Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
J Med Chem ; 66(15): 10381-10412, 2023 08 10.
Article in English | MEDLINE | ID: mdl-37489798

ABSTRACT

Because of the wide use of Fingolimod for the treatment of multiple sclerosis (MS) and its cardiovascular side effects such as bradycardia, second-generation sphingosine 1-phosphate receptor 1 (S1P1) agonist drugs for MS have been developed and approved by FDA. The issue of bradycardia is still present with the new drugs, however, which necessitates further exploration of S1P1 agonists with improved safety profiles for next-generation MS drugs. Herein, we report a tetrahydroisoquinoline or a benzo[c]azepine core-based S1P1 agonists such as 32 and 60 after systematic examination of hydrophilic groups and cores. We investigated the binding modes of our representative compounds and their molecular interactions with S1P1 employing recent S1P1 cryo-EM structures. Also, favorable ADME properties of our compounds were shown. Furthermore, in vivo efficacy of our compounds was clearly demonstrated with PLC and EAE studies. Also, the preliminary in vitro cardiovascular safety of our compound was verified with human iPSC-derived cardiomyocytes.


Subject(s)
Multiple Sclerosis , Tetrahydroisoquinolines , Humans , Multiple Sclerosis/drug therapy , Multiple Sclerosis/metabolism , Sphingosine-1-Phosphate Receptors , Bradycardia/chemically induced , Receptors, Lysosphingolipid/agonists , Receptors, Lysosphingolipid/metabolism , Receptors, Lysosphingolipid/therapeutic use , Fingolimod Hydrochloride/pharmacology , Fingolimod Hydrochloride/therapeutic use , Tetrahydroisoquinolines/therapeutic use , Sphingosine/metabolism
2.
ESC Heart Fail ; 10(1): 334-341, 2023 Feb.
Article in English | MEDLINE | ID: mdl-36217778

ABSTRACT

AIMS: Therapeutic options targeting post-ischaemic cardiac remodelling are sparse. The bioactive sphingolipid sphingosine-1-phosphate (S1P) reduces ischaemia/reperfusion injury. However, its impact on post-ischaemic remodelling independently of its infarct size (IS)-reducing effect is yet unknown and was addressed in this study. METHODS AND RESULTS: Acute myocardial infarction (AMI) in mice was induced by permanent ligation of the left anterior descending artery (LAD). C57Bl6 were treated with the S1P lyase inhibitor 4-deoxypyridoxine (DOP) starting 7 days prior to AMI to increase endogenous S1P concentrations. Cardiac function and myocardial healing were assessed by cardiovascular magnetic resonance imaging (cMRI), murine echocardiography, histomorphology, and gene expression analysis. DOP effects were investigated in cardiomyocyte-specific S1P receptor 1 deficient (S1PR1 Cardio Cre+) and Cre- control mice and S1P concentrations measured by LC-MS/MS. IS and cardiac function did not differ between control and DOP-treated groups on day one after LAD-ligation despite fourfold increase in plasma S1P. In contrast, cardiac function was clearly improved and myocardial scar size reduced, respectively, on Day 21 in DOP-treated mice. The latter also exhibited smaller cardiomyocyte size and reduced embryonic gene expression. The benefit of DOP treatment was abolished in S1PR1 Cardio Cre+. CONCLUSIONS: S1P improves cardiac function and myocardial healing post AMI independently of initial infarct size and accomplishes this via the cardiomyocyte S1PR1. Hence, in addition to its beneficial effects on I/R injury, S1PR1 may be a promising target in post-infarction myocardial remodelling as adjunctive therapy to revascularization as well as in patients not eligible for standard interventional procedures.


Subject(s)
Myocardial Infarction , Receptors, Lysosphingolipid , Mice , Animals , Sphingosine-1-Phosphate Receptors/therapeutic use , Chromatography, Liquid , Receptors, Lysosphingolipid/genetics , Receptors, Lysosphingolipid/metabolism , Receptors, Lysosphingolipid/therapeutic use , Tandem Mass Spectrometry , Myocardial Infarction/drug therapy
3.
Cell Mol Neurobiol ; 43(3): 1219-1236, 2023 Apr.
Article in English | MEDLINE | ID: mdl-35917044

ABSTRACT

Multiple sclerosis (MS) is an inflammatory-demyelinating disease of the central nervous system (CNS) mediated by aberrant auto-reactive immune responses. The current immune-modulatory therapies are unable to protect and repair immune-mediated neural tissue damage. One of the therapeutic targets in MS is the sphingosine-1-phosphate (S1P) pathway which signals via sphingosine-1-phosphate receptors 1-5 (S1P1-5). S1P receptors are expressed predominantly on immune and CNS cells. Considering the potential neuroprotective properties of S1P signaling, we utilized S1P1-GFP (Green fluorescent protein) reporter mice in the cuprizone-induced demyelination model to investigate in vivo S1P - S1P1 signaling in the CNS. We observed S1P1 signaling in a subset of neural stem cells in the subventricular zone (SVZ) during demyelination. During remyelination, S1P1 signaling is expressed in oligodendrocyte progenitor cells in the SVZ and mature oligodendrocytes in the medial corpus callosum (MCC). In the cuprizone model, we did not observe S1P1 signaling in neurons and astrocytes. We also observed ß-arrestin-dependent S1P1 signaling in lymphocytes during demyelination and CNS inflammation. Our findings reveal ß-arrestin-dependent S1P1 signaling in oligodendrocyte lineage cells implying a role of S1P1 signaling in remyelination.


Subject(s)
Multiple Sclerosis , Remyelination , Mice , Animals , Sphingosine-1-Phosphate Receptors/metabolism , Sphingosine-1-Phosphate Receptors/therapeutic use , Cuprizone , Receptors, Lysosphingolipid/metabolism , Receptors, Lysosphingolipid/therapeutic use , Central Nervous System/metabolism , Multiple Sclerosis/metabolism , Oligodendroglia/metabolism , beta-Arrestins/metabolism , beta-Arrestins/therapeutic use , Mice, Inbred C57BL
4.
Cells ; 11(13)2022 06 29.
Article in English | MEDLINE | ID: mdl-35805142

ABSTRACT

Sphingosine-1-phosphate (S1P) and S1P receptors (S1PR) are bioactive lipid molecules that are ubiquitously expressed in the human body and play an important role in the immune system. S1P-S1PR signaling has been well characterized in immune trafficking and activation in both innate and adaptive immune systems. Despite this knowledge, the full scope in the pathogenesis of autoimmune disorders is not well characterized yet. From the discovery of fingolimod, the first S1P modulator, until siponimod, the new molecule recently approved for the treatment of secondary progressive multiple sclerosis (SPMS), there has been a great advance in understanding the S1P functions and their involvement in immune diseases, including multiple sclerosis (MS). Modulation on S1P is an interesting target for the treatment of various autoimmune disorders. Improved understanding of the mechanism of action of fingolimod has allowed the development of the more selective second-generation S1PR modulators. Subtype 1 of the S1PR (S1PR1) is expressed on the cell surface of lymphocytes, which are known to play a major role in MS pathogenesis. The understanding of S1PR1's role facilitated the development of pharmacological strategies directed to this target, and theoretically reduced the safety concerns derived from the use of fingolimod. A great advance in the MS treatment was achieved in March 2019 when the Food and Drug Association (FDA) approved Siponimod, for both active secondary progressive MS and relapsing-remitting MS. Siponimod became the first oral disease modifying therapy (DMT) specifically approved for active forms of secondary progressive MS. Additionally, for the treatment of relapsing forms of MS, ozanimod was approved by FDA in March 2020. Currently, there are ongoing trials focused on other new-generation S1PR1 modulators. This review approaches the fundamental aspects of the sphingosine phosphate modulators and their main similarities and differences.


Subject(s)
Autoimmune Diseases , Multiple Sclerosis , Autoimmune Diseases/drug therapy , Fingolimod Hydrochloride/pharmacology , Fingolimod Hydrochloride/therapeutic use , Humans , Lysophospholipids , Multiple Sclerosis/drug therapy , Multiple Sclerosis/metabolism , Receptors, Lysosphingolipid/metabolism , Receptors, Lysosphingolipid/therapeutic use , Signal Transduction , Sphingosine/analogs & derivatives , Sphingosine/metabolism
5.
Nat Rev Rheumatol ; 18(6): 335-351, 2022 06.
Article in English | MEDLINE | ID: mdl-35508810

ABSTRACT

Sphingosine 1-phosphate (S1P), which acts via G protein-coupled S1P receptors (S1PRs), is a bioactive lipid essential for vascular integrity and lymphocyte trafficking. The S1P-S1PR signalling axis is a key component of the inflammatory response in autoimmune rheumatic diseases. Several drugs that target S1PRs have been approved for the treatment of multiple sclerosis and inflammatory bowel disease and are under clinical testing for patients with systemic lupus erythematosus (SLE). Preclinical studies support the hypothesis that targeting the S1P-S1PR axis would be beneficial to patients with SLE, rheumatoid arthritis (RA) and systemic sclerosis (SSc) by reducing pathological inflammation. Whereas most preclinical research and development efforts are focused on reducing lymphocyte trafficking, protective effects of circulating S1P on endothelial S1PRs, which maintain the vascular barrier and enable blood circulation while dampening leukocyte extravasation, have been largely overlooked. In this Review, we take a holistic view of S1P-S1PR signalling in lymphocyte and vascular pathobiology. We focus on the potential of S1PR modulators for the treatment of SLE, RA and SSc and summarize the rationale, pathobiology and evidence from preclinical models and clinical studies. Improved understanding of S1P pathobiology in autoimmune rheumatic diseases and S1PR therapeutic modulation is anticipated to lead to efficacious and safer management of these diseases.


Subject(s)
Lupus Erythematosus, Systemic , Multiple Sclerosis , Rheumatic Diseases , Humans , Multiple Sclerosis/drug therapy , Receptors, Lysosphingolipid/therapeutic use , Rheumatic Diseases/drug therapy , Signal Transduction , Sphingosine-1-Phosphate Receptors
6.
Adv Sci (Weinh) ; 9(14): e2104452, 2022 05.
Article in English | MEDLINE | ID: mdl-35289120

ABSTRACT

SPHK1 (sphingosine kinase-1) catalyzes the phosphorylation of sphingosine to sphingosine-1-phosphate (S1P), is found to be highly expressed in solid tumors. Here, extracellular vesicles (EVs) are identified as the key transporters of SPHK1 to the tumor microenvironment. Consequently, SPHK1-packaged EVs elevate S1P levels in the tumor microenvironment, where S1P appears as an immunosuppressive agent. However, the exact mechanism of how S1P mediates its immunosuppressive effects in cancer is not understood. It is investigated that S1P can induce T cell exhaustion. S1P can also upregulate programmed death ligand-1 (PDL-1) expression through E2F1-mediated transcription. Notably, an SPHK1 inhibitor PF543 improves T cell-mediated cytotoxicity. Furthermore, combining PF543 with an anti-PD-1 antibody reduces tumor burden and metastasis more effectively than PF543 alone in vivo. These data demonstrate a previously unrecognized mechanism of how SPHK1-packaged EVs contribute to the progression of ovarian cancer and thus present the potential clinical application of inhibiting SPHK1/S1P signaling to improve immune checkpoint blockage (anti-PD-1 antibody) therapy in ovarian cancer.


Subject(s)
Extracellular Vesicles , Ovarian Neoplasms , Carcinoma, Ovarian Epithelial , Extracellular Vesicles/metabolism , Female , Humans , Immunotherapy , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/metabolism , Receptors, Lysosphingolipid/metabolism , Receptors, Lysosphingolipid/therapeutic use , Sphingosine/metabolism , Sphingosine/therapeutic use , T-Lymphocytes/metabolism , T-Lymphocytes/pathology , Tumor Microenvironment
7.
J Am Soc Nephrol ; 27(11): 3383-3393, 2016 Nov.
Article in English | MEDLINE | ID: mdl-26961351

ABSTRACT

Epithelial and endothelial injury and a cascade of immune and interstitial cell activation in the kidney lead to AKI. After mild to moderate AKI, the epithelium can regenerate and restore kidney function, yet little is known about the endothelium during these repair processes. Sphingosine 1-phosphate receptor 1 (S1P1), a G protein-coupled receptor, is necessary for vascular homeostasis. Here, we used an inducible genetic approach in a mouse model of AKI, ischemia-reperfusion injury (IRI), to determine the temporal effects of endothelial S1P1 during AKI. Deletion of endothelial S1P1 before IRI exacerbated kidney injury and inflammation, and the delayed deletion of S1P1 after IRI prevented kidney recovery, resulting in chronic inflammation and progressive fibrosis. Specifically, S1P1 directly suppressed endothelial activation of leukocyte adhesion molecule expression and inflammation. Altogether, the data indicate activation of endothelial S1P1 is necessary to protect from IRI and permit recovery from AKI. Endothelial S1P1 may be a therapeutic target for the prevention of early injury as well as prevention of progressive kidney fibrosis after AKI.


Subject(s)
Acute Kidney Injury/prevention & control , Receptors, Lysosphingolipid/physiology , Receptors, Lysosphingolipid/therapeutic use , Animals , Endothelium , Fibrosis/prevention & control , Kidney/blood supply , Kidney/pathology , Male , Mice , Mice, Inbred BALB C , Recovery of Function , Reperfusion Injury/prevention & control
8.
Cell Mol Immunol ; 13(1): 3-10, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26189369

ABSTRACT

Severe influenza remains unusual in its virulence for humans. Complications or ultimately death arising from these infections are often associated with hyperinduction of proinflammatory cytokine production, which is also known as 'cytokine storm'. For this disease, it has been proposed that immunomodulatory therapy may improve the outcome, with or without the combination of antiviral agents. Here, we review the current literature on how various effectors of the immune system initiate the cytokine storm and exacerbate pathological damage in hosts. We also review some of the current immunomodulatory strategies for the treatment of cytokine storms in severe influenza, including corticosteroids, peroxisome proliferator-activated receptor agonists, sphingosine-1-phosphate receptor 1 agonists, cyclooxygenase-2 inhibitors, antioxidants, anti-tumour-necrosis factor therapy, intravenous immunoglobulin therapy, statins, arbidol, herbs, and other potential therapeutic strategies.


Subject(s)
Antiviral Agents/therapeutic use , Cytokines/antagonists & inhibitors , Immunologic Factors/therapeutic use , Immunomodulation , Influenza, Human/drug therapy , Orthomyxoviridae/drug effects , Adrenal Cortex Hormones/therapeutic use , Antioxidants/therapeutic use , Cyclooxygenase 2/genetics , Cyclooxygenase 2/immunology , Cyclooxygenase Inhibitors/therapeutic use , Cytokines/genetics , Cytokines/immunology , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Immunoglobulins, Intravenous/therapeutic use , Indoles/therapeutic use , Influenza, Human/genetics , Influenza, Human/immunology , Influenza, Human/virology , Orthomyxoviridae/immunology , Peroxisome Proliferator-Activated Receptors/antagonists & inhibitors , Peroxisome Proliferator-Activated Receptors/genetics , Peroxisome Proliferator-Activated Receptors/immunology , Plant Preparations/therapeutic use , Receptors, Lysosphingolipid/therapeutic use
9.
Acta Ophthalmol ; 92(1): e12-21, 2014 Feb.
Article in English | MEDLINE | ID: mdl-23910624

ABSTRACT

PURPOSE: This study aims to investigate the effects of systemic application of sphingosine 1-phosphate receptor 1(S1P1) on allogeneic corneal transplantation in mice. METHODS: A total of 112 BALB/c mice received corneal grafts from C57BL/6 donors. Recipients were randomly divided into seven groups and treated with intraperitoneal injections of S1P1 (5 mg/kg/days), cyclosporine A (5 mg/kg/days), dexamethasone (1 mg/kg/days) and rapamycin (2 mg/kg/days). S1P1was combined with rapamycin or cyclosporine A, and saline served as negative control. Serum levels of IL-2, IL-10, TGF-ß1 and IFN-γ were measured by Elisa. The numbers of CD4+ T and regulatory (Treg) cell phenotype were measured by flow cytometry. Cytokine mRNA expression was analysed by real-time quantitative PCR. CD4+ T cells and cytokines were histologically identified by immunofluorescence staining. RESULTS: Corneal graft survival was prolonged by intraperitoneal injections in S1P1 alone (mean survival time MST, 35.3 ± 5.6 days), S1P1 combined with rapamycin (MST, 38.7 ± 6.5 days) or S1P1 and cyclosporine A (MST, 32.7 ± 4.8 days) compared with the controls (MST, 14.6 ± 0.2 days; n = 5, p < 0.01). S1P1 alone increased CD4+ T (p < 0.01) and Treg cells (p < 0.01; n = 5) in the cervical and mesenteric lymph nodes compared with the controls and S1P1 + rapamycin (p < 0.05; n = 5). TGF-ß1 and IL-10 mRNA transcriptions in corneal grafts following S1P1+ rapamycin increased (both p < 0.01; n = 3), and TGF-ß1 and IL-10 in the serum level following S1P1 alone increased (both p < 0.01; n = 3). These results paralleled the findings obtained from immunofluorescence. CONCLUSION: S1P1 has significant effect in corneal allograft rejection inhibition. The combined treatment of S1P1 and rapamycin results in synergistic effect.


Subject(s)
Graft Rejection/prevention & control , Receptors, Lysosphingolipid/therapeutic use , Allografts , Animals , CD4-Positive T-Lymphocytes/physiology , Corneal Transplantation , Cyclosporine/therapeutic use , Cytokines/blood , Cytokines/genetics , Dexamethasone/therapeutic use , Drug Therapy, Combination , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Graft Rejection/blood , Graft Survival/drug effects , Immunosuppressive Agents/therapeutic use , Injections, Intraperitoneal , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , RNA, Messenger/genetics , Sirolimus/therapeutic use , T-Lymphocytes, Regulatory/physiology
10.
Nat Chem Biol ; 7(5): 254-6, 2011 May.
Article in English | MEDLINE | ID: mdl-21445057

ABSTRACT

Sphingosine 1-phosphate receptor 1 (S1P(1)) is critical for lymphocyte recirculation and is a clinical target for treatment of multiple sclerosis. By generating a short-duration S1P(1) agonist and mice in which fluorescently tagged S1P(1) replaces wild-type receptor, we elucidate physiological and agonist-perturbed changes in expression of S1P(1) at a subcellular level in vivo. We demonstrate differential downregulation of S1P(1) on lymphocytes and endothelia after agonist treatment.


Subject(s)
Gene Knock-In Techniques , Green Fluorescent Proteins/chemistry , Multiple Sclerosis/drug therapy , Receptors, Lysosphingolipid/agonists , Receptors, Lysosphingolipid/therapeutic use , Animals , Down-Regulation/drug effects , Endothelium/drug effects , Endothelium/metabolism , Flow Cytometry , Fluorescent Dyes/chemistry , Fluorescent Dyes/metabolism , Green Fluorescent Proteins/metabolism , Lymphocytes/drug effects , Lymphocytes/metabolism , Mice , Multiple Sclerosis/metabolism , Multiple Sclerosis/pathology , Receptors, Lysosphingolipid/metabolism , Time Factors
11.
Eur J Clin Invest ; 37(3): 171-9, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17359484

ABSTRACT

All stages of atherosclerosis have been identified as a chronic vascular inflammatory disease. In the last few years there is increasing evidence that endogenous lysophospholipids such as sphingosine-1-phosphate (S1P) have potent anti-inflammatory properties. The S1P analogue FTY720 that has been developed as a potent, orally active, immunosuppressant in the field of transplantation and autoimmune disease has interesting effects on inflammatory processes in the arterial vessel wall. S1P targets five specific S1P receptors (S1P(1-5)), which are ubiquitously expressed. S1P(1-3) receptor expression is identified in arterial vessels. S1P and FTY720 show potent silencing effects on some vascular proinflammatory mechanisms in endothelial and vascular smooth muscle cells. In addition, the interaction of monocytes with the vessel wall is inhibited. As shown recently, FTY720 can effectively reduce the progression of atherosclerosis in apolipoprotein E-deficient mice having a high-cholesterol diet. It is not entirely clear which S1P receptor subtype is mainly involved in this process. However, it is currently speculated that the S1P(3) and probably the S1P(1) is involved in the anti-atherosclerotic effects of FTY720. This review summarizes the current knowledge about S1P- and FTY720-effects on mechanisms of vascular inflammatory disease. In addition S1P receptor subtypes are identified which might be interesting for molecular drug targeting.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Atherosclerosis/drug therapy , Immunosuppressive Agents/therapeutic use , Lysophospholipids/therapeutic use , Propylene Glycols/therapeutic use , Receptors, Lysosphingolipid/therapeutic use , Sphingosine/analogs & derivatives , Apoptosis/drug effects , Endothelial Cells/metabolism , Fingolimod Hydrochloride , Humans , Sphingosine/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL
...