Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
1.
J Virol ; 98(9): e0053524, 2024 Sep 17.
Article in English | MEDLINE | ID: mdl-39158273

ABSTRACT

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.


Subject(s)
Antibodies, Neutralizing , Antibodies, Viral , Coronavirus Infections , Spike Glycoprotein, Coronavirus , Swine Diseases , Viral Vaccines , Animals , Swine , Coronavirus Infections/prevention & control , Coronavirus Infections/immunology , Coronavirus Infections/veterinary , Coronavirus Infections/virology , Antibodies, Neutralizing/immunology , Antibodies, Neutralizing/blood , Spike Glycoprotein, Coronavirus/immunology , Spike Glycoprotein, Coronavirus/genetics , Mice , Swine Diseases/prevention & control , Swine Diseases/virology , Swine Diseases/immunology , Antibodies, Viral/immunology , Antibodies, Viral/blood , Viral Vaccines/immunology , Viral Vaccines/administration & dosage , mRNA Vaccines , Deltacoronavirus/immunology , Deltacoronavirus/genetics , Nanoparticles , RNA, Messenger/genetics , RNA, Messenger/immunology , Immunoglobulin G/blood , Immunoglobulin G/immunology , Mice, Inbred BALB C , Female , Immunity, Humoral , Liposomes
2.
Funct Integr Genomics ; 24(3): 79, 2024 Apr 24.
Article in English | MEDLINE | ID: mdl-38653845

ABSTRACT

Coronaviruses have been identified as pathogens of gastrointestinal and respiratory diseases in humans and various animal species. In recent years, the global spread of new coronaviruses has had profound influences for global public health and economies worldwide. As highly pathogenic zoonotic viruses, coronaviruses have become the focus of current research. Porcine Deltacoronavirus (PDCoV), an enterovirus belonging to the family of coronaviruses, has emerged on a global scale in the past decade and significantly influenced the swine industry. Moreover, PDCoV infects not only pigs but also other species, including humans, chickens and cattles, exhibiting a broad host tropism. This emphasizes the need for in-depth studies on coronaviruses to mitigate their potential threats. In this review, we provided a comprehensive summary of the current studies on PDCoV. We first reviewed the epidemiological investigations on the global prevalence and distribution of PDCoV. Then, we delved into the studies on the pathogenesis of PDCoV to understand the mechanisms how the virus impacts its hosts. Furthermore, we also presented some exploration studies on the immune evasion mechanisms of the virus to enhance the understanding of host-virus interactions. Despite current limitations in vaccine development for PDCoV, we highlighted the inhibitory effects observed with certain substances, which offers a potential direction for future research endeavors. In conclusion, this review summarized the scientific findings in epidemiology, pathogenesis, immune evasion mechanisms and vaccine development of PDCoV. The ongoing exploration of potential vaccine candidates and the insights gained from inhibitory substances have provided a solid foundation for future vaccine development to prevent and control diseases associated with PDCoV.


Subject(s)
Coronavirus Infections , Deltacoronavirus , Immune Evasion , Swine Diseases , Viral Vaccines , Animals , Swine , Coronavirus Infections/immunology , Coronavirus Infections/prevention & control , Coronavirus Infections/virology , Coronavirus Infections/epidemiology , Deltacoronavirus/pathogenicity , Deltacoronavirus/immunology , Deltacoronavirus/genetics , Swine Diseases/virology , Swine Diseases/immunology , Swine Diseases/prevention & control , Swine Diseases/epidemiology , Viral Vaccines/immunology , Vaccine Development , Humans
3.
J Virol ; 97(11): e0147023, 2023 Nov 30.
Article in English | MEDLINE | ID: mdl-37882521

ABSTRACT

IMPORTANCE: As a member of the δ-coronavirus family, porcine deltacoronavirus (PDCoV) is a vital reason for diarrhea in piglets, which can contribute to high morbidity and mortality rates. Initially identified in Hong Kong in 2012, the virus has rapidly spread worldwide. During PDCoV infection, the virus employs evasion mechanisms to evade host surveillance, while the host mounts corresponding responses to impede viral replication. Our research has revealed that PDCoV infection down-regulates the expression of PGAM5 to promote virus replication. In contrast, PGAM5 degrades PDCoV N through autophagy by interacting with the cargo receptor P62 and the E3 ubiquitination ligase STUB1. Additionally, PGAM5 interacts with MyD88 and TRAF3 to activate the IFN signal pathway, resulting in the inhibition of viral replication.


Subject(s)
Coronavirus Infections , Coronavirus Nucleocapsid Proteins , Deltacoronavirus , Interferon Type I , Mitochondrial Proteins , Phosphoprotein Phosphatases , Proteolysis , Swine Diseases , Swine , Virus Replication , Animals , Coronavirus Infections/immunology , Coronavirus Infections/veterinary , Coronavirus Infections/virology , Interferon Type I/immunology , Signal Transduction , Swine/virology , Swine Diseases/virology , Ubiquitin-Protein Ligases/metabolism , Virus Replication/immunology , Coronavirus Nucleocapsid Proteins/metabolism , Deltacoronavirus/immunology , Deltacoronavirus/metabolism , Phosphoprotein Phosphatases/metabolism , Mitochondrial Proteins/metabolism , Down-Regulation , Immune Evasion , RNA-Binding Proteins/metabolism
4.
Int J Mol Sci ; 25(14)2024 Jul 12.
Article in English | MEDLINE | ID: mdl-39062886

ABSTRACT

Porcine deltacoronavirus (PDCoV) is an emerging enteric pathogen that causes substantial economic losses in the swine industry worldwide. The PDCoV NS6 protein is an accessory protein that plays a pivotal role in the viral life cycle and immune evasion. However, the functions of NS6 and its role in PDCoV pathogenesis remain largely unknown. In this study, we prepared a monoclonal antibody (mAb) 5-A11 that specifically recognizes the PDCoV NS6 protein. The mAb 5-A11 exhibited high specificity for PDCoV, with no cross-reactivity with several major porcine pathogenic viruses. Furthermore, the epitope recognized by mAb 5-A11 was precisely mapped to residues 70EYGSIYGKDFI80 of the NS6 protein using Western blot analysis. Notably, this epitope is highly conserved among different PDCoV isolates. Substantial variations were observed when comparing this epitope with the corresponding regions in the NS6 proteins of other δ coronaviruses, suggesting potential differences in the structure, function, and antigenicity of their NS6 proteins. Our findings provide valuable tools and insights for further elucidating the functions of the NS6 protein and its role in PDCoV pathogenesis, as well as for developing diagnostic and therapeutic strategies against PDCoV infection.


Subject(s)
Antibodies, Monoclonal , Deltacoronavirus , Epitopes , Viral Nonstructural Proteins , Animals , Antibodies, Monoclonal/immunology , Swine , Deltacoronavirus/immunology , Viral Nonstructural Proteins/immunology , Viral Nonstructural Proteins/metabolism , Epitopes/immunology , Epitope Mapping , Coronavirus Infections/immunology , Coronavirus Infections/virology , Antibodies, Viral/immunology , Swine Diseases/virology , Swine Diseases/immunology , Mice , Amino Acid Sequence , Mice, Inbred BALB C
5.
J Virol ; 96(7): e0015822, 2022 04 13.
Article in English | MEDLINE | ID: mdl-35311551

ABSTRACT

Major histocompatibility complex class I (MHC-I) and MHC-II molecules, mainly being responsible for the processing and presentation of intracellular or extracellular antigen, respectively, are critical for antiviral immunity. Here, we reported that porcine deltacoronavirus (PDCoV) with the zoonotic potential and potential spillover from pigs to humans, upregulated the expressions of porcine MHC-I (swine leukocyte antigen class I, SLA-I) molecules and SLA-I antigen presentation associated genes instead of porcine MHC-II (SLA-II) molecules both in primary porcine enteroids and swine testicular (ST) cells at the late stage of infection, and this finding was verified in vivo. Moreover, the induction of SLA-I molecules by PDCoV infection was mediated through enhancing the expression of NOD-like receptor (NLR) family caspase recruitment domain-containing 5 (NLRC5). Mechanistic studies demonstrated that PDCoV infection robustly elevated retinoic acid-inducible gene I (RIG-I) expression, and further initiated the downstream type I interferon beta (IFN-ß) production, which led to the upregulation of NLRC5 and SLA-I genes. Likewise, interferon regulatory factor 1 (IRF1) elicited by PDCoV infection directly activated the promoter activity of NLRC5, resulting in an increased expression of NLRC5 and SLA-I upregulation. Taken together, our findings advance our understanding of how PDCoV manipulates MHC molecules, and knowledge that could help inform the development of therapies and vaccines against PDCoV. IMPORTANCE MHC-I molecules play a crucial role in antiviral immunity by presenting intracellular antigens to CD8+T lymphocytes and eliminating virus-infected cells by natural killer cells' "missing-self recognition." However, the manipulation of MHC molecules by coronaviruses remains poorly understood. Here, we demonstrated that PDCoV, a zoonotic potential coronavirus efficiently infecting cells from broad species, greatly increased the expressions of porcine MHC-I (SLA-I) molecules and MHC-I antigen presentation associated genes but not porcine MHC-II (SLA-II) molecules both in vitro and in vivo. Mechanistically, the upregulation of MHC-I molecules by PDCoV infection required the master transactivator of MHC-I, NLRC5, which was mediated not only by RIG-I-initiated type I IFN signaling pathway but also by IRF1 induced by PDCoV as it could activate NLRC5 promoter activity. These results provide significant insights into the modification of the MHC class I pathway and may provide a potential therapeutic intervention for PDCoV.


Subject(s)
Coronavirus Infections , Deltacoronavirus , Histocompatibility Antigens Class I , Animals , Coronavirus Infections/immunology , Deltacoronavirus/immunology , Gene Expression Regulation/immunology , Histocompatibility Antigens Class I/genetics , Histocompatibility Antigens Class I/immunology , Interferon Regulatory Factor-1/genetics , Interferon Regulatory Factor-1/metabolism , Swine
6.
Nat Commun ; 15(1): 5330, 2024 Jun 22.
Article in English | MEDLINE | ID: mdl-38909062

ABSTRACT

Porcine deltacoronavirus (PDCoV) is an emerging enteric pathogen that has recently been detected in humans. Despite this zoonotic concern, the antigenic structure of PDCoV remains unknown. The virus relies on its spike (S) protein for cell entry, making it a prime target for neutralizing antibodies. Here, we generate and characterize a set of neutralizing antibodies targeting the S protein, shedding light on PDCoV S interdomain crosstalk and its vulnerable sites. Among the four identified antibodies, one targets the S1A domain, causing local and long-range conformational changes, resulting in partial exposure of the S1B domain. The other antibodies bind the S1B domain, disrupting binding to aminopeptidase N (APN), the entry receptor for PDCoV. Notably, the epitopes of these S1B-targeting antibodies are concealed in the prefusion S trimer conformation, highlighting the necessity for conformational changes for effective antibody binding. The binding footprint of one S1B binder entirely overlaps with APN-interacting residues and thus targets a highly conserved epitope. These findings provide structural insights into the humoral immune response against the PDCoV S protein, potentially guiding vaccine and therapeutic development for this zoonotic pathogen.


Subject(s)
Antibodies, Neutralizing , Antibodies, Viral , Deltacoronavirus , Epitopes , Spike Glycoprotein, Coronavirus , Spike Glycoprotein, Coronavirus/immunology , Spike Glycoprotein, Coronavirus/metabolism , Spike Glycoprotein, Coronavirus/chemistry , Animals , Antibodies, Neutralizing/immunology , Swine , Antibodies, Viral/immunology , Epitopes/immunology , Humans , Deltacoronavirus/immunology , Deltacoronavirus/metabolism , CD13 Antigens/metabolism , CD13 Antigens/immunology , Coronavirus Infections/immunology , Coronavirus Infections/virology , Protein Domains , Protein Binding , Swine Diseases/virology , Swine Diseases/immunology , HEK293 Cells
7.
Sheng Wu Gong Cheng Xue Bao ; 40(7): 2150-2161, 2024 Jul 25.
Article in Zh | MEDLINE | ID: mdl-39044581

ABSTRACT

This study aims to develop an effective bivalent subunit vaccine that is promising to prevent both porcine deltacoronavirus (PDCoV) and porcine epidemic diarrhea virus (PEDV). The receptor-binding domains (RBDs) of PDCoV and PEDV were fused and cloned into the eukaryotic expression vector pCDNA3.1(+). The fusion protein PDCoV-RBD-PEDV-RBD (pdRBD-peRBD) was expressed by the ExpiCHOTM expression system and purified. Mice were immunized with the fusion protein at three different doses (10, 20, and 30 µg). The humoral immune response and cellular immune response induced by the fusion protein were evaluated by ELISA and flow cytometry. The neutralization titers of the serum of immunized mice against PDCoV and PEDV were determined by the microneutralization test. The results showed that high levels of IgG antibodies were induced in the three different dose groups after booster immunization, and there was no significant difference in the antibody level between different dose groups, indicating that the immunization dose of 10 µg could achieve the fine immune effect. The results of flow cytometry showed that the immunization groups demonstrated increased proportion of CD3+CD4+ T cells and decreased proportion of CD3+CD8+ T cells, which was consistent with the expectation about the humoral immune response induced by the subunit vaccine. At the same time, the levels of interleukin (IL)-2, IL-4, and interferon (IFN)-γ in the serum were determined. The results showed that the fusion protein induced both humoral immune effect and cellular immune response. The results of the neutralization test showed that the antibody induced by 10 µg fusion protein neutralized both PDCoV and PEDV in vitro, with the titers of 1:179.25 and 1:141.21, respectively. The above results suggested that the pdRBD-peRBD could induce a high level of humoral immune response at a dose of 10 µg, and the induced antibody could neutralize both PDCoV and PEDV. Therefore, the fusion protein pdRBD-peRBD is expected to be an effective subunit vaccine that can simultaneously prevent PDCoV and PEDV.


Subject(s)
Antibodies, Viral , Coronavirus Infections , Porcine epidemic diarrhea virus , Recombinant Fusion Proteins , Viral Vaccines , Animals , Porcine epidemic diarrhea virus/immunology , Porcine epidemic diarrhea virus/genetics , Mice , Swine , Viral Vaccines/immunology , Viral Vaccines/genetics , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/genetics , Coronavirus Infections/prevention & control , Coronavirus Infections/immunology , Antibodies, Viral/blood , Antibodies, Viral/immunology , Deltacoronavirus/immunology , Deltacoronavirus/genetics , Swine Diseases/prevention & control , Swine Diseases/immunology , Vaccines, Subunit/immunology , Vaccines, Subunit/genetics , Mice, Inbred BALB C , Female , Antibodies, Neutralizing/blood , Antibodies, Neutralizing/immunology , Protein Domains , Immunogenicity, Vaccine , Immunity, Humoral
8.
Virology ; 597: 110150, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38917690

ABSTRACT

Coronaviruses (CoVs) comprise a group of important human and animal pathogens that threaten public health because of their interspecies transmission potential to humans. However, virus-like particles (VLPs) constitute versatile tools in CoVs vaccine development due to their favorable immunological characteristics. Here, we engineered the VLPs composed of the spike (S), membrane (M), and envelope (E) structural proteins of the Porcine deltacoronavirus (PDCoV) and examined their immune responses in mice. Neutralization assays and flow Cytometry demonstrated that PDCoV VLPs induced highly robust neutralizing antibodies (NAbs) and elicited cellular immunity. To assess the protective efficacy of VLPs in newborn piglets, pregnant sows received vaccinations with either a PDCoV-inactivated vaccine or VLPs at 40 and 20 days before delivery. Five days post-farrowing, piglets were orally challenged with the PDCoV strain. Severe diarrhea, high viral RNA copies, and substantial intestinal villus atrophy were detected in piglets born to unimmunized sows. However, piglets from sows immunized with VLPs exhibited high NAbs titers and markedly reduced microscopic damage to the intestinal tissues, with no piglet showing diarrhea. Hence, the results indicate that the VLPs are a potential clinical candidate for PDCoV vaccination, while the strategy may serve as a platform for developing other coronavirus vaccines.


Subject(s)
Antibodies, Neutralizing , Antibodies, Viral , Coronavirus Infections , Deltacoronavirus , Swine Diseases , Vaccines, Virus-Like Particle , Viral Vaccines , Animals , Swine , Vaccines, Virus-Like Particle/immunology , Vaccines, Virus-Like Particle/administration & dosage , Coronavirus Infections/prevention & control , Coronavirus Infections/veterinary , Coronavirus Infections/immunology , Coronavirus Infections/virology , Antibodies, Viral/blood , Antibodies, Viral/immunology , Antibodies, Neutralizing/blood , Antibodies, Neutralizing/immunology , Swine Diseases/prevention & control , Swine Diseases/virology , Swine Diseases/immunology , Viral Vaccines/immunology , Viral Vaccines/administration & dosage , Female , Deltacoronavirus/immunology , Mice , Pregnancy , Spike Glycoprotein, Coronavirus/immunology , Spike Glycoprotein, Coronavirus/genetics , Animals, Newborn
9.
Virus Res ; 302: 198497, 2021 09.
Article in English | MEDLINE | ID: mdl-34217778

ABSTRACT

Porcine deltacoronavirus (PDCoV) is an emerging coronavirus that causes vomiting, diarrhea, dehydration, and even death of piglets, resulting in significant losses to the pig industry worldwide. However, the epitopes of PDCoV remain largely unknown. In this study, a monoclonal antibody (mAb) against the PDCoV nucleocapsid (N) protein, termed 9G1, was prepared using the lymphocyte hybridoma technique, and was identified as a type IgG1 with a κ light chain and reacted with the native N protein of PDCoV. Furthermore, the epitope recognized by the 9G1 mAb was subjected to western blot and an ELISA using truncated recombinant proteins and synthetic polypeptides of the PDCoV N protein. The results indicate that 9G1 mAb recognized the epitope, G59TPIPPSYAFYY70 (EP-9G1), a novel linear B cell epitope of the PDCoV N protein. A comparison analysis revealed that the EP-9G1 epitope was highly conserved among PDCoV strains, in which four residues (G59-F68YY70) were observed among different coronavirus genera. These data demonstrate that the EP-9G1 epitope identified in this study provides some basic information for further characterization of the antigenic structure of the PDCoV N protein and has potential use for developing diagnostic reagents for PDCoV.


Subject(s)
Antibodies, Monoclonal/immunology , Coronavirus Infections/veterinary , Deltacoronavirus/immunology , Epitopes, B-Lymphocyte/immunology , Nucleocapsid Proteins/immunology , Amino Acid Sequence , Animals , Coronavirus Infections/diagnosis , Coronavirus Infections/immunology , Coronavirus Infections/virology , Deltacoronavirus/genetics , Enzyme-Linked Immunosorbent Assay/veterinary , Female , Immunoglobulin G/immunology , Mice, Inbred BALB C , Nucleocapsid Proteins/genetics , Recombinant Proteins , Sequence Alignment , Swine
10.
Viruses ; 13(12)2021 11 30.
Article in English | MEDLINE | ID: mdl-34960672

ABSTRACT

Porcine deltacoronavirus (PDCoV) can cause diarrhea and dehydration in newborn piglets. Here, we developed a double antibody sandwich quantitative enzyme-linked immunosorbent assay (DAS-ELISA) for detection of PDCoV by using a specific monoclonal antibody against the PDCoV N protein and an anti-PDCoV rabbit polyclonal antibody. Using DAS-ELISA, the detection limit of recombinant PDCoV N protein and virus titer were approximately 0.5 ng/mL and 103.0 TCID50/mL, respectively. A total of 59 intestinal and 205 fecal samples were screened for the presence of PDCoV by using DAS-ELISA and reverse transcriptase real-time PCR (RT-qPCR). The coincidence rate of the DAS-ELISA and RT-qPCR was 89.8%. DAS-ELISA had a sensitivity of 80.8% and specificity of 95.6%. More importantly, the DAS-ELISA could detect the antigen of PDCoV inactivated virus, and the viral antigen concentrations remained unchanged in the inactivated virus. These results suggest that DAS-ELISA could be used for antigen detection of clinical samples and inactivated vaccines. It is a novel method for detecting PDCoV infections and evaluating the PDCoV vaccine.


Subject(s)
Antibodies, Viral/blood , Antigens, Viral/blood , Coronavirus Infections/blood , Coronavirus Infections/veterinary , Deltacoronavirus/immunology , Enzyme-Linked Immunosorbent Assay/methods , Swine Diseases/diagnosis , Animals , Antibodies, Viral/immunology , Antigens, Viral/immunology , Coronavirus Infections/immunology , Coronavirus Infections/virology , Deltacoronavirus/genetics , Deltacoronavirus/isolation & purification , Rabbits , Swine , Swine Diseases/blood , Swine Diseases/virology
11.
Virology ; 559: 196-209, 2021 07.
Article in English | MEDLINE | ID: mdl-33964685

ABSTRACT

This study was conducted to examine the role of stress-activated protein kinases (SAPKs), including c-Jun NH2-terminal kinases (JNK1/2) and p38 mitogen-activated protein kinase (MAPK), in porcine deltacoronavirus (PDCoV) infection. Results demonstrated the activation of JNK1/2 and p38 MAPK in PDCoV-infected cells, which occurred concomitant with viral biosynthesis and irrespective of cell type. Pharmacological inhibition or knockdown of either SAPK significantly attenuated PDCoV replication, whereas addition of a signaling activator augmented virus infectivity. Moreover, pharmacological inhibition of JNK1/2 or p38 MAPK activation was innocuous to viral entry but significantly detrimental to post uncoating stages of the replication cycle. Remarkably, cytokine gene expression in PDCoV-infected IPEC-J2 cells was modified by inhibiting the activation of either SAPK. Collectively, these data indicate that JNK1/2 and p38 MAPK signaling pathways contribute to viral biosynthesis and regulate immune responses, thereby favoring the replication of PDCoV.


Subject(s)
Cytokines/immunology , Deltacoronavirus/physiology , Protein Kinases/metabolism , Stress, Physiological , Virus Replication , Animals , Cell Line , Cytokines/genetics , Deltacoronavirus/immunology , Protein Kinases/genetics , Signal Transduction , Stress, Physiological/genetics , Stress, Physiological/physiology , Swine
12.
Front Immunol ; 12: 688758, 2021.
Article in English | MEDLINE | ID: mdl-34220846

ABSTRACT

Coronaviruses (CoVs) are a known global threat, and most recently the ongoing COVID-19 pandemic has claimed more than 2 million human lives. Delays and interference with IFN responses are closely associated with the severity of disease caused by CoV infection. As the most abundant viral protein in infected cells just after the entry step, the CoV nucleocapsid (N) protein likely plays a key role in IFN interruption. We have conducted a comprehensive comparative analysis and report herein that the N proteins of representative human and animal CoVs from four different genera [swine acute diarrhea syndrome CoV (SADS-CoV), porcine epidemic diarrhea virus (PEDV), severe acute respiratory syndrome CoV (SARS-CoV), SARS-CoV-2, Middle East respiratory syndrome CoV (MERS-CoV), infectious bronchitis virus (IBV) and porcine deltacoronavirus (PDCoV)] suppress IFN responses by multiple strategies. In particular, we found that the N protein of SADS-CoV interacted with RIG-I independent of its RNA binding activity, mediating K27-, K48- and K63-linked ubiquitination of RIG-I and its subsequent proteasome-dependent degradation, thus inhibiting the host IFN response. These data provide insight into the interaction between CoVs and host, and offer new clues for the development of therapies against these important viruses.


Subject(s)
Coronavirus Nucleocapsid Proteins/genetics , Coronavirus Nucleocapsid Proteins/immunology , DEAD Box Protein 58/metabolism , Interferons/antagonists & inhibitors , Interferons/immunology , Receptors, Immunologic/metabolism , Amino Acid Sequence/genetics , Animals , COVID-19/pathology , DEAD Box Protein 58/immunology , Deltacoronavirus/genetics , Deltacoronavirus/immunology , Humans , Infectious bronchitis virus/genetics , Infectious bronchitis virus/immunology , Interferon Regulatory Factor-3/metabolism , Middle East Respiratory Syndrome Coronavirus/genetics , Middle East Respiratory Syndrome Coronavirus/immunology , Phosphorylation , Porcine epidemic diarrhea virus/genetics , Porcine epidemic diarrhea virus/immunology , Receptors, Immunologic/immunology , Severe acute respiratory syndrome-related coronavirus/genetics , Severe acute respiratory syndrome-related coronavirus/immunology , SARS-CoV-2/genetics , SARS-CoV-2/immunology , Swine , Ubiquitination/physiology
13.
Int Immunopharmacol ; 93: 107317, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33493866

ABSTRACT

Porcine deltacoronavirus (PDCoV) is an emerging swine enteropathogenic coronavirus (CoV) that poses economic and public health burdens. Currently, there are no effective antiviral agents against PDCoV. Cryptoporus volvatus often serves as an antimicrobial agent in Traditional Chinese Medicines. This study aimed to evaluate the antiviral activities of ergosterol peroxide (EP) from C. volvatus against PDCoV infection. The inhibitory activity of EP against PDCoV was assessed by using virus titration and performing Quantitative Reverse transcription PCR (RT-qPCR), Western blotting and immunofluorescence assays in LLC-PK1 cells. The mechanism of EP against PDCoV was analyzed by flow cytometry, RT-qPCR and Western blotting. We found that EP treatment inhibited PDCoV infection in LLC-PK1 cells in a dose-dependent manner. Subsequently, we demonstrated that EP blocked virus attachment and entry using RT-qPCR. Time-of-addition assays indicated that EP mainly exerted its inhibitory effect at the early and middle stages in the PDCoV replication cycle. EP also inactivated PDCoV infectivity directly as well as suppressed PDCoV-induced apoptosis. Furthermore, EP treatment decreased the phosphorylation of IκBα and p38 MAPK induced by PDCoV infection as well as the mRNA levels of cytokines (IL-1ß, IL-6, IL-12, TNF-α, IFN-α, IFN-ß, Mx1 and PKR). These results imply that EP can inhibit PDCoV infection and regulate host immune responses by downregulating the activation of the NF-κB and p38/MAPK signaling pathways in vitro. EP can be used as a potential candidate for the development of a new anti-PDCoV therapy.


Subject(s)
Antiviral Agents/pharmacology , Deltacoronavirus/drug effects , Deltacoronavirus/immunology , Ergosterol/analogs & derivatives , MAP Kinase Signaling System/drug effects , NF-kappa B/metabolism , Animals , Apoptosis/drug effects , Cell Line , Coronavirus Infections/drug therapy , Coronavirus Infections/immunology , Coronavirus Infections/virology , Cytokines/metabolism , Ergosterol/chemistry , Ergosterol/pharmacology , I-kappa B Proteins/metabolism , Immunologic Factors/chemistry , Immunologic Factors/pharmacology , LLC-PK1 Cells , Polyporaceae , Swine , Swine Diseases , Transcription Factor RelA/metabolism , Virion/drug effects , Virus Replication/drug effects
14.
J Immunol Methods ; 483: 112808, 2020 08.
Article in English | MEDLINE | ID: mdl-32562689

ABSTRACT

Three different porcine enteric coronaviruses (PECs), i.e., porcine epidemic diarrhea virus (PEDV), transmissible gastroenteritis virus (TGEV) and porcine Deltacoronavirus (PDCoV) are currently circulating in U.S. commercial swine herds. Differential diagnosis of PECs relies on laboratory methods. This study describes the development of an ELISA-like multiplex planar immunoassay based on virus-specific recombinant S1 proteins printed in an array of spots at the bottom of a 96-well microplate for simultaneous detection differential serodiagnosis of PEDV, TGEV, PDCoV in a single sample. The technology overall format and working principle is similar to the solid-phase standard ELISA. After the three typical incubation steps, the reaction was visualized as blue spots which intensity correlated with antibody levels to specific viral antigen target in the array. The diagnostic performance of the assay was evaluated on known status serum samples (n = 480) collected over time (day post-inoculation -7, 0, 7, 14, 21, 28, 35, and 42) from pigs inoculated with PEDV, TGEV Purdue, TGEV Miller, PDCoV (USA/IL/2014), or mock inoculated with culture media under experimental conditions. Antigen-specific cut-offs were selected to ensure 100% diagnostic and analytical specificity for each given antigen target. The overall diagnostic sensitivity was 92% (44/48 positives, 95% confidence interval (CI) 98,100) for PEDV S1, 100% (95/95 positives, 95% CI 98, 100) for TGEV S1, and 98% (47/48 positives, 95% CI 97, 100) for PDCoV S1. The results of this study demonstrate that the AgroDiag PEC multiplex immunoassay is an efficient and reliable test for differential detection and serodiagnosis of PEDV, TGEV and PDCoV.


Subject(s)
Alphacoronavirus/immunology , Antibodies, Viral/blood , Coronavirus Infections/veterinary , Enzyme-Linked Immunosorbent Assay/veterinary , Immunoglobulin G/blood , Serologic Tests/veterinary , Animals , Biomarkers/blood , Coronavirus Infections/diagnosis , Coronavirus Infections/immunology , Coronavirus Infections/virology , Deltacoronavirus/immunology , Diagnosis, Differential , Gastroenteritis, Transmissible, of Swine/diagnosis , Gastroenteritis, Transmissible, of Swine/immunology , Gastroenteritis, Transmissible, of Swine/virology , Porcine epidemic diarrhea virus/immunology , Predictive Value of Tests , Reproducibility of Results , Swine , Transmissible gastroenteritis virus/immunology
SELECTION OF CITATIONS
SEARCH DETAIL