Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 357
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
J Virol ; 94(21)2020 10 14.
Artigo em Inglês | MEDLINE | ID: mdl-32796075

RESUMO

The intestinal organoid culture system is a pathbreaking working model for investigating pathogen-host interactions in the intestines. However, due to the limitations of the first generation of intestinal organoids, basal-out structure and growth in Matrigel, most pathogens can rarely attach to the apical membrane directly and hardly initiate infection. In this study, we first developed a next-generation porcine intestinal organoid culture system, characterized by an apical membrane on the surface, called apical-out. To investigate the infectivity and antiviral immune responses of this apical-out porcine intestinal organoid, a swine enteric virus, transmissible gastroenteritis virus (TGEV), was employed to inoculate the culture system. Both reverse transcription-quantitative PCR (RT-qPCR) and immunofluorescence assay (IFA) analysis demonstrated that TGEV replicated in the apical-out porcine intestinal organoid culture system. Additionally, our results illustrated that TGEV infection significantly upregulated the expression levels of alpha interferon (IFN-α), IFN-λ1, interferon-stimulated gene 15 (ISG15), ISG58, tumor necrosis factor alpha (TNF-α), and interleukin 6 (IL-6) in this culture system. Hence, we successfully developed a porcine intestinal apical-out organoid culture system, which will facilitate the investigation of pathogen-host interactions in pig intestines.IMPORTANCE Intestinal organoids are a newly developed culture system for investigating pathogen-host interactions. Intestinal organoid models have been widely used since their development, because the results obtained from this type of culture model better represent physiological conditions than those from well-established cell lines. The three-dimensional (3D) porcine intestinal organoid model was reported in 2018 and 2019 for the investigation of intestinal pathogens. However, those organoid culture models were basal-out intestinal organoids, which are not suitable for porcine enteric virus research because they invade the intestines via the apical side of epithelial cells on villi. In this study, we developed a porcine apical-out intestinal organoid culture system and verified its infectivity, type I and type III interferon (IFN) antiviral responses, and inflammatory responses following infection by a swine enteric virus. Our results imply that this apical-out porcine intestinal organoid culture system is an ideal model for the investigation of interactions between swine enteric viruses and the intestines.


Assuntos
Células Epiteliais/imunologia , Gastroenterite Suína Transmissível/imunologia , Interações Hospedeiro-Patógeno/imunologia , Mucosa Intestinal/imunologia , Organoides/imunologia , Vírus da Gastroenterite Transmissível/imunologia , Animais , Bioensaio , Quimiocina CXCL10/genética , Quimiocina CXCL10/imunologia , Células Epiteliais/patologia , Células Epiteliais/virologia , Gastroenterite Suína Transmissível/genética , Gastroenterite Suína Transmissível/virologia , Regulação da Expressão Gênica , Interações Hospedeiro-Patógeno/genética , Interferon-alfa/genética , Interferon-alfa/imunologia , Interferons/genética , Interferons/imunologia , Interleucina-6/genética , Interleucina-6/imunologia , Mucosa Intestinal/patologia , Mucosa Intestinal/virologia , Organoides/patologia , Organoides/virologia , Suínos , Vírus da Gastroenterite Transmissível/crescimento & desenvolvimento , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/imunologia , Ubiquitinas/genética , Ubiquitinas/imunologia , Replicação Viral
2.
J Gen Virol ; 100(2): 206-216, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30652967

RESUMO

Both porcine epidemic diarrhoea virus (PEDV) and porcine transmissible gastroenteritis virus (TGEV), which cause high mortality in piglets and produce similar clinical symptoms and histopathological morphology, belong to the genus Alphacoronavirus. Serological diagnosis plays an important role in distinguishing pathogen species. Together with the spike (S) protein, the nucleocapsid (N) protein is one of the immunodominant regions among coronaviruses. In this study, two-way antigenic cross-reactivity between the N proteins of PEDV and TGEV was observed by indirect immunofluorescence assay (IFA) and Western blot analysis. Furthermore, the PEDV N protein harbouring truncations of amino acids (aa) 1 to 170 or aa 125 to 301 was demonstrated to cross-react with the anti-TGEV N polyclonal antibody (PAb), whereas the truncation-expressing aa 302 to 401 resulted in a specific reaction with the anti-PEDV N PAb but not with the anti-TGEV N PAb. Mutants of the PEDV N protein were generated based on sequence alignment and structural analysis; we then confirmed that the N-terminal residues 58-RWRMRRGERIE-68 and 78-LGTGPHAD-85 contributed to the cross-reactivity. All the results provide vital clues for the development of precise diagnostic assays for porcine coronaviruses.


Assuntos
Anticorpos Antivirais/imunologia , Reações Cruzadas , Epitopos/imunologia , Proteínas do Nucleocapsídeo/imunologia , Vírus da Diarreia Epidêmica Suína/imunologia , Vírus da Gastroenterite Transmissível/imunologia , Animais , Western Blotting , Mapeamento de Epitopos , Técnica Indireta de Fluorescência para Anticorpo , Suínos
3.
J Virol ; 92(22)2018 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-30185587

RESUMO

In host innate immunity, type I interferons (IFN-I) are major antiviral molecules, and coronaviruses have evolved diverse strategies to counter the IFN-I response during infection. Transmissible gastroenteritis virus (TGEV), a member of the Alphacoronavirus family, induces endoplasmic reticulum (ER) stress and significant IFN-I production after infection. However, how TGEV evades the IFN-I antiviral response despite the marked induction of endogenous IFN-I has remained unclear. Inositol-requiring enzyme 1 α (IRE1α), a highly conserved ER stress sensor with both kinase and RNase activities, is involved in the IFN response. In this study, IRE1α facilitated TGEV replication via downmodulating the host microRNA (miR) miR-30a-5p abundance. miR-30a-5p normally enhances IFN-I antiviral activity by directly targeting the negative regulators of Janus family kinase (JAK)-signal transducer and activator of transcription (STAT), the suppressor of cytokine signaling protein 1 (SOCS1), and SOCS3. Furthermore, TGEV infection increased SOCS1 and SOCS3 expression, which dampened the IFN-I antiviral response and facilitated TGEV replication. Importantly, compared with mock infection, TGEV infection in vivo resulted in decreased miR-30a-5p levels and significantly elevated SOCS1 and SOCS3 expression in the piglet ileum. Taken together, our data reveal a new strategy used by TGEV to escape the IFN-I response by engaging the IRE1α-miR-30a-5p/SOCS1/3 axis, thus improving our understanding of how TGEV escapes host innate immune defenses.IMPORTANCE Type I interferons (IFN-I) play essential roles in restricting viral infections. Coronavirus infection induces ER stress and the interferon response, which reflects different adaptive cellular processes. An understanding of how coronavirus-elicited ER stress is actively involved in viral replication and manipulates the host IFN-I response has remained elusive. Here, TGEV inhibited host miR-30a-5p via the ER stress sensor IRE1α, which led to the increased expression of negative regulators of JAK-STAT signaling cascades, namely, SOCS1 and SOCS3. Increased SOCS1 or SOCS3 expression impaired the IFN-I antiviral response, promoting TGEV replication. These findings enhance our understanding of the strategies used by coronaviruses to antagonize IFN-I innate immunity via IRE1α-mediated manipulation of the miR-30a-5p/SOCS axis, highlighting the crucial role of IRE1α in innate antiviral resistance and the potential of IRE1α as a novel target against coronavirus infection.


Assuntos
Estresse do Retículo Endoplasmático/imunologia , Endorribonucleases/metabolismo , Evasão da Resposta Imune/imunologia , Interferon Tipo I/imunologia , MicroRNAs/genética , Proteína 1 Supressora da Sinalização de Citocina/metabolismo , Proteína 3 Supressora da Sinalização de Citocinas/metabolismo , Vírus da Gastroenterite Transmissível/imunologia , Animais , Linhagem Celular , Estresse do Retículo Endoplasmático/genética , Interações Hospedeiro-Patógeno/imunologia , Evasão da Resposta Imune/genética , Imunidade Inata/imunologia , Janus Quinases/metabolismo , Sus scrofa , Vírus da Gastroenterite Transmissível/genética , Replicação Viral/fisiologia
4.
Virol J ; 16(1): 97, 2019 08 05.
Artigo em Inglês | MEDLINE | ID: mdl-31382996

RESUMO

BACKGROUND: Transmissible gastroenteritis virus (TGEV), a member of the family Coronaviridae, causes lethal watery diarrhea in piglets. Previous studies have revealed that the coronaviruses develop various strategies to evade the host innate immunity through the inhibition of nuclear factor kappa B (NF-κB) signaling pathway. However, the ability of TGEV to inhibit the host innate immune response by modulating the NF-κB signaling pathway is not clear. METHODS: In this study, a dual luciferase reporter assay was used to confirm the inhibition of NF-κB by TGEV infection and to identify the major viral proteins involved in the inhibition of NF-κB signaling. Real-time quantitative PCR was used to quantify the mRNA expression of inflammatory factors. The deubiquitination of Nsp3 domains and its effect on IκBα and p65 were analyzed by western blotting. The ubiquitination level of IκBα was analyzed by immunoprecipitation. RESULTS: In ST and IPEC-J2 cells, TGEV exhibited a dose-dependent inhibition of NF-κB activity. Individual TGEV protein screening revealed the high potential of non-structural protein 3 (Nsp3) to inhibit NF-κB signaling, and leading to the downregulation of the NF-κB-induced cytokine production. We demonstrated that the inhibitory effect of Nsp3 was mainly mediated through the suppression of IκBα degradation as well as the inhibition of p65 phosphorylation and nuclear translocation. Furthermore, the amino acid residues at positions 590-1,215 in Nsp3 were demonstrated to inhibit the degradation of IκBα by inhibiting the IκBα ubiquitination. CONCLUSION: TGEV infection can inhibit the activation of the NF-κB signaling pathway, which is mainly mediated by Nsp3 through the canonical pathway. The amino acid residues at positions 590-1,215 in Nsp3 compose the critical domain that mediates NF-κB inhibition. We speculate that this inhibitory effect is likely to be related to the structure of PLP2 with deubiquitinating enzyme activity of the amino acid residues at positions 590-1,215 in Nsp3. Our study provides a better understanding of the TGEV-mediated innate immune modulation and lays the basis for studies on the pathogenesis of coronavirus.


Assuntos
Gastroenterite Suína Transmissível/imunologia , Evasão da Resposta Imune , Imunidade Inata , NF-kappa B/antagonistas & inibidores , Transdução de Sinais , Vírus da Gastroenterite Transmissível/imunologia , Proteínas não Estruturais Virais/genética , Animais , Linhagem Celular , Regulação para Baixo , Interações entre Hospedeiro e Microrganismos , NF-kappa B/genética , Suínos , Vírus da Gastroenterite Transmissível/fisiologia , Proteínas não Estruturais Virais/imunologia , Replicação Viral
5.
Arch Virol ; 164(4): 983-994, 2019 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-30729994

RESUMO

Transmissible gastroenteritis virus (TGEV) infection causes severe diarrhea in piglets and imposes a significant economic burden on pig farms. Single-chain fragment variable (scFv) antibodies effectively inhibit virus infection and could be a potential therapeutic reagent for preventing disease. In this study, a recombinant scFv antibody phage display library was constructed from peripheral blood lymphocytes of piglets infected with TGEV. The library was screened with four rounds of biopanning using purified TGEV antigen, and scFv antibodies that bound to TGEV were obtained. The scFv gene was subcloned into the pET-28a(+), and the constituted plasmid was introduced into Escherichia coli BL21 (DE3) for protein expression. All three scFv clones identified had neutralizing activity against TGEV. An immunofluorescence assay and western blot analysis demonstrated that two scFv antibodies reacted with the spike protein of TGEV. These results indicate that scFv antibodies provide protection against viral infection in vitro and may be a therapeutic candidate for both prevention and treatment of TGEV infection in swine.


Assuntos
Anticorpos Antivirais/imunologia , Gastroenterite Suína Transmissível/virologia , Anticorpos de Cadeia Única/imunologia , Vírus da Gastroenterite Transmissível/imunologia , Animais , Anticorpos Antivirais/genética , Gastroenterite Suína Transmissível/imunologia , Testes de Neutralização , Anticorpos de Cadeia Única/genética , Suínos , Vírus da Gastroenterite Transmissível/genética
6.
Arch Virol ; 164(11): 2659-2669, 2019 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-31385116

RESUMO

Interferon gamma (IFN-γ) is best known for its ability to regulate host immune responses; however, its direct antiviral activity is less well studied. Transmissible gastroenteritis virus (TGEV) is an economically important swine enteric coronavirus and causes acute diarrhea in piglets. At present, little is known about the function of IFN-γ in the control of TGEV infection. In this study, we demonstrated that IFN-γ inhibited TGEV infection directly in ST cells and intestine epithelial IPEC-J2 cells and that the anti-TGEV activity of IFN-γ was independent of IFN-α/ß. Moreover, IFN-γ suppressed TGEV infection in ST cells more efficiently than did IFN-α, and the combination of IFN-γ and IFN-α displayed a synergistic effect against TGEV. Mechanistically, using overexpression and functional knockdown experiments, we demonstrated that porcine interferon regulatory factor 1 (poIRF1) elicited by IFN-γ primarily mediated IFN-γ signaling cascades and the inhibition of TGEV infection by IFN-γ. Importantly, we found that TGEV elevated the expression of poIRF1 and IFN-γ in infected small intestines and peripheral blood mononuclear cells. Thus, IFN-γ plays a crucial role in curtailing enteric coronavirus infection and may serve as an effective prophylactic and/or therapeutic agent against TGEV infection.


Assuntos
Gastroenterite Suína Transmissível/imunologia , Fator Regulador 1 de Interferon/metabolismo , Interferon gama/imunologia , Vírus da Gastroenterite Transmissível/imunologia , Animais , Linhagem Celular , Chlorocebus aethiops , Fator Regulador 1 de Interferon/genética , Interferon-alfa/imunologia , Interferência de RNA , RNA Interferente Pequeno/genética , Transdução de Sinais/imunologia , Suínos , Células Vero
7.
Appl Microbiol Biotechnol ; 103(1): 437-447, 2019 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-30402771

RESUMO

Recent years have seen extremely fast development of new viral nanovaccines and diagnostic agents using nanostructures prepared by biological and chemical synthesis. We used spherical gold nanoparticles (average diameter, 15 nm) as a platform for the antigen for swine transmissible gastroenteritis virus (TGEV). The literature data demonstrate that immunization of animals with the TGEV antigen coupled to gold nanoparticles (GNPs) not only activates antigen-presenting cells but also increases the proliferative activity of splenic lymphoid (antibody-forming) cells. The contents of γ-IFN, IL-1ß, and IL-6 in animals immunized with GNP-antigen conjugates were found to be higher than those in intact animals or in animals given the antigen alone. The increased concentration of IL-1ß in the immunized animals directly correlated with the activity of macrophages and stimulated B cells, which produce this cytokine when activated. The increased concentration of IL-6 indicates that the injected preparations are stimulatory to cellular immunity. Immunization with the TGEV antigen conjugated to GNPs as a carrier activates the respiratory activity of lymphoid cells and peritoneal macrophages, which is directly related to their transforming activity and to the activation of antibody generation. Furthermore, the use of this conjugate allows marked improvement of the structure of the animals' immune organs and restores the morphological-functional state of these organs. The microanatomical changes (increased number of follicles) indicate the activation of the B-dependent zone of the spleen and, consequently, the development of a humoral-type immunological reaction. The degradative processes observed in the animals immunized with TGEV antigen alone are evidence of weak resistance to pathogen attack. These results can be used to develop vaccines against this infection by employing TGEV antigen coupled to gold nanoparticles as a carrier.


Assuntos
Portadores de Fármacos/farmacologia , Imunização/métodos , Nanopartículas Metálicas/administração & dosagem , Vírus da Gastroenterite Transmissível/imunologia , Animais , Antígenos Virais/química , Portadores de Fármacos/administração & dosagem , Ouro , Cobaias , Interferon gama/metabolismo , Macrófagos Peritoneais/imunologia , Masculino , Nanopartículas Metálicas/química , Baço/citologia , Baço/imunologia , Vírus da Gastroenterite Transmissível/genética , Vírus da Gastroenterite Transmissível/patogenicidade
8.
Appl Microbiol Biotechnol ; 102(19): 8307-8318, 2018 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-30056514

RESUMO

The highly infectious porcine transmissible gastroenteritis virus (TGEV), which belongs to the coronaviruses (CoVs), causes diarrhea and high mortality rates in piglets, resulting in severe economic losses in the pork industry worldwide. In this study, we used Lactobacillus plantarum (L. plantarum) to anchor the expression of TGEV antigen (S) to dendritic cells (DCs) via dendritic cell-targeting peptides (DCpep). The results show that S antigen could be detected on the surface of L. plantarum by different detection methods. Furthermore, flow cytometry and ELISA techniques were used to measure the cellular, mucosal, and humoral immune responses of the different orally gavaged mouse groups. The obtained results demonstrated the significant effect of the constructed L. plantarum expressing S-DCpep fusion proteins in inducing high expression levels of B7 molecules on DCs, as well as high levels of IgG, secretory IgA, and IFN-γ and IL-4 cytokines compared with the other groups. Accordingly, surface expression of DC-targeted antigens successfully induced cellular, mucosal, and humoral immunity in mice and could be used as a vaccine.


Assuntos
Antígenos de Bactérias/imunologia , Lactobacillus plantarum/imunologia , Vírus da Gastroenterite Transmissível/imunologia , Animais , Anticorpos Antivirais/imunologia , Células Dendríticas/imunologia , Imunidade Humoral/imunologia , Imunização/métodos , Imunoglobulina A Secretora/imunologia , Camundongos , Suínos , Vacinação/métodos , Vacinas Virais/imunologia
9.
Appl Microbiol Biotechnol ; 102(19): 8403-8417, 2018 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-30022263

RESUMO

Transmissible gastroenteritis coronavirus (TGEV) is one of the most severe threats to the swine industry. In this study, we constructed a suite of recombinant Lactobacillus plantarum with surface displaying the spike (S) protein coming from TGEV and fused with DC cells targeting peptides (DCpep) to develop an effective, safe, and convenient vaccine against transmissible gastroenteritis. Our research results found that the recombinant Lactobacillus plantarum (NC8-pSIP409-pgsA-S-DCpep) group expressing S fused with DCpep could not only significantly increase the percentages of MHC-II+CD80+ B cells and CD3+CD4+ T cells but also the number of IgA+ B cells and CD3+CD4+ T cells of ileum lamina propria, which elevated the specific secretory immunoglobulin A (SIgA) titers in feces and IgG titers in serum. Taken together, these results suggest that NC8-pSIP409-pgsA-S-DCpep expressing the S of TGEV fused with DCpep could effectively induce immune responses and provide a feasible original strategy and approach for the design of TGEV vaccines.


Assuntos
Proteínas de Bactérias/imunologia , Peptídeos e Proteínas de Sinalização Intracelular/imunologia , Lactobacillus plantarum/imunologia , Vírus da Gastroenterite Transmissível/imunologia , Animais , Anticorpos Antivirais/imunologia , Linfócitos B/imunologia , Gastroenterite Suína Transmissível/imunologia , Imunoglobulina A Secretora/imunologia , Imunoglobulina G/imunologia , Suínos , Linfócitos T/imunologia , Vacinas Virais/imunologia
10.
Nanomedicine ; 14(4): 1201-1212, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29501635

RESUMO

Inactivated transmissible gastroenteritis virus (TGEV) vaccines are widely used in swine herds in China. These are limited, however, by the need to elicit both humoral and cellular immunity, as well as the efficiency of adjuvants. In this study, a 70-nm nano silicon particle was applied with inactivated TGEV vaccine in mice, and its immune-enhancing effects and mechanism of action investigated. We found that nano silicon applied with inactivated TGEV vaccine induced high antibody titers, increase IL-6, TNF-α and IFN-γ expression, and stimulate CD3+ T cell proliferation with a high CD4+/CD8+ T lymphocyte ratio. Nano silicon could quickly activate innate and adaptive immunity by stimulating Toll-like receptor signaling pathways, indicating that the nano silicon adjuvant enhanced long-term humoral and early cellular immune responses when combined with inactivated TGEV vaccine. Nano silicon could be considered for use as an antigen- carrier and adjuvant for veterinary vaccines.


Assuntos
Imunidade Celular/imunologia , Imunidade Humoral/imunologia , Silício/química , Vírus da Gastroenterite Transmissível/imunologia , Vacinas de DNA/imunologia , Vacinas de DNA/uso terapêutico , Adjuvantes Imunológicos , Animais , Linhagem Celular , Ensaio de Imunoadsorção Enzimática , Gastroenterite/imunologia , Gastroenterite/prevenção & controle , Interferon gama/metabolismo , Interleucina-6/metabolismo , Masculino , Camundongos Endogâmicos BALB C , Suínos , Fator de Necrose Tumoral alfa/metabolismo
11.
J Clin Microbiol ; 55(5): 1426-1436, 2017 05.
Artigo em Inglês | MEDLINE | ID: mdl-28202790

RESUMO

The development of porcine epidemic diarrhea virus (PEDV) antibody-based assays is important for detecting infected animals, confirming previous virus exposure, and monitoring sow herd immunity. However, the potential cross-reactivity among porcine coronaviruses is a major concern for the development of pathogen-specific assays. In this study, we used serum samples (n = 792) from pigs of precisely known infection status and a multiplex fluorescent microbead-based immunoassay and/or enzyme-linked immunoassay platform to characterize the antibody response to PEDV whole-virus (WV) particles and recombinant polypeptides derived from the four PEDV structural proteins, i.e., spike (S), nucleocapsid (N), membrane (M), and envelope (E). Antibody assay cutoff values were selected to provide 100% diagnostic specificity for each target. The earliest IgG antibody response, mainly directed against S1 polypeptides, was observed at days 7 to 10 postinfection. With the exception of nonreactive protein E, we observed similar antibody ontogenies and patterns of seroconversion for S1, N, M, and WV antigens. Recombinant S1 provided the best diagnostic sensitivity, regardless of the PEDV strain, with no cross-reactivity detected against transmissible gastroenteritis virus (TGEV), porcine respiratory coronavirus (PRCV), or porcine deltacoronavirus (PDCoV) pig antisera. The WV particles showed some cross-reactivity to TGEV Miller and TGEV Purdue antisera, while N protein presented some cross-reactivity to TGEV Miller. The M protein was highly cross-reactive to TGEV and PRCV antisera. Differences in the antibody responses to specific PEDV structural proteins have important implications in the development and performance of antibody assays for the diagnosis of PEDV enteric disease.


Assuntos
Antígenos Virais/imunologia , Infecções por Coronavirus , Vírus da Diarreia Epidêmica Suína/imunologia , Doenças dos Suínos/diagnóstico , Suínos/virologia , Vírus da Gastroenterite Transmissível/imunologia , Animais , Anticorpos Antivirais/sangue , Anticorpos Antivirais/imunologia , Infecções por Coronavirus/diagnóstico , Infecções por Coronavirus/veterinária , Infecções por Coronavirus/virologia , Proteínas M de Coronavírus , Proteínas do Nucleocapsídeo de Coronavírus , Reações Cruzadas/imunologia , Diagnóstico Diferencial , Imunoglobulina G/sangue , Imunoglobulina G/imunologia , Proteínas do Nucleocapsídeo/imunologia , Doenças dos Suínos/virologia , Proteínas da Matriz Viral/imunologia
12.
J Virol ; 89(6): 3332-42, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25589635

RESUMO

UNLABELLED: Porcine epidemic diarrhea virus (PEDV) and transmissible gastroenteritis virus (TGEV) are economically important swine enteropathogenic coronaviruses. These two viruses belong to two distinct species of the Alphacoronavirus genus within Coronaviridae and induce similar clinical signs and pathological lesions in newborn piglets, but they are presumed to be antigenically distinct. In the present study, two-way antigenic cross-reactivity examinations between the prototype PEDV CV777 strain, three distinct U.S. PEDV strains (the original highly virulent PC22A, S indel Iowa106, and S 197del PC177), and two representative U.S. TGEV strains (Miller and Purdue) were conducted by cell culture immunofluorescent (CCIF) and viral neutralization (VN) assays. None of the pig TGEV antisera neutralized PEDV and vice versa. One-way cross-reactions were observed by CCIF between TGEV Miller hyperimmune pig antisera and all PEDV strains. Enzyme-linked immunosorbent assays, immunoblotting using monoclonal antibodies and Escherichia coli-expressed recombinant PEDV and TGEV nucleocapsid (N) proteins, and sequence analysis suggested at least one epitope on the N-terminal region of PEDV/TGEV N protein that contributed to this cross-reactivity. Biologically, PEDV strain CV777 induced greater cell fusion in Vero cells than did U.S. PEDV strains. Consistent with the reported genetic differences, the results of CCIF and VN assays also revealed higher antigenic variation between PEDV CV777 and U.S. strains. IMPORTANCE: Evidence of antigenic cross-reactivity between porcine enteric coronaviruses, PEDV and TGEV, in CCIF assays supports the idea that these two species are evolutionarily related, but they are distinct species defined by VN assays. Identification of PEDV- or TGEV-specific antigenic regions allows the development of more specific immunoassays for each virus. Antigenic and biologic variations between the prototype and current PEDV strains could explain, at least partially, the recurrence of PEDV epidemics. Information on the conserved antigenicity among PEDV strains is important for the development of PEDV vaccines to protect swine from current highly virulent PEDV infections.


Assuntos
Anticorpos Antivirais/imunologia , Infecções por Coronavirus/veterinária , Vírus da Diarreia Epidêmica Suína/imunologia , Doenças dos Suínos/imunologia , Vírus da Gastroenterite Transmissível/imunologia , Sequência de Aminoácidos , Animais , Variação Antigênica , Infecções por Coronavirus/imunologia , Infecções por Coronavirus/virologia , Reações Cruzadas , Gastroenterite Suína Transmissível/imunologia , Gastroenterite Suína Transmissível/virologia , Dados de Sequência Molecular , Vírus da Diarreia Epidêmica Suína/química , Vírus da Diarreia Epidêmica Suína/classificação , Vírus da Diarreia Epidêmica Suína/genética , Alinhamento de Sequência , Suínos , Doenças dos Suínos/virologia , Vírus da Gastroenterite Transmissível/química , Vírus da Gastroenterite Transmissível/classificação , Vírus da Gastroenterite Transmissível/genética
13.
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
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.
Appl Microbiol Biotechnol ; 100(17): 7457-69, 2016 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-27020282

RESUMO

Transmissible gastroenteritis coronavirus (TGEV) is a member of the genus Coronavirus, family Coronaviridae, order Nidovirales. TGEV is an enteropathogenic coronavirus that causes highly fatal acute diarrhoea in newborn pigs. An oral Lactobacillus casei (L. casei) vaccine against anti-transmissible gastroenteritis virus developed in our laboratory was used to study mucosal immune responses. In this L. casei vaccine, repetitive peptides expressed by L. casei (specifically the MDP and tuftsin fusion protein (MT)) were repeated 20 times and the D antigenic site of the TGEV spike (S) protein was repeated 6 times. Immunization with recombinant Lactobacillus is crucial for investigations of the effect of immunization, such as the first immunization time and dose. The first immunization is more important than the last immunization in the series. The recombinant Lactobacillus elicited specific systemic and mucosal immune responses. Recombinant L. casei had a strong potentiating effect on the cellular immunity induced by the oral L. casei vaccine. However, during TGEV infection, the systemic and local immune responses switched from Th1 to Th2-based immune responses. The systemic humoral immune response was stronger than the cellular immune response after TGEV infection. We found that the recombinant Lactobacillus stimulated IL-17 expression in both the systemic and mucosal immune responses against TGEV infection. Furthermore, the Lactobacillus vaccine stimulated an anti-TGEV infection Th17 pathway. The histopathological examination showed tremendous potential for recombinant Lactobacillus to enable rapid and effective treatment for TGEV with an intestinal tropism in piglets. The TGEV immune protection was primarily dependent on mucosal immunity.


Assuntos
Vacinas Bacterianas/imunologia , Gastroenterite/prevenção & controle , Lacticaseibacillus casei/imunologia , Células Th1/imunologia , Células Th2/imunologia , Vírus da Gastroenterite Transmissível/imunologia , Vacinas Virais/imunologia , Animais , Células Cultivadas , Gastroenterite/imunologia , Imunidade nas Mucosas/imunologia , Imunização , Imunoglobulina A/imunologia , Interleucina-17/biossíntese , Interleucina-17/imunologia , Suínos , Receptor 2 Toll-Like/biossíntese , Receptor 4 Toll-Like/biossíntese , Receptor Toll-Like 9/biossíntese , Tuftsina/genética
16.
J Virol ; 87(17): 9754-67, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23824792

RESUMO

Innate immune response is the first line of antiviral defense resulting, in most cases, in pathogen clearance with minimal clinical consequences. Viruses have developed diverse strategies to subvert host defense mechanisms and increase their survival. In the transmissible gastroenteritis virus (TGEV) as a model, we previously reported that accessory gene 7 counteracts the host antiviral response by associating with the catalytic subunit of protein phosphatase 1 (PP1c). In the present work, the effect of the absence of gene 7 on the host cell, during infection, was further analyzed by transcriptomic analysis. The pattern of gene expression of cells infected with a recombinant mutant TGEV, lacking gene 7 expression (rTGEV-Δ7), was compared to that of cells infected with the parental virus (rTGEV-wt). Genes involved in the immune response, the interferon response, and inflammation were upregulated during TGEV infection in the absence of gene 7. An exacerbated innate immune response during infection with rTGEV-Δ7 virus was observed both in vitro and in vivo. An increase in macrophage recruitment and activation in lung tissues infected with rTGEV-Δ7 virus was observed compared to cells infected with the parental virus. In summary, the absence of protein 7 both in vitro and in vivo led to increased proinflammatory responses and acute tissue damage after infection. In a porcine animal model, which is immunologically similar to humans, we present a novel example of how viral proteins counteract host antiviral pathways to determine the infection outcome and pathogenesis.


Assuntos
Imunidade Inata , Vírus da Gastroenterite Transmissível/imunologia , Proteínas Virais/genética , Proteínas Virais/imunologia , Animais , Células Cultivadas , Citocinas/genética , Citocinas/metabolismo , Deleção de Genes , Genes Virais , Interações Hospedeiro-Patógeno/genética , Interações Hospedeiro-Patógeno/imunologia , Imunidade Inata/genética , Mediadores da Inflamação/metabolismo , Masculino , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Suínos , Transcriptoma , Vírus da Gastroenterite Transmissível/genética , Vírus da Gastroenterite Transmissível/patogenicidade
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 Pathog ; 7(6): e1002090, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21695242

RESUMO

Transmissible gastroenteritis virus (TGEV) genome contains three accessory genes: 3a, 3b and 7. Gene 7 is only present in members of coronavirus genus a1, and encodes a hydrophobic protein of 78 aa. To study gene 7 function, a recombinant TGEV virus lacking gene 7 was engineered (rTGEV-Δ7). Both the mutant and the parental (rTGEV-wt) viruses showed the same growth and viral RNA accumulation kinetics in tissue cultures. Nevertheless, cells infected with rTGEV-Δ7 virus showed an increased cytopathic effect caused by an enhanced apoptosis mediated by caspase activation. Macromolecular synthesis analysis showed that rTGEV-Δ7 virus infection led to host translational shut-off and increased cellular RNA degradation compared with rTGEV-wt infection. An increase of eukaryotic translation initiation factor 2 (eIF2α) phosphorylation and an enhanced nuclease, most likely RNase L, activity were observed in rTGEV-Δ7 virus infected cells. These results suggested that the removal of gene 7 promoted an intensified dsRNA-activated host antiviral response. In protein 7 a conserved sequence motif that potentially mediates binding to protein phosphatase 1 catalytic subunit (PP1c), a key regulator of the cell antiviral defenses, was identified. We postulated that TGEV protein 7 may counteract host antiviral response by its association with PP1c. In fact, pull-down assays demonstrated the interaction between TGEV protein 7, but not a protein 7 mutant lacking PP1c binding motif, with PP1. Moreover, the interaction between protein 7 and PP1 was required, during the infection, for eIF2α dephosphorylation and inhibition of cell RNA degradation. Inoculation of newborn piglets with rTGEV-Δ7 and rTGEV-wt viruses showed that rTGEV-Δ7 virus presented accelerated growth kinetics and pathology compared with the parental virus. Overall, the results indicated that gene 7 counteracted host cell defenses, and modified TGEV persistence increasing TGEV survival. Therefore, the acquisition of gene 7 by the TGEV genome most likely has provided a selective advantage to the virus.


Assuntos
Genes Virais/imunologia , Interações Hospedeiro-Patógeno/imunologia , Vírus da Gastroenterite Transmissível/imunologia , Animais , Animais Recém-Nascidos , Linhagem Celular , Gastroenterite Suína Transmissível/mortalidade , Gastroenterite Suína Transmissível/virologia , Imunidade Inata , Suínos , Vírus da Gastroenterite Transmissível/genética , Vírus da Gastroenterite Transmissível/patogenicidade , Proteínas Virais/genética , Proteínas Virais/fisiologia , Virulência , Replicação Viral
19.
J Virol ; 85(6): 2703-13, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21191013

RESUMO

Although enveloped viruses typically trigger the prodigious secretion of alpha interferon (IFN-α) by plasmacytoid dendritic cells (pDC), porcine pDC remain quiescent when exposed to porcine reproductive and respiratory syndrome virus (PRRSV). This inactivity is likely due to virus-mediated interference since the typical IFN-α response by either purified or nonsorted porcine pDC to transmissible gastroenteritis virus (TGEV) or the Toll-like receptor 9 agonist, oligodeoxynucleotide (ODN) D19, was markedly reduced in the presence of PRRSV. Suppression occurred independently of virus viability and acidification of pDC early endosomes but correlated with diminished levels of IFN-α mRNA. This change was attributed to an abrogation of transcription resulting from a decrease in the otherwise enhanced amounts of the requisite interferon regulatory factor 7 (IRF-7), whose gene expression in turn was limited as a consequence of a lessened availability of nuclear-localized signal transducer and activator of transcription 1 (STAT1). While PRRSV also inhibited tumor necrosis factor alpha (TNF-α) synthesis by pDC responding to either agent, only the interleukin-2 (IL-2) and IL-6 production instigated by ODN D19 exposure was blocked. Likewise, PRRSV did not impact a specific TGEV-associated enhancement of IL-8 expression. Moreover, an augmented phosphorylation of NF-κB seen in activated pDC was not only unaffected by PRRSV but actually occurred in its presence. Thus, as supported by a demonstrated resilience of pDC to PRRSV infection, this pathogen may interact with a cell surface protein(s) to selectively impede the completion of cascades involved in cytokine production by stimulated pDC.


Assuntos
Células Dendríticas/imunologia , Células Dendríticas/virologia , Interferon Tipo I/antagonistas & inibidores , Síndrome Respiratória e Reprodutiva Suína/virologia , Vírus da Síndrome Respiratória e Reprodutiva Suína/imunologia , Vírus da Síndrome Respiratória e Reprodutiva Suína/patogenicidade , Animais , Regulação para Baixo , Expressão Gênica , Fator Regulador 7 de Interferon/biossíntese , América do Norte , Oligodesoxirribonucleotídeos/imunologia , RNA Mensageiro/biossíntese , Suínos , Vírus da Gastroenterite Transmissível/imunologia
20.
New Microbiol ; 35(4): 487-90, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23109017

RESUMO

The coronavirus heptide repeat (HR) region in the spike protein induces neutralizing antibodies that block the postfusion core formation and inhibit virus entry into target cells. The HR2 regions for coronaviruses of the same serogroup share high homology. We found that polyclonal antibodies derived from transmissible gastroenteritis coronavirus HR2 and upstream region were cross-reactive with the S proteins of the same serogroup in western blotting. The polyclonal antibodies also potently cross-neutralized viruses from the same serogroup. This study provides new insight for designing vaccine and therapeutic reagents against coronavirus infections.


Assuntos
Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/imunologia , Coronavirus/imunologia , Glicoproteínas de Membrana/química , Glicoproteínas de Membrana/imunologia , Sequências Repetitivas de Aminoácidos , Proteínas do Envelope Viral/química , Proteínas do Envelope Viral/imunologia , Motivos de Aminoácidos , Sequência de Aminoácidos , Animais , Coronavirus/classificação , Coronavirus/genética , Reações Cruzadas , Humanos , Glicoproteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos BALB C , Dados de Sequência Molecular , Glicoproteína da Espícula de Coronavírus , Sequências de Repetição em Tandem , Vírus da Gastroenterite Transmissível/química , Vírus da Gastroenterite Transmissível/genética , Vírus da Gastroenterite Transmissível/imunologia , Proteínas do Envelope Viral/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA