Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 97
Filter
1.
Vet Pathol ; 47(4): 677-89, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20442421

ABSTRACT

Human T-lymphotropic virus type-1 (HTLV-1), the first human retrovirus discovered, is the causative agent of adult T-cell leukemia/lymphoma (ATL) and a number of lymphocyte-mediated inflammatory conditions including HTLV-1-associated myelopathy/tropical spastic paraparesis. Development of animal models to study the pathogenesis of HTLV-1-associated diseases has been problematic. Mechanisms of early infection and cell-to-cell transmission can be studied in rabbits and nonhuman primates, but lesion development and reagents are limited in these species. The mouse provides a cost-effective, highly reproducible model in which to study factors related to lymphoma development and the preclinical efficacy of potential therapies against ATL. The ability to manipulate transgenic mice has provided important insight into viral genes responsible for lymphocyte transformation. Expansion of various strains of immunodeficient mice has accelerated the testing of drugs and targeted therapy against ATL. This review compares various mouse models to illustrate recent advances in the understanding of HTLV-1-associated ATL development and how improvements in these models are critical to the future development of targeted therapies against this aggressive T-cell lymphoma.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Human T-lymphotropic virus 1/immunology , Immunity, Innate/immunology , Leukemia-Lymphoma, Adult T-Cell/immunology , Paraparesis, Tropical Spastic/immunology , Animals , Disease Models, Animal , Human T-lymphotropic virus 1/genetics , Humans , Leukemia-Lymphoma, Adult T-Cell/genetics , Mice , Mice, Inbred NOD , Mice, SCID , Mice, Transgenic , Paraparesis, Tropical Spastic/genetics
2.
J Virol ; 83(22): 11467-76, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19726513

ABSTRACT

Human T-lymphotropic virus type 1 (HTLV-1) is the etiological agent of adult T-cell leukemia/lymphoma, and it encodes a number of nonstructural proteins that are involved in virus replication and immune evasion. The viral protein p12 previously has been characterized to interfere with major histocompatibility complex class, ICAM-1, and ICAM-2 expression, and it activates STAT5. Using a previously established T-cell line immortalized with an HTLV-1 molecular clone deleted for p12, we assessed the role of p12 in regulating cellular growth and virus transmission. These cells were complemented for p12 expression by the transduction of a lentivirus vector expressing p12. We report that p12 conferred a selective growth advantage in vitro and increased the colony formation of human T cells in soft-agar assays. Consistently with previous studies, p12- and p12+ cell lines produced similar amounts of virus particles released into the supernatant of cultured cells, although we found that p12 expression greatly enhanced virus transmission. Moreover, we found that interleukin-2 (IL-2) stimulation also increased HTLV-1 transmission whether p12 was expressed or not, and inversely, that the inhibition of Jak signaling significantly reduced HTLV-1 transmission. Intriguingly, IL-2/Jak signaling was not associated with changes in viral gene expression, viral RNA encapsidation, the maturation of the virus particle, cell-cell adherence, or Gag polarization and virological synapse formation. We do demonstrate, however, that IL-2 stimulation and p12 expression significantly increased the rate of syncytium formation, revealing a novel role for IL-2 signaling and Jak activation in HTLV-1 virus transmission.


Subject(s)
Human T-lymphotropic virus 1/physiology , Janus Kinases/physiology , Receptors, Interleukin-2/physiology , Viral Regulatory and Accessory Proteins/physiology , Cell Division/physiology , Cell Line, Tumor , Cell Transformation, Viral/physiology , Humans , Interleukin-2/physiology , Retroviridae Proteins, Oncogenic/physiology , STAT Transcription Factors/physiology , Signal Transduction/physiology , Virus Replication/physiology
3.
Vet Pathol ; 46(5): 1003-14, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19429977

ABSTRACT

Infection with human T-cell leukemia virus type 1 (HTLV-1) leads sometimes to the development of adult T-cell lymphoma/leukemia (ATL), which is invariably fatal and often associated with humoral hypercalcemia of malignancy. The transformation of infected CD4 T cells and the pathogenesis of leukemia have been studied with great limitation in tissue culture and patients. To better understand the pathogenesis and perform preclinical drug studies, animal models of ATL are urgently needed. In mice, inoculation of HTLV-1 cell lines mostly leads to development of localized lymphomas. To develop an ATL animal model with leukemic spread of ATL cells, mouse strains with different well-defined immune deficiencies were inoculated intraperitoneally with different HTLV-1-infected cell lines (ACH.2, C8166, MT-2, MET-1). Inoculation of MET-1 cells into NOD/SCID mice provided the best model system for slowly developing T-cell leukemia with multiple organ involvement. In leukemic mice, an increase in serum calcium levels correlated with expression of receptor activator of nuclear factor kappa-light-chain-enhancer of activated B cells ligand on leukemic cells and secretion of parathyroid hormone-related protein and interleukin-6. In contrast to the other cell lines that did not spread systemically, MET-1 expressed both the adhesion molecules CD11a (LFA-1alpha) and CD49d (VLA-4alpha) and produced or induced expression of matrix metalloproteinases 1, 2, 3, and 9, thus underlining the importance of these molecules in the spread of adult T-cell leukemia cells. The MET-1/NOD/SCID model will be useful for developing interventions against invasion and spread of leukemic cells and subsequent humoral hypercalcemia of malignancy.


Subject(s)
Disease Models, Animal , Hypercalcemia/etiology , Leukemia-Lymphoma, Adult T-Cell/immunology , Mice, Inbred NOD/immunology , Mice, SCID/immunology , Animals , Basigin/immunology , Blotting, Western/veterinary , CD11a Antigen/immunology , Cell Line , Female , Humans , Hypercalcemia/immunology , Immunohistochemistry/veterinary , Integrin alpha4/immunology , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinase 9/immunology , Mice , Mice, Knockout , Parathyroid Hormone-Related Protein/genetics , Parathyroid Hormone-Related Protein/immunology , RANK Ligand/immunology , RNA/chemistry , RNA/genetics , Receptors, Chemokine/genetics , Receptors, Chemokine/immunology , Reverse Transcriptase Polymerase Chain Reaction/veterinary
4.
Leukemia ; 19(7): 1175-83, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15889157

ABSTRACT

Parathyroid hormone-related protein (PTHrP) plays a primary role in the development of humoral hypercalcemia of malignancy seen in the majority of adult T-cell leukemia/lymphoma (ATLL) patients with human T-cell lymphotropic virus type-1 (HTLV-1) infection. HTLV-1 Tax has been shown to complex with ETS-1 and SP1 to transactivate the PTHrP P3 promoter. Previously, we established a SCID/bg mouse model of human ATL with RV-ATL cells and showed that PTHrP expression was independent of Tax. In this study, we report an inverse correlation of PTHrP with tax/rex mRNA in multiple HTLV-1-positive cell lines and RV-ATL cells. Stimulation of Jurkat T cells with PMA/ionomycin upregulated the PTHrP P3 promoter by a previously characterized Ets binding site and also induced protein/DNA complex formation identical to that observed in RV-ATL cells. Further, we provide evidence that cotransfection with Ets-1 and constitutively active Mek-1 in HTLV-1-negative transformed T cells with stimulation by PMA/ionomycin not only resulted in a robust induction of PTHrP P3 but also formed a complex with ETS-1/P3 EBS similar to that in ATLL cells. Our data demonstrate that transcriptional regulation of PTHrP in ATLL cells can be controlled by T-cell receptor signaling and the ETS and MAPK ERK pathway in a Tax-independent manner.


Subject(s)
Leukemia-Lymphoma, Adult T-Cell/metabolism , Parathyroid Hormone-Related Protein/metabolism , Promoter Regions, Genetic , Proto-Oncogene Proteins/metabolism , Transcription Factors/metabolism , Transcription, Genetic , Adult , Animals , Cell Line, Tumor , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Products, rex/genetics , Gene Products, rex/metabolism , Gene Products, tax/genetics , Gene Products, tax/metabolism , HTLV-I Infections/metabolism , HTLV-I Infections/virology , Human T-lymphotropic virus 1/genetics , Human T-lymphotropic virus 1/metabolism , Humans , Leukemia-Lymphoma, Adult T-Cell/genetics , Leukemia-Lymphoma, Adult T-Cell/virology , Male , Mice , Parathyroid Hormone-Related Protein/genetics , Proto-Oncogene Protein c-ets-1 , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-ets , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Antigen, T-Cell/metabolism , Signal Transduction/physiology , Transcription Factors/genetics
5.
Cell Death Differ ; 12 Suppl 1: 905-15, 2005 Aug.
Article in English | MEDLINE | ID: mdl-15761473

ABSTRACT

p13(II) of human T-cell leukemia virus type 1 (HTLV-1) is an 87-amino-acid protein that is targeted to the inner mitochondrial membrane. p13(II) alters mitochondrial membrane permeability, producing a rapid, membrane potential-dependent influx of K(+). These changes result in increased mitochondrial matrix volume and fragmentation and may lead to depolarization and alterations in mitochondrial Ca(2+) uptake/retention capacity. At the cellular level, p13(II) has been found to interfere with cell proliferation and transformation and to promote apoptosis induced by ceramide and Fas ligand. Assays carried out in T cells (the major targets of HTLV-1 infection in vivo) demonstrate that p13(II)-mediated sensitization to Fas ligand-induced apoptosis can be blocked by an inhibitor of Ras farnesylation, thus implicating Ras signaling as a downstream target of p13(II) function.


Subject(s)
Human T-lymphotropic virus 1 , Mitochondria/drug effects , Retroviridae Proteins/pharmacology , Amino Acid Sequence , Animals , Apoptosis/drug effects , Calcium/metabolism , Cell Death/drug effects , Cell Proliferation/drug effects , Cloning, Molecular , Human T-lymphotropic virus 1/genetics , Human T-lymphotropic virus 1/metabolism , Humans , Intracellular Membranes/drug effects , Mitochondria/physiology , Molecular Sequence Data , Retroviridae Proteins/biosynthesis , Retroviridae Proteins/genetics , Signal Transduction/drug effects
6.
J Virol ; 75(20): 9885-95, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11559821

ABSTRACT

The highly conserved coadapters CREB binding protein (CBP) and p300 form complexes with CREB as well as other DNA binding transcription factors to modulate chromatin remodeling and thus transcription. Human T-lymphotropic virus type 1 (HTLV-1) transcription is controlled, in part, by the CREB/ATF family of transcription factors which bind promoter sequences and function as complexes with the viral oncogenic protein Tax. We have reported that the nuclear localizing protein p30(II) of HTLV-1 functions as a transcription factor, differentially modulates CREB-responsive promoters, and is critical for maintenance of proviral loads in rabbits. In this study, we tested whether p30(II) directly interacts with CBP/p300 to modulate gene transcription. Gal4(BD)-p30(II)-mediated transactivation was enhanced following exogenous expression of p300 and was competitively repressed by the p300 binding protein, adenovirus E1A, and E1ACR2 (mutated for retinoblastoma binding but retaining p300 binding). In contrast, E1ACR1 (mutated for p300 binding) failed to alter Gal4(BD)-p30(II)-mediated transactivation. In addition, Gal4(BD)-p30(II)-mediated transactivation was competitively inhibited by the cotransfection of CMV-p30(II)-HA and CMV-Tax but could be rescued by exogenous p300. Importantly, we demonstrate that p30(II) colocalizes with p300 in cell nuclei and directly binds to CBP/p300 in cells. Deletion mutants of CBP/p300 were used to localize the site critical for binding p30(II) to a highly conserved KIX region. DNA binding assays confirmed the interference of p30(II) with the assembly of CREB-Tax-p300/CBP multiprotein complexes on 21-bp repeat oligonucleotides in vitro. Collectively, our results demonstrate that CBP/p300 is a cellular protein target for HTLV-1 p30(II) and mediates its transcriptional effects in vivo.


Subject(s)
Human T-lymphotropic virus 1/physiology , Nuclear Proteins/metabolism , Retroviridae Proteins/metabolism , Trans-Activators/metabolism , Transcription, Genetic , Adenovirus E1A Proteins/genetics , Adenovirus E1A Proteins/metabolism , Cell Line , Cyclic AMP Response Element-Binding Protein/metabolism , Human T-lymphotropic virus 1/genetics , Human T-lymphotropic virus 1/metabolism , Humans , Mutation , Nuclear Proteins/genetics , Protein Binding , Trans-Activators/genetics
7.
Virus Res ; 78(1-2): 17-34, 2001 Oct 30.
Article in English | MEDLINE | ID: mdl-11520577

ABSTRACT

Efficient entry into, and infection of, human cells by human T-cell leukaemia virus type-1 (HTLV-1) is mediated by the viral envelope glycoproteins, gp46 and gp21. The gp46 surface glycoprotein binds to an as yet unidentified cell surface receptor, thereby, allowing the gp21 transmembrane glycoprotein to initiate fusion of the viral and cellular membranes. In the absence of membrane fusion viral penetration and entry into the host cell cannot occur. The envelope glycoproteins are also a major target for neutralising antibodies and cytotoxic T lymphocytes following a protective immune response, and represent ideal constituents for a recombinant HTLV-1 vaccine. Given the importance of the envelope proteins in HTLV-1 pathogenesis there is increasing interest in obtaining sufficient quantities of these proteins for biochemical, biophysical and biological analyses. We have now developed a system for production of large amounts of a glycosylated and functional form of soluble recombinant gp46 (sRgp46), and have used this recombinant material for analysis of envelope function and receptor binding activity. We find that, the sRgp46 molecules expressed in our system are immunologically indistinguishable from the native virally expressed surface glycoproteins; that sRgp46 binds to T-cells in a dose dependent and saturable manner; and that cell surface binding by sRgp46 can be inhibited by neutralising antibodies. Importantly, we demonstrate that these sRgp46 molecules potently inhibit syncytia formation and viral infection of target cells, and that regions outwith the SU domain of envelope are not required for binding to target cells or for inhibiting membrane fusion. The sRgp46 produced in our study will provide new opportunities to investigate envelope-receptor interactions, and will be of utility in defining the conformationally sensitive antigenic determinants of the HTLV-1 surface glycoprotein.


Subject(s)
Gene Products, env/pharmacology , Giant Cells/drug effects , HTLV-I Antigens/pharmacology , HTLV-I Infections/virology , Human T-lymphotropic virus 1/physiology , Retroviridae Proteins, Oncogenic/pharmacology , Animals , Binding, Competitive , Cells, Cultured , Dose-Response Relationship, Drug , Drosophila , Giant Cells/virology , HTLV-I Antigens/biosynthesis , HeLa Cells , Human T-lymphotropic virus 1/pathogenicity , Humans , Jurkat Cells , Neutralization Tests , Protein Binding , Receptors, Virus/metabolism , Recombinant Proteins/pharmacology , T-Lymphocytes/metabolism
8.
J Virol ; 75(16): 7672-82, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11462039

ABSTRACT

Human T-lymphotropic virus type 1 (HTLV-1) is a complex retrovirus encoding regulatory and accessory genes in four open reading frames (ORF I to IV) of the pX region. We have demonstrated an important role of pX ORF I expression, which encodes p12(I), in establishment of HTLV-1 infection in a rabbit model and for optimal viral infectivity in quiescent primary lymphocytes. These data indicated that p12(I) may enhance lymphocyte activation and thereby promote virus infection. To further define the role of p12(I) in cell activation, we characterized the subcellular localization of p12(I) in transfected 293T cells and HeLa-Tat cells by multiple methods, including immunofluorescence confocal microscopy, electron microscopy, and subcellular fractionation. Herein, we demonstrate that p12(I) accumulates in the endoplasmic reticulum (ER) and cis-Golgi apparatus. The location of p12(I) was unchanged following treatments with both cycloheximide (blocking de novo protein synthesis) and brefeldin A (disrupting ER-to-Golgi protein transport), indicating that the protein is retained in the ER and cis-Golgi. Moreover, using coimmunoprecipitation assays, we identify the direct binding of p12(I) with both calreticulin and calnexin, resident ER proteins which regulate calcium storage. Our results indicate that p12(I) directly binds key regulatory proteins involved in calcium-mediated cell signaling and suggest a role of p12(I) in the establishment of HTLV-1 infection by activation of host cells.


Subject(s)
Calcium-Binding Proteins/physiology , HTLV-I Infections/virology , Human T-lymphotropic virus 1/physiology , Ribonucleoproteins/physiology , Biological Transport , Calnexin , Calreticulin , Cell Line , Endoplasmic Reticulum/virology , Golgi Apparatus/virology , HTLV-I Infections/metabolism , HTLV-I Infections/pathology , Humans , Virus Replication
9.
Am J Pathol ; 158(6): 2219-28, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11395400

ABSTRACT

The majority of patients with adult T-cell leukemia/lymphoma (ATL) resulting from human T-cell lymphotropic virus type-1 (HTLV-1) infection develop humoral hypercalcemia of malignancy (HHM). We used an animal model using severe combined immunodeficient (SCID)/beige mice to study the pathogenesis of HHM. SCID/beige mice were inoculated intraperitoneally with a human ATL line (RV-ATL) and were euthanized 20 to 32 days after inoculation. SCID/beige mice with engrafted RV-ATL cells developed lymphoma in the mesentery, liver, thymus, lungs, and spleen. The lymphomas stained positively for human CD45RO surface receptor and normal mouse lymphocytes stained negatively confirming the human origin of the tumors. The ATL cells were immunohistochemically positive for parathyroid hormone-related protein (PTHrP). In addition, PTHrP mRNA was highly expressed in lymphomas when compared to MT-2 cells (HTLV-1-positive cell line). Mice with lymphoma developed severe hypercalcemia. Plasma PTHrP concentrations were markedly increased in mice with hypercalcemia, and correlated with the increase in plasma calcium concentrations. Bone densitometry and histomorphometry in lymphoma-bearing mice revealed significant bone loss because of a marked increase in osteoclastic bone resorption. RV-ATL cells contained 1.5 HTLV-1 proviral copies of the tax gene as determined by quantitative real-time polymerase chain reaction (PCR). However, tax expression was not detected by Western blot or reverse transcriptase (RT)-PCR in RV-ATL cells, which suggests that factors other than Tax are modulators of PTHrP gene expression. The SCID/beige mouse model mimics HHM as it occurs in ATL patients, and will be useful to investigate the regulation of PTHrP expression by ATL cells in vivo.


Subject(s)
Disease Models, Animal , Gene Products, tax/genetics , Hypercalcemia/etiology , Leukemia-Lymphoma, Adult T-Cell/complications , Mice, SCID , Animals , Bone Density , Calcium/blood , Cell Division , Gene Products, tax/biosynthesis , Human T-lymphotropic virus 1/genetics , Humans , Hypercalcemia/genetics , Hypercalcemia/metabolism , Immunophenotyping , Leukemia-Lymphoma, Adult T-Cell/pathology , Leukemia-Lymphoma, Adult T-Cell/virology , Mice , Neoplasm Proteins/blood , Neoplasm Transplantation , Parathyroid Hormone-Related Protein , Protein Biosynthesis , Proteins/genetics , Proviruses/genetics , RNA, Messenger/biosynthesis , RNA, Neoplasm/biosynthesis , Transplantation, Heterologous
10.
J Virol ; 74(23): 11270-7, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11070026

ABSTRACT

Human T-lymphotropic virus type 1 (HTLV-1), a complex retrovirus, causes adult T-cell lymphoma/leukemia and is linked to a variety of immune-mediated disorders. The roles of proteins encoded in the pX open reading frame (ORF) II gene region in HTLV-1 replication or in mediating virus-associated diseases remain to be defined. A nucleus-localizing 30-kDa protein, p30(II), encoded within pX ORF II has limited homology with the POU family of transcription factors. Recently, we reported that selected mutations in pX ORF II diminish the ability of HTLV-1 to maintain high viral loads in infected rabbits. Herein we have tested the transcriptional ability of p30(II) in mammalian cells by using yeast Gal4 fusion protein vectors and transfection of luciferase reporter genes driven by CREB-responsive promoters. p30(II) as a Gal4 DNA-binding domain (DBD) fusion protein transactivates Gal4-driven luciferase reporter gene activity up to 25-fold in 293 and HeLa-tat cells. We confirmed nuclear localization of p30(II) and demonstrate dose-dependent binding of p30(II)-Gal4(DBD) to Gal4 DNA-binding sites. The transcriptional activity of p30(II)-Gal4(DBD) was independent of TATA box flanking sequences, as shown by using two different Gal4 reporter systems. Studies of selected p30(II) mutants indicated that domains that mediate transcription are restricted to a central core region of the protein between amino acids 62 and 220. Transfection of a p30(II)-expressing plasmid repressed cellular CRE-driven reporter gene activity, with or without Tax expression. In contrast, p30(II) at lower concentrations enhanced HTLV-1 long terminal repeat-driven reporter gene activity independent of Tax expression. These data are the first to demonstrate a transcriptional function for p30(II) and suggest a mechanism by which this nuclear protein may influence HTLV-1 replication or cellular gene expression in vivo.


Subject(s)
Cyclic AMP Response Element-Binding Protein/pharmacology , Human T-lymphotropic virus 1/genetics , Promoter Regions, Genetic , Transcription Factors/physiology , Viral Proteins/physiology , Animals , Cell Nucleus/chemistry , Gene Expression Regulation , HeLa Cells , Humans , Open Reading Frames , Rabbits , Response Elements , Terminal Repeat Sequences , Transcriptional Activation , Viral Proteins/analysis , Virus Replication
11.
AIDS Res Hum Retroviruses ; 16(16): 1661-8, 2000 Nov 01.
Article in English | MEDLINE | ID: mdl-11080807

ABSTRACT

Human T cell leukemia virus type 1 (HTLV-1) and 2 (HTLV-2) are distinct oncogenic retroviruses that infect several cell types, but display their biologic/pathogenic activity only in T lymphocytes. HTLV-1 is associated with adult T cell leukemia, a malignancy of mature CD4(+) T cells, and a chronic neurological disorder termed HTLV-1-associated myelopathy/tropical spastic paraparesis (HAM/TSP). HTLV-2 is less pathogenic and has been associated with a few cases of a variant of hairy cell leukemia and neurological disease. Previous studies have indicated that in vivo HTLV-1 has a preferential tropism for CD4(+) T cells, whereas HTLV-2 in vivo tropism is less clear, but appears to favor CD8(+) T cells. The molecular mechanism that determines the cellular tropism of HTLV-1 and HTLV-2 has not been precisely determined. However, one study by our group has provided evidence that HTLV-1-enhanced viral transcription in CD4(+) T cells may be responsible for its tropism. In an effort to understand HTLV-2 tropism we tested the ability of HTLV-2 to infect, replicate in, and transform purified CD4(+) or CD8(+) T cells in cell culture. After cocultures of purified primary human CD4(+) and CD8(+) T cells with an HTLV-2-producer cell line we measured viral transcription by reverse transcription PCR analysis, virus production by p19(gag) ELISA, proviral integration by DNA slot-blot analysis, surface phenotype by FACS analysis, and cellular transformation. We also measured HTLV-2 long terminal repeat-directed transcription in the presence and absence of Tax in purified CD4(+) and CD8(+) T cells, using transient transfection assays. Our data indicate that CD4(+) and CD8(+) T cells are equally susceptible to HTLV-2 infection. We observed no significant difference in viral transcription based on mRNA and virus production in CD4(+) and CD8(+) T cell cocultures. Although LTR transcription was enhanced 12- to 16-fold in the presence of Tax, there was no significant difference in CD4(+) or CD8(+) T cells. Interestingly, we show that HTLV-2 preferentially transforms CD8(+) T cells in culture. Together, our data indicate that, unlike HTLV-1, HTLV-2 cell tropism is not due to inhibition of viral infection and inefficient gene expression in CD4(+) versus CD8(+) T cells, and likely involves unique interactions with viral and CD8(+) T cell-specific proteins.


Subject(s)
CD4-Positive T-Lymphocytes/virology , CD8-Positive T-Lymphocytes/virology , Human T-lymphotropic virus 2/pathogenicity , Cell Transformation, Viral , Cells, Cultured , Coculture Techniques , Human T-lymphotropic virus 2/genetics , Human T-lymphotropic virus 2/physiology , Humans , Luciferases/genetics , Luciferases/metabolism , RNA, Viral/metabolism , Terminal Repeat Sequences/genetics , Transfection , Virus Replication
12.
AIDS Res Hum Retroviruses ; 16(16): 1757-64, 2000 Nov 01.
Article in English | MEDLINE | ID: mdl-11080823

ABSTRACT

Human T lymphotropic virus type 1 (HTLV-1) is a complex retrovirus containing regulatory and accessory genes encoded in four open reading frames (ORF I-IV) of the pX region. It is not clear what role pX ORFs I and II-encoded proteins have in the pathogenesis of the lymphoproliferative diseases associated with HTLV-1 infection. The conserved ORF I encodes for a hydrophobic 12-kDa protein, p12, (I) that contains four SH3 binding motifs (PXXP) that localizes to cellular endomembranes when overexpressed in cultured cells. Differential splicing of pX ORF II results in the production of two nuclear proteins, p13(II) and p30(II). p13(II) also localizes to mitochondria. p30(II) shares homology with the POU family of transcription factors. We have identified functional roles of pX ORF I and ORF II in establishment and maintenance of infection in a rabbit model. To functionally study p12(I) we have tested a proviral clone with selective ablation of ORF I (ACH.p12(I)) for its ability to infect quiescent peripheral blood mononuclear cells (PBMC). Our data indicate that T cells infected with the wild-type clone of HTLV-1 (ACH) are more efficient than ACH.p12(I) in infecting quiescent PBMC. These findings parallel our animal model data and suggest a role for p12(I) in the activation of quiescent lymphocytes, a prerequisite for effective viral replication in vivo. To test the ability of p30(II) to function as a transcription factor we have constructed p30(II) as a Gal4-fusion protein. When transfected with Gal4-driven luciferase reporter genes, the p30(II)-Gal4-fusion protein induces transcriptional activity up to 50-fold in both 293 and HeLa-Tat cells. These systems will be useful to identify molecular mechanisms that explain the functional role of pX ORF I and ORF II-encoded proteins in HTLV-1 replication.


Subject(s)
Genes, pX/genetics , Genes, pX/physiology , Human T-lymphotropic virus 1/immunology , Human T-lymphotropic virus 1/pathogenicity , Open Reading Frames , Transcription Factors , Animals , Cell Transformation, Viral , HTLV-I Infections/virology , Human T-lymphotropic virus 1/genetics , Humans , Leukocytes, Mononuclear/virology , Lymphocyte Activation , Oncogene Proteins, Viral/genetics , Oncogene Proteins, Viral/metabolism , Rabbits , Retroviridae Proteins/genetics , Retroviridae Proteins/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/virology , Transcriptional Activation , Viral Regulatory and Accessory Proteins , Virus Replication
13.
J Virol ; 74(21): 9828-35, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11024109

ABSTRACT

Human T-lymphotropic virus type 1 (HTLV-1) is a complex retrovirus encoding regulatory and accessory genes in four open reading frames (ORF I to IV) of the pX region. Emerging evidence indicates an important role for the pX ORF I-encoded accessory protein p12(I) in viral replication, but its contribution to viral pathogenesis remains to be defined. p12(I) is a conserved, membrane-associated protein containing four SH3-binding motifs (PXXP). Its interaction with the interleukin-2 (IL-2) receptor beta- and gamma-chains implies an involvement of p12(I) in intracellular signaling pathways. In addition, we have demonstrated that expression of pX ORF I p12(I) is essential for persistent infection in rabbits. In contrast, standard in vitro systems have thus far failed to demonstrate a contribution of p12(I) to viral infectivity and ultimately cellular transformation. In this study we developed multiple in vitro coculture assays to evaluate the role of p12(I) in viral infectivity in quiescent peripheral blood mononuclear cells to more accurately reflect the virus-cell interactions as they occur in vivo. Using these assays, we demonstrate a dramatic reduction in viral infectivity in quiescent T lymphocytes for a p12 mutant viral clone (ACH.p12) in comparison to the wild-type clone ACH. Moreover, addition of IL-2 and phytohemagglutinin during the infection completely rescued the ability of ACH.p12 to infect primary lymphocytes. When newly infected primary lymphocytes are used to passage virus, ACH.p12 also exhibited a reduced ability to productively infect activated lymphocytes. Our data are the first to demonstrate a functional role for pX ORF I in the infection of primary lymphocytes and suggest a role for p12(I) in activation of host cells during early stages of infection.


Subject(s)
Human T-lymphotropic virus 1/physiology , Human T-lymphotropic virus 1/pathogenicity , Oncogene Proteins, Viral/metabolism , T-Lymphocytes/virology , Transcription Factors , Animals , Antigens, Viral/metabolism , Cell Line, Transformed , Coculture Techniques , Enzyme-Linked Immunosorbent Assay , Human T-lymphotropic virus 1/genetics , Humans , Lymphocyte Activation , Oncogene Proteins, Viral/genetics , Rabbits , Transfection , Viral Regulatory and Accessory Proteins
14.
J Med Virol ; 62(2): 286-92, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11002260

ABSTRACT

Human T-lymphotropic virus type 1 (HTLV-1) infection causes adult T-cell leukemia and is characterized by long periods of clinical latency with low levels of viral production. Transcription of HTLV-1 is controlled through sequences in the promoter and enhancer regions of the long terminal repeat of the integrated provirus. Important among these sequences are three 21 bp imperfect repeats responsive to the viral oncogenic protein Tax (TRE). Members of the CREB/ATF-1/CREM family of transcription factors bind to TRE-1 and are critical for HTLV-1 transcription. Other less studied family members include the inducible cAMP early repressor (ICER) proteins. ICER proteins lack phosphorylation and activation domains and are potent inhibitors of transcription. The ability of ICER to bind TRE-1 and its effects on HTLV-1 Tax mediated transcription have not been studied in the natural cell targets of the virus, peripheral blood mononuclear cells (PBMC). We show that ICER mRNA levels are low in quiescent PBMC, but rise and remain elevated for up to 18 hr after mitogenic stimulation of these cells. Electrophoretic mobility shift assays using recombinant Tax and ICER demonstrate that ICER binds TRE-1 and that binding is increased in the presence of Tax. Furthermore, over expression of ICER IIgamma suppressed Tax-mediated transcription whereas an anti-sense ICER II plasmid designed to block endogenous ICER enhanced Tax-mediated transcription in activated PBMC. Together our data indicate that ICER inhibits Tax-mediated transcription in activated PBMC and suggest a role for ICER in maintenance of HTLV-1 persistence.


Subject(s)
DNA-Binding Proteins/metabolism , Gene Products, tax/genetics , Gene Products, tax/metabolism , Human T-lymphotropic virus 1/genetics , Leukocytes, Mononuclear/virology , Repressor Proteins , Transcription, Genetic , Adult , Cells, Cultured , Cyclic AMP Response Element Modulator , DNA-Binding Proteins/genetics , Gene Expression Regulation, Viral , Human T-lymphotropic virus 1/physiology , Humans , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Virus Latency
15.
Proc Natl Acad Sci U S A ; 97(11): 6114-9, 2000 May 23.
Article in English | MEDLINE | ID: mdl-10811912

ABSTRACT

Walleye dermal sarcoma (WDS) is a common disease of walleye fish in the United States and Canada. These proliferative lesions are present autumn through winter and regress in the spring. Walleye dermal sarcoma virus (WDSV), a retrovirus distantly related to other members of the family Retroviridae, has been etiologically linked to the development of WDS. We have reported that the D-cyclin homologue [retroviral (rv) cyclin] encoded by WDSV rescues yeast conditionally deficient for cyclin synthesis from growth arrest and that WDSV-cyclin mRNA is present in developing tumors. These data strongly suggest that the rv-cyclin plays a central role in the development of WDS. To test the ability of the WDSV rv-cyclin to induce cell proliferation, we have generated transgenic mice expressing the rv-cyclin in squamous epithelia from the bovine keratin-5 promoter. The transgenic animals were smaller than littermates, had reduced numbers of hair follicles, and transgenic females did not lactate properly. Following injury the transgenic animals developed severe squamous epithelial hyperplasia and dysplasia with ultrastructural characteristics of neoplastic squamous epithelium. Immunocytochemistry studies demonstrated that the hyperplastic epithelium stained positive for cytokeratin and were abnormally differentiated. Furthermore, the rv-cyclin protein was detected in the thickened basal cell layers of the proliferating lesions. These data are the first to indicate that the highly divergent WDSV rv-cyclin is a very potent stimulator of eukaryotic cell proliferation and to demonstrate the potential of a cyclin homologue encoded by a retrovirus to induce hyperplastic skin lesions.


Subject(s)
Cyclins/physiology , Retroviridae/pathogenicity , Skin/pathology , Alopecia/etiology , Alopecia/pathology , Animals , Cell Differentiation , Cell Division , Cyclins/genetics , Epithelial Cells/pathology , Esocidae/virology , Female , Fish Diseases/virology , Hyperplasia , Keratosis/etiology , Keratosis/pathology , Male , Mice , Mice, Transgenic , Phenotype , Retroviridae/genetics , Sarcoma/veterinary , Sarcoma/virology , Skin Ulcer/etiology , Skin Ulcer/pathology , Tail/pathology
16.
J Virol ; 74(3): 1094-100, 2000 Feb.
Article in English | MEDLINE | ID: mdl-10627519

ABSTRACT

Human T-lymphotropic virus type 1 (HTLV-1) causes adult T-cell leukemia/lymphoma and is associated with a variety of immune-mediated disorders. The role of four open reading frames (ORFs), located between env and the 3' long terminal repeat of HTLV-1, in mediating disease is not entirely clear. By differential splicing, ORF II encodes two proteins, p13(II) and p30(II), both of which have not been functionally defined. p13(II) localizes to mitochondria and may alter the configuration of the tubular network of this cellular organelle. p30(II) localizes to the nucleolus and shares homology with the transcription factors Oct-1 and -2, Pit-1, and POU-M1. Both p13(II) and p30(II) are dispensable for infection and immortalization of primary human and rabbit lymphocytes in vitro. To test the role of ORF II gene products in vivo, we inoculated rabbits with lethally irradiated cell lines expressing the wild-type molecular clone of HTLV-1 (ACH.1) or a clone containing selected mutations in ORF II (ACH.30/13.1). ACH.1-inoculated animals maintained higher HTLV-1-specific antibody titers than animals inoculated with ACH.30/13.1. Viral p19 antigen was transiently detected in ex vivo cultures of peripheral blood mononuclear cells (PBMC) from only two ACH.30/13.1-inoculated rabbits, while PBMC cultures from all ACH.1-inoculated rabbits routinely produced p19 antigen. In only three of six animals exposed to the ACH. p30(II)/p13(II) clone could provirus be consistently PCR amplified from extracted PBMC DNA and quantitative competitive PCR showed the proviral loads in PBMC from ACH.p30(II)/p13(II)-infected rabbits to be dramatically lower than the proviral loads in rabbits exposed to ACH. Our data indicate selected mutations in pX ORF II diminish the ability of HTLV-1 to maintain high viral loads in vivo and suggest an important function for p13(II) and p30(II) in viral pathogenesis.


Subject(s)
HTLV-I Infections/virology , Human T-lymphotropic virus 1/genetics , Human T-lymphotropic virus 1/physiology , Open Reading Frames/genetics , Viral Load , Animals , Blotting, Western , Cell Line , Cloning, Molecular , Enzyme-Linked Immunosorbent Assay , Gene Products, gag/blood , HTLV-I Antibodies/blood , Human T-lymphotropic virus 1/isolation & purification , Humans , Leukocytes, Mononuclear/virology , Mutation , Polymerase Chain Reaction , Proviruses , Rabbits , Retroviridae Proteins, Oncogenic/blood , gag Gene Products, Human Immunodeficiency Virus
17.
Vaccine ; 19(9-10): 1068-81, 2000 Dec 08.
Article in English | MEDLINE | ID: mdl-11137241

ABSTRACT

The ability of a peptide vaccine derived from the human T-lymphotropic virus type 1 (HTLV-1) surface envelope glycoprotein protein (gp46) to mimic the native protein and elicit a protective immune response has been examined. This peptide construct, designated MVFMF2, comprises amino acids (aa) 175-218 of gp46 linked by a four residue turn (GPSL) to a promiscuous T-cell epitope from the measles virus fusion protein (MVF, aa 288-302). The peptide was structurally characterized by circular dichroism (CD) spectroscopy and was found to contain alpha-helical secondary structure. The immunogenicity of MVFMF2 in rabbits and mice was evaluated by direct ELISA and competitive ELISA using peptide constructs and the recombinant protein ACH-RE3 (aa 165-306). This peptide, when administered with adjuvant (N-acetyl-glucosamine-3yl-acetyl-L-alanyl-D-isoglutamine, nor-MDP) was immunogenic in an outbred population of both rabbits and mice. Furthermore, the peptide construct was encapsulated in biodegradable microspheres of poly(D,L-lactide-co-glycolide) to eliminate booster immunization and to examine adjuvant requirements. The data indicate that MVFMF2 shows enhanced immunogenicity when encapsulated in biodegradable microspheres. Inoculation of the encapsulated peptide produced a similar humoral response to that of the free peptide, but did not require the use of adjuvant. Elicited anti-rabbit and anti-mouse antibodies recognized whole viral preparations and the recombinant protein ACH-RE3 in ELISA assays. Additionally, inoculated rabbits exhibited enhanced reactivity to viral antigens by western blot compared to non-vaccinated controls. Although anti-rabbit and anti-mouse antibodies were capable of inhibiting syncytium formation at low dilutions, rabbits were not protected from cell-associated viral challenge. Future development of vaccines to HTLV-1 may need to incorporate the ability to elicit cell-mediated immune responses in order to protect against cell-associated viral infection.


Subject(s)
Human T-lymphotropic virus 1/immunology , Measles virus/immunology , Peptide Fragments/immunology , Vaccines, Synthetic/immunology , Viral Envelope Proteins/immunology , Viral Fusion Proteins/immunology , Viral Vaccines/immunology , Amino Acid Sequence , Animals , Antibodies, Viral/biosynthesis , Epitopes , Male , Mice , Mice, Inbred ICR , Molecular Sequence Data , Protein Folding , Rabbits , T-Lymphocytes, Cytotoxic/immunology , Viral Envelope Proteins/chemistry
18.
J Virol ; 73(11): 9642-9, 1999 Nov.
Article in English | MEDLINE | ID: mdl-10516077

ABSTRACT

Human T-cell lymphotropic virus type 1 (HTLV-1), a complex retrovirus, encodes a hydrophobic 12-kD protein from pX open reading frame (ORF) I that localizes to cellular endomembranes and contains four minimal SH3 binding motifs (PXXP). We have demonstrated the importance of ORF I expression in the establishment of infection and hypothesize that p12(I) has a role in T-cell activation. In this study, we tested interleukin-2 (IL-2) receptor expression, IL-2-mediated proliferation, and Jak/Stat activation in T-cell lines immortalized with either wild-type or ORF I mutant clones of HTLV-1. All cell lines exhibited typical patterns of T-cell markers and maintained mutation fidelity. No significant differences between cell lines were observed in IL-2 receptor chain (alpha, beta, or gamma(c)) expression, in IL-2-mediated proliferation, or in IL-2-induced phosphorylated forms of Stat3, Stat5, Jak1, or Jak3. The expression of ORF I is more likely to play a role in early HTLV-1 infection, such as in the activation of quiescent T cells in vivo.


Subject(s)
Human T-lymphotropic virus 1/physiology , Interleukin-2/pharmacology , Lymphocyte Activation , Protein-Tyrosine Kinases/metabolism , T-Lymphocytes/immunology , Trans-Activators/metabolism , Cell Line, Transformed , Cell Transformation, Viral , Human T-lymphotropic virus 1/genetics , Human T-lymphotropic virus 1/immunology , Humans , Open Reading Frames/genetics , Phosphorylation , Receptors, Interleukin-2/metabolism , T-Lymphocytes/metabolism
19.
Can J Vet Res ; 63(3): 207-11, 1999 Jul.
Article in English | MEDLINE | ID: mdl-10480463

ABSTRACT

In order to investigate the role of retroviruses in the recently described porcine postweaning multisystemic wasting syndrome (PMWS) serum and leukocytes were screened for reverse transcriptase (RT) activity, and tissues were examined for the presence of conserved lentiviral sequences using degenerate primers in a polymerase chain reaction (PCR). Serum and stimulated leukocytes from the blood and lymph nodes from pigs with PMWS, as well as from control pigs had RT activity that was detected by the sensitive Amp-RT assay. A 257-bp fragment was amplified from DNA from the blood and bone marrow of pigs with PMWS. This fragment was identical in size to conserved lentiviral sequences that were amplified from plasmids containing DNA from several lentiviruses. Cloning and sequencing of the fragment from affected pigs, however, did not reveal homology with the recognized lentiviruses. Together the results of these analyses suggest that the RT activity present in tissues from control and affected pigs is the result of endogenous retrovirus expression, and that a lentivirus is not a primary pathogen in PMWS.


Subject(s)
Lentivirus/genetics , Swine Diseases/virology , Wasting Syndrome/veterinary , Amino Acid Sequence , Animals , DNA, Viral/analysis , Lentivirus/pathogenicity , Molecular Sequence Data , Polymerase Chain Reaction , Sequence Alignment , Sequence Analysis, DNA , Swine , Swine Diseases/genetics , Wasting Syndrome/genetics , Wasting Syndrome/virology
20.
Article in English | MEDLINE | ID: mdl-9928723

ABSTRACT

HTLV-I is the etiologic agent of adult T-cell leukemia/lymphoma and is associated with tropical spastic paraparesis/HTLV-I-associated myelopathy. Following integration into the host cell genome, HTLV-I replication is regulated by both host and viral mechanisms that control transcription. Low levels of viral transcription (basal transcription) occur before expression of the virally encoded Tax protein (Tax-mediated transcription). Members of the cyclic adenosine monophosphate (cAMP) response element binding (CREB)/activating transcription factor 1 (ATF-1) family of transcription factors bind three 21-bp repeats (Tax-responsive element-1, or TRE-1) within the viral promoter and are important for basal and Tax-mediated transcription. Using mitogen stimulated and quiescent peripheral blood mononuclear cells (PBMC) and Jurkat cells, we compared differences in basal transcription and amounts and binding of transcription factors with TRE-1. We demonstrate that amounts of transcriptionally active phosphorylated CREB protein (P-CREB) differ between activated PBMC and Jurkat cells. Following stimulation, P-CREB levels remain elevated in PBMC for up to 24 hours whereas CREB is dephosphorylated in Jurkat cells within 4 hours following stimulation. The differences in P-CREB levels between PBMC and Jurkat cells were directly correlated with basal transcription of HTLV-I in the two cell types. Using electrophoretic mobility shift assays, we determined that the pattern of band migration differed between the two cell types. These data demonstrate that PBMC differentially regulate basal HTLV-I transcription compared with Jurkat T cells, and this differential regulation is due, in part to differential phosphorylation and binding of CREB/ATF-1 to TRE-1 in the HTLV-I promoter. We demonstrate the utility of using primary lymphocyte models to study HTLV-I transcription in the context of cell signaling and suggest that activated PBMC maintain elevated levels of P-CREB, which promote basal HTLV-I transcription and enhance viral persistence in vivo.


Subject(s)
DNA-Binding Proteins , Human T-lymphotropic virus 1/genetics , Leukocytes, Mononuclear/virology , Transcription Factors/genetics , Transcription, Genetic , Activating Transcription Factor 1 , Humans , Immunoblotting , Jurkat Cells , Response Elements , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...