Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 175
Filter
1.
PLoS Pathog ; 17(5): e1009577, 2021 05.
Article in English | MEDLINE | ID: mdl-34019588

ABSTRACT

Human T-cell leukemia virus type 1 (HTLV-1) is a retrovirus that causes an aggressive T-cell malignancy and a variety of inflammatory conditions. The integrated provirus includes a single binding site for the epigenomic insulator, CCCTC-binding protein (CTCF), but its function remains unclear. In the current study, a mutant virus was examined that eliminates the CTCF-binding site. The mutation did not disrupt the kinetics and levels of virus gene expression, or establishment of or reactivation from latency. However, the mutation disrupted the epigenetic barrier function, resulting in enhanced DNA CpG methylation downstream of the CTCF binding site on both strands of the integrated provirus and H3K4Me3, H3K36Me3, and H3K27Me3 chromatin modifications both up- and downstream of the site. A majority of clonal cell lines infected with wild type HTLV-1 exhibited increased plus strand gene expression with CTCF knockdown, while expression in mutant HTLV-1 clonal lines was unaffected. These findings indicate that CTCF binding regulates HTLV-1 gene expression, DNA and histone methylation in an integration site dependent fashion.


Subject(s)
Epigenesis, Genetic , Genome, Viral/genetics , HTLV-I Infections/virology , Human T-lymphotropic virus 1/genetics , Leukemia, T-Cell/virology , Binding Sites , CCCTC-Binding Factor/genetics , CCCTC-Binding Factor/metabolism , Cell Line , Chromatin/genetics , DNA Methylation , Epigenomics , Human T-lymphotropic virus 1/physiology , Humans , Mutation , Virus Integration , Virus Latency/genetics
2.
PLoS Negl Trop Dis ; 15(2): e0009066, 2021 02.
Article in English | MEDLINE | ID: mdl-33544713

ABSTRACT

Human T-cell leukemia virus type 1 (HTLV-1) has worldwide distribution and is considered endemic in southwestern Japan. HTLV-1 infection has been associated with adult T-cell leukemia/lymphoma (ATL) and HTLV-1-associated myelopathy/tropical spastic paraparesis (HAM/TSP) besides other diseases. This cross-sectional study aimed to investigate the prevalence, risk factors and molecular characterization of HTLV-1, among the world's largest population of Japanese immigrants and their descendants outside of Japan, in São Paulo, Southeast Brazil, as well as to analyze the phylogenetic relationship among isolates of HTLV-1. From July to December 2017, 2,139 individuals from five Japanese associations were interviewed and submitted to blood collection. All serum samples were first tested for the presence of anti-HTLV-1/2 antibodies by ELISA and then peripheral blood from individuals with positive serological results were analyzed for the presence of HTLV-1 5'LTR proviral DNA. Partial sequencing of the 5'LTR region of HTLV-1 proviral DNA was performed by Sanger. The prevalence of HTLV-1 infection was 5.1% (CI 95%: 4.2-6.0). In the multiple logistic regression model, HTLV-1 infection was associated with age ≥ 45 years, female sex, being first and second-generation Japanese immigrants, and having sexual partners with history of blood transfusion. The phylogenetic analysis revealed that all HTLV-1 were classified as Cosmopolitan (1a) subtype. Of them, 47.8% were classified as Transcontinental (A) subgroup and 52.2% as belonging to the Japanese (B) subgroup. Although most HTLV-1-infected patients were asymptomatic (97.3%), blurred vision was associated with HTLV-1 infection. The high prevalence of HTLV-1 infection found in this studied population and especially the intra- and interfamily HTLV-1 transmission presents an urgent call for preventive and control responses of this infection in Brazil.


Subject(s)
Emigrants and Immigrants , HTLV-I Infections/epidemiology , Human T-lymphotropic virus 1 , Leukemia, T-Cell/epidemiology , Leukemia, T-Cell/prevention & control , Adult , Asymptomatic Diseases , Brazil/epidemiology , Cross-Sectional Studies , Enzyme-Linked Immunosorbent Assay , Female , Human T-lymphotropic virus 1/classification , Human T-lymphotropic virus 1/genetics , Humans , Japan , Leukemia, T-Cell/virology , Male , Middle Aged , Molecular Epidemiology , Paraparesis, Tropical Spastic/virology , Pedigree , Phylogeny , Prevalence , Proviruses , Risk Factors
3.
Emerg Microbes Infect ; 9(1): 913-923, 2020 Dec.
Article in English | MEDLINE | ID: mdl-32249692

ABSTRACT

Africa is the largest endemic area for HTLV-1, with many molecular genotypes. We previously demonstrated that some strains from North Africa (a-NA clade) originated from a recombinant event between Senegalese and West African strains. A series of 52 new HTLV-1 strains from 13 North and West African countries were sequenced in the LTR region and/or a env gene fragment. Four samples from French Guyanese of African origin were also added. Furthermore, 7 complete sequences from different genotypes were characterized. Phylogenetic analyses showed that most of the new African strains belong to the Cosmopolitan a-genotype. Ten new strains from the a-NA clade were found in Morocco, Western Sahara, Mali, Guinea, Côte d'Ivoire and Ghana. A new a-G-Rec clade, which arose from a distinct recombination event between Senegalese and West African strains, was identified in Guinea and Ghana. The complete sequences suggest that recombination occur in the LTR as well as the env/pol region of the genome, thus a-NA and a-G-Rec strains have a mosaic profile with genetic segments from either a-WA or a-Sen strains. Our work demonstrates that recombination in HTLV-1 may not be as rare an event as previously proposed.


Subject(s)
Human T-lymphotropic virus 1/genetics , Terminal Repeat Sequences/genetics , Africa/epidemiology , DNA, Viral , Genome, Viral , Genotype , Human T-lymphotropic virus 1/isolation & purification , Humans , Leukemia, T-Cell/virology , Phylogeny , Phylogeography , Recombination, Genetic
4.
Mol Cancer Res ; 17(12): 2522-2536, 2019 12.
Article in English | MEDLINE | ID: mdl-31594868

ABSTRACT

The genetic and molecular alterations responsible for leukemogenesis and progression of HTLV-infected adult T-cell leukemia (ATL) have not been fully clarified. Previously, we reported that various genes are not only overexpressed but also abnormally spliced in ATL cells. Here, we identified various CASP8 transcript variants in PBMCs from a smoldering-type ATL patient, which encode aberrant truncated caspase 8 (Casp8) isoforms. Among those, we focus on the three transcript variants, CASP8L (including the first 136 bp of the intron 8 between exon 8 and exon 9), CASP8-ΔE4 (without the exon 4), and CASP8-ΔE7 (without the exon 7), because they encode isoforms, Casp8L, Casp8-ΔE4, and Casp8-ΔE7, respectively, without the C-terminal catalytic domains. In this study, we conducted in vitro characterization and functional analysis of those mutant Casp8 isoforms to clarify their changed functions compared with the wild-type (WT)-Casp8. We demonstrated that these abnormal Casp8 isoforms showed lower ability to induce apoptosis than WT-Casp8 due to their dominant-negative interactions with WT-Casp8, which impair WT-Casp8 homodimerization that is essential for induction of apoptosis. Moreover, Casp8L and Casp8-ΔE7, which have only two death-effector domains, significantly activated NFκB by forming filament-like structures, which probably function as scaffolds for the IKK complex formation. In view of increasing levels of these abnormal CASP8 transcripts in primary PBMCs from HTLV-1 carriers and patients with ATL, we propose a possibility that overexpression of those Casp8 mutants, with lower proapoptotic activities and higher NFκB-activating functions than WT-Casp8, may be one of the molecular abnormalities causing malignant transformation and growth of ATL cells. IMPLICATIONS: We describe naturally occurring CASP8 transcription variants in PBMCs from patients with ATL, which encode truncated Casp8-mutant isoforms with lower proapoptotic activities and higher NFκB-activating functions compared with WT-Casp8.


Subject(s)
Alternative Splicing/genetics , Caspase 8/genetics , Deltaretrovirus/genetics , Leukemia, T-Cell/genetics , Apoptosis/genetics , Caspase 8/blood , Cell Line, Tumor , Cell Proliferation/genetics , Exons/genetics , Female , Gene Expression Regulation, Neoplastic , Humans , Leukemia, T-Cell/blood , Leukemia, T-Cell/pathology , Leukemia, T-Cell/virology , Male , RNA Splicing/genetics , Signal Transduction/genetics
5.
J Invest Dermatol ; 139(1): 157-166, 2019 01.
Article in English | MEDLINE | ID: mdl-30048652

ABSTRACT

Human T-cell leukemia virus type 1 (HTLV-1) propagates within and between individuals via cell-to-cell transmission, and primary infection typically occurs across juxtaposed mucosal surfaces during breastfeeding or sexual intercourse. It is therefore likely that dendritic cells (DCs) are among the first potential targets for HTLV-1. However, it remains unclear how DCs contribute to virus transmission and dissemination in the early stages of infection. We show that an HTLV-1-infected cell line (MT-2) and naturally infected CD4+ T cells transfer p19+ viral particles to the surface of allogeneic DCs via cell-to-cell contacts. Similarly organized cell-to-cell contacts also facilitate DC-mediated transfer of HTLV-1 to autologous CD4+ T cells. These findings shed light on the cellular structures involved in anterograde and retrograde transmission and suggest a key role for DCs in the natural history and pathogenesis of HTLV-1 infection.


Subject(s)
CD4-Positive T-Lymphocytes/virology , Dendritic Cells/virology , Human T-lymphotropic virus 1/physiology , Leukemia, T-Cell/pathology , Virus Replication , CD4-Positive T-Lymphocytes/immunology , Dendritic Cells/metabolism , Dendritic Cells/ultrastructure , Humans , Leukemia, T-Cell/metabolism , Leukemia, T-Cell/virology , Microscopy, Electron, Scanning , Tumor Cells, Cultured
6.
Int J Mol Sci ; 19(11)2018 Nov 08.
Article in English | MEDLINE | ID: mdl-30413005

ABSTRACT

Human T-cell leukemia virus type 1 (HTLV-1) is the causative agent of illnesses, such as adult T-cell leukemia/lymphoma, myelopathy/tropical spastic paraparesis (a neurodegenerative disorder), and other diseases. Therefore, HTLV-1 infection is a serious public health concern. Currently, diseases caused by HTLV-1 cannot be prevented or cured. Hence, there is a pressing need to comprehensively understand the mechanisms of HTLV-1 infection and intervention in host cell physiology. HTLV-1-encoded non-structural proteins that reside and function in the cellular membranes are of particular interest, because they alter cellular components, signaling pathways, and transcriptional mechanisms. Summarized herein is the current knowledge about the functions of the membrane-associated p8I, p12I, and p13II regulatory non-structural proteins. p12I resides in endomembranes and interacts with host proteins on the pathways of signal transduction, thus preventing immune responses to the virus. p8I is a proteolytic product of p12I residing in the plasma membrane, where it contributes to T-cell deactivation and participates in cellular conduits, enhancing virus transmission. p13II associates with the inner mitochondrial membrane, where it is proposed to function as a potassium channel. Potassium influx through p13II in the matrix causes membrane depolarization and triggers processes that lead to either T-cell activation or cell death through apoptosis.


Subject(s)
Cell Membrane/genetics , Human T-lymphotropic virus 1/genetics , Leukemia, T-Cell/genetics , Viral Proteins/genetics , Apoptosis/genetics , Cell Membrane/virology , Cell Proliferation/genetics , Gene Expression Regulation, Viral/genetics , Humans , Leukemia, T-Cell/pathology , Leukemia, T-Cell/virology , T-Lymphocytes/virology
7.
Microbiol Immunol ; 62(10): 673-676, 2018 Oct.
Article in English | MEDLINE | ID: mdl-30125970

ABSTRACT

Quantitative PCR (qPCR) of human T-cell leukemia virus type 1 (HTLV-1) provirus is used for HTLV-1 testing and for assessment of risk of HTLV-1-related diseases. In this study, a reference material was developed for standardizing HTLV-1 qPCR. Freeze-dried TL-Om1 cells diluted with Jurkat cells were prepared and an assigned value for proviral load (PVL) of 2.71 copies/100 cells was determined by digital PCR. Nine Japanese laboratories using their own methods evaluated the PVLs of this reference material as 1.08-3.49 copies/100 cells. The maximum difference between laboratories was 3.2-fold. Correcting measured PVLs by using a formula incorporating the assigned value of this reference material should minimize such discrepancies.


Subject(s)
DNA, Viral/analysis , Human T-lymphotropic virus 1/genetics , Leukemia, T-Cell/virology , Real-Time Polymerase Chain Reaction/methods , Real-Time Polymerase Chain Reaction/standards , Cell Line, Tumor , DNA, Viral/genetics , Disaccharides/genetics , HTLV-I Infections/genetics , HTLV-I Infections/virology , Humans , Japan , Jurkat Cells , Proviruses/genetics , Reference Standards , Viral Load/genetics
8.
Sci Rep ; 8(1): 11118, 2018 07 24.
Article in English | MEDLINE | ID: mdl-30042514

ABSTRACT

The human T-cell leukemia virus type 1 (HTLV-1) is highly dependent on cell-to-cell interaction for transmission and productive infection. Cell-to-cell interactions through the virological synapse, biofilm-like structures and cellular conduits have been reported, but the relative contribution of each mechanism on HTLV-1 transmission still remains vastly unknown. The HTLV-1 protein p8 has been found to increase viral transmission and cellular conduits. Here we show that HTLV-1 expressing cells are interconnected by tunneling nanotubes (TNTs) defined as thin structures containing F-actin and lack of tubulin connecting two cells. TNTs connected HTLV-1 expressing cells and uninfected T-cells and monocytes and the viral proteins Tax and Gag localized to these TNTs. The HTLV-1 expressing protein p8 was found to induce TNT formation. Treatment of MT-2 cells with the nucleoside analog cytarabine (cytosine arabinoside, AraC) reduced number of TNTs and furthermore reduced TNT formation induced by the p8 protein. Intercellular transmission of HTLV-1 through TNTs provides a means of escape from recognition by the immune system. Cytarabine could represent a novel anti-HTLV-1 drug interfering with viral transmission.


Subject(s)
Cell Communication/genetics , HTLV-I Infections/genetics , Human T-lymphotropic virus 1/genetics , Nanotubes/chemistry , Tubulin/genetics , Actin Cytoskeleton/drug effects , Actins/genetics , Cell Communication/immunology , Cytarabine/pharmacology , Gene Products, tax/genetics , HTLV-I Infections/transmission , HTLV-I Infections/virology , Human T-lymphotropic virus 1/drug effects , Human T-lymphotropic virus 1/pathogenicity , Humans , Immune System , Jurkat Cells/virology , Leukemia, T-Cell/genetics , Leukemia, T-Cell/pathology , Leukemia, T-Cell/virology , T-Lymphocytes/immunology , Viral Proteins/genetics
9.
Chest ; 154(1): e23-e26, 2018 07.
Article in English | MEDLINE | ID: mdl-30044750

ABSTRACT

CASE PRESENTATION: A 61-year-old Caribbean man presented to the ED with dyspnea that had progressed over the previous week with associated cough and high fevers. Four days prior to admission, his primary care physician noted oral thrush and obtained a chest radiograph that revealed a right middle lobe infiltrate. He was prescribed levofloxacin and clotrimazole. Despite therapy, his symptoms progressed. He had an 11 pack-year smoking history and hypertension but had been in good health. He denied recent travel, alcohol or illicit drug use, or high-risk sexual behaviors, and his only previous medicine was amlodipine. Institutional review board approval was not obtained for this case report, as all patient data are anonymous and obtained during routine patient care activities.


Subject(s)
Antibodies, Viral/analysis , Deltaretrovirus Infections/complications , Leukemia, T-Cell/complications , Primate T-lymphotropic virus 1/immunology , Respiratory Insufficiency/etiology , Tumor Virus Infections/complications , Biopsy , Bronchoscopy , Caribbean Region , Deltaretrovirus Infections/diagnosis , Deltaretrovirus Infections/virology , Diagnosis, Differential , Humans , Leukemia, T-Cell/diagnosis , Leukemia, T-Cell/virology , Male , Middle Aged , Respiratory Insufficiency/diagnosis , Tomography, X-Ray Computed , Tumor Virus Infections/diagnosis , Tumor Virus Infections/virology
10.
Acta Pharmacol Sin ; 38(8): 1093-1103, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28392570

ABSTRACT

Human T-cell lymphotropic virus type 1 (HTLV-1) is the etiologic agent of adult T-cell leukemia/lymphoma (ATL), a rapidly progressing clonal malignancy of CD4+ T lymphocytes. Exploring the host-HTLV-1 interactions and the molecular mechanisms underlying HTLV-1-mediated tumorigenesis is critical for developing efficient therapies against the viral infection and associated leukemia/lymphoma. It has been demonstrated to date that several HTLV-1 proteins play key roles in the cellular transformation and immortalization of infected T lymphocytes. Of note, the HTLV-1 oncoprotein Tax inhibits the innate IFN response through interaction with MAVS, STING and RIP1, causing the suppression of TBK1-mediated phosphorylation of IRF3/IRF7. The HTLV-1 protein HBZ disrupts genomic integrity and inhibits apoptosis and autophagy of the target cells. Furthermore, it is revealed that HBZ enhances the proliferation of ATL cells and facilitates evasion of the infected cells from immunosurveillance. These studies provide insights into the molecular mechanisms by which HTLV-1 mediates the formation of cancer as well as useful strategies for the development of new therapeutic interventions against ATL. In this article, we review the recent advances in the understanding of the pathogenesis, the underlying mechanisms, clinical diagnosis and treatment of the disease caused by HTLV-1 infection. In addition, we discuss the future direction for targeting HTLV-1-associated cancers and strategies against HTLV-1.


Subject(s)
Carcinogenesis/metabolism , Human T-lymphotropic virus 1/physiology , Cell Transformation, Neoplastic/metabolism , HTLV-I Infections/complications , HTLV-I Infections/virology , Human T-lymphotropic virus 1/genetics , Humans , Leukemia, T-Cell/virology , Lymphoma, T-Cell/virology
11.
Transpl Infect Dis ; 19(1)2017 Feb.
Article in English | MEDLINE | ID: mdl-27860061

ABSTRACT

Strongyloides stercoralis has the potential to cause accelerated autoinfection in immunocompromised hosts. Screening tests for strongyloidiasis may be falsely negative in the setting of immunosuppression. We report a case of Strongyloides hyperinfection syndrome in a patient with human T-lymphotropic virus type 1-associated T-cell leukemia early after hematopoietic stem cell transplant. The diagnosis was made by stool ova and parasite examination, despite a negative screening enzyme-linked immunosorbent assay. Because of anticipated prolonged neutropenia, an extended course of treatment was utilized.


Subject(s)
HTLV-I Infections/complications , Hematopoietic Stem Cell Transplantation/adverse effects , Human T-lymphotropic virus 1/isolation & purification , Leukemia, T-Cell/complications , Lymphoma, T-Cell/complications , Strongyloides stercoralis/isolation & purification , Strongyloidiasis/diagnosis , Transplantation Conditioning/adverse effects , Adult , Animals , Antineoplastic Agents/therapeutic use , Antiprotozoal Agents/therapeutic use , Enzyme-Linked Immunosorbent Assay , Fatal Outcome , HTLV-I Infections/therapy , HTLV-I Infections/virology , Hepatitis B, Chronic/complications , Humans , Immunocompromised Host , Leukemia, T-Cell/therapy , Leukemia, T-Cell/virology , Lymphoma, T-Cell/therapy , Lymphoma, T-Cell/virology , Male , Respiratory Distress Syndrome/complications , Respiratory Insufficiency/etiology , Strongyloidiasis/drug therapy , Strongyloidiasis/parasitology , Transplantation Conditioning/methods
12.
Bing Du Xue Bao ; 32(2): 235-42, 2016 Mar.
Article in Chinese | MEDLINE | ID: mdl-27396170

ABSTRACT

Human T-cell leukemia virus type 1 (HTLV-1) is a retrovirus demonstrated to be associated with human disease. Infection by the HTLV-1 can cause T-cell leukemia (ATL) in adults. HTLV-1 bZIP factor (HBZ) is a viral protein encoded by the minus strand of the HTLV-1 provirus. Among the regulatory and accessory genes of HTLV-1, HBZ is the only gene that remains intact and which is expressed consistently in all patients with ATL. Moreover, HBZ has a critical role in the leukemogenesis of ATL. Here, we review the function of HBZ in the oncogenesis of HTLV-1 and its molecular mechanism of action.


Subject(s)
Basic-Leucine Zipper Transcription Factors/metabolism , HTLV-I Infections/virology , Human T-lymphotropic virus 1/metabolism , Leukemia, T-Cell/virology , Retroviridae Proteins/metabolism , Animals , Basic-Leucine Zipper Transcription Factors/genetics , Carcinogenesis , HTLV-I Infections/pathology , Human T-lymphotropic virus 1/genetics , Humans , Leukemia, T-Cell/pathology , Retroviridae Proteins/genetics
13.
J Clin Microbiol ; 53(11): 3485-91, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26292315

ABSTRACT

Quantitative PCR (qPCR) analysis of human T-cell leukemia virus type 1 (HTLV-1) was used to assess the amount of HTLV-1 provirus DNA integrated into the genomic DNA of host blood cells. Accumulating evidence indicates that a high proviral load is one of the risk factors for the development of adult T-cell leukemia/lymphoma and HTLV-1-associated myelopathy/tropical spastic paraparesis. However, interlaboratory variability in qPCR results makes it difficult to assess the differences in reported proviral loads between laboratories. To remedy this situation, we attempted to minimize discrepancies between laboratories through standardization of HTLV-1 qPCR in a collaborative study. TL-Om1 cells that harbor the HTLV-1 provirus were serially diluted with peripheral blood mononuclear cells to prepare a candidate standard. By statistically evaluating the proviral loads of the standard and those determined using in-house qPCR methods at each laboratory, we determined the relative ratios of the measured values in the laboratories to the theoretical values of the TL-Om1 standard. The relative ratios of the laboratories ranged from 0.84 to 4.45. Next, we corrected the proviral loads of the clinical samples from HTLV-1 carriers using the relative ratio. As expected, the overall differences between the laboratories were reduced by half, from 7.4-fold to 3.8-fold on average, after applying the correction. HTLV-1 qPCR can be standardized using TL-Om1 cells as a standard and by determining the relative ratio of the measured to the theoretical standard values in each laboratory.


Subject(s)
DNA, Viral/analysis , Human T-lymphotropic virus 1/genetics , Real-Time Polymerase Chain Reaction/methods , Real-Time Polymerase Chain Reaction/standards , Viral Load/genetics , Cell Line, Tumor , DNA, Viral/genetics , HTLV-I Infections/genetics , HTLV-I Infections/virology , Humans , Japan , Jurkat Cells , Leukemia, T-Cell/genetics , Leukemia, T-Cell/virology , Leukocytes, Mononuclear/virology , Proviruses/genetics , Virus Integration/genetics
14.
Mediators Inflamm ; 2014: 564296, 2014.
Article in English | MEDLINE | ID: mdl-25525301

ABSTRACT

Virus infections are involved in chronic inflammation and, in some cases, cancer development. Although a viral infection activates the immune system's response that eradicates the pathogen mainly through inflammatory mechanisms, it is now recognized that this inflammatory condition is also favorable to the development of tumors. Indeed, it is well described that viruses, such as hepatitis C virus (HCV), Epstein Barr virus (EBV), human papillomavirus (HPV) or human T-cell lymphotropic virus type-1 (HTLV-1), are important risk factors for tumor malignancies. The inflammatory response is a fundamental immune mechanism which involves several molecular and cellular components consisting of cytokines and chemokines that are released by various proinflammatory cells. In parallel to this process, some endogenous recruited components release anti-inflammatory mediators to restore homeostasis. The development of tools and strategies using viruses to hijack the immune response is mostly linked to the presence of regulatory T-cells (Treg) that can inhibit inflammation and antiviral responses of other effector cells. In this review, we will focus on current understanding of the role of natural and induced Treg in the control and the resolution of inflammatory response in HCV-, HTLV-1-, and EBV-associated cancers.


Subject(s)
Hepacivirus/metabolism , Herpesvirus 4, Human/metabolism , Human T-lymphotropic virus 1/metabolism , Inflammation/metabolism , Neoplasms/virology , T-Lymphocytes, Regulatory/cytology , Animals , Carcinoma/virology , Carcinoma, Hepatocellular/virology , Hodgkin Disease/virology , Homeostasis , Humans , Immunity, Innate , Leukemia, T-Cell/virology , Liver Neoplasms/virology , Lymphoma, T-Cell/virology , Nasopharyngeal Carcinoma , Nasopharyngeal Neoplasms/virology , Risk Factors
15.
Int J Oncol ; 45(5): 2159-66, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25189759

ABSTRACT

Experimental and clinical studies have revealed the effectiveness of a specific nutrient synergy (SNS) mixture composed of ascorbic acid (AA), lysine, proline, arginine, epigallocatechin gallate (EGCG) and other micronutrients in targeting crucial physiological mechanisms involved in cancer progression and metastasis. HTLV-1 causes adult T-cell leukemia (ATL). The spread and metastases of ATL as well as other tumors has been associated with matrix metalloproteinases, especially the gelatinases MMP-2 and MMP-9. The objective of this study was to investigate whether SNS, AA and EGCG affects the gelatinolytic activity of MMP-2 and its transcriptional and translational levels in HTLV-1-positive and -negative malignant T-cells. The results indicated that SNS and EGCG caused a dose-dependent decline in the activity, transcription and translation of MMP-2 after treatment with SNS and EGCG, while AA was only able to inhibit the activity at maximum doses tested and to some extent, the protein expression levels of MMP-2, without affecting their transcriptional levels. The highest activity was noted in the case of SNS which is likely to be due to a synergistic effect of the different constituents in the formulation. These results point towards the potential integration of SNS in the anti-invasive treatment of ATL and related diseases.


Subject(s)
Ascorbic Acid/administration & dosage , Catechin/analogs & derivatives , Leukemia, T-Cell/drug therapy , Matrix Metalloproteinase 2/biosynthesis , Catechin/administration & dosage , Drug Synergism , Gene Expression Regulation, Neoplastic/drug effects , HTLV-I Infections/drug therapy , HTLV-I Infections/genetics , HTLV-I Infections/pathology , Human T-lymphotropic virus 1/drug effects , Human T-lymphotropic virus 1/pathogenicity , Humans , Leukemia, T-Cell/genetics , Leukemia, T-Cell/virology , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase 9/biosynthesis , Protein Biosynthesis/drug effects , Transcription, Genetic/drug effects
16.
Life Sci ; 104(1-2): 9-14, 2014 May 28.
Article in English | MEDLINE | ID: mdl-24704970

ABSTRACT

Human T-cell lymphotropic virus type 1 (HTLV-1) is associated with adult T-cell leukemia/lymphoma (ATL) and HTLV-1-associated myelopathy/tropical spastic paraparesis (HAM/TSP). Only a limited percentage of infected individuals develop disease in response to the virus while the majority remain asymptomatic, and HAM/TSP is the most common clinical manifestation of the virus. HAM/TSP is an inflammatory disease of the central nervous system (CNS); however, the mechanism by which HTLV-1 induces HAM/TSP is not yet clear. CD4(+) T lymphocytes are the main reservoirs of HTLV-1 in vivo and perform an important role in the immunological response to this retrovirus. This virus-host interaction may provoke changes in the immunological response, such as the enhanced production of inflammatory cytokines and the spontaneous proliferation of T CD4(+) lymphocytes, which are implicated in the pathogenesis of HAM/TSP.


Subject(s)
Human T-lymphotropic virus 1 , Paraparesis, Tropical Spastic/immunology , Paraparesis, Tropical Spastic/virology , CD4-Positive T-Lymphocytes/cytology , Cell Proliferation , Central Nervous System/metabolism , Cytokines/metabolism , Humans , Immune System , Inflammation , Leukemia, T-Cell/virology
17.
Retrovirology ; 10: 118, 2013 Oct 24.
Article in English | MEDLINE | ID: mdl-24156738

ABSTRACT

BACKGROUND: Human T-cell leukemia virus type 1 (HTLV-1) causes chronic infection leading to development of adult T-cell leukemia (ATL) and inflammatory diseases. Non-human primates infected with simian T-cell leukemia virus type 1 (STLV-1) are considered to constitute a suitable animal model for HTLV-1 research. However, the function of the regulatory and accessory genes of STLV-1 has not been analyzed in detail. In this study, STLV-1 in naturally infected Japanese macaques was analyzed. RESULTS: We identified spliced transcripts of STLV-1 corresponding to HTLV-1 tax and HTLV-1 bZIP factor (HBZ). STLV-1 Tax activated the NFAT, AP-1 and NF-κB signaling pathways, whereas STLV-1 bZIP factor (SBZ) suppressed them. Conversely, SBZ enhanced TGF-ß signaling and induced Foxp3 expression. Furthermore, STLV-1 Tax activated the canonical Wnt pathway while SBZ suppressed it. STLV-1 Tax enhanced the viral promoter activity while SBZ suppressed its activation. Then we addressed the clonal proliferation of STLV-1⁺ cells by massively sequencing the provirus integration sites. Some clones proliferated distinctively in monkeys with higher STLV-1 proviral loads. Notably, one of the monkeys surveyed in this study developed T-cell lymphoma in the brain; STLV-1 provirus was integrated in the lymphoma cell genome. When anti-CCR4 antibody, mogamulizumab, was administered into STLV-1-infected monkeys, the proviral load decreased dramatically within 2 weeks. We observed that some abundant clones recovered after discontinuation of mogamulizumab administration. CONCLUSIONS: STLV-1 Tax and SBZ have functions similar to those of their counterparts in HTLV-1. This study demonstrates that Japanese macaques naturally infected with STLV-1 resemble HTLV-1 carriers and are a suitable model for the investigation of persistent HTLV-1 infection and asymptomatic HTLV-1 carrier state. Using these animals, we verified that mogamulizumab, which is currently used as a drug for relapsed ATL, is also effective in reducing the proviral load in asymptomatic individuals.


Subject(s)
Deltaretrovirus Infections/veterinary , Disease Models, Animal , Leukemia, T-Cell/veterinary , Primate Diseases/pathology , Primate Diseases/virology , Primate T-lymphotropic virus 1/isolation & purification , Tumor Virus Infections/veterinary , Animals , Deltaretrovirus Infections/pathology , Deltaretrovirus Infections/virology , Humans , Leukemia, T-Cell/pathology , Leukemia, T-Cell/virology , Macaca , Primate T-lymphotropic virus 1/growth & development , Primate T-lymphotropic virus 1/pathogenicity , Tumor Virus Infections/pathology , Tumor Virus Infections/virology
18.
J Virol ; 87(13): 7435-44, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23616667

ABSTRACT

Human T-cell leukemia virus type 1 (HTLV-1) is the etiologic agent of adult T-cell leukemia (ATL). The viral regulatory protein Tax1 plays a pivotal role in T-cell transformation and ATL development. Previous studies in our laboratory, using the yeast 2-hybrid approach to screen a T-cell library for Tax1-interacting partners, identified the cellular Four and a Half Lim domain protein 3 (FHL3) as a possible Tax1-interacting candidate. FHL3 is a member of the FHL family of proteins, which function as transcriptional coactivators and cytoskeleton regulators and have a role in cancer progression and development. The aim of this study was to investigate the physical and functional interaction between Tax1 and members of the FHL family of proteins. We show that Tax1 and FHL3 interact both in vitro and in vivo. Furthermore, both FHL1 and -2 also interact with Tax1. We have demonstrated that FHL3 enhances Tax1-mediated activation of the viral long terminal repeat (LTR) without affecting basal activity and that FHL1 to -3 regulate NF-κB activation by Tax1 in a cell-specific manner. In addition, we have found that the interaction between Tax1 and FHL1 to -3 affects the localization of these proteins, leading to their redistribution in cells. Tax1 also affected FHL3 cytoskeleton function by increasing FHL3-mediated cell spreading. Overall, our results suggest that the interaction between Tax1 and the FHL family alters both the transactivating activity and the subcellular localization of Tax1 and provide new insights into molecular mechanisms that underlie the oncogenic nature of this HTLV-1 protein.


Subject(s)
Human T-lymphotropic virus 1/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , LIM Domain Proteins/metabolism , Leukemia, T-Cell/virology , Neoplasm Proteins/metabolism , Animals , Blotting, Western , COS Cells , Chlorocebus aethiops , Chromatin Immunoprecipitation , Cytoskeleton/metabolism , Cytoskeleton/physiology , Fluorescent Antibody Technique , Glutathione Transferase , HeLa Cells , Humans , Immunoprecipitation , Leukemia, T-Cell/metabolism , Plasmids/genetics , Two-Hybrid System Techniques
20.
J Biol Chem ; 287(41): 34683-93, 2012 Oct 05.
Article in English | MEDLINE | ID: mdl-22891252

ABSTRACT

Human T cell leukemia virus type 1 and type 2 (HTLV-1 and -2) are two closely related retroviruses with the former causing adult T cell leukemia. HTLV-2 infection is prevalent among intravenous drug users, and the viral genome encodes the viral transactivator Tax, which is highly homologous to the transforming protein Tax from HTLV-1. However, the link between HTLV-2 infection and leukemia has not been established. In the present study, we evaluated the activity of HTLV-2 Tax in promoting aberrant proliferation of human CD4 T lymphocytes. Tax2 efficiently immortalized CD4(+) memory T lymphocytes with a CD3/TCRαß/CD4/CD25/CD45RO/CD69 immunophenotype, promoted constitutive activation of PI3K/Akt, IκB kinase/NF-κB, mitogen-activated protein kinase, and STAT3, and it also increased the level of Mcl-1. Disruption of these oncogenic pathways led to growth retardation and apoptotic cell death of the Tax2-established T cell lines. We further found that Tax2 induced autophagy by interacting with the autophagy molecule complex containing Beclin1 and PI3K class III to form the LC3(+) autophagosome. Tax2-mediated autophagy promoted survival and proliferation of the immortalized T cells. The present study demonstrated the oncogenic properties of Tax2 in human T cells and also implicated Tax2 in serving as a molecular tool to generate distinct T cell subtype lines.


Subject(s)
Autophagy , CD4-Positive T-Lymphocytes/metabolism , Cell Transformation, Viral , Gene Products, tax/metabolism , Human T-lymphotropic virus 2/metabolism , Immunologic Memory , Antigens, CD/genetics , Antigens, CD/metabolism , CD4-Positive T-Lymphocytes/pathology , Cell Line , Cell Proliferation , Cell Survival/genetics , Enzyme Activation/genetics , Extracellular Signal-Regulated MAP Kinases/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Products, tax/genetics , HTLV-II Infections/genetics , HTLV-II Infections/metabolism , HTLV-II Infections/pathology , Human T-lymphotropic virus 2/genetics , Humans , I-kappa B Kinase/genetics , I-kappa B Kinase/metabolism , Leukemia, T-Cell/genetics , Leukemia, T-Cell/metabolism , Leukemia, T-Cell/pathology , Leukemia, T-Cell/virology , Myeloid Cell Leukemia Sequence 1 Protein , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL