Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
1.
Proc Natl Acad Sci U S A ; 110(38): E3640-9, 2013 Sep 17.
Artigo em Inglês | MEDLINE | ID: mdl-24003131

RESUMO

Despite considerable efforts to sequence hypermutated cancers such as melanoma, distinguishing cancer-driving genes from thousands of recurrently mutated genes remains a significant challenge. To circumvent the problematic background mutation rates and identify new melanoma driver genes, we carried out a low-copy piggyBac transposon mutagenesis screen in mice. We induced eleven melanomas with mutation burdens that were 100-fold lower relative to human melanomas. Thirty-eight implicated genes, including two known drivers of human melanoma, were classified into three groups based on high, low, or background-level mutation frequencies in human melanomas, and we further explored the functional significance of genes in each group. For two genes overlooked by prevailing discovery methods, we found that loss of membrane associated guanylate kinase, WW and PDZ domain containing 2 and protein tyrosine phosphatase, receptor type, O cooperated with the v-raf murine sarcoma viral oncogene homolog B (BRAF) recurrent V600E mutation to promote cellular transformation. Moreover, for infrequently mutated genes often disregarded by current methods, we discovered recurrent mitogen-activated protein kinase kinase kinase 1 (Map3k1)-activating insertions in our screen, mirroring recurrent MAP3K1 up-regulation in human melanomas. Aberrant expression of Map3k1 enabled growth factor-autonomous proliferation and drove BRAF-independent ERK signaling, thus shedding light on alternative means of activating this prominent signaling pathway in melanoma. In summary, our study contributes several previously undescribed genes involved in melanoma and establishes an important proof-of-principle for the utility of the low-copy transposon mutagenesis approach for identifying cancer-driving genes, especially those masked by hypermutation.


Assuntos
Elementos de DNA Transponíveis/genética , Regulação Neoplásica da Expressão Gênica/fisiologia , MAP Quinase Quinase Quinase 1/metabolismo , Melanoma/genética , Mutagênese Insercional/genética , Transdução de Sinais/fisiologia , Animais , Western Blotting , Primers do DNA/genética , Regulação Neoplásica da Expressão Gênica/genética , Testes Genéticos , Células HEK293 , Humanos , Imuno-Histoquímica , Camundongos , Camundongos Transgênicos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/genética , Especificidade da Espécie
2.
Cell Rep ; 28(13): 3435-3449.e5, 2019 09 24.
Artigo em Inglês | MEDLINE | ID: mdl-31553912

RESUMO

Comprehensive sequencing approaches have allowed for the identification of the most frequent contributors to cancer, known as drivers. They have also revealed a class of mutations in understudied, infrequently altered genes, referred to as "long tail" (LT) drivers. A key challenge has been to find clinically relevant LT drivers and to understand how they cooperate to drive disease. Here, we identified far upstream binding protein 1 (FUBP1) as an LT driver using an in vivo CRISPR screen. FUBP1 cooperates with other tumor suppressor genes to transform mammary epithelial cells by disrupting cellular differentiation and tissue architecture. Mechanistically, FUBP1 participates in regulating N6-methyladenosine (m6A) RNA methylation, and its loss leads to global changes in RNA splicing and widespread expression of aberrant driver isoforms. These findings suggest that somatic alteration of a single gene involved in RNA splicing and m6A methylation can produce the necessary panoply of contributors for neoplastic transformation.


Assuntos
Processamento Alternativo/genética , Proteínas de Ligação a DNA/genética , Neoplasias/genética , Oncogenes/genética , Proteínas de Ligação a RNA/genética , Genes Supressores de Tumor , Humanos
3.
Sci Rep ; 8(1): 1415, 2018 01 23.
Artigo em Inglês | MEDLINE | ID: mdl-29362392

RESUMO

In cancer, tumor suppressor genes (TSGs) are frequently truncated, causing their encoded products to be non-functional or dominant-negative. We previously showed that premature polyadenylation (pPA) of MAGI3 truncates the gene, switching its functional role from a TSG to a dominant-negative oncogene. Here we report that MAGI3 undergoes pPA at the intron immediately downstream of its large internal exon, which is normally highly modified by N6-methyladenosine (m6A). In breast cancer cells that upregulate MAGI3 pPA , m6A levels in the large internal exon of MAGI3 are significantly reduced compared to cells that do not express MAGI3 pPA . We further find that MAGI3 pPA transcripts are significantly depleted of m6A modifications, in contrast to highly m6A-modified full-length MAGI3 mRNA. Finally, we analyze public expression data and find that other TSGs, including LATS1 and BRCA1, also undergo intronic pPA following large internal exons, and that m6A levels in these exons are reduced in pPA-activated breast cancer cells relative to untransformed mammary cells. Our study suggests that m6A may play a role in regulating intronic pPA of MAGI3 and possibly other TSGs, warranting further investigation.


Assuntos
Adenosina/análogos & derivados , Proteína BRCA1/genética , Neoplasias da Mama/genética , Proteínas de Membrana/genética , Proteínas Serina-Treonina Quinases/genética , Adenosina/metabolismo , Proteína BRCA1/metabolismo , Linhagem Celular Tumoral , Éxons , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Íntrons , Proteínas de Membrana/metabolismo , Poliadenilação , Proteínas Serina-Treonina Quinases/metabolismo , RNA Mensageiro/metabolismo , Regulação para Cima
4.
Elife ; 52016 05 20.
Artigo em Inglês | MEDLINE | ID: mdl-27205883

RESUMO

Genetic mutation, chromosomal rearrangement and copy number amplification are common mechanisms responsible for generating gain-of-function, cancer-causing alterations. Here we report a new mechanism by which premature cleavage and polyadenylation (pPA) of RNA can produce an oncogenic protein. We identify a pPA event at a cryptic intronic poly(A) signal in MAGI3, occurring in the absence of local exonic and intronic mutations. The altered mRNA isoform, called MAGI3(pPA), produces a truncated protein that acts in a dominant-negative manner to prevent full-length MAGI3 from interacting with the YAP oncoprotein, thereby relieving YAP inhibition and promoting malignant transformation of human mammary epithelial cells. We additionally find evidence for recurrent expression of MAGI3(pPA) in primary human breast tumors but not in tumor-adjacent normal tissues. Our results provide an example of how pPA contributes to cancer by generating a truncated mRNA isoform that encodes an oncogenic, gain-of-function protein.


Assuntos
Neoplasias da Mama/patologia , Proteínas de Membrana/biossíntese , Proteínas Mutantes/biossíntese , Poliadenilação , Isoformas de RNA/metabolismo , Linhagem Celular Tumoral , Humanos , Proteínas de Membrana/genética , Proteínas Mutantes/genética , Isoformas de RNA/genética
5.
Cell Rep ; 17(5): 1302-1317, 2016 10 25.
Artigo em Inglês | MEDLINE | ID: mdl-27783945

RESUMO

Overabundance of Slug protein is common in human cancer and represents an important determinant underlying the aggressiveness of basal-like breast cancer (BLBC). Despite its importance, this transcription factor is rarely mutated in BLBC, and the mechanism of its deregulation in cancer remains unknown. Here, we report that Slug undergoes acetylation-dependent protein degradation and identify the deacetylase SIRT2 as a key mediator of this post-translational mechanism. SIRT2 inhibition rapidly destabilizes Slug, whereas SIRT2 overexpression extends Slug stability. We show that SIRT2 deacetylates Slug protein at lysine residue K116 to prevent Slug degradation. Interestingly, SIRT2 is frequently amplified and highly expressed in BLBC. Genetic depletion and pharmacological inactivation of SIRT2 in BLBC cells reverse Slug stabilization, cause the loss of clinically relevant pathological features of BLBC, and inhibit tumor growth. Our results suggest that targeting SIRT2 may be a rational strategy for diminishing Slug abundance and its associated malignant traits in BLBC.


Assuntos
Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Sirtuína 2/metabolismo , Fatores de Transcrição da Família Snail/metabolismo , Sequência de Aminoácidos , Animais , Linhagem Celular Tumoral , Proliferação de Células , Feminino , Inativação Gênica , Células HEK293 , Humanos , Lisina/metabolismo , Camundongos Endogâmicos NOD , Camundongos SCID , Invasividade Neoplásica , Ligação Proteica , Estabilidade Proteica , Proteômica , Fatores de Transcrição da Família Snail/química , Especificidade por Substrato
6.
PLoS One ; 6(10): e26650, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-22039523

RESUMO

Somatic forward genetic screens have the power to interrogate thousands of genes in a single animal. Retroviral and transposon mutagenesis systems in mice have been designed and deployed in somatic tissues for surveying hematopoietic and solid tumor formation. In the context of cancer, the ability to visually mark mutant cells would present tremendous advantages for identifying tumor formation, monitoring tumor growth over time, and tracking tumor infiltrations and metastases into wild-type tissues. Furthermore, locating mutant clones is a prerequisite for screening and analyzing most other somatic phenotypes. For this purpose, we developed a system using the piggyBac (PB) transposon for somatic mutagenesis with an activated reporter and tracker, called PB-SMART. The PB-SMART mouse genetic screening system can simultaneously induce somatic mutations and mark mutated cells using bioluminescence or fluorescence. The marking of mutant cells enable analyses that are not possible with current somatic mutagenesis systems, such as tracking cell proliferation and tumor growth, detecting tumor cell infiltrations, and reporting tissue mutagenesis levels by a simple ex vivo visual readout. We demonstrate that PB-SMART is highly mutagenic, capable of tumor induction with low copy transposons, which facilitates the mapping and identification of causative insertions. We further integrated a conditional transposase with the PB-SMART system, permitting tissue-specific mutagenesis with a single cross to any available Cre line. Targeting the germline, the system could also be used to conduct F1 screens. With these features, PB-SMART provides an integrated platform for individual investigators to harness the power of somatic mutagenesis and phenotypic screens to decipher the genetic basis of mammalian biology and disease.


Assuntos
Elementos de DNA Transponíveis , Genes Reporter , Mutagênese , Animais , Camundongos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA