Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
1.
PLoS Pathog ; 18(2): e1010343, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-35176124

RESUMO

The continuous emergence of severe acute respiratory coronavirus 2 (SARS-CoV-2) variants and the increasing number of breakthrough infection cases among vaccinated people support the urgent need for research and development of antiviral drugs. Viral entry is an intriguing target for antiviral drug development. We found that diltiazem, a blocker of the L-type calcium channel Cav1.2 pore-forming subunit (Cav1.2 α1c) and an FDA-approved drug, inhibits the binding and internalization of SARS-CoV-2, and decreases SARS-CoV-2 infection in cells and mouse lung. Cav1.2 α1c interacts with SARS-CoV-2 spike protein and ACE2, and affects the attachment and internalization of SARS-CoV-2. Our finding suggests that diltiazem has potential as a drug against SARS-CoV-2 infection and that Cav1.2 α1c is a promising target for antiviral drug development for COVID-19.


Assuntos
Tratamento Farmacológico da COVID-19 , COVID-19 , Diltiazem/farmacologia , Pulmão/efeitos dos fármacos , SARS-CoV-2/efeitos dos fármacos , Células A549 , Animais , COVID-19/patologia , COVID-19/virologia , Células Cultivadas , Chlorocebus aethiops , Diltiazem/uso terapêutico , Modelos Animais de Doenças , Feminino , Células HEK293 , Células HeLa , Humanos , Pulmão/patologia , Pulmão/virologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , SARS-CoV-2/fisiologia , Células Vero , Ligação Viral/efeitos dos fármacos , Internalização do Vírus/efeitos dos fármacos
2.
PLoS Pathog ; 14(7): e1007189, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-30028877

RESUMO

Rabies virus (RABV) invades the central nervous system and nearly always causes fatal disease in humans. How RABV interacts with host neuron membrane receptors to become internalized and cause rabid symptoms is not yet fully understood. Here, we identified a novel receptor of RABV, which RABV uses to infect neurons. We found that metabotropic glutamate receptor subtype 2 (mGluR2), a member of the G protein-coupled receptor family that is abundant in the central nervous system, directly interacts with RABV glycoprotein to mediate virus entry. RABV infection was drastically decreased after mGluR2 siRNA knock-down in cells. Antibodies to mGluR2 blocked RABV infection in cells in vitro. Moreover, mGluR2 ectodomain soluble protein neutralized the infectivity of RABV cell-adapted strains and a street strain in cells (in vitro) and in mice (in vivo). We further found that RABV and mGluR2 are internalized into cells and transported to early and late endosomes together. These results suggest that mGluR2 is a functional cellular entry receptor for RABV. Our findings may open a door to explore and understand the neuropathogenesis of rabies.


Assuntos
Vírus da Raiva/patogenicidade , Raiva/metabolismo , Receptores de Glutamato Metabotrópico/metabolismo , Internalização do Vírus , Animais , Linhagem Celular , Humanos , Camundongos , Raiva/fisiopatologia
3.
Sensors (Basel) ; 20(8)2020 Apr 23.
Artigo em Inglês | MEDLINE | ID: mdl-32340181

RESUMO

The best water vapor information layer (BWIL), based on Himawari-8 water vapor bands over a typical region of East Asia, is investigated with the U.S. standard atmospheric profile and European Centre for Medium-Range Weather Forecasts Re-Analysis-interim (ERA-interim) dataset. The sensitivity tests reveal that the height of the BWIL is connected heavily to the amount of water vapor in the atmosphere, and to the satellite zenith angle. According to the temporal and spatial distribution analysis of BWIL, there are two basic features of BWIL. First, it lifts from January to July gradually and descends from July to October in the whole region. Second, it is higher over sea than land. These characteristics may stem from the transport of water vapor by monsoon and the concentration of water vapor in different areas. With multiple water vapor absorption IR bands, Himawari-8 can present water vapor information at multiple pressure layers. The water vapor content of ERA-interim in July 2016 is assessed as an example. By comparing the brightness temperatures from satellite observation and simulation under clear sky conditions, the ERA-interim reanalysis dataset may underestimate the amount of water vapor at pressure layers higher than 280 hPa and overestimate the water vapor quantity at pressure layers from 394 to 328 hPa, yet perform well at 320~260 hPa during this month.

4.
Arch Virol ; 162(2): 359-367, 2017 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-27757685

RESUMO

Bovine ephemeral fever (BEF) is caused by the arthropod-borne bovine ephemeral fever virus (BEFV), which is a member of the family Rhabdoviridae and the genus Ephemerovirus. BEFV causes an acute febrile infection in cattle and water buffalo. In this study, a recombinant Newcastle disease virus (NDV) expressing the glycoprotein (G) of BEFV (rL-BEFV-G) was constructed, and its biological characteristics in vitro and in vivo, pathogenicity, and immune response in mice and cattle were evaluated. BEFV G enabled NDV to spread from cell to cell. rL-BEFV-G remained nonvirulent in poultry and mice compared with vector LaSota virus. rL-BEFV-G triggered a high titer of neutralizing antibodies against BEFV in mice and cattle. These results suggest that rL-BEFV-G might be a suitable candidate vaccine against BEF.


Assuntos
Anticorpos Neutralizantes/biossíntese , Anticorpos Antivirais/biossíntese , Vírus da Febre Efêmera Bovina/genética , Febre Efêmera/prevenção & controle , Vírus da Doença de Newcastle/genética , Vacinas Virais/genética , Animais , Bovinos , Embrião de Galinha , Cricetinae , Cães , Febre Efêmera/imunologia , Febre Efêmera/virologia , Vírus da Febre Efêmera Bovina/efeitos dos fármacos , Vírus da Febre Efêmera Bovina/imunologia , Células Epiteliais/virologia , Expressão Gênica , Vetores Genéticos/química , Vetores Genéticos/imunologia , Glicoproteínas/administração & dosagem , Glicoproteínas/genética , Glicoproteínas/imunologia , Imunização , Células Madin Darby de Rim Canino , Camundongos , Camundongos Endogâmicos BALB C , Vírus da Doença de Newcastle/imunologia , Vírus Reordenados/genética , Vírus Reordenados/imunologia , Vacinas Sintéticas , Proteínas Virais/administração & dosagem , Proteínas Virais/genética , Proteínas Virais/imunologia , Vacinas Virais/administração & dosagem , Vacinas Virais/imunologia
5.
Virol J ; 13: 109, 2016 06 24.
Artigo em Inglês | MEDLINE | ID: mdl-27342050

RESUMO

BACKGROUND: West Nile virus (WNV) is an emerging zoonotic pathogen which is harmful to human and animal health. Effective vaccination in susceptible hosts should protect against WNV infection and significantly reduce viral transmission between animals and from animals to humans. A versatile vaccine suitable for different species that can be delivered via flexible routes remains an essential unmet medical need. In this study, we developed a recombinant avirulent Newcastle disease virus (NDV) LaSota strain expressing WNV premembrane/envelope (PrM/E) proteins (designated rLa-WNV-PrM/E) and evaluated its immunogenicity in mice, horses, chickens, ducks and geese. RESULTS: Mouse immunization experiments disclosed that rLa-WNV-PrM/E induces significant levels of WNV-neutralizing antibodies and E protein-specific CD4+ and CD8+ T-cell responses. Moreover, recombinant rLa-WNV-PrM/E elicited significant levels of WNV-specific IgG in horses upon delivery via intramuscular immunization, and in chickens, ducks and geese via intramuscular, oral or intranasal immunization. CONCLUSIONS: Our results collectively support the utility of rLa-WNV-PrM/E as a promising WNV veterinary vaccine candidate for mammals and poultry.


Assuntos
Mamíferos/imunologia , Vírus da Doença de Newcastle/genética , Doenças das Aves Domésticas/imunologia , Vacinas Virais/imunologia , Febre do Nilo Ocidental/prevenção & controle , Vírus do Nilo Ocidental/imunologia , Animais , Anticorpos Antivirais/imunologia , Galinhas , Feminino , Vetores Genéticos/genética , Vetores Genéticos/imunologia , Humanos , Mamíferos/virologia , Camundongos , Vírus da Doença de Newcastle/metabolismo , Doenças das Aves Domésticas/prevenção & controle , Doenças das Aves Domésticas/virologia , Vacinação , Proteínas do Envelope Viral/administração & dosagem , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/imunologia , Vacinas Virais/administração & dosagem , Vacinas Virais/genética , Febre do Nilo Ocidental/imunologia , Febre do Nilo Ocidental/virologia , Vírus do Nilo Ocidental/genética
6.
Exp Parasitol ; 153: 39-44, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25746893

RESUMO

For the first time, we show here that perforin-like (TgPLP1) is expressed in bradyzoites of Toxoplasma gondii. An immunofluorescence assay (IFA) and immunohistochemistry (IHC) revealed that TgPLP1 is expressed in T. gondii-encysted and in vitro-induced bradyzoites, TgPLP1 is distributed in micronemes in a manner similar to its distribution in tachyzoites. To shed light on the function of TgPLP1 in bradyzoites, quantitative PCR revealed that the expression level of TgPLP1 gene decreased over time during differentiation into bradyzoites in vitro. This finding suggests that TgPLP1 may play a role in the rupture of tissue cysts or the maintenance of cyst structure, although the exact function of this gene in the bradyzoites is still unknown.


Assuntos
Perforina/metabolismo , Proteínas de Protozoários/metabolismo , Toxoplasma/metabolismo , Toxoplasmose/parasitologia , Animais , Linhagem Celular , Feminino , Humanos , Estágios do Ciclo de Vida , Camundongos Endogâmicos BALB C , Microssomos/parasitologia , Perforina/genética , Transporte Proteico , Proteínas de Protozoários/genética , Coelhos , Toxoplasma/genética , Toxoplasma/crescimento & desenvolvimento
7.
Parasitol Res ; 113(12): 4335-48, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25248513

RESUMO

Avian coccidiosis, caused by Eimeria spp., is one of the major parasitic diseases in birds. Cysteine protease is a major virulence factor in parasitic protozoa, and it may be a suitable chemotherapeutic target and vaccine candidate molecule. A 100 amino acid (aa.) partial sequence of cathepsin L, which is a cysteine protease, was reported by Katrib et al. (Ac. No. CDJ41293) (2012). A 219 aa. sequence was reported by Reid et al. (Ac. No. AFV92863) (2013). However, the open reading frame (ORF) was not reported. In this study, a full sequence of a cathepsin-L-like peptidase in Eimeria tenella (EtcatL) was obtained and its biochemical characterizations and expression profiles were analyzed across different stages of the parasite's life cycle. Results showed that the EtcatL gene encodes a protein 470 aa. in length, with 47 and 49% identity to Toxoplasma gondii and Eimeria acervulina. Considering the close phylogenetic relationship, TgcatL (PDB. ID 3F75) was selected for use as a template for homology modeling with quality factors of 90.9. Gelatin SDS-PAGE showed it to exert protease activity at ≈38 and ≈26 kDa. Further analysis showed the kinetic parameters of the recombinant peptidase to be K m = 8.9 µM and V max = 5.7 RFU/s µM at pH 5.5 containing 10 mM dithiothreitol (DTT) in the reaction matrix, and the IC50 value of E64 was 65.32 ± 3.02 nM. The recombinant protein was active from 25 to 50 °C, with optimal activity at 42 °C. The RT-PCR and Western blot results showed it to be expressed mainly at the endogenous stages and the initial phase of the sporulation. The protective experiment showed that chickens immunized with 100 and 200 µg rEtcatL had reduction of weight loss values 48.7 and 57.9% those of infected controls, respectively. Their reduction of lesion scores (RLS) were 25.0 and 47.2% that of control chickens, and relative oocyst production (ROP) was 39.6 and 15.5% that of control chickens. These results indicate that the EtcatL can be used as an effective immunogen, and further studies are needed to enhance its potential as a vaccine candidate molecule.


Assuntos
Anticorpos Antiprotozoários/imunologia , Catepsina L/genética , Galinhas/imunologia , Coccidiose/veterinária , Eimeria tenella/enzimologia , Motivos de Aminoácidos , Sequência de Aminoácidos , Animais , Sequência de Bases , Catepsina L/imunologia , Catepsina L/metabolismo , Galinhas/parasitologia , Coccidiose/imunologia , Coccidiose/parasitologia , Coccidiose/prevenção & controle , Eimeria tenella/genética , Feminino , Camundongos , Camundongos Endogâmicos BALB C , Modelos Moleculares , Dados de Sequência Molecular , Oocistos , Filogenia , Proteínas Recombinantes
8.
Emerg Microbes Infect ; 13(1): 2300464, 2024 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-38164797

RESUMO

Genetic changes have occurred in the genomes of prevalent African swine fever viruses (ASFVs) in the field in China, which may change their antigenic properties and result in immune escape. There is usually poor cross-protection between heterogonous isolates, and, therefore, it is important to test the cross-protection of the live attenuated ASFV vaccines against current prevalent heterogonous isolates. In this study, we evaluated the protective efficacy of the ASFV vaccine candidate HLJ/18-7GD against emerging isolates. HLJ/18-7GD provided protection against a highly virulent variant and a lower lethal isolate, both derived from genotype II Georgia07-like ASFV and isolated in 2020. HLJ/18-7GD vaccination prevented pigs from developing ASF-specific clinical signs and death, decreased viral shedding via the oral and rectal routes, and suppressed viral replication after challenges. However, HLJ/18-7GD vaccination did not provide solid cross-protection against genotype I NH/P68-like ASFV challenge in pigs. HLJ/18-7GD vaccination thus shows great promise as an alternative strategy for preventing and controlling genotype II ASFVs, but vaccines providing cross-protection against different ASFV genotypes may be needed in China.


Assuntos
Vírus da Febre Suína Africana , Febre Suína Africana , Vacinas Virais , Suínos , Animais , Febre Suína Africana/prevenção & controle , Vacinas Atenuadas/genética , Proteínas Virais/genética , Genótipo , Vacinas Virais/genética
9.
Int J Biol Macromol ; 268(Pt 1): 131695, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38642684

RESUMO

Due to the absence of effective vaccine and treatment, African swine fever virus (ASFV) control is entirely dependent on accurate and early diagnosis, along with culling of infected pigs. The B646L/p72 is the major capsid protein of ASFV and is an important target for developing a diagnostic assays and vaccines. Herein, we generated a monoclonal antibody (mAb) (designated as 2F11) against the trimeric p72 protein, and a blocking ELISA (bELISA) was established for the detection of both genotype I and II ASFV antibodies. To evaluate the performance of the diagnostic test, a total of 506 porcine serum samples were tested. The average value of percent of inhibition (PI) of 133 negative pig serum was 8.4 % with standard deviation (SD) 6.5 %. Accordingly, the cut-off value of the newly established method was set at 28 % (mean + 3SD). Similarly, a receiver operating characteristic (ROC) was applied to determine the cut off value and the p72-bELISA exhibited a sensitivity of 100 % and a specificity of 99.33 % when the detection threshold was set at 28 %. The bELISA was also able to specifically recognize anti-ASFV sera without cross-reacting with other positive serums for other major swine pathogens. Moreover, by designing a series of overlapped p72 truncated proteins, the linear B cell epitope recognized by 2F11 mAb was defined to be 283NSHNIQ288. Amino acid sequence comparison revealed that the amino acid sequence 283NSHNIQ288 is highly conserved between different ASFV isolates. Our findings indicate that the newly established mAb based blocking ELISA may have a great potential in improving the detection of ASFV antibodies and provides solid foundation for further studies.


Assuntos
Vírus da Febre Suína Africana , Anticorpos Monoclonais , Anticorpos Antivirais , Ensaio de Imunoadsorção Enzimática , Epitopos de Linfócito B , Animais , Vírus da Febre Suína Africana/imunologia , Ensaio de Imunoadsorção Enzimática/métodos , Anticorpos Monoclonais/imunologia , Anticorpos Antivirais/imunologia , Anticorpos Antivirais/sangue , Suínos , Epitopos de Linfócito B/imunologia , Proteínas do Capsídeo/imunologia , Febre Suína Africana/imunologia , Febre Suína Africana/diagnóstico , Febre Suína Africana/virologia , Sequência de Aminoácidos , Mapeamento de Epitopos
10.
Virus Res ; 346: 199412, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38838820

RESUMO

African swine fever virus (ASFV) is a large double-stranded DNA virus with a complex structural architecture and encodes more than 150 proteins, where many are with unknown functions. E184L has been reported as one of the immunogenic ASFV proteins that may contribute to ASFV pathogenesis and immune evasion. However, the antigenic epitopes of E184L are not yet characterized. In this study, recombinant E184L protein was expressed in prokaryotic expression system and four monoclonal antibodies (mAbs), designated as 1A10, 2D2, 3H6, and 4C10 were generated. All four mAbs reacted specifically with ASFV infected cells. To identify the epitopes of the mAbs, a series of overlapped peptides of E184L were designed and expressed as maltose binding fusion proteins. Accordingly, the expressed fusion proteins were probed with each E184L mAb separately by using Western blot. Following a fine mapping, the minimal linear epitope recognized by mAb 1A10 was identified as 119IQRQGFL125, and mAbs 2D2, 3H6, and 4C10 recognized a region located between 153DPTEFF158. Alignment of amino acids of E184L revealed that the two linear epitopes are highly conserved among different ASFV isolates. Furthermore, the potential application of the two epitopes in ASFV diagnosis was assessed through epitope-based ELISA using 24 ASFV positive and 18 negative pig serum and the method were able to distinguish positive and negative samples, indicating the two epitopes are dominant antigenic sites. To our knowledge, this is the first study to characterize the B cell epitopes of the antigenic E184L protein of ASFV, offering valuable tools for future research, as well as laying a foundation for serological diagnosis and epitope-based marker vaccine development.


Assuntos
Vírus da Febre Suína Africana , Anticorpos Monoclonais , Anticorpos Antivirais , Mapeamento de Epitopos , Epitopos de Linfócito B , Vírus da Febre Suína Africana/imunologia , Vírus da Febre Suína Africana/genética , Anticorpos Monoclonais/imunologia , Epitopos de Linfócito B/imunologia , Animais , Anticorpos Antivirais/imunologia , Suínos , Febre Suína Africana/imunologia , Febre Suína Africana/virologia , Camundongos , Proteínas Virais/imunologia , Proteínas Virais/genética , Proteínas Virais/química , Antígenos Virais/imunologia , Antígenos Virais/genética , Antígenos Virais/química , Camundongos Endogâmicos BALB C
11.
Exp Parasitol ; 133(2): 121-30, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23201216

RESUMO

Although IL-17 is a key factor in Th17 lineage host responses and plays critical roles in immunological control of a variety of infectious diseases, the contribution of IL-17 to immune function during Eimeria tenella infection is unknown. In the present study, we used an experimental E. tenella infection model to clarify the role of Th17-associated response in the resulting immune response by quantitative real-time PCR assays. We observed robust production of STAT-3 (the transcription factors), IL-1ß, IL-6 and IL-17 in cecal intraepithelial lymphocytes during the early infection, peaking at 6h p.i. and declining thereafter. The expression of TGF-ß was moderately upregulated and had 2 peaks at 6 and 72h p.i. during the early infection. To further investigate the role of chIL-17 during the infection, we treated the infected chickens with IL-17 and its neutralized antibody. As a result, the reduced fecal oocyst shedding and cecal lesion scores, but enhanced body weight gains were observed in IL-17 neutralized chickens. The results of histopathology showed that the neutrophils recruitment diminished and the parasite burden in IL-17 neutralized chickens decreased. These results may be due to the significant decrease in the production of IL-17, IL-6 and TGF-ß, but enhanced IL-12 and IFN-γ expression in IL-17 neutralized chickens. The converse results were shown in IL-17 treated infected-chickens in which chickens showed increased fecal oocyst shedding, exacerbated lesion scores, and reduced body weight gains. These results suggested that chicken IL-17 might mediate E. tenella - induced immunopathology during the infection.


Assuntos
Galinhas/parasitologia , Coccidiose/veterinária , Eimeria tenella/imunologia , Interleucina-17/imunologia , Doenças das Aves Domésticas/imunologia , Células Th17/imunologia , Animais , Anticorpos Antiprotozoários/imunologia , Ceco/patologia , Clonagem Molecular , Coccidiose/imunologia , Coccidiose/patologia , Eimeria tenella/patogenicidade , Fezes/parasitologia , Regulação da Expressão Gênica , Interferon gama/metabolismo , Interleucina-12/metabolismo , Interleucina-17/genética , Interleucina-17/metabolismo , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Interleucina-6/genética , Interleucina-6/metabolismo , Neutrófilos/imunologia , Reação em Cadeia da Polimerase/veterinária , Doenças das Aves Domésticas/parasitologia , Doenças das Aves Domésticas/patologia , Coelhos , Proteínas Recombinantes/genética , Proteínas Recombinantes/imunologia , Proteínas Recombinantes/metabolismo , Fator de Transcrição STAT3/genética , Fator de Transcrição STAT3/metabolismo , Organismos Livres de Patógenos Específicos , Fator de Crescimento Transformador beta/genética , Fator de Crescimento Transformador beta/metabolismo , Aumento de Peso
12.
Viruses ; 15(7)2023 06 28.
Artigo em Inglês | MEDLINE | ID: mdl-37515155

RESUMO

A cell line expressing the CD2v protein of ASFV was generated. The efficient expression of CD2v protein was determined by immunofluorescence and Western blotting. The CD2v protein was Ni-affinity purified from the supernatant of cell cultures. The CD2v-expressing cells showed properties of hemadsorption, and the secreted CD2v protein exhibited hemagglutinating activity. The antigenicity and immunoprotection ability of CD2v were evaluated by immunizing pigs alone, combined with a cell-line-expressed p30 protein or triple combined with p30 and K205R protein. Immunized pigs were challenged with the highly virulent ASFV strain HLJ/18. Virus challenge results showed that CD2v immunization alone could provide partial protection at the early infection stage. Protein p30 did not show synergistic protection effects in immunization combined with CD2v. Interestingly, immunization with the triple combination of CD2V, p30 and K205R reversed the protection effect. The viremia onset time was delayed, and one pig out of three recovered after the challenge. The pig recovered from ASFV clinical symptoms, the rectal temperature returned to normal levels and the viremia was cleared. The mechanism of this protection effect warrants further investigation.


Assuntos
Vírus da Febre Suína Africana , Febre Suína Africana , Vacinas Virais , Suínos , Animais , Vírus da Febre Suína Africana/genética , Proteínas Virais , Viremia/prevenção & controle , Linhagem Celular , Mamíferos
13.
Virus Res ; 328: 199085, 2023 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-36889544

RESUMO

African swine fever virus causes an acute, highly contagious swine disease with high mortality, leading to enormous losses in the pig industry. The K205R, a nonstructural protein of African swine fever virus, is abundantly expressed in the cytoplasm of infected cells at the early stage of infection and induces a strong immune response. However, to date, the antigenic epitopes of this immunodeterminant have not been characterized. In the present study, the K205R protein was expressed in a mammalian cell line and purified using Ni-affinity chromatography. Furthermore, three monoclonal antibodies (mAbs; 5D6, 7A8, and 7H10) against K205R were generated. Indirect immunofluorescence assay and western blot results showed that all three mAbs recognized native and denatured K205R in African swine fever virus (ASFV)-infected cells. To identify the epitopes of the mAbs, a series of overlapping short peptides were designed and expressed as fusion proteins with maltose-binding protein. Subsequently, the peptide fusion proteins were probed with monoclonal antibodies using western blot and enzyme-linked immunosorbent assay. The three target epitopes were fine-mapped; the core sequences of recognized by the mAbs 5D6, 7A8, and 7H10 were identified as 157FLTPEIQAILDE168, 154REKFLTP160, and 136PTNAMFFTRSEWA148, respectively. Probing with sera from ASFV-infected pigs in a dot blot assay demonstrated that epitope 7H10 was the immunodominant epitope of K205R. Sequence alignment showed that all epitopes were conserved across ASFV strains and genotypes. To our knowledge, this is the first study to characterize the epitopes of the antigenic K205R protein of ASFV. These findings may serve as a basis for the development of serological diagnostic methods and subunit vaccines.


Assuntos
Vírus da Febre Suína Africana , Febre Suína Africana , Suínos , Animais , Epitopos de Linfócito B/genética , Anticorpos Monoclonais , Linhagem Celular , Anticorpos Antivirais , Mamíferos
14.
Exp Parasitol ; 130(4): 442-8, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22326592

RESUMO

Coccidiosis is an economically important protozoan disease worldwide caused by Eimeria parasites. Toll-like receptors (TLRs), a family of highly conserved proteins, are involved in pathogen detection by initiating host responses, and play important roles in the reduction and clearance of pathogens. Little is known about the roles of chicken TLRs during Eimeria tenella infection. We detected the dynamic changes in the expression of TLRs and associated cytokines in the cecum of E. tenella-infected chickens during the early stage of infection. Day-old (Experiment 1) and three-week-old (Experiment 2) chickens were orally gavaged with 10,000 oocysts (30 chickens each experiment), and their cecum intraepithelial lymphocytes were collected at 3, 6, 12, 24, 48, and 72h post-infection (hpi). Expression profiling of TLR1LA, TLR4, TLR5, TLR7, TLR21, and IFN-α, IFN-ß, IFN-γ, IL-1ß, IL-12 genes were analyzed using quantitative real-time polymerase chain reaction. Almost all TLR transcripts were transiently increased at 3hpi in Experiment 1. In three-week-old chickens, TLR1LA, TLR4, TLR5, TLR7, and TLR21 expression was upregulated at 12hpi, and TLR1LA, TLR5, and TLR21 were highly expressed at 72hpi. In day-old chickens, IFN-α, IFN-ß, IFN-γ, IL-1ß, and IL-12 expression was significantly upregulated at 3hpi (156.1-1117.1-fold change), in comparison to the different peak level times and relatively small changes for these cytokines in the three-week-old chickens. Our results provide a valuable overview for the expression pattern of TLRs and associated cytokines during the early stage of E. tenella infection in chickens.


Assuntos
Galinhas/parasitologia , Coccidiose/veterinária , Citocinas/metabolismo , Eimeria tenella/metabolismo , Doenças das Aves Domésticas/imunologia , Receptores Toll-Like/metabolismo , Animais , Ceco/imunologia , Ceco/parasitologia , Coccidiose/imunologia , Coccidiose/parasitologia , Citocinas/genética , Citocinas/imunologia , DNA Complementar/metabolismo , Eimeria tenella/genética , Eimeria tenella/imunologia , Perfilação da Expressão Gênica , Imunidade Inata , Linfócitos/imunologia , Doenças das Aves Domésticas/parasitologia , RNA de Protozoário/análise , Reação em Cadeia da Polimerase em Tempo Real/veterinária , Organismos Livres de Patógenos Específicos , Receptores Toll-Like/genética , Receptores Toll-Like/imunologia
15.
Virol Sin ; 37(2): 248-255, 2022 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-35234625

RESUMO

Severe acute respiratory syndrome (SARS) is a highly contagious zoonotic disease caused by SARS coronavirus (SARS-CoV). Since its outbreak in Guangdong Province of China in 2002, SARS has caused 8096 infections and 774 deaths by December 31st, 2003. Although there have been no more SARS cases reported in human populations since 2004, the recent emergence of a novel coronavirus disease (COVID-19) indicates the potential of the recurrence of SARS and other coronavirus disease among humans. Thus, developing a rapid response SARS vaccine to provide protection for human populations is still needed. Spike (S) protein of SARS-CoV can induce neutralizing antibodies, which is a pivotal immunogenic antigen for vaccine development. Here we constructed a recombinant chimeric vesicular stomatitis virus (VSV) VSVΔG-SARS, in which the glycoprotein (G) gene is replaced with the SARS-CoV S gene. VSVΔG-SARS maintains the bullet-like shape of the native VSV, with the heterogeneous S protein incorporated into its surface instead of G protein. The results of safety trials revealed that VSVΔG-SARS is safe and effective in mice at a dose of 1 â€‹× â€‹106 TCID50. More importantly, only a single-dose immunization of 2 â€‹× â€‹107 TCID50 can provide high-level neutralizing antibodies and robust T cell responses to non-human primate animal models. Thus, our data indicate that VSVΔG-SARS can be used as a rapid response vaccine candidate. Our study on the recombinant VSV-vectored SARS-CoV vaccines can accumulate experience and provide a foundation for the new coronavirus disease in the future.


Assuntos
COVID-19 , Coronavírus Relacionado à Síndrome Respiratória Aguda Grave , Animais , Anticorpos Neutralizantes , Anticorpos Antivirais , Imunização , Imunogenicidade da Vacina , Macaca mulatta , Camundongos , Coronavírus Relacionado à Síndrome Respiratória Aguda Grave/genética , Glicoproteína da Espícula de Coronavírus , Vacinas Sintéticas/genética , Vírus da Estomatite Vesicular Indiana/genética , Vírus da Estomatite Vesicular Indiana/metabolismo
16.
Viruses ; 14(9)2022 09 10.
Artigo em Inglês | MEDLINE | ID: mdl-36146810

RESUMO

African swine fever (ASF) is a highly contagious and fatal disease caused by the African swine fever virus. Recently, the multigene family and CD2v gene-deleted ASF vaccine candidate HLJ/18-7GD was found to be safe and effective in laboratory and clinical trials. However, the immune-protective mechanisms underlying the effects of HLJ/18-7GD remain unclear. We assessed samples from pigs immunized with a single dose of 106 TCID50 HLJ/18-7GD. We found that pigs immunized with HLJ/18-7GD showed high levels of specific antibodies. T lymphocyte subsets (helper T cells (Th); cytotoxic T lymphocytes (CTL); double-positive T cells (DP-T cells)) were temporarily increased in peripheral blood mononuclear cells (PBMCs) after HLJ/18-7GD immunization. Once the HLJ/18-7GD-immunized pigs had been challenged with virulent HLJ/18, the percentage of Th, CTL, and DP-T cells increased significantly. PBMCs extracted from the pigs induced higher levels of CD8+ T cells after infection with the HLJ/18 strain in vitro. The levels of GM-CSF, IFN-γ, and TNF-α were upregulated at 7 days post-inoculation; this finding was contrary to the results obtained after HLJ/18 or HLJ/18ΔCD2v infection. The immune protection from HLJ/18-7GD resulted from many synergies, which could provide a theoretical basis for HLJ/18-7GD as a safe and effective ASF vaccine.


Assuntos
Vírus da Febre Suína Africana , Febre Suína Africana , Vacinas Virais , Animais , Linfócitos T CD8-Positivos , Fator Estimulador de Colônias de Granulócitos e Macrófagos , Leucócitos Mononucleares , Suínos , Fator de Necrose Tumoral alfa , Proteínas Virais
17.
mBio ; 13(1): e0244321, 2022 02 22.
Artigo em Inglês | MEDLINE | ID: mdl-35012335

RESUMO

Loss of the furin cleavage motif in the SARS-CoV-2 spike protein reduces the virulence and transmission of SARS-CoV-2, suggesting that furin is an attractive antiviral drug target. However, lack of understanding of the regulation of furin activity has largely limited the development of furin-based therapeutic strategies. Here, we find that alpha-soluble NSF attachment protein (α-SNAP), an indispensable component of vesicle trafficking machinery, inhibits the cleavage of SARS-CoV-2 spike protein and other furin-dependent virus glycoproteins. SARS-CoV-2 infection increases the expression of α-SNAP, and overexpression of α-SNAP reduces SARS-CoV-2 infection in cells. We further reveal that α-SNAP is an interferon-upregulated furin inhibitor that inhibits furin function by interacting with its P domain. Our study demonstrates that α-SNAP, in addition to its role in vesicle trafficking, plays an important role in the host defense against furin-dependent virus infection and therefore could be a target for the development of therapeutic options for COVID-19. IMPORTANCE Some key mutations of SARS-CoV-2 spike protein, such as D614G and P681R mutations, increase the transmission or pathogenicity by enhancing the cleavage efficacy of spike protein by furin. Loss of the furin cleavage motif of SARS-CoV-2 spike protein reduces the virulence and transmission, suggesting that furin is an attractive antiviral drug target. However, lack of understanding of the regulation of furin activity has largely limited the development of furin-based therapeutic strategies. Here, we found that in addition to its canonical role in vesicle trafficking, alpha-soluble NSF attachment protein (α-SNAP) plays an important role in the host defense against furin-dependent virus infection. we identified that α-SNAP is a novel interferon-upregulated furin inhibitor and inhibits the cleavage of SARS-CoV-2 spike protein and other furin-dependent virus glycoproteins by interacting with P domain of furin. Our study demonstrates that α-SNAP could be a target for the development of therapeutic options for COVID-19.


Assuntos
COVID-19 , Glicoproteína da Espícula de Coronavírus , Humanos , Glicoproteína da Espícula de Coronavírus/metabolismo , SARS-CoV-2/metabolismo , Furina/metabolismo , Proteínas de Ligação a Fator Solúvel Sensível a N-Etilmaleimida/metabolismo , Interferons/metabolismo , Proteínas de Transporte , Antivirais , Glicoproteínas/metabolismo
18.
Cell Discov ; 7(1): 119, 2021 Dec 14.
Artigo em Inglês | MEDLINE | ID: mdl-34903715

RESUMO

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) uses angiotensin-converting enzyme 2 (ACE2) as a binding receptor to enter cells via clathrin-mediated endocytosis (CME). However, receptors involved in other steps of SARS-CoV-2 infection remain largely unknown. Here, we found that metabotropic glutamate receptor subtype 2 (mGluR2) is an internalization factor for SARS-CoV-2. Our results show that mGluR2 directly interacts with the SARS-CoV-2 spike protein and that knockdown of mGluR2 decreases internalization of SARS-CoV-2 but not cell binding. Further, mGluR2 is uncovered to cooperate with ACE2 to facilitate SARS-CoV-2 internalization through CME and mGluR2 knockout in mice abolished SARS-CoV-2 infection in the nasal turbinates and significantly reduced viral infection in the lungs. Notably, mGluR2 is also important for SARS-CoV spike protein- and Middle East respiratory syndrome coronavirus spike protein-mediated internalization. Thus, our study identifies a novel internalization factor used by SARS-CoV-2 and opens a new door for antiviral development against coronavirus infection.

19.
Natl Sci Rev ; 8(3): nwaa291, 2021 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-34676095

RESUMO

Minks are raised in many countries and have transmitted severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) to humans. However, the biologic properties of SARS-CoV-2 in minks are largely unknown. Here, we investigated and found that SARS-CoV-2 replicates efficiently in both the upper and lower respiratory tracts, and transmits efficiently in minks via respiratory droplets; pulmonary lesions caused by SARS-CoV-2 in minks are similar to those seen in humans with COVID-19. We further found that a spike protein-based subunit vaccine largely prevented SARS-CoV-2 replication and lung damage caused by SARS-CoV-2 infection in minks. Our study indicates that minks are a useful animal model for evaluating the efficacy of drugs or vaccines against COVID-19 and that vaccination is a potential strategy to prevent minks from transmitting SARS-CoV-2.

20.
Emerg Microbes Infect ; 10(1): 2183-2193, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-34709128

RESUMO

The Georgia-07-like genotype II African swine fever virus (ASFV) with high virulence has been prevalent in China since 2018. Here, we report that genotype I ASFVs have now also emerged in China. Two non-haemadsorbing genotype I ASFVs, HeN/ZZ-P1/21 and SD/DY-I/21, were isolated from pig farms in Henan and Shandong province, respectively. Phylogenetic analysis of the whole genome sequences suggested that both isolates share high similarity with NH/P68 and OURT88/3, two genotype I ASFVs isolated in Portugal in the last century. Animal challenge testing revealed that SD/DY-I/21 shows low virulence and efficient transmissibility in pigs, and causes mild onset of infection and chronic disease. SD/DY-I/21 was found to cause necrotic skin lesions and joint swelling. The emergence of genotype I ASFVs will present more problems and challenges for the control and prevention of African swine fever in China.


Assuntos
Vírus da Febre Suína Africana/genética , Vírus da Febre Suína Africana/isolamento & purificação , Febre Suína Africana/virologia , Febre Suína Africana/epidemiologia , Febre Suína Africana/transmissão , Vírus da Febre Suína Africana/classificação , Vírus da Febre Suína Africana/patogenicidade , Animais , China/epidemiologia , Genoma Viral , Genótipo , Filogenia , Sus scrofa/virologia , Suínos , Virulência
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA