Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
1.
Nature ; 583(7815): 286-289, 2020 07.
Article in English | MEDLINE | ID: mdl-32380510

ABSTRACT

The current outbreak of coronavirus disease-2019 (COVID-19) poses unprecedented challenges to global health1. The new coronavirus responsible for this outbreak-severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)-shares high sequence identity to SARS-CoV and a bat coronavirus, RaTG132. Although bats may be the reservoir host for a variety of coronaviruses3,4, it remains unknown whether SARS-CoV-2 has additional host species. Here we show that a coronavirus, which we name pangolin-CoV, isolated from a Malayan pangolin has 100%, 98.6%, 97.8% and 90.7% amino acid identity with SARS-CoV-2 in the E, M, N and S proteins, respectively. In particular, the receptor-binding domain of the S protein of pangolin-CoV is almost identical to that of SARS-CoV-2, with one difference in a noncritical amino acid. Our comparative genomic analysis suggests that SARS-CoV-2 may have originated in the recombination of a virus similar to pangolin-CoV with one similar to RaTG13. Pangolin-CoV was detected in 17 out of the 25 Malayan pangolins that we analysed. Infected pangolins showed clinical signs and histological changes, and circulating antibodies against pangolin-CoV reacted with the S protein of SARS-CoV-2. The isolation of a coronavirus from pangolins that is closely related to SARS-CoV-2 suggests that these animals have the potential to act as an intermediate host of SARS-CoV-2. This newly identified coronavirus from pangolins-the most-trafficked mammal in the illegal wildlife trade-could represent a future threat to public health if wildlife trade is not effectively controlled.


Subject(s)
Betacoronavirus/genetics , Betacoronavirus/isolation & purification , Eutheria/virology , Evolution, Molecular , Genome, Viral/genetics , Sequence Homology, Nucleic Acid , Animals , Betacoronavirus/classification , COVID-19 , China , Chiroptera/virology , Chlorocebus aethiops , Coronavirus Envelope Proteins , Coronavirus Infections/epidemiology , Coronavirus Infections/pathology , Coronavirus Infections/transmission , Coronavirus Infections/veterinary , Coronavirus Infections/virology , Coronavirus M Proteins , Coronavirus Nucleocapsid Proteins , Disease Reservoirs/virology , Genomics , Host Specificity , Humans , Lung/pathology , Lung/virology , Malaysia , Nucleocapsid Proteins/genetics , Pandemics , Phosphoproteins , Phylogeny , Pneumonia, Viral/epidemiology , Pneumonia, Viral/transmission , Pneumonia, Viral/virology , Polymerase Chain Reaction , Recombination, Genetic , SARS-CoV-2 , Sequence Alignment , Sequence Analysis, RNA , Spike Glycoprotein, Coronavirus/genetics , Vero Cells , Viral Envelope Proteins/genetics , Viral Matrix Proteins/genetics , Zoonoses/transmission , Zoonoses/virology
2.
PLoS Pathog ; 19(5): e1011384, 2023 05.
Article in English | MEDLINE | ID: mdl-37196026

ABSTRACT

Malayan pangolin SARS-CoV-2-related coronavirus (SARSr-CoV-2) is closely related to SARS-CoV-2. However, little is known about its pathogenicity in pangolins. Using CT scans we show that SARSr-CoV-2 positive Malayan pangolins are characterized by bilateral ground-glass opacities in lungs in a similar manner to COVID-19 patients. Histological examination and blood gas tests are indicative of dyspnea. SARSr-CoV-2 infected multiple organs in pangolins, with the lungs the major target, and histological expression data revealed that ACE2 and TMPRSS2 were co-expressed with viral RNA. Transcriptome analysis indicated that virus-positive pangolins were likely to have inadequate interferon responses, with relative greater cytokine and chemokine activity in the lung and spleen. Notably, both viral RNA and viral proteins were detected in three pangolin fetuses, providing initial evidence for vertical virus transmission. In sum, our study outlines the biological framework of SARSr-CoV-2 in pangolins, revealing striking similarities to COVID-19 in humans.


Subject(s)
COVID-19 , Chiroptera , Animals , Humans , Pangolins/genetics , SARS-CoV-2/genetics , Virulence , Phylogeny , RNA, Viral , Tropism
3.
J Virol ; 96(5): e0208621, 2022 03 09.
Article in English | MEDLINE | ID: mdl-34985993

ABSTRACT

Coronavirus infections induce the expression of multiple proinflammatory cytokines and chemokines. We have previously shown that in cells infected with gammacoronavirus infectious bronchitis virus (IBV), interleukin 6 (IL-6), and IL-8 were drastically upregulated, and the MAP kinase p38 and the integrated stress response pathways were implicated in this process. In this study, we report that coronavirus infection activates a negative regulatory loop that restricts the upregulation of a number of proinflammatory genes. As revealed by the initial transcriptomic and subsequent validation analyses, the anti-inflammatory adenine-uridine (AU)-rich element (ARE)-binding protein, zinc finger protein 36 (ZFP36), and its related family members were upregulated in cells infected with IBV and three other coronaviruses, alphacoronaviruses porcine epidemic diarrhea virus (PEDV), human coronavirus 229E (HCoV-229E), and betacoronavirus HCoV-OC43, respectively. Characterization of the functional roles of ZFP36 during IBV infection demonstrated that ZFP36 promoted the degradation of transcripts coding for IL-6, IL-8, dual-specificity phosphatase 1 (DUSP1), prostaglandin-endoperoxide synthase 2 (PTGS2) and TNF-α-induced protein 3 (TNFAIP3), through binding to AREs in these transcripts. Consistently, knockdown and inhibition of JNK and p38 kinase activities reduced the expression of ZFP36, as well as the expression of IL-6 and IL-8. On the contrary, overexpression of mitogen-activated protein kinase kinase 3 (MKK3) and MAPKAP kinase-2 (MK2), the upstream and downstream kinases of p38, respectively, increased the expression of ZFP36 and decreased the expression of IL-8. Taken together, this study reveals an important regulatory role of the MKK3-p38-MK2-ZFP36 axis in coronavirus infection-induced proinflammatory response. IMPORTANCE Excessive and uncontrolled induction and release of proinflammatory cytokines and chemokines, the so-called cytokine release syndrome (CRS), would cause life-threatening complications and multiple organ failure in severe coronavirus infections, including severe acute respiratory syndrome (SARS), Middle East respiratory syndrome (MERS) and COVID-19. This study reveals that coronavirus infection also induces the expression of ZFP36, an anti-inflammatory ARE-binding protein, promoting the degradation of ARE-containing transcripts coding for IL-6 and IL-8 as well as a number of other proteins related to inflammatory response. Furthermore, the p38 MAP kinase, its upstream kinase MKK3 and downstream kinase MK2 were shown to play a regulatory role in upregulation of ZFP36 during coronavirus infection cycles. This MKK3-p38-MK2-ZFP36 axis would constitute a potential therapeutic target for severe coronavirus infections.


Subject(s)
Coronavirus Infections/metabolism , Interleukin-6/metabolism , Interleukin-8/metabolism , Tristetraprolin/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Adenine/metabolism , Animals , Cell Line , Chlorocebus aethiops , Coronavirus Infections/genetics , Gene Expression Regulation , Humans , Infectious bronchitis virus/metabolism , Infectious bronchitis virus/pathogenicity , Interleukin-6/genetics , Interleukin-8/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , Transcriptional Activation , Up-Regulation , Uridine/metabolism , Vero Cells
5.
Int J Mol Sci ; 22(11)2021 May 26.
Article in English | MEDLINE | ID: mdl-34073283

ABSTRACT

Infection induces the production of proinflammatory cytokines and chemokines such as interleukin-8 (IL-8) and 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 this study, we investigated the roles of the integrated stress response (ISR) and activator protein-1 (AP-1) family proteins in regulating coronavirus-induced IL-8 and IL-6 upregulation. The mRNA expression of IL-8 and IL-6 was significantly induced in cells infected with infectious bronchitis virus (IBV), a gammacoronavirus, and porcine epidemic diarrhea virus, an alphacoronavirus. Overexpression of a constitutively active phosphomimetic mutant of eukaryotic translation initiation factor 2α (eIF2α), chemical inhibition of its dephosphorylation, or overexpression of its upstream double-stranded RNA-dependent protein kinase (PKR) significantly enhanced IL-8 mRNA expression in IBV-infected cells. Overexpression of the AP-1 protein cJUN or its upstream kinase also increased the IBV-induced IL-8 mRNA expression, which was synergistically enhanced by overexpression of cFOS. Taken together, this study demonstrated the important regulatory roles of ISR and AP-1 proteins in IL-8 production during coronavirus infection, highlighting the complex interactions between cellular stress pathways and the innate immune response.


Subject(s)
Coronavirus Infections/metabolism , Endoplasmic Reticulum Stress/genetics , Eukaryotic Initiation Factor-2/metabolism , Interleukin-8/metabolism , Unfolded Protein Response/genetics , Alphacoronavirus/metabolism , Alphacoronavirus/pathogenicity , Animals , Cell Line , Chlorocebus aethiops , Coronavirus Infections/genetics , Gammacoronavirus/metabolism , Gammacoronavirus/pathogenicity , Gene Expression Regulation , Humans , Immunity, Innate , Infectious bronchitis virus/metabolism , Infectious bronchitis virus/pathogenicity , Interleukin-8/genetics , Phosphorylation , Porcine epidemic diarrhea virus/metabolism , Porcine epidemic diarrhea virus/pathogenicity , Proto-Oncogene Proteins c-fos/genetics , Proto-Oncogene Proteins c-fos/metabolism , Proto-Oncogene Proteins c-jun/genetics , Proto-Oncogene Proteins c-jun/metabolism , Signal Transduction/genetics , Transcription Factor AP-1/genetics , Transcription Factor AP-1/metabolism , Up-Regulation , Vero Cells , eIF-2 Kinase/genetics , eIF-2 Kinase/metabolism
6.
Fish Shellfish Immunol ; 67: 459-466, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28602680

ABSTRACT

P38 mitogen-activated protein kinases (MAPKs) are one of the most important central regulatory proteins response to extra environmental stresses. In this study, two novel p38 MAPKs, Ec-P38γ and Ec-P38δ, were identified from Epinephelus coioides, an economically important cultured fish in China and Southeast Asian counties. Both of Ec-p38γ and Ec-p38δ sequences contain a serine/threonine protein kinase (S_TKc) domain and a highly conserved Thr-Gly-Tyr (TGY) motif. Analysis of phylogenetic relationships illustrated that p38 amino acid sequences were conserved between different species indicating that the functions may be similar. The four subtypes of p38 (α, ß, γ, and δ) mRNA can be detected in all thirteen tissues examined, but the expression level is different in these tissues. The expression patterns of the four Ec-p38 subtypes in E. coioides were also detected response to Cryptocaryon irritans infection, one of the most important protozoan pathogens of marine fish. The expression of four p38 subtypes was up-regulated in the tissues examined, with the highest expressions of Ec-p38α (5.2 times) and Ec-p38δ (4.2 times) occurring in the skin, while Ec-p38ß (24.8 times) and γ (16.6 times) occurred in the spleen. There was no significantly correlation between the expression of Ec-p38γ/Ec-p38δ and the expression of nuclear factor kappaB (NF-kB). The results indicated the sequences and the characters of Ec-p38γ and Ec-p38δ were conserved, the p38 subtypes showed tissue-specific expression patterns in healthy grouper, and their expressions were significantly up-regulated post C. irritans infection, suggesting these p38 MAPKs may play important roles in these tissues during pathogen-caused inflammation.


Subject(s)
Bass , Fish Diseases/immunology , Immunity, Innate/genetics , p38 Mitogen-Activated Protein Kinases/genetics , p38 Mitogen-Activated Protein Kinases/immunology , Amino Acid Sequence , Animals , Ciliophora/physiology , Ciliophora Infections/immunology , Ciliophora Infections/veterinary , Fish Proteins/chemistry , Fish Proteins/genetics , Fish Proteins/immunology , Gene Expression Profiling/veterinary , Phylogeny , Sequence Alignment/veterinary , p38 Mitogen-Activated Protein Kinases/chemistry
7.
Arch Virol ; 161(7): 2007-11, 2016 Jul.
Article in English | MEDLINE | ID: mdl-27120185

ABSTRACT

Reticuloendotheliosis virus (REV), an important immunosuppressive pathogen, has many hosts, including chickens, ducks, geese, turkeys, and wild birds. Clinically, REV may lead to increased susceptibility to other pathogens, resulting in serious tissue damage (especially tumors) and the death of its host. In this study, we encountered a disease outbreak resulting in a large number of deaths of pigeons in Guangdong Province, Southern China. Histopathological analysis revealed apparent tumor-like lesions in multiple organs of pigeons. PCR assays for detection of tumor-associated pathogens (REV, avian leukosis virus, and Marek's disease virus) in poultry revealed the presence of REV sequences only. Moreover, fowlpox virus (FPV) with an insertion of REV long terminal repeat (LTR) sequences was also considered, but it was excluded using a specific PCR assay. To gain more genetic information, two full-length REV genome sequences were determined and found to have the highest nucleotide sequence similarity (99.9 %) and the closest genetic relationship to a vaccine strain (MD-2) and had a more distant genetic relationship (94.3 %) to a duck-origin strain (ATCC-VR775). To confirm the presence of REVs in pigeons, specific-pathogen-free (SPF) chickens and healthy pigeons were inoculated with microfiltered tumor tissue homogenates and were found to be susceptible to infection with REV. To our knowledge, this is the first report of REV in pigeons, and the data suggest that pigeons may be the natural host of REV.


Subject(s)
Bird Diseases/virology , Columbidae/virology , Reticuloendotheliosis virus/isolation & purification , Animals , Bird Diseases/epidemiology , Bird Diseases/pathology , Chickens , China/epidemiology , Ducks , Genome, Viral , Phylogeny , Poultry Diseases/virology , Reticuloendotheliosis virus/classification , Reticuloendotheliosis virus/genetics , Reticuloendotheliosis virus/physiology
8.
Arch Virol ; 161(11): 3237-44, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27530112

ABSTRACT

Porcine circovirus type 2 (PCV2) is considered the major etiological pathogen of porcine circovirus-associated diseases (PCVADs) in pigs. Recently, PCV2 was also found in non-porcine animals such as cattle, rats, and mice. However, there was no record of PCV2 in rats in China. The goal of this study was to investigate whether PCV2 was present in rats (Rattus norvegicus, RN) on three swine farms, using molecular tools. PCR results showed that 30 of 95 (31.6 %) rat samples were positive for PCV2. Moreover, further genotype analysis suggested that 10 of 30 (33.3 %) were positive for PCV2a, 19 of 30 (63.3 %) were positive for PCV2b, and only one sample (1/30, 3.33 %) was co-infected by PCV2a and PCV2b. To determine the possible origin of PCV2, 60 serum samples were also collected from weaned pigs on those swine farms, and 23 out of 60 samples were positive for PCV2. In addition, two distinct RN-origin and two distinct porcine-origin PCV2 full-length nucleotide sequences were obtained from the farms. Sequence and phylogenetic analysis indicated that they had the highest nucleotide similarity and closest genetic relationships to each other. In this study, we report the infection and genome characterization of PCV2 in rats and compare RN-origin and porcine-origin PCV2 sequences obtained from the same pig farm, revealing possible cross-species transmission of PCV2.


Subject(s)
Circoviridae Infections/veterinary , Circovirus/classification , Circovirus/isolation & purification , Farms , Rats/virology , Animals , China , Circoviridae Infections/virology , Circovirus/genetics , Cluster Analysis , DNA, Viral/chemistry , DNA, Viral/genetics , Genome, Viral , Phylogeny , Polymerase Chain Reaction , Sequence Analysis, DNA , Sequence Homology , Swine/virology
9.
Exp Parasitol ; 164: 79-83, 2016 May.
Article in English | MEDLINE | ID: mdl-26921717

ABSTRACT

The excretion frequencies of cecal and intestinal droppings of Chinese Lingnan yellow chickens were observed for 10 consecutive days. The chickens were then orally inoculated with a precocious line of Eimeria necatrix, and the oocysts present in the cecal and intestinal droppings were separately collected and monitored using the McMaster method. The results showed that the excretion frequency of cecal droppings was significantly lower than that of intestinal droppings, and the oocysts of E. necatrix were distributed primarily in the cecal droppings. This distribution affects the homogeneity of the second and third generation of oocysts ingested by the chickens and therefore affects the immune effect observed during E. necatrix immunization. To artificially strengthen the immunologic homogeneity against E. necatrix, a method of artificially strengthening the second immunization was applied, and the immune effect was evaluated based on oocyst excretion, body weight gain, fecal scores, intestinal lesion scores and survival percentages. The results showed that no significant intestinal damage was caused by immunization reactions in the chickens. In addition, the number of excreted oocysts in the immunized chicken groups could be significantly increased, and the immunologic homogeneity of the immunized chickens could be improved by artificially strengthening the second immunization, which could in turn improve the immune protective effect.


Subject(s)
Chickens/parasitology , Coccidiosis/veterinary , Eimeria/isolation & purification , Immunization/veterinary , Poultry Diseases/parasitology , Animals , Cecum/parasitology , Coccidiosis/immunology , Coccidiosis/parasitology , Coccidiosis/prevention & control , Eimeria/immunology , Feces/parasitology , Immunization, Secondary/veterinary , Intestines/parasitology , Intestines/pathology , Oocysts , Poultry Diseases/immunology , Poultry Diseases/prevention & control , Random Allocation
10.
Fish Shellfish Immunol ; 47(1): 280-8, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26384847

ABSTRACT

CCR6 have been demonstrated playing an important role in immune cells homing to mucosal tissues, mediating antigen presentation and immune response in mammals. CCR6 in lower vertebrate leukocyte homing has not yet been revealed. Cryptocaryon irritans is believed to be a good pathogen model for skin and gill mucosal immunity. In this study, we identified two CCR6s and their three possible ligands CCL20 like cDNA sequences, designated as grouper EcCCR6A, EcCCR6B, EcCCL20L1, EcCCL20L2 and EcCCL20L3. It is interesting to find that EcCCR6A has a longer second extracellular loop than EcCCR6B, which is more similar to mammalian CCR6. Tissue distribution analysis showed that EcCCR6A pronouncedly dominates in gill and brain while EcCCR6B dominates in head kidney, trunk kidney and thymus. Three chemokine ligands have their own distinct expression pattern in health grouper tissues. EcCCL20L1 dominates in spleen and head kidney, EcCCL20L2 dominates in gill and thymus, whereas EcCCL20L3 dominates in skin and brain. The expression patterns of these chemokines and chemokine receptors were detected in C. irritans infected grouper and the results showed that EcCCR6A, EcCCR6B and EcCCL20L1 were significantly up-regulated in the skin of C. irritans infected fish, which indicated these two chemokine receptors and their ligand may play important role in immune cells' homing to skin mucosal immune tissues under pathogen caused inflammation.


Subject(s)
Bass , Chemokine CCL20/genetics , Ciliophora Infections/veterinary , Fish Diseases/genetics , Fish Proteins/genetics , Immunity, Mucosal , Receptors, CCR6/genetics , Amino Acid Sequence , Animals , Chemokine CCL20/chemistry , Chemokine CCL20/metabolism , Ciliophora/physiology , Ciliophora Infections/genetics , Ciliophora Infections/immunology , Ciliophora Infections/parasitology , Fish Diseases/immunology , Fish Diseases/parasitology , Fish Proteins/chemistry , Fish Proteins/metabolism , Ligands , Organ Specificity , Phylogeny , Receptors, CCR6/chemistry , Receptors, CCR6/metabolism , Sequence Alignment/veterinary
11.
Virus Genes ; 51(3): 361-6, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26573282

ABSTRACT

Since 2013, the second outbreak of peste des petits ruminants (PPR) caused by Peste des petits ruminants virus (PPRV) has spread over more than 20 provinces, municipalities, and autonomous regions in China, resulting in major economic losses for livestock industry. In 2014, we encountered a clinical PPR case on a goat farm in Guangdong province, southern China. The complete genome of this PPRV strain, named CH/GDDG/2014, was sequenced to determine its similarities and differences with other strains. The CH/GDDG/2014 genome comprised 15,954 nucleotides (six nucleotides more than classical PPRVs identified before 2013, but complying with the rule of six) with six open reading frames encoding nucleocapsid protein, phosphoprotein, matrix protein, fusion protein, hemagglutinin, and large polymerase protein, respectively. The whole-genome-based alignment analysis indicated that CH/GDDG/2014 had the most proximate consensus (99.8 %) to China/XJYL/2013 and the least consensus (87.2 %) to KN5/2011. The phylogenetic analysis showed that CH/GDDG/2014 was clustered in one branch (lineage IV) with other emerging strains during the second outbreak. This study is the first report describing the whole-genome sequence of PPRV in Guangdong province, southern China and also suggests the PPR outbreak may be closely related to illegal cross-regional importation of goats.


Subject(s)
Goat Diseases/virology , Peste-des-Petits-Ruminants/genetics , Peste-des-Petits-Ruminants/virology , Peste-des-petits-ruminants virus/genetics , Phylogeny , Animals , Base Sequence , China/epidemiology , Cluster Analysis , Disease Outbreaks , Genes, Viral , Goat Diseases/epidemiology , Goats , Nucleocapsid Proteins/genetics , Peste-des-Petits-Ruminants/mortality , Peste-des-petits-ruminants virus/isolation & purification , Sequence Alignment , Sequence Analysis, DNA
12.
Virus Genes ; 49(3): 507-11, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25248785

ABSTRACT

For the worldwide pig industries, porcine circovirus type 2 (PCV2) is an economically important pathogen. At present, the prevalence of PCV2 is common in Chinese swine herds. However, there is little information on PCV2 prevalence in non-porcine animals in China, such as bovids. Therefore, the goal of this study is to obtain the firsthand prevalence data of PCV2 in bovids in China. Two hundred and eighty serum and muscle samples from dairy cows (n = 180), buffalo (n = 50), and yellow cattle (n = 50) were analyzed by PCR. The detection results show that PCV2 infections (16 %, 8/50) only exist in buffaloes. In addition, there are different PCV2 viral DNAs identified by differential PCR in the same buffalo sample. Nucleotide sequencing and phylogenetic analysis results based on partial ORF1 and ORF2 sequences suggest that PCV2 strains have genetic diversity in buffaloes and they are divided into three different genotypes (PCV2b, PCV2d, and PCV2e, respectively). Moreover, to our knowledge, the PCV2d and PCV2e genotypes have not been previously reported in bovids. Through this study, the firsthand data of PCV2 prevalence in bovids in China was documented.


Subject(s)
Cattle Diseases/epidemiology , Cattle Diseases/virology , Circoviridae Infections/veterinary , Circovirus/classification , Circovirus/isolation & purification , Genetic Variation , Animals , Cattle , China/epidemiology , Circoviridae Infections/epidemiology , Circoviridae Infections/virology , Circovirus/genetics , Cluster Analysis , DNA, Viral/chemistry , DNA, Viral/genetics , Genotype , Molecular Sequence Data , Muscles/virology , Phylogeny , Polymerase Chain Reaction , Sequence Analysis, DNA , Sequence Homology , Serum/virology
13.
Viruses ; 16(5)2024 05 16.
Article in English | MEDLINE | ID: mdl-38793679

ABSTRACT

In recent years, an increasing number of viruses have triggered outbreaks that pose a severe threat to both human and animal life, as well as caused substantial economic losses. It is crucial to understand the genomic structure and epidemiology of these viruses to guide effective clinical prevention and treatment strategies. Nanopore sequencing, a third-generation sequencing technology, has been widely used in genomic research since 2014. This technology offers several advantages over traditional methods and next-generation sequencing (NGS), such as the ability to generate ultra-long reads, high efficiency, real-time monitoring and analysis, portability, and the ability to directly sequence RNA or DNA molecules. As a result, it exhibits excellent applicability and flexibility in virus research, including viral detection and surveillance, genome assembly, the discovery of new variants and novel viruses, and the identification of chemical modifications. In this paper, we provide a comprehensive review of the development, principles, advantages, and applications of nanopore sequencing technology in animal and human virus research, aiming to offer fresh perspectives for future studies in this field.


Subject(s)
Genome, Viral , High-Throughput Nucleotide Sequencing , Nanopore Sequencing , Viruses , Nanopore Sequencing/methods , Animals , Humans , Viruses/genetics , Viruses/classification , Viruses/isolation & purification , High-Throughput Nucleotide Sequencing/methods , Virus Diseases/virology , Virus Diseases/diagnosis , Genomics/methods , Nanopores
14.
Emerg Microbes Infect ; 12(1): e2176008, 2023 Dec.
Article in English | MEDLINE | ID: mdl-36724449

ABSTRACT

Disruption of the cell cycle is a common strategy shared by many viruses to create a conducible cellular microenvironment for their efficient replication. We have previously shown that infection of cells with gammacoronavirus infectious bronchitis virus (IBV) activated the theataxia-telangiectasia mutated (ATM) Rad3-related (ATR)/checkpoint kinase 1 (Chk1) pathway and induced cell cycle arrest in S and G2/M phases, partially through the interaction of nonstructural protein 13 (nsp13) with the p125 catalytic subunit of DNA polymerase delta (pol δ). In this study, we show, by GST pulldown, co-immunoprecipitation and immunofluorescent staining, that IBV nsp12 directly interacts with the p50 regulatory subunit of pol δ in vitro and in cells overexpressing the two proteins as well as in cells infected with a recombinant IBV harbouring an HA-tagged nsp12. Furthermore, nsp12 from severe acute respiratory syndrome coronavirus (SARS-CoV) and SARS-CoV-2 was also able to interact with p50. These interactions play a synergistic role with nsp13 in the induction of S phase arrest. The fact that subunits of an essential cellular DNA replication machinery physically associate with two core replication enzymes from three different coronaviruses highlights the importance of these associations in coronavirus replication and virus-host interaction, and reveals the potential of targeting these subunits for antiviral intervention.


Subject(s)
COVID-19 , Infectious bronchitis virus , Humans , DNA Polymerase III/chemistry , DNA Polymerase III/genetics , DNA Polymerase III/metabolism , S Phase , Coronavirus RNA-Dependent RNA Polymerase , RNA Helicases/genetics , RNA Helicases/metabolism , SARS-CoV-2/metabolism , Cell Cycle Checkpoints , Infectious bronchitis virus/genetics , Infectious bronchitis virus/metabolism , DNA Damage
15.
Front Microbiol ; 14: 1227485, 2023.
Article in English | MEDLINE | ID: mdl-37547693

ABSTRACT

Porcine reproductive and respiratory syndrome virus (PRRSV) infection of pigs causes a variety of clinical manifestations, depending on the pathogenicity and virulence of the specific strain. Identification and characterization of potential determinant(s) for the pathogenicity and virulence of these strains would be an essential step to precisely design and develop effective anti-PRRSV intervention. In this study, we report the construction of an infectious clone system based on PRRSV vaccine strain SP by homologous recombination technique, and the rescue of a chimeric rSP-HUB2 strain by replacing the GP5 and M protein-coding region from SP strain with the corresponding region from a highly pathogenic strain PRRSV-HUB2. The two recombinant viruses were shown to be genetically stable and share similar growth kinetics, with rSP-HUB2 exhibiting apparent growth and fitness advantages. Compared to in cells infected with PRRSV-rSP, infection of cells with rSP-HUB2 showed significantly more inhibition of the induction of type I interferon (IFN-ß) and interferon stimulator gene 56 (ISG56), and significantly more promotion of the induction of proinflammatory cytokines IL-6, IL-8, ISG15 and ISG20. Further overexpression, deletion and mutagenesis studies demonstrated that amino acid residue F16 in the N-terminal region of the GP5 protein from HUB2 was a determinant for the phenotypic difference between the two recombinant viruses. This study provides evidence that GP5 may function as a potential determinant for the pathogenicity and virulence of highly pathogenic PRRSV.

16.
Virology ; 582: 114-127, 2023 05.
Article in English | MEDLINE | ID: mdl-37058744

ABSTRACT

Coronavirus infection induces a variety of cellular antiviral responses either dependent on or independent of type I interferons (IFNs). Our previous studies using Affymetrix microarray and transcriptomic analysis revealed the differential induction of three IFN-stimulated genes (ISGs), IRF1, ISG15 and ISG20, by gammacoronavirus infectious bronchitis virus (IBV) infection of IFN-deficient Vero cells and IFN-competent, p53-defcient H1299 cells, respectively. In this report, the induction kinetics and anti-IBV functions of these ISGs as well as mechanisms underlying their differential induction are characterized. The results confirmed that these three ISGs were indeed differentially induced in H1299 and Vero cells infected with IBV, significantly more upregulation of IRF1, ISG15 and ISG20 was elicited in IBV-infected Vero cells than that in H1299 cells. Induction of these ISGs was also detected in cells infected with human coronavirus-OC43 (HCoV-OC43) and porcine epidemic diarrhea virus (PEDV), respectively. Manipulation of their expression by overexpression, knockdown and/or knockout demonstrated that IRF1 played an active role in suppressing IBV replication, mainly through the activation of the IFN pathway. However, a minor, if any, role in inhibiting IBV replication was played by ISG15 and ISG20. Furthermore, p53, but not IRF1, was implicated in regulating the IBV infection-induced upregulation of ISG15 and ISG20. This study provides new information on the mechanisms underlying the induction of these ISGs and their contributions to the host cell antiviral response during IBV infection.


Subject(s)
Coronavirus Infections , Gammacoronavirus , Infectious bronchitis virus , Animals , Humans , Antiviral Agents/pharmacology , Chlorocebus aethiops , Coronavirus Infections/veterinary , Cytokines/genetics , Exoribonucleases , Infectious bronchitis virus/genetics , Swine , Tumor Suppressor Protein p53 , Ubiquitins , Vero Cells
17.
Antibiotics (Basel) ; 12(6)2023 May 25.
Article in English | MEDLINE | ID: mdl-37370280

ABSTRACT

We determined the prevalence and molecular characteristics of blaCTX-M-55-positive Escherichia coli (E. coli) isolated from duck-fish polyculture farms in Guangzhou, China. A total of 914 E. coli strains were isolated from 2008 duck and environmental samples (water, soil and plants) collected from four duck fish polyculture farms between 2017 and 2019. Among them, 196 strains were CTX-M-1G-positive strains by PCR, and 177 (90%) blaCTX-M-1G-producing strains were blaCTX-M-55-positive. MIC results showed that the 177 blaCTX-M-55-positive strains were highly resistant to ciprofloxacin, ceftiofur and florfenicol, with antibiotic resistance rates above 95%. Among the 177 strains, 37 strains carrying the F18:A-:B1 plasmid and 10 strains carrying the F33:A-:B- plasmid were selected for further study. Pulse field gel electrophoresis (PFGE) combined with S1-PFGE, Southern hybridization and whole-genome sequencing (WGS) analysis showed that both horizontal transfer and clonal spread contributed to dissemination of the blaCTX-M-55 gene among the E. coli. blaCTX-M-55 was located on different F18:A-:B1 plasmids with sizes between ~76 and ~173 kb. In addition, the presence of blaCTX-M-55 with other resistance genes (e.g., tetA, floR, fosA3, blaTEM, aadA5 CmlA and InuF) on the same F18:A-:B1 plasmid may result in co-selection of resistance determinants and accelerate the dissemination of blaCTX-M-55 in E. coli. In summary, the F18:A-:B1 plasmid may play an important role in the transmission of blaCTX-M-55 in E. coli, and the continuous monitoring of the prevalence and transmission mechanism of blaCTX-M-55 in duck-fish polyculture farms remains important.

18.
Nat Commun ; 14(1): 2488, 2023 04 29.
Article in English | MEDLINE | ID: mdl-37120646

ABSTRACT

Wildlife is reservoir of emerging viruses. Here we identified 27 families of mammalian viruses from 1981 wild animals and 194 zoo animals collected from south China between 2015 and 2022, isolated and characterized the pathogenicity of eight viruses. Bats harbor high diversity of coronaviruses, picornaviruses and astroviruses, and a potentially novel genus of Bornaviridae. In addition to the reported SARSr-CoV-2 and HKU4-CoV-like viruses, picornavirus and respiroviruses also likely circulate between bats and pangolins. Pikas harbor a new clade of Embecovirus and a new genus of arenaviruses. Further, the potential cross-species transmission of RNA viruses (paramyxovirus and astrovirus) and DNA viruses (pseudorabies virus, porcine circovirus 2, porcine circovirus 3 and parvovirus) between wildlife and domestic animals was identified, complicating wildlife protection and the prevention and control of these diseases in domestic animals. This study provides a nuanced view of the frequency of host-jumping events, as well as assessments of zoonotic risk.


Subject(s)
COVID-19 , Chiroptera , Viruses , Animals , Animals, Domestic/virology , Animals, Wild/virology , Animals, Zoo/virology , Chiroptera/virology , Mammals/virology , Pangolins/virology , Phylogeny , Zoonoses/virology
19.
Virus Genes ; 45(1): 181-5, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22528639

ABSTRACT

A total of 127 porcine samples were collected from 48 farms in six provinces in south China. The positive rate of porcine epidemic diarrhea virus (PEDV) was 43.0 % (55/127), and the co-infection rate of PEDV and transmissible gastroenteritis virus (TGEV) was 12.0 % (15/127). The partial S gene and complete M gene were amplified from PEDV-positive strains by RT-PCR, cloned, sequenced and compared with each other, as well as with the reference strains in GenBank. Sequence homology results of the partial S gene and complete M gene showed that all south China field PEDV strains had nucleotide (deduced amino acid) sequence identities of 86.7-98.7 % (83.2-99.3 %) and 96.1-100 % (95.0-100%), respectively, with the foreign reference strains reported in GenBank. Phylogenetic analysis of the partial S gene showed that all the south China PEDV strains and two Thailand strains (08UB01 and 08RB07) belong to the same group and differ genetically from European strains and early domestic strains. Phylogenetic analysis of the complete M gene showed that all south China PEDV strains have a close relationship with most of the strains in Korea and Thailand, but differ genetically from the vaccine strain (CV777).


Subject(s)
Coronavirus Infections/veterinary , Phylogeny , Porcine epidemic diarrhea virus/genetics , Swine Diseases/epidemiology , Swine/virology , Animals , China/epidemiology , Coronavirus Infections/epidemiology , Coronavirus Infections/virology , Coronavirus M Proteins , Membrane Glycoproteins/genetics , Porcine epidemic diarrhea virus/classification , Porcine epidemic diarrhea virus/isolation & purification , Reverse Transcriptase Polymerase Chain Reaction , Sequence Analysis, DNA , Spike Glycoprotein, Coronavirus , Swine Diseases/virology , Viral Envelope Proteins/genetics , Viral Matrix Proteins/genetics
20.
Emerg Microbes Infect ; 11(1): 1717-1729, 2022 Dec.
Article in English | MEDLINE | ID: mdl-35727266

ABSTRACT

Coronaviruses have evolved a variety of strategies to exploit normal cellular processes and signalling pathways for their efficient reproduction in a generally hostile cellular environment. One immediate-early response gene (IEG) family, the AP-1 gene family, was previously shown to be activated by coronavirus infection. In this study, we report that another IEG family, the EGR family, is also activated in cells infected with four different coronaviruses in three genera, i.e. gammacoronavirus infectious bronchitis virus (IBV), alphacoronaviruses porcine epidemic diarrhoea virus (PEDV) and human coronavirus-229E (HCoV-229E), and betacoronavirus HCoV-OC43. Knockdown of EGR1 reduced the expression of cJUN and cFOS, and knockdown of cJUN and/or cFOS reduced the expression of EGR1, demonstrating that these two IEG families may be cross-activated and mutual regulated. Furthermore, ERK1/2 was identified as an upstream kinase, and JNK and p38 as inhibitors of EGR1 activation in coronavirus-infected cells. However, upregulation of EGR family genes, in particular EGR1, appears to play a differential role in regulating viral replication, apoptosis and antiviral response. EGR1 was shown to play a limited role in regulation of coronavirus replication, and an anti-apoptotic role in cells infected with IBV or PEDV, but not in cells infected with HCoV-229E. Upregulation of EGR1 may also play a differential role in the regulation of antiviral response against different coronaviruses. This study reveals a novel regulatory network shared by different coronaviruses in the immediate-early response of host cells to infection.


Subject(s)
Coronavirus Infections , Coronavirus OC43, Human , Coronavirus , Animals , Antiviral Agents/pharmacology , Apoptosis , Coronavirus/genetics , Swine , Transcription Factor AP-1/genetics , Transcription Factor AP-1/pharmacology , Transcription Factor AP-1/therapeutic use , Virus Replication
SELECTION OF CITATIONS
SEARCH DETAIL