Your browser doesn't support javascript.
loading
A CRISPR/Cas9-Engineered ARID1A-Deficient Human Gastric Cancer Organoid Model Reveals Essential and Nonessential Modes of Oncogenic Transformation.
Lo, Yuan-Hung; Kolahi, Kevin S; Du, Yuhong; Chang, Chiung-Ying; Krokhotin, Andrey; Nair, Ajay; Sobba, Walter D; Karlsson, Kasper; Jones, Sunny J; Longacre, Teri A; Mah, Amanda T; Tercan, Bahar; Sockell, Alexandra; Xu, Hang; Seoane, Jose A; Chen, Jin; Shmulevich, Ilya; Weissman, Jonathan S; Curtis, Christina; Califano, Andrea; Fu, Haian; Crabtree, Gerald R; Kuo, Calvin J.
Afiliação
  • Lo YH; Department of Medicine, Division of Hematology, Stanford University School of Medicine, Stanford, California.
  • Kolahi KS; Department of Pathology, Stanford University School of Medicine, Stanford, California.
  • Du Y; Department of Pharmacology and Chemical Biology and Emory Chemical Biology Discovery Center, Emory University School of Medicine, Atlanta, Georgia.
  • Chang CY; Department of Pathology, Stanford University School of Medicine, Stanford, California.
  • Krokhotin A; Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, California.
  • Nair A; Department of Systems Biology, Columbia University, New York, New York.
  • Sobba WD; Department of Medicine, Division of Hematology, Stanford University School of Medicine, Stanford, California.
  • Karlsson K; Department of Medicine, Division of Hematology, Stanford University School of Medicine, Stanford, California.
  • Jones SJ; Division of Oncology, Stanford University School of Medicine, Stanford, California.
  • Longacre TA; Department of Systems Biology, Columbia University, New York, New York.
  • Mah AT; Department of Pathology, Stanford University School of Medicine, Stanford, California.
  • Tercan B; Department of Medicine, Division of Hematology, Stanford University School of Medicine, Stanford, California.
  • Sockell A; Institute for Systems Biology, Seattle, Washington.
  • Xu H; Division of Oncology, Stanford University School of Medicine, Stanford, California.
  • Seoane JA; Division of Oncology, Stanford University School of Medicine, Stanford, California.
  • Chen J; Division of Oncology, Stanford University School of Medicine, Stanford, California.
  • Shmulevich I; Howard Hughes Medical Institute, Department of Cellular and Molecular Pharmacology, University of California, San Francisco, California.
  • Weissman JS; Department of Pharmacology and Cecil H. and Ida Green Center for Reproductive Biology Sciences, The University of Texas Southwestern Medical Center, Dallas, Texas.
  • Curtis C; Institute for Systems Biology, Seattle, Washington.
  • Califano A; Howard Hughes Medical Institute, Department of Cellular and Molecular Pharmacology, University of California, San Francisco, California.
  • Fu H; Division of Oncology, Stanford University School of Medicine, Stanford, California.
  • Crabtree GR; Department of Systems Biology, Columbia University, New York, New York.
  • Kuo CJ; Department of Pharmacology and Chemical Biology and Emory Chemical Biology Discovery Center, Emory University School of Medicine, Atlanta, Georgia.
Cancer Discov ; 11(6): 1562-1581, 2021 06.
Article em En | MEDLINE | ID: mdl-33451982
ABSTRACT
Mutations in ARID1A rank among the most common molecular aberrations in human cancer. However, oncogenic consequences of ARID1A mutation in human cells remain poorly defined due to lack of forward genetic models. Here, CRISPR/Cas9-mediated ARID1A knockout (KO) in primary TP53-/- human gastric organoids induced morphologic dysplasia, tumorigenicity, and mucinous differentiation. Genetic WNT/ß-catenin activation rescued mucinous differentiation, but not hyperproliferation, suggesting alternative pathways of ARID1A KO-mediated transformation. ARID1A mutation induced transcriptional regulatory modules characteristic of microsatellite instability and Epstein-Barr virus-associated subtype human gastric cancer, including FOXM1-associated mitotic genes and BIRC5/survivin. Convergently, high-throughput compound screening indicated selective vulnerability of ARID1A-deficient organoids to inhibition of BIRC5/survivin, functionally implicating this pathway as an essential mediator of ARID1A KO-dependent early-stage gastric tumorigenesis. Overall, we define distinct pathways downstream of oncogenic ARID1A mutation, with nonessential WNT-inhibited mucinous differentiation in parallel with essential transcriptional FOXM1/BIRC5-stimulated proliferation, illustrating the general utility of organoid-based forward genetic cancer analysis in human cells.

SIGNIFICANCE:

We establish the first human forward genetic modeling of a commonly mutated tumor suppressor gene, ARID1A. Our study integrates diverse modalities including CRISPR/Cas9 genome editing, organoid culture, systems biology, and small-molecule screening to derive novel insights into early transformation mechanisms of ARID1A-deficient gastric cancers.See related commentary by Zafra and Dow, p. 1327.This article is highlighted in the In This Issue feature, p. 1307.
Assuntos

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Assunto principal: Neoplasias Gástricas / Fatores de Transcrição / Transformação Celular Neoplásica / Proteínas de Ligação a DNA / Sistemas CRISPR-Cas Tipo de estudo: Prognostic_studies Limite: Humans Idioma: En Revista: Cancer Discov Ano de publicação: 2021 Tipo de documento: Article

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Assunto principal: Neoplasias Gástricas / Fatores de Transcrição / Transformação Celular Neoplásica / Proteínas de Ligação a DNA / Sistemas CRISPR-Cas Tipo de estudo: Prognostic_studies Limite: Humans Idioma: En Revista: Cancer Discov Ano de publicação: 2021 Tipo de documento: Article