Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
1.
Eur Heart J ; 45(36): 3751-3763, 2024 Sep 29.
Artículo en Inglés | MEDLINE | ID: mdl-39115049

RESUMEN

BACKGROUND AND AIMS: Type 1 long QT syndrome (LQT1) is caused by pathogenic variants in the KCNQ1-encoded Kv7.1 potassium channels, which pathologically prolong ventricular action potential duration (APD). Herein, the pathologic phenotype in transgenic LQT1 rabbits is rescued using a novel KCNQ1 suppression-replacement (SupRep) gene therapy. METHODS: KCNQ1-SupRep gene therapy was developed by combining into a single construct a KCNQ1 shRNA (suppression) and an shRNA-immune KCNQ1 cDNA (replacement), packaged into adeno-associated virus serotype 9, and delivered in vivo via an intra-aortic root injection (1E10 vg/kg). To ascertain the efficacy of SupRep, 12-lead electrocardiograms were assessed in adult LQT1 and wild-type (WT) rabbits and patch-clamp experiments were performed on isolated ventricular cardiomyocytes. RESULTS: KCNQ1-SupRep treatment of LQT1 rabbits resulted in significant shortening of the pathologically prolonged QT index (QTi) towards WT levels. Ventricular cardiomyocytes isolated from treated LQT1 rabbits demonstrated pronounced shortening of APD compared to LQT1 controls, leading to levels similar to WT (LQT1-UT vs. LQT1-SupRep, P < .0001, LQT1-SupRep vs. WT, P = ns). Under ß-adrenergic stimulation with isoproterenol, SupRep-treated rabbits demonstrated a WT-like physiological QTi and APD90 behaviour. CONCLUSIONS: This study provides the first animal-model, proof-of-concept gene therapy for correction of LQT1. In LQT1 rabbits, treatment with KCNQ1-SupRep gene therapy normalized the clinical QTi and cellular APD90 to near WT levels both at baseline and after isoproterenol. If similar QT/APD correction can be achieved with intravenous administration of KCNQ1-SupRep gene therapy in LQT1 rabbits, these encouraging data should compel continued development of this gene therapy for patients with LQT1.


Asunto(s)
Terapia Genética , Canal de Potasio KCNQ1 , Miocitos Cardíacos , Síndrome de Romano-Ward , Animales , Conejos , Canal de Potasio KCNQ1/genética , Terapia Genética/métodos , Síndrome de Romano-Ward/genética , Síndrome de Romano-Ward/terapia , Animales Modificados Genéticamente , Potenciales de Acción , Electrocardiografía , ARN Interferente Pequeño/genética , Síndrome de QT Prolongado/genética , Síndrome de QT Prolongado/terapia , Modelos Animales de Enfermedad
2.
PLoS Pathog ; 15(10): e1008060, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31658294

RESUMEN

The loss of Memory CD4 T-cells (Mem) is a major hallmark of HIV-1 immuno-pathogenesis and occurs early during the first months of primary infection. A lot of effort has been put into understanding the molecular mechanisms behind this loss, yet they still have not been fully identified. In this study, we unveil the unreported role of USP18 in the deleterious effects of sustained type I IFN signaling on Mem, including HIV-1-specific CD4 T-cells. We find that interfering with IFN-I signaling pathway in infected patients, notably by targeting the interferon-stimulated gene USP18, resulted in reduced PTEN expression similar to those observed in uninfected control donors. We show that AKT activation in response to cytokine treatment, T-cell receptor (TcR) triggering, as well as HIV-1 Gag stimulation was significantly improved in infected patients when PTEN or USP18 were inhibited. Finally, our data demonstrate that higher USP18 in Mem from infected patients prevent proper cell survival and long-lasting maintenance in an AKT-dependent manner. Altogether, we establish a direct role for type I IFN/USP18 signaling in the maintenance of total and virus-specific Mem and provide a new mechanism for the reduced survival of these populations during primary HIV-1 infection.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , VIH-1/inmunología , Memoria Inmunológica/inmunología , Interferón Tipo I/inmunología , Fosfohidrolasa PTEN/biosíntesis , Ubiquitina Tiolesterasa/metabolismo , Apoptosis/inmunología , Supervivencia Celular , Infecciones por VIH/inmunología , Infecciones por VIH/patología , VIH-1/patogenicidad , Humanos , Fosfohidrolasa PTEN/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptores de Antígenos de Linfocitos T/inmunología , Transducción de Señal/inmunología , Ubiquitina Tiolesterasa/genética , Productos del Gen gag del Virus de la Inmunodeficiencia Humana/inmunología
3.
J Biol Chem ; 294(27): 10503-10518, 2019 07 05.
Artículo en Inglés | MEDLINE | ID: mdl-31118237

RESUMEN

Type I interferons (IFN-I) are key innate immune effectors predominantly produced by activated plasmacytoid dendritic cells (pDCs). By modulating immune responses at their foundation, IFNs can widely reshape immunity to control infectious diseases and malignancies. Nevertheless, their biological activities can also be detrimental to surrounding healthy cells, as prolonged IFN-I signaling is associated with excessive inflammation and immune dysfunction. The interaction of the human pDC receptor immunoglobulin-like transcript 7 (ILT7) with its IFN-I-regulated ligand, bone marrow stromal cell antigen 2 (BST2) plays a key role in controlling the IFN-I amounts produced by pDCs in response to Toll-like receptor (TLR) activation. However, the structural determinants and molecular features of BST2 that govern ILT7 engagement and activation are largely undefined. Using two functional assays to measure BST2-stimulated ILT7 activation as well as biophysical studies, here we identified two structurally-distinct regions of the BST2 ectodomain that play divergent roles during ILT7 activation. We found that although the coiled-coil region contains a newly defined ILT7-binding surface, the N-terminal region appears to suppress ILT7 activation. We further show that a stable BST2 homodimer binds to ILT7, but post-binding events associated with the unique BST2 coiled-coil plasticity are required to trigger receptor signaling. Hence, BST2 with an unstable or a rigid coiled-coil fails to activate ILT7, whereas substitutions in its N-terminal region enhance activation. Importantly, the biological relevance of these newly defined domains of BST2 is underscored by the identification of substitutions having opposing potentials to activate ILT7 in pathological malignant conditions.


Asunto(s)
Antígeno 2 del Estroma de la Médula Ósea/metabolismo , Receptores Inmunológicos/metabolismo , Secuencia de Aminoácidos , Antígeno 2 del Estroma de la Médula Ósea/química , Antígeno 2 del Estroma de la Médula Ósea/genética , Línea Celular , Dimerización , Humanos , Mutagénesis , Unión Proteica , Conformación Proteica en Hélice alfa , Dominios Proteicos , Alineación de Secuencia
4.
J Virol ; 90(22): 10236-10246, 2016 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-27581991

RESUMEN

BST2/tetherin is a type I interferon (IFN-I)-stimulated host factor that restricts the release of HIV-1 by entrapping budding virions at the cell surface. This membrane-associated protein can also engage and activate the plasmacytoid dendritic cell (pDC)-specific immunoglobulin-like transcript 7 (ILT7) inhibitory receptor to downregulate the IFN-I response by pDCs. Pandemic HIV-1 group M uses Vpu (M-Vpu) to counteract the two BST2 isoforms (long and short) that are expressed in human cells. M-Vpu efficiently downregulates surface long BST2, while it displaces short BST2 molecules away from viral assembly sites. We recently found that this attribute is used by M-Vpu to activate the BST2/ILT7-dependent negative-feedback pathway and to suppress pDC IFN-I responses during sensing of infected cells. However, whether this property is conserved in endemic HIV-1 group O, which has evolved Nef (O-Nef) to counteract specifically the long BST2 isoform, remains unknown. In the present study, we validated that O-Nefs have the capacity to downregulate surface BST2 and enhance HIV-1 particle release although less efficiently than M-Vpu. In contrast to M-Vpu, O-Nef did not efficiently enhance viral spread in T cell culture or displace short BST2 from viral assembly sites to prevent its occlusion by tethered HIV-1 particles. Consequently, O-Nef impairs the ability of BST2 to activate negative ILT7 signaling to suppress the IFN-I response by pDC-containing peripheral blood mononuclear cells (PBMCs) during sensing of infected cells. These distinctive features of BST2 counteraction by O-Nefs may in part explain the limited spread of HIV-1 group O in the human population. IMPORTANCE: The geographical distributions and prevalences of different HIV-1 groups show large variations. Understanding drivers of distinctive viral spread may aid in the development of therapeutic strategies for controlling the spread of HIV-1 pandemic strains. The differential spread of HIV-1 groups appears to be linked to their capacities to antagonize the long and short isoforms of the BST2 restriction factor. We found that the endemic HIV-1 group O-encoded BST2 antagonist Nef is unable to counteract the restriction mediated by short BST2, a condition that impairs its ability to activate ILT7 and suppress pDC antiviral responses. This is in contrast to the pandemic HIV-1 group M-specified BST2 countermeasure Vpu, which displays a diverse array of mechanisms to counteract short and long BST2 isoforms, an attribute that allows the effective control of pDC antiviral responses. These findings may help explain the limited spread of HIV-1 group O as well as the continued predominance of HIV-1 group M throughout the world.


Asunto(s)
Antígenos CD/metabolismo , Células Dendríticas/metabolismo , VIH-1/metabolismo , Antivirales/metabolismo , Línea Celular , Células Dendríticas/virología , Proteínas Ligadas a GPI/metabolismo , Células HEK293 , Infecciones por VIH/metabolismo , Infecciones por VIH/virología , Humanos , Interferones/metabolismo , Leucocitos Mononucleares/metabolismo , Leucocitos Mononucleares/virología , Receptores Inmunológicos/metabolismo , Proteínas Reguladoras y Accesorias Virales/metabolismo
5.
PLoS Pathog ; 11(7): e1005024, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-26172439

RESUMEN

Plasmacytoid dendritic cells (pDCs) constitute a major source of type-I interferon (IFN-I) production during acute HIV infection. Their activation results primarily from TLR7-mediated sensing of HIV-infected cells. However, the interactions between HIV-infected T cells and pDCs that modulate this sensing process remain poorly understood. BST2/Tetherin is a restriction factor that inhibits HIV release by cross-linking virions onto infected cell surface. BST2 was also shown to engage the ILT7 pDC-specific inhibitory receptor and repress TLR7/9-mediated IFN-I production by activated pDCs. Here, we show that Vpu, the HIV-1 antagonist of BST2, suppresses TLR7-mediated IFN-I production by pDC through a mechanism that relies on the interaction of BST2 on HIV-producing cells with ILT7. Even though Vpu downregulates surface BST2 as a mean to counteract the restriction on HIV-1 release, we also find that the viral protein re-locates remaining BST2 molecules outside viral assembly sites where they are free to bind and activate ILT7 upon cell-to-cell contact. This study shows that through a targeted regulation of surface BST2, Vpu promotes HIV-1 release and limits pDC antiviral responses upon sensing of infected cells. This mechanism of innate immune evasion is likely to be important for an efficient early viral dissemination during acute infection.


Asunto(s)
Antígenos CD/inmunología , Células Dendríticas/inmunología , Infecciones por VIH/inmunología , VIH-1/inmunología , Proteínas del Virus de la Inmunodeficiencia Humana/inmunología , Proteínas Reguladoras y Accesorias Virales/inmunología , Antígenos CD/metabolismo , Linfocitos T CD4-Positivos/inmunología , Línea Celular , Técnicas de Cocultivo , Células Dendríticas/metabolismo , Citometría de Flujo , Proteínas Ligadas a GPI/inmunología , Proteínas Ligadas a GPI/metabolismo , Infecciones por VIH/metabolismo , Proteínas del Virus de la Inmunodeficiencia Humana/metabolismo , Humanos , Evasión Inmune/inmunología , Microscopía Confocal , Receptor Cross-Talk/inmunología , Receptores Inmunológicos/inmunología , Receptores Inmunológicos/metabolismo , Resonancia por Plasmón de Superficie , Proteínas Reguladoras y Accesorias Virales/metabolismo
6.
Traffic ; 12(12): 1714-29, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21902775

RESUMEN

Bone marrow stromal cell antigen-2 (BST-2) inhibits human immunodeficiency virus type 1 (HIV-1) release by cross-linking nascent virions on infected cell surface. HIV-1 Vpu is thought to antagonize BST-2 by downregulating its surface levels via a mechanism that involves intracellular sequestration and lysosomal degradation. Here, we investigated the functional importance of cell-surface BST-2 downregulation and the BST-2 pools targeted by Vpu using an inducible proviral expression system. Vpu established a surface BST-2 equilibrium at ∼60% of its initial levels within 6 h, a condition that coincided with detection of viral release. Analysis of BST-2 post-endocytic trafficking revealed that the protein is engaged in a late endosomal pathway independent of Vpu. While Vpu moderately enhanced cell-surface BST-2 clearance, it strongly affected the protein resupply to the plasma membrane via newly synthesized proteins. Noticeably, Vpu affected clearance of surface BST-2 more substantially in Jurkat T cells than in HeLa cells, suggesting a cell-dependent impact of Vpu on the pool of surface BST-2. Collectively, our data reveal that Vpu imposes a new BST-2 equilibrium, incompatible with efficient restriction of HIV-1 release, by combining an acceleration of surface BST-2 natural clearance, whose degree might be cell-type dependent, to a severe impairment of the protein resupply to the plasma membrane.


Asunto(s)
Membrana Celular/metabolismo , VIH-1/metabolismo , Proteínas del Virus de la Inmunodeficiencia Humana/genética , Proteínas del Virus de la Inmunodeficiencia Humana/metabolismo , Proteínas Reguladoras y Accesorias Virales/genética , Proteínas Reguladoras y Accesorias Virales/metabolismo , Antígenos CD/genética , Antígenos CD/metabolismo , Línea Celular Transformada , Línea Celular Tumoral , Membrana Celular/genética , Regulación hacia Abajo , Endocitosis/genética , Endosomas/genética , Endosomas/metabolismo , Proteínas Ligadas a GPI/antagonistas & inhibidores , Proteínas Ligadas a GPI/genética , Proteínas Ligadas a GPI/metabolismo , Células HEK293 , Células HeLa , Humanos , Células Jurkat , Transporte de Proteínas , Linfocitos T
7.
J Virol ; 86(7): 3513-27, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22301143

RESUMEN

BST-2/tetherin is an interferon (IFN)-inducible host restriction factor that inhibits the release of many enveloped viruses and functions as a negative-feedback regulator of IFN production by plasmacytoid dendritic cells. Currently, mechanisms underlying BST2 transcriptional regulation by type I IFN remain largely unknown. Here, we demonstrate that the BST2 promoter is a secondary target of the IFN cascade and show that a single IRF binding site is sufficient to render this promoter responsive to IFN-α. Interestingly, expression of IRF-1 or virus-activated forms of IRF-3 and IRF-7 stimulated the BST2 promoter even under conditions where type I IFN signaling was inhibited. Indeed, vesicular stomatitis virus could directly upregulate BST-2 during infection of mouse embryonic fibroblasts through a process that required IRF-7 but was independent from the type I IFN cascade; however, in order to achieve optimal BST-2 induction, the type I IFN cascade needed to be engaged through activation of IRF-3. Furthermore, using human peripheral blood mononuclear cells, we show that BST-2 upregulation is part of an early intrinsic immune response since TLR8 and TLR3 agonists, known to trigger pathways that mediate activation of IRF proteins, could upregulate BST-2 prior to engagement of the type I IFN pathway. Collectively, our findings reveal that BST2 is activated by the same signals that trigger type I IFN production, outlining a regulatory mechanism ensuring that production of type I IFN and expression of a host restriction factor involved in the IFN negative-feedback loop are closely coordinated.


Asunto(s)
Antígenos CD/genética , Factor 7 Regulador del Interferón/genética , Interferones/metabolismo , Transducción de Señal , Estomatitis Vesicular/genética , Virus de la Estomatitis Vesicular Indiana/fisiología , Animales , Antígenos CD/metabolismo , Secuencia de Bases , Línea Celular , Proteínas Ligadas a GPI/genética , Proteínas Ligadas a GPI/metabolismo , Humanos , Factor 7 Regulador del Interferón/metabolismo , Leucocitos Mononucleares/metabolismo , Leucocitos Mononucleares/virología , Datos de Secuencia Molecular , Regiones Promotoras Genéticas , Activación Transcripcional , Regulación hacia Arriba , Estomatitis Vesicular/metabolismo , Estomatitis Vesicular/virología , Virus de la Estomatitis Vesicular Indiana/genética
8.
J Gen Virol ; 93(Pt 4): 722-732, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22238232

RESUMEN

Human cytomegalovirus (HCMV) is a large DNA virus belonging to the subfamily Betaherpesvirinae. Haematopoietic cells of the myeloid lineage have been shown to harbour latent HCMV. However, following terminal differentiation of these cells, virus is reactivated, and in an immunocompromised host acute infection can occur. It is currently unknown which viral and cellular factors are involved in regulating the switch between lytic and latent infections. Cyclophilin A (CyPA) is a cellular protein that acts as a major factor in virus replication and/or virion maturation for a number of different viruses, including human immunodeficiency virus, hepatitis C virus, murine cytomegalovirus, influenza A virus and vaccinia virus. This study investigated the role of CyPA during HCMV infection. CyPA expression was silenced in human foreskin fibroblast (HF) and THP-1 cells using small interfering RNA (siRNA) technology, or the cells were treated with cyclosporin A (CsA) to inhibit CyPA activity. Silencing CyPA in HF cells with siRNA resulted in an overall reduction in virus production characterized by delayed expression of immediate-early (IE) proteins, decreased viral DNA loads and reduced titres. Furthermore, silencing of CyPA in THP-1 cells pre- and post-differentiation prevented IE protein expression and virus reactivation from a non-productive state. Interestingly, it was observed that treatment of THP-1 cells with CsA prevented the cells from establishing a fully latent infection. In summary, these results demonstrate that CyPA expression is an important factor in HCMV IE protein expression and virus production in lytically infected HF cells, and is a major component in virus reactivation from infected THP-1 cells.


Asunto(s)
Ciclofilina A/fisiología , Infecciones por Citomegalovirus/virología , Citomegalovirus/fisiología , Activación Viral/fisiología , Replicación Viral/fisiología , Western Blotting , Infecciones por Citomegalovirus/metabolismo , Replicación del ADN/fisiología , ADN Viral/metabolismo , Fibroblastos/virología , Silenciador del Gen , Genes Virales/fisiología , Humanos , Proteínas Inmediatas-Precoces/metabolismo , Proteínas Inmediatas-Precoces/fisiología , Reacción en Cadena en Tiempo Real de la Polimerasa
9.
Open Forum Infect Dis ; 9(2): ofab632, 2022 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-35103246

RESUMEN

Population-level immune surveillance, which includes monitoring exposure and assessing vaccine-induced immunity, is a crucial component of public health decision-making during a pandemic. Serosurveys estimating the prevalence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) antibodies in the population played a key role in characterizing SARS-CoV-2 epidemiology during the early phases of the pandemic. Existing serosurveys provide infrastructure to continue immune surveillance but must be adapted to remain relevant in the SARS-CoV-2 vaccine era. Here, we delineate how SARS-CoV-2 serosurveys should be designed to distinguish infection- and vaccine-induced humoral immune responses to efficiently monitor the evolution of the pandemic. We discuss how serosurvey results can inform vaccine distribution to improve allocation efficiency in countries with scarce vaccine supplies and help assess the need for booster doses in countries with substantial vaccine coverage.

10.
Arch Virol ; 156(10): 1847-51, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21625978

RESUMEN

Human cytomegalovirus (HCMV) latency is poorly understood. We previously described a novel HCMV latency-associated transcript, UL81-82ast, coding for a protein designated LUNA (latency unique natural antigen). The aim of this study was to confirm the presence of LUNA in HCMV-seropositive donors. Standard co-immunoprecipitation and ELISA assays were used to detect antibodies against the LUNA protein in the sera of HCMV-seropositive donors. Specific antibodies against LUNA were detected in all HCMV-seropositive donors but in none of the seronegative donors. These data confirm that LUNA is expressed during in vivo infections and is capable of eliciting an immune response.


Asunto(s)
Infecciones por Citomegalovirus/virología , Citomegalovirus/genética , Regulación Viral de la Expresión Génica , Proteínas Virales/genética , Anticuerpos Antivirales/inmunología , Citomegalovirus/inmunología , Citomegalovirus/fisiología , Infecciones por Citomegalovirus/inmunología , Humanos , Proteínas Virales/inmunología , Latencia del Virus
11.
iScience ; 24(11): 103225, 2021 Nov 19.
Artículo en Inglés | MEDLINE | ID: mdl-34712922

RESUMEN

The crosstalk between intestinal epithelial cells (IECs) and Th17-polarized CD4+ T cells is critical for mucosal homeostasis, with HIV-1 causing significant alterations in people living with HIV (PLWH) despite antiretroviral therapy (ART). In a model of IEC and T cell co-cultures, we investigated the effects of IL-17A, the Th17 hallmark cytokine, on IEC ability to promote de novo HIV infection and viral reservoir reactivation. Our results demonstrate that IL-17A acts in synergy with TNF to boost IEC production of CCL20, a Th17-attractant chemokine, and promote HIV trans-infection of CD4+ T cells and viral outgrowth from reservoir cells of ART-treated PLWH. Importantly, the Illumina RNA-sequencing revealed an IL-17A-mediated pro-inflammatory and pro-viral molecular signature, including a decreased expression of type I interferon (IFN-I)-induced HIV restriction factors. These findings point to the deleterious features of IL-17A and raise awareness for caution when designing therapies aimed at restoring the paucity of mucosal Th17 cells in ART-treated PLWH.

12.
Retrovirology ; 7: 114, 2010 Dec 22.
Artículo en Inglés | MEDLINE | ID: mdl-21176220

RESUMEN

Viral protein U (Vpu) is a type 1 membrane-associated accessory protein that is unique to human immunodeficiency virus type 1 (HIV-1) and a subset of related simian immunodeficiency virus (SIV). The Vpu protein encoded by HIV-1 is associated with two primary functions during the viral life cycle. First, it contributes to HIV-1-induced CD4 receptor downregulation by mediating the proteasomal degradation of newly synthesized CD4 molecules in the endoplasmic reticulum (ER). Second, it enhances the release of progeny virions from infected cells by antagonizing Tetherin, an interferon (IFN)-regulated host restriction factor that directly cross-links virions on host cell-surface. This review will mostly focus on recent advances on the role of Vpu in CD4 downregulation and Tetherin antagonism and will discuss how these two functions may have impacted primate immunodeficiency virus cross-species transmission and the emergence of pandemic strain of HIV-1.


Asunto(s)
Infecciones por VIH/virología , VIH-1/fisiología , Proteínas del Virus de la Inmunodeficiencia Humana/metabolismo , Proteínas Reguladoras y Accesorias Virales/metabolismo , Animales , Antígenos CD/genética , Antígenos CD/metabolismo , Antígenos CD4/genética , Antígenos CD4/metabolismo , Proteínas Ligadas a GPI/antagonistas & inhibidores , Proteínas Ligadas a GPI/genética , Proteínas Ligadas a GPI/metabolismo , Infecciones por VIH/genética , Infecciones por VIH/metabolismo , VIH-1/genética , Interacciones Huésped-Patógeno , Proteínas del Virus de la Inmunodeficiencia Humana/genética , Humanos , Datos de Secuencia Molecular , Proteínas Reguladoras y Accesorias Virales/genética
13.
J Virol ; 83(24): 13032-6, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19793820

RESUMEN

The human immunodeficiency virus type 1 (HIV-1) accessory protein Vpu enhances virus particle release by counteracting a host factor that retains virions at the surfaces of infected cells. It was recently demonstrated that cellular protein BST-2/CD317/Tetherin restricts HIV-1 release in a Vpu-dependent manner. Calcium-modulating cyclophilin ligand (CAML) was also proposed to be involved in this process. We investigated whether CAML is involved in cell surface expression of Tetherin. Here, we show that CAML overexpression in permissive Cos-7 cells or CAML depletion in restrictive HeLa cells has no effect on HIV-1 release or on Tetherin surface expression, indicating that CAML is not required for Tetherin-mediated restriction of HIV-1 release.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/fisiología , Antígenos CD/fisiología , VIH-1/fisiología , Glicoproteínas de Membrana/fisiología , Virión/fisiología , Proteínas Adaptadoras Transductoras de Señales/antagonistas & inhibidores , Animales , Antígenos CD/análisis , Células COS , Chlorocebus aethiops , Proteínas Ligadas a GPI , Células HeLa , Humanos , Glicoproteínas de Membrana/análisis
14.
J Virol Methods ; 151(2): 204-210, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18586333

RESUMEN

Peromyscus maniculatus (deer mouse) is the primary reservoir for Sin Nombre virus (SNV). Although the presence of IgG antibodies is often used as a marker of infection, it provides little information on active infections in a population but usually is an indicator of past infections. The presence of IgM antibodies is a much better marker for determining whether active infections are present in a population. A mu-capture SNV-specific IgM enzyme linked immunosorbent assay (ELISA) was developed. From live-trap and release studies a total of 68 rodent sera were studied for the presence of Sin Nombre virus-specific IgG and IgM antibodies. In these studies, IgM responses were detected in a number of animals. In some cases early SNV infection was determined through the presence of anti-SNV IgM before IgG antibodies could be detected. From the set of animals analyzed, it was concluded that the IgM response against SNV can persist anywhere from 1 to up to over 2 months, with a median of less than 1 month. Most importantly, it was demonstrated that anti-Sin Nombre virus IgM is an important tool for detection of early infections in rodents and should be considered as a key diagnostic tool.


Asunto(s)
Inmunoglobulina G/análisis , Inmunoglobulina M/análisis , Peromyscus/inmunología , Peromyscus/virología , Virus Sin Nombre/inmunología , Animales , Anticuerpos Antivirales/análisis , Especificidad de Anticuerpos , Ensayo de Inmunoadsorción Enzimática/métodos , Síndrome Pulmonar por Hantavirus/inmunología , Síndrome Pulmonar por Hantavirus/veterinaria
16.
Cell Rep ; 21(1): 141-153, 2017 Oct 03.
Artículo en Inglés | MEDLINE | ID: mdl-28978468

RESUMEN

Macrophages are heterogeneous immune cells with distinct origins, phenotypes, functions, and tissue localization. Their susceptibility to HIV-1 is subject to variations from permissiveness to resistance, owing in part to regulatory microRNAs. Here, we used RNA sequencing (RNA-seq) to examine the expression of >400 microRNAs in productively infected and bystander cells of HIV-1-exposed macrophage cultures. Two microRNAs upregulated in bystander macrophages, miR-221 and miR-222, were identified as negative regulators of CD4 expression and CD4-mediated HIV-1 entry. Both microRNAs were enhanced by tumor necrosis factor alpha (TNF-α), an inhibitor of CD4 expression. MiR-221/miR-222 inhibitors recovered HIV-1 entry in TNF-α-treated macrophages by enhancing CD4 expression and increased HIV-1 replication and spread in macrophages by countering TNF-α-enhanced miR-221/miR-222 expression in bystander cells. In line with these findings, HIV-1-resistant intestinal myeloid cells express higher levels of miR-221 than peripheral blood monocytes. Thus, miR-221/miR-222 act as effectors of the antiviral host response activated during macrophage infection that restrict HIV-1 entry.


Asunto(s)
Antígenos CD4/genética , VIH-1/genética , Interacciones Huésped-Patógeno , Macrófagos/virología , MicroARNs/genética , Efecto Espectador , Antígenos CD4/metabolismo , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Células HEK293 , VIH-1/crecimiento & desarrollo , VIH-1/metabolismo , Humanos , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , MicroARNs/metabolismo , Cultivo Primario de Células , Análisis de Secuencia de ARN , Transducción de Señal , Células THP-1 , Factor de Necrosis Tumoral alfa/farmacología , Replicación Viral
17.
Viruses ; 8(3): 67, 2016 Mar 03.
Artículo en Inglés | MEDLINE | ID: mdl-26950141

RESUMEN

The plasma membrane protects the cell from its surroundings and regulates cellular communication, homing, and metabolism. Not surprisingly, the composition of this membrane is highly controlled through the vesicular trafficking of proteins to and from the cell surface. As intracellular pathogens, most viruses exploit the host plasma membrane to promote viral replication while avoiding immune detection. This is particularly true for the enveloped human immunodeficiency virus (HIV), which assembles and obtains its lipid shell directly at the plasma membrane. HIV-1 encodes two proteins, negative factor (Nef) and viral protein U (Vpu), which function primarily by altering the quantity and localization of cell surface molecules to increase virus fitness despite host antiviral immune responses. These proteins are expressed at different stages in the HIV-1 life cycle and employ a variety of mechanisms to target both unique and redundant surface proteins, including the viral receptor CD4, host restriction factors, immunoreceptors, homing molecules, tetraspanins and membrane transporters. In this review, we discuss recent progress in the study of the Nef and Vpu targeting of host membrane proteins with an emphasis on how remodeling of the cell membrane allows HIV-1 to avoid host antiviral immune responses leading to the establishment of systemic and persistent infection.


Asunto(s)
Membrana Celular/metabolismo , VIH-1/fisiología , Interacciones Huésped-Patógeno , Proteínas del Virus de la Inmunodeficiencia Humana/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas Reguladoras y Accesorias Virales/metabolismo , Productos del Gen nef del Virus de la Inmunodeficiencia Humana/metabolismo
18.
Cell Metab ; 23(5): 852-66, 2016 05 10.
Artículo en Inglés | MEDLINE | ID: mdl-27166946

RESUMEN

Plasmacytoid dendritic cells (pDCs) are unique bone-marrow-derived cells that produce large amounts of type I interferon in response to microbial stimulation. Furthermore, pDCs also promote T cell tolerance in sterile-inflammation conditions. However, the immunomodulatory role of aortic pDCs in atherosclerosis has been poorly understood. Here, we identified functional mouse and human pDCs in the aortic intima and showed that selective, inducible pDC depletion in mice exacerbates atherosclerosis. Aortic pDCs expressed CCR9 and indoleamine 2,3-dioxygenase 1 (IDO-1), an enzyme involved in driving the generation of regulatory T cells (Tregs). As a consequence, loss of pDCs resulted in decreased numbers of Tregs and reduced IL-10 levels in the aorta. Moreover, antigen presentation by pDCs expanded antigen-specific Tregs in the atherosclerotic aorta. Notably, Tregs ablation affected pDC homeostasis in diseased aorta. Accordingly, pDCs in human atherosclerotic aortas colocalized with Tregs. Collectively, we identified a mechanism of atheroprotection mediated by tolerogenic aortic pDCs.


Asunto(s)
Aorta/patología , Aterosclerosis/enzimología , Aterosclerosis/prevención & control , Células Dendríticas/enzimología , Indolamina-Pirrol 2,3,-Dioxigenasa/metabolismo , Linfocitos T Reguladores/inmunología , Animales , Anticuerpos/farmacología , Aterosclerosis/inmunología , Aterosclerosis/patología , Médula Ósea/patología , Recuento de Células , Proliferación Celular/efectos de los fármacos , Epítopos , Homeostasis/efectos de los fármacos , Humanos , Interferón Tipo I/metabolismo , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Proteínas de la Membrana/metabolismo , Ratones , Ratones Transgénicos , Receptores de LDL/metabolismo , Factores de Tiempo , Receptor Toll-Like 9/metabolismo , Tirosina Quinasa 3 Similar a fms/metabolismo
19.
J Virol Methods ; 123(1): 9-15, 2005 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-15582693

RESUMEN

In this study we identified and characterized a monoclonal antibody against the matrix protein of a cytomegalovirus isolated from the common deer mouse (Peromyscus maniculatus) (PCMV). The monoclonal antibody was isolated using previously described technology which could be applied to the production of monoclonal antibodies against zoonotic disease. The antibody was found to react with a protein homologous to the human cytomegalovirus (HCMV) matrix protein (pp71), the product of the UL82 open reading frame (ORF). mAbs were generated from heterologous fusion of spleen cells from PCMV-positive mice and Balb/C P3X63-Ag8.653 myeloma cells. Using this approach, four monoclonal antibodies: B8C4, C12E8, G6A2 and P4E5 were generated. Antibody G6A2 reacted strongly with PCMV-infected cells as well as purified virions on ELISA and immunofluorescence. Western blot analysis, using sucrose gradient-purified virions, demonstrated that this mAb reacted specifically to a single protein with an apparent molecular weight of 71 kDa. The protein band was excised from the gel, purified and subjected to trypsin digestion followed by mass spectrometry. The protein sequences obtained were found to have identity to HCMV UL82 gene product. Sequence analysis indicated that it is the putative HCMV pp71 protein homolog of PCMV. G6A2 mAb did not cross-react with either human or murine recombinant pp71 proteins expressed in mammalian cells.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/metabolismo , Muromegalovirus/inmunología , Peromyscus/virología , Proteínas Virales/metabolismo , Secuencia de Aminoácidos , Animales , Anticuerpos Monoclonales/biosíntesis , Anticuerpos Antivirales/biosíntesis , Anticuerpos Antivirales/inmunología , Anticuerpos Antivirales/metabolismo , Línea Celular Tumoral , Infecciones por Herpesviridae/inmunología , Infecciones por Herpesviridae/virología , Humanos , Hibridomas , Ratones , Ratones Endogámicos BALB C , Datos de Secuencia Molecular , Análisis de Secuencia de Proteína , Proteínas Virales/química , Proteínas Virales/inmunología
20.
J Vis Exp ; (103)2015 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-26383704

RESUMEN

HIV-1 innate sensing requires direct contact of infected CD4+ T cells with plasmacytoid dendritic cells (pDCs). In order to study this process, the protocols described here use freshly isolated human peripheral blood mononuclear cells (PBMCs) or plasmacytoid dendritic cells (pDCs) to sense infections in either T cell line (MT4) or heterologous primary CD4+ T cells. In order to ensure proper sensing, it is essential that PBMC are isolated immediately after blood collection and that optimal percentage of infected T cells are used. Furthermore, multi-parametric flow cytometric staining can be used to confirm that PBMC samples contain the different cell lineages at physiological ratios. A number of controls can also be included to evaluate viability and functionality of pDCs. These include, the presence of specific surface markers, assessing cellular responses to known agonist of Toll-Like Receptors (TLR) pathways, and confirming a lack of spontaneous type-I interferon (IFN) production. In this system, freshly isolated PBMCs or pDCs are co-cultured with HIV-1 infected cells in 96 well plates for 18-22 hr. Supernatants from these co-cultures are then used to determine the levels of bioactive type-I IFNs by monitoring the activation of the ISGF3 pathway in HEK-Blue IFN-α/ß cells. Prior and during co-culture conditions, target cells can be subjected to flow cytometric analysis to determine a number of parameters, including the percentage of infected cells, levels of specific surface markers, and differential killing of infected cells. Although, these protocols were initially developed to follow type-I IFN production, they could potentially be used to study other imuno-modulatory molecules released from pDCs and to gain further insight into the molecular mechanisms governing HIV-1 innate sensing.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/virología , Técnicas de Cocultivo/métodos , VIH-1/inmunología , Linfocitos T CD4-Positivos/citología , Células Dendríticas/citología , Células Dendríticas/inmunología , Células Dendríticas/virología , Citometría de Flujo , Humanos , Interferón Tipo I/inmunología , Leucocitos Mononucleares/citología , Leucocitos Mononucleares/inmunología , Leucocitos Mononucleares/virología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA