Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 52
Filtrar
1.
Virology ; 566: 143-152, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34929590

RESUMO

Annual repeat influenza vaccination raises concerns about protective efficacy against mismatched viruses. We investigated the impact of heterologous prime-boost vaccination on inducing cross protection by designing recombinant influenza viruses with chimeric hemagglutinin (HA) carrying M2 extracellular domains (M2e-HA). Heterologous prime-boost vaccination of C57BL/6 mice with M2e-HA chimeric virus more effectively induced M2e and HA stalk specific IgG antibodies correlating with cross protection than homologous prime-boost vaccination. Induction of M2e and HA stalk specific IgG antibodies was compromised in 1-year old mice, indicating significant aging effects on priming subdominant M2e and HA stalk IgG antibody responses. This study demonstrates that a heterologous prime-boost strategy with recombinant influenza virus expressing extra M2e epitopes provides more effective cross protection than homologous vaccination.


Assuntos
Envelhecimento/imunologia , Anticorpos Antivirais/biossíntese , Glicoproteínas de Hemaglutininação de Vírus da Influenza/genética , Imunoglobulina G/biossíntese , Vacinas contra Influenza/genética , Influenza Humana/prevenção & controle , Infecções por Orthomyxoviridae/prevenção & controle , Fatores Etários , Envelhecimento/genética , Animais , Antígenos Virais/genética , Antígenos Virais/imunologia , Proteção Cruzada , Feminino , Glicoproteínas de Hemaglutininação de Vírus da Influenza/imunologia , Humanos , Imunização Secundária/métodos , Imunogenicidade da Vacina , Vírus da Influenza A Subtipo H1N1/imunologia , Vírus da Influenza A Subtipo H1N1/patogenicidade , Vírus da Influenza A Subtipo H3N2/imunologia , Vírus da Influenza A Subtipo H3N2/patogenicidade , Vacinas contra Influenza/administração & dosagem , Vacinas contra Influenza/biossíntese , Influenza Humana/imunologia , Influenza Humana/virologia , Camundongos , Camundongos Endogâmicos C57BL , Modelos Moleculares , Infecções por Orthomyxoviridae/imunologia , Infecções por Orthomyxoviridae/virologia , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/imunologia , Vacinação/métodos , Vacinas Sintéticas , Proteínas da Matriz Viral/genética , Proteínas da Matriz Viral/imunologia
2.
Sci Rep ; 11(1): 4151, 2021 02 18.
Artigo em Inglês | MEDLINE | ID: mdl-33603072

RESUMO

Hemagglutinin (HA)-based current vaccines provide suboptimum cross protection. Influenza A virus contains an ion channel protein M2 conserved extracellular domain (M2e), a target for developing universal vaccines. Here we generated reassortant influenza virus rgH3N2 4xM2e virus (HA and NA from A/Switzerland/9715293/2013/(H3N2)) expressing chimeric 4xM2e-HA fusion proteins with 4xM2e epitopes inserted into the H3 HA N-terminus. Recombinant rgH3N2 4xM2e virus was found to retain equivalent growth kinetics as rgH3N2 in egg substrates. Intranasal single inoculation of mice with live rgH3N2 4xM2e virus was effective in priming the induction of M2e specific IgG antibody responses in mucosal and systemic sites as well as T cell responses. The rgH3N2 4xM2e primed mice were protected against a broad range of different influenza A virus subtypes including H1N1, H3N2, H5N1, H7N9, and H9N2. The findings support a new approach to improve the efficacy of current vaccine platforms by recombinant influenza virus inducing immunity to HA and cross protective M2e antigens.


Assuntos
Proteção Cruzada/imunologia , Hemaglutininas/imunologia , Vírus da Influenza A Subtipo H3N2/imunologia , Vacinas contra Influenza/imunologia , Influenza Humana/imunologia , Infecções por Orthomyxoviridae/imunologia , Proteínas Recombinantes de Fusão/imunologia , Animais , Anticorpos Antivirais/imunologia , Antígenos Virais/imunologia , Células HEK293 , Humanos , Imunoglobulina G/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Vacinação/métodos
3.
Vaccine ; 37(44): 6656-6664, 2019 10 16.
Artigo em Inglês | MEDLINE | ID: mdl-31542260

RESUMO

Pre-fusion stabilizing mutations (DS-Cav1) in soluble fusion (F) proteins of human respiratory syncytial virus (RSV) were previously reported. Here we investigated the antigenic and immunogenic properties of pre-fusion like RSV F proteins on enveloped virus-like particles (VLP). Additional mutations were introduced to DS-Cav1 (F-dcmTM VLP); fusion peptide deletion and cleavage mutation site 1 (F1d-dcmTM VLP) or both sites (F12d-dcmTM VLP). F1d-dcmTM VLP and F12d-dcmTM VLP displayed higher reactivity against pre-fusion specific site Ø and antigenic site I and II specific monoclonal antibodies, compared to F-dcmTM VLP with DS-Cav1 only. Mice immunized with F1d-dcmTM VLP and F12d-dcmTM VLP induced higher levels of DS-Cav1 pre-fusion specific IgG antibodies, RSV neutralizing activity titers, and effective lung viral clearance after challenge. These results suggest that cleavage site mutations and fusion peptide deletion in addition to DS-Cav1 mutations have contributed to structural stabilization of pre-fusion like F conformation on enveloped VLP, capable of inducing high levels of pre-fusion F specific and RSV neutralizing antibodies.


Assuntos
Imunogenicidade da Vacina , Infecções por Vírus Respiratório Sincicial/prevenção & controle , Vacinas contra Vírus Sincicial Respiratório/imunologia , Vírus Sincicial Respiratório Humano/imunologia , Vacinas de Partículas Semelhantes a Vírus/imunologia , Proteínas Virais de Fusão/imunologia , Animais , Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/imunologia , Linhagem Celular , Chlorocebus aethiops , Modelos Animais de Doenças , Feminino , Humanos , Imunização , Camundongos , Mutação , Testes de Neutralização , Vírus Sincicial Respiratório Humano/genética , Células Vero , Proteínas Virais de Fusão/genética
4.
Immune Netw ; 19(3): e18, 2019 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-31281715

RESUMO

Formalin-inactivated respiratory syncytial virus (RSV) vaccination causes vaccine-enhanced disease (VED) after RSV infection. It is considered that vaccine platforms enabling endogenous synthesis of RSV immunogens would induce favorable immune responses than non-replicating subunit vaccines in avoiding VED. Here, we investigated the immunogenicity, protection, and disease in mice after vaccination with RSV fusion protein (F) encoding plasmid DNA (F-DNA) or virus-like particles presenting RSV F (F-VLP). F-DNA vaccination induced CD8 T cells and RSV neutralizing Abs, whereas F-VLP elicited higher levels of IgG2a isotype and neutralizing Abs, and germinal center B cells, contributing to protection by controlling lung viral loads after RSV challenge. However, mice that were immunized with F-DNA displayed weight loss and pulmonary histopathology, and induced F specific CD8 T cell responses and recruitment of monocytes and plasmacytoid dendritic cells into the lungs. These innate immune parameters, RSV disease, and pulmonary histopathology were lower in mice that were immunized with F-VLP after challenge. This study provides important insight into developing effective and safe RSV vaccines.

5.
Virology ; 534: 1-13, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31163351

RESUMO

Alum adjuvanted formalin-inactivated respiratory syncytial virus (RSV) vaccination resulted in enhanced respiratory disease in young children upon natural infection. Here, we investigated the adjuvant effects of monophosphoryl lipid A (MPL) and oligodeoxynucleotide CpG (CpG) on vaccine-enhanced respiratory disease after fusion (F) protein prime vaccination and RSV challenge in infant and adult mouse models. Combination CpG + MPL adjuvant in RSV F protein single dose priming of infant and adult age mice was found to promote the induction of IgG2a isotype antibodies and neutralizing activity, and lung viral clearance after challenge. CpG + MPL adjuvanted F protein (Fp) priming of infant and adult age mice was effective in avoiding lung histopathology, in reducing interleukin-4+ CD4 T cells and cellular infiltration of monocytes and neutrophils after RSV challenge. This study suggests that combination CpG and MPL adjuvant in RSV subunit vaccination might contribute to priming protective immune responses and preventing inflammatory RSV disease after infection.


Assuntos
Adjuvantes Imunológicos/administração & dosagem , Pulmão/imunologia , Infecções por Vírus Respiratório Sincicial/imunologia , Vacinas contra Vírus Sincicial Respiratório/administração & dosagem , Vírus Sincicial Respiratório Humano/imunologia , Proteínas Virais de Fusão/administração & dosagem , Animais , Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/imunologia , Linfócitos T CD4-Positivos/imunologia , Feminino , Humanos , Pulmão/patologia , Pulmão/virologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Neutrófilos/imunologia , Oligodesoxirribonucleotídeos/administração & dosagem , Oligodesoxirribonucleotídeos/imunologia , Infecções por Vírus Respiratório Sincicial/patologia , Infecções por Vírus Respiratório Sincicial/prevenção & controle , Infecções por Vírus Respiratório Sincicial/virologia , Vacinas contra Vírus Sincicial Respiratório/genética , Vacinas contra Vírus Sincicial Respiratório/imunologia , Vírus Sincicial Respiratório Humano/genética , Vírus Sincicial Respiratório Humano/fisiologia , Vacinação , Proteínas Virais de Fusão/genética , Proteínas Virais de Fusão/imunologia
6.
Antiviral Res ; 168: 100-108, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31150678

RESUMO

Clinical trials with alum-adjuvanted formalin-inactivated human respiratory syncytial virus (FI-RSV) vaccine failed in children due to vaccine-enhanced disease upon RSV infection. In this study, we found that inactivated, detergent-split RSV vaccine (Split) displayed higher reactivity against neutralizing antibodies in vitro and less histopathology in primed adult mice after challenge, compared to FI-RSV. The immunogenicity and efficacy of FI-RSV and Split RSV vaccine were further determined in 2 weeks old mice after a single dose in the absence or presence of monophosphoryl lipid A (MPL) + CpG combination adjuvant. Split RSV with MPL + CpG adjuvant was effective in increasing T helper type 1 (Th1) immune responses and IgG2a isotype antibodies, neutralizing activity, and lung viral clearance as well as modulating immune responses to prevent pulmonary histopathology after RSV vaccination and challenge. This study demonstrates the efficacy of Split RSV as an effective vaccine candidate.


Assuntos
Adjuvantes Imunológicos/administração & dosagem , Infecções por Vírus Respiratório Sincicial/prevenção & controle , Vacinas contra Vírus Sincicial Respiratório/administração & dosagem , Vírus Sincicial Respiratório Humano/imunologia , Compostos de Alúmen/administração & dosagem , Animais , Anticorpos Neutralizantes/sangue , Linhagem Celular , Humanos , Imunoglobulina G/sangue , Lipídeo A/administração & dosagem , Lipídeo A/análogos & derivados , Pulmão/efeitos dos fármacos , Pulmão/patologia , Pulmão/virologia , Camundongos , Camundongos Endogâmicos BALB C , Oligodesoxirribonucleotídeos/administração & dosagem , Infecções por Vírus Respiratório Sincicial/imunologia , Infecções por Vírus Respiratório Sincicial/patologia , Infecções por Vírus Respiratório Sincicial/virologia , Vacinas contra Vírus Sincicial Respiratório/imunologia , Células Th1/imunologia , Vacinas de Produtos Inativados , Carga Viral
7.
PLoS One ; 13(10): e0205071, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30365561

RESUMO

Intramuscular (IM) vaccination with formalin-inactivated respiratory syncytial virus (FI-RSV) failed in clinical trials due to vaccine-enhanced respiratory disease. To test the efficacy of skin vaccination against respiratory syncytial virus (RSV), we investigated the immunogenicity, efficacy, and inflammatory disease after microneedle (MN) patch delivery of FI-RSV vaccine (FI-RSV MN) to the mouse skin with or without an adjuvant of monophosphoryl lipid A (MPL). Compared to IM vaccination, MN patch delivery of FI-RSV was more effective in clearing lung viral loads and preventing weight loss, and in diminishing inflammation, infiltrating immune cells, and T helper type 2 (Th2) CD4 T cell responses after RSV challenge. With MPL adjuvant, MN patch delivery of FI-RSV significantly increased the immunogenicity and efficacy as well as preventing RSV disease as evidenced by lung viral clearance and avoiding pulmonary histopathology. Improved efficacy and prevention of disease by FI-RSV MN with MPL were correlated with no sign of airway resistance, lower levels of Th2 cytokines and infiltrating innate inflammatory cells, and higher levels of Th1 T cell responses into the lung. This study suggests that MN patch delivery of RSV vaccines to the skin with MPL adjuvant would be a promising vaccination method.


Assuntos
Adjuvantes Imunológicos/administração & dosagem , Lipídeo A/análogos & derivados , Infecções por Vírus Respiratório Sincicial/prevenção & controle , Vacinas contra Vírus Sincicial Respiratório/administração & dosagem , Vacinação/métodos , Animais , Anticorpos Antivirais/metabolismo , Linhagem Celular , Humanos , Inflamação/imunologia , Inflamação/patologia , Inflamação/prevenção & controle , Injeções Intramusculares , Lipídeo A/administração & dosagem , Pulmão/imunologia , Pulmão/patologia , Pulmão/virologia , Camundongos Endogâmicos BALB C , Microtecnologia , Agulhas , Infecções por Vírus Respiratório Sincicial/imunologia , Infecções por Vírus Respiratório Sincicial/patologia , Vacinas contra Vírus Sincicial Respiratório/imunologia , Pele , Vacinas de Produtos Inativados/administração & dosagem , Vacinas de Produtos Inativados/imunologia , Carga Viral
8.
Vaccines (Basel) ; 6(4)2018 Sep 20.
Artigo em Inglês | MEDLINE | ID: mdl-30241300

RESUMO

Influenza virus M2 protein has a highly conserved ectodomain (M2e) as a cross-protective antigenic target. We investigated the antigenic and immunogenic properties of tandem repeat M2e (5xM2e) proteins and virus-like particles (5xM2e VLP) to better understand how VLP and protein platform vaccines induce innate and protective adaptive immune responses. Despite the high antigenic properties of 5xM2e proteins, the 5xM2e VLP was superior to 5xM2e proteins in inducing IgG2a isotype antibodies, T cell responses, plasma cells and germinal center B cells as well as in conferring cross protection. Mice primed with 5xM2e VLP were found to be highly responsive to 5xM2e protein boost, overcoming the low immunogenicity and protective efficacy of 5xM2e proteins. Immunogenic differences between VLPs and proteins in priming immune responses might be due to an intrinsic ability of 5xM2e VLP to stimulate dendritic cells secreting T helper type 1 (Th1) cytokines. We also found that 5xM2e VLP was effective in inducing inflammatory cytokines and chemokines, and in recruiting macrophages, monocytes, neutrophils, and CD11b⁺ dendritic cells at the injection site. Therefore, this study provides evidence that 5xM2e VLP is an effective vaccine platform, inducing cross-protection by stimulating innate and adaptive immune responses.

9.
Virology ; 524: 172-181, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30199754

RESUMO

Current licensed adjuvants including aluminum hydroxide (alum) bias immune responses toward T helper type 2 (Th2) immune responses. We tested whether virus-like particles presenting flagellin (Flag VLP) exhibit adjuvant effects on eliciting Th1 type immune responses and improving the efficacy of poor immunogenic tandem repeat M2e (M2e5x) protein vaccine against influenza virus. Co-immunization of mice with Flag VLP and M2e5x protein vaccine induced significantly higher levels of IgG2a isotype (Th1) antibodies in sera and mucosal sites, effector CD4+ T cells secreting IFN-γ and granzyme B, and more effective lung viral clearance and protection compared to alum adjuvant. Flag VLP stimulated primary macrophages and dendritic cells to secrete inflammatory cytokines, which is partially dependent on the Toll-like receptor 5. This study provides insight into developing effective vaccine adjuvants.


Assuntos
Adjuvantes Imunológicos , Flagelina/imunologia , Vírus da Influenza A/imunologia , Vacinas contra Influenza/imunologia , Influenza Humana/prevenção & controle , Vacinas de Partículas Semelhantes a Vírus/imunologia , Proteínas da Matriz Viral/imunologia , Animais , Linfócitos T CD4-Positivos , Feminino , Flagelina/genética , Humanos , Imunidade Celular , Imunidade Humoral , Imunogenicidade da Vacina , Imunoglobulina G/sangue , Influenza Humana/virologia , Camundongos , Camundongos Endogâmicos BALB C , Células Th1/imunologia , Células Th2/imunologia , Proteínas da Matriz Viral/genética
10.
J Virol ; 92(20)2018 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-30068650

RESUMO

The complement pathway is involved in eliminating antigen immune complexes. However, the role of the C3 complement system remains largely unknown in influenza virus M2 extracellular (M2e) domain or hemagglutinin (HA) vaccine-mediated protection after vaccination. Using a C3 knockout (C3 KO) mouse model, we found that complement protein C3 was required for effective induction of immune responses to vaccination with M2e-based or HA-based vaccines, which include isotype class-switched antibodies and effector CD4 and CD8 T cell responses. C3 KO mice after active immunization with cross-protective nonneutralizing M2e-based vaccine were not protected against influenza virus, although low levels of M2e-specific antibodies were protective after passive coadministration with virus in wild-type mice. In contrast, C3 KO mice that were immunized with strain-specific neutralizing HA-based vaccine were protected against homologous virus challenge despite lower levels of HA antibody responses. C3 KO mice showed impaired maintenance of innate immune cells and a defect in innate immune responses upon exposure to antigens. The findings in this study suggest that C3 is required for effective induction of humoral and cellular adaptive immune responses as well as protective immunity after nonneutralizing influenza M2e vaccination.IMPORTANCE Complement is the well-known innate immune defense system involved in the opsonization and lysis of pathogens but is less studied in establishing adaptive immunity after vaccination. Influenza virus HA-based vaccination confers protection via strain-specific neutralizing antibodies, whereas M2e-based vaccination induces a broad spectrum of protection by immunity against the conserved M2e epitopes. This study revealed the critical roles of C3 complement in inducing humoral and cellular immune responses after immunization with M2e or HA vaccines. C3 was found to be required for protection by M2e-based but not by HA-based active vaccination as well as for maintaining innate antigen-presenting cells. Findings in this study have insight into better understanding the roles of C3 complement in inducing effective innate and adaptive immunity as well as in conferring protection by cross-protective conserved M2e vaccination.


Assuntos
Complemento C3/metabolismo , Glicoproteínas de Hemaglutininação de Vírus da Influenza/imunologia , Imunidade Celular , Imunidade Heteróloga , Imunidade Humoral , Vacinas contra Influenza/imunologia , Proteínas da Matriz Viral/imunologia , Animais , Anticorpos Antivirais/sangue , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Complemento C3/deficiência , Proteção Cruzada , Vacinas contra Influenza/administração & dosagem , Camundongos Endogâmicos C57BL , Camundongos Knockout
11.
Artigo em Inglês | MEDLINE | ID: mdl-30044449

RESUMO

Although the effects of age, period, and cohort (APC) on suicide are important, previous work in this area may have been invalid because of an identification problem. We analyzed these effects under three different scenarios to identify vulnerable groups and thus overcame the identification problem. We extracted the annual numbers of suicides from the National Death Register of Korea (1992⁻2015) and estimated the APC effects. The annual average suicide rates in 1992⁻2015 were 31.5 and 14.7 per 100,000 males and females, respectively. The APC effects on suicide were similar in both sexes. The age effect was clearly higher in older subjects, in contrast to the minimal changes apparent during earlier adulthood. The birth cohort effect showed an inverted U shape; a higher cohort effect was evident in females born in the early 1980s when period drift was larger than 3.7%/year. Period effect increased sharply during the early 1990s and 2000s. We found that elderly and young females may be at a particularly high risk of suicide in Korea.


Assuntos
Suicídio/tendências , Adolescente , Adulto , Fatores Etários , Idoso , Efeito de Coortes , Estudos de Coortes , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , República da Coreia/epidemiologia
12.
Nat Commun ; 9(1): 359, 2018 01 24.
Artigo em Inglês | MEDLINE | ID: mdl-29367723

RESUMO

Current influenza vaccines provide limited protection against circulating influenza A viruses. A universal influenza vaccine will eliminate the intrinsic limitations of the seasonal flu vaccines. Here we report methodology to generate double-layered protein nanoparticles as a universal influenza vaccine. Layered nanoparticles are fabricated by desolvating tetrameric M2e into protein nanoparticle cores and coating these cores by crosslinking headless HAs. Representative headless HAs of two HA phylogenetic groups are constructed and purified. Vaccinations with the resulting protein nanoparticles in mice induces robust long-lasting immunity, fully protecting the mice against challenges by divergent influenza A viruses of the same group or both groups. The results demonstrate the importance of incorporating both structure-stabilized HA stalk domains and M2e into a universal influenza vaccine to improve its protective potency and breadth. These potent disassemblable protein nanoparticles indicate a wide application in protein drug delivery and controlled release.


Assuntos
Anticorpos Antivirais/imunologia , Hemaglutininas/imunologia , Vírus da Influenza A/imunologia , Vacinas contra Influenza/imunologia , Influenza Humana/virologia , Nanopartículas , Infecções por Orthomyxoviridae/virologia , Animais , Humanos , Vacinas contra Influenza/uso terapêutico , Influenza Humana/prevenção & controle , Camundongos , Complexos Multiproteicos/imunologia , Infecções por Orthomyxoviridae/prevenção & controle , Filogenia
13.
Vaccines (Basel) ; 5(4)2017 Dec 19.
Artigo em Inglês | MEDLINE | ID: mdl-29257056

RESUMO

Heterologous prime boost with DNA and recombinant modified vaccinia virus Ankara (rMVA) vaccines is considered as a promising vaccination approach against human immunodeficiency virus (HIV-1). To further enhance the efficacy of DNA-rMVA vaccination, we investigated humoral and cellular immune responses in mice after three sequential immunizations with DNA, a combination of DNA and virus-like particles (VLP), and rMVA expressing HIV-1 89.6 gp120 envelope proteins (Env). DNA prime and boost with a combination of VLP and DNA vaccines followed by an rMVA boost induced over a 100-fold increase in Env-specific IgG antibody titers compared to three sequential immunizations with DNA and rMVA. Cellular immune responses were induced by VLP-DNA and rMVA vaccinations at high levels in CD8 T cells, CD4 T cells, and peripheral blood mononuclear cells secreting interferon (IFN)-γ, and spleen cells producing interleukin (IL)-2, 4, 5 cytokines. This study suggests that a DNA and VLP combination vaccine with MVA is a promising strategy in enhancing the efficacy of DNA-rMVA vaccination against HIV-1.

14.
Hum Vaccin Immunother ; 13(11): 2594-2605, 2017 11 02.
Artigo em Inglês | MEDLINE | ID: mdl-28854003

RESUMO

Respiratory syncytial virus (RSV) fusion (F) protein is suggested to be a protective vaccine target although its efficacy and safety concerns remain not well understood. We investigated immunogenicity, efficacy, and safety of F proteins in a soluble form or on virus-like particle (F-VLP). F VLP preferentially elicited IgG2a antibody and T helper type 1 (Th1) immune responses whereas F protein induced IgG1 isotype and Th2 responses. Despite lung viral clearance after prime or prime-boost and then RSV challenge, F protein immune mice displayed weight loss and lung histopathology and high mucus production and eosinophils. In contrast, prime or prime-boost vaccination of F VLP induced effective protection, prevented infiltration of eosinophils and vaccine- enhanced disease after challenge. This study provides insight into developing an effective and safe RSV vaccine candidate.


Assuntos
Pulmão/patologia , Infecções por Vírus Respiratório Sincicial/prevenção & controle , Vacinas contra Vírus Sincicial Respiratório/efeitos adversos , Vacinas de Partículas Semelhantes a Vírus/imunologia , Proteínas Virais de Fusão/efeitos adversos , Proteínas Virais de Fusão/imunologia , Animais , Anticorpos Neutralizantes/biossíntese , Anticorpos Antivirais , Humanos , Imunogenicidade da Vacina , Imunoglobulina G/biossíntese , Pulmão/virologia , Camundongos , Camundongos Endogâmicos BALB C , Muco , Infecções por Vírus Respiratório Sincicial/imunologia , Infecções por Vírus Respiratório Sincicial/virologia , Vacinas contra Vírus Sincicial Respiratório/administração & dosagem , Vacinas contra Vírus Sincicial Respiratório/imunologia , Vírus Sincicial Respiratório Humano/imunologia , Células Th1 , Células Th2 , Vacinação , Vacinas de Partículas Semelhantes a Vírus/administração & dosagem , Vacinas de Partículas Semelhantes a Vírus/efeitos adversos , Proteínas Virais de Fusão/administração & dosagem , Redução de Peso
15.
Virology ; 511: 142-151, 2017 11.
Artigo em Inglês | MEDLINE | ID: mdl-28846899

RESUMO

Formalin inactivated respiratory syncytial virus (FI-RSV) vaccination caused vaccine-enhanced respiratory disease (ERD) upon exposure to RSV in children. Virus-like particles presenting RSV F fusion protein (F VLP) are known to increase T helper type-1 (Th1) immune responses and avoid ERD in animal models. We hypothesized that F VLP would prime immune responses preventing ERD upon subsequent exposure to ERD-prone FI-RSV. Here, we demonstrated that heterologous F VLP priming and FI-RSV boosting of mice prevented FI-RSV vaccine-enhanced lung inflammation and eosinophilia upon RSV challenge. F VLP priming redirected pulmonary T cells toward effector CD8 T cells producing Th1 cytokines and significantly suppressed pulmonary Th2 cytokines. This study suggests that RSV F VLP priming would modulate and shift immune responses to subsequent exposure to ERD-prone FI-RSV vaccine and RSV infection, suppressing Th2 immune-mediated pulmonary histopathology and eosinophilia.


Assuntos
Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos/prevenção & controle , Infecções por Vírus Respiratório Sincicial/prevenção & controle , Vacinas contra Vírus Sincicial Respiratório/efeitos adversos , Vacinas contra Vírus Sincicial Respiratório/imunologia , Vírus Sinciciais Respiratórios/imunologia , Vacinas de Partículas Semelhantes a Vírus/imunologia , Animais , Linfócitos T CD8-Positivos/imunologia , Citocinas/metabolismo , Camundongos , Células Th1/imunologia , Vacinas de Produtos Inativados/efeitos adversos , Vacinas de Produtos Inativados/imunologia
16.
Hum Vaccin Immunother ; 13(5): 1031-1039, 2017 05 04.
Artigo em Inglês | MEDLINE | ID: mdl-28129031

RESUMO

Vaccine-enhanced disease has been a major obstacle in developing a safe vaccine against respiratory syncytial virus (RSV). This study demonstrates the immunogenicity, efficacy, and safety of virus-like particle (VLP) vaccines containing RSV F (F VLP), G (G VLP), or F and G proteins (FG VLP) in cotton rats. RSV specific antibodies were effectively induced by vaccination of cotton rats with F VLP or FG VLP vaccines. After challenge, lung RSV clearance was observed with RSV F, G, FG VLP, and formalin inactivated RSV (FI-RSV) vaccines. Upon RSV infection, cotton rats with RSV VLP vaccines were protected against airway hyper-responsiveness and weight loss, which are different from FI-RSV vaccination exhibiting vaccine-enhanced disease of airway obstruction, weight loss, and severe histopathology with eosinophilia and mucus production. FG VLP and F VLP vaccines did not cause pulmonary inflammation whereas G VLP induced moderate lung inflammation with eosinophilia and mucus production. In particular, F VLP and FG VLP vaccines were found to be effective in inducing antibody secreting cell responses in bone marrow and lymphoid organs as well as avoiding the induction of T helper type 2 cytokines. These results provide further evidence to develop a safe RSV vaccine based on VLP platforms.


Assuntos
Infecções por Vírus Respiratório Sincicial/prevenção & controle , Vacinas contra Vírus Sincicial Respiratório/imunologia , Vírus Sincicial Respiratório Humano/imunologia , Vacinas de Partículas Semelhantes a Vírus/imunologia , Proteínas do Envelope Viral/imunologia , Proteínas Virais de Fusão/imunologia , Animais , Anticorpos Neutralizantes , Anticorpos Antivirais/sangue , Imunogenicidade da Vacina , Pulmão/patologia , Pneumonia , Ratos , Infecções por Vírus Respiratório Sincicial/imunologia , Vacinas contra Vírus Sincicial Respiratório/administração & dosagem , Vacinas contra Vírus Sincicial Respiratório/efeitos adversos , Sigmodontinae , Vacinas de Partículas Semelhantes a Vírus/administração & dosagem , Vacinas de Partículas Semelhantes a Vírus/efeitos adversos , Proteínas do Envelope Viral/genética , Proteínas Virais de Fusão/genética
17.
J Biomed Nanotechnol ; 13(1): 84-98, 2017 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-29302248

RESUMO

Cellular immune correlates conferring protection against respiratory syncytial virus (RSV) but preventing vaccine-enhanced respiratory disease largely remain unclear. We investigated cellular immune correlates that contribute to preventing disease against human respiratory syncytial virus (RSV) by nanoparticle vaccine delivery. Formalin-inactivated RSV (FI-RSV) vaccines and virus-like nanoparticles carrying RSV fusion proteins (F VLP) were investigated in mice. The FI-RSV vaccination caused severe weight loss and histopathology by inducing interleukin (IL)-4+, interferon (IFN)-γ+, IL-4+IFN-γ+ CD4+ T cells, eosinophils, and lung plasmacytoid dendritic cells (DCs), CD103+ DCs, and CD11b+ DCs. In contrast, the F VLP-immune mice induced protection against RSV without disease by inducing natural killer cells, activated IFN-γ+, and IFN-γ+ tumor necrosis factor (TNF)-α+ CD8+ T cells in the lung and bronchiolar airways during RSV infection but not disease-inducing DCs and effector T cells. Clodronate-mediated depletion studies provided evidence that alveolar macrophages that were present at high levels in the F VLP-immune mice play a role in modulating protective cellular immune phenotypes. There was an intrinsic difference between the F VLP and FI-RSV treatments in stimulating proinflammatory cytokines. The F VLP nanoparticle vaccination induced distinct innate and adaptive cellular subsets that potentially prevented lung disease after RSV infection.


Assuntos
Nanopartículas/química , Proteínas Recombinantes de Fusão/imunologia , Infecções por Vírus Respiratório Sincicial/imunologia , Vírus Sinciciais Respiratórios/imunologia , Vacinas de Produtos Inativados/imunologia , Vacinas Virais/imunologia , Animais , Ácido Clodrônico , Citocinas/análise , Citocinas/imunologia , Pulmão/patologia , Macrófagos Alveolares/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Nanomedicina , Vacinas de Produtos Inativados/efeitos adversos , Vacinas Virais/efeitos adversos , Vírion/imunologia
18.
Virology ; 494: 215-24, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27123586

RESUMO

A safe and effective vaccine against respiratory syncytial virus (RSV) should confer protection without causing vaccine-enhanced disease. Here, using a cotton rat model, we investigated the protective efficacy and safety of an RSV combination vaccine composed of F-encoding plasmid DNA and virus-like particles containing RSV fusion (F) and attachment (G) glycoproteins (FFG-VLP). Cotton rats with FFG-VLP vaccination controlled lung viral replication below the detection limit, and effectively induced neutralizing activity and antibody-secreting cell responses. In comparison with formalin inactivated RSV (FI-RSV) causing severe RSV disease after challenge, FFG-VLP vaccination did not cause weight loss, airway hyper-responsiveness, IL-4 cytokines, histopathology, and infiltrates of proinflammatory cells such as eosinophils. FFG-VLP was even more effective in preventing RSV-induced pulmonary inflammation than live RSV infections. This study provides evidence that FFG-VLP can be developed into a safe and effective RSV vaccine candidate.


Assuntos
Infecções por Vírus Respiratório Sincicial/imunologia , Infecções por Vírus Respiratório Sincicial/prevenção & controle , Vacinas contra Vírus Sincicial Respiratório/imunologia , Vírus Sinciciais Respiratórios/genética , Vírus Sinciciais Respiratórios/imunologia , Vacinas de DNA/imunologia , Vacinas de Partículas Semelhantes a Vírus/imunologia , Animais , Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/imunologia , Citocinas/metabolismo , Modelos Animais de Doenças , Imunização , Imunoglobulina G/imunologia , Ratos , Infecções por Vírus Respiratório Sincicial/patologia , Infecções por Vírus Respiratório Sincicial/virologia , Vacinas contra Vírus Sincicial Respiratório/administração & dosagem , Vacinas contra Vírus Sincicial Respiratório/efeitos adversos , Vacinas contra Vírus Sincicial Respiratório/genética , Sigmodontinae , Vacinas de DNA/administração & dosagem , Vacinas de DNA/efeitos adversos , Vacinas de DNA/genética , Vacinas de Partículas Semelhantes a Vírus/administração & dosagem , Vacinas de Partículas Semelhantes a Vírus/efeitos adversos , Vacinas de Partículas Semelhantes a Vírus/genética , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/imunologia , Proteínas Virais de Fusão/genética , Proteínas Virais de Fusão/imunologia
20.
PLoS One ; 10(10): e0139916, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26468884

RESUMO

Respiratory syncytial virus (RSV) is well-known for inducing vaccine-enhanced respiratory disease after vaccination of young children with formalin-inactivated RSV (FI-RSV) in alum formulation. Here, we investigated alum adjuvant effects on protection and disease after FI-RSV immunization with or without alum in comparison with live RSV reinfections. Despite viral clearance, live RSV reinfections caused weight loss and substantial pulmonary inflammation probably due to high levels of RSV specific IFN-γ+IL4-, IFN-γ-TNF-α+, IFN-γ+TNF-α- effector CD4 and CD8 T cells. Alum adjuvant significantly improved protection as evidenced by effective viral clearance compared to unadjuvanted FI-RSV. However, in contrast to unadjuvanted FI-RSV, alum-adjuvanted FI-RSV (FI-RSV-A) induced severe vaccine-enhanced RSV disease including weight loss, eosinophilia, and lung histopathology. Alum adjuvant in the FI-RSV-A was found to be mainly responsible for inducing high levels of RSV-specific IFN-γ-IL4+, IFN-γ-TNF-α+ CD4+ T cells, and proinflammatory cytokines IL-6 and IL-4 as well as B220+ plasmacytoid and CD4+ dendritic cells, and inhibiting the induction of IFN-γ+CD8 T cells. This study suggests that alum adjuvant in FI-RSV vaccines increases immunogenicity and viral clearance but also induces atypical T helper CD4+ T cells and multiple inflammatory dendritic cell subsets responsible for vaccine-enhanced severe RSV disease.


Assuntos
Imunidade Adaptativa/imunologia , Compostos de Alúmen/administração & dosagem , Imunidade Inata/imunologia , Pneumonia/patologia , Infecções por Vírus Respiratório Sincicial/prevenção & controle , Vírus Sinciciais Respiratórios/imunologia , Adjuvantes Imunológicos/administração & dosagem , Animais , Linfócitos T CD4-Positivos/imunologia , Ensaio de Imunoadsorção Enzimática , Feminino , Citometria de Fluxo , Imunização , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Pneumonia/imunologia , Pneumonia/virologia , Infecções por Vírus Respiratório Sincicial/imunologia , Infecções por Vírus Respiratório Sincicial/virologia , Linfócitos T Auxiliares-Indutores/imunologia , Vacinas Virais/uso terapêutico
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA