Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 87
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 184(12): 3205-3221.e24, 2021 06 10.
Artículo en Inglés | MEDLINE | ID: mdl-34015271

RESUMEN

Monoclonal antibodies (mAbs) are a focus in vaccine and therapeutic design to counteract severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and its variants. Here, we combined B cell sorting with single-cell VDJ and RNA sequencing (RNA-seq) and mAb structures to characterize B cell responses against SARS-CoV-2. We show that the SARS-CoV-2-specific B cell repertoire consists of transcriptionally distinct B cell populations with cells producing potently neutralizing antibodies (nAbs) localized in two clusters that resemble memory and activated B cells. Cryo-electron microscopy structures of selected nAbs from these two clusters complexed with SARS-CoV-2 spike trimers show recognition of various receptor-binding domain (RBD) epitopes. One of these mAbs, BG10-19, locks the spike trimer in a closed conformation to potently neutralize SARS-CoV-2, the recently arising mutants B.1.1.7 and B.1.351, and SARS-CoV and cross-reacts with heterologous RBDs. Together, our results characterize transcriptional differences among SARS-CoV-2-specific B cells and uncover cross-neutralizing Ab targets that will inform immunogen and therapeutic design against coronaviruses.


Asunto(s)
Anticuerpos Neutralizantes/inmunología , Linfocitos B/metabolismo , SARS-CoV-2/inmunología , Glicoproteína de la Espiga del Coronavirus/inmunología , Anticuerpos Monoclonales/química , Anticuerpos Monoclonales/inmunología , Anticuerpos Neutralizantes/sangre , Anticuerpos Neutralizantes/química , Anticuerpos Antivirales/sangre , Anticuerpos Antivirales/química , Anticuerpos Antivirales/inmunología , Complejo Antígeno-Anticuerpo/química , Complejo Antígeno-Anticuerpo/metabolismo , Reacciones Antígeno-Anticuerpo , Linfocitos B/citología , Linfocitos B/virología , COVID-19/patología , COVID-19/virología , Microscopía por Crioelectrón , Cristalografía por Rayos X , Perfilación de la Expresión Génica , Humanos , Inmunoglobulina A/inmunología , Región Variable de Inmunoglobulina/química , Región Variable de Inmunoglobulina/genética , Dominios Proteicos/inmunología , Multimerización de Proteína , Estructura Cuaternaria de Proteína , SARS-CoV-2/aislamiento & purificación , SARS-CoV-2/metabolismo , Análisis de Secuencia de ARN , Glicoproteína de la Espiga del Coronavirus/química , Glicoproteína de la Espiga del Coronavirus/genética , Glicoproteína de la Espiga del Coronavirus/metabolismo
2.
Nat Immunol ; 16(6): 546-53, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25988886

RESUMEN

To replicate in their hosts, viruses have to navigate the complexities of the mammalian cell, co-opting mechanisms of cellular physiology while defeating restriction factors that are dedicated to halting their progression. Primate lentiviruses devote a relatively large portion of their coding capacity to counteracting restriction factors by encoding accessory proteins dedicated to neutralizing the antiviral function of these intracellular inhibitors. Research into the roles of the accessory proteins has revealed the existence of previously undetected intrinsic defenses, provided insight into the evolution of primate lentiviruses as they adapt to new species and uncovered new targets for the development of therapeutics. This Review discusses the biology of the restriction factors APOBEC3, SAMHD1 and tetherin and the viral accessory proteins that counteract them.


Asunto(s)
Antígenos CD/metabolismo , Citosina Desaminasa/metabolismo , Infecciones por VIH/inmunología , VIH-1/fisiología , Especificidad del Huésped , Evasión Inmune , Proteínas de Unión al GTP Monoméricas/metabolismo , Proteínas Reguladoras y Accesorias Virales/metabolismo , Desaminasas APOBEC , Animales , Evolución Biológica , Citidina Desaminasa , Proteínas Ligadas a GPI/metabolismo , Infecciones por VIH/virología , Humanos , Terapia Molecular Dirigida , Proteína 1 que Contiene Dominios SAM y HD
3.
Proc Natl Acad Sci U S A ; 120(23): e2303509120, 2023 06 06.
Artículo en Inglés | MEDLINE | ID: mdl-37252952

RESUMEN

Vectored immunoprophylaxis was first developed as a means of establishing engineered immunity to HIV using an adenoassociated viral vector expressing a broadly neutralizing antibody. We applied this concept to establish long-term prophylaxis against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in a mouse model using adenoassociated virus and lentiviral vectors expressing a high-affinity angiotensin-converting enzyme 2 (ACE2) decoy. Administration of decoy-expressing (adenoassociated virus) AAV2.retro and AAV6.2 vectors by intranasal instillation or intramuscular injection protected mice against high-titered SARS-CoV-2 infection. AAV and lentiviral vectored immunoprophylaxis was durable and was active against SARS-CoV-2 Omicron subvariants. The AAV vectors were also effective therapeutically when administered postinfection. Vectored immunoprophylaxis could be of value for immunocompromised individuals for whom vaccination is not practical and as a means to rapidly establish protection from infection. Unlike monoclonal antibody therapy, the approach is expected to remain active despite continued evolution viral variants.


Asunto(s)
COVID-19 , Animales , Ratones , COVID-19/prevención & control , SARS-CoV-2/genética , Inmunización , Inmunoterapia , Vacunación , Dependovirus/genética , Glicoproteína de la Espiga del Coronavirus , Anticuerpos Neutralizantes/uso terapéutico , Anticuerpos Antivirales/uso terapéutico
4.
PLoS Biol ; 20(3): e3001592, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-35358182

RESUMEN

Gastrointestinal effects associated with Coronavirus Disease 2019 (COVID-19) are highly variable for reasons that are not understood. In this study, we used intestinal organoid-derived cultures differentiated from primary human specimens as a model to examine interindividual variability. Infection of intestinal organoids derived from different donors with Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) resulted in orders of magnitude differences in virus replication in small intestinal and colonic organoid-derived monolayers. Susceptibility to infection correlated with angiotensin I converting enzyme 2 (ACE2) expression level and was independent of donor demographic or clinical features. ACE2 transcript levels in cell culture matched the amount of ACE2 in primary tissue, indicating that this feature of the intestinal epithelium is retained in the organoids. Longitudinal transcriptomics of organoid-derived monolayers identified a delayed yet robust interferon signature, the magnitude of which corresponded to the degree of SARS-CoV-2 infection. Interestingly, virus with the Omicron variant spike (S) protein infected the organoids with the highest infectivity, suggesting increased tropism of the virus for intestinal tissue. These results suggest that heterogeneity in SARS-CoV-2 replication in intestinal tissues results from differences in ACE2 levels, which may underlie variable patient outcomes.


Asunto(s)
COVID-19 , Enzima Convertidora de Angiotensina 2/genética , Humanos , Organoides , SARS-CoV-2
5.
Mol Ther ; 32(1): 124-139, 2024 Jan 03.
Artículo en Inglés | MEDLINE | ID: mdl-37990494

RESUMEN

Quiescent human hematopoietic stem cells (HSC) are ideal targets for gene therapy applications due to their preserved stemness and repopulation capacities; however, they have not been exploited extensively because of their resistance to genetic manipulation. We report here the development of a lentiviral transduction protocol that overcomes this resistance in long-term repopulating quiescent HSC, allowing their efficient genetic manipulation. Mechanistically, lentiviral vector transduction of quiescent HSC was found to be restricted at the level of vector entry and by limited pyrimidine pools. These restrictions were overcome by the combined addition of cyclosporin H (CsH) and deoxynucleosides (dNs) during lentiviral vector transduction. Clinically relevant transduction levels were paired with higher polyclonal engraftment of long-term repopulating HSC as compared with standard ex vivo cultured controls. These findings identify the cell-intrinsic barriers that restrict the transduction of quiescent HSC and provide a means to overcome them, paving the way for the genetic engineering of unstimulated HSC.


Asunto(s)
Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas , Humanos , Transducción Genética , Lentivirus/genética , Terapia Genética/métodos , Inmunidad Innata , Vectores Genéticos/genética , Antígenos CD34
6.
Nat Immunol ; 13(3): 223-228, 2012 Feb 12.
Artículo en Inglés | MEDLINE | ID: mdl-22327569

RESUMEN

SAMHD1 restricts the infection of dendritic and other myeloid cells by human immunodeficiency virus type 1 (HIV-1), but in lentiviruses of the simian immunodeficiency virus of sooty mangabey (SIVsm)-HIV-2 lineage, SAMHD1 is counteracted by the virion-packaged accessory protein Vpx. Here we found that SAMHD1 restricted infection by hydrolyzing intracellular deoxynucleoside triphosphates (dNTPs), lowering their concentrations to below those required for the synthesis of the viral DNA by reverse transcriptase (RT). SAMHD1-mediated restriction was alleviated by the addition of exogenous deoxynucleosides. An HIV-1 with a mutant RT with low affinity for dNTPs was particularly sensitive to SAMHD1-mediated restriction. Vpx prevented the SAMHD1-mediated decrease in dNTP concentration and induced the degradation of human and rhesus macaque SAMHD1 but had no effect on mouse SAMHD1. Nucleotide-pool depletion could be a general mechanism for protecting cells from infectious agents that replicate through a DNA intermediate.


Asunto(s)
VIH-1/fisiología , Proteínas de Unión al GTP Monoméricas/metabolismo , Nucleótidos/metabolismo , Replicación Viral , Animales , Línea Celular , Humanos , Espacio Intracelular/metabolismo , Macaca mulatta , Macrófagos/inmunología , Ratones , Proteínas de Unión al GTP Monoméricas/genética , Proteínas de Unión al GTP Monoméricas/inmunología , Proteína 1 que Contiene Dominios SAM y HD
7.
Biochem Eng J ; 187: 108596, 2022 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-36034180

RESUMEN

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has caused a global pandemic since December 2019, and with it, a push for innovations in rapid testing and neutralizing antibody treatments in an effort to solve the spread and fatality of the disease. One such solution to both of these prevailing issues is targeting the interaction of SARS-CoV-2 spike receptor binding domain (RBD) with the human angiotensin-converting enzyme 2 (ACE2) receptor protein. Structural studies have shown that the N-terminal alpha-helix comprised of the first 23 residues of ACE2 plays an important role in this interaction. Where it is typical to design a binding domain to fit a target, we have engineered a protein that relies on multivalency rather than the sensitivity of a monomeric ligand to provide avidity to its target by fusing the N-terminal helix of ACE2 to the coiled-coil domain of the cartilage oligomeric matrix protein. The resulting ACE-MAP is able to bind to the SARS-CoV-2 RBD with improved binding affinity, is expressible in E. coli, and is thermally stable and relatively small (62 kDa). These properties suggest ACE-MAP and the MAP scaffold to be a promising route towards developing future diagnostics and therapeutics to SARS-CoV-2.

8.
Mol Ther ; 28(8): 1795-1805, 2020 08 05.
Artículo en Inglés | MEDLINE | ID: mdl-32497512

RESUMEN

Dendritic cell vaccines are a promising strategy for the treatment of cancer and infectious diseases but have met with mixed success. We report on a lentiviral vector-based dendritic cell vaccine strategy that generates a cluster of differentiation 8 (CD8) T cell response that is much stronger than that achieved by standard peptide-pulsing approaches. The strategy was tested in the mouse lymphocytic choriomeningitis virus (LCMV) model. Bone marrow-derived dendritic cells from SAMHD1 knockout mice were transduced with a lentiviral vector expressing the GP33 major-histocompatibility-complex (MHC)-class-I-restricted peptide epitope and CD40 ligand (CD40L) and injected into wild-type mice. The mice were highly protected against acute and chronic variant CL-13 LCMVs, resulting in a 100-fold greater decrease than that achieved with peptide epitope-pulsed dendritic cells. Inclusion of an MHC-class-II-restricted epitope in the lentiviral vector further increased the CD8 T cell response and resulted in antigen-specific CD8 T cells that exhibited a phenotype associated with functional cytotoxic T cells. The vaccination synergized with checkpoint blockade to reduce the viral load of mice chronically infected with CL-13 to an undetectable level. The strategy improves upon current dendritic cell vaccine strategies; is applicable to the treatment of disease, including AIDS and cancer; and supports the utility of Vpx-containing vectors.


Asunto(s)
Células Dendríticas/efectos de los fármacos , Células Dendríticas/inmunología , Vectores Genéticos , Inhibidores de Puntos de Control Inmunológico/farmacología , Lentivirus , Vacunas Virales/inmunología , Virosis/prevención & control , Animales , Biomarcadores , Células Dendríticas/virología , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades , Vectores Genéticos/administración & dosificación , Vectores Genéticos/genética , Antígenos de Histocompatibilidad Clase II , Interacciones Huésped-Patógeno/inmunología , Humanos , Lentivirus/genética , Coriomeningitis Linfocítica/prevención & control , Virus de la Coriomeningitis Linfocítica/inmunología , Ratones , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Vacunas Virales/administración & dosificación , Virosis/etiología , Virosis/inmunología
9.
Mol Ther ; 27(5): 960-973, 2019 05 08.
Artículo en Inglés | MEDLINE | ID: mdl-30962161

RESUMEN

HIV-1-infected individuals are treated with lifelong antiretroviral drugs to control the infection. A means to strengthen the antiviral T cell response might allow them to control viral loads without antiretroviral drugs. We report the development of a lentiviral vector-based dendritic cell (DC) vaccine in which HIV-1 antigen is co-expressed with CD40 ligand (CD40L) and a soluble, high-affinity programmed cell death 1 (PD-1) dimer. CD40L activates the DCs, whereas PD-1 binds programmed death ligand 1 (PD-L1) to prevent checkpoint activation and strengthen the cytotoxic T lymphocyte (CTL) response. The injection of humanized mice with DCs transduced with vector expressing CD40L and the HIV-1 SL9 epitope induced antigen-specific T cell proliferation and memory differentiation. Upon HIV-1 challenge of vaccinated mice, viral load was suppressed by 2 logs for 6 weeks. Introduction of the soluble PD-1 dimer into a vector that expressed full-length HIV-1 proteins accelerated the antiviral response. The results support development of this approach as a therapeutic vaccine that might allow HIV-1-infected individuals to control virus replication without antiretroviral therapy.


Asunto(s)
Células Dendríticas/inmunología , Infecciones por VIH/terapia , Receptor de Muerte Celular Programada 1/inmunología , Linfocitos T Citotóxicos/inmunología , Replicación Viral/inmunología , Vacunas contra el SIDA/inmunología , Vacunas contra el SIDA/farmacología , Animales , Ligando de CD40 , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/virología , Epítopos de Linfocito T/inmunología , Vectores Genéticos/inmunología , Vectores Genéticos/farmacología , Infecciones por VIH/inmunología , Infecciones por VIH/virología , VIH-1/inmunología , VIH-1/patogenicidad , Humanos , Activación de Linfocitos/inmunología , Ratones
10.
Nature ; 493(7430): 51-5, 2013 Jan 03.
Artículo en Inglés | MEDLINE | ID: mdl-23235831

RESUMEN

Pore-forming toxins are critical virulence factors for many bacterial pathogens and are central to Staphylococcus aureus-mediated killing of host cells. S. aureus encodes pore-forming bi-component leukotoxins that are toxic towards neutrophils, but also specifically target other immune cells. Despite decades since the first description of staphylococcal leukocidal activity, the host factors responsible for the selectivity of leukotoxins towards different immune cells remain unknown. Here we identify the human immunodeficiency virus (HIV) co-receptor CCR5 as a cellular determinant required for cytotoxic targeting of subsets of myeloid cells and T lymphocytes by the S. aureus leukotoxin ED (LukED). We further demonstrate that LukED-dependent cell killing is blocked by CCR5 receptor antagonists, including the HIV drug maraviroc. Remarkably, CCR5-deficient mice are largely resistant to lethal S. aureus infection, highlighting the importance of CCR5 targeting in S. aureus pathogenesis. Thus, depletion of CCR5(+) leukocytes by LukED suggests a new immune evasion mechanism of S. aureus that can be therapeutically targeted.


Asunto(s)
Toxinas Bacterianas/metabolismo , Exotoxinas/metabolismo , Receptores CCR5/metabolismo , Staphylococcus aureus/patogenicidad , Animales , Antagonistas de los Receptores CCR5 , Muerte Celular , Células Cultivadas , Células Dendríticas/citología , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Femenino , Humanos , Evasión Inmune , Memoria Inmunológica , Células Jurkat , Ratones , Células Mieloides/citología , Células Mieloides/inmunología , Células Mieloides/metabolismo , Staphylococcus aureus/inmunología , Linfocitos T/citología , Linfocitos T/inmunología , Linfocitos T/metabolismo
11.
J Biol Chem ; 292(3): 1068-1080, 2017 01 20.
Artículo en Inglés | MEDLINE | ID: mdl-27920203

RESUMEN

The triphosphohydrolase SAMHD1 (sterile α motif and histidine-aspartate domain-containing protein 1) restricts HIV-1 replication in nondividing myeloid cells by depleting the dNTP pool, preventing reverse transcription. SAMHD1 is also reported to have ribonuclease activity that degrades the virus genomic RNA. Human SAMHD1 is regulated by phosphorylation of its carboxyl terminus at Thr-592, which abrogates its antiviral function yet has only a small effect on its phosphohydrolase activity. In the mouse, SAMHD1 is expressed as two isoforms (ISF1 and ISF2) that differ at the carboxyl terminus due to alternative splicing of the last coding exon. In this study we characterized the biochemical and antiviral properties of the two mouse isoforms of SAMHD1. Both are antiviral in nondividing cells. Mass spectrometry analysis showed that SAMHD1 is phosphorylated at several amino acid residues, one of which (Thr-634) is homologous to Thr-592. Phosphomimetic mutation at Thr-634 of ISF1 ablates its antiviral activity yet has little effect on phosphohydrolase activity in vitro dGTP caused ISF1 to tetramerize, activating its catalytic activity. In contrast, ISF2, which lacks the phosphorylation site, was significantly more active, tetramerized, and was active without added dGTP. Neither isoform nor human SAMHD1 had detectable RNase activity in vitro or affected HIV-1 genomic RNA stability in newly infected cells. These data support a model in which SAMHD1 catalytic activity is regulated through tetramer stabilization by the carboxyl-terminal tail, phosphorylation destabilizing the complexes and inactivating the enzyme. ISF2 may serve to reduce the dNTP pool to very low levels as a means of restricting virus replication.


Asunto(s)
Infecciones por VIH/enzimología , VIH-1/fisiología , Proteínas de Unión al GTP Monoméricas/metabolismo , Multimerización de Proteína , ARN Viral/metabolismo , Replicación Viral/fisiología , Sustitución de Aminoácidos , Animales , Infecciones por VIH/genética , Humanos , Isoenzimas/química , Isoenzimas/genética , Isoenzimas/metabolismo , Ratones , Modelos Moleculares , Proteínas de Unión al GTP Monoméricas/química , Proteínas de Unión al GTP Monoméricas/genética , Mutación Missense , Fosforilación , ARN Viral/genética , Proteína 1 que Contiene Dominios SAM y HD , Células U937
12.
Retrovirology ; 13(1): 83, 2016 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-27905985

RESUMEN

BACKGROUND: Monocytes, the primary myeloid cell-type in peripheral blood, are resistant to HIV-1 infection as a result of the lentiviral restriction factor SAMHD1. Toll-like receptors recognize microbial pathogen components, inducing the expression of antiviral host proteins and proinflammatory cytokines. TLR agonists that mimic microbial ligands have been found to have activity against HIV-1 in macrophages. The induction of restriction factors in monocytes by TLR agonist activation has not been well studied. To analyze restriction factor induction by TLR activation in monocytes, we used the imidazoquinoline TLR7/8 agonist R848 and infected with HIV-1 reporter virus that contained packaged viral accessory protein Vpx, which allows the virus to escape SAMHD1-mediated restriction.  RESULTS: R848 prevented the replication of Vpx-containing HIV-1 and HIV-2 in peripheral blood mononuclear cells and monocytes. The block was post-entry but prior to reverse transcription of the viral genomic RNA. The restriction was associated with destabilization of the genomic RNA molecules of the in-coming virus particle. R848 treatment of activated T cells did not protect them from infection but treated monocytes produced high levels of proinflammatory cytokines, including type-I IFN that protected bystander activated T cells from infection. CONCLUSION: The activation of TLR7/8 induces two independent restrictions to HIV-1 replication in monocytes: a cell-intrinsic block that acts post-entry to prevent reverse transcription; and a cell-extrinsic block, in which monocytes produce high levels of proinflammatory cytokines (primarily type-I IFN) that protects bystander monocytes and T lymphocytes. The cell-intrinsic block may result from the induction of a novel restriction factor, which can be termed Lv5 and acts by destabilizing the in-coming viral genomic RNA, either by the induction of a host ribonuclease or by disrupting the viral capsid. TLR agonists are being developed for therapeutic use to diminish the size of the latent provirus reservoir in HIV-1 infected individuals. Such drugs may both induce latent provirus expression and restrict virus replication during treatment.


Asunto(s)
Fármacos Anti-VIH/farmacología , VIH-1/efectos de los fármacos , Imidazoles/farmacología , Monocitos/efectos de los fármacos , Monocitos/virología , Línea Celular , Células HEK293 , VIH-1/fisiología , Humanos , Interferón Tipo I/biosíntesis , Leucocitos Mononucleares/efectos de los fármacos , Leucocitos Mononucleares/virología , Activación de Linfocitos/efectos de los fármacos , Monocitos/inmunología , Proteínas de Unión al GTP Monoméricas/genética , ARN Viral , Transcripción Reversa , Proteína 1 que Contiene Dominios SAM y HD , Receptor Toll-Like 7/agonistas , Receptor Toll-Like 8/agonistas , Replicación Viral/efectos de los fármacos
13.
J Virol ; 89(10): 5701-13, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25762741

RESUMEN

UNLABELLED: Sterile alpha motif domain and HD domain-containing protein 1 (SAMHD1) restricts human immunodeficiency virus type 1 (HIV-1) replication in myeloid and resting T cells. Lentiviruses such as HIV-2 and some simian immunodeficiency viruses (SIVs) counteract the restriction by encoding Vpx or Vpr, accessory proteins that are packaged in virions and which, upon entry of the virus into the cytoplasm, induce the proteasomal degradation of SAMHD1. As a tool to study these mechanisms, we generated HeLa cell lines that express a fusion protein termed NLS.GFP.SAM595 in which the Vpx binding domain of SAMHD1 is fused to the carboxy terminus of green fluorescent protein (GFP) and a nuclear localization signal is fused to the amino terminus of GFP. Upon incubation of Vpx-containing virions with the cells, the NLS.GFP.SAM595 fusion protein was degraded over several hours and the levels remained low over 5 days as the result of continued targeting of the CRL4 E3 ubiquitin ligase. Degradation of the fusion protein required that it contain a nuclear localization sequence. Fusion to the cytoplasmic protein muNS rendered the protein resistant to Vpx-mediated degradation, confirming that SAMHD1 is targeted in the nucleus. Virions treated with protease inhibitors failed to release Vpx, indicating that Gag processing was required for Vpx release from the virion. Mutations in the capsid protein that altered the kinetics of virus uncoating and the Gag binding drug PF74 had no effect on the Vpx-mediated degradation. These results suggest that Vpx is released from virions without a need for uncoating of the capsid, allowing Vpx to transit to the nucleus rapidly upon entry into the cytoplasm. IMPORTANCE: SAMHD1 restricts lentiviral replication in myeloid cells and resting T cells. Its importance is highlighted by the fact that viruses such as HIV-2 encode an accessory protein that is packaged in the virion and is dedicated to inducing SAMHD1 degradation. Vpx needs to act rapidly upon infection to allow reverse transcription to proceed. The limited number of Vpx molecules in a virion also needs to clear the cell of SAMHD1 over a prolonged period of time. Using an engineered HeLa cell line that expresses a green fluorescent protein (GFP)-SAMHD1 fusion protein, we showed that the Vpx-dependent degradation occurs without a need for viral capsid uncoating. In addition, the fusion protein was degraded only when it was localized to the nucleus, confirming that SAMHD1 is targeted in the nucleus and thus explaining why Vpx also localizes to the nucleus.


Asunto(s)
VIH-2/fisiología , VIH-2/patogenicidad , Proteínas de Unión al GTP Monoméricas/metabolismo , Proteínas Reguladoras y Accesorias Virales/metabolismo , Desencapsidación Viral/fisiología , Proteínas de la Cápside/genética , Proteínas de la Cápside/fisiología , Células HEK293 , VIH-1/genética , VIH-1/fisiología , VIH-2/genética , Células HeLa , Interacciones Huésped-Patógeno , Humanos , Proteínas de Unión al GTP Monoméricas/genética , Mutación , Procesamiento Proteico-Postraduccional , Proteolisis , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Proteína 1 que Contiene Dominios SAM y HD , Virus de la Inmunodeficiencia de los Simios/genética , Virus de la Inmunodeficiencia de los Simios/fisiología , Proteínas Reguladoras y Accesorias Virales/genética , Replicación Viral , Productos del Gen gag del Virus de la Inmunodeficiencia Humana/metabolismo
14.
Antimicrob Agents Chemother ; 58(8): 4915-9, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24867973

RESUMEN

Sterile alpha motif- and histidine/aspartic acid domain-containing protein 1 (SAMHD1) limits HIV-1 replication by hydrolyzing deoxynucleoside triphosphates (dNTPs) necessary for reverse transcription. Nucleoside reverse transcriptase inhibitors (NRTIs) are components of anti-HIV therapies. We report here that SAMHD1 cleaves NRTI triphosphates (TPs) at significantly lower rates than dNTPs and that SAMHD1 depletion from monocytic cells affects the susceptibility of HIV-1 infections to NRTIs in complex ways that depend not only on the relative changes in dNTP and NRTI-TP concentrations but also on the NRTI activation pathways.


Asunto(s)
Didesoxinucleótidos/metabolismo , Transcriptasa Inversa del VIH/antagonistas & inhibidores , VIH-1/efectos de los fármacos , Proteínas de Unión al GTP Monoméricas/metabolismo , Inhibidores de la Transcriptasa Inversa/farmacología , Adenina/análogos & derivados , Adenina/farmacología , Línea Celular , Expresión Génica , Genes Reporteros , Transcriptasa Inversa del VIH/genética , Transcriptasa Inversa del VIH/metabolismo , VIH-1/enzimología , Interacciones Huésped-Patógeno , Humanos , Lamivudine/farmacología , Luciferasas/genética , Luciferasas/metabolismo , Monocitos/efectos de los fármacos , Monocitos/metabolismo , Monocitos/virología , Proteínas de Unión al GTP Monoméricas/antagonistas & inhibidores , Proteínas de Unión al GTP Monoméricas/genética , Organofosfonatos/farmacología , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Proteína 1 que Contiene Dominios SAM y HD , Estavudina/farmacología , Tenofovir , Replicación Viral/efectos de los fármacos , Zidovudina/farmacología
15.
J Virol ; 87(21): 11741-50, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23986575

RESUMEN

The deoxynucleoside triphosphohydrolase SAMHD1 restricts retroviral replication in myeloid cells. Human immunodeficiency virus type 2 (HIV-2) and a simian immunodeficiency virus from rhesus macaques (SIVmac) encode Vpx, a virion-packaged accessory protein that counteracts SAMHD1 by inducing its degradation. SAMHD1 is thought to work by depleting the pool of intracellular deoxynucleoside triphosphates but has also been reported to have exonuclease activity that could allow it to degrade the viral genomic RNA or viral reverse-transcribed DNA. To induce the degradation of SAMHD1, Vpx co-opts the cullin4a-based E3 ubiquitin ligase, CRL4. E3 ubiquitin ligases are regulated by the covalent attachment of the ubiquitin-like protein Nedd8 to the cullin subunit. Neddylation can be prevented by MLN4924, a drug that inhibits the nedd8-activating enzyme. We report that MLN4924 inhibits the neddylation of CRL4, blocking Vpx-induced degradation of SAMHD1 and maintaining the restriction. Removal of the drug several hours postinfection released the block. Similarly, Vpx-containing virus-like particles and deoxynucleosides added to the cells more than 24 h postinfection released the SAMHD1-mediated block. Taken together, these findings support deoxynucleoside triphosphate pool depletion as the primary mechanism of SAMHD1 restriction and argue against a nucleolytic mechanism, which would not be reversible.


Asunto(s)
Proteínas Cullin/metabolismo , VIH-1/inmunología , Proteínas de Unión al GTP Monoméricas/metabolismo , Ubiquitinas/metabolismo , Replicación Viral , Línea Celular , Proteínas Cullin/inmunología , Ciclopentanos/metabolismo , Inhibidores Enzimáticos/metabolismo , VIH-1/fisiología , Humanos , Proteínas de Unión al GTP Monoméricas/inmunología , Proteína NEDD8 , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Pirimidinas/metabolismo , Proteína 1 que Contiene Dominios SAM y HD , Ubiquitinas/antagonistas & inhibidores
16.
Virol J ; 11: 108, 2014 Jun 09.
Artículo en Inglés | MEDLINE | ID: mdl-24912982

RESUMEN

BACKGROUND: HIV-1 Vpr-mediated G2 cell cycle arrest is dependent on the interaction of Vpr with an E3 ubiquitin ligase that contains damage-specific DNA binding protein 1 (DDB1), Cullin 4A (Cul4A), DDB1 and Cul4-associated factor 1 (DCAF1), and Rbx1. Vpr is thought to associate directly with DCAF1 in the E3 ubiquitin ligase complex although the exact interaction pattern of the proteins in the complex is not completely defined. The Vpr of SIVagm induces G2 arrest of cognate African Green Monkey (AGM) cells but not human cells. The molecular mechanism by which SIVagm Vpr exhibits its species-specific function remained unknown. METHODS: Physical interaction of proteins in the E3 ubiquitin ligase complex was assessed by co-immunoprecipitation followed by western blotting. In addition, co-localization of the proteins in cells was investigated by confocal microscopy. The cell cycle was analyzed by propidium iodide staining and flow cytometry. DNA damage response elicited by Vpr was evaluated by detecting phosphorylation of H2AX, a marker for DNA damage response. RESULTS: We show that RNAi knock-down of DCAF1 prevented the co-immunoprecipitation of DDB1 with HIV-1 Vpr while DDB1 knock-down did not influence the binding of Vpr to DCAF1. HIV-1 Vpr mutants with a L64P or a R90K mutation maintained the ability to associate with DCAF1 but did not appear to be in a complex with DDB1. SIVagm Vpr associated with AGM DCAF1 and DDB1 while, in human cells, it binds to human DCAF1 but hardly binds to human DDB1, resulting in the reduced activation of H2AX. CONCLUSIONS: The identification of Vpr mutants which associate with DCAF1 but only poorly with DDB1 suggests that DCAF1 is necessary but the simple binding of Vpr to DCAF1 is not sufficient for the Vpr association with DDB1-containing E3 ligase complex. Vpr may interact both with DCAF1 and DDB1 in the E3 ligase complex. Alternatively, the interaction of Vpr and DCAF1 may induce a conformational change in DCAF1 or Vpr that promotes the interaction with DDB1. The ability of SIVagm Vpr to associate with DDB1, but not DCAF1, can explain the species-specificity of SIVagm Vpr-mediated G2 arrest.


Asunto(s)
Proteínas Portadoras/metabolismo , Ciclo Celular , Proteínas de Unión al ADN/metabolismo , VIH-1/fisiología , Ubiquitina-Proteína Ligasas/metabolismo , Productos del Gen vpr del Virus de la Inmunodeficiencia Humana/metabolismo , Animales , Western Blotting , Chlorocebus aethiops , Citometría de Flujo , Células HeLa , Humanos , Inmunoprecipitación , Microscopía Confocal , Modelos Biológicos , Proteínas Mutantes/metabolismo , Unión Proteica , Proteínas Serina-Treonina Quinasas
17.
J Immunother Cancer ; 12(4)2024 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-38658032

RESUMEN

BACKGROUND: While immunotherapy has been highly successful for the treatment of some cancers, for others, the immune response to tumor antigens is weak leading to treatment failure. The resistance of tumors to checkpoint inhibitor therapy may be caused by T cell exhaustion resulting from checkpoint activation. METHODS: In this study, lentiviral vectors that expressed T cell epitopes of an experimentally introduced tumor antigen, ovalbumin, or the endogenous tumor antigen, Trp1 were developed. The vectors coexpressed CD40 ligand (CD40L), which served to mature the dendritic cells (DCs), and a soluble programmed cell death protein 1 (PD-1) microbody to prevent checkpoint activation. Vaccination of mice bearing B16.OVA melanomas with vector-transduced DCs induced the proliferation and activation of functional, antigen-specific, cytolytic CD8 T cells. RESULTS: Vaccination induced the expansion of CD8 T cells that infiltrated the tumors to suppress tumor growth. Vector-encoded CD40L and PD-1 microbody increased the extent of tumor growth suppression. Adoptive transfer demonstrated that the effect was mediated by CD8 T cells. Direct injection of the vector, without the need for ex vivo transduction of DCs, was also effective. CONCLUSIONS: This study suggests that therapeutic vaccination that induces tumor antigen-specific CD8 T cells coupled with a vector-expressed checkpoint inhibitor can be an effective means to suppress the growth of tumors that are resistant to conventional immunotherapy.


Asunto(s)
Vacunas contra el Cáncer , Inhibidores de Puntos de Control Inmunológico , Lentivirus , Animales , Ratones , Vacunas contra el Cáncer/inmunología , Vacunas contra el Cáncer/uso terapéutico , Lentivirus/genética , Inhibidores de Puntos de Control Inmunológico/farmacología , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Humanos , Células Dendríticas/inmunología , Modelos Animales de Enfermedad , Linfocitos T CD8-positivos/inmunología , Melanoma Experimental/inmunología , Melanoma Experimental/terapia , Línea Celular Tumoral , Ratones Endogámicos C57BL , Femenino
18.
Retrovirology ; 10: 26, 2013 Mar 05.
Artículo en Inglés | MEDLINE | ID: mdl-23497255

RESUMEN

BACKGROUND: SAMHD1 is a triphosphohydrolase that restricts the replication of HIV-1 and SIV in myeloid cells. In macrophages and dendritic cells, SAMHD1 restricts virus replication by diminishing the deoxynucleotide triphosphate pool to a level below that which supports lentiviral reverse transcription. HIV-2 and related SIVs encode the accessory protein Vpx to induce the proteasomal degradation of SAMHD1 following virus entry. While SAMHD1 has been shown to restrict HIV-1 and SIV, the breadth of its restriction is not known and whether other viruses have a means to counteract the restriction has not been determined. RESULTS: We show that SAMHD1 restricts a wide array of divergent retroviruses, including the alpha, beta and gamma classes. Murine leukemia virus was restricted by SAMHD1 in macrophages yet removal of SAMHD1 did not alleviate the block to infection because of an additional block to viral nuclear import. Prototype foamy virus (PFV) and Human T cell leukemia virus type I (HTLV-1) were the only retroviruses tested that were not restricted by SAMHD1. PFV reverse transcribes predominantly prior to entry and thus is unaffected by the dNTP level in the target cell. It is possible that HTLV-1 has a mechanism to render the virus resistant to SAMHD1-mediated restriction. CONCLUSION: The results suggest that SAMHD1 has broad anti-retroviral activity against which most viruses have not found an escape.


Asunto(s)
Macrófagos/virología , Proteínas de Unión al GTP Monoméricas/farmacología , Células Mieloides/virología , Retroviridae/efectos de los fármacos , Retroviridae/patogenicidad , Replicación Viral/efectos de los fármacos , Línea Celular , Células Dendríticas/metabolismo , Células Dendríticas/virología , VIH-1/efectos de los fármacos , VIH-1/fisiología , Humanos , Células Jurkat , Macrófagos/inmunología , Proteínas de Unión al GTP Monoméricas/metabolismo , Células Mieloides/metabolismo , Retroviridae/clasificación , Retroviridae/fisiología , Proteína 1 que Contiene Dominios SAM y HD
19.
J Virol ; 86(23): 12552-60, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22973040

RESUMEN

Sterile alpha motif domain- and HD domain-containing protein 1 (SAMHD1) is a deoxynucleoside triphosphohydrolase that restricts the replication of lentiviruses in myeloid cells by hydrolyzing the cellular deoxynucleotide triphosphates to a level below that which is required for reverse transcription. Human immunodeficiency virus type 2 (HIV-2) and some simian immunodeficiency viruses (SIVs) encode the accessory protein viral protein X (Vpx) that counteracts SAMHD1. Vpx recruits SAMHD1 to a cullin4A-RING E3 ubiquitin ligase (CRL4), which targets the enzyme for proteasomal degradation. Vpx and SAMHD1 both localize to the nucleus of the cell. We identified the nuclear localization sequence (NLS) of SAMHD1 as a conserved KRPR sequence at amino acid residues 11 to 14. SAMHD1 lacking a functional NLS localized to the cytoplasm but retained its triphosphohydrolase and antiviral activities. However, cytoplasmic SAMHD1 was resistant to Vpx-induced degradation, and its antiviral activity was not counteracted by Vpx. Cytoplasmic SAMHD1 interacted with Vpx and retained it in the cytoplasm. The inhibition of nuclear export with leptomycin B did not impair the ability of Vpx to degrade SAMHD1. These findings suggest that SAMHD1 is targeted by Vpx for ubiquitination and degradation in the nucleus.


Asunto(s)
Núcleo Celular/metabolismo , Proteínas de Unión al GTP Monoméricas/metabolismo , Proteolisis , Ubiquitina-Proteína Ligasas/metabolismo , Proteínas Reguladoras y Accesorias Virales/metabolismo , Secuencia de Aminoácidos/genética , Proteínas Cullin/metabolismo , Células HeLa , Humanos , Immunoblotting , Inmunoprecipitación , Microscopía Fluorescente , Señales de Localización Nuclear/genética , Plásmidos/genética , Proteína 1 que Contiene Dominios SAM y HD , Ubiquitinación
20.
bioRxiv ; 2023 Jan 12.
Artículo en Inglés | MEDLINE | ID: mdl-36711584

RESUMEN

Vectored immunoprophylaxis was first developed as a means to establish engineered immunity to HIV through the use of an adeno-associated viral vector expressing a broadly neutralizing antibody. We have applied this concept to establish long-term prophylaxis against SARS-CoV-2 by adeno-associated and lentiviral vectors expressing a high affinity ACE2 decoy receptor. Administration of decoy-expressing AAV vectors based on AAV2.retro and AAV6.2 by intranasal instillation or intramuscular injection protected mice against high-titered SARS-CoV-2 infection. AAV and lentiviral vectored immunoprophylaxis was durable and active against recent SARS-CoV-2 Omicron subvariants. The AAV vectors were also effective when administered up to 24 hours post-infection. Vectored immunoprophylaxis could be of value for immunocompromised individuals for whom vaccination is not practical and as a means to rapidly establish protection from infection. Unlike monoclonal antibody therapy, the approach is expected to remain active despite continued evolution viral variants.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA