Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
J Virol ; 96(13): e0014322, 2022 07 13.
Artigo em Inglês | MEDLINE | ID: mdl-35658531

RESUMO

Differentiation of infected from vaccinated hosts (DIVH) is a critical step in virus eradication programs. DIVH-compatible vaccines, however, take years to develop, and are therefore unavailable for fighting the sudden outbreaks that typically drive pandemics. Here, we establish a protocol for the swift and efficient development of DIVH assays, and show that this approach is compatible with any type of vaccines. Using porcine circovirus 2 (PCV2) as the experimental model, the first step is to use Immunoglobin G (IgG) sero-dynamics (IsD) curves to aid epitope discovery (IsDAED): PCV2 Cap peptides were categorized into three types: null interaction, nonspecific interaction (NSI), and specific interaction (SI). We subsequently compared IsDAED approach and traditional approach, and demonstrated identifying SI peptides and excluding NSI peptides supports efficient diagnostic kit development, specifically using a protein-peptide hybrid microarray (PPHM). IsDAED directed the design of a DIVH protocol for three types of PCV2 vaccines (while using a single PPHM). Finally, the DIVH protocol successfully differentiated infected pigs from vaccinated pigs at five farms. This IsDAED approach is almost certainly extendable to other viruses and host species. IMPORTANCE Sudden outbreaks of pandemics caused by virus, such as SARS-CoV-2, has been determined as a public health emergency of international concern. However, the development of a DIVH-compatible vaccine is time-consuming and full of uncertainty, which is unsuitable for an emergent situation like the ongoing COVID-19 pandemic. Along with the development and public health implementation of new vaccines to prevent human diseases, e.g., human papillomavirus vaccines for cervical cancer; enterovirus 71 vaccines for hand, foot, and mouth disease; and most recently SARS-CoV-2, there is an increasing demand for DIVH. Here, we use the IsDAED approach to confirm SI peptides and to exclude NSI peptides, finally to direct the design of a DIVH protocol. It is plausible that our IsDAED approach is applicable for other infectious disease.


Assuntos
Anticorpos Antivirais , Infecções por Circoviridae , Epitopos , Imunoglobulina G , Vacinas Virais , Animais , Anticorpos Antivirais/sangue , COVID-19 , Infecções por Circoviridae/imunologia , Circovirus , Modelos Animais de Doenças , Epitopos/análise , Epitopos/imunologia , Humanos , Imunoglobulina G/sangue , Imunoglobulina G/imunologia , Peptídeos , SARS-CoV-2 , Suínos , Doenças dos Suínos/imunologia , Vacinas Virais/imunologia
2.
PLoS One ; 17(2): e0263310, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35202392

RESUMO

Broccoli (Brassica oleracea var. italica) is an important B. oleracea cultivar, with high economic and agronomic value. However, comparative genome analyses are still needed to clarify variation among cultivars and phylogenetic relationships within the family Brassicaceae. Herein, the complete chloroplast (cp) genome of broccoli was generated by Illumina sequencing platform to provide basic information for genetic studies and to establish phylogenetic relationships within Brassicaceae. The whole genome was 153,364 bp, including two inverted repeat (IR) regions of 26,197 bp each, separated by a small single copy (SSC) region of 17,834 bp and a large single copy (LSC) region of 83,136 bp. The total GC content of the entire chloroplast genome accounts for 36%, while the GC content in each region of SSC,LSC, and IR accounts for 29.1%, 34.15% and 42.35%, respectively. The genome harbored 133 genes, including 88 protein-coding genes, 37 tRNAs, and 8 rRNAs, with 17 duplicates in IRs. The most abundant amino acid was leucine and the least abundant was cysteine. Codon usage analyses revealed a bias for A/T-ending codons. A total of 35 repeat sequences and 92 simple sequence repeats were detected, and the SC-IR boundary regions were variable between the seven cp genomes. A phylogenetic analysis suggested that broccoli is closely related to Brassica oleracea var. italica MH388764.1, Brassica oleracea var. italica MH388765.1, and Brassica oleracea NC_0441167.1. Our results are expected to be useful for further species identification, population genetics analyses, and biological research on broccoli.


Assuntos
Brassicaceae/genética , Genoma de Cloroplastos/genética , Filogenia , Sequenciamento Completo do Genoma , Composição de Bases/genética , Brassicaceae/classificação , Cloroplastos/genética , Códon/genética , Evolução Molecular , Sequenciamento de Nucleotídeos em Larga Escala , Repetições de Microssatélites/genética , Anotação de Sequência Molecular , RNA Ribossômico/genética , RNA de Transferência/genética , Análise de Sequência de DNA
3.
Antiviral Res ; 151: 63-70, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29337166

RESUMO

Porcine reproductive and respiratory syndrome virus (PRRSV) causes severe economic losses to current swine production worldwide. Highly pathogenic PRRSV (HP-PRRSV), originated from a genotype 2 PRRSV, is more virulent than classical PRRSV and further exacerbates the economic impact. HP-PRRSV has become the predominant circulating field strain in China since 2006. CD163 is a cellular receptor for PRRSV. The depletion of CD163 whole protein or SRCR5 region (interaction site for the virus) confers resistance to infection of several PRRSV isolates in pigs or cultured host cells. In this study, we described the generation of a CD163 knockout (KO) pig in which the CD163 protein was ablated by using CRISPR/Cas9 gene targeting and somatic cell nuclear transfer (SCNT) technologies. Challenge with HP-PRRSV TP strain showed that CD163 KO pigs are completely resistant to viral infection manifested by the absence of viremia, antibody response, high fever or any other PRRS-associated clinical signs. By comparison, wild-type (WT) controls displayed typical signs of PRRSV infection and died within 2 weeks after infection. Deletion of CD163 showed no adverse effects to the macrophages on immunophenotyping and biological activity as hemoglobin-haptoglobin scavenger. The results demonstrated that CD163 knockout confers full resistance to HP-PRRSV infection to pigs without impairing the biological function associated with the gene.


Assuntos
Resistência à Doença , Síndrome Respiratória e Reprodutiva Suína/prevenção & controle , Vírus da Síndrome Respiratória e Reprodutiva Suína/patogenicidade , Receptores de Superfície Celular/deficiência , Animais , Antígenos CD/química , Antígenos CD/genética , Antígenos de Diferenciação Mielomonocítica/química , Antígenos de Diferenciação Mielomonocítica/genética , Edição de Genes , Genótipo , Macrófagos/imunologia , Macrófagos/metabolismo , Macrófagos/virologia , Técnicas de Transferência Nuclear , Domínios e Motivos de Interação entre Proteínas/genética , Receptores de Superfície Celular/química , Receptores de Superfície Celular/genética , Suínos , Viremia/prevenção & controle
5.
Int J Cancer ; 126(11): 2741-51, 2010 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-19824041

RESUMO

Apoptin, a chicken anemia virus-derived, p53-independent, bcl-2-insenstive apoptotic protein with the ability to specifically induce apoptosis in tumor or transformed cells, is a promising tool for cancer gene therapy. In this study, pseudotype baculovirus, a recently developed alternative gene delivery system, was used as a vector to express Apoptin. The resultant recombinant baculovirus (BV-Apoptin) efficiently expressed the Apoptin protein and induced apoptosis in HepG2 and H22 cells. Studies in vivo showed that intratumoral injection of BV-Apoptin into a xenogeneic tumor (derived from H22 murine hepatoma cells in C57BL/6 mice) significantly suppressed tumor growth, and significantly prolonged the survival of tumor-bearing mice compared to a control pseudotype baculovirus that expressed EGFP. Taken together, these results suggest that Apoptin, expressed from the pseudotype baculovirus vector, has the potential to become a therapeutic agent for the treatment of solid tumors.


Assuntos
Proteínas do Capsídeo/genética , Carcinoma Hepatocelular/patologia , Animais , Apoptose , Western Blotting , Proteínas do Capsídeo/uso terapêutico , Linhagem Celular , Linhagem Celular Tumoral , Vírus da Anemia da Galinha/genética , Citometria de Fluxo , Terapia Genética/métodos , Vetores Genéticos , Humanos , Rim , Neoplasias Hepáticas Experimentais/mortalidade , Neoplasias Hepáticas Experimentais/patologia , Camundongos , Camundongos Endogâmicos C57BL , Taxa de Sobrevida
6.
Biotechnol Lett ; 31(4): 509-18, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19116689

RESUMO

We constructed a suicidal DNA vaccine pSFV-ORF5m/ORF6 co-expressing GP5m (a modified GP5) and M proteins of porcine reproductive and respiratory syndrome virus (PRRSV). In mice immunization, specific immune responses were elicited by the suicidal DNA vaccine pSFV-ORF5m/ORF6. The immunogenicity and protective efficiency was then evaluated in piglets immunized with pSFV-ORF5m/ORF6 before virus challenge: PRRSV-specific neutralizing antibodies and lymphocyte proliferative responses were developed. Post-PRRSV challenge, these immune responses were further boosted and partial protection was obtained.


Assuntos
Síndrome Respiratória e Reprodutiva Suína/prevenção & controle , Vírus da Síndrome Respiratória e Reprodutiva Suína/genética , Vacinas de DNA/genética , Vacinas de DNA/imunologia , Proteínas do Envelope Viral/imunologia , Proteínas da Matriz Viral/imunologia , Animais , Anticorpos Antivirais/sangue , Rim/virologia , Pulmão/patologia , Pulmão/virologia , Linfonodos/virologia , Testes de Neutralização , Tonsila Palatina/virologia , Síndrome Respiratória e Reprodutiva Suína/imunologia , Vírus da Síndrome Respiratória e Reprodutiva Suína/imunologia , Baço/virologia , Suínos , Proteínas do Envelope Viral/genética , Proteínas da Matriz Viral/genética
7.
Zhongguo Yi Xue Ke Xue Yuan Xue Bao ; 30(4): 498-505, 2008 Aug.
Artigo em Chinês | MEDLINE | ID: mdl-18795628

RESUMO

OBJECTIVE: To construct the recombinant adenovirus containing herpes simplex virus-1 virion protein (VP) 22 and human microdystrophin gene, then the adenovirus was transfected into C2C12 myoblast and studied on the property of protein transduction with VP22-mediated microdystrophin in C2C12 myoblast. METHODS: The full-length VP22 cDNA was obtained from recombinant plasmid pSINrep5-VP22 with PCR, and the product was directionally inserted into pShuttle-CMV to acquire the plasmid pCMV-VP22. Microdystrophin cDNA was obtained from recombinant plasmid pBSK-micro digested with restrictive endonuclease NotI, and the product was directionally inserted into pCMV-VP22 to acquire the plasmid pCMV-VP22-MICDYS. The plasmid of pCMV-VP22-MICDYS was lined with Pme I, and the fragment containing VP22-microdystrophin was reclaimed and transfected into E1 coli BJ5183 with plasmid pAdeasy-1. After having been screened by selected media, the extracted plasmid of positive bacteria was transfected into HEK293 cells with liposome and was identified by observing the cytopathic effect of cells and by PCR method to acquire the recombinant adenovirus Ad-VP22-MICDYS. Finally, the C2C12 myoblast were transfected with the recombinant adenovirus Ad-VP22-MICDYS and Ad-MICDYS, and the expression of microdystrophin was detected by RT-PCR, Western blot and immunocytochemistry. RESULTS: The recombinant adenovirus including VP22 and microdystrophin gene was successfully constructed. VP22 transferred VP22-microdystrophin fused protein from infected C2C12 myoblast into uninfected cells and enhance the expression of microdystrophin in myoblast. CONCLUSIONS: Recombinant adenovirus containing VP22 and microdystrophin gene was constructed successfully. VP22 can enhance the expression with microdystrophin in myoblast. It lays the foundation for further studying on VP22-mediated recombinant including microdystrophin gene to cure Duchenne muscular dystrophy.


Assuntos
Adenoviridae/genética , Distrofina/genética , Vetores Genéticos/genética , Transdução Genética , Proteínas Estruturais Virais/genética , Adenoviridae/fisiologia , Animais , Linhagem Celular , Distrofina/metabolismo , Vetores Genéticos/metabolismo , Humanos , Camundongos , Mioblastos/metabolismo , Mioblastos/virologia , Simplexvirus/genética , Simplexvirus/metabolismo , Proteínas Estruturais Virais/metabolismo , Vírion/genética , Vírion/metabolismo
8.
Vaccine ; 25(49): 8220-7, 2007 Nov 28.
Artigo em Inglês | MEDLINE | ID: mdl-17980465

RESUMO

Baculovirus, which is extensively utilized as an excellent tool for production of recombinant protein in insect cells, has recently emerged as a novel and attractive gene delivery vehicle for mammalian cells. In the present study, a pseudotype baculovirus (with the glycoprotein of vesicular stomatitis virus (VSV-G) on the envelope) was used as vector to construct recombinant baculovirus coexpressing GP5 and M protein of porcine reproductive and respiratory syndrome virus (PRRSV), under the transcriptional control of two independent cytomegalovirus immediate early (CMV-IE) enhancer/promoters. The resultant recombinant baculovirus (BV-G-5m6) efficiently expressed PRRSV GP5 and M protein in mammalian cells. Intramuscular injection of BV-G-5m6 with various doses (1 x 10(8), 1 x 10(9), and 1 x 10(10)PFU/mouse) induced the production of PRRSV-specific neutralizing antibodies and gamma interferon (IFN-gamma) under dose-dependent pattern. Furthermore, BV-G-5m6 performed better immunogenicity, even at low dose (10(8)PFU), than DNA construct (pCI-5m6) encoding the same antigens, as demonstrated by significantly enhanced neutralizing antibodies and IFN-gamma production. These results indicate that pseudotype baculovirus-mediated gene delivery can be utilized as an alternative strategy to develop new generation of vaccine against PRRSV infection.


Assuntos
Anticorpos Antivirais/sangue , Baculoviridae/genética , Recombinação Genética , Proteínas do Envelope Viral , Proteínas da Matriz Viral , Animais , Baculoviridae/imunologia , Baculoviridae/metabolismo , Linhagem Celular , Feminino , Interferon gama/biossíntese , Camundongos , Camundongos Endogâmicos BALB C , Testes de Neutralização , Síndrome Respiratória e Reprodutiva Suína/prevenção & controle , Vírus da Síndrome Respiratória e Reprodutiva Suína/genética , Vírus da Síndrome Respiratória e Reprodutiva Suína/imunologia , Vírus da Síndrome Respiratória e Reprodutiva Suína/metabolismo , Suínos , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/imunologia , Proteínas do Envelope Viral/metabolismo , Proteínas da Matriz Viral/genética , Proteínas da Matriz Viral/imunologia , Proteínas da Matriz Viral/metabolismo , Vacinas Virais/administração & dosagem , Vacinas Virais/genética , Vacinas Virais/imunologia
9.
Vaccine ; 25(3): 547-60, 2007 Jan 05.
Artigo em Inglês | MEDLINE | ID: mdl-16920232

RESUMO

Porcine reproductive and respiratory syndrome virus (PRRSV) infection still remains today as the most significant health threat to swine and poses a challenge to current vaccination strategies. To develop a new generation of vaccine against PRRSV, a live attenuated pseudorabies virus (PRV) was used as vaccine vector to express the two major membrane-associated proteins (GP5 or M) of PRRSV in various forms. Four PRV recombinants, rPRV-GP5 (expressing native GP5), rPRV-GP5m (expressing GP5m, a modified GP5), rPRV-GP5-M (co-expressing GP5 and M proteins), rPRV-GP5m-M (co-expressing GP5m and M proteins) were generated. Mouse immunized with all these recombinants developed comparable PRV-specific humoral immune responses and provided complete protection against a lethal PRV challenge. However, the highest level of PRRSV-specific neutralizing antibodies and lymphocyte proliferative responses was observed in mice immunized with rPRV-GP5m-M. The immunogenicity and protective efficiency of rPRV-GP5m-M were further evaluated in the piglets. Compared to commercial PRRSV killed vaccine, detectable PRRSV-specific neutralizing antibody and higher lymphocyte proliferative responses could be developed in piglets immunized with rPRV-GP5m-M before virus challenge. Furthermore, more efficient protection against a PRRSV challenge was obtained in piglets immunized with rPRV-GP5m-M, as showed by the balanced body-temperature fluctuation, shorter-term viremia, lower proportion of virus load in nasal and oropharyngeal scrapings and tissues, and milder lung lesions. These data indicate that the recombinant rPRV-GP5m-M is a promising candidate bivalent vaccine against both PRV and PRRSV infection.


Assuntos
Herpesvirus Suídeo 1/genética , Herpesvirus Suídeo 1/imunologia , Síndrome Respiratória e Reprodutiva Suína/imunologia , Síndrome Respiratória e Reprodutiva Suína/prevenção & controle , Vírus da Síndrome Respiratória e Reprodutiva Suína/genética , Vírus da Síndrome Respiratória e Reprodutiva Suína/imunologia , Vacinas contra Pseudorraiva/imunologia , Pseudorraiva/imunologia , Pseudorraiva/prevenção & controle , Animais , Temperatura Corporal/fisiologia , Proliferação de Células , Feminino , Linfócitos/imunologia , Proteínas de Membrana/genética , Proteínas de Membrana/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Plasmídeos/genética , Síndrome Respiratória e Reprodutiva Suína/patologia , Pseudorraiva/patologia , RNA Viral/biossíntese , RNA Viral/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Suínos , Vacinas Atenuadas , Proteínas do Envelope Viral/imunologia , Viremia/virologia , Eliminação de Partículas Virais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA