Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 214
Filter
1.
J Neurol ; 271(6): 3471-3485, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38430272

ABSTRACT

Human T-cell leukemia virus type 1 (HTLV-1)-associated myelopathy/tropical spastic paraparesis (HAM/TSP) is a chronic neurodegenerative disease. This multicenter, randomized phase 3 study evaluated the efficacy and safety of 0.3 mg/kg intravenous mogamulizumab, a monoclonal antibody targeting-CC chemokine receptor 4, every 12 weeks in HAM/TSP patients. This study comprised a 24-week double-blind, placebo-controlled period, 24-week open-label period, and extension treatment period. The primary endpoint was the proportion of patients with a ≥ 1-grade improvement in the Osame motor disability score (OMDS). Secondary endpoints were changes in HTLV-1 proviral load, 10-m timed walk, cerebrospinal fluid (CSF) neopterin levels, and safety. The exploratory endpoint was CSF chemokine C-X-C motif ligand 10 (CXCL10) levels. Thirty-four and 33 patients were randomized to mogamulizumab and placebo arms, respectively. At the end of the double-blind period, no significant difference was found in the OMDS improvement rate or other secondary efficacy endpoints assessing motor activities. However, the mogamulizumab arm showed a significant decrease in HTLV-1 proviral load (- 59.39 ± 29.91% vs. placebo 2.32 ± 36.31%) and CSF neopterin (p < 0.001)/CXCL10 levels (p = 0.004). The baseline OMDS pattern and the 60-80% HTLV-1 proviral load reduction were sustained through the open-label and extension treatment periods. Although a higher incidence of rash (69.2%) was reported, the safety profile was similar compared with a previous phase 1/2a study. We found no significant difference in clinical benefit; however, mogamulizumab may provide long-term clinical benefit by preventing disease progression, as CSF neopterin/CXCL10 levels are associated with long-term prognosis in HAM/TSP.Clinical Trial Registration Number: NCT03191526 (registered date: 6-June-2017).


Subject(s)
Antibodies, Monoclonal, Humanized , Human T-lymphotropic virus 1 , Neopterin , Paraparesis, Tropical Spastic , Humans , Double-Blind Method , Antibodies, Monoclonal, Humanized/administration & dosage , Male , Middle Aged , Female , Paraparesis, Tropical Spastic/drug therapy , Paraparesis, Tropical Spastic/cerebrospinal fluid , Adult , Aged , Neopterin/cerebrospinal fluid , Human T-lymphotropic virus 1/drug effects , Chemokine CXCL10/cerebrospinal fluid , Viral Load/drug effects , Treatment Outcome
2.
Nat Commun ; 12(1): 4996, 2021 08 17.
Article in English | MEDLINE | ID: mdl-34404793

ABSTRACT

Between 10 and 20 million people worldwide are infected with the human T-cell lymphotropic virus type 1 (HTLV-1). Despite causing life-threatening pathologies there is no therapeutic regimen for this deltaretrovirus. Here, we screened a library of integrase strand transfer inhibitor (INSTI) candidates built around several chemical scaffolds to determine their effectiveness in limiting HTLV-1 infection. Naphthyridines with substituents in position 6 emerged as the most potent compounds against HTLV-1, with XZ450 having highest efficacy in vitro. Using single-particle cryo-electron microscopy we visualised XZ450 as well as the clinical HIV-1 INSTIs raltegravir and bictegravir bound to the active site of the deltaretroviral intasome. The structures reveal subtle differences in the coordination environment of the Mg2+ ion pair involved in the interaction with the INSTIs. Our results elucidate the binding of INSTIs to the HTLV-1 intasome and support their use for pre-exposure prophylaxis and possibly future treatment of HTLV-1 infection.


Subject(s)
Antiviral Agents/chemistry , Antiviral Agents/pharmacology , Cryoelectron Microscopy , HTLV-I Infections/drug therapy , Human T-lymphotropic virus 1/drug effects , Amides , Catalytic Domain , Deltaretrovirus , Drug Resistance, Viral/drug effects , HIV Integrase/drug effects , HIV-1 , Heterocyclic Compounds, 3-Ring , Human T-lymphotropic virus 1/genetics , Humans , Naphthyridines/pharmacology , Piperazines , Pyridones , Recombinant Proteins
3.
Biochem Biophys Res Commun ; 574: 104-109, 2021 10 15.
Article in English | MEDLINE | ID: mdl-34455369

ABSTRACT

Immunomodulatory imide drugs (IMiDs), such as lenalidomide and pomalidomide, exert pleiotropic effects, e.g., antitumor effects in multiple myeloma, by binding the protein Cereblon and altering its substrate specificity. Lenalidomide is approved for the treatment of adult T-cell leukemia/lymphoma (ATL) caused by human T-cell leukemia virus type 1 (HTLV-1), although the precise mechanisms responsible for its effectiveness have not been fully elucidated. Here, we used HTLV-1-infected cell lines to investigate how IMiDs exert anti-ATL effects. In three of four tested HTLV-1-infected cell lines, the cells treated with lenalidomide or pomalidomide exhibited mild growth suppression without apoptosis, which was associated with decreased IRF4, c-Myc, and phosphorylated STAT3 levels as well as enhanced SOCS3 expression. Additionally, the levels of enhancer of zeste homolog 2 (EZH2) and trimethyl histone 3 Lys27 (H3K27me3) were decreased following IMiD treatment in all three susceptible cell lines. An IMiD-mediated reduction of EZH2 and H3K27me3 levels was also observed in a multiple myeloma cell line. Furthermore, treatment with an EZH2-inhibitor reproduced the IMiD-mediated effects in HTLV-1-infected cells and multiple myeloma cells. These findings strongly suggest that a reduction of EZH2 expression is involved in the mechanism underlying the antitumor effects of IMiD.


Subject(s)
Antiviral Agents/pharmacology , Enhancer of Zeste Homolog 2 Protein/antagonists & inhibitors , HTLV-I Infections/drug therapy , Human T-lymphotropic virus 1/drug effects , Lenalidomide/pharmacology , Thalidomide/analogs & derivatives , Cell Line , Cell Proliferation/drug effects , Enhancer of Zeste Homolog 2 Protein/metabolism , HTLV-I Infections/pathology , Humans , Microbial Sensitivity Tests , Thalidomide/pharmacology
4.
Int J Mol Sci ; 22(11)2021 May 24.
Article in English | MEDLINE | ID: mdl-34073995

ABSTRACT

Human T-cell leukemia virus type 1 (HTLV-1), the cause of adult T-cell leukemia/lymphoma (ATLL), is a retrovirus, which integrates into the host genome and persistently infects CD4+ T-cells. Virus propagation is stimulated by (1) clonal expansion of infected cells and (2) de novo infection. Viral gene expression is induced by the transactivator protein Tax, which recruits host factors like positive transcription elongation factor b (P-TEFb) to the viral promoter. Since HTLV-1 gene expression is repressed in vivo by viral, cellular, and epigenetic mechanisms in late phases of infection, HTLV-1 avoids an efficient CD8+ cytotoxic T-cell (CTL) response directed against the immunodominant viral Tax antigen. Hence, therapeutic strategies using latency reversing agents (LRAs) sought to transiently activate viral gene expression and antigen presentation of Tax to enhance CTL responses towards HTLV-1, and thus, to expose the latent HTLV-1 reservoir to immune destruction. Here, we review strategies that aimed at enhancing Tax expression and Tax-specific CTL responses to interfere with HTLV-1 latency. Further, we provide an overview of LRAs including (1) histone deacetylase inhibitors (HDACi) and (2) activators of P-TEFb, that have mainly been studied in context of human immunodeficiency virus (HIV), but which may also be powerful in the context of HTLV-1.


Subject(s)
Gene Expression Regulation, Viral/drug effects , Gene Products, tax/immunology , Gene Products, tax/metabolism , Histone Deacetylase Inhibitors/pharmacology , Human T-lymphotropic virus 1/drug effects , Leukemia-Lymphoma, Adult T-Cell/immunology , Positive Transcriptional Elongation Factor B/metabolism , Virus Latency/drug effects , CD4-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/virology , Gene Expression Regulation, Viral/genetics , Gene Products, tax/genetics , Histone Deacetylase Inhibitors/therapeutic use , Histones/metabolism , Human T-lymphotropic virus 1/genetics , Human T-lymphotropic virus 1/immunology , Human T-lymphotropic virus 1/metabolism , Humans , Leukemia-Lymphoma, Adult T-Cell/metabolism , Leukemia-Lymphoma, Adult T-Cell/pathology , Leukemia-Lymphoma, Adult T-Cell/virology , Phosphorylation , Virus Latency/genetics
5.
Front Immunol ; 12: 608890, 2021.
Article in English | MEDLINE | ID: mdl-33767694

ABSTRACT

Human T-cell lymphotropic virus type 1 (HTLV-1)-associated myelopathy/tropical spastic paraparesis (HAM/TSP) develops in 1-5% of HTLV-1-infected individuals. Previous studies by us and others have shown that the expression of negative immune checkpoint receptors (NCRs) is significantly increased on CD8 T cells in various chronic viral infections and are associated with poor anti-viral immunity. We have previously identified the differential expression of NCRs on CD8 T cells in blood from patients with HAM/TSP and in central nervous system (CNS) tissues of HTLV-1 infected humanized mice and defined the association with neurological complications. In this study, we determined the co-expression patterns of several key NCRs (PD-1, TIGIT, TIM-3, and LAG-3) and their cognate ligands in HTLV-1 infection and assessed how combination strategies targeting these pathways would impact HTLV-1-specific CD8 T-cell effector functions as an approach to reduce CNS disease outcomes. We found that global CD8 T cells from HAM/TSP patients co-express multiple NCRs at significantly higher frequencies than asymptomatic carriers (AC). Moreover, NCR ligands (PVR and PD-LI) on both plasmacytoid and myeloid dendritic cells were also expressed at higher frequencies in HAM/TSP compared to AC. In both AC and HAM/TSP subjects, combination dual PD-L1/TIGIT or triple PD-L1/TIGIT/TIM-3 blockade with monoclonal antibodies resulted in increases in intracellular cytokine expression in CD8 T cells after virus stimulation, particularly CD107a, a marker of degranulation, and TNF-α, a key cytokine that can directly inhibit viral replication. Interestingly, almost all blockade combinations resulted in reduced IL-2+ HTLV-1-specific CD8 T cell frequencies in HAM/TSP subjects, but not in AC. These results define a novel combinatorial NCR immunotherapeutic blockade strategy to reduce HAM/TSP disease burden.


Subject(s)
Anti-Retroviral Agents/pharmacology , HTLV-I Infections/genetics , HTLV-I Infections/immunology , Human T-lymphotropic virus 1/drug effects , Human T-lymphotropic virus 1/immunology , Immune Checkpoint Inhibitors/pharmacology , Adult , Anti-Retroviral Agents/therapeutic use , Biomarkers , Clinical Decision-Making , Cytokines , Disease Management , Drug Therapy, Combination , Female , Gene Expression Regulation/drug effects , HTLV-I Infections/drug therapy , HTLV-I Infections/virology , Host-Pathogen Interactions/immunology , Humans , Immune Checkpoint Inhibitors/therapeutic use , Immune Checkpoint Proteins/genetics , Immune Checkpoint Proteins/metabolism , Immunologic Memory , Immunophenotyping , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/metabolism , Lymphocyte Count , Male , Middle Aged , T-Lymphocyte Subsets/drug effects , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Treatment Outcome , Viral Load
6.
ACS Chem Biol ; 16(3): 529-538, 2021 03 19.
Article in English | MEDLINE | ID: mdl-33619959

ABSTRACT

Human T-cell lymphotropic virus type 1 (HTLV-1) is a retrovirus that can cause severe paralytic neurologic disease and immune disorders as well as cancer. An estimated 20 million people worldwide are infected with HTLV-1, with prevalence reaching 30% in some parts of the world. In stark contrast to HIV-1, no direct acting antivirals (DAAs) exist against HTLV-1. The aspartyl protease of HTLV-1 is a dimer similar to that of HIV-1 and processes the viral polyprotein to permit viral maturation. We report that the FDA-approved HIV-1 protease inhibitor darunavir (DRV) inhibits the enzyme with 0.8 µM potency and provides a scaffold for drug design against HTLV-1. Analogs of DRV that we designed and synthesized achieved submicromolar inhibition against HTLV-1 protease and inhibited Gag processing in viral maturation assays and in a chronically HTLV-1 infected cell line. Cocrystal structures of these inhibitors with HTLV-1 protease highlight opportunities for future inhibitor design. Our results show promise toward developing highly potent HTLV-1 protease inhibitors as therapeutic agents against HTLV-1 infections.


Subject(s)
Antiviral Agents/chemistry , Aspartic Acid Endopeptidases/antagonists & inhibitors , Darunavir/analogs & derivatives , Human T-lymphotropic virus 1/drug effects , Protease Inhibitors/chemistry , Amino Acid Sequence , Antiviral Agents/pharmacology , Aspartic Acid Endopeptidases/chemistry , Aspartic Acid Endopeptidases/genetics , Darunavir/pharmacology , Drug Discovery , Escherichia coli/genetics , Humans , Molecular Dynamics Simulation , Molecular Structure , Molecular Targeted Therapy , Protease Inhibitors/pharmacology , Protein Binding , Protein Conformation , Structure-Activity Relationship
7.
Bioorg Med Chem ; 28(22): 115746, 2020 11 15.
Article in English | MEDLINE | ID: mdl-33007558

ABSTRACT

Human T cell lymphotropic virus type 1 (HTLV-1) is a human retrovirus that infects approximately 10-20 million people worldwide and causes an aggressive neoplasia (adult T-cell leukemia/lymphoma - ATL). Therapeutic approaches for the treatment of ATL have variable effectiveness and poor prognosis, thus requiring strategies to identify novel compounds with activity on infected cells. In this sense, we initially screened a small series of 25 1,2,3-triazole derivatives to discover cell proliferation inhibitors and apoptosis inducers in HTLV-1-infected T-cell line (MT-2) for further assessment of their effect on viral tax activity through inducible-tax reporter cell line (Jurkat LTR-GFP). Eight promising compounds (02, 05, 06, 13, 15, 21, 22 and 25) with activity ≥70% were initially selected, based on a suitable cell-based assay using resazurin reduction method, and evaluated towards cell cycle, apoptosis and Tax/GFP expression analyses through flow cytometry. Compound 02 induced S phase cell cycle arrest and compounds 05, 06, 22 and 25 promoted apoptosis. Remarkably, compounds 22 and 25 also reduced GFP expression in an inducible-tax reporter cell, which suggests an effect on Tax viral protein. More importantly, compounds 02, 22 and 25 were not cytotoxic in human hepatoma cell line (Huh-7). Therefore, the discovery of 3 active and non-cytotoxic compounds against HTLV-1-infected cells can potentially contribute, as an initial promising strategy, to the development process of new drugs against ATL.


Subject(s)
Antiviral Agents/pharmacology , Gene Products, tax/antagonists & inhibitors , Heterocyclic Compounds/pharmacology , Human T-lymphotropic virus 1/drug effects , Triazoles/pharmacology , Antiviral Agents/chemical synthesis , Antiviral Agents/chemistry , Cell Cycle Checkpoints/drug effects , Cell Survival/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Gene Products, tax/metabolism , Heterocyclic Compounds/chemistry , Humans , Molecular Structure , Structure-Activity Relationship , Triazoles/chemistry
8.
Biosci Rep ; 40(10)2020 10 30.
Article in English | MEDLINE | ID: mdl-33048123

ABSTRACT

Ceramide (Cer) is a bioactive cellular lipid with compartmentalized and tightly regulated levels. Distinct metabolic pathways lead to the generation of Cer species with distinguishable roles in oncogenesis. Deregulation of Cer pathways has emerged as an important mechanism for acquired chemotherapeutic resistance. Adult T-cell leukemia (ATL) cells are defective in Cer synthesis. ATL is an aggressive neoplasm that develops following infection with human T-cell lymphotropic virus-1 (HTLV-1) where the viral oncogene Tax contributes to the pathogenesis of the disease. ATL cells, resistant to all-trans-retinoic acid, are sensitive to pharmacologically achievable concentrations of the synthetic retinoid ST1926. We studied the effects of ST1926 on Cer pathways in ATL cells. ST1926 treatment resulted in early Tax oncoprotein degradation in HTLV-1-treated cells. ST1926 induced cell death and a dose- and time-dependent accumulation of Cer in malignant T cells. The kinetics and degree of Cer production showed an early response upon ST1926 treatment. ST1926 enhanced de novo Cer synthesis via activation of ceramide synthase CerS(s) without inhibiting dihydroceramide desaturase, thereby accumulating Cer rather than the less bioactive dihydroceramide. Using labeling experiments with the unnatural 17-carbon sphinganine and measuring the generated Cer species, we showed that ST1926 preferentially induces the activities of a distinct set of CerS(s). We detected a delay in cell death response and interruption of Cer generation in response to ST1926 in Molt-4 cells overexpressing Bcl-2. These results highlight the potential role of ST1926 in inducing Cer levels, thus lowering the threshold for cell death in ATL cells.


Subject(s)
Adamantane/analogs & derivatives , Antineoplastic Agents/pharmacology , Ceramides/biosynthesis , Cinnamates/pharmacology , Leukemia-Lymphoma, Adult T-Cell/drug therapy , Adamantane/pharmacology , Cell Death/drug effects , Enzyme Activation , Gene Products, tax/metabolism , Human T-lymphotropic virus 1/drug effects , Human T-lymphotropic virus 1/metabolism , Humans , Jurkat Cells , Leukemia-Lymphoma, Adult T-Cell/metabolism , Leukemia-Lymphoma, Adult T-Cell/pathology , Leukemia-Lymphoma, Adult T-Cell/virology , Oxidoreductases/genetics , Oxidoreductases/metabolism , Proteolysis , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Time Factors
9.
J Neurovirol ; 26(5): 652-663, 2020 10.
Article in English | MEDLINE | ID: mdl-32705480

ABSTRACT

Human T-lymphotropic virus type 1 (HTLV-1) is associated with adult T cell leukemia/lymphoma and HTLV-1-associated myelopathy/tropical spastic paraparesis (HAM/TSP). HAM/TSP is an inflammatory disease of the spinal cord and clinically characterized by progressive spastic paraparesis, urinary incontinence, and mild sensory disturbance. The interaction between the host immune response and HTLV-1-infected cells regulates the development of HAM/TSP. HTLV-1 preferentially infects CD4+ T cells and is maintained by proliferation of the infected T cells. HTLV-1-infected cells rarely express viral antigens in vivo; however, they easily express the antigens after short-term culture. Therefore, such virus-expressing cells may lead to activation and expansion of antigen-specific T cell responses. Infected T cells with HTLV-1 and HTLV-1-specific CD8+ cytotoxic T lymphocytes invade the central nervous system and produce various proinflammatory cytokines and chemokines, leading to neuronal damage and degeneration. Therefore, cellular immune responses to HTLV-1 have been considered to play important roles in disease development of HAM/TSP. Recent studies have clarified the viral strategy for persistence in the host through genetic and epigenetic changes by HTLV-1 and host immune responses including T cell function and differentiation. Newly developed animal models could provide the opportunity to uncover the precise pathogenesis and development of clinically effective treatment. Several molecular target drugs are undergoing clinical trials with promising efficacy. In this review, we summarize recent advances in the immunopathogenesis of HAM/TSP and discuss the perspectives of the research on this disease.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Cytokines/immunology , Human T-lymphotropic virus 1/pathogenicity , Leukemia-Lymphoma, Adult T-Cell/immunology , Paraparesis, Tropical Spastic/immunology , Urinary Incontinence/immunology , Animals , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/virology , Cell Proliferation/drug effects , Cytokines/biosynthesis , Disease Models, Animal , Host-Pathogen Interactions/immunology , Human T-lymphotropic virus 1/drug effects , Human T-lymphotropic virus 1/immunology , Humans , Immunity, Cellular/drug effects , Immunologic Factors/therapeutic use , Leukemia-Lymphoma, Adult T-Cell/drug therapy , Leukemia-Lymphoma, Adult T-Cell/pathology , Leukemia-Lymphoma, Adult T-Cell/virology , Lymphocyte Activation/drug effects , Neurons/drug effects , Neurons/immunology , Neurons/pathology , Neurons/virology , Neuroprotective Agents/therapeutic use , Paraparesis, Tropical Spastic/drug therapy , Paraparesis, Tropical Spastic/pathology , Paraparesis, Tropical Spastic/virology , Spinal Cord/drug effects , Spinal Cord/immunology , Spinal Cord/virology , T-Lymphocytes, Cytotoxic/drug effects , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/virology , Urinary Incontinence/drug therapy , Urinary Incontinence/pathology , Urinary Incontinence/virology
10.
Blood ; 136(7): 871-884, 2020 08 13.
Article in English | MEDLINE | ID: mdl-32391874

ABSTRACT

Adult T-cell leukemia-lymphoma (ATL) is an aggressive hematological malignancy of CD4+ T cells transformed by human T-cell lymphotropic virus-1 (HTLV-1). Most HTLV-1-infected individuals are asymptomatic, and only 3% to 5% of carriers develop ATL. Here, we describe the contribution of aberrant DNA methylation to ATL leukemogenesis. HTLV-1-infected T-cells and their uninfected counterparts were separately isolated based on CADM1 and CD7 expression status, and differentially methylated positions (DMPs) specific to HTLV-infected T cells were identified through genome-wide DNA methylation profiling. Accumulation of DNA methylation at hypermethylated DMPs correlated strongly with ATL development and progression. In addition, we identified 22 genes downregulated because of promoter hypermethylation in HTLV-1-infected T cells, including THEMIS, LAIR1, and RNF130, which negatively regulate T-cell receptor (TCR) signaling. Phosphorylation of ZAP-70, a transducer of TCR signaling, was dysregulated in HTLV-1-infected cell lines but was normalized by reexpression of THEMIS. Therefore, we hypothesized that DNA hypermethylation contributes to growth advantages in HTLV-1-infected cells during ATL leukemogenesis. To test this idea, we investigated the anti-ATL activities of OR-1200 and OR-2100 (OR21), novel decitabine (DAC) prodrugs with enhanced oral bioavailability. Both DAC and OR21 inhibited cell growth, accompanied by global DNA hypomethylation, in xenograft tumors established by implantation of HTLV-1-infected cells. OR21 was less hematotoxic than DAC, whereas tumor growth inhibition was almost identical between the 2 compounds, making it suitable for long-term treatment of ATL patient-derived xenograft mice. Our results demonstrate that regional DNA hypermethylation is functionally important for ATL leukemogenesis and an effective therapeutic target.


Subject(s)
Antineoplastic Agents/administration & dosage , DNA Methylation/drug effects , HTLV-I Infections/drug therapy , Leukemia-Lymphoma, Adult T-Cell/drug therapy , Pyridines/administration & dosage , Administration, Oral , Adult , Aged , Animals , Cell Transformation, Viral/drug effects , Cell Transformation, Viral/genetics , Cells, Cultured , DNA Methylation/genetics , Demethylation/drug effects , Drugs, Investigational/therapeutic use , Female , Gene Expression Regulation, Leukemic/drug effects , HTLV-I Infections/complications , HTLV-I Infections/genetics , Human T-lymphotropic virus 1/drug effects , Human T-lymphotropic virus 1/physiology , Humans , Leukemia-Lymphoma, Adult T-Cell/genetics , Mice , Mice, Inbred BALB C , Mice, Knockout , Molecular Targeted Therapy/methods , Xenograft Model Antitumor Assays , Young Adult
11.
Microb Pathog ; 144: 104198, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32283259

ABSTRACT

HTLV-1, the first human oncogenic retrovirus, is a type C retrovirus that belongs to the Deltaretrovirus genus. The HTLV-1 genome has 8.5 kbp length, and consists of major genes such as gag, pol, pro, env, and pX region. This retrovirus is considered as one of the most deadly infectious agent for peripheral-blood mononuclear cells (PBMC). The infection of HTLV-1 can lead to dangerous complications, such as infective dermatitis (ID), uveitis, arthritis, lymphadenitis, arthropathies, Sjögren's Syndrome (SS), and particularly HTLV-1-associated myelopathy/tropical spastic paraparesis (HAM/TSP) or Adult T-Cell Leukemia Lymphoma (ATLL). At the moment, Zidovudine (AZT) plus IFN-α is the only treatment available for HTLV-1 infections. Based on scientific studies, alongside the therapeutic regimens, intrinsic mechanisms also play a determinant role in reducing the signs of disease. Programmed cell death-1 (PD-1) signaling pathway, one of the most important checkpoints, has recently received interest, such as the development of a novel generation of anti-tumors. In the present study, we discuss the role of PD-1 signaling pathway in HTLV-1 infection as well as its application as a novel approach for treatment of HTLV-1 infections.


Subject(s)
HTLV-I Infections/drug therapy , Human T-lymphotropic virus 1/drug effects , Human T-lymphotropic virus 1/genetics , Molecular Targeted Therapy/methods , Programmed Cell Death 1 Receptor/drug effects , Signal Transduction/drug effects , Adult , Antigens, CD/chemistry , CTLA-4 Antigen/chemistry , Chronic Disease , HTLV-I Infections/complications , HTLV-I Infections/virology , Hepatitis A Virus Cellular Receptor 2/chemistry , Humans , Interferon-alpha/pharmacology , Leukocytes, Mononuclear/virology , Paraparesis, Tropical Spastic , Programmed Cell Death 1 Receptor/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 11/chemistry , Receptors, Immunologic/chemistry , Zidovudine/pharmacology , Lymphocyte Activation Gene 3 Protein
12.
Retrovirology ; 17(1): 5, 2020 03 21.
Article in English | MEDLINE | ID: mdl-32199462

ABSTRACT

BACKGROUND: Adult T-cell leukemia-lymphoma (ATL) is an aggressive mature lymphoid proliferation associated with poor prognosis. Standard of care includes chemotherapy and/or the combination of zidovudine and interferon-alpha. However, most patients experience relapse less than 6 months after diagnosis. Allogeneic stem cell transplantation is the only curative treatment, but is only feasible in a minority of cases. We previously showed in a mouse model that Arsenic trioxide (As2O3) targets ATL leukemia initiating cells. RESULTS: As2O3 consolidation was given in 9 patients with ATL (lymphoma n = 4; acute n = 2; and indolent n = 3), who were in complete (n = 4) and partial (n = 3) remission, in stable (n = 1) and in progressive (n = 1) disease. Patients received up to 8 weeks of As2O3 at the dose of 0.15 mg/kg/day intravenously in combination with zidovudine and interferon-alpha. One patient in progression died rapidly. Of the remaining eight patients, three with indolent ATL subtype showed overall survivals of 48, 53 and 97 months, and duration of response to As2O3 of 22, 25 and 73 months. The other 5 patients with aggressive ATL subtype had median OS of 36 months and a median duration of response of 10 months. Side effects were mostly hematological and cutaneous (one grade 3) and reversible with dose reduction of AZT/IFN and/or As2O3 discontinuation. The virus integration analysis revealed the regression of the predominant malignant clone in one patient with a chronic subtype. CONCLUSION: These results suggest that consolidation with As2O3 could be an option for patients with ATL in response after induction therapy and who are not eligible for allogeneic stem cell transplantation.


Subject(s)
Arsenic Trioxide/therapeutic use , Leukemia-Lymphoma, Adult T-Cell/drug therapy , Administration, Intravenous , Adult , Arsenic Trioxide/administration & dosage , Drug Therapy, Combination , Female , Human T-lymphotropic virus 1/drug effects , Humans , Interferon-alpha/therapeutic use , Longitudinal Studies , Male , Middle Aged , Retrospective Studies , Zidovudine/therapeutic use
14.
J Virol ; 94(1)2019 12 12.
Article in English | MEDLINE | ID: mdl-31597781

ABSTRACT

The divergent clinical outcomes of human T cell leukemia virus type 1 (HTLV-1) and HTLV-2 infections have been attributed to functional differences in their antisense proteins. In contrast to HTLV-1 bZIP factor (HBZ), the role of the antisense protein of HTLV-2 (APH-2) in HTLV-2 infection is poorly understood. In previous studies, we identified the endosomal sorting complex required for transport 0 (ESCRT-0) subunit HRS as a novel interaction partner of APH-2 but not HBZ. HRS is a master regulator of endosomal protein sorting for lysosomal degradation and is hijacked by many viruses to promote replication. However, no studies to date have shown a link between HTLVs and HRS. In this study, we sought to characterize the interaction between HRS and APH-2 and to investigate the impact of HRS on the life cycle of HTLV-2. We confirmed a direct specific interaction between APH-2 and HRS and showed that the CC2 domain of HRS and the N-terminal domain of APH-2 mediate their interaction. We demonstrated that HRS recruits APH-2 to early endosomes, possibly furnishing an entry route into the endosomal/lysosomal pathway. We demonstrated that inhibition of this pathway using either bafilomycin or HRS overexpression substantially extends the half-life of APH-2 and stabilizes Tax2B expression levels. We found that HRS enhances Tax2B-mediated long terminal repeat (LTR) activation, while depletion of HRS enhances HTLV-2 production and release, indicating that HRS may have a negative impact on HTLV-2 replication. Overall, our study provides important new insights into the role of the ESCRT-0 HRS protein, and by extension the ESCRT machinery and the endosomal/lysosomal pathway, in HTLV-2 infection.IMPORTANCE While APH-2 is the only viral protein consistently expressed in infected carriers, its role in HTLV-2 infection is poorly understood. In this study, we characterized the interaction between the ESCRT-0 component HRS and APH-2 and explored the role of HRS in HTLV-2 replication. HRS is a master regulator of protein sorting for lysosomal degradation, a feature that is manipulated by several viruses to promote replication. Unexpectedly, we found that HRS targets APH-2 and possibly Tax2B for lysosomal degradation and has an overall negative impact on HTLV-2 replication and release. The negative impact of interactions between HTLV-2 regulatory proteins and HRS, and by extension the ESCRT machinery, may represent an important strategy used by HTLV-2 to limit virus production and to promote persistence, features that may contribute to the limited pathogenic potential of this infection.


Subject(s)
Endosomal Sorting Complexes Required for Transport/genetics , Gene Products, tax/genetics , Human T-lymphotropic virus 2/genetics , Phosphoproteins/genetics , Retroviridae Proteins/genetics , Basic-Leucine Zipper Transcription Factors/genetics , Basic-Leucine Zipper Transcription Factors/metabolism , Cycloheximide/pharmacology , Endosomal Sorting Complexes Required for Transport/antagonists & inhibitors , Endosomal Sorting Complexes Required for Transport/metabolism , Gene Expression Regulation , Gene Products, tax/metabolism , HEK293 Cells , HeLa Cells , Human T-lymphotropic virus 1/drug effects , Human T-lymphotropic virus 1/genetics , Human T-lymphotropic virus 1/metabolism , Human T-lymphotropic virus 1/pathogenicity , Human T-lymphotropic virus 2/drug effects , Human T-lymphotropic virus 2/metabolism , Human T-lymphotropic virus 2/pathogenicity , Humans , Lysosomes/drug effects , Lysosomes/metabolism , Lysosomes/virology , Macrolides/pharmacology , Phosphoproteins/metabolism , Retroviridae Proteins/metabolism , Signal Transduction , Virus Replication/drug effects
15.
Virus Res ; 270: 197653, 2019 09.
Article in English | MEDLINE | ID: mdl-31299194

ABSTRACT

Adult T-cell leukemia and human T-cell leukemia virus type 1 (HTLV-1) - associated myelopathy/tropical spastic paraparesis, which develop after HTLV-1 infection, are difficult to cure. In particular, the mode of HTLV-1 propagation is not well understood. Poly (ADP-ribose) polymerase-1 is reported to be a co-activator of HTLV-1 Tax protein; however, the effects of polyADP-ribosylation on infectivity of HTLV-1 have not been fully clarified. We studied the effects of a PARP inhibitor on two modes of HTLV-1 transmission: through cell adhesion between MT-2 cells (an HTLV-1-infected cell line) and uninfected cells and through virus particles produced by HTLV-1-producing c77 cells. Although the PARP inhibitor decreased HTLV-1 infection through cell adhesion, it increased HTLV-1 infection through virion production and caused apoptosis of HTLV-1-infected cells. Thus, careful consideration is required for clinical application of PARP inhibitors in HTLV-1 patients.


Subject(s)
Apoptosis/drug effects , Human T-lymphotropic virus 1/drug effects , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Virion/drug effects , Virus Attachment/drug effects , Cell Line , Cell Proliferation/drug effects , Cell Survival/drug effects , Human T-lymphotropic virus 1/physiology , Humans , T-Lymphocytes/drug effects , T-Lymphocytes/virology
16.
J Virol ; 93(16)2019 08 15.
Article in English | MEDLINE | ID: mdl-31167921

ABSTRACT

Human T-cell leukemia virus type 1 (HTLV-1) infection causes T-cell leukemia and inflammatory diseases, most notably including HTLV-1-associated myelopathy/tropical spastic paraparesis (HAM/TSP). The underlying mechanism for the pathogenesis of HAM/TSP remains unclear. According to a recent clinical trial, a humanized antibody that targets CCR4+ cells ameliorates inflammation by reducing the number of infected cells in the central nervous system; this result suggests that the transmigration of HTLV-1-infected cells plays a crucial role in HAM/TSP. Partly due to the blood-brain barrier, current treatments for HAM/TSP are mostly palliative. Pentosan polysulfate (PPS), a semisynthetic glycosaminoglycan, has recently been used to treat HAM/TSP and was found to alleviate the symptoms. In this study, we investigated the effect of PPS on HTLV-1-infected cells and provide evidence for its efficacy in HAM/TSP. PPS was cytotoxic to certain HTLV-1-infected cells and significantly suppressed HTLV-1 virion production. PPS also efficiently inhibited HTLV-1 cell-cell transmission in T cells. In addition, PPS blocked HTLV-1 infection of primary endothelial cells (human umbilical vascular endothelial cells) and suppressed the subsequent induction of proinflammatory cytokine expression. Furthermore, PPS was found to inhibit the adhesion and transmigration of HTLV-1-infected cells. We also confirmed the anti-HTLV-1 effect of PPS in vivo using two mouse models. PPS blocked HTLV-1 infection in a mouse model with peripheral blood mononuclear cell (PBMC)-humanized NOD-scid IL2Rgammanull (huPBMC NSG) mice. PPS was also found to suppress the development of dermatitis and lung damage in HTLV-1 bZIP factor (HBZ)-transgenic (HBZ-Tg) mice, an HTLV-1 transgenic mouse model in which the mice develop systemic inflammation.IMPORTANCE HTLV-1 is the first human retrovirus to have been identified and is endemic in certain areas worldwide. HTLV-1 infection leads to the development of an inflammatory disease called HAM/TSP, a myelopathy characterized by slowly progressive spastic paraparesis. There have been no effective therapeutics available for HAM/TSP, but recently, a semisynthetic glycosaminoglycan, named pentosan polysulfate (PPS), has been found to alleviate the symptoms of HAM/TSP. Here we conducted a comprehensive study on the effect of PPS both in vitro and in vivo PPS demonstrated anti-HTLV-1 potential in infected cell lines, as shown by its suppressive effects on HTLV-1 replication and transmission and on the transmigration of infected T cells. Moreover, results obtained from two HTLV-1 mouse models demonstrate that PPS inhibits HTLV-1 infection and inflammation development in vivo Our work offers insights into the treatment of HAM/TSP by PPS and also suggests its possible use for treating other HTLV-1-induced inflammatory diseases.


Subject(s)
Antineoplastic Agents/pharmacology , HTLV-I Infections/virology , Human T-lymphotropic virus 1/drug effects , Human T-lymphotropic virus 1/physiology , Pentosan Sulfuric Polyester/pharmacology , Animals , Cell Adhesion , Cell Line, Tumor , Disease Models, Animal , Endothelial Cells/virology , HTLV-I Infections/drug therapy , HTLV-I Infections/transmission , Humans , Leukocytes, Mononuclear/metabolism , Leukocytes, Mononuclear/virology , Mice , Mice, Inbred NOD , Mice, Knockout , Mice, Transgenic , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , T-Lymphocytes/virology , Virus Replication/drug effects
17.
Retrovirology ; 16(1): 5, 2019 02 19.
Article in English | MEDLINE | ID: mdl-30782173

ABSTRACT

BACKGROUND: Adult T-cell leukemia (ATL) is an aggressive neoplasm caused by human T-cell leukemia virus type 1 (HTLV-1). ATL carries a poor prognosis due to chemotherapy resistance. Thus, it is urgent to develop new treatment strategies. Hypericin (HY) is a new-type of photosensitizer in the context of photodynamic therapy (PDT) due to its excellent photosensitizing properties and anti-tumor activities. RESULTS: In the present study, we investigated the efficacy of hypericin in ATL cells. Clinically achievable concentrations of hypericin in association with PDT induced the inhibition of cell proliferation in ATL cell lines with minimal effect on peripheral blood CD4+ T lymphocytes. Moreover, hypericin-PDT treatment caused apoptosis and G2/M phase cell cycle arrest in leukemic cells. Western blot analyses revealed that hypericin-PDT treatment resulted in downregulation of Bcl-2 and enhanced the expression of Bad, cytochrome C, and AIF. Cleavage of caspases-3/-7/-9/-8, Bid, and PARP was increased in hypericin-PDT-treated ATL cells. In a luciferase assay, hypericin-PDT treatment was able to activate the promoter activity of Bax and p53, resulting in enhanced expression of Bax and p53 proteins. Finally, hypericin-PDT treatment suppressed the expression of viral protein HBZ and Tax by blocking the promoter activity via HTLV-1 5'LTR and 3'LTR. CONCLUSIONS: Our results revealed that hypericin-PDT is highly effective against ATL cells by induction of apoptosis and suppression of viral transcription. These studies highlight the promising use of hypericin-PDT as a targeted therapy for ATL.


Subject(s)
Apoptosis/drug effects , Human T-lymphotropic virus 1/drug effects , Leukemia-Lymphoma, Adult T-Cell/drug therapy , Light , Perylene/analogs & derivatives , Photochemotherapy/methods , Photosensitizing Agents/pharmacology , Anthracenes , Apoptosis/radiation effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/radiation effects , Cell Survival/drug effects , Cell Survival/radiation effects , Human T-lymphotropic virus 1/radiation effects , Humans , Models, Biological , Perylene/pharmacology , Transcription, Genetic/drug effects , Transcription, Genetic/radiation effects , Tumor Stem Cell Assay
18.
Int J Biol Macromol ; 124: 282-290, 2019 Mar 01.
Article in English | MEDLINE | ID: mdl-30452989

ABSTRACT

We examined the effects of various acidic polysaccharides isolated from marine algae on the infection and replication of human immunodeficiency virus type-1 (HIV-1), hepatitis B virus (HBV), hepatitis C virus (HCV), and human T-cell leukemia virus type-1 (HTLV-1). It was found that sulfated fucan polysaccharides, ascophyllan, and two fucoidans derived from different sources significantly inhibited the early step of HIV-1 (R9 and JR-FL) infection, while they did not affect the late step. The alginate oligomer consisted of uronic acids and sulfated-galactan porphyran showed no significant inhibitory effects. In addition, ascophyllan and two fucoidans inhibited the early step of HBV infection in a dose-dependent manner. Furthermore, these polysaccharides inhibited the early step of HCV infection but had no inhibitory effects on HTLV-1 replication. To further examine the specificity of these polysaccharides in viral infections, we used vesicular stomatitis virus (VSV)-G-pseudotyped HIV-1 infection. Ascophyllan, the two fucoidans, and alginate oligomer also potently inhibited VSV-G-pseudotyped HIV-1 infection in HeLa cells. Taken together, these results suggest that the acidic polysaccharides used in this study are capable of inhibiting the early step of viral infections depending on the polysaccharides but not in a strict species-specific manner.


Subject(s)
Aquatic Organisms/chemistry , Polysaccharides/chemistry , Virus Diseases/drug therapy , Virus Replication/drug effects , Acids/chemistry , Cyanobacteria/chemistry , HIV-1/drug effects , HIV-1/pathogenicity , Hepacivirus/drug effects , Hepacivirus/pathogenicity , Hepatitis B virus/drug effects , Hepatitis B virus/pathogenicity , Human T-lymphotropic virus 1/drug effects , Human T-lymphotropic virus 1/pathogenicity , Humans , Polysaccharides/isolation & purification , Polysaccharides/pharmacology , Virus Diseases/virology
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...