Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
2.
Oncogene ; 36(18): 2589-2598, 2017 05 04.
Article in English | MEDLINE | ID: mdl-27819671

ABSTRACT

Eradication of leukemia stem cells (LSCs) is the ultimate goal of treating acute myeloid leukemia (AML). We recently showed that the combined loss of Runx1/Cbfb inhibited the development of MLL-AF9-induced AML. However, c-Kit+/Gr-1- cells remained viable in Runx1/Cbfb-deleted cells, indicating that suppressing RUNX activity may not eradicate the most immature LSCs. In this study, we found upregulation of several hemostasis-related genes, including the thrombin-activatable receptor PAR-1 (protease-activated receptor-1), in Runx1/Cbfb-deleted MLL-AF9 cells. Similar to the effect of Runx1/Cbfb deletion, PAR-1 overexpression induced CDKN1A/p21 expression and attenuated proliferation in MLL-AF9 cells. To our surprise, PAR-1 deficiency also prevented leukemia development induced by a small number of MLL-AF9 leukemia stem cells (LSCs) in vivo. PAR-1 deficiency also reduced leukemogenicity of AML1-ETO-induced leukemia. Re-expression of PAR-1 in PAR-1-deficient cells combined with a limiting-dilution transplantation assay demonstrated the cell-dose-dependent role of PAR-1 in MLL-AF9 leukemia: PAR-1 inhibited rapid leukemic proliferation when there were a large number of LSCs, while a small number of LSCs required PAR-1 for their efficient growth. Mechanistically, PAR-1 increased the adherence properties of MLL-AF9 cells and promoted their engraftment to bone marrow. Taken together, these data revealed a multifaceted role for PAR-1 in leukemogenesis, and highlight this receptor as a potential target to eradicate primitive LSCs in AML.


Subject(s)
Core Binding Factor Alpha 2 Subunit/genetics , Cyclin-Dependent Kinase Inhibitor p21/genetics , Leukemia, Myeloid, Acute/genetics , Receptor, PAR-1/genetics , Animals , Cell Line, Tumor , Cell Proliferation/genetics , Disease Models, Animal , Gene Expression Regulation, Neoplastic , Humans , Leukemia, Myeloid, Acute/pathology , Mice , Neoplastic Stem Cells/pathology , Receptor, PAR-1/biosynthesis
3.
Leukemia ; 30(3): 728-39, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26449661

ABSTRACT

The t(8;21) rearrangement, which creates the AML1-ETO fusion protein, represents the most common chromosomal translocation in acute myeloid leukemia (AML). Clinical data suggest that CBL mutations are a frequent event in t(8;21) AML, but the role of CBL in AML1-ETO-induced leukemia has not been investigated. In this study, we demonstrate that CBL mutations collaborate with AML1-ETO to expand human CD34+ cells both in vitro and in a xenograft model. CBL depletion by shRNA also promotes the growth of AML1-ETO cells, demonstrating the inhibitory function of endogenous CBL in t(8;21) AML. Mechanistically, loss of CBL function confers hyper-responsiveness to thrombopoietin and enhances STAT5/AKT/ERK/Src signaling in AML1-ETO cells. Interestingly, we found the protein tyrosine phosphatase UBASH3B/Sts-1, which is known to inhibit CBL function, is upregulated by AML1-ETO through transcriptional and miR-9-mediated regulation. UBASH3B/Sts-1 depletion induces an aberrant pattern of CBL phosphorylation and impairs proliferation in AML1-ETO cells. The growth inhibition caused by UBASH3B/Sts-1 depletion can be rescued by ectopic expression of CBL mutants, suggesting that UBASH3B/Sts-1 supports the growth of AML1-ETO cells partly through modulation of CBL function. Our study reveals a role of CBL in restricting myeloid proliferation of human AML1-ETO-induced leukemia, and identifies UBASH3B/Sts-1 as a potential target for pharmaceutical intervention.


Subject(s)
Core Binding Factor Alpha 2 Subunit/genetics , Gene Expression Regulation, Leukemic , Leukemia, Myeloid, Acute/genetics , Oncogene Proteins, Fusion/genetics , Preleukemia/genetics , Protein Tyrosine Phosphatases/genetics , Proto-Oncogene Proteins c-cbl/genetics , Animals , Cell Proliferation , Chromosomes, Human, Pair 21 , Chromosomes, Human, Pair 8 , Core Binding Factor Alpha 2 Subunit/metabolism , Extracellular Signal-Regulated MAP Kinases/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Fetal Blood/cytology , Fetal Blood/drug effects , Fetal Blood/metabolism , Heterografts , Humans , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Mice , Mice, SCID , MicroRNAs/genetics , MicroRNAs/metabolism , Myeloid Cells/cytology , Myeloid Cells/drug effects , Myeloid Cells/metabolism , Oncogene Proteins, Fusion/metabolism , Preleukemia/metabolism , Preleukemia/pathology , Protein Tyrosine Phosphatases/metabolism , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-cbl/antagonists & inhibitors , Proto-Oncogene Proteins c-cbl/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , RUNX1 Translocation Partner 1 Protein , STAT5 Transcription Factor/genetics , STAT5 Transcription Factor/metabolism , Thrombopoietin/pharmacology , Transgenes , Translocation, Genetic , src-Family Kinases/genetics , src-Family Kinases/metabolism
4.
Oncogene ; 34(27): 3483-92, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25263451

ABSTRACT

The transcription factor RUNX1 is a master regulator of hematopoiesis. Disruption of RUNX1 activity has been implicated in the development of hematopoietic neoplasms. Recent studies also highlight the importance of RUNX1 in solid tumors both as a tumor promoter and a suppressor. Given its central role in cancer development, RUNX1 is an excellent candidate for targeted therapy. A potential strategy to target RUNX1 is through modulation of its posttranslational modifications (PTMs). Numerous studies have shown that RUNX1 activity is regulated by PTMs, including phosphorylation, acetylation, methylation and ubiquitination. These PTMs regulate RUNX1 activity either positively or negatively by altering RUNX1-mediated transcription, promoting protein degradation and affecting protein interactions. In this review, we first summarize the available data on the context- and dosage-dependent roles of RUNX1 in various types of neoplasms. We then provide a comprehensive overview of RUNX1 PTMs from biochemical and biologic perspectives. Finally, we discuss how aberrant PTMs of RUNX1 might contribute to tumorigenesis and also strategies to develop anticancer therapies targeting RUNX1 PTMs.


Subject(s)
Antineoplastic Agents/therapeutic use , Core Binding Factor Alpha 2 Subunit/metabolism , Molecular Targeted Therapy/methods , Protein Processing, Post-Translational , Animals , Base Sequence , Carcinogenesis/genetics , Carcinogenesis/metabolism , Core Binding Factor Alpha 2 Subunit/chemistry , Core Binding Factor Alpha 2 Subunit/genetics , Hematologic Neoplasms/drug therapy , Hematologic Neoplasms/genetics , Humans , Lysine/genetics , Molecular Sequence Data , Protein Processing, Post-Translational/drug effects , Protein Processing, Post-Translational/genetics , Tyrosine/genetics
6.
Leukemia ; 26(6): 1329-37, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22289984

ABSTRACT

The AML1-ETO fusion protein, which is present in 10-15% of cases of acute myeloid leukemia, is known to repress myeloid differentiation genes through DNA binding and recruitment of chromatin-modifying proteins and transcription factors in target genes. ChIP-chip analysis of human hematopoietic stem/progenitor cells transduced with the AML1-ETO fusion gene enabled us to identify 1168 AML1-ETO target genes, 103 of which were co-occupied by histone deacetylase 1 (HDAC1) and had lost the hyperacetylation mark at histone H4, and 264 showed a K9 trimethylation at histone H3. Enrichment of genes involved in hematopoietic differentiation and in specific signaling pathways was observed in the presence of these epigenetic modifications associated with an 'inactive' chromatin status. Furthermore, AML1-ETO target genes had a significant correlation between the chromatin marks studied and transcriptional silencing. Interestingly, AML1 binding sites were absent on a large number of selected AML1-ETO promoters and an Sp1 binding site was found in over 50% of them. Reversible silencing induced by the fusion protein in the presence of AML1 and/or Sp1 transcription factor binding site was confirmed. Therefore, this study provides a global analysis of AML1-ETO functional chromatin modifications and identifies the important role of Sp1 in the DNA binding pattern of AML1-ETO, suggesting a role for Sp1-targeted therapy in this leukemia subtype.


Subject(s)
Chromatin/genetics , Core Binding Factor Alpha 2 Subunit/genetics , Core Binding Factor Alpha 2 Subunit/metabolism , Hematopoietic Stem Cells/metabolism , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/metabolism , Oncogene Proteins, Fusion/genetics , Oncogene Proteins, Fusion/metabolism , Sp1 Transcription Factor/metabolism , Acetylation , Binding Sites , Cells, Cultured , Chromatin Immunoprecipitation , Core Binding Factor Alpha 2 Subunit/antagonists & inhibitors , Epigenesis, Genetic , Genomics , Hematopoietic Stem Cells/cytology , Histone Deacetylase 1/genetics , Histone Deacetylase 1/metabolism , Histones/metabolism , Humans , Oncogene Proteins, Fusion/antagonists & inhibitors , Promoter Regions, Genetic/genetics , RNA, Messenger/genetics , RUNX1 Translocation Partner 1 Protein , Real-Time Polymerase Chain Reaction , Sp1 Transcription Factor/antagonists & inhibitors , Sp1 Transcription Factor/genetics , Umbilical Cord/cytology , Umbilical Cord/metabolism
7.
Leukemia ; 26(2): 244-54, 2012 Feb.
Article in English | MEDLINE | ID: mdl-21836612

ABSTRACT

The DNA hypomethylating drug decitabine maintains normal hematopoietic stem cell (HSC) self-renewal but induces terminal differentiation in acute myeloid leukemia (AML) cells. The basis for these contrasting cell fates, and for selective CpG hypomethylation by decitabine, is poorly understood. Promoter CpGs, with methylation measured by microarray, were classified by the direction of methylation change with normal myeloid maturation. In AML cells, the methylation pattern at maturation-responsive CpGs suggested at least partial maturation. Consistent with partial maturation, in gene expression analyses, AML cells expressed high levels of the key lineage-specifying factor CEBPA, but relatively low levels of the key late-differentiation driver CEBPE. In methylation analysis by mass spectrometry, CEBPE promoter CpGs that are usually hypomethylated during granulocyte maturation were significantly hypermethylated in AML cells. Decitabine-induced hypomethylation was greatest at these and other promoter CpGs that are usually hypomethylated with myeloid maturation, accompanied by cellular differentiation of AML cells. In contrast, decitabine-treated normal HSCs retained immature morphology, and methylation significantly decreased at CpGs that are less methylated in immature cells. High expression of lineage-specifying factor and aberrant epigenetic repression of some key late-differentiation driver genes distinguishes AML cells from normal HSCs, and could explain the contrasting differentiation and methylation responses to decitabine.


Subject(s)
Antineoplastic Agents/therapeutic use , Azacitidine/analogs & derivatives , CpG Islands , DNA Methylation , Hematopoietic Stem Cells/metabolism , Leukemia, Myeloid, Acute/drug therapy , Azacitidine/therapeutic use , Base Sequence , Cell Lineage , DNA Primers , Decitabine , Humans , Leukemia, Myeloid, Acute/pathology , Polymerase Chain Reaction , Promoter Regions, Genetic
8.
Leukemia ; 25(11): 1739-50, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21701495

ABSTRACT

Suppression of apoptosis by TP53 mutation contributes to resistance of acute myeloid leukemia (AML) to conventional cytotoxic treatment. Using differentiation to induce irreversible cell cycle exit in AML cells could be a p53-independent treatment alternative, however, this possibility requires evaluation. In vitro and in vivo regimens of the deoxycytidine analogue decitabine that deplete the chromatin-modifying enzyme DNA methyl-transferase 1 without phosphorylating p53 or inducing early apoptosis were determined. These decitabine regimens but not equimolar DNA-damaging cytarabine upregulated the key late differentiation factors CCAAT enhancer-binding protein ɛ and p27/cyclin dependent kinase inhibitor 1B (CDKN1B), induced cellular differentiation and terminated AML cell cycle, even in cytarabine-resistant p53- and p16/CDKN2A-null AML cells. Leukemia initiation by xenotransplanted AML cells was abrogated but normal hematopoietic stem cell engraftment was preserved. In vivo, the low toxicity allowed frequent drug administration to increase exposure, an important consideration for S phase specific decitabine therapy. In xenotransplant models of p53-null and relapsed/refractory AML, the non-cytotoxic regimen significantly extended survival compared with conventional cytotoxic cytarabine. Modifying in vivo dose and schedule to emphasize this pathway of decitabine action can bypass a mechanism of resistance to standard therapy.


Subject(s)
Epigenesis, Genetic , Genes, p53 , Leukemia, Myeloid, Acute/drug therapy , Transplantation, Heterologous , Animals , Antineoplastic Agents/therapeutic use , Apoptosis , Azacitidine/analogs & derivatives , Azacitidine/therapeutic use , Blotting, Western , Cell Differentiation , Cytarabine/therapeutic use , DNA Damage , Decitabine , Electrophoresis, Polyacrylamide Gel , Humans , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Mice , Phosphorylation
10.
Cell Mol Life Sci ; 66(3): 370-4, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19151919

ABSTRACT

Rac1, a member of the Rho family of GTPases, is an intracellular transducer known to regulate multiple signaling pathways that control cytoskeleton organization, transcription, and cell proliferation. Deregulated expression or activation patterns of Rac1 can result in aberrant cell signaling and numerous pathological conditions. Here, we highlight the physiological functions and signaling mechanisms of Rac1 and their relevance to disease.


Subject(s)
Protein Isoforms/metabolism , Signal Transduction/physiology , rac GTP-Binding Proteins/metabolism , Actins/metabolism , Animals , Endocytosis/physiology , Humans , Protein Isoforms/genetics , Reactive Oxygen Species/metabolism , rac GTP-Binding Proteins/genetics
11.
Blood ; 98(3): 823-9, 2001 Aug 01.
Article in English | MEDLINE | ID: mdl-11468184

ABSTRACT

The p12(I) protein, encoded by the pX open reading frame I of the human T-lymphotropic virus type 1 (HTLV-1), is a hydrophobic protein that localizes to the endoplasmic reticulum and the Golgi. Although p12(I) contains 4 minimal proline-rich, src homology 3-binding motifs (PXXP), a characteristic commonly found in proteins involved in signaling pathways, it has not been known whether p12(I) has a role in modulating intracellular signaling pathways. This study demonstrated that p12(I) binds to the cytoplasmic domain of the interleukin-2 receptor (IL-2R) beta chain that is involved in the recruitment of the Jak1 and Jak3 kinases. As a result of this interaction, p12(I) increases signal transducers and activators of transcription 5 (STAT5) DNA binding and transcriptional activity and this effect depends on the presence of both IL-2R beta and gamma(c) chains and Jak3. Transduction of primary human peripheral blood mononuclear cells (PBMCs) with a human immunodeficiency virus type 1-based retroviral vector expressing p12(I) also resulted in increased STAT5 phosphorylation and DNA binding. However, p12(I) could increase proliferation of human PBMCs only after stimulation of T-cell receptors by treatment of cells with low concentrations of alphaCD3 and alphaCD28 antibodies. In addition, the proliferative advantage of p12(I)-transduced PBMCs was evident mainly at low concentrations of IL-2. Together, these data indicate that p12(I) may confer a proliferative advantage on HTLV-1-infected cells in the presence of suboptimal antigen stimulation and that this event may account for the clonal proliferation of infected T cells in vivo. (Blood. 2001;98:823-829)


Subject(s)
DNA-Binding Proteins/drug effects , Interleukin-2/pharmacology , Milk Proteins , Oncogene Proteins, Viral/pharmacology , T-Lymphocytes/virology , Trans-Activators/drug effects , Transcription Factors , Cell Culture Techniques , Cell Division/drug effects , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Drug Synergism , HTLV-I Infections/metabolism , HTLV-I Infections/pathology , Humans , Protein Binding , Receptors, Interleukin-2/metabolism , STAT5 Transcription Factor , Signal Transduction/drug effects , T-Lymphocytes/cytology , T-Lymphocytes/drug effects , Trans-Activators/genetics , Trans-Activators/metabolism , Transcriptional Activation/drug effects , Viral Regulatory and Accessory Proteins
12.
J Virol ; 75(13): 6086-94, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11390610

ABSTRACT

Human T-cell leukemia virus type 1 (HTLV-1) establishes a persistent infection in the host despite a vigorous virus-specific immune response. Here we demonstrate that an HTLV-1-encoded protein, p12(I), resides in the endoplasmic reticulum (ER) and Golgi and physically binds to the free human major histocompatibility complex class I heavy chains (MHC-I-Hc) encoded by the HLA-A2, -B7, and -Cw4 alleles. As a result of this interaction, the newly synthesized MHC-I-Hc fails to associate with beta(2)-microglobulin and is retrotranslocated to the cytosol, where it is degraded by the proteasome complex. Targeting of the free MHC-I-Hc, and not the MHC-I-Hc-beta(2)-microglobulin complex, by p12(I) represents a novel mechanism of viral interference and disrupts the intracellular trafficking of MHC-I, which results in a significant decrease in surface levels of MHC-I on human T-cells. These findings suggest that the interaction of p12(I) with MHC-1-Hc may interfere with antigen presentation in vivo and facilitate escape of HTLV-1-infected cells from immune recognition.


Subject(s)
Histocompatibility Antigens Class I/metabolism , Oncogene Proteins, Viral/physiology , Transcription Factors , Biological Transport , Cysteine Endopeptidases/physiology , Endoplasmic Reticulum/metabolism , Golgi Apparatus/metabolism , HLA-A2 Antigen/metabolism , HLA-B7 Antigen/metabolism , HLA-C Antigens/metabolism , HeLa Cells , Humans , Multienzyme Complexes/physiology , Proteasome Endopeptidase Complex , Viral Regulatory and Accessory Proteins , beta 2-Microglobulin/metabolism
13.
AIDS Res Hum Retroviruses ; 16(16): 1777-81, 2000 Nov 01.
Article in English | MEDLINE | ID: mdl-11080826

ABSTRACT

Human T cell leukemia/lymphotropic virus types 1 and 2 are two immunologically and phylogenetically related retroviruses that differ in their pathogenicity in vivo. The overall genetic structure of HTLV-1 and -2 is similar. Each contains a unique region at the 3' end of the genome, designated the pX region. p12(I) is a membrane-associated protein encoded by the open reading frame I (ORF I) region of HTLV-1, which lies within the pX region. A corresponding protein, p10(I) is encoded by the ORF I region of HTLV-2 and an additional protein, p11(V), is encoded by ORF V, which overlaps the HTLV-2 ORF I region. As with HTLV-1, the small proteins encoded by the pX region of HTLV-2 appear to be dispensable for viral replication and cellular transformation in vitro. However, the small open reading frames of both viruses are important for viral replication in vivo, which suggests they may play an important role during the viral life cycle. This study was undertaken to investigate and compare the cellular targets of the p10(I), p11(V), and p12(I) putative proteins.


Subject(s)
Histocompatibility Antigens Class I/metabolism , Human T-lymphotropic virus 1/metabolism , Human T-lymphotropic virus 2/metabolism , Retroviridae Proteins/genetics , Retroviridae Proteins/metabolism , 3' Untranslated Regions/genetics , Cell Line, Transformed , HTLV-I Infections/virology , HTLV-II Infections/virology , HeLa Cells , Human T-lymphotropic virus 1/genetics , Human T-lymphotropic virus 1/pathogenicity , Human T-lymphotropic virus 2/genetics , Human T-lymphotropic virus 2/pathogenicity , Humans , Proton-Translocating ATPases/metabolism , Receptors, Interleukin-2/metabolism , T-Lymphocytes/virology
14.
Virology ; 274(1): 86-93, 2000 Aug 15.
Article in English | MEDLINE | ID: mdl-10936091

ABSTRACT

The mechanism of T-cell transformation by human T-cell lymphotropic virus type I (HTLV-I), though not completely understood, appears to involve the interactions of several viral and cellular proteins. One of these viral proteins, p12(I), encoded by HTLV-I orfI, is a weak oncogene that binds the 16-kDa subunit of the vacuolar ATPase and interacts with the immature beta and gamma(c) chains of the IL-2 receptor. We have expressed the singly spliced orfI cDNA in the baculovirus system and used the recombinant protein as a tool to assess the presence of antibodies in naturally or experimentally infected hosts. In addition, rabbit antisera were raised against various p12(I) synthetic peptides and used to identify three antigenic regions within p12(I), one between the two putative transmembrane regions of p12(I) and two at the carboxy-terminus of the protein. More importantly, sera from a naturally infected human (1 of 32) and experimentally infected rabbits (9 of 20) recognized the rp12(I), demonstrating orfI expression and immunogenicity in vivo. Taken together these data provide the first evidence of orfI expression during HTLV-I infections.


Subject(s)
Deltaretrovirus Antibodies/immunology , Deltaretrovirus Antigens/immunology , Human T-lymphotropic virus 1/immunology , Oncogene Proteins, Viral/immunology , Transcription Factors , Amino Acid Sequence , Animals , Cell Line , Deltaretrovirus Antibodies/blood , Deltaretrovirus Antigens/genetics , Disease Models, Animal , HTLV-I Infections/blood , HTLV-I Infections/immunology , HeLa Cells , Human T-lymphotropic virus 1/genetics , Humans , Molecular Sequence Data , Oncogene Proteins, Viral/genetics , Peptides/immunology , Precipitin Tests , Rabbits , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Spodoptera/cytology , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology , Viral Regulatory and Accessory Proteins
15.
Biochem Pharmacol ; 58(6): 935-50, 1999 Sep 15.
Article in English | MEDLINE | ID: mdl-10509746

ABSTRACT

Intracellular cyclic AMP, determined in part by cyclic nucleotide phosphodiesterases (PDEs), regulates proliferation and immune functions in lymphoid cells. Total PDE, PDE3, and PDE4 activities were measured in phytohemagglutinin (PHA)-activated peripheral blood mononuclear cells (PBMC-PHA), normal natural killer (NK) cells, Jurkat and Kit225-K6 leukemic T-cells, T-cell lines transformed with human T-lymphotropic virus (HTLV)-I (a retrovirus that causes adult T-cell leukemia/lymphoma) and HTLV-II (a nonpathogenic retrovirus), normal B-cells, and B-cells transformed with Epstein-Barr virus (EBV). All cells exhibited PDE3 and PDE4 activities but in different proportions. In EBV-transformed B cells, PDE4 was much higher than PDE3. HTLV-I+ T-cells differed significantly from other T-lymphocyte-derived cells in also having a higher proportion of PDE4 activities, which apparently were not related to selective induction of any one PDE4 mRNA (judged by reverse transcription-polymerase chain reaction) or expression of the HTLV-I regulatory protein Tax. In MJ cells (an HTLV-I+ T-cell line), Jurkat cells, and PBMC-PHA cells, the tyrosine kinase inhibitor herbimycin A strongly inhibited PDE activity. Growth of MJ cells was inhibited by herbimycin A and a protein kinase C (PKC) inhibitor, and was arrested in G1 by rolipram, a specific PDE4 inhibitor. Proliferation of several HTLV-I+ T-cell lines, PBMC-PHA, and Jurkat cells was inhibited differentially by forskolin (which activates adenylyl cyclase), the selective PDE inhibitors cilostamide and rolipram, and the nonselective PDE inhibitors pentoxifylline and isobutyl methylxanthine. These results suggest that PDE4 isoforms may be functionally up-regulated in HTLV-I+ T-cells and may contribute to the virus-induced proliferation, and that PDEs could be therapeutic targets in immune/inflammatory and neoplastic diseases.


Subject(s)
3',5'-Cyclic-AMP Phosphodiesterases/metabolism , Cell Transformation, Viral , Human T-lymphotropic virus 1/physiology , Lymphocytes/enzymology , 3',5'-Cyclic-AMP Phosphodiesterases/antagonists & inhibitors , 3',5'-Cyclic-AMP Phosphodiesterases/genetics , Adult , B-Lymphocytes/enzymology , Benzoquinones , Cell Division/drug effects , Cell Line, Transformed/enzymology , Colforsin/pharmacology , Cyclic Nucleotide Phosphodiesterases, Type 3 , Cyclic Nucleotide Phosphodiesterases, Type 4 , Enzyme Inhibitors/pharmacology , Gene Products, tax/biosynthesis , Gene Products, tax/metabolism , Humans , Interleukin-2/metabolism , Jurkat Cells/enzymology , Killer Cells, Natural/enzymology , Lactams, Macrocyclic , Leukocytes, Mononuclear/enzymology , Lymphocytes/virology , Protein Kinase Inhibitors , Quinones/pharmacology , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Rifabutin/analogs & derivatives , T-Lymphocytes/enzymology
16.
J Virol ; 73(8): 6460-7, 1999 Aug.
Article in English | MEDLINE | ID: mdl-10400740

ABSTRACT

The HTLV-1 singly spliced open reading frame I protein, p12(I), is highly unstable and appears to be necessary for persistent infection in rabbits. Here we demonstrate that p12(I) forms dimers through two putative leucine zipper domains and that its stability is augmented by specific proteasome inhibitors. p12(I) is ubiquitylated, and mutations of its unique carboxy-terminus lysine residue to an arginine greatly enhance its stability. Interestingly, analysis of 53 independent HTLV-1 strains revealed that the natural p12(I) alleles found in ex vivo samples of tropical spastic paraparesis-HTLV-1-associated myelopathy patients contain a Lys at position 88 in some cases, whereas arginine is consistently found at position 88 in HTLV-1 strains from all adult T-cell leukemia-lymphoma (ATLL) cases and healthy carriers studied. This apparent segregation of different alleles in tropical spastic paraparesis-HTLV-associated myelopathy and ATLL or healthy carriers may be relevant in vivo, since p12(I) binds the interleukin-2 receptor beta and gammac chains, raising the possibility that the two natural alleles might affect differently the regulation of these molecules.


Subject(s)
Alleles , Amino Acid Substitution , Arginine/genetics , Human T-lymphotropic virus 1/genetics , Human T-lymphotropic virus 1/physiology , Leucine Zippers , Leukemia-Lymphoma, Adult T-Cell/virology , Lysine/genetics , Oncogene Proteins, Viral/genetics , Paraparesis, Tropical Spastic/virology , Transcription Factors , Adult , Amino Acid Sequence , Arginine/metabolism , Arginine/physiology , Binding Sites , Carrier State , Cysteine Endopeptidases/metabolism , Human T-lymphotropic virus 1/metabolism , Humans , Lysine/metabolism , Lysine/physiology , Molecular Sequence Data , Multienzyme Complexes/metabolism , Oncogene Proteins, Viral/metabolism , Oncogene Proteins, Viral/physiology , Proteasome Endopeptidase Complex , Ubiquitins , Viral Regulatory and Accessory Proteins
17.
J Virol ; 72(11): 8852-60, 1998 Nov.
Article in English | MEDLINE | ID: mdl-9765430

ABSTRACT

Human T-cell lymphotropic/leukemia virus type 1 (HTLV-1) transforms human T cells in vitro, and Tax, a potent transactivator of viral and cellular genes, plays a key role in cell immortalization. Tax activity is mediated by interaction with cellular transcription factors including members of the CREB/ATF family, the NF-kappaB/c-Rel family, serum response factor, and the coactivators CREB binding protein-p300. Although p53 is usually not mutated in HTLV-1-infected T cells, its half-life is increased and its function is impaired. Here we report that transient coexpression of p53 and Tax results in the suppression of p53 transcriptional activity. Expression of Tax abrogates p53-induced G1 arrest in the Calu-6 cell line and prevents the apoptosis induced by overexpressing p53 in the HeLa/Tat cell line. The Tax mutants M22 and G148V, which selectively activate the CREB/ATF pathway, exert these same biological effects on p53 function. In contrast, the NF-kappaB-active Tax mutant M47 has no effect on p53 activity in any of these systems. Consistent with the negative effect of Tax on p53, no activity on a p53-responsive promoter was observed upon transfection of HTLV-1-infected T-cell lines. The p53 protein is expressed at high levels in the nucleus, and nuclear extracts of HTLV-1-infected T cells bind constitutively to a DNA oligonucleotide containing the p53 response element, indicating that Tax does not interfere with p53 binding to DNA. Tax is able to suppress the transactivation function of p53 in three different cell lines, and this suppression required Tax-mediated activation of the CREB/ATF, but not the NF-kappaB/c-Rel, pathway. Tax and the active Tax mutants were able to abrogate the G1 arrest and apoptosis induced by p53, and this effect does not correlate with an altered localization of nuclear p53 or with the disruption of p53-DNA complexes. The suppression of p53 activity by Tax could be important in T-cell immortalization induced by HTLV-1.


Subject(s)
Cyclic AMP Response Element-Binding Protein/physiology , Gene Products, tax/physiology , Human T-lymphotropic virus 1/pathogenicity , Transcription Factors/physiology , Tumor Suppressor Protein p53/physiology , Activating Transcription Factor 2 , Apoptosis , Base Sequence , Cell Cycle , Cell Line , Cell Nucleus/metabolism , Cell Transformation, Viral , Cyclic AMP Response Element-Binding Protein/genetics , DNA/metabolism , Gene Products, tax/genetics , Genes, pX , Human T-lymphotropic virus 1/genetics , Human T-lymphotropic virus 1/physiology , Humans , Jurkat Cells , Plasmids/genetics , Transcription Factors/genetics , Transcription, Genetic , Tumor Suppressor Protein p53/antagonists & inhibitors
18.
J Virol ; 72(5): 4408-12, 1998 May.
Article in English | MEDLINE | ID: mdl-9557732

ABSTRACT

Human T-lymphotropic virus type 1 (HTLV-1) and HTLV-2 differ in pathogenicity in vivo. HTLV-1 causes leukemia and neurologic and inflammatory diseases, whereas HTLV-2 is less clearly associated with human disease. Both retroviruses transform human T cells in vitro, and transformation by HTLV-1 was found to be associated with the constitutive activation of the Jak/STAT pathway. To assess whether HTLV-2 transformation may also result in constitutive activation of the Jak/STAT pathway, six interleukin-2-independent, HTLV-2-transformed T-cell lines were analyzed for the presence of activated Jak and STAT proteins by electrophoretic mobility shift assay. In addition, the phosphorylation status of Jak and STAT proteins was assessed directly by immunoprecipitation and immunoblotting with an antiphosphotyrosine antibody. Jak/STAT proteins were not found to be constitutively activated in any of the T-cell lines infected by the type 2 human and nonhuman primate viruses, suggesting that HTLV-2 and the cognate virus simian T-lymphotropic virus type 2 from Pan paniscus transform T cells in vitro by mechanisms at least partially different from those used by HTLV-1.


Subject(s)
Cell Transformation, Viral , DNA-Binding Proteins/metabolism , Human T-lymphotropic virus 2/physiology , Milk Proteins , Protein-Tyrosine Kinases/metabolism , Simian T-lymphotropic virus 1/physiology , Trans-Activators/metabolism , Animals , Caseins/genetics , Cell Line, Transformed , Genes, fos , Haplorhini , Human T-lymphotropic virus 1/physiology , Humans , Janus Kinase 1 , Janus Kinase 3 , Phenotype , Promoter Regions, Genetic , Receptors, IgG/genetics , STAT1 Transcription Factor , STAT3 Transcription Factor , STAT5 Transcription Factor
19.
Proc Natl Acad Sci U S A ; 94(25): 13897-902, 1997 Dec 09.
Article in English | MEDLINE | ID: mdl-9391124

ABSTRACT

Human T cell leukemia/lymphotropic virus type I (HTLV-I) induces adult T cell leukemia/lymphoma (ATLL). The mechanism of HTLV-I oncogenesis in T cells remains partly elusive. In vitro, HTLV-I induces ligand-independent transformation of human CD4+ T cells, an event that correlates with acquisition of constitutive phosphorylation of Janus kinases (JAK) and signal transducers and activators of transcription (STAT) proteins. However, it is unclear whether the in vitro model of HTLV-I transformation has relevance to viral leukemogenesis in vivo. Here we tested the status of JAK/STAT phosphorylation and DNA-binding activity of STAT proteins in cell extracts of uncultured leukemic cells from 12 patients with ATLL by either DNA-binding assays, using DNA oligonucleotides specific for STAT-1 and STAT-3, STAT-5 and STAT-6 or, more directly, by immunoprecipitation and immunoblotting with anti-phosphotyrosine antibody for JAK and STAT proteins. Leukemic cells from 8 of 12 patients studied displayed constitutive DNA-binding activity of one or more STAT proteins, and the constitutive activation of the JAK/STAT pathway was found to persist over time in the 2 patients followed longitudinally. Furthermore, an association between JAK3 and STAT-1, STAT-3, and STAT-5 activation and cell-cycle progression was demonstrated by both propidium iodide staining and bromodeoxyuridine incorporation in cells of four patients tested. These results imply that JAK/STAT activation is associated with replication of leukemic cells and that therapeutic approaches aimed at JAK/STAT inhibition may be considered to halt neoplastic growth.


Subject(s)
DNA-Binding Proteins/metabolism , Leukemia-Lymphoma, Adult T-Cell/metabolism , Leukemia-Lymphoma, Adult T-Cell/pathology , Milk Proteins , Protein-Tyrosine Kinases/metabolism , Trans-Activators/metabolism , Adult , Base Sequence , Cell Division , DNA Probes/genetics , Enzyme Activation , Human T-lymphotropic virus 1/pathogenicity , Humans , Janus Kinase 3 , Leukemia-Lymphoma, Adult T-Cell/genetics , STAT1 Transcription Factor , STAT3 Transcription Factor , STAT5 Transcription Factor , Time Factors
20.
Blood ; 88(5): 1551-60, 1996 Sep 01.
Article in English | MEDLINE | ID: mdl-8781409

ABSTRACT

Human T-cell lymphotropic/leukemia virus type I (HTLV-I) is associated with T-cell transformation both in vivo and in vitro. Although some of the mechanisms responsible for transformation remain unknown, increasing evidence supports a direct role of viral as well as dysregulated cellular proteins in transformation. We investigated the potential role of the tumor suppressor gene p53 and of the p53-regulated gene, p21waf1/cip1 (wild-type p53 activated fragment 1/cycling dependent kinases [cdks] interacting protein 1), in HTLV-I-infected T cells. We have found that the majority of HTLV-I-infected T cells have the wild-type p53 gene. However, its function in HTLV-I-transformed cells appears to be impaired, as shown by the lack of appropriate p53-mediated responses to ionizing radiation (IR). Interestingly, the expression of the p53 inducible gene, p21waf1/cip1, is elevated at the messenger ribonucleic acid and protein levels in all HTLV-I-infected T-cell lines examined as well as in Taxl-1, a human T-cell line stably expressing Tax. Additionally, Tax induces upregulation of a p21waf1/cip1 promoter-driven luciferase gene in p53 null cells, and increases p21waf1/cip1 expression in Jurkat T cells. These findings suggest that the Tax protein is at least partially responsible for the p53-independent expression of p21waf1/cip1 in HTLV-I-infected cells. Dysregulation of p53 and p21waf1/cip1 proteins regulating cell-cycle progression, may represent an important step in HTLV-I-induced T-cell transformation.


Subject(s)
Cell Cycle/physiology , Cell Transformation, Viral , Cyclins/biosynthesis , Gene Expression Regulation, Viral , Human T-lymphotropic virus 1/physiology , T-Lymphocytes/metabolism , Tumor Suppressor Protein p53/physiology , Base Sequence , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/genetics , DNA Damage , DNA Repair , Gene Expression Regulation, Viral/radiation effects , Gene Products, tax/physiology , Genes, p53 , Humans , Interleukin-2/pharmacology , Intracellular Signaling Peptides and Proteins , Molecular Sequence Data , Protein Biosynthesis , Proteins/genetics , T-Lymphocytes/radiation effects , T-Lymphocytes/virology , GADD45 Proteins
SELECTION OF CITATIONS
SEARCH DETAIL
...