Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 79
Filtrar
Mais filtros

Bases de dados
País/Região como assunto
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
Nature ; 579(7798): 270-273, 2020 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-32015507

RESUMO

Since the outbreak of severe acute respiratory syndrome (SARS) 18 years ago, a large number of SARS-related coronaviruses (SARSr-CoVs) have been discovered in their natural reservoir host, bats1-4. Previous studies have shown that some bat SARSr-CoVs have the potential to infect humans5-7. Here we report the identification and characterization of a new coronavirus (2019-nCoV), which caused an epidemic of acute respiratory syndrome in humans in Wuhan, China. The epidemic, which started on 12 December 2019, had caused 2,794 laboratory-confirmed infections including 80 deaths by 26 January 2020. Full-length genome sequences were obtained from five patients at an early stage of the outbreak. The sequences are almost identical and share 79.6% sequence identity to SARS-CoV. Furthermore, we show that 2019-nCoV is 96% identical at the whole-genome level to a bat coronavirus. Pairwise protein sequence analysis of seven conserved non-structural proteins domains show that this virus belongs to the species of SARSr-CoV. In addition, 2019-nCoV virus isolated from the bronchoalveolar lavage fluid of a critically ill patient could be neutralized by sera from several patients. Notably, we confirmed that 2019-nCoV uses the same cell entry receptor-angiotensin converting enzyme II (ACE2)-as SARS-CoV.


Assuntos
Betacoronavirus/classificação , Betacoronavirus/genética , Quirópteros/virologia , Infecções por Coronavirus/epidemiologia , Infecções por Coronavirus/virologia , Surtos de Doenças , Pneumonia Viral/epidemiologia , Pneumonia Viral/virologia , Enzima de Conversão de Angiotensina 2 , Animais , Anticorpos Antivirais/sangue , Betacoronavirus/metabolismo , Betacoronavirus/ultraestrutura , COVID-19 , Linhagem Celular , China/epidemiologia , Chlorocebus aethiops , Feminino , Genoma Viral/genética , Humanos , Masculino , Peptidil Dipeptidase A/metabolismo , Filogenia , Coronavírus Relacionado à Síndrome Respiratória Aguda Grave/classificação , Coronavírus Relacionado à Síndrome Respiratória Aguda Grave/genética , SARS-CoV-2 , Homologia de Sequência do Ácido Nucleico , Síndrome Respiratória Aguda Grave , Células Vero
2.
J Gen Virol ; 105(1)2024 01.
Artigo em Inglês | MEDLINE | ID: mdl-38180473

RESUMO

Nipah virus (NiV) is a deadly zoonotic pathogen with high potential to cause another pandemic. Owing to biosafety concerns, studies on living NiV must be performed in biosafety level 4 (BSL-4) laboratories, which greatly hinders the development of anti-NiV drugs. To overcome this issue, minigenome systems have been developed to study viral replication and screen for antiviral drugs. This study aimed to develop two minigenome systems (transient and stable expression) based on a helper cell line expressing the NiV P, N and L proteins required to initiate NiV RNA replication. Stable minigenome cells were resistant to ribavirin, remdesivir and favipiravir but sensitive to interferons. Cells of the transient replication system were sensitive to ribavirin and favipiravir and suitable for drug screening. Our study demonstrates a feasible and effective platform for studying NiV replication and shows great potential for high-throughput drug screening in a BSL-2 laboratory environment.


Assuntos
Vírus Nipah , Vírus Nipah/genética , Ribavirina , Replicação Viral , Antivirais/farmacologia
3.
J Virol ; 96(12): e0049422, 2022 06 22.
Artigo em Inglês | MEDLINE | ID: mdl-35604143

RESUMO

G protein subunit ß1 (GNB1), the beta subunit of the G protein family, plays an important role in regulating transmembrane signal transduction. Although a recent study has demonstrated that GNB1 can bind the matrix protein 1 (M1) to facilitate M1 transport to budding sites and promote the release of progeny influenza A virus (IAV), whether the GNB1 protein has other functions in IAV replication requires further study. Here, we found that GNB1 promoted IAV replication, as virus yield decreased in GNB1 knockdown or knockout cells. GNB1 interacted with polymerase subunits PB2, PB1, and PA. Overexpressed GNB1 facilitated PB2 binding to importin α3, α5, and α7 promoting the nuclear import of PB2, enhancing viral RNA synthesis and polymerase activity. Altogether, our results demonstrated that GNB1 positively regulates virus replication by interacting with polymerase subunits and facilitating the nuclear import of PB2, which provide novel insights into the molecular mechanism of IAV. IMPORTANCE Until now, there has been only one article on the role of GNB1 in IAV budding. No study has investigated the role of GNB1 in IAV replication. In this study, our research demonstrated that GNB1 could increase the interaction between PB2 and the importin α isoform and mediate the nuclear import of PB2. Therefore, GNB1 could promote viral replication and transcription. Our results provide a better understanding of the molecular mechanisms of viral replication and provide potential antiviral drug targets.


Assuntos
Transporte Ativo do Núcleo Celular , Subunidades beta da Proteína de Ligação ao GTP , Vírus da Influenza A , Influenza Humana , Proteínas Virais , Subunidades beta da Proteína de Ligação ao GTP/metabolismo , Humanos , Vírus da Influenza A/genética , Vírus da Influenza A/fisiologia , Influenza Humana/genética , Carioferinas/metabolismo , Proteínas Virais/genética , Proteínas Virais/metabolismo , Replicação Viral
4.
J Med Virol ; 95(3): e28657, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36912367

RESUMO

Novel immune escape variants have emerged as severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) continues to spread worldwide. Many of the variants cause breakthrough infections in vaccinated populations, posing great challenges to current antiviral strategies targeting the immunodominance of the receptor-binding domain within the spike protein. Here, we found that a novel broadly neutralizing monoclonal antibody (mAb), G5, provided efficient protection against SARS-CoV-2 variants of concern (VOCs) in vitro and in vivo. A single dose of mAb G5 could significantly inhibit the viral burden in mice challenged with the mouse-adapted SARS-CoV-2 or SARS-CoV-2 Omicron BA.1 variant, as well as the body weight loss and cytokine release induced by mouse-adapted SARS-CoV-2. The refined epitope recognized by mAb G5 was identified as 1148 FKEELDKYF1156 in the stem helix of subunit S2. In addition, a human-mouse chimeric mAb was generated based on the variable region of heavy chain and VL genes of mAb G5. Our study provides a broad antibody drug candidate against SARS-CoV-2 VOCs and reveals a novel target for developing pan-SARS-CoV-2 vaccines.


Assuntos
Anticorpos Monoclonais , COVID-19 , Humanos , Animais , Camundongos , Anticorpos Monoclonais/uso terapêutico , Vacinas contra COVID-19 , SARS-CoV-2/genética , Imunossupressores , Glicoproteína da Espícula de Coronavírus/genética , Anticorpos Neutralizantes , Anticorpos Antivirais/uso terapêutico
5.
Clin Infect Dis ; 73(3): 361-366, 2021 08 02.
Artigo em Inglês | MEDLINE | ID: mdl-32638022

RESUMO

BACKGROUND: Currently, there is no direct evidence to prove the active replication of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in the intestinal tract and relevant pathological changes in the colon and rectum. We investigated the presence of virions and pathological changes in surgical rectal tissues of a patient with clinically confirmed coronavirus disease 2019 (COVID-19) with rectal adenocarcinoma. METHODS: The clinical data were collected during hospitalization and follow-up of this patient. Quantitative reverse transcriptase-polymerasechain reaction (RT-PCR) was performed on the rectal tissue specimens obtained from surgical resection, succus entericus and intestinal mucosa of ileostomy, and rectal mucosa during follow-up after recovery. Ultrathin sections of surgical samples were observed for SARS-CoV-2 virions using electron microscopy. Histopathological examination was performed using hematoxylin-eosin stain. Immunohistochemical analysis and immunofluorescence were carried out on rectal tissues to evaluate the distribution of SARS-CoV-2 antigen and immune cell infiltrations. RESULTS: The patient had fever and cough on day 3 postoperatively, was diagnosed with COVID-19 on day 7, and was discharged from the hospital on day 41. RNA of SARS-CoV-2 was detected in surgically resected rectal specimens but not in samples collected 37 days after discharge. Notably, coincident with rectal tissues of surgical specimens testing nucleic acid positive for SARS-CoV-2, typical coronavirus virions in rectal tissue were observed under electron microscopy. Moreover, abundant lymphocytes and macrophages (some were SARS-CoV-2 positive) infiltrating the lamina propria were found with no significant mucosal damage. CONCLUSIONS: We first report the direct evidence of active SARS-CoV-2 replication in a patient's rectum during the incubation period, which might explain SARS-CoV-2 fecal-oral transmission.


Assuntos
COVID-19 , SARS-CoV-2 , Hospitalização , Humanos , Intestinos , Alta do Paciente , RNA Viral
6.
Am J Physiol Gastrointest Liver Physiol ; 321(4): G436-G447, 2021 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-34405716

RESUMO

Chronic constipation (CC) is a gastrointestinal disorder that adversely affects the quality of life. MicroRNAs are involved in the pathogenesis of functional gastrointestinal disorders. This study aims to investigate the molecular mechanism of microRNA-128 in CC. Here, we successfully constructed a murine model of CC based on morphine and rhubarb. The expression of stem cell factor (SCF) and neuron-specific enolase (NSE) was low in the models. Using miRNA array and bioinformatic analysis, we predicted and confirmed the expression of miR-128 and its downstream target genes in CC model. Compared with the control group, CC group showed a significant downregulation of miR-128 and upregulation of p38α and macrophage colony-stimulating factors (M-CSFs). Moreover, we observed elevated inflammatory cytokine and decreased anti-inflammatory cytokine levels in colonic tissues. Furthermore, coculture assays indicated that regulating expression of miR-128 in colonic epithelial cells induced the secretion of IL-6 and TNF-α by macrophages. In conclusion, our study demonstrated that miR-128 regulated the p38α/M-CSF signaling pathway to promote chronic inflammatory responses and changes in the immune microenvironment of the colon, thereby offering potential insights into the pathogenesis of CC and therapeutic targets for its treatment.NEW & NOTEWORTHY In this study, we constructed a murine model and identified a novel signaling mechanism involved in the chronic constipation progression. Our findings on the role of miR-128/p38α/M-CSF axis provide new insights into the treatment of chronic constipation.


Assuntos
Constipação Intestinal/metabolismo , Fator Estimulador de Colônias de Macrófagos/metabolismo , MicroRNAs/metabolismo , Transdução de Sinais , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Animais , Linhagem Celular Tumoral , Colo/metabolismo , Constipação Intestinal/genética , Feminino , Interleucina-6/metabolismo , Mucosa Intestinal/metabolismo , Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos ICR , MicroRNAs/genética , Células RAW 264.7 , Fator de Necrose Tumoral alfa/metabolismo
8.
Langmuir ; 36(50): 15212-15219, 2020 12 22.
Artigo em Inglês | MEDLINE | ID: mdl-33307709

RESUMO

The resistance of drugs to the new influenza A virus (IAV) strains and the limited efficiency of vaccines to prevent seasonal flu epidemics underscore the urgency in finding novel strategies to block IAV infection, which is required to gain insights into the mechanism of the initial step of IAV adhesion. While it is well established that IAVs bind to respiratory tract cells by recognizing sialylated glycans on host cell membranes through a multivalency effect, how IAVs dynamically respond to multiple glycan receptors via distinct valencies has not been fully understood, limiting the discovery of novel anti-flu strategies. Using single-particle tracking to record the 2D mobilities and surface residence times of highly pathogenic H5N1 avian IAVs adhered to fluidic membranes containing α2-3 sialylated GM3 glycolipids, we quantified the univalent and multivalent IAV adhesion channels, which provide insights into the mechanism of IAV binding; IAV can guide the clustering of dynamic glycolipids to statistically match the multivalent binding affinities for IAV adhesion. This mechanism can be inhibited by disrupting the dynamic glycan clustering on membranes of varying fluidities, like the gel phase membrane. This work facilitates a deeper fundamental understanding of IAV infection as well as the development of novel anti-flu strategies.


Assuntos
Virus da Influenza A Subtipo H5N1 , Vírus da Influenza A , Lipídeos , Membranas , Polissacarídeos
10.
Emerg Infect Dis ; 23(4): 637-641, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-28169827

RESUMO

In May 2016, a highly pathogenic avian influenza A(H5N8) virus strain caused deaths among 3 species of wild migratory birds in Qinghai Lake, China. Genetic analysis showed that the novel reassortant virus belongs to group B H5N8 viruses and that the reassortment events likely occurred in early 2016.


Assuntos
Migração Animal , Animais Selvagens , Anseriformes , Charadriiformes , Vírus da Influenza A Subtipo H5N8 , Influenza Aviária/virologia , Animais , China/epidemiologia , Surtos de Doenças/veterinária , Vírus da Influenza A Subtipo H5N8/genética , Influenza Aviária/epidemiologia , Filogenia , Vírus Reordenados
11.
J Virol ; 90(4): 2142-9, 2016 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-26656694

RESUMO

The neuraminidase stalk of the newly emerged H7N9 influenza virus possesses a 5-amino-acid deletion. This study focuses on characterizing the biological functions of H7N9 with varied neuraminidase stalk lengths. Results indicate that the 5-amino-acid deletion had no impact on virus infectivity or replication in vitro or in vivo compared to that of a virus with a full-length stalk, but enhanced virulence in mice was observed for H7N9 encoding a 19- to 20-amino-acid deletion, suggesting that N9 stalk length impacts virulence in mammals, as N1 stalk length does.


Assuntos
Subtipo H7N9 do Vírus da Influenza A/genética , Subtipo H7N9 do Vírus da Influenza A/patogenicidade , Neuraminidase/genética , Infecções por Orthomyxoviridae/virologia , Proteínas Virais/genética , Fatores de Virulência/genética , Animais , Peso Corporal , Citocinas/análise , Histocitoquímica , Pulmão/patologia , Pulmão/virologia , Camundongos Endogâmicos BALB C , Infecções por Orthomyxoviridae/patologia , Deleção de Sequência , Carga Viral , Virulência
12.
J Gen Virol ; 96(Pt 5): 975-981, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25604926

RESUMO

Eight avian influenza A (H5N6) viruses were isolated from live poultry markets (LPMs) in Sichuan and Jiangxi Provinces in China in 2014, including those close to the county where the human H5N6 infection occurred. Genetic and phylogenetic analyses revealed that these H5N6 viruses were novel reassortants between H5N1 clade 2.3.4 and H6N6 viruses, and had evolved into two distinct lineages (Sichuan and Jiangxi). Moreover, the human H5N6 virus was closely related to the avian-source viruses of Sichuan lineage. Notably, H5N6 viruses contained a T160A substitution in the haemagglutinin protein and an 11 aa deletion in the neuraminidase stalk, which may aid in enhancing viral affinity for human-like receptors and virulence in mammals. As the H5N1 virus infects humans through direct contact, infection with the novel H5N6 virus raised significant concerns that the H5 subtype was a likely candidate for a pandemic. Therefore, extensive and long-term surveillance of avian influenza viruses in LPMs is essential.


Assuntos
Vírus da Influenza A/genética , Vírus da Influenza A/isolamento & purificação , Influenza Aviária/virologia , Vírus Reordenados/genética , Vírus Reordenados/isolamento & purificação , Substituição de Aminoácidos , Animais , China , Análise por Conglomerados , Glicoproteínas de Hemaglutininação de Vírus da Influenza/genética , Dados de Sequência Molecular , Neuraminidase/genética , Filogenia , Aves Domésticas , RNA Viral/genética , Análise de Sequência de DNA , Deleção de Sequência , Homologia de Sequência , Proteínas Virais/genética , Virulência
13.
Emerg Infect Dis ; 20(12): 2076-9, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25425075

RESUMO

Human infection with avian influenza virus A(H10N8) was initially reported in China in December 2013. We characterized H10N8 strains from a human patient and from poultry in live markets that infected persons had visited. Results of genome sequencing and virus characterization suggest that the virus strains that infected humans originated from these markets.


Assuntos
Vírus da Influenza A Subtipo H10N8/classificação , Influenza Humana/epidemiologia , Influenza Humana/transmissão , Animais , China/epidemiologia , Genes Virais , Humanos , Vírus da Influenza A Subtipo H10N8/genética , Influenza Humana/virologia , Filogenia , Aves Domésticas/virologia
14.
J Gen Virol ; 95(Pt 7): 1430-1435, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24671753

RESUMO

Water bodies are an important route for the spread and transmission of avian influenza virus (AIV). The determining factor for an AIV to transmit through diffusion in water is the term of viability of the virus in the water body. To better understand the perpetuation of AIV in natural water bodies, and thus the risks of AIV spread and transmission via such bodies, we systematically studied the inactivation dynamics of two AIV strains (H5N1 and H9N2) at different temperatures in water bodies of important migratory bird habitats within China (Dongting Lake, Poyang Lake, the Hubei segment of the Yangtze River and Qinghai Lake). We also studied the impact of water-borne micro-organisms on the perpetuation of AIV. Our findings indicated that water is very likely an important route for the epidemic spread of AIV, especially during the autumn and winter seasons. In addition, water-borne micro-organisms might antagonize the persistence of AIV.


Assuntos
Virus da Influenza A Subtipo H5N1/fisiologia , Vírus da Influenza A Subtipo H9N2/fisiologia , Viabilidade Microbiana , Microbiologia da Água , Animais , Aves , China , Influenza Aviária/transmissão , Lagos/virologia , Estações do Ano , Temperatura , Fatores de Tempo
15.
J Virol ; 87(2): 767-78, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23115280

RESUMO

Influenza A virus NS2 protein, also called nuclear export protein (NEP), is crucial for the nuclear export of viral ribonucleoproteins. However, the molecular mechanisms of NEP mediation in this process remain incompletely understood. A leucine-rich nuclear export signal (NES2) in NEP, located at the predicted N2 helix of the N-terminal domain, was identified in the present study. NES2 was demonstrated to be a transferable NES, with its nuclear export activity depending on the nuclear export receptor chromosome region maintenance 1 (CRM1)-mediated pathway. The interaction between NEP and CRM1 is coordinately regulated by both the previously reported NES (NES1) and now the new NES2. Deletion of the NES1 enhances the interaction between NEP and CRM1, and deletion of the NES1 and NES2 motifs completely abolishes this interaction. Moreover, NES2 interacts with CRM1 in the mammalian two-hybrid system. Mutant viruses containing NES2 alterations generated by reversed genetics exhibit reduced viral growth and delay in the nuclear export of viral ribonucleoproteins (vRNPs). The NES2 motif is highly conserved in the influenza A and B viruses. The results demonstrate that leucine-rich NES2 is involved in the nuclear export of vRNPs and contributes to the understanding of nucleocytoplasmic transport of influenza virus vRNPs.


Assuntos
Transporte Ativo do Núcleo Celular , Vírus da Influenza A/fisiologia , Sinais de Exportação Nuclear , Ribonucleoproteínas/metabolismo , Proteínas não Estruturais Virais/metabolismo , Replicação Viral , Linhagem Celular , Sequência Conservada , Humanos , Vírus da Influenza A/genética , Ligação Proteica , Mapeamento de Interação de Proteínas , Deleção de Sequência , Técnicas do Sistema de Duplo-Híbrido , Proteínas não Estruturais Virais/genética
16.
Arch Virol ; 159(4): 689-700, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24132721

RESUMO

Plasmid DNA vaccines are considered alternatives to inactivated influenza virus vaccines to control influenza. Vaccination with a hemagglutinin (HA)-, HA ectodomain (HAe)-, or HA subunit 1 (HA1)-based vaccine can stimulate protective immunity in animals. The aim of this study was to compare their capacity to induce an antibody response and protection against influenza virus infection in mice after DNA vaccination. We constructed three expression vectors encoding full-length HA, HAe, or HA1 of the A/California/07/2009 influenza A virus and designed three animal experiments: (i) BALB/c mice were immunized twice with 30 µg of the HA, HAe, or HA1 DNA vaccine with high-voltage electroporation (100 V), and 3 weeks after boosting, they were challenged with a lethal dose of virus. (ii) Immunization and challenge were as in experiment i, but with low-voltage electroporation (10 V). (iii) Mice were immunized once with 50 µg of DNA and challenged 1 week later. The immunogenic effects of the three DNA vaccines were evaluated in terms of antibody titer, survival rate, bodyweight change, and lung viral titer. In all three experiments, both HA and HAe induced higher antibody and neutralization titers than HA1. Following challenge with a lethal mouse-adapted homologous virus, both HA and HAe reduced the viral titers in lung washes or offered better protection from weight loss than HA1 in experiments ii and iii. Thus, HA1 induces a lower immune response than HA or HAe when used as a DNA vaccination. Our data should be valuable in choosing the optimal candidate vaccine when faced with the threat of pandemic influenza.


Assuntos
Anticorpos Antivirais/sangue , Glicoproteínas de Hemaglutininação de Vírus da Influenza/imunologia , Vírus da Influenza A/imunologia , Vacinas contra Influenza/imunologia , Infecções por Orthomyxoviridae/prevenção & controle , Vacinação/métodos , Vacinas de DNA/imunologia , Animais , Peso Corporal , Modelos Animais de Doenças , Feminino , Glicoproteínas de Hemaglutininação de Vírus da Influenza/genética , Vírus da Influenza A/genética , Vacinas contra Influenza/genética , Pulmão/virologia , Camundongos , Camundongos Endogâmicos BALB C , Infecções por Orthomyxoviridae/patologia , Análise de Sobrevida , Vacinas de DNA/genética , Carga Viral
17.
Arch Virol ; 159(3): 471-83, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24057757

RESUMO

Pandemic influenza A virus (H1N1) 2009 poses a serious public-health challenge worldwide. To characterize the neutralizing epitopes of this virus, we generated a panel of eight monoclonal antibodies (mAbs) against the HA of the A/California/07/2009 virus. The antibodies were specific for the 2009 pdm H1N1 HA, as the antibodies displayed HA-specific ELISA, hemagglutination inhibition (HAI) and neutralization activity. One mAb (mAb12) showed significantly higher HAI and neutralizing titers than the other mAbs. We mapped the antigenic epitopes of the HA by characterizing escape mutants of a 2009 H1N1 vaccine strain (NYMC X-179A). The amino acid changes suggested that these eight mAbs recognized HA antigenic epitopes located in the Sa, Sb, Ca1 and Ca2 sites. Passive immunization with mAbs showed that mAb12 displayed more efficient neutralizing activity in vivo than the other mAbs. mAb12 was also found to be protective, both prophylactically and therapeutically, against a lethal viral challenge in mice. In addition, a single injection of 10 mg/kg mAb12 outperformed a 5-day course of treatment with oseltamivir (10 mg/kg/day by gavage) with respect to both prophylaxis and treatment of lethal viral infection. Taken together, our results showed that mouse-origin mAbs displayed neutralizing effectiveness in vitro and in vivo. One mAb in particular (mAb12) recognized an epitope within the Sb site and demonstrated outstanding neutralizing effectiveness.


Assuntos
Anticorpos Monoclonais/imunologia , Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/imunologia , Vírus da Influenza A Subtipo H1N1/imunologia , Animais , Anticorpos Monoclonais/isolamento & purificação , Anticorpos Monoclonais/uso terapêutico , Anticorpos Neutralizantes/isolamento & purificação , Anticorpos Neutralizantes/uso terapêutico , Anticorpos Antivirais/isolamento & purificação , Anticorpos Antivirais/uso terapêutico , Modelos Animais de Doenças , Feminino , Glicoproteínas de Hemaglutininação de Vírus da Influenza/imunologia , Imunização Passiva , Camundongos , Camundongos Endogâmicos BALB C , Infecções por Orthomyxoviridae/prevenção & controle , Infecções por Orthomyxoviridae/terapia , Análise de Sobrevida
18.
BMC Infect Dis ; 14: 195, 2014 Apr 11.
Artigo em Inglês | MEDLINE | ID: mdl-24720840

RESUMO

BACKGROUND: Human cytomegalovirus (HCMV) is a ubiquitous pathogen that causes serious problems in immunocompromised or immunologically immature hosts. Vaccination is the preferred approach for prevention of HCMV infection, but so far no approved HCMV vaccine is available. In this study, we assessed the immunogenicity and protective immunity of a formalin-inactivated murine cytomegalovirus vaccine (FI-MCMV) in a mouse model in combination with adjuvants MF59, alum, or chitosan. METHODS: Specific-pathogen-free BALB/c mice aged 6-8 weeks were immunized twice, 3 weeks apart, with various doses of FI-MCMV (0.25 µg, 1 µg, 4 µg) with or without adjuvant. Mice were challenged with a lethal dose (5 × LD50) of a more virulent mouse salivary gland-passaged MCMV 3 weeks after the second immunization. The protective immunity of the vaccine was evaluated by determining the survival rates, residual spleen and salivary gland viral loads, body weight changes, and serum anti-MCMV IgG titers. RESULTS: Immunization with FI-MCMV vaccine induced a high level of specific antibody response. Antigen sparing was achieved by the addition of an adjuvant, which significantly enhanced the humoral response to vaccine antigens with a wide range of doses. The level of live virus detected in the spleen on day 5 and in the salivary glands on day 21 after the lethal challenge was significantly lower in adjuvant-treated groups than in controls. Survival rates in adjuvant-treated groups also increased significantly. Furthermore, these protective immune responses were sustained for at least 6 months following immunization. CONCLUSIONS: These results show that inactivated MCMV vaccine is effective, and that the adjuvanted FI-MCMV vaccine provides more effective and longer-term protection than the adjuvant-free vaccine.


Assuntos
Adjuvantes Imunológicos , Vacinas contra Citomegalovirus/imunologia , Infecções por Herpesviridae/prevenção & controle , Muromegalovirus/imunologia , Animais , Feminino , Infecções por Herpesviridae/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Organismos Livres de Patógenos Específicos , Vacinas de Produtos Inativados/imunologia , Carga Viral
19.
Virol Sin ; 39(1): 56-70, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-37967718

RESUMO

Avian H9N2 viruses have wide host range among the influenza A viruses. However, knowledge of H9N2 mammalian adaptation is limited. To explore the molecular basis of the adaptation to mammals, we performed serial lung passaging of the H9N2 strain A/chicken/Hunan/8.27 YYGK3W3-OC/2018 (3W3) in mice and identified six mutations in the hemagglutinin (HA) and polymerase acidic (PA) proteins. Mutations L226Q, T511I, and A528V of HA were responsible for enhanced pathogenicity and viral replication in mice; notably, HA-L226Q was the key determinant. Mutations T97I, I545V, and S594G of PA contributed to enhanced polymerase activity in mammalian cells and increased viral replication levels in vitro and in vivo. PA-T97I increased viral polymerase activity by accelerating the viral polymerase complex assembly. Our findings revealed that the viral replication was affected by the presence of PA-97I and/or PA-545V in combination with a triple-point HA mutation. Furthermore, the double- and triple-point PA mutations demonstrated antagonistic effect on viral replication when combined with HA-226Q. Notably, any combination of PA mutations, along with double-point HA mutations, resulted in antagonistic effect on viral replication. We also observed antagonism in viral replication between PA-545V and PA-97I, as well as between HA-528V and PA-545V. Our findings demonstrated that several antagonistic mutations in HA and PA proteins affect viral replication, which may contribute to the H9N2 virus adaptation to mice and mammalian cells. These findings can potentially contribute to the monitoring of H9N2 field strains for assessing their potential risk in mammals.


Assuntos
Vírus da Influenza A Subtipo H9N2 , Influenza Aviária , Infecções por Orthomyxoviridae , Animais , Camundongos , Vírus da Influenza A Subtipo H9N2/genética , Hemaglutininas , Proteínas Virais/genética , Proteínas Virais/metabolismo , Mutação , Replicação Viral/genética , Nucleotidiltransferases , Galinhas , Mamíferos/metabolismo
20.
Adv Sci (Weinh) ; : e2309840, 2024 May 20.
Artigo em Inglês | MEDLINE | ID: mdl-38769664

RESUMO

The RNA modification, 5-methylcytosine (m5C), has recently gained prominence as a pivotal post-transcriptional regulator of gene expression, intricately intertwined with various tumorigenic processes. However, the exact mechanisms governing m5C modifications during the onset and progression of colorectal cancer (CRC) remain unclear. Here, it is determined that the m5C methyltransferase NSUN2 exhibits significantly elevated expression and exerts an oncogenic function in CRC. Mechanistically, NSUN2 and YBX1 are identified as the "writer" and "reader" of ENO1, culminating in the reprogramming of the glucose metabolism and increased production of lactic acid in an m5C-dependent manner. The accumulation of lactic acid derived from CRC cells, in turn, activates the transcription of NSUN2 through histone H3K18 lactylation (H3K18la), and induces the lactylation of NSUN2 at the Lys356 residue (K356), which is crucial for capturing target RNAs. Together, these findings reveal an intriguing positive feedback loop involving the NSUN2/YBX1/m5C-ENO1 signaling axis, thereby bridging the connection between metabolic reprogramming and epigenetic remodeling, which may shed light on the therapeutic potential of combining an NSUN2 inhibitor with immunotherapy for CRC.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA