Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
J Virol ; 97(2): e0189022, 2023 02 28.
Artículo en Inglés | MEDLINE | ID: mdl-36688652

RESUMEN

Roseoloviruses (human herpesvirus 6A [HHV-6A], -6B, and -7) infect >90% of the human population during early childhood and are thought to remain latent or persistent throughout the life of the host. As such, these viruses are among the most pervasive and stealthy of all viruses; they must necessarily excel at escaping immune detection throughout the life of the host, and yet, very little is known about how these viruses so successfully escape host defenses. Here, we characterize the expression, trafficking, and posttranslational modifications of the HHV6B U20 gene product, which is encoded within a block of genes unique to the roseoloviruses. HHV-6B U20 trafficked slowly through the secretory system, receiving several posttranslational modifications to its N-linked glycans, indicative of surface-expressed glycoproteins, and eventually reaching the cell surface before being internalized. Interestingly, U20 is also phosphorylated on at least one Ser, Thr, or Tyr residue. These results provide a framework to understand the role(s) of U20 in evading host defenses. IMPORTANCE The roseolovirus U20 proteins are virus-encoded integral membrane glycoproteins possessing class I major histocompatibility complex (MHC)-like folds. Surprisingly, although U20 proteins from HHV-6A and -6B share 92% identity, recent studies ascribe different functions to HHV6A U20 and HHV6B U20. HHV6A U20 was shown to downregulate NKG2D ligands, while HHV6B U20 was shown to inhibit tumor necrosis factor alpha (TNF-α)-induced apoptosis during nonproductive infection with HHV6B (E. Kofod-Olsen, K. Ross-Hansen, M. H. Schleimann, D. K. Jensen, et al., J Virol 86:11483-11492, 2012, https://doi.org/10.1128/jvi.00847-12; A. E. Chaouat, B. Seliger, O. Mandelboim, D. Schmiedel, Front Immunol 12:714799, 2021, https://doi.org/10.3389/fimmu.2021.714799). Here, we have performed cell biological and biochemical characterization of the trafficking, glycosylation, and posttranslational modifications occurring on HHV6B U20.


Asunto(s)
Glicoproteínas de Membrana , Infecciones por Roseolovirus , Proteínas Virales , Humanos , Herpesvirus Humano 6/genética , Herpesvirus Humano 6/inmunología , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Infecciones por Roseolovirus/inmunología , Infecciones por Roseolovirus/virología , Proteínas Virales/genética , Proteínas Virales/inmunología , Evasión Inmune
2.
J Virol ; 95(14): e0162820, 2021 06 24.
Artículo en Inglés | MEDLINE | ID: mdl-33952641

RESUMEN

Like all herpesviruses, the roseoloviruses (HHV6A, -6B, and -7) establish lifelong infection within their host, requiring these viruses to evade host antiviral responses. One common host-evasion strategy is the downregulation of host-encoded, surface-expressed glycoproteins. Roseoloviruses have been shown to evade the host immune response by downregulating NK-activating ligands, class I MHC, and the TCR/CD3 complex. To more globally identify glycoproteins that are differentially expressed on the surface of HHV6A-infected cells, we performed cell surface capture of N-linked glycoproteins present on the surface of T cells infected with HHV6A, and compared these to proteins present on the surface of uninfected T cells. We found that the protein tyrosine phosphatase CD45 is downregulated in T cells infected with HHV6A. We also demonstrated that CD45 is similarly downregulated in cells infected with HHV7. CD45 is essential for signaling through the T cell receptor and, as such, is necessary for developing a fully functional immune response. Interestingly, the closely related betaherpesviruses human cytomegalovirus (HCMV) and murine cytomegalovirus (MCMV) have also separately evolved unique mechanisms to target CD45. While HCMV and MCMV target CD45 signaling and trafficking, HHV6A acts to downregulate CD45 transcripts. IMPORTANCE Human herpesviruses-6 and -7 infect essentially 100% of the world's population before the age of 5 and then remain latent or persistent in their host throughout life. As such, these viruses are among the most pervasive and stealthy of all viruses. Host immune cells rely on the presence of surface-expressed proteins to identify and target virus-infected cells. Here, we investigated the changes that occur to proteins expressed on the cell surface of T cells after infection with human herpesvirus-6A. We discovered that HHV-6A infection results in a reduction of CD45 on the surface of infected T cells and impaired activation in response to T cell receptor stimulation.


Asunto(s)
Regulación Enzimológica de la Expresión Génica , Regulación Viral de la Expresión Génica , Herpesvirus Humano 6/genética , Herpesvirus Humano 7/genética , Antígenos Comunes de Leucocito/genética , Linfocitos T/virología , Línea Celular , Regulación hacia Abajo , Células HEK293 , Herpesvirus Humano 6/metabolismo , Herpesvirus Humano 7/metabolismo , Humanos , Estabilidad Proteica , Receptores de Antígenos de Linfocitos T/metabolismo , Linfocitos T/inmunología , Linfocitos T/metabolismo
3.
J Immunol ; 205(1): 272-281, 2020 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-32444392

RESUMEN

Acute graft-versus-host disease (GVHD) is a frequent complication of hematopoietic transplantation, yet patient risk stratification remains difficult, and prognostic biomarkers to guide early clinical interventions are lacking. We developed an approach to evaluate the potential of human T cells from hematopoietic grafts to produce GVHD. Nonconditioned NBSGW mice transplanted with titrated doses of human bone marrow developed GVHD that was characterized by widespread lymphocyte infiltration and organ pathology. Interestingly, GVHD was not an inevitable outcome in our system and was influenced by transplant dose, inflammatory status of the host, and type of graft. Mice that went on to develop GVHD showed signs of rapid proliferation in the human T cell population during the first 1-3 wk posttransplant and had elevated human IFN-γ in plasma that correlated negatively with the expansion of the human hematopoietic compartment. Furthermore, these early T cell activation metrics were predictive of GVHD onset 3-6 wk before phenotypic pathology. These results reveal an early window of susceptibility for pathological T cell activation following hematopoietic transplantation that is not simply determined by transient inflammation resulting from conditioning-associated damage and show that T cell parameters during this window can serve as prognostic biomarkers for risk of later GVHD development.


Asunto(s)
Enfermedad Injerto contra Huésped/diagnóstico , Trasplante de Células Madre Hematopoyéticas/efectos adversos , Linfocitos T/inmunología , Animales , Proliferación Celular , Células Cultivadas , Modelos Animales de Enfermedad , Femenino , Enfermedad Injerto contra Huésped/sangre , Enfermedad Injerto contra Huésped/inmunología , Humanos , Interferón gamma/sangre , Interferón gamma/inmunología , Activación de Linfocitos , Masculino , Ratones , Periodo Posoperatorio , Cultivo Primario de Células , Pronóstico , Factores de Tiempo , Quimera por Trasplante/inmunología , Acondicionamiento Pretrasplante/efectos adversos , Trasplante Heterólogo/efectos adversos
4.
PLoS Pathog ; 12(8): e1005868, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27580123

RESUMEN

The natural killer cell receptor NKG2D activates NK cells by engaging one of several ligands (NKG2DLs) belonging to either the MIC or ULBP families. Human cytomegalovirus (HCMV) UL16 and UL142 counteract this activation by retaining NKG2DLs and US18 and US20 act via lysomal degradation but the importance of NK cell evasion for infection is unknown. Since NKG2DLs are highly conserved in rhesus macaques, we characterized how NKG2DL interception by rhesus cytomegalovirus (RhCMV) impacts infection in vivo. Interestingly, RhCMV lacks homologs of UL16 and UL142 but instead employs Rh159, the homolog of UL148, to prevent NKG2DL surface expression. Rh159 resides in the endoplasmic reticulum and retains several NKG2DLs whereas UL148 does not interfere with NKG2DL expression. Deletion of Rh159 releases human and rhesus MIC proteins, but not ULBPs, from retention while increasing NK cell stimulation by infected cells. Importantly, RhCMV lacking Rh159 cannot infect CMV-naïve animals unless CD8+ cells, including NK cells, are depleted. However, infection can be rescued by replacing Rh159 with HCMV UL16 suggesting that Rh159 and UL16 perform similar functions in vivo. We therefore conclude that cytomegaloviral interference with NK cell activation is essential to establish but not to maintain chronic infection.


Asunto(s)
Infecciones por Citomegalovirus/inmunología , Citomegalovirus/inmunología , Evasión Inmune , Células Asesinas Naturales/inmunología , Activación de Linfocitos , Animales , Humanos , Células K562 , Macaca fascicularis , Glicoproteínas de Membrana/inmunología , Subfamilia K de Receptores Similares a Lectina de Células NK/inmunología , Proteínas Virales/inmunología
5.
J Virol ; 88(6): 3298-308, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24390327

RESUMEN

UNLABELLED: The U21 gene product from human herpesvirus 7 binds to and redirects class I major histocompatibility complex (MHC) molecules to a lysosomal compartment. The molecular mechanism by which U21 reroutes class I MHC molecules to lysosomes is not known. Here, we have reconstituted the interaction between purified soluble U21 and class I MHC molecules, suggesting that U21 does not require additional cellular proteins to interact with class I MHC molecules. Our results demonstrate that U21, itself predicted to contain an MHC class I-like protein fold, interacts tightly with class I MHC molecules as a tetramer, in a 4:2 stoichiometry. These observations have helped to elucidate a refined model describing the mechanism by which U21 escorts class I MHC molecules to the lysosomal compartment. IMPORTANCE: In this report, we show that the human herpesvirus 7 (HHV-7) immunoevasin U21, itself a class I MHC-like protein, binds with high affinity to class I MHC molecules as a tetramer and escorts them to lysosomes, where they are degraded. While many class I MHC-like molecules have been described in detail, this unusual viral class I-like protein functions as a tetramer, associating with class I MHC molecules in a 4:2 ratio, illuminating a functional significance of homooligomerization of a class I MHC-like protein.


Asunto(s)
Proteínas Portadoras/química , Proteínas Portadoras/metabolismo , Herpesvirus Humano 7/metabolismo , Antígenos de Histocompatibilidad Clase I/metabolismo , Infecciones por Roseolovirus/metabolismo , Infecciones por Roseolovirus/virología , Proteínas Virales/química , Proteínas Virales/metabolismo , Proteínas Portadoras/genética , Herpesvirus Humano 7/química , Herpesvirus Humano 7/genética , Humanos , Unión Proteica , Multimerización de Proteína , Proteínas Virales/genética
6.
Front Immunol ; 15: 1363156, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38953028

RESUMEN

Introduction: Human Herpesvirus 6B (HHV-6B) impedes host immune responses by downregulating class I MHC molecules (MHC-I), hindering antigen presentation to CD8+ T cells. Downregulation of MHC-I disengages inhibitory receptors on natural killer (NK) cells, resulting in activation and killing of the target cell if NK cell activating receptors such as NKG2D have engaged stress ligands upregulated on the target cells. Previous work has shown that HHV-6B downregulates three MHC-like stress ligands MICB, ULBP1, and ULBP3, which are recognized by NKG2D. The U20 glycoprotein of the related virus HHV-6A has been implicated in the downregulation of ULBP1, but the precise mechanism remains undetermined. Methods: We set out to investigate the role of HHV-6B U20 in modulating NK cell activity. We used HHV-6B U20 expressed as a recombinant protein or transduced into target cells, as well as HHV-6B infection, to investigate binding interactions with NK cell ligands and receptors and to assess effects on NK cell activation. Small-angle X-ray scattering was used to align molecular models derived from machine-learning approaches. Results: We demonstrate that U20 binds directly to ULBP1 with sub-micromolar affinity. Transduction of U20 decreases NKG2D binding to ULBP1 at the cell surface but does not decrease ULBP1 protein levels, either at the cell surface or in toto. HHV-6B infection and soluble U20 have the same effect. Transduction of U20 blocks NK cell activation in response to cell-surface ULBP1. Structural modeling of the U20 - ULBP1 complex indicates some similarities to the m152-RAE1γ complex.


Asunto(s)
Proteínas Ligadas a GPI , Herpesvirus Humano 6 , Células Asesinas Naturales , Activación de Linfocitos , Subfamilia K de Receptores Similares a Lectina de Células NK , Humanos , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/metabolismo , Herpesvirus Humano 6/inmunología , Proteínas Ligadas a GPI/metabolismo , Proteínas Ligadas a GPI/inmunología , Subfamilia K de Receptores Similares a Lectina de Células NK/metabolismo , Subfamilia K de Receptores Similares a Lectina de Células NK/inmunología , Activación de Linfocitos/inmunología , Unión Proteica , Proteínas Virales/inmunología , Proteínas Virales/metabolismo , Glicoproteínas/inmunología , Glicoproteínas/metabolismo , Péptidos y Proteínas de Señalización Intracelular
7.
Cancer Immun ; 13: 9, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23885215

RESUMEN

Invariant natural killer T (iNKT) cells are innate T lymphocytes that specifically recognize α-linked glycosphingolipids (α-GSLs) as antigens presented by CD1d molecules. Activating iNKT cells by administering α-GSLs improves disease outcomes in murine cancer models and, thus, there is great interest in the clinical potential of these lipids for treating human cancers. However, humans possess several other CD1 isoforms that are not present in mice and it is not clear whether these CD1 molecules, which also bind lipids, affect human iNKT cell responses. We demonstrate here that CD1c, which is co-expressed with CD1d on blood dendritic cells and on a fraction of B cells, is able to present α-galactosylceramide (α-GalCer) as a weak agonist to human iNKT cells, and that the presence of CD1c synergistically enhances α-GalCerdependent activation of iNKT cells by CD1d. Primary human B cells expressing CD1c induced stronger iNKT cell responses to α-GalCer than the CD1c- subset, and an antibody against CD1c inhibited iNKT cell cytokine secretion. These results suggest that therapeutic activation of human iNKT cells by α-GSLs will be driven preferentially by CD1c+ cell types. Thus, B cell neoplasias that co-express CD1c and CD1d may be particularly susceptible to α-GSL therapy, and cancer vaccines using α-GSLs as adjuvants may be most effective when presented by CD1c+ antigen-presenting cells.


Asunto(s)
Antígenos CD1/biosíntesis , Galactosilceramidas/inmunología , Glicoproteínas/biosíntesis , Células T Asesinas Naturales/inmunología , Secuencia de Aminoácidos , Animales , Antígenos CD1/inmunología , Antígenos CD1/metabolismo , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Glicoproteínas/inmunología , Glicoproteínas/metabolismo , Células HeLa , Humanos , Activación de Linfocitos/inmunología , Ratones , Modelos Moleculares , Datos de Secuencia Molecular , Unión Proteica
8.
PLoS Pathog ; 7(11): e1002362, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22102813

RESUMEN

Herpesviruses have evolved numerous immune evasion strategies to facilitate establishment of lifelong persistent infections. Many herpesviruses encode gene products devoted to preventing viral antigen presentation as a means of escaping detection by cytotoxic T lymphocytes. The human herpesvirus-7 (HHV-7) U21 gene product, for example, is an immunoevasin that binds to class I major histocompatibility complex molecules and redirects them to the lysosomal compartment. Virus infection can also induce the upregulation of surface ligands that activate NK cells. Accordingly, the herpesviruses have evolved a diverse array of mechanisms to prevent NK cell engagement of NK-activating ligands on virus-infected cells. Here we demonstrate that the HHV-7 U21 gene product interferes with NK recognition. U21 can bind to the NK activating ligand ULBP1 and reroute it to the lysosomal compartment. In addition, U21 downregulates the surface expression of the NK activating ligands MICA and MICB, resulting in a reduction in NK-mediated cytotoxicity. These results suggest that this single viral protein may interfere both with CTL-mediated recognition through the downregulation of class I MHC molecules as well as NK-mediated recognition through downregulation of NK activating ligands.


Asunto(s)
Proteínas Portadoras/metabolismo , Citotoxicidad Inmunológica , Herpesvirus Humano 7/patogenicidad , Antígenos de Histocompatibilidad Clase I/metabolismo , Células Asesinas Naturales/inmunología , Proteínas Virales/metabolismo , Presentación de Antígeno , Línea Celular , Proteínas Ligadas a GPI/metabolismo , Células HEK293 , Herpesvirus Humano 7/inmunología , Herpesvirus Humano 7/metabolismo , Humanos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Células Asesinas Naturales/metabolismo , Células Asesinas Naturales/virología , Lisosomas , Infecciones por Roseolovirus/inmunología , Proteínas Virales/inmunología
9.
Front Immunol ; 13: 864898, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35444636

RESUMEN

Human roseolovirus U20 and U21 are type I membrane glycoproteins that have been implicated in immune evasion by interfering with recognition of classical and non-classical MHC proteins. U20 and U21 are predicted to be type I glycoproteins with extracytosolic immunoglobulin-like domains, but detailed structural information is lacking. AlphaFold and RoseTTAfold are next generation machine-learning-based prediction engines that recently have revolutionized the field of computational three-dimensional protein structure prediction. Here, we review the structural biology of viral immunoevasins and the current status of computational structure prediction algorithms. We use these computational tools to generate structural models for U20 and U21 proteins, which are predicted to adopt MHC-Ia-like folds with closed MHC platforms and immunoglobulin-like domains. We evaluate these structural models and place them within current understanding of the structural basis for viral immune evasion of T cell and natural killer cell recognition.


Asunto(s)
Herpesvirus Humano 6 , Herpesvirus Humano 7 , Infecciones por Roseolovirus , Herpesvirus Humano 6/metabolismo , Herpesvirus Humano 7/metabolismo , Humanos , Modelos Estructurales , Proteínas Virales/metabolismo
10.
Front Immunol ; 13: 936968, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35677042

RESUMEN

[This corrects the article DOI: 10.3389/fimmu.2022.864898.].

11.
J Biol Chem ; 285(47): 37016-29, 2010 Nov 19.
Artículo en Inglés | MEDLINE | ID: mdl-20833720

RESUMEN

The U21 open reading frame from human herpesvirus-7 encodes a membrane protein that associates with and redirects class I MHC molecules to the lysosomal compartment. The mechanism by which U21 accomplishes this trafficking excursion is unknown. Here we have examined the contribution of localization, glycosylation, domain structure, and the absence of substrate class I MHC molecules on the ability of U21 to traffic to lysosomes. Our results suggest the existence of a cellular protein necessary for U21-mediated rerouting of class I MHC molecules.


Asunto(s)
Proteínas Portadoras/metabolismo , Glioblastoma/metabolismo , Antígeno HLA-A2/metabolismo , Herpesvirus Humano 7/metabolismo , Lisosomas/metabolismo , Proteínas Virales/metabolismo , Western Blotting , Proteínas Portadoras/antagonistas & inhibidores , Proteínas Portadoras/genética , Diferenciación Celular , Citoplasma/metabolismo , Retículo Endoplásmico/metabolismo , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Glicosilación , Antígeno HLA-A2/genética , Humanos , Inmunoprecipitación , Fragmentos de Péptidos/metabolismo , Transporte de Proteínas , Interferencia de ARN , ARN Mensajero/genética , ARN Interferente Pequeño/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células Tumorales Cultivadas , Proteínas Virales/antagonistas & inhibidores , Proteínas Virales/genética , Microglobulina beta-2/antagonistas & inhibidores , Microglobulina beta-2/genética , Microglobulina beta-2/metabolismo
12.
J Virol ; 84(8): 3738-51, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20106916

RESUMEN

Herpesviruses have evolved numerous strategies to evade detection by the immune system. Notably, most of the herpesviruses interfere with viral antigen presentation to cytotoxic T lymphocytes (CTLs) by removing class I major histocompatibility complex (MHC) molecules from the infected cell surface. Clearly, since the herpesviruses have evolved an extensive array of mechanisms to remove class I MHC molecules from the cell surface, this strategy serves them well. However, class I MHC molecules often serve as inhibitory ligands for NK cells, so viral downregulation of all class I MHC molecules should leave the infected cell open to NK cell attack. Some viruses solve this problem by selectively downregulating certain class I MHC products, leaving other class I products at the cell surface to serve as inhibitory NK cell ligands. Here, we show that human herpesvirus 7 (HHV-7) U21 binds to and downregulates all of the human class I MHC gene products, as well as the murine class I molecule H-2K(b). HHV-7-infected cells must therefore possess other means of escaping NK cell detection.


Asunto(s)
Proteínas Portadoras/fisiología , Regulación hacia Abajo , Herpesvirus Humano 7/inmunología , Herpesvirus Humano 7/patogenicidad , Antígenos de Histocompatibilidad Clase I/biosíntesis , Antígenos de Histocompatibilidad Clase I/inmunología , Proteínas Virales/fisiología , Animales , Línea Celular , Células Cultivadas , Humanos , Ratones , Unión Proteica , Mapeo de Interacción de Proteínas
13.
Life Sci Alliance ; 4(7)2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34112724

RESUMEN

Invariant natural killer T (iNKT) cells are a conserved population of innate T lymphocytes that interact with key antigen-presenting cells to modulate adaptive T-cell responses in ways that can either promote protective immunity, or limit pathological immune activation. Understanding the immunological networks engaged by iNKT cells to mediate these opposing functions is a key pre-requisite to effectively using iNKT cells for therapeutic applications. Using a human umbilical cord blood xenotransplantation model, we show here that co-transplanted allogeneic CD4+ iNKT cells interact with monocytes and T cells in the graft to coordinate pro-hematopoietic and immunoregulatory pathways. The nexus of iNKT cells, monocytes, and cord blood T cells led to the release of cytokines (IL-3, GM-CSF) that enhance hematopoietic stem and progenitor cell activity, and concurrently induced PGE2-mediated suppression of T-cell inflammatory responses that limit hematopoietic stem and progenitor cell engraftment. This resulted in successful long-term hematopoietic engraftment without pretransplant conditioning, including multi-lineage human chimerism and colonization of the spleen by antibody-producing human B cells. These results highlight the potential for using iNKT cellular immunotherapy to improve rates of hematopoietic engraftment independently of pretransplant conditioning.


Asunto(s)
Células T Asesinas Naturales/inmunología , Células T Asesinas Naturales/metabolismo , Inmunología del Trasplante/inmunología , Animales , Células Presentadoras de Antígenos/inmunología , Citocinas/inmunología , Femenino , Sangre Fetal/inmunología , Humanos , Inmunidad Innata/inmunología , Inmunoterapia/métodos , Activación de Linfocitos/inmunología , Ratones , Ratones Endogámicos NOD , Trasplante de Tejidos/métodos
14.
Mol Biol Cell ; 31(3): 196-208, 2020 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-31851583

RESUMEN

The human herpesvirus-7 (HHV-7) U21 glycoprotein binds to class I major histocompatibility complex (MHC) molecules in the endoplasmic reticulum (ER) and reroutes them to lysosomes. How this single viral glycoprotein efficiently redirects the U21/class I MHC complex to the lysosomal compartment is poorly understood. To investigate the trafficking of HHV-7 U21, we followed synchronous release of U21 from the ER as it traffics through the secretory system. Sorting of integral membrane proteins from the trans-Golgi network (TGN) has been shown to occur through tubular carriers that emanate from the TGN or through vesicular carriers that recruit GGA (Golgi-localized, γ-ear-containing, ARF-binding protein), clathrin adaptors, and clathrin. Here, we present evidence for the existence of a third type of Golgi-derived carrier that is vesicular, yet clathrin independent. This U21-containing carrier also carries a Golgi membrane protein engineered to form inducible oligomers. We propose that U21 employs the novel mechanism of forming oligomeric complexes with class I MHC molecules that result in sorting of the oligomeric U21/class I MHC complexes to Golgi--derived quality control carriers destined for lysosomes.


Asunto(s)
Proteínas Portadoras/metabolismo , Herpesvirus Humano 7/metabolismo , Proteínas Virales/metabolismo , Proteínas Adaptadoras del Transporte Vesicular/metabolismo , Clatrina/metabolismo , Retículo Endoplásmico/metabolismo , Aparato de Golgi/virología , Células HeLa , Antígenos de Histocompatibilidad Clase I/metabolismo , Antígenos de Histocompatibilidad Clase I/fisiología , Humanos , Lisosomas/metabolismo , Lisosomas/fisiología , Unión Proteica , Transporte de Proteínas , Red trans-Golgi/metabolismo
15.
Front Immunol ; 11: 573406, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33193358

RESUMEN

NBSGW mice are highly immunodeficient and carry a hypomorphic mutation in the c-kit gene, providing a host environment that supports robust human hematopoietic expansion without pre-conditioning. These mice thus provide a model to investigate human hematopoietic engraftment in the absence of conditioning-associated damage. We compared transplantation of human CD34+ HSPCs purified from three different sources: umbilical cord blood, adult bone marrow, and adult G-CSF mobilized peripheral blood. HSPCs from mobilized peripheral blood were significantly more efficient (as a function of starting HSPC dose) than either cord blood or bone marrow HSPCs at generating high levels of human chimerism in the murine blood and bone marrow by 12 weeks post-transplantation. While T cells do not develop in this model due to thymic atrophy, all three HSPC sources generated a human compartment that included B lymphocytic, myeloid, and granulocytic lineages. However, the proportions of these lineages varied significantly according to HSPC source. Mobilized blood HSPCs produced a strikingly higher proportion of granulocyte lineage cells (~35% as compared to ~5%), whereas bone marrow HSPC output was dominated by B lymphocytic cells, and cord blood HSPC output was enriched for myeloid lineages. Following transplantation, all three HSPC sources showed a shift in the CD34+ subset towards CD45RA+ progenitors along with a complete loss of the CD45RA-CD49f+ long-term HSC subpopulation, suggesting this model promotes mainly short-term HSC activity. Mice transplanted with cord blood HSPCs maintained a diversified human immune compartment for at least 36 weeks after the primary transplant, although mice given adult bone marrow HSPCs had lost diversity and contained only myeloid cells by this time point. Finally, to assess the impact of non-HSPCs on transplantation outcome, we also tested mice transplanted with total or T cell-depleted adult bone marrow mononuclear cells. Total bone marrow mononuclear cell transplants produced significantly lower human chimerism compared to purified HSPCs, and T-depletion rescued B cell levels but not other lineages. Together these results reveal marked differences in engraftment efficiency and lineage commitment according to HSPC source and suggest that T cells and other non-HSPC populations affect lineage output even in the absence of conditioning-associated inflammation.


Asunto(s)
Linaje de la Célula , Trasplante de Células Madre de Sangre del Cordón Umbilical , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/metabolismo , Huésped Inmunocomprometido/genética , Mutación , Proteínas Proto-Oncogénicas c-kit/genética , Animales , Antígenos CD34/metabolismo , Linfocitos B/inmunología , Linfocitos B/metabolismo , Supervivencia Celular , Células Cultivadas , Femenino , Movilización de Célula Madre Hematopoyética , Células Madre Hematopoyéticas/inmunología , Humanos , Integrina alfa6/metabolismo , Antígenos Comunes de Leucocito/metabolismo , Masculino , Ratones Mutantes , Trasplante de Células Madre de Sangre Periférica , Fenotipo , Linfocitos T/inmunología , Linfocitos T/metabolismo , Factores de Tiempo , Quimera por Trasplante
16.
JCI Insight ; 2(13)2017 Jul 06.
Artículo en Inglés | MEDLINE | ID: mdl-28679955

RESUMEN

A central issue for adoptive cellular immunotherapy is overcoming immunosuppressive signals to achieve tumor clearance. While γδ T cells are known to be potent cytolytic effectors that can kill a variety of cancers, it is not clear whether they are inhibited by suppressive ligands expressed in tumor microenvironments. Here, we have used a powerful preclinical model where EBV infection drives the de novo generation of human B cell lymphomas in vivo, and autologous T lymphocytes are held in check by PD-1/CTLA-4-mediated inhibition. We show that a single dose of adoptively transferred Vδ2+ T cells has potent antitumor effects, even in the absence of checkpoint blockade or activating compounds. Vδ2+ T cell immunotherapy given within the first 5 days of EBV infection almost completely prevented the outgrowth of tumors. Vδ2+ T cell immunotherapy given more than 3 weeks after infection (after neoplastic transformation is evident) resulted in a dramatic reduction in tumor burden. The immunotherapeutic Vδ2+ T cells maintained low cell surface expression of PD-1 in vivo, and their recruitment to tumors was followed by a decrease in B cells expressing PD-L1 and PD-L2 inhibitory ligands. These results suggest that adoptively transferred PD-1lo Vδ2+ T cells circumvent the tumor checkpoint environment in vivo.

17.
Curr Opin Virol ; 9: 178-87, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25462451

RESUMEN

Human cytomegalovirus (HCMV), the prototypical human ß-herpesvirus, encodes approximately 40 known gene products that function to subvert our host defense mechanisms. From HCMV, we have learned about interferon signaling, cytokine function, chemokine signaling, natural killer (NK) cells' cytotoxicity toward tumors and virus-infected cells, antigen processing and presentation, and protective initiation of the apoptotic signaling cascade. With each successive discovery of novel host evasion mechanism encoded by the cytomegaloviruses, we illuminate what these herpesviruses have learned over the course of their 100 MYr-long evolution with their hosts. As much as we have learned from HCMV, the other members of the human ß-herpesvirus family, HHV-6 and HHV-7, are closely-related and yet largely unexplored. These viruses likely have much yet to teach us.


Asunto(s)
Herpesvirus Humano 6/inmunología , Herpesvirus Humano 6/fisiología , Herpesvirus Humano 7/inmunología , Herpesvirus Humano 7/fisiología , Interacciones Huésped-Patógeno , Evasión Inmune , Humanos
18.
PLoS One ; 9(6): e99139, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24901711

RESUMEN

The human herpesvirus-7 (HHV-7) U21 gene product binds to class I major histocompatibility complex (MHC) molecules and reroutes them to a lysosomal compartment. Trafficking of integral membrane proteins to lysosomes is mediated through cytoplasmic sorting signals that recruit heterotetrameric clathrin adaptor protein (AP) complexes, which in turn mediate protein sorting in post-Golgi vesicular transport. Since U21 can mediate rerouting of class I molecules to lysosomes even when lacking its cytoplasmic tail, we hypothesize the existence of a cellular protein that contains the lysosomal sorting information required to escort class I molecules to the lysosomal compartment. If such a protein exists, we expect that it might recruit clathrin adaptor protein complexes as a means of lysosomal sorting. Here we describe experiments demonstrating that the µ adaptins from AP-1 and AP-3 are involved in U21-mediated trafficking of class I molecules to lysosomes. These experiments support the idea that a cellular protein(s) is necessary for U21-mediated lysosomal sorting of class I molecules. We also examine the impact of transient versus chronic knockdown of these adaptor protein complexes, and show that the few remaining µ subunits in the cells are eventually able to reroute class I molecules to lysosomes.


Asunto(s)
Complejo 1 de Proteína Adaptadora/metabolismo , Complejo 3 de Proteína Adaptadora/metabolismo , Proteínas Portadoras/metabolismo , Herpesvirus Humano 7/metabolismo , Antígenos de Histocompatibilidad Clase I/metabolismo , Lisosomas/metabolismo , Proteínas Virales/metabolismo , Complejo 1 de Proteína Adaptadora/antagonistas & inhibidores , Complejo 1 de Proteína Adaptadora/genética , Complejo 2 de Proteína Adaptadora/antagonistas & inhibidores , Complejo 2 de Proteína Adaptadora/genética , Complejo 2 de Proteína Adaptadora/metabolismo , Complejo 3 de Proteína Adaptadora/antagonistas & inhibidores , Complejo 3 de Proteína Adaptadora/genética , Subunidades mu de Complejo de Proteína Adaptadora/metabolismo , Línea Celular , Membrana Celular/metabolismo , Células HEK293 , Humanos , Muromegalovirus/metabolismo , Transporte de Proteínas , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Proteínas del Envoltorio Viral/metabolismo
19.
Virology ; 365(1): 125-35, 2007 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-17467766

RESUMEN

Like all other members of the herpesvirus family, the closely related human herpesviruses-6 and -7 (HHV-6,7) persist in their host throughout life. In so doing, without exception, every member of the herpesvirus family has evolved mechanisms to avoid detection by the immune system. In particular, human cytomegalovirus (HCMV), mouse cytomegalovirus (MCMV), human herpesvirus-8 (HHV-8), and herpes simplex virus (HSV) all encode multiple proteins that interfere with proper MHC class I antigen presentation. The mechanisms employed by these viruses to effect removal of MHC class I from the cell surface vary. The U21 open reading frame from HHV-7 diverts class I MHC molecules to an endolysosomal compartment using an as-yet unknown mechanism. The two variants of HHV-6, HHV-6A and -6B, both possess a U21 open reading frame which contain only approximately 30% amino acid identity to the U21 sequence from HHV-7. Here we describe the characterization of the U21 gene products from HHV-6A and HHV-6B. Like HHV-7 U21, both of the HHV-6 U21 molecules bind to and divert class I MHC molecules to an endolysosomal compartment, effectively removing them from the cell surface, and providing a possible means of escape from immune detection.


Asunto(s)
Herpesvirus Humano 6/metabolismo , Antígenos de Histocompatibilidad Clase I/metabolismo , Proteínas Virales/fisiología , Secuencia de Aminoácidos , Proteínas Portadoras/química , Proteínas Portadoras/farmacología , Línea Celular , Regulación hacia Abajo/efectos de los fármacos , Herpesvirus Humano 6/genética , Antígenos de Histocompatibilidad Clase I/química , Humanos , Proteínas Virales/química , Proteínas Virales/farmacología
20.
J Biol Chem ; 279(39): 41208-17, 2004 Sep 24.
Artículo en Inglés | MEDLINE | ID: mdl-15263005

RESUMEN

Angelman syndrome is a severe neurological disorder characterized by mental retardation, absent speech, ataxia, seizures, and hyperactivity. The gene affected in this disorder is UBE3A, the gene encoding the E6-associated protein (E6AP) ubiquitin-protein ligase. Most patients have chromosomal deletions that remove the entire maternal allele of UBE3A. However, a small subset of patients have E6AP point mutations that result in single amino acid changes or short in-frame deletions that still allow translation of a full-length protein. By studying these point mutations in E6AP, we found a strong correlation between Angelman-associated mutations and a loss of E3 ubiquitin ligase activity. Interestingly the point mutations affect E6AP activity in different ways. Some mutant proteins cannot form thiol ester intermediates with ubiquitin, others retain the thiol ester formation activity but cannot efficiently transfer ubiquitin to a substrate, and still others are unstable in cells. Our results suggest that the loss of E6AP catalytic activity and likely the improper regulation of E6AP substrate(s) are important in the development of Angelman syndrome.


Asunto(s)
Síndrome de Angelman/genética , Mutación , Ubiquitina-Proteína Ligasas/química , Ubiquitina-Proteína Ligasas/genética , Animales , Unión Competitiva , Catálisis , Cristalografía por Rayos X , Escherichia coli/metabolismo , Ésteres/metabolismo , Fibroblastos/metabolismo , Eliminación de Gen , Glutatión Transferasa/metabolismo , Humanos , Ratones , Plásmidos/metabolismo , Mutación Puntual , Unión Proteica , Conformación Proteica , Estructura Terciaria de Proteína , Compuestos de Sulfhidrilo/metabolismo , Factores de Tiempo , Transgenes , Técnicas del Sistema de Dos Híbridos , Ubiquitina/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA