1
|
Wang L, Kong Q, Leng X, Leung H, Li Y. The sphingosine-1-phosphate signaling pathway (sphingosine-1-phosphate and its receptor, sphingosine kinase) and epilepsy. Epilepsia Open 2024. [PMID: 39727628 DOI: 10.1002/epi4.13112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 11/08/2024] [Accepted: 11/20/2024] [Indexed: 12/28/2024] Open
Abstract
Epilepsy is one of the common chronic neurological diseases, affecting more than 70 million people worldwide. The brains of people with epilepsy exhibit a pathological and persistent propensity for recurrent seizures. Epilepsy often coexists with cardiovascular disease, cognitive dysfunction, depression, etc., which seriously affects the patient's quality of life. Although our understanding of epilepsy has advanced, the pathophysiological mechanisms leading to epileptogenesis, drug resistance, and associated comorbidities remain largely unknown. The use of newer antiepileptic drugs has increased, but this has not improved overall outcomes. We need to deeply study the pathogenesis of epilepsy and find drugs that can not only prevent the epileptogenesis and interfere with the process of epileptogenesis but also treat epilepsy comorbidities. Sphingosine-1-phosphate (S1P) is an important lipid molecule. It not only forms the basis of cell membranes but is also an important bioactive mediator. It can not only act as a second messenger in cells to activate downstream signaling pathways but can also exert biological effects by being secreted outside cells and binding to S1P receptors on the cell membrane. Fingolimod (FTY720) is the first S1P receptor modulator developed and approved for the treatment of multiple sclerosis. More and more studies have proven that the S1P signaling pathway is closely related to epilepsy, drug-resistant epilepsy, epilepsy comorbidities, or other epilepsy-causing diseases. However, there is much controversy over the role of certain natural molecules in the pathway and receptor modulators (such as FTY720) in epilepsy. Here, we summarize and analyze the role of the S1P signaling pathway in epilepsy, provide a basis for finding potential therapeutic targets and/or epileptogenic biomarkers, analyze the reasons for these controversies, and put forward our opinions. PLAIN LANGUAGE SUMMARY: This article combines the latest research literature at home and abroad to review the sphingosine 1-phosphate signaling pathway and epileptogenesis, drug-resistant epilepsy, epilepsy comorbidities, other diseases that can cause epilepsy, as well as the sphingosine-1-phosphate signaling pathway regulators and epilepsy, with the expectation of providing a certain theoretical basis for finding potential epilepsy treatment targets and/or epileptogenic biomarkers in the sphingosine-1-phosphate signaling pathway.
Collapse
Affiliation(s)
- Lin Wang
- Department of Neurology, Affiliated Hospital of Jining Medical University, Jining City, China
- Epilepsy Center, Affiliated Hospital of Jining Medical University, Jining City, China
- The Chinese University of Hong Kong, Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Central Ave, Hong Kong, Hong Kong
| | - Qingxia Kong
- Department of Neurology, Affiliated Hospital of Jining Medical University, Jining City, China
- Epilepsy Center, Affiliated Hospital of Jining Medical University, Jining City, China
| | - Xinyi Leng
- The Chinese University of Hong Kong, Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Central Ave, Hong Kong, Hong Kong
| | - Howan Leung
- Division of Neurology, Department of Medicine and Therapeutics, Prince of Wales Hospital, 7/F Clinical Science Building, Prince of Wales Hospital, Hong Kong, Hong Kong
| | - Yang Li
- Department of Oncology, Affiliated Hospital of Jining Medical University, Jining City, China
| |
Collapse
|
2
|
Roggeri A, Olivero G, Usai C, Vanmierlo T, Pittaluga A. Presynaptic Release-Regulating Sphingosine 1-Phosphate 1/3 Receptors in Cortical Glutamatergic Terminals: Adaptations in EAE Mice and Impact of Therapeutic FTY720. Cells 2023; 12:2343. [PMID: 37830557 PMCID: PMC10571862 DOI: 10.3390/cells12192343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 09/13/2023] [Accepted: 09/20/2023] [Indexed: 10/14/2023] Open
Abstract
This study provides evidence of the existence of presynaptic inhibitory sphingosine-1-phosphate receptor 1 (S1P1R) and facilitatory S1P3R in cortical nerve endings (synaptosomes) of healthy mice. The conclusion relies on the findings that (i) the S1P1R agonist CS-2100 (0.1-30 nM) inhibits the 12 mM KCl-evoked glutamate exocytosis (quantified as the release of [3H]D-aspartate) while the S1P3R allosteric agonist CYM-5541 potentiates it and (ii) these effects are inhibited by the S1P1R antagonist Ex 26 (30-300 nM) and the S1P3R antagonist TY-52156 (100-1000 nM), respectively. Confocal microscopy and western blot analysis confirmed the presence of S1P1R and S1P3R proteins in cortical glutamatergic synaptosomes, which were scarcely accessible to biotin in a biotinylation study. Then, we demonstrated that S1P1R and S1P3R densities and their release activity are amplified in cortical synaptosomes of mice suffering from experimental autoimmune encephalomyelitis (EAE), despite receptors maintain their preferential internal distribution. Receptor changes recover following chronic oral therapeutic FTY720 (0.03 mg/Kg/day). These results improve our knowledge of the role of presynaptic release-regulating S1P1Rs and S1P3Rs controlling glutamate transmission in the CNS also unravelling functional adaptations during EAE that recover following chronic FTY720. In a whole, these findings provide new information on the central neuroprotectant activities of FTY720.
Collapse
Affiliation(s)
- Alessandra Roggeri
- Department of Pharmacy (DiFar), University of Genoa, Viale Cembrano 4, 16148 Genoa, Italy; (A.R.); (G.O.)
| | - Guendalina Olivero
- Department of Pharmacy (DiFar), University of Genoa, Viale Cembrano 4, 16148 Genoa, Italy; (A.R.); (G.O.)
| | - Cesare Usai
- Institute of Biophysics, National Research Council, Via De Marini 6, 16149 Genoa, Italy;
| | - Tim Vanmierlo
- Department of Neuroscience, Biomedical Research Institute, European Graduate School of Neuroscience, Hasselt University, B-3590 Hasselt, Belgium;
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neurosciences, Division Translational Neuroscience, Maastricht University, 6200 MD Maastricht, The Netherlands
| | - Anna Pittaluga
- Department of Pharmacy (DiFar), Center of Excellence for Biomedical Research, 3Rs Center, University of Genoa, Viale Cembrano 4, 16148 Genoa, Italy
- IRCCS Ospedale Policlinico San Martino, 16145 Genoa, Italy
| |
Collapse
|
3
|
van Echten-Deckert G. The role of sphingosine 1-phosphate metabolism in brain health and disease. Pharmacol Ther 2023; 244:108381. [PMID: 36907249 DOI: 10.1016/j.pharmthera.2023.108381] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 03/02/2023] [Accepted: 03/06/2023] [Indexed: 03/13/2023]
Abstract
Lipids are essential structural and functional components of the central nervous system (CNS). Sphingolipids are ubiquitous membrane components which were discovered in the brain in the late 19th century. In mammals, the brain contains the highest concentration of sphingolipids in the body. Sphingosine 1-phosphate (S1P) derived from membrane sphingolipids evokes multiple cellular responses which, depending on its concentration and localization, make S1P a double-edged sword in the brain. In the present review we highlight the role of S1P in brain development and focus on the often contrasting findings regarding its contributions to the initiation, progression and potential recovery of different brain pathologies, including neurodegeneration, multiple sclerosis (MS), brain cancers, and psychiatric illnesses. A detailed understanding of the critical implications of S1P in brain health and disease may open the door for new therapeutic options. Thus, targeting S1P-metabolizing enzymes and/or signaling pathways might help overcome, or at least ameliorate, several brain illnesses.
Collapse
|
4
|
Al-Otaibi KM, Alghamdi BS, Al-Ghamdi MA, Mansouri RA, Ashraf GM, Omar UM. Therapeutic effect of combination vitamin D3 and siponimod on remyelination and modulate microglia activation in cuprizone mouse model of multiple sclerosis. Front Behav Neurosci 2023; 16:1068736. [PMID: 36688131 PMCID: PMC9849768 DOI: 10.3389/fnbeh.2022.1068736] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Accepted: 11/29/2022] [Indexed: 01/07/2023] Open
Abstract
Stimulation of remyelination is critical for the treatment of multiple sclerosis (MS) to alleviate symptoms and protect the myelin sheath from further damage. The current study aimed to investigate the possible therapeutic effects of combining vitamin D3 (Vit D3) and siponimod (Sipo) on enhancing remyelination and modulating microglia phenotypes in the cuprizone (CPZ) demyelination mouse model. The study was divided into two stages; demyelination (first 5 weeks) and remyelination (last 4 weeks). In the first 5 weeks, 85 mice were randomly divided into two groups, control (n = 20, standard rodent chow) and CPZ (n = 65, 0.3% CPZ mixed with chow for 6 weeks, followed by 3 weeks of standard rodent chow). At week 5, the CPZ group was re-divided into four groups (n = 14) for remyelination stages; untreated CPZ (0.2 ml of CMC orally), CPZ+Vit D3 (800 IU/kg Vit D3 orally), CPZ+Sipo (1.5 mg/kg Sipo orally), and CPZ+Vit D3 (800 IU/kg Vit D3) + Sipo (1.5 mg/kg Sipo orally). Various behavioral tasks were performed to evaluate motor performance. Luxol Fast Blue (LFB) staining, the expression level of myelin basic protein (MBP), and M1/M2 microglia phenotype genes were assessed in the corpus callosum (CC). The results showed that the combination of Vit D3 and Sipo improved behavioral deficits, significantly promoted remyelination, and modulated expression levels of microglia phenotype genes in the CC at early and late remyelination stages. These results demonstrate for the first time that a combination of Vit D3 and Sipo can improve the remyelination process in the cuprizone (CPZ) mouse model by attenuating the M1 microglia phenotype. This may help to improve the treatment of MS patients.
Collapse
Affiliation(s)
- Kholoud M. Al-Otaibi
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia,Department of Chemistry, Faculty of Science, Albaha University, Albaha, Saudi Arabia,*Correspondence: Badrah S. Alghamdi Kholoud M. Al-Otaibi
| | - Badrah S. Alghamdi
- Department of Physiology, Neuroscience Unit, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia,Pre-Clinical Research Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia,*Correspondence: Badrah S. Alghamdi Kholoud M. Al-Otaibi
| | - Maryam A. Al-Ghamdi
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia,Vitamin D Pharmacogenomics Research Group, King Abdulaziz University, Jeddah, Saudi Arabia,Experimental Biochemistry Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Rasha A. Mansouri
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Ghulam Md Ashraf
- Pre-Clinical Research Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia,Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Ulfat M. Omar
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia,Princess Dr. Najla Bint Saud Al-Saud Center for Excellence Research in Biotechnology, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
5
|
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: 1.7] [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
|
6
|
Lauro F, Giancotti LA, Kolar G, Harada CM, Harmon TA, Garrett TJ, Salvemini D. Role of Adenosine Kinase in Sphingosine-1-Phosphate Receptor 1-Induced Mechano-Hypersensitivities. Cell Mol Neurobiol 2022; 42:2909-2918. [PMID: 34773542 PMCID: PMC9098694 DOI: 10.1007/s10571-021-01162-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Accepted: 10/22/2021] [Indexed: 12/21/2022]
Abstract
Emerging evidence implicates the sphingosine-1-phosphate receptor subtype 1 (S1PR1) in the development of neuropathic pain. Continued investigation of the signaling pathways downstream of S1PR1 are needed to support development of S1PR1 antagonists. In rodents, intrathecal (i.th.) injection of SEW2871, a selective S1PR1 agonist, activates the nod-like receptor family, pyrin domain containing 3 inflammasome, increases interleukin-1β (IL-1β) and causes behavioral hypersensitivity. I.th. injection of a IL-1β receptor antagonist blocks SEW2871-induced hypersensitivity, suggesting that IL-1β contributes to S1PR1's actions. Interestingly, previous studies have suggested that IL-1β increases the expression/activity of adenosine kinase (ADK), a key regulator of adenosine signaling at its receptors (ARs). Increased ADK expression reduces adenosine signaling whereas inhibiting ADK restores the action of adenosine. Here, we show that SEW287-induced behavioral hypersensitivity is associated with increased expression of ADK in astrocytes of the dorsal horn of the spinal cord. Moreover, the ADK inhibitor, ABT702, blocks SEW2871-induced hypersensitivity. These findings link ADK activation to S1PR1. If SEW2871-induced pain is mediated by IL-1β, which in turn activates ADK and leads to mechano-allodynia, then blocking ADK should attenuate IL-1β effects. In support of this idea, recombinant rat (rrIL-1β)-induced allodynia was blocked by at least 90% with ABT702, functionally linking ADK to IL-1β. Moreover, the selective A3AR antagonist, MRS1523, prevents the ability of ABT702 to block SEW2871 and IL-1β-induced allodynia, implicating A3AR signaling in the beneficial effects exerted by ABT702. Our findings provide novel mechanistic insight into how S1PR1 signaling in the spinal cord produces hypersensitivity through IL1-β and ADK activation.
Collapse
Affiliation(s)
- Filomena Lauro
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, 1402 South Grand Blvd., St. Louis, MO, 63104, USA
- Henry and Amelia Nasrallah Center for Neuroscience, Saint Louis University School of Medicine, 1402 South Grand Blvd., St. Louis, MO, 63104, USA
| | - Luigino Antonio Giancotti
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, 1402 South Grand Blvd., St. Louis, MO, 63104, USA
- Henry and Amelia Nasrallah Center for Neuroscience, Saint Louis University School of Medicine, 1402 South Grand Blvd., St. Louis, MO, 63104, USA
| | - Grant Kolar
- Henry and Amelia Nasrallah Center for Neuroscience, Saint Louis University School of Medicine, 1402 South Grand Blvd., St. Louis, MO, 63104, USA
- Department of Pathology, Saint Louis University School of Medicine, 1402 South Grand Blvd., St. Louis, MO, 63104, USA
| | - Caron Mitsue Harada
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, 1402 South Grand Blvd., St. Louis, MO, 63104, USA
- Henry and Amelia Nasrallah Center for Neuroscience, Saint Louis University School of Medicine, 1402 South Grand Blvd., St. Louis, MO, 63104, USA
| | - Taylor A Harmon
- Department of Chemistry, University of Florida, Gainesville, FL, 32610, USA
| | - Timothy J Garrett
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, FL, 32610, USA
| | - Daniela Salvemini
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, 1402 South Grand Blvd., St. Louis, MO, 63104, USA.
- Henry and Amelia Nasrallah Center for Neuroscience, Saint Louis University School of Medicine, 1402 South Grand Blvd., St. Louis, MO, 63104, USA.
| |
Collapse
|
7
|
Molecular, Pathological, Clinical, and Therapeutic Aspects of Perihematomal Edema in Different Stages of Intracerebral Hemorrhage. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:3948921. [PMID: 36164392 PMCID: PMC9509250 DOI: 10.1155/2022/3948921] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/12/2022] [Revised: 08/17/2022] [Accepted: 09/03/2022] [Indexed: 02/07/2023]
Abstract
Acute intracerebral hemorrhage (ICH) is a devastating type of stroke worldwide. Neuronal destruction involved in the brain damage process caused by ICH includes a primary injury formed by the mass effect of the hematoma and a secondary injury induced by the degradation products of a blood clot. Additionally, factors in the coagulation cascade and complement activation process also contribute to secondary brain injury by promoting the disruption of the blood-brain barrier and neuronal cell degeneration by enhancing the inflammatory response, oxidative stress, etc. Although treatment options for direct damage are limited, various strategies have been proposed to treat secondary injury post-ICH. Perihematomal edema (PHE) is a potential surrogate marker for secondary injury and may contribute to poor outcomes after ICH. Therefore, it is essential to investigate the underlying pathological mechanism, evolution, and potential therapeutic strategies to treat PHE. Here, we review the pathophysiology and imaging characteristics of PHE at different stages after acute ICH. As illustrated in preclinical and clinical studies, we discussed the merits and limitations of varying PHE quantification protocols, including absolute PHE volume, relative PHE volume, and extension distance calculated with images and other techniques. Importantly, this review summarizes the factors that affect PHE by focusing on traditional variables, the cerebral venous drainage system, and the brain lymphatic drainage system. Finally, to facilitate translational research, we analyze why the relationship between PHE and the functional outcome of ICH is currently controversial. We also emphasize promising therapeutic approaches that modulate multiple targets to alleviate PHE and promote neurologic recovery after acute ICH.
Collapse
|
8
|
Sphingosine-1-Phosphate (S1P) and S1P Signaling Pathway Modulators, from Current Insights to Future Perspectives. Cells 2022; 11:cells11132058. [PMID: 35805142 PMCID: PMC9265592 DOI: 10.3390/cells11132058] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 06/23/2022] [Indexed: 01/27/2023] Open
Abstract
Sphingosine-1-phosphate (S1P) and S1P receptors (S1PR) are bioactive lipid molecules that are ubiquitously expressed in the human body and play an important role in the immune system. S1P-S1PR signaling has been well characterized in immune trafficking and activation in both innate and adaptive immune systems. Despite this knowledge, the full scope in the pathogenesis of autoimmune disorders is not well characterized yet. From the discovery of fingolimod, the first S1P modulator, until siponimod, the new molecule recently approved for the treatment of secondary progressive multiple sclerosis (SPMS), there has been a great advance in understanding the S1P functions and their involvement in immune diseases, including multiple sclerosis (MS). Modulation on S1P is an interesting target for the treatment of various autoimmune disorders. Improved understanding of the mechanism of action of fingolimod has allowed the development of the more selective second-generation S1PR modulators. Subtype 1 of the S1PR (S1PR1) is expressed on the cell surface of lymphocytes, which are known to play a major role in MS pathogenesis. The understanding of S1PR1’s role facilitated the development of pharmacological strategies directed to this target, and theoretically reduced the safety concerns derived from the use of fingolimod. A great advance in the MS treatment was achieved in March 2019 when the Food and Drug Association (FDA) approved Siponimod, for both active secondary progressive MS and relapsing–remitting MS. Siponimod became the first oral disease modifying therapy (DMT) specifically approved for active forms of secondary progressive MS. Additionally, for the treatment of relapsing forms of MS, ozanimod was approved by FDA in March 2020. Currently, there are ongoing trials focused on other new-generation S1PR1 modulators. This review approaches the fundamental aspects of the sphingosine phosphate modulators and their main similarities and differences.
Collapse
|
9
|
Dietrich M, Hecker C, Martin E, Langui D, Gliem M, Stankoff B, Lubetzki C, Gruchot J, Göttle P, Issberner A, Nasiri M, Ramseier P, Beerli C, Tisserand S, Beckmann N, Shimshek D, Petzsch P, Akbar D, Levkau B, Stark H, Köhrer K, Hartung HP, Küry P, Meuth SG, Bigaud M, Zalc B, Albrecht P. Increased Remyelination and Proregenerative Microglia Under Siponimod Therapy in Mechanistic Models. NEUROLOGY(R) NEUROIMMUNOLOGY & NEUROINFLAMMATION 2022; 9:9/3/e1161. [PMID: 35354603 PMCID: PMC8969301 DOI: 10.1212/nxi.0000000000001161] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Accepted: 01/27/2022] [Indexed: 12/11/2022]
Abstract
Background and Objectives Siponimod is an oral, selective sphingosine-1-phosphate receptor-1/5 modulator approved for treatment of multiple sclerosis. Methods Mouse MRI was used to investigate remyelination in the cuprizone model. We then used a conditional demyelination Xenopus laevis model to assess the dose-response of siponimod on remyelination. In experimental autoimmune encephalomyelitis–optic neuritis (EAEON) in C57Bl/6J mice, we monitored the retinal thickness and the visual acuity using optical coherence tomography and optomotor response. Optic nerve inflammatory infiltrates, demyelination, and microglial and oligodendroglial differentiation were assessed by immunohistochemistry, quantitative real-time PCR, and bulk RNA sequencing. Results An increased remyelination was observed in the cuprizone model. Siponimod treatment of demyelinated tadpoles improved remyelination in comparison to control in a bell-shaped dose-response curve. Siponimod in the EAEON model attenuated the clinical score, reduced the retinal degeneration, and improved the visual function after prophylactic and therapeutic treatment, also in a bell-shaped manner. Inflammatory infiltrates and demyelination of the optic nerve were reduced, the latter even after therapeutic treatment, which also shifted microglial differentiation to a promyelinating phenotype. Discussion These results confirm the immunomodulatory effects of siponimod and suggest additional regenerative and promyelinating effects, which follow the dynamics of a bell-shaped curve with high being less efficient than low concentrations.
Collapse
Affiliation(s)
- Michael Dietrich
- From the Department of Neurology (M.D., C.H., M.G., J.G., P.G., A.I., M.N., H.-P.H., P.K., S.G.M.), Heinrich Heine University Düsseldorf, Medical Faculty (P.A.), Düsseldorf, Germany; Sorbonne Université (E.M., D.L., B.S., C.L., D.A., B.Z.), Inserm, CNRS, Institut du Cerveau, Pitié-Salpêtrière Hospital; AP-HP (B.S.), Saint-Antoine Hospital; AP-HP (C.L.), Pitié-Salpêtrière Hospital, Paris, France; Novartis Institutes for BioMedical Research (P.R., C.B., S.T., N.B., D.S., M.B.), Basel, Switzerland; Biological and Medical Research Center (BMFZ) (P.P., K.K.), Heinrich Heine University Düsseldorf, Medical Faculty; Institute for Molecular Medicine III (B.L.), University Hospital Düsseldorf and Heinrich Heine University Düsseldorf; Institute of Pharmaceutical and Medicinal Chemistry (H.S.), Heinrich Heine University Düsseldorf, Duesseldorf, Germany; Brain and Mind Center (H.-P.H.), University of Sydney, NSW, Australia; and Medical University of Vienna (H.-P.H.), Vienna, Austria
| | - Christina Hecker
- From the Department of Neurology (M.D., C.H., M.G., J.G., P.G., A.I., M.N., H.-P.H., P.K., S.G.M.), Heinrich Heine University Düsseldorf, Medical Faculty (P.A.), Düsseldorf, Germany; Sorbonne Université (E.M., D.L., B.S., C.L., D.A., B.Z.), Inserm, CNRS, Institut du Cerveau, Pitié-Salpêtrière Hospital; AP-HP (B.S.), Saint-Antoine Hospital; AP-HP (C.L.), Pitié-Salpêtrière Hospital, Paris, France; Novartis Institutes for BioMedical Research (P.R., C.B., S.T., N.B., D.S., M.B.), Basel, Switzerland; Biological and Medical Research Center (BMFZ) (P.P., K.K.), Heinrich Heine University Düsseldorf, Medical Faculty; Institute for Molecular Medicine III (B.L.), University Hospital Düsseldorf and Heinrich Heine University Düsseldorf; Institute of Pharmaceutical and Medicinal Chemistry (H.S.), Heinrich Heine University Düsseldorf, Duesseldorf, Germany; Brain and Mind Center (H.-P.H.), University of Sydney, NSW, Australia; and Medical University of Vienna (H.-P.H.), Vienna, Austria
| | - Elodie Martin
- From the Department of Neurology (M.D., C.H., M.G., J.G., P.G., A.I., M.N., H.-P.H., P.K., S.G.M.), Heinrich Heine University Düsseldorf, Medical Faculty (P.A.), Düsseldorf, Germany; Sorbonne Université (E.M., D.L., B.S., C.L., D.A., B.Z.), Inserm, CNRS, Institut du Cerveau, Pitié-Salpêtrière Hospital; AP-HP (B.S.), Saint-Antoine Hospital; AP-HP (C.L.), Pitié-Salpêtrière Hospital, Paris, France; Novartis Institutes for BioMedical Research (P.R., C.B., S.T., N.B., D.S., M.B.), Basel, Switzerland; Biological and Medical Research Center (BMFZ) (P.P., K.K.), Heinrich Heine University Düsseldorf, Medical Faculty; Institute for Molecular Medicine III (B.L.), University Hospital Düsseldorf and Heinrich Heine University Düsseldorf; Institute of Pharmaceutical and Medicinal Chemistry (H.S.), Heinrich Heine University Düsseldorf, Duesseldorf, Germany; Brain and Mind Center (H.-P.H.), University of Sydney, NSW, Australia; and Medical University of Vienna (H.-P.H.), Vienna, Austria
| | - Dominique Langui
- From the Department of Neurology (M.D., C.H., M.G., J.G., P.G., A.I., M.N., H.-P.H., P.K., S.G.M.), Heinrich Heine University Düsseldorf, Medical Faculty (P.A.), Düsseldorf, Germany; Sorbonne Université (E.M., D.L., B.S., C.L., D.A., B.Z.), Inserm, CNRS, Institut du Cerveau, Pitié-Salpêtrière Hospital; AP-HP (B.S.), Saint-Antoine Hospital; AP-HP (C.L.), Pitié-Salpêtrière Hospital, Paris, France; Novartis Institutes for BioMedical Research (P.R., C.B., S.T., N.B., D.S., M.B.), Basel, Switzerland; Biological and Medical Research Center (BMFZ) (P.P., K.K.), Heinrich Heine University Düsseldorf, Medical Faculty; Institute for Molecular Medicine III (B.L.), University Hospital Düsseldorf and Heinrich Heine University Düsseldorf; Institute of Pharmaceutical and Medicinal Chemistry (H.S.), Heinrich Heine University Düsseldorf, Duesseldorf, Germany; Brain and Mind Center (H.-P.H.), University of Sydney, NSW, Australia; and Medical University of Vienna (H.-P.H.), Vienna, Austria
| | - Michael Gliem
- From the Department of Neurology (M.D., C.H., M.G., J.G., P.G., A.I., M.N., H.-P.H., P.K., S.G.M.), Heinrich Heine University Düsseldorf, Medical Faculty (P.A.), Düsseldorf, Germany; Sorbonne Université (E.M., D.L., B.S., C.L., D.A., B.Z.), Inserm, CNRS, Institut du Cerveau, Pitié-Salpêtrière Hospital; AP-HP (B.S.), Saint-Antoine Hospital; AP-HP (C.L.), Pitié-Salpêtrière Hospital, Paris, France; Novartis Institutes for BioMedical Research (P.R., C.B., S.T., N.B., D.S., M.B.), Basel, Switzerland; Biological and Medical Research Center (BMFZ) (P.P., K.K.), Heinrich Heine University Düsseldorf, Medical Faculty; Institute for Molecular Medicine III (B.L.), University Hospital Düsseldorf and Heinrich Heine University Düsseldorf; Institute of Pharmaceutical and Medicinal Chemistry (H.S.), Heinrich Heine University Düsseldorf, Duesseldorf, Germany; Brain and Mind Center (H.-P.H.), University of Sydney, NSW, Australia; and Medical University of Vienna (H.-P.H.), Vienna, Austria
| | - Bruno Stankoff
- From the Department of Neurology (M.D., C.H., M.G., J.G., P.G., A.I., M.N., H.-P.H., P.K., S.G.M.), Heinrich Heine University Düsseldorf, Medical Faculty (P.A.), Düsseldorf, Germany; Sorbonne Université (E.M., D.L., B.S., C.L., D.A., B.Z.), Inserm, CNRS, Institut du Cerveau, Pitié-Salpêtrière Hospital; AP-HP (B.S.), Saint-Antoine Hospital; AP-HP (C.L.), Pitié-Salpêtrière Hospital, Paris, France; Novartis Institutes for BioMedical Research (P.R., C.B., S.T., N.B., D.S., M.B.), Basel, Switzerland; Biological and Medical Research Center (BMFZ) (P.P., K.K.), Heinrich Heine University Düsseldorf, Medical Faculty; Institute for Molecular Medicine III (B.L.), University Hospital Düsseldorf and Heinrich Heine University Düsseldorf; Institute of Pharmaceutical and Medicinal Chemistry (H.S.), Heinrich Heine University Düsseldorf, Duesseldorf, Germany; Brain and Mind Center (H.-P.H.), University of Sydney, NSW, Australia; and Medical University of Vienna (H.-P.H.), Vienna, Austria
| | - Catherine Lubetzki
- From the Department of Neurology (M.D., C.H., M.G., J.G., P.G., A.I., M.N., H.-P.H., P.K., S.G.M.), Heinrich Heine University Düsseldorf, Medical Faculty (P.A.), Düsseldorf, Germany; Sorbonne Université (E.M., D.L., B.S., C.L., D.A., B.Z.), Inserm, CNRS, Institut du Cerveau, Pitié-Salpêtrière Hospital; AP-HP (B.S.), Saint-Antoine Hospital; AP-HP (C.L.), Pitié-Salpêtrière Hospital, Paris, France; Novartis Institutes for BioMedical Research (P.R., C.B., S.T., N.B., D.S., M.B.), Basel, Switzerland; Biological and Medical Research Center (BMFZ) (P.P., K.K.), Heinrich Heine University Düsseldorf, Medical Faculty; Institute for Molecular Medicine III (B.L.), University Hospital Düsseldorf and Heinrich Heine University Düsseldorf; Institute of Pharmaceutical and Medicinal Chemistry (H.S.), Heinrich Heine University Düsseldorf, Duesseldorf, Germany; Brain and Mind Center (H.-P.H.), University of Sydney, NSW, Australia; and Medical University of Vienna (H.-P.H.), Vienna, Austria
| | - Joel Gruchot
- From the Department of Neurology (M.D., C.H., M.G., J.G., P.G., A.I., M.N., H.-P.H., P.K., S.G.M.), Heinrich Heine University Düsseldorf, Medical Faculty (P.A.), Düsseldorf, Germany; Sorbonne Université (E.M., D.L., B.S., C.L., D.A., B.Z.), Inserm, CNRS, Institut du Cerveau, Pitié-Salpêtrière Hospital; AP-HP (B.S.), Saint-Antoine Hospital; AP-HP (C.L.), Pitié-Salpêtrière Hospital, Paris, France; Novartis Institutes for BioMedical Research (P.R., C.B., S.T., N.B., D.S., M.B.), Basel, Switzerland; Biological and Medical Research Center (BMFZ) (P.P., K.K.), Heinrich Heine University Düsseldorf, Medical Faculty; Institute for Molecular Medicine III (B.L.), University Hospital Düsseldorf and Heinrich Heine University Düsseldorf; Institute of Pharmaceutical and Medicinal Chemistry (H.S.), Heinrich Heine University Düsseldorf, Duesseldorf, Germany; Brain and Mind Center (H.-P.H.), University of Sydney, NSW, Australia; and Medical University of Vienna (H.-P.H.), Vienna, Austria
| | - Peter Göttle
- From the Department of Neurology (M.D., C.H., M.G., J.G., P.G., A.I., M.N., H.-P.H., P.K., S.G.M.), Heinrich Heine University Düsseldorf, Medical Faculty (P.A.), Düsseldorf, Germany; Sorbonne Université (E.M., D.L., B.S., C.L., D.A., B.Z.), Inserm, CNRS, Institut du Cerveau, Pitié-Salpêtrière Hospital; AP-HP (B.S.), Saint-Antoine Hospital; AP-HP (C.L.), Pitié-Salpêtrière Hospital, Paris, France; Novartis Institutes for BioMedical Research (P.R., C.B., S.T., N.B., D.S., M.B.), Basel, Switzerland; Biological and Medical Research Center (BMFZ) (P.P., K.K.), Heinrich Heine University Düsseldorf, Medical Faculty; Institute for Molecular Medicine III (B.L.), University Hospital Düsseldorf and Heinrich Heine University Düsseldorf; Institute of Pharmaceutical and Medicinal Chemistry (H.S.), Heinrich Heine University Düsseldorf, Duesseldorf, Germany; Brain and Mind Center (H.-P.H.), University of Sydney, NSW, Australia; and Medical University of Vienna (H.-P.H.), Vienna, Austria
| | - Andrea Issberner
- From the Department of Neurology (M.D., C.H., M.G., J.G., P.G., A.I., M.N., H.-P.H., P.K., S.G.M.), Heinrich Heine University Düsseldorf, Medical Faculty (P.A.), Düsseldorf, Germany; Sorbonne Université (E.M., D.L., B.S., C.L., D.A., B.Z.), Inserm, CNRS, Institut du Cerveau, Pitié-Salpêtrière Hospital; AP-HP (B.S.), Saint-Antoine Hospital; AP-HP (C.L.), Pitié-Salpêtrière Hospital, Paris, France; Novartis Institutes for BioMedical Research (P.R., C.B., S.T., N.B., D.S., M.B.), Basel, Switzerland; Biological and Medical Research Center (BMFZ) (P.P., K.K.), Heinrich Heine University Düsseldorf, Medical Faculty; Institute for Molecular Medicine III (B.L.), University Hospital Düsseldorf and Heinrich Heine University Düsseldorf; Institute of Pharmaceutical and Medicinal Chemistry (H.S.), Heinrich Heine University Düsseldorf, Duesseldorf, Germany; Brain and Mind Center (H.-P.H.), University of Sydney, NSW, Australia; and Medical University of Vienna (H.-P.H.), Vienna, Austria
| | - Milad Nasiri
- From the Department of Neurology (M.D., C.H., M.G., J.G., P.G., A.I., M.N., H.-P.H., P.K., S.G.M.), Heinrich Heine University Düsseldorf, Medical Faculty (P.A.), Düsseldorf, Germany; Sorbonne Université (E.M., D.L., B.S., C.L., D.A., B.Z.), Inserm, CNRS, Institut du Cerveau, Pitié-Salpêtrière Hospital; AP-HP (B.S.), Saint-Antoine Hospital; AP-HP (C.L.), Pitié-Salpêtrière Hospital, Paris, France; Novartis Institutes for BioMedical Research (P.R., C.B., S.T., N.B., D.S., M.B.), Basel, Switzerland; Biological and Medical Research Center (BMFZ) (P.P., K.K.), Heinrich Heine University Düsseldorf, Medical Faculty; Institute for Molecular Medicine III (B.L.), University Hospital Düsseldorf and Heinrich Heine University Düsseldorf; Institute of Pharmaceutical and Medicinal Chemistry (H.S.), Heinrich Heine University Düsseldorf, Duesseldorf, Germany; Brain and Mind Center (H.-P.H.), University of Sydney, NSW, Australia; and Medical University of Vienna (H.-P.H.), Vienna, Austria
| | - Pamela Ramseier
- From the Department of Neurology (M.D., C.H., M.G., J.G., P.G., A.I., M.N., H.-P.H., P.K., S.G.M.), Heinrich Heine University Düsseldorf, Medical Faculty (P.A.), Düsseldorf, Germany; Sorbonne Université (E.M., D.L., B.S., C.L., D.A., B.Z.), Inserm, CNRS, Institut du Cerveau, Pitié-Salpêtrière Hospital; AP-HP (B.S.), Saint-Antoine Hospital; AP-HP (C.L.), Pitié-Salpêtrière Hospital, Paris, France; Novartis Institutes for BioMedical Research (P.R., C.B., S.T., N.B., D.S., M.B.), Basel, Switzerland; Biological and Medical Research Center (BMFZ) (P.P., K.K.), Heinrich Heine University Düsseldorf, Medical Faculty; Institute for Molecular Medicine III (B.L.), University Hospital Düsseldorf and Heinrich Heine University Düsseldorf; Institute of Pharmaceutical and Medicinal Chemistry (H.S.), Heinrich Heine University Düsseldorf, Duesseldorf, Germany; Brain and Mind Center (H.-P.H.), University of Sydney, NSW, Australia; and Medical University of Vienna (H.-P.H.), Vienna, Austria
| | - Christian Beerli
- From the Department of Neurology (M.D., C.H., M.G., J.G., P.G., A.I., M.N., H.-P.H., P.K., S.G.M.), Heinrich Heine University Düsseldorf, Medical Faculty (P.A.), Düsseldorf, Germany; Sorbonne Université (E.M., D.L., B.S., C.L., D.A., B.Z.), Inserm, CNRS, Institut du Cerveau, Pitié-Salpêtrière Hospital; AP-HP (B.S.), Saint-Antoine Hospital; AP-HP (C.L.), Pitié-Salpêtrière Hospital, Paris, France; Novartis Institutes for BioMedical Research (P.R., C.B., S.T., N.B., D.S., M.B.), Basel, Switzerland; Biological and Medical Research Center (BMFZ) (P.P., K.K.), Heinrich Heine University Düsseldorf, Medical Faculty; Institute for Molecular Medicine III (B.L.), University Hospital Düsseldorf and Heinrich Heine University Düsseldorf; Institute of Pharmaceutical and Medicinal Chemistry (H.S.), Heinrich Heine University Düsseldorf, Duesseldorf, Germany; Brain and Mind Center (H.-P.H.), University of Sydney, NSW, Australia; and Medical University of Vienna (H.-P.H.), Vienna, Austria
| | - Sarah Tisserand
- From the Department of Neurology (M.D., C.H., M.G., J.G., P.G., A.I., M.N., H.-P.H., P.K., S.G.M.), Heinrich Heine University Düsseldorf, Medical Faculty (P.A.), Düsseldorf, Germany; Sorbonne Université (E.M., D.L., B.S., C.L., D.A., B.Z.), Inserm, CNRS, Institut du Cerveau, Pitié-Salpêtrière Hospital; AP-HP (B.S.), Saint-Antoine Hospital; AP-HP (C.L.), Pitié-Salpêtrière Hospital, Paris, France; Novartis Institutes for BioMedical Research (P.R., C.B., S.T., N.B., D.S., M.B.), Basel, Switzerland; Biological and Medical Research Center (BMFZ) (P.P., K.K.), Heinrich Heine University Düsseldorf, Medical Faculty; Institute for Molecular Medicine III (B.L.), University Hospital Düsseldorf and Heinrich Heine University Düsseldorf; Institute of Pharmaceutical and Medicinal Chemistry (H.S.), Heinrich Heine University Düsseldorf, Duesseldorf, Germany; Brain and Mind Center (H.-P.H.), University of Sydney, NSW, Australia; and Medical University of Vienna (H.-P.H.), Vienna, Austria
| | - Nicolau Beckmann
- From the Department of Neurology (M.D., C.H., M.G., J.G., P.G., A.I., M.N., H.-P.H., P.K., S.G.M.), Heinrich Heine University Düsseldorf, Medical Faculty (P.A.), Düsseldorf, Germany; Sorbonne Université (E.M., D.L., B.S., C.L., D.A., B.Z.), Inserm, CNRS, Institut du Cerveau, Pitié-Salpêtrière Hospital; AP-HP (B.S.), Saint-Antoine Hospital; AP-HP (C.L.), Pitié-Salpêtrière Hospital, Paris, France; Novartis Institutes for BioMedical Research (P.R., C.B., S.T., N.B., D.S., M.B.), Basel, Switzerland; Biological and Medical Research Center (BMFZ) (P.P., K.K.), Heinrich Heine University Düsseldorf, Medical Faculty; Institute for Molecular Medicine III (B.L.), University Hospital Düsseldorf and Heinrich Heine University Düsseldorf; Institute of Pharmaceutical and Medicinal Chemistry (H.S.), Heinrich Heine University Düsseldorf, Duesseldorf, Germany; Brain and Mind Center (H.-P.H.), University of Sydney, NSW, Australia; and Medical University of Vienna (H.-P.H.), Vienna, Austria
| | - Derya Shimshek
- From the Department of Neurology (M.D., C.H., M.G., J.G., P.G., A.I., M.N., H.-P.H., P.K., S.G.M.), Heinrich Heine University Düsseldorf, Medical Faculty (P.A.), Düsseldorf, Germany; Sorbonne Université (E.M., D.L., B.S., C.L., D.A., B.Z.), Inserm, CNRS, Institut du Cerveau, Pitié-Salpêtrière Hospital; AP-HP (B.S.), Saint-Antoine Hospital; AP-HP (C.L.), Pitié-Salpêtrière Hospital, Paris, France; Novartis Institutes for BioMedical Research (P.R., C.B., S.T., N.B., D.S., M.B.), Basel, Switzerland; Biological and Medical Research Center (BMFZ) (P.P., K.K.), Heinrich Heine University Düsseldorf, Medical Faculty; Institute for Molecular Medicine III (B.L.), University Hospital Düsseldorf and Heinrich Heine University Düsseldorf; Institute of Pharmaceutical and Medicinal Chemistry (H.S.), Heinrich Heine University Düsseldorf, Duesseldorf, Germany; Brain and Mind Center (H.-P.H.), University of Sydney, NSW, Australia; and Medical University of Vienna (H.-P.H.), Vienna, Austria
| | - Patrick Petzsch
- From the Department of Neurology (M.D., C.H., M.G., J.G., P.G., A.I., M.N., H.-P.H., P.K., S.G.M.), Heinrich Heine University Düsseldorf, Medical Faculty (P.A.), Düsseldorf, Germany; Sorbonne Université (E.M., D.L., B.S., C.L., D.A., B.Z.), Inserm, CNRS, Institut du Cerveau, Pitié-Salpêtrière Hospital; AP-HP (B.S.), Saint-Antoine Hospital; AP-HP (C.L.), Pitié-Salpêtrière Hospital, Paris, France; Novartis Institutes for BioMedical Research (P.R., C.B., S.T., N.B., D.S., M.B.), Basel, Switzerland; Biological and Medical Research Center (BMFZ) (P.P., K.K.), Heinrich Heine University Düsseldorf, Medical Faculty; Institute for Molecular Medicine III (B.L.), University Hospital Düsseldorf and Heinrich Heine University Düsseldorf; Institute of Pharmaceutical and Medicinal Chemistry (H.S.), Heinrich Heine University Düsseldorf, Duesseldorf, Germany; Brain and Mind Center (H.-P.H.), University of Sydney, NSW, Australia; and Medical University of Vienna (H.-P.H.), Vienna, Austria
| | - David Akbar
- From the Department of Neurology (M.D., C.H., M.G., J.G., P.G., A.I., M.N., H.-P.H., P.K., S.G.M.), Heinrich Heine University Düsseldorf, Medical Faculty (P.A.), Düsseldorf, Germany; Sorbonne Université (E.M., D.L., B.S., C.L., D.A., B.Z.), Inserm, CNRS, Institut du Cerveau, Pitié-Salpêtrière Hospital; AP-HP (B.S.), Saint-Antoine Hospital; AP-HP (C.L.), Pitié-Salpêtrière Hospital, Paris, France; Novartis Institutes for BioMedical Research (P.R., C.B., S.T., N.B., D.S., M.B.), Basel, Switzerland; Biological and Medical Research Center (BMFZ) (P.P., K.K.), Heinrich Heine University Düsseldorf, Medical Faculty; Institute for Molecular Medicine III (B.L.), University Hospital Düsseldorf and Heinrich Heine University Düsseldorf; Institute of Pharmaceutical and Medicinal Chemistry (H.S.), Heinrich Heine University Düsseldorf, Duesseldorf, Germany; Brain and Mind Center (H.-P.H.), University of Sydney, NSW, Australia; and Medical University of Vienna (H.-P.H.), Vienna, Austria
| | - Bodo Levkau
- From the Department of Neurology (M.D., C.H., M.G., J.G., P.G., A.I., M.N., H.-P.H., P.K., S.G.M.), Heinrich Heine University Düsseldorf, Medical Faculty (P.A.), Düsseldorf, Germany; Sorbonne Université (E.M., D.L., B.S., C.L., D.A., B.Z.), Inserm, CNRS, Institut du Cerveau, Pitié-Salpêtrière Hospital; AP-HP (B.S.), Saint-Antoine Hospital; AP-HP (C.L.), Pitié-Salpêtrière Hospital, Paris, France; Novartis Institutes for BioMedical Research (P.R., C.B., S.T., N.B., D.S., M.B.), Basel, Switzerland; Biological and Medical Research Center (BMFZ) (P.P., K.K.), Heinrich Heine University Düsseldorf, Medical Faculty; Institute for Molecular Medicine III (B.L.), University Hospital Düsseldorf and Heinrich Heine University Düsseldorf; Institute of Pharmaceutical and Medicinal Chemistry (H.S.), Heinrich Heine University Düsseldorf, Duesseldorf, Germany; Brain and Mind Center (H.-P.H.), University of Sydney, NSW, Australia; and Medical University of Vienna (H.-P.H.), Vienna, Austria
| | - Holger Stark
- From the Department of Neurology (M.D., C.H., M.G., J.G., P.G., A.I., M.N., H.-P.H., P.K., S.G.M.), Heinrich Heine University Düsseldorf, Medical Faculty (P.A.), Düsseldorf, Germany; Sorbonne Université (E.M., D.L., B.S., C.L., D.A., B.Z.), Inserm, CNRS, Institut du Cerveau, Pitié-Salpêtrière Hospital; AP-HP (B.S.), Saint-Antoine Hospital; AP-HP (C.L.), Pitié-Salpêtrière Hospital, Paris, France; Novartis Institutes for BioMedical Research (P.R., C.B., S.T., N.B., D.S., M.B.), Basel, Switzerland; Biological and Medical Research Center (BMFZ) (P.P., K.K.), Heinrich Heine University Düsseldorf, Medical Faculty; Institute for Molecular Medicine III (B.L.), University Hospital Düsseldorf and Heinrich Heine University Düsseldorf; Institute of Pharmaceutical and Medicinal Chemistry (H.S.), Heinrich Heine University Düsseldorf, Duesseldorf, Germany; Brain and Mind Center (H.-P.H.), University of Sydney, NSW, Australia; and Medical University of Vienna (H.-P.H.), Vienna, Austria
| | - Karl Köhrer
- From the Department of Neurology (M.D., C.H., M.G., J.G., P.G., A.I., M.N., H.-P.H., P.K., S.G.M.), Heinrich Heine University Düsseldorf, Medical Faculty (P.A.), Düsseldorf, Germany; Sorbonne Université (E.M., D.L., B.S., C.L., D.A., B.Z.), Inserm, CNRS, Institut du Cerveau, Pitié-Salpêtrière Hospital; AP-HP (B.S.), Saint-Antoine Hospital; AP-HP (C.L.), Pitié-Salpêtrière Hospital, Paris, France; Novartis Institutes for BioMedical Research (P.R., C.B., S.T., N.B., D.S., M.B.), Basel, Switzerland; Biological and Medical Research Center (BMFZ) (P.P., K.K.), Heinrich Heine University Düsseldorf, Medical Faculty; Institute for Molecular Medicine III (B.L.), University Hospital Düsseldorf and Heinrich Heine University Düsseldorf; Institute of Pharmaceutical and Medicinal Chemistry (H.S.), Heinrich Heine University Düsseldorf, Duesseldorf, Germany; Brain and Mind Center (H.-P.H.), University of Sydney, NSW, Australia; and Medical University of Vienna (H.-P.H.), Vienna, Austria
| | - Hans-Peter Hartung
- From the Department of Neurology (M.D., C.H., M.G., J.G., P.G., A.I., M.N., H.-P.H., P.K., S.G.M.), Heinrich Heine University Düsseldorf, Medical Faculty (P.A.), Düsseldorf, Germany; Sorbonne Université (E.M., D.L., B.S., C.L., D.A., B.Z.), Inserm, CNRS, Institut du Cerveau, Pitié-Salpêtrière Hospital; AP-HP (B.S.), Saint-Antoine Hospital; AP-HP (C.L.), Pitié-Salpêtrière Hospital, Paris, France; Novartis Institutes for BioMedical Research (P.R., C.B., S.T., N.B., D.S., M.B.), Basel, Switzerland; Biological and Medical Research Center (BMFZ) (P.P., K.K.), Heinrich Heine University Düsseldorf, Medical Faculty; Institute for Molecular Medicine III (B.L.), University Hospital Düsseldorf and Heinrich Heine University Düsseldorf; Institute of Pharmaceutical and Medicinal Chemistry (H.S.), Heinrich Heine University Düsseldorf, Duesseldorf, Germany; Brain and Mind Center (H.-P.H.), University of Sydney, NSW, Australia; and Medical University of Vienna (H.-P.H.), Vienna, Austria
| | - Patrick Küry
- From the Department of Neurology (M.D., C.H., M.G., J.G., P.G., A.I., M.N., H.-P.H., P.K., S.G.M.), Heinrich Heine University Düsseldorf, Medical Faculty (P.A.), Düsseldorf, Germany; Sorbonne Université (E.M., D.L., B.S., C.L., D.A., B.Z.), Inserm, CNRS, Institut du Cerveau, Pitié-Salpêtrière Hospital; AP-HP (B.S.), Saint-Antoine Hospital; AP-HP (C.L.), Pitié-Salpêtrière Hospital, Paris, France; Novartis Institutes for BioMedical Research (P.R., C.B., S.T., N.B., D.S., M.B.), Basel, Switzerland; Biological and Medical Research Center (BMFZ) (P.P., K.K.), Heinrich Heine University Düsseldorf, Medical Faculty; Institute for Molecular Medicine III (B.L.), University Hospital Düsseldorf and Heinrich Heine University Düsseldorf; Institute of Pharmaceutical and Medicinal Chemistry (H.S.), Heinrich Heine University Düsseldorf, Duesseldorf, Germany; Brain and Mind Center (H.-P.H.), University of Sydney, NSW, Australia; and Medical University of Vienna (H.-P.H.), Vienna, Austria
| | - Sven Günther Meuth
- From the Department of Neurology (M.D., C.H., M.G., J.G., P.G., A.I., M.N., H.-P.H., P.K., S.G.M.), Heinrich Heine University Düsseldorf, Medical Faculty (P.A.), Düsseldorf, Germany; Sorbonne Université (E.M., D.L., B.S., C.L., D.A., B.Z.), Inserm, CNRS, Institut du Cerveau, Pitié-Salpêtrière Hospital; AP-HP (B.S.), Saint-Antoine Hospital; AP-HP (C.L.), Pitié-Salpêtrière Hospital, Paris, France; Novartis Institutes for BioMedical Research (P.R., C.B., S.T., N.B., D.S., M.B.), Basel, Switzerland; Biological and Medical Research Center (BMFZ) (P.P., K.K.), Heinrich Heine University Düsseldorf, Medical Faculty; Institute for Molecular Medicine III (B.L.), University Hospital Düsseldorf and Heinrich Heine University Düsseldorf; Institute of Pharmaceutical and Medicinal Chemistry (H.S.), Heinrich Heine University Düsseldorf, Duesseldorf, Germany; Brain and Mind Center (H.-P.H.), University of Sydney, NSW, Australia; and Medical University of Vienna (H.-P.H.), Vienna, Austria
| | - Marc Bigaud
- From the Department of Neurology (M.D., C.H., M.G., J.G., P.G., A.I., M.N., H.-P.H., P.K., S.G.M.), Heinrich Heine University Düsseldorf, Medical Faculty (P.A.), Düsseldorf, Germany; Sorbonne Université (E.M., D.L., B.S., C.L., D.A., B.Z.), Inserm, CNRS, Institut du Cerveau, Pitié-Salpêtrière Hospital; AP-HP (B.S.), Saint-Antoine Hospital; AP-HP (C.L.), Pitié-Salpêtrière Hospital, Paris, France; Novartis Institutes for BioMedical Research (P.R., C.B., S.T., N.B., D.S., M.B.), Basel, Switzerland; Biological and Medical Research Center (BMFZ) (P.P., K.K.), Heinrich Heine University Düsseldorf, Medical Faculty; Institute for Molecular Medicine III (B.L.), University Hospital Düsseldorf and Heinrich Heine University Düsseldorf; Institute of Pharmaceutical and Medicinal Chemistry (H.S.), Heinrich Heine University Düsseldorf, Duesseldorf, Germany; Brain and Mind Center (H.-P.H.), University of Sydney, NSW, Australia; and Medical University of Vienna (H.-P.H.), Vienna, Austria
| | - Bernard Zalc
- From the Department of Neurology (M.D., C.H., M.G., J.G., P.G., A.I., M.N., H.-P.H., P.K., S.G.M.), Heinrich Heine University Düsseldorf, Medical Faculty (P.A.), Düsseldorf, Germany; Sorbonne Université (E.M., D.L., B.S., C.L., D.A., B.Z.), Inserm, CNRS, Institut du Cerveau, Pitié-Salpêtrière Hospital; AP-HP (B.S.), Saint-Antoine Hospital; AP-HP (C.L.), Pitié-Salpêtrière Hospital, Paris, France; Novartis Institutes for BioMedical Research (P.R., C.B., S.T., N.B., D.S., M.B.), Basel, Switzerland; Biological and Medical Research Center (BMFZ) (P.P., K.K.), Heinrich Heine University Düsseldorf, Medical Faculty; Institute for Molecular Medicine III (B.L.), University Hospital Düsseldorf and Heinrich Heine University Düsseldorf; Institute of Pharmaceutical and Medicinal Chemistry (H.S.), Heinrich Heine University Düsseldorf, Duesseldorf, Germany; Brain and Mind Center (H.-P.H.), University of Sydney, NSW, Australia; and Medical University of Vienna (H.-P.H.), Vienna, Austria
| | - Philipp Albrecht
- From the Department of Neurology (M.D., C.H., M.G., J.G., P.G., A.I., M.N., H.-P.H., P.K., S.G.M.), Heinrich Heine University Düsseldorf, Medical Faculty (P.A.), Düsseldorf, Germany; Sorbonne Université (E.M., D.L., B.S., C.L., D.A., B.Z.), Inserm, CNRS, Institut du Cerveau, Pitié-Salpêtrière Hospital; AP-HP (B.S.), Saint-Antoine Hospital; AP-HP (C.L.), Pitié-Salpêtrière Hospital, Paris, France; Novartis Institutes for BioMedical Research (P.R., C.B., S.T., N.B., D.S., M.B.), Basel, Switzerland; Biological and Medical Research Center (BMFZ) (P.P., K.K.), Heinrich Heine University Düsseldorf, Medical Faculty; Institute for Molecular Medicine III (B.L.), University Hospital Düsseldorf and Heinrich Heine University Düsseldorf; Institute of Pharmaceutical and Medicinal Chemistry (H.S.), Heinrich Heine University Düsseldorf, Duesseldorf, Germany; Brain and Mind Center (H.-P.H.), University of Sydney, NSW, Australia; and Medical University of Vienna (H.-P.H.), Vienna, Austria
| |
Collapse
|
10
|
Wang J, Goren I, Yang B, Lin S, Li J, Elias M, Fiocchi C, Rieder F. Review article: the sphingosine 1 phosphate/sphingosine 1 phosphate receptor axis - a unique therapeutic target in inflammatory bowel disease. Aliment Pharmacol Ther 2022; 55:277-291. [PMID: 34932238 PMCID: PMC8766911 DOI: 10.1111/apt.16741] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 10/03/2021] [Accepted: 12/09/2021] [Indexed: 02/03/2023]
Abstract
BACKGROUND Ozanimod, a high selective sphingosine 1 phosphate (S1P) receptor (S1PR) 1/5 modulator was approved by the Food and Drug Administration for the treatment of adult patients with moderately to severely active ulcerative colitis. Additional S1PR modulators are being tested in clinical development programmes for both ulcerative colitis and Crohn's disease. AIM To provide an overview of advances in understanding S1PRs biology and summarise preclinical and clinical investigations of S1P receptor modulators in chronic inflammatory disease with special emphasis on inflammatory bowel diseases (IBD). METHODS We performed a narrative review using PubMed and ClinicalTrials.gov. RESULTS Through S1PRs, S1P regulates multiple cellular processes, including proliferation, migration, survival, and vascular barrier integrity. The S1PRs function of regulating lymphocyte trafficking is well known, but new functions of S1PRs expand our knowledge of S1PRs biology. Several S1PR modulators are in clinical development for both ulcerative colitis and Crohn's disease and have shown promise in phase II and III studies with ozanimod now being approved for ulcerative colitis. CONCLUSIONS S1P receptor modulators constitute a novel, promising, safe, and convenient strategy for the treatment of IBD.
Collapse
Affiliation(s)
- Jie Wang
- Henan Key Laboratory of Immunology and Targeted Drug, Xinxiang Medical University, Xinxiang 453003, Henan Province, China,Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, Ohio, USA
| | - Idan Goren
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, Ohio, USA,Division of Gastroenterology, Rabin Medical Center, Petah Tikva, Israel, Affiliated with Sackler Faculty of Medicine, Tel Aviv University, Israel
| | - Bo Yang
- Henan Key Laboratory of Immunology and Targeted Drug, Xinxiang Medical University, Xinxiang 453003, Henan Province, China
| | - Sinan Lin
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, Ohio, USA,Department of Gastroenterology and Hepatology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Jiannan Li
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, Ohio, USA
| | - Michael Elias
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, Ohio, USA
| | - Claudio Fiocchi
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, Ohio, USA,Department of Gastroenterology, Hepatology and Nutrition, Digestive Diseases and Surgery Institute; Cleveland Clinic Foundation, Cleveland, USA
| | - Florian Rieder
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, Ohio, USA,Department of Gastroenterology, Hepatology and Nutrition, Digestive Diseases and Surgery Institute; Cleveland Clinic Foundation, Cleveland, USA
| |
Collapse
|
11
|
Yuan M, Wu H. Astrocytes in the Traumatic Brain Injury: the Good and the Bad. Exp Neurol 2021; 348:113943. [PMID: 34863998 DOI: 10.1016/j.expneurol.2021.113943] [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: 08/17/2021] [Revised: 11/08/2021] [Accepted: 11/29/2021] [Indexed: 12/21/2022]
Abstract
Astrocytes control many processes of the nervous system in health and disease, and respond to injury quickly. Astrocytes produce neuroprotective factors in the injured brain to clear cellular debris and to orchestrate neurorestorative processes that are beneficial for neurological recovery after traumatic brain injury (TBI). However, astrocytes also become dysregulated and produce cytotoxic mediators that hinder CNS repair by induction of neuronal dysfunction and cell death. Hence, we discuss the potential role of astrocytes in neuropathological processes such as neuroinflammation, neurogenesis, synaptogenesis and blood-brain barrier repair after TBI. Thus, an improved understanding of the dual role of astrocytes may advance our knowledge of post-brain injury recovery, and provide opportunities for the development of novel therapeutic strategies for TBI.
Collapse
Affiliation(s)
- Mengqi Yuan
- Institute of Neuroscience, Hengyang Medical College, University of South China, Hengyang, 421001, Hunan, China
| | - Haitao Wu
- Beijing Institute of Basic Medical Sciences, 100850 Beijing, China; Key Laboratory of Neuroregeneration, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, 226019, Jiangsu, China; Chinese Institute for Brain Research (CIBR), 102206 Beijing, China.
| |
Collapse
|
12
|
Labus JS, Mayer EA, Tillisch K, Aagaard KM, Stains J, Broniowska K, Van Remortel C, Tun G, Rapkin A. Dysregulation in Sphingolipid Signaling Pathways is Associated With Symptoms and Functional Connectivity of Pain Processing Brain Regions in Provoked Vestibulodynia. THE JOURNAL OF PAIN 2021; 22:1586-1605. [PMID: 34029688 PMCID: PMC10460622 DOI: 10.1016/j.jpain.2021.04.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 03/27/2021] [Accepted: 04/29/2021] [Indexed: 10/21/2022]
Abstract
Provoked vestibulodynia (PVD) is a chronic pain disorder characterized by local hypersensitivity and severe pain with pressure localized to the vulvar vestibule. Despite decades of study, the lack of identified biomarkers has slowed the development of effective therapies. The primary aim of this study was to use metabolomics to identify novel biochemical mechanisms in vagina and blood underlying brain biomarkers and symptoms in PVD, thereby closing this knowledge gap. Using a cross-sectional case-control observational study design, untargeted and unbiased metabolomic profiling of vaginal fluid and plasma was performed in women with PVD compared to healthy controls. In women with PVD, we also obtained assessments of vulvar pain, vestibular and vaginal muscle tenderness, and 24-hour symptom intensity alongside resting-state brain functional connectivity of brain regions involved in pain processing and modulation. Compared to healthy controls, women with PVD demonstrated differences primarily in vaginal (but not plasma) concentrations of metabolites of the sphingolipid signaling pathways, suggesting localized effects in vagina and vulvar vestibule rather than systemic effects. Our findings reveal that dysregulation of sphingolipid metabolism in PVD is associated with increased vulvar pain and muscle tenderness, sexual dysfunction, and decreased functional connectivity strength in pain processing/modulatory brain regions. This data collectively suggests that alterations in sphingolipid signaling pathways are likely an important molecular biomarker in PVD that could lead to new targets for therapeutic intervention. PERSPECTIVE: This manuscript presents the results of a robust, unbiased molecular assessment of plasma and vaginal fluid samples in women with provoked vestibulodynia compared to healthy controls. The findings suggest that alterations in sphingolipid signaling pathways are associated with symptoms and brain biomarkers and may be an important molecular marker that could provide new targets for therapeutic intervention.
Collapse
Affiliation(s)
- Jennifer S Labus
- G. Oppenheimer Center for Neurobiology of Stress and Resilience, Vatche and Tamar Manoukian Division of Digestive Diseases, David Geffen School of Medicine at the University of California, Los Angeles, California; Brain Research Institute UCLA, Gonda (Goldschmied) Neuroscience and Genetics Research Center, Los Angeles, California.
| | - Emeran A Mayer
- G. Oppenheimer Center for Neurobiology of Stress and Resilience, Vatche and Tamar Manoukian Division of Digestive Diseases, David Geffen School of Medicine at the University of California, Los Angeles, California
| | - Kirsten Tillisch
- G. Oppenheimer Center for Neurobiology of Stress and Resilience, Vatche and Tamar Manoukian Division of Digestive Diseases, David Geffen School of Medicine at the University of California, Los Angeles, California
| | - Kjersti M Aagaard
- Division of Maternal-Fetal Medicine, Departments of Obstetrics and Gynecology, Baylor College of Medicine and Texas Children's Hospital, Houston, Texas; Department of Molecular and Human Genetics, Bioinformatics Research Laboratory, Baylor College of Medicine, Houston, Texas; Department of Molecular and Cell Biology, Baylor College of Medicine, Houston, Texas
| | - Jean Stains
- G. Oppenheimer Center for Neurobiology of Stress and Resilience, Vatche and Tamar Manoukian Division of Digestive Diseases, David Geffen School of Medicine at the University of California, Los Angeles, California
| | | | - Charlotte Van Remortel
- G. Oppenheimer Center for Neurobiology of Stress and Resilience, Vatche and Tamar Manoukian Division of Digestive Diseases, David Geffen School of Medicine at the University of California, Los Angeles, California
| | - Guistinna Tun
- G. Oppenheimer Center for Neurobiology of Stress and Resilience, Vatche and Tamar Manoukian Division of Digestive Diseases, David Geffen School of Medicine at the University of California, Los Angeles, California
| | - Andrea Rapkin
- G. Oppenheimer Center for Neurobiology of Stress and Resilience, Vatche and Tamar Manoukian Division of Digestive Diseases, David Geffen School of Medicine at the University of California, Los Angeles, California; Department of Obstetrics and Gynecology, David Geffen School of Medicine at the University of California, Los Angeles, California
| |
Collapse
|
13
|
Sphingolipid metabolism governs Purkinje cell patterned degeneration in Atxn1[82Q]/+ mice. Proc Natl Acad Sci U S A 2021; 118:2016969118. [PMID: 34479994 PMCID: PMC8433568 DOI: 10.1073/pnas.2016969118] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Accepted: 07/19/2021] [Indexed: 01/02/2023] Open
Abstract
Neuronal subtypes are differentially affected by neuropathologies. For example, Purkinje cells, the principal neurons of the cerebellum, can be divided in subpopulations based on their sensitivity to pathological insult. However, the molecular mechanisms explaining why, among seemingly identical neurons, some will degenerate while others survive remain unknown. Here, we analyzed, in a disease model of cerebellar neurodegeneration, the metabolism of sphingolipids, complex lipids involved in cell apoptosis, and found that specific sphingolipids accumulate in the cerebellar region primarily affected by neurodegeneration. Preventing this accumulation by disrupting sphingolipid metabolism via genetic mutation caused a neuroprotective effect on subpopulations of Purkinje cells. Thus, our data indicate that sphingolipid metabolism is involved in the predisposition of neuronal subtypes to neurodegeneration. Patterned degeneration of Purkinje cells (PCs) can be observed in a wide range of neuropathologies, but mechanisms behind nonrandom cerebellar neurodegeneration remain unclear. Sphingolipid metabolism dyshomeostasis typically leads to PC neurodegeneration; hence, we questioned whether local sphingolipid balance underlies regional sensitivity to pathological insults. Here, we investigated the regional compartmentalization of sphingolipids and their related enzymes in the cerebellar cortex in healthy and pathological conditions. Analysis in wild-type animals revealed higher sphingosine kinase 1 (Sphk1) levels in the flocculonodular cerebellum, while sphingosine-1-phosphate (S1P) levels were higher in the anterior cerebellum. Next, we investigated a model for spinocerebellar ataxia type 1 (SCA1) driven by the transgenic expression of the expanded Ataxin 1 protein with 82 glutamine (82Q), exhibiting severe PC degeneration in the anterior cerebellum while the flocculonodular region is preserved. In Atxn1[82Q]/+ mice, we found that levels of Sphk1 and Sphk2 were region-specific decreased and S1P levels increased, particularly in the anterior cerebellum. To determine if there is a causal link between sphingolipid levels and neurodegeneration, we deleted the Sphk1 gene in Atxn1[82Q]/+ mice. Analysis of Atxn1[82Q]/+; Sphk1−/− mice confirmed a neuroprotective effect, rescuing a subset of PCs in the anterior cerebellum, in domains reminiscent of the modules defined by AldolaseC expression. Finally, we showed that Sphk1 deletion acts on the ATXN1[82Q] protein expression and prevents PC degeneration. Taken together, our results demonstrate that there are regional differences in sphingolipid metabolism and that this metabolism is directly involved in PC degeneration in Atxn1[82Q]/+ mice.
Collapse
|
14
|
Akbari E, Spychalski GB, Menyhert MM, Rangharajan KK, Tinapple JW, Prakash S, Song JW. Endothelial barrier function is co-regulated at vessel bifurcations by fluid forces and sphingosine-1-phosphate. BIOMATERIALS AND BIOSYSTEMS 2021; 3:100020. [PMID: 35317095 PMCID: PMC8936769 DOI: 10.1016/j.bbiosy.2021.100020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 05/12/2021] [Accepted: 05/29/2021] [Indexed: 12/31/2022] Open
Abstract
Sphingosine-1-phosphate (S1P) is a bioactive sphingolipid mediator of endothelial barrier function. Prior studies have implicated mechanical stimulation due to intravascular laminar shear stress in co-regulating S1P signaling in endothelial cells (ECs). Yet, vascular networks in vivo consist of vessel bifurcations, and this geometry generates hemodynamic forces at the bifurcation point distinct from laminar shear stress. However, the role of these forces at vessel bifurcations in regulating S1P-dependent endothelial barrier function is not known. In this study, we implemented a microfluidic platform that recapitulates the flow dynamics of vessel bifurcations with in situ quantification of the permeability of microvessel analogues. Co-application of S1P with impinging bifurcated fluid flow, which is characterized by approximately zero shear stress and 38 dyn•cm-2 stagnation pressure at the vessel bifurcation point, promotes vessel stabilization. Similarly, co-treatment of S1P with 3 dyn•cm-2 laminar shear stress is also protective of endothelial barrier function. Moreover, it is shown that vessel stabilization due to bifurcated fluid flow and laminar shear stress is dependent on S1P receptor 1 or 2 signaling. Collectively, these findings demonstrate the endothelium-protective function of fluid forces at vessel bifurcations and their involvement in coordinating S1P-dependent regulation of vessel permeability.
Collapse
Affiliation(s)
- Ehsan Akbari
- Department of Mechanical and Aerospace Engineering, The Ohio State University, Columbus, OH, United States, 43210
| | - Griffin B. Spychalski
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH, United States, 43210
| | - Miles M. Menyhert
- Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, OH, United States, 43210
| | - Kaushik K. Rangharajan
- Department of Mechanical and Aerospace Engineering, The Ohio State University, Columbus, OH, United States, 43210
| | - Joseph W. Tinapple
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH, United States, 43210
| | - Shaurya Prakash
- Department of Mechanical and Aerospace Engineering, The Ohio State University, Columbus, OH, United States, 43210
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States, 43210
| | - Jonathan W. Song
- Department of Mechanical and Aerospace Engineering, The Ohio State University, Columbus, OH, United States, 43210
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States, 43210
| |
Collapse
|
15
|
Song H, McEwen HP, Duncan T, Lee JY, Teo JD, Don AS. Sphingosine kinase 2 is essential for remyelination following cuprizone intoxication. Glia 2021; 69:2863-2881. [PMID: 34399014 DOI: 10.1002/glia.24074] [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: 02/04/2021] [Revised: 08/03/2021] [Accepted: 08/05/2021] [Indexed: 01/16/2023]
Abstract
Therapeutics that promote oligodendrocyte survival and remyelination are needed to restore neurological function in demyelinating diseases. Sphingosine 1-phosphate (S1P) is an essential lipid metabolite that signals through five G-protein coupled receptors. S1P receptor agonists such as Fingolimod are valuable immunosuppressants used to treat multiple sclerosis, and promote oligodendrocyte survival. However, the role for endogenous S1P, synthesized by the enzyme sphingosine kinase 2 (SphK2), in oligodendrocyte survival and myelination has not been established. This study investigated the requirement for SphK2 in oligodendrocyte survival and remyelination using the cuprizone mouse model of acute demyelination, followed by spontaneous remyelination. Oligodendrocyte density did not differ between untreated wild-type (WT) and SphK2 knockout (SphK2-/- ) mice. However, cuprizone treatment caused significantly greater loss of mature oligodendrocytes in SphK2-/- compared to WT mice. Following cuprizone withdrawal, spontaneous remyelination occurred in WT but not SphK2-/- mice, even though progenitor and mature oligodendrocyte density increased in both genotypes. Levels of cytotoxic sphingosine and ceramide were higher in the corpus callosum of SphK2-/- mice, and in contrast to WT mice, did not decline following cuprizone withdrawal in SphK2-/- mice. We also observed a significant reduction in myelin thickness with aging in SphK2-/- compared to WT mice. These results provide the first evidence that SphK2, the dominant enzyme catalyzing S1P synthesis in the adult brain, is essential for remyelination following a demyelinating insult and myelin maintenance with aging. We propose that persistently high levels of sphingosine and ceramide, a direct consequence of SphK2 deficiency, may block remyelination.
Collapse
Affiliation(s)
- Huitong Song
- Centenary Institute, The University of Sydney, Camperdown, New South Wales, Australia
| | - Holly P McEwen
- Centenary Institute, The University of Sydney, Camperdown, New South Wales, Australia
| | - Thomas Duncan
- School of Medical Sciences, UNSW Sydney, Sydney, New South Wales, Australia
| | - Jun Yup Lee
- Centenary Institute, The University of Sydney, Camperdown, New South Wales, Australia
| | - Jonathan D Teo
- Centenary Institute, The University of Sydney, Camperdown, New South Wales, Australia
| | - Anthony S Don
- Centenary Institute, The University of Sydney, Camperdown, New South Wales, Australia.,School of Medical Sciences, The University of Sydney, Camperdown, New South Wales, Australia
| |
Collapse
|
16
|
Activation of sphingosine-1-phosphate receptor subtype 1 in the central nervous system contributes to morphine-induced hyperalgesia and antinociceptive tolerance in rodents. Pain 2021; 161:2107-2118. [PMID: 32301840 DOI: 10.1097/j.pain.0000000000001888] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Accepted: 03/30/2020] [Indexed: 12/31/2022]
Abstract
ABSTRACT Morphine-induced alterations in sphingolipid metabolism in the spinal cord and increased formation of the bioactive sphingolipid metabolite sphingosine-1-phosphate (S1P) have been implicated in the development of morphine-induced hyperalgesia (OIH; increased pain sensitivity) and antinociceptive tolerance. These adverse effects hamper opioid use for treating chronic pain and contribute to dependence and abuse. S1P produces distinct effects through 5 G-protein-coupled receptors (S1PR1-5) and several intracellular targets. How S1P exerts its effects in response to morphine remains unknown. Here, we report that S1P contributes to the development of morphine-induced hyperalgesia and tolerance through S1P receptor subtype 1 (S1PR1) signaling in uninjured male and female rodents, which can be blocked by targeting S1PR1 with S1PR1 antagonists or RNA silencing. In mouse neuropathic pain models, S1PR1 antagonists blocked the development of tolerance to the antiallodynic effects of morphine without altering morphine pharmacokinetics and prevented prolonged morphine-induced neuropathic pain. Targeting S1PR1 reduced morphine-induced neuroinflammatory events in the dorsal horn of the spinal cord: increased glial marker expression, mitogen-activated protein kinase p38 and nuclear factor κB activation, and increased inflammatory cytokine expression, such as interleukin-1β, a cytokine central in the modulation of opioid-induced neural plasticity. Our results identify S1PR1 as a critical path for S1P signaling in response to sustained morphine and reveal downstream neuroinflammatory pathways impacted by S1PR1 activation. Our data support investigating S1PR1 antagonists as a clinical approach to mitigate opioid-induced adverse effects and repurposing the functional S1PR1 antagonist FTY720, which is FDA-approved for multiple sclerosis, as an opioid adjunct.
Collapse
|
17
|
Goto H, Miyamoto M, Kihara A. Direct uptake of sphingosine-1-phosphate independent of phospholipid phosphatases. J Biol Chem 2021; 296:100605. [PMID: 33785361 PMCID: PMC8093947 DOI: 10.1016/j.jbc.2021.100605] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 03/23/2021] [Accepted: 03/26/2021] [Indexed: 12/20/2022] Open
Abstract
Sphingosine-1-phosphate (S1P) is a lipid mediator that is relatively abundant in plasma and plays an important role in the vascular and immune systems. To date, the only known mechanism for removing S1P from plasma has been dephosphorylation by phospholipid phosphatases (PLPPs) on the surface of cells in contact with the plasma. However, there remains a possibility that PLPP-independent dephosphorylation or direct S1P uptake into cells could occur. To examine these possibilities, here we generated triple KO (TKO) HAP1 cells that lacked all PLPPs (PLPP1–3) present in mammals. In the TKO cells, the intracellular metabolism of externally added deuterium-labeled S1P to ceramide was reduced to 17% compared with the WT cells, indicating that most extracellular S1P is dephosphorylated by PLPPs and then taken up into cells. However, this result also reveals the existence of a PLPP-independent S1P uptake pathway. Tracer experiments using [32P]S1P showed the existence of a direct S1P uptake pathway that functions without prior dephosphorylation. Overexpression of sphingolipid transporter 2 (SPNS2) or of major facilitator superfamily domain containing 2B (MFSD2B), both known S1P efflux transporters, in TKO cells increased the direct uptake of S1P, whereas KO of MFSD2B in TKO cells reduced this uptake. These results suggest that these are channel-type transporters and capable of not only exporting but also importing S1P. Furthermore, we observed that erythroid cells expressing MFSD2B, exhibited high S1P uptake activity. Our findings describing direct S1P uptake may contribute to the elucidation of the molecular mechanisms that regulate plasma S1P concentration.
Collapse
Affiliation(s)
- Hirotaka Goto
- Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo, Japan
| | | | - Akio Kihara
- Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo, Japan.
| |
Collapse
|
18
|
Chun J, Giovannoni G, Hunter SF. Sphingosine 1-phosphate Receptor Modulator Therapy for Multiple Sclerosis: Differential Downstream Receptor Signalling and Clinical Profile Effects. Drugs 2021; 81:207-231. [PMID: 33289881 PMCID: PMC7932974 DOI: 10.1007/s40265-020-01431-8] [Citation(s) in RCA: 87] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Lysophospholipids are a class of bioactive lipid molecules that produce their effects through various G protein-coupled receptors (GPCRs). Sphingosine 1-phosphate (S1P) is perhaps the most studied lysophospholipid and has a role in a wide range of physiological and pathophysiological events, via signalling through five distinct GPCR subtypes, S1PR1 to S1PR5. Previous and continuing investigation of the S1P pathway has led to the approval of three S1PR modulators, fingolimod, siponimod and ozanimod, as medicines for patients with multiple sclerosis (MS), as well as the identification of new S1PR modulators currently in clinical development, including ponesimod and etrasimod. S1PR modulators have complex effects on S1PRs, in some cases acting both as traditional agonists as well as agonists that produce functional antagonism. S1PR subtype specificity influences their downstream effects, including aspects of their benefit:risk profile. Some S1PR modulators are prodrugs, which require metabolic modification such as phosphorylation via sphingosine kinases, resulting in different pharmacokinetics and bioavailability, contrasting with others that are direct modulators of the receptors. The complex interplay of these characteristics dictates the clinical profile of S1PR modulators. This review focuses on the S1P pathway, the characteristics and S1PR binding profiles of S1PR modulators, the mechanisms of action of S1PR modulators with regard to immune cell trafficking and neuroprotection in MS, together with a summary of the clinical effectiveness of the S1PR modulators that are approved or in late-stage development for patients with MS. Sphingosine 1-phosphate receptor modulator therapy for multiple sclerosis: differential downstream receptor signalling and clinical profile effects (MP4 65540 kb).
Collapse
Affiliation(s)
- Jerold Chun
- Sanford Burnham Prebys Medical Discovery Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037 USA
| | - Gavin Giovannoni
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, 4 Newark St, London, E1 2AT UK
| | - Samuel F. Hunter
- Advanced Neurosciences Institute, 101 Forrest Crossing Blvd STE 103, Franklin, TN 37064 USA
| |
Collapse
|
19
|
Bouscary A, Quessada C, René F, Spedding M, Turner BJ, Henriques A, Ngo ST, Loeffler JP. Sphingolipids metabolism alteration in the central nervous system: Amyotrophic lateral sclerosis (ALS) and other neurodegenerative diseases. Semin Cell Dev Biol 2020; 112:82-91. [PMID: 33160824 DOI: 10.1016/j.semcdb.2020.10.008] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 10/14/2020] [Accepted: 10/19/2020] [Indexed: 12/11/2022]
Abstract
Sphingolipids are complex lipids. They play a structural role in neurons, but are also involved in regulating cellular communication, and neuronal differentiation and maturation. There is increasing evidence to suggest that dysregulated metabolism of sphingolipids is linked to neurodegenerative processes in amyotrophic lateral sclerosis (ALS), Parkinson's disease and Gaucher's disease. In this review, we provide an overview of the role of sphingolipids in the development and maintenance of the nervous system. We describe the implications of altered metabolism of sphingolipids in the pathophysiology of certain neurodegenerative diseases, with a primary focus on ALS. Finally, we provide an update of potential treatments that could be used to target the metabolism of sphingolipids in neurodegenerative diseases.
Collapse
Affiliation(s)
- Alexandra Bouscary
- Université de Strasbourg, UMR_S 1118, Fédération de Médecine Translationnelle, CRBS, 1 rue Eugène Boeckel, 67000 Strasbourg, France; INSERM, U1118, Mécanismes Centraux et Périphériques de la Neurodégénérescence, CRBS, 1 rue Eugène Boeckel, 67000 Strasbourg, France
| | - Cyril Quessada
- Université de Strasbourg, UMR_S 1118, Fédération de Médecine Translationnelle, CRBS, 1 rue Eugène Boeckel, 67000 Strasbourg, France; INSERM, U1118, Mécanismes Centraux et Périphériques de la Neurodégénérescence, CRBS, 1 rue Eugène Boeckel, 67000 Strasbourg, France
| | - Frédérique René
- Université de Strasbourg, UMR_S 1118, Fédération de Médecine Translationnelle, CRBS, 1 rue Eugène Boeckel, 67000 Strasbourg, France; INSERM, U1118, Mécanismes Centraux et Périphériques de la Neurodégénérescence, CRBS, 1 rue Eugène Boeckel, 67000 Strasbourg, France
| | - Michael Spedding
- Spedding Research Solutions SAS, 6 rue Ampere, 78650 Le Vesinet, France
| | - Bradley J Turner
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, 30 Royal Parade, Parkville, VIC 3052, Australia
| | | | - Shyuan T Ngo
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Building 75, Cnr College Rd & Cooper Rd, Brisbane city, QLD 4072, Australia; Centre for Clinical Research, The University of Queensland, Building 71/918, Royal Brisbane & Women's Hospital Campus, Herston, QLD 4029, Australia; Queensland Brain Institute Building 79, The University of Queensland, St Lucia, QLD 4072, Australia
| | - Jean-Philippe Loeffler
- Université de Strasbourg, UMR_S 1118, Fédération de Médecine Translationnelle, CRBS, 1 rue Eugène Boeckel, 67000 Strasbourg, France; INSERM, U1118, Mécanismes Centraux et Périphériques de la Neurodégénérescence, CRBS, 1 rue Eugène Boeckel, 67000 Strasbourg, France.
| |
Collapse
|
20
|
Squillace S, Spiegel S, Salvemini D. Targeting the Sphingosine-1-Phosphate Axis for Developing Non-narcotic Pain Therapeutics. Trends Pharmacol Sci 2020; 41:851-867. [PMID: 33010954 DOI: 10.1016/j.tips.2020.09.006] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 09/02/2020] [Accepted: 09/10/2020] [Indexed: 02/07/2023]
Abstract
Chronic pain is a life-altering condition affecting millions of people. Current treatments are inadequate and prolonged therapies come with severe side effects, especially dependence and addiction to opiates. Identification of non-narcotic analgesics is of paramount importance. Preclinical and clinical studies suggest that sphingolipid metabolism alterations contribute to neuropathic pain development. Functional sphingosine-1-phosphate (S1P) receptor 1 (S1PR1) antagonists, such as FTY720/fingolimod, used clinically for non-pain conditions, are emerging as non-narcotic analgesics, supporting the repurposing of fingolimod for chronic pain treatment and energizing drug discovery focused on S1P signaling. Here, we summarize the role of S1P in pain to highlight the potential of targeting the S1P axis towards development of non-narcotic therapeutics, which, in turn, will hopefully help lessen misuse of opioid pain medications and address the ongoing opioid epidemic.
Collapse
Affiliation(s)
- Silvia Squillace
- Department of Pharmacology and Physiology and the Henry and Amelia Nasrallah Center for Neuroscience, Saint Louis University School of Medicine, St. Louis, MO 63104, USA
| | - Sarah Spiegel
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University School of Medicine, Richmond, VA 23298, USA
| | - Daniela Salvemini
- Department of Pharmacology and Physiology and the Henry and Amelia Nasrallah Center for Neuroscience, Saint Louis University School of Medicine, St. Louis, MO 63104, USA.
| |
Collapse
|
21
|
Positron Emission Tomography in the Inflamed Cerebellum: Addressing Novel Targets among G Protein-Coupled Receptors and Immune Receptors. Pharmaceutics 2020; 12:pharmaceutics12100925. [PMID: 32998351 PMCID: PMC7601272 DOI: 10.3390/pharmaceutics12100925] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 09/17/2020] [Accepted: 09/25/2020] [Indexed: 01/12/2023] Open
Abstract
Inflammatory processes preceding clinical manifestation of brain diseases are moving increasingly into the focus of positron emission tomographic (PET) investigations. A key role in inflammation and as a target of PET imaging efforts is attributed to microglia. Cerebellar microglia, with a predominant ameboid and activated subtype, is of special interest also regarding improved and changing knowledge on functional involvement of the cerebellum in mental activities in addition to its regulatory role in motor function. The present contribution considers small molecule ligands as potential PET tools for the visualization of several receptors recognized to be overexpressed in microglia and which can potentially serve as indicators of inflammatory processes in the cerebellum. The sphingosine 1 phosphate receptor 1 (S1P1), neuropeptide Y receptor 2 (NPY2) and purinoceptor Y12 (P2Y12) cannabinoid receptors and the chemokine receptor CX3CR1 as G-protein-coupled receptors and the ionotropic purinoceptor P2X7 provide structures with rather classical binding behavior, while the immune receptor for advanced glycation end products (RAGE) and the triggering receptor expressed on myeloid cells 2 (TREM2) might depend for instance on further accessory proteins. Improvement in differentiation between microglial functional subtypes in comparison to the presently used 18 kDa translocator protein ligands as well as of the knowledge on the role of polymorphisms are special challenges in such developments.
Collapse
|
22
|
Giussani P, Prinetti A, Tringali C. The role of Sphingolipids in myelination and myelin stability and their involvement in childhood and adult demyelinating disorders. J Neurochem 2020; 156:403-414. [PMID: 33448358 DOI: 10.1111/jnc.15133] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 07/14/2020] [Accepted: 07/17/2020] [Indexed: 01/02/2023]
Abstract
Multiple sclerosis (MS) represents the most common demyelinating disease affecting the central nervous system (CNS) in adults as well as in children. Furthermore, in children, in addition to acquired diseases such as MS, genetically inherited diseases significantly contribute to the incidence of demyelinating disorders. Some genetic defects lead to sphingolipid alterations that are able to elicit neurological symptoms. Sphingolipids are essential for brain development, and their aberrant functionality may thus contribute to demyelinating diseases such as MS. In particular, sphingolipidoses caused by deficits of sphingolipid-metabolizing enzymes, are often associated with demyelination. Sphingolipids are not only structural molecules but also bioactive molecules involved in the regulation of cellular events such as development of the nervous system, myelination and maintenance of myelin stability. Changes in the sphingolipid metabolism deeply affect plasma membrane organization. Thus, changes in myelin sphingolipid composition might crucially contribute to the phenotype of diseases characterized by demyelinalization. Here, we review key features of several sphingolipids such as ceramide/dihydroceramide, sphingosine/dihydrosphingosine, glucosylceramide and, galactosylceramide which act in myelin formation during rat brain development and in human brain demyelination during the pathogenesis of MS, suggesting that this knowledge could be useful in identifying targets for possible therapies.
Collapse
Affiliation(s)
- Paola Giussani
- Department of Medical Biotechnology and Translational Medicine, Università di Milano, LITA Segrate, Segrate, Italy
| | - Alessandro Prinetti
- Department of Medical Biotechnology and Translational Medicine, Università di Milano, LITA Segrate, Segrate, Italy
| | - Cristina Tringali
- Department of Medical Biotechnology and Translational Medicine, Università di Milano, LITA Segrate, Segrate, Italy
| |
Collapse
|
23
|
Weigel C, Hüttner SS, Ludwig K, Krieg N, Hofmann S, Schröder NH, Robbe L, Kluge S, Nierhaus A, Winkler MS, Rubio I, von Maltzahn J, Spiegel S, Gräler MH. S1P lyase inhibition protects against sepsis by promoting disease tolerance via the S1P/S1PR3 axis. EBioMedicine 2020; 58:102898. [PMID: 32711251 PMCID: PMC7381498 DOI: 10.1016/j.ebiom.2020.102898] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 06/19/2020] [Accepted: 07/01/2020] [Indexed: 12/29/2022] Open
Abstract
Background One-third of all deaths in hospitals are caused by sepsis. Despite its demonstrated prevalence and high case fatality rate, antibiotics remain the only target-oriented treatment option currently available. Starting from results showing that low-dose anthracyclines protect against sepsis in mice, we sought to find new causative treatment options to improve sepsis outcomes. Methods Sepsis was induced in mice, and different treatment options were evaluated regarding cytokine and biomarker expression, lung epithelial cell permeability, autophagy induction, and survival benefit. Results were validated in cell culture experiments and correlated with patient samples. Findings Effective low-dose epirubicin treatment resulted in substantial downregulation of the sphingosine 1-phosphate (S1P) degrading enzyme S1P lyase (SPL). Consequent accumulation and secretion of S1P in lung parenchyma cells stimulated the S1P-receptor type 3 (S1PR3) and mitogen-activated protein kinases p38 and ERK, reducing tissue damage via increased disease tolerance. The protective effects of SPL inhibition were absent in S1PR3 deficient mice. Sepsis patients showed increased expression of SPL, stable expression of S1PR3, and increased levels of mucin-1 and surfactant protein D as indicators of lung damage. Interpretation Our work highlights a tissue-protective effect of SPL inhibition in sepsis due to activation of the S1P/S1PR3 axis and implies that SPL inhibitors and S1PR3 agonists might be potential therapeutics to protect against sepsis by increasing disease tolerance against infections. Funding This study was supported by the Center for Sepsis Control and Care (CSCC), the German Research Foundation (DFG), RTG 1715 (to M. H. G. and I. R.) and the National Institutes of Health, Grant R01GM043880 (to S. S.).
Collapse
Affiliation(s)
- Cynthia Weigel
- Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Am Klinikum 1, 07747 Jena, Germany; Center for Molecular Biomedicine, Jena University Hospital, 07745 Jena, Germany; Leibniz Institute on Aging - Fritz Lipmann Institute (FLI), 07745 Jena, Germany
| | - Sören S Hüttner
- Leibniz Institute on Aging - Fritz Lipmann Institute (FLI), 07745 Jena, Germany
| | - Kristin Ludwig
- Center for Molecular Biomedicine, Jena University Hospital, 07745 Jena, Germany; Institute of Molecular Cell Biology, Jena University Hospital, 07745 Jena, Germany
| | - Nadine Krieg
- Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Am Klinikum 1, 07747 Jena, Germany; Center for Molecular Biomedicine, Jena University Hospital, 07745 Jena, Germany
| | - Susann Hofmann
- Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Am Klinikum 1, 07747 Jena, Germany; Center for Molecular Biomedicine, Jena University Hospital, 07745 Jena, Germany; Center for Sepsis Control and Care, Jena University Hospital, 07740 Jena, Germany
| | - Nathalie H Schröder
- Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Am Klinikum 1, 07747 Jena, Germany; Center for Molecular Biomedicine, Jena University Hospital, 07745 Jena, Germany
| | - Linda Robbe
- Department of Intensive Care Medicine, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Stefan Kluge
- Department of Intensive Care Medicine, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Axel Nierhaus
- Department of Intensive Care Medicine, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Martin S Winkler
- Department of Anesthesiology and Intensive Care Medicine, University Medical Center Göttingen, 37075 Göttingen, Germany
| | - Ignacio Rubio
- Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Am Klinikum 1, 07747 Jena, Germany; Center for Sepsis Control and Care, Jena University Hospital, 07740 Jena, Germany
| | - Julia von Maltzahn
- Leibniz Institute on Aging - Fritz Lipmann Institute (FLI), 07745 Jena, Germany
| | - Sarah Spiegel
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University School of Medicine, Richmond, VA 23298, USA
| | - Markus H Gräler
- Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Am Klinikum 1, 07747 Jena, Germany; Center for Molecular Biomedicine, Jena University Hospital, 07745 Jena, Germany; Center for Sepsis Control and Care, Jena University Hospital, 07740 Jena, Germany.
| |
Collapse
|
24
|
Murine platelet production is suppressed by S1P release in the hematopoietic niche, not facilitated by blood S1P sensing. Blood Adv 2020; 3:1702-1713. [PMID: 31171507 DOI: 10.1182/bloodadvances.2019031948] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Accepted: 04/17/2019] [Indexed: 02/06/2023] Open
Abstract
The bioactive lipid mediator sphingosine 1-phosphate (S1P) was recently assigned critical roles in platelet biology: whereas S1P1 receptor-mediated S1P gradient sensing was reported to be essential for directing proplatelet extensions from megakaryocytes (MKs) toward bone marrow sinusoids, MK sphingosine kinase 2 (Sphk2)-derived S1P was reported to further promote platelet shedding through receptor-independent intracellular actions, and platelet aggregation through S1P1 Yet clinical use of S1P pathway modulators including fingolimod has not been associated with risk of bleeding or thrombosis. We therefore revisited the role of S1P in platelet biology in mice. Surprisingly, no reduction in platelet counts was observed when the vascular S1P gradient was ablated by impairing S1P provision to plasma or S1P degradation in interstitial fluids, nor when gradient sensing was impaired by S1pr1 deletion selectively in MKs. Moreover, S1P1 expression and signaling were both undetectable in mature MKs in situ, and MK S1pr1 deletion did not affect platelet aggregation or spreading. When S1pr1 deletion was induced in hematopoietic progenitor cells, platelet counts were instead significantly elevated. Isolated global Sphk2 deficiency was associated with thrombocytopenia, but this was not replicated by MK-restricted Sphk2 deletion and was reversed by compound deletion of either Sphk1 or S1pr2, suggesting that this phenotype arises from increased S1P export and S1P2 activation secondary to redistribution of sphingosine to Sphk1. Consistent with clinical observations, we thus observe no essential role for S1P1 in facilitating platelet production or activation. Instead, S1P restricts megakaryopoiesis through S1P1, and can further suppress thrombopoiesis through S1P2 when aberrantly secreted in the hematopoietic niche.
Collapse
|
25
|
Langeslag M, Kress M. The ceramide-S1P pathway as a druggable target to alleviate peripheral neuropathic pain. Expert Opin Ther Targets 2020; 24:869-884. [PMID: 32589067 DOI: 10.1080/14728222.2020.1787989] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Introduction: Neuropathic pain disorders are diverse, and the currently available therapies are ineffective in the majority of cases. Therefore, there is a major need for gaining novel mechanistic insights and developing new treatment strategies for neuropathic pain. Areas covered: We performed an in-depth literature search on the molecular mechanisms and systemic importance of the ceramide-to-S1P rheostat regulating neuron function and neuroimmune interactions in the development of neuropathic pain. Expert opinion: The S1P receptor modulator FTY720 (fingolimod, Gilenya®), LPA receptor antagonists and several mechanistically related compounds in clinical development raise great expectations for treating neuropathic pain disorders. Research on S1P receptors, S1P receptor modulators or SPHK inhibitors with distinct selectivity, pharmacokinetics and safety must provide more mechanistic insight into whether they may qualify as useful treatment options for neuropathic pain disorders. The functional relevance of genetic variations within the ceramide-to-S1P rheostat should be explored for an enhanced understanding of neuropathic pain pathogenesis. The ceramide-to-S1P rheostat is emerging as a critically important regulator hub of neuroimmune interactions along the pain pathway, and improved mechanistic insight is required to develop more precise and effective drug treatment options for patients suffering from neuropathic pain disorders.
Collapse
Affiliation(s)
- Michiel Langeslag
- Institute of Physiology, DPMP, Medical University Innsbruck , Austria
| | - Michaela Kress
- Institute of Physiology, DPMP, Medical University Innsbruck , Austria
| |
Collapse
|
26
|
Stepanovska B, Huwiler A. Targeting the S1P receptor signaling pathways as a promising approach for treatment of autoimmune and inflammatory diseases. Pharmacol Res 2020; 154:104170. [DOI: 10.1016/j.phrs.2019.02.009] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Revised: 02/04/2019] [Accepted: 02/05/2019] [Indexed: 11/26/2022]
|
27
|
Marciniak A, Camp SM, Garcia JGN, Polt R. In silico Docking Studies of Fingolimod and S1P 1 Agonists. Front Pharmacol 2020; 11:247. [PMID: 32210822 PMCID: PMC7076195 DOI: 10.3389/fphar.2020.00247] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Accepted: 02/24/2020] [Indexed: 11/13/2022] Open
Abstract
The sphingosine-1-phosphate receptor 1 (S1P1), originally the endothelial differentiation gene 1 receptor (EDG-1), is one of five G protein–coupled receptors (GPCRs) S1P1–5 that bind to and are activated by sphingosine-1-phosphate (S1P). The lipid S1P is an intermediate in sphingolipid homeostasis, and S1P1 is a major medical target for immune system modulation; agonism of the receptor produces a myriad of biological responses, including endothelial cell barrier integrity, chemotaxis, lymphocyte trafficking/targeting, angiogenesis, as well as regulation of the cardiovascular system. Use of in silico docking simulations on the crystal structure of S1P1 allows for pinpointing the residues within the receptor’s active site that actively contribute to the binding of S1P, and point to how these specific interactions can be exploited to design more effective synthetic analogs to specifically target S1P1 in the presence of the closely related receptors S1P2, S1P3, S1P4, and S1P5. We examined the binding properties of the endogenous substrate as well as a selection of synthetic sphingosine-derived S1P1 modulators of S1P1 with in silico docking simulations using the software package Molecular Operating Environment® (MOE®). The modeling studies reveal the relevance of phosphorylation, i.e., the presence of a phosphate or phosphonate moiety within the substrate for successful binding to occur, and indicate which residues are responsible for S1P1 binding of the most prominent sphingosine-1-phosphate receptor (S1PR) modulators, including fingolimod and its structural relatives. Furthermore, trends in steric preferences as for the binding of enantiomers to S1P1 could be observed, facilitating future design of receptor-specific substrates to precisely target the active site of S1P1.
Collapse
Affiliation(s)
- Alexander Marciniak
- Department of Chemistry and Biochemistry, The University of Arizona, Tucson, AZ, United States
| | - Sara M Camp
- Department of Medicine, The University of Arizona, Tucson, AZ, United States
| | - Joe G N Garcia
- Department of Medicine, The University of Arizona, Tucson, AZ, United States
| | - Robin Polt
- Department of Chemistry and Biochemistry, The University of Arizona, Tucson, AZ, United States
| |
Collapse
|
28
|
Dorsey SG, Kleckner IR, Barton D, Mustian K, O'Mara A, St Germain D, Cavaletti G, Danhauer SC, Hershman DL, Hohmann AG, Hoke A, Hopkins JO, Kelly KP, Loprinzi CL, McLeod HL, Mohile S, Paice J, Rowland JH, Salvemini D, Segal RA, Smith EL, Stevens WM, Janelsins MC. The National Cancer Institute Clinical Trials Planning Meeting for Prevention and Treatment of Chemotherapy-Induced Peripheral Neuropathy. J Natl Cancer Inst 2020; 111:531-537. [PMID: 30715378 DOI: 10.1093/jnci/djz011] [Citation(s) in RCA: 81] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Revised: 01/04/2019] [Accepted: 01/25/2019] [Indexed: 12/25/2022] Open
Abstract
Although recent scientific advances have improved our understanding of basic biological mechanisms underlying chemotherapy-induced peripheral neuropathy (CIPN), few interventions are available to prevent or treat CIPN. Although some biological targets from preclinical studies show promise in nonhuman animal models, few targets have been translated to successful clinical trials. To address this problem, the National Cancer Institute's Symptom Management and Health-Related Quality of Life Steering Committee convened a meeting of experts in the CIPN and oncology symptom management fields to participate in a Clinical Trials Planning Meeting (CTPM). Investigators presented data from preclinical and translational studies for possible CIPN interventions; these were evaluated for readiness of randomized clinical trial testing by experts, and recommendations were provided. Breakout sessions were convened to discuss and develop future studies. The CTPM experts concluded that there is compelling evidence to move forward with selected pharmacological and nonpharmacological clinical trials for the prevention and treatment of CIPN. Several key feasibility issues need to be addressed, however. These include identification of optimal outcome measures to define the CIPN phenotype, establishment of parameters that guide the evaluation of clinically meaningful effects, and adoption of approaches for inclusion of translational and biomarker and/or genetic measures. The results of the CTPM provide support for conducting clinical trials that include both pharmacological and nonpharmacological approaches, alone or in combination, with biomarkers, genetics, or other measures designed to inform underlying CIPN mechanisms. Several working groups were formed to design rigorous CIPN clinical trials, the results of which are ongoing.
Collapse
Affiliation(s)
- Susan G Dorsey
- Department of Pain and Translational Symptom Science, University of Maryland School of Nursing, Center to Advance Chronic Pain Research, Baltimore, MD
| | - Ian R Kleckner
- Department of Surgery, University of Rochester Medical Center, Cancer Control Program, Wilmot Cancer Institute, Rochester, NY
| | - Debra Barton
- University of Michigan School of Nursing, Ann Arbor, MI
| | - Karen Mustian
- Department of Surgery, University of Rochester Medical Center, Cancer Control Program, Wilmot Cancer Institute, Rochester, NY
| | - Ann O'Mara
- Division of Cancer Prevention, National Cancer Institute, Bethesda, MD
| | - Diane St Germain
- Division of Cancer Prevention, National Cancer Institute, Bethesda, MD
| | - Guido Cavaletti
- Experimental Neurology Unit, School of Medicine and Surgery, University Milano-Bicocca, Monza, Italy
| | - Suzanne C Danhauer
- Division of Public Health Sciences, Department of Social Sciences and Health Policy, Wake Forest School of Medicine, Winston Salem, NC
| | - Dawn L Hershman
- Department of Medicine, Columbia University Medical Center, New York, NY
| | - Andrea G Hohmann
- Department of Psychological and Brain Sciences, Program in Neuroscience and Gill Center for Biomolecular Science, Indiana University, Bloomington, IN
| | - Ahmet Hoke
- Department of Neurology, School of Medicine, Johns Hopkins University, Baltimore, MD
| | | | - Katherine P Kelly
- Children's National Health System, Department of Nursing Science, Professional Practice, and Quality, George Washington School of Medicine and Health Sciences, Washington, DC
| | | | | | - Supriya Mohile
- Department of Surgery, University of Rochester Medical Center, Cancer Control Program, Wilmot Cancer Institute, Rochester, NY
| | - Judith Paice
- Hematology-Oncology Division, Northwestern University, Chicago, IL
| | | | - Daniela Salvemini
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, St. Louis, MO
| | - Rosalind A Segal
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA
| | | | | | - Michelle C Janelsins
- Department of Surgery, University of Rochester Medical Center, Cancer Control Program, Wilmot Cancer Institute, Rochester, NY
| |
Collapse
|
29
|
Abstract
There is substantial evidence that the enzymes, sphingosine kinase 1 and 2, which catalyse the formation of the bioactive lipid sphingosine 1-phosphate, are involved in pathophysiological processes. In this chapter, we appraise the evidence that both enzymes are druggable and describe how isoform-specific inhibitors can be developed based on the plasticity of the sphingosine-binding site. This is contextualised with the effect of sphingosine kinase inhibitors in cancer, pulmonary hypertension, neurodegeneration, inflammation and sickling.
Collapse
Affiliation(s)
- Susan Pyne
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde , Glasgow, Scotland, UK
| | - David R Adams
- Institute of Chemical Sciences, Heriot-Watt University, Edinburgh, Scotland, UK
| | - Nigel J Pyne
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde , Glasgow, Scotland, UK.
| |
Collapse
|
30
|
Stockstill K, Wahlman C, Braden K, Chen Z, Yosten GL, Tosh D, Jacobson K, Doyle T, Samson W, Salvemini D. Sexually dimorphic therapeutic response in bortezomib-induced neuropathic pain reveals altered pain physiology in female rodents. Pain 2020; 161:177-184. [PMID: 31490328 PMCID: PMC6923586 DOI: 10.1097/j.pain.0000000000001697] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Chemotherapy-induced neuropathic pain (CINP) in both sexes compromises many current chemotherapeutics and lacks an FDA-approved therapy. We recently identified the sphingosine-1-phosphate receptor subtype 1 (S1PR1) and A3 adenosine receptor subtype (A3AR) as novel targets for therapeutic intervention. Our work in male rodents using paclitaxel, oxaliplatin, and bortezomib showed robust inhibition of CINP with either S1PR1 antagonists or A3AR agonists. The S1PR1 functional antagonist FTY720 (Gilenya) is FDA-approved for treating multiple sclerosis, and selective A3AR agonists are in advanced clinical trials for cancer and inflammatory disorders, underscoring the need for their expedited trials in patients with CINP as chemotherapy adjuncts. Our findings reveal that S1PR1 antagonists and A3AR agonists mitigate paclitaxel and oxaliplatin CINP in female and male rodents, but failed to block or reverse bortezomib-induced neuropathic pain (BINP) in females. Although numerous mechanisms likely underlie these differences, we focused on receptor levels. We found that BINP in male rats, but not in female rats, was associated with increased expression of A3AR in the spinal cord dorsal horn, whereas S1PR1 levels were similar in both sexes. Thus, alternative mechanisms beyond receptor expression may account for sex differences in response to S1PR1 antagonists. Morphine and duloxetine, both clinical analgesics, reversed BINP in female mice, demonstrating that the lack of response is specific to S1PR1 and A3AR agents. Our findings suggest that A3AR- and S1PR1-based therapies are not viable approaches in preventing and treating BINP in females and should inform future clinical trials of these drugs as adjuncts to chemotherapy.
Collapse
Affiliation(s)
- Katherine Stockstill
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, 1402 South Grand Blvd., St. Louis, MO 63104, USA
| | - Carrie Wahlman
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, 1402 South Grand Blvd., St. Louis, MO 63104, USA
| | - Kathryn Braden
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, 1402 South Grand Blvd., St. Louis, MO 63104, USA
| | - Zhoumou Chen
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, 1402 South Grand Blvd., St. Louis, MO 63104, USA
| | - G. L. Yosten
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, 1402 South Grand Blvd., St. Louis, MO 63104, USA
| | - D.K. Tosh
- Molecular Recognition Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland 20892-0810, USA
| | - K.A. Jacobson
- Molecular Recognition Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland 20892-0810, USA
| | - T.M. Doyle
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, 1402 South Grand Blvd., St. Louis, MO 63104, USA
| | - W.K. Samson
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, 1402 South Grand Blvd., St. Louis, MO 63104, USA
| | - Daniela Salvemini
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, 1402 South Grand Blvd., St. Louis, MO 63104, USA
| |
Collapse
|
31
|
Tran C, Heng B, Teo JD, Humphrey SJ, Qi Y, Couttas TA, Stefen H, Brettle M, Fath T, Guillemin GJ, Don AS. Sphingosine 1-phosphate but not Fingolimod protects neurons against excitotoxic cell death by inducing neurotrophic gene expression in astrocytes. J Neurochem 2019; 153:173-188. [PMID: 31742704 DOI: 10.1111/jnc.14917] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Revised: 11/13/2019] [Accepted: 11/15/2019] [Indexed: 12/19/2022]
Abstract
Sphingosine 1-phosphate (S1P) is an essential lipid metabolite that signals through a family of five G protein-coupled receptors, S1PR1-S1PR5, to regulate cell physiology. The multiple sclerosis drug Fingolimod (FTY720) is a potent S1P receptor agonist that causes peripheral lymphopenia. Recent research has demonstrated direct neuroprotective properties of FTY720 in several neurodegenerative paradigms; however, neuroprotective properties of the native ligand S1P have not been established. We aimed to establish the significance of neurotrophic factor up-regulation by S1P for neuroprotection, comparing S1P with FTY720. S1P induced brain-derived neurotrophic factor (BDNF), leukemia inhibitory factor (LIF), platelet-derived growth factor B (PDGFB), and heparin-binding EGF-like growth factor (HBEGF) gene expression in primary human and murine astrocytes, but not in neurons, and to a much greater extent than FTY720. Accordingly, S1P but not FTY720 protected cultured neurons against excitotoxic cell death in a primary murine neuron-glia coculture model, and a neutralizing antibody to LIF blocked this S1P-mediated neuroprotection. Antagonists of S1PR1 and S1PR2 both inhibited S1P-mediated neurotrophic gene induction in human astrocytes, indicating that simultaneous activation of both receptors is required. S1PR2 signaling was transduced through Gα13 and the small GTPase Rho, and was necessary for the up-regulation and activation of the transcription factors FOS and JUN, which regulate LIF, BDNF, and HBEGF transcription. In summary, we show that S1P protects hippocampal neurons against excitotoxic cell death through up-regulation of neurotrophic gene expression, particularly LIF, in astrocytes. This up-regulation requires both S1PR1 and S1PR2 signaling. FTY720 does not activate S1PR2, explaining its relative inefficacy compared to S1P.
Collapse
Affiliation(s)
- Collin Tran
- School of Medical Sciences, UNSW Sydney, Kensington, NSW, Australia.,Centenary Institute, The University of Sydney, Camperdown, NSW, Australia
| | - Benjamin Heng
- MND Research Centre, Neuroinflammation group, Macquarie University, Sydney, NSW, Australia
| | - Jonathan D Teo
- Centenary Institute, The University of Sydney, Camperdown, NSW, Australia
| | - Sean J Humphrey
- School of Life and Environmental Sciences, The University of Sydney, Camperdown, NSW, Australia
| | - Yanfei Qi
- Centenary Institute, The University of Sydney, Camperdown, NSW, Australia
| | - Timothy A Couttas
- Centenary Institute, The University of Sydney, Camperdown, NSW, Australia
| | - Holly Stefen
- Dementia Research Centre and Department of Biomedical Sciences, Faculty of Medicine and Sciences, Macquarie University, Sydney, NSW, Australia
| | - Merryn Brettle
- School of Medical Sciences, UNSW Sydney, Kensington, NSW, Australia
| | - Thomas Fath
- School of Medical Sciences, UNSW Sydney, Kensington, NSW, Australia.,Dementia Research Centre and Department of Biomedical Sciences, Faculty of Medicine and Sciences, Macquarie University, Sydney, NSW, Australia
| | - Gilles J Guillemin
- MND Research Centre, Neuroinflammation group, Macquarie University, Sydney, NSW, Australia
| | - Anthony S Don
- Centenary Institute, The University of Sydney, Camperdown, NSW, Australia.,NHMRC Clinical Trials Centre, The University of Sydney, Camperdown, NSW, Australia
| |
Collapse
|
32
|
Shimano K, Maeda Y, Kataoka H, Murase M, Mochizuki S, Utsumi H, Oshita K, Sugahara K. Amiselimod (MT-1303), a novel sphingosine 1-phosphate receptor-1 functional antagonist, inhibits progress of chronic colitis induced by transfer of CD4+CD45RBhigh T cells. PLoS One 2019; 14:e0226154. [PMID: 31805144 PMCID: PMC6894856 DOI: 10.1371/journal.pone.0226154] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Accepted: 11/20/2019] [Indexed: 02/07/2023] Open
Abstract
Amiselimod (MT-1303) is a novel sphingosine 1-phosphate receptor-1 (S1P1 receptor) modulator with a more favorable cardiac safety profile than other S1P1 receptor modulators. MT-1303 phosphate (MT-1303-P), an active metabolite of MT-1303, exhibits S1P1 receptor agonism at a lower EC50 value than other S1P1 receptor modulators currently being developed. We aimed to evaluate the efficacy of MT-1303 and its mode of action in chronic colitis using an inflammatory bowel disease (IBD) model. Oral administration of MT-1303 (0.3 mg/kg) once daily for 3 days to mice almost completely abolished S1P1 receptor expression on CD4+ T cells from mesenteric lymph nodes, which corresponded to a marked decrease in CD4+ T cell count in peripheral blood, indicating that MT-1303-P acts as a functional antagonist of the S1P1 receptor. The potential benefit of MT-1303 for IBD was assessed using immunodeficient SCID mice with chronic colitis induced by adoptive transfer of CD4+CD45RBhigh T cells from BALB/c mice. An oral dose of 0.1 and 0.3 mg/kg MT-1303 administered daily one week after the cell transfer inhibited the development of chronic colitis with an efficacy comparable to that of an anti-mTNF-α mAb (250 μg/mouse). In addition, MT-1303 administration significantly reduced the number of infiltrating Th1 and Th17 cells into the lamina propria of the colon in colitis mice. Our results suggest that MT-1303 acts as a functional antagonist of the S1P1 receptor on lymphocytes, regulates lymphocyte trafficking, and inhibits infiltration of colitogenic Th1 and Th17 cells into the colon to inhibit the development of chronic colitis.
Collapse
Affiliation(s)
- Kyoko Shimano
- Sohyaku, Innovative Research Division, Mitsubishi Tanabe Pharma Corporation, Yokohama, Japan
| | - Yasuhiro Maeda
- Sohyaku, Innovative Research Division, Mitsubishi Tanabe Pharma Corporation, Yokohama, Japan
| | - Hirotoshi Kataoka
- Sohyaku, Innovative Research Division, Mitsubishi Tanabe Pharma Corporation, Yokohama, Japan
| | - Mikako Murase
- Sohyaku, Innovative Research Division, Mitsubishi Tanabe Pharma Corporation, Yokohama, Japan
| | - Sachiko Mochizuki
- Sohyaku, Innovative Research Division, Mitsubishi Tanabe Pharma Corporation, Yokohama, Japan
| | - Hiroyuki Utsumi
- Sohyaku, Innovative Research Division, Mitsubishi Tanabe Pharma Corporation, Yokohama, Japan
| | - Koichi Oshita
- Sohyaku, Innovative Research Division, Mitsubishi Tanabe Pharma Corporation, Yokohama, Japan
| | - Kunio Sugahara
- Sohyaku, Innovative Research Division, Mitsubishi Tanabe Pharma Corporation, Yokohama, Japan
- * E-mail:
| |
Collapse
|
33
|
Sphingosine Kinase 2 Potentiates Amyloid Deposition but Protects against Hippocampal Volume Loss and Demyelination in a Mouse Model of Alzheimer's Disease. J Neurosci 2019; 39:9645-9659. [PMID: 31641049 DOI: 10.1523/jneurosci.0524-19.2019] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2019] [Revised: 09/19/2019] [Accepted: 10/10/2019] [Indexed: 01/20/2023] Open
Abstract
Sphingosine 1-phosphate (S1P) is a potent vasculoprotective and neuroprotective signaling lipid, synthesized primarily by sphingosine kinase 2 (SK2) in the brain. We have reported pronounced loss of S1P and SK2 activity early in Alzheimer's disease (AD) pathogenesis, and an inverse correlation between hippocampal S1P levels and age in females, leading us to speculate that loss of S1P is a sensitizing influence for AD. Paradoxically, SK2 was reported to mediate amyloid β (Aβ) formation from amyloid precursor protein (APP) in vitro To determine whether loss of S1P sensitizes to Aβ-mediated neurodegeneration, we investigated whether SK2 deficiency worsens pathology and memory in male J20 (PDGFB-APPSwInd) mice. SK2 deficiency greatly reduced Aβ content in J20 mice, associated with significant improvements in epileptiform activity and cross-frequency coupling measured by hippocampal electroencephalography. However, several key measures of APPSwInd-dependent neurodegeneration were enhanced on the SK2-null background, despite reduced Aβ burden. These included hippocampal volume loss, oligodendrocyte attrition and myelin loss, and impaired performance in Y-maze and social novelty memory tests. Inhibition of the endosomal cholesterol exporter NPC1 greatly reduced sphingosine phosphorylation in glial cells, linking loss of SK2 activity and S1P in AD to perturbed endosomal lipid metabolism. Our findings establish SK2 as an important endogenous regulator of both APP processing to Aβ, and oligodendrocyte survival, in vivo These results urge greater consideration of the roles played by oligodendrocyte dysfunction and altered membrane lipid metabolic flux as drivers of neurodegeneration in AD.SIGNIFICANCE STATEMENT Genetic, neuropathological, and functional studies implicate both Aβ and altered lipid metabolism and/or signaling as key pathogenic drivers of Alzheimer's disease. In this study, we first demonstrate that the enzyme SK2, which generates the signaling lipid S1P, is required for Aβ formation from APP in vivo Second, we establish a new role for SK2 in the protection of oligodendrocytes and myelin. Loss of SK2 sensitizes to Aβ-mediated neurodegeneration by attenuating oligodendrocyte survival and promoting hippocampal atrophy, despite reduced Aβ burden. Our findings support a model in which Aβ-independent sensitizing influences such as loss of neuroprotective S1P are more important drivers of neurodegeneration than gross Aβ concentration or plaque density.
Collapse
|
34
|
Bobinger T, Manaenko A, Burkardt P, Beuscher V, Sprügel MI, Roeder SS, Bäuerle T, Seyler L, Nagel AM, Linker RA, Engelhorn T, Dörfler A, Horsten SV, Schwab S, Huttner HB. Siponimod (BAF-312) Attenuates Perihemorrhagic Edema And Improves Survival in Experimental Intracerebral Hemorrhage. Stroke 2019; 50:3246-3254. [PMID: 31558140 DOI: 10.1161/strokeaha.119.027134] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background and Purpose- Perihemorrhagic edema (PHE) is associated with poor outcome after intracerebral hemorrhage (ICH). Infiltration of immune cells is considered a major contributor of PHE. Recent studies suggest that immunomodulation via S1PR (sphingosine-1-phosphate receptor) modulators improve outcome in ICH. Siponimod, a selective modulator of sphingosine 1-phosphate receptors type 1 and type 5, demonstrated an excellent safety profile in a large study of patients with multiple sclerosis. Here, we investigated the impact of siponimod treatment on perihemorrhagic edema, neurological deficits, and survival in a mouse model of ICH. Methods- ICH was induced by intracranial injection of 0.075 U of bacterial collagenase in 123 mice. Mice were randomly assigned to different treatment groups: vehicle, siponimod given as a single dosage 30 minutes after the operation or given 3× for 3 consecutive days starting 30 minutes after operation. The primary outcome of our study was evolution of PHE measured by magnetic resonance-imaging on T2-maps 72 hours after ICH, secondary outcomes included evolution of PHE 24 hours after ICH, survival and neurological deficits, as well as effects on circulating blood cells and body weight. Results- Siponimod significantly reduced PHE measured by magnetic resonance imaging (P=0.021) as well as wet-dry method (P=0.04) 72 hours after ICH. Evaluation of PHE 24 hours after ICH showed a tendency toward attenuated brain edema in the low-dosage group (P=0.08). Multiple treatments with siponimod significantly improved neurological deficits measured by Garcia Score (P=0.03). Survival at day 10 was improved in mice treated with multiple dosages of siponimod (P=0.037). Mice treated with siponimod showed a reduced weight loss after ICH (P=0.036). Conclusions- Siponimod (BAF-312) attenuated PHE after ICH, increased survival, and reduced ICH-induced sensorimotor deficits in our experimental ICH-model. Findings encourage further investigation of inflammatory modulators as well as the translation of BAF-312 to a human study of ICH patients.
Collapse
Affiliation(s)
- Tobias Bobinger
- From the Department of Neurology (T.B., A.M., P.B., V.B., M.I.S., S.S.R., R.A.L., S.S., H.B.H.), University of Erlangen, Germany
| | - Anatol Manaenko
- From the Department of Neurology (T.B., A.M., P.B., V.B., M.I.S., S.S.R., R.A.L., S.S., H.B.H.), University of Erlangen, Germany
| | - Petra Burkardt
- From the Department of Neurology (T.B., A.M., P.B., V.B., M.I.S., S.S.R., R.A.L., S.S., H.B.H.), University of Erlangen, Germany
| | - Vanessa Beuscher
- From the Department of Neurology (T.B., A.M., P.B., V.B., M.I.S., S.S.R., R.A.L., S.S., H.B.H.), University of Erlangen, Germany
| | - Maximilian I Sprügel
- From the Department of Neurology (T.B., A.M., P.B., V.B., M.I.S., S.S.R., R.A.L., S.S., H.B.H.), University of Erlangen, Germany
| | - Sebastian S Roeder
- From the Department of Neurology (T.B., A.M., P.B., V.B., M.I.S., S.S.R., R.A.L., S.S., H.B.H.), University of Erlangen, Germany
| | - Tobias Bäuerle
- Department of Radiology (T.B., L.S., A.M.N.), University of Erlangen, Germany
| | - Lisa Seyler
- Department of Radiology (T.B., L.S., A.M.N.), University of Erlangen, Germany
| | - Armin M Nagel
- Department of Radiology (T.B., L.S., A.M.N.), University of Erlangen, Germany
| | - Ralf A Linker
- From the Department of Neurology (T.B., A.M., P.B., V.B., M.I.S., S.S.R., R.A.L., S.S., H.B.H.), University of Erlangen, Germany.,Department of Neurology, University of Regensburg, Germany (R.A.L.)
| | - Tobias Engelhorn
- Department of Neuroradiology (T.E., A.D.), University of Erlangen, Germany
| | - Arnd Dörfler
- Department of Neuroradiology (T.E., A.D.), University of Erlangen, Germany
| | - S V Horsten
- Department of Experimental Therapy and Preclinical Center (S.v.H.), University of Erlangen, Germany
| | - Stefan Schwab
- From the Department of Neurology (T.B., A.M., P.B., V.B., M.I.S., S.S.R., R.A.L., S.S., H.B.H.), University of Erlangen, Germany
| | - Hagen B Huttner
- From the Department of Neurology (T.B., A.M., P.B., V.B., M.I.S., S.S.R., R.A.L., S.S., H.B.H.), University of Erlangen, Germany
| |
Collapse
|
35
|
Faissner S, Plemel JR, Gold R, Yong VW. Progressive multiple sclerosis: from pathophysiology to therapeutic strategies. Nat Rev Drug Discov 2019; 18:905-922. [PMID: 31399729 DOI: 10.1038/s41573-019-0035-2] [Citation(s) in RCA: 281] [Impact Index Per Article: 46.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/04/2019] [Indexed: 02/07/2023]
Abstract
Multiple sclerosis (MS) is a chronic inflammatory disease of the central nervous system that involves demyelination and axonal degeneration. Although substantial progress has been made in drug development for relapsing-remitting MS, treatment of the progressive forms of the disease, which are characterized clinically by the accumulation of disability in the absence of relapses, remains unsatisfactory. This unmet clinical need is related to the complexity of the pathophysiological mechanisms involved in MS progression. Chronic inflammation, which occurs behind a closed blood-brain barrier with activation of microglia and continued involvement of T cells and B cells, is a hallmark pathophysiological feature. Inflammation can enhance mitochondrial damage in neurons, which, consequently, develop an energy deficit, further reducing axonal health. The growth-inhibitory and inflammatory environment of lesions also impairs remyelination, a repair process that might protect axons from degeneration. Moreover, neurodegeneration is accelerated by the altered expression of ion channels on denuded axons. In this Review, we discuss the current understanding of these disease mechanisms and highlight emerging therapeutic strategies based on these insights, including those targeting the neuroinflammatory and degenerative aspects as well as remyelination-promoting approaches.
Collapse
Affiliation(s)
- Simon Faissner
- Department of Neurology, St. Josef-Hospital, Ruhr-University Bochum, Bochum, Germany. .,Hotchkiss Brain Institute and Department of Clinical Neurosciences, University of Calgary, Calgary, Alberta, Canada.
| | - Jason R Plemel
- Department of Medicine, University of Alberta, Edmonton, Alberta, Canada
| | - Ralf Gold
- Department of Neurology, St. Josef-Hospital, Ruhr-University Bochum, Bochum, Germany
| | - V Wee Yong
- Hotchkiss Brain Institute and Department of Clinical Neurosciences, University of Calgary, Calgary, Alberta, Canada.
| |
Collapse
|
36
|
Huang Y, Mao K, Germain RN. Thinking differently about ILCs-Not just tissue resident and not just the same as CD4 + T-cell effectors. Immunol Rev 2019; 286:160-171. [PMID: 30294968 DOI: 10.1111/imr.12704] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Accepted: 08/10/2018] [Indexed: 12/16/2022]
Abstract
Innate lymphoid cells (ILCs) resemble adaptive T lymphocytes based on transcription factor expression, cytokine production, and their presumptive roles in immunity, but are activated for effector function through cytokine signaling and not antigen-specific receptors. The prevailing view is that ILCs adapt to specific microenvironments during development and operate as tissue-resident cells in co-operation with antigen-specific T cells to provide host protection and contribute to tissue maintenance. In particular, conventional models equate the activity of different ILC subsets with CD4+ effector T-cell types based on corresponding transcription factor expression and a potential for comparable cytokine production. Based on recent data from our laboratory, we suggest that these views on tissue residence and parallel functioning to CD4+ T cells are too restrictive. Our findings show that ILC2s can be mobilized from the gut under inflammatory conditions and contribute to distal immunity in the lungs during infection, whereas gut-resident ILC3s operate in a quite distinct manner from Th17 CD4+ effector cells in responding to commensal microbes, with important implications for control of metabolic homeostasis. In this review, we discuss the recent advances leading to these revised views of ILC inter-organ trafficking and the distinct and complementary function of ILCs with respect to adaptive T cells in establishing and maintaining a physiologic host environment.
Collapse
Affiliation(s)
- Yuefeng Huang
- Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National institute of Health, Bethesda, Maryland.,Department of Microbiology & Immunology, College of Physicians & Surgeons, Columbia University, New York, New York
| | - Kairui Mao
- Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National institute of Health, Bethesda, Maryland
| | - Ronald N Germain
- Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National institute of Health, Bethesda, Maryland
| |
Collapse
|
37
|
Sphingosine-1-phosphate receptor 1 activation in astrocytes contributes to neuropathic pain. Proc Natl Acad Sci U S A 2019; 116:10557-10562. [PMID: 31068460 DOI: 10.1073/pnas.1820466116] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Neuropathic pain afflicts millions of individuals and represents a major health problem for which there is limited effective and safe therapy. Emerging literature links altered sphingolipid metabolism to nociceptive processing. However, the neuropharmacology of sphingolipid signaling in the central nervous system in the context of chronic pain remains largely unexplored and controversial. We now provide evidence that sphingosine-1-phosphate (S1P) generated in the dorsal horn of the spinal cord in response to nerve injury drives neuropathic pain by selectively activating the S1P receptor subtype 1 (S1PR1) in astrocytes. Accordingly, genetic and pharmacological inhibition of S1PR1 with multiple antagonists in distinct chemical classes, but not agonists, attenuated and even reversed neuropathic pain in rodents of both sexes and in two models of traumatic nerve injury. These S1PR1 antagonists retained their ability to inhibit neuropathic pain during sustained drug administration, and their effects were independent of endogenous opioid circuits. Moreover, mice with astrocyte-specific knockout of S1pr1 did not develop neuropathic pain following nerve injury, thereby identifying astrocytes as the primary cellular substrate of S1PR1 activity. On a molecular level, the beneficial reductions in neuropathic pain resulting from S1PR1 inhibition were driven by interleukin 10 (IL-10), a potent neuroprotective and anti-inflammatory cytokine. Collectively, our results provide fundamental neurobiological insights that identify the cellular and molecular mechanisms engaged by the S1PR1 axis in neuropathic pain and establish S1PR1 as a target for therapeutic intervention with S1PR1 antagonists as a class of nonnarcotic analgesics.
Collapse
|
38
|
Fischl AS, Wang X, Falcon BL, Almonte-Baldonado R, Bodenmiller D, Evans G, Stewart J, Wilson T, Hipskind P, Manro J, Uhlik MT, Chintharlapalli S, Gerald D, Alsop DC, Benjamin LE, Bhatt RS. Inhibition of Sphingosine Phosphate Receptor 1 Signaling Enhances the Efficacy of VEGF Receptor Inhibition. Mol Cancer Ther 2019; 18:856-867. [PMID: 30787172 DOI: 10.1158/1535-7163.mct-18-0548] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Revised: 10/04/2018] [Accepted: 02/04/2019] [Indexed: 01/15/2023]
Abstract
Inhibition of VEGFR signaling is an effective treatment for renal cell carcinoma, but resistance continues to be a major problem. Recently, the sphingosine phosphate (S1P) signaling pathway has been implicated in tumor growth, angiogenesis, and resistance to antiangiogenic therapy. S1P is a bioactive lipid that serves an essential role in developmental and pathologic angiogenesis via activation of the S1P receptor 1 (S1P1). S1P1 signaling counteracts VEGF signaling and is required for vascular stabilization. We used in vivo and in vitro angiogenesis models including a postnatal retinal angiogenesis model and a renal cell carcinoma murine tumor model to test whether simultaneous inhibition of S1P1 and VEGF leads to improved angiogenic inhibition. Here, we show that inhibition of S1P signaling reduces the endothelial cell barrier and leads to excessive angiogenic sprouting. Simultaneous inhibition of S1P and VEGF signaling further disrupts the tumor vascular beds, decreases tumor volume, and increases tumor cell death compared with monotherapies. These studies suggest that inhibition of angiogenesis at two stages of the multistep process may maximize the effects of antiangiogenic therapy. Together, these data suggest that combination of S1P1 and VEGFR-targeted therapy may be a useful therapeutic strategy for the treatment of renal cell carcinoma and other tumor types.
Collapse
MESH Headings
- Angiogenesis Inhibitors/pharmacology
- Animals
- Antibodies, Monoclonal/pharmacology
- Carcinoma, Renal Cell/blood supply
- Carcinoma, Renal Cell/drug therapy
- Carcinoma, Renal Cell/metabolism
- Carcinoma, Renal Cell/pathology
- Cell Line, Tumor
- Drug Therapy, Combination
- Endothelial Cells/drug effects
- Endothelial Cells/metabolism
- Female
- Humans
- Kidney Neoplasms/blood supply
- Kidney Neoplasms/drug therapy
- Kidney Neoplasms/metabolism
- Kidney Neoplasms/pathology
- Lysophospholipids/antagonists & inhibitors
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Nude
- Neovascularization, Pathologic/drug therapy
- Sphingosine/analogs & derivatives
- Sphingosine/antagonists & inhibitors
- Sphingosine-1-Phosphate Receptors/antagonists & inhibitors
- Sunitinib/pharmacology
- Treatment Outcome
- Tumor Burden/drug effects
- Vascular Endothelial Growth Factor A/pharmacology
- Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
| | - Xiaoen Wang
- Department of Radiology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | | | | | | | | | | | | | | | | | | | | | | | - David C Alsop
- Department of Radiology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | | | - Rupal S Bhatt
- Division of Hematology and Oncology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts.
| |
Collapse
|
39
|
Hundehege P, Cerina M, Eichler S, Thomas C, Herrmann AM, Göbel K, Müntefering T, Fernandez-Orth J, Bock S, Narayanan V, Budde T, Speckmann EJ, Wiendl H, Schubart A, Ruck T, Meuth SG. The next-generation sphingosine-1 receptor modulator BAF312 (siponimod) improves cortical network functionality in focal autoimmune encephalomyelitis. Neural Regen Res 2019; 14:1950-1960. [PMID: 31290453 PMCID: PMC6676873 DOI: 10.4103/1673-5374.259622] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Autoimmune diseases of the central nervous system (CNS) like multiple sclerosis (MS) are characterized by inflammation and demyelinated lesions in white and grey matter regions. While inflammation is present at all stages of MS, it is more pronounced in the relapsing forms of the disease, whereas progressive MS (PMS) shows significant neuroaxonal damage and grey and white matter atrophy. Hence, disease-modifying treatments beneficial in patients with relapsing MS have limited success in PMS. BAF312 (siponimod) is a novel sphingosine-1-phosphate receptor modulator shown to delay progression in PMS. Besides reducing inflammation by sequestering lymphocytes in lymphoid tissues, BAF312 crosses the blood-brain barrier and binds its receptors on neurons, astrocytes and oligodendrocytes. To evaluate potential direct neuroprotective effects, BAF312 was systemically or locally administered in the CNS of experimental autoimmune encephalomyelitis mice with distinct grey- and white-matter lesions (focal experimental autoimmune encephalomyelitis using an osmotic mini-pump). Ex-vivo flow cytometry revealed that systemic but not local BAF312 administration lowered immune cell infiltration in animals with both grey and white matter lesions. Ex-vivo voltage-sensitive dye imaging of acute brain slices revealed an altered spatio-temporal pattern of activation in the lesioned cortex compared to controls in response to electrical stimulation of incoming white-matter fiber tracts. Here, BAF312 administration showed partial restore of cortical neuronal circuit function. The data suggest that BAF312 exerts a neuroprotective effect after crossing the blood-brain barrier independently of peripheral effects on immune cells. Experiments were carried out in accordance with German and EU animal protection law and approved by local authorities (Landesamt für Natur, Umwelt und Verbraucherschutz Nordrhein-Westfalen; 87-51.04.2010.A331) on December 28, 2010.
Collapse
Affiliation(s)
- Petra Hundehege
- Department of Neurology with Institute of Translational Neurology, Westfälische Wilhelms-Universität, Münster, Germany
| | - Manuela Cerina
- Department of Neurology with Institute of Translational Neurology, Westfälische Wilhelms-Universität, Münster, Germany
| | - Susann Eichler
- Department of Neurology with Institute of Translational Neurology, Westfälische Wilhelms-Universität, Münster, Germany
| | - Christian Thomas
- Department of Neurology with Institute of Translational Neurology, Westfälische Wilhelms-Universität, Münster, Germany
| | - Alexander M Herrmann
- Department of Neurology with Institute of Translational Neurology, Westfälische Wilhelms-Universität, Münster, Germany
| | - Kerstin Göbel
- Department of Neurology with Institute of Translational Neurology, Westfälische Wilhelms-Universität, Münster, Germany
| | - Thomas Müntefering
- Department of Neurology with Institute of Translational Neurology, Westfälische Wilhelms-Universität, Münster, Germany
| | - Juncal Fernandez-Orth
- Department of Neurology with Institute of Translational Neurology, Westfälische Wilhelms-Universität, Münster, Germany
| | - Stefanie Bock
- Department of Neurology with Institute of Translational Neurology, Westfälische Wilhelms-Universität, Münster, Germany
| | - Venu Narayanan
- Department of Neurology with Institute of Translational Neurology, Westfälische Wilhelms-Universität, Münster, Germany
| | - Thomas Budde
- Institute of Physiology I, Westfälische Wilhelms-Universität, Münster, Germany
| | | | - Heinz Wiendl
- Department of Neurology with Institute of Translational Neurology, Westfälische Wilhelms-Universität, Münster, Germany
| | - Anna Schubart
- Novartis Institutes of Biomedical Research, Basel, Switzerland
| | - Tobias Ruck
- Department of Neurology with Institute of Translational Neurology, Westfälische Wilhelms-Universität, Münster, Germany
| | - Sven G Meuth
- Department of Neurology with Institute of Translational Neurology, Westfälische Wilhelms-Universität, Münster, Germany
| |
Collapse
|
40
|
Kifuji T, Inoue S, Furukawa M, Perez Madera B, Goto T, Kumagai H, Mair SJ, Kawaguchi A. Absorption, disposition and metabolic pathway of amiselimod (MT-1303) in healthy volunteers in a mass balance study. Xenobiotica 2018; 49:1033-1043. [PMID: 30231665 DOI: 10.1080/00498254.2018.1525508] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Abstract
The absorption, metabolism and excretion of MT-1303 were investigated in healthy male subjects after a single oral dose of 0.4 mg [14C]-MT-1303 (ClinicalTrials.gov NCT02293967). The MT-1303 concentration in the plasma reached a maximum at 12 h after administration. Thereafter, the concentration declined with a half-life of 451 h. At the final assessment on Day 57, 91.16% of the administered radioactivity was excreted, and the cumulative excretion in the urine and faeces was 35.32% and 55.84%, respectively. The most abundant metabolite in plasma was MT-1303-P, which accounted for 42.6% of the area under the plasma concentration-time curve (AUC) of the total radioactivity. The major component excreted in urine was Human Urine (HU)4 (3066434), accounting for 28.1% of radioactivity in the sample (4.05% of the dose), whereas MT-1303 was a major component in the faeces, accounting for 89.8% of radioactivity in the sample (25.49% of the dose) up to 240 h after administration. This study indicates that multiple metabolic pathways are involved in the elimination of MT-1303 from the human body and the excretion of MT-1303 and MT-1303-P via the kidney is low. Therefore, MT-1303 is unlikely to cause conspicuous drug interactions or alter pharmacokinetics in patients with renal impairment.
Collapse
Affiliation(s)
- Takayuki Kifuji
- a Data Science Department, Ikuyaku. Integrated Value Development Division , Mitsubishi Tanabe Pharma Corporation , Chuo-ku , Tokyo , Japan
| | - Shinsuke Inoue
- a Data Science Department, Ikuyaku. Integrated Value Development Division , Mitsubishi Tanabe Pharma Corporation , Chuo-ku , Tokyo , Japan
| | - Megumi Furukawa
- a Data Science Department, Ikuyaku. Integrated Value Development Division , Mitsubishi Tanabe Pharma Corporation , Chuo-ku , Tokyo , Japan
| | | | - Takahiro Goto
- c DMPK Research Laboratories, Sohyaku. Innovative Research Division , Mitsubishi Tanabe Pharma Corporation , Yokohama , Kanagawa , Japan
| | - Hiroshi Kumagai
- c DMPK Research Laboratories, Sohyaku. Innovative Research Division , Mitsubishi Tanabe Pharma Corporation , Yokohama , Kanagawa , Japan
| | | | - Atsuhiro Kawaguchi
- a Data Science Department, Ikuyaku. Integrated Value Development Division , Mitsubishi Tanabe Pharma Corporation , Chuo-ku , Tokyo , Japan
| |
Collapse
|
41
|
Zheng YH, Ma YY, Ding Y, Chen XQ, Gao GX. An insight into new strategies to combat antifungal drug resistance. DRUG DESIGN DEVELOPMENT AND THERAPY 2018; 12:3807-3816. [PMID: 30464412 PMCID: PMC6225914 DOI: 10.2147/dddt.s185833] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Invasive fungal infections especially in immunocompromised patients represent a dominating cause of mortality. The most commonly used antifungal agents can be divided into three broad categories, including triazoles, echinocandins and polyenes. Antifungal resistance is on the increase, posing a growing threat to the stewardship of immunocompromised patients with fungal infections. The paucity of currently available antifungals leads to the rapid emergence of drug resistance and thus aggravates the refractoriness of invasive fungal infections. Therefore, deep exploration into mechanisms of drug resistance and search for new antifungal targets are required. This review highlights the therapeutic strategies targeting Hsp90, calcineurin, trehalose biosynthesis and sphingolipids biosynthesis, in an attempt to provide clinical evidence for overcoming drug resistance and to form the rationale for combination therapy of conventional antifungals and agents with novel mechanisms of action. What’s more, this review also gives a concise introduction of three new-fashioned antifungals, including carboxymethyl chitosan, silver nanoparticles and chromogranin A-N46.
Collapse
Affiliation(s)
- Yan-Hua Zheng
- Department of Hematology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China, ;
| | - Yue-Yun Ma
- Department of Clinical Laboratory Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Yi Ding
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Xie-Qun Chen
- Department of Hematology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China, ;
| | - Guang-Xun Gao
- Department of Hematology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China, ;
| |
Collapse
|
42
|
A selective inhibitor of ceramide synthase 1 reveals a novel role in fat metabolism. Nat Commun 2018; 9:3165. [PMID: 30131496 PMCID: PMC6104039 DOI: 10.1038/s41467-018-05613-7] [Citation(s) in RCA: 95] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Accepted: 07/17/2018] [Indexed: 02/06/2023] Open
Abstract
Specific forms of the lipid ceramide, synthesized by the ceramide synthase enzyme family, are believed to regulate metabolic physiology. Genetic mouse models have established C16 ceramide as a driver of insulin resistance in liver and adipose tissue. C18 ceramide, synthesized by ceramide synthase 1 (CerS1), is abundant in skeletal muscle and suggested to promote insulin resistance in humans. We herein describe the first isoform-specific ceramide synthase inhibitor, P053, which inhibits CerS1 with nanomolar potency. Lipidomic profiling shows that P053 is highly selective for CerS1. Daily P053 administration to mice fed a high-fat diet (HFD) increases fatty acid oxidation in skeletal muscle and impedes increases in muscle triglycerides and adiposity, but does not protect against HFD-induced insulin resistance. Our inhibitor therefore allowed us to define a role for CerS1 as an endogenous inhibitor of mitochondrial fatty acid oxidation in muscle and regulator of whole-body adiposity. Ceramides are signalling molecules that regulate several physiological functions including insulin sensitivity. Here the authors report a selective ceramide synthase 1 inhibitor that counteracts lipid accumulation within the muscle and adiposity by increasing fatty acid oxidation but without affecting insulin sensitivity in mice fed with an obesogenic diet.
Collapse
|
43
|
Yuan L, Basdeo S, Ji QC. Overcoming the stability, solubility and extraction challenges in reversed-phase UHPLC–MS/MS bioanalysis of a phosphate drug and its prodrug in blood lysate. J Pharm Biomed Anal 2018; 157:36-43. [DOI: 10.1016/j.jpba.2018.05.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Revised: 05/03/2018] [Accepted: 05/04/2018] [Indexed: 01/17/2023]
|
44
|
Stockstill K, Doyle TM, Yan X, Chen Z, Janes K, Little JW, Braden K, Lauro F, Giancotti LA, Harada CM, Yadav R, Xiao WH, Lionberger JM, Neumann WL, Bennett GJ, Weng HR, Spiegel S, Salvemini D. Dysregulation of sphingolipid metabolism contributes to bortezomib-induced neuropathic pain. J Exp Med 2018; 215:1301-1313. [PMID: 29703731 PMCID: PMC5940258 DOI: 10.1084/jem.20170584] [Citation(s) in RCA: 86] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Revised: 07/31/2017] [Accepted: 03/21/2018] [Indexed: 11/04/2022] Open
Abstract
The development of chemotherapy-induced painful peripheral neuropathy is a major dose-limiting side effect of many chemotherapeutics, including bortezomib, but the mechanisms remain poorly understood. We now report that bortezomib causes the dysregulation of de novo sphingolipid metabolism in the spinal cord dorsal horn to increase the levels of sphingosine-1-phosphate (S1P) receptor 1 (S1PR1) ligands, S1P and dihydro-S1P. Accordingly, genetic and pharmacological disruption of S1PR1 with multiple S1PR1 antagonists, including FTY720, blocked and reversed neuropathic pain. Mice with astrocyte-specific alterations of S1pr1 did not develop neuropathic pain and lost their ability to respond to S1PR1 inhibition, strongly implicating astrocytes as a primary cellular substrate for S1PR1 activity. At the molecular level, S1PR1 engaged astrocyte-driven neuroinflammation and altered glutamatergic homeostasis, processes blocked by S1PR1 antagonism. Our findings establish S1PR1 as a target for therapeutic intervention and provide insight into cellular and molecular pathways. As FTY720 also shows promising anticancer potential and is FDA approved, rapid clinical translation of our findings is anticipated.
Collapse
Affiliation(s)
- Katherine Stockstill
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, St. Louis, MO
| | - Timothy M Doyle
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, St. Louis, MO
| | - Xisheng Yan
- Department of Pharmaceutical and Biomedical Sciences, College of Pharmacy, University of Georgia, Athens, GA
| | - Zhoumou Chen
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, St. Louis, MO
| | - Kali Janes
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, St. Louis, MO
| | - Joshua W Little
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, St. Louis, MO
- Department of Surgery, Center for Anatomical Science and Education, Saint Louis University School of Medicine, St. Louis, MO
| | - Kathryn Braden
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, St. Louis, MO
| | - Filomena Lauro
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, St. Louis, MO
| | | | - Caron Mitsue Harada
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, St. Louis, MO
| | - Ruchi Yadav
- Department of Pharmaceutical and Biomedical Sciences, College of Pharmacy, University of Georgia, Athens, GA
| | - Wen Hua Xiao
- Department of Anesthesiology, University of California, San Diego, La Jolla, CA
| | - Jack M Lionberger
- Department of Internal Medicine, Division of Hematology, Oncology, and Cellular Therapeutics, Saint Louis University School of Medicine, St. Louis, MO
| | - William L Neumann
- Department of Pharmaceutical Sciences, School of Pharmacy, Southern Illinois University Edwardsville, Edwardsville, IL
| | - Gary J Bennett
- Department of Anesthesiology, University of California, San Diego, La Jolla, CA
| | - Han-Rong Weng
- Department of Pharmaceutical and Biomedical Sciences, College of Pharmacy, University of Georgia, Athens, GA
| | - Sarah Spiegel
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, School of Medicine, Richmond, VA
| | - Daniela Salvemini
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, St. Louis, MO
| |
Collapse
|
45
|
Abstract
Metastatic bone pain is the single most common form of cancer pain and persists as a result of peripheral and central inflammatory, as well as neuropathic mechanisms. Here, we provide the first characterization of sphingolipid metabolism alterations in the spinal cord occurring during cancer-induced bone pain (CIBP). Following femoral arthrotomy and syngenic tumor implantation in mice, ceramides decreased with corresponding increases in sphingosine and the bioactive sphingolipid metabolite, sphingosine 1-phosphate (S1P). Intriguingly, de novo sphingolipid biosynthesis was increased as shown by the elevations of dihydro-ceramides and dihydro-S1P. We next identified the S1P receptor subtype 1 (S1PR1) as a novel target for therapeutic intervention. Intrathecal or systemic administration of the competitive and functional S1PR1 antagonists, TASP0277308 and FTY720/Fingolimod, respectively, attenuated cancer-induced spontaneous flinching and guarding. Inhibiting CIBP by systemic delivery of FTY720 did not result in antinociceptive tolerance over 7 days. FTY720 administration enhanced IL-10 in the lumbar ipsilateral spinal cord of CIBP animals and intrathecal injection of an IL-10 neutralizing antibody mitigated the ability of systemic FTY720 to reverse CIBP. FTY720 treatment was not associated with alterations in bone metabolism in vivo. Studies here identify a novel mechanism to inhibit bone cancer pain by blocking the actions of the bioactive metabolites S1P and dihydro-S1P in lumbar spinal cord induced by bone cancer and support potential fast-track clinical application of the FDA-approved drug, FTY720, as a therapeutic avenue for CIBP.
Collapse
|
46
|
Cuzzocrea S, Doyle T, Campolo M, Paterniti I, Esposito E, Farr SA, Salvemini D. Sphingosine 1-Phosphate Receptor Subtype 1 as a Therapeutic Target for Brain Trauma. J Neurotrauma 2018; 35:1452-1466. [PMID: 29310513 DOI: 10.1089/neu.2017.5391] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Traumatic brain injury (TBI) provokes secondary pathological mechanisms, including ischemic and inflammatory processes. The new research in sphingosine 1-phosphate (S1P) receptor modulators has opened the door for an effective mechanism of reducing central nervous system (CNS) inflammatory lesion activity. Thus, the aim of this study was to characterize the immunomodulatory effect of the functional S1PR1 antagonist, siponimod, in phase III clinical trials for autoimmune disorders and of the competitive sphingosine 1-phosphate receptor subtype 1 (S1PR1) antagonist, TASP0277308, in pre-clinical development in an in vivo model of TBI in mice. We used the well-characterized model of TBI caused by controlled cortical impact. Mice were injected intraperitoneally with siponimod or TASP0277308 (1 mg/kg) at 1 and 4 h post-trauma. Our results demonstrated that these agents exerted significant beneficial effects on TBI pre-clinical scores in term of anti-inflammatory and immunomodulatory effects, in particular, attenuation of astrocytes and microglia activation, cytokines release, and rescue of the reduction of adhesion molecules (i.e., occludin and zonula occludens-1). Moreover, these compounds were able to decrease T-cell activation visible by reduction of CD4+ and CD8+, reduce the lesioned area (measured by 2,3,5-triphenyltetrazolium chloride staining), and to preserve tissue architecture, microtubule stability, and neural plasticity. Moreover, our findings provide pre-clinical evidence for the use of low-dose oral S1PR1 antagonists as neuroprotective strategies for TBI and broaden our understanding of the underlying S1PR1-driven neuroinflammatory processes in the pathophysiology of TBI. Altogether, our results showed that blocking the S1PR1 axis is an effective therapeutic strategy to mitigate neuropathological effects engaged in the CNS by TBI.
Collapse
Affiliation(s)
- Salvatore Cuzzocrea
- 1 Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina , Viale Ferdinando Stagno D'Alcontres, Messina, Italy .,2 Department of Pharmacology and Physiology Saint Louis University , St. Louis, Missouri
| | - Timothy Doyle
- 2 Department of Pharmacology and Physiology Saint Louis University , St. Louis, Missouri
| | - Michela Campolo
- 1 Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina , Viale Ferdinando Stagno D'Alcontres, Messina, Italy
| | - Irene Paterniti
- 1 Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina , Viale Ferdinando Stagno D'Alcontres, Messina, Italy
| | - Emanuela Esposito
- 1 Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina , Viale Ferdinando Stagno D'Alcontres, Messina, Italy
| | - Susan A Farr
- 3 VA Medical Center Saint Louis , St. Louis, Missouri.,4 Division of Geriatric Medicine, Saint Louis University , St. Louis, Missouri
| | - Daniela Salvemini
- 2 Department of Pharmacology and Physiology Saint Louis University , St. Louis, Missouri
| |
Collapse
|
47
|
Lidington D, Kroetsch JT, Bolz SS. Cerebral artery myogenic reactivity: The next frontier in developing effective interventions for subarachnoid hemorrhage. J Cereb Blood Flow Metab 2018; 38:17-37. [PMID: 29135346 PMCID: PMC5757446 DOI: 10.1177/0271678x17742548] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Revised: 10/24/2017] [Accepted: 10/25/2017] [Indexed: 12/26/2022]
Abstract
Aneurysmal subarachnoid hemorrhage (SAH) is a devastating cerebral event that kills or debilitates the majority of those afflicted. The blood that spills into the subarachnoid space stimulates profound cerebral artery vasoconstriction and consequently, cerebral ischemia. Thus, once the initial bleeding in SAH is appropriately managed, the clinical focus shifts to maintaining/improving cerebral perfusion. However, current therapeutic interventions largely fail to improve clinical outcome, because they do not effectively restore normal cerebral artery function. This review discusses emerging evidence that perturbed cerebrovascular "myogenic reactivity," a crucial microvascular process that potently dictates cerebral perfusion, is the critical element underlying cerebral ischemia in SAH. In fact, the myogenic mechanism could be the reason why many therapeutic interventions, including "Triple H" therapy, fail to deliver benefit to patients. Understanding the molecular basis for myogenic reactivity changes in SAH holds the key to develop more effective therapeutic interventions; indeed, promising recent advancements fuel optimism that vascular dysfunction in SAH can be corrected to improve outcome.
Collapse
Affiliation(s)
- Darcy Lidington
- Department of Physiology, University of Toronto, Toronto, Canada
- Toronto Centre for Microvascular Medicine at TBEP, University of Toronto, Toronto, Canada
| | - Jeffrey T Kroetsch
- Department of Physiology, University of Toronto, Toronto, Canada
- Toronto Centre for Microvascular Medicine at TBEP, University of Toronto, Toronto, Canada
| | - Steffen-Sebastian Bolz
- Department of Physiology, University of Toronto, Toronto, Canada
- Toronto Centre for Microvascular Medicine at TBEP, University of Toronto, Toronto, Canada
- Heart & Stroke/Richard Lewar Centre of Excellence for Cardiovascular Research, University of Toronto, Toronto, Canada
| |
Collapse
|
48
|
Pharmacokinetics, Pharmacodynamics, Tolerability, and Food Effect of Cenerimod, a Selective S1P₁ Receptor Modulator in Healthy Subjects. Int J Mol Sci 2017; 18:ijms18122636. [PMID: 29211013 PMCID: PMC5751239 DOI: 10.3390/ijms18122636] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Revised: 11/28/2017] [Accepted: 12/01/2017] [Indexed: 11/26/2022] Open
Abstract
The pharmacokinetics, pharmacodynamics, tolerability, and food effect of cenerimod, a potent sphingosine-1-phosphate subtype 1 receptor modulator, were investigated in three sub-studies. Two double-blind, placebo-controlled, randomised studies in healthy male subjects were performed. Cenerimod was administered either as single dose (1, 3, 10 or 25 mg; Study 1) or once daily for 35 days (0.5, 1, 2 or 4 mg; Study 2). A two-period cross-over, open-label study was performed to assess the food effect (1 mg, Study 3). The pharmacokinetic profile of cenerimod was characterised by a tmax of 5.0–6.2 h. Terminal half-life after single and multiple doses ranged from 170 to 199 h and 283 to 539 h, respectively. Food had no relevant effect on the pharmacokinetics of cenerimod. A dose-dependent decrease in lymphocyte count was observed after initiation of cenerimod and reached a plateau (maximum change from baseline: −64%) after 20–23 days of treatment. Lymphocyte counts returned to baseline values at end-of-study examination. One serious adverse event of circulatory collapse (25 mg dose group, maximum tolerated dose: 10 mg) and adverse events of mild-to-moderate intensity were reported. Treatment initiation was associated with transient decreases in heart rate and blood pressure at doses >1 and ≥10 mg, respectively.
Collapse
|
49
|
Sphingosine 1-phosphate (S1P) signalling: Role in bone biology and potential therapeutic target for bone repair. Pharmacol Res 2017; 125:232-245. [PMID: 28855094 DOI: 10.1016/j.phrs.2017.08.013] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Revised: 08/22/2017] [Accepted: 08/23/2017] [Indexed: 12/30/2022]
Abstract
The lipid mediator sphingosine 1-phosphate (S1P) affects cellular functions in most systems. Interest in its therapeutic potential has increased following the discovery of its G protein-coupled receptors and the recent availability of agents that can be safely administered in humans. Although the role of S1P in bone biology has been the focus of much less research than its role in the nervous, cardiovascular and immune systems, it is becoming clear that this lipid influences many of the functions, pathways and cell types that play a key role in bone maintenance and repair. Indeed, S1P is implicated in many osteogenesis-related processes including stem cell recruitment and subsequent differentiation, differentiation and survival of osteoblasts, and coupling of the latter cell type with osteoclasts. In addition, S1P's role in promoting angiogenesis is well-established. The pleiotropic effects of S1P on bone and blood vessels have significant potential therapeutic implications, as current therapeutic approaches for critical bone defects show significant limitations. Because of the complex effects of S1P on bone, the pharmacology of S1P-like agents and their physico-chemical properties, it is likely that therapeutic delivery of S1P agents will offer significant advantages compared to larger molecular weight factors. Hence, it is important to explore novel methods of utilizing S1P agents therapeutically, and improve our understanding of how S1P and its receptors modulate bone physiology and repair.
Collapse
|
50
|
Meshcheryakova A, Mechtcheriakova D, Pietschmann P. Sphingosine 1-phosphate signaling in bone remodeling: multifaceted roles and therapeutic potential. Expert Opin Ther Targets 2017; 21:725-737. [PMID: 28524744 PMCID: PMC5470107 DOI: 10.1080/14728222.2017.1332180] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Introduction: Sphingolipids belong to a complex class of lipid molecules that are crucially involved in the regulation of important biological processes including proliferation, migration and apoptosis. Given the significant progress made in understanding the sphingolipid pathobiology of several diseases, sphingolipid-related checkpoints emerge as attractive targets. Recent data indicate the multifaceted contribution of the sphingolipid machinery to osteoclast – osteoblast crosstalk, representing one of the pivotal interactions underlying bone homeostasis. Imbalances in the interplay of osteoblasts and osteoclasts might lead to bone-related diseases such as osteoporosis, rheumatoid arthritis, and bone metastases. Areas covered: We summarize and analyze the progress made in bone research in the context of the current knowledge of sphingolipid-related mechanisms regulating bone remodeling. Particular emphasis was given to bioactive sphingosine 1-phosphate (S1P) and S1P receptors (S1PRs). Moreover, the mechanisms of how dysregulations of this machinery cause bone diseases, are covered. Expert opinion: In the context of bone diseases, pharmacological interference with sphingolipid machinery may lead to novel directions in therapeutic strategies. Implementation of knowledge derived from in vivo animal models and in vitro studies using pharmacological agents to manipulate the S1P/S1PRs axes suggests S1PR2 and S1PR3 as potential drug targets, particularly in conjunction with technology for local drug delivery.
Collapse
Affiliation(s)
- Anastasia Meshcheryakova
- a Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology , Medical University of Vienna , Vienna , Austria
| | - Diana Mechtcheriakova
- a Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology , Medical University of Vienna , Vienna , Austria
| | - Peter Pietschmann
- a Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology , Medical University of Vienna , Vienna , Austria
| |
Collapse
|