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

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Nat Immunol ; 24(4): 714-728, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36928414

RESUMEN

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.


Asunto(s)
Células Dendríticas , Interferón Tipo I , Animales , Ratones , Interferón Tipo I/metabolismo , Perfilación de la Expresión Génica , Fenotipo , Transcriptoma
2.
Immunity ; 56(4): 744-746, 2023 04 11.
Artículo en Inglés | MEDLINE | ID: mdl-37044064

RESUMEN

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.


Asunto(s)
Proteínas de Caenorhabditis elegans , Animales , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/genética , Receptores Citoplasmáticos y Nucleares/genética , Receptores Citoplasmáticos y Nucleares/metabolismo , Regulación de la Expresión Génica , Transducción de Señal
3.
Nat Immunol ; 20(4): 407-419, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30886419

RESUMEN

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.


Asunto(s)
Sistema Nervioso Central/inmunología , Macrófagos/inmunología , Meningitis Viral/inmunología , Monocitos/inmunología , Animales , Inmunidad , Inflamación/inmunología , Interferón gamma/fisiología , Meninges/inmunología , Ratones
4.
Immunity ; 55(11): 2103-2117.e10, 2022 11 08.
Artículo en Inglés | MEDLINE | ID: mdl-36323311

RESUMEN

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.


Asunto(s)
COVID-19 , Coriomeningitis Linfocítica , Animales , Ratones , Lipopolisacáridos , Ratones Endogámicos C57BL , SARS-CoV-2 , Virus de la Coriomeningitis Linfocítica/fisiología , Macrófagos , Meninges
5.
Nat Immunol ; 19(8): 898, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29959442

RESUMEN

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'.

6.
Nat Immunol ; 18(10): 1117-1127, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28805812

RESUMEN

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.


Asunto(s)
Inmunidad Innata , Subgrupos Linfocitarios/inmunología , Subgrupos Linfocitarios/metabolismo , Receptor 1 Gatillante de la Citotoxidad Natural/metabolismo , Células Th17/inmunología , Células Th17/metabolismo , Animales , Biomarcadores , Movimiento Celular/genética , Movimiento Celular/inmunología , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Modelos Animales de Enfermedad , Encefalomielitis Autoinmune Experimental/genética , Encefalomielitis Autoinmune Experimental/inmunología , Encefalomielitis Autoinmune Experimental/metabolismo , Expresión Génica , Inmunofenotipificación , Ratones , Ratones Noqueados , Vaina de Mielina/inmunología , Receptor 1 Gatillante de la Citotoxidad Natural/genética , Proteínas de Dominio T Box , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo
7.
Nat Immunol ; 21(11): 1315-1316, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32958929
8.
Trends Immunol ; 45(5): 346-357, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38632001

RESUMEN

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).


Asunto(s)
Envejecimiento , Enfermedad de Alzheimer , Encéfalo , Macrófagos , Animales , Humanos , Envejecimiento/inmunología , Enfermedad de Alzheimer/inmunología , Encéfalo/inmunología , Encéfalo/patología , Infecciones/inmunología , Macrófagos/inmunología
9.
Glia ; 72(3): 475-503, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-37909340

RESUMEN

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.


Asunto(s)
Lesiones Encefálicas , Nacimiento Prematuro , Lactante , Embarazo , Animales , Femenino , Recién Nacido , Humanos , Recien Nacido Prematuro , Neuroglía , Encéfalo
10.
Eur J Immunol ; 53(9): e2250267, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37402972

RESUMEN

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.


Asunto(s)
Infecciones del Sistema Nervioso Central , Meninges , Adulto , Recién Nacido , Humanos , Encéfalo , Streptococcus pneumoniae
11.
PLoS Pathog ; 14(10): e1007293, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30296302

RESUMEN

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.


Asunto(s)
Anticuerpos Neutralizantes/sangre , Vectores de Enfermedades , Epítopos/inmunología , Hominidae/inmunología , Infecciones por Retroviridae/transmisión , Virus Espumoso de los Simios/aislamiento & purificación , Proteínas del Envoltorio Viral/inmunología , Adulto , Secuencia de Aminoácidos , Animales , Anticuerpos Neutralizantes/inmunología , Sitios de Unión , Gorilla gorilla/virología , Hominidae/sangre , Hominidae/virología , Humanos , Masculino , Persona de Mediana Edad , Pan troglodytes/virología , Infecciones por Retroviridae/virología
12.
Methods ; 158: 22-26, 2019 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-30742997

RESUMEN

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.


Asunto(s)
Bioensayo/métodos , Quimiocinas/análisis , Citocinas/análisis , Ensayos Analíticos de Alto Rendimiento/métodos , Malaria Cerebral/diagnóstico , Animales , Bioensayo/instrumentación , Biomarcadores/análisis , Encéfalo/inmunología , Encéfalo/patología , Quimiocinas/inmunología , Citocinas/inmunología , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática/instrumentación , Ensayo de Inmunoadsorción Enzimática/métodos , Ensayos Analíticos de Alto Rendimiento/instrumentación , Humanos , Malaria Cerebral/inmunología , Malaria Cerebral/parasitología , Malaria Cerebral/patología , Ratones , Microesferas , Plasmodium berghei/inmunología
13.
J Virol ; 89(23): 12118-30, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26401039

RESUMEN

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.


Asunto(s)
Regulación de la Expresión Génica/inmunología , VIH-1/inmunología , Linfocitos T/virología , Factor de Necrosis Tumoral alfa/inmunología , Productos del Gen vpr del Virus de la Inmunodeficiencia Humana/inmunología , Línea Celular , Proteínas de Unión al ADN/metabolismo , Humanos , Quinasas Quinasa Quinasa PAM/metabolismo , FN-kappa B/metabolismo , Linfocitos T/inmunología , Linfocitos T/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
14.
J Virol ; 89(24): 12480-91, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26446599

RESUMEN

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.


Asunto(s)
Productos del Gen env/genética , Filogenia , Recombinación Genética , Infecciones por Retroviridae/genética , Virus Espumoso de los Simios/genética , Animales , Enfermedades del Simio Antropoideo/epidemiología , Enfermedades del Simio Antropoideo/genética , Enfermedades del Simio Antropoideo/virología , Camerún , Gabón , Gorilla gorilla , Humanos , Pan troglodytes , Infecciones por Retroviridae/epidemiología , Infecciones por Retroviridae/veterinaria
15.
J Virol ; 88(22): 13429-35, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25210185

RESUMEN

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.


Asunto(s)
Leucocitos Mononucleares/virología , Subgrupos Linfocitarios/virología , Infecciones por Retroviridae/virología , Virus Espumoso de los Simios/fisiología , Tropismo Viral , Adulto , Anciano , Animales , ADN Viral/análisis , ADN Viral/química , ADN Viral/genética , Humanos , Masculino , Persona de Mediana Edad , Datos de Secuencia Molecular , Reacción en Cadena en Tiempo Real de la Polimerasa , Análisis de Secuencia de ADN , Carga Viral , Adulto Joven
16.
J Inflamm (Lond) ; 21(1): 3, 2024 Jan 30.
Artículo en Inglés | MEDLINE | ID: mdl-38291415

RESUMEN

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.

17.
J Virol ; 86(2): 909-18, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22090096

RESUMEN

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.


Asunto(s)
Células Dendríticas/inmunología , Inmunidad Innata , Leucocitos Mononucleares/inmunología , Infecciones por Retroviridae/inmunología , Spumavirus/inmunología , Animales , Línea Celular , Células Cultivadas , Células Dendríticas/virología , Humanos , Interferón Tipo I/genética , Interferón Tipo I/inmunología , Leucocitos Mononucleares/virología , Infecciones por Retroviridae/genética , Infecciones por Retroviridae/virología , Spumavirus/fisiología , Receptor Toll-Like 7/genética , Receptor Toll-Like 7/inmunología
18.
J Virol ; 86(24): 13350-9, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23015714

RESUMEN

Simian foamy viruses (SFVs) are retroviruses that are widespread among nonhuman primates (NHPs). SFVs actively replicate in their oral cavity and can be transmitted to humans after NHP bites, giving rise to a persistent infection even decades after primary infection. Very few data on the genetic structure of such SFVs found in humans are available. In the framework of ongoing studies searching for SFV-infected humans in south Cameroon rainforest villages, we studied 38 SFV-infected hunters whose times of infection had presumably been determined. By long-term cocultures of peripheral blood mononuclear cells with BHK-21 cells, we isolated five new SFV strains and obtained complete genomes of SFV strains from chimpanzee (Pan troglodytes troglodytes; strains BAD327 and AG15), monkey (Cercopithecus nictitans; strain AG16), and gorilla (Gorilla gorilla; strains BAK74 and BAD468). These zoonotic strains share a very high degree of similarity with their NHP counterparts and have a high degree of conservation of the genetic elements important for viral replication. Interestingly, analysis of FV DNA sequences obtained before cultivation revealed variants with deletions in both the U3 region and tas that may correlate with in vivo chronicity in humans. Genomic changes in bet (a premature stop codon) and gag were also observed. To determine if such changes were specific to zoonotic strains, we studied local SFV-infected chimpanzees and found the same genomic changes. Our study reveals that natural polymorphism of SFV strains does exist at both the intersubspecies level (gag, bet) and the intrasubspecies (U3, tas) levels but does not seem to reflect a viral adaptation specific to zoonotic SFV strains.


Asunto(s)
Genes Virales , Virus Espumoso de los Simios/genética , Animales , Secuencia de Bases , Camerún , ADN Viral/genética , Humanos , Datos de Secuencia Molecular , Filogenia , Polimorfismo Genético , Primates/virología , Homología de Secuencia de Ácido Nucleico , Virus Espumoso de los Simios/clasificación , Virus Espumoso de los Simios/fisiología , Replicación Viral
19.
PLoS Pathog ; 7(10): e1002306, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22046126

RESUMEN

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.


Asunto(s)
Enfermedades del Simio Antropoideo/transmisión , Hominidae/virología , Infecciones por Retroviridae/transmisión , Virus Espumoso de los Simios/aislamiento & purificación , Adolescente , Adulto , África Central/epidemiología , Anciano , Anciano de 80 o más Años , Animales , Anticuerpos Antivirales , Enfermedades del Simio Antropoideo/virología , Mordeduras y Picaduras , Población Negra/etnología , Niño , Preescolar , ADN Viral/análisis , Femenino , Humanos , Masculino , Persona de Mediana Edad , Infecciones por Retroviridae/etnología , Infecciones por Retroviridae/virología , Análisis de Secuencia de ADN , Estudios Seroepidemiológicos , Virus Espumoso de los Simios/genética , Virus Espumoso de los Simios/inmunología , Adulto Joven , Zoonosis/epidemiología , Zoonosis/virología
20.
STAR Protoc ; 4(1): 102119, 2023 03 17.
Artículo en Inglés | MEDLINE | ID: mdl-36853673

RESUMEN

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.


Asunto(s)
Sistema Nervioso Central , Meninges , Animales , Ratones , Citometría de Flujo , Inmunohistoquímica , Meninges/diagnóstico por imagen , Cabeza
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA