Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Lancet Reg Health West Pac ; 34: 100707, 2023 May.
Artigo em Inglês | MEDLINE | ID: mdl-37283962

RESUMO

Background: Despite the success in decreasing varicella-related disease burden, live-attenuated Oka vaccine strain of varicella-zoster virus (vOka) remains neuro-virulence and may establish latency and reactivate, raising safety concerns. Here we aimed to evaluate the safety and immunogenicity of a skin- and neuro-attenuated varicella vaccine candidate (v7D). Methods: This is a randomized, double-blind, placebo-controlled, dose-escalation and age de-escalation phase 1 clinical trial conducted in Liuzhou, China (ChiCTR1900022284). Eligible healthy participants aged 1-49 years, with no history of varicella vaccination and had no history of varicella or herpes zoster were sequentially enrolled and allocated to subcutaneously receive one of the three doses (3.3, 3.9, and 4.2 lg PFU) of v7D, vOka or placebo in a dose-escalation and age de-escalation manner. The primary outcome was safety, assessed by adverse events/reactions within 42 days after vaccination and serious adverse events (SAEs) throughout six months after vaccination. The secondary outcome was immunogenicity, assessed by the VZV IgG antibodies measured with fluorescent antibody to membrane antigen (FAMA) assay. Findings: Between April 2019 and March 2020, totally 224 participants were enrolled. Within 42 days post-vaccination, the incidences of adverse reactions were 37.5%-38.7% in the three doses of v7D groups which were similar to that of the vOka (37.5%) and placebo (34.4%) groups. No SAE has been judged as causally related to vaccination. At 42 days post-vaccination, 100% of children aged 1-12 years in the per-protocol set of immunogenicity cohort of the v7D groups became seropositive. Meanwhile, in the intent-to-treat set of immunogenicity cohort of subjects aged 1-49 years, the geometric mean increases of the three groups of v7D vaccine were 3.8, 5.8 and 3.2, respectively, which were similar to that of the vOka vaccine group (4.4) and significantly higher than that of the placebo group (1.3). Interpretation: The candidate v7D vaccine has been preliminarily shown to be well-tolerated and immunogenic in humans. The data warrant further evaluation of the safety advantage and efficacy of v7D as a varicella vaccine. Funding: The National Natural Science Foundation of China, CAMS Innovation Fund for Medical Sciences, and Beijing Wantai CO., LTD.

2.
Microorganisms ; 11(2)2023 Feb 17.
Artigo em Inglês | MEDLINE | ID: mdl-36838484

RESUMO

Infection with the varicella-zoster virus (VZV) causes chickenpox and shingles, which lead to significant morbidity and mortality globally. The detection of serum VZV-specific antibodies is important for the clinical diagnosis and sero-epidemiological research of VZV infection, and for assessing the effect of VZV vaccine immunization. Over recent decades, a variety of methods for VZV antibody detection have been developed. This review summarizes and compares the current methods for detecting VZV antibodies, and discussed future directions for this field.

3.
Viruses ; 14(5)2022 04 20.
Artigo em Inglês | MEDLINE | ID: mdl-35632591

RESUMO

Primary varicella-zoster virus (VZV) infection causes varicella, which remains a prominent public health concern in children. Current varicella vaccines adopt the live-attenuated Oka strain, vOka, which retains the ability to infect neurons, establish latency and reactivate, leading to vaccine-associated zoster in some vaccinees. Therefore, it is necessary to develop a safer next-generation varicella vaccine to help reduce vaccine hesitancy. This paper reviews the discovery and identification of the skin- and neuro-tropic factor, the open reading frame 7 (ORF7) of VZV, as well as the development of a skin- and neuro-attenuated live varicella vaccine comprising an ORF7-deficient mutant, v7D. This work could provide insights into the research of novel virus vaccines based on functional genomics and reverse genetics.


Assuntos
Varicela , Herpes Zoster , Varicela/prevenção & controle , Vacina contra Varicela/efeitos adversos , Criança , Herpesvirus Humano 3/genética , Humanos , Vacinas Atenuadas
4.
Nat Commun ; 13(1): 824, 2022 02 11.
Artigo em Inglês | MEDLINE | ID: mdl-35149692

RESUMO

Varicella caused by the primary infection of varicella-zoster virus (VZV) exerts a considerable disease burden globally. Current varicella vaccines consisting of the live-attenuated vOka strain of VZV are generally safe and effective. However, vOka retains full neurovirulence and can establish latency and reactivate to cause herpes zoster in vaccine recipients, raising safety concerns. Here, we rationally design a live-attenuated varicella vaccine candidate, v7D. This virus replicates like wild-type virus in MRC-5 fibroblasts and human PBMCs, the carrier for VZV dissemination, but is severely impaired for infection of human skin and neuronal cells. Meanwhile, v7D shows immunogenicity comparable to vOka both in vitro and in multiple small animal species. Finally, v7D is proven well-tolerated and immunogenic in nonhuman primates. Our preclinical data suggest that v7D is a promising candidate as a safer live varicella vaccine with reduced risk of vaccine-related complications, and could inform the design of other herpes virus vaccines.


Assuntos
Vacina contra Varicela/imunologia , Varicela/imunologia , Pele/imunologia , Vacinas Atenuadas/imunologia , Animais , Linhagem Celular , Varicela/prevenção & controle , Feminino , Fibroblastos , Cobaias , Herpes Zoster/virologia , Herpesvirus Humano 3 , Humanos , Imunogenicidade da Vacina , Pulmão , Masculino , Camundongos , Neurônios/patologia , Coelhos , Ratos , Pele/patologia , Vacinação , Vacinas Virais
5.
Theranostics ; 11(13): 6607-6615, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33995679

RESUMO

SARS-CoV-2 infection, which is responsible for the current COVID-19 pandemic, can cause life-threatening pneumonia, respiratory failure and even death. Characterizing SARS-CoV-2 pathogenesis in primary human target cells and tissues is crucial for developing vaccines and therapeutics. However, given the limited access to clinical samples from COVID-19 patients, there is a pressing need for in vitro/in vivo models to investigate authentic SARS-CoV-2 infection in primary human lung cells or tissues with mature structures. The present study was designed to evaluate a humanized mouse model carrying human lung xenografts for SARS-CoV-2 infection in vivo. Methods: Human fetal lung tissue surgically grafted under the dorsal skin of SCID mice were assessed for growth and development after 8 weeks. Following SARS-CoV-2 inoculation into the differentiated lung xenografts, viral replication, cell-type tropism and histopathology of SARS-CoV-2 infection, and local cytokine/chemokine expression were determined over a 6-day period. The effect of IFN-α treatment against SARS-CoV-2 infection was tested in the lung xenografts. Results: Human lung xenografts expanded and developed mature structures closely resembling normal human lung. SARS-CoV-2 replicated and spread efficiently in the lung xenografts with the epithelial cells as the main target, caused severe lung damage, and induced a robust pro-inflammatory response. IFN-α treatment effectively inhibited SARS-CoV-2 replication in the lung xenografts. Conclusions: These data support the human lung xenograft mouse model as a useful and biological relevant tool that should facilitate studies on the pathogenesis of SARS-CoV-2 lung infection and the evaluation of potential antiviral therapies.


Assuntos
COVID-19/imunologia , Modelos Animais de Doenças , Pulmão/patologia , Mucosa Respiratória/citologia , SARS-CoV-2/imunologia , Feto Abortado , Animais , COVID-19/patologia , COVID-19/virologia , Células Cultivadas , Células Epiteliais/virologia , Xenoenxertos , Humanos , Pulmão/imunologia , Pulmão/virologia , Transplante de Pulmão , Masculino , Camundongos , Camundongos SCID , Cultura Primária de Células , SARS-CoV-2/patogenicidade , Replicação Viral
6.
Nat Microbiol ; 5(12): 1542-1552, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-32895526

RESUMO

Varicella-zoster virus (VZV) is a medically important human herpesvirus that causes chickenpox and shingles, but its cell-associated nature has hindered structure studies. Here we report the cryo-electron microscopy structures of purified VZV A-capsid and C-capsid, as well as of the DNA-containing capsid inside the virion. Atomic models derived from these structures show that, despite enclosing a genome that is substantially smaller than those of other human herpesviruses, VZV has a similarly sized capsid, consisting of 955 major capsid protein (MCP), 900 small capsid protein (SCP), 640 triplex dimer (Tri2) and 320 triplex monomer (Tri1) subunits. The VZV capsid has high thermal stability, although with relatively fewer intra- and inter-capsid protein interactions and less stably associated tegument proteins compared with other human herpesviruses. Analysis with antibodies targeting the N and C termini of the VZV SCP indicates that the hexon-capping SCP-the largest among human herpesviruses-uses its N-terminal half to bridge hexon MCP subunits and possesses a C-terminal flexible half emanating from the inner rim of the upper hexon channel into the tegument layer. Correlation of these structural features and functional observations provide insights into VZV assembly and pathogenesis and should help efforts to engineer gene delivery and anticancer vectors based on the currently available VZV vaccine.


Assuntos
Capsídeo/ultraestrutura , Herpesvirus Humano 3/ultraestrutura , Infecção pelo Vírus da Varicela-Zoster/virologia , Capsídeo/metabolismo , Proteínas do Capsídeo/química , Proteínas do Capsídeo/metabolismo , Microscopia Crioeletrônica , Herpesvirus Humano 3/química , Herpesvirus Humano 3/metabolismo , Humanos , Modelos Moleculares , Domínios Proteicos , Vírion/metabolismo , Vírion/ultraestrutura
7.
Virol J ; 15(1): 65, 2018 04 10.
Artigo em Inglês | MEDLINE | ID: mdl-29631630

RESUMO

BACKGROUND: Endometrial cancer (EC) is one of the most common gynecological malignancies globally. Although progress has been made in surgical and other adjuvant therapies, there is still a great need to develop new approaches to further reduce the incidence and mortality of EC. Oncolytic virotherapy offers a novel promising option of cancer treatment and has demonstrated good efficacy in preclinical models and clinical trials. However, only few oncolytic viruses have been tested for EC treatment. In this study, the potential of an oncolytic coxsackievirus B3 (CV-B3) strain 2035A (CV-B3/2035A) was investigated as a novel biotherapeutic agent against EC. METHODS: Human EC cell lines (Ishikawa, HEC-1-A and HEC-1-B) were infected with CV-B3/2035A, and viral replication and cytotoxic effects were evaluated in vitro. CV-B3/2035A-induced oncolysis was also investigated in nude mice bearing EC xenografts in vivo and in patient-derived EC samples ex vivo. RESULTS: Human EC cell lines expressing different levels of CAR and DAF were all susceptible to infection by CV-B3/2035A and supported efficient viral replication in vitro. In the EC xenograft/nude mouse model, both intratumoral and intravenous administrations of CV-B3-2035A exerted significant therapeutic effects against pre-established EC tumors without causing significant treatment-related toxicity and mortality in nude mice. Moreover, CV-B3/2035A treatment resulted in decreased viability of patient-derived EC samples ex vivo. CONCLUSIONS: CV-B3/2035A showed oncolytic activity in human EC cell lines both in vitro and in vivo as well as in patient-derived EC samples ex vivo and thus could be used as an alternative virotherapy agent for the treatment of EC.


Assuntos
Neoplasias do Endométrio/terapia , Neoplasias do Endométrio/virologia , Enterovirus/fisiologia , Terapia Viral Oncolítica , Vírus Oncolíticos/fisiologia , Animais , Linhagem Celular Tumoral , Neoplasias do Endométrio/patologia , Enterovirus/isolamento & purificação , Feminino , Humanos , Camundongos Endogâmicos BALB C , Camundongos Nus , Transplante de Neoplasias , Vírus Oncolíticos/isolamento & purificação , Receptores Virais/metabolismo , Replicação Viral , Ensaios Antitumorais Modelo de Xenoenxerto
8.
Virol Sin ; 32(5): 387-395, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-29116592

RESUMO

Varicella-zoster virus (VZV) is a neurotropic alphaherpesvirus that causes chickenpox and shingles. ORF7 is an important virulence determinant of VZV in both human skin and nerve tissues, however, its specific function and involved molecular mechanism in VZV pathogenesis remain largely elusive. Previous yeast two-hybrid studies on intraviral protein-protein interaction network in herpesviruses have revealed that VZV ORF7 may interact with ORF53, which is a virtually unstudied but essential viral protein. The aim of this study is to identify and characterize VZV ORF53, and to investigate its relationship with ORF7. For this purpose, we prepared monoclonal antibodies against ORF53 and, for the first time, characterized it as a ~40 kDa viral protein predominantly localizing to the trans-Golgi network of the infected host cell. Next, we further confirmed the interaction between ORF7 and ORF53 by co-immunoprecipitation and co-localization studies in both plasmid-transfected and VZV-infected cells. Moreover, interestingly, we found that ORF53 lost its trans-Golgi network localization and became dispersed in the cytoplasm of host cells infected with an ORF7-deleted recombinant VZV, and thus ORF7 seems to play a role in normal subcellular localization of ORF53. Collectively, these results suggested that ORF7 and ORF53 may function as a complex during infection, which may be implicated in VZV pathogenesis.


Assuntos
Herpesvirus Humano 3/metabolismo , Proteínas Virais/metabolismo , Rede trans-Golgi/metabolismo , Imunoprecipitação , Ligação Proteica
9.
Virology ; 512: 34-38, 2017 12.
Artigo em Inglês | MEDLINE | ID: mdl-28910710

RESUMO

Syncytia formation has been considered important for cell-to-cell spread and pathogenesis of many viruses. As a syncytium forms, individual nuclei often congregate together, allowing close contact of nuclear membranes and possibly fusion to occur. However, there is currently no reported evidence of nuclear membrane fusion between adjacent nuclei in wild-type virus-induced syncytia. Varicella-zoster virus (VZV) is one typical syncytia-inducing virus that causes chickenpox and shingles in humans. Here, we report, for the first time, an interesting observation of apparent fusion of the outer nuclear membranes from juxtaposed nuclei that comprise VZV syncytia both in ARPE-19 human epithelial cells in vitro and in human skin xenografts in the SCID-hu mouse model in vivo. This work reveals a novel aspect of VZV-related cytopathic effect in the context of multinucleated syncytia. Additionally, the information provided by this study could be helpful for future studies on interactions of viruses with host cell nuclei.


Assuntos
Células Epiteliais/patologia , Células Epiteliais/virologia , Células Gigantes/virologia , Herpesvirus Humano 3/fisiologia , Membrana Nuclear/patologia , Fusão Celular , Linhagem Celular , Humanos , Membrana Nuclear/virologia , Pele/citologia
10.
Antiviral Res ; 146: 45-53, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-28827121

RESUMO

Varicella pneumonia is one of the most serious, potentially life-threatening complications of primary varicella-zoster virus (VZV) infection in adults and immunocompromised individuals. However, studies on the lung pathogenesis of VZV infection as well as development and testing of antivirals have long been hindered by limited access to clinical samples and a lack of suitable animal models. In this study, we report for the first time the use of human lung xenografts in SCID mice for investigating VZV infection. Human fetal lung tissues grafted under the kidney capsule of SCID mice rapidly grew and developed mature structures closely resembling normal human lung. Following infection, VZV replicated and spread efficiently in human lung xenografts, where the virus targeted both alveolar epithelial and mesenchymal cells, and resulted in formation of large viral lesions. VZV particles were readily detected in the nuclei and cytoplasm of infected lung cells by electron microscopy. Additionally, VZV infection resulted in a robust pro-inflammatory cytokine response in human lung xenografts. In conclusion, infecting human lung xenografts in SCID mice provides a useful, biological relevant tool for future mechanistic studies on VZV lung pathogenesis, and may potentially facilitate the evaluation of new antiviral therapies for VZV lung infection.


Assuntos
Modelos Animais de Doenças , Herpesvirus Humano 3/fisiologia , Pulmão/virologia , Infecção pelo Vírus da Varicela-Zoster/virologia , Animais , Citocinas/biossíntese , Citocinas/imunologia , Herpesvirus Humano 3/imunologia , Herpesvirus Humano 3/isolamento & purificação , Xenoenxertos , Humanos , Pulmão/fisiopatologia , Transplante de Pulmão , Camundongos , Camundongos SCID , Transplante Heterólogo , Infecção pelo Vírus da Varicela-Zoster/imunologia , Replicação Viral
11.
Virology ; 491: 96-105, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26891237

RESUMO

Varicella-zoster virus (VZV) is the causative agent of both chickenpox (varicella) and shingles (zoster). VZV survives host defenses, even with an intact immune system, and disseminates in the host before causing disease. To date, several diverse immunomodulatory strategies used by VZV to undermine host immunity have been identified; however, few studies have addressed the complement evasion strategies used by this virus. Here, we show that expression of CD59, which is a key member of host regulators of complement activation (RCA), is significantly upregulated in response to VZV infection in human T cells and dorsal root ganglia (DRG) but not in human skin xenografts in SCID-hu mice in vivo. This is the first report demonstrating that VZV infection upregulates host CD59 expression in a tissue-specific manner in vivo, which may aid VZV in complement evasion and pathogenesis.


Assuntos
Antígenos CD59/genética , Varicela/genética , Herpesvirus Humano 3/fisiologia , Animais , Antígenos CD59/metabolismo , Varicela/metabolismo , Varicela/patologia , Varicela/virologia , Modelos Animais de Doenças , Gânglios Espinais/metabolismo , Gânglios Espinais/patologia , Gânglios Espinais/virologia , Herpesvirus Humano 3/genética , Interações Hospedeiro-Patógeno , Humanos , Fígado/metabolismo , Fígado/patologia , Fígado/virologia , Masculino , Camundongos , Camundongos SCID , Timo/metabolismo , Timo/patologia , Timo/virologia
12.
Dev Comp Immunol ; 34(9): 969-76, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20444427

RESUMO

Interferon-gamma-inducible lysosomal thiol reductase (GILT) is an important thiol reductase, involved in class, MHC-restricted antigen processing by catalyzing disulfide bond reduction in mammals. Herein, we describe the identification and characterization of pearl oyster Pinctada fucata GILT (designated as poGILT). The poGILT cDNA was 1273bp long and consisted of a 5'-untranslated region (UTR) of 24bp, a 3'-UTR of 484bp with two cytokine RNA instability motifs (ATTTA), and an open reading frame (ORF) of 765bp encoding a polypeptide of 254 amino acids with an estimated molecular mass of 28.9kDa and a theoretical isoelectric point of 7.4. The N-terminus of the poGILT was found to have a putative signal peptide with a cleavage site amino acid position at 19-20. SMART analysis showed that the poGILT contained a GILT active-site C(69)PDC(72) motif and a GILT signature motif C(115)QHGKEECIGNLIETC(130). Homology analysis of the deduced amino acid sequence of the poGILT with other known GILT sequences by MatGAT software revealed that the poGILT shared 42.9-67.3% similarity and 22.9-49.8% identity to the other known GILT sequences. The expression level of poGILT mRNA was higher in digestive gland, moderate in adductor muscle, gills, gonad, intestine and mantle, and lower in hemocytes. The poGILT mRNA expression was significantly up-regulated in gill and digestive gland after LPS or V. alginolyticus stimulation, respectively. These results suggested that the poGILT was a constitutively expressed acute-phase protein, the expression of which can be enhanced after LPS or V. algrinolyticus stimulation, perhaps involved in the innate immune response of pearl oyster.


Assuntos
Interferon gama/metabolismo , Oxirredutases atuantes sobre Doadores de Grupo Enxofre/metabolismo , Pinctada/imunologia , Vibrioses/imunologia , Vibrio alginolyticus/imunologia , Motivos de Aminoácidos/genética , Sequência de Aminoácidos , Animais , Clonagem Molecular , Perfilação da Expressão Gênica , Imunidade Inata , Interferon gama/imunologia , Lisossomos/metabolismo , Dados de Sequência Molecular , Oxirredutases atuantes sobre Doadores de Grupo Enxofre/genética , Oxirredutases atuantes sobre Doadores de Grupo Enxofre/imunologia , Filogenia , Pinctada/microbiologia , Vibrioses/enzimologia , Vibrioses/genética , Vibrio alginolyticus/patogenicidade
13.
Fish Shellfish Immunol ; 26(4): 662-8, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19254766

RESUMO

The clip-domain serine proteases (SPs) are the essential components of extracellular signaling cascade in various biological processes, especially in embryonic development and the innate immune responses of invertebrate. Herein, we described the isolation and characterization of pearl oyster Pinctada fucata clip-domain SP gene (designated as poSP). The poSP cDNA was 1080 bp long and consisted of a 5'-untranslated region (UTR) of 13 bp, a 3'-UTR of 68 bp with a polyadenylation signal (AATAAA) at 22 nucleotides upstream of the poly(A) tail, and an open reading frame (ORF) of 999 bp encoding a polypeptide of 332 amino acids with an estimated molecular mass of 36.5 kDa and a theoretical isoelectric point of 7.3. A clip-domain and a trypsin-like serine protease domain were identified in the poSP using SMART analysis. Homology analysis of the deduced amino acid sequence of the poSP with other known SP sequences by MatGAT software revealed that the poSP shared 47.0-68.4% similarity to the other known SP sequences. The poSP mRNA was expressed in haemocytes, gonad, digestive gland and mantle, but not expressed in adductor muscle and gill. The poSP mRNA was up-regulated and increased nearly double-fold after LPS or Vibrio alginolyticus stimulation, respectively. These results suggested that the poSP was an inducible acute-phase protein that perhaps involved in the innate immune response of pearl oyster.


Assuntos
Regulação Enzimológica da Expressão Gênica , Pinctada/enzimologia , Pinctada/genética , Serina Endopeptidases/genética , Serina Endopeptidases/metabolismo , Adjuvantes Imunológicos/farmacologia , Sequência de Aminoácidos , Animais , Sequência de Bases , Clonagem Molecular , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Lipopolissacarídeos/farmacologia , Dados de Sequência Molecular , Filogenia , Pinctada/classificação , Pinctada/microbiologia , Homologia de Sequência de Aminoácidos , Fatores de Tempo , Vibrio/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...