Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
1.
J Virol ; 98(3): e0170923, 2024 Mar 19.
Article in English | MEDLINE | ID: mdl-38305156

ABSTRACT

Tick-borne flaviviruses (TBFs) are transmitted to humans through milk and tick bites. Although a case of possible mother-to-child transmission of tick-borne encephalitis virus (TBEV) through breast milk has been reported, this route has not been confirmed in experimental models. Therefore, in this study, using type I interferon receptor-deficient A129 mice infected with Langat virus (LGTV), we aimed to demonstrate the presence of infectious virus in the milk and mammary glands of infected mice. Our results showed viral RNA of LGTV in the pup's stomach milk clots (SMCs) and blood, indicating that the virus can be transmitted from dam to pup through breast milk. In addition, we observed that LGTV infection causes tissue lesions in the mammary gland, and viral particles were present in mammary gland epithelial cells. Furthermore, we found that milk from infected mice could infect adult mice via the intragastric route, which has a milder infection process, longer infection time, and a lower rate of weight loss than other modes of infection. Specifically, we developed a nano-luciferase-LGTV reporter virus system to monitor the dynamics of different infection routes and observed dam-to-pup infection using in vivo bioluminescence imaging. This study provides comprehensive evidence to support breast milk transmission of TBF in mice and has helped provide useful data for studying TBF transmission routes.IMPORTANCETo date, no experimental models have confirmed mother-to-child transmission of tick-borne flavivirus (TBF) through breastfeeding. In this study, we used a mouse model to demonstrate the presence of infectious viruses in mouse breast milk and mammary gland epithelial cells. Our results showed that pups could become infected through the gastrointestinal route by suckling milk, and the infection dynamics could be monitored using a reporter virus system during breastfeeding in vivo. We believe our findings have provided substantial evidence to understand the underlying mechanism of breast milk transmission of TBF in mice, which has important implications for understanding and preventing TBF transmission in humans.


Subject(s)
Encephalitis Viruses, Tick-Borne , Encephalitis, Tick-Borne , Infectious Disease Transmission, Vertical , Mammary Glands, Animal , Milk , Animals , Female , Mice , Encephalitis Viruses, Tick-Borne/growth & development , Encephalitis Viruses, Tick-Borne/physiology , Encephalitis, Tick-Borne/transmission , Encephalitis, Tick-Borne/virology , Mammary Glands, Animal/virology , Milk/virology , Animals, Newborn/virology
2.
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
3.
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
4.
Arch Virol ; 167(8): 1649-1657, 2022 Aug.
Article in English | MEDLINE | ID: mdl-35661915

ABSTRACT

Porcine deltacoronavirus (PDCoV) and porcine epidemic diarrhea virus (PEDV) have often been detected simultaneously in piglets with coronavirus diarrhea. However, the intestinal immune response to the interaction between circulating PDCoV and PEDV is unknown. Therefore, this study was conducted to investigate the intestinal immunity of neonatal piglets that were exposed first to PDCoV and then to PEDV. The amounts and distribution of CD3+ T lymphocytes, B lymphocytes, and goblet cells (GCs) in the small intestine were analyzed by immunohistochemistry and periodic acid-Schiff staining, respectively. The expression levels of pattern recognition receptors and downstream mediator cytokines were analyzed by qPCR and ELISA. The results showed that the numbers of GCs, CD3+ T lymphocytes, and B lymphocytes in the duodenum and jejunum of the PDCoV + PEDV coinoculated piglets were increased compared with those of piglets inoculated with PEDV alone. The piglets in the PDCoV + PEDV group had significantly upregulated IFN-α and IFN-λ1 compared with the PEDV single-inoculated piglets. These results suggest that PDCoV + PEDV-coinfected piglets can activate intestinal antiviral immunity more strongly than piglets infected with PEDV alone, which provides new insight into the pathogenesis mechanism of swine enteric coronavirus coinfection that may be used for vaccination in the future.


Subject(s)
Coronavirus Infections , Coronavirus , Porcine epidemic diarrhea virus , Swine Diseases , Animals , Coronavirus Infections/epidemiology , Deltacoronavirus , Diarrhea/veterinary , Porcine epidemic diarrhea virus/physiology , Swine
5.
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
6.
J Neurovirol ; 26(5): 687-695, 2020 10.
Article in English | MEDLINE | ID: mdl-32671812

ABSTRACT

Pseudorabies virus (PRV) establishes a lifelong latent infection in swine trigeminal ganglion (TG) following acute infection. Increased corticosteroid levels, due to stress, increases the incidence of reactivation from latency. Muscle injection combined with intravenous deliver of the synthetic corticosteroid dexamethasone (DEX) consistently induces reactivation from latency in pigs. In this study, PRV-free piglets were infected with PRV. Viral shedding in nasal and ocular swabs demonstrated that PRV infection entered the latent period. The anti-PRV antibody was detected by enzyme-linked immunosorbent assay and the serum neutralization test, which suggested that the PRV could establish latent infection in the presence of humoral immunity. Immunohistochemistry and viral genome detection of TG neurons suggested that PRV was reactivated from latency. Viral gene expressions of IE180, EP0, VP16, and LLT-intron were readily detected at 3-h post-DEX treatment, but gB, a γ1 gene, was not detectable. The differentially expressed phosphorylated proteins of TG neurons were analyzed by ITRAQ coupled with LC-MS/MS, and p-EIF2S2 differentially expression was confirmed by western blot assay. Taken together, our study provides the evidence that typical gene expression in PRV reactivation from latency in TG is disordered compared with known lytic infection in epithelial cells.


Subject(s)
Dexamethasone/pharmacology , Gene Expression Regulation, Viral/drug effects , Herpesvirus 1, Suid/drug effects , Pseudorabies/virology , Swine Diseases/virology , Trigeminal Ganglion/drug effects , Virus Activation/drug effects , Animals , Antibodies, Viral/blood , Eye/virology , Glucocorticoids/pharmacology , Herpes Simplex Virus Protein Vmw65/genetics , Herpes Simplex Virus Protein Vmw65/immunology , Herpesvirus 1, Suid/genetics , Herpesvirus 1, Suid/immunology , Herpesvirus 1, Suid/pathogenicity , Immediate-Early Proteins/genetics , Immediate-Early Proteins/immunology , Immunity, Humoral/drug effects , Nasal Cavity/virology , Neurons/drug effects , Neurons/immunology , Neurons/virology , Pseudorabies/immunology , Pseudorabies/pathology , Swine , Swine Diseases/immunology , Swine Diseases/pathology , Trigeminal Ganglion/immunology , Trigeminal Ganglion/virology , Virus Latency/drug effects , Virus Shedding/drug effects
7.
J Nutr ; 149(6): 894-901, 2019 06 01.
Article in English | MEDLINE | ID: mdl-31070734

ABSTRACT

BACKGROUND: Selenium (Se) plays a protective role in aflatoxin B1 (AFB1)-induced splenic immunotoxicity in chicks. OBJECTIVE: This study was designed to reveal the underlying mechanism of Se-mediated protection against AFB1-induced splenic injury in broilers. METHODS: Four groups of 1-d-old Cobb male broilers (n = 5 cages/diet, 6 chicks/cage) were arranged in a 3-wk 2 × 2 factorial design trial whereby they were fed an Se-deficient, corn- and soy-based diet [base diet (BD), 36 µg Se/kg], BD plus 1.0 mg AFB1/kg, BD plus 0.3 mg Se/kg, or BD plus 1.0 mg AFB1/kg and 0.3 mg Se/kg (as 2-hydroxy-4-methylselenobutanoic acid). Serum and spleen were collected at week 3 to assay for cytokines, histology, redox status, selected inflammation- and apoptosis-related genes and proteins, and the selenogenome. RESULTS: Dietary AFB1 induced growth retardation and spleen injury, decreasing (P < 0.05) body weight gain, feed intake, feed conversion efficiency, and serum interleukin-1ß by 17.8-98.1% and increasing (P < 0.05) the spleen index and serum interleukin-6 by 37.6-113%. It also reduced the splenic lymphocyte number, the white pulp region, and histiocyte proliferation in Se-adequate groups. However, Se deficiency aggravated (P < 0.05) these AFB1-induced alterations by 16.2-103%. Moreover, Se deficiency decreased (P < 0.05) splenic glutathione peroxidase (GPX) activity and glutathione-S transferase and glutathione concentrations by 35.6-89.4% in AFB1-exposed groups. Furthermore, Se deficiency upregulated (P < 0.05) the apoptotic (Caspase 3 and Caspase 9) and antimicrobial (ß defensin 1 and 2) genes, but downregulated (P < 0.05) antiapoptotic (B-cell lymphoma 2) and inflammatory (E3 ubiquitin-protein ligase CBL-B) genes at the mRNA and/or protein level in AFB1 supplementation groups. Additionally, Se deficiency downregulated (P < 0.05) GPX3, thioredoxin reductase 1 (TXNRD 1), GPX4, and selenoprotein (SELENO) S, and upregulated (P < 0.05) SELENOT and SELENOU in spleen in AFB1 administered groups. CONCLUSIONS: Dietary Se deficiency exacerbated AFB1-induced spleen injury in chicks, partially through the regulation of oxidative stress, inflammatory and apoptotic signaling, and 6 selenoproteins.


Subject(s)
Aflatoxin B1/toxicity , Avian Proteins/genetics , Selenium/deficiency , Selenoproteins/genetics , Spleen/drug effects , Spleen/immunology , Animals , Animals, Newborn , Antioxidants/metabolism , Apoptosis/drug effects , Apoptosis/genetics , Apoptosis/immunology , Chickens , Gene Expression Regulation/drug effects , Inflammation/etiology , Inflammation/genetics , Inflammation/immunology , Male , Oxidation-Reduction , Signal Transduction/drug effects , Spleen/metabolism
8.
J Neuroinflammation ; 15(1): 275, 2018 Sep 21.
Article in English | MEDLINE | ID: mdl-30241539

ABSTRACT

BACKGROUND: Many viruses depend on the extensive membranous network of the endoplasmic reticulum (ER) for their translation, replication, and packaging. Certain membrane modifications of the ER can be a trigger for ER stress, as well as the accumulation of viral protein in the ER by viral infection. Then, unfolded protein response (UPR) is activated to alleviate the stress. Zika virus (ZIKV) is a mosquito-borne flavivirus and its infection causes microcephaly in newborns and serious neurological complications in adults. Here, we investigated ER stress and the regulating model of UPR in ZIKV-infected neural cells in vitro and in vivo. METHODS: Mice deficient in type I and II IFN receptors were infected with ZIKV via intraperitoneal injection and the nervous tissues of the mice were assayed at 5 days post-infection. The expression of phospho-IRE1, XBP1, and ATF6 which were the key markers of ER stress were analyzed by immunohistochemistry assay in vivo. Additionally, the nuclear localization of XBP1s and ATF6n were analyzed by immunohistofluorescence. Furthermore, two representative neural cells, neuroblastoma cell line (SK-N-SH) and astrocytoma cell line (CCF-STTG1), were selected to verify the ER stress in vitro. The expression of BIP, phospho-elF2α, phospho-IRE1, and ATF6 were analyzed through western blot and the nuclear localization of XBP1s was performed by confocal immunofluorescence microscopy. RT-qPCR was also used to quantify the mRNA level of the UPR downstream genes in vitro and in vivo. RESULTS: ZIKV infection significantly upregulated the expression of ER stress markers in vitro and in vivo. Phospho-IRE1 and XBP1 expression significantly increased in the cerebellum and mesocephalon, while ATF6 expression significantly increased in the mesocephalon. ATF6n and XBP1s were translocated into the cell nucleus. The levels of BIP, ATF6, phospho-elf2α, and spliced xbp1 also significantly increased in vitro. Furthermore, the downstream genes of UPR were detected to investigate the regulating model of the UPR during ZIKV infection in vitro and in vivo. The transcriptional levels of atf4, gadd34, chop, and edem-1 in vivo and that of gadd34 and chop in vitro significantly increased. CONCLUSION: Findings in this study demonstrated that ZIKV infection activates ER stress in neural cells. The results offer clues to further study the mechanism of neuropathogenesis caused by ZIKV infection.


Subject(s)
Activating Transcription Factor 6/metabolism , Membrane Proteins/metabolism , Neurons/metabolism , Protein Serine-Threonine Kinases/metabolism , X-Box Binding Protein 1/metabolism , Zika Virus Infection/metabolism , Zika Virus Infection/pathology , Animals , Brain/metabolism , Brain/pathology , Brain/virology , Cell Line, Transformed , Disease Models, Animal , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum/pathology , Endoplasmic Reticulum Stress/drug effects , Endoplasmic Reticulum Stress/genetics , Gene Expression Regulation, Viral/genetics , Gene Expression Regulation, Viral/physiology , Humans , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neurons/virology , Protein Serine-Threonine Kinases/genetics , Receptor, Interferon alpha-beta/deficiency , Receptor, Interferon alpha-beta/genetics , X-Box Binding Protein 1/genetics , Zika Virus/genetics , Zika Virus/physiology
9.
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
10.
Virology ; 597: 110128, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38861876

ABSTRACT

Porcine epidemic diarrhea virus (PEDV) remains one of the major causative microorganisms of viral diarrhea in piglets worldwide, with no approved drugs for treatment. We identified a natural molecule, flavonol, which is widely found in tea, vegetables and herbs. Subsequently, the antiviral activity of compound flavonol was evaluated in Vero cells and IPEC-J2 cells, and its anti-PEDV mechanism was analyzed by molecular docking and molecular dynamics. The results showed that flavonol could effectively inhibit viral progeny production, RNA synthesis and protein expression of PEDV strains in a dose-dependent manner. When flavonol was added simultaneously with viral infection in Vero cells, it demonstrated potent anti-PEDV activity by affecting the viral attachment and internalization phases. Similarly, in IPEC-J2 cells, flavonol effectively inhibited PEDV infection at different stages of infection, except for the release phase. Moreover, flavonol mainly interacts with PEDV Mpro through hydrogen bonds and hydrophobic forces, and the complex formed by it has high stability. Importantly, flavonol also showed broad-spectrum activity against other porcine enteric coronaviruses such as TGEV and PDCoV in vitro. These findings suggest that flavonol may exert antiviral effects by interacting with viral Mpro, thereby affecting viral replication. This means that flavonol is expected to become a potential drug to prevent or treat porcine enteric coronavirus.


Subject(s)
Antiviral Agents , Flavonols , Porcine epidemic diarrhea virus , Virus Replication , Porcine epidemic diarrhea virus/drug effects , Animals , Antiviral Agents/pharmacology , Antiviral Agents/chemistry , Flavonols/pharmacology , Chlorocebus aethiops , Swine , Vero Cells , Virus Replication/drug effects , Molecular Docking Simulation , Coronavirus Infections/virology , Coronavirus Infections/veterinary , Coronavirus Infections/drug therapy , Virus Internalization/drug effects , Swine Diseases/virology , Swine Diseases/drug therapy , Cell Line , Molecular Dynamics Simulation , Virus Attachment/drug effects
11.
Anal Chem ; 85(5): 2645-51, 2013 Mar 05.
Article in English | MEDLINE | ID: mdl-23391352

ABSTRACT

In this work, we reported a simple rapid and point-of-care magnetic immunofluorescence assay for avian influenza virus (AIV) and developed a portable experimental setup equipped with an optical fiber spectrometer and a microfluidic device. We achieved the integration of immunomagnetic target capture, concentration, and fluorescence detection in the microfluidic chip. By optimizing flow rate and incubation time, we could get a limit of detection low up to 3.7 × 10(4) copy/µL with a sample consumption of 2 µL and a total assay time of less than 55 min. This approach had proved to possess high portability, fast analysis, high specificity, high precision, and reproducibility with an intra-assay variability of 2.87% and an interassay variability of 4.36%. As a whole, this microfluidic system may provide a powerful platform for the rapid detection of AIV and may be extended for detection of other viral pathogens; in addition, this portable experimental setup enables the development of point-of-care diagnostic systems while retaining adequate sensitivity.


Subject(s)
Immunomagnetic Separation/instrumentation , Influenza A virus/isolation & purification , Microfluidic Analytical Techniques/instrumentation , Point-of-Care Systems , Animals , Influenza A Virus, H1N1 Subtype/isolation & purification , Influenza A Virus, H5N1 Subtype/isolation & purification , Limit of Detection , Magnetic Fields , Models, Theoretical , Optical Fibers , Quantum Dots , Spectrometry, Fluorescence , Time Factors
12.
Arch Virol ; 158(3): 717-21, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23160706

ABSTRACT

To analyze the epidemiology of PRRSV in Hubei Province of China, 668 serum samples collected from 14 pig-breeding farms were tested. We found that the PRRSV-positive rate was 5.24 % and that HP-PRRSV had become the dominant strain. To further investigate the genetic variation of PRRSV strains in this region, the complete gene sequences of nsp2, orf5, and orf7 from nine PRRSV strains collected during 2011-2012 were determined and compared with 33 known sequences. The results revealed that diverse HP-PRRSV strains are present in this region. An analysis of orf5 gene sequences showed that the strains collected during 2009-2010 formed a tightly clustered branch. When compared with the JXA1 strain, they had one mutation (V29 → A29) in a decoy epitope. Furthermore, we found that the number of potential N-glycosylation sites had apparently increased since 2006. These findings increase our knowledge of PRRSV epidemiology in Central China.


Subject(s)
Genetic Variation , Open Reading Frames , Porcine Reproductive and Respiratory Syndrome/epidemiology , Porcine Reproductive and Respiratory Syndrome/virology , Porcine respiratory and reproductive syndrome virus/classification , Porcine respiratory and reproductive syndrome virus/genetics , Viral Nonstructural Proteins/genetics , Amino Acid Sequence , Animals , Base Sequence , China/epidemiology , Genome, Viral , Phylogeny , RNA, Viral/genetics , Sequence Analysis, RNA , Swine , Swine Diseases , Viral Nonstructural Proteins/chemistry , Viral Proteins/chemistry , Viral Proteins/genetics
13.
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
14.
Microbiol Spectr ; 11(6): e0220923, 2023 Dec 12.
Article in English | MEDLINE | ID: mdl-37938022

ABSTRACT

IMPORTANCE: Understanding the role of the endoribonuclease non-structural protein 15 (nsp15) (EndoU) in coronavirus (CoV) infection and pathogenesis is essential for vaccine target discovery. Whether the EndoU activity of CoV nsp15, as a virulence-related protein, has a diverse effect on viral virulence needs to be further explored. Here, we found that the transmissible gastroenteritis virus (TGEV) and feline infectious peritonitis virus (FIPV) nsp15 proteins antagonize SeV-induced interferon-ß (IFN-ß) production in human embryonic kidney 293 cells. Interestingly, compared with wild-type infection, infection with EnUmt-TGEV or EnUmt-FIPV did not change the IFN-ß response or reduce viral propagation in immunocompetent cells. The results of animal experiments showed that EnUmt viruses did not reduce the clinical presentation and mortality caused by TGEV and FIPV. Our findings enrich the understanding of nsp15-mediated regulation of alpha-CoV propagation and virulence and reveal that the conserved functions of nonstructural proteins have diverse effects on the pathogenicity of CoVs.


Subject(s)
Coronavirus Infections , Coronavirus , Animals , Humans , Virulence , Endoribonucleases/metabolism , Uridylate-Specific Endoribonucleases
15.
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
16.
Anal Chem ; 84(5): 2358-65, 2012 Mar 06.
Article in English | MEDLINE | ID: mdl-22309154

ABSTRACT

In this work, robust approach for a highly sensitive point-of-care virus detection was established based on immunomagnetic nanobeads and fluorescent quantum dots (QDs). Taking advantage of immunomagnetic nanobeads functionalized with the monoclonal antibody (mAb) to the surface protein hemagglutinin (HA) of avian influenza virus (AIV) H9N2 subtype, H9N2 viruses were efficiently captured through antibody affinity binding, without pretreatment of samples. The capture kinetics could be fitted well with a first-order bimolecular reaction with a high capturing rate constant k(f) of 4.25 × 10(9) (mol/L)(-1) s(-1), which suggested that the viruses could be quickly captured by the well-dispersed and comparable-size immunomagnetic nanobeads. In order to improve the sensitivity, high-luminance QDs conjugated with streptavidin (QDs-SA) were introduced to this assay through the high affinity biotin-streptavidin system by using the biotinylated mAb in an immuno sandwich mode. We ensured the selective binding of QDs-SA to the available biotin-sites on biotinylated mAb and optimized the conditions to reduce the nonspecific adsorption of QDs-SA to get a limit of detection low up to 60 copies of viruses in 200 µL. This approach is robust for application at the point-of-care due to its very good specificity, precision, and reproducibility with an intra-assay variability of 1.35% and an interassay variability of 3.0%, as well as its high selectivity also demonstrated by analysis of synthetic biological samples with mashed tissues and feces. Moreover, this method has been validated through a double-blind trial with 30 throat swab samples with a coincidence of 96.7% with the expected results.


Subject(s)
Immunomagnetic Separation , Influenza A Virus, H9N2 Subtype/isolation & purification , Point-of-Care Systems , Animals , Antibodies/chemistry , Antibodies/immunology , Biotin/chemistry , Biotin/metabolism , Chickens , Feces/virology , Influenza A Virus, H9N2 Subtype/chemistry , Influenza A Virus, H9N2 Subtype/immunology , Influenza in Birds/virology , Kinetics , Liver/virology , Lung/virology , Microscopy, Confocal , Quantum Dots , Streptavidin/chemistry , Streptavidin/metabolism
17.
Infect Genet Evol ; 104: 105362, 2022 10.
Article in English | MEDLINE | ID: mdl-36084837

ABSTRACT

Porcine kobuvirus (PKV) infection is very common in both healthy pigs and diarrhea pigs throughout the world. However, there is no proof that it causes diarrhea, and little is known about its role in diarrhea. There are only a few reports concerning porcine kobuvirus separation at present, which makes investigating its invasion and pathogenesis mechanisms difficult. This study sequenced the entire genome of a porcine kobuvirus strain termed "Wuhan2020" after it was isolated from intestinal tissue samples of healthy piglets. The analysis results revealed that it shared the most resemblance with the WUH1 strain (89.5%) and belonged to the same evolutionary branch as the Hungarian strain S-1-SUN. The PKV was located using the in situ hybridization (ISH) approach, which revealed that it was colonized in intestinal villus epithelial cells and lymphocytes in the Peyer's patch. In general, we analyzed the genetic evolution of PKV, discovered PKV susceptible cells and determined PKV localization in the intestine of infected pigs, providing a reference for future research.


Subject(s)
Kobuvirus , Picornaviridae Infections , Swine Diseases , Animals , China , Diarrhea , Feces , Genomics , Intestines , Kobuvirus/genetics , Phylogeny , Picornaviridae Infections/veterinary , Sequence Analysis , Swine
18.
Antioxidants (Basel) ; 11(2)2022 Feb 06.
Article in English | MEDLINE | ID: mdl-35204204

ABSTRACT

Methionine, as an essential amino acid, play roles in antioxidant defense and the regulation of immune responses. This study was designed to determine the effects and mechanisms of increased consumption of methionine by sows and piglets on the capacity of the progeny to counteract lipopolysaccharide (LPS) challenge-induced injury in the liver and spleen of piglets. Primiparous sows (n = 10/diet) and their progeny were fed a diet that was adequate in sulfur amino acids (CON) or CON + 25% total sulfur amino acids as methionine from gestation day 85 to postnatal day 35. A total of ten male piglets were selected from each treatment and divided into 2 groups (n = 5/treatment) for a 2 × 2 factorial design [diets (CON, Methionine) and challenge (saline or LPS)] at 35 d old. After 24 h challenge, the piglets were euthanized to collect the liver and spleen for the histopathology, redox status, and gene expression analysis. The histopathological results showed that LPS challenge induced liver and spleen injury, while dietary methionine supplementation alleviated these damages that were induced by the LPS challenge. Furthermore, the LPS challenge also decreased the activities of GPX, SOD, and CAT and upregulated the mRNA and(or) protein expression of TLR4, MyD88, TRAF6, NOD1, NOD2, NF-kB, TNF-α, IL-8, p53, BCL2, and COX2 in the liver and (or) spleen. The alterations of GPX and SOD activities and the former nine genes were prevented or alleviated by the methionine supplementation. In conclusion, the maternal and neonatal dietary supplementation of methionine improved the ability of piglets to resist LPS challenge-induced liver and spleen injury, potentially through the increased antioxidant capacity and inhibition of TLR4 and NOD signaling pathway.

19.
Anal Chem ; 83(19): 7316-22, 2011 Oct 01.
Article in English | MEDLINE | ID: mdl-21838323

ABSTRACT

We have developed a new fluorescent immune ensemble probe comprised of a conjugated lower toxic water-soluble CdTe:Zn(2+) quantum dots (QDs) and Ru(bpy)(2)(mcbpy-O-Su-ester)(PF(6))(2)- antibody complex (Ru-Ab) for the dual-color determination of human enterovirus 71 (EV71) in homogeneous solution. EV71 monoantibody was easily covalently conjugated with Ru(bpy)(2)(mcbpy-O-Su-ester)(PF(6))(2) to form a stable complex Ru-Ab, which acted both as an effective quencher of QDs fluorescence and the capture probe of virus antigen EV71. Herein, the target EV71 can break up the low fluorescent ionic ensemble by antigen-antibody combination to set free the fluorescent QDs and restore the fluorescence of QDs whereas the fluorescence intensity of Ru-Ab remains the same. Thus, the determination of EV71 by the complex Ru-Ab and QDs can be realized via the restoration of QDs fluorescence upon addition of EV71 and even can be directly evaluated by the ratio of green-colored QDs fluorescence intensity to Ru-Ab red-colored fluorescence intensity. The green-colored fluorescence of QDs was very sensitive to the change of EV71 concentration, and its fluorescence intensity increased with the increase of EV71 concentration between 1.8 ng/mL and 12 µg/mL. With this method, EV71 was detected at subnanogram per milliliter concentration in the presence of 160 µg/mL bovine serum albumin. More importantly, this strategy can be used as a universal method for any protein or virus by changing conjugated antibodies in disease early diagnosis providing a fast and promising clinical approach for virus determination. In a word, a simple, fast, sensitive, and highly selective assay for EV71 has been described. It could be applied in real sample analysis with a satisfactory result. It was notable that the sensor could not only achieve rapid and precise quantitative determination of protein/virus by fluorescent intensity but also could be applied in semiquantitative protein/virus determination by digital visualization.


Subject(s)
Enterovirus A, Human/immunology , Enterovirus A, Human/isolation & purification , Fluorescence , Immunoassay/methods , Animals , Antibodies/chemistry , Antibodies/immunology , Antigen-Antibody Complex/chemistry , Antigen-Antibody Complex/immunology , Biosensing Techniques , Cattle , Cells, Cultured , Chlorocebus aethiops , Colorimetry , Humans , Hydrogen-Ion Concentration , Organometallic Compounds/chemistry , Quantum Dots , Serum Albumin, Bovine/chemistry , Spectrometry, Fluorescence , Vero Cells
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL