1
|
Robichon K, Bibi R, Kiernan M, Denny L, Prisinzano TE, Kivell BM, La Flamme AC. Enhanced and complementary benefits of a nalfurafine and fingolimod combination to treat immune-driven demyelination. Clin Transl Immunology 2023; 12:e1480. [PMID: 38090669 PMCID: PMC10714663 DOI: 10.1002/cti2.1480] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2023] [Revised: 10/19/2023] [Accepted: 11/28/2023] [Indexed: 04/20/2024] Open
Abstract
OBJECTIVES Multiple sclerosis (MS) is a neurodegenerative disease characterised by inflammation and damage to myelin sheaths. While all current disease-modifying treatments (DMTs) are very effective at reducing relapses, they do not slow the progression of the disease, and there is little evidence that these treatments are able to repair or remyelinate damaged axons. Recent evidence suggests that activating kappa opioid receptors (KORs) has a beneficial effect on the progression of MS, and this study investigates the effects of KOR agonists treatment in combination with two current DMTs. METHODS Using the well-established murine model for immune-driven demyelination of MS, experimental autoimmune encephalomyelitis, the effect of KOR agonists in combination with DMTs fingolimod or dimethyl fumarate on disease progression, immune cell infiltration and activation as well as myelination were analysed. RESULTS Fingolimod in combination with the KOR agonist, nalfurafine, significantly increased each individual beneficial effect as measured by increased recovery of mice and reduced relapses. These beneficial effects correlated with a reduction in immune cell infiltration into the CNS as well as peripheral immune cell alterations including a reduction in autoreactive CD4+ T-cell cytokine production as well as increased myelination in the spinal cords of co-treated animals. In contrast, while the use of dimethyl fumarate in combination with nalfurafine did not adversely affect the benefits of nalfurafine, the combination did not significantly enhance those benefits. CONCLUSION This study indicates that KOR agonists can be used in combination with fingolimod and dimethyl fumarate with the nalfurafine-fingolimod combination providing enhanced benefits.
Collapse
Affiliation(s)
- Katharina Robichon
- School of Biological SciencesVictoria University of WellingtonWellingtonNew Zealand
- Centre for Biodiscovery Wellington Victoria University of WellingtonWellingtonNew Zealand
| | - Rabia Bibi
- School of Biological SciencesVictoria University of WellingtonWellingtonNew Zealand
- Centre for Biodiscovery Wellington Victoria University of WellingtonWellingtonNew Zealand
| | - Mackenzie Kiernan
- School of Biological SciencesVictoria University of WellingtonWellingtonNew Zealand
- Centre for Biodiscovery Wellington Victoria University of WellingtonWellingtonNew Zealand
| | - Lisa Denny
- School of Biological SciencesVictoria University of WellingtonWellingtonNew Zealand
- Centre for Biodiscovery Wellington Victoria University of WellingtonWellingtonNew Zealand
| | | | - Bronwyn M Kivell
- School of Biological SciencesVictoria University of WellingtonWellingtonNew Zealand
- Centre for Biodiscovery Wellington Victoria University of WellingtonWellingtonNew Zealand
| | - Anne Camille La Flamme
- School of Biological SciencesVictoria University of WellingtonWellingtonNew Zealand
- Centre for Biodiscovery Wellington Victoria University of WellingtonWellingtonNew Zealand
- Malaghan Institute of Medical ResearchWellingtonNew Zealand
| |
Collapse
|
2
|
Ikumi N, Fujita H. Generalized pustular psoriasis occurring in a patient with multiple sclerosis during treatment with fingolimod. J Dermatol 2023; 50:e295-e296. [PMID: 37017450 DOI: 10.1111/1346-8138.16802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 03/10/2023] [Accepted: 03/24/2023] [Indexed: 04/06/2023]
Affiliation(s)
- Natsumi Ikumi
- Department of Dermatology, Kumagaya General Hospital, Saitama, Japan
- Division of Cutaneous Science, Department of Dermatology, Nihon University School of Medicine, Tokyo, Japan
| | - Hideki Fujita
- Division of Cutaneous Science, Department of Dermatology, Nihon University School of Medicine, Tokyo, Japan
| |
Collapse
|
3
|
Rial SA, Shishani R, Cummings BP, Lim GE. Is 14-3-3 the Combination to Unlock New Pathways to Improve Metabolic Homeostasis and β-Cell Function? Diabetes 2023; 72:1045-1054. [PMID: 37471599 PMCID: PMC10382651 DOI: 10.2337/db23-0094] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Accepted: 05/10/2023] [Indexed: 07/22/2023]
Abstract
Since their discovery nearly five decades ago, molecular scaffolds belonging to the 14-3-3 protein family have been recognized as pleiotropic regulators of diverse cellular and physiological functions. With their ability to bind to proteins harboring specific serine and threonine phosphorylation motifs, 14-3-3 proteins can interact with and influence the function of docking proteins, enzymes, transcription factors, and transporters that have essential roles in metabolism and glucose homeostasis. Here, we will discuss the regulatory functions of 14-3-3 proteins that will be of great interest to the fields of metabolism, pancreatic β-cell biology, and diabetes. We first describe how 14-3-3 proteins play a central role in glucose and lipid homeostasis by modulating key pathways of glucose uptake, glycolysis, oxidative phosphorylation, and adipogenesis. This is followed by a discussion of the contributions of 14-3-3 proteins to calcium-dependent exocytosis and how this relates to insulin secretion from β-cells. As 14-3-3 proteins are major modulators of apoptosis and cell cycle progression, we will explore if 14-3-3 proteins represent a viable target for promoting β-cell regeneration and discuss the feasibility of targeting 14-3-3 proteins to treat metabolic diseases such as diabetes. ARTICLE HIGHLIGHTS 14-3-3 proteins are ubiquitously expressed scaffolds with multiple roles in glucose homeostasis and metabolism. 14-3-3ζ regulates adipogenesis via distinct mechanisms and is required for postnatal adiposity and adipocyte function. 14-3-3ζ controls glucose-stimulated insulin secretion from pancreatic β-cells by regulating mitochondrial function and ATP synthesis as well as facilitating cross talk between β-cells and α-cells.
Collapse
Affiliation(s)
- Sabri A. Rial
- Department of Medicine, University of Montreal, Montreal, Quebec, Canada
- Cardiometabolic Axis, University of Montreal Hospital Research Center (CRCHUM), Montreal, Quebec, Canada
| | - Rahaf Shishani
- Department of Surgery, Center for Alimentary and Metabolic Sciences, School of Medicine, University of California, Davis, Sacramento, CA
| | - Bethany P. Cummings
- Department of Surgery, Center for Alimentary and Metabolic Sciences, School of Medicine, University of California, Davis, Sacramento, CA
| | - Gareth E. Lim
- Department of Medicine, University of Montreal, Montreal, Quebec, Canada
- Cardiometabolic Axis, University of Montreal Hospital Research Center (CRCHUM), Montreal, Quebec, Canada
| |
Collapse
|
4
|
Liao CY, Barrow F, Venkatesan N, Nakao Y, Mauer AS, Fredrickson G, Song MJ, Sehrawat TS, Dasgupta D, Graham RP, Revelo XS, Malhi H. Modulating sphingosine 1-phosphate receptor signaling skews intrahepatic leukocytes and attenuates murine nonalcoholic steatohepatitis. Front Immunol 2023; 14:1130184. [PMID: 37153573 PMCID: PMC10160388 DOI: 10.3389/fimmu.2023.1130184] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Accepted: 04/10/2023] [Indexed: 05/09/2023] Open
Abstract
Sphingosine 1-phosphate (S1P) is a bioactive sphingolipid associated with nonalcoholic steatohepatitis (NASH). Immune cell-driven inflammation is a key determinant of NASH progression. Macrophages, monocytes, NK cells, T cells, NKT cells, and B cells variably express S1P receptors from a repertoire of 5 receptors termed S1P1 - S1P5. We have previously demonstrated that non-specific S1P receptor antagonism ameliorates NASH and attenuates hepatic macrophage accumulation. However, the effect of S1P receptor antagonism on additional immune cell populations in NASH remains unknown. We hypothesized that S1P receptor specific modulation may ameliorate NASH by altering leukocyte recruitment. A murine NASH model was established by dietary feeding of C57BL/6 male mice with a diet high in fructose, saturated fat, and cholesterol (FFC) for 24 weeks. In the last 4 weeks of dietary feeding, the mice received the S1P1,4,5 modulator Etrasimod or the S1P1 modulator Amiselimod, daily by oral gavage. Liver injury and inflammation were determined by histological and gene expression analyses. Intrahepatic leukocyte populations were analyzed by flow cytometry, immunohistochemistry, and mRNA expression. Alanine aminotransferase, a sensitive circulating marker for liver injury, was reduced in response to Etrasimod and Amiselimod treatment. Liver histology showed a reduction in inflammatory foci in Etrasimod-treated mice. Etrasimod treatment substantially altered the intrahepatic leukocyte populations through a reduction in the frequency of T cells, B cells, and NKT cells and a proportional increase in CD11b+ myeloid cells, polymorphonuclear cells, and double negative T cells in FFC-fed and control standard chow diet (CD)-fed mice. In contrast, FFC-fed Amiselimod-treated mice showed no changes in the frequencies of intrahepatic leukocytes. Consistent with the improvement in liver injury and inflammation, hepatic macrophage accumulation and the gene expression of proinflammatory markers such as Lgals3 and Mcp-1 were decreased in Etrasimod-treated FFC-fed mice. Etrasimod treated mouse livers demonstrated an increase in non-inflammatory (Marco) and lipid associated (Trem2) macrophage markers. Thus, S1P1,4,5 modulation by Etrasimod is more effective than S1P1 antagonism by Amiselimod, at the dose tested, in ameliorating NASH, likely due to the alteration of leukocyte trafficking and recruitment. Etrasimod treatment results in a substantial attenuation of liver injury and inflammation in murine NASH.
Collapse
Affiliation(s)
- Chieh-Yu Liao
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, United States
| | - Fanta Barrow
- Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, MN, United States
| | - Nanditha Venkatesan
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, United States
| | - Yasuhiko Nakao
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, United States
| | - Amy S. Mauer
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, United States
| | - Gavin Fredrickson
- Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, MN, United States
| | - Myeong Jun Song
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, United States
- Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Tejasav S. Sehrawat
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, United States
| | - Debanjali Dasgupta
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, United States
| | - Rondell P. Graham
- Division of Anatomic Pathology, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, United States
| | - Xavier S. Revelo
- Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, MN, United States
| | - Harmeet Malhi
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, United States
| |
Collapse
|
5
|
Tourkochristou E, Mouzaki A, Triantos C. Unveiling the biological role of sphingosine-1-phosphate receptor modulators in inflammatory bowel diseases. World J Gastroenterol 2023; 29:110-125. [PMID: 36683721 PMCID: PMC9850947 DOI: 10.3748/wjg.v29.i1.110] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 11/16/2022] [Accepted: 12/14/2022] [Indexed: 01/04/2023] Open
Abstract
Inflammatory bowel disease (IBD) is chronic inflammation of the gastrointestinal tract that has a high epidemiological prevalence worldwide. The increasing disease burden worldwide, lack of response to current biologic therapeutics, and treatment-related immunogenicity have led to major concerns regarding the clinical management of IBD patients and treatment efficacy. Understanding disease pathogenesis and disease-related molecular mechanisms is the most important goal in developing new and effective therapeutics. Sphingosine-1-phosphate (S1P) receptor (S1PR) modulators form a class of oral small molecule drugs currently in clinical development for IBD have shown promising effects on disease improvement. S1P is a sphingosine-derived phospholipid that acts by binding to its receptor S1PR and is involved in the regulation of several biological processes including cell survival, differentiation, migration, proliferation, immune response, and lymphocyte trafficking. T lymphocytes play an important role in regulating inflammatory responses. In inflamed IBD tissue, an imbalance between T helper (Th) and regulatory T lymphocytes and Th cytokine levels was found. The S1P/S1PR signaling axis and metabolism have been linked to inflammatory responses in IBD. S1P modulators targeting S1PRs and S1P metabolism have been developed and shown to regulate inflammatory responses by affecting lymphocyte trafficking, lymphocyte number, lymphocyte activity, cytokine production, and contributing to gut barrier function.
Collapse
Affiliation(s)
- Evanthia Tourkochristou
- Division of Gastroenterology, Department of Internal Medicine, Medical School, University of Patras, Patras 26504, Greece
| | - Athanasia Mouzaki
- Division of Hematology, Department of Internal Medicine, Medical School, University of Patras, Patras 26504, Greece
| | - Christos Triantos
- Division of Gastroenterology, Department of Internal Medicine, Medical School, University of Patras, Patras 26504, Greece
| |
Collapse
|
6
|
Tetorou K, Sisa C, Iqbal A, Dhillon K, Hristova M. Current Therapies for Neonatal Hypoxic-Ischaemic and Infection-Sensitised Hypoxic-Ischaemic Brain Damage. Front Synaptic Neurosci 2021; 13:709301. [PMID: 34504417 PMCID: PMC8421799 DOI: 10.3389/fnsyn.2021.709301] [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: 05/13/2021] [Accepted: 07/19/2021] [Indexed: 12/15/2022] Open
Abstract
Neonatal hypoxic-ischaemic brain damage is a leading cause of child mortality and morbidity, including cerebral palsy, epilepsy, and cognitive disabilities. The majority of neonatal hypoxic-ischaemic cases arise as a result of impaired cerebral perfusion to the foetus attributed to uterine, placental, or umbilical cord compromise prior to or during delivery. Bacterial infection is a factor contributing to the damage and is recorded in more than half of preterm births. Exposure to infection exacerbates neuronal hypoxic-ischaemic damage thus leading to a phenomenon called infection-sensitised hypoxic-ischaemic brain injury. Models of neonatal hypoxia-ischaemia (HI) have been developed in different animals. Both human and animal studies show that the developmental stage and the severity of the HI insult affect the selective regional vulnerability of the brain to damage, as well as the subsequent clinical manifestations. Therapeutic hypothermia (TH) is the only clinically approved treatment for neonatal HI. However, the number of HI infants needed to treat with TH for one to be saved from death or disability at age of 18-22 months, is approximately 6-7, which highlights the need for additional or alternative treatments to replace TH or increase its efficiency. In this review we discuss the mechanisms of HI injury to the immature brain and the new experimental treatments studied for neonatal HI and infection-sensitised neonatal HI.
Collapse
Affiliation(s)
| | | | | | | | - Mariya Hristova
- Perinatal Brain Repair Group, Department of Maternal and Fetal Medicine, UCL Institute for Women’s Health, London, United Kingdom
| |
Collapse
|
7
|
Chiba K. Discovery of fingolimod based on the chemical modification of a natural product from the fungus, Isaria sinclairii. J Antibiot (Tokyo) 2020; 73:666-678. [PMID: 32681100 DOI: 10.1038/s41429-020-0351-0] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 06/30/2020] [Indexed: 12/15/2022]
Abstract
Fingolimod is a first-in-class of sphingosine-1-phosphate (S1P) receptor modulator and is widely used a therapeutic drug for multiple sclerosis (MS), autoimmune disease in the central nervous system. About 25 year ago, a natural product, myriocin was isolated from culture broths of the fungus Isaria sinclairii. Myriocin, a rather complex amino acid having three successive asymmetric centers, was found to show a potent immunosuppressive activity in vitro; however, it induced a strong toxicity in vivo. To find out a less toxic immunosuppressive candidate, the chemical structure of myriocin was simplified to a nonchiral symmetric 2-substituted-2-aminoproane-1,3-diol framework. Finally, a highly potent immunosuppressant, fingolimod was found by the extensive chemical modification and pharmacological evaluation using skin allograft model in vivo. Throughout the analyses of the mechanism action of fingolimod, it is revealed that S1P receptor 1 (S1P1) plays an essential role in lymphocyte circulation and that the molecular target of fingolimod is S1P1. Phosphorylated fingolimod acts as a "functional" antagonist at S1P1, modulates lymphocyte circulation, and shows a potent immunosuppressive activity. Fingolimod significantly reduced the relapse rate of MS in the clinical studies and has been approved as a new therapeutic drug for MS in more than 80 countries.
Collapse
Affiliation(s)
- Kenji Chiba
- Research Unit Immunology & Inflammation, Innovative Research Division, Mitsubishi Tanabe Pharma Corporation, 1000, Kamoshida-cho, Aoba-ku, Yokohama, Kanagawa, 227-0033, Japan.
| |
Collapse
|
8
|
Cohan S, Lucassen E, Smoot K, Brink J, Chen C. Sphingosine-1-Phosphate: Its Pharmacological Regulation and the Treatment of Multiple Sclerosis: A Review Article. Biomedicines 2020; 8:biomedicines8070227. [PMID: 32708516 PMCID: PMC7400006 DOI: 10.3390/biomedicines8070227] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 07/11/2020] [Accepted: 07/15/2020] [Indexed: 12/11/2022] Open
Abstract
Sphingosine-1-phosphate (S1P), via its G-protein-coupled receptors, is a signaling molecule with important regulatory properties on numerous, widely varied cell types. Five S1P receptors (S1PR1-5) have been identified, each with effects determined by their unique G-protein-driven downstream pathways. The discovery that lymphocyte egress from peripheral lymphoid organs is promoted by S1P via S1PR-1 stimulation led to the development of pharmacological agents which are S1PR antagonists. These agents promote lymphocyte sequestration and reduce lymphocyte-driven inflammatory damage of the central nervous system (CNS) in animal models, encouraging their examination of efficacy in the treatment of multiple sclerosis (MS). Preclinical research has also demonstrated direct protective effects of S1PR antagonists within the CNS, by modulation of S1PRs, particularly S1PR-1 and S1PR-5, and possibly S1PR-2, independent of effects upon lymphocytes. Three of these agents, fingolimod, siponimod and ozanimod have been approved, and ponesimod has been submitted for regulatory approval. In patients with MS, these agents reduce relapse risk, sustained disability progression, magnetic resonance imaging markers of disease activity, and whole brain and/or cortical and deep gray matter atrophy. Future opportunities in the development of more selective and intracellular S1PR-driven downstream pathway modulators may expand the breadth of agents to treat MS.
Collapse
|
9
|
Luo RY, Luo C, Zhong F, Shen WY, Li H, Hu ZL, Dai RP. ProBDNF promotes sepsis-associated encephalopathy in mice by dampening the immune activity of meningeal CD4 + T cells. J Neuroinflammation 2020; 17:169. [PMID: 32466783 PMCID: PMC7257240 DOI: 10.1186/s12974-020-01850-0] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2020] [Accepted: 05/19/2020] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Sepsis-associated encephalopathy (SAE) increases the mortality of septic patients, but its mechanism remains unclear. The present study aimed to investigate the roles of T lymphocytes, proBDNF, and their interaction in the pathogenesis of SAE. METHODS Fear conditioning tests were conducted for cognitive assessment in the lipopolysaccharide (LPS, 5 mg kg-1)-induced septic mice. Meninges and peripheral blood were harvested for flow cytometry or qPCR. FTY720 and monoclonal anti-proBDNF antibody (McAb-proB) were used to investigate the effect of lymphocyte depletion and blocking proBDNF on the impaired cognitive functions in the septic mice. RESULTS In the septic mice, cognitive function was impaired, the percentage of CD4+ T cells were decreased in the meninges (P = 0.0021) and circulation (P = 0.0222), and pro-inflammatory cytokines were upregulated, but the anti-inflammatory cytokines interleukin (IL)-4 (P < 0.0001) and IL-13 (P = 0.0350) were downregulated in the meninges. Lymphocyte depletion by intragastrically treated FTY720 (1 mg kg-1) for 1 week ameliorated LPS-induced learning deficit. In addition, proBDNF was increased in the meningeal (P = 0.0042) and peripheral (P = 0.0090) CD4+ T cells. Intraperitoneal injection of McAb-proB (100 μg) before LPS treatment significantly alleviated cognitive dysfunction, inhibited the downregulation of meningeal (P = 0.0264) and peripheral (P = 0.0080) CD4+ T cells, and normalized the gene expression of cytokines in the meninges. However, intra-cerebroventricular McAb-proB injection (1 μg) did not have such effect. Finally, exogenous proBDNF downregulated the percentage of CD4+ T cells in cultured splenocytes from septic mice (P = 0.0021). CONCLUSION Upregulated proBDNF in immune system promoted the pathogenesis of SAE through downregulating the circulating CD4+ T cells, limiting its infiltration into the meninges and perturbing the meningeal pro-/anti-inflammatory homeostasis.
Collapse
Affiliation(s)
- Ru-Yi Luo
- Department of Anesthesiology, The Second Xiangya Hospital, Central South University, Central Ren-Min Road No. 139, Changsha, Hunan Province, China.,Anesthesia Medical Research Center, Central South University, Changsha, China
| | - Cong Luo
- Department of Anesthesiology, The Second Xiangya Hospital, Central South University, Central Ren-Min Road No. 139, Changsha, Hunan Province, China.,Anesthesia Medical Research Center, Central South University, Changsha, China
| | - Feng Zhong
- Anesthesia Medical Research Center, Central South University, Changsha, China.,Department of Anesthesiology, Guangdong Cardiovascular Institute, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Wei-Yun Shen
- Department of Anesthesiology, The Second Xiangya Hospital, Central South University, Central Ren-Min Road No. 139, Changsha, Hunan Province, China.,Anesthesia Medical Research Center, Central South University, Changsha, China
| | - Hui Li
- Department of Anesthesiology, The Second Xiangya Hospital, Central South University, Central Ren-Min Road No. 139, Changsha, Hunan Province, China.,Anesthesia Medical Research Center, Central South University, Changsha, China
| | - Zhao-Lan Hu
- Department of Anesthesiology, The Second Xiangya Hospital, Central South University, Central Ren-Min Road No. 139, Changsha, Hunan Province, China.,Anesthesia Medical Research Center, Central South University, Changsha, China
| | - Ru-Ping Dai
- Department of Anesthesiology, The Second Xiangya Hospital, Central South University, Central Ren-Min Road No. 139, Changsha, Hunan Province, China. .,Anesthesia Medical Research Center, Central South University, Changsha, China.
| |
Collapse
|
10
|
Nystad AE, Lereim RR, Wergeland S, Oveland E, Myhr KM, Bø L, Torkildsen Ø. Fingolimod downregulates brain sphingosine-1-phosphate receptor 1 levels but does not promote remyelination or neuroprotection in the cuprizone model. J Neuroimmunol 2019; 339:577091. [PMID: 31739156 DOI: 10.1016/j.jneuroim.2019.577091] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Revised: 10/22/2019] [Accepted: 10/22/2019] [Indexed: 11/28/2022]
Abstract
Fingolimod is used to treat patients with relapsing-remitting multiple sclerosis; it crosses the blood-brain barrier and modulates sphingosine-1-phosphate receptors (S1PRs). Oligodendrocytes, astrocytes, microglia, and neuronal cells express S1PRs, and fingolimod could potentially improve remyelination and be neuroprotective. We used the cuprizone animal model, histo-, immunohistochemistry, and quantitative proteomics to study the effect of fingolimod on remyelination and axonal damage. Fingolimod was functionally active during remyelination by downregulating S1PR1 brain levels, and fingolimod-treated mice had more oligodendrocytes in the secondary motor cortex after three weeks of remyelination. However, there were no differences in remyelination or axonal damage compared to placebo. Thus, fingolimod does not seem to directly promote remyelination or protect against axonal injury or loss when given after cuprizone-induced demyelination.
Collapse
Affiliation(s)
- Agnes E Nystad
- Norwegian Multiple Sclerosis Competence Centre, Department of Neurology, Haukeland University Hospital, Bergen, Norway; Department of Clinical Medicine, University of Bergen, Bergen, Norway.
| | - Ragnhild Reehorst Lereim
- Proteomics Unit at University of Bergen (PROBE), Department of Biomedicine, University of Bergen, Norway; Computational Biology Unit (CBU), Department of Informatics, University of Bergen, Bergen, Norway
| | - Stig Wergeland
- Norwegian Multiple Sclerosis Competence Centre, Department of Neurology, Haukeland University Hospital, Bergen, Norway; Department of Clinical Medicine, University of Bergen, Bergen, Norway
| | - Eystein Oveland
- Proteomics Unit at University of Bergen (PROBE), Department of Biomedicine, University of Bergen, Norway
| | - Kjell-Morten Myhr
- Department of Clinical Medicine, University of Bergen, Bergen, Norway; Neuro-SysMed, Department of Neurology, Haukeland University Hospital, Bergen, Norway
| | - Lars Bø
- Norwegian Multiple Sclerosis Competence Centre, Department of Neurology, Haukeland University Hospital, Bergen, Norway
| | - Øivind Torkildsen
- Norwegian Multiple Sclerosis Competence Centre, Department of Neurology, Haukeland University Hospital, Bergen, Norway; Department of Clinical Medicine, University of Bergen, Bergen, Norway
| |
Collapse
|
11
|
Volpi C, Orabona C, Macchiarulo A, Bianchi R, Puccetti P, Grohmann U. Preclinical discovery and development of fingolimod for the treatment of multiple sclerosis. Expert Opin Drug Discov 2019; 14:1199-1212. [PMID: 31389262 DOI: 10.1080/17460441.2019.1646244] [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] [Indexed: 12/30/2022]
Abstract
Introduction: Fingolimod, the first oral disease-modifying treatment (DMT) in multiple sclerosis (MS), is a sphingosine 1-phosphate receptor (S1PR) ligand. Approved in 2010, fingolimod has been extensively studied and has been credited with several mechanisms of actions that contribute to its efficacy in MS, among which is the regulation of lymphocyte circulation between the central nervous system and the periphery. Concerns about toxicity, off-target effects, and real-life performance have been raised over time in post-marketing studies of such that next-generation sphingosine-1 phosphate receptor ligands are now being developed. Areas covered: Herein, the authors expand upon previous systematic reviews obtained via PubMed and through their expert opinion on fingolimod use in clinical practice. Long-term data including long-term efficacy, safety, tolerability, and management especially within growing DMT options and pre-treatment constellation in MS patients are discussed, together with the results of an increased understanding of the chemistry underlying the structure-activity relationship. Expert opinion: Despite the limitations illustrated in this article, fingolimod still constitutes a paradigm shift in MS treatment. However, although immunomodulation via S1PRs on lymphocytes has represented a major breakthrough in the clinical management of MS, modifying the evolution of progressive MS will likely require the development of approaches other than merely targeting S1PRs.
Collapse
Affiliation(s)
- Claudia Volpi
- Department of Experimental Medicine, University of Perugia , Perugia , Italy
| | - Ciriana Orabona
- Department of Experimental Medicine, University of Perugia , Perugia , Italy
| | - Antonio Macchiarulo
- Department of Pharmaceutical Sciences, University of Perugia , Perugia , Italy
| | - Roberta Bianchi
- Department of Experimental Medicine, University of Perugia , Perugia , Italy
| | - Paolo Puccetti
- Department of Experimental Medicine, University of Perugia , Perugia , Italy
| | - Ursula Grohmann
- Department of Experimental Medicine, University of Perugia , Perugia , Italy
| |
Collapse
|
12
|
Pelz A, Schaffert H, Diallo R, Hiepe F, Meisel A, Kohler S. S1P receptor antagonists fingolimod and siponimod do not improve the outcome of experimental autoimmune myasthenia gravis mice after disease onset. Eur J Immunol 2017; 48:498-508. [DOI: 10.1002/eji.201747187] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2017] [Revised: 10/30/2017] [Accepted: 11/28/2017] [Indexed: 11/12/2022]
Affiliation(s)
- Andreas Pelz
- Department of Experimental Neurology; Charité - Universitätsmedizin Berlin; Berlin Germany
- Department of Rheumatology and Clinical Immunology; Charité - Universitätsmedizin Berlin; Berlin Germany
| | - Hanne Schaffert
- Department of Experimental Neurology; Charité - Universitätsmedizin Berlin; Berlin Germany
| | - Radharani Diallo
- Department of Experimental Neurology; Charité - Universitätsmedizin Berlin; Berlin Germany
| | - Falk Hiepe
- Department of Rheumatology and Clinical Immunology; Charité - Universitätsmedizin Berlin; Berlin Germany
| | - Andreas Meisel
- Department of Experimental Neurology; Charité - Universitätsmedizin Berlin; Berlin Germany
- Department of Neurology; Charité - Universitätsmedizin Berlin; Berlin Germany
- NeuroCure Clinical Research Center (NCRC); Charité - Universitätsmedizin Berlin; Berlin Germany
| | - Siegfried Kohler
- Department of Experimental Neurology; Charité - Universitätsmedizin Berlin; Berlin Germany
- Department of Neurology; Charité - Universitätsmedizin Berlin; Berlin Germany
- NeuroCure Clinical Research Center (NCRC); Charité - Universitätsmedizin Berlin; Berlin Germany
| |
Collapse
|
13
|
Klotz L, Berthele A, Brück W, Chan A, Flachenecker P, Gold R, Haghikia A, Hellwig K, Hemmer B, Hohlfeld R, Korn T, Kümpfel T, Lang M, Limmroth V, Linker RA, Meier U, Meuth SG, Paul F, Salmen A, Stangel M, Tackenberg B, Tumani H, Warnke C, Weber MS, Ziemssen T, Zipp F, Wiendl H. [Monitoring of blood parameters under course-modified MS therapy : Substance-specific relevance and current recommendations for action]. DER NERVENARZT 2017; 87:645-59. [PMID: 26927677 DOI: 10.1007/s00115-016-0077-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
With the approval of various substances for the immunotherapy of multiple sclerosis (MS), treatment possibilities have improved significantly over the last few years. Indeed, the choice of individually tailored preparations and treatment monitoring for the treating doctor is becoming increasingly more complex. This is particularly applicable for monitoring for a treatment-induced compromise of the immune system. The following article by members of the German Multiple Sclerosis Skills Network (KKNMS) and the task force "Provision Structures and Therapeutics" summarizes the practical recommendations for approved immunotherapy for mild to moderate and for (highly) active courses of MS. The focus is on elucidating the substance-specific relevance of particular laboratory parameters with regard to the mechanism of action and the side effects profile. To enable appropriate action to be taken in clinical practice, any blood work changes that can be expected, in addition to any undesirable laboratory findings and their causes and relevance, should be elucidated.
Collapse
Affiliation(s)
- L Klotz
- Department für Neurologie, Universitätsklinikum Münster, Albert-Schweitzer-Campus 1, Gebäude A1, 48149, Münster, Deutschland
| | - A Berthele
- Neurologische Klinik und Poliklinik, Klinikum rechts der Isar der TU München, Ismaninger Straße 22, 81675, München, Deutschland
| | - W Brück
- Institut für Neuropathologie, Universitätsmedizin Göttingen der Georg-August-Universität, Robert-Koch-Str. 40, 37075, Göttingen, Deutschland
| | - A Chan
- Neurologische Klinik, St. Josef-Hospital, Universitätsklinikum der Ruhr-Universität Bochum, Gudrunstr. 56, 44791, Bochum, Deutschland
| | - P Flachenecker
- Neurologisches Rehabilitationszentrum Quellenhof in Bad Wildbad GmbH, Kuranlagenallee 2, 75323, Bad Wildbad, Deutschland
| | - R Gold
- Neurologische Klinik, St. Josef-Hospital, Universitätsklinikum der Ruhr-Universität Bochum, Gudrunstr. 56, 44791, Bochum, Deutschland
| | - A Haghikia
- Neurologische Klinik, St. Josef-Hospital, Universitätsklinikum der Ruhr-Universität Bochum, Gudrunstr. 56, 44791, Bochum, Deutschland
| | - K Hellwig
- Neurologische Klinik, St. Josef-Hospital, Universitätsklinikum der Ruhr-Universität Bochum, Gudrunstr. 56, 44791, Bochum, Deutschland
| | - B Hemmer
- Neurologische Klinik und Poliklinik, Klinikum rechts der Isar der TU München, Ismaninger Straße 22, 81675, München, Deutschland
| | - R Hohlfeld
- Institut für Klinische Neuroimmunologie, Klinikum der Universität München, Campus Großhadern, Marchioninistr. 15, 81377, München, Deutschland
| | - T Korn
- Neurologische Klinik und Poliklinik, Klinikum rechts der Isar der TU München, Ismaninger Straße 22, 81675, München, Deutschland
| | - T Kümpfel
- Institut für Klinische Neuroimmunologie, Klinikum der Universität München, Campus Großhadern, Marchioninistr. 15, 81377, München, Deutschland
| | - M Lang
- NeuroTransConcept GmbH, Centers of Excellence, Pfauengasse 8, 89073, Ulm, Deutschland
| | - V Limmroth
- Klinik für Neurologie und Palliativmedizin, Kliniken der Stadt Köln, Ostmerheimer Str. 200, 51109, Köln - Merheim, Deutschland
| | - R A Linker
- Neurologische Klinik, Universitätsklinikum Erlangen, Schwabachanlage 6, 91054, Erlangen, Deutschland
| | - U Meier
- Berufsverband Deutscher Neurologen BDN, Am Ziegelkamp 1f, 41515, Grevenbroich, Deutschland
| | - S G Meuth
- Department für Neurologie, Universitätsklinikum Münster, Albert-Schweitzer-Campus 1, Gebäude A1, 48149, Münster, Deutschland
| | - F Paul
- Institut für Neuroimmunologie, Universitätsklinikum Charité, Schumannstr. 20/21, 10117, Berlin, Deutschland
| | - A Salmen
- Neurologische Klinik, St. Josef-Hospital, Universitätsklinikum der Ruhr-Universität Bochum, Gudrunstr. 56, 44791, Bochum, Deutschland
| | - M Stangel
- Klinik für Neurologie, Medizinische Hochschule Hannover, Carl-Neuberg-Str. 1, 30625, Hannover, Deutschland
| | - B Tackenberg
- Klinik für Neurologie, Philipps-Universität und Universitätsklinikum Marburg, Baldingerstr. 1, 35043, Marburg, Deutschland
| | - H Tumani
- Neurologische Universitätsklinik der Universität Ulm, Oberer Eselsberg 45, 89081, Ulm, Deutschland.,Fachklinik für Neurologie Dietenbronn, Dietenbronn 7, 88477, Schwendi, Deutschland
| | - C Warnke
- Klinik für Neurologie, Universitätsklinikum Düsseldorf, Moorenstraße 5, 40225, Düsseldorf, Deutschland
| | - M S Weber
- Institut für Neuropathologie, Universitätsmedizin Göttingen der Georg-August-Universität, Robert-Koch-Str. 40, 37075, Göttingen, Deutschland
| | - T Ziemssen
- Klinik und Poliklinik für Neurologie, Universitätsklinikum Carl Gustav Carus der TU Dresden, Fetscherstr. 74, 01307, Dresden, Deutschland
| | - F Zipp
- Klinik für Neurologie, Universitätsmedizin der Johannes-Gutenberg-Universität Mainz, Langenbeckstr. 1, 55131, Mainz, Deutschland
| | - H Wiendl
- Department für Neurologie, Universitätsklinikum Münster, Albert-Schweitzer-Campus 1, Gebäude A1, 48149, Münster, Deutschland.
| |
Collapse
|
14
|
Hemdan NYA, Weigel C, Reimann CM, Gräler MH. Modulating sphingosine 1-phosphate signaling with DOP or FTY720 alleviates vascular and immune defects in mouse sepsis. Eur J Immunol 2016; 46:2767-2777. [PMID: 27683081 DOI: 10.1002/eji.201646417] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2016] [Revised: 09/01/2016] [Accepted: 09/27/2016] [Indexed: 12/12/2022]
Abstract
Sepsis is a systemic inflammatory response to pathogens and a leading cause of hospital related mortality worldwide. Sphingosine 1-phosphate (S1P) regulates multiple cellular processes potentially involved in the pathogenesis of sepsis, including antigen presentation, lymphocyte egress, and maintenance of vascular integrity. We thus explored the impact of manipulating S1P signaling in experimental polymicrobial sepsis in mice. Administration of 4-deoxypyridoxine (DOP), an inhibitor of the S1P-degrading enzyme S1P-lyase, or of the sphingosine analog FTY720 that serves as an S1P receptor agonist after phosphorylation ameliorated morbidity, improved recovery from sepsis in surviving mice, and reduced sepsis-elicited hypothermia and body weight loss. Treated mice developed lymphopenia, leading to an accumulation of lymphocytes in peripheral lymph nodes, and reduced bacterial burden in liver, but not in blood. Sepsis-induced upregulation of mRNA expression of cytokines in spleen remained unchanged, but reduction of IL-6, TNF-α, MCP-1, and IL-10 in plasma was evident. DOP and FTY720 treatment significantly reduced levels of Evans blue leakage from blood into liver and lung, decreased hematocrit values, and lowered plasma levels of VEGF-A in septic mice. Collectively, our results indicate that modulation of S1P signaling showed a protective phenotype in experimental sepsis by modulating vascular and immune functions.
Collapse
Affiliation(s)
- Nasr Y A Hemdan
- Department of Anesthesiology and Intensive Care Medicine, Center for Sepsis Control and Care (CSCC), and the Center for Molecular Biomedicine (CMB), University Hospital Jena, Jena, Germany
| | - Cynthia Weigel
- Department of Anesthesiology and Intensive Care Medicine, Center for Sepsis Control and Care (CSCC), and the Center for Molecular Biomedicine (CMB), University Hospital Jena, Jena, Germany.,Fritz Lipmann Institute, Leibniz Institute on Aging, Jena, Germany
| | - Christina-Maria Reimann
- Department of Anesthesiology and Intensive Care Medicine, Center for Sepsis Control and Care (CSCC), and the Center for Molecular Biomedicine (CMB), University Hospital Jena, Jena, Germany
| | - Markus H Gräler
- Department of Anesthesiology and Intensive Care Medicine, Center for Sepsis Control and Care (CSCC), and the Center for Molecular Biomedicine (CMB), University Hospital Jena, Jena, Germany
| |
Collapse
|
15
|
Abstract
Objective To review the pharmacology, pharmacokinetics, efficacy, and safety of mycophenolate sodium, everolimus, and FTY720. Study Selection and Data Extraction Clinical trials and abstracts evaluating mycophenolate sodium, everolimus, and FTY720 in solid-organ transplantation were considered for evaluation. English-language studies and published abstracts were selected for inclusion. Data Synthesis Mycophenolate sodium has recently been approved by the Food and Drug Adminstration for marketing in the United States; everolimus and FTY720 are immunosuppressive agents that may soon be available in the United States. These agents have proven efficacy in reducing the incidence of acute rejection in solid-organ transplantation. Clinical trials have shown that these newer agents are relatively well tolerated. The most common adverse events associated with these agents were gastrointestinal and hematologic effects (mycophenolate sodium); hyperlipidemia, increased serum creatinine, and hematologic effects (everolimus); and gastrointestinal effects, headache, and bradycardia (FTY720). Conclusion Mycophenolate sodium has been approved in some European countries and the United States. Everolimus has been approved in some European countries and a new drug application has been submitted to the Food and Drug Administration. FTY720 is currently in phase III clinical trials and submission to the Food and Drug Administration for approval is a few years away. The approval of these agents will furnish the transplant practitioner with even more options for immunosuppression.
Collapse
|
16
|
Tamakuwala M, Ratna W, Joshi A, Stagni G. Fingolimod hydrochloride gel shows promising therapeutic effects in a mouse model of atopic dermatitis. ACTA ACUST UNITED AC 2016; 68:1268-77. [PMID: 27465785 DOI: 10.1111/jphp.12588] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2016] [Accepted: 05/14/2016] [Indexed: 11/26/2022]
Abstract
OBJECTIVES To assess the efficacy of topically applied 2% hydroxypropyl cellulose gels containing 0.5% fingolimod hydrochloride (FNGL) with or without 6% colloidal oatmeal in an in vivo model of atopic dermatitis (AD). METHODS AD-like lesions were induced in SKH1/Hr hairless mice and were treated with FNGL gels, non-medicated base gels and Elidel(®) cream for 6 weeks. The severity/improvement of the lesions was assessed regularly using the Eczema Area and Severity Index (EASI), pH of the skin, transepidermal water loss, g/m(2) /h (TEWL), humidity and temperature. At the end of the experiments, the plasma levels of cytokines, FNGL and white blood cells were determined. KEY FINDINGS The EASI score was almost unchanged for the vehicle-only groups compared to before the treatments, whereas the medicated groups showed a significant decrease in the overall EASI score (P < 0.01), although there was non-significant differences among them (P > 0.081). Both the FNGL groups also showed a significant (P ˂ 0.05) reduction in blood WBC. CONCLUSION This study shows that the gels containing 0.50% FNGL and FNGL 0.50% plus 6% colloidal oatmeal have potential for the treatment of AD. The presence of colloidal oatmeal may provide additional benefits.
Collapse
Affiliation(s)
- Mayurkumar Tamakuwala
- Division of Pharmaceutical Sciences, Arnold and Marie Schwartz College of Pharmacy and Health Sciences, Long Island University, Brooklyn, NY, USA
| | - Warren Ratna
- Division of Pharmaceutical Sciences, Arnold and Marie Schwartz College of Pharmacy and Health Sciences, Long Island University, Brooklyn, NY, USA
| | - Amit Joshi
- Division of Pharmaceutical Sciences, Arnold and Marie Schwartz College of Pharmacy and Health Sciences, Long Island University, Brooklyn, NY, USA
| | - Grazia Stagni
- Division of Pharmaceutical Sciences, Arnold and Marie Schwartz College of Pharmacy and Health Sciences, Long Island University, Brooklyn, NY, USA.
| |
Collapse
|
17
|
Therapeutic approach to mite-induced intractable dermatitis using novel immunomodulator FTY720 ointment (fingolimod) in NC/Nga mice. Allergol Int 2016; 65:172-179. [PMID: 26666476 DOI: 10.1016/j.alit.2015.10.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2015] [Revised: 10/15/2015] [Accepted: 10/22/2015] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND The increasing incidence and prevalence of atopic dermatitis (AD) demands new therapeutic approaches for treating the disease. We investigated the therapeutic efficacy of immunomodulator FTY720 ointment (fingolimod) for mite-induced intractable AD using an NC/Nga mouse model. METHODS Female NC/Nga mice that developed severe AD were divided into four groups: (1) FTY720 (0.001% FTY720 ointment), (2) tacrolimus (tacrolimus hydrate ointment) (3) betamethasone (betamethasone ointment), and (4) ointment base (hydrophilic petrolatum), all of which received treatment six times per week. Therapeutic efficacy after two weeks was evaluated in terms of AD severity, histochemical observations (epidermal hypertrophy, mast cell accumulation, and CD3(+) T cell infiltration), transepidermal water loss (TEWL), and epidermal barrier function (filaggrin expression). RESULTS Betamethasone treatment showed little effect, confirming that the AD was intractable. In the FTY720 group, AD improved significantly compared with the ointment base group, as did epidermal hypertrophy, mast cell accumulation, and CD3(+) T cell infiltration. In contrast, AD in the tacrolimus and betamethasone groups did not improve significantly, nor did epidermal hypertrophy or mast cell accumulation. Furthermore, in the FTY720 group, TEWL decreased significantly compared with the ointment base group, and filaggrin expression significantly increased compared with the betamethasone and ointment base groups. CONCLUSIONS FTY720 ointment is a promising candidate for treatment of intractable AD. These findings also provide the first evidence that FTY720 ointment ameliorates epidermal barrier function.
Collapse
|
18
|
Cruickshanks N, Roberts JL, Bareford MD, Tavallai M, Poklepovic A, Booth L, Spiegel S, Dent P. Differential regulation of autophagy and cell viability by ceramide species. Cancer Biol Ther 2016; 16:733-42. [PMID: 25803131 DOI: 10.1080/15384047.2015.1026509] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
The present studies sought to determine whether the anti-folate pemetrexed (Alimta) and the sphingosine-1-phosphate receptor modulator FTY720 (Fingolimod, Gilenya) interacted to kill tumor cells. FTY720 and pemetrexed interacted in a greater than additive fashion to kill breast, brain and colorectal cancer cells. Loss of p53 function weakly enhanced the toxicity of FTY720 whereas deletion of activated RAS strongly or expression of catalytically inactive AKT facilitated killing. Combined drug exposure reduced the activity of AKT, p70 S6K and mTOR and activated JNK and p38 MAPK. Expression of activated forms of AKT, p70 S6K and mTOR or inhibition of JNK and p38 MAPK suppressed the interaction between FTY720 and pemetrexed. Treatment of cells with FTY720 and pemetrexed increased the numbers of early autophagosomes but not autolysosomes, which correlated with increased LC3II processing and increased p62 levels, suggestive of stalled autophagic flux. Knock down of ATG5 or Beclin1 suppressed autophagosome formation and cell killing. Knock down of ceramide synthase 6 suppressed autophagosome production and cell killing whereas knock down of ceramide synthase 2 enhanced vesicle formation and facilitated death. Collectively our findings argue that pemetrexed and FTY720 could be a novel adjunct modality for breast cancer treatment.
Collapse
Key Words
- Ad, adenovirus
- Alimta
- CMV, empty vector plasmid or virus
- CerS, ceramide synthase
- CerS2
- CerS6
- ER, endoplasmic reticulum
- ERK, extracellular regulated kinase
- FTY720
- Gilenya
- IP, immunoprecipitation
- LASS, longevity assurance gene
- MAPK, mitogen activated protein kinase
- MEK, mitogen activated extracellular regulated kinase
- PI3K, phosphatidyl inositol 3 kinase
- PTEN, phosphatase and tensin homolog on chromosome 10
- PTX, pemetrexed
- Pemetrexed
- ROS, reactive oxygen species
- S1P
- SCR, scrambled
- VEH, vehicle.
- autophagy
- ca, constitutively active
- ceramide
- dn, dominant negative
- mTOR, mammalian target of rapamycin
- si, small interfering
Collapse
Affiliation(s)
- Nichola Cruickshanks
- a Department of Biochemistry and Molecular Biology; Virginia Commonwealth University ; Richmond , VA , USA
| | | | | | | | | | | | | | | |
Collapse
|
19
|
Farrokhi M, Beni AA, Etemadifar M, Rezaei A, Rivard L, Zadeh AR, Sedaghat N, Ghadimi M. Effect of Fingolimod on Platelet Count Among Multiple Sclerosis Patients. Int J Prev Med 2016; 6:125. [PMID: 26900439 PMCID: PMC4736130 DOI: 10.4103/2008-7802.172539] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2015] [Accepted: 08/23/2015] [Indexed: 11/10/2022] Open
Abstract
Background: While many studies have previously focused on fingolimod's effect on immune cells, the effect it has on circulating and local central nervous system platelets (Plts) has not yet been investigated. This study will elucidate what effects fingolimod treatment has on multiple sclerosis (MS) patients’ plasma Plt levels. In addition, it will propose possible reasoning for these effects and suggest further investigation into this topic. Methods: This quasi-experimental study used patients from the Isfahan Multiple Sclerosis Society to produce a subject pool of 80 patients, including 14 patients who ceased fingolimod use due to complications. The patients had their blood analyzed to determine Plt levels both 1-month prior to fingolimod treatment and 1-month after fingolimod treatment had been started. Results: The mean level of Plts before initiation of fingolimod therapy (Plt1) among these MS patients was 256.53 ± 66.26. After 1-month of fingolimod treatment, the Plt level yielded an average of 229.96 ± 49.67 (Plt2). This number is significantly lower than the average Plt count before treatment (P < 0.01). Conclusions: MS patients taking oral fingolimod treatment may be at risk for side-effects caused by low Plt levels. This may not be a factor for patients with higher or normal Plt levels. However, a patient with naturally low Plt levels may experience a drop below the normal level and be at risk for excessive bleeding. In addition to these possible harmful side-effects, the decreased Plt population may pose positive effects for MS patients.
Collapse
Affiliation(s)
- Mehrdad Farrokhi
- Department of Immunology, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran; Isfahan Neurosciences Research Centre, Affiliated to Isfahan University of Medical Sciences, Isfahan, Iran
| | - Ali Amani Beni
- Department of Immunology, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran; Isfahan Neurosciences Research Centre, Affiliated to Isfahan University of Medical Sciences, Isfahan, Iran
| | - Masoud Etemadifar
- Isfahan Neurosciences Research Centre, Affiliated to Isfahan University of Medical Sciences, Isfahan, Iran; Department of Neurology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran; Multiple Sclerosis and Neuroimmunology Research Center, Isfahan, Iran
| | - Ali Rezaei
- Department of Molecular Biology, Cell Biology, and Biochemistry, Brown University, Providence, Rhode Island 02912, USA; Department of Neuroscience, Brown University, Providence, Rhode Island 02912, USA
| | - Leah Rivard
- Department of Molecular Biology, Cell Biology, and Biochemistry, Brown University, Providence, Rhode Island 02912, USA; Department of Neuroscience, Brown University, Providence, Rhode Island 02912, USA
| | - Aryan Rafiee Zadeh
- Department of Immunology, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Nahid Sedaghat
- Isfahan Neurosciences Research Centre, Affiliated to Isfahan University of Medical Sciences, Isfahan, Iran
| | - Milad Ghadimi
- Department of Electrical and Computer Engineering, Isfahan University of Technology, Isfahan, Iran; Department of Immunology, School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| |
Collapse
|
20
|
Gardner NM, Riley RT, Showker JL, Voss KA, Sachs AJ, Maddox JR, Gelineau-van Waes JB. Elevated Nuclear and Cytoplasmic FTY720-Phosphate in Mouse Embryonic Fibroblasts Suggests the Potential for Multiple Mechanisms in FTY720-Induced Neural Tube Defects. Toxicol Sci 2015; 150:161-8. [PMID: 26719367 DOI: 10.1093/toxsci/kfv321] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
FTY720 (fingolimod) is a U.S. Food and Drug Administration-approved drug to treat relapsing remitting multiple sclerosis. FTY720 treatment in pregnant inbred LM/Bc mice results in approximately 60% of embryos having a neural tube defect (NTD). Sphingosine kinases (Sphk1, Sphk2) phosphorylate FTY720 in vivo to form the bioactive metabolite FTY720-1-phosphate (FTY720-P). Cytoplasmic FTY720-P is an agonist for 4 of the 5 sphingosine-1-phosphate (S1P) receptors (S1P1, 3-5) and can also act as a functional antagonist of S1P1, whereas FTY720-P generated in the nucleus inhibits histone deacetylases (HDACs), leading to increased histone acetylation. This study demonstrates that treatment of LM/Bc mouse embryonic fibroblasts (MEFs) with FTY720 results in a significant accumulation of FTY720-P in both the cytoplasmic and nuclear compartments. Elevated nuclear FTY720-P is associated with decreased HDAC activity and increased histone acetylation at H3K18 and H3K23 in LM/Bc MEFs. Treatment of LM/Bc MEFs with FTY720 and a selective Sphk2 inhibitor, ABC294640, significantly reduces the amount of FTY720-P that accumulates in the nucleus. The data provide insight into the relative amounts of FTY720-P generated in the nuclear versus cytoplasmic subcellular compartments after FTY720 treatment and the specific Sphk isoforms involved. The results of this study suggest that FTY720-induced NTDs may involve multiple mechanisms, including: (1) sustained and/or altered S1P receptor activation and signaling by FTY720-P produced in the cytoplasm and (2) HDAC inhibition and histone hyperacetylation by FTY720-P generated in the nucleus that could lead to epigenetic changes in gene regulation.
Collapse
Affiliation(s)
- Nicole M Gardner
- *Department of Pharmacology, Creighton University School of Medicine, Omaha, Nebraska 68178 and
| | - Ronald T Riley
- USDA-ARS, Toxicology and Mycotoxin Research Unit, Athens, Georgia 30605
| | - Jency L Showker
- USDA-ARS, Toxicology and Mycotoxin Research Unit, Athens, Georgia 30605
| | - Kenneth A Voss
- USDA-ARS, Toxicology and Mycotoxin Research Unit, Athens, Georgia 30605
| | - Andrew J Sachs
- *Department of Pharmacology, Creighton University School of Medicine, Omaha, Nebraska 68178 and
| | - Joyce R Maddox
- *Department of Pharmacology, Creighton University School of Medicine, Omaha, Nebraska 68178 and
| | | |
Collapse
|
21
|
Fingolimod ameliorates the development of experimental autoimmune encephalomyelitis by inhibiting Akt-mTOR axis in mice. Int Immunopharmacol 2015; 30:171-178. [PMID: 26632437 DOI: 10.1016/j.intimp.2015.11.024] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2015] [Revised: 11/19/2015] [Accepted: 11/19/2015] [Indexed: 11/20/2022]
Abstract
Fingolimod is a new immunosuppressive agent approved by Food and Drug Administration (FDA) for treating multiple sclerosis (MS). It acts as a functional antagonist to downregulate the S1P1 receptor, which is known to signal through the Akt-mTOR pathway. We investigated the mechanism of fingolimod action in the classical animal model of MS: experimental autoimmune encephalomyelitis (EAE). Fingolimod treatment significantly reduced clinical scores and histopathology in this model, even when treatment was begun after the onset of pathology. The Akt-mTOR signaling pathway was shown to be activated in the EAE model, by measuring the abundance of downstream activation markers, pAkt and ps6k. And this pathway was inhibited when EAE mice were treated with fingolimod. Mice with EAE exhibited an increased frequency of Th1 cells in the spleen, with concomitant increases in the mRNA levels of Tbet and Ifng and increased IFN-γ production by activated splenocytes; the frequency of Treg cells, as well as mRNA levels of Foxp3 and Tgfb, was reduced, as was TGF-β production by activated splenocytes. After treatment with fingolimod, these parameters were reversed, suggesting that fingolimod treatment inhibits the Akt-mTOR axis in EAE, which affects the differentiation and function of Th1 and Treg cells. These results provide an insight into the mechanism of action of fingolimod treatment and may provide new ideas for treating EAE and MS.
Collapse
|
22
|
Yoshida Y, Mikami N, Matsushima Y, Otani F, Miyawaki M, Takatsuji M, Banno R, Tsuji T, Fujita T, Tsujikawa K, Kohno T. Functional Mechanism(s) of the Inhibition of Disease Progression by Combination Treatment with Fingolimod Plus Pathogenic Antigen in a Glucose-6-phosphate Isomerase Peptide-Induced Arthritis Mouse Model. Biol Pharm Bull 2015; 38:1120-5. [PMID: 25994913 DOI: 10.1248/bpb.b14-00873] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
We previously reported that combination treatment with fingolimod (FTY720) plus antigenic peptide of glucose-6-phosphate isomerase (residues 325-339) (GPI325-339) from the onset of symptoms significantly inhibited disease progression in a mouse model of GPI325-339-induced arthritis. In this study, we investigated the mechanism(s) involved. The model mice were treated from arthritis onset with FTY720 alone, GPI325-339 alone, or the combination of FTY720 plus GPI325-339. At the end of treatment, inguinal lymph nodes (LNs) were excised and examined histologically and in flow cytometry. Levels of apoptotic cells, programmed death-1-expressing CD4(+)forkhead box P3(-) nonregulatory T cells (non-Tregs), and cytotoxic T-lymphocyte antigen 4-expressing non-Tregs in inguinal LNs were markedly increased in the combination treatment group mice. Regulatory T cells (Tregs) were also increased. These results indicate that combination treatment with FTY720 plus GPI325-339 inhibits the progression of arthritis by inducing clonal deletion and anergy of pathogenic T cells and also by immune suppression via Tregs.
Collapse
Affiliation(s)
- Yuya Yoshida
- Department of Pathological Biochemistry, Faculty of Pharmaceutical Sciences, Setsunan University
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Abstract
The interface of multiple sclerosis (MS) and infection occurs on several levels. First, infectious disease has been postulated as a potential trigger, if not cause, of MS. Second, exacerbation of MS has been well-documented as a consequence of infection, and, lastly, infectious diseases have been recognized as a complication of the therapies currently employed in the treatment of MS. MS is a disease in which immune dysregulation is a key component. Examination of central nervous system (CNS) tissue of people affected by MS demonstrates immune cell infiltration, activation and inflammation. Therapies that alter the immune response have demonstrated efficacy in reducing relapse rates and evidence of brain inflammation on magnetic resonance imaging (MRI). Despite the altered immune response in MS, there is a lack of evidence that these patients are at increased risk of infectious disease in the absence of treatment or debility. Links between infections and disease-modifying therapies (DMTs) used in MS will be discussed in this review, as well as estimates of occurrence and ways to potentially minimize these risks. We address infection in MS in a comprehensive fashion, including (1) the impact of infections on relapse rates in patients with MS; (2) a review of available infection data from pivotal trials and postmarketing studies for the approved and experimental DMTs, including frequency, types and severity of infections; and (3) relevant risk minimization strategies, particularly as they pertain to progressive multifocal leukoencephalopathy (PML).
Collapse
|
24
|
Brunkhorst R, Vutukuri R, Pfeilschifter W. Fingolimod for the treatment of neurological diseases-state of play and future perspectives. Front Cell Neurosci 2014; 8:283. [PMID: 25309325 PMCID: PMC4162362 DOI: 10.3389/fncel.2014.00283] [Citation(s) in RCA: 111] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2014] [Accepted: 08/25/2014] [Indexed: 11/25/2022] Open
Abstract
Sphingolipids are a fascinating class of signaling molecules derived from the membrane lipid sphingomyelin. They show abundant expression in the brain. Complex sphingolipids such as glycosphingolipids (gangliosides and cerebrosides) regulate vesicular transport and lysosomal degradation and their dysregulation can lead to storage diseases with a neurological phenotype. More recently, simple sphingolipids such ceramide, sphingosine and sphingosine 1-phosphate (S1P) were discovered to signal in response to many extracellular stimuli. Forming an intricate signaling network, the balance of these readily interchangeable mediators is decisive for cell fate under stressful conditions. The immunomodulator fingolimod is the prodrug of an S1P receptor agonist. Following receptor activation, the drug leads to downregulation of the S1P1 receptor inducing functional antagonism. As the first drug to modulate the sphingolipid signaling pathway, it was marketed in 2010 for the treatment of multiple sclerosis (MS). At that time, immunomodulation was widely accepted as the key mechanism of fingolimod’s efficacy in MS. But given the excellent passage of this lipophilic compound into the brain and its massive brain accumulation as well as the abundant expression of S1P receptors on brain cells, it is conceivable that fingolimod also affects brain cells directly. Indeed, a seminal study showed that the protective effect of fingolimod in experimental autoimmune encephalitis (EAE), a murine MS model, is lost in mice lacking the S1P1 receptor on astrocytes, arguing for a specific role of astrocytic S1P signaling in MS. In this review, we discuss the role of sphingolipid mediators and their metabolizing enzymes in neurologic diseases and putative therapeutic strategies arising thereof.
Collapse
Affiliation(s)
- Robert Brunkhorst
- Cerebrovascular Research Group, Department of Neurology, Frankfurt University Hospital Frankfurt am Main, Germany
| | - Rajkumar Vutukuri
- Institute of General Pharmacology and Toxicology, pharmazentrum frankfurt, Goethe University Frankfurt Frankfurt am Main, Germany
| | - Waltraud Pfeilschifter
- Cerebrovascular Research Group, Department of Neurology, Frankfurt University Hospital Frankfurt am Main, Germany
| |
Collapse
|
25
|
Tsuji T, Yoshida Y, Fujita T, Kohno T. Oral therapy for type 1 diabetes mellitus using a novel immunomodulator, FTY720 (fingolimod), in combination with sitagliptin, a dipeptidyl peptidase-4 inhibitor, examined in non-obese diabetic mice. J Diabetes Investig 2014; 3:441-8. [PMID: 24843604 PMCID: PMC4019244 DOI: 10.1111/j.2040-1124.2012.00218.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Aims/Introduction: The therapeutic effectiveness against type 1 diabetes mellitus of a novel immunomodulator, FTY720 (fingolimod), in combination with sitagliptin, a dipeptidyl peptidase‐4 inhibitor, was examined in the non‐obese diabetic (NOD) mouse model. Materials and Methods: Female NOD mice that had developed type 1 diabetes mellitus spontaneously were divided into four groups according to which therapy they received: (i) FTY720 (0.1 mg/kg, orally, six times a week) plus sitagliptin (1 mg/kg, orally, six times a week); (ii) FTY720 (0.1 mg/kg, orally, six times a week); (iii) sitagliptin (1 mg/kg, orally, six times a week); and (iv) the vehicle (water) alone. Therapeutic efficacy was evaluated in terms of survival rate, ratio of insulin‐positive β‐cells/total islet area, extent of islet inflammation (insulitis score) and blood‐glucose level. Results: The therapeutic administration of FTY720 plus sitagliptin significantly improved survival (83% at 70 days after onset, P < 0.05) compared with sitagliptin alone (17%) or vehicle alone (0%). The fasting‐blood glucose level, the ratio of insulin‐positive β‐cells/total islet area and the insulitis score in the surviving mice, which had been treated with FTY720 plus sitagliptin, were improved to the normal levels as in age‐matched NOD mice with normoglycemia. Conclusions: Combination therapy with FTY720 and sitagliptin is a promising candidate for type 1 diabetes mellitus treatment, and might allow the treatment of type 1 diabetes mellitus with only oral agents. (J Diabetes Invest, doi: 10.1111/j.2040‐1124.2012.00218.x, 2012)
Collapse
Affiliation(s)
- Takumi Tsuji
- Department of Pathological Biochemistry, Faculty of Pharmaceutical Sciences, Setsunan University, Osaka
| | - Yuya Yoshida
- Department of Pathological Biochemistry, Faculty of Pharmaceutical Sciences, Setsunan University, Osaka
| | - Tetsuro Fujita
- Research Institute for Production and Development, Kyoto, Japan
| | - Takeyuki Kohno
- Department of Pathological Biochemistry, Faculty of Pharmaceutical Sciences, Setsunan University, Osaka ; Research Institute for Production and Development, Kyoto, Japan
| |
Collapse
|
26
|
Tsuji T, Inoue M, Yoshida Y, Fujita T, Kaino Y, Kohno T. Therapeutic approach for type 1 diabetes mellitus using the novel immunomodulator FTY720 (fingolimod) in combination with once-daily injection of insulin glargine in non-obese diabetic mice. J Diabetes Investig 2014; 3:132-7. [PMID: 24843556 PMCID: PMC4020730 DOI: 10.1111/j.2040-1124.2011.00160.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Aims/Introduction: The therapeutic effectiveness against type 1 diabetes mellitus (DM) of the novel immunomodulator FTY720 (fingolimod), alone and in combination with insulin glargine, was examined in the non‐obese diabetic (NOD) mouse model. Materials and Methods: Female NOD mice that had developed DM spontaneously were divided into four groups: (i) an FTY720 (0.1 mg/kg, p.o., twice weekly)‐treated group; (ii) an insulin glargine (1.0 IU, s.c., once daily)‐treated group; (iii) a combination FTY720 + insulin glargine (0.1–1.0 IU, s.c., once daily)‐treated group; and (iv) a placebo (vehicle)‐treated group. Treatment was initiated at the time of onset of DM and continued for 70 days or until death. The therapeutic efficacy of FTY720, insulin glargine and FTY720 + insulin glargine was evaluated by measuring the ratio of insulin‐positive β‐cells/total islet area, the extent of islet inflammation (insulitis score), blood glucose levels, and serum C‐peptide levels. Results: Therapeutic administration of FTY720 to NOD mice with hyperglycemia (i.e. overt DM) significantly prolonged survival (P < 0.05 vs placebo). In the placebo group, all mice died within 63 days on the onset of DM; in contrast, 45% of FTY720‐treated mice survived during the observation period (up to 70 days after the onset of DM). Therapeutic administration of FTY720 in combination with insulin glargine to NOD mice with hyperglycemia further improved survival (P < 0.05) compared with either FTY720 or insulin glargine alone (i.e. 85% of FTY720 + insulin glargine‐treated mice survived to the end of the observation period). The efficacy of FTY720 in combination with insulin glargine was confirmed by histochemical, immunohistochemical and endocrinologic observations. Conclusions: Combination therapy with FTY720 plus insulin glargine is a promising candidate for the treatment of DM and may allow for a reduction in the frequency of insulin self‐injections. (J Diabetes Invest, doi:10.1111/j.2040‐1124.2011.00160.x, 2011)
Collapse
Affiliation(s)
- Takumi Tsuji
- Department of Pathological Biochemistry, Faculty of Pharmaceutical Sciences, Setsunan University, Osaka
| | - Mariko Inoue
- Department of Pathological Biochemistry, Faculty of Pharmaceutical Sciences, Setsunan University, Osaka
| | - Yuya Yoshida
- Department of Pathological Biochemistry, Faculty of Pharmaceutical Sciences, Setsunan University, Osaka
| | - Tetsuro Fujita
- Research Institute for Production and Development, Kyoto
| | | | - Takeyuki Kohno
- Department of Pathological Biochemistry, Faculty of Pharmaceutical Sciences, Setsunan University, Osaka
| |
Collapse
|
27
|
1,25-Dihydroxyvitamin D3 selectively and reversibly impairs T helper-cell CNS localization. Proc Natl Acad Sci U S A 2013; 110:21101-6. [PMID: 24324134 DOI: 10.1073/pnas.1306072110] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Pharmacologic targeting of T helper (TH) cell trafficking poses an attractive opportunity for amelioration of autoimmune diseases such as multiple sclerosis (MS). MS risk is associated with vitamin D deficiency, and its bioactive form, 1,25-dihydroxyvitamin D3 [1,25(OH)2D3], has been shown to prevent experimental autoimmune encephalomyelitis, a mouse model of MS, via an incompletely understood mechanism. Herein, we systematically examined 1,25(OH)2D3 effects on TH cells during their migration from the lymph nodes to the CNS. Our data demonstrate that myelin-reactive TH cells are successfully generated in the presence of 1,25(OH)2D3, secrete proinflammatory cytokines, and do not preferentially differentiate into suppressor T cells. These cells are able to leave the lymph node, enter the peripheral circulation, and migrate to the s.c. immunization sites. However, TH cells from 1,25(OH)2D3-treated mice are unable to enter the CNS parenchyma but are instead maintained in the periphery. Upon treatment cessation, mice rapidly develop experimental autoimmune encephalomyelitis, demonstrating that 1,25(OH)2D3 prevents the disease only temporarily likely by halting TH cell migration into the CNS.
Collapse
|
28
|
Bigaud M, Guerini D, Billich A, Bassilana F, Brinkmann V. Second generation S1P pathway modulators: research strategies and clinical developments. Biochim Biophys Acta Mol Cell Biol Lipids 2013; 1841:745-58. [PMID: 24239768 DOI: 10.1016/j.bbalip.2013.11.001] [Citation(s) in RCA: 97] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2013] [Revised: 10/30/2013] [Accepted: 11/04/2013] [Indexed: 11/17/2022]
Abstract
Multiple Sclerosis (MS) is a chronic autoimmune disorder affecting the central nervous system (CNS) through demyelination and neurodegeneration. Until recently, major therapeutic treatments have relied on agents requiring injection delivery. In September 2010, fingolimod/FTY720 (Gilenya, Novartis) was approved as the first oral treatment for relapsing forms of MS. Fingolimod causes down-modulation of S1P1 receptors on lymphocytes which prevents the invasion of autoaggressive T cells into the CNS. In astrocytes, down-modulation of S1P1 by the drug reduces astrogliosis, a hallmark of MS, thereby allowing restoration of productive astrocyte communication with other neural cells and the blood brain barrier. Animal data further suggest that the drug directly supports the recovery of nerve conduction and remyelination. In human MS, such mechanisms may explain the significant decrease in the number of inflammatory markers on brain magnetic resonance imaging in recent clinical trials, and the reduction of brain atrophy by the drug. Fingolimod binds to 4 of the 5 known S1P receptor subtypes, and significant efforts were made over the past 5 years to develop next generation S1P receptor modulators and determine the minimal receptor selectivity needed for maximal therapeutic efficacy in MS patients. Other approaches considered were competitive antagonists of the S1P1 receptor, inhibitors of the S1P lyase to prevent S1P degradation, and anti-S1P antibodies. Below we discuss the current status of the field, and the functional properties of the most advanced compounds. This article is part of a Special Issue entitled New Frontiers in Sphingolipid Biology.
Collapse
Affiliation(s)
- Marc Bigaud
- Novartis Institutes for Biomedical Research, CH-4056 Basel, Switzerland.
| | - Danilo Guerini
- Novartis Institutes for Biomedical Research, CH-4056 Basel, Switzerland
| | - Andreas Billich
- Novartis Institutes for Biomedical Research, CH-4056 Basel, Switzerland
| | | | - Volker Brinkmann
- Novartis Institutes for Biomedical Research, CH-4056 Basel, Switzerland.
| |
Collapse
|
29
|
Therapeutic use of a selective S1P1 receptor modulator ponesimod in autoimmune diabetes. PLoS One 2013; 8:e77296. [PMID: 24204793 PMCID: PMC3811978 DOI: 10.1371/journal.pone.0077296] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2013] [Accepted: 08/30/2013] [Indexed: 12/27/2022] Open
Abstract
In the present study, we investigated the therapeutic potential of a selective S1P1 receptor modulator, ponesimod, to protect and reverse autoimmune diabetes in non-obese diabetic (NOD) mice. Ponesimod was administered orally to NOD mice starting at 6, 10, 13 and 16 weeks of age up to 35 weeks of age or to NOD mice showing recent onset diabetes. Peripheral blood and spleen B and T cell counts were significantly reduced after ponesimod administration. In pancreatic lymph nodes, B lymphocytes were increased and expressed a transitional 1-like phenotype. Chronic oral ponesimod treatment efficiently prevented autoimmune diabetes in 6, 10 and 16 week-old pre-diabetic NOD mice. Treatment withdrawal led to synchronized disease relapse. Ponesimod did not inhibit the differentiation of autoreactive T cells as assessed by adoptive transfer of lymphocytes from treated disease-free NOD mice. In addition, it did not affect the migration, proliferation and activation of transgenic BDC2.5 cells into the target tissue. However, ponesimod inhibited spreading of the T cell responses to islet-specific glucose-6-phosphatase catalytic subunit-related protein (IGRP). Treatment of diabetic NOD mice with ponesimod induced disease remission. However, here again, upon treatment cessation, the disease rapidly recurred. This recurrence was effectively prevented by combination treatment with a CD3 antibody leading to the restoration of self-tolerance. In conclusion, treatment with a selective S1P1 modulator in combination with CD3 antibody represents a promising therapeutic approach for the treatment of autoimmune diabetes.
Collapse
|
30
|
Tsuiki A, Luo W, Henning T, Vishwanathan S, Dinh C, Adams D, Sweeney E, Mitchell J, Bachman S, Sharma P, Powell N, Hendry M, McNicholl J, Kersh E. Evaluation of the lymphocyte trafficking drug FTY720 in vaginal tissues. J Med Primatol 2013; 42:89-100. [PMID: 23311598 DOI: 10.1111/jmp.12033] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/10/2012] [Indexed: 11/27/2022]
Abstract
BACKGROUND FTY720 is an immunomodulatory agent that reduces lymphocytes in peripheral tissues and circulation. Such agents may be effective as vaginal microbicides for HIV prevention. Systemic or vaginal application of FTY720 may reduce lymphocyte concentrations in genital tissues, reducing HIV target cell numbers. METHODS Five female pigtail macaques received topical vaginal gel FTY720 (n = 2), intravenous (i.v.) FTY720 (n = 2), or placebo gel (n = 1) in this pilot study. Circulating and mucosal lymphocytes and genital mucosa, cytokines, and tissue histology were analyzed to document topical and i.v. FTY720 effects. RESULTS Topical and i.v. FTY720 appeared to decrease the levels of cervicovaginal IL-8, IL-1ra, and genital inflammatory cells. Small sample size precluded statistical analysis. Topical administration had no overt adverse effects. CONCLUSIONS This study introduces FTY720 as an immunomodulatory agent for the vaginal mucosa, compares topical effects to those of i.v. administration, and provides the basis for future studies involving FTY720 for HIV prevention.
Collapse
Affiliation(s)
- Ai Tsuiki
- Division of Scientific Resources, NCEZID, CDC, Atlanta, GA 30333, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Abstract
OBJECTIVES This study aimed at T-cell inhibition by immunosuppressants to reduce cell damage and improve the course of severe acute pancreatitis (SAP). METHODS A taurocholate-induced SAP was used and 5 groups were compared: (1) rapamycin + FTY720, (2) rapamycin, (3) FTY720, (4) cortisol, and (5) control: sodium chloride. Drugs were applied intravenously at SAP induction; 6 hours later, rats were killed. Interleukin (IL)-1, IL-6, IL-10, tumor necrosis factor α, platelet-activating factor, amylase, and lipase were measured in serum and myeloperoxidase tissue activity in pancreas, kidney, lung, liver, and spleen. Edema, inflammation, and necrosis were histologically determined in pancreas. CD4/CD8 immunohistochemistry was performed. RESULTS Inflammation was ameliorated in all 4 treated groups. Necrosis development was suppressed by FTY720, FTY720 + rapamycin, and cortisol. IL-6 and IL-10 were significantly lower in these groups. Amylase was higher in all treatment groups compared to the controls except for the cortisol group. Tumor necrosis factor α, lipase, and myeloperoxidase activity were not affected by therapy. CD4+/CD8+ cells were significantly less in FTY720-treated pancreata. CONCLUSION Rapamycin and FTY720 ameliorated the severity of SAP, which may be due to early suppression of helper T cells. FTY720 reduced the development of pancreatic necrosis. The combination of both immunosuppressants did not show advantage to treatment with FTY720 alone.
Collapse
|
32
|
Discovery of fingolimod, the sphingosine 1-phosphate receptor modulator and its application for the therapy of multiple sclerosis. Future Med Chem 2012; 4:771-81. [PMID: 22530640 DOI: 10.4155/fmc.12.25] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Fingolimod (FTY720) is a first-in-class, orally active, sphingosine 1-phosphate (S1P)-receptor modulator with a structure closely related to sphingosine. The compound was discovered by chemical modification of a natural product, myriocin. Phosphorylated form of FTY720 acts as a functional antagonist at S1P receptor type 1 (S1P(1)), inhibits lymphocyte egress from secondary lymphoid organs and shows immunomodulating effects. Phase III studies in multiple sclerosis demonstrated that oral FTY720 had superior efficacy compared with intramuscular IFN-β1a (AVONEX(®)) with regard to reducing the rate of relapse and the number of inflammatory lesions in the CNS. FTY720 has been approved as a new therapeutic drug for multiple sclerosis in more than 50 countries, including the USA, Japan and some of those in the EU.
Collapse
|
33
|
Chiba K, Yoshii N. [Pharmacological properties and clinical efficacy of fingolimod hydrochloride (Imusera®/Gilenya®) for the treatment of multiple sclerosis]. Nihon Yakurigaku Zasshi 2012; 139:265-74. [PMID: 22728990 DOI: 10.1254/fpj.139.265] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
|
34
|
Karliner JS. Sphingosine kinase and sphingosine 1-phosphate in the heart: a decade of progress. Biochim Biophys Acta Mol Cell Biol Lipids 2012; 1831:203-12. [PMID: 22735359 DOI: 10.1016/j.bbalip.2012.06.006] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2012] [Revised: 06/14/2012] [Accepted: 06/17/2012] [Indexed: 12/11/2022]
Abstract
Activation of sphingosine kinase/sphingosine 1-phosphate (SK/S1P)-mediated signaling has emerged as a critical cardioprotective pathway in response to acute ischemia/reperfusion injury. S1P is released in both ischemic pre- and post-conditioning. Application of exogenous S1P to cultured cardiac myocytes subjected to hypoxia or treatment of isolated hearts either before ischemia or at the onset of reperfusion exerts prosurvival effects. Synthetic congeners of S1P such as FTY720 mimic these responses. Gene targeted mice null for the SK1 isoform whose hearts are subjected to ischemia/reperfusion injury exhibit increased infarct size and respond poorly either to ischemic pre- or postconditioning. Measurements of cardiac SK activity and S1P parallel these observations. Experiments in SK2 knockout mice have revealed that this isoform is necessary for survival in the heart. High density lipoprotein (HDL) is a major carrier of S1P, and studies of hearts in which selected S1P receptors have been inhibited implicate the S1P cargo of HDL in cardioprotection. Inhibition of S1P lyase, an endogenous enzyme that degrades S1P, also leads to cardioprotection. These observations have considerable relevance for future therapeutic approaches to acute and chronic myocardial injury. This article is part of a Special Issue entitled Advances in Lysophospholipid Research.
Collapse
|
35
|
Sphingosine 1-phosphate receptor 1 as a useful target for treatment of multiple sclerosis. Pharmaceuticals (Basel) 2012; 5:514-28. [PMID: 24281561 PMCID: PMC3763654 DOI: 10.3390/ph5050514] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2012] [Revised: 04/24/2012] [Accepted: 05/15/2012] [Indexed: 12/20/2022] Open
Abstract
Sphingosine 1-phosphate (S1P), a lysophospholipid mediator, is generated from sphingosine by sphingosine kinases and binds five known cell surface receptors. S1P receptor 1 (S1P1) plays an essential role in lymphocyte egress from secondary lymphoid organs (SLO), as evinced by the inability of lymphocytes to exit from the SLO in mice lacking lymphocytic S1P1. Fingolimod hydrochloride (FTY720) is a first-in-class, orally active, S1P receptor modulator with a structure closely related to sphingosine. FTY720 was first synthesized by chemical modification of a natural product, myriocin. FTY720 is effectively converted to an active metabolite, FTY720 phosphate (FTY720-P) by sphingosine kinases. FTY720-P shows high affinity to 4 of the S1P receptors (S1P1, S1P3, S1P4, and S1P5). In particular, FTY720-P strongly induces internalization and degradation of S1P1, inhibits S1P responsiveness of lymphocytes in the SLO, and acts as a functional antagonist at lymphocytic S1P1. Consequently, FTY720 inhibits S1P1-dependent lymphocyte egress from the SLO to decrease circulation of lymphocytes including autoreactive Th17 cells and is highly effective in experimental autoimmune encephalomyelitis (EAE), an animal model of multiple sclerosis (MS). Because FTY720 shows a superior efficacy in relapsing remitting MS patients compared to intramuscular interferon-β-1a (Avonex®), S1P1 is presumed to be a useful target for the therapy of MS.
Collapse
|
36
|
Emmerich J, Mumm JB, Chan IH, LaFace D, Truong H, McClanahan T, Gorman DM, Oft M. IL-10 Directly Activates and Expands Tumor-Resident CD8+ T Cells without De Novo Infiltration from Secondary Lymphoid Organs. Cancer Res 2012; 72:3570-81. [DOI: 10.1158/0008-5472.can-12-0721] [Citation(s) in RCA: 182] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
37
|
Strader CR, Pearce CJ, Oberlies NH. Fingolimod (FTY720): a recently approved multiple sclerosis drug based on a fungal secondary metabolite. JOURNAL OF NATURAL PRODUCTS 2011; 74:900-907. [PMID: 21456524 DOI: 10.1021/np2000528] [Citation(s) in RCA: 133] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/28/2023]
Abstract
Fingolimod (Gilenya; FTY720), a synthetic compound based on the fungal secondary metabolite myriocin (ISP-I), is a potent immunosuppressant that was approved (September 2010) by the U.S. FDA as a new treatment for multiple sclerosis (MS). Fingolimod was synthesized by the research group of Tetsuro Fujita at Kyoto University in 1992 while investigating structure-activity relationships of derivatives of the fungal metabolite ISP-I, isolated from Isaria sinclairii. Fingolimod becomes active in vivo following phosphorylation by sphingosine kinase 2 to form fingolimod-phosphate, which binds to extracellular G protein-coupled receptors, sphingosine 1-phosphates, and prevents the release of lymphocytes from lymphoid tissue. Fingolimod is orally active, which is unique among current first-line MS therapies, and it has the potential to be used in the treatment of organ transplants and cancer. This review highlights the discovery and development of fingolimod, from an isolated lead natural product, through synthetic analogues, to an approved drug.
Collapse
Affiliation(s)
- Cherilyn R Strader
- Department of Chemistry and Biochemistry, University of North Carolina at Greensboro, P.O. Box 26170, Greensboro, North Carolina 27402-6170, USA
| | | | | |
Collapse
|
38
|
Bradaric MJ, Barua A, Penumatsa K, Yi Y, Edassery SL, Sharma S, Abramowicz JS, Bahr JM, Luborsky JL. Sphingosine-1 phosphate receptor (S1p1), a critical receptor controlling human lymphocyte trafficking, is expressed in hen and human ovaries and ovarian tumors. J Ovarian Res 2011; 4:4. [PMID: 21356044 PMCID: PMC3058111 DOI: 10.1186/1757-2215-4-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2010] [Accepted: 02/28/2011] [Indexed: 01/05/2023] Open
Abstract
Background Sphingosine-1 receptor 1 (S1P1) plays a major role in regulating lymphocyte egress from peripheral lymph tissue. Lymphocyte trafficking is potentially a critical response to tumors and to tumor vaccines. Also, the receptor has been shown to influence metastasis. However, there is little information on its expression in the aged ovary or ovarian tumors. As a basis for further studies in the laying hen model of spontaneous ovarian cancer, the objective of this study was to determine if S1P1 is expressed in hens, and if the morphological distribution of S1P1 is similar in hen and human ovary and ovarian tumors. Methods S1P1 mRNA was ascertained in hen tissue by RT-PCR using hen specific primers. S1P1 protein expression and localization was evaluated in hen and human tissue with a human S1P1 antibody by Western blot and immunohistochemistry. Results S1P1 mRNA was expressed in all hen tissues examined. Protein was detected in human and hen ovary and ovarian tumors at 47, 72 and 108 kDa in Western blots. S1P1 was similarly expressed on endothelial cells, lymphocytes and surface epithelial cells in normal ovaries and tumor-containing ovaries of the hen. In addition, S1P1 distribution was heterogeneous in both hen and human ovarian tumors by immunohistochemistry. Conclusion The results show that S1P1 is expressed in the hen and human ovary as well as in ovarian tumors. These findings support the use of the hen in further studies of the role of S1P1 in metastasis and immune cell trafficking in ovarian tumor development.
Collapse
Affiliation(s)
- Michael J Bradaric
- Department of Pharmacology, Rush University Medical Center, Chicago, IL USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Yoshida Y, Tsuji T, Fujita T, Kohno T. Relapse of Experimental Autoimmune Encephalomyelitis after Discontinuation of FTY720 (Fingolimod) Treatment, but Not after Combination of FTY720 and Pathogenic Autoantigen. Biol Pharm Bull 2011; 34:933-6. [DOI: 10.1248/bpb.34.933] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Yuya Yoshida
- Department of Pathological Biochemistry, Faculty of Pharmaceutical Sciences, Setsunan University
| | - Takumi Tsuji
- Department of Pathological Biochemistry, Faculty of Pharmaceutical Sciences, Setsunan University
| | | | - Takeyuki Kohno
- Department of Pathological Biochemistry, Faculty of Pharmaceutical Sciences, Setsunan University
- Research Institute for Production and Development
| |
Collapse
|
40
|
Chiba K, Kataoka H, Seki N, Maeda Y, Sugahara K. Fingolimod (FTY720), the Sphingosine 1-Phosphate Receptor Modulator, as a New Therapeutic Drug in Multiple Sclerosis. Inflamm Regen 2011. [DOI: 10.2492/inflammregen.31.167] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
|
41
|
Fingolimod (FTY720): discovery and development of an oral drug to treat multiple sclerosis. Nat Rev Drug Discov 2010; 9:883-97. [PMID: 21031003 DOI: 10.1038/nrd3248] [Citation(s) in RCA: 996] [Impact Index Per Article: 66.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The discovery of fingolimod (FTY720/Gilenya; Novartis), an orally active immunomodulatory drug, has opened up new approaches to the treatment of multiple sclerosis, the most common inflammatory disorder of the central nervous system. Elucidation of the effects of fingolimod--mediated by the modulation of sphingosine 1-phosphate (S1P) receptors--has indicated that its therapeutic activity could be due to regulation of the migration of selected lymphocyte subsets into the central nervous system and direct effects on neural cells, particularly astrocytes. An improved understanding of the biology of S1P receptors has also been gained. This article describes the discovery and development of fingolimod, which was approved by the US Food and Drug Administration in September 2010 as a first-line treatment for relapsing forms of multiple sclerosis, thereby becoming the first oral disease-modifying therapy to be approved for multiple sclerosis in the United States.
Collapse
|
42
|
Horga A, Castilló J, Montalban X. Fingolimod for relapsing multiple sclerosis: an update. Expert Opin Pharmacother 2010; 11:1183-96. [DOI: 10.1517/14656561003769866] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
43
|
Bajwa A, Jo SK, Ye H, Huang L, Dondeti KR, Rosin DL, Haase VH, Macdonald TL, Lynch KR, Okusa MD. Activation of sphingosine-1-phosphate 1 receptor in the proximal tubule protects against ischemia-reperfusion injury. J Am Soc Nephrol 2010; 21:955-65. [PMID: 20338995 DOI: 10.1681/asn.2009060662] [Citation(s) in RCA: 94] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
Agonists of the sphingosine-1-phosphate receptor (S1PR) attenuate kidney ischemia-reperfusion injury (IRI). Previous studies suggested that S1P1R-induced lymphopenia mediates this protective effect, but lymphocyte-independent mechanisms could also contribute. Here, we investigated the effects of S1PR agonists on kidney IRI in mice that lack T and B lymphocytes (Rag-1 knockout mice). Administration of the nonselective S1PR agonist FTY720 or the selective S1P1R agonist SEW2871 reduced injury in both Rag-1 knockout and wild-type mice. In vitro, SEW2871 significantly attenuated LPS- or hypoxia/reoxygenation-induced apoptosis in cultured mouse proximal tubule epithelial cells, supporting a direct protective effect of S1P1R agonists via mitogen-activated protein kinase and/or Akt pathways. S1P1Rs in the proximal tubule mediated IRI in vivo as well: Mice deficient in proximal tubule S1P1Rs experienced a greater decline in renal function after IRI than control mice and their kidneys were no longer protected by SEW2871 administration. In summary, S1PRs in the proximal tubule are necessary for stress-induced cell survival, and S1P1R agonists are renoprotective via direct effects on the tubule cells. Selective agonists of S1P1Rs may hold therapeutic potential for the prevention and treatment of acute kidney injury.
Collapse
Affiliation(s)
- Amandeep Bajwa
- Department of Medicine, Division of Nephrology, Box 800133, University of Virginia Health System, Charlottesville, VA 22908, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Lee KD, Chow WN, Sato-Bigbee C, Graf MR, Graham RS, Colello RJ, Young HF, Mathern BE. FTY720 reduces inflammation and promotes functional recovery after spinal cord injury. J Neurotrauma 2010; 26:2335-44. [PMID: 19624262 DOI: 10.1089/neu.2008.0840] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
A robust and complex inflammatory cascade is known to be a prominent component of secondary injury following spinal cord injury (SCI). Specifically, the concept of trauma-induced autoimmunity has linked the lymphocyte population with neural tissue injury and neurologic deficit. FTY720, a sphingosine receptor modulator that sequesters lymphocytes in secondary lymphoid organs, has been shown to be effective in the treatment of a variety of experimental autoimmune disorders. Accordingly, by reducing lymphocyte infiltration into the spinal cord following SCI, this novel immunomodulator may enhance tissue preservation and functional recovery. In the present study, a moderate to severe contusion SCI was simulated in adult Long-Evans hooded rats. Using flow cytometry we showed that daily FTY720 treatment dramatically reduced T-cell infiltration into the SCI lesion site at 4 and 7 days post-injury, while other inflammatory cell populations were relatively unaltered. To assess functional recovery, three groups of injured animals (treated, vehicle, and injury only) were evaluated weekly for hindlimb recovery. Animals in the treated group consistently exhibited higher functional scores than animals in the control groups after 2 weeks post-injury. This finding was associated with a greater degree of white matter sparing at the lesion epicenter when cords were later sectioned and stained. Furthermore, treated animals were found to exhibit improved bladder function and a reduced incidence of hemorrhagic cystitis compared to control counterparts. Collectively these results demonstrate the neuroprotective potential of FTY720 treatment after experimental SCI.
Collapse
Affiliation(s)
- Kangmin D Lee
- Department of Neurosurgery, Virginia Commonwealth University, Richmond, Virginia, USA
| | | | | | | | | | | | | | | |
Collapse
|
45
|
|
46
|
Chiba K. Sphingosine 1-phosphate receptor type 1 as a novel target for the therapy of autoimmune diseases. Inflamm Regen 2010. [DOI: 10.2492/inflammregen.30.160] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
|
47
|
Hwang SJ, Kim JH, Kim HY, Kim S, Chung DH. FTY720, a sphingosine 1-phosphate receptor modulator, inhibits CD1d-restricted NKT cells by suppressing cytokine production but not migration. J Transl Med 2010; 90:9-19. [PMID: 19823172 DOI: 10.1038/labinvest.2009.109] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
FTY720, a sphingosine 1-phosphate (S1P) receptor modulator, suppresses immune responses by inhibiting T-cell migration into target tissues; however, it does not alter T-cell functions. In this study, we investigated the biological effects of FTY720 on NKT cells. Unlike T cells, FTY720 suppressed the production of IL-4, IFN-gamma, IL-10, and IL-13 by NKT cells through the S1P1 receptor (S1P(1)). Moreover, FTY720 also inhibited the expression of T-bet and GATA-3 of NKT cells in the presence of TCR engagement. However, it did not inhibit NKT cell migration in vitro or in vivo. In a K/BxN serum transfer arthritis model, FTY720 suppressed arthritis in B6, but not in CD1d(-/-) mice. Moreover, the adoptive transfer of control NKT cells restored arthritis in CD1d(-/-) mice, whereas FTY720-pretreated NKT cells did not. The number of NKT cells in the joints of B6 mice given FTY720 was similar to that in the joints of untreated B6 mice, whereas the production of IL-4 and IFN-gamma was reduced in the FTY720-treated B6 mice. Taken together, these data show that FTY720 suppresses cytokine production in NKT cells through S1P(1), but not NKT cell migration. Thus, FTY720 may be useful in the treatment of NKT cell-promoted immune diseases.
Collapse
Affiliation(s)
- Su Jin Hwang
- Department of Pathology, Seoul National University College of Medicine, Seoul 110-744, Korea
| | | | | | | | | |
Collapse
|
48
|
Chiba K, Kataoka H, Maeda Y, Seki N, Sugahara K. Sphingosine 1-phosphate receptor modulator, fingolimod (FTY720), provides a new therapeutic approach for autoimmune diseases. Inflamm Regen 2010. [DOI: 10.2492/inflammregen.30.419] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
|
49
|
Chiba K. [New therapeutic approach for autoimmune diseases by the sphingosine 1-phosphate receptor modulator, fingolimod (FTY720)]. YAKUGAKU ZASSHI 2009; 129:655-65. [PMID: 19483408 DOI: 10.1248/yakushi.129.655] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Fingolimod (FTY720) is the first substance in the new immunomodulator class called sphingosine 1-phosphate (S1P) receptor modulators. We isolated an immunosuppressive natural product, myrocin, from the culture broth of Isaria sinclairii, a kind of vegetative wasp. The chemical modification of myriocin yielded a new compound, FTY720, which has more potent immunosuppressive activity and less toxicity than myriocin. FTY720 has been shown to be highly effective in experimental allograft models and autoimmune disease models such as autoimmune encephalomyelitis, collagen-induced arthritis, and lupus nephritis. The most striking feature of FTY720 is the induction of a marked decrease in peripheral blood lymphocytes at doses that show immunosuppressive activity in these models. FTY720 is rapidly converted to FTY720-phosphate (FTY720-P) by sphingosine kinases. FTY720-P acts as a potent agonist at S1P receptor type 1 (S1P(1)), internalizes S1P(1) on lymphocytes, and inhibits the migration of lymphocytes toward S1P. It is highly likely that the reduction of peripheral blood lymphocytes by FTY720 is due to the inhibition of S1P(1)-dependent lymphocyte egress from secondary lymphoid organs and thymus. Recently, it has been reported that FTY720 exerted considerable therapeutic effects in a placebo-controlled clinical trail involving patients with relapsing multiple sclerosis. Patients who received FTY720 orally had a significant reduction in the clinical disease activity, the number of lesions in the central nervous system, and the relapse rates. Since FTY720 possesses a new mechanism of action that has not been observed with other immunosuppressive agents, it is believed that FTY720 provides a new therapeutic approach for autoimmune diseases including multiple sclerosis.
Collapse
Affiliation(s)
- Kenji Chiba
- Pharmacology Laboratory, Research Division, Mitsubishi Tanabe Pharma Corporation, Yokohama, Japan.
| |
Collapse
|
50
|
Rosen H, Gonzalez-Cabrera PJ, Sanna MG, Brown S. Sphingosine 1-phosphate receptor signaling. Annu Rev Biochem 2009; 78:743-68. [PMID: 19231986 DOI: 10.1146/annurev.biochem.78.072407.103733] [Citation(s) in RCA: 327] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The sphingosine 1-phosphate (S1P) receptor signaling system is a productive model system. A hydrophobic zwitterionic lysophospholipid ligand with difficult physical properties interacts with five high-affinity G protein-coupled receptors to generate multiple downstream signals. These signals modulate homeostasis and pathology on a steep agonist concentration-response curve. Ligand presence is essential for vascular development and endothelial integrity, while acute increases in ligand concentrations result in cardiac death. Understanding this integrated biochemical system has exemplified the impact of both genetics and chemistry. Developing specific tools with defined biochemical properties for the reversible modulation of signals in real time has been essential to complement insights gained from genetic approaches that may be irreversible and compensated. Despite its knife-edge between life and death, this system, based in part on receptor subtype-selectivity and in part on differential attenuation of deleterious signals, now appears to be on the cusp of meaningful therapy for multiple sclerosis.
Collapse
Affiliation(s)
- Hugh Rosen
- Departments of Chemical Physiology and Immunology and The Scripps Research Institute Molecular Screening Center, The Scripps Research Institute, La Jolla, CA 92037, USA.
| | | | | | | |
Collapse
|