Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 52
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Curr Top Microbiol Immunol ; 438: 103-134, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-34904194

RESUMEN

Latency and reactivation in neurons are critical aspects of VZV pathogenesis that have historically been difficult to investigate. Viral genomes are retained in many human ganglia after the primary infection, varicella; and about one-third of the naturally infected VZV seropositive population reactivates latent virus, which most often clinically manifests as herpes zoster (HZ or Shingles). HZ is frequently complicated by acute and chronic debilitating pain for which there remains a need for more effective treatment options. Understanding of the latent state is likely to be essential in the design of strategies to reduce reactivation. Experimentally addressing VZV latency has been difficult because of the strict human species specificity of VZV and the fact that until recently, experimental reactivation had not been achieved. We do not yet know the neuron subtypes that harbor latent genomes, whether all can potentially reactivate, what the drivers of VZV reactivation are, and how immunity interplays with the latent state to control reactivation. However, recent advances have enabled a picture of VZV latency to start to emerge. The first is the ability to detect the latent viral genome and its expression in human ganglionic tissues with extraordinary sensitivity. The second, the subject of this chapter, is the development of in vitro human neuron systems permitting the modeling of latent states that can be experimentally reactivated. This review will summarize recent advances of in vitro models of neuronal VZV latency and reactivation, the limitations of the current systems, and discuss outstanding questions and future directions regarding these processes using these and yet to be developed models. Results obtained from the in vitro models to date will also be discussed in light of the recent data gleaned from studies of VZV latency and gene expression learned from human cadaver ganglia, especially the discovery of VZV latency transcripts that seem to parallel the long-studied latency-associated transcripts of other neurotropic alphaherpesviruses.


Asunto(s)
Varicela , Herpes Zóster , Humanos , Herpesvirus Humano 3/genética , Activación Viral/genética , Latencia del Virus/genética , Herpes Zóster/patología , Neuronas/patología
2.
J Virol ; 94(13)2020 06 16.
Artículo en Inglés | MEDLINE | ID: mdl-32295909

RESUMEN

Small noncoding RNAs (sncRNA), including microRNA (miR), are expressed by many viruses to provide an additional layer of gene expression regulation. Our work has shown that varicella-zoster virus (VZV; also called human herpesvirus 3 [HHV3]), the human alphaherpesvirus causing varicella and herpes zoster, expresses 24 virally encoded sncRNA (VZVsncRNA) in infected cells. Here, we demonstrate that several VZVsncRNA can modulate VZV growth, including four VZVsncRNA (VZVsncRNA10, -11, -12, and -13) that are antisense to VLT, a transcript made in lytic infections and associated with VZV latency. The influence on productive VZV growth and spread was assessed in epithelial cells transfected with locked nucleotide analog antagonists (LNAA). LNAA to the four VZVsncRNA antisense to VLT significantly reduced viral spread and progeny titers of infectious virus, suggesting that these sncRNA promoted lytic infection. The LNAA to VZVsncRNA12, encoded in the leader to ORF61, also significantly increased the levels of VLT transcripts. Conversely, overexpression of VZVsncRNA13 using adeno-associated virus consistently increased VZV spread and progeny titers. These results suggest that sncRNA antisense to VZV may regulate VZV growth, possibly by affecting VLT expression. Transfection of LNAA to VZVsncRNA14 and VZVsncRNA9 decreased and increased VZV growth, respectively, while LNAA to three other VZVsncRNA had no significant effects on replication. These data strongly support the conclusion that VZV replication is modulated by multiple virally encoded sncRNA, revealing an additional layer of complexity of VZV regulation of lytic infections. This may inform the development of novel anti-sncRNA-based therapies for treatment of VZV diseases.IMPORTANCE Varicella-zoster virus (VZV) causes herpes zoster, a major health issue in the aging and immunocompromised populations. Small noncoding RNAs (sncRNA) are recognized as important actors in modulating gene expression. This study extends our previous work and shows that four VZVsncRNA clustering in and near ORF61 and antisense to the latency-associated transcript of VZV can positively influence productive VZV infection. The ability of multiple exogenous small oligonucleotides targeting VZVsncRNA to inhibit VZV replication strengthens the possibility that they may inform development of novel treatments for painful herpes zoster.


