Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
STAR Protoc ; 4(1): 101995, 2023 03 17.
Artículo en Inglés | MEDLINE | ID: mdl-36602901

RESUMEN

Specialized T cells are located in the nasal cavity and act as the first line of defense against respiratory viral infection. Here, we present a protocol for the detection and characterization of antigen-specific nasal-resident T cells. We detail steps for localized nasal swabbing to collect the nasal samples. We then describe IFN-γ ELISpot and an activation-induced marker assay to detect and characterize antigen-specific nasal-resident T cells. For complete details on the use and execution of this protocol, please refer to Lim et al. (2022).1.


Asunto(s)
Bioensayo , Linfocitos T , Humanos
2.
J Exp Med ; 219(10)2022 10 03.
Artículo en Inglés | MEDLINE | ID: mdl-35972472

RESUMEN

Rapid recognition of SARS-CoV-2-infected cells by resident T cells in the upper airway might provide an important layer of protection against COVID-19. Whether parenteral SARS-CoV-2 vaccination or infection induces nasal-resident T cells specific for distinct SARS-CoV-2 proteins is unknown. We isolated T cells from the nasal mucosa of COVID-19 vaccinees who either experienced SARS-CoV-2 infection after vaccination (n = 34) or not (n = 16) and analyzed their phenotype, SARS-CoV-2 specificity, function, and persistence. Nasal-resident SARS-CoV-2-specific CD8+ and CD4+ T cells were detected almost exclusively in vaccinees who experienced SARS-CoV-2 breakthrough infection. Importantly, the Spike-specific T cells primed by vaccination did not suppress the induction of T cells specific for other SARS-CoV-2 proteins. The nasal-resident T cell responses persisted for ≥140 d, with minimal sign of waning. These data highlight the importance of viral nasal challenge in the formation of SARS-CoV-2-specific antiviral immunity at the site of primary infection and further define the immunological features of SARS-CoV-2 hybrid immunity.


Asunto(s)
COVID-19 , Anticuerpos Antivirales , Linfocitos T CD4-Positivos , Linfocitos T CD8-positivos , Vacunas contra la COVID-19 , Humanos , SARS-CoV-2 , Vacunación
3.
Cell Rep Med ; 3(11): 100793, 2022 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-36257326

RESUMEN

Unlike mRNA vaccines based only on the spike protein, inactivated severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) vaccines should induce a diversified T cell response recognizing distinct structural proteins. Here, we perform a comparative analysis of SARS-CoV-2-specific T cells in healthy individuals following vaccination with inactivated SARS-CoV-2 or mRNA vaccines. Relative to spike mRNA vaccination, inactivated vaccines elicit a lower magnitude of spike-specific T cells, but the combination of membrane, nucleoprotein, and spike-specific T cell response is quantitatively comparable with the sole spike T cell response induced by mRNA vaccine, and they efficiently tolerate the mutations characterizing the Omicron lineage. However, this multi-protein-specific T cell response is not mediated by a coordinated CD4 and CD8 T cell expansion but by selective priming of CD4 T cells. These findings can help in understanding the role of CD4 and CD8 T cells in the efficacy of the different vaccines to control severe COVID-19 after Omicron infection.


Asunto(s)
COVID-19 , Vacunas Virales , Humanos , SARS-CoV-2/genética , Vacunas contra la COVID-19 , Vacunas Virales/genética , ARN Mensajero/genética , COVID-19/prevención & control , Vacunas de ARNm
4.
Med ; 3(2): 104-118.e4, 2022 Feb 11.
Artículo en Inglés | MEDLINE | ID: mdl-35072129

RESUMEN

BACKGROUND: Protection offered by coronavirus disease 2019 (COVID-19) vaccines wanes over time, requiring an evaluation of different boosting strategies to revert such a trend and enhance the quantity and quality of Spike-specific humoral and cellular immune responses. These immunological parameters in homologous or heterologous vaccination boosts have thus far been studied for mRNA and ChAdOx1 nCoV-19 vaccines, but knowledge on individuals who received a single dose of Ad26.COV2.S is lacking. METHODS: We studied Spike-specific humoral and cellular immunity in Ad26.COV2.S-vaccinated individuals (n = 55) who were either primed with Ad26.COV2.S only (n = 13) or were boosted with a homologous (Ad26.COV2.S, n = 28) or heterologous (BNT162b2, n = 14) second dose. We compared our findings with the results found in individuals vaccinated with a single (n = 16) or double (n = 44) dose of BNT162b2. FINDINGS: We observed that a strategy of heterologous vaccination enhanced the quantity and breadth of both Spike-specific humoral and cellular immunity in Ad26.COV2.S-vaccinated individuals. In contrast, the impact of the homologous boost was quantitatively minimal in Ad26.COV2.S-vaccinated individuals, and Spike-specific antibodies and T cells were narrowly focused to the S1 region. CONCLUSIONS: Despite the small sample size of the study and the lack of well-defined correlates of protection against COVID-19, the immunological features detected support the utilization of a heterologous vaccine boost in individuals who received Ad26.COV2.S vaccination. FUNDING: This study is partially supported by the Singapore Ministry of Health's National Medical Research Council under its COVID-19 Research Fund (COVID19RF3-0060, COVID19RF-001, and COVID19RF-008), The Medical College St. Bartholomew's Hospital Trustees - Pump Priming Fund for SMD COVID-19 Research.


Asunto(s)
Ad26COVS1 , COVID-19 , Anticuerpos Neutralizantes , Vacuna BNT162 , COVID-19/prevención & control , Vacunas contra la COVID-19 , ChAdOx1 nCoV-19 , Humanos , SARS-CoV-2
5.
Nat Biotechnol ; 40(11): 1680-1689, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-35697804

RESUMEN

Fast, high-throughput methods for measuring the level and duration of protective immune responses to SARS-CoV-2 are needed to anticipate the risk of breakthrough infections. Here we report the development of two quantitative PCR assays for SARS-CoV-2-specific T cell activation. The assays are rapid, internally normalized and probe-based: qTACT requires RNA extraction and dqTACT avoids sample preparation steps. Both assays rely on the quantification of CXCL10 messenger RNA, a chemokine whose expression is strongly correlated with activation of antigen-specific T cells. On restimulation of whole-blood cells with SARS-CoV-2 viral antigens, viral-specific T cells secrete IFN-γ, which stimulates monocytes to produce CXCL10. CXCL10 mRNA can thus serve as a proxy to quantify cellular immunity. Our assays may allow large-scale monitoring of the magnitude and duration of functional T cell immunity to SARS-CoV-2, thus helping to prioritize revaccination strategies in vulnerable populations.


Asunto(s)
COVID-19 , SARS-CoV-2 , Humanos , SARS-CoV-2/genética , Inmunidad Celular , Reacción en Cadena de la Polimerasa , Linfocitos T
6.
Nat Commun ; 13(1): 4615, 2022 08 08.
Artículo en Inglés | MEDLINE | ID: mdl-35941158

RESUMEN

Understanding the impact of age on vaccinations is essential for the design and delivery of vaccines against SARS-CoV-2. Here, we present findings from a comprehensive analysis of multiple compartments of the memory immune response in 312 individuals vaccinated with the BNT162b2 SARS-CoV-2 mRNA vaccine. Two vaccine doses induce high antibody and T cell responses in most individuals. However, antibody recognition of the Spike protein of the Delta and Omicron variants is less efficient than that of the ancestral Wuhan strain. Age-stratified analyses identify a group of low antibody responders where individuals ≥60 years are overrepresented. Waning of the antibody and cellular responses is observed in 30% of the vaccinees after 6 months. However, age does not influence the waning of these responses. Taken together, while individuals ≥60 years old take longer to acquire vaccine-induced immunity, they develop more sustained acquired immunity at 6 months post-vaccination. A third dose strongly boosts the low antibody responses in the older individuals against the ancestral Wuhan strain, Delta and Omicron variants.


Asunto(s)
COVID-19 , Vacunas Virales , Anciano , Anticuerpos Antivirales , Formación de Anticuerpos , Vacuna BNT162 , COVID-19/prevención & control , Vacunas contra la COVID-19 , Humanos , Persona de Mediana Edad , SARS-CoV-2 , Vacunación , Vacunas Sintéticas , Vacunas de ARNm
7.
Immunobiology ; 225(1): 151868, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31784043

RESUMEN

The HLA-B*15:02 allele is associated with an increased risk of developing carbamazepine (CBZ)-induced Stevens-Johnson syndrome (SJS) and toxic epidermal necrolysis (TEN). Many studies, however, have demonstrated that a large majority of HLA-B*15:02 individuals are unlikely to develop the adverse drug reaction while on CBZ. This phenomenon suggests that other factors that modulate the allergic immune response, such as regulatory T cells (Tregs), might contribute to an uncontrolled immune response in SJS/TEN. Peripheral blood mononuclear cells (PBMCs) from 15 healthy HLA-B*15:02 carriers were isolated to investigate the role of Tregs in controlling the immune response towards CBZ. Recognition of CBZ was assessed using enzyme linked immunosorbent spot (ELISPOT) assay for IFN-γ, and the donor T-cell profiles were quantified by flow cytometry to differentiate CBZ responders from non-responders. As CD39 expression on Tregs promotes immune tolerance, we investigated the mechanisms of Treg suppression using inhibitors targeting the CD39/adenosinergic pathway. PBMCs from seven donors (responders) produced high levels of IFN-γ when re-exposed to CBZ, while eight donors (non-responders) did not. Flow cytometric analysis revealed that non-responders produced significantly higher frequencies of CD4+CD25+CD127loCD39+FoxP3+ Tregs compared to responders. CD39 inhibition using POM-1 inhibitor converted five of the eight non-responders into responders (P < 0.05). Higher frequencies of CD4+CD25+CD127loCD39+FoxP3+ Tregs was correlated with lower production of IFN-γ (P < 0.01). Our data suggest that CD4+CD25+CD127loCD39+FoxP3+ Tregs may play a role in promoting CBZ tolerance in HLA-B*15:02 carriers. The CD39/adenosinergic axis can be a potential target to alleviate the uncontrolled immune response during this adverse drug event.


Asunto(s)
Hipersensibilidad a las Drogas/inmunología , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos/inmunología , Antígeno HLA-B15/genética , Linfocitos T Reguladores/inmunología , Adenosina/metabolismo , Alérgenos/inmunología , Antígenos CD/metabolismo , Apirasa/metabolismo , Carbamazepina/inmunología , Estudios de Casos y Controles , Células Cultivadas , Ensayo de Immunospot Ligado a Enzimas , Factores de Transcripción Forkhead/metabolismo , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Antígeno HLA-B15/metabolismo , Humanos , Tolerancia Inmunológica/genética , Inmunidad Celular , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA