Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Genome Res ; 33(3): 299-313, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36859333

RESUMEN

Insights into host-virus interactions during SARS-CoV-2 infection are needed to understand COVID-19 pathogenesis and may help to guide the design of novel antiviral therapeutics. N 6-Methyladenosine modification (m6A), one of the most abundant cellular RNA modifications, regulates key processes in RNA metabolism during stress response. Gene expression profiles observed postinfection with different SARS-CoV-2 variants show changes in the expression of genes related to RNA catabolism, including m6A readers and erasers. We found that infection with SARS-CoV-2 variants causes a loss of m6A in cellular RNAs, whereas m6A is detected abundantly in viral RNA. METTL3, the m6A methyltransferase, shows an unusual cytoplasmic localization postinfection. The B.1.351 variant has a less-pronounced effect on METTL3 localization and loss of m6A than did the B.1 and B.1.1.7 variants. We also observed a loss of m6A upon SARS-CoV-2 infection in air/liquid interface cultures of human airway epithelia, confirming that m6A loss is characteristic of SARS-CoV-2-infected cells. Further, transcripts with m6A modification are preferentially down-regulated postinfection. Inhibition of the export protein XPO1 results in the restoration of METTL3 localization, recovery of m6A on cellular RNA, and increased mRNA expression. Stress granule formation, which is compromised by SARS-CoV-2 infection, is restored by XPO1 inhibition and accompanied by a reduced viral infection in vitro. Together, our study elucidates how SARS-CoV-2 inhibits the stress response and perturbs cellular gene expression in an m6A-dependent manner.


Asunto(s)
COVID-19 , SARS-CoV-2 , Humanos , SARS-CoV-2/genética , COVID-19/genética , Metilación , ARN , ARN Viral/genética , Metiltransferasas/genética
2.
EMBO J ; 40(24): e108944, 2021 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-34601723

RESUMEN

Severe COVID-19 is characterized by lung abnormalities, including the presence of syncytial pneumocytes. Syncytia form when SARS-CoV-2 spike protein expressed on the surface of infected cells interacts with the ACE2 receptor on neighboring cells. The syncytia forming potential of spike variant proteins remain poorly characterized. Here, we first assessed Alpha (B.1.1.7) and Beta (B.1.351) spread and fusion in cell cultures, compared with the ancestral D614G strain. Alpha and Beta replicated similarly to D614G strain in Vero, Caco-2, Calu-3, and primary airway cells. However, Alpha and Beta formed larger and more numerous syncytia. Variant spike proteins displayed higher ACE2 affinity compared with D614G. Alpha, Beta, and D614G fusion was similarly inhibited by interferon-induced transmembrane proteins (IFITMs). Individual mutations present in Alpha and Beta spikes modified fusogenicity, binding to ACE2 or recognition by monoclonal antibodies. We further show that Delta spike also triggers faster fusion relative to D614G. Thus, SARS-CoV-2 emerging variants display enhanced syncytia formation.


Asunto(s)
Enzima Convertidora de Angiotensina 2/metabolismo , Anticuerpos Monoclonales/farmacología , Células Gigantes/virología , Mutación , SARS-CoV-2/fisiología , Glicoproteína de la Espiga del Coronavirus/genética , Animales , Células CACO-2 , Línea Celular , Chlorocebus aethiops , Células Gigantes/efectos de los fármacos , Células Gigantes/metabolismo , Células HEK293 , Humanos , SARS-CoV-2/efectos de los fármacos , SARS-CoV-2/genética , Células Vero , Replicación Viral/efectos de los fármacos
3.
J Gen Virol ; 104(11)2023 11.
Artículo en Inglés | MEDLINE | ID: mdl-37909282

RESUMEN

Enterovirus 71 (EV-A71) is a major public health problem, causing a range of illnesses from hand-foot-and-mouth disease to severe neurological manifestations. EV-A71 strains have been phylogenetically classified into eight genogroups (A to H), based on their capsid-coding genomic region. Genogroups B and C have caused large outbreaks worldwide and represent the two canonical circulating EV-A71 subtypes. Little is known about the antigenic diversity of new genogroups as compared to the canonical ones. Here, we compared the antigenic features of EV-A71 strains that belong to the canonical B and C genogroups and to genogroups E and F, which circulate in Africa. Analysis of the peptide sequences of EV-A71 strains belonging to different genogroups revealed a high level of conservation of the capsid residues involved in known linear and conformational neutralization antigenic sites. Using a published crystal structure of the EV-A71 capsid as a model, we found that most of the residues that are seemingly specific to some genogroups were mapped outside known antigenic sites or external loops. These observations suggest a cross-neutralization activity of anti-genogroup B or C antibodies against strains of genogroups E and F. Neutralization assays were performed with diverse rabbit and mouse anti-EV-A71 sera, anti-EV-A71 human standards and a monoclonal neutralizing antibody. All the batches of antibodies that were tested successfully neutralized all available isolates, indicating an overall broad cross-neutralization between the canonical genogroups B and C and genogroups E and F. A panel constituted of more than 80 individual human serum samples from Cambodia with neutralizing antibodies against EV-A71 subgenogroup C4 showed quite similar cross-neutralization activities between isolates of genogroups C4, E and F. Our results thus indicate that the genetic drift underlying the separation of EV-A71 strains into genogroups A, B, C, E and F does not correlate with the emergence of antigenically distinct variants.


Asunto(s)
Enterovirus Humano A , Infecciones por Enterovirus , Enterovirus , Enfermedad de Boca, Mano y Pie , Humanos , Ratones , Animales , Conejos , Enterovirus Humano A/genética , Antígenos Virales/genética , Proteínas de la Cápside/genética , Genotipo , Anticuerpos Monoclonales
4.
J Virol ; 96(19): e0130122, 2022 10 12.
Artículo en Inglés | MEDLINE | ID: mdl-36121299

RESUMEN

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) remained genetically stable during the first 3 months of the pandemic, before acquiring a D614G spike mutation that rapidly spread worldwide and then generating successive waves of viral variants with increasingly high transmissibility. We set out to evaluate possible epistatic interactions between the early-occurring D614G mutation and the more recently emerged cleavage site mutations present in spike of the Alpha, Delta, and Omicron variants of concern. The P681H/R mutations at the S1/S2 cleavage site increased spike processing and fusogenicity but limited its incorporation into pseudoviruses. In addition, the higher cleavage rate led to higher shedding of the spike S1 subunit, resulting in a lower infectivity of the P681H/R-carrying pseudoviruses compared to those expressing the Wuhan wild-type spike. The D614G mutation increased spike expression at the cell surface and limited S1 shedding from pseudovirions. As a consequence, the D614G mutation preferentially increased the infectivity of P681H/R-carrying pseudoviruses. This enhancement was more marked in cells where the endosomal route predominated, suggesting that more stable spikes could better withstand the endosomal environment. Taken together, these findings suggest that the D614G mutation stabilized S1/S2 association and enabled the selection of mutations that increased S1/S2 cleavage, leading to the emergence of SARS-CoV-2 variants expressing highly fusogenic spikes. IMPORTANCE The first SARS-CoV-2 variant that spread worldwide in early 2020 carried a D614G mutation in the viral spike, making this protein more stable in its cleaved form at the surface of virions. The Alpha and Delta variants, which spread in late 2020 and early 2021, respectively, proved increasingly transmissible and pathogenic compared to the original strain. Interestingly, Alpha and Delta both carried the mutations P681H/R in a cleavage site that made the spike more cleaved and more efficient at mediating viral fusion. We show here that variants with increased spike cleavage due to P681H/R were even more dependent on the stabilizing effect of the D614G mutation, which limited the shedding of cleaved S1 subunits from viral particles. These findings suggest that the worldwide spread of the D614G mutation was a prerequisite for the emergence of more pathogenic SARS-CoV-2 variants with highly fusogenic spikes.


Asunto(s)
COVID-19 , SARS-CoV-2 , Glicoproteína de la Espiga del Coronavirus , COVID-19/virología , Humanos , Mutación , SARS-CoV-2/genética , SARS-CoV-2/patogenicidad , Glicoproteína de la Espiga del Coronavirus/genética
5.
J Virol ; 96(7): e0216121, 2022 04 13.
Artículo en Inglés | MEDLINE | ID: mdl-35297660

RESUMEN

Vaccine-induced protective T cell immunity is necessary for HIV-1 functional cure. We previously reported that rhesus PD1-Gag-based DNA vaccination sustained simian-human immunodeficiency virus (SHIV) suppression by inducing effector-memory CD8+ T cells. Here, we investigated a human PD1-Gag-based DNA vaccine, namely, ICVAX, for clinical translation. PD1-based dendritic cell targeting and mosaic antigenic designs were combined to generate the ICVAX by fusing the human soluble PD1 domain with a bivalent HIV-1 Gag-p41 mosaic antigen. The mosaic antigen was cross-reactive with patients infected with B, CRF07/08_BC, and CRF01_AE variants. In mice, ICVAX elicited stronger, broader, and more polyfunctional T cell responses than mosaic Gag-p41 alone, and suppressed EcoHIV infection more efficiently. In macaques, ICVAX elicited polyfunctional effector-memory T cell responses that targeted multiple nonoverlapping epitopes of the Gag-p41 antigen. Furthermore, ICVAX manufactured following good manufacturing practices proved potent immunogenicity in macaques after biannual homologous vaccination, warranting clinical evaluation of ICVAX as an immunotherapy against HIV-1. IMPORTANCE This study presents that ICVAX, a PD1-based DNA vaccine against HIV-1, could induce broad and polyfunctional T cell responses against different HIV-1 subtypes. ICVAX encodes a recombinant antigen consisting of the human soluble PD1 domain fused with two mosaic Gag-p41 antigens. The mosaic antigens cover more than 500 HIV-1 strains circulating in China including the subtypes B/B', CRF01_AE, and CRF07/08_BC. In mice, ICVAX elicited stronger, broader, and more polyfunctional T cell responses, with better EcoHIV suppression than the nontargeting mosaic Gag-p41 DNA vaccine. Moreover, both lab-generated and GMP-grade ICVAX also elicited strong polyfunctional effector-memory T cell responses in rhesus macaques with good immunogenicity against multiple nonoverlapping epitopes of the Gag-p41 antigen. This study therefore highlights the great potential to translate the PD1-based DNA vaccine approach into clinical use, and opens up new avenues for alternative HIV-1 vaccine design for HIV-1 preventive and functional cure.


Asunto(s)
Infecciones por VIH , VIH-1 , Vacunas Combinadas , Vacunas de ADN , Vacunas Virales , Vacunas contra el SIDA/inmunología , Animales , Antígenos Virales , Antígeno CD48 , Linfocitos T CD8-positivos , Epítopos/inmunología , Productos del Gen gag/genética , Productos del Gen gag/inmunología , Infecciones por VIH/prevención & control , VIH-1/genética , Humanos , Macaca mulatta , Células T de Memoria , Ratones , Vacunas Combinadas/genética , Vacunas Combinadas/inmunología , Vacunas de ADN/genética , Vacunas de ADN/inmunología , Vacunas Virales/genética , Vacunas Virales/inmunología
6.
PLoS Pathog ; 17(6): e1009647, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-34125864

RESUMEN

HIV-1 functional cure requires sustained viral suppression without antiretroviral therapy. While effector-memory CD8+ T lymphocytes are essential for viremia control, few vaccines elicit such cellular immunity that could be potently recalled upon viral infection. Here, we investigated a program death-1 (PD1)-based vaccine by fusion of simian immunodeficiency virus capsid antigen to soluble PD1. Homologous vaccinations suppressed setpoint viremia to undetectable levels in vaccinated macaques following a high-dose intravenous challenge by the pathogenic SHIVSF162P3CN. Poly-functional effector-memory CD8+ T cells were not only induced after vaccination, but were also recalled upon viral challenge for viremia control as determined by CD8 depletion. Vaccine-induced effector memory CD8+ subsets displayed high cytotoxicity-related genes by single-cell analysis. Vaccinees with sustained viremia suppression for over two years responded to boost vaccination without viral rebound. These results demonstrated that PD1-based vaccine-induced effector-memory CD8+ T cells were recalled by AIDS virus infection, providing a potential immunotherapy for functional cure.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Memoria Inmunológica/inmunología , Receptor de Muerte Celular Programada 1/inmunología , Vacunas contra el SIDAS/inmunología , Viremia/prevención & control , Animales , Femenino , Macaca mulatta , Ratones , Ratones Endogámicos C57BL , Síndrome de Inmunodeficiencia Adquirida del Simio/inmunología , Síndrome de Inmunodeficiencia Adquirida del Simio/prevención & control , Virus de la Inmunodeficiencia de los Simios , Vacunas de ADN/inmunología , Productos del Gen gag del Virus de la Inmunodeficiencia Humana/inmunología
7.
Int J Mol Sci ; 22(2)2021 Jan 07.
Artículo en Inglés | MEDLINE | ID: mdl-33430234

RESUMEN

CD4+ T cells orchestrate adaptive immune responses through their capacity to recruit and provide help to multiple immune effectors, in addition to exerting direct effector functions. CD4+ T cells are increasingly recognized as playing an essential role in the control of chronic viral infections. In this review, we present recent advances in understanding the nature of CD4+ T cell help provided to antiviral effectors. Drawing from our studies of natural human immunodeficiency virus (HIV) control, we then focus on the role of high-affinity T cell receptor (TCR) clonotypes in mediating antiviral CD4+ T cell responses. Last, we discuss the role of TCR affinity in determining CD4+ T cell differentiation, reviewing the at times divergent studies associating TCR signal strength to the choice of a T helper 1 (Th1) or a T follicular helper (Tfh) cell fate.


Asunto(s)
Infecciones por VIH/inmunología , Receptores de Antígenos de Linfocitos T/inmunología , Células T Auxiliares Foliculares/citología , Células T Auxiliares Foliculares/inmunología , Inmunidad Adaptativa/inmunología , Linfocitos T CD8-positivos/inmunología , Diferenciación Celular/inmunología , Infecciones por VIH/virología , Humanos , Inmunidad Humoral/inmunología , Linfocitos T Reguladores/inmunología
8.
J Immunol ; 199(10): 3437-3452, 2017 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-28993513

RESUMEN

Rare patients who spontaneously control HIV replication provide a useful model to inform HIV vaccine development. HIV controllers develop particularly efficient antiviral CD4+ T cell responses mediated by shared high-affinity TCRs. To determine whether the candidate DNA vaccine ADVAX could induce similar responses, we analyzed Gag-specific primary CD4+ T cells from healthy volunteers who received ADVAX DNA by electroporation. Vaccinated volunteers had an immunodominant response to the Gag293 epitope with a functional avidity intermediate between that of controllers and treated patients. The TCR repertoire of Gag293-specific CD4+ T cells proved highly biased, with a predominant usage of the TCRß variable gene 2 (TRBV2) in vaccinees as well as controllers. TCRα variable gene (TRAV) gene usage was more diverse, with the dominance of TRAV29 over TRAV24 genes in vaccinees, whereas TRAV24 predominated in controllers. Sequence analysis revealed an unexpected degree of overlap between the specific repertoires of vaccinees and controllers, with the sharing of TRAV24 and TRBV2 public motifs (>30%) and of public clonotypes characteristic of high-affinity TCRs. MHC class II tetramer binding revealed a broad HLA-DR cross-restriction, explaining how Gag293-specific public clonotypes could be selected in individuals with diverse genetic backgrounds. TRAV29 clonotypes also proved cross-restricted, but conferred responses of lower functional avidity upon TCR transfer. In conclusion, DNA vaccination by electroporation primed for TCR clonotypes that were associated with HIV control, highlighting the potential of this vaccine delivery method. To our knowledge, this study provides the first proof-of-concept that clonotypic analysis may be used as a tool to monitor the quality of vaccine-induced responses and modulate these toward "controller-like" responses.


Asunto(s)
Vacunas contra el SIDA/inmunología , Linfocitos T CD4-Positivos/inmunología , Infecciones por VIH/inmunología , VIH/fisiología , Receptores de Antígenos de Linfocitos T alfa-beta/genética , Productos del Gen gag del Virus de la Inmunodeficiencia Humana/inmunología , Células Clonales , Reacciones Cruzadas , Electroporación , Ensayo de Immunospot Ligado a Enzimas , Antígenos HLA-DR/metabolismo , Humanos , Activación de Linfocitos , Unión Proteica , Vacunas de ADN , Replicación Viral
9.
Med Sci (Paris) ; 31(1): 53-9, 2015 Jan.
Artículo en Francés | MEDLINE | ID: mdl-25658731

RESUMEN

Co-infection with Mycobacterium tuberculosis (Mtb) and human immunodeficiency virus (HIV) represents a major threat to public health worldwide. The treatment of patients coinfected by Mtb and HIV is often complicated by the occurrence of an immune reconstitution inflammatory syndrome (IRIS), resulting in the unexpected resumption of tuberculosis symptoms after the initiation of antiretroviral therapy. IRIS is associated with a rapid reconstitution of CD4(+) T cell responses specific for Mtb, which is promoted by the control of HIV replication and a high concentration of available interleukin-7. Macrophages, whose activity is suddenly stimulated by CD4(+) T cell help, respond by an exacerbated inflammatory response in Mtb-rich tissues. A major research objective remains to identify biomarkers which could allow a reliable prediction of IRIS occurrence, in order to optimize medical care for the many patients affected by both HIV and tuberculosis in resource-limited settings.


Asunto(s)
Infecciones por VIH/terapia , Síndrome Inflamatorio de Reconstitución Inmune/etiología , Tuberculosis/terapia , Coinfección , Infecciones por VIH/complicaciones , Infecciones por VIH/inmunología , Humanos , Síndrome Inflamatorio de Reconstitución Inmune/diagnóstico , Síndrome Inflamatorio de Reconstitución Inmune/inmunología , Inflamación/etiología , Inflamación/inmunología , Factores de Riesgo , Tuberculosis/complicaciones , Tuberculosis/inmunología
10.
J Virol ; 87(18): 10368-80, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23864638

RESUMEN

The expression of certain HLA class I alleles, including HLA-B*27 and HLA-B*57, is associated with better control of human immunodeficiency virus type 1 (HIV-1) infection, but the mechanisms responsible are not fully understood. We sought evidence that pressure from the human restriction factor TRIM5α (hTRIM5α) could contribute to viral control. The hTRIM5α sensitivity of viruses from both HLA-B*57-positive (HLA-B*57(+)) and HLA-B*27(+) patients who spontaneously controlled viral replication, but not viruses from viremic patients expressing these alleles, was significantly greater than that of viruses from patients not expressing these protective HLA-B alleles. Overall, a significant negative correlation between hTRIM5α sensitivity and viral load was observed. In HLA-B*57(+) patients, the T242N mutation in the HLA-B*57-restricted TW10 CD8(+) T lymphocyte (CTL) epitope was strongly associated with hTRIM5α sensitivity. In HLA-B*27(+) controllers, hTRIM5α sensitivity was associated with a significant reduction in emergence of key CTL mutations. In several patients, viral evolution to avoid hTRIM5α sensitivity was observed but could be associated with reduced viral replicative capacity. Thus, in individuals expressing protective HLA-B alleles, the combined pressures exerted by CTL, hTRIM5α, and capsid structural constraints can prevent viral escape both by impeding the selection of necessary resistance/compensatory mutations and forcing the selection of escape mutations that increase hTRIM5α sensitivity or impair viral replicative capacity.


Asunto(s)
Proteínas Portadoras/inmunología , Infecciones por VIH/inmunología , VIH-1/inmunología , Antígeno HLA-B27/inmunología , Adulto , Factores de Restricción Antivirales , Femenino , Antígeno HLA-B27/genética , Humanos , Masculino , Persona de Mediana Edad , Proteínas de Motivos Tripartitos , Ubiquitina-Proteína Ligasas , Carga Viral
11.
J Virol ; 86(19): 10661-74, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22837194

RESUMEN

HIV controllers are rare individuals who spontaneously control HIV replication in the absence of antiretroviral therapy. To identify parameters of the CD4 response that may contribute to viral control rather than merely reflect a persistently low viremia, we compared the T helper profiles in two groups of patients with more than 10 years of viral suppression: HIV controllers from the Agence Nationale de Recherche sur le SIDA et les Hépatites Virales (ANRS) CO18 cohort (n = 26) and efficiently treated patients (n = 16). Cells specific for immunodominant Gag and cytomegalovirus (CMV) peptides were evaluated for the production of 10 cytokines and cytotoxicity markers and were also directly quantified ex vivo by major histocompatibility complex (MHC) class II tetramer staining. HIV controller CD4(+) T cells were characterized by a higher frequency of gamma interferon (IFN-γ) production, perforin(+)/CD107a(+) expression, and polyfunctionality in response to Gag peptides. While interleukin 4 (IL-4), IL-17, and IL-21 production did not differ between groups, the cells of treated patients produced more IL-10 in response to Gag and CMV peptides, pointing to persistent negative immunoregulation after long-term antiretroviral therapy. Gag293 tetramer-positive cells were detected at a high frequency (0.12%) and correlated positively with IFN-γ-producing CD4(+) T cells in the controller group (R = 0.73; P = 0.003). Tetramer-positive cells were fewer in the highly active antiretroviral therapy (HAART) group (0.04%) and did not correlate with IFN-γ production, supporting the notion of a persistent immune dysfunction in HIV-specific CD4(+) T cells of treated patients. In conclusion, HIV controllers maintained a population of highly efficient Th1 effectors directed against Gag in spite of a persistently low antigenemia, while patients treated in the long term showed a loss of CD4 effector functions.


Asunto(s)
Infecciones por VIH/sangre , Infecciones por VIH/virología , Células TH1/virología , ADP-Ribosil Ciclasa 1/biosíntesis , Adulto , Anciano , Antirretrovirales/farmacología , Linfocitos T CD4-Positivos/citología , Estudios de Cohortes , Citocinas/metabolismo , Citometría de Flujo/métodos , Productos del Gen gag/metabolismo , Humanos , Interferón gamma/metabolismo , Interleucina-10/metabolismo , Interleucina-17/metabolismo , Interleucina-4/metabolismo , Interleucinas/metabolismo , Leucocitos Mononucleares/citología , Proteína 1 de la Membrana Asociada a los Lisosomas/biosíntesis , Persona de Mediana Edad , Fenotipo
12.
Front Immunol ; 14: 1221961, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37559726

RESUMEN

Background: The role of adaptive immune responses in long COVID remains poorly understood, with contrasting hypotheses suggesting either an insufficient antiviral response or an excessive immune response associated with inflammatory damage. To address this issue, we set to characterize humoral and CD4+ T cell responses in long COVID patients prior to SARS-CoV-2 vaccination. Methods: Long COVID patients who were seropositive (LC+, n=28) or seronegative (LC-, n=23) by spike ELISA assay were recruited based on (i) an initial SARS-CoV-2 infection documented by PCR or the conjunction of three major signs of COVID-19 and (ii) the persistence or resurgence of at least 3 symptoms for over 3 months. They were compared to COVID patients with resolved symptoms (RE, n=29) and uninfected control individuals (HD, n=29). Results: The spectrum of persistent symptoms proved similar in both long COVID groups, with a trend for a higher number of symptoms in the seronegative group (median=6 vs 4.5; P=0.01). The use a highly sensitive S-flow assay enabled the detection of low levels of SARS-CoV-2 spike-specific IgG in 22.7% of ELISA-seronegative long COVID (LC-) patients. In contrast, spike-specific IgG levels were uniformly high in the LC+ and RE groups. Multiplexed antibody analyses to 30 different viral antigens showed that LC- patients had defective antibody responses to all SARS-CoV-2 proteins tested but had in most cases preserved responses to other viruses. A sensitive primary T cell line assay revealed low but detectable SARS-CoV-2-specific CD4 responses in 39.1% of LC- patients, while response frequencies were high in the LC+ and RE groups. Correlation analyses showed overall strong associations between humoral and cellular responses, with exceptions in the LC- group. Conclusions: These findings provide evidence for two major types of antiviral immune responses in long COVID. Seropositive patients showed coordinated cellular and humoral responses at least as high as those of recovered patients. In contrast, ELISA-seronegative long COVID patients showed overall low antiviral responses, with detectable specific CD4+ T cells and/or antibodies in close to half of patients (52.2%). These divergent findings in patients sharing a comparable spectrum of persistent symptoms raise the possibility of multiple etiologies in long COVID.


Asunto(s)
COVID-19 , Síndrome Post Agudo de COVID-19 , Humanos , Vacunas contra la COVID-19 , SARS-CoV-2 , Anticuerpos Antivirales , Antivirales , Inmunoglobulina G
13.
Emerg Microbes Infect ; 12(2): 2245921, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37542391

RESUMEN

Prevention of robust severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) infection in nasal turbinate (NT) requires in vivo evaluation of IgA neutralizing antibodies. Here, we report the efficacy of receptor binding domain (RBD)-specific monomeric B8-mIgA1 and B8-mIgA2, and dimeric B8-dIgA1, B8-dIgA2 and TH335-dIgA1 against intranasal SARS-CoV-2 challenge in Syrian hamsters. These antibodies exhibited comparable neutralization potency against authentic virus by competing with human angiotensin converting enzyme-2 (ACE2) receptor for RBD binding. While reducing viral loads in lungs significantly, prophylactic intranasal B8-dIgA unexpectedly led to high amount of infectious viruses and extended damage in NT compared to controls. Mechanistically, B8-dIgA failed to inhibit SARS-CoV-2 cell-to-cell transmission, but was hijacked by the virus through dendritic cell-mediated trans-infection of NT epithelia leading to robust nasal infection. Cryo-EM further revealed B8 as a class II antibody binding trimeric RBDs in 3-up or 2-up/1-down conformation. Neutralizing dIgA, therefore, may engage an unexpected mode of SARS-CoV-2 nasal infection and injury.


Asunto(s)
COVID-19 , Resfriado Común , Cricetinae , Animales , Humanos , SARS-CoV-2 , Mesocricetus , Anticuerpos Antivirales , Anticuerpos Neutralizantes , Inmunoglobulina A , Glicoproteína de la Espiga del Coronavirus
14.
PLoS Pathog ; 6(2): e1000780, 2010 Feb 26.
Artículo en Inglés | MEDLINE | ID: mdl-20195518

RESUMEN

HIV controllers are rare individuals who spontaneously control HIV replication in the absence of antiretroviral treatment. Emerging evidence indicates that HIV control is mediated through very active cellular immune responses, though how such responses can persist over time without immune exhaustion is not yet understood. To investigate the nature of memory CD4+ T cells responsible for long-term anti-HIV responses, we characterized the growth kinetics, Vbeta repertoire, and avidity for antigen of patient-derived primary CD4+ T cell lines. Specific cell lines were obtained at a high rate for both HIV controllers (16/17) and efficiently treated patients (19/20) in response to the immunodominant Gag293 peptide. However, lines from controllers showed faster growth kinetics than those of treated patients. After normalizing for growth rates, IFN-gamma responses directed against the immunodominant Gag293 peptide showed higher functional avidity in HIV controllers, indicating differentiation into highly efficient effector cells. In contrast, responses to Gag161, Gag263, or CMV peptides did not differ between groups. Gag293-specific CD4+ T cells were characterized by a diverse Vbeta repertoire, suggesting that multiple clones contributed to the high avidity CD4+ T cell population in controllers. The high functional avidity of the Gag293-specific response could be explained by a high avidity interaction between the TCR and the peptide-MHC complex, as demonstrated by MHC class II tetramer binding. Thus, HIV controllers harbor a pool of memory CD4+ T cells with the intrinsic ability to recognize minimal amounts of Gag antigen, which may explain how they maintain an active antiviral response in the face of very low viremia.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/virología , Productos del Gen gag/inmunología , Infecciones por VIH/inmunología , Receptores de Antígenos de Linfocitos T/inmunología , VIH/inmunología , Humanos , Activación de Linfocitos/inmunología , Carga Viral
15.
Blood ; 115(18): 3708-17, 2010 May 06.
Artículo en Inglés | MEDLINE | ID: mdl-20038787

RESUMEN

Idiopathic CD4(+) T-cell lymphocytopenia (ICL) is a rare acquired T-cell immunodeficiency of unknown pathogenic basis. Six adults with ICL who developed opportunistic infections were investigated using extensive immunophenotyping analysis and functional evaluation of the chemokine receptor CXCR4. For all 6 patients studied, a profound defect in CXCR4 expression was detected at the surface of CD4(+) T lymphocytes, in association with an abnormal intracellular accumulation of CXCR4 and of its natural ligand, the chemokine CXCL12. For all patients studied, CD4(+) T-cell chemotactic response toward CXCL12 was decreased, whereas sensitivity to CXCL8 was preserved. CXCR4 recovery after ligand-induced endocytosis was impaired in ICL CD4(+) T cells. Upon in vitro addition of interleukin-2 (IL-2), membrane expression of CXCR4 returned to normal levels in 5 of 6 patients, whereas intracellular accumulation of CXCR4 and CXCL12 disappeared. Upon therapeutic administration of IL-2, CD4(+) T-cell count and membrane CXCR4 expression and function improved over time in 3 of 4 patients treated. Therefore, our data indicate that ICL is associated with defective surface expression of CXCR4, which may be reversed by IL-2.


Asunto(s)
Linfocitos T CD4-Positivos/metabolismo , Membrana Celular/metabolismo , Infecciones Oportunistas/metabolismo , Receptores CXCR4/metabolismo , Linfocitopenia-T Idiopática CD4-Positiva/metabolismo , Adolescente , Adulto , Células Cultivadas , Quimiocinas , Quimiotaxis , Citometría de Flujo , Humanos , Inmunofenotipificación , Interleucina-2/uso terapéutico , Persona de Mediana Edad , Linfocitopenia-T Idiopática CD4-Positiva/terapia , Adulto Joven
16.
J Cell Biol ; 221(7)2022 07 04.
Artículo en Inglés | MEDLINE | ID: mdl-35695891

RESUMEN

Wang et al. report in this issue (2022. J. Cell Biol.https://doi.org/10.1083/jcb.202108015) that the SARS-CoV-2 protein ORF10 increases the activity of the E3 ligase CUL2ZYG11B, leading to the degradation of multiple ciliary proteins. The resulting loss of cilia may facilitate the spread of SARS-CoV-2 in the respiratory tree.


Asunto(s)
COVID-19 , Cilios , Ubiquitina-Proteína Ligasas , COVID-19/patología , Proteínas de Ciclo Celular , Cilios/patología , Proteínas Cullin , Genes Virales , Humanos , Proteínas/metabolismo , SARS-CoV-2 , Ubiquitina-Proteína Ligasas/metabolismo
17.
Nat Commun ; 13(1): 521, 2022 01 26.
Artículo en Inglés | MEDLINE | ID: mdl-35082297

RESUMEN

HIV elite controllers maintain a population of CD4 + T cells endowed with high avidity for Gag antigens and potent effector functions. How these HIV-specific cells avoid infection and depletion upon encounter with the virus remains incompletely understood. Ex vivo characterization of single Gag-specific CD4 + T cells reveals an advanced Th1 differentiation pattern in controllers, except for the CCR5 marker, which is downregulated compared to specific cells of treated patients. Accordingly, controller specific CD4 + T cells show decreased susceptibility to CCR5-dependent HIV entry. Two controllers carried biallelic mutations impairing CCR5 surface expression, indicating that in rare cases CCR5 downregulation can have a direct genetic cause. Increased expression of ß-chemokine ligands upon high-avidity antigen/TCR interactions contributes to autocrine CCR5 downregulation in controllers without CCR5 mutations. These findings suggest that genetic and functional regulation of the primary HIV coreceptor CCR5 play a key role in promoting natural HIV control.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Controladores de Élite , Infecciones por VIH/inmunología , VIH-1/inmunología , Receptores CCR5/metabolismo , Internalización del Virus , Quimiocinas , Regulación hacia Abajo , Regulación de la Expresión Génica , Productos del Gen gag/metabolismo , Infecciones por VIH/virología , Antígenos de Histocompatibilidad Clase II , Humanos , Mutación , Receptores CCR5/genética , Receptores CXCR3
18.
Nat Cell Biol ; 24(1): 24-34, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-35027731

RESUMEN

SARS-CoV-2 infection of human cells is initiated by the binding of the viral Spike protein to its cell-surface receptor ACE2. We conducted a targeted CRISPRi screen to uncover druggable pathways controlling Spike protein binding to human cells. Here we show that the protein BRD2 is required for ACE2 transcription in human lung epithelial cells and cardiomyocytes, and BRD2 inhibitors currently evaluated in clinical trials potently block endogenous ACE2 expression and SARS-CoV-2 infection of human cells, including those of human nasal epithelia. Moreover, pharmacological BRD2 inhibition with the drug ABBV-744 inhibited SARS-CoV-2 replication in Syrian hamsters. We also found that BRD2 controls transcription of several other genes induced upon SARS-CoV-2 infection, including the interferon response, which in turn regulates the antiviral response. Together, our results pinpoint BRD2 as a potent and essential regulator of the host response to SARS-CoV-2 infection and highlight the potential of BRD2 as a therapeutic target for COVID-19.


Asunto(s)
Enzima Convertidora de Angiotensina 2/metabolismo , Antivirales/farmacología , Células Epiteliales/virología , SARS-CoV-2/metabolismo , Factores de Transcripción/efectos de los fármacos , Enzima Convertidora de Angiotensina 2/efectos de los fármacos , COVID-19/metabolismo , COVID-19/virología , Línea Celular , Células Epiteliales/metabolismo , Humanos , Glicoproteínas de Membrana/metabolismo , SARS-CoV-2/efectos de los fármacos , SARS-CoV-2/patogenicidad , Factores de Transcripción/metabolismo , Tratamiento Farmacológico de COVID-19
19.
Clin Immunol ; 141(1): 15-30, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21865089

RESUMEN

A small minority of HIV-infected individuals, known as HIV controllers, is able to exert long-term control over HIV replication in the absence of treatment. Increasing evidence suggests that the adaptive immune system plays a critical role in this control but also that a combination of several host and/or viral factors, rather than a single cause, leads to this rare phenotype. Here, we review recent advances in the study of these remarkable individuals. We summarize the epidemiology and clinical characteristics of HIV controllers, and subsequently describe contributing roles of host genetic factors, innate and adaptive immune responses, and viral factors to this phenotype. We emphasize distinctive characteristics of HIV-specific CD4 T cell responses and of CD4 T cell subpopulations that are frequently found in HIV controllers. We discuss major controversies in the field and the relevance of the study of HIV controllers for the development of novel therapeutic strategies and vaccines.


Asunto(s)
Infecciones por VIH/inmunología , Infecciones por VIH/virología , Sobrevivientes de VIH a Largo Plazo , Inmunidad Adaptativa , Animales , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/virología , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/virología , Modelos Animales de Enfermedad , Estudio de Asociación del Genoma Completo , VIH/inmunología , VIH/patogenicidad , VIH/fisiología , Infecciones por VIH/genética , Interacciones Huésped-Patógeno/inmunología , Humanos , Inmunidad Humoral , Inmunidad Innata , Inmunidad Mucosa , Fenotipo , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/virología , Células Th17/inmunología , Células Th17/virología , Replicación Viral
20.
J Virol ; 84(1): 96-108, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19864382

RESUMEN

Interleukin-7 (IL-7) plays a central role in controlling the homeostasis of both naive and long-term-memory CD4(+) T cells. To better understand how human immunodeficiency virus (HIV) perturbs CD4(+) T-cell homeostasis, we performed a detailed analysis of IL-7R expression, IL-7 binding, and IL-7-dependent early and late signaling events in CD4(+) T-cell subsets from viremic and efficiently treated patients. HIV infection differentially affected the expression of IL-7 receptor (IL-7R) chains, with decreases in IL-7Ralpha/CD127 expression in the memory subset and increases in gammac/CD132 expression in all CD4(+) T cells. This resulted in preserved IL-7 binding in the naive compartment and decreased IL-7 binding in the memory compartment of viremic patients. Accordingly, the percentages of cells signaling in response to IL-7, as measured by pSTAT5 induction, were decreased in memory subsets, including conventional CD4(+) T cells and regulatory T cells. However, the levels of pSTAT5 induction per responding cell, as measured by pSTAT5 fluorescence intensity, were increased within all naive and memory CD4(+) T-cell subsets of viremic patients. The basal level of pSTAT5 was also increased, indicating a constitutive activation of the JAK/STAT5 pathway. IL-7 functional responses, as measured by Bcl-2, CD25, and Foxp3 induction, were impaired in viremic patient CD4(+) T cells, suggesting that chronic activation led to downstream defects in the STAT5 signaling pathway. Thus, HIV infection perturbs IL-7 responses at both receptor binding and signaling steps, which likely compromises the regenerative capacity of the CD4(+) T-cell pool and may contribute to CD4(+) T-cell depletion.


Asunto(s)
Linfocitos T CD4-Positivos/virología , Infecciones por VIH/inmunología , Interleucina-7/metabolismo , Quinasas Janus/metabolismo , Factor de Transcripción STAT5/metabolismo , Adulto , Recuento de Linfocito CD4 , Linfocitos T CD4-Positivos/patología , Estudios de Casos y Controles , Homeostasis , Humanos , Persona de Mediana Edad , Unión Proteica , Transducción de Señal , Carga Viral
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA