Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 44
Filter
1.
Immunity ; 41(3): 440-450, 2014 Sep 18.
Article in English | MEDLINE | ID: mdl-25238098

ABSTRACT

Pathologically swollen lymph nodes (LNs), or buboes, characterize Yersinia pestis infection, yet how they form and function is unknown. We report that colonization of the draining LN (dLN) occurred due to trafficking of infected dendritic cells and monocytes in temporally distinct waves in response to redundant chemotactic signals, including through CCR7, CCR2, and sphingosine-1-phospate (S1P) receptors. Retention of multiple subsets of phagocytes within peripheral LNs using the S1P receptor agonist FTY720 or S1P1-specific agonist SEW2871 increased survival, reduced colonization of downstream LNs, and limited progression to transmission-associated septicemic or pneumonic disease states. Conditional deletion of S1P1 in mononuclear phagocytes abolished node-to-node trafficking of infected cells. Thus, Y. pestis-orchestrated LN remodeling promoted its dissemination via host cells through the lymphatic system but can be blocked by prevention of leukocyte egress from DLNs. These findings define a novel trafficking route of mononuclear phagocytes and identify S1P as a therapeutic target during infection.


Subject(s)
Lymph Nodes/pathology , Lysophospholipids/genetics , Plague/pathology , Receptors, Lysosphingolipid/immunology , Sphingosine/analogs & derivatives , Yersinia pestis/pathogenicity , Animals , CD11 Antigens/metabolism , CD11b Antigen/metabolism , Cell Movement , Chemokine CCL21/genetics , Dendritic Cells/microbiology , Female , Fingolimod Hydrochloride , Integrin alpha Chains/metabolism , Lymph Nodes/cytology , Lymph Nodes/microbiology , Lysophospholipids/agonists , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Monocytes/microbiology , Oxadiazoles/pharmacology , Phagocytes/immunology , Plague/immunology , Propylene Glycols/pharmacology , Receptors, CCR2/immunology , Receptors, CCR7/genetics , Receptors, CCR7/immunology , Receptors, Lysosphingolipid/agonists , Sphingosine/agonists , Sphingosine/genetics , Sphingosine/pharmacology , Thiophenes/pharmacology , Yersinia pestis/immunology
2.
Nature ; 523(7560): 342-6, 2015 Jul 16.
Article in English | MEDLINE | ID: mdl-26053123

ABSTRACT

Lipid mediators influence immunity in myriad ways. For example, circulating sphingosine-1-phosphate (S1P) is a key regulator of lymphocyte egress. Although the majority of plasma S1P is bound to apolipoprotein M (ApoM) in the high-density lipoprotein (HDL) particle, the immunological functions of the ApoM-S1P complex are unknown. Here we show that ApoM-S1P is dispensable for lymphocyte trafficking yet restrains lymphopoiesis by activating the S1P1 receptor on bone marrow lymphocyte progenitors. Mice that lacked ApoM (Apom(-/-)) had increased proliferation of Lin(-) Sca-1(+) cKit(+) haematopoietic progenitor cells (LSKs) and common lymphoid progenitors (CLPs) in bone marrow. Pharmacological activation or genetic overexpression of S1P1 suppressed LSK and CLP cell proliferation in vivo. ApoM was stably associated with bone marrow CLPs, which showed active S1P1 signalling in vivo. Moreover, ApoM-bound S1P, but not albumin-bound S1P, inhibited lymphopoiesis in vitro. Upon immune stimulation, Apom(-/-) mice developed more severe experimental autoimmune encephalomyelitis, characterized by increased lymphocytes in the central nervous system and breakdown of the blood-brain barrier. Thus, the ApoM-S1P-S1P1 signalling axis restrains the lymphocyte compartment and, subsequently, adaptive immune responses. Unique biological functions imparted by specific S1P chaperones could be exploited for novel therapeutic opportunities.


Subject(s)
Apolipoproteins/metabolism , Central Nervous System/pathology , Lipoproteins, HDL/metabolism , Lymphocytes/cytology , Lymphocytes/metabolism , Lymphopoiesis , Lysophospholipids/metabolism , Sphingosine/analogs & derivatives , Animals , Apolipoproteins/deficiency , Apolipoproteins/genetics , Apolipoproteins M , Blood-Brain Barrier/pathology , Cell Movement , Cell Proliferation/genetics , Central Nervous System/immunology , Central Nervous System/metabolism , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/metabolism , Encephalomyelitis, Autoimmune, Experimental/pathology , Female , Fingolimod Hydrochloride/pharmacology , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Inflammation/immunology , Inflammation/metabolism , Inflammation/pathology , Lymphocytes/immunology , Lymphoid Progenitor Cells/cytology , Lymphoid Progenitor Cells/metabolism , Lysophospholipids/agonists , Lysophospholipids/blood , Lysophospholipids/genetics , Male , Mice , Mice, Inbred C57BL , Protein Binding , Receptors, Lysosphingolipid/metabolism , Signal Transduction , Sphingosine/agonists , Sphingosine/blood , Sphingosine/genetics , Sphingosine/metabolism
3.
Molecules ; 26(17)2021 Aug 24.
Article in English | MEDLINE | ID: mdl-34500564

ABSTRACT

Sphingosine 1-phosphate (S1P) is an extensively studied signaling molecule that contributes to cell proliferation, survival, migration and other functions through binding to specific S1P receptors. The cycle of S1P1 internalization upon S1P binding and recycling to the cell surface when local S1P concentrations are low drives T cell trafficking. S1P1 modulators, such as fingolimod, disrupt this recycling by inducing persistent S1P1 internalization and receptor degradation, which results in blocked egress of T cells from the secondary lymphoid tissues. The approval of these compounds for the treatment of multiple sclerosis has placed the development of S1PR modulators in the focus of pharmacological research, mostly for autoimmune indications. Here, we report on a novel anellated bismorpholino derivative of oxy-fingolimod, named ST-2191, which exerts selective S1P1 agonist and functional antagonist potency. ST-2191 is also effective in reducing the lymphocyte number in mice, and this effect is not dependent on phosphorylation by sphingosine kinase 2 for activity. These data show that ST-2191 is a novel S1P1 modulator, but further experiments are needed to analyze the therapeutic impact of ST-2191 in animal models of autoimmune diseases.


Subject(s)
Fingolimod Hydrochloride/pharmacology , Lysophospholipids/agonists , Lysophospholipids/antagonists & inhibitors , Sphingosine/analogs & derivatives , Animals , CHO Cells , Cricetulus , Humans , Lymphocyte Count/methods , Lysophospholipids/metabolism , Mice , Mice, Inbred C57BL , Multiple Sclerosis/drug therapy , Multiple Sclerosis/metabolism , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Receptors, Lysosphingolipid/metabolism , Signal Transduction/drug effects , Sphingosine/agonists , Sphingosine/antagonists & inhibitors , Sphingosine/metabolism , T-Lymphocytes/metabolism
4.
Mol Cell Biochem ; 469(1-2): 89-95, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32301060

ABSTRACT

Lysophosphatidic acid (LPA) signaling through LPA receptors (LPA1 to LPA6) regulates a variety of malignant properties in cancer cells. Recently, we show that LPA2 expression is elevated by long-term cisplatin (CDDP) treatment in melanoma A375 cells. In the present study, we investigated whether LPA2-mediated signaling is involved in the modulation of chemoresistance in A375 cells. In cell survival assay, cells were treated with CDDP and dacarbazine (DTIC) every 24 h for 2 days. The cell survival rates to CDDP and DTIC were markedly increased by an LPA2 agonist, GRI-977143. To validate the effects of LPA2 on cell survival, LPA2 knockdown cells were generated from A375 cells. The cell survival rates elevated by GRI-977143 were suppressed by LPA2 knockdown. To evaluate the roles of LPA2-mediated signaling in cell survival, cells were pretreated with a Gi protein inhibitor, pertussis toxin (PTX). In the presence of GRI-977143, the cell survival rates to CDDP and DTIC were significantly lower in PTX-treated cells than in untreated cells. In addition, pretreatment of an adenylyl cyclase inhibitor, SQ22536, increased the cell survival of A375 cells treated with CDDP and DTIC. These results suggest that LPA2-mediated signaling plays an important role in the enhancement of chemoresistance of A375 cells treated with anticancer drugs.


Subject(s)
Antineoplastic Agents/pharmacology , Cisplatin/pharmacology , Dacarbazine/pharmacology , Drug Resistance, Neoplasm/genetics , Lysophospholipids/metabolism , Melanoma/metabolism , Receptors, Lysophosphatidic Acid/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Gene Knockdown Techniques , Humans , Lysophospholipids/agonists , Lysophospholipids/genetics , Melanoma/genetics , Pertussis Toxin/toxicity , Receptors, Lysophosphatidic Acid/genetics , Signal Transduction/drug effects , Signal Transduction/genetics
5.
Biochim Biophys Acta Mol Cell Biol Lipids ; 1863(9): 980-990, 2018 09.
Article in English | MEDLINE | ID: mdl-29787912

ABSTRACT

Given their important role in neuronal function, there has been an increasing focus on altered lipid levels in brain disorders. The effect of a high-fat (HF) diet on the lipid profiles of the cortex, hippocampus, hypothalamus, and olfactory bulb of the mouse brain was investigated using nanoflow ultrahigh pressure liquid chromatography-electrospray ionization-tandem mass spectrometry in the current study. For 8 weeks, two groups of 5-week-old mice were fed either an HF or normal diet (6 mice from each group analyzed as the F and N groups, respectively). The remaining mice in both groups then received a 4-week normal diet. Each group was then subdivided into two groups for another 4-week HF or normal diet. Quantitative analysis of 270 of the 359 lipids identified from brain tissue revealed that an HF diet significantly affected the brain lipidome in all brain regions that were analyzed. The HF diet significantly increased diacylglycerols, which play a role in insulin resistance in all regions that were analyzed. Although the HF diet increased most lipid species, the majority of phosphatidylserine species were decreased, while lysophosphatidylserine species, with the same acyl chain, were substantially increased. This result can be attributed to increased oxidative stress due to the HF diet. Further, weight-cycling (yo-yo effect) was found more critical for the perturbation of brain lipid profiles than weight gain without a preliminary experience of an HF diet. The present study reveals systematic alterations in brain lipid levels upon HF diet analyzed either by lipid class and molecular levels.


Subject(s)
Cerebral Cortex/drug effects , Diet, High-Fat , Dietary Fats/administration & dosage , Hippocampus/drug effects , Hypothalamus/drug effects , Metabolome , Olfactory Bulb/drug effects , Animals , Brain Chemistry , Cerebral Cortex/metabolism , Chromatography, High Pressure Liquid , Diglycerides/agonists , Diglycerides/metabolism , Hippocampus/metabolism , Hypothalamus/metabolism , Insulin Resistance , Lipid Metabolism/drug effects , Lysophospholipids/agonists , Lysophospholipids/metabolism , Male , Mice , Mice, Inbred C57BL , Olfactory Bulb/metabolism , Oxidative Stress , Phosphatidylserines/antagonists & inhibitors , Phosphatidylserines/metabolism , Spectrometry, Mass, Electrospray Ionization
6.
Stroke ; 47(12): 3053-3056, 2016 12.
Article in English | MEDLINE | ID: mdl-27827329

ABSTRACT

BACKGROUND AND PURPOSE: Growing evidence supports that the immunomodulatory drug fingolimod is protective in stroke. Fingolimod binds to 4 of 5 sphingosine-1-phosphate (S1P) receptors and, among other actions, it induces lymphopenia. In this study, we investigated whether a selective S1P1 agonist is protective in experimental stroke. METHODS: Drug selectivity was studied in vitro in cells overexpressing the human S1P receptors. Mice (n=54) received different doses of LASW1238 (3 or 10 mg/kg), fingolimod (1 mg/kg), or the vehicle intraperitoneal, and lymphopenia was studied at different time points. After intraluminal middle cerebral artery occlusion for 45 minutes and immediately after reperfusion, mice (n=56) received the drug treatment. At 24 hours, a neurological test was performed and infarct volume was measured. Treatment and all the analyses were performed in a blind fashion. RESULTS: In vitro functional assays showed that LASW1238 is a selective agonist of the S1P1 receptor. At 10 mg/kg, this compound induced sustained lymphopenia in mice comparable with fingolimod, whereas at 3 mg/kg it induced short-lasting lymphopenia. After ischemia, both LASW1238 (10 mg/kg) and fingolimod reduced infarct volume, but only LASW1238 (10 mg/kg) showed statistically significant differences versus the vehicle. The neurological function and plasma cytokine levels were not different between groups. CONCLUSIONS: The selective S1P1 agonist LASW1238 reduces infarct volume after ischemia/reperfusion in mice, but only when lymphopenia is sustained for at least 24 hours. S1P1 and lymphocytes are potential targets for drug treatment in stroke. Defining the best drug dosing regimens to control the extent and duration of lymphopenia is critical to achieve the desired effects.


Subject(s)
Cerebral Infarction/drug therapy , Lymphopenia/chemically induced , Lysophospholipids/agonists , Neuroprotective Agents/pharmacology , Receptors, Lysosphingolipid/agonists , Reperfusion Injury/drug therapy , Sphingosine/analogs & derivatives , Animals , Fingolimod Hydrochloride/pharmacology , Immunosuppressive Agents/pharmacology , Male , Mice , Mice, Inbred C57BL , Sphingosine/agonists
7.
Bioorg Med Chem Lett ; 26(10): 2470-2474, 2016 05 15.
Article in English | MEDLINE | ID: mdl-27055941

ABSTRACT

The synthesis and structure-activity relationship (SAR) of a series of pyridyl-isoxazole based agonists of S1P1 are discussed. Compound 5b provided potent in vitro activity with selectivity, had an acceptable pharmacokinetic profile, and demonstrated efficacy in a dose dependent manner when administered orally in a rodent model of arthritis.


Subject(s)
Arthritis, Experimental/drug therapy , Lysophospholipids/agonists , Sphingosine/analogs & derivatives , Structure-Activity Relationship , Administration, Oral , Animals , Chemistry Techniques, Synthetic , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical/methods , Humans , Isoxazoles/chemistry , Isoxazoles/pharmacology , Lymphocyte Count , Male , Rats, Inbred Lew , Receptors, Lysosphingolipid/agonists , Sphingosine/agonists
8.
J Immunol ; 193(4): 1966-74, 2014 Aug 15.
Article in English | MEDLINE | ID: mdl-25015824

ABSTRACT

Although much is described about the molecules involved in neutrophil migration from circulation into tissues, less is known about the molecular mechanisms that regulate neutrophil entry into lymph nodes (LNs) draining a local inflammatory site. In this study, we investigated neutrophil migration toward LNs in a context of inflammation induced by immunization of BALB/c mice with OVA emulsified in CFA. We demonstrated that neutrophils can enter LNs of OVA/CFA-immunized mice not only via lymphatic vessels but also from blood, across high endothelial venules. By adoptive transfer experiments, we showed that this influx was dependent on an inflammatory-state condition and previous neutrophil stimulation with OVA/anti-OVA immune complexes. Importantly, we have demonstrated that, in the migratory pattern to LNs, neutrophils used L-selectin and P-selectin glycoprotein ligand-1, macrophage-1 Ag and LFA-1 integrins, and CXCR4 to get access across high endothelial venules, whereas macrophage-1 Ag, LFA-1, and CXCR4 were involved in their trafficking through afferent lymphatics. Strikingly, we found that stimulation with immune complexes significantly upregulated the expression of sphingosine-1-phosphate receptor 4 on neutrophils, and that treatment with the sphingosine-1-phosphate agonist FTY720 altered neutrophil LN-homing ability. These findings summarized in this article disclose the molecular pattern that controls neutrophil recruitment to LNs.


Subject(s)
Antigen-Antibody Complex/immunology , Immune System Diseases/immunology , Leukocyte Disorders/immunology , Lymph Nodes/immunology , Neutrophils/immunology , Adoptive Transfer , Animals , Cell Movement/immunology , Female , Fingolimod Hydrochloride , Immunosuppressive Agents/pharmacology , Inflammation/immunology , L-Selectin/immunology , Lymph Nodes/cytology , Lymphatic Vessels/immunology , Lymphocyte Function-Associated Antigen-1/immunology , Lysophospholipids/agonists , Macrophage-1 Antigen/immunology , Mice , Mice, Inbred BALB C , Neutrophils/transplantation , P-Selectin/immunology , Propylene Glycols/pharmacology , Receptors, CXCR4/immunology , Receptors, Lysosphingolipid/metabolism , Sphingosine/agonists , Sphingosine/analogs & derivatives , Sphingosine/pharmacology
9.
Clin Exp Pharmacol Physiol ; 43(2): 166-73, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26582369

ABSTRACT

There is growing evidence that diabetes mellitus causes attenuation of the bioactive metabolite of membrane sphingolipids, sphingosine-1-phosphate, and this may be a key mechanism in the decreased cardioprotective effect of ischaemic preconditioning (IPC) in the diabetic heart. Thus, this study has been designed to investigate the role and pharmacological potential of sphingosine-1-phosphate in diabetic rat heart. Diabetes was produced in Wistar rats by administration of a low dose of streptozotocin (STZ) (35 mg/kg, i.p., once) and feeding a high fat diet (HFD) for 6 weeks. Isolated rat heart was subjected to 30 min ischaemia followed by 120 min of reperfusion (I/R). The heart was subjected to pre-ischaemic treatment (before ischaemia for 20 min) and pharmacological preconditioning with the S1P agonist FTY720 (0.6 µmol/L) with and without atractyloside (an mPTP opener; in the last episode of reperfusion before I/R). Myocardial infarction was assessed in terms of increase in lactate dehydrogenase (LDH), creatinine kinase-MB (CK-MB), myeloperoxidase (MPO) level and infarct size (triphenyltetrazolium chloride staining). Immunohistochemistry analysis was done for assessment of tumour necrosis factor (TNF)-α and glycogen synthase kinase (GSK)-3ß level in cardiac tissue. Pre-ischaemic treatment and pharmacological preconditioning with FTY720 significantly decreased I/R-induced myocardial infarction, TNF-alpha, GSK-3ß level and release of LDH and CK-MB as compared to control group. The cardioprotective effect of S1P agonist was significantly attenuated by atractyloside. It may be concluded that S1P agonist FTY720 prevents the diabetic heart from ischaemic reperfusion injury, possibly through inhibition of GSK-3ß and regulation of opening of mitochondrial permeability transition pore.


Subject(s)
Diabetes Mellitus, Experimental/complications , Fingolimod Hydrochloride/pharmacology , Glycogen Synthase Kinase 3/metabolism , Lysophospholipids/agonists , Mitochondrial Membrane Transport Proteins/metabolism , Myocardial Reperfusion Injury/prevention & control , Myocardium/metabolism , Sphingosine/analogs & derivatives , Animals , Blood Glucose/metabolism , Cardiotonic Agents/pharmacology , Creatine Kinase, MB Form/metabolism , Diet, High-Fat/adverse effects , Glycogen Synthase Kinase 3 beta , Ischemic Preconditioning , Lipids/blood , Male , Mitochondrial Permeability Transition Pore , Myocardial Reperfusion Injury/blood , Myocardial Reperfusion Injury/complications , Myocardial Reperfusion Injury/metabolism , Rats , Rats, Wistar , Sphingosine/agonists
10.
Curr Top Microbiol Immunol ; 378: 149-70, 2014.
Article in English | MEDLINE | ID: mdl-24728597

ABSTRACT

The development of fingolimod, an unselective functional antagonist of the interactions between sphingosine 1 phosphate (S1P) and sphingosine 1 phosphate receptors (S1PRs), as the first oral therapy for multiple sclerosis (MS) has been a milestone. The parallel intensive research on the role of S1P, sphingosine kinases, and the five known S1PRs, their tissue distribution and expression in physiological and pathological conditions have led to a wide range of interesting findings. The initial focus of this research in the context of developing fingolimod as a treatment of MS has been on its immunological effects. The wide distribution and important roles of sphingosine, its metabolites, and their receptors in the central nervous system (CNS) in general, in myelin, and in all cell types of this organ have spurred interest to examine S1P and its five receptors in the brain as well. The present review will concentrate on the latter area and give a brief overview of what is known about S1P/S1PR interactions in the CNS in physiological and pathological conditions.


Subject(s)
Central Nervous System Diseases/metabolism , Lysophospholipids/therapeutic use , Sphingosine/analogs & derivatives , Animals , Central Nervous System Diseases/drug therapy , Humans , Lysophospholipids/agonists , Lysophospholipids/metabolism , Receptors, Lysosphingolipid/metabolism , Sphingosine/agonists , Sphingosine/metabolism , Sphingosine/therapeutic use
11.
Curr Top Microbiol Immunol ; 378: 129-47, 2014.
Article in English | MEDLINE | ID: mdl-24728596

ABSTRACT

Cytokine storm defines a dysregulation of and an excessively exaggerated immune response most often accompanying selected viral infections and several autoimmune diseases. Newly emerging and re-emerging infections of the respiratory tract, especially influenza, SARS, and hantavirus post considerable medical problems. Their morbidities and mortalities are often a direct result of cytokine storm. This chapter visits primarily influenza virus infection and resultant cytokine storm. It provides the compelling evidence that illuminates cytokine storm in influenza pathogenesis and the clear findings that cytokine storm is chemically tractable by therapy directed toward sphingosine-1-phosphate receptor (S1PR) modulation, specifically S1P1R agonist therapy. The mechanism(s) of how S1P1R signaling works and the pathways involved are subjects of this review.


Subject(s)
Cytokines/immunology , Influenza, Human/drug therapy , Influenza, Human/mortality , Lysophospholipids/agonists , Lysophospholipids/therapeutic use , Orthomyxoviridae/drug effects , Sphingosine/analogs & derivatives , Animals , Humans , Influenza, Human/immunology , Influenza, Human/virology , Orthomyxoviridae/immunology , Sphingosine/agonists , Sphingosine/therapeutic use
12.
Acta Anaesthesiol Scand ; 57(6): 713-8, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23683126

ABSTRACT

BACKGROUND: Increased vascular leakage leading to hypovolaemia and tissue oedema is common in severe sepsis. Hypovolaemia together with oedema formation may contribute to hypoxia and result in multiorgan failure and death. To improve treatment during sepsis, a potential therapeutic target may be to reduce the vascular leakage. Substances affecting the endothelial barrier are interesting in this respect, as it is suggested that increase in vascular leakage depends on reorganisation of the endothelial cells and breakdown of the endothelial barrier. The agonist of the bioactive lipid sphingosine-1-phosphate, FTY720, has been shown to modulate the integrity of the endothelium and reduce permeability both in vitro and in vivo. The aim of the present study was to determine if FTY720 could reduce the loss of plasma volume during experimental sepsis in rats. METHODS: Sepsis was induced by ligation and incision of the caecum in the rat. Plasma volume was determined before and 4.5 h after induction of sepsis by a dilution technique using (125) I-labelled albumin. RESULTS: FTY720 in a dose of 0.2 mg/kg reduced the loss of plasma during sepsis by approximately 30% compared with vehicle, without any adverse effects on haemodynamic and physiological parameters. The increase in hematocrit and haemoglobin concentration was also found to be higher in the vehicle group. CONCLUSION: FTY720 in a dose without haemodynamic side effects reduces loss of plasma volume during experimental sepsis most likely because of reduction in permeability and may therefore be beneficial in sepsis.


Subject(s)
Lysophospholipids/agonists , Plasma Volume/drug effects , Propylene Glycols/therapeutic use , Sepsis/physiopathology , Sphingosine/analogs & derivatives , Animals , Capillary Leak Syndrome/drug therapy , Capillary Leak Syndrome/etiology , Capillary Permeability/drug effects , Cecum/injuries , Disease Models, Animal , Diuresis/drug effects , Drug Evaluation, Preclinical , Edema/etiology , Edema/prevention & control , Endothelium, Vascular/drug effects , Fingolimod Hydrochloride , Hematocrit , Hemodynamics/drug effects , Hemoglobins/analysis , Intestinal Perforation/complications , Male , Propylene Glycols/pharmacology , Random Allocation , Rats , Rats, Sprague-Dawley , Sepsis/blood , Sepsis/etiology , Sphingosine/agonists , Sphingosine/pharmacology , Sphingosine/therapeutic use
13.
Am J Physiol Lung Cell Mol Physiol ; 302(8): L736-45, 2012 Apr 15.
Article in English | MEDLINE | ID: mdl-22287614

ABSTRACT

Sphingosine-1-phosphate (S1P) is an immunomodulatory lipid mediator that plays an important role in lymphocyte trafficking. Elevated levels of S1P are found in bronchoalveolar lavage (BAL) fluid of patients with asthma; however, its role in disease is not known. FTY720, a synthetic analog of S1P, has been shown to abrogate allergic inflammation and airway hyperresponsiveness following acute allergen challenge. However, its effects on asthmatic airway remodeling induced by repeated allergen exposure are unknown. Ovalbumin (OVA)-sensitized rats were challenged on days 14, 19, and 24 after sensitization. FTY720 or vehicle (PBS) therapy was administered 1 h prior to each challenge. BAL fluid and quantitative histological analysis were performed 48 h after the last challenge. FTY720 inhibited OVA-induced features of airway remodeling including increased airway smooth muscle mass and bronchial neovascularization, without affecting lymphocyte numbers in secondary lymphoid organs. Furthermore, CD3+ cells adjacent to airway smooth muscle bundles were increased in OVA-challenged rats but the increase was inhibited by FTY720. There was an expansion of bronchus-associated lymphoid tissue following FTY720 treatment of OVA-challenged animals. Real-time quantitative PCR revealed that Th2-associated transcription factors were inhibited following FTY720 therapy. Airway remodeling is a cardinal feature of severe asthma. These results demonstrate that allergen-driven airway remodeling can be inhibited by FTY720, offering potential new therapies for the treatment of severe asthma.


Subject(s)
Airway Remodeling/drug effects , Allergens/adverse effects , Immunosuppressive Agents/therapeutic use , Lysophospholipids/agonists , Lysophospholipids/therapeutic use , Propylene Glycols/therapeutic use , Sphingosine/analogs & derivatives , Animals , Anti-Asthmatic Agents/therapeutic use , Asthma/drug therapy , Bronchi/blood supply , Bronchi/drug effects , Bronchi/pathology , Bronchoalveolar Lavage Fluid , CD3 Complex/analysis , Fingolimod Hydrochloride , Male , Muscle, Smooth/drug effects , Rats , Severity of Illness Index , Sphingosine/agonists , Sphingosine/therapeutic use , Treatment Outcome
14.
Xenobiotica ; 42(7): 671-86, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22225501

ABSTRACT

Sphingosine-1-phosphate (S1P(1)) receptor agonists such as Fingolimod (FTY-720) are a novel class of immunomodulators that have clinical utility in the treatment of remitting relapsing multiples sclerosis. This class of compound act by inducing peripheral lymphopenia. Using an integrated pharmacokinetic/pharmacodynamic (PK-PD) approach based on an in vivo rat model, novel S1P(1) agonists were identified with a predicted more rapid rate of reversibility of lymphocyte reduction in human compared to Fingolimod. The in vivo potency of 15 compounds based on PK-PD modelling of the rat lymphocyte reduction model was correlated with in vitro measures of potency at the S1P(1) receptor using ß arrestin recruitment and G-protein signalling. A structurally novel S1P(1) agonist was identified and predictions of human pharmacokinetics and clinical dose are presented.


Subject(s)
Lysophospholipids/agonists , Propylene Glycols/pharmacokinetics , Sphingosine/analogs & derivatives , Animals , Arrestin/metabolism , Fingolimod Hydrochloride , GTP-Binding Proteins/metabolism , Lymphocytes/drug effects , Lymphocytes/metabolism , Lysophospholipids/metabolism , Male , Multiple Sclerosis, Relapsing-Remitting/drug therapy , Propylene Glycols/pharmacology , Propylene Glycols/therapeutic use , Rats , Rats, Inbred Strains , Signal Transduction , Sphingosine/agonists , Sphingosine/metabolism , Sphingosine/pharmacokinetics , Sphingosine/pharmacology , Sphingosine/therapeutic use
15.
Rheumatol Int ; 31(7): 967-9, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21113810

ABSTRACT

Therapeutics targeting sphingosine-1-phosphate (S1P), a kind of lipid mediator regulating immune cell trafficking, has been emerging rapidly as a novel line of regimen for autoimmune diseases, including rheumatoid arthritis (RA). Here, we propose that S1P-targeted therapy is beneficial not only for limiting inflammation but for preventing bone-resorptive disorders, such as osteoporosis, by controlling the migratory behavior of osteoclast precursors and therefore would be good for treating elderly female RA patients who suffer from postmenopausal osteoporosis and arthritis simultaneously.


Subject(s)
Arthritis, Rheumatoid/drug therapy , Lysophospholipids/agonists , Osteoclasts/drug effects , Osteoporosis, Postmenopausal/drug therapy , Receptors, Lysosphingolipid/agonists , Sphingosine/analogs & derivatives , Age Factors , Aged , Animals , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Arthritis, Rheumatoid/complications , Female , Humans , Lysophospholipids/physiology , Mice , Osteoclasts/physiology , Osteoporosis, Postmenopausal/immunology , Receptors, Lysosphingolipid/physiology , Sphingosine/agonists , Sphingosine/physiology , Treatment Outcome
16.
Sci Rep ; 11(1): 15308, 2021 07 28.
Article in English | MEDLINE | ID: mdl-34321503

ABSTRACT

Sphingosine 1-phosphate (S1P) is a bioactive signalling sphingolipid that is increased in diseases such as obesity and diabetes. S1P can modulate platelet function, however the direction of effect and S1P receptors (S1PRs) involved are controversial. Here we describe the role of S1P in regulating human platelet function and identify the receptor subtypes responsible for S1P priming. Human platelets were treated with protease-activated receptor 1 (PAR-1)-activating peptide in the presence or absence of S1P, S1PR agonists or antagonists, and sphingosine kinases inhibitors. S1P alone did not induce platelet aggregation but at low concentrations S1P enhanced PAR1-mediated platelet responses, whereas PAR1 responses were inhibited by high concentrations of S1P. This biphasic effect was mimicked by pan-S1PR agonists. Specific agonists revealed that S1PR1 receptor activation has a positive priming effect, S1PR2 and S1PR3 have no effect on platelet function, whereas S1PR4 and S1PR5 receptor activation have an inhibitory effect on PAR-1 mediated platelet function. Although platelets express both sphingosine kinase 1/2, enzymes which phosphorylate sphingosine to produce S1P, only dual and SphK2 inhibition reduced platelet function. These results support a role for SphK2-mediated S1P generation in concentration-dependent positive and negative priming of platelet function, through S1PR1 and S1PR4/5 receptors, respectively.


Subject(s)
Lysophospholipids/pharmacology , Platelet Activation/drug effects , Sphingosine-1-Phosphate Receptors/drug effects , Sphingosine/analogs & derivatives , Blood Platelets/drug effects , Blood Platelets/ultrastructure , Carrier Proteins/pharmacology , Cell Shape/drug effects , Dose-Response Relationship, Drug , Humans , Lysophospholipids/agonists , Lysophospholipids/antagonists & inhibitors , Peptide Fragments/pharmacology , Peptides/pharmacology , Phosphotransferases (Alcohol Group Acceptor)/antagonists & inhibitors , Phosphotransferases (Alcohol Group Acceptor)/physiology , Platelet Aggregation/drug effects , Receptor, PAR-1/agonists , Sphingosine/agonists , Sphingosine/antagonists & inhibitors , Sphingosine/pharmacology , Sphingosine-1-Phosphate Receptors/physiology
17.
Biol Pharm Bull ; 33(2): 204-8, 2010.
Article in English | MEDLINE | ID: mdl-20118541

ABSTRACT

Lysophosphatidic acid (LPA) is a lipid mediator that is known to exhibit chemotactic activity toward a variety of cancer cells. However, its effect on the immune system has not been studied extensively. Another lipid mediator, sphingosine-1-phosphate (S1P), has been shown to influence lymphocyte recirculation by regulating lymphocyte egress from lymphoid organs. In this study, we found that LPA inhibits spontaneous migration of mouse splenic lymphocytes through a chemorepulsive effect. We also demonstrated that LPA inhibits chemokine CCL21-induced lymphocyte migration. This inhibitory effect on CCL21-induced migration was observed for both T and B cells. The involvement of a receptor, LPA(1), LPA(2) or LPA(3), in the inhibition of the CCL21-induced migration was confirmed with a synthetic agonist, oleyl thiophosphate. Considering that the signaling by CCL21 through cognate receptor CCR7 contributes to lymphocyte homing and dendritic cell trafficking to lymph nodes, LPA may play a role as a key regulator of these processes. The inhibitory effect of LPA is in remarkable contrast to the effect of S1P receptor signaling, which is known to potentiate lymphocyte chemotaxis involving CCR7.


Subject(s)
Chemotaxis, Leukocyte/immunology , Lymphocytes/cytology , Lysophospholipids/physiology , Animals , Cell Migration Inhibition/drug effects , Cell Migration Inhibition/immunology , Chemokine CCL21/antagonists & inhibitors , Chemokine CCL21/physiology , Chemotaxis, Leukocyte/drug effects , Fatty Alcohols/pharmacology , Female , Lymphocytes/drug effects , Lymphocytes/immunology , Lysophospholipids/agonists , Lysophospholipids/pharmacology , Mice , Mice, Inbred BALB C , Organophosphates/pharmacology
18.
Front Immunol ; 11: 1102, 2020.
Article in English | MEDLINE | ID: mdl-32670273

ABSTRACT

With the sudden outbreak of COVID-19 patient worldwide and associated mortality, it is critical to come up with an effective treatment against SARS-CoV-2. Studies suggest that mortality due to COVID 19 is mainly attributed to the hyper inflammatory response leading to cytokine storm and ARDS in infected patients. Sphingosine-1-phosphate receptor 1 (S1PR1) analogs, AAL-R and RP-002, have earlier provided in-vivo protection from the pathophysiological response during H1N1 influenza infection and improved mortality. Recently, it was shown that the treatment with sphingosine-1-phosphate receptor 1 analog, CYM5442, resulted in the significant dampening of the immune response upon H1N1 challenge in mice and improved survival of H1N1 infected mice in combination with an antiviral drug, oseltamivir. Hence, here we suggest to investigate the possible utility of using S1P analogs to treat COVID-19.


Subject(s)
Coronavirus Infections/drug therapy , Cytokine Release Syndrome/prevention & control , Indans/therapeutic use , Lysophospholipids/agonists , Oxadiazoles/therapeutic use , Pneumonia, Viral/drug therapy , Sphingosine-1-Phosphate Receptors/metabolism , Sphingosine/analogs & derivatives , Animals , Betacoronavirus/drug effects , Betacoronavirus/immunology , COVID-19 , Humans , Influenza A Virus, H1N1 Subtype/drug effects , Mice , Orthomyxoviridae Infections/drug therapy , Orthomyxoviridae Infections/prevention & control , Oseltamivir/therapeutic use , Pandemics , SARS-CoV-2 , Sphingosine/agonists
19.
Transplant Proc ; 51(6): 2081-2098, 2019.
Article in English | MEDLINE | ID: mdl-31399186

ABSTRACT

Sphingosine-1-phosphate (S1P) is a biologically active sphingolipid that acts through the members of a family of 5 G protein-coupled receptors (S1P1 to S1P5). Among these, S1P1 is a major regulator of lymphocyte trafficking. Fingolimod, whose active metabolite, fingolimod phosphate, acts as a nonselective S1P-receptor agonist, exerts its immunomodulatory effect, at least in part, by regulating lymphocyte trafficking via downregulation of S1P1 expression on lymphocytes. Here, we describe the pharmacologic profile of a novel S1P1 agonist, ASP1126. ASP1126 preferentially activated S1P1 compared to S1P3 in rat and human guanosine-5'-(γ-thio)-triphosphate (GTPγS) assays. Oral single administration of ASP1126 decreased the number of peripheral lymphocytes and repeated dosing showed a cumulative effect on lymphopenia in both rats and monkeys. ASP1126 prolonged allograft survival in a rat heterotopic heart transplantation model in combination with a subtherapeutic dose of tacrolimus that was independent of drug-drug interactions. In addition, in nonhuman primate (NHP) renal transplantation, pretreatment with ASP1126 reduced not only the number of naive T cells and central memory T cells but also effector memory T cells in the peripheral blood, all of which could contribute to acute graft rejection and prolonged allograft survival in combination with tacrolimus. Further, we confirmed that ASP1126 has a broad ranging safety margin with respect to its effect on lung weight in rats and bradycardia in NHPs, which were the adverse events found in clinical studies of fingolimod. ASP1126 with improved safety profile has the potential to be an adjunct therapy in combination with tacrolimus in clinical transplantation.


Subject(s)
Graft Rejection/prevention & control , Graft Survival/drug effects , Immunosuppressive Agents/pharmacology , Lysophospholipids/agonists , Sphingosine/analogs & derivatives , Allografts/drug effects , Allografts/metabolism , Animals , Bradycardia/chemically induced , Drug Synergism , Humans , Lymphocytes/drug effects , Macaca fascicularis , Male , Rats , Sphingosine/agonists , Tacrolimus/pharmacology , Transplantation, Homologous/methods
20.
Stroke ; 39(12): 3411-7, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18757288

ABSTRACT

BACKGROUND AND PURPOSE: We have previously shown that the sphingosine 1-phosphate (S1P)/S1P receptor-1 (S1P(1)R) axis contributes to the migration of transplanted neural progenitor cells (NPCs) toward areas of spinal cord injury. In the current study, we examined a strategy to increase endogenous NPC migration toward the injured central nervous system to modify S1PR. METHODS: S1P concentration in the ischemic brain was measured in a mouse thrombosis model of the middle cerebral artery. NPC migration in vitro was assessed by a Boyden chamber assay. Endogenous NPC migration toward the insult was evaluated after ventricular administration of the S1P(2)R antagonist JTE-013. RESULTS: The concentration of S1P in the brain was increased after ischemia and was maximal 14 days after the insult. The increase in S1P in the infarcted brain was primarily caused by accumulation of microglia at the insult. Mouse NPCs mainly expressed S1P(1)R and S1P(2)R as S1PRs, and S1P significantly induced the migration of NPCs in vitro through activation of S1P(1)R. However, an S1P(1)R agonist failed to have any synergistic effect on S1P-mediated NPC migration, whereas pharmacologic or genetic inhibition of S1P(2)R by JTE-013 or short hairpin RNA expression enhanced S1P-mediated NPC migration but did not affect proliferation and differentiation. Interestingly, administration of JTE-013 into a brain ventricle significantly enhanced endogenous NPC migration toward the area of ischemia. CONCLUSIONS: Our findings suggest that S1P is a chemoattractant for NPCs released from an infarcted area and regulation of S1P(2)R function further enhances the migration of NPCs toward a brain infarction.


Subject(s)
Brain/cytology , Cerebral Infarction/therapy , Chemotaxis/drug effects , Embryonic Stem Cells/transplantation , Lysophospholipids/physiology , Pyrazoles/therapeutic use , Pyridines/therapeutic use , Receptors, Lysosphingolipid/antagonists & inhibitors , Sphingosine/analogs & derivatives , Animals , Brain Ischemia/complications , Cell Differentiation/drug effects , Cell Division/drug effects , Cell Movement/drug effects , Cells, Cultured , Cerebral Infarction/drug therapy , Cerebral Infarction/physiopathology , Chemotaxis/physiology , Drug Evaluation, Preclinical , Embryonic Stem Cells/cytology , Embryonic Stem Cells/drug effects , Female , Injections, Intraventricular , Lymphocyte Subsets/drug effects , Lysophospholipids/agonists , Mice , Mice, Inbred C57BL , Microglia/drug effects , Pyrazoles/pharmacology , Pyridines/pharmacology , RNA Interference , RNA, Small Interfering/pharmacology , Receptors, Lysosphingolipid/physiology , Sphingosine/agonists , Sphingosine/physiology , Sphingosine-1-Phosphate Receptors
SELECTION OF CITATIONS
SEARCH DETAIL