Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Antiviral Res ; 221: 105781, 2024 01.
Artículo en Inglés | MEDLINE | ID: mdl-38097049

RESUMEN

Coxsackievirus B6 (CVB6), a member of the human enterovirus family, is associated with severe diseases such as myocarditis in children. However, to date, only a limited number of CVB6 strains have been identified, and their characterization in animal models has been lacking. To address this gap, in this study, a neonatal murine model of CVB6 infection was established to compare the replication and virulence of three infectious-clone-derived CVB6 strains in vivo. The results showed that following challenge with a lethal dose of CVB6 strains, the neonatal mice rapidly exhibited a series of clinical signs, such as weight loss, limb paralysis, and death. For the two high-virulence CVB6 strains, histological examination revealed myocyte necrosis in skeletal and cardiac muscle, and immunohistochemistry confirmed the expression of CVB6 viral protein in these tissues. Real-time PCR assay also revealed higher viral loads in the skeletal and cardiac muscle than in other tissues at different time points post infection. Furthermore, the protective effect of passive immunization with antisera and a neutralizing monoclonal antibody against CVB6 infection was evaluated in the neonatal mouse model. This study should provide insights into the pathogenesis of CVB6 and facilitate further research in the development of vaccines and antivirals against CVBs.


Asunto(s)
Infecciones por Coxsackievirus , Enterovirus , Niño , Animales , Ratones , Humanos , Modelos Animales de Enfermedad , Virulencia , Enterovirus Humano B , Anticuerpos Neutralizantes/uso terapéutico , Ratones Endogámicos C57BL , Antivirales/farmacología , Antivirales/uso terapéutico
2.
J Virol ; 96(21): e0137322, 2022 11 09.
Artículo en Inglés | MEDLINE | ID: mdl-36226984

RESUMEN

Zika virus (ZIKV) is transmitted mostly via mosquito bites and no vaccine is available, so it may reemerge. We and others previously demonstrated that neonatal infection of ZIKV results in heart failure and can be fatal. Animal models implicated ZIKV involvement in viral heart diseases. It is unknown whether and how ZIKV causes heart failure in adults. Herein, we studied the effects of ZIKV infection on the heart function of adult A129 mice. First, we found that ZIKV productively infects the rat-, mouse-, or human-originated heart cell lines and caused ubiquitination-mediated degradation of and distortive effects on connexin 43 (Cx43) protein that is important for communications between cardiomyocytes. Second, ZIKV infection caused 100% death of the A129 mice with decreasing body weight, worsening health score, shrugging fur, and paralysis. The viral replication was detected in multiple organs. In searching for the viral effects on heart of the A129 mice, we found that ZIKV infection resulted in the increase of cardiac muscle enzymes, implicating a viral acute myocardial injury. ZIKV-caused heart injury was also demonstrated by electrocardiogram (ECG) showing widened and fragmented QRS waves, prolonged PR interval, and slower heart rate. The intercalated disc (ICD) between two cardiomyocytes was destroyed, as shown by the electronic microscopy, and the Cx43 distribution in the ICDs was less organized in the ZIKV-infected mice compared to that in the phosphate-buffered saline (PBS)-treated mice. Consistently, ZIKV productively infected the heart of A129 mice and decreased Cx43 protein. Therefore, we demonstrated that ZIKV infection caused heart failure, which might lead to fatal sequelae in ZIKV-infected A129 mice. IMPORTANCE Zika virus (ZIKV) is a teratogen causing devastating sequelae to the newborns who suffer a congenital ZIKV infection while it brings about only mild symptoms to the health-competent older children or adults. Mouse models have played an important role in mechanistic and pathogenic studies of ZIKV. In this study, we employed 3 to 4 week-old A129 mice for ZIKV infection. RT-qPCR assays discovered that ZIKV replicated in multiple organs, including the heart. As a result of ZIKV infection, the A129 mice experienced weight loss, health score worsening, paralysis, and deaths. We revealed that the ZIKV infection caused abnormal electrocardiogram presentations, increased cardiac muscle enzymes, downregulated Cx43, and destroyed the gap junction and the intercalated disc between the cardiomyocytes, implicating that ZIKV may cause an acute myocardial injury in A129 mice. Therefore, our data imply that ZIKV infection may jeopardize the immunocompromised population with a severe clinical consequence, such as heart defect.


Asunto(s)
Cardiopatías , Insuficiencia Cardíaca , Infección por el Virus Zika , Virus Zika , Recién Nacido , Niño , Animales , Ratones , Humanos , Ratas , Adolescente , Conexina 43 , Miocitos Cardíacos/patología , Modelos Animales de Enfermedad , Uniones Comunicantes/patología , Parálisis
3.
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
4.
Virol Sin ; 36(6): 1575-1584, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34581960

RESUMEN

Coxsackievirus B1 (CVB1) is a leading causative agent of severe infectious diseases in humans and has been reported to be associated with outbreaks of aseptic meningitis, myocarditis, and the development of chronic diseases such as type 1 diabetes mellitus (T1DM). There is no approved vaccine or effective antiviral therapy to treat CBV1 infection. And animal models to assess the effects of antiviral agents and vaccine remain limited. In this study, we established a neonatal mouse model of CVB1 using a clinically isolated strain to characterize the pathological manifestations of virus infection and to promote the development of vaccines and antiviral drugs against CVB1. One-day-old BALB/c mice were susceptible to CVB1 infection by intraperitoneal injection. Mice challenged with CVB1 at a low dose [10 median tissue culture infective dose (TCID50)] exhibited a series of clinical symptoms, such as inactivity, emaciation, limb weakness, hair thinning, hunching and even death. Pathological examination and tissue viral load analysis showed that positive signals of CVB1 were detected in the heart, spinal cord, limb muscle and kidney without pathological damage. Particularly, CVB1 had a strong tropism towards the pancreas, causing severe cellular necrosis with inflammatory infiltration, and was spread by viraemia. Notably, the monoclonal antibody (mAb) 6H5 and antisera elicited from CVB1-vaccinated mice effectively protected the mice from CVB1 infection in the mouse model. In summary, the established neonatal mouse model is an effective tool for evaluating the efficacy of CVB1 antiviral reagents and vaccines.


Asunto(s)
Infecciones por Coxsackievirus , Vacunas Virales , Animales , Animales Recién Nacidos , Antivirales/farmacología , Infecciones por Coxsackievirus/tratamiento farmacológico , Modelos Animales de Enfermedad , Ratones
5.
Theranostics ; 11(13): 6607-6615, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33995679

RESUMEN

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.


Asunto(s)
COVID-19/inmunología , Modelos Animales de Enfermedad , Pulmón/patología , Mucosa Respiratoria/citología , SARS-CoV-2/inmunología , Feto Abortado , Animales , COVID-19/patología , COVID-19/virología , Células Cultivadas , Células Epiteliales/virología , Xenoinjertos , Humanos , Pulmón/inmunología , Pulmón/virología , Trasplante de Pulmón , Masculino , Ratones , Ratones SCID , Cultivo Primario de Células , SARS-CoV-2/patogenicidad , Replicación Viral
6.
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
7.
Virol J ; 15(1): 65, 2018 04 10.
Artículo en Inglés | MEDLINE | ID: mdl-29631630

RESUMEN

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.


Asunto(s)
Neoplasias Endometriales/terapia , Neoplasias Endometriales/virología , Enterovirus/fisiología , Viroterapia Oncolítica , Virus Oncolíticos/fisiología , Animales , Línea Celular Tumoral , Neoplasias Endometriales/patología , Enterovirus/aislamiento & purificación , Femenino , Humanos , Ratones Endogámicos BALB C , Ratones Desnudos , Trasplante de Neoplasias , Virus Oncolíticos/aislamiento & purificación , Receptores Virales/metabolismo , Replicación Viral , Ensayos Antitumor por Modelo de Xenoinjerto
8.
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
9.
Virology ; 512: 34-38, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-28910710

RESUMEN

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.


Asunto(s)
Células Epiteliales/patología , Células Epiteliales/virología , Células Gigantes/virología , Herpesvirus Humano 3/fisiología , Membrana Nuclear/patología , Fusión Celular , Línea Celular , Humanos , Membrana Nuclear/virología , Piel/citología
10.
Antiviral Res ; 146: 45-53, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28827121

RESUMEN

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.


Asunto(s)
Modelos Animales de Enfermedad , Herpesvirus Humano 3/fisiología , Pulmón/virología , Infección por el Virus de la Varicela-Zóster/virología , Animales , Citocinas/biosíntesis , Citocinas/inmunología , Herpesvirus Humano 3/inmunología , Herpesvirus Humano 3/aislamiento & purificación , Xenoinjertos , Humanos , Pulmón/fisiopatología , Trasplante de Pulmón , Ratones , Ratones SCID , Trasplante Heterólogo , Infección por el Virus de la Varicela-Zóster/inmunología , Replicación Viral
11.
Antiviral Res ; 143: 69-73, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28404225

RESUMEN

Cholesterol 25-hydroxylase (Ch25h) is an interferon-inducible protein, and recent studies have demonstrated that it inhibited the replication of many enveloped viruses. However, in this study, we found that cells infected with wild-type (WT) HSV-1 reduced the expression of Ch25h, and ectopic expression of Ch25h could not inhibit the replication of WT-HSV-1. By screening assay, HSV-1 UL41 protein was found to down-regulate the expression of Ch25h. In addition, UL41 abrogated the antiviral activity of Ch25h via degrading its mRNA. Furthermore, ectopic expression of Ch25h inhibited the replication of UL41-null mutant HSV-1 (R2621), but not WT-HSV-1, and knockdown of Ch25h did not affect the replication of WT-HSV-1, but promoted the replication of the R2621. For the first time, HSV-1 UL41 was demonstrated to evade the antiviral function of Ch25h via its endonuclease activity.


Asunto(s)
Antivirales/farmacología , Herpesvirus Humano 1/efectos de los fármacos , Esteroide Hidroxilasas/farmacología , Virión/efectos de los fármacos , Regulación Viral de la Expresión Génica , Técnicas de Silenciamiento del Gen , Células HEK293 , Herpes Simple/tratamiento farmacológico , Herpesvirus Humano 1/patogenicidad , Humanos , ARN Mensajero/genética , Esteroide Hidroxilasas/genética , Proteínas Virales/genética , Proteínas Virales/metabolismo , Replicación Viral/efectos de los fármacos
12.
Virology ; 491: 96-105, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26891237

RESUMEN

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.


Asunto(s)
Antígenos CD59/genética , Varicela/genética , Herpesvirus Humano 3/fisiología , Animales , Antígenos CD59/metabolismo , Varicela/metabolismo , Varicela/patología , Varicela/virología , Modelos Animales de Enfermedad , Ganglios Espinales/metabolismo , Ganglios Espinales/patología , Ganglios Espinales/virología , Herpesvirus Humano 3/genética , Interacciones Huésped-Patógeno , Humanos , Hígado/metabolismo , Hígado/patología , Hígado/virología , Masculino , Ratones , Ratones SCID , Timo/metabolismo , Timo/patología , Timo/virología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA