1.Research advances of the roles of sphingosine-1-phosphate in acute lung injury.
Meng Yan WANG ; Pei CUI ; Hai Ming XIN
Chinese Journal of Burns 2022;38(5):496-500
Sphingosine-1-phosphate (S1P) is the main metabolite produced in the process of phospholipid metabolism, which can promote proliferation, migration, and apoptosis of cells, and maintain the barrier function of vascular endothelium. The latest researches showed that S1P can alleviate acute lung injury (ALI) and the inflammation caused by ALI, while the dosage of S1P is still needed to be considered. Mesenchymal stem cells (MSCs) have been a emerging therapy with potential therapeutic effects on ALI because of their characteristics of self-replication and multi-directional differentiation, and their advantages in hematopoiesis, immune regulation, and tissue repair. S1P can promote differentiation of MSCs and participate in immune regulation, while MSCs can regulate the homeostasis of S1P in the body. The synergistic effect of S1P and MSC provides a new treatment method for ALI. This article reviews the production and biological function of S1P, receptor and signal pathway of S1P, the therapeutic effects of S1P on ALI, and the research advances of S1P combined with MSCs in the treatment of ALI, aiming to provide theoretical references for the development of S1P targeted drugs in the treatment of ALI and the search for new combined treatment schemes for ALI.
Acute Lung Injury
;
Animals
;
Lung/metabolism*
;
Lysophospholipids/pharmacology*
;
Mice
;
Mice, Inbred C57BL
;
Sphingosine/pharmacology*
2.Ceramide participates in cell programmed death induced by Type II anti-CD20 mAb.
Yan HUANG ; Sun WU ; Yuan ZHANG ; Youmei ZI ; Man YANG ; Yan GUO ; Lingxiu ZHANG ; Lihua WANG
Journal of Central South University(Medical Sciences) 2015;40(12):1292-1297
OBJECTIVE:
To explore the exact mechanisms of programmed cell death (PCD) induced by Type II anti-CD20 mAb in CD20+ non-Hodgkin lymphoma (NHL) cells, and to provide theoretical basis for anti-tumor ability of new CD20 mAb.
METHODS:
After incubation with Rituximab (a Type I anti-CD20 mAb) and Tositumomab (a Type II anti-CD20 mAb), Raji cells were stained by annexin V & propidium iodide (PI). The ratio of programmed death cells were measured by two channel flow cytometry (FCM). Before the treatment of anti-CD20 mAbs, Raji cells was incubated with a caspase inhibitor carbobenzoxy-valyl-alanyl-aspartyl-[O-methyl]- fluoromethylketone (Z-VAD-FMK) and a dihydroceramide synthase inhibitor fumonisin B1 (FB1) for 30 minutes to assess their inhibitory effect on PCD. High performance liquid chromatography (HPLC) was utilized to compare the ratio of programmed death cells between the pretreatment group (treated by Rituximab and Tositumomab) and the non-pretreatment group. The anti-CD20 mAbs-treated Raji cells were collected, and the ceramide levels in the Raji cells in the different pretreatment groups were also examined by HPLC, and the inhibitory effect of FB1 on the changes of ceramide levels in the Raji cells was measured. The Raji cells were incubated with different concentration C2-ceramide, C2-Ceramide-induced PCD was also evaluated by annexin V & PI staining after 16 hours.
RESULTS:
Tositumomab (10 µg/mL) but not Rituximab (10 µg/mL) can induce significant PCD (28.6±4.2)% in Raji cells, with significant difference (t=26.48, P<0.01), which cannot be blocked by Z-VAD-FMK with a concentration range from 10 to 30 µmol/L (F=3.01, P>0.05). The cellular ceramide levels in Raji cells were significantly elevated after the treatment of Tositumomab (t=28.48, P<0.01). C2-ceramide can significantly induce PCD in Raji cells in a dose-dependent manner with a concentration range from 5 to 40 µmol/L (F=2.71, P>0.05). The dihydroceramide synthase inhibitor FB1 can significantly inhibit the elevated cellular ceramide levels (F=20.18, P<0.01) and cell programmed death induced by Tositumomab (F=17.02, P<0.01).
CONCLUSION
Type II but not Type I anti-CD20 mAbs can induce caspase independent PCD in CD20+ NHL cells through the elevation of cellular ceramide levels. The PCD is not associated with classic caspase pathway.
Amino Acid Chloromethyl Ketones
;
Apoptosis
;
drug effects
;
Cell Line, Tumor
;
drug effects
;
Humans
;
Lymphoma, Non-Hodgkin
;
Rituximab
;
pharmacology
;
Sphingosine
;
analogs & derivatives
;
pharmacology
3.Effect of a novel selective S1P1 agonist, Syl948, on mouse skin transplantation.
Jing JIN ; Hai-Jing ZHANG ; Xiao-Jian WANG ; Wan-Qi ZHOU ; Da-Li YIN ; Xiao-Guang CHEN
Acta Pharmaceutica Sinica 2014;49(5):627-631
Syl948 is a synthesized selective S1P1 agonist with novel structure. HTRF-IP1 test indicated that Syl948-P, the active form of Syl948 in vitro, has strong activity against S1P1 (EC50: 83 +/- 16 nmol x L(-1)), but its effect on S1P3 was very weak (EC50: 1 026 +/- 90 nmol x L(-1)). In SD rats, oral administration of Syl948 10 mg x kg(-1) significantly decreased the peripheral blood lymphocytes (PBL), with the maximal PBL inhibition rate of 63%, which was as similar as equal dose of fingolimod (FTY720). Oral administration of Syl948 10 mg x kg(-1) had no effect on heart rate of SD rats, which was better than FTY720. Daily oral administration with Syl948 (2 or 4 mg x kg(-1)) significantly prolonged the survival time of the allografts of skin slice on mice. In summary, the above results demonstrated that Syl948 has great selectivity in vitro and good activity in vivo, which indicated its potential use as an anti-rejection drug in skin transplantation.
Animals
;
Fingolimod Hydrochloride
;
Graft Survival
;
drug effects
;
Immunosuppressive Agents
;
pharmacology
;
Lymphocytes
;
drug effects
;
Mice
;
Propylene Glycols
;
pharmacology
;
Rats
;
Receptors, Lysosphingolipid
;
agonists
;
Skin Transplantation
;
Sphingosine
;
analogs & derivatives
;
pharmacology
;
Transplantation, Homologous
4.Sphingosine kinase 1 enhances the proliferation and invasion of human colon cancer LoVo cells through up-regulating FAK pathway and the expression of ICAM-1 and VCAM-1.
Shi-quan LIU ; Ying-jie SU ; Jie-an HUANG ; Meng-bin QIN ; Guo-du TANG
Chinese Journal of Oncology 2013;35(5):331-336
OBJECTIVETo investigate the effects of sphingosine kinase 1 (SphK1) on the proliferation, migration and invasion of human colon cancer LoVo cells, and to explore the related mechanisms.
METHODSHuman colon cancer LoVo cells were divided into three groups: phorbol 12-myristate 13-acetate (PMA) was used to induce the activation of SphK1 in the PMA group, N,N-dimethylsphingosine (DMS) used to suppress the activity of SphK1 in DMS group, and the cells treated with equal amount of 0.9 % NaCl instead of drugs served as the control group. The activity of SphK1 was assayed by autoradiography, the cell proliferation was assessed by MTT assay, cell migration and invasion were examined by Boyden chamber assay, concentrations of sICAM-1 and sVCAM-1 were assayed by ELISA, and RT-PCR and Western blot were used to evaluate the mRNA and protein expression in the cells.
RESULTSThe activity of SphK1 was efficiently induced by PMA and significantly suppressed by DMS. PMA induced cell proliferation in a time- and dose-dependent manner. On the contrast, DMS suppressed cell proliferation in a time- and dose-dependent manner. After treating with PMA, the number of migrating and invasing cells were increased to 143.36 ± 8.73 and 118.46 ± 6.25, significantly higher than those of the control group (75.48 ± 6.12 and 64.19 ± 5.36). After treating with DMS, the number of migrating and invasing cells were decreased to 38.57 ± 3.24 and 32.48 ± 4.27, significantly lower than those of the control group (P < 0.01). The relative expression levels of FAK, ICAM-1 and VCAM-1 mRNA in the PMA group were 0.82 ± 0.06, 0.74 ± 0.05 and 0.89 ± 0.09, and those in the DMS group were 0.23 ± 0.02, 0.26 ± 0.03 and 0.37 ± 0.04, with significant differences between the PMA, DMS and control groups (P < 0.01). Compared with the control group, the relative expression levels of FAK and p-FAK proteins in the PMA group (0.52 ± 0.06 and 0.51 ± 0.06) were significantly elevated, and those of the DMS group (0.20 ± 0.03 and 0.09 ± 0.02) were significantly decreased. In addition, the concentrations of sICAM-1 and sVCAM-1 were significantly elevated with the activation of SphK1. On the contrary, those of the DMS group were significantly reduced with the suppression of SphK1 (Both P < 0.01).
CONCLUSIONSSphK1 may enhance the proliferation, migration and invasion of colon cancer LoVo cells through activating FAK pathway and up-regulating the expression of ICAM-1 and VCAM-1.
Cell Line, Tumor ; Cell Movement ; drug effects ; Cell Proliferation ; drug effects ; Colonic Neoplasms ; enzymology ; metabolism ; pathology ; Dose-Response Relationship, Drug ; Enzyme Inhibitors ; pharmacology ; Focal Adhesion Kinase 1 ; genetics ; metabolism ; Humans ; Intercellular Adhesion Molecule-1 ; genetics ; metabolism ; Neoplasm Invasiveness ; Phosphorylation ; drug effects ; Phosphotransferases (Alcohol Group Acceptor) ; metabolism ; RNA, Messenger ; metabolism ; Signal Transduction ; Sphingosine ; analogs & derivatives ; pharmacology ; Tetradecanoylphorbol Acetate ; pharmacology ; Vascular Cell Adhesion Molecule-1 ; genetics ; metabolism
5.Sphingosine kinase 1 and tumor.
Cai-Xia ZHANG ; Hong-Wei HE ; Rong-Guang SHAO
Acta Pharmaceutica Sinica 2013;48(7):971-978
Sphingolipids as an important regulator play a critical role in the cell biological functions. Among them, ceramide (Cer) and sphingosine (Sph) induce apoptosis and inhibit cell proliferation; on the contrary sphingosine 1-phosphate (S1P) promotes cell survival and proliferation. The balance between ceramide/sphingosine and S1P forms a so-called "sphingolipid-rheostat", which decides the cell fate. Sphingosine kinases, which catalyze the phosphorylation of sphingosine to S1P, are critical regulators of this balance. Here, we review the role of sphingosine kinase 1 (SphK1) in regulating fundamental biological processes and tumorigenesis and the potential of SphK1 as a new target for cancer therapeutics.
Amino Alcohols
;
pharmacology
;
Animals
;
Apoptosis
;
drug effects
;
Cell Movement
;
drug effects
;
Cell Proliferation
;
drug effects
;
Ceramides
;
metabolism
;
Enzyme Activation
;
Enzyme Inhibitors
;
pharmacology
;
Humans
;
Lysophospholipids
;
metabolism
;
Neoplasms
;
metabolism
;
pathology
;
Neovascularization, Pathologic
;
Phosphorylation
;
Phosphotransferases (Alcohol Group Acceptor)
;
antagonists & inhibitors
;
metabolism
;
Sphingosine
;
analogs & derivatives
;
metabolism
;
Thiazoles
;
pharmacology
6.Mechanisms of ROS in U266 cell death induced by FTY720.
Ying-Chun LI ; Zhuo-Gang LIU ; Kun YAO ; Hui-Han WANG ; Rong HU ; Wei YANG ; Ai-Jun LIAO
Journal of Experimental Hematology 2013;21(3):643-646
This study was purpose to investigate the role of reactive oxygen species (ROS) in apoptosis and autophagy induced by FTY720 in multiple myeloma cell line U266. U266 cells were treated by different concentrations of FTY720 for 24 h, the apoptotic rates were detected by flow cytometry, and the expression of LC3B was detected by Western blot. The results indicated that apoptosis and autophagy were induced by FTY720 in U266 cells. Autophagy induced by FTY720 could lead to cell death. Bafilomycin A1, the inhibitor of autophagy, could enhance the cell viability in U266 cells treated with FTY720. NAC or Tiron, ROS scavenger, could decrease the FTY720 induced apoptosis and the expression of LC3B-II was reduced in combination of FTY720 with NAC or Tiron as compared with treatment with FTY720 only. It is concluded that FTY720 can induce U266 cell apoptosis and autophagy. ROS is the mediator that regulates both the apoptosis and autophagy in multiple myeloma cells.
1,2-Dihydroxybenzene-3,5-Disulfonic Acid Disodium Salt
;
Apoptosis
;
drug effects
;
Autophagy
;
drug effects
;
Cell Line, Tumor
;
Fingolimod Hydrochloride
;
Humans
;
Macrolides
;
Microtubule-Associated Proteins
;
metabolism
;
Multiple Myeloma
;
metabolism
;
pathology
;
Propylene Glycols
;
pharmacology
;
Reactive Oxygen Species
;
metabolism
;
Sphingosine
;
analogs & derivatives
;
pharmacology
7.Stimulation of sphingosine-1-phosphate on cardiomyogenic differentiation of mesenchymal stem cells.
Lili JIANG ; Tianqing LIU ; Kedong SONG ; Shui GUAN ; Xiangqin LI ; Dan GE
Chinese Journal of Biotechnology 2013;29(11):1617-1628
To study the effect of sphingosine-1-phosphate (S1P) on the cardiomyogenic differentiation of human umbilical cord mesenchymal stem cells (UC-MSCs) and human adipose-derived mesenchymal stem cells (AD-MSCs), we seeded the cells in the culture plates and used cardiomyocyte culture medium (CMCM) combining with different concentration of S1P to induce UC-MSCs and AD-MSCs in vitro for 7, 14 and 28 days. Cardiomyogenic differentiations were identified through immunofluorescence staining, and the results were observed with fluorescence microscopy and confocal microscopy. The effects of S1P and CMCM on cell activity were evaluated by the methyl thiazolyl tetrazolium assay. The functional characteristic similar to cardiomyocytes was evaluated through detecting calcium transient. Our results showed that cardiomyogenic differentiation of UC-MSCs or AD-MSCs were enhanced with S1P concentration increasing, but cell activities declined. Results showed that the suitable differentiation time was 14 days, and the optimal concentration of S1P was 0.5 micromol/L. When working together with CMCM, S1P could promote the differentiation of UC-MSCs or AD-MSCs into functional cardiomyocytes, giving rise to specific electrophysiological properties (the calcium transient). Taken together, our results suggested that S1P could promote the differentiation of UC-MSCs or AD-MSCs into functional cardiomyocytes when being cultured in CMCM.
Adipose Tissue
;
cytology
;
metabolism
;
Cell Differentiation
;
drug effects
;
Cells, Cultured
;
Culture Media
;
Humans
;
Lysophospholipids
;
pharmacology
;
Mesenchymal Stromal Cells
;
cytology
;
drug effects
;
Myocytes, Cardiac
;
cytology
;
Sphingosine
;
analogs & derivatives
;
pharmacology
;
Umbilical Cord
;
cytology
8.Simvastatin inhibits sphingosylphosphorylcholine-induced differentiation of human mesenchymal stem cells into smooth muscle cells.
Kyung Hye KIM ; Young Mi KIM ; Mi Jeong LEE ; Hyun Chang KO ; Moon Bum KIM ; Jae Ho KIM
Experimental & Molecular Medicine 2012;44(2):159-166
Sphingosylphosphorylcholine (SPC) induces differentiation of human adipose tissue-derived mesenchymal stem cells (hASCs) into smooth muscle-like cells expressing alpha-smooth muscle actin (alpha-SMA) via transforming growth factor-beta1/Smad2- and RhoA/Rho kinase-dependent mechanisms. 3-Hydroxy-3-methylglutaryl-coenzyme A reductase inhibitors (statins) have been known to have beneficial effects in the treatment of cardiovascular diseases. In the present study, we examined the effects of simvastatin on the SPC-induced alpha-SMA expression and Smad2 phosphorylation in hASCs. Simvastatin inhibited the SPC-induced alpha-SMA expression and sustained phosphorylation of Smad2 in hASCs. SPC treatment caused RhoA activation via a simvastatin-sensitive mechanism. The SPC-induced alpha-SMA expression and Smad2 phosphorylation were abrogated by pretreatment of the cells with the Rho kinase inhibitor Y27632 or overexpression of a dominant negative RhoA mutant. Furthermore, SPC induced secretion of TGF-beta1 and pretreatment with either Y27632 or simvastatin inhibited the SPC-induced TGF-beta1 secretion. These results suggest that simvastatin inhibits SPC-induced differentiation of hASCs into smooth muscle cells by attenuating the RhoA/Rho kinase-dependent activation of autocrine TGF-beta1/Smad2 signaling pathway.
Amides/pharmacology
;
Blotting, Western
;
Cell Differentiation/*drug effects
;
Cells, Cultured
;
Enzyme-Linked Immunosorbent Assay
;
Humans
;
Immunohistochemistry
;
Mesenchymal Stem Cells/*cytology/*drug effects
;
Myocytes, Smooth Muscle/*cytology/*drug effects
;
Phosphorylcholine/*analogs & derivatives/pharmacology
;
Pyridines/pharmacology
;
Simvastatin/*pharmacology
;
Sphingosine/*analogs & derivatives/pharmacology
;
rhoA GTP-Binding Protein/antagonists & inhibitors/metabolism
9.Antitumor effect of sphingosine kinase 1 inhibitor in combination with chemotherapy on SGC7901 gastric cancer cells in vitro.
Guo-Jian YIN ; Kang-Hua LAN ; Chuang-Ying HU ; Qin LU ; Wen TANG ; Shao-Feng WANG
Chinese Journal of Oncology 2012;34(2):96-99
OBJECTIVETo study the effect of the sphingosine kinase 1 (SphK1) inhibitor N,N-dimethylsphingosine (DMS) in combination with chemotherapeutic drugs (DDP, 5-Fu, MMC) on the proliferation of gastric cancer cells (SGC7901) in vitro, and to evaluate whether SphK1 inhibitors could be used as synergetic agents in chemotherapy.
METHODSSGC7901 cells were incubated in vitro with DMS (1 micromol/L) and 5-Fu, DDP, MMC at different concentrations in combination or separately for 24 h. The effects on the growth and survival of SGC7901 cells were determined by MTT assay. The inhibition rates were assessed by response surface analysis and the interactive relationships between the combined drugs were evaluated on the basis of positive/negative values of the cross product coefficients in the response surface equation.
RESULTSThe growth inhibition rate of the gastric cancer cells by treatment with DMS (1 micromol/L) was (10.23 +/- 0.74)%. The growth inhibition rates of the gastric cancer cells treated with 5-Fu (1, 5 and 25 microg/ml) for 24 h were (9.95 +/- 3.24)%, (21.04 +/- 2.19)%, and (45.49 +/- 3.60)%, respectively. The growth inhibition rates of the gastric cancer cells treated with DDP (0.5, 2.5 and 12.5 microg/ml) for 24 h were (9.38 +/- 0.79)%, (19.61 +/- 0.90)%, and (29.83 +/- 0.54)%, respectively. The growth inhibition rates of the gastric cancer cells treated with MMC (0.1, 0.5 and 2.5 microg/ml) for 24 h were (15.35 +/- 0.77)%, (24.72 +/- 0.83)%, and (30.68 +/- 0.28)%, respectively. There were significant differences among the inhibition rates caused by different concentrations of the drugs (P < 0.05). When 1 micromol/L DMS was used in combination with 5-Fu (1, 5, and 25 microg/ml) for 24 h, the growth inhibition rates of the cancer cells were (16.76 +/- 0.41)%, (27.28 +/- 0.29)% and (52.56 +/- 3.60)%, respectively. When 1 micromol/L DMS was used in combination with DDP (0.5, 2.5, and 12.5 microg/ml) for 24 h, the growth inhibition rates of the cancer cells were (15.35 +/- 0.86)%, (25.57 +/- 0.27)%, (36.37 +/- 0.51)%, respectively. When 1 micromol/L DMS was used in combination with MMC (0.1, 0.5, and 2.5 microg/ml) for 24 h, the growth inhibition rates of the cancer cells were (21.02 +/- 0.28)%, (32.10 +/- 0.27)%, (36.36 +/- 0.28)%, respectively. There were also significant differences among the growth inhibition rates caused by different concentrations of the drugs alone and in combination groups (P < 0.05).
CONCLUSIONSDMS can suppress the proliferation of SGC7901 cells in vitro, and there are evident synergetic effects when it is used in combination with chemotherapeutic drugs. The results of this study indicate that SphK1 inhibitors may become novel and promising chemotherapeutic sensitizers.
Antibiotics, Antineoplastic ; pharmacology ; Antimetabolites, Antineoplastic ; pharmacology ; Antineoplastic Agents ; pharmacology ; Cell Line, Tumor ; Cell Proliferation ; drug effects ; Cisplatin ; pharmacology ; Drug Synergism ; Enzyme Inhibitors ; pharmacology ; Fluorouracil ; pharmacology ; Humans ; Mitomycin ; pharmacology ; Phosphotransferases (Alcohol Group Acceptor) ; antagonists & inhibitors ; Sphingosine ; analogs & derivatives ; pharmacology ; Stomach Neoplasms ; pathology
10.Antiobesity activity of a sphingosine 1-phosphate analogue FTY720 observed in adipocytes and obese mouse model.
Myung Hee MOON ; Jae Kyo JEONG ; Ju Hee LEE ; Yang Gyu PARK ; You Jin LEE ; Jae Won SEOL ; Sang Youel PARK
Experimental & Molecular Medicine 2012;44(10):603-614
Higher levels of body fat are associated with an increased risk for development numerous adverse health conditions. FTY720 is an immune modulator and a synthetic analogue of sphingosine 1-phosphate (S1P), activated S1P receptors and is effective in experimental models of transplantation and autoimmunity. Whereas immune modulation by FTY720 has been extensively studied, other actions of FTY720 are not well understood. Here we describe a novel role of FTY720 in the prevention of obesity, involving the regulation of adipogenesis and lipolysis in vivo and in vitro. Male C57B/6J mice were fed a standard diet or a high fat diet (HFD) without or with FTY720 (0.04 mg/kg, twice a week) for 6 weeks. The HFD induced an accumulation of large adipocytes, down-regulation of phosphorylated AMP-activated protein kinase alpha (p-AMPKalpha) and Akt (p-Akt); down-regulation of hormone-sensitive lipase (HSL), adipose triglyceride lipase (ATGL) and perilipin mRNA as well as up-regulation of phosphorylated HSL (p-HSL, Ser563) and glycogen synthase kinase 3 alpha/beta (p-GSK3alpha/beta). All these effects were blunted by FTY720 treatment, which inhibited adipogenesis and promoted lipolysis. Also, FTY720 significantly decreased lipid accumulation in maturing preadipocytes. FTY720 down-regulated the transcriptional levels of the PPARgamma, C/EBPalpha and adiponectin, which are markers of adipogenic differentiation. FTY720 significantly increased the release of glycerol and the expression of the HSL, ATGL and perilipin, which are regulators of lipolysis. These results show that FTY720 prevented obesity by modulating adipogenesis and lipolysis, and suggest that FTY720 is used for the treatment of obesity.
3T3-L1 Cells
;
AMP-Activated Protein Kinases/metabolism
;
Adipocytes/*drug effects/physiology
;
Adipogenesis/drug effects
;
Animals
;
Anti-Obesity Agents/*pharmacology/therapeutic use
;
Antigens, Differentiation/genetics/metabolism
;
Carrier Proteins/genetics/metabolism
;
Cell Size
;
Diet, High-Fat/adverse effects
;
Disease Models, Animal
;
Enzyme Activation
;
Gene Expression Regulation, Enzymologic/drug effects
;
Glycogen Synthase Kinase 3/genetics/metabolism
;
Lipase/genetics/metabolism
;
Lipolysis/drug effects
;
Male
;
Mice
;
Mice, Inbred C57BL
;
Obesity/etiology/metabolism/*prevention & control
;
Phosphoproteins/genetics/metabolism
;
Phosphorylation
;
Propylene Glycols/*pharmacology/therapeutic use
;
Protein Processing, Post-Translational
;
Proto-Oncogene Proteins c-akt/metabolism
;
Sphingosine/*analogs & derivatives/pharmacology/therapeutic use
;
Sterol Esterase/metabolism

Result Analysis
Print
Save
E-mail