Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Biochemistry ; 62(17): 2517-2529, 2023 09 05.
Artigo em Inglês | MEDLINE | ID: mdl-37554055

RESUMO

Antigen conformation shapes CD4+ T-cell specificity through mechanisms of antigen processing, and the consequences for immunity may rival those from conformational effects on antibody specificity. CD4+ T cells initiate and control immunity to pathogens and cancer and are at least partly responsible for immunopathology associated with infection, autoimmunity, and allergy. The primary trigger for CD4+ T-cell maturation is the presentation of an epitope peptide in the MHC class II antigen-presenting protein (MHCII), most commonly on an activated dendritic cell, and then the T-cell responses are recalled by subsequent presentations of the epitope peptide by the same or other antigen-presenting cells. Peptide presentation depends on the proteolytic fragmentation of the antigen in an endosomal/lysosomal compartment and concomitant loading of the fragments into the MHCII, a multistep mechanism called antigen processing and presentation. Although the role of peptide affinity for MHCII has been well studied, the role of proteolytic fragmentation has received less attention. In this Perspective, we will briefly summarize evidence that antigen resistance to unfolding and proteolytic fragmentation shapes the specificity of the CD4+ T-cell response to selected viral envelope proteins, identify several remarkable examples in which the immunodominant CD4+ epitopes most likely depend on the interaction of processing machinery with antigen conformation, and outline how knowledge of antigen conformation can inform future efforts to design vaccines.


Assuntos
Linfócitos T CD4-Positivos , Epitopos de Linfócito T , Linfócitos T CD4-Positivos/metabolismo , Epitopos de Linfócito T/química , Epitopos de Linfócito T/metabolismo , Proteínas Virais de Fusão/metabolismo , Antígenos de Histocompatibilidade Classe II/metabolismo , Apresentação de Antígeno , Epitopos Imunodominantes/química , Epitopos Imunodominantes/metabolismo
2.
Sci Rep ; 12(1): 20117, 2022 11 22.
Artigo em Inglês | MEDLINE | ID: mdl-36418458

RESUMO

SARS-CoV-2 gains cell entry via angiotensin-converting enzyme (ACE) 2, a membrane-bound enzyme of the "alternative" (alt) renin-angiotensin system (RAS). ACE2 counteracts angiotensin II by converting it to potentially protective angiotensin 1-7. Using mass spectrometry, we assessed key metabolites of the classical RAS (angiotensins I-II) and alt-RAS (angiotensins 1-7 and 1-5) pathways as well as ACE and ACE2 concentrations in 159 patients hospitalized with COVID-19, stratified by disease severity (severe, n = 76; non-severe: n = 83). Plasma renin activity (PRA-S) was calculated as the sum of RAS metabolites. We estimated ACE activity using the angiotensin II:I ratio (ACE-S) and estimated systemic alt-RAS activation using the ratio of alt-RAS axis metabolites to PRA-S (ALT-S). We applied mixed linear models to assess how PRA-S and ACE/ACE2 concentrations affected ALT-S, ACE-S, and angiotensins II and 1-7. Median angiotensin I and II levels were higher with severe versus non-severe COVID-19 (angiotensin I: 86 versus 30 pmol/L, p < 0.01; angiotensin II: 114 versus 58 pmol/L, p < 0.05), demonstrating activation of classical RAS. The difference disappeared with analysis limited to patients not taking a RAS inhibitor (angiotensin I: 40 versus 31 pmol/L, p = 0.251; angiotensin II: 76 versus 99 pmol/L, p = 0.833). ALT-S in severe COVID-19 increased with time (days 1-6: 0.12; days 11-16: 0.22) and correlated with ACE2 concentration (r = 0.831). ACE-S was lower in severe versus non-severe COVID-19 (1.6 versus 2.6; p < 0.001), but ACE concentrations were similar between groups and correlated weakly with ACE-S (r = 0.232). ACE2 and ACE-S trajectories in severe COVID-19, however, did not differ between survivors and non-survivors. Overall RAS alteration in severe COVID-19 resembled severity of disease-matched patients with influenza. In mixed linear models, renin activity most strongly predicted angiotensin II and 1-7 levels. ACE2 also predicted angiotensin 1-7 levels and ALT-S. No single factor or the combined model, however, could fully explain ACE-S. ACE2 and ACE-S trajectories in severe COVID-19 did not differ between survivors and non-survivors. In conclusion, angiotensin II was elevated in severe COVID-19 but was markedly influenced by RAS inhibitors and driven by overall RAS activation. ACE-S was significantly lower with severe COVID-19 and did not correlate with ACE concentrations. A shift to the alt-RAS axis because of increased ACE2 could partially explain the relative reduction in angiotensin II levels.


Assuntos
COVID-19 , Hormônios Peptídicos , Humanos , Enzima de Conversão de Angiotensina 2 , Sistema Renina-Angiotensina , Angiotensina I , Angiotensina II , SARS-CoV-2 , Renina , Anti-Hipertensivos
3.
Front Immunol ; 13: 946318, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35928813

RESUMO

Background and Methods: The SARS-CoV-2 (severe acute respiratory syndrome coronavirus 2) Omicron (B.1.1.529) variant is the antigenically most distinct variant to date. As the heavily mutated spike protein enables neutralization escape, we studied serum-neutralizing activities of naïve and vaccinated individuals after Omicron BA.1 or BA.2 sub-lineage infections in live virus neutralization tests with Omicron BA.1, Omicron BA.2, wildtype (WT, B1.1), and Delta (B.1.617.2) strains. Serum samples obtained after WT infections and three-dose mRNA vaccinations with and without prior infection were included as controls. Results: Primary BA.1 infections yielded reduced neutralizing antibody levels against WT, Delta, and Omicron BA.2, while samples from BA.2-infected individuals showed almost no cross-neutralization against the other variants. Serum neutralization of Omicron BA.1 and BA.2 variants was detectable after three-dose mRNA vaccinations, but with reduced titers. Vaccination-breakthrough infections with either Omicron BA.1 or BA.2, however, generated equal cross-neutralizing antibody levels against all SARS-CoV-2 variants tested. Conclusions: Our study demonstrates that although Omicron variants are able to enhance cross-neutralizing antibody levels in pre-immune individuals, primary infections with BA.1 or BA.2 induced mostly variant-specific neutralizing antibodies, emphasizing the differently shaped humoral immunity induced by the two Omicron variants. These data thus contribute substantially to the understanding of antibody responses induced by primary Omicron infections or multiple exposures to different SARS-CoV-2 variants and are of particular importance for developing vaccination strategies in the light of future emerging variants.


Assuntos
COVID-19 , SARS-CoV-2 , Anticorpos Neutralizantes , Anticorpos Antivirais , Anticorpos Amplamente Neutralizantes , Humanos , Glicoproteínas de Membrana , Testes de Neutralização , RNA Mensageiro , Glicoproteína da Espícula de Coronavírus/genética , Proteínas do Envelope Viral
4.
Front Immunol ; 13: 888794, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35711424

RESUMO

Background: Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants of concern (VOCs) with different resistance levels to existing immunity have recently emerged. Antibodies that recognize the SARS-CoV-2 spike (S) protein and exhibit neutralizing activities are considered the best correlate of protection and an understanding of humoral immunity is crucial for controlling the pandemic. We thus analyzed such antibodies in individuals recovered from infection in 2020 as well as vaccinees after two doses of an mRNA vaccine. Methods: Neutralizing antibody responses against three SARS-CoV-2 variants (D614G, VOCs Beta and Delta) were determined in serum samples from 54 infected individuals (24 non-hospitalized, 30 hospitalized) and 34 vaccinees shortly after symptom onset or second vaccination, respectively, as well as six months later. In addition, the effect of the S sequence of the infecting strain on neutralization was studied. Results: Non-hospitalized patients had the lowest neutralization titers against all variants, while those of hospitalized patients equaled or exceeded those of vaccinees. Neutralizing activity was lower against the two VOCs and declined significantly in all cohorts after six months. This decrease was more pronounced in hospitalized and vaccinated individuals than in non-hospitalized patients. Of note, the specific neutralizing activity (NT titer/ELISA value ratio) was higher in the infected cohorts than in vaccinees and did not differ between non-hospitalized and hospitalized patients. Patients infected with viral strains carrying mutations in the N-terminal domain of the spike protein were impaired in Beta VOC neutralization. Conclusions: Specific neutralizing activities were higher in infected than in vaccinated individuals, and no difference in the quality of these antibodies was observed between hospitalized and non-hospitalized patients, despite significantly lower titers in the latter group. Additionally, antibody responses of infected individuals showed greater heterogeneity than those of vaccinees, which was associated with mutations in the spike protein of the infecting strain. Overall, our findings yielded novel insights into SARS-CoV-2-specific neutralizing antibodies, evolving differently after virus infection and COVID-19 vaccination, which is an important issue to consider in ongoing vaccine strategy improvements.


Assuntos
COVID-19 , SARS-CoV-2 , Anticorpos Antivirais , COVID-19/prevenção & controle , Vacinas contra COVID-19 , Humanos , Glicoproteínas de Membrana , Testes de Neutralização , SARS-CoV-2/genética , Glicoproteína da Espícula de Coronavírus/genética , Vacinação , Vacinas Sintéticas , Proteínas do Envelope Viral , Vacinas de mRNA
5.
Cancers (Basel) ; 14(8)2022 Apr 13.
Artigo em Inglês | MEDLINE | ID: mdl-35454867

RESUMO

Here we analyzed SARS-CoV-2-specific antibodies and T-cell responses after two coronavirus disease 2019 vaccinations over a six-month period in patients with hematological malignancies and assessed the effect of a third vaccination in a subgroup. Sixty-six patients and 66 healthy controls were included. After two vaccinations seroconversion was seen in 52% and a T-cell-specific response in 59% of patients compared with 100% in controls (p = 0.001). Risk factors for a poor serological response were age (<65a), history of anti-CD20 therapy within the year preceding vaccination, CD19+ B-cells < 110/µL, and CD4+ T-cells > 310/µL. The magnitude of T-cell response was higher in patients <65a and with CD19+ B-cells < 110/µL. Patients and healthy controls demonstrated a significant decrease in SARS-CoV-2 S antibody levels over the period of six months (p < 0.001). A third vaccination demonstrated a strong serological response in patients who had responded to the previous doses (p < 0.001). The third vaccination yielded seroconversion in three out of 19 patients in those without serological response. We conclude that both humoral and cellular responses after SARS-CoV-2 immunization are impaired in patients with hematological malignancies. A third vaccination enhanced B-cell response in patients who previously responded to the second vaccination but may be of limited benefit in patients without prior seroconversion.

6.
Vaccines (Basel) ; 9(8)2021 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-34452033

RESUMO

The aim of this prospective study was to assess lymphocyte proliferative and cytokine response prior to and following tick-borne encephalitis (TBE) immunization among patients after allogeneic hematopoietic stem cell transplantation (HSCT). Seventeen adult patients 11-13 months after HSCT and eight unvaccinated healthy adults received up to three TBE vaccinations. Following in vitro stimulation with TBE-antigen, lymphocyte proliferation and cytokine secretion (IL-2, IL-10, IL-13, TNF-alpha, IFN-gamma, GM-CSF) were analyzed by thymidine incorporation assay and the Luminex system. Ten patients (59%) showed significant baseline TBE-specific lymphocyte proliferation (stimulation index (SI) > 3) prior to vaccination, but none of the unvaccinated controls (p = 0.002). All patients with a TBE-specific antibody response after two vaccinations (at least 2-fold increase of neutralization test titers) exhibited a strong TBE-specific lymphocyte proliferative response at baseline (SI > 10). Patients with sibling donors had a significantly stronger baseline TBE-specific lymphocyte proliferative and IL-13 cytokine response than patients with unrelated donors (p < 0.05). In conclusion, a relevant proportion of patients showed TBE-specific lymphocyte proliferative and cytokine responses prior to vaccination after HSCT, which predicted the humoral response to the vaccine. Patients with vaccinated sibling donors were more likely to elicit a cellular immune response than patients with unrelated donors of unknown vaccination status.

7.
Sci Immunol ; 6(57)2021 03 04.
Artigo em Inglês | MEDLINE | ID: mdl-33664060

RESUMO

CD8+ T cell immunity to SARS-CoV-2 has been implicated in COVID-19 severity and virus control. Here, we identified nonsynonymous mutations in MHC-I-restricted CD8+ T cell epitopes after deep sequencing of 747 SARS-CoV-2 virus isolates. Mutant peptides exhibited diminished or abrogated MHC-I binding in a cell-free in vitro assay. Reduced MHC-I binding of mutant peptides was associated with decreased proliferation, IFN-γ production and cytotoxic activity of CD8+ T cells isolated from HLA-matched COVID-19 patients. Single cell RNA sequencing of ex vivo expanded, tetramer-sorted CD8+ T cells from COVID-19 patients further revealed qualitative differences in the transcriptional response to mutant peptides. Our findings highlight the capacity of SARS-CoV-2 to subvert CD8+ T cell surveillance through point mutations in MHC-I-restricted viral epitopes.


Assuntos
Linfócitos T CD8-Positivos/imunologia , COVID-19 , Epitopos de Linfócito T , Antígenos HLA-A/imunologia , Imunidade Celular , Mutação , SARS-CoV-2 , Linfócitos T CD8-Positivos/patologia , COVID-19/genética , COVID-19/imunologia , COVID-19/patologia , Proliferação de Células , Epitopos de Linfócito T/genética , Epitopos de Linfócito T/imunologia , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Interferon gama/imunologia , Peptídeos/genética , Peptídeos/imunologia , SARS-CoV-2/genética , SARS-CoV-2/imunologia
8.
NPJ Vaccines ; 5(1): 67, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32728481

RESUMO

The aim of this prospective study was to characterize the humoral immune response to TBE vaccination after hematopoietic stem cell transplantation (HSCT). Nineteen adult patients 11-13 months after HSCT and 15 age-matched immunocompetent adults received up to three TBE vaccinations. Antibodies against TBE virus were measured by neutralization test (NT). As primary endpoint, the antibody response (NT titer of ≥10 and at least a twofold increase from baseline 4 weeks after second vaccination) was compared between patients and controls using Fisher exact test. Prior vaccination, 15 (79%) HSCT patients still had detectable neutralizing antibodies. At primary endpoint, the antibody response was significantly lower in patients than in controls (35% versus 93%; p < 0.001). The CD4+ cell count was a predictor for an antibody response in patients (p = 0.019). Interestingly, the majority of HSCT patients still had detectable antibodies prior vaccination. Following vaccination, antibody response in HSCT patients was associated with the CD4+ cell count.

9.
Front Immunol ; 11: 16, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32038660

RESUMO

West Nile (WN) virus infection of humans is frequently asymptomatic, but can also lead to WN fever or neuroinvasive disease. CD4 T cells and B cells are critical in the defense against WN virus, and neutralizing antibodies, which are directed against the viral glycoprotein E, are an accepted correlate of protection. For the efficient production of these antibodies, B cells interact directly with CD4 helper T cells that recognize peptides from E or the two other structural proteins (capsid-C and membrane-prM/M) of the virus. However, the specific protein sites yielding such helper epitopes remain unknown. Here, we explored the CD4 T cell response in humans after WN virus infection using a comprehensive library of overlapping peptides covering all three structural proteins. By measuring T cell responses in 29 individuals with either WN virus disease or asymptomatic infection, we showed that CD4 T cells focus on peptides in specific structural elements of C and at the exposed surface of the pre- and postfusion forms of the E protein. Our data indicate that these immunodominant epitopes are recognized in the context of multiple different HLA molecules. Furthermore, we observed that immunodominant antigen regions are structurally conserved and similarly targeted in other mosquito-borne flaviviruses, including dengue, yellow fever, and Zika viruses. Together, these findings indicate a strong impact of virion protein structure on epitope selection and antigenicity, which is an important issue to consider in future vaccine design.


Assuntos
Infecções Assintomáticas , Linfócitos T CD4-Positivos/imunologia , Epitopos de Linfócito T/imunologia , Febre do Nilo Ocidental/imunologia , Vírus do Nilo Ocidental/imunologia , Adulto , Idoso , Anticorpos Neutralizantes/sangue , Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/sangue , Anticorpos Antivirais/imunologia , Proteínas do Capsídeo/imunologia , Estudos de Coortes , Vírus da Dengue/química , Vírus da Dengue/imunologia , Epitopos de Linfócito T/química , Feminino , Antígenos HLA-D/genética , Humanos , Epitopos Imunodominantes/imunologia , Masculino , Pessoa de Meia-Idade , Biblioteca de Peptídeos , RNA Viral/sangue , Proteínas do Envelope Viral/imunologia , Febre do Nilo Ocidental/virologia , Vírus do Nilo Ocidental/química , Vírus da Febre Amarela/química , Vírus da Febre Amarela/imunologia , Zika virus/química , Zika virus/imunologia
10.
PLoS Negl Trop Dis ; 14(2): e0008034, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-32017766

RESUMO

BACKGROUND: Zika virus has recently spread to South- and Central America, causing congenital birth defects and neurological complications. Many people at risk are flavivirus pre-immune due to prior infections with other flaviviruses (e.g. dengue virus) or flavivirus vaccinations. Since pre-existing cross-reactive immunity can potentially modulate antibody responses to Zika virus infection and may affect the outcome of disease, we analyzed fine-specificity as well as virus-neutralizing and infection-enhancing activities of antibodies induced by a primary Zika virus infection in flavivirus-naïve as well as yellow fever- and/or tick-borne encephalitis-vaccinated individuals. METHODOLOGY: Antibodies in sera from convalescent Zika patients with and without vaccine-induced immunity were assessed by ELISA with respect to Zika virus-specificity and flavivirus cross-reactivity. Functional analyses included virus neutralization and infection-enhancement. The contribution of IgM and cross-reactive antibodies to these properties was determined by depletion experiments. PRINCIPAL FINDINGS: Pre-existing flavivirus immunity had a strong influence on the antibody response in primary Zika virus infections, resulting in higher titers of broadly flavivirus cross-reactive antibodies and slightly lower levels of Zika virus-specific IgM. Antibody-dependent enhancement (ADE) of Zika virus was mediated by sub-neutralizing concentrations of specific IgG but not by cross-reactive antibodies. This effect was potently counteracted by the presence of neutralizing IgM. Broadly cross-reactive antibodies were able to both neutralize and enhance infection of dengue virus but not Zika virus, indicating a different exposure of conserved sequence elements in the two viruses. CONCLUSIONS: Our data point to an important role of flavivirus-specific IgM during the transient early stages of infection, by contributing substantially to neutralization and by counteracting ADE. In addition, our results highlight structural differences between strains of Zika and dengue viruses that are used for analyzing infection-enhancement by cross-reactive antibodies. These findings underscore the possible impact of specific antibody patterns on flavivirus disease and vaccination efficacy.


Assuntos
Anticorpos Antivirais/sangue , Vacinas Virais/imunologia , Infecção por Zika virus/prevenção & controle , Zika virus/imunologia , Afinidade de Anticorpos , Antígenos Virais/imunologia , Ensaio de Imunoadsorção Enzimática , Humanos , Imunoglobulina G/sangue , Testes de Neutralização , Polietilenoglicóis , Proteínas do Envelope Viral/imunologia , Zika virus/genética
11.
Front Immunol ; 9: 1196, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29899743

RESUMO

Zika virus (ZIKV) has recently caused explosive outbreaks in Pacific islands, South- and Central America. Like with other flaviviruses, protective immunity is strongly dependent on potently neutralizing antibodies (Abs) directed against the viral envelope protein E. Such Ab formation is promoted by CD4 T cells through direct interaction with B cells that present epitopes derived from E or other structural proteins of the virus. Here, we examined the extent and epitope dominance of CD4 T cell responses to capsid (C) and envelope proteins in Zika patients. All patients developed ZIKV-specific CD4 T cell responses, with substantial contributions of C and E. In both proteins, immunodominant epitopes clustered at sites that are structurally conserved among flaviviruses but have highly variable sequences, suggesting a strong impact of protein structural features on immunodominant CD4 T cell responses. Our data are particularly relevant for designing flavivirus vaccines and their evaluation in T cell assays and provide insights into the importance of viral protein structure for epitope selection and antigenicity.


Assuntos
Linfócitos B/imunologia , Linfócitos T CD4-Positivos/imunologia , Proteínas do Capsídeo/genética , Epitopos de Linfócito T/genética , Epitopos Imunodominantes/genética , Proteínas do Envelope Viral/genética , Vacinas Virais/imunologia , Infecção por Zika virus/imunologia , Zika virus/fisiologia , Adulto , Idoso , Anticorpos Neutralizantes/metabolismo , Anticorpos Antivirais/metabolismo , Proteínas do Capsídeo/imunologia , Células Cultivadas , Sequência Conservada/genética , Epitopos de Linfócito T/imunologia , Feminino , Humanos , Epitopos Imunodominantes/imunologia , Masculino , Pessoa de Meia-Idade , Relação Estrutura-Atividade , Proteínas do Envelope Viral/imunologia , Adulto Jovem
12.
Sci Rep ; 7(1): 8907, 2017 08 21.
Artigo em Inglês | MEDLINE | ID: mdl-28827760

RESUMO

The live attenuated yellow fever (YF) vaccine is a highly effective human vaccine and induces long-term protective neutralizing antibodies directed against the viral envelope protein E. The generation of such antibodies requires the help of CD4 T cells which recognize peptides derived from proteins in virus particles internalized and processed by E-specific B cells. The CD4 T helper cell response is restricted to few immunodominant epitopes, but the mechanisms of their selection are largely unknown. Here, we report that CD4 T cell responses elicited by the YF-17D vaccine are focused to hotspots of two helices of the viral capsid protein and to exposed strands and loops of E. We found that the locations of immunodominant epitopes within three-dimensional protein structures exhibit a high degree of overlap between YF virus and the structurally homologous flavivirus tick-borne encephalitis virus, although amino acid sequence identity of the epitope regions is only 15-45%. The restriction of epitopes to exposed E protein surfaces and their strikingly similar positioning within proteins of distantly related flaviviruses are consistent with a strong influence of protein structure that shapes CD4 T cell responses and provide leads for a rational design of immunogens for vaccination.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Epitopos Imunodominantes/química , Epitopos Imunodominantes/imunologia , Conformação Proteica , Vacina contra Febre Amarela/imunologia , Vírus da Febre Amarela/imunologia , Sequência de Aminoácidos , Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais , Sítios de Ligação , Linfócitos T CD4-Positivos/metabolismo , Sequência Conservada , Mapeamento de Epitopos , Humanos , Modelos Moleculares , Ligação Proteica , Relação Estrutura-Atividade , Especificidade do Receptor de Antígeno de Linfócitos T/imunologia , Vacinação , Vacinas Atenuadas/imunologia , Proteínas do Envelope Viral/química , Proteínas do Envelope Viral/imunologia , Proteínas Estruturais Virais/química , Proteínas Estruturais Virais/imunologia , Febre Amarela/imunologia , Febre Amarela/prevenção & controle
13.
J Clin Virol ; 70: 109-119, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26305832

RESUMO

Opportunistic viral infections are still a major complication following solid organ transplantation. Immune monitoring may allow the identification of patients at risk of infection and, eventually, the modulation of immunosuppressive strategies. Immune monitoring can be performed using virus-specific and non virus-specific assays. This article describes and summarizes the pros and cons of the different technical approaches. Among the assays based on non virus-specific antigens, the enumeration of T-cell subsets, the quantification of cytokines and chemokines and the quantification of intracellular adenosine triphosphate following mitogen stimulation are described and their clinical applications to determine the risk for viral infection are discussed. In addition, current specific methods available for monitoring viral-specific T-cell responses are summarized, such as peptide-MHC multimer staining, intracellular cytokine staining, enzyme-linked immunospot and virus-specific IFN-γ ELISA assays, and their clinical applications to determine the individual risk for opportunistic viral infections with human cytomegalovirus, Epstein-Barr virus and polyoma BK virus are discussed. The standardization of the procedure, the choice of the antigen(s) and the criteria to define cut-off values for positive responses are needed for some of these approaches before their implementation in the clinic. Nevertheless, immune monitoring combined with virological monitoring in transplant recipients is increasingly regarded as a helpful tool to identify patients at risk of infection as well as to assess treatment efficacy.


Assuntos
Infecções Oportunistas/etiologia , Transplante de Órgãos , Especificidade do Receptor de Antígeno de Linfócitos T/imunologia , Linfócitos T/imunologia , Transplantados , Viroses/etiologia , Antígenos/imunologia , Citocinas/biossíntese , Humanos , Imunossupressores/efeitos adversos , Contagem de Linfócitos , Infecções Oportunistas/metabolismo , Infecções Oportunistas/virologia , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo , Linfócitos T/metabolismo , Viroses/metabolismo , Viroses/virologia
14.
J Virol ; 88(23): 13845-57, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25253341

RESUMO

UNLABELLED: Tick-borne encephalitis (TBE) virus is an important human-pathogenic flavivirus endemic in large parts of Europe and Central and Eastern Asia. Neutralizing antibodies specific for the viral envelope protein E are believed to mediate long-lasting protection after natural infection and vaccination. To study the specificity and individual variation of human antibody responses, we developed immunoassays with recombinant antigens representing viral surface protein domains and domain combinations. These allowed us to dissect and quantify antibody populations of different fine specificities in sera of TBE patients and vaccinees. Postinfection and postvaccination sera both displayed strong individual variation of antibody titers as well as the relative proportions of antibodies to different domains of E, indicating that the immunodominance patterns observed were strongly influenced by individual-specific factors. The contributions of these antibody populations to virus neutralization were quantified by serum depletion analyses and revealed a significantly biased pattern. Antibodies to domain III, in contrast to what was found in mouse immunization studies with TBE and other flaviviruses, did not play any role in the human neutralizing antibody response, which was dominated by antibodies to domains I and II. Importantly, most of the neutralizing activity could be depleted from sera by a dimeric soluble form of the E protein, which is the building block of the icosahedral herringbone-like shell of flaviviruses, suggesting that antibodies to more complex quaternary epitopes involving residues from adjacent dimers play only a minor role in the total response to natural infection and vaccination in humans. IMPORTANCE: Tick-borne encephalitis (TBE) virus is a close relative of yellow fever, dengue, Japanese encephalitis, and West Nile viruses and distributed in large parts of Europe and Central and Eastern Asia. Antibodies to the viral envelope protein E prevent viral attachment and entry into cells and thus mediate virus neutralization and protection from disease. However, the fine specificity and individual variation of neutralizing antibody responses are currently not known. We have therefore developed new in vitro assays for dissecting the antibody populations present in blood serum and determining their contribution to virus neutralization. In our analysis of human postinfection and postvaccination sera, we found an extensive variation of the antibody populations present in sera, indicating substantial influences of individual-specific factors that control the specificity of the antibody response. Our study provides new insights into the immune response to an important human pathogen that is of relevance for the design of novel vaccines.


Assuntos
Anticorpos Neutralizantes/sangue , Anticorpos Antivirais/sangue , Vírus da Encefalite Transmitidos por Carrapatos/imunologia , Encefalite Transmitida por Carrapatos/imunologia , Vacinas Virais/imunologia , Adulto , Idoso , Estudos de Coortes , Epitopos/imunologia , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Proteínas do Envelope Viral/imunologia , Vacinas Virais/administração & dosagem , Adulto Jovem
15.
Exp Gerontol ; 47(8): 654-7, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22564865

RESUMO

Human cytomegalovirus (HCMV) may play an important role in immune system aging, due to its ability to modulate the host immune system. A significant age-related increase has been demonstrated for HCMV-specific serum antibody levels but so far, no information exists whether and to which extent the magnitude of the HCMV-specific memory B cell response develops with increasing age. We examined the size of the HCMV-specific memory B cell response and assessed whether there are quantitative differences in HCMV-specific memory B cell numbers and serum antibody titers against HCMV in young and older persons. The quantities of HCMV-specific memory B cells were determined in 20 young (20-31 years) and 21 older (60-80 years) healthy volunteers by limiting dilution analysis. HCMV-specific antibody levels were analyzed by ELISA. Our study demonstrates that higher HCMV-specific plasma antibody levels correlated well with the numbers of circulating HCMV-specific memory B cells and were not due to a generally higher antibody production in these individuals. The magnitude of HCMV-specific memory B cell responses was significantly higher in the group of older as compared to young subjects, but showed a high degree of individual variation in elderly persons. Together, the results of the present study indicate that the circulating memory B cell pool against HCMV increases during aging in humans. The expansion of HCMV-specific memory B cells and antibody titers possibly contribute to the sustained control of HCMV infection during old age.


Assuntos
Envelhecimento/imunologia , Subpopulações de Linfócitos B/imunologia , Citomegalovirus/imunologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Anticorpos Antivirais/sangue , Infecções por Citomegalovirus/imunologia , Feminino , Humanos , Imunoglobulina G/sangue , Memória Imunológica/fisiologia , Contagem de Linfócitos , Masculino , Pessoa de Meia-Idade , Adulto Jovem
16.
J Infect Dis ; 202(12): 1770-9, 2010 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-21067369

RESUMO

INTRODUCTION: Acute hepatitis C virus (HCV) infection is rarely studied, but virus sequence evolution and host-virus dynamics during this early stage may influence the outcome of infection. Hypervariable region 1 (HVR1) is genetically diverse and under selective pressure from the host immune response. We analyzed HVR1 evolution by frequent sampling of an acutely infected HCV cohort. METHODS: Three or more pretreatment samples were obtained from each of 10 acutely infected subjects. Polymerase chain reaction amplification was performed with multiple primer combinations to identify the full range of sequences present. Positive samples were cloned and sequenced. Phylogenetic analyses were used to assess viral diversity. RESULTS: Eight of the 10 subjects were coinfected with at least 2 HCV subtypes. Multiple subtypes were detected in individual samples, and their relative proportions changed through acute infection. The subjects with the most complex subtype structure also had a dynamic viral load; however, changes in viral load were not directly linked to changes in subtype. CONCLUSIONS: This well-sampled cohort with acute HCV infection was characterized by dynamic coinfection with multiple viral subtypes, representing a highly complex virologic landscape extremely early in infection.


Assuntos
Hepacivirus/imunologia , Hepacivirus/patogenicidade , Hepatite C/virologia , Interações Hospedeiro-Patógeno , Polimorfismo Genético , RNA Viral/genética , Adolescente , Adulto , Áustria , Clonagem Molecular , Feminino , Genótipo , Hepacivirus/genética , Hepacivirus/isolamento & purificação , Hepatite C/diagnóstico , Humanos , Estudos Longitudinais , Masculino , Pessoa de Meia-Idade , Dados de Sequência Molecular , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Análise de Sequência de DNA , Proteínas do Envelope Viral/genética
17.
Pediatr Infect Dis J ; 24(7): 605-10, 2005 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15999001

RESUMO

BACKGROUND: Dual respiratory viral infections are frequently associated with lower respiratory tract illness in infants. This study aimed to determine the impact of a dual respiratory viral infection on specific aspects of the infant's immune response and the clinical course of illness. METHODS: A prospective study was performed with 772 infants hospitalized from October 2000 through July 2004. Sensitive polymerase chain reaction methodology revealed the presence of a single respiratory virus in 443 (57%) of 772 cases, whereas dual infections were identified in 153 (20%) of cases. From 250 infants with confirmed respiratory viral infection, fresh heparinized blood was analyzed for interferon-gamma (IFN-gamma) responses by flow cytometry. Of these, 191 patients had a single infection with respiratory syncytial virus (RSV), rhinoviruses, adenoviruses or influenza viruses; and 59 patients had a dual infection with RSV and rhinoviruses, RSV and adenoviruses, influenza viruses and rhinoviruses or adenoviruses and rhinoviruses. The clinical features and peripheral lymphocyte IFN-gamma responses were compared among infants with single or dual infections. RESULTS: It was found that dual infections with non-RSV respiratory viruses induced peripheral blood mononuclear cell IFN-gamma responses that mimic those of single infections, whereas coinfection with RSV was associated with reduced IFN-gamma responses and a more severe clinical course of lower respiratory tract disease. CONCLUSIONS: The results indicate that the clinical characteristics and the IFN-gamma response differ significantly in single and dual respiratory viral infection, depending on the nature of the simultaneously detected viruses. In dual infections, RSV involvement was associated with a decreased IFN-gamma response in peripheral blood mononuclear cell and an increase in severity of illness.


Assuntos
Hospitalização , Doenças do Prematuro , Infecções Respiratórias , Viroses/complicações , Viroses/virologia , Feminino , Humanos , Recém-Nascido , Recém-Nascido Prematuro , Doenças do Prematuro/imunologia , Doenças do Prematuro/fisiopatologia , Doenças do Prematuro/virologia , Interferon gama/biossíntese , Leucócitos Mononucleares/imunologia , Masculino , Estudos Prospectivos , Infecções Respiratórias/complicações , Infecções Respiratórias/imunologia , Infecções Respiratórias/fisiopatologia , Infecções Respiratórias/virologia , Viroses/imunologia , Viroses/fisiopatologia , Vírus/genética , Vírus/isolamento & purificação
18.
J Infect Dis ; 187(2): 311-4, 2003 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-12552457

RESUMO

Serum samples from 68 immunocompetent infants (mean age, 12.6 months) with an acute adenovirus infection of the respiratory tract (39 experiencing their first adenovirus infection) were tested for the presence of adenovirus DNA, to investigate whether viral dissemination via the blood is usually present in the immunocompetent patient. Using a nested polymerase chain reaction assay, adenovirus DNA could be detected in acute-phase serum samples from 28 (41%) children. Adenovirus DNA was never found in follow-up serum samples, indicating a short period ( approximately 1 week) of viral dissemination. In children experiencing their first adenovirus infection, viral DNA could be detected in 72% of the acute-phase serum samples collected within the first week after onset of symptoms. Adenovirus DNA could also be detected in 25% of the acute-phase serum samples from patients with reinfection.


Assuntos
Adenoviridae/genética , Adenoviridae/isolamento & purificação , Infecções por Adenovirus Humanos/virologia , DNA Viral/sangue , Infecções Respiratórias/virologia , Doença Aguda , Infecções por Adenovirus Humanos/sangue , Feminino , Hospitalização , Humanos , Lactente , Masculino , Infecções Respiratórias/sangue , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA