1
|
Cour M, Pedretti S, Nduhirabandi F, Hacking D, Frias MA, Hausenloy DJ, Lecour S. Interplay between the SAFE and the sphingolipid pathway for cardioprotection. Life Sci 2024; 358:123145. [PMID: 39401691 DOI: 10.1016/j.lfs.2024.123145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2024] [Revised: 10/07/2024] [Accepted: 10/10/2024] [Indexed: 10/19/2024]
Abstract
AIM Activation of both the Survivor Activating Factor Enhancement (SAFE) pathway (including Tumor Necrosis Factor-alpha (TNF-α) and Signal Transducer and Activator of Transcription-3 (STAT-3)) and the sphingolipid signalling pathway (including sphingosine kinase-1 (SK1) and sphingosine-1 phosphate (S1P)) play a key role in promoting cardioprotection against ischemia-reperfusion injury (IRI). We investigated whether the activation of the SAFE pathway by exogenous S1P is dependent on the activation of SK1 for cardioprotection. MATERIALS AND METHODS Isolated cardiomyocytes from TNF-α knockout (KO) mice, cardiomyocyte-specific STAT-3 KO mice and their wild-type (WT) littermates were exposed to simulated ischemia in the presence of a trigger of the SAFE pathway (S1P) and SK1 inhibitor (SK1-I). Similarly, isolated perfused hearts from adult TNF-α KO, STAT-3 KO and WT mice were subjected to IRI with S1P and/or SK1-I. Cell viability, infarct size (IS) and SK1 activity were assessed. KEY FINDINGS In isolated cardiomyocytes and in isolated hearts subjected to simulated ischemia/IRI, S1P pretreatment decreased cell death in WT mice, an effect that was abrogated in the presence of SK1-I. S1P failed to reduce cell death after simulated ischemia/IRI in both cardiomyocytes or hearts isolated from TNF-α KO and STAT-3 KO mice. Interestingly, S1P pretreatment increased SK1 activity in WT and STAT-3 KO mice, with no changes in TNF-α KO mice. SIGNIFICANCE Our data strongly suggest SK1 as a key component to activate STAT-3 downstream of TNF-α in the SAFE pathway, paving the way for the development of novel cardioprotective strategies that may target SK1 to modulate the SAFE pathway and increase cell survival following IRI.
Collapse
Affiliation(s)
- Martin Cour
- Hatter Institute/Cape Heart Institute, Department of Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Sarah Pedretti
- Hatter Institute/Cape Heart Institute, Department of Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Frederic Nduhirabandi
- Hatter Institute/Cape Heart Institute, Department of Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Damian Hacking
- Hatter Institute/Cape Heart Institute, Department of Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Miguel A Frias
- Division of Laboratory Medicine, Diagnostic Department, Geneva University Hospitals, Geneva, Switzerland
| | - Derek J Hausenloy
- The Hatter Cardiovascular Institute, University College London, London, UK
| | - Sandrine Lecour
- Hatter Institute/Cape Heart Institute, Department of Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa.
| |
Collapse
|
2
|
Nikiforova AB, Baburina YL, Borisova MP, Surin AK, Kharechkina ES, Krestinina OV, Suvorina MY, Kruglova SA, Kruglov AG. Mitochondrial F-ATP Synthase Co-Migrating Proteins and Ca 2+-Dependent Formation of Large Channels. Cells 2023; 12:2414. [PMID: 37830628 PMCID: PMC10572550 DOI: 10.3390/cells12192414] [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] [Received: 06/30/2023] [Revised: 09/18/2023] [Accepted: 10/02/2023] [Indexed: 10/14/2023] Open
Abstract
Monomers, dimers, and individual FOF1-ATP synthase subunits are, presumably, involved in the formation of the mitochondrial permeability transition pore (PTP), whose molecular structure, however, is still unknown. We hypothesized that, during the Ca2+-dependent assembly of a PTP complex, the F-ATP synthase (subunits) recruits mitochondrial proteins that do not interact or weakly interact with the F-ATP synthase under normal conditions. Therefore, we examined whether the PTP opening in mitochondria before the separation of supercomplexes via BN-PAGE will increase the channel stability and channel-forming capacity of isolated F-ATP synthase dimers and monomers in planar lipid membranes. Additionally, we studied the specific activity and the protein composition of F-ATP synthase dimers and monomers from rat liver and heart mitochondria before and after PTP opening. Against our expectations, preliminary PTP opening dramatically suppressed the high-conductance channel activity of F-ATP synthase dimers and monomers and decreased their specific "in-gel" activity. The decline in the channel-forming activity correlated with the reduced levels of as few as two proteins in the bands: methylmalonate-semialdehyde dehydrogenase and prohibitin 2. These results indicate that proteins co-migrating with the F-ATP synthase may be important players in PTP formation and stabilization.
Collapse
Affiliation(s)
- Anna B. Nikiforova
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Institutskaya 3, 142290 Pushchino, Russia; (A.B.N.); (Y.L.B.); (M.P.B.); (E.S.K.); (O.V.K.)
| | - Yulia L. Baburina
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Institutskaya 3, 142290 Pushchino, Russia; (A.B.N.); (Y.L.B.); (M.P.B.); (E.S.K.); (O.V.K.)
| | - Marina P. Borisova
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Institutskaya 3, 142290 Pushchino, Russia; (A.B.N.); (Y.L.B.); (M.P.B.); (E.S.K.); (O.V.K.)
| | - Alexey K. Surin
- Branch of the Shemyakin—Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Prospekt Nauki 6, 142290 Pushchino, Russia;
- State Research Centre for Applied Microbiology and Biotechnology, 142279 Obolensk, Russia
- Institute of Protein Research, Russian Academy of Sciences, Institutskaya 4, 142290 Pushchino, Russia;
| | - Ekaterina S. Kharechkina
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Institutskaya 3, 142290 Pushchino, Russia; (A.B.N.); (Y.L.B.); (M.P.B.); (E.S.K.); (O.V.K.)
| | - Olga V. Krestinina
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Institutskaya 3, 142290 Pushchino, Russia; (A.B.N.); (Y.L.B.); (M.P.B.); (E.S.K.); (O.V.K.)
| | - Maria Y. Suvorina
- Institute of Protein Research, Russian Academy of Sciences, Institutskaya 4, 142290 Pushchino, Russia;
| | - Svetlana A. Kruglova
- Institute of Basic Biological Problems, Russian Academy of Sciences, Institutskaya 2, 142290 Pushchino, Russia;
| | - Alexey G. Kruglov
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Institutskaya 3, 142290 Pushchino, Russia; (A.B.N.); (Y.L.B.); (M.P.B.); (E.S.K.); (O.V.K.)
| |
Collapse
|
3
|
Chen Q, Kovilakath A, Allegood J, Thompson J, Hu Y, Cowart LA, Lesnefsky EJ. Endoplasmic reticulum stress and mitochondrial dysfunction during aging: Role of sphingolipids. Biochim Biophys Acta Mol Cell Biol Lipids 2023; 1868:159366. [PMID: 37473835 PMCID: PMC11154090 DOI: 10.1016/j.bbalip.2023.159366] [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] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Revised: 06/24/2023] [Accepted: 07/11/2023] [Indexed: 07/22/2023]
Abstract
The endoplasmic reticulum (ER) plays a key role in the regulation of protein folding, lipid synthesis, calcium homeostasis, and serves as a primary site of sphingolipid biosynthesis. ER stress (ER dysfunction) participates in the development of mitochondrial dysfunction during aging. Mitochondria are in close contact with the ER through shared mitochondria associated membranes (MAM). Alteration of sphingolipids contributes to mitochondria-driven cell injury. Cardiolipin is a phospholipid that is critical to maintain enzyme activity in the electron transport chain. The aim of the current study was to characterize the changes in sphingolipids and cardiolipin in ER, MAM, and mitochondria during the progression of aging in young (3 mo.), middle (18 mo.), and aged (24 mo.) C57Bl/6 mouse hearts. ER stress increased in hearts from 18 mo. mice and mice exhibited mitochondrial dysfunction by 24 mo. Hearts were pooled to isolate ER, MAM, and subsarcolemmal mitochondria (SSM). LC-MS/MS quantification of lipid content showed that aging increased ceramide content in ER and MAM. In addition, the contents of sphingomyelin and monohexosylceramides are also increased in the ER from aged mice. Aging increased the total cardiolipin content in the ER. Aging did not alter the total cardiolipin content in mitochondria or MAM yet altered the composition of cardiolipin with aging in line with increased oxidative stress compared to young mice. These results indicate that alteration of sphingolipids can contribute to the ER stress and mitochondrial dysfunction that occurs during aging.
Collapse
Affiliation(s)
- Qun Chen
- Department of Medicine (Division of Cardiology), Virginia Commonwealth University, Richmond, VA 23298, United States of America
| | - Anna Kovilakath
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, VA 23298, United States of America
| | - Jeremy Allegood
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, VA 23298, United States of America
| | - Jeremy Thompson
- Department of Medicine (Division of Cardiology), Virginia Commonwealth University, Richmond, VA 23298, United States of America
| | - Ying Hu
- Department of Medicine (Division of Cardiology), Virginia Commonwealth University, Richmond, VA 23298, United States of America
| | - L Ashley Cowart
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, VA 23298, United States of America; Richmond Department of Veterans Affairs Medical Center, Richmond, VA 23249, United States of America
| | - Edward J Lesnefsky
- Department of Medicine (Division of Cardiology), Virginia Commonwealth University, Richmond, VA 23298, United States of America; Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, VA 23298, United States of America; Department of Physiology and Biophysics, Virginia Commonwealth University, Richmond, VA 23298, United States of America; Richmond Department of Veterans Affairs Medical Center, Richmond, VA 23249, United States of America.
| |
Collapse
|
4
|
Loss of sphingosine kinase 2 promotes the expansion of hematopoietic stem cells by improving their metabolic fitness. Blood 2022; 140:1686-1701. [PMID: 35881840 DOI: 10.1182/blood.2022016112] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 07/20/2022] [Indexed: 11/20/2022] Open
Abstract
Hematopoietic stem cells (HSCs) have reduced capacities to properly maintain and replenish the hematopoietic system during myelosuppressive injury or aging. Expanding and rejuvenating HSCs for therapeutic purposes has been a long-sought goal with limited progress. Here, we show that the enzyme Sphk2 (sphingosine kinase 2), which generates the lipid metabolite sphingosine-1-phosphate, is highly expressed in HSCs. The deletion of Sphk2 markedly promotes self-renewal and increases the regenerative potential of HSCs. More importantly, Sphk2 deletion globally preserves the young HSC gene expression pattern, improves the function, and sustains the multilineage potential of HSCs during aging. Mechanistically, Sphk2 interacts with prolyl hydroxylase 2 and the Von Hippel-Lindau protein to facilitate HIF1α ubiquitination in the nucleus independent of the Sphk2 catalytic activity. Deletion of Sphk2 increases hypoxic responses by stabilizing the HIF1α protein to upregulate PDK3, a glycolysis checkpoint protein for HSC quiescence, which subsequently enhances the function of HSCs by improving their metabolic fitness; specifically, it enhances anaerobic glycolysis but suppresses mitochondrial oxidative phosphorylation and generation of reactive oxygen species. Overall, targeting Sphk2 to enhance the metabolic fitness of HSCs is a promising strategy to expand and rejuvenate functional HSCs.
Collapse
|
5
|
Duan M, Gao P, Chen SX, Novák P, Yin K, Zhu X. Sphingosine-1-phosphate in mitochondrial function and metabolic diseases. Obes Rev 2022; 23:e13426. [PMID: 35122459 DOI: 10.1111/obr.13426] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 01/02/2022] [Accepted: 01/02/2022] [Indexed: 01/23/2023]
Abstract
Sphingosine-1-phosphate (S1P) is a bioactive sphingolipid metabolite. The past decade has witnessed exponential growth in the field of S1P research, partly attributed to drugs targeting its receptors or kinases. Accumulating evidence indicates that changes in the S1P axis (i.e., S1P production, transport, and receptors) may modify metabolism and eventually mediate metabolic diseases. Dysfunction of the mitochondria on a master monitor of cellular metabolism is considered the leading cause of metabolic diseases, with aberrations typically induced by abnormal biogenesis, respiratory chain complex disorders, reactive oxygen species overproduction, calcium deposition, and mitophagy impairment. Accordingly, we discuss decades of investigation into changes in the S1P axis and how it controls mitochondrial function. Furthermore, we summarize recent scientific advances in disorders associated with the S1P axis and their involvement in the pathogenesis of metabolic diseases in humans, including type 2 diabetes mellitus and cardiovascular disease, from the perspective of mitochondrial function. Finally, we review potential challenges and prospects for S1P axis application to the regulation of mitochondrial function and metabolic diseases; these data may provide theoretical guidance for the treatment of metabolic diseases.
Collapse
Affiliation(s)
- Meng Duan
- Guangxi Key Laboratory of Diabetic Systems Medicine, Guilin Medical University, Guilin, Guangxi, China
| | - Pan Gao
- Guangxi Key Laboratory of Diabetic Systems Medicine, Guilin Medical University, Guilin, Guangxi, China
| | - Sheng-Xi Chen
- Guangxi Key Laboratory of Diabetic Systems Medicine, Guilin Medical University, Guilin, Guangxi, China
| | - Petr Novák
- Guangxi Key Laboratory of Diabetic Systems Medicine, Guilin Medical University, Guilin, Guangxi, China
| | - Kai Yin
- Guangxi Key Laboratory of Diabetic Systems Medicine, Guilin Medical University, Guilin, Guangxi, China.,Department of Cardiology, The Second Affiliated Hospital of Guilin Medical University, Guilin, Guangxi, China
| | - Xiao Zhu
- Guangxi Key Laboratory of Diabetic Systems Medicine, Guilin Medical University, Guilin, Guangxi, China
| |
Collapse
|
6
|
Chung LH, Liu D, Liu XT, Qi Y. Ceramide Transfer Protein (CERT): An Overlooked Molecular Player in Cancer. Int J Mol Sci 2021; 22:13184. [PMID: 34947980 PMCID: PMC8705978 DOI: 10.3390/ijms222413184] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 12/02/2021] [Accepted: 12/05/2021] [Indexed: 12/26/2022] Open
Abstract
Sphingolipids are a class of essential lipids implicated in constructing cellular membranes and regulating nearly all cellular functions. Sphingolipid metabolic network is centered with the ceramide-sphingomyelin axis. Ceramide is well-recognized as a pro-apoptotic signal; while sphingomyelin, as the most abundant type of sphingolipids, is required for cell growth. Therefore, the balance between these two sphingolipids can be critical for cancer cell survival and functioning. Ceramide transfer protein (CERT) dictates the ratio of ceramide to sphingomyelin within the cell. It is the only lipid transfer protein that specifically delivers ceramide from the endoplasmic reticulum to the Golgi apparatus, where ceramide serves as the substrate for sphingomyelin synthesis. In the past two decades, an increasing body of evidence has suggested a critical role of CERT in cancer, but much more intensive efforts are required to draw a definite conclusion. Herein, we review all research findings of CERT, focusing on its molecular structure, cellular functions and implications in cancer. This comprehensive review of CERT will help to better understand the molecular mechanism of cancer and inspire to identify novel druggable targets.
Collapse
Affiliation(s)
- Long Hoa Chung
- Centenary Institute of Cancer Medicine and Cell Biology, University of Sydney, Camperdown, NSW 2050, Australia; (D.L.); (X.T.L.)
| | | | | | - Yanfei Qi
- Centenary Institute of Cancer Medicine and Cell Biology, University of Sydney, Camperdown, NSW 2050, Australia; (D.L.); (X.T.L.)
| |
Collapse
|
7
|
Gaggini M, Pingitore A, Vassalle C. Plasma Ceramides Pathophysiology, Measurements, Challenges, and Opportunities. Metabolites 2021; 11:metabo11110719. [PMID: 34822377 PMCID: PMC8622894 DOI: 10.3390/metabo11110719] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 10/15/2021] [Accepted: 10/19/2021] [Indexed: 01/22/2023] Open
Abstract
Ceramides are a family of lipid molecules, composed of sphingosine and a fatty acid, and transported by lipoproteins (primarily by low-density lipoproteins) in the bloodstream. They are not only structural lipids, but multifunctional and bioactive molecules with key roles in many important cellular pathways, such as inflammatory processes and apoptosis, representing potential biomarkers of cardiometabolic diseases as well as pharmacological targets. Recent data reported ceramide modulation by diet and aerobic exercise, suggesting nutrients and exercise-targeting sphingolipid pathways as a countermeasure, also in combination with other therapies, for risk and progression of chronic disease prevention and health maintenance. In this review, we focus on the available data regarding remarks on ceramide structure and metabolism, their pathophysiologic roles, and the effect of dietary habit and aerobic exercise on ceramide levels. Moreover, advancements and limitations of lipidomic techniques and simplification attempts to overcome difficulties of interpretation and to facilitate practical applications, such as the proposal of scores, are also discussed.
Collapse
Affiliation(s)
- Melania Gaggini
- Institute of Clinical Physiology, National Research Council, 56124 Pisa, Italy; (M.G.); (A.P.)
| | - Alessandro Pingitore
- Institute of Clinical Physiology, National Research Council, 56124 Pisa, Italy; (M.G.); (A.P.)
| | - Cristina Vassalle
- Fondazione CNR-Regione Toscana G. Monasterio, Via Moruzzi, 1, 56124 Pisa, Italy
- Correspondence: ; Tel.: +39-050-3153525
| |
Collapse
|
8
|
Cui M, Göbel V, Zhang H. Uncovering the 'sphinx' of sphingosine 1-phosphate signalling: from cellular events to organ morphogenesis. Biol Rev Camb Philos Soc 2021; 97:251-272. [PMID: 34585505 PMCID: PMC9292677 DOI: 10.1111/brv.12798] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 09/11/2021] [Accepted: 09/16/2021] [Indexed: 11/02/2022]
Abstract
Sphingosine 1-phosphate (S1P) is a bioactive sphingolipid metabolite, functioning as a signalling molecule in diverse cellular processes. Over the past few decades, studies of S1P signalling have revealed that the physiological activity of S1P largely depends on S1P metabolizing enzymes, transporters and receptors on the plasma membrane, as well as on the intracellular proteins that S1P binds directly to. In addition to its roles in cancer signalling, immunity and inflammation, a large body of evidence has identified a close link of S1P signalling with organ morphogenesis. Here we discuss the vital role of S1P signalling in orchestrating various cellular events during organ morphogenesis through analysing each component along the extracellular and intracellular S1P signalling axes. For each component, we review advances in our understanding of S1P signalling and function from the upstream regulators to the downstream effectors and from cellular behaviours to tissue organization, primarily in the context of morphogenetic mechanisms. S1P-mediated vesicular trafficking is also discussed as a function independent of its signalling function. A picture emerges that reveals a multifaceted role of S1P-dependent pathways in the development and maintenance of organ structure and function.
Collapse
Affiliation(s)
- Mengqiao Cui
- Centre of Reproduction, Development and Aging, Faculty of Health Sciences, University of Macau, Taipa, Macau SAR, 999078, China
| | - Verena Göbel
- Mucosal Immunology and Biology Research Center, Department of Pediatrics, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, U.S.A
| | - Hongjie Zhang
- Centre of Reproduction, Development and Aging, Faculty of Health Sciences, University of Macau, Taipa, Macau SAR, 999078, China.,MoE Frontiers Science Center for Precision Oncology, University of Macau, Taipa, Macau SAR, 999078, China
| |
Collapse
|
9
|
A Rationale for Hypoxic and Chemical Conditioning in Huntington's Disease. Int J Mol Sci 2021; 22:ijms22020582. [PMID: 33430140 PMCID: PMC7826574 DOI: 10.3390/ijms22020582] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 12/23/2020] [Accepted: 01/05/2021] [Indexed: 12/17/2022] Open
Abstract
Neurodegenerative diseases are characterized by adverse cellular environments and pathological alterations causing neurodegeneration in distinct brain regions. This development is triggered or facilitated by conditions such as hypoxia, ischemia or inflammation and is associated with disruptions of fundamental cellular functions, including metabolic and ion homeostasis. Targeting intracellular downstream consequences to specifically reverse these pathological changes proved difficult to translate to clinical settings. Here, we discuss the potential of more holistic approaches with the purpose to re-establish a healthy cellular environment and to promote cellular resilience. We review the involvement of important molecular pathways (e.g., the sphingosine, δ-opioid receptor or N-Methyl-D-aspartate (NMDA) receptor pathways) in neuroprotective hypoxic conditioning effects and how these pathways can be targeted for chemical conditioning. Despite the present scarcity of knowledge on the efficacy of such approaches in neurodegeneration, the specific characteristics of Huntington’s disease may make it particularly amenable for such conditioning techniques. Not only do classical features of neurodegenerative diseases like mitochondrial dysfunction, oxidative stress and inflammation support this assumption, but also specific Huntington’s disease characteristics: a relatively young age of neurodegeneration, molecular overlap of related pathologies with hypoxic adaptations and sensitivity to brain hypoxia. The aim of this review is to discuss several molecular pathways in relation to hypoxic adaptations that have potential as drug targets in neurodegenerative diseases. We will extract the relevance for Huntington’s disease from this knowledge base.
Collapse
|
10
|
Yang CC, Hsiao LD, Su MH, Yang CM. Sphingosine 1-Phosphate Induces Cyclooxygenase-2/Prostaglandin E 2 Expression via PKCα-dependent Mitogen-Activated Protein Kinases and NF-κB Cascade in Human Cardiac Fibroblasts. Front Pharmacol 2020; 11:569802. [PMID: 33192511 PMCID: PMC7662885 DOI: 10.3389/fphar.2020.569802] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Accepted: 09/22/2020] [Indexed: 12/12/2022] Open
Abstract
In the regions of tissue injuries and inflammatory diseases, sphingosine 1-phosphate (S1P), a proinflammatory mediator, is increased. S1P may induce the upregulation of cyclooxygenase-2 (COX-2)/prostaglandin E2 (PGE2) system in various types of cells to exacerbate heart inflammation. However, the detailed molecular mechanisms by which S1P induces COX-2 expression in human cardiac fibroblasts (HCFs) remain unknown. HCFs were incubated with S1P and analyzed by Western blotting, real time-Polymerase chain reaction (RT-PCR), and immunofluorescent staining. Our results indicated that S1P activated S1PR1/3-dependent transcriptional activity to induce COX-2 expression and PGE2 production. S1P recruited and activated PTX-sensitive Gi or -insensitive Gq protein-coupled S1PR and then stimulated PKCα-dependent phosphorylation of p42/p44 MAPK, p38 MAPK, and JNK1/2, leading to activating transcription factor NF-κB. Moreover, S1P-activated NF-κB was translocated into the nucleus and bound to its corresponding binding sites on COX-2 promoters determined by chromatin immunoprecipitation (ChIP) and promoter-reporter assays, thereby turning on COX-2 gene transcription associated with PGE2 production in HCFs. These results concluded that in HCFs, activation of NF-κB by PKCα-mediated MAPK cascades was essential for S1P-induced up-regulation of the COX-2/PGE2 system. Understanding the mechanisms of COX-2 expression and PGE2 production regulated by the S1P/S1PRs system on cardiac fibroblasts may provide rationally therapeutic interventions for heart injury or inflammatory diseases.
Collapse
Affiliation(s)
- Chien-Chung Yang
- Department of Traditional Chinese Medicine, Chang Gung Memorial Hospital at Tao-Yuan, Tao-Yuan, Taiwan.,School of Traditional Chinese Medicine, College of Medicine, Chang Gung University, Tao-Yuan, Taiwan
| | - Li-Der Hsiao
- Department of Pharmacology, College of Medicine, China Medical University, Taichung, Taiwan
| | - Mei-Hsiu Su
- Department of Pharmacology, College of Medicine, China Medical University, Taichung, Taiwan
| | - Chuen-Mao Yang
- Department of Pharmacology, College of Medicine, China Medical University, Taichung, Taiwan.,Department of Post-Baccalaureate Veterinary Medicine, College of Medical and Health Science, Asia University, Wufeng, Taichung, Taiwan
| |
Collapse
|
11
|
Wang D, Tabti R, Elderwish S, Djehal A, Chouha N, Pinot F, Yu P, Nebigil CG, Désaubry L. SFPH proteins as therapeutic targets for a myriad of diseases. Bioorg Med Chem Lett 2020; 30:127600. [PMID: 33035678 PMCID: PMC7536521 DOI: 10.1016/j.bmcl.2020.127600] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 09/27/2020] [Accepted: 09/30/2020] [Indexed: 12/21/2022]
Abstract
The stomatin/prohibitin/flotillin/HflK/HflC (SPFH) domain is present in an evolutionarily conserved family of proteins that regulate a myriad of signaling pathways in archaea, bacteria and eukaryotes. The most studied SPFH proteins, prohibitins, have already been targeted by different families of small molecules to induce anticancer, cardioprotective, anti-inflammatory, antiviral, and antiosteoporotic activities. Ligands of other SPFH proteins have also been identified and shown to act as anesthetics, anti-allodynia, anticancer, and anti-inflammatory agents. These findings indicate that modulators of human or bacterial SPFH proteins can be developed to treat a wide variety of human disorders.
Collapse
Affiliation(s)
- Dong Wang
- Sino-French Joint Lab of Food Nutrition/Safety and Medicinal Chemistry, College of Biotechnology, Tianjin University of Science and Technology, Tianjin, China
| | - Redouane Tabti
- INSERM-University of Strasbourg, Regenerative Nanomedicine Laboratory (UMR1260), Faculty of Medicine, FMTS, Strasbourg, France
| | - Sabria Elderwish
- INSERM-University of Strasbourg, Regenerative Nanomedicine Laboratory (UMR1260), Faculty of Medicine, FMTS, Strasbourg, France
| | - Amel Djehal
- Superior National School Biotechnology Taoufik Khaznadar, Constantine, Algeria
| | - Nora Chouha
- University of Batna 2, Faculty of Biology, Batna, Algeria
| | - Franck Pinot
- University of Strasbourg, CNRS, IBMP UPR 2357, Strasbourg, France
| | - Peng Yu
- Sino-French Joint Lab of Food Nutrition/Safety and Medicinal Chemistry, College of Biotechnology, Tianjin University of Science and Technology, Tianjin, China
| | - Canan G Nebigil
- INSERM-University of Strasbourg, Regenerative Nanomedicine Laboratory (UMR1260), Faculty of Medicine, FMTS, Strasbourg, France
| | - Laurent Désaubry
- Sino-French Joint Lab of Food Nutrition/Safety and Medicinal Chemistry, College of Biotechnology, Tianjin University of Science and Technology, Tianjin, China; INSERM-University of Strasbourg, Regenerative Nanomedicine Laboratory (UMR1260), Faculty of Medicine, FMTS, Strasbourg, France.
| |
Collapse
|
12
|
Wang D, Tabti R, Elderwish S, Abou-Hamdan H, Djehal A, Yu P, Yurugi H, Rajalingam K, Nebigil CG, Désaubry L. Prohibitin ligands: a growing armamentarium to tackle cancers, osteoporosis, inflammatory, cardiac and neurological diseases. Cell Mol Life Sci 2020; 77:3525-3546. [PMID: 32062751 PMCID: PMC11104971 DOI: 10.1007/s00018-020-03475-1] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Revised: 01/28/2020] [Accepted: 01/30/2020] [Indexed: 02/08/2023]
Abstract
Over the last three decades, the scaffold proteins prohibitins-1 and -2 (PHB1/2) have emerged as key signaling proteins regulating a myriad of signaling pathways in health and diseases. Small molecules targeting PHBs display promising effects against cancers, osteoporosis, inflammatory, cardiac and neurodegenerative diseases. This review provides an updated overview of the various classes of PHB ligands, with an emphasis on their mechanism of action and therapeutic potential. We also describe how these ligands have been used to explore PHB signaling in different physiological and pathological settings.
Collapse
Affiliation(s)
- Dong Wang
- Sino-French Joint Lab of Food Nutrition/Safety and Medicinal Chemistry, College of Biotechnology, Tianjin University of Science and Technology, Tianjin, China
| | - Redouane Tabti
- Laboratory of Cardio-Oncology and Medicinal Chemistry (FRE 2033), CNRS, Institut Le Bel, 4 rue Blaise Pascal, CS 90032, 67081, Strasbourg, France
| | - Sabria Elderwish
- Laboratory of Cardio-Oncology and Medicinal Chemistry (FRE 2033), CNRS, Institut Le Bel, 4 rue Blaise Pascal, CS 90032, 67081, Strasbourg, France
| | - Hussein Abou-Hamdan
- Laboratory of Cardio-Oncology and Medicinal Chemistry (FRE 2033), CNRS, Institut Le Bel, 4 rue Blaise Pascal, CS 90032, 67081, Strasbourg, France
| | - Amel Djehal
- Laboratory of Cardio-Oncology and Medicinal Chemistry (FRE 2033), CNRS, Institut Le Bel, 4 rue Blaise Pascal, CS 90032, 67081, Strasbourg, France
- Superior National School Biotechnology Taoufik Khaznadar, Ville universitaire Ali Mendjeli, BP E66 25100, Constantine, Algeria
| | - Peng Yu
- Sino-French Joint Lab of Food Nutrition/Safety and Medicinal Chemistry, College of Biotechnology, Tianjin University of Science and Technology, Tianjin, China
| | - Hajime Yurugi
- Cell Biology Unit, University Medical Center Mainz, JGU-Mainz, Mainz, Germany
| | | | - Canan G Nebigil
- Laboratory of Cardio-Oncology and Medicinal Chemistry (FRE 2033), CNRS, Institut Le Bel, 4 rue Blaise Pascal, CS 90032, 67081, Strasbourg, France
| | - Laurent Désaubry
- Sino-French Joint Lab of Food Nutrition/Safety and Medicinal Chemistry, College of Biotechnology, Tianjin University of Science and Technology, Tianjin, China.
- Laboratory of Cardio-Oncology and Medicinal Chemistry (FRE 2033), CNRS, Institut Le Bel, 4 rue Blaise Pascal, CS 90032, 67081, Strasbourg, France.
| |
Collapse
|
13
|
Qian Y, Gao C, Zhao X, Song Y, Luo H, An S, Huang J, Zhang J, Jiang R. Fingolimod Attenuates Lung Injury and Cardiac Dysfunction after Traumatic Brain Injury. J Neurotrauma 2020; 37:2131-2140. [PMID: 32434456 DOI: 10.1089/neu.2019.6951] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Acute lung injury (ALI) and cardiac dysfunction are common in traumatic brain injury (TBI) patients and always indicate poor outcomes. Inflammatory responses play important roles in TBI-induced cardiac and pulmonary damage. Fingolimod, an immunomodulatory agent, alleviates brain edema, restores the integrity of the blood-brain barrier (BBB), and improves functional deficits by inhibiting multiple inflammatory responses. Fingolimod (1 mg/kg) was injected intraperitoneally at 2 h after the controlled cortical impact (CCI) model was established in adult male mice. The concentration of inflammatory cytokines in the lung and heart after TBI was measured with a cytokine array. The lung wet/dry weight ratio and Evans blue dye leakage were used to quantify pulmonary edema and capillary leakage. Immunofluorescence, electron microscopy, and echocardiographic examination were used to assess the pathology and functional deficits in hearts. We found that TBI caused significant heart and lung damage. The administration of fingolimod significantly reduced the elevated inflammatory cytokine production, neutrophil infiltration, the leakage of protein in bronchoalveolar lavage fluid (BALF), and the wet/dry weight ratio in lung tissue at 3 days after TBI. In addition, fingolimod treatment also alleviated the inflammatory response in the heart; decreased cardiac apoptosis, fibrosis, and histological microstructural changes; and improved cardiac function from 3 days after TBI and maintained it for 30 days after TBI as measured by echocardiography. These results suggest that TBI resulted in significant cardiac and pulmonary damage accompanied by significant inflammatory responses in heart and lung tissue. Fingolimod treatment reduced the inflammatory response and alleviated TBI-induced lung and heart injury.
Collapse
Affiliation(s)
- Yu Qian
- Department of Neurology, Tianjin Medical University General Hospital, Tianjin, China.,Tianjin Neurological Institute, Key Laboratory of Post-Neuroinjury Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin, China
| | - Chuang Gao
- Tianjin Neurological Institute, Key Laboratory of Post-Neuroinjury Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin, China.,Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
| | | | - Yiming Song
- Tianjin Neurological Institute, Key Laboratory of Post-Neuroinjury Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin, China.,Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Hongliang Luo
- Tianjin Neurological Institute, Key Laboratory of Post-Neuroinjury Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin, China.,Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Shuo An
- Tianjin Neurological Institute, Key Laboratory of Post-Neuroinjury Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin, China.,Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Jinhao Huang
- Tianjin Neurological Institute, Key Laboratory of Post-Neuroinjury Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin, China.,Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Jianning Zhang
- Tianjin Neurological Institute, Key Laboratory of Post-Neuroinjury Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin, China.,Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Rongcai Jiang
- Tianjin Neurological Institute, Key Laboratory of Post-Neuroinjury Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin, China.,Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
| |
Collapse
|
14
|
Raza Z, Saleem U, Naureen Z. Sphingosine 1-phosphate signaling in ischemia and reperfusion injury. Prostaglandins Other Lipid Mediat 2020; 149:106436. [PMID: 32173486 DOI: 10.1016/j.prostaglandins.2020.106436] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 02/28/2020] [Accepted: 03/02/2020] [Indexed: 02/07/2023]
Abstract
Ischemia and reperfusion injury is a complex hemodynamic pathological phenomenon that engages the metabolic to inflammatory machinery in development of disease conditions like heart failure, stroke and acute kidney failure. Target specific therapeutic approaches for ischemia reperfusion injury remains critical despite the extensive studies contributing to the understanding of its pathogenesis. Ischemic or pharmacological conditionings have been long established manipulations to harness the endogenous protective mechanisms against ischemia reperfusion injury that fostered the development of potential therapeutic targets such as sphingolipids signaling. Sphingosine 1-phosphate has been emerged as a crucial metabolite of sphingolipids to regulate the cell survival, vascular integrity and inflammatory cascades in ischemia reperfusion injury. Sphingosine 1-phosphate signaling process has been implicated to downgrade the mitochondrial dysfunction, apoptotic assembly along with upregulation of RISK and SAFE pro-survival pathways. It also regulates the endothelial dysfunction and immune cells behavior to control the vascular permeability and immune cells infiltration at ischemia reperfusion injury site. Targeting the signaling of this single moiety holds the vast potential to extensively influence the detrimental signaling of ischemia reperfusion injury. This review highlights the role and significance of S1P signaling that can be therapeutically exploit to treat ischemia reperfusion injury mediated pathological conditions in different organs.
Collapse
Affiliation(s)
- Zohaib Raza
- Government College University, Faisalabad, Pakistan.
| | - Uzma Saleem
- Government College University, Faisalabad, Pakistan
| | | |
Collapse
|
15
|
Blais-Lecours P, Laouafa S, Arias-Reyes C, Santos WL, Joseph V, Burgess JK, Halayko AJ, Soliz J, Marsolais D. Metabolic Adaptation of Airway Smooth Muscle Cells to an SPHK2 Substrate Precedes Cytostasis. Am J Respir Cell Mol Biol 2020; 62:35-42. [PMID: 31247144 PMCID: PMC6938129 DOI: 10.1165/rcmb.2018-0397oc] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Accepted: 06/26/2019] [Indexed: 12/31/2022] Open
Abstract
Thickening of the airway smooth muscle is central to bronchial hyperreactivity. We have shown that the sphingosine analog (R)-2-amino-4-(4-heptyloxyphenyl)-2-methylbutanol (AAL-R) can reverse preestablished airway hyperreactivity in a chronic asthma model. Because sphingosine analogs can be metabolized by SPHK2 (sphingosine kinase 2), we investigated whether this enzyme was required for AAL-R to perturb mechanisms sustaining airway smooth muscle cell proliferation. We found that AAL-R pretreatment reduced the capacity of live airway smooth muscle cells to use oxygen for oxidative phosphorylation and increased lactate dehydrogenase activity. We also determined that SPHK2 was upregulated in airway smooth muscle cells bearing the proliferation marker Ki67 relative to their Ki67-negative counterpart. Comparing different stromal cell subsets of the lung, we found that high SPHK2 concentrations were associated with the ability of AAL-R to inhibit metabolic activity assessed by conversion of the tetrazolium dye MTT. Knockdown or pharmacological inhibition of SPHK2 reversed the effect of AAL-R on MTT conversion, indicating the essential role for this kinase in the metabolic perturbations induced by sphingosine analogs. Our results support the hypothesis that increased SPHK2 levels in proliferating airway smooth muscle cells could be exploited to counteract airway smooth muscle thickening with synthetic substrates.
Collapse
Affiliation(s)
- Pascale Blais-Lecours
- Centre de Recherche de l’Institut Universitaire de Cardiologie et de Pneumologie de Québec and
| | - Sofien Laouafa
- Centre de Recherche de l’Institut Universitaire de Cardiologie et de Pneumologie de Québec and
| | - Christian Arias-Reyes
- Centre de Recherche de l’Institut Universitaire de Cardiologie et de Pneumologie de Québec and
| | - Webster L. Santos
- Department of Chemistry and Virginia Tech Center for Drug Discovery, Virginia Tech, Blacksburg, Virginia
| | - Vincent Joseph
- Centre de Recherche de l’Institut Universitaire de Cardiologie et de Pneumologie de Québec and
- Faculty of Medicine, Université Laval, Québec City, Québec, Canada
| | - Janette K. Burgess
- Department of Pathology and Medical Biology, Experimental Pulmonology and Inflammation Research and
- GRIAC (Groningen Research Institute for Asthma and COPD), University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Andrew J. Halayko
- Department of Physiology and Pathophysiology, University of Manitoba, Winnipeg, Manitoba, Canada; and
- Biology of Breathing Group, Children’s Hospital Research Institute of Manitoba, Winnipeg, Manitoba, Canada
| | - Jorge Soliz
- Centre de Recherche de l’Institut Universitaire de Cardiologie et de Pneumologie de Québec and
- Faculty of Medicine, Université Laval, Québec City, Québec, Canada
| | - David Marsolais
- Centre de Recherche de l’Institut Universitaire de Cardiologie et de Pneumologie de Québec and
- Faculty of Medicine, Université Laval, Québec City, Québec, Canada
| |
Collapse
|
16
|
S1P/S1P Receptor Signaling in Neuromuscolar Disorders. Int J Mol Sci 2019; 20:ijms20246364. [PMID: 31861214 PMCID: PMC6941007 DOI: 10.3390/ijms20246364] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Revised: 12/06/2019] [Accepted: 12/13/2019] [Indexed: 12/13/2022] Open
Abstract
The bioactive sphingolipid metabolite, sphingosine 1-phosphate (S1P), and the signaling pathways triggered by its binding to specific G protein-coupled receptors play a critical regulatory role in many pathophysiological processes, including skeletal muscle and nervous system degeneration. The signaling transduced by S1P binding appears to be much more complex than previously thought, with important implications for clinical applications and for personalized medicine. In particular, the understanding of S1P/S1P receptor signaling functions in specific compartmentalized locations of the cell is worthy of being better investigated, because in various circumstances it might be crucial for the development or/and the progression of neuromuscular diseases, such as Charcot-Marie-Tooth disease, myasthenia gravis, and Duchenne muscular dystrophy.
Collapse
|
17
|
Ahmed N, Laghari AH, AlBkhoor B, Tabassum S, Meo SA, Muhammad N, Linardi D, Al-Masri AA, Fumagalli G, Luciani GB, Faggian G, Rungatscher A. Fingolimod Plays Role in Attenuation of Myocardial Injury Related to Experimental Model of Cardiac Arrest and Extracorporeal Life Support Resuscitation. Int J Mol Sci 2019; 20:ijms20246237. [PMID: 31835656 PMCID: PMC6940876 DOI: 10.3390/ijms20246237] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Revised: 11/22/2019] [Accepted: 12/05/2019] [Indexed: 12/21/2022] Open
Abstract
Background: Sudden cardiac arrest is a major global health concern, and survival of patients with ischemia–reperfusion injury is a leading cause of myocardial dysfunction. The mechanism of this phenomenon is not well understood because of the complex pathophysiological nature of the disease. Aim of the study was to investigate the cardioprotective role of fingolimod in an in vivo model of cardiac arrest and resuscitation. Methods: In this study, an in vivo rat model of cardiac arrest using extracorporeal membrane oxygenation resuscitation monitored by invasive hemodynamic measurement was developed. At the beginning of extracorporeal life support (ECLS), animals were randomly treated with fingolimod (Group A, n = 30) or saline (Group B, n = 30). Half of the animals in each group (Group A1 and B1, n = 15 each) were sacrificed after 1 h, and the remaining animals (Group A2 and B2) after 24 h of reperfusion. Blood and myocardial tissues were collected for analysis of cardiac features, inflammatory biomarkers, and cell signaling pathways. Results: Treatment with fingolimod resulted in activation of survival pathways resulting into reduced inflammation, myocardial oxidative stress and apoptosis of cardiomyocytes. This led to significant improvement in systolic and diastolic functions of the left ventricle and improved contractility index. Conclusions: Sphingosine1phosphate receptor activation with fingolimod improved cardiac function after cardiac arrest supported with ECLS. Present study findings strongly support a cardioprotective role of fingolimod through sphingosine-1-phosphate receptor activation during reperfusion after circulatory arrest.
Collapse
Affiliation(s)
- Naseer Ahmed
- Department of Biological and Biomedical Sciences, The Aga Khan University, Karachi 74800, Pakistan
- Department of Surgery, Cardiac Surgery Division, University of Verona Medical School, 37129 Verona, Italy; (D.L.); (G.B.L.); (G.F.); (A.R.)
- Correspondence:
| | - Abid H. Laghari
- Department of Medicine, section of Cardiology, Aga Khan University, Karachi 74800, Pakistan;
| | | | - Sobia Tabassum
- Department of Biological Sciences, International Islamic University, Islamabad 44000, Pakistan;
| | - Sultan Ayoub Meo
- Department of Physiology, College of Medicine, King Saud University, Riyadh 11461, Saudi Arabia; (S.A.M.); (A.A.A.-M.)
| | - Nazeer Muhammad
- COMSATS University Islamabad, Wah Campus, Rawalpindi 47040, Pakistan;
| | - Daniele Linardi
- Department of Surgery, Cardiac Surgery Division, University of Verona Medical School, 37129 Verona, Italy; (D.L.); (G.B.L.); (G.F.); (A.R.)
| | - Abeer A. Al-Masri
- Department of Physiology, College of Medicine, King Saud University, Riyadh 11461, Saudi Arabia; (S.A.M.); (A.A.A.-M.)
| | - Guido Fumagalli
- Department of Diagnostics and Public Health, Section of Pharmacology, University of Verona Medical School, 37134 Verona, Italy;
| | - Giovanni Battista Luciani
- Department of Surgery, Cardiac Surgery Division, University of Verona Medical School, 37129 Verona, Italy; (D.L.); (G.B.L.); (G.F.); (A.R.)
| | - Giuseppe Faggian
- Department of Surgery, Cardiac Surgery Division, University of Verona Medical School, 37129 Verona, Italy; (D.L.); (G.B.L.); (G.F.); (A.R.)
| | - Alessio Rungatscher
- Department of Surgery, Cardiac Surgery Division, University of Verona Medical School, 37129 Verona, Italy; (D.L.); (G.B.L.); (G.F.); (A.R.)
| |
Collapse
|
18
|
Elderwish S, Audebrand A, Nebigil CG, Désaubry L. Discovery of 3,3'-pyrrolidinyl-spirooxindoles as cardioprotectant prohibitin ligands. Eur J Med Chem 2019; 186:111859. [PMID: 31735574 DOI: 10.1016/j.ejmech.2019.111859] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 11/04/2019] [Accepted: 11/05/2019] [Indexed: 01/08/2023]
Abstract
The scaffold proteins prohibitins-1 and 2 (PHB1/2) play many important roles in coordinating many cell signaling pathways and represent emerging targets in cardiology and oncology. We previously reported that a family of natural products derivatives, flavaglines, binds to PHB1/2 to exert cardioprotectant and anti-cancer effects. However, flavaglines also target the initiation factor of translation eIF4A, which doesn't contribute to cardioprotection and may even induce some adverse effects. Herein, we report the development of a convenient and robust synthesis of the new PHB2 ligand 2'-phenylpyrrolidinyl-spirooxindole, and its analogues. We discovered that these compounds displays cardioprotective effect against doxorubicin mediated cardiotoxicity and uncovered the structural requirement for this activity. We identified in particular some analogues that are more cardioprotectant than flavaglines. Pull-down experiments demonstrated that these compounds bind not only to PHB2 but also PHB1. These novel PHB ligands may provide the basis for the development of new drugs candidates to protect the heart against the adverse effects of anticancer treatments.
Collapse
Affiliation(s)
- Sabria Elderwish
- Laboratory of Medicinal Chemistry and Cardio-oncology, CNRS, 4 rue Blaise Pascal, 67081, Strasbourg, France
| | - Anaïs Audebrand
- Laboratory of Medicinal Chemistry and Cardio-oncology, CNRS, Ecole Supérieure de Biotechnologie de Strasbourg, Illkirch, France
| | - Canan G Nebigil
- Laboratory of Medicinal Chemistry and Cardio-oncology, CNRS, Ecole Supérieure de Biotechnologie de Strasbourg, Illkirch, France
| | - Laurent Désaubry
- Laboratory of Medicinal Chemistry and Cardio-oncology, CNRS, 4 rue Blaise Pascal, 67081, Strasbourg, France.
| |
Collapse
|
19
|
Pulli I, Löf C, Blom T, Asghar M, Lassila T, Bäck N, Lin KL, Nyström J, Kemppainen K, Toivola D, Dufour E, Sanz A, Cooper H, Parys J, Törnquist K. Sphingosine kinase 1 overexpression induces MFN2 fragmentation and alters mitochondrial matrix Ca2+ handling in HeLa cells. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2019; 1866:1475-1486. [DOI: 10.1016/j.bbamcr.2019.06.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Revised: 06/02/2019] [Accepted: 06/13/2019] [Indexed: 01/08/2023]
|
20
|
Ahmed N. Cardioprotective mechanism of FTY720 in ischemia reperfusion injury. J Basic Clin Physiol Pharmacol 2019; 30:jbcpp-2019-0063. [PMID: 31469655 DOI: 10.1515/jbcpp-2019-0063] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Accepted: 07/06/2019] [Indexed: 12/17/2022]
Abstract
Cardioprotection is a very challenging area in the field of cardiovascular sciences. Myocardial damage accounts for nearly 50% of injury due to reperfusion, yet there is no effective strategy to prevent this to reduce the burden of heart failure. During last couple of decades, by combining genetic and bimolecular studies, many new drugs have been developed to treat hypertension, heart failure, and cancer. The use of percutaneous coronary intervention has reduced the mortality and morbidity of acute coronary syndrome dramatically. However, there is no standard therapy available that can mitigate cardiac reperfusion injury, which contributes to up to half of myocardial infarcts. Literature shows that the activation of sphingosine receptors, which are G protein-coupled receptors, induces cardioprotection both in vitro and in vivo. The exact mechanism of this protection is not clear yet. In this review, we discuss the mechanism of ischemia reperfusion injury and the role of the FDA-approved sphingosine 1 phosphate drug fingolimod in cardioprotection.
Collapse
Affiliation(s)
- Naseer Ahmed
- The Aga Khan University, Medical College, Karachi, Pakistan, Phone: +92 21 3486 4465
| |
Collapse
|
21
|
Ahmed N, Mehmood A, Linardi D, Sadiq S, Tessari M, Meo SA, Rehman R, Hajjar WM, Muhammad N, Iqbal MP, Gilani AUH, Faggian G, Rungatscher A. Cardioprotective Effects of Sphingosine-1-Phosphate Receptor Immunomodulator FTY720 in a Clinically Relevant Model of Cardioplegic Arrest and Cardiopulmonary Bypass. Front Pharmacol 2019; 10:802. [PMID: 31379576 PMCID: PMC6656862 DOI: 10.3389/fphar.2019.00802] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Accepted: 06/21/2019] [Indexed: 12/28/2022] Open
Abstract
Objective: FTY720, an immunomodulator derived from sphingosine-1-phosphate, has recently demonstrated its immunomodulatory, anti-inflammatory, anti-oxidant, anti-apoptotic and anti-inflammatory properties. Furthermore, FTY720 might be a key pharmacological target for preconditioning. In this preclinical model, we have investigated the effects of FTY720 on myocardium during reperfusion in an experimental model of cardioplegic arrest (CPA) and cardiopulmonary bypass. Methods: 30 Sprague–Dawley rats (300–350 g) were randomized into two groups: Group-A, treated with FTY720 1 mg/kg via intravenous cannulation, and Group-B, as control. After 15 min of treatment, rats underwent CPA for 30 min followed by initiation of extracorporeal life support for 2 h. Support weaning was done, and blood and myocardial tissues were collected for analysis. Hemodynamic parameters, inflammatory mediators, nitro-oxidative stress, neutrophil infiltration, immunoblotting analysis, and immunohistochemical staining were analyzed and compared between groups. Results: FTY720 treatment activated the Akt/Erk1/2 signaling pathways, reduced the level of inflammatory mediators, activated antiapoptotic proteins, and inhibited proapoptotic proteins, leading to reduced nitro-oxidative stress and cardiomyocyte apoptosis. Moreover, significant preservation of high-energy phosphates were observed in the FTY720-treated group. This resulted in improved recovery of left ventricular systolic and diastolic functions. Conclusion: The cardioprotective mechanism in CPA is associated with activation of prosurvival cell signaling pathways that prevents myocardial damage. FTY720 preserves high-energy phosphates attenuates myocardial inflammation and oxidative stress, and improves cardiac function.
Collapse
Affiliation(s)
- Naseer Ahmed
- Department of Biological and Biomedical Sciences, The Aga Khan University, Karachi, Pakistan.,Department of Surgery, Cardiac Surgery Division, University of Verona Medical School, Verona, Italy
| | - Adeela Mehmood
- Department of Pharmacology, Liaqat National Medical College, Karachi, Pakistan
| | - Daniele Linardi
- Department of Surgery, Cardiac Surgery Division, University of Verona Medical School, Verona, Italy
| | - Soban Sadiq
- Pharmacology and Molecular Lab, University of Liverpool, United Kingdom
| | - Maddalena Tessari
- Department of Surgery, Cardiac Surgery Division, University of Verona Medical School, Verona, Italy
| | - Sultan Ayoub Meo
- Department of Physiology, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Rehana Rehman
- Department of Biological and Biomedical Sciences, The Aga Khan University, Karachi, Pakistan
| | - Waseem M Hajjar
- Department of Thoracic Surgery, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Nazeer Muhammad
- Department of Mathematics, COMSATS University Islamabad, Wah Campus, Pakistan
| | - Muhammad Perwaiz Iqbal
- Department of Biological and Biomedical Sciences, The Aga Khan University, Karachi, Pakistan
| | - Anwar-Ul-Hassan Gilani
- Department of Biological and Biomedical Sciences, The Aga Khan University, Karachi, Pakistan
| | - Giuseppe Faggian
- Department of Surgery, Cardiac Surgery Division, University of Verona Medical School, Verona, Italy
| | - Alessio Rungatscher
- Department of Surgery, Cardiac Surgery Division, University of Verona Medical School, Verona, Italy
| |
Collapse
|
22
|
Heusch G. There Is More to β-Blockade Than Just Blockade of β-Receptors: A Case for Cardioprotective Cross-Signaling. J Am Coll Cardiol 2019; 70:193-195. [PMID: 28683967 DOI: 10.1016/j.jacc.2017.05.017] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Accepted: 05/09/2017] [Indexed: 11/25/2022]
Affiliation(s)
- Gerd Heusch
- Institute for Pathophysiology, West German Heart and Vascular Center, University of Essen Medical School, Essen, Germany.
| |
Collapse
|
23
|
Abstract
Long noncoding RNAs (lncRNAs) are an important group of pervasive noncoding RNAs (>200nt) proposed to be crucial regulators of numerous physiological and pathological processes. Through interactions with RNA, chromatin, and protein, lncRNAs modulate mRNA stability, chromatin structure, and the function of proteins (including transcription factors). In addition, to their well-known roles in the modulation of cell growth, apoptosis, neurological disease progression and cancer metastasis, these large molecules have also been identified as likely mediators of lipid metabolism. In particular, lncRNAs orchestrate adipogenesis; fatty acid, cholesterol, and phospholipid metabolism and transport; and the formation of high-density and low-density lipoproteins (HDLs and LDLs). LncRNAs also appear to target several transcription factors that play essential roles in the regulation of lipid metabolism, such as liver X receptors (LXRs), sterol regulatory element binding proteins (SREBPs), and peroxisome proliferator-activated receptor γ (PPARγ). Better understanding the regulatory roles of lncRNAs in dyslipidemia, atherosclerosis, and adipogenesis will reveal appropriate strategies to treat these diseases. In this review, we review recent progress in lncRNA-mediated regulation of lipid metabolism, as well as its role in the regulation of adipogenesis.
Collapse
|
24
|
Di Pardo A, Maglione V. Sphingolipid Metabolism: A New Therapeutic Opportunity for Brain Degenerative Disorders. Front Neurosci 2018; 12:249. [PMID: 29719499 PMCID: PMC5913346 DOI: 10.3389/fnins.2018.00249] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2017] [Accepted: 03/29/2018] [Indexed: 01/01/2023] Open
Abstract
Neurodegenerative diseases represent a class of fatal brain disorders for which the number of effective therapeutic options remains limited with only symptomatic treatment accessible. Multiple studies show that defects in sphingolipid pathways are shared among different brain disorders including neurodegenerative diseases and may contribute to their complex pathogenesis. In this mini review, we discuss the hypothesis that modulation of sphingolipid metabolism and their related signaling pathways may represent a potential therapeutic approach for those devastating conditions. The plausible “druggability” of sphingolipid pathways is greatly promising and represent a relevant feature that brings real advantage to the development of new therapeutic options for these conditions. Indeed, several molecules that selectively target sphingolipds are already available and many of them currently in clinical trial for human diseases. A deeper understanding of the “sphingolipid scenario” in neurodegenerative disorders would certainly enhance therapeutic perspectives for these conditions, by taking advantage from the already available molecules and by promoting the development of new ones.
Collapse
|
25
|
The Role of Sphingosine-1-Phosphate and Ceramide-1-Phosphate in Inflammation and Cancer. Mediators Inflamm 2017; 2017:4806541. [PMID: 29269995 PMCID: PMC5705877 DOI: 10.1155/2017/4806541] [Citation(s) in RCA: 132] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Revised: 08/01/2017] [Accepted: 08/30/2017] [Indexed: 01/02/2023] Open
Abstract
Inflammation is part of our body's response to tissue injury and pathogens. It helps to recruit various immune cells to the site of inflammation and activates the production of mediators to mobilize systemic protective processes. However, chronic inflammation can increase the risk of diseases like cancer. Apart from cytokines and chemokines, lipid mediators, particularly sphingosine-1-phosphate (S1P) and ceramide-1-phosphate (C1P), contribute to inflammation and cancer. S1P is an important player in inflammation-associated colon cancer progression. On the other hand, C1P has been recognized to be involved in cancer cell growth, migration, survival, and inflammation. However, whether C1P is involved in inflammation-associated cancer is not yet established. In contrast, few studies have also suggested that S1P and C1P are involved in anti-inflammatory pathways regulated in certain cell types. Ceramide is the substrate for ceramide kinase (CERK) to yield C1P, and sphingosine is phosphorylated to S1P by sphingosine kinases (SphKs). Biological functions of sphingolipid metabolites have been studied extensively. Ceramide is associated with cell growth inhibition and enhancement of apoptosis while S1P and C1P are associated with enhancement of cell growth and survival. Altogether, S1P and C1P are important regulators of ceramide level and cell fate. This review focuses on S1P and C1P involvement in inflammation and cancer with emphasis on recent progress in the field.
Collapse
|
26
|
Ganta VC, Annex BH. LMO2 (LIM Domain Only 2) and Endothelial Cell Migration in Developmental and Postnatal Angiogenesis. Arterioscler Thromb Vasc Biol 2017; 37:1806-1808. [PMID: 28954807 DOI: 10.1161/atvbaha.117.309953] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Vijay Chaitanya Ganta
- From the Robert M. Berne Cardiovascular Research Center (V.C.G., B.H.A.) and Division of Cardiovascular Medicine, Department of Medicine (B.H.A.), University of Virginia School of Medicine, Charlottesville
| | - Brian H Annex
- From the Robert M. Berne Cardiovascular Research Center (V.C.G., B.H.A.) and Division of Cardiovascular Medicine, Department of Medicine (B.H.A.), University of Virginia School of Medicine, Charlottesville.
| |
Collapse
|
27
|
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: 72] [Impact Index Per Article: 9.0] [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
|
28
|
Fang R, Zhang LL, Zhang LZ, Li W, Li M, Wen K. Sphingosine 1-Phosphate Postconditioning Protects Against Myocardial Ischemia/reperfusion Injury in Rats via Mitochondrial Signaling and Akt-Gsk3β Phosphorylation. Arch Med Res 2017. [PMID: 28625317 DOI: 10.1016/j.arcmed.2017.03.013] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
BACKGROUND AND AIMS Although preconditioning of sphingosine 1-phosphate (S1P) has been shown to protect myocytes from hypoxia reoxgenation injury in vitro, the role of S1P postconditioning on myocardial ischemia reperfusion injury (MIRI) in vivo and its related mechanism are unknown. The aim of this study was to investigate the protective role of sphingosine 1-phosphate (S1P) postconditioning in MIRI via its effects on mitochondrial signaling and Akt/Gsk3β phosphorylation. METHODS Rats were subjected to MIRI, consisting of 30 min of ischemia followed by 120 min of reperfusion, with S1P administered at the beginning of the reperfusion. Myocardial infarct size and apoptotic index were measured by triphenyltetrazolium (TTC) and terminal deoxynucleotide transferase dUTP nick-end labeling (TUNEL) assays, respectively. Akt and Gsk3β phosphorylation, caspase-3 cleavage, and cytochrome c translocation were assessed by western blot. Mitochondrial permeability transition pore (MPTP) opening and mitochondrial membrane potential (MMP, ΔΨ) were also examined to determine overall mitochondrial function. RESULTS S1P postconditioning significantly decreased myocardial infarct size and apoptosis, as well as enhanced Akt and Gsk3β phosphorylation, attenuated caspase-3 cleavage and cytosolic cytochrome c translocation, and inhibited MPTP opening, which subsequently preserved Δψ. Electron microscopy also confirmed that S1P helped maintain myocardial mitochondria integrity. Moreover, the protective effects of S1P treatment were blocked by cotreatment with a PI3K inhibitor, LY294002. CONCLUSIONS These results suggest that S1P postconditioning protects against MIRI by regulating mitochondrial signaling and Akt/Gsk3β phosphorylation.
Collapse
Affiliation(s)
- Rui Fang
- Department of Pharmacology, College of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Lu-Lu Zhang
- Department of Pharmacology, College of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Li-Zhi Zhang
- Department of Obstetrics and Gynecology, Tianjin First Centre Hospital, Tianjin, China
| | - Wenchang Li
- Department of Pharmacology, College of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Mengmeng Li
- Department of Pharmacology, College of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Ke Wen
- Department of Pharmacology, College of Basic Medical Sciences, Tianjin Medical University, Tianjin, China.
| |
Collapse
|
29
|
Defective Sphingosine-1-phosphate metabolism is a druggable target in Huntington's disease. Sci Rep 2017; 7:5280. [PMID: 28706199 PMCID: PMC5509685 DOI: 10.1038/s41598-017-05709-y] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Accepted: 06/01/2017] [Indexed: 12/22/2022] Open
Abstract
Huntington’s disease is characterized by a complex and heterogeneous pathogenic profile. Studies have shown that disturbance in lipid homeostasis may represent a critical determinant in the progression of several neurodegenerative disorders. The recognition of perturbed lipid metabolism is only recently becoming evident in HD. In order to provide more insight into the nature of such a perturbation and into the effect its modulation may have in HD pathology, we investigated the metabolism of Sphingosine-1-phosphate (S1P), one of the most important bioactive lipids, in both animal models and patient samples. Here, we demonstrated that S1P metabolism is significantly disrupted in HD even at early stage of the disease and importantly, we revealed that such a dysfunction represents a common denominator among multiple disease models ranging from cells to humans through mouse models. Interestingly, the in vitro anti-apoptotic and the pro-survival actions seen after modulation of S1P-metabolizing enzymes allows this axis to emerge as a new druggable target and unfolds its promising therapeutic potential for the development of more effective and targeted interventions against this incurable condition.
Collapse
|
30
|
Vestri A, Pierucci F, Frati A, Monaco L, Meacci E. Sphingosine 1-Phosphate Receptors: Do They Have a Therapeutic Potential in Cardiac Fibrosis? Front Pharmacol 2017. [PMID: 28626422 PMCID: PMC5454082 DOI: 10.3389/fphar.2017.00296] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Sphingosine 1-phosphate (S1P) is a bioactive lipid that is characterized by a peculiar mechanism of action. In fact, S1P, which is produced inside the cell, can act as an intracellular mediator, whereas after its export outside the cell, it can act as ligand of specific G-protein coupled receptors, which were initially named endothelial differentiation gene (Edg) and eventually renamed sphingosine 1-phosphate receptors (S1PRs). Among the five S1PR subtypes, S1PR1, S1PR2 and S1PR3 isoforms show broad tissue gene expression, while S1PR4 is primarily expressed in immune system cells, and S1PR5 is expressed in the central nervous system. There is accumulating evidence for the important role of S1P as a mediator of many processes, such as angiogenesis, carcinogenesis and immunity, and, ultimately, fibrosis. After a tissue injury, the imbalance between the production of extracellular matrix (ECM) and its degradation, which occurs due to chronic inflammatory conditions, leads to an accumulation of ECM and, consequential, organ dysfunction. In these pathological conditions, many factors have been described to act as pro- and anti-fibrotic agents, including S1P. This bioactive lipid exhibits both pro- and anti-fibrotic effects, depending on its site of action. In this review, after a brief description of sphingolipid metabolism and signaling, we emphasize the involvement of the S1P/S1PR axis and the downstream signaling pathways in the development of fibrosis. The current knowledge of the therapeutic potential of S1PR subtype modulators in the treatment of the cardiac functions and fibrinogenesis are also examined.
Collapse
Affiliation(s)
- Ambra Vestri
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", Molecular and Applied Biology Research Unit, University of FlorenceFlorence, Italy
| | - Federica Pierucci
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", Molecular and Applied Biology Research Unit, University of FlorenceFlorence, Italy.,Interuniversity Institutes of MyologyFirenze, Italy
| | - Alessia Frati
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", Molecular and Applied Biology Research Unit, University of FlorenceFlorence, Italy
| | - Lucia Monaco
- Department of Physiology and Pharmacology "Vittorio Erspamer", Sapienza University of RomeRome, Italy
| | - Elisabetta Meacci
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", Molecular and Applied Biology Research Unit, University of FlorenceFlorence, Italy.,Interuniversity Institutes of MyologyFirenze, Italy
| |
Collapse
|
31
|
Hatoum D, Haddadi N, Lin Y, Nassif NT, McGowan EM. Mammalian sphingosine kinase (SphK) isoenzymes and isoform expression: challenges for SphK as an oncotarget. Oncotarget 2017; 8:36898-36929. [PMID: 28415564 PMCID: PMC5482707 DOI: 10.18632/oncotarget.16370] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2017] [Accepted: 03/02/2017] [Indexed: 12/16/2022] Open
Abstract
The various sphingosine kinase (SphK) isoenzymes (isozymes) and isoforms, key players in normal cellular physiology, are strongly implicated in cancer and other diseases. Mutations in SphKs, that may justify abnormal physiological function, have not been recorded. Nonetheless, there is a large and growing body of evidence demonstrating the contribution of gain or loss of function and the imbalance in the SphK/S1P rheostat to a plethora of pathological conditions including cancer, diabetes and inflammatory diseases. SphK is expressed as two isozymes SphK1 and SphK2, transcribed from genes located on different chromosomes and both isozymes catalyze the phosphorylation of sphingosine to S1P. Expression of each SphK isozyme produces alternately spliced isoforms. In recent years the importance of the contribution of SpK1 expression to treatment resistance in cancer has been highlighted and, additionally, differences in treatment outcome appear to also be dependent upon SphK isoform expression. This review focuses on an exciting emerging area of research involving SphKs functions, expression and subcellular localization, highlighting the complexity of targeting SphK in cancer and also comorbid diseases. This review also covers the SphK isoenzymes and isoforms from a historical perspective, from their first discovery in murine species and then in humans, their role(s) in normal cellular function and in disease processes, to advancement of SphK as an oncotarget.
Collapse
Affiliation(s)
- Diana Hatoum
- School of Life Sciences, University of Technology Sydney, Ultimo, Sydney, NSW 2007, Australia
| | - Nahal Haddadi
- School of Life Sciences, University of Technology Sydney, Ultimo, Sydney, NSW 2007, Australia
| | - Yiguang Lin
- School of Life Sciences, University of Technology Sydney, Ultimo, Sydney, NSW 2007, Australia
| | - Najah T. Nassif
- School of Life Sciences, University of Technology Sydney, Ultimo, Sydney, NSW 2007, Australia
| | - Eileen M. McGowan
- School of Life Sciences, University of Technology Sydney, Ultimo, Sydney, NSW 2007, Australia
| |
Collapse
|
32
|
Selective replacement of mitochondrial DNA increases the cardioprotective effect of chronic continuous hypoxia in spontaneously hypertensive rats. Clin Sci (Lond) 2017; 131:865-881. [PMID: 28292971 DOI: 10.1042/cs20170083] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Revised: 03/08/2017] [Accepted: 03/14/2017] [Indexed: 12/13/2022]
Abstract
Mitochondria play an essential role in improved cardiac ischaemic tolerance conferred by adaptation to chronic hypoxia. In the present study, we analysed the effects of continuous normobaric hypoxia (CNH) on mitochondrial functions, including the sensitivity of the mitochondrial permeability transition pore (MPTP) to opening, and infarct size (IS) in hearts of spontaneously hypertensive rats (SHR) and the conplastic SHR-mtBN strain, characterized by the selective replacement of the mitochondrial genome of SHR with that of the more ischaemia-resistant brown Norway (BN) strain. Rats were adapted to CNH (10% O2, 3 weeks) or kept at room air as normoxic controls. In the left ventricular mitochondria, respiration and cytochrome c oxidase (COX) activity were measured using an Oxygraph-2k and the sensitivity of MPTP opening was assessed spectrophotometrically as Ca2+-induced swelling. Myocardial infarction was analysed in anaesthetized open-chest rats subjected to 20 min of coronary artery occlusion and 3 h of reperfusion. The IS reached 68±3.0% and 65±5% of the area at risk in normoxic SHR and SHR-mtBN strains, respectively. CNH significantly decreased myocardial infarction to 46±3% in SHR. In hypoxic SHR-mtBN strain, IS reached 33±2% and was significantly smaller compared with hypoxic SHR. Mitochondria isolated from hypoxic hearts of both strains had increased detergent-stimulated COX activity and were less sensitive to MPTP opening. The maximum swelling rate was significantly lower in hypoxic SHR-mtBN strain compared with hypoxic SHR, and positively correlated with myocardial infarction in all experimental groups. In conclusion, the mitochondrial genome of SHR modulates the IS-limiting effect of adaptation to CNH by affecting mitochondrial energetics and MPTP sensitivity to opening.
Collapse
|
33
|
Huang L, Li T, Liu YW, Zhang L, Dong ZH, Liu SY, Gao YT. Plasma Metabolic Profile Determination in Young ST-segment Elevation Myocardial Infarction Patients with Ischemia and Reperfusion: Ultra-performance Liquid Chromatography and Mass Spectrometry for Pathway Analysis. Chin Med J (Engl) 2017; 129:1078-86. [PMID: 27098794 PMCID: PMC4852676 DOI: 10.4103/0366-6999.180527] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Background: This study was to establish a disease differentiation model for ST-segment elevation myocardial infarction (STEMI) youth patients experiencing ischemia and reperfusion via ultra-performance liquid chromatography and mass spectrometry (UPLC/MS) platform, which searches for closely related characteristic metabolites and metabolic pathways to evaluate their predictive value in the prognosis after discharge. Methods: Forty-seven consecutive STEMI patients (23 patients under 45 years of age, referred to here as “youth,” and 24 “elderly” patients) and 48 healthy control group members (24 youth, 24 elderly) were registered prospectively. The youth patients were required to provide a second blood draw during a follow-up visit one year after morbidity (n = 22, one lost). Characteristic metabolites and relative metabolic pathways were screened via UPLC/MS platform base on the Kyoto encyclopedia of genes and genomes (KEGG) and Human Metabolome Database. Receiver operating characteristic (ROC) curves were drawn to evaluate the predictive value of characteristic metabolites in the prognosis after discharge. Results: We successfully established an orthogonal partial least squares discriminated analysis model (R2X = 71.2%, R2Y = 79.6%, and Q2 = 55.9%) and screened out 24 ions; the sphingolipid metabolism pathway showed the most drastic change. The ROC curve analysis showed that ceramide [Cer(d18:0/16:0), Cer(t18:0/12:0)] and sphinganine in the sphingolipid pathway have high sensitivity and specificity on the prognosis related to major adverse cardiovascular events after youth patients were discharged. The area under curve (AUC) was 0.671, 0.750, and 0.711, respectively. A follow-up validation one year after morbidity showed corresponding AUC of 0.778, 0.833, and 0.806. Conclusions: By analyzing the plasma metabolism of myocardial infarction patients, we successfully established a model that can distinguish two different factors simultaneously: pathological conditions and age. Sphingolipid metabolism is the top most altered pathway in young STEMI patients and as such may represent a valuable prognostic factor and potential therapeutic target.
Collapse
Affiliation(s)
| | - Tong Li
- Department of Heart Center, Tianjin Medical University, the Third Central Clinical Medicine College, Tianjin 300170, China
| | | | | | | | | | | |
Collapse
|
34
|
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
|
35
|
White CR, Datta G, Giordano S. High-Density Lipoprotein Regulation of Mitochondrial Function. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 982:407-429. [PMID: 28551800 DOI: 10.1007/978-3-319-55330-6_22] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Lipoproteins play a key role in regulating plasma and tissue levels of cholesterol. Apolipoprotein B (apoB)-containing lipoproteins, including chylomicrons, very-low density lipoprotein (VLDL) and low-density lipoprotein (LDL), serve as carriers of triglycerides and cholesterol and deliver these metabolites to peripheral tissues. In contrast, high-density lipoprotein (HDL) mediates Reverse Cholesterol Transport (RCT), a process by which excess cholesterol is removed from the periphery and taken up by hepatocytes where it is metabolized and excreted. Anti-atherogenic properties of HDL have been largely ascribed to apoA-I, the major protein component of the lipoprotein particle. The inflammatory response associated with atherosclerosis and ischemia-reperfusion (I-R) injury has been linked to the development of mitochondrial dysfunction. Under these conditions, an increase in reactive oxygen species (ROS) formation induces damage to mitochondrial structural elements, leading to a reduction in ATP synthesis and initiation of the apoptotic program. Recent studies suggest that HDL-associated apoA-I and lysosphingolipids attenuate mitochondrial injury by multiple mechanisms, including the suppression of ROS formation and induction of autophagy. Other apolipoproteins, however, present in lower abundance in HDL particles may exert opposing effects on mitochondrial function. This chapter examines the role of HDL-associated apolipoproteins and lipids in the regulation of mitochondrial function and bioenergetics.
Collapse
Affiliation(s)
- C Roger White
- Department of Medicine, Division of Cardiovascular Disease, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Geeta Datta
- Department of Medicine, Division of Cardiovascular Disease, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Samantha Giordano
- Department of Medicine, Division of Cardiovascular Disease, University of Alabama at Birmingham, Birmingham, AL, USA.
| |
Collapse
|
36
|
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: 31] [Impact Index Per Article: 3.4] [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
|
37
|
White CR, Giordano S, Anantharamaiah GM. High-density lipoprotein, mitochondrial dysfunction and cell survival mechanisms. Chem Phys Lipids 2016; 199:161-169. [PMID: 27150975 DOI: 10.1016/j.chemphyslip.2016.04.007] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2016] [Revised: 04/22/2016] [Accepted: 04/23/2016] [Indexed: 01/08/2023]
Abstract
Ischemic injury is associated with acute myocardial infarction, percutaneous coronary intervention, coronary artery bypass grafting and open heart surgery. The timely re-establishment of blood flow is critical in order to minimize cardiac complications. Reperfusion after a prolonged ischemic period, however, can induce severe cardiomyocyte dysfunction with mitochondria serving as a major target of ischemia/reperfusion (I/R) injury. An increase in the formation of reactive oxygen species (ROS) induces damage to mitochondrial respiratory complexes leading to uncoupling of oxidative phosphorylation. Mitochondrial membrane perturbations also contribute to calcium overload, opening of the mitochondrial permeability transition pore (mPTP) and the release of apoptotic mediators into the cytoplasm. Clinical and experimental studies show that ischemic preconditioning (ICPRE) and postconditioning (ICPOST) attenuate mitochondrial injury and improve cardiac function in the context of I/R injury. This is achieved by the activation of two principal cell survival cascades: 1) the Reperfusion Injury Salvage Kinase (RISK) pathway; and 2) the Survivor Activating Factor Enhancement (SAFE) pathway. Recent data suggest that high density lipoprotein (HDL) mimics the effects of conditioning protocols and attenuates myocardial I/R injury via activation of the RISK and SAFE signaling cascades. In this review, we discuss the roles of apolipoproteinA-I (apoA-I), the major protein constituent of HDL, and sphingosine 1-phosphate (S1P), a lysosphingolipid associated with small, dense HDL particles as mediators of cardiomyocyte survival. Both apoA-I and S1P exert an infarct-sparing effect by preventing ROS-dependent injury and inhibiting the opening of the mPTP.
Collapse
Affiliation(s)
- C Roger White
- Department of Medicine, Division of Cardiovascular Disease, University of Alabama at Birmingham, Birmingham, AL, USA.
| | - Samantha Giordano
- Department of Medicine, Division of Cardiovascular Disease, University of Alabama at Birmingham, Birmingham, AL, USA
| | - G M Anantharamaiah
- The Division of Gerontology, Geriatric Medicine and Palliative Care, University of Alabama at Birmingham, Birmingham, AL, USA; Biochemistry and Molecular Genetics, University of Alabama at Birmingham, Birmingham, AL, USA
| |
Collapse
|
38
|
Frati A, Ricci B, Pierucci F, Nistri S, Bani D, Meacci E. Role of sphingosine kinase/S1P axis in ECM remodeling of cardiac cells elicited by relaxin. Mol Endocrinol 2016; 29:53-67. [PMID: 25415609 DOI: 10.1210/me.2014-1201] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
The initiation and progression of heart failure is linked to adverse cardiac remodeling of the extracellular matrix (ECM) during disease mainly through the deregulation of myocardial metalloproteinases (MMPs). Relaxin (RLX), a peptide hormone acting as a physiological cardiac effector, is a key regulator of ECM remodeling in reproductive and nonreproductive tissues. Studying primary cultures of mouse cardiac muscle cells and rat H9c2 cardiomyoblasts, we have obtained evidence for a new signaling pathway activated by RLX to induce ECM remodeling that involves the bioactive sphingolipids sphingosine-1-phosphate (S1P) and ceramide. In both cell populations, recombinant human RLX increased sphingosine kinase activity and S1P formation, whereas sphingomyelin and ceramide content were decreased in [(3)H]serine-labeled cells. According to the literature, RLX promoted MMP-2 and MMP-9 expression/release. Pharmacological inhibition of sphingolipid metabolism and silencing of sphingosine kinase 1, the enzyme responsible for S1P formation, were able to prevent MMP expression/release elicited by the hormone and induce the expression of tissue inhibitor of MMPs. In addition, we found that sphingolipid signaling is required for the regulation of connective tissue growth factor, a member of the CCN 1-3 family of genes that are involved in cell proliferation and differentiation. Finally, the induction of cardiomyoblast maturation induced by RLX was also found to be counteracted by inhibition of S1P formation. In conclusion, these findings provide a novel mechanism by which RLX acts on cardiac ECM remodeling and cardiac cell differentiation and offer interesting therapeutic options to prevent heart fibrosis and to favor myocardial regeneration.
Collapse
Affiliation(s)
- Alessia Frati
- Department of Biomedical, Experimental, and Clinical Sciences (A.F., B.R., F.P., E.M.), Research Unit of Biochemistry, and Department of Experimental and Clinical Medicine (S.N., D.B.), Research Unit of Histology and Embryology, University of Florence, 50134 Florence, Italy
| | | | | | | | | | | |
Collapse
|
39
|
Pyne S, Adams DR, Pyne NJ. Sphingosine 1-phosphate and sphingosine kinases in health and disease: Recent advances. Prog Lipid Res 2016; 62:93-106. [PMID: 26970273 DOI: 10.1016/j.plipres.2016.03.001] [Citation(s) in RCA: 146] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Revised: 03/07/2016] [Accepted: 03/08/2016] [Indexed: 12/24/2022]
Abstract
Sphingosine kinases (isoforms SK1 and SK2) catalyse the formation of a bioactive lipid, sphingosine 1-phosphate (S1P). S1P is a well-established ligand of a family of five S1P-specific G protein coupled receptors but also has intracellular signalling roles. There is substantial evidence to support a role for sphingosine kinases and S1P in health and disease. This review summarises recent advances in the area in relation to receptor-mediated signalling by S1P and novel intracellular targets of this lipid. New evidence for a role of each sphingosine kinase isoform in cancer, the cardiovascular system, central nervous system, inflammation and diabetes is discussed. There is continued research to develop isoform selective SK inhibitors, summarised here. Analysis of the crystal structure of SK1 with the SK1-selective inhibitor, PF-543, is used to identify residues that could be exploited to improve selectivity in SK inhibitor development for future therapeutic application.
Collapse
Affiliation(s)
- Susan Pyne
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral St, Glasgow, G4 0RE, Scotland, UK.
| | - David R Adams
- School of Engineering & Physical Sciences, Heriot-Watt University, Edinburgh EH14 4AS, Scotland, UK.
| | - Nigel J Pyne
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral St, Glasgow, G4 0RE, Scotland, UK.
| |
Collapse
|
40
|
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: 16.7] [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
|
41
|
Inhibition of ceramide de novo synthesis as a postischemic strategy to reduce myocardial reperfusion injury. Basic Res Cardiol 2016; 111:12. [PMID: 26786259 DOI: 10.1007/s00395-016-0533-x] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2015] [Accepted: 01/11/2016] [Indexed: 12/17/2022]
Abstract
The injury caused by myocardial reperfusion after ischemia can be contained by interventions aimed at reducing the inflammation and the oxidative stress that underlie exacerbation of tissue damage. Sphingolipids are a class of structural and signaling lipid molecules; among them, the inflammation mediator ceramide accumulates in the myocardium upon ischemia/reperfusion. Here, we show that, after transient coronary occlusion in mice, an increased de novo ceramide synthesis takes place at reperfusion in the ischemic area surrounding necrosis (area at risk). This correlates with the enhanced expression of the first and rate-limiting enzyme of the de novo pathway, serine palmitoyltransferase (SPT). The intraventricular administration at reperfusion of myriocin, an inhibitor of SPT, significantly protected the area at risk from damage, reducing the infarcted area by 40.9 % relative to controls not treated with the drug. In the area at risk, myriocin downregulated ceramide, reduced the content in other mediators of inflammation and reactive oxygen species, and activated the Nrf2-HO1 cytoprotective response. We conclude that an enhanced ceramide synthesis takes part in ischemia/reperfusion injury and that myriocin treatment can be proposed as a strategy for myocardial pharmacological postconditioning.
Collapse
|
42
|
The SR/ER-mitochondria calcium crosstalk is regulated by GSK3β during reperfusion injury. Cell Death Differ 2015. [PMID: 26206086 DOI: 10.1038/cdd.2015.101] [Citation(s) in RCA: 86] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Glycogen synthase kinase-3β (GSK3β) is a multifunctional kinase whose inhibition is known to limit myocardial ischemia-reperfusion injury. However, the mechanism mediating this beneficial effect still remains unclear. Mitochondria and sarco/endoplasmic reticulum (SR/ER) are key players in cell death signaling. Their involvement in myocardial ischemia-reperfusion injury has gained recognition recently, but the underlying mechanisms are not yet well understood. We questioned here whether GSK3β might have a role in the Ca(2+) transfer from SR/ER to mitochondria at reperfusion. We showed that a fraction of GSK3β protein is localized to the SR/ER and mitochondria-associated ER membranes (MAMs) in the heart, and that GSK3β specifically interacted with the inositol 1,4,5-trisphosphate receptors (IP3Rs) Ca(2+) channeling complex in MAMs. We demonstrated that both pharmacological and genetic inhibition of GSK3β decreased protein interaction of IP3R with the Ca(2+) channeling complex, impaired SR/ER Ca(2+) release and reduced the histamine-stimulated Ca(2+) exchange between SR/ER and mitochondria in cardiomyocytes. During hypoxia reoxygenation, cell death is associated with an increase of GSK3β activity and IP3R phosphorylation, which leads to enhanced transfer of Ca(2+) from SR/ER to mitochondria. Inhibition of GSK3β at reperfusion reduced both IP3R phosphorylation and SR/ER Ca(2+) release, which consequently diminished both cytosolic and mitochondrial Ca(2+) concentrations, as well as sensitivity to apoptosis. We conclude that inhibition of GSK3β at reperfusion diminishes Ca(2+) leak from IP3R at MAMs in the heart, which limits both cytosolic and mitochondrial Ca(2+) overload and subsequent cell death.
Collapse
|
43
|
Ali F, Khan AQ, Khan R, Sultana S. Trichloroethylene-mediated cytotoxicity in human epidermal keratinocytes is mediated by the rapid accumulation of intracellular calcium: Interception by naringenin. Hum Exp Toxicol 2015; 35:147-61. [PMID: 25855085 DOI: 10.1177/0960327115578865] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Industrial solvents pose a significant threat to the humankind. The mechanisms of their toxicity still remain in debate. Trichloroethylene (TCE) is a widespread industrial solvent responsible for severe liver dysfunction, cutaneous toxicity in occupationally exposed humans. We utilized an in vitro system of human epidermal keratinocyte (HaCaT) cells in this study to avoid complex cell and extracellular interactions. We report the cytotoxicity of organic solvent TCE in HaCaT and its reversal by a natural flavanone, naringenin (Nar). The cytotoxicity was attributed to the rapid intracellular free calcium (Ca(2+)) release, which might lead to the elevation of protein kinase C along with robust free radical generation, instability due to energy depletion, and sensitization of intracellular stress signal transducer nuclear factor κB. These effects were actually seen to induce significant amount of genomic DNA fragmentation. Furthermore, all these effects of TCE were effectively reversed by the treatment of Nar, a natural flavanone. Our studies identify intracellular Ca as a unique target used by organic solvents in the cytotoxicity and highlight the Ca(2+) ion stabilizer properties of Nar.
Collapse
Affiliation(s)
- F Ali
- Section of Molecular Carcinogenesis and Chemoprevention, Department of Medical Elementology and Toxicology, Faculty of Science, Hamdard University, Jamia Hamdard, New Delhi, India
| | - A Q Khan
- Section of Molecular Carcinogenesis and Chemoprevention, Department of Medical Elementology and Toxicology, Faculty of Science, Hamdard University, Jamia Hamdard, New Delhi, India
| | - R Khan
- Section of Molecular Carcinogenesis and Chemoprevention, Department of Medical Elementology and Toxicology, Faculty of Science, Hamdard University, Jamia Hamdard, New Delhi, India
| | - S Sultana
- Section of Molecular Carcinogenesis and Chemoprevention, Department of Medical Elementology and Toxicology, Faculty of Science, Hamdard University, Jamia Hamdard, New Delhi, India
| |
Collapse
|
44
|
Abstract
Reperfusion is mandatory to salvage ischemic myocardium from infarction, but reperfusion per se contributes to injury and ultimate infarct size. Therefore, cardioprotection beyond that by timely reperfusion is needed to reduce infarct size and improve the prognosis of patients with acute myocardial infarction. The conditioning phenomena provide such cardioprotection, insofar as brief episodes of coronary occlusion/reperfusion preceding (ischemic preconditioning) or following (ischemic postconditioning) sustained myocardial ischemia with reperfusion reduce infarct size. Even ischemia/reperfusion in organs remote from the heart provides cardioprotection (remote ischemic conditioning). The present review characterizes the signal transduction underlying the conditioning phenomena, including their physical and chemical triggers, intracellular signal transduction, and effector mechanisms, notably in the mitochondria. Cardioprotective signal transduction appears as a highly concerted spatiotemporal program. Although the translation of ischemic postconditioning and remote ischemic conditioning protocols to patients with acute myocardial infarction has been fairly successful, the pharmacological recruitment of cardioprotective signaling has been largely disappointing to date.
Collapse
Affiliation(s)
- Gerd Heusch
- From the Institute for Pathophysiology, West German Heart and Vascular Centre, University of Essen Medical School, Essen, Germany.
| |
Collapse
|
45
|
Bajwa A, Rosin DL, Chroscicki P, Lee S, Dondeti K, Ye H, Kinsey GR, Stevens BK, Jobin K, Kenwood BM, Hoehn KL, Lynch KR, Okusa MD. Sphingosine 1-phosphate receptor-1 enhances mitochondrial function and reduces cisplatin-induced tubule injury. J Am Soc Nephrol 2014; 26:908-25. [PMID: 25145931 DOI: 10.1681/asn.2013121351] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Sphingosine 1-phosphate (S1P), the natural sphingolipid ligand for a family of five G protein- coupled receptors (S1P1-S1P5Rs), regulates cell survival and lymphocyte circulation. We have shown that the pan-S1PR agonist, FTY720, attenuates kidney ischemia-reperfusion injury by directly activating S1P1 on proximal tubule (PT) cells, independent of the canonical lymphopenic effects of S1P1 activation on B and T cells. FTY720 also reduces cisplatin-induced AKI. Therefore, in this study, we used conditional PT-S1P1-null (PepckCreS1pr1(fl/fl)) and control (PepckCreS1pr1(w/wt)) mice to determine whether the protective effect of FTY720 in AKI is mediated by PT-S1P1. Cisplatin induced more renal injury in PT-S1P1-null mice than in controls. Although FTY720 produced lymphopenia in both control and PT-S1P1-null mice, it reduced injury only in control mice. Furthermore, the increase in proinflammatory cytokine (CXCL1, MCP-1, TNF-α, and IL-6) expression and infiltration of neutrophils and macrophages induced by cisplatin treatment was attenuated by FTY720 in control mice but not in PT-S1P1-null mice. Similarly, S1P1 deletion rendered cultured PT cells more susceptible to cisplatin-induced injury, whereas S1P1 overexpression protected PT cells from injury and preserved mitochondrial function. We conclude that S1P1 may have an important role in stabilizing mitochondrial function and that FTY720 administration represents a novel strategy in the prevention of cisplatin-induced AKI.
Collapse
Affiliation(s)
- Amandeep Bajwa
- Departments of Medicine and Center for Immunity, Inflammation, and Regenerative Medicine, University of Virginia School of Medicine, Charlottesville, Virginia
| | - Diane L Rosin
- Center for Immunity, Inflammation, and Regenerative Medicine, University of Virginia School of Medicine, Charlottesville, Virginia Pharmacology, and
| | - Piotr Chroscicki
- Departments of Medicine and Center for Immunity, Inflammation, and Regenerative Medicine, University of Virginia School of Medicine, Charlottesville, Virginia
| | - Sangju Lee
- Departments of Medicine and Center for Immunity, Inflammation, and Regenerative Medicine, University of Virginia School of Medicine, Charlottesville, Virginia
| | - Krishna Dondeti
- Departments of Medicine and Center for Immunity, Inflammation, and Regenerative Medicine, University of Virginia School of Medicine, Charlottesville, Virginia
| | - Hong Ye
- Departments of Medicine and Center for Immunity, Inflammation, and Regenerative Medicine, University of Virginia School of Medicine, Charlottesville, Virginia
| | - Gilbert R Kinsey
- Departments of Medicine and Center for Immunity, Inflammation, and Regenerative Medicine, University of Virginia School of Medicine, Charlottesville, Virginia
| | - Brian K Stevens
- Departments of Medicine and Center for Immunity, Inflammation, and Regenerative Medicine, University of Virginia School of Medicine, Charlottesville, Virginia
| | - Katarzyna Jobin
- Departments of Medicine and Center for Immunity, Inflammation, and Regenerative Medicine, University of Virginia School of Medicine, Charlottesville, Virginia
| | | | | | | | - Mark D Okusa
- Departments of Medicine and Center for Immunity, Inflammation, and Regenerative Medicine, University of Virginia School of Medicine, Charlottesville, Virginia
| |
Collapse
|
46
|
Chowdhury I, Thompson WE, Thomas K. Prohibitins role in cellular survival through Ras-Raf-MEK-ERK pathway. J Cell Physiol 2014; 229:998-1004. [PMID: 24347342 DOI: 10.1002/jcp.24531] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2013] [Accepted: 12/11/2013] [Indexed: 12/15/2022]
Abstract
Prohibitins are members of a highly conserved protein family containing the stomatin/prohibitin/flotillin/HflK/C (SPFH) domain (also known as the prohibitin [PHB] domain) found in unicellular eukaryotes, fungi, plants, animals, and humans. Two highly homologous members of prohibitins expressed in eukaryotes are prohibitin (PHB; B-cell receptor associated protein-32, BAP-32) and prohibitin 2/repressor of estrogen receptor activity (PHB2, REA, BAP-37). Both PHB and REA/PHB2 are ubiquitously expressed and are present in multiple cellular compartments including the mitochondria, nucleus, and the plasma membrane. Multiple functions have been attributed to the mitochondrial and nuclear PHB and PHB2/REA including cellular differentiation, anti-proliferation, and morphogenesis. One of the major functions of the prohibitins are in maintaining the functional integrity of the mitochondria and protecting cells from various stresses. In the present review, we focus on the recent research developments indicating that PHB and PHB2/REA are involved in maintaining cellular survival through the Ras-Raf-MEK-Erk pathway. Understanding the molecular mechanisms by which the intracellular signaling pathways utilize prohibitins in governing cellular survival is likely to result in development of therapeutic strategies to overcome various human pathological disorders such as diabetes, obesity, neurological diseases, inflammatory bowel disease, and cancer.
Collapse
Affiliation(s)
- Indrajit Chowdhury
- Department of Obstetrics and Gynecology, Morehouse School of Medicine, Atlanta, Georgia; Reproductive Science Research Program, Morehouse School of Medicine, Atlanta, Georgia
| | | | | |
Collapse
|
47
|
Yan W, Zhang F, Zhang R, Zhang X, Wang Y, Zhou F, Xia Y, Liu P, Gao C, Wang H, Zhang L, Zhou J, Gao F, Gao E, Koch WJ, Wang H, Cheng H, Qu Y, Tao L. Adiponectin regulates SR Ca(2+) cycling following ischemia/reperfusion via sphingosine 1-phosphate-CaMKII signaling in mice. J Mol Cell Cardiol 2014; 74:183-92. [PMID: 24852843 DOI: 10.1016/j.yjmcc.2014.05.010] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2013] [Revised: 04/15/2014] [Accepted: 05/08/2014] [Indexed: 01/01/2023]
Abstract
The adipocyte-secreted hormone adiponectin (APN) exerts protective effects on the heart under stress conditions. Recent studies have demonstrated that APN induces a marked Ca(2+) influx in skeletal muscle. However, whether APN modulates [Ca(2+)]i activity, especially [Ca(2+)]i transients in cardiomyocytes, is still unknown. This study was designed to determine whether APN modulates [Ca(2+)]i transients in cardiomyocytes. Adult male wild-type (WT) and APN knockout (APN KO) mice were subjected to myocardial ischemia/reperfusion (I/R, 30min/30min) injury. CaMKII-PLB phosphorylation and SR Ca(2+)-ATPase (SERCA2) activity were downregulated in I/R hearts of WT mice and further decreased in those of APN KO mice. Both the globular domain of APN and full-length APN significantly reversed the decrease in CaMKII-PLB phosphorylation and SERCA2 activity in WT and APN KO mice. Interestingly, compared with WT littermates, single myocytes isolated from APN KO mice had remarkably decreased [Ca(2+)]i transients, cell shortening, and a prolonged Ca(2+) decay rate. Further examination revealed that APN enhances SERCA2 activity via CaMKII-PLB signaling. In in vivo and in vitro experiments, both APN receptor 1/2 and S1P were necessary for the APN-stimulated CaMKII-PLB-SERCA2 activation. In addition, S1P activated CaMKII-PLB signaling in neonatal cardiomyocytes in a dose dependent manner and improved [Ca(2+)]i transients in APN KO myocytes via the S1P receptor (S1PR1/3). Further in vivo experiments revealed that pharmacological inhibition of S1PR1/3 and SERCA2 siRNA suppressed APN-mediated cardioprotection during I/R. These data demonstrate that S1P is a novel regulator of SERCA2 that activates CaMKII-PLB signaling and mediates APN-induced cardioprotection.
Collapse
Affiliation(s)
- Wenjun Yan
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Fuyang Zhang
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Ronghuai Zhang
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Xing Zhang
- Department of Physiology, School of Basic Medical Sciences, Fourth Military Medical University, Xi'an 710032, China
| | - Yanru Wang
- Institutes of Molecular Medicine, Peking University, Beijing 100083, China
| | - Fen Zhou
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Yunlong Xia
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Peilin Liu
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Chao Gao
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Han Wang
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Lijian Zhang
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Jingjun Zhou
- Department of Physiology, School of Basic Medical Sciences, Fourth Military Medical University, Xi'an 710032, China
| | - Feng Gao
- Department of Physiology, School of Basic Medical Sciences, Fourth Military Medical University, Xi'an 710032, China
| | - Erhe Gao
- Center for Translational Medicine, School of Medicine, Temple University, Philadelphia, PA 19107, USA
| | - Walter J Koch
- Center for Translational Medicine, School of Medicine, Temple University, Philadelphia, PA 19107, USA
| | - Haichang Wang
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Heping Cheng
- Institutes of Molecular Medicine, Peking University, Beijing 100083, China
| | - Yan Qu
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China.
| | - Ling Tao
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China.
| |
Collapse
|
48
|
Xu A, Szczepanek K, Maceyka MW, Ross T, Bowler E, Hu Y, Kenny B, Mehfoud C, Desai PN, Baumgarten CM, Chen Q, Lesnefsky EJ. Transient complex I inhibition at the onset of reperfusion by extracellular acidification decreases cardiac injury. Am J Physiol Cell Physiol 2014; 306:C1142-53. [PMID: 24696146 DOI: 10.1152/ajpcell.00241.2013] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
A reversible inhibition of mitochondrial respiration by complex I inhibition at the onset of reperfusion decreases injury in buffer-perfused hearts. Administration of acidic reperfusate for a brief period at reperfusion decreases cardiac injury. We asked if acidification treatment decreased cardiac injury during reperfusion by inhibiting complex I. Exposure of isolated mouse heart mitochondria to acidic buffer decreased the complex I substrate-stimulated respiration, whereas respiration with complex II substrates was unaltered. Evidence of the rapid and reversible inhibition of complex I by an acidic environment was obtained at the level of isolated complex, intact mitochondria and in situ mitochondria in digitonin-permeabilized cardiac myocytes. Moreover, ischemia-damaged complex I was also reversibly inhibited by an acidic environment. In the buffer-perfused mouse heart, reperfusion with pH 6.6 buffer for the initial 5 min decreased infarction. Compared with untreated hearts, acidification treatment markedly decreased the mitochondrial generation of reactive oxygen species and improved mitochondrial calcium retention capacity and inner mitochondrial membrane integrity. The decrease in infarct size achieved by acidic reperfusion approximates the reduction obtained by a reversible, partial blockade of complex I at reperfusion. Extracellular acidification decreases cardiac injury during reperfusion in part via the transient and reversible inhibition of complex I, leading to a reduction of oxyradical generation accompanied by a decreased susceptibility to mitochondrial permeability transition during early reperfusion.
Collapse
Affiliation(s)
- Aijun Xu
- Department of Medicine, Division of Cardiology, Pauley Heart Center, Virginia Commonwealth University School of Medicine, Richmond, Virginia; Department of Anesthesiology, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China; and
| | - Karol Szczepanek
- Department of Medicine, Division of Cardiology, Pauley Heart Center, Virginia Commonwealth University School of Medicine, Richmond, Virginia
| | - Michael W Maceyka
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University School of Medicine, Richmond, Virginia
| | - Thomas Ross
- Department of Medicine, Division of Cardiology, Pauley Heart Center, Virginia Commonwealth University School of Medicine, Richmond, Virginia
| | - Elizabeth Bowler
- Department of Medicine, Division of Cardiology, Pauley Heart Center, Virginia Commonwealth University School of Medicine, Richmond, Virginia; University of the West of England, Bristol, United Kingdom
| | - Ying Hu
- Department of Medicine, Division of Cardiology, Pauley Heart Center, Virginia Commonwealth University School of Medicine, Richmond, Virginia
| | - Barrett Kenny
- Department of Medicine, Division of Cardiology, Pauley Heart Center, Virginia Commonwealth University School of Medicine, Richmond, Virginia
| | - Chris Mehfoud
- Department of Medicine, Division of Cardiology, Pauley Heart Center, Virginia Commonwealth University School of Medicine, Richmond, Virginia
| | - Pooja N Desai
- Department of Physiology and Biophysics, Virginia Commonwealth University School of Medicine, Richmond, Virginia
| | - Clive M Baumgarten
- Department of Medicine, Division of Cardiology, Pauley Heart Center, Virginia Commonwealth University School of Medicine, Richmond, Virginia; Department of Physiology and Biophysics, Virginia Commonwealth University School of Medicine, Richmond, Virginia
| | - Qun Chen
- Department of Medicine, Division of Cardiology, Pauley Heart Center, Virginia Commonwealth University School of Medicine, Richmond, Virginia
| | - Edward J Lesnefsky
- Department of Medicine, Division of Cardiology, Pauley Heart Center, Virginia Commonwealth University School of Medicine, Richmond, Virginia; Department of Biochemistry and Molecular Biology, Virginia Commonwealth University School of Medicine, Richmond, Virginia; Department of Physiology and Biophysics, Virginia Commonwealth University School of Medicine, Richmond, Virginia; McGuire Veterans Affairs Medical Center, Richmond, Virginia;
| |
Collapse
|
49
|
Wallington-Beddoe CT, Powell JA, Tong D, Pitson SM, Bradstock KF, Bendall LJ. Sphingosine kinase 2 promotes acute lymphoblastic leukemia by enhancing MYC expression. Cancer Res 2014; 74:2803-15. [PMID: 24686171 DOI: 10.1158/0008-5472.can-13-2732] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Sphingosine kinase 2 (SK2) may have utility as a prognostic marker in inflammatory diseases such as cancer in which it has been rationalized as a candidate therapeutic target. Here, we show that SK2 has an oncogenic role in acute lymphoblastic leukemia (ALL) by influencing expression of MYC. Genetic ablation of SK2 impaired leukemia development in a mouse model of ALL and pharmacologic inhibition extended survival in mouse xenograft models of human disease. SK2 attenuation in both the settings reduced MYC expression in leukemic cells, with reduced levels of acetylated histone H3 within the MYC gene associated with reduced levels of MYC protein and expression of MYC-regulated genes. Our results demonstrated that SK2 regulates MYC, which has a pivotal role in hematologic malignancies, providing a preclinical proof of concept for this pathway as a broad-based therapeutic target in this setting.
Collapse
Affiliation(s)
- Craig T Wallington-Beddoe
- Authors' Affiliations: Westmead Institute for Cancer Research, Westmead Millennium Institute, University of Sydney; Hematology Department, Westmead Hospital, Westmead, Sydney, New South Wales; and Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, South Australia, Australia
| | - Jason A Powell
- Authors' Affiliations: Westmead Institute for Cancer Research, Westmead Millennium Institute, University of Sydney; Hematology Department, Westmead Hospital, Westmead, Sydney, New South Wales; and Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, South Australia, Australia
| | - Daochen Tong
- Authors' Affiliations: Westmead Institute for Cancer Research, Westmead Millennium Institute, University of Sydney; Hematology Department, Westmead Hospital, Westmead, Sydney, New South Wales; and Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, South Australia, Australia
| | - Stuart M Pitson
- Authors' Affiliations: Westmead Institute for Cancer Research, Westmead Millennium Institute, University of Sydney; Hematology Department, Westmead Hospital, Westmead, Sydney, New South Wales; and Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, South Australia, Australia
| | - Kenneth F Bradstock
- Authors' Affiliations: Westmead Institute for Cancer Research, Westmead Millennium Institute, University of Sydney; Hematology Department, Westmead Hospital, Westmead, Sydney, New South Wales; and Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, South Australia, Australia
| | - Linda J Bendall
- Authors' Affiliations: Westmead Institute for Cancer Research, Westmead Millennium Institute, University of Sydney; Hematology Department, Westmead Hospital, Westmead, Sydney, New South Wales; and Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, South Australia, Australia
| |
Collapse
|
50
|
Waeber C, Walther T. Sphingosine-1-phosphate as a potential target for the treatment of myocardial infarction. Circ J 2014; 78:795-802. [PMID: 24632793 DOI: 10.1253/circj.cj-14-0178] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
This review focuses on the role of sphingosine-1-phosphate (S1P) signaling in the heart, with particular emphasis on how it could be modulated therapeutically in the context of myocardial infarction (MI). After a brief general description of sphingolipid metabolism and signaling, this review will examine the relationship between S1P and the beneficial effects of high-density lipoprotein (HDL), and finally focus on the known actions of S1P on different mechanisms relevant to MI pathophysiology (cardiomyocyte protection, fibrosis, remodeling, arrhythmia, control of vascular tone and potential repair mechanisms). The potential of particular enzyme isoforms or receptor subtypes for the development of therapeutic agents for MI will also be explored.
Collapse
Affiliation(s)
- Christian Waeber
- Department of Pharmacology and Therapeutics, School of Medicine, School of Pharmacy, University College Cork
| | | |
Collapse
|