Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Signal Transduct Target Ther ; 9(1): 118, 2024 May 03.
Artículo en Inglés | MEDLINE | ID: mdl-38702343

RESUMEN

Antitumor therapies based on adoptively transferred T cells or oncolytic viruses have made significant progress in recent years, but the limited efficiency of their infiltration into solid tumors makes it difficult to achieve desired antitumor effects when used alone. In this study, an oncolytic virus (rVSV-LCMVG) that is not prone to induce virus-neutralizing antibodies was designed and combined with adoptively transferred T cells. By transforming the immunosuppressive tumor microenvironment into an immunosensitive one, in B16 tumor-bearing mice, combination therapy showed superior antitumor effects than monotherapy. This occurred whether the OV was administered intratumorally or intravenously. Combination therapy significantly increased cytokine and chemokine levels within tumors and recruited CD8+ T cells to the TME to trigger antitumor immune responses. Pretreatment with adoptively transferred T cells and subsequent oncolytic virotherapy sensitizes refractory tumors by boosting T-cell recruitment, down-regulating the expression of PD-1, and restoring effector T-cell function. To offer a combination therapy with greater translational value, mRNA vaccines were introduced to induce tumor-specific T cells instead of adoptively transferred T cells. The combination of OVs and mRNA vaccine also displays a significant reduction in tumor burden and prolonged survival. This study proposed a rational combination therapy of OVs with adoptive T-cell transfer or mRNA vaccines encoding tumor-associated antigens, in terms of synergistic efficacy and mechanism.


Asunto(s)
Viroterapia Oncolítica , Virus Oncolíticos , Animales , Ratones , Virus Oncolíticos/genética , Virus Oncolíticos/inmunología , Viroterapia Oncolítica/métodos , Terapia Combinada , Vacunas de ARNm/inmunología , Melanoma Experimental/terapia , Melanoma Experimental/inmunología , Microambiente Tumoral/inmunología , Linfocitos T CD8-positivos/inmunología , Linfocitos T/inmunología , Humanos , Línea Celular Tumoral , Vacunas contra el Cáncer/inmunología , Vacunas contra el Cáncer/genética , Vacunas contra el Cáncer/administración & dosificación
2.
J Hepatol ; 80(5): 714-729, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38336348

RESUMEN

BACKGROUND & AIMS: Mechanisms behind the impaired response of antigen-specific B cells to therapeutic vaccination in chronic hepatitis B virus (HBV) infection remain unclear. The development of vaccines or strategies to overcome this obstacle is vital for advancing the management of chronic hepatitis B. METHODS: A mouse model, denominated as E6F6-B, was engineered to feature a knock-in of a B-cell receptor (BCR) that specifically recognizes HBsAg. This model served as a valuable tool for investigating the temporal and spatial dynamics of humoral responses following therapeutic vaccination under continuous antigen exposure. Using a suite of immunological techniques, we elucidated the differentiation trajectory of HBsAg-specific B cells post-therapeutic vaccination in HBV carrier mice. RESULTS: Utilizing the E6F6-B transfer model, we observed a marked decline in antibody-secreting cells 2 weeks after vaccination. A dysfunctional and atypical pre-plasma cell population (BLIMP-1+ IRF4+ CD40- CD138- BCMA-) emerged, manifested by sustained BCR signaling. By deploying an antibody to purge persistent HBsAg, we effectively prompted the therapeutic vaccine to provoke conventional plasma cell differentiation. This resulted in an enhanced anti-HBs antibody response and facilitated HBsAg clearance. CONCLUSIONS: Sustained high levels of HBsAg limit the ability of therapeutic hepatitis B vaccines to induce the canonical plasma cell differentiation necessary for anti-HBs antibody production. Employing a strategy combining antibodies with vaccines can surmount this altered humoral response associated with atypical pre-plasma cells, leading to improved therapeutic efficacy in HBV carrier mice. IMPACT AND IMPLICATIONS: Therapeutic vaccines aimed at combatting HBV encounter suboptimal humoral responses in clinical settings, and the mechanisms impeding their effectiveness have remained obscure. Our research, utilizing the innovative E6F6-B mouse transfer model, reveals that the persistence of HBsAg can lead to the emergence of an atypical pre-plasma cell population, which proves to be relevant to the potency of therapeutic HBV vaccines. Targeting the aberrant differentiation process of these atypical pre-plasma cells stands out as a critical strategy to amplify the humoral response elicited by HBV therapeutic vaccines in carrier mouse models. This discovery suggests a compelling avenue for further study in the context of human chronic hepatitis B. Encouragingly, our findings indicate that synergistic therapy combining HBV-specific antibodies with vaccines offers a promising approach that could significantly advance the pursuit of a functional cure for HBV.


Asunto(s)
Hepatitis B Crónica , Hepatitis B , Ratones , Humanos , Animales , Antígenos de Superficie de la Hepatitis B , Virus de la Hepatitis B , Vacunas contra Hepatitis B/uso terapéutico , Anticuerpos contra la Hepatitis B , Diferenciación Celular , Hepatitis B/prevención & control , Hepatitis B/tratamiento farmacológico
3.
J Control Release ; 365: 369-383, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-37972764

RESUMEN

Micro/Nano-scale particles are widely used as vaccine adjuvants to enhance immune response and improve antigen stability. While aluminum salt is one of the most common adjuvants approved for human use, its immunostimulatory capacity is suboptimal. In this study, we modified risedronate, an immunostimulant and anti-osteoporotic drug, to create zinc salt particle-based risedronate (Zn-RS), also termed particulate risedronate. Compared to soluble risedronate, micronanoparticled Zn-RS adjuvant demonstrated increased recruitment of innate cells, enhanced antigen uptake locally, and a similar antigen depot effect as aluminum salt. Furthermore, Zn-RS adjuvant directly and quickly stimulated immune cells, accelerated the formulation of germinal centers in lymph nodes, and facilitated the rapid production of antibodies. Importantly, Zn-RS adjuvant exhibited superior performance in both young and aged mice, effectively protecting against respiratory diseases such as SARS-CoV-2 challenge. Consequently, particulate risedronate showed great potential as an immune-enhancing vaccine adjuvant, particularly beneficial for vaccines targeting the susceptible elderly.


Asunto(s)
Adyuvantes de Vacunas , Vacunas , Animales , Ratones , Humanos , Anciano , Ácido Risedrónico/uso terapéutico , Aluminio , Adyuvantes Inmunológicos , Inmunización , Antígenos
4.
Protein Cell ; 15(2): 121-134, 2024 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-37470320

RESUMEN

Continual evolution of the severe acute respiratory syndrome coronavirus (SARS-CoV-2) virus has allowed for its gradual evasion of neutralizing antibodies (nAbs) produced in response to natural infection or vaccination. The rapid nature of these changes has incited a need for the development of superior broad nAbs (bnAbs) and/or the rational design of an antibody cocktail that can protect against the mutated virus strain. Here, we report two angiotensin-converting enzyme 2 competing nAbs-8H12 and 3E2-with synergistic neutralization but evaded by some Omicron subvariants. Cryo-electron microscopy reveals the two nAbs synergistic neutralizing virus through a rigorous pairing permitted by rearrangement of the 472-489 loop in the receptor-binding domain to avoid steric clashing. Bispecific antibodies based on these two nAbs tremendously extend the neutralizing breadth and restore neutralization against recent variants including currently dominant XBB.1.5. Together, these findings expand our understanding of the potential strategies for the neutralization of SARS-CoV-2 variants toward the design of broad-acting antibody therapeutics and vaccines.


Asunto(s)
COVID-19 , SARS-CoV-2 , Humanos , Microscopía por Crioelectrón , Anticuerpos Neutralizantes , Anticuerpos Antivirales
5.
Nat Commun ; 14(1): 3609, 2023 06 17.
Artículo en Inglés | MEDLINE | ID: mdl-37330604

RESUMEN

Tailed bacteriophages (order, Caudovirales) account for the majority of all phages. However, the long flexible tail of siphophages hinders comprehensive investigation of the mechanism of viral gene delivery. Here, we report the atomic capsid and in-situ structures of the tail machine of the marine siphophage, vB_DshS-R4C (R4C), which infects Roseobacter. The R4C virion, comprising 12 distinct structural protein components, has a unique five-fold vertex of the icosahedral capsid that allows genome delivery. The specific position and interaction pattern of the tail tube proteins determine the atypical long rigid tail of R4C, and further provide negative charge distribution within the tail tube. A ratchet mechanism assists in DNA transmission, which is initiated by an absorption device that structurally resembles the phage-like particle, RcGTA. Overall, these results provide in-depth knowledge into the intact structure and underlining DNA delivery mechanism for the ecologically important siphophages.


Asunto(s)
Bacteriófagos , Caudovirales , Genoma Viral , Bacteriófagos/genética , Bacteriófagos/química , Genes Virales , Caudovirales/genética , Proteínas de la Cápside/genética , ADN , ADN Viral/genética
6.
Front Microbiol ; 13: 983656, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36212859

RESUMEN

Human Rhinoviruses (RVs) are dominant pathogens causing a wide range of respiratory tract diseases, posing a huge threat to public health worldwide. Viruses belonging to the RV-C species are more likely to cause severe illnesses and are strongly associated with asthma onset or exacerbations than RV-A or RV-B. Rapid and sensitive detection of neutralizing antibodies (NAbs) against RV-C can promote the development of vaccines and antiviral drugs and help in the diagnosis of viral infection. In this study, a rapid neutralization testing system for RV-C15, based on an enzyme-linked immunospot assay (Nt-ELISPOT) was developed. A monoclonal antibody (MAb), named 9F9, with high binding efficacy for RV-C15 conjugated to horseradish peroxidase (HRP), was used to detect RV-C15-infected cells at a concentration of 2 µg/ml. The optimal infectious dose of RV-C15 was set at 1 × 104 TCID50/well and the cells were fixed with 0.5% formaldehyde diluted in PBS after incubation for 20 h. Compared with the traditional cytopathic effect (CPE)-based neutralization assay (Nt-CPE), Nt-ELISPOT significantly shortened the detection period and showed good consistency with the detection of neutralizing titers of both sera and NAbs. Using Nt-ELISPOT, three anti-RV-C15 NAbs were obtained with IC50 values of 0.16, 0.27, and 11.8 µg/ml, respectively. Moreover, 64 human serum samples collected from a wide range of age groups were tested for NAb against RV-C15 by Nt-ELISPOT. The total seroprevalence was 48.4% (31/64) and the positive rate was lowest in the group under 6 years old. Thus, the Nt-ELISPOT established in this study can be used as a high-throughput and rapid neutralization assay for the screening of NAbs and for seroepidemiological investigation against RV-C15.

7.
Cell Host Microbe ; 30(9): 1279-1294.e6, 2022 09 14.
Artículo en Inglés | MEDLINE | ID: mdl-36002016

RESUMEN

Coxsackievirus B1 (CVB1) is an emerging pathogen associated with severe neonatal diseases including aseptic meningitis, myocarditis, and pancreatitis and also with the development of type 1 diabetes. We characterize the binding and therapeutic efficacies of three CVB1-specific neutralizing antibodies (nAbs) identified for their ability to inhibit host receptor engagement. High-resolution cryo-EM structures showed that these antibodies recognize different epitopes but with an overlapping region in the capsid VP2 protein and specifically the highly variable EF loop. Moreover, they perturb capsid-receptor interactions by binding various viral particle forms. Antibody combinations achieve synergetic neutralization via a stepwise capsid transition and virion disruption, indicating dynamic changes in the virion in response to multiple nAbs targeting the receptor-binding site. Furthermore, this three-antibody cocktail protects against lethal challenge in neonatal mice and limits pancreatitis and viral replication in a non-obese diabetic mouse model. These results illustrate the utility of nAbs for rational design of therapeutics against picornaviruses such as CVB.


Asunto(s)
Anticuerpos Antivirales , Pancreatitis , Animales , Anticuerpos Neutralizantes , Cápside/química , Proteínas de la Cápside , Epítopos , Ratones
8.
Nat Commun ; 13(1): 1533, 2022 03 22.
Artículo en Inglés | MEDLINE | ID: mdl-35318331

RESUMEN

Pseudorabies virus (PRV) is a major etiological agent of swine infectious diseases and is responsible for significant economic losses in the swine industry. Recent data points to human viral encephalitis caused by PRV infection, suggesting that PRV may be able to overcome the species barrier to infect humans. To date, there is no available therapeutic for PRV infection. Here, we report the near-atomic structures of the PRV A-capsid and C-capsid, and illustrate the interaction that occurs between these subunits. We show that the C-capsid portal complex is decorated with capsid-associated tegument complexes. The PRV capsid structure is highly reminiscent of other α-herpesviruses, with some additional structural features of ß- and γ-herpesviruses. These results illustrate the structure of the PRV capsid and elucidate the underlying assembly mechanism at the molecular level. This knowledge may be useful for the development of oncolytic agents or specific therapeutics against this arm of the herpesvirus family.


Asunto(s)
Herpesvirus Suido 1 , Animales , Cápside , Proteínas de la Cápside , Porcinos , Estructuras Virales
9.
Nat Commun ; 13(1): 824, 2022 02 11.
Artículo en Inglés | MEDLINE | ID: mdl-35149692

RESUMEN

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.


Asunto(s)
Vacuna contra la Varicela/inmunología , Varicela/inmunología , Piel/inmunología , Vacunas Atenuadas/inmunología , Animales , Línea Celular , Varicela/prevención & control , Femenino , Fibroblastos , Cobayas , Herpes Zóster/virología , Herpesvirus Humano 3 , Humanos , Inmunogenicidad Vacunal , Pulmón , Masculino , Ratones , Neuronas/patología , Conejos , Ratas , Piel/patología , Vacunación , Vacunas Virales
10.
Cell Host Microbe ; 29(3): 448-462.e5, 2021 03 10.
Artículo en Inglés | MEDLINE | ID: mdl-33539764

RESUMEN

Enterovirus uncoating receptors bind at the surface depression ("canyon") that encircles each capsid vertex causing the release of a host-derived lipid called "pocket factor" that is buried in a hydrophobic pocket formed by the major viral capsid protein, VP1. Coxsackievirus and adenovirus receptor (CAR) is a universal uncoating receptor of group B coxsackieviruses (CVB). Here, we present five high-resolution cryoEM structures of CVB representing different stages of virus infection. Structural comparisons show that the CAR penetrates deeper into the canyon than other uncoating receptors, leading to a cascade of events: collapse of the VP1 hydrophobic pocket, high-efficiency release of the pocket factor and viral uncoating and genome release under neutral pH, as compared with low pH. Furthermore, we identified a potent therapeutic antibody that can neutralize viral infection by interfering with virion-CAR interactions, destabilizing the capsid and inducing virion disruption. Together, these results define the structural basis of CVB cell entry and antibody neutralization.


Asunto(s)
Microscopía por Crioelectrón , Enterovirus/metabolismo , Enterovirus/ultraestructura , Animales , Anticuerpos Neutralizantes , Cápside/metabolismo , Proteínas de la Cápside/ultraestructura , Enterovirus Humano B/metabolismo , Infecciones por Enterovirus/inmunología , Infecciones por Enterovirus/metabolismo , Infecciones por Enterovirus/virología , Femenino , Ratones , Ratones Endogámicos BALB C , Modelos Moleculares , Dominios y Motivos de Interacción de Proteínas , Receptores Virales , Virión/metabolismo , Virión/ultraestructura , Desencapsidación Viral
11.
Nat Microbiol ; 5(12): 1542-1552, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32895526

RESUMEN

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.


Asunto(s)
Cápside/ultraestructura , Herpesvirus Humano 3/ultraestructura , Infección por el Virus de la Varicela-Zóster/virología , Cápside/metabolismo , Proteínas de la Cápside/química , Proteínas de la Cápside/metabolismo , Microscopía por Crioelectrón , Herpesvirus Humano 3/química , Herpesvirus Humano 3/metabolismo , Humanos , Modelos Moleculares , Dominios Proteicos , Virión/metabolismo , Virión/ultraestructura
12.
Cell Host Microbe ; 27(2): 249-261.e5, 2020 02 12.
Artículo en Inglés | MEDLINE | ID: mdl-32027857

RESUMEN

Hand, foot, and mouth disease is a common childhood illness primarily caused by coxsackievirus A16 (CVA16), for which there are no current vaccines or treatments. We identify three CVA16-specific neutralizing monoclonal antibodies (nAbs) with therapeutic potential: 18A7, 14B10, and NA9D7. We present atomic structures of these nAbs bound to all three viral particle forms-the mature virion, A-particle, and empty particle-and show that each Fab can simultaneously occupy the mature virion. Additionally, 14B10 or NA9D7 provide 100% protection against lethal CVA16 infection in a neonatal mouse model. 18A7 binds to a non-conserved epitope present in all three particles, whereas 14B10 and NA9D7 recognize broad protective epitopes but only bind the mature virion. NA9D7 targets an immunodominant site, which may overlap the receptor-binding site. These findings indicate that CVA16 vaccines should be based on mature virions and that these antibodies could be used to discriminate optimal virion-based immunogens.


Asunto(s)
Anticuerpos Neutralizantes , Enterovirus Humano A/inmunología , Enfermedad de Boca, Mano y Pie/virología , Animales , Anticuerpos Neutralizantes/inmunología , Anticuerpos Neutralizantes/ultraestructura , Anticuerpos Antivirales/inmunología , Anticuerpos Antivirales/ultraestructura , Proteínas de la Cápside/inmunología , Línea Celular , Microscopía por Crioelectrón , Enterovirus/inmunología , Enterovirus/ultraestructura , Enterovirus Humano A/ultraestructura , Enfermedad de Boca, Mano y Pie/inmunología , Enfermedad de Boca, Mano y Pie/prevención & control , Humanos , Ratones , Vacunas Virales/inmunología , Virión/inmunología
13.
Sci Adv ; 4(9): eaat7459, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-30255146

RESUMEN

Coxsackievirus A10 (CVA10) recently emerged as a major pathogen of hand, foot, and mouth disease and herpangina in children worldwide, and lack of a vaccine or a cure against CVA10 infections has made therapeutic antibody identification a public health priority. By targeting a local isolate, CVA10-FJ-01, we obtained a potent antibody, 2G8, against all three capsid forms of CVA10. We show that 2G8 exhibited both 100% preventive and 100% therapeutic efficacy against CVA10 infection in mice. Comparisons of the near-atomic cryo-electron microscopy structures of the three forms of CVA10 capsid and their complexes with 2G8 Fab reveal that a single Fab binds a border region across the three capsid proteins (VP1 to VP3) and explain 2G8's remarkable cross-reactivities against all three capsid forms. The atomic structures of this first neutralizing antibody of CVA10 should inform strategies for designing vaccines and therapeutics against CVA10 infections.


Asunto(s)
Anticuerpos Neutralizantes/farmacología , Enterovirus Humano A/inmunología , Vacunas Virales/farmacología , Virión/química , Animales , Anticuerpos Neutralizantes/química , Anticuerpos Neutralizantes/inmunología , Cápside/química , Reacciones Cruzadas , Microscopía por Crioelectrón , Enfermedad de Boca, Mano y Pie/prevención & control , Humanos , Ratones Endogámicos BALB C , Pruebas de Neutralización , Vacunas Virales/inmunología , Virión/inmunología
14.
Emerg Microbes Infect ; 7(1): 144, 2018 Aug 10.
Artículo en Inglés | MEDLINE | ID: mdl-30097574

RESUMEN

The human liver chimeric mouse with primary human hepatocytes (PHHs) engraftment has been demonstrated to be a useful animal model to study hepatitis B virus (HBV) pathogenesis and evaluate anti-HBV drugs. However, the disadvantages of using PHHs include the inability for cellular expansion in vitro, limited donor availability, individual differences, and ethical issues, necessitating the development of alternatives. To obtain in vitro expandable hepatocytes, we optimized the hepatic differentiation procedure of the human liver progenitor cell line, HepaRG, using four functional small molecules (4SM) and enriched the precursor hepatocyte-like cells (HLCs). HepaRG cells of different hepatic differentiation states were engrafted to immunodeficient mice (FRGS) with weekly 4SM treatment. The HepaRG-engrafted mice were challenged with HBV and/or treated with several antivirals to evaluate their effects. We demonstrated that the 4SM treatment enhanced hepatic differentiation and promoted cell proliferation capacity both in vitro and in vivo. Mice engrafted with enriched HepaRG of prehepatic differentiation and treated with 4SM displayed approximately 10% liver chimerism at week 8 after engraftment and were maintained at this level for another 16 weeks. Therefore, we developed a HepaRG-based human liver chimeric mouse model: HepaRG-FRGS. Our experimental results showed that the liver chimerism of the mice was adequate to support chronic HBV infection for 24 weeks and to evaluate antivirals. We also demonstrated that HBV infection in HepaRG cells was dependent on their hepatic differentiation state and liver chimerism in vivo. Overall, HepaRG-FRGS mice provide a novel human liver chimeric mouse model to study chronic HBV infection and evaluate anti-HBV drugs.


Asunto(s)
Quimera/virología , Modelos Animales de Enfermedad , Virus de la Hepatitis B/fisiología , Hepatitis B Crónica/virología , Hepatocitos/virología , Hígado/virología , Animales , Diferenciación Celular , Línea Celular , Quimera/genética , Virus de la Hepatitis B/genética , Hepatitis B Crónica/fisiopatología , Hepatocitos/citología , Humanos , Hígado/citología , Ratones , Ratones Endogámicos BALB C , Ratones SCID , Replicación Viral
15.
Emerg Microbes Infect ; 7(1): 2, 2018 Jan 10.
Artículo en Inglés | MEDLINE | ID: mdl-29323107

RESUMEN

Human enteroviruses (EVs) are the most common causative agents infecting human, causing many harmful diseases, such as hand, foot, and mouth disease (HFMD), herpangina (HA), myocarditis, encephalitis, and aseptic meningitis. EV-related diseases pose a serious worldwide threat to public health. To gain comprehensive insight into the seroepidemiology of major prevalent EVs in humans, we firstly performed a serological survey for neutralizing antibodies (nAbs) against Enterovirus A71 (EV-A71), Coxsackie virus A16 (CV-A16), Coxsackie virus A6 (CV-A6), Coxsackie virus A10 (CV-A10), Coxsackie virus B3 (CV-B3), Coxsackie virus B5 (CV-B5), Echovirus 25 (ECHO25), and Echovirus 30 (ECHO30) among the healthy population in Xiamen City in 2016, using micro-neutralization assay. A total of 515 subjects aged 5 months to 83 years were recruited by stratified random sampling. Most major human EVs are widely circulated in Xiamen City and usually infect infants and children. The overall seroprevalence of these eight EVs were ranged from 14.4% to 42.7%, and most of them increased with age and subsequently reached a plateau. The co-existence of nAbs against various EVs are common among people ≥ 7 years of age, due to the alternate infections or co-infections with different serotypes of EVs, while most children were negative for nAb against EVs, especially those < 1 year of age. This is the first report detailing the seroepidemiology of eight prevalent EVs in the same population, which provides scientific data supporting further studies on the improvement of EV-related disease prevention and control.


Asunto(s)
Anticuerpos Neutralizantes/sangre , Infecciones por Enterovirus/inmunología , Enterovirus/inmunología , Vigilancia Inmunológica , Estudios Seroepidemiológicos , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Anticuerpos Neutralizantes/inmunología , Anticuerpos Neutralizantes/aislamiento & purificación , Niño , Preescolar , Enterovirus Humano A/inmunología , Enterovirus Humano B/inmunología , Femenino , Voluntarios Sanos , Humanos , Lactante , Masculino , Persona de Mediana Edad , Adulto Joven
16.
Virol Sin ; 32(5): 387-395, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-29116592

RESUMEN

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.


Asunto(s)
Herpesvirus Humano 3/metabolismo , Proteínas Virales/metabolismo , Red trans-Golgi/metabolismo , Inmunoprecipitación , Unión Proteica
17.
Sci Rep ; 5: 12973, 2015 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-26243660

RESUMEN

Human enterovirus 71 (EV71) and coxsackievirus A16 (CA16) are the major etiological agents of hand, foot and mouth disease (HFMD) and are often associated with neurological complications. Currently, several vaccine types are being developed for EV71 and CA16. In this study, we constructed a bivalent chimeric virus-like particle (VLP) presenting the VP1 (aa208-222) and VP2 (aa141-155) epitopes of EV71 using hepatitis B virus core protein (HBc) as a carrier, designated HBc-E1/2. Immunization with the chimeric VLPs HBc-E1/2 induced higher IgG titers and neutralization titers against EV71 and CA16 in vitro than immunization with only one epitope incorporated into HBc. Importantly, passive immunization with the recombinant HBc-E2 particles protected neonatal mice against lethal EV71 and CA16 infections. We demonstrate that anti-VP2 (aa141-155) sera bound authentic CA16 viral particles, whereas anti-VP1 (aa208-222) sera could not. Moreover, the anti-VP2 (aa141-155) antibodies inhibited the binding of human serum to virions, which demonstrated that the VP2 epitope is immunodominant between EV71 and CA16. These results illustrated that the chimeric VLP HBc-E1/2 is a promising candidate for a broad-spectrum HFMD vaccine, and also reveals mechanisms of protection by the neighboring linear epitopes of the VP1 GH and VP2 EF loops.


Asunto(s)
Proteínas de la Cápside/inmunología , Enterovirus Humano A/inmunología , Enfermedad de Boca, Mano y Pie/prevención & control , Vacunas Virales/inmunología , Secuencias de Aminoácidos/inmunología , Animales , Anticuerpos Neutralizantes/sangre , Anticuerpos Antivirales/sangre , Reacciones Cruzadas , Epítopos/inmunología , Femenino , Enfermedad de Boca, Mano y Pie/inmunología , Ratones Endogámicos BALB C , Ratas Wistar , Vacunación , Virión/inmunología
18.
Virus Res ; 205: 41-4, 2015 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-26004198

RESUMEN

Echovirus 25 (E-25) is a member of the enterovirus family and a common pathogen that induces hand, foot, and mouth disease (HFMD), meningitis, skin rash, and respiratory illnesses. In this study, we constructed and characterized an infectious full-length E-25 cDNA clone derived from the XM0297 strain, which was the first subgenotype D6 strain isolated in Xiamen, China. The 5'-Untranslated Regions (5'-UTR), P3 (3A-3B, 3D) and P3 (3C) regions of this E-25 (XM0297) strain were highly similar to EV-B77, E-16 and E-13, respectively. Our data demonstrate that the rescued E-25 viruses exhibited similar growth kinetics to the prototype virus strain XM0297. We observed the rescued viral particles using transmission electron microscope (TEM) and found them to possess an icosahedral structure, with a diameter of approximately 30 nm. The cross neutralization test demonstrated that the E-25 (XM0297) strain immune serum could not neutralize EV-A71, CV-A16 or CV-B3; likewise, the EV-A71 and CV-A16 immune serum could not neutralize E-25 (XM0297). The availability of this infectious clone will greatly enhance future virological investigations and possible vaccine development against E-25.


Asunto(s)
ADN Complementario/genética , ADN Viral/genética , Enterovirus Humano B/genética , Infecciones por Enterovirus/virología , Anticuerpos Antivirales/inmunología , China , ADN Complementario/metabolismo , ADN Viral/metabolismo , Enterovirus Humano B/clasificación , Enterovirus Humano B/inmunología , Enterovirus Humano B/fisiología , Enfermedad de Boca, Mano y Pie/inmunología , Enfermedad de Boca, Mano y Pie/virología , Humanos , Datos de Secuencia Molecular , Filogenia
19.
Theranostics ; 4(5): 498-513, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24669278

RESUMEN

Human enterovirus 71 (EV71) is the main causative agent of hand, foot, and mouth disease (HFMD) and is associated with several severe neurological complications in the Asia-Pacific region. Here, we evaluated that while passive transfer of neutralizing monoclonal antibody (nMAb) against the VP2 protein protect against lethal EV71 infection in BALB/c mice. Protective nMAb were mapped to residues 141-155 of VP2 by peptide ELISA. High-resolution structural analysis showed that the epitope is part of the VP2 EF loop, which is the "puff" region that forms the "southern rim" of the canyon. Moreover, a three-dimensional structural characterization for the puff region with prior neutralizing epitopes and receptor-binding sites that can serve to inform vaccine strategies. Interestingly, using hepatitis B virus core protein (HBc) as a carrier, we demonstrated that the cross-neutralizing EV71 antibodies were induced, and the VP2 epitope immunized mice serum also conferred 100% in vivo passive protection. The mechanism of in vivo protection conferred by VP2 nMAb is in part attributed to the in vitro neutralizing titer and ability to bind authentic viral particles. Importantly, the anti-VP2(aa141-155) antibodies could inhibit the binding of human serum to EV71 virions showed that the VP2 epitope is immunodominant. Collectively, our results suggest that a broad-spectrum vaccine strategy targeting the high-affinity epitope of VP2 EF loop may elicits effective immune responses against EV71 infection.


Asunto(s)
Enterovirus Humano A/inmunología , Epítopos de Linfocito B/inmunología , Enfermedad de Boca, Mano y Pie/prevención & control , Proteínas Estructurales Virales/inmunología , Vacunas Virales/inmunología , Animales , Anticuerpos Neutralizantes/sangre , Anticuerpos Neutralizantes/uso terapéutico , Anticuerpos Antivirales/sangre , Anticuerpos Antivirales/uso terapéutico , Antígenos Virales/genética , Antígenos Virales/inmunología , Enterovirus Humano A/genética , Epítopos de Linfocito B/genética , Femenino , Enfermedad de Boca, Mano y Pie/inmunología , Ratones , Ratones Endogámicos BALB C , Análisis de Supervivencia , Resultado del Tratamiento , Vacunas Sintéticas/administración & dosificación , Vacunas Sintéticas/genética , Vacunas Sintéticas/inmunología , Proteínas Estructurales Virales/genética , Vacunas Virales/administración & dosificación , Vacunas Virales/genética
20.
Biomed Environ Sci ; 22(6): 496-501, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20337223

RESUMEN

OBJECTIVE: To study the feasibility of using tetracysteine (TC) reporter in gene therapy. METHODS: Effects of TC reporter and conventional reporter genes encoding green fluorescence protein (GFP) and luciferase (Luc) on expression and function of the therapeutic gene MGMT(P140K) were compared. Cytotoxicity and drug resistance were studied by Western blot. TC reporter used in therapy was analyzed by flow cytometry (FCM). RESULTS: The TC reporter had no toxicity to cells and neither affected the expression or activity of therapeutic gene as compared to GFP and Luc. TC could be used in blood sample detection. CONCLUSION: TC is a new kind of reporter gene for lentiviral vector in future gene therapy.


Asunto(s)
Cisteína , Genes Reporteros , Linfocitos/metabolismo , Animales , Células CHO , Cricetinae , Cricetulus , Cisteína/análogos & derivados , Cisteína/genética , Cisteína/metabolismo , Regulación de la Expresión Génica , Terapia Genética , Humanos , Lentivirus/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...