Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
1.
Genes Dev ; 27(13): 1435-40, 2013 Jul 01.
Article in English | MEDLINE | ID: mdl-23796898

ABSTRACT

Androgen-deprived prostate cancer (PCa) is infiltrated by B lymphocytes that produce cytokines that activate IκB kinase α (IKKα) to accelerate the emergence of castration-resistant tumors. We now demonstrate that infiltrating B lymphocytes and IKKα are also required for androgen-dependent expansion of epithelial progenitors responsible for prostate regeneration. In these cells and in PCa cells, IKKα phosphorylates transcription factor E2F1 on a site that promotes its nuclear translocation, association with the coactivator CBP, and recruitment to critical genomic targets that include Bmi1, a key regulator of normal and cancerous prostate stem cell renewal. The IKKα-BMI1 pathway is also activated in human PCa.


Subject(s)
B-Lymphocytes/physiology , E2F1 Transcription Factor/metabolism , I-kappa B Kinase/metabolism , Polycomb Repressive Complex 1/metabolism , Prostate/physiopathology , Proto-Oncogene Proteins/metabolism , Regeneration , Androgens/pharmacology , Animals , Cells, Cultured , E2F1 Transcription Factor/genetics , Gene Expression Regulation, Developmental/drug effects , Humans , I-kappa B Kinase/genetics , Male , Mice , Neoplasm Recurrence, Local/physiopathology , Orchiectomy , Polycomb Repressive Complex 1/genetics , Prostate/drug effects , Prostatic Neoplasms/pathology , Proto-Oncogene Proteins/genetics
2.
Mol Cell ; 39(6): 873-85, 2010 Sep 24.
Article in English | MEDLINE | ID: mdl-20864035

ABSTRACT

During an immune response, B cells undergo rapid proliferation and activation-induced cytidine deaminase (AID)-dependent remodeling of immunoglobulin (IG) genes within germinal centers (GCs) to generate memory B and plasma cells. Unfortunately, the genotoxic stress associated with the GC reaction also promotes most B cell malignancies. Here, we report that exogenous and intrinsic AID-induced DNA strand breaks activate ATM, which signals through an LKB1 intermediate to inactivate CRTC2, a transcriptional coactivator of CREB. Using genome-wide location analysis, we determined that CRTC2 inactivation unexpectedly represses a genetic program that controls GC B cell proliferation, self-renewal, and differentiation while opposing lymphomagenesis. Inhibition of this pathway results in increased GC B cell proliferation, reduced antibody secretion, and impaired terminal differentiation. Multiple distinct pathway disruptions were also identified in human GC B cell lymphoma patient samples. Combined, our data show that CRTC2 inactivation, via physiologic DNA damage response signaling, promotes B cell differentiation in response to genotoxic stress.


Subject(s)
B-Lymphocytes/cytology , Cell Cycle Proteins/metabolism , Cell Differentiation/immunology , Cytidine Deaminase/genetics , DNA Damage/immunology , DNA-Binding Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Tumor Suppressor Proteins/metabolism , AMP-Activated Protein Kinase Kinases , Active Transport, Cell Nucleus/drug effects , Active Transport, Cell Nucleus/radiation effects , Animals , Ataxia Telangiectasia Mutated Proteins , B-Lymphocytes/drug effects , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , B-Lymphocytes/radiation effects , Cell Cycle Proteins/antagonists & inhibitors , Cell Cycle Proteins/genetics , Cell Differentiation/drug effects , Cell Differentiation/radiation effects , Cell Line, Tumor , DNA Breaks, Double-Stranded/drug effects , DNA Breaks, Double-Stranded/radiation effects , DNA Damage/drug effects , DNA Damage/radiation effects , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/genetics , Gene Expression/drug effects , Gene Expression/immunology , Gene Expression/radiation effects , Gene Expression Regulation/immunology , Germinal Center/cytology , Humans , Immunoglobulin Class Switching/physiology , Lymphoma, B-Cell/genetics , Lymphoma, B-Cell/metabolism , Metformin/pharmacology , Mice , Mice, Knockout , Phosphorylation/drug effects , Phosphorylation/radiation effects , Plasma Cells/cytology , Plasma Cells/immunology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins/genetics , Signal Transduction/drug effects , Signal Transduction/genetics , Signal Transduction/immunology , Signal Transduction/radiation effects , Transcription Factors/genetics , Transcription Factors/metabolism , Tumor Suppressor Proteins/antagonists & inhibitors , Tumor Suppressor Proteins/genetics
3.
Stem Cells ; 26(6): 1484-9, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18369100

ABSTRACT

Differences between individual DNA sequences provide the basis for human genetic variability. Forms of genetic variation include single-nucleotide polymorphisms, insertions/duplications, deletions, and inversions/translocations. The genome of human embryonic stem cells (hESCs) has been characterized mainly by karyotyping and comparative genomic hybridization (CGH), techniques whose relatively low resolution at 2-10 megabases (Mb) cannot accurately determine most copy number variability, which is estimated to involve 10%-20% of the genome. In this brief technical study, we examined HSF1 and HSF6 hESCs using array-comparative genomic hybridization (aCGH) to determine copy number variants (CNVs) as a higher-resolution method for characterizing hESCs. Our approach used five samples for each hESC line and showed four consistent CNVs for HSF1 and five consistent CNVs for HSF6. These consistent CNVs included amplifications and deletions that ranged in size from 20 kilobases to 1.48 megabases, involved seven different chromosomes, were both shared and unique between hESCs, and were maintained during neuronal stem/progenitor cell differentiation or drug selection. Thirty HSF1 and 40 HSF6 less consistently scored but still highly significant candidate CNVs were also identified. Overall, aCGH provides a promising approach for uniquely identifying hESCs and their derivatives and highlights a potential genomic source for distinct differentiation and functional potentials that lower-resolution karyotype and CGH techniques could miss. Disclosure of potential conflicts of interest is found at the end of this article.


Subject(s)
Embryonic Stem Cells/cytology , Embryonic Stem Cells/physiology , Genetic Variation , Genome, Human , Cell Culture Techniques , Cell Division/genetics , DNA/genetics , DNA-Binding Proteins/genetics , Heat Shock Transcription Factors , Heat-Shock Proteins/genetics , Humans , Neurons/cytology , Neurons/physiology , Nucleic Acid Hybridization/methods , Polymerase Chain Reaction/methods , Transcription Factors/genetics
4.
J Mol Biol ; 362(2): 173-83, 2006 Sep 15.
Article in English | MEDLINE | ID: mdl-16920149

ABSTRACT

B cell-specific B29 (Igbeta, CD79b) genes in rat, mouse, and human are situated between the 5' growth hormone (GH) locus control region and the 3' GH gene cluster. The entire GH genomic region is DNase 1 hypersensitive in GH-expressing pituitary cells, which predicts an "open" chromatin configuration, and yet B29 is not expressed. The B29 promoter and enhancers exhibit histone deacetylation in pituitary cells, but histone deacetylase inhibition failed to activate B29 expression. The B29 promoter and a 3' enhancer showed local dense DNA methylation in both pituitary and non-lymphoid cells consistent with gene silencing. However, DNA methyltransferase inhibition did not activate B29 expression either. B29 promoter constructs were minimally activated in transfected pituitary cells. Co-transfection of the B cell-specific octamer transcriptional co-activator Bob1 with the B29 promoter construct resulted in high level promoter activity in pituitary cells comparable to B29 promoter activity in transfected B cells. Unexpectedly, inclusion of the B29 3' enhancer in B29 promoter constructs strongly inhibited B29 transcriptional activity even when pituitary cells were co-transfected with Bob1. Both Oct-1 and Pit-1 bind the B29 3' enhancer in in vitro electrophoretic mobility shift assay and in in vivo chromatin immunoprecipitation analyses. These data indicate that the GH locus-embedded, tissue-specific B29 gene is silenced in GH-expressing pituitary cells by epigenetic mechanisms, the lack of a B cell-specific transcription factor, and likely by the B29 3' enhancer acting as a powerful silencer in a context and tissue-specific manner.


Subject(s)
CD79 Antigens/genetics , Enhancer Elements, Genetic , Gene Expression Regulation , Gene Silencing , Immunologic Factors/genetics , Pituitary Gland/cytology , Animals , Azacitidine/metabolism , CD79 Antigens/metabolism , Cell Line , Enzyme Inhibitors/metabolism , Histone Deacetylase Inhibitors , Humans , Hydroxamic Acids/metabolism , Immunologic Factors/metabolism , Mice , Octamer Transcription Factor-1/metabolism , Pituitary Gland/immunology , Promoter Regions, Genetic , Rats , Trans-Activators/genetics , Trans-Activators/metabolism , Transcription Factor Pit-1/metabolism
5.
Proc Natl Acad Sci U S A ; 104(24): 10175-80, 2007 Jun 12.
Article in English | MEDLINE | ID: mdl-17548807

ABSTRACT

Aberrant expression of the TCL1 oncoprotein promotes malignant transformation of germinal center (GC) B cells. Repression of TCL1 in GC B cells facilitates FAS-mediated apoptosis and prevents lymphoma formation. However, the mechanism for this repression is unknown. Here we show that the CREB coactivator TORC2 directly regulates TCL1 expression independent of CREB Ser-133 phosphorylation and CBP/p300 recruitment. GC signaling through CD40 or the BCR, which activates pCREB-dependent genes, caused TORC2 phosphorylation, cytosolic emigration, and TCL1 repression. Signaling via cAMP-inducible pathways inhibited TCL1 repression and reduced apoptosis, consistent with a prosurvival role for TCL1 before GC selection and supporting an initiating role for aberrant TCL1 expression during GC lymphomagenesis. Our data indicate that a novel CREB/TORC2 regulatory mode controls the normal program of GC gene activation and repression that promotes B cell development and circumvents oncogenic progression. Our results also reconcile a paradox in which signals that activate pCREB/CBP/p300 genes concurrently repress TCL1 to initiate its silencing.


Subject(s)
B-Lymphocytes/metabolism , Cell Transformation, Neoplastic/genetics , Gene Expression Regulation, Neoplastic , Proto-Oncogene Proteins/genetics , Transcription Factors/genetics , Apoptosis , B-Lymphocytes/immunology , B-Lymphocytes/pathology , Cell Line , Cell Line, Tumor , Genetic Vectors , Germinal Center/metabolism , Germinal Center/pathology , Humans , Jurkat Cells , Models, Biological , Plasmids , Proto-Oncogene Proteins/metabolism , Retroviridae/genetics , Signal Transduction , Transcription Factors/metabolism , Transcriptional Activation
SELECTION OF CITATIONS
SEARCH DETAIL