首页 | 本学科首页   官方微博 | 高级检索  
相似文献
 共查询到19条相似文献,搜索用时 109 毫秒
1.
生长抑素(somatostatin,SST)通过与细胞膜上的G蛋白偶联的生长抑素受体(somatostatin receptors,SSTRs)结合而发挥其抑制细胞增殖的作用,因而生长抑素类似物(somatostatin analogue, SSA)常被用于肿瘤辅助治疗。然而,治疗效果存在相当大的个体差异,推测生长抑素类似物治疗效果不佳,与内源性生长抑素受体表达缺失或者表达量和亚型组合有关。为此,检测各亚型SSTR在几例罕见的神经内分泌肿瘤中的表达,并检测过表达SSTR2和SSTR5以及受体激活对细胞增殖的抑制效果,分析受体激活的可能机制,有助于临床筛选适合SSA肿瘤辅助治疗的病例,预估SSA的治疗效果。免疫组化检测肿瘤组织SSTR1-5的表达。在培养的293T细胞中过表达SSTR2和SSTR5,免疫共沉淀检测受体相互作用,免疫荧光和共聚焦显微镜检测受体细胞内定位。用MTT法检测受体过表达及激活对培养的人肺癌细胞NCI-H460细胞增殖的影响,用流式细胞技术检测细胞周期分布。SSTR1-5在10例神经内分泌肿瘤组织中均有不同程度的表达,表达亚型及表达量与肿瘤类型和年龄无关,SSTR5在所有肿瘤组织中均表达。SSTR2与SSTR5可形成受体相互作用。SSTR2与SSTR5活化后相互作用增加并定位于细胞质。共表达SSTR2和SSTR5显著抑制细胞增殖,并与受体激活剂呈现剂量相关性。SSTR2/SSTR5的共表达及激活显著减少S期的细胞而滞留于G1期。  相似文献   

2.
游离脂肪酸(free fatty acid,FFA)是动物一种重要能量来源,同时它还是一种重要的信号分子,其生理功能和作用机制长期以来倍受关注. 最近研究表明,细胞膜存在FFA的特定孤儿型G蛋白偶联膜受体家族.中长链游离脂肪酸是GPR40和GPR120的配基,而短链游离脂肪酸则是GPR41和GPR43的配基. 该受体家族可以介导游离脂肪酸,通过ERK、PI3K-Akt和MAPK信号通路,在维持机体内的葡萄糖稳态、脂肪形成、白细胞功能和细胞增殖等生理过程中发挥重要作用. 本文就游离脂肪酸G蛋白偶联受体的结构、分布、配体选择性、下游信号通路,及其介导FFA生理功能的最新研究进展进行简要综述.  相似文献   

3.
G蛋白偶联受体家族卵巢癌G蛋白偶联受体1(ovarian cancer G protein-coupled receptor 1, OGR1)亚家族的OGR1、T细胞死亡偶联基因8(T-cell death associated gene 8, TDAG8)、G 蛋白偶联受体4(G protein-coupled receptor 4, GPR4)及诱导细胞停滞于G2/M期的G蛋白偶联受体G2A(from G2 accumulation)4 种受体是最新发现的一类质子感知受体.除了质子,体内又有它们各自特定的脂质分 子配体活化这些受体来调节细胞机能.该类受体广泛分布于人的各种正常组织和肿瘤 组织细胞中,在肿瘤的发生与转移、细胞骨架重组等生理病理过程中发挥双重作用. 正常表达时它们有一定的抑制肿瘤作用,但这些受体的异常表达或过表达使某些组织 和细胞恶性转化,导致肿瘤的发生.本文综述了在肿瘤组织的酸性微环境中,细胞表 达的质子(pH)感知受体对肿瘤发生与肿瘤转移的调节作用及其相关的信号通路.  相似文献   

4.
细胞外钙受体(CaR)为G蛋白偶联受体超家族中的成员,它的大部分作用是以Gαi,Gαq和Gα12/13为中介的,但由G蛋白α亚基介导的作用并不能完全解释CaR的生物学效应.与CaR相互作用蛋白如抑制蛋白、G蛋白受体激酶、受体激活修饰蛋白、丝蛋白、钾通道、小窝蛋白等结构和信号蛋白赋予CaR独特的信号转导特征,并能够更充分说明CaR在不同组织和细胞中所发挥的作用.本文将对上述几种与其相互作用蛋白及它们所产生的生物学效应做一综述.  相似文献   

5.
代谢型谷氨酸受体l/5(mGluR1/5)是G蛋白偶联受体家族C的重要成员之一,该受体及其介导的下游信号在调节神经系统的正常生理功能起着非常重要作用,并与相关神经系统退行性疾病密切相关。文章介绍了mGluR1/5所介导的信号通路、信号通路调控的分子机制以及其他GPCR受体的相互作用对信号共同调节的分子机制等方面最新研究进展。  相似文献   

6.
《遗传》2020,(8)
G蛋白偶联受体(G protein-coupled receptors, GPCRs)作为最大的一类膜蛋白受体家族,可被多种配体激活并发挥相应的信号转导功能,参与生物体内重要的生理过程。G蛋白偶联受体相关分选蛋白(G protein-coupled receptors associated sorting proteins, GASPs)则对内吞后的GPCRs分选过程发挥着重要的作用,并介导受体进入降解或再循环途径,进而调控细胞的信号转导等过程。研究发现GASPs的功能缺陷与多种疾病相关,包括神经系统疾病、肿瘤和耳聋等。本文重点介绍了G蛋白偶联受体相关分选蛋白的功能特征及其相关信号通路,描述了GASPs功能缺陷与疾病的关联性及家族蛋白与GPCRs的相互作用、GASPs分选途径的发现、参与的信号通路及对基因转录调控,以期为GASPs相关多种疾病的治疗提供新的思路和策略。  相似文献   

7.
G蛋白偶联受体3(Gpr3)属于G蛋白偶联受体视紫质家族成员. Gpr3通过激活Gs蛋白介导的下游信号通路,维持卵泡卵母细胞减数分裂的前期阻滞,但在卵泡颗粒细胞中的作用不清. 为了明确Gpr3在猪卵泡颗粒细胞中的功能,构建了Gpr3基因的真核表达载体,利用过表达的方式激活其介导的信号通路,并利用MTT、流式细胞术和real-time PCR等方法检测了过表达Gpr3对猪卵泡颗粒细胞增殖及凋亡的影响. 结果显示,过表达Gpr3后,猪颗粒细胞的增殖水平显著下调,G0/G1期细胞的百分比增加,S期细胞减少,Cyclin B1和CDK1 mRNA的表达量也显著降低;同时,显著增加了颗粒细胞的凋亡率,在抑制Bcl-2表达的同时,促进了Bax的表达. 结果表明:过表达Gpr3在猪颗粒细胞中具有抑增殖促凋亡的作用,丰富了其在调节卵泡发育过程中的生物学功能.  相似文献   

8.
程智洁  裴钢 《生命科学》1999,11(4):169-171
对Arrestins家族的研究是当今生物学中信号传导研究领域的热点之一。Arrestins可以作用于G蛋白偶联受体,使受体与下游的G蛋白解偶联;并充当受体与笼形蛋白的接头,促进衣被蛋白介导的受体内吞;在受体内吞过程中,Arrestins、受体和c-Src激酶共同起始MAPK信号传导途径。本综述概括了近年来Arrestins研究的最新进展,包括其分类、主要蛋白功能结构域、基因定位和在G蛋白偶联受体信号传导中的地位与作用。  相似文献   

9.
G蛋白偶联受体激酶(GRK)是G蛋白偶联受体(GPCR)信号通路的负性调节因子。近来的研究发现,GRK除了磷酸化G蛋白偶联受体使其脱敏外,还能与其他非受体底物结合,功能呈现多样性。GRK5是GRK家族成员之一,该研究探索了GRK5在细胞周期和有丝分裂中的作用,结果显示:在细胞内干扰GRK5的表达导致分裂中期的细胞数目增多和细胞凋亡。进一步的研究发现,干扰GRK5的表达导致有丝分裂中期的染色体不能正常排列到赤道板,而对分裂后期染色质分离以及胞质分裂没有影响。在细胞内干扰GRK蛋白家族的另一个成员GRK2对有丝分裂则没有明显影响。该研究提示GRK5是细胞有丝分裂的重要调控蛋白。  相似文献   

10.
Raf激酶抑制蛋白(RKIP)的生物学功能研究   总被引:4,自引:0,他引:4  
胡新宙  王旭  张伟 《生命科学》2007,19(1):31-36
Raf激酶抑制蛋白(Raf kinase inhibitor protein,RKIP)属于磷脂酰乙醇胺结合蛋白(phosphatidylethanolamine-binding protein,PEBP)家族,广泛存在于各种生物中,参与了对细胞内多种信号转导通路的调节作用。RKIP可以与Raf-1结合,从而抑制MAPK信号转导通路,并参与了对G蛋白偶联受体信号通路和NF-κB信号通路的调控。RKIP在膜的生物合成、精子发生、神经发育和细胞凋亡等生理过程中发挥重要作用,并参与了老年痴呆症及糖尿病等的病理过程。此外,近年来的研究表明RKIP是一个新的转移抑制因子,可以抑制前列腺癌、人乳腺癌和黑色素瘤细胞的转移,并已成为一个新的前列腺癌诊断标志物。  相似文献   

11.
12.
Somatostatins are a diverse family of peptide hormones that regulate various aspects of growth, development, and metabolism through interactions with numerous somatostatin receptor subtypes (SSTRs) on target tissues. In this study, we used rainbow trout to evaluate the effects of growth hormone (GH), insulin (INS), and insulin-like growth factor-I (IGF-I) on the expression of SSTR 1A, 1B and 2 mRNAs. GH regulated the expression of SSTRs in a subtype- and tissue-specific manner. GH reduced SSTR 1A, 1B, and 2 expression in optic tectum, reduced SSTR 1A and 1B expression in pancreas, reduced SSTR 1A expression in liver, and increased hepatic SSTR 1B expression. INS also regulated SSTR expression in a subtype- and tissue-specific manner. INS reduced SSTR 1B expression in optic tectum, increased SSTR 2 expression in pancreas, and increased SSTR 1B and 2 expression in liver. IGF-I generally decreased the expression of all SSTRs. These data indicate that GH, INS, and IGF-I modulate the expression of SSTRs and suggest that independent mechanisms may serve to regulate the various receptor subtypes.  相似文献   

13.
The present study describes the status of somatostatin receptors (SSTRs) and their colocalization with insulin (β), glucagon (α) and somatostatin (δ) producing cells in the pancreatic islets of 11 weeks old R6/2 Huntington's Disease transgenic (HD tg) and age-matched wild type (wt) mice. We also determined expression of tyrosine hydroxylase (TH), glutamic acid decarboxylase (GAD) and presynaptic marker synaptophysin (SYP) in addition to signal transduction pathways associated with diabetes. In R6/2 mice, islets are relatively smaller in size, exhibit enhanced expression and nuclear inclusion of mHtt along with the loss of insulin, glucagon and somatostatin expression. In comparison to wt, R6/2 mice display enhanced mRNA for all SSTRs except SSTR2. In the pancreatic lysate, SSTR1, 4 and 5 immunoreactivity decreases whereas SSTR3 immunoreactivity increases with no discernible changes in SSTR2 immunoreactivity. Furthermore, at the cellular level, R6/2 mice exhibit a receptor specific distributional pattern of SSTRs like immunoreactivity and colocalization with β, α and δ cells. While GAD expression is increased, TH and SYP immunoreactivity was decreased in R6/2 mice, anticipating a cross-talk between the CNS and pancreas in diabetes pathophysiology. We also dissected out the changes in signaling pathway and found decreased activation and expression of PKA, AKT, ERK1/2 and STAT3 in R6/2 mice pancreas. These findings suggest that the impaired organization of SSTRs within islets may lead to perturbed hormonal regulation and signaling. These interconnected complex events might shed new light on the pathogenesis of diabetes in neurodegenerative diseases and the role of SSTRs in potential therapeutic intervention.  相似文献   

14.
Expression of somatostatin receptors 1 and 2 in the adult mouse kidney   总被引:6,自引:0,他引:6  
  相似文献   

15.
Neuroendocrine tumours (NETs) may occur at many sites in the body although the majority occur within the gastroenteropancreatic axis. Non-gastroenteropancreatic NETs encompass phaeochromocytomas and paragangliomas, medullary thyroid carcinoma, anterior pituitary tumour, broncho-pulmonary NETs and parathyroid tumours. Like most endocrine tumours, NETs also express somatostatin (SST) receptors (subtypes 1–5) whose ligand SST is known to inhibit endocrine and exocrine secretions and have anti-tumour effects. In the light of this knowledge, the idea of using SST analogues in the treatment of NETs has become increasingly popular and new studies have centred upon the development of new SST analogues. We attempt to review SST receptor (SSTR) biology primarily in neuroendocrine tissues, focusing on pituitary tumours. A full data search was performed through PubMed over the years 2000–2009 with keywords ‘somatostatin, molecular biology, somatostatin receptors, somatostatin signalling, NET, pituitary’ and all relevant publications have been included, together with selected publications prior to that date. SSTR signalling in non-neuroendocrine solid tumours is beyond the scope of this review. SST is a potent anti-proliferative and anti-secretory agent for some NETs. The successful therapeutic use of SST analogues in the treatment of these tumours depends on a thorough understanding of the diverse effects of SSTR subtypes in different tissues and cell types. Further studies will focus on critical points of SSTR biology such as homo- and heterodimerization of SSTRs and the differences between post-receptor signalling pathways of SSTR subtypes.  相似文献   

16.
Somatostatin regulates multiple biological functions by acting through a family of five G protein-coupled receptors, somatostatin receptors (SSTRs) 1-5. Although all five receptor subtypes inhibit adenylate cyclase activity and decrease intracellular cAMP levels, specific receptor subtypes also couple to additional signaling pathways. In CCL39 fibroblasts expressing either human SSTR1 or SSTR2, we demonstrate that activation of SSTR1 (but not SSTR2) attenuated both thrombin- and integrin-stimulated Rho-GTP complex formation. The reduction in Rho-GTP formation in the presence of somatostatin was associated with decreased translocation of Rho and LIM kinase to the plasma membrane and fewer focal contacts. Activation of Rho resulted in the formation of intracellular actin stress fibers and cell migration. In CCL39-R1 cells, somatostatin treatment prevented actin stress fiber assembly and attenuated thrombin-stimulated cell migration through Transwell membranes to basal levels. To show that native SSTR1 shares the ability to inhibit Rho activation, we demonstrated that somatostatin treatment of human umbilical vein endothelial cells attenuated thrombin-stimulated Rho-GTP accumulation. These data show for the first time that a G protein-coupled receptor, SSTR1, inhibits the activation of Rho, the assembly of focal adhesions and actin stress fibers, and cell migration.  相似文献   

17.
The role of somatostatin (SST) and epidermal growth factor (EGF) in breast cancer is undisputed; however, the molecular mechanisms underlying their antiproliferative or proliferative effects are not well understood. We initially confirmed that breast tumour tissues express all five somatostatin receptors (SSTR1-5) and four epidermal growth factor receptors (ErbB1-4). Subsequently, to gain insight into the function of SSTRs and ErbBs in oestrogen receptor (ER)-positive (MCF-7) or ERα-negative (MDA-MB-231) breast cancer cells, we defined SSTR1, SSTR5 and ErbB1 mRNA and protein expression in these two tumour cell lines. Consistent with previous studies showing SSTR1/SSTR5 heterodimerization and having seen cell-specific and ligand-selective alterations in receptor expression, we next elucidated whether SSTR1 and SSTR5 functionally interact with ErbB1 using pbFRET analysis. We subsequently determined the effects of SST and EGF either alone, or in combination, on selected downstream signalling molecules such as erk1/2, p38 and JNK. Here, we showed that both SST and EGF influenced erk1/2 phosphorylation and that SST modulated the effects of EGF in a cell-specific manner. We also demonstrated agonist-, time and cell-dependent regulation of p38 phosphorylation. We further investigated modulation of Grb2, SOS, Shc, SH-PTP1 and SH-PTP2. ErbB1 adaptor proteins known to play a role in MAPK activation, Shc, Grb2 and SOS, changed in an agonist- and cell-specific manner whereas, SH-PTP1 and SH-PTP2, adaptor proteins reported to interact with SSTRs, translocated from the cytosol to membrane in a cell-specific manner following SST and/or EGF treatment. Although several previous studies have shown crosstalk between RTKs and GPCRs, there are no reports describing SSTR (GPCR) modulation of ErbBs (RTK) in breast cancer. To the best of our knowledge, this is the first report describing crosstalk/interactions between SSTRs and ErbBs.  相似文献   

18.
Five somatostatin receptors (SSTRs) bind somatostatin-14 (S-14) and somatostatin-28 (S-28), but SSTR5 has the highest affinity for S-28. To determine whether S-28 acting through SSTR5 mediates inhibition of glucagon-like peptide-1 (GLP-1), fetal rat intestinal cell cultures were treated with somatostatin analogs with relatively high specificity for SSTRs 2-5. S-28 dose-dependently inhibited GLP-1 secretion stimulated by gastrin-releasing peptide more potently than S-14 (EC(50) 0.01 vs. 5.8 nM). GLP-1 secretion was inhibited by an SSTR5 analog, BIM-23268, more potently than S-14 and nearly as effectively as S-28. The SSTR5 analog L-372,588 also suppressed GLP-1 secretion equivalent to S-28, but a structurally similar peptide, L-362,855 (Tyr to Phe at position 7), was ineffective. An SSTR2-selective analog was less effective than S-28, and an SSTR3 analog was inactive. Separate treatment with GLP-1-(7-36)-NH(2) increased S-28 and S-14 secretion by three- and fivefold; BIM-23268 abolished S-28 without altering S-14, whereas the SSTR2 analog was inactive. The results indicate that somatostatin regulation of GLP-1 secretion occurs via S-28 through activation of SSTR5. GLP-1-stimulated S-28 secretion is also autoregulated by SSTR5 activation, suggesting a feedback loop between GLP-1 and S-28 modulated by SSTR5.  相似文献   

19.
Somatostatin (SST) inhibition of hormone hypersecretion from tumors is mediated by somatostatin receptors (SSTRs). SSTRs also play an important role in controlling tumor growth through specific antiproliferative actions. These receptors are well expressed in numerous normal and tumor tissues and are susceptible to regulation by a variety of factors. Estradiol, a potent trophic and mitogenic hormone in its target tissues, is known to modulate the expression of SST and its receptors. Accordingly, in the present study, we determined the effects of tamoxifen, a selective estrogen receptor (ER) modulator (SERM), and estradiol on SSTR1 and SSTR2 expression at the mRNA and protein levels in ER-positive and -negative breast cancer cells. We found that SSTR1 was upregulated by tamoxifen in a dose-dependent manner but no effect was seen with estradiol. In contrast, SSTR2 was upregulated by both tamoxifen and estradiol. Combined treatment caused suppression of SSTR1 below control levels but had no significant effect on SSTR2. Treatment with SSTR1-specific agonist was significantly more effective in suppressing cell proliferation of cells pre-treated with tamoxifen. Taking these data into consideration, we suggest that tamoxifen and estradiol exert variable effects on SSTR1 and SSTR2 mRNA and protein expression and distributional pattern of the receptors. These changes are cell subtype-specific and affect the ability of SSTR agonists to inhibit cell proliferation.  相似文献   

设为首页 | 免责声明 | 关于勤云 | 加入收藏

Copyright©北京勤云科技发展有限公司  京ICP备09084417号