Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Mol Cell ; 83(8): 1350-1367.e7, 2023 04 20.
Article in English | MEDLINE | ID: mdl-37028419

ABSTRACT

The mammalian SWI/SNF (mSWI/SNF or BAF) family of chromatin remodeling complexes play critical roles in regulating DNA accessibility and gene expression. The three final-form subcomplexes-cBAF, PBAF, and ncBAF-are distinct in biochemical componentry, chromatin targeting, and roles in disease; however, the contributions of their constituent subunits to gene expression remain incompletely defined. Here, we performed Perturb-seq-based CRISPR-Cas9 knockout screens targeting mSWI/SNF subunits individually and in select combinations, followed by single-cell RNA-seq and SHARE-seq. We uncovered complex-, module-, and subunit-specific contributions to distinct regulatory networks and defined paralog subunit relationships and shifted subcomplex functions upon perturbations. Synergistic, intra-complex genetic interactions between subunits reveal functional redundancy and modularity. Importantly, single-cell subunit perturbation signatures mapped across bulk primary human tumor expression profiles both mirror and predict cBAF loss-of-function status in cancer. Our findings highlight the utility of Perturb-seq to dissect disease-relevant gene regulatory impacts of heterogeneous, multi-component master regulatory complexes.


Subject(s)
Chromatin Assembly and Disassembly , Neoplasms , Animals , Humans , Chromosomal Proteins, Non-Histone/genetics , Chromosomal Proteins, Non-Histone/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Chromatin/genetics , Mammals/metabolism
2.
Cell Rep ; 41(5): 111581, 2022 11 01.
Article in English | MEDLINE | ID: mdl-36323264

ABSTRACT

The dynamic regulation of ß-cell abundance is poorly understood. Since chromatin remodeling plays critical roles in liver regeneration, these mechanisms could be generally important for regeneration in other tissues. Here, we show that the ARID1A mammalian SWI/SNF complex subunit is a critical regulator of ß-cell regeneration. Arid1a is highly expressed in quiescent ß-cells but is physiologically suppressed when ß-cells proliferate during pregnancy or after pancreas resection. Whole-body Arid1a knockout mice are protected against streptozotocin-induced diabetes. Cell-type and temporally specific genetic dissection show that ß-cell-specific Arid1a deletion can potentiate ß-cell regeneration in multiple contexts. Transcriptomic and epigenomic profiling of mutant islets reveal increased neuregulin-ERBB-NR4A signaling. Chemical inhibition of ERBB or NR4A1 blocks increased regeneration associated with Arid1a loss. Mammalian SWI/SNF (mSWI/SNF) complex activity is a barrier to ß-cell regeneration in physiologic and disease states.


Subject(s)
Epidermal Growth Factor , Nuclear Proteins , Mice , Animals , Pregnancy , Female , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Chromatin Assembly and Disassembly , Signal Transduction , Liver Regeneration , Mammals/metabolism , DNA-Binding Proteins/genetics , Transcription Factors/genetics
3.
Nat Genet ; 52(2): 198-207, 2020 02.
Article in English | MEDLINE | ID: mdl-31932695

ABSTRACT

Mutations in ARID1A, a subunit of the SWI/SNF chromatin remodeling complex, are the most common alterations of the SWI/SNF complex in estrogen-receptor-positive (ER+) breast cancer. We identify that ARID1A inactivating mutations are present at a high frequency in advanced endocrine-resistant ER+ breast cancer. An epigenome CRISPR-CAS9 knockout (KO) screen identifies ARID1A as the top candidate whose loss determines resistance to the ER degrader fulvestrant. ARID1A inactivation in cells and in patients leads to resistance to ER degraders by facilitating a switch from ER-dependent luminal cells to ER-independent basal-like cells. Cellular plasticity is mediated by loss of ARID1A-dependent SWI/SNF complex targeting to genomic sites of the luminal lineage-determining transcription factors including ER, forkhead box protein A1 (FOXA1) and GATA-binding factor 3 (GATA3). ARID1A also regulates genome-wide ER-FOXA1 chromatin interactions and ER-dependent transcription. Altogether, we uncover a critical role for ARID1A in maintaining luminal cell identity and endocrine therapeutic response in ER+ breast cancer.


Subject(s)
Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , DNA-Binding Proteins/genetics , Drug Resistance, Neoplasm/genetics , Receptors, Estrogen/metabolism , Transcription Factors/genetics , Animals , Breast Neoplasms/mortality , Breast Neoplasms/pathology , Case-Control Studies , Cell Line, Tumor , Chromatin/genetics , Chromatin/metabolism , DNA-Binding Proteins/metabolism , Female , GATA3 Transcription Factor/genetics , GATA3 Transcription Factor/metabolism , Hepatocyte Nuclear Factor 3-alpha/genetics , Hepatocyte Nuclear Factor 3-alpha/metabolism , Humans , Mice , Mutation , Receptors, Estrogen/genetics , Transcription Factors/metabolism , Xenograft Model Antitumor Assays
4.
Cell ; 175(5): 1272-1288.e20, 2018 11 15.
Article in English | MEDLINE | ID: mdl-30343899

ABSTRACT

Mammalian SWI/SNF (mSWI/SNF) ATP-dependent chromatin remodeling complexes are multi-subunit molecular machines that play vital roles in regulating genomic architecture and are frequently disrupted in human cancer and developmental disorders. To date, the modular organization and pathways of assembly of these chromatin regulators remain unknown, presenting a major barrier to structural and functional determination. Here, we elucidate the architecture and assembly pathway across three classes of mSWI/SNF complexes-canonical BRG1/BRM-associated factor (BAF), polybromo-associated BAF (PBAF), and newly defined ncBAF complexes-and define the requirement of each subunit for complex formation and stability. Using affinity purification of endogenous complexes from mammalian and Drosophila cells coupled with cross-linking mass spectrometry (CX-MS) and mutagenesis, we uncover three distinct and evolutionarily conserved modules, their organization, and the temporal incorporation of these modules into each complete mSWI/SNF complex class. Finally, we map human disease-associated mutations within subunits and modules, defining specific topological regions that are affected upon subunit perturbation.


Subject(s)
Chromatin Assembly and Disassembly , Chromatin/metabolism , Chromosomal Proteins, Non-Histone/metabolism , Transcription Factors/metabolism , Animals , Chromatin/chemistry , Chromosomal Proteins, Non-Histone/analysis , Chromosomal Proteins, Non-Histone/genetics , Drosophila/metabolism , Gene Knockout Techniques , HEK293 Cells , Humans , Mass Spectrometry , Mutagenesis , Protein Subunits/analysis , Protein Subunits/genetics , Protein Subunits/metabolism , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification , Transcription Factors/analysis , Transcription Factors/genetics
5.
Cancer Cell ; 32(5): 542-543, 2017 11 13.
Article in English | MEDLINE | ID: mdl-29136498

ABSTRACT

In this issue of Cancer Cell, Sun et al. describe context-dependent oncogenic and tumor-suppressive functions for the mammalian SWI/SNF (BAF) complex subunit ARID1A in the development and progression of hepatocellular carcinoma (HCC).


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Adult , Animals , Genes, Tumor Suppressor , Oncogenes
SELECTION OF CITATIONS
SEARCH DETAIL
...