Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
AIDS ; 36(9): 1315-1317, 2022 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-35833684

RESUMEN

Current coronavirus disease 2019 (COVID-19) mRNA vaccines induce robust SARS-CoV-2-specific humoral and cellular responses in people with HIV (PWH). However, the rate of decay of effector immune responses has not been studied in these individuals. Here, we report a significant waning of antibody responses but persistent T-cell responses 6 months post vaccination in virally suppressed PWH with high CD4+ T-cell counts. These responses are comparable with those seen in healthy donors.


Asunto(s)
COVID-19 , Infecciones por VIH , Vacunas Virales , Animales , Anticuerpos Antivirales , COVID-19/prevención & control , Vacunas contra la COVID-19 , Humanos , Inmunidad Celular , Inmunidad Humoral , Ratones , Ratones Endogámicos BALB C , ARN Mensajero , SARS-CoV-2 , Vacunación , Vacunas Sintéticas , Vacunas de ARNm
3.
EBioMedicine ; 80: 104048, 2022 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-35533495

RESUMEN

BACKGROUND: COVID-19 mRNA vaccines elicit strong T and B cell responses to the SARS-CoV-2 spike glycoprotein in both SARS-CoV-2 naïve and experienced patients. However, it is unknown whether the post-vaccine CD4+ T cell responses seen in patients with a history of COVID-19 are due to restimulation of T cell clonotypes that were first activated during natural infection or if they are the result of new clones activated by the vaccine. METHODS: To address this question, we analyzed the SARS-CoV-2 spike glycoprotein-specific CD4+ T cell receptor repertoire before and after vaccination in 10 COVID-19 convalescent patients and 4 SARS-CoV-2 naïve healthy donor vaccine recipients. We used the viral Functional Expansion of Specific T cells (ViraFEST) assay to quantitatively identify specific SARS-CoV-2 and common cold coronavirus CD4+ T cell clonotypes post COVID-19 disease resolution and post mRNA SARS-CoV-2 vaccination. FINDINGS: We found that while some preexisting T cell receptor clonotypes persisted, the post-vaccine repertoire consisted mainly of vaccine-induced clones and was largely distinct from the repertoire induced by natural infection. Vaccination-induced clones led to an overall maintenance of the total number of SARS-CoV-2 reactive clonotypes over time through expansion of novel clonotypes only stimulated through vaccination. Additionally, we demonstrated that the vaccine preferentially induces T cells that are only specific for SARS-CoV-2 antigens, rather than T cells that cross-recognize SARS-CoV-2/common cold coronaviruses. INTERPRETATION: These data demonstrate that SARS-CoV-2 vaccination in patients with prior SARS-CoV-2 infection induces a new antigen-specific repertoire and sheds light on the differential immune responses induced by vaccination versus natural infection. FUNDING: Bloomberg∼Kimmel Institute for Cancer Immunotherapy, The Johns Hopkins University, The Bill and Melinda Gates Foundation, NCI U54CA260492, NIH.


Asunto(s)
COVID-19 , Resfriado Común , Vacunas Virales , Anticuerpos Antivirales , Linfocitos T CD4-Positivos , Vacunas contra la COVID-19 , Humanos , SARS-CoV-2 , Vacunación
4.
Clin Infect Dis ; 75(9): 1652-1654, 2022 10 29.
Artículo en Inglés | MEDLINE | ID: mdl-35438751

RESUMEN

We compared antibody and T-cell responses against the severe acute respiratory syndrome coronavirus 2 vaccine strain spike protein to responses against the Omicron variant in 15 messenger RNA vaccine recipients. While these individuals had significantly lower levels of antibodies that inhibited Omicron spike protein binding to ACE2, there was no difference in T-cell responses.


Asunto(s)
Vacunas contra la COVID-19 , COVID-19 , Humanos , SARS-CoV-2/genética , ARN Mensajero/genética , Linfocitos T , Anticuerpos Antivirales , Anticuerpos Neutralizantes , Vacunas de ARNm
5.
JCI Insight ; 7(10)2022 05 23.
Artículo en Inglés | MEDLINE | ID: mdl-35389888

RESUMEN

BackgroundBreakthrough SARS-CoV-2 infections in vaccinated individuals have been previously associated with suboptimal humoral immunity. However, less is known about breakthrough infections with the Omicron variant.MethodsWe analyzed SARS-CoV-2-specific antibody and cellular responses in healthy vaccine recipients who experienced breakthrough infections a median of 50 days after receiving a booster mRNA vaccine with an ACE2 binding inhibition assay and an ELISpot assay, respectively.ResultsWe found that high levels of antibodies inhibited vaccine strain spike protein binding to ACE2 but that lower levels inhibited Omicron variant spike protein binding to ACE2 in 4 boosted vaccine recipients prior to infection. The levels of antibodies that inhibited vaccine strain and Omicron spike protein binding after breakthrough in 18 boosted vaccine recipients were similar to levels seen in COVID-19-negative boosted vaccine recipients. In contrast, boosted vaccine recipients had significantly stronger T cell responses to both vaccine strain and Omicron variant spike proteins at the time of breakthrough.ConclusionOur data suggest that breakthrough infections with the Omicron variant can occur despite robust immune responses to the vaccine strain spike protein.FundingThis work was supported by the Johns Hopkins COVID-19 Vaccine-related Research Fund and by funds from the National Institute of Allergy and Infectious Disease intramural program as well as awards from the National Cancer Institute (U54CA260491) and the National Institutes of Allergy and Infectious Disease (K08AI156021 and U01AI138897).


Asunto(s)
COVID-19 , Enfermedades Transmisibles , Hipersensibilidad , Enzima Convertidora de Angiotensina 2 , Anticuerpos Antivirales , Vacuna BNT162 , COVID-19/prevención & control , Vacunas contra la COVID-19 , Humanos , Inmunidad , SARS-CoV-2 , Vacunas Sintéticas , Vacunas de ARNm
6.
JCI Insight ; 7(5)2022 03 08.
Artículo en Inglés | MEDLINE | ID: mdl-35104245

RESUMEN

Benchmarks for protective immunity from infection or severe disease after SARS-CoV-2 vaccination are still being defined. Here, we characterized virus neutralizing and ELISA antibody levels, cellular immune responses, and viral variants in 4 separate groups: healthy controls (HCs) weeks (early) or months (late) following vaccination in comparison with symptomatic patients with SARS-CoV-2 after partial or full mRNA vaccination. During the period of the study, most symptomatic breakthrough infections were caused by the SARS-CoV-2 Alpha variant. Neutralizing antibody levels in the HCs were sustained over time against the vaccine parent virus but decreased against the Alpha variant, whereas IgG titers and T cell responses against the parent virus and Alpha variant declined over time. Both partially and fully vaccinated patients with symptomatic infections had lower virus neutralizing antibody levels against the parent virus than the HCs, similar IgG antibody titers, and similar virus-specific T cell responses measured by IFN-γ. Compared with HCs, neutralization activity against the Alpha variant was lower in the partially vaccinated infected patients and tended to be lower in the fully vaccinated infected patients. In this cohort of breakthrough infections, parent virus neutralization was the superior predictor of breakthrough infections with the Alpha variant of SARS-CoV-2.


Asunto(s)
Inmunidad Adaptativa , Anticuerpos Antivirales/inmunología , Vacunas contra la COVID-19/farmacología , COVID-19/virología , SARS-CoV-2/inmunología , Vacunación/métodos , Vacunas Sintéticas/farmacología , Vacunas de ARNm/farmacología , Adulto , Anciano , COVID-19/epidemiología , COVID-19/prevención & control , Femenino , Estudios de Seguimiento , Humanos , Masculino , Persona de Mediana Edad , Pandemias , Vigilancia de la Población , Estudios Retrospectivos , Estados Unidos/epidemiología , Adulto Joven
7.
J Clin Invest ; 132(5)2022 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-35061630

RESUMEN

Recent studies have shown that vaccinated individuals harbor T cells that can cross-recognize SARS-CoV-2 and endemic human common cold coronaviruses. However, it is still unknown whether CD4+ T cells from vaccinated individuals recognize peptides from bat coronaviruses that may have the potential of causing future pandemics. In this study, we identified a SARS-CoV-2 spike protein epitope (S815-827) that is conserved in coronaviruses from different genera and subgenera, including SARS-CoV, MERS-CoV, multiple bat coronaviruses, and a feline coronavirus. Our results showed that S815-827 was recognized by 42% of vaccinated participants in our study who received the Pfizer-BioNTech (BNT162b2) or Moderna (mRNA-1273) COVID-19 vaccines. Using T cell expansion and T cell receptor sequencing assays, we demonstrated that S815-827-reactive CD4+ T cells from the majority of responders cross-recognized homologous peptides from at least 6 other diverse coronaviruses. Our results support the hypothesis that the current mRNA vaccines elicit T cell responses that can cross-recognize bat coronaviruses and thus might induce some protection against potential zoonotic outbreaks. Furthermore, our data provide important insights that inform the development of T cell-based pan-coronavirus vaccine strategies.


Asunto(s)
Vacuna nCoV-2019 mRNA-1273/inmunología , Vacuna BNT162/inmunología , Linfocitos T CD4-Positivos/inmunología , COVID-19/inmunología , Epítopos de Linfocito T/inmunología , Receptores de Antígenos de Linfocitos T/inmunología , SARS-CoV-2/inmunología , Glicoproteína de la Espiga del Coronavirus/inmunología , Vacuna nCoV-2019 mRNA-1273/administración & dosificación , Vacuna BNT162/administración & dosificación , COVID-19/prevención & control , Femenino , Humanos , Masculino , Péptidos/inmunología
8.
Clin Infect Dis ; 74(7): 1268-1270, 2022 04 09.
Artículo en Inglés | MEDLINE | ID: mdl-34293114

RESUMEN

Previous studies have shown that certain vaccines induce suboptimal responses in people living with human immunodeficiency virus (HIV, PLWH). However, responses to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) vaccines have not been fully characterized in these patients. Here we show that the BNT162b2 vaccine induces robust immune responses comparable to responses in healthy donors.


Asunto(s)
COVID-19 , Infecciones por VIH , Anticuerpos Antivirales , Vacuna BNT162 , COVID-19/prevención & control , VIH , Humanos , Inmunidad Celular , Inmunidad Humoral , SARS-CoV-2 , Vacunas Sintéticas , Vacunas de ARNm
9.
Clin Infect Dis ; 75(1): e898-e901, 2022 08 24.
Artículo en Inglés | MEDLINE | ID: mdl-34694362

RESUMEN

Little is known about the decay kinetics of coronavirus disease 2019 vaccine-elicited severe acute respiratory syndrome coronavirus 2-specific T cells. In this study we show a modest decline in the frequency of these T cells at 6 months and demonstrate robust expansion in response to antigen and recognition of spike peptides from the Delta variant.


Asunto(s)
COVID-19 , SARS-CoV-2 , Anticuerpos Antivirales , COVID-19/prevención & control , Vacunas contra la COVID-19 , Humanos , Linfocitos T , Vacunas Sintéticas , Vacunas de ARNm
10.
Front Immunol ; 12: 708355, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34394110

RESUMEN

HIV-specific CD8 T cells and broadly neutralizing antibodies (bNAbs) both contribute to the control of viremia, but in most cases, neither can completely suppress viral replication. To date, therapeutic vaccines have not been successful in eliciting HIV-specific CD8 T cell or bNAb responses that are capable of preventing long-term viral rebound upon ART cessation. These challenges suggest that a combinatorial approach that harnesses both bNAbs and CD8 T cell responses may be necessary for long term control of viral replication. In this study we demonstrate a synergistic interaction between CD8 T cells and bNAbs using an in vitro model. Our data suggest that this combinatorial approach is very effective at suppressing viral replication in vitro and should be considered in future therapeutic studies.


Asunto(s)
Anticuerpos ampliamente neutralizantes/inmunología , Linfocitos T CD8-positivos/inmunología , Anticuerpos Anti-VIH/inmunología , Células Cultivadas , Infecciones por VIH/inmunología , Humanos , Replicación Viral
11.
J Clin Invest ; 131(10)2021 05 17.
Artículo en Inglés | MEDLINE | ID: mdl-33822770

RESUMEN

Recent studies have shown T cell cross-recognition of SARS-CoV-2 and common cold coronavirus spike proteins. However, the effect of SARS-CoV-2 vaccines on T cell responses to common cold coronaviruses (CCCs) remains unknown. In this study, we analyzed CD4+ T cell responses to spike peptides from SARS-CoV-2 and 3 CCCs (HCoV-229E, HCoV-NL63, and HCoV-OC43) before and after study participants received Pfizer-BioNTech (BNT162b2) or Moderna (mRNA-1273) mRNA-based COVID-19 vaccines. Vaccine recipients showed broad T cell responses to the SARS-CoV-2 spike protein, and we identified 23 distinct targeted peptides in 9 participants, including 1 peptide that was targeted in 6 individuals. Only 4 of these 23 targeted peptides would potentially be affected by mutations in the UK (B.1.1.7) and South African (B.1.351) variants, and CD4+ T cells from vaccine recipients recognized the 2 variant spike proteins as effectively as they recognized the spike protein from the ancestral virus. Interestingly, we observed a 3-fold increase in the CD4+ T cell responses to HCoV-NL63 spike peptides after vaccination. Our results suggest that T cell responses elicited or enhanced by SARS-CoV-2 mRNA vaccines may be able to control SARS-CoV-2 variants and lead to cross-protection against some endemic coronaviruses.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Vacunas contra la COVID-19/inmunología , Coronavirus Humano 229E , Coronavirus Humano NL63 , Coronavirus Humano OC43 , ARN Mensajero , SARS-CoV-2 , Glicoproteína de la Espiga del Coronavirus , Adulto , Vacuna BNT162 , Coronavirus Humano 229E/genética , Coronavirus Humano 229E/inmunología , Coronavirus Humano NL63/genética , Coronavirus Humano NL63/inmunología , Coronavirus Humano OC43/genética , Coronavirus Humano OC43/inmunología , Reacciones Cruzadas , Femenino , Humanos , Masculino , Persona de Mediana Edad , ARN Mensajero/genética , ARN Mensajero/inmunología , SARS-CoV-2/genética , SARS-CoV-2/inmunología , Glicoproteína de la Espiga del Coronavirus/genética , Glicoproteína de la Espiga del Coronavirus/inmunología
12.
J Clin Invest ; 131(10)2021 05 17.
Artículo en Inglés | MEDLINE | ID: mdl-33830946

RESUMEN

BACKGROUNDRecent studies have reported T cell immunity to the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in unexposed donors, possibly due to crossrecognition by T cells specific for common cold coronaviruses (CCCs). True T cell crossreactivity, defined as the recognition by a single TCR of more than one distinct peptide-MHC ligand, has never been shown in the context of SARS-CoV-2.METHODSWe used the viral functional expansion of specific T cells (ViraFEST) platform to identify T cell responses crossreactive for the spike (S) glycoproteins of SARS-CoV-2 and CCCs at the T cell receptor (TCR) clonotype level in convalescent COVID-19 patients (CCPs) and SARS-CoV-2-unexposed donors. Confirmation of SARS-CoV-2/CCC crossreactivity and assessments of functional avidity were performed using a TCR cloning and transfection system.RESULTSMemory CD4+ T cell clonotypes that crossrecognized the S proteins of SARS-CoV-2 and at least one other CCC were detected in 65% of CCPs and unexposed donors. Several of these TCRs were shared among multiple donors. Crossreactive T cells demonstrated significantly impaired SARS-CoV-2-specific proliferation in vitro relative to monospecific CD4+ T cells, which was consistent with lower functional avidity of their TCRs for SARS-CoV-2 relative to CCC.CONCLUSIONSOur data confirm, for what we believe is the first time, the existence of unique memory CD4+ T cell clonotypes crossrecognizing SARS-CoV-2 and CCCs. The lower avidity of crossreactive TCRs for SARS-CoV-2 may be the result of antigenic imprinting, such that preexisting CCC-specific memory T cells have reduced expansive capacity upon SARS-CoV-2 infection. Further studies are needed to determine how these crossreactive T cell responses affect clinical outcomes in COVID-19 patients.FUNDINGNIH funding (U54CA260492, P30CA006973, P41EB028239, R01AI153349, R01AI145435-A1, R21AI149760, and U19A1088791) was provided by the National Institute of Allergy and Infectious Diseases, the National Cancer Institute, and the National Institute of Biomedical Imaging and Bioengineering. The Bloomberg~Kimmel Institute for Cancer Immunotherapy, The Johns Hopkins University Provost, and The Bill and Melinda Gates Foundation provided funding for this study.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , COVID-19/inmunología , Epítopos de Linfocito T/inmunología , Memoria Inmunológica , Receptores de Antígenos de Linfocitos T/inmunología , SARS-CoV-2/inmunología , Adulto , Anciano , Reacciones Cruzadas , Femenino , Humanos , Células Jurkat , Masculino , Persona de Mediana Edad
13.
J Clin Invest ; 130(12): 6631-6638, 2020 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-32966269

RESUMEN

BACKGROUNDT cell responses to the common cold coronaviruses have not been well characterized. Preexisting T cell immunity to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has been reported, and a recent study suggested that this immunity was due to cross-recognition of the novel coronavirus by T cells specific for the common cold coronaviruses.METHODSWe used the enzyme-linked immunospot (ELISPOT) assay to characterize the T cell responses against peptide pools derived from the spike protein of 3 common cold coronaviruses (HCoV-229E, HCoV-NL63, and HCoV-OC43) and SARS-CoV-2 in 21 healthy donors (HDs) who were seronegative for SARS-CoV-2 and had no known exposure to the virus. An in vitro expansion culture assay was also used to analyze memory T cell responses.RESULTSWe found responses to the spike protein of the 3 common cold coronaviruses in many of the donors. We then focused on HCoV-NL63 and detected broad T cell responses to the spike protein and identified 22 targeted peptides. Interestingly, only 1 study participant had a significant response to SARS-CoV-2 spike or nucleocapsid protein in the ELISPOT assay. In vitro expansion studies suggested that T cells specific for the HCoV-NL63 spike protein in this individual could also recognize SARS-CoV-2 spike protein peptide pools.CONCLUSIONHDs have circulating T cells specific for the spike proteins of HCoV-NL63, HCoV-229E, and HCoV-OC43. T cell responses to SARS-CoV-2 spike and nucleocapsid proteins were present in only 1 participant and were potentially the result of cross-recognition by T cells specific for the common cold coronaviruses. Further studies are needed to determine whether this cross-recognition influences coronavirus disease 2019 (COVID-19) outcomes.


Asunto(s)
COVID-19/inmunología , Resfriado Común/inmunología , Coronavirus Humano 229E/inmunología , Coronavirus Humano NL63/inmunología , Coronavirus Humano OC43/inmunología , Inmunidad Celular , SARS-CoV-2/inmunología , Linfocitos T/inmunología , Adulto , Reacciones Cruzadas , Femenino , Humanos , Masculino , Persona de Mediana Edad
14.
Front Immunol ; 11: 591, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32318072

RESUMEN

HIV-1 positive elite controllers or suppressors control viral replication without antiretroviral therapy, likely via CTL-mediated elimination of infected cells, and therefore represent a model of an HIV-1 functional cure. Efforts to cure HIV-1 accordingly rely on the existence or generation of antigen-specific cytotoxic T lymphocytes (CTL) to eradicate infected cells upon reversal of latency. Detecting and quantifying these HIV-1-specific CTL responses will be crucial for developing vaccine and T cell-based immunotherapies. A recently developed assay, called MANAFEST, uses T cell receptor (TCR) Vß sequencing of peptide-stimulated cultures followed by a bioinformatic pipeline to identify neoantigen-specific T cells in cancer patients. This assay is more sensitive than conventional immune assays and therefore has the possibility to identify HIV-1 antigenic targets that have not been previously explored for vaccine or T cell immunotherapeutic strategies. Here we show that a modified version of the MANAFEST assay, called ViraFEST, can identify memory CD8+ T cell responses against autologous HIV-1 Gag and Nef epitope variants in an elite suppressor. Nine TCR Vß clonotypes were identified and 6 of these were cross-reactive for autologous variants or known escape variants. Our findings are a proof of principle that the ViraFEST assay can be used to detect and monitor these responses for downstream use in immunotherapeutic treatment approaches.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Epítopos de Linfocito T/inmunología , Infecciones por VIH/inmunología , VIH-1/inmunología , Técnicas Inmunológicas , Receptores de Antígenos de Linfocitos T/análisis , Reacciones Cruzadas , Humanos , Receptores de Antígenos de Linfocitos T/inmunología
15.
Open Forum Infect Dis ; 7(4): ofaa082, 2020 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-32284948

RESUMEN

Resting CD4+ T cells are the best characterized component of the latent reservoir. Activation of these CD4+ T cells is needed to optimize transcription and viral replication, and this strategy has been used to measure the inducible reservoir. There are several methods that can be used to activate CD4+ T cells, and in this study, we compared 3 different strategies: the combination of the lectin phytohaemagglutinin (PHA) and irradiated allogeneic feeders, a combination of PHA and a superagonistic anti-CD28 antibody, and the combination of the protein kinase C agonist phorbol 12-myristate 13-acetate and the calcium ionophore ionomycin. We show that each strategy induces a different pattern of expression of activation markers on CD4+ T cells. However, the different activation strategies induced similar frequencies of latently infected CD4+ T cells from people living with HIV on suppressive antiretroviral therapy regimens to produce replication-competent virus. Furthermore, the frequency of infectious units per million induced by each regimen was positively correlated with the copies of intact proviral DNA per million CD4+ T cells. Our results suggest that no single pattern of activation marker expression is most associated with latency reversal and demonstrate that different immune activation strategies reverse latency in a low frequency of CD4+ T cells that harbor intact proviral DNA.

16.
AIDS ; 34(4): 641-643, 2020 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-31895150

RESUMEN

: Elite controllers or suppressors control viral replication without antiretroviral therapy. We used the intact proviral DNA assay to approximate the size of the inducible latent reservoir in elite suppressors and found that, while the median frequency of both total and intact proviral DNA was markedly lower than the frequencies seen in chronic progressors on antiretroviral therapy there was no significant difference in the ratio of intact to total proviral DNA between elite suppressors and chronic progressors.


Asunto(s)
Infecciones por VIH/virología , VIH-1/genética , Provirus/genética , Carga Viral , Recuento de Linfocito CD4 , Infecciones por VIH/inmunología , VIH-1/fisiología , Humanos , Replicación Viral
17.
JCI Insight ; 3(18)2018 09 20.
Artículo en Inglés | MEDLINE | ID: mdl-30232278

RESUMEN

Clonal expansion of T cells harboring replication-competent virus has recently been demonstrated in patients on suppressive antiretroviral therapy (ART) regimens. However, there has not been direct evidence of this phenomenon in settings of natural control, including in posttreatment controllers who maintain control of viral replication after treatment when ART is discontinued. We present a case of an individual who has had undetectable viral loads for more than 15 years following the cessation of ART. Using near-full-genome sequence analysis, we demonstrate that 9 of 12 replication-competent isolates cultured from this subject were identical and that this identity was maintained 6 months later. A similar pattern of replication-competent virus clonality was seen in a treatment-naive HLA-B*57 elite controller. In both cases, we show that CD8+ T cells are capable of suppressing the replication of the clonally expanded viruses in vitro. Our data suggest that, while clonal expansion of replication-competent virus can present a barrier to viral eradication, these viral isolates remain susceptible to HIV-specific immune responses and can be controlled in patients with long-term suppression of viral replication.


Asunto(s)
Infecciones por VIH/inmunología , Sobrevivientes de VIH a Largo Plazo , VIH-1/genética , Replicación Viral , Vacunas contra el SIDA , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/virología , Linfocitos T CD8-positivos/inmunología , Epítopos/inmunología , Epítopos de Linfocito T/genética , Epítopos de Linfocito T/inmunología , Infecciones por VIH/terapia , Infecciones por VIH/virología , Antígenos HLA-B , Humanos , Factores de Tiempo , Carga Viral , Productos del Gen gag del Virus de la Inmunodeficiencia Humana , Productos del Gen nef del Virus de la Inmunodeficiencia Humana
18.
PLoS One ; 13(2): e0192098, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29466365

RESUMEN

Heterologous immunity is an important aspect of the adaptive immune response. We hypothesized that this process could modulate the HIV-1-specific CD8+ T cell response, which has been shown to play an important role in HIV-1 immunity and control. We found that stimulation of peripheral blood mononuclear cells (PBMCs) from HIV-1-positive subjects with microbial peptides that were cross-reactive with immunodominant HIV-1 epitopes resulted in dramatic expansion of HIV-1-specific CD8+ T cells. Interestingly, the TCR repertoire of HIV-1-specific CD8+ T cells generated by ex vivo stimulation of PBMCs using HIV-1 peptide was different from that of cells stimulated with cross-reactive microbial peptides in some HIV-1-positive subjects. Despite these differences, CD8+ T cells stimulated with either HIV-1 or cross-reactive peptides effectively suppressed HIV-1 replication in autologous CD4+ T cells. These data suggest that exposure to cross-reactive microbial antigens can modulate HIV-1-specific immunity.


Asunto(s)
Linfocitos T CD8-positivos/efectos de los fármacos , Reacciones Cruzadas , Infecciones por VIH/inmunología , VIH-1/efectos de los fármacos , Linfocitos T CD8-positivos/inmunología , VIH-1/inmunología , VIH-1/fisiología , Humanos , Replicación Viral/efectos de los fármacos
19.
J Virus Erad ; 3(2): 85-89, 2017 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-28435692

RESUMEN

AIMS: Latently infected resting CD4 T cells represent a major barrier to HIV-1 eradication efforts. The standard assays used for measuring this reservoir induce activation of resting CD4 T cells with either phytohaemagglutinin (PHA) with irradiated feeder cells, or with anti-CD3 antibodies. We designed a study to compare the sensitivity of a new assay (based on the stimulation of CD4 T cells with anti-CD3 and anti-CD28 coated microbeads) with that of the traditional PHA- and feeder-based viral outgrowth assay. METHODS: Resting CD4 T cells from 10 HIV-1-infected patients on suppressive combination antiretroviral therapy (cART) regimens were cultured in the traditional PHA/feeders viral outgrowth assay and the new CD3/CD28 bead-based assay. Flow cytometry was used to assess the kinetics of activation of resting CD4 T cells in the two different assays. RESULTS: There was no significant difference in the sensitivity of the two assays. The median frequency of latently infected cells was 0.83 infectious units per million (IUPM) for the PHA/feeders assay and 0.54 IUPM with the CD3/CD28 bead-based assay. However, while virus was obtained from all 10 patients with the traditional PHA/feeders outgrowth assay, no virus was obtained from two of 10 patients with the novel anti-CD3/CD28 bead-based viral outgrowth assay (IUPM < 0.02). CONCLUSION: The new CD3/CD28 bead-based assay has comparable sensitivity to the PHA/feeders assay and does not require the addition of feeders, making it a simpler and less labour-intensive alternative to the standard PHA/feeders-based assay.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...