Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
Add more filters










Publication year range
1.
Vet Microbiol ; 293: 110100, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38718527

ABSTRACT

Recent epidemiological studies have discovered that a lot of cases of porcine epidemic diarrhea virus (PEDV) infection are frequently accompanied by porcine kobuvirus (PKV) infection, suggesting a potential relationship between the two viruses in the development of diarrhea. To investigate the impact of PKV on PEDV pathogenicity and the number of intestinal lymphocytes, piglets were infected with PKV or PEDV or co-infected with both viruses. Our findings demonstrate that co-infected piglets exhibit more severe symptoms, acute gastroenteritis, and higher PEDV replication compared to those infected with PEDV alone. Notably, PKV alone does not cause significant intestinal damage but enhances PEDV's pathogenicity and alters the number of intestinal lymphocytes. These results underscore the complexity of viral interactions in swine diseases and highlight the need for comprehensive diagnostic and treatment strategies addressing co-infections.


Subject(s)
Coinfection , Coronavirus Infections , Intestines , Kobuvirus , Lymphocytes , Porcine epidemic diarrhea virus , Swine Diseases , Animals , Porcine epidemic diarrhea virus/pathogenicity , Porcine epidemic diarrhea virus/physiology , Swine , Swine Diseases/virology , Coinfection/virology , Coinfection/veterinary , Coronavirus Infections/veterinary , Coronavirus Infections/virology , Lymphocytes/virology , Kobuvirus/pathogenicity , Kobuvirus/genetics , Intestines/virology , Diarrhea/virology , Diarrhea/veterinary , Virus Replication , Gastroenteritis/virology , Gastroenteritis/veterinary , Picornaviridae Infections/veterinary , Picornaviridae Infections/virology
2.
Antiviral Res ; 217: 105688, 2023 09.
Article in English | MEDLINE | ID: mdl-37516153

ABSTRACT

Vaccines that trigger mucosal immune responses at the entry portals of pathogens are highly desired. Here, we showed that antigen-decorated nanoparticle generated through CRISPR engineering of T4 bacteriophage can serve as a universal platform for the rapid development of mucosal vaccines. Insertion of Flu viral M2e into phage T4 genome through fusion to Soc (Small Outer Capsid protein) generated a recombinant phage, and the Soc-M2e proteins self-assembled onto phage capsids to form 3M2e-T4 nanoparticles during propagation of T4 in E. coli. Intranasal administration of 3M2e-T4 nanoparticles maintains antigen persistence in the lungs, resulting in increased uptake and presentation by antigen-presenting cells. M2e-specific secretory IgA, effector (TEM), central (TCM), and tissue-resident memory CD4+ T cells (TRM) were efficiently induced in the local mucosal sites, which mediated protections against divergent influenza viruses. Our studies demonstrated the mechanisms of immune protection following 3M2e-T4 nanoparticles vaccination and provide a versatile T4 platform that can be customized to rapidly develop mucosal vaccines against future emerging epidemics.


Subject(s)
Influenza Vaccines , Nanoparticles , Orthomyxoviridae Infections , Animals , Mice , Influenza Vaccines/genetics , Bacteriophage T4/genetics , Clustered Regularly Interspaced Short Palindromic Repeats , Escherichia coli/genetics , Orthomyxoviridae Infections/prevention & control , Mice, Inbred BALB C , Viral Matrix Proteins
3.
Virol J ; 20(1): 151, 2023 Jul 14.
Article in English | MEDLINE | ID: mdl-37452371

ABSTRACT

Pseudorabies virus (PRV) can infect multiple hosts and lead to fatal encephalitis. There is a significant increase in the number of microglia in the brain of animals infected with PRV. However, whether and how microglia contribute to central nervous system damage in PRV infection remain unknown. In the present study, we elucidated that PRV infection can cause more severe inflammatory cell infiltration, thicker and more numerous vessel sleeve walls, and more severe inflammatory responses in the brains of natural hosts (pigs) than in those of nonnatural hosts (mice). In a mice infection model, activated microglia restricted viral replication in the early stage of infection. Acute neuroinflammation caused by microglia hyperactivation at late-stage of infection. Furthermore, in vitro experiments revealed that microglia restricted viral replication and decreased viral infectivity. This may be associated with the phagocytic ability of microglia because we observed a significant increase in the expression of the membrane receptor TREM2 in microglia, which is closely related to phagocytosis, we observed that depletion of microglia exacerbated neurological symptoms, blood-brain barrier breakdown, and peripheral lymphocyte infiltration. Taken together, we revealed the dual role of microglia in protecting the host and neurons from PRV infection.


Subject(s)
Herpesvirus 1, Suid , Pseudorabies , Mice , Animals , Swine , Microglia , Brain , Immunity
4.
Vet Res Commun ; 47(4): 1949-1962, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37266866

ABSTRACT

The thymus, the central immune organ in mammals, plays an important role in immune defense. Porcine reproductive and respiratory syndrome virus (PRRSV) infection in piglets can cause thymus injury and immunosuppression. However, the mechanisms of thymus injury remain unknown. This study was aimed at investigating the specific manifestations of thymus injury through the construction of a PRRSV-infected piglet model and histopathological observation. In this study, fourteen 40-day-old PRRSV-free piglets were randomly divided into two groups, eleven of which were intramuscularly injected with 3 mL of PRRSV WUH3 virus suspension (106 PFU /mL) in the infection group, and three of which were sham-inoculated with 3 mL of RPMI-1640 medium in the control group. Clinical necropsy and samples collection were performed on day 8 after artificial infection. With the Illumina platform, the transcriptomes of piglet thymus tissues from infected and control piglets were sequenced to explore the relationships of differentially expressed genes (DEGs) and signaling pathways with thymus injury. The immune organs of PRRSV-infected piglets were severely damaged. The histopathological findings in the thymus indicated that PRRSV infection was associated with a large decrease in lymphocytes, cell necrosis and cell apoptosis; an increase in blood vessels and macrophages; thymic corpuscle hyperplasia; and interstitial widening of the thymic lobules. The transcriptomic analysis results revealed that the Gene Ontology functions of DEGs were enriched primarily in biological processes such as angiogenesis, regulation of angiogenesis and positive regulation of cell migration. Moreover, greater numbers of blood vessels and macrophages were observed in the thymus in PRRSV-infected than control piglets. KEGG pathway enrichment analysis revealed that the DEGs were significantly enriched in the Toll-like receptor signaling pathway, chemokine signaling pathway, IL-17 signaling pathway and TNF signaling pathway. The expression of TLR8, IRF5, the chemokines CCL2, CCL3L1 and CCL5; and their receptors CCR1, CCR2 and CCR5 was significantly up-regulated in PRRSV infection, thus suggesting that these cytokines were associated with the pathological processes of thymus injury.


Subject(s)
Porcine Reproductive and Respiratory Syndrome , Porcine respiratory and reproductive syndrome virus , Swine Diseases , Animals , Swine , Porcine Reproductive and Respiratory Syndrome/genetics , Transcriptome , Thymus Gland/pathology , Apoptosis , Mammals , Swine Diseases/genetics
5.
Arch Virol ; 168(6): 169, 2023 May 26.
Article in English | MEDLINE | ID: mdl-37233865

ABSTRACT

Japanese encephalitis virus (JEV) infection can cause brain tissue lesions characterized by neuronal death, and apoptosis is involved in JEV-induced neuronopathy. In the present study, mouse microglia were infected with JEV, and pyknosis with dark-staining nuclei of infected cells was detected using Hoechst 33342 staining. TUNEL staining showed that JEV infection promoted the apoptosis of BV2 cells, and the apoptosis rate was significantly increased at 24-60 hours postinfection (hpi) (P < 0.01) and was the highest at 36 h (P < 0.0001). Western blot results showed that the expression of the Bcl-2 protein in JEV-infected cells was downregulated significantly at 60 hpi (P < 0.001), whereas that of the Bax protein was observably upregulated at 60 hpi (P < 0.001). At the same time, the level of cytochrome c (Cyt c) was significantly increased (P < 0.001), and the expression levels of two apoptosis-related proteins, namely, cleaved caspase-3 (P < 0.01) and caspase-9 (P < 0.001), were elevated significantly. Immunofluorescence staining showed that the amount of Cyt c increased with time after infection. After BV2 cells were infected with JEV, the expression of RIG-1 increased significantly from 24 hpi to 60 h (P < 0.001). The expression of MAVS increased significantly at 24 h (P < 0.001) and decreased gradually from 24 h to 60 hpi. The expression of TBK1 and NF-κB (p65) was not significantly changed. The expression of p-TBK1 and p-NF-κB (p-p65) increased significantly within 24 h (P < 0.001) and decreased from 24 to 60 hpi. The expression levels of IRF3 and p-IRF3 peaked at 24 hpi (P < 0.001) and decreased gradually from 24 to 60 hpi. However, the expression levels of JEV proteins showed no significant change at 24 and 36 hpi but were markedly elevated at 48 and 60 hpi. Interference with the expression of the RIG-1 protein in BV2 cells resulted in a dramatic increase in the expression of the anti-apoptotic protein Bcl-2 (P < 0.05), whereas the pro-apoptotic protein Bax, cleaved caspase-9, and especially cleaved caspase-3 were downregulated (P < 0.05), and viral protein expression was notably reduced (P < 0.05). These results indicate that JEV induces apoptosis through mitochondrial-dependent apoptosis pathways, interfering with the expression of RIG-1 in BV2 cells can inhibit viral replication and inhibit apoptosis.


Subject(s)
Encephalitis Virus, Japanese , Encephalitis, Japanese , Animals , Mice , Encephalitis Virus, Japanese/physiology , Caspase 3/metabolism , Caspase 9/genetics , Caspase 9/metabolism , NF-kappa B/metabolism , Cell Line , Apoptosis , Signal Transduction , Proto-Oncogene Proteins c-bcl-2/metabolism
6.
Elife ; 112022 08 16.
Article in English | MEDLINE | ID: mdl-35972780

ABSTRACT

Zika virus (ZIKV) can be transmitted from mother to fetus during pregnancy, causing adverse fetal outcomes. Several studies have indicated that ZIKV can damage the fetal brain directly; however, whether the ZIKV-induced maternal placental injury contributes to adverse fetal outcomes is sparsely defined. Here, we demonstrated that ZIKV causes the pyroptosis of placental cells by activating the executor gasdermin E (GSDME) in vitro and in vivo. Mechanistically, TNF-α release is induced upon the recognition of viral genomic RNA by RIG-I, followed by activation of caspase-8 and caspase-3 to ultimately escalate the GSDME cleavage. Further analyses revealed that the ablation of GSDME or treatment with TNF-α receptor antagonist in ZIKV-infected pregnant mice attenuates placental pyroptosis, which consequently confers protection against adverse fetal outcomes. In conclusion, our study unveils a novel mechanism of ZIKV-induced adverse fetal outcomes via causing placental cell pyroptosis, which provides new clues for developing therapies for ZIKV-associated diseases.


Subject(s)
Placenta , Pregnancy Complications, Infectious , Pyroptosis , Zika Virus Infection , Animals , Female , Fetus , Humans , Mice , Placenta/pathology , Placenta/virology , Pore Forming Cytotoxic Proteins , Pregnancy , Pregnancy Complications, Infectious/virology , RNA, Viral , Tumor Necrosis Factor-alpha , Zika Virus/pathogenicity , Zika Virus Infection/complications
7.
Arch Virol ; 167(3): 849-859, 2022 Mar.
Article in English | MEDLINE | ID: mdl-35119507

ABSTRACT

Japanese encephalitis (JE) is a zoonotic epidemic disease caused by Japanese encephalitis virus (JEV), and currently, no medicines are available to treat this disease. Autophagy modulators play an important role in the treatment of tumors, heart disease, and some viral diseases. The aim of this study was to investigate the effects of autophagy modulators on JEV infection and the host response in mice. The experimental mice were grouped as follows: DMEM (control), JEV, JEV+rapamycin (JEV+Rapa), JEV+wortmannin (JEV+Wort), JEV+chloroquine (JEV+CQ), Rapa, Wort, and CQ. The control group was treated with DMEM. The mice in other groups were infected with 105 PFU of JEV, and Rapa, Wort, and CQ were administered 2 h prior to JEV challenge and then administered daily for 10 consecutive days. All mice were monitored for neurological signs and survival. The damage of subcellular structures in the mouse brain was evaluated by transmission electron microscopy. The distribution of virus in the mouse brain was determined by RNAScope staining and immunohistochemical staining. The neuroinflammatory responses in the brain were examined via quantitative real-time PCR, and the signal pathways involved in neuroinflammation were identified by Western blot. The mice in the JEV+Wort and JEV+CQ groups showed milder neurological symptoms, less damage to the mitochondria in the brain tissue, and a higher survival rate than those in the JEV+Rapa and JEV groups. Compared with the JEV+Rapa and JEV groups, the distribution of JEV in the brain of mice in the JEV+Wort and JEV+CQ groups was lower, and the inflammatory response was weaker. No significant difference was observed in the expression of the PI3K/AKT/NF-κB pathway in mouse brain among the different groups. Our study suggests that the autophagy inhibitors Wort and CQ reduce JEV infection and weaken the inflammatory response, which does not depend on the PI3K/AKT/NF-κB pathway in mouse brain.


Subject(s)
Encephalitis Virus, Japanese , Encephalitis, Japanese , Animals , Autophagy , Encephalitis Virus, Japanese/physiology , Encephalitis, Japanese/drug therapy , Inflammation/drug therapy , Mice , Phosphatidylinositol 3-Kinases
8.
Front Immunol ; 12: 745625, 2021.
Article in English | MEDLINE | ID: mdl-34712234

ABSTRACT

Developing influenza vaccines that protect against a broad range of viruses is a global health priority. Several conserved viral proteins or domains have been identified as promising targets for such vaccine development. However, none of the targets is sufficiently immunogenic to elicit complete protection, and vaccine platforms that can enhance immunogenicity and deliver multiple antigens are desperately needed. Here, we report proof-of-concept studies for the development of next-generation influenza vaccines using the bacteriophage T4 virus-like particle (VLP) platform. Using the extracellular domain of influenza matrix protein 2 (M2e) as a readout, we demonstrate that up to ~1,281 M2e molecules can be assembled on a 120 x 86 nanometer phage capsid to generate M2e-T4 VLPs. These M2e-decorated nanoparticles, without any adjuvant, are highly immunogenic, stimulate robust humoral as well as cellular immune responses, and conferred complete protection against lethal influenza virus challenge. Potentially, additional conserved antigens could be incorporated into the M2e-T4 VLPs and mass-produced in E. coli in a short amount of time to deal with an emerging influenza pandemic.


Subject(s)
Bacteriophage T4/immunology , Capsid Proteins/immunology , Influenza Vaccines , Vaccine Development/methods , Viral Matrix Proteins/immunology , Viroporin Proteins/immunology , Animals , Antibodies, Viral/biosynthesis , Antibodies, Viral/blood , Bronchoalveolar Lavage Fluid/immunology , Capsid Proteins/genetics , Female , Humans , Immunogenicity, Vaccine , Influenza A virus/immunology , Influenza, Human/prevention & control , Influenza, Human/virology , Mice , Mice, Inbred BALB C , Nanoparticle Drug Delivery System , Orthomyxoviridae Infections/prevention & control , Orthomyxoviridae Infections/virology , Peptide Library , Proof of Concept Study , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Viral Matrix Proteins/genetics , Viroporin Proteins/genetics
9.
PLoS Negl Trop Dis ; 15(6): e0008442, 2021 06.
Article in English | MEDLINE | ID: mdl-34153060

ABSTRACT

Japanese encephalitis virus (JEV) is a pathogen that causes severe vector-borne zoonotic diseases, thereby posing a serious threat to human health. Although JEV is potentially neurotropic, its pathogenesis and distribution in the host have not been fully elucidated. In this study, an infected mouse model was established using a highly virulent P3 strain of JEV. Immunohistochemistry and in situ hybridization, combined with anatomical imaging of the mouse brain, were used to dynamically localize the virus and construct three-dimensional (3D) images. Consequently, onset of mild clinical signs occurred in some mice at 3.5 d post JEV infection, while most mice displayed typical neurological signs at 6 d post-infection (dpi). Moreover, brain pathology revealed typical changes associated with non-suppurative encephalitis, which lasted up to 8 d. The earliest detection of viral antigen was achieved at 3 dpi in the thalamus and medulla oblongata. At 6 dpi, the positive viral antigen signals were mainly distributed in the cerebral cortex, olfactory area, basal ganglia, thalamus, and brainstem regions in mice. At 8 dpi, the antigen signals gradually decreased, and the localization of JEV tended to concentrate in the cerebrum and thalamus, while no viral antigen was detected in the brain at 21 dpi. In this model, the viral antigen was first expressed in the reticular thalamic nucleus (Rt), and the virus content is relatively stable. The expression of the viral antigen in the hippocampal CA2 region, the anterior olfactory nucleus, and the deep mesencephalic nucleus was high and persistent. The 3D images showed that viral signals were mostly concentrated in the parietal cortex, occipital lobe, and hippocampus, near the mid-sagittal plane. In the early stages of infection in mice, a large number of viral antigens were detected in denatured and necrotic neurons, suggesting that JEV directly causes neuronal damage. From the time of its entry, JEV is widely distributed in the central nervous system thereby causing extensive damage.


Subject(s)
Brain/virology , Encephalitis Virus, Japanese/physiology , Encephalitis, Japanese/virology , Animals , Brain/pathology , Imaging, Three-Dimensional , Immunohistochemistry , In Situ Hybridization , Mice , Time Factors
10.
Virology ; 558: 119-125, 2021 06.
Article in English | MEDLINE | ID: mdl-33756424

ABSTRACT

Coinfection of porcine epidemic diarrhea virus (PEDV) and porcine deltacoronavirus (PDCoV) is one of common findings in diarrheal piglets that cause massive economic losses to the pig industry globally. However, the mechanism of the co-infection is unclear. In this study, neonatal non-colostrum-fed piglets were exposed orally with a single infection of PDCoV or PEDV, or coinfection of PDCoV and PEDV. Clinically all viral infected piglets developed watery diarrhea and dehydration in 24 h post-exposure (hpe) and were succumbed to viral diarrhea disease and euthanized at 72 hpe. Histopathologically, acute gastroenteritis is evident in all viral infected piglet. Immunohistochemistry, RNAscope and RT-qPCR demonstrated that PEDV tropism changes from epithelial cells of small intestine to gastric epithelial cells and macrophages in Peyer's patches in the ileum. These findings suggest that coinfection of PDCoV and PEDV can alter PEDV tropism that may affect the outcome of viral disease in piglets. This animal model can be used for the pathogenesis and vaccination of viral coinfection in piglet in the future.


Subject(s)
Coinfection/virology , Coronavirus Infections/veterinary , Deltacoronavirus/pathogenicity , Gastrointestinal Tract/virology , Porcine epidemic diarrhea virus/pathogenicity , Viral Tropism , Animals , Coronavirus Infections/virology , Diarrhea/virology , Disease Models, Animal , Epithelial Cells/virology , Ileum/virology , Swine
11.
Avian Dis ; 64(3): 386-392, 2020 09 01.
Article in English | MEDLINE | ID: mdl-33205165

ABSTRACT

Clostridium perfringens (CP) is the etiologic agent of necrotic enteritis (NE) in broiler chickens that is responsible for massive economic losses in the poultry industry in response to voluntary reduction and withdrawal of antibiotic growth promoters. Large variations exist in the CP isolates in inducing intestinal NE lesions. However, limited information is available on CP isolate genetics in inducing NE with other predisposing factors. This study investigated the ability of five CP isolates from different sources to influence NE pathogenesis by using an Eimeria maxima (EM) coinfection NE model: Str.13 (from soil), LLY_N11 (healthy chicken intestine), SM101 (food poisoning), Del1 (netB+tpeL-) and LLY_Tpel17 (netB+tpeL+) for NE-afflicted chickens. The 2-wk-old broiler chickens were preinfected with EM (5 × 103 oocysts) followed by CP infection (around 1 × 109 colony-forming units per chicken). The group of the LLY_Tpel17 isolate with EM coinfection had 25% mortality. No mortality was observed in the groups infected with EM alone, all CP alone, or dual infections of EM/other CP isolates. In this model of EM/CP coinfections, the relative percentages of body weight gain showed statistically significant decreases in all EM/CP groups except the EM/SM101 group when compared with the sham control group. Evident gut lesions were only observed in the three groups of EM/LLY_N11, EM/Del1, and EM/LLY_Tpel17, all of which possessed an essential NE pathogenesis locus in their genomes. Our studies indicate that LLY_Tpel17 is highly pathogenic to induce severe gut lesions and would be a good CP challenge strain for studies investigating pathogenesis and evaluating the protection efficacy for antibiotic alternative approaches.


Subject(s)
Clostridium Infections/veterinary , Clostridium perfringens/pathogenicity , Coccidiosis/veterinary , Coinfection/veterinary , Enteritis/veterinary , Necrosis/veterinary , Poultry Diseases/microbiology , Animals , Chickens , Clostridium Infections/microbiology , Clostridium perfringens/physiology , Coccidiosis/parasitology , Coinfection/microbiology , Coinfection/parasitology , Disease Models, Animal , Eimeria/physiology , Enteritis/microbiology , Enteritis/parasitology , Necrosis/microbiology , Necrosis/parasitology , Poultry Diseases/parasitology , Virulence
12.
Sci Rep ; 10(1): 6340, 2020 04 14.
Article in English | MEDLINE | ID: mdl-32286320

ABSTRACT

Fowl cholera is a serious, highly contagious disease caused by the bacterium Pasteurella multocida (P. multocida) in a range of avian species and is characterized by an acute form of septicaemia. The pathogenic mechanism of chicken lung injury caused by the bacterium is unclear. Therefore, P. multocida Q (a reference standard strain isolated from chicken) and 1G1 (a clinic isolated strain from duck) were selected to infect chickens, establishing fowl cholera-induced laying hen models. Several important proteins involved in the process of lung injury were identified and quantified using immunohistochemistry and WB. The results showed that chicken lungs infected with bacteria for 24 h showed congestion and edema. The inflammatory factors HMGB1 and IL-6, intercellular matrix MMP, the cell apoptosis-associated caspase-3 and necrotic apoptosis signal molecules RIPK1 and RIPK3 were widely expressed in the lungs of group Q and were significantly different compared with those of 1G1 group and uninfected group (P < 0.05). The results indicated that RIPK1 and RIPK3 are involved in the injury process of chicken lungs after infection with P. multocida, and the mechanisms of lung injury induced by different strains are different.


Subject(s)
Avian Proteins/metabolism , Lung/metabolism , Pasteurella Infections/veterinary , Poultry Diseases/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Animals , Apoptosis , Avian Proteins/genetics , Chickens , HMGB1 Protein/genetics , HMGB1 Protein/metabolism , Inflammation , Interleukin-6/genetics , Interleukin-6/metabolism , Lung/microbiology , Lung/pathology , Matrix Metalloproteinases/genetics , Matrix Metalloproteinases/metabolism , Pasteurella Infections/metabolism , Pasteurella Infections/microbiology , Pasteurella multocida/pathogenicity , Poultry Diseases/microbiology , Receptor-Interacting Protein Serine-Threonine Kinases/genetics
13.
Article in English | MEDLINE | ID: mdl-32117795

ABSTRACT

Temperate phages are considered as natural vectors for gene transmission among bacteria due to the ability to integrate their genomes into a host chromosome, therefore, affect the fitness and phenotype of host bacteria. Many virulence genes of pathogenic bacteria were identified in temperate phage genomes, supporting the concept that temperate phages play important roles in increasing the bacterial pathogenicity through delivery of the virulence genes. However, little is known about the roles of temperate phages in attenuation of bacterial virulence. Here, we report a novel Bordetella bronchiseptica temperate phage, vB_BbrS_PHB09 (PHB09), which has a 42,129-bp dsDNA genome with a G+C content of 62.8%. Phylogenetic analysis based on large terminase subunit indicated that phage PHB09 represented a new member of the family Siphoviridae. The genome of PHB09 contains genes encoding lysogen-associated proteins, including integrase and cI protein. The integration site of PHB09 is specifically located within a pilin gene of B. bronchiseptica. Importantly, we found that the integration of phage PHB09 significantly decreased the virulence of parental strain B. bronchiseptica Bb01 in mice, most likely through disruption the expression of pilin gene. Moreover, a single shot of the prophage bearing B. bronchiseptica strain completely protected mice against lethal challenge with wild-type virulent B. bronchiseptica, indicating the vaccine potential of lysogenized strain. Our findings not only indicate the complicated roles of temperate phages in bacterial virulence other than simple delivery of virulent genes but also provide a potential strategy for developing bacterial vaccines.


Subject(s)
Bordetella Infections/microbiology , Bordetella bronchiseptica/pathogenicity , Bordetella bronchiseptica/virology , Lysogeny , Siphoviridae/physiology , Animals , Bacterial Vaccines/immunology , Bordetella Infections/prevention & control , Bordetella bronchiseptica/growth & development , Bordetella bronchiseptica/immunology , DNA, Viral/genetics , Female , Genome, Viral , Mice , Mice, Inbred BALB C , Phylogeny , Prophages/genetics , Prophages/physiology , Siphoviridae/classification , Siphoviridae/genetics , Siphoviridae/isolation & purification , Vaccines, Attenuated/immunology , Virulence
14.
Sci Rep ; 10(1): 1210, 2020 Jan 27.
Article in English | MEDLINE | ID: mdl-31988368

ABSTRACT

Swine pneumonia is a great threat for pig industry around the world, which is usually accompanied with neutrophils infiltration in the airway. Although interleukin-8 (CXCL8) and its receptors, CXC chemokine receptor 1 and 2 (CXCR1/2) in human have been well documented, the expression and function of CXCR1/2 is still unknown in swine. To explore the feasibility to develop new veterinary anti-inflammatory drugs targeting porcine CXCR1/2, we detected CXCR1/2 expression in swine pneumonia through Real-Time PCR and immunohistochemistry for the first time. Two porcine CXCR1/2 antagonists, CXCL8(3-72)N11R/G31P (pN11R) and CXCL8(3-72)G31P (pG31P) were prepared and their anti-inflammatory effects were evaluated using cell chemotaxis assays and animal experiments. Our data showed that CXCR1/2 expression, which was closely related to neutrophil infiltration in the lung, was significantly up-regulated in swine pneumonia. The pN11R and pG31P could effectively inhibit the directional migration of neutrophils in vitro. In vivo data also indicated that both pN11R and pG31P significantly relieved LPS-induced pneumonia in mice through decreasing the expression of TNF-α, CXCL8, and IL-1ß, and inhibiting neutrophil influx into the lung. pG31P was more efficient. Our study suggested that it is possible to develop new veterinary anti-inflammatory drugs targeting porcine CXCR1/2, and pG31P is a promising candidate.


Subject(s)
Interleukin-8/pharmacology , Interleukin-8/therapeutic use , Peptide Fragments/pharmacology , Peptide Fragments/therapeutic use , Pneumonia/drug therapy , Pneumonia/veterinary , Receptors, Interleukin-8A/antagonists & inhibitors , Receptors, Interleukin-8B/antagonists & inhibitors , Swine Diseases/drug therapy , Animals , Cell Movement/drug effects , Disease Models, Animal , Drug Discovery/methods , Female , Immunohistochemistry , Interleukin-8/metabolism , Lipopolysaccharides/adverse effects , Lipopolysaccharides/pharmacology , Mice , Mice, Inbred BALB C , Neutrophils/metabolism , Pneumonia/chemically induced , Pneumonia/pathology , Real-Time Polymerase Chain Reaction , Receptors, Interleukin-8A/genetics , Receptors, Interleukin-8A/immunology , Receptors, Interleukin-8A/isolation & purification , Receptors, Interleukin-8B/genetics , Receptors, Interleukin-8B/immunology , Receptors, Interleukin-8B/isolation & purification , Signal Transduction/drug effects , Swine
15.
Vet Microbiol ; 238: 108430, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31648727

ABSTRACT

Japanese Encephalitis Virus (JEV) is an important zoonotic flavivirus transmitted by mosquitos. JEV infection in sows primarily manifests as a reproductive disease such as abortion and transient infertility while in infected boars, it can cause orchitis. Previous studies mainly focused on the pathogenesis of human encephalitis caused by JEV infection, while few concentrations have been made to unveil the potential mechanism of reproductive dysfunction in JEV-infected pigs. In this study, histopathological analysis and immunohistochemistry staining was performed on testis of JEV-infected boars, indicating that JEV could infect testicular cells and cause inflammatory changes in testis. In vitro assays reveal that primary swine testicular cells and swine testis (ST) cells are highly permissive to JEV and significant inflammatory response was shown during JEV infection. Mechanically, we found that JEV infection increases the expression of retinoic acid-inducible gene I (RIG-I) and activates transcription factor NF-κB. Production of pro-inflammatory cytokines was greatly reduced in JEV infected testicular cells after knockout of RIG-I or treatment with the NF-κB specific inhibitor. In addition, activation of NF-κB was also significantly suppressed upon RIG-I knockout. Taken together, our results reveal that JEV could infect boar testicles, and RIG-I-NF-κB signaling pathway is involved in JEV-induced inflammation in swine testicular cells.


Subject(s)
DEAD Box Protein 58/metabolism , Encephalitis, Japanese/veterinary , NF-kappa B/metabolism , Orchitis/veterinary , Sus scrofa , Swine Diseases/physiopathology , Animals , Cells, Cultured , Encephalitis Virus, Japanese/physiology , Encephalitis, Japanese/complications , Encephalitis, Japanese/physiopathology , In Vitro Techniques , Inflammation , Male , Orchitis/etiology , Signal Transduction/immunology , Swine , Swine Diseases/virology
16.
Parasit Vectors ; 12(1): 285, 2019 Jun 04.
Article in English | MEDLINE | ID: mdl-31164143

ABSTRACT

BACKGROUND: Apicomplexan protozoans of the genus Eimeria cause coccidiosis, one of the most economically relevant parasitic diseases in chickens. The lack of a complete understanding of molecular mechanisms in the host-parasite interaction limits the development of effective control measures. In the present study, RNA sequencing (RNA-Seq) was applied to investigate the host mRNA profiles of the cecal mucosa collected at day 5 post-infection with Eimeria maxima (EM). RESULTS: Total RNA from cecal samples of the uninfected naïve control and the EM groups was used to make libraries, generating 354,924,372 and 356,229,250 usable reads, respectively, which were assembled into a total of 386,088 high-quality unigenes (transcripts) in Trinity software. RNA-Seq analysis of cecal samples in the two groups revealed 332 upregulated and 363 downregulated genes with significant differences (P ≤ 0.05), including several significant immune-related gene families, such as the major histocompatibility complex (MHC) class I alpha chain, granzyme A and immunoglobulin subtype genes among upregulated differentially expressed genes. In addition, a total of 60 clusters of differentiation (CD) molecular genes and 570 novel genes were found. The completeness of the assembled transcriptome was further assessed using the Kyoto Encyclopedia of Genes and Genomes (KEGG) database, Gene ontology (GO), eggNOG and CAZy for gene annotation. The broad gene categories represented by the highly differentiated host genes suggested enrichment in immune responses, and downregulation in the metabolic pathway, MARK signaling pathway, vascular smooth muscle contraction, and proteins processing in endoplasmic reticulum after EM infection. CONCLUSIONS: Eimeria maxima induced statistically significant differences in the cecal mucosal gene expression of infected chickens. These findings provide new insights into the host-parasite interaction and enhance our understanding of the molecular mechanism of avian coccidiosis.


Subject(s)
Cecum/parasitology , Chickens/parasitology , Coccidiosis/veterinary , Eimeria/pathogenicity , Gene Expression Profiling , Mucous Membrane/parasitology , Animals , Gene Expression , Gene Expression Regulation , Gene Ontology , Poultry Diseases/parasitology , RNA, Messenger
17.
Food Chem Toxicol ; 127: 270-279, 2019 May.
Article in English | MEDLINE | ID: mdl-30922968

ABSTRACT

Mequindox (MEQ) is a synthetic antibacterial agent. Recent studies showed that MEQ and its primary metabolites exhibit strong genotoxicity to mammalian cells, and MEQ induced carcinogenicity in mice. These findings suggest that chronic exposure to MEQ could lead to an increased risk of cancer later in life. In the present study, four groups of Wistar rats (55 rats/sex/group) were fed with diets containing MEQ (0, 25, 55, and 110 mg/kg) for 2 years. The results showed that the hematological system, liver, kidneys, and adrenal glands, as well as the developmental and reproductive systems, were the main targets for MEQ. Liver toxicity mediated by MEQ was associated with apoptosis and the nuclear factor κB (NF-κB) signaling pathway. In addition, MEQ increased the incidence of tumors in rats. Phosphorylated histone H2AX (γ-H2AX) is identified as a biomarker of cellular response to DNA double-strand breaks (DSB). Our data demonstrated that γ-H2AX expression was significantly increased in tumors. Thus, high levels of DSB might be responsible for carcinogenesis in rats, and further investigation is absolutely required to clarify the exact molecular mechanisms for carcinogenicity caused by MEQ in vivo.


Subject(s)
Apoptosis/drug effects , Carcinogens/toxicity , DNA Damage , Quinoxalines/toxicity , Animals , Body Weight/drug effects , Dietary Exposure , Female , Histones/biosynthesis , Immunohistochemistry , Liver/drug effects , Liver/metabolism , Male , NF-kappa B/metabolism , Neoplasms, Experimental/metabolism , Organ Size/drug effects , Phosphoproteins/biosynthesis , Rats, Wistar , Survival Analysis
18.
Avian Dis ; 63(3): 461-467, 2019 09 01.
Article in English | MEDLINE | ID: mdl-31967429

ABSTRACT

Clostridium perfringens (CP) type A and newly created type G strains are the key etiological factors in the induction of necrotic enteritis (NE), an important enteric disease that is responsible for the annual loss of $6 billion in the worldwide poultry industry. Several CP toxin genes were found to be critical in NE pathogenesis in chickens, but limited information is available on the CP lethal toxin tpeL gene. In this study, 19 CP strains isolated from NE-affected chicken farms were characterized microbiologically and molecularly and evaluated for their pathogenicity in commercial broiler chickens. Toxin typing by PCR revealed that all strains tested were positive for the netB toxin gene, but only five strains were positive for the tpeL toxin gene (LLY-TpeL 13, -TpeL 15, -TpeL 17, -TpeL 18, and -TpeL 19, simplified as TpeL 13, TpeL 15, TpeL 17, TpeL 18, and TpeL 19). High levels of TpeL proteins were detected in the concentrated culture supernatant from strains TpeL 13, 15, 17, and 19 by western blotting. Quantitative PCR showed that strains TpeL 13, 15, 17, 18, and 19 harbored a high number of copies of tpeL genes, while TpeL 18 had the highest number of copies of the tpeL gene among all CP strains tested when normalized with copy numbers of 16S rRNA gene as a housekeeping gene marker. The in vivo NE challenge test using multiple oral CP inoculations combined with a high-protein diet showed that TpeL 17 was the most virulent in inducing typical NE lesions, followed by TpeL 19 as the next most virulent, when tested in commercial broiler chickens. Infection with TpeL 17 reduced the growth rate significantly, as shown by reduced relative body weight gain percentage at day 5 postinfection. Availability of the virulent netB+tpeL+ CP strains is essential for the development of a CP-alone NE challenge model that could provide significant tools for understanding CP pathogenesis and for development of alternative to antibiotics.


Caracterización de cepas virulentas de Clostridium perfringens netB+/tpeL+ de granjas de pollos de engorde con enteritis necrótica. Clostridium perfringens (CP) tipo A y los tipos nuevos G son los factores etiológicos clave en la inducción de enteritis necrótica (NE), una enfermedad entérica importante que es responsable de pérdidas anuales de $6 mil millones en la industria avícola mundial. Se ha determinado que varios genes de toxinas de C. perfringens son críticos en la patogénesis de la enteritis necrótica en pollos, pero se dispone de información limitada sobre el gene tpeL de la toxina letal de C. perfringens. En este estudio, se caracterizaron microbiológicamente y molecularmente 19 cepas de C. perfringens aisladas de granjas avícolas afectadas por enteritis necrótica y se evaluaron en su patogenicidad en pollos de engorde comerciales. La tipificación de toxinas por PCR reveló que todas las cepas analizadas fueron positivas para el gene de la toxina netB, pero solo cinco fueron positivas para el gene de la toxina tpeL (LLY-TpeL 13, -TpeL 15, -TpeL 17, -TpeL 18, y -TpeL 19, simplificado como TpeL 13, TpeL 15, TpeL 17, TpeL 18 y TpeL 19). Los altos niveles de proteínas TpeL se detectaron en el sobrenadante del cultivo concentrado de las cepas TpeL 13, 15, 17 y 19 mediante inmunoelectrotransferencia tipo Western. La PCR cuantitativa mostró que las cepas TpeL 13, 15, 17, 18 y 19 albergaban un alto número de copias de los genes tpeL, mientras que TpeL 18 mostró el mayor número de copias del gene tpeL entre todas las cepas de C. perfringens analizadas cuando se normalizó con los números de copias del gene 16S rRNA como un marcador genético constitutivo. La prueba in vivo de desafío de enteritis necrótica utilizando múltiples inoculaciones orales de C. perfringens combinadas con una dieta alta en proteínas mostró que TpeL 17 fue la más virulenta en la inducción de lesiones típicas de enteritis necrótica, seguida de TpeL 19 como la siguiente más virulenta cuando se inoculó en pollos de engorde comerciales. La infección con TpeL 17 redujo significativamente la tasa de crecimiento, como lo demuestra la reducción del porcentaje de aumento de peso corporal relativo en el día cinco posterior a la infección. La disponibilidad de las cepas netB+ tpeL+ de C. perfringens virulentas es esencial para el desarrollo de un modelo de desafío enteritis necrótica únicamente con C. perfringens que podría proporcionar herramientas significativas para comprender la patogénesis de C. perfringens y para el desarrollo de alternativas a los antibióticos.


Subject(s)
Chickens , Clostridium Infections/veterinary , Clostridium perfringens/genetics , Clostridium perfringens/pathogenicity , Enteritis/veterinary , Necrosis/veterinary , Poultry Diseases/microbiology , Animals , Clostridium Infections/microbiology , Diet, High-Protein/veterinary , Enteritis/microbiology , Necrosis/microbiology , RNA, Bacterial/analysis , RNA, Ribosomal, 16S/analysis , Virulence
19.
J Nutr ; 148(8): 1209-1216, 2018 08 01.
Article in English | MEDLINE | ID: mdl-30137478

ABSTRACT

Background: Zearalenone (ZEN) can cause serious defects in development and reproduction in humans and animals. Silymarin shows antioxidant and estrogenic effects. Objective: This study was conducted to determine if silymarin can antagonize ZEN-induced hepatic and reproductive toxicities. Methods: Thirty-five 21-d-old female Sprague-Dawley rats (n = 7/diet) were fed a control diet (Ctrl) or Ctrl plus 20 mg ZEN/kg or Ctrl plus 20 mg ZEN/kg with 100, 200, or 500 mg silymarin/kg for 6 wk. Serum, livers, ovaries, and uterus were collected at week 6 for biochemistry, hormone, and redox status and selected gene and protein assays. Results: The consumption of ZEN decreased (P < 0.05) the final body weight by 17.9%, induced liver injury, increased (P < 0.05) aspartate aminotransferase and alkaline phosphatase activities, and decreased (P < 0.05) total protein and albumin concentrations in serum by 16.7-40.6%. ZEN also caused reproductive toxicity, including decreased (P < 0.05) 17ß-estradiol and increased (P < 0.05) follicle-stimulating hormone concentrations in serum by 12.7-46.3% and induced histopathologic alterations in the liver, ovaries, and uterus. Interestingly, these alterations induced by ZEN were alleviated (P < 0.05) by silymarin supplementation at 100, 200, and 500 mg/kg. Moreover, silymarin supplementation at the 3 doses mitigated (P < 0.05) ZEN-induced impairment in hepatic glutathione peroxidase activity, total antioxidant capacity, and malondialdehyde concentration by 17.6-100%. Meanwhile, silymarin supplementation at all doses upregulated (P < 0.05) phospho-ribosomal protein S6 kinase 1 (p-RPS6KB1) and 3ß-hydroxysteroid dehydrogenase (HSD3B) by 43.0-121% but downregulated (P < 0.05) AMP-activated protein kinase (AMPK) and 3α-hydroxysteroid dehydrogenase (HSD3A) in the liver relative to the ZEN group by 11.2-40.6%. In addition, silymarin supplementation at all doses elevated (P < 0.05) HSD3B by 1.8- to 2.5-fold and decreased (P < 0.05) estrogen receptor 1 (ESR1), ATP binding cassette (ABC) c1, and Abcc5 in ovaries and the uterus by 10.7-63.2%. Conclusion: Dietary silymarin supplementation at 100, 200, and 500 mg/kg protected rats from ZEN-induced hepatotoxicity and reproductive toxicity, potentially through improvement in the antioxidant capacity and regulation in the genes related to protein synthesis, ZEN metabolism, hormone synthesis, and ABC transporters in the tissues.


Subject(s)
Antioxidants/therapeutic use , Chemical and Drug Induced Liver Injury/prevention & control , Liver/drug effects , Reproduction/drug effects , Silybum marianum/chemistry , Silymarin/therapeutic use , Zearalenone/toxicity , AMP-Activated Protein Kinases/metabolism , ATP-Binding Cassette Transporters/metabolism , Animals , Antioxidants/metabolism , Antioxidants/pharmacology , Blood Proteins/metabolism , Chemical and Drug Induced Liver Injury/metabolism , Chemical and Drug Induced Liver Injury/pathology , Dietary Supplements , Estrogen Receptor alpha/blood , Female , Glutathione Peroxidase/metabolism , Hormones/blood , Hydroxysteroid Dehydrogenases/metabolism , Liver/enzymology , Liver/pathology , Malondialdehyde/blood , Multidrug Resistance-Associated Proteins/metabolism , Ovary/drug effects , Ovary/pathology , Phytotherapy , Rats, Sprague-Dawley , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , Silymarin/pharmacology , Uterus/drug effects , Uterus/pathology
20.
Virol J ; 14(1): 227, 2017 11 21.
Article in English | MEDLINE | ID: mdl-29157279

ABSTRACT

BACKGROUND: Postweaning multisystemic wasting syndrome (PMWS) is an emerging disease in swine. Pigs with PMWS are often infected with a variety of other pathogens, including bacteria, viruses and mycoplasm, in addition to porcine circovirus type 2 (PCV2). PCV2 and Haemophilus parasuis serovar 4 (HPS4) coinfection remain epidemic in China. METHODS: Here we report construction of a three-week-old naturally farrowed, colostrum-deprived (NFCD) piglet's infection model and demonstrate that PCV2-infected piglets with the HPS4 coinfection increased the virulence of PCV2 and these pathogens interact acquired PMWS. RESULTS: All the single infected piglets were transiently bacteremic or viremic. All the PCV2/HPS4 coinfected piglets developed PMWS, characterized by dyspnea, anorexia, prostration and lose weight severely. Co-infection with PCV2 and HPS4 resulted in an increased amount of virus in serum and tissues, presented a slower generation and lower levels of antibodies against PCV2. Co-infection with PCV2 and HPS4 resulted in further reductions in total and differential peripheral blood leukocyte counts. Meantime, PCV2/ HPS4 coinfection potentiated the severity of lung and lymphoid lesions by PCV2-associated, increased the virulence of PCV2-antigen and enhanced the incidence of PMWS in piglets. CONCLUSION: Co-infection with PCV2 and HPS4 induce the exacerbation of system injuries and enhance the pathogenicity of PCV2 in piglets.


Subject(s)
Circovirus/pathogenicity , Coinfection/veterinary , Haemophilus Infections/veterinary , Haemophilus parasuis/physiology , Porcine Postweaning Multisystemic Wasting Syndrome/microbiology , Porcine Postweaning Multisystemic Wasting Syndrome/virology , Virulence/physiology , Animals , Antibodies, Viral/blood , China , Coinfection/microbiology , Coinfection/pathology , Coinfection/virology , DNA, Viral/blood , Haemophilus Infections/pathology , Haemophilus Infections/virology , Leukocyte Count/veterinary , Polymerase Chain Reaction/veterinary , Porcine Postweaning Multisystemic Wasting Syndrome/pathology , Swine
SELECTION OF CITATIONS
SEARCH DETAIL
...