Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 3 de 3
Filter
Add more filters










Database
Language
Publication year range
1.
Clin Cancer Res ; 30(6): 1079-1092, 2024 Mar 15.
Article in English | MEDLINE | ID: mdl-37916971

ABSTRACT

Epithelioid sarcoma (EpS) is an ultra-rare malignant soft-tissue cancer mostly affecting adolescents and young adults. EpS often exhibits an unfavorable clinical course with fatal outcome in ∼50% of cases despite aggressive multimodal therapies combining surgery, chemotherapy, and irradiation. EpS is traditionally classified in a more common, less aggressive distal (classic) type and a rarer aggressive proximal type. Both subtypes are characterized by a loss of nuclear INI1 expression, most often following homozygous deletion of its encoding gene, SMARCB1-a core subunit of the SWI/SNF chromatin remodeling complex. In 2020, the EZH2 inhibitor tazemetostat was the first targeted therapy approved for EpS, raising new hopes. Still, the vast majority of patients did not benefit from this drug or relapsed rapidly. Further, other recent therapeutic modalities, including immunotherapy, are only effective in a fraction of patients. Thus, novel strategies, specifically targeted to EpS, are urgently needed. To accelerate translational research on EpS and eventually boost the discovery and development of new diagnostic tools and therapeutic options, a vibrant translational research community has formed in past years and held two international EpS digital expert meetings in 2021 and 2023. This review summarizes our current understanding of EpS from the translational research perspective and points to innovative research directions to address the most pressing questions in the field, as defined by expert consensus and patient advocacy groups.


Subject(s)
Sarcoma , Transcription Factors , Adolescent , Young Adult , Humans , Transcription Factors/genetics , DNA-Binding Proteins/genetics , Chromosomal Proteins, Non-Histone/genetics , Homozygote , Consensus , Sequence Deletion , SMARCB1 Protein/genetics , SMARCB1 Protein/metabolism , Sarcoma/diagnosis , Sarcoma/genetics , Sarcoma/therapy
2.
Cancer Cell ; 33(6): 1128-1141.e7, 2018 06 11.
Article in English | MEDLINE | ID: mdl-29861296

ABSTRACT

Synovial sarcoma (SS) is defined by the hallmark SS18-SSX fusion oncoprotein, which renders BAF complexes aberrant in two manners: gain of SSX to the SS18 subunit and concomitant loss of BAF47 subunit assembly. Here we demonstrate that SS18-SSX globally hijacks BAF complexes on chromatin to activate an SS transcriptional signature that we define using primary tumors and cell lines. Specifically, SS18-SSX retargets BAF complexes from enhancers to broad polycomb domains to oppose PRC2-mediated repression and activate bivalent genes. Upon suppression of SS18-SSX, reassembly of BAF47 restores enhancer activation, but is not required for proliferative arrest. These results establish a global hijacking mechanism for SS18-SSX on chromatin, and define the distinct contributions of two concurrent BAF complex perturbations.


Subject(s)
Chromatin/genetics , Oncogene Proteins, Fusion/genetics , SMARCB1 Protein/genetics , Sarcoma, Synovial/genetics , Cell Line, Tumor , Chromatin/metabolism , Enhancer Elements, Genetic/genetics , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques/methods , HEK293 Cells , Humans , Oncogene Proteins, Fusion/metabolism , SMARCB1 Protein/metabolism , Sarcoma, Synovial/metabolism , Sarcoma, Synovial/pathology , Exome Sequencing/methods
3.
Nat Genet ; 49(11): 1613-1623, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28945250

ABSTRACT

Perturbations to mammalian SWI/SNF (mSWI/SNF or BAF) complexes contribute to more than 20% of human cancers, with driving roles first identified in malignant rhabdoid tumor, an aggressive pediatric cancer characterized by biallelic inactivation of the core BAF complex subunit SMARCB1 (BAF47). However, the mechanism by which this alteration contributes to tumorigenesis remains poorly understood. We find that BAF47 loss destabilizes BAF complexes on chromatin, absent significant changes in complex assembly or integrity. Rescue of BAF47 in BAF47-deficient sarcoma cell lines results in increased genome-wide BAF complex occupancy, facilitating widespread enhancer activation and opposition of Polycomb-mediated repression at bivalent promoters. We demonstrate differential regulation by two distinct mSWI/SNF assemblies, BAF and PBAF complexes, enhancers and promoters, respectively, suggesting that each complex has distinct functions that are perturbed upon BAF47 loss. Our results demonstrate collaborative mechanisms of mSWI/SNF-mediated gene activation, identifying functions that are co-opted or abated to drive human cancers and developmental disorders.


Subject(s)
Carcinogenesis/genetics , Chromosomal Proteins, Non-Histone/genetics , Gene Expression Regulation, Neoplastic , Rhabdoid Tumor/genetics , SMARCB1 Protein/genetics , Sarcoma/genetics , Transcription Factors/genetics , Carcinogenesis/metabolism , Carcinogenesis/pathology , Cell Line, Tumor , Chromatin/chemistry , Chromatin/metabolism , Chromatin Assembly and Disassembly , Chromosomal Proteins, Non-Histone/metabolism , Enhancer Elements, Genetic , Genetic Complementation Test , Humans , Polycomb-Group Proteins/genetics , Polycomb-Group Proteins/metabolism , Promoter Regions, Genetic , Rhabdoid Tumor/metabolism , Rhabdoid Tumor/pathology , SMARCB1 Protein/deficiency , Sarcoma/metabolism , Sarcoma/pathology , Transcription Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...