Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Front Oncol ; 10: 448, 2020.
Article in English | MEDLINE | ID: mdl-32309216

ABSTRACT

The interaction of lymphoma cells with their microenvironment has an important role in disease pathogenesis and is being actively pursued therapeutically using immunomodulatory drugs, including immune checkpoint inhibitors. Diffuse large B-cell lymphoma (DLBCL) is an aggressive high-grade disease that remains incurable in ~40% of patients treated with R-CHOP immunochemotherapy. The FOXP1 transcription factor is abundantly expressed in such high-risk DLBCL and we recently identified its regulation of immune response signatures, in particular, its suppression of the cell surface expression of major histocompatibility class II (MHC-II), which has a critical role in antigen presentation to T cells. Using CRISPR/Cas9 genome editing we have depleted Foxp1 expression in the aggressive murine A20 lymphoma cell line. When grown in BALB/c mice, this cell line provides a high-fidelity immunocompetent disseminated lymphoma model that displays many characteristics of human DLBCL. Transient Foxp1-depletion using siRNA, and stable depletion using CRISPR (generated by independently targeting Foxp1 exon six or seven) upregulated cell surface I-Ab (MHC-II) expression without impairing cell viability in vitro. RNA sequencing of Foxp1-depleted A20 clones identified commonly deregulated genes, such as the B-cell marker Cd19, and hallmark DLBCL signatures such as MYC-targets and oxidative phosphorylation. Immunocompetent animals bearing Foxp1-depleted A20 lymphomas showed significantly-improved survival, and 20% did not develop tumors; consistent with modulating immune surveillance, this was not observed in immunodeficient NOD SCIDγ mice. The A20 Foxp1 CRISPR model will help to further characterize the contribution of Foxp1 to lymphoma immune evasion and the potential for Foxp1 targeting to synergize with other immunotherapies.

2.
Endocrinology ; 158(3): 503-515, 2017 03 01.
Article in English | MEDLINE | ID: mdl-28001444

ABSTRACT

Plasmablastic B-cell malignancies include plasmablastic lymphoma and subsets of multiple myeloma and diffuse large B-cell lymphomaDLBCL. These diseases can be difficult to diagnose and treat, and they lack well-characterized cell line models. Here, immunophenotyping and FOXP1 expression profiling identified plasmablastic characteristics in DLBCL cell lines HLY-1 and SU-DHL-9, associated with CTNNAL1, HPGD, RORA, IGF1, and/or vitamin D receptor (VDR) transcription. We demonstrated VDR protein expression in primary plasmablastic tumor cells and confirmed in cell lines expression of both VDR and the metabolic enzyme CYP27B1, which catalyzes active vitamin D3 production. Although Vdr and Cyp27b1 transcription in normal B cells were activated by interleukin 4 (IL-4) and CD40 signaling, respectively, unstimulated malignant plasmablastic cells lacking IL-4 expressed both VDR and CYP27B1. Positive autoregulation evidenced intact VDR function in all plasmablastic lines, and inhibition of growth by active vitamin D3 was both dependent on MYC protein inhibition and could be enhanced by cotreatment with a synthetic ROR ligand SR-1078. Furthermore, a VDR polymorphism, FOK1, was associated with greater vitamin D3-dependent growth inhibition. In summary, HLY-1 provides an important model of strongly plasmablastic lymphoma, and disruption of VDR pathway activity may be of therapeutic benefit in both plasmablastic lymphoma and myeloma.


Subject(s)
Cholecalciferol/therapeutic use , Multiple Myeloma/metabolism , Plasmablastic Lymphoma/metabolism , Receptors, Calcitriol/metabolism , Animals , Benzamides , Cell Cycle/drug effects , Cell Line, Tumor , Cholecalciferol/pharmacology , Female , Forkhead Transcription Factors/metabolism , Humans , Immunophenotyping , Mice, Inbred C57BL , Multiple Myeloma/drug therapy , Multiple Myeloma/genetics , Plasmablastic Lymphoma/drug therapy , Plasmablastic Lymphoma/genetics , Receptors, Calcitriol/genetics , Repressor Proteins/metabolism
3.
Leuk Lymphoma ; 58(5): 1037-1051, 2017 05.
Article in English | MEDLINE | ID: mdl-27678023

ABSTRACT

Diffuse large B-cell lymphoma (DLBCL) is the most common subtype of mature B-cell lymphoma. While the majority of patients are cured with immunochemotherapy incorporating the anti-CD20 monoclonal antibody rituximab (R-CHOP), relapsed and refractory patients still have a dismal prognosis. DLBCL subtypes including an aggressive activated B-cell-like (ABC) and a more favorable prognosis germinal center-like (GCB) DLBCL have been identified by gene expression profiling and are characterized by distinct genetic abnormalities and oncogenic pathways. This identification of novel molecular targets is now enabling clinical trials to evaluate more effective personalized approaches to DLBCL therapy. The forkhead transcription factor FOXP1 is highly expressed in the ABC-DLBCL gene signature and has been extensively studied within the context of DLBCL for more than a decade. Here, we review the significance of FOXP1 in the pathogenesis of DLBCL, summarizing data supporting its utility as a prognostic and subtyping marker, its targeting by genetic aberrations, the importance of specific isoforms, and emerging data demonstrating a functional role in lymphoma biology. FOXP1 is one of the critical transcription factors whose deregulated expression makes important contributions to DLBCL pathogenesis. Thus, FOXP1 warrants further study as a potential theranostic in ABC-DLBCL.


Subject(s)
Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Lymphoma, Large B-Cell, Diffuse/genetics , Lymphoma, Large B-Cell, Diffuse/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Animals , Biomarkers, Tumor , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Gene Expression Regulation, Neoplastic , Genetic Variation , Humans , Lymphoma, Large B-Cell, Diffuse/drug therapy , Lymphoma, Large B-Cell, Diffuse/mortality , MicroRNAs/genetics , Molecular Targeted Therapy , Multigene Family , Prognosis , Protein Isoforms , RNA Interference , Signal Transduction , Translocation, Genetic
4.
Oncotarget ; 7(33): 52940-52956, 2016 Aug 16.
Article in English | MEDLINE | ID: mdl-27224915

ABSTRACT

FOXP2 shares partially overlapping normal tissue expression and functionality with FOXP1; an established diffuse large B-cell lymphoma (DLBCL) oncogene and marker of poor prognosis. FOXP2 is expressed in the plasma cell malignancy multiple myeloma but has not been studied in DLBCL, where a poor prognosis activated B-cell (ABC)-like subtype display partially blocked plasma cell differentiation. FOXP2 protein expression was detected in ABC-DLBCL cell lines, and in primary DLBCL samples tumoral FOXP2 protein expression was detected in both germinal center B-cell-like (GCB) and non-GCB DLBCL. In biopsies from DLBCL patients treated with immunochemotherapy (R-CHOP), ≥ 20% nuclear tumoral FOXP2-positivity (n = 24/158) correlated with significantly inferior overall survival (OS: P = 0.0017) and progression-free survival (PFS: P = 0.0096). This remained significant in multivariate analysis against either the international prognostic index score or the non-GCB DLBCL phenotype (P < 0.05 for both OS and PFS). Expression of BLIMP1, a marker of plasmacytic differentiation that is commonly inactivated in ABC-DLBCL, did not correlate with patient outcome or FOXP2 expression in this series. Increased frequency of FOXP2 expression significantly correlated with FOXP1-positivity (P = 0.0187), and FOXP1 co-immunoprecipitated FOXP2 from ABC-DLBCL cells indicating that these proteins can co-localize in a multi-protein complex. FOXP2-positive DLBCL had reduced expression of HIP1R (P = 0.0348), which is directly repressed by FOXP1, and exhibited distinct patterns of gene expression. Specifically in ABC-DLBCL these were associated with lower expression of immune response and T-cell receptor signaling pathways. Further studies are warranted to investigate the potential functional cooperativity between FOXP1 and FOXP2 in repressing immune responses during the pathogenesis of high-risk DLBCL.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Forkhead Transcription Factors/metabolism , Lymphoma, Large B-Cell, Diffuse/drug therapy , Signal Transduction/drug effects , Adult , Aged , Aged, 80 and over , Antibodies, Monoclonal, Murine-Derived/administration & dosage , Cell Line, Tumor , Cyclophosphamide/administration & dosage , Doxorubicin/administration & dosage , Female , Forkhead Transcription Factors/genetics , Gene Expression Profiling/methods , Gene Expression Regulation, Neoplastic , Gene Ontology , Humans , Kaplan-Meier Estimate , Lymphoma, Large B-Cell, Diffuse/genetics , Lymphoma, Large B-Cell, Diffuse/metabolism , Male , Middle Aged , Prednisone/administration & dosage , Protein Binding , Repressor Proteins/genetics , Repressor Proteins/metabolism , Signal Transduction/genetics , Transcriptome/genetics , Vincristine/administration & dosage , Young Adult
5.
Haematologica ; 101(7): 861-71, 2016 07.
Article in English | MEDLINE | ID: mdl-27056922

ABSTRACT

Strong FOXP1 protein expression is a poor risk factor in diffuse large B-cell lymphoma and has been linked to an activated B-cell-like subtype, which preferentially expresses short FOXP1 (FOXP1S) proteins. However, both short isoform generation and function are incompletely understood. Here we prove by mass spectrometry and N-terminal antibody staining that FOXP1S proteins in activated B-cell-like diffuse large B-cell lymphoma are N-terminally truncated. Furthermore, a rare strongly FOXP1-expressing population of normal germinal center B cells lacking the N-terminus of the regular long protein (FOXP1L) was identified. Exon-targeted silencing and transcript analyses identified three alternate 5' non-coding exons [FOXP1-Ex6b(s), FOXP1-Ex7b and FOXP1-Ex7c], downstream of at least two predicted promoters, giving rise to FOXP1S proteins. These were differentially controlled by B-cell activation and methylation, conserved in murine lymphoma cells, and significantly correlated with FOXP1S protein expression in primary diffuse large B-cell lymphoma samples. Alternatively spliced isoforms lacking exon 9 (e.g. isoform 3) did not encode FOXP1S, and an alternate long human FOXP1 protein (FOXP1AL) likely generated from a FOXP1-Ex6b(L) transcript was detected. The ratio of FOXP1L:FOXP1S isoforms correlated with differential expression of plasmacytic differentiation markers in U-2932 subpopulations, and altering this ratio was sufficient to modulate CD19 expression in diffuse large B-cell lymphoma cell lines. Thus, the activity of multiple alternate FOXP1 promoters to produce multiple protein isoforms is likely to regulate B-cell maturation.


Subject(s)
B-Lymphocytes/metabolism , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Gene Expression Regulation, Neoplastic , Lymphoma, Large B-Cell, Diffuse/genetics , Promoter Regions, Genetic , Protein Interaction Domains and Motifs/genetics , Repressor Proteins/genetics , Repressor Proteins/metabolism , Alternative Splicing , Animals , Antigens, CD19/genetics , Antigens, CD19/metabolism , Cell Line, Tumor , Exons , Forkhead Transcription Factors/chemistry , Humans , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Lymphoma, Large B-Cell, Diffuse/pathology , Mice , Protein Isoforms , RNA, Messenger/genetics , RNA, Messenger/metabolism , Repressor Proteins/chemistry
6.
PLoS One ; 10(6): e0128513, 2015.
Article in English | MEDLINE | ID: mdl-26034982

ABSTRACT

Mutations of the forkhead transcription factor FOXP2 gene have been implicated in inherited speech-and-language disorders, and specific Foxp2 expression patterns in neuronal populations and neuronal phenotypes arising from Foxp2 disruption have been described. However, molecular functions of FOXP2 are not completely understood. Here we report a requirement for FOXP2 in growth arrest of the osteosarcoma cell line 143B. We observed endogenous expression of this transcription factor both transiently in normally developing murine osteoblasts and constitutively in human SAOS-2 osteosarcoma cells blocked in early osteoblast development. Critically, we demonstrate that in 143B osteosarcoma cells with minimal endogenous expression, FOXP2 induced by growth arrest is required for up-regulation of p21WAF1/CIP1. Upon growth factor withdrawal, FOXP2 induction occurs rapidly and precedes p21WAF1/CIP1 activation. Additionally, FOXP2 expression could be induced by MAPK pathway inhibition in growth-arrested 143B cells, but not in traditional cell line models of osteoblast differentiation (MG-63, C2C12, MC3T3-E1). Our data are consistent with a model in which transient upregulation of Foxp2 in pre-osteoblast mesenchymal cells regulates a p21-dependent growth arrest checkpoint, which may have implications for normal mesenchymal and osteosarcoma biology.


Subject(s)
Apoptosis , Bone Neoplasms/pathology , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Forkhead Transcription Factors/metabolism , Gene Expression Regulation, Neoplastic , Osteoblasts/cytology , Osteosarcoma/pathology , Animals , Blotting, Western , Bone Neoplasms/genetics , Bone Neoplasms/metabolism , Cell Cycle , Cell Proliferation , Cells, Cultured , Chromatin Immunoprecipitation , Cyclin-Dependent Kinase Inhibitor p21/genetics , Forkhead Transcription Factors/antagonists & inhibitors , Forkhead Transcription Factors/genetics , Humans , Immunoenzyme Techniques , Male , Mice , Mice, Inbred C57BL , Osteoblasts/metabolism , Osteosarcoma/genetics , Osteosarcoma/metabolism , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction
7.
Trends Immunol ; 33(2): 98-102, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22075207

ABSTRACT

Until recently, the basic leucine zipper transcription factor E4BP4 (also known as NFIL3) was of little interest to immunologists, being best known for its role in regulating circadian rhythm in chick pineal gland. However, characterisation of E4bp4(-/-) mice, independently generated in four different laboratories, has revealed roles for E4BP4 in diverse haematopoietic lineages. E4BP4 is essential for the development of NK cells and CD8α(+) conventional dendritic cells, and is also involved in macrophage activation, polarisation of CD4(+) T cell responses and B cell class switching to IgE. Here, we discuss the role of E4BP4 as a regulator of the immune response and highlight future questions for the field.


Subject(s)
Basic-Leucine Zipper Transcription Factors/immunology , Animals , Cytokines/biosynthesis , Cytokines/immunology , Dendritic Cells/immunology , Humans , Killer Cells, Natural/immunology , T-Lymphocytes, Helper-Inducer/immunology
9.
Cancer Res ; 70(10): 3985-95, 2010 May 15.
Article in English | MEDLINE | ID: mdl-20460523

ABSTRACT

A variety of genetic lesions, including chromosomal translocations, internal tandem duplications, and mutations, have been described in acute myeloid leukemia (AML). Expression profiling has shown that chromosomal translocations, in particular, are associated with distinctive patterns of gene expression. AML exhibiting the translocation t(8;21), which fuses the AML1 and ETO genes, has such a characteristic expression profile. One gene whose expression is highly correlated with the presence of the AML1/ETO fusion is POU4F1, which encodes the POU homeodomain transcription factor BRN3A. Here we show using specific siRNA in t(8;21) cells and overexpression studies in progenitor cells that AML1/ETO promotes expression of POU4F1/BRN3A. This effect requires DNA-binding function of AML1/ETO, and accordingly, AML1/ETO is bound to the POU4F1 locus in t(8;21) cells. Functionally, whereas overexpression of Brn3a in murine hematopoietic progenitor cells induces terminal myeloid differentiation, coexpression of AML1/ETO or AML1/ETO9a blocks this effect. Furthermore, Brn3a reduction by shRNA impairs AML1/ETO-induced immortalization of murine progenitors. In summary, we identify POU4F1/BRN3A as a novel potential upregulated AML1/ETO target gene whose dramatically high expression may cooperate with AML1/ETO in t(8;21) cells.


Subject(s)
Core Binding Factor Alpha 2 Subunit/metabolism , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/metabolism , Oncogene Proteins, Fusion/metabolism , Transcription Factor Brn-3A/metabolism , Adult , Animals , Base Sequence , Blotting, Western , Cell Differentiation , Chromatin Immunoprecipitation , Chromosomes, Human, Pair 21/genetics , Chromosomes, Human, Pair 8/genetics , Core Binding Factor Alpha 2 Subunit/genetics , Electrophoretic Mobility Shift Assay , Embryonic Stem Cells/cytology , Embryonic Stem Cells/physiology , Fetus , Fluorescent Antibody Technique , Hematopoietic Stem Cells/physiology , Humans , Immunoenzyme Techniques , Leukemia, Myeloid, Acute/pathology , Liver/cytology , Liver/metabolism , Luciferases/metabolism , Mice , Molecular Sequence Data , Oncogene Proteins, Fusion/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/pharmacology , RUNX1 Translocation Partner 1 Protein , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factor Brn-3A/antagonists & inhibitors , Transcription Factor Brn-3A/genetics , Transfection , Translocation, Genetic/genetics
10.
Nat Immunol ; 10(10): 1118-24, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19749763

ABSTRACT

Natural killer (NK) cells are a subset of lymphocytes crucial for innate immunity and modification of adaptive immune responses. In contrast to commitment to the T cell or B cell lineage, little is known about NK cell lineage commitment. Here we show that the basic leucine zipper (bZIP) transcription factor E4BP4 (also called NFIL3) is essential for generation of the NK cell lineage. E4BP4-deficient mice (Nfil3(-/-); called 'E4bp4(-/-)' here) had B cells, T cells and NKT cells but specifically lack NK cells and showed severely impaired NK cell-mediated cytotoxicity. Overexpression of E4bp4 was sufficient to increase NK cell production from hematopoietic progenitor cells. E4BP4 acted in a cell-intrinsic manner 'downstream' of the interleukin 15 receptor (IL-15R) and through the transcription factor Id2. E4bp4(-/-) mice may provide a model for definitive analysis of the contribution of NK cells to immune responses and pathologies.


Subject(s)
Basic-Leucine Zipper Transcription Factors/immunology , Cell Differentiation/immunology , Cytotoxicity, Immunologic/immunology , Killer Cells, Natural/cytology , Animals , B-Lymphocytes/immunology , Basic-Leucine Zipper Transcription Factors/genetics , Basic-Leucine Zipper Transcription Factors/metabolism , Cell Differentiation/genetics , Cell Lineage/genetics , Cell Lineage/immunology , Cytotoxicity, Immunologic/genetics , Flow Cytometry , Gene Expression/immunology , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/immunology , Inhibitor of Differentiation Protein 2/genetics , Inhibitor of Differentiation Protein 2/immunology , Inhibitor of Differentiation Protein 2/metabolism , Killer Cells, Natural/immunology , Lymphocyte Subsets/immunology , Mice , Mice, Knockout , Natural Killer T-Cells/immunology , Receptors, Interleukin-15/genetics , Receptors, Interleukin-15/immunology , Receptors, Interleukin-15/metabolism , Reverse Transcriptase Polymerase Chain Reaction , T-Lymphocytes/immunology
11.
Oncogene ; 23(21): 3830-40, 2004 May 06.
Article in English | MEDLINE | ID: mdl-15021903

ABSTRACT

The Brn-3 family of POU (Pit-Oct-Unc) homeodomain transcription factors regulate differentiation of neuronal cell types. The transcriptional activator Brn-3a is expressed in Ewing's sarcomas, which also express characteristic chimaeric proteins as a consequence of fusion of the TET family gene EWS to one of several ETS genes. We have previously demonstrated a physical interaction between Brn-3a and EWS proteins, and show here that the C-terminal POU domain but not N-terminal activation domain of Brn-3a can interact in vitro with the RNA-binding domain of EWS. Likely due to POU domain homology, the related factor Brn-3b can also interact with EWS, but to a lesser extent than Brn-3a. Importantly, Brn-3a but not Brn-3b interacts in vitro with chimaeric EWS/Fli-1, EWS/ATF-1 and EWS/ERG proteins. Furthermore, overexpression of EWS/Fli-1 but not EWS or Fli-1 inhibits Brn-3a-associated growth arrest and neurite outgrowth in neuronal cells, and specifically inhibits Brn-3a-dependent activation of p21 and SNAP-25 transcription. In contrast, upregulation of Bcl-2 expression and inhibition of apoptosis by Brn-3a is antagonized more by EWS than by EWS/Fli-1. These data demonstrate that oncogenic rearrangement of EWS to produce EWS/Fli-1 may enhance the antiapoptotic effect of Brn-3a and inhibit its ability to promote neuronal differentiation.


Subject(s)
Apoptosis , DNA-Binding Proteins/physiology , Neurons/cytology , Oncogene Proteins, Fusion/physiology , RNA-Binding Protein EWS/physiology , Transcription Factors/physiology , Animals , Cell Differentiation , Cells, Cultured , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/genetics , Humans , Membrane Proteins/analysis , Nerve Tissue Proteins/analysis , Neurites/physiology , Promoter Regions, Genetic , Proto-Oncogene Protein c-fli-1 , RNA/metabolism , Synaptosomal-Associated Protein 25 , Transcription Factor Brn-3 , Transcription Factor Brn-3A , Transcription Factor Brn-3B
12.
Cell Cycle ; 2(3): 238-45, 2003.
Article in English | MEDLINE | ID: mdl-12734433

ABSTRACT

Mice carrying the bovine papillomavirus type I genome develop dermal fibrosarcomas in a multiple step process characterized by distinctive proliferative stages. Chromosomal aberrations are identified early in this tumorigenic pathway, however, the mechanism that originates them is unknown. Using a functional assay, we investigated the status of the mitotic spindle cell cycle checkpoint (MSCCC) that regulates the metaphase to anaphase transition, in cells representing different stages of fibrosarcoma progression. Loss of MSCCC activity was apparent in mild fibromatosis and completely abolished in aggressive fibromatosis and fibrosarcoma lesions. This altered MSCCC status was confirmed biochemically by deregulated expression of Cks1 protein and unscheduled cyclin B metabolism. Immunoprecipitation and sequencing analyses indicated that mutation of p53 was not required for the abrogation of the MSCCC. These results demonstrate that loss of mitotic spindle checkpoint activity predisposes to chromosomal instability at early stages of fibrosarcoma development. To our knowledge, these studies constitute the first report of a transition in MSCCC activity in a tumorigenesis model.


Subject(s)
Chromosome Aberrations , DNA Damage/genetics , Fibrosarcoma/genetics , Genes, cdc/physiology , Genetic Predisposition to Disease/genetics , Mitosis/genetics , Mutation/genetics , Spindle Apparatus/genetics , Adaptor Proteins, Signal Transducing , Animals , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Transformation, Neoplastic/genetics , Cyclin B/genetics , Cyclin B/metabolism , Disease Progression , Fibroma/genetics , Mice , Models, Biological , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Tumor Cells, Cultured , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
13.
J Biol Chem ; 278(20): 18022-9, 2003 May 16.
Article in English | MEDLINE | ID: mdl-12637494

ABSTRACT

Glucocorticoids influence many physiological processes, and in particular apoptosis, often with opposite effects depending on the cell type examined. We found that during fibrosarcoma development there is a strong increase in apoptosis at the tumor stage, which is repressed by dexamethasone to levels observed in normal fibroblasts. The anti-apoptotic Bcl-2 family protein Bcl-x(L) is induced by dexamethasone at the transcriptional level at all stages of fibrosarcoma development. The ligand-activated glucocorticoid receptor (GR) activates the Bcl-x promoter in transient transfection experiments, and GR binds to specific Bcl-x promoter sequences in vitro and in vivo. Furthermore, a GR antagonist abolishes this effect, indicating that Bcl-x(L) induction is mediated by GR. Importantly, exogenous Bcl-x(L) inhibits apoptosis and caspase-3 activity in fibrosarcoma cells to levels found in dexamethasone-treated fibrosarcoma cells. We conclude that Bcl-x(L) is a key target mediating the anti-apoptotic effects of glucocorticoids during fibrosarcoma development. These observations provide further understanding of the molecular basis of glucocorticoid regulation of cell death during tumorigenesis.


Subject(s)
Apoptosis , Fibrosarcoma/metabolism , Fibrosarcoma/pathology , Gene Expression Regulation, Neoplastic , Glucocorticoids/pharmacology , Proto-Oncogene Proteins c-bcl-2/metabolism , Transcriptional Activation , Blotting, Northern , Caspases/metabolism , Cell Death , Chromatin/metabolism , DNA/metabolism , Dexamethasone/pharmacology , Disease Progression , Humans , Immunoblotting , Models, Genetic , Neoplasms/pathology , Plasmids/metabolism , Precipitin Tests , Promoter Regions, Genetic , Receptors, Glucocorticoid/metabolism , Transfection , Tumor Cells, Cultured , bcl-X Protein
SELECTION OF CITATIONS
SEARCH DETAIL
...