Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
1.
Artigo em Inglês | MEDLINE | ID: mdl-38536635

RESUMO

Porcine epidemic diarrhea virus (PEDV) infection results in significant mortality among newborn piglets, leading to substantial economic setbacks in the pig industry. Short-chain fatty acids (SCFA), the metabolites of intestinal probiotics, play pivotal roles in modulating intestinal function, enhancing the intestinal barrier, and bolstering immune responses through diverse mechanisms. The protective potential of Lactobacillus delbrueckii, Lactobacillus johnsonii, and Lactococcus lactis was first noted when administered to PEDV-infected piglets. Histological evaluations, combined with immunofluorescence studies, indicated that piglets receiving L. lactis displayed less intestinal damage, with diminished epithelial cell necrosis and milder injury levels. Differences in immunofluorescence intensity revealed a significant disparity in antigen content between the L. lactis and PEDV groups, suggesting that L. lactis might suppress PEDV replication, the intestine. We then assessed short-chain fatty acid content through targeted metabolomics, finding that acetate levels markedly varied from other groups. This protective impact was confirmed by administering acetate to PEDV-infected piglets. Data suggested that piglets receiving acetate exhibited resistance to PEDV. Flow cytometry analyses were conducted to evaluate the expression of innate and adaptive immune cells in piglets. Sodium acetate appeared to bolster innate immune defenses against PEDV, marked by elevated NK cell and macrophage counts in mesenteric lymph nodes, along with increased NK cells in the spleen and macrophages in the bloodstream. Acetic acid was also found to enhance the populations of CD8+ IFN-γ T cells in the blood, spleen, and mesenteric lymph, CD4+ IFN-γ T cells in mesenteric lymph nodes and spleen, and CD4+ IL-4+T cells in the bloodstream. Transcriptome analyses were carried out on the jejunal mucosa from piglets with PEDV-induced intestinal damage and from healthy counterparts with intact barriers. Through bioinformatics analysis, we pinpointed 189 significantly upregulated genes and 333 downregulated ones, with the PI3K-AKT, ECM-receptor interaction, and pancreatic secretion pathways being notably enriched. This transcriptomic evidence was further corroborated by western blot and qPCR. Short-chain fatty acids (SCFA) were found to modulate G protein-coupled receptor 41 (GPR41) and 43 (GPR43) in porcine intestinal epithelial cells (IPEC-J2). Post-acetic acid exposure, there was a notable upsurge in the ZO-1 barrier protein expression in IPEC-J2 compared to the unexposed control group (WT), while GPR43 knockdown inversely affected ZO-1 expression. Acetic acid amplified the concentrations of phosphorylated PI3K and AKT pivotal components of the PI3K/AKT pathway. Concurrently, the co-administration of AKT agonist SC79 and PI3K inhibitor LY294002 revealed acetic acid's role in augmenting ZO-1 expression via the P13K/AKT signaling pathway. This study demonstrates that acetic acid produced by Lactobacillus strains regulates intestinal barrier and immune functions to alleviate PEDV infection. These findings provide valuable insights for mitigating the impact of PEDV in the pig industry.

2.
Microbiome ; 12(1): 20, 2024 Feb 05.
Artigo em Inglês | MEDLINE | ID: mdl-38317217

RESUMO

BACKGROUND: The gut microbiota is a critical factor in the regulation of host health, but the relationship between the differential resistance of hosts to pathogens and the interaction of gut microbes is not yet clear. Herein, we investigated the potential correlation between the gut microbiota of piglets and their disease resistance using single-cell transcriptomics, 16S amplicon sequencing, metagenomics, and untargeted metabolomics. RESULTS: Porcine epidemic diarrhea virus (PEDV) infection leads to significant changes in the gut microbiota of piglets. Notably, Landrace pigs lose their resistance quickly after being infected with PEDV, but transplanting the fecal microbiota of Min pigs to Landrace pigs alleviated the infection status. Macrogenomic and animal protection models identified Lactobacillus reuteri and Lactobacillus amylovorus in the gut microbiota as playing an anti-infective role. Moreover, metabolomic screening of the secondary bile acids' deoxycholic acid (DCA) and lithocholic acid (LCA) correlated significantly with Lactobacillus reuteri and Lactobacillus amylovorus, but only LCA exerted a protective function in the animal model. In addition, LCA supplementation altered the distribution of intestinal T-cell populations and resulted in significantly enriched CD8+ CTLs, and in vivo and in vitro experiments showed that LCA increased SLA-I expression in porcine intestinal epithelial cells via FXR receptors, thereby recruiting CD8+ CTLs to exert antiviral effects. CONCLUSIONS: Overall, our findings indicate that the diversity of gut microbiota influences the development of the disease, and manipulating Lactobacillus reuteri and Lactobacillus amylovorus, as well as LCA, represents a promising strategy to improve PEDV infection in piglets. Video Abstract.


Assuntos
Infecções por Coronavirus , Microbioma Gastrointestinal , Vírus da Diarreia Epidêmica Suína , Doenças dos Suínos , Animais , Suínos , Infecções por Coronavirus/prevenção & controle , Infecções por Coronavirus/veterinária , Doenças dos Suínos/prevenção & controle , Resistência à Doença
3.
Int Immunopharmacol ; 121: 110568, 2023 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-37390563

RESUMO

Trichinellosis is a food-borne parasitic disease with a worldwide distribution that not only endangers human health but also leads to economic loss. Infection of pregnant animals with Trichinella spiralis (T. spiralis) may lead to abortion and other adverse consequences, so it is necessary to treat the infection during pregnancy. Albendazole (ABZ) is an effective therapeutic drug for adult T. spiralis worms. The safety of this drug during pregnancy, especially whether it has any effect on offspring, should be fully evaluated. A change in the immune response to T. spiralis in the offspring of pregnant mice treated with ABZ may lead to a difference in susceptibility to T. spiralis compared to that of the offspring of normal mice. However, the safety of ABZ treatment in pregnant mice and the effects on the immune response and susceptibility of their offspring to T. spiralis are poorly understood. Therefore, we assessed whether maternal ABZ treatment during pregnancy affects the immune response or susceptibility to T. spiralis in infected offspring. In this study, mice were infected with T. spiralis at 10 days of pregnancy and treated with ABZ at 3 days post infection (dpi), and the specific immune response in the pregnant mice and the survival rate and worm burden of their 6-week-old offspring after T. spiralis infection were examined. The results showed that the antiparasitic immune response in pregnant mice was activated by T. spiralis infection. Treatment of pregnant mice with ABZ increased the percentage of CD4 + T cells. The percentages of Th2 and Treg cells in the PP, MLN and spleen of pregnant mice in the infection group were significantly increased compared with those of normal mice. ABZ treatment during pregnancy promoted the Th2 and Treg immune responses in pregnant mice infected with T. spiralis. The transcriptional levels of the Th2 and Treg cytokines IL-4, IL-5, IL-13, and TGF-ß in the small intestine, MLN and spleen of pregnant mice in the treatment group were significantly higher than those of pregnant mice in the T. spiralis infection only group. The results indicated that ABZ treatment did not cause abortion in pregnant mice or affect the survival rate of their offspring. Furthermore, treatment of pregnant mice with ABZ had no significant effect on the above immune responses in their T. spiralis-infected offspring compared to those of T. spiralis-infected offspring of mice in the normal group. The results also indicated that treatment of pregnant mice infected with T. spiralis with ABZ shifted the immune response to a Th2- and Treg-skewed immune response and that this drug had no effects on the offspring survival rate, immune response or worm burden after T. spiralis infection. This study further indicated that ABZ administration to treat T. spiralis infection in pregnant mice is safe for the select immune response and susceptibility of their offspring.


Assuntos
Trichinella spiralis , Triquinelose , Gravidez , Feminino , Humanos , Camundongos , Animais , Albendazol/uso terapêutico , Citocinas , Imunidade
4.
Virus Res ; 322: 198931, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36130654

RESUMO

African swine fever virus (ASFV) employs diverse strategies to confront or evade host type I interferon (IFN-I)-induced antiviral responses. Moreover, the mechanisms of this process are largely unknown. Here, we assessed 27 ASFV proteins to determine whether any of them suppressed the cGAS-STING pathway to facilitate immune evasion. Using dual-luciferase assays, we found that ASFV MGF505-7R suppressed the activity of the IFN-ß and ISRE promoters and the expression of IFN-I and ISGs. MGF505-7R interacted with IRF7 and TBK1, degrading IRF7 by autophagy, cysteine, and proteasome pathways and TBK1 by the proteasome pathway. Moreover, TBK1 and IRF3 were phosphorylated by cGAS-STING stimulation. Finally, small interfering RNA (siRNA)-based silencing of MGF505-7R enhanced IFN-I antiviral activity. Taken together, these results preliminarily clarified the immune escape mechanism of ASFV MGF505-7R, which provides a potential target for developing antiviral agents.


Assuntos
Vírus da Febre Suína Africana , Febre Suína Africana , Interferon Tipo I , Suínos , Animais , Vírus da Febre Suína Africana/genética , Complexo de Endopeptidases do Proteassoma/metabolismo , Proteínas Virais/genética , Proteínas Virais/metabolismo , Nucleotidiltransferases/metabolismo , Antivirais/farmacologia , Antivirais/metabolismo
5.
Vaccines (Basel) ; 10(3)2022 Feb 24.
Artigo em Inglês | MEDLINE | ID: mdl-35334986

RESUMO

African Swine Fever Virus (ASFV) has spread worldwide, and the lack of vaccines severely negatively impacts the pig industry. In this study, the p14.5 protein encoded by ASFV was used as the antigen, and the p14.5 gene was expressed in vitro using the Lactobacillus expression system. Three new functionally recombinant Lactobacillus plantarum (L. plantarum) were constructed and the expressions of the p14.5 protein, p14.5-IL-33-Mus fusion protein and CTA1-p14.5-D-D fusion protein were successfully detected using Western blot analysis. After oral immunization of SPF mice with recombinant L. plantarum, flow cytometry and ELISA were performed to detect the differentiation and maturity of T lymphocytes, B lymphocytes and DCs of the mice, which were higher than those of the control group. Specific antibodies were produced. The immunogenicity of the adjuvant group was stronger than that of the single antigen group, and the IL-33 adjuvant effect was stronger than that of the CTA1-DD adjuvant.

6.
Vet Res ; 53(1): 7, 2022 Jan 24.
Artigo em Inglês | MEDLINE | ID: mdl-35073979

RESUMO

The type I interferon (IFN-I) signaling pathway is an important part of the innate immune response and plays a vital role in controlling and eliminating pathogens. African swine fever virus (ASFV) encodes various proteins to evade the host's natural immunity. However, the molecular mechanism by which the ASFV-encoded proteins inhibit interferon production remains poorly understood. In the present study, ASFV MGF360-11L inhibited cGAS, STING, TBK1, IKKε, IRF7 and IRF3-5D mediated activation of the IFN-ß and ISRE promoters, accompanied by decreases in IFN-ß, ISG15 and ISG56 mRNA expression. ASFV MGF360-11L interacted with TBK1 and IRF7, degrading TBK1 and IRF7 through the cysteine, ubiquitin-proteasome and autophagy pathways. Moreover, ASFV MGF360-11L also inhibited the phosphorylation of TBK1 and IRF3 stimulated by cGAS-STING overexpression. Truncation mutation analysis revealed that aa 167-353 of ASFV MGF360-11L could inhibit cGAS-STING-mediated activation of the IFN-ß and ISRE promoters. Finally, the results indicated that ASFV MGF360-11L plays a significant role in inhibiting IL-1ß, IL-6 and IFN-ß production in PAM cells (PAMs) infected with ASFV. In short, these results demonstrated that ASFV MGF360-11L was involved in regulating IFN-I expression by negatively regulating the cGAS signaling pathway. In summary, this study preliminarily clarified the molecular mechanism by which the ASFV MGF360-11L protein antagonizes IFN-I-mediated antiviral activity, which will help to provide new strategies for the treatment and prevention of ASF.


Assuntos
Vírus da Febre Suína Africana , Febre Suína Africana , Interferon Tipo I , Doenças dos Suínos , Febre Suína Africana/patologia , Febre Suína Africana/virologia , Vírus da Febre Suína Africana/metabolismo , Animais , Interferon Tipo I/genética , Interferon beta , Interferons/imunologia , Nucleotidiltransferases/genética , Nucleotidiltransferases/metabolismo , Transdução de Sinais , Suínos , Doenças dos Suínos/patologia
7.
Front Immunol ; 12: 800965, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34925386

RESUMO

Avian influenza viruses can be efficiently transmitted through mucous membranes, and conventional vaccines are not effective in protecting against mucosal infection by influenza viruses. To induce multiple immune responses in an organism, we constructed a recombinant Lactobacillus plantarum expressing the influenza virus antigen HA1 with the adjuvant dendritic cell-targeting peptide (DCpep). The recombinant L. plantarum strains NC8Δ-pWCF-HA1 and NC8Δ-pWCF-HA1-DCpep were used to immunize mice via oral administration, and the humoral, cellular and mucosal immune responses were evaluated. In addition, the serum levels of specific antibodies and hemagglutination inhibition (HI) levels were also measured. Our results showed that recombinant L. plantarum activated dendritic cells in Peyer's patches (PPs), increased the numbers of CD4+IFN-γ+ and CD8+IFN-γ+ cells in the spleen and mesenteric lymph nodes (MLNs), and affected the ability of CD4+ and CD8+ cells to proliferate in the spleen and MLNs. Additionally, recombinant L. plantarum increased the number of B220+IgA+ cells in PPs and the level of IgA in the lungs and different intestinal segments. In addition, specific IgG, IgG1 and IgG2a antibodies were induced at high levels in the mice serum, specific IgA antibodies were induced at high levels in the mice feces, and HI potency was significantly increased. Thus, the recombinant L. plantarum strains NC8Δ-pWCF-HA1 and NC8Δ-pWCF-HA1-DCpep have potential as vaccine candidates for avian influenza virus.


Assuntos
Glicoproteínas de Hemaglutininação de Vírus da Influenza/imunologia , Vacinas contra Influenza/imunologia , Lactobacillus plantarum , Animais , Anticorpos Antivirais/imunologia , Antígenos Virais/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Infecções por Orthomyxoviridae/imunologia , Infecções por Orthomyxoviridae/prevenção & controle , Vacinas Sintéticas/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA