Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Ann Neurol ; 63(5): 591-601, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18360829

RESUMO

OBJECTIVE: We assessed whether peripheral activation of microglia by a nasal proteosome-based adjuvant (Protollin) that has been given safely to humans can prevent amyloid deposition in young mice and affect amyloid deposition and memory function in old mice with a large amyloid load. METHODS: Amyloid precursor protein (APP) transgenic (Tg) J20 mice received nasal treatment with Protollin weekly for 8 months beginning at age 5 months. Twenty-four-month-old J20 mice were treated weekly for 6 weeks. RESULTS: We found reduction in the level of fibrillar amyloid (93%), insoluble beta-amyloid (Abeta; 68%), and soluble Abeta (45%) fragments in 14-month-old mice treated with Protollin beginning at age 5 months. Twenty-four-month-old mice treated with nasal Protollin for 6 weeks had decreased soluble and insoluble Abeta (1-40) and (1-42) and improved memory function. Activated microglia (CD11b+ cells) colocalized with Abeta fibrils in the 24-month-old animals, and microglial activation correlated with the decrease in Abeta. No microglial activation was observed in 14-month-old mice, suggesting that once Abeta is cleared, there is downregulation of microglial activation. Both groups had reduction in astrocytosis. Protollin was observed in the nasal cavity and cervical lymph node but not in the brain. Activated CD11b+SRA+ (scavenger receptor A) cells were found in blood and cervical lymph node and increased interleukin-10 in cervical lymph node. No toxicity was associated with treatment. INTERPRETATION: Our results demonstrate a novel antibody-independent immunotherapy for both prevention and treatment of Alzheimer's disease that is mediated by peripheral activation of microglia with no apparent toxicity.


Assuntos
Envelhecimento/metabolismo , Amiloide/metabolismo , Encéfalo/metabolismo , Cisteína Endopeptidases/administração & dosagem , Lipopolissacarídeos/administração & dosagem , Microglia/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Administração por Inalação , Envelhecimento/efeitos dos fármacos , Animais , Encéfalo/efeitos dos fármacos , Combinação de Medicamentos , Camundongos , Camundongos Transgênicos , Microglia/efeitos dos fármacos , Complexo de Endopeptidases do Proteassoma/efeitos dos fármacos
2.
J Clin Invest ; 115(9): 2423-33, 2005 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16100572

RESUMO

Amyloid beta-peptide (Abeta) appears to play a key pathogenic role in Alzheimer disease (AD). Immune therapy in mouse models of AD via Abeta immunization or passive administration of Abeta antibodies markedly reduces Abeta levels and reverses behavioral impairment. However, a human trial of Abeta immunization led to meningoencephalitis in some patients and was discontinued. Here we show that nasal vaccination with a proteosome-based adjuvant that is well tolerated in humans plus glatiramer acetate, an FDA-approved synthetic copolymer used to treat multiple sclerosis, potently decreases Abeta plaques in an AD mouse model. This effect did not require the presence of antibody, as it was observed in B cell-deficient (Ig mu-null) mice. Vaccinated animals developed activated microglia that colocalized with Abeta fibrils, and the extent of microglial activation correlated strongly with the decrease in Abeta fibrils. Activation of microglia and clearing of Abeta occurred with the adjuvant alone, although to a lesser degree. Our results identify a novel approach to immune therapy for AD that involves clearing of Abeta through the utilization of compounds that have been safely tested on or are currently in use in humans.


Assuntos
Adjuvantes Imunológicos , Administração Intranasal , Doença de Alzheimer , Peptídeos beta-Amiloides/metabolismo , Cisteína Endopeptidases , Lipopolissacarídeos , Peptídeos/uso terapêutico , Vacinação , Adjuvantes Imunológicos/administração & dosagem , Adjuvantes Imunológicos/uso terapêutico , Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/genética , Animais , Antígenos CD/metabolismo , Barreira Hematoencefálica/citologia , Encéfalo/citologia , Encéfalo/metabolismo , Encéfalo/patologia , Cisteína Endopeptidases/administração & dosagem , Cisteína Endopeptidases/uso terapêutico , Modelos Animais de Doenças , Combinação de Medicamentos , Feminino , Acetato de Glatiramer , Humanos , Lipopolissacarídeos/administração & dosagem , Lipopolissacarídeos/uso terapêutico , Masculino , Camundongos , Camundongos Transgênicos , Complexo de Endopeptidases do Proteassoma
3.
Vaccine ; 35(25): 3318-3325, 2017 06 05.
Artigo em Inglês | MEDLINE | ID: mdl-28499553

RESUMO

Sporadic, yet frequent human infections with avian H5N1 influenza A viruses continue to pose a potential pandemic threat. Poor immunogenicity of unadjuvanted H5N1 vaccines warrants developing novel adjuvants and formulations as well as alternate delivery systems to improve their immunogenicity and efficacy. Here, we show that Protollin, a nasal adjuvant composed of Neisseria meningitides outer membrane proteins non-covalently linked to Shigella flexneri 2a lipopolysaccharide, is a potent nasal adjuvant for an inactivated split virion H5N1 clade 1 A/Viet Nam1203/2004 (A/VN/1203/04) vaccine in a mouse model. Protollin-adjuvanted vaccines elicited enhanced serum protective hemagglutination inhibition titers, mucosal IgA responses, and H5N1-specific cell-mediated immunity that resulted in complete protection against a lethal challenge with a homologous virus as well as a heterologous clade 2 virus A/Indonesia/05/2005 (A/IN/05/05). Detailed analysis of adaptive immunity revealed that Protollin increased the frequency of lymphoid- as well as local tissue-resident antibody-secreting cells, local germinal center reaction of B cells, broad-spectrum of CD4 T cell response. Our findings suggest that nasal delivery of H5N1 vaccine with Protollin adjuvant can overcome the poor immunogenicity of H5N1 vaccines, induce both cellular and humoral immune responses, enhance protection against challenge with clade 1 and clade 2 H5N1 viruses and achieve significant antigen dose-sparing.


Assuntos
Adjuvantes Imunológicos , Cisteína Endopeptidases/imunologia , Imunidade nas Mucosas , Virus da Influenza A Subtipo H5N1/imunologia , Vacinas contra Influenza/imunologia , Lipopolissacarídeos/imunologia , Infecções por Orthomyxoviridae/imunologia , Animais , Anticorpos Antivirais/sangue , Células Produtoras de Anticorpos/imunologia , Linfócitos T CD4-Positivos/imunologia , Modelos Animais de Doenças , Combinação de Medicamentos , Testes de Inibição da Hemaglutinação , Imunidade Celular , Imunidade Humoral , Imunogenicidade da Vacina , Imunoglobulina A/biossíntese , Imunoglobulina A/imunologia , Vacinas contra Influenza/administração & dosagem , Camundongos , Infecções por Orthomyxoviridae/prevenção & controle
4.
Vaccine ; 34(12): 1444-51, 2016 Mar 14.
Artigo em Inglês | MEDLINE | ID: mdl-26873056

RESUMO

INTRODUCTION: There is great interest in developing more effective influenza vaccines for the elderly. Oil-in-water adjuvants can boost humoral responses to seasonal vaccines in elderly subjects but relatively little is known about their mechanism of action. METHODS: We compared humoral and cellular immune profiles in young adult (2 months), mature (11-12 months) or aged (16-17 month) female BALB/c mice following two doses of Alum or AS03-adjuvanted A/H3N2 split-virus antigen (A/Uruguay/716/2007) at 0.75 or 3 µg hemagglutinin (HA) per dose intramuscularly versus 3 µg HA without adjuvant. RESULTS: Overall, hemagglutination inhibition (HAI), microneutralization (MN) and end-point ELISA titres were higher in the young mice and when an adjuvant was used. Both adjuvants increased humoral responses in older animals but the highest titres across all groups were observed in the AS03-adjuvanted groups. Neither IgG avidity nor A/H3N2-specific splenocyte proliferation was influenced by age, antigen dose or adjuvant. In contrast, cytokine production by ex vivo-stimulated splenocytes differed widely between groups. Most cytokine levels in older mice vaccinated with antigen alone (3 µg HA/dose) were ≤ 50% of those in young animals. In young mice, cytokine levels increased modestly with Alum and significantly with AS03. Increases tended to be greatest at the lower antigen dose (0.75 µg versus 3 µg HA). In the older animals, Alum had little impact on cytokine production but responses in the AS03 groups paralleled those of the young mice (broad activation of Th1, Th2, and Th17-type cytokines) and the greatest increases were seen with the higher antigen dose (3 µg HA). CONCLUSIONS: In both young and aged mice, Alum and AS03 increased the magnitude of humoral and cellular responses to split influenza virus vaccination. Overall, these effects were most pronounced in the younger animals and the groups receiving AS03. These data support the use of oil-in-water adjuvants in influenza vaccines targeting the elderly.


Assuntos
Adjuvantes Imunológicos/administração & dosagem , Fatores Etários , Compostos de Alúmen/administração & dosagem , Vacinas contra Influenza/administração & dosagem , Polissorbatos/administração & dosagem , Esqualeno/administração & dosagem , alfa-Tocoferol/administração & dosagem , Animais , Anticorpos Antivirais/sangue , Citocinas/imunologia , Relação Dose-Resposta Imunológica , Combinação de Medicamentos , Feminino , Testes de Inibição da Hemaglutinação , Imunidade Celular , Imunidade Humoral , Vírus da Influenza A Subtipo H3N2 , Camundongos Endogâmicos BALB C , Testes de Neutralização , Infecções por Orthomyxoviridae/prevenção & controle , Baço/imunologia
5.
Front Immunol ; 6: 207, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25972874

RESUMO

During the 2009-2010 influenza pandemic, an adjuvanted, dose-sparing vaccine was recommended for most Canadians. We hypothesize that differences exist in the responses to AS03-adjuvanted, low antigen (Ag) dose versus unadjuvanted, full-dose vaccines. We investigated the relationship between Ag dose and the oil-in-water emulsion Adjuvant System AS03. BALB/c mice received two IM doses of AS03A or AS03B with exaggerated dilutions of A/Uruguay/716/2007 H3N2 split virion vaccine Ag. Immune responses were assessed 3 weeks after the booster. Unadjuvanted "high" (3 µg) and low-dose (0.03-0.003 µg) vaccines generated similar serum antibody titers and cytokine secretion patterns in restimulated splenocytes. Compared to unadjuvanted "high-dose" vaccination, both AS03A and AS03B-adjuvanted low-dose vaccines tended to elicit higher serum antibody titers, broader induction of cytokine secretion and generated more influenza-specific antibody secreting cells and cytokine-secreting CD4 and CD8 T cells in splenocytes. We show that varying Ag and/or AS03 dose in this influenza vaccination mouse model can strongly influence both the magnitude and pattern of the immune response elicited. These findings are highly relevant given the likelihood of expanded use of adjuvanted, dose-sparing vaccines and raise questions about the use of "standard" doses of vaccines in pre-clinical vaccine studies.

6.
Vaccine ; 32(2): 238-45, 2014 Jan 03.
Artigo em Inglês | MEDLINE | ID: mdl-24252699

RESUMO

An inactivated, mucosal mumps virus (MuV) vaccine would address many of the problems associated with current live-attenuated formulations. Protollin (Prl)-based adjuvants (containing TLR2 and TLR4 ligands) are well-suited for nasal administration. We sought to develop an inactivated whole-virus nasal vaccine for MuV using the Prl adjuvant/delivery vehicle and to test tolerability and immunogenicity in a mouse model. BALB/c mice exhibited signs of transient reactogenicity (hunched posture, erect fur, weight loss ≤10% of total body weight) following administration of intranasal MuV-Prl vaccines, though most of these manifestations resolved within 24h. Compared to high-dose unadjuvanted vaccine (8µgMuV), administration of high-dose adjuvanted formulation (8µgMuV-Prl) induced greater MuV-specific serum IgG (3.26E6ng/mL vs. 2.2E5ng/mL, 8µgMuV-Prl vs. 8µgMuV, p<0.001) and mucosal IgA (128ng/mL vs. 45ng/mL, 8µgMuV-Prl vs. 8µgMuV, p<0.05). Serum IgG isotypes and splenocyte cytokine secretion induced by MuV-Prl suggested a predominant T helper cell (Th)1-type immune response. This response was characterized by: (1) ≥four-fold increase of IgG2a levels compared to IgG1; and (2) high IL-2 (644pg/mL)/IFN-γ (228pg/mL) and low IL-5 (31pg/mL) secretion in MuV-restimulated splenocytes from animals receiving MuV-Prl formulations. MuV-Prl vaccination induced higher levels of serum antibodies capable of neutralizing MuV in vitro than MuV alone, particularly for high-dose 8µg formulations (357 neutralizing units (NU)/mL vs. 32NU/mL, 8µgMuV-Prl vs. 8µgMuV, p<0.001). Thus, nasal MuV-Prl vaccines are fairly well-tolerated and highly immunogenic in mice.


Assuntos
Adjuvantes Imunológicos/administração & dosagem , Formação de Anticorpos , Cisteína Endopeptidases/farmacologia , Imunidade nas Mucosas , Lipopolissacarídeos/farmacologia , Vacina contra Caxumba/imunologia , Adjuvantes Imunológicos/farmacologia , Administração Intranasal , Animais , Anticorpos Neutralizantes/sangue , Anticorpos Neutralizantes/imunologia , Células Cultivadas , Cisteína Endopeptidases/administração & dosagem , Citocinas/imunologia , Combinação de Medicamentos , Imunoglobulina A/imunologia , Imunoglobulina G/sangue , Imunoglobulina G/imunologia , Lipopolissacarídeos/administração & dosagem , Camundongos , Camundongos Endogâmicos BALB C , Caxumba/prevenção & controle , Vacina contra Caxumba/administração & dosagem , Baço/citologia , Baço/imunologia , Vacinas de Produtos Inativados/administração & dosagem , Vacinas de Produtos Inativados/imunologia
7.
J Clin Virol ; 44(4): 287-91, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19233722

RESUMO

BACKGROUND: Immunization of mice with the G protein of respiratory syncytial virus (RSV) characteristically induces an immune response that is partially protective, but which can prime for pulmonary eosinophilia. We have shown previously that the N191A mutation in a recombinant RSV G protein fragment is associated with reduced pulmonary eosinophilic infiltration when administered with alum subcutaneously in BALB/c mice followed by RSV challenge. We hypothesize that the performance of this "epitope enhanced" recombinant G protein fragment may be further improved by combining with the newly developed adjuvant, Protollin, coupled with intranasal delivery. OBJECTIVES: To investigate efficacy of an intranasally delivered, Protollin-adjuvanted, epitope-enhanced recombinant G protein vaccine in BALB/c mice. STUDY DESIGN: Recombinant protein, designated Trx-G128-229, consisted of a bacterially expressed central fragment (amino acids 128-229) of the RSV Long strain G protein fused to a fragment of thioredoxin (Trx). BALB/c mice were chosen to evaluate the effectiveness of wild type and epitope-enhanced Trx-G128-229 as a nasal vaccine with the adjuvant Protollin. RESULTS: The intranasal administration of Trx-G128-229 with Protollin conferred similar protection against RSV challenge as subcutaneously administered Trx-G128-229 with alum, but with markedly reduced eosinophilia and the Th2 cytokine IL-13. CONCLUSIONS: These results support the concept of an RSV vaccine optimized by combined strategies, including epitope enhancement and judicious selection of adjuvants.


Assuntos
Adjuvantes Imunológicos/administração & dosagem , Cisteína Endopeptidases/administração & dosagem , Lipopolissacarídeos/administração & dosagem , Infecções por Vírus Respiratório Sincicial/prevenção & controle , Vacinas contra Vírus Sincicial Respiratório/imunologia , Proteínas do Envelope Viral/imunologia , Adjuvantes Imunológicos/farmacologia , Administração Intranasal , Animais , Anticorpos Antivirais/sangue , Cisteína Endopeptidases/farmacologia , Combinação de Medicamentos , Eosinofilia/prevenção & controle , Feminino , Imunoglobulina G/sangue , Lipopolissacarídeos/farmacologia , Pulmão/virologia , Camundongos , Camundongos Endogâmicos BALB C , Vacinas contra Vírus Sincicial Respiratório/administração & dosagem , Vacinas contra Vírus Sincicial Respiratório/genética , Vírus Sinciciais Respiratórios/genética , Vírus Sinciciais Respiratórios/imunologia , Vacinas de Subunidades Antigênicas/administração & dosagem , Vacinas de Subunidades Antigênicas/genética , Vacinas de Subunidades Antigênicas/imunologia , Vacinas Sintéticas/administração & dosagem , Vacinas Sintéticas/genética , Vacinas Sintéticas/imunologia , Proteínas do Envelope Viral/administração & dosagem , Proteínas do Envelope Viral/genética
8.
Vaccine ; 27(3): 421-30, 2009 Jan 14.
Artigo em Inglês | MEDLINE | ID: mdl-19013492

RESUMO

An intranasal vaccine composed of Toll-like receptor 2 (TLR2) ligand Neisseria meningitidis outer membrane proteins and Toll-like receptor 4 (TLR4) ligand Shigella flexneri lipopolysaccharide (LPS) (Protollin) and enriched respiratory syncytial virus (RSV) proteins (eRSV) has been demonstrated to promote balanced Th1/Th2 responses without eosinophil recruitment and to protect against challenge in mouse models. We used TLR2, TLR4 and myeloid differentiation factor 88 (MyD88) knock-out (-/-) mice to investigate the roles of these signalling pathways on immunogenicity, protection and pulmonary infiltrates following RSV immunization and challenge. Antigen-specific systemic and mucosal antibody production was significantly impaired only in TLR4-/- mice following Protollin-eRSV immunization. In contrast, an intact MyD88 pathway was crucial to elicit a balanced type 1:type 2 immune response, characterized by increased splenocyte production of antigen-induced IFNgamma and IL-10 with concomitant reduction of IL5, IgG2a isotype switching and abrogation of pulmonary eosinophil recruitment following challenge. MyD88-dependent signalling also contributed to neutrophil recruitment to the lungs following immunization with eRSV antigen, in the presence or absence of Protollin, compared to a mock antigen or vaccine. Both TLR4 and MyD88-signalling were required for optimal protection against challenge. The upregulation of early signalling molecules IFN-beta, TNFalpha, CD40 and CD86 were studied in splenocytes isolated from naïve TLR2, TLR4 and MyD88-/- mice following stimulation with vaccine components. Splenocytes from TLR4-/- mice displayed reduced IFN-beta while those of MyD88-/- mice elicited less TNFalpha and lower expression of CD40 and CD86 on CD11c+ cells. Together, our results suggest that optimal immunogenicity and protection against RSV without risk of enhanced pulmonary inflammation requires intact TLR4/MyD88-dependent signalling.


Assuntos
Adjuvantes Imunológicos/farmacologia , Proteínas da Membrana Bacteriana Externa/farmacologia , Lipopolissacarídeos/farmacologia , Fator 88 de Diferenciação Mieloide/imunologia , Vacinas contra Vírus Sincicial Respiratório/imunologia , Receptor 4 Toll-Like/imunologia , Animais , Anticorpos Antivirais/sangue , Líquido da Lavagem Broncoalveolar/citologia , Líquido da Lavagem Broncoalveolar/imunologia , Citocinas/biossíntese , Feminino , Leucócitos Mononucleares/imunologia , Pulmão/imunologia , Pulmão/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fator 88 de Diferenciação Mieloide/deficiência , Neisseria meningitidis/imunologia , Neutrófilos/imunologia , Infecções por Vírus Respiratório Sincicial/imunologia , Infecções por Vírus Respiratório Sincicial/prevenção & controle , Shigella flexneri/imunologia , Baço/imunologia , Receptor 2 Toll-Like/deficiência , Receptor 2 Toll-Like/imunologia , Receptor 4 Toll-Like/deficiência
9.
Vaccine ; 25(34): 6334-40, 2007 Aug 21.
Artigo em Inglês | MEDLINE | ID: mdl-17640780

RESUMO

The feasibility of developing a prophylactic vaccine against SARS was assessed by comparing the immune responses elicited by immunizing mice with a recombinant SARS spike glycoprotein (S-protein) formulated with different adjuvants, given by different routes. In both young and aged mice, an intranasal Protollin-formulated S-protein vaccine elicited high levels of antigen-specific IgG in serum, comparable to those elicited by an intramuscular Alum-adsorbed S-protein vaccine. Serum antibodies were shown to be virus neutralizing. Intranasal immunization of young mice with the Protollin-formulated vaccine elicited significant levels of antigen-specific lung IgA in contrast to mice immunized with the intramuscular vaccine in which no antigen-specific lung IgA was detected. Following live virus challenge of aged mice, no virus was detected in the lungs of intranasally immunized mice, in contrast to intramuscularly immunized mice whose lung virus titers were comparable to those observed in control mice.


Assuntos
Anticorpos Antivirais/biossíntese , Cisteína Endopeptidases/administração & dosagem , Lipopolissacarídeos/administração & dosagem , Pulmão/imunologia , Glicoproteínas de Membrana/imunologia , Coronavírus Relacionado à Síndrome Respiratória Aguda Grave/imunologia , Vacinas Sintéticas/administração & dosagem , Proteínas do Envelope Viral/imunologia , Vacinas Virais/administração & dosagem , Administração Intranasal , Animais , Cisteína Endopeptidases/imunologia , Citocinas/biossíntese , Combinação de Medicamentos , Feminino , Imunização , Lipopolissacarídeos/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Glicoproteína da Espícula de Coronavírus , Vacinas Sintéticas/imunologia , Vacinas Virais/imunologia
10.
Vaccine ; 25(29): 5348-58, 2007 Jul 20.
Artigo em Inglês | MEDLINE | ID: mdl-17582662

RESUMO

Proteosome-based vaccines have TLR2-based adjuvant activity and show promise for mucosal immunization. We examined the effects of proteosomes on mucosal uptake in Peyer's patches in vivo. Proteosomes accelerated transepithelial transport of microparticles by M cells and induced migration of dendritic cells (DCs) into the follicle-associated epithelium (FAE); both effects were dependent on TLR2. Proteosomes induced the release of the DC-attracting chemokine MIP3alpha from Caco-2 epithelial cells in vitro. In HEK cells, proteosome-mediated MIP3alpha release was dependent on TLR2 expression and matrix metalloproteinase activation. Thus, TLR2 activation by proteosomes may promote mucosal uptake of particulate vaccines, and this may contribute to their adjuvanticity.


Assuntos
Complexo de Endopeptidases do Proteassoma/imunologia , Receptor 2 Toll-Like/agonistas , Receptor 2 Toll-Like/imunologia , Vacinação/métodos , Animais , Vacinas Bacterianas/imunologia , Vacinas Bacterianas/metabolismo , Linhagem Celular , Quimiocina CCL20 , Quimiocinas CC/biossíntese , Células Dendríticas/imunologia , Feminino , Imunidade nas Mucosas , Imuno-Histoquímica , Mucosa Intestinal/imunologia , Proteínas Inflamatórias de Macrófagos/biossíntese , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Microscopia Confocal , Modelos Animais , Nódulos Linfáticos Agregados/imunologia , Complexo de Endopeptidases do Proteassoma/metabolismo
11.
Vaccine ; 24(10): 1625-32, 2006 Mar 06.
Artigo em Inglês | MEDLINE | ID: mdl-16243411

RESUMO

F1-V is a recombinant plague antigen comprising the capsular (F1) and virulence-associated (V) proteins. Given intramuscularly with Alhydrogel, it protects mice against challenge, but is less effective in non-human primates against high-dose aerosolized Yersinia pestis challenge, perhaps because it fails to induce respiratory immunity. Intranasal immunization of mice with F1-V formulated with a Proteosome-based adjuvant (Protollin), elicited high titers of specific IgA in lungs whereas intranasal F1-V alone or intramuscular Alhydrogel-adsorbed F1-V did not. The Protollin-adjuvanted F1-V vaccine also induced high serum titers of specific IgG, comparable to those induced by intramuscular Alhydrogel-adsorbed F1-V. Mice immunized intranasally with Protollin-F1-V were 100% protected against aerosol challenge with 170 LD50 of Y. pestis and 80% against 255 LD50.


Assuntos
Anticorpos Antibacterianos/biossíntese , Antígenos de Bactérias/imunologia , Proteínas de Bactérias/imunologia , Pulmão/imunologia , Vacina contra a Peste/administração & dosagem , Peste/prevenção & controle , Vacinas Sintéticas/administração & dosagem , Administração Intranasal , Aerossóis , Animais , Feminino , Imunização , Imunoglobulina A/biossíntese , Imunoglobulina G/biossíntese , Interleucina-10/biossíntese , Camundongos , Vacina contra a Peste/imunologia , Proteínas Citotóxicas Formadoras de Poros , Fator de Necrose Tumoral alfa/biossíntese , Vacinas Sintéticas/imunologia
12.
Vaccine ; 23(11): 1374-83, 2005 Feb 03.
Artigo em Inglês | MEDLINE | ID: mdl-15661386

RESUMO

Protollin-MV is a vaccine produced by mixing split measles virus (MV) antigen with the novel adjuvant Protollin (Neisseria meningitidis outer membrane proteins non-covalently complexed with Shigella flexneri 2a lipopolysaccharide). Intranasal immunization of mice with two or three doses of Protollin-MV induces both serum IgG and mucosal IgA with strong neutralizing activity. There is a dose-dependent shift towards lower IgG1:IgG2a ratios and MV-specific IFNgamma production in splenocytes. Intranasal Protollin-MV can therefore induce systemic and mucosal neutralizing antibody responses as well as elicit a balanced TH1/TH2-type response.


Assuntos
Adjuvantes Imunológicos/farmacologia , Anticorpos Antivirais/imunologia , Cisteína Endopeptidases/farmacologia , Imunidade Celular , Lipopolissacarídeos/farmacologia , Vacina contra Sarampo/imunologia , Adjuvantes Imunológicos/administração & dosagem , Administração Intranasal , Animais , Cisteína Endopeptidases/administração & dosagem , Combinação de Medicamentos , Feminino , Imunidade nas Mucosas , Imunoglobulina A/análise , Imunoglobulina G/sangue , Interferon gama/biossíntese , Leucócitos/imunologia , Lipopolissacarídeos/administração & dosagem , Sarampo/prevenção & controle , Vacina contra Sarampo/administração & dosagem , Camundongos , Camundongos Endogâmicos BALB C , Neisseria meningitidis/química , Testes de Neutralização , Shigella flexneri/química , Baço/imunologia , Linfócitos T Auxiliares-Indutores/imunologia , Vacinas de Subunidades Antigênicas/imunologia
13.
J Infect Dis ; 192(8): 1450-5, 2005 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-16170764

RESUMO

Infection with group A streptococcus (GAS) may result in a number of clinical conditions, including the potentially life-threatening postinfectious sequelae of rheumatic fever and rheumatic heart disease. As part of the search for a vaccine to prevent GAS infection, a conformationally constrained and minimally conserved peptide, J14, from the M protein of GAS has been defined. In the present study, J14 was formulated with bacterial outer membrane proteins (proteosomes) and then intranasally administered to outbred mice without additional adjuvant. Such immunization led to high titers of J14-specific serum immunoglobulin (Ig) G and mucosal IgA. After upper respiratory tract GAS challenge, immunized mice demonstrated increased survival and reduced GAS colonization of the throat.


Assuntos
Antígenos de Bactérias/imunologia , Epitopos de Linfócito B/imunologia , Infecções Estreptocócicas/prevenção & controle , Vacinas Estreptocócicas/administração & dosagem , Streptococcus pyogenes/imunologia , Administração Intranasal , Animais , Transmissão de Doença Infecciosa/prevenção & controle , Fezes , Imunoglobulina G/biossíntese , Imunoglobulina G/sangue , Camundongos , Mucosa Nasal/microbiologia , Saliva , Infecções Estreptocócicas/transmissão , Streptococcus pyogenes/patogenicidade
14.
Vaccine ; 22(27-28): 3691-7, 2004 Sep 09.
Artigo em Inglês | MEDLINE | ID: mdl-15315848

RESUMO

Protollin is a novel adjuvant comprising Proteosomes non-covalently complexed with LPS. Intranasal immunization of mice with Protollin combined with detergent-split influenza antigens (HA) or recombinant influenza hemagglutinin (rHA) enhanced serum IgG and mucosal IgA levels by up to 250-fold compared with immunization with the antigens alone. IFN-gamma responses were also enhanced compared to the levels produced by splenocytes from mice immunized with antigen alone, while production of IL-5 was abrogated. Mice immunized with Protollin-rHA were completely protected against lethal challenge with influenza virus, demonstrating that Protollin is an effective mucosal adjuvant for prophylactic vaccines.


Assuntos
Adjuvantes Imunológicos/farmacologia , Cisteína Endopeptidases/farmacologia , Imunidade nas Mucosas/efeitos dos fármacos , Lipopolissacarídeos/farmacologia , Vacinas/imunologia , Administração Intranasal , Animais , Anticorpos Antivirais/análise , Anticorpos Antivirais/biossíntese , Química Farmacêutica , Citocinas/análise , Citocinas/biossíntese , Portadores de Fármacos , Combinação de Medicamentos , Feminino , Hemaglutininas/análise , Hemaglutininas/imunologia , Imunoglobulina A/análise , Imunoglobulina A/biossíntese , Imunoglobulina G/análise , Imunoglobulina G/biossíntese , Vacinas contra Influenza/uso terapêutico , Lipopolissacarídeos/imunologia , Lipossomos , Camundongos , Camundongos Endogâmicos BALB C , Infecções por Orthomyxoviridae/imunologia , Infecções por Orthomyxoviridae/prevenção & controle , Baço/citologia , Vacinas/administração & dosagem , Vacinas Sintéticas/administração & dosagem , Vacinas Sintéticas/imunologia
15.
Infect Immun ; 72(5): 2507-12, 2004 May.
Artigo em Inglês | MEDLINE | ID: mdl-15102757

RESUMO

We have previously shown that a hexavalent group A streptococcal M protein-based vaccine evoked bactericidal antibodies after intramuscular injection. In the present study, we show that the hexavalent vaccine formulated with several different mucosal adjuvants and delivered intranasally induced serum and salivary antibodies that protected mice from intranasal challenge infections with virulent group A streptococci. The hexavalent vaccine was formulated with liposomes with or without monophosphorylated lipid A (MPL), cholera toxin B subunit with or without holotoxin, or proteosomes from Neisseria meningitidis outer membrane proteins complexed with lipopolysaccharide from Shigella flexneri. Intranasal immunization with the hexavalent vaccine mixed with these adjuvants resulted in significant levels of antibodies in serum 2 weeks after the final dose. Mean serum antibody titers were equivalent in all groups of mice except those that were immunized with hexavalent protein plus liposomes without MPL, which were significantly lower. Salivary antibodies were also detected in mice that received the vaccine formulated with the four strongest adjuvants. T-cell proliferative assays and cytokine assays using lymphocytes from cervical lymph nodes and spleens from mice immunized with the hexavalent vaccine formulated with proteosomes indicated the presence of hexavalent protein-specific T cells and a Th1-weighted mixed Th1-Th2 cytokine profile. Intranasal immunization with adjuvanted formulations of the hexavalent vaccine resulted in significant levels of protection (80 to 100%) following intranasal challenge infections with type 24 group A streptococci. Our results indicate that intranasal delivery of adjuvanted multivalent M protein vaccines induces protective antibody responses and may provide an alternative to parenteral vaccine formulations.


Assuntos
Infecções Estreptocócicas/imunologia , Infecções Estreptocócicas/prevenção & controle , Vacinas Estreptocócicas/administração & dosagem , Streptococcus pyogenes/imunologia , Adjuvantes Imunológicos/administração & dosagem , Administração Intranasal , Animais , Anticorpos Antibacterianos/biossíntese , Anticorpos Antibacterianos/sangue , Especificidade de Anticorpos , Células Produtoras de Anticorpos/imunologia , Antígenos de Bactérias , Proteínas de Bactérias/imunologia , Citocinas/biossíntese , Feminino , Técnicas In Vitro , Lipossomos , Ativação Linfocitária , Camundongos , Camundongos Endogâmicos BALB C , Saliva/imunologia , Linfócitos T/imunologia
16.
Vaccine ; 21(25-26): 3706-12, 2003 Sep 08.
Artigo em Inglês | MEDLINE | ID: mdl-12922101

RESUMO

The potential for enhancing the immunogenicity of recombinant (baculovirus-derived) influenza hemagglutinin (rHA) was investigated by comparing the immune responses elicited in mice by an intranasal (i.n.) rHA formulated with Proteosomes, with those induced by intramuscular (i.m.) or i.n. rHA alone. The Proteosome-rHA vaccine induced mucosal responses in the respiratory tract, as well as high serum IgG and hemagglutination inhibition (HAI) titers. In contrast, rHA alone given i.m. induced serum IgG without mucosal responses and was ineffective at inducing either mucosal or systemic responses when given i.n. Only mice immunized with the Proteosome-rHA vaccine were completely protected from both death and acute morbidity following live virus challenge, indicating that the i.n. Proteosome-rHA vaccine induced more complete protective immunity than the same doses of unformulated rHA given i.n. or i.m.


Assuntos
Adjuvantes Imunológicos/farmacologia , Baculoviridae/imunologia , Hemaglutininas/imunologia , Imunidade nas Mucosas/imunologia , Vacinas contra Influenza/imunologia , Vacinas de DNA/imunologia , Administração Intranasal , Animais , Anticorpos Antivirais/biossíntese , Química Farmacêutica , Citocinas/biossíntese , Feminino , Imunoglobulina A/biossíntese , Imunoglobulina G/biossíntese , Indicadores e Reagentes , Vírus da Influenza A/imunologia , Vacinas contra Influenza/administração & dosagem , Camundongos , Camundongos Endogâmicos BALB C , Infecções por Orthomyxoviridae/imunologia , Infecções por Orthomyxoviridae/prevenção & controle , Fenótipo , Baço/citologia , Baço/imunologia , Vacinas de DNA/administração & dosagem , Vacinas Sintéticas/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA