Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Mais filtros

Base de dados
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
Proc Natl Acad Sci U S A ; 114(17): E3434-E3443, 2017 04 25.
Artigo em Inglês | MEDLINE | ID: mdl-28396387

RESUMO

Oncogenic PIK3CA mutations are found in a significant fraction of human cancers, but therapeutic inhibition of PI3K has only shown limited success in clinical trials. To understand how mutant PIK3CA contributes to cancer cell proliferation, we used genome scale loss-of-function screening in a large number of genomically annotated cancer cell lines. As expected, we found that PIK3CA mutant cancer cells require PIK3CA but also require the expression of the TCA cycle enzyme 2-oxoglutarate dehydrogenase (OGDH). To understand the relationship between oncogenic PIK3CA and OGDH function, we interrogated metabolic requirements and found an increased reliance on glucose metabolism to sustain PIK3CA mutant cell proliferation. Functional metabolic studies revealed that OGDH suppression increased levels of the metabolite 2-oxoglutarate (2OG). We found that this increase in 2OG levels, either by OGDH suppression or exogenous 2OG treatment, resulted in aspartate depletion that was specifically manifested as auxotrophy within PIK3CA mutant cells. Reduced levels of aspartate deregulated the malate-aspartate shuttle, which is important for cytoplasmic NAD+ regeneration that sustains rapid glucose breakdown through glycolysis. Consequently, because PIK3CA mutant cells exhibit a profound reliance on glucose metabolism, malate-aspartate shuttle deregulation leads to a specific proliferative block due to the inability to maintain NAD+/NADH homeostasis. Together these observations define a precise metabolic vulnerability imposed by a recurrently mutated oncogene.


Assuntos
Classe I de Fosfatidilinositol 3-Quinases , Complexo Cetoglutarato Desidrogenase , Mutação , Proteínas de Neoplasias , Neoplasias , Animais , Linhagem Celular Tumoral , Ciclo do Ácido Cítrico/genética , Classe I de Fosfatidilinositol 3-Quinases/genética , Classe I de Fosfatidilinositol 3-Quinases/metabolismo , Glicólise/genética , Humanos , Complexo Cetoglutarato Desidrogenase/biossíntese , Complexo Cetoglutarato Desidrogenase/genética , Camundongos , Camundongos Nus , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Neoplasias/enzimologia , Neoplasias/genética , Neoplasias/patologia
2.
J Virol ; 81(23): 13265-70, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17898070

RESUMO

Fanconi anemia (FA) patients have an increased risk for squamous cell carcinomas (SCCs) at sites of predilection for infection with high-risk human papillomavirus (HPV) types, including the oral cavity and the anogenital tract. We show here that activation of the FA pathway is a frequent event in cervical SCCs. We found that FA pathway activation is triggered mainly by the HPV type 16 (HPV-16) E7 oncoprotein and is associated with an enhanced formation of large FANCD2 foci and recruitment of FANCD2 as well as FANCD1/BRCA2 to chromatin. Episomal expression of HPV-16 oncoproteins was sufficient to activate the FA pathway. Importantly, the expression of HPV-16 E7 in FA-deficient cells led to accelerated chromosomal instability. Taken together, our findings establish the FA pathway as an early host cell response to high-risk HPV infection and may help to explain the greatly enhanced susceptibility of FA patients to squamous cell carcinogenesis at anatomic sites that are frequently infected by high-risk HPVs.


Assuntos
Proteína BRCA2/metabolismo , Instabilidade Cromossômica , Proteína do Grupo de Complementação D2 da Anemia de Fanconi/metabolismo , Papillomavirus Humano 16/metabolismo , Proteínas Oncogênicas Virais/metabolismo , Proteínas Reguladoras de Apoptose , Linhagem Celular , Cromatina/metabolismo , Humanos , Proteínas E7 de Papillomavirus , Ligação Proteica
3.
Cancer Discov ; 3(9): 1044-57, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23764425

RESUMO

UNLABELLED: 3q26 is frequently amplified in several cancer types with a common amplified region containing 20 genes. To identify cancer driver genes in this region, we interrogated the function of each of these genes by loss- and gain-of-function genetic screens. Specifically, we found that TLOC1 (SEC62) was selectively required for the proliferation of cell lines with 3q26 amplification. Increased TLOC1 expression induced anchorage-independent growth, and a second 3q26 gene, SKIL (SNON), facilitated cell invasion in immortalized human mammary epithelial cells. Expression of both TLOC1 and SKIL induced subcutaneous tumor growth. Proteomic studies showed that TLOC1 binds to DDX3X, which is essential for TLOC1-induced transformation and affected protein translation. SKIL induced invasion through upregulation of SLUG (SNAI2) expression. Together, these studies identify TLOC1 and SKIL as driver genes at 3q26 and more broadly suggest that cooperating genes may be coamplified in other regions with somatic copy number gain. SIGNIFICANCE: These studies identify TLOC1 and SKIL as driver genes in 3q26. These observations provide evidence that regions of somatic copy number gain may harbor cooperating genes of different but complementary functions.


Assuntos
Cromossomos Humanos Par 3/genética , Peptídeos e Proteínas de Sinalização Intracelular/genética , Proteínas de Membrana Transportadoras/genética , Invasividade Neoplásica/genética , Neoplasias/genética , Proteínas Proto-Oncogênicas/genética , Neoplasias da Mama/genética , Carcinoma Pulmonar de Células não Pequenas/genética , Linhagem Celular Tumoral , Proliferação de Células , RNA Helicases DEAD-box/metabolismo , Variações do Número de Cópias de DNA/genética , Transição Epitelial-Mesenquimal/genética , Feminino , Amplificação de Genes/genética , Regulação Neoplásica da Expressão Gênica , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Neoplasias Pulmonares/genética , Glândulas Mamárias Humanas/citologia , Proteínas de Membrana Transportadoras/metabolismo , Neoplasias Ovarianas/genética , Ligação Proteica , Proteínas Proto-Oncogênicas/metabolismo , Interferência de RNA , RNA Interferente Pequeno , Fatores de Transcrição da Família Snail , Fatores de Transcrição/biossíntese
4.
Cancer Lett ; 305(2): 113-22, 2011 Jun 28.
Artigo em Inglês | MEDLINE | ID: mdl-21075512

RESUMO

High-risk HPV E6 and E7 oncoproteins cooperate to subvert critical host cell cycle checkpoint control mechanisms in order to promote viral genome replication. This results not only in aberrant proliferation but also in host cellular changes that can promote genomic instability. The HPV-16 E7 oncoprotein was found to induce centrosome abnormalities thereby disrupting mitotic fidelity and increasing the risk for chromosome missegregation and aneuploidy. In addition, expression of the high-risk HPV E7 oncoprotein stimulates DNA replication stress as a potential source of DNA breakage and structural chromosomal instability. Proliferation of genomically unstable cells is sustained by several mechanisms including the accelerated degradation of claspin by HPV-16 E7 and the degradation of p53 by the high-risk HPV E6 oncoprotein. These results highlight the oncogenic potential of aberrant proliferation and opens new avenues for prevention of malignant progression, not only in HPV-associated cervical cancer but also in non-virally associated malignancies with disrupted cell cycle checkpoint control mechanisms.


Assuntos
Instabilidade Genômica , Papillomaviridae/genética , Neoplasias do Colo do Útero/prevenção & controle , Centrossomo/metabolismo , Dano ao DNA , Replicação do DNA , Progressão da Doença , Feminino , Papillomavirus Humano 16/genética , Humanos , Modelos Biológicos , Proteínas Oncogênicas Virais/genética , Fases de Leitura Aberta , Proteínas E7 de Papillomavirus/genética , Proteínas Repressoras/genética , Neoplasias do Colo do Útero/genética , Neoplasias do Colo do Útero/virologia , Replicação Viral
5.
Nat Genet ; 43(10): 964-968, 2011 Sep 04.
Artigo em Inglês | MEDLINE | ID: mdl-21892161

RESUMO

Prior studies have identified recurrent oncogenic mutations in colorectal adenocarcinoma and have surveyed exons of protein-coding genes for mutations in 11 affected individuals. Here we report whole-genome sequencing from nine individuals with colorectal cancer, including primary colorectal tumors and matched adjacent non-tumor tissues, at an average of 30.7× and 31.9× coverage, respectively. We identify an average of 75 somatic rearrangements per tumor, including complex networks of translocations between pairs of chromosomes. Eleven rearrangements encode predicted in-frame fusion proteins, including a fusion of VTI1A and TCF7L2 found in 3 out of 97 colorectal cancers. Although TCF7L2 encodes TCF4, which cooperates with ß-catenin in colorectal carcinogenesis, the fusion lacks the TCF4 ß-catenin-binding domain. We found a colorectal carcinoma cell line harboring the fusion gene to be dependent on VTI1A-TCF7L2 for anchorage-independent growth using RNA interference-mediated knockdown. This study shows previously unidentified levels of genomic rearrangements in colorectal carcinoma that can lead to essential gene fusions and other oncogenic events.


Assuntos
Adenocarcinoma/genética , Neoplasias Colorretais/genética , Proteínas de Fusão Oncogênica , Proteínas Qb-SNARE/genética , Proteína 2 Semelhante ao Fator 7 de Transcrição/genética , Adenocarcinoma/patologia , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/genética , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/metabolismo , Linhagem Celular Tumoral , Transformação Celular Neoplásica/genética , Neoplasias Colorretais/patologia , Éxons , Deleção de Genes , Dosagem de Genes , Técnicas de Silenciamento de Genes , Rearranjo Gênico , Genoma Humano , Humanos , Proteínas Qb-SNARE/metabolismo , Interferência de RNA , Alinhamento de Sequência , Análise de Sequência de DNA , Fator de Transcrição 4 , Proteína 2 Semelhante ao Fator 7 de Transcrição/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , beta Catenina/genética , beta Catenina/metabolismo
6.
Cancer Res ; 70(23): 9959-68, 2010 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-20935219

RESUMO

Patients with the rare genetic disease, Fanconi anemia (FA), are highly susceptible to squamous cell carcinomas arising at multiple anatomic sites including the head and neck region. Human papillomaviruses (HPVs), particularly HPV16, are associated with ∼20% of head and neck squamous cell carcinomas (HNSCCs) in the general population. Some but not other investigators have reported that HNSCCs in FA patients are much more frequently positive for HPV. In addition, studies have demonstrated an interaction between the HPV16 E7 oncoprotein and the FA pathway, a DNA damage response pathway deficient in FA patients. On the basis of these studies, it was hypothesized that the FA pathway contributes to repair of DNA damage induced by HPV16 E7, providing one explanation for why FA patients are predisposed to HPV-associated HNSCCs. To determine the importance of the FA pathway in modulating the oncogenic abilities of E7, we crossed K14E7 transgenic (K14E7) and fancD2 knockout mice (FancD2(-/-)) to establish K14E7/FancD2(-/-) and K14E7/FancD2(+/+) mice and monitored their susceptibility to HNSCC when treated with a chemical carcinogen. K14E7/FancD2(-/-) mice had a significantly higher incidence of HNSCC compared with K14E7/FancD2(+/+) mice. This difference correlated with an increased proliferative index and the increase in expression of biomarkers that are used to assess levels of DNA damage. These animal studies support the hypotheses that FA patients have increased susceptibility to HPV-associated cancer and that the FA DNA damage response pathway normally attenuates the oncogenic potential of HPV16 E7.


Assuntos
Proteína do Grupo de Complementação D2 da Anemia de Fanconi/genética , Predisposição Genética para Doença/genética , Neoplasias de Cabeça e Pescoço/genética , Proteínas E7 de Papillomavirus/genética , 4-Nitroquinolina-1-Óxido , Animais , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/metabolismo , Dano ao DNA , Anemia de Fanconi/genética , Anemia de Fanconi/metabolismo , Proteína do Grupo de Complementação D2 da Anemia de Fanconi/metabolismo , Feminino , Imunofluorescência , Neoplasias de Cabeça e Pescoço/induzido quimicamente , Neoplasias de Cabeça e Pescoço/metabolismo , Humanos , Masculino , Camundongos , Camundongos da Linhagem 129 , Camundongos Knockout , Camundongos Transgênicos , Proteínas E7 de Papillomavirus/metabolismo , Quinolonas , Transdução de Sinais
7.
Cancer Res ; 69(17): 7022-9, 2009 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-19706760

RESUMO

The human papillomavirus (HPV) 16 E7 oncoprotein has been reported previously to stimulate DNA damage and to activate host cell DNA damage checkpoints. How HPV-16 E7 maintains proliferation despite activated DNA damage checkpoints is incompletely understood. Here, we provide evidence that cells expressing the HPV-16 E7 oncoprotein can enter mitosis in the presence of DNA damage. We show that this activity of HPV-16 E7 involves attenuation of DNA damage checkpoint control by accelerating the proteolytic turnover of claspin. Claspin mediates the activation of CHK1 by ATR in response to replication stress, and its degradation plays a critical role in DNA damage checkpoint recovery. Expression of a nondegradable mutant of claspin was shown to inhibit mitotic entry in HPV-16 E7-expressing cells. Multiple components of the SCF(beta-TrCP)-based claspin degradation machinery were found deregulated in the presence of HPV-16 E7, including cullin 1, beta-TrCP, Aurora A, and Polo-like kinase-1 (PLK1). In contrast, no difference in the expression level of the claspin deubiquitinating enzyme USP7 was detected. Levels of Aurora A and PLK1 as well as phosphorylated PLK1 at threonine 210, a prerequisite for DNA damage checkpoint recovery, remained detectable following replication stress in HPV-16 E7-expressing cells but not in control cells. In summary, our results suggest that the HPV-16 E7 oncoprotein alleviates DNA damage checkpoint responses and promotes mitotic entry by accelerating claspin degradation through a mechanism that involves deregulation of components of the SCF(beta-TrCP)-based claspin degradation machinery.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Dano ao DNA , Papillomavirus Humano 16/genética , Mitose/genética , Proteínas Oncogênicas Virais/genética , Infecções por Papillomavirus/genética , Proteínas Adaptadoras de Transdução de Sinal/genética , Aurora Quinases , Proteínas de Ciclo Celular/metabolismo , Proteínas Culina/metabolismo , Regulação Viral da Expressão Gênica , Humanos , Hidrólise , Proteínas E7 de Papillomavirus , Infecções por Papillomavirus/metabolismo , Infecções por Papillomavirus/patologia , Infecções por Papillomavirus/virologia , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Contendo Repetições de beta-Transducina/metabolismo , Quinase 1 Polo-Like
8.
Environ Mol Mutagen ; 50(8): 741-7, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19326465

RESUMO

Centrosome aberrations are a frequent finding in human tumors. However, very little is known about the molecular mechanisms leading to disruption of centrosome duplication control and the functional consequences of aberrant centrosome numbers. The high-risk human papillomavirus Type 16 (HPV-16) E6 and E7 oncoproteins are overexpressed in HPV-associated malignancies of the anogenital tract and have been instrumental in delineating different pathways of centrosome amplification. Whereas the E6 oncoprotein was found to provoke centrosome accumulation, the HPV-16 E7 oncoprotein triggers a genuine disruption of the centrosome duplication cycle. Importantly, the E7 oncoprotein can rapidly cause centrosome overduplication through a pathway that involves the concurrent formation of multiple daughters at single maternal centrioles (centriole flowers). Several lines of evidence suggest that cyclin E/CDK2 complexes and Polo-like kinase 4 (PLK4) are crucial players in this process. These findings underscore that the HPV-16 E7 oncoprotein is a unique tool to dissect normal and abnormal centriole biogenesis and the underlying molecular circuitry.


Assuntos
Centrossomo , Instabilidade Cromossômica , Aberrações Cromossômicas , Papillomaviridae/fisiologia , Proteínas Virais/fisiologia , Humanos
9.
Cancer Res ; 68(23): 9954-63, 2008 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-19047177

RESUMO

Expression of the high-risk human papillomavirus (HPV-16) E7 oncoprotein extends the life span of primary human keratinocytes and partially restores telomere length in the absence of telomerase. The molecular basis of this activity is incompletely understood. Here, we show that HPV-16 E7 induces an increased formation of alternative lengthening of telomeres (ALT)-associated promyelocytic leukemia bodies (APBs) in early passage primary human keratinocytes as well as HPV-negative tumor cells. This activity was found to require sequences of HPV-16 E7 involved in degradation of the retinoblastoma tumor suppressor protein as well as regions in the COOH terminus. HPV-16 E7-induced APBs contained ssDNA and several proteins that are involved in the response to DNA replication stress, most notably the Fanconi anemia D2 protein (FANCD2) as well as BRCA2 and MUS81. In line with these results, we found that FANCD2-containing APBs form in an ATR-dependent manner in HPV-16 E7-expressing cells. To directly show a role of FANCD2 in ALT, we provide evidence that knockdown of FANCD2 rapidly causes telomere dysfunction in cells that rely on ALT to maintain telomeres. Taken together, our results suggest a novel link between replication stress and recombination-based telomere maintenance that may play a role in HPV-16 E7-mediated extension of host cell life span and immortalization.


Assuntos
Corpos de Inclusão Intranuclear/metabolismo , Leucemia Promielocítica Aguda/metabolismo , Proteínas Oncogênicas Virais/biossíntese , Telômero/genética , Linhagem Celular Tumoral , Núcleo Celular/metabolismo , Replicação do DNA , DNA de Neoplasias/biossíntese , DNA de Neoplasias/genética , DNA de Cadeia Simples/genética , DNA de Cadeia Simples/metabolismo , Proteína do Grupo de Complementação D2 da Anemia de Fanconi/biossíntese , Proteína do Grupo de Complementação D2 da Anemia de Fanconi/metabolismo , Células HeLa , Humanos , Corpos de Inclusão Intranuclear/genética , Corpos de Inclusão Intranuclear/virologia , Queratinócitos/patologia , Queratinócitos/virologia , Leucemia Promielocítica Aguda/genética , Leucemia Promielocítica Aguda/virologia , Proteínas E7 de Papillomavirus , RNA Interferente Pequeno/genética , Telômero/metabolismo , Transfecção
10.
J Cell Biochem ; 99(3): 759-69, 2006 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-16676346

RESUMO

Replication stress is a frequent and early event during tumorigenesis. Whereas the cellular responses to a persistent block of replication fork progression have been extensively studied, relatively little is known about how cells respond to low-intensity replication stress. However, transient replication fork perturbations are likely to occur even more frequently in tumor cells than a permanent replication arrest. We report here that transient, low intensity replication stress leads to a rapid activation of the DNA replication checkpoint but to a significantly delayed apoptotic response in a small but significant number of cells. This late apoptotic response was independent of p53 and we found evidence for cell death during mitosis in a proportion of cells. To further explore the role of p53 in the response to replication stress, we analyzed mouse embryonic fibroblasts (MEFs) deficient of p53 in comparison to wild-type or p63- or p73-deficient MEFs. We detected a significant increase of apoptosis and morphological signs of failed mitosis such as multinucleation in p53-deficient MEFs following replication stress, but not in wild-type or p63- or p73-deficient cells. Multinucleated p53-deficient MEFs frequently retained cyclin B1 expression indicating a persistently activated mitotic spindle checkpoint. Collectively, our results suggest that the cellular response to replication stress involves the mitotic spindle checkpoint in a proportion of cells. These findings imply that the mitotic spindle checkpoint may act in concert with DNA damage and cell-cycle checkpoints as an early anti-tumor barrier and provide a possible explanation for its frequent relaxation in human cancer.


Assuntos
Ciclo Celular/fisiologia , Dano ao DNA , Replicação do DNA , Fuso Acromático/metabolismo , Animais , Apoptose/fisiologia , Forma Celular , Células Cultivadas , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Fibroblastos/citologia , Fibroblastos/fisiologia , Histonas/genética , Histonas/metabolismo , Humanos , Camundongos , Camundongos Knockout , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Transdução de Sinais/fisiologia , Transativadores/genética , Transativadores/metabolismo , Proteína Tumoral p73 , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo , Proteínas Supressoras de Tumor/genética , Proteínas Supressoras de Tumor/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA