Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 90
Filtrar
Mais filtros

País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Immunity ; 56(10): 2358-2372.e5, 2023 10 10.
Artigo em Inglês | MEDLINE | ID: mdl-37699392

RESUMO

Lung-resident memory B cells (lung-BRMs) differentiate into plasma cells after reinfection, providing enhanced pulmonary protection. Here, we investigated the determinants of lung-BRM differentiation upon influenza infection. Kinetic analyses revealed that influenza nucleoprotein (NP)-specific BRMs preferentially differentiated early after infection and required T follicular helper (Tfh) cell help. BRM differentiation temporally coincided with transient interferon (IFN)-γ production by Tfh cells. Depletion of IFN-γ in Tfh cells prevented lung-BRM differentiation and impaired protection against heterosubtypic infection. IFN-γ was required for expression of the transcription factor T-bet by germinal center (GC) B cells, which promoted differentiation of a CXCR3+ GC B cell subset that were precursors of lung-BRMs and CXCR3+ memory B cells in the mediastinal lymph node. Absence of IFN-γ signaling or T-bet in GC B cells prevented CXCR3+ pre-memory precursor development and hampered CXCR3+ memory B cell differentiation and subsequent lung-BRM responses. Thus, Tfh-cell-derived IFN-γ is critical for lung-BRM development and pulmonary immunity, with implications for vaccination strategies targeting BRMs.


Assuntos
Influenza Humana , Linfócitos T Auxiliares-Indutores , Humanos , Interferon gama/metabolismo , Células B de Memória , Células T Auxiliares Foliculares/metabolismo , Centro Germinativo , Diferenciação Celular , Receptores CXCR3/metabolismo
2.
Mol Ther ; 2024 Sep 18.
Artigo em Inglês | MEDLINE | ID: mdl-39295146

RESUMO

Intradermal Mycobacterium bovis Bacillus Calmette-Guérin (BCG) vaccination is currently the only licensed strategy for preventing tuberculosis (TB). It provides limited protection against pulmonary TB. To enhance the efficacy of BCG, we developed a recombinant BCG expressing exogenous monocyte chemoattractant CC chemokine ligand 2 (CCL2) called rBCG-CCL2. Co-culturing macrophages with rBCG-CCL2 enhances their abilities in migration, phagocytosis, and effector molecule expression. In the mouse model, intranasal vaccination with rBCG-CCL2 induced greater immune cell infiltration and a more extensive innate immune response in lung compared to vaccination with parental BCG, as determined by multiparameter flow cytometry, transcriptomic analysis, and pathological assessments. Moreover, rBCG-CCL2 induced a high frequency of activated macrophages and antigen-specific T helper 1 (Th1) and Th17 T cells in lungs. The enhanced immune microenvironment responded more effectively to intravenous challenge with Mycobacterium tuberculosis (Mtb) H37Ra, leading to significant reductions in H37Ra burden and pathological damage to the lungs and spleen. Intranasal rBCG-CCL2-vaccinated mice rapidly initiated pro-inflammatory Th1 cytokine release and reduced pathological damage to the lungs and spleen during the early stage of H37Ra challenge. The finding that co-expression of CCL2 synergistically enhances the immune barrier induced by BCG provides a model for defining immune correlates and mechanisms of vaccine-elicited protection against TB.

3.
Proc Natl Acad Sci U S A ; 118(19)2021 05 11.
Artigo em Inglês | MEDLINE | ID: mdl-33941704

RESUMO

Intranasal (i.n.) immunization is a promising vaccination route for infectious respiratory diseases such as influenza. Recombinant protein vaccines can overcome the safety concerns and long production phase of virus-based influenza vaccines. However, soluble protein vaccines are poorly immunogenic if administered by an i.n. route. Here, we report that polyethyleneimine-functionalized graphene oxide nanoparticles (GP nanoparticles) showed high antigen-loading capacities and superior immunoenhancing properties. Via a facile electrostatic adsorption approach, influenza hemagglutinin (HA) was incorporated into GP nanoparticles and maintained structural integrity and antigenicity. The resulting GP nanoparticles enhanced antigen internalization and promoted inflammatory cytokine production and JAWS II dendritic cell maturation. Compared with soluble HA, GP nanoparticle formulations induced significantly enhanced and cross-reactive immune responses at both systemic sites and mucosal surfaces in mice after i.n. immunization. In the absence of any additional adjuvant, the GP nanoparticle significantly boosted antigen-specific humoral and cellular immune responses, comparable to the acknowledged potent mucosal immunomodulator CpG. The robust immune responses conferred immune protection against challenges by homologous and heterologous viruses. Additionally, the solid self-adjuvant effect of GP nanoparticles may mask the role of CpG when coincorporated. In the absence of currently approved mucosal adjuvants, GP nanoparticles can be developed into potent i.n. influenza vaccines, providing broad protection. With versatility and flexibility, the GP nanoplatform can be easily adapted for constructing mucosal vaccines for different respiratory pathogens.


Assuntos
Reações Cruzadas/imunologia , Vírus da Influenza A Subtipo H3N2/imunologia , Vacinas contra Influenza/imunologia , Influenza Humana/imunologia , Nanopartículas/química , Infecções por Orthomyxoviridae/imunologia , Administração Intranasal , Animais , Linhagem Celular , Citocinas/imunologia , Citocinas/metabolismo , Feminino , Grafite/química , Grafite/imunologia , Glicoproteínas de Hemaglutininação de Vírus da Influenza/química , Glicoproteínas de Hemaglutininação de Vírus da Influenza/imunologia , Humanos , Imunidade Humoral/efeitos dos fármacos , Imunidade Humoral/imunologia , Imunidade nas Mucosas/efeitos dos fármacos , Imunidade nas Mucosas/imunologia , Vírus da Influenza A Subtipo H3N2/efeitos dos fármacos , Vírus da Influenza A Subtipo H3N2/fisiologia , Vacinas contra Influenza/administração & dosagem , Vacinas contra Influenza/química , Influenza Humana/prevenção & controle , Influenza Humana/virologia , Camundongos Endogâmicos BALB C , Nanopartículas/administração & dosagem , Oligodesoxirribonucleotídeos/química , Oligodesoxirribonucleotídeos/imunologia , Infecções por Orthomyxoviridae/prevenção & controle , Infecções por Orthomyxoviridae/virologia , Polietilenoimina/química , Vacinação/métodos
4.
J Infect Dis ; 228(Suppl 7): S648-S659, 2023 11 13.
Artigo em Inglês | MEDLINE | ID: mdl-37469133

RESUMO

BACKGROUND: Ebolaviruses Ebola (EBOV), Sudan (SUDV), and Bundibugyo (BDBV) cause severe human disease, which may be accompanied by hemorrhagic syndrome, with high case fatality rates. Monovalent vaccines do not offer cross-protection against these viruses whose endemic areas overlap. Therefore, development of a panebolavirus vaccine is a priority. As a vaccine vector, human parainfluenza virus type 3 (HPIV3) has the advantages of needle-free administration and induction of both systemic and local mucosal antibody responses in the respiratory tract. METHODS: To minimize the antivector immunity, genes encoding the HPIV3 envelope proteins F and HN were removed from the vaccine constructs, resulting in expression of only the ebolavirus envelope protein-glycoprotein. These second-generation vaccine constructs were used to develop a combination vaccine against EBOV, SUDV, and BDBV. RESULTS: A single intranasal vaccination of guinea pigs or ferrets with the trivalent combination vaccine elicited humoral responses to each of the targeted ebolaviruses, including binding and neutralizing antibodies, as well as Fc-mediated effector functions. This vaccine protected animals from death and disease caused by lethal challenges with EBOV, SUDV, or BDBV. CONCLUSIONS: The combination vaccine elicited protection that was comparable to that induced by the monovalent vaccines, thus demonstrating the value of this combination trivalent vaccine.


Assuntos
Vacinas contra Ebola , Ebolavirus , Doença pelo Vírus Ebola , Animais , Humanos , Cobaias , Anticorpos Antivirais , Furões , Anticorpos Neutralizantes , Vacinas Combinadas
5.
Mol Ther ; 30(9): 2984-2997, 2022 09 07.
Artigo em Inglês | MEDLINE | ID: mdl-35484842

RESUMO

As the coronavirus disease 2019 (COVID-19) pandemic continues and new severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants of concern emerge, the adaptive immunity initially induced by the first-generation COVID-19 vaccines starts waning and needs to be strengthened and broadened in specificity. Vaccination by the nasal route induces mucosal, humoral, and cellular immunity at the entry point of SARS-CoV-2 into the host organism and has been shown to be the most effective for reducing viral transmission. The lentiviral vaccination vector (LV) is particularly suitable for this route of immunization owing to its non-cytopathic, non-replicative, and scarcely inflammatory properties. Here, to set up an optimized cross-protective intranasal booster against COVID-19, we generated an LV encoding stabilized spike of SARS-CoV-2 Beta variant (LV::SBeta-2P). mRNA vaccine-primed and -boosted mice, with waning primary humoral immunity at 4 months after vaccination, were boosted intranasally with LV::SBeta-2P. A strong boost effect was detected on cross-sero-neutralizing activity and systemic T cell immunity. In addition, mucosal anti-spike IgG and IgA, lung-resident B cells, and effector memory and resident T cells were efficiently induced, correlating with complete pulmonary protection against the SARS-CoV-2 Delta variant, demonstrating the suitability of the LV::SBeta-2P vaccine candidate as an intranasal booster against COVID-19. LV::SBeta-2P vaccination was also fully protective against Omicron infection of the lungs and central nervous system, in the highly susceptible B6.K18-hACE2IP-THV transgenic mice.


Assuntos
COVID-19 , Vacinas Virais , Animais , Anticorpos Neutralizantes , Anticorpos Antivirais , COVID-19/prevenção & controle , Vacinas contra COVID-19 , Humanos , Pulmão , Camundongos , Mucosa , SARS-CoV-2/genética , Vacinação , Vacinas Sintéticas , Vacinas de mRNA
6.
J Nanobiotechnology ; 21(1): 60, 2023 Feb 22.
Artigo em Inglês | MEDLINE | ID: mdl-36814238

RESUMO

BACKGROUND: Unlike the injectable vaccines, intranasal lipid nanoparticle (NP)-based adjuvanted vaccine is promising to protect against local infection and viral transmission. Infection of ferrets with SARS-CoV-2 results in typical respiratory disease and pathology akin to in humans, suggesting that the ferret model may be ideal for intranasal vaccine studies. RESULTS: We developed SARS-CoV-2 subunit vaccine containing both Spike receptor binding domain (S-RBD) and Nucleocapsid (N) proteins (NP-COVID-Proteins) or their mRNA (NP-COVID-mRNA) and NP-monosodium urate adjuvant. Both the candidate vaccines in intranasal vaccinated aged ferrets substantially reduced the replicating virus in the entire respiratory tract. Specifically, the NP-COVID-Proteins vaccine did relatively better in clearing the virus from the nasal passage early post challenge infection. The immune gene expression in NP-COVID-Proteins vaccinates indicated increased levels of mRNA of IFNα, MCP1 and IL-4 in lungs and nasal turbinates, and IFNγ and IL-2 in lungs; while proinflammatory mediators IL-1ß and IL-8 mRNA levels in lungs were downregulated. In NP-COVID-Proteins vaccinated ferrets S-RBD and N protein specific IgG antibodies in the serum were substantially increased at both day post challenge (DPC) 7 and DPC 14, while the virus neutralizing antibody titers were relatively better induced by mRNA versus the proteins-based vaccine. In conclusion, intranasal NP-COVID-Proteins vaccine induced balanced Th1 and Th2 immune responses in the respiratory tract, while NP-COVID-mRNA vaccine primarily elicited antibody responses. CONCLUSIONS: Intranasal NP-COVID-Proteins vaccine may be an ideal candidate to elicit increased breadth of immunity against SARS-CoV-2 variants.


Assuntos
COVID-19 , Vacinas contra Influenza , Humanos , Animais , Idoso , Furões , Imunidade nas Mucosas , SARS-CoV-2 , Carga Viral , Anticorpos Antivirais , Pulmão/patologia , Anticorpos Neutralizantes , Adjuvantes Imunológicos , Vacinas contra COVID-19 , Vacinas de mRNA
7.
J Cell Mol Med ; 25(5): 2621-2632, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33481318

RESUMO

The field of tissue-resident B cells has received increasing attention, yet the feature of tissue B cells in respiratory system is unclear. Here, we first show that non-circulating B cells obtained from nasal, trachea and lung tissues are numerically and phenotypically distinct from their circulating counterparts. Analysis of single cell transcriptome sequence identified multiple differentially expressed genes between non-circulating B cells and circulating B cells, which illustrated their heterogeneity. Furthermore, we found high expression of CXCR3 on non-circulating B cells, and the chemokine CXCL11 was also up-regulated in the respiratory tissues, suggesting that CXCR3-CXCL11 axis might accelerate the local resident of non-circulating B cells in respiratory tract. Interestingly, intranasal immunization with BCG in mice elicited a sustained humoral immune response via induction of IgA and IgG Abs, which revealed the role of B cells. Meanwhile, tissue-resident B cells, IgA+ and IgG+ memory B cells (MBCs) in respiratory tissues, as well as plasma cells in bone marrow, were expanded and maintained, and these subsets probably developed into antibody-producing cells to participate in the local humoral immunity. Our data illustrate the phenotype and function of tissue B cells in the upper and lower airways, provide references for the prospective development of vaccines.


Assuntos
Linfócitos B/imunologia , Linfócitos B/metabolismo , Suscetibilidade a Doenças , Homeostase , Fenótipo , Sistema Respiratório/imunologia , Sistema Respiratório/metabolismo , Imunidade Adaptativa , Animais , Antígenos/imunologia , Biomarcadores , Biologia Computacional/métodos , Suscetibilidade a Doenças/imunologia , Feminino , Perfilação da Expressão Gênica , Imunização , Memória Imunológica , Imunofenotipagem , Camundongos , Especificidade de Órgãos/imunologia , Transcriptoma , Vacinas/administração & dosagem , Vacinas/imunologia
8.
J Immunoassay Immunochem ; 42(4): 424-443, 2021 Jul 04.
Artigo em Inglês | MEDLINE | ID: mdl-33724901

RESUMO

This study reports the influence of peste des petits ruminants (PPR) vaccination on the clinico-pathological outcomes of PPR in the face of an outbreak. Twenty-two West African dwarf goats procured for a different study started showing early signs of PPR during acclimatization. In response, PPR vaccine was administered either intranasally with phytogenic mucoadhesive gum (Group A; n = 6) or without gum (Group B; n = 6); subcutaneously (Group C; n = 6) or not vaccinated (Group D; n = 4) and studied for 21 days. The clinical scores, hematology, serology and pathology scores were evaluated. Clinical signs of PPR were present in all groups, presenting a percentage mortality of 33%; 33%; 64% and 100% for Groups A, B, C, and D, respectively. Polycythemia and mild leukopenia were observed in all groups, and all animals were seropositive by day 7 post-vaccination. The lung consolidation scores were low in Groups A and B, compared to Group C. Histopathological lesions consistent with PPR was observed in the lymphoid organs, gastrointestinal tract, and lungs with the presence of PPR antigen as detected by immunohistochemistry. The findings suggest that intranasal vaccination with or without mucoadhesive gum may influence the outcome of PPR infection more than the subcutaneous route in the face of an outbreak.


Assuntos
Sistemas de Liberação de Medicamentos , Peste dos Pequenos Ruminantes/imunologia , Vacinas Virais/imunologia , Administração Intranasal , Animais , Gengiva/imunologia , Cabras , Injeções Subcutâneas , Masculino , Vírus da Peste dos Pequenos Ruminantes/imunologia , Polímeros/administração & dosagem , Resultado do Tratamento , Vacinas Virais/administração & dosagem
9.
BMC Vet Res ; 16(1): 73, 2020 Mar 04.
Artigo em Inglês | MEDLINE | ID: mdl-32131825

RESUMO

BACKGROUND: Bovine respiratory disease (BRD) continues to be great challenge in calf rearing units. The urgent need to decrease the use of antibiotics and increase animal welfare in beef production has forced us to introduce new preventive methods. Vaccinations could contribute to the solution, but the high incidence of BRD already at an early age has made it difficult to introduce suitable vaccination programs. Challenge studies have shown promising results in 3-14 day old calves vaccinated with intranasal BRD vaccine, but very few field trials are available to assess the efficacy of the intranasal vaccines in field conditions. We evaluated the effect of one dose of commercial intranasal vaccination on calf mortality, daily gain, and treatment incidence for BRD in one calf rearing unit. In total, 497 calves (mean age 19 days) were included in our study, 247 of which were vaccinated at the time of arrival to the unit and 250 served as negative controls (unvaccinated). Vaccinated and unvaccinated calves were situated in separate compartments until weaning. Daily gain, treatment incidence, and mortality were recorded until the calves were transported to the finishing unit, which averaged 154.5 days from arrival. RESULTS: Average daily gain over the complete study period was 1151.9 g/day (SD 137.9) for the vaccinated calves and 1139.5 g/day (SD 135.9) for the unvaccinated calves. Intranasal vaccination combined with older arrival age (17 days or older) resulted in a higher daily gain (47.8 g/day) compared with unvaccinated calves (coef. 0.0478, p = 0.003). This association was not recorded in calves that were younger than 17 days upon arrival. Intranasal vaccination was not significantly associated either with mortality (OR 0.976, p = 0.968) or treatment incidence for BRD (OR 1.341, p = 0.120). In total, six vaccinated calves (2.43%) and six unvaccinated calves (2.40%) died during the study period. CONCLUSIONS: Vaccinating arriving calves with intranasal vaccine in the calf rearing unit did not decrease the mortality or treatment incidence for BRD, but it significantly improved the weight gain in calves transported to the unit at the age of 17 days or older.


Assuntos
Administração Intranasal/veterinária , Complexo Respiratório Bovino/prevenção & controle , Vacinas Virais/administração & dosagem , Animais , Complexo Respiratório Bovino/imunologia , Bovinos , Feminino , Incidência , Masculino , Vacinação/métodos , Vacinação/veterinária , Aumento de Peso
10.
J Immunoassay Immunochem ; 41(3): 311-321, 2020 May 03.
Artigo em Inglês | MEDLINE | ID: mdl-32119592

RESUMO

A study was conducted to evaluate mucoadhesive property and immunomodulatory effect of phytogenic gums from Boswellia frereana, Boswellia carteri andCommiphora myrrha on intranasal Peste des petits ruminants (PPR) vaccination in goats and sheep in an ex-vivo and in-vivo situations. Plant gums were purified, dried and compressed into 500gm tablets. Modified shear stress measurement technique was used on freshly excised trachea and intestine tissues of goat to measure peak adhesion time. Forty eight animals (24 goats and 24 sheep) were divided into eight groups (of 3 goats and 3 sheep) and immunized intranasally with gum-vaccine combinations in two ratios (1:1, 1:2). Antibody against PPR virus was measured on day 14, 28, 42 and 56 post vaccination using H-based PPR bELISA. The peak adhesion time of the different gums was transient. PPR virus antibodies were detected in all immunized goats and sheep but not in unvaccinated control. The best percentage inhibition was recorded for Boswellia carteri-vaccine combination group at a ratio of 1:1. Administration of Boswellia carteri-PPR vaccine combination through intranasal or subcutaneous route, elicited similar antibody titre, implying that the intranasal route may be used as a non-invasive alternative delivery in PPR vaccination of small ruminants.


Assuntos
Anticorpos Antivirais/imunologia , Boswellia/química , Boswellia/imunologia , Peste dos Pequenos Ruminantes/imunologia , Resinas Vegetais/administração & dosagem , Resinas Vegetais/farmacologia , Vacinação , Vacinas Virais/imunologia , Adesividade , Administração Intranasal , Animais , Anticorpos Antivirais/administração & dosagem , Anticorpos Antivirais/isolamento & purificação , Mucosa Gástrica , Cabras , Peste dos Pequenos Ruminantes/terapia , Vírus da Peste dos Pequenos Ruminantes/imunologia , Resinas Vegetais/isolamento & purificação , Ovinos , Vacinas Virais/administração & dosagem , Vacinas Virais/isolamento & purificação
11.
Exp Parasitol ; 201: 1-10, 2019 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-31004570

RESUMO

The aim of this study was to observe the intestinal mucosal/systemic responses triggered by intranasal vaccination using recombinant Trichinella spiralis serine protease (rTsSP) and its capacity to elicit immune protection against larva challenge in a murine model. rTsSP coupled with cholera toxin B subunit (CTB) was used to vaccinate mice via intranasal route. The results revealed that intranasal vaccination with rTsSP plus CTB elicited significantly intestinal local sIgA response and a TsSP-specific systemic antibody response in vaccinated mice. Furthermore, more goblet cells/acidic mucins and IgA-secreting cells were observed in jejunum from vaccinated mice. Anti-rTsSP immune serum strongly recognized the cuticle of various worm stages (muscle larva, intestinal infective larva and adult worm). The level of IFN-γ, IL-4 and IL-10 of rTsSP-vaccinated mice was significantly elevated relative to CTB and PBS control groups. The vaccinated mice exhibited a 71.10% adult reduction at 9 days pi and a 62.10% muscle larva reduction at 42 days pi following larva challenge. Additionally, vaccination with rTsSP also dampened intestinal T. spiralis development and decreased the female fecundity. Our results showed that intranasal vaccination using rTsSP adjuvanted with CTB triggered significantly local sIgA response and systemic concurrent Th1/Th2 response that induced an obvious protection against Trichinella infection.


Assuntos
Serina Proteases/imunologia , Trichinella spiralis/imunologia , Administração Intranasal , Animais , Anticorpos Anti-Helmínticos/sangue , Antígenos de Helmintos/administração & dosagem , Antígenos de Helmintos/imunologia , Citocinas/análise , Duodeno/química , Duodeno/citologia , Ensaio de Imunoadsorção Enzimática , Feminino , Técnica Indireta de Fluorescência para Anticorpo , Células Caliciformes/química , Soros Imunes/imunologia , Imunoglobulina A/sangue , Imunoglobulina A Secretora/análise , Imunoglobulina A Secretora/metabolismo , Imunoglobulina G/sangue , Imunoglobulina M/sangue , Linfonodos/citologia , Linfonodos/imunologia , Masculino , Mesentério , Camundongos , Camundongos Endogâmicos BALB C , Mucinas/isolamento & purificação , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/imunologia , Serina Proteases/administração & dosagem , Organismos Livres de Patógenos Específicos , Baço/citologia , Baço/imunologia , Trichinella spiralis/enzimologia
12.
Infect Immun ; 85(10)2017 10.
Artigo em Inglês | MEDLINE | ID: mdl-28717032

RESUMO

Serotype-specific protection against Streptococcus pneumoniae is an important limitation of the current polysaccharide-based vaccines. To prevent serotype replacement, reduce transmission, and limit the emergence of new variants, it is essential to induce broad protection and restrict pneumococcal colonization. In this study, we used a prototype vaccine formulation consisting of lipopolysaccharide (LPS)-detoxified outer membrane vesicles (OMVs) from Salmonella enterica serovar Typhimurium displaying the variable N terminus of PspA (α1α2) for intranasal vaccination, which induced strong Th17 immunity associated with a substantial reduction of pneumococcal colonization. Despite the variable nature of this protein, a common major histocompatibility complex class (MHC-II) epitope was identified, based on in silico prediction combined with ex vivo screening, and was essential for interleukin-17 A (IL-17A)-mediated cross-reactivity and associated with cross protection. Based on 1,352 PspA sequences derived from a pneumococcal carriage cohort, this OMV-based vaccine formulation containing a single α1α2 type was estimated to cover 19.1% of strains, illustrating the potential of Th17-mediated cross protection.


Assuntos
Proteção Cruzada , Interleucina-17/imunologia , Infecções Pneumocócicas/prevenção & controle , Vacinas Pneumocócicas/imunologia , Salmonella typhimurium/química , Streptococcus pneumoniae/imunologia , Células Th17/imunologia , Administração Intranasal , Animais , Antígenos de Bactérias/imunologia , Antígenos de Bactérias/isolamento & purificação , Proteínas da Membrana Bacteriana Externa/imunologia , Proteínas de Bactérias/genética , Proteínas de Bactérias/imunologia , Simulação por Computador , Epitopos/química , Epitopos/genética , Epitopos/imunologia , Epitopos/isolamento & purificação , Genes MHC da Classe II , Proteínas de Choque Térmico/genética , Proteínas de Choque Térmico/imunologia , Interleucina-17/biossíntese , Lipopolissacarídeos/imunologia , Camundongos , Infecções Pneumocócicas/imunologia , Vacinas Pneumocócicas/química , Salmonella typhimurium/imunologia , Vesículas Secretórias/química , Vesículas Secretórias/imunologia , Vacinação
13.
Immunology ; 150(3): 276-289, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-27775159

RESUMO

Preventive influenza vaccines must be reformulated annually because of antigen shift and drift of circulating influenza viral strains. However, seasonal vaccines do not always match the circulating strains, and there is the ever-present threat that avian influenza viruses may adapt to humans. Hence, a universal influenza vaccine is needed to provide protective immunity against a broad range of influenza viruses. We designed an influenza antigen consisting of three tandem M2e repeats plus HA2, in combination with a detoxified anthrax oedema toxin delivery system (EFn plus PA) to enhance immune responses. The EFn-3×M2e-HA2 plus PA vaccine formulation elicited robust, antigen-specific, IgG responses; and was protective against heterologous influenza viral challenge when intranasally delivered to mice three times. Moreover, use of the detoxified anthrax toxin system as an adjuvant had the additional benefit of generating protective immunity against anthrax. Hence, this novel vaccine strategy could potentially address two major emerging public health and biodefence threats.


Assuntos
Adjuvantes Imunológicos/administração & dosagem , Antraz/imunologia , Antígenos de Bactérias/administração & dosagem , Toxinas Bacterianas/administração & dosagem , Vacinas contra Influenza/imunologia , Influenza Humana/imunologia , Linfócitos T/imunologia , Animais , Anticorpos Antibacterianos/sangue , Anticorpos Antivirais/sangue , Bioterrorismo , Células Cultivadas , Citocinas/metabolismo , Humanos , Ativação Linfocitária , Camundongos , Camundongos Endogâmicos BALB C , Infecções por Orthomyxoviridae/imunologia , Linfócitos T/microbiologia , Linfócitos T/virologia , Vacinação
14.
Virol J ; 14(1): 44, 2017 03 03.
Artigo em Inglês | MEDLINE | ID: mdl-28253901

RESUMO

Despite availability of annual influenza vaccines, influenza causes significant morbidity and mortality in the elderly. This is at least in part a result of immunosenescence; the age-dependent decrease in immunological competence that results in greater susceptibility to infections and reduced responses to vaccination. To improve protective immune responses in this age group, new vaccines strategies, such as the use of adjuvants, are needed. Here, we evaluated the mucosal vaccine adjuvant Endocine™, formulated with split influenza antigen and administered intranasally in aged (20-month old) mice. Humoral immune responses were assessed and compared to unadjuvanted intranasal and subcutaneous vaccines. We show that formulation with Endocine™ significantly enhances hemagglutination inhibition (HI) titers, as well as serum IgG and mucosal IgA antibody titers, compared to both types of unadjuvanted vaccines. Thus, our results indicate that intranasal vaccination with Endocine™ is a possible approach for the development of mucosal influenza vaccines for the elderly.


Assuntos
Adjuvantes Imunológicos/administração & dosagem , Formação de Anticorpos , Antígenos Virais/administração & dosagem , Antígenos Virais/imunologia , Imunidade nas Mucosas , Vacinas contra Influenza/administração & dosagem , Vacinas contra Influenza/imunologia , Administração Intranasal , Animais , Anticorpos Antivirais/sangue , Feminino , Testes de Hemaglutinação , Imunoglobulina A/análise , Imunoglobulina G/sangue , Camundongos Endogâmicos BALB C , Vacinas de Subunidades Antigênicas/administração & dosagem , Vacinas de Subunidades Antigênicas/imunologia
15.
Acta Biochim Biophys Sin (Shanghai) ; 49(1): 74-82, 2017 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-27974288

RESUMO

Respiratory syncytial virus (RSV) is a leading cause of lower respiratory infection in infants and children, but there is still no licensed vaccine available. In this report, we developed virus-like particle (VLP) vaccines based on the Bac-to-Bac baculovirus expression system, consisting of an influenza virus matrix (M1) protein and the RSV fusion protein (F) or glycoprotein (G). These RSV VLPs were identified by western blot analysis and electron microscopy. Female BALB/c mice immunized intranasally (i.n.) with RSV-F VLPs, RSV-G VLPs, or both showed viral-specific antibody responses against RSV. Total IgG, IgG1, IgG2a, and mucosal IgA were detected in mice with RSV-F plus RSV-G VLPs, revealing potent cellular and mucosal immune responses. Moreover, we found that these mixed RSV VLPs conferred enhanced protection against live RSV challenges, showing significant decreases in lung viral replication and obvious attenuation of histopathological changes associated with viral infections. These results demonstrate that RSV-F plus RSV-G VLPs by intranasal vaccination is a promising vaccine candidate that warrants further evaluation using cotton rat and primate models.


Assuntos
Infecções por Vírus Respiratório Sincicial/prevenção & controle , Vacinas de Partículas Semelhantes a Vírus/administração & dosagem , Administração Intranasal , Animais , Feminino , Imunidade Celular , Imunidade nas Mucosas , Camundongos , Camundongos Endogâmicos BALB C , Infecções por Vírus Respiratório Sincicial/imunologia , Vacinas de Partículas Semelhantes a Vírus/imunologia
16.
J Infect Dis ; 214(3): 438-46, 2016 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-27122591

RESUMO

Salmonella infection is an increasingly important public health problem owing to the emergence of multidrug resistance and the lack of broadly efficient vaccines. Novel strategies of vaccination are required to induce protective immune responses at mucosal surfaces and in the circulation, to limit bacteria entry and dissemination. To this aim, intranasal anti-Salmonella vaccination with an innovative formulation composed of gas-filled microbubbles and the pathogen-derived protective protein serodominant secreted effector protein B (SseB-MB) was evaluated in a mouse infection model. Intranasal application of SseB-MB induced gut and systemic immunoglobulin A, T-helper type 17 cell (Th17), and Th1 responses, all of which are associated with natural immunity against Salmonella In vaccinated mice, a significant reduction in bacterial load was observed in intestinal tissues and the spleen after an otherwise lethal oral infection. Therefore, MB serve as an efficient carrier for nasal delivery of a Salmonella antigen that results in protection upon activation of the common mucosal immune system.


Assuntos
Antígenos de Bactérias/imunologia , Proteínas de Bactérias/imunologia , Portadores de Fármacos/administração & dosagem , Trato Gastrointestinal/imunologia , Chaperonas Moleculares/imunologia , Infecções por Salmonella/prevenção & controle , Vacinas contra Salmonella/imunologia , Salmonella/imunologia , Administração Intranasal , Animais , Anticorpos Antibacterianos/análise , Anticorpos Antibacterianos/sangue , Antígenos de Bactérias/administração & dosagem , Carga Bacteriana , Proteínas de Bactérias/administração & dosagem , Modelos Animais de Doenças , Feminino , Trato Gastrointestinal/microbiologia , Imunidade nas Mucosas , Imunoglobulina A/análise , Imunoglobulina A/sangue , Camundongos Endogâmicos BALB C , Chaperonas Moleculares/administração & dosagem , Infecções por Salmonella/imunologia , Vacinas contra Salmonella/administração & dosagem , Células Th1/imunologia , Células Th17/imunologia , Resultado do Tratamento
17.
J Zoo Wildl Med ; 47(2): 551-7, 2016 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-27468028

RESUMO

Three methods for delivering a live attenuated canine distemper virus (CDV) vaccine to domestic cats ( Felis catus ) were investigated, as models for developing vaccination protocols for tigers (Panthera tigris). Twenty domestic cats were randomly divided into four treatment groups: saline injection (negative controls); and oral, intranasal, and subcutaneous vaccinates. Cats were injected with saline or a CDV vaccine (Nobivac DP, Merck) at wk 0 and 4. Blood and nasal swabs were collected at wk 0 (prior to the initial vaccination) and weekly thereafter for 9 wk. Urine samples were collected on wk 1 to 9 after initial vaccination. Forty-nine weeks following the initial vaccination series, three cats from the subcutaneous group and three cats from the intranasal group were revaccinated. Blood was collected immediately prior, and 7 and 21 days subsequent to revaccination. Nasal swabs and urine samples were collected from each cat prior to wk 49 revaccination and daily for 7 days thereafter. Nasal swabs and urine were analyzed by quantitative PCR for vaccine virus presence. Sera were tested for CDV antibodies by virus neutralization. All cats were sero-negative for CDV antibodies at the beginning of the study, and saline-injected cats remained sero-negative throughout the study. A dramatic anamnestic response was seen following wk 4 subcutaneous vaccinations, with titers peaking at wk 6 (geometric mean = 2,435.5). Following wk 49 revaccination, subcutaneous vaccinates again mounted impressive titers (wk 52 geometric mean = 2,048). Revaccination of the intranasal group cats at wk 49 produced a small increase in titers (wk 52 geometric mean = 203). CDV viral RNA was detected in six nasal swabs but no urine samples, demonstrating low viral shedding postvaccination. The strong antibody response to subcutaneous vaccination and the lack of adverse effects suggest this vaccine is safe and potentially protective against CDV infection in domestic cats.


Assuntos
Anticorpos Antivirais/sangue , Doenças do Gato/prevenção & controle , Vírus da Cinomose Canina/imunologia , Cinomose/prevenção & controle , Vacinas Virais/imunologia , Administração Intranasal , Administração Oral , Animais , Doenças do Gato/sangue , Doenças do Gato/virologia , Gatos , Cinomose/sangue , Feminino , Injeções Subcutâneas , Masculino , Vacinas Atenuadas/imunologia , Vacinas Virais/administração & dosagem
18.
Microbiol Immunol ; 59(12): 735-43, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26577130

RESUMO

Although the BCG vaccine can prevent tuberculosis (TB) in infants, its ability to prevent adult pulmonary TB is reportedly limited. Therefore, development of a novel effective vaccine against pulmonary TB has become an international research priority. We have previously reported that intranasal vaccination of mice with a mycobacterial heparin-binding hemagglutinin adhesin (HBHA) plus mucosal adjuvant cholera toxin (CT) enhances production of IFN-γ and anti-HBHA antibody and suppresses extrapulmonary bacterial dissemination after intranasal infection with BCG. In the present study, the effects of intranasal HBHA + CT vaccine on murine pulmonary Mycobacterium tuberculosis (Mtb) infection were examined. Intranasal HBHA + CT vaccination alone failed to reduce the bacterial burden in the infected lung. However, a combination vaccine consisting of s.c. BCG priming and an intranasal HBHA + CT booster significantly enhanced protective immunity against pulmonary Mtb infection on day 14 compared with BCG vaccine alone. Further, it was found that intranasal HBHA + CT vaccine enhanced not only IFN-γ but also IL-17A production by HBHA-specific T cells in the lung after pulmonary Mtb infection. Therefore, this combination vaccine may be a good candidate for a new vaccine strategy against pulmonary TB.


Assuntos
Adesinas Bacterianas/imunologia , Vacina BCG/farmacologia , Lectinas/imunologia , Pulmão/imunologia , Mycobacterium tuberculosis/imunologia , Células Th17/imunologia , Tuberculose Pulmonar/prevenção & controle , Adjuvantes Imunológicos/farmacologia , Animais , Antígenos de Bactérias/imunologia , Vacina BCG/imunologia , Toxina da Cólera/imunologia , Feminino , Interferon gama/biossíntese , Interleucina-17/biossíntese , Interleucina-17/imunologia , Lectinas/genética , Pulmão/patologia , Camundongos , Camundongos Endogâmicos C57BL , Linfócitos T/metabolismo , Células Th1/imunologia , Tuberculose Pulmonar/imunologia
19.
Parasitology ; 142(13): 1640-6, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26394776

RESUMO

The CAF01 adjuvant has previously been shown to be safe for human use and to be a potent adjuvant for several vaccine antigens. In the present work, we sought to optimize the Leishmania amazonensis antigens (LaAg) intranasal vaccine in an attempt to enhance the protective immune responses against Leishmania (infantum) chagasi by using the CAF01 association. LaAg/CAF01 vaccinated mice that were challenged 15 days after booster dose with L. (infantum) chagasi showed a significant reduction in their parasite burden in both the spleen and liver, which is associated with an increase in specific production of IFN-γ and nitrite, and a decrease in IL-4 production. In addition, LaAg/CAF01 intranasal delivery was able to increase lymphoproliferative immune responses after parasite antigen recall. These results suggest the feasibility of using the intranasal route for the delivery of crude antigens and of a human-compatible adjuvant against visceral leishmaniasis.


Assuntos
Antígenos de Protozoários/imunologia , Leishmania infantum/imunologia , Leishmania mexicana/imunologia , Leishmaniose Visceral/prevenção & controle , Vacinas Protozoárias/administração & dosagem , Adjuvantes Imunológicos/administração & dosagem , Administração Intranasal , Animais , Anticorpos Antiprotozoários/sangue , Citocinas/biossíntese , Epitopos , Feminino , Imunidade Celular , Imunoglobulina G/sangue , Fígado/parasitologia , Camundongos , Camundongos Endogâmicos BALB C , Nitritos/metabolismo , Baço/citologia , Baço/imunologia , Baço/parasitologia , Vacinas de Produtos Inativados/administração & dosagem
20.
APMIS ; 2024 Jul 03.
Artigo em Inglês | MEDLINE | ID: mdl-38961516

RESUMO

This study was to evaluate the sufficient safety and effect of the novel influenza vaccine program. It prepared new reassortant influenza virus, with high yield on Vero cells. According to the plaque counting, one dose LAIV was composed with 105 PFU of H1, H3, BY, and BV, respectively. Then mixed this LAIV with compound adjuvant, containing 500 µg/mL of carbopol971P and 50 µg/mL of tetanus toxin. That vaccination was called catt-flu. And it employed the GYZZ02 vaccine (commercialized freeze-dried LAIV, listed in China) as cohort analysis control. All mice received two doses of the vaccine, administered on days 0 and 14, respectively. That catt-flu program could induce more cross-protection with neutralizing antibody against heterogeneous types of influenza virus, not only based on HA but also NA protective antigen, through convenient nasal immunization, which had non-inferiority titter compared with the chicken embryo-derived GYZZ02 vaccine on safe and effect. The Vero cell-derived vaccine (LAIV) combined compound catt adjuvant (contain carbopol971P and tetanus toxin) could provide another safety and protective program of influenza vaccine by intranasal administration, as catt-flu program.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA