Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 316
Filtrar
1.
Cell ; 166(2): 328-342, 2016 Jul 14.
Artigo em Inglês | MEDLINE | ID: mdl-27374332

RESUMO

Metastases are the main cause of cancer deaths, but the mechanisms underlying metastatic progression remain poorly understood. We isolated pure populations of cancer cells from primary tumors and metastases from a genetically engineered mouse model of human small cell lung cancer (SCLC) to investigate the mechanisms that drive the metastatic spread of this lethal cancer. Genome-wide characterization of chromatin accessibility revealed the opening of large numbers of distal regulatory elements across the genome during metastatic progression. These changes correlate with copy number amplification of the Nfib locus, and differentially accessible sites were highly enriched for Nfib transcription factor binding sites. Nfib is necessary and sufficient to increase chromatin accessibility at a large subset of the intergenic regions. Nfib promotes pro-metastatic neuronal gene expression programs and drives the metastatic ability of SCLC cells. The identification of widespread chromatin changes during SCLC progression reveals an unexpected global reprogramming during metastatic progression.


Assuntos
Neoplasias Pulmonares/patologia , Fatores de Transcrição NFI/metabolismo , Metástase Neoplásica/patologia , Carcinoma de Pequenas Células do Pulmão/patologia , Motivos de Aminoácidos , Animais , Linhagem Celular Tumoral , Células Cultivadas , Modelos Animais de Doenças , Regulação Neoplásica da Expressão Gênica , Técnicas de Silenciamento de Genes , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Camundongos , Fatores de Transcrição NFI/genética , Regiões Promotoras Genéticas , Carcinoma de Pequenas Células do Pulmão/genética , Carcinoma de Pequenas Células do Pulmão/metabolismo , Regulação para Cima
2.
Mol Cell ; 82(16): 3103-3118.e8, 2022 08 18.
Artigo em Inglês | MEDLINE | ID: mdl-35752172

RESUMO

The development of CRISPR-based barcoding methods creates an exciting opportunity to understand cellular phylogenies. We present a compact, tunable, high-capacity Cas12a barcoding system called dual acting inverted site array (DAISY). We combined high-throughput screening and machine learning to predict and optimize the 60-bp DAISY barcode sequences. After optimization, top-performing barcodes had ∼10-fold increased capacity relative to the best random-screened designs and performed reliably across diverse cell types. DAISY barcode arrays generated ∼12 bits of entropy and ∼66,000 unique barcodes. Thus, DAISY barcodes-at a fraction of the size of Cas9 barcodes-achieved high-capacity barcoding. We coupled DAISY barcoding with single-cell RNA-seq to recover lineages and gene expression profiles from ∼47,000 human melanoma cells. A single DAISY barcode recovered up to ∼700 lineages from one parental cell. This analysis revealed heritable single-cell gene expression and potential epigenetic modulation of memory gene transcription. Overall, Cas12a DAISY barcoding is an efficient tool for investigating cell-state dynamics.


Assuntos
Sistemas CRISPR-Cas , Código de Barras de DNA Taxonômico , Linhagem da Célula/genética , Código de Barras de DNA Taxonômico/métodos , Humanos , Aprendizado de Máquina , Filogenia
3.
Genes Dev ; 35(15-16): 1109-1122, 2021 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-34301766

RESUMO

Lung adenocarcinoma, the most prevalent lung cancer subtype, is characterized by its high propensity to metastasize. Despite the importance of metastasis in lung cancer mortality, its underlying cellular and molecular mechanisms remain largely elusive. Here, we identified miR-200 miRNAs as potent suppressors for lung adenocarcinoma metastasis. miR-200 expression is specifically repressed in mouse metastatic lung adenocarcinomas, and miR-200 decrease strongly correlates with poor patient survival. Consistently, deletion of mir-200c/141 in the KrasLSL-G12D/+ ; Trp53flox/flox lung adenocarcinoma mouse model significantly promoted metastasis, generating a desmoplastic tumor stroma highly reminiscent of metastatic human lung cancer. miR-200 deficiency in lung cancer cells promotes the proliferation and activation of adjacent cancer-associated fibroblasts (CAFs), which in turn elevates the metastatic potential of cancer cells. miR-200 regulates the functional interaction between cancer cells and CAFs, at least in part, by targeting Notch ligand Jagged1 and Jagged2 in cancer cells and inducing Notch activation in adjacent CAFs. Hence, the interaction between cancer cells and CAFs constitutes an essential mechanism to promote metastatic potential.


Assuntos
Fibroblastos Associados a Câncer , Neoplasias Pulmonares , MicroRNAs , Animais , Linhagem Celular Tumoral , Proliferação de Células/genética , Fibroblastos/metabolismo , Regulação Neoplásica da Expressão Gênica , Humanos , Neoplasias Pulmonares/metabolismo , Camundongos , MicroRNAs/genética , MicroRNAs/metabolismo , Metástase Neoplásica/patologia
4.
Mol Cell ; 80(3): 452-469.e9, 2020 11 05.
Artigo em Inglês | MEDLINE | ID: mdl-33157015

RESUMO

Although TP53 is the most commonly mutated gene in human cancers, the p53-dependent transcriptional programs mediating tumor suppression remain incompletely understood. Here, to uncover critical components downstream of p53 in tumor suppression, we perform unbiased RNAi and CRISPR-Cas9-based genetic screens in vivo. These screens converge upon the p53-inducible gene Zmat3, encoding an RNA-binding protein, and we demonstrate that ZMAT3 is an important tumor suppressor downstream of p53 in mouse KrasG12D-driven lung and liver cancers and human carcinomas. Integrative analysis of the ZMAT3 RNA-binding landscape and transcriptomic profiling reveals that ZMAT3 directly modulates exon inclusion in transcripts encoding proteins of diverse functions, including the p53 inhibitors MDM4 and MDM2, splicing regulators, and components of varied cellular processes. Interestingly, these exons are enriched in NMD signals, and, accordingly, ZMAT3 broadly affects target transcript stability. Collectively, these studies reveal ZMAT3 as a novel RNA-splicing and homeostasis regulator and a key component of p53-mediated tumor suppression.


Assuntos
Proteínas de Ligação a RNA/genética , Proteína Supressora de Tumor p53/genética , Adenocarcinoma/genética , Processamento Alternativo , Animais , Proteínas de Ciclo Celular/metabolismo , Éxons , Perfilação da Expressão Gênica/métodos , Genes Supressores de Tumor , Humanos , Neoplasias Hepáticas/genética , Masculino , Camundongos , Camundongos Endogâmicos ICR , Camundongos SCID , Interferência de RNA , Splicing de RNA , Proteínas de Ligação a RNA/metabolismo , Proteína Supressora de Tumor p53/metabolismo
5.
Nature ; 592(7856): 794-798, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33854239

RESUMO

The initiation of cell division integrates a large number of intra- and extracellular inputs. D-type cyclins (hereafter, cyclin D) couple these inputs to the initiation of DNA replication1. Increased levels of cyclin D promote cell division by activating cyclin-dependent kinases 4 and 6 (hereafter, CDK4/6), which in turn phosphorylate and inactivate the retinoblastoma tumour suppressor. Accordingly, increased levels and activity of cyclin D-CDK4/6 complexes are strongly linked to unchecked cell proliferation and cancer2,3. However, the mechanisms that regulate levels of cyclin D are incompletely understood4,5. Here we show that autophagy and beclin 1 regulator 1 (AMBRA1) is the main regulator of the degradation of cyclin D. We identified AMBRA1 in a genome-wide screen to investigate the genetic basis of  the response to CDK4/6 inhibition. Loss of AMBRA1 results in high levels of cyclin D in cells and in mice, which promotes proliferation and decreases sensitivity to CDK4/6 inhibition. Mechanistically, AMBRA1 mediates ubiquitylation and proteasomal degradation of cyclin D as a substrate receptor for the cullin 4 E3 ligase complex. Loss of AMBRA1 enhances the growth of lung adenocarcinoma in a mouse model, and low levels of AMBRA1 correlate with worse survival in patients with lung adenocarcinoma. Thus, AMBRA1 regulates cellular levels of cyclin D, and contributes to cancer development and the response of cancer cells to CDK4/6 inhibitors.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Ciclina D/metabolismo , Adenocarcinoma de Pulmão/genética , Animais , Divisão Celular , Quinase 4 Dependente de Ciclina/antagonistas & inibidores , Quinase 4 Dependente de Ciclina/metabolismo , Quinase 6 Dependente de Ciclina/antagonistas & inibidores , Quinase 6 Dependente de Ciclina/metabolismo , Genes Supressores de Tumor , Humanos , Neoplasias Pulmonares/genética , Camundongos , Piperazinas/farmacologia , Piridinas/farmacologia , Células U937 , Ubiquitinação
6.
Nat Methods ; 20(7): 1070-1081, 2023 07.
Artigo em Inglês | MEDLINE | ID: mdl-37291262

RESUMO

The development of transgenic mouse models that express genes of interest in specific cell types has transformed our understanding of basic biology and disease. However, generating these models is time- and resource-intensive. Here we describe a model system, SELective Expression and Controlled Transduction In Vivo (SELECTIV), that enables efficient and specific expression of transgenes by coupling adeno-associated virus (AAV) vectors with Cre-inducible overexpression of the multi-serotype AAV receptor, AAVR. We demonstrate that transgenic AAVR overexpression greatly increases the efficiency of transduction of many diverse cell types, including muscle stem cells, which are normally refractory to AAV transduction. Superior specificity is achieved by combining Cre-mediated AAVR overexpression with whole-body knockout of endogenous Aavr, which is demonstrated in heart cardiomyocytes, liver hepatocytes and cholinergic neurons. The enhanced efficacy and exquisite specificity of SELECTIV has broad utility in development of new mouse model systems and expands the use of AAV for gene delivery in vivo.


Assuntos
Técnicas de Transferência de Genes , Vetores Genéticos , Camundongos , Animais , Vetores Genéticos/genética , Camundongos Transgênicos , Terapia Genética , Transgenes , Dependovirus/genética , Transdução Genética
7.
Nature ; 580(7801): 136-141, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-32238925

RESUMO

Cancer genomics studies have identified thousands of putative cancer driver genes1. Development of high-throughput and accurate models to define the functions of these genes is a major challenge. Here we devised a scalable cancer-spheroid model and performed genome-wide CRISPR screens in 2D monolayers and 3D lung-cancer spheroids. CRISPR phenotypes in 3D more accurately recapitulated those of in vivo tumours, and genes with differential sensitivities between 2D and 3D conditions were highly enriched for genes that are mutated in lung cancers. These analyses also revealed drivers that are essential for cancer growth in 3D and in vivo, but not in 2D. Notably, we found that carboxypeptidase D is responsible for removal of a C-terminal RKRR motif2 from the α-chain of the insulin-like growth factor 1 receptor that is critical for receptor activity. Carboxypeptidase D expression correlates with patient outcomes in patients with lung cancer, and loss of carboxypeptidase D reduced tumour growth. Our results reveal key differences between 2D and 3D cancer models, and establish a generalizable strategy for performing CRISPR screens in spheroids to reveal cancer vulnerabilities.


Assuntos
Sistemas CRISPR-Cas/genética , Técnicas de Cultura de Células/métodos , Proliferação de Células/genética , Genoma Humano/genética , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patologia , Esferoides Celulares/patologia , Adenocarcinoma/genética , Adenocarcinoma/metabolismo , Adenocarcinoma/patologia , Motivos de Aminoácidos , Animais , Carboxipeptidases/antagonistas & inibidores , Carboxipeptidases/deficiência , Carboxipeptidases/genética , Carboxipeptidases/metabolismo , Feminino , Humanos , Neoplasias Pulmonares/metabolismo , Camundongos , Terapia de Alvo Molecular , Mutação , Fenótipo , Receptor IGF Tipo 1/química , Receptor IGF Tipo 1/metabolismo , Transdução de Sinais , Esferoides Celulares/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
8.
Proc Natl Acad Sci U S A ; 120(38): e2303224120, 2023 09 19.
Artigo em Inglês | MEDLINE | ID: mdl-37695905

RESUMO

Cancer genomes are almost invariably complex with genomic alterations cooperating during each step of carcinogenesis. In cancers that lack a single dominant oncogene mutation, cooperation between the inactivation of multiple tumor suppressor genes can drive tumor initiation and growth. Here, we shed light on how the sequential acquisition of genomic alterations generates oncogene-negative lung tumors. We couple tumor barcoding with combinatorial and multiplexed somatic genome editing to characterize the fitness landscapes of three tumor suppressor genes NF1, RASA1, and PTEN, the inactivation of which jointly drives oncogene-negative lung adenocarcinoma initiation and growth. The fitness landscape was surprisingly accessible, with each additional mutation leading to growth advantage. Furthermore, the fitness landscapes remained fully accessible across backgrounds with the inactivation of additional tumor suppressor genes. These results suggest that while predicting cancer evolution will be challenging, acquiring the multiple alterations that drive the growth of oncogene-negative tumors can be facilitated by the lack of constraints on mutational order.


Assuntos
Adenocarcinoma de Pulmão , Neoplasias Pulmonares , Humanos , Oncogenes/genética , Adenocarcinoma de Pulmão/genética , Mutação , Neoplasias Pulmonares/genética , Transformação Celular Neoplásica , Proteína p120 Ativadora de GTPase
9.
J Biol Chem ; : 107859, 2024 Oct 05.
Artigo em Inglês | MEDLINE | ID: mdl-39374781

RESUMO

RAS clustering at the cell membrane is critical to activate signaling in cells, but whether this clustering is mediated exclusively by its c-terminal hypervariable region, receives contributions from the G-domain of RAS, and/or is influenced by secondary effectors has been intensely debated. Reports that G-domain mutations do not modulate RAS-RAS interactions, have led some to question the validity of previous experiments that indicate the G-domain plays a role in RAS clustering/interactions. Here we reconcile these findings by clarifying the impact of experimental variables, such as protein expression levels, cellular context, RAS zygosity, and secondary effector interactions on RAS clustering. Lack of control over these variables impact the results using G-domain mutations across various assay systems and can lead to unsound conclusions.

10.
Nat Rev Genet ; 19(12): 801, 2018 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-30327497

RESUMO

The originally published article failed to acknowledge the equal first authorship contribution of I. P. Winters and C. W. Murray. The article has now been corrected online. The editors apologize for this error.

11.
Nat Rev Genet ; 19(12): 741-755, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30267031

RESUMO

Large-scale sequencing of human tumours has uncovered a vast array of genomic alterations. Genetically engineered mouse models recapitulate many features of human cancer and have been instrumental in assigning biological meaning to specific cancer-associated alterations. However, their time, cost and labour-intensive nature limits their broad utility; thus, the functional importance of the majority of genomic aberrations in cancer remains unknown. Recent advances have accelerated the functional interrogation of cancer-associated alterations within in vivo models. Specifically, the past few years have seen the emergence of CRISPR-Cas9-based strategies to rapidly generate increasingly complex somatic alterations and the development of multiplexed and quantitative approaches to ascertain gene function in vivo.


Assuntos
Sistemas CRISPR-Cas , Genes Neoplásicos , Genômica/métodos , Neoplasias/genética , Animais , Humanos , Camundongos , Neoplasias/metabolismo , Neoplasias/patologia
12.
Cell ; 132(5): 875-86, 2008 Mar 07.
Artigo em Inglês | MEDLINE | ID: mdl-18329372

RESUMO

miR-17 approximately 92, miR-106b approximately 25, and miR-106a approximately 363 belong to a family of highly conserved miRNA clusters. Amplification and overexpression of miR-1792 is observed in human cancers, and its oncogenic properties have been confirmed in a mouse model of B cell lymphoma. Here we show that mice deficient for miR-17 approximately 92 die shortly after birth with lung hypoplasia and a ventricular septal defect. The miR-17 approximately 92 cluster is also essential for B cell development. Absence of miR-17 approximately 92 leads to increased levels of the proapoptotic protein Bim and inhibits B cell development at the pro-B to pre-B transition. Furthermore, while ablation of miR-106b approximately 25 or miR-106a approximately 363 has no obvious phenotypic consequences, compound mutant embryos lacking both miR-106b approximately 25 and miR-17 approximately 92 die at midgestation. These results provide key insights into the physiologic functions of this family of microRNAs and suggest a link between the oncogenic properties of miR-17 approximately 92 and its functions during B lymphopoiesis and lung development.


Assuntos
MicroRNAs/genética , MicroRNAs/metabolismo , Família Multigênica , Deleção de Sequência , Regiões 3' não Traduzidas/metabolismo , Animais , Proteínas Reguladoras de Apoptose/metabolismo , Linfócitos B/citologia , Proteína 11 Semelhante a Bcl-2 , Sobrevivência Celular , Células-Tronco Embrionárias/metabolismo , Feto/citologia , Genes Letais , Comunicação Interventricular/genética , Pneumopatias/genética , Proteínas de Membrana/metabolismo , Camundongos , Proteínas Proto-Oncogênicas/metabolismo
13.
Nucleic Acids Res ; 49(6): e36, 2021 04 06.
Artigo em Inglês | MEDLINE | ID: mdl-33619540

RESUMO

Several existing technologies enable short genomic alterations including generating indels and short nucleotide variants, however, engineering more significant genomic changes is more challenging due to reduced efficiency and precision. Here, we developed RecT Editor via Designer-Cas9-Initiated Targeting (REDIT), which leverages phage single-stranded DNA-annealing proteins (SSAP) RecT for mammalian genome engineering. Relative to Cas9-mediated homology-directed repair (HDR), REDIT yielded up to a 5-fold increase of efficiency to insert kilobase-scale exogenous sequences at defined genomic regions. We validated our REDIT approach using different formats and lengths of knock-in templates. We further demonstrated that REDIT tools using Cas9 nickase have efficient gene-editing activities and reduced off-target errors, measured using a combination of targeted sequencing, genome-wide indel, and insertion mapping assays. Our experiments inhibiting repair enzyme activities suggested that REDIT has the potential to overcome limitations of endogenous DNA repair steps. Finally, our REDIT method is applicable across cell types including human stem cells, and is generalizable to different Cas9 enzymes.


Assuntos
Proteína 9 Associada à CRISPR , Proteínas de Ligação a DNA , Proteínas de Escherichia coli , Edição de Genes/métodos , Linhagem Celular , Genoma , Humanos , Reparo de DNA por Recombinação , Células-Tronco/metabolismo
14.
Genes Dev ; 29(14): 1576-85, 2015 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-26178787

RESUMO

Pancreatic ductal adenocarcinoma (PDAC) is a genomically diverse, prevalent, and almost invariably fatal malignancy. Although conventional genetically engineered mouse models of human PDAC have been instrumental in understanding pancreatic cancer development, these models are much too labor-intensive, expensive, and slow to perform the extensive molecular analyses needed to adequately understand this disease. Here we demonstrate that retrograde pancreatic ductal injection of either adenoviral-Cre or lentiviral-Cre vectors allows titratable initiation of pancreatic neoplasias that progress into invasive and metastatic PDAC. To enable in vivo CRISPR/Cas9-mediated gene inactivation in the pancreas, we generated a Cre-regulated Cas9 allele and lentiviral vectors that express Cre and a single-guide RNA. CRISPR-mediated targeting of Lkb1 in combination with oncogenic Kras expression led to selection for inactivating genomic alterations, absence of Lkb1 protein, and rapid tumor growth that phenocopied Cre-mediated genetic deletion of Lkb1. This method will transform our ability to rapidly interrogate gene function during the development of this recalcitrant cancer.


Assuntos
Adenocarcinoma/fisiopatologia , Carcinoma Ductal Pancreático/fisiopatologia , Modelos Animais de Doenças , Adenocarcinoma/genética , Animais , Carcinoma Ductal Pancreático/genética , Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas , Regulação Neoplásica da Expressão Gênica , Vetores Genéticos/genética , Genoma/genética , Humanos , Lentivirus/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos
15.
Mol Cell ; 55(3): 436-50, 2014 Aug 07.
Artigo em Inglês | MEDLINE | ID: mdl-25042806

RESUMO

The serine/threonine kinase LKB1 is a tumor suppressor whose loss is associated with increased metastatic potential. In an effort to define biochemical signatures of metastasis associated with LKB1 loss, we discovered that the epithelial-to-mesenchymal transition transcription factor Snail1 was uniquely upregulated upon LKB1 deficiency across cell types. The ability of LKB1 to suppress Snail1 levels was independent of AMPK but required the related kinases MARK1 and MARK4. In a screen for substrates of these kinases involved in Snail regulation, we identified the scaffolding protein DIXDC1. Similar to loss of LKB1, DIXDC1 depletion results in upregulation of Snail1 in a FAK-dependent manner, leading to increased cell invasion. MARK1 phosphorylation of DIXDC1 is required for its localization to focal adhesions and ability to suppress metastasis in mice. DIXDC1 is frequently downregulated in human cancers, which correlates with poor survival. This study defines an AMPK-independent phosphorylation cascade essential for LKB1-dependent control of metastatic behavior.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/química , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteínas dos Microfilamentos/química , Proteínas dos Microfilamentos/metabolismo , Invasividade Neoplásica/genética , Proteínas Serina-Treonina Quinases/metabolismo , Quinases Proteína-Quinases Ativadas por AMP , Animais , Linhagem Celular Tumoral , Transição Epitelial-Mesenquimal/genética , Transição Epitelial-Mesenquimal/fisiologia , Regulação Neoplásica da Expressão Gênica , Células HEK293 , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/genética , Neoplasias Pulmonares , Camundongos , Proteínas dos Microfilamentos/genética , Invasividade Neoplásica/patologia , Fosforilação , Proteínas Serina-Treonina Quinases/genética , Transdução de Sinais , Fatores de Transcrição da Família Snail , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
16.
Actas Dermosifiliogr ; 113(10): 961-965, 2022.
Artigo em Inglês, Espanhol | MEDLINE | ID: mdl-35526565

RESUMO

Dedicated dermoscopy units assess individuals at high risk for melanoma. Understanding the reasons for referral to these units and the epidemiological profile of referred patients can help optimize health care resources and determine who benefits most from dermoscopic evaluation. We analyzed reasons for referral and epidemiological characteristics of 413 patients with at least 1 high-risk factor for melanoma seen at a dedicated dermoscopy unit over a period of 10 years. We also analyzed the number of necessary excisions (NNE) for each melanoma diagnosed, histologic features, and associations between nonenvironmental factors and diagnosis. The main reasons for referral were a past history of melanoma (21.5%), changes detected by the patient or a relative (20%), clinical and/or dermoscopic findings suggestive of malignancy (19.4%), and a family history of melanoma (17.4%). Seventy-six of the 178 excised lesions were melanomas (NNE per melanoma detected, 2.34). Older age was the only risk factor significantly associated with the development of melanoma.


Assuntos
Melanoma , Nevo Pigmentado , Neoplasias Cutâneas , Humanos , Dermoscopia , Nevo Pigmentado/patologia , Neoplasias Cutâneas/diagnóstico , Neoplasias Cutâneas/epidemiologia , Neoplasias Cutâneas/patologia , Centros de Atenção Terciária , Estudos Retrospectivos , Melanoma/diagnóstico , Melanoma/epidemiologia , Melanoma/patologia , Encaminhamento e Consulta
17.
Genes Dev ; 27(14): 1557-67, 2013 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-23873940

RESUMO

Metastasis accounts for the vast majority of cancer-related deaths, yet the molecular mechanisms that drive metastatic spread remain poorly understood. Here we report that Tks5, which has been linked to the formation of proteolytic cellular protrusions known as invadopodia, undergoes an isoform switch during metastatic progression in a genetically engineered mouse model of lung adenocarcinoma. Nonmetastatic primary tumor-derived cells predominantly expressed a short isoform, Tks5short, while metastatic primary tumor- and metastasis-derived cells acquired increased expression of the full-length isoform Tks5long. This elevation of Tks5long to Tks5short ratio correlated with a commensurate increase in invadopodia activity in metastatic cells compared with nonmetastatic cells. Further characterization of these isoforms by knockdown and overexpression experiments demonstrated that Tks5long promoted invadopodia in vitro and increased metastasis in transplant models and an autochthonous model of lung adenocarcinoma. Conversely, Tks5short decreased invadopodia stability and proteolysis, acting as a natural dominant-negative inhibitor to Tks5long. Importantly, high Tks5long and low Tks5short expressions in human lung adenocarcinomas correlated with metastatic disease and predicted worse survival of early stage patients. These data indicate that tipping the Tks5 isoform balance to a high Tks5long to Tks5short ratio promotes invadopodia-mediated invasion and metastasis.


Assuntos
Proteínas Adaptadoras de Transporte Vesicular/genética , Adenocarcinoma/genética , Adenocarcinoma/patologia , Regulação Neoplásica da Expressão Gênica , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patologia , Fosfoproteínas/genética , Adenocarcinoma/mortalidade , Adenocarcinoma de Pulmão , Animais , Linhagem Celular Tumoral , Técnicas de Silenciamento de Genes , Humanos , Neoplasias Pulmonares/mortalidade , Camundongos , Camundongos Nus , Invasividade Neoplásica/genética , Metástase Neoplásica/genética , Proteínas de Ligação a Fosfato , Isoformas de Proteínas , Análise de Sobrevida
18.
Nat Methods ; 14(7): 737-742, 2017 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-28530655

RESUMO

Cancer growth is a multistage, stochastic evolutionary process. While cancer genome sequencing has been instrumental in identifying the genomic alterations that occur in human tumors, the consequences of these alterations on tumor growth remain largely unexplored. Conventional genetically engineered mouse models enable the study of tumor growth in vivo, but they are neither readily scalable nor sufficiently quantitative to unravel the magnitude and mode of action of many tumor-suppressor genes. Here, we present a method that integrates tumor barcoding with ultradeep barcode sequencing (Tuba-seq) to interrogate tumor-suppressor function in mouse models of human cancer. Tuba-seq uncovers genotype-dependent distributions of tumor sizes. By combining Tuba-seq with multiplexed CRISPR-Cas9-mediated genome editing, we quantified the effects of 11 tumor-suppressor pathways that are frequently altered in human lung adenocarcinoma. Tuba-seq enables the broad quantification of the function of tumor-suppressor genes with unprecedented resolution, parallelization, and precision.


Assuntos
Neoplasias Experimentais/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Adenocarcinoma/genética , Animais , DNA/genética , DNA/isolamento & purificação , DNA/metabolismo , Código de Barras de DNA Taxonômico , Feminino , Engenharia Genética , Humanos , Lentivirus/genética , Pulmão/metabolismo , Neoplasias Pulmonares/genética , Masculino , Camundongos , Modelos Genéticos , Plasmídeos , Proteínas Proto-Oncogênicas p21(ras)/genética , Proteínas Proto-Oncogênicas p21(ras)/metabolismo , Proteínas Supressoras de Tumor/genética
19.
Nature ; 505(7482): 212-7, 2014 Jan 09.
Artigo em Inglês | MEDLINE | ID: mdl-24305048

RESUMO

Non-small-cell lung cancer (NSCLC) is the most prevalent histological cancer subtype worldwide. As the majority of patients present with invasive, metastatic disease, it is vital to understand the basis for lung cancer progression. Hmga2 is highly expressed in metastatic lung adenocarcinoma, in which it contributes to cancer progression and metastasis. Here we show that Hmga2 promotes lung cancer progression in mouse and human cells by operating as a competing endogenous RNA (ceRNA) for the let-7 microRNA (miRNA) family. Hmga2 can promote the transformation of lung cancer cells independent of protein-coding function but dependent upon the presence of let-7 sites; this occurs without changes in the levels of let-7 isoforms, suggesting that Hmga2 affects let-7 activity by altering miRNA targeting. These effects are also observed in vivo, where Hmga2 ceRNA activity drives lung cancer growth, invasion and dissemination. Integrated analysis of miRNA target prediction algorithms and metastatic lung cancer gene expression data reveals the TGF-ß co-receptor Tgfbr3 (ref. 12) as a putative target of Hmga2 ceRNA function. Tgfbr3 expression is regulated by the Hmga2 ceRNA through differential recruitment to Argonaute 2 (Ago2), and TGF-ß signalling driven by Tgfbr3 is important for Hmga2 to promote lung cancer progression. Finally, analysis of NSCLC-patient gene-expression data reveals that HMGA2 and TGFBR3 are coordinately regulated in NSCLC-patient material, a vital corollary to ceRNA function. Taken together, these results suggest that Hmga2 promotes lung carcinogenesis both as a protein-coding gene and as a non-coding RNA; such dual-function regulation of gene-expression networks reflects a novel means by which oncogenes promote disease progression.


Assuntos
Progressão da Doença , Proteína HMGA2/genética , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patologia , Animais , Proteínas Argonautas/metabolismo , Ligação Competitiva/genética , Carcinoma Pulmonar de Células não Pequenas/genética , Carcinoma Pulmonar de Células não Pequenas/patologia , Linhagem Celular Tumoral , Proliferação de Células , Modelos Animais de Doenças , Regulação Neoplásica da Expressão Gênica/genética , Humanos , Camundongos , MicroRNAs/genética , MicroRNAs/metabolismo , Invasividade Neoplásica/genética , Metástase Neoplásica/genética , Proteoglicanas/biossíntese , Proteoglicanas/deficiência , Proteoglicanas/genética , Isoformas de RNA/genética , Isoformas de RNA/metabolismo , Receptores de Fatores de Crescimento Transformadores beta/biossíntese , Receptores de Fatores de Crescimento Transformadores beta/deficiência , Receptores de Fatores de Crescimento Transformadores beta/genética , Transcrição Gênica/genética , Fator de Crescimento Transformador beta/metabolismo
20.
Proc Natl Acad Sci U S A ; 114(28): E5625-E5634, 2017 07 11.
Artigo em Inglês | MEDLINE | ID: mdl-28652369

RESUMO

The extracellular microenvironment is an integral component of normal and diseased tissues that is poorly understood owing to its complexity. To investigate the contribution of the microenvironment to lung fibrosis and adenocarcinoma progression, two pathologies characterized by excessive stromal expansion, we used mouse models to characterize the extracellular matrix (ECM) composition of normal lung, fibrotic lung, lung tumors, and metastases. Using quantitative proteomics, we identified and assayed the abundance of 113 ECM proteins, which revealed robust ECM protein signatures unique to fibrosis, primary tumors, or metastases. These analyses indicated significantly increased abundance of several S100 proteins, including Fibronectin and Tenascin-C (Tnc), in primary lung tumors and associated lymph node metastases compared with normal tissue. We further showed that Tnc expression is repressed by the transcription factor Nkx2-1, a well-established suppressor of metastatic progression. We found that increasing the levels of Tnc, via CRISPR-mediated transcriptional activation of the endogenous gene, enhanced the metastatic dissemination of lung adenocarcinoma cells. Interrogation of human cancer gene expression data revealed that high TNC expression correlates with worse prognosis for lung adenocarcinoma, and that a three-gene expression signature comprising TNC, S100A10, and S100A11 is a robust predictor of patient survival independent of age, sex, smoking history, and mutational load. Our findings suggest that the poorly understood ECM composition of the fibrotic and tumor microenvironment is an underexplored source of diagnostic markers and potential therapeutic targets for cancer patients.


Assuntos
Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/mortalidade , Proteômica/métodos , Tenascina/fisiologia , Adenocarcinoma/metabolismo , Animais , Anexina A2/metabolismo , Sistemas CRISPR-Cas , Progressão da Doença , Matriz Extracelular/metabolismo , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Neoplasias Pulmonares/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Análise Multivariada , Metástase Neoplásica , Prognóstico , Proteínas S100/metabolismo , Fator Nuclear 1 de Tireoide/metabolismo , Resultado do Tratamento , Microambiente Tumoral
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa