Your browser doesn't support javascript.
loading
Smarca4 Inactivation Promotes Lineage-Specific Transformation and Early Metastatic Features in the Lung.
Concepcion, Carla P; Ma, Sai; LaFave, Lindsay M; Bhutkar, Arjun; Liu, Manyuan; DeAngelo, Lydia P; Kim, Jonathan Y; Del Priore, Isabella; Schoenfeld, Adam J; Miller, Manon; Kartha, Vinay K; Westcott, Peter M K; Sánchez-Rivera, Francisco J; Meli, Kevin; Gupta, Manav; Bronson, Roderick T; Riely, Gregory J; Rekhtman, Natasha; Rudin, Charles M; Kim, Carla F; Regev, Aviv; Buenrostro, Jason D; Jacks, Tyler.
Afiliação
  • Concepcion CP; Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts.
  • Ma S; Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts.
  • LaFave LM; Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, Massachusetts.
  • Bhutkar A; Broad Institute of MIT and Harvard, Cambridge, Massachusetts.
  • Liu M; Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts.
  • DeAngelo LP; Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, Massachusetts.
  • Kim JY; Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts.
  • Del Priore I; Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts.
  • Schoenfeld AJ; Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts.
  • Miller M; Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts.
  • Kartha VK; Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts.
  • Westcott PMK; Thoracic Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York.
  • Sánchez-Rivera FJ; Department of Medicine, Weill Cornell Medical College, New York, New York.
  • Meli K; Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts.
  • Gupta M; Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, Massachusetts.
  • Bronson RT; Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts.
  • Riely GJ; Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, New York.
  • Rekhtman N; Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts.
  • Rudin CM; Stem Cell Program, Division of Hematology/Oncology and Division of Respiratory Disease, Boston Children's Hospital, Boston, Massachusetts.
  • Kim CF; Biological and Biomedical Sciences PhD Program, Harvard Medical School, Boston, Massachusetts.
  • Regev A; Department of Pathology, Harvard Medical School, Boston, Massachusetts.
  • Buenrostro JD; Thoracic Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York.
  • Jacks T; Department of Medicine, Weill Cornell Medical College, New York, New York.
Cancer Discov ; 12(2): 562-585, 2022 02.
Article em En | MEDLINE | ID: mdl-34561242
ABSTRACT
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

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Assunto principal: Fatores de Transcrição / Proteínas Nucleares / Transformação Celular Neoplásica / DNA Helicases / Adenocarcinoma de Pulmão / Neoplasias Pulmonares / Metástase Neoplásica Tipo de estudo: Prognostic_studies Limite: Animals / Humans Idioma: En Revista: Cancer Discov Ano de publicação: 2022 Tipo de documento: Article

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Assunto principal: Fatores de Transcrição / Proteínas Nucleares / Transformação Celular Neoplásica / DNA Helicases / Adenocarcinoma de Pulmão / Neoplasias Pulmonares / Metástase Neoplásica Tipo de estudo: Prognostic_studies Limite: Animals / Humans Idioma: En Revista: Cancer Discov Ano de publicação: 2022 Tipo de documento: Article