Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Cancer Discov ; 12(2): 562-585, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34561242

RESUMO

SMARCA4/BRG1 encodes for one of two mutually exclusive ATPases present in mammalian SWI/SNF chromatin remodeling complexes and is frequently mutated in human lung adenocarcinoma. However, the functional consequences of SMARCA4 mutation on tumor initiation, progression, and chromatin regulation in lung cancer remain poorly understood. Here, we demonstrate that loss of Smarca4 sensitizes club cell secretory protein-positive cells within the lung in a cell type-dependent fashion to malignant transformation and tumor progression, resulting in highly advanced dedifferentiated tumors and increased metastatic incidence. Consistent with these phenotypes, Smarca4-deficient primary tumors lack lung lineage transcription factor activities and resemble a metastatic cell state. Mechanistically, we show that Smarca4 loss impairs the function of all three classes of SWI/SNF complexes, resulting in decreased chromatin accessibility at lung lineage motifs and ultimately accelerating tumor progression. Thus, we propose that the SWI/SNF complex via Smarca4 acts as a gatekeeper for lineage-specific cellular transformation and metastasis during lung cancer evolution. SIGNIFICANCE: We demonstrate cell-type specificity in the tumor-suppressive functions of SMARCA4 in the lung, pointing toward a critical role of the cell-of-origin in driving SWI/SNF-mutant lung adenocarcinoma. We further show the direct effects of SMARCA4 loss on SWI/SNF function and chromatin regulation that cause aggressive malignancy during lung cancer evolution.This article is highlighted in the In This Issue feature, p. 275.


Assuntos
Adenocarcinoma de Pulmão/genética , Transformação Celular Neoplásica , DNA Helicases/genética , Neoplasias Pulmonares/genética , Metástase Neoplásica , Proteínas Nucleares/genética , Fatores de Transcrição/genética , Adenocarcinoma de Pulmão/secundário , Animais , Modelos Animais de Doenças , Humanos , Neoplasias Pulmonares/patologia , Camundongos
2.
Cancer Res ; 80(18): 3841-3854, 2020 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-32690724

RESUMO

Inactivation of SMARCA4/BRG1, the core ATPase subunit of mammalian SWI/SNF complexes, occurs at very high frequencies in non-small cell lung cancers (NSCLC). There are no targeted therapies for this subset of lung cancers, nor is it known how mutations in BRG1 contribute to lung cancer progression. Using a combination of gain- and loss-of-function approaches, we demonstrate that deletion of BRG1 in lung cancer leads to activation of replication stress responses. Single-molecule assessment of replication fork dynamics in BRG1-deficient cells revealed increased origin firing mediated by the prelicensing protein, CDC6. Quantitative mass spectrometry and coimmunoprecipitation assays showed that BRG1-containing SWI/SNF complexes interact with RPA complexes. Finally, BRG1-deficient lung cancers were sensitive to pharmacologic inhibition of ATR. These findings provide novel mechanistic insight into BRG1-mutant lung cancers and suggest that their dependency on ATR can be leveraged therapeutically and potentially expanded to BRG1-mutant cancers in other tissues. SIGNIFICANCE: These findings indicate that inhibition of ATR is a promising therapy for the 10% of non-small cell lung cancer patients harboring mutations in SMARCA4/BRG1. GRAPHICAL ABSTRACT: http://cancerres.aacrjournals.org/content/canres/80/18/3841/F1.large.jpg.


Assuntos
Proteínas Mutadas de Ataxia Telangiectasia/antagonistas & inibidores , Carcinoma Pulmonar de Células não Pequenas/genética , DNA Helicases/genética , Deleção de Genes , Neoplasias Pulmonares/genética , Proteínas Nucleares/genética , Fatores de Transcrição/genética , Animais , Proteínas de Ciclo Celular/metabolismo , Proteínas Cromossômicas não Histona , DNA Helicases/deficiência , Progressão da Doença , Feminino , Fatores de Transcrição Forkhead , Edição de Genes , Humanos , Imunoprecipitação , Espectrometria de Massas , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Nus , Proteínas Nucleares/deficiência , Proteínas Nucleares/metabolismo , Análise de Sequência de RNA , Fatores de Transcrição/deficiência
3.
Am J Respir Cell Mol Biol ; 59(2): 237-245, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29447458

RESUMO

Metastatic disease is the primary cause of death of patients with lung cancer, but the mouse models of lung adenocarcinoma do not accurately recapitulate the tumor microenvironment or metastatic disease observed in patients. In this study, we conditionally deleted E-cadherin in an autochthonous lung adenocarcinoma mouse model driven by activated oncogenic Kras and p53 loss. Loss of E-cadherin significantly accelerated lung adenocarcinoma progression and decreased survival of the mice. Kras;p53;E-cadherin mice had a 41% lung tumor burden, invasive grade 4 tumors, and a desmoplastic stroma just 8 weeks after tumor initiation. One hundred percent of the mice developed local metastases to the lymph nodes or chest wall, and 38% developed distant metastases to the liver or kidney. Lung adenocarcinoma cancer cell lines derived from these tumors also had high migratory rates. These studies demonstrate that the Kras;p53;E-cadherin mouse model better emulates the tumor microenvironment and metastases observed in patients with lung adenocarcinoma than previous models and may therefore be useful for studying metastasis and testing new lung cancer treatments in vivo.


Assuntos
Adenocarcinoma/patologia , Caderinas/metabolismo , Neoplasias Pulmonares/patologia , Metástase Neoplásica , Proteínas Proto-Oncogênicas p21(ras)/metabolismo , Adenocarcinoma/genética , Animais , Modelos Animais de Doenças , Neoplasias Hepáticas/patologia , Neoplasias Pulmonares/genética , Camundongos Endogâmicos C57BL , Metástase Neoplásica/genética , Proteínas Proto-Oncogênicas p21(ras)/genética
4.
Mol Ther ; 25(9): 1999-2013, 2017 09 06.
Artigo em Inglês | MEDLINE | ID: mdl-28619647

RESUMO

Patient-derived induced pluripotent stem cells (iPSCs) hold great promise for autologous cell replacement. However, for many inherited diseases, treatment will likely require genetic repair pre-transplantation. Genome editing technologies are useful for this application. The purpose of this study was to develop CRISPR-Cas9-mediated genome editing strategies to target and correct the three most common types of disease-causing variants in patient-derived iPSCs: (1) exonic, (2) deep intronic, and (3) dominant gain of function. We developed a homology-directed repair strategy targeting a homozygous Alu insertion in exon 9 of male germ cell-associated kinase (MAK) and demonstrated restoration of the retinal transcript and protein in patient cells. We generated a CRISPR-Cas9-mediated non-homologous end joining (NHEJ) approach to excise a major contributor to Leber congenital amaurosis, the IVS26 cryptic-splice mutation in CEP290, and demonstrated correction of the transcript and protein in patient iPSCs. Lastly, we designed allele-specific CRISPR guides that selectively target the mutant Pro23His rhodopsin (RHO) allele, which, following delivery to both patient iPSCs in vitro and pig retina in vivo, created a frameshift and premature stop that would prevent transcription of the disease-causing variant. The strategies developed in this study will prove useful for correcting a wide range of genetic variants in genes that cause inherited retinal degeneration.


Assuntos
Sistemas CRISPR-Cas , Edição de Genes , Marcação de Genes , Células-Tronco Pluripotentes Induzidas/metabolismo , Degeneração Retiniana/genética , Transplante de Células-Tronco , Alelos , Animais , Linhagem Celular , Ordem dos Genes , Loci Gênicos , Terapia Genética , Vetores Genéticos/genética , Recombinação Homóloga , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Íntrons , Mutação , Proteínas Serina-Treonina Quinases/genética , RNA Guia de Cinetoplastídeos , Degeneração Retiniana/terapia , Transplante de Células-Tronco/métodos , Transplante Autólogo
6.
Nat Commun ; 8: 14922, 2017 04 07.
Artigo em Inglês | MEDLINE | ID: mdl-28387316

RESUMO

Adenosquamous lung tumours, which are extremely poor prognosis, may result from cellular plasticity. Here, we demonstrate lineage switching of KRAS+ lung adenocarcinomas (ADC) to squamous cell carcinoma (SCC) through deletion of Lkb1 (Stk11) in autochthonous and transplant models. Chromatin analysis reveals loss of H3K27me3 and gain of H3K27ac and H3K4me3 at squamous lineage genes, including Sox2, ΔNp63 and Ngfr. SCC lesions have higher levels of the H3K27 methyltransferase EZH2 than the ADC lesions, but there is a clear lack of the essential Polycomb Repressive Complex 2 (PRC2) subunit EED in the SCC lesions. The pattern of high EZH2, but low H3K27me3 mark, is also prevalent in human lung SCC and SCC regions within ADSCC tumours. Using FACS-isolated populations, we demonstrate that bronchioalveolar stem cells and club cells are the likely cells-of-origin for SCC transitioned tumours. These findings shed light on the epigenetics and cellular origins of lineage-specific lung tumours.


Assuntos
Adenocarcinoma/genética , Carcinoma de Células Escamosas/genética , Neoplasias Pulmonares/genética , Complexo Repressor Polycomb 2/genética , Proteínas Serina-Treonina Quinases/genética , Proteínas Quinases Ativadas por AMP , Adenocarcinoma/metabolismo , Adenocarcinoma/patologia , Animais , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patologia , Proteína Potenciadora do Homólogo 2 de Zeste/genética , Proteína Potenciadora do Homólogo 2 de Zeste/metabolismo , Perfilação da Expressão Gênica/métodos , Regulação Neoplásica da Expressão Gênica , Histonas/metabolismo , Estimativa de Kaplan-Meier , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patologia , Metilação , Camundongos da Linhagem 129 , Camundongos Knockout , Complexo Repressor Polycomb 2/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas p21(ras)/genética , Proteínas Proto-Oncogênicas p21(ras)/metabolismo , Células Tumorais Cultivadas
7.
Sci Transl Med ; 9(376)2017 02 08.
Artigo em Inglês | MEDLINE | ID: mdl-28179501

RESUMO

Diamond-Blackfan anemia (DBA) is a congenital disorder characterized by the failure of erythroid progenitor differentiation, severely curtailing red blood cell production. Because many DBA patients fail to respond to corticosteroid therapy, there is considerable need for therapeutics for this disorder. Identifying therapeutics for DBA requires circumventing the paucity of primary patient blood stem and progenitor cells. To this end, we adopted a reprogramming strategy to generate expandable hematopoietic progenitor cells from induced pluripotent stem cells (iPSCs) from DBA patients. Reprogrammed DBA progenitors recapitulate defects in erythroid differentiation, which were rescued by gene complementation. Unbiased chemical screens identified SMER28, a small-molecule inducer of autophagy, which enhanced erythropoiesis in a range of in vitro and in vivo models of DBA. SMER28 acted through autophagy factor ATG5 to stimulate erythropoiesis and up-regulate expression of globin genes. These findings present an unbiased drug screen for hematological disease using iPSCs and identify autophagy as a therapeutic pathway in DBA.


Assuntos
Anemia de Diamond-Blackfan/tratamento farmacológico , Descoberta de Drogas , Células-Tronco Hematopoéticas/metabolismo , Compostos Alílicos/farmacologia , Anemia de Diamond-Blackfan/patologia , Antígenos CD34/metabolismo , Autofagia/efeitos dos fármacos , Proteína 5 Relacionada à Autofagia/metabolismo , Diferenciação Celular/efeitos dos fármacos , Reprogramação Celular , Células Eritroides/efeitos dos fármacos , Células Eritroides/patologia , Eritropoese/efeitos dos fármacos , Teste de Complementação Genética , Globinas/metabolismo , Transplante de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/efeitos dos fármacos , Humanos , Células-Tronco Pluripotentes Induzidas/efeitos dos fármacos , Células-Tronco Pluripotentes Induzidas/metabolismo , Quinazolinas/farmacologia
8.
Nat Biotechnol ; 32(11): 1151-1157, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25306246

RESUMO

The ability to differentiate human pluripotent stem cells into endothelial cells with properties of cord-blood endothelial colony-forming cells (CB-ECFCs) may enable the derivation of clinically relevant numbers of highly proliferative blood vessel-forming cells to restore endothelial function in patients with vascular disease. We describe a protocol to convert human induced pluripotent stem cells (hiPSCs) or embryonic stem cells (hESCs) into cells similar to CB-ECFCs at an efficiency of >10(8) ECFCs produced from each starting pluripotent stem cell. The CB-ECFC-like cells display a stable endothelial phenotype with high clonal proliferative potential and the capacity to form human vessels in mice and to repair the ischemic mouse retina and limb, and they lack teratoma formation potential. We identify Neuropilin-1 (NRP-1)-mediated activation of KDR signaling through VEGF165 as a critical mechanism for the emergence and maintenance of CB-ECFC-like cells.


Assuntos
Diferenciação Celular/genética , Células-Tronco Embrionárias/citologia , Células Endoteliais/citologia , Células-Tronco Pluripotentes/citologia , Animais , Proliferação de Células/genética , Células Endoteliais/metabolismo , Sangue Fetal/citologia , Humanos , Camundongos , Neuropilina-1/metabolismo , Células-Tronco/citologia , Fator A de Crescimento do Endotélio Vascular/metabolismo
9.
J Am Chem Soc ; 130(46): 15246-7, 2008 Nov 19.
Artigo em Inglês | MEDLINE | ID: mdl-18939837

RESUMO

A bis-phenanthroline [2]catenane copper complex, consisting of one olefinic macrocycle and one nonolefinic macrocycle, underwent an entropy-driven ring-opening olefin metathesis polymerization (ROMP) to provide a polypseudorotaxane. The polymerization featured an average degree of polymerization of ca. 63 wherein the polymer was effectively saturated with threaded macrocycles. Removal of the copper led to near complete release of the macrocycles from the polymer backbone.


Assuntos
Catenanos/química , Polímeros/química , Rotaxanos/química , Estrutura Molecular
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA