Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 121
Filter
1.
JCI Insight ; 9(11)2024 Jun 10.
Article in English | MEDLINE | ID: mdl-38855867

ABSTRACT

In rheumatoid arthritis, inflammatory mediators extravasate from blood into joints via gaps between endothelial cells (ECs), but the contribution of ECs is not known. Sphingosine 1-phosphate receptor 1 (S1PR1), widely expressed on ECs, maintains the vascular barrier. Here, we assessed the contribution of vascular integrity and EC S1PR1 signaling to joint damage in mice exposed to serum-induced arthritis (SIA). EC-specific deletion of S1PR1 or pharmacological blockade of S1PR1 promoted vascular leak and amplified SIA, whereas overexpression of EC S1PR1 or treatment with an S1PR1 agonist delayed SIA. Blockade of EC S1PR1 induced membrane metalloproteinase-dependent cleavage of vascular endothelial cadherin (VE-cadherin), a principal adhesion molecule that maintains EC junctional integrity. We identified a disintegrin and a metalloproteinase domain 10 (ADAM10) as the principal VE-cadherin "sheddase." Mice expressing a stabilized VE-cadherin construct had decreased extravascular VE-cadherin and vascular leakage in response to S1PR1 blockade, and they were protected from SIA. Importantly, patients with active rheumatoid arthritis had decreased circulating S1P and microvascular expression of S1PR1, suggesting a dysregulated S1P/S1PR1 axis favoring vascular permeability and vulnerability. We present a model in which EC S1PR1 signaling maintains homeostatic vascular barrier function by limiting VE-cadherin shedding mediated by ADAM10 and suggest this signaling axis as a therapeutic target in inflammatory arthritis.


Subject(s)
ADAM10 Protein , Antigens, CD , Arthritis, Experimental , Arthritis, Rheumatoid , Cadherins , Endothelial Cells , Sphingosine-1-Phosphate Receptors , Animals , Cadherins/metabolism , Sphingosine-1-Phosphate Receptors/metabolism , Sphingosine-1-Phosphate Receptors/genetics , Mice , Arthritis, Experimental/metabolism , Arthritis, Experimental/pathology , Antigens, CD/metabolism , Antigens, CD/genetics , Endothelial Cells/metabolism , Humans , Arthritis, Rheumatoid/metabolism , Arthritis, Rheumatoid/pathology , Arthritis, Rheumatoid/genetics , ADAM10 Protein/metabolism , ADAM10 Protein/genetics , Amyloid Precursor Protein Secretases/metabolism , Signal Transduction , Mice, Knockout , Membrane Proteins/metabolism , Membrane Proteins/genetics , Male , Sphingosine/analogs & derivatives , Sphingosine/metabolism , Lysophospholipids/metabolism , Capillary Permeability , Female
2.
Front Immunol ; 15: 1405622, 2024.
Article in English | MEDLINE | ID: mdl-38827741

ABSTRACT

Background: Severe acute pancreatitis (SAP) is an inflammatory disorder affecting the gastrointestinal system. Intestinal injury plays an important role in the treatment of severe acute pancreatitis. In this study, we mainly investigated the role of S1PR2 in regulating macrophage pyroptosis in the intestinal injury of severe acute pancreatitis. Methods: The SAP model was constructed using cerulein and lipopolysaccharide, and the expression of S1PR2 was inhibited by JTE-013 to detect the degree of pancreatitis and intestinal tissue damage in mice. Meanwhile, the level of pyroptosis-related protein was detected by western blot, the level of related mRNA was detected by PCR, and the level of serum inflammatory factors was detected by ELISA. In vitro experiments, LPS+ATP was used to construct the pyroptosis model of THP-1. After knockdown and overexpression of S1PR2, the pyroptosis proteins level was detected by western blot, the related mRNA level was detected by PCR, and the level of cell supernatant inflammatory factors were detected by ELISA. A rescue experiment was used to verify the sufficient necessity of the RhoA/ROCK pathway in S1PR2-induced pyroptosis. Meanwhile, THP-1 and FHC were co-cultured to verify that cytokines released by THP-1 after damage could regulate FHC damage. Results: Our results demonstrated that JTE-013 effectively attenuated intestinal injury and inflammation in mice with SAP. Furthermore, we observed a significant reduction in the expression of pyroptosis-related proteins within the intestinal tissue of SAP mice upon treatment with JTE-013. We confirmed the involvement of S1PR2 in THP-1 cell pyroptosis in vitro. Specifically, activation of S1PR2 triggered pyroptosis in THP-1 cells through the RhoA/ROCK signaling pathway. Moreover, it was observed that inflammatory factors released during THP-1 cell pyroptosis exerted an impact on cohesin expression in FHC cells. Conclusion: The involvement of S1PR2 in SAP-induced intestinal mucosal injury may be attributed to its regulation of macrophage pyroptosis.


Subject(s)
Disease Models, Animal , Macrophages , Pancreatitis , Pyroptosis , Sphingosine-1-Phosphate Receptors , Animals , Mice , Humans , Macrophages/metabolism , Macrophages/immunology , Pancreatitis/metabolism , Pancreatitis/immunology , Pancreatitis/pathology , Pancreatitis/chemically induced , Sphingosine-1-Phosphate Receptors/metabolism , Sphingosine-1-Phosphate Receptors/genetics , Male , Signal Transduction , Mice, Inbred C57BL , rhoA GTP-Binding Protein/metabolism , THP-1 Cells , rho-Associated Kinases/metabolism , rho-Associated Kinases/genetics , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Intestines/pathology , Intestines/immunology , Cytokines/metabolism , Lipopolysaccharides , Pyrazoles , Pyridines
3.
Mol Cell Biol ; 44(5): 178-193, 2024.
Article in English | MEDLINE | ID: mdl-38767243

ABSTRACT

Transcription factor 12 (TCF12) is a known oncogene in many cancers. However, whether TCF12 can regulate malignant phenotypes and angiogenesis in osteosarcoma is not elucidated. In this study, we demonstrated increased expression of TCF12 in osteosarcoma tissues and cell lines. High TCF12 expression was associated with metastasis and poor survival rate of osteosarcoma patients. Knockdown of TCF12 reduced the proliferation, migration, and invasion of osteosarcoma cells. TCF12 was found to bind to the promoter region of sphingosine kinase 1 (SPHK1) to induce transcriptional activation of SPHK1 expression and enhance the secretion of sphingosine-1-phosphate (S1P), which eventually resulted in the malignant phenotypes of osteosarcoma cells. In addition, S1P secreted by osteosarcoma cells promoted the angiogenesis of HUVECs by targeting S1PR4 on the cell membrane to activate the STAT3 signaling pathway. These findings suggest that TCF12 may induce transcriptional activation of SPHK1 to promote the synthesis and secretion of S1P. This process likely enhances the malignant phenotypes of osteosarcoma cells and induces angiogenesis via the S1PR4/STAT3 signaling pathway.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors , Lysophospholipids , Neovascularization, Pathologic , Osteosarcoma , Phosphotransferases (Alcohol Group Acceptor) , STAT3 Transcription Factor , Signal Transduction , Sphingosine , Humans , Osteosarcoma/genetics , Osteosarcoma/metabolism , Osteosarcoma/pathology , STAT3 Transcription Factor/metabolism , STAT3 Transcription Factor/genetics , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Phosphotransferases (Alcohol Group Acceptor)/genetics , Lysophospholipids/metabolism , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/genetics , Cell Line, Tumor , Sphingosine/analogs & derivatives , Sphingosine/metabolism , Basic Helix-Loop-Helix Transcription Factors/metabolism , Basic Helix-Loop-Helix Transcription Factors/genetics , Gene Expression Regulation, Neoplastic , Cell Proliferation/genetics , Human Umbilical Vein Endothelial Cells/metabolism , Bone Neoplasms/metabolism , Bone Neoplasms/genetics , Bone Neoplasms/pathology , Transcriptional Activation/genetics , Sphingosine-1-Phosphate Receptors/metabolism , Sphingosine-1-Phosphate Receptors/genetics , Receptors, Lysosphingolipid/metabolism , Receptors, Lysosphingolipid/genetics , Cell Movement/genetics , Male , Animals , Female , Angiogenesis
4.
Exp Cell Res ; 439(1): 114071, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38729336

ABSTRACT

Atherosclerosis preferentially occurs in areas with low shear stress (LSS) and oscillatory flow. LSS has been demonstrated to correlate with the development of atherosclerosis. The sphingosine 1-phosphate receptor 1 (S1PR1), involving intravascular blood flow sensing, regulates vascular development and vascular barrier function. However, whether LSS affects atherosclerosis via regulating S1PR1 remains incompletely clear. In this study, immunostaining results of F-actin, ß-catenin, and VE-cadherin indicated that LSS impaired endothelial barrier function in human umbilical vein endothelial cells (HUVECs). Western blot analysis showed that LSS resulted in blockage of autophagic flux in HUVECs. In addition, autophagy agonist Rapamycin (Rapa) antagonized LSS-induced endothelial barrier dysfunction, whereas autophagic flux inhibitor Bafilomycin A1 (BafA1) exacerbated it, indicating that LSS promoted endothelial barrier dysfunction by triggering autophagic flux blockage. Notably, gene expression analysis revealed that LSS downregulated S1PR1 expression, which was antagonized by Rapa. Selective S1PR1 antagonist W146 impaired endothelial barrier function of HUVECs under high shear stress (HSS) conditions. Moreover, our data showed that expression of GAPARAPL2, a member of autophagy-related gene 8 (Atg8) proteins, was decreased in HUVECs under LSS conditions. Autophagic flux blockage induced by GAPARAPL2 knockdown inhibited S1PR1, aggravated endothelial barrier dysfunction of HUVECs in vitro, and promoted aortic atherosclerosis in ApoE-/- mice in vivo. Our study demonstrates that autophagic flux blockage induced by LSS downregulates S1PR1 expression and impairs endothelial barrier function. GABARAPL2 inhibition is involved in LSS-induced autophagic flux blockage, which impairs endothelial barrier function via downregulation of S1PR1.


Subject(s)
Atherosclerosis , Autophagy , Human Umbilical Vein Endothelial Cells , Sphingosine-1-Phosphate Receptors , Stress, Mechanical , Animals , Autophagy/drug effects , Atherosclerosis/metabolism , Atherosclerosis/pathology , Atherosclerosis/genetics , Humans , Human Umbilical Vein Endothelial Cells/metabolism , Sphingosine-1-Phosphate Receptors/metabolism , Sphingosine-1-Phosphate Receptors/genetics , Mice , Mice, Inbred C57BL , Male , Endothelium, Vascular/metabolism , Endothelium, Vascular/pathology , Endothelium, Vascular/drug effects
5.
Cardiovasc Res ; 120(5): 476-489, 2024 Apr 30.
Article in English | MEDLINE | ID: mdl-38109696

ABSTRACT

AIMS: The entry of lipoproteins from blood into the arterial wall is a rate-limiting step in atherosclerosis. It is controversial whether this happens by filtration or regulated transendothelial transport.Because sphingosine-1-phosphate (S1P) preserves the endothelial barrier, we investigated in vivo and in vitro, whether S1P and its cognate S1P-receptor 3 (S1P3) regulate the transendothelial transport of lipoproteins. METHODS AND RESULTS: Compared to apoE-haploinsufficient mice (CTRL), apoE-haploinsufficient mice with additional endothelium-specific knock-in of S1P3 (S1P3-iECKI) showed decreased transport of LDL and Evan's Blue but increased transport of HDL from blood into the peritoneal cave. After 30 weeks of high-fat diet feeding, S1P3-iECKI mice had lower levels of non-HDL-cholesterol and less atherosclerosis than CTRL mice. In vitro stimulation with an S1P3 agonist increased the transport of 125I-HDL but decreased the transport of 125I-LDL through human aortic endothelial cells (HAECs). Conversely, inhibition or knock-down of S1P3 decreased the transport of 125I-HDL but increased the transport of 125I-LDL. Silencing of SCARB1 encoding scavenger receptor B1 (SR-BI) abrogated the stimulation of 125I-HDL transport by the S1P3 agonist. The transendothelial transport of 125I-LDL was decreased by silencing of SCARB1 or ACVLR1 encoding activin-like kinase 1 but not by interference with LDLR. None of the three knock-downs prevented the stimulatory effect of S1P3 inhibition on transendothelial 125I-LDL transport. CONCLUSION: S1P3 regulates the transendothelial transport of HDL and LDL oppositely by SR-BI-dependent and SR-BI-independent mechanisms, respectively. This divergence supports a contention that lipoproteins pass the endothelial barrier by specifically regulated mechanisms rather than passive filtration.


Subject(s)
Atherosclerosis , Endothelial Cells , Lipoproteins, HDL , Lipoproteins, LDL , Protein Transport , Sphingosine-1-Phosphate Receptors , Animals , Humans , Mice , Atherosclerosis/metabolism , Atherosclerosis/genetics , Atherosclerosis/pathology , Atherosclerosis/prevention & control , Biological Transport , Cells, Cultured , Disease Models, Animal , Endothelial Cells/metabolism , Lipoproteins, HDL/metabolism , Lipoproteins, LDL/metabolism , Lysophospholipids , Mice, Inbred C57BL , Mice, Knockout, ApoE , Scavenger Receptors, Class B/metabolism , Scavenger Receptors, Class B/genetics , Sphingosine-1-Phosphate Receptors/metabolism , Sphingosine-1-Phosphate Receptors/genetics , Protein Transport/genetics
6.
Arthritis Res Ther ; 25(1): 206, 2023 10 19.
Article in English | MEDLINE | ID: mdl-37858140

ABSTRACT

BACKGROUND: Circular RNAs are involved in autoimmune disease pathogenesis. Our previous study indicated that circPTPN22 is involved in autoimmune diseases, such as systemic lupus erythematosus (SLE) and rheumatoid arthritis, but the underlying mechanisms remain unclear. METHODS: First, the expression of circPTPN22 was detected by real-time PCR and western blotting. After overexpression or knockdown of circPTPN22, the proliferation of Jurkat cells was detected by the CCK-8 assay, and the apoptosis of Jurkat cells was detected by flow cytometry. In addition, the relationship between circPTPN22-miR-4689-S1PR1 was confirmed by bioinformatic analyses, fluorescence in situ hybridization assays, RNA-binding protein immunoprecipitation, and dual luciferase reporter assays. RESULTS: We found that circPTPN22 expression was downregulated in the PBMCs of SLE patients compared to those of healthy controls. Overexpression of circPTPN22 increased proliferation and inhibited apoptosis of Jurkat T cells, whereas knockdown of circPTPN22 exerted the opposite effects. CircPTPN22 acts as a miR-4689 sponge, and S1PR1 is a direct target of miR-4689. Importantly, the circPTPN22/miR-4689/S1PR1 axis inhibited the secretion of TNF-α and IL-6 in Jurkat T cells. CONCLUSIONS: CircPTPN22 acts as a miR-4689 sponge to regulate T-cell activation by targeting S1PR1, providing a novel mechanism for the pathogenesis of SLE.


Subject(s)
Lupus Erythematosus, Systemic , MicroRNAs , Protein Tyrosine Phosphatase, Non-Receptor Type 22 , RNA, Circular , Sphingosine-1-Phosphate Receptors , T-Lymphocytes , Humans , In Situ Hybridization, Fluorescence , Jurkat Cells , Lupus Erythematosus, Systemic/genetics , Lupus Erythematosus, Systemic/immunology , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , MicroRNAs/genetics , MicroRNAs/immunology , Protein Tyrosine Phosphatase, Non-Receptor Type 22/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 22/immunology , RNA, Circular/genetics , RNA, Circular/immunology , Sphingosine-1-Phosphate Receptors/genetics , Sphingosine-1-Phosphate Receptors/immunology , T-Lymphocytes/immunology
7.
Oncogene ; 42(47): 3491-3502, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37828220

ABSTRACT

Cell senescence deters the activation of various oncogenes. Induction of senescence is, therefore, a potentially effective strategy to interfere with vital processes in tumor cells. Sphingosine-1-phosphate receptor 1 (S1PR1) has been implicated in various cancer types, including ovarian cancer. The mechanism by which S1PR1 regulates ovarian cancer cell senescence is currently elusive. In this study, we demonstrate that S1PR1 was highly expressed in human ovarian cancer tissues and cell lines. S1PR1 deletion inhibited the proliferation and migration of ovarian cancer cells. S1PR1 deletion promoted ovarian cancer cell senescence and sensitized ovarian cancer cells to cisplatin chemotherapy. Exposure of ovarian cancer cells to sphingosine-1-phosphate (S1P) increased the expression of 3-phosphatidylinositol-dependent protein kinase 1 (PDK1), decreased the expression of large tumor suppressor 1/2 (LATS1/2), and induced phosphorylation of Yes-associated protein (p-YAP). Opposite results were obtained in S1PR1 knockout cells following pharmacological inhibition. After silencing LATS1/2 in S1PR1-deficient ovarian cancer cells, senescence was suppressed and S1PR1 expression was increased concomitantly with YAP expression. Transcriptional regulation of S1PR1 by YAP was confirmed by chromatin immunoprecipitation. Accordingly, the S1PR1-PDK1-LATS1/2-YAP pathway regulates ovarian cancer cell senescence and does so through a YAP-mediated feedback loop. S1PR1 constitutes a druggable target for the induction of senescence in ovarian cancer cells. Pharmacological intervention in the S1PR1-PDK1-LATS1/2-YAP signaling axis may augment the efficacy of standard chemotherapy.


Subject(s)
Ovarian Neoplasms , Protein Kinases , Female , Humans , Sphingosine-1-Phosphate Receptors/genetics , Ovarian Neoplasms/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Cellular Senescence/genetics , Cell Proliferation/genetics
8.
Sci Signal ; 16(798): eade6737, 2023 08 15.
Article in English | MEDLINE | ID: mdl-37582160

ABSTRACT

The G protein-coupled receptor (GPCR) US28 encoded by the human cytomegalovirus (HCMV) is associated with accelerated progression of glioblastomas, aggressive brain tumors with a generally poor prognosis. Here, we showed that US28 increased the malignancy of U251 glioblastoma cells by enhancing signaling mediated by sphingosine-1-phosphate (S1P), a bioactive lipid that stimulates oncogenic pathways in glioblastoma. US28 expression increased the abundance of the key components of the S1P signaling axis, including an enzyme that generates S1P [sphingosine kinase 1 (SK1)], an S1P receptor [S1P receptor 1 (S1P1)], and S1P itself. Enhanced S1P signaling promoted glioblastoma cell proliferation and survival by activating the kinases AKT and CHK1 and the transcriptional regulators cMYC and STAT3 and by increasing the abundance of cancerous inhibitor of PP2A (CIP2A), driving several feed-forward signaling loops. Inhibition of S1P signaling abrogated the proliferative and anti-apoptotic effects of US28. US28 also activated the S1P signaling axis in HCMV-infected cells. This study uncovers central roles for S1P and CIP2A in feed-forward signaling that contributes to the US28-mediated exacerbation of glioblastoma.


Subject(s)
Glioblastoma , Humans , Glioblastoma/genetics , Glioblastoma/metabolism , Glioblastoma/pathology , Sphingosine-1-Phosphate Receptors/genetics , Signal Transduction , Lysophospholipids/metabolism , Sphingosine/metabolism , Receptors, Lysosphingolipid/genetics , Receptors, Lysosphingolipid/metabolism
9.
Am J Hypertens ; 36(9): 509-516, 2023 08 05.
Article in English | MEDLINE | ID: mdl-37171128

ABSTRACT

BACKGROUND: Our previous studies showed that renal medullary sphingosine-1-phosphate receptor 1 (S1PR1) mediated sodium excretion, high salt intake increased S1PR1 level, deoxycorticosterone acetate (DOCA) blocked high salt-induced S1PR1 in the renal medulla, and that conditional knockout of S1PR1 in the collecting duct aggravated DOCA-salt hypertension. The present study tested the hypothesis that overexpression of S1PR1 transgene in the renal medulla attenuates the sodium retention and hypertension in DOCA-salt mouse model. METHODS: Male C57BL/6J mice received renal medullary transfection of control or S1PR1-expressing plasmids and then DOCA-salt treatment. Renal sodium excretion and arterial pressure were compared between control and S1PR1-overexpressed mice in response to high salt loading or pressure natriuresis. RESULTS: S1PR1-transfected mice showed significantly enhanced urinary sodium excretion in response to acute sodium loading (0.93 ± 0.27 in control vs. 4.72 ± 1.12 µmol/min/gKW in S1PR1-overexpressed mice, P < 0.05) and the pressure natriuresis (3.58 ± 1.77 vs. 9.52 ± 1.38, P < 0.05), less positive sodium balance in response to chronic high-salt intake (3.05 ± 0.39 vs. 1.65 ± 0.39 mmol/72 hr, P < 0.05), and consequently, the attenuation of DOCA-salt hypertension (134.2 ± 6.79 vs. 109.8 ± 3.54 mm Hg, P < 0.05). The αENaC protein amount in the renal medulla was not changed, however, the ßENaC was significantly decreased and the γENaC was significantly increased in S1PR1-overexpressed mice. The immunostaining showed apical membrane translocation of γENaC, while no change of αENaC and ßENaC in control mice, and that the apical membrane translocation of γENaC was blocked in S1PR1-treasffected mice. CONCLUSIONS: These results suggested that activation of S1PR1 in the renal medulla attenuates DOCA-induced sodium retention and salt-sensitive hypertension associated with inhibition of ENaC.


Subject(s)
Desoxycorticosterone Acetate , Hypertension , Male , Mice , Animals , Desoxycorticosterone Acetate/adverse effects , Sodium Chloride, Dietary/adverse effects , Sphingosine-1-Phosphate Receptors/genetics , Sphingosine-1-Phosphate Receptors/metabolism , Mice, Inbred C57BL , Hypertension/chemically induced , Hypertension/genetics , Hypertension/metabolism , Blood Pressure , Sodium/metabolism , Sodium Chloride/adverse effects , Transgenes , Acetates/adverse effects , Acetates/metabolism , Kidney
10.
Int J Biol Sci ; 19(5): 1597-1615, 2023.
Article in English | MEDLINE | ID: mdl-37056936

ABSTRACT

Patients with eosinophilic asthma react well to conventional treatment of asthma while individualized therapy for non-eosinophilic endotypes have yet to be developed. Dysregulated sphingosine metabolites are associated with the pathophysiology of different asthma endotypes with their receptors involved. However, whether the sphingosine-1-phosphate receptor 4 (S1PR4) contributes to disease progression of asthma remains underappreciated. In this study, we demonstrated that sphingosine metabolism was disturbed in asthma while it could not be used to distinguish between different endotypes of asthma. S1PR4, a vital receptor of bioactive sphingosine metabolites and mainly expressed in macrophages, exhibited lower expression both in patients and experimental mice with neutrophilic airway inflammation. Additionally, S1pr4 deficiency aggravated the OVA/LPS-induced pulmonary inflammation in mice along with a significant up-regulation in M1 macrophage activation. Mechanistic studies showed that S1PR4 was strongly connected to bioactive oxylipins concurrent with bounding to formyl peptide receptor 2 to influence the phosphorylation of JNK and contributed to the macrophage M1 program, which in turn secreted amounts of inflammatory cytokines associated to the inflammatory response of neutrophilic asthma. Furthermore, treating mice with S1PR4 agonist CYM50308 was characterized by less pulmonary inflammatory infiltration. Our research indicates S1PR4 a promising therapeutic target for non-eosinophilic phenotypes of asthma.


Subject(s)
Asthma , Sphingosine , Mice , Animals , Sphingosine-1-Phosphate Receptors/genetics , Sphingosine-1-Phosphate Receptors/therapeutic use , Sphingosine/metabolism , Sphingosine/pharmacology , Sphingosine/therapeutic use , Macrophage Activation , Asthma/metabolism , Inflammation , Disease Models, Animal
11.
Sci Rep ; 12(1): 18027, 2022 10 27.
Article in English | MEDLINE | ID: mdl-36302829

ABSTRACT

This study aimed to explore the regulatory role of lncRNA ANRIL/miR-181b-5p/S1PR1 in UC. UC mouse model was established by 5/6th nephrectomy. We detected body weight, serum levels of renal function and inflammatory factors (biochemical analyzer/ELISA), and cardiac parameters (echocardiography). HE and Masson staining showed the pathological changes and fibrosis in myocardial and nephridial tissues. The expression of ANRIL, miR-181b-5p, and S1PR1 were detected by qRT-PCR or Western blot/immunofluorescence. T cells activation was analyzed by Flow cytometry. ANRIL/S1PR1 were up-regulated and miR-181b-5p was down-regulated in UC mice. ANRIL silencing up-regulated miR-181b-5p and down-regulated S1PR1 (a target of miR-181b-5p). ANRIL silencing increased the body weight, recovered renal function [decreased blood urea nitrogen (BUN) and serum creatinine (Scr)] and cardiac function [decreased left ventricular end-diastolic diameter (LVEDD), LV end-systolic diameter (LVESD), LV systolic anterior wall thickness (LVAWS), LV end-diastolic anterior wall thickness (LVAWD), myocardial performance index (MPI), and isovolumic relaxation time (IVRT); increased LV ejection fraction (LVEF), LVEF/MPI, fractional shortening (FS), and E- and A-waves (E/A)], inhibited the inflammation [decreased interferon (IFN)-γ, interleukin (IL)-2, IL-10, and tumor necrosis factor (TNF)-α], and relieved pathological injuries and fibrosis. ANRIL silencing also recovered the viability and inhibited the inflammation of activated T cells in vitro, and inhibited T cell activation in UC mice in vivo. In addition, miR-181b-5p overexpression exhibited same effects with ANRIL silencing in UC. ANRIL silencing inhibited T cell activation through regulating miR-181b-5p/S1PR1, contributing to the remission of UC.


Subject(s)
Cardiomyopathies , MicroRNAs , RNA, Long Noncoding , Sphingosine-1-Phosphate Receptors , T-Lymphocytes , Animals , Mice , Body Weight , Cardiomyopathies/pathology , Cell Line , Fibrosis , Inflammation/pathology , MicroRNAs/genetics , MicroRNAs/metabolism , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , T-Lymphocytes/metabolism , Tumor Necrosis Factor-alpha , Sphingosine-1-Phosphate Receptors/genetics , Sphingosine-1-Phosphate Receptors/metabolism
12.
J Clin Invest ; 132(17)2022 09 01.
Article in English | MEDLINE | ID: mdl-36047496

ABSTRACT

Cancer-related cognitive impairment (CRCI) is a major neurotoxicity affecting more than 50% of cancer survivors. The underpinning mechanisms are mostly unknown, and there are no FDA-approved interventions. Sphingolipidomic analysis of mouse prefrontal cortex and hippocampus, key sites of cognitive function, revealed that cisplatin increased levels of the potent signaling molecule sphingosine-1-phosphate (S1P) and led to cognitive impairment. At the biochemical level, S1P induced mitochondrial dysfunction, activation of NOD-, LRR-, and pyrin domain-containing protein 3 inflammasomes, and increased IL-1ß formation. These events were attenuated by systemic administration of the functional S1P receptor 1 (S1PR1) antagonist FTY720, which also attenuated cognitive impairment without adversely affecting locomotor activity. Similar attenuation was observed with ozanimod, another FDA-approved functional S1PR1 antagonist. Mice with astrocyte-specific deletion of S1pr1 lost their ability to respond to FTY720, implicating involvement of astrocytic S1PR1. Remarkably, our pharmacological and genetic approaches, coupled with computational modeling studies, revealed that cisplatin increased S1P production by activating TLR4. Collectively, our results identify the molecular mechanisms engaged by the S1P/S1PR1 axis in CRCI and establish S1PR1 antagonism as an approach to target CRCI with therapeutics that have fast-track clinical application.


Subject(s)
Cognitive Dysfunction , Fingolimod Hydrochloride , Animals , Central Nervous System/metabolism , Cisplatin/adverse effects , Cognitive Dysfunction/chemically induced , Cognitive Dysfunction/drug therapy , Cognitive Dysfunction/genetics , Fingolimod Hydrochloride/pharmacology , Lysophospholipids/metabolism , Mice , Mice, Inbred NOD , Receptors, Lysosphingolipid/genetics , Receptors, Lysosphingolipid/metabolism , Sphingosine/metabolism , Sphingosine-1-Phosphate Receptors/genetics
13.
Bioengineered ; 13(5): 13703-13717, 2022 05.
Article in English | MEDLINE | ID: mdl-35707833

ABSTRACT

Sphingosine-1-phosphate receptor 2 (S1PR2) was highly expressed in intestinal epithelial cells (IECs) and facilitated the proliferation of IECs. However, the specific function of S1PR2 in intestinal diseases, such as ulcerative colitis (UC), remains unclear. Accordingly, the current study set out to investigate the function of S1PR2 in maintaining intestinal barrier and inducing UC. S1PR2-overexpressed and knockdown Caco-2 cells were established to explore the function of S1PR2 on the permeability of IECs barrier. The UC-like mouse model in which UC is induced by dextran sulfate sodium (DSS) was established and utilized to investigate the role for S1PR2. The results showed that S1PR2 functioned as a maintainer of IECs permeability and a pathogenic factor for UC. A series of in vitro and in vivo experiments were conducted, and it was found that S1PR2 played an important role in intestinal epithelial cell proliferation and maintenance of intestinal epithelial cell barrier, possibly by the regulation on the expression level of SphK2, HDAC1, HDAC2, and ERK1/2 signaling pathway. The expression of S1PR2 was upregulated in UC mice and the colonic pathological damage in UC mice could be alleviated by the inhibition of S1PR2. Collectively, these results suggest that S1PR2 functions as a maintainer of IECs permeability and a pathogenic factor for UC. The research suggests S1PR2 may be an effective target for developing therapeutic strategies against UC.Abbreviations: S1PR2, Sphingosine-1-phosphate receptor 2; UC, ulcerative colitis; IECs, intestinal epithelial cells; DSS, dextran sulfate sodium; IBD, inflammation bowel disease; CD, Crohn's disease; S1P, sphingosin-1-phosphate; SphK, sphingosine kinase; HIECs, human IECs; siRNA, small interfering RNA; CCK-8, cell counting kit-8; TEER, transepithelial electrical resistance; TEM, transmission electron microscope; RT-PCR, real-time reverse transcriptase polymerase-chain reaction; ELISA, enzyme-linked immunosorbent assay; HE, hematoxylin and eosin.


Subject(s)
Colitis, Ulcerative , Colitis , Sphingosine-1-Phosphate Receptors , Animals , Caco-2 Cells , Colitis/chemically induced , Colitis/metabolism , Colitis/pathology , Colitis, Ulcerative/metabolism , Colitis, Ulcerative/pathology , Dextran Sulfate , Disease Models, Animal , Humans , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Mice , Mice, Inbred C57BL , Sphingosine-1-Phosphate Receptors/genetics
14.
Nat Commun ; 13(1): 731, 2022 02 08.
Article in English | MEDLINE | ID: mdl-35136060

ABSTRACT

Lysophospholipids are bioactive lipids and can signal through G-protein-coupled receptors (GPCRs). The best studied lysophospholipids are lysophosphatidic acid (LPA) and sphingosine 1-phosphate (S1P). The mechanisms of lysophospholipid recognition by an active GPCR, and the activations of lysophospholipid GPCR-G-protein complexes remain unclear. Here we report single-particle cryo-EM structures of human S1P receptor 1 (S1P1) and heterotrimeric Gi complexes formed with bound S1P or the multiple sclerosis (MS) treatment drug Siponimod, as well as human LPA receptor 1 (LPA1) and Gi complexes in the presence of LPA. Our structural and functional data provide insights into how LPA and S1P adopt different conformations to interact with their cognate GPCRs, the selectivity of the homologous lipid GPCRs for S1P versus LPA, and the different activation mechanisms of these GPCRs by LPA and S1P. Our studies also reveal specific optimization strategies to improve the MS-treating S1P1-targeting drugs.


Subject(s)
GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Receptors, Lysophosphatidic Acid/metabolism , Sphingosine-1-Phosphate Receptors/metabolism , Animals , Azetidines/pharmacology , Azetidines/therapeutic use , Benzyl Compounds/pharmacology , Benzyl Compounds/therapeutic use , Cryoelectron Microscopy , GTP-Binding Protein alpha Subunits, Gi-Go/genetics , GTP-Binding Protein alpha Subunits, Gi-Go/isolation & purification , GTP-Binding Protein alpha Subunits, Gi-Go/ultrastructure , Humans , Lysophospholipids/metabolism , Molecular Conformation/drug effects , Molecular Docking Simulation , Multiple Sclerosis/drug therapy , Multiple Sclerosis/pathology , Receptors, Lysophosphatidic Acid/genetics , Receptors, Lysophosphatidic Acid/isolation & purification , Receptors, Lysophosphatidic Acid/ultrastructure , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Recombinant Proteins/ultrastructure , Sf9 Cells , Single Molecule Imaging , Sphingosine/analogs & derivatives , Sphingosine/metabolism , Sphingosine-1-Phosphate Receptors/genetics , Sphingosine-1-Phosphate Receptors/isolation & purification , Sphingosine-1-Phosphate Receptors/ultrastructure , Spodoptera
15.
J Headache Pain ; 23(1): 25, 2022 Feb 10.
Article in English | MEDLINE | ID: mdl-35144528

ABSTRACT

BACKGROUND: Central sensitization is an important pathophysiological mechanism of chronic migraine (CM), and microglia activation in trigeminocervical complex (TCC) contributes to the development of central sensitization. Emerging evidence implicates that blocking sphingosine-1-phosphate receptor 1 (S1PR1) can relieve the development of chronic pain and inhibit the activation of microglia. However, it is unclear whether S1PR1 is involved in the central sensitization of CM. Therefore, the purpose of this study is to explore the role of S1PR1 and its downstream signal transducers and activators of transcription 3 (STAT3) signaling pathway in the CM, mainly in inflammation. METHODS: Chronic intermittent intraperitoneal injection of nitroglycerin (NTG) established a mouse model of CM. First, we observed the changes and subcellular localization of S1PR1 in the trigeminocervical complex (TCC). Then, W146, a S1PR1 antagonist; SEW2871, a S1PR1 agonist; AG490, a STAT3 inhibitor were applied by intraperitoneal injection to investigate the related molecular mechanism. The changes in the number of microglia and the expression of calcitonin gene-related peptide (CGRP) and c-fos in the TCC site were explored by immunofluorescence. In addition, we studied the effect of S1PR1 inhibitors on STAT3 in lipopolysaccharide-treated BV-2 microglia. RESULTS: Our results showed that the expression of S1PR1 was increased after NTG injection and S1PR1 was colocalized with in neurons and glial cells in the TCC. The S1PR1 antagonist W146 alleviated NTG-induced hyperalgesia and suppressed the upregulation of CGRP, c-fos and pSTAT3 in the TCC. Importantly, blocking S1PR1 reduced activation of microglia. In addition, we found that inhibiting STAT3 signal also attenuated NTG-induced basal mechanical and thermal hyperalgesia. CONCLUSIONS: Our results indicate that inhibiting S1PR1 signal could alleviate central sensitization and inhibit microglia activity caused by chronic NTG administration via STAT3 signal pathway, which provide a new clue for the clinical treatment of CM.


Subject(s)
Migraine Disorders , Nitroglycerin , Sphingosine-1-Phosphate Receptors/genetics , Animals , Central Nervous System Sensitization , Disease Models, Animal , Hyperalgesia/chemically induced , Hyperalgesia/drug therapy , Mice , Migraine Disorders/chemically induced , Migraine Disorders/drug therapy
16.
Sci Rep ; 12(1): 454, 2022 01 10.
Article in English | MEDLINE | ID: mdl-35013382

ABSTRACT

Sphingosine 1-phosphate (S1P) is a signaling lipid that has broad roles, working either intracellularly through various protein targets, or extracellularly via a family of five G-protein coupled receptors. Agents that selectively and specifically target each of the S1P receptors have been sought as both biological tools and potential therapeutics. JTE-013, a small molecule antagonist of S1P receptors 2 and 4 (S1P2 and S1P4) has been widely used in defining the roles of these receptors in various biological processes. Indeed, our previous studies showed that JTE-013 had anti-acute myeloid leukaemia (AML) activity, supporting a role for S1P2 in the biology and therapeutic targeting of AML. Here we examined this further and describe lipidomic analysis of AML cells that revealed JTE-013 caused alterations in sphingolipid metabolism, increasing cellular ceramides, dihydroceramides, sphingosine and dihydrosphingosine. Further examination of the mechanisms behind these observations showed that JTE-013, at concentrations frequently used in the literature to target S1P2/4, inhibits several sphingolipid metabolic enzymes, including dihydroceramide desaturase 1 and both sphingosine kinases. Collectively, these findings demonstrate that JTE-013 can have broad off-target effects on sphingolipid metabolism and highlight that caution must be employed in interpreting the use of this reagent in defining the roles of S1P2/4.


Subject(s)
Pyrazoles/chemistry , Pyridines/chemistry , Sphingolipids/metabolism , Sphingosine-1-Phosphate Receptors/antagonists & inhibitors , Sphingosine-1-Phosphate Receptors/metabolism , HEK293 Cells , Humans , Kinetics , Oxidoreductases/chemistry , Oxidoreductases/genetics , Oxidoreductases/metabolism , Pyrazoles/pharmacology , Pyridines/pharmacology , Sphingosine-1-Phosphate Receptors/genetics
17.
Hypertension ; 79(2): 399-412, 2022 02.
Article in English | MEDLINE | ID: mdl-34865521

ABSTRACT

Incomplete spiral artery remodeling, caused by impaired extravillous trophoblast invasion, is a fundamental pathogenic process associated with malplacentation and the development of preeclampsia. Nevertheless, the mechanisms controlling this regulation of trophoblast invasion are largely unknown. We report that sphingosine-1-phosphate synthesis and expression is abundant in healthy trophoblast, whereas in pregnancies complicated by preeclampsia the placentae are associated with reduced sphingosine-1-phosphate and lower SPHK1 (sphingosine kinase 1) expression and activity. In vivo inhibition of sphingosine kinase 1 activity during placentation in pregnant mice led to decreased placental sphingosine-1-phosphate production and defective placentation, resulting in a preeclampsia phenotype. Moreover, sphingosine-1-phosphate increased HTR8/SVneo (immortalized human trophoblst cells) cell invasion in a Hippo-signaling-dependent transcriptional coactivator YAP (Yes-associated protein) dependent manner, which is activated by S1PR2 (sphingosine-1-phosphate receptor-2) and downstream RhoA (Ras homolog gene family, member A)/ROCK (Rho-associated protein kinase) induced actin polymerization. Mutation-based YAP-5SA (S61A, S109A, S127A, S164A, S381A) demonstrated that sphingosine-1-phosphate activation of YAP could be either dependent or independent of Hippo signaling. Together, these findings suggest a novel pathogenic pathway of preeclampsia via disrupted sphingosine-1-phosphate metabolism and signaling-induced, interrupted actin dynamics and YAP deactivation; this may lead to potential novel intervention targets for the prevention and management of preeclampsia.


Subject(s)
Actins/metabolism , Lysophospholipids/metabolism , Pre-Eclampsia/metabolism , Proto-Oncogene Proteins c-yes/metabolism , Sphingosine-1-Phosphate Receptors/metabolism , Sphingosine/analogs & derivatives , Trophoblasts/metabolism , Animals , Female , Humans , Lysophospholipids/genetics , Mice , Placenta/metabolism , Placentation/physiology , Pregnancy , Proto-Oncogene Proteins c-yes/genetics , Signal Transduction/physiology , Sphingosine/genetics , Sphingosine/metabolism , Sphingosine-1-Phosphate Receptors/genetics
18.
J Recept Signal Transduct Res ; 42(3): 225-229, 2022 Jun.
Article in English | MEDLINE | ID: mdl-33685344

ABSTRACT

Several intracellular pathological processes have been reported to be regulated by the FAM19A5/S1PR1 signaling pathway. However, the role of FAM19A5/S1PR1 signaling pathway in the viability and proliferation of mantle cell lymphoma is not been completely understood. The task of this study is to explore the influence of FAM19A5/S1PR1 signaling pathway in affecting the survival and growth of mantle cell lymphoma. shRNAs against FAM19A5 or S1PR1 were transfected into mantle cell lymphom. Cell viability and proliferation were measured through MTT assay and CCK8 assay, respectively. Our results demonstrated that loss of FAM19A5 significantly reduced the viability of mantle cell lymphom, an effect that was followed by a drop in cell proliferation capacity. Besides, inhibition of S1PR1 also impairs cell survival and interrupt mantle cell lymphom proliferation in vitro. Taken together, our results illustrate that FAM19A5/S1PR1 signaling pathway is associated with the regulation of mantle cell lymphom viability and proliferation. This finding will provide a potential target for the treatment of malignant lymphoma in the clinical practice.


Subject(s)
Cytokines , Lymphoma, Mantle-Cell , Sphingosine-1-Phosphate Receptors , Cell Line, Tumor , Cell Proliferation/genetics , Cell Survival/genetics , Cytokines/genetics , Humans , Lymphoma, Mantle-Cell/genetics , Signal Transduction/genetics , Sphingosine-1-Phosphate Receptors/genetics
19.
Anticancer Agents Med Chem ; 22(2): 254-260, 2022.
Article in English | MEDLINE | ID: mdl-33797381

ABSTRACT

BACKGROUND: SphK1 is a conserved lipid kinase, which can catalyze the formation of tumorpromoting factor sphingosine phosphate-1 (S1P). OBJECTIVE: This study aimed to investigate the effect of SphK1 on the proliferation/migration of colon cancer cells and associated mechanisms. METHODS: Transcription of the SphK1 gene in colon cancer cells was detected. Gene transcription of SphK1 was inhibited by transfecting with the si-SphK1 gene in colon cancer cells. Effects of SphK1 inhibition (si-SphK1) on cell migration/proliferation were detected using the transwell system and MTS. Gene transcription of SIP, S1PR1, S1PR2, S1PR3, and activation of JAK/STAT3 pathway were examined using RT-PCR and western blot assay. S1PR1 over-expressing plasmid was constructed and transfected into cells. Effects of S1PR1 overexpression on migration/proliferation of si-SphK1 transfected colon cancer cells and activation of JAK/STAT3 pathway were determined using RT-PCR and western blotting. RESULTS: Gene transcription of SphK1 in SW480 and HT-29 colon cancer cells was significantly inhibited by transfection of the si-SphK1 gene. Transwell migration and MTS findings showed that si-SphK1 transfection (si- SphK1 group) could reduce migration quantity and cell viability of colon cancer cells compared to the negative control (NC) (p<0.0001). SphK1 inhibition (si-SphK1 group) significantly down-regulated S1PR1 and S1PR3 gene transcription in SW480 and HT-29 cells (p<0.0001) and decreased activation level of JAKSTAT3 signaling pathway compared to NC group (p<0.05). Over-expression of S1PR1 reversed inhibitory effects of si-SphK1 on migration/proliferation of SW480 and activation of JAK/Stat3. CONCLUSION: SphK1 promoted proliferation and migration of colon cancer cells through promoting JAK/STAT activation and up-regulating S1PR1 expression.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Colonic Neoplasms/metabolism , Janus Kinase 1/metabolism , STAT3 Transcription Factor/metabolism , Sphingosine-1-Phosphate Receptors/genetics , Up-Regulation , Adaptor Proteins, Signal Transducing/genetics , Cell Movement , Cell Proliferation , Colonic Neoplasms/pathology , Humans , Sphingosine-1-Phosphate Receptors/metabolism , Tumor Cells, Cultured
20.
J Exp Med ; 219(1)2022 01 03.
Article in English | MEDLINE | ID: mdl-34677611

ABSTRACT

Tissue-resident memory T (TRM) cells provide long-lasting immune protection. One of the key events controlling TRM cell development is the local retention of TRM cell precursors coupled to downregulation of molecules necessary for tissue exit. Sphingosine-1-phosphate receptor 5 (S1PR5) is a migratory receptor with an uncharted function in T cells. Here, we show that S1PR5 plays a critical role in T cell infiltration and emigration from peripheral organs, as well as being specifically downregulated in TRM cells. Consequentially, TRM cell development was selectively impaired upon ectopic expression of S1pr5, whereas loss of S1pr5 enhanced skin TRM cell formation by promoting peripheral T cell sequestration. Importantly, we found that T-bet and ZEB2 were required for S1pr5 induction and that local TGF-ß signaling was necessary to promote coordinated Tbx21, Zeb2, and S1pr5 downregulation. Moreover, S1PR5-mediated control of tissue residency was conserved across innate and adaptive immune compartments. Together, these results identify the T-bet-ZEB2-S1PR5 axis as a previously unappreciated mechanism modulating the generation of tissue-resident lymphocytes.


Subject(s)
Cell Differentiation/genetics , Lymphoid Tissue/metabolism , Memory T Cells/metabolism , Sphingosine-1-Phosphate Receptors/genetics , T-Lymphocytes/metabolism , Animals , CD8-Positive T-Lymphocytes/metabolism , Cell Movement/genetics , Cells, Cultured , Gene Expression Profiling/methods , Humans , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , RNA-Seq/methods , Sphingosine-1-Phosphate Receptors/metabolism , T-Box Domain Proteins/genetics , T-Box Domain Proteins/metabolism , Zinc Finger E-box Binding Homeobox 2/genetics , Zinc Finger E-box Binding Homeobox 2/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...