Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 79
Filtrar
1.
Antiviral Res ; 220: 105752, 2023 12.
Artículo en Inglés | MEDLINE | ID: mdl-37949318

RESUMEN

The outbreak of SARS-CoV-2 infections had led to the COVID-19 pandemic which has a significant impact on global public health and the economy. The spike (S) protein of SARS-CoV-2 contains the receptor binding domain (RBD) which binds to human angiotensin-converting enzyme 2 receptor. Numerous RBD-based vaccines have been developed and recently focused on the induction of neutralizing antibodies against the immune evasive Omicron BQ.1.1 and XBB.1.5 subvariants. In this preclinical study, we reported the use of a direct fusion of the type IIb Escherichia coli heat-labile enterotoxin A subunit with SARS CoV-2 RBD protein (RBD-LTA) as an intranasal vaccine candidate. The results showed that intranasal immunization with the RBD-LTA fusion protein in BALB/c mice elicited potent neutralizing antibodies against the Wuhan-Hu-1 and several SARS-CoV-2 variants as well as the production of IgA antibodies in bronchoalveolar lavage fluids (BALFs). Furthermore, the heterologous RBD representing the same strains used in the bivalent mRNA vaccine were used as a second-dose RBD-LTA/RBD protein booster after bivalent mRNA vaccination. The results showed that the neutralizing antibody titers elicited by the intranasal bivalent RBD-LTA/RBD protein booster were similar to the intramuscular bivalent mRNA booster, but the RBD-specific IgA titers in sera and BALFs significantly increased. Overall, this preclinical study suggests that the RBD-LTA fusion protein could be a promising candidate as a mucosal booster COVID-19 vaccine.


Asunto(s)
COVID-19 , Glicoproteína de la Espiga del Coronavirus , Animales , Ratones , Humanos , Glicoproteína de la Espiga del Coronavirus/genética , Escherichia coli , Vacunas contra la COVID-19 , Calor , Pandemias , COVID-19/prevención & control , SARS-CoV-2/genética , Enterotoxinas/genética , Vacunación , Inmunización , Anticuerpos Neutralizantes , ARN Mensajero , Anticuerpos Antivirales
2.
J Med Virol ; 95(8): e29040, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37635380

RESUMEN

Protein subunit vaccines have been used as prophylactic vaccines for a long time. The well-established properties of these vaccines make them the first choice for the coronavirus disease 2019 (COVID-19) outbreak. However, it is not easy to develop a protein vaccine that induces cytotoxic T lymphocyte responses and requires a longer time for manufacturing, which limits the usage of this vaccine type. Here, we report the combination of a recombinant spike (S)-trimer protein with a DNA vaccine-encoded S protein as a novel COVID-19 vaccine. The recombinant S protein was formulated with different adjuvants and mixed with the DNA plasmid before injection. We found that the recombinant S protein formulated with the adjuvant aluminum hydroxide and mixed with the DNA plasmid could enhance antigen-specific antibody titers, neutralizing antibody titers. We further evaluated the IgG2a/IgG1 isotype and cytokine profiles of the specific boosted T-cell response, which indicated that the combined vaccine induced a T-helper 1 cell-biased immune response. Immunized hamsters were challenged with severe acute respiratory syndrome coronavirus 2, and the body weight of the hamsters that received the recombinant S protein with aluminum hydroxide and/or the DNA plasmid was not reduced. Alternatively, those that received control or only the DNA plasmid immunization were reduced. Interestingly, after the third day of the viral load in the lungs, the viral challenge could not be detected in hamsters immunized with the recombinant S protein in aluminum hydroxide mixed with DNA (tissue culture infectious dose < 10). The viral load in the lungs was 109 , 106 , and 107 for the phosphate-buffered saline, protein in aluminum hydroxide, and DNA-only immunizations, respectively. These results indicated that antiviral mechanisms neutralizing antibodies play important roles. Furthermore, we found that the combination of protein and DNA vaccination could induce relatively strong CD8+ T-cell responses. In summary, the protein subunit vaccine combined with a DNA vaccine could induce strong CD8+ T-cell responses to increase antiviral immunity for disease control.


Asunto(s)
COVID-19 , Vacunas de ADN , Humanos , Animales , Cricetinae , SARS-CoV-2/genética , Hidróxido de Aluminio , Vacunas contra la COVID-19 , Subunidades de Proteína , COVID-19/prevención & control , ADN , Inmunidad Celular , Adyuvantes Inmunológicos , Adyuvantes Farmacéuticos , Antivirales
3.
Sci Rep ; 13(1): 13468, 2023 08 18.
Artículo en Inglés | MEDLINE | ID: mdl-37596329

RESUMEN

The COVID-19 pandemic has had a widespread impact on a global scale, and the evolution of considerable dominants has already taken place. Some variants contained certain key mutations located on the receptor binding domain (RBD) of spike protein, such as E484K and N501Y. It is increasingly worrying that these variants could impair the efficacy of current vaccines or therapies. Therefore, analyzing and predicting the high-risk mutations of SARS-CoV-2 spike glycoprotein is crucial to design future vaccines against the different variants. In this work, we proposed an in silico approach, immune-escaping score (IES), to predict high-risk immune-escaping hot spots on the receptor-binding domain (RBD), implemented through integrated delta binding free energy measured by computational mutagenesis of spike-antibody complexes and mutation frequency calculated from viral genome sequencing data. We identified 23 potentially immune-escaping mutations on the RBD by using IES, nine of which occurred in omicron variants (R346K, K417N, N440K, L452Q, L452R, S477N, T478K, F490S, and N501Y), despite our dataset being curated before the omicron first appeared. The highest immune-escaping score (IES = 1) was found for E484K, which agrees with recent studies stating that the mutation significantly reduced the efficacy of neutralization antibodies. Furthermore, our predicted delta binding free energy and IES show a high correlation with high-throughput deep mutational scanning data (Pearson's r = 0.70) and experimentally measured neutralization titers data (mean Pearson's r = -0.80). In summary, our work presents a new method to identify the potentially immune-escaping mutations on the RBD and provides valuable insights into future COVID-19 vaccine design.


Asunto(s)
COVID-19 , Dermatitis , Humanos , Vacunas contra la COVID-19/genética , COVID-19/prevención & control , Pandemias , SARS-CoV-2/genética
4.
Virus Res ; 329: 199101, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-36958398

RESUMEN

Coxsackievirus A10 (CVA10) is one of enteroviral pathogens that cause the hand, foot, and mouth disease (HFMD). Since CVA10 was reported to be not easily propagated in the Vero cell culture, a feasible manufacture process for producing formalin-inactivated CVA10 vaccine is urgently needed. Several cell lines that commonly used for viral vaccine production was tested for CVA10 (M2014 strain) culture in this study, and our result showed that CVA10 could be easily propagated in the HEK293A cells. A serum-free HEK293A cell culture system was developed for CVA10 production and the yields have reached over 108 TCID50/mL. The biochemical and immunogenic properties of CVA10 particles obtained from this serum-free HEK293A culture were identical to our previous study. Two major particles of CVA10 were separated by ultracentrifugation, and only the infectious mature particles were capable of inducing CVA10 neutralizing antibody responses in the mouse immunogenicity studies. Additionally, we found that coxsackievirus A6 and enterovirus A71 could also be easily propagated using this serum-free HEK293A cell culture system. Our results provide a solution to overcome the obstacle in the propagation of CVA10 and facilitate the development of multivalent vaccines for prevention of HFMD.


Asunto(s)
Enterovirus Humano A , Enterovirus , Enfermedad de Boca, Mano y Pie , Animales , Ratones , Enfermedad de Boca, Mano y Pie/prevención & control , Anticuerpos Neutralizantes , Anticuerpos Antivirales , Vacunas de Productos Inactivados , Enterovirus Humano A/genética
5.
Viruses ; 14(6)2022 06 14.
Artículo en Inglés | MEDLINE | ID: mdl-35746775

RESUMEN

Hemagglutinin (HA) and neuraminidase (NA) are the two major envelope proteins of influenza viruses. The spatial organization of HA and NA on the virus surface needs to be optimized to promote viral fitness, host specificity, transmissibility, infectivity, and virulence. We previously demonstrated that the recombinant NA protein of the 2009 pandemic H1N1 (pH1N1) with the I365T/S366N mutation in the NA 370-loop elicited higher NA-inhibition antibody titers against the homologous pH1N1 virus and three heterologous H5N1, H3N2, and H7N9 viruses in mice. In this study, we used PR8-based reverse genetics (RG) by replacing the HA and NA genes of A/Texas/05/2009 pH1N1 virus to obtain the wild-type pH1N1 and three NA 370-loop mutant viruses of pH1N1 (I365T/S366N), RG pH1N1 (I365E/S366D), and RG pH1N1 (I365T/S366A). Our results revealed that the viral NA enzyme activity increased for the RG pH1N1(I365T/S366N) and RG pH1N1 (I365E/S366D) viruses but reduced for the RG pH1N1 (I365T/S366A) virus. The increased or decreased NA enzyme activity was found to correlate with the increase or decrease in HA titers of these NA 370-loop mutant viruses. All of these three NA 370-loop mutant RG pH1N1 viruses were less virulent than the wild-type RG pH1N1 virus in mice. Immunizations with the inactivated viruses carrying the three NA 370-loop mutations and the wild-type RG pH1N1 virus were found to elicit approximately the same titers of NA-inhibition antibodies against H1N1 and H5N1 viruses. These results may provide information for developing NA-based influenza virus vaccines.


Asunto(s)
Subtipo H1N1 del Virus de la Influenza A , Subtipo H5N1 del Virus de la Influenza A , Subtipo H7N9 del Virus de la Influenza A , Vacunas contra la Influenza , Infecciones por Orthomyxoviridae , Animales , Anticuerpos Antivirales , Antígenos Virales , Hemaglutinación , Hemaglutininas , Subtipo H3N2 del Virus de la Influenza A/genética , Subtipo H7N9 del Virus de la Influenza A/genética , Ratones , Mutación , Neuraminidasa , Virulencia/genética
6.
Pharmaceutics ; 14(5)2022 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-35631599

RESUMEN

Zika virus (ZIKV) infections in humans are mainly transmitted by the mosquito vectors, but human-to-human sexual transmission is also another important route. Developing a ZIKV mucosal vaccine that can elicit both systemic and mucosal immune responses is of particular interest. In this study, we constructed a recombinant ZIKV envelope DIII (ZDIII) protein genetically fused with Salmonella typhimurium flagellin (FliC-ZDIII) as a novel mucosal antigen for intranasal immunization. The results indicated that the FliC-ZDIII fusion proteins formulated with E. coli heat-labile enterotoxin B subunit (LTIIb-B5) adjuvant greatly increased the ZDIII-specific IgG, IgA, and neutralizing titers in sera, and the ZDIII-specific IgA titers in bronchoalveolar lavage and vaginal fluids. Protective immunity was further assessed by subcutaneous and intravaginal ZIKV challenges. The second-generation FliCΔD3-2ZDIII was shown to result in a reduced titer of anti-FliC IgG antibodies in sera and still retained the same levels of serum IgG, IgA, and neutralizing antibodies and mucosal IgA antibodies without compromising the vaccine antigenicity. Therefore, intranasal immunization with FliCΔD3-2ZDIII fusion proteins formulated with LTIIb-B5 adjuvant elicited the greatest protective immunity against subcutaneous and intravaginal ZIKV challenges. Our findings indicated that the combination of FliCΔD3-2ZDIII fusion proteins and LTIIb-B5 adjuvant for intranasal immunization can be used for developing ZIKV mucosal vaccines.

7.
Int J Mol Sci ; 23(9)2022 Apr 28.
Artículo en Inglés | MEDLINE | ID: mdl-35563292

RESUMEN

During the sustained COVID-19 pandemic, global mass vaccination to achieve herd immunity can prevent further viral spread and mutation. A protein subunit vaccine that is safe, effective, stable, has few storage restrictions, and involves a liable manufacturing process would be advantageous to distribute around the world. Here, we designed and produced a recombinant spike (S)-Trimer that is maintained in a prefusion state and exhibits a high ACE2 binding affinity. Rodents received different doses of S-Trimer (0.5, 5, or 20 µg) antigen formulated with aluminum hydroxide (Alum) or an emulsion-type adjuvant (SWE), or no adjuvant. After two vaccinations, the antibody response, T-cell responses, and number of follicular helper T-cells (Tfh) or germinal center (GC) B cells were assessed in mice; the protective efficacy was evaluated on a Syrian hamster infection model. The mouse studies demonstrated that adjuvating the S-Trimer with SWE induced a potent humoral immune response and Th1-biased cellular immune responses (in low dose) that were superior to those induced by Alum. In the Syrian hamster studies, when S-Trimer was adjuvanted with SWE, higher levels of neutralizing antibodies were induced against live SARS-CoV-2 from the original lineage and against the emergence of variants (Beta or Delta) with a slightly decreased potency. In addition, the SWE adjuvant demonstrated a dose-sparing effect; thus, a lower dose of S-Trimer as an antigen (0.5 µg) can induce comparable antisera and provide complete protection from viral infection. These data support the utility of SWE as an adjuvant to enhance the immunogenicity of the S-Trimer vaccine, which is feasible for further clinical testing.


Asunto(s)
Vacunas contra la COVID-19 , COVID-19 , SARS-CoV-2 , Glicoproteína de la Espiga del Coronavirus , Células TH1 , Adyuvantes Inmunológicos/farmacología , Adyuvantes Farmacéuticos , Animales , Anticuerpos Neutralizantes , Anticuerpos Antivirales , COVID-19/prevención & control , Vacunas contra la COVID-19/farmacología , Cricetinae , Emulsiones , Humanos , Ratones , Glicoproteína de la Espiga del Coronavirus/química , Glicoproteína de la Espiga del Coronavirus/inmunología , Células TH1/inmunología
8.
Front Immunol ; 12: 795741, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34925381

RESUMEN

Glycan-masking the vaccine antigen by mutating the undesired antigenic sites with an additional N-linked glycosylation motif can refocus B-cell responses to desired epitopes, without affecting the antigen's overall-folded structure. This study examined the impact of glycan-masking mutants of the N-terminal domain (NTD) and receptor-binding domain (RBD) of SARS-CoV-2, and found that the antigenic design of the S protein increases the neutralizing antibody titers against the Wuhan-Hu-1 ancestral strain and the recently emerged SARS-CoV-2 variants Alpha (B.1.1.7), Beta (B.1.351), and Delta (B.1.617.2). Our results demonstrated that the use of glycan-masking Ad-S-R158N/Y160T in the NTD elicited a 2.8-fold, 6.5-fold, and 4.6-fold increase in the IC-50 NT titer against the Alpha (B.1.1.7), Beta (B.1.351) and Delta (B.1.617.2) variants, respectively. Glycan-masking of Ad-S-D428N in the RBD resulted in a 3.0-fold and 2.0-fold increase in the IC-50 neutralization titer against the Alpha (B.1.1.7) and Beta (B.1.351) variants, respectively. The use of glycan-masking in Ad-S-R158N/Y160T and Ad-S-D428N antigen design may help develop universal COVID-19 vaccines against current and future emerging SARS-CoV-2 variants.


Asunto(s)
Antígenos Virales/inmunología , COVID-19/inmunología , Epítopos/inmunología , Dominios y Motivos de Interacción de Proteínas/inmunología , SARS-CoV-2/inmunología , Glicoproteína de la Espiga del Coronavirus/inmunología , Adenoviridae/genética , Animales , Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/inmunología , Formación de Anticuerpos/inmunología , COVID-19/prevención & control , COVID-19/virología , Vacunas contra la COVID-19/genética , Vacunas contra la COVID-19/inmunología , Modelos Animales de Enfermedad , Relación Dosis-Respuesta Inmunológica , Femenino , Ingeniería Genética , Vectores Genéticos/genética , Humanos , Inmunización , Ratones , Pruebas de Neutralización , Polisacáridos , SARS-CoV-2/genética , Glicoproteína de la Espiga del Coronavirus/química , Glicoproteína de la Espiga del Coronavirus/genética , Glicoproteína de la Espiga del Coronavirus/metabolismo , Relación Estructura-Actividad
9.
Front Immunol ; 12: 692700, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34335603

RESUMEN

The highly pathogenic avian influenza (HPAI) H5N1 viruses with the capability of transmission from birds to humans have a serious impact on public health. To date, HPAI H5N1 viruses have evolved into ten antigenically distinct clades that could cause a mismatch of vaccine strains and reduce vaccine efficacy. In this study, the glycan masking and unmasking strategies on hemagglutinin antigen were used for designing two antigens: H5-dm/st2 and H5-tm/st2, and investigated for their elicited immunity using two-dose recombinant H5 (rH5) immunization and a first-dose adenovirus vector prime, followed by a second-dose rH5 protein booster immunization. The H5-dm/st2 antigen was found to elicit broadly neutralizing antibodies against different H5N1 clade/subclade viruses, as well as more stem-binding antibodies to inhibit HA-facilitated membrane fusion activity. Mice immunized with the H5-dm/st2 antigen had a higher survival rate when challenged with homologous and heterologous clades of H5N1 viruses. Mutant influenza virus replaced with the H5-dm/st2 gene generated by reverse genetics (RG) technology amplified well in MDCK cells and embryonated chicken eggs. Again, the inactivated H5N1-dm/st2 RG virus elicited more potent cross-clade neutralizing and anti-fusion antibodies in sera. Therefore, the H5N1-dm/st2 RG virus with the site-specific glycan-masking on the globular head and the glycan-unmasking on the stem region of H5 antigen can be used for further development of cross-protective H5N1 vaccines.


Asunto(s)
Anticuerpos Antivirales/inmunología , Antígenos Virales/administración & dosificación , Anticuerpos ampliamente neutralizantes/sangre , Glicoproteínas Hemaglutininas del Virus de la Influenza/administración & dosificación , Epítopos Inmunodominantes , Inmunogenicidad Vacunal , Subtipo H5N1 del Virus de la Influenza A/inmunología , Vacunas contra la Influenza/administración & dosificación , Infecciones por Orthomyxoviridae/prevención & control , Polisacáridos/administración & dosificación , Animales , Antígenos Virales/inmunología , Embrión de Pollo , Modelos Animales de Enfermedad , Perros , Femenino , Glicoproteínas Hemaglutininas del Virus de la Influenza/inmunología , Inmunización , Subtipo H5N1 del Virus de la Influenza A/genética , Subtipo H5N1 del Virus de la Influenza A/patogenicidad , Vacunas contra la Influenza/inmunología , Células de Riñón Canino Madin Darby , Ratones Endogámicos BALB C , Infecciones por Orthomyxoviridae/sangre , Infecciones por Orthomyxoviridae/inmunología , Infecciones por Orthomyxoviridae/virología , Polisacáridos/inmunología
10.
Methods Mol Biol ; 2248: 139-153, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33185873

RESUMEN

Virus-like particle (VLP) technology is an alternative platform for developing vaccines to combat seasonal and pandemic influenza. Influenza VLPs are non-infectious nanoparticles that can elicit effective vaccine immunogenicity in hosts. B-cell-activating factor (BAFF, or BLyS) and a proliferation-inducing ligand (APRIL) are members of the tumor necrosis factor (TNF) superfamily of cytokines. Both BAFF and APRIL are homotrimers that interact with homotrimeric receptors. Here, we report a method of the production of influenza VLPs by molecular incorporation with BAFF or APRIL homotrimers to interact with their receptors. We engineered the VLPs by direct fusion of BAFF or APRIL to the transmembrane anchored domain of the hemagglutinin (HA) gene. We also describe procedures for the production of BAFF-VLPs containing H5H7 and H1H5H7 for multi-subtype vaccine development.


Asunto(s)
Antígenos Virales/inmunología , Factor Activador de Células B/inmunología , Vacunas contra la Influenza/inmunología , Proteínas Recombinantes de Fusión , Miembro 13 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/inmunología , Vacunas de Partículas Similares a Virus/inmunología , Animales , Antígenos Virales/genética , Factor Activador de Células B/genética , Baculoviridae/genética , Clonación Molecular , Expresión Génica , Pruebas de Hemaglutinación , Glicoproteínas Hemaglutininas del Virus de la Influenza/inmunología , Humanos , Subtipo H5N1 del Virus de la Influenza A/inmunología , Plásmidos/genética , Proteínas Recombinantes de Fusión/inmunología , Miembro 13 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/genética
11.
Vaccines (Basel) ; 8(4)2020 Nov 30.
Artículo en Inglés | MEDLINE | ID: mdl-33266210

RESUMEN

Human infections with highly pathogenic avian influenza H5N1 viruses persist as a major global health concern. Vaccination remains the primary protective strategy against H5N1 and other novel avian influenza virus infections. We investigated the use of E. coli type IIb heat labile enterotoxin B subunit (LTIIb-B5) as a mucosal adjuvant for intranasal immunizations with recombinant HA proteins against H5N1 avian influenza viruses. Use of LTIIb-B5 adjuvant elicited more potent IgG, IgA, and neutralizing antibody titers in both sera and bronchoalveolar lavage fluids, thus increasing protection against lethal virus challenges. LTIIb-B5 mucosal adjuvanticity was found to trigger stronger Th17 cellular response in spleen lymphocytes and cervical lymph nodes. Studies of anti-IL-17A monoclonal antibody depletion and IL-17A knockout mice also suggest the contribution from Th17 cellular response to anti-H5N1 protective immunity. Our results indicate a link between improved protection against H5N1 live virus challenges and increased Th17 response due to the use of LTIIb-B5 mucosal adjuvant with HA subunit proteins.

12.
J Immunother Cancer ; 8(2)2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-33037116

RESUMEN

BACKGROUND: Emulsion adjuvants are a potent tool for effective vaccination; however, the size matters on mucosal signatures and the mechanism of action following intranasal vaccination remains unclear. Here, we launch a mechanistic study to address how mucosal membrane interacts with nanoemulsion of a well-defined size at cellular level and to elucidate the impact of size on tumor-associated antigen therapy. METHODS: The squalene-based emulsified particles at the submicron/nanoscale could be elaborated by homogenization/extrusion. The mucosal signatures following intranasal delivery in mice were evaluated by combining whole-mouse genome microarray and immunohistochemical analysis. The immunological signatures were tested by assessing their ability to influence the transportation of a model antigen ovalbumin (OVA) across nasal mucosal membranes and drive cellular immunity in vivo. Finally, the cancer immunotherapeutic efficacy is monitored by assessing tumor-associated antigen models consisting of OVA protein and tumor cells expressing OVA epitope. RESULTS: Uniform structures with ~200 nm in size induce the emergence of membranous epithelial cells and natural killer cells in nasal mucosal tissues, facilitate the delivery of protein antigen across the nasal mucosal membrane and drive broad-spectrum antigen-specific T-cell immunity in nasal mucosal tissues as well as in the spleen. Further, intranasal vaccination of the nanoemulsion could assist the antigen to generate potent antigen-specific CD8+ cytotoxic T-lymphocyte response. When combined with immunotherapeutic models, such an effective antigen-specific cytotoxic activity allowed the tumor-bearing mice to reach up to 50% survival 40 days after tumor inoculation; moreover, the optimal formulation significantly attenuated lung metastasis. CONCLUSIONS: In the absence of any immunostimulator, only 0.1% content of squalene-based nanoemulsion could rephrase the mucosal signatures following intranasal vaccination and induce broad-spectrum antigen-specific cellular immunity, thereby improving the efficacy of tumor-associated antigen therapy against in situ and metastatic tumors. These results provide critical mechanistic insights into the adjuvant activity of nanoemulsion and give directions for the design and optimization of mucosal delivery for vaccine and immunotherapy.


Asunto(s)
Adyuvantes Inmunológicos/uso terapéutico , Administración Intranasal/métodos , Inmunomodulación/inmunología , Inmunoterapia/métodos , Membrana Mucosa/inmunología , Nanopartículas/química , Vacunación/métodos , Adyuvantes Inmunológicos/farmacología , Animales , Femenino , Humanos , Ratones
13.
Hum Vaccin Immunother ; 16(9): 2176-2182, 2020 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-32530355

RESUMEN

The class II membrane fusion induced by flavivirus E proteins is a unique pH-dependent membrane fusion process differently from the class I or III membrane fusion by other enveloped virus proteins. The fusion peptide on the DII of the flavivirus E proteins can insert into the cell membrane as a cell entry process besides the receptor bindings. A traditional assay using C6/36 mosquito cells infected by dengue viruses has been previously reported but did not provide efficient quantitation to measure the virus-triggered membrane fusion. Here we reported the development of a quantitative cell fusion assay for four serotypes of dengue viruses and the recently emerged Zika viruses. We used a pCI-neo vector encoding the prME genes of dengue and Zika viruses and investigated the cell fusion in transfected 293, Vero and CHO cells. Donor cells were co-transfection of the prME genes of dengue and Zika prME gene and T7 RNA polymerase to react with the indicator cells transfected with the luciferase gene under the control of the T7 promoter. Quantification of the virus-induced cell fusion was determined by the luciferase expression levels under a switch of pH from 7.4 to 5.4 in the co-cultured donor and indicator cells. The quantitative luciferase-based assay was applied to measure the anti-fusion activity by two monoclonal antibodies mAb 4G2 and mAb DB42 against dengue virus infections. This assay could quality as a quantitative bioassay for testing the potency of anti-fusion monoclonal antibodies.


Asunto(s)
Virus del Dengue , Dengue , Infección por el Virus Zika , Virus Zika , Animales , Anticuerpos Antivirales , Bioensayo , Fusión Celular , Cricetinae , Cricetulus , Luciferasas , Fusión de Membrana , Serogrupo , Internalización del Virus
14.
Vaccines (Basel) ; 8(2)2020 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-32455704

RESUMEN

Human infections with H7N9 avian influenza A virus can result in severe diseases with high mortality. Developing an effective vaccine is urgently needed to prevent its pandemic potential. Vaccine delivery routes via mucosal surfaces are known to elicit mucosal immune responses such as secretory IgA antibodies in mucosal fluids, thus providing first-line protection at infection sites. PEG-b-PLACL (PELC) is a squalene-based oil-in-water emulsion adjuvant system that can enhance antigen penetration and uptake in nasal mucosal layers with enhanced mucin interactions. In this study, intranasal immunizations with recombinant H7 (rH7) proteins with a PELC/CpG adjuvant, as compared to the use of poly (I:C) or bacterial flagellin adjuvant, elicited higher titers of H7-specific IgG, IgA, hemagglutination inhibition, and neutralizing antibodies in sera, and increased numbers of H7-specific IgG- and IgA-antibody secreting cells in the spleen. Both PELC/CpG and poly (I:C) adjuvants at a dose as low as 5 µg HA provided an 80% survival rate against live virus challenges, but a lower degree of PELC/CpG-induced Th17 responses was observed. Therefore, the mucosal delivery of rH7 proteins formulated in a PELC/CpG adjuvant can be used for H7N9 mucosal vaccine development.

16.
Vaccines (Basel) ; 7(4)2019 Nov 22.
Artículo en Inglés | MEDLINE | ID: mdl-31766677

RESUMEN

Highly pathogenic avian influenza viruses are classified by the World Organization for Animal Health (OIE) as causes of devastating avian diseases. This study aimed to develop type IIb Escherichia coli heat-labile enterotoxin (LTIIb) as novel mucosal adjuvants for mucosal vaccine development. The fusion protein of H5 and LTIIb-A subunit was expressed and purified for mouse and chicken intranasal immunizations. Intranasal immunization with the H5-LTIIb-A fusion protein in mice elicited potent neutralizing antibodies in sera and bronchoalveolar lavage fluids, induced stronger Th1 and Th17 cellular responses in spleen and cervical lymph nodes, and improved protection against H5N1 influenza virus challenge. More interestingly, intranasal immunization with the H5-LTIIb-A fusion protein in chickens elicited high titers of IgY, IgA, hemagglutinin inhibition (HAI), and neutralizing antibodies in their antisera. This study employed the novel adjuvants of LTIIb for the development of a new generation of mucosal vaccines against highly pathogenic avian influenza viruses.

17.
Theranostics ; 9(16): 4811-4826, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31367259

RESUMEN

The viral E proteins of dengue virus (DENV) and Zika virus (ZIKV) are the major viral proteins involved in receptor binding and fusion, and for the induction of protective antibodies against viral infections. DIII of the E proteins is an independent domain and stretches out on the virion surface that can elicit type-specific neutralizing antibodies. For recombinant DIII vaccine development, prime-boost immunizations can provide an advantage of eliciting more type-specific neutralizing antibodies by recalling DIII antigens after DIII booster to improve protection. Methods: The DIII of the E genes of DENV and ZIKV were fused with bacterial fliC gene for the expression of flagellin-DIII (FliC-DIII) fusion proteins. Prime-boost immunization strategies by the second-dose booster of four DENV serotype or ZIKV FliC-DIII fusion proteins were used to investigate the induction of neutralizing antibodies and protection against viral infections. Cross-reactive non-neutralizing antibodies in each group of antisera were also examined using in vitro antibody-dependent enhancement (ADE) assay. A series of glycan-masking E antigens were finally constructed for prime-boost immunizations to abolish the elicitation of cross-reactive non-neutralizing antibodies for ADE activity. Results: We showed that inclusion of a bivalent live-attenuated vaccine with a FliC-DIII booster is superior in eliciting neutralization titers and protection in vivo against all four-serotype DENVs. We also demonstrated that recombinant adenovirus vectors encoding four-serotype DENV prMEs with a FliC-DIII prime-boost scheme is capable of eliciting good antibody responses. In contract, recombinant adenovirus vector of ZIKV prME gene priming, followed by ZIKV FliC-DIII booster did not improve vaccine efficacy. The glycan-masking mutation on the ZIKV E protein ij loop (E-248NHT), but not on DENV2 E protein ij loop (E-242NHT), resulted in abolishing the elicitation of cross-reactive antibodies for DENV and ZIKV infection enhancements. Conclusions: Our findings can provide useful information for designing novel immunogens and vaccination strategies in an attempt to develop a safe and efficacious DENV or ZIKV vaccine.


Asunto(s)
Virus del Dengue/inmunología , Dengue/inmunología , Flagelina/inmunología , Proteínas del Envoltorio Viral/inmunología , Infección por el Virus Zika/inmunología , Virus Zika/inmunología , Animales , Anticuerpos Antivirales/inmunología , Reacciones Cruzadas , Dengue/prevención & control , Dengue/virología , Virus del Dengue/química , Virus del Dengue/genética , Flagelina/administración & dosificación , Flagelina/genética , Humanos , Inmunización , Inmunización Secundaria , Ratones , Ratones Endogámicos BALB C , Polisacáridos/administración & dosificación , Polisacáridos/inmunología , Dominios Proteicos , Salmonella typhimurium/genética , Salmonella typhimurium/inmunología , Proteínas del Envoltorio Viral/administración & dosificación , Proteínas del Envoltorio Viral/química , Proteínas del Envoltorio Viral/genética , Vacunas Virales/administración & dosificación , Vacunas Virales/genética , Vacunas Virales/inmunología , Virus Zika/química , Virus Zika/genética , Infección por el Virus Zika/prevención & control , Infección por el Virus Zika/virología
18.
Vaccine ; 37(47): 6933-6941, 2019 11 08.
Artículo en Inglés | MEDLINE | ID: mdl-31383491

RESUMEN

The novel H7N9 avian influenza A virus has caused human infections in China since 2013; some isolates from the fifth wave of infections have emerged as highly pathogenic avian influenza viruses. Recombinant hemagglutinin proteins of H7N9 viruses can be rapidly and efficiently produced with low-level biocontainment facilities. In this study, recombinant H7 antigen was obtained from engineered stable clones of Chinese Hamster Ovary (CHO) cells for subsequent large-scale production. The stable CHO cell clones were also adapted to grow in serum-free suspension cultures. To improve the immunogenicity of the recombinant H7 antigens, we evaluated the use of a novel combination adjuvant of PELC and CpG (PELC/CpG) to augment the anti-H7N9 immune responses in mice. We compared the effects with other adjuvants such as alum, AddaVax (MF59-like), and several Toll-like receptor ligands such as R848, CpG, and poly (I:C). With the PELC/CpG combination adjuvant, CHO cell-expressed rH7 antigens containing terminally sialylated complex type N-glycans were able to induce high titers of neutralizing antibodies in sera and conferred protection following live virus challenges. These data indicate that the CHO cell-expressed recombinant H7 antigens and a PELC/CpG combination adjuvant can be used for H7N9 subunit vaccine development.


Asunto(s)
Adyuvantes Inmunológicos/administración & dosificación , Islas de CpG/inmunología , Hemaglutininas/inmunología , Subtipo H7N9 del Virus de la Influenza A/inmunología , Vacunas contra la Influenza/inmunología , Infecciones por Orthomyxoviridae/inmunología , Proteínas Recombinantes/inmunología , Compuestos de Alumbre/química , Animales , Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/inmunología , Células CHO , Línea Celular , Cricetulus , Femenino , Pruebas de Inhibición de Hemaglutinación/métodos , Glicoproteínas Hemaglutininas del Virus de la Influenza/inmunología , Ratones , Ratones Endogámicos BALB C , Polisorbatos/química , Escualeno/química , Vacunas de Subunidad/inmunología
19.
Antiviral Res ; 164: 12-22, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30738089

RESUMEN

Virus-like particle (VLP) technology is an attractive platform for the development of seasonal and pandemic influenza vaccines. Influenza VLPs can be obtained by the overexpression of HA, M1, NA, and/or M2 viral proteins in insect, mammalian, or plant cells. In this study, we reported to obtain highly immunogenic influenza VLPs by molecular incorporation with B-cell-activating factor (BAFF) or proliferation-inducing ligand (APRIL). Since BAFF and APRIL act as homotrimers to interact with their receptors, we engineered the VLPs by direct fusion of BAFF or APRIL to the transmembrane anchored domain of H5HA gene. Results showed that immunizations with the HA-transmembrane anchored BAFF- or APRIL-VLPs only formulated in alum but not MPL adjuvant elicited significantly higher IgG titers in sera. However, only the BAFF-VLPs formulated in alum adjuvant elicited more broadly neutralizing antibodies against the homologous and two heterologous H5N1 clade/subclade viruses and conferred protective immunity against live virus challenges. As the multi-subtype influenza vaccines containing a variety of HA subtypes can confer broader protective immunity, we also obtained multi-subtype H5H7 BAFF-VLPs and H1H5H7 BAFF-VLPs and demonstrated that these multi-subtype BAFF-VLPs were able to induce the production of neutralizing antibodies against multiple HA subtypes. Our findings provided useful information for the development of highly immunogenic, multi-subtype influenza VLP vaccines.


Asunto(s)
Anticuerpos Antivirales/sangre , Factor Activador de Células B/inmunología , Subtipo H5N1 del Virus de la Influenza A/inmunología , Vacunas contra la Influenza/inmunología , Infecciones por Orthomyxoviridae/prevención & control , Vacunas de Partículas Similares a Virus/inmunología , Animales , Anticuerpos Neutralizantes/sangre , Factor Activador de Células B/administración & dosificación , Reacciones Cruzadas , Femenino , Glicoproteínas Hemaglutininas del Virus de la Influenza/inmunología , Inmunoglobulina G/inmunología , Vacunas contra la Influenza/administración & dosificación , Ratones , Ratones Endogámicos BALB C , Neuraminidasa/inmunología , Infecciones por Orthomyxoviridae/inmunología , Miembro 13 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/administración & dosificación , Miembro 13 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/inmunología , Vacunas de Partículas Similares a Virus/administración & dosificación , Proteínas Virales/inmunología
20.
Biotechnol Bioeng ; 116(3): 598-609, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30080931

RESUMEN

Refocusing of B-cell responses can be achieved by preserving the overall fold of the antigen structure but selectively mutating the undesired antigenic sites with additional N-linked glycosylation motifs for glycan masking the vaccine antigen. We previously reported that glycan-masking recombinant H5 hemagglutinin (rH5HA) antigens on residues 83, 127, and 138 (g127 + g138 or g83 + g127 + 138 rH5HA) elicited broader neutralizing antibodies and protection against heterologous clades/subclades of high pathogenic avian influenza H5N1 viruses. In this study, we engineered the stably expressing Chinese hamster ovary (CHO) cell clones for producing the glycan-masking g127 + g138 and g83 + g127 + g138 rH5HA antigens. All of these glycan-masking rH5HA antigens produced in stable CHO cell clones were found to be mostly oligomeric structures. Only the immunization with the glycan-masking g127 + g138 but not g83 + g127 + g138 rH5HA antigens elicited more potent neutralizing antibody titers against four out of five heterologous clades/subclades of H5N1 viral strains. The increased neutralizing antibody titers against these heterologous viral strains were correlated with the increased amounts of stem-binding antibodies, only the glycan-masking g127 + g138 rH5HA antigens can translate into more protection against live viral challenges. The stable CHO cell line-produced glycan-masking g127 + g138 rH5HA can be used for H5N1 subunit vaccine development.


Asunto(s)
Glicoproteínas Hemaglutininas del Virus de la Influenza , Subtipo H5N1 del Virus de la Influenza A , Vacunas contra la Influenza , Ingeniería de Proteínas/métodos , Proteínas Recombinantes , Animales , Anticuerpos Antivirales/sangre , Anticuerpos Antivirales/inmunología , Antígenos Virales/química , Antígenos Virales/genética , Antígenos Virales/inmunología , Antígenos Virales/metabolismo , Células CHO , Cricetinae , Cricetulus , Femenino , Glicoproteínas Hemaglutininas del Virus de la Influenza/química , Glicoproteínas Hemaglutininas del Virus de la Influenza/genética , Glicoproteínas Hemaglutininas del Virus de la Influenza/inmunología , Glicoproteínas Hemaglutininas del Virus de la Influenza/metabolismo , Subtipo H5N1 del Virus de la Influenza A/genética , Subtipo H5N1 del Virus de la Influenza A/inmunología , Vacunas contra la Influenza/química , Vacunas contra la Influenza/genética , Vacunas contra la Influenza/inmunología , Vacunas contra la Influenza/metabolismo , Ratones , Ratones Endogámicos BALB C , Polisacáridos/química , Polisacáridos/genética , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/inmunología , Proteínas Recombinantes/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA