Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Nature ; 620(7973): 374-380, 2023 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-37532932

RESUMEN

Low-grade inflammation is a hallmark of old age and a central driver of ageing-associated impairment and disease1. Multiple factors can contribute to ageing-associated inflammation2; however, the molecular pathways that transduce aberrant inflammatory signalling and their impact in natural ageing remain unclear. Here we show that the cGAS-STING signalling pathway, which mediates immune sensing of DNA3, is a critical driver of chronic inflammation and functional decline during ageing. Blockade of STING suppresses the inflammatory phenotypes of senescent human cells and tissues, attenuates ageing-related inflammation in multiple peripheral organs and the brain in mice, and leads to an improvement in tissue function. Focusing on the ageing brain, we reveal that activation of STING triggers reactive microglial transcriptional states, neurodegeneration and cognitive decline. Cytosolic DNA released from perturbed mitochondria elicits cGAS activity in old microglia, defining a mechanism by which cGAS-STING signalling is engaged in the ageing brain. Single-nucleus RNA-sequencing analysis of microglia and hippocampi of a cGAS gain-of-function mouse model demonstrates that engagement of cGAS in microglia is sufficient to direct ageing-associated transcriptional microglial states leading to bystander cell inflammation, neurotoxicity and impaired memory capacity. Our findings establish the cGAS-STING pathway as a driver of ageing-related inflammation in peripheral organs and the brain, and reveal blockade of cGAS-STING signalling as a potential strategy to halt neurodegenerative processes during old age.


Asunto(s)
Envejecimiento , Encéfalo , Disfunción Cognitiva , Inflamación , Proteínas de la Membrana , Enfermedades Neurodegenerativas , Nucleotidiltransferasas , Animales , Humanos , Ratones , Envejecimiento/metabolismo , Envejecimiento/patología , Encéfalo/metabolismo , Encéfalo/patología , Efecto Espectador , Disfunción Cognitiva/metabolismo , Disfunción Cognitiva/patología , ADN/inmunología , Inflamación/enzimología , Inflamación/metabolismo , Proteínas de la Membrana/metabolismo , Trastornos de la Memoria/enzimología , Trastornos de la Memoria/metabolismo , Microglía/metabolismo , Mitocondrias/metabolismo , Enfermedades Neurodegenerativas/enzimología , Enfermedades Neurodegenerativas/metabolismo , Nucleotidiltransferasas/metabolismo , Especificidad de Órganos , Transducción de Señal , Hipocampo/metabolismo , Hipocampo/patología
2.
J Cell Sci ; 137(1)2024 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-38197775

RESUMEN

The septation initiation network (SIN) is a conserved signal transduction network, which is important for cytokinesis in Schizosaccharomyces pombe. The SIN component Etd1p is required for association of some SIN proteins with the spindle pole body (SPB) during anaphase and for contractile ring formation. We show that tethering of Cdc7p or Sid1p to the SIN scaffold Cdc11p at the SPB, rescues etd1-Δ. Analysis of a suppressor of the mutant etd1-M9 revealed that SIN signalling is influenced by the carbon source of the cell. Growth on a non-fermentable carbon source glycerol reduces the requirement for SIN signalling but does not bypass it. The decreased need for SIN signalling is mediated largely by reduction of protein kinase A activity, and it is phenocopied by deletion of pka1 on glucose medium. We conclude that protein kinase A is an important regulator of the SIN, and that SIN signalling is regulated by the carbon source of the cell.


Asunto(s)
Proteínas Quinasas Dependientes de AMP Cíclico , Schizosaccharomyces , Proteínas Quinasas Dependientes de AMP Cíclico/genética , Schizosaccharomyces/genética , Citoesqueleto de Actina , Carbono , Transducción de Señal
3.
PLoS Biol ; 19(3): e3001158, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33780434

RESUMEN

Since its emergence in December 2019, Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) has spread globally and become a major public health burden. Despite its close phylogenetic relationship to SARS-CoV, SARS-CoV-2 exhibits increased human-to-human transmission dynamics, likely due to efficient early replication in the upper respiratory epithelium of infected individuals. Since different temperatures encountered in the human upper and lower respiratory tract (33°C and 37°C, respectively) have been shown to affect the replication kinetics of several respiratory viruses, as well as host innate immune response dynamics, we investigated the impact of temperature on SARS-CoV-2 and SARS-CoV infection using the primary human airway epithelial cell culture model. SARS-CoV-2, in contrast to SARS-CoV, replicated to higher titers when infections were performed at 33°C rather than 37°C. Although both viruses were highly sensitive to type I and type III interferon pretreatment, a detailed time-resolved transcriptome analysis revealed temperature-dependent interferon and pro-inflammatory responses induced by SARS-CoV-2 that were inversely proportional to its replication efficiency at 33°C or 37°C. These data provide crucial insight on pivotal virus-host interaction dynamics and are in line with characteristic clinical features of SARS-CoV-2 and SARS-CoV, as well as their respective transmission efficiencies.


Asunto(s)
Perfilación de la Expresión Génica/métodos , Regulación Viral de la Expresión Génica/genética , SARS-CoV-2/genética , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/genética , Animales , Antivirales/farmacología , Células Cultivadas , Chlorocebus aethiops , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Células Epiteliales/virología , Regulación Viral de la Expresión Génica/efectos de los fármacos , Interacciones Huésped-Patógeno/efectos de los fármacos , Interacciones Huésped-Patógeno/genética , Humanos , Interferones/farmacología , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/efectos de los fármacos , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/fisiología , SARS-CoV-2/efectos de los fármacos , SARS-CoV-2/fisiología , Especificidad de la Especie , Temperatura , Células Vero , Replicación Viral/efectos de los fármacos , Replicación Viral/genética
4.
Genome Res ; 24(8): 1251-9, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24879558

RESUMEN

Endogenous retroelements (EREs) account for about half of the mouse or human genome, and their potential as insertional mutagens and transcriptional perturbators is suppressed by early embryonic epigenetic silencing. Here, we asked how ERE control is maintained during the generation of induced pluripotent stem cells (iPSCs), as this procedure involves profound epigenetic remodeling. We found that all EREs tested were markedly up-regulated during the reprogramming of either mouse embryonic fibroblasts, human CD34(+) cells, or human primary hepatocytes. At the iPSC stage, EREs of some classes were repressed, whereas others remained highly expressed, yielding a pattern somewhat reminiscent of that recorded in embryonic stem cells. However, variability persisted between individual iPSC clones in the control of specific ERE integrants. Both during reprogramming and in iPS cells, the up-regulation of specific EREs significantly impacted on the transcription of nearby cellular genes. While transcription triggered by specific ERE integrants at highly precise developmental stages may be an essential step toward obtaining pluripotent cells, the broad and unspecific unleashing of the repetitive genome observed here may contribute to the inefficiency of the reprogramming process and to the phenotypic heterogeneity of iPSCs.


Asunto(s)
Retrovirus Endógenos/genética , Células Madre Pluripotentes Inducidas/fisiología , Transcriptoma , Animales , Células Cultivadas , Reprogramación Celular , Silenciador del Gen , Humanos , Ratones , Regulación hacia Arriba
5.
J Biol Chem ; 287(26): 22015-29, 2012 Jun 22.
Artículo en Inglés | MEDLINE | ID: mdl-22493439

RESUMEN

Influenza virus infections lead to a burst of type I interferon (IFN) in the human respiratory tract, which most probably accounts for a rapid control of the virus. Although in mice, IFN-induced Mx1 factor mediates a major part of this response, the situation is less clear in humans. Interestingly, a recently identified IFN-induced cellular protein, tetherin (also known as CD317, BST-2, or HM1.24), exerts potent antiviral activity against a broad range of retroviruses, as well as several other enveloped viruses, by impeding the release of newly generated viral particles from the cell surface. Here we show that influenza virus belongs to the targets of this potent antiviral factor. Ectopic expression of tetherin strongly inhibited fully replicative influenza virus. In addition, depleting endogenous tetherin increased viral production of influenza virions, both in cells constitutively expressing tetherin and upon its induction by IFN. We further demonstrate, by biochemical and morphological means, that tetherin exerts its antiviral action by tethering newly budded viral particles, a mechanism similar to the one that operates against HIV-1. In addition, we determined that the magnitude of tetherin antiviral activity is comparable with or higher than the one of several previously identified anti-influenza cellular factors, such as MxA, ADAR1, ISG15, and viperin. Finally, we demonstrate that influenza virus reduces the impact of tetherin-mediated restriction on its replication by several mechanisms. First, the influenza virus NS1 protein impedes IFN-mediated tetherin induction. Second, influenza infection leads to a decrease of tetherin steady state levels, and the neuraminidase surface protein partly counteracts its activity. Overall, our study helps to delineate the intricate molecular battle taking place between influenza virus and its host cells.


Asunto(s)
Antígenos CD/fisiología , Gripe Humana/metabolismo , Orthomyxoviridae/metabolismo , Animales , Antígenos CD/metabolismo , Línea Celular , Línea Celular Tumoral , Perros , Proteínas Ligadas a GPI/metabolismo , Proteínas Ligadas a GPI/fisiología , Regulación Viral de la Expresión Génica , Glicósido Hidrolasas/metabolismo , Células HeLa , Humanos , Gripe Humana/virología , Microscopía Electrónica/métodos , Modelos Biológicos , Plásmidos/metabolismo , Interferencia de ARN , Subtilisina/metabolismo
6.
Retrovirology ; 10: 6, 2013 Jan 11.
Artículo en Inglés | MEDLINE | ID: mdl-23311681

RESUMEN

BACKGROUND: Dendritic cells and their subsets, located at mucosal surfaces, are among the first immune cells to encounter disseminating pathogens. The cellular restriction factor BST-2/tetherin (also known as CD317 or HM1.24) potently restricts HIV-1 release by retaining viral particles at the cell surface in many cell types, including primary cells such as macrophages. However, BST-2/tetherin does not efficiently restrict HIV-1 infection in immature dendritic cells. RESULTS: We now report that BST-2/tetherin expression in myeloid (myDC) and monocyte-derived dendritic cells (DC) can be significantly up-regulated by IFN-α treatment and TLR-4 engagement with LPS. In contrast to HeLa or 293T cells, infectious HIV-1 release in immature DC and IFN-α-matured DC was only modestly affected in the absence of Vpu compared to wild-type viruses. Strikingly, immunofluorescence analysis revealed that BST-2/tetherin was excluded from HIV containing tetraspanin-enriched microdomains (TEMs) in both immature DC and IFN-α-matured DC. In contrast, in LPS-mediated mature DC, BST-2/tetherin exerted a significant restriction in transfer of HIV-1 infection to CD4+ T cells. Additionally, LPS, but not IFN-α stimulation of immature DC, leads to a dramatic redistribution of cellular restriction factors to the TEM as well as at the virological synapse between DC and CD4+ T cells. CONCLUSIONS: In conclusion, we demonstrate that TLR-4 engagement in immature DC significantly up-regulates the intrinsic antiviral activity of BST-2/tetherin, during cis-infection of CD4+ T cells across the DC/T cell virological synapse. Manipulating the function and potency of cellular restriction factors such as BST-2/tetherin to HIV-1 infection, has implications in the design of antiviral therapeutic strategies.


Asunto(s)
Antígenos CD/inmunología , Linfocitos T CD4-Positivos/inmunología , Células Dendríticas/inmunología , VIH-1/inmunología , Sinapsis Inmunológicas/virología , Receptor Toll-Like 4/inmunología , Virión/inmunología , Antígenos CD/genética , Linfocitos T CD4-Positivos/virología , Diferenciación Celular , Células Dendríticas/efectos de los fármacos , Células Dendríticas/virología , Proteínas Ligadas a GPI/genética , Proteínas Ligadas a GPI/inmunología , Regulación de la Expresión Génica/inmunología , VIH-1/efectos de los fármacos , Células HeLa , Humanos , Sinapsis Inmunológicas/efectos de los fármacos , Interferón-alfa/farmacología , Lipopolisacáridos/farmacología , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Macrófagos/virología , Monocitos/efectos de los fármacos , Monocitos/inmunología , Monocitos/virología , Cultivo Primario de Células , Transducción de Señal , Tetraspaninas/genética , Tetraspaninas/inmunología , Receptor Toll-Like 4/genética , Virión/efectos de los fármacos , Liberación del Virus/efectos de los fármacos , Replicación Viral/efectos de los fármacos
7.
PLoS Pathog ; 7(12): e1002456, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22194693

RESUMEN

Virus assembly and interaction with host-cell proteins occur at length scales below the diffraction limit of visible light. Novel super-resolution microscopy techniques achieve nanometer resolution of fluorescently labeled molecules. The cellular restriction factor tetherin (also known as CD317, BST-2 or HM1.24) inhibits the release of human immunodeficiency virus 1 (HIV-1) through direct incorporation into viral membranes and is counteracted by the HIV-1 protein Vpu. For super-resolution analysis of HIV-1 and tetherin interactions, we established fluorescence labeling of HIV-1 proteins and tetherin that preserved HIV-1 particle formation and Vpu-dependent restriction, respectively. Multicolor super-resolution microscopy revealed important structural features of individual HIV-1 virions, virus assembly sites and their interaction with tetherin at the plasma membrane. Tetherin localization to micro-domains was dependent on both tetherin membrane anchors. Tetherin clusters containing on average 4 to 7 tetherin dimers were visualized at HIV-1 assembly sites. Combined biochemical and super-resolution analysis revealed that extended tetherin dimers incorporate both N-termini into assembling virus particles and restrict HIV-1 release. Neither tetherin domains nor HIV-1 assembly sites showed enrichment of the raft marker GM1. Together, our super-resolution microscopy analysis of HIV-1 interactions with tetherin provides new insights into the mechanism of tetherin-mediated HIV-1 restriction and paves the way for future studies of virus-host interactions.


Asunto(s)
Antígenos CD/metabolismo , Membrana Celular/metabolismo , VIH-1/metabolismo , Proteínas del Virus de la Inmunodeficiencia Humana/metabolismo , Virión/metabolismo , Antígenos CD/química , Línea Celular , Membrana Celular/inmunología , Color , Técnica del Anticuerpo Fluorescente/métodos , Proteínas Ligadas a GPI/química , Proteínas Ligadas a GPI/metabolismo , Humanos , Microscopía Confocal , Microscopía Electrónica de Transmisión/métodos , Transfección , Virión/inmunología
8.
J Exp Med ; 203(13): 2887-93, 2006 Dec 25.
Artículo en Inglés | MEDLINE | ID: mdl-17145955

RESUMEN

HIV-1 infects immature dendritic cells (iDCs), but infection is inefficient compared with activated CD4+ T cells and only involves a small subset of iDCs. We analyzed whether this could be attributed to specific cellular restrictions during the viral life cycle. To study env-independent restriction to HIV-1 infection, we used a single-round infection assay with HIV-1 pseudotyped with vesicular stomatitis virus G protein (HIV-VSVG). Small interfering RNA-mediated depletion of APOBEC3G/3F (A3G/3F), but not TRIM5alpha, enhanced HIV-1 infection of iDCs, indicating that A3G/3F controls the sensitivity of iDCs to HIV-1 infection. Furthermore, sequences of HIV reverse transcripts revealed G-to-A hypermutation of HIV genomes during iDC infection, demonstrating A3G/3F cytidine deaminase activity in iDCs. When we separated the fraction of iDCs that was susceptible to HIV, we found the cells to be deficient in A3G messenger RNA and protein. We also noted that during DC maturation, which further reduces susceptibility to infection, A3G levels increased. These findings highlight a role for A3G/3F in explaining the resistance of most DCs to HIV-1 infection, as well as the susceptibility of a fraction of iDCs. An increase in the A3G/3F-mediated intrinsic resistance of iDCs could result in a block of HIV infection at its mucosal point of entry.


Asunto(s)
Citosina Desaminasa/fisiología , Células Dendríticas/virología , VIH-1/crecimiento & desarrollo , Monocitos/citología , Nucleósido Desaminasas/fisiología , Proteínas Represoras/fisiología , Desaminasa APOBEC-3G , Factores de Restricción Antivirales , Secuencia de Bases , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Proteínas Portadoras/fisiología , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/genética , Línea Celular , Citidina Desaminasa , Citosina Desaminasa/genética , Citosina Desaminasa/metabolismo , ADN Viral/genética , Células Dendríticas/efectos de los fármacos , Células Dendríticas/metabolismo , Citometría de Flujo , Expresión Génica , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , VIH-1/genética , Células HeLa , Humanos , Células Jurkat , Lipopolisacáridos/farmacología , Glicoproteínas de Membrana/genética , Datos de Secuencia Molecular , Nucleósido Desaminasas/genética , Nucleósido Desaminasas/metabolismo , Mutación Puntual , ARN Interferente Pequeño/genética , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Homología de Secuencia de Ácido Nucleico , Transfección , Proteínas de Motivos Tripartitos , Ubiquitina-Proteína Ligasas , Proteínas del Envoltorio Viral/genética , Replicación Viral/genética
9.
PLoS Pathog ; 5(9): e1000574, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19730691

RESUMEN

Host cells impose a broad range of obstacles to the replication of retroviruses. Tetherin (also known as CD317, BST-2 or HM1.24) impedes viral release by retaining newly budded HIV-1 virions on the surface of cells. HIV-1 Vpu efficiently counteracts this restriction. Here, we show that HIV-1 Vpu induces the depletion of tetherin from cells. We demonstrate that this phenomenon correlates with the ability of Vpu to counteract the antiviral activity of both overexpressed and interferon-induced endogenous tetherin. In addition, we show that Vpu co-immunoprecipitates with tetherin and beta-TrCP in a tri-molecular complex. This interaction leads to Vpu-mediated proteasomal degradation of tetherin in a beta-TrCP2-dependent manner. Accordingly, in conditions where Vpu-beta-TrCP2-tetherin interplay was not operative, including cells stably knocked down for beta-TrCP2 expression or cells expressing a dominant negative form of beta-TrCP, the ability of Vpu to antagonize the antiviral activity of tetherin was severely impaired. Nevertheless, tetherin degradation did not account for the totality of Vpu-mediated counteraction against the antiviral factor, as binding of Vpu to tetherin was sufficient for a partial relief of the restriction. Finally, we show that the mechanism used by Vpu to induce tetherin depletion implicates the cellular ER-associated degradation (ERAD) pathway, which mediates the dislocation of ER membrane proteins into the cytosol for subsequent proteasomal degradation. In conclusion, we show that Vpu interacts with tetherin to direct its beta-TrCP2-dependent proteasomal degradation, thereby alleviating the blockade to the release of infectious virions. Identification of tetherin binding to Vpu provides a potential novel target for the development of drugs aimed at inhibiting HIV-1 replication.


Asunto(s)
Antígenos CD/metabolismo , VIH-1/patogenicidad , Proteínas del Virus de la Inmunodeficiencia Humana/metabolismo , Glicoproteínas de Membrana/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Proteínas Reguladoras y Accesorias Virales/metabolismo , Proteínas con Repetición de beta-Transducina/metabolismo , Animales , Línea Celular , Retículo Endoplásmico , Proteínas Ligadas a GPI , Células HeLa , Humanos , Inmunoprecipitación , Interferón-alfa/metabolismo , Ratones , Poliubiquitina/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Dominios y Motivos de Interacción de Proteínas , Virión/metabolismo
10.
J Virol ; 83(23): 12611-21, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19776130

RESUMEN

Retroelements are important evolutionary forces but can be deleterious if left uncontrolled. Members of the human APOBEC3 family of cytidine deaminases can inhibit a wide range of endogenous, as well as exogenous, retroelements. These enzymes are structurally organized in one or two domains comprising a zinc-coordinating motif. APOBEC3G contains two such domains, only the C terminal of which is endowed with editing activity, while its N-terminal counterpart binds RNA, promotes homo-oligomerization, and is necessary for packaging into human immunodeficiency virus type 1 (HIV-1) virions. Here, we performed a large-scale mutagenesis-based analysis of the APOBEC3G N terminus, testing mutants for (i) inhibition of vif-defective HIV-1 infection and Alu retrotransposition, (ii) RNA binding, and (iii) oligomerization. Furthermore, in the absence of structural information on this domain, we used homology modeling to examine the positions of functionally important residues and of residues found to be under positive selection by phylogenetic analyses of primate APOBEC3G genes. Our results reveal the importance of a predicted RNA binding dimerization interface both for packaging into HIV-1 virions and inhibition of both HIV-1 infection and Alu transposition. We further found that the HIV-1-blocking activity of APOBEC3G N-terminal mutants defective for packaging can be almost entirely rescued if their virion incorporation is forced by fusion with Vpr, indicating that the corresponding region of APOBEC3G plays little role in other aspects of its action against this pathogen. Interestingly, residues forming the APOBEC3G dimer interface are highly conserved, contrasting with the rapid evolution of two neighboring surface-exposed amino acid patches, one targeted by the Vif protein of primate lentiviruses and the other of yet-undefined function.


Asunto(s)
Citidina Desaminasa/genética , Citidina Desaminasa/inmunología , VIH-1/inmunología , Desaminasa APOBEC-3G , Secuencia de Aminoácidos , Animales , Secuencia Conservada , Citidina Desaminasa/metabolismo , Dimerización , Humanos , Lentivirus de los Primates , Modelos Moleculares , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Primates , Unión Proteica , Estructura Terciaria de Proteína , ARN Viral/metabolismo , Homología de Secuencia de Aminoácido
11.
Nature ; 424(6944): 99-103, 2003 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-12808466

RESUMEN

Viral replication usually requires that innate intracellular lines of defence be overcome, a task usually accomplished by specialized viral gene products. The virion infectivity factor (Vif) protein of human immunodeficiency virus (HIV) is required during the late stages of viral production to counter the antiviral activity of APOBEC3G (apolipoprotein B mRNA-editing enzyme, catalytic polypeptide-like 3G; also known as CEM15), a protein expressed notably in human T lymphocytes. When produced in the presence of APOBEC3G, vif-defective virus is non-infectious. APOBEC3G is closely related to APOBEC1, the central component of an RNA-editing complex that deaminates a cytosine residue in apoB messenger RNA. APOBEC family members also have potent DNA mutator activity through dC deamination; however, whether the editing potential of APOBEC3G has any relevance to HIV inhibition is unknown. Here, we demonstrate that it does, as APOBEC3G exerts its antiviral effect during reverse transcription to trigger G-to-A hypermutation in the nascent retroviral DNA. We also find that APOBEC3G can act on a broad range of retroviruses in addition to HIV, suggesting that hypermutation by editing is a general innate defence mechanism against this important group of pathogens.


Asunto(s)
ADN Viral/biosíntesis , ADN Viral/genética , VIH-1/genética , Mutagénesis/genética , Proteínas/metabolismo , Transcripción Genética/genética , Desaminasa APOBEC-3G , Antivirales/metabolismo , Secuencia de Bases , Línea Celular , Citidina Desaminasa , Productos del Gen vif/deficiencia , Productos del Gen vif/genética , Productos del Gen vif/metabolismo , VIH-1/fisiología , Humanos , Datos de Secuencia Molecular , Nucleósido Desaminasas , Mutación Puntual/genética , Proteínas/genética , Edición de ARN/genética , ARN Mensajero/biosíntesis , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteínas Represoras , Carga Viral , Replicación Viral , Productos del Gen vif del Virus de la Inmunodeficiencia Humana
12.
J Virol ; 82(13): 6585-90, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18434399

RESUMEN

Interferon (IFN) has been part of the standard treatment of chronic hepatitis B infection for more than 2 decades, yet the mechanism of action of this antiviral remains poorly understood. It was recently observed that members of the human APOBEC family of cytidine deaminases endowed with anti-hepatitis B virus (HBV) activity are upregulated by type I and II IFNs. However, we demonstrated that, in tissue culture, these cellular enzymes are not essential effectors of the anti-HBV action of these cytokines. Here, we show that murine APOBEC3 (muA3) can also block HBV replication. While expressed at low levels in the mouse liver at baseline, muA3 is upregulated upon IFN induction. However, in HBV-transgenic muA3 knockout mice, IFN induction blocked HBV DNA production as efficiently as in control HBV-transgenic muA3-competent animals. We conclude that APOBEC3 is not an essential mediator of the IFN-mediated inhibition of HBV in vivo.


Asunto(s)
Citidina Desaminasa/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Virus de la Hepatitis B , Hepatitis B/virología , Interferones/farmacología , Replicación Viral/genética , Animales , Cartilla de ADN/genética , Genotipo , Hepatitis B/tratamiento farmacológico , Inmunoprecipitación , Interferones/uso terapéutico , Ratones , Ratones Transgénicos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Replicación Viral/efectos de los fármacos
13.
Genome Biol ; 20(1): 71, 2019 04 19.
Artículo en Inglés | MEDLINE | ID: mdl-30999927

RESUMEN

Despite its widespread use, RNA-seq is still too laborious and expensive to replace RT-qPCR as the default gene expression analysis method. We present a novel approach, BRB-seq, which uses early multiplexing to produce 3' cDNA libraries for dozens of samples, requiring just 2 hours of hands-on time. BRB-seq has a comparable performance to the standard TruSeq approach while showing greater tolerance for lower RNA quality and being up to 25 times cheaper. We anticipate that BRB-seq will transform basic laboratory practice given its capacity to generate genome-wide transcriptomic data at a similar cost as profiling four genes using RT-qPCR.


Asunto(s)
Perfilación de la Expresión Génica/métodos , Biblioteca de Genes , Análisis de Secuencia de ARN , Secuenciación de Nucleótidos de Alto Rendimiento
14.
Retrovirology ; 5: 54, 2008 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-18597676

RESUMEN

Human APOBEC3 proteins are editing enzymes that can interfere with the replication of exogenous retroviruses such as human immunodeficiency virus (HIV), hepadnaviruses such as hepatitis B virus (HBV), and with the retrotransposition of endogenous retroelements such as long-interspersed nuclear elements (LINE) and Alu. Here, we show that APOBEC3G, but not other APOBEC3 family members, binds 7SL RNA, the common ancestor of Alu RNAs that is specifically recruited into HIV virions. Our data further indicate that APOBEC3G recognizes 7SL RNA and Alu RNA by its common structure, the Alu domain, suggesting a mechanism for APOBEC3G- mediated inhibition of Alu retrotransposition. However, we also demonstrate that APOBEC3F and APOBEC3G are normally recruited into and inhibit the infectivity of DeltaVif HIV1 virions when 7SLRNA is prevented from accessing particles by RNA interference against SRP14 or by over expression of SRP19, both components of the signal recognition particle. We thus conclude that 7SL RNA is not an essential mediator of the virion packaging of these antiviral cytidine deaminases.


Asunto(s)
Elementos Alu/genética , Citidina Desaminasa/metabolismo , ARN Citoplasmático Pequeño/metabolismo , Partícula de Reconocimiento de Señal/metabolismo , Desaminasa APOBEC-3G , Elementos Alu/fisiología , Secuencia de Bases , Línea Celular , Citidina Desaminasa/genética , VIH-1/metabolismo , Humanos , Datos de Secuencia Molecular , Mutación , ARN Citoplasmático Pequeño/química , Partícula de Reconocimiento de Señal/química , Virión/metabolismo , Ensamble de Virus
15.
Haematologica ; 92(1): 127-8, 2007 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17229647

RESUMEN

We describe a point mutation creating an additional ATG codon in the 5' untranslated region (UTR) of the HAMP gene, in a patient with juvenile hemochromatosis. By transient in vitro transfection studies, we provide evidence that the additional ATG is functional and prevents normal hepcidin production by inducing an aberrant translation initiation of the pre-hepcidin mRNA.


Asunto(s)
Péptidos Catiónicos Antimicrobianos/sangre , Péptidos Catiónicos Antimicrobianos/deficiencia , Péptidos Catiónicos Antimicrobianos/genética , Hemocromatosis/sangre , Mutación Puntual , Biosíntesis de Proteínas , Regiones no Traducidas 5' , Línea Celular , Codón Iniciador , Hepcidinas , Humanos , Modelos Biológicos , Mutación , ARN Mensajero/metabolismo , Transfección
16.
Clin Vaccine Immunol ; 24(1)2017 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-27795301

RESUMEN

Subunit vaccines for prevention of congenital cytomegalovirus (CMV) infection based on glycoprotein B (gB) and pp65 are in clinical trials, but it is unclear whether simultaneous vaccination with both antigens enhances protection. We undertook evaluation of a novel bivalent vaccine based on nonreplicating lymphocytic choriomeningitis virus (rLCMV) vectors expressing a cytoplasmic tail-deleted gB [gB(dCt)] and full-length pp65 from human CMV in mice. Immunization with the gB(dCt) vector alone elicited a comparable gB-binding antibody response and a superior neutralizing response to that elicited by adjuvanted subunit gB. Immunization with the pp65 vector alone elicited robust T cell responses. Comparable immunogenicity of the combined gB(dCt) and pp65 vectors with the individual monovalent formulations was demonstrated. To demonstrate proof of principle for a bivalent rLCMV-based HCMV vaccine, the congenital guinea pig cytomegalovirus (GPCMV) infection model was used to compare rLCMV vectors encoding homologs of pp65 (GP83) and gB(dCt), alone and in combination versus Freund's adjuvanted recombinant gB. Both vectors elicited significant immune responses, and no loss of gB immunogenicity was noted with the bivalent formulation. Combined vaccination with rLCMV-vectored GPCMV gB(dCt) and pp65 (GP83) conferred better protection against maternal viremia than subunit or either monovalent rLCMV vaccine. The bivalent vaccine also was significantly more effective in reducing pup mortality than the monovalent vaccines. In summary, bivalent vaccines with rLCMV vectors expressing gB and pp65 elicited potent humoral and cellular responses and conferred protection in the GPCMV model. Further clinical trials of LCMV-vectored HCMV vaccines are warranted.


Asunto(s)
Infecciones por Citomegalovirus/prevención & control , Vacunas contra Citomegalovirus/inmunología , Portadores de Fármacos , Virus de la Coriomeningitis Linfocítica/genética , Fosfoproteínas/inmunología , Proteínas del Envoltorio Viral/inmunología , Proteínas de la Matriz Viral/inmunología , Animales , Anticuerpos Neutralizantes/sangre , Anticuerpos Antivirales/sangre , Antígenos Virales/genética , Antígenos Virales/inmunología , Infecciones por Citomegalovirus/congénito , Vacunas contra Citomegalovirus/administración & dosificación , Modelos Animales de Enfermedad , Femenino , Cobayas , Ratones Endogámicos C57BL , Fosfoproteínas/genética , Linfocitos T/inmunología , Vacunas Combinadas/administración & dosificación , Vacunas Combinadas/inmunología , Vacunas Sintéticas/administración & dosificación , Vacunas Sintéticas/inmunología , Proteínas del Envoltorio Viral/genética , Proteínas de la Matriz Viral/genética
17.
Vaccine ; 35(1): 1-9, 2017 01 03.
Artículo en Inglés | MEDLINE | ID: mdl-27899229

RESUMEN

An important focus in vaccine research is the design of vaccine vectors with low seroprevalence and high immunogenicity. Replication-incompetent lymphocytic choriomeningitis virus (rLCMV) vectors do not elicit vector-neutralizing antibody responses, and homologous prime-boost regimens with rLCMV vectors induce boostable and protective T cell responses to model antigens in mice. However, cellular and humoral immune responses following homologous rLCMV vaccine regimens have not been rigorously evaluated in non-human primates (NHPs). To test whether rLCMV vectors constitute an effective vaccine platform in NHPs, we developed rLCMV vectors expressing SIVmac239 Env and Gag antigens and assessed their immunogenicity in mice and cynomolgus macaques. Immunization with rLCMV vaccine vectors expressing SIV Env and Gag was effective at generating SIV-specific T cell and antibody responses in both mice and NHPs. Epitope mapping using SIV Env in C57BL/6 mice demonstrated that rLCMV vectors induced sustained poly-functional responses to both dominant and subdominant epitopes. Our results suggest the potential of rLCMV vectors as vaccine candidates. Future SIV challenge experiments in rhesus macaques will be needed to assess immune protection by these vaccine vectors.


Asunto(s)
Antígenos Virales/inmunología , Portadores de Fármacos , Virus de la Coriomeningitis Linfocítica/genética , Vacunas contra el SIDAS/inmunología , Virus de la Inmunodeficiencia de los Simios/inmunología , Animales , Anticuerpos Antivirales/sangre , Antígenos Virales/genética , Macaca fascicularis , Ratones Endogámicos C57BL , Vacunas contra el SIDAS/administración & dosificación , Vacunas contra el SIDAS/genética , Virus de la Inmunodeficiencia de los Simios/genética , Linfocitos T/inmunología , Vacunas Atenuadas/administración & dosificación , Vacunas Atenuadas/genética , Vacunas Atenuadas/inmunología , Vacunas Sintéticas/administración & dosificación , Vacunas Sintéticas/genética , Vacunas Sintéticas/inmunología
18.
Hum Gene Ther ; 16(8): 913-20, 2005 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16076249

RESUMEN

Multicellular organisms have evolved under relentless attacks from pathogens, and as a consequence have spiked their genomes with numerous genes that serve to thwart these threats, notably through the building of the innate and adaptive arms of the immune system. The innate immune system is by far the most ancient, being found as widely as in plants and Drosophila, while adaptive immunity arose with the emergence of cartilaginous fishes. Innate immunity enters rapidly into the game during the course of an infection and generally involves the recognition by specific cellular receptors of common pathogen-associated patterns to elicit broad defensive responses, mediated in humans by interferons, macrophages, and natural killer cells, amongst others. When innate immunity fails to eradicate the infection quickly, adaptive immune responses enter into play, to generate exquisitely specific defenses to virtually any pathogen, thanks to a quasi-infinite repertoire of nonself receptors and effectors. A specific form of innate immunity, coined "intrinsic immunity," completes this protection by providing a constant, always-on, line of defense, generally through intracellular obstacles to the replication of pathogens. This component of the immune system has gained much attention as it was discovered that it is a cornerstone of the resistance of mammals against retroviruses. One of these newly discovered intracellular molecular weapons, the APOBEC family of proteins, is active against several classes of retroelements. We present here the current state of knowledge on this rapidly evolving field and discuss implications for gene therapy.


Asunto(s)
Terapia Genética/tendencias , Inmunidad Innata , Lentivirus/genética , Proteínas/genética , Proteínas/inmunología , Retroelementos , Desaminasa APOBEC-3G , Adaptación Fisiológica , Evolución Biológica , Citidina Desaminasa/metabolismo , Vectores Genéticos , Hepatitis B Crónica/genética , Hepatitis B Crónica/terapia , Humanos , Neoplasias/genética , Neoplasias/terapia , Nucleósido Desaminasas , Proteínas Represoras , Transducción Genética , Virus/patogenicidad
19.
PLoS One ; 8(9): e73780, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24040067

RESUMEN

Ribosome-inactivating proteins (RIPs) are endowed with several medicinal properties, including antiviral activity. We demonstrate here that the recently identified type I RIP from Momordica balsamina also possesses antiviral activity, as determined by viral growth curve assays and single-round infection experiments. Importantly, this activity is at play even as doses where the RIP has no cytotoxic effect. In addition, balsamin inhibits HIV-1 replication not only in T cell lines but also in human primary CD4(+) T cells. This antiviral compound exerts its activity at a viral replicative step occurring later than reverse-transcription, most likely on viral protein translation, prior to viral budding and release. Finally, we demonstrate that balsamin antiviral activity is broad since it also impedes influenza virus replication. Altogether our results demonstrate that type I RIP can exert a potent anti-HIV-1 activity which paves the way for new therapeutic avenues for the treatment of viral infections.


Asunto(s)
VIH-1/efectos de los fármacos , Momordica/metabolismo , Proteínas de Plantas/farmacología , Proteínas Inactivadoras de Ribosomas/farmacología , Replicación Viral/efectos de los fármacos , Animales , Linfocitos T CD4-Positivos/virología , Línea Celular , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Relación Dosis-Respuesta a Droga , Farmacorresistencia Viral/genética , VIH-1/genética , VIH-1/fisiología , Interacciones Huésped-Patógeno/efectos de los fármacos , Humanos , Concentración 50 Inhibidora , Células Jurkat , Mutación , Linfocitos T/patología , Linfocitos T/virología
20.
J Leukoc Biol ; 88(6): 1251-8, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20807705

RESUMEN

DC are major targets of HIV-1 during the early events of infection. Yet, HIV-1 infects these cells only inefficiently in vitro as compared with CD4+T lymphocytes. Accordingly, we have previously identified a strong post-entry block to HIV-1 replication in MDDC as a result of the cellular restriction factor A3G. Furthermore, we have demonstrated that As2O3, a drug used to treat acute promyelocytic leukemia, can fully eliminate the potent post-entry restriction of HIV-1 infection in MDDC and in blood-derived MyDC by mechanisms that were unclear. We are now exploring the interplay between As2O3 and A3G-mediated restriction in primary DC subsets. Here, we report that As2O3 counteracts A3G-mediated restriction in MyDC but not in MDDC. RNAi of A3G in MyDC indicated that the As2O3-mediated increase of HIV-1 infection was largely dependent on the presence of the cellular restriction factor. This study reveals an unexpected interplay between As2O3 and A3G-mediated restriction to HIV-1 infection in primary human MyDC.


Asunto(s)
Antineoplásicos/farmacología , Arsenicales/farmacología , Citidina Desaminasa/fisiología , Células Dendríticas/virología , VIH-1/efectos de los fármacos , Células Mieloides/virología , Óxidos/farmacología , Desaminasa APOBEC-3G , Elementos Alu , Trióxido de Arsénico , Células HEK293 , Humanos , Interferón-alfa/farmacología , Retroelementos , Tropismo Viral
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA