1
|
Hagemann N, Qi Y, Mohamud Yusuf A, Li A, Squire A, Tertel T, Giebel B, Ludewig P, Spangenberg P, Chen J, Mosig A, Gunzer M, Hermann DM. Microvascular Network Remodeling in the Ischemic Mouse Brain Defined by Light Sheet Microscopy. Arterioscler Thromb Vasc Biol 2024; 44:915-929. [PMID: 38357819 DOI: 10.1161/atvbaha.123.320339] [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: 10/27/2023] [Accepted: 02/01/2024] [Indexed: 02/16/2024]
Abstract
BACKGROUND Until now, the analysis of microvascular networks in the reperfused ischemic brain has been limited due to tissue transparency challenges. METHODS Using light sheet microscopy, we assessed microvascular network remodeling in the striatum from 3 hours to 56 days post-ischemia in 2 mouse models of transient middle cerebral artery occlusion lasting 20 or 40 minutes, resulting in mild ischemic brain injury or brain infarction, respectively. We also examined the effect of a clinically applicable S1P (sphingosine-1-phosphate) analog, FTY720 (fingolimod), on microvascular network remodeling. RESULTS Over 56 days, we observed progressive microvascular degeneration in the reperfused striatum, that is, the lesion core, which was followed by robust angiogenesis after mild ischemic injury induced by 20-minute middle cerebral artery occlusion. However, more severe ischemic injury elicited by 40-minute middle cerebral artery occlusion resulted in incomplete microvascular remodeling. In both cases, microvascular networks did not return to their preischemic state but displayed a chronically altered pattern characterized by higher branching point density, shorter branches, higher unconnected branch density, and lower tortuosity, indicating enhanced network connectivity. FTY720 effectively increased microvascular length density, branching point density, and volume density in both models, indicating an angiogenic effect of this drug. CONCLUSIONS Utilizing light sheet microscopy together with automated image analysis, we characterized microvascular remodeling in the ischemic lesion core in unprecedented detail. This technology will significantly advance our understanding of microvascular restorative processes and pave the way for novel treatment developments in the stroke field.
Collapse
Affiliation(s)
- Nina Hagemann
- Department of Neurology and Center for Translational Neuro- and Behavioral Sciences, University Hospital Essen, University of Duisburg-Essen, Germany (N.H., Y.Q., A.M.Y., A.L., D.M.H.)
| | - Yachao Qi
- Department of Neurology and Center for Translational Neuro- and Behavioral Sciences, University Hospital Essen, University of Duisburg-Essen, Germany (N.H., Y.Q., A.M.Y., A.L., D.M.H.)
| | - Ayan Mohamud Yusuf
- Department of Neurology and Center for Translational Neuro- and Behavioral Sciences, University Hospital Essen, University of Duisburg-Essen, Germany (N.H., Y.Q., A.M.Y., A.L., D.M.H.)
| | - AnRan Li
- Department of Neurology and Center for Translational Neuro- and Behavioral Sciences, University Hospital Essen, University of Duisburg-Essen, Germany (N.H., Y.Q., A.M.Y., A.L., D.M.H.)
| | - Anthony Squire
- Institute for Experimental Immunology and Imaging and Imaging Center Essen, University Hospital Essen, University of Duisburg-Essen, Germany (A.S., P.S., M.G.)
| | - Tobias Tertel
- Institute for Transfusion Medicine, University Hospital Essen, University of Duisburg-Essen, Germany (T.T., B.G.)
| | - Bernd Giebel
- Institute for Transfusion Medicine, University Hospital Essen, University of Duisburg-Essen, Germany (T.T., B.G.)
| | - Peter Ludewig
- Department of Neurology, University Medical Center Hamburg-Eppendorf, Germany (P.L.)
| | - Philippa Spangenberg
- Institute for Experimental Immunology and Imaging and Imaging Center Essen, University Hospital Essen, University of Duisburg-Essen, Germany (A.S., P.S., M.G.)
| | - Jianxu Chen
- Leibniz-Institut für Analytische Wissenschaften-ISAS-e.V., Dortmund, Germany (J.C., M.G.)
| | - Axel Mosig
- Bioinformatics Group, Faculty for Biology and Biotechnology and Center for Protein Diagnostics, Ruhr-University Bochum, Germany (A.M.)
| | - Matthias Gunzer
- Institute for Experimental Immunology and Imaging and Imaging Center Essen, University Hospital Essen, University of Duisburg-Essen, Germany (A.S., P.S., M.G.)
- Leibniz-Institut für Analytische Wissenschaften-ISAS-e.V., Dortmund, Germany (J.C., M.G.)
| | - Dirk M Hermann
- Department of Neurology and Center for Translational Neuro- and Behavioral Sciences, University Hospital Essen, University of Duisburg-Essen, Germany (N.H., Y.Q., A.M.Y., A.L., D.M.H.)
| |
Collapse
|
2
|
Constantinescu V, Haase R, Akgün K, Ziemssen T. S1P receptor modulators and the cardiovascular autonomic nervous system in multiple sclerosis: a narrative review. Ther Adv Neurol Disord 2022; 15:17562864221133163. [PMID: 36437849 PMCID: PMC9685213 DOI: 10.1177/17562864221133163] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 09/29/2022] [Indexed: 01/21/2024] Open
Abstract
UNLABELLED Sphingosine 1-phosphate (S1P) receptor (S1PR) modulators have a complex mechanism of action, which are among the most efficient therapeutic options in multiple sclerosis (MS) and represent a promising approach for other immune-mediated diseases. The S1P signaling pathway involves the activation of five extracellular S1PR subtypes (S1PR1-S1PR5) that are ubiquitous and have a wide range of effects. Besides the immunomodulatory beneficial outcome in MS, S1P signaling regulates the cardiovascular function via S1PR1-S1PR3 subtypes, which reside on cardiac myocytes, endothelial, and vascular smooth muscle cells. In our review, we describe the mechanisms and clinical effects of S1PR modulators on the cardiovascular system. In the past, mostly short-term effects of S1PR modulators on the cardiovascular system have been studied, while data on long-term effects still need to be investigated. Immediate effects detected after treatment initiation are due to parasympathetic overactivation. In contrast, long-term effects may arise from a shift of the autonomic regulation toward sympathetic predominance along with S1PR1 downregulation. A mild increase in blood pressure has been reported in long-term studies, as well as decreased baroreflex sensitivity. In most studies, sustained hypertension was found to represent a significant adverse event related to treatment. The shift in the autonomic control and blood pressure values could not be just a consequence of disease progression but also related to S1PR modulation. Reduced cardiac autonomic activation and decreased heart rate variability during the long-term treatment with S1PR modulators may increase the risk for subsequent cardiac events. For second-generation S1PR modulators, this observation has to be confirmed in further studies with longer follow-ups. The periodic surveillance of cardiovascular function and detection of any cardiac autonomic dysfunction can help predict cardiac outcomes not only after the first dose but also throughout treatment. PLAIN LANGUAGE SUMMARY What is the cardiovascular effect of S1P receptor modulator therapy in multiple sclerosis? Sphingosine 1-phosphate (S1P) receptor (S1PR) modulators are among the most efficient therapies for multiple sclerosis. As small molecules, they are not only acting on the immune but on cardiovascular and nervous systems as well. Short-term effects of S1PR modulators on the cardiovascular system have already been extensively described, while long-term effects are less known. Our review describes the mechanisms of action and the short- and long-term effects of these therapeutic agents on the cardiovascular system in different clinical trials. We systematically reviewed the literature that had been published by January 2022. One hundred seven articles were initially identified by title and abstract using targeted keywords, and thirty-nine articles with relevance to cardiovascular effects of S1PR therapy in multiple sclerosis patients were thereafter considered, including their references for further accurate clarification. Studies on fingolimod, the first S1PR modulator approved for treating multiple sclerosis, primarily support the safety profile of this therapeutic class. The second-generation therapeutic agents along with a different treatment initiation approach helped mitigate several of the cardiovascular adverse effects that had previously been observed at the start of treatment. The heart rate may decrease when initiating S1PR modulators and, less commonly, the atrioventricular conduction may be prolonged, requiring cardiac monitoring for the first 6 h of medication. Continuous therapy with S1PR modulators can increase blood pressure values; therefore, the presence of arterial hypertension should be checked during long-term treatment. Periodic surveillance of the cardiovascular and autonomic functions can help predict cardiac outcomes and prevent possible adverse events in S1PR modulators treatment. Further studies with longer follow-ups are needed, especially for the second-generation of S1PR modulators, to confirm the safety profile of this therapeutic class.
Collapse
Affiliation(s)
- Victor Constantinescu
- Department of Neurology, Center of Clinical
Neuroscience, University Hospital Carl Gustav Carus, Dresden University of
Technology, Dresden, Germany
| | - Rocco Haase
- Department of Neurology, Center of Clinical
Neuroscience, University Hospital Carl Gustav Carus, Dresden University of
Technology, Dresden, Germany
| | - Katja Akgün
- Department of Neurology, Center of Clinical
Neuroscience, University Hospital Carl Gustav Carus, Dresden University of
Technology, Dresden, Germany
| | - Tjalf Ziemssen
- Department of Neurology, Center of Clinical
Neuroscience, University Hospital Carl Gustav Carus, Dresden University of
Technology, Fetscherstrasse 74, D-01307 Dresden, Germany
| |
Collapse
|
3
|
B-Cell Targeted Therapies in Patients with Multiple Sclerosis and Incidence of Headache: A Systematic Review and Meta-Analysis. J Pers Med 2022; 12:jpm12091474. [PMID: 36143259 PMCID: PMC9504525 DOI: 10.3390/jpm12091474] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 08/31/2022] [Accepted: 09/06/2022] [Indexed: 11/17/2022] Open
Abstract
Background: Multiple Sclerosis treatment with B-cell targeted therapies may be associated with an increased incidence of headache. We aimed to find and compare the association of B-cell targeted therapies with the incidence of headache in patients with Multiple Sclerosis. Methods: In a systematic based approach, the following databases were searched from inception until the 6th of June 2020: Pubmed/MEDLINE, ClinicalTrials.gov, EU Clinical Trials Register. Only randomized clinical trials (RCTs) enrolling patients with Multiple Sclerosis comparing B-cell targeted therapies (Rituximab, Ocrelizumab, Ofatumumab, Ublituximab or Cladribine) with placebo were selected for the systematic review and further meta-analysis. PRISMA guidelines were followed at all stages of the systematic review. The primary outcome was an all-cause headache of B-cell targeting therapy in patients with Multiple Sclerosis. Results: Nine RCTs were included. Compared with placebo, treatment with B-cell targeting therapies revealed a trend in headache risk, but it was not statistically significant (Relative Risk 1.12 [95% Confidence Interval 0.96–1.30]; p = 0.15; I2 = 9.32%). Surprisingly, in a sub-group analysis, Cladribine was statistically significant for an increase in headache risk (RR 1.20 [95% CI 1.006–1.42]; p = 0.042; I2 = 0%; 3 studies with 2107 participants). Conclusions: Even though a trend is shown, B-cell targeted therapies do not correlate with an increased incidence of headache as an adverse effect. Sub-analyses revealed a significant association between Cladribine alone and an increased incidence of headache. Whereas a purinergic signaling cascade is proposed as a mechanism of action, further research is needed to unravel the underlying pathogenetic mechanism of headache induction and establish headache prevention strategies.
Collapse
|
4
|
Pournajaf S, Dargahi L, Javan M, Pourgholami MH. Molecular Pharmacology and Novel Potential Therapeutic Applications of Fingolimod. Front Pharmacol 2022; 13:807639. [PMID: 35250559 PMCID: PMC8889014 DOI: 10.3389/fphar.2022.807639] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 01/31/2022] [Indexed: 12/14/2022] Open
Abstract
Fingolimod is a well-tolerated, highly effective disease-modifying therapy successfully utilized in the management of multiple sclerosis. The active metabolite, fingolimod-phosphate, acts on sphingosine-1-phosphate receptors (S1PRs) to bring about an array of pharmacological effects. While being initially recognized as a novel agent that can profoundly reduce T-cell numbers in circulation and the CNS, thereby suppressing inflammation and MS, there is now rapidly increasing knowledge on its previously unrecognized molecular and potential therapeutic effects in diverse pathological conditions. In addition to exerting inhibitory effects on sphingolipid pathway enzymes, fingolimod also inhibits histone deacetylases, transient receptor potential cation channel subfamily M member 7 (TRMP7), cytosolic phospholipase A2α (cPLA2α), reduces lysophosphatidic acid (LPA) plasma levels, and activates protein phosphatase 2A (PP2A). Furthermore, fingolimod induces apoptosis, autophagy, cell cycle arrest, epigenetic regulations, macrophages M1/M2 shift and enhances BDNF expression. According to recent evidence, fingolimod modulates a range of other molecular pathways deeply rooted in disease initiation or progression. Experimental reports have firmly associated the drug with potentially beneficial therapeutic effects in immunomodulatory diseases, CNS injuries, and diseases including Alzheimer's disease (AD), Parkinson's disease (PD), epilepsy, and even cancer. Attractive pharmacological effects, relative safety, favorable pharmacokinetics, and positive experimental data have collectively led to its testing in clinical trials. Based on the recent reports, fingolimod may soon find its way as an adjunct therapy in various disparate pathological conditions. This review summarizes the up-to-date knowledge about molecular pharmacology and potential therapeutic uses of fingolimod.
Collapse
Affiliation(s)
- Safura Pournajaf
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Leila Dargahi
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Javan
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | | |
Collapse
|
5
|
Sasset L, Di Lorenzo A. Sphingolipid Metabolism and Signaling in Endothelial Cell Functions. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1372:87-117. [PMID: 35503177 DOI: 10.1007/978-981-19-0394-6_8] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
The endothelium, inner layer of blood vessels, constitutes a metabolically active paracrine, endocrine, and autocrine organ, able to sense the neighboring environment and exert a variety of biological functions important to preserve the health of vasculature, tissues, and organs. Sphingolipids are both fundamental structural components of the eukaryotic membranes and signaling molecules regulating a variety of biological functions. Ceramide and sphingosine-1-phosphate (S1P), bioactive sphingolipids, have emerged as important regulators of cardiovascular functions in health and disease. In this review we discuss recent insights into the role of ceramide and S1P biosynthesis and signaling in regulating endothelial cell functions, in health and diseases. We also highlight advances into the mechanisms regulating serine palmitoyltransferase, the first and rate-limiting enzyme of de novo sphingolipid biosynthesis, with an emphasis on its inhibitors, ORMDL and NOGO-B. Understanding the molecular mechanisms regulating the sphingolipid de novo biosynthesis may provide the foundation for therapeutic modulation of this pathway in a variety of conditions, including cardiovascular diseases, associated with derangement of this pathway.
Collapse
Affiliation(s)
- Linda Sasset
- Department of Pathology and Laboratory Medicine, Cardiovascular Research Institute, Feil Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Annarita Di Lorenzo
- Department of Pathology and Laboratory Medicine, Cardiovascular Research Institute, Feil Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
6
|
Roy R, Alotaibi AA, Freedman MS. Sphingosine 1-Phosphate Receptor Modulators for Multiple Sclerosis. CNS Drugs 2021; 35:385-402. [PMID: 33797705 DOI: 10.1007/s40263-021-00798-w] [Citation(s) in RCA: 53] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/19/2021] [Indexed: 12/13/2022]
Abstract
Fingolimod (Gilenya) received regulatory approval from the US FDA in 2010 as the first-in-class sphingosine 1-phosphate (S1P) receptor (S1PR) modulator and was the first oral disease-modifying therapy (DMT) used for the treatment of the relapsing forms of multiple sclerosis (MS). Development of this new class of therapeutic compounds has continued to be a pharmacological goal of high interest in clinical trials for treatment of various autoimmune disorders, including MS. S1P is a physiologic signaling molecule that acts as a ligand for a group of cell surface receptors. S1PRs are expressed on various body tissues and regulate diverse physiological and pathological cellular responses involved in innate and adaptive immune, cardiovascular, and neurological functions. Subtype 1 of the S1PR (S1PR1) is expressed on the cell surface of lymphocytes, which are well known for their major role in MS pathogenesis and play an important regulatory role in the egress of lymphocytes from lymphoid organs to the lymphatic circulation. Thus, S1PR1-directed pharmacological interventions aim to modulate its role in immune cell trafficking through sequestration of autoreactive lymphocytes in the lymphoid organs to reduce their recirculation and subsequent infiltration into the central nervous system. Indeed, receptor subtype selectivity for S1PR1 is theoretically favored to minimize safety concerns related to interaction with other S1PR subtypes. Improved understanding of fingolimod's mechanism of action has provided strategies for the development of the more selective second-generation S1PR modulators. This selectivity serves to reduce the most important safety concern regarding cardiac-related side effects, such as bradycardia, which requires prolonged first-dose monitoring. It has led to the generation of smaller molecules with shorter half-lives, improved onset of action with no requirement for phosphorylation for activation, and preserved efficacy. The shorter half-lives of the second-generation agents allow for more rapid reversal of their pharmacological effects following treatment discontinuation. This may be beneficial in addressing further treatment-related complications in case of adverse events, managing serious or opportunistic infections such as progressive multifocal leukoencephalopathy, and eliminating the drug in pregnancies. In March 2019, a breakthrough in MS treatment was achieved with the FDA approval for the second S1PR modulator, siponimod (Mayzent), for both active secondary progressive MS and relapsing-remitting MS. This was the first oral DMT specifically approved for active forms of secondary progressive MS. Furthermore, ozanimod received FDA approval in March 2020 for treatment of relapsing forms of MS, followed by subsequent approvals from Health Canada and the European Commission. Other second-generation selective S1PR modulators that have been tested for MS, with statistically significant data from phase II and phase III clinical studies, include ponesimod (ACT-128800), ceralifimod (ONO-4641), and amiselimod (MT-1303). This review covers the available data about the mechanisms of action, pharmacodynamics and kinetics, efficacy, safety, and tolerability of the various S1PR modulators for patients with relapsing-remitting, secondary progressive, and, for fingolimod, primary progressive MS.
Collapse
Affiliation(s)
- Reshmi Roy
- Department of Medicine, The Ottawa Hospital Research Institute, University of Ottawa, Ottawa, Canada.
| | - Alaa A Alotaibi
- Department of Medicine, The Ottawa Hospital Research Institute, University of Ottawa, Ottawa, Canada
| | - Mark S Freedman
- Department of Medicine, The Ottawa Hospital Research Institute, University of Ottawa, Ottawa, Canada
| |
Collapse
|
7
|
Dhangadamajhi G, Singh S. Malaria link of hypertension: a hidden syndicate of angiotensin II, bradykinin and sphingosine 1-phosphate. Hum Cell 2021; 34:734-744. [PMID: 33683655 DOI: 10.1007/s13577-021-00513-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Accepted: 02/22/2021] [Indexed: 01/22/2023]
Abstract
In malaria-endemic countries, the burden of hypertension is on the rise. Although malaria and hypertension seem to have no direct link, several studies in recent years support their possible link. Three bioactive molecules such as angiotensin II (Ang II), bradykinin (BK) and sphingosine 1-phosphate (S1P) are crucial in regulating blood pressure. While the increased level of Ang II and S1P are responsible for inducing hypertension, BK is arthero-protective and anti-hypertensive. Therefore, in the present review, based on available literatures we highlight the present knowledge on the production and bioavailability of these molecules, the mechanism of their regulation of hypertension, and patho-physiological role in malaria. Further, a possible link between malaria and hypertension is hypothesized through various arguments based on experimental evidence. Understanding of their mechanisms of blood pressure regulation during malaria infection may open up avenues for drug therapeutics and management of malaria in co-morbidity with hypertension.
Collapse
Affiliation(s)
- Gunanidhi Dhangadamajhi
- Department of Biotechnology, Maharaja Sriramchandra Bhanjadeo University, Takatpur, Baripada, Odisha, 75003, India.
| | - Shailja Singh
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi, 110067, India
| |
Collapse
|
8
|
Fan X, Liu L, Shi Y, Guo F, He X, Zhao X, Zhong D, Li G. Recent advances of the function of sphingosine 1-phosphate (S1P) receptor S1P3. J Cell Physiol 2020; 236:1564-1578. [PMID: 33410533 DOI: 10.1002/jcp.29958] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Revised: 07/08/2020] [Accepted: 07/09/2020] [Indexed: 12/18/2022]
Abstract
Known as a variety of sphingolipid metabolites capable of performing various biological activities, sphingosine 1-phosphate (S1P) is commonly found in platelets, red blood cells, neutrophils, lymph fluid, and blood, as well as other cells and body fluids. S1P comprises five receptors, namely, S1P1-S1P5, with the distribution of S1P receptors exhibiting tissue selectivity to some degree. S1P1, S1P2, and S1P3 are extensively expressed in a wide variety of different tissues. The expression of S1P4 is restricted to lymphoid and hematopoietic tissues, while S1P5 is primarily expressed in the nervous system. S1P3 plays an essential role in the pathophysiological processes related to inflammation, cell proliferation, cell migration, tumor invasion and metastasis, ischemia-reperfusion, tissue fibrosis, and vascular tone. In this paper, the relevant mechanism in the role of S1P3 is summarized.
Collapse
Affiliation(s)
- Xuehui Fan
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Lili Liu
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Yue Shi
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Fanghan Guo
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Xiao He
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Xiuli Zhao
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Di Zhong
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Guozhong Li
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| |
Collapse
|
9
|
Potential sphingosine-1-phosphate-related therapeutic targets in the treatment of cerebral ischemia reperfusion injury. Life Sci 2020; 249:117542. [PMID: 32169519 DOI: 10.1016/j.lfs.2020.117542] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 02/29/2020] [Accepted: 03/09/2020] [Indexed: 12/17/2022]
Abstract
Sphingosine-1-phosphate (S1P) is a bioactive sphingolipid that regulates lymphocyte trafficking, glial cell activation, vasoconstriction, endothelial barrier function, and neuronal death pathways in the brain. Research has increasingly implicated S1P in the pathology of cerebral ischemia reperfusion (IR) injury. As a high-affinity agonist of S1P receptor, fingolimod exhibits excellent neuroprotective effects against ischemic challenge both in vivo and in vitro. By summarizing recent progress on how S1P participates in the development of brain IR injury, this review identifies potential therapeutic targets for the treatment of brain IR injury.
Collapse
|
10
|
Panta CR, Ruisanchez É, Móré D, Dancs PT, Balogh A, Fülöp Á, Kerék M, Proia RL, Offermanns S, Tigyi GJ, Benyó Z. Sphingosine-1-Phosphate Enhances α 1-Adrenergic Vasoconstriction via S1P2-G 12/13-ROCK Mediated Signaling. Int J Mol Sci 2019; 20:ijms20246361. [PMID: 31861195 PMCID: PMC6941080 DOI: 10.3390/ijms20246361] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 12/04/2019] [Accepted: 12/13/2019] [Indexed: 01/21/2023] Open
Abstract
Sphingosine-1-phosphate (S1P) has been implicated recently in the physiology and pathology of the cardiovascular system including regulation of vascular tone. Pilot experiments showed that the vasoconstrictor effect of S1P was enhanced markedly in the presence of phenylephrine (PE). Based on this observation, we hypothesized that S1P might modulate α1-adrenergic vasoactivity. In murine aortas, a 20-minute exposure to S1P but not to its vehicle increased the Emax and decreased the EC50 of PE-induced contractions indicating a hyperreactivity to α1-adrenergic stimulation. The potentiating effect of S1P disappeared in S1P2 but not in S1P3 receptor-deficient vessels. In addition, smooth muscle specific conditional deletion of G12/13 proteins or pharmacological inhibition of the Rho-associated protein kinase (ROCK) by Y-27632 or fasudil abolished the effect of S1P on α1-adrenergic vasoconstriction. Unexpectedly, PE-induced contractions remained enhanced markedly as late as three hours after S1P-exposure in wild-type (WT) and S1P3 KO but not in S1P2 KO vessels. In conclusion, the S1P–S1P2–G12/13–ROCK signaling pathway appears to have a major influence on α1-adrenergic vasoactivity. This cooperativity might lead to sustained vasoconstriction when increased sympathetic tone is accompanied by increased S1P production as it occurs during acute coronary syndrome and stroke.
Collapse
Affiliation(s)
- Cecília R. Panta
- Institute of Translational Medicine, Semmelweis University, 1094 Budapest, Hungary (D.M.); (P.T.D.); (A.B.); (M.K.); (G.J.T.)
- Correspondence: (C.R.P.); (Z.B.)
| | - Éva Ruisanchez
- Institute of Translational Medicine, Semmelweis University, 1094 Budapest, Hungary (D.M.); (P.T.D.); (A.B.); (M.K.); (G.J.T.)
| | - Dorottya Móré
- Institute of Translational Medicine, Semmelweis University, 1094 Budapest, Hungary (D.M.); (P.T.D.); (A.B.); (M.K.); (G.J.T.)
| | - Péter T. Dancs
- Institute of Translational Medicine, Semmelweis University, 1094 Budapest, Hungary (D.M.); (P.T.D.); (A.B.); (M.K.); (G.J.T.)
| | - Andrea Balogh
- Institute of Translational Medicine, Semmelweis University, 1094 Budapest, Hungary (D.M.); (P.T.D.); (A.B.); (M.K.); (G.J.T.)
| | - Ágnes Fülöp
- Institute of Translational Medicine, Semmelweis University, 1094 Budapest, Hungary (D.M.); (P.T.D.); (A.B.); (M.K.); (G.J.T.)
| | - Margit Kerék
- Institute of Translational Medicine, Semmelweis University, 1094 Budapest, Hungary (D.M.); (P.T.D.); (A.B.); (M.K.); (G.J.T.)
| | - Richard L. Proia
- National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), Bethesda, MD 20892, USA;
| | - Stefan Offermanns
- Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany;
| | - Gábor J. Tigyi
- Institute of Translational Medicine, Semmelweis University, 1094 Budapest, Hungary (D.M.); (P.T.D.); (A.B.); (M.K.); (G.J.T.)
- Department of Physiology, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Zoltán Benyó
- Institute of Translational Medicine, Semmelweis University, 1094 Budapest, Hungary (D.M.); (P.T.D.); (A.B.); (M.K.); (G.J.T.)
- Correspondence: (C.R.P.); (Z.B.)
| |
Collapse
|
11
|
Sphingosine 1-Phosphate (S1P)/ S1P Receptor Signaling and Mechanotransduction: Implications for Intrinsic Tissue Repair/Regeneration. Int J Mol Sci 2019; 20:ijms20225545. [PMID: 31703256 PMCID: PMC6888058 DOI: 10.3390/ijms20225545] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Revised: 10/31/2019] [Accepted: 11/05/2019] [Indexed: 12/16/2022] Open
Abstract
Tissue damage, irrespective from the underlying etiology, destroys tissue structure and, eventually, function. In attempt to achieve a morpho-functional recover of the damaged tissue, reparative/regenerative processes start in those tissues endowed with regenerative potential, mainly mediated by activated resident stem cells. These cells reside in a specialized niche that includes different components, cells and surrounding extracellular matrix (ECM), which, reciprocally interacting with stem cells, direct their cell behavior. Evidence suggests that ECM stiffness represents an instructive signal for the activation of stem cells sensing it by various mechanosensors, able to transduce mechanical cues into gene/protein expression responses. The actin cytoskeleton network dynamic acts as key mechanotransducer of ECM signal. The identification of signaling pathways influencing stem cell mechanobiology may offer therapeutic perspectives in the regenerative medicine field. Sphingosine 1-phosphate (S1P)/S1P receptor (S1PR) signaling, acting as modulator of ECM, ECM-cytoskeleton linking proteins and cytoskeleton dynamics appears a promising candidate. This review focuses on the current knowledge on the contribution of S1P/S1PR signaling in the control of mechanotransduction in stem/progenitor cells. The potential contribution of S1P/S1PR signaling in the mechanobiology of skeletal muscle stem cells will be argued based on the intriguing findings on S1P/S1PR action in this mechanically dynamic tissue.
Collapse
|
12
|
The potential application of
Cordyceps
in metabolic‐related disorders. Phytother Res 2019; 34:295-305. [DOI: 10.1002/ptr.6536] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 09/15/2019] [Accepted: 10/09/2019] [Indexed: 01/26/2023]
|
13
|
Alganga H, Almabrouk TAM, Katwan OJ, Daly CJ, Pyne S, Pyne NJ, Kennedy S. Short Periods of Hypoxia Upregulate Sphingosine Kinase 1 and Increase Vasodilation of Arteries to Sphingosine 1-Phosphate (S1P) via S1P 3. J Pharmacol Exp Ther 2019; 371:63-74. [PMID: 31371480 DOI: 10.1124/jpet.119.257931] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Accepted: 07/30/2019] [Indexed: 02/06/2023] Open
Abstract
Sphingosine kinase [(SK), isoforms SK1 and SK2] catalyzes the formation of the bioactive lipid, sphingosine 1-phosphate (S1P). This can be exported from cells and bind to S1P receptors to modulate vascular function. We investigated the effect of short-term hypoxia on SK1 expression and the response of arteries to S1P. SK1 expression in rat aortic and coronary artery endothelial cells was studied using immunofluorescence and confocal microscopy. Responses of rat aortic rings were studied using wire myography and reversible hypoxia induced by bubbling myography chambers with 95% N2:5% CO2 Inhibitors were added 30 minutes before induction of hypoxia. S1P induced endothelium-dependent vasodilation via activation of S1P3 receptors and generation of nitric oxide. Hypoxia significantly increased relaxation to S1P and this was attenuated by (2R)-1-[[(4-[[3-methyl-5-[(phenylsulfonyl)methyl] phenoxy]methyl]phenyl]methyl]-2-pyrrolidinemethanol [(PF-543), SK1 inhibitor] but not (R)-FTY720 methyl ether [(ROMe), SK2 inhibitor]. Hypoxia also increased vessel contractility to the thromboxane mimetic, 9,11-dideoxy-11α,9α-epoxymethanoprostaglandin F2α, which was further increased by PF-543 and ROMe. Hypoxia upregulated SK1 expression in aortic and coronary artery endothelial cells and this was blocked by PF-543 and 2-(p-hydroxyanilino)-4-(p-chlorophenyl)thiazole [(SKi), SK1/2 inhibitor]. The effects of PF-543 and SKi were associated with increased proteasomal/lysosomal degradation of SK1. A short period of hypoxia increases the expression of SK1, which may generate S1P to oppose vessel contraction. Under hypoxic conditions, upregulation of SK1 is likely to lead to increased export of S1P from the cell and vasodilation via activation of endothelial S1P3 receptors. These data have significance for perfusion of tissue during episodes of ischemia.
Collapse
Affiliation(s)
- H Alganga
- Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary & Life Sciences, University of Glasgow, Glasgow, United Kingdom (H.A., T.A.M.A., O.J.K., C.J.D., S.K.); Strathclyde Institute of Pharmacy and Biomedical Science, University of Strathclyde, Glasgow, United Kingdom (S.P., N.J.P.); Department of Pharmacology, School of Medicine, University of Zawia, Zawia, Libya (H.A., T.A.M.A.); and Department of Biochemistry, College of Medicine, University of Diyala, Baqubah, Iraq (O.J.K.)
| | - T A M Almabrouk
- Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary & Life Sciences, University of Glasgow, Glasgow, United Kingdom (H.A., T.A.M.A., O.J.K., C.J.D., S.K.); Strathclyde Institute of Pharmacy and Biomedical Science, University of Strathclyde, Glasgow, United Kingdom (S.P., N.J.P.); Department of Pharmacology, School of Medicine, University of Zawia, Zawia, Libya (H.A., T.A.M.A.); and Department of Biochemistry, College of Medicine, University of Diyala, Baqubah, Iraq (O.J.K.)
| | - O J Katwan
- Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary & Life Sciences, University of Glasgow, Glasgow, United Kingdom (H.A., T.A.M.A., O.J.K., C.J.D., S.K.); Strathclyde Institute of Pharmacy and Biomedical Science, University of Strathclyde, Glasgow, United Kingdom (S.P., N.J.P.); Department of Pharmacology, School of Medicine, University of Zawia, Zawia, Libya (H.A., T.A.M.A.); and Department of Biochemistry, College of Medicine, University of Diyala, Baqubah, Iraq (O.J.K.)
| | - C J Daly
- Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary & Life Sciences, University of Glasgow, Glasgow, United Kingdom (H.A., T.A.M.A., O.J.K., C.J.D., S.K.); Strathclyde Institute of Pharmacy and Biomedical Science, University of Strathclyde, Glasgow, United Kingdom (S.P., N.J.P.); Department of Pharmacology, School of Medicine, University of Zawia, Zawia, Libya (H.A., T.A.M.A.); and Department of Biochemistry, College of Medicine, University of Diyala, Baqubah, Iraq (O.J.K.)
| | - S Pyne
- Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary & Life Sciences, University of Glasgow, Glasgow, United Kingdom (H.A., T.A.M.A., O.J.K., C.J.D., S.K.); Strathclyde Institute of Pharmacy and Biomedical Science, University of Strathclyde, Glasgow, United Kingdom (S.P., N.J.P.); Department of Pharmacology, School of Medicine, University of Zawia, Zawia, Libya (H.A., T.A.M.A.); and Department of Biochemistry, College of Medicine, University of Diyala, Baqubah, Iraq (O.J.K.)
| | - N J Pyne
- Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary & Life Sciences, University of Glasgow, Glasgow, United Kingdom (H.A., T.A.M.A., O.J.K., C.J.D., S.K.); Strathclyde Institute of Pharmacy and Biomedical Science, University of Strathclyde, Glasgow, United Kingdom (S.P., N.J.P.); Department of Pharmacology, School of Medicine, University of Zawia, Zawia, Libya (H.A., T.A.M.A.); and Department of Biochemistry, College of Medicine, University of Diyala, Baqubah, Iraq (O.J.K.)
| | - S Kennedy
- Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary & Life Sciences, University of Glasgow, Glasgow, United Kingdom (H.A., T.A.M.A., O.J.K., C.J.D., S.K.); Strathclyde Institute of Pharmacy and Biomedical Science, University of Strathclyde, Glasgow, United Kingdom (S.P., N.J.P.); Department of Pharmacology, School of Medicine, University of Zawia, Zawia, Libya (H.A., T.A.M.A.); and Department of Biochemistry, College of Medicine, University of Diyala, Baqubah, Iraq (O.J.K.)
| |
Collapse
|
14
|
Sposito AC, de Lima-Junior JC, Moura FA, Barreto J, Bonilha I, Santana M, Virginio VW, Sun L, Carvalho LSF, Soares AA, Nadruz W, Feinstein SB, Nofer JR, Zanotti I, Kontush A, Remaley AT. Reciprocal Multifaceted Interaction Between HDL (High-Density Lipoprotein) and Myocardial Infarction. Arterioscler Thromb Vasc Biol 2019; 39:1550-1564. [DOI: 10.1161/atvbaha.119.312880] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Despite decades of therapeutic advances, myocardial infarction remains a leading cause of death worldwide. Recent studies have identified HDLs (high-density lipoproteins) as a potential candidate for mitigating coronary ischemia/reperfusion injury via a broad spectrum of signaling pathways. HDL ligands, such as S1P (sphingosine-1-phosphate), Apo (apolipoprotein) A-I, clusterin, and miRNA, may influence the opening of the mitochondrial channel, insulin sensitivity, and production of vascular autacoids, such as NO, prostacyclin, and endothelin-1. In parallel, antioxidant activity and sequestration of oxidized molecules provided by HDL can attenuate the oxidative stress that triggers ischemia/reperfusion. Nevertheless, during myocardial infarction, oxidation and the capture of oxidized and proinflammatory molecules generate large phenotypic and functional changes in HDL, potentially limiting its beneficial properties. In this review, new findings from cellular and animal models, as well as from clinical studies, will be discussed to describe the cardioprotective benefits of HDL on myocardial infarction. Furthermore, mechanisms by which HDL modulates cardiac function and potential strategies to mitigate postmyocardial infarction risk damage by HDL will be detailed throughout the review.
Collapse
Affiliation(s)
- Andrei C. Sposito
- From the Atherosclerosis and Vascular Biology Laboratory, Cardiology Department, State University of Campinas, Brazil (A.C.S., J.C.d.L.-J., F.A.M., J.B., I.B., M.S., V.W.V., L.S.F.C., A.A.S.S., W.N.)
| | - José Carlos de Lima-Junior
- From the Atherosclerosis and Vascular Biology Laboratory, Cardiology Department, State University of Campinas, Brazil (A.C.S., J.C.d.L.-J., F.A.M., J.B., I.B., M.S., V.W.V., L.S.F.C., A.A.S.S., W.N.)
| | - Filipe A. Moura
- From the Atherosclerosis and Vascular Biology Laboratory, Cardiology Department, State University of Campinas, Brazil (A.C.S., J.C.d.L.-J., F.A.M., J.B., I.B., M.S., V.W.V., L.S.F.C., A.A.S.S., W.N.)
- Department of Medicine, Weill-Cornell Medical College, New York, NY (F.A.M.)
| | - Joaquim Barreto
- From the Atherosclerosis and Vascular Biology Laboratory, Cardiology Department, State University of Campinas, Brazil (A.C.S., J.C.d.L.-J., F.A.M., J.B., I.B., M.S., V.W.V., L.S.F.C., A.A.S.S., W.N.)
| | - Isabella Bonilha
- From the Atherosclerosis and Vascular Biology Laboratory, Cardiology Department, State University of Campinas, Brazil (A.C.S., J.C.d.L.-J., F.A.M., J.B., I.B., M.S., V.W.V., L.S.F.C., A.A.S.S., W.N.)
| | - Michele Santana
- From the Atherosclerosis and Vascular Biology Laboratory, Cardiology Department, State University of Campinas, Brazil (A.C.S., J.C.d.L.-J., F.A.M., J.B., I.B., M.S., V.W.V., L.S.F.C., A.A.S.S., W.N.)
| | - Vitor W. Virginio
- From the Atherosclerosis and Vascular Biology Laboratory, Cardiology Department, State University of Campinas, Brazil (A.C.S., J.C.d.L.-J., F.A.M., J.B., I.B., M.S., V.W.V., L.S.F.C., A.A.S.S., W.N.)
| | - Lufan Sun
- Lipoprotein Metabolism Section, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD (L.S., A.T.R.)
- Department of Cardiology, The First Hospital of China Medical University, Shenyang, Liaoning Province, China (L.S.)
| | - Luiz Sergio F. Carvalho
- From the Atherosclerosis and Vascular Biology Laboratory, Cardiology Department, State University of Campinas, Brazil (A.C.S., J.C.d.L.-J., F.A.M., J.B., I.B., M.S., V.W.V., L.S.F.C., A.A.S.S., W.N.)
| | - Alexandre A.S. Soares
- From the Atherosclerosis and Vascular Biology Laboratory, Cardiology Department, State University of Campinas, Brazil (A.C.S., J.C.d.L.-J., F.A.M., J.B., I.B., M.S., V.W.V., L.S.F.C., A.A.S.S., W.N.)
| | - Wilson Nadruz
- From the Atherosclerosis and Vascular Biology Laboratory, Cardiology Department, State University of Campinas, Brazil (A.C.S., J.C.d.L.-J., F.A.M., J.B., I.B., M.S., V.W.V., L.S.F.C., A.A.S.S., W.N.)
| | - Steve B. Feinstein
- Division of Cardiology, Rush University Medical Center, Chicago, IL (S.B.F.)
| | - Jerzy-Roch Nofer
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf, Germany (J.-R.N.)
| | - Ilaria Zanotti
- Department of Food and Drugs, University of Parma, Italy (I.Z.)
| | - Anatol Kontush
- UMR-ICAN 1166, National Institute for Health and Medical Research (INSERM), Sorbonne University, Paris, France (A.K.)
| | - Alan T. Remaley
- Lipoprotein Metabolism Section, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD (L.S., A.T.R.)
| |
Collapse
|
15
|
Multiple sclerosis treatment with fingolimod: profile of non-cardiologic adverse events. Acta Neurol Belg 2017; 117:821-827. [PMID: 28528469 DOI: 10.1007/s13760-017-0794-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Accepted: 05/15/2017] [Indexed: 01/25/2023]
Abstract
Fingolimod was the first oral medication approved for management of multiple sclerosis and is currently used by tens of thousands patients worldwide. Fingolimod acts via the sphingosine 1-phosphate (S1P) receptor, maintaining peripheral lymphocytes entrapped in the lymph nodes. In consequence, there is a reduction in the infiltration of aggressive lymphocytes into the central nervous system. The drug is safe and effective, and its first hours of use are associated with related to S1P receptors in the heart. This side effect is well known by all doctors prescribing fingolimod. However, the drug has other potential adverse events that, although relatively rare, require awareness from the neurologist. Among these there are infections (herpes simplex, herpes zoster, Cryptococcus, Epstein-Barr virus, hepatitis, Molluscum Contagiosum, and leishmaniosis), lung and thyroid complications, refractory headaches, encephalopathy, vasculopathy, tumefactive lesions in magnetic resonance imaging and ophthalmological disorders. The present review lists the non-cardiologic adverse events that all neurologists prescribing fingolimod should be aware of.
Collapse
|
16
|
Nielsen OH, Li Y, Johansson-Lindbom B, Coskun M. Sphingosine-1-Phosphate Signaling in Inflammatory Bowel Disease. Trends Mol Med 2017; 23:362-374. [DOI: 10.1016/j.molmed.2017.02.002] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2016] [Revised: 02/10/2017] [Accepted: 02/14/2017] [Indexed: 12/14/2022]
|
17
|
Siedlinski M, Nosalski R, Szczepaniak P, Ludwig-Gałęzowska AH, Mikołajczyk T, Filip M, Osmenda G, Wilk G, Nowak M, Wołkow P, Guzik TJ. Vascular transcriptome profiling identifies Sphingosine kinase 1 as a modulator of angiotensin II-induced vascular dysfunction. Sci Rep 2017; 7:44131. [PMID: 28276483 PMCID: PMC5343497 DOI: 10.1038/srep44131] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Accepted: 02/03/2017] [Indexed: 12/22/2022] Open
Abstract
Vascular dysfunction is an important phenomenon in hypertension. We hypothesized that angiotensin II (AngII) affects transcriptome in the vasculature in a region-specific manner, which may help to identify genes related to vascular dysfunction in AngII-induced hypertension. Mesenteric artery and aortic transcriptome was profiled using Illumina WG-6v2.0 chip in control and AngII infused (490 ng/kg/min) hypertensive mice. Gene set enrichment and leading edge analyses identified Sphingosine kinase 1 (Sphk1) in the highest number of pathways affected by AngII. Sphk1 mRNA, protein and activity were up-regulated in the hypertensive vasculature. Chronic sphingosine-1-phosphate (S1P) infusion resulted in a development of significantly increased vasoconstriction and endothelial dysfunction. AngII-induced hypertension was blunted in Sphk1-/- mice (systolic BP 167 ± 4.2 vs. 180 ± 3.3 mmHg, p < 0.05), which was associated with decreased aortic and mesenteric vasoconstriction in hypertensive Sphk1-/- mice. Pharmacological inhibition of S1P synthesis reduced vasoconstriction of mesenteric arteries. While Sphk1 is important in mediating vasoconstriction in hypertension, Sphk1-/- mice were characterized by enhanced endothelial dysfunction, suggesting a local protective role of Sphk1 in the endothelium. S1P serum level in humans was correlated with endothelial function (arterial tonometry). Thus, vascular transcriptome analysis shows that S1P pathway is critical in the regulation of vascular function in AngII-induced hypertension, although Sphk1 may have opposing roles in the regulation of vasoconstriction and endothelium-dependent vasorelaxation.
Collapse
Affiliation(s)
- Mateusz Siedlinski
- Department of Internal and Agricultural Medicine, Faculty of Medicine, Jagiellonian University Medical College, Kraków, Poland
| | - Ryszard Nosalski
- Department of Internal and Agricultural Medicine, Faculty of Medicine, Jagiellonian University Medical College, Kraków, Poland.,British Heart Foundation Centre for Excellence, Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, Scotland, UK
| | - Piotr Szczepaniak
- Department of Internal and Agricultural Medicine, Faculty of Medicine, Jagiellonian University Medical College, Kraków, Poland
| | | | - Tomasz Mikołajczyk
- Department of Internal and Agricultural Medicine, Faculty of Medicine, Jagiellonian University Medical College, Kraków, Poland.,British Heart Foundation Centre for Excellence, Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, Scotland, UK
| | - Magdalena Filip
- Department of Internal and Agricultural Medicine, Faculty of Medicine, Jagiellonian University Medical College, Kraków, Poland
| | - Grzegorz Osmenda
- Department of Internal and Agricultural Medicine, Faculty of Medicine, Jagiellonian University Medical College, Kraków, Poland
| | - Grzegorz Wilk
- Department of Internal and Agricultural Medicine, Faculty of Medicine, Jagiellonian University Medical College, Kraków, Poland
| | - Michał Nowak
- Department of Internal and Agricultural Medicine, Faculty of Medicine, Jagiellonian University Medical College, Kraków, Poland
| | - Paweł Wołkow
- Centre for Medical Genomics-OMICRON, Jagiellonian University Medical College, Kraków, Poland
| | - Tomasz J Guzik
- Department of Internal and Agricultural Medicine, Faculty of Medicine, Jagiellonian University Medical College, Kraków, Poland.,British Heart Foundation Centre for Excellence, Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, Scotland, UK
| |
Collapse
|
18
|
Cui K, Ruan Y, Wang T, Rao K, Chen Z, Wang S, Liu J. FTY720 Supplementation Partially Improves Erectile Dysfunction in Rats With Streptozotocin-Induced Type 1 Diabetes Through Inhibition of Endothelial Dysfunction and Corporal Fibrosis. J Sex Med 2017; 14:323-335. [DOI: 10.1016/j.jsxm.2017.01.006] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2016] [Revised: 01/10/2017] [Accepted: 01/11/2017] [Indexed: 10/20/2022]
|
19
|
van Vuuren D, Marais E, Genade S, Lochner A. The differential effects of FTY720 on functional recovery and infarct size following myocardial ischaemia/reperfusion. Cardiovasc J Afr 2017; 27:375-386. [PMID: 27966000 PMCID: PMC5408499 DOI: 10.5830/cvja-2016-039] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2015] [Accepted: 03/30/2016] [Indexed: 01/08/2023] Open
Abstract
AIM The aim of this study was to evaluate the effects of the sphingosine analogue, FTY720 (Fingolimod), on the outcomes of myocardial ischaemia/reperfusion (I/R) injury. METHODS Two concentrations of FTY720 (1 or 2.5 µM were administered either prior to (PreFTY), or following (PostFTY) 20 minutes' global (GI) or 35 minutes' regional ischaemia (RI) in the isolated, perfused, working rat heart. Functional recovery during reperfusion was assessed following both models of ischaemia, while infarct size (IFS) was determined following RI. RESULTS FTY720 at 1 µM exerted no effect on functional recovery, while 2.5 µM significantly impaired aortic output (AO) recovery when administered prior to GI (% recovery: control: 33.88 ± 6.12% vs PreFTY: 0%, n = 6-10; p < 0.001), as well as before and after RI ( % recovery: control: 27.86 ± 13.22% vs PreFTY: 0.62% ; p < 0.05; and PostFTY: 2.08%; p = 0.0585, n = 6). FTY720 at 1 µM administered during reperfusion reduced IFS (% of area at risk (AAR): control: 39.89 ± 3.93% vs PostFTY: 26.56 ± 4.32%, n = 6-8; p < 0.05), while 2.5 µM FTY720 reduced IFS irrespective of the time of administration ( % of AAR: control: 39.89 ± 3.93% vs PreFTY: 29.97 ± 1.03% ; and PostFTY: 30.45 ± 2.16%, n = 6; p < 0.05). CONCLUSION FTY720 exerted divergent outcomes on function and tissue survival depending on the concentration administered, as well as the timing of administration.
Collapse
Affiliation(s)
- Derick van Vuuren
- Division of Medical Physiology, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Stellenbosch University, Tygerberg, South Africa.
| | - Erna Marais
- Division of Medical Physiology, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Stellenbosch University, Tygerberg, South Africa
| | - Sonia Genade
- Division of Medical Physiology, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Stellenbosch University, Tygerberg, South Africa
| | - Amanda Lochner
- Division of Medical Physiology, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Stellenbosch University, Tygerberg, South Africa
| |
Collapse
|
20
|
Tölle M, Klöckl L, Wiedon A, Zidek W, van der Giet M, Schuchardt M. Regulation of endothelial nitric oxide synthase activation in endothelial cells by S1P1 and S1P3. Biochem Biophys Res Commun 2016; 476:627-634. [DOI: 10.1016/j.bbrc.2016.06.009] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Accepted: 06/01/2016] [Indexed: 12/16/2022]
|
21
|
Cantalupo A, Di Lorenzo A. S1P Signaling and De Novo Biosynthesis in Blood Pressure Homeostasis. J Pharmacol Exp Ther 2016; 358:359-70. [PMID: 27317800 DOI: 10.1124/jpet.116.233205] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Accepted: 06/13/2016] [Indexed: 01/12/2023] Open
Abstract
Initially discovered as abundant components of eukaryotic cell membranes, sphingolipids are now recognized as important bioactive signaling molecules that modulate a variety of cellular functions, including those relevant to cancer and immunologic, inflammatory, and cardiovascular disorders. In this review, we discuss recent advances in our understanding of the role of sphingosine-1-phosphate (S1P) receptors in the regulation of vascular function, and focus on how de novo biosynthesized sphingolipids play a role in blood pressure homeostasis. The therapeutic potential of new drugs that target S1P signaling is also discussed.
Collapse
Affiliation(s)
- Anna Cantalupo
- Department of Pathology and Laboratory Medicine, Center for Vascular Biology, Weill Cornell Medicine, Cornell University, New York, New York
| | - Annarita Di Lorenzo
- Department of Pathology and Laboratory Medicine, Center for Vascular Biology, Weill Cornell Medicine, Cornell University, New York, New York
| |
Collapse
|
22
|
White CR, Giordano S, Anantharamaiah GM. High-density lipoprotein, mitochondrial dysfunction and cell survival mechanisms. Chem Phys Lipids 2016; 199:161-169. [PMID: 27150975 DOI: 10.1016/j.chemphyslip.2016.04.007] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2016] [Revised: 04/22/2016] [Accepted: 04/23/2016] [Indexed: 01/08/2023]
Abstract
Ischemic injury is associated with acute myocardial infarction, percutaneous coronary intervention, coronary artery bypass grafting and open heart surgery. The timely re-establishment of blood flow is critical in order to minimize cardiac complications. Reperfusion after a prolonged ischemic period, however, can induce severe cardiomyocyte dysfunction with mitochondria serving as a major target of ischemia/reperfusion (I/R) injury. An increase in the formation of reactive oxygen species (ROS) induces damage to mitochondrial respiratory complexes leading to uncoupling of oxidative phosphorylation. Mitochondrial membrane perturbations also contribute to calcium overload, opening of the mitochondrial permeability transition pore (mPTP) and the release of apoptotic mediators into the cytoplasm. Clinical and experimental studies show that ischemic preconditioning (ICPRE) and postconditioning (ICPOST) attenuate mitochondrial injury and improve cardiac function in the context of I/R injury. This is achieved by the activation of two principal cell survival cascades: 1) the Reperfusion Injury Salvage Kinase (RISK) pathway; and 2) the Survivor Activating Factor Enhancement (SAFE) pathway. Recent data suggest that high density lipoprotein (HDL) mimics the effects of conditioning protocols and attenuates myocardial I/R injury via activation of the RISK and SAFE signaling cascades. In this review, we discuss the roles of apolipoproteinA-I (apoA-I), the major protein constituent of HDL, and sphingosine 1-phosphate (S1P), a lysosphingolipid associated with small, dense HDL particles as mediators of cardiomyocyte survival. Both apoA-I and S1P exert an infarct-sparing effect by preventing ROS-dependent injury and inhibiting the opening of the mPTP.
Collapse
Affiliation(s)
- C Roger White
- Department of Medicine, Division of Cardiovascular Disease, University of Alabama at Birmingham, Birmingham, AL, USA.
| | - Samantha Giordano
- Department of Medicine, Division of Cardiovascular Disease, University of Alabama at Birmingham, Birmingham, AL, USA
| | - G M Anantharamaiah
- The Division of Gerontology, Geriatric Medicine and Palliative Care, University of Alabama at Birmingham, Birmingham, AL, USA; Biochemistry and Molecular Genetics, University of Alabama at Birmingham, Birmingham, AL, USA
| |
Collapse
|
23
|
Imeri F, Blanchard O, Jenni A, Schwalm S, Wünsche C, Zivkovic A, Stark H, Pfeilschifter J, Huwiler A. FTY720 and two novel butterfly derivatives exert a general anti-inflammatory potential by reducing immune cell adhesion to endothelial cells through activation of S1P(3) and phosphoinositide 3-kinase. Naunyn Schmiedebergs Arch Pharmacol 2015; 388:1283-92. [PMID: 26267293 DOI: 10.1007/s00210-015-1159-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2014] [Accepted: 07/27/2015] [Indexed: 10/23/2022]
Abstract
Sphingosine-1-phosphate (S1P) is a key lipid regulator of a variety of cellular responses including cell proliferation and survival, cell migration, and inflammatory reactions. Here, we investigated the effect of S1P receptor activation on immune cell adhesion to endothelial cells under inflammatory conditions. We show that S1P reduces both tumor necrosis factor (TNF)-α- and lipopolysaccharide (LPS)-stimulated adhesion of Jurkat and U937 cells to an endothelial monolayer. The reducing effect of S1P was reversed by the S1P1+3 antagonist VPC23019 but not by the S1P1 antagonist W146. Additionally, knockdown of S1P3, but not S1P1, by short hairpin RNA (shRNA) abolished the reducing effect of S1P, suggesting the involvement of S1P3. A suppression of immune cell adhesion was also seen with the immunomodulatory drug FTY720 and two novel butterfly derivatives ST-968 and ST-1071. On the molecular level, S1P and all FTY720 derivatives reduced the mRNA expression of LPS- and TNF-α-induced adhesion molecules including ICAM-1, VCAM-1, E-selectin, and CD44 which was reversed by the PI3K inhibitor LY294002, but not by the MEK inhibitor U0126.In summary, our data demonstrate a novel molecular mechanism by which S1P, FTY720, and two novel butterfly derivatives acted anti-inflammatory that is by suppressing gene transcription of various endothelial adhesion molecules and thereby preventing adhesion of immune cells to endothelial cells and subsequent extravasation.
Collapse
Affiliation(s)
- Faik Imeri
- Institute of Pharmacology, University of Bern, Inselspital, INO-F, CH-3010, Bern, Switzerland
| | - Olivier Blanchard
- Institute of Pharmacology, University of Bern, Inselspital, INO-F, CH-3010, Bern, Switzerland
| | - Aurelio Jenni
- Institute of Pharmacology, University of Bern, Inselspital, INO-F, CH-3010, Bern, Switzerland
| | - Stephanie Schwalm
- Institute of Pharmacology, University of Bern, Inselspital, INO-F, CH-3010, Bern, Switzerland.,Pharmazentrum Frankfurt/ZAFES, Klinikum der Goethe-Universität Frankfurt am Main, Frankfurt am Main, Germany
| | - Christin Wünsche
- Institute of Pharmacology, University of Bern, Inselspital, INO-F, CH-3010, Bern, Switzerland.,Pharmazentrum Frankfurt/ZAFES, Klinikum der Goethe-Universität Frankfurt am Main, Frankfurt am Main, Germany
| | - Aleksandra Zivkovic
- Institute of Pharmaceutical and Medicinal Chemistry, Heinrich Heine-University Düsseldorf, Universitätsstr. 1, 40225, Düsseldorf, Germany
| | - Holger Stark
- Institute of Pharmaceutical and Medicinal Chemistry, Heinrich Heine-University Düsseldorf, Universitätsstr. 1, 40225, Düsseldorf, Germany
| | - Josef Pfeilschifter
- Pharmazentrum Frankfurt/ZAFES, Klinikum der Goethe-Universität Frankfurt am Main, Frankfurt am Main, Germany
| | - Andrea Huwiler
- Institute of Pharmacology, University of Bern, Inselspital, INO-F, CH-3010, Bern, Switzerland.
| |
Collapse
|
24
|
Aguiar C, Batista S, Pacheco R. Cardiovascular effects of fingolimod: Relevance, detection and approach. Rev Port Cardiol 2015; 34:279-85. [PMID: 25843307 DOI: 10.1016/j.repc.2014.11.012] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2014] [Accepted: 11/15/2014] [Indexed: 01/10/2023] Open
Abstract
Fingolimod, a structural analogue of sphingosine, is the first oral treatment available for multiple sclerosis. The presence of sphingosine-1-phosphate receptors in the sinus and atrioventricular nodes, myocardial cells, endothelial cells and arterial smooth muscle cells is responsible for fingolimod's cardiovascular effects. We provide a comprehensive review of the mechanisms of these effects and characterize their clinical relevance.
Collapse
Affiliation(s)
- Carlos Aguiar
- Cardiology Department, Hospital Santa Cruz, CHLO, Carnaxide, Portugal.
| | - Sónia Batista
- Multiple Sclerosis Outpatient Clinic, Coimbra University, Coimbra, Portugal
| | - Ricardo Pacheco
- Medical Department, Novartis Farma S.A., Porto Salvo, Portugal
| |
Collapse
|
25
|
Aguiar C, Batista S, Pacheco R. Cardiovascular effects of fingolimod: Relevance, detection and approach. REVISTA PORTUGUESA DE CARDIOLOGIA (ENGLISH EDITION) 2015. [DOI: 10.1016/j.repce.2014.11.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
|
26
|
Lindå H, von Heijne A. A case of posterior reversible encephalopathy syndrome associated with gilenya(®) (fingolimod) treatment for multiple sclerosis. Front Neurol 2015; 6:39. [PMID: 25788891 PMCID: PMC4349179 DOI: 10.3389/fneur.2015.00039] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2014] [Accepted: 02/16/2015] [Indexed: 11/25/2022] Open
Abstract
We describe posterior reversible encephalopathy syndrome (PRES) in a woman with multiple sclerosis treated with Gilenya® (Fingolimod). The first symptoms appeared after 21 months of fingolimod treatment. She experienced headache, altered mental status, cognitive deficits, seizures, and visual disturbances. Not at any time during the course of the disease could any signs of infection or rheumatic disorder be detected. Test for anti-neuronal antibodies was also negative. Her blood pressure was normal. MRI showed widespread cortical and subcortical changes with some mass-effect in the temporo-occipital-parietal lobes in the left hemisphere. Contrast enhancement was seen in the leptomeninges and, in addition, there were no areas with restricted diffusion and no signs of hemorrhage. Her condition deteriorated until fingolimod was discontinued. Slowly her condition improved and after 8 months, the only symptoms that remained were two small, non-corresponding, right inferior scotomas. We believe that all symptoms, the clinical course, and the MRI findings in this case can all be explained by considering PRES, a probably rare, but serious, side effect of fingolimod treatment.
Collapse
Affiliation(s)
- Hans Lindå
- Neurology Unit, Division of Internal Medicine, Karolinska Institute, Danderyd Hospital , Danderyd , Sweden ; Neurology Clinic, Sophiahemmet , Stockholm , Sweden
| | | |
Collapse
|
27
|
Xu HL, Pelligrino DA, Paisansathan C, Testai FD. Protective role of fingolimod (FTY720) in rats subjected to subarachnoid hemorrhage. J Neuroinflammation 2015; 12:16. [PMID: 25622980 PMCID: PMC4324852 DOI: 10.1186/s12974-015-0234-7] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2014] [Accepted: 01/01/2015] [Indexed: 12/20/2022] Open
Abstract
Background Subarachnoid hemorrhage (SAH) is a neurological emergency with limited pharmacological treatment options. Inflammation is increasingly recognized as a key pathogenic contributor to brain injury in this condition. In the present study, we examined the neuroprotective effects of the immunomodulatory agent, fingolimod, in rats subjected to SAH. Methods We utilized an endovascular rat perforation model of SAH. Animals were divided into four groups: (1) sham-vehicle; (2) sham-fingolimod; (3) SAH-vehicle; and (4) SAH-fingolimod. Rats received either vehicle solution or fingolimod (0.5 mg/kg) intraperitoneally 3 hours after sham surgery or SAH. A closed cranial window and intravital microscope system was used at 48 hours to assess neuroinflammation, which was represented by rhodamine-6G-labeled leukocyte trafficking in pial venules, and pial arteriolar dilating responses to a variety of vasodilators, including hypercapnia, and topically-applied acetylcholine, adenosine, and S-nitroso-N-acetyl penicillamine. In addition, motor-sensory function was evaluated. Results Compared to sham-vehicle rats, SAH-vehicle animals displayed a four-times greater increase in pial venular intraluminal leukocyte adhesion. Treatment with fingolimod largely reduced the intravascular leukocyte adhesion. Vehicle-treated SAH animals displayed a significant decrease in pial arteriolar responses to all the vasodilators tested and vascular reactivity was preserved, to a significant degree, in the presence of fingolimod. In addition, neurological scores obtained at 48 hours post-SAH indicated significant neurological deficits in the vehicle-treated group (versus sham-vehicle surgical control). Those deficiencies were partially reduced by fingolimod (P < 0.0001 compared to the vehicle-treated SAH group). Conclusions Treatment of rats with fingolimod was associated with a marked limitation in the intravascular adhesion of leukocytes to pial venules, preserved pial arteriolar dilating function, and improved neurological outcome in rats subjected to SAH.
Collapse
Affiliation(s)
- Hao-Liang Xu
- Neuroanesthesia Research Laboratory, University of Illinois College of Medicine, Chicago, IL, USA.
| | - Dale A Pelligrino
- Neuroanesthesia Research Laboratory, University of Illinois College of Medicine, Chicago, IL, USA.
| | - Chanannait Paisansathan
- Department of Anesthesiology of the University of Illinois College of Medicine, Chicago, IL, USA.
| | - Fernando D Testai
- Department of Neurology and Rehabilitation of the University of Illinois College of Medicine, 912 S Wood Street, Rm 855 N NPI (MC 796), Chicago, IL, 60612-7330, USA.
| |
Collapse
|
28
|
Camm J, Hla T, Bakshi R, Brinkmann V. Cardiac and vascular effects of fingolimod: mechanistic basis and clinical implications. Am Heart J 2014; 168:632-44. [PMID: 25440790 DOI: 10.1016/j.ahj.2014.06.028] [Citation(s) in RCA: 161] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2014] [Accepted: 06/15/2014] [Indexed: 12/19/2022]
Abstract
Fingolimod, a sphingosine-1-phosphate receptor (S1PR) modulator, was the first oral disease-modifying therapy approved for relapsing forms of multiple sclerosis; it reduces autoreactive lymphocytes' egress from lymphoid tissues by down-regulating S1PRs. Sphingosine-1-phosphate signaling is implicated in a range of physiologic functions, and S1PRs are expressed differentially in various tissues, including the cardiovascular system. Modulation of S1PRs on cardiac cells provides an explanation for the transient effects of fingolimod on heart rate and atrioventricular conduction at initiation of fingolimod therapy, and for the mild but more persistent effects on blood pressure observed in some patients on long-term treatment. This review describes the nontherapeutic actions of fingolimod in the context of sphingosine-1-phosphate signaling in the cardiovascular system, as well as providing a summary of the associated clinical implications useful to physicians considering initiation of fingolimod therapy in patients. A transient reduction in heart rate (mean decrease of 8 beats per minute) and, less commonly, a temporary delay in atrioventricular conduction observed in some patients when initiating fingolimod therapy are both due to activation of S1PR subtype 1 on cardiac myocytes. These effects are a reflection of fingolimod first acting as a full S1PR agonist and thereafter functioning as an S1PR antagonist after down-regulation of S1PR subtype 1 at the cell surface. For most individuals, first-dose effects of fingolimod are asymptomatic, but all patients need to be monitored for at least 6 hours after the first dose, in accordance with the label recommendations.
Collapse
|
29
|
Cheng Q, Ma S, Lin D, Mei Y, Gong H, Lei L, Chen Y, Zhao Y, Hu B, Wu Y, Yu X, Zhao L, Liu H. The S1P1 receptor-selective agonist CYM-5442 reduces the severity of acute GVHD by inhibiting macrophage recruitment. Cell Mol Immunol 2014; 12:681-91. [PMID: 25088224 DOI: 10.1038/cmi.2014.59] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2014] [Revised: 06/15/2014] [Accepted: 06/15/2014] [Indexed: 01/04/2023] Open
Abstract
FTY720, an agonist for four of the five known sphingosine-1-phosphate (S1P) receptors, has been reported to inhibit acute graft-versus-host disease (aGVHD). Because FTY720 functions through multiple S1P receptors, the mechanism of action through one or more of these receptors may account for its side effects. Thus, more selective S1P receptor modulators are needed to evaluate the roles of different S1P receptors and their therapeutic efficacies. In this study, we investigated the effect of an S1P1-selective agonist, CYM-5442, on the progression of aGVHD. We showed that CYM-5442 significantly inhibited but did not prevent aGVHD. CYM-5442 did not affect the infiltration of the donor T cells into the target organs, while the number of macrophages in GVHD organs was significantly reduced by CYM-5442 treatment. In vivo proliferation assays showed that the proliferation of macrophages was not suppressed by CYM-5442. Further studies using human endothelial cells demonstrated that CYM-5442 treatment downregulated CCL2 and CCL7 expression in endothelial cells, therefore reducing the migration of monocytes, from which tissue macrophages originate. Our data demonstrate the therapeutic efficacy of an S1P1-selective agonist in aGVHD and its possible mechanism of action. The results suggest that further investigations are needed regarding CYM-5442 as a potential therapeutic regimen for aGVHD.
Collapse
Affiliation(s)
- Qiao Cheng
- Laboratory of Cellular and Molecular Tumor Immunology, Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, China
| | - Shoubao Ma
- Laboratory of Cellular and Molecular Tumor Immunology, Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, China
| | - Dandan Lin
- Laboratory of Cellular and Molecular Tumor Immunology, Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, China
| | - Yu Mei
- Laboratory of Cellular and Molecular Tumor Immunology, Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, China
| | - Huanle Gong
- Laboratory of Cellular and Molecular Tumor Immunology, Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, China
| | - Lei Lei
- Laboratory of Cellular and Molecular Tumor Immunology, Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, China
| | - Yuanyuan Chen
- Laboratory of Cellular and Molecular Tumor Immunology, Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, China
| | - Ye Zhao
- Laboratory of Cellular and Molecular Tumor Immunology, Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, China.,Department of Hematology, First Affiliated Hospital of Soochow University, Suzhou, China
| | - Bo Hu
- Laboratory of Cellular and Molecular Tumor Immunology, Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, China
| | - Yan Wu
- Laboratory of Cellular and Molecular Tumor Immunology, Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, China
| | - Xiao Yu
- Laboratory of Cellular and Molecular Tumor Immunology, Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, China
| | - Lixiang Zhao
- Laboratory of Cellular and Molecular Tumor Immunology, Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, China
| | - Haiyan Liu
- Laboratory of Cellular and Molecular Tumor Immunology, Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, China.,Cyrus Tang Hematology Center, Jiangsu Institute of Hematology, First Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
30
|
Samarska IV, Bouma HR, Buikema H, Mungroop HE, Houwertjes MC, Absalom AR, Epema AH, Henning RH. S1P1 receptor modulation preserves vascular function in mesenteric and coronary arteries after CPB in the rat independent of depletion of lymphocytes. PLoS One 2014; 9:e97196. [PMID: 24819611 PMCID: PMC4018292 DOI: 10.1371/journal.pone.0097196] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2014] [Accepted: 04/16/2014] [Indexed: 11/25/2022] Open
Abstract
Background Cardiopulmonary bypass (CPB) may induce systemic inflammation and vascular dysfunction. Sphingosine 1-phosphate (S1P) modulates various vascular and immune responses. Here we explored whether agonists of the S1P receptors, FTY720 and SEW2871 improve vascular reactivity after CPB in the rat. Methods Experiments were done in male Wistar rats (total n = 127). Anesthesia was induced by isoflurane (2.5–3%) and maintained by fentanyl and midazolam during CPB. After catheterization of the left femoral artery, carotid artery and the right atrium, normothermic extracorporeal circulation was instituted for 60 minutes. In the first part of the study animals were euthanized after either 1 hour, 1 day, 2 or 5 days of the recovery period. In second part of the study animals were euthanized after 1 day of postoperative period. We evaluated the contractile response to phenylephrine (mesenteric arteries) or to serotonin (coronary artery) and vasodilatory response to acethylcholine (both arteries). Results Contractile responses to phenylephrine were reduced at 1 day recovery after CPB and Sham as compared to healthy control animals (Emax, mN: 7.9±1.9, 6.5±1.5, and 11.3±1.3, respectively). Mainly FTY720, but not SEW2871, caused lymphopenia in both Sham and CPB groups. In coronary and mesenteric arteries, both FTY720 and SEW2871 normalized serotonin and phenylephrine-mediated vascular reactivity after CPB (p<0.05) and FTY720 increased relaxation to acetylcholine as compared with untreated rats that underwent CPB. Conclusion Pretreatment with FTY720 or SEW2871 preserves vascular function in mesenteric and coronary artery after CPB. Therefore, pharmacological activation of S1P1 receptors may provide a promising therapeutic intervention to prevent CPB-related vascular dysfunction in patients.
Collapse
Affiliation(s)
- Iryna V. Samarska
- Department of Anesthesiology, University of Groningen, University Medical Center Groningen, The Netherlands
- * E-mail:
| | - Hjalmar R. Bouma
- Department of Clinical Pharmacy and Pharmacology, University of Groningen, University Medical Center Groningen, The Netherlands
| | - Hendrik Buikema
- Department of Clinical Pharmacy and Pharmacology, University of Groningen, University Medical Center Groningen, The Netherlands
| | - Hubert E. Mungroop
- Department of Anesthesiology, University of Groningen, University Medical Center Groningen, The Netherlands
| | - Martin C. Houwertjes
- Department of Anesthesiology, University of Groningen, University Medical Center Groningen, The Netherlands
| | - Anthony R. Absalom
- Department of Anesthesiology, University of Groningen, University Medical Center Groningen, The Netherlands
| | - Anne H. Epema
- Department of Anesthesiology, University of Groningen, University Medical Center Groningen, The Netherlands
| | - Robert H. Henning
- Department of Clinical Pharmacy and Pharmacology, University of Groningen, University Medical Center Groningen, The Netherlands
| |
Collapse
|
31
|
Cruz-Orengo L, Daniels BP, Dorsey D, Basak SA, Grajales-Reyes JG, McCandless EE, Piccio L, Schmidt RE, Cross AH, Crosby SD, Klein RS. Enhanced sphingosine-1-phosphate receptor 2 expression underlies female CNS autoimmunity susceptibility. J Clin Invest 2014; 124:2571-84. [PMID: 24812668 DOI: 10.1172/jci73408] [Citation(s) in RCA: 96] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2013] [Accepted: 03/20/2014] [Indexed: 11/17/2022] Open
Abstract
Multiple sclerosis (MS) is an inflammatory disease of the CNS that is characterized by BBB dysfunction and has a much higher incidence in females. Compared with other strains of mice, EAE in the SJL mouse strain models multiple features of MS, including an enhanced sensitivity of female mice to disease; however, the molecular mechanisms that underlie the sex- and strain-dependent differences in disease susceptibility have not been described. We identified sphingosine-1-phosphate receptor 2 (S1PR2) as a sex- and strain-specific, disease-modifying molecule that regulates BBB permeability by destabilizing adherens junctions. S1PR2 expression was increased in disease-susceptible regions of the CNS of both female SJL EAE mice and female patients with MS compared with their male counterparts. Pharmacological blockade or lack of S1PR2 signaling decreased EAE disease severity as the result of enhanced endothelial barrier function. Enhanced S1PR2 signaling in an in vitro BBB model altered adherens junction formation via activation of Rho/ROCK, CDC42, and caveolin endocytosis-dependent pathways, resulting in loss of apicobasal polarity and relocation of abluminal CXCL12 to vessel lumina. Furthermore, S1PR2-dependent BBB disruption and CXCL12 relocation were observed in vivo. These results identify a link between S1PR2 signaling and BBB polarity and implicate S1PR2 in sex-specific patterns of disease during CNS autoimmunity.
Collapse
MESH Headings
- Animals
- Autoimmunity/genetics
- Blood-Brain Barrier/immunology
- Blood-Brain Barrier/metabolism
- Case-Control Studies
- Central Nervous System/immunology
- Central Nervous System/metabolism
- Central Nervous System/pathology
- Encephalomyelitis, Autoimmune, Experimental/etiology
- Encephalomyelitis, Autoimmune, Experimental/genetics
- Encephalomyelitis, Autoimmune, Experimental/metabolism
- Female
- Gene Expression Profiling
- Genetic Predisposition to Disease
- Humans
- Male
- Mice
- Mice, 129 Strain
- Mice, Inbred C57BL
- Mice, Knockout
- Multiple Sclerosis/etiology
- Multiple Sclerosis/genetics
- Multiple Sclerosis/metabolism
- Receptors, Lysosphingolipid/deficiency
- Receptors, Lysosphingolipid/genetics
- Receptors, Lysosphingolipid/metabolism
- Sex Characteristics
- Species Specificity
- Sphingosine-1-Phosphate Receptors
Collapse
|
32
|
Abstract
Amenorrhea has not been reported as an adverse event in fingolimod phase III clinical trials in patients with multiple sclerosis (MS) with either 0.5 mg or 1.25 mg dosages. Here we report three cases of young women with MS who developed amenorrhea within 6 months of initiation of fingolimod. They experienced irregularities in their menstrual cycles in the first 3 months, which progressed to amenorrhea by 5th or 6th month. Gynecology evaluations showed no other etiologies. Menses returned to baseline after discontinuation of fingolimod for 2–3 months. Amenorrhea could be associated with fingolimod in the first year. Future surveillance is advised to determine the incidence rate of this adverse event.
Collapse
Affiliation(s)
- R Alroughani
- Division of Neurology, Department of Medicine, Amiri Hospital, Sharq, Kuwait
- Neurology Clinic, Dasman Diabetes Institute, Kuwait
| |
Collapse
|
33
|
Review: Novel insights into the regulation of vascular tone by sphingosine 1-phosphate. Placenta 2014; 35 Suppl:S86-92. [DOI: 10.1016/j.placenta.2013.12.006] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2013] [Revised: 12/10/2013] [Accepted: 12/11/2013] [Indexed: 11/17/2022]
|
34
|
Bigaud M, Guerini D, Billich A, Bassilana F, Brinkmann V. Second generation S1P pathway modulators: research strategies and clinical developments. Biochim Biophys Acta Mol Cell Biol Lipids 2013; 1841:745-58. [PMID: 24239768 DOI: 10.1016/j.bbalip.2013.11.001] [Citation(s) in RCA: 97] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2013] [Revised: 10/30/2013] [Accepted: 11/04/2013] [Indexed: 11/17/2022]
Abstract
Multiple Sclerosis (MS) is a chronic autoimmune disorder affecting the central nervous system (CNS) through demyelination and neurodegeneration. Until recently, major therapeutic treatments have relied on agents requiring injection delivery. In September 2010, fingolimod/FTY720 (Gilenya, Novartis) was approved as the first oral treatment for relapsing forms of MS. Fingolimod causes down-modulation of S1P1 receptors on lymphocytes which prevents the invasion of autoaggressive T cells into the CNS. In astrocytes, down-modulation of S1P1 by the drug reduces astrogliosis, a hallmark of MS, thereby allowing restoration of productive astrocyte communication with other neural cells and the blood brain barrier. Animal data further suggest that the drug directly supports the recovery of nerve conduction and remyelination. In human MS, such mechanisms may explain the significant decrease in the number of inflammatory markers on brain magnetic resonance imaging in recent clinical trials, and the reduction of brain atrophy by the drug. Fingolimod binds to 4 of the 5 known S1P receptor subtypes, and significant efforts were made over the past 5 years to develop next generation S1P receptor modulators and determine the minimal receptor selectivity needed for maximal therapeutic efficacy in MS patients. Other approaches considered were competitive antagonists of the S1P1 receptor, inhibitors of the S1P lyase to prevent S1P degradation, and anti-S1P antibodies. Below we discuss the current status of the field, and the functional properties of the most advanced compounds. This article is part of a Special Issue entitled New Frontiers in Sphingolipid Biology.
Collapse
Affiliation(s)
- Marc Bigaud
- Novartis Institutes for Biomedical Research, CH-4056 Basel, Switzerland.
| | - Danilo Guerini
- Novartis Institutes for Biomedical Research, CH-4056 Basel, Switzerland
| | - Andreas Billich
- Novartis Institutes for Biomedical Research, CH-4056 Basel, Switzerland
| | | | - Volker Brinkmann
- Novartis Institutes for Biomedical Research, CH-4056 Basel, Switzerland.
| |
Collapse
|
35
|
Stephenson M, Wong A, Rotella JA, Crump N, Kerr F, Greene SL. Deliberate fingolimod overdose presenting with delayed hypotension and bradycardia responsive to atropine. J Med Toxicol 2013; 10:215-8. [PMID: 24178903 DOI: 10.1007/s13181-013-0354-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
INTRODUCTION Fingolimod is an immunomodulating agent used in multiple sclerosis (MS). It is a sphingosine-1-phosphate (S1P) receptor agonist prescribed for relapsing forms of MS to delay onset of physical disability. As fingolimod is known to cause first-dose bradycardia, telemetry is recommended for the first 6 h post-dose. We present the first reported case of deliberate fingolimod overdose requiring atropine administration for bradycardia and hemodynamic instability. CASE REPORT A 33-year-old woman ingested 14 mg of fingolimod and 2 g of phenoxymethylpenicillin. After presenting to the emergency department 19 h later, she was initially hemodynamically stable (heart rate (HR) 60, blood pressure (BP) 113/89 mmHg). Two hours later, she then developed bradycardia (HR 48) and hypotension (87/57 mmHg). Despite intravenous fluids, stabilisation was only achieved after administration of atropine (300 μg). She was then admitted to the intensive care unit (ICU) for further monitoring where another episode of bradycardia and hypotension required atropine. She was monitored in the ICU for 48 h and then discharged on day 5 with no further episodes. DISCUSSION Fingolimod is known to cause bradycardia in the first 6 h post first therapeutic dose. Following intentional overdose, onset of bradycardia occurred at 21 h post-ingestion and was associated with hypotension. Atropine was successful in treating bradycardia and associated hypotension.
Collapse
Affiliation(s)
- M Stephenson
- Department of Emergency Medicine, Austin Hospital, Heidelberg, Victoria, Australia
| | | | | | | | | | | |
Collapse
|
36
|
Del Galdo S, Vettel C, Heringdorf DMZ, Wieland T. The activation of RhoC in vascular endothelial cells is required for the S1P receptor type 2-induced inhibition of angiogenesis. Cell Signal 2013; 25:2478-84. [PMID: 23993968 DOI: 10.1016/j.cellsig.2013.08.017] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2013] [Accepted: 08/24/2013] [Indexed: 12/12/2022]
Abstract
Sphingosine-1-phosphate (S1P) is a multifunctional phospholipid inducing a variety of cellular responses in endothelial cells (EC). S1P responses are mediated by five G protein coupled receptors of which three types (S1P1R-S1P3R) have been described to be of importance in vascular endothelial cells (EC). Whereas the S1P1R regulates endothelial barrier function by coupling to Gαi and the monomeric GTPase Rac1, the signaling pathways involved in the S1P-induced regulation of angiogenesis are ill defined. We therefore studied the sprouting of human umbilical vein EC (HUVEC) in vitro and analyzed the activation of the RhoGTPases RhoA and RhoC. Physiological relevant concentrations of S1P (100-300nM) induce a moderate activation of RhoA and RhoC. Inhibition or siRNA-mediated depletion of the S1P2R preferentially decreased the activation of RhoC. Both manipulations caused an increase of sprouting in a spheroid based in vitro sprouting assay. Interestingly, a similar increase in sprouting was detected after effective siRNA-mediated knockdown of RhoC. In contrast, the depletion of RhoA had no influence on sprouting. Furthermore, suppression of the activity of G proteins of the Gα12/13 subfamily by adenoviral overexpression of the regulator of G protein signaling domain of LSC as well as siRNA-mediated knockdown of the Rho specific guanine nucleotide exchange factor leukemia associated RhoGEF (LARG) inhibited the S1P-induced activation of RhoC and concomitantly increased sprouting of HUVEC with similar efficacy. We conclude that the angiogenic sprouting of EC is suppressed via the S1P2R subtype. Thus, the increase in basal sprouting can be attributed to blocking of the inhibitory action of autocrine S1P stimulating the S1P2R. This inhibitory pathway involves the activation of RhoC via Gα12/13 and LARG, while the simultaneously occurring activation of RhoA is apparently dispensable here.
Collapse
Affiliation(s)
- Sabrina Del Galdo
- Institute of Experimental and Clinical Pharmacology and Toxicology, Mannheim Medical Faculty, Heidelberg University, Maybachstrasse 14, 68169 Mannheim, Germany
| | | | | | | |
Collapse
|
37
|
Urina MA, Rojas C, Urina D. Efectos cardiovasculares de fingolimod en el tratamiento de la esclerosis múltiple. REVISTA COLOMBIANA DE CARDIOLOGÍA 2013. [DOI: 10.1016/s0120-5633(13)70044-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
|
38
|
Abstract
Sphingosine-1-phosphate (S1P) regulates important functions in cardiac and vascular homeostasis. It has been implied to play causal roles in the pathogenesis of many cardiovascular disorders such as coronary artery disease, atherosclerosis, myocardial infarction, and heart failure. The majority of S1P in plasma is associated with high-density lipoproteins (HDL), and their S1P content has been shown to be responsible, at least in part, for several of the beneficial effects of HDL on cardiovascular risk. The attractiveness of S1P-based drugs for potential cardiovascular applications is increasing in the wake of the clinical approval of FTY720, but answers to important questions on the effects of S1P in cardiovascular biology and medicine must still be found. This chapter focuses on the current understanding of the role of S1P and its receptors in cardiovascular physiology, pathology, and disease.
Collapse
Affiliation(s)
- Bodo Levkau
- University of Duisburg-Essen, Essen, Germany.
| |
Collapse
|
39
|
Spijkers LJA, Alewijnse AE, Peters SLM. FTY720 (fingolimod) increases vascular tone and blood pressure in spontaneously hypertensive rats via inhibition of sphingosine kinase. Br J Pharmacol 2012; 166:1411-8. [PMID: 22251137 DOI: 10.1111/j.1476-5381.2012.01865.x] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
BACKGROUND AND PURPOSE FTY720 (Fingolimod) is a recently approved orally administered drug for the treatment of multiple sclerosis. Phase II and III clinical trials have demonstrated that this drug modestly increases BP. We previously showed that inhibition of sphingosine kinase increases vascular tone and BP in hypertensive, but not normotensive rats. Since FTY720 is reported to have inhibitory effects on sphingosine kinase, we investigated whether FTY720 increases vascular tone and BP only in hypertensive rats via this mechanism. EXPERIMENTAL APPROACH The contractile and BP modulating effects of FTY720 were studied in vivo and ex vivo (wire myography) in age-matched normotensive Wistar Kyoto (WKY) rats and spontaneously hypertensive rats (SHRs). KEY RESULTS Oral administration of FTY720 induced an increase in mean arterial pressure in SHR, whereas a decrease in BP was observed in WKY rats, as measured 24 h after administration. Similar to the sphingosine kinase inhibitor dimethylsphingosine (DMS), FTY720 induced large contractions in isolated carotid arteries from SHR, but not in those from WKY. In contrast, the phosphorylated form of FTY720 did not induce contractions in isolated carotid arteries from SHR. FTY720-induced contractions were inhibited by endothelium denudation, COX and thromboxane synthase inhibitors, and by thromboxane receptor antagonism, indicating that (like DMS-induced contractions) they were endothelium-dependent and mediated by thromboxane A₂. CONCLUSIONS AND IMPLICATIONS These data demonstrate that FTY720 increases vascular tone and BP only in hypertensive rats, most likely as a result of its inhibitory effect on sphingosine kinase.
Collapse
Affiliation(s)
- Léon J A Spijkers
- Department of Pharmacology & Pharmacotherapy, Academic Medical Center, Amsterdam, The Netherlands
| | | | | |
Collapse
|
40
|
Ontaneda D, Cohen JA. Potential mechanisms of efficacy and adverse effects in the use of fingolimod (FTY720). Expert Rev Clin Pharmacol 2012; 4:567-70. [PMID: 22114884 DOI: 10.1586/ecp.11.46] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
The interactions between sphingosine 1-phosphate (S1P) and S1P receptors (S1PRs) mediate a wide range of biological functions in the CNS and immune and cardiovascular systems. Fingolimod (FTY720), an S1PR modulator with potent immunomodulatory effects, was recently approved to treat multiple sclerosis. Some adverse effects of fingolimod reflecting vascular leak phenomena may be mediated through endothelial S1PRs, particularly macular edema. Oo et al. characterized the molecular interactions of fingolimod phosphate and S1PR type 1 (S1P(1)) leading to functional antagonism - phosphorylation of S1P(1) with subsequent receptor complex internalization, polyubiquitinylation and degradation. Differences along the pharmacological pathways that mediate vascular leak and lymphopenia were demonstrated, suggesting that distinct S1P(1) mechanisms mediate the adverse effects and efficacy of fingolimod.
Collapse
Affiliation(s)
- Daniel Ontaneda
- The Mellen Center for Multiple Sclerosis Treatment and Research, Neurological Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | | |
Collapse
|
41
|
Sphingosine 1-phosphate protects primary human keratinocytes from apoptosis via nitric oxide formation through the receptor subtype S1P₃. Mol Cell Biochem 2012; 371:165-76. [PMID: 22899173 DOI: 10.1007/s11010-012-1433-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2012] [Accepted: 08/03/2012] [Indexed: 12/20/2022]
Abstract
Although the lipid mediator sphingosine 1-phosphate (S1P) has been identified to induce cell growth arrest of human keratinocytes, the sphingolipid effectively protects these epidermal cells from apoptosis. The molecular mechanism of the anti-apoptotic action induced by S1P is less characterized. Apart from S1P, endogenously produced nitric oxide (NO•) has been recognized as a potent modulator of apoptosis in keratinocytes. Therefore, it was of great interest to elucidate whether S1P protects human keratinocytes via a NO•-dependent signalling pathway. Indeed, S1P induced an activation of endothelial nitric oxide synthase (eNOS) in human keratinocytes leading to an enhanced formation of NO•. Most interestingly, the cell protective effect of S1P was almost completely abolished in the presence of the eNOS inhibitor L-NAME as well as in eNOS-deficient keratinocytes indicating that the sphingolipid metabolite S1P protects human keratinocytes from apoptosis via eNOS activation and subsequent production of protective amounts of NO•. It is well established that most of the known actions of S1P are mediated by a family of five specific G protein-coupled receptors. Therefore, the involvement of S1P-receptor subtypes in S1P-mediated eNOS activation has been examined. Indeed, this study clearly shows that the S1P(3) is the exclusive receptor subtype in human keratinocytes which mediates eNOS activation and NO• formation in response to S1P. In congruence, when the S1P(3) receptor subtype is abrogated, S1P almost completely lost its ability to protect human keratinocytes from apoptosis.
Collapse
|
42
|
Bajwa A, Huang L, Ye H, Dondeti K, Song S, Rosin DL, Lynch KR, Lobo PI, Li L, Okusa MD. Dendritic cell sphingosine 1-phosphate receptor-3 regulates Th1-Th2 polarity in kidney ischemia-reperfusion injury. THE JOURNAL OF IMMUNOLOGY 2012; 189:2584-96. [PMID: 22855711 DOI: 10.4049/jimmunol.1200999] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Dendritic cells (DCs) are central to innate and adaptive immunity of early kidney ischemia-reperfusion injury (IRI), and strategies to alter DC function may provide new therapeutic opportunities. Sphingosine 1-phosphate (S1P) modulates immunity through binding to its receptors (S1P1-5), and protection from kidney IRI occurs in S1P3-deficient mice. Through a series of experiments we determined that this protective effect was owing in part to differences between S1P3-sufficient and -deficient DCs. Mice lacking S1P3 on bone marrow cells were protected from IRI, and S1P3-deficient DCs displayed an immature phenotype. Wild-type (WT) but not S1P3-deficient DCs injected into mice depleted of DCs prior to kidney IR reconstituted injury. Adoptive transfer (i.e., i.v. injection) of glycolipid (Ag)-loaded WT but not S1P3-deficient DCs into WT mice exacerbated IRI, suggesting that WT but not S1P3-deficient DCs activated NKT cells. Whereas WT DC transfers activated the Th1/IFN-γ pathway, S1P3-deficient DCs activated the Th2/IL-4 pathway, and an IL-4-blocking Ab reversed protection from IRI, supporting the concept that IL-4 mediates the protective effect of S1P3-deficient DCs. Administration of S1P3-deficient DCs 7 d prior to or 3 h after IRI protected mice from IRI and suggests their potential use in cell-based therapy. We conclude that absence of DC S1P3 prevents DC maturation and promotes a Th2/IL-4 response. These findings highlight the importance of DC S1P3 in modulating NKT cell function and IRI and support development of selective S1P3 antagonists for tolerizing DCs for cell-based therapy or for systemic administration for the prevention and treatment of IRI and autoimmune diseases.
Collapse
Affiliation(s)
- Amandeep Bajwa
- Department of Medicine, University of Virginia, Charlottesville, VA 22908, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Choi JW, Chun J. Lysophospholipids and their receptors in the central nervous system. Biochim Biophys Acta Mol Cell Biol Lipids 2012; 1831:20-32. [PMID: 22884303 DOI: 10.1016/j.bbalip.2012.07.015] [Citation(s) in RCA: 194] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2012] [Revised: 07/17/2012] [Accepted: 07/18/2012] [Indexed: 02/05/2023]
Abstract
Lysophosphatidic acid (LPA) and sphingosine 1-phosphate (S1P), two of the best-studied lysophospholipids, are known to influence diverse biological events, including organismal development as well as function and pathogenesis within multiple organ systems. These functional roles are due to a family of at least 11 G protein-coupled receptors (GPCRs), named LPA(1-6) and S1P(1-5), which are widely distributed throughout the body and that activate multiple effector pathways initiated by a range of heterotrimeric G proteins including G(i/o), G(12/13), G(q) and G(s), with actual activation dependent on receptor subtypes. In the central nervous system (CNS), a major locus for these signaling pathways, LPA and S1P have been shown to influence myriad responses in neurons and glial cell types through their cognate receptors. These receptor-mediated activities can contribute to disease pathogenesis and have therapeutic relevance to human CNS disorders as demonstrated for multiple sclerosis (MS) and possibly others that include congenital hydrocephalus, ischemic stroke, neurotrauma, neuropsychiatric disorders, developmental disorders, seizures, hearing loss, and Sandhoff disease, based upon the experimental literature. In particular, FTY720 (fingolimod, Gilenya, Novartis Pharma, AG) that becomes an analog of S1P upon phosphorylation, was approved by the FDA in 2010 as a first oral treatment for MS, validating this class of receptors as medicinal targets. This review will provide an overview and update on the biological functions of LPA and S1P signaling in the CNS, with a focus on results from studies using genetic null mutants for LPA and S1P receptors. This article is part of a Special Issue entitled Advances in Lysophospholipid Research.
Collapse
Affiliation(s)
- Ji Woong Choi
- Department of Molecular Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | | |
Collapse
|
44
|
Krishna SM, Seto SW, Moxon JV, Rush C, Walker PJ, Norman PE, Golledge J. Fenofibrate increases high-density lipoprotein and sphingosine 1 phosphate concentrations limiting abdominal aortic aneurysm progression in a mouse model. THE AMERICAN JOURNAL OF PATHOLOGY 2012; 181:706-18. [PMID: 22698985 DOI: 10.1016/j.ajpath.2012.04.015] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/30/2011] [Revised: 04/03/2012] [Accepted: 04/12/2012] [Indexed: 02/03/2023]
Abstract
There are currently no acceptable treatments to limit progression of abdominal aortic aneurysm (AAA). Increased serum concentrations of high-density lipoprotein (HDL) are associated with reduced risk of developing an AAA. The present study aimed to assess the effects of fenofibrate on aortic dilatation in a mouse model of AAA. Male low-density lipoprotein receptor-deficient (Ldlr(-/-)) mice were maintained on a high-fat diet for 3 weeks followed by 6 weeks of oral administration of vehicle or fenofibrate. From 14 to 18 weeks of age, all mice were infused with angiotensin II (AngII). At 18 weeks of age, blood and aortas were collected for assessment of serum lipoproteins, aortic pathology, aortic Akt1 and endothelial nitric oxide synthase (eNOS) activities, immune cell infiltration, eNOS and inducible NOS (iNOS) expression, sphingosine 1 phosphate (S1P) receptor status, and apoptosis. Mice receiving fenofibrate had reduced suprarenal aortic diameter, reduced aortic arch Sudan IV staining, higher serum HDL levels, increased serum S1P concentrations, and increased aortic Akt1 and eNOS activities compared with control mice. Macrophages, T lymphocytes, and apoptotic cells were less evident and eNOS, iNOS, and S1P receptors 1 and 3 were up-regulated in aortas from mice receiving fenofibrate. The present findings suggest that fenofibrate antagonizes AngII-induced AAA and atherosclerosis by up-regulating serum HDL and S1P levels, with associated activation of NO-producing enzymes and reduction of aortic inflammation.
Collapse
Affiliation(s)
- Smriti M Krishna
- Vascular Biology Unit, School of Medicine and Dentistry, James Cook University, Townsville, QLD, Australia
| | | | | | | | | | | | | |
Collapse
|
45
|
Shimizu T, De Wispelaere A, Winkler M, D'Souza T, Caylor J, Chen L, Dastvan F, Deou J, Cho A, Larena-Avellaneda A, Reidy M, Daum G. Sphingosine-1-phosphate receptor 3 promotes neointimal hyperplasia in mouse iliac-femoral arteries. Arterioscler Thromb Vasc Biol 2012; 32:955-61. [PMID: 22308044 DOI: 10.1161/atvbaha.111.241034] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE The objective of this study was to define a role for sphingosine-1-phosphate receptor 3 (S1PR3) in intimal hyperplasia. METHODS AND RESULTS A denudation model of the iliac-femoral artery in wild-type and S1PR3-null mice was used to define a role for S1PR3 in the arterial injury response because we found in humans and mice that expression of S1PR3 was higher in these arteries compared with carotid arteries. At 28 days after surgery, wild-type arteries formed significantly larger lesions than S1PR3-null arteries. Bromodeoxyuridine labeling experiments demonstrated that on injury, wild-type arteries exhibited higher medial as well as intimal proliferation than S1PR3-null arteries. Because S1PR3 expression in vitro was low, we expressed S1PR3 in S1PR3-null smooth muscle cells (SMCs) using retroviral-mediated gene transfer to study the effects of S1PR3 on cell functions and signaling. SMCs expressing S1PR3, but not vector-transfected controls, responded to sphingosine-1-phosphate stimulation with activation of Rac, Erk, and Akt. SMCs expressing S1PR3 also migrated more. CONCLUSIONS In humans and mice, S1PR3 expression was higher in iliac-femoral arteries compared with carotid arteries. S1PR3 promoted neointimal hyperplasia on denudation of iliac-femoral arteries in mice, likely by stimulating cell migration and proliferation through activation of signaling pathways involving Erk, Akt, and Rac.
Collapse
Affiliation(s)
- Takuya Shimizu
- Department of Pathology, University of Washington, Seattle, 98109, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Schuchardt M, Tölle M, Prüfer J, van der Giet M. Pharmacological relevance and potential of sphingosine 1-phosphate in the vascular system. Br J Pharmacol 2011; 163:1140-62. [PMID: 21309759 DOI: 10.1111/j.1476-5381.2011.01260.x] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Sphingosine-1-phosphate (S1P) was identified as a crucial molecule for regulating immune responses, inflammatory processes as well as influencing the cardiovascular system. S1P mediates differentiation, proliferation and migration during vascular development and homoeostasis. S1P is a naturally occurring lipid metabolite and is present in human blood in nanomolar concentrations. S1P is not only involved in physiological but also in pathophysiological processes. Therefore, this complex signalling system is potentially interesting for pharmacological intervention. Modulation of the system might influence inflammatory, angiogenic or vasoregulatory processes. S1P activates G-protein coupled receptors, namely S1P(1-5) , whereas only S1P(1-3) is present in vascular cells. S1P can also act as an intracellular signalling molecule. This review highlights the pharmacological potential of S1P signalling in the vascular system by giving an overview of S1P-mediated processes in endothelial cells (ECs) and vascular smooth muscle cells (VSMCs). After a short summary of S1P metabolism and signalling pathways, the role of S1P in EC and VSMC proliferation and migration, the cause of relaxation and constriction of arterial blood vessels, the protective functions on endothelial apoptosis, as well as the regulatory function in leukocyte adhesion and inflammatory responses are summarized. This is followed by a detailed description of currently known pharmacological agonists and antagonists as new tools for mediating S1P signalling in the vasculature. The variety of effects influenced by S1P provides plenty of therapeutic targets currently under investigation for potential pharmacological intervention.
Collapse
Affiliation(s)
- Mirjam Schuchardt
- Charité- Universitätsmedizin Berlin, CharitéCentrum 10, Department of Nephrology, Campus Benjamin Franklin, Hindenburgdamm 30, Berlin, Germany
| | | | | | | |
Collapse
|
47
|
Yonesu K, Nakamura T, Mizuno Y, Suzuki C, Nagayama T, Satoh S, Nara F. A novel sphingosine-1-phosphate receptor 1 antagonist prevents the proliferation and relaxation of vascular endothelial cells by sphingosine-1-phosphate. Biol Pharm Bull 2011; 33:1500-5. [PMID: 20823564 DOI: 10.1248/bpb.33.1500] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
A sphingosine-1-phosphate receptor 1 (S1P1) antagonist is expected to be an anti-angiogenic compound; however, there are few reports that demonstrated that a S1P1 inhibitor improved the disease state in an angiogenic animal model. Since we determined that a prototype S1P1 antagonist was an in vivo angiogenesis inhibitor, we developed the derivatives to acquire more effective compounds. In this report, we show the S1P1 antagonistic activity of some representatives, especially compound 5 {sodium 4-[(4-butoxyphenyl)thio]-2'-[{4-[(heptylthio)methyl]-2-hydroxyphenyl}(hydroxy)methyl]biphenyl-3-sulfonate}. The IC50 values calculated from an intracellular cyclic AMP measurement assay and a [33P]sphingosine-1-phosphate (Sph-1-P)/S1P1 binding assay were 38 and 200 nM, respectively. A subtype specificity test for the other Sph-1-P receptors showed that compound 5 was the S1P1-directional antagonist. It also inhibited the proliferation, migration, and tube formation of human umbilical vein endothelial cells stimulated by Sph-1-P with the IC50 values of 18, 650, and 230 nM, respectively. A cytotoxicity assay concurrently performed with a tube formation assay supported the hypothesis that these biological effects were not due to its cytotoxicity. Furthermore, administration (10 mg/kg, intravenously) to anesthetized Sprague-Dawley rats inhibited Sph-1-P-induced hypotension by 100-90% for 30 min. This is presumably through the inhibition of Sph-1-P-induced vasorelaxation, mainly by the blocking of S1P1 and/or S1P3. Taken together, these results show that compound 5 is an inhibitor of in vitro and in vivo Sph-1-P signaling, and that it will be useful to elucidate the in vivo effect of Sph-1-P on vascular endothelial cells.
Collapse
Affiliation(s)
- Kiyoaki Yonesu
- Cardiovascular-Metabolics Research Laboratories, Daiichi Sankyo Co., Ltd., Tokyo, Japan.
| | | | | | | | | | | | | |
Collapse
|
48
|
Cohen JA, Chun J. Mechanisms of fingolimod's efficacy and adverse effects in multiple sclerosis. Ann Neurol 2011; 69:759-77. [PMID: 21520239 DOI: 10.1002/ana.22426] [Citation(s) in RCA: 280] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Until recently, all approved multiple sclerosis (MS) disease treatments were administered parenterally. Oral fingolimod was approved in September 2010 by the US Food and Drug Administration to reduce relapses and disability progression in relapsing forms of MS. In the clinical trials that led to approval, fingolimod reduced not only acute relapses and magnetic resonance imaging lesion activity but also disability progression and brain volume loss, suggesting preservation of tissue. Fingolimod's mechanism of action in MS is not known with certainty. Its active form, fingolimod-phosphate (fingolimod-P), is a sphingosine 1-phosphate receptor (S1PR) modulator that inhibits egress of lymphocytes from lymph nodes and their recirculation, potentially reducing trafficking of pathogenic cells into the central nervous system (CNS). Fingolimod also readily penetrates the CNS, and fingolimod-P formed in situ may have direct effects on neural cells. Fingolimod potently inhibits the MS animal model, experimental autoimmune encephalomyelitis, but is ineffective in mice with selective deficiency of the S1P₁ S1PR subtype on astrocytes despite normal expression in the immune compartment. These findings suggest that S1PR modulation by fingolimod in both the immune system and CNS, producing a combination of beneficial anti-inflammatory and possibly neuroprotective/reparative effects, may contribute to its efficacy in MS. In clinical trials, fingolimod was generally safe and well tolerated. Its interaction with S1PRs in a variety of tissues largely accounts for the reported adverse effects, which were seen more frequently with doses 2.5 to 10x the approved 0.5 mg dose. Fingolimod's unique mechanism of action distinguishes it from all other currently approved MS therapies.
Collapse
Affiliation(s)
- Jeffrey A Cohen
- Mellen Center for Multiple Sclerosis Treatment and Research, Neurological Institute, Cleveland Clinic, Cleveland, OH 44195, USA.
| | | |
Collapse
|
49
|
Thennes T, Mehta D. Heterotrimeric G proteins, focal adhesion kinase, and endothelial barrier function. Microvasc Res 2011; 83:31-44. [PMID: 21640127 DOI: 10.1016/j.mvr.2011.05.004] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2011] [Revised: 05/04/2011] [Accepted: 05/12/2011] [Indexed: 12/18/2022]
Abstract
Ligands by binding to G protein coupled receptors (GPCRs) stimulate dissociation of heterotrimeric G proteins into Gα and Gβγ subunits. Released Gα and Gβγ subunits induce discrete signaling cues that differentially regulate focal adhesion kinase (FAK) activity and endothelial barrier function. Activation of G proteins downstream of receptors such as protease activated receptor 1 (PAR1) and histamine receptors rapidly increases endothelial permeability which reverses naturally within the following 1-2 h. However, activation of G proteins coupled to the sphingosine-1-phosphate receptor 1 (S1P1) signal cues that enhance basal barrier endothelial function and restore endothelial barrier function following the increase in endothelial permeability by edemagenic agents. Intriguingly, both PAR1 and S1P1 activation stimulates FAK activity, which associates with alteration in endothelial barrier function by these agonists. In this review, we focus on the role of the G protein subunits downstream of PAR1 and S1P1 in regulating FAK activity and endothelial barrier function.
Collapse
Affiliation(s)
- Tracy Thennes
- Department of Pharmacology, University of Illinois at Chicago, Chicago, IL 60612, USA
| | | |
Collapse
|
50
|
Sphingosine-1-Phosphate-Specific G Protein-Coupled Receptors as Novel Therapeutic Targets for Atherosclerosis. Pharmaceuticals (Basel) 2011. [PMCID: PMC4052545 DOI: 10.3390/ph4010117] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
Atherosclerosis is a chronic inflammatory process involving complex interactions of modified lipoproteins, monocyte-derived macrophages or foam cells, lymphocytes, endothelial cells (ECs), and vascular smooth muscle cells. Sphingosine-1-phosphate (S1P), a biologically active blood-borne lipid mediator, exerts pleiotropic effects such as cell proliferation, migration and cell-cell adhesion in a variety of cell types via five members of S1P-specific high-affinity G protein-coupled receptors (S1P1-S1P5). Among them, S1P1, S1P2 and S1P3 are major receptor subtypes which are widely expressed in various tissues. Available evidence suggest that S1P and HDL-bound S1P exert atheroprotective effects including inhibition of leukocyte adhesion and stimulation of endothelial nitric oxide synthase (eNOS) in endothelial cells (ECs) through the activation of Gi signaling pathway via S1P3 and probably S1P1, although there is still controversy. FTY720, the phosphorylation product of which is a high-affinity agonist for all S1P receptors except S1P2 and act as an immunosuppressant by downregulating S1P1 on lymphocytes, inhibits atherosclerosis in LDL receptor-null mice and apoE-null mice through the inhibition of lymphocyte and macrophage functions and probably stimulation of EC functions, without influencing plasma lipid concentrations. In contrast to S1P1 and S1P3, S1P2 facilitates atherosclerosis by activating G12/13-Rho-Rho kinase (ROCK) in apoE-null mice. S1P2 mediates transmigration of monocytes into the arterial intima, oxidized LDL accumulation and cytokine secretion in monocyte-derived macrophages, and eNOS inhibition and cytokine secretion in ECs through Rac inhibition, NF-κB activation and 3′-specific phosphoinositide phosphatase (PTEN) stimulation downstream of G12/13-Rho-ROCK. Systemic long-term administration of a selective S1P2-blocker remarkably inhibits atherosclerosis without overt toxicity. Thus, multiple S1P receptors positively and negatively regulate atherosclerosis through multitudes of mechanisms. Considering the essential and multi-faceted role of S1P2 in atherogenesis and the impact of S1P2 inactivation on atherosclerosis, S1P2 is a particularly promising therapeutic target for atherosclerosis.
Collapse
|