Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 350
Filter
1.
Sci Rep ; 14(1): 9700, 2024 04 27.
Article in English | MEDLINE | ID: mdl-38678148

ABSTRACT

Ocular abnormal angiogenesis and edema are featured in several ocular diseases. S1P signaling via S1P1 likely is part of the negative feedback mechanism necessary to maintain vascular health. In this study, we conducted pharmacological experiments to determine whether ASP4058, a sphingosine 1-phosphate receptor 1/5 (S1P1/5) agonist, is useful in abnormal vascular pathology in the eye. First, human retinal microvascular endothelial cells (HRMECs) were examined using vascular endothelial growth factor (VEGF)-induced cell proliferation and hyperpermeability. ASP4058 showed high affinity and inhibited VEGF-induced proliferation and hyperpermeability of HRMECs. Furthermore, S1P1 expression and localization changes were examined in the murine laser-induced choroidal neovascularization (CNV) model, a mouse model of exudative age-related macular degeneration, and the efficacy of ASP4058 was verified. In the CNV model mice, S1P1 tended to decrease in expression immediately after laser irradiation and colocalized with endothelial cells and Müller glial cells. Oral administration of ASP4058 also suppressed vascular hyperpermeability and CNV, and the effect was comparable to that of the intravitreal administration of aflibercept, an anti-VEGF drug. Next, efficacy was also examined in a retinal vein occlusion (RVO) model in which retinal vascular permeability was increased. ASP4058 dose-dependently suppressed the intraretinal edema. In addition, it suppressed the expansion of the perfusion area observed in the RVO model. ASP4058 also suppressed the production of VEGF in the eye. Collectively, ASP4058 can be a potential therapeutic agent that normalizes abnormal vascular pathology, such as age-related macular degeneration and RVO, through its direct action on endothelial cells.


Subject(s)
Choroidal Neovascularization , Disease Models, Animal , Animals , Humans , Choroidal Neovascularization/drug therapy , Choroidal Neovascularization/metabolism , Choroidal Neovascularization/pathology , Mice , Sphingosine-1-Phosphate Receptors/metabolism , Sphingosine-1-Phosphate Receptors/agonists , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Vascular Endothelial Growth Factor A/metabolism , Cell Proliferation/drug effects , Mice, Inbred C57BL , Receptors, Lysosphingolipid/agonists , Receptors, Lysosphingolipid/metabolism , Male
2.
Biomed Pharmacother ; 174: 116575, 2024 May.
Article in English | MEDLINE | ID: mdl-38599060

ABSTRACT

Sepsis is characterized as life-threatening organ dysfunction caused by a dysregulated host response to an infection. Despite numerous clinical trials that addressed this syndrome, there is still no causative treatment available to dampen its severity. Curtailing the infection at an early stage with anti-infectives is the only effective treatment regime besides intensive care. In search for additional treatment options, we recently discovered the inhibition of the sphingosine 1-phosphate (S1P) lyase and subsequent activation of the S1P receptor type 3 (S1PR3) in pre-conditioning experiments as promising targets for sepsis prevention. Here, we demonstrate that treatment of septic mice with the direct S1P lyase inhibitor C31 or the S1PR3 agonist CYM5541 in the advanced phase of sepsis resulted in a significantly increased survival rate. A single dose of each compound led to a rapid decline of sepsis severity in treated mice and coincided with decreased cytokine release and increased lung barrier function with unaltered bacterial load. The survival benefit of both compounds was completely lost in S1PR3 deficient mice. Treatment of the murine macrophage cell line J774.1 with either C31 or CYM5541 resulted in decreased protein kinase B (Akt) and stress-activated protein kinase/c-Jun N-terminal kinase (SAPK/JNK) phosphorylation without alteration of the mitogen-activated protein kinase (MAPK) p38 and p44/42 phosphorylation. Thus, activation of S1PR3 in the acute phase of sepsis by direct agonism or S1P lyase inhibition dampened Akt and JNK phosphorylation, resulting in decreased cytokine release, improved lung barrier stability, rapid decline of sepsis severity and better survival in mice.


Subject(s)
Aldehyde-Lyases , Mice, Inbred C57BL , Sepsis , Sphingosine-1-Phosphate Receptors , Animals , Sepsis/drug therapy , Sepsis/metabolism , Sphingosine-1-Phosphate Receptors/metabolism , Sphingosine-1-Phosphate Receptors/antagonists & inhibitors , Aldehyde-Lyases/antagonists & inhibitors , Aldehyde-Lyases/metabolism , Mice , Receptors, Lysosphingolipid/agonists , Receptors, Lysosphingolipid/metabolism , Male , Disease Models, Animal , Cell Line , Proto-Oncogene Proteins c-akt/metabolism , Cytokines/metabolism , Mice, Knockout
3.
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
4.
EMBO Mol Med ; 15(5): e16645, 2023 05 08.
Article in English | MEDLINE | ID: mdl-36912000

ABSTRACT

Sphingosine-1-phosphate (S1P), the circulating HDL-bound lipid mediator that acts via S1P receptors (S1PR), is required for normal vascular development. The role of this signaling axis in vascular retinopathies is unclear. Here, we show in a mouse model of oxygen-induced retinopathy (OIR) that endothelial overexpression of S1pr1 suppresses while endothelial knockout of S1pr1 worsens neovascular tuft formation. Furthermore, neovascular tufts are increased in Apom-/- mice which lack HDL-bound S1P while they are suppressed in ApomTG mice which have more circulating HDL-S1P. These results suggest that circulating HDL-S1P activation of endothelial S1PR1 suppresses neovascular pathology in OIR. Additionally, systemic administration of ApoM-Fc-bound S1P or a small-molecule Gi-biased S1PR1 agonist suppressed neovascular tuft formation. Circulating HDL-S1P activation of endothelial S1PR1 may be a key protective mechanism to guard against neovascular retinopathies that occur not only in premature infants but also in diabetic patients and aging people.


Subject(s)
Retinal Neovascularization , Mice , Animals , Sphingosine-1-Phosphate Receptors , Receptors, Lysosphingolipid/genetics , Receptors, Lysosphingolipid/agonists , Lipoproteins, HDL , Sphingosine , Lysophospholipids
5.
Mol Cell Biochem ; 478(10): 2271-2279, 2023 Oct.
Article in English | MEDLINE | ID: mdl-36652045

ABSTRACT

Severe acute respiratory syndrome coronavirus type 2 (SARS-CoV-2) infection leads to hyper-inflammation and amplified immune response in severe cases that may progress to cytokine storm and multi-organ injuries like acute respiratory distress syndrome and acute lung injury. In addition to pro-inflammatory cytokines, different mediators are involved in SARS-CoV-2 pathogenesis and infection, such as sphingosine-1-phosphate (S1P). S1P is a bioactive lipid found at a high level in plasma, and it is synthesized from sphingomyelin by the action of sphingosine kinase. It is involved in inflammation, immunity, angiogenesis, vascular permeability, and lymphocyte trafficking through G-protein coupled S1P receptors. Reduction of the circulating S1P level correlates with COVID-19 severity. S1P binding to sphingosine-1-phosphate receptor 1 (S1PR1) elicits endothelial protection and anti-inflammatory effects during SARS-CoV-2 infection, by limiting excessive INF-α response and hindering mitogen-activated protein kinase and nuclear factor kappa B action. However, binding to S1PR2 opposes the effect of S1PR1 with vascular inflammation, endothelial permeability, and dysfunction as the concomitant outcome. This binding also promotes nod-like receptor pyrin 3 (NLRP3) inflammasome activation, causing liver inflammation and fibrogenesis. Thus, higher expression of macrophage S1PR2 contributes to the activation of the NLRP3 inflammasome and the release of pro-inflammatory cytokines. In conclusion, S1PR1 agonists and S1PR2 antagonists might effectively manage COVID-19 and its severe effects. Further studies are recommended to elucidate the potential conflict in the effects of S1P in COVID-19.


Subject(s)
COVID-19 , Humans , Receptors, Lysosphingolipid/agonists , Receptors, Lysosphingolipid/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein , Inflammasomes , SARS-CoV-2 , Sphingosine , Cytokines/metabolism , Lysophospholipids/metabolism , Inflammation
6.
Front Immunol ; 12: 759276, 2021.
Article in English | MEDLINE | ID: mdl-34992595

ABSTRACT

Background: Psoriasis is an autoimmune skin disease associated with lipid metabolism. Sphingosine-1-phosphate (S1P) is a bioactive lipid that plays a key role in the development of autoimmune diseases. However, there is currently a lack of comprehensive evidence of the effectiveness of S1P on psoriasis. Objective: To assess the efficacy and possible mechanism of S1P and its signal modulators in the treatment of psoriasis-like dermatitis. Methods: Six databases were searched through May 8, 2021, for studies reporting S1P and its signal modulators. Two reviewers independently extracted information from the enrolled studies. Methodological quality was assessed using SYRCLE's risk of bias tool. RevMan 5.3 software was used to analyze the data. For clinical studies, the Psoriasis Area and Severity Index score were the main outcomes. For preclinical studies, we clarified the role of S1P and its regulators in psoriasis in terms of phenotype and mechanism. Results: One randomized double-blind placebo-controlled trial and nine animal studies were included in this study. The pooled results showed that compared with control treatment, S1P receptor agonists [mean difference (MD): -6.80; 95% confidence interval (CI): -8.23 to -5.38; p<0.00001], and sphingosine kinase 2 inhibitors (MD: -0.95; 95% CI: -1.26 to -0.65; p<0.00001) alleviated psoriasis-like dermatitis in mice. The mechanism of S1P receptor agonists in treating psoriasis might be related to a decrease in the number of white blood cells, topical lymph node weight, interleukin-23 mRNA levels, and percentage of CD3+ T cells (p<0.05). Sphingosine kinase 2 inhibitors ameliorated psoriasis in mice, possibly by reducing spleen weight and cell numbers (p<0.05). Conclusions: S1P receptor agonists and sphingosine kinase 2 inhibitors could be potential methods for treating psoriasis by decreasing immune responses and inflammatory factors.


Subject(s)
Lysophospholipids/immunology , Psoriasis/immunology , Sphingosine/analogs & derivatives , Animals , Enzyme Inhibitors/pharmacology , Humans , Immunosuppressive Agents/pharmacology , Mice , Phosphotransferases (Alcohol Group Acceptor)/antagonists & inhibitors , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Psoriasis/drug therapy , Randomized Controlled Trials as Topic , Receptors, Lysosphingolipid/agonists , Software , Sphingosine/immunology
7.
J Neuroinflammation ; 16(1): 54, 2019 Mar 02.
Article in English | MEDLINE | ID: mdl-30825874

ABSTRACT

BACKGROUND: Chronic inflammatory demyelinating polyradiculoneuropathy (CIDP) is an autoimmune-mediated inflammatory disease of the peripheral nervous system characterized by a response directed against certain myelin proteins and for which therapies are limited. Previous studies have suggested a beneficial role of FTY720, a sphingosine 1-phosphate (S1P) receptor agonist, known to deplete lymphocytes from the peripheral blood by sequestering them into lymph nodes, in the treatment of experimental autoimmune neuritis (EAN). Therefore, we investigated whether FTY720 is also beneficial in chronic experimental autoimmune neuritis (c-EAN), a recently developed rat model mimicking human CIDP. METHODS: c-EAN was induced in Lewis rats by immunization with S-palm P0(180-199) peptide. Rats were treated with FTY720 (1 mg/kg) or vehicle intraperitoneally once daily from the onset of clinical signs for 18 days; clinical signs were assessed daily until 60 days post-immunization (dpi). Electrophysiological and histological features were examined at different time points. We also evaluated the serum levels of different pro- and anti-inflammatory cytokines by ELISA or flow cytometry at 18, 40, and 60 dpi. RESULTS: Our data demonstrate that FTY720 decreased the severity and abolished the chronicity of the disease in c-EAN rats. Therapeutic FTY720 treatment reversed electrophysiological and histological anomalies, suggesting that myelinated fibers were subsequently preserved, it inhibited macrophage and IL-17+ cell infiltration in PNS, and it significantly reduced circulating pro-inflammatory cytokines. CONCLUSIONS: FTY720 treatment has beneficial effects on c-EAN, a new animal model mimicking human CIDP. We have shown that FTY720 is an effective immunomodulatory agent, improving the disease course of c-EAN, preserving the myelinated fibers, attenuating the axonal degeneration, and decreasing the number of infiltrated inflammatory cells in peripheral nerves. These data confirm the interest of testing FTY720 or molecules targeting S1P in human peripheral neuropathies.


Subject(s)
Fingolimod Hydrochloride/pharmacology , Immunosuppressive Agents/pharmacology , Neuritis, Autoimmune, Experimental/pathology , Sciatic Nerve/drug effects , Sciatic Nerve/pathology , Animals , Male , Neurites/drug effects , Polyradiculoneuropathy, Chronic Inflammatory Demyelinating , Rats , Rats, Inbred Lew , Receptors, Lysosphingolipid/agonists , Severity of Illness Index
8.
Acta Biochim Biophys Sin (Shanghai) ; 51(4): 402-410, 2019 Apr 01.
Article in English | MEDLINE | ID: mdl-30877755

ABSTRACT

Epicardial progenitor cells (EpiCs) which are derived from the proepicardium have the potential to differentiate into coronary vascular smooth muscle cells during development. Whether sphingosine 1-phosphate (S1P), a highly hydrophobic zwitterionic lysophospholipid in signal transduction, induces the differentiation of EpiCs is unknown. In the present study, we demonstrated that S1P significantly induced the expression of smooth muscle cell specific markers α-smooth muscle actin and myosin heavy chain 11 in the EpiCs. And the smooth muscle cells differentiated from the EpiCs stimulated by S1P were further evaluated by gel contraction assay. To further confirm the major subtype of sphingosine 1-phosphate receptors (S1PRs) involved in the differentiation of EpiCs, we used the agonists and antagonists of different S1PRs. The results showed that the S1P1/S1P3 antagonist VPC23019 and the S1P2 antagonist JTE013 significantly attenuated EpiCs differentiation, while the S1P1 agonist SEW2871 and antagonist W146 did not affect EpiCs differentiation. These results collectively suggested that S1P, principally through its receptor S1P3, increases EpiCs differentiation into VSMCs and thus indicated the importance of S1P signaling in the embryonic coronary vasculature, while S1P2 plays a secondary role.


Subject(s)
Cell Differentiation/drug effects , Lysophospholipids/pharmacology , Mouse Embryonic Stem Cells/cytology , Myocytes, Smooth Muscle/drug effects , Pericardium/cytology , Sphingosine/analogs & derivatives , Actins/genetics , Actins/metabolism , Animals , Cell Differentiation/genetics , Cells, Cultured , Gene Expression Regulation/drug effects , Mice , Mice, Inbred C57BL , Mouse Embryonic Stem Cells/metabolism , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/metabolism , Pericardium/embryology , Phosphoserine/analogs & derivatives , Phosphoserine/pharmacology , Pyrazoles/pharmacology , Pyridines/pharmacology , Receptors, Lysosphingolipid/agonists , Receptors, Lysosphingolipid/antagonists & inhibitors , Receptors, Lysosphingolipid/genetics , Sphingosine/pharmacology
9.
Pharmazie ; 74(2): 107-110, 2019 02 01.
Article in English | MEDLINE | ID: mdl-30782260

ABSTRACT

SYL-927 is a selective sphingosine-1-phosphate receptor 1 (S1P1) agonist for autoimmune diseases. It undergoes phosphorylation to the active SYL-927-P in vivo, which activates S1P1 on lymphocytes, causing lymphopenia by retention of lymphocytes in the lymph nodes. The aim of this study was to identify the involvement of blood cells in the phosphorylation of SYL-927. In addition, pharmacokinetics of SYL-927 and SYL-927-P in blood and plasma were compared in rats. The results demonstrated that SYL-927 can be converted to SYL-927-P in rat blood, but not in rat plasma. However, both rat blood and plasma are capable of dephosphorylating SYL-927-P to SYL-927. SYL-927-P generation and release were observed after incubating SYL-927 with rat and human erythrocytes and platelets. The addition of sphingosine kinases (SPHKs) inhibitors N,N-dimethylsphingosine (DMS) and FTY720 significantly inhibited SYL-927-P generation, indicating the involvement of SPHKs. In addition, SYL-927 and SYL-927-P levels in blood were significantly higher than those in plasma after oral administration of SYL-927 in rats, suggesting the blood cells for the production of SYL-927-P. In summary, the blood cells such as erythrocytes and platelets contribute to the generation and release of SYL-927-P, which is important for maintaining plasma active phosphate levels for prolonged effects.


Subject(s)
Blood Platelets/metabolism , Erythrocytes/metabolism , Fingolimod Hydrochloride/analogs & derivatives , Immunosuppressive Agents/blood , Receptors, Lysosphingolipid/agonists , Administration, Oral , Animals , Fingolimod Hydrochloride/administration & dosage , Fingolimod Hydrochloride/blood , Humans , Immunosuppressive Agents/administration & dosage , Phosphorylation , Phosphotransferases (Alcohol Group Acceptor)/antagonists & inhibitors , Phosphotransferases (Alcohol Group Acceptor)/blood , Rats , Sphingosine/administration & dosage , Sphingosine/analogs & derivatives , Sphingosine/blood
10.
Heart Vessels ; 34(6): 1052-1063, 2019 Jun.
Article in English | MEDLINE | ID: mdl-30604190

ABSTRACT

It has been demonstrated that S1P receptors affect heart ischaemia-reperfusion (IR) induced injury. However, whether S1P receptors affect IR-induced cardiac death has not been investigated. The aim of this paper is to demonstrate the role of S1P receptors in IR-induced cardiac death. Healthy adult male Sprague-Dawley rats were assigned to the following groups: non-operation control group, sham operation group, IR group, IR group pretreated with DMSO, IR group pretreated with S1P3 agonist, IR group pretreated with an antagonist of S1P3, IR group pretreated with S1P2 and S1P3 antagonists, IR group pretreated with heptanol and antagonists of S1P2/3, and IR group pretreated with Gap26 and antagonists of S1P2/3 (heptanol acts as a Cx43 uncoupler and the mimic peptide Gap26 as Cx43 blocker). The groups with S1P2 or S1P3 agonist application before reperfusion were used to assess whether these can be used for therapy of IR. The haemodynamics, electrocardiograms (ECG), infarction area, and mortality rates were recorded. Immunohistological connexin 43 (Cx43) expression in the heart was detected in each group. Blocking S1P2/3 receptors with specific antagonists resulted in an increment of IR-induced mortality, increased infarction size, redistribution of Cx43 expression, as well as affecting the heart function. The infarction size, heart function, and mortality were totally or partially restored in the S1P2, S1P3 agonist-pretreated IR group, and the heptanol/Gap26-treated S1P2/3-blocked IR group. The S1P receptor S1P2/3 and Cx43 are involved in the IR-induced cardiac death.


Subject(s)
Death, Sudden, Cardiac/prevention & control , Myocardial Ischemia/pathology , Myocardial Reperfusion Injury/pathology , Peptides/pharmacology , Receptors, Lysosphingolipid/metabolism , Animals , Connexin 43/antagonists & inhibitors , Connexin 43/metabolism , Death, Sudden, Cardiac/etiology , Heptanol/pharmacology , Male , Rats , Rats, Sprague-Dawley , Receptors, Lysosphingolipid/agonists , Receptors, Lysosphingolipid/antagonists & inhibitors , Signal Transduction/drug effects , Sphingosine-1-Phosphate Receptors
11.
Mult Scler Relat Disord ; 27: 276-280, 2019 Jan.
Article in English | MEDLINE | ID: mdl-30447536

ABSTRACT

BACKGROUND: Fingolimod, a sphingosine-1-phosphate receptor agonist, is used for treatment of relapsing-remitting multiple sclerosis (RRMS). S1P receptors that fingolimod acts upon have also been shown to be expressed on atrial myocytes. This expression pattern has been linked with the drug's cardiovascular effects, such as bradycardia. We aimed to evaluate the clinical and electrocardiographic predictors of heart rate (HR) reduction in patients receiving first-dose fingolimod. METHODS: We retrospectively analyzed subjects diagnosed with RRMS who were allocated to fingolimod treatment. HR, systolic and diastolic blood pressure values and electrocardiography during the first dose of fingolimod were accessed. RESULTS: A total of 114 RRMS patients (65.8% female, 33.58 ±â€¯8.63 years) were included. After the initial dose of fingolimod, the heart rate decreased significantly at each hour (each p < 0.001). Nadir heart rate was reached at 4 h. The multivariate binary logistic regression analysis revealed that BMI (OR: 0.878, p = 0.045), optic nerve involvement (OR: 3.205, p = 0.018), baseline HR (OR: 1.079, p = 0.002) and T-peak-T-end interval (OR: 1.046, p = 0.030) were independent predictors of greater HR reduction. During 6-h monitorization, none of the patients had relevant adverse reactions. CONCLUSION: Our findings provide an insight on clinical and electrocardiographic predictors of HR reduction that occurs in RRMS patients receiving first dose of fingolimod.


Subject(s)
Bradycardia/chemically induced , Bradycardia/diagnosis , Fingolimod Hydrochloride/adverse effects , Heart Rate/drug effects , Multiple Sclerosis, Relapsing-Remitting/drug therapy , Adult , Electrocardiography , Female , Humans , Male , Multiple Sclerosis, Relapsing-Remitting/complications , Multiple Sclerosis, Relapsing-Remitting/physiopathology , Receptors, Lysosphingolipid/agonists , Retrospective Studies
12.
Infect Immun ; 87(2)2019 02.
Article in English | MEDLINE | ID: mdl-30510103

ABSTRACT

Incidence of whooping cough (pertussis), a bacterial infection of the respiratory tract caused by the bacterium Bordetella pertussis, has reached levels not seen since the 1950s. Antibiotics fail to improve the course of disease unless administered early in infection. Therefore, there is an urgent need for the development of antipertussis therapeutics. Sphingosine-1-phosphate receptor (S1PR) agonists have been shown to reduce pulmonary inflammation during Bordetella pertussis infection in mouse models. However, the mechanisms by which S1PR agonists attenuate pertussis disease are unknown. We report the results of a transcriptome sequencing study examining pulmonary transcriptional responses in B. pertussis-infected mice treated with S1PR agonist AAL-R or vehicle control. This study identified peptidoglycan recognition protein 4 (PGLYRP4) as one of the most highly upregulated genes in the lungs of infected mice following S1PR agonism. PGLYRP4, a secreted, innate mediator of host defenses, was found to limit early inflammatory pathology in knockout mouse studies. Further, S1PR agonist AAL-R failed to attenuate pertussis disease in PGLYRP4 knockout (KO) mice. B. pertussis virulence factor tracheal cytotoxin (TCT), a secreted peptidoglycan breakdown product, induces host tissue damage. TCT-oversecreting strains were found to drive an early inflammatory response similar to that observed in PGLYRP4 KO mice. Further, TCT-oversecreting strains induced significantly greater pathology in PGLYRP4-deficient animals than their wild-type counterparts. Together, these data indicate that S1PR agonist-mediated protection against pertussis disease is PGLYRP4 dependent. Our data suggest PGLYRP4 functions, in part, by preventing TCT-induced airway damage.


Subject(s)
Bordetella pertussis/immunology , Carrier Proteins/metabolism , Receptors, Lysosphingolipid/agonists , Whooping Cough/immunology , Animals , Disease Models, Animal , Gene Expression Regulation, Bacterial/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout
13.
Bioorg Chem ; 82: 41-57, 2019 02.
Article in English | MEDLINE | ID: mdl-30268973

ABSTRACT

Agonism of S1P1 receptor has been proven to be responsible for peripheral blood lymphopenia and elicts the identification of various S1P1 modulators. In this paper we described a series of oxadiazole-based S1P1 direct-acting agonists disubstituted on terminal benzene ring, with high potency for S1P1 receptor and favorable selectivity against S1P3 receptor. In addition, two representative agents named 16-3b and 16-3g demonstrated impressive efficacy in lymphocyte reduction along with reduced effect on heart rate when orally administered. Furthermore, these compounds have been shown to possess desired pharmacokinetic (PK) and physicochemical profiles. The binding mode between 16-3b and the activated S1P1 model was also studied.


Subject(s)
Oxadiazoles/pharmacology , Receptors, Lysosphingolipid/agonists , Animals , CHO Cells , Cricetulus , Drug Design , Humans , Lymphocytes/drug effects , Lymphopenia/chemically induced , Male , Mice , Molecular Docking Simulation , Molecular Structure , Oxadiazoles/chemical synthesis , Oxadiazoles/chemistry , Oxadiazoles/pharmacokinetics , Rats, Sprague-Dawley , Sphingosine-1-Phosphate Receptors , Structure-Activity Relationship
14.
Folia Neuropathol ; 56(3): 196-205, 2018.
Article in English | MEDLINE | ID: mdl-30509041

ABSTRACT

Alzheimer's disease (AD) is characterized by alterations of amyloid precursor protein (APP) metabolism, accumulation of amyloid  peptides (A), hyperphosphorylation of Tau proteins and also by sphingolipids disturbances. These changes lead to oxidative stress, mitochondria dysfunction, synaptic loss and neuro-inflammation. It is known that A may promote ceramides formation and reversely, ceramides could stimulate A peptides release. However, the effect of ceramide and sphingosine-1-phosphate (S1P) on APP metabolism has not been fully elucidated. In this study we investigated the role of ceramide and S1P on APP metabolism. Moreover, the effect of ceramide and SEW 2871 (agonist for S1P receptor-1) on Sirt1 (NAD+-dependent nuclear enzyme responsible for stress response) gene expression under A toxicity was analyzed. Experiments were carried out using pheochromocytoma cells (PC-12) transfected with: an empty vector (used as a control), human wild-type APP gene (APPwt) and Swedish mutated (K670M/N671L) APP gene (APPsw). Our results indicated that C2-ceramide significantly decreased the viability of the APPwt, APPsw as well as empty vector-transfected PC12 cells. It was observed that C2-ceramide had no significant effect on the mRNA level of - and -secretase in APPwt and APPsw cells. However, it significantly decreased transcription of -secretase in control cells. Results also showed a significant increase in Psen1 (crucial subunit of -secretase) gene expression in APPsw cells after incubation with C2-ceramide. We observed that SEW 2871 significantly upregulated the mRNA level of -secretase in control-empty vector-transfected cells subjected to C2-ceramide toxicity. The same tendency, though insignificant, was observed in APPwt and APPsw cells. Moreover, SEW 2871 enhanced the mRNA level of -secretase and Psen1 in APPsw cells after C2-ceramide treatment. Additionally, SEW 2871 significantly upregulated a gene expression of Sirt1 in APPwt and also APPsw cells subjected to C2-ceramide toxicity. Furthermore, it was observed that SEW 2871 significantly enhanced the viability of all investigated cells' lines probably through its positive influence on Sirt1.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Protein Precursor/metabolism , Ceramides/pharmacology , Lysophospholipids/metabolism , Oxadiazoles/pharmacology , Sphingosine/analogs & derivatives , Thiophenes/pharmacology , Animals , Ceramides/metabolism , Humans , Models, Theoretical , Neurons/metabolism , Oxadiazoles/metabolism , PC12 Cells , Rats , Receptors, Lysosphingolipid/agonists , Sphingosine/metabolism , Thiophenes/metabolism , Transcription, Genetic/drug effects
15.
Bioorg Med Chem Lett ; 28(23-24): 3585-3591, 2018 12 15.
Article in English | MEDLINE | ID: mdl-30409535

ABSTRACT

Sphingolipids represent an essential class of lipids found in all eukaryotes, and strongly influence cellular signal transduction. Autoimmune diseases like asthma and multiple sclerosis (MS) are mediated by the sphingosine-1-phosphate receptor 1 (S1P1) to express a variety of symptoms and disease patterns. Inspired by its natural substrate, an array of artificial sphingolipid derivatives has been developed to target this specific G protein-coupled receptor (GPCR) in an attempt to suppress autoimmune disorders. FTY720, also known as fingolimod, is the first oral disease-modifying therapy for MS on the market. In pursuit of improved stability, bioavailability, and efficiency, structural analogues of this initial prodrug have emerged over time. This review covers a brief introduction to the sphingolipid metabolism, the mechanism of action on S1P1, and an updated overview of synthetic sphingosine S1P1 agonists.


Subject(s)
Autoimmune Diseases/pathology , Receptors, Lysosphingolipid/metabolism , Autoimmune Diseases/drug therapy , Autoimmune Diseases/metabolism , Biological Availability , Fingolimod Hydrochloride/chemistry , Fingolimod Hydrochloride/pharmacokinetics , Fingolimod Hydrochloride/therapeutic use , Humans , Immunosuppressive Agents/chemistry , Immunosuppressive Agents/pharmacokinetics , Immunosuppressive Agents/therapeutic use , Multiple Sclerosis/drug therapy , Multiple Sclerosis/metabolism , Multiple Sclerosis/pathology , Oxadiazoles/chemistry , Oxadiazoles/pharmacokinetics , Oxadiazoles/therapeutic use , Receptors, Lysosphingolipid/agonists , Receptors, Lysosphingolipid/antagonists & inhibitors , Sphingolipids/metabolism
16.
Chem Commun (Camb) ; 54(90): 12758-12761, 2018 Nov 08.
Article in English | MEDLINE | ID: mdl-30362470

ABSTRACT

Ginkgolic acid obtained as a sphingomyelin synthase inhibitor from a plant extract library inspired the concept of sphingolipid mimics. Ginkgolic acid-derived N-acyl anilines and ginkgolic acid 2-phosphate (GA2P) respectively mimic ceramide and sphingosine 1-phosphate (S1P) in structure and function. The GA2P-induced phosphorylation of ERK and internalization of S1P receptor 1 (S1P1) indicated potent agonist activity. Docking studies revealed that GA2P adopts a similar binding conformation to the bound ligand ML5, which is a strong antagonist of S1P1.


Subject(s)
Biological Products/pharmacology , Enzyme Inhibitors/pharmacology , Receptors, Lysosphingolipid/agonists , Salicylates/pharmacology , Sphingolipids/agonists , Transferases (Other Substituted Phosphate Groups)/antagonists & inhibitors , Animals , Biological Products/chemical synthesis , Biological Products/chemistry , CHO Cells , Cricetulus , Dose-Response Relationship, Drug , Drug Design , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Humans , Molecular Docking Simulation , Molecular Structure , Receptors, Lysosphingolipid/metabolism , Salicylates/chemical synthesis , Salicylates/chemistry , Sphingolipids/metabolism , Structure-Activity Relationship , Transferases (Other Substituted Phosphate Groups)/metabolism
17.
Front Immunol ; 9: 1696, 2018.
Article in English | MEDLINE | ID: mdl-30127782

ABSTRACT

Hypoxic-ischemic injury to the developing brain remains a major cause of significant long-term morbidity and mortality. Emerging evidence from neonatal brain injury models suggests a detrimental role for peripheral lymphocytes. The immunomodulatory substance FTY720, a sphingosine-1-phosphate receptor agonist, was shown to reduce adult ischemia-induced neurodegeneration through its lymphopenic mode of action. In the present study, we hypothesized that FTY720 promotes neuroprotection by reducing peripheral lymphocytes and their infiltration into the injured neonatal brain. Term-born equivalent postnatal day 9 C57BL/6 mice were exposed to hypoxia ischemia (HI) followed by a single injection of 1 mg/kg FTY720 or vehicle (0.9% sodium chloride). Brain injury, microglia, and endothelial activation were assessed 7 days post HI using histology and western blot. Peripheral and cerebral leukocyte subsets were analyzed by multichannel flow cytometry. Whether FTY720s' effects could be attributed to its lymphopenic mode of action was determined in T cell-depleted mice. In contrast to our hypothesis, FTY720 exacerbated HI-induced neuropathology including loss of gray and white matter structures. While microglia and endothelial activation remained unchanged, FTY720 induced a strong and sustained depletion of peripheral T cells resulting in significantly reduced cerebral infiltration of CD4 T cells. CD4 T cell subset analysis revealed that circulating regulatory and effector T cells counts were similarly decreased after FTY720 treatment. However, since neonatal HI per se induces a selective infiltration of Foxp3 positive regulatory T cells compared to Foxp3 negative effector T cells effects of FTY720 on cerebral regulatory T cell infiltration were more pronounced than on effector T cells. Reductions in T lymphocytes, and particularly regulatory T cells coincided with an increased infiltration of innate immune cells, mainly neutrophils and inflammatory macrophages. Importantly anti-CD3-mediated T cell depletion resulted in a similar exacerbation of brain injury, which was not further enhanced by an additional FTY720 treatment. In summary, peripheral T cell depletion by FTY720 resulted in increased infiltration of innate immune cells concomitant to reduced T cell infiltration and exacerbation HI-induced brain injury. This study indicates that neonatal T cells may promote endogenous neuroprotection in the term-born equivalent hypoxic-ischemic brain potentially providing new opportunities for therapeutic intervention.


Subject(s)
Fingolimod Hydrochloride/pharmacology , Hypoxia-Ischemia, Brain/immunology , Immunosuppressive Agents/pharmacology , Lymphocyte Depletion , Neuroprotective Agents/pharmacology , T-Lymphocytes/drug effects , Animals , Animals, Newborn , Brain/drug effects , Brain/immunology , Female , Inflammation , Lymphocyte Subsets/immunology , Male , Mice , Mice, Inbred C57BL , Receptors, Lysosphingolipid/agonists , T-Lymphocytes, Regulatory/drug effects
18.
J Crohns Colitis ; 12(suppl_2): S633-S640, 2018 Aug 22.
Article in English | MEDLINE | ID: mdl-30137311

ABSTRACT

Studies in the 1990s using animal models of intestinal inflammation delineated the crucial molecules involved in leukocyte attraction and retention to the inflamed gut and associated lymphoid tissues. The first drug targeting leukocyte trafficking tested in inflammatory bowel diseases was the anti-ICAM-1 antisense oligonucleotide alicaforsen, showing only modest efficacy. Subsequently, the anti-α4 monoclonal antibody natalizumab proved efficacious for induction and maintenance of remission in Crohn's disease, but was associated with progressive multifocal leukoencephalopathy due to its ability to interfere with both α4ß1 and α4ß7 function. Later developments in this area took advantage of the fairly selective expression of MAdCAM-1 in the digestive organs, showing that vedolizumab, a more specific monoclonal antibody selectively blocking MAdCAM-1 binding to integrin α4ß7, was efficacious for induction and maintenance of remission in ulcerative colitis and Crohn's disease, and it was not associated with neurological complications. Currently, other drugs targeting the ß7 subunit, immunoglobulin superfamily molecules expressed on the endothelium, as well as blockade of lymphocyte recirculation in lymph nodes through modulation of sphingosine 1-phosphate receptors are under development. The potential use and risks of combined anti-trafficking therapy will be examined in this review.


Subject(s)
Chemotaxis, Leukocyte/drug effects , Gastrointestinal Agents/therapeutic use , Inflammatory Bowel Diseases/drug therapy , Inflammatory Bowel Diseases/immunology , Integrins/antagonists & inhibitors , Leukocytes/physiology , Animals , Antibodies, Monoclonal, Humanized/therapeutic use , Cell Adhesion Molecules , Cell Communication/drug effects , Endothelial Cells/physiology , Fingolimod Hydrochloride/therapeutic use , Gastrointestinal Agents/pharmacology , Gastrointestinal Tract/immunology , Humans , Immunoglobulins , Immunosuppressive Agents/therapeutic use , Indans/therapeutic use , Intercellular Adhesion Molecule-1 , Lysophospholipids/agonists , Mucoproteins/antagonists & inhibitors , Natalizumab/therapeutic use , Oxadiazoles/therapeutic use , Phosphorothioate Oligonucleotides/therapeutic use , Receptors, Lysosphingolipid/agonists , Sphingosine/agonists , Sphingosine/analogs & derivatives
19.
J Pharmacol Exp Ther ; 366(3): 509-518, 2018 09.
Article in English | MEDLINE | ID: mdl-29945931

ABSTRACT

The immunomodulatory prodrug 2-amino-2-(2-[4-octylphenyl]ethyl)-1,3-propanediol (FTY720), which acts as an agonist for sphingosine-1-phosphate (S1P) receptors (S1PR) when phosphorylated, is proposed as a novel pain therapeutic. In this study, we assessed FTY720-mediated antinociception in the radiant heat tail-flick test and in the chronic constriction injury (CCI) model of neuropathic pain in mice. FTY720 produced antinociception and antiallodynia, respectively, and these effects were dose-dependent and mimicked by the S1PR1-selective agonist CYM-5442. Repeated administration of FTY720 for 1 week produced tolerance to acute thermal antinociception, but not to antiallodynia in the CCI model. S1PR-stimulated [35S]GTPγS autoradiography revealed apparent desensitization of G protein activation by S1P or the S1PR1 agonist 5-[4-phenyl-5-(trifluoromethyl)-2-thienyl]-3-[3-(trifluoromethyl)phenyl]-1,2,4-oxadiazole (SEW-2871) throughout the brain. Similar results were seen in spinal cord membranes, whereby the Emax value of S1PR-stimulated [35S]GTPγS binding was greatly reduced in repeated FTY720-treated mice. These results suggest that S1PR1 is a primary target of FTY720 in alleviating both acute thermal nociception and chronic neuropathic nociception. Furthermore, the finding that tolerance develops to antinociception in the tail-flick test but not in chronic neuropathic pain suggests a differential mechanism of FTY720 action between these models. The observation that repeated FTY720 administration led to desensitized S1PR1 signaling throughout the central nervous system suggests the possibility that S1PR1 activation drives the acute thermal antinociceptive effects, whereas S1PR1 desensitization mediates the following: 1) tolerance to thermal antinociceptive actions of FTY720 and 2) the persistent antiallodynic effects of FTY720 in neuropathic pain by producing functional antagonism of pronociceptive S1PR1 signaling.


Subject(s)
Fingolimod Hydrochloride/pharmacology , Neuralgia/drug therapy , Neuralgia/metabolism , Opioid Peptides/drug effects , Receptors, Lysosphingolipid/metabolism , Temperature , Animals , Disease Models, Animal , Fingolimod Hydrochloride/therapeutic use , Male , Mice , Mice, Inbred ICR , Neuralgia/physiopathology , Receptors, Lysosphingolipid/agonists , Nociceptin
20.
Acta Biochim Biophys Sin (Shanghai) ; 50(7): 651-657, 2018 Jul 01.
Article in English | MEDLINE | ID: mdl-29901713

ABSTRACT

Ischemia/reperfusion (I/R) is a major cause of acute kidney injury (AKI), along with delayed graft function, which can trigger chronic kidney injury by stimulating epithelial to mesenchymal transition (EMT) in the kidney canaliculus. Sphingosine 1-phosphate receptor 1 (S1P1) is a G protein-coupled receptor that is indispensable for vessel homeostasis. This study aimed to investigate the influence of S1P1 on the mechanisms underlying I/R-induced EMT in the kidney using in vivo and in vitro models. Wild-type (WT) and S1P1-overexpressing kidney canaliculus cells were subject to hypoxic conditions followed by reoxygenation in the presence or absence of FTY720-P, a potent S1P1 agonist. In vivo, bilateral arteria renalis in wild-type mice and mice with silenced S1P1 were clamped for 30 min to obtain I/R models. We found that hypoxia/reoxygenation (H/R) significantly enhanced the expressions of EMT biomarkers and down-regulated S1P1 expression in wild-type canaliculus cells. In contrast, FTY720-P treatment or overexpression of S1P1 significantly suppressed EMT in wild-type canaliculus cells. Furthermore, after 48-72 h, a significant upregulation of EMT biomarker expression was triggered by I/R in mice with silenced S1P1, while the expressions of these markers did not change in wild-type mice. A kt activity was increased with H/R-induced EMT, suggesting that the protective influence of FTY720-P was due to its inhibition of PI3K/Akt. Therefore, the results of this study provide evidence that down-regulation of S1P1 expression is essential for the generation and progression of EMT triggered by I/R. S1P1 exhibits a prominent inhibitory effect on kidney I/R-induced EMT in the kidney by affecting the PI3K/Akt pathway.


Subject(s)
Epithelial-Mesenchymal Transition , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Lysosphingolipid/metabolism , Reperfusion Injury/metabolism , Animals , Cell Line , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Gene Expression/drug effects , Humans , Kidney/cytology , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Organophosphates/pharmacology , Receptors, Lysosphingolipid/agonists , Receptors, Lysosphingolipid/genetics , Reperfusion Injury/genetics , Signal Transduction , Sphingosine/analogs & derivatives , Sphingosine/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL