首页 | 本学科首页   官方微博 | 高级检索  
相似文献
 共查询到20条相似文献,搜索用时 109 毫秒
1.
哺乳动物雷帕霉素靶(mTOR)和蛋白激酶B(Akt/PKB)与肿瘤发生的密切关系已被广泛地认可.mTOR是一种丝/苏氨酸激酶,可以通过影响mRNA转录、代谢、自噬等方式调控细胞的生长.它既是PI3K的效应分子,也可以是PI3K的反馈调控因子.mTORC1 和mTORC2是mTOR的两种不同复合物. 对雷帕霉素敏感的mTORC1受到营养、生长因子、能量和应激4种因素的影响.生长因子通过PI3K/Akt信号通路调控mTORC1是最具特征性调节路径.而mTORC2最为人熟知的是作为Akt473磷酸化位点的上游激酶. 同样,Akt/PKB在细胞增殖分化、迁移生长过程中发挥着重要作用. 随着Thr308和Ser473两个位点激活,Akt/PKB也得以全面活化.因此,mTORC2-Akt-mTORC1的信号通路在肿瘤形成和生长中是可以存在的.目前临床肿瘤治疗中,PI3K/Akt/mTOR是重要的靶向治疗信号通路.然而,仅抑制mTORC1活性,不是所有的肿瘤都能得到预期控制.雷帕霉素虽然能抑制mTORC1,但也能反馈性地增加PI3K信号活跃度,从而影响治疗预后.近来发现的第二代抑制剂可以同时抑制mTORC1/2和PI3K活性,这种抑制剂被认为在肿瘤治疗上颇具前景.本综述着重阐述了PI3K/Akt/mTOR信号通路的传导、各因子之间的相互调控以及相关抑制剂的发展.  相似文献   

2.
脂肪组织是一种主要的能量储存和内分泌器官。脂肪生成是一系列复杂的细胞分化过程,受到细胞营养水平、激素和代谢物等调节。哺乳动物雷帕霉素靶蛋白(mammalian target of rapamycin, mTOR)复合物包括哺乳动物雷帕霉素靶蛋白复合体1(mammalian target of rapamycin complex 1,mTORC1)和mTORC2两种蛋白质复合体。mTOR复合物含有的脂质激酶样域奠定了mTOR通路调控脂肪生成的基础。对mTORC1和mTORC2的部分组成蛋白质研究也验证了mTOR调控成脂的功能。基于前期的研究,我们综述了miR-199a-3p、miR-103、miR-188、68 kD有丝分裂中的Src相关底物(Src-associated substrate in mitosis of 68 kD,Sam68)、内皮抑素等物质通过mTORC1和mTORC2蛋白质复合体调控脂肪生成的机制。同时,进一步构建了包括胰岛素/IGF通路、PI3K-AKT通路、氨基酸通路、AMPK通路、cAMP通路、cGMP通路、NOTCH通路以及影响上述通路的bta-miR-150、4-O-甲基蔗糖和多种蛋白质在内的mTOR信号通路调控脂肪生成的网络。本文主要综述了mTOR复合物的特性和mTOR通路调控脂肪生成方面的最新研究进展,指出mTORC2具有调控脂质摄取和脂质分解的作用,调控mTORC1功能的作用,但是有关mTORC2的研究相对mTORC1较少,因此,对脂肪生成和脂质代谢的进一步研究需要集中于mTORC2。  相似文献   

3.
近几年来,关于哺乳动物雷帕霉素靶(mammalian target of rapamycin,mTOR)在各种哺乳动物细胞中调节肌动蛋白微丝极化及肌球蛋白微丝网形成的研究一直在不断地取得新的进展。尽管到目前为止,包括mTORC2上游和下游在内的相关的调控路径还未明确,但是因为mTORC6,的物学多样性,使其成为了当今生物学研究的焦点之一。基于长久以来特别是近五年对mTORC2的研究,在涉及细胞运动迁移、增殖分化、蛋白质合成、凋亡及自噬等生物学功能的研究中,一些重要的下游相关调控分子和蛋白相继被发现,比如P—Rexl/2、Rho家族GTPases、PKC、cAMP、p27kip1等。该综述着重总结了mTORC2实现这些生物学功能所可能通过的四条路径。当然,仍然需要大量的实验数据和研究证据进一步地证实和完善这些已经发现的可能存在的路径。  相似文献   

4.
哺乳动物雷帕霉素靶蛋白mTOR是一个进化上十分保守的蛋白激酶,属于PIKK超家族。在细胞内mTOR存在两种功能不同的复合体mTORC1和mTORC2。mTOR主要通过接受上游信号分子Rheb、TSC1/TSC2的调控来整合细胞内外信号,其下游效应器是4E-BP和p70S6K,通过影响特定mRNA的翻译调节细胞的生长和增殖。在神经系统方面,神经元的发育、突触可塑性的调节、学习和记忆的形成都依赖于适当的mTOR通路的活化。新近的研究显示,神经退行性疾病阿尔茨海默病患者表现mTOR通路的异常,在双转基因鼠中,APP和PS1表达与mTOR/P70S6K下调关联,并影响精神状态评分。mTOR信号通路生理功能和调节机制的研究对了解AD的发病机理和寻找药物靶点具有重要意义。  相似文献   

5.
mTOR信号通路与癌症治疗   总被引:1,自引:0,他引:1  
陈樑  张红锋 《生命的化学》2005,25(2):127-129
哺乳动物雷帕霉素靶蛋白(mammalian target of rapamycin,mTOR)是一种非典型的丝氨酸/苏氨酸蛋白激酶,在细胞的生长、分化、增殖、迁移和存活上扮演了重要的角色。由于mTOR信号转导通路在细胞周期进程中发挥了重要作用,而细胞周期进程调节异常与许多疾病尤其是癌症的发生、发展有关,因此mTOR信号通路的失调可引起多种癌症。mTOR的特异性抑制剂雷帕霉素及其衍生物CCI-779能抑制mTOR的功能,使细胞阻滞在G。期,并引起凋亡。CCI-779作为抗癌药物已分别进入Ⅱ期临床。通过临床实验CCI-779显示出较高的抗癌活性和相对较小的副作用。越来越多的实验证据显示,mTOR信号转导通路的抑制剂可开发成为潜在的肿瘤特异性治疗药物。  相似文献   

6.
mTOR是细胞生长和增殖的中枢调控因子。mTOR形成2个不同的复合物mTORC1和mTORC2。mTORC1受多种信号调节,如生长因子、氨基酸和细胞能量,同时,mTORC1调节许多重要的细胞过程,包括翻译、转录和自噬。AMPK作为一种关键的生理能量传感器,是细胞和有机体能量平衡的主要调节因子,协调多种代谢途径,平衡能量的供应和需求,最终调节细胞和器官的生长。能量代谢平衡调控是由多个与之相关的信号通路所介导,其中AMPK/mTOR信号通路在细胞内共同构成一个合成代谢和分解代谢过程的开关。此外,AMPK/mTOR信号通路还是一个自噬的重要调控途径。本文着重于目前对AMPK和mTOR信号传导之间关系的了解,讨论了AMPK/mTOR在细胞和有机体能量稳态中的作用。  相似文献   

7.
哺乳动物雷帕霉素靶蛋白(mammalian target of rapamycin, mTOR)是一种特异性的蛋白激酶,在调控细胞生长、增殖、代谢等多项生命活动中都具有重要意义。mTOR调控功能的失活与异常激活,会导致相关肿瘤和疾病的发生。近年来已有多种mTOR抑制剂用于治疗该信号转导通路异常引起的肿瘤。该文探究多种调控mTOR的信号通路和mTOR抑制剂用于肿瘤治疗的最新进展,还探讨肿瘤细胞对mTOR抑制剂产生耐药性的潜在机制和应对策略。因此,对mTOR信号通路及其调控机制的探索有助于研发全新的肿瘤治疗技术。  相似文献   

8.
正哺乳动物雷帕霉素靶蛋白(mTOR)是存在于哺乳动物体内的一种高度保守的丝氨酸/苏氨酸蛋白激酶,能够调节细胞内多种物质的代谢。它参与组成哺乳动物雷帕霉素靶蛋白复合体1(mTORC1)和哺乳动物雷帕霉素靶蛋白复合体2(mTORC2)2种复合体。在2015年发现的人类mT ORC1结构基础上,Saxton等人揭示了亮氨酸对于mT ORC1通路复杂的调控机制。他  相似文献   

9.
哺乳动物雷帕霉素靶蛋白(mTOR)是一种丝氨酸/苏氨酸激酶,可集合胞内和胞外信号调节细胞生长、增殖、代谢和存活。在肿瘤的发生发展过程中,mTOR信号多处于异常激活状态,已被确证为肿瘤药物治疗的重要靶标。因雷帕霉素临床应用存在局限性,近期ATP竞争性mTOR抑制剂的研发十分活跃。本文综述了ATP竞争性mTOR抑制剂的研究进展,重点讨论药物的作用特点。  相似文献   

10.
哺乳动物雷帕霉素靶蛋白(mammalian target of rapamycin,mTOR)是一种Ser/Thr激酶,属于PIKK超家族,对调节细胞周期、蛋白质合成等具有重要作用,是细胞生长、增殖、分化、凋亡的中心调控器,但在哺乳动物卵母细胞中的研究还未见报道.以小鼠卵母细胞为研究对象,采用免疫荧光为主要研究方法,对mTOR在小鼠卵母细胞中的表达进行研究,并通过mTOR的特异性抑制剂雷帕霉素( rapamycin,RAPA )对卵母细胞进行处理,对mTOR在卵母细胞成熟过程中的作用进行研究.结果显示:小鼠卵母细胞成熟过程中,生发泡( germinal vesicle,GV )期mTOR主要集中在核膜处表达,生发泡破裂 ( germinal vesicle breakdown,GVBD )后mTOR伴随染色体分布,第二次减数分裂中期( second metaphase,MⅡ期 ) mTOR伴随纺锤体分布;雷帕霉素处理后,小鼠卵母细胞的成熟受到抑制,且这种抑制作用具有浓度依赖性,同时其mTOR的表达部位和形态也发生变化.研究表明,在小鼠卵母细胞成熟过程中,mTOR在各个时期的表达及分布具有阶段特异性,并对小鼠卵母细胞GVBD的发生和第一极体的排放都具有重要作用.  相似文献   

11.
cAMP and mTOR signalling pathways control a number of critical cellular processes including metabolism, protein synthesis, proliferation and cell survival and therefore understanding the signalling events which integrate these two signalling pathways is of particular interest. In this study, we show that the pharmacological elevation of [cAMP]i in mouse embryonic fibroblasts (MEFs) and human embryonic kidney 293 (HEK293) cells inhibits mTORC1 activation via a PKA-dependent mechanism. Although the inhibitory effect of cAMP on mTOR could be mediated by impinging on signalling cascades (i.e. PKB, MAPK and AMPK) that inhibit TSC1/2, an upstream negative regulator of mTORC1, we show that cAMP inhibits mTORC1 in TSC2 knockout (TSC2−/−) MEFs. We also show that cAMP inhibits insulin and amino acid-stimulated mTORC1 activation independently of Rheb, Rag GTPases, TSC2, PKB, MAPK and AMPK, indicating that cAMP may act independently of known regulatory inputs into mTOR. Moreover, we show that the prolonged elevation in [cAMP]i can also inhibit mTORC2. We provide evidence that this cAMP-dependent inhibition of mTORC1/2 is caused by the dissociation of mTORC1 and 2 and a reduction in mTOR catalytic activity, as determined by its auto-phosphorylation on Ser2481. Taken together, these results provide an important insight into how cAMP signals to mTOR and down-regulates its activity, which may lead to the identification of novel drug targets to inhibit mTOR that could be used for the treatment and prevention of human diseases such as cancer.  相似文献   

12.
13.

Background

mTOR, which can form mTOR Complex 1 (mTORC1) or mTOR Complex 2 (mTORC2) depending on its binding partners, is frequently deregulated in the pulmonary neoplastic conditions and interstitial lung diseases of the patients treated with rapalogs. In this study, we investigated the relationship between mTOR signaling and epithelial mesenchymal transition (EMT) by dissecting mTOR pathways.

Methods

Components of mTOR signaling pathway were silenced by shRNA in a panel of non-small cell lung cancer cell lines and protein expression of epithelial and mesenchymal markers were evaluated by immunoblotting and immunocytochemistry. mRNA level of the E-cadherin repressor complexes were evaluated by qRT-PCR.

Results

IGF-1 treatment decreased expression of the E-cadherin and rapamycin increased its expression, suggesting hyperactivation of mTOR signaling relates to the loss of E-cadherin. Genetic ablation of rapamycin-insensitive companion of mTOR (Rictor), a component of mTORC2, did not influence E-cadherin expression, whereas genetic ablation of regulatory-associated protein of mTOR (Raptor), a component of mTORC1, led to a decrease in E-cadherin expression at the mRNA level. Increased phosphorylation of AKT at Ser473 and GSK-3β at Ser9 were observed in the Raptor-silenced NSCLC cells. Of the E-cadherin repressor complexes tested, Snail, Zeb2, and Twist1 mRNAs were elevated in raptor-silenced A549 cells, and Zeb2 and Twist1 mRNAs were elevated in Raptor-silenced H2009 cells. These findings were recapitulated by treatment with the GSK-3β inhibitor, LiCl. Raptor knockdown A549 cells showed increased expression of N-cadherin and vimentin with mesenchymal phenotypic changes.

Conclusions

In conclusion, selective inhibition of mTORC1 leads to hyperactivation of the AKT/GSK-3β pathway, inducing E-cadherin repressor complexes and EMT. These findings imply the existence of a feedback inhibition loop of mTORC1 onto mTORC2 that plays a role in the homeostasis of E-cadherin expression and EMT, requiring caution in the clinical use of rapalog and selective mTORC1 inhibitors.  相似文献   

14.
The mammalian target of rapamycin (mTOR) is centrally involved in growth, survival and metabolism. In cancer, mTOR is frequently hyperactivated and is a clinically validated target for drug development. Until recently, we have relied largely on the use of rapamycin to study mTOR function and its anticancer potential. Recent insights now indicate that rapamycin is a partial inhibitor of mTOR through allosteric inhibition of mTOR complex-1 (mTORC1) but not mTOR complex-2 (mTORC2). Both the mechanism of action and the cellular response to mTORC1 inhibition by rapamycin and related drugs may limit the effectiveness of these compounds as antitumor agents. We and others have recently reported the discovery of second-generation ATP-competitive mTOR kinase inhibitors (TKIs) that bind to the active sites of mTORC1 and mTORC2, thereby targeting mTOR signaling function globally (see refs. 1-4). The discovery of specific, active-site mTOR inhibitors has opened a new chapter in the 40-plus year old odyssey that began with the discovery of rapamycin from a soil sample collected on Easter Island (see Vézina C, et al. J Antibiot 1975). Here, we discuss recent studies that highlight the emergence of rapamycin-resistant mTOR function in protein synthesis, cell growth, survival and metabolism. It is shown that these rapamycin-resistant mTOR functions are profoundly inhibited by TKIs. A more complete suppression of mTOR global signaling network by the new inhibitors is expected to yield a deeper and broader antitumor response in the clinic.  相似文献   

15.
Traumatic brain injury (TBI) provokes primary and secondary damage on endothelium and brain parenchyma, leading neurons die rapidly by necrosis. The mammalian target of rapamycin signalling pathway (mTOR) manages numerous aspects of cellular growth, and it is up-regulated after moderate to severe traumatic brain injury (TBI). Currently, the significance of this increased signalling event for the recovery of brain function is unclear; therefore, we used two different selective inhibitors of mTOR activity to discover the functional role of mTOR inhibition in a mouse model of TBI performed by a controlled cortical impact injury (CCI). Treatment with KU0063794, a dual mTORC1 and mTORC2 inhibitor, and with rapamycin as well-known inhibitor of mTOR, was performed 1 and 4 hours subsequent to TBI. Results proved that mTOR inhibitors, especially KU0063794, significantly improved cognitive and motor recovery after TBI, reducing lesion volumes. Also, treatment with mTOR inhibitors ameliorated the neuroinflammation associated with TBI, showing a diminished neuronal death and astrogliosis after trauma. Our findings propose that the involvement of selective mTORC1/2 inhibitor may represent a therapeutic strategy to improve recovery after brain trauma.  相似文献   

16.
17.
The mechanistic target of rapamycin (mTOR) plays a central role in cellular growth and metabolism. mTOR forms two distinct protein complexes, mTORC1 and mTORC2. Much is known about the regulation and functions of mTORC1 due to availability of a natural compound, rapamycin, that inhibits this complex. Studies that define mTORC2 cellular functions and signaling have lagged behind. The development of pharmacological inhibitors that block mTOR kinase activity, and thereby inhibit both mTOR complexes, along with availability of mice with genetic knockouts in mTOR complex components have now provided new insights on mTORC2 function and regulation. Since prolonged effects of rapamycin can also disrupt mTORC2, it is worth re-evaluating the contribution of this less-studied mTOR complex in cancer, metabolic disorders and aging. In this review, we focus on recent developments on mammalian mTORC2 signaling mechanisms and its cellular and tissue-specific functions.  相似文献   

18.
New potent mTORC1/mTORC2 dual inhibitors, 5,7-dihydro-6H-pyrrolo[2,3-d]pyrimidin-6-one derivatives, were obtained by optimizing functional groups on our previously reported PI3Kα inhibitor. All the target compounds were synthesized and structural optimization on the structure of the lead compound based on cytotoxic activity. The results showed that some of the target compounds exhibited moderate to high cytotoxic activity against cell line U87MG and PC-3. The activities against mTOR kinase were investigated and the compound 12q showed excellent activity with an IC50 value of 54 nM in the same level of the positive control BEZ235 with IC50 value of 55 nM under the same test conditions. The western blot and cell cycle results demonstrate that compound 12q is a candidate as an mTORC1/mTORC2 dual-target inhibitor. The theoretical calculations were also performed to better understanding the binding modes of the compound 12q in the mTOR active site.  相似文献   

19.
The development of drug-resistance by neoplastic cells is recognized as a major cause of targeted therapy failure and disease progression. The mechanistic (previously mammalian) target of rapamycin (mTOR) is a highly conserved Ser/Thr kinase that acts as the catalytic subunit of two structurally and functionally distinct large multiprotein complexes, referred to as mTOR complex 1 (mTORC1) and mTORC2. Both mTORC1 and mTORC2 play key roles in a variety of healthy cell types/tissues by regulating physiological anabolic and catabolic processes in response to external cues. However, a body of evidence identified aberrant activation of mTOR signaling as a common event in many human tumors. Therefore, mTOR is an attractive target for therapeutic targeting in cancer and this fact has driven the development of numerous mTOR inhibitors, several of which have progressed to clinical trials. Nevertheless, mTOR inhibitors have met with a very limited success as anticancer therapeutics. Among other reasons, this failure was initially ascribed to the activation of several compensatory signaling pathways that dampen the efficacy of mTOR inhibitors. The discovery of these regulatory feedback mechanisms greatly contributed to a better understanding of cancer cell resistance to mTOR targeting agents. However, over the last few years, other mechanisms of resistance have emerged, including epigenetic alterations, compensatory metabolism rewiring and the occurrence of mTOR mutations. In this article, we provide the reader with an updated overview of the mechanisms that could explain resistance of cancer cells to the various classes of mTOR inhibitors.  相似文献   

20.
TBK1 responds to microbes to initiate cellular responses critical for host innate immune defense. We found previously that TBK1 phosphorylates mTOR (mechanistic target of rapamycin) on S2159 to increase mTOR complex 1 (mTORC1) signaling in response to the growth factor EGF and the viral dsRNA mimetic poly(I:C). mTORC1 and the less well studied mTORC2 respond to diverse cues to control cellular metabolism, proliferation, and survival. Although TBK1 has been linked to Akt phosphorylation, a direct relationship between TBK1 and mTORC2, an Akt kinase, has not been described. By studying MEFs lacking TBK1, as well as MEFs, macrophages, and mice bearing an Mtor S2159A knock-in allele (MtorA/A) using in vitro kinase assays and cell-based approaches, we demonstrate here that TBK1 activates mTOR complex 2 (mTORC2) directly to increase Akt phosphorylation. We find that TBK1 and mTOR S2159 phosphorylation promotes mTOR-dependent phosphorylation of Akt in response to several growth factors and poly(I:C). Mechanistically, TBK1 coimmunoprecipitates with mTORC2 and phosphorylates mTOR S2159 within mTORC2 in cells. Kinase assays demonstrate that TBK1 and mTOR S2159 phosphorylation increase mTORC2 intrinsic catalytic activity. Growth factors failed to activate TBK1 or increase mTOR S2159 phosphorylation in MEFs. Thus, basal TBK1 activity cooperates with growth factors in parallel to increase mTORC2 (and mTORC1) signaling. Collectively, these results reveal cross talk between TBK1 and mTOR, key regulatory nodes within two major signaling networks. As TBK1 and mTOR contribute to tumorigenesis and metabolic disorders, these kinases may work together in a direct manner in a variety of physiological and pathological settings.  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