Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 34
1.
Viral Immunol ; 37(3): 139-148, 2024 04.
Article En | MEDLINE | ID: mdl-38574260

Goose astrovirus type 2 (GAstV-2) is a novel pathogen causing visceral gout in goslings; it not only causes necrosis of renal epithelial cells but also causes spleen damage, indicating that GAstV-2 induces immunosuppression in goslings. However, to date, the interaction between GAstV-2 and immune cells remains unclear. In this study, peripheral blood lymphocytes and macrophages were isolated from goslings without GAstV-2 infection and then inoculated in vitro with GAstV-2, and the virus localization in the lymphocytes and macrophages, proliferation and apoptosis of lymphocytes, and phagocytic activity, reactive oxygen species (ROS) and nitric oxide (NO) production, and cell polarity in macrophages were determined. The results showed that GAstV-2 was observed in the cytoplasm of CD4 and CD8 T cells and macrophages, indicating that GAstV-2 can infect both lymphocytes and macrophages. GAstV-2 infection reduced the lymphocyte proliferation induced by Concanavalin A and lipopolysaccharide stimulation and increased the lymphocyte apoptosis rate and mRNA expression of Fas, demonstrating that GAstV-2 causes damage to lymphocytes. Moreover, GAstV-2 infection enhanced phagocytic activity and production of ROS and NO and induced a proinflammatory phenotype in macrophages (M1 macrophages), indicating that macrophages play an antiviral role during GAstV-2 infection. In conclusion, these results demonstrate that GAstV-2 infection causes damages to lymphocytes, and host macrophages inhibit GAstV-2 invasion during infection.


Astroviridae Infections , Geese , Animals , Humans , Geese/metabolism , Reactive Oxygen Species , Lymphocytes/metabolism , Macrophages , Astroviridae Infections/veterinary , Nitric Oxide/metabolism , Nitric Oxide/pharmacology
2.
Poult Sci ; 103(7): 103774, 2024 Apr 18.
Article En | MEDLINE | ID: mdl-38669820

Goose astrovirus genotype 2 (GAstV-2) mainly causes gout in goslings; therefore, it is a major pathogen threatening to goose flocks. However, the mechanisms underlying host-GAstV-2 interactions remain unclear because host cells suitable for GAstV-2 replication have been unavailable. We previously noted that GAstV-2 is primarily located in goose renal epithelial cells, where it causes kidney damage. Therefore, here, we derived goose primary renal tubular epithelial (RTE) cells (GRTE cells) from the kidneys of goose embryos after collagenase I digestion. After culture in Dulbecco's modified Eagle medium/Nutrient mixture F-12 with 10% fetal bovine serum (FBS), the isolated cells had polygonal with roadstone-like morphology; they were identified to be epithelial cells based on the presence of cytokeratin 18 expression detected through immunofluorescence assay (IFA). GAstV-2 infection in GRTE cells led to no obvious cytopathic effects; the maximum amounts of infectious virions were observed 48 h post infection through IFA and quantitative PCR. Next, RNA-seq was performed to identify and map post-GAstV-2 infection differentially expressed genes. The downregulated pathways were mainly related to metabolism, including tryptophan metabolism, drug metabolism by cytochrome P450, xenobiotic metabolism by cytochrome P450, retinol metabolism, butanoate metabolism, starch and sucrose metabolism, ascorbate and aldarate metabolism, and drug metabolism by other enzymes and peroxisome. In contrast, the upregulated pathways were mostly related to the host cell defense and proliferation, including extracellular matrix-receptor interaction, complement and coagulation cascades, phagosome, PI3K-Akt signaling pathway, human T-lymphotropic virus 1 infection, lysosome, and tumor necrosis factor signaling pathway. In conclusion, we developed a GRTE cell line for GAstV-2 replication and analyzed the potential host-GAstV-2 interactions through RNA-seq; our results may aid in further investigating the pathogenic mechanisms underlying GAstV-2 infection and provide strategies for its prevention and control.

3.
Poult Sci ; 103(4): 103484, 2024 Apr.
Article En | MEDLINE | ID: mdl-38306918

Goose astrovirus (GAstV)-2, a novel pathogen identified in 2018, mainly causes visceral gout in goslings, leading to approximately 50% mortality. At present, no commercial veterinary products are available to prevent and treat the disease. Our previous studies showed that nitric oxide (NO) and inducible NO synthase (iNOS) were markedly higher in the kidney and spleen of goslings infected with GAstV-2, but their effects during GAstV-2 infection remain unclear. In the present study, goslings were intraperitoneally injected with aminoguanidine (AG)-an iNOS inhibitor-to examine the role of NO during GAstV-2 infection. AG significantly decreased the serum NO concentration and iNOS mRNA expression in the kidney. Moreover, AG reduced the mortality, serum uric acid and creatinine content, and urate deposition in visceral organs and joints. Histopathological analysis demonstrated that AG reduced renal tubular cell necrosis, inflammatory cell infiltration, glycogen deposition in glomerular mesangium, and interstitial fibrosis, suggesting alleviation of kidney lesions. Furthermore, AG decreased the expression of renal injury markers such as KIM-1 and desmin; inflammatory cytokine-related genes such as IL-1ß, IL-8, and MMP-9; and autophagy-related genes and proteins such as LC3II, ATG5, and Beclin1. However, quantitative real-time PCR and immunohistochemistry showed that treatment with AG did not affect the kidney and liver viral load. These findings suggest that AG decreases the mortality rate and kidney lesions in goslings infected with GAstV-2 through mechanisms associated with autophagy and inhibition of inflammatory cytokine production in the kidney but not with GAstV-2 replication.


Astroviridae Infections , Astroviridae , Avastrovirus , Gout , Guanidines , Animals , Geese , Uric Acid , Chickens , Astroviridae/genetics , Gout/drug therapy , Gout/veterinary , Astroviridae Infections/drug therapy , Astroviridae Infections/veterinary , Kidney/pathology , Cytokines , Avastrovirus/genetics
4.
Vet Microbiol ; 287: 109887, 2023 Dec.
Article En | MEDLINE | ID: mdl-37925877

N6-methyladenosine (m6A), the most common modification in mammalian mRNA and viral RNA, regulates mRNA structure, stability, translation, and nuclear export. The Japanese encephalitis virus (JEV) is a mosquito-borne flavivirus causing severe neurologic disease in humans. To date, the role of m6A modification in JEV infection remains unclear. Herein, we aimed to determine the impact of m6A methylation modification on JEV replication in vitro and in vivo. Our results demonstrated that the overexpression of the m6A reader protein YTHDF1 in vitro significantly inhibits JEV proliferation. Additionally, YTHDF1 negatively regulates JEV proliferation in YTHDF1 knockdown cells and YTHDF1 knockout mice. MeRIP-seq analysis indicated that YTHDF1 interacts with several interferon-stimulated genes (ISGs), especially in IFIT3. Overall, our data showed that YTHDF1 played a vital role in inhibiting JEV replication. These findings bring novel insights into the specific mechanisms involved in the innate immune response to infection with JEV. They can be used in the development of novel therapeutics for controlling JEV infection.


Encephalitis Virus, Japanese , Encephalitis, Japanese , Humans , Mice , Animals , Encephalitis Virus, Japanese/genetics , Host-Pathogen Interactions , Encephalitis, Japanese/veterinary , Cell Line , RNA, Messenger , Virus Replication , Mammals , RNA-Binding Proteins/genetics
5.
Int J Pharm ; 625: 122091, 2022 Sep 25.
Article En | MEDLINE | ID: mdl-35964826

Triple-negative breast cancer (TNBC) is featured by aggression and metastasis and remains an unmet medical challenge due to high death rate. We aimed to repurpose maduramicin (MAD) as an effective drug against TNBC, and develop a nanoemulsion system to enhance anticancer efficacy of MAD. MDA-MB-231 and 4 T1 cells were used as in vitro model, and cell viability was determined by performing cell counting kit-8 and a colony-formation assay. Furthermore, MAD loaded nanoemulsion (MAD-NEs) was manufactured and characterized by a series of tests. The anticancer and anti-metastasis mechanism of MAD-NEs were assessed by performing cell cycle, apoptosis, wound-healing, transwell assay and Western blotting assays. Herein, MAD was firstly demonstrated to be an effective agent to suppress growth of TNBC cells. Subsequently, the optimized MAD-NEs were shown to have stability and high encapsulation efficiency, and could arrested cells in G0/G1 phase and induced apoptosis in TNBC cells. More importantly, MAD-NEs significantly impeded the metastasis of tumor cells, which was further demonstrated by the significant altered expression of epithelial-mesenchymal transition and extracellular matrix markers in vitro and in vivo. Moreover, compared to MAD, MAD-NEs exhibited higher efficacy in shrinking breast tumor size and repressing liver and lung metastasis in vivo, and showed excellent biocompatibility in tumor-bearing mice. The successfully prepared MAD-NEs are expected to be harnessed to suppress tumor growth, invasion and metastasis in the battle against malignant TNBC.


Triple Negative Breast Neoplasms , Animals , Cell Line, Tumor , Cell Movement , Cell Proliferation , Drug Repositioning , Humans , Lactones , Mice , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/pathology
6.
Sensors (Basel) ; 22(13)2022 Jun 23.
Article En | MEDLINE | ID: mdl-35808245

Video watermarking is an important means of video and multimedia copyright protection, but the current watermarking algorithm is difficult to ensure high robustness under various attacks. In this paper, a video watermarking algorithm based on NSCT, pseudo 3D-DCT and NMF has been proposed. Combined with NSCT, 3D-DCT and NMF, the algorithm embeds the encrypted QR code copyright watermark into the NMF base matrix to improve the anti-attack ability of the watermark under the condition of invisibility. The experimental results show that the algorithm ensures the invisibility of the watermark with a high signal-to-noise ratio of the video, and meanwhile has high ability and robustness against common single and combined attacks, such as filtering, noise, compression, shear, rotation and so on. The issue that the video watermarking algorithm has poor resistance to various attacks, especially the shearing attack, has been solved in this paper; thus, it can be used for digital multimedia video copyright protection.

7.
Int J Nanomedicine ; 17: 2475-2491, 2022.
Article En | MEDLINE | ID: mdl-35668999

Background: Halofuginone (HF)-loaded TPGS polymeric micelles (HTPM) were successfully fabricated using the thin-film hydration technique. HTPM via intravenous injection have been demonstrated to exert an excellent anticancer effect against triple-negative breast cancer (TNBC) cells and subcutaneous xenografts. In the present study, we further explored the potential treatment effect and mechanism of orally administered HTPM alone and in combination with surgical therapy on TNBC in subcutaneous and orthotopic mouse models. Methods: Herein, the stability and in vitro release behavior of HTPM were first evaluated in the simulated gastrointestinal fluids. Caco-2 cell monolayers were then used to investigate the absorption and transport patterns of HF with/without encapsulation in TPGS polymeric micelles. Subsequently, the therapeutic effect of orally administered HTPM was checked on subcutaneous xenografts of TNBC in nude mice. Ultimately, orally administered HTPM, combined with surgical therapy, were utilized to treat orthotopic TNBC in nude mice. Results: Our data confirmed that HTPM exhibited good stability and sustained release in the simulated gastrointestinal fluids. HF was authenticated to be a substrate of P-glycoprotein (P-gp), and its permeability across Caco-2 cell monolayers was markedly enhanced via heightening intracellular absorption and inhibiting P-gp efflux due to encapsulation in TPGS polymeric micelles. Compared with HF alone, HTPM showed stronger tumor-suppressing effects in subcutaneous xenografts of MDA-MB-231 cells when orally administered. Moreover, compared with HTPM or surgical therapy alone, peroral HTPM combined with partial surgical excision synergistically retarded the growth of orthotopic TNBC. Fundamentally, HTPM orally administered at the therapeutic dose did not cause any pathological injury, while HF alone led to weight loss and jejunal bleeding in the investigated mice. Conclusion: Taken together, HTPM could be applied as a potential anticancer agent for TNBC by oral administration.


Micelles , Triple Negative Breast Neoplasms , Animals , Caco-2 Cells , Cell Line, Tumor , Humans , Mice , Mice, Nude , Piperidines , Polymers , Quinazolinones , Triple Negative Breast Neoplasms/drug therapy , Vitamin E
8.
J Virol Methods ; 306: 114542, 2022 08.
Article En | MEDLINE | ID: mdl-35577125

Goose astrovirus (GAstV) is a novel pathogen that was discovered in 2018. It has two genotypes, GAstV-1 and GAstV-2, and both can cause visceral gout of goslings and result in significant economic losses. The present work aimed to develop a duplex TaqMan real-time quantitative reverse transcription PCR (RT-qPCR) assay to distinguish the two genotypes. MegAlign software was used to design two pairs of primers and a pair of matched probes based on the open reading frame 2 (ORF2) sequence with the greatest difference between GAstV-1 and GAstV-2, and primer and probe concentrations and annealing temperatures were optimised. Fluorescence signals were obtained for GAstV-1 and GAstV-2 in the FAM and VIC channels, respectively, but no fluorescent signal was observed for other pathogens. The detection limit for GAstV-1 and GAstV-2 was 33.3 and 33.7 DNA copies/µL, respectively. Intra- and inter-assay variability tests revealed excellent reproducibility. Furthermore, the assay detected GAstV-1 and GAstV-2 in allantoic fluids (100% positive) spiked with viruses, and 70 clinical gout gosling samples were examined, of which 11.4% were positive for GAstV-1, 74.3% were positive for GAstV-2%, and 5.7% were positive for mixed infection. In summary, the developed duplex RT-qPCR assay has high specificity, sensitivity, and reproducibility, and can be used in the clinic for detection of GAstV-1 and GAstV-2.


Astroviridae Infections , Avastrovirus , Gout , Animals , Astroviridae Infections/diagnosis , Astroviridae Infections/veterinary , Avastrovirus/genetics , Geese , Genotype , Real-Time Polymerase Chain Reaction , Reproducibility of Results , Reverse Transcriptase Polymerase Chain Reaction , Sensitivity and Specificity
9.
Front Vet Sci ; 9: 810977, 2022.
Article En | MEDLINE | ID: mdl-35359686

The aim of this study was to describe radiography, computed tomography (CT), and magnetic resonance imaging (MRI) findings of enzootic nasal tumors in goats infected with enzootic nasal tumor viruses. Five of six goats with a mean age of 2 years, showed clinical signs of respiratory disease. Head radiographs showed increased density of the unilateral or bilateral nasal cavity in four goats, and a CT scan showed that the space-occupying lesion of the nasal cavity originated from the ethmoid bone and was enhanced homogeneously postcontrast in all goats. The nasal concha was destroyed and the paranasal sinus mucosa was thickened and filled with fluid in some goats. On MRI, the mass exhibited equal or slightly higher signal intensity on T2 weighted images, equal signal intensity on T1 weighted images, a high signal on fluid-attenuated inversion recovery images and heterogeneous enhancement postcontrast. After dissection, histopathological examination of the mass and virus genome detection of the nasal secretions confirmed that the intranasal mass was a low-grade adenocarcinoma and that the goats were infected with enzootic nasal tumor virus type 2. In conclusion, CT and MRI have high diagnostic values for enzootic nasal tumors because they match the postmortem findings and are more accurate than radiography.

10.
Vet Microbiol ; 263: 109244, 2021 Dec.
Article En | MEDLINE | ID: mdl-34649010

Goose nephritic astrovirus (GNAstV) has recently been identified, which causes kidney swelling and visceral gout in goslings. However, the pathological changes in kidney tissue due to GNAstV infection have not yet been described. In the study, fifty goslings were orally infected with GNAstV, and fifty goslings received PBS as a control. Kidney tissue was collected at different days following infection (dpi) to assess the injury. GNAstV infection reduced body weight, increased the relative weight of the kidney, and increased serum uric acid and creatinine levels. GNAstV was found within renal epithelial cells, and the viral load in the kidney peaked at 7 dpi. Pale and swollen kidney tissue was observed in infected goslings, especially at 5 and 7 dpi. GNAstV infection caused degeneration and necrosis of renal epithelial cells, structural destruction of the brush border, glycogen deposition in the glomerular mesangium, increased fibrosis, and infiltration of inflammatory cells into the renal interstitium. Moreover, swollen mitochondria, broken mitochondrial ridges, autophagosomes, and autophagolysosomes were observed under ultrahistopathological examination. GNAstV infection increased levels of LC3B, ATG5, and Beclin 1, and decreased p62, and downregulated WT1 mRNA and upregulated desmin mRNA. At early stages, GNAstV infection decreased expression of intercellular junction-related genes, including ZO-1, occludin, claudin-10, and catenin-α2. In conclusion, GNAstV infection causes renal epithelial cell autophagy, destruction of brush border and intercellular junctions, podocyte damage, and increased fibrosis, ultimately resulting in damage to the kidney.


Astroviridae Infections , Autophagy , Podocytes , Poultry Diseases , Animals , Astroviridae Infections/pathology , Astroviridae Infections/veterinary , Avastrovirus , Fibrosis , Geese , Intercellular Junctions , Kidney/cytology , Kidney/virology , Podocytes/virology , Poultry Diseases/pathology , Poultry Diseases/virology , RNA, Messenger , Uric Acid
11.
Viruses ; 13(6)2021 06 09.
Article En | MEDLINE | ID: mdl-34207913

The emergence of a novel goose nephritic astrovirus (GNAstV) has caused economic losses to the Chinese goose industry. High viral load is found in the spleen of goslings infected with GNAstV, but pathological injuries to the spleen due to GNAstV are largely unknown. In this study, 50 two-day-old goslings were infected orally with GNAstV, and 50 goslings were treated with PBS as control. Spleens were collected at different times following infection to assess damage. GNAstV infection caused visceral gout and urate deposition in joints, and resulted in 16% mortality. GNAstV was found in the lymphocytes and macrophages within the spleen. Lymphocyte loss, especially around the white pulp, and destruction and decline in the number of reticular fibers was observed in GNAstV-infected goslings. Moreover, in GNAstV-infected goslings, ultrahistopathological examination found that splenic lymphocytes exhibited condensed chromatin and apoptotic bodies, and reticular cells displayed damage to plasma membrane integrity and swollen mitochondria. Furthermore, TUNEL staining confirmed apoptosis of lymphocytes, and the mRNA levels of Fas and FasL were significantly increased in the GNAstV-infected goslings. In addition, GNAstV infection reduced the number and protein expression of CD8. In conclusion, GNAstV infection causes lymphocyte depletion, reticular cell necrosis, reticular fiber destruction, lymphocyte apoptosis, and reduction in CD8 levels, which contribute to spleen injury.


Apoptosis , Avastrovirus/physiology , Geese/virology , Lymphocytes/metabolism , Poultry Diseases/etiology , Poultry Diseases/metabolism , Animals , Avastrovirus/classification , Avastrovirus/genetics , Biopsy , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes , Immunohistochemistry , Lymphocyte Count , Lymphocytes/immunology , Lymphocytes/pathology , Poultry Diseases/diagnosis , Spleen/immunology , Spleen/metabolism , Spleen/pathology , Spleen/virology , Viral Load
12.
Poult Sci ; 100(4): 100990, 2021 Apr.
Article En | MEDLINE | ID: mdl-33647718

Goose nephritic astrovirus (GNAstV) was first isolated in 2018, causing great economic losses to the goose industry. However, little is known about host immune response to GNAstV infection. In this study, forty 2-day-old goslings were randomly divided into 2 groups: infection and negative control groups. Each gosling in the infection group was challenged with 0.5 mL GNAstV-JSHA intramuscularly, whereas the gosling in the negative control group was inoculated with the same amount of PBS. Histopathological changes and virus location in the spleen and kidney were examined, and the expression of immune-related genes was determined by qPCR at 7 and 14 d after infection. Our results showed that GNAstV infection induced degeneration and necrosis of splenic lymphocytes and renal epithelial cells, and these cells were positive for the virus. In addition, GNAstV infection induced the activation of pattern recognition receptors (RIG-I, MDA-5, and TLR3) and key adaptor molecules (MyD88, MAVS, and IRF7) in the spleen and kidney, and upregulated the gene expression of interferon-α in the spleen and antiviral proteins (MX1, OASL, and IFITM3) in the spleen and kidney. Moreover, high expression levels of interleukin (IL)-1ß and IL-8 in the spleen and iNOS in the spleen and kidney were found. These results indicated that GNAstV infection activated host innate immune response. Furthermore, GNAstV infection increased the expression levels of CD8+, MHCI, and MHCII, indicating that adaptive immune response was activated. Besides, TGF-ß was highly expressed in the spleen and kidney, which may be an immune evasion strategy of GNAstV to cause infection. Interestingly, both IL-1ß and IL-6 mRNA levels were decreased in the kidney, which may help reduce kidney lesions. This is the first study to report changes in immune-related gene expression in response to GNAstV infection, and our results provide insights into viral pathogenesis.


Astroviridae Infections , Geese , Gene Expression Regulation , Spleen , Animals , Astroviridae Infections/immunology , Astroviridae Infections/veterinary , Avastrovirus/immunology , Chickens , Gene Expression Profiling , Gene Expression Regulation/immunology , Immunity, Innate/immunology , Kidney/virology , Spleen/immunology , Spleen/virology
13.
Virus Res ; 291: 198220, 2021 01 02.
Article En | MEDLINE | ID: mdl-33152381

Cyclic GMP-AMP synthase (cGAS) is a main sensor used to detect microbial DNA in the cytoplasm, which subsequently induces the production of interferon (IFN) via the cGAS/STING/IRF3 signaling pathway, leading to an antiviral response. However, some viruses have evolved multiple strategies to escape this process. Pseudorabies virus (PRV) is a double-stranded DNA virus belonging to the Alphaherpesvirinae subfamily, which can cause serious damage to the porcine industry. Many herpesvirus components have been reported to counteract IFN production, whereas little is known of PRV. In the present study, we found that PRV glycoprotein E (gE) was involved in counteracting cGAS/STING-mediated IFN production. Ectopic expression of gE decreased cGAS/STING-mediated IFN-ß promoter activity and the level of mRNA expression. Moreover, gE targeted at or downstream of IRF3 was found to inhibit IFN-ß production. However, gE did not affect the phosphorylation, dimerization and nuclear translocation of IRF3. Furthermore, gE is located on the nuclear membrane and could subsequently degrade CREB-binding protein (CBP). MG132, a proteasome inhibitor, decreased CBP degradation and restored the IFN-ß production induced by gE. Finally, gE-deleted PRV induced a higher level of IFN-ß production and reduced CBP degradation compared to wild-type PRV. Together, these results demonstrate that PRV gE can inhibit cGAS/STING-mediated IFN-ß production by degrading CBP to interrupt the enhanced assembly of IRF3 and CBP.


CREB-Binding Protein/metabolism , Herpesvirus 1, Suid/immunology , Immunity, Innate , Interferon-beta/antagonists & inhibitors , Viral Envelope Proteins/genetics , Viral Envelope Proteins/immunology , Animals , CREB-Binding Protein/genetics , Chlorocebus aethiops , HEK293 Cells , Herpesvirus 1, Suid/chemistry , Herpesvirus 1, Suid/genetics , Humans , Interferon Regulatory Factor-3/genetics , Interferon Regulatory Factor-3/metabolism , Interferon-beta/immunology , Phosphorylation , Signal Transduction/immunology , Swine , Vero Cells
14.
Virology ; 548: 31-38, 2020 09.
Article En | MEDLINE | ID: mdl-32838944

Entering the nucleus is important for Porcine circovirus type 2 (PCV2) replication. Karyopherins (KPNs) mediate the nuclear import of many cytoplasmic proteins. Our previous study showed that KPNA3 is involved in interferon production during PCV2 infection induced by Poly I:C and ISD (Interferon stimulatory DNA). However, it remains unclear whether PCV2 replication is associated with KPNA3. In the present study, knockdown of KPNA3 promoted the replication of PCV2, whereas overexpression of KPNA3 inhibited PCV2 replication in PK-15 cells. Furthermore, KPNA3 knockdown inhibited IRF3 and reduced the expression of antiviral genes including IFN-ß, ISG54, Mx1 and ISG56, while the opposite results were obtained after KPNA3 overexpression. KPNA3 knockdown also promoted p65 nuclear translocation and increased the mRNA expression of IL-10 and IL-1ß. These results suggested that KPNA3 facilitates IRF3 entry into the nucleus and the production of an antiviral response, resulting in PCV2 replication inhibition and blockage of NF-κB signal activation.


Cell Nucleus/metabolism , Circoviridae Infections/veterinary , Circovirus/physiology , Swine Diseases/metabolism , alpha Karyopherins/metabolism , Animals , Cell Nucleus/genetics , Circoviridae Infections/genetics , Circoviridae Infections/metabolism , Circoviridae Infections/virology , Circovirus/genetics , Host-Pathogen Interactions , Interleukin-10/genetics , Interleukin-10/metabolism , NF-kappa B/genetics , NF-kappa B/metabolism , Protein Transport , Signal Transduction , Swine , Swine Diseases/genetics , Swine Diseases/virology , Virus Replication , alpha Karyopherins/genetics
15.
Poult Sci ; 99(4): 1967-1974, 2020 Apr.
Article En | MEDLINE | ID: mdl-32241477

In 2018, a new goose astrovirus (GAstrV) was reported in China, which causes 2 to 20% deaths in 4- to 16-day-old goslings causing great damages to the livestock industry. Gout is the typical feature of GAstrV infection in goslings. However, the mechanism of gout formation remains unclear. In the present study, 2-day-old goslings were infected intramuscularly with GAstrV for 14 D. One quarter of the infected goslings died, and typical gout pathological changes were found in the dead infected goslings. Pathological changes were observed in the morphology of the kidney and liver, such as degeneration, necrosis, and inflammatory cell infiltration. Accordingly, a high virus load was found in both organs. The serum level of uric acid in the inoculated goslings was higher, whereas no differences were found in levels of creatinine, calcium, and phosphorus. Moreover, the xanthine dehydrogenase (XOD) and adenosine deaminase (ADA) activities and the mRNA levels of xanthine dehydrogenase, adenosine deaminase, phosphoribosyl pyrophosphate amidotransferase, and phosphoribosyl pyrophosphate synthetase 1 in livers increased, wheres the multidrug resistance-associated protein 4 mRNA level and Na-K-ATPase activity in the kidneys decreased. These results showed that GAstrV infection could cause lesions on the liver and kidney and then increase the expression or activity of enzymes related to uric acid production in the liver and decrease renal excretion function, which contribute to hyperuricemia and gout formation.


Astroviridae Infections/veterinary , Avian Proteins/genetics , Carrier Proteins/genetics , Geese , Gout/veterinary , Poultry Diseases/metabolism , Uric Acid/metabolism , Animals , Astroviridae Infections/complications , Astroviridae Infections/metabolism , Astroviridae Infections/virology , Avastrovirus/physiology , Avian Proteins/metabolism , Carrier Proteins/metabolism , Disease Models, Animal , Feces/chemistry , Gout/metabolism , Gout/virology , Kidney/metabolism , Liver/chemistry , Poultry Diseases/virology , Purines/metabolism
16.
Res Vet Sci ; 130: 179-183, 2020 Jun.
Article En | MEDLINE | ID: mdl-32199176

S. Pullorum is a causative agent of enteric disease of poultry with serious diarrhea. However, the detailed mechanism behind its injury to intestinal mucosa barrier, especially for intestinal stem cells, is unclear. In this study, S. Pullorum were orally administrated to 3 days old chicken to investigate the pathogenesis of S. Pullorum on intestinal mucosal barrier, especially on the proliferation of epithelial cells. We found that S. Pullorum could colonize in the cecum and invade into the liver through intestinal mucosa damage, which caused obvious pathological changes in liver and intestine and even leaded to death, as well as significant reduction of body weight. We also found that S. Pullorum infection enhanced the mRNA expression of IL-1ß and IL-6 through TLR4/MyD88 pathway, which was also further verified by the increased lipopolysaccharide (LPS) levels in serum. Furthermore, S. Pullorum increased the depth of crypt and density of PCNA+ cells significantly through the over-activation of Wnt/ß-catenin signaling pathway. The expression of intestinal stem cells markers Lgr5 and Bmi1 was also increased after S. Pullorum infection to support the crypt hyperplasia. In addition, we verified that S. Pullorum infection enhanced the mRNA expression of IL-1ß, TLR4, Lgr5 and Bmi1. Our study indicated that S. Pullorum infection damaged the intestinal mucosa barrier to induce diarrhea, affected the abnormal proliferation of intestinal stem cells by over-activation of Wnt/ß-catenin pathway in chicken.


Chickens , Hyperplasia/veterinary , Intestinal Diseases/veterinary , Poultry Diseases/physiopathology , Salmonella Infections, Animal/physiopathology , Salmonella enterica/physiology , Animals , Avian Proteins/physiology , Hyperplasia/microbiology , Hyperplasia/physiopathology , Intestinal Diseases/microbiology , Intestinal Diseases/physiopathology , Intestines/physiopathology , Poultry Diseases/microbiology , Salmonella Infections, Animal/microbiology , Salmonella enterica/pathogenicity , Signal Transduction , Stem Cells/metabolism , Virulence , Wnt Signaling Pathway , beta Catenin/physiology
17.
Res Vet Sci ; 125: 148-152, 2019 Aug.
Article En | MEDLINE | ID: mdl-31228738

In the porcine industry, some piglets show slightly enlarged and bluish inguinal lymph nodes. However, the causative factors for these signs and prevention of these signs remain unclear. Tylvalosin is a broad-spectrum antibiotic with an immunomodulatory function. This study was aimed at evaluating the effect of tylvalosin on the abovementioned signs. Thus, fifteen 90-day pregnant sows were divided into an untreated control group and 0.1 and 0.2 g/kg feed tylvalosin-treated groups until delivery. Forty-five piglets on day 2 after birth (15 each group) were blooded, then oxidative stress, serum cytokine levels, routine blood analysis, and effect of sera on macrophage phagocytic activity were examined. Fifteen piglets on day 2 after birth (5 in each group) were euthanized and pathological changes in the inguinal lymph nodes were observed. The untreated piglets showed hemorrhage, hemosiderin accumulation, and increased macrophages in the inguinal lymph nodes. However, tylvalosin administration in sows alleviated these signs in their piglets; increased total antioxidant capacity and serum glutathione levels; decreased serum IL-1ß, TNF-α, and IL-10 levels; improved the percentages of neutrophils and lymphocytes in the blood; and increased the body weight of the weaning piglets. In addition, the serum of newborn piglets also showed enhanced RAW264.7 macrophage phagocytic activity. These results demonstrated that tylvalosin administration in pregnant sows attenuates the enlargement and bluish coloration of inguinal lymph nodes in newborn piglets.


Anti-Bacterial Agents/administration & dosage , Lymph Nodes/drug effects , Sus scrofa/physiology , Tylosin/analogs & derivatives , Animals , Animals, Newborn/immunology , Animals, Newborn/physiology , Color , Dose-Response Relationship, Drug , Female , Lymph Nodes/physiopathology , Organ Size/drug effects , Pregnancy , Random Allocation , Sus scrofa/immunology , Tylosin/administration & dosage
18.
Vet Microbiol ; 227: 34-40, 2018 Dec.
Article En | MEDLINE | ID: mdl-30473349

PCV2 is a single-stranded DNA virus that we previously found to induce IFN-ß production via RIG-I and MDA-5. cGAS is known to be the most important DNA sensor for the recognition of cytosolic DNA; however, it remains unclear whether the interferon production induced by PCV2 is associated with cGAS. In the present study, PCV2 infection was found to increase the level of cGAS and STING expression, promote the release of cyclic dinucleotide cGAMP, and induce STING dimerization and translocation into the nucleus of PK-15 cells. These findings indicate that PCV2 infection activates both cGAS and STING. Furthermore, the knockdown of cGAS and STING decreased both the mRNA expression and promoter activity of IFN-ß, demonstrating that the cGAS/STING signaling pathway contributes to the production of IFN-ß. In addition, a knockdown of cGAS and STING also decreased the PCV2 viral load and infectivity. Therefore, PCV2 infection activates the cGAS/STING signaling pathway to induce IFN-ß production and the knockdown of cGAS and STING decreases viral replication in PK-15 cells. These results provide further insight into the relationship between PCV2 and host innate immunity.


Circovirus/immunology , Interferon-beta/immunology , Membrane Proteins/immunology , Nucleotidyltransferases/immunology , Signal Transduction/immunology , Virus Replication/immunology , Animals , Cell Line , Circoviridae Infections/virology , Circovirus/genetics , Circovirus/physiology , Immunity, Innate , Interferon-beta/biosynthesis , Interferon-beta/genetics , Membrane Proteins/genetics , Membrane Proteins/metabolism , Nucleotides, Cyclic/genetics , Nucleotides, Cyclic/immunology , Nucleotidyltransferases/genetics , Nucleotidyltransferases/metabolism , Signal Transduction/genetics , Swine
19.
Virology ; 520: 75-82, 2018 07.
Article En | MEDLINE | ID: mdl-29793076

Interferon (IFN)-mediated antiviral response is an important part of host defense. Previous studies reported that porcine circovirus type 2 (PCV2) inhibits interferon production, but the mechanism is still poorly understood. In this study, PCV2 suppresses IFN-ß and IRF3 promoters and mRNA level of IFN-ß induced by ISD or Poly(I:C), but has no effect on the activation of AP-1 and NF-κB. Furthermore, PCV2 decreases the mRNA level of IFN-ß and IFN-ß promoter activity driven by STING, TBK1, IRF3, and IRF3/5D, and causes a reduction in the protein level of nuclear p-IRF3. In addition, PCV2 interrupts the interaction of KPNA3, rather than KPNA4, with p-IRF3. Overexpression of KPNA3 restores IFN-ß promoter activity. These results indicate that PCV2 disrupts the interaction of KPNA3 with p-IRF3 and blocks p-IRF3 translocation to the nucleus, thereby inhibiting IFN-ß induction in PK-15 cells.


Circovirus/physiology , Interferon-beta/genetics , alpha Karyopherins/metabolism , Animals , Cell Line , Circovirus/drug effects , Circovirus/genetics , Epithelial Cells/virology , Host-Pathogen Interactions , Interferon Regulatory Factor-3/genetics , NF-kappa B/genetics , Poly I-C/pharmacology , Promoter Regions, Genetic , Signal Transduction , Swine , alpha Karyopherins/genetics
20.
Vet Microbiol ; 211: 36-42, 2017 Nov.
Article En | MEDLINE | ID: mdl-29102119

Type I Interferons (IFNs) is known for its antiviral activity; however, it is surprising that in vitro treatment of IFN-α and IFN-γ enhanced the replication of porcine circovirus type 2 (PCV2), indicating a complex relationship between interferon and PCV2. To date, it remains poorly understood how the interferon is produced during PCV2 infection and whether the interferon induced by PCV2 itself can promote viral replication. In this study, PCV2 induced the up-regulation of IFN-ß in PK-15 cells, while treatment of PCV2-infected cells with the interferon regulatory factor-3 (IRF3) inhibitor, BX795, decreased the expression of IFN-ß, whereas treatment with the nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) inhibitor, BAY11-7082, did not. These findings indicate that PCV2 can induce IFN-ß production via the IRF3-mediated rather than the NF-κB-mediated signal pathway. Moreover, PCV2 increased the protein expression levels of phosphorylation-IRF3 (p-IRF3), mitochondria antiviral-signaling protein (MAVS), retinoic acid-inducible gene I (RIG-1) and melanoma differentiation-associated protein 5 (MDA-5), and the knockdown of RIG-1 and MDA-5 decreased the expression level of IFN-ß in PK-15 cells. Therefore, PCV2 induces IFN-ß production via the RIG-1/MDA-5/MAVS/IRF signaling pathway. Furthermore, the PCV2 load and PCV2 infectivity decreased after knockdown of RIG-1 and MDA-5, indicating that RIG-1 and MDA-5 signaling pathways contribute to PCV2 replication. In conclusion, PCV2 induces the production of IFN-ß via the RIG-1 and MDA-5 signaling pathways, and the IFN-ß produced during PCV2 infection facilitates viral replication. These results will help us further understand the pathogenic mechanisms of PCV2.


Circoviridae Infections/veterinary , Circovirus/physiology , Signal Transduction , Swine Diseases/virology , Virus Replication , Animals , Cell Line , Circoviridae Infections/immunology , Circoviridae Infections/virology , Circovirus/genetics , Circovirus/immunology , Circovirus/pathogenicity , Genes, Reporter , Interferon-beta/metabolism , Swine , Swine Diseases/immunology , Viral Load/veterinary
...