Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Int J Mol Sci ; 24(8)2023 Apr 17.
Article in English | MEDLINE | ID: mdl-37108539

ABSTRACT

FTY720 is an FDA-approved sphingosine derivative drug for the treatment of multiple sclerosis. This compound blocks lymphocyte egress from lymphoid organs and autoimmunity through sphingosine 1-phosphate (S1P) receptor blockage. Drug repurposing of FTY720 has revealed improvements in glucose metabolism and metabolic diseases. Studies also demonstrate that preconditioning with this compound preserves the ATP levels during cardiac ischemia in rats. The molecular mechanisms by which FTY720 promotes metabolism are not well understood. Here, we demonstrate that nanomolar concentrations of the phosphorylated form of FTY720 (FTY720-P), the active ligand of S1P receptor (S1PR), activates mitochondrial respiration and the mitochondrial ATP production rate in AC16 human cardiomyocyte cells. Additionally, FTY720-P increases the number of mitochondrial nucleoids, promotes mitochondrial morphology alterations, and induces activation of STAT3, a transcription factor that promotes mitochondrial function. Notably, the effect of FTY720-P on mitochondrial function was suppressed in the presence of a STAT3 inhibitor. In summary, our results suggest that FTY720 promotes the activation of mitochondrial function, in part, through a STAT3 action.


Subject(s)
Fingolimod Hydrochloride , Sphingosine , Rats , Humans , Animals , Fingolimod Hydrochloride/pharmacology , Propylene Glycols/pharmacology , Ligands , Receptors, Lysosphingolipid/metabolism , Sphingosine-1-Phosphate Receptors/metabolism , Mitochondria/metabolism , Adenosine Triphosphate , Immunosuppressive Agents/pharmacology , STAT3 Transcription Factor/metabolism
2.
Mol Neurobiol ; 60(2): 901-922, 2023 Feb.
Article in English | MEDLINE | ID: mdl-36385233

ABSTRACT

Sphingosine receptors (S1PRs) are implicated in the progression of neurodegenerative diseases and metabolic disorders like obesity and type 2 diabetes (T2D). The link between S1PRs and cognition in type 2 diabetes, as well as the mechanisms that underpin it, are yet unknown. Neuroinflammation is the common pathology shared among T2D and cognitive impairment. However, the interplay between the M1 and M2 polarization state of microglia, a primary driver of neuroinflammation, could be the driving factor for impaired learning and memory in diabetes. In the present study, we investigated the effects of fingolimod (S1PR1 modulator) on cognition in high-fat diet and streptozotocin-induced diabetic mice. We further assessed the potential pathways linking microglial polarization and cognition in T2D. Fingolimod (0.5 mg/kg and 1 mg/kg) improved M2 polarization and synaptic plasticity while ameliorating cognitive decline and neuroinflammation. Sphingolipid dysregulation was mimicked in vitro using palmitate in BV2 cells, followed by conditioned media exposure to Neuro2A cells. Mechanistically, type 2 diabetes induced microglial activation, priming microglia towards the M1 phenotype. In the hippocampus and cortex of type 2 diabetic mice, there was a substantial drop in pSTAT3, which was reversed by fingolimod. This protective effect of fingolimod on microglial M2 polarization was primarily suppressed by selective jmjd3 blockade in vitro using GSK-J4, revealing that jmjd3 was involved downstream of STAT3 in the fingolimod-enabled shift of microglia from M1 to M2 polarization state. This study suggested that fingolimod might effectively improve cognition in type 2 diabetes by promoting M2 polarization.


Subject(s)
Cognitive Dysfunction , Diabetes Mellitus, Experimental , Diabetes Mellitus, Type 2 , Animals , Mice , Cell Polarity , Cognition , Cognitive Dysfunction/complications , Cognitive Dysfunction/drug therapy , Cognitive Dysfunction/metabolism , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/metabolism , Fingolimod Hydrochloride/pharmacology , Fingolimod Hydrochloride/therapeutic use , Microglia/metabolism , Neuroinflammatory Diseases , Signal Transduction
3.
J Appl Toxicol ; 43(2): 220-229, 2023 02.
Article in English | MEDLINE | ID: mdl-35946054

ABSTRACT

FTY720P, an analogue of sphingosine 1-phosphate, has emerged lately as a potential causative agent of inflammatory bowel disease, in which electrolytes movements driven by the sodium gradient established by the Na+ /K+ ATPase are altered. We showed previously in Caco-2 cells, a 50% FTY720P-induced decrease in the ATPase activity, mediated via S1PR2 and PGE2. This work aims at delineating the mechanism underlying PGE2 release and at investigating if the ATPase inhibition is due to changes in its abundance. The activity of the ATPase and the localization of a GFP-tagged Na+ /K+ -ATPase α1 -subunit were assessed in cells treated with 7.5 nM FTY720P. The involvement of ERK, p38 MAPK, PKC, and PI3K was studied in cells treated with 7.5 nM FTY720P or 1 nM PGE2 in presence of their inhibitors, or by determining changes in the protein expression of their activated phosphorylated forms. Imaging data showed ∼30% reduction in the GFP-tagged Na+ /K+ ATPase at the plasma membrane. Both FTY720P and PGE2 showed, respectively, 50% and 60% reduction in ATPase activity that disappeared when p38 MAPK, PKC, and PI3K were inhibited individually but not with ERK inhibition. The effect of FTY720P was imitated by PMA, an activator of PKC. Western blotting revealed inhibition of ERK by FTY720P. It was concluded that FTY720P, through activation of S1PR2, downregulates the Na+ /K+ ATPase by inhibiting ERK, which in turn activates p38 MAPK leading to the sequential activation of PKC and PI3K, PGE2 release, and a decrease in the Na+ /K+ ATPase activity and membrane abundance.


Subject(s)
Protein Kinase C , Signal Transduction , Humans , Phosphatidylinositol 3-Kinases/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Sodium-Potassium-Exchanging ATPase/metabolism , Caco-2 Cells , Dinoprostone/metabolism , Dinoprostone/pharmacology , Sodium/metabolism
4.
Cell Physiol Biochem ; 56(4): 418-435, 2022 Aug 29.
Article in English | MEDLINE | ID: mdl-36041048

ABSTRACT

BACKGROUND/AIMS: In renal ischemia, the Na+/K+ ATPase of the kidney epithelial cells translocates to intracellular compartments, resulting in altered kidney functions. Sphingosine-1-phosphate (S1P) was shown to play a protective role against this ischemic injury. Whether the sphingolipid targets the Na+/K+ ATPase is a possibility that has not been explored before. This work aims at investigating the effect of S1P on renal Na+/K+ ATPase using its analogue FTY720P and LLC-PK1 cells. METHODS: The activity of the Na+/K+ ATPase was assayed by measuring the amount of inorganic phosphate liberated in presence and absence of ouabain, a specific inhibitor of the enzyme while its protein expression was studied by western blot analysis. RESULTS: FTY720P increased the activity of the ATPase in a dose and time dependent manner, with a highest effect observed at 15 minutes and a dose of 80 nM. The protein expression was also increased. The stimulation of the Na+/K+ ATPase disappeared completely in presence of JTE-013, a specific blocker of S1PR2, as well as in presence of Y-27632, a Rho kinase inhibitor, BAPTA-AM, a Ca2+ chelator, wortmannin, a PI3K inhibitor, carboxy-PTIO, a scavenger for nitric oxide (NO), and KT 5823, a PKG inhibitor. CYM 5520, a S1PR2 agonist mimicked the effect of FTY720P. FTY720P increased the expression of p-Akt, a direct effector of PI3K, however, this increase disappeared when Rho kinase was inhibited, revealing that Rho kinase acts upstream PI3K. Glyco-SNAP-1, a NO donor, activated the pump in both presence and absence of wortmannin, indicating that PI3K is upstream NO. Interestingly, glyco-SNAP-1 and 8-bromo-cGMP, a PKG activator, exerted no effect on the Na+/K+ ATPase in absence of free Ca2+ revealing that the NO mediated effect is calcium-dependent. The involvement of calcium was further confirmed by the translocation of NFAT to the nucleus. The presence of verapamil or extracellular EGTA abolished the stimulatory effect of FTY720P, indicating that the source of calcium is extracellular. CONCLUSION: The results suggest that FTY720P activates sequentially S1PR2, Rho kinase, PI3K, leading to NO release and PKG stimulation. The latter phosphorylates calcium channels in the cell membrane, leading to calcium influx, and translocation of the ATPase units to the membrane.


