Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters











Database
Type of study
Language
Publication year range
1.
Proc Natl Acad Sci U S A ; 120(31): e2216127120, 2023 08.
Article in English | MEDLINE | ID: mdl-37487091

ABSTRACT

Retroviruses and their host have coevolved in a delicate balance between viral replication and survival of the infected cell. In this equilibrium, restriction factors expressed by infected cells control different steps of retroviral replication such as entry, uncoating, nuclear import, expression, or budding. Here, we describe a mechanism of restriction against human T cell leukemia virus type 1 (HTLV-1) by the helicase-like transcription factor (HLTF). We show that RNA and protein levels of HLTF are reduced in primary T cells of HTLV-1-infected subjects, suggesting a clinical relevance. We further demonstrate that the viral oncogene Tax represses HLTF transcription via the Enhancer of zeste homolog 2 methyltransferase of the Polycomb repressive complex 2. The Tax protein also directly interacts with HLTF and induces its proteasomal degradation. RNA interference and gene transduction in HTLV-1-infected T cells derived from patients indicate that HLTF is a restriction factor. Restoring the normal levels of HLTF expression induces the dispersal of the Golgi apparatus and overproduction of secretory granules. By synergizing with Tax-mediated NF-κB activation, physiologically relevant levels of HLTF intensify the autophagic flux. Increased vesicular trafficking leads to an enlargement of the lysosomes and the production of large vacuoles containing viral particles. HLTF induction in HTLV-1-infected cells significantly increases the percentage of defective virions. In conclusion, HLTF-mediated activation of the autophagic flux blunts the infectious replication cycle of HTLV-1, revealing an original mode of viral restriction.


Subject(s)
Human T-lymphotropic virus 1 , Leukemia, T-Cell , Humans , Human T-lymphotropic virus 1/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Gene Products, tax/genetics , Gene Products, tax/metabolism , T-Lymphocytes/metabolism , NF-kappa B/metabolism , DNA-Binding Proteins
2.
Front Immunol ; 13: 980514, 2022.
Article in English | MEDLINE | ID: mdl-36032174

ABSTRACT

Previous attempts to develop a vaccine against bovine leukemia virus (BLV) have not been successful because of inadequate or short-lived stimulation of all immunity components. In this study, we designed an approach based on an attenuated BLV provirus by deleting genes dispensable for infectivity but required for efficient replication. The ability of the vaccine to protect from natural BLV infection was investigated in the context of dairy productive conditions in an endemic region. The attenuated vaccine was tested in a farm in which the prevalence rose from 16.7% in young cattle at the beginning of the study to more than 90% in adult individuals. Sterilizing immunity was obtained in 28 out of 29 vaccinated heifers over a period of 48 months, demonstrating the effectiveness of the vaccine. As indicated by the antiviral antibody titers, the humoral response was slightly reduced compared to wild-type infection. After initial post-vaccination bursts, the proviral loads of the attenuated vaccine remained most frequently undetectable. During the first dairy cycle, proviral DNA was not detected by nested-PCR in milk samples from vaccinated cows. During the second dairy cycle, provirus was sporadically detected in milk of two vaccinated cows. Forty-two calves born from vaccinated cows were negative for proviral DNA but had antiviral antibodies in their peripheral blood. The attenuated strain was not transmitted to sentinels, further supporting the safety of the vaccine. Altogether, these data thus demonstrate that the vaccine against BLV is safe and effective in herd conditions characterized by a very high incidence. This cost-effective approach will thus decrease the prevalence of BLV without modification of production practices. After facing a series of challenges pertaining to effectiveness and biosafety, the vaccine is now available for further large-scale delivery. The different challenges and hurdles that were bypassed may be informative for the development of a vaccine against HTLV-1.


Subject(s)
Enzootic Bovine Leukosis , Leukemia Virus, Bovine , Animals , Antiviral Agents , Cattle , Female , Proviruses , Vaccines, Attenuated
3.
Cancers (Basel) ; 14(9)2022 Apr 26.
Article in English | MEDLINE | ID: mdl-35565292

ABSTRACT

The composition of the tumor microenvironment (TME) mediates the outcome of chemo- and immunotherapies in malignant pleural mesothelioma (MPM). Tumor-associated macrophages (TAMs) and monocyte myeloid-derived immunosuppressive cells (M-MDSCs) constitute a major fraction of the TME. As central cells of the innate immune system, monocytes exert well-characterized functions of phagocytosis, cytokine production, and antibody-dependent cell-mediated cytotoxicity (ADCC). The objective of this study was to evaluate the ability of monocytes to exert a direct cytotoxicity by cell-to-cell contact with MPM cells. The experimental model is based on cocultures between human blood-derived monocytes sorted by negative selection and mesothelioma cell lines. Data show (i) that blood-derived human monocytes induce tumor cell death by direct cell-to-cell contact, (ii) that VPA is a pharmacological enhancer of this cytotoxic activity, (iii) that VPA increases monocyte migration and their aggregation with MPM cells, and (iv) that the molecular mechanisms behind VPA modulation of monocytes involve a downregulation of the membrane receptors associated with the M2 phenotype, i.e., CD163, CD206, and CD209. These conclusions, thus, broaden our understanding about the molecular mechanisms involved in immunosurveillance of the tumor microenvironment and open new prospects for further improvement of still unsatisfactory MPM therapies.

4.
PLoS Pathog ; 16(5): e1008502, 2020 05.
Article in English | MEDLINE | ID: mdl-32407379

ABSTRACT

Viruses have developed different strategies to escape from immune response. Among these, viral non-coding RNAs are invisible to the immune system and may affect the fate of the host cell. Bovine leukemia virus (BLV) encodes both short (miRNAs) and long (antisense AS1 and AS2) non-coding RNAs. To elucidate the mechanisms associated with BLV non-coding RNAs, we performed phenotypic and transcriptomic analyzes in a reverse genetics system. RNA sequencing of B-lymphocytes revealed that cell proliferation is the most significant mechanism associated with ablation of the viral non-coding RNAs. To assess the biological relevance of this observation, we determined the cell kinetic parameters in vivo using intravenous injection of BrdU and CFSE. Fitting the data to a mathematical model provided the rates of cell proliferation and death. Our data show that deletion of miRNAs correlates with reduced proliferation of the infected cell and lack of pathogenesis.


Subject(s)
B-Lymphocytes , Cell Transformation, Viral , Leukemia Virus, Bovine , MicroRNAs , RNA, Antisense , RNA, Viral , Animals , B-Lymphocytes/metabolism , B-Lymphocytes/pathology , B-Lymphocytes/virology , Cattle , Cell Proliferation , Leukemia Virus, Bovine/genetics , Leukemia Virus, Bovine/metabolism , Leukemia Virus, Bovine/pathogenicity , MicroRNAs/genetics , MicroRNAs/metabolism , RNA, Antisense/genetics , RNA, Antisense/metabolism , RNA, Viral/genetics , RNA, Viral/metabolism
5.
Retrovirology ; 16(1): 26, 2019 10 07.
Article in English | MEDLINE | ID: mdl-31590667

ABSTRACT

Vaccination against retroviruses is a challenge because of their ability to stably integrate into the host genome, undergo long-term latency in a proportion of infected cells and thereby escape immune response. Since clearance of the virus is almost impossible once infection is established, the primary goal is to achieve sterilizing immunity. Besides efficacy, safety is the major issue since vaccination has been associated with increased infection or reversion to pathogenicity. In this review, we discuss the different issues that we faced during the development of an efficient vaccine against bovine leukemia virus (BLV). We summarize the historical failures of inactivated vaccines, the efficacy and safety of a live-attenuated vaccine and the economical constraints of further industrial development.


Subject(s)
Enzootic Bovine Leukosis/prevention & control , Leukemia Virus, Bovine/immunology , Viral Vaccines/immunology , Animals , Antibodies, Viral/immunology , Cattle , Vaccination/veterinary , Vaccines, Attenuated/immunology
6.
Viruses ; 7(11): 6080-8, 2015 Nov 24.
Article in English | MEDLINE | ID: mdl-26610551

ABSTRACT

Different animal models have been proposed to investigate the mechanisms of Human T-lymphotropic Virus (HTLV)-induced pathogenesis: rats, transgenic and NOD-SCID/γcnull (NOG) mice, rabbits, squirrel monkeys, baboons and macaques. These systems indeed provide useful information but have intrinsic limitations such as lack of disease relevance, species specificity or inadequate immune response. Another strategy based on a comparative virology approach is to characterize a related pathogen and to speculate on possible shared mechanisms. In this perspective, bovine leukemia virus (BLV), another member of the deltaretrovirus genus, is evolutionary related to HTLV-1. BLV induces lymphoproliferative disorders in ruminants providing useful information on the mechanisms of viral persistence, genetic determinants of pathogenesis and potential novel therapies.


Subject(s)
Host-Pathogen Interactions , Human T-lymphotropic virus 1/physiology , Leukemia Virus, Bovine/physiology , Animals , Biomedical Research/trends , Disease Models, Animal , Human T-lymphotropic virus 1/pathogenicity , Humans , Leukemia Virus, Bovine/pathogenicity , Virology/trends
7.
J Virol ; 89(17): 8945-56, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26085161

ABSTRACT

UNLABELLED: Viruses have coevolved with their host to ensure efficient replication and transmission without inducing excessive pathogenicity that would indirectly impair their persistence. This is exemplified by the bovine leukemia virus (BLV) system in which lymphoproliferative disorders develop in ruminants after latency periods of several years. In principle, the equilibrium reached between the virus and its host could be disrupted by emergence of more pathogenic strains. Intriguingly but fortunately, such a hyperpathogenic BLV strain was never observed in the field or designed in vitro. In this study, we sought to understand the role of envelope N-linked glycosylation with the hypothesis that this posttranslational modification could either favor BLV infection by allowing viral entry or allow immune escape by using glycans as a shield. Using reverse genetics of an infectious molecular provirus, we identified a N-linked envelope glycosylation site (N230) that limits viral replication and pathogenicity. Indeed, mutation N230E unexpectedly leads to enhanced fusogenicity and protein stability. IMPORTANCE: Infection by retroviruses requires the interaction of the viral envelope protein (SU) with a membrane-associated receptor allowing fusion and release of the viral genomic RNA into the cell. We show that N-linked glycosylation of the bovine leukemia virus (BLV) SU protein is, as expected, essential for cell infection in vitro. Consistently, mutation of all glycosylation sites of a BLV provirus destroys infectivity in vivo. However, single mutations do not significantly modify replication in vivo. Instead, a particular mutation at SU codon 230 increases replication and accelerates pathogenesis. This unexpected observation has important consequences in terms of disease control and managing.


Subject(s)
Leukemia Virus, Bovine/genetics , Leukemia Virus, Bovine/pathogenicity , Viral Envelope Proteins/genetics , Virus Replication/genetics , Amino Acid Sequence , Animals , Base Sequence , COS Cells , Cats , Cell Fusion , Chlorocebus aethiops , Glycosylation , HEK293 Cells , HeLa Cells , Humans , Leukemia Virus, Bovine/metabolism , Membrane Fusion/genetics , Mutation , Protein Stability , Sequence Alignment , Sequence Analysis, RNA , Sheep , Viral Envelope Proteins/metabolism , Viral Load
SELECTION OF CITATIONS
SEARCH DETAIL