Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
bioRxiv ; 2024 Feb 27.
Artigo em Inglês | MEDLINE | ID: mdl-36711811

RESUMO

Cancer cells encounter stresses during tumor progression and metastatic spread, however, how they survive these challenges is not fully understood. We now identify a mechanism for cancer cell survival through the discovery of a multiprotein signaling complex that includes the GTPase Cdc42, the Cdc42 GEF/effector protein Dock7, AKT, mTOR and the mTORC1 regulatory partners TSC1, TSC2, and Rheb. This pro-survival signaling complex sustains the activated state of AKT by preventing its dephosphorylation at Ser473 during serum starvation, resulting in a low but critical activation of a Raptor-independent mTOR/S6K activity. We demonstrate that the Dock7 DHR1 domain, previously of unknown function, is responsible for preserving AKT phosphorylation through an interaction requiring its C2-like motif. Collectively, these findings help address long-standing questions of how Cdc42 signals mTOR activation by elucidating the unique functions of its signaling partner Dock7 as an AKT regulator necessary for resistance to anoikis and apoptosis in cancer cells.

2.
bioRxiv ; 2023 Sep 28.
Artigo em Inglês | MEDLINE | ID: mdl-37808692

RESUMO

Developing therapeutic strategies against COVID-19 has gained widespread interest given the likelihood that new viral variants will continue to emerge. Here we describe one potential therapeutic strategy which involves targeting members of the glutaminase family of mitochondrial metabolic enzymes (GLS and GLS2), which catalyze the first step in glutamine metabolism, the hydrolysis of glutamine to glutamate. We show three examples where GLS expression increases during coronavirus infection of host cells, and another in which GLS2 is upregulated. The viruses hijack the metabolic machinery responsible for glutamine metabolism to generate the building blocks for biosynthetic processes and satisfy the bioenergetic requirements demanded by the 'glutamine addiction' of virus-infected host cells. We demonstrate how genetic silencing of glutaminase enzymes reduces coronavirus infection and that newer members of two classes of small molecule allosteric inhibitors targeting these enzymes, designated as SU1, a pan-GLS/GLS2 inhibitor, and UP4, which is specific for GLS, block viral replication in mammalian epithelial cells. Overall, these findings highlight the importance of glutamine metabolism for coronavirus replication in human cells and show that glutaminase inhibitors can block coronavirus infection and thereby may represent a novel class of anti-viral drug candidates. Teaser: Inhibitors targeting glutaminase enzymes block coronavirus replication and may represent a new class of anti-viral drugs.

3.
Cell Death Discov ; 8(1): 357, 2022 Aug 13.
Artigo em Inglês | MEDLINE | ID: mdl-35963851

RESUMO

Proliferating cancer cells are dependent on glutamine metabolism for survival when challenged with oxidative stresses caused by reactive oxygen species, hypoxia, nutrient deprivation and matrix detachment. ATF4, a key stress responsive transcription factor, is essential for cancer cells to sustain glutamine metabolism when challenged with these various types of stress. While it is well documented how the ATF4 transcript is translated into protein as a stress response, an important question concerns how the ATF4 message levels are sustained to enable cancer cells to survive the challenges of nutrient deprivation and damaging reactive oxygen species. Here, we now identify the pathway in triple negative breast cancer cells that provides a sustained ATF4 response and enables their survival when encountering these challenges. This signaling pathway starts with mTORC2, which upon sensing cellular stresses arising from glutamine deprivation or an acute inhibition of glutamine metabolism, initiates a cascade of events that triggers an increase in ATF4 transcription. Surprisingly, this signaling pathway is not dependent on AKT activation, but rather requires the mTORC2 target, PKC, which activates the transcription factor Nrf2 that then induces ATF4 expression. Additionally, we identify a sirtuin family member, the NAD+-dependent de-succinylase Sirt5, as a key transcriptional target for ATF4 that promotes cancer cell survival during metabolic stress. Sirt5 plays fundamental roles in supporting cancer cell metabolism by regulating various enzymatic activities and by protecting an enzyme essential for glutaminolysis, glutaminase C (GAC), from degradation. We demonstrate that ectopic expression of Sirt5 compensates for knockdowns of ATF4 in cells exposed to glutamine deprivation-induced stress. These findings provide important new insights into the signaling cues that lead to sustained ATF4 expression as a general stress-induced regulator of glutamine metabolism, as well as highlight Sirt5 an essential effector of the ATF4 response to metabolic stress.

4.
J Extracell Vesicles ; 10(3): e12051, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33473262

RESUMO

Tumour-derived microvesicles (MVs) serve as critical mediators of cell-to-cell communication in the tumour microenvironment. So far, the underlying mechanisms of MV biogenesis, especially how key tumorigenesis signals such as abnormal EGF signalling regulates MV release, remain unclear. Here, we set out to establish reliable readouts for MV biogenesis and then explore the molecular mechanisms that regulate MV generation. We found that Rho family small G protein Cdc42 is a convergent node of multiple regulatory signals that occur in MV biogenesis. The binding of activated GTP-bound Cdc42 and its downstream effector, Ras GTPase-activating-like protein 1 (IQGAP1), is required for MV shedding. Activated Cdc42 maintains sustained EGF signalling by inhibiting the internalization of cell surface receptors, including EGFR and the VEGF oligomer, VEGF90K, and then facilitates MV release. Subsequently, we further demonstrated that blocking these signalling pathways using the corresponding mutants effectively reduced MV shedding and significantly inhibited MV-promoted in vivo tumour angiogenesis. These findings reveal a complex regulation of MV shedding by tumour cells, shedding light on the regulatory mechanism of MV biogenesis, and potentially contributing to strategies that target MVs in cancer therapy.


Assuntos
Vesículas Extracelulares/metabolismo , Neoplasias/metabolismo , Microambiente Tumoral , Proteína cdc42 de Ligação ao GTP/metabolismo , Células 3T3 , Animais , Comunicação Celular , Linhagem Celular , Linhagem Celular Tumoral , Micropartículas Derivadas de Células/metabolismo , Humanos , Camundongos , Neovascularização Patológica/metabolismo , Transdução de Sinais , Proteínas Ativadoras de ras GTPase/metabolismo
5.
Dev Cell ; 56(3): 277-291.e6, 2021 02 08.
Artigo em Inglês | MEDLINE | ID: mdl-33321103

RESUMO

It is critical that epiblast cells within blastocyst-stage embryos receive the necessary regulatory cues to remain pluripotent until the appropriate time when they are stimulated to undergo differentiation, ultimately to give rise to an entire organism. Here, we show that exposure of embryonic stem cells (ESCs), which are the in vitro equivalents of epiblasts, to ESC-derived extracellular vesicles (EVs) helps to maintain their stem cell properties even under culture conditions that would otherwise induce differentiation. EV-treated ESCs continued to express stemness genes, preserving their pluripotency and ability to generate chimeric mice. These effects were triggered by fibronectin bound to the surfaces of EVs, enabling them to interact with ESC-associated integrins and activate FAK more effectively than fibronectin alone. Overall, these findings highlight a potential regulatory mechanism whereby epiblast cells, via their shed EVs, create an environment within the blastocyst that prevents their premature differentiation and maintains their pluripotent state.


Assuntos
Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/enzimologia , Vesículas Extracelulares/metabolismo , Proteína-Tirosina Quinases de Adesão Focal/metabolismo , Animais , Células Cultivadas , Quimera/metabolismo , Embrião de Mamíferos/metabolismo , Células-Tronco Embrionárias/ultraestrutura , Ativação Enzimática , Fibroblastos/metabolismo , Fibronectinas/metabolismo , Camundongos , Modelos Biológicos , Fenótipo
6.
Proc Natl Acad Sci U S A ; 116(52): 26625-26632, 2019 Dec 26.
Artigo em Inglês | MEDLINE | ID: mdl-31843902

RESUMO

The mitochondrial enzyme glutaminase (GLS) is frequently up-regulated during tumorigenesis and is being evaluated as a target for cancer therapy. GLS catalyzes the hydrolysis of glutamine to glutamate, which then supplies diverse metabolic pathways with carbon and/or nitrogen. Here, we report that SIRT5, a mitochondrial NAD+-dependent lysine deacylase, plays a key role in stabilizing GLS. In transformed cells, SIRT5 regulates glutamine metabolism by desuccinylating GLS and thereby protecting it from ubiquitin-mediated degradation. Moreover, we show that SIRT5 is up-regulated during cellular transformation and supports proliferation and tumorigenesis. Elevated SIRT5 expression in human breast tumors correlates with poor patient prognosis. These findings reveal a mechanism for increasing GLS expression in cancer cells and establish a role for SIRT5 in metabolic reprogramming and mammary tumorigenesis.

8.
Oncotarget ; 8(14): 22325-22343, 2017 Apr 04.
Artigo em Inglês | MEDLINE | ID: mdl-28423611

RESUMO

Tissue transglutaminase (tTG), a dual-function enzyme with GTP-binding and acyltransferase activities, has been implicated in the survival and chemotherapy resistance of aggressive cancer cells and cancer stem cells, including glioma stem cells (GSCs). Using a model system comprising two distinct subtypes of GSCs referred to as proneural (PN) and mesenchymal (MES), we find that the phenotypically aggressive and radiation therapy-resistant MES GSCs exclusively express tTG relative to PN GSCs. As such, the self-renewal, proliferation, and survival of these cells was sensitive to treatment with tTG inhibitors, with a benefit being observed when combined with the standard of care for high grade gliomas (i.e. radiation or temozolomide). Efforts to understand the molecular drivers of tTG expression in MES GSCs revealed an unexpected link between tTG and a common marker for stem cells and cancer stem cells, Aldehyde dehydrogenase 1A3 (ALDH1A3). ALDH1A3, as well as other members of the ALDH1 subfamily, can function in cells as a retinaldehyde dehydrogenase to generate retinoic acid (RA) from retinal. We show that the enzymatic activity of ALDH1A3 and its product, RA, are necessary for the observed expression of tTG in MES GSCs. Additionally, the ectopic expression of ALDH1A3 in PN GSCs is sufficient to induce the expression of tTG in these cells, further demonstrating a causal link between ALDH1A3 and tTG. Together, these findings ascribe a novel function for ALDH1A3 in an aggressive GSC phenotype via the up-regulation of tTG, and suggest the potential for a similar role by ALDH1 family members across cancer types.


Assuntos
Aldeído Oxirredutases/metabolismo , Neoplasias Encefálicas/metabolismo , Proteínas de Ligação ao GTP/metabolismo , Glioma/metabolismo , Células-Tronco Mesenquimais/fisiologia , Células-Tronco Neoplásicas/fisiologia , Células-Tronco/fisiologia , Transglutaminases/metabolismo , Aldeído Oxirredutases/genética , Biomarcadores Tumorais/metabolismo , Neoplasias Encefálicas/genética , Linhagem Celular Tumoral , Proliferação de Células , Sobrevivência Celular , Dacarbazina/análogos & derivados , Dacarbazina/farmacologia , Proteínas de Ligação ao GTP/genética , Regulação Neoplásica da Expressão Gênica , Glioma/genética , Humanos , Proteína 2 Glutamina gama-Glutamiltransferase , RNA Interferente Pequeno/genética , Temozolomida , Transglutaminases/genética , Tretinoína/metabolismo , Regulação para Cima
9.
Nat Commun ; 8: 14450, 2017 02 16.
Artigo em Inglês | MEDLINE | ID: mdl-28205552

RESUMO

Non-classical secretory vesicles, collectively referred to as extracellular vesicles (EVs), have been implicated in different aspects of cancer cell survival and metastasis. Here, we describe how a specific class of EVs, called microvesicles (MVs), activates VEGF receptors and tumour angiogenesis through a unique 90 kDa form of VEGF (VEGF90K). We show that VEGF90K is generated by the crosslinking of VEGF165, catalysed by the enzyme tissue transglutaminase, and associates with MVs through its interaction with the chaperone Hsp90. We further demonstrate that MV-associated VEGF90K has a weakened affinity for Bevacizumab, causing Bevacizumab to be ineffective in blocking MV-dependent VEGF receptor activation. However, treatment with an Hsp90 inhibitor releases VEGF90K from MVs, restoring the sensitivity of VEGF90K to Bevacizumab. These findings reveal a novel mechanism by which cancer cell-derived MVs influence the tumour microenvironment and highlight the importance of recognizing their unique properties when considering drug treatment strategies.


Assuntos
Benzoquinonas/farmacologia , Bevacizumab/farmacologia , Neoplasias da Mama/patologia , Vesículas Extracelulares/classificação , Vesículas Extracelulares/metabolismo , Lactamas Macrocíclicas/farmacologia , Neovascularização Patológica/metabolismo , Receptores de Fatores de Crescimento do Endotélio Vascular/metabolismo , Indutores da Angiogênese/metabolismo , Animais , Benzoquinonas/metabolismo , Bevacizumab/metabolismo , Linhagem Celular Tumoral/efeitos dos fármacos , Linhagem Celular Tumoral/metabolismo , Micropartículas Derivadas de Células/classificação , Micropartículas Derivadas de Células/metabolismo , Modelos Animais de Doenças , Combinação de Medicamentos , Feminino , Proteínas de Choque Térmico HSP90/efeitos dos fármacos , Proteínas de Choque Térmico HSP90/metabolismo , Células HeLa , Humanos , Lactamas Macrocíclicas/metabolismo , Camundongos , Neovascularização Patológica/patologia , Vesículas Secretórias , Transdução de Sinais , Transglutaminases , Transplante Heterólogo , Microambiente Tumoral , Fator A de Crescimento do Endotélio Vascular/efeitos dos fármacos , Fator A de Crescimento do Endotélio Vascular/metabolismo
10.
Nat Commun ; 7: 11321, 2016 Apr 18.
Artigo em Inglês | MEDLINE | ID: mdl-27089238

RESUMO

Many transformed cells exhibit altered glucose metabolism and increased utilization of glutamine for anabolic and bioenergetic processes. These metabolic adaptations, which accompany tumorigenesis, are driven by oncogenic signals. Here we report that the transcription factor c-Jun, product of the proto-oncogene JUN, is a key regulator of mitochondrial glutaminase (GLS) levels. Activation of c-Jun downstream of oncogenic Rho GTPase signalling leads to elevated GLS gene expression and glutaminase activity. In human breast cancer cells, GLS protein levels and sensitivity to GLS inhibition correlate strongly with c-Jun levels. We show that c-Jun directly binds to the GLS promoter region, and is sufficient to increase gene expression. Furthermore, ectopic overexpression of c-Jun renders breast cancer cells dependent on GLS activity. These findings reveal a role for c-Jun as a driver of cancer cell metabolic reprogramming, and suggest that cancers overexpressing JUN may be especially sensitive to GLS-targeted therapies.


Assuntos
Fibroblastos/citologia , Fibroblastos/metabolismo , Regulação Neoplásica da Expressão Gênica , Glutaminase/genética , Neoplasias/genética , Proteínas Proto-Oncogênicas c-jun/genética , Animais , Sequência de Bases , Western Blotting , Linhagem Celular Tumoral , Proliferação de Células/genética , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/metabolismo , Células Cultivadas , Embrião de Mamíferos/citologia , Glutaminase/metabolismo , Glutamina/metabolismo , Humanos , Células MCF-7 , Camundongos , Dados de Sequência Molecular , Neoplasias/metabolismo , Neoplasias/terapia , Regiões Promotoras Genéticas/genética , Ligação Proteica , Proto-Oncogene Mas , Proteínas Proto-Oncogênicas c-jun/metabolismo , Interferência de RNA , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Proteínas rho de Ligação ao GTP/metabolismo
11.
Mol Cancer Res ; 12(6): 940-52, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24615340

RESUMO

UNLABELLED: Overexpression of the receptor tyrosine kinase HER2/ErbB2 (ERBB2) has been linked to a poor prognosis for patients with breast cancer; thus, its activity is a central target for cancer therapy. Likewise, overexpression of heregulin (HRG/NRG1), a growth factor responsible for ErbB2 activation, has also been shown to be a driver of breast cancer progression. Although ErbB2 inhibitors offer a major advancement in the treatment of ErbB2-dependent breast cancers, patients are highly susceptible to developing clinical resistance to these drugs. Therefore, a detailed understanding of the molecular mechanism that underlies HRG/ErbB2-induced tumorigenesis is essential for the development of effective therapeutic strategies for this subset of patients with breast cancer. Here, it was demonstrated that HRG promoted anchorage-independent breast cancer cell growth more potently than EGF, and that the HRG-dependent activation of phosphoinositide 3-kinase and mTORC1 are necessary events for cell transformation. Functional evaluation of two distinct mTOR (MTOR) inhibitors, rapamycin and INK-128, on HRG-dependent signaling activities, uncovered a necessary role for mTORC2 in the regulation of the AKT/TSC2/mTORC1 axis by affecting the phosphorylation of AKT at the PDK1(PDPK1)-dependent site (T308) as well as at the mTORC2-dependent site (S473). The elimination of Rictor (RICTOR), a critical component of mTORC2, is detrimental to both the activation of mTORC1 and HRG-mediated cellular transformation. Similar results were obtained in multiple breast cancer model systems, highlighting an important role for mTORC2 in HRG/ErbB2-dependent breast cancer. IMPLICATIONS: These findings suggest the potential benefits of targeting mTORC2 in HRG/ErbB2-induced breast cancer.


Assuntos
Transformação Celular Neoplásica/metabolismo , Neuregulina-1/metabolismo , Receptor ErbB-2/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Linhagem Celular Tumoral , Humanos , Células MCF-7 , Fosforilação , Transdução de Sinais , Serina-Treonina Quinases TOR/genética
12.
Future Med Chem ; 5(14): 1685-700, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24047273

RESUMO

The metabolic adaptations that support oncogenic growth can also render cancer cells dependent on certain nutrients. Along with the Warburg effect, increased utilization of glutamine is one of the metabolic hallmarks of the transformed state. Glutamine catabolism is positively regulated by multiple oncogenic signals, including those transmitted by the Rho family of GTPases and by c-Myc. The recent identification of mechanistically distinct inhibitors of glutaminase, which can selectively block cellular transformation, has revived interest in the possibility of targeting glutamine metabolism in cancer therapy. Here, we outline the regulation and roles of glutamine metabolism within cancer cells and discuss possible strategies for, and the consequences of, impacting these processes therapeutically.


Assuntos
Antineoplásicos/farmacologia , Glutaminase/antagonistas & inibidores , Glutamina/metabolismo , Terapia de Alvo Molecular/métodos , Neoplasias/tratamento farmacológico , Neoplasias/metabolismo , Animais , Antineoplásicos/química , Transformação Celular Neoplásica/metabolismo , Glutaminase/metabolismo , Humanos , Modelos Moleculares , Transdução de Sinais/efeitos dos fármacos
13.
Trends Mol Med ; 19(2): 74-82, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23219172

RESUMO

Recently, the small molecule 968 was found to block the Rho GTPase-dependent growth of cancer cells in cell culture and mouse xenografts, and when the target of 968 was found to be the mitochondrial enzyme glutaminase (GLS1), it revealed a surprising link between Rho GTPases and mitochondrial glutamine metabolism. Signal transduction via the Rho GTPases, together with NF-κB, appears to elevate mitochondrial glutaminase activity in cancer cells, thereby helping cancer cells satisfy their altered metabolic demands. Here, we review what is known about the mechanism of glutaminase activation in cancer cells, compare the properties of two distinct glutaminase inhibitors, and discuss recent findings that shed new light on how glutamine metabolism might affect cancer progression.


Assuntos
Neoplasias/metabolismo , Proteínas rho de Ligação ao GTP/metabolismo , Animais , Ativação Enzimática/efeitos dos fármacos , Inibidores Enzimáticos , Glutaminase/antagonistas & inibidores , Glutaminase/metabolismo , Humanos , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Transdução de Sinais , Vesículas Transportadoras , Proteínas rho de Ligação ao GTP/antagonistas & inibidores
14.
Small GTPases ; 3(4): 219-24, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22906997

RESUMO

A novel form of cell-to-cell communication involving the formation and shedding of large vesicular structures, called microvesicles (MVs), from the surfaces of highly aggressive forms of human cancer cells has been attracting increasing amounts of attention. This is in large part due to the fact that MVs contain a variety of cargo that is not typically thought to be released from cells including cell-surface receptor tyrosine kinases, cytosolic and nuclear signaling proteins and RNA transcripts. MVs, by sharing their contents with other cells, can greatly impact cancer progression by increasing primary tumor growth, as well as by promoting the development of the pre-metastatic niche. We have recently shown that the small GTPase RhoA is critical for MV biogenesis in human cancer cells. Moreover, we have now obtained evidence that implicates the highly related small GTPases, Rac and Cdc42, in regulating the loading of specific cargo into MVs, as well as in the shedding of MVs from cancer cells. Thus, linking the Rho family of small GTPases to MV biogenesis has begun to shed some light on a new and unexpected way that these signaling proteins contribute to human cancer progression.


Assuntos
Transformação Celular Neoplásica , Micropartículas Derivadas de Células/metabolismo , Neoplasias/metabolismo , Proteína rhoA de Ligação ao GTP/metabolismo , Animais , Comunicação Celular , Humanos
15.
Cancer Cell ; 18(3): 207-19, 2010 Sep 14.
Artigo em Inglês | MEDLINE | ID: mdl-20832749

RESUMO

Rho GTPases impact a number of activities important for oncogenesis. We describe a small molecule inhibitor that blocks oncogenic transformation induced by various Rho GTPases in fibroblasts, and the growth of human breast cancer and B lymphoma cells, without affecting normal cells. We identify the target of this inhibitor to be the metabolic enzyme glutaminase, which catalyzes the hydrolysis of glutamine to glutamate. We show that transformed fibroblasts and breast cancer cells exhibit elevated glutaminase activity that is dependent on Rho GTPases and NF-κB activity, and is blocked by the small molecule inhibitor. These findings highlight a previously unappreciated connection between Rho GTPase activation and cellular metabolism and demonstrate that targeting glutaminase activity can inhibit oncogenic transformation.


Assuntos
Transformação Celular Neoplásica/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Glutaminase/antagonistas & inibidores , Mitocôndrias/enzimologia , Proteínas rho de Ligação ao GTP/antagonistas & inibidores , Animais , Neoplasias da Mama/enzimologia , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/metabolismo , Feminino , Fibroblastos/efeitos dos fármacos , Fibroblastos/enzimologia , Fibroblastos/patologia , Glutaminase/metabolismo , Humanos , Camundongos , Mitocôndrias/efeitos dos fármacos , Células NIH 3T3 , Transdução de Sinais/efeitos dos fármacos , Transfecção , Proteínas rho de Ligação ao GTP/metabolismo
16.
Nucleus ; 1(2): 139-43, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-21326945

RESUMO

The nuclear cap-binding complex (CBC), a heterodimer comprised of a 20 kDa subunit (CBP20) and an 80 kDa regulatory subunit (CBP80), binds to nascent RNA polymerase II transcripts and is important throughout different aspects of RNA metabolism. In a recent publication, using a combination of X-ray crystallographic information, mutagenesis studies, small-angle scattering experiments, analytical ultracentrifugation and in vivo assays, we presented evidence that importin-α and importin-ß, two nucleocytoplasmic transport proteins, play key roles in regulating the binding of capped RNA by the CBC in cells. A model for how complexes between CBC and the importins cycle in and out of the nucleus and direct the proper positional binding and release of capped RNA is presented here and is discussed in light of recent publications.


Assuntos
Citoplasma/metabolismo , Carioferinas/metabolismo , Dobramento de RNA , Animais , Cristalografia por Raios X , Citosol/metabolismo , Humanos , Carioferinas/química , Complexo Proteico Nuclear de Ligação ao Cap/metabolismo
17.
J Biol Chem ; 285(8): 5815-26, 2010 Feb 19.
Artigo em Inglês | MEDLINE | ID: mdl-20028979

RESUMO

Although the small GTPase Ran is best known for its roles in nucleocytoplasmic transport, mitotic spindle assembly, and nuclear envelope formation, recent studies have demonstrated the overexpression of Ran in multiple tumor types and that its expression is correlated with a poor patient prognosis, providing evidence for the importance of this GTPase in cell growth regulation. Here we show that Ran is subject to growth factor regulation by demonstrating that it is activated in a serum-dependent manner in human breast cancer cells and, in particular, in response to heregulin, a growth factor that activates the Neu/ErbB2 tyrosine kinase. The heregulin-dependent activation of Ran requires mTOR (mammalian target of rapamycin) and stimulates the capped RNA binding capability of the cap-binding complex in the nucleus, thus influencing gene expression at the level of mRNA processing. We further demonstrate that the excessive activation of Ran has important consequences for cell growth by showing that a novel, activated Ran mutant is sufficient to transform NIH-3T3 cells in an mTOR- and epidermal growth factor receptor-dependent manner and that Ran-transformed cells form tumors in mice.


Assuntos
Neoplasias da Mama/enzimologia , Núcleo Celular/enzimologia , Transformação Celular Neoplásica/metabolismo , Neuregulina-1/metabolismo , Proteína ran de Ligação ao GTP/metabolismo , Animais , Neoplasias da Mama/genética , Linhagem Celular Tumoral , Núcleo Celular/genética , Transformação Celular Neoplásica/genética , Ativação Enzimática/efeitos dos fármacos , Feminino , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Camundongos , Mutação , Células NIH 3T3 , Neuregulina-1/farmacologia , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas de Ligação ao Cap de RNA/genética , Proteínas de Ligação ao Cap de RNA/metabolismo , Processamento Pós-Transcricional do RNA/efeitos dos fármacos , Processamento Pós-Transcricional do RNA/genética , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Receptor ErbB-2/genética , Receptor ErbB-2/metabolismo , Serina-Treonina Quinases TOR , Proteína ran de Ligação ao GTP/genética
18.
Nat Struct Mol Biol ; 16(9): 930-7, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19668212

RESUMO

The binding of capped RNAs to the cap-binding complex (CBC) in the nucleus, and their dissociation from the CBC in the cytosol, represent essential steps in RNA processing. Here we show how the nucleocytoplasmic transport proteins importin-alpha and importin-beta have key roles in regulating these events. As a first step toward understanding the molecular basis for this regulation, we determined a 2.2-A resolution X-ray structure for a CBC-importin-alpha complex that provides a detailed picture for how importin-alpha binds to the CBP80 subunit of the CBC. Through a combination of biochemical studies, X-ray crystallographic information and small-angle scattering experiments, we then determined how importin-beta binds to the CBC through its CBP20 subunit. Together, these studies enable us to propose a model describing how importin-beta stimulates the dissociation of capped RNA from the CBC in the cytosol following its nuclear export.


Assuntos
Complexo Proteico Nuclear de Ligação ao Cap/química , alfa Carioferinas/química , beta Carioferinas/química , Sequência de Aminoácidos , Cristalografia por Raios X , Células HeLa , Humanos , Modelos Moleculares , Dados de Sequência Molecular , Complexo Proteico Nuclear de Ligação ao Cap/metabolismo , Ligação Proteica , Estrutura Quaternária de Proteína , Estrutura Terciária de Proteína , Subunidades Proteicas/química , Subunidades Proteicas/metabolismo , Capuzes de RNA/metabolismo , alfa Carioferinas/metabolismo , beta Carioferinas/metabolismo
19.
Nat Struct Biol ; 9(12): 912-7, 2002 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-12434151

RESUMO

The 7-methyl guanosine cap structure of RNA is essential for key aspects of RNA processing, including pre-mRNA splicing, 3' end formation, U snRNA transport, nonsense-mediated decay and translation. Two cap-binding proteins mediate these effects: cytosolic eIF-4E and nuclear cap-binding protein complex (CBC). The latter consists of a CBP20 subunit, which binds the cap, and a CBP80 subunit, which ensures high-affinity cap binding. Here we report the 2.1 A resolution structure of human CBC with the cap analog m7GpppG, as well as the structure of unliganded CBC. Comparisons between these structures indicate that the cap induces substantial conformational changes within the N-terminal loop of CBP20, enabling Tyr 20 to join Tyr 43 in pi-pi stacking interactions with the methylated guanosine base. CBP80 stabilizes the movement of the N-terminal loop of CBP20 and locks the CBC into a high affinity cap-binding state. The structure for the CBC bound to m7GpppG highlights interesting similarities and differences between CBC and eIF-4E, and provides insights into the regulatory mechanisms used by growth factors and other extracellular stimuli to influence the cap-binding state of the CBC.


Assuntos
Fosfatos de Dinucleosídeos/química , Modelos Moleculares , Complexo Proteico Nuclear de Ligação ao Cap/química , Capuzes de RNA/química , Sítios de Ligação , Cristalografia por Raios X , Fosfatos de Dinucleosídeos/metabolismo , Fator de Iniciação 4E em Eucariotos/química , Humanos , Complexo Proteico Nuclear de Ligação ao Cap/metabolismo , Ligação Proteica , Conformação Proteica , Subunidades Proteicas , Capuzes de RNA/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...