Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
1.
Genes Dev ; 31(23-24): 2343-2360, 2017 12 01.
Article in English | MEDLINE | ID: mdl-29326336

ABSTRACT

The oncogenic transcription factor TAL1/SCL induces an aberrant transcriptional program in T-cell acute lymphoblastic leukemia (T-ALL) cells. However, the critical factors that are directly activated by TAL1 and contribute to T-ALL pathogenesis are largely unknown. Here, we identified AT-rich interactive domain 5B (ARID5B) as a collaborating oncogenic factor involved in the transcriptional program in T-ALL. ARID5B expression is down-regulated at the double-negative 2-4 stages in normal thymocytes, while it is induced by the TAL1 complex in human T-ALL cells. The enhancer located 135 kb upstream of the ARID5B gene locus is activated under a superenhancer in T-ALL cells but not in normal T cells. Notably, ARID5B-bound regions are associated predominantly with active transcription. ARID5B and TAL1 frequently co-occupy target genes and coordinately control their expression. ARID5B positively regulates the expression of TAL1 and its regulatory partners. ARID5B also activates the expression of the oncogene MYC Importantly, ARID5B is required for the survival and growth of T-ALL cells, and forced expression of ARID5B in immature thymocytes results in thymus retention, differentiation arrest, radioresistance, and tumor formation in zebrafish. Our results indicate that ARID5B reinforces the oncogenic transcriptional program by positively regulating the TAL1-induced regulatory circuit and MYC in T-ALL, thereby contributing to T-cell leukemogenesis.


Subject(s)
Carcinogenesis/genetics , DNA-Binding Proteins/metabolism , Gene Expression Regulation, Neoplastic , T-Cell Acute Lymphocytic Leukemia Protein 1/metabolism , Transcription Factors/metabolism , Animals , Cell Line, Tumor , Cell Survival/genetics , DNA-Binding Proteins/genetics , Enhancer Elements, Genetic/genetics , Gene Expression Profiling , Genes, myc/genetics , HEK293 Cells , Humans , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma , Protein Binding , Protein Domains/genetics , Thymocytes/metabolism , Thymus Gland/growth & development , Transcription Factors/genetics , Transcriptional Activation/genetics , Zebrafish
2.
Blood ; 135(12): 934-947, 2020 03 19.
Article in English | MEDLINE | ID: mdl-31972002

ABSTRACT

Adult T-cell leukemia/lymphoma (ATL) is a highly aggressive hematological malignancy derived from mature CD4+ T-lymphocytes. Here, we demonstrate the transcriptional regulatory network driven by 2 oncogenic transcription factors, IRF4 and NF-κB, in ATL cells. Gene expression profiling of primary ATL samples demonstrated that the IRF4 gene was more highly expressed in ATL cells than in normal T cells. Chromatin immunoprecipitation sequencing analysis revealed that IRF4-bound regions were more frequently found in super-enhancers than in typical enhancers. NF-κB was found to co-occupy IRF4-bound regulatory elements and formed a coherent feed-forward loop to coordinately regulate genes involved in T-cell functions and development. Importantly, IRF4 and NF-κB regulated several cancer genes associated with super-enhancers in ATL cells, including MYC, CCR4, and BIRC3. Genetic inhibition of BIRC3 induced growth inhibition in ATL cells, implicating its role as a critical effector molecule downstream of the IRF4-NF-κB transcriptional network.


Subject(s)
Interferon Regulatory Factors/metabolism , Leukemia-Lymphoma, Adult T-Cell/etiology , Leukemia-Lymphoma, Adult T-Cell/metabolism , NF-kappa B/metabolism , Signal Transduction , Apoptosis/genetics , Baculoviral IAP Repeat-Containing 3 Protein/metabolism , Cell Line, Tumor , Cell Proliferation , Cell Survival/genetics , Computational Biology , Gene Expression Profiling , Gene Knockdown Techniques , Humans , Models, Biological , RNA, Small Interfering/genetics , Receptors, CCR4/metabolism
3.
Blood ; 134(3): 239-251, 2019 07 18.
Article in English | MEDLINE | ID: mdl-31076442

ABSTRACT

The oncogenic transcription factor TAL1 regulates the transcriptional program in T-ALL. ARID5B is one of the critical downstream targets of TAL1, which further activates the oncogenic regulatory circuit in T-ALL cells. Here, we elucidated the molecular functions of the noncoding RNA, ARID5B-inducing enhancer associated long noncoding RNA (ARIEL), in T-ALL pathogenesis. We demonstrated that ARIEL is specifically activated in TAL1 + T-ALL cases, and its expression is associated with ARID5B enhancer activity. ARIEL recruits mediator proteins to the ARID5B enhancer, promotes enhancer-promoter interactions, and activates the expression of ARID5B, thereby positively regulating the TAL1-induced transcriptional program and the MYC oncogene. The TAL1 complex coordinately regulates the expression of ARIEL Knockdown of ARIEL inhibits cell growth and survival of T-ALL cells in culture and blocks disease progression in a murine xenograft model. Our results indicate that ARIEL plays an oncogenic role as an enhancer RNA in T-ALL.


Subject(s)
Carcinogenesis/genetics , Gene Expression Regulation, Leukemic , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , RNA, Long Noncoding/genetics , Transcription, Genetic , Animals , Binding Sites , Cell Line, Tumor , Cell Proliferation , Cell Survival/genetics , Chromatin Immunoprecipitation Sequencing , DNA-Binding Proteins/metabolism , Disease Models, Animal , Disease Progression , Enhancer Elements, Genetic , Gene Knockdown Techniques , Gene Targeting , Heterografts , Humans , Mice , Models, Biological , Multiprotein Complexes , Oncogenes , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Promoter Regions, Genetic , Protein Binding , T-Cell Acute Lymphocytic Leukemia Protein 1/metabolism , Transcription Factors/metabolism
4.
Blood ; 130(21): 2326-2338, 2017 11 23.
Article in English | MEDLINE | ID: mdl-28978570

ABSTRACT

A number of studies have recently demonstrated that super-enhancers, which are large cluster of enhancers typically marked by a high level of acetylation of histone H3 lysine 27 and mediator bindings, are frequently associated with genes that control and define cell identity during normal development. Super-enhancers are also often enriched at cancer genes in various malignancies. The identification of such enhancers would pinpoint critical factors that directly contribute to pathogenesis. In this study, we performed enhancer profiling using primary leukemia samples from adult T-cell leukemia/lymphoma (ATL), which is a genetically heterogeneous intractable cancer. Super-enhancers were enriched at genes involved in the T-cell activation pathway, including IL2RA/CD25, CD30, and FYN, in both ATL and normal mature T cells, which reflected the origin of the leukemic cells. Super-enhancers were found at several known cancer gene loci, including CCR4, PIK3R1, and TP73, in multiple ATL samples, but not in normal mature T cells, which implicated those genes in ATL pathogenesis. A small-molecule CDK7 inhibitor, THZ1, efficiently inhibited cell growth, induced apoptosis, and downregulated the expression of super-enhancer-associated genes in ATL cells. Furthermore, enhancer profiling combined with gene expression analysis identified a previously uncharacterized gene, TIAM2, that was associated with super-enhancers in all ATL samples, but not in normal T cells. Knockdown of TIAM2 induced apoptosis in ATL cell lines, whereas overexpression of this gene promoted cell growth. Our study provides a novel strategy for identifying critical cancer genes.


Subject(s)
Enhancer Elements, Genetic/genetics , Gene Expression Profiling , Genes, Neoplasm , Leukemia-Lymphoma, Adult T-Cell/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Cell Survival/genetics , Cyclin-Dependent Kinases/antagonists & inhibitors , Cyclin-Dependent Kinases/metabolism , Down-Regulation/drug effects , Down-Regulation/genetics , Gene Expression Regulation, Leukemic/drug effects , Genetic Association Studies , Humans , Leukemia-Lymphoma, Adult T-Cell/drug therapy , Leukemia-Lymphoma, Adult T-Cell/immunology , Leukemia-Lymphoma, Adult T-Cell/pathology , Lymphocyte Activation/genetics , Phenylenediamines/pharmacology , Phenylenediamines/therapeutic use , Phosphorylation/drug effects , Protein Kinase Inhibitors/pharmacology , Pyrimidines/pharmacology , Pyrimidines/therapeutic use , RNA Polymerase II/metabolism , T-Lymphocytes/drug effects , T-Lymphocytes/metabolism , Cyclin-Dependent Kinase-Activating Kinase
5.
Cell Metab ; 33(5): 1027-1041.e8, 2021 05 04.
Article in English | MEDLINE | ID: mdl-33770508

ABSTRACT

Mitochondria have an independent genome (mtDNA) and protein synthesis machinery that coordinately activate for mitochondrial generation. Here, we report that the Krebs cycle intermediate fumarate links metabolism to mitobiogenesis through binding to malic enzyme 2 (ME2). Mechanistically, fumarate binds ME2 with two complementary consequences. First, promoting the formation of ME2 dimers, which activate deoxyuridine 5'-triphosphate nucleotidohydrolase (DUT). DUT fosters thymidine generation and an increase of mtDNA. Second, fumarate-induced ME2 dimers abrogate ME2 monomer binding to mitochondrial ribosome protein L45, freeing it for mitoribosome assembly and mtDNA-encoded protein production. Methylation of the ME2-fumarate binding site by protein arginine methyltransferase-1 inhibits fumarate signaling to constrain mitobiogenesis. Notably, acute myeloid leukemia is highly dependent on mitochondrial function and is sensitive to targeting of the fumarate-ME2 axis. Therefore, mitobiogenesis can be manipulated in normal and malignant cells through ME2, an unanticipated governor of mitochondrial biomass production that senses nutrient availability through fumarate.


Subject(s)
Fumarates/metabolism , Malate Dehydrogenase/metabolism , Mitochondria/metabolism , Animals , Cell Line , Citric Acid Cycle , DNA, Mitochondrial/metabolism , Dimerization , Humans , Leukemia/pathology , Leukemia/veterinary , Malate Dehydrogenase/antagonists & inhibitors , Malate Dehydrogenase/genetics , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mitochondria/genetics , Protein Binding , Protein-Arginine N-Methyltransferases/antagonists & inhibitors , Protein-Arginine N-Methyltransferases/genetics , Protein-Arginine N-Methyltransferases/metabolism , Pyrophosphatases/metabolism , RNA Interference , RNA, Small Interfering/metabolism , Ribosomal Proteins/metabolism , Thymidine/metabolism
6.
Exp Hematol ; 53: 7-15, 2017 09.
Article in English | MEDLINE | ID: mdl-28652130

ABSTRACT

In hematopoietic cell development, the transcriptional program is strictly regulated in a lineage- and stage-specific manner that requires a number of transcription factors to work in a cascade or in a loop, in addition to interactions with nonhematopoietic cells in the microenvironment. Disruption of the transcriptional program alters the cellular state and may predispose cells to the acquisition of genetic abnormalities. Early studies have shown that proteins that promote cell differentiation often serve as tumor suppressors, whereas inhibitors of those proteins act as oncogenes in the context of acute leukemia. A prime example is T-cell acute lymphoblastic leukemia (T-ALL), a malignant disorder characterized by clonal proliferation of immature stage thymocytes. Although a relatively small number of genetic abnormalities are observed in T-ALL, these abnormalities are crucial for leukemogenesis. Many oncogenes and tumor suppressors in T-ALL are transcription factors that are required for normal hematopoiesis. The transformation process in T-ALL is efficient and orchestrated; the oncogene disrupts the transcriptional program directing T-cell differentiation and also uses its native ability as a master transcription factor in hematopoiesis. This imbalance in the transcriptional program is a primary determinant underlying the molecular pathogenesis of T-ALL. In this review, we focus on the oncogenic transcription factor TAL1 and the tumor-suppressor E-proteins and discuss the malignant cell state, the transcriptional circuit, and the consequence of molecular abnormalities in T-ALL.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/physiology , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/etiology , Proto-Oncogene Proteins/physiology , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Differentiation , GTP-Binding Proteins/physiology , Hematopoiesis , Humans , Membrane Proteins/physiology , Mice , Oncogenes , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Proto-Oncogene Proteins/genetics , Receptors, Antigen, T-Cell/physiology , T-Cell Acute Lymphocytic Leukemia Protein 1 , Tumor Suppressor Proteins/physiology
7.
Oncotarget ; 5(13): 4881-94, 2014 Jul 15.
Article in English | MEDLINE | ID: mdl-24952679

ABSTRACT

Endoplasmic reticulum stress from unfolded proteins is associated with the proliferation of pancreatic tumor cells, making the many regulatory molecules of this pathway appealing targets for therapy. The objective of our study was to assess potential therapeutic efficacy of inhibitors of unfolded protein response (UPR) in pancreatic cancers focusing on IRE1α inhibitors. IRE1α-mediated XBP-1 mRNA splicing encodes a transcription factor that enhances transcription of chaperone proteins in order to reverse UPR. Proliferation assays using a panel of 14 pancreatic cancer cell lines showed a dose- and time-dependent growth inhibition by IRE1α-specific inhibitors (STF-083010, 2-Hydroxy-1-naphthaldehyde, 3-Ethoxy-5,6-dibromosalicylaldehyde, toyocamycin). Growth inhibition was also noted using a clonogenic growth assay in soft agar, as well as a xenograft in vivo model of pancreatic cancer. Cell cycle analysis showed that these IRE1α inhibitors caused growth arrest at either the G1 or G2/M phases (SU8686, MiaPaCa2) and induced apoptosis (Panc0327, Panc0403). Western blot analysis showed cleavage of caspase 3 and PARP, and prominent induction of the apoptotic molecule BIM. In addition, synergistic effects were found between either STF-083010, 2-Hydroxy-1-naphthaldehyde, 3-Ethoxy-5,6-dibromosalicylaldehyde, or toyocamycin and either gemcitabine or bortezomib. Our data suggest that use of an IRE1α inhibitor is a novel therapeutic approach for treatment of pancreatic cancers.


Subject(s)
Apoptosis/drug effects , Enzyme Inhibitors/pharmacology , Pancreatic Neoplasms/drug therapy , Unfolded Protein Response/drug effects , Animals , Blotting, Western , Boronic Acids/pharmacology , Bortezomib , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Deoxycytidine/analogs & derivatives , Deoxycytidine/pharmacology , Drug Synergism , Endoribonucleases/antagonists & inhibitors , Endoribonucleases/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Humans , Mice, Inbred NOD , Mice, SCID , Naphthalenes/pharmacology , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/metabolism , Pyrazines/pharmacology , RNA Interference , RNA Splicing/drug effects , Regulatory Factor X Transcription Factors , Reverse Transcriptase Polymerase Chain Reaction , Sulfonamides/pharmacology , Thiophenes/pharmacology , Toyocamycin/pharmacology , Transcription Factors/genetics , Transcription Factors/metabolism , Unfolded Protein Response/genetics , X-Box Binding Protein 1 , Xenograft Model Antitumor Assays , Gemcitabine
SELECTION OF CITATIONS
SEARCH DETAIL