Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
1.
Virus Res ; 341: 199329, 2024 03.
Artigo em Inglês | MEDLINE | ID: mdl-38262568

RESUMO

Porcine deltacoronavirus (PDCoV) is a novel enteric coronavirus that can cause vomiting, watery diarrhea in pigs and the death of piglets. The open reading frame (ORF) 5 is one of the accessory genes in PDCoV genome and encodes an accessory protein NS6. To date, the function of NS6 is still unclear. In this study, the recombinant NS6 was successfully expressed in prokaryotic expression system and purified. To prepare monoclonal antibody (mAb), six-week-old female BALB/c mice were primed subcutaneously with purified NS6. A novel mouse mAb against NS6 was obtained and designated as 3D5. The isotype of 3D5 is IgG2b with kappa (κ) light chain. 3D5 can specifically recognizes the natural NS6 in swine testis (ST) cells infected with PDCoV and expressed NS6 in human embryonic kidney 293T (HEK 293T) cells transfected with mammalian vector. The minimal linear B cell epitope recognised by 3D5 on NS6 was 25VPELIDPLVK34 determined by peptide scanning and named EP-3D5. The sequence of EP-3D5 is completely conserved among PDCoV strains. Moreover, six to nine residues of EP-3D5 were identified to be conserved in non-PDCoV strains. These results provide valuable insights into the antigenic structure and function of NS6 in virus pathogenesis, and aid for the development of PDCoV epitope-associated diagnostics and vaccine design.


Assuntos
Infecções por Coronavirus , Doenças dos Suínos , Masculino , Camundongos , Suínos , Animais , Feminino , Humanos , Deltacoronavirus , Diarreia , Epitopos de Linfócito B , Infecções por Coronavirus/veterinária , Mamíferos
2.
Vet Microbiol ; 288: 109953, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-38118371

RESUMO

The discovery of antiviral molecules is crucial for controlling porcine deltacoronavirus (PDCoV). Previous studies have provided evidence that the IFN-inducible transmembrane protein 3 (IFITM3), which is coded by an interferon-stimulated gene, prevents the infections of a number of enveloped viruses. Nevertheless, the involvement of IFITM3 in PDCoV infection remains unexplored. In this study, it was observed that the overexpression of IFITM3 successfully restrictes the infection of PDCoV in cell cultures. Conversely, the suppression of IFITM3 facilitates the infection of PDCoV in IPI-2I and IPEC-J2 cells. Further studies revealed that IFITM3 limits the attachment phase of viral infection by interacting with the S1 subunit of the PDCoV Spike (S) protein. In addition, IFITM3 is verified as a member of the CD225 family, the GxxxG conserved motif of this family is important for it to limit PDCoV infection. In summary, this study reveals the mechanism of IFITM3 as an antiviral molecule to inhibit PDCoV infection, and also provides theoretical supports for screening effective anti-PDCoV drugs.


Assuntos
Infecções por Coronavirus , Coronavirus , Doenças dos Suínos , Suínos , Animais , Coronavirus/genética , Infecções por Coronavirus/veterinária , Glicoproteína da Espícula de Coronavírus/genética , Antivirais/metabolismo
3.
PLoS Pathog ; 19(10): e1011702, 2023 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-37801439

RESUMO

Coronaviruses (CoVs) are a family of the largest RNA viruses that typically cause respiratory, enteric, and hepatic diseases in animals and humans, imposing great threats to the public safety and animal health. Porcine deltacoronavirus (PDCoV), a newly emerging enteropathogenic coronavirus, causes severe diarrhea in suckling piglets all over the world and poses potential risks of cross-species transmission. Here, we use PDCoV as a model of CoVs to illustrate the reciprocal regulation between CoVs infection and host antiviral responses. In this study, downregulation of DNA polymerase delta interacting protein 3 (POLDIP3) was confirmed in PDCoV infected IPEC-J2 cells by isobaric tags for relative and absolute quantification (iTRAQ) and Western blotting analysis. Overexpression of POLDIP3 inhibits PDCoV infection, whereas POLDIP3 knockout (POLDIP3-/-) by CRISPR-Cas9 editing significantly promotes PDCoV infection, indicating POLDIP3 as a novel antiviral regulator against PDCoV infection. Surprisingly, an antagonistic strategy was revealed that PDCoV encoded nonstructural protein 5 (nsp5) was responsible for POLDIP3 reduction via its 3C-like protease cleavage of POLDIP3 at the glutamine acid 176 (Q176), facilitating PDCoV infection due to the loss of antiviral effects of the cleaved fragments. Consistent with the obtained data in IPEC-J2 cell model in vitro, POLDIP3 reduction by cleavage was also corroborated in PDCoV infected-SPF piglets in vivo. Collectively, we unveiled a new antagonistic strategy evolved by PDCoV to counteract antiviral innate immunity by nsp5-mediated POLDIP3 cleavage, eventually ensuring productive virus replication. Importantly, we further demonstrated that nsp5s from PEDV and TGEV harbor the conserved function to cleave porcine POLDIP3 at the Q176 to despair POLDIP3-mediated antiviral effects. In addition, nsp5 from SARS-CoV-2 also cleaves human POLDIP3. Therefore, we speculate that coronaviruses employ similar POLDIP3 cleavage mechanisms mediated by nsp5 to antagonize the host antiviral responses to sustain efficient virus infection.


Assuntos
Infecções por Coronavirus , Doenças dos Suínos , Animais , Humanos , Suínos , Imunidade Inata , Replicação Viral , Antivirais , Proteínas de Ligação a RNA
4.
PLoS Pathog ; 19(3): e1011201, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36888569

RESUMO

Autophagy plays an important role in the infectious processes of diverse pathogens. For instance, cellular autophagy could be harnessed by viruses to facilitate replication. However, it is still uncertain about the interplay of autophagy and swine acute diarrhea syndrome coronavirus (SADS-CoV) in cells. In this study, we reported that SADS-CoV infection could induce a complete autophagy process both in vitro and in vivo, and an inhibition of autophagy significantly decreased SADS-CoV production, thus suggesting that autophagy facilitated the replication of SADS-CoV. We found that ER stress and its downstream IRE1 pathway were indispensable in the processes of SADS-CoV-induced autophagy. We also demonstrated that IRE1-JNK-Beclin 1 signaling pathway, neither PERK-EIF2S1 nor ATF6 pathways, was essential during SADS-CoV-induced autophagy. Importantly, our work provided the first evidence that expression of SADS-CoV PLP2-TM protein induced autophagy through the IRE1-JNK-Beclin 1 signaling pathway. Furthermore, the interaction of viral PLP2-TMF451-L490 domain and substrate-binding domain of GRP78 was identified to activate the IRE1-JNK-Beclin 1 signaling pathway, and thus resulting in autophagy, and in turn, enhancing SADS-CoV replication. Collectively, these results not only showed that autophagy promoted SADS-CoV replication in cultured cells, but also revealed that the molecular mechanism underlying SADS-CoV-induced autophagy in cells.


Assuntos
Chaperona BiP do Retículo Endoplasmático , Papaína , Papaína/metabolismo , Proteína Beclina-1 , Peptídeo Hidrolases/metabolismo , Autofagia , Transdução de Sinais , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo
5.
J Virol ; 97(2): e0187122, 2023 02 28.
Artigo em Inglês | MEDLINE | ID: mdl-36648234

RESUMO

Pseudorabies virus (PRV) is a neurotropic virus causing obvious neurological disorders and reproductive failure in pigs. PRV entry into target cells is a complex multistep process initiated by interacting viral envelope glycoproteins with cellular receptors. In the current study, we found that thrombospondin 3 (THBS3) plays an important role in PRV entry into target cells, indicating that THBS3 is a new PRV coreceptor. To confirm this hypothesis, the knockdown of THBS3 in several permissive cells inhibited PRV primary infection, and overexpression of THBS3 in PK15 cells promoted PRV infection. CRISPR-Cas9 knockout markedly reduced PRV infection in PK15 cells. Antibodies against THBS3 blocked PRV infection in naturally permissive target cells. Moreover, soluble THBS3 protein neutralized the infectivity of PRV. Mechanistically, THBS3 interacted with the PRV gD via its N and C termini to facilitate PRV binding in permissive and nonpermissive cells. Also, in the absence of Nectin-1, THBS3 promoted cell-to-cell fusion mediated by virus glycoproteins. While THBS3 alone could not increase virus entry, overexpression of it in the presence of Nectin-1 promoted virus entry into CHO-K1 cells. Our results have identified THBS3 as a critical player in PRV binding and subsequent membrane fusion and entry. IMPORTANCE Herpesvirus entry occurs through a cascade of virus-cell interactions, and multiple surface glycoproteins play a role in virus binding and entry during the virus invasion process. Early studies showed that attachment to cells by PRV, as well as other alphaherpesviruses, is mediated by interactions between the viral glycoprotein gC and cell membrane proteoglycans carrying heparan sulfate chains (HSPGs). However, gD may also be involved in virus binding in an HSPG-independent manner. To date, the respective cellular receptors are still unknown. In this report, we identified a host molecule, THBS3, involved in gD-mediated PRV binding and subsequent membrane fusion and entry, which increases our understanding of the initial events in alpha herpesvirus infections.


Assuntos
Herpesvirus Suídeo 1 , Pseudorraiva , Ligação Viral , Internalização do Vírus , Animais , Cricetinae , Células CHO , Herpesvirus Suídeo 1/metabolismo , Herpesvirus Suídeo 1/patogenicidade , Nectinas/genética , Nectinas/metabolismo , Suínos , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/metabolismo , Técnicas de Silenciamento de Genes
6.
Vet Microbiol ; 274: 109553, 2022 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-36181744

RESUMO

Infection induces the production of proinflammatory cytokines and chemokines such as interleukin-8 (IL-8) and interleukin-6 (IL-6). Although they facilitate local antiviral immunity, their excessive release leads to life-threatening cytokine release syndrome, exemplified by the severe cases of coronavirus disease 2019 (COVID-19) caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. In the present study, we found that interleukin-8 (IL-8) was upregulated by PDCoV infection. We then demonstrated that PDCoV E protein induced IL-8 production and that the TM domain and the C-terminal domain of the E protein were important for IL-8 production. Subsequently, we showed here that deleting the AP-1 and NF-κB binding motif in porcine IL-8 promoter abrogated its activation, suggesting that IL-8 expression was dependent on AP-1 and NF-κB. Furthermore, PDCoV E induced IL-8 production, which was also dependent on the NF-κB pathway through activating nuclear factor p65 phosphorylation and NF-κB inhibitor alpha (IκBα) protein phosphorylation, as well as inducing the nuclear translocation of p65, eventually resulting in the promotion of IL-8 production. PDCoV E also activated c-fos and c-jun, both of which are members of the AP-1 family. These findings provide new insights into the molecular mechanisms of PDCoV-induced IL-8 production and help us further understand the pathogenesis of PDCoV infection.


Assuntos
COVID-19 , Doenças dos Suínos , Suínos , Animais , NF-kappa B/metabolismo , Inibidor de NF-kappaB alfa , Interleucina-6/genética , Fator de Transcrição AP-1/genética , Fator de Transcrição AP-1/metabolismo , Interleucina-8/genética , Interleucina-8/metabolismo , SARS-CoV-2 , COVID-19/veterinária , Citocinas , Antivirais/farmacologia
7.
Biomed Res Int ; 2022: 6790665, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36082155

RESUMO

Since the early 1980s when MRI imaging technology was put into clinical use, the number of MRI clinical tests has steadily increased by more than 10% every year. At the same time, exogenous MRI contrast agents have also been developed with the development of MRI technology. However, there are still challenges in the preparation of contrast agents for magnetic resonance imaging, such as how to prepare high-efficiency contrast agents with high stability and low biological toxicity. In order to study the contrast agent with simple preparation method, low cost, and good imaging effect, a magnetic resonance contrast agent was prepared by magnetic nanoparticles. By acting on magnetic resonance imaging detection method, and using polymer ligands to synthesize magnetic nanoparticles, experiments and tests of P(MA-alt-VAc) polymer ligand-modified magnetic nanoparticles were carried out. The experimental results showed that when nanoparticles containing different iron ion concentrations were incubated with DC 2.4 normal cells for 48 hours, the cell viability was still higher than 80% at concentrations up to 200 µm. It shows that the nanoparticle has high cell activity and good biological adaptability. The transverse relaxation (r 2) value of the nanoparticles in aqueous solution at 37°C and 1.5 T magnetic field is 231.1 m-1 s-1, which is much higher than that of PTMP-PMAA (r 2 = 35.1 mM-1 s-1), which is also more than five times the relaxation of SHU-555C (r 2 = 44 mM-1 s-1). It shows that the nanoparticles prepared in this paper have good effect and can be used as a contrast agent in human brain for magnetic resonance imaging.


Assuntos
Meios de Contraste , Nanopartículas , Humanos , Imageamento por Ressonância Magnética/métodos , Espectroscopia de Ressonância Magnética , Neuroimagem , Polímeros
8.
Transbound Emerg Dis ; 69(6): 3506-3517, 2022 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-36150417

RESUMO

In recent years, increasing numbers of cases of acute gastroenteritis caused by Group A rotavirus (RVA) G12 strains have been reported in humans from many countries around the world, but G12 RVA detection in animals is currently less reported. Pigs are an important animal reservoir of zoonotic RVs and a mixing vessel for RVs. In 2020, RVA infection cases in piglets increased in China, which attracted more attention. During an epidemiological survey, a new type of porcine G12P[7] strain (CN127) was detected in pig farms across several provinces. Complete genome analyses revealed that strain CN127 possessed a Wa-like backbone with a genotype constellation of G12-P[7]-I1-C1-M1-R1-A8-N1-T1-E1-H1. The A8 genotype is indicative of its porcine rotavirus origin. Sequence identities and phylogenetic analyses showed that the VP2, VP4, NSP1, NSP4 and NSP5 genes were most closely related to those of porcine rotaviruses, but the VP1, VP6, VP7 and NSP2-3 genes were most closely related to those of human rotaviruses. CN127 likely emerged due to genetic reassortment between porcine and human rotavirus. In vivo experiments showed that CN127 infection caused gastrointestinal tract lesions in piglets and histopathological changes in the lung, liver and mesenteric lymph nodes (MLNs). In the small intestine, RVA antigen was detected in the duodenum and jejunum but not in the ileum. In the extra-intestinal tissues, RVA antigen was detected in the lung but not in the MLNs. Viral RNA was detected in the intestinal and extra-intestinal tissues as well as blood. This study reveals that RVA G12P[7] may become an epidemic strain in China and also provides further evidence that cocirculating human and porcine strains could produce new genotype rotaviruses with high virulence in piglets.


Assuntos
Infecções por Rotavirus , Rotavirus , Doenças dos Suínos , Humanos , Suínos , Animais , Rotavirus/genética , Infecções por Rotavirus/epidemiologia , Infecções por Rotavirus/veterinária , Virulência , Filogenia , Genoma Viral , Genótipo
9.
Viruses ; 14(8)2022 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-36016405

RESUMO

Porcine deltacoronavirus (PDCoV) is a recently discovered enteropathogenic coronavirus and has caused significant economic impacts on the pork industry. Although studies have partly uncovered the molecular mechanism of PDCoV-host interaction, it requires further research. In this study, we explored the roles of Stromal Antigen 2 (STAG2) in PDCoV infection. We found that STAG2-deficient cells inhibited infection with vesicular stomatitis virus (VSV) and PDCoV, whereas restoration of STAG2 expression in STAG2-depleted (STAG2-/-) IPEC-J2 cells line restored PDCoV infection, suggesting that STAG2 is involved in the PDCoV replication. Furthermore, we found that STAG2 deficiency results in robust interferon (IFN) expression. Subsequently, we found that STAG2 deficiency results in the activation of JAK-STAT signaling and the expression of IFN stimulated gene (ISG), which establish an antiviral state. Taken together, the depletion of STAG2 activates the JAK-STAT signaling and induces the expression of ISG, thereby inhibiting PDCoV replication. Our study provides new insights and potential therapeutic targets for unraveling the mechanism of PDCoV replication.


Assuntos
Infecções por Coronavirus , Coronavirus , Doenças dos Suínos , Animais , Antivirais/metabolismo , Coronavirus/fisiologia , Deltacoronavirus , Interferons/metabolismo , Suínos
10.
J Virol ; 96(5): e0088921, 2022 03 09.
Artigo em Inglês | MEDLINE | ID: mdl-34495699

RESUMO

Porcine epidemic diarrhea virus (PEDV) causes a porcine disease associated with swine epidemic diarrhea. Different antagonistic strategies have been identified, and the mechanism by which PEDV infection impairs the production of interferon (IFN) and delays the activation of the IFN response to escape host innate immunity has been determined, but the pathogenic mechanisms of PEDV infection remain enigmatic. Our preliminary results revealed that endogenous F-box and WD repeat domain-containing 7 (FBXW7) protein, the substrate recognition component of the SCF-type E3 ubiquitin ligase, is downregulated in PEDV-infected Vero E6 cells, according to the results from an isobaric tags for relative and absolute quantification (iTRAQ) analysis. Overexpression of FBXW7 in target cells makes them more resistant to PEDV infection, whereas ablation of FBXW7 expression by small interfering RNA (siRNA) significantly promotes PEDV infection. In addition, FBXW7 was verified as an innate antiviral factor capable of enhancing the expression of RIG-I and TBK1, and it was found to induce interferon-stimulated genes (ISGs), which led to an elevated antiviral state of the host cells. Moreover, we revealed that PEDV nonstructural protein 2 (nsp2) interacts with FBXW7 and targets FBXW7 for degradation through the K48-linked ubiquitin-proteasome pathway. Consistent with the results proven in vitro, FBXW7 reduction was also confirmed in different intestinal tissues from PEDV-infected specific-pathogen-free (SPF) pigs. Taken together, the data indicated that PEDV has evolved with a distinct antagonistic strategy to circumvent the host antiviral response by targeting the ubiquitin-proteasome-mediated degradation of FBXW7. Our findings provide novel insights into PEDV infection and pathogenesis. IMPORTANCE To counteract the host antiviral defenses, most viruses, including coronaviruses, have evolved with diverse strategies to dampen host IFN-mediated antiviral response, by interfering with or evading specific host regulators at multiple steps of this response. In this study, a novel antagonistic strategy was revealed showing that PEDV infection could circumvent the host innate response by targeted degradation of endogenous FBXW7 in target cells, a process that was verified to be a positive modulator for the host innate immune system. Degradation of FBXW7 hampers host innate antiviral activation and facilitates PEDV replication. Our findings reveal a new mechanism exploited by PEDV to suppress the host antiviral response.


Assuntos
Infecções por Coronavirus/veterinária , Proteína 7 com Repetições F-Box-WD/metabolismo , Evasão da Resposta Imune , Imunidade Inata , Vírus da Diarreia Epidêmica Suína/imunologia , Doenças dos Suínos/imunologia , Animais , Antivirais/imunologia , Chlorocebus aethiops , Infecções por Coronavirus/imunologia , Infecções por Coronavirus/prevenção & controle , Infecções por Coronavirus/virologia , Interferon Tipo I/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Transdução de Sinais/imunologia , Suínos , Doenças dos Suínos/prevenção & controle , Doenças dos Suínos/virologia , Ubiquitinas/metabolismo , Células Vero
11.
Vet Microbiol ; 264: 109299, 2022 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-34896854

RESUMO

Porcine deltacoronavirus (PDCoV) is an emerging porcine enteric coronavirus that causes severe diarrhea in piglets and results in serious economic losses. There are no effective vaccines and antiviral drugs to prevent and treat PDCoV infection currently. Griffithsin (GRFT) is a lectin with potent antiviral activity against enveloped viruses because of its ability to specifically bind N-linked high-mannose oligosaccharides. GRFT has been reported to possess antiviral activity against severe acute respiratory syndrome coronavirus (SARS-CoV), Middle East respiratory syndrome coronavirus (MERS-CoV), and porcine epidemic diarrhea virus (PEDV). Here, we first confirmed the antiviral activity of GRFT against PDCoV in vitro. The infected cells (%) and virus titers were significantly decreased at concentration 1 µg/mL or above of GRFT. Time-course experiments revealed that GRFT inhibits PDCoV infection at the adsorption and penetration step. GRFT binding to PDCoV spike (S) protein on the surface wraps the virus and blocks its entry. The outstanding antiviral potency indicates that GRFT has the potential value as a candidate drug for the prevention and treatment of PDCoV infection.


Assuntos
Deltacoronavirus , Lectinas de Plantas , Animais , Antivirais/farmacologia , Técnicas de Cultura de Células/veterinária , Infecções por Coronavirus/tratamento farmacológico , Infecções por Coronavirus/veterinária , Deltacoronavirus/efeitos dos fármacos , Lectinas de Plantas/farmacologia , Suínos , Doenças dos Suínos/tratamento farmacológico
12.
Front Vet Sci ; 9: 1083605, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36619958

RESUMO

Swine enteric coronavirus (SeCoV) causes acute diarrhea, vomiting, dehydration, and high mortality in neonatal piglets, causing severe losses worldwide. SeCoV includes the following four members: transmissible gastroenteritis virus (TGEV), porcine epidemic diarrhea virus (PEDV), porcine delta coronavirus (PDCoV), and swine acute diarrhea syndrome coronavirus (SADS-CoV). Clinically, mixed infections with several SeCoVs, which are more common in global farms, cause widespread infections. It is worth noting that PDCoV has a broader host range, suggesting the risk of PDCoV transmission across species, posing a serious threat to public health and global security. Studies have begun to focus on investigating the interaction between SeCoV and its host. Here, we summarize the effects of viral proteins on apoptosis, autophagy, and innate immunity induced by SeCoV, providing a theoretical basis for an in-depth understanding of the pathogenic mechanism of coronavirus.

13.
J Virol ; 95(21): e0124621, 2021 10 13.
Artigo em Inglês | MEDLINE | ID: mdl-34379449

RESUMO

Rotaviruses are the causative agents of severe and dehydrating gastroenteritis in children, piglets, and many other young animals. They replicate their genomes and assemble double-layered particles in cytoplasmic electron-dense inclusion bodies called "viroplasms." The formation of viroplasms is reportedly associated with the stability of microtubules. Although material transport is an important function of microtubules, whether and how microtubule-based transport influences the formation of viroplasms are still unclear. Here, we demonstrate that small viroplasms move and fuse in living cells. We show that microtubule-based dynein transport affects rotavirus infection, viroplasm formation, and the assembly of transient enveloped particles (TEPs) and triple-layered particles (TLPs). The dynein intermediate chain (DIC) is shown to localize in the viroplasm and to interact directly with nonstructural protein 2 (NSP2), indicating that the DIC is responsible for connecting the viroplasm to dynein. The WD40 repeat domain of the DIC regulates the interaction between the DIC and NSP2, and the knockdown of the DIC inhibited rotaviral infection, viroplasm formation, and the assembly of TEPs and TLPs. Our findings show that rotavirus viroplasms hijack dynein transport for fusion events, required for maximal assembly of infectious viral progeny. This study provides novel insights into the intracellular transport of viroplasms, which is involved in their biogenesis. IMPORTANCE Because the viroplasm is the viral factory for rotavirus replication, viroplasm formation undoubtedly determines the effective production of progeny rotavirus. Therefore, an understanding of the virus-host interactions involved in the biogenesis of the viroplasm is critical for the future development of prophylactic and therapeutic strategies. Previous studies have reported that the formation of viroplasms is associated with the stability of microtubules, whereas little is known about its specific mechanism. Here, we demonstrate that rotavirus viroplasm formation takes advantage of microtubule-based dynein transport mediated by an interaction between NSP2 and the DIC. These findings provide new insight into the intracellular transport of viroplasms.


Assuntos
Dineínas/metabolismo , Proteínas de Ligação a RNA/metabolismo , Infecções por Rotavirus/virologia , Rotavirus/fisiologia , Proteínas não Estruturais Virais/metabolismo , Compartimentos de Replicação Viral/metabolismo , Animais , Linhagem Celular , Chlorocebus aethiops , Células HEK293 , Interações entre Hospedeiro e Microrganismos , Humanos , Microtúbulos/metabolismo , Domínios Proteicos , Transporte Proteico , Suínos , Imagem com Lapso de Tempo , Montagem de Vírus , Replicação Viral
14.
Viruses ; 12(6)2020 05 31.
Artigo em Inglês | MEDLINE | ID: mdl-32486349

RESUMO

Porcine epidemic diarrhea virus (PEDV) causes a porcine disease associated with swine epidemic diarrhea. The type I interferon (IFN-I or IFN α/ß) is a key mediator of innate antiviral response during virus infection. Different antagonistic strategies have been identified and determined as to how PEDV infection inhibits the host's IFN responses to escape the host innate immune pathway, but the pathogenic mechanisms of PEDV infection are not fully elucidated. Our preliminary results revealed that endogenous TANK-binding kinase 1 (TBK1) and interferon regulatory factor 3 (IRF3), the key components in the IFN signaling pathway were downregulated in PEDV infected IPEC-J2 cells by iTRAQ analysis. In this study, we screened nsp15 as the most important viral encoded protein involved in TBK1 and IRF3 reduction. Endoribonuclease (EndoU) activity has been well determined for coronavirus nsp15. Three residues (H226, H241, and K282) of PEDV nsp15 were identified as critical amino acids for PEDV EndoU but not D265, which was not well correlated with published results of other coronaviruses, such as severe acute respiratory syndrome virus (SARS-CoV). Moreover, PEDV nsp15 can directly degrade the RNA levels of TBK1 and IRF3 dependent on its EndoU activity to suppress IFN production and constrain the induction of IFN stimulated genes (ISGs), by which PEDV antagonizes the host innate response to facilitate its replication. Collectively, these results have confirmed that PEDV nsp15 was capable of subverting the IFN response by the RNA degradation of TBK1 and IRF3.


Assuntos
Endorribonucleases/imunologia , Fator Regulador 3 de Interferon/metabolismo , Interferon Tipo I/imunologia , Vírus da Diarreia Epidêmica Suína/imunologia , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas não Estruturais Virais/imunologia , Animais , Linhagem Celular , Chlorocebus aethiops , Infecções por Coronavirus/patologia , Infecções por Coronavirus/veterinária , Regulação para Baixo , Células HEK293 , Humanos , Fator Regulador 3 de Interferon/genética , Interferon Tipo I/antagonistas & inibidores , Proteínas Serina-Treonina Quinases/genética , Estabilidade de RNA/genética , RNA Mensageiro/metabolismo , Transdução de Sinais/imunologia , Suínos , Doenças dos Suínos/imunologia , Doenças dos Suínos/patologia , Células Vero
15.
Mol Immunol ; 108: 68-74, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30784764

RESUMO

p53 is a tumor suppressor gene that can be activated in many contexts, such as DNA damage or stressful conditions. p53 has also been shown to be important for responses to certain viral infections. Porcine epidemic diarrhea virus (PEDV) is a major enteric pathogen of the coronavirus family that causes extensive mortality among piglets. The involvement of p53 during PEDV infection has not previously been investigated. In this study, we detected p53 upregulation in response to PEDV infection. Treatment with a p53 specific activator or p53 overexpression markedly decreased viral replication, and we showed that there was more viral progeny produced in p53 knock-out cells than in p53 wild-type cells. Finally, we demonstrated that inhibition of viral infection by p53 was mediated via p53-dependent IFN signaling, leading to IFN-stimulated response element (ISRE) activation, as well as the upregulation of IFN-stimulated genes (ISGs) and IFN-ß released from infected cells. These findings demonstrate that p53 suppresses PEDV infection, offering a novel therapeutic strategy for combatting this deadly disease in piglets.


Assuntos
Infecções por Coronavirus/imunologia , Infecções por Coronavirus/virologia , Imunidade , Interferons/metabolismo , Vírus da Diarreia Epidêmica Suína/fisiologia , Proteína Supressora de Tumor p53/metabolismo , Células HEK293 , Humanos , Vírus da Diarreia Epidêmica Suína/crescimento & desenvolvimento , Replicação Viral
16.
Int J Legal Med ; 133(2): 465-466, 2019 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-29737420

RESUMO

Using AGCU Y24 Plus PCR Amplification Kit, 32 Y short tandem repeat (STR) loci were analyzed in 355 unrelated male participants of Meizhou city in Guangdong Province of China. By analyzing 341 different haplotypes, it was found that haplotype diversity (HD) and discrimination capacity (DC) were 0.9995 and 0.9605, respectively. Population relationships were analyzed by comparing Hakka population with ten other populations. The results indicate that Meizhou Hakka population was closely related to Guangdong Han population. These data were valuable for both forensic applications and population genetics.


Assuntos
Cromossomos Humanos Y , Etnicidade/genética , Genética Populacional , Repetições de Microssatélites , Polimorfismo Genético , China , Haplótipos , Humanos , Masculino , Reação em Cadeia da Polimerase
17.
J Virol ; 92(8)2018 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-29386292

RESUMO

Porcine epidemic diarrhea virus (PEDV) causes acute and devastating enteric disease in suckling piglets and results in huge economic losses in the pig industry worldwide. To establish productive infection, viruses must first circumvent the host innate immune response. In this study, we found that PEDV infection stimulated epidermal growth factor receptor (EGFR) activation, which has been linked to not only anticancer therapeutics, but also antiviral signaling. Therefore, we determined whether EGFR activation affected PEDV infection by using an activator or overexpression assay. The data showed that EGFR activation enhanced virus replication in both cases. We also found that specific inhibition of EGFR by either inhibitors or small interfering RNA (siRNA) led to a decrease in virus yields. Further analysis revealed that inhibition of EGFR produced augmentation of type I interferon genes. We next observed that the EGFR downstream cascade STAT3 was also activated upon PEDV infection. Similar to the case of EGFR, specific inhibition of STAT3 by either inhibitor or siRNA increased the antiviral activity of interferon and resulted in decreased PEDV RNA levels, and vice versa. The data on STAT3 depletion in combination with EGFR activation suggest that the attenuation of antiviral activity by EGFR activation requires activation of the STAT3 signaling pathway. Taken together, these data demonstrate that PEDV-induced EGFR activation serves as a negative regulator of the type I interferon response and provides a novel therapeutic target for virus infection.IMPORTANCE EGFR is a transmembrane tyrosine receptor that mediates various cellular events, as well as several types of human cancers. In this study, we investigated for the first time the role of EGFR in PEDV infection. We observed that PEDV infection induced EGFR activation. The role of EGFR activation is to impair the antiviral activity of type I interferon, which requires the involvement of the EGFR downstream signaling cascade STAT3. Our findings reveal a new mechanism evolved by PEDV to circumvent the host antiviral response, which might serve as a therapeutic target against virus infection.


Assuntos
Infecções por Coronavirus/metabolismo , Infecções por Coronavirus/veterinária , Receptores ErbB/metabolismo , Interferon Tipo I/metabolismo , Vírus da Diarreia Epidêmica Suína/metabolismo , Transdução de Sinais , Animais , Infecções por Coronavirus/genética , Infecções por Coronavirus/patologia , Receptores ErbB/genética , Células HEK293 , Humanos , Interferon Tipo I/genética , Vírus da Diarreia Epidêmica Suína/genética , Fator de Transcrição STAT3/genética , Fator de Transcrição STAT3/metabolismo , Suínos
18.
Virology ; 517: 24-29, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29475600

RESUMO

Porcine epidemic diarrhea virus (PEDV) is a causative agent of porcine epidemic diarrhea (PED). PED, characterized by acute diarrhea, vomiting, dehydration, has caused serious economic losses in pig industry worldwide. Here, we demonstrate that activation of a disintergrin and metalloprotease 17 (ADAM17) induced the decrease of PEDV infection in HEK293 and IPEC-J2 cells and the downregulation of cell surface aminopeptidase N (APN) expression, an important entry factor for PEDV infection. Furthermore, overexpression of ADAM17 suppressed PEDV infection in HEK293 and IPEC-J2 cells, whereas ablation of ADAM17 expression using ADAM17 specific siRNA resulted in a corresponding increase of PEDV infection and an upregulation of cell surface APN expression. Taken together, these data demonstrate that modulation of APN expression by metalloprotease ADAM17 regulates PEDV infection. Hence, the reduction in APN expression represents another component of the anti-PEDV infection response initiated by ADAM17.


Assuntos
Proteína ADAM17/metabolismo , Antígenos CD13/metabolismo , Vírus da Diarreia Epidêmica Suína/fisiologia , Proteína ADAM17/genética , Animais , Linhagem Celular , Células Epiteliais/virologia , Humanos , Internalização do Vírus
19.
Arch Virol ; 162(8): 2415-2419, 2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28470417

RESUMO

Porcine epidemic diarrhea virus (PEDV), a causative agent of porcine epidemic diarrhea, causes economic loss in the global swine industry. Vero cell, an African green monkey kidney cell line, has been commonly used to isolate and propagate PEDV. However, since the production of interferon in these cells is defective, Vero cells are not the ideal cell type to study the molecular mechanisms of PEDV infection and the host antiviral innate immune response. In this study, we observed that human embryonic kidney 293 (HEK293) cells were susceptible to infection with PEDV vaccine strain CV777 (G1 subtype) and field isolate LNCT2 (G2 subtype). The one-step growth curve showed that the growth dynamics of PEDV in HEK293 cells was similar to that observed in Vero cells. Furthermore, we revealed that aminopeptidase N was involved in PEDV infection in HEK293 cells. Taken together, our findings suggest that HEK293 cells can be efficiently infected by PEDV, which might provide a useful tool for understanding the fundamental mechanisms of PEDV infection in vitro.


Assuntos
Vírus da Diarreia Epidêmica Suína/crescimento & desenvolvimento , Vírus da Diarreia Epidêmica Suína/fisiologia , Animais , Técnicas de Cultura de Células , Células HEK293 , Humanos , Vírus da Diarreia Epidêmica Suína/isolamento & purificação , Cultura de Vírus
20.
J Virol ; 91(10)2017 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-28275187

RESUMO

Porcine epidemic diarrhea virus (PEDV), the causative agent of porcine epidemic diarrhea, has caused huge economic losses in pig-producing countries. Although PEDV was long believed to replicate in the intestinal epithelium by using aminopeptidase N as a receptor, the mechanisms of PEDV infection are not fully characterized. In this study, we found that PEDV infection of epithelial cells results in disruption of the tight junctional distribution of occludin to its intracellular location. Overexpression of occludin in target cells makes them more susceptible to PEDV infection, whereas ablation of occludin expression by use of small interfering RNA (siRNA) in target cells significantly reduces their susceptibility to virus infection. However, the results observed with occludin siRNA indicate that occludin is not required for virus attachment. We conclude that occludin plays an essential role in PEDV infection at the postbinding stages. Furthermore, we observed that macropinocytosis inhibitors blocked occludin internalization and virus entry, indicating that virus entry and occludin internalization are closely coupled. However, the macropinocytosis inhibitors could not impede virus replication once the virus had entered host cells. This suggests that occludin internalization by macropinocytosis or a macropinocytosis-like process is involved in the virus entry events. Immunofluorescence confocal microscopy showed that PEDV was trapped at cellular junctional regions upon macropinocytosis inhibitor treatment, indicating that occludin may serve as a scaffold in the vicinity of virus entry. Collectively, these data show that occludin plays an essential role in PEDV infection during late entry events. Our observation may provide novel insights into PEDV infection and related pathogenesis.IMPORTANCE Tight junctions are highly specialized membrane domains whose main function is to attach adjacent cells to each other, thereby forming intercellular seals. Here we investigate, for the first time, the role of the tight junction protein occludin in PEDV infection. We observed that PEDV infection induced the internalization of occludin. By using genetic modification methods, we demonstrate that occludin plays an essential role in PEDV infection. Moreover, PEDV entry and occludin internalization seem to be closely coupled. Our findings reveal a new mechanism of PEDV infection.


Assuntos
Ocludina/metabolismo , Vírus da Diarreia Epidêmica Suína/fisiologia , Junções Íntimas/química , Ligação Viral , Internalização do Vírus , Animais , Linhagem Celular , Chlorocebus aethiops , Citoplasma/metabolismo , Células Epiteliais/virologia , Ocludina/deficiência , Ocludina/genética , Vírus da Diarreia Epidêmica Suína/efeitos dos fármacos , Vírus da Diarreia Epidêmica Suína/patogenicidade , RNA Interferente Pequeno , Suínos , Junções Íntimas/patologia , Junções Íntimas/virologia , Células Vero , Internalização do Vírus/efeitos dos fármacos , Replicação Viral/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...