Subject(s)
Phosphatidylinositol 3-Kinases , rho-Associated Kinases , Animals , Calcium/metabolism , Nitric Oxide/metabolism , Organophosphates , Phosphatidylinositol 3-Kinases/metabolism , Sodium-Potassium-Exchanging ATPase/metabolism , Sphingosine/analogs & derivatives , Swine , Wortmannin/pharmacology , rho-Associated Kinases/metabolism
5.
Prostaglandins Other Lipid Mediat ; 149: 106436, 2020 08.
Article in English | MEDLINE | ID: mdl-32173486

ABSTRACT

Ischemia and reperfusion injury is a complex hemodynamic pathological phenomenon that engages the metabolic to inflammatory machinery in development of disease conditions like heart failure, stroke and acute kidney failure. Target specific therapeutic approaches for ischemia reperfusion injury remains critical despite the extensive studies contributing to the understanding of its pathogenesis. Ischemic or pharmacological conditionings have been long established manipulations to harness the endogenous protective mechanisms against ischemia reperfusion injury that fostered the development of potential therapeutic targets such as sphingolipids signaling. Sphingosine 1-phosphate has been emerged as a crucial metabolite of sphingolipids to regulate the cell survival, vascular integrity and inflammatory cascades in ischemia reperfusion injury. Sphingosine 1-phosphate signaling process has been implicated to downgrade the mitochondrial dysfunction, apoptotic assembly along with upregulation of RISK and SAFE pro-survival pathways. It also regulates the endothelial dysfunction and immune cells behavior to control the vascular permeability and immune cells infiltration at ischemia reperfusion injury site. Targeting the signaling of this single moiety holds the vast potential to extensively influence the detrimental signaling of ischemia reperfusion injury. This review highlights the role and significance of S1P signaling that can be therapeutically exploit to treat ischemia reperfusion injury mediated pathological conditions in different organs.


Subject(s)
Ischemia/pathology , Lysophospholipids/metabolism , Reperfusion Injury/pathology , Signal Transduction , Sphingosine/analogs & derivatives , Animals , Brain/blood supply , Humans , Ischemia/metabolism , Kidney/blood supply , Reperfusion Injury/metabolism , Sphingosine/metabolism
6.
Cell Physiol Biochem ; 53(3): 518-531, 2019.
Article in English | MEDLINE | ID: mdl-31502430

ABSTRACT

BACKGROUND/AIMS: Liver regeneration is induced by S1P and accompanied with an increase in hepatic Na+/K+ ATPase activity, suggesting a potential modulatory role of the sphingolipid on the ATPase activity. The ability of S1P to alter the ATPase activity was confirmed in a previous work which showed a time dependent effect, with an inhibition appearing at 15min and a stimulation at two hours. The aim of this work was to investigate if FTY720-P, an analogue of S1P used in the treatment of multiple sclerosis, exerts a similar effect at 2 hours. METHODS: HepG2 cells were treated with FTY720-P for two hours and the activity of the Na+/K+ ATPase was assayed by measuring the amount of inorganic phosphate liberated in presence and absence of ouabain. The involvement of NF-κB in the pathway was investigated by determining changes in the protein expression of IκB. RESULTS: FTY720-P induced a 2.5-fold increase in the activity of the Na+/K+ ATPase which was maintained in the presence of JTE-013, a specific blocker of S1PR2, but disappeared completely in presence of CAY 10444, a specific S1PR3 antagonist. The involvement of S1PR3 was supported by the stimulation observed with Cym5541, a S1PR3 agonist. FTY720-P increased the expression of COX2, and reduced that of IκB. Its effect was not manifested in presence of indomethacin, a COX inhibitor, or in presence of an NF-κB inhibitor. Exogenous PGE2 induced a significant stimulatory effect. Inhibiting PKC and ERK with respectively calphostin C and PD98059 abolished the effect of FTY720-P on the ATPase and on IκB, but not that of exogenous PGE2 indicating that the two kinases are upstream of NF-κB and PGE2. The PKC activator PMA increased the activity of the Na+/K+ ATPase as well as the expression of phopho-ERK, inferring that PKC is upstream of ERK. CONCLUSION: It was concluded that FTY720-P stimulates the Na+/K+ ATPase via PGE2 by activating sequentially S1PR3, PKC, ERK, NF-κB. The latter enhances COX-2 expression leading to PGE2 release.


Subject(s)
Dinoprostone/metabolism , Organophosphates/pharmacology , Receptors, Lysosphingolipid/metabolism , Sodium-Potassium-Exchanging ATPase/metabolism , Sphingosine/analogs & derivatives , Blotting, Western , Enzyme Activation/drug effects , Hep G2 Cells , Humans , MAP Kinase Signaling System/drug effects , NF-kappa B/metabolism , Protein Kinase C/metabolism , Signal Transduction/drug effects , Sphingosine/pharmacology , Sphingosine-1-Phosphate Receptors
7.
Life Sci ; 215: 198-206, 2018 Dec 15.
Article in English | MEDLINE | ID: mdl-30439376

ABSTRACT

AIMS: Sphingosine-1-phosphate (S1P) has been implicated lately in inflammatory bowel disease which has diarrhea as one of its symptoms. Diarrhea is due to altered water movements as a result of altered electrolyte transport, and in particular sodium. Sodium movements are geared by the sodium gradient established by the Na+/K+ ATPase. The aim of this work was to investigate if S1P can modulate the activity of the ATPase, using Caco-2 cells as a model and the S1P analogue, FTY720P. MATERIALS AND METHODS: The activity of the ATPase was assayed by measuring the amount of inorganic phosphate liberated in presence and absence of ouabain. Protein expression of the various S1P receptors was studied by western blot analysis. KEY FINDINGS: Caco-2 cells were found to express mainly S1PR2 and S1PR3. FTY720P (7.5 nM) reduced significantly the activity of the Na+/K+ ATPase when applied for 15 min. This inhibitory effect disappeared in presence of JTE-013, a specific blocker of S1PR2, and indomethacin, an inhibitor of cyclooxygenase enzymes, and was mimicked by CYM5520, a S1PR2 agonist and by exogenous PGE2. The inhibitory effect of PGE2 did not appear when EP3 receptors were blocked or when a nitric oxide scavenger was added. RpcAMP, a PKA inhibitor, reduced the activity of the Na+/K+ ATPase, while dbcAMP, a PKA activator was without any effect and when added, abrogated the effect of PGE2. SIGNIFICANCE: It was concluded that FTY720P inhibits the Na+/K+ ATPase via activation of S1PR2 and generation of PGE2 nitric oxide.


Subject(s)
Dinoprostone/metabolism , Nitric Oxide/metabolism , Organophosphates/pharmacology , Receptors, Lysosphingolipid/metabolism , Sodium-Potassium-Exchanging ATPase/antagonists & inhibitors , Sphingosine/analogs & derivatives , Blotting, Western , Caco-2 Cells , Humans , Indomethacin/pharmacology , Lysophospholipids/metabolism , Pyrazoles/pharmacology , Pyridines/pharmacology , Pyrroles/pharmacology , Signal Transduction/drug effects , Sodium-Potassium-Exchanging ATPase/metabolism , Sphingosine/metabolism , Sphingosine/pharmacology , Sphingosine-1-Phosphate Receptors
8.
Zhongguo Xiu Fu Chong Jian Wai Ke Za Zhi ; 32(5): 575-580, 2018 05 15.
Article in Chinese | MEDLINE | ID: mdl-29806345

ABSTRACT

Objective: To investigate the effects of FTY720-P on EphA2-EphrinA2 bidirectional signaling in osteoclasts. Methods: Murine RAW264.7 macrophages were induced into osteoclasts by dexamethasone and 1α, 25-dihydroxyvitamin D 3, and identified by tartrate resistant acid phosphatase (TRAP) staining. Then, the osteoclasts were divided into 2 groups. The osteoclasts were treated with 400 ng/mL FTY720-P in experimental group and without FTY720-P in control group, respectively. After 48 hours of culture, the cells in 2 groups were detected by real-time fluorescent quantitative PCR, Western blot, and immunofluorescence staining. The expressions of EphA2, EphrinA2, RhoA, and the bone reconstruction associated proteinsï¼»bone morphogenetic protein 2 (BMP-2) and transform growth factor ß 1 (TGF-ß 1)ï¼½were analyzed and compared. Results: RAW264.7 cells were successfully induced into osteoclasts identified by TRAP staining. Compared with control group, the relative expressions of EphA2 and EphrinA2 mRNAs and proteins in experimental group significantly decreased after 48 hours ( P<0.05), and the relative expression of RhoA protein also significantly decreased ( P<0.05). The relative expressions of BMP-2 and TGF-ß 1 mRNAs were significantly increased ( P<0.05), and those protein expressions were enhanced. Conclusion: FTY720-P can down-regulate the expression of RhoA and promote the expressions of TGF- ß 1 and BMP-2 by affecting the transduction of EphA2-EphrinA2 bidirectional signaling in osteoclasts.


Subject(s)
Cell Differentiation , Ephrin-A2/metabolism , Organophosphates/metabolism , Osteoclasts/metabolism , Receptor, EphA2/metabolism , Signal Transduction , Sphingosine/analogs & derivatives , Animals , Bone Morphogenetic Protein 2 , Macrophages , Mice , Sphingosine/metabolism
9.
Zhongguo Xiu Fu Chong Jian Wai Ke Za Zhi ; 32(3): 285-290, 2018 03 15.
Article in Chinese | MEDLINE | ID: mdl-29806276

ABSTRACT

Objective: To investigate the effect of FTY720-P on the differentiation and maturation of MC3T3-E1 cells. Methods: The MC3T3-E1 cells were divided into the experimental group and the control group. In the experimental group, the cells were induced by the medium containing 400 ng/mL FTY720-P (chloroform as solubilizer) in vitro. In the control group, the cells were cultured with the medium only containing chloroform. The cell morphology of 2 groups were observed by inverted phase contrast microscope; the expression of osteoblast related protein (collagen type Ⅰ and collagen type Ⅲ) was detected by immunofluorescence staining; the alkaline phosphatase (ALP) staining and alizarin red staining were used to observe the formation of osteoblasts and the formation of mineralized nodules in 2 groups; and the TUNEL fluorescence assay was used to detect the cell apoptosis. Results: After 48 hours of culture, the cells of 2 groups had grown into slender fusiform at the bottom of the bottle, and there was no significant difference in cell morphology between 2 groups. Immunofluorescence staining showed that the expression of collagen type Ⅰ was positive in the experimental group and weakly positive in the control group; the integrated absorbance ( IA) value of the experimental group was 187 600±7 944, which was significantly higher than that of the control group (14 230±1 070) ( t=43.680, P=0.001). The expression of collagen type Ⅲ was weakly positive in the experimental group and the control group, and there was no significant difference in IA value between 2 groups ( t=1.976, P=0.119). ALP staining and alizarin red staining were positive in the experimental group and negative in the control group. TUNEL staining was positive in the experimental group and negative in the control group; the rate of TUNEL staining positive cells in the experimental group was 35.82%±2.99%, which was significantly higher than that in the control group (2.28%±0.51%) ( t=23.420, P=0.002). Conclusion: FTY720-P can promote the osteogenic differentiation of MC3T3-E1 cells with speeding up maturation and mineralization of extracellular matrix and affect the apoptosis of the cells.


Subject(s)
Cell Differentiation/drug effects , Cell Differentiation/physiology , Organophosphates/pharmacology , Osteoblasts/physiology , Osteogenesis/drug effects , Sphingosine/analogs & derivatives , Alkaline Phosphatase , Animals , Cell Count , Cell Division/drug effects , Cell Division/physiology , Cell Line , Cells, Cultured , Collagen Type I , Mice , Sphingosine/pharmacology
10.
Article in Chinese | WPRIM (Western Pacific) | ID: wpr-856812

ABSTRACT

Methods: The MC3T3-E1 cells were divided into the experimental group and the control group. In the experimental group, the cells were induced by the medium containing 400 ng/mL FTY720-P (chloroform as solubilizer) in vitro. In the control group, the cells were cultured with the medium only containing chloroform. The cell morphology of 2 groups were observed by inverted phase contrast microscope; the expression of osteoblast related protein (collagen type Ⅰ and collagen type Ⅲ) was detected by immunofluorescence staining; the alkaline phosphatase (ALP) staining and alizarin red staining were used to observe the formation of osteoblasts and the formation of mineralized nodules in 2 groups; and the TUNEL fluorescence assay was used to detect the cell apoptosis.

11.
Article in Chinese | WPRIM (Western Pacific) | ID: wpr-856782

ABSTRACT

Objective: To investigate the effects of FTY720-P on EphA2-EphrinA2 bidirectional signaling in osteoclasts. Methods: Murine RAW264.7 macrophages were induced into osteoclasts by dexamethasone and 1α, 25-dihydroxyvitamin D 3, and identified by tartrate resistant acid phosphatase (TRAP) staining. Then, the osteoclasts were divided into 2 groups. The osteoclasts were treated with 400 ng/mL FTY720-P in experimental group and without FTY720-P in control group, respectively. After 48 hours of culture, the cells in 2 groups were detected by real-time fluorescent quantitative PCR, Western blot, and immunofluorescence staining. The expressions of EphA2, EphrinA2, RhoA, and the bone reconstruction associated proteins[bone morphogenetic protein 2 (BMP-2) and transform growth factor β 1 (TGF-β 1)]were analyzed and compared. Results: RAW264.7 cells were successfully induced into osteoclasts identified by TRAP staining. Compared with control group, the relative expressions of EphA2 and EphrinA2 mRNAs and proteins in experimental group significantly decreased after 48 hours ( P<0.05), and the relative expression of RhoA protein also significantly decreased ( P<0.05). The relative expressions of BMP-2 and TGF-β 1 mRNAs were significantly increased ( P<0.05), and those protein expressions were enhanced. Conclusion: FTY720-P can down-regulate the expression of RhoA and promote the expressions of TGF- β 1 and BMP-2 by affecting the transduction of EphA2-EphrinA2 bidirectional signaling in osteoclasts.

12.
Biochem Cell Biol ; 94(4): 371-7, 2016 08.
Article in English | MEDLINE | ID: mdl-27501354

ABSTRACT

Sphingosine-1-phosphate (S1P) was found previously to inhibit Na(+)-K(+) ATPase in HepG2 cells. Whether fingolimod (FTY720), a S1P receptor (S1PR) agonist, similarly inhibits the ATPase is a question that needs to be addressed. The aim of this work was to study the effect of FTY720P, the active form of the drug, on the activity of Na(+)-K(+) ATPase in HepG2 cells and determine its mechanism of action. The activity of the ATPase was assayed by measuring the amount of inorganic phosphate liberated in the presence and the absence of ouabain. FTY720-P (7.5 nmol/L, 15 min) significantly reduced the activity of the ATPase. This effect disappeared completely in the presence of JTE-013, which is a specific blocker of sphingosine-1-phosphate receptor 2 (S1PR2), as well as in the presence of calphostin and indomethacin, which are inhibitors of protein kinase C (PKC) and COX-2, respectively. The effect of FTY720P was mimicked by prostaglandin E2 (PGE2) and PMA, but abrogated by NF-κB inhibition. When NF-κB was inhibited, the effect of exogenous PGE2 still appeared, but that of PMA did not manifest, suggesting that NF-κB is upstream of PGE2 and downstream of PKC. It was concluded that FTY720P activates via S1PR2, PKC, and NF-κB. The latter induces PGE2 generation and inhibits Na(+)-K(+) ATPase.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Dinoprostone/metabolism , Liver Neoplasms/metabolism , Organophosphates/pharmacology , Receptors, Lysosphingolipid/metabolism , Sodium-Potassium-Exchanging ATPase/antagonists & inhibitors , Sphingosine/analogs & derivatives , Blotting, Western , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/pathology , Humans , Liver Neoplasms/drug therapy , Liver Neoplasms/pathology , Lysophospholipids/metabolism , NF-kappa B/metabolism , Protein Kinase C/metabolism , Sphingosine/metabolism , Sphingosine/pharmacology , Sphingosine-1-Phosphate Receptors , Tumor Cells, Cultured
13.
Biochem Biophys Res Commun ; 476(4): 627-634, 2016 08 05.
Article in English | MEDLINE | ID: mdl-27282481

ABSTRACT

Endothelial nitric oxide synthase (eNOS) plays a crucial role in vascular homeostasis. Lysophospholipid interaction with sphingosine 1-phosphat (S1P) receptors results in eNOS activation in different cells. In endothelial cells, eNOS activation via S1P1 or S1P3 was shown controversially. The aim of this study is to investigate the meaning of both S1P receptors for eNOS activation in human endothelial cells. Therefore, several S1P1 and S1P3 agonists in combination with antagonists and specific RNAi approach were used. eNOS activation was measured in human umbilical vein endothelial cells (HUVEC) via DAF2-DA-based fluorescence microscopy. For investigation of the signaling pathway, agonists/antagonist studies, RNAi approach, Luminex™ multiplex, and Western Blot were used. In HUVEC, both the S1P1 agonist AUY954 as well as the S1P1,3 agonist FTY720P induced eNOS activation in a time- and dose-dependent manner. Other S1P1 agonists activated eNOS to a lesser extent. The AUY954-induced eNOS activation was blocked by the S1P1 antagonist W146, the combination of W146 and the S1P3 antagonist CAY10444 and the S1P1,3 antagonist VPC23019, but not by CAY10444 indicating the meaning of S1P1 for the AUY954-induced eNOS activation. The FTY720P-induced eNOS activation was blocked only by the combination of W146 and CAY10444 and the combined S1P1,3 antagonist VPC23019, but not by W146 or CAY10444 indicating the importance of both S1P1 and S1P3 for FTY720-induced eNOS activation. These results were confirmed using specific siRNA against S1P1 and S1P3. The S1P1,3 activation results in Akt phosphorylation and subsequent activation of eNOS via phosphorylation at serine(1177) and dephosphorylation at threonine(495). Beside former investigations with rather unspecific S1P receptor activation these data show potent selective S1P1 activation by using AUY954 and with selective S1P receptor inhibition evidence was provided that both S1P1 and S1P3 lead to downstream activation of eNOS in HUVEC in the same experimental setting. Inhibition or knockdown of one of these receptor subtypes did not abolish the eNOS activation and subsequent NO production.


Subject(s)
Endothelial Cells/metabolism , Nitric Oxide Synthase Type III/metabolism , Receptors, Lysosphingolipid/metabolism , Anilides/pharmacology , Endothelial Cells/drug effects , Enzyme Activation/drug effects , Gene Knockdown Techniques , Human Umbilical Vein Endothelial Cells , Humans , Nitric Oxide/biosynthesis , Nitric Oxide Synthase Type III/chemistry , Organophosphates/pharmacology , Organophosphonates/pharmacology , Phosphorylation , Phosphoserine/analogs & derivatives , Phosphoserine/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , RNA, Small Interfering/genetics , Receptors, Lysosphingolipid/antagonists & inhibitors , Receptors, Lysosphingolipid/genetics , Signal Transduction , Sphingosine/analogs & derivatives , Sphingosine/pharmacology , Sphingosine-1-Phosphate Receptors , Thiazolidines/pharmacology , Thiophenes/pharmacology , beta-Alanine/analogs & derivatives , beta-Alanine/pharmacology
14.
Front Pharmacol ; 7: 94, 2016.
Article in English | MEDLINE | ID: mdl-27148053

ABSTRACT

Sphingosine 1-phosphate (S1P) is an immune modulatory lipid mediator and has been implicated in numerous pathophysiological processes. S1P is produced by sphingosine kinase 1 (Sphk1) and Sphk2. Dendritic cells (DCs) are central for the direction of immune responses and crucially involved in autoimmunity and cancerogenesis. In this study we examined the function and survival of bone marrow-derived DCs under long-term inflammatory stimulation. We observed that differentiated cells undergo activation-induced cell death (AICD) upon LPS stimulation with an increased metabolic activity shortly after stimulation, followed by a rapid activation of caspase 3 and subsequent augmented apoptosis. Importantly, we highlight a profound role of Sphk1 in secretion of inflammatory cytokines and survival of dendritic cells that might be mediated by a change in sphingolipid levels as well as by a change in STAT3 expression. Cell growth during differentiation of Sphk1-deficient cells treated with the functional S1P receptor antagonist FTYP was reduced. Importantly, in dendritic cells we did not observe a compensatory regulation of Sphk2 mRNA in Sphk1-deficient cells. Instead, we discovered a massive increase in Sphk1 mRNA concentration upon long-term stimulation with LPS in wild type cells that might function as an attempt to rescue from inflammation-caused cell death. Taken together, in this investigation we describe details of a crucial involvement of sphingolipids and Sphk1 in AICD during long-term immunogenic activity of DCs that might play an important role in autoimmunity and might explain the differences in immune response observed in in vivo studies of Sphk1 modulation.

15.
Int J Mol Sci ; 16(12): 29454-66, 2015 Dec 10.
Article in English | MEDLINE | ID: mdl-26690412

ABSTRACT

Breakdown of the blood-brain barrier (BBB) is an early hallmark of multiple sclerosis (MS), a progressive inflammatory disease of the central nervous system. Cell adhesion in the BBB is modulated by sphingosine-1-phosphate (S1P), a signaling protein, via S1P receptors (S1P1). Fingolimod phosphate (FTY720-P) a functional S1P1 antagonist has been shown to improve the relapse rate in relapsing-remitting MS by preventing the egress of lymphocytes from lymph nodes. However, its role in modulating BBB permeability-in particular, on the tight junction proteins occludin, claudin 5 and ZO-1-has not been well elucidated to date. In the present study, FTY720-P did not change the transendothelial electrical resistance in a rat brain microvascular endothelial cell (RBMEC) culture exposed to inflammatory conditions and thus did not decrease endothelial barrier permeability. In contrast, occludin was reduced in RBMEC culture after adding FTY720-P. Additionally, FTY720-P did not alter the amount of endothelial matrix metalloproteinase (MMP)-9 and MMP-2 in RBMEC cultures. Taken together, our observations support the assumption that S1P1 plays a dual role in vascular permeability, depending on its ligand. Thus, S1P1 provides a mechanistic basis for FTY720-P-associated disruption of endothelial barriers-such as the blood-retinal barrier-which might result in macular edema.


Subject(s)
Capillary Permeability/drug effects , Endothelial Cells/drug effects , Fingolimod Hydrochloride/pharmacology , Immunosuppressive Agents/pharmacology , Animals , Apoptosis , Blood-Brain Barrier/pathology , Cells, Cultured , Drug Evaluation, Preclinical , Endothelial Cells/metabolism , MAP Kinase Signaling System , Matrix Metalloproteinase 2/metabolism , Microvessels/pathology , Multiple Sclerosis/drug therapy , Multiple Sclerosis/pathology , Occludin/metabolism , Rats
SELECTION OF CITATIONS
SEARCH DETAIL
...