Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Trends Immunol ; 45(5): 346-357, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38632001

RESUMO

Mammalian brain border-associated macrophages (BAMs) are strategically positioned to support vital properties and processes: for example, the composition of the brain's perivascular extracellular matrix and cerebrospinal fluid flow via the glymphatic pathway. BAMs also effectively restrict the spread of infectious microbes into the brain. However, while fighting infections, BAMs sustain long-term transcriptomic changes and can be replaced by inflammatory monocytes, potentially leading to a gradual loss of their beneficial homeostatic functions. We hypothesize that by expediting the deterioration of BAMs, multiple infection episodes might be associated with accelerated brain aging and the putative development of neurodegenerative diseases. Our viewpoint is supported by recent studies suggesting that rejuvenating aged BAMs, and counterbalancing their detrimental inflammatory signatures during infections, might hold promise in treating aging-related neurological disorders, including Alzheimer's disease (AD).


Assuntos
Envelhecimento , Doença de Alzheimer , Encéfalo , Macrófagos , Animais , Humanos , Envelhecimento/imunologia , Doença de Alzheimer/imunologia , Encéfalo/imunologia , Encéfalo/patologia , Infecções/imunologia , Macrófagos/imunologia
2.
J Inflamm (Lond) ; 21(1): 3, 2024 Jan 30.
Artigo em Inglês | MEDLINE | ID: mdl-38291415

RESUMO

The brain and spinal cord collectively referred to as the Central Nervous System (CNS) are protected by the blood-brain barrier that limits molecular, microbial and immunological trafficking. However, in the last decade, many studies have emphasized the protective role of 'border regions' at the surface of the CNS which are highly immunologically active, in contrast with the CNS parenchyma. In the steady-state, lymphoid and myeloid cells residing in the cranial meninges can affect brain function and behavior. Upon infection, they provide a first layer of protection against microbial neuroinvasion. The maturation of border sites over time enables more effective brain protection in adults as compared to neonates. Here, we provide a comprehensive update on the meningeal immune system and its role in physiological brain function and protection against infectious agents.

3.
Glia ; 72(3): 475-503, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-37909340

RESUMO

Across the globe, approximately one in 10 babies are born preterm, that is, before 37 weeks of a typical 40 weeks of gestation. Up to 50% of preterm born infants develop brain injury, encephalopathy of prematurity (EoP), that substantially increases their risk for developing lifelong defects in motor skills and domains of learning, memory, emotional regulation, and cognition. We are still severely limited in our abilities to prevent or predict preterm birth. No longer just the "support cells," we now clearly understand that during development glia are key for building a healthy brain. Glial dysfunction is a hallmark of EoP, notably, microgliosis, astrogliosis, and oligodendrocyte injury. Our knowledge of glial biology during development is exponentially expanding but hasn't developed sufficiently for development of effective neuroregenerative therapies. This review summarizes the current state of knowledge for the roles of glia in infants with EoP and its animal models, and a description of known glial-cell interactions in the context of EoP, such as the roles for border-associated macrophages. The field of perinatal medicine is relatively small but has worked passionately to improve our understanding of the etiology of EoP coupled with detailed mechanistic studies of pre-clinical and human cohorts. A primary finding from this review is that expanding our collaborations with computational biologists, working together to understand the complexity of glial subtypes, glial maturation, and the impacts of EoP in the short and long term will be key to the design of therapies that improve outcomes.


Assuntos
Lesões Encefálicas , Nascimento Prematuro , Lactente , Gravidez , Animais , Feminino , Recém-Nascido , Humanos , Recém-Nascido Prematuro , Neuroglia , Encéfalo
4.
Nat Commun ; 14(1): 4290, 2023 07 18.
Artigo em Inglês | MEDLINE | ID: mdl-37463962

RESUMO

Endo-lysosomes transport along microtubules and clustering in the perinuclear area are two necessary steps for microbes to activate specialized phagocyte functions. We report that RUN and FYVE domain-containing protein 3 (RUFY3) exists as two alternative isoforms distinguishable by the presence of a C-terminal FYVE domain and by their affinity for phosphatidylinositol 3-phosphate on endosomal membranes. The FYVE domain-bearing isoform (iRUFY3) is preferentially expressed in primary immune cells and up-regulated upon activation by microbes and Interferons. iRUFY3 is necessary for ARL8b + /LAMP1+ endo-lysosomes positioning in the pericentriolar organelles cloud of LPS-activated macrophages. We show that iRUFY3 controls macrophages migration, MHC II presentation and responses to Interferon-γ, while being important for intracellular Salmonella replication. Specific inactivation of rufy3 in phagocytes leads to aggravated pathologies in mouse upon LPS injection or bacterial pneumonia. This study highlights the role of iRUFY3 in controlling endo-lysosomal dynamics, which contributes to phagocyte activation and immune response regulation.


Assuntos
Apresentação de Antígeno , Lipopolissacarídeos , Animais , Camundongos , Endossomos/metabolismo , Lipopolissacarídeos/metabolismo , Lisossomos/metabolismo , Fagócitos
5.
Eur J Immunol ; 53(9): e2250267, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37402972

RESUMO

In the past 10 years, important discoveries have been made in the field of neuroimmunology, especially regarding brain borders. Indeed, meninges are protective envelopes surrounding the CNS and are currently in the spotlight, with multiple studies showing their involvement in brain infection and cognitive disorders. In this review, we describe the meningeal layers and their protective role in the CNS against bacterial, viral, fungal, and parasitic infections, by immune and nonimmune cells. Moreover, we discuss the neurological and cognitive consequences resulting from meningeal infections in neonates (e.g. infection with group B Streptococcus, cytomegalovirus, …) or adults (e.g. infection with Trypanosoma brucei, Streptococcus pneumoniae, …). We hope that this review will bring to light an integrated view of meningeal immune regulations during CNS infections and their neurological consequences.


Assuntos
Infecções do Sistema Nervoso Central , Meninges , Adulto , Recém-Nascido , Humanos , Encéfalo , Streptococcus pneumoniae
6.
Immunity ; 56(4): 744-746, 2023 04 11.
Artigo em Inglês | MEDLINE | ID: mdl-37044064

RESUMO

How can beneficial microorganisms be distinguished from pathogenic ones? In this issue of Immunity, Peterson et al. discovered that a specific phenazine, which is part of a family of toxic metabolites expressed by pathogenic bacteria, is detected by Caenorhabditis elegans by directly binding to a nuclear hormone receptor, promoting the expression of detoxifying enzymes and immunity-related genes, thus protecting the worm.


Assuntos
Proteínas de Caenorhabditis elegans , Animais , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/genética , Receptores Citoplasmáticos e Nucleares/genética , Receptores Citoplasmáticos e Nucleares/metabolismo , Regulação da Expressão Gênica , Transdução de Sinais
7.
Nat Immunol ; 24(4): 714-728, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-36928414

RESUMO

Plasmacytoid dendritic cells (pDCs) are the main source of type I interferon (IFN-I) during viral infections. Their other functions are debated, due to a lack of tools to identify and target them in vivo without affecting pDC-like cells and transitional DCs (tDCs), which harbor overlapping phenotypes and transcriptomes but a higher efficacy for T cell activation. In the present report, we present a reporter mouse, pDC-Tom, designed through intersectional genetics based on unique Siglech and Pacsin1 coexpression in pDCs. The pDC-Tom mice specifically tagged pDCs and, on breeding with Zbtb46GFP mice, enabled transcriptomic profiling of all splenic DC types, unraveling diverging activation of pDC-like cells versus tDCs during a viral infection. The pDC-Tom mice also revealed initially similar but later divergent microanatomical relocation of splenic IFN+ versus IFN- pDCs during infection. The mouse models and specific gene modules we report here will be useful to delineate the physiological functions of pDCs versus other DC types.


Assuntos
Células Dendríticas , Interferon Tipo I , Animais , Camundongos , Interferon Tipo I/metabolismo , Perfilação da Expressão Gênica , Fenótipo , Transcriptoma
8.
STAR Protoc ; 4(1): 102119, 2023 03 17.
Artigo em Inglês | MEDLINE | ID: mdl-36853673

RESUMO

The highly vascularized meninges protect the surface of the central nervous system and contain a dense network of immune cells controlling neuroinfection and neuroinflammation. Here, we present techniques for the immunological and virological assessment of mouse dural meninges. We describe steps for immunophenotyping including meninges extraction and digestion, immunostaining, and flow cytometry. We then describe viral assessment upon lymphocytic choriomeningitis virus infection including steps for fixation of the meninges in the skull, whole-mount immunohistochemistry, and confocal imaging. For complete details on the use and execution of this protocol, please refer to Rebejac et al. (2022).1.


Assuntos
Sistema Nervoso Central , Meninges , Animais , Camundongos , Citometria de Fluxo , Imuno-Histoquímica , Meninges/diagnóstico por imagem , Cabeça
9.
Immunity ; 55(11): 2103-2117.e10, 2022 11 08.
Artigo em Inglês | MEDLINE | ID: mdl-36323311

RESUMO

The surface of the central nervous system (CNS) is protected by the meninges, which contain a dense network of meningeal macrophages (MMs). Here, we examined the role of tissue-resident MM in viral infection. MHC-II- MM were abundant neonatally, whereas MHC-II+ MM appeared over time. These barrier macrophages differentially responded to in vivo peripheral challenges such as LPS, SARS-CoV-2, and lymphocytic choriomeningitis virus (LCMV). Peripheral LCMV infection, which was asymptomatic, led to a transient infection and activation of the meninges. Mice lacking macrophages but conserving brain microglia, or mice bearing macrophage-specific deletion of Stat1 or Ifnar, exhibited extensive viral spread into the CNS. Transcranial pharmacological depletion strategies targeting MM locally resulted in several areas of the meninges becoming infected and fatal meningitis. Low numbers of MHC-II+ MM, which is seen upon LPS challenge or in neonates, corelated with higher viral load upon infection. Thus, MMs protect against viral infection and may present targets for therapeutic manipulation.


Assuntos
COVID-19 , Coriomeningite Linfocítica , Animais , Camundongos , Lipopolissacarídeos , Camundongos Endogâmicos C57BL , SARS-CoV-2 , Vírus da Coriomeningite Linfocítica/fisiologia , Macrófagos , Meninges
10.
Sci Rep ; 12(1): 8804, 2022 05 25.
Artigo em Inglês | MEDLINE | ID: mdl-35614095

RESUMO

A system of lymphatic vessels has been recently characterized in the meninges, with a postulated role in 'cleaning' the brain via cerebral fluid drainage. As meninges are the origin site of migraine pain, we hypothesized that malfunctioning of the lymphatic system should affect the local trigeminal nociception. To test this hypothesis, we studied nociceptive and inflammatory mechanisms in the hemiskull preparations (containing the meninges) of K14-VEGFR3-Ig (K14) mice lacking the meningeal lymphatic system. We recorded the spiking activity of meningeal afferents and estimated the local mast cells population, calcitonin gene-related peptide (CGRP) and cytokine levels as well as the dural trigeminal innervation in freshly-isolated hemiskull preparations from K14-VEGFR3-Ig (K14) or wild type C57BL/6 mice (WT). Spiking activity data have been confirmed in an acquired model of meningeal lymphatic dysfunction (AAV-mVEGFR3(1-4)Ig induced lymphatic ablation). We found that levels of the pro-inflammatory cytokine IL12-p70 and CGRP, implicated in migraine, were reduced in the meninges of K14 mice, while the levels of the mast cell activator MCP-1 were increased. The other migraine-related pro-inflammatory cytokines (basal and stimulated), did not differ between the two genotypes. The patterns of trigeminal innervation in meninges remained unchanged and we did not observe alterations in basal or ATP-induced nociceptive firing in the meningeal afferents associated with meningeal lymphatic dysfunction. In summary, the lack of meningeal lymphatic system is associated with a new balance between pro- and anti-migraine mediators but does not directly trigger meningeal nociceptive state.


Assuntos
Peptídeo Relacionado com Gene de Calcitonina , Transtornos de Enxaqueca , Animais , Citocinas , Inflamação , Sistema Linfático , Meninges , Camundongos , Camundongos Endogâmicos C57BL , Nociceptividade
11.
Nat Immunol ; 21(11): 1315-1316, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32958929
12.
Nat Immunol ; 20(4): 407-419, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30886419

RESUMO

Tissue macrophages have an embryonic origin and can be replenished in some tissues under steady-state conditions by blood monocytes. However, little is known about the residency and properties of infiltrating monocytes after an inflammatory challenge. The meninges of the central nervous system (CNS) are populated by a dense network of macrophages that act as resident immune sentinels. Here we show that, following lymphocytic choriomeningitis virus infection, resident meningeal macrophages (MMs) acquired viral antigen and interacted directly with infiltrating cytotoxic T lymphocytes, which led to macrophage depletion. Concurrently, the meninges were infiltrated by inflammatory monocytes that engrafted the meningeal niche and remained in situ for months after viral clearance. This engraftment led to interferon-γ-dependent functional changes in the pool of MMs, including loss of bacterial and immunoregulatory sensors. Collectively, these data indicate that peripheral monocytes can engraft the meninges after an inflammatory challenge, imprinting the compartment with long-term defects in immune function.


Assuntos
Sistema Nervoso Central/imunologia , Macrófagos/imunologia , Meningite Viral/imunologia , Monócitos/imunologia , Animais , Imunidade , Inflamação/imunologia , Interferon gama/fisiologia , Meninges/imunologia , Camundongos
13.
Methods ; 158: 22-26, 2019 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-30742997

RESUMO

This protocol describes how to prepare mouse brain tissue for quantification of multiple inflammatory mediators using a multiplex bead-based immunoassay. It is important to have methods that allow quantification of multiple analytes from small amounts of tissue. Bio-Plex is a Luminex xMAP-based multiplex bead-based immunoassay technology that permits simultaneous analysis of up to 100 analytes from a single tissue sample. This assay has been used extensively to investigate analytes in plasma and serum samples as well as cultured and primary cells. Here, we describe a method for simultaneous analysis of 33 different inflammatory cytokines and chemokines from mouse brain tissue using the Bio-Plex Pro Mouse Chemokine Panel 33-Plex.


Assuntos
Bioensaio/métodos , Quimiocinas/análise , Citocinas/análise , Ensaios de Triagem em Larga Escala/métodos , Malária Cerebral/diagnóstico , Animais , Bioensaio/instrumentação , Biomarcadores/análise , Encéfalo/imunologia , Encéfalo/patologia , Quimiocinas/imunologia , Citocinas/imunologia , Modelos Animais de Doenças , Ensaio de Imunoadsorção Enzimática/instrumentação , Ensaio de Imunoadsorção Enzimática/métodos , Ensaios de Triagem em Larga Escala/instrumentação , Humanos , Malária Cerebral/imunologia , Malária Cerebral/parasitologia , Malária Cerebral/patologia , Camundongos , Microesferas , Plasmodium berghei/imunologia
14.
PLoS Pathog ; 14(10): e1007293, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-30296302

RESUMO

Human diseases of zoonotic origin are a major public health problem. Simian foamy viruses (SFVs) are complex retroviruses which are currently spilling over to humans. Replication-competent SFVs persist over the lifetime of their human hosts, without spreading to secondary hosts, suggesting the presence of efficient immune control. Accordingly, we aimed to perform an in-depth characterization of neutralizing antibodies raised by humans infected with a zoonotic SFV. We quantified the neutralizing capacity of plasma samples from 58 SFV-infected hunters against primary zoonotic gorilla and chimpanzee SFV strains, and laboratory-adapted chimpanzee SFV. The genotype of the strain infecting each hunter was identified by direct sequencing of the env gene amplified from the buffy coat with genotype-specific primers. Foamy virus vector particles (FVV) enveloped by wild-type and chimeric gorilla SFV were used to map the envelope region targeted by antibodies. Here, we showed high titers of neutralizing antibodies in the plasma of most SFV-infected individuals. Neutralizing antibodies target the dimorphic portion of the envelope protein surface domain. Epitopes recognized by neutralizing antibodies have been conserved during the cospeciation of SFV with their nonhuman primate host. Greater neutralization breadth in plasma samples of SFV-infected humans was statistically associated with smaller SFV-related hematological changes. The neutralization patterns provide evidence for persistent expression of viral proteins and a high prevalence of coinfection. In conclusion, neutralizing antibodies raised against zoonotic SFV target immunodominant and conserved epitopes located in the receptor binding domain. These properties support their potential role in restricting the spread of SFV in the human population.


Assuntos
Anticorpos Neutralizantes/sangue , Vetores de Doenças , Epitopos/imunologia , Hominidae/imunologia , Infecções por Retroviridae/transmissão , Vírus Espumoso dos Símios/isolamento & purificação , Proteínas do Envelope Viral/imunologia , Adulto , Sequência de Aminoácidos , Animais , Anticorpos Neutralizantes/imunologia , Sítios de Ligação , Gorilla gorilla/virologia , Hominidae/sangue , Hominidae/virologia , Humanos , Masculino , Pessoa de Meia-Idade , Pan troglodytes/virologia , Infecções por Retroviridae/virologia
15.
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
16.
Nat Immunol ; 19(8): 898, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29959442

RESUMO

In the version of this article initially published, in second paragraph of the second subsection of Results ('Peripheral licensing of CD4+ TH17 cells in Tbx21-/- hosts'), the figure citation ('Fig. 1c') in the sentence that begins "In addition to" was incorrect. The correct citation is 'Fig. 1d'.

17.
Trends Mol Med ; 24(6): 542-559, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29731353

RESUMO

The central nervous system (CNS) is an immunologically specialized tissue protected by a blood-brain barrier. The CNS parenchyma is enveloped by a series of overlapping membranes that are collectively referred to as the meninges. The meninges provide an additional CNS barrier, harbor a diverse array of resident immune cells, and serve as a crucial interface with the periphery. Recent studies have significantly advanced our understanding of meningeal immunity, demonstrating how a complex immune landscape influences CNS functions under steady-state and inflammatory conditions. The location and activation state of meningeal immune cells can profoundly influence CNS homeostasis and contribute to neurological disorders, but these cells are also well equipped to protect the CNS from pathogens. In this review, we discuss advances in our understanding of the meningeal immune repertoire and provide insights into how this CNS barrier operates immunologically under conditions ranging from neurocognition to inflammatory diseases.


Assuntos
Imunidade , Meninges/imunologia , Animais , Sistema Nervoso Central/imunologia , Sistema Nervoso Central/metabolismo , Suscetibilidade a Doenças , Homeostase , Humanos , Sistema Imunitário/citologia , Sistema Imunitário/imunologia , Sistema Imunitário/metabolismo , Meninges/anatomia & histologia , Meninges/irrigação sanguínea , Meningite/etiologia , Meningite/metabolismo , Meningite/patologia
18.
Nat Immunol ; 18(10): 1117-1127, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-28805812

RESUMO

The transcription factor T-bet has been associated with increased susceptibility to systemic and organ-specific autoimmunity, but the mechanism by which T-bet expression promotes neuroinflammation remains unknown. In this study, we demonstrate a cardinal role of T-bet-dependent NKp46+ innate lymphoid cells (ILCs) in the initiation of CD4+ TH17-mediated neuroinflammation. Loss of T-bet specifically in NKp46+ ILCs profoundly impaired the ability of myelin-reactive TH17 cells to invade central nervous system (CNS) tissue and protected the mice from autoimmunity. T-bet-dependent NKp46+ ILCs localized in the meninges and acted as chief coordinators of meningeal inflammation by inducing the expression of proinflammatory cytokines, chemokines and matrix metalloproteinases, which together facilitated T cell entry into CNS parenchyma. Our findings uncover a detrimental role of T-bet-dependent NKp46+ ILCs in the development of CNS autoimmune disease.


Assuntos
Imunidade Inata , Subpopulações de Linfócitos/imunologia , Subpopulações de Linfócitos/metabolismo , Receptor 1 Desencadeador da Citotoxicidade Natural/metabolismo , Células Th17/imunologia , Células Th17/metabolismo , Animais , Biomarcadores , Movimento Celular/genética , Movimento Celular/imunologia , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Modelos Animais de Doenças , Encefalomielite Autoimune Experimental/genética , Encefalomielite Autoimune Experimental/imunologia , Encefalomielite Autoimune Experimental/metabolismo , Expressão Gênica , Imunofenotipagem , Camundongos , Camundongos Knockout , Bainha de Mielina/imunologia , Receptor 1 Desencadeador da Citotoxicidade Natural/genética , Proteínas com Domínio T , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo
19.
Cell Rep ; 17(2): 413-424, 2016 10 04.
Artigo em Inglês | MEDLINE | ID: mdl-27705790

RESUMO

Several pattern-recognition receptors sense HIV-1 replication products and induce type I interferon (IFN-I) production under specific experimental conditions. However, it is thought that viral sensing and IFN induction are virtually absent in the main target cells of HIV-1 in vivo. Here, we show that activated CD4+ T cells sense HIV-1 infection through the cytosolic DNA sensor cGAS and mount a bioactive IFN-I response. Efficient induction of IFN-I by HIV-1 infection requires proviral integration and is regulated by newly expressed viral accessory proteins: Vpr potentiates, while Vpu suppresses cGAS-dependent IFN-I induction. Furthermore, Vpr also amplifies innate sensing of HIV-1 infection in Vpx-treated dendritic cells. Our results identify cGAS as mediator of an IFN-I response to HIV-1 infection in CD4+ T cells and demonstrate that this response is modulated by the viral accessory proteins Vpr and Vpu. Thus, viral innate immune evasion is incomplete in the main target cells of HIV-1.


Assuntos
Infecções por HIV/imunologia , Proteínas do Vírus da Imunodeficiência Humana/genética , Interferon Tipo I/imunologia , Nucleotidiltransferases/genética , Proteínas Virais Reguladoras e Acessórias/genética , Produtos do Gene vpr do Vírus da Imunodeficiência Humana/genética , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/metabolismo , Linfócitos T CD4-Positivos/virologia , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Células Dendríticas/virologia , Células HEK293 , Infecções por HIV/genética , Infecções por HIV/virologia , HIV-1/imunologia , HIV-1/patogenicidade , Proteínas do Vírus da Imunodeficiência Humana/imunologia , Humanos , Imunidade Inata/genética , Interferon Tipo I/genética , Lentivirus/genética , Nucleotidiltransferases/imunologia , Proteínas Virais Reguladoras e Acessórias/imunologia , Produtos do Gene vpr do Vírus da Imunodeficiência Humana/imunologia
20.
Virology ; 496: 219-226, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27348053

RESUMO

The majority of currently identified simian foamy virus (SFV)-infected Cameroonian and Gabonese individuals harbor SFV from the gorilla lineage. We constructed an indicator cell line for the quantification of gorilla SFVs, in which the U3 sequence of a gorilla SFV directs the expression of the ß-galactosidase protein. The gorilla foamy virus activated ß-galactosidase (GFAB) cells efficiently quantified two zoonotic primary gorilla isolates and SFVs from three chimpanzee subspecies. Primary gorilla SFVs replicated more slowly and at lower levels than primary chimpanzee SFVs. Analysis of previously described motifs of Tas proteins and U3 LTRs involved in viral gene synthesis revealed conservation of such motifs in Tas proteins from gorilla and chimpanzee SFVs, but little sequence homology in the LTR regions previously shown to interact with viral and cellular factors.


Assuntos
Vírus Espumoso dos Símios/fisiologia , Sequências Repetidas Terminais , Ativação Transcricional , Sequência de Aminoácidos , Animais , Sequência de Bases , Sítios de Ligação , Linhagem Celular , Células Cultivadas , Cricetinae , Expressão Gênica , Genes Reporter , Vetores Genéticos , Gorilla gorilla , Motivos de Nucleotídeos , Pan troglodytes , Ligação Proteica , Especificidade da Espécie
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA