Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 127
Filtrar
1.
Viruses ; 13(11)2021 10 26.
Artigo em Inglês | MEDLINE | ID: mdl-34834964

RESUMO

A transmissible gastroenteritis virus (TGEV) is a porcine enteropathogenic coronavirus, causing acute swine enteric disease especially in suckling piglets. Mesoporous silica nanoparticles (MSNs) are safe vaccine adjuvant, which could enhance immune responses. Our previous research confirmed that nano silicon had immune-enhancing effects with inactivated TGEV vaccine. In this study, we further clarified the immune-enhancing mechanism of the inactivated TGEV vaccine with MSNs on porcine dendritic cells (DCs). Our results indicated that the inactivated TGEV vaccine with MSNs strongly enhanced the activation of the DCs. Expressions of TLR3, TLR5, TLR7, TLR9, and TLR10, cytokines IFN-α, IL-1ß, IL-6, IL-12, and TNF-α, cytokine receptor CCR-7 of immature DCs were characterized and showed themselves to be significantly higher in the inactivated TGEV vaccine with the MSN group. In summary, the inactivated TGEV vaccine with MSNs has effects on the phenotype and function of porcine DCs, which helps to better understand the immune-enhancing mechanism.


Assuntos
Citocinas/metabolismo , Células Dendríticas/imunologia , Gastroenterite Suína Transmissível/imunologia , Gastroenterite Suína Transmissível/prevenção & controle , Receptores Toll-Like/metabolismo , Vírus da Gastroenterite Transmissível/imunologia , Vacinas de Produtos Inativados/imunologia , Adjuvantes de Vacinas/uso terapêutico , Animais , Citocinas/imunologia , Células Dendríticas/citologia , Feminino , Imunidade Inata , Nanopartículas/uso terapêutico , Fenótipo , Silício/uso terapêutico , Suínos , Receptores Toll-Like/imunologia , Vacinas de Produtos Inativados/uso terapêutico
2.
Vet Microbiol ; 252: 108930, 2021 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-33290999

RESUMO

Our previous research obtained purified recombinant porcine interferon-α (rPoIFN-α) containing thioredoxin (Trx) fusion tag in E. coli Rosetta (DE3). Here, we evaluate the efficacy of this rPoIFN-α to prevent piglets from the infection of the transmissible gastroenteritis virus (TGEV) attack. In this experiment, twenty-five TGEV-seronegative piglets were randomly divided into five groups. Group 1 was positive control and only challenged with TGEV; Pigs in groups 2-4 were pretreated with 2 × 10(7)IU/pig, 2 × 10(6)IU/pig, and 2 × 10(5)IU/pig rPoIFN-α before TGEV challenge. The fifth group is a negative control group. The animals of this group are pretreated only with Trx protein-containing PBS solution without TGEV challenge. After 48 h of rPoIFN-α pretreatment, the pigs in groups 1-4 were challenged by TGEV, and the pigs in group 5 were administered with PBS. The surveillance results show that Pigs pre-treated with 2 × 10 (7) IU/pig rPoIFN-α are fully aligned with the violent TGEV attack. Pigs pretreated with 2 × 10 (6) IU/pig rPoIFN-α are partially aligned with the violent TGEV attack. Though piglets pretreated with 2 × 10(6) IU/pig or 2 × 10(5)IU/pig rPoIFN-α cannot be adapted to the challenge of TGEV. However, the use of this dose of rPoIFN-α could put off the clinical signs of pigs than the positive control group of the above. These results indicate that rPoIFN-α can protect pigs from the infection of potential TGEV or delay the appearance of clinical symptoms, and its effect is dose-dependent.


Assuntos
Escherichia coli/genética , Gastroenterite Suína Transmissível/prevenção & controle , Interferon-alfa/metabolismo , Vírus da Gastroenterite Transmissível/imunologia , Animais , Escherichia coli/isolamento & purificação , Gastroenterite Suína Transmissível/virologia , Interferon-alfa/genética , Proteínas Recombinantes , Suínos
3.
Virol J ; 17(1): 163, 2020 10 23.
Artigo em Inglês | MEDLINE | ID: mdl-33097081

RESUMO

BACKGROUND: Transmissible gastroenteritis virus (TGEV) causes enteric infection in piglets, characterized by vomiting, severe diarrhea and dehydration, and the mortality in suckling piglets is often high up to 100%. Vaccination is an effective measure to control the disease caused by TGEV. METHODS: In this study, cell-cultured TGEV HN-2012 strain was inactivated by formaldehyde (FA), ß-propiolactone (BPL) or binaryethylenimine (BEI), respectively. Then the inactivated TGEV vaccine was prepared with freund's adjuvant, and the immunization effects were evaluated in mice. The TGEV-specific IgG level was detected by ELISA. The positive rates of CD4+, CD8+, CD4+IFN-γ+, CD4+IL-4+ T lymphocytes were detected by flow cytometry assay. Lymphocyte proliferation assay and gross pathology and histopathology examination were also performed to assess the three different inactivating reagents in formulating TGEV vaccine. RESULTS: The results showed that the TGEV-specific IgG level in FA group (n = 17) was earlier and stronger, while the BEI group produced much longer-term IgG level. The lymphocyte proliferation test demonstrated that the BEI group had a stronger ability to induce spleen lymphocyte proliferation. The positive rates of CD4+ and CD8+ T lymphocyte subsets of peripheral blood lymphocyte in BEI group was higher than that in FA group and BPL groups by flow cytometry assay. The positive rate of CD4+IFN-γ+ T lymphocyte subset was the highest in the BPL group, and the positive rate of CD4+IL-4+ T lymphocyte subset was the highest in the FA group. There were no obvious pathological changes in the vaccinated mice and the control group after the macroscopic and histopathological examination. CONCLUSIONS: These results indicated that all the three experimental groups could induce cellular and humoral immunity, and the FA group had the best humoral immunity effect, while the BEI group showed its excellent cellular immunity effect.


Assuntos
Anticorpos Antivirais/sangue , Gastroenterite Suína Transmissível/prevenção & controle , Indicadores e Reagentes/farmacologia , Vírus da Gastroenterite Transmissível/efeitos dos fármacos , Vacinas Virais/imunologia , Inativação de Vírus/efeitos dos fármacos , Animais , Feminino , Imunidade Celular , Imunidade Humoral , Imunoglobulina G/sangue , Indicadores e Reagentes/classificação , Ativação Linfocitária , Camundongos , Camundongos Endogâmicos BALB C , Suínos , Linfócitos T/imunologia , Vacinas de Produtos Inativados/administração & dosagem , Vacinas de Produtos Inativados/imunologia , Vacinas Virais/administração & dosagem
4.
Elife ; 92020 09 02.
Artigo em Inglês | MEDLINE | ID: mdl-32876563

RESUMO

Porcine reproductive and respiratory syndrome virus (PRRSV) and transmissible gastroenteritis virus (TGEV) are two highly infectious and lethal viruses causing major economic losses to pig production. Here, we report generation of double-gene-knockout (DKO) pigs harboring edited knockout alleles for known receptor proteins CD163 and pAPN and show that DKO pigs are completely resistant to genotype 2 PRRSV and TGEV. We found no differences in meat-production or reproductive-performance traits between wild-type and DKO pigs, but detected increased iron in DKO muscle. Additional infection challenge experiments showed that DKO pigs exhibited decreased susceptibility to porcine deltacoronavirus (PDCoV), thus offering unprecedented in vivo evidence of pAPN as one of PDCoV receptors. Beyond showing that multiple gene edits can be combined in a livestock animal to achieve simultaneous resistance to two major viruses, our study introduces a valuable model for investigating infection mechanisms of porcine pathogenic viruses that exploit pAPN or CD163 for entry.


Pig epidemics are the biggest threat to the pork industry. In 2019 alone, hundreds of billions of dollars worldwide were lost due to various pig diseases, many of them caused by viruses. The porcine reproductive and respiratory virus (PRRS virus for short), for instance, leads to reproductive disorders such as stillbirths and premature labor. Two coronaviruses ­ the transmissible gastroenteritis virus (or TGEV) and the porcine delta coronavirus ­ cause deadly diarrhea and could potentially cross over into humans. Unfortunately, there are still no safe and effective methods to prevent or control these pig illnesses, but growing disease-resistant pigs could reduce both financial and animal losses. Traditionally, breeding pigs to have a particular trait is a slow process that can take many years. But with gene editing technology, it is possible to change or remove specific genes in a single generation of animals. When viruses infect a host, they use certain proteins on the surface of the host's cells to find their inside: the PRRS virus relies a protein called CD163, and TGEV uses pAPN. Xu, Zhou, Mu et al. used gene editing technology to delete the genes that encode the CD163 and pAPN proteins in pigs. When the animals were infected with PRRS virus or TGEV, the non-edited pigs got sick but the gene-edited animals remained healthy. Unexpectedly, pigs without CD163 and pAPN also coped better with porcine delta coronavirus infections, suggesting that CD163 and pAPN may also help this coronavirus infect cells. Finally, the gene-edited pigs reproduced and produced meat as well as the control pigs. These experiments show that gene editing can be a powerful technology for producing animals with desirable traits. The gene-edited pigs also provide new knowledge about how porcine viruses infect pigs, and may offer a starting point to breed disease-resistant animals on a larger scale.


Assuntos
Antígenos CD13/deficiência , Infecções por Coronavirus/prevenção & controle , Coronavirus/patogenicidade , Gastroenterite Suína Transmissível/prevenção & controle , Síndrome Respiratória e Reprodutiva Suína/prevenção & controle , Vírus da Síndrome Respiratória e Reprodutiva Suína/patogenicidade , Receptores de Superfície Celular/deficiência , Vírus da Gastroenterite Transmissível/patogenicidade , Animais , Animais Geneticamente Modificados , Antígenos CD/genética , Antígenos CD/imunologia , Antígenos de Diferenciação Mielomonocítica/genética , Antígenos de Diferenciação Mielomonocítica/imunologia , Composição Corporal , Antígenos CD13/genética , Antígenos CD13/imunologia , Coronavirus/imunologia , Infecções por Coronavirus/genética , Infecções por Coronavirus/imunologia , Infecções por Coronavirus/virologia , Suscetibilidade a Doenças , Gastroenterite Suína Transmissível/genética , Gastroenterite Suína Transmissível/imunologia , Gastroenterite Suína Transmissível/virologia , Técnicas de Silenciamento de Genes , Interações entre Hospedeiro e Microrganismos , Indústria de Embalagem de Carne , Fenótipo , Síndrome Respiratória e Reprodutiva Suína/genética , Síndrome Respiratória e Reprodutiva Suína/imunologia , Síndrome Respiratória e Reprodutiva Suína/virologia , Vírus da Síndrome Respiratória e Reprodutiva Suína/imunologia , Receptores de Superfície Celular/genética , Receptores de Superfície Celular/imunologia , Sus scrofa/genética , Suínos , Vírus da Gastroenterite Transmissível/imunologia , Aumento de Peso
5.
J Microbiol Biotechnol ; 30(4): 515-525, 2020 04 28.
Artigo em Inglês | MEDLINE | ID: mdl-31838830

RESUMO

Interferon (IFN)-λ plays an essential role in mucosal cells which exhibit strong antiviral activity. Lactobacillus plantarum (L. plantarum) has substantial application potential in the food and medical industries because of its probiotic properties. Alphacoronaviruses, especially porcine epidemic diarrhea virus (PEDV) and transmissible gastroenteritis virus (TGEV), cause high morbidity and mortality in piglets resulting in economic loss. Co-infection by these two viruses is becoming increasingly frequent. Therefore, it is particularly important to develop a new drug to prevent diarrhea infected with mixed viruses in piglets. In this study, we first constructed an anchored expression vector with CWA (C-terminal cell wall anchor) on L. plantarum. Second, we constructed two recombinant L. plantarum strains that anchored IFN-λ3 via pgsA (N-terminal transmembrane anchor) and CWA. Third, we demonstrated that both recombinant strains possess strong antiviral effects against coronavirus infection in the intestinal porcine epithelial cell line J2 (IPEC-J2). However, recombinant L. plantarum with the CWA anchor exhibited a more powerful antiviral effect than recombinant L. plantarum with pgsA. Consistent with this finding, Lb.plantarum-pSIP-409-IFN-λ3-CWA enhanced the expression levels of IFN-stimulated genes (ISGs) (ISG15, OASL, and Mx1) in IPEC-J2 cells more than did recombinant Lb.plantarum-pSIP-409-pgsA'-IFN-λ3. Our study verifies that recombinant L. plantarum inhibits PEDV and TGEV infection in IPEC-J2 cells, which may offer great potential for use as a novel oral antiviral agent in therapeutic applications for combating porcine epidemic diarrhea and transmissible gastroenteritis. This study is the first to show that recombinant L. plantarum suppresses PEDV and TGEV infection of IPEC-J2 cells.


Assuntos
Infecções por Coronavirus/veterinária , Gastroenterite Suína Transmissível/prevenção & controle , Interferons/administração & dosagem , Lactobacillus plantarum/genética , Doenças dos Suínos/prevenção & controle , Animais , Infecções por Coronavirus/imunologia , Infecções por Coronavirus/prevenção & controle , Infecções por Coronavirus/virologia , Células Epiteliais/imunologia , Células Epiteliais/virologia , Feminino , Gastroenterite Suína Transmissível/genética , Gastroenterite Suína Transmissível/imunologia , Gastroenterite Suína Transmissível/virologia , Expressão Gênica , Interferons/genética , Interferons/imunologia , Lactobacillus plantarum/metabolismo , Masculino , Vírus da Diarreia Epidêmica Suína/fisiologia , Suínos , Doenças dos Suínos/genética , Doenças dos Suínos/imunologia , Doenças dos Suínos/virologia , Vírus da Gastroenterite Transmissível/fisiologia
6.
Sci Rep ; 9(1): 13186, 2019 Sep 12.
Artigo em Inglês | MEDLINE | ID: mdl-31515498

RESUMO

Swine enteric diseases have caused significant economic loss and have been considered as the major threat to the global swine industry. Several coronaviruses, including transmissible gastroenteritis virus (TGEV) and porcine epidemic diarrhea virus (PEDV), have been identified as the causative agents of these diseases. Effective measures to control these diseases are lacking. The major host cells of transmissible gastroenteritis virus and porcine epidemic diarrhea virus have thought to be epithelial cells on small intestine villi. Aminopeptidase-N (APN) has been described as the putative receptor for entry of transmissible gastroenteritis virus and porcine epidemic diarrhea virus into cells in vitro. Recently, Whitworth et al. have reported that APN knockout pigs are resistant to TGEV but not PEDV after weaning. However, it remains unclear if APN-null neonatal pigs are protected from TGEV. Here we report the generation of APN-null pigs by using CRISPR/Cas9 technology followed by somatic cell nuclear transfer. APN-null pigs are produced with normal pregnancy rate and viability, indicating lack of APN is not embryonic lethal. After viral challenge, APN-null neonatal piglets are resistant to highly virulent transmissible gastroenteritis virus. Histopathological analyses indicate APN-null pigs exhibit normal small intestine villi, while wildtype pigs show typical lesions in small intestines. Immunochemistry analyses confirm that no transmissible gastroenteritis virus antigen is detected in target tissues in APN-null piglets. However, upon porcine epidemic diarrhea virus challenge, APN-null pigs are still susceptible with 100% mortality. Collectively, this report provides a viable tool for producing animals with enhanced resistance to TGEV and clarifies that APN is dispensable for the PEDV infection in pigs.


Assuntos
Animais Geneticamente Modificados , Antígenos CD13/deficiência , Infecções por Coronavirus , Gastroenterite Suína Transmissível , Vírus da Diarreia Epidêmica Suína/metabolismo , Suínos , Vírus da Gastroenterite Transmissível/metabolismo , Animais , Animais Geneticamente Modificados/genética , Animais Geneticamente Modificados/metabolismo , Animais Geneticamente Modificados/virologia , Antígenos CD13/metabolismo , Infecções por Coronavirus/enzimologia , Infecções por Coronavirus/genética , Infecções por Coronavirus/virologia , Gastroenterite Suína Transmissível/enzimologia , Gastroenterite Suína Transmissível/genética , Gastroenterite Suína Transmissível/prevenção & controle , Gastroenterite Suína Transmissível/virologia , Vírus da Diarreia Epidêmica Suína/genética , Suínos/genética , Suínos/metabolismo , Suínos/virologia , Vírus da Gastroenterite Transmissível/genética
7.
J Vet Med Sci ; 81(10): 1438-1444, 2019 Oct 18.
Artigo em Inglês | MEDLINE | ID: mdl-31474664

RESUMO

Transmissible gastroenteritis (TGE), caused by transmissible gastroenteritis virus (TGEV), is a highly infectious disease in pigs. Vaccination is an effective approach to prevent TGEV infection. Here, we evaluated the potential of TGEV S1 as a DNA vaccine and porcine interleukin (pIL)-12 as an adjuvant in a mouse model. A DNA vaccine was constructed with the TGEV S1 gene to induce immune response in an experimental mouse model; pIL-12 was chosen as the immunological adjuvant within this DNA vaccine. The pVAX1-(TGEV-S1) and pVAX1-(pIL-12) vectors were transfected into BHK-21 cells and expressed in vitro. Experimental mice were separately immunized with each of the recombinant plasmids and controls through the intramuscular route. The lymphocytes isolated from the blood and spleen were analyzed for proliferation, cytotoxic activities, and populations of CD4+ and CD8+ cells. The titers of TGEV S1 in an enzyme-linked immunosorbent assay (ELISA) and TGEV neutralizing antibodies and the concentrations of interferon (IFN)-γ and IL-4 were also analyzed in the serum. The plasmids pVAX1-(TGEV-S1) and pVAX1-(pIL-12) could be expressed in BHK-21 cells, and the combination of pVAX1-(TGEV-S1) and pVAX1-(pIL-12) could induce a significant increase in all markers. pIL-12 could act as an immunological adjuvant in the DNA vaccine for TGEV-S1. Furthermore, the DNA vaccine prepared using TGEV-S1 and porcine IL-12 could induce excellent humoral and cellular immune responses.


Assuntos
Interleucina-12/imunologia , Plasmídeos/imunologia , Glicoproteína da Espícula de Coronavírus/imunologia , Vacinas de DNA/virologia , Adjuvantes Imunológicos , Animais , Gastroenterite Suína Transmissível/prevenção & controle , Imunidade Celular , Imunidade Humoral , Interleucina-12/genética , Camundongos , Modelos Animais , Glicoproteína da Espícula de Coronavírus/genética , Suínos , Vírus da Gastroenterite Transmissível/genética , Vírus da Gastroenterite Transmissível/imunologia , Vacinas de DNA/imunologia , Vacinas Virais/imunologia
8.
Viruses ; 11(8)2019 07 25.
Artigo em Inglês | MEDLINE | ID: mdl-31349683

RESUMO

Porcine epidemic diarrhea virus (PEDV) is an enteric coronavirus causing high morbidity and mortality in porcine herds worldwide. Although both inactivated and live attenuated vaccines have been extensively used, the emergence of highly virulent strains and the recurrent outbreaks even in vaccinated farms highlight the need of effective vaccines. Engineering of genetically defined live attenuated vaccines is a rational approach for novel vaccine development. In this line, we engineered an attenuated virus based on the transmissible gastroenteritis virus (TGEV) genome, expressing a chimeric spike protein from a virulent United States (US) PEDV strain. This virus (rTGEV-RS-SPEDV) was attenuated in highly-sensitive five-day-old piglets, as infected animals did not lose weight and none of them died. In addition, the virus caused very minor tissue damage compared with a virulent virus. The rTGEV-RS-SPEDV vaccine candidate was also attenuated in three-week-old animals that were used to evaluate the protection conferred by this virus, compared with the protection induced by infection with a virulent PEDV US strain (PEDV-NVSL). The rTGEV-RS-SPEDV virus protected against challenge with a virulent PEDV strain, reducing challenge virus titers in jejunum and leading to undetectable challenge virus RNA levels in feces. The rTGEV-RS-SPEDV virus induced a humoral immune response specific for PEDV, including neutralizing antibodies. Altogether, the data indicated that rTGEV-RS-SPEDV is a promising vaccine candidate against virulent PEDV infection.


Assuntos
Gastroenterite Suína Transmissível/prevenção & controle , Vírus da Diarreia Epidêmica Suína/genética , Doenças dos Suínos/prevenção & controle , Vírus da Gastroenterite Transmissível/genética , Vacinas Virais/imunologia , Animais , Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/sangue , Genoma Viral , Vírus da Diarreia Epidêmica Suína/patogenicidade , Recombinação Genética , Suínos , Doenças dos Suínos/imunologia , Doenças dos Suínos/virologia , Estados Unidos , Vacinas Atenuadas/imunologia , Vacinas Sintéticas/imunologia , Vacinas Virais/genética
9.
Anim Sci J ; 90(3): 366-371, 2019 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-30623527

RESUMO

Porcine viral diarrhea is an acute and highly contagious enteric disease in pigs which causes huge economic losses in pig industry worldwide. Transmissible gastroenteritis virus (TGEV) is main pathogens responsible for piglets viral diarrhea. Knockout the host cellular surface receptor for TGEV may be an effective way to accelerate the breeding of resistant pigs. In this study, we applied site-specific editing pAPN which is effective in swine testis (ST) cells. Site-specific editing of pAPN reduced TGEV proliferation in ST cells by 96%-99% at different time periods post-infection. Next, the site-specific editing of pAPN porcine fetal fibroblasts were produced, and then the cell colonies were used as donor cells to generate the site-specific editing of pAPN pigs. Our research findings will not only offer a more thorough understanding of the pathogenesis of piglet diarrhea and lay the foundation for breeding TGEV-resistant piglets, but also understanding the molecular mechanisms involved in coronaviral infections.


Assuntos
Cruzamento/métodos , Antígenos CD13/genética , Diarreia/prevenção & controle , Diarreia/veterinária , Gastroenterite Suína Transmissível/prevenção & controle , Edição de Genes/métodos , Técnicas de Inativação de Genes/métodos , Receptores Virais/genética , Doenças dos Suínos/prevenção & controle , Suínos , Animais , Diarreia/virologia , Fibroblastos/enzimologia , Gastroenterite Suína Transmissível/virologia , Masculino , Doenças dos Suínos/virologia , Testículo/citologia , Testículo/virologia , Vírus da Gastroenterite Transmissível/patogenicidade , Vírus da Gastroenterite Transmissível/fisiologia , Replicação Viral
10.
Viruses ; 9(10)2017 09 25.
Artigo em Inglês | MEDLINE | ID: mdl-28946696

RESUMO

Porcine transmissible gastroenteritis virus (TGEV) and porcine epidemic diarrhea virus (PEDV) are the causative agents of highly fatal acute diarrhea in pigs, resulting in enormous losses in the pig industry worldwide. To develop an effective bivalent oral vaccine against TGEV and PEDV infection, the D antigenic site of the TGEV spike (S) protein and the major antigen site (core neutralizing epitope-COE) of the PEDV S protein were used as immunogens, and the enhanced green fluorescent protein (eGFP) gene was used as a reporter to construct genetically engineered Lactobacillus casei rLpPGF-T7g10-eGFP-6D-COE. The expression of proteins of interest by the recombinant L. casei was confirmed by confocal laser scanning microscopy and a Western blot assay, and the immunogenicity of rLpPGF-T7g10-eGFP-6D-COE in orally immunized mice was evaluated. The results showed that levels of anti-PEDV and anti-TGEV serum immunoglobulin G (IgG) and mucosal secreted immunoglobulin A (sIgA) antibodies obtained from the mice immunized with rLpPGF-T7g10-eGFP-6D-COE, as well as the proliferation levels of lymphocytes, were significantly higher than those in mice orally administered phosphate-buffered saline (PBS) or rLpPG-T7g10. Moreover, the serum IgG antibodies showed neutralizing effects against PEDV and TGEV. Our data suggest that the antibiotic resistance-free genetically engineered L. casei bivalent oral vaccine provides a safe and promising strategy for vaccine development against PEDV and TGEV.


Assuntos
Gastroenterite Suína Transmissível/prevenção & controle , Imunogenicidade da Vacina , Lacticaseibacillus casei/imunologia , Vírus da Diarreia Epidêmica Suína/imunologia , Vírus da Gastroenterite Transmissível/imunologia , Vacinas Virais/imunologia , Animais , Antibacterianos/farmacologia , Proliferação de Células , Células Cultivadas , Resistência Microbiana a Medicamentos , Feminino , Engenharia Genética , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Imunização , Imunoglobulina A Secretora/sangue , Imunoglobulina G/sangue , Lacticaseibacillus casei/efeitos dos fármacos , Lacticaseibacillus casei/genética , Linfócitos/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Testes de Neutralização , Vírus da Diarreia Epidêmica Suína/genética , Suínos , Vírus da Gastroenterite Transmissível/genética , Vacinas Sintéticas/administração & dosagem , Vacinas Sintéticas/genética , Vacinas Sintéticas/imunologia , Proteínas Virais/genética , Proteínas Virais/imunologia , Vacinas Virais/administração & dosagem , Vacinas Virais/genética
11.
Vet Immunol Immunopathol ; 172: 1-8, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-27032496

RESUMO

Transmissible gastroenteritis virus (TGEV) replicates in the small intestine and induces enteritis and watery diarrhea. Establishment of local immunity in the intestine would thus prevent TGEV transmission. CpG DNA has been reported as a promising mucosal adjuvant in some animals. The effects of oral immunization of CpG DNA together with inactivated TGEV (ITGEV) were investigated in this study. Pigs (6 weeks old) were orally immunized with ITGEV plus CpG DNA. The TGEV-specific IgA level in the intestinal tract and the TGEV-specific IgG level in serum significantly increased following immunization with ITGEV plus CpG DNA (P ≤ 0.05). Moreover, populations of IgA-secreting cells, CD3+ T lymphocytes and intraepithelial lymphocytes (IELs), in the intestine increased significantly after immunization with ITGEV plus CpG DNA (P ≤ 0.05). Furthermore, the expression of IL-6, IL-12 and interferon-γ (IFN-γ) in ligated intestine segments increased significantly after injection with ITGEV plus CpG DNA (P ≤ 0.05). Taken together, these data suggest that oral immunization of ITGEV plus CpG DNA elicits a local immune response. Further studies are required to determine whether this immunity provides protection against TGEV in pigs.


Assuntos
Adjuvantes Imunológicos , Suínos , Vírus da Gastroenterite Transmissível/imunologia , Vacinas de DNA/imunologia , Vacinas Virais/imunologia , Adjuvantes Imunológicos/administração & dosagem , Administração Oftálmica , Animais , Anticorpos Antivirais/imunologia , Nucleotídeos de Citosina/administração & dosagem , Nucleotídeos de Citosina/imunologia , Sequência Rica em GC/imunologia , Gastroenterite Suína Transmissível/imunologia , Gastroenterite Suína Transmissível/prevenção & controle , Nucleotídeos de Guanina/administração & dosagem , Nucleotídeos de Guanina/imunologia , Imunoglobulina A/imunologia , Imunoglobulina A/metabolismo , Interferon gama/biossíntese , Interleucina-12/biossíntese , Interleucina-6/biossíntese , Intestinos/imunologia , Doenças dos Suínos/imunologia , Doenças dos Suínos/prevenção & controle , Linfócitos T/imunologia , Vacinas de Produtos Inativados/administração & dosagem , Vacinas de Produtos Inativados/imunologia , Vacinas Virais/administração & dosagem
12.
Sci Rep ; 6: 24152, 2016 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-27080036

RESUMO

The digestive tract is the entry site for transmissible gastroenteritis virus (TGEV). TGEV transmission can be prevented if local immunity is established with increased lymphocytes. The current parenteral mode of vaccination stimulates systemic immunity well, but it does not induce sufficient mucosal immunity. Retinoic acid (RA) plays an important role in the induction of cells that imprint gut-homing molecules. We examined whether RA assist parenteral vaccination of pigs could improve mucosal immunity. We demonstrated that elevated numbers of gut-homing CD8(+) T cells (which express α4ß7 and CCR9 molecules) were presented in porcine inguinal lymph nodes and were recruited to the small intestine by RA. Intestinal mucosal immunity (IgA titre) and systemic immunity (serum IgG titre) were enhanced by RA. Therefore, we hypothesized that RA could induce DCs to form an immature mucosal phenotype and could recruit them to the small intestinal submucosa. Porcine T-cells expressed ß7 integrin and CCR9 receptors and migrated to CCL25 by a mechanism that was dependent of activation by RA-pretreated DCs, rather than direct activation by RA. Together, our results provide powerful evidence that RA can assist whole inactivated TGEV (WI-TGEV) via subcutaneous (s.c.) immunization to generate intestinal immunity, and offer new vaccination strategies against TGEV.


Assuntos
Linfócitos T CD8-Positivos/efeitos dos fármacos , Linfócitos T CD8-Positivos/imunologia , Quimiotaxia de Leucócito/efeitos dos fármacos , Quimiotaxia de Leucócito/imunologia , Trato Gastrointestinal/imunologia , Vírus da Gastroenterite Transmissível/imunologia , Tretinoína/farmacologia , Animais , Linfócitos T CD8-Positivos/metabolismo , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Modelos Animais de Doenças , Gastroenterite Suína Transmissível/prevenção & controle , Imunidade nas Mucosas , Imunoglobulina A Secretora/imunologia , Imunoglobulina G/imunologia , Imunoglobulina G/metabolismo , Mucosa Intestinal/imunologia , Fenótipo , Receptores de Retorno de Linfócitos/metabolismo , Suínos , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo , Vacinas de Produtos Inativados
13.
Antiviral Res ; 131: 74-84, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-26988122

RESUMO

Transmissible gastroenteritis (TGE) causes severe diarrhea in suckling piglets, results in enormous economic loss in swine-producing areas of the world. To develop an effective, safe, and convenient vaccine for the prevention of TGE, we have constructed a recombinant Bacillus subtilis strain (B. subtilis CotGSG) displaying the transmissible gastroenteritis virus (TGEV) spike (S) protein and discussed its immune function to intestinal submucosal dendritic cells (DCs). Our results showed that the recombinant B. subtilis had the ability to recruit more DCs to sample B. subtilis CotGSG, migrate to MLNs, and induce immune responses. Immunized piglets with B. subtilis CotGSG could significantly elevate the specific SIgA titers in feces, IgG titers and neutralizing antibodies in serum. Collectively, our results suggested that recombinant B. subtilis CotGSG expressing the TGEV S protein could effectively induce immune responses via DCs, and provided a perspective on potential novel strategy and approach that may be applicable to the development of the next generation of TGEV vaccines.


Assuntos
Anticorpos Antivirais/sangue , Bacillus subtilis/imunologia , Gastroenterite Suína Transmissível/imunologia , Vírus da Gastroenterite Transmissível/química , Proteínas Virais/imunologia , Vacinas Virais/imunologia , Animais , Anticorpos Neutralizantes/sangue , Antígenos Virais/imunologia , Bacillus subtilis/genética , Movimento Celular , Células Dendríticas/imunologia , Gastroenterite Suína Transmissível/prevenção & controle , Imunoglobulina A Secretora/análise , Imunoglobulina G/sangue , Imunoglobulina G/imunologia , Mucosa Intestinal/citologia , Mucosa Intestinal/imunologia , Mucosa Intestinal/virologia , Suínos , Vírus da Gastroenterite Transmissível/genética , Vírus da Gastroenterite Transmissível/imunologia , Vacinas Sintéticas/imunologia , Proteínas Virais/administração & dosagem , Proteínas Virais/genética , Vacinas Virais/administração & dosagem , Vacinas Virais/genética
14.
Virus Genes ; 52(3): 354-64, 2016 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-26980672

RESUMO

Porcine transmissible gastroenteritis virus (TGEV) and porcine epidemic diarrhea virus (PEDV) can cause severe diarrhea in newborn piglets and led to significant economic losses. The S proteins are the main structural proteins of PEDV and TGEV capable of inducing neutralizing antibodies in vivo. In this study, a DNA vaccine SL7207 (pVAXD-PS1-TS) co-expressing S proteins of TGEV and PEDV delivered by attenuated Salmonella typhimurium was constructed and its immunogenicity in piglets was investigated. Twenty-day-old piglets were orally immunized with SL7207 (pVAXD-PS1-TS) at a dosage of 1.6 × 10(11) CFU per piglet and then booster immunized with 2.0 × 10(11) CFU after 2 weeks. Humoral immune responses, as reflected by virus neutralizing antibodies and specific IgG and sIgA, and cellular immune responses, as reflected by IFN-γ, IL-4, and lymphocyte proliferation, were evaluated. SL7207 (pVAXD-PS1-TS) simultaneously elicited immune responses against TGEV and PEDV after oral immunization. The immune levels started to increase at 2 weeks after immunization and increased to levels statistically significantly different than controls at 4 weeks post-immunization, peaking at 6 weeks and declined at 8 weeks. The humoral, mucosal, and cellular immune responses induced by SL7207 (pAXD-PS1-TS) were significantly higher than those of the PBS and SL7207 (pVAXD) (p < 0.01). In particular, the levels of IFN-γ and IL-4 were higher than those induced by the single-gene vaccine SL7207 (pVAXD-PS1) (p < 0.05). These results demonstrated that SL7207 (pVAXD-PS1-TS) possess the immunological functions of the two S proteins of TGEV and PEDV, indicating that SL7207 (pVAXD-PS1-TS) is a candidate oral vaccine for TGE and PED.


Assuntos
Vírus da Diarreia Epidêmica Suína/imunologia , Salmonella typhimurium/genética , Salmonella typhimurium/imunologia , Vírus da Gastroenterite Transmissível/imunologia , Vacinas de DNA/imunologia , Proteínas Virais/imunologia , Animais , Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/sangue , Anticorpos Antivirais/imunologia , Células COS , DNA Recombinante/genética , DNA Recombinante/imunologia , DNA Viral/genética , DNA Viral/imunologia , Gastroenterite Suína Transmissível/imunologia , Gastroenterite Suína Transmissível/prevenção & controle , Genes Virais , Imunoglobulina G/sangue , Interferon gama/sangue , Interleucina-4/sangue , Vírus da Diarreia Epidêmica Suína/genética , Suínos , Vírus da Gastroenterite Transmissível/genética , Vacinas Atenuadas/administração & dosagem , Vacinas Atenuadas/genética , Vacinas Atenuadas/imunologia , Vacinas de DNA/administração & dosagem , Vacinas de DNA/genética , Proteínas Virais/genética , Vacinas Virais/administração & dosagem , Vacinas Virais/genética , Vacinas Virais/imunologia
15.
Virus Genes ; 52(2): 218-27, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26837896

RESUMO

Attenuated Salmonella typhimurium (S. typhimurium) was selected as a transgenic vehicle for the development of live mucosal vaccines against transmissible gastroenteritis virus (TGEV) based on the M gene. An approximate 1.0 kb DNA fragment, encoding for glycoprotein M, was amplified by RT-PCR and cloned into eukaryotic expression vector pVAX1. The recombinant plasmid pVAX-M was transformed by electroporation into attenuated S. typhimurium SL7207, and the expression and translation of the pVAX-M delivered by recombinant S. typhimurium SL7207 (pVAX-M) was detected both in vitro and in vivo. BALB/c mice were inoculated orally with SL7207 (pVAX-M) at different dosages to evaluate safety of the vaccines. The bacterium was safe to mice at a dosage of 2 × 10(9) CFU, almost eliminated from the spleen and liver at week 4 post-immunization and eventually cleared at week 6. Mice immunized with 1 × 10(9) CFU of SL7207 (pVAX-M) elicited specific anti-TGEV local mucosal and humoral responses including levels of IgA, IgG, IL-4, and IFN-γ as measured by indirect ELISA assay. Moreover, the control groups (pVAX group, PBS group) maintained at a normal level during week 4-8 post-immunization. The results indicated that attenuated S. typhimurium could be used as a delivery vector for oral immunization of TGEV M gene vaccine.


Assuntos
Salmonella typhimurium/genética , Salmonella typhimurium/imunologia , Vacinas Atenuadas/genética , Vacinas Atenuadas/imunologia , Proteínas da Matriz Viral/genética , Proteínas da Matriz Viral/imunologia , Animais , Anticorpos Antivirais/imunologia , Linhagem Celular , Proteínas M de Coronavírus , Citocinas/biossíntese , Modelos Animais de Doenças , Feminino , Gastroenterite Suína Transmissível/prevenção & controle , Expressão Gênica , Células HEK293 , Humanos , Imunidade nas Mucosas , Imunização , Imunoglobulina A/imunologia , Camundongos , Suínos , Transformação Bacteriana , Vírus da Gastroenterite Transmissível/genética , Vírus da Gastroenterite Transmissível/imunologia , Vacinas de DNA , Vacinas Virais
16.
Vaccine ; 33(32): 3900-6, 2015 Jul 31.
Artigo em Inglês | MEDLINE | ID: mdl-26116254

RESUMO

To explore the possibility of developing a vaccine against transmissible gastroenteritis virus (TGEV) infection, a recombinant swinepox virus (rSPV-SA) expressing a TGEV protective antigen has been constructed. Immune responses and protection efficacy of the vaccination vector were assessed in both mice and pig models. An indirect ELISA assay suggested that when mice were vaccinated with rSPV-SA, the level of IgG against TGEV was enhanced dramatically. The cytokine assays were employed and the results indicated that both the Th1-type and Th2-type cytokine levels raised after vaccination with rSPV-SA in mice models. Results from the passive immunity protection test of new born piglets demonstrated that the recombinant live-vector vaccine, rSPV-SA, could 100% protect piglets from the SPV infection, and there was no significant clinical symptom in the rSPV-SA treatment group during this experiment. The data suggest that the novel recombinant swinepox virus is a potential vaccine against TGEV infection.


Assuntos
Portadores de Fármacos , Epitopos/imunologia , Gastroenterite Suína Transmissível/prevenção & controle , Suipoxvirus/genética , Vírus da Gastroenterite Transmissível/imunologia , Vacinas Virais/imunologia , Animais , Anticorpos Antivirais/sangue , Citocinas/metabolismo , Epitopos/genética , Gastroenterite Suína Transmissível/imunologia , Vetores Genéticos , Imunização Passiva , Imunoglobulina G/sangue , Camundongos Endogâmicos BALB C , Suínos , Linfócitos T/imunologia , Vírus da Gastroenterite Transmissível/genética , Resultado do Tratamento , Vacinas Sintéticas/administração & dosagem , Vacinas Sintéticas/genética , Vacinas Sintéticas/imunologia , Vacinas Virais/administração & dosagem , Vacinas Virais/genética
17.
Appl Microbiol Biotechnol ; 98(19): 8301-12, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24993357

RESUMO

The role of muramyl dipeptide (MDP) and tuftsin in oral immune adjustment remains unclear, particularly in a Lactobacillus casei (L. casei) vaccine. To address this, we investigated the effects of different repetitive peptides expressed by L. casei, specifically the MDP and tuftsin fusion protein (MT) repeated 20 and 40 times (20MT and 40MT), in mice also expressing the D antigenic site of the spike (S) protein of transmissible gastroenteritis virus (TGEV) on intestinal and systemic immune responses and confirmed the immunoregulation of these peptides. Treatment of mice with a different vaccine consisting of L. casei expressing MDP and tuftsin stimulated humoral and cellular immune responses. Both 20MT and 40MT induced an increase in IgG and IgA levels against TGEV, as determined using enzyme-linked immunosorbent assay. Increased IgG and IgA resulted in the activation of TGEV-neutralising antibody activity in vitro. In addition, 20MT and 40MT stimulated the differentiation of innate immune cells, including T helper cell subclasses and regulatory T (Treg) cells, which induced robust T helper type 1 and T helper type 17 (Th17) responses and reduced Treg T cell immune responses in the 20MT and 40MT groups, respectively. Notably, treatment of mice with L. casei expressing 20MT and 40MT enhanced the anti-TGEV antibody immune responses of both the humoral and mucosal immune systems. These findings suggest that L. casei expressing MDP and tuftsin possesses substantial immunopotentiating properties, as it can induce humoral and T cell-mediated immune responses upon oral administration, and it may be useful in oral vaccines against TGEV challenge.


Assuntos
Acetilmuramil-Alanil-Isoglutamina/genética , Gastroenterite Suína Transmissível/imunologia , Lacticaseibacillus casei/genética , Células Th1/imunologia , Células Th17/imunologia , Vírus da Gastroenterite Transmissível/imunologia , Tuftsina/genética , Vacinas Virais/imunologia , Acetilmuramil-Alanil-Isoglutamina/administração & dosagem , Acetilmuramil-Alanil-Isoglutamina/imunologia , Administração Oral , Animais , Feminino , Gastroenterite Suína Transmissível/prevenção & controle , Gastroenterite Suína Transmissível/virologia , Lacticaseibacillus casei/imunologia , Masculino , Camundongos , Glicoproteína da Espícula de Coronavírus/administração & dosagem , Glicoproteína da Espícula de Coronavírus/genética , Glicoproteína da Espícula de Coronavírus/imunologia , Suínos , Vírus da Gastroenterite Transmissível/genética , Tuftsina/administração & dosagem , Tuftsina/imunologia , Regulação para Cima , Vacinas Virais/administração & dosagem , Vacinas Virais/genética
18.
PLoS One ; 8(3): e57468, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23526943

RESUMO

Porcine transmissible gastroenteritis virus (TGEV) and porcine epidemic diarrhea virus (PDEV) can cause severe diarrhea in pigs. Development of effective vaccines against TGEV and PEDV is one of important prevention measures. The spike (S) protein is the surface glycoprotein of TGEV and PEDV, which can induce specific neutralization antibodies and is a candidate antigen for vaccination attempts. In this study, the open reading frames of the TGEV S1 protein and in addition of the S or S1 proteins of PEDV were inserted into the eukaryotic expression vector, pIRES, resulting in recombinant plasmids, pIRES-(TGEV-S1-PEDV-S1) and pIRES-(TGEV-S1-PEDV-S). Subsequently, 6-8 weeks old Kunming mice were inoculated with both DNA plasmids. Lymphocyte proliferation assay, virus neutralization assay, IFN-γ assay and CTL activity assay were performed. TGEV/PEDV specific antibody responses as well as kinetic changes of T lymphocyte subgroups of the immunized mice were analyzed. The results showed that the recombinant DNA plasmids increased the proliferation of T lymphocytes and the number of CD4+ and CD8+ T lymphocyte subgroups. In addition, the DNA vaccines induced a high level of IFN-γ in the immunized mice. The specific CTL activity in the pIRES-(TGEV-S1-PEDV-S) group became significant at 42 days post-immunization. At 35 days post-immunization, the recombinant DNA plasmids bearing full-length S genes of TGEV and PEDV stimulated higher levels of specific antibodies and neutralizing antibodies in immunized mice.


Assuntos
Vírus da Diarreia Epidêmica Suína/genética , Vírus da Diarreia Epidêmica Suína/imunologia , Vírus da Gastroenterite Transmissível/genética , Vírus da Gastroenterite Transmissível/imunologia , Vacinas de DNA/genética , Vacinas de DNA/imunologia , Proteínas Virais/genética , Proteínas Virais/imunologia , Vacinas Virais/genética , Vacinas Virais/imunologia , Animais , Anticorpos Antivirais/sangue , Infecções por Coronavirus/imunologia , Infecções por Coronavirus/prevenção & controle , Infecções por Coronavirus/veterinária , DNA Recombinante/genética , DNA Recombinante/imunologia , DNA Viral/genética , DNA Viral/imunologia , Gastroenterite Suína Transmissível/imunologia , Gastroenterite Suína Transmissível/prevenção & controle , Genes Virais , Interferon gama/sangue , Interleucina-4/sangue , Camundongos , Plasmídeos/genética , Plasmídeos/imunologia , Suínos , Doenças dos Suínos/imunologia , Doenças dos Suínos/prevenção & controle , Linfócitos T/imunologia
19.
Arch Virol ; 158(4): 799-807, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23188495

RESUMO

The enteropathogenic coronavirus transmissible gastroenteritis virus (TGEV) causes severe disease in young piglets. We have studied the protective effects of the probiotic Enterococcus faecium NCIMB 10415 (E. faecium), which is approved as a feed additive in the European Union, against TGEV infection. E. faecium was added to swine testicle (ST) cells before, concomitantly with, or after TGEV infection. Viability assays revealed that E. faecium led to a dose-dependent rescue of viability of TGEV-infected cells reaching nearly to complete protection. Virus yields of the E. faecium-treated cultures were reduced by up to three log10 units. Western blot analysis of purified TGEV revealed that the levels of all viral structural proteins were reduced after E. faecium treatment. Using transmission electron microscopy, we observed attachment of TGEV particles to the surface of E. faecium which might be a means to trap virus and to prevent infection. Increased production of nitric oxide in the cells treated with E. faecium and elevated expression of interleukin 6 and 8 pointed to stimulated cellular defense as a mechanism to fight TGEV infection.


Assuntos
Enterococcus faecium/fisiologia , Gastroenterite Suína Transmissível/prevenção & controle , Probióticos , Vírus da Gastroenterite Transmissível , Ração Animal , Animais , Linhagem Celular , Sobrevivência Celular , Citocinas/genética , Citocinas/metabolismo , Dieta/veterinária , Regulação da Expressão Gênica , Masculino , Óxido Nítrico/metabolismo , Suínos , Testículo/citologia , Cultura de Vírus
20.
Hybridoma (Larchmt) ; 30(5): 457-62, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22008073

RESUMO

The entire pig aminopeptidase N (pAPN) gene was amplified by RT-PCR using total RNA extracted from intestinal brush border membrane of a newborn piglet. The amplified products of the pAPN gene were cloned into the vector pMD18-T, generating a recombinant plasmid pMD18-T-pAPN. The C subunit of pAPN (pAPN-C) produced by PCR from the plasmid pMD18-T-pAPN was expressed in Escherichia coli using vector pET-32a with His tag. After confirming reactivity of the recombinant protein pAPN-C to antibody against native pAPN, polyclonal antibody against the recombinant protein pAPN-C was prepared in rabbit using purified protein as immunogen. In Western blot analysis, the antibody elicited by the recombinant protein pAPN-C could recognize the native pAPN. These data demonstrate that the pAPN-C recombinant protein and its polyclonal antibody can provide some basis for further receptor antagonist.


Assuntos
Antígenos CD13/imunologia , Escherichia coli/genética , Soros Imunes , Subunidades Proteicas/imunologia , Proteínas Recombinantes de Fusão/imunologia , Animais , Especificidade de Anticorpos , Antígenos CD13/biossíntese , Antígenos CD13/isolamento & purificação , Clonagem Molecular , Infecções por Coronavirus/prevenção & controle , Ensaio de Imunoadsorção Enzimática , Gastroenterite Suína Transmissível/prevenção & controle , Vetores Genéticos , Reação em Cadeia da Polimerase , Subunidades Proteicas/biossíntese , Subunidades Proteicas/isolamento & purificação , Coelhos , Proteínas Recombinantes de Fusão/biossíntese , Proteínas Recombinantes de Fusão/isolamento & purificação , Suínos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...