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
|
Kashikar R, Senapati S, Dudhipala N, Basu SK, Mandal N, Majumdar S. Ophthalmic Nanoemulsion Fingolimod Formulation for Topical Application. J Ocul Pharmacol Ther 2024; 40:504-512. [PMID: 38976488 DOI: 10.1089/jop.2024.0055] [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] [Indexed: 07/10/2024] Open
Abstract
Purpose: Fingolimod (FTY720; FT), a structural analog of sphingosine, has potential ocular applications. The goal of this study was to develop an FT-loaded nanoemulsion (NE; FT-NE) formulation for the efficient and prolonged delivery of FT to the posterior segment of the eye through the topical route. Methods: FT-NE formulations were prepared using homogenization followed by the probe sonication method. The lead FT-NE formulations (0.15% and 0.3% w/v loading), comprising soybean oil as oil and Tween® 80 and Poloxamer 188 as surfactants, were further evaluated for in vitro release, surface morphology, filtration sterilization, and stability at refrigerated temperature. Ocular bioavailability following topical application of FT-NE (0.3%) was examined in Sprague-Dawley rats. Results: The formulation, at both dose levels, showed desirable physicochemical characteristics, a nearly spherical shape with homogenous nanometric size distribution, and was stable for 180 days (last time point checked) at refrigerated temperature postfiltration through a polyethersulfone (0.22 µm) membrane. In vitro release studies showed prolonged release over 24 h, compared with the control FT solution (FT-S). In vivo studies revealed that effective concentrations of FT were achieved in the vitreous humor and retina following topical application of FT-NE. Conclusions: The results from these studies demonstrate that the FT-NE formulation can serve as a viable platform for the ocular delivery of FT through the topical route.
Collapse
Affiliation(s)
- Rama Kashikar
- Department of Pharmaceutics and Drug Delivery, University of Mississippi, Mississippi, USA
| | - Samir Senapati
- Department of Pharmaceutics and Drug Delivery, University of Mississippi, Mississippi, USA
| | - Narendar Dudhipala
- Department of Pharmaceutics and Drug Delivery, University of Mississippi, Mississippi, USA
| | - Sandip K Basu
- Department of Ophthalmology, The University of Tennessee Health Science Center, Hamilton Eye Institute, Memphis, Tennessee, USA
| | - Nawajes Mandal
- Department of Ophthalmology, The University of Tennessee Health Science Center, Hamilton Eye Institute, Memphis, Tennessee, USA
- Department of Anatomy and Neurobiology, The University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Soumyajit Majumdar
- Department of Pharmaceutics and Drug Delivery, University of Mississippi, Mississippi, USA
- Research Institute of Pharmaceutical Sciences, University of Mississippi, Mississippi, USA
| |
Collapse
|
3
|
Qian Z, Zheng K, Xu Y, Chen S, Chen S, Liang J, Cao Y, Ng TK, Qiu K. Longitudinal in vivo evaluation of retinal ganglion cell complex layer and dendrites in mice with experimental autoimmune encephalomyelitis. Exp Eye Res 2023; 237:109708. [PMID: 37913917 DOI: 10.1016/j.exer.2023.109708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 10/24/2023] [Accepted: 10/28/2023] [Indexed: 11/03/2023]
Abstract
Experimental autoimmune encephalomyelitis (EAE), induced by the immunization of myelin oligodendrocyte glycoprotein (MOG), is related to human MOG antibody-associated disease (MOGAD). Neuroinflammation and demyelination of the optic nerve can lead to retinal ganglion cell (RGC) death and axonal damage in MOGAD. Here, we aimed to evaluate the structural changes in RGCs longitudinally by in vivo imaging in mice with RGCs expressing yellow fluorescent protein along the course of EAE. Successful induction of EAE was confirmed by the neurological function scores and histology analyses. The changes in the thickness of ganglion cell complex (GCC) layer and RGC survival and dendrites were monitored longitudinally along the course of EAE. Before the onset of EAE, there were no significant changes in the number and morphology of RGCs and the thickness of the GCC layer as compared to the mice without EAE induction. After the onset of EAE, the thickness of the GCC layer and the RGC number and dendritic network all gradually decreased along the course of EAE. Notably, dendritic shrinkage could be detected earlier than the thinning of the GCC layer. In summary, this study delineated the longitudinal profile of RGC structural changes in EAE mice, providing an assessment platform for monitoring outcomes of RGC treatments.
Collapse
Affiliation(s)
- Zhen Qian
- Joint Shantou International Eye Center of Shantou University and the Chinese University of Hong Kong, Shantou, Guangdong, China; Shantou University Medical College, Shantou, Guangdong, China
| | - Ke Zheng
- Joint Shantou International Eye Center of Shantou University and the Chinese University of Hong Kong, Shantou, Guangdong, China; Shantou University Medical College, Shantou, Guangdong, China
| | - Yanxuan Xu
- Joint Shantou International Eye Center of Shantou University and the Chinese University of Hong Kong, Shantou, Guangdong, China
| | - Si Chen
- Joint Shantou International Eye Center of Shantou University and the Chinese University of Hong Kong, Shantou, Guangdong, China
| | - Shaowan Chen
- Joint Shantou International Eye Center of Shantou University and the Chinese University of Hong Kong, Shantou, Guangdong, China
| | - Jiajian Liang
- Joint Shantou International Eye Center of Shantou University and the Chinese University of Hong Kong, Shantou, Guangdong, China
| | - Yingjie Cao
- Joint Shantou International Eye Center of Shantou University and the Chinese University of Hong Kong, Shantou, Guangdong, China
| | - Tsz Kin Ng
- Joint Shantou International Eye Center of Shantou University and the Chinese University of Hong Kong, Shantou, Guangdong, China; Department of Ophthalmology and Visual Sciences, The Chinese University of Hong Kong, Hong Kong.
| | - Kunliang Qiu
- Joint Shantou International Eye Center of Shantou University and the Chinese University of Hong Kong, Shantou, Guangdong, China.
| |
Collapse
|
4
|
Fan H, Yang Y, Bai Q, Wang D, Shi X, Zhang L, Yang Y. Neuroprotective Effects of Sinomenine on Experimental Autoimmune Encephalomyelitis via Anti-Inflammatory and Nrf2-Dependent Anti-Oxidative Stress Activity. Neuromolecular Med 2023; 25:545-562. [PMID: 37735290 DOI: 10.1007/s12017-023-08756-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Accepted: 08/24/2023] [Indexed: 09/23/2023]
Abstract
Multiple sclerosis (MS) is an autoimmune inflammatory disease of the central nervous system (CNS). Sinomenine (SIN), a bioactive alkaloid extracted from the Chinese medicinal plant Sinomenium acutum, has powerful anti-inflammatory and immunosuppressive therapeutic benefits. In our previous research, we found that SIN increased resistance to oxidative stress via the nuclear factor erythroid 2-related factor 2 (Nrf2) signaling pathway in PC12 neuronal cells. However, whether SIN can improve the symptoms and pathological features of experimental autoimmune encephalomyelitis (EAE), a murine model of MS, via the Nrf2 signaling pathway remains unclear. EAE was immunized followed by SIN treatment. Then we evaluated the effects of SIN in EAE. Subsequently, primary microglia were cultured to explore the effect of SIN on microglia activation. Further, the levels of Nrf2 and its downstream molecules were detected to assess the molecular mechanisms of SIN. We demonstrated that SIN effectively ameliorated the severity of EAE, accompanied by a reduction in the demyelination, axonal damage and inhibition of inflammatory cell infiltration. Mechanistically, SIN decreased the inflammatory cytokines expression, and suppressed microglia and astrocytes activation in EAE mice. Furthermore, SIN suppressed lipopolysaccharide (LPS)-induced microglial activation and the production of pro-inflammatory factors in vitro. Moreover, SIN inhibited oxidative stress via the activation of the Nrf2 signaling pathway. Our work proves that SIN exerts its neuroprotective effects by the Nrf2-dependent anti-oxidative stress and diminishing neuroinflammation, suggesting that the "antioxiflammation" effect of SIN is expected to be an ideal treatment strategy for MS/EAE.
Collapse
Affiliation(s)
- Hua Fan
- Office of Research & Innovation, The First Affiliated Hospital, College of Clinical Medicine, Henan University of Science and Technology, Luoyang, 471003, China.
| | - Yang Yang
- Office of Research & Innovation, The First Affiliated Hospital, College of Clinical Medicine, Henan University of Science and Technology, Luoyang, 471003, China
| | - Qianqian Bai
- Office of Research & Innovation, The First Affiliated Hospital, College of Clinical Medicine, Henan University of Science and Technology, Luoyang, 471003, China
| | - Dongmei Wang
- School of Basic Medical Sciences, Henan University of Science and Technology, Luoyang, 471003, China
| | - Xiaofei Shi
- Department of Rheumatology and Immunology, The First Affiliated Hospital, College of Clinical Medicine, Henan University of Science and Technology, Luoyang, 471003, China
| | - Lele Zhang
- Department of traditional Chinese medicine, The First Affiliated Hospital, College of Clinical Medicine, Henan University of Science and Technology, Luoyang, 471003, China
| | - Yanhui Yang
- Department of Trauma center, The First Affiliated Hospital, College of Clinical Medicine , Henan University of Science and Technology, Luoyang, 471003, China.
| |
Collapse
|
5
|
Feng Y, Feng F, Pan S, Zhang J, Li W. Fingolimod ameliorates chronic experimental autoimmune neuritis by modulating inflammatory cytokines and Akt/mTOR/NF-κB signaling. Brain Behav 2023; 13:e2965. [PMID: 36917739 PMCID: PMC10097075 DOI: 10.1002/brb3.2965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 02/14/2023] [Accepted: 02/17/2023] [Indexed: 03/16/2023] Open
Abstract
OBJECTIVE Chronic inflammatory demyelinating polyradiculoneuropathy (CIDP) is an immune-mediated disease that targets the myelin sheaths of the peripheral nerves. Fingolimod is a sphingosine 1 phosphate (S1P) receptor antagonist with a high affinity for S1P receptors through the Akt-mTOR pathway, and prior research has suggested that it might be helpful in autoimmune illnesses. METHODS Chronic experimental autoimmune neuritis (c-EAN) was induced by immunizing Lewis rats with the S-palm P0(180-199) peptide, and then the treatment group was intraperitoneally injected with fingolimod (1 mg/kg) daily. Hematoxylin and eosin staining was used to assess the severity of nerve injury. Immunohistochemistry staining showed that fingolimod's anti-inflammatory effects on c-EAN rats might be realized through the NF-κB signaling pathway. Tumor necrosis factor-α (TNF-α), interferon-γ (INF-γ), interleukin-1beta (IL-1β), interleukin 6 (IL-6), inducible nitric oxide synthase (iNOS), and intercellular adhesion molecule-1 (ICAM-1) were measured to evaluate the inflammation levels, and pAkt, p-S6, and p-p65 were used to measure the abundance of downstream activation markers to determine whether the Akt/mTOR/NF-κB signaling pathway was activated in the c-EAN model. RESULTS Fingolimod treatment reduced the inflammatory reaction and the expression of NF-κB in sciatic nerves. It also decreased the mRNA levels of the proinflammatory cytokines TNF-α, IFN-γ, IL-1β, IL-6, iNOS, and ICAM-1 and pAkt, p-S6, and p-p65, representing the Akt/mTOR/NF-κB signaling pathway. CONCLUSION Our data showed that fingolimod could improve the disease course, alleviate the decrease in inflammation, and reduce proinflammatory cytokines through the Akt/mTOR/NF-κB axis in c-EAN rats, which could be beneficial for the development of CIDP-related research.
Collapse
Affiliation(s)
- Yuan Feng
- Department of Neurology, Henan Provincial People's Hospital, Zhengzhou, China.,Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Fang Feng
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Shuyi Pan
- Department of Hyperbaric Medicine, 6th Medical Center of PLA General Hospital, Beijing, China
| | - Jiewen Zhang
- Department of Neurology, Henan Provincial People's Hospital, Zhengzhou, China
| | - Wei Li
- Department of Neurology, Henan Provincial People's Hospital, Zhengzhou, China
| |
Collapse
|
6
|
Jeong YE, Rajbhandari L, Kim BW, Venkatesan A, Hoke A. Downregulation of SF3B2 protects CNS neurons in models of multiple sclerosis. Ann Clin Transl Neurol 2023; 10:246-265. [PMID: 36574260 PMCID: PMC9930435 DOI: 10.1002/acn3.51717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 11/21/2022] [Accepted: 11/21/2022] [Indexed: 12/29/2022] Open
Abstract
OBJECTIVE Neurodegeneration induced by inflammatory stress in multiple sclerosis (MS) leads to long-term neurological disabilities that are not amenable to current immunomodulatory therapies. METHODS AND RESULTS Here, we report that neuronal downregulation of Splicing factor 3b subunit 2 (SF3B2), a component of U2 small nuclear ribonucleoprotein (snRNP), preserves retinal ganglion cell (RGC) survival and axonal integrity in experimental autoimmune encephalomyelitis (EAE)-induced mice. By employing an in vitro system recapitulating the inflammatory environment of MS lesion, we show that when SF3B2 levels are downregulated, cell viability and axon integrity are preserved in cortical neurons against inflammatory toxicity. Notably, knockdown of SF3B2 suppresses the expression of injury-response and necroptosis genes and prevents activation of Sterile Alpha and TIR Motif Containing 1 (Sarm1), a key enzyme that mediates programmed axon degeneration. INTERPRETATION Together, these findings suggest that the downregulation of SF3B2 is a novel potential therapeutic target to prevent secondary neurodegeneration in MS.
Collapse
Affiliation(s)
- Ye Eun Jeong
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland, 21205, USA
| | - Labchan Rajbhandari
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland, 21205, USA
| | - Byung Woo Kim
- Division of Neuropathology, Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, 21205, USA
| | - Arun Venkatesan
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland, 21205, USA
| | - Ahmet Hoke
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland, 21205, USA
| |
Collapse
|
7
|
Camacho-Toledano C, Machín-Díaz I, Calahorra L, Cabañas-Cotillas M, Otaegui D, Castillo-Triviño T, Villar LM, Costa-Frossard L, Comabella M, Midaglia L, García-Domínguez JM, García-Arocha J, Ortega MC, Clemente D. Peripheral myeloid-derived suppressor cells are good biomarkers of the efficacy of fingolimod in multiple sclerosis. J Neuroinflammation 2022; 19:277. [PMCID: PMC9675277 DOI: 10.1186/s12974-022-02635-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Accepted: 10/30/2022] [Indexed: 11/21/2022] Open
Abstract
Background The increasing number of treatments that are now available to manage patients with multiple sclerosis (MS) highlights the need to develop biomarkers that can be used within the framework of individualized medicine. Fingolimod is a disease-modifying treatment that belongs to the sphingosine-1-phosphate receptor modulators. In addition to inhibiting T cell egress from lymph nodes, fingolimod promotes the immunosuppressive activity of myeloid-derived suppressor cells (MDSCs), whose monocytic subset (M-MDSCs) can be used as a biomarker of disease severity, as well as the degree of demyelination and extent of axonal damage in the experimental autoimmune encephalomyelitis (EAE) model of MS. In the present study, we have assessed whether the abundance of circulating M-MDSCs may represent a useful biomarker of fingolimod efficacy in EAE and in the clinical context of MS patients. Methods Treatment with vehicle or fingolimod was orally administered to EAE mice for 14 days in an individualized manner, starting the day when each mouse began to develop clinical signs. Peripheral blood from EAE mice was collected previous to treatment and human peripheral blood mononuclear cells (PBMCs) were collected from fingolimod to treat MS patients’ peripheral blood. In both cases, M-MDSCs abundance was analyzed by flow cytometry and its relationship with the future clinical affectation of each individual animal or patient was assessed. Results Fingolimod-treated animals presented a milder EAE course with less demyelination and axonal damage, although a few animals did not respond well to treatment and they invariably had fewer M-MDSCs prior to initiating the treatment. Remarkably, M-MDSC abundance was also found to be an important and specific parameter to distinguish EAE mice prone to better fingolimod efficacy. Finally, in a translational effort, M-MDSCs were quantified in MS patients at baseline and correlated with different clinical parameters after 12 months of fingolimod treatment. M-MDSCs at baseline were highly representative of a good therapeutic response to fingolimod, i.e., patients who met at least two of the criteria used to define non-evidence of disease activity-3 (NEDA-3) 12 months after treatment. Conclusion Our data indicate that M-MDSCs might be a useful predictive biomarker of the response of MS patients to fingolimod. Supplementary Information The online version contains supplementary material available at 10.1186/s12974-022-02635-3.
Collapse
Affiliation(s)
- Celia Camacho-Toledano
- grid.414883.20000 0004 1767 1847Neuroimmuno-Repair Group, Hospital Nacional de Parapléjicos-SESCAM, Finca La Peraleda s/n, 45071 Toledo, Spain
| | - Isabel Machín-Díaz
- grid.414883.20000 0004 1767 1847Neuroimmuno-Repair Group, Hospital Nacional de Parapléjicos-SESCAM, Finca La Peraleda s/n, 45071 Toledo, Spain
| | - Leticia Calahorra
- grid.414883.20000 0004 1767 1847Neuroimmuno-Repair Group, Hospital Nacional de Parapléjicos-SESCAM, Finca La Peraleda s/n, 45071 Toledo, Spain
| | - María Cabañas-Cotillas
- grid.414883.20000 0004 1767 1847Neuroimmuno-Repair Group, Hospital Nacional de Parapléjicos-SESCAM, Finca La Peraleda s/n, 45071 Toledo, Spain
| | - David Otaegui
- grid.432380.eMultiple Sclerosis Unit, Biodonostia Health Institute, 20014 Donostia-San Sebastián, Spain
| | - Tamara Castillo-Triviño
- grid.432380.eMultiple Sclerosis Unit, Biodonostia Health Institute, 20014 Donostia-San Sebastián, Spain ,grid.414651.30000 0000 9920 5292Neurology Department, Hospital Universitario Donostia, San Sebastián, Spain
| | - Luisa María Villar
- grid.411347.40000 0000 9248 5770Immunology Department, Hospital Universitario Ramón y Cajal, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
| | - Lucienne Costa-Frossard
- grid.411347.40000 0000 9248 5770Immunology Department, Hospital Universitario Ramón y Cajal, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain ,grid.411347.40000 0000 9248 5770Multiple Sclerosis Unit, Neurology, Ramón y Cajal University Hospital, Madrid, Spain
| | - Manuel Comabella
- grid.411083.f0000 0001 0675 8654Neurology-Neuroimmunology Service, Centre d’Esclerosi Múltiple de Catalunya (Cemcat), Institut de Recerca Vall d’Hebron, Hospital Universitari Vall d’Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Luciana Midaglia
- grid.411083.f0000 0001 0675 8654Neurology-Neuroimmunology Service, Centre d’Esclerosi Múltiple de Catalunya (Cemcat), Institut de Recerca Vall d’Hebron, Hospital Universitari Vall d’Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - José Manuel García-Domínguez
- grid.410526.40000 0001 0277 7938Multiple Sclerosis Unit, Department of Neurology, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | - Jennifer García-Arocha
- grid.414883.20000 0004 1767 1847Neuroimmuno-Repair Group, Hospital Nacional de Parapléjicos-SESCAM, Finca La Peraleda s/n, 45071 Toledo, Spain
| | - María Cristina Ortega
- grid.414883.20000 0004 1767 1847Neuroimmuno-Repair Group, Hospital Nacional de Parapléjicos-SESCAM, Finca La Peraleda s/n, 45071 Toledo, Spain
| | - Diego Clemente
- grid.414883.20000 0004 1767 1847Neuroimmuno-Repair Group, Hospital Nacional de Parapléjicos-SESCAM, Finca La Peraleda s/n, 45071 Toledo, Spain
| |
Collapse
|
8
|
Pournajaf S, Dargahi L, Javan M, Pourgholami MH. Molecular Pharmacology and Novel Potential Therapeutic Applications of Fingolimod. Front Pharmacol 2022; 13:807639. [PMID: 35250559 PMCID: PMC8889014 DOI: 10.3389/fphar.2022.807639] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 01/31/2022] [Indexed: 12/14/2022] Open
Abstract
Fingolimod is a well-tolerated, highly effective disease-modifying therapy successfully utilized in the management of multiple sclerosis. The active metabolite, fingolimod-phosphate, acts on sphingosine-1-phosphate receptors (S1PRs) to bring about an array of pharmacological effects. While being initially recognized as a novel agent that can profoundly reduce T-cell numbers in circulation and the CNS, thereby suppressing inflammation and MS, there is now rapidly increasing knowledge on its previously unrecognized molecular and potential therapeutic effects in diverse pathological conditions. In addition to exerting inhibitory effects on sphingolipid pathway enzymes, fingolimod also inhibits histone deacetylases, transient receptor potential cation channel subfamily M member 7 (TRMP7), cytosolic phospholipase A2α (cPLA2α), reduces lysophosphatidic acid (LPA) plasma levels, and activates protein phosphatase 2A (PP2A). Furthermore, fingolimod induces apoptosis, autophagy, cell cycle arrest, epigenetic regulations, macrophages M1/M2 shift and enhances BDNF expression. According to recent evidence, fingolimod modulates a range of other molecular pathways deeply rooted in disease initiation or progression. Experimental reports have firmly associated the drug with potentially beneficial therapeutic effects in immunomodulatory diseases, CNS injuries, and diseases including Alzheimer's disease (AD), Parkinson's disease (PD), epilepsy, and even cancer. Attractive pharmacological effects, relative safety, favorable pharmacokinetics, and positive experimental data have collectively led to its testing in clinical trials. Based on the recent reports, fingolimod may soon find its way as an adjunct therapy in various disparate pathological conditions. This review summarizes the up-to-date knowledge about molecular pharmacology and potential therapeutic uses of fingolimod.
Collapse
Affiliation(s)
- Safura Pournajaf
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Leila Dargahi
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Javan
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | | |
Collapse
|
9
|
Candadai AA, Liu F, Verma A, Adil MS, Alfarhan M, Fagan SC, Somanath PR, Narayanan SP. Neuroprotective Effects of Fingolimod in a Cellular Model of Optic Neuritis. Cells 2021; 10:cells10112938. [PMID: 34831161 PMCID: PMC8616192 DOI: 10.3390/cells10112938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 10/19/2021] [Accepted: 10/21/2021] [Indexed: 11/29/2022] Open
Abstract
Visual dysfunction resulting from optic neuritis (ON) is one of the most common clinical manifestations of multiple sclerosis (MS), characterized by loss of retinal ganglion cells, thinning of the nerve fiber layer, and inflammation to the optic nerve. Current treatments available for ON or MS are only partially effective, specifically target the inflammatory phase, and have limited effects on long-term disability. Fingolimod (FTY) is an FDA-approved immunomodulatory agent for MS therapy. The objective of the current study was to evaluate the neuroprotective properties of FTY in the cellular model of ON-associated neuronal damage. R28 retinal neuronal cell damage was induced through treatment with tumor necrosis factor-α (TNFα). In our cell viability analysis, FTY treatment showed significantly reduced TNFα-induced neuronal death. Treatment with FTY attenuated the TNFα-induced changes in cell survival and cell stress signaling molecules. Furthermore, immunofluorescence studies performed using various markers indicated that FTY treatment protects the R28 cells against the TNFα-induced neurodegenerative changes by suppressing reactive oxygen species generation and promoting the expression of neuronal markers. In conclusion, our study suggests neuroprotective effects of FTY in an in vitro model of optic neuritis.
Collapse
Affiliation(s)
- Amritha A. Candadai
- Clinical and Experimental Therapeutics Program, College of Pharmacy, University of Georgia, Augusta, GA 30912, USA; (A.A.C.); (F.L.); (A.V.); (M.S.A.); (M.A.); (S.C.F.); (P.R.S.)
- Charlie Norwood VA Medical Center, Augusta, GA 30912, USA
- Culver Vision Discovery Institute, Augusta University, Augusta, GA 30912, USA
| | - Fang Liu
- Clinical and Experimental Therapeutics Program, College of Pharmacy, University of Georgia, Augusta, GA 30912, USA; (A.A.C.); (F.L.); (A.V.); (M.S.A.); (M.A.); (S.C.F.); (P.R.S.)
- Charlie Norwood VA Medical Center, Augusta, GA 30912, USA
- Culver Vision Discovery Institute, Augusta University, Augusta, GA 30912, USA
| | - Arti Verma
- Clinical and Experimental Therapeutics Program, College of Pharmacy, University of Georgia, Augusta, GA 30912, USA; (A.A.C.); (F.L.); (A.V.); (M.S.A.); (M.A.); (S.C.F.); (P.R.S.)
- Charlie Norwood VA Medical Center, Augusta, GA 30912, USA
| | - Mir S. Adil
- Clinical and Experimental Therapeutics Program, College of Pharmacy, University of Georgia, Augusta, GA 30912, USA; (A.A.C.); (F.L.); (A.V.); (M.S.A.); (M.A.); (S.C.F.); (P.R.S.)
- Charlie Norwood VA Medical Center, Augusta, GA 30912, USA
| | - Moaddey Alfarhan
- Clinical and Experimental Therapeutics Program, College of Pharmacy, University of Georgia, Augusta, GA 30912, USA; (A.A.C.); (F.L.); (A.V.); (M.S.A.); (M.A.); (S.C.F.); (P.R.S.)
- Charlie Norwood VA Medical Center, Augusta, GA 30912, USA
- Culver Vision Discovery Institute, Augusta University, Augusta, GA 30912, USA
| | - Susan C. Fagan
- Clinical and Experimental Therapeutics Program, College of Pharmacy, University of Georgia, Augusta, GA 30912, USA; (A.A.C.); (F.L.); (A.V.); (M.S.A.); (M.A.); (S.C.F.); (P.R.S.)
- Charlie Norwood VA Medical Center, Augusta, GA 30912, USA
| | - Payaningal R. Somanath
- Clinical and Experimental Therapeutics Program, College of Pharmacy, University of Georgia, Augusta, GA 30912, USA; (A.A.C.); (F.L.); (A.V.); (M.S.A.); (M.A.); (S.C.F.); (P.R.S.)
- Charlie Norwood VA Medical Center, Augusta, GA 30912, USA
| | - S. Priya Narayanan
- Clinical and Experimental Therapeutics Program, College of Pharmacy, University of Georgia, Augusta, GA 30912, USA; (A.A.C.); (F.L.); (A.V.); (M.S.A.); (M.A.); (S.C.F.); (P.R.S.)
- Charlie Norwood VA Medical Center, Augusta, GA 30912, USA
- Culver Vision Discovery Institute, Augusta University, Augusta, GA 30912, USA
- Correspondence:
| |
Collapse
|