1-磷酸鞘氨醇对2型糖尿病小鼠胰岛β细胞损伤的保护作用
详细信息    查看全文 | 推荐本文 |
  • 英文篇名:Protective effects of sphingosine-1-phosphate on islet β-cell proliferation and less cell apoptosis in type 2 diabetic mice
  • 作者:隋婧 ; 和一之 ; 邓梅 ; 赵心蕊 ; 施秉银
  • 英文作者:SUI Jing;HE Yi-zhi;DENG Mei;ZHAO Xin-rui;SHI Bing-yin;Department of Endocrinology and International Medical Center,The First Affiliated Hospital of Xi'an Jiaotong University;Department of Endocrinology,Xi'an No.3 Hospital;Department of Endocrinology,Shanxian Central Hospital;Department of Immunology and Rheumatology,The Second Affiliated Hospital of Xi'an Jiaotong University;Department of Endocrinology,The First Affiliated Hospital of Xi'an Jiaotong University;
  • 关键词:1-磷酸鞘氨醇 ; 1-磷酸鞘氨醇受体 ; 2型糖尿病 ; 胰岛β细胞 ; 增殖 ; 凋亡 ; S1P/S1PR信号通路
  • 英文关键词:sphingosine-1-phosphate;;sphingosine-1-phosphate receptor;;type 2 diabetes mellitus;;islet β-cell;;proliferation;;apoptosis;;S1P/S1PR signaling pathway
  • 中文刊名:XAYX
  • 英文刊名:Journal of Xi'an Jiaotong University(Medical Sciences)
  • 机构:西安交通大学第一附属医院内分泌科涉外病房;西安市第三医院内分泌科;山东省单县中心医院内分泌科;西安交通大学第二附属医院风湿免疫科;西安交通大学第一附属医院内分泌科;
  • 出版日期:2018-10-16 09:45
  • 出版单位:西安交通大学学报(医学版)
  • 年:2018
  • 期:v.39;No.215
  • 基金:中华医学会临床医学科研专项基金项目(No.13040470432);; 西安交通大学第一附属医院院基金(No.2013YK20)~~
  • 语种:中文;
  • 页:XAYX201806012
  • 页数:8
  • CN:06
  • ISSN:61-1399/R
  • 分类号:63-70
摘要
目的研究1-磷酸鞘氨醇(S1P)对2型糖尿病(T2DM)小鼠胰岛β细胞的促进增殖、抑制凋亡情况和对血糖的影响,以及S1P受体S1PR1-3在T2DM小鼠胰腺中的表达定位,探讨S1P对糖尿病胰岛β细胞损伤的保护作用。方法雄性C57BL/6J小鼠,高脂饮食联合小剂量链脲佐菌素(STZ)制备T2DM小鼠模型,随机分为S1P高剂量组(S1PH)、S1P低剂量组(S1PL)、糖尿病对照组(DC)及正常对照组(NC),每日予S1PH组和S1PL组S1P溶液100μg/kg及20μg/kg腹腔注射,DC组及NC组予同体积溶剂替代。给药3周后行葡萄糖耐量试验(IPGTT),胰腺组织HE及TUNEL染色,免疫组化检测胰岛中胰岛素、Ki67及S1PR1-3蛋白的表达。结果 S1P给药3周后,S1PL组及S1PH组空腹血糖及IPGTT 2h血糖较DC组轻度下降。胰岛组织HE染色及胰岛素免疫组化染色示S1PL组及S1PH组较DC组胰岛病变减轻。Ki67免疫组化染色示S1PL组及S1PH组胰岛β细胞增殖率较DC组明显升高(P<0.05);TUNEL染色示S1PL组及S1PH组胰岛β细胞凋亡率较DC组明显降低(P<0.05)。胰岛组织免疫组化示S1P受体S1PR1-3均表达于小鼠胰岛β细胞中,在胰腺组织外分泌部表达甚微或不表达。其中S1PL组、S1PH组和DC组S1PR1及S1PR2的表达均较NC组明显增多,各组间S1PR3表达差异无统计学意义。结论 S1P与其受体S1PR1及S1PR2结合能够显著改善T2DM小鼠胰岛β细胞形态,促进小鼠胰岛β细胞增殖、抑制凋亡,提示S1P对T2DM胰岛β细胞损伤有保护作用。S1P/S1PR信号通路在T2DM中发挥着重要作用,有可能成为未来药物治疗的新靶点。
        Objective To explore the potential protective effects of sphingosine-1-phosphate(S1P)on pancreatic isletβ-cell proliferation and less cell apoptosis in type 2 diabetic mice and to detect the localization and expression of S1P receptors in the islets of type 2 diabetic mice.Methods Mice with type 2 diabetes induced by high-fat diet with low-dose streptozotocin were randomly divided into 4 groups:high-dose S1P administration group (S1PH),low-dose S1P administration group(S1PL),diabetic model control group(DC),and normal control group(NC).Exogenous S1P was intraperitoneally injected daily at a dose of 100μg/kg for S1PH group and 20μg/kg for S1PL group for 3 weeks.The potential protective effects of S1P onβ-cell proliferation and apoptosis were determined.Intraperitoneal glucose tolerance test(IPGTT)and homeostasis model assessment(HOMA)index were assessed to test the effects of S1P on islet function.Expressions of insulin,Ki67,S1P receptor isoforms(S1PR1,S1PR2 and S1PR3)proteins in pancreatic islets were tested by immunohistochemical staining.Results Three weeks after S1P administration,fasting blood glucose and 2-hour glucose after injection during IPGTT in the two S1P administration groups were decreased slightly compared to DC group.β-cell proliferation rate was higher and the apoptosis rate was lower in S1PH and S1PL groups than in DC group(P<0.05).S1P receptors S1PR1,S1PR2 and S1PR3 were positively stained in the islet cell cytoplasm and membrane.S1PR1 and S1PR2 proteins were significantly expressed in S1PH and S1PL groups and DC diabetic groups compared with NC group(P<0.01),while S1PR3 expression did not significantly differ among these groups.Conclusion Extracellular S1P promotesβ-cell proliferation and suppresses cell apoptosis via S1P receptor isoforms S1PR1 and S1PR2 in diabetic mice.Therefore,S1P/S1PR signaling can be considered as a potential therapeutic target for the treatment of diabetes for its important role in T2DM.
引文
[1]WANG L,GAO P,ZHANG M,et al.Prevalence and ethnic pattern of diabetes and prediabetes in China in 2013[J].JAMA,2017,317(24):2515-2523.
    [2]ALSHEHRY ZH,MUNDRA PA,BARLOW CK,et al.Plasma lipidomic profiles improve on traditional risk factors for the prediction of cardiovascular events in type 2 diabetes mellitus[J].Circulation,2016,134(21):1637-1650.
    [3]JESSUP CF,BONDER CS,PITSON SM,et al.The sphingolipid rheostat:A potential target for improving pancreatic islet survival and function[J].Endocr Metab Immune Disord Drug Targets,2011,11(4):262-272.
    [4]MEYER ZU HERINGDORF D.Lysophospholipid receptor-dependent and-independent calcium signaling[J].J Cell Biochem,2004,92(5):937-948.
    [5]SPIEGEL S,MILSTIEN S.Sphingosine 1-phosphate,a key cell signaling molecule[J].J Biol Chem,2002,277(29):25851-25854.
    [6]MASTRANDREA LD,SESSANNA SM,LAYCHOCK SG.Sphingosine kinase activity and sphingosine-1 phosphate production in rat pancreatic islets and INS-1 cells:Response to cytokines[J].Diabetes,2005,54(5):1429-1436.
    [7]CHUN J,HLA T,LYNCH KR,et al.International Union of Basic and Clinical Pharmacology.LXXVIII.Lysophospholipid receptornomenclature[J].Pharmacol Rev,2010,62(4):579-587.
    [8]VOLZKE A,KOCH A,MEYER ZU HERINGDORF D,et al.Sphingosine 1-phosphate(S1P)induces COX-2 expression and PGE2 formation via S1P receptor 2 in renal mesangial cells[J].Biochim Biophys Acta,2014,1841(1):11-21.
    [9]LEE SY,LEE HY,SONG JH,et al.Adipocyte-specific deficiency of de novo sphingolipid biosynthesis leads to lipodystrophy and insulin resistance[J].Diabetes,2017,66(10):2596-2609.
    [10]CUVILLIER O.Sphingosine 1-phosphate receptors:from biology to physiopathology[J].Med Sci(Paris),2012,28(11):951-957.
    [11]HATOUM D,HADDADI N,LIN Y,et al.Mammalian sphingosine kinase(SphK)isoenzymes and isoform expression:Challenges for SphK as an oncotarget[J].Oncotarget,2017,8(22):36898-36929.
    [12]MATTHEWS DR,HOSKER JP,RUDENSKI AS,et al.Homeostasis model assessment:insulin resistance and beta-cell function from fasting plasma glucose and insulin concentrations in man[J].Diabetologia,1985,28(7):412-419.
    [13]PULKOSKI-GROSS MJ,DONALDSON JC,OBEID LM.Sphingosine-1-phosphate metabolism:A structural perspective[J].Crit Rev Biochem Mol Biol,2015,50(4):298-313.
    [14]MACEYKA M,HARIKUMAR KB,MILSTIEN S,et al.Sphingosine-1-phosphate signaling and its role in disease[J].Trends Cell Biol,2012,22(1):50-60.
    [15]SOLTAU I,MUDERSBACH E,GEISSEN M,et al.Serumsphingosine-1-phosphate concentrations are inversely associated with atherosclerotic diseases in humans[J].PLoS One,2016,11(12):e0168302.
    [16]HAASS NK,NASSIF N,MCGOWAN EM.Switching the sphingolipid rheostat in the treatment of diabetes and cancer comorbidity from a problem to an advantage[J].Biomed Res Int,2015,2015:165105.
    [17]MOON H,CHON J,JOO J,et al.FTY720 preserved isletβ-cell mass by inhibiting apoptosis and increasing survival ofβ-cells in db/db mice[J].Diabetes Metab Res Rev,2013,29(1):19-24.
    [18]SHIMIZU H,OKAJIMA F,KIMURA T,et al.Sphingosine 1-phosphate stimulates insulin secretion in HIT-T 15 cells and mouse islets[J].Endocr J,2000,47(3):261-269.
    [19]SERAFIMIDIS I,RODRIGUEZ-AZNAR E,LESCHE M,et al.Pancreas lineage allocation and specification are regulated by sphingosine-1-phosphate signalling[J].PLoS Biol,2017,15(3):e2000949.
    [20]LAYCHOCK SG,TIAN Y,SESSANNA SM.Endothelial differentiation gene receptors in pancreatic islets and INS-1 cells[J].Diabetes,2003,52(8):1986-1993.
    [21]KITADA Y,KAJITA K,TAGUCHI K,et al.Blockade of sphingosine 1-phosphate receptor 2 signaling attenuates high-fat diet-induced adipocyte hypertrophy and systemic glucose intolerance in mice[J].Endocrinology,2016,157(5):1839-1851.