov
- Cardiology Research Institute, Tomsk National Research Medical Center of the RAS, Tomsk, Russia
| | - Alla A. Boshchenko
- Cardiology Research Institute, Tomsk National Research Medical Center of the RAS, Tomsk, Russia
| | - N. Rajendra Prasad
- Department of Biochemistry and Biotechnology, Faculty of Science, Annamalai University, Annamalainagar, Tamilnadu, India
| | | | | | | |
Collapse
|
14
|
Comità S, Femmino S, Thairi C, Alloatti G, Boengler K, Pagliaro P, Penna C. Regulation of STAT3 and its role in cardioprotection by conditioning: focus on non-genomic roles targeting mitochondrial function. Basic Res Cardiol 2021; 116:56. [PMID: 34642818 PMCID: PMC8510947 DOI: 10.1007/s00395-021-00898-0] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 09/23/2021] [Accepted: 09/27/2021] [Indexed: 12/11/2022]
Abstract
Ischemia–reperfusion injury (IRI) is one of the biggest challenges for cardiovascular researchers given the huge death toll caused by myocardial ischemic disease. Cardioprotective conditioning strategies, namely pre- and post-conditioning maneuvers, represent the most important strategies for stimulating pro-survival pathways essential to preserve cardiac health. Conditioning maneuvers have proved to be fundamental for the knowledge of the molecular basis of both IRI and cardioprotection. Among this evidence, the importance of signal transducer and activator of transcription 3 (STAT3) emerged. STAT3 is not only a transcription factor but also exhibits non-genomic pro-survival functions preserving mitochondrial function from IRI. Indeed, STAT3 is emerging as an influencer of mitochondrial function to explain the cardioprotection phenomena. Studying cardioprotection, STAT3 proved to be crucial as an element of the survivor activating factor enhancement (SAFE) pathway, which converges on mitochondria and influences their function by cross-talking with other cardioprotective pathways. Clearly there are still some functional properties of STAT3 to be discovered. Therefore, in this review, we highlight the evidence that places STAT3 as a promoter of the metabolic network. In particular, we focus on the possible interactions of STAT3 with processes aimed at maintaining mitochondrial functions, including the regulation of the electron transport chain, the production of reactive oxygen species, the homeostasis of Ca2+ and the inhibition of opening of mitochondrial permeability transition pore. Then we consider the role of STAT3 and the parallels between STA3/STAT5 in cardioprotection by conditioning, giving emphasis to the human heart and confounders.
Collapse
Affiliation(s)
- Stefano Comità
- Department of Clinical and Biological Sciences, University of Turin, Regione Gonzole 10, Orbassano, 10043, Torino, TO, Italy
| | - Saveria Femmino
- Department of Medical Sciences, University of Turin, Torino, Italy
| | - Cecilia Thairi
- Department of Clinical and Biological Sciences, University of Turin, Regione Gonzole 10, Orbassano, 10043, Torino, TO, Italy
| | | | - Kerstin Boengler
- Institute of Physiology, University of Giessen, Giessen, Germany
| | - Pasquale Pagliaro
- Department of Clinical and Biological Sciences, University of Turin, Regione Gonzole 10, Orbassano, 10043, Torino, TO, Italy.
| | - Claudia Penna
- Department of Clinical and Biological Sciences, University of Turin, Regione Gonzole 10, Orbassano, 10043, Torino, TO, Italy.
| |
Collapse
|
15
|
Yang LG, Wang AL, Li L, Yang H, Jie X, Zhu ZF, Zhang XJ, Zhao HP, Chi RF, Li B, Qin FZ, Wang JP, Wang K. Sphingosine-1-phosphate induces myocyte autophagy after myocardial infarction through mTOR inhibition. Eur J Pharmacol 2021; 907:174260. [PMID: 34144026 DOI: 10.1016/j.ejphar.2021.174260] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 06/05/2021] [Accepted: 06/07/2021] [Indexed: 11/22/2022]
Abstract
Sphingosine-1-phosphate (S1P)/S1P receptor 1 signaling exerts cardioprotective effects including inhibition of myocyte apoptosis. However, little is known about the effect of S1P treatment on myocyte autophagy after myocardial infarction (MI). In the present study, we tested the hypothesis that S1P induces myocyte autophagy through inhibition of the mammalian target of rapamycin (mTOR), leading to improvement of left ventricular (LV) function after MI. Sprague-Dawley rats underwent MI or sham operation. The animals were randomized to receive S1P (50 μg/kg/day, i.p.) or placebo for one week. H9C2 cardiomyocytes cultured in serum- and glucose-deficient medium were treated with or without S1P for 3 h. MI rats exhibited an increase in LV end-diastolic dimension (EDD) and decreases in LV fractional shortening (FS) and the maximal rate of LV pressure rise (+dP/dt). S1P treatment attenuated the increase in LV EDD and decreases in LV FS and +dP/dt. In the MI placebo group, the LC3 II/I ratio, a marker of autophagy, was increased, and increased further by S1P treatment. S1P also enhanced the autophagy-related proteins Atg4b and Atg5 after MI. Similarly, in cultured cardiomyocytes, autophagy was increased under glucose and serum deprivation, and increased further by S1P treatment. The effect of S1P on myocyte autophagy was associated with mTOR inhibition after MI or in cultured cardiomyocytes under glucose and serum deprivation. S1P treatment prevents LV remodeling, enhances myocyte autophagy and inhibits mTOR activity after MI. These findings suggest that S1P treatment induces myocyte autophagy through mTOR inhibition, leading to the attenuation of LV dysfunction after MI.
Collapse
Affiliation(s)
- Li-Guo Yang
- The Second Hospital of Shanxi Medical University, Taiyuan, 030001, Shanxi, PR China; Shanxi Medical University, Taiyuan, 030001, Shanxi, PR China; Shanxi Provincial People's Hospital, Taiyuan, 030001, Shanxi, PR China
| | - Ai-Ling Wang
- The Second Hospital of Shanxi Medical University, Taiyuan, 030001, Shanxi, PR China; Shanxi Medical University, Taiyuan, 030001, Shanxi, PR China
| | - Lu Li
- Shanxi Medical University, Taiyuan, 030001, Shanxi, PR China
| | - Hong Yang
- The Second Hospital of Shanxi Medical University, Taiyuan, 030001, Shanxi, PR China; Shanxi Medical University, Taiyuan, 030001, Shanxi, PR China
| | - Xi Jie
- The Second Hospital of Shanxi Medical University, Taiyuan, 030001, Shanxi, PR China; Shanxi Medical University, Taiyuan, 030001, Shanxi, PR China
| | - Zong-Feng Zhu
- The Second Hospital of Shanxi Medical University, Taiyuan, 030001, Shanxi, PR China; Shanxi Medical University, Taiyuan, 030001, Shanxi, PR China
| | - Xiao-Juan Zhang
- The Second Hospital of Shanxi Medical University, Taiyuan, 030001, Shanxi, PR China; Shanxi Medical University, Taiyuan, 030001, Shanxi, PR China; Shanxi Province Cardiovascular Hospital, Taiyuan, 030024, Shanxi, PR China
| | - Hui-Ping Zhao
- The Second Hospital of Shanxi Medical University, Taiyuan, 030001, Shanxi, PR China; Shanxi Medical University, Taiyuan, 030001, Shanxi, PR China
| | - Rui-Fang Chi
- The Second Hospital of Shanxi Medical University, Taiyuan, 030001, Shanxi, PR China; Shanxi Medical University, Taiyuan, 030001, Shanxi, PR China
| | - Bao Li
- The Second Hospital of Shanxi Medical University, Taiyuan, 030001, Shanxi, PR China; Shanxi Medical University, Taiyuan, 030001, Shanxi, PR China
| | - Fu-Zhong Qin
- The Second Hospital of Shanxi Medical University, Taiyuan, 030001, Shanxi, PR China; Shanxi Medical University, Taiyuan, 030001, Shanxi, PR China.
| | - Jia-Pu Wang
- Shanxi Medical University, Taiyuan, 030001, Shanxi, PR China; Shanxi Province Cardiovascular Hospital, Taiyuan, 030024, Shanxi, PR China
| | - Ke Wang
- The Second Hospital of Shanxi Medical University, Taiyuan, 030001, Shanxi, PR China; Shanxi Medical University, Taiyuan, 030001, Shanxi, PR China
| |
Collapse
|
16
|
Katsur M, He Z, Vinokur V, Corteling R, Yellon DM, Davidson SM. Exosomes from neuronal stem cells may protect the heart from ischaemia/reperfusion injury via JAK1/2 and gp130. J Cell Mol Med 2021; 25:4455-4465. [PMID: 33797200 PMCID: PMC8093960 DOI: 10.1111/jcmm.16515] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 03/17/2021] [Accepted: 03/22/2021] [Indexed: 12/22/2022] Open
Abstract
Myocardial infarction requires urgent reperfusion to salvage viable heart tissue. However, reperfusion increases infarct size further by promoting mitochondrial damage in cardiomyocytes. Exosomes from a wide range of different cell sources have been shown to activate cardioprotective pathways in cardiomyocytes, thereby reducing infarct size. Yet, it is currently challenging to obtain highly pure exosomes in quantities enough for clinical studies. To overcome this problem, we used exosomes isolated from CTX0E03 neuronal stem cells, which are genetically stable, conditionally inducible and can be produced on an industrial scale. However, it is unknown whether exosomes from neuronal stem cells may reduce cardiac ischaemia/reperfusion injury. In this study, we demonstrate that exosomes from differentiating CTX0E03 cells can reduce infarct size in mice. In an in vitro assay, these exosomes delayed cardiomyocyte mitochondrial permeability transition pore opening, which is responsible for cardiomyocyte death after reperfusion. The mechanism of MPTP inhibition was via gp130 signalling and the downstream JAK/STAT pathway. Our results support previous findings that exosomes from non‐cardiomyocyte‐related cells produce exosomes capable of protecting cardiomyocytes from myocardial infarction. We anticipate our findings may encourage scientists to use exosomes obtained from reproducible clinical‐grade stocks of cells for their ischaemia/reperfusion studies.
Collapse
Affiliation(s)
- Miroslava Katsur
- The Hatter Cardiovascular Institute, University College London, London, UK
| | - Zhenhe He
- The Hatter Cardiovascular Institute, University College London, London, UK
| | - Vladimir Vinokur
- Department of Biochemistry and Molecular Biology, Institute of Medical Research Israel-Canada, The Hebrew University of Jerusalem, Jerusalem, Israel
| | | | - Derek M Yellon
- The Hatter Cardiovascular Institute, University College London, London, UK
| | - Sean M Davidson
- The Hatter Cardiovascular Institute, University College London, London, UK
| |
Collapse
|
17
|
Nieuwenhuijs-Moeke GJ, Bosch DJ, Leuvenink HG. Molecular Aspects of Volatile Anesthetic-Induced Organ Protection and Its Potential in Kidney Transplantation. Int J Mol Sci 2021; 22:ijms22052727. [PMID: 33800423 PMCID: PMC7962839 DOI: 10.3390/ijms22052727] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 02/25/2021] [Accepted: 03/03/2021] [Indexed: 12/16/2022] Open
Abstract
Ischemia reperfusion injury (IRI) is inevitable in kidney transplantation and negatively impacts graft and patient outcome. Reperfusion takes place in the recipient and most of the injury following ischemia and reperfusion occurs during this reperfusion phase; therefore, the intra-operative period seems an attractive window of opportunity to modulate IRI and improve short- and potentially long-term graft outcome. Commonly used volatile anesthetics such as sevoflurane and isoflurane have been shown to interfere with many of the pathophysiological processes involved in the injurious cascade of IRI. Therefore, volatile anesthetic (VA) agents might be the preferred anesthetics used during the transplantation procedure. This review highlights the molecular and cellular protective points of engagement of VA shown in in vitro studies and in vivo animal experiments, and the potential translation of these results to the clinical setting of kidney transplantation.
Collapse
Affiliation(s)
- Gertrude J. Nieuwenhuijs-Moeke
- Department of Anesthesiology, University Medical Centre Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands;
- Correspondence: ; Tel.: +31-631623075
| | - Dirk J. Bosch
- Department of Anesthesiology, University Medical Centre Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands;
| | - Henri G.D. Leuvenink
- Department of Surgery, University Medical Centre Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands;
| |
Collapse
|
18
|
Morris G, Puri BK, Bortolasci CC, Carvalho A, Berk M, Walder K, Moreira EG, Maes M. The role of high-density lipoprotein cholesterol, apolipoprotein A and paraoxonase-1 in the pathophysiology of neuroprogressive disorders. Neurosci Biobehav Rev 2021; 125:244-263. [PMID: 33657433 DOI: 10.1016/j.neubiorev.2021.02.037] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 01/29/2021] [Accepted: 02/23/2021] [Indexed: 12/11/2022]
Abstract
Lowered high-density lipoprotein (HDL) cholesterol has been reported in major depressive disorder, bipolar disorder, first episode of psychosis, and schizophrenia. HDL, its major apolipoprotein component, ApoA1, and the antioxidant enzyme paraoxonase (PON)1 (which is normally bound to ApoA1) all have anti-atherogenic, antioxidant, anti-inflammatory, and immunomodulatory roles, which are discussed in this paper. The paper details the pathways mediating the anti-inflammatory effects of HDL, ApoA1 and PON1 and describes the mechanisms leading to compromised HDL and PON1 levels and function in an environment of chronic inflammation. The molecular mechanisms by which changes in HDL, ApoA1 and PON1 might contribute to the pathophysiology of the neuroprogressive disorders are explained. Moreover, the anti-inflammatory actions of ApoM-mediated sphingosine 1-phosphate (S1P) signalling are reviewed as well as the deleterious effects of chronic inflammation and oxidative stress on ApoM/S1P signalling. Finally, therapeutic interventions specifically aimed at improving the levels and function of HDL and PON1 while reducing levels of inflammation and oxidative stress are considered. These include the so-called Mediterranean diet, extra virgin olive oil, polyphenols, flavonoids, isoflavones, pomegranate juice, melatonin and the Mediterranean diet combined with the ketogenic diet.
Collapse
Affiliation(s)
- Gerwyn Morris
- Deakin University, IMPACT - The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia
| | | | - Chiara C Bortolasci
- Deakin University, IMPACT - The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia; Deakin University, CMMR Strategic Research Centre, School of Medicine, Geelong, Victoria, Australia.
| | - Andre Carvalho
- Deakin University, IMPACT - The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia; Department of Psychiatry, University of Toronto, Toronto, ON, Canada; Centre for Addiction and Mental Health (CAMH), Toronto, ON, Canada
| | - Michael Berk
- Deakin University, IMPACT - The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia; Orygen, The National Centre of Excellence in Youth Mental Health, The Department of Psychiatry and The Florey Institute for Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria, Australia
| | - Ken Walder
- Deakin University, IMPACT - The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia; Deakin University, CMMR Strategic Research Centre, School of Medicine, Geelong, Victoria, Australia
| | - Estefania G Moreira
- Post-Graduation Program in Health Sciences, State University of Londrina, Londrina, PR, Brazil
| | - Michael Maes
- Deakin University, IMPACT - The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia; Department of Psychiatry, King Chulalongkorn University Hospital, Bangkok, Thailand; Department of Psychiatry, Medical University of Plovdiv, Plovdiv, Bulgaria
| |
Collapse
|
19
|
Cirillo F, Piccoli M, Ghiroldi A, Monasky MM, Rota P, La Rocca P, Tarantino A, D'Imperio S, Signorelli P, Pappone C, Anastasia L. The antithetic role of ceramide and sphingosine-1-phosphate in cardiac dysfunction. J Cell Physiol 2021; 236:4857-4873. [PMID: 33432663 DOI: 10.1002/jcp.30235] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 12/09/2020] [Accepted: 12/11/2020] [Indexed: 12/27/2022]
Abstract
Cardiovascular diseases (CVDs) are the leading cause of death globally and the number of cardiovascular patients, which is estimated to be over 30 million in 2018, represent a challenging issue for the healthcare systems worldwide. Therefore, the identification of novel molecular targets to develop new treatments is an ongoing challenge for the scientific community. In this context, sphingolipids (SLs) have been progressively recognized as potent bioactive compounds that play crucial roles in the modulation of several key biological processes, such as proliferation, differentiation, and apoptosis. Furthermore, SLs involvement in cardiac physiology and pathophysiology attracted much attention, since these molecules could be crucial in the development of CVDs. Among SLs, ceramide and sphingosine-1-phosphate (S1P) represent the most studied bioactive lipid mediators, which are characterized by opposing activities in the regulation of the fate of cardiac cells. In particular, maintaining the balance of the so-called ceramide/S1P rheostat emerged as an important novel therapeutical target to counteract CVDs. Thus, this review aims at critically summarizing the current knowledge about the antithetic roles of ceramide and S1P in cardiomyocytes dysfunctions, highlighting how the modulation of their metabolism through specific molecules, such as myriocin and FTY720, could represent a novel and interesting therapeutic approach to improve the management of CVDs.
Collapse
Affiliation(s)
- Federica Cirillo
- Laboratory of Stem Cells for Tissue Engineering, IRCCS Policlinico San Donato, Milan, Italy
| | - Marco Piccoli
- Laboratory of Stem Cells for Tissue Engineering, IRCCS Policlinico San Donato, Milan, Italy
| | - Andrea Ghiroldi
- Laboratory of Stem Cells for Tissue Engineering, IRCCS Policlinico San Donato, Milan, Italy
| | | | - Paola Rota
- Department of Biomedical, Surgical and Dental Sciences, University of Milan, Milan, Italy
| | - Paolo La Rocca
- Department of Biomedical Sciences for Health, University of Milan, Milan, Italy
| | - Adriana Tarantino
- Laboratory of Stem Cells for Tissue Engineering, IRCCS Policlinico San Donato, Milan, Italy.,Department of Arrhythmology, IRCCS Policlinico San Donato, Milan, Italy
| | - Sara D'Imperio
- Laboratory of Stem Cells for Tissue Engineering, IRCCS Policlinico San Donato, Milan, Italy.,Department of Arrhythmology, IRCCS Policlinico San Donato, Milan, Italy
| | - Paola Signorelli
- Department of Health Sciences, Biochemistry and Molecular Biology Laboratory, University of Milan, Milan, Italy
| | - Carlo Pappone
- Department of Arrhythmology, IRCCS Policlinico San Donato, Milan, Italy.,Faculty of Medicine and Surgery, University of Vita-Salute San Raffaele, Milan, Italy
| | - Luigi Anastasia
- Laboratory of Stem Cells for Tissue Engineering, IRCCS Policlinico San Donato, Milan, Italy.,Faculty of Medicine and Surgery, University of Vita-Salute San Raffaele, Milan, Italy
| |
Collapse
|
20
|
Ticagrelor Conditioning Effects Are Not Additive to Cardioprotection Induced by Direct NLRP3 Inflammasome Inhibition: Role of RISK, NLRP3, and Redox Cascades. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:9219825. [PMID: 32832010 PMCID: PMC7424511 DOI: 10.1155/2020/9219825] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 06/07/2020] [Accepted: 06/26/2020] [Indexed: 12/11/2022]
Abstract
Inhibition of either P2Y12 receptor or the nucleotide-binding oligomerization domain- (NOD-) like receptor pyrin domain containing 3 (NLRP3) inflammasome provides cardioprotective effects. Here, we investigate whether direct NLRP3 inflammasome inhibition exerts additive effects on myocardial protection induced by the P2Y12 receptor antagonist Ticagrelor. Ticagrelor (150 mg/kg) was orally administered to rats for three consecutive days. Then, isolated hearts underwent an ischemia/reperfusion (30 min ischemia/60 min reperfusion; IR) protocol. The selective NLRP3 inflammasome inhibitor INF (50 μM) was infused before the IR protocol to the hearts from untreated animals or pretreated with Ticagrelor. In parallel experiments, the hearts isolated from untreated animals were perfused with Ticagrelor (3.70 μM) before ischemia and subjected to IR. The hearts of animals pretreated with Ticagrelor showed a significantly reduced infarct size (IS, 49 ± 3% of area at risk, AAR) when compared to control IR group (69 ± 2% of AAR). Similarly, ex vivo administration of INF before the IR injury resulted in significant IS reduction (38 ± 3% of AAR). Myocardial IR induced the NLRP3 inflammasome complex formation, which was attenuated by either INF pretreatment ex vivo, or by repeated oral treatment with Ticagrelor. The beneficial effects induced by either treatment were associated with the protective Reperfusion Injury Salvage Kinase (RISK) pathway activation and redox defence upregulation. In contrast, no protective effects nor NLRP3/RISK modulation were recorded when Ticagrelor was administered before ischemia in isolated heart, indicating that Ticagrelor direct target is not in the myocardium. Our results confirm that Ticagrelor conditioning effects are likely mediated through platelets, but are not additives to the ones achieved by directly inhibiting NLRP3.
Collapse
|
21
|
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: 202] [Impact Index Per Article: 40.4] [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
|
22
|
Jozefczuk E, Guzik TJ, Siedlinski M. Significance of sphingosine-1-phosphate in cardiovascular physiology and pathology. Pharmacol Res 2020; 156:104793. [PMID: 32278039 DOI: 10.1016/j.phrs.2020.104793] [Citation(s) in RCA: 124] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Revised: 03/27/2020] [Accepted: 03/27/2020] [Indexed: 02/07/2023]
Abstract
Sphingosine-1-phosphate (S1P) is a signaling lipid, synthetized by sphingosine kinases (SPHK1 and SPHK2), that affects cardiovascular function in various ways. S1P signaling is complex, particularly since its molecular action is reliant on the differential expression of its receptors (S1PR1, S1PR2, S1PR3, S1PR4, S1PR5) within various tissues. Significance of this sphingolipid is manifested early in vertebrate development as certain defects in S1P signaling result in embryonic lethality due to defective vasculo- or cardiogenesis. Similar in the mature organism, S1P orchestrates both physiological and pathological processes occurring in the heart and vasculature of higher eukaryotes. S1P regulates cell fate, vascular tone, endothelial function and integrity as well as lymphocyte trafficking, thus disbalance in its production and signaling has been linked with development of such pathologies as arterial hypertension, atherosclerosis, endothelial dysfunction and aberrant angiogenesis. Number of signaling mechanisms are critical - from endothelial nitric oxide synthase through STAT3, MAPK and Akt pathways to HDL particles involved in redox and inflammatory balance. Moreover, S1P controls both acute cardiac responses (cardiac inotropy and chronotropy), as well as chronic processes (such as apoptosis and hypertrophy), hence numerous studies demonstrate significance of S1P in the pathogenesis of hypertrophic/fibrotic heart disease, myocardial infarction and heart failure. This review presents current knowledge concerning the role of S1P in the cardiovascular system, as well as potential therapeutic approaches to target S1P signaling in cardiovascular diseases.
Collapse
Affiliation(s)
- E Jozefczuk
- Department of Internal and Agricultural Medicine, Faculty of Medicine, Jagiellonian University Medical College, Cracow, Poland
| | - T J Guzik
- Department of Internal and Agricultural Medicine, Faculty of Medicine, Jagiellonian University Medical College, Cracow, Poland; Institute of Cardiovascular and Medical Sciences, BHF Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, UK
| | - M Siedlinski
- Department of Internal and Agricultural Medicine, Faculty of Medicine, Jagiellonian University Medical College, Cracow, Poland; Institute of Cardiovascular and Medical Sciences, BHF Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, UK.
| |
Collapse
|
23
|
Raza Z, Saleem U, Naureen Z. Sphingosine 1-phosphate signaling in ischemia and reperfusion injury. Prostaglandins Other Lipid Mediat 2020; 149:106436. [PMID: 32173486 DOI: 10.1016/j.prostaglandins.2020.106436] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 02/28/2020] [Accepted: 03/02/2020] [Indexed: 02/07/2023]
Abstract
Ischemia and reperfusion injury is a complex hemodynamic pathological phenomenon that engages the metabolic to inflammatory machinery in development of disease conditions like heart failure, stroke and acute kidney failure. Target specific therapeutic approaches for ischemia reperfusion injury remains critical despite the extensive studies contributing to the understanding of its pathogenesis. Ischemic or pharmacological conditionings have been long established manipulations to harness the endogenous protective mechanisms against ischemia reperfusion injury that fostered the development of potential therapeutic targets such as sphingolipids signaling. Sphingosine 1-phosphate has been emerged as a crucial metabolite of sphingolipids to regulate the cell survival, vascular integrity and inflammatory cascades in ischemia reperfusion injury. Sphingosine 1-phosphate signaling process has been implicated to downgrade the mitochondrial dysfunction, apoptotic assembly along with upregulation of RISK and SAFE pro-survival pathways. It also regulates the endothelial dysfunction and immune cells behavior to control the vascular permeability and immune cells infiltration at ischemia reperfusion injury site. Targeting the signaling of this single moiety holds the vast potential to extensively influence the detrimental signaling of ischemia reperfusion injury. This review highlights the role and significance of S1P signaling that can be therapeutically exploit to treat ischemia reperfusion injury mediated pathological conditions in different organs.
Collapse
Affiliation(s)
- Zohaib Raza
- Government College University, Faisalabad, Pakistan.
| | - Uzma Saleem
- Government College University, Faisalabad, Pakistan
| | | |
Collapse
|
24
|
Plouffe B, Thomsen ARB, Irannejad R. Emerging Role of Compartmentalized G Protein-Coupled Receptor Signaling in the Cardiovascular Field. ACS Pharmacol Transl Sci 2020; 3:221-236. [PMID: 32296764 PMCID: PMC7155194 DOI: 10.1021/acsptsci.0c00006] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2020] [Indexed: 02/06/2023]
Abstract
G protein-coupled receptors (GPCRs) are cell surface receptors that for many years have been considered to function exclusively at the plasma membrane, where they bind to extracellular ligands and activate G protein signaling cascades. According to the conventional model, these signaling events are rapidly terminated by β-arrestin (β-arr) recruitment to the activated GPCR resulting in signal desensitization and receptor internalization. However, during the past decade, emerging evidence suggest that many GPCRs can continue to activate G proteins from intracellular compartments after they have been internalized. G protein signaling from intracellular compartments is in general more sustained compared to G protein signaling at the plasma membrane. Notably, the particular location closer to the nucleus is beneficial for selective cellular functions such as regulation of gene transcription. Here, we review key GPCRs that undergo compartmentalized G protein signaling and discuss molecular considerations and requirements for this signaling to occur. Our main focus will be on receptors involved in the regulation of important physiological and pathological cardiovascular functions. We also discuss how sustained G protein activation from intracellular compartments may be involved in cellular functions that are distinct from functions regulated by plasma membrane G protein signaling, and the corresponding significance in cardiovascular physiology.
Collapse
Affiliation(s)
- Bianca Plouffe
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, Belfast BT9 7BL, United Kingdom
| | - Alex R B Thomsen
- Department of Basic Science and Craniofacial Biology, NYU College of Dentistry, New York, New York 10010, United States
| | - Roshanak Irannejad
- Department of Biochemistry and Biophysics, Cardiovascular Research Institute, University of California, San Francisco, California 94158, United States
| |
Collapse
|
25
|
Abstract
Acute myocardial infarction is a major cause of death and disability worldwide. This study was designed to elucidate the effect of resveratrol (RES) in isoproterenol (ISO)-challenged myocardial injury in rats. Male Sprague-Dawley rats were randomly allocated to four groups (10 rats/group): negative, control positive ISO (85 mg/kg), Propranolol/ISO, and RES/ISO. RES (50 mg/kg) improved plasma lactate dehydrogenase, creatine kinase, and cardiac troponin T; brain natriuretic peptide, interleukin-10, vascular endothelial growth factor, and transforming growth factor-β1; as well as cardiac superoxide dismutase, malondialdehyde, and total protein kinase-1 (Akt-1) levels. In addition, RES reduced the expression of cardiac inducible nitric oxide synthase and microRNA-34a, as well as p38 mitogen-activated protein kinase levels compared with positive control group. In conclusion, RES could reduce the degree of MI induced by ISO by improving the antioxidant, antiapoptotic, and anti-inflammatory capacities of the body.
Collapse
Affiliation(s)
- Sylvia A Boshra
- Department of Biochemistry, Faculty of Pharmacy, October 6 University, Giza, Egypt
| |
Collapse
|
26
|
Harhous Z, Booz GW, Ovize M, Bidaux G, Kurdi M. An Update on the Multifaceted Roles of STAT3 in the Heart. Front Cardiovasc Med 2019; 6:150. [PMID: 31709266 PMCID: PMC6823716 DOI: 10.3389/fcvm.2019.00150] [Citation(s) in RCA: 94] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2019] [Accepted: 10/07/2019] [Indexed: 12/18/2022] Open
Abstract
Signal transducer and activator of transcription 3 (STAT3) is a signaling molecule and transcription factor that plays important protective roles in the heart. The protection mediated by STAT3 is attributed to its genomic actions as a transcription factor and other non-genomic roles targeting mitochondrial function and autophagy. As a transcription factor, STAT3 upregulates genes that are anti-oxidative, anti-apoptotic, and pro-angiogenic, but suppresses anti-inflammatory and anti-fibrotic genes. Its suppressive effects on gene expression are achieved through competing with other transcription factors or cofactors. STAT3 is also linked to the modification of mRNA expression profiles in cardiac cells by inhibiting or inducing miRNA. In addition to these genomic roles, STAT3 is suggested to function protectively in mitochondria, where it regulates ROS production, in part by regulating the activities of the electron transport chain complexes, although our recent evidence calls this role into question. Nonetheless, STAT3 is a key player known to be activated in the cardioprotective ischemic conditioning protocols. Through these varied roles, STAT3 participates in various mechanisms that contribute to cardioprotection against different heart pathologies, including myocardial infarction, hypertrophy, diabetic cardiomyopathy, and peripartum cardiomyopathy. Understanding how STAT3 is involved in the protective mechanisms against these different cardiac pathologies could lead to novel therapeutic strategies to treat them.
Collapse
Affiliation(s)
- Zeina Harhous
- Laboratory of Experimental and Clinical Pharmacology, Faculty of Sciences, Doctoral School of Sciences and Technology, Lebanese University, Beirut, Lebanon
- Univ-Lyon, CarMeN Laboratory, INSERM 1060, INRA 1397, University Claude Bernard Lyon1, INSA Lyon, Oullins, France
- IHU OPeRa, Groupement Hospitalier EST, Bron, France
| | - George W. Booz
- Department of Pharmacology and Toxicology, School of Medicine, The University of Mississippi Medical Center, Jackson, MS, United States
| | - Michel Ovize
- Univ-Lyon, CarMeN Laboratory, INSERM 1060, INRA 1397, University Claude Bernard Lyon1, INSA Lyon, Oullins, France
- IHU OPeRa, Groupement Hospitalier EST, Bron, France
| | - Gabriel Bidaux
- Univ-Lyon, CarMeN Laboratory, INSERM 1060, INRA 1397, University Claude Bernard Lyon1, INSA Lyon, Oullins, France
- IHU OPeRa, Groupement Hospitalier EST, Bron, France
| | - Mazen Kurdi
- Laboratory of Experimental and Clinical Pharmacology, Faculty of Sciences, Doctoral School of Sciences and Technology, Lebanese University, Beirut, Lebanon
| |
Collapse
|
27
|
Caccioppo A, Franchin L, Grosso A, Angelini F, D'Ascenzo F, Brizzi MF. Ischemia Reperfusion Injury: Mechanisms of Damage/Protection and Novel Strategies for Cardiac Recovery/Regeneration. Int J Mol Sci 2019; 20:E5024. [PMID: 31614414 PMCID: PMC6834134 DOI: 10.3390/ijms20205024] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Revised: 09/24/2019] [Accepted: 10/08/2019] [Indexed: 12/11/2022] Open
Abstract
Ischemic diseases in an aging population pose a heavy social encumbrance. Moreover, current therapeutic approaches, which aimed to prevent or minimize ischemia-induced damage, are associated with relevant costs for healthcare systems. Early reperfusion by primary percutaneous coronary intervention (PPCI) has undoubtedly improved patient's outcomes; however, the prevention of long-term complications is still an unmet need. To face these hurdles and improve patient's outcomes, novel pharmacological and interventional approaches, alone or in combination, reducing myocardium oxygen consumption or supplying blood flow via collateral vessels have been proposed. A number of clinical trials are ongoing to validate their efficacy on patient's outcomes. Alternative options, including stem cell-based therapies, have been evaluated to improve cardiac regeneration and prevent scar formation. However, due to the lack of long-term engraftment, more recently, great attention has been devoted to their paracrine mediators, including exosomes (Exo) and microvesicles (MV). Indeed, Exo and MV are both currently considered to be one of the most promising therapeutic strategies in regenerative medicine. As a matter of fact, MV and Exo that are released from stem cells of different origin have been evaluated for their healing properties in ischemia reperfusion (I/R) settings. Therefore, this review will first summarize mechanisms of cardiac damage and protection after I/R damage to track the paths through which more appropriate interventional and/or molecular-based targeted therapies should be addressed. Moreover, it will provide insights on novel non-invasive/invasive interventional strategies and on Exo-based therapies as a challenge for improving patient's long-term complications. Finally, approaches for improving Exo healing properties, and topics still unsolved to move towards Exo clinical application will be discussed.
Collapse
Affiliation(s)
- Andrea Caccioppo
- Department of Medical Sciences, University of Turin, 10124 Torino, Italy.
| | - Luca Franchin
- Division of Cardiology, Department of Medical Sciences, University of Turin, 10124 Torino, Italy.
| | - Alberto Grosso
- Department of Medical Sciences, University of Turin, 10124 Torino, Italy.
| | - Filippo Angelini
- Division of Cardiology, Department of Medical Sciences, University of Turin, 10124 Torino, Italy.
| | - Fabrizio D'Ascenzo
- Division of Cardiology, Department of Medical Sciences, University of Turin, 10124 Torino, Italy.
| | | |
Collapse
|
28
|
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: 2.0] [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
|
29
|
Shu Z, Yang Y, Yang L, Jiang H, Yu X, Wang Y. Cardioprotective effects of dihydroquercetin against ischemia reperfusion injury by inhibiting oxidative stress and endoplasmic reticulum stress-induced apoptosis via the PI3K/Akt pathway. Food Funct 2019; 10:203-215. [PMID: 30525169 DOI: 10.1039/c8fo01256c] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Dihydroquercetin (DHQ), a dihydroxyflavone, possesses potent antioxidant properties and is proposed to be useful in the prevention and treatment of cardiovascular disease. In this study, we aimed to investigate whether DHQ has protective effects against MIRI and to elucidate the mechanism of attenuation of oxidative stress-and ERS-induced apoptosis via the PI3K/Akt pathway. Isolated rat hearts and H9c2 cardiomyocytes were treated with or without DHQ, and then subjected to I/R and H/R, respectively. Our results showed that DHQ pretreatment markedly alleviated cardiac dysfunction, scavenged free radicals, reduced lipid peroxidation, and increased the activity of antioxidant enzymes ex vivo and in vitro. The result of western blot analysis showed that DHQ inhibited the apoptotic pathway and the expression of pro-apoptotic proteins CHOP, Caspase-12, and p-JNK. In addition, DHQ delayed the onset of ERs by reducing GRP78, p-PERK, and p-eif2α expression levels and by increasing HO-1 expression and Nrf2 binding to antioxidant response elements. These cardioprotective effects of DHQ were partially counteracted by the PI3K/Akt inhibitor LY294002. The protective effects of DHQ on the inhibition of MIRI may be mediated by activating the PI3K/Akt pathway to reduce oxidative stress-and ERS-induced apoptosis.
Collapse
Affiliation(s)
- Zunpeng Shu
- Guangdong Pharmaceutical University, Guangzhou 510006, P.R. China.
| | | | | | | | | | | |
Collapse
|
30
|
Ke M, Tang Q, Pan Z, Yin Y, Zhang L, Wen K. Sphingosine-1-phosphate attenuates hypoxia/reoxygenation-induced cardiomyocyte injury via a mitochondrial pathway. Biochem Biophys Res Commun 2019; 510:142-148. [PMID: 30661785 DOI: 10.1016/j.bbrc.2019.01.067] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2018] [Accepted: 01/13/2019] [Indexed: 12/14/2022]
Abstract
Our previous study showed that Sphingosine-1-phosphate (S1P) could protect cardiomyocytes against hypoxia/reoxygenation (H/R) injury via the JAK-STAT pathway and maintain normal myocardial mitochondria integrity in vivo. However, it is not known yet whether S1P can relieve mitochondrial dysfunction via the mitochondrial apoptotic pathway and its detailed mechanism remains to be investigated. The aim of this study was to demonstrate the mitochondrial protective effects of S1P in a cardiomyocyte H/R injury model. In the present study, we established a H/R model in H9c2 cells. Cell viability was determined by the MTT assay, and apoptosis was evaluated by annexin V-FITC/PI staining. Mitochondrial calcium ion concentration, mitochondrial membrane potential (ΔΨm), opening of the mitochondrial permeability transition pore (mPTP), and release of cytochrome C were detected by laser confocal microscopy. The results showed that S1P inhibited the decrease in cell viability induced by H/R injury and reduced apoptosis. Confocal microscopy showed that S1P prevented loss of ΔΨm, relieved mitochondrial calcium overload, and inhibited opening of the mPTP and release of cytochrome C. The STAT3 inhibitor STATTIC can reverse the antiapoptotic effects of S1P and block the effect of S1P on mitochondria. Taken together, our results indicate that S1P protects cardiomyocytes against H/R injury by relieving mitochondrial dysfunction via the STAT3 pathway.
Collapse
Affiliation(s)
- Mengran Ke
- Department of Pharmacology, College of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Qiqi Tang
- Department of Pharmacology, College of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Ziang Pan
- Department of Pharmacology, College of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Yongqiang Yin
- Department of Pharmacology, College of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Lizhi Zhang
- Department of Obstetrics and Gynecology, Tianjin First Centre Hospital, Tianjin, China.
| | - Ke Wen
- Department of Pharmacology, College of Basic Medical Sciences, Tianjin Medical University, Tianjin, China.
| |
Collapse
|
31
|
Involvement of sphingosine-1-phosphate receptors 2/3 in IR-induced sudden cardiac death. Heart Vessels 2019; 34:1052-1063. [DOI: 10.1007/s00380-018-01323-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Accepted: 12/21/2018] [Indexed: 12/22/2022]
|
32
|
Rosenberg JH, Werner JH, Moulton MJ, Agrawal DK. Current Modalities and Mechanisms Underlying Cardioprotection by Ischemic Conditioning. J Cardiovasc Transl Res 2018; 11:292-307. [PMID: 29797232 PMCID: PMC6117210 DOI: 10.1007/s12265-018-9813-1] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Accepted: 05/14/2018] [Indexed: 02/07/2023]
Abstract
Ischemic preconditioning is a process which serves to mitigate reperfusion injury. Preconditioning of the heart can be achieved through natural, pharmacological, and mechanical means. Mechanical preconditioning appears to have the greatest chance of good outcomes while methods employing pharmacologic preconditioning have been largely unsuccessful. Remote ischemic preconditioning achieves a cardioprotective effect by applying cycles of ischemia and reperfusion in a distal limb, stimulating the release of a neurohumoral cardioprotective factor incited by stimulation of afferent neurons. The cardioprotective factor stimulates the reperfusion injury salvage kinase (RISK) and survivor activator factor enhancement (SAFE) signaling cascades in cardiomyocytes which promote cell survival by the expression of anti-apoptotic genes and inhibition of the opening of mitochondrial permeability transition pores. Clinical application of ischemic preconditioning involving targets in the RISK and SAFE signaling appears promising in the treatment of acute myocardial infarction; however, clinical trials have yet to demonstrate additional benefit to current therapy.
Collapse
Affiliation(s)
- John H Rosenberg
- Department of Clinical & Translational Science, The Peekie Nash Carpenter Endowed Chair in Medicine, Creighton University School of Medicine, CRISS II Room 510, 2500 California Plaza, Omaha, NE, 68178, USA
| | - John H Werner
- Department of Clinical & Translational Science, The Peekie Nash Carpenter Endowed Chair in Medicine, Creighton University School of Medicine, CRISS II Room 510, 2500 California Plaza, Omaha, NE, 68178, USA
| | - Michael J Moulton
- Department of Cardiothoracic Surgery, University of Nebraska Medical Center, Omaha, NE, USA
| | - Devendra K Agrawal
- Department of Clinical & Translational Science, The Peekie Nash Carpenter Endowed Chair in Medicine, Creighton University School of Medicine, CRISS II Room 510, 2500 California Plaza, Omaha, NE, 68178, USA.
| |
Collapse
|
33
|
Russo I, Femminò S, Barale C, Tullio F, Geuna S, Cavalot F, Pagliaro P, Penna C. Cardioprotective Properties of Human Platelets Are Lost in Uncontrolled Diabetes Mellitus: A Study in Isolated Rat Hearts. Front Physiol 2018; 9:875. [PMID: 30042694 PMCID: PMC6048273 DOI: 10.3389/fphys.2018.00875] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Accepted: 06/19/2018] [Indexed: 12/13/2022] Open
Abstract
Platelets affect myocardial damage from ischemia/reperfusion. Redox-dependent sphingosine-1-phosphate production and release are altered in diabetic platelets. Sphingosine-1-phosphate is a double-edged sword for ischemia/reperfusion injury. Therefore, we aimed to verify whether: (1) human healthy- or diabetic-platelets are cardioprotective, (2) sphingosine-1-phosphate receptors and downstream kinases play a role in platelet-induced cardioprotection, and (3) a correlation between platelet redox status and myocardial ischemia/reperfusion injury exists. Isolated rat hearts were subjected to 30-min ischemia and 1-h reperfusion. Infarct size was studied in hearts pretreated with healthy- or diabetic-platelets. Healthy-platelets were co-infused with sphingosine-1-phosphate receptor blocker, ERK-1/2 inhibitor, PI3K antagonist or PKC inhibitor to ascertain the cardioprotective mechanisms. In platelets we assessed (i) aggregation response to ADP, collagen, and arachidonic-acid, (ii) cyclooxygenase-1 levels, and (iii) AKT and ERK-phosphorylation. Platelet sphingosine-1-phosphate production and platelet levels of reactive oxygen species (ROS) were quantified and correlated to infarct size. Infarct size was reduced by about 22% in healthy-platelets pretreated hearts only. This cardioprotective effect was abrogated by either sphingosine-1-phosphate receptors or ERK/PI3K/PKC pathway blockade. Cyclooxygenase-1 levels and aggregation indices were higher in diabetic-platelets than healthy-platelets. Diabetic-platelets released less sphingosine-1-phosphate than healthy-platelets when mechanical or chemically stimulated in vitro. Yet, ROS levels were higher in diabetic-platelets and correlated with infarct size. Cardioprotective effects of healthy-platelet depend on the platelet’s capacity to activate cardiac sphingosine-1-phosphate receptors and ERK/PI3K/PKC pathways. However, diabetic-platelets release less S1P and lose cardioprotective effects. Platelet ROS levels correlate with infarct size. Whether these redox alterations are responsible for sphingosine-1-phosphate dysfunction in diabetic-platelets remains to be ascertained.
Collapse
Affiliation(s)
- Isabella Russo
- Department of Clinical and Biological Sciences, AOU San Luigi, University of Turin, Turin, Italy
| | - Saveria Femminò
- Department of Clinical and Biological Sciences, AOU San Luigi, University of Turin, Turin, Italy.,Istituto Nazionale Ricerche Cardiovascolari (INRC), Bologna, Italy
| | - Cristina Barale
- Department of Clinical and Biological Sciences, AOU San Luigi, University of Turin, Turin, Italy
| | - Francesca Tullio
- Department of Clinical and Biological Sciences, AOU San Luigi, University of Turin, Turin, Italy.,Istituto Nazionale Ricerche Cardiovascolari (INRC), Bologna, Italy
| | - Stefano Geuna
- Department of Clinical and Biological Sciences, AOU San Luigi, University of Turin, Turin, Italy
| | - Franco Cavalot
- Internal Medicine and Metabolic Disease Unit, AOU San Luigi, University of Turin, Turin, Italy.,Ospedale San Luigi Gonzaga, Orbassano, Italy
| | - Pasquale Pagliaro
- Department of Clinical and Biological Sciences, AOU San Luigi, University of Turin, Turin, Italy.,Istituto Nazionale Ricerche Cardiovascolari (INRC), Bologna, Italy
| | - Claudia Penna
- Department of Clinical and Biological Sciences, AOU San Luigi, University of Turin, Turin, Italy.,Istituto Nazionale Ricerche Cardiovascolari (INRC), Bologna, Italy
| |
Collapse
|
34
|
Deshpande GP, Imamdin A, Lecour S, Opie LH. Sphingosine-1-phosphate (S1P) activates STAT3 to protect against de novo acute heart failure (AHF). Life Sci 2018; 196:127-132. [DOI: 10.1016/j.lfs.2018.01.023] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Revised: 01/19/2018] [Accepted: 01/22/2018] [Indexed: 01/23/2023]
|
35
|
Rossello X, Riquelme JA, Davidson SM, Yellon DM. Role of PI3K in myocardial ischaemic preconditioning: mapping pro-survival cascades at the trigger phase and at reperfusion. J Cell Mol Med 2018; 22:926-935. [PMID: 29159980 PMCID: PMC5783840 DOI: 10.1111/jcmm.13394] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Accepted: 08/21/2017] [Indexed: 01/16/2023] Open
Abstract
The Reperfusion Injury Salvage Kinase (RISK) pathway is considered the main pro-survival kinase cascade mediating the ischaemic preconditioning (IPC) cardioprotective effect. To assess the role of PI3K-Akt, its negative regulator PTEN and other pro-survival proteins such as ERK and STAT3 in the context of IPC, C57BL/6 mouse hearts were retrogradely perfused in a Langendorff system and subjected to 4 cycles of 5 min. ischaemia and 5 min. reperfusion prior to 35 min. of global ischaemia and 120 min. of reperfusion. Wortmannin, a PI3K inhibitor, was administered either at the stabilization period or during reperfusion. Infarct size was assessed using triphenyl tetrazolium staining, and phosphorylation levels of Akt, PTEN, ERK, GSK3β and STAT3 were evaluated using Western blot analyses. IPC reduced infarct size in hearts subjected to lethal ischaemia and reperfusion, but this effect was lost in the presence of Wortmannin, whether it was present only during preconditioning or only during early reperfusion. IPC increased the levels of Akt phosphorylation during both phases and this effect was fully abrogated by PI3K, whilst its downstream GSK3β was phosphorylated only during the trigger phase after IPC. Both PTEN and STAT3 were phosphorylated during both phases after IPC, but this was PI3K independent. IPC increases ERK phosphorylation during both phases, being only PI3K-dependent during the IPC phase. In conclusion, PI3K-Akt plays a major role in IPC-induced cardioprotection. However, PTEN, ERK and STAT3 are also phosphorylated by IPC through a PI3K-independent pathway, suggesting that cardioprotection is mediated through more than one cell signalling cascade.
Collapse
Affiliation(s)
- Xavier Rossello
- The Hatter Cardiovascular InstituteUniversity College LondonLondonUK
| | - Jaime A Riquelme
- The Hatter Cardiovascular InstituteUniversity College LondonLondonUK
- Advanced Center for Chronic Diseases (ACCDiS)Facultad de Ciencias Quimicas y Farmaceuticas & Facultad de MedicinaUniversidad de ChileSantiagoChile
| | - Sean M Davidson
- The Hatter Cardiovascular InstituteUniversity College LondonLondonUK
| | - Derek M Yellon
- The Hatter Cardiovascular InstituteUniversity College LondonLondonUK
| |
Collapse
|
36
|
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: 7.9] [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]
|
37
|
Ahmed N, Linardi D, Muhammad N, Chiamulera C, Fumagalli G, Biagio LS, Gebrie MA, Aslam M, Luciani GB, Faggian G, Rungatscher A. Sphingosine 1-Phosphate Receptor Modulator Fingolimod (FTY720) Attenuates Myocardial Fibrosis in Post-heterotopic Heart Transplantation. Front Pharmacol 2017; 8:645. [PMID: 28966593 PMCID: PMC5605636 DOI: 10.3389/fphar.2017.00645] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Accepted: 08/31/2017] [Indexed: 12/21/2022] Open
Abstract
Background and Objective: Sphingosine 1-phosphate (S1P), and S1P receptor modulator fingolimod have been suggested to play important cardioprotective role in animal models of myocardial ischemia/reperfusion injuries. To understand the cardioprotective function of S1P and its mechanism in vivo, we analyzed apoptotic, inflammatory biomarkers, and myocardial fibrosis in an in vivo heterotopic rat heart transplantation model. Methods: Heterotopic heart transplantation is performed in 60 Sprague–Dawley (SD) rats (350–400 g). The heart transplant recipients (n = 60) are categorized into Group A (control) and Group B (fingolimod treated 1 mg/kg intravenous). At baseline with 24 h after heart transplantation, blood and myocardial tissue are collected for analysis of myocardial biomarkers, apoptosis, inflammatory markers, oxidative stress, and phosphorylation of Akt/Erk/STAT-3 signaling pathways. Myocardial fibrosis was investigated using Masson’s trichrome staining and L-hydroxyline. Results: Fingolimod treatment activates both Reperfusion Injury Salvage Kinase (RISK) and Survivor Activating Factor Enhancement (SAFE) pathways as evident from activation of anti-apoptotic and anti-inflammatory pathways. Fingolimod treatment caused a reduction in myocardial oxidative stress and hence cardiomyocyte apoptosis resulting in a decrease in myocardial reperfusion injury. Moreover, a significant (p < 0.001) reduction in collagen staining and hydroxyproline content was observed in fingolimod treated animals 30 days after transplantation demonstrating a reduction in cardiac fibrosis. Conclusion: S1P receptor activation with fingolimod activates anti-apoptotic and anti-inflammatory pathways, leading to improved myocardial salvage causing a reduction in cardiac fibrosis.
Collapse
Affiliation(s)
- Naseer Ahmed
- Section of Cardiac Surgery, Department of Surgery, Dentistry, Paediatrics and Gynaecology, University of VeronaVerona, Italy.,Faculty of Health Sciences, University of PunjabLahore, Pakistan.,Research Unit, Faculty of Allied Health Sciences, University of LahoreLahore, Pakistan.,Section of Pharmacology, Department of Diagnostics and Public Health, University of VeronaVerona, Italy
| | - Daniele Linardi
- Section of Cardiac Surgery, Department of Surgery, Dentistry, Paediatrics and Gynaecology, University of VeronaVerona, Italy
| | - Nazeer Muhammad
- COMSATS Institute of Information TechnologyWah Cantt, Pakistan
| | - Cristiano Chiamulera
- Section of Pharmacology, Department of Diagnostics and Public Health, University of VeronaVerona, Italy
| | - Guido Fumagalli
- Section of Pharmacology, Department of Diagnostics and Public Health, University of VeronaVerona, Italy
| | - Livio San Biagio
- Section of Cardiac Surgery, Department of Surgery, Dentistry, Paediatrics and Gynaecology, University of VeronaVerona, Italy
| | - Mebratu A Gebrie
- Section of Cardiac Surgery, Department of Surgery, Dentistry, Paediatrics and Gynaecology, University of VeronaVerona, Italy.,Department of Anatomy, Università di Addis AbebaAddis Ababa, Ethiopia
| | - Muhammad Aslam
- Department of Internal Medicine, Cardiology and Angiology, University Hospital, Justus Liebig UniversityGiessen, Germany
| | - Giovanni Battista Luciani
- Section of Cardiac Surgery, Department of Surgery, Dentistry, Paediatrics and Gynaecology, University of VeronaVerona, Italy
| | - Giuseppe Faggian
- Section of Cardiac Surgery, Department of Surgery, Dentistry, Paediatrics and Gynaecology, University of VeronaVerona, Italy
| | - Alessio Rungatscher
- Section of Cardiac Surgery, Department of Surgery, Dentistry, Paediatrics and Gynaecology, University of VeronaVerona, Italy
| |
Collapse
|
38
|
van Vuuren D, Marais E, Genade S, Lochner A. The differential effects of FTY720 on functional recovery and infarct size following myocardial ischaemia/reperfusion. Cardiovasc J Afr 2017; 27:375-386. [PMID: 27966000 PMCID: PMC5408499 DOI: 10.5830/cvja-2016-039] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2015] [Accepted: 03/30/2016] [Indexed: 01/08/2023] Open
Abstract
AIM The aim of this study was to evaluate the effects of the sphingosine analogue, FTY720 (Fingolimod), on the outcomes of myocardial ischaemia/reperfusion (I/R) injury. METHODS Two concentrations of FTY720 (1 or 2.5 µM were administered either prior to (PreFTY), or following (PostFTY) 20 minutes' global (GI) or 35 minutes' regional ischaemia (RI) in the isolated, perfused, working rat heart. Functional recovery during reperfusion was assessed following both models of ischaemia, while infarct size (IFS) was determined following RI. RESULTS FTY720 at 1 µM exerted no effect on functional recovery, while 2.5 µM significantly impaired aortic output (AO) recovery when administered prior to GI (% recovery: control: 33.88 ± 6.12% vs PreFTY: 0%, n = 6-10; p < 0.001), as well as before and after RI ( % recovery: control: 27.86 ± 13.22% vs PreFTY: 0.62% ; p < 0.05; and PostFTY: 2.08%; p = 0.0585, n = 6). FTY720 at 1 µM administered during reperfusion reduced IFS (% of area at risk (AAR): control: 39.89 ± 3.93% vs PostFTY: 26.56 ± 4.32%, n = 6-8; p < 0.05), while 2.5 µM FTY720 reduced IFS irrespective of the time of administration ( % of AAR: control: 39.89 ± 3.93% vs PreFTY: 29.97 ± 1.03% ; and PostFTY: 30.45 ± 2.16%, n = 6; p < 0.05). CONCLUSION FTY720 exerted divergent outcomes on function and tissue survival depending on the concentration administered, as well as the timing of administration.
Collapse
Affiliation(s)
- Derick van Vuuren
- Division of Medical Physiology, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Stellenbosch University, Tygerberg, South Africa.
| | - Erna Marais
- Division of Medical Physiology, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Stellenbosch University, Tygerberg, South Africa
| | - Sonia Genade
- Division of Medical Physiology, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Stellenbosch University, Tygerberg, South Africa
| | - Amanda Lochner
- Division of Medical Physiology, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Stellenbosch University, Tygerberg, South Africa
| |
Collapse
|
39
|
Abstract
In animal models platelet P2Y12 receptor antagonists put the heart into a protected state, not as a result of suppressed thrombosis but rather through protective signaling, similar to that for ischemic postconditioning. While both ischemic postconditioning and the P2Y12 blocker cangrelor protect blood-perfused hearts, only the former protects buffer-perfused hearts indicating that the blocker requires a blood-borne constituent or factor to protect. We used an anti-platelet antibody to make thrombocytopenic rats to test if that factor resides within the platelet. Infarct size was measured in open-chest rats subjected to 30-min ischemia/2-h reperfusion. Infarct size was not different in thrombocytopenic rats showing that preventing aggregation alone is not protective. While ischemic preconditioning could reduce infarct size in thrombocytopenic rats, the P2Y12 inhibitor cangrelor could not, indicating that it protects by interacting with some factor in the platelet. Ischemic preconditioning is known to require phosphorylation of sphingosine. In rats treated with dimethylsphingosine to block sphingosine kinase, cangrelor was no longer protective. Thus cangrelor's protective mechanism appears to also involve sphingosine kinase revealing yet another similarity to conditioning's mechanism.
Collapse
|
40
|
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: 39] [Impact Index Per Article: 4.9] [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
|
41
|
Hausenloy DJ, Barrabes JA, Bøtker HE, Davidson SM, Di Lisa F, Downey J, Engstrom T, Ferdinandy P, Carbrera-Fuentes HA, Heusch G, Ibanez B, Iliodromitis EK, Inserte J, Jennings R, Kalia N, Kharbanda R, Lecour S, Marber M, Miura T, Ovize M, Perez-Pinzon MA, Piper HM, Przyklenk K, Schmidt MR, Redington A, Ruiz-Meana M, Vilahur G, Vinten-Johansen J, Yellon DM, Garcia-Dorado D. Ischaemic conditioning and targeting reperfusion injury: a 30 year voyage of discovery. Basic Res Cardiol 2016; 111:70. [PMID: 27766474 PMCID: PMC5073120 DOI: 10.1007/s00395-016-0588-8] [Citation(s) in RCA: 244] [Impact Index Per Article: 27.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2016] [Accepted: 10/11/2016] [Indexed: 01/12/2023]
Abstract
To commemorate the auspicious occasion of the 30th anniversary of IPC, leading pioneers in the field of cardioprotection gathered in Barcelona in May 2016 to review and discuss the history of IPC, its evolution to IPost and RIC, myocardial reperfusion injury as a therapeutic target, and future targets and strategies for cardioprotection. This article provides an overview of the major topics discussed at this special meeting and underscores the huge importance and impact, the discovery of IPC has made in the field of cardiovascular research.
Collapse
Affiliation(s)
- Derek J Hausenloy
- The Hatter Cardiovascular Institute, University College London, London, UK. .,The National Institute of Health Research University College London Hospitals Biomedical Research Centre, London, UK. .,Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore, 8 College Road, Singapore, 169857, Singapore. .,National Heart Research Institute Singapore, National Heart Centre Singapore, Singapore, Singapore.
| | - Jose A Barrabes
- Department of Cardiology, Vall d'Hebron University Hospital and Research Institute, Universitat Autònoma, Barcelona, Spain
| | - Hans Erik Bøtker
- Department of Cardiology, Aarhus University Hospital Skejby, 8200, Aarhus N, Denmark
| | - Sean M Davidson
- The Hatter Cardiovascular Institute, University College London, London, UK
| | - Fabio Di Lisa
- Department of Biomedical Sciences and CNR Institute of Neurosciences, University of Padova, Padua, Italy
| | - James Downey
- Department of Physiology and Cell Biology, College of Medicine, University of South Alabama, Mobile, AL, USA
| | - Thomas Engstrom
- Department of Cardiology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Péter Ferdinandy
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary.,Pharmahungary Group, Szeged, Hungary
| | - Hector A Carbrera-Fuentes
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore, 8 College Road, Singapore, 169857, Singapore.,National Heart Research Institute Singapore, National Heart Centre Singapore, Singapore, Singapore.,Institute for Biochemistry, Medical Faculty Justus-Liebig-University, Giessen, Germany.,Department of Microbiology, Kazan Federal University, Kazan, Russian Federation
| | - Gerd Heusch
- Institute for Pathophysiology, West-German Heart and Vascular Center, University of Essen Medical School, Essen, Germany
| | - Borja Ibanez
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain.,IIS-Fundación Jiménez Díaz Hospital, Madrid, Spain
| | - Efstathios K Iliodromitis
- 2nd University Department of Cardiology, National and Kapodistrian University of Athens, Athens, Greece
| | - Javier Inserte
- Department of Cardiology, Vall d'Hebron University Hospital and Research Institute, Universitat Autònoma, Barcelona, Spain
| | | | - Neena Kalia
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, UK
| | - Rajesh Kharbanda
- Oxford Heart Centre, The John Radcliffe Hospital, Oxford University Hospitals, Oxford, UK
| | - Sandrine Lecour
- Department of Medicine, Hatter Institute for Cardiovascular Research in Africa and South African Medical Research Council Inter-University Cape Heart Group, Faculty of Health Sciences, University of Cape Town, Chris Barnard Building, Anzio Road, Observatory, Cape Town, Western Cape, 7925, South Africa
| | - Michael Marber
- King's College London BHF Centre, The Rayne Institute, St. Thomas' Hospital, London, UK
| | - Tetsuji Miura
- Department of Cardiovascular, Renal, and Metabolic Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Michel Ovize
- Explorations Fonctionnelles Cardiovasculaires, Hôpital Louis Pradel, Lyon, France.,UMR 1060 (CarMeN), Université Claude Bernard, Lyon 1, France
| | - Miguel A Perez-Pinzon
- Cerebral Vascular Disease Research Laboratories, University of Miami Miller School of Medicine, Miami, FL, 33136, USA.,Neuroscience Program, University of Miami Miller School of Medicine, Miami, FL, 33136, USA.,Department of Neurology, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
| | - Hans Michael Piper
- Carl von Ossietzky Universität Oldenburg, Ökologiezentrum, Raum 2-116, Uhlhornsweg 99 b, 26129, Oldenburg, Germany
| | - Karin Przyklenk
- Department of Physiology and Emergency Medicine, Cardiovascular Research Institute, Wayne State University, Detroit, MI, USA
| | - Michael Rahbek Schmidt
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore, 8 College Road, Singapore, 169857, Singapore
| | - Andrew Redington
- Division of Cardiology, Department of Pediatrics, Heart Institute, Cincinnati College of Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Marisol Ruiz-Meana
- Department of Cardiology, Vall d'Hebron University Hospital and Research Institute, Universitat Autònoma, Barcelona, Spain
| | - Gemma Vilahur
- Cardiovascular Research Center, CSIC-ICCC, IIB-Hospital Sant Pau, c/Sant Antoni Maria Claret 167, 08025, Barcelona, Spain
| | - Jakob Vinten-Johansen
- Division of Cardiothoracic Surgery, Department of Surgery, Emory University, Atlanta, USA
| | - Derek M Yellon
- The Hatter Cardiovascular Institute, University College London, London, UK.,The National Institute of Health Research University College London Hospitals Biomedical Research Centre, London, UK
| | - David Garcia-Dorado
- Department of Cardiology, Vall d'Hebron University Hospital and Research Institute, Universitat Autònoma, Barcelona, Spain.
| |
Collapse
|
42
|
Role of JAK-STAT pathway in reducing cardiomyocytes hypoxia/reoxygenation injury induced by S1P postconditioning. Eur J Pharmacol 2016; 784:129-36. [PMID: 27215146 DOI: 10.1016/j.ejphar.2016.05.024] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Revised: 05/19/2016] [Accepted: 05/19/2016] [Indexed: 01/01/2023]
Abstract
This experiment was designed to explore the protection of sphingosine1-phosphate (S1P) postconditioning on rat myocardial cells injured by hypoxia/reoxygenation acting via the Janus kinase-signal transducer and activator of transcription (JAK-STAT) signal pathway. The data showed that S1P could significantly increase cell viability, lower the rate of apoptosis, decrease the content of lactate dehydrogenase (LDH) and caspase3 activity in the culture medium, increase the activity of total superoxide dismutase (T-SOD) and manganese superoxide dismutase (Mn-SOD), reduce the loss of mitochondrial membrane potential and the fluorescence intensity of intracellular calcium, as well as increase the phosphorylation of JAK2 and STAT3 in comparison with the H/R group. When the JAK inhibitor AG490 or the STAT inhibitor stattic were added, the effects of S1P were inhibited. Our date shows that S1P protects H9c2 cells from hypoxia/reoxygenation injury and that the protection by S1P was inhibited by AG490 and stattic. Therefore S1P protects H9c2 cells against hypoxia/reoxygenation injury via the JAK-STAT pathway.
Collapse
|
43
|
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: 1.4] [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
|
44
|
Understanding STAT3 signaling in cardiac ischemia. Basic Res Cardiol 2016; 111:27. [PMID: 27017613 DOI: 10.1007/s00395-016-0543-8] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2015] [Revised: 01/25/2016] [Accepted: 02/17/2016] [Indexed: 10/22/2022]
Abstract
Cardiovascular disease is the leading cause of death worldwide. It remains one of the greatest challenges to global health and will continue to dominate mortality trends in the future. Acute myocardial infarction results in 7.4 million deaths globally per annum. Current management strategies are centered on restoration of coronary blood flow via percutaneous coronary intervention, coronary artery bypass grafting and administration of anti-platelet agents. Such myocardial reperfusion accounts for 40-50 % of the final infarct size in most cases. Signaling transducer and activator of transcription 3 (STAT3) has been shown to have cardioprotective effects via canonical and non-canonical activation and modulation of mitochondrial and transcriptional responses. A significant body of in vitro and in vivo evidence suggests that activation of the STAT3 signal transduction pathway results in a cardio protective response to ischemia and attempts have been made to modulate this with therapeutic effect. Not only is STAT3 important for cardiomyocyte function, but it also modulates the cardiac microenvironment and communicates with cardiac fibroblasts. To this end, we here review the current evidence supporting the manipulation of STAT3 for therapeutic benefit in cardiac ischemia and identify areas for future research.
Collapse
|
45
|
Brinck JW, Thomas A, Lauer E, Jornayvaz FR, Brulhart-Meynet MC, Prost JC, Pataky Z, Löfgren P, Hoffstedt J, Eriksson M, Pramfalk C, Morel S, Kwak BR, van Eck M, James RW, Frias MA. Diabetes Mellitus Is Associated With Reduced High-Density Lipoprotein Sphingosine-1-Phosphate Content and Impaired High-Density Lipoprotein Cardiac Cell Protection. Arterioscler Thromb Vasc Biol 2016; 36:817-24. [PMID: 26966278 DOI: 10.1161/atvbaha.115.307049] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2015] [Accepted: 02/29/2016] [Indexed: 11/16/2022]
Abstract
OBJECTIVE The dyslipidemia of type 2 diabetes mellitus has multiple etiologies and impairs lipoprotein functionality, thereby increasing risk for cardiovascular disease. High-density lipoproteins (HDLs) have several beneficial effects, notably protecting the heart from myocardial ischemia. We hypothesized that glycation of HDL could compromise this cardioprotective effect. APPROACH AND RESULTS We used in vitro (cardiomyocytes) and ex vivo (whole heart) models subjected to oxidative stress together with HDL isolated from diabetic patients and nondiabetic HDL glycated in vitro (methylglyoxal). Diabetic and in vitro glycated HDL were less effective (P<0.05) than control HDL in protecting from oxidative stress. Protection was significantly, inversely correlated with the degree of in vitro glycation (P<0.001) and the levels of hemoglobin A1c in diabetic patients (P<0.007). The ability to activate protective, intracellular survival pathways involving Akt, Stat3, and Erk1/2 was significantly reduced (P<0.05) using glycated HDL. Glycation reduced the sphingosine-1-phosphate (S1P) content of HDL, whereas the S1P concentrations of diabetic HDL were inversely correlated with hemoglobin A1c (P<0.005). The S1P contents of in vitro glycated and diabetic HDL were significantly, positively correlated (both <0.01) with cardiomyocyte survival during oxidative stress. Adding S1P to diabetic HDL increased its S1P content and restored its cardioprotective function. CONCLUSIONS Our data demonstrate that glycation can reduce the S1P content of HDL, leading to increased cardiomyocyte cell death because of less effective activation of intracellular survival pathways. It has important implications for the functionality of HDL in diabetes mellitus because HDL-S1P has several beneficial effects on the vasculature.
Collapse
Affiliation(s)
- Jonas W Brinck
- From the Department of Internal Medicine, Division of Endocrinology, Diabetology, Hypertension and Nutrition, Medical Faculty, Geneva University, Geneva, Switzerland (J.W.B., F.R.J., M.-C.B.-M., R.W.J., M.A.F.); University Centre of Legal Medicine, Unit of Toxicology, Lausanne-Geneva, Switzerland (A.T., E.L., J.-C.P.); Faculty of Biology and Medicine, Lausanne University Hospital, Lausanne University, Lausanne, Switzerland (A.T.); Department of Community Medicine, Service of Therapeutic Education for Chronic Diseases, WHO Collaborating Centre, University Hospitals of Geneva and University of Geneva, Geneva, Switzerland (Z.P.); Department of Medicine, Karolinska Institute, Stockholm, Sweden (J.W.B., P.L., J.H., M.E.); Molecular Nutrition Unit, Department of Biosciences and Nutrition, Karolinska Institute, Stockholm, Sweden (C.P.); Division of Clinical Chemistry, Department of Laboratory Medicine, Karolinska Institute, Stockholm, Sweden (C.P.); Department of Pathology and Immunology, Medical Faculty, Geneva University, Geneva, Switzerland (S.M., B.R.K.); and Leiden Academic Centre for Drug Research, Division of Biopharmaceutics, Cluster of BioTherapeutics, Leiden University, Leiden, The Netherlands (M.v.E.).
| | - Aurélien Thomas
- From the Department of Internal Medicine, Division of Endocrinology, Diabetology, Hypertension and Nutrition, Medical Faculty, Geneva University, Geneva, Switzerland (J.W.B., F.R.J., M.-C.B.-M., R.W.J., M.A.F.); University Centre of Legal Medicine, Unit of Toxicology, Lausanne-Geneva, Switzerland (A.T., E.L., J.-C.P.); Faculty of Biology and Medicine, Lausanne University Hospital, Lausanne University, Lausanne, Switzerland (A.T.); Department of Community Medicine, Service of Therapeutic Education for Chronic Diseases, WHO Collaborating Centre, University Hospitals of Geneva and University of Geneva, Geneva, Switzerland (Z.P.); Department of Medicine, Karolinska Institute, Stockholm, Sweden (J.W.B., P.L., J.H., M.E.); Molecular Nutrition Unit, Department of Biosciences and Nutrition, Karolinska Institute, Stockholm, Sweden (C.P.); Division of Clinical Chemistry, Department of Laboratory Medicine, Karolinska Institute, Stockholm, Sweden (C.P.); Department of Pathology and Immunology, Medical Faculty, Geneva University, Geneva, Switzerland (S.M., B.R.K.); and Leiden Academic Centre for Drug Research, Division of Biopharmaceutics, Cluster of BioTherapeutics, Leiden University, Leiden, The Netherlands (M.v.E.)
| | - Estelle Lauer
- From the Department of Internal Medicine, Division of Endocrinology, Diabetology, Hypertension and Nutrition, Medical Faculty, Geneva University, Geneva, Switzerland (J.W.B., F.R.J., M.-C.B.-M., R.W.J., M.A.F.); University Centre of Legal Medicine, Unit of Toxicology, Lausanne-Geneva, Switzerland (A.T., E.L., J.-C.P.); Faculty of Biology and Medicine, Lausanne University Hospital, Lausanne University, Lausanne, Switzerland (A.T.); Department of Community Medicine, Service of Therapeutic Education for Chronic Diseases, WHO Collaborating Centre, University Hospitals of Geneva and University of Geneva, Geneva, Switzerland (Z.P.); Department of Medicine, Karolinska Institute, Stockholm, Sweden (J.W.B., P.L., J.H., M.E.); Molecular Nutrition Unit, Department of Biosciences and Nutrition, Karolinska Institute, Stockholm, Sweden (C.P.); Division of Clinical Chemistry, Department of Laboratory Medicine, Karolinska Institute, Stockholm, Sweden (C.P.); Department of Pathology and Immunology, Medical Faculty, Geneva University, Geneva, Switzerland (S.M., B.R.K.); and Leiden Academic Centre for Drug Research, Division of Biopharmaceutics, Cluster of BioTherapeutics, Leiden University, Leiden, The Netherlands (M.v.E.)
| | - François R Jornayvaz
- From the Department of Internal Medicine, Division of Endocrinology, Diabetology, Hypertension and Nutrition, Medical Faculty, Geneva University, Geneva, Switzerland (J.W.B., F.R.J., M.-C.B.-M., R.W.J., M.A.F.); University Centre of Legal Medicine, Unit of Toxicology, Lausanne-Geneva, Switzerland (A.T., E.L., J.-C.P.); Faculty of Biology and Medicine, Lausanne University Hospital, Lausanne University, Lausanne, Switzerland (A.T.); Department of Community Medicine, Service of Therapeutic Education for Chronic Diseases, WHO Collaborating Centre, University Hospitals of Geneva and University of Geneva, Geneva, Switzerland (Z.P.); Department of Medicine, Karolinska Institute, Stockholm, Sweden (J.W.B., P.L., J.H., M.E.); Molecular Nutrition Unit, Department of Biosciences and Nutrition, Karolinska Institute, Stockholm, Sweden (C.P.); Division of Clinical Chemistry, Department of Laboratory Medicine, Karolinska Institute, Stockholm, Sweden (C.P.); Department of Pathology and Immunology, Medical Faculty, Geneva University, Geneva, Switzerland (S.M., B.R.K.); and Leiden Academic Centre for Drug Research, Division of Biopharmaceutics, Cluster of BioTherapeutics, Leiden University, Leiden, The Netherlands (M.v.E.)
| | - Marie-Claude Brulhart-Meynet
- From the Department of Internal Medicine, Division of Endocrinology, Diabetology, Hypertension and Nutrition, Medical Faculty, Geneva University, Geneva, Switzerland (J.W.B., F.R.J., M.-C.B.-M., R.W.J., M.A.F.); University Centre of Legal Medicine, Unit of Toxicology, Lausanne-Geneva, Switzerland (A.T., E.L., J.-C.P.); Faculty of Biology and Medicine, Lausanne University Hospital, Lausanne University, Lausanne, Switzerland (A.T.); Department of Community Medicine, Service of Therapeutic Education for Chronic Diseases, WHO Collaborating Centre, University Hospitals of Geneva and University of Geneva, Geneva, Switzerland (Z.P.); Department of Medicine, Karolinska Institute, Stockholm, Sweden (J.W.B., P.L., J.H., M.E.); Molecular Nutrition Unit, Department of Biosciences and Nutrition, Karolinska Institute, Stockholm, Sweden (C.P.); Division of Clinical Chemistry, Department of Laboratory Medicine, Karolinska Institute, Stockholm, Sweden (C.P.); Department of Pathology and Immunology, Medical Faculty, Geneva University, Geneva, Switzerland (S.M., B.R.K.); and Leiden Academic Centre for Drug Research, Division of Biopharmaceutics, Cluster of BioTherapeutics, Leiden University, Leiden, The Netherlands (M.v.E.)
| | - Jean-Christophe Prost
- From the Department of Internal Medicine, Division of Endocrinology, Diabetology, Hypertension and Nutrition, Medical Faculty, Geneva University, Geneva, Switzerland (J.W.B., F.R.J., M.-C.B.-M., R.W.J., M.A.F.); University Centre of Legal Medicine, Unit of Toxicology, Lausanne-Geneva, Switzerland (A.T., E.L., J.-C.P.); Faculty of Biology and Medicine, Lausanne University Hospital, Lausanne University, Lausanne, Switzerland (A.T.); Department of Community Medicine, Service of Therapeutic Education for Chronic Diseases, WHO Collaborating Centre, University Hospitals of Geneva and University of Geneva, Geneva, Switzerland (Z.P.); Department of Medicine, Karolinska Institute, Stockholm, Sweden (J.W.B., P.L., J.H., M.E.); Molecular Nutrition Unit, Department of Biosciences and Nutrition, Karolinska Institute, Stockholm, Sweden (C.P.); Division of Clinical Chemistry, Department of Laboratory Medicine, Karolinska Institute, Stockholm, Sweden (C.P.); Department of Pathology and Immunology, Medical Faculty, Geneva University, Geneva, Switzerland (S.M., B.R.K.); and Leiden Academic Centre for Drug Research, Division of Biopharmaceutics, Cluster of BioTherapeutics, Leiden University, Leiden, The Netherlands (M.v.E.)
| | - Zoltan Pataky
- From the Department of Internal Medicine, Division of Endocrinology, Diabetology, Hypertension and Nutrition, Medical Faculty, Geneva University, Geneva, Switzerland (J.W.B., F.R.J., M.-C.B.-M., R.W.J., M.A.F.); University Centre of Legal Medicine, Unit of Toxicology, Lausanne-Geneva, Switzerland (A.T., E.L., J.-C.P.); Faculty of Biology and Medicine, Lausanne University Hospital, Lausanne University, Lausanne, Switzerland (A.T.); Department of Community Medicine, Service of Therapeutic Education for Chronic Diseases, WHO Collaborating Centre, University Hospitals of Geneva and University of Geneva, Geneva, Switzerland (Z.P.); Department of Medicine, Karolinska Institute, Stockholm, Sweden (J.W.B., P.L., J.H., M.E.); Molecular Nutrition Unit, Department of Biosciences and Nutrition, Karolinska Institute, Stockholm, Sweden (C.P.); Division of Clinical Chemistry, Department of Laboratory Medicine, Karolinska Institute, Stockholm, Sweden (C.P.); Department of Pathology and Immunology, Medical Faculty, Geneva University, Geneva, Switzerland (S.M., B.R.K.); and Leiden Academic Centre for Drug Research, Division of Biopharmaceutics, Cluster of BioTherapeutics, Leiden University, Leiden, The Netherlands (M.v.E.)
| | - Patrik Löfgren
- From the Department of Internal Medicine, Division of Endocrinology, Diabetology, Hypertension and Nutrition, Medical Faculty, Geneva University, Geneva, Switzerland (J.W.B., F.R.J., M.-C.B.-M., R.W.J., M.A.F.); University Centre of Legal Medicine, Unit of Toxicology, Lausanne-Geneva, Switzerland (A.T., E.L., J.-C.P.); Faculty of Biology and Medicine, Lausanne University Hospital, Lausanne University, Lausanne, Switzerland (A.T.); Department of Community Medicine, Service of Therapeutic Education for Chronic Diseases, WHO Collaborating Centre, University Hospitals of Geneva and University of Geneva, Geneva, Switzerland (Z.P.); Department of Medicine, Karolinska Institute, Stockholm, Sweden (J.W.B., P.L., J.H., M.E.); Molecular Nutrition Unit, Department of Biosciences and Nutrition, Karolinska Institute, Stockholm, Sweden (C.P.); Division of Clinical Chemistry, Department of Laboratory Medicine, Karolinska Institute, Stockholm, Sweden (C.P.); Department of Pathology and Immunology, Medical Faculty, Geneva University, Geneva, Switzerland (S.M., B.R.K.); and Leiden Academic Centre for Drug Research, Division of Biopharmaceutics, Cluster of BioTherapeutics, Leiden University, Leiden, The Netherlands (M.v.E.)
| | - Johan Hoffstedt
- From the Department of Internal Medicine, Division of Endocrinology, Diabetology, Hypertension and Nutrition, Medical Faculty, Geneva University, Geneva, Switzerland (J.W.B., F.R.J., M.-C.B.-M., R.W.J., M.A.F.); University Centre of Legal Medicine, Unit of Toxicology, Lausanne-Geneva, Switzerland (A.T., E.L., J.-C.P.); Faculty of Biology and Medicine, Lausanne University Hospital, Lausanne University, Lausanne, Switzerland (A.T.); Department of Community Medicine, Service of Therapeutic Education for Chronic Diseases, WHO Collaborating Centre, University Hospitals of Geneva and University of Geneva, Geneva, Switzerland (Z.P.); Department of Medicine, Karolinska Institute, Stockholm, Sweden (J.W.B., P.L., J.H., M.E.); Molecular Nutrition Unit, Department of Biosciences and Nutrition, Karolinska Institute, Stockholm, Sweden (C.P.); Division of Clinical Chemistry, Department of Laboratory Medicine, Karolinska Institute, Stockholm, Sweden (C.P.); Department of Pathology and Immunology, Medical Faculty, Geneva University, Geneva, Switzerland (S.M., B.R.K.); and Leiden Academic Centre for Drug Research, Division of Biopharmaceutics, Cluster of BioTherapeutics, Leiden University, Leiden, The Netherlands (M.v.E.)
| | - Mats Eriksson
- From the Department of Internal Medicine, Division of Endocrinology, Diabetology, Hypertension and Nutrition, Medical Faculty, Geneva University, Geneva, Switzerland (J.W.B., F.R.J., M.-C.B.-M., R.W.J., M.A.F.); University Centre of Legal Medicine, Unit of Toxicology, Lausanne-Geneva, Switzerland (A.T., E.L., J.-C.P.); Faculty of Biology and Medicine, Lausanne University Hospital, Lausanne University, Lausanne, Switzerland (A.T.); Department of Community Medicine, Service of Therapeutic Education for Chronic Diseases, WHO Collaborating Centre, University Hospitals of Geneva and University of Geneva, Geneva, Switzerland (Z.P.); Department of Medicine, Karolinska Institute, Stockholm, Sweden (J.W.B., P.L., J.H., M.E.); Molecular Nutrition Unit, Department of Biosciences and Nutrition, Karolinska Institute, Stockholm, Sweden (C.P.); Division of Clinical Chemistry, Department of Laboratory Medicine, Karolinska Institute, Stockholm, Sweden (C.P.); Department of Pathology and Immunology, Medical Faculty, Geneva University, Geneva, Switzerland (S.M., B.R.K.); and Leiden Academic Centre for Drug Research, Division of Biopharmaceutics, Cluster of BioTherapeutics, Leiden University, Leiden, The Netherlands (M.v.E.)
| | - Camilla Pramfalk
- From the Department of Internal Medicine, Division of Endocrinology, Diabetology, Hypertension and Nutrition, Medical Faculty, Geneva University, Geneva, Switzerland (J.W.B., F.R.J., M.-C.B.-M., R.W.J., M.A.F.); University Centre of Legal Medicine, Unit of Toxicology, Lausanne-Geneva, Switzerland (A.T., E.L., J.-C.P.); Faculty of Biology and Medicine, Lausanne University Hospital, Lausanne University, Lausanne, Switzerland (A.T.); Department of Community Medicine, Service of Therapeutic Education for Chronic Diseases, WHO Collaborating Centre, University Hospitals of Geneva and University of Geneva, Geneva, Switzerland (Z.P.); Department of Medicine, Karolinska Institute, Stockholm, Sweden (J.W.B., P.L., J.H., M.E.); Molecular Nutrition Unit, Department of Biosciences and Nutrition, Karolinska Institute, Stockholm, Sweden (C.P.); Division of Clinical Chemistry, Department of Laboratory Medicine, Karolinska Institute, Stockholm, Sweden (C.P.); Department of Pathology and Immunology, Medical Faculty, Geneva University, Geneva, Switzerland (S.M., B.R.K.); and Leiden Academic Centre for Drug Research, Division of Biopharmaceutics, Cluster of BioTherapeutics, Leiden University, Leiden, The Netherlands (M.v.E.)
| | - Sandrine Morel
- From the Department of Internal Medicine, Division of Endocrinology, Diabetology, Hypertension and Nutrition, Medical Faculty, Geneva University, Geneva, Switzerland (J.W.B., F.R.J., M.-C.B.-M., R.W.J., M.A.F.); University Centre of Legal Medicine, Unit of Toxicology, Lausanne-Geneva, Switzerland (A.T., E.L., J.-C.P.); Faculty of Biology and Medicine, Lausanne University Hospital, Lausanne University, Lausanne, Switzerland (A.T.); Department of Community Medicine, Service of Therapeutic Education for Chronic Diseases, WHO Collaborating Centre, University Hospitals of Geneva and University of Geneva, Geneva, Switzerland (Z.P.); Department of Medicine, Karolinska Institute, Stockholm, Sweden (J.W.B., P.L., J.H., M.E.); Molecular Nutrition Unit, Department of Biosciences and Nutrition, Karolinska Institute, Stockholm, Sweden (C.P.); Division of Clinical Chemistry, Department of Laboratory Medicine, Karolinska Institute, Stockholm, Sweden (C.P.); Department of Pathology and Immunology, Medical Faculty, Geneva University, Geneva, Switzerland (S.M., B.R.K.); and Leiden Academic Centre for Drug Research, Division of Biopharmaceutics, Cluster of BioTherapeutics, Leiden University, Leiden, The Netherlands (M.v.E.)
| | - Brenda R Kwak
- From the Department of Internal Medicine, Division of Endocrinology, Diabetology, Hypertension and Nutrition, Medical Faculty, Geneva University, Geneva, Switzerland (J.W.B., F.R.J., M.-C.B.-M., R.W.J., M.A.F.); University Centre of Legal Medicine, Unit of Toxicology, Lausanne-Geneva, Switzerland (A.T., E.L., J.-C.P.); Faculty of Biology and Medicine, Lausanne University Hospital, Lausanne University, Lausanne, Switzerland (A.T.); Department of Community Medicine, Service of Therapeutic Education for Chronic Diseases, WHO Collaborating Centre, University Hospitals of Geneva and University of Geneva, Geneva, Switzerland (Z.P.); Department of Medicine, Karolinska Institute, Stockholm, Sweden (J.W.B., P.L., J.H., M.E.); Molecular Nutrition Unit, Department of Biosciences and Nutrition, Karolinska Institute, Stockholm, Sweden (C.P.); Division of Clinical Chemistry, Department of Laboratory Medicine, Karolinska Institute, Stockholm, Sweden (C.P.); Department of Pathology and Immunology, Medical Faculty, Geneva University, Geneva, Switzerland (S.M., B.R.K.); and Leiden Academic Centre for Drug Research, Division of Biopharmaceutics, Cluster of BioTherapeutics, Leiden University, Leiden, The Netherlands (M.v.E.)
| | - Miranda van Eck
- From the Department of Internal Medicine, Division of Endocrinology, Diabetology, Hypertension and Nutrition, Medical Faculty, Geneva University, Geneva, Switzerland (J.W.B., F.R.J., M.-C.B.-M., R.W.J., M.A.F.); University Centre of Legal Medicine, Unit of Toxicology, Lausanne-Geneva, Switzerland (A.T., E.L., J.-C.P.); Faculty of Biology and Medicine, Lausanne University Hospital, Lausanne University, Lausanne, Switzerland (A.T.); Department of Community Medicine, Service of Therapeutic Education for Chronic Diseases, WHO Collaborating Centre, University Hospitals of Geneva and University of Geneva, Geneva, Switzerland (Z.P.); Department of Medicine, Karolinska Institute, Stockholm, Sweden (J.W.B., P.L., J.H., M.E.); Molecular Nutrition Unit, Department of Biosciences and Nutrition, Karolinska Institute, Stockholm, Sweden (C.P.); Division of Clinical Chemistry, Department of Laboratory Medicine, Karolinska Institute, Stockholm, Sweden (C.P.); Department of Pathology and Immunology, Medical Faculty, Geneva University, Geneva, Switzerland (S.M., B.R.K.); and Leiden Academic Centre for Drug Research, Division of Biopharmaceutics, Cluster of BioTherapeutics, Leiden University, Leiden, The Netherlands (M.v.E.)
| | - Richard W James
- From the Department of Internal Medicine, Division of Endocrinology, Diabetology, Hypertension and Nutrition, Medical Faculty, Geneva University, Geneva, Switzerland (J.W.B., F.R.J., M.-C.B.-M., R.W.J., M.A.F.); University Centre of Legal Medicine, Unit of Toxicology, Lausanne-Geneva, Switzerland (A.T., E.L., J.-C.P.); Faculty of Biology and Medicine, Lausanne University Hospital, Lausanne University, Lausanne, Switzerland (A.T.); Department of Community Medicine, Service of Therapeutic Education for Chronic Diseases, WHO Collaborating Centre, University Hospitals of Geneva and University of Geneva, Geneva, Switzerland (Z.P.); Department of Medicine, Karolinska Institute, Stockholm, Sweden (J.W.B., P.L., J.H., M.E.); Molecular Nutrition Unit, Department of Biosciences and Nutrition, Karolinska Institute, Stockholm, Sweden (C.P.); Division of Clinical Chemistry, Department of Laboratory Medicine, Karolinska Institute, Stockholm, Sweden (C.P.); Department of Pathology and Immunology, Medical Faculty, Geneva University, Geneva, Switzerland (S.M., B.R.K.); and Leiden Academic Centre for Drug Research, Division of Biopharmaceutics, Cluster of BioTherapeutics, Leiden University, Leiden, The Netherlands (M.v.E.)
| | - Miguel A Frias
- From the Department of Internal Medicine, Division of Endocrinology, Diabetology, Hypertension and Nutrition, Medical Faculty, Geneva University, Geneva, Switzerland (J.W.B., F.R.J., M.-C.B.-M., R.W.J., M.A.F.); University Centre of Legal Medicine, Unit of Toxicology, Lausanne-Geneva, Switzerland (A.T., E.L., J.-C.P.); Faculty of Biology and Medicine, Lausanne University Hospital, Lausanne University, Lausanne, Switzerland (A.T.); Department of Community Medicine, Service of Therapeutic Education for Chronic Diseases, WHO Collaborating Centre, University Hospitals of Geneva and University of Geneva, Geneva, Switzerland (Z.P.); Department of Medicine, Karolinska Institute, Stockholm, Sweden (J.W.B., P.L., J.H., M.E.); Molecular Nutrition Unit, Department of Biosciences and Nutrition, Karolinska Institute, Stockholm, Sweden (C.P.); Division of Clinical Chemistry, Department of Laboratory Medicine, Karolinska Institute, Stockholm, Sweden (C.P.); Department of Pathology and Immunology, Medical Faculty, Geneva University, Geneva, Switzerland (S.M., B.R.K.); and Leiden Academic Centre for Drug Research, Division of Biopharmaceutics, Cluster of BioTherapeutics, Leiden University, Leiden, The Netherlands (M.v.E.)
| |
Collapse
|
46
|
Morel S, Christoffersen C, Axelsen LN, Montecucco F, Rochemont V, Frias MA, Mach F, James RW, Naus CC, Chanson M, Lampe PD, Nielsen MS, Nielsen LB, Kwak BR. Sphingosine-1-phosphate reduces ischaemia-reperfusion injury by phosphorylating the gap junction protein Connexin43. Cardiovasc Res 2016; 109:385-396. [PMID: 26762268 PMCID: PMC4752044 DOI: 10.1093/cvr/cvw004] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2015] [Accepted: 01/09/2016] [Indexed: 01/29/2023] Open
Abstract
AIM Increasing evidence points to lipoprotein composition rather than reverse cholesterol transport in the cardioprotective properties of high-density lipoproteins (HDLs). HDL binding to receptors at the surface of cardiomyocytes activates signalling pathways promoting survival, but downstream targets are largely unknown. Here, we investigate the pathways by which the sphingosine-1-phosphate (S1P) constituent of HDL limits cell death induced by cardiac ischaemia-reperfusion (I/R). METHODS AND RESULTS Apolipoprotein M (ApoM) transgenic (Apom-Tg) mice, in which plasma S1P is increased by 296%, and wild-type (WT) mice were subjected to in vivo I/R. Infarct size, neutrophil infiltration into the infarcted area, and serum Troponin I were less pronounced in Apom-Tg mice. In vitro experiments suggest that this cardioprotection depends on direct effects of S1P on cardiomyocytes, whereas leucocyte recruitment seems only indirectly affected. Importantly, short-term S1P treatment at the onset of reperfusion was sufficient to reduce I/R injury in isolated perfused hearts. Mechanistic in vitro and ex vivo studies revealed that 5 min of S1P treatment induced phosphorylation of the gap junction protein Connexin43 (Cx43) on Serine368 (S368), which was mediated by S1P2 and S1P3, but not by S1P1, receptors in cardiomyocytes. Finally, S1P-induced reduction of infarct size after ex vivo I/R was lost in hearts of mice with a truncated C-terminus of Cx43 (Cx43(K258/KO)) or in which the S368 is mutated to a non-phosphorylatable alanine (Cx43(S368A/S368A)). CONCLUSION Our study reveals an important molecular pathway by which modulating the apoM/S1P axis has a therapeutic potential in the fight against I/R injury in the heart.
Collapse
Affiliation(s)
- Sandrine Morel
- Department of Pathology and Immunology, University of Geneva, CMU, F06.2747.a, Rue Michel-Servet 1, Geneva 1211, Switzerland
| | - Christina Christoffersen
- Department of Clinical Biochemistry, Rigshospitalet, Copenhagen 2100, Denmark Department of Biomedical Sciences, Copenhagen 2200, Denmark
| | - Lene N Axelsen
- Department of Biomedical Sciences, Copenhagen 2200, Denmark The Danish National Research Foundation Centre for Cardiac Arrhythmia, Copenhagen 2200, Denmark
| | - Fabrizio Montecucco
- Department of Medical Specialties-Cardiology, University of Geneva, Geneva 1211, Switzerland
| | - Viviane Rochemont
- Department of Pathology and Immunology, University of Geneva, CMU, F06.2747.a, Rue Michel-Servet 1, Geneva 1211, Switzerland
| | - Miguel A Frias
- Department of Medical Specialties-Endocrinology, Diabetology, Hypertension and Nutrition, University of Geneva, Geneva 1211, Switzerland
| | - Francois Mach
- Department of Medical Specialties-Cardiology, University of Geneva, Geneva 1211, Switzerland
| | - Richard W James
- Department of Medical Specialties-Endocrinology, Diabetology, Hypertension and Nutrition, University of Geneva, Geneva 1211, Switzerland
| | - Christian C Naus
- Department of Cellular and Physiological Science, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Marc Chanson
- Department of Pediatrics, University of Geneva, Geneva 1211, Switzerland Department of Cell Physiology and Metabolism, University of Geneva, Geneva 1211, Switzerland
| | - Paul D Lampe
- Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Morten S Nielsen
- Department of Biomedical Sciences, Copenhagen 2200, Denmark The Danish National Research Foundation Centre for Cardiac Arrhythmia, Copenhagen 2200, Denmark
| | - Lars B Nielsen
- Department of Clinical Biochemistry, Rigshospitalet, Copenhagen 2100, Denmark Department of Biomedical Sciences, Copenhagen 2200, Denmark Department of Clinical Medicine, University of Copenhagen, Copenhagen 2100, Denmark
| | - Brenda R Kwak
- Department of Pathology and Immunology, University of Geneva, CMU, F06.2747.a, Rue Michel-Servet 1, Geneva 1211, Switzerland Department of Medical Specialties-Cardiology, University of Geneva, Geneva 1211, Switzerland
| |
Collapse
|
47
|
Hildebrandt HA, Kreienkamp V, Gent S, Kahlert P, Heusch G, Kleinbongard P. Kinetics and Signal Activation Properties of Circulating Factor(s) From Healthy Volunteers Undergoing Remote Ischemic Pre-Conditioning. ACTA ACUST UNITED AC 2016; 1:3-13. [PMID: 27642642 PMCID: PMC5012372 DOI: 10.1016/j.jacbts.2016.01.007] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Revised: 01/13/2016] [Accepted: 01/14/2016] [Indexed: 12/11/2022]
Abstract
Although remote ischemic pre-conditioning (RIPC) reduced infarct size in animal experiments and proof-of-concept clinical trials, recent phase III trials failed to confirm cardioprotection during cardiac surgery. Here, we characterized the kinetic properties of humoral factors that are released after RIPC, as well as the signal transduction pathways that were responsible for cardioprotection in an ex vivo model of global ischemia reperfusion injury. Venous blood from 20 healthy volunteers was collected at baseline and 5 min, 30 min, 1 h, 6 h, and daily from 1 to 7 days after RIPC (3 × 5/5 min upper-limb ischemia/reperfusion). Plasma-dialysates (cut-off: 12 to 14 kDa; dilution: 1:20) were infused into Langendorff-perfused mouse hearts subjected to 20/120 min global ischemia/reperfusion. Infarct size and phosphorylation of signal transducer and activator of transcription (STAT)3, STAT5, extracellular-regulated kinase 1/2 and protein kinase B were determined. In a subgroup of plasma-dialysates, an inhibitor of STAT3 (Stattic) was used in mouse hearts. Perfusion with baseline-dialysate resulted in an infarct size of 39% of ventricular mass (interquartile range: 36% to 42%). Perfusion with dialysates obtained 5 min to 6 days after RIPC significantly reduced infarct size by ∼50% and increased STAT3 phosphorylation beyond that with baseline-dialysate. Inhibition of STAT3 abrogated these effects. These results suggest that RIPC induces the release of cardioprotective, dialyzable factor(s) within 5 min, and that circulate for up to 6 days. STAT3 is activated in murine myocardium by RIPC-induced human humoral factors and is causally involved in cardioprotection. Pre-clinical and early phase clinical studies with remote ischemic preconditioning (RIPC) appeared promising; however, RIPC was not effective in phase III clinical trials. To improve the translation of RIPC into clinical practice, the kinetic properties and functional effects of humoral factors released after RIPC in humans were characterized ex vivo. Venous blood from 20 healthy volunteers was collected at baseline and 5 min, 30 min, 1 h, 6 h and daily from 1 to 7 days after RIPC. Plasma dialysates were infused into Langendorff-perfused mouse hearts subjected to 20/120 min global ischemia/reperfusion. Perfusion with dialysates obtained 5 min to 6 days after RIPC significantly reduced infarct size by ∼50% when compared to perfusion with dialysates obtained at baseline prior to RIPC, and increased STAT3 phosphorylation beyond values obtained with baseline-dialysate.
Collapse
Key Words
- AKT, protein kinase B
- ERK, extracellular-regulated kinase
- IQR, interquartile range
- LV+RV, left and right ventricular
- LVDP, left ventricular developed pressure
- RIC, remote ischemic conditioning
- RIPC, remote ischemic pre-conditioning
- SAFE, survival activating factor enhancement
- STAT, signal transducer and activator of transcription
- TTC, 2,3,5-triphenyltetrazolium chloride
- cardioprotection
- human
- humoral factor
- kinetics
- remote ischemic pre-conditioning
- signaling
Collapse
Affiliation(s)
- Heike A. Hildebrandt
- Institute for Pathophysiology, West-German Heart and Vascular Center Essen, University of Essen Medical School, Essen, Germany
- Department of Cardiology of the West-German Heart and Vascular Center Essen, University of Essen Medical School, Essen, Germany
| | - Vincent Kreienkamp
- Institute for Pathophysiology, West-German Heart and Vascular Center Essen, University of Essen Medical School, Essen, Germany
| | - Sabine Gent
- Institute for Pathophysiology, West-German Heart and Vascular Center Essen, University of Essen Medical School, Essen, Germany
| | - Philipp Kahlert
- Department of Cardiology of the West-German Heart and Vascular Center Essen, University of Essen Medical School, Essen, Germany
| | - Gerd Heusch
- Institute for Pathophysiology, West-German Heart and Vascular Center Essen, University of Essen Medical School, Essen, Germany
| | - Petra Kleinbongard
- Institute for Pathophysiology, West-German Heart and Vascular Center Essen, University of Essen Medical School, Essen, Germany
- Reprint requests and correspondence: Dr. Petra Kleinbongard, Institute for Pathophysiology, West-German Heart and Vascular Center Essen, University of Essen Medical School, Hufelandstrasse 55, 45122 Essen, NRW, Germany.
| |
Collapse
|
48
|
Lin J, Wang T, Li Y, Wang M, Li H, Irwin MG, Xia Z. N-Acetylcysteine Restores Sevoflurane Postconditioning Cardioprotection against Myocardial Ischemia-Reperfusion Injury in Diabetic Rats. J Diabetes Res 2016; 2016:9213034. [PMID: 26783539 PMCID: PMC4691468 DOI: 10.1155/2016/9213034] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2015] [Revised: 07/30/2015] [Accepted: 08/26/2015] [Indexed: 02/07/2023] Open
Abstract
The effect of sevoflurane postconditioning (sevo-postC) cardioprotection is compromised in diabetes which is associated with increased oxidative stress. We hypothesized that antioxidant N-Acetylcysteine may enhance or restore sevo-postC cardioprotection in diabetes. Control or streptozotocin-induced Type 1 diabetic rats were either untreated or treated with N-Acetylcysteine for four weeks starting at five weeks after streptozotocin injection and were subjected to myocardial ischemia-reperfusion injury (IRI), in the absence or presence of sevo-postC. Diabetes showed reduction of cardiac STAT3 activation (p-STAT3) and adiponectin with concomitantly increase of FoxO1 and CD36, which associated with reduced sevo-postC cardioprotection. N-Acetylcysteine and sevo-postC synergistically reduced the infarct size in diabetic groups. N-Acetylcysteine remarkably increased cardiac p-STAT3 which was further enhanced by sevo-postC. N-Acetylcysteine but not sevo-postC decreased myocardial FoxO1 while sevo-postC but not N-Acetylcysteine significantly increased myocardiac adiponectin in diabetic rats. It is concluded that late stage diabetic rats displayed reduction of cardiac p-STAT3, adiponectin deficiency, and increase of FoxO1 and CD36 expression, which may be responsible for the loss of myocardial responsiveness to sevo-postC cardioprotection. N-Acetylcysteine restored Sevo-postC cardioprotection in diabetes possibly through enhancing cardiac p-STAT3 and adiponectin and reducing Fox1 and CD36.
Collapse
Affiliation(s)
- Jiefu Lin
- Department of Anesthesiology, The First Affiliated Hospital of Jinan University, Guangzhou 510642, China
| | - Tingting Wang
- Department of Anesthesiology, The University of Hong Kong, Pokfulam, Hong Kong
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yalan Li
- Department of Anesthesiology, The First Affiliated Hospital of Jinan University, Guangzhou 510642, China
- *Yalan Li: and
| | - Mengxia Wang
- Department of Anesthesiology, The First Affiliated Hospital of Jinan University, Guangzhou 510642, China
| | - Haobo Li
- Department of Anesthesiology, The University of Hong Kong, Pokfulam, Hong Kong
| | - Michael G. Irwin
- Department of Anesthesiology, The University of Hong Kong, Pokfulam, Hong Kong
| | - Zhengyuan Xia
- Department of Anesthesiology, The University of Hong Kong, Pokfulam, Hong Kong
- Department of Anesthesiology, Affiliated Hospital of Guangdong Medical College, Zhanjiang 524023, China
- *Zhengyuan Xia:
| |
Collapse
|
49
|
Zouein FA, Altara R, Chen Q, Lesnefsky EJ, Kurdi M, Booz GW. Pivotal Importance of STAT3 in Protecting the Heart from Acute and Chronic Stress: New Advancement and Unresolved Issues. Front Cardiovasc Med 2015; 2:36. [PMID: 26664907 PMCID: PMC4671345 DOI: 10.3389/fcvm.2015.00036] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2015] [Accepted: 11/12/2015] [Indexed: 12/25/2022] Open
Abstract
The transcription factor, signal transducer and activator of transcription 3 (STAT3), has been implicated in protecting the heart from acute ischemic injury under both basal conditions and as a crucial component of pre- and post-conditioning protocols. A number of anti-oxidant and antiapoptotic genes are upregulated by STAT3 via canonical means involving phosphorylation on Y705 and S727, although other incompletely defined posttranslational modifications are involved. In addition, STAT3 is now known to be present in cardiac mitochondria and to exert actions that regulate the electron transport chain, reactive oxygen species production, and mitochondrial permeability transition pore opening. These non-canonical actions of STAT3 are enhanced by S727 phosphorylation. The molecular basis for the mitochondrial actions of STAT3 is poorly understood, but STAT3 is known to interact with a critical subunit of complex I and to regulate complex I function. Dysfunctional complex I has been implicated in ischemic injury, heart failure, and the aging process. Evidence also indicates that STAT3 is protective to the heart under chronic stress conditions, including hypertension, pregnancy, and advanced age. Paradoxically, the accumulation of unphosphorylated STAT3 (U-STAT3) in the nucleus has been suggested to drive pathological cardiac hypertrophy and inflammation via non-canonical gene expression, perhaps involving a distinct acetylation profile. U-STAT3 may also regulate chromatin stability. Our understanding of how the non-canonical genomic and mitochondrial actions of STAT3 in the heart are regulated and coordinated with the canonical actions of STAT3 is rudimentary. Here, we present an overview of what is currently known about the pleotropic actions of STAT3 in the heart in order to highlight controversies and unresolved issues.
Collapse
Affiliation(s)
- Fouad A Zouein
- American University of Beirut Faculty of Medicine , Beirut , Lebanon
| | - Raffaele Altara
- Department of Pharmacology and Toxicology, School of Medicine, The University of Mississippi Medical Center , Jackson, MS , USA
| | - Qun Chen
- Division of Cardiology, Department of Internal Medicine, Pauley Heart Center, Virginia Commonwealth University , Richmond, VA , USA
| | - Edward J Lesnefsky
- Division of Cardiology, Department of Internal Medicine, Pauley Heart Center, Virginia Commonwealth University , Richmond, VA , USA ; Department of Biochemistry and Molecular Biology, Virginia Commonwealth University , Richmond, VA , USA ; McGuire Department of Veterans Affairs Medical Center , Richmond, VA , USA
| | - Mazen Kurdi
- Department of Pharmacology and Toxicology, School of Medicine, The University of Mississippi Medical Center , Jackson, MS , USA ; Department of Chemistry and Biochemistry, Faculty of Sciences, Lebanese University , Hadath , Lebanon
| | - George W Booz
- Department of Pharmacology and Toxicology, School of Medicine, The University of Mississippi Medical Center , Jackson, MS , USA
| |
Collapse
|
50
|
Kelly-Laubscher RF, King JC, Hacking D, Somers S, Hastie S, Stewart T, Imamdin A, Maarman G, Pedretti S, Lecour S. Cardiac preconditioning with sphingosine-1-phosphate requires activation of signal transducer and activator of transcription-3. Cardiovasc J Afr 2015; 25:118-23. [PMID: 25000441 PMCID: PMC4120131 DOI: 10.5830/cvja-2014-016] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2013] [Accepted: 03/31/2014] [Indexed: 02/05/2023] Open
Abstract
Aims Sphingosine-1-phosphate (S1P) is a cardioprotective agent. Signal transducer and activator of transcription 3 (STAT-3) is a key mediator of many cardioprotective agents. We aimed to explore whether STAT-3 is a key mediator in S1P-induced preconditioning. Methods Langendorff-perfused hearts from Wistar rats and wild-type or cardiomyocyte-specific STAT-3 knockout mice were pre-treated with S1P (10 nmol/l), with or without the STAT-3 pathway inhibitor AG490, before an ischaemia–reperfusion insult. Triphenyltetrazolium chloride and Evans blue staining were used for the determination of infarct size. Western blot analysis was carried out on the S1P pre-treated hearts for detection of cytosolic, nuclear and mitochondrial phosphorylated and total STAT-3 proteins. Results Pre-treatment with S1P decreased the infarct size in isolated rat (5 ± 3% vs control 26 ± 8%, p < 0.01) and wild-type mouse hearts (13 ± 1% vs control 33 ± 3%, p < 0.05). This protective effect was abolished in the rat hearts pre-treated with AG490 (30 ± 10%, p = ns vs control) and in the hearts from STAT-3 knockout mice (35 ± 4% vs control 30 ± 3%, p = ns). Levels of phosphorylated STAT-3 were significantly increased in both the nuclear (p < 0.05 vs control) and mitochondrial (p < 0.05 vs control) fractions in the S1P pre-treated hearts, but remained unchanged in the cytosolic fraction (p = ns vs control). Conclusion These novel results demonstrate that pharmacological preconditioning with S1P in the isolated heart is mediated by activation of mitochondrial and nuclear STAT-3, therefore suggesting that S1P may be a novel therapeutic target to modulate mitochondrial and nuclear function in cardiovascular disease in order to protect the heart against ischaemia–reperfusion.
Collapse
Affiliation(s)
- Roisin F Kelly-Laubscher
- Hatter Institute for Cardiovascular Research in Africa, Chris Barnard Building, Medical School Campus, University of Cape Town, Cape Town, South Africa
| | - Jonathan C King
- Hatter Institute for Cardiovascular Research in Africa, Chris Barnard Building, Medical School Campus, University of Cape Town, Cape Town, South Africa
| | - Damian Hacking
- Hatter Institute for Cardiovascular Research in Africa, Chris Barnard Building, Medical School Campus, University of Cape Town, Cape Town, South Africa
| | - Sarin Somers
- Hatter Institute for Cardiovascular Research in Africa, Chris Barnard Building, Medical School Campus, University of Cape Town, Cape Town, South Africa
| | - Samantha Hastie
- Hatter Institute for Cardiovascular Research in Africa, Chris Barnard Building, Medical School Campus, University of Cape Town, Cape Town, South Africa
| | - Tessa Stewart
- Hatter Institute for Cardiovascular Research in Africa, Chris Barnard Building, Medical School Campus, University of Cape Town, Cape Town, South Africa
| | - Aqeela Imamdin
- Hatter Institute for Cardiovascular Research in Africa, Chris Barnard Building, Medical School Campus, University of Cape Town, Cape Town, South Africa
| | - Gerald Maarman
- Hatter Institute for Cardiovascular Research in Africa, Chris Barnard Building, Medical School Campus, University of Cape Town, Cape Town, South Africa
| | - Sarah Pedretti
- Hatter Institute for Cardiovascular Research in Africa, Chris Barnard Building, Medical School Campus, University of Cape Town, Cape Town, South Africa
| | - Sandrine Lecour
- Hatter Institute for Cardiovascular Research in Africa, Chris Barnard Building, Medical School Campus, University of Cape Town, Cape Town, South Africa
| |
Collapse
|