1
|
Xie YX, Yao H, Peng JF, Ni D, Liu WT, Li CQ, Yi GH. Insight into modulators of sphingosine-1-phosphate receptor and implications for cardiovascular therapeutics. J Drug Target 2024; 32:300-310. [PMID: 38269855 DOI: 10.1080/1061186x.2024.2309577] [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: 04/26/2023] [Accepted: 10/21/2023] [Indexed: 01/26/2024]
Abstract
Cardiovascular disease is the leading cause of death worldwide, and it's of great importance to understand its underlying mechanisms and find new treatments. Sphingosine 1-phosphate (S1P) is an active lipid that exerts its effects through S1P receptors on the cell surface or intracellular signal, and regulates many cellular processes such as cell growth, cell proliferation, cell migration, cell survival, and so on. S1PR modulators are a class of modulators that can interact with S1PR subtypes to activate receptors or block their activity, exerting either agonist or functional antagonist effects. Many studies have shown that S1P plays a protective role in the cardiovascular system and regulates cardiac physiological functions mainly through interaction with cell surface S1P receptors (S1PRs). Therefore, S1PR modulators may play a therapeutic role in cardiovascular diseases. Here, we review five S1PRs and their functions and the progress of S1PR modulators. In addition, we focus on the effects of S1PR modulators on atherosclerosis, myocardial infarction, myocardial ischaemia/reperfusion injury, diabetic cardiovascular diseases, and myocarditis, which may provide valuable insights into potential therapeutic strategies for cardiovascular disease.
Collapse
Affiliation(s)
- Yu-Xin Xie
- Hunan province Cooperative Innovation Center for Molecular Target New Drug Study, Institute of Pharmacy and Pharmacology, University of South China, Hengyang, Hunan, China
- Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical School, Institute of Cardiovascular Disease, University of South China, Hengyang, Hunan, China
| | - Hui Yao
- Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical School, Institute of Cardiovascular Disease, University of South China, Hengyang, Hunan, China
| | - Jin-Fu Peng
- Hunan province Cooperative Innovation Center for Molecular Target New Drug Study, Institute of Pharmacy and Pharmacology, University of South China, Hengyang, Hunan, China
- Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical School, Institute of Cardiovascular Disease, University of South China, Hengyang, Hunan, China
| | - Dan Ni
- Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical School, Institute of Cardiovascular Disease, University of South China, Hengyang, Hunan, China
| | - Wan-Ting Liu
- Hunan province Cooperative Innovation Center for Molecular Target New Drug Study, Institute of Pharmacy and Pharmacology, University of South China, Hengyang, Hunan, China
- Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical School, Institute of Cardiovascular Disease, University of South China, Hengyang, Hunan, China
| | - Chao-Quan Li
- Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical School, Institute of Cardiovascular Disease, University of South China, Hengyang, Hunan, China
| | - Guang-Hui Yi
- Hunan province Cooperative Innovation Center for Molecular Target New Drug Study, Institute of Pharmacy and Pharmacology, University of South China, Hengyang, Hunan, China
- Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical School, Institute of Cardiovascular Disease, University of South China, Hengyang, Hunan, China
| |
Collapse
|
2
|
Foran D, Antoniades C, Akoumianakis I. Emerging Roles for Sphingolipids in Cardiometabolic Disease: A Rational Therapeutic Target? Nutrients 2024; 16:3296. [PMID: 39408263 PMCID: PMC11478599 DOI: 10.3390/nu16193296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 09/19/2024] [Accepted: 09/25/2024] [Indexed: 10/20/2024] Open
Abstract
Cardiovascular disease is a leading cause of morbidity and mortality. New research elucidates increasingly complex relationships between cardiac and metabolic health, giving rise to new possible therapeutic targets. Sphingolipids are a heterogeneous class of bioactive lipids with critical roles in normal human physiology. They have also been shown to play both protective and deleterious roles in the pathogenesis of cardiovascular disease. Ceramides are implicated in dysregulating insulin signalling, vascular endothelial function, inflammation, oxidative stress, and lipoprotein aggregation, thereby promoting atherosclerosis and vascular disease. Ceramides also advance myocardial disease by enhancing pathological cardiac remodelling and cardiomyocyte death. Glucosylceramides similarly contribute to insulin resistance and vascular inflammation, thus playing a role in atherogenesis and cardiometabolic dysfunction. Sphingosing-1-phosphate, on the other hand, may ameliorate some of the pathological functions of ceramide by protecting endothelial barrier integrity and promoting cell survival. Sphingosine-1-phosphate is, however, implicated in the development of cardiac fibrosis. This review will explore the roles of sphingolipids in vascular, cardiac, and metabolic pathologies and will evaluate the therapeutic potential in targeting sphingolipids with the aim of prevention and reversal of cardiovascular disease in order to improve long-term cardiovascular outcomes.
Collapse
Affiliation(s)
| | | | - Ioannis Akoumianakis
- Cardiovascular Medicine Division, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 9DU, UK; (D.F.); (C.A.)
| |
Collapse
|
3
|
Phan F, Bourron O, Foufelle F, Le Stunff H, Hajduch E. Sphingosine-1-phosphate signalling in the heart: exploring emerging perspectives in cardiopathology. FEBS Lett 2024. [PMID: 38965662 DOI: 10.1002/1873-3468.14973] [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: 03/18/2024] [Revised: 05/23/2024] [Accepted: 06/12/2024] [Indexed: 07/06/2024]
Abstract
Cardiometabolic disorders contribute to the global burden of cardiovascular diseases. Emerging sphingolipid metabolites like sphingosine-1-phosphate (S1P) and its receptors, S1PRs, present a dynamic signalling axis significantly impacting cardiac homeostasis. S1P's intricate mechanisms extend to its transportation in the bloodstream by two specific carriers: high-density lipoprotein particles and albumin. This intricate transport system ensures the accessibility of S1P to distant target tissues, influencing several physiological processes critical for cardiovascular health. This review delves into the diverse functions of S1P and S1PRs in both physiological and pathophysiological conditions of the heart. Emphasis is placed on their diverse roles in modulating cardiac health, spanning from cardiac contractility, angiogenesis, inflammation, atherosclerosis and myocardial infarction. The intricate interplays involving S1P and its receptors are analysed concerning different cardiac cell types, shedding light on their respective roles in different heart diseases. We also review the therapeutic applications of targeting S1P/S1PRs in cardiac diseases, considering existing drugs like Fingolimod, as well as the prospects and challenges in developing novel therapies that selectively modulate S1PRs.
Collapse
Affiliation(s)
- Franck Phan
- INSERM, Centre de Recherche des Cordeliers, Sorbonne Université, Paris, France
- Diabetology Department, Assistance Publique-Hôpitaux de Paris (APHP), La Pitié-Salpêtrière-Charles Foix University Hospital, Paris, France
- Institut Hospitalo-Universitaire ICAN, Paris, France
| | - Olivier Bourron
- INSERM, Centre de Recherche des Cordeliers, Sorbonne Université, Paris, France
- Diabetology Department, Assistance Publique-Hôpitaux de Paris (APHP), La Pitié-Salpêtrière-Charles Foix University Hospital, Paris, France
- Institut Hospitalo-Universitaire ICAN, Paris, France
| | - Fabienne Foufelle
- INSERM, Centre de Recherche des Cordeliers, Sorbonne Université, Paris, France
- Institut Hospitalo-Universitaire ICAN, Paris, France
| | - Hervé Le Stunff
- Institut des Neurosciences Paris-Saclay, CNRS UMR 9197, Université Paris-Saclay, France
| | - Eric Hajduch
- INSERM, Centre de Recherche des Cordeliers, Sorbonne Université, Paris, France
- Institut Hospitalo-Universitaire ICAN, Paris, France
| |
Collapse
|
4
|
Wang Y, Kim B, Gong S, Park J, Zhu M, Wong EM, Park AY, Chernoff J, Guo F. Control of OPC proliferation and repopulation by the intellectual disability gene PAK1 under homeostatic and demyelinating conditions. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.26.591153. [PMID: 38746444 PMCID: PMC11092442 DOI: 10.1101/2024.04.26.591153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
Appropriate proliferation and repopulation of oligodendrocyte progenitor cells (OPCs) determine successful (re)myelination in homeostatic and demyelinating brains. Activating mutations in p21-activated kinase 1 (PAK1) cause intellectual disability, neurodevelopmental abnormality, and white matter anomaly in children. It remains unclear if and how PAK1 regulates oligodendroglial development. Here, we report that PAK1 controls proliferation and regeneration of OPCs. Unlike differentiating oligodendrocytes, OPCs display high PAK1 activity which maintains them in a proliferative state by modulating PDGFRa-mediated mitogenic signaling. PAK1-deficient or kinase-inhibited OPCs reduce their proliferation capacity and population expansion. Mice carrying OPC-specific PAK1 deletion or kinase inhibition are populated with fewer OPCs in the homeostatic and demyelinated CNS than control mice. Together, our findings suggest that kinase-activating PAK1 mutations stall OPCs in a progenitor state, impacting timely oligodendroglial differentiation in the CNS of affected children and that PAK1 is a potential molecular target for replenishing OPCs in demyelinating lesions.
Collapse
Affiliation(s)
- Yan Wang
- Department of Neurology, UC Davis School of Medicine; Institute for Pediatric Regenerative Medicine (IPRM), Shriners Hospitals for Children, Sacramento, CA 95817
| | - Bokyung Kim
- Department of Neurology, UC Davis School of Medicine; Institute for Pediatric Regenerative Medicine (IPRM), Shriners Hospitals for Children, Sacramento, CA 95817
| | - Shuaishuai Gong
- Department of Neurology, UC Davis School of Medicine; Institute for Pediatric Regenerative Medicine (IPRM), Shriners Hospitals for Children, Sacramento, CA 95817
| | - Joohyun Park
- Department of Neurology, UC Davis School of Medicine; Institute for Pediatric Regenerative Medicine (IPRM), Shriners Hospitals for Children, Sacramento, CA 95817
| | - Meina Zhu
- Department of Neurology, UC Davis School of Medicine; Institute for Pediatric Regenerative Medicine (IPRM), Shriners Hospitals for Children, Sacramento, CA 95817
| | - Evelyn M. Wong
- Department of Neurology, UC Davis School of Medicine; Institute for Pediatric Regenerative Medicine (IPRM), Shriners Hospitals for Children, Sacramento, CA 95817
| | - Audrey Y. Park
- Department of Neurology, UC Davis School of Medicine; Institute for Pediatric Regenerative Medicine (IPRM), Shriners Hospitals for Children, Sacramento, CA 95817
| | - Jonathan Chernoff
- Cancer Signaling and Microenvironment Program, Fox Chase Cancer Center, 333 Cottman Avenue, Philadelphia, PA 19111
| | - Fuzheng Guo
- Department of Neurology, UC Davis School of Medicine; Institute for Pediatric Regenerative Medicine (IPRM), Shriners Hospitals for Children, Sacramento, CA 95817
| |
Collapse
|
5
|
Wang L, Zhang X, Ma C, Wu N. 1-Phosphate receptor agonists: A promising therapeutic avenue for ischemia-reperfusion injury management. Int Immunopharmacol 2024; 131:111835. [PMID: 38508097 DOI: 10.1016/j.intimp.2024.111835] [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: 02/01/2024] [Accepted: 03/07/2024] [Indexed: 03/22/2024]
Abstract
Ischemia-reperfusion injury (IRI) - a complex pathological condition occurring when blood supply is abruptly restored to ischemic tissues, leading to further tissue damage - poses a significant clinical challenge. Sphingosine-1-phosphate receptors (S1PRs), a specialized set of G-protein-coupled receptors comprising five subtypes (S1PR1 to S1PR5), are prominently present in various cell membranes, including those of lymphocytes, cardiac myocytes, and endothelial cells. Increasing evidence highlights the potential of targeting S1PRs for IRI therapeutic intervention. Notably, preconditioning and postconditioning strategies involving S1PR agonists like FTY720 have demonstrated efficacy in mitigating IRI. As the synthesis of a diverse array of S1PR agonists continues, with FTY720 being a prime example, the body of experimental evidence advocating for their role in IRI treatment is expanding. Despite this progress, comprehensive reviews delineating the therapeutic landscape of S1PR agonists in IRI remain limited. This review aspires to meticulously elucidate the protective roles and mechanisms of S1PR agonists in preventing and managing IRI affecting various organs, including the heart, kidney, liver, lungs, intestines, and brain, to foster novel pharmacological approaches in clinical settings.
Collapse
Affiliation(s)
- Linyuan Wang
- Department of Cardiovascular Ultrasound, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China; The Central Laboratory of The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China
| | - Xiaowen Zhang
- Medical Research Center, Shengjing Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China
| | - Chunyan Ma
- Department of Cardiovascular Ultrasound, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China.
| | - Nan Wu
- The Central Laboratory of The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China.
| |
Collapse
|
6
|
Ya'ar Bar S, Pintel N, Abd Alghne H, Khattib H, Avni D. The therapeutic potential of sphingolipids for cardiovascular diseases. Front Cardiovasc Med 2023; 10:1224743. [PMID: 37608809 PMCID: PMC10440740 DOI: 10.3389/fcvm.2023.1224743] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Accepted: 07/17/2023] [Indexed: 08/24/2023] Open
Abstract
Cardiovascular diseases (CVDs) are the leading cause of morbidity and mortality worldwide and Inflammation plays a critical role in the development of CVD. Despite considerable progress in understanding the underlying mechanisms and various treatment options available, significant gaps in therapy necessitate the identification of novel therapeutic targets. Sphingolipids are a family of lipids that have gained attention in recent years as important players in CVDs and the inflammatory processes that underlie their development. As preclinical studies have shown that targeting sphingolipids can modulate inflammation and ameliorate CVDs, targeting sphingolipids has emerged as a promising therapeutic strategy. This review discusses the current understanding of sphingolipids' involvement in inflammation and cardiovascular diseases, the existing therapeutic approaches and gaps in therapy, and explores the potential of sphingolipids-based drugs as a future avenue for CVD treatment.
Collapse
Affiliation(s)
- Sapir Ya'ar Bar
- Department of Natural Compound, Nutrition, and Health, MIGAL, Kiryat Shmona, Israel
| | - Noam Pintel
- Department of Natural Compound, Nutrition, and Health, MIGAL, Kiryat Shmona, Israel
| | - Hesen Abd Alghne
- Department of Natural Compound, Nutrition, and Health, MIGAL, Kiryat Shmona, Israel
- Tel-Hai College Department of Biotechnology, Kiryat Shmona, Israel
| | - Hamdan Khattib
- Department of Natural Compound, Nutrition, and Health, MIGAL, Kiryat Shmona, Israel
- Department of Gastroenterology and Hepatology, Tel Aviv University Sackler Faculty of Medicine, Tel Aviv, Israel
| | - Dorit Avni
- Department of Natural Compound, Nutrition, and Health, MIGAL, Kiryat Shmona, Israel
- Tel-Hai College Department of Biotechnology, Kiryat Shmona, Israel
| |
Collapse
|
7
|
Jung E, Capel R, Jiang C, Venturi E, Neagu G, Pearcey S, Zhou Y, Zhang Y, Lei M. Cardiac deficiency of P21-activated kinase 1 promotes atrial arrhythmogenesis in mice following adrenergic challenge. Philos Trans R Soc Lond B Biol Sci 2023; 378:20220168. [PMID: 37122217 PMCID: PMC10150202 DOI: 10.1098/rstb.2022.0168] [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: 09/16/2022] [Accepted: 11/24/2022] [Indexed: 05/02/2023] Open
Abstract
P21-activated kinase 1 (Pak1) signalling plays a vital and overall protective role in the heart. However, the phenotypes of Pak1 deficiency in the cardiac atria have not been well explored. In this study, Pak1 cardiac-conditional knock-out (cKO) mice were studied under baseline and adrenergic challenge conditions. Pak1 cKO mice show atrial arrhythmias including atrial fibrillation (AF) in vivo, detected during anaesthetized electrocardiography without evidence of interstitial fibrosis upon Masson's trichrome staining. Optical mapping of left atrial preparations from Pak1 cKO mice revealed a higher incidence of Ca2+ and action potential alternans under isoprenaline challenge and differences in baseline action potential and calcium transient characteristics. Type-2 ryanodine receptor (RyR2) channels from Pak1 cKO hearts had a higher open probability than those from wild-type. Reverse transcription-quantitative polymerase chain reaction and Western blotting indicated that pCamkIIδ and RyR2 are highly phosphorylated at baseline in the atria of Pak1 cKO mice, while the expression of Slc8a2 and Slc8a3 as a Na+-Ca2+ exchanger, controlling the influx of Ca2+ from outside of the cell and efflux of Na+ from the cytoplasm, are augmented. Chromatin immunoprecipitation study showed that pCreb1 interacts with Slc8a2 and Slc8a3. Our study thus demonstrates that deficiency of Pak1 promotes atrial arrhythmogenesis under adrenergic stress, probably through post-translational and transcriptional modifications of key molecules that are critical to Ca2+ homeostasis. This article is part of the theme issue 'The heartbeat: its molecular basis and physiological mechanisms'.
Collapse
Affiliation(s)
- Eunjeong Jung
- Department of Pharmacology, University of Oxford, Oxford OX1 3QT, UK
| | - Rebecca Capel
- Department of Pharmacology, University of Oxford, Oxford OX1 3QT, UK
| | - Congshan Jiang
- National Regional Children's Medical Center (Northwest); Key Laboratory of Precision Medicine to Pediatric Diseases of Shaanxi Province; Xi'an Key Laboratory of Children's Health and Diseases, Shaanxi Institute for Pediatric Diseases; Xi'an Children's Hospital, Affiliated Children's Hospital of Xi'an Jiaotong University. No. 69, Xijuyuan Lane, Xi'an 710003, People's Republic of China
| | - Elisa Venturi
- Department of Pharmacology, University of Oxford, Oxford OX1 3QT, UK
| | - Georgiana Neagu
- Department of Pharmacology, University of Oxford, Oxford OX1 3QT, UK
| | - Sarah Pearcey
- Paediatric Intensive Care, Addenbrooke's Hospital, Cambridge CB2 1QY, UK
| | - Yafei Zhou
- National Regional Children's Medical Center (Northwest); Key Laboratory of Precision Medicine to Pediatric Diseases of Shaanxi Province; Xi'an Key Laboratory of Children's Health and Diseases, Shaanxi Institute for Pediatric Diseases; Xi'an Children's Hospital, Affiliated Children's Hospital of Xi'an Jiaotong University. No. 69, Xijuyuan Lane, Xi'an 710003, People's Republic of China
- Key Laboratory of Medical Electrophysiology of the Ministry of Education, Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan, 646000, People's Republic of China
| | - Yanmin Zhang
- National Regional Children's Medical Center (Northwest); Key Laboratory of Precision Medicine to Pediatric Diseases of Shaanxi Province; Xi'an Key Laboratory of Children's Health and Diseases, Shaanxi Institute for Pediatric Diseases; Xi'an Children's Hospital, Affiliated Children's Hospital of Xi'an Jiaotong University. No. 69, Xijuyuan Lane, Xi'an 710003, People's Republic of China
- Institute of Cardiovascular Sciences, Faculty of Medicine and Human Science, University of Manchester, Manchester, M13, 9GB UK
| | - Ming Lei
- Department of Pharmacology, University of Oxford, Oxford OX1 3QT, UK
- Key Laboratory of Medical Electrophysiology of the Ministry of Education, Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan, 646000, People's Republic of China
| |
Collapse
|
8
|
Pereira CH, Bare DJ, Rosas PC, Dias FAL, Banach K. The role of P21-activated kinase (Pak1) in sinus node function. J Mol Cell Cardiol 2023; 179:90-101. [PMID: 37086972 PMCID: PMC10294268 DOI: 10.1016/j.yjmcc.2023.04.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 04/11/2023] [Accepted: 04/11/2023] [Indexed: 04/24/2023]
Abstract
Sinoatrial node (SAN) dysfunction (SND) and atrial arrhythmia frequently occur simultaneously with a hazard ratio of 4.2 for new onset atrial fibrillation (AF) in SND patients. In the atrial muscle attenuated activity of p21-activated kinase 1 (Pak1) increases the risk for AF by enhancing NADPH oxidase 2 dependent production of reactive oxygen species (ROS). However, the role of Pak1 dependent ROS regulation in SAN function has not yet been determined. We hypothesize that Pak1 activity maintains SAN activity by regulating the expression of the hyperpolarization activated cyclic nucleotide gated cation channel (HCN). To determine Pak1 dependent changes in heart rate (HR) regulation we quantified the intrinsic sinus rhythm in wild type (WT) and Pak1 deficient (Pak1-/-) mice of both sexes in vivo and in isolated Langendorff perfused hearts. Pak1-/- hearts displayed an attenuated HR in vivo after autonomic blockage and in isolated hearts. The contribution of the Ca2+ clock to pacemaker activity remained unchanged, but Ivabradine (3 μM), a blocker of HCN channels that are a membrane clock component, eliminated the differences in SAN activity between WT and Pak1-/- hearts. Reduced HCN4 expression was confirmed in Pak1-/- right atria. The reduced HCN activity in Pak1-/- could be rescued by class II HDAC inhibition (LMK235), ROS scavenging (TEMPOL) or attenuation of Extracellular Signal-Regulated Kinase (ERK) 1/2 activity (SCH772984). No sex specific differences in Pak1 dependent SAN regulation were determined. Our results establish Pak1 as a class II HDAC regulator and a potential therapeutic target to attenuate SAN bradycardia and AF susceptibility.
Collapse
Affiliation(s)
- Carlos H Pereira
- Dept. of Internal Medicine/Cardiology, Rush University Medical Center, 1750 W. Harrison St., Chicago, IL 60612, USA; Biological Science Center, Department of Physiology, Av. Cel Francisco H. dos Santos 100, 19031 Centro Politécnico-Curitiba, Brazil.
| | - Dan J Bare
- Dept. of Physiology & Biophysics, The Ohio State University, 5018 Graves Hall, 333 W.10th Ave., Columbus, OH 4321, USA.
| | - Paola C Rosas
- Dept. of Pharmacy Practice, College of Pharmacy, 833 S Wood St., Chicago, IL 60612, USA.
| | - Fernando A L Dias
- Biological Science Center, Department of Physiology, Av. Cel Francisco H. dos Santos 100, 19031 Centro Politécnico-Curitiba, Brazil.
| | - Kathrin Banach
- Dept. of Internal Medicine/Cardiology, Rush University Medical Center, 1750 W. Harrison St., Chicago, IL 60612, USA.
| |
Collapse
|
9
|
Che YJ, Ren XH, Wang ZW, Wu Q, Xing K, Zhang M, Xu C, Han D, Yuan S, Zheng SH, Chen YY, Liao XR, Shi F, Zhong XH, Cai X, Cheng SX. Lymph-Node-Targeted Drug Delivery for Effective Immunomodulation to Prolong the Long-Term Survival After Heart Transplantation. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2207227. [PMID: 36314402 DOI: 10.1002/adma.202207227] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Indexed: 06/16/2023]
Abstract
The chronic rejection responses and side effects of the systematic administration of immunosuppressants are the main obstacles to heart allograft and patient survival. The development of xenotransplantation also urgently requires more efficient immune regulation strategies. Herein, it is demonstrated that lymph-node (LN)-targeted drug delivery can realize LN-specific immunomodulation with attenuated immune suppression on distant peripheral immune organs to effectively prolong long-term survival after heart transplantation in a chronic murine heart transplantation model. A chemokine C-C motif ligand 21 (CCL21) specific aptamer for LN targeting is decorated onto the surface of the hybrid nanoparticular delivery vector mainly composed of CaCO3 /CaP/heparin. The targeting delivery system can dramatically enhance accumulation of the loaded immunosuppressant, fingolimod hydrochloride (FTY720), in draining lymph nodes (dLNs) for inducing powerful immune suppression. By promoting the generation of endogenous regulatory T cells (Tregs ) and decreasing the proportion of effector T cells (Teffs ) in dLNs after heart transplantation, the LN-targeting strategy can effectively regulate local immune responses instead of systemic immunity, which reduces the incidence of long-term complications. This study provides an efficient strategy to improve the survival rate after organ transplantation by precise and localized immunoregulation with minimized side effects of immunosuppression.
Collapse
Affiliation(s)
- Yan-Jia Che
- Department of Cardiovascular Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, P. R. China
| | - Xiao-He Ren
- Key Laboratory of Biomedical Polymers of Ministry of Education, Department of Chemistry, Wuhan University, Wuhan, 430072, P. R. China
| | - Zhi-Wei Wang
- Department of Cardiovascular Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, P. R. China
| | - Qi Wu
- Department of Cardiovascular Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, P. R. China
| | - Kai Xing
- Department of Cardiovascular Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, P. R. China
| | - Min Zhang
- Department of Cardiovascular Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, P. R. China
| | - Chang Xu
- Key Laboratory of Biomedical Polymers of Ministry of Education, Department of Chemistry, Wuhan University, Wuhan, 430072, P. R. China
| | - Di Han
- Key Laboratory of Biomedical Polymers of Ministry of Education, Department of Chemistry, Wuhan University, Wuhan, 430072, P. R. China
| | - Shun Yuan
- Department of Cardiovascular Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, P. R. China
| | - Si-Hao Zheng
- Department of Cardiovascular Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, P. R. China
| | - Yuan-Yang Chen
- Department of Cardiovascular Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, P. R. China
| | - Xin-Ru Liao
- Key Laboratory of Biomedical Polymers of Ministry of Education, Department of Chemistry, Wuhan University, Wuhan, 430072, P. R. China
| | - Feng Shi
- Department of Cardiovascular Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, P. R. China
| | - Xiao-Han Zhong
- Department of Cardiovascular Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, P. R. China
| | - Xin Cai
- Department of Cardiovascular Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, P. R. China
| | - Si-Xue Cheng
- Key Laboratory of Biomedical Polymers of Ministry of Education, Department of Chemistry, Wuhan University, Wuhan, 430072, P. R. China
| |
Collapse
|
10
|
Spampinato SF, Sortino MA, Salomone S. Sphingosine-1-phosphate and Sphingosine-1-phosphate receptors in the cardiovascular system: pharmacology and clinical implications. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2022; 94:95-139. [PMID: 35659378 DOI: 10.1016/bs.apha.2022.02.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Sphingosine-1-phosphate (S1P) is a lipid that binds and activates five distinct receptor subtypes, S1P1, S1P2, S1P3, S1P4, S1P5, widely expressed in different cells, tissues and organs. In the cardiovascular system these receptors have been extensively studied, but no drug acting on them has been approved so far for treating cardiovascular diseases. In contrast, a number of S1P receptor agonists are approved as immunomodulators, mainly for multiple sclerosis, because of their action on lymphocyte trafficking. This chapter summarizes the available information on S1P receptors in the cardiovascular system and discusses their potential for treating cardiovascular conditions and/or their role on the clinical pharmacology of drugs so far approved for non-cardiovascular conditions. Basic research has recently produced data useful to understand the molecular pharmacology of S1P and S1P receptors, regarding biased agonism, S1P storage, release and vehiculation and chaperoning by lipoproteins, paracrine actions, intracellular non-receptorial S1P actions. On the other hand, the approval of fingolimod and newer generation S1P receptor ligands as immunomodulators, provides information on a number of clinical observations on the impact of these drugs on cardiovascular system which need to be integrated with preclinical data. S1P receptors are potential targets for prevention and treatment of major cardiovascular conditions, including hypertension, myocardial infarction, heart failure and stroke.
Collapse
Affiliation(s)
| | - Maria Angela Sortino
- Department of Biomedical and Biotechnological Science, University of Catania, Catania, Italy
| | - Salvatore Salomone
- Department of Biomedical and Biotechnological Science, University of Catania, Catania, Italy.
| |
Collapse
|
11
|
Yang B, Jiang Q, He S, Li T, Ou X, Chen T, Fan X, Jiang F, Zeng X, Huang CLH, Lei M, Tan X. Ventricular SK2 upregulation following angiotensin II challenge: Modulation by p21-activated kinase-1. J Mol Cell Cardiol 2022; 164:110-125. [PMID: 34774547 DOI: 10.1016/j.yjmcc.2021.11.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Revised: 10/15/2021] [Accepted: 11/03/2021] [Indexed: 12/11/2022]
Abstract
Effects of hypertrophic challenge on small-conductance, Ca2+-activated K+(SK2) channel expression were explored in intact murine hearts, isolated ventricular myocytes and neonatal rat cardiomyocytes (NRCMs). An established experimental platform applied angiotensin II (Ang II) challenge in the presence and absence of reduced p21-activated kinase (PAK1) (PAK1cko vs. PAK1f/f, or shRNA-PAK1 interference) expression. SK2 current contributions were detected through their sensitivity to apamin block. Ang II treatment increased such SK2 contributions to optically mapped action potential durations (APD80) and their heterogeneity, and to patch-clamp currents. Such changes were accentuated in PAK1cko compared to PAK1f/f, intact hearts and isolated cardiomyocytes. They paralleled increased histological and echocardiographic hypertrophic indices, reduced cardiac contractility, and increased SK2 protein expression, changes similarly greater with PAK1cko than PAK1f/f. In NRCMs, Ang II challenge replicated such increases in apamin-sensitive SK patch clamp currents as well as in real-time PCR and western blot measures of SK2 mRNA and protein expression and cell hypertrophy. Furthermore, the latter were enhanced by shRNA-PAK1 interference and mitigated by the PAK1 agonist FTY720. Increased CaMKII and CREB phosphorylation accompanied these effects. These were rescued by both FTY720 as well as the CaMKII inhibitor KN93, but not its inactive analogue KN92. Such CREB then specifically bound to the KCNN2 promoter sequence in luciferase assays. These findings associate Ang II induced hypertrophy with increased SK2 expression brought about by a CaMKII/CREB signaling convergent with the PAK1 pathway thence upregulating the KCNN2 promoter activity. SK2 may then influence cardiac electrophysiology under conditions of cardiac hypertrophy and failure.
Collapse
Affiliation(s)
- Binbin Yang
- Key Laboratory of Medical Electrophysiology of the Ministry of Education, Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan 646000, China; Oral & Maxillofacial Reconstruction and Regeneration Laboratory, The Affiliated Stomatology Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Qin Jiang
- Key Laboratory of Medical Electrophysiology of the Ministry of Education, Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan 646000, China; Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Shicheng He
- Key Laboratory of Medical Electrophysiology of the Ministry of Education, Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan 646000, China; Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Tao Li
- Key Laboratory of Medical Electrophysiology of the Ministry of Education, Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Xianhong Ou
- Key Laboratory of Medical Electrophysiology of the Ministry of Education, Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Tangting Chen
- Key Laboratory of Medical Electrophysiology of the Ministry of Education, Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Xuehui Fan
- Key Laboratory of Medical Electrophysiology of the Ministry of Education, Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Feng Jiang
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Xiaorong Zeng
- Key Laboratory of Medical Electrophysiology of the Ministry of Education, Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Christopher L-H Huang
- Key Laboratory of Medical Electrophysiology of the Ministry of Education, Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan 646000, China; Physiological Laboratory and Department of Biochemistry, University of Cambridge, Cambridge CB2 3EG, UK
| | - Ming Lei
- Key Laboratory of Medical Electrophysiology of the Ministry of Education, Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan 646000, China; Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, UK.
| | - Xiaoqiu Tan
- Key Laboratory of Medical Electrophysiology of the Ministry of Education, Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan 646000, China; Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China.
| |
Collapse
|
12
|
Yoon KS, Cha HJ, Choi SO, Lee JM. 2-((2-(4-Iodo-2,5-dimethoxyphenyl)ethylamino)methyl)phenol (25I-NBOH) and 2-(((2-(4-chloro-2,5-dimethoxyphenyl)ethyl)amino)methyl)phenol (25C-NBOH) induce adverse effects on the cardiovascular system. Toxicol Lett 2022; 355:160-169. [PMID: 34843874 DOI: 10.1016/j.toxlet.2021.11.016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 10/30/2021] [Accepted: 11/24/2021] [Indexed: 12/14/2022]
Abstract
Two new psychoactive substances (NPSs) classified as phenethylamines, namely 2-((2-(4-Iodo-2,5-dimethoxyphenyl)ethylamino)methyl)phenol (25I-NBOH) and 2-(((2-(4-chloro-2,5-dimethoxyphenyl)ethyl)amino)methyl)phenol (25C-NBOH), are being abused by people seeking recreational hallucinogens. These NPSs may cause serious health problems as their adverse effects are not known in most cases. Therefore, in the present study, we evaluated the cardiotoxicity of 25I-NBOH and 25C-NBOH using 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay, rat electrocardiography (ECG), Langendorff test, and human ether-a-go-go-related gene (hERG) assay. Furthermore, we analyzed the expression levels of p21 CDC42/RAC1-activated kinase 1 (PAK1), which is known to play various roles in the cardiovascular system. In the MTT assay, treatment with 25I-NBOH or 25C-NBOH dramatically decreased viability of H9c2 cardiomyocytes. Meanwhile, these two compounds significantly increased QT intervals and RR intervals in the rat ECG measurement. 25I-NBOH down-regulated the PAK1 protein expression in rat primary cardiomyocytes as well as H9c2 cells. However, 25C-NBOH had no effect on the PAK1 expression in H9c2 cells. In an in-depth study, 25I-NBOH inhibited potassium channels in the hERG assay, but in ex vivo test, the substance did not affect the left ventricular developed pressure (LVDP) and heart rate of the isolated rat hearts. Taken together, these results suggest that both 25I-NBOH and 25C-NBOH may have adverse cardiovascular effect. Further investigation would be needed to determine which factors mainly influence the relationship between PAK1 expression and cardiotoxicity.
Collapse
Affiliation(s)
- Kyung Sik Yoon
- National Institute of Food and Drug Safety Evaluation, Ministry of Food and Drug Safety, Osong, Cheongju, Republic of Korea.
| | - Hye Jin Cha
- National Institute of Food and Drug Safety Evaluation, Ministry of Food and Drug Safety, Osong, Cheongju, Republic of Korea.
| | - Sun Ok Choi
- National Institute of Food and Drug Safety Evaluation, Ministry of Food and Drug Safety, Osong, Cheongju, Republic of Korea.
| | - Jin-Moo Lee
- National Institute of Food and Drug Safety Evaluation, Ministry of Food and Drug Safety, Osong, Cheongju, Republic of Korea.
| |
Collapse
|
13
|
Alatrag F, Amoni M, Kelly-Laubscher R, Gwanyanya A. Cardioprotective effect of fingolimod against calcium paradox-induced myocardial injury in the isolated rat heart. Can J Physiol Pharmacol 2022; 100:134-141. [PMID: 34559972 DOI: 10.1139/cjpp-2021-0381] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Fingolimod (FTY720) inhibits Ca2+-permeable, Mg2+-sensitive channels called transient receptor potential melastatin 7 (TRPM7), but its effects on Ca2+ paradox (CP) - induced myocardial damage has not been evaluated. We studied the effect of FTY720 on CP-induced myocardial damage and used other TRPM7 channel inhibitors nordihydroguaiaretic acid (NDGA) and Mg2+ to test if any effect of FTY720 was via TRPM7 inhibition. Langendorff-perfused Wistar rat hearts were treated with FTY720 or NDGA and subjected to a CP protocol consisting of Ca2+ depletion followed by Ca2+ repletion. Hearts of rats pre-treated with MgSO4 were also subjected to CP. Hemodynamic parameters were measured using an intraventricular balloon, and myocardial infarct size was quantified using triphenyltetrazolium chloride stain. TRPM7 proteins in ventricular tissue were detected using immunoblot analysis. FTY720, but not NDGA, decreased CP-induced infarct size. Both FTY720 and NDGA minimized the CP-induced elevation of left ventricular end-diastolic pressure, but only FTY720 ultimately improved ventricular developed pressure. Mg2+ pre-treatment had no effect on CP-induced infarct size, nor hemodynamic parameters during CP, nor the level of TRPM7 protein expression in ventricular tissue. Overall, FTY720 attenuated CP-induced myocardial damage, with potential therapeutic implications on Ca2+-mediated cardiotoxicity; however, the cardioprotective mechanism of FTY720 seems to be unrelated to TRPM7 channel modulation.
Collapse
Affiliation(s)
- Fatma Alatrag
- Department of Human Biology, Faculty of Health Sciences, University of Cape Town, Observatory 7925, Cape Town, South Africa
| | - Matthew Amoni
- Department of Human Biology, Faculty of Health Sciences, University of Cape Town, Observatory 7925, Cape Town, South Africa
| | - Roisin Kelly-Laubscher
- Department of Pharmacology and Therapeutics, The College of Medicine and Health, University College Cork, Ireland
- Department of Biological Sciences, Faculty of Science, University of Cape Town, Rondebosch 7700, Cape Town, South Africa
| | - Asfree Gwanyanya
- Department of Human Biology, Faculty of Health Sciences, University of Cape Town, Observatory 7925, Cape Town, South Africa
| |
Collapse
|
14
|
Yoon KS, Gu SM, Cha HJ, Kim YH, Yun J, Lee JM. 25I-NBOMe, a phenethylamine derivative, induces adverse cardiovascular effects in rodents: possible involvement of p21 (CDC42/RAC)-activated kinase 1. Drug Chem Toxicol 2020; 45:898-906. [PMID: 32597268 DOI: 10.1080/01480545.2020.1784924] [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] [Indexed: 02/06/2023]
Abstract
Abuse of new psychoactive substances is an emerging social problem. Several phenethylamines are internationally controlled substances as they are likely to be abused and have adverse effects. Phenethylamine analog 2-(4-iodo-2,5-dimethoxyphenyl)-N-(2-methoxybenzyl)ethanamine (25I-NBOMe) has been reported as one of the most commonly abused psychoactive substance. However, the cardiotoxicity of this compound has not been extensively evaluated. Thus, in this study, we investigated the adverse cardiovascular effects of 25I-NBOMe, related to p21 (CDC42/RAC)-activated kinase 1 (PAK1). The cardiotoxicity of 25I-NBOMe was evaluated using the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay, live/dead cytotoxicity assay, PAK1/CDC42 kinase assay, and in vivo electrocardiography (ECG). Also, we analyzed the expression level of PAK1, which is known to play key roles in the cardiovascular system. In the MTT assay, cell viability of 25I-NBOMe-treated H9c2 cells or primary cardiomyocytes of ICR mice decreased in a concentration-dependent manner. Results from the in vitro cytotoxicity assay in cardiomyocytes showed that 25I-NBOMe decreased the viability of H9c2 rat cardiomyocytes, and TC50 of 25I-NBOMe was found to be 70.4 μM. We also observed that 25I-NBOMe reduced PAK1 activity in vitro. Surface ECG measurement revealed that intravenous injection of 25I-NBOMe (doses of 1.0 and 3.0 mg/kg, corresponding to serum concentrations of 18.1 and 28.6 ng/mL, respectively) prolonged the QTc interval in SD rats. Furthermore, treatment with 25I-NBOMe downregulated the expression of PAK1 in the hearts of SD rats and H9c2 cells. In summary, our findings indicate that PAK1-related adverse effects of 25I-NBOMe can cause toxicity to cardiomyocytes and induce an abnormal ECG pattern in animals.
Collapse
Affiliation(s)
- Kyung Sik Yoon
- Ministry of Food and Drug Safety, National Institute of Food and Drug Safety Evaluation, Cheongju-si, Republic of Korea
| | - Sun Mi Gu
- College of Pharmacy and Medical Research Center, Chungbuk National University, Cheongju-si, Republic of Korea
| | - Hye Jin Cha
- Ministry of Food and Drug Safety, National Institute of Food and Drug Safety Evaluation, Cheongju-si, Republic of Korea
| | - Young-Hoon Kim
- Ministry of Food and Drug Safety, National Institute of Food and Drug Safety Evaluation, Cheongju-si, Republic of Korea
| | - Jaesuk Yun
- College of Pharmacy and Medical Research Center, Chungbuk National University, Cheongju-si, Republic of Korea
| | - Jin-Moo Lee
- Ministry of Food and Drug Safety, National Institute of Food and Drug Safety Evaluation, Cheongju-si, Republic of Korea
| |
Collapse
|
15
|
Liu C, Tang M, Zhang X, Li J, Cao G. Knockdown of miR-665 Protects Against Cardiomyocyte Ischemia/Reperfusion Injury-Induced ROS Accumulation and Apoptosis Through the Activation of Pak1/Akt Signaling in Myocardial Infarction. Int Heart J 2020; 61:347-354. [DOI: 10.1536/ihj.19-416] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Affiliation(s)
- Chuanzhen Liu
- Department of Cardiovascular Surgery, Qilu Hospital of Shandong University
| | - Mengmeng Tang
- Department of Cardiovascular Surgery, Qilu Hospital of Shandong University
| | - Xiquan Zhang
- Department of Cardiovascular Surgery, Qilu Hospital of Shandong University
| | - Jianhua Li
- Department of Cardiovascular Surgery, Qilu Hospital of Shandong University
| | - Guangqing Cao
- Department of Cardiovascular Surgery, Qilu Hospital of Shandong University
| |
Collapse
|
16
|
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: 20] [Impact Index Per Article: 5.0] [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
|
17
|
Nassal D, Gratz D, Hund TJ. Challenges and Opportunities for Therapeutic Targeting of Calmodulin Kinase II in Heart. Front Pharmacol 2020; 11:35. [PMID: 32116711 PMCID: PMC7012788 DOI: 10.3389/fphar.2020.00035] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Accepted: 01/14/2020] [Indexed: 12/19/2022] Open
Abstract
Heart failure remains a major health burden around the world. Despite great progress in delineation of molecular mechanisms underlying development of disease, standard therapy has not advanced at the same pace. The multifunctional signaling molecule Ca2+/calmodulin-dependent protein kinase II (CaMKII) has received considerable attention over recent years for its central role in maladaptive remodeling and arrhythmias in the setting of chronic disease. However, these basic science discoveries have yet to translate into new therapies for human patients. This review addresses both the promise and barriers to developing translational therapies that target CaMKII signaling to abrogate pathologic remodeling in the setting of chronic disease. Efforts in small molecule design are discussed, as well as alternative targeting approaches that exploit novel avenues for compound delivery and/or genetic approaches to affect cardiac CaMKII signaling. These alternative strategies provide hope for overcoming some of the challenges that have limited the development of new therapies.
Collapse
Affiliation(s)
- Drew Nassal
- The Frick Center for Heart Failure and Arrhythmia and Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| | - Daniel Gratz
- The Frick Center for Heart Failure and Arrhythmia and Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, United States.,Department of Biomedical Engineering, College of Engineering, The Ohio State University, Columbus, OH, United States
| | - Thomas J Hund
- The Frick Center for Heart Failure and Arrhythmia and Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, United States.,Department of Biomedical Engineering, College of Engineering, The Ohio State University, Columbus, OH, United States.,Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| |
Collapse
|
18
|
Kovilakath A, Jamil M, Cowart LA. Sphingolipids in the Heart: From Cradle to Grave. Front Endocrinol (Lausanne) 2020; 11:652. [PMID: 33042014 PMCID: PMC7522163 DOI: 10.3389/fendo.2020.00652] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Accepted: 08/11/2020] [Indexed: 01/10/2023] Open
Abstract
Cardiovascular diseases are the leading cause of mortality worldwide and this has largely been driven by the increase in metabolic disease in recent decades. Metabolic disease alters metabolism, distribution, and profiles of sphingolipids in multiple organs and tissues; as such, sphingolipid metabolism and signaling have been vigorously studied as contributors to metabolic pathophysiology in various pathological outcomes of obesity, including cardiovascular disease. Much experimental evidence suggests that targeting sphingolipid metabolism may be advantageous in the context of cardiometabolic disease. The heart, however, is a structurally and functionally complex organ where bioactive sphingolipids have been shown not only to mediate pathological processes, but also to contribute to essential functions in cardiogenesis and cardiac function. Additionally, some sphingolipids are protective in the context of ischemia/reperfusion injury. In addition to mechanistic contributions, untargeted lipidomics approaches used in recent years have identified some specific circulating sphingolipids as novel biomarkers in the context of cardiovascular disease. In this review, we summarize recent literature on both deleterious and beneficial contributions of sphingolipids to cardiogenesis and myocardial function as well as recent identification of novel sphingolipid biomarkers for cardiovascular disease risk prediction and diagnosis.
Collapse
Affiliation(s)
- Anna Kovilakath
- Department of Human and Molecular Genetics, Virginia Commonwealth University, Richmond, VA, United States
| | - Maryam Jamil
- Department of Human and Molecular Genetics, Virginia Commonwealth University, Richmond, VA, United States
| | - Lauren Ashley Cowart
- Department of Biochemistry and Molecular Biology and the Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, United States
- Hunter Holmes McGuire Veteran's Affairs Medical Center, Richmond, VA, United States
- *Correspondence: Lauren Ashley Cowart
| |
Collapse
|
19
|
Liu X, Wu J, Zhu C, Liu J, Chen X, Zhuang T, Kuang Y, Wang Y, Hu H, Yu P, Fan H, Zhang Y, Liu Z, Zhang L. Endothelial S1pr1 regulates pressure overload-induced cardiac remodelling through AKT-eNOS pathway. J Cell Mol Med 2019; 24:2013-2026. [PMID: 31854513 PMCID: PMC6991681 DOI: 10.1111/jcmm.14900] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 11/07/2019] [Accepted: 11/19/2019] [Indexed: 12/15/2022] Open
Abstract
Cardiac vascular microenvironment is crucial for cardiac remodelling during the process of heart failure. Sphingosine 1‐phosphate (S1P) tightly regulates vascular homeostasis via its receptor, S1pr1. We therefore hypothesize that endothelial S1pr1 might be involved in pathological cardiac remodelling. In this study, heart failure was induced by transverse aortic constriction (TAC) operation. S1pr1 expression is significantly increased in microvascular endothelial cells (ECs) of post‐TAC hearts. Endothelial‐specific deletion of S1pr1 significantly aggravated cardiac dysfunction and deteriorated cardiac hypertrophy and fibrosis in myocardium. In vitro experiments demonstrated that S1P/S1pr1 praxis activated AKT/eNOS signalling pathway, leading to more production of nitric oxide (NO), which is an essential cardiac protective factor. Inhibition of AKT/eNOS pathway reversed the inhibitory effect of EC‐S1pr1‐overexpression on angiotensin II (AngII)‐induced cardiomyocyte (CM) hypertrophy, as well as on TGF‐β‐mediated cardiac fibroblast proliferation and transformation towards myofibroblasts. Finally, pharmacological activation of S1pr1 ameliorated TAC‐induced cardiac hypertrophy and fibrosis, leading to an improvement in cardiac function. Together, our results suggest that EC‐S1pr1 might prevent the development of pressure overload‐induced heart failure via AKT/eNOS pathway, and thus pharmacological activation of S1pr1 or EC‐targeting S1pr1‐AKT‐eNOS pathway could provide a future novel therapy to improve cardiac function during heart failure development.
Collapse
Affiliation(s)
- Xiuxiang Liu
- Key Laboratory of Arrhythmias of the Ministry of Education of China, Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jinjin Wu
- Cardiovascular Department, Shanghai Children's Medical Center, Shanghai Jiaotong University, Shanghai, China
| | - Chenying Zhu
- Heart Failure Institute, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jie Liu
- Key Laboratory of Arrhythmias of the Ministry of Education of China, Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Xiaoli Chen
- Key Laboratory of Arrhythmias of the Ministry of Education of China, Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Tao Zhuang
- Key Laboratory of Arrhythmias of the Ministry of Education of China, Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yashu Kuang
- Key Laboratory of Arrhythmias of the Ministry of Education of China, Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yanfang Wang
- Key Laboratory of Arrhythmias of the Ministry of Education of China, Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Hao Hu
- Heart Failure Institute, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Ping Yu
- Heart Failure Institute, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Huimin Fan
- Heart Failure Institute, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yuzhen Zhang
- Key Laboratory of Arrhythmias of the Ministry of Education of China, Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Zhongmin Liu
- Department of Cardiology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Lin Zhang
- Key Laboratory of Arrhythmias of the Ministry of Education of China, Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
20
|
Yoon KS, Lee JM, Kim YH, Suh SK, Cha HJ. Cardiotoxic effects of [3-[2-(diethylamino)ethyl]-1H-indol-4-yl] acetate and 3-[2-[ethyl(methyl)amino]ethyl]-1H-indol-4-ol. Toxicol Lett 2019; 319:40-48. [PMID: 31706004 DOI: 10.1016/j.toxlet.2019.10.022] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Revised: 10/06/2019] [Accepted: 10/27/2019] [Indexed: 12/28/2022]
Abstract
Two synthetic tryptamines, namely [3-[2-(diethylamino)ethyl]-1H-indol-4-yl] acetate (4-AcO-DET) and 3-[2-[ethyl(methyl)amino]ethyl]-1H-indol-4-ol (4-HO-MET), are abused by individuals seeking recreational hallucinogens. These new psychoactive substances (NPSs) can cause serious health problems because their adverse effects are mostly unknown. In the present study, we evaluated the cardiotoxicity of 4-AcO-DET and 4-HO-MET using the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay, electrocardiography (ECG), and the human ether-a-go-go-related gene (hERG) assay. In addition, we analyzed the expression level of p21 (CDC42/RAC)-activated kinase 1 (PAK1), which is known to play various roles in the cardiovascular system. In the MTT assay, 4-AcO-DET- and 4-HO-MET-treated H9c2 cells proliferated in a concentration-dependent manner. Moreover, both substances increased QT intervals (as determined using ECG) in Sprague-Dawley rats and inhibited potassium channels (as verified by the hERG assay) in Chinese hamster ovary cells. However, there was no change in PAK1 expression. Collectively, the results indicated that 4-AcO-DET and 4-HO-MET might cause adverse effects on the cardiovascular system. Further studies are required to confirm the relationship between PAK1 expression and cardiotoxicity. The findings of the present study would provide science-based evidence for scheduling the two NPSs.
Collapse
Affiliation(s)
- Kyung Sik Yoon
- National Institute of Drug and Safety Evaluation, Ministry of Food and Drug Safety, Osong, Cheongju, Republic of Korea.
| | - Jin-Moo Lee
- National Institute of Drug and Safety Evaluation, Ministry of Food and Drug Safety, Osong, Cheongju, Republic of Korea.
| | - Young-Hoon Kim
- National Institute of Drug and Safety Evaluation, Ministry of Food and Drug Safety, Osong, Cheongju, Republic of Korea.
| | - Soo Kyung Suh
- National Institute of Drug and Safety Evaluation, Ministry of Food and Drug Safety, Osong, Cheongju, Republic of Korea.
| | - Hye Jin Cha
- National Institute of Drug and Safety Evaluation, Ministry of Food and Drug Safety, Osong, Cheongju, Republic of Korea.
| |
Collapse
|
21
|
Alessenko AV, Lebedev AT, Kurochkin IN. The Role of Sphingolipids in Cardiovascular Pathologies. BIOCHEMISTRY (MOSCOW), SUPPLEMENT SERIES B: BIOMEDICAL CHEMISTRY 2019. [DOI: 10.1134/s1990750819020021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
22
|
Alessenko AV, Lebedev АТ, Kurochkin IN. [The role of sphingolipids in cardiovascular pathologies]. BIOMEDIT︠S︡INSKAI︠A︡ KHIMII︠A︡ 2019; 64:487-495. [PMID: 30632976 DOI: 10.18097/pbmc20186406487] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Cardiovascular diseases (CVD) remain the leading cause of death in industrialized countries. One of the most significant risk factors for atherosclerosis is hypercholesterolemia. Its diagnostics is based on routine lipid profile analysis, including the determination of total cholesterol, low and high density lipoprotein cholesterol, and triglycerides. However in recent years, much attention has been paid to the crosstalk between the metabolic pathways of the cholesterol and sphingolipids biosynthesis. Sphingolipids are a group of lipids, containing a molecule of aliphatic alcohol sphingosine. These include sphingomyelins, cerebrosides, gangliosides and ceramides, sphingosines, and sphingosine-1-phosphate (S-1-P). It has been found that catabolism of sphingolipids is associated with catabolism of cholesterol. However, the exact mechanism of this interaction is still unknown. Particular attention as CVD inducer attracts ceramide (Cer). Lipoprotein aggregates isolated from atherosclerotic pluques are enriched with Cer. The level of Cer and sphingosine increases after ischemia reperfusion of the heart, in the infarction zone and in the blood, and also in hypertension. S-1-P exhibits pronounced cardioprotective properties. Its content sharply decreases with ischemia and myocardial infarction. S-1-P presents predominantly in HDL, and influences their multiple functions. Increased levels of Cer and sphingosine and decreased levels of S-1-P formed in the course of coronary heart disease can be an important factor in the development of atherosclerosis. It is proposed to use determination of sphingolipids in blood plasma as markers for early diagnosis of cardiac ischemia and for hypertension in humans. There are intensive studies aimed at correction of metabolism S-1-P. The most successful drugs are those that use S-1-P receptors as a targets, since all of its actions are receptor-mediated.
Collapse
Affiliation(s)
- A V Alessenko
- Institute of Biochemical Physics, Russian Academy of Sciences, Moscow, Russia
| | | | - I N Kurochkin
- Institute of Biochemical Physics, Russian Academy of Sciences, Moscow, Russia
| |
Collapse
|
23
|
Brand CS, Lighthouse JK, Trembley MA. Protective transcriptional mechanisms in cardiomyocytes and cardiac fibroblasts. J Mol Cell Cardiol 2019; 132:1-12. [PMID: 31042488 DOI: 10.1016/j.yjmcc.2019.04.023] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Revised: 04/19/2019] [Accepted: 04/22/2019] [Indexed: 12/13/2022]
Abstract
Heart failure is the leading cause of morbidity and mortality worldwide. Several lines of evidence suggest that physical activity and exercise can pre-condition the heart to improve the response to acute cardiac injury such as myocardial infarction or ischemia/reperfusion injury, preventing the progression to heart failure. It is becoming more apparent that cardioprotection is a concerted effort between multiple cell types and converging signaling pathways. However, the molecular mechanisms of cardioprotection are not completely understood. What is clear is that the mechanisms underlying this protection involve acute activation of transcriptional activators and their corresponding gene expression programs. Here, we review the known stress-dependent transcriptional programs that are activated in cardiomyocytes and cardiac fibroblasts to preserve function in the adult heart after injury. Focus is given to prominent transcriptional pathways such as mechanical stress or reactive oxygen species (ROS)-dependent activation of myocardin-related transcription factors (MRTFs) and transforming growth factor beta (TGFβ), and gene expression that positively regulates protective PI3K/Akt signaling. Together, these pathways modulate both beneficial and pathological responses to cardiac injury in a cell-specific manner.
Collapse
Affiliation(s)
- Cameron S Brand
- Department of Pharmacology, School of Medicine, University of California - San Diego, 9500 Gilman Drive, Biomedical Sciences Building, La Jolla, CA 92093, USA.
| | - Janet K Lighthouse
- Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, 601 Elmwood Avenue, Box CVRI, Rochester, NY 14624, USA.
| | - Michael A Trembley
- Department of Cardiology, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA 02115, USA.
| |
Collapse
|
24
|
Solution structures and biophysical analysis of full-length group A PAKs reveal they are monomeric and auto-inhibited in cis. Biochem J 2019; 476:1037-1051. [PMID: 30858169 PMCID: PMC6448136 DOI: 10.1042/bcj20180867] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Revised: 03/05/2019] [Accepted: 03/08/2019] [Indexed: 12/13/2022]
Abstract
The group A p21-activated kinases (PAKs) exist in an auto-inhibited form until activated by GTPase binding and auto-phosphorylation. In the auto-inhibited form, a regulatory domain binds to the kinase domain (KD) blocking the binding of substrates, and CDC42 or Rac binding to the regulatory domain relieves this auto-inhibition allowing auto-phosphorylation on the KD activation loop. We have determined the crystal structure of the PAK3 catalytic domain and by small angle X-ray scattering, the solution-phase structures of full-length inactive PAK1 and PAK3. The structures reveal a compact but elongated molecular shape that demonstrates that, together with multiple independent biophysical measurements and in contrast with previous assumptions, group A PAKs are monomeric both before and after activation, consistent with an activation mechanism of cis-auto-inhibition and initial cis-auto-phosphorylation, followed by transient dimerisation to allow trans-auto-phosphorylation for full activation, yielding a monomeric active PAK protein.
Collapse
|
25
|
Wang X, Li M, Yu Y, Liu G, Yu Y, Zou Y, Ge J, Chen R. FTY720 alleviates coxsackievirus B3‐induced myocarditis and inhibits viral replication through regulating sphingosine 1‐phosphate receptors and AKT/caspase‐3 pathways. J Cell Physiol 2019; 234:18029-18040. [PMID: 30843214 DOI: 10.1002/jcp.28434] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2018] [Revised: 02/03/2019] [Accepted: 02/14/2019] [Indexed: 01/08/2023]
Affiliation(s)
- Xinggang Wang
- Department of Cardiology, Key Laboratory of Viral Heart Diseases, Shanghai Institute of Cardiovascular Diseases Zhongshan Hospital, Fudan University, Ministry of Public Health Shanghai China
| | - Minghui Li
- Department of Cardiology, Key Laboratory of Viral Heart Diseases, Shanghai Institute of Cardiovascular Diseases Zhongshan Hospital, Fudan University, Ministry of Public Health Shanghai China
| | - Ying Yu
- Department of Cardiology, Key Laboratory of Viral Heart Diseases, Shanghai Institute of Cardiovascular Diseases Zhongshan Hospital, Fudan University, Ministry of Public Health Shanghai China
| | - Guijian Liu
- Department of Cardiology, Key Laboratory of Viral Heart Diseases, Shanghai Institute of Cardiovascular Diseases Zhongshan Hospital, Fudan University, Ministry of Public Health Shanghai China
| | - Yong Yu
- Department of Cardiology, Key Laboratory of Viral Heart Diseases, Shanghai Institute of Cardiovascular Diseases Zhongshan Hospital, Fudan University, Ministry of Public Health Shanghai China
| | - Yunzeng Zou
- Department of Cardiology, Key Laboratory of Viral Heart Diseases, Shanghai Institute of Cardiovascular Diseases Zhongshan Hospital, Fudan University, Ministry of Public Health Shanghai China
| | - Junbo Ge
- Department of Cardiology, Key Laboratory of Viral Heart Diseases, Shanghai Institute of Cardiovascular Diseases Zhongshan Hospital, Fudan University, Ministry of Public Health Shanghai China
| | - Ruizhen Chen
- Department of Cardiology, Key Laboratory of Viral Heart Diseases, Shanghai Institute of Cardiovascular Diseases Zhongshan Hospital, Fudan University, Ministry of Public Health Shanghai China
| |
Collapse
|
26
|
Yoon KS, Yun J, Kim YH, Shin J, Kim SJ, Seo JW, Hyun SA, Suh SK, Cha HJ. 2-(2,5-Dimethoxy-4-methylphenyl)-N-(2-methoxybenzyl)ethanamine (25D-NBOMe) and N-(2-methoxybenzyl)-2,5-dimethoxy-4-chlorophenethylamine (25C-NBOMe) induce adverse cardiac effects in vitro and in vivo. Toxicol Lett 2019; 304:50-57. [PMID: 30658151 DOI: 10.1016/j.toxlet.2019.01.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Revised: 11/21/2018] [Accepted: 01/13/2019] [Indexed: 01/08/2023]
Abstract
Two emerging psychoactive substances, 2-(2,5-dimethoxy-4-methylphenyl)-N-(2-methoxybenzyl)ethanamine (25D-NBOMe) and N-(2-methoxybenzyl)-2,5-dimethoxy-4-chlorophenethylamine (25C-NBOMe), are being abused, leading to fatal and non-fatal intoxications. However, most of their adverse effects have been reported anecdotally. In the present study, cardiotoxicity was evaluated through 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay, rat electrocardiography (ECG), and human ether-a-go-go-related gene (hERG) assay. Expression levels of p21 (CDC42/RAC)-activated kinase 1 (PAK1), one of known biomarkers for cardiotoxicity, were also analyzed. Both 25D-NBOMe and 25C-NBOMe at 100 μM reduced cell viability in MTT assay. At 2.0 mg/kg and 0.75 mg/kg, they prolonged QT intervals in rat ECG. PAK1 was down-regulated by treatment with these two test compounds. Furthermore, potassium channels were inhibited by 25D-NBOMe treatment in hERG assay. Taken together, these results suggest that both 25D-NBOMe and 25C-NBOMe have potential cardiotoxicity, especially regarding cardiac rhythm. Further studies are needed to confirm the relationship between PAK1 down-regulation and cardiotoxicity.
Collapse
Affiliation(s)
- Kyung Sik Yoon
- National Institute of Drug and Safety Evaluation, Ministry of Food and Drug Safety, Osong, Cheongju, Republic of Korea.
| | - Jaesuk Yun
- College of Pharmacy, Chungbuk National University, Chungju, Republic of Korea.
| | - Young-Hoon Kim
- National Institute of Drug and Safety Evaluation, Ministry of Food and Drug Safety, Osong, Cheongju, Republic of Korea.
| | - Jisoon Shin
- National Institute of Drug and Safety Evaluation, Ministry of Food and Drug Safety, Osong, Cheongju, Republic of Korea.
| | - Sung Jin Kim
- Cosmetics Policy Division, Ministry of Food and Drug Safety, Osong, Cheongju, Republic of Korea.
| | - Jung-Wook Seo
- Research Group for Safety Pharmacology, Korea Institute of Toxicology, KRICT, Daejeon, Republic of Korea.
| | - Sung-Ae Hyun
- Research Group for Safety Pharmacology, Korea Institute of Toxicology, KRICT, Daejeon, Republic of Korea.
| | - Soo Kyung Suh
- National Institute of Drug and Safety Evaluation, Ministry of Food and Drug Safety, Osong, Cheongju, Republic of Korea.
| | - Hye Jin Cha
- National Institute of Drug and Safety Evaluation, Ministry of Food and Drug Safety, Osong, Cheongju, Republic of Korea.
| |
Collapse
|
27
|
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: 0.8] [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]
|
28
|
Mihanfar A, Nejabati HR, Fattahi A, Latifi Z, Pezeshkian M, Afrasiabi A, Safaie N, Jodati AR, Nouri M. The role of sphingosine 1 phosphate in coronary artery disease and ischemia reperfusion injury. J Cell Physiol 2018; 234:2083-2094. [PMID: 30341893 DOI: 10.1002/jcp.27353] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Accepted: 08/17/2018] [Indexed: 12/15/2022]
Abstract
Coronary artery disease (CAD) is a common cause of morbidity and mortality worldwide. Atherosclerotic plaques, as a hallmark of CAD, cause chronic narrowing of coronary arteries over time and could also result in acute myocardial infarction (AMI). The standard treatments for ameliorating AMI are reperfusion strategies, which paradoxically result in ischemic reperfusion (I/R) injury. Sphingosine 1 phosphate (S1P), as a potent lysophospholipid, plays an important role in various organs, including immune and cardiovascular systems. In addition, high-density lipoprotein, as a negative predictor of atherosclerosis and CAD, is a major carrier of S1P in blood circulation. S1P mediates its effects through binding to specific G protein-coupled receptors, and its signaling contributes to a variety of responses, including cardiac inflammation, dysfunction, and I/R injury protection. In this review, we will focus on the role of S1P in CAD and I/R injury as a potential therapeutic target.
Collapse
Affiliation(s)
- Aynaz Mihanfar
- Department of Biochemistry, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Hamid Reza Nejabati
- Stem Cell and Regenerative Medicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amir Fattahi
- Department of Reproductive Biology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Zeinab Latifi
- Stem Cell and Regenerative Medicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Masoud Pezeshkian
- Cardiovascular Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Abbas Afrasiabi
- Cardiovascular Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Naser Safaie
- Cardiovascular Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ahmad Reza Jodati
- Cardiovascular Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Nouri
- Stem Cell and Regenerative Medicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
29
|
Zhang N, Li X, Liu X, Cao Y, Chen D, Liu X, Wang Q, Du J, Weng J, Ma W. p21-activated kinase 1 activity is required for histone H3 Ser 10 phosphorylation and chromatin condensation in mouse oocyte meiosis. Reprod Fertil Dev 2018; 29:1287-1296. [PMID: 27166635 DOI: 10.1071/rd16026] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2016] [Accepted: 04/06/2016] [Indexed: 01/04/2023] Open
Abstract
p21-activated kinase 1 (Pak1) is essential for a variety of cellular events, including gene transcription, cytoskeletal organisation, cell proliferation and apoptosis. Pak1 is activated upon autophosphorylation on many amino residues; in particular, phosphorylation on Thr423 maintains maximal Pak1 activation. In the present study we investigated the protein expression, subcellular localisation and function of Pak1 phosphorylated on Thr423 (pPak1Thr423) in mouse oocytes. pPak1Thr423 was detected upon meiotic resumption and localised on the condensing chromatin. Thr423 phosphorylation was markedly suppressed by the Pak1 ATP-competitive inhibitor PF-3758309, but not by the allosteric inhibitors IPA-3 (2.5 μM and 10μM) (1, 1'-dithiobis-2-naphthalenol) and TAT-PAK18 (10 μM), which prevent the binding of Pak1 to its upstream activators GTPase Cdc42/Rac and Pak-interacting exchange factor (PIX), respectively, implying that Pak1 activation may be independent of GTPase and PIX in oocyte meiosis. Inhibition of Pak1 activation concomitantly restrained histone H3 phosphorylation on Ser10 and consequently inhibited chromatin condensation; however, this phenotype was reversed by concomitant administration of the Pak1 activator FTY720. The changes in the pattern of expression of phosphorylated extracellular signal-regulated kinase 1/2 in response to PF-3758309 or FTY720 were the same as seen for pPak1Thr423. These results show that activated Pak1 regulates chromatin condensation by promoting H3 Ser10 phosphorylation in oocytes after the resumption of meiotic progression.
Collapse
Affiliation(s)
- Nana Zhang
- Department of Histology and Embryology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Xiuhong Li
- Biospecimen and Clinical Data Repository, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China
| | - Xiaoyun Liu
- Department of Histology and Embryology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Yan Cao
- Department of Histology and Embryology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Dandan Chen
- Department of Histology and Embryology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Xiaoyu Liu
- Department of Histology and Embryology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Qian Wang
- Department of Histology and Embryology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Juan Du
- Department of Histology and Embryology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Jing Weng
- Experimental Center for Basic Medical Teaching, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Wei Ma
- Department of Histology and Embryology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| |
Collapse
|
30
|
Endothelial Cdc42 deficiency impairs endothelial regeneration and vascular repair after inflammatory vascular injury. Respir Res 2018; 19:27. [PMID: 29422044 PMCID: PMC5806471 DOI: 10.1186/s12931-018-0729-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Accepted: 01/26/2018] [Indexed: 01/11/2023] Open
Abstract
Background Endothelial cell (EC) regeneration is essential for inflammation resolution and vascular integrity recovery after inflammatory vascular injury. Cdc42 is a central regulator of cell survival and vessel formation in EC development. However, it is unknown that whether Cdc42 could be a regulating role of EC repair following the inflammatory injury in the lung. The study sought to test the hypothesis that Cdc42 is required for endothelial regeneration and vascular integrity recovery after LPS-induced inflammatory injury. Methods and results The role of Cdc42 for the regulation of pulmonary vascular endothelial repair was tested in vitro and in vivo. In LPS-induced acute lung injury (ALI) mouse models, knockout of the Cdc42 gene in ECs increased inflammatory cell infiltration and pulmonary vascular leakage and inhibited vascular EC proliferation, which eventually resulted in more severe inflammatory lung injury. In addition, siRNA-mediated knockdown of Cdc42 protein on ECs disrupted cell proliferation and migration and tube formation, which are necessary processes for recovery after inflammatory vascular injury, resulting in inflammatory vascular injury recovery defects. Conclusion We found that Cdc42 deficiency impairs EC function and regeneration, which are crucial in the post-inflammatory vascular injury repair process. These findings indicate that Cdc42 is a potential target for novel treatments designed to facilitate endothelial regeneration and vascular repair in inflammatory pulmonary vascular diseases, such as ALI/ARDS. Electronic supplementary material The online version of this article (10.1186/s12931-018-0729-8) contains supplementary material, which is available to authorized users.
Collapse
|
31
|
White C, Alshaker H, Cooper C, Winkler M, Pchejetski D. The emerging role of FTY720 (Fingolimod) in cancer treatment. Oncotarget 2018; 7:23106-27. [PMID: 27036015 PMCID: PMC5029614 DOI: 10.18632/oncotarget.7145] [Citation(s) in RCA: 116] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2015] [Accepted: 01/19/2016] [Indexed: 02/07/2023] Open
Abstract
FTY720 (Fingolimod) is a clinically approved immunomodulating therapy for multiple sclerosis that sequesters T-cells to lymph nodes through functional antagonism of sphingosine-1-phosphate 1 receptor. FTY720 also demonstrates a proven efficacy in multiple in vitro and in vivo cancer models, suggesting a potential therapeutic role in cancer patients. A potential anticancer mechanism of FTY720 is through the inhibition of sphingosine kinase 1, a proto-oncogene with in vitro and clinical cancer association. In addition, FTY720's anticancer properties may be attributable to actions on several other molecular targets. This study focuses on reviewing the emerging evidence regarding the anticancer properties and molecular targets of FTY720. While the clinical transition of FTY720 is currently limited by its immune suppression effects, studies aiming at FTY720 delivery and release together with identifying its key synergetic combinations and relevant patient subsets may lead to its rapid introduction into the clinic.
Collapse
Affiliation(s)
| | - Heba Alshaker
- Department of Pharmacology and Biomedical Sciences, Faculty of Pharmacy and Medical Sciences, University of Petra, Amman, Jordan.,School of Medicine, University of East Anglia, Norwich, UK
| | - Colin Cooper
- School of Medicine, University of East Anglia, Norwich, UK
| | - Matthias Winkler
- Department of Surgery and Cancer, Imperial College London, London, UK
| | | |
Collapse
|
32
|
Cannavo A, Liccardo D, Komici K, Corbi G, de Lucia C, Femminella GD, Elia A, Bencivenga L, Ferrara N, Koch WJ, Paolocci N, Rengo G. Sphingosine Kinases and Sphingosine 1-Phosphate Receptors: Signaling and Actions in the Cardiovascular System. Front Pharmacol 2017; 8:556. [PMID: 28878674 PMCID: PMC5572949 DOI: 10.3389/fphar.2017.00556] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Accepted: 08/07/2017] [Indexed: 12/20/2022] Open
Abstract
The sphingosine kinases 1 and 2 (SphK1 and 2) catalyze the phosphorylation of the lipid, sphingosine, generating the signal transmitter, sphingosine 1-phosphate (S1P). The activation of such kinases and the subsequent S1P generation and secretion in the blood serum of mammals represent a major checkpoint in many cellular signaling cascades. In fact, activating the SphK/S1P system is critical for cell motility and proliferation, cytoskeletal organization, cell growth, survival, and response to stress. In the cardiovascular system, the physiological effects of S1P intervene through the binding and activation of a family of five highly selective G protein-coupled receptors, called S1PR1-5. Importantly, SphK/S1P signal is present on both vascular and myocardial cells. S1P is a well-recognized survival factor in many tissues. Therefore, it is not surprising that the last two decades have seen a flourishing of interest and investigative efforts directed to obtain additional mechanistic insights into the signaling, as well as the biological activity of this phospholipid, and of its receptors, especially in the cardiovascular system. Here, we will provide an up-to-date account on the structure and function of sphingosine kinases, discussing the generation, release, and function of S1P. Keeping the bull's eye on the cardiovascular system, we will review the structure and signaling cascades and biological actions emanating from the stimulation of different S1P receptors. We will end this article with a summary of the most recent, experimental and clinical observations targeting S1PRs and SphKs as possible new therapeutic avenues for cardiovascular disorders, such as heart failure.
Collapse
Affiliation(s)
- Alessandro Cannavo
- Lewis Katz School of Medicine, Center for Translational Medicine, Temple University, PhiladelphiaPA, United States.,Department of Translational Medical Sciences, University of Naples Federico IINaples, Italy
| | - Daniela Liccardo
- Lewis Katz School of Medicine, Center for Translational Medicine, Temple University, PhiladelphiaPA, United States.,Department of Translational Medical Sciences, University of Naples Federico IINaples, Italy
| | - Klara Komici
- Department of Translational Medical Sciences, University of Naples Federico IINaples, Italy
| | - Graziamaria Corbi
- Department of Medicine and Health Science, University of MoliseCampobasso, Italy
| | - Claudio de Lucia
- Lewis Katz School of Medicine, Center for Translational Medicine, Temple University, PhiladelphiaPA, United States
| | | | - Andrea Elia
- Istituti Clinici Scientifici Maugeri SpA Società Benefit, Telese Terme Institute (BN)Telese, Italy
| | - Leonardo Bencivenga
- Department of Translational Medical Sciences, University of Naples Federico IINaples, Italy
| | - Nicola Ferrara
- Department of Translational Medical Sciences, University of Naples Federico IINaples, Italy.,Istituti Clinici Scientifici Maugeri SpA Società Benefit, Telese Terme Institute (BN)Telese, Italy
| | - Walter J Koch
- Lewis Katz School of Medicine, Center for Translational Medicine, Temple University, PhiladelphiaPA, United States
| | - Nazareno Paolocci
- Division of Cardiology, Johns Hopkins University Medical Institutions, BaltimoreMD, United States.,Department of Experimental Medicine, University of PerugiaPerugia, Italy
| | - Giuseppe Rengo
- Department of Translational Medical Sciences, University of Naples Federico IINaples, Italy.,Istituti Clinici Scientifici Maugeri SpA Società Benefit, Telese Terme Institute (BN)Telese, Italy
| |
Collapse
|
33
|
Wang Y, Wang S, Lei M, Boyett M, Tsui H, Liu W, Wang X. The p21-activated kinase 1 (Pak1) signalling pathway in cardiac disease: from mechanistic study to therapeutic exploration. Br J Pharmacol 2017; 175:1362-1374. [PMID: 28574147 DOI: 10.1111/bph.13872] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Revised: 05/12/2017] [Accepted: 05/17/2017] [Indexed: 01/01/2023] Open
Abstract
p21-activated kinase 1 (Pak1) is a member of the highly conserved family of serine/threonine protein kinases regulated by Ras-related small G-proteins, Cdc42/Rac1. It has been previously demonstrated to be involved in cardiac protection. Based on recent studies, this review provides an overview of the role of Pak1 in cardiac diseases including disrupted Ca2+ homoeostasis-related cardiac arrhythmias, adrenergic stress- and pressure overload-induced hypertrophy, and ischaemia/reperfusion injury. These findings demonstrate the important role of Pak1 mediated through the phosphorylation and transcriptional modification of hypertrophy and/or arrhythmia-related genes. This review also discusses the anti-arrhythmic and anti-hypertrophic, protective function of Pak1 and the beneficial effects of fingolimod (an FDA-approved sphingolipid drug), a Pak1 activator, and its ability to prevent arrhythmias and cardiac hypertrophy. These findings also highlight the therapeutic potential of Pak1 signalling in the treatment and prevention of cardiac diseases. LINKED ARTICLES This article is part of a themed section on Spotlight on Small Molecules in Cardiovascular Diseases. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v175.8/issuetoc.
Collapse
Affiliation(s)
- Yanwen Wang
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
| | - Shunyao Wang
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
| | - Ming Lei
- Department of Pharmacology, The University of Oxford, Oxford, UK
| | - Mark Boyett
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
| | - Hoyee Tsui
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
| | - Wei Liu
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
| | - Xin Wang
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
| |
Collapse
|
34
|
Marino A, Sakamoto T, Robador PA, Tomita K, Levi R. S1P receptor 1-Mediated Anti-Renin-Angiotensin System Cardioprotection: Pivotal Role of Mast Cell Aldehyde Dehydrogenase Type 2. J Pharmacol Exp Ther 2017; 362:230-242. [PMID: 28500264 DOI: 10.1124/jpet.117.241976] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Accepted: 05/09/2017] [Indexed: 12/13/2022] Open
Abstract
In the ischemic-reperfused (I/R) heart, renin-containing mast cells (MC) release enzymatically active renin, activating a local renin-angiotensin system (RAS), causing excessive norepinephrine release and arrhythmic dysfunction. Activation of Gi-receptors on MC and/or ischemic preconditioning (IPC) prevent renin release, thus providing anti-RAS cardioprotection. We questioned whether sphingosine-1-phosphate (S1P), a sphingolipid produced in the I/R heart, might afford anti-RAS cardioprotection by activating Gi-coupled S1P1 receptors (S1P1R) on MC. We report that activation of Gi-coupled S1P1R in cardiac MC confers IPC-like anti-RAS cardioprotection due to S1P1R-mediated inhibition of I/R-induced cardiac MC degranulation and renin release. This results from an initial translocation of protein kinase C subtype-ε and subsequent activation of aldehyde dehydrogenase type 2 (ALDH2), culminating in the elimination of the MC-degranulating effects of acetaldehyde and other toxic species produced during I/R. Inhibition of toxic aldehydes-induced MC-renin release prevents local RAS activation, reduces infarct size, and alleviates arrhythmias. Notably, these cardioprotective effects are lacking in hearts and MC from gene-targeted knock-in mice (ALDH2*2) in which ALDH2 enzymatic activity is maximally reduced. Thus, ALDH2 appears to play a pivotal role in this protective process. Our findings suggest that MC S1P1R may represent a new pharmacologic and therapeutic target for the direct alleviation of RAS-induced cardiac dysfunctions, including ischemic heart disease and congestive heart failure.
Collapse
Affiliation(s)
- Alice Marino
- Department of Pharmacology, Weill Cornell Medicine, New York, New York
| | - Takuya Sakamoto
- Department of Pharmacology, Weill Cornell Medicine, New York, New York
| | - Pablo A Robador
- Department of Pharmacology, Weill Cornell Medicine, New York, New York
| | - Kengo Tomita
- Department of Pharmacology, Weill Cornell Medicine, New York, New York
| | - Roberto Levi
- Department of Pharmacology, Weill Cornell Medicine, New York, New York
| |
Collapse
|
35
|
Peyrin-Biroulet L, Christopher R, Behan D, Lassen C. Modulation of sphingosine-1-phosphate in inflammatory bowel disease. Autoimmun Rev 2017; 16:495-503. [PMID: 28279838 DOI: 10.1016/j.autrev.2017.03.007] [Citation(s) in RCA: 98] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Accepted: 02/14/2017] [Indexed: 12/15/2022]
Abstract
Inflammatory bowel diseases (IBD), including ulcerative colitis and Crohn's disease, involve an inappropriate immune reaction in the digestive tract, causing a variety of disabling symptoms. The advent of monoclonal antibodies (anti-tumor necrosis factor, anti-integrin, anti-interleukin -23) has revolutionized IBD management. Nevertheless, these agents, with potential for immunogenicity, are associated with high rates of response loss and disease relapse over time. They are also associated with high production costs. Sphingosine-1-phosphate (S1P), a membrane-derived lysophospholipid signaling molecule, is implicated in a vast array of physiological and pathophysiological processes, primarily via extracellular activation of S1P1-S1P5 receptors. S1P1, S1P4 and S1P5 are involved in regulation of the immune system, while S1P2 and S1P3 may be associated with cardiovascular, pulmonary, and theoretical cancer-related risks. Targeting S1P receptors for inflammatory conditions has been successful in clinical trials leading to approval of the non-selective S1P modulator, fingolimod, for relapsing forms of multiple sclerosis. However, the association of this non-selective S1P modulator with serious adverse events provides the rationale for developing more selective S1P receptor modulators. Until recently, three S1P modulators with differing selectivity for S1P receptors were in clinical development for IBD: ozanimod (RPC1063), etrasimod (APD334) and amiselimod (MT-1303). The development of amiselimod has been stopped as Biogen are currently focusing on other drugs in its portfolio. Following encouraging results from the Phase 2 TOUCHSTONE trial, a Phase 3 trial of the S1P modulator ozanimod in patients with moderate-to-severe ulcerative colitis is ongoing. Etrasimod is also being tested in a phase 2 trial in ulcerative colitis. These pipeline medications can be administered orally and may avoid the formation of anti-drug antibodies that can lead to treatment failure with injectable biologic therapies for IBD. Data from ongoing clinical trials will establish the relationship between the selectivity of S1P modulators and their safety and efficacy in IBD, as well as their potential place in the clinical armamentarium for IBD.
Collapse
Affiliation(s)
- Laurent Peyrin-Biroulet
- Department of Gastroenterology and Inserm U954, Nancy University Hospital, Lorraine University, 54500 Vandoeuvre, France.
| | - Ronald Christopher
- Arena Pharmaceuticals, Inc., 6154 Nancy Ridge Drive, San Diego, CA 92121, USA
| | - Dominic Behan
- Arena Pharmaceuticals, Inc., 6154 Nancy Ridge Drive, San Diego, CA 92121, USA
| | - Cheryl Lassen
- Arena Pharmaceuticals GmbH, Untere Brühlstrasse 4, CH-4800 Zofingen, Switzerland
| |
Collapse
|
36
|
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: 0.9] [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
|
37
|
Yung BS, Brand CS, Xiang SY, Gray CBB, Means CK, Rosen H, Chun J, Purcell NH, Brown JH, Miyamoto S. Selective coupling of the S1P 3 receptor subtype to S1P-mediated RhoA activation and cardioprotection. J Mol Cell Cardiol 2017; 103:1-10. [PMID: 28017639 PMCID: PMC5410967 DOI: 10.1016/j.yjmcc.2016.12.008] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Revised: 12/16/2016] [Accepted: 12/19/2016] [Indexed: 01/17/2023]
Abstract
Sphingosine-1-phosphate (S1P), a bioactive lysophospholipid, is generated and released at sites of tissue injury in the heart and can act on S1P1, S1P2, and S1P3 receptor subtypes to affect cardiovascular responses. We established that S1P causes little phosphoinositide hydrolysis and does not induce hypertrophy indicating that it does not cause receptor coupling to Gq. We previously demonstrated that S1P confers cardioprotection against ischemia/reperfusion by activating RhoA and its downstream effector PKD. The S1P receptor subtypes and G proteins that regulate RhoA activation and downstream responses in the heart have not been determined. Using siRNA or pertussis toxin to inhibit different G proteins in NRVMs we established that S1P regulates RhoA activation through Gα13 but not Gα12, Gαq, or Gαi. Knockdown of the three major S1P receptors using siRNA demonstrated a requirement for S1P3 in RhoA activation and subsequent phosphorylation of PKD, and this was confirmed in studies using isolated hearts from S1P3 knockout (KO) mice. S1P treatment reduced infarct size induced by ischemia/reperfusion in Langendorff perfused wild-type (WT) hearts and this protection was abolished in the S1P3 KO mouse heart. CYM-51736, an S1P3-specific agonist, also decreased infarct size after ischemia/reperfusion to a degree similar to that achieved by S1P. The finding that S1P3 receptor- and Gα13-mediated RhoA activation is responsible for protection against ischemia/reperfusion suggests that selective targeting of S1P3 receptors could provide therapeutic benefits in ischemic heart disease.
Collapse
Affiliation(s)
- Bryan S Yung
- Department of Pharmacology, School of Medicine, University of California, San Diego, La Jolla, CA 92093, United States
| | - Cameron S Brand
- Department of Pharmacology, School of Medicine, University of California, San Diego, La Jolla, CA 92093, United States
| | - Sunny Y Xiang
- Department of Pharmacology, School of Medicine, University of California, San Diego, La Jolla, CA 92093, United States
| | - Charles B B Gray
- Department of Pharmacology, School of Medicine, University of California, San Diego, La Jolla, CA 92093, United States
| | | | - Hugh Rosen
- Department of Chemical Physiology, Scripps Research Institute, La Jolla, CA 92037, United States
| | - Jerold Chun
- Department of Molecular and Cellular Neuroscience, Dorris Neuroscience Center, Scripps Research Institute, La Jolla, CA 92037, United States
| | - Nicole H Purcell
- Department of Pharmacology, School of Medicine, University of California, San Diego, La Jolla, CA 92093, United States
| | - Joan Heller Brown
- Department of Pharmacology, School of Medicine, University of California, San Diego, La Jolla, CA 92093, United States.
| | - Shigeki Miyamoto
- Department of Pharmacology, School of Medicine, University of California, San Diego, La Jolla, CA 92093, United States.
| |
Collapse
|
38
|
Cannavo A, Liccardo D, Komici K, Corbi G, de Lucia C, Femminella GD, Elia A, Bencivenga L, Ferrara N, Koch WJ, Paolocci N, Rengo G. Sphingosine Kinases and Sphingosine 1-Phosphate Receptors: Signaling and Actions in the Cardiovascular System. Front Pharmacol 2017. [PMID: 28878674 DOI: 10.3389/fphar.2017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/01/2023] Open
Abstract
The sphingosine kinases 1 and 2 (SphK1 and 2) catalyze the phosphorylation of the lipid, sphingosine, generating the signal transmitter, sphingosine 1-phosphate (S1P). The activation of such kinases and the subsequent S1P generation and secretion in the blood serum of mammals represent a major checkpoint in many cellular signaling cascades. In fact, activating the SphK/S1P system is critical for cell motility and proliferation, cytoskeletal organization, cell growth, survival, and response to stress. In the cardiovascular system, the physiological effects of S1P intervene through the binding and activation of a family of five highly selective G protein-coupled receptors, called S1PR1-5. Importantly, SphK/S1P signal is present on both vascular and myocardial cells. S1P is a well-recognized survival factor in many tissues. Therefore, it is not surprising that the last two decades have seen a flourishing of interest and investigative efforts directed to obtain additional mechanistic insights into the signaling, as well as the biological activity of this phospholipid, and of its receptors, especially in the cardiovascular system. Here, we will provide an up-to-date account on the structure and function of sphingosine kinases, discussing the generation, release, and function of S1P. Keeping the bull's eye on the cardiovascular system, we will review the structure and signaling cascades and biological actions emanating from the stimulation of different S1P receptors. We will end this article with a summary of the most recent, experimental and clinical observations targeting S1PRs and SphKs as possible new therapeutic avenues for cardiovascular disorders, such as heart failure.
Collapse
Affiliation(s)
- Alessandro Cannavo
- Lewis Katz School of Medicine, Center for Translational Medicine, Temple University, PhiladelphiaPA, United States
- Department of Translational Medical Sciences, University of Naples Federico IINaples, Italy
| | - Daniela Liccardo
- Lewis Katz School of Medicine, Center for Translational Medicine, Temple University, PhiladelphiaPA, United States
- Department of Translational Medical Sciences, University of Naples Federico IINaples, Italy
| | - Klara Komici
- Department of Translational Medical Sciences, University of Naples Federico IINaples, Italy
| | - Graziamaria Corbi
- Department of Medicine and Health Science, University of MoliseCampobasso, Italy
| | - Claudio de Lucia
- Lewis Katz School of Medicine, Center for Translational Medicine, Temple University, PhiladelphiaPA, United States
| | | | - Andrea Elia
- Istituti Clinici Scientifici Maugeri SpA Società Benefit, Telese Terme Institute (BN)Telese, Italy
| | - Leonardo Bencivenga
- Department of Translational Medical Sciences, University of Naples Federico IINaples, Italy
| | - Nicola Ferrara
- Department of Translational Medical Sciences, University of Naples Federico IINaples, Italy
- Istituti Clinici Scientifici Maugeri SpA Società Benefit, Telese Terme Institute (BN)Telese, Italy
| | - Walter J Koch
- Lewis Katz School of Medicine, Center for Translational Medicine, Temple University, PhiladelphiaPA, United States
| | - Nazareno Paolocci
- Division of Cardiology, Johns Hopkins University Medical Institutions, BaltimoreMD, United States
- Department of Experimental Medicine, University of PerugiaPerugia, Italy
| | - Giuseppe Rengo
- Department of Translational Medical Sciences, University of Naples Federico IINaples, Italy
- Istituti Clinici Scientifici Maugeri SpA Società Benefit, Telese Terme Institute (BN)Telese, Italy
| |
Collapse
|
39
|
Lubbers ER, Mohler PJ. Roles and regulation of protein phosphatase 2A (PP2A) in the heart. J Mol Cell Cardiol 2016; 101:127-133. [PMID: 27832939 DOI: 10.1016/j.yjmcc.2016.11.003] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2016] [Revised: 10/28/2016] [Accepted: 11/03/2016] [Indexed: 01/21/2023]
Abstract
Reversible protein phosphorylation is central to a variety of cardiac processes including excitation-contraction coupling, Ca2+ handling, cell metabolism, myofilament regulation, and cell-cell communication. While kinase pathways linked with elevated adrenergic signaling have been a major focus for the cardiovascular field over the past half century, new findings support the critical role of protein phosphatases in both health and disease. Protein phosphatase 2A (PP2A) is a central cardiac phosphatase that regulates diverse myocyte functions through a host of target molecules. Notably, multiple mechanisms have evolved to dynamically tune PP2A function, including modulation of the composition, phosphorylation, methylation, and localization of PP2A holoenzyme populations. Further, aberrations in this regulation of PP2A function may contribute to cardiac pathophysiology. In summary, PP2A is a critical regulatory molecule in both health and disease, with a myriad of targets in heart. Based on their unique structure, localization, and regulatory properties, PP2A subunits represent exciting therapeutic targets to modulate altered adrenergic signaling in cardiovascular disease.
Collapse
Affiliation(s)
- Ellen R Lubbers
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, United States; Department of Physiology & Cell Biology, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| | - Peter J Mohler
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, United States; Department of Physiology & Cell Biology, The Ohio State University Wexner Medical Center, Columbus, OH, United States; Department of Internal Medicine, Division of Cardiovascular Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, United States.
| |
Collapse
|
40
|
Abdar M, Ebrahimifar P, Etemadifar M. The outbreak fingolimod cardiovascular side effects in relapsing-remitting multiple sclerosis patient: A longitudinal study in an Iranian population. ARYA ATHEROSCLEROSIS 2016; 12:274-280. [PMID: 28607567 PMCID: PMC5455326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
BACKGROUND Fingolimod (FTY-720) has shown efficacy in relapsing multiple sclerosis (MS), while some side effects of this drug have been recognized that the most important is cardiovascular side effects. The aim of this study was to evaluate the cardiovascular side effects of FTY-720. However, the effect of fingolimod on cardiac has not been well recognized. This study was designed to evaluate the cardiovascular side effects of fingolimod in relapsing-remitting multiple sclerosis (RRMS) patient in an Iranian population. METHODS This prospective clinical trial study was performed on 200 RRMS patients. The patients received a single daily oral dose of fingolimod 0.5 mg. During the first 6 hours after the first fingolimod dose, the patients' vital signs and electrocardiographic traces were continuously monitored. Moreover, the patients followed up over 6 months after receiving fingolimod. RESULTS The results showed that pulse rate (P < 0.001), systolic blood pressure (BP) (P < 0.001), and diastolic BP (P < 0.001) were decreased significantly during 6 hours after receiving the first dose of fingolimod. The most reduction in vital sign was observed in 3 hours. Arrhythmia, bradycardia, and dizziness were the other complications of fingolimod, which were detected in our study. CONCLUSION All the side effects such as hypotension and bradycardia were happened in first 3 hours after receiving the fingolimod. Indeed, we advise clinicians to monitor the patients for first 6 hours after initiation of fingolimod to decrease worse side effects.
Collapse
Affiliation(s)
- Morteza Abdar
- Professor, Department of Cardiology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Payam Ebrahimifar
- Cardiac Rehabilitation Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran,Correspondence to: Payam Ebrahimifar,
| | - Masoud Etemadifar
- Professor, Department of Neurology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
41
|
Linker RA, Wendt G. Cardiac Safety Profile of First Dose of Fingolimod for Relapsing-Remitting Multiple Sclerosis in Real-World Settings: Data from a German Prospective Multi-Center Observational Study. Neurol Ther 2016; 5:193-201. [PMID: 27624575 PMCID: PMC5130918 DOI: 10.1007/s40120-016-0051-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Indexed: 01/24/2023] Open
Abstract
Introduction Fingolimod was the first oral therapy approved for the treatment of relapsing-remitting multiple sclerosis (RRMS). Due to its action on cardiac sphingosine 1-phosphate receptors, fingolimod is leading to a transient decrease in heart rate (HR) and the occurrence of rare and asymptomatic self-limited atrioventricular (AV) blocks. This German non-interventional clinical study aimed to assess the cardiac safety profile in RRMS patients during at least 6 h after the initial treatment or restart after interruption of fingolimod in real-world settings. Methods The GoCARD study (German National Health Authorities, BfArM, CFTY720DDE18, NIS334) was a prospective, multi-center non-interventional study which was conducted in neurological and other medical practices or hospitals, qualified to routinely assess electrocardiogram (ECG) findings. Data were collected through interviews, clinical evaluations (notably ECGs), and laboratory tests. Medical history, vital signs, and a 12-lead ECG were assessed before fingolimod administration. After the first dose, a 6 h ECG was performed and vital signs (blood pressure and HR) were measured hourly. The occurrence of bradycardia (HR ≤45 beats per minute [BPM]), AV blocks (2nd degree Mobitz type I or higher), and corrected QT interval (QTc) intervals was also documented. Results More than 95% of physicians adhered to the cardiac monitoring recommendations. The observation of 217 patients in 42 study centers showed that while 35.9% of the patients had any cardiac risk profile, none of them experienced a bradycardia during the 6 h post-dose observation. Overall, only 1.8% of all patients displayed bradycardia (HR ≤45 BPM) during 6 h after treatment initiation. Moreover, in this cohort, none of the patients showed a new or persistent onset AV block (2nd degree Mobitz type I or higher) or QTc ≥500 ms. Conclusion Altogether, these data confirm that the first-dose observation after fingolimod initiation is usually uneventful (even in patients with pre-existing cardiovascular risk factors of this cohort) and that the rarely observed events remained asymptomatic and self-limited. Funding Novartis Pharma GmbH, Nürnberg, Germany.
Collapse
Affiliation(s)
- Ralf A Linker
- Department of Neurology, University Hospital Erlangen, Erlangen, Germany.
| | | |
Collapse
|
42
|
Terentyev D, Hamilton S. Regulation of sarcoplasmic reticulum Ca 2+ release by serine-threonine phosphatases in the heart. J Mol Cell Cardiol 2016; 101:156-164. [PMID: 27585747 DOI: 10.1016/j.yjmcc.2016.08.020] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Revised: 08/26/2016] [Accepted: 08/27/2016] [Indexed: 12/17/2022]
Abstract
The amount and timing of Ca2+ release from the sarcoplasmic reticulum (SR) during cardiac cycle are the main determinants of cardiac contractility. Reversible phosphorylation of the SR Ca2+ release channel, ryanodine receptor type 2 (RyR2) is the central mechanism of regulation of Ca2+ release in cardiomyocytes. Three major serine-threonine phosphatases including PP1, PP2A and PP2B (calcineurin) have been implicated in modulation of RyR2 function. Changes in expression levels of these phosphatases, their activity and targeting to the RyR2 macromolecular complex were demonstrated in many animal models of cardiac disease and humans and are implicated in cardiac arrhythmia and heart failure. Here we review evidence in support of regulation of RyR2-mediated SR Ca2+ release by serine-threonine phosphatases and the role and mechanisms of dysregulation of phosphatases in various disease states.
Collapse
Affiliation(s)
- Dmitry Terentyev
- The Warren Alpert Medical School of Brown University, Rhode Island Hospital, Department of Medicine, Cardiovascular Research Center, United States.
| | - Shanna Hamilton
- Cardiff University, School of Medicine, Wales Heart Research Institute, United Kingdom
| |
Collapse
|
43
|
Egom EEA, Bae JS, Capel R, Richards M, Ke Y, Pharithi RB, Maher V, Kruzliak P, Lei M. Effect of sphingosine-1-phosphate on L-type calcium current and Ca2+ transient in rat ventricular myocytes. Mol Cell Biochem 2016; 419:83-92. [DOI: 10.1007/s11010-016-2752-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Accepted: 06/21/2016] [Indexed: 01/05/2023]
|
44
|
Li N, Zhang F. Implication of sphingosin-1-phosphate in cardiovascular regulation. Front Biosci (Landmark Ed) 2016; 21:1296-313. [PMID: 27100508 DOI: 10.2741/4458] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Sphingosine-1-phosphate (S1P) is a bioactive sphingolipid metabolite generated by phosphorylation of sphingosine catalyzed by sphingosine kinase. S1P acts mainly through its high affinity G-protein-coupled receptors and participates in the regulation of multiple systems, including cardiovascular system. It has been shown that S1P signaling is involved in the regulation of cardiac chronotropy and inotropy and contributes to cardioprotection as well as cardiac remodeling; S1P signaling regulates vascular function, such as vascular tone and endothelial barrier, and possesses an anti-atherosclerotic effect; S1P signaling is also implicated in the regulation of blood pressure. Therefore, manipulation of S1P signaling may offer novel therapeutic approaches to cardiovascular diseases. As several S1P receptor modulators and sphingosine kinase inhibitors have been approved or under clinical trials for the treatment of other diseases, it may expedite the test and implementation of these S1P-based drugs in cardiovascular diseases.
Collapse
Affiliation(s)
- Ningjun Li
- Department of Pharmacology and Toxicology, Medical College of Virginia Campus, Virginia Commonwealth University, Richmond, Virginia, USA,
| | - Fan Zhang
- Department of Pharmacology and Toxicology, Medical College of Virginia Campus, Virginia Commonwealth University, Richmond, Virginia, USA
| |
Collapse
|
45
|
Peng X, He Q, Li G, Ma J, Zhong TP. Rac1-PAK2 pathway is essential for zebrafish heart regeneration. Biochem Biophys Res Commun 2016; 472:637-42. [PMID: 26966072 DOI: 10.1016/j.bbrc.2016.03.011] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2016] [Accepted: 03/05/2016] [Indexed: 01/08/2023]
Abstract
P-21 activated kinases, or PAKs, are serine-threonine kinases that play important roles in diverse heart functions include heart development, cardiovascular development and function in a range of models; however, the mechanisms by which PAKs mediate heart regeneration are unknown. Here, we demonstrate that PAK2 and PAK4 expression is induced in cardiomyocytes and vessels, respectively, following zebrafish heart injury. Inhibition of PAK2 and PAK4 using a specific small molecule inhibitor impedes cardiomyocyte proliferation/dedifferentiation and cardiovascular regeneration, respectively. Cdc42 is specifically expressed in the ventricle and may function upstream of PAK2 but not PAK4 under normal conditions and that cardiomyocyte proliferentation during heart regeneration relies on Rac1-mediated activation of Pak2. Our results indicate that PAKs play a key role in heart regeneration.
Collapse
Affiliation(s)
- Xiangwen Peng
- State Key Laboratory of Genetic Engineering, Department of Genetics, School of Life Sciences, Fudan University, Shanghai 201203, China
| | - Quanze He
- Center for Reproduction and Genetics, Suzhou Municipal Hospital, Jiangsu 215002, China
| | - Guobao Li
- State Key Laboratory of Genetic Engineering, Department of Genetics, School of Life Sciences, Fudan University, Shanghai 201203, China
| | - Jinmin Ma
- State Key Laboratory of Genetic Engineering, Department of Genetics, School of Life Sciences, Fudan University, Shanghai 201203, China
| | - Tao P Zhong
- State Key Laboratory of Genetic Engineering, Department of Genetics, School of Life Sciences, Fudan University, Shanghai 201203, China; Department of Medicine, Vanderbilt University School of Medicine, TN 37232, USA.
| |
Collapse
|
46
|
Santos-Gallego CG, Vahl TP, Goliasch G, Picatoste B, Arias T, Ishikawa K, Njerve IU, Sanz J, Narula J, Sengupta PP, Hajjar RJ, Fuster V, Badimon JJ. Sphingosine-1-Phosphate Receptor Agonist Fingolimod Increases Myocardial Salvage and Decreases Adverse Postinfarction Left Ventricular Remodeling in a Porcine Model of Ischemia/Reperfusion. Circulation 2016; 133:954-66. [PMID: 26826180 DOI: 10.1161/circulationaha.115.012427] [Citation(s) in RCA: 150] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2014] [Accepted: 01/08/2016] [Indexed: 01/01/2023]
Abstract
BACKGROUND Fingolimod, a sphingosine-1-phosphate receptor agonist, is used for the treatment of multiple sclerosis and exerts antiapoptotic properties. We hypothesized that sphingosine-1-phosphate receptor activation with fingolimod during acute myocardial infarction (MI) inhibits apoptosis, leading to increased myocardial salvage, reduced infarct size, and mitigated left ventricular (LV) remodeling in a porcine model of ischemia/reperfusion. METHODS AND RESULTS Ischemia/reperfusion was induced in pigs by balloon occlusion of the left anterior descending artery, followed by reperfusion. Animals randomly received fingolimod or saline (control). In short-term experiments, fingolimod treatment activated the cardioprotective reperfusion injury salvage kinase and survivor activating factor enhancement pathways in the infarct border zone 24 hours after MI, leading to decreased cardiomyocyte apoptosis and reduced myocardial oxidative stress. These effects were abolished by specific inhibitors of both pathways, demonstrating that fingolimod-induced cardioprotection was mediated by reperfusion injury salvage kinase and survivor activating factor enhancement pathways. In long-term experiments, fingolimod significantly improved myocardial salvage, reduced infarct size, and improved systolic LV function measured by cardiac magnetic resonance 1 week and 1 month after MI. Importantly, fingolimod mitigated the development of adverse post-MI LV remodeling 1 month after MI. Specifically, fingolimod treatment led to a significant reduction in LV mass, LV dilatation, and neurohormonal activation, and it preserved LV geometry. Furthermore, fingolimod decreased interstitial fibrosis, cardiomyocyte hypertrophy, and chronic activation of Akt and extracellular receptor kinase 1/2 in the remote noninfarcted myocardium. CONCLUSIONS Sphingosine-1-phosphate receptor activation with fingolimod during acute MI reduced infarct size via the reperfusion injury salvage kinase and survivor activating factor enhancement pathways, improved systolic LV function, and mitigated post-MI LV remodeling. Our data strongly support a cardioprotective role for sphingosine-1-phosphate receptor activation during MI.
Collapse
Affiliation(s)
- Carlos G Santos-Gallego
- From Icahn School of Medicine at Mount Sinai, Mount Sinai Heart, Zena and Michael A. Wiener Cardiovascular Institute, and Marie-Josée and Henry R. Kravis Center for Cardiovascular Health, New York, NY (C.G.S.-G., T.P.V., G.G., B.P., T.A., K.I., I.U.N., J.S., J.N., P.P.S., R.J.H., V.F., J.J.B.); Columbia University Medical Center, New York Presbyterian Hospital, NY (T.P.V.); Department of Cardiology, Medical University of Vienna, Austria (G.G.); Center for Clinical Heart Research, Department of Cardiology, Oslo University Hospital Ullevaal, Norway (I.U.N.); and Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain (T.A., V.F.).
| | - Torsten P Vahl
- From Icahn School of Medicine at Mount Sinai, Mount Sinai Heart, Zena and Michael A. Wiener Cardiovascular Institute, and Marie-Josée and Henry R. Kravis Center for Cardiovascular Health, New York, NY (C.G.S.-G., T.P.V., G.G., B.P., T.A., K.I., I.U.N., J.S., J.N., P.P.S., R.J.H., V.F., J.J.B.); Columbia University Medical Center, New York Presbyterian Hospital, NY (T.P.V.); Department of Cardiology, Medical University of Vienna, Austria (G.G.); Center for Clinical Heart Research, Department of Cardiology, Oslo University Hospital Ullevaal, Norway (I.U.N.); and Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain (T.A., V.F.)
| | - Georg Goliasch
- From Icahn School of Medicine at Mount Sinai, Mount Sinai Heart, Zena and Michael A. Wiener Cardiovascular Institute, and Marie-Josée and Henry R. Kravis Center for Cardiovascular Health, New York, NY (C.G.S.-G., T.P.V., G.G., B.P., T.A., K.I., I.U.N., J.S., J.N., P.P.S., R.J.H., V.F., J.J.B.); Columbia University Medical Center, New York Presbyterian Hospital, NY (T.P.V.); Department of Cardiology, Medical University of Vienna, Austria (G.G.); Center for Clinical Heart Research, Department of Cardiology, Oslo University Hospital Ullevaal, Norway (I.U.N.); and Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain (T.A., V.F.)
| | - Belen Picatoste
- From Icahn School of Medicine at Mount Sinai, Mount Sinai Heart, Zena and Michael A. Wiener Cardiovascular Institute, and Marie-Josée and Henry R. Kravis Center for Cardiovascular Health, New York, NY (C.G.S.-G., T.P.V., G.G., B.P., T.A., K.I., I.U.N., J.S., J.N., P.P.S., R.J.H., V.F., J.J.B.); Columbia University Medical Center, New York Presbyterian Hospital, NY (T.P.V.); Department of Cardiology, Medical University of Vienna, Austria (G.G.); Center for Clinical Heart Research, Department of Cardiology, Oslo University Hospital Ullevaal, Norway (I.U.N.); and Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain (T.A., V.F.)
| | - Teresa Arias
- From Icahn School of Medicine at Mount Sinai, Mount Sinai Heart, Zena and Michael A. Wiener Cardiovascular Institute, and Marie-Josée and Henry R. Kravis Center for Cardiovascular Health, New York, NY (C.G.S.-G., T.P.V., G.G., B.P., T.A., K.I., I.U.N., J.S., J.N., P.P.S., R.J.H., V.F., J.J.B.); Columbia University Medical Center, New York Presbyterian Hospital, NY (T.P.V.); Department of Cardiology, Medical University of Vienna, Austria (G.G.); Center for Clinical Heart Research, Department of Cardiology, Oslo University Hospital Ullevaal, Norway (I.U.N.); and Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain (T.A., V.F.)
| | - Kiyotake Ishikawa
- From Icahn School of Medicine at Mount Sinai, Mount Sinai Heart, Zena and Michael A. Wiener Cardiovascular Institute, and Marie-Josée and Henry R. Kravis Center for Cardiovascular Health, New York, NY (C.G.S.-G., T.P.V., G.G., B.P., T.A., K.I., I.U.N., J.S., J.N., P.P.S., R.J.H., V.F., J.J.B.); Columbia University Medical Center, New York Presbyterian Hospital, NY (T.P.V.); Department of Cardiology, Medical University of Vienna, Austria (G.G.); Center for Clinical Heart Research, Department of Cardiology, Oslo University Hospital Ullevaal, Norway (I.U.N.); and Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain (T.A., V.F.)
| | - Ida U Njerve
- From Icahn School of Medicine at Mount Sinai, Mount Sinai Heart, Zena and Michael A. Wiener Cardiovascular Institute, and Marie-Josée and Henry R. Kravis Center for Cardiovascular Health, New York, NY (C.G.S.-G., T.P.V., G.G., B.P., T.A., K.I., I.U.N., J.S., J.N., P.P.S., R.J.H., V.F., J.J.B.); Columbia University Medical Center, New York Presbyterian Hospital, NY (T.P.V.); Department of Cardiology, Medical University of Vienna, Austria (G.G.); Center for Clinical Heart Research, Department of Cardiology, Oslo University Hospital Ullevaal, Norway (I.U.N.); and Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain (T.A., V.F.)
| | - Javier Sanz
- From Icahn School of Medicine at Mount Sinai, Mount Sinai Heart, Zena and Michael A. Wiener Cardiovascular Institute, and Marie-Josée and Henry R. Kravis Center for Cardiovascular Health, New York, NY (C.G.S.-G., T.P.V., G.G., B.P., T.A., K.I., I.U.N., J.S., J.N., P.P.S., R.J.H., V.F., J.J.B.); Columbia University Medical Center, New York Presbyterian Hospital, NY (T.P.V.); Department of Cardiology, Medical University of Vienna, Austria (G.G.); Center for Clinical Heart Research, Department of Cardiology, Oslo University Hospital Ullevaal, Norway (I.U.N.); and Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain (T.A., V.F.)
| | - Jagat Narula
- From Icahn School of Medicine at Mount Sinai, Mount Sinai Heart, Zena and Michael A. Wiener Cardiovascular Institute, and Marie-Josée and Henry R. Kravis Center for Cardiovascular Health, New York, NY (C.G.S.-G., T.P.V., G.G., B.P., T.A., K.I., I.U.N., J.S., J.N., P.P.S., R.J.H., V.F., J.J.B.); Columbia University Medical Center, New York Presbyterian Hospital, NY (T.P.V.); Department of Cardiology, Medical University of Vienna, Austria (G.G.); Center for Clinical Heart Research, Department of Cardiology, Oslo University Hospital Ullevaal, Norway (I.U.N.); and Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain (T.A., V.F.)
| | - Partho P Sengupta
- From Icahn School of Medicine at Mount Sinai, Mount Sinai Heart, Zena and Michael A. Wiener Cardiovascular Institute, and Marie-Josée and Henry R. Kravis Center for Cardiovascular Health, New York, NY (C.G.S.-G., T.P.V., G.G., B.P., T.A., K.I., I.U.N., J.S., J.N., P.P.S., R.J.H., V.F., J.J.B.); Columbia University Medical Center, New York Presbyterian Hospital, NY (T.P.V.); Department of Cardiology, Medical University of Vienna, Austria (G.G.); Center for Clinical Heart Research, Department of Cardiology, Oslo University Hospital Ullevaal, Norway (I.U.N.); and Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain (T.A., V.F.)
| | - Roger J Hajjar
- From Icahn School of Medicine at Mount Sinai, Mount Sinai Heart, Zena and Michael A. Wiener Cardiovascular Institute, and Marie-Josée and Henry R. Kravis Center for Cardiovascular Health, New York, NY (C.G.S.-G., T.P.V., G.G., B.P., T.A., K.I., I.U.N., J.S., J.N., P.P.S., R.J.H., V.F., J.J.B.); Columbia University Medical Center, New York Presbyterian Hospital, NY (T.P.V.); Department of Cardiology, Medical University of Vienna, Austria (G.G.); Center for Clinical Heart Research, Department of Cardiology, Oslo University Hospital Ullevaal, Norway (I.U.N.); and Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain (T.A., V.F.)
| | - Valentin Fuster
- From Icahn School of Medicine at Mount Sinai, Mount Sinai Heart, Zena and Michael A. Wiener Cardiovascular Institute, and Marie-Josée and Henry R. Kravis Center for Cardiovascular Health, New York, NY (C.G.S.-G., T.P.V., G.G., B.P., T.A., K.I., I.U.N., J.S., J.N., P.P.S., R.J.H., V.F., J.J.B.); Columbia University Medical Center, New York Presbyterian Hospital, NY (T.P.V.); Department of Cardiology, Medical University of Vienna, Austria (G.G.); Center for Clinical Heart Research, Department of Cardiology, Oslo University Hospital Ullevaal, Norway (I.U.N.); and Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain (T.A., V.F.)
| | - Juan J Badimon
- From Icahn School of Medicine at Mount Sinai, Mount Sinai Heart, Zena and Michael A. Wiener Cardiovascular Institute, and Marie-Josée and Henry R. Kravis Center for Cardiovascular Health, New York, NY (C.G.S.-G., T.P.V., G.G., B.P., T.A., K.I., I.U.N., J.S., J.N., P.P.S., R.J.H., V.F., J.J.B.); Columbia University Medical Center, New York Presbyterian Hospital, NY (T.P.V.); Department of Cardiology, Medical University of Vienna, Austria (G.G.); Center for Clinical Heart Research, Department of Cardiology, Oslo University Hospital Ullevaal, Norway (I.U.N.); and Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain (T.A., V.F.)
| |
Collapse
|
47
|
Davis RT, Simon JN, Utter M, Mungai P, Alvarez MG, Chowdhury SAK, Heydemann A, Ke Y, Wolska BM, Solaro RJ. Knockout of p21-activated kinase-1 attenuates exercise-induced cardiac remodelling through altered calcineurin signalling. Cardiovasc Res 2015; 108:335-47. [PMID: 26464331 DOI: 10.1093/cvr/cvv234] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Accepted: 10/03/2015] [Indexed: 01/14/2023] Open
Abstract
AIMS Despite its known cardiovascular benefits, the intracellular signalling mechanisms underlying physiological cardiac growth remain poorly understood. Therefore, the purpose of this study was to investigate a novel role of p21-activated kinase-1 (Pak1) in the regulation of exercise-induced cardiac hypertrophy. METHODS AND RESULTS Wild-type (WT) and Pak1 KO mice were subjected to 6 weeks of treadmill endurance exercise training (ex-training). Cardiac function was assessed via echocardiography, in situ haemodynamics, and the pCa-force relations in skinned fibre preparations at baseline and at the end of the training regimen. Post-translational modifications to the sarcomeric proteins and expression levels of calcium-regulating proteins were also assessed following ex-training. Heart weight/tibia length and echocardiography data revealed that there was marked hypertrophy following ex-training in the WT mice, which was not evident in the KO mice. Additionally, following ex-training, WT mice demonstrated an increase in cardiac contractility, myofilament calcium sensitivity, and phosphorylation of cardiac myosin-binding protein C, cardiac TnT, and tropomyosin compared with KO mice. With ex-training in WT mice, there were also increased protein levels of calcineurin and increased phosphorylation of phospholamban. CONCLUSIONS Our data suggest that Pak1 is essential for adaptive physiological cardiac remodelling and support previous evidence that demonstrates Pak1 signalling is important for cardiac growth and survival.
Collapse
Affiliation(s)
- Robert T Davis
- Department of Physiology and Biophysics, Center for Cardiovascular Research, University of Illinois at Chicago, 835 S. Wolcott Ave-Rm. E202, Chicago, IL 60612, USA
| | - Jillian N Simon
- Department of Physiology and Biophysics, Center for Cardiovascular Research, University of Illinois at Chicago, 835 S. Wolcott Ave-Rm. E202, Chicago, IL 60612, USA
| | - Megan Utter
- Department of Physiology and Biophysics, Center for Cardiovascular Research, University of Illinois at Chicago, 835 S. Wolcott Ave-Rm. E202, Chicago, IL 60612, USA
| | - Paul Mungai
- Department of Physiology and Biophysics, Center for Cardiovascular Research, University of Illinois at Chicago, 835 S. Wolcott Ave-Rm. E202, Chicago, IL 60612, USA
| | - Manuel G Alvarez
- Department of Physiology and Biophysics, Center for Cardiovascular Research, University of Illinois at Chicago, 835 S. Wolcott Ave-Rm. E202, Chicago, IL 60612, USA
| | - Shamim A K Chowdhury
- Department of Physiology and Biophysics, Center for Cardiovascular Research, University of Illinois at Chicago, 835 S. Wolcott Ave-Rm. E202, Chicago, IL 60612, USA
| | - Ahlke Heydemann
- Department of Physiology and Biophysics, Center for Cardiovascular Research, University of Illinois at Chicago, 835 S. Wolcott Ave-Rm. E202, Chicago, IL 60612, USA
| | - Yunbo Ke
- Department of Physiology and Biophysics, Center for Cardiovascular Research, University of Illinois at Chicago, 835 S. Wolcott Ave-Rm. E202, Chicago, IL 60612, USA
| | - Beata M Wolska
- Department of Physiology and Biophysics, Center for Cardiovascular Research, University of Illinois at Chicago, 835 S. Wolcott Ave-Rm. E202, Chicago, IL 60612, USA Department of Medicine, Section of Cardiology, Center for Cardiovascular Research, University of Illinois, Chicago, IL 60612, USA
| | - R John Solaro
- Department of Physiology and Biophysics, Center for Cardiovascular Research, University of Illinois at Chicago, 835 S. Wolcott Ave-Rm. E202, Chicago, IL 60612, USA
| |
Collapse
|
48
|
Goltz D, Huss S, Ramadori E, Büttner R, Diehl L, Meyer R. Immunomodulation by splenectomy or by FTY720 protects the heart against ischemia reperfusion injury. Clin Exp Pharmacol Physiol 2015. [DOI: 10.1111/1440-1681.12465] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- D Goltz
- Institute of Pathology; University of Bonn; Bonn Germany
| | - S Huss
- Institute of Pathology; University of Cologne; Cologne Germany
| | - E Ramadori
- Institute of Pathology; University of Bonn; Bonn Germany
| | - R Büttner
- Institute of Pathology; University of Cologne; Cologne Germany
| | - L Diehl
- Experimental Immunology and Hepatology; Centre for Experimental Medicine; University Medical Centre Hamburg-Eppendorf; Hamburg Germany
| | - R Meyer
- Institute of Physiology II; University of Bonn; Bonn Germany
| |
Collapse
|
49
|
Xiao W, Xu GT, Zhang J, Zhang J, Zhang Y, Ye W. FTY720 ameliorates Dry Eye Disease in NOD mice: Involvement of leukocytes inhibition and goblet cells regeneration in ocular surface tissue. Exp Eye Res 2015; 138:145-52. [PMID: 26187517 DOI: 10.1016/j.exer.2015.06.032] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2015] [Revised: 06/06/2015] [Accepted: 06/24/2015] [Indexed: 10/23/2022]
Abstract
UNLABELLED FTY720 is a promising drug in attenuating multiple sclerosis, prolonging survival of organ allograft, and many other protective effects. Its mechanism of action is considered to be mediated by the internalization of sphingosine 1-phosphate receptors (S1PRs). In the current study, we investigated the efficacy of FTY720 in Non-Obese Diabetic (NOD) mice, serving as a model of Dry Eye Disease (DED). NOD mice were divided into six study groups, i.e., FTY720-treated groups with 3 concentrations of FTY720 (0.05%, 0.005%, and 0.001%), 0.05% Cyclosporin A (CsA) treated group, normal saline treated group, and no treatment control group. FTY720 was reconstituted with normal saline and prepared as eye drop. The stability and production of tear film was measured by Tear Break up Time test (TBUT) and phenol red cotton thread test (PRCTT), respectively. Tear fluid washings were collected and assessed by ELISA. Cytokines were detected in lacrimal glands by RT-PCR. Inflammation in conjunctiva was assessed by immunohistochemistry, goblet cells and conjunctival epithelia were examined and evaluated by impression cytology. Our results indicated that FTY720 had a significantly therapeutic effect in NOD mice. After FTY720 intervention, TBUT and PRCTT data were greatly improved (p < 0.01), the interleukin 1β (IL-1β) level was markedly decreased in tear fluid washings compared to control and normal saline groups after 2 weeks ( CONTROL 1.06 ± 0.12, Normal saline:0.97 ± 0.09 pg/ml, CsA:0.22 ± 0.02 pg/ml, 0.001% FTY720:0.23 ± 0.02 pg/ml, 0.005% FTY720:0.14 ± 0.03 pg/ml, 0.05% FTY720: 0.18 ± 0.03 pg/ml. CsA group and 3 FTY720 groups VS. control group and normal saline groups: p < 0.01). Proinflammatory factors were greatly decreased in lacrimal glands (p < 0.01). Leukocytes were identified and markedly decreased in conujnctiva (p < 0.01), inflammatory reaction of DED was greatly relieved. More importantly, the goblet cells were largely restored and ocular surface lesions were significantly ameliorated (p < 0.01). Thus, we observed FTY720 alleviated DED in NOD mice by inhibiting leukocytes, the function of ocular surface tissue in NOD mice was partially restored via inhibiting ocular surface inflammation and increasing the density of goblet cells and conjunctival epithelia. FTY720 may offer a novel strategy for the treatment of inflammatory disorders in the ocular surface.
Collapse
Affiliation(s)
- Weibao Xiao
- Department of Ophthalmology, Huashan Hospital Affiliated to Fudan University, 12 Middle Wulumuqi Road, Buiding 3, Room 802, Shanghai 200040, China
| | - Guo-Tong Xu
- Tongji Eye Institute and Department of Regenerative Medicine, Tongji University School of Medicine, 1239 Siping Road, Medical School Building, Room 521, Shanghai 200092, China
| | - Jingfa Zhang
- Tongji Eye Institute and Department of Regenerative Medicine, Tongji University School of Medicine, 1239 Siping Road, Medical School Building, Room 521, Shanghai 200092, China
| | - Jiaying Zhang
- Department of Ophthalmology, Huashan Hospital Affiliated to Fudan University, 12 Middle Wulumuqi Road, Buiding 3, Room 802, Shanghai 200040, China
| | - Yu Zhang
- Department of Ophthalmology, Huashan Hospital Affiliated to Fudan University, 12 Middle Wulumuqi Road, Buiding 3, Room 802, Shanghai 200040, China
| | - Wen Ye
- Department of Ophthalmology, Huashan Hospital Affiliated to Fudan University, 12 Middle Wulumuqi Road, Buiding 3, Room 802, Shanghai 200040, China.
| |
Collapse
|
50
|
Egom EE, Kruzliak P, Rotrekl V, Lei M. The effect of the sphingosine-1-phosphate analogue FTY720 on atrioventricular nodal tissue. J Cell Mol Med 2015; 19:1729-34. [PMID: 25864579 PMCID: PMC4511369 DOI: 10.1111/jcmm.12549] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2014] [Accepted: 01/08/2015] [Indexed: 12/12/2022] Open
Abstract
The sphingosine‐1‐phosphate (S1P) receptor modulator, fingolimod (FTY720), has been used for the treatment of patients with relapsing forms of multiple sclerosis, but atrioventricular (AV) conduction block have been reported in some patients after the first dose. The underlying mechanism of this AV node conduction blockade is still not well‐understood. In this study, we hypothesize that expression of this particular arrhythmia might be related to a direct effect of FTY720 on AV node rather than a parasympathetic mimetic action. We, therefore, investigated the effect of FTY720 on AV nodal, using in vitro rat model preparation, under both basal as well as ischaemia/reperfusion conditions. We first look at the expression pattern of S1P receptors on the AV node using real‐time PCR. Although all three S1P receptor isoforms were expressed in AVN tissues, S1P1 receptor isoform expression level was higher than S1P2 and S1P3. The effect of 25 nM FTY720 on cycle length (CL) was subsequently studied via extracellular potentials recordings. FTY720 caused a mild to moderate prolongation in CL by an average 9% in AVN (n = 10, P < 0.05) preparations. We also show that FTY720 attenuated both ischaemia and reperfusion induced AVN rhythmic disturbance. To our knowledge, these remarkable findings have not been previously reported in the literature, and stress the importance for extensive monitoring period in certain cases, especially in patients taking concurrently AV node blocker agents.
Collapse
Affiliation(s)
- Emmanuel E Egom
- Egom Clinical and Translational Research Services, Halifax, NS, Canada
| | - Peter Kruzliak
- Department of Cardiovascular Diseases, International Clinical Research Center, St. Anne's University Hospital and Masaryk University, Brno, Czech Republic
| | - Vladimir Rotrekl
- Department of Cardiovascular Diseases, International Clinical Research Center, St. Anne's University Hospital and Masaryk University, Brno, Czech Republic.,Department of Medical Physics and Biophysics, Faculty of Medicine, Pavol Jozef Safarik University, Kosice, Slovak Republic
| | - Ming Lei
- Department of Biology, Faculty of Medicine, Masaryk University, Brno, Czech Republic.,Department of Pharmacology, University of Oxford, Oxford & Institute of Cardiovascular Sciences, UK
| |
Collapse
|