Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
1.
Front Immunol ; 15: 1387454, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38799468

RESUMO

Introduction: Mycobacteria are known to exert a range of heterologous effects on the immune system. The mycobacteria-based Freund's Complete Adjuvant is a potent non-specific stimulator of the immune response used in immunization protocols promoting antibody production, and Mycobacterium bovis Bacille Calmette Guérin (BCG) vaccination has been linked with decreased morbidity and mortality beyond the specific protection it provides against tuberculosis (TB) in some populations and age groups. The role of heterologous antibodies in this phenomenon, if any, remains unclear and under-studied. Methods: We set out to evaluate antibody responses to a range of unrelated pathogens following infection with Mycobacterium tuberculosis (M.tb) and vaccination with BCG or a candidate TB vaccine, MTBVAC, in non-human primates. Results: We demonstrate a significant increase in the titer of antibodies against SARS-CoV-2, cytomegalovirus, Epstein-Barr virus, tetanus toxoid, and respiratory syncytial virus antigens following low-dose aerosol infection with M.tb. The magnitude of some of these responses correlated with TB disease severity. However, vaccination with BCG administered by the intradermal, intravenous or aerosol routes, or intradermal delivery of MTBVAC, did not increase antibody responses against unrelated pathogens. Discussion: Our findings suggest that it is unlikely that heterologous antibodies contribute to the non-specific effects of these vaccines. The apparent dysregulation of B cell responses associated with TB disease warrants further investigation, with potential implications for risk of B cell cancers and novel therapeutic strategies.


Assuntos
Vacina BCG , Mycobacterium tuberculosis , Tuberculose , Vacinação , Animais , Vacina BCG/imunologia , Vacina BCG/administração & dosagem , Tuberculose/imunologia , Tuberculose/prevenção & controle , Mycobacterium tuberculosis/imunologia , Anticorpos Antibacterianos/imunologia , Anticorpos Antibacterianos/sangue , Anticorpos Antivirais/imunologia , Anticorpos Antivirais/sangue , Vacinas contra a Tuberculose/imunologia , Vacinas contra a Tuberculose/administração & dosagem , Feminino , Macaca mulatta , SARS-CoV-2/imunologia , COVID-19/imunologia , COVID-19/prevenção & controle , Imunidade Heteróloga , Masculino
2.
J Immunol ; 206(3): 455-462, 2021 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-33468558

RESUMO

Memory T cells (Tmem) rapidly mount Ag-specific responses during pathogen reencounter. However, Tmem also respond to inflammatory cues in the absence of an activating TCR signal, a phenomenon termed bystander activation. Although bystander activation was first described over 20 years ago, the physiological relevance and the consequences of T cell bystander activation have only become more evident in recent years. In this review, we discuss the scenarios that trigger CD8 Tmem bystander activation including acute and chronic infections that are either systemic or localized, as well as evidence for bystander CD8 Tmem within tumors and following vaccination. We summarize the possible consequences of bystander activation for the T cell itself, the subsequent immune response, and the host. We highlight when T cell bystander activation appears to benefit or harm the host and briefly discuss our current knowledge gaps regarding regulatory signals that can control bystander activation.


Assuntos
Efeito Espectador/imunologia , Linfócitos T CD8-Positivos/imunologia , Infecções/imunologia , Neoplasias/imunologia , Vacinas/imunologia , Animais , Humanos , Imunidade Heteróloga , Memória Imunológica , Ativação Linfocitária
3.
Nat Med ; 26(6): 932-940, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32393800

RESUMO

Recent efforts toward an HIV vaccine focus on inducing broadly neutralizing antibodies, but eliciting both neutralizing antibodies (nAbs) and cellular responses may be superior. Here, we immunized macaques with an HIV envelope trimer, either alone to induce nAbs, or together with a heterologous viral vector regimen to elicit nAbs and cellular immunity, including CD8+ tissue-resident memory T cells. After ten vaginal challenges with autologous virus, protection was observed in both vaccine groups at 53.3% and 66.7%, respectively. A nAb titer >300 was generally associated with protection but in the heterologous viral vector + nAb group, titers <300 were sufficient. In this group, protection was durable as the animals resisted six more challenges 5 months later. Antigen stimulation of T cells in ex vivo vaginal tissue cultures triggered antiviral responses in myeloid and CD4+ T cells. We propose that cellular immune responses reduce the threshold of nAbs required to confer superior and durable protection.


Assuntos
Anticorpos Neutralizantes/efeitos dos fármacos , Anticorpos Antivirais/efeitos dos fármacos , Linfócitos T CD8-Positivos/efeitos dos fármacos , Produtos do Gene gag/genética , Imunidade Celular/efeitos dos fármacos , Vacinas contra a SAIDS/farmacologia , Síndrome de Imunodeficiência Adquirida dos Símios/prevenção & controle , Vírus da Imunodeficiência Símia/imunologia , Animais , Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/imunologia , Linfócitos T CD4-Positivos/efeitos dos fármacos , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Feminino , Produtos do Gene gag/imunologia , Vetores Genéticos , Imunidade Celular/imunologia , Imunidade Heteróloga , Imunogenicidade da Vacina , Memória Imunológica/imunologia , Macaca mulatta , Mucosa , Vagina
4.
Front Immunol ; 10: 2564, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31787970

RESUMO

Enterovirus and Coxsackievirus are the major viruses that cause hand, foot, and mouth disease (HFMD) outbreaks worldwide. Several studies have shown the potential of viral envelope protein 1 (VP1) on providing protective effects from viral strains of different genotypes. However, whether VP1 has the cross-protection in Enteroviruses or Coxsackievirus has not been studied in-depth. In this study, the vp1 gene of Enterovirus 71 (EV71) and Coxsackievirus B3 (CB3) was inserted into the vector pET22b (+) to form the respective expression plasmids pEVP1 or pCVP1, and then transformed into Escherichia coli strain BL21 (DE3). The recombinant EVP1 or CVP1 protein was overexpressed successfully and effectively purified to homogeneity. Then, we identified that EVP1 and CVP1 protein could generate effectively specific humoral immunity and cellular immunity in mice, what's more, we determined the cross-protection of VP1 between EV71 and CB3 in a murine model. The results showed that immunization with EVP1 could effectively induce specific IgG and secretory IgA against CVP1 and the sera from EVP1-immunized mice could neutralize CB3 with mean titers 1:440. In contrast, no measurable neutralizing antibodies to EV71 were detected in CVP1-immunized mice. Then, newborn BALB/C mice, whose mother was immunized with EVP1 or CVP1, were administered with different lethal doses of EV71 or CB3. The EVP1 immunized group showed a 90% protective efficacy for a CB3 dosage of 120 LD50, but the CVP1 immunized group showed no significantly different protective efficacy against 15 LD50 of EV71 compared with the BSA immunized group. Hence, EVP1 is a promising subunit vaccine candidate against Enterovirus 71 and Coxsackievirus B3 caused HFMD.


Assuntos
Infecções por Coxsackievirus/prevenção & controle , Enterovirus Humano A/imunologia , Enterovirus Humano B/imunologia , Infecções por Enterovirus/prevenção & controle , Proteínas Estruturais Virais/imunologia , Animais , Anticorpos Neutralizantes/sangue , Anticorpos Antivirais/sangue , Infecções por Coxsackievirus/imunologia , Infecções por Coxsackievirus/virologia , Proteção Cruzada/imunologia , Modelos Animais de Doenças , Enterovirus Humano A/genética , Enterovirus Humano A/patogenicidade , Infecções por Enterovirus/imunologia , Infecções por Enterovirus/virologia , Feminino , Humanos , Imunidade Heteróloga , Interferon gama/sangue , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Vacinas de Subunidades Antigênicas/genética , Vacinas de Subunidades Antigênicas/imunologia , Proteínas Estruturais Virais/genética , Vacinas Virais/genética , Vacinas Virais/imunologia
5.
Viruses ; 11(9)2019 08 25.
Artigo em Inglês | MEDLINE | ID: mdl-31450681

RESUMO

Newcastle disease (ND) is responsible for significant economic losses in the poultry industry. The disease is caused by virulent strains of Avian avulavirus 1 (AAvV-1), a species within the family Paramyxoviridae. We developed a recombinant construct based on the herpesvirus of turkeys (HVT) as a vector expressing two genes: F and HN (HVT-NDV-F-HN) derived from the AAvV-1 genotype VI ("pigeon variant" of AAvV-1). This recombinant viral vaccine candidate was used to subcutaneously immunize one group of specific pathogen-free (SPF) chickens and two groups of broiler chickens (20 one-day-old birds/group). Humoral immune response was evaluated by hemagglutination-inhibition test and enzyme-linked immunosorbent assay (ELISA). The efficacy of the immunization was assessed in two separate challenge studies performed at 6 weeks of age with the use of virulent AAvV-1 strains representing heterologous genotypes IV and VII. The developed vaccine candidate elicited complete protection in SPF chickens since none of the birds became sick or died during the 2-week observation period. In the broiler groups, 90% and 100% clinical protection were achieved after challenges with AAvV-1 of IV and VII genotypes, respectively. We found no obvious relationship between antibody levels and protection assessed in broilers in the challenge study. The developed recombinant HVT-NDV-F-HN construct containing genes from a genotype VI AAvV-1 offers promising results as a potential vaccine candidate against ND in chickens.


Assuntos
Proteína HN/imunologia , Imunização/veterinária , Vírus da Doença de Newcastle , Vacinas Sintéticas/imunologia , Proteínas Virais de Fusão/imunologia , Animais , Antígenos Virais/biossíntese , Antígenos Virais/genética , Galinhas/virologia , Proteção Cruzada , Genes Virais , Proteína HN/biossíntese , Proteína HN/genética , Testes de Inibição da Hemaglutinação , Herpesvirus Meleagrídeo 1/genética , Herpesvirus Meleagrídeo 1/imunologia , Herpesvirus Meleagrídeo 1/metabolismo , Imunidade Heteróloga , Doença de Newcastle/imunologia , Doença de Newcastle/prevenção & controle , Vírus da Doença de Newcastle/genética , Vírus da Doença de Newcastle/imunologia , Doenças das Aves Domésticas/virologia , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/genética , Proteínas Recombinantes/imunologia , Vacinas Sintéticas/virologia , Proteínas Virais de Fusão/biossíntese , Proteínas Virais de Fusão/genética , Vacinas Virais/biossíntese , Vacinas Virais/imunologia
6.
Curr Opin Immunol ; 59: 101-108, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31265968

RESUMO

The human lung contains a heterogeneous population of immune cells which mediate protective responses, maintain tissue homeostasis, but can also promote immunopathology in disease. The majority of T cells in the human lung are tissue resident memory T cells (TRM) which have been shown in mouse models to provide vital roles in the protection against multiple respiratory pathogens, and contribute to heterosubtypic protection in the context of vaccination. In this review, we will discuss recent studies in humans identifying lung TRM, their role in maintaining tissue homeostasis, and emerging evidence implicating TRM in anti-tumor immunity and immune surveillance as well as their potential for immunopathology in chronic airway inflammation.


Assuntos
Inflamação/imunologia , Pulmão/imunologia , Neoplasias/imunologia , Infecções Respiratórias/imunologia , Linfócitos T/imunologia , Vacinas/imunologia , Animais , Modelos Animais de Doenças , Homeostase , Humanos , Imunidade Heteróloga , Memória Imunológica , Vigilância Imunológica , Camundongos , Vacinação
7.
Cells ; 8(5)2019 05 26.
Artigo em Inglês | MEDLINE | ID: mdl-31130710

RESUMO

Host immune responses play an important role in the outcome of infection with hepatitis C virus (HCV). They can lead to viral clearance and a positive outcome, or progression and severity of chronic disease. Extensive research in the past >25 years into understanding the immune responses against HCV have still resulted in many unanswered questions implicating a role for unknown factors and events. In our earlier studies, we made a surprising discovery that peptides derived from structural and non-structural proteins of HCV have substantial amino acid sequence homologies with various proteins of adenoviruses and that immunizing mice with a non-replicating, non-recombinant adenovirus vector leads to induction of a robust cross-reactive cellular and humoral response against various HCV antigens. In this work, we further demonstrate antibody cross-reactivity between Ad and HCV in vivo. We also extend this observation to show that recombinant adenoviruses containing antigens from unrelated pathogens also possess the ability to induce cross-reactive immune responses against HCV antigens along with the induction of transgene antigen-specific immunity. This cross-reactive immunity can (a) accommodate the making of dual-pathogen vaccines, (b) play an important role in the natural course of HCV infection and (c) provide a plausible answer to many unexplained questions regarding immunity to HCV.


Assuntos
Infecções por Adenoviridae/virologia , Adenoviridae/imunologia , Vacinas contra Adenovirus/imunologia , Reações Cruzadas/imunologia , Vetores Genéticos/imunologia , Hepacivirus/imunologia , Antígenos da Hepatite C/imunologia , Hepatite C/virologia , Imunidade Heteróloga/imunologia , Animais , Antígenos de Bactérias/imunologia , Células Cultivadas , Feminino , Antígenos HIV/imunologia , Imunidade Celular/imunologia , Imunidade Humoral/imunologia , Imunização/métodos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Baço/patologia , Vacinação/métodos
8.
Front Immunol ; 10: 730, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31024558

RESUMO

An ideal malaria vaccine platform should potently induce protective immune responses and block parasite transmission from mosquito to human, and it should maintain these effects for an extended period. Here, we have focused on vaccine development based on adeno-associated virus serotype 1 (AAV1), a viral vector widely studied in the field of clinical gene therapy that is able to induce long-term transgene expression without causing toxicity in vivo. Our results show the potential utility of AAV1 vectors as an extremely potent booster vaccine to induce durable immunity when combined with an adenovirus-priming vaccine in a rodent malaria model. We generated a series of recombinant AAV1s and human adenovirus type 5 (AdHu5) expressing either Plasmodium falciparum circumsporozoite protein (PfCSP) or P25 (Pfs25) protein. Heterologous two-dose immunization with an AdHu5-prime and AAV1-boost (AdHu5-AAV1) elicited robust and durable PfCSP- or Pfs25-specific functional antibodies over 280 days. Regarding protective efficacy, AdHu5-AAV1 PfCSP achieved high sterile protection (up to 80% protection rate) against challenge with transgenic Plasmodium berghei sporozoites expressing PfCSP. When examining transmission-blocking (TB) efficacy, we found that immunization with AdHu5-AAV1 Pfs25 maintained TB activity in vivo against transgenic P. berghei expressing Pfs25 for 287 days (99% reduction in oocyst intensity, 85% reduction in oocyst prevalence). Our data indicate that AAV1-based malaria vaccines can confer potent and durable protection as well as TB efficacy when administered following an AdHu5 priming vaccine, supporting the further evaluation of this regimen in clinical trials as a next-generation malaria vaccine platform.


Assuntos
Dependovirus/genética , Vetores Genéticos/genética , Vacinas Antimaláricas/imunologia , Malária/imunologia , Plasmodium falciparum/fisiologia , Animais , Anticorpos Antiprotozoários/sangue , Células Cultivadas , Resistência à Doença , Terapia Genética , Humanos , Imunidade Heteróloga , Imunização Secundária , Malária/transmissão , Camundongos , Camundongos Endogâmicos BALB C , Proteínas de Protozoários/genética , Vacinação
9.
Cell ; 176(6): 1340-1355.e15, 2019 03 07.
Artigo em Inglês | MEDLINE | ID: mdl-30799037

RESUMO

Th17 cells provide protection at barrier tissues but may also contribute to immune pathology. The relevance and induction mechanisms of pathologic Th17 responses in humans are poorly understood. Here, we identify the mucocutaneous pathobiont Candida albicans as the major direct inducer of human anti-fungal Th17 cells. Th17 cells directed against other fungi are induced by cross-reactivity to C. albicans. Intestinal inflammation expands total C. albicans and cross-reactive Th17 cells. Strikingly, Th17 cells cross-reactive to the airborne fungus Aspergillus fumigatus are selectively activated and expanded in patients with airway inflammation, especially during acute allergic bronchopulmonary aspergillosis. This indicates a direct link between protective intestinal Th17 responses against C. albicans and lung inflammation caused by airborne fungi. We identify heterologous immunity to a single, ubiquitous member of the microbiota as a central mechanism for systemic induction of human anti-fungal Th17 responses and as a potential risk factor for pulmonary inflammatory diseases.


Assuntos
Candida albicans/imunologia , Células Th17/imunologia , Células Th17/metabolismo , Aspergillus fumigatus/imunologia , Aspergillus fumigatus/patogenicidade , Candida albicans/patogenicidade , Reações Cruzadas/imunologia , Fibrose Cística/imunologia , Fibrose Cística/microbiologia , Humanos , Imunidade , Imunidade Heteróloga/imunologia , Células Th17/fisiologia
10.
Eur J Clin Microbiol Infect Dis ; 38(3): 449-456, 2019 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-30719592

RESUMO

Vaccines such as Vaccinia or BCG have non-specific effects conferring protection against other diseases than their target infection, which are likely partly mediated through induction of innate immune memory (trained immunity). MVA85A, a recombinant strain of modified Vaccinia Ankara (MVA), has been suggested as an alternative vaccine against tuberculosis, but its capacity to induce positive or negative non-specific immune effects has not been studied. This study assesses whether Vaccinia and MVA are able to induce trained innate immunity in monocytes. Human primary monocytes were primed in an in vitro model with Vaccinia or MVA for 1 day, after which the stimulus was washed off and the cells were rechallenged with unrelated microbial ligands after 1 week. Heterologous cytokine responses were assessed and the capacity of MVA to induce epigenetic changes at the level of cytokine genes was investigated using chromatin immunoprecipitation and pharmacological inhibitors. Monocytes trained with Vaccinia showed significantly increased IL-6 and TNF-α production to stimulation with non-related stimuli, compared to non-trained monocytes. In contrast, monocytes primed with MVA showed significant decreased heterologous IL-6 and TNF-α responses, an effect which was abrogated by the addition of a histone methyltransferase inhibitor. No effects on H3K4me3 were observed after priming with MVA. It can be thus concluded that Vaccinia induces trained immunity in vitro, whereas MVA induces innate immune tolerance. This suggests the induction of trained immunity as an immunological mechanism involved in the non-specific effects of Vaccinia vaccination and points to a possible explanation for the lack of effect of MVA85A against tuberculosis.


Assuntos
Imunidade Heteróloga , Vacinas contra a Tuberculose/imunologia , Vaccinia virus/imunologia , Vacínia/imunologia , Vacinas Virais/imunologia , Histona Metiltransferases/antagonistas & inibidores , Humanos , Imunidade Heteróloga/efeitos dos fármacos , Imunidade Inata , Interleucina-6/metabolismo , Leucócitos Mononucleares/imunologia , Pemetrexede/farmacologia , Fator de Necrose Tumoral alfa/metabolismo , Vacinas de DNA , Vaccinia virus/genética
12.
Clin Exp Immunol ; 195(1): 96-108, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30194852

RESUMO

Submicroscopic Plasmodium infections in pregnancy are common in endemic areas, and it is important to understand the impact of these low-level infections. Asymptomatic, chronic infections are advantageous for parasite persistence, particularly in areas where the optimal eco-epidemiological conditions for parasite transmission fluctuate. In chronic infections, the persistence of the antigenic stimulus changes the expression of immune mediators and promotes constant immune regulation, including increases in regulatory T cell populations. These alterations of the immune system could compromise the response to routine vaccination. This study aimed to evaluate the effect of submicroscopic plasmodial infection with P. falciparum and P. vivax during pregnancy on the immune response to the tetanus toxoid vaccine in Colombian women. Expression of different cytokines and mediators of immune regulation and levels of anti-tetanus toxoid (TT) immunoglobulin (Ig)G were quantified in pregnant women with and without submicroscopic plasmodial infection. The anti-TT IgG levels were significantly lower in the infected group compared with the uninfected group. The expression of interferon (IFN)-γ, tumour necrosis factor (TNF) and forkhead box protein 3 (FoxP3) was significantly higher in the infected group, while the expression of cytotoxic T lymphocyte antigen 4 (CTLA-4) and transforming growth factor (TGF)-ß was lower in the group of infected. In conclusion, submicroscopic Plasmodium infection altered the development of the immune response to the TT vaccine in Colombian pregnant women. The impact of Plasmodium infections on the immune regulatory pathways warrants further exploration.


Assuntos
Anticorpos Antibacterianos/sangue , Malária/imunologia , Plasmodium falciparum/fisiologia , Plasmodium vivax/fisiologia , Complicações Infecciosas na Gravidez/imunologia , Linfócitos T Reguladores/imunologia , Toxoide Tetânico/imunologia , Adolescente , Adulto , Doença Crônica , Colômbia , Citocinas/metabolismo , Feminino , Humanos , Imunidade Heteróloga , Gravidez , Vacinação , Adulto Jovem
13.
Semin Immunol ; 39: 35-43, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-30007489

RESUMO

Besides protection against specific microorganisms, vaccines can induce heterologous or non-specific effects (NSE). Epidemiological data suggest that vaccination with live-attenuated vaccines such as Bacillus Calmette-Guérin (BCG), measles vaccine, and oral polio vaccine results in increased overall childhood survival, and several of these observations have been confirmed in randomized trials. Immunological mechanisms mediating NSE include heterologous lymphocyte effects and induction of innate immune memory (trained immunity). Trained immunity induces long-term functional upregulation of innate immune cells through epigenetic and metabolic reprogramming. An overview of the epidemiological evidence of non-specific effects of vaccines and the latest insights regarding the biological mechanisms behind this phenomenon is presented, and future research priorities and potential implications are discussed.


Assuntos
Reprogramação Celular/imunologia , Imunidade Heteróloga , Imunidade Inata/efeitos dos fármacos , Memória Imunológica/efeitos dos fármacos , Vacinação , Vacina BCG/administração & dosagem , Reprogramação Celular/genética , Criança , Epigênese Genética , Humanos , Linfócitos/citologia , Linfócitos/efeitos dos fármacos , Linfócitos/imunologia , Macrófagos/citologia , Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Vacina contra Sarampo/administração & dosagem , Vacinas contra Poliovirus/administração & dosagem , Ensaios Clínicos Controlados Aleatórios como Assunto , Fatores Sexuais
14.
Artigo em Inglês | MEDLINE | ID: mdl-29888215

RESUMO

Most vertebrates are infected with one or more herpesviruses and remain so for the rest of their lives. The relationship of immunocompetent healthy host with herpesviruses may sometime be considered as harmonious. However, clinically severe diseases can occur when host immunity is compromised due to aging, during some stress response, co-infections or during neoplastic disease conditions. Discord can also occur during iatrogenic immunosuppression used for controlling graft rejection, in some primary genetic immunodeficiencies as well as when the virus infects a non-native host. In this review, we discuss such issues and their influence on host-herpesvirus interaction.


Assuntos
Coinfecção/complicações , Coinfecção/imunologia , Infecções por Herpesviridae/imunologia , Herpesviridae/imunologia , Herpesviridae/patogenicidade , Interações Hospedeiro-Parasita/imunologia , Animais , Doenças Autoimunes/complicações , Doenças Autoimunes/imunologia , Infecções por HIV/complicações , Infecções por HIV/imunologia , Infecções por Herpesviridae/complicações , Infecções por Herpesviridae/virologia , Interações Hospedeiro-Parasita/fisiologia , Humanos , Sistema Imunitário/imunologia , Sistema Imunitário/virologia , Imunidade Heteróloga , Síndromes de Imunodeficiência/complicações , Síndromes de Imunodeficiência/imunologia , Malária/complicações , Malária/imunologia
15.
Mucosal Immunol ; 11(4): 1265-1278, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29545648

RESUMO

A universal influenza vaccine must provide protection against antigenically divergent influenza viruses either through broadly neutralizing antibodies or cross-reactive T cells. Here, intranasal immunizations with recombinant adenoviral vectors (rAd) encoding hemagglutinin (HA) and nucleoprotein (NP) in combination with rAd-Interleukin-(IL)-1ß or rAd-IL-18 were evaluated for their efficacy in BALB/c mice. Mucosal delivery of rAd-IL-1ß enhanced HA-specific antibody responses including strain-specific neutralizing antibodies. Nevertheless, the beneficial effects on the local T cell responses were much more impressive reflected by increased numbers of CD103+CD69+ tissue-resident memory T cells (TRM). This increased immunogenicity translated into superior protection against infections with homologous and heterologous strains including H1N1, pH1N1, H3N2, and H7N7. Inhibition of the egress of circulating T cells out of the lymph nodes during the heterologous infection had no impact on the degree of protection underscoring the unique potential of TRM for the local containment of mucosal infections. The local co-expression of IL-1ß and antigen lead to the activation of critical checkpoints in the formation of TRM including activation of epithelial cells, expression of chemokines and adhesion molecules, recruitment of lung-derived CD103+ DCs, and finally local TRM imprinting. Given the importance of TRM-mediated protection at mucosal barriers, this study has major implications for vaccine development.


Assuntos
Células Dendríticas/imunologia , Vírus da Influenza A/fisiologia , Vacinas contra Influenza/imunologia , Influenza Humana/imunologia , Interleucina-1beta/metabolismo , Infecções por Orthomyxoviridae/imunologia , Linfócitos T/imunologia , Adenoviridae/genética , Adjuvantes Imunológicos , Animais , Anticorpos Neutralizantes/sangue , Anticorpos Antivirais/sangue , Células Cultivadas , Feminino , Vetores Genéticos , Humanos , Imunidade Heteróloga , Memória Imunológica , Interleucina-18/genética , Interleucina-1beta/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Especificidade de Órgãos
16.
mBio ; 8(6)2017 Dec 05.
Artigo em Inglês | MEDLINE | ID: mdl-29208744

RESUMO

Fifty years after the discovery of Epstein-Barr virus (EBV), it remains unclear how primary infection with this virus leads to massive CD8 T-cell expansion and acute infectious mononucleosis (AIM) in young adults. AIM can vary greatly in severity, from a mild transient influenza-like illness to a prolonged severe syndrome. We questioned whether expansion of a unique HLA-A2.01-restricted, cross-reactive CD8 T-cell response between influenza virus A-M158 (IAV-M1) and EBV BMLF1280 (EBV-BM) could modulate the immune response to EBV and play a role in determining the severity of AIM in 32 college students. Only ex vivo total IAV-M1 and IAV-M1+EBV-BM cross-reactive tetramer+ frequencies directly correlated with AIM severity and were predictive of severe disease. Expansion of specific cross-reactive memory IAV-M1 T-cell receptor (TCR) Vß repertoires correlated with levels of disease severity. There were unique profiles of qualitatively different functional responses in the cross-reactive and EBV-specific CD8 T-cell responses in each of the three groups studied, severe-AIM patients, mild-AIM patients, and seropositive persistently EBV-infected healthy donors, that may result from differences in TCR repertoire use. IAV-M1 tetramer+ cells were functionally cross-reactive in short-term cultures, were associated with the highest disease severity in AIM, and displayed enhanced production of gamma interferon, a cytokine that greatly amplifies immune responses, thus frequently contributing to induction of immunopathology. Altogether, these data link heterologous immunity via CD8 T-cell cross-reactivity to CD8 T-cell repertoire selection, function, and resultant disease severity in a common and important human infection. In particular, it highlights for the first time a direct link between the TCR repertoire with pathogenesis and the diversity of outcomes upon pathogen encounter.IMPORTANCE The pathogenic impact of immune responses that by chance cross-react to unrelated viruses has not been established in human infections. Here, we demonstrate that the severity of acute infectious mononucleosis (AIM), an Epstein-Barr virus (EBV)-induced disease prevalent in young adults but not children, is associated with increased frequencies of T cells cross-reactive to EBV and the commonly acquired influenza A virus (IAV). The T-cell receptor (TCR) repertoire and functions of these cross-reactive T cells differed between mild- and severe-AIM patients, most likely because these two groups of patients had selected different memory TCR repertoires in response to IAV infections encountered earlier. This heterologous immunity may explain variability in disease outcome and why young adults with more-developed IAV-specific memory T-cell pools have more-severe disease than children, who have less-developed memory pools. This study provides a new framework for understanding the role of heterologous immunity in human health and disease and highlights an important developing field examining the role of T-cell repertoires in the mediation of immunopathology.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Memória Imunológica , Mononucleose Infecciosa/imunologia , Influenza Humana/imunologia , Receptores de Antígenos de Linfócitos T/imunologia , Proteínas da Matriz Viral/imunologia , Antígenos Virais/imunologia , Reações Cruzadas/imunologia , Epitopos de Linfócito T/imunologia , Epitopos de Linfócito T/metabolismo , Feminino , Antígeno HLA-A2/imunologia , Herpesvirus Humano 4/imunologia , Humanos , Imunidade Heteróloga , Vírus da Influenza A/imunologia , Interferon gama/metabolismo , Ativação Linfocitária , Masculino , Receptores de Antígenos de Linfócitos T/metabolismo , Índice de Gravidade de Doença , Adulto Jovem
17.
Vet Res ; 48(1): 79, 2017 11 21.
Artigo em Inglês | MEDLINE | ID: mdl-29157291

RESUMO

Peste des petits ruminants virus (PPRV) causes an economically important disease that limits productivity in small domestic ruminants and often affects the livestock of the poorest populations in developing countries. Animals that survive PPRV develop strong cellular and humoral responses, which are probably necessary for protection. Vaccination should thus aim at mimicking these natural responses. Immunization strategies against this morbillivirus using recombinant adenoviruses expressing PPRV-F or -H proteins can protect PPRV-challenged animals and permit differentiation of infected from vaccinated animals. Little is known of the T cell repertoire these recombinant vaccines induce. In the present work, we identified several CD4+ and CD8+ T cell epitopes in sheep infected with PPRV. We also show that recombinant adenovirus vaccination induced T cell responses to the same epitopes, and led to memory T cell differentiation. T cells primed by these recombinant adenovirus vaccines expanded after PPRV challenge and probably contributed to protection. These data validate the use of recombinant adenovirus expressing PPRV genes as DIVA strategies to control this highly contagious disease.


Assuntos
Epitopos de Linfócito T/imunologia , Peste dos Pequenos Ruminantes/imunologia , Vírus da Peste dos Pequenos Ruminantes/imunologia , Doenças dos Ovinos/imunologia , Vacinas Virais/imunologia , Animais , Imunidade Heteróloga/imunologia , Camundongos , Ovinos , Linfócitos T/imunologia , Vacinas Sintéticas/imunologia
18.
Int Immunopharmacol ; 52: 136-142, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-28898770

RESUMO

Immunoglobulin therapy is the administration of human polyvalent IgG and represents the most effective treatment to prevent recurrent infections in antibody deficiency patients. Primary antibody deficiency represents the main indication of immunoglobulin replacement therapy and includes a wide range of disorders characterized by impaired antibody production in response to pathogens and recurrent infections. However, not all primary antibody deficiency patients require immunoglobulin replacement. Indeed, immunoglobulin preparations are expensive and, once prescribed, usually result in lifelong therapy. Moreover, many patients significantly benefit from a long-term antibiotic prophylaxis and a prompt begin of antibiotic therapy in case of infectious events. Even more controversial is the decision to initiate immunoglobulin replacement therapy in secondary antibody deficiency, a heterogeneous and expanding group including B-cell lymphoproliferative syndromes, protein losing states and therapeutic agents. This review seeks to define the indication to immunoglobulin replacement in primary and secondary antibody deficiency disorders, distinguishing those in which the beginning of immunoglobulin therapy is always indicated at the same time as the diagnosis has been made, from those lacking of defined indication to replacement therapy. In addition, we propose a clinical approach, mainly based on the evaluation of infectious history, vaccine response and bronchiectasis finding, to support the decision to initiate immunoglobulin therapy in an individual patient.


Assuntos
Síndrome Linfoproliferativa Autoimune/terapia , Imunoglobulina G/uso terapêutico , Síndromes de Imunodeficiência/terapia , Imunoterapia/métodos , Vacinas/imunologia , Antibioticoprofilaxia , Bronquiectasia , Humanos , Imunidade Heteróloga , Seleção de Pacientes
19.
Expert Rev Vaccines ; 16(7): 1-10, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-28532186

RESUMO

INTRODUCTION: Bacillus Calmette-Guérin (BCG) is the only available vaccine against tuberculosis. Although its protective efficacy against pulmonary tuberculosis is still under debate, it provides protection against other mycobacterial diseases. BCG is also an effective therapy against superficial bladder cancer and potentially decreases overall childhood mortality. Areas covered: The purpose of this paper is to provide a state-of-the-art summary of the beneficial effects of BCG in inflammatory and auto-immune diseases. As a strong inducer of Th1 type immunity, BCG has been reported to protect against atopic conditions, such as allergic asthma, a Th2-driven disorder. Its protective effect has been well documented in mice, but still awaits definitive evidence in humans. Similarly, murine studies have shown a protective effect of BCG against auto-immune diseases, such as multiple sclerosis and insulin-dependent diabetes, but studies in humans have come to conflicting conclusions. Expert commentary: Studies in mice have shown a beneficial effect of the BCG vaccine against allergic asthma, multiple sclerosis and diabetes. However, the understanding of its mechanism is still fragmentary and requires further in depth research. Some observational or intervention studies in humans have also suggested a beneficial effect, but definitive evidence for this requires confirmation in carefully conducted prospective studies.


Assuntos
Doenças Autoimunes/prevenção & controle , Autoimunidade , Vacina BCG/uso terapêutico , Inflamação/prevenção & controle , Animais , Doenças Autoimunes/imunologia , Vacina BCG/efeitos adversos , Vacina BCG/imunologia , Modelos Animais de Doenças , Humanos , Imunidade Heteróloga , Inflamação/imunologia , Células Th1/imunologia , Células Th2/imunologia
20.
Trials ; 18(1): 152, 2017 03 31.
Artigo em Inglês | MEDLINE | ID: mdl-28359325

RESUMO

BACKGROUND: Bacillus Calmette-Guérin (BCG) vaccination may have nonspecific effects, i.e., effects on childhood morbidity and mortality that go beyond its effect on the risk of childhood tuberculosis (TB). Though the available scientific literature is mostly from observational studies, and is fraught with controversy, BCG vaccination at birth may protect infants in high-mortality populations against serious infections other than TB. Yet, other studies indicate that giving BCG later in infancy may modify immune responses to non-TB antigens and potentially enhance immunity, potentially also against tuberculosis (TB). It is unclear whether BCG vaccination very early in life offers adequate protection against TB and other infections among HIV-1-exposed children because even those who remain uninfected with HIV-1 show signs of impaired immunocompetence early in infancy. This study will compare BCG vaccination at birth with BCG vaccination at 14 weeks of age in HIV-1-exposed infants. METHODS: This is an individually randomized controlled trial in 2200 HIV-1-exposed infants. The intervention is BCG vaccination within 24 h of birth while the comparator is BCG given at 14 weeks of age. The study co-primary outcomes are severe illness in the first 14 weeks of life, and production of tumor necrosis factor, interleukin (IL)-1ß, IL-6 and interferon-γ in response to mycobacterial and nonmycobacterial antigens. The study is being conducted in three health centers in Uganda. DISCUSSION: A well-timed BCG vaccination could have important nonspecific effects in HIV-1-exposed infants. This trial could inform the development of appropriate timing of BCG vaccination for HIV-1-exposed infants. TRIAL REGISTRATION: ClinicalTrials.gov, identifier: NCT02606526 . Registered on 12 November 2015.


Assuntos
Vacina BCG/administração & dosagem , Infecções por HIV/imunologia , HIV-1/imunologia , Esquemas de Imunização , Tuberculose/prevenção & controle , Vacinação , Vacina BCG/efeitos adversos , Vacina BCG/imunologia , Biomarcadores/sangue , Protocolos Clínicos , Citocinas/sangue , Citocinas/imunologia , Infecções por HIV/transmissão , Infecções por HIV/virologia , HIV-1/patogenicidade , Humanos , Imunidade Heteróloga , Lactente , Recém-Nascido , Transmissão Vertical de Doenças Infecciosas , Projetos de Pesquisa , Fatores de Tempo , Resultado do Tratamento , Tuberculose/imunologia , Tuberculose/microbiologia , Uganda
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA