Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
J Immunol ; 208(2): 429-443, 2022 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-34903642

RESUMEN

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) induces T cell, B cell, and Ab responses that are detected for several months in recovered individuals. Whether this response resembles a typical respiratory viral infection is a matter of debate. In this study, we followed T cell and Ab responses in 24 mainly nonhospitalized human subjects who had recovered from PCR-confirmed SARS-CoV-2 infection at two time points (median of 45 and 145 d after symptom onset). Ab responses were detected in 95% of subjects, with a strong correlation between plasma and salivary anti-spike (anti-S) and anti-receptor binding domain IgG, as well as a correlation between circulating T follicular helper cells and the SARS-CoV-2-specific IgG response. T cell responses to SARS-CoV-2 peptides were determined using intracellular cytokine staining, activation markers, proliferation, and cytokine secretion. All study subjects had a T cell response to at least one SARS-CoV-2 Ag based on at least one T cell assay. CD4+ responses were largely of the Th1 phenotype, but with a lower ratio of IFN-γ- to IL-2-producing cells and a lower frequency of CD8+:CD4+ T cells than in influenza A virus (IAV)-specific memory responses within the same subjects. Analysis of secreted molecules also revealed a lower ratio of IFN-γ to IL-2 and an altered cytotoxic profile for SARS-CoV-2 S- and nucleocapsid-specific responses compared with IAV-specific responses. These data suggest that the memory T cell phenotype after a single infection with SARS-CoV-2 persists over time, with an altered cytokine and cytotoxicity profile compared with long-term memory to whole IAV within the same subjects.


Asunto(s)
Formación de Anticuerpos , COVID-19/inmunología , Inmunidad Celular , Inmunoglobulina G/inmunología , SARS-CoV-2/inmunología , Glicoproteína de la Espiga del Coronavirus/inmunología , Células TH1/inmunología , Adulto , Anciano , Femenino , Humanos , Masculino , Persona de Mediana Edad , Factores de Tiempo
2.
Clin Immunol ; 237: 108963, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35259543

RESUMEN

Convalescent coronavirus disease 2019 (COVID-19) subjects who receive BNT162b2 develop robust antibody responses against SARS-CoV-2. However, our understanding of the clonal B cell response pre- and post-vaccination in such individuals is limited. Here we characterized B cell phenotypes and the BCR repertoire after BNT162b2 immunization in two convalescent COVID-19 subjects. BNT162b2 stimulated many B cell clones that were under-represented during SARS-CoV-2 infection. In addition, the vaccine generated B cell clusters with >65% similarity in CDR3 VH and VL region consensus sequences both within and between subjects. This result suggests that the CDR3 region plays a dominant role adjacent to heavy and light chain V/J pairing in the recognition of the SARS-CoV-2 spike protein. Antigen-specific B cell populations with homology to published SARS-CoV-2 antibody sequences from the CoV-AbDab database were observed in both subjects. These results point towards the development of convergent antibody responses against the virus in different individuals.


Asunto(s)
Anticuerpos Antivirales , Vacuna BNT162 , COVID-19 , Regiones Determinantes de Complementariedad , Anticuerpos Antivirales/inmunología , Formación de Anticuerpos , Vacuna BNT162/inmunología , COVID-19/inmunología , COVID-19/prevención & control , Regiones Determinantes de Complementariedad/genética , Humanos , SARS-CoV-2 , Glicoproteína de la Espiga del Coronavirus/inmunología
3.
J Virol ; 93(6)2019 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-30602611

RESUMEN

The HIV accessory protein Nef modulates key immune evasion and pathogenic functions, and its encoding gene region exhibits high sequence diversity. Given the recent identification of early HIV-specific adaptive immune responses as novel correlates of HIV reservoir size, we hypothesized that viral factors that facilitate the evasion of such responses-namely, Nef genetic and functional diversity-might also influence reservoir establishment and/or persistence. We isolated baseline plasma HIV RNA-derived nef clones from 30 acute/early-infected individuals who participated in a clinical trial of early combination antiretroviral therapy (cART) (<6 months following infection) and assessed each Nef clone's ability to downregulate CD4 and human leukocyte antigen (HLA) class I in vitro We then explored the relationships between baseline clinical, immunological, and virological characteristics and the HIV reservoir size measured 48 weeks following initiation of suppressive cART (where the reservoir size was quantified in terms of the proviral DNA loads as well as the levels of replication-competent HIV in CD4+ T cells). Maximal within-host Nef-mediated downregulation of HLA, but not CD4, correlated positively with post-cART proviral DNA levels (Spearman's R = 0.61, P = 0.0004) and replication-competent reservoir sizes (Spearman's R = 0.36, P = 0.056) in univariable analyses. Furthermore, the Nef-mediated HLA downregulation function was retained in final multivariable models adjusting for established clinical and immunological correlates of reservoir size. Finally, HIV subtype B-infected persons (n = 25) harbored significantly larger viral reservoirs than non-subtype B-infected persons (2 infected with subtype CRF01_AE and 3 infected with subtype G). Our results highlight a potentially important role of viral factors-in particular, HIV subtype and accessory protein function-in modulating viral reservoir establishment and persistence.IMPORTANCE While combination antiretroviral therapies (cART) have transformed HIV infection into a chronic manageable condition, they do not act upon the latent HIV reservoir and are therefore not curative. As HIV cure or remission should be more readily achievable in individuals with smaller HIV reservoirs, achieving a deeper understanding of the clinical, immunological, and virological determinants of reservoir size is critical to eradication efforts. We performed a post hoc analysis of 30 participants of a clinical trial of early cART who had previously been assessed in detail for their clinical, immunological, and reservoir size characteristics. We observed that the HIV subtype and autologous Nef-mediated HLA downregulation function correlated with the viral reservoir size measured approximately 1 year post-cART initiation. Our findings highlight virological characteristics-both genetic and functional-as possible novel determinants of HIV reservoir establishment and persistence.


Asunto(s)
Infecciones por VIH/inmunología , VIH/inmunología , Evasión Inmune/inmunología , Productos del Gen nef del Virus de la Inmunodeficiencia Humana/inmunología , Adulto , Antirretrovirales/farmacología , Antígenos CD4/inmunología , Linfocitos T CD4-Positivos/inmunología , Regulación hacia Abajo/efectos de los fármacos , Regulación hacia Abajo/inmunología , VIH/efectos de los fármacos , Infecciones por VIH/tratamiento farmacológico , Antígenos HLA/inmunología , Humanos , Evasión Inmune/efectos de los fármacos , Masculino , Persona de Mediana Edad , Latencia del Virus/efectos de los fármacos , Latencia del Virus/inmunología , Adulto Joven
4.
J Immunol ; 198(8): 3181-3194, 2017 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-28264968

RESUMEN

In chronic diseases, such as HIV infection, plasmacytoid dendritic cells (pDCs) are rendered dysfunctional, as measured by their decreased capacity to produce IFN-α. In this study, we identified elevated levels of T cell Ig and mucin-domain containing molecule-3 (Tim-3)-expressing pDCs in the blood of HIV-infected donors. The frequency of Tim-3-expressing pDCs correlated inversely with CD4 T cell counts and positively with HIV viral loads. A lower frequency of pDCs expressing Tim-3 produced IFN-α or TNF-α in response to the TLR7 agonists imiquimod and Sendai virus and to the TLR9 agonist CpG. Thus, Tim-3 may serve as a biomarker of pDC dysfunction in HIV infection. The source and function of Tim-3 was investigated on enriched pDC populations from donors not infected with HIV. Tim-3 induction was achieved in response to viral and artificial stimuli, as well as exogenous IFN-α, and was PI3K dependent. Potent pDC-activating stimuli, such as CpG, imiquimod, and Sendai virus, induced the most Tim-3 expression and subsequent dysfunction. Small interfering RNA knockdown of Tim-3 increased IFN-α secretion in response to activation. Intracellular Tim-3, as measured by confocal microscopy, was dispersed throughout the cytoplasm prior to activation. Postactivation, Tim-3 accumulated at the plasma membrane and associated with disrupted TLR9 at the submembrane. Tim-3-expressing pDCs had reduced IRF7 levels. Furthermore, intracellular Tim-3 colocalized with p85 and IRF7 within LAMP1+ lysosomes, suggestive of a role in degradation. We conclude that Tim-3 is a biomarker of dysfunctional pDCs and may negatively regulate IFN-α, possibly through interference with TLR signaling and recruitment of IRF7 and p85 into lysosomes, enhancing their degradation.


Asunto(s)
Biomarcadores/análisis , Células Dendríticas/inmunología , Infecciones por VIH/inmunología , Receptor 2 Celular del Virus de la Hepatitis A/inmunología , Transducción de Señal/inmunología , Adulto , Separación Celular , Células Dendríticas/metabolismo , Femenino , Infecciones por VIH/metabolismo , Receptor 2 Celular del Virus de la Hepatitis A/metabolismo , Humanos , Factor 7 Regulador del Interferón/inmunología , Factor 7 Regulador del Interferón/metabolismo , Lisosomas/inmunología , Lisosomas/metabolismo , Masculino , Microscopía Confocal , Persona de Mediana Edad , Receptor Toll-Like 9/inmunología , Receptor Toll-Like 9/metabolismo , Adulto Joven
6.
J Virol ; 91(16)2017 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-28592534

RESUMEN

Immunotherapy with passive administration of broadly neutralizing HIV-1 envelope-specific antibodies (bnAbs) in the setting of established infection in vivo has yielded mixed results. The contribution of different antibodies toward the direct elimination of infected cells is poorly understood. In this study, we determined the ability of 12 well-characterized anti-HIV-1 neutralizing antibodies to recognize and eliminate primary CD4 T cells infected with HIV-1 belonging to clades A, B, C, and D, via antibody-dependent complement-mediated lysis (ADCML) and antibody-dependent cell-mediated cytotoxicity (ADCC), in vitro We further tested unique combinations of these antibodies to determine the optimal antibody cocktails to be tested in future clinical trials. We report that antibody binding to infected CD4 T cells is highly variable and correlates with ADCML and ADCC processes. Particularly, antibodies targeting the envelope glycan shield (2G12) and V1/V2 site (PG9, PG16, and PGT145) are best at recognizing HIV-1-infected CD4 T cells. However, only PG9 and PG16 and their combinations with other bnAbs sufficiently induced the elimination of HIV-1-infected CD4 T cells by ADCML, ADCC, or both. Notably, CD4 binding site antibodies VRC01, 3BNC117, and NIH45-46 G54W did not exhibit recognition of infected cells and were unable to induce their killing. Future trials geared toward the development of a cure for HIV/AIDS should incorporate V1/V2 antibodies for maximal clearance of infected cells. With the use of only primary immune cells, we conducted a comprehensive cross-clade physiological analysis to aid the direction of antibodies as therapeutics toward the development of a cure for HIV/AIDS.IMPORTANCE Several antibodies capable of neutralizing the majority of circulating HIV-1 strains have been identified to date and have been shown to prevent infection in animal models. However, the use of combinations of such broadly neutralizing antibodies (bnAbs) for the treatment and eradication of HIV-1 in infected humans remains uncertain. In this study, we tested the ability of bnAbs to directly recognize and eliminate primary human CD4 T cells infected with diverse HIV-1 strains representative of the global epidemic by antibody-dependent pathways. We also tested several combinations of bnAbs in our assays in order to maximize the clearance of infected cells. We show that the ability of bnAbs to identify and kill infected cells is highly variable and that only a few of them are able to exert this function. Our data will help guide the formulation of bnAbs to test in future human trials aimed at the development of a cure.


Asunto(s)
Anticuerpos Neutralizantes/inmunología , Citotoxicidad Celular Dependiente de Anticuerpos , Proteínas del Sistema Complemento/inmunología , Reacciones Cruzadas , Anticuerpos Anti-VIH/inmunología , VIH-1/inmunología , Humanos
7.
J Virol ; 91(8)2017 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-28179527

RESUMEN

A major barrier to a human immunodeficiency virus type 1 (HIV-1) infection cure is the establishment of a viral reservoir in spite of combined antiretroviral therapy (cART). It is unclear how HIV-specific cytotoxic T lymphocytes (CTLs) influence the size of the reservoir in early HIV infection. Twenty-eight subjects with early HIV infection were recruited to receive cART and followed for 48 weeks. HIV reservoirs in peripheral CD4+ T cells measured by cell-associated proviral DNA and viral outgrowth cultures were determined at baseline and after 48 weeks of cART. At baseline, granzyme B and gamma interferon (IFN-γ) enzyme-linked immunosorbent spot (ELISpot) assays were performed with peptides spanning the HIV proteome. All subjects had detectable HIV-specific granzyme B and IFN-γ responses at baseline. The quantity and specificity of granzyme B responses did not correlate with IFN-γ responses. For granzyme B, Tat/Rev was the most dominant whereas for IFN-γ, Gag predominated. HIV-specific granzyme B T cell responses negatively correlated with HIV proviral loads at baseline and at 48 weeks and with replication-competent viral infectious units per million (IUPM) CD4+ T cells at baseline but not significantly at 48 weeks. Tat/Rev-, Env-, Gag-, and Vif-specific granzyme B responses correlated most strongly with reservoir control. There was no correlation of HIV-specific IFN-γ responses with reservoir size at baseline or at 48 weeks. The majority of granzyme B responses were contributed by CD8+ T cells. Thus, our findings suggest that the induction of potent granzyme B-producing CTLs to Tat, Rev, Env, Gag, and Vif during early infection may be able to prevent the establishment of a large viral reservoir, thereby facilitating a reduced HIV burden.IMPORTANCE A major barrier to the cure of human immunodeficiency virus type 1 (HIV-1) infection is the establishment of a viral reservoir that must be significantly reduced or eradicated entirely to enable a cure. Combined antiretroviral therapy (cART) alone is unable to clear this viral reservoir. It has been shown that CD8+ cytotoxic T lymphocytes (CTLs) are important in controlling early HIV infection by reducing plasma viremia. However, it is not known if these HIV-specific CTLs influence the establishment of the viral reservoir in early HIV infection. We show that HIV-specific granzyme B responses targeting HIV Tat/Rev, Env, Gag, and Vif, but not IFN-γ responses, are associated with reduced virus reservoirs at baseline and at 48 weeks of cART. These findings shed light on the nature of the effector CTL response that might limit reservoir size with implications for cure research and HIV vaccines.


Asunto(s)
Linfocitos T CD4-Positivos/virología , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Citotoxinas/metabolismo , Reservorios de Enfermedades/virología , Granzimas/metabolismo , Infecciones por VIH/inmunología , Adulto , Antivirales , Linfocitos T CD8-positivos/virología , Humanos , Interferón gamma/metabolismo , Masculino , Persona de Mediana Edad , Adulto Joven
8.
J Virol ; 85(1): 254-63, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20980521

RESUMEN

The immunologic mechanisms underlying the faster progression of hepatitis C virus (HCV) disease in the presence of human immunodeficiency virus (HIV) coinfection are not clearly understood. T-cell cross-reactivity between HCV and influenza virus-specific epitopes has been associated with rapid progression of HCV disease (S. Urbani, B. Amadei, P. Fisicaro, M. Pilli, G. Missale, A. Bertoletti, and C. Ferrari, J. Exp. Med. 201:675-680, 2005). We asked whether T-cell cross-reactivity between HCV and HIV could exist during HCV/HIV coinfection and affect pathogenesis. Our search for amino acid sequence homology between the HCV and HIV proteomes revealed two similar HLA-A2-restricted epitopes, HIV-Gag (SLYNTVATL [HIV-SL9]) and HCV-NS5b (ALYDVVSKL [HCV-AL9]). We found that 4 out of 20 HLA-A2-positive (HLA-A2(+)) HIV-infected individuals had CD8(+) T cells that recognized both the HIV-SL9 and HCV-AL9 epitopes. However, the AL9 epitope was generally shown to be a weak agonist. Although HCV-monoinfected individuals in our study did not show AL9-specific responses, we found that about half of HCV/HIV-coinfected individuals had dual responses to both epitopes. High dual T-cell recognition among coinfected subjects was usually due to separate T-cell populations targeting each epitope, as determined by pentamer staining. The one individual demonstrating cross-reactive T cells to both epitopes showed the most advanced degree of liver disease. In coinfected individuals, we observed a positive correlation between the magnitudes of T-cell responses to both the SL9 and the AL9 epitopes, which was also positively associated with the clinical parameter of liver damage. Thus, we find that HIV infection induces T cells that can cross-react to heterologous viruses or prime for T cells that are closely related in sequence. However, the induction of cross-reactive T cells may not be associated with control of disease caused by the heterologous virus. This demonstrates that degeneracy of HIV-specific T cells may play a role in the immunopathology of HCV/HIV coinfection.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Epítopos de Linfocito T/inmunología , Productos del Gen gag/inmunología , Infecciones por VIH/complicaciones , Antígeno HLA-A2/metabolismo , Hepatitis C/complicaciones , Proteínas no Estructurales Virales/inmunología , Secuencia de Aminoácidos , Linfocitos T CD8-positivos/virología , Reacciones Cruzadas , Epítopos de Linfocito T/química , Epítopos de Linfocito T/genética , Productos del Gen gag/química , Productos del Gen gag/genética , Infecciones por VIH/inmunología , Infecciones por VIH/virología , VIH-1/inmunología , Antígeno HLA-A2/genética , Hepacivirus/inmunología , Hepatitis C/inmunología , Hepatitis C/virología , Humanos , Datos de Secuencia Molecular , Proteínas no Estructurales Virales/química , Proteínas no Estructurales Virales/genética
9.
J Immunol ; 185(1): 498-506, 2010 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-20519650

RESUMEN

We examined the role of CD4(+) T cell IL-21 production in viral control of HIV infection. HIV-infected individuals had greater circulating IL-21-producing CD4(+) T cells in blood compared with uninfected volunteers. HIV-specific IL-21-producing CD4(+) T cells were detected in blood during untreated acute and chronic HIV infection, and elevated frequencies of these cells correlated with relative viral control. These cells had an effector memory or end effector phenotype and expressed CXCR5. HIV-specific CD8(+) T cells exhibited high levels of IL-21R, indicating sensitivity to IL-21. Low or aviremic long-term nonprogressors, however, showed absent or low HIV-specific IL-21 CD4(+) T cells, but more easily detectable HIV-specific IL-2-producing CD4(+) T cells, suggesting changing requirements for particular gamma-chain cytokines depending on Ag abundance. Thus, IL-21-producing CD4(+) T cells are induced in viremic HIV infection and likely contribute to viral control by affecting CD8(+) T cell maintenance.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/virología , Epítopos de Linfocito T/inmunología , Infecciones por VIH/inmunología , Infecciones por VIH/virología , VIH-1/inmunología , Interleucinas/biosíntesis , Activación de Linfocitos/inmunología , Enfermedad Aguda , Linfocitos T CD4-Positivos/patología , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/patología , Linfocitos T CD8-positivos/virología , Enfermedad Crónica , Progresión de la Enfermedad , Infecciones por VIH/patología , VIH-1/genética , Memoria Inmunológica , Inmunofenotipificación , Carga Viral/inmunología
10.
Sci Adv ; 8(3): eabj9815, 2022 Jan 21.
Artículo en Inglés | MEDLINE | ID: mdl-35044832

RESUMEN

Safe and effective vaccines are needed to end the COVID-19 pandemic. Here, we report the preclinical development of a lipid nanoparticle­formulated SARS-CoV-2 mRNA vaccine, PTX-COVID19-B. PTX-COVID19-B was chosen among three candidates after the initial mouse vaccination results showed that it elicited the strongest neutralizing antibody response against SARS-CoV-2. Further tests in mice and hamsters indicated that PTX-COVID19-B induced robust humoral and cellular immune responses and completely protected the vaccinated animals from SARS-CoV-2 infection in the lung. Studies in hamsters also showed that PTX-COVID19-B protected the upper respiratory tract from SARS-CoV-2 infection. Mouse immune sera elicited by PTX-COVID19-B vaccination were able to neutralize SARS-CoV-2 variants of concern, including the Alpha, Beta, Gamma, and Delta lineages. No adverse effects were induced by PTX-COVID19-B in either mice or hamsters. Based on these results, PTX-COVID19-B was authorized by Health Canada to enter clinical trials in December 2020 with a phase 2 clinical trial ongoing.


Asunto(s)
Vacunas contra la COVID-19/inmunología , COVID-19/prevención & control , SARS-CoV-2/inmunología , Glicoproteína de la Espiga del Coronavirus/inmunología , Vacunas Sintéticas/inmunología , Vacunas de ARNm/inmunología , Animales , Anticuerpos Neutralizantes/sangre , Anticuerpos Antivirales/sangre , Recuento de Linfocito CD4 , Linfocitos T CD8-positivos/inmunología , COVID-19/inmunología , Vacunas contra la COVID-19/efectos adversos , Canadá , Línea Celular , Cricetinae , Evaluación Preclínica de Medicamentos , Femenino , Células HEK293 , Humanos , Inmunidad Celular/inmunología , Inmunidad Humoral/inmunología , Liposomas/farmacología , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Nanopartículas , Glicoproteína de la Espiga del Coronavirus/genética , Células TH1/inmunología
11.
Eur J Immunol ; 40(9): 2493-505, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20623550

RESUMEN

Co-infection of HCV with HIV has been associated with more rapid progression of HCV-related disease. HCV-specific T-cell immune responses, which are essential for disease control, are attenuated in co-infection with HIV. T-cell exhaustion has recently been implicated in the deficient control of chronic viral infections. In the current study, we investigated the role of programmed death-1 (PD-1) and T-cell immunoglobulin and mucin domain-containing molecule-3 (Tim-3) expression in T-cell exhaustion during HCV/HIV co-infection. We show that in HCV/HIV co-infection, both total and HCV-specific T cells co-express Tim-3 and PD-1 in significantly higher frequencies, compared with HCV mono-infection. Co-expression of these two markers on HCV-specific CD8(+) T cells positively correlated with a clinical parameter of liver disease progression. HCV-specific CD8(+) T cells showed greater frequencies of Tim-3/PD-1 co-expression than HIV-specific CD8(+) T cells, which may indicate a greater degree of exhaustion in the former. Blocking Tim-3 or PD-1 pathways restored both HIV- and HCV-specific CD8(+) T-cell expansion in the blood of co-infected individuals. These data demonstrate that co-expression of Tim-3 and PD-1 may play a significant role in HCV-specific T-cell dysfunction, especially in the setting of HIV co-infection.


Asunto(s)
Antígenos CD/metabolismo , Proteínas Reguladoras de la Apoptosis/metabolismo , Linfocitos T CD8-positivos/metabolismo , Infecciones por VIH/inmunología , VIH-1/inmunología , Hepacivirus/inmunología , Hepatitis C/inmunología , Proteínas de la Membrana/metabolismo , Anticuerpos Bloqueadores/farmacología , Antígenos CD/genética , Antígenos CD/inmunología , Antígenos Virales/inmunología , Proteínas Reguladoras de la Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/inmunología , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/patología , Linfocitos T CD8-positivos/virología , Proliferación Celular/efectos de los fármacos , Separación Celular , Células Cultivadas , Progresión de la Enfermedad , Femenino , Citometría de Flujo , Infecciones por VIH/complicaciones , Infecciones por VIH/patología , Infecciones por VIH/fisiopatología , VIH-1/patogenicidad , Antígenos HLA-A/inmunología , Antígenos HLA-A/metabolismo , Antígeno HLA-A2 , Hepacivirus/patogenicidad , Receptor 2 Celular del Virus de la Hepatitis A , Hepatitis C/complicaciones , Hepatitis C/patología , Hepatitis C/fisiopatología , Humanos , Hígado/inmunología , Hígado/patología , Hígado/virología , Masculino , Proteínas de la Membrana/genética , Proteínas de la Membrana/inmunología , Receptor de Muerte Celular Programada 1
12.
Cell Microbiol ; 12(12): 1703-17, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20735437

RESUMEN

Clinical and epidemiological research provides evidence for a positive correlation between Neisseria gonorrhoeae infection and HIV transmission; however, mechanistic studies examining this relationship have yielded conflicting results. To explore this interaction, we exposed ex vivo cultured peripheral blood cells from acute HIV(+) individuals to N. gonorrhoeae. Unexpectedly, we observed a profound inhibition in HIV-1 replication in the ex vivo cultures, and this was recapitulated when peripheral blood mononuclear cells (PBMCs) from healthy donors were co-infected with HIV-1 and N. gonorrhoeae. Next, we established that gonococcal-infected PBMCs liberated a soluble factor that effectively blocked HIV-1 replication. Cytokine analyses and antibody blocking experiments revealed that the type I interferon, interferon-α (IFNα), was expressed upon exposure to N. gonorrhoeae and was responsible for the inhibition of HIV-1. Intracellular staining, TLR9-blocking and cell depletion-based studies demonstrated that the IFNα was elicited by plasmacytoid dendritic cells (pDCs) in a TLR9-dependent manner. The pDC response to N. gonorrhoeae was unexpected given pDCs more established role in innate defence against intracellular pathogens, suggesting this may be a bacterial immune evasion strategy. In the context of HIV, this overcomes the virus's otherwise effective avoidance of the interferon response and represents a previously unrecognized intersection between these two sexually transmitted pathogens.


Asunto(s)
Células Dendríticas/inmunología , VIH-1/crecimiento & desarrollo , Interferón-alfa/inmunología , Leucocitos Mononucleares/microbiología , Leucocitos Mononucleares/virología , Neisseria gonorrhoeae/crecimiento & desarrollo , Receptor Toll-Like 9/inmunología , Células Cultivadas , Infecciones por VIH/inmunología , Infecciones por VIH/microbiología , Infecciones por VIH/virología , VIH-1/inmunología , Humanos , Interferón-alfa/biosíntesis , Neisseria gonorrhoeae/inmunología , Replicación Viral
13.
Life Sci Alliance ; 4(11)2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34548381

RESUMEN

The mechanisms inducing exhaustion of HIV-specific CD8+ T cells are not fully understood. Metabolic programming directly influences T-cell differentiation, effector function, and memory. We evaluated metabolic profiles of ex vivo CD8+ T cells in HIV-infected individuals. The baseline oxygen consumption rate of CD8+ T cells was elevated in all infected individuals and CD8+ T cells were working at maximal respiratory capacity. The baseline glycolysis rate was enhanced only during early untreated HIV and in viral controllers, but glycolytic capacity was conserved at all stages of infection. CD8+ T-cell mTOR activity was found to be reduced. Enhanced glycolysis was crucial for HIV-specific killing of CD8+ T cells. CD8+ T-cell cytoplasmic GAPDH content was reduced in HIV, but less in early infection and viral controllers. Thus, CD8+ T-cell exhaustion in HIV is characterized by reduced glycolytic activity, enhanced OXPHOS demands, dysregulated mTOR, and reduced cytoplasmic GAPDH. These data provide potential metabolic strategies to reverse CD8+ T-cell dysfunction in HIV.


Asunto(s)
Linfocitos T CD8-positivos/metabolismo , Glucólisis/inmunología , Infecciones por VIH/metabolismo , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/fisiología , Gliceraldehído-3-Fosfato Deshidrogenasa (Fosforilante)/metabolismo , Glucólisis/fisiología , Infecciones por VIH/inmunología , VIH-1/patogenicidad , Humanos , Activación de Linfocitos , Fosforilación Oxidativa , Consumo de Oxígeno/inmunología , Consumo de Oxígeno/fisiología , Serina-Treonina Quinasas TOR/metabolismo
14.
Nat Commun ; 12(1): 1806, 2021 03 22.
Artículo en Inglés | MEDLINE | ID: mdl-33753733

RESUMEN

Better diagnostic tools are needed to combat the ongoing COVID-19 pandemic. Here, to meet this urgent demand, we report a homogeneous immunoassay to detect IgG antibodies against SARS-CoV-2. This serological assay, called SATiN, is based on a tri-part Nanoluciferase (tNLuc) approach, in which the spike protein of SARS-CoV-2 and protein G, fused respectively to two different tNLuc tags, are used as antibody probes. Target engagement of the probes allows reconstitution of a functional luciferase in the presence of the third tNLuc component. The assay is performed directly in the liquid phase of patient sera and enables rapid, quantitative and low-cost detection. We show that SATiN has a similar sensitivity to ELISA, and its readouts are consistent with various neutralizing antibody assays. This proof-of-principle study suggests potential applications in diagnostics, as well as disease and vaccination management.


Asunto(s)
Anticuerpos Antivirales/sangre , Prueba de COVID-19/métodos , COVID-19/diagnóstico , Inmunoensayo/métodos , Luciferasas/metabolismo , SARS-CoV-2/inmunología , SARS-CoV-2/aislamiento & purificación , Anticuerpos Neutralizantes/sangre , Anticuerpos Antivirales/inmunología , COVID-19/sangre , COVID-19/virología , Ensayo de Inmunoadsorción Enzimática , Células HEK293 , Humanos , Inmunoglobulina G/sangre , Inmunoglobulina M/sangre , Glicoproteína de la Espiga del Coronavirus/inmunología
15.
J Virol ; 83(17): 8722-32, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19553327

RESUMEN

The presence of interleukin-2 (IL-2)-producing human immunodeficiency virus type 1 (HIV-1)-specific CD4(+) T-cell responses has been associated with the immunological control of HIV-1 replication; however, the causal relationship between these factors remains unclear. Here we show that IL-2-producing HIV-1-specific CD4(+) T cells can be cloned from acutely HIV-1-infected individuals. Despite the early presence of these cells, each of the individuals in the present study exhibited progressive disease, with one individual showing rapid progression. In this rapid progressor, three IL-2-producing HIV-1 Gag-specific CD4(+) T-cell responses were identified and mapped to the following optimal epitopes: HIVWASRELER, REPRGSDIAGT, and FRDYVDRFYKT. Responses to these epitopes in peripheral blood mononuclear cells were monitored longitudinally to >1 year postinfection, and contemporaneous circulating plasma viruses were sequenced. A variant of the FRDYVDRFYKT epitope sequence, FRDYVDQFYKT, was observed in 1/21 plasma viruses sequenced at 5 months postinfection and 1/10 viruses at 7 months postinfection. This variant failed to stimulate the corresponding CD4(+) T-cell clone and thus constitutes an escape mutant. Responses to each of the three Gag epitopes were rapidly lost, and this loss was accompanied by a loss of antigen-specific cells in the periphery as measured by using an FRDYVDRFYKT-presenting major histocompatibility complex class II tetramer. Highly active antiretroviral therapy was associated with the reemergence of FRDYVDRFYKT-specific cells by tetramer. Thus, our data support that IL-2-producing HIV-1-specific CD4(+) T-cell responses can exert immune pressure during early HIV-1 infection but that the inability of these responses to enforce enduring control of viral replication is related to the deletion and/or dysfunction of HIV-1-specific CD4(+) T cells rather than to the fixation of escape mutations at high frequencies.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Infecciones por VIH/inmunología , Infecciones por VIH/virología , VIH-1/genética , VIH-1/inmunología , Interleucina-2/metabolismo , Mutación Missense/inmunología , Adulto , Animales , Mapeo Epitopo , Epítopos de Linfocito T/genética , Epítopos de Linfocito T/inmunología , VIH-1/aislamiento & purificación , Humanos , Tolerancia Inmunológica , Estudios Longitudinales , Masculino , Productos del Gen gag del Virus de la Inmunodeficiencia Humana/genética , Productos del Gen gag del Virus de la Inmunodeficiencia Humana/inmunología
16.
Vaccine ; 38(9): 2149-2159, 2020 02 24.
Artículo en Inglés | MEDLINE | ID: mdl-32014267

RESUMEN

HIV-1 envelope (Env)-specific antibody present at mucosal surfaces can block entry of HIV-1 into these portals and thus should be elicited by an HIV-1 preventive vaccine. Since three molecules of tumor necrosis factor superfamily (TNFSF), APRIL, BAFF, and CD40L, could promote mucosal antibody responses, we made fusion constructs of them with an HIV-1 gp140 trimer and tested the mucosal gp140-specific antibody elicited by the fusion constructs in mice using a DNA prime-protein boost vaccination regimen. The fusion constructs formed trimers and displayed both broadly neutralizing antibody epitopes and non-broadly neutralizing antibody epitopes. Compared with the control construct, trimeric gp140, trimeric gp140-APRIL and gp140-BAFF fusion proteins mildly promoted B cell proliferation in vitro, enhanced HIV-1 gp140-binding IgG responses in vaginal lavage or fecal pellets, respectively, and decreased HIV-1 gp140-binding IgA in sera. Gp140-APRIL also augmented HIV-1 gp140-binding IgG in sera. Surprisingly, gp140-CD40L did not promote B cell proliferation in vitro and inhibited mucosal and systemic HIV-1 gp140-binding IgG or IgA. These results suggest that APRIL and BAFF should be further explored as molecular adjuvants for HIV-1 vaccines to enhance mucosal antibody responses, but covalent fusion of TNFSFs to gp140 may hinder their adjuvancy due to steric interactions.


Asunto(s)
Vacunas contra el SIDA/inmunología , Factor Activador de Células B/inmunología , Ligando de CD40/inmunología , Infecciones por VIH , Inmunidad Mucosa , Miembro 13 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/inmunología , Productos del Gen env del Virus de la Inmunodeficiencia Humana/inmunología , Animales , Anticuerpos Neutralizantes/sangre , Formación de Anticuerpos , Femenino , Anticuerpos Anti-VIH/sangre , Infecciones por VIH/prevención & control , VIH-1/inmunología , Inmunoglobulina A/sangre , Inmunoglobulina G/sangre , Ratones , Proteínas Recombinantes de Fusión/inmunología
17.
Sci Immunol ; 5(52)2020 10 08.
Artículo en Inglés | MEDLINE | ID: mdl-33033173

RESUMEN

While the antibody response to SARS-CoV-2 has been extensively studied in blood, relatively little is known about the antibody response in saliva and its relationship to systemic antibody levels. Here, we profiled by enzyme-linked immunosorbent assays (ELISAs) IgG, IgA and IgM responses to the SARS-CoV-2 spike protein (full length trimer) and its receptor-binding domain (RBD) in serum and saliva of acute and convalescent patients with laboratory-diagnosed COVID-19 ranging from 3-115 days post-symptom onset (PSO), compared to negative controls. Anti-SARS-CoV-2 antibody responses were readily detected in serum and saliva, with peak IgG levels attained by 16-30 days PSO. Longitudinal analysis revealed that anti-SARS-CoV-2 IgA and IgM antibodies rapidly decayed, while IgG antibodies remained relatively stable up to 105 days PSO in both biofluids. Lastly, IgG, IgM and to a lesser extent IgA responses to spike and RBD in the serum positively correlated with matched saliva samples. This study confirms that serum and saliva IgG antibodies to SARS-CoV-2 are maintained in the majority of COVID-19 patients for at least 3 months PSO. IgG responses in saliva may serve as a surrogate measure of systemic immunity to SARS-CoV-2 based on their correlation with serum IgG responses.


Asunto(s)
Anticuerpos Antivirales/sangre , Antígenos Virales/inmunología , Betacoronavirus/inmunología , Infecciones por Coronavirus/inmunología , Neumonía Viral/inmunología , Saliva/inmunología , Glicoproteína de la Espiga del Coronavirus/inmunología , Adulto , COVID-19 , Infecciones por Coronavirus/virología , Estudios Transversales , Ensayo de Inmunoadsorción Enzimática , Femenino , Humanos , Inmunoglobulina A/sangre , Inmunoglobulina A/inmunología , Inmunoglobulina G/sangre , Inmunoglobulina G/inmunología , Inmunoglobulina M/sangre , Inmunoglobulina M/inmunología , Estudios Longitudinales , Masculino , Persona de Mediana Edad , Pandemias , Neumonía Viral/virología , SARS-CoV-2
18.
J Virol ; 82(13): 6767-71, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18434403

RESUMEN

T(H)-17 cells have been shown to play a role in bacterial defense, acute inflammation, and autoimmunity. We examined the role of interleukin 17 (IL-17) production in human immunodeficiency virus type 1 (HIV-1) infection. Both HIV-1- and cytomegalovirus (CMV)-specific IL-17-producing CD4(+) T cells were detectable in early HIV-1 infection but were reduced to nondetectable levels in chronic and nonprogressive HIV-1 infection. IL-17-producing CMV-specific cells were not detected in blood from HIV-1-uninfected normal volunteers. Virus-specific T(H)-17 cells could coexpress other cytokines and could express CCR4 or CXCR3. Although the etiology of these cells has yet to be established, we propose that microbial translocation may induce them.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Infecciones por VIH/inmunología , VIH-1/inmunología , Interleucina-17/inmunología , Animales , Western Blotting , Citometría de Flujo , Humanos , Ratones
19.
JCI Insight ; 2(17)2017 09 07.
Artículo en Inglés | MEDLINE | ID: mdl-28878119

RESUMEN

Eradication of the HIV-1 latent reservoir represents the current paradigm to developing a cure for AIDS. HIV-1 has evolved multiple mechanisms to evade CD8 T cell responses, including HIV-1 Nef-mediated downregulation of MHC-I from the surface of infected cells. Nef transcripts and protein are detectable in samples from aviremic donors, suggesting that Nef expression in latently HIV-1-infected CD4 T cells protects them from immune-mediated clearance. Here, we tested 4 small molecule inhibitors of HIV-1 Nef in an in vitro primary CD4 T cell latency model and measured the ability of autologous ex vivo or HIV-1 peptide-expanded CD8 T cells to recognize and kill latently infected cells as a function of inhibitor treatment. Nef inhibition enhanced cytokine secretion by autologous CD8 T cells against latently HIV-1-infected targets in an IFN-γ release assay. Additionally, CD8 T cell-mediated elimination of latently HIV-1-infected cells was significantly enhanced following Nef blockade, measured as a reduction in the frequency of infected cells and Gag protein in cultures following viral outgrowth assays. We demonstrate for the first time to our knowledge that Nef blockade, in combination with HIV-specific CD8 T cell expansion, might be a feasible strategy to target the HIV-1 latent reservoir that should be tested further in vivo.


Asunto(s)
Fármacos Anti-VIH/farmacología , Productos del Gen nef/antagonistas & inhibidores , VIH-1/metabolismo , Latencia del Virus , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/virología , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/virología , Células Cultivadas , Regulación hacia Abajo , Productos del Gen nef/genética , Productos del Gen nef/metabolismo , VIH-1/efectos de los fármacos , Humanos , Complejo Mayor de Histocompatibilidad/inmunología
20.
Immunol Res ; 35(1-2): 89-102, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-17003512

RESUMEN

Globally, at least 60 million people have been infected with the human immunodeficiency virus type 1 (HIV-1), the majority of whom will develop the acquired immunodeficiency syndrome (AIDS) leading to tremendous morbidity and the mortality. Understanding the immunopathogenesis of AIDS and the immune correlates of viral protection are necessary to develop effective vaccines and immunotherapies. A major focus of our laboratory has been to understand the CD4+ T cell immune response directed against HIV- 1, and to determine mechanisms of T cell dysfunction that lead to viral escape. In addition, we are interested in evaluating the TNF-TNFR family members as potential molecular adjuvants that could be incorporated into vaccines which could be used to further boost T cell immunogenicity in healthy or HIV-1-infected individuals, as many of these molecules have been shown to replace the functions of CD4+ T cell help.


Asunto(s)
Síndrome de Inmunodeficiencia Adquirida/inmunología , Linfocitos T CD4-Positivos/inmunología , VIH-1/inmunología , Vacunas contra el SIDA/inmunología , Vacunas contra el SIDA/uso terapéutico , Síndrome de Inmunodeficiencia Adquirida/prevención & control , Humanos , Sistema Inmunológico , Linfocitos T Citotóxicos/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA