38 resultados para MTORC1


Relevância:

20.00% 20.00%

Publicador:

Resumo:

Purpose Cancer cells have been shown to be more susceptible to Ran knockdown than normal cells. We now investigate whether Ran is a potential therapeutic target of cancers with frequently found mutations that lead to higher Ras/MEK/ERK [mitogen-activated protein/extracellular signal-regulated kinase (ERK; MEK)] and phosphoinositide 3-kinase (PI3K)/Akt/mTORC1 activities. Experimental Design Apoptosis was measured by flow cytometry [propidium iodide (PI) and Annexin V staining] and MTT assay in cancer cells grown under different conditions after knockdown of Ran. The correlations between Ran expression and patient survival were examined in breast and lung cancers. Results Cancer cells with their PI3K/Akt/mTORC1 and Ras/MEK/ERK pathways inhibited are less susceptible to Ran silencing-induced apoptosis. K-Ras-mutated, c-Met-amplified, and Pten-deleted cancer cells are also more susceptible to Ran silencing-induced apoptosis than their wild-type counterparts and this effect is reduced by inhibitors of the PI3K/Akt/mTORC1 and MEK/ERK pathways. Overexpression of Ran in clinical specimens is significantly associated with poor patient outcome in both breast and lung cancers. This association is dramatically enhanced in cancers with increased c-Met or osteopontin expression, or with oncogenic mutations of K-Ras or PIK3CA, all of which are mutations that potentially correlate with activation of the PI3K/Akt/mTORC1 and/or Ras/MEK/ERK pathways. Silencing Ran also results in dysregulation of nucleocytoplasmic transport of transcription factors and downregulation of Mcl-1 expression, at the transcriptional level, which are reversed by inhibitors of the PI3K/Akt/mTORC1 and MEK/ERK pathways. Conclusion Ran is a potential therapeutic target for treatment of cancers with mutations/changes of expression in protooncogenes that lead to activation of the PI3K/Akt/mTORC1 and Ras/MEK/ERK pathways. ©2011 AACR.

Relevância:

20.00% 20.00%

Publicador:

Resumo:

Purpose: Cancer cells have been shown to be more susceptible to Ran knockdown than normal cells. We nowinvestigate whether Ran is a potential therapeutic target of cancers with frequently found mutations that lead to higher Ras/MEK/ERK [mitogen-activated protein/extracellular signal-regulated kinase (ERK; MEK)] and phosphoinositide 3-kinase (PI3K)/Akt/mTORC1 activities.

Relevância:

20.00% 20.00%

Publicador:

Resumo:

PRL-3, a metastasis-associated phosphatase, is known to exert its oncogenic functions through activation of PI3K/Akt, which is a key regulator of the rapamycin-sensitive mTOR complex 1 (mTORC1), but a coherent link between PRL-3 and activation of mTOR has not yet been formally demonstrated. We report a positive correlation between PRL-3 expression and mTOR phospho-activation in clinical tumour samples and mouse models of cancer and demonstrate that PRL-3 increased downstream signalling to the mTOR substrates, p70S6K and 4E-BP1, by increasing PI3K/Akt-mediated activation of Rheb-GTP via TSC2 suppression. We also show that PRL-3 increases mTOR translocation to lysosomes via increased mTOR binding affinity to Rag GTPases in an Akt-independent manner, demonstrating a previously undescribed mechanism of action for PRL-3. PRL-3 also enhanced matrix metalloproteinase-2 secretion and cellular invasiveness via activation of mTOR, attributes which were sensitive to rapamycin treatment. The downstream effects of PRL-3 were maintained even under conditions of environmental stress, suggesting that PRL-3 provides a strategic survival advantage to tumour cells via its effects on mTOR.

Relevância:

20.00% 20.00%

Publicador:

Resumo:

The mammalian target of rapamycin complex 1 (mTORC1) is a highly conserved protein complex regulating key pathways in cell growth. Hyperactivation of mTORC1 is implicated in numerous cancers, thus making it a potential broad-spectrum chemotherapeutic target. Here, we characterized how mTORC1 responds to cell death induced by various anticancer drugs such rapamycin, etoposide, cisplatin, curcumin, staurosporine and Fas ligand. All treatments induced cleavage in the mTORC1 component, raptor, resulting in decreased raptor-mTOR interaction and subsequent inhibition of the mTORC1-mediated phosphorylation of downstream substrates (S6K and 4E-BP1). The cleavage was primarily mediated by caspase-6 and occurred at two sites. Mutagenesis at one of these sites, conferred resistance to cell death, indicating that raptor cleavage is important in chemotherapeutic apoptosis.

Relevância:

20.00% 20.00%

Publicador:

Resumo:

Spinocerebellar ataxia type 1 (SCA1), due to the expansion of a polyglutamine repeat within the ubiquitously expressed Ataxin-1 protein, leads to the premature degeneration of Purkinje cells (PCs), the cause of which is poorly understood. Here, we identified the unique proteomic signature of Sca1(154Q/2Q) PCs at an early stage of disease, highlighting extensive alterations in proteins associated with synaptic functioning, maintenance, and transmission. Focusing on Homer-3, a PC-enriched scaffold protein regulating neuronal activity, revealed an early decline in its expression. Impaired climbing fiber-mediated synaptic transmission diminished mTORC1 signaling, paralleling Homer-3 reduction in Sca1(154Q/2Q) PCs. Ablating mTORC1 within PCs or pharmacological inhibition of mTORC1 identified Homer-3 as its downstream target. mTORC1 knockout in Sca1(154Q/2Q) PCs exacerbated and accelerated pathology. Reinstating Homer-3 expression in Sca1(154Q/2Q) PCs attenuated cellular dysfunctions and improved motor deficits. Our work reveals that impaired mTORC1-Homer-3 activity underlies PC susceptibility in SCA1 and presents a promising therapeutic target.

Relevância:

20.00% 20.00%

Publicador:

Resumo:

As Neoplasias Mieloproliferativas (NMPs) se caracterizam por apresentarem acúmulo de eritrócitos, leucócitos e plaquetas morfologicamente normais e seus precursores. Nos últimos anos vários estudos buscaram conhecer os mecanismos celulares e moleculares envolvidos na fisiopatologia e evolução dessas desordens, com o intuito de encontrar marcadores de diagnóstico, prognóstico e terapias eficazes. A mutação pontual no gene que codifica a enzima Janus Kinase 2 (JAK2 V617F), presente em aproximadamente 90% dos pacientes com PV e em 50% dos pacientes com TE e MF, foi o principal achado genético anormal associado a essas doenças. Essa mutação resulta na ativação constitutiva da enzima JAK2 e na desregulação da proliferação celular e resistência à apoptose. Nosso grupo de pesquisa descreveu em PV, TE e MF a expressão alterada de genes reguladores da apoptose e dados da literatura indicam que a desregulação do ciclo celular contribui para a fisiopatologia das NMPs. Nesse projeto o intuito foi investigar a associação da via de sinalização m-TOR com as alterações do ciclo celular e via JAK/STAT nas NMPs. A via de sinalização m-TOR participa dos processos celulares de sobrevivência e proliferação. A estratégia experimental foi avaliar a expressão de genes e proteínas, reguladores da via m-TOR, em leucócitos de pacientes com NPMC e linhagens celulares JAK2+ tratadas com inibidores de JAK2 e AKT. Para determinar a relação da via m-TOR nas NMPs foi escolhido o gene eIF4E, alterado nessas doenças, para observar sua modulação diante da inibição farmacológica nas linhagens celulares JAK2 positivas. Os resultados desse estudo contribuem para a descrição de novos alvos terapêuticos dependentes e indepentendes da atividade quinase JAK2 e para o melhor conhecimento da participação da via de sinalização m-TOR na fisiopatologia das NMPs.

Relevância:

10.00% 10.00%

Publicador:

Resumo:

Glutamine is conditionally essential in cancer cells, being utilized as a carbon and nitrogen source for macromolecule production, as well as for anaplerotic reactions fuelling the tricarboxylic acid (TCA) cycle. In this study, we demonstrated that the glutamine transporter ASCT2 (SLC1A5) is highly expressed in prostate cancer patient samples. Using LNCaP and PC-3 prostate cancer cell lines, we showed that chemical or shRNA-mediated inhibition of ASCT2 function in vitro decreases glutamine uptake, cell cycle progression through E2F transcription factors, mTORC1 pathway activation and cell growth. Chemical inhibition also reduces basal oxygen consumption and fatty acid synthesis, showing that downstream metabolic function is reliant on ASCT2-mediated glutamine uptake. Furthermore, shRNA knockdown of ASCT2 in PC-3 cell xenografts significantly inhibits tumour growth and metastasis in vivo, associated with the down-regulation of E2F cell cycle pathway proteins. In conclusion, ASCT2-mediated glutamine uptake is essential for multiple pathways regulating the cell cycle and cell growth, and is therefore a putative therapeutic target in prostate cancer.

Relevância:

10.00% 10.00%

Publicador:

Resumo:

The TSC2 gene, mutated in patients with tuberous sclerosis complex (TSC), encodes a 200 kDa protein TSC2 (tuberin). The importance of TSC2 in the regulation of cell growth and proliferation is irrefutable. TSC2 in complex with TSC1 negatively regulates the mTOR complex 1 (mTORC1) via RHEB in the PI3K-AKT-mTOR pathway and in turn regulates cell proliferation. It shows nuclear as well as cytoplasmic localization. However, its nuclear function remains elusive. In order to identify the nuclear function of TSC2, a whole-genome expression profiling of TSC2 overexpressing cells was performed, and the results showed differential regulation of 266 genes. Interestingly, transcription was found to be the most populated functional category. EREG (Epiregulin), a member of the epidermal growth factor family, was found to be the most downregulated gene in the microarray analysis. Previous reports have documented elevated levels of EREG in TSC lesions, making its regulatory aspects intriguing. Using the luciferase reporter, ChIP and EMSA techniques, we show that TSC2 binds to the EREG promoter between -352 bp and -303 bp and negatively regulates its expression. This is the first evidence for the role of TSC2 as a transcription factor and of TSC2 binding to the promoter of any gene.

Relevância:

10.00% 10.00%

Publicador:

Resumo:

Nitric oxide ( NO) has been shown to be effective in cancer chemoprevention and therefore drugs that help generate NO would be preferable for combination chemotherapy or solo use. This study shows a new evidence of NO as a mediator of acute leukemia cell death induced by fisetin, a promising chemotherapeutic agent. Fisetin was able to kill THP-1 cells in vivo resulting in tumor shrinkage in the mouse xenograft model. Death induction in vitro was mediated by an increase in NO resulting in double strand DNA breaks and the activation of both the extrinsic and the intrinsic apoptotic pathways. Double strand DNA breaks could be reduced if NO inhibitor was present during fisetin treatment. Fisetin also inhibited the downstream components of the mTORC1 pathway through downregulation of levels of p70 S6 kinase and inducing hypo-phosphorylation of S6 Ri P kinase, eIF4B and eEF2K. NO inhibition restored phosphorylation of downstream effectors of mTORC1 and rescued cells from death. Fisetin induced Ca2+ entry through L-type Ca2+ channels and abrogation of Ca2+ influx reduced caspase activation and cell death. NO increase and increased Ca2+ were independent phenomenon. It was inferred that apoptotic death of acute monocytic leukemia cells was induced by fisetin through increased generation of NO and elevated Ca2+ entry activating the caspase dependent apoptotic pathways. Therefore, manipulation of NO production could be viewed as a potential strategy to increase efficacy of chemotherapy in acute monocytic leukemia.

Relevância:

10.00% 10.00%

Publicador:

Resumo:

Lymphangioleiomyomatosis (LAM) is a rare lung-metastasizing neoplasm caused by the proliferation of smooth muscle-like cells that commonly carry loss-of-function mutations in either the tuberous sclerosis complex 1 or 2 (TSC1 or TSC2) genes. While allosteric inhibition of the mechanistic target of rapamycin (mTOR) has shown substantial clinical benefit, complementary therapies are required to improve response and/or to treat specific patients. However, there is a lack of LAM biomarkers that could potentially be used to monitor the disease and to develop other targeted therapies. We hypothesized that the mediators of cancer metastasis to lung, particularly in breast cancer, also play a relevant role in LAM. Analyses across independent breast cancer datasets revealed associations between low TSC1/2 expression, altered mTOR complex 1 (mTORC1) pathway signaling, and metastasis to lung. Subsequently, immunohistochemical analyses of 23 LAM lesions revealed positivity in all cases for the lung metastasis mediators fascin 1 (FSCN1) and inhibitor of DNA binding 1 (ID1). Moreover, assessment of breast cancer stem or luminal progenitor cell biomarkers showed positivity in most LAM tissue for the aldehyde dehydrogenase 1 (ALDH1), integrin-beta 3 (ITGB3/CD61), and/or the sex-determining region Y-box 9 (SOX9) proteins. The immunohistochemical analyses also provided evidence of heterogeneity between and within LAM cases. The analysis of Tsc2-deficient cells revealed relative over-expression of FSCN1 and ID1; however, Tsc2-deficient cells did not show higher sensitivity to ID1-based cancer inhibitors. Collectively, the results of this study reveal novel LAM biomarkers linked to breast cancer metastasis to lung and to cell stemness, which in turn might guide the assessment of additional or complementary therapeutic opportunities for LAM.

Relevância:

10.00% 10.00%

Publicador:

Resumo:

Projeto de Pós-Graduação/Dissertação apresentado à Universidade Fernando Pessoa como parte dos requisitos para obtenção do grau de Mestre em Ciências Farmacêuticas

Relevância:

10.00% 10.00%

Publicador:

Resumo:

Following resistance exercise in the fasted state, both protein synthesis and degradation in skeletal muscle are increased. The addition of essential amino acids potentiates the synthetic response suggesting that an amino acid sensor, which is involved in both synthesis and degradation, may be activated by resistance exercise. One such candidate protein is the class 3 phosphatidylinositol 3OH-kinase (PI3K) Vps34. To determine whether mammalian Vps34 (mVps34) is modulated by high-resistance contractions, mVps34 and S6K1 (an index of mTORC1) activity were measured in the distal hindlimb muscles of rats 0.5, 3, 6 and 18 h after acute unilateral high-resistance contractions with the contralateral muscles serving as a control. In the lengthening tibialis anterior (TA) muscle, S6K1 (0.5 h = 366.3 +/- 112.08%, 3 h = 124.7 +/- 15.96% and 6 h = 129.2 +/- 0%) and mVps34 (3 h = 68.8 +/- 15.1% and 6 h = 36.0 +/- 8.79%) activity both increased, whereas in the shortening soleus and plantaris (PLN) muscles the increase was significantly lower (PLN S6K1 0.5 h = 33.1 +/- 2.29% and 3 h = 47.0 +/- 6.65%; mVps34 3 h = 24.5 +/- 7.92%). HPLC analysis of the TA demonstrated a 25% increase in intramuscular leucine concentration in rats 1.5 h after exercise. A similar level of leucine added to C2C12 cells in vitro increased mVps34 activity 3.2-fold. These data suggest that, following high-resistance contractions, mVps34 activity is stimulated by an influx of essential amino acids such as leucine and this may prolong mTORC1 signalling and contribute to muscle hypertrophy.

Relevância:

10.00% 10.00%

Publicador:

Resumo:

Inhibition of the PI3K (phosphoinositide 3-kinase)/Akt/mTORC1 (mammalian target of rapamycin complex 1) and Ras/MEK [MAPK (mitogen-activated protein kinase)/ERK (extracellular-signal-regulated kinase) kinase]/ERK pathways for cancer therapy has been pursued for over a decade with limited success. Emerging data have indicated that only discrete subsets of cancer patients have favourable responses to these inhibitors. This is due to genetic mutations that confer drug insensitivity and compensatory mechanisms. Therefore understanding of the feedback mechanisms that occur with respect to specific genetic mutations may aid identification of novel biomarkers that predict patient response. In the present paper, we show that feedback between the PI3K/Akt/mTORC1 and Ras/MEK/ERK pathways is cell-line-specific and highly dependent on the activating mutation of K-Ras or overexpression c-Met. We found that cell lines exhibited differential signalling and apoptotic responses to PD184352, a specific MEK inhibitor, and PI103, a second-generation class I PI3K inhibitor. We reveal that feedback from the PI3K/Akt/mTORC1 to the Ras/MEK/ERK pathway is present in cancer cells harbouring either K-Ras activating mutations or amplification of c-Met but not the wild-type counterparts. Moreover, we demonstrate that inhibition of protein phosphatase activity by OA (okadaic acid) restored PI103-mediated feedback in wild-type cells. Together, our results demonstrate a novel mechanism for feedback between the PI3K/Akt/mTORC1 and the Ras/MEK/ERK pathways that only occurs in K-Ras mutant and c-Met amplified cells but not the isogenic wild-type cells through a mechanism that may involve inhibition of a specific endogenous phosphatase(s) activity. We conclude that monitoring K-Ras and c-Met status are important biomarkers for determining the efficacy of PI103 and other PI3K/Akt inhibitors in cancer therapy.

Relevância:

10.00% 10.00%

Publicador:

Resumo:

Le système rénine-angiotensine-aldostérone (SRAA) régule l’homéostasie de la contraction des artères. Or, suivant la liaison de l’angiotensine II (Ang II) à son récepteur AT1, le SRAA est également impliqué dans l’activation de voies de signalisation à l’origine de l’inflammation et de l’hypertrophie des cellules musculaires lisses vasculaires (CMLV), soit deux processus participant au remodelage vasculaire caractéristique de diverses maladies cardiovasculaires, telles l’hypertension et l’athérosclérose. Ces pathologies sont les premières causes de mortalité naturelle en Amérique et les traitements les ciblant ne sont pas optimaux puisqu’ils visent seulement quelques facteurs de risque qui leur sont associés. Ainsi, la détermination des effecteurs intracellulaires régulant ces voies délétères est nécessaire à l'identification de nouvelles cibles thérapeutiques. L’inflammation Ang II-dépendante dans les CMLV est attribuée au facteur de transcription nuclear factor-kappa B (NF-κB). Cependant, les processus moléculaires couplant le récepteur AT1 à son activation sont peu caractérisés. L’étude abordant cette question démontre in vitro que NF-κB est activé par la protéine IκB kinase β (IKKβ) dans les CMLV exposées à l’Ang II et que cette kinase est régulée par deux voies de signalisation indépendantes, mais complémentaires afin d’assurer son activation rigoureuse et soutenue. L’une des voies est précoce et dépend des seconds messagers ainsi que de deux nouveaux effecteurs sous-jacents au récepteur AT1, soit la E3 ligase TNF receptor-associated factor 6 (TRAF6) et la IKK kinase transforming growth factor-beta-activated kinase 1 (TAK1) tandis que la seconde est tardive et résulte de la signalisation mitogen-activated protein kinase kinase 1/2 (MEK1/2) - extracellular signal-regulated kinase 1/2 (ERK1/2) - ribosomal S6 kinase (RSK). L’inhibition conjointe de ces voies abroge complètement la réponse inflammatoire, ce qui indique qu’elles en sont la seule source. Ainsi, l’inhibition d’IKKβ pourrait suffire à contrer l’inflammation impliquée dans le remodelage vasculaire associé à une suractivation du SRAA. Une découverte des plus novatrices découle de cette étude, qui veut que la E3 ligase TRAF6 est un nouvel effecteur des récepteurs couplés aux protéines G et est à l’origine de la formation d’un nouveau type de second messager, soit des chaînes libres de poly-ubiquitines. Les mécanismes moléculaires à la base de l’hypertrophie Ang II-dépendante dans les CMLV sont également peu définis. Or, suivant la parution d’un article démontrant qu’IKKβ dans les cellules cancéreuses participe aux mécanismes d’initiation de la traduction en réponse au facteur de nécrose tumorale α (TNFα) via la phosphorylation de la protéine Tuberous sclerosis 1 (TSC1) et donc l’activation du complexe mammalian target of rapamycin (mTORC1), une hypothèse a été émise selon laquelle cette kinase serait impliquée dans la synthèse protéique Ang II-dépendante dans les CMLV. Les expériences effectuées in vitro dans des CMLV exposées à l’Ang II démontrent qu’IKKβ induit la phosphorylation de TSC1 ainsi que l’activation de mTORC1 et de ses substrats S6 kinase 1 (S6K1) et translational regulators eukaryotic translation initiation factor 4E-binding protein (4E-BP1), deux protéines impliquées directement dans l’hypertrophie. Par ailleurs, la synthèse protéique au niveau des CMLV exposées à l’Ang II est réduite de 75% suivant la diminution de l’expression d’IKKβ et suivant la surexpression d’un mutant de TSC1 dont le site consensus d’IKKβ a été modifié, faisant de cette kinase un médiateur majeur au niveau de ce processus. Ainsi, in vitro IKKβ en réponse à l’Ang II est en amont de deux processus impliqués dans un remodelage vasculaire à l’origine de maladies cardiovasculaires. De plus, plusieurs facteurs de risque de ces pathologies convergent à l’activation d’IKKβ, ce qui en fait une cible thérapeutique particulièrement attrayante. Qui plus est, l’administration d’un inhibiteur d’IKKβ à des rats diminue non seulement la synthèse protéique dépendante de l’Ang II au niveau de l’aorte et des artères mésentériques, mais également la synthèse de la protéine pro-inflammatoire VCAM-1 par les cellules composant l’aorte, ce qui confirme son envergure en tant que cible.

Relevância:

10.00% 10.00%

Publicador:

Resumo:

La plasticité synaptique activité-dépendante forme la base physiologique de l’apprentissage et de la mémoire dépendants de l’hippocampe. Le rôle joué par les différents sous-types d’interneurones dans l’apprentissage et la mémoire hippocampiques reste inconnu, mais repose probablement sur des mécanismes de la plasticité spécifique aux synapses de certains sous-types d’interneurones. Les synapses excitatrices établies sur les interneurones de l’oriens-alveus dans l’aire CA1 exhibent une forme persistante de potentialisation à long terme induite par la stimulation chimique des récepteurs métabotropiques du glutamate de type 1 (mGluR1) [mGluR1-mediated chemical late long-term potentiation (cL-LTPmGluR1)]. Le présent projet de recherche avait pour objectifs d’identifier les sous-types d’interneurones de l’oriens-alveus exprimant la cL-LTPmGluR1 et d’examiner les mécanismes d’induction et d’expression de celle-ci. Nous avons déterminé que la stimulation répétée des mGluR1 induit de la cL-LTPmGluR1 aux synapses excitatrices établies sur le sous-type d’interneurones exprimant le peptide somatostatine (SOM-INs). Des enregistrements électrophysiologiques couplés à des inhibiteurs pharmacologiques et à un knock-out fonctionnel de mammalian target of rapamycin complexe 1 (mTORC1) ont montré que l’induction de la cL-LTPmGluR1 (qui consiste en trois applications de l’agoniste des mGluR1/5, le (S)-3,5-dihydroxyphénylglycine (DHPG) en présence de l’antagoniste des récepteurs métabotropiques du glutamate de type 5 (mGluR5), le 2-méthyl-6-(phényléthynyl)-pyridine (MPEP)) des SOM-INs requiert les voies de signalisation des mGluR1, de extracellular signal-regulated protein kinase (ERK) et de mTORC1. L’ensemble de nos résultats montre qu’une forme persistante de plasticité synaptique sous-tendue par mTORC1 est induite par la stimulation répétée des mGluR1 dans les interneurones hippocampiques exprimant le peptide somatostatine. La connaissance des mécanismes sous-tendant la cL-LTPmGluR1, couplée à l’utilisation de modèles animal in vivo, rendront maintenant possible le blocage de la cL-LTPmGluR1 dans les SOM-INs et l’examen de son rôle dans l’apprentissage et la mémoire dépendants de l’hippocampe.