Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
mBio ; 14(2): e0352622, 2023 04 25.
Artigo em Inglês | MEDLINE | ID: mdl-36802226

RESUMO

The human T-cell leukemia virus (HTLV)-1 is responsible for an aggressive neurodegenerative disease (HAM/TSP) and multiple neurological alterations. The capacity of HTLV-1 to infect central nervous system (CNS) resident cells, together with the neuroimmune-driven response, has not been well-established. Here, we combined the use of human induced pluripotent stem cells (hiPSC) and of naturally STLV-1-infected nonhuman primates (NHP) as models with which to investigate HTLV-1 neurotropism. Hence, neuronal cells obtained after hiPSC differentiation in neural polycultures were the main cell population infected by HTLV-1. Further, we report the infection of neurons with STLV-1 in spinal cord regions as well as in brain cortical and cerebellar sections of postmortem NHP. Additionally, reactive microglial cells were found in infected areas, suggesting an immune antiviral response. These results emphasize the need to develop new efficient models by which to understand HTLV-1 neuroinfection and suggest an alternative mechanism that leads to HAM/TSP.


Assuntos
Vírus Linfotrópico T Tipo 1 Humano , Células-Tronco Pluripotentes Induzidas , Doenças Neurodegenerativas , Vírus Linfotrópico T Tipo 1 de Símios , Animais , Humanos , Encéfalo , Vírus Linfotrópico T Tipo 1 Humano/fisiologia , Primatas , Neurônios
3.
Retrovirology ; 18(1): 6, 2021 02 23.
Artigo em Inglês | MEDLINE | ID: mdl-33622348

RESUMO

BACKGROUND: The Human T-cell Lymphotropic Virus Type-1 (HTLV-1) is a blood-borne pathogen and etiological agent of Adult T-cell Leukemia/Lymphoma (ATLL) and HTLV-1 Associated Myelopathy/Tropical Spastic Paraparesis (HAM/TSP). HTLV-1 has currently infected up to 10 million globally with highly endemic areas in Japan, Africa, the Caribbean and South America. We have previously shown that Extracellular Vesicles (EVs) enhance HTLV-1 transmission by promoting cell-cell contact. RESULTS: Here, we separated EVs into subpopulations using differential ultracentrifugation (DUC) at speeds of 2 k (2000×g), 10 k (10,000×g), and 100 k (100,000×g) from infected cell supernatants. Proteomic analysis revealed that EVs contain the highest viral/host protein abundance in the 2 k subpopulation (2 k > 10 k > 100 k). The 2 k and 10 k populations contained viral proteins (i.e., p19 and Tax), and autophagy proteins (i.e., LC3 and p62) suggesting presence of autophagosomes as well as core histones. Interestingly, the use of 2 k EVs in an angiogenesis assay (mesenchymal stem cells + endothelial cells) caused deterioration of vascular-like-tubules. Cells commonly associated with the neurovascular unit (i.e., astrocytes, neurons, and macrophages) in the blood-brain barrier (BBB) showed that HTLV-1 EVs may induce expression of cytokines involved in migration (i.e., IL-8; 100 k > 2 k > 10 k) from astrocytes and monocyte-derived macrophages (i.e., IL-8; 2 k > 10 k). Finally, we found that EVs were able to promote cell-cell contact and viral transmission in monocytic cell-derived dendritic cell. The EVs from both 2 k and 10 k increased HTLV-1 spread in a humanized mouse model, as evidenced by an increase in proviral DNA and RNA in the Blood, Lymph Node, and Spleen. CONCLUSIONS: Altogether, these data suggest that various EV subpopulations induce cytokine expression, tissue damage, and viral spread.


Assuntos
Células Endoteliais/virologia , Vesículas Extracelulares/virologia , Vírus Linfotrópico T Tipo 1 Humano/fisiologia , Animais , Comunicação Celular , Citocinas/análise , Citocinas/genética , Citocinas/imunologia , Vesículas Extracelulares/imunologia , Vesículas Extracelulares/fisiologia , Feminino , Infecções por HTLV-I/virologia , Humanos , Camundongos , Camundongos Endogâmicos NOD , Camundongos Transgênicos , Proteômica , Células THP-1 , Células U937
4.
PLoS Pathog ; 15(10): e1008093, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31600344

RESUMO

ISG20 is a broad spectrum antiviral protein thought to directly degrade viral RNA. However, this mechanism of inhibition remains controversial. Using the Vesicular Stomatitis Virus (VSV) as a model RNA virus, we show here that ISG20 interferes with viral replication by decreasing protein synthesis in the absence of RNA degradation. Importantly, we demonstrate that ISG20 exerts a translational control over a large panel of non-self RNA substrates including those originating from transfected DNA, while sparing endogenous transcripts. This activity correlates with the protein's ability to localize in cytoplasmic processing bodies. Finally, these functions are conserved in the ISG20 murine ortholog, whose genetic ablation results in mice with increased susceptibility to viral infection. Overall, our results posit ISG20 as an important defense factor able to discriminate the self/non-self origins of the RNA through translation modulation.


Assuntos
Antivirais/farmacologia , Exorribonucleases/farmacologia , Biossíntese de Proteínas , RNA Viral/metabolismo , Estomatite Vesicular/imunologia , Vesiculovirus/imunologia , Replicação Viral/efeitos dos fármacos , Animais , Exorribonucleases/fisiologia , Células HeLa , Humanos , Camundongos , Camundongos Knockout , Estabilidade de RNA , RNA Viral/genética , Estomatite Vesicular/tratamento farmacológico , Estomatite Vesicular/virologia , Vesiculovirus/efeitos dos fármacos
5.
PLoS Pathog ; 15(2): e1007589, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30818370

RESUMO

Human T Lymphotropic virus (HTLV) infection can persist in individuals resulting, at least in part, from viral escape of the innate immunity, including inhibition of type I interferon response in infected T-cells. Plasmacytoid dendritic cells (pDCs) are known to bypass viral escape by their robust type I interferon production. Here, we demonstrated that pDCs produce type I interferons upon physical cell contact with HTLV-infected cells, yet pDC activation inversely correlates with the ability of the HTLV-producing cells to transmit infection. We show that pDCs sense surface associated-HTLV present with glycan-rich structure referred to as biofilm-like structure, which thus represents a newly described viral structure triggering the antiviral response by pDCs. Consistently, heparan sulfate proteoglycans and especially the cell surface pattern of terminal ß-galactoside glycosylation, modulate the transmission of the immunostimulatory RNA to pDCs. Altogether, our results uncover a function of virus-containing cell surface-associated glycosylated structures in the activation of innate immunity.


Assuntos
Células Dendríticas/fisiologia , Infecções por HTLV-I/metabolismo , Citocinas , Galactosídeos/metabolismo , Glicosilação , Infecções por HTLV-I/imunologia , Vírus Linfotrópico T Tipo 1 Humano/imunologia , Vírus Linfotrópico T Tipo 1 Humano/patogenicidade , Vírus Linfotrópico T Tipo 2 Humano/imunologia , Vírus Linfotrópico T Tipo 2 Humano/patogenicidade , Humanos , Imunidade Inata/fisiologia , Interferon Tipo I/imunologia , Interferon-alfa/imunologia , Interferon-alfa/metabolismo , Células Jurkat , Linfócitos T/imunologia , Linfócitos T/fisiologia
6.
PLoS Negl Trop Dis ; 12(10): e0006812, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-30273350

RESUMO

Simian T-Leukemia Virus type 1 and Simian Foamy Virus infect non-human primates. While STLV-1, as HTLV-1, causes Adult T-cell Leukemia/lymphoma, SFV infection is asymptomatic. Both retroviruses can be transmitted from NHPs to humans through bites that allow contact between infected saliva and recipient blood. Because both viruses infect CD4+ T-cells, they might interfere with each other replication, and this might impact viral transmission. Impact of STLV-1 co-infection on SFV replication was analyzed in 18 SFV-positive/STLV-1-negative and 18 naturally SFV/STLV-1 co-infected Papio anubis. Even if 9 animals were found STLV-1-positive in saliva, STLV-1 PVL was much higher in the blood. SFV proviruses were detected in the saliva of all animals. Interestingly, SFV proviral load was much higher in the blood of STLV-1/SFV co-infected animals, compared to STLV-1-negative animals. Given that soluble Tax protein can enter uninfected cells, we tested its effect on foamy virus promoter and we show that Tax protein can transactivate the foamy LTR. This demonstrates that true STLV-1 co-infection or Tax only has an impact on SFV replication and may influence the ability of the virus to be zoonotically transmitted as well as its ability to promote hematological abnormalities.


Assuntos
Coinfecção/virologia , Infecções por Deltaretrovirus/virologia , Infecções por Retroviridae/virologia , Vírus Linfotrópico T Tipo 1 de Símios/isolamento & purificação , Vírus Espumoso dos Símios/isolamento & purificação , Carga Viral , Animais , Sangue/virologia , Infecções por Deltaretrovirus/complicações , Transmissão de Doença Infecciosa , Papio anubis , Provírus/isolamento & purificação , Infecções por Retroviridae/complicações , Saliva/virologia , Replicação Viral
7.
Front Microbiol ; 9: 278, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29593659

RESUMO

Five to ten million individuals are infected by Human T-cell Leukemia Virus type 1 (HTLV-1). HTLV-1 is transmitted through prolonged breast-feeding, by sexual contacts and by transmission of infected T lymphocytes through blood transfusion. One to ten percent of infected carriers will develop a severe HTLV-1-associated disease: Adult-T-cell leukemia/lymphoma (ATLL), or a neurological disorder named Tropical Spastic Paraparesis/HTLV-1 Associated Myelopathy (TSP/HAM). In vivo, HTLV-1 is mostly detected in CD4+ T-cells, and to a lesser extent in CD8+ T cells and dendritic cells. There is a strong correlation between HTLV-1 proviral load (PVL) and clinical status of infected individuals. Thus, reducing PVL could be part of a strategy to prevent or treat HTLV-1-associated diseases among carriers. Treatment of ATLL patients using conventional chemotherapy has very limited benefit. Some chronic and acute ATLL patients are, however, efficiently treated with a combination of interferon α and zidovudine (IFN-α/AZT), to which arsenic trioxide is added in some cases. On the other hand, no efficient treatment for TSP/HAM patients has been described yet. It is therefore crucial to develop therapies that could either prevent the occurrence of HTLV-1-associated diseases or at least block the evolution of the disease in the early stages. In vivo, reverse transcriptase (RT) activity is low in infected cells, which is correlated with a clonal mode of viral replication. This renders infected cells resistant to nucleoside RT inhibitors such as AZT. However, histone deacetylase inhibitors (HDACi) associated to AZT efficiently induces viral expression and prevent de novo cellular infection. In asymptomatic STLV-1 infected non-human primates, HDACi/AZT combination allows a strong decrease in the PVL. Unfortunately, rebound in the PVL occurs when the treatment is stopped, highlighting the need for better antiviral compounds. Here, we review previously used strategies targeting HTLV-1 replication. We also tested a series of HIV-1 RT inhibitors in an in vitro anti-HTLV-1 screen, and report that bis-POM-PMEA (adefovir dipivoxil) and bis-POC-PMPA (tenofovir disoproxil) are much more efficient compared to AZT to decrease HTLV-1 cell-to-cell transmission in vitro. Our results suggest that revisiting already established antiviral drugs is an interesting approach to discover new anti-HTLV-1 drugs.

9.
PLoS Pathog ; 13(4): e1006353, 2017 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-28426803

RESUMO

Human T lymphotropic Virus type 1 (HTLV-1) is the etiological agent of Adult T cell Leukemia/Lymphoma (ATLL) and HTLV-1-Associated Myelopathy/Tropical Spastic Paraparesis (HAM/TSP). Both CD4+ T-cells and dendritic cells (DCs) infected with HTLV-1 are found in peripheral blood from HTLV-1 carriers. We previously demonstrated that monocyte-derived IL-4 DCs are more susceptible to HTLV-1 infection than autologous primary T-cells, suggesting that DC infection precedes T-cell infection. However, during blood transmission, breast-feeding or sexual transmission, HTLV-1 may encounter different DC subsets present in the blood, the intestinal or genital mucosa respectively. These different contacts may impact HTLV-1 ability to infect DCs and its subsequent transfer to T-cells. Using in vitro monocyte-derived IL-4 DCs, TGF-ß DCs and IFN-α DCs that mimic DCs contacting HTLV-1 in vivo, we show here that despite their increased ability to capture HTLV-1 virions, IFN-α DCs restrict HTLV-1 productive infection. Surprisingly, we then demonstrate that it is not due to the antiviral activity of type-I interferon produced by IFN-α DCs, but that it is likely to be linked to a distinct trafficking route of HTLV-1 in IL-4 DCs vs. IFN-α DCs. Finally, we demonstrate that, in contrast to IL-4 DCs, IFN-α DCs are impaired in their capacity to transfer HTLV-1 to CD4 T-cells, both after viral capture and trans-infection and after their productive infection. In conclusion, the nature of the DCs encountered by HTLV-1 upon primo-infection and the viral trafficking route through the vesicular pathway of these cells determine the efficiency of viral transmission to T-cells, which may condition the fate of infection.


Assuntos
Antivirais/farmacologia , Citocinas/imunologia , Células Dendríticas/imunologia , Infecções por HTLV-I/imunologia , Vírus Linfotrópico T Tipo 1 Humano/imunologia , Paraparesia Espástica Tropical/imunologia , Adulto , Células Dendríticas/virologia , Infecções por HTLV-I/transmissão , Infecções por HTLV-I/virologia , Vírus Linfotrópico T Tipo 1 Humano/patogenicidade , Vírus Linfotrópico T Tipo 1 Humano/fisiologia , Humanos , Interferon Tipo I/imunologia , Modelos Biológicos , Paraparesia Espástica Tropical/patologia , Paraparesia Espástica Tropical/virologia , Linfócitos T/imunologia , Linfócitos T/virologia
10.
Methods Mol Biol ; 1582: 47-55, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28357661

RESUMO

Unlike HIV-1, HTLV-1 viral transmission requires cell-to-cell contacts, while cell-free virions are poorly infectious and almost absent from body fluids. Though the virus uses three nonexclusive mechanisms to infect new target cells: (1) MTOC polarization followed by formation of a virological synapse and viral transfer into a synaptic cleft, (2) genesis of a viral biofilm and its transfer of embedded viruses, or (3) HTLV-1 transmission using conduits. The Tax transactivator and the p8 viral proteins are involved in virological synapse and nanotube formation respectively.HTLV-1 transcription from the viral promoter (i.e., LTR) requires the Tax protein that is absent from the viral particle and is expressed after productive infection. The present chapter focuses on a series of protocols used to quantify HTLV-1 de novo infection of target cells. These techniques do not discriminate between the different modes of transmission, but allow an accurate measure of productive infection. We used cell lines that are stably transfected with LTR-GFP or LTR-luciferase plasmids and quantified Green Fluorescent Protein expression or luciferase activity, since both of them reflect Tax expression.


Assuntos
Produtos do Gene tax/metabolismo , Genes Reporter , Infecções por HTLV-I/metabolismo , Infecções por HTLV-I/transmissão , Vírus Linfotrópico T Tipo 1 Humano/metabolismo , Luciferases/biossíntese , Técnicas de Cocultura , Produtos do Gene tax/genética , Infecções por HTLV-I/genética , Vírus Linfotrópico T Tipo 1 Humano/genética , Humanos , Células Jurkat , Luciferases/genética
11.
Cancer Lett ; 389: 78-85, 2017 03 28.
Artigo em Inglês | MEDLINE | ID: mdl-28034804

RESUMO

HTLV-1 causes Adult T cell Leukemia/Lymphoma (ATLL) in humans. We describe an ATL-like disease in a 9 year-old female baboon naturally infected with STLV-1 (the simian counterpart of HTLV-1), with a lymphocyte count over 1010/L, lymphocytes with abnormal nuclear morphology, and pulmonary and skin lesions. The animal was treated with a combination of AZT and alpha interferon. Proviral load (PVL) was measured every week. Because the disease continued to progress, the animal was euthanized. Abnormal infiltrates of CD3+CD25+ lymphocytes and Tax-positive cells were found by histological analyses in both lymphoid and non-lymphoid organs. PVL was measured and clonal diversity was assessed by LM-PCR (Ligation-Mediated Polymerase Chain Reaction) and high throughput sequencing, in blood during treatment and in 14 different organs. The highest PVL was found in lymph nodes, spleen and lungs. One major clone and a number of intermediate abundance clones were present in blood throughout the course of treatment, and in organs. These results represent the first multi-organ clonality study in ATLL. We demonstrate a previously undescribed clonal complexity in ATLL. Our data reinforce the usefulness of natural STLV-1 infection as a model of ATLL.


Assuntos
Infecções por Deltaretrovirus/veterinária , Doenças dos Macacos/patologia , Vírus Linfotrópico T Tipo 1 de Símios , Animais , Infecções por Deltaretrovirus/tratamento farmacológico , Infecções por Deltaretrovirus/patologia , Infecções por Deltaretrovirus/virologia , Modelos Animais de Doenças , Feminino , Interferon-alfa/farmacologia , Leucemia-Linfoma de Células T do Adulto/patologia , Linfócitos/patologia , Doenças dos Macacos/tratamento farmacológico , Doenças dos Macacos/virologia , Papio , Carga Viral , Zidovudina/farmacologia
12.
J Virol ; 89(20): 10580-90, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26269171

RESUMO

UNLABELLED: Human T-cell leukemia virus type 1 (HTLV-1)-infected CD4(+) T cells and dendritic cells (DCs) are present in peripheral blood from HTLV-1 carriers. While T-cell infection requires cell-cell contact, DCs might be infected with cell-free virus, at least in vitro. However, a thorough comparison of the susceptibilities of the two cell types to HTLV-1 infection using cell-associated and cell-free viral sources has not been performed. We first determined that human primary monocyte-derived dendritic cells (MDDCs) were more susceptible to HTLV-1 infection than their autologous lymphocyte counterparts after contact with chronically infected cells. Next, a comparison of infection efficiency using nonconcentrated or concentrated supernatants from infected cells as well as purified viral biofilm was performed. Integrated provirus was found after exposure of MDDCs or primary lymphocytes to viral biofilm but not to a viral supernatant. Using a large series of primary cell samples (n = 21), we demonstrated a higher proviral load in MDDCs exposed to viral biofilm than in lymphocytes. This higher susceptibility is correlated to a higher expression of neuropilin-1 on MDDCs than on autologous activated T lymphocytes. Moreover, we show that MDDCs infected with viral biofilm can transmit the virus to lymphocytes. In conclusion, MDDCs are more susceptible to HTLV-1 infection than autologous lymphocytes in vitro, supporting a model in which DC infection might represent an important step during primo-infection in vivo. IMPORTANCE: HTLV-1 is able to infect several cell types, but viral DNA is mainly found in T lymphocytes in vivo. This supports a model in which T lymphocytes are the main target of infection. However, during the primo-infection of new individuals, incoming viruses might first encounter dendritic cells (DCs), the specialized immune cells responsible for the antiviral response of the host. HTLV-1 cell-free purified viruses can infect dendritic cells in vitro, while T-cell infection is restricted to cell-to-cell transmission. In order to understand the sequence of HTLV-1 dissemination, we undertook a direct comparison of the susceptibilities of the two cell types using cell-associated and cell-free viral sources. We report here that MDDCs are more susceptible to HTLV-1 infection than autologous lymphocytes in vitro and are able to efficiently transmit the virus to lymphocytes. Our results suggest that DCs may represent a true viral reservoir, as the first cell type to be infected in vivo.


Assuntos
Células Dendríticas/virologia , Interações Hospedeiro-Patógeno/imunologia , Vírus Linfotrópico T Tipo 1 Humano/fisiologia , Linfócitos T/virologia , Linhagem Celular , Técnicas de Cocultura , Células Dendríticas/imunologia , Expressão Gênica , Vírus Linfotrópico T Tipo 1 Humano/patogenicidade , Humanos , Células Jurkat , Neuropilina-1/genética , Neuropilina-1/imunologia , Especificidade de Órgãos , Cultura Primária de Células , Transdução de Sinais , Linfócitos T/imunologia , Carga Viral/fisiologia , Replicação Viral/fisiologia
13.
Cell Mol Life Sci ; 72(22): 4409-27, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26047659

RESUMO

Exosomes are secreted membrane vesicles of endosomal origin present in biological fluids. Exosomes may serve as shuttles for amyloidogenic proteins, notably infectious prions, and may participate in their spreading in vivo. To explore the significance of the exosome pathway on prion infectivity and release, we investigated the role of the endosomal sorting complex required for transport (ESCRT) machinery and the need for ceramide, both involved in exosome biogenesis. Silencing of HRS-ESCRT-0 subunit drastically impairs the formation of cellular infectious prion due to an altered trafficking of cholesterol. Depletion of Tsg101-ESCRT-I subunit or impairment of the production of ceramide significantly strongly decreases infectious prion release. Together, our data reveal that ESCRT-dependent and -independent pathways can concomitantly regulate the exosomal secretion of infectious prion, showing that both pathways operate for the exosomal trafficking of a particular cargo. These data open up a new avenue to regulate prion release and propagation.


Assuntos
Complexos Endossomais de Distribuição Requeridos para Transporte/genética , Exossomos/genética , Príons/genética , Transdução de Sinais/genética , Compostos de Anilina/farmacologia , Animais , Compostos de Benzilideno/farmacologia , Linhagem Celular , Linhagem Celular Tumoral , Ceramidas/metabolismo , Proteínas de Ligação a DNA/genética , Exossomos/metabolismo , Exossomos/ultraestrutura , Humanos , Immunoblotting , Camundongos Transgênicos , Microscopia Confocal , Microscopia Eletrônica , Príons/metabolismo , Transporte Proteico/efeitos dos fármacos , Transporte Proteico/genética , Interferência de RNA , Coelhos , Ovinos , Fatores de Transcrição/genética
14.
Retrovirology ; 11: 93, 2014 Nov 12.
Artigo em Inglês | MEDLINE | ID: mdl-25389016

RESUMO

BACKGROUND: The role of innate immunity in general and of type I interferon (IFN-I) in particular in HTLV-1 pathogenesis is still a matter of debate. ADAR1-p150 is an Interferon Stimulated Gene (ISG) induced by IFN-I that can edit viral RNAs. We therefore investigated whether it could play the role of an anti-HTLV factor. RESULTS: We demonstrate here that ADAR1 is also expressed in the absence of IFN stimulation in activated primary T-lymphocytes that are the natural target of this virus and in HTLV-1 or HTLV-2 chronically infected T-cells. ADAR1 expression is also increased in primary lymphocytes obtained from HTLV-1 infected individuals. We show that ADAR1 enhances HTLV-1 and HTLV-2 infection in T-lymphocytes and that this proviral effect is independent from its editing activity. ADAR1 expression suppresses IFN-α inhibitory effect on HTLV-1 and HTLV-2 and acts through the repression of PKR phosphorylation. DISCUSSION: This study demonstrates that two interferon stimulated genes, i.e. PKR and ADAR1 have opposite effects on HTLV replication in vivo. The balanced expression of those proteins could determine the fate of the viral cycle in the course of infection.


Assuntos
Adenosina Desaminase/metabolismo , Interações Hospedeiro-Patógeno , Vírus Linfotrópico T Tipo 1 Humano/fisiologia , Vírus Linfotrópico T Tipo 2 Humano/fisiologia , Proteínas de Ligação a RNA/metabolismo , Replicação Viral , eIF-2 Quinase/antagonistas & inibidores , Células Cultivadas , Humanos , Inibição Psicológica , Dados de Sequência Molecular , Análise de Sequência de DNA , Linfócitos T/imunologia , Linfócitos T/virologia
15.
J Virol ; 87(24): 13386-96, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24089560

RESUMO

Type I interferon (IFN-I) inhibits the replication of different viruses. However, the effect of IFN-I on the human T-lymphotropic virus type 1 (HTLV-1) viral cycle is controversial. Here, we investigated the consequences of IFN-α addition for different steps of HTLV-1 and HTLV-2 infection. We first show that alpha interferon (IFN-α) efficiently impairs HTLV-1 and HTLV-2 de novo infection in a T cell line and in primary lymphocytes. Using pseudotyped viruses expressing HTLV-1 envelope, we then show that cell-free infection is insensitive to IFN-α, demonstrating that the cytokine does not affect the early stages of the viral cycle. In contrast, intracellular levels of Gag, Env, or Tax protein are affected by IFN-α treatment in T cells, primary lymphocytes, or 293T cells transfected with HTLV-1 or HTLV-2 molecular clones, demonstrating that IFN-α acts during the late stages of infection. We show that IFN-α does not affect Tax-mediated transcription and acts at a posttranscriptional level. Using either small interfering RNA (siRNA) directed against PKR or a PKR inhibitor, we demonstrate that PKR, whose expression is induced by interferon, plays a major role in IFN-α-induced HTLV-1/2 inhibition. These results indicate that IFN-α has a strong repressive effect on the HTLV-1 and HTLV-2 viral cycle during de novo infection of cells that are natural targets of the viruses.


Assuntos
Infecções por HTLV-I/enzimologia , Infecções por HTLV-II/enzimologia , Vírus Linfotrópico T Tipo 1 Humano/fisiologia , Vírus Linfotrópico T Tipo 2 Humano/fisiologia , Interferon-alfa/metabolismo , eIF-2 Quinase/metabolismo , Linhagem Celular , Ativação Enzimática , Infecções por HTLV-I/genética , Infecções por HTLV-I/virologia , Infecções por HTLV-II/genética , Infecções por HTLV-II/virologia , Interações Hospedeiro-Patógeno , Vírus Linfotrópico T Tipo 1 Humano/genética , Vírus Linfotrópico T Tipo 2 Humano/genética , Humanos , Interferon alfa-2 , Proteínas Recombinantes/metabolismo , eIF-2 Quinase/genética
16.
Nat Struct Mol Biol ; 19(5): 517-24, S1, 2012 Apr 08.
Artigo em Inglês | MEDLINE | ID: mdl-22484317

RESUMO

Despite intense research in the context of neurodegenerative diseases associated with its misfolding, the endogenous human prion protein PrP(C) (or PRNP) has poorly understood physiological functions. Whereas most PrP(C) is exposed to the extracellular environment, conserved domains result in transmembrane forms of PrP(C) that traffic in the endolysosomal system and are linked to inherited and infectious neuropathologies. One transmembrane PrP(C) variant orients the N-terminal 'octarepeat' domain into the cytoplasm. Here we demonstrate that the octarepeat domain of human PrP(C) contains GW/WG motifs that bind Argonaute (AGO) proteins, the essential components of microRNA (miRNA)-induced silencing complexes (miRISCs). Transmembrane PrP(C) preferentially binds AGO, and PrP(C) promotes formation or stability of miRISC effector complexes containing the trinucleotide repeat-containing gene 6 proteins (TNRC6) and miRNA-repressed mRNA. Accordingly, effective repression of several miRNA targets requires PrP(C). We propose that dynamic interactions between PrP(C)-enriched endosomes and subcellular foci of AGO underpin these effects.


Assuntos
Proteínas Argonautas/metabolismo , MicroRNAs/metabolismo , Proteínas PrPC/metabolismo , Sequência de Aminoácidos , Animais , Proteínas Argonautas/análise , Autoantígenos/metabolismo , Linhagem Celular , Humanos , Camundongos , Dados de Sequência Molecular , Proteínas PrPC/análise , Proteínas PrPC/química , Ligação Proteica , Estrutura Terciária de Proteína , RNA Mensageiro/metabolismo , Proteínas de Ligação a RNA/metabolismo , Ribonuclease III/metabolismo
17.
PLoS One ; 7(1): e30872, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22295118

RESUMO

Prion diseases are fatal, transmissible neurodegenerative diseases of the central nervous system. An abnormally protease-resistant and insoluble form (PrP(Sc)) of the normally soluble protease-sensitive host prion protein (PrP(C)) is the major component of the infectious prion. During the course of prion disease, PrP(Sc) accumulates primarily in the lymphoreticular and central nervous systems. Recent studies have shown that co-infection of prion-infected fibroblast cells with the Moloney murine leukemia virus (Mo-MuLV) strongly enhanced the release and spread of scrapie infectivity in cell culture, suggesting that retroviral coinfection might significantly influence prion spread and disease incubation times in vivo. We now show that another retrovirus, the murine leukemia virus Friend (F-MuLV), also enhanced the release and spread of scrapie infectivity in cell culture. However, peripheral co-infection of mice with both Friend virus and the mouse scrapie strain 22L did not alter scrapie disease incubation times, the levels of PrP(Sc) in the brain or spleen, or the distribution of pathological lesions in the brain. Thus, retroviral co-infection does not necessarily alter prion disease pathogenesis in vivo, most likely because of different cell-specific sites of replication for scrapie and F-MuLV.


Assuntos
Coinfecção , Vírus da Leucemia Murina de Friend/fisiologia , Proteínas PrPSc/metabolismo , Doenças Priônicas/virologia , Animais , Células Dendríticas Foliculares/metabolismo , Células Dendríticas Foliculares/virologia , Suscetibilidade a Doenças , Exossomos/metabolismo , Exossomos/virologia , Período de Incubação de Doenças Infecciosas , Camundongos , Células NIH 3T3 , Baço/imunologia
18.
Cell Mol Life Sci ; 69(8): 1331-52, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22076653

RESUMO

The cellular prion protein PrP(C)/CD230 is a GPI-anchor protein highly expressed in cells from the nervous and immune systems and well conserved among vertebrates. In the last decade, several studies suggested that PrP(C) displays antiviral properties by restricting the replication of different viruses, and in particular retroviruses such as murine leukemia virus (MuLV) and the human immunodeficiency virus type 1 (HIV-1). In this context, we previously showed that PrP(C) displays important similarities with the HIV-1 nucleocapsid protein and found that PrP(C) expression in a human cell line strongly reduced HIV-1 expression and virus production. Using different PrP(C) mutants, we report here that the anti-HIV-1 properties are mostly associated with the amino-terminal 24-KRPKP-28 basic domain. In agreement with its reported RNA chaperone activity, we found that PrP(C) binds to the viral genomic RNA of HIV-1 and negatively affects its translation. Using a combination of biochemical and cell imaging strategies, we found that PrP(C) colocalizes with the virus assembly machinery at the plasma membrane and at the virological synapse in infected T cells. Depletion of PrP(C) in infected T cells and microglial cells favors HIV-1 replication, confirming its negative impact on the HIV-1 life cycle.


Assuntos
HIV-1/crescimento & desenvolvimento , Proteínas PrPC/metabolismo , Linfócitos T/virologia , Antígenos CD/genética , Linhagem Celular , Células Cultivadas , Proteínas Ligadas por GPI/genética , Expressão Gênica , Técnicas de Silenciamento de Genes , Produtos do Gene gag/análise , HIV-1/genética , HIV-1/metabolismo , Humanos , Mutação , Proteínas PrPC/análise , Proteínas PrPC/química , Proteínas PrPC/genética , Biossíntese de Proteínas , Estrutura Terciária de Proteína , RNA Viral/genética , RNA Viral/metabolismo
19.
Prion ; 4(4): 292-301, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20930564

RESUMO

Neurodegenerative diseases are often associated with misfolding and deposition of specific proteins in the nervous system. The prion protein, which is associated with transmissible spongiform encephalopathies (TSEs), is one of them. The normal function of the cellular form of the prion protein (PrP(C)) is mediated through specific signal transduction pathways and is linked to resistance to oxidative stress, neuronal outgrowth and cell survival. In TSEs, PrP(C) is converted into an abnormally folded isoform, called PrP(Sc), that may impair the normal function of the protein and/or generate toxic aggregates. To investigate these molecular events we performed a two-dimensional gel electrophoresis comparison of neuroblastoma N2a cells expressing different amounts of PrP(C) and eventually infected with the 22L prion strain. Mass spectrometry and peptide mass fingerprint analysis identified a series of proteins with modified expression. They included the chaperones Grp78/BiP, protein disulfide-isomerase A6, Grp75 and Hsp60 which had an opposite expression upon PrPC expression and PrP(Sc) production. The detection of these proteins was coherent with the idea that protein misfolding plays an important role in TSEs. Other proteins, such as calreticulin, tubulin, vimentin or the laminin receptor had their expression modified in infected cells, which was reminiscent of previous results. Altogether our data provide molecular information linking PrP expression and misfolding, which could be the basis of further therapeutic and pathophysiological research in this field.


Assuntos
Neuroblastoma/metabolismo , Proteínas PrPC/metabolismo , Proteômica/métodos , Animais , Western Blotting , Extratos Celulares , Linhagem Celular Tumoral , Eletroforese em Gel Bidimensional , Chaperona BiP do Retículo Endoplasmático , Camundongos , Modelos Biológicos , Proteínas PrPC/patogenicidade , Proteínas PrPSc/metabolismo
20.
Biol Cell ; 100(10): 603-15, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18422484

RESUMO

BACKGROUND INFORMATION: TSEs (transmissible spongiform encephalopathies) are neurodegenerative disorders affecting humans and animals. PrP(Sc), a conformationally altered isoform of the normal prion protein (PrP(C)), is thought to be the pathogenic agent. However, the biochemical composition of the prion agent is still matter of debate. The potential transmission risk of the prion agent through biological fluids has been shown, but the development of competitive diagnostic tests and treatment for TSEs requires a more comprehensive knowledge of the agent and the cellular mechanisms by which it is disseminated. With this aim, we initiated characterization of the prion agent and the pathways by which it can be propagated using the cellular model system neuroblastoma (N2a). RESULTS: The present study shows that N2a cells infected with scrapie release the prion agent into the cell culture medium in association with exosome-like structures and viral particles of endogenous origin. We found that both prion proteins and scrapie infectivity are mainly associated with exosome-like structures that contain viral envelope glycoprotein and nucleic acids, such as RNAs. CONCLUSIONS: The dissemination of prions in N2a cell culture is mediated through the exosomal pathway.


Assuntos
Exossomos/metabolismo , Proteínas PrPC/metabolismo , Proteínas PrPSc/metabolismo , Doenças Priônicas/metabolismo , Scrapie/metabolismo , Animais , Linhagem Celular Tumoral , Exossomos/virologia , Camundongos , Neuroblastoma , Doenças Priônicas/virologia , Scrapie/virologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA