Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Vaccines (Basel) ; 12(4)2024 Mar 27.
Artículo en Inglés | MEDLINE | ID: mdl-38675744

RESUMEN

The emergence of new SARS-CoV-2 variants continues to cause challenging problems for the effective control of COVID-19. In this study, we tested the hypothesis of whether a strategy of multivalent and sequential heterologous spike protein vaccinations would induce a broader range and higher levels of neutralizing antibodies against SARS-CoV-2 variants and more effective protection than homologous spike protein vaccination in a mouse model. We determined spike-specific IgG, receptor-binding inhibition titers, and protective efficacy in the groups of mice that were vaccinated with multivalent recombinant spike proteins (Wuhan, Delta, Omicron), sequentially with heterologous spike protein variants, or with homologous spike proteins. Trivalent (Wuhan + Delta + Omicron) and sequential heterologous spike protein vaccinations were more effective in inducing serum inhibition activities of receptor binding to spike variants and virus neutralizing antibody titers than homologous spike protein vaccination. The higher efficacy of protection was observed in mice with trivalent and sequential heterologous spike protein vaccination after a challenge with a mouse-adapted SARS-CoV-2 MA10 strain compared to homologous spike protein vaccination. This study provides evidence that a strategy of multivalent and sequential heterologous variant spike vaccination might provide more effective protection against emerging SARS-CoV-2 variants than homologous spike vaccination and significantly alleviate severe inflammation due to COVID-19.

2.
Antiviral Res ; 225: 105877, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38561077

RESUMEN

The conventional inactivated split seasonal influenza vaccine offers low efficacy, particularly in the elderly and against antigenic variants. Here, to improve the efficacy of seasonal vaccination for the elderly population, we tested whether supplementing seasonal bivalent (H1N1 + H3N2) split (S) vaccine with M2 ectodomain repeat and multi-subtype consensus neuraminidase (NA) proteins (N1 NA + N2 NA + flu B NA) on a virus-like particle (NA-M2e) would induce enhanced cross-protection against different influenza viruses in aged mice. Immunization with split vaccine plus NA-M2e (S + NA-M2e) increased vaccine-specific IgG antibodies towards T-helper type 1 responses and hemagglutination inhibition titers. Aged mice with NA-M2e supplemented vaccination were protected against homologous and heterologous viruses at higher efficacies, as evidenced by preventing weight loss, lowering lung viral loads, inducing broadly cross-protective humoral immunity, and IFN-γ+ CD4 and CD8 T cell responses than those with seasonal vaccine. Overall, this study supports a new strategy of NA-M2e supplemented vaccination to enhance protection against homologous and antigenically different viruses in the elderly.


Asunto(s)
Subtipo H1N1 del Virus de la Influenza A , Vacunas contra la Influenza , Gripe Humana , Infecciones por Orthomyxoviridae , Anciano , Humanos , Ratones , Animales , Infecciones por Orthomyxoviridae/prevención & control , Neuraminidasa , Subtipo H3N2 del Virus de la Influenza A , Estaciones del Año , Anticuerpos Antivirales , Protección Cruzada , Ratones Endogámicos BALB C
3.
Vaccines (Basel) ; 11(7)2023 Jul 06.
Artículo en Inglés | MEDLINE | ID: mdl-37515025

RESUMEN

With concerns about the efficacy of repeat annual influenza vaccination, it is important to better understand the impact of priming vaccine immunity and develop an effective vaccination strategy. Here, we determined the impact of heterologous prime-boost vaccination on inducing broader protective immunity compared to repeat vaccination with the same antigen. The primed mice that were intramuscularly boosted with a heterologous inactivated influenza A virus (H1N1, H3N2, H5N1, H7N9, H9N2) vaccine showed increased strain-specific hemagglutination inhibition titers against prime and boost vaccine strains. Heterologous prime-boost vaccination of mice with inactivated viruses was more effective in inducing high levels of IgG antibodies specific for groups 1 and 2 hemagglutinin stalk domains, as well as cross-protection, compared to homologous vaccination. Both humoral and T cell immunity were found to play a critical role in conferring cross-protection by heterologous prime-boost vaccination. These results support a strategy to enhance cross-protective efficacy by heterologous prime-boost influenza vaccination.

4.
Immun Ageing ; 20(1): 20, 2023 May 11.
Artículo en Inglés | MEDLINE | ID: mdl-37170231

RESUMEN

BACKGROUND: Current influenza vaccines deliver satisfactory results in young people but are less effective in the elderly. Development of vaccines for an ever-increasing aging population has been an arduous challenge due to immunosenescence that impairs the immune response in the aged, both quantitatively and qualitatively. RESULTS: To potentially enhance vaccine efficacy in the elderly, we investigated the immunogenicity and cross-protection of influenza hemagglutinin virus-like particles (HA-VLP) incorporated with glycosylphosphatidylinositol (GPI)-anchored cytokine-adjuvants (GPI-GM-CSF and GPI-IL-12) via protein transfer in aged mice. Lung viral replication against homologous and heterologous influenza viruses was significantly reduced in aged mice after vaccination with cytokine incorporated VLPs (HA-VLP-Cyt) in comparison to HA-VLP alone. Enhanced IFN-γ+CD4+ and IFN-γ+CD8+ T cell responses were also observed in aged mice immunized with HA-VLP-Cyt when compared to HA-VLP alone. CONCLUSIONS: Cytokine-adjuvanted influenza HA-VLP vaccine induced enhanced protective response against homologous influenza A virus infection in aged mice. Influenza HA-VLP vaccine with GPI-cytokines also induced enhanced T cell responses correlating with better protection against heterologous infection in the absence of neutralizing antibodies. The results suggest that a vaccination strategy using cytokine-adjuvanted influenza HA-VLPs could be used to enhance protection against influenza A virus in the elderly.

5.
Vaccines (Basel) ; 10(9)2022 Aug 24.
Artículo en Inglés | MEDLINE | ID: mdl-36146461

RESUMEN

Adjuvants can increase the magnitude and durability of the immune response generated by the vaccine antigen. Aluminum salts (Alum) remain the main adjuvant licensed for human use. A few new adjuvants have been licensed for use in human vaccines since the 1990s. QS-21, a mixture of saponin compounds, was included in the AS01-adjuvanted Shingrix vaccine. Here, we investigated the adjuvant effects of VSA-1, a newly developed semisynthetic analog of QS-21, on promoting protection in mice after vaccination with the inactivated split virus vaccine. The adjuvant effects of VSA-1 on improving vaccine efficacy after prime immunization were evident as shown by significantly higher levels of hemagglutination-inhibiting antibody titers and enhanced homologous protection compared to those by QS-21 and Alum adjuvants. The adjuvant effects of VSA-1 on enhancing heterosubtypic protection after two doses of adjuvanted vaccination were comparable to those of QS-21. T cell immunity played an important role in conferring cross-protection by VSA-1-adjuvanted vaccination. Overall, the findings in this study suggest that VSA-1 exhibits desirable adjuvant properties and a unique pattern of innate and adaptive immune responses, contributing to improved homologous and heterosubtypic protection by inactivated split influenza vaccination in mice.

6.
Virology ; 574: 37-46, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35914365

RESUMEN

To improve cross-protection of influenza vaccination, we tested conjugation of conserved M2e epitopes to the surface of inactivated influenza virus (iPR8-M2e*). Treatment of virus with chemical cross-linker led to diminished hemagglutination activity and failure to induce hemagglutination inhibiting antibodies. Conjugated iPR8-M2e* vaccine was less protective against homologous and heterosubtypic viruses, despite the induction of virus-specific binding IgG antibodies. In alternative approaches to enhance cross-protection, we developed a genetically linked chimeric protein (M2e-B stalk) vaccine with M2e of influenza A and hemagglutinin (HA) stalk of influenza B virus. Vaccination of mice with inactivated influenza A virus supplemented with M2e-B stalk effectively induced hemagglutination inhibiting antibodies, humoral and cellular M2e immune responses, and enhanced heterosubtypic protection. This study demonstrates the importance of HA functional integrity in influenza vaccine efficacy and that supplementation of influenza vaccines with M2e-B stalk protein could be a feasible strategy of improving cross-protection against influenza viruses.


Asunto(s)
Vacunas contra la Influenza , Gripe Humana , Infecciones por Orthomyxoviridae , Orthomyxoviridae , Animales , Anticuerpos Antivirales , Protección Cruzada , Hemaglutinación , Humanos , Gripe Humana/prevención & control , Ratones , Ratones Endogámicos BALB C
7.
PLoS Pathog ; 18(8): e1010755, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-36006890

RESUMEN

Annual influenza vaccination is recommended to update the variable hemagglutinin antigens. Here, we first designed a virus-like particle (VLP) displaying consensus multi-neuraminidase (NA) subtypes (cN1, cN2, B cNA) and M2 ectodomain (M2e) tandem repeat (m-cNA-M2e VLP). Vaccination of mice with m-cNA-M2e VLP induced broad NA inhibition (NAI), and M2e antibodies as well as interferon-gamma secreting T cell responses. Mice vaccinated with m-cNA-M2e VLP were protected against influenza A (H1N1, H5N1, H3N2, H9N2, H7N9) and influenza B (Yamagata and Victoria lineage) viruses containing substantial antigenic variations. Protective immune contributors include cellular and humoral immunity as well as antibody-dependent cellular cytotoxicity. Furthermore, comparable cross protection by m-cNA-M2e VLP vaccination was induced in aged mice. This study supports a novel strategy of developing a universal vaccine against influenza A and B viruses potentially in both young and aged populations by inducing multi-NA subtype and M2e immunity with a single VLP entity.


Asunto(s)
Vacunas contra la Influenza , Gripe Humana , Infecciones por Orthomyxoviridae , Animales , Anticuerpos Antivirales , Humanos , Virus de la Influenza A/clasificación , Vacunas contra la Influenza/inmunología , Gripe Humana/prevención & control , Ratones , Ratones Endogámicos BALB C , Infecciones por Orthomyxoviridae/prevención & control , Proteínas de la Matriz Viral/genética
8.
NPJ Vaccines ; 7(1): 68, 2022 Jun 29.
Artículo en Inglés | MEDLINE | ID: mdl-35768475

RESUMEN

We developed a new chimeric M2e and H3 hemagglutinin (HA) stalk protein vaccine (M2e-H3 stalk) by genetic engineering of modified H3 stalk domain conjugated with conserved M2e epitopes to overcome the drawbacks of low efficacy by monomeric domain-based universal vaccines. M2e-H3 stalk protein expressed and purified from Escherichia coli was thermostable, displaying native-like antigenic epitopes recognized by antisera of different HA subtype proteins and influenza A virus infections. Adjuvanted M2e-H3 stalk vaccination induced M2e and stalk-specific IgG antibodies recognizing viral antigens on virus particles and on the infected cell surface, CD4+ and CD8+ T-cell responses, and antibody-dependent cytotoxic cell surrogate activity in mice. M2e-H3 stalk was found to confer protection against heterologous and heterosubtypic cross-group subtype viruses (H1N1, H5N1, H9N2, H3N2, H7N9) at similar levels in adult and aged mice. These results provide evidence that M2e-H3 stalk chimeric proteins can be developed as a universal influenza A virus vaccine candidate for young and aged populations.

9.
Mol Ther Methods Clin Dev ; 26: 38-51, 2022 Sep 08.
Artículo en Inglés | MEDLINE | ID: mdl-35755946

RESUMEN

Hemagglutinin (HA) stem-based vaccines have limitations in providing broad and effective protection against cross-group influenza viruses, despite being a promising universal vaccine target. To overcome the limited cross-protection and low efficacy by HA stem vaccination, we genetically engineered a chimeric conjugate of thermostable H1 HA stem and highly conserved M2e repeat (M2e-H1stem), which was expressed at high yields in Escherichia coli. M2e-H1stem protein presented native-like epitopes reactive to antisera of live virus infection. M2e-H1stem protein vaccination of mice induced strong M2e- and HA stem-specific immune responses, conferring broadly effective cross-protection against both antigenically distinct group 1 (H1N1, H5N1, and H9N2 subtypes) and group 2 (H3N2 and H7N9 subtypes) seasonal and pandemic potential influenza viruses. M2e-H1stem vaccination generated CD4+ and CD8+ T cell responses and antibody-dependent cytotoxic cellular and humoral immunity, which contributed to enhancing cross-protection. Furthermore, comparable broad cross-group protection was observed in older aged mice after M2e-H1stem vaccination. This study provides evidence warranting further development of chimeric M2e-stem proteins as a promising universal influenza vaccine candidate in adult and aged populations.

10.
FASEB J ; 36(3): e22182, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-35113455

RESUMEN

Pre-pandemic influenza H5N1 vaccine has relatively low immunogenicity and often requires high antigen amounts and two immunizations to induce protective immunity. Incorporation of vaccine adjuvants is promising to stretch vaccine doses during pandemic outbreaks. This study presents a physical radiofrequency (RF) adjuvant (RFA) to conveniently and effectively increase the immunogenicity and efficacy of H5N1 vaccine without modification of vaccine preparation. Physical RFA is based on a brief RF treatment of the skin to induce thermal stress to enhance intradermal vaccine-induced immune responses with minimal local or systemic adverse reactions. We found that physical RFA could significantly increase H5N1 vaccine-induced hemagglutination inhibition antibody titers in murine models. Intradermal H5N1 vaccine in the presence of RFA but not vaccine alone significantly lowered lung viral titers, reduced body weight loss, and improved survival rates after lethal viral challenges. The improved protection in the presence of RFA was correlated with enhanced humoral and cellular immune responses to H5N1 vaccination in both male and female mice, indicating no gender difference of RFA effects in murine models. Our data support further development of the physical RFA to conveniently enhance the efficacy of H5N1 vaccine.


Asunto(s)
Inmunidad Celular/inmunología , Inmunidad Humoral/inmunología , Subtipo H5N1 del Virus de la Influenza A/inmunología , Vacunas contra la Influenza/inmunología , Infecciones por Orthomyxoviridae/inmunología , Adyuvantes Inmunológicos/farmacología , Animales , Anticuerpos Antivirales/inmunología , Femenino , Pruebas de Inhibición de Hemaglutinación/métodos , Pulmón/inmunología , Pulmón/virología , Masculino , Ratones , Ratones Endogámicos C57BL , Pandemias/prevención & control , Vacunación/métodos
11.
Membranes (Basel) ; 12(1)2022 Jan 17.
Artículo en Inglés | MEDLINE | ID: mdl-35054623

RESUMEN

A general finite element model and a new solution method were developed to simulate the permeances of Lintz Donawiz converter gas (LDG) components and the performance of a polysulfone membrane separation unit. The permeances at eight bars of CO, N2, and H2 in LDG simulated using the developed model equations employing the experimental mixed gas data were obtained by controlling the finite element numbers and comparing them with pure gas permeation data. At the optimal finite element numbers (s = 15, n = 1), the gas permeances under the mixed-gas condition were 6.3% (CO), 3.9% (N2), and 7.2% (H2) larger than those of the pure gases, On the other hand, the mixed-gas permeance of CO2 was 4.5% smaller than that of pure gas. These differences were attributed to the plasticization phenomenon of the polysulfone membrane used by CO2. The newly adopted solution method for the stiff nonlinear model functions enabled the simulation of the performance (in terms of gas recovery, concentration, and flow rate) of the first-stage membrane within two seconds under most gas flow conditions. The performance of a first-stage membrane unit separating LDG could be predicted by the developed model with a small error of <2.1%. These model and solution methods could be utilized effectively for simulating gas permeances of the membrane that is plasticized severely by the permeating gas and the separation performance of two- or multi-stage membrane processes.

12.
Antiviral Res ; 197: 105229, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34933043

RESUMEN

Vaccination against influenza viruses suffers from low efficacy in conferring homologous and cross-protection, particularly in older adults. Here, we compared the effects of three different adjuvant types (QS-21+MPL, CpG+MPL and bacterial cell wall CWS) on enhancing the immunogenicity and homologous and heterosubtypic protection of influenza vaccination in young adult and aged mouse models. A combination of saponin QS-21 and monophosphoryl lipid A (QS-21+MPL) was most effective in inducing T helper type 1 (Th1) T cell and cross-reactive IgG as well as hemagglutination inhibiting antibody responses to influenza vaccination. Both combination adjuvants (QS-21+MPL and CpG+MPL) exhibited high potency by preventing weight loss and reducing viral loads and enhanced homologous and cross-protection by influenza vaccination in adult and aged mouse models. Bacillus Calmette-Guerin cell-wall skeleton (CWS) displayed substantial adjuvant effects on immune responses to influenza vaccination but lower adjuvant efficacy in inducing Th1 IgG responses, cross-protection in adult mice, and in conferring homologous protection in aged mice. This study has significance in comparing the effects of potent adjuvants on enhancing humoral and cellular immune responses to influenza virus vaccination, inducing homologous and cross-protection in adult and aged populations.


Asunto(s)
Adyuvantes Inmunológicos/administración & dosificación , Adyuvantes Inmunológicos/normas , Anticuerpos Antivirales/sangre , Protección Cruzada/inmunología , Inmunogenicidad Vacunal , Vacunas contra la Influenza/inmunología , Adyuvantes Inmunológicos/clasificación , Factores de Edad , Animales , Femenino , Humanos , Inmunidad Celular , Vacunas contra la Influenza/administración & dosificación , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Infecciones por Orthomyxoviridae/inmunología , Infecciones por Orthomyxoviridae/prevención & control , Células TH1/inmunología , Células Th2/inmunología , Vacunación
13.
Virology ; 566: 143-152, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34929590

RESUMEN

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.


Asunto(s)
Envejecimiento/inmunología , Anticuerpos Antivirales/biosíntesis , Glicoproteínas Hemaglutininas del Virus de la Influenza/genética , Inmunoglobulina G/biosíntesis , Vacunas contra la Influenza/genética , Gripe Humana/prevención & control , Infecciones por Orthomyxoviridae/prevención & control , Factores de Edad , Envejecimiento/genética , Animales , Antígenos Virales/genética , Antígenos Virales/inmunología , Protección Cruzada , Femenino , Glicoproteínas Hemaglutininas del Virus de la Influenza/inmunología , Humanos , Inmunización Secundaria/métodos , Inmunogenicidad Vacunal , Subtipo H1N1 del Virus de la Influenza A/inmunología , Subtipo H1N1 del Virus de la Influenza A/patogenicidad , Subtipo H3N2 del Virus de la Influenza A/inmunología , Subtipo H3N2 del Virus de la Influenza A/patogenicidad , Vacunas contra la Influenza/administración & dosificación , Vacunas contra la Influenza/biosíntesis , Gripe Humana/inmunología , Gripe Humana/virología , Ratones , Ratones Endogámicos C57BL , Modelos Moleculares , Infecciones por Orthomyxoviridae/inmunología , Infecciones por Orthomyxoviridae/virología , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/inmunología , Vacunación/métodos , Vacunas Sintéticas , Proteínas de la Matriz Viral/genética , Proteínas de la Matriz Viral/inmunología
14.
Vaccines (Basel) ; 9(4)2021 Mar 29.
Artículo en Inglés | MEDLINE | ID: mdl-33805473

RESUMEN

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) continues to be expanding the pandemic disease across the globe. Although SARS-CoV-2 vaccines were rapidly developed and approved for emergency use of vaccination in humans, supply and production difficulties are slowing down the global vaccination program. The efficacy of many different versions of vaccine candidates and adjuvant effects remain unknown, particularly in the elderly. In this study, we compared the immunogenic properties of SARS-CoV-2 full-length spike (S) ectodomain in young adult and aged mice, S1 with receptor binding domain, and S2 with fusion domain. Full-length S was more immunogenic and effective in inducing IgG antibodies after low dose vaccination, compared to the S1 subunit. Old-aged mice induced SARS-CoV-2 spike-specific IgG antibodies with neutralizing activity after high dose S vaccination. With an increased vaccine dose, S1 was highly effective in inducing neutralizing and receptor-binding inhibiting antibodies, although both S1 and S2 subunit domain vaccines were similarly immunogenic. Adjuvant effects were significant for effective induction of IgG1 and IgG2a isotypes, neutralizing and receptor-binding inhibiting antibodies, and antibody-secreting B cell and interferon-γ secreting T cell immune responses. Results of this study provide information in designing SARS-CoV-2 spike vaccine antigens and effective vaccination in the elderly.

15.
Sci Rep ; 11(1): 4151, 2021 02 18.
Artículo en Inglés | MEDLINE | ID: mdl-33603072

RESUMEN

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.


Asunto(s)
Protección Cruzada/inmunología , Hemaglutininas/inmunología , Subtipo H3N2 del Virus de la Influenza A/inmunología , Vacunas contra la Influenza/inmunología , Gripe Humana/inmunología , Infecciones por Orthomyxoviridae/inmunología , Proteínas Recombinantes de Fusión/inmunología , Animales , Anticuerpos Antivirales/inmunología , Antígenos Virales/inmunología , Células HEK293 , Humanos , Inmunoglobulina G/inmunología , Ratones , Ratones Endogámicos BALB C , Vacunación/métodos
16.
Vaccines (Basel) ; 8(4)2020 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-33271920

RESUMEN

Respiratory syncytial virus (RSV) is one of the most important pathogens causing significant morbidity and mortality in infants and the elderly. Live attenuated influenza vaccine (LAIV) is a licensed vaccine platform in humans and it is known to induce broader immune responses. RSV G attachment proteins mediate virus binding to the target cells and they contain a conserved central domain with neutralizing epitopes. Here, we generated recombinant LAIV based on the attenuated A/Puerto Rico/8/1934 virus backbone, expressing an RSV conserved G-domain in a chimeric hemagglutinin (HA) fusion molecule (HA-G). The attenuated phenotypes of chimeric HA-G LAIV were evident by restricted replication in the upper respiratory tract and low temperature growth characteristics. The immunization of mice with chimeric HA-G LAIV induced significant increases in G-protein specific IgG2a (T helper type 1) and IgG antibody-secreting cell responses in lung, bronchioalveolar fluid, bone marrow, and spleens after RSV challenge. Vaccine-enhanced disease that is typically caused by inactivated-RSV vaccination was not observed in chimeric HA-G LAIV as analyzed by lung histopathology. These results in this study suggest a new approach of developing an RSV vaccine candidate while using recombinant LAIV, potentially conferring protection against influenza virus and RSV.

17.
Vaccines (Basel) ; 8(4)2020 Nov 03.
Artículo en Inglés | MEDLINE | ID: mdl-33153089

RESUMEN

Influenza viruses constantly evolve, reducing the overall protective effect of routine vaccination campaigns. Many different strategies are being explored to design universal influenza vaccines capable of protecting against evolutionary diverged viruses. The ectodomain of influenza A M2e protein (M2e) is among the most promising targets for universal vaccine design. Here, we generated two recombinant live attenuated influenza vaccines (LAIVs) expressing additional four M2e tandem repeats (4M2e) from the N-terminus of the viral hemagglutinin (HA) protein, in an attempt to enhance the M2e-mediated cross-protection. The recombinant H1N1+4M2e and H3N2+4M2e viruses retained growth characteristics attributable to traditional LAIV viruses and induced robust influenza-specific antibody responses in BALB/c mice, although M2e-specific antibodies were raised only after two-dose vaccination with LAIV+4M2e viruses. Mice immunized with either LAIV or LAIV+4M2e viruses were fully protected against a panel of heterologous influenza challenge viruses suggesting that antibody and cell-mediated immunity contributed to the protection. The protective role of the M2e-specific antibody was seen in passive serum transfer experiments, where enhancement in the survival rates between classical LAIV and chimeric H3N2+4M2e LAIV was demonstrated for H3N2 and H5N1 heterologous challenge viruses. Overall, the results of our study suggest that M2e-specific antibodies induced by recombinant LAIV+4M2e in addition to cellular immunity by LAIV play an important role in conferring protection against heterologous viruses.

18.
Virology ; 550: 51-60, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32882637

RESUMEN

Influenza virus neuraminidase (NA) contains a universally conserved epitope (NAe, NA222-230). However, no studies have reported vaccines targeting this NA conserved epitope and inducing antibodies recognizing NAe. The extracellular domain of M2 (M2e) is considered as an attractive target for a universal influenza vaccine. We generated recombinant influenza H1N1 viruses expressing conserved epitopes in hemagglutinin (HA) molecules: NAe (NAe-HA) or M2e (M2e-HA) within the HA head domain. Inactivated recombinant NAe-HA and M2e-HA viruses were more effective in inducing IgG antibodies specific for an inserted conserved epitope than live recombinant virus. Recombinant inactivated M2e-HA virus vaccination induced cross protection against H3N2 virus with less weight loss compared to NAe-HA and was more effective in inducing humoral and cellular M2e immune responses. This study provides insight into developing recombinant influenza virus vaccines compatible with current platforms to induce antibody responses to conserved poorly immunogenic epitopes.


Asunto(s)
Glicoproteínas Hemaglutininas del Virus de la Influenza/inmunología , Subtipo H1N1 del Virus de la Influenza A/inmunología , Vacunas contra la Influenza/biosíntesis , Neuraminidasa/inmunología , Infecciones por Orthomyxoviridae/prevención & control , Virus Reordenados/inmunología , Proteínas de la Matriz Viral/inmunología , Animales , Anticuerpos Antivirales/biosíntesis , Protección Cruzada , Epítopos/química , Epítopos/inmunología , Femenino , Glicoproteínas Hemaglutininas del Virus de la Influenza/genética , Humanos , Inmunidad Celular/efectos de los fármacos , Inmunidad Humoral/efectos de los fármacos , Subtipo H1N1 del Virus de la Influenza A/efectos de los fármacos , Subtipo H1N1 del Virus de la Influenza A/genética , Subtipo H3N2 del Virus de la Influenza A/efectos de los fármacos , Subtipo H3N2 del Virus de la Influenza A/genética , Subtipo H3N2 del Virus de la Influenza A/inmunología , Subtipo H5N1 del Virus de la Influenza A/efectos de los fármacos , Subtipo H5N1 del Virus de la Influenza A/genética , Subtipo H5N1 del Virus de la Influenza A/inmunología , Subtipo H9N2 del Virus de la Influenza A/efectos de los fármacos , Subtipo H9N2 del Virus de la Influenza A/genética , Subtipo H9N2 del Virus de la Influenza A/inmunología , Vacunas contra la Influenza/administración & dosificación , Vacunas contra la Influenza/genética , Ratones , Ratones Endogámicos BALB C , Neuraminidasa/genética , Infecciones por Orthomyxoviridae/inmunología , Infecciones por Orthomyxoviridae/virología , Virus Reordenados/efectos de los fármacos , Virus Reordenados/genética , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/inmunología , Vacunación/métodos , Vacunas de Productos Inactivados , Proteínas de la Matriz Viral/genética
19.
Vaccine ; 38(36): 5783-5792, 2020 08 10.
Artículo en Inglés | MEDLINE | ID: mdl-32674907

RESUMEN

Lactic acid bacteria Lactobacillus casei DK128 isolated from fermented vegetable foods was suggested to stimulate innate immune responses. Here, we investigated whether heat-killed DK128 would exhibit adjuvant effects on enhancing the efficacy of influenza vaccination. Immunization of mice with split influenza virus vaccine in the presence of heat-killed DK128 induced significantly higher levels of both IgG1 and IgG2c isotype antibodies than those by vaccine only. A single dose DK128-adjuvanted influenza vaccination conferred higher efficacy of protection, as evidenced by intact lung function, less weight loss, enhanced clearance of lung viral loads, and lower levels of inflammatory cytokines and infiltrates. Immunization of CD4 T cell-knockout (CD4KO) mice with influenza vaccine and DK128, but not with vaccine alone, induced isotype-switched IgG antibodies and protection against lethal challenge in CD4KO mice. The results in this study suggest heat-killed DK128 as a potential vaccine adjuvant, promoting the induction of IgG isotype switching in CD4-deficient condition and enhancing protective efficacy of split influenza vaccination in immunocompromised and immune-competent subjects.


Asunto(s)
Vacunas contra la Influenza , Gripe Humana , Lacticaseibacillus casei , Infecciones por Orthomyxoviridae , Adyuvantes Inmunológicos , Animales , Anticuerpos Antivirales , Inmunidad , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Infecciones por Orthomyxoviridae/prevención & control , Vacunación
20.
PLoS One ; 14(6): e0216871, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31246961

RESUMEN

Despite the ability to induce a broad range of cross protection, M2e5x virus-like particles (VLPs) alone provide limited vaccine efficacy and confer low efficacy of protection against highly pathogenic avian influenza virus (HPAIV) infection in chickens. Avian influenza hemagglutinin (HA) has been a major antigenic target that enhances humoral immunity, but the efficacy of avian HA-based vaccines against HPAIV needs to be further improved. In this study, we evaluated the vaccine efficacy induced by combination of conserved tandem repeat M2e5x VLPs and HA VLPs against an avian influenza virus. We found that combinatorial vaccine elicited higher levels of reassortant H5N1 (rgH5N1) virus-specific IgG, IgG1, IgG2a and IgA antibody responses compared to M2e5x VLPs in sera and lungs of mice. Combinatorial VLPs vaccination induced higher levels of CD8+ T cell and germinal center B cell responses in lung and spleen compared to M2e5x VLPs. Combinatorial VLPs vaccination showed significantly reduced inflammatory responses and lung viral loads upon rgH5N1 virus challenge infection, resulting in less body weight loss compared to M2e5x VLPs alone. These results indicate that immune responses to both M2e and HA might provide a strategy of vaccination inducing enhanced protection against avian influenza virus.


Asunto(s)
Ingeniería Genética , Subtipo H5N1 del Virus de la Influenza A/inmunología , Vacunas de Partículas Similares a Virus/genética , Vacunas de Partículas Similares a Virus/inmunología , Proteínas de la Matriz Viral/genética , Animales , Anticuerpos Antivirales/sangre , Anticuerpos Antivirales/inmunología , Inmunización , Inmunoglobulinas/sangre , Inmunoglobulinas/inmunología , Pulmón/inmunología , Ratones , Células Sf9 , Spodoptera
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...