Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
2.
Curr Opin Struct Biol ; 71: 65-70, 2021 12.
Article in English | MEDLINE | ID: mdl-34225008

ABSTRACT

Cellular identity and physiologic function in mammary epithelial cells and in many breast cancers flow from the action of a network of master transcriptional regulators including estrogen receptor alpha, GATA3, and FOXA1. The last decade has seen the completion of multiple large sequencing projects focusing on breast cancer. These massive compendia of sequence data have provided a wealth of new information linking mutation in these transcription factors to alterations in tumor biology and transcriptional program. The emerging details on mutation in cancer, and direct experimental exploration of hypotheses based on it, are now providing a wealth of new information on the roles played by estrogen receptor alpha, GATA3, and FOXA1 in regulating gene transcription and how their combined action contributes to a network shaping cell function in both physiologic and disease states.


Subject(s)
Breast Neoplasms , Breast Neoplasms/genetics , Estrogen Receptor alpha/genetics , Estrogen Receptor alpha/metabolism , Female , GATA3 Transcription Factor/genetics , Gene Expression Regulation, Neoplastic , Gene Regulatory Networks , Hepatocyte Nuclear Factor 3-alpha/genetics , Humans , Receptors, Estrogen
3.
Clin Cancer Res ; 26(15): 3908-3917, 2020 08 01.
Article in English | MEDLINE | ID: mdl-32156746

ABSTRACT

Small-cell carcinoma of the ovary, hypercalcemic type (SCCOHT) is a rare and highly aggressive ovarian malignancy. In almost all cases, it is associated with somatic and often germline pathogenic variants in SMARCA4, which encodes for the SMARCA4 protein (BRG1), a subunit of the SWI/SNF chromatin remodeling complex. Approximately 20% of human cancers possess pathogenic variants in at least one SWI/SNF subunit. Because of their role in regulating many important cellular processes including transcriptional control, DNA repair, differentiation, cell division, and DNA replication, SWI/SNF complexes with mutant subunits are thought to contribute to cancer initiation and progression. Fewer than 500 cases of SCCOHT have been reported in the literature and approximately 60% are associated with hypercalcemia. SCCOHT primarily affects females under 40 years of age who usually present with symptoms related to a pelvic mass. SCCOHT is an aggressive cancer, with long-term survival rates of 30% in early-stage cases. Although various treatment approaches have been proposed, there is no consensus on surveillance and therapeutic strategy. An international group of multidisciplinary clinicians and researchers recently formed the International SCCOHT Consortium to evaluate current knowledge and propose consensus surveillance and therapeutic recommendations, with the aim of improving outcomes. Here, we present an overview of the genetics of this cancer, provide updates on new treatment targets, and propose management guidelines for this challenging cancer.


Subject(s)
Carcinoma, Small Cell/genetics , DNA Helicases/genetics , Hypercalcemia/genetics , Nuclear Proteins/genetics , Ovarian Neoplasms/genetics , Transcription Factors/genetics , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Small Cell/blood , Carcinoma, Small Cell/mortality , Carcinoma, Small Cell/therapy , Chemotherapy, Adjuvant/methods , Chemotherapy, Adjuvant/standards , Chromatin Assembly and Disassembly/genetics , Female , Gynecology/standards , Humans , Hypercalcemia/blood , Hypercalcemia/pathology , Hypercalcemia/therapy , Medical Oncology/standards , Mutation , Ovarian Neoplasms/blood , Ovarian Neoplasms/mortality , Ovarian Neoplasms/therapy , Ovariectomy/standards , Ovary/pathology , Ovary/surgery , Practice Guidelines as Topic , Radiotherapy, Adjuvant/standards , Stem Cell Transplantation/standards , Survival Rate , Treatment Outcome
4.
Neurosci J ; 2019: 5950818, 2019.
Article in English | MEDLINE | ID: mdl-31275953

ABSTRACT

There is an excellent correlation between the age when alcohol consumption begins and the likelihood of lifelong problems with alcohol abuse. Alcohol use often begins in adolescence, a time marked by brain development and maturation of numerous brain systems. Rats are an important model, wherein the emergence of alcohol withdrawal symptoms serves as a gauge of dependency following chronic alcohol consumption. Previous work has shown that adolescent Long-Evans rats consume high levels of alcohol and develop a severe alcohol withdrawal syndrome when fed alcohol as part of a liquid diet. Acutely, alcohol inhibits two important excitatory receptors for glutamate (NMDA and AMPA) and may further decrease glutamate activity through modulatory adenosine receptors. The present study focuses on potential adaptive changes in expression of these receptors that may create a receptor imbalance during chronic alcohol consumption and lead to severe overexcitation of the adolescent brain during alcohol withdrawal. Levels of brain expression of NMDA, AMPA, and adenosine A1 and A2a receptors were determined by Western blotting after adolescent rats consumed an alcohol-containing liquid diet for 4, 11, or 18 days. Severity of alcohol withdrawal was also assessed at these time points. Levels increased for both AMPA and NMDA receptors, significant and approaching maximal by day 11. In contrast, A1 receptor density showed a slow decline reaching significance at 18 days. There were no changes in expression of adenosine A2a receptor. The most severe withdrawal symptoms appear to coincide with the later downregulation of adenosine A1 receptors coming on top of maximal upregulation of excitatory AMPA and NMDA glutamate receptors. Thus, loss of adenosine "brakes" on glutamate excitation may punctuate receptor imbalance in alcohol-consuming adolescents by allowing the upregulation of the excitatory receptors to have full impact during early alcohol withdrawal.

5.
Expert Rev Anticancer Ther ; 19(5): 375-391, 2019 05.
Article in English | MEDLINE | ID: mdl-30986130

ABSTRACT

INTRODUCTION: Cancer genome sequencing studies have discovered mutations in members of the SWItch/Sucrose Non-Fermentable (SWI/SNF) chromatin-remodeling complex in nearly 25% of human cancers. The SWI/SNF complex, first discovered in S. cerevisiae, shows strong conservation from yeast to Drosophila to mammals, contains approximately 10-12 subunits and regulates nucleosome positioning through the energy generated by its ATPase subunits. The unexpected finding of frequent mutations in the complex has fueled studies to identify the mechanisms that drive tumor development and the accompanying therapeutic vulnerabilities. Areas covered: In the review, we focus upon the potential roles different SWI/SNF subunit mutations play in human oncogenesis, their common and unique mechanisms of transformation and the potential for translating these mechanisms into targeted therapies for SWI/SNF-mutant tumors. Expert opinion: We currently have limited insights into how mutations in different SWI/SNF subunits drive the development of human tumors. Because the SWI/SNF complex participates in a broad range of normal cellular functions, defining specific oncogenic pathways has proved difficult. In addition, therapeutic options for SWI/SNF-mutant cancers have mainly evolved from high-throughput screens of cell lines with mutations in different subunits. Future studies should follow a more coherent plan to pinpoint common vulnerabilities among these tumors.


Subject(s)
Chromosomal Proteins, Non-Histone/genetics , Molecular Targeted Therapy , Neoplasms/therapy , Transcription Factors/genetics , Animals , Carcinogenesis/genetics , Epigenome , High-Throughput Screening Assays , Humans , Mutation , Neoplasms/genetics , Neoplasms/pathology
6.
Mol Cancer Ther ; 17(12): 2767-2779, 2018 12.
Article in English | MEDLINE | ID: mdl-30232145

ABSTRACT

Small cell carcinoma of the ovary, hypercalcemic type (SCCOHT) is a rare but extremely lethal malignancy that mainly impacts young women. SCCOHT is characterized by a diploid genome with loss of SMARCA4 and lack of SMARCA2 expression, two mutually exclusive ATPases of the SWI/SNF chromatin-remodeling complex. We and others have identified the histone methyltransferase EZH2 as a promising therapeutic target for SCCOHT, suggesting that SCCOHT cells depend on the alternation of epigenetic pathways for survival. In this study, we found that SCCOHT cells were more sensitive to pan-HDAC inhibitors compared with other ovarian cancer lines or immortalized cell lines tested. Pan-HDAC inhibitors, such as quisinostat, reversed the expression of a group of proteins that were deregulated in SCCOHT cells due to SMARCA4 loss, leading to growth arrest, apoptosis, and differentiation in vitro and suppressed tumor growth of xenografted tumors of SCCOHT cells. Moreover, combined treatment of HDAC inhibitors and EZH2 inhibitors at sublethal doses synergistically induced histone H3K27 acetylation and target gene expression, leading to rapid induction of apoptosis and growth suppression of SCCOHT cells and xenografted tumors. Therefore, our preclinical study highlighted the therapeutic potential of combined treatment of HDAC inhibitors with EZH2 catalytic inhibitors to treat SCCOHT.


Subject(s)
Antineoplastic Agents/therapeutic use , Carcinoma, Small Cell/drug therapy , Enhancer of Zeste Homolog 2 Protein/antagonists & inhibitors , Histone Deacetylase Inhibitors/therapeutic use , Hypercalcemia/drug therapy , Ovarian Neoplasms/drug therapy , Animals , Apoptosis/drug effects , Biocatalysis/drug effects , Carcinoma, Small Cell/complications , Carcinoma, Small Cell/genetics , Carcinoma, Small Cell/pathology , Cell Cycle Checkpoints/drug effects , Cell Differentiation/drug effects , Cell Line, Tumor , Cell Lineage/drug effects , DNA Helicases/metabolism , Drug Synergism , Enhancer of Zeste Homolog 2 Protein/metabolism , Epigenesis, Genetic/drug effects , Female , Gene Expression Regulation, Neoplastic/drug effects , Histone Deacetylase Inhibitors/pharmacology , Humans , Hydroxamic Acids/pharmacology , Hydroxamic Acids/therapeutic use , Hypercalcemia/complications , Mice , Nuclear Proteins/metabolism , Ovarian Neoplasms/complications , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Proteome/metabolism , Transcription Factors/metabolism , Xenograft Model Antitumor Assays
7.
Clin Cancer Res ; 24(8): 1932-1943, 2018 04 15.
Article in English | MEDLINE | ID: mdl-29440177

ABSTRACT

Purpose: Small cell carcinoma of the ovary, hypercalcemic type (SCCOHT) is a rare, aggressive ovarian cancer in young women that is universally driven by loss of the SWI/SNF ATPase subunits SMARCA4 and SMARCA2. A great need exists for effective targeted therapies for SCCOHT.Experimental Design: To identify underlying therapeutic vulnerabilities in SCCOHT, we conducted high-throughput siRNA and drug screens. Complementary proteomics approaches profiled kinases inhibited by ponatinib. Ponatinib was tested for efficacy in two patient-derived xenograft (PDX) models and one cell-line xenograft model of SCCOHT.Results: The receptor tyrosine kinase (RTK) family was enriched in siRNA screen hits, with FGFRs and PDGFRs being overlapping hits between drug and siRNA screens. Of multiple potent drug classes in SCCOHT cell lines, RTK inhibitors were only one of two classes with selectivity in SCCOHT relative to three SWI/SNF wild-type ovarian cancer cell lines. We further identified ponatinib as the most effective clinically approved RTK inhibitor. Reexpression of SMARCA4 was shown to confer a 1.7-fold increase in resistance to ponatinib. Subsequent proteomic assessment of ponatinib target modulation in SCCOHT cell models confirmed inhibition of nine known ponatinib target kinases alongside 77 noncanonical ponatinib targets in SCCOHT. Finally, ponatinib delayed tumor doubling time 4-fold in SCCOHT-1 xenografts while reducing final tumor volumes in SCCOHT PDX models by 58.6% and 42.5%.Conclusions: Ponatinib is an effective agent for SMARCA4-mutant SCCOHT in both in vitro and in vivo preclinical models through its inhibition of multiple kinases. Clinical investigation of this FDA-approved oncology drug in SCCOHT is warranted. Clin Cancer Res; 24(8); 1932-43. ©2018 AACR.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Small Cell/metabolism , Carcinoma, Small Cell/pathology , Imidazoles/pharmacology , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Protein Kinase Inhibitors/pharmacology , Pyridazines/pharmacology , Animals , Carcinoma, Small Cell/drug therapy , Cell Line, Tumor , Computational Biology/methods , Disease Models, Animal , Female , Humans , Mice , Ovarian Neoplasms/drug therapy , Protein Interaction Mapping , Protein Interaction Maps , RNA, Small Interfering/genetics , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Xenograft Model Antitumor Assays
8.
J Pathol ; 242(3): 371-383, 2017 07.
Article in English | MEDLINE | ID: mdl-28444909

ABSTRACT

Small cell carcinoma of the ovary, hypercalcaemic type (SCCOHT) is a rare but aggressive and untreatable malignancy affecting young women. We and others recently discovered that SMARCA4, a gene encoding the ATPase of the SWI/SNF chromatin-remodelling complex, is the only gene recurrently mutated in the majority of SCCOHT. The low somatic complexity of SCCOHT genomes and the prominent role of the SWI/SNF chromatin-remodelling complex in transcriptional control of genes suggest that SCCOHT cells may rely on epigenetic rewiring for oncogenic transformation. Herein, we report that approximately 80% (19/24) of SCCOHT tumour samples have strong expression of the histone methyltransferase EZH2 by immunohistochemistry, with the rest expressing variable amounts of EZH2. Re-expression of SMARCA4 suppressed the expression of EZH2 in SCCOHT cells. In comparison to other ovarian cell lines, SCCOHT cells displayed hypersensitivity to EZH2 shRNAs and two selective EZH2 inhibitors, GSK126 and EPZ-6438. EZH2 inhibitors induced cell cycle arrest, apoptosis, and cell differentiation in SCCOHT cells, along with the induction of genes involved in cell cycle regulation, apoptosis, and neuron-like differentiation. EZH2 inhibitors suppressed tumour growth and improved the survival of mice bearing SCCOHT xenografts. Therefore, our data suggest that loss of SMARCA4 creates a dependency on the catalytic activity of EZH2 in SCCOHT cells and that pharmacological inhibition of EZH2 is a promising therapeutic strategy for treating this disease. Copyright © 2017 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Subject(s)
Carcinoma, Small Cell/enzymology , Enhancer of Zeste Homolog 2 Protein/antagonists & inhibitors , Histone-Lysine N-Methyltransferase/metabolism , Hypercalcemia/enzymology , Ovarian Neoplasms/enzymology , Animals , Apoptosis/physiology , Carcinoma, Ovarian Epithelial , Cell Cycle Checkpoints/physiology , Cell Line, Tumor , Cell Transformation, Neoplastic , DNA Helicases/deficiency , Down-Regulation , Enhancer of Zeste Homolog 2 Protein/metabolism , Female , Histone Methyltransferases , Humans , Neoplasm Transplantation , Neoplasms, Glandular and Epithelial/enzymology , Nuclear Proteins/deficiency , Transcription Factors/deficiency , Transplantation, Heterologous , Tumor Cells, Cultured , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL
...