Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 98
Filtrar
1.
Virus Res ; 329: 199101, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-36958398

RESUMO

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.


Assuntos
Enterovirus Humano A , Enterovirus , Doença de Mão, Pé e Boca , Animais , Camundongos , Doença de Mão, Pé e Boca/prevenção & controle , Anticorpos Neutralizantes , Anticorpos Antivirais , Vacinas de Produtos Inativados , Enterovirus Humano A/genética
2.
Biol Open ; 11(9)2022 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-35929543

RESUMO

Enterovirus 71 (EV71) is one of the causative agents of hand-foot-and-mouth disease, which in some circumstances could lead to severe neurological diseases. Despite of its importance for human health, little is known about the early stages of EV71 infection. EV71 starts uncoating with its receptor, human scavenger receptor B2 (hSCARB2), at low pH. We show that EV71 was not targeted to lysosomes in human rhabdomyosarcoma cells overexpressing hSCARB2 and that the autophagic pathway is not essential for EV71 productive uncoating. Instead, EV71 was efficiently uncoated 30 min after infection in late endosomes (LEs) containing hSCARB2, mannose-6-phosphate receptor (M6PR), RAB9, bis(monoacylglycero)phosphate and lysosomal associated membrane protein 2 (LAMP2). Furthering the notion that mature LEs are crucial for EV71 uncoating, cation-dependent (CD)-M6PR knockdown impairs EV71 infection. Since hSCARB2 interacts with cation-independent (CI)-M6PR through M6P-binding sites and CD-M6PR also harbor a M6P-binding site, CD-M6PR is likely to play important roles in EV71 uncoating in LEs.


Assuntos
Enterovirus Humano A , Infecções por Enterovirus , Enterovirus , Animais , Cátions/metabolismo , Endossomos/metabolismo , Enterovirus/metabolismo , Enterovirus Humano A/metabolismo , Humanos , Proteínas de Membrana Lisossomal/química , Proteínas de Membrana Lisossomal/genética , Proteínas de Membrana Lisossomal/metabolismo , Receptor IGF Tipo 2/metabolismo , Receptores Depuradores/química , Receptores Depuradores/genética , Receptores Depuradores/metabolismo
3.
Cells ; 9(5)2020 05 09.
Artigo em Inglês | MEDLINE | ID: mdl-32397494

RESUMO

Microbial proteins have recently been found to have more benefits in clinical disease treatment because of their better-developed strategy and properties than traditional medicine. In this study, we investigated the effectiveness of a truncated peptide synthesized from the C-terminal sequence of pneumolysin, i.e., C70PLY4, in Streptococcus pneumoniae, in treating chronic inflammatory conditions. It has been shown that C70PLY4 significantly blocks the transendothelial migration of neutrophils and attenuates the formation of atherosclerotic plaque and the secretion of soluble forms of the intercellular adhesion molecule-1 (ICAM-1), the vascular cell adhesion molecule 1 (VCAM-1), and E-selectin in high-fat-diet/streptozotocin-induced inflammatory rats. The mechanism and the docking simulation analysis further indicated that C70PLY4 might serve as a Toll-like receptor 4 (TLR4) antagonist by competing for the binding site of MD2, an indispensable protein for lipopolysaccharide (LPS)-TLR4 interaction signaling, on the TLR4 structure. Moreover, compared to the full-length PLY, C70PLY4 seems to have no cytotoxicity in human vascular endothelial cells. Our study elucidated a possible therapeutic efficacy of C70PLY4 in reducing chronic inflammatory conditions and clarified the underlying mechanism. Thus, our findings identify a new drug candidate that, by blocking TLR4 activity, could be an effective treatment for patients with chronic inflammatory diseases.


Assuntos
Inflamação/tratamento farmacológico , Proteínas Mutantes/farmacologia , Proteínas Mutantes/uso terapêutico , Streptococcus pneumoniae/metabolismo , Estreptolisinas/farmacologia , Receptor 4 Toll-Like/antagonistas & inibidores , Sequência de Aminoácidos , Animais , Apoptose/efeitos dos fármacos , Proteínas de Bactérias/química , Proteínas de Bactérias/farmacologia , Sítios de Ligação , Caspase 3/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Dieta Hiperlipídica , Selectina E/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Molécula 1 de Adesão Intercelular/metabolismo , Lipopolissacarídeos , Camundongos , Simulação de Acoplamento Molecular , Proteínas Mutantes/química , NF-kappa B/metabolismo , Neutrófilos/citologia , Neutrófilos/efeitos dos fármacos , Fosforilação/efeitos dos fármacos , Solubilidade , Estreptolisinas/química , Estreptozocina , Receptor 4 Toll-Like/metabolismo , Migração Transendotelial e Transepitelial/efeitos dos fármacos , Molécula 1 de Adesão de Célula Vascular/metabolismo
4.
ACS Appl Mater Interfaces ; 10(15): 12553-12561, 2018 Apr 18.
Artigo em Inglês | MEDLINE | ID: mdl-29595053

RESUMO

To accomplish an innovative vaccine design, there are two key challenges: developing formulations that avoid cold chain shipment and finding a delivery vehicle that is absorbable in vivo. Here, we explored the design and performance of a colloidal vesicle that enabled us to consider both challenges. We used polymeric bioresorbable amphiphiles as surface-active agents for stabilizing oily/aqueous interfaces and formed a colloidal vehicle named polysorbasome (PSS, polymeric absorbable vesicle), without using conventional emulsifiers such as sorbitan esters or their ethoxylates. Homogenizing the oil/water compartments forms a colloid containing an aqueous solution in its core and provides an oily barrier that isolates the encapsulated material from external materials. In this form, the PSS serves as a depot for sustained delivery of vaccine antigens. Following vaccination, the antigen-specific antibodies and the cell-mediated immunity can be manipulated after the antigen being formulated with PSS particles. Then, the degradability intrinsic to the polymeric bioresorbable amphiphiles complies with the destruction and further absorbance of the vehicles in vivo. The structural features of these versatile vesicles based on bioresorbable amphiphilic engineering may provide new insights into vaccine delivery.


Assuntos
Implantes Absorvíveis , Coloides , Sistemas de Liberação de Medicamentos , Polímeros , Vacinas
5.
Vaccine ; 36(22): 3134-3139, 2018 05 24.
Artigo em Inglês | MEDLINE | ID: mdl-28274636

RESUMO

Hand, foot and mouth diseases (HFMD) are mainly caused by Enterovirus A71 (EV-A71) infections. Clinical trials in Asia conducted with formalin-inactivated EV-A71 vaccine candidates produced from serum-free Vero cell culture using either roller bottle or cell factory technology, are found to be safe and highly efficacious. To increase vaccine yields and reduce the production costs, the bioprocess improvement for EV-A71 vaccine manufacturing is currently being investigated. The parameters that could affect and enhance the production yields of EV-A71 virus growth in the microcarrier bioreactor were investigated. The medium replacement culture strategy included a multi-harvested semi-batch process and perfusion technology and was found to increase the production yields more than 7-14 folds. Based on the western blot and cryo-EM analyses of the EV-A71 virus particles produced from either the multi-harvested semi-batch (MHSBC) or perfusion cultures were found to be similar to those virus particles obtained from the single batch culture. Mouse immunogenicity studies indicate that the EV-A71 vaccine candidates produced from the perfusion culture have similar potency to those obtained from single batch bioprocess. The physical structures of the EV-A71 particles revealed by the cryo-EM analysis were found to be spherical capsid particles. These results provide feasible technical bioprocesses for increasing virus yields and the scale up of EV-A71 vaccine manufacturing using the bioreactor cell culture methods.


Assuntos
Reatores Biológicos/virologia , Técnicas de Cultura de Células/métodos , Enterovirus Humano A/crescimento & desenvolvimento , Vacinas Virais/biossíntese , Cultura de Vírus/métodos , Animais , Técnicas de Cultura Celular por Lotes , Chlorocebus aethiops , Imunogenicidade da Vacina , Camundongos , Testes de Neutralização , Vacinas de Produtos Inativados/biossíntese , Células Vero
6.
Sci Rep ; 6: 36732, 2016 11 09.
Artigo em Inglês | MEDLINE | ID: mdl-27827451

RESUMO

This study describes the feasibility and adjuvant mechanism of a degradable emulsion for tuning adaptive immune responses to a vaccine antigen. We featured a mouse model with ovalbumin (OVA) as the antigen to deepen our understanding of the properties of a degradable emulsion-based adjuvant, dubbed PELC, interacting with immune cells and to elucidate their roles in vaccine immunogenicity in vivo. First, we demonstrated that the emulsion, which is stabilized by an amphiphilic bioresorbable polymer, shows degradation in mimic human body conditions and considerable tolerance in vivo. Then, we confirmed the model protein could be loaded into the emulsion and released from the matrix in a sustained manner, subsequently driving the production of antigen-specific antibodies. We also comprehended that PELC not only recruits antigen-presenting cells (APCs) to the injection site but also induces the activation of the recruited APCs and migration to the draining lymph nodes. As an adjuvant for cancer immunotherapy, PELC-formulated OVA could strongly enhance antigen-specific T-cell responses as well as anti-tumor ability with respected to non-formulated OVA, using OVA protein/EG7 cells as a tumor antigen/tumor cell model. Accordingly, our data paved the way for the clinical application of degradable emulsions based on amphiphilic bioresorbable polymers as vaccine adjuvants.


Assuntos
Adjuvantes Imunológicos , Antígenos de Neoplasias , Poliésteres , Polietilenoglicóis , Vacinas , Adjuvantes Imunológicos/química , Adjuvantes Imunológicos/farmacocinética , Adjuvantes Imunológicos/farmacologia , Animais , Anticorpos/imunologia , Células Apresentadoras de Antígenos/imunologia , Antígenos de Neoplasias/química , Antígenos de Neoplasias/imunologia , Antígenos de Neoplasias/farmacologia , Linhagem Celular , Emulsões , Feminino , Camundongos , Camundongos Endogâmicos BALB C , Poliésteres/química , Poliésteres/farmacocinética , Poliésteres/farmacologia , Polietilenoglicóis/química , Polietilenoglicóis/farmacocinética , Polietilenoglicóis/farmacologia , Linfócitos T/imunologia , Vacinas/química , Vacinas/imunologia , Vacinas/farmacocinética , Vacinas/farmacologia
7.
Antiviral Res ; 132: 225-32, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27387826

RESUMO

Enterovirus A71 (EV-A71) is responsible for epidemics of hand, foot and mouth disease (HFMD) in young children. To circumvent difficulties in obtaining clinical enterovirus isolates that might be contaminated with other viruses, a platform technology was developed to quickly generate vaccine virus strains based on the published enterovirus genomic sequences. A recombinant plasmid containing the full-length infectious cDNA clone of EV-A71 vaccine strain E59 was directly generated after transfecting the recombinant plasmid into Vero, RD or HEK293A cells, and phenotypic characteristics similar to the parental strain were observed. The cDNA-derived infectious EV-A71 virus grown in Vero cells produced relatively stable virus titers in both T-flasks and microcarrier culture systems. To evaluate the genetic stability of the cDNA-derived EV-A71 viruses, the immunodominant structural proteins, VP1 and VP2, of the recombinant EV-A71 viruses were sequenced and analyzed. The cDNA-derived EV-A71 virus showed weak pathogenicity in a human SCARB2 mouse model. These results show the successful generation of a recombinant virus derived from a published viral genomic sequence that demonstrated good genetic stability and viral yields, which could represent an efficient and safe vaccine strain for cGMP-grade manufacturing.


Assuntos
DNA Complementar , Enterovirus Humano A/genética , Enterovirus Humano A/imunologia , Infecções por Enterovirus/imunologia , Genoma Viral , Vacinas de DNA/genética , Vacinas de DNA/imunologia , Substituição de Aminoácidos , Animais , Anticorpos Neutralizantes/sangue , Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/sangue , Anticorpos Antivirais/imunologia , Antígenos Virais/genética , Antígenos Virais/imunologia , Chlorocebus aethiops , Modelos Animais de Doenças , Enterovirus Humano A/patogenicidade , Infecções por Enterovirus/prevenção & controle , Infecções por Enterovirus/virologia , Ordem dos Genes , Instabilidade Genômica , Humanos , Epitopos Imunodominantes/genética , Epitopos Imunodominantes/imunologia , Camundongos , Camundongos Transgênicos , Mutação , Células Vero , Replicação Viral
8.
Microbes Infect ; 18(11): 706-709, 2016 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-27394146

RESUMO

Here we investigate the immunogenicity of a combination adjuvant comprising emulsified fine particles (namely PELC) together with LD-indolicidin, a peptiomimetic stereoisomer of the bovine neutrophil peptide indolicidin. We demonstrated that intranasal vaccination with either PELC/LD-indolicidin or Alum enhances protective influenza-specific serological immunity in mice; however, the adjuvant potency of Alum was rather reduced when the mice vaccinated orally with formulated influenza vaccines. The information gathered from this study will enhance our effort in the formulation design as well as the optimization of alternative administration routes for prophylactic vaccines against emerging infectious diseases, in particular pandemic influenza.


Assuntos
Adjuvantes Imunológicos/farmacologia , Peptídeos Catiônicos Antimicrobianos/farmacologia , Vacinas contra Influenza/imunologia , Orthomyxoviridae/imunologia , Poliésteres/farmacologia , Polietilenoglicóis/farmacologia , Esqualeno/farmacologia , Adjuvantes Imunológicos/administração & dosagem , Administração Intranasal , Administração Oral , Compostos de Alúmen/administração & dosagem , Compostos de Alúmen/farmacologia , Animais , Anticorpos Antivirais/sangue , Peptídeos Catiônicos Antimicrobianos/administração & dosagem , Feminino , Vacinas contra Influenza/administração & dosagem , Camundongos Endogâmicos BALB C , Poliésteres/administração & dosagem , Polietilenoglicóis/administração & dosagem , Esqualeno/administração & dosagem
9.
Sci Rep ; 6: 30648, 2016 07 29.
Artigo em Inglês | MEDLINE | ID: mdl-27470096

RESUMO

We developed a novel platform to express high levels of recombinant lipoproteins with intrinsic adjuvant properties. Based on this technology, our group developed recombinant lipidated dengue envelope protein domain IIIs as vaccine candidates against dengue virus. This work aims to evaluate the immune responses in mice to the tetravalent formulation. We demonstrate that 4 serotypes of recombinant lipidated dengue envelope protein domain III induced both humoral and cellular immunity against all 4 serotypes of dengue virus on the mixture that formed the tetravalent formulation. Importantly, the immune responses induced by the tetravalent formulation in the absence of the exogenous adjuvant were functional in clearing the 4 serotypes of dengue virus in vivo. We affirm that the tetravalent formulation of recombinant lipidated dengue envelope protein domain III is a potential vaccine candidate against dengue virus and suggest further detailed studies of this formulation in nonhuman primates.


Assuntos
Vacinas contra Dengue/imunologia , Vírus da Dengue/imunologia , Dengue/prevenção & controle , Lipoproteínas/imunologia , Proteínas Recombinantes/imunologia , Proteínas do Envelope Viral/imunologia , Animais , Vacinas contra Dengue/administração & dosagem , Vacinas contra Dengue/genética , Modelos Animais de Doenças , ELISPOT , Feminino , Imunidade Celular , Imunidade Humoral , Lipoproteínas/genética , Camundongos Endogâmicos BALB C , Testes de Neutralização , Domínios Proteicos , Proteínas Recombinantes/genética , Sorogrupo , Vacinas Sintéticas/administração & dosagem , Vacinas Sintéticas/genética , Vacinas Sintéticas/imunologia , Proteínas do Envelope Viral/genética
10.
Hum Vaccin Immunother ; 12(7): 1678-89, 2016 07 02.
Artigo em Inglês | MEDLINE | ID: mdl-26901482

RESUMO

Dengue has a major impact on global public health, and the use of dengue vaccine is very limited. In this study, we evaluated the immunogenicity and protective efficacy of a dengue vaccine made from a recombinant measles virus (MV) that expresses envelope protein domain III (ED3) of dengue-1 to 4. Following immunization with the MV-vectored dengue vaccine, mice developed specific interferon-gamma and antibody responses against dengue virus and MV. Neutralizing antibodies against MV and dengue viruses were also induced, and protective levels of FRNT50 ≥ 10 to 4 serotypes of dengue viruses were detected in the MV-vectored dengue vaccine-immunized mice. In addition, specific interferon-gamma and antibody responses to dengue viruses were still induced by the MV-vectored dengue vaccine in mice that were pre-infected with MV. This finding suggests that the pre-existing immunity to MV did not block the initiation of immune responses. By contrast, mice that were pre-infected with dengue-3 exhibited no effect in terms of their antibody responses to MV and dengue viruses, but a dominant dengue-3-specific T-cell response was observed. After injection with dengue-2, a detectable but significantly lower viremia and a higher titer of anti-dengue-2 neutralizing antibodies were observed in MV-vectored dengue vaccine-immunized mice versus the vector control, suggesting that an anamnestic antibody response that provided partial protection against dengue-2 was elicited. Our results with regard to T-cell responses and the effect of pre-immunity to MV or dengue viruses provide clues for the future applications of an MV-vectored dengue vaccine.


Assuntos
Formação de Anticorpos , Vacinas contra Dengue/imunologia , Portadores de Fármacos , Vírus do Sarampo/genética , Linfócitos T/imunologia , Animais , Anticorpos Neutralizantes/sangue , Anticorpos Antivirais/sangue , Vacinas contra Dengue/administração & dosagem , Vacinas contra Dengue/genética , Interferon gama/metabolismo , Vírus do Sarampo/imunologia , Camundongos Endogâmicos C57BL , Vacinas Sintéticas/administração & dosagem , Vacinas Sintéticas/genética , Vacinas Sintéticas/imunologia
11.
Antiviral Res ; 129: 58-66, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-26899790

RESUMO

Childhood exanthema caused by different serotypes of coxsackievirus (CV-A) and enterovirus A71 (EV-A71) has become a serious global health problem; it is commonly known as hand, foot, and mouth disease (HFMD). Current EV-A71 vaccine clinical trials have demonstrated that human antibody responses generated by EV-A71 vaccinations do not cross-neutralize coxsackievirus A16 (CV-A16). An effective multivalent HFMD vaccine is urgently needed. From molecular epidemiological studies in Southeast Asia, CV-A6 and CV-A10 are commonly found in HFMD outbreaks. In this study, CV-A6 and CV-A10 were individually cultured in rhabdomyosarcoma (RD) cells grown in medium containing serum, harvested and concentrated. In viral downstream purification, two viral fractions were separated by sucrose gradient zonal ultracentrifugation and detected using a SDS-PAGE analysis and a virus infectivity assay. These two viral fractions were formalin-inactivated, and only the infectious particle fraction was found to be capable of inducing CV-A serotype-specific neutralizing antibody responses in animal immunogenicity studies. These mouse and rabbit antisera also failed to cross-neutralize EV-A71 and CV-A16 infections. Only a combination of formalin-inactivated EV-A71, CV-A6, CV-A10 and CV-A16 multivalent vaccine candidates elicited cross-neutralizing antibody responses in both mouse and rabbit immunogenicity studies. The current results certainly provide important information for multivalent HFMD vaccine development.


Assuntos
Anticorpos Antivirais/imunologia , Enterovirus Humano A/imunologia , Vacinas Virais/imunologia , Vírion/imunologia , Animais , Anticorpos Neutralizantes/biossíntese , Anticorpos Neutralizantes/sangue , Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/biossíntese , Anticorpos Antivirais/sangue , Antígenos Virais/química , Antígenos Virais/imunologia , Reações Cruzadas , Enterovirus Humano A/química , Enterovirus Humano A/isolamento & purificação , Enterovirus Humano A/ultraestrutura , Infecções por Enterovirus/imunologia , Genótipo , Doença de Mão, Pé e Boca/imunologia , Doença de Mão, Pé e Boca/virologia , Camundongos , Coelhos , Alinhamento de Sequência , Vacinação , Vacinas de Produtos Inativados/imunologia , Tropismo Viral , Vírion/química , Vírion/isolamento & purificação
12.
Vaccine ; 34(8): 1054-61, 2016 Feb 17.
Artigo em Inglês | MEDLINE | ID: mdl-26776472

RESUMO

The linkage of an immunogen with a toll-like receptor ligand has great potential to induce highly potent immune responses with the initial features of antigen-presenting cell activation. In the current study, we expressed recombinant dengue-3 envelope protein domain III (D3ED III) in lipidated form using an Escherichia coli-based system. The recombinant lipidated dengue-3 envelope protein domain III (LD3ED III) augments the expression levels of IL-12 family cytokines. LD3ED III-immunized mice enhance wide ranges of T cell responses as indicated by IFN-γ, IL-17, IL-21 production. Additionally, LD3ED III-immunized mice increase the frequencies of anti-D3ED III antibody producing cells. The boosted antibody titers cover various IgG isotypes, including IgG1, IgG2a, IgG2b, and IgG3. Importantly, LD3ED III-immunized mice induce neutralizing antibody capacity associated with a reduction of viremia levels after challenges. In contrast, mice that are immunized with D3ED III formulated with aluminum phosphate (D3ED III/Alum) only enhance Th2 responses and boost IgG1 antibody titers. Neither neutralizing antibody responses nor the inhibition of viremia levels after challenge is observed in mice that are immunized with D3ED III/Alum. These results suggest that LD3ED III can induce broad profiles of cellular and humoral immune responses.


Assuntos
Vacinas contra Dengue/imunologia , Dengue/prevenção & controle , Proteínas do Envelope Viral/imunologia , Adjuvantes Imunológicos/farmacologia , Animais , Anticorpos Neutralizantes/sangue , Anticorpos Antivirais/sangue , Citocinas/imunologia , Células Dendríticas/imunologia , Vírus da Dengue , Feminino , Imunidade Celular , Imunidade Humoral , Imunoglobulina G/sangue , Lipídeos/química , Camundongos , Camundongos Endogâmicos BALB C , Proteínas Recombinantes/imunologia , Linfócitos T/imunologia , Viremia/prevenção & controle
13.
PLoS One ; 10(12): e0145717, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26714037

RESUMO

Dengue is the leading cause of mosquito-borne viral infections and no vaccine is available now. Envelope protein domain III (ED3) is the major target for the binding of dengue virus neutralizing antibodies; however, the ED3-specifc T-cell response is less well understood. To investigate the T-cell responses to four serotypes of dengue virus (DENV-1 to 4), we immunized mice using either a tetravalent ED3-based DNA or protein vaccine, or combined both as a DNA prime-protein boost strategy (prime-boost). A significant serotype-dependent IFN-γ or IL-4 response was observed in mice immunized with either the DNA or protein vaccine. The IFN-γ response was dominant to DENV-1 to 3, whereas the IL-4 response was dominant to DENV-4. Although the similar IgG titers for the four serotypes were observed in mice immunized with the tetravalent vaccines, the neutralizing antibody titers varied and followed the order of 2 = 3>1>4. Interestingly, the lower IFN-γ response to DENV-4 is attributable to the immunodominance change between two CD4+ T-cell epitopes; one T-cell epitope located at E349-363 of DENV-1 to 3 was more immunogenic than the DENV-4 epitope E313-327. Despite DENV-4 specific IFN-γ responses were suppressed by immunodominance change, either DENV-4-specific IFN-γ or neutralizing antibody responses were still recalled after DENV-4 challenge and contributed to virus clearance. Immunization with the prime-boost elicited both IFN-γ and neutralizing antibody responses and provided better protection than either DNA or protein immunization. Our findings shed light on how ED3-based tetravalent dengue vaccines sharpen host CD4 T-cell responses and contribute to protection against dengue virus.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Vacinas contra Dengue/química , Vacinas contra Dengue/imunologia , Epitopos Imunodominantes/imunologia , Proteínas do Envelope Viral/química , Proteínas do Envelope Viral/imunologia , Sequência de Aminoácidos , Animais , Feminino , Imunização , Camundongos , Camundongos Endogâmicos BALB C , Dados de Sequência Molecular , Estrutura Terciária de Proteína
14.
Int J Infect Dis ; 41: 56-64, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26555647

RESUMO

OBJECTIVES: The regulation of the immunopathology of respiratory syncytial virus (RSV) by regulatory T-cells (CD4(+)CD25(+)Foxp3(+); Tregs) is not understood. METHODS: To deduce the same, Tregs were depleted in BALB/c mice by injecting anti-CD25 antibody followed by RSV infection (anti-CD25-RSV mice). RESULTS: In this model, a decrease in anti-fusion (F) antibody and neutralizing activity, and an increase in anti-nucleocapsid (N) antibody in serum, were seen. Decreased antibody-dependent cell-mediated cytotoxicity (ADCC) activity, increased IgG2a, and an influx of activated CD8(+) T-cells into the lungs were also observed. Co-culture of splenic CD45RA(+) B-cells from RSV-infected normal mice with CD4(+) cells isolated from anti-CD25-RSV mice (B/CD4) increased anti-F antibody secretion. The inclusion of CD25(+) Tregs isolated from isotype Ig-RSV mice into the B/CD4 co-culture substantially enhanced the frequency of anti-F antibody production. However, the same effect was not seen in the co-culture of CD45RA(+) B-cells with dendritic cells (DCs) (B/DCs) or CD8(+) cells (B/CD8) that were obtained from anti-CD25-RSV mice. The transfer of enriched B-cells from anti-CD25-RSV mice into RSV-infected SCID mice increased severe lung inflammation associated with the increased viral load and eosinophil number. CONCLUSIONS: These results indicate that Tregs modulate B-cell activity, particularly in producing F-specific neutralizing antibodies, to regulate RSV-mediated exacerbated diseases.


Assuntos
Linfócitos B/fisiologia , Infecções por Vírus Respiratório Sincicial/imunologia , Linfócitos T Reguladores/fisiologia , Animais , Anticorpos Neutralizantes , Anticorpos Antivirais , Linfócitos T CD8-Positivos/imunologia , Feminino , Pulmão/patologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos SCID , Infecções por Vírus Respiratório Sincicial/virologia , Vírus Sinciciais Respiratórios/imunologia , Baço/patologia
15.
Expert Rev Vaccines ; 14(12): 1623-32, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26420467

RESUMO

Recombinant bacterial lipoproteins (RLP) with built-in immuno-stimulating properties for novel subunit vaccine development are reviewed. This platform technology offers the following advantages: easily converts antigens into highly immunogenic RLP using a fusion sequence containing lipobox; the lipid moiety of RLP is recognized as the danger signals in the immune system through the Toll-like receptor 2, so both innate and adaptive immune responses can be induced by RLP; serves as an efficient and cost-effective bioprocess for producing RLP in Escherichia coli and the feasibility and safety of this core platform technology has been successfully demonstrated in animal model studies including meningococcal group B subunit vaccine, dengue subunit vaccine, novel subunit vaccine against Clostridium difficile-associated diseases and HPV-based immunotherapeutic vaccines.


Assuntos
Antígenos de Bactérias/imunologia , Desenho de Fármacos , Lipoproteínas/imunologia , Vacinas de Subunidades Antigênicas/imunologia , Vacinas Sintéticas/imunologia , Adjuvantes Imunológicos , Antígenos de Superfície/imunologia , Cápsulas Bacterianas/imunologia , Proteínas da Membrana Bacteriana Externa/imunologia , Vacinas Bacterianas/imunologia , Borrelia burgdorferi/imunologia , Clostridioides difficile/imunologia , Dengue/prevenção & controle , Vacinas contra Dengue/imunologia , Vírus da Dengue/imunologia , Humanos , Neisseria meningitidis/imunologia , Vacinação
16.
PLoS One ; 10(8): e0135045, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26271033

RESUMO

Clostridium difficile is an emerging pathogen responsible for opportunistic infections in hospitals worldwide and is the main cause of antibiotic-associated pseudo-membranous colitis and diarrhea in humans. Clostridial toxins A and B (TcdA and TcdB) specifically bind to unknown glycoprotein(s) on the surface of epithelial cells in the host intestine, disrupting the intestinal barrier and ultimately leading to acute inflammation and diarrhea. The C-terminal receptor-binding domain (RBD) of TcdA, which is responsible for the initial binding of the toxin to host glycoproteins, has been predicted to contain 7 potential oligosaccharide-binding sites. To study the specific roles and functions of these 7 putative lectin-like binding regions, a consensus sequence of TcdA RBD derived from different C. difficile strains deposited in the NCBI protein database and three truncated fragments corresponding to the N-terminal (residues 1-411), middle (residues 296-701), and C-terminal portions (residues 524-911) of the RBD (F1, F2 and F3, respectively) were designed and expressed in Escherichia coli. In this study, the recombinant RBD (rRBD) and its truncated fragments were purified, characterized biologically and found to have the following similar properties: (a) are capable of binding to the cell surface of both Vero and Caco-2 cells; (b) possess Toll-like receptor agonist-like adjuvant activities that can activate dendritic cell maturation and increase the secretion of pro-inflammatory cytokines; and (c) function as potent adjuvants in the intramuscular immunization route to enhance immune responses against weak immunogens. Although F1, F2 and F3 have similar repetitive amino acid sequences and putative oligosaccharide-binding domains, they do not possess the same biological and immunological properties: (i) TcdA rRBD and its fragments bind to the cell surface, but only TcdA rRBD and F3 internalize into Vero cells within 15 min; (ii) the fragments exhibit various levels of hemagglutinin (HA) activity, with the exception of the F1 fragment, which demonstrates no HA activity; and (iii) in the presence of alum, all fragments elicit various levels of anti-toxin A-neutralizing antibody responses, but those neutralizing antibodies elicited by F2 did not protect mice against a TcdA challenge. Because TcdA rRBD, F1 and F3 formulated with alum can elicit immune protective responses against the cytotoxicity of TcdA, they represent potential components of future candidate vaccines against C. difficile-associated diseases.


Assuntos
Toxinas Bacterianas/química , Toxinas Bacterianas/imunologia , Enterotoxinas/química , Enterotoxinas/imunologia , Domínios e Motivos de Interação entre Proteínas , Sequência de Aminoácidos , Animais , Toxinas Bacterianas/metabolismo , Vacinas Bacterianas/imunologia , Vacinas Bacterianas/metabolismo , Células CACO-2 , Células Cultivadas , Chlorocebus aethiops , Clostridioides difficile , Enterotoxinas/metabolismo , Feminino , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Dados de Sequência Molecular , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/imunologia , Fragmentos de Peptídeos/metabolismo , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas/imunologia , Células Vero
17.
J Biomed Sci ; 22: 65, 2015 Aug 07.
Artigo em Inglês | MEDLINE | ID: mdl-26245825

RESUMO

BACKGROUND: Opportunistically nosocomial infections in hospitalized patients are often related to Clostridium difficile infections (CDI) due to disruption of the intestinal micro-flora by antibiotic therapies during hospitalization. Clostridial exotoxins A and B (TcdA and TcdB) specifically bind to unknown glycoprotein(s) in the host intestine, disrupt the intestinal barrier leading to acute inflammation and diarrhea. The C-terminal receptor binding domain of TcdA (A-rRBD) has been shown to elicit antibody responses that neutralize TcdA toxicity in Vero cell cytotoxicity assays, but not effectively protect hamsters against a lethal dose challenge of C. difficile spores. To develop an effective recombinant subunit vaccine against CDI, A-rRBD was lipidated (rlipoA-RBD) as a rational design to contain an intrinsic adjuvant, a toll-like receptor 2 agonist and expressed in Escherichia coli. RESULTS: The purified rlipoA-RBD was characterized immunologically and found to have the following properties: (a) mice, hamsters and rabbits vaccinated with 3 µg of rlipoA-RBD produced strong antibody responses that neutralized TcdA toxicity in Vero cell cytotoxicity assays; furthermore, the neutralization titer was comparable to those obtained from antisera immunized either with 10 µg of TcdA toxoid or 30 µg of A-rRBD; (b) rlipoA-RBD elicited immune responses and protected mice from TcdA challenge, but offered insignificant protection (10 to 20 %) against C. difficile spores challenge in hamster models; (c) only rlipoA-RBD formulated with B-rRBD consistently confers protection (90 to 100 %) in the hamster challenge model; and (d) rlipoA-RBD was found to be 10-fold more potent than A-rRBD as an adjuvant to enhancing immune responses against a poor antigen such as ovalbumin. CONCLUSION: These results indicate that rlipoA-RBD formulated with B-rRBD could be an excellent vaccine candidate for preclinical studies and future clinical trials.


Assuntos
Vacinas Bacterianas/imunologia , Clostridioides difficile/imunologia , Enterocolite Pseudomembranosa/imunologia , Lipoproteínas/imunologia , Animais , Toxinas Bacterianas/genética , Toxinas Bacterianas/imunologia , Vacinas Bacterianas/genética , Vacinas Bacterianas/farmacologia , Chlorocebus aethiops , Clostridioides difficile/genética , Cricetinae , Enterocolite Pseudomembranosa/genética , Enterocolite Pseudomembranosa/patologia , Enterocolite Pseudomembranosa/prevenção & controle , Enterotoxinas/genética , Enterotoxinas/imunologia , Lipoproteínas/genética , Lipoproteínas/farmacologia , Camundongos , Camundongos Endogâmicos BALB C , Coelhos , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/imunologia , Proteínas Recombinantes de Fusão/farmacologia , Células Vero
18.
Artigo em Inglês | MEDLINE | ID: mdl-26067816

RESUMO

A core platform technology for high production of recombinant lipoproteins with built-in immunostimulator for novel subunit vaccine development has been established. This platform technology has the following advantages: (1) easily convert antigen into lipidated recombinant protein using a fusion sequence containing lipobox and express high level (50-150mg/L) in Escherichia coli; (2) a robust high-yield up- and downstream bioprocess for lipoprotein production is successfully developed to devoid endotoxin contamination; (3) the lipid moiety of recombinant lipoproteins, which is identical to that of bacterial lipoproteins is recognized as danger signals by the immune system (Toll-like receptor 2 agonist), so both innate and adaptive immune responses can be induced by lipoproteins; and (4) successfully demonstrate the feasibility and safety of this core platform technology in meningococcal group B subunit vaccine, dengue subunit vaccine, novel subunit vaccine against Clostridium difficile-associated diseases, and HPV-based immunotherapeutic vaccines in animal model studies.


Assuntos
Adjuvantes Imunológicos , Lipoproteínas/imunologia , Vacinas/imunologia , Humanos
19.
Hum Vaccin Immunother ; 11(11): 2688-704, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26009802

RESUMO

Enterovirus A infections are the primary cause of hand, foot and mouth disease (HFMD) in infants and young children. Although enterovirus 71 (EV-A71) and coxsackievirus A16 (CV-A16) are the predominant causes of HFMD epidemics worldwide, EV-A71 has emerged as a major neurovirulent virus responsible for severe neurological complications and fatal outcomes. HFMD is a serious health threat and economic burden across the Asia-Pacific region. Inactivated EV-A71 vaccines have elicited protection against EV-A71 but not against CV-A16 infections in large efficacy trials. The current development of a bivalent inactivated EV-A71/CV-A16 vaccine is the next step toward that of multivalent HFMD vaccines. These vaccines should ultimately include other prevalent pathogenic coxsackieviruses A (CV-A6 and CV-A10), coxsackieviruses B (B3 and B5) and echovirus 30 that often co-circulate during HFMD epidemics and can cause severe HFMD, aseptic meningitis and acute viral myocarditis. The prospect and challenges for the development of such multivalent vaccines are discussed.


Assuntos
Enterovirus Humano A/imunologia , Enterovirus/imunologia , Doença de Mão, Pé e Boca/prevenção & controle , Imunização/métodos , Vacinas Virais/administração & dosagem , Vacinas Virais/imunologia , Animais , Descoberta de Drogas/métodos , Descoberta de Drogas/tendências , Doença de Mão, Pé e Boca/imunologia , Humanos
20.
PLoS Negl Trop Dis ; 9(4): e0003692, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25855976

RESUMO

Enterovirus 71 (EV71) and coxsackieviruses (CV) are the major causative agents of hand, foot and mouth disease (HFMD). There is not currently a vaccine available against HFMD, even though a newly developed formalin-inactivated EV71 (FI-EV71) vaccine has been tested in clinical trial and has shown efficacy against EV71. We have designed and genetically engineered a recombinant adenovirus Ad-EVVLP with the EV71 P1 and 3CD genes inserted into the E1/E3-deleted adenoviral genome. Ad-EVVLP were produced in HEK-293A cells. In addition to Ad-EVVLP particles, virus-like particles (VLPs) formed from the physical association of EV71 capsid proteins, VP0, VP1, and VP3 expressed from P1 gene products. They were digested by 3CD protease and confirmed to be produced by Ad-EVVLP-producing cells, as determined using transmission electron microscopy and western blotting. Mouse immunogenicity studies showed that Ad-EVVLP-immunized antisera neutralized the EV71 B4 and C2 genotypes. Activation of VLP-specific CD4+ and CD8+/IFN-γ T cells associated with Th1/Th2-balanced IFN-ɣ, IL-17, IL-4, and IL-13 was induced; in contrast, FI-EV71 induced only Th2-mediated neutralizing antibody against EV71 and low VLP-specific CD4+ and CD8+ T cell responses. The antiviral immunity against EV71 was clearly demonstrated in mice vaccinated with Ad-EVVLP in a hSCARB2 transgenic (hSCARB2-Tg) mouse challenge model. Ad-EVVLP-vaccinated mice were 100% protected and demonstrated reduced viral load in both the CNS and muscle tissues. Ad-EVVLP successfully induced anti-CVA16 immunities. Although antisera had no neutralizing activity against CVA16, the 3C-specific CD4+ and CD8+/IFN-γ T cells were identified, which could mediate protection against CVA16 challenge. FI-EV71 did not induce 3C-mediated immunity and had no efficacy against the CVA16 challenge. These results suggest that Ad-EVVLP can enhance neutralizing antibody and protective cellular immune responses to prevent EV71 infection and cellular immune responses against CV infection.


Assuntos
Proteínas do Capsídeo/imunologia , Enterovirus Humano A/imunologia , Doença de Mão, Pé e Boca/prevenção & controle , Vacinas Virais/imunologia , Animais , Anticorpos Neutralizantes/imunologia , Feminino , Engenharia Genética , Doença de Mão, Pé e Boca/virologia , Soros Imunes/imunologia , Interleucina-13/imunologia , Interleucina-17 , Interleucina-4/imunologia , Camundongos , Camundongos Transgênicos , Vacinas de Produtos Inativados/imunologia , Vacinas Sintéticas/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA