Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 54
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
PLoS Biol ; 21(3): e3002039, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36930652

RESUMEN

Coronaviruses (CoVs) comprise a group of important human and animal pathogens. Despite extensive research in the past 3 years, the host innate immune defense mechanisms against CoVs remain incompletely understood, limiting the development of effective antivirals and non-antibody-based therapeutics. Here, we performed an integrated transcriptomic analysis of porcine jejunal epithelial cells infected with porcine epidemic diarrhea virus (PEDV) and identified cytidine/uridine monophosphate kinase 2 (CMPK2) as a potential host restriction factor. CMPK2 exhibited modest antiviral activity against PEDV infection in multiple cell types. CMPK2 transcription was regulated by interferon-dependent and interferon regulatory factor 1 (IRF1)-dependent pathways post-PEDV infection. We demonstrated that 3'-deoxy-3',4'-didehydro-cytidine triphosphate (ddhCTP) catalysis by Viperin, another interferon-stimulated protein, was essential for CMPK2's antiviral activity. Both the classical catalytic domain and the newly identified antiviral key domain of CMPK2 played crucial roles in this process. Together, CMPK2, viperin, and ddhCTP suppressed the replication of several other CoVs of different genera through inhibition of the RNA-dependent RNA polymerase activities. Our results revealed a previously unknown function of CMPK2 as a restriction factor for CoVs, implying that CMPK2 might be an alternative target of interfering with the viral polymerase activity.


Asunto(s)
Infecciones por Coronavirus , Coronavirus , Virus de la Diarrea Epidémica Porcina , Humanos , Animales , Porcinos , Interferones , Antivirales/farmacología , Proteínas/genética , Virus de la Diarrea Epidémica Porcina/genética
2.
J Virol ; 98(8): e0061124, 2024 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-39078151

RESUMEN

Porcine deltacoronavirus (PDCoV), an emerging enteropathogenic coronavirus, is a serious threat to piglets and has zoonotic potential. Here, we aimed to further explore the role of aminopeptidase N (APN) as a receptor for PDCoV and test the inhibitory effect of a chimeric APN protein strategy on PDCoV infection. PK-15 cells and LLC-PK1 cells expressing chimeric APN were selected and infected with PDCoV. Viral replication was significantly decreased in these chimeric APN cells compared with that in control group cells. To further characterize the effect of the chimeric APN strategy on PDCoV infection in vitro, primary intestinal epithelial cells isolated from chimeric APN pigs were inoculated with PDCoV. Viral challenge of these cells led to decreased PDCoV infection. More importantly, virally challenged chimeric APN neonatal piglets displayed reduced viral load, significantly fewer microscopic lesions in the intestinal tissue, and no diarrhea. Taken together, these findings deepen our understanding of the mechanism of PDCoV infection and provide a valuable model for the production of disease-resistant animals. IMPORTANCE: Porcine deltacoronavirus (PDCoV), an emerging enteropathogenic coronavirus, causes diarrhea in piglets and possesses the potential to infect humans. However, there are currently no effective measures for the prevention or control of PDCoV infection. Here, we have developed PK-15 cells, LLC-PK1 cells, and primary intestinal epithelial cells expressing chimeric APN, and viral challenge of these cells led to decreased PDCoV infection. Furthermore, virally challenged chimeric APN neonatal piglets displayed reduced viral load, significantly fewer microscopic lesions in the intestinal tissue, and no diarrhea. These data show that chimeric APN is a promising strategy to combat PDCoV infection.


Asunto(s)
Animales Recién Nacidos , Antígenos CD13 , Infecciones por Coronavirus , Deltacoronavirus , Enfermedades de los Porcinos , Replicación Viral , Animales , Porcinos , Antígenos CD13/genética , Antígenos CD13/metabolismo , Enfermedades de los Porcinos/virología , Deltacoronavirus/genética , Infecciones por Coronavirus/virología , Infecciones por Coronavirus/veterinaria , Infecciones por Coronavirus/prevención & control , Carga Viral , Edición Génica/métodos , Línea Celular , Células Epiteliales/virología , Diarrea/virología
3.
J Virol ; : e0053524, 2024 Aug 19.
Artículo en Inglés | MEDLINE | ID: mdl-39158273

RESUMEN

Coronaviruses (CoVs) are important pathogens for humans and other vertebrates, causing severe respiratory and intestinal infections that have become a threat to public health because of the potential for interspecies transmission between animals and humans. Therefore, the development of safe, effective vaccines remains a top priority for the control of CoV infection. The unique immunological characteristics of vaccines featuring messenger RNA (mRNA) present an advantageous tool for coronavirus vaccine development. Here, we designed two lipid nanoparticle (LNP)-encapsulated mRNA (mRNA-LNP) vaccines: one encoding full-length spike (S) protein and the other encoding the spike ectodomain (Se) from porcine deltacoronavirus (PDCoV). Fourteen days after primary immunization, both mRNA vaccines induced high levels of immunoglobulin G and neutralizing antibodies in mice, with the S vaccine showing better performance than the Se vaccine. Passive immune protection of the S mRNA vaccine in suckling piglets was confirmed by the induction of robust PDCoV-specific humoral and cellular immune responses. The S mRNA vaccine also showed better protective effects than the inactivated vaccine. Our results suggest that the novel PDCoV-S mRNA-LNP vaccine may have the potential to combat PDCoV infection. IMPORTANCE: As an emerging porcine enteropathogenic coronavirus, porcine deltacoronavirus (PDCoV) has the potential for cross-species transmission, attracting extensive attention. Messenger RNA (mRNA) vaccines are a promising option for combating emerging and re-emerging infectious diseases, as evidenced by the demonstrated efficacy of the COVID-19 mRNA vaccine. Here, we first demonstrated that PDCoV-S mRNA-lipid nanoparticle (LNP) vaccines could induce potent humoral and cellular immune responses in mice. An evaluation of passive immune protection of S mRNA vaccines in suckling piglets confirmed that the protective effect of mRNA vaccine was better than that of inactivated vaccine. This study suggests that the PDCoV-S mRNA-LNP vaccine may serve as a potential and novel vaccine candidate for combating PDCoV infection.

4.
J Immunol ; 210(3): 271-282, 2023 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-36548460

RESUMEN

Swine coronavirus-porcine epidemic diarrhea virus (PEDV) with specific susceptibility to pigs has existed for decades, and recurrent epidemics caused by mutant strains have swept the world again since 2010. In this study, single-cell RNA sequencing was used to perform for the first time, to our knowledge, a systematic analysis of pig jejunum infected with PEDV. Pig intestinal cell types were identified by representative markers and identified a new tuft cell marker, DNAH11. Excepting enterocyte cells, the goblet and tuft cells confirmed susceptibility to PEDV. Enrichment analyses showed that PEDV infection resulted in upregulation of cell apoptosis, junctions, and the MAPK signaling pathway and downregulation of oxidative phosphorylation in intestinal epithelial cell types. The T cell differentiation and IgA production were decreased in T and B cells, respectively. Cytokine gene analyses revealed that PEDV infection downregulated CXCL8, CXCL16, and IL34 in tuft cells and upregulated IL22 in Th17 cells. Further studies found that infection of goblet cells with PEDV decreased the expression of MUC2, as well as other mucin components. Moreover, the antimicrobial peptide REG3G was obviously upregulated through the IL33-STAT3 signaling pathway in enterocyte cells in the PEDV-infected group, and REG3G inhibited the PEDV replication. Finally, enterocyte cells expressed almost all coronavirus entry factors, and PEDV infection caused significant upregulation of the coronavirus receptor ACE2 in enterocyte cells. In summary, this study systematically investigated the responses of different cell types in the jejunum of piglets after PEDV infection, which deepened the understanding of viral pathogenesis.


Asunto(s)
Infecciones por Coronavirus , Virus de la Diarrea Epidémica Porcina , Porcinos , Animales , Virus de la Diarrea Epidémica Porcina/genética , Transcriptoma , Intestino Delgado/patología , Intestinos/patología , Análisis de Secuencia de ARN
5.
J Virol ; 97(11): e0095823, 2023 Nov 30.
Artículo en Inglés | MEDLINE | ID: mdl-37846983

RESUMEN

IMPORTANCE: As an emerging porcine enteropathogenic coronavirus that has the potential to infect humans, porcine deltacoronavirus (PDCoV) is receiving increasing attention. However, no effective commercially available vaccines against this virus are available. In this work, we designed a spike (S) protein and receptor-binding domain (RBD) trimer as a candidate PDCoV subunit vaccine. We demonstrated that S protein induced more robust humoral and cellular immune responses than the RBD trimer in mice. Furthermore, the protective efficacy of the S protein was compared with that of inactivated PDCoV vaccines in piglets and sows. Of note, the immunized piglets and suckling pig showed a high level of NAbs and were associated with reduced virus shedding and mild diarrhea, and the high level of NAbs was maintained for at least 4 months. Importantly, we demonstrated that S protein-based subunit vaccines conferred significant protection against PDCoV infection.


Asunto(s)
Infecciones por Coronavirus , Coronavirus , Enfermedades de los Porcinos , Vacunas de Subunidad , Animales , Femenino , Humanos , Ratones , Coronavirus/genética , Infecciones por Coronavirus/prevención & control , Infecciones por Coronavirus/veterinaria , Deltacoronavirus , Porcinos , Vacunas de Subunidad/administración & dosificación
6.
Microb Pathog ; 192: 106714, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38801864

RESUMEN

Porcine deltacoronavirus (PDCoV), a novel enteropathogenic coronavirus, causes diarrhea mainly in suckling piglets and has the potential to infect humans. Whereas, there is no commercially available vaccine which can effectively prevent this disease. In this study, to ascertain the duration of immune protection of inactivated PDCoV vaccine, suckling piglets were injected subcutaneously with inactivated PDCoV vaccine using a prime/boost strategy at 3 and 17-day-old. Neutralizing antibody assay showed that the level of the inactivated PDCoV group was still ≥1:64 at three months after prime vaccination. The three-month-old pigs were orally challenged with PDCoV strain CZ2020. Two pigs in challenge control group showed mild to severe diarrhea at 10-11 day-post-challenge (DPC), while the inactivated PDCoV group had no diarrhea. High levels of viral shedding, substantial intestinal villus atrophy, and positive straining of viral antigens in ileum were detected in challenge control group, while the pigs in inactivated PDCoV group exhibited significantly reduced viral load, minor intestinal villi damage and negative straining of viral antigens. These results demonstrated that PDCoV was pathogenic against three-month-old pigs and inactivated PDCoV vaccine can provide effective protection in pigs lasting for three months.


Asunto(s)
Anticuerpos Neutralizantes , Anticuerpos Antivirales , Infecciones por Coronavirus , Diarrea , Enfermedades de los Porcinos , Vacunas de Productos Inactivados , Vacunas Virales , Esparcimiento de Virus , Animales , Anticuerpos Neutralizantes/sangre , Anticuerpos Neutralizantes/inmunología , Vacunas de Productos Inactivados/inmunología , Vacunas de Productos Inactivados/administración & dosificación , Porcinos , Enfermedades de los Porcinos/prevención & control , Enfermedades de los Porcinos/inmunología , Enfermedades de los Porcinos/virología , Anticuerpos Antivirales/sangre , Anticuerpos Antivirales/inmunología , Vacunas Virales/inmunología , Vacunas Virales/administración & dosificación , Infecciones por Coronavirus/prevención & control , Infecciones por Coronavirus/inmunología , Infecciones por Coronavirus/veterinaria , Diarrea/prevención & control , Diarrea/inmunología , Diarrea/virología , Vacunación , Coronavirus/inmunología , Carga Viral , Antígenos Virales/inmunología
7.
Microb Pathog ; 190: 106612, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38467166

RESUMEN

Rotavirus group A (RVA) is a main pathogen causing diarrheal diseases in humans and animals. Various genotypes are prevalent in the Chinese pig herd. The genetic diversity of RVA lead to distinctly characteristics. In the present study, a porcine RVA strain, named AHFY2022, was successfully isolated from the small intestine tissue of piglets with severe diarrhea. The AHFY2022 strain was identified by cytopathic effects (CPE) observation, indirect immunofluorescence assay (IFA), electron microscopy (EM), high-throughput sequencing, and pathogenesis to piglets. The genomic investigation using NGS data revealed that AHFY2022 exhibited the genotypes G9-P[23]-I5-R1-C1-M1-A8-N1-T1-E1-H1, using the online platform the Bacterial and Viral Bioinformatics Resource Center (BV-BRC) (https://www.bv-brc.org/). Moreover, experimental inoculation in 5-day-old and 27-day-old piglets demonstrated that AHFY2022 caused severe diarrhea, fecal shedding, small intestinal villi damage, and colonization in all challenged piglets. Taken together, our results detailed the virological features of the porcine rotavirus G9P[23] from China, including the whole-genome sequences, genotypes, growth kinetics in MA104 cells and the pathogenicity in suckling piglets.


Asunto(s)
Diarrea , Genoma Viral , Genotipo , Filogenia , Infecciones por Rotavirus , Rotavirus , Enfermedades de los Porcinos , Animales , Rotavirus/genética , Rotavirus/aislamiento & purificación , Rotavirus/clasificación , Rotavirus/patogenicidad , Porcinos , Infecciones por Rotavirus/virología , Infecciones por Rotavirus/veterinaria , China , Enfermedades de los Porcinos/virología , Diarrea/virología , Diarrea/veterinaria , Intestino Delgado/virología , Intestino Delgado/patología , Heces/virología , Secuenciación de Nucleótidos de Alto Rendimiento
8.
Arch Virol ; 169(9): 180, 2024 Aug 16.
Artículo en Inglés | MEDLINE | ID: mdl-39150572

RESUMEN

Porcine epidemic diarrhea virus (PEDV) is an enteric coronavirus that has been the main cause of diarrhea in piglets since 2010 in China. The aim of this study was to investigate sequence variation and recombination events in the spike (S) gene of PEDV isolates from China. Thirty complete S gene sequences were obtained from PEDV-positive samples collected in six provinces in China from 2020 to 2023. Phylogenetic analysis showed that 10% (3/30) belonged to subtype GII-a, 6.67% (2/30) were categorized as subtype GII-b, 66.67% (20/30) were categorized as subtype GII-c, and 16.66% (5/30) were clustered with the S-INDEL strains. Amino acid sequence alignments showed that, when compared to strains of other subtypes, the GII-c strains had two characteristic amino acid substitutions (N139D and I289M). Five S-INDEL subtype strains had a single amino acid deletion (139N) and four amino acid substitutions (N118G, T137S, A138S, and D141G). Recombination analysis allowed six putative recombination events to be identified, one involving recombination between GII-c strains, two involving GII-c and GII-b strains, two involving GII-c and GI-a strains, and one involving GII-a and GI-b strains. These results suggest that recombination between PEDV strains has been common and complex in recent years and is one of the main reasons for the continuous variation of PEDV strains.


Asunto(s)
Infecciones por Coronavirus , Virus de la Diarrea Epidémica Porcina , Recombinación Genética , Glicoproteína de la Espiga del Coronavirus , Enfermedades de los Porcinos , Animales , Secuencia de Aminoácidos , Sustitución de Aminoácidos , China/epidemiología , Infecciones por Coronavirus/virología , Infecciones por Coronavirus/veterinaria , Infecciones por Coronavirus/epidemiología , Diarrea/virología , Diarrea/veterinaria , Diarrea/epidemiología , Variación Genética , Genotipo , Filogenia , Virus de la Diarrea Epidémica Porcina/genética , Virus de la Diarrea Epidémica Porcina/clasificación , Virus de la Diarrea Epidémica Porcina/aislamiento & purificación , Glicoproteína de la Espiga del Coronavirus/genética , Porcinos , Enfermedades de los Porcinos/virología , Enfermedades de los Porcinos/epidemiología
9.
Microb Pathog ; 169: 105642, 2022 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-35710089

RESUMEN

Caprine parainfluenza virus type 3 (CPIV3), a new strain of virus, was isolated from the goats in 2014 in China. Studies have shown that viral infection can induce changes in the expression profile of host miRNAs, which modulate natural immune responses and viral infection. In this study, we report that bta-miR-677 suppressed CPIV3 replication in Madin-Darby bovine kidney (MDBK) cells and guinea pigs. Bta-miR-677 overexpression promoted type I interferon (IFN-I) and IFN-stimulated genes (ISGs) production, thereby inhibiting CPIV3 replication, while bta-miR-677 inhibitor suppressed the antiviral innate immune response to promoted viral replication in MDBK cells. We showed that bta-miR-677 suppresses CPIV3 replication via directly targeted the 3'-untranslated region (3'-UTR) of mitochondrial antiviral signaling protein (MAVS) thus enhancing IFN pathway in MDBK cells. We also demonstrated that bta-miR-677 agomir could inhibit CPIV3 proliferation in guinea pigs, with much lower viral RNA levels in lung and trachea. Guinea pigs showed no obvious pathological changes and less severe lung lesions in bta-miR-677 agomir treated group at 7 dpi. This study contributes to our understanding of the molecular mechanisms underlying CPIV3 pathogenesis.


Asunto(s)
Interferón Tipo I , MicroARNs , Regiones no Traducidas 3' , Animales , Antivirales/farmacología , Bovinos , Línea Celular , Proliferación Celular , Cabras , Cobayas , Interferón Tipo I/genética , MicroARNs/genética , MicroARNs/metabolismo , Virus de la Parainfluenza 3 Humana/genética , Replicación Viral
10.
Microb Pathog ; 166: 105501, 2022 May.
Artículo en Inglés | MEDLINE | ID: mdl-35378245

RESUMEN

Pathogenic Escherichia coli (E. coli) can cause a variety of intestinal and extra-intestinal infections in humans and animals. Similar with the intestinal disease, respiratory disease is also a major threat to the breeding industry of goats. But the reports on respiratory disease associated E. coli are very limited. In this study, E. coli and other pathogens were examined for the 77 submitted respiratory cases. The serotypes, virulence genes, phylogenetic group and antimicrobial resistance characteristics of the E. coli isolates were identified. The results showed that 34 cases (44.16%) were associated with E. coli and 22 cases showed mixed infections of E. coli with Mycoplasma ovipneumoniae, Mannheimia haemolytica or Pasteurella . Among the 49 E. coli isolates, O8 (32.65%), O9 (20.41%) and O89 (10.20%) were the predominant serotypes (31/49, 63.27%). 22 virulence genes were detected and the most prevalent genes were fimH (100%), yijp (100%), mat (97.96%), ompA (95.92%) ibeB (91.84%) and fyuA (77.55%). In addition, ibeA was detected in 6.12% (3/49) of the strains. Markers of extra-intestinal pathogenic E. coli (ExPEC) were also identified and 14 strains were classified as ExPECs. 14 (28.57%), 25 (51.02%), 3 (6.12%) and 7 (14.29%) strains belonged to phylogenetic group A, B1, B2 and D, respectively, and group A and B1 were the predominant ones. The E. coli strains showed high resistant (48.98%-100%) to the 14 selected antimicrobials and all of them were defined as multiple drug resistant (MDR) strains. This is the first systemically study on E. coli of goats respiratory diseases origin in eastern China. The results suggest that E. coli infection may play an important role in goat respiratory diseases and that goats are reservoir hosts of ExPECs, which needs continuous monitoring in the future.


Asunto(s)
Infecciones por Escherichia coli , Escherichia coli , Animales , Antibacterianos/farmacología , Antibacterianos/uso terapéutico , Farmacorresistencia Bacteriana , Infecciones por Escherichia coli/tratamiento farmacológico , Infecciones por Escherichia coli/epidemiología , Infecciones por Escherichia coli/veterinaria , Cabras , Filogenia , Factores de Virulencia/genética
11.
Microb Pathog ; 170: 105723, 2022 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-35981694

RESUMEN

Porcine deltacoronavirus (PDCoV) is an emenging swine enteropathogenic coronavirus that can cause high mortality rate. It affects pigs of all ages, but most several in neonatal piglets. Little is known regarding the pathogenicity of PDCoV against 27-day-old piglets. In this study, 27-day-old piglets were experimentally infected with PDCoV CZ2020 from cell culture, the challenged piglets do not have obvious symptoms from 1 to 7 days post-challenge (DPC), while viral shedding was detected in rectal swab at 1 DPC. Tissues of small intestines displayed slight macroscopic and microscopic lesions with no viral antigen detection. On the other hand, 27-day-old piglets were infected with PDCoV from intestinal contents, the piglets developed mild to severe diarrhea, shedding increasing from 2 to 7 DPC, and developed macroscopic and microscopic lesions in small intestines with clear viral antigen confirmed by immunohistochemistry staining. Indicating the small intestine was still the major target organ in PDCoV-challenged pigs at the age of 27-day-old. Diarrhea caused by PDCoV from intestinal contents in 27-day-old piglets is less reported. Thus, our results might provide new insights into the pathogenesis of PDCoV.


Asunto(s)
Enfermedades de los Porcinos , Animales , Técnicas de Cultivo de Célula , Deltacoronavirus , Diarrea/patología , Contenido Digestivo , Porcinos , Virulencia
12.
J Gen Virol ; 101(7): 717-734, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32427096

RESUMEN

Caprine parainfluenza virus type 3 (CPIV3) is a novel important pathogen causing respiratory disease in goats, but the pathogenic mechanism is not clear yet. Evidence suggests that exosomes transfer biologically active molecules between cells. Viral infections can cause profound changes in exosome components, and exosomes have been involved in viral transmission and pathogenicity. In this study, we explored the characteristics and functions of exosomes purified from the supernatant of Madin-Darby bovine kidney (MDBK) cells inoculated with CPIV3. Infection of CPIV3 showed increased exosome secretion and the loading of viral proteins and RNA into exosomes. These exosomes were capable of transferring CPIV3 genetic materials to recipient cells to establish a productive infection and promote the viral replication. To explore the potential mechanism, small RNA deep sequencing revealed that CPIV3 exosomes contained a diverse range of RNA species, including miRNA and piRNA, in proportions different from exosomes isolated from mock-infected cells. Expression patterns of 11 differentially expressed miRNAs were subsequently validated by quantitative reverse transcriptase PCR (qRT-PCR). Targets of miRNAs were predicted and functional annotation analysis showed that the main pathways involved were autophagy signalling ways. Autophagy inhibited by the CPIV3-exosome was further verified, and miR-126-3 p_2 packaged in the vesicles was an important regulation factor in this process. Inhibition of autophagy may be one of the responsible reasons for promoting efficient replication of exosome-mediated CPIV3 infection. The study suggests that exosomes are key in pathogenesis or protection against CPIV3. Further understating of their role in CPIV3 infection may bring novel insight to the development of protection measures.


Asunto(s)
Autofagia , Exosomas/metabolismo , Enfermedades de las Cabras/metabolismo , Enfermedades de las Cabras/virología , Virus de la Parainfluenza 3 Humana/fisiología , Infecciones por Respirovirus/veterinaria , Animales , Línea Celular , Exosomas/ultraestructura , Regulación de la Expresión Génica , Enfermedades de las Cabras/genética , Cabras , Interacciones Huésped-Patógeno , MicroARNs/genética
13.
BMC Genomics ; 20(1): 516, 2019 Jun 21.
Artículo en Inglés | MEDLINE | ID: mdl-31226933

RESUMEN

BACKGROUND: Bovine viral diarrhea virus (BVDV) is an economically important viral pathogen of domestic and wild ruminants. Apart from cattle, small ruminants (goats and sheep) are also the susceptible hosts for BVDV. BVDV infection could interfere both of the innate and adaptive immunity of the host, while the genes and mechanisms responsible for these effects have not yet been fully understood. Peripheral blood mononuclear cells (PBMCs) play a pivotal role in the immune responses to viral infection, and these cells were the target of BVDV infection. In the present study, the transcriptome of goat peripheral blood mononuclear cells (PBMCs) infected with BVDV-2 was explored by using RNA-Seq technology. RESULTS: Goat PBMCs were successfully infected by BVDV-2, as determined by RT-PCR and quantitative real-time RT-PCR (qRT-PCR). RNA-Seq analysis results at 12 h post-infection (hpi) revealed 499 differentially expressed genes (DEGs, fold-change ≥ ± 2, p < 0.05) between infected and mock-infected PBMCs. Of these genes, 97 were up-regulated and the remaining 352 genes were down-regulated. The identified DEGs were found to be significantly enriched for locomotion/ localization, immune response, inflammatory response, defense response, regulation of cytokine production, etc., under GO enrichment analysis. Cytokine-cytokine receptor interaction, TNF signaling pathway, chemokine signaling pathway, etc., were found to be significantly enriched in KEGG pathway database. Protein-protein interaction (PPI) network analysis indicated most of the DEGs related to innate or adaptive immune responses, inflammatory response, and cytokine/chemokine-mediated signaling pathway. TNF, IL-6, IL-10, IL-12B, GM-CSF, ICAM1, EDN1, CCL5, CCL20, CXCL10, CCL2, MAPK11, MAPK13, CSF1R and LRRK1 were located in the core of the network and highly connected with other DGEs. CONCLUSIONS: BVDV-2 infection of goat PBMCs causes the transcription changes of a series of DEGs related to host immune responses, including inflammation, defense response, cell locomotion, cytokine/chemokine-mediated signaling, etc. The results will be useful for exploring and further understanding the host responses to BVDV-2 infection in goats.


Asunto(s)
Virus de la Diarrea Viral Bovina Tipo 2/genética , Enfermedades de las Cabras/inmunología , Enfermedades de las Cabras/virología , Infecciones por Pestivirus/veterinaria , Animales , Virus de la Diarrea Viral Bovina Tipo 2/inmunología , Perfilación de la Expresión Génica , Regulación Viral de la Expresión Génica , Enfermedades de las Cabras/genética , Cabras , Inmunidad/genética , Leucocitos Mononucleares/virología , Infecciones por Pestivirus/genética , Infecciones por Pestivirus/inmunología , Análisis de Secuencia de ARN , Replicación Viral
14.
Microb Pathog ; 137: 103795, 2019 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-31610219

RESUMEN

Paratyphoid fever caused by Salmonella Paratyphi A is a serious public health problem in many countries. In order to and develop a live attenuated candidate vaccine of Salmonella Paratyphi A, a Salmonella pathogenicity island 2 (SPI2, approximate 40 kb) deletion mutant of Salmonella Paratyphi A was constructed by lambda Red recombination, then the biological characteristics and protective ability of the Salmonella Paratyphi A SPI2 mutant were evaluated. Our results showed that the growth and biochemical properties of the SPI2 mutant were consistent with that of its parent strain, and the mutant was stable with the loss of SPI2. The mice lethal test showed that the virulence of the SPI2 mutant was significantly decreased, it can colonize and persistent more than 14 days in the liver and spleen of mice. Vaccination with the SPI2 mutant in mice revealed no significant effect on body weight and clinical symptoms compared to control animals, and specific humoral and cellular immune responses were also significantly induced. Immunization of mice offered efficient protection against Salmonella Paratyphi A strain challenge at 14 days post vaccination based on mortality and clinical symptoms relative to control group. Overall, these findings suggested that SPI2 plays an important role in pathogenicity of Salmonella Paratyphi A, and the SPI2 mutant showed its potential to develop a live attenuated vaccine candidate.


Asunto(s)
Islas Genómicas , Fiebre Paratifoidea/prevención & control , Vacunas contra la Salmonella/administración & dosificación , Salmonella paratyphi A/genética , Vacunas Tifoides-Paratifoides/administración & dosificación , Animales , Anticuerpos Antibacterianos/inmunología , Femenino , Humanos , Inmunización , Hígado/inmunología , Ratones , Ratones Endogámicos BALB C , Fiebre Paratifoidea/inmunología , Fiebre Paratifoidea/microbiología , Vacunas contra la Salmonella/genética , Vacunas contra la Salmonella/inmunología , Salmonella paratyphi A/inmunología , Salmonella paratyphi A/patogenicidad , Eliminación de Secuencia , Bazo/inmunología , Vacunas Tifoides-Paratifoides/genética , Vacunas Tifoides-Paratifoides/inmunología , Virulencia
15.
Microb Pathog ; 134: 103569, 2019 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-31163247

RESUMEN

Caprine parainfluenza virus type 3 (CPIV3) is one of the important viral respiratory tract agents in goats. The pathogenicity of CPIV3 has been examined in goats but it has not been explored in other laboratory animals. In the present study, an experimental infection of guinea pigs with CPIV3 was performed. The virus-inoculated guinea pigs displayed clinical signs related to the respiratory disease at 2-12 days post inoculation (dpi). Five infected guinea pigs died during 2 and 7 dpi. Apparent gross pneumonic lesions including consolidation and congestion in one or more lung lobes were observed in necropsied and dead animals. Histo-pathological changes in lungs including expansions of the alveolar interstitium, congestion, macrophage infiltration and compensatory emphysema were also observed. Virus was detectable at 2-10 dpi, 2-10 dpi and 2-7 dpi, as detected by virus isolation, real-time RT-PCR and immunohistochemistry staining, respectively. Viremia was also confirmed after CPIV3 infection during 3-7 dpi. The severe pathological lesions and highest viral load were observed before 7 dpi. Viral specific hemagglutination inhibition and neutralizing antibodies were produced from 7 dpi and 10 dpi, respectively, which related to the clearance of virus. The results present here indicated that guinea pig could be an ideal laboratory animal model for CPIV3 studies in the future.


Asunto(s)
Enfermedades de las Cabras/virología , Virus de la Parainfluenza 3 Humana/patogenicidad , Infecciones por Respirovirus/veterinaria , Animales , Anticuerpos Neutralizantes , Anticuerpos Antivirales , Modelos Animales de Enfermedad , Femenino , Cabras , Cobayas , Pulmón/patología , Pulmón/virología , Reacción en Cadena en Tiempo Real de la Polimerasa , Infecciones por Respirovirus/inmunología , Carga Viral , Viremia , Virulencia
16.
BMC Vet Res ; 15(1): 47, 2019 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-30709350

RESUMEN

BACKGROUND: Porcine circovirus type 2 (PCV2) is the causal agent of postweaning multisystemic wasting syndrome (PMWS), causing large economical losses of the global swine industry. Nitric oxide (NO), as an important signaling molecule, has antiviral activity on some viruses. To date, there is little information on the role of NO during PCV2 infection. RESULTS: We used indirect fluorescence assay (IFA), TCID50, real-time RT-qPCR and western blot assay to reveal the role of NO in restricting PCV2 replication. PCV2 replication was inhibited by a form of NO, NO•, whereas PCV2 was not susceptible to another form of NO, NO+. CONCLUSION: Our findings indicate that the form of NO• has a potential role in the fight against PCV2 infection.


Asunto(s)
Antivirales/farmacología , Circovirus/efectos de los fármacos , Óxido Nítrico/farmacología , Replicación Viral/efectos de los fármacos , Animales , Western Blotting , Línea Celular , Circovirus/fisiología , Relación Dosis-Respuesta a Droga , Técnica del Anticuerpo Fluorescente Indirecta , Radicales Libres , Técnicas In Vitro , Donantes de Óxido Nítrico/farmacología , Nitroprusiato/farmacología , Reacción en Cadena en Tiempo Real de la Polimerasa , Porcinos
17.
BMC Vet Res ; 15(1): 151, 2019 May 17.
Artículo en Inglés | MEDLINE | ID: mdl-31101113

RESUMEN

BACKGROUND: Caprine parainfluenza virus type 3 (CPIV3) is major pathogen of goat herds causing serious respiratory tract disease and economic losses to the goat industry in China. We analyzed the differential proteomics of CPIV3-infected Madin-Darby bovine kidney (MDBK) cells using quantitative iTRAQ coupled LC-MS/MS. In addition, four DEPs were validated by qRT-PCR and western blot analysis. RESULTS: Quantitative proteomics analysis revealed 163 differentially expressed proteins (DEPs) between CPIV3-infected and mock-infected groups (p-value < 0.05 and fold change > 1.2), among which 91 were down-regulated and 72 were up-regulated. Gene ontology (GO) analysis showed that these DEPs were involved in molecular functions, cellular components and biological processes. Biological functions in which the DEPs were involved in included diseases, genetic information processing, metabolism, environmental information processing, cellular processes, and organismal systems. STRING analysis revealed that four heat shock proteins (HSPs) included HSPA5, HSPA1B, HSP90B1 and HSPA6 may be associated with proliferation of CPIV3 in MDBK cells. qRT-PCR and western blot analysis showed that the selected HSPs were identical to the quantitative proteomics data. CONCLUSION: To our knowledge, this is the first report of the proteomic changes in MDBK cells after CPIV3 infection.


Asunto(s)
Proteínas de Choque Térmico/metabolismo , Proteómica , Infecciones por Respirovirus/veterinaria , Respirovirus/fisiología , Animales , Western Blotting , Bovinos , Línea Celular , Cromatografía Liquida , Perfilación de la Expresión Génica , Proteínas de Choque Térmico/genética , Riñón/virología , Reacción en Cadena en Tiempo Real de la Polimerasa , Respirovirus/genética , Infecciones por Respirovirus/genética , Infecciones por Respirovirus/metabolismo , Espectrometría de Masas en Tándem , Replicación Viral/fisiología
18.
Arch Virol ; 162(1): 235-246, 2017 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-27722993

RESUMEN

Porcine circovirus type 2 (PCV2) is subdivided into four genotypes: PCV2a, PCV2b, PCV2c and PCV2d. Here, for the first time, we compared the efficacy of two experimental inactivated chimeric PCV1-2 vaccines based on genotypes 2b and 2d. Seventeen 3-week-old pigs were divided randomly into four groups. Group 1 and 2 pigs were inoculated with genotype 2b- and 2d-based inactivated vaccines, respectively. At 28 days post-vaccination (DPV), pigs in groups 1-3 were challenged with the PCV2b 0233 strain. All experimental pigs were necropsied at 21 days post-challenge (DPC). Pigs vaccinated with the genotype 2b- or 2d-based vaccine had high antibody titres and lower PCV2b copy numbers in samples of sera, faeces and nasal secretions compared with pigs in the unvaccinated challenge group. Interestingly, we detected no DNA from the challenge strain in the superficial inguinal lymph nodes of the pigs immunized with the PCV2b vaccine, while one pig in the PCV2d- immunized group had detectable DNA from the challenge strain at 21 DPC. We found no significant differences in the humoral immune response, PCV2b load, or PCV-related microscopic lesions between the two vaccinated groups post-challenge. Therefore, both vaccines were equally effective at inducing immunity against challenge with PCV2b strain 0233.


Asunto(s)
Infecciones por Circoviridae/veterinaria , Circovirus/inmunología , Genotipo , Enfermedades de los Porcinos/prevención & control , Vacunas Virales/administración & dosificación , Vacunas Virales/inmunología , Animales , Anticuerpos Antivirales/sangre , Secreciones Corporales/virología , Infecciones por Circoviridae/prevención & control , Infecciones por Circoviridae/virología , Circovirus/clasificación , Circovirus/genética , Heces/virología , Suero/virología , Porcinos , Enfermedades de los Porcinos/virología , Resultado del Tratamiento , Vacunas de Productos Inactivados/administración & dosificación , Vacunas de Productos Inactivados/inmunología , Vacunas Sintéticas/administración & dosificación , Vacunas Sintéticas/inmunología , Carga Viral
19.
Virol J ; 12: 113, 2015 Jul 30.
Artículo en Inglés | MEDLINE | ID: mdl-26220290

RESUMEN

BACKGROUND: Porcine circovirus type-2b (PCV2b) is recognized as the etiological agent of the various clinical manifestations of porcine circovirus-associated disease (PCVAD). Previous studies have demonstrated effectiveness of chimeric PCV1-2 vaccines against PCV2b challenge. In this study, the efficacy of inactivated and live-attenuated (2 × 10(3.5) or 2 × 10(4.0) 50% tissue culture infective dose [TCID50] dose) chimeric PCV1-2b vaccines was compared side-by-side in conventional pigs. METHODS: Twenty-seven non-PCV2 viremic pigs without PCV2-specific antibody were randomly divided into six groups, including four vaccinated and challenged groups, a nonvaccinated challenged group, and a mock group. All pigs except those in the mock group were challenged at 28 days post vaccination (DPV) using PCV2b. RESULTS: Both inactivated and live-attenuated chimeric PCV1-2b vaccines induced a robust antibody responses, and significantly decreased microscopic lesion and lower viral loads in serum or superficial inguinal lymph nodes (SILN) compared with that in the nonvaccinated challenged group. PCV2 antibody titers decreased after 7 days post challenge (DPC) in pigs administered the inactivated PCV1-2b vaccine and they were lower than those in pigs inoculated with live-attenuated PCV1-2b on the day of necropsy. Moreover, no viremia was present in pigs inoculated with live-attenuated PCV1-2b vaccine at 21 DPC regardless of the dose difference. CONCLUSIONS: The results demonstrated that both inactivated and live-attenuated chimeric PCV1-2b vaccines were effective to induce protective immunity against PCV2b infection.


Asunto(s)
Infecciones por Circoviridae/veterinaria , Circovirus/inmunología , Enfermedades de los Porcinos/prevención & control , Vacunas Virales/administración & dosificación , Vacunas Virales/inmunología , Estructuras Animales/patología , Estructuras Animales/virología , Animales , Anticuerpos Antivirales/sangre , China , Infecciones por Circoviridae/inmunología , Infecciones por Circoviridae/patología , Infecciones por Circoviridae/prevención & control , Circovirus/genética , Circovirus/aislamiento & purificación , Datos de Secuencia Molecular , Análisis de Secuencia de ADN , Porcinos , Enfermedades de los Porcinos/inmunología , Enfermedades de los Porcinos/patología , Enfermedades de los Porcinos/virología , Resultado del Tratamiento , Vacunas Atenuadas/administración & dosificación , Vacunas Atenuadas/genética , Vacunas Atenuadas/inmunología , Vacunas de Productos Inactivados/administración & dosificación , Vacunas de Productos Inactivados/genética , Vacunas de Productos Inactivados/inmunología , Vacunas Sintéticas/administración & dosificación , Vacunas Sintéticas/genética , Vacunas Sintéticas/inmunología , Vacunas Virales/genética
20.
Vet Microbiol ; 297: 110211, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39096790

RESUMEN

Porcine deltacoronavirus (PDCoV), a cross-species transmissible enterovirus, frequently induces severe diarrhea and vomiting symptoms in piglets, which not only pose a significant menace to the global pig industry but also a potential public safety risk. In a previous study, we isolated a vaccine candidate, PDCoV CZ2020-P100, by passaging a parental PDCoV strain in vitro, exhibiting attenuated virulence and enhanced replication. However, the factors underlying these differences between primary and passaged strains remain unknown. In this study, we present the transcriptional landscapes of porcine kidney epithelial cells (LLC-PK1) cells infected with PDCoV CZ2020-P1 strain and P100 strain using the RNA-sequencing. We identified 105 differentially expressed genes (DEGs) in P1-infected cells and 295 DEGs in P100-infected cells. Enrichment analyses indicated that many DEGs showed enrichment in immune and inflammatory responses, with a more and higher upregulation of DEGs enriched in the P100-infected group. Notably, the DEGs were concentrated in the MAPK pathway within the P100-infected group, with significant upregulation in EphA2 and c-Fos. Knockdown of EphA2 and c-Fos reduced PDCoV infection and significantly impaired P100 replication compared to P1, suggesting a novel mechanism in which EphA2 and c-Fos are highly involved in passaged virus replication. Our findings illuminate the resemblances and distinctions in the gene expression patterns of host cells infected with P1 and P100, confirming that EphA2 and c-Fos play key roles in high-passage PDCoV replication. These results enhance our understanding of the changes in virulence and replication capacity during the process of passaging.


Asunto(s)
Deltacoronavirus , Receptor EphA2 , Transcriptoma , Replicación Viral , Animales , Porcinos , Deltacoronavirus/genética , Deltacoronavirus/fisiología , Deltacoronavirus/patogenicidad , Receptor EphA2/genética , Enfermedades de los Porcinos/virología , Proteínas Proto-Oncogénicas c-fos/genética , Proteínas Proto-Oncogénicas c-fos/metabolismo , Células LLC-PK1 , Línea Celular , Infecciones por Coronavirus/virología , Infecciones por Coronavirus/veterinaria
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA