Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
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.
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
3.
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
4.
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
5.
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
6.
J Virol ; 89(24): 12480-91, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26446599

RESUMO

UNLABELLED: Simian foamy virus (SFV) is a ubiquitous retrovirus in nonhuman primates (NHPs) that can be transmitted to humans, mostly through severe bites. In the past few years, our laboratory has identified more than 50 hunters from central Africa infected with zoonotic SFVs. Analysis of the complete sequences of five SFVs obtained from these individuals revealed that env was the most variable gene. Furthermore, recombinant SFV strains, some of which involve sequences in the env gene, were recently identified. Here, we investigated the variability of the env genes of zoonotic SFV strains and searched for possible recombinants. We sequenced the complete env gene or its surface glycoprotein region (SU) from DNA amplified from the blood of (i) a series of 40 individuals from Cameroon or Gabon infected with a gorilla or chimpanzee foamy virus (FV) strain and (ii) 1 gorilla and 3 infected chimpanzees living in the same areas as these hunters. Phylogenetic analyses revealed the existence of two env variants among both the gorilla and chimpanzee FV strains that were present in zoonotic and NHP strains. These variants differ greatly (>30% variability) in a 753-bp-long region located in the receptor-binding domain of SU, whereas the rest of the gene is very conserved. Although the organizations of the Env protein sequences are similar, the potential glycosylation patterns differ between variants. Analysis of recombination suggests that the variants emerged through recombination between different strains, although all parental strains could not be identified. IMPORTANCE: SFV infection in humans is a great example of a zoonotic retroviral infection that has not spread among human populations, in contrast to human immunodeficiency viruses (HIVs) and human T-lymphotropic viruses (HTLVs). Recombination was a major mechanism leading to the emergence of HIV. Here, we show that two SFV molecular envelope gene variants circulate among ape populations in Central Africa and that both can be transmitted to humans. These variants differ greatly in the SU region that corresponds to the part of the Env protein in contact with the environment. These variants may have emerged through recombination between SFV strains infecting different NHP species.


Assuntos
Produtos do Gene env/genética , Filogenia , Recombinação Genética , Infecções por Retroviridae/genética , Vírus Espumoso dos Símios/genética , Animais , Doenças dos Símios Antropoides/epidemiologia , Doenças dos Símios Antropoides/genética , Doenças dos Símios Antropoides/virologia , Camarões , Gabão , Gorilla gorilla , Humanos , Pan troglodytes , Infecções por Retroviridae/epidemiologia , Infecções por Retroviridae/veterinária
7.
J Virol ; 89(23): 12118-30, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26401039

RESUMO

UNLABELLED: The HIV-1 accessory protein Vpr displays different activities potentially impacting viral replication, including the arrest of the cell cycle in the G2 phase and the stimulation of apoptosis and DNA damage response pathways. Vpr also modulates cytokine production by infected cells, but this property remains partly characterized. Here, we investigated the effect of Vpr on the production of the proinflammatory cytokine tumor necrosis factor (TNF). We report that Vpr significantly increases TNF secretion by infected lymphocytes. De novo production of Vpr is required for this effect. Vpr mutants known to be defective for G2 cell cycle arrest induce lower levels of TNF secretion, suggesting a link between these two functions. Silencing experiments and the use of chemical inhibitors further implicated the cellular proteins DDB1 and TAK1 in this activity of Vpr. TNF secreted by HIV-1-infected cells triggers NF-κB activity in bystander cells and allows viral reactivation in a model of latently infected cells. Thus, the stimulation of the proinflammatory pathway by Vpr may impact HIV-1 replication in vivo. IMPORTANCE: The role of the HIV-1 accessory protein Vpr remains only partially characterized. This protein is important for viral pathogenesis in infected individuals but is dispensable for viral replication in most cell culture systems. Some of the functions described for Vpr remain controversial. In particular, it remains unclear whether Vpr promotes or instead prevents proinflammatory and antiviral immune responses. In this report, we show that Vpr promotes the release of TNF, a proinflammatory cytokine associated with rapid disease progression. Using Vpr mutants or inhibiting selected cellular genes, we show that the cellular proteins DDB1 and TAK1 are involved in the release of TNF by HIV-infected cells. This report provides novel insights into how Vpr manipulates TNF production and helps clarify the role of Vpr in innate immune responses and inflammation.


Assuntos
Regulação da Expressão Gênica/imunologia , HIV-1/imunologia , Linfócitos T/virologia , Fator de Necrose Tumoral alfa/imunologia , Produtos do Gene vpr do Vírus da Imunodeficiência Humana/imunologia , Linhagem Celular , Proteínas de Ligação a DNA/metabolismo , Humanos , MAP Quinase Quinase Quinases/metabolismo , NF-kappa B/metabolismo , Linfócitos T/imunologia , Linfócitos T/metabolismo , Fator de Necrose Tumoral alfa/metabolismo
8.
J Leukoc Biol ; 98(3): 319-32, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26162402

RESUMO

Monocytes and macrophages are a diverse population of innate immune cells that play a critical role in homeostasis and inflammation. These cells are surveillant by nature and closely monitor the vasculature and surrounding tissue during states of health and disease. Given their abundance and strategic positioning throughout the body, myeloid cells are among the first responders to any inflammatory challenge and are active participants in most immune-mediated diseases. Recent studies have shed new light on myeloid cell dynamics and function by use of an imaging technique referred to as intravital microscopy (IVM). This powerful approach allows researchers to gain real-time insights into monocytes and macrophages performing homeostatic and inflammatory tasks in living tissues. In this review, we will present a contemporary synopsis of how intravital microscopy has revolutionized our understanding of myeloid cell contributions to vascular maintenance, microbial defense, autoimmunity, tumorigenesis, and acute/chronic inflammatory diseases.


Assuntos
Imageamento Tridimensional/métodos , Macrófagos/citologia , Monócitos/citologia , Animais , Vasos Sanguíneos/citologia , Comunicação Celular , Humanos , Modelos Imunológicos
10.
Curr Opin Virol ; 10: 47-55, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25698621

RESUMO

Most viral pathogens that have emerged in humans have originated from various animal species. Emergence is a multistep process involving an initial spill-over of the infectious agent into single individuals and its subsequent dissemination into the human population. Similar to simian immunodeficiency viruses and simian T lymphotropic viruses, simian foamy viruses (SFV) are retroviruses that are widespread among non-human primates and can be transmitted to humans, giving rise to a persistent infection, which seems to be controlled in the case of SFV. In this review, we present current data on the discovery, cross-species transmission, and molecular evolution of SFV in human populations initially infected and thus at risk for zoonotic emergence.


Assuntos
Infecções por Retroviridae/imunologia , Infecções por Retroviridae/transmissão , Vírus Espumoso dos Símios/genética , Vírus Espumoso dos Símios/imunologia , Animais , DNA Viral/genética , Evolução Molecular , Genoma Viral , Humanos , Doenças dos Primatas/virologia , Primatas , Infecções por Retroviridae/veterinária , Infecções por Retroviridae/virologia , Vírus Espumoso dos Símios/isolamento & purificação , Vírus Espumoso dos Símios/fisiologia , Vírus Espumoso dos Símios/ultraestrutura , Proteínas Estruturais Virais/genética , Proteínas Estruturais Virais/metabolismo , Tropismo Viral , Zoonoses
11.
J Virol ; 88(22): 13429-35, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25210185

RESUMO

UNLABELLED: Simian foamy viruses (SFV) are retroviruses that are widespread among nonhuman primates. SFV can be transmitted to humans, giving rise to a persistent infection. Only a few data are available concerning the distribution of SFV in human blood cells. Here we purified blood mononuclear cell subsets from 11 individuals infected with a Gorilla gorilla SFV strain and quantified SFV DNA levels by quantitative PCR. SFV DNA was detected in the majority of the CD8(+), CD4(+), and CD19(+) lymphocyte samples and rarely in CD14(+) monocyte and CD56(+) NK lymphocyte samples. The median (interquartile range [IQR]) SFV DNA counts were 16.0 (11.0 to 49.8), 11.3 (5.9 to 28.3), and 17.2 (2.0 to 25.2) copies/10(5) cells in CD8(+) T lymphocytes, CD4(+) T lymphocytes, and CD19(+) B lymphocytes, respectively. In the CD4 compartment, SFV DNA was detected in both memory and naive CD4(+) T lymphocytes. SFV DNA levels in CD4(+) T cells were positively correlated with the duration of the infection. Our study shows with a quantitative method that CD8(+), CD4(+), and B lymphocytes are major cellular targets of SFV in the blood of infected humans. IMPORTANCE: Investigation of SFV infections in humans is important due to the origin of human immunodeficiency viruses (HIV) and human T cell lymphotropic viruses (HTLV) from cross-species transmission of their simian counterparts to humans. Surprisingly little is known about many aspects of the biology of SFV in infected humans, including quantitative data concerning the cellular targets of SFV in vivo. Here we show that the distribution of SFV DNA among the different leukocyte populations is not homogeneous and that viral load in CD4(+) T lymphocytes is correlated with the duration of infection. These new data will help in understanding the biology of retroviral infections in humans and can be useful in the growing field of SFV-based gene therapy.


Assuntos
Leucócitos Mononucleares/virologia , Subpopulações de Linfócitos/virologia , Infecções por Retroviridae/virologia , Vírus Espumoso dos Símios/fisiologia , Tropismo Viral , Adulto , Idoso , Animais , DNA Viral/análise , DNA Viral/química , DNA Viral/genética , Humanos , Masculino , Pessoa de Meia-Idade , Dados de Sequência Molecular , Reação em Cadeia da Polimerase em Tempo Real , Análise de Sequência de DNA , Carga Viral , Adulto Jovem
12.
J Exp Med ; 210(13): 2813-21, 2013 Dec 16.
Artigo em Inglês | MEDLINE | ID: mdl-24277152

RESUMO

The neutralizing activity of anti-HIV-1 antibodies is typically measured in assays where cell-free virions enter reporter cell lines. However, HIV-1 cell to cell transmission is a major mechanism of viral spread, and the effect of the recently described broadly neutralizing antibodies (bNAbs) on this mode of transmission remains unknown. Here we identify a subset of bNAbs that inhibit both cell-free and cell-mediated infection in primary CD4(+) lymphocytes. These antibodies target either the CD4-binding site (NIH45-46 and 3BNC60) or the glycan/V3 loop (10-1074 and PGT121) on HIV-1 gp120 and act at low concentrations by inhibiting multiple steps of viral cell to cell transmission. These antibodies accumulate at virological synapses and impair the clustering and fusion of infected and target cells and the transfer of viral material to uninfected T cells. In addition, they block viral cell to cell transmission to plasmacytoid DCs and thereby interfere with type-I IFN production. Thus, only a subset of bNAbs can efficiently prevent HIV-1 cell to cell transmission, and this property should be considered an important characteristic defining antibody potency for therapeutic or prophylactic antiviral strategies.


Assuntos
Anticorpos Neutralizantes/imunologia , Anticorpos Anti-HIV/imunologia , Infecções por HIV/tratamento farmacológico , Infecções por HIV/imunologia , HIV/fisiologia , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/virologia , Técnicas de Cocultura , Células Dendríticas/citologia , Células Dendríticas/imunologia , Células HEK293 , Células HeLa , Humanos , Concentração Inibidora 50 , Microscopia de Fluorescência , Fatores de Tempo , Vírion/fisiologia
13.
PLoS One ; 8(10): e77072, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24116202

RESUMO

Simian foamy viruses (SFV) are widespread retroviruses among non-human primates (NHP). SFV actively replicate in the oral cavity and can be transmitted to humans through NHP bites, giving rise to a persistent infection. We aimed at studying the natural history of SFV infection in human. We have analyzed viral load and gene expression in 14 hunters from Cameroon previously shown to be infected with a gorilla SFV strain. Viral DNA could be detected by quantitative polymerase chain reaction (q-PCR) targeting the pol-in region, in most samples of peripheral blood mononuclear cells (PBMCs) (7.1 ± 6.0 SFV DNA copies/105 PBMCs) and saliva (2.4 ± 4.3 SFV DNA copies/105 cells) derived from the hunters. However, quantitative real-time reverse-transcription polymerase chain reaction (RT)-qPCR revealed the absence of SFV viral gene expression in both PBMCs and saliva, suggesting that SFV was latent in the human samples. Our study demonstrates that a latent infection can occur in humans and persist for years, both in PBMCs and saliva. Such a scenario may contribute to the putative lack of secondary human-to-human transmissions of SFV.


Assuntos
Infecções por Retroviridae/sangue , Infecções por Retroviridae/virologia , Saliva/virologia , Vírus Espumoso dos Símios/fisiologia , Latência Viral , Adulto , Animais , Sangue/virologia , DNA Viral/isolamento & purificação , Humanos , Leucócitos Mononucleares/virologia , Pessoa de Meia-Idade , Infecções por Retroviridae/diagnóstico , Vírus Espumoso dos Símios/isolamento & purificação , Adulto Jovem
14.
BMC Genomics ; 13: 677, 2012 Nov 28.
Artigo em Inglês | MEDLINE | ID: mdl-23190615

RESUMO

BACKGROUND: HIV-associated dementia (HAD) is the most common dementia type in young adults less than 40 years of age. Although the neurotoxins, oxidative/metabolic stress and impaired activity of neurotrophic factors are believed to be underlying reasons for the development of HAD, the genomic basis, which ultimately defines the virus-host interaction and leads to neurologic manifestation of HIV disease is lacking. Therefore, identifying HIV fingerprints on the host gene machinery and its regulation by microRNA holds a great promise and potential for improving our understanding of HAD pathogenesis, its diagnosis and therapy. RESULTS: A parallel profiling of mRNA and miRNA of the frontal cortex autopsies from HIV positive patients with and without dementia was performed using Illumina Human-6 BeadChip and Affymetrix version 1.0 miRNA array, respectively. The gene ontology and pathway analysis of the two data sets showed high concordance between miRNA and mRNAs, revealing significant interference with the host axon guidance and its downstream signalling pathways in HAD brains. Moreover, the differentially expressed (DE) miRNAs identified in this study, in particular miR-137, 153 and 218, based on which most correlations were built cumulatively targeted neurodegeneration related pathways, implying their future potential in diagnosis, prognosis and possible therapies for HIV-mediated and possibly other neurodegenerative diseases. Furthermore, this relationship between DE miRNAs and DE mRNAs was also reflected in correlation analysis using Bayesian networks by splitting-averaging strategy (SA-BNs), which revealed 195 statistically significant correlated miRNA-mRNA pairs according to Pearson's correlation test (P<0.05). CONCLUSIONS: Our study provides the first evidence on unambiguous support for intrinsic functional relationship between mRNA and miRNA in the context of HIV-mediated neurodegeneration, which shows that neurologic manifestation in HIV patients possibly occurs through the interference with the host axon guidance and its downstream signalling pathways. These data provide an excellent avenue for the development of new generation of diagnostic/prognostic biomarkers and therapeutic intervention strategies for HIV-associated neurodegeneration.


Assuntos
Complexo AIDS Demência/genética , Axônios/patologia , Lobo Frontal/metabolismo , Infecções por HIV/genética , HIV/fisiologia , MicroRNAs/metabolismo , Transdução de Sinais/genética , Complexo AIDS Demência/diagnóstico , Complexo AIDS Demência/patologia , Adulto , Diagnóstico Precoce , Lobo Frontal/patologia , Lobo Frontal/virologia , Perfilação da Expressão Gênica , Genoma Humano/genética , Genômica , Infecções por HIV/complicações , Infecções por HIV/patologia , Humanos , Masculino , MicroRNAs/genética , Pessoa de Meia-Idade , Análise de Sequência com Séries de Oligonucleotídeos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo
15.
J Virol ; 86(2): 909-18, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22090096

RESUMO

Foamy viruses (FV) are nonpathogenic retroviruses that have cospeciated with primates for millions of years. FV can be transmitted through severe bites from monkeys to humans. Viral loads remain generally low in infected humans, and no secondary transmission has been reported. Very little is known about the ability of FV to trigger an innate immune response in human cells. A few previous reports suggested that FV do not induce type I interferon (IFN) in nonhematopoietic cells. Here, we examined how human hematopoietic cells sense FV particles and FV-infected cells. We show that peripheral blood mononuclear cells (PBMCs), plasmacytoid dendritic cells (pDCs), and the pDC-like cell line Gen2.2 detect FV, produce high levels of type I IFN, and express the IFN-stimulated gene MxA. Fewer than 20 FV-infected cells are sufficient to trigger an IFN response. Both prototypic and primary viruses stimulated IFN release. Donor cells expressing a replication-defective virus, carrying a mutated reverse transcriptase, induced IFN production by target cells as potently as wild-type virus. In contrast, an FV strain with env deleted, which does not produce viral particles, was inactive. IFN production was blocked by an inhibitor of endosomal acidification (bafilomycin A1) and by an endosomal Toll-like receptor (TLR) antagonist (A151). Silencing experiments in Gen2.2 further demonstrated that TLR7 is involved in FV recognition. Therefore, FV are potent inducers of type I IFN by pDCs and by PBMCs. This previously underestimated activation of the innate immune response may be involved in the control of viral replication in humans.


Assuntos
Células Dendríticas/imunologia , Imunidade Inata , Leucócitos Mononucleares/imunologia , Infecções por Retroviridae/imunologia , Spumavirus/imunologia , Animais , Linhagem Celular , Células Cultivadas , Células Dendríticas/virologia , Humanos , Interferon Tipo I/genética , Interferon Tipo I/imunologia , Leucócitos Mononucleares/virologia , Infecções por Retroviridae/genética , Infecções por Retroviridae/virologia , Spumavirus/fisiologia , Receptor 7 Toll-Like/genética , Receptor 7 Toll-Like/imunologia
16.
PLoS Pathog ; 7(10): e1002306, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22046126

RESUMO

Human infection by simian foamy viruses (SFV) can be acquired by persons occupationally exposed to non-human primates (NHP) or in natural settings. This study aimed at getting better knowledge on SFV transmission dynamics, risk factors for such a zoonotic infection and, searching for intra-familial dissemination and the level of peripheral blood (pro)viral loads in infected individuals. We studied 1,321 people from the general adult population (mean age 49 yrs, 640 women and 681 men) and 198 individuals, mostly men, all of whom had encountered a NHP with a resulting bite or scratch. All of these, either Pygmies (436) or Bantus (1085) live in villages in South Cameroon. A specific SFV Western blot was used and two nested PCRs (polymerase, and LTR) were done on all the positive/borderline samples by serology. In the general population, 2/1,321 (0.2%) persons were found to be infected. In the second group, 37/198 (18.6%) persons were SFV positive. They were mostly infected by apes (37/39) FV (mainly gorilla). Infection by monkey FV was less frequent (2/39). The viral origin of the amplified sequences matched with the history reported by the hunters, most of which (83%) are aged 20 to 40 years and acquired the infection during the last twenty years. The (pro)viral load in 33 individuals infected by a gorilla FV was quite low (<1 to 145 copies per 10(5) cells) in the peripheral blood leucocytes. Of the 30 wives and 12 children from families of FV infected persons, only one woman was seropositive in WB without subsequent viral DNA amplification. We demonstrate a high level of recent transmission of SFVs to humans in natural settings specifically following severe gorilla bites during hunting activities. The virus was found to persist over several years, with low SFV loads in infected persons. Secondary transmission remains an open question.


Assuntos
Doenças dos Símios Antropoides/transmissão , Hominidae/virologia , Infecções por Retroviridae/transmissão , Vírus Espumoso dos Símios/isolamento & purificação , Adolescente , Adulto , África Central/epidemiologia , Idoso , Idoso de 80 Anos ou mais , Animais , Anticorpos Antivirais , Doenças dos Símios Antropoides/virologia , Mordeduras e Picadas , População Negra/etnologia , Criança , Pré-Escolar , DNA Viral/análise , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Infecções por Retroviridae/etnologia , Infecções por Retroviridae/virologia , Análise de Sequência de DNA , Estudos Soroepidemiológicos , Vírus Espumoso dos Símios/genética , Vírus Espumoso dos Símios/imunologia , Adulto Jovem , Zoonoses/epidemiologia , Zoonoses/virologia
17.
BMC Infect Dis ; 9: 192, 2009 Dec 02.
Artigo em Inglês | MEDLINE | ID: mdl-19951441

RESUMO

BACKGROUND: HIV-1 penetrates the central nervous system, which is vital for HIV-associated dementia (HAD). But the role of cellular infiltration and activation together with HIV in the development of HAD is poorly understood. METHODS: To study activation and infiltration patterns of macrophages, CD8+ T cells in relation to HIV in diverse CNS areas of patients with and without dementia. 46 brain regions from two rapidly progressing severely demented patients and 53 regions from 4 HIV+ non-dementia patients were analyzed. Macrophage and CD8+ T cell infiltration of the CNS in relation to HIV was assessed using immuno-histochemical analysis with anti-HIV (P24), anti-CD8 and anti-CD68, anti-S-100A8 and granzyme B antibodies (cellular activation). Statistical analysis was performed with SPSS 12.0 with Student's t test and ANOVA. RESULTS: Overall, the patterns of infiltration of macrophages and CD8+ T cells were indiscernible between patients with and without dementia, but the co-localization of macrophages and CD8+ T cells along with HIV P24 antigen in the deeper midline and mesial temporal structures of the brain segregated the two groups. This predilection of infected macrophages and CD8+ T cells to the middle part of the brain was unique to both HAD patients, along with unique nature of provirus gag gene sequences derived from macrophages in the midline and mesial temporal structures. CONCLUSION: Strong predilection of infected macrophages and CD8+ T cells was typical of the deeper midline and mesial temporal structures uniquely in HAD patients, which has some influence on neurocognitive impairment during HIV infection.


Assuntos
Complexo AIDS Demência/imunologia , Encéfalo/patologia , Linfócitos T CD8-Positivos/virologia , Macrófagos/virologia , Complexo AIDS Demência/patologia , Adulto , Sequência de Bases , Encéfalo/imunologia , Encéfalo/virologia , Linfócitos T CD8-Positivos/imunologia , Pré-Escolar , Feminino , Proteína do Núcleo p24 do HIV/metabolismo , HIV-1/genética , HIV-1/imunologia , Humanos , Ativação Linfocitária/imunologia , Macrófagos/imunologia , Masculino , Pessoa de Meia-Idade , Dados de Sequência Molecular , Filogenia , Reação em Cadeia da Polimerase
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA