首页 | 官方网站   微博 | 高级检索  
相似文献
 共查询到20条相似文献,搜索用时 140 毫秒
1.
用稳定过表达并带有myc表位的葡萄糖转运子1(glucose transporter 1, GLUT1)或葡萄糖转运子4(glucose transporter 4, GLUT4)的L6骨骼肌细胞株定征GLUT1和GLUT4对胰岛素的响应. 所筛选的L6-GLUT1myc细胞克隆分化前后的葡萄糖摄取量均在线性范围. 100 nmol/L胰岛素使L6-GLUT1myc和L6-GLUT4myc肌原细胞膜上GLUT1或GLUT4的量分别达到基础组的(1.58±0.01)倍和(1.96±0.11)倍, 2-脱氧葡萄糖摄取量分别达到了(1.53±0.09)倍和(1.86±0.17)倍, 此作用可被渥曼青霉素(wortmannin)抑制. 胰岛素刺激了此2种细胞中的Akt磷酸化. L6-GLUT1myc肌原细胞的葡萄糖摄取量对胰岛素浓度呈剂量依赖性, 但与野生型细胞相比, 其对胰岛素的敏感性和最大响应没有改变. 但L6-GLUT4myc肌原细胞的葡萄糖摄取量对胰岛素的敏感性和最大响应均增加. 以前的研究提示毛喉素(forskolin)可能影响胰岛素刺激的GLUT4转位. 本研究表明, 在L6-GLUT4myc细胞中, 毛喉素使胰岛素刺激的葡萄糖摄取减少了65%, 此作用是由它对GLUT4的直接抑制而不是由其对GLUT4转位的影响造成的. 毛喉素和dipyridamole对GLUT4比对GLUT1有更强的抑制作用, 而戊巴比妥(pentobarbital)对GLUT1的抑制作用强于GLUT4. 应用这些抑制剂的结果表明、L6肌原细胞中基础状态下和胰岛素刺激状态下的葡萄糖主要由过表达的GLUT1或GLUT4转运. 因此, L6-GLUT1myc和L6-GLUT4myc细胞株为筛查对肌肉细胞GLUT1或GLUT4的活性或转位有不同作用的化合物提供了一个平台.  相似文献   

2.
葡萄糖转运蛋白4(glucose transporter 4,GLUT4)参与胰岛素敏感的脂肪细胞和肌肉细胞中的葡萄糖转运,对机体葡萄糖代谢至关重要。磷脂酰肌醇作为各种蛋白质的定位信号,参与调控细胞生长和新陈代谢,在胰岛素信号转导过程中起着关键作用。在过去的几十年里,关于磷脂酰肌醇信号调控GLUT4囊泡转运方面已有了很大的进展。该文总结了磷脂酰肌醇在GLUT4囊泡转运中的调控作用。  相似文献   

3.
葡萄糖转运蛋白4(GLUT4)。主要分布于骨胳肌,心肌及脂肪组织中,当胰岛素与细胞膜受体结合后。产生一系列信号,促进GLUT4从胞内易位至细胞膜,GLUT4通过自身构象改变。将葡萄糖摄入细胞内,从而协助维持血糖的稳定,这些具体信号正在被广泛深入的研究。现在发现至少有两条独立的信号传导途径。一条是经典的PI3K途径。另一条是新近发现的Cb1/CAP途径。深入了解这些信号传导途径。对于揭示2型糖尿病的发病机制有重要的意义。  相似文献   

4.
GLUT4在胰岛素作用下的转运上膜是血糖调控的一个关键途径.其中包含了两个重要的过程-胰岛素信号转导以及GLUT4转运途径.在这两个过程中新的特异分子的发现以及它们功能特点的研究是发展有效的药物治疗糖尿病的关键因素.本文主要从GLUT4在胞内的循环途径,胰岛素调节的GLUT4的转运以及转运中的调控蛋白三个方面着手,综述了GLUT4的转运调控研究进展.  相似文献   

5.
胰岛素抵抗(insulin resistance,IR)是2型糖尿病(type 2 diabetes mellitus,T2DM)的主要诱因,运动因其在改善骨骼肌胰岛素敏感性方面的显著作用,已被临床采用作为防治IR和T2DM的有效手段。运动能增加葡萄糖转运子4 (glucose transporter type 4,Glut4)的转位,而影响Glut4转位和葡萄糖摄取的途径包括胰岛素信号通路和肌肉收缩两大类。研究发现运动通过增加骨骼肌血流灌注、毛细血管募集、胰岛素信号途径和Sestrins-mTOR (mammalian target of rapamycin)信号通路而改善Glut4转位。因此,全面理解运动调节骨骼肌Glut4转位和葡萄糖摄取的分子机制对于揭示运动疗法防治糖代谢异常的机制具有重要意义。  相似文献   

6.
目的:研究量子点标记活细胞内GLUT4蛋白的方法,用于长时程观察活细胞内GLUT4的转运过程。方法:使用在GLUT4蛋白膜外区构建了myc位点的L6-GLUT4myc细胞系,用胰岛素刺激L6细胞内的GLUT4myc转运到细胞膜上,通过抗体抗原反应先后将一抗9E10和偶联二抗IgG的量子点与特异性位点结合。结果:通过量子点标记固定细胞内GLUT4的实验,证明了标记方法的特异性和灵敏性。量子点能够标记细胞膜表面的GLUT4蛋白并伴随GLUT4的胞吞进入细胞。适当调整实验温度,用量子点标记细胞膜上的GLUT4并且在实验过程结束后将标记了量子点的GLUT4保持在细胞膜表面,能够观察活细胞内GLUT4蛋白内化和胞内循环的过程。结论:发展了量子点标记活细胞内GLUT4的方法,为进一步研究活细胞内GLUT4的转运过程打下了基础。  相似文献   

7.
葡萄糖转位载体4(GLUT4)转位的受损与2型糖尿病密切相关,所以筛选促进GLUT4转位的药物并研究其相关的信号通路对治疗2型糖尿病具有十分重要的意义。药物促进GLUT4转位主要通过AMP活化蛋白激酶(AMPK)、磷脂酰肌醇3激酶/蛋白激酶B(PI3K/Akt)和蛋白激酶C(PKC)信号通路。本文分别介绍了这三种信号通路与GLUT4转位的关系以及相关的促进GLUT4转位的药物,为寻找治疗2型糖尿病的潜在新药提供参考。  相似文献   

8.
动物脂肪和肌肉组织中葡萄糖的摄取是通过受胰岛素调控的GLUT4储存囊泡的运输实现的.Sec1p的同源物Munc18c被认为是通过控制SNARE复合物的装配来使GLUT4囊泡锚定到质膜上的重要物质.我们发现Munc18c的缺失没有影响GLUT4的转运上膜,也没有影响Syntaxin4在细胞膜上的定位.在缺少Munc18c和功能性Syntaxin2的时候,GLUT4的转运可能和Munc18b有关.在3T3-L1脂肪细胞中与Syntaxin4具有强烈相互作用的是Munc18c而不是Munc18a和Munc18b.然而,当缺少Munc18c时,Munc18a和Munc18b与Syntaxin4体现出较弱的相互作用.因此,Syntaxin4可能在胰岛素刺激GLUT4转运过程中起到重要的作用,且与SM蛋白的相互作用是有代偿性的.  相似文献   

9.
猪是研究糖尿病最理想的模型动物, 研究胰岛素和胰岛素抵抗是研究糖尿病的重要环节。为明确SOCS-3在胰岛素抵抗中的作用, 分别用100 nmol/L的胰岛素, 300 nmol/L的地塞米松处理原代培养的猪脂肪细胞诱导胰岛素抵抗; 利用半定量RT-PCR技术分别检测SOCS-3、OB、GLUT4和PPARg 基因表达变化。结果发现, 胰岛素增加了GLUT4、SOCS-3和PPARg 基因的表达, 对OB基因表达变化没有显著性影响; 地塞米松诱导的胰岛素抵抗状态下OB和SOCS-3基因表达水平升高, 而GLUT4和PPARγ基因表达水平显著下调。研究结果表明, GLUT4基因表达量水平的升高可能是由于PPARg的高表达引起, SOCS-3基因的不同表达水平对胰岛素信号的抑制效果不同。地塞米松诱导的胰岛素抵抗不仅表现在对葡萄糖转运的抑制, 也反映在抑制了胰岛素信号; 而SOCS-3基因可能是消除胰岛素抵抗的一个有效靶基因。  相似文献   

10.
动物脂肪和肌肉组织中葡萄糖的摄取是通过受胰岛素调控的GLUT4储存囊泡的运输实现的.Sec1p的同源物Munc18c被认为是通过控制SNARE复合物的装配来使GLUT4囊泡锚定到质膜上的重要物质.我们发现Munc18c的缺失没有影响GLUT4的转运上膜,也没有影响Syntaxin4在细胞膜上的定位.在缺少Munc18c和功能性Syntaxin2的时候,GLUT4的转运可能和Munc18b有关.在3T3-L1脂肪细胞中与Syntaxin4具有强烈相互作用的是Munc18c而不是Munc18a和Munc18b.然而,当缺少Munc18c时,Munc18a和Munc18b与Syntaxin4体现出较弱的相互作用.因此,Syntaxin4可能在胰岛素刺激GLUT4转运过程中起到重要的作用,且与SM蛋白的相互作用是有代偿性的.  相似文献   

11.
Glucose is the main source of energy for the body, requiring constant regulation of its blood concentration. Insulin release by the pancreas induces glucose uptake by insulin-sensitive tissues, most notably the brain, skeletal muscle, and adipocytes. Patients suffering from type-2 diabetes and/or obesity often develop insulin resistance and are unable to control their glucose homeostasis. New insights into the mechanisms of insulin resistance may provide new treatment strategies for type-2 diabetes.The GLUT family of glucose transporters consists of thirteen members distributed on different tissues throughout the body1. Glucose transporter type 4 (GLUT4) is the major transporter that mediates glucose uptake by insulin sensitive tissues, such as the skeletal muscle. Upon binding of insulin to its receptor, vesicles containing GLUT4 translocate from the cytoplasm to the plasma membrane, inducing glucose uptake. Reduced GLUT4 translocation is one of the causes of insulin resistance in type-2 diabetes2,3.The translocation of GLUT4 from the cytoplasm to the plasma membrane can be visualized by immunocytochemistry, using fluorophore-conjugated GLUT4-specific antibodies.Here, we describe a technique to quantify total amounts of GLUT4 translocation to the plasma membrane of cells during a chosen duration, using flow cytometry. This protocol is rapid (less than 4 hours, including incubation with insulin) and allows the analysis of as few as 3,000 cells or as many as 1 million cells per condition in a single experiment. It relies on anti-GLUT4 antibodies directed to an external epitope of the transporter that bind to it as soon as it is exposed to the extracellular medium after translocation to the plasma membrane.  相似文献   

12.
Ins (endocytosis) and outs (exocytosis) of GLUT4 trafficking   总被引:3,自引:0,他引:3  
Glucose transporter 4 (GLUT4) is the major insulin-regulated glucose transporter expressed mainly in muscle and adipose tissue. GLUT4 is stored in a poorly characterized intracellular vesicular compartment and translocates to the cell surface in response to insulin stimulation resulting in an increased glucose uptake. This process is essential for the maintenance of normal glucose homeostasis and involves a complex interplay of trafficking events and intracellular signaling cascades. Recent studies have identified sortilin as an essential element for the formation of GLUT4 storage vesicles during adipogenesis and Golgi-localized gamma-ear-containing Arf-binding protein (GGA) as a key coat adaptor for the entry of newly synthesized GLUT4 into the specialized compartment. Insulin-stimulated GLUT4 translocation from this compartment to the plasma membrane appears to require the Akt/protein kinase B substrate termed AS160 (Akt substrate of 160kDa). In addition, the VPS9 domain-containing protein Gapex-5 in complex with CIP4 appears to function as a Rab31 guanylnucleotide exchange factor that is necessary for insulin-stimulated GLUT4 translocation. Here, we attempt to summarize recent advances in GLUT4 vesicle biogenesis, intracellular trafficking and membrane fusion.  相似文献   

13.
Insulin stimulated GLUT4 (glucose transporter 4) translocation and glucose uptake in muscles and adipocytes is important for the maintenance of blood glucose homeostasis in our body. In this paper, we report the identification of kaempferitrin (kaempferol 3,7-dirhamnoside), a glycosylated flavonoid, as a compound that inhibits insulin stimulated GLUT4 translocation and glucose uptake in 3T3-L1 adipocytes. In the absence of insulin, we observed that addition of kaempferitrin did not affect GLUT4 translocation or glucose uptake. On the other hand, kaempferitrin acted as an inhibitor of insulin-stimulated GLUT4 translocation and glucose uptake in 3T3-L1 adipocytes by inhibiting Akt activation. Molecular docking studies using a homology model of GLUT4 showed that kaempferitrin binds directly to GLUT4 at the glucose transportation channel, suggesting the possibility of a competition between kaempferitrin and glucose during the transport. Taken together, our data demonstrates that kaempferitrin inhibits GLUT4 mediated glucose uptake at least by two different mechanisms, one by interfering with the insulin signaling pathway and the other by a possible competition with glucose during the transport.  相似文献   

14.
The facilitative glucose transporter GLUT4 plays a key role in regulating whole body glucose homeostasis. GLUT4 dramatically changes its distribution upon insulin stimulation, and insulin-resistant diabetes is often linked with compromised translocation of GLUT4 under insulin stimulation. To elucidate the functional significance of the sole N-glycan chain on GLUT4, wild-type GLUT4 and a GLUT4 glycosylation mutant conjugated with enhanced GFP were stably expressed in HeLa cells. The N-glycan contributed to the overall stability of newly synthesized GLUT4. Moreover, cell surface expression of wild-type GLUT4 in HeLa cells was elevated upon insulin treatment, whereas the glycosylation mutant lost the ability to respond to insulin. Subcellular distribution of the mutant was distinct from that of wild-type GLUT4, implying that the subcellular localization required for insulin-mediated translocation was impaired in the mutant protein. Interestingly, kifunensine-treated cells also lost sensitivity to insulin, suggesting the functional importance of the N-glycan structure for GLUT4 trafficking. The K(m) or turnover rates of wild-type and mutant GLUT4, however, were similar, suggesting that the N-glycan had little effect on transporter activity. These findings underscore the critical roles of the N-glycan chain in quality control as well as intracellular trafficking of GLUT4.  相似文献   

15.
Exploring the whereabouts of GLUT4 in skeletal muscle (review)   总被引:4,自引:0,他引:4  
The glucose transporter GLUT4 is expressed in muscle, fat cells, brain and kidney. In contrast to other glucose transporters, GLUT4 in unstimulated cells is mostly intracellular. Stimuli such as insulin and muscle contractions then cause the translocation of GLUT4 to the cell surface. Questions related to GLUT4 storage compartments, trafficking to the surface membrane, and nature of the intracellular pools, have kept many groups busy for the past 20 years. Yet, one of the main questions in the field remains the universality of GLUT4 features. Can one extrapolate work done on fat cells to muscle or brain? Or vice-versa? Can one use cultures to predict GLUT4 behaviour in fully differentiated tissues? This review summarizes the authors' knowledge of GLUT4 biology in skeletal muscle, which is the predominant tissue for glucose homeostasis. The results are compared to those obtained with the fat cell system, and an attempt is made to assess the universality principle.  相似文献   

16.
Zhang Y  Zhang H  Yao XG  Shen H  Chen J  Li C  Chen L  Zheng M  Ye J  Hu L  Shen X  Jiang H 《PloS one》2012,7(2):e31811
Glucose transporter 4 (GLUT4) is a principal glucose transporter in response to insulin, and impaired translocation or decreased expression of GLUT4 is believed to be one of the major pathological features of type 2 diabetes mellitus (T2DM). Therefore, induction of GLUT4 translocation or/and expression is a promising strategy for anti-T2DM drug discovery. Here we report that the natural product (+)-Rutamarin (Rut) functions as an efficient dual inducer on both insulin-induced GLUT4 translocation and expression. Rut-treated 3T3-L1 adipocytes exhibit efficiently enhanced insulin-induced glucose uptake, while diet-induced obese (DIO) mice based assays further confirm the Rut-induced improvement of glucose homeostasis and insulin sensitivity in vivo. Subsequent investigation of Rut acting targets indicates that as a specific protein tyrosine phosphatase 1B (PTP1B) inhibitor Rut induces basal GLUT4 translocation to some extent and largely enhances insulin-induced GLUT4 translocation through PI3 kinase-AKT/PKB pathway, while as an agonist of retinoid X receptor α (RXRα), Rut potently increases GLUT4 expression. Furthermore, by using molecular modeling and crystallographic approaches, the possible binding modes of Rut to these two targets have been also determined at atomic levels. All our results have thus highlighted the potential of Rut as both a valuable lead compound for anti-T2DM drug discovery and a promising chemical probe for GLUT4 associated pathways exploration.  相似文献   

17.
Background information. Insulin‐stimulated glucose uptake into skeletal muscle is crucial for glucose homoeostasis, and depends on the recruitment of GLUT4 (glucose transporter 4) to the plasma membrane. Mechanisms underlying insulin‐dependent GLUT4 translocation, particularly the role of Rho family GTPases, remain controversial. Results. In the present study, we show that constitutively active Rac1, but not other Rho family GTPases tested, induced GLUT4 translocation in the absence of insulin, suggesting that Rac1 activation is sufficient for GLUT4 translocation in muscle cells. Rac1 activation occurred in dorsal membrane ruffles of insulin‐stimulated cells as revealed by a novel method to visualize activated Rac1 in situ. We further identified FLJ00068 as a GEF (guanine‐nucleotide‐exchange factor) responsible for this Rac1 activation. Indeed, constitutively active FLJ00068 caused Rac1 activation in dorsal membrane ruffles and GLUT4 translocation without insulin stimulation. Down‐regulation of Rac1 or FLJ00068 by RNA interference, on the other hand, abrogated insulin‐induced GLUT4 translocation. Basal, but not insulin‐stimulated, activity of the serine/threonine kinase Akt was required for the induction of GLUT4 translocation by constitutively active Rac1 or FLJ00068. Conclusion. Collectively, Rac1 activation specifically in membrane ruffles by the GEF FLJ00068 is sufficient for insulin induction of glucose uptake into skeletal‐muscle cells.  相似文献   

18.
Glucose is used aerobically and anaerobically to generate energy for cells. Glucose transporters (GLUTs) are transmembrane proteins that transport glucose across the cell membrane. Insulin promotes glucose utilization in part through promoting glucose entry into the skeletal and adipose tissues. This has been thought to be achieved through insulin-induced GLUT4 translocation from intracellular compartments to the cell membrane, which increases the overall rate of glucose flux into a cell. The insulin-induced GLUT4 translocation has been investigated extensively. Recently, significant progress has been made in our understanding of GLUT4 expression and translocation. Here, we summarized the methods and reagents used to determine the expression levels of Slc2a4 mRNA and GLUT4 protein, and GLUT4 translocation in the skeletal muscle, adipose tissues, heart and brain. Overall, a variety of methods such real-time polymerase chain reaction, immunohistochemistry, fluorescence microscopy, fusion proteins, stable cell line and transgenic animals have been used to answer particular questions related to GLUT4 system and insulin action. It seems that insulin-induced GLUT4 translocation can be observed in the heart and brain in addition to the skeletal muscle and adipocytes. Hormones other than insulin can induce GLUT4 translocation. Clearly, more studies of GLUT4 are warranted in the future to advance of our understanding of glucose homeostasis.  相似文献   

19.
Insulin stimulates glucose uptake into skeletal muscle tissue mainly through the translocation of glucose transporter 4 (GLUT4) to the plasma membrane. The precise mechanism involved in this process is presently unknown. In the cascade of events leading to insulin-induced glucose transport, insulin activates specific protein kinase C (PKC) isoforms. In this study we investigated the roles of PKC zeta in insulin-stimulated glucose uptake and GLUT4 translocation in primary cultures of rat skeletal muscle. We found that insulin initially caused PKC zeta to associate specifically with the GLUT4 compartments and that PKC zeta together with the GLUT4 compartments were then translocated to the plasma membrane as a complex. PKC zeta and GLUT4 recycled independently of one another. To further establish the importance of PKC zeta in glucose transport, we used adenovirus constructs containing wild-type or kinase-inactive, dominant-negative PKC zeta (DNPKC zeta) cDNA to overexpress this isoform in skeletal muscle myotube cultures. We found that overexpression of PKC zeta was associated with a marked increase in the activity of this isoform. The overexpressed, active PKC zeta coprecipitated with the GLUT4 compartments. Moreover, overexpression of PKC zeta caused GLUT4 translocation to the plasma membrane and increased glucose uptake in the absence of insulin. Finally, either insulin or overexpression of PKC zeta induced serine phosphorylation of the GLUT4-compartment-associated vesicle-associated membrane protein 2. Furthermore, DNPKC zeta disrupted the GLUT4 compartment integrity and abrogated insulin-induced GLUT4 translocation and glucose uptake. These results demonstrate that PKC zeta regulates insulin-stimulated GLUT4 translocation and glucose transport through the unique colocalization of this isoform with the GLUT4 compartments.  相似文献   

20.
The skeletal muscle cells are one of the main sites of glucose uptake through glucose transporter 4 (GLUT4) in response to insulin. In muscle cells, 5' adenosine monophosphate-activated protein kinase (AMPK) is known as another GLUT4 translocation promoter. Natural compounds that activate AMPK have a possibility to overcome insulin resistance in the diabetic state. Piceatannol is a natural analog and a metabolite of resveratrol, a known AMPK activator. In this study, we investigate the in vitro effect of piceatannol on glucose uptake, AMPK phosphorylation and GLUT4 translocation to plasma membrane in L6 myocytes, and its in vivo effect on blood glucose levels in type 2 diabetic model db/db mice. Piceatannol was found to promote glucose uptake, AMPK phosphorylation and GLUT4 translocation by Western blotting analyses in L6 myotubes under a condition of insulin absence. Promotion by piceatannol of glucose uptake as well as GLUT4 translocation to plasma membrane by immunocytochemistry was also demonstrated in L6 myoblasts transfected with a glut4 cDNA-coding vector. Piceatannol suppressed the rises in blood glucose levels at early stages and improved the impaired glucose tolerance at late stages in db/db mice. These in vitro and in vivo findings suggest that piceatannol may be preventive and remedial for type 2 diabetes and become an antidiabetic phytochemical.  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-23

京公网安备 11010802026262号