Asunto(s)
Herpesvirus Humano 3/genética , ARN Pequeño no Traducido/genética , ARN Pequeño no Traducido/metabolismo , Varicela/genética , Varicela/virología , Herpes Zóster/genética , Herpes Zóster/virología , Herpesvirus Humano 3/crecimiento & desarrollo , Humanos , MicroARNs/metabolismo , Neuronas/virología , Latencia del Virus , Replicación Viral
3.
Exp Eye Res ; 180: 29-38, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30447199

RESUMEN

Cell replacement therapy is a promising approach for treatment of retinal degenerative diseases. Several protocols for the generation of photoreceptor precursors (PRP) from human embryonic stem cells (hESC) have been reported with variable efficiency. Herein, we show the advantages of use of size-controlled embryoid bodies in the ESC differentiation process using two differentiation protocols. We further explored cell-labeling methods for following the survival of PRP transplanted subretinally in rat eyes. Size-controlled embryoid bodies (EBs) generated using microwell dishes and non-size-controlled EBs generated using V-shaped 96-well plates were differentiated into PRP using two differentiation protocols. The differentiation protocols utilized two different combinations of growth factors. The first, Dkk1, Noggin, and IGF1, and the second protocol used IWR1e, SAG, and CHIR99021. Differentiation efficiency to PRP was analyzed by qPCR, immunocytochemistry, and fluorescence-assisted cell sorting (FACS). Size-controlled IWR1e yielded a significantly higher percent (86.4%) of PRP cells expressing CRX, compared with non-size-controlled IWR1e (51.4%, P = 0.026) or the size-controlled DKK1 protocol (70.5%, p = 0.007). In addition, the IWR1e differentiated cells exhibited a significantly higher fluorescence intensity of CRX immunostaining, compared with the DKK1 protocol, consistent with higher protein expression levels. The IWR1e cells exhibited higher maturation levels, as manifested by lower early neuronal marker PAX6 and pluripotency marker OCT4 levels compared with the DKK1 protocol. The expression of other late photoreceptor markers (NRL, recoverin) were similar among the differentiation groups. PRP cells were labeled by using hESC constitutively expressing EGFP or by AAV-GFP transduction. Finally, we transplanted the cells in the subretinal space of wild-type rats and monitored their survival over several weeks. The AAV2 serotype efficiently transduced the PRP cells, whereas other serotypes yielded low or no transduction. Following subretinal transplantation of GFP-labeled PRP, 63% of the cells were detected at 4 weeks post-transplantation. In conclusion, we show here that the IWR1e protocol using size-controlled EBs efficiently generated of PRP that could be labeled and followed in-vivo for weeks. The data from this study is an advance toward the goal of PRP transplantation therapy for retinal degenerative diseases.


Asunto(s)
Células Madre Embrionarias Humanas/citología , Células Fotorreceptoras/citología , Coloración y Etiquetado/métodos , Trasplante de Células Madre , Células Madre/citología , Animales , Diferenciación Celular , Supervivencia Celular , Dependovirus , Citometría de Flujo , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Inmunohistoquímica , Parvovirinae/genética , Ratas , Ratas Long-Evans , Reacción en Cadena en Tiempo Real de la Polimerasa
4.
J Virol ; 91(24)2017 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-29021397

RESUMEN

Many herpesviruses express small noncoding RNAs (sncRNAs), including microRNAs (miRNAs), that may play roles in regulating lytic and latent infections. None have yet been reported in varicella-zoster virus (VZV; also known as human herpesvirus 3 [HHV-3]). Here we analyzed next-generation sequencing (NGS) data for small RNAs in VZV-infected fibroblasts and human embryonic stem cell-derived (hESC) neurons. Two independent bioinformatics analyses identified more than 20 VZV-encoded 20- to 24-nucleotide RNAs, some of which are predicted to have stem-loop precursors potentially representing miRNAs. These sequences are perfectly conserved between viruses from three clades of VZV. One NGS-identified sequence common to both bioinformatics analyses mapped to the repeat regions of the VZV genome, upstream of the predicted promoter of the immediate early gene open reading frame 63 (ORF63). This miRNA candidate was detected in each of 3 independent biological repetitions of NGS of RNA from fibroblasts and neurons productively infected with VZV using TaqMan quantitative PCR (qPCR). Importantly, transfected synthetic RNA oligonucleotides antagonistic to the miRNA candidate significantly enhanced VZV plaque growth rates. The presence of 6 additional small noncoding RNAs was also verified by TaqMan qPCR in productively infected fibroblasts and ARPE19 cells. Our results show VZV, like other human herpesviruses, encodes several sncRNAs and miRNAs, and some may regulate infection of host cells.IMPORTANCE Varicella-zoster virus is an important human pathogen, with herpes zoster being a major health issue in the aging and immunocompromised populations. Small noncoding RNAs (sncRNAs) are recognized as important actors in modulating gene expression, and this study demonstrates the first reported VZV-encoded sncRNAs. Many are clustered to a small genomic region, as seen in other human herpesviruses. At least one VZV sncRNA was expressed in productive infection of neurons and fibroblasts that is likely to reduce viral replication. Since sncRNAs have been suggested to be potential targets for antiviral therapies, identification of these molecules in VZV may provide a new direction for development of treatments for painful herpes zoster.


Asunto(s)
Herpesvirus Humano 3/genética , MicroARNs/genética , ARN Pequeño no Traducido/genética , Biología Computacional , Fibroblastos/virología , Genoma Viral , Herpes Zóster/virología , Herpesvirus Humano 3/fisiología , Humanos , MicroARNs/biosíntesis , Neuronas/virología , Sistemas de Lectura Abierta , ARN Pequeño no Traducido/biosíntesis , ARN Pequeño no Traducido/clasificación , Análisis de Secuencia de ADN , Latencia del Virus , Replicación Viral
5.
PLoS Pathog ; 11(6): e1004885, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-26042814

RESUMEN

Varicella zoster virus (VZV) latency in sensory and autonomic neurons has remained enigmatic and difficult to study, and experimental reactivation has not yet been achieved. We have previously shown that human embryonic stem cell (hESC)-derived neurons are permissive to a productive and spreading VZV infection. We now demonstrate that hESC-derived neurons can also host a persistent non-productive infection lasting for weeks which can subsequently be reactivated by multiple experimental stimuli. Quiescent infections were established by exposing neurons to low titer cell-free VZV either by using acyclovir or by infection of axons in compartmented microfluidic chambers without acyclovir. VZV DNA and low levels of viral transcription were detectable by qPCR for up to seven weeks. Quiescently-infected human neuronal cultures were induced to undergo renewed viral gene and protein expression by growth factor removal or by inhibition of PI3-Kinase activity. Strikingly, incubation of cultures induced to reactivate at a lower temperature (34°C) resulted in enhanced VZV reactivation, resulting in spreading, productive infections. Comparison of VZV genome transcription in quiescently-infected to productively-infected neurons using RNASeq revealed preferential transcription from specific genome regions, especially the duplicated regions. These experiments establish a powerful new system for modeling the VZV latent state, and reveal a potential role for temperature in VZV reactivation and disease.


Asunto(s)
Herpesvirus Humano 3/fisiología , Células-Madre Neurales/virología , Neuronas/virología , Activación Viral/fisiología , Latencia del Virus/fisiología , Células Madre Embrionarias/virología , Herpes Zóster/virología , Humanos , Hibridación in Situ , Técnicas In Vitro , Reacción en Cadena de la Polimerasa
6.
J Virol ; 88(9): 5079-86, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24574392

RESUMEN

UNLABELLED: The two human neurotropic alphaherpesviruses varicella-zoster virus (VZV) and herpes simplex virus type 1 (HSV1) both establish latency in sensory ganglia. Human trigeminal ganglia are known to frequently harbor both viruses, and there is evidence to suggest the presence of both VZV and HSV1 DNA in the same neuron. We ask here whether VZV and HSV1 can exclude themselves and each other and whether they can productively infect the same cells in human neurons and human foreskin fibroblasts (HFF). Simultaneous infection (coinfection) or consecutive infection (superinfection) was assessed using cell-free HSV1 and VZV expressing fluorescent reporter proteins. Automated analysis was carried out to detect singly and dually infected cells. We demonstrate that VZV and HSV1 both display efficient superinfection exclusion (SE) in HFF, with each virus excluding either itself or the other virus. While SE also occurred in neurons, it was with much lower efficiency. Both alphaherpesviruses productively infected the same neurons, whether applied simultaneously or even consecutively, albeit at lower frequencies. IMPORTANCE: Superinfection exclusion by VZV for itself or the related neurotropic alphaherpesvirus HSV1 has been studied here for the first time. We find that while these viruses display classic SE in fibroblasts, SE is less efficient for both HSV1 and VZV in human neurons. The ability of multiple VZV strains to productively infect the same neurons has important implications in terms of recombination of both wild-type and vaccine strains in patients.


Asunto(s)
Herpesvirus Humano 1/fisiología , Herpesvirus Humano 3/fisiología , Neuronas/virología , Interferencia Viral , Células Cultivadas , Fibroblastos/virología , Herpesvirus Humano 1/crecimiento & desarrollo , Herpesvirus Humano 3/crecimiento & desarrollo , Humanos
7.
J Virol ; 88(13): 7674-7, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24741086

RESUMEN

Transcriptional changes following varicella-zoster virus (VZV) infection of cultured human neurons derived from embryonic stem cells were compared to those in VZV-infected human foreskin fibroblasts. Transcription of 340 neuronal genes significantly altered by VZV infection included 223 transcript changes unique to neurons. Strikingly, genes inhibiting apoptosis were upregulated in neurons, while proapoptotic gene transcription was increased in fibroblasts. These data are a basis for discovery of differences in virus-host interactions between these VZV targets.


Asunto(s)
Apoptosis/genética , Biomarcadores/metabolismo , Células Madre Embrionarias/metabolismo , Fibroblastos/metabolismo , Perfilación de la Expresión Génica , Herpesvirus Humano 3/fisiología , Neuronas/metabolismo , Células Cultivadas , Células Madre Embrionarias/citología , Células Madre Embrionarias/virología , Fibroblastos/citología , Fibroblastos/virología , Humanos , Neuronas/citología , Neuronas/virología , Análisis de Secuencia por Matrices de Oligonucleótidos
8.
Am J Pathol ; 183(5): 1621-1633, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24055371

RESUMEN

The nephron is composed of a monolayer of epithelial cells that make up its various compartments. In development, these cells begin as mesenchyme. NCAM1, abundant in the mesenchyme and early nephron lineage, ceases to express in mature kidney epithelia. We show that, once placed in culture and released from quiescence, adult human kidney epithelial cells (hKEpCs), uniformly positive for CD24/CD133, re-express NCAM1 in a specific cell subset that attains a stem/progenitor state. Immunosorted NCAM1(+) cells overexpressed early nephron progenitor markers (PAX2, SALL1, SIX2, WT1) and acquired a mesenchymal fate, indicated by high vimentim and reduced E-cadherin levels. Gene expression and microarray analysis disclosed both a proximal tubular origin of these cells and molecules regulating epithelial-mesenchymal transition. NCAM1(+) cells generated clonal progeny when cultured in the presence of fetal kidney conditioned medium, differentiated along mesenchymal lineages but retained the unique propensity to generate epithelial kidney spheres and produce epithelial renal tissue on single-cell grafting in chick CAM and mouse. Depletion of NCAM1(+) cells from hKEpCs abrogated stemness traits in vitro. Eliminating these cells during the regenerative response that follows glycerol-induced acute tubular necrosis worsened peak renal injury in vivo. Thus, higher clone-forming and developmental capacities characterize a distinct subset of adult kidney-derived cells. The ability to influence an endogenous regenerative response via NCAM1 targeting may lead to novel therapeutics for renal diseases.


Asunto(s)
Antígeno CD56/metabolismo , Células Epiteliales/metabolismo , Células Epiteliales/patología , Riñón/patología , Células Madre/metabolismo , Lesión Renal Aguda/metabolismo , Lesión Renal Aguda/patología , Adulto , Animales , Anticuerpos/metabolismo , Biomarcadores/metabolismo , Nitrógeno de la Urea Sanguínea , Diferenciación Celular/genética , Proliferación Celular , Pollos , Células Clonales , Regulación hacia Abajo/genética , Ontología de Genes , Células HEK293 , Humanos , Mesodermo/patología , Ratones , Anotación de Secuencia Molecular , Nefronas/metabolismo , Nefronas/patología , Análisis de Secuencia por Matrices de Oligonucleótidos , Esferoides Celulares/metabolismo , Esferoides Celulares/patología , Transcriptoma/genética , Regulación hacia Arriba/genética
9.
Virology ; 595: 110096, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38710129

RESUMEN

Herpes stromal keratitis is the leading cause of infectious blindness in the western world. Infection by HSV1 is most common, but VZV and hCMV also infect the cornea. Multiple models of HSV1 corneal infection exist, but none for VZV and hCMV because of their host specificity. Here, we used commercially available 3D human corneal epithelial equivalents (HCEE) to study infection by these herpesviruses. HCEE was infected by HSV-1 and hCMV without requiring scarification and resulted in spreading infections. Spread of HSV-1 infection was rapid, while that of hCMV was slow. In contrast, infections with VZV required damage to the HCEE and did not spread. Acyclovir dramatically reduced replication of HSV-1 in this model. We conclude that highly quality-controlled, readily available HCEE is a useful model to study human-restricted herpesvirus infection of the human corneal epithelium and for screening of antiviral drugs for treating HSK in an 3D model system.


Asunto(s)
Antivirales , Epitelio Corneal , Herpesvirus Humano 1 , Queratitis Herpética , Humanos , Queratitis Herpética/virología , Queratitis Herpética/tratamiento farmacológico , Epitelio Corneal/virología , Epitelio Corneal/patología , Herpesvirus Humano 1/fisiología , Herpesvirus Humano 1/efectos de los fármacos , Antivirales/farmacología , Antivirales/uso terapéutico , Herpesvirus Humano 3/fisiología , Herpesvirus Humano 3/efectos de los fármacos , Citomegalovirus/fisiología , Citomegalovirus/efectos de los fármacos , Replicación Viral , Aciclovir/farmacología , Aciclovir/uso terapéutico , Células Epiteliales/virología , Modelos Biológicos
10.
J Virol ; 86(6): 3211-8, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22238301

RESUMEN

Pluripotent human stem cells are a powerful tool for the generation of differentiated cells that can be used for the study of human disease. We recently demonstrated that neurons derived from pluripotent human embryonic stem cells (hESC) can be infected by the highly host-restricted human alphaherpesvirus varicella-zoster virus (VZV), permitting the interaction of VZV with neurons to be readily evaluated in culture. In the present study, we examine whether pluripotent hESC and neural progenitors at intermediate stages of differentiation are permissive for VZV infection. We demonstrate here that VZV infection is blocked in naïve hESC. A block to VZV replication is also seen when a bacterial artificial chromosome (BAC) containing the VZV genome is transfected into hESC. In contrast, related alphaherpesviruses herpes simplex virus 1 (HSV-1) and pseudorabies virus (PrV) productively infect naïve hESC in a cell-free manner, and PrV replicates from a BAC transfected into hESC. Neurons differentiate from hESC via neural progenitor intermediates, as is the case in the embryo. The first in vitro stage at which permissiveness of hESC-derived neural precursors to VZV replication is observed is upon formation of "neurospheres," immediately after detachment from the inductive stromal feeder layer. These findings suggest that hESC may be useful in deciphering the yet enigmatic mechanisms of specificity of VZV infection and replication.


Asunto(s)
Células Madre Embrionarias/virología , Herpesvirus Humano 3/fisiología , Neuronas/virología , Células Madre Pluripotentes/virología , Diferenciación Celular , Células Cultivadas , Células Madre Embrionarias/citología , Herpesvirus Humano 3/genética , Humanos , Neuronas/citología , Células Madre Pluripotentes/citología , Replicación Viral
11.
J Virol ; 86(16): 8614-24, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22674980

RESUMEN

Varicella-zoster virus (VZV) is the causative agent of chickenpox and herpes zoster (shingles). After the primary infection, the virus remains latent in sensory ganglia and reactivates upon weakening of the cellular immune system due to various conditions, erupting from sensory neurons and infecting the corresponding skin tissue. The current varicella vaccine is highly attenuated in the skin and yet retains its neurovirulence and may reactivate and damage sensory neurons. The factors involved in neuronal invasion and establishment of latency are still elusive. Previously, we constructed a library of whole-gene deletion mutants carrying a bacterial artificial chromosome sequence and a luciferase marker in order to perform a comprehensive VZV genome functional analysis. Here, screening of dispensable gene deletion mutants in differentiated neuronal cells led to the identification of ORF7 as the first known, likely a main, VZV neurotropic factor. ORF7 is a virion component localized to the Golgi compartment in infected cells, whose deletion causes loss of polykaryon formation in epithelial cell culture. Interestingly, ORF7 deletion completely abolishes viral spread in human nervous tissue ex vivo and in an in vivo mouse model. This finding adds to our previous report that ORF7 is also a skin-tropic factor. The results of our investigation will not only lead to a better understanding of VZV neurotropism but could also contribute to the development of a neuroattenuated vaccine candidate against shingles or a vector for delivery of other antigens.


Asunto(s)
Herpesvirus Humano 3/patogenicidad , Neuronas/virología , Proteínas Virales/metabolismo , Factores de Virulencia/metabolismo , Animales , Modelos Animales de Enfermedad , Eliminación de Gen , Herpes Zóster/patología , Herpes Zóster/virología , Herpesvirus Humano 3/genética , Humanos , Ratones , Técnicas de Cultivo de Órganos , Proteínas Virales/genética , Virulencia , Factores de Virulencia/genética
12.
J Virol ; 85(13): 6220-33, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21525353

RESUMEN

Study of the human neurotrophic herpesvirus varicella-zoster virus (VZV) and of its ability to infect neurons has been severely limited by strict viral human tropism and limited availability of human neurons for experimentation. Human embryonic stem cells (hESC) can be differentiated to all the cell types of the body including neurons and are therefore a potentially unlimited source of human neurons to study their interactions with human neurotropic viruses. We report here reproducible infection of hESC-derived neurons by cell-associated green fluorescent protein (GFP)-expressing VZV. hESC-derived neurons expressed GFP within 2 days after incubation with mitotically inhibited MeWo cells infected with recombinant VZV expressing GFP as GFP fusions to VZV proteins or under an independent promoter. VZV infection was confirmed by immunostaining for immediate-early and viral capsid proteins. Infection of hESC-derived neurons was productive, resulting in release into the medium of infectious virions that appeared fully assembled when observed by electron microscopy. We also demonstrated, for the first time, VZV infection of axons and retrograde transport from axons to neuronal cell bodies using compartmented microfluidic chambers. The use of hESC-derived human neurons in conjunction with fluorescently tagged VZV shows great promise for the study of VZV neuronal infection and axonal transport and has potential for the establishment of a model for VZV latency in human neurons.


Asunto(s)
Axones/virología , Diferenciación Celular , Células Madre Embrionarias/citología , Herpesvirus Humano 3/patogenicidad , Neuronas/virología , Transporte Axonal , Axones/ultraestructura , Cápside/metabolismo , Cápside/ultraestructura , Células Cultivadas , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Herpesvirus Humano 3/genética , Herpesvirus Humano 3/fisiología , Humanos , Microscopía Electrónica de Transmisión , Neuronas/ultraestructura , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Proteínas Virales/genética , Proteínas Virales/metabolismo , Virión/metabolismo , Virión/ultraestructura , Liberación del Virus
13.
J Neurovirol ; 18(6): 462-70, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22918852

RESUMEN

Retrograde axonal transport of the neurotropic alphaherpesvirus Varicella zoster virus (VZV) from vesicles at the skin results in sensory neuron infection and establishment of latency. Reactivation from latency leads to painful herpes zoster. The lack of a suitable animal model of these processes for the highly human-restricted VZV has resulted in a dearth of knowledge regarding the axonal transport of VZV. We recently demonstrated VZV infection of distal axons, leading to subsequent capsid transport to the neuronal somata, and replication and release of infectious virus using a new model based on neurons derived from human embryonic stem cells (hESC). In the present study, we perform a kinetic analysis of the retrograde transport of green fluorescent protein-tagged ORF23 in VZV capsids using hESC-derived neurons compartmentalized microfluidic chambers and time-lapse video microscopy. The motion of the VZV was discontinuous, showing net retrograde movement with numerous short pauses and reversals in direction. Velocities measured were higher 1 h after infection than 6 h after infection, while run lengths were similar at both time points. The hESC-derived neuron model was also used to show that reduced neuronal spread by a VZV loss-of-function mutant for ORF7 is not due to the prevention of axonal infection and transport of the virus to the neuronal somata. hESC-derived neurons are, therefore, a powerful model for studying axonal transport of VZV and molecular characteristics of neuronal infection.


Asunto(s)
Herpesvirus Humano 3/fisiología , Neuronas/metabolismo , Neuronas/virología , Proteínas Virales/metabolismo , Latencia del Virus , Transporte Axonal , Cápside/metabolismo , Diferenciación Celular , Células Cultivadas , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Genes Reporteros , Proteínas Fluorescentes Verdes , Humanos , Cinética , Técnicas Analíticas Microfluídicas , Modelos Biológicos , Neuronas/ultraestructura , Sistemas de Lectura Abierta , Imagen de Lapso de Tiempo , Proteínas Virales/genética , Replicación Viral
14.
Viruses ; 14(5)2022 05 10.
Artículo en Inglés | MEDLINE | ID: mdl-35632756

RESUMEN

Locked-nucleotide analog antagonists (LNAA) to four varicella zoster virus small non-coding RNA (VZVsncRNA 10-13) derived from the mRNA of the open reading frame (ORF) 61 gene individually reduce VZV replication in epithelial cells and fibroblasts. To study the potential roles VZVsncRNA 10-13 have in neuronal infection we generated two recombinant VZV; one in which 8 nucleotides were changed in VZVsncRNA10 without altering the encoded residues of ORF61 (VZVsnc10MUT) and a second containing a 12-nucleotide deletion of the sequence common to VZVsncRNA12 and 13, located in the ORF61 mRNA leader sequence (VZVsnc12-13DEL). Both were developed from a VZV BAC with a green fluorescent protein (GFP) reporter fused to the N terminal of the capsid protein encoded by ORF23. The growth of both mutant VZV in epithelial cells and fibroblasts was similar to that of the parental recombinant virus. Both mutants established productive infections and experimental latency in neurons derived from human embryonic stem cells (hESC). However, neurons that were latently infected with both VZV mutant viruses showed impaired ability to reactivate when given stimuli that successfully reactivated the parental virus. These results suggest that these VZVsncRNA may have a role in VZV latency maintenance and/or reactivation. The extension of these studies and confirmation of such roles could potentially inform the development of a non-reactivating, live VZV vaccine.


Asunto(s)
Herpesvirus Humano 3 , ARN Pequeño no Traducido , Herpesvirus Humano 3/fisiología , Humanos , Mutación , Nucleótidos , ARN Pequeño no Traducido/genética , Latencia del Virus/genética
15.
Viruses ; 14(2)2022 02 12.
Artículo en Inglés | MEDLINE | ID: mdl-35215971

RESUMEN

Varicella Zoster Virus (VZV) causes Herpes Zoster (HZ), a common debilitating and complicated disease affecting up to a third of unvaccinated populations. Novel antiviral treatments for VZV reactivation and HZ are still in need. Here, we evaluated the potential of targeting the replicating and reactivating VZV genome using Clustered Regularly Interspaced Short Palindromic Repeat-Cas9 nucleases (CRISPR/Cas9) delivered by adeno-associated virus (AAV) vectors. After AAV serotype and guide RNA (gRNA) optimization, we report that a single treatment with AAV2-expressing Staphylococcus aureus CRISPR/Cas9 (saCas9) with gRNA to the duplicated and essential VZV genes ORF62/71 (AAV2-62gRsaCas9) greatly reduced VZV progeny yield and cell-to-cell spread in representative epithelial cells and in lytically infected human embryonic stem cell (hESC)-derived neurons. In contrast, AAV2-62gRsaCas9 did not reduce the replication of a recombinant virus mutated in the ORF62 targeted sequence, establishing that antiviral effects were a consequence of VZV-genome targeting. Delivery to latently infected and reactivation-induced neuron cultures also greatly reduced infectious-virus production. These results demonstrate the potential of AAV-delivered genome editors to limit VZV productive replication in epithelial cells, infected human neurons, and upon reactivation. The approach could be developed into a strategy for the treatment of VZV disease and virus spread in HZ.


Asunto(s)
Sistemas CRISPR-Cas , Dependovirus/genética , Herpesvirus Humano 3/fisiología , Neuronas/virología , Sistemas de Lectura Abierta/genética , Antivirales/farmacología , Línea Celular , Descubrimiento de Drogas , Herpesvirus Humano 3/efectos de los fármacos , Células Madre Embrionarias Humanas , Humanos , Proteínas Inmediatas-Precoces , Transactivadores , Proteínas del Envoltorio Viral , Latencia del Virus , Replicación Viral
16.
Dev Biol ; 348(1): 12-21, 2010 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-20691678

RESUMEN

TMF/ARA160 is a Golgi-associated protein to which several cellular activities have been attributed. These include, trafficking of Golgi-derived vesicles and E3 ubiquitin ligase activity. Here we show that TMF/ARA160 is required for the onset of key processes which underlie the development of mature sperm in mammals. TMF/ARA160 is highly expressed in specific spermatogenic stages. While the protein is not detected in the spermatogenic progenitor cells - spermatogonia, it accumulates in the Golgi of spermatocytes and spermatids but then disappears and is absent from spermatozoa and epididymal sperm cells. Mice that are homozygous null for TMF develop normally are healthy and the females are fertile. However, the males are sterile and their spermatids suffer from several developmental defects. They lack homing of Golgi-derived proacrosomal vesicles to the perinuclear surface, resulting in spermatozoa and epididymal sperm cells which lack acrosome. In a later developmental stage, the cytoplasm is not properly removed, thus resulting in spermatids which bare the nucleus with tightly packed DNA, surrounded by a cytoplasm. Finally, the spermatozoa of TMF(-/-) mice also suffer from misshapen heads, tails coiling around the sperm heads, and lack of motility. Taken together our findings portray TMF/ARA160 as a key regulator which is essential for the onset of key events in the differentiation and maturation of mammalian sperm and whose absence severely compromises their ability to fertilize ova.


Asunto(s)
Infertilidad Masculina/fisiopatología , Maduración del Esperma/fisiología , Espermatozoides/fisiología , Ubiquitina-Proteína Ligasas/fisiología , Proteínas de Transporte Vesicular/fisiología , Acrosoma/química , Acrosoma/ultraestructura , Citoesqueleto de Actina/ultraestructura , Animales , Diferenciación Celular , Citoplasma/metabolismo , Proteínas de Unión al ADN , Femenino , Aparato de Golgi/metabolismo , Proteínas de la Matriz de Golgi , Infertilidad Masculina/genética , Masculino , Ratones , Ratones Noqueados , Microscopía Electrónica , Mitocondrias/ultraestructura , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Cabeza del Espermatozoide/ultraestructura , Motilidad Espermática , Cola del Espermatozoide/ultraestructura , Interacciones Espermatozoide-Óvulo/fisiología , Espermátides/metabolismo , Espermátides/ultraestructura , Espermatocitos/metabolismo , Espermatocitos/ultraestructura , Espermatozoides/anomalías , Espermatozoides/ultraestructura , Factores de Transcripción , Ubiquitina-Proteína Ligasas/deficiencia , Ubiquitina-Proteína Ligasas/genética , Proteínas de Transporte Vesicular/deficiencia , Proteínas de Transporte Vesicular/genética
17.
Antiviral Res ; 193: 105144, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34303746

RESUMEN

Herpes zoster (HZ) remains a significant health burden with millions of cases in North America and Europe annually. HZ is frequently followed by long-term pain or post-herpetic neuralgia (PHN). Although effective vaccines for HZ are available, currently used nucleotide analogues often have limited effectiveness against HZ and especially PHN, so there remains a need for additional antiviral therapies for HZ. We recently identified a population of small non-coding RNA (sncRNA) encoded by Varicella Zoster Virus (VZV) and showed that single locked-nucleic acid antagonists (LNAA) to some sncRNA can modulate VZV replication in cell culture. In this work, we explored the antiviral effects of combinations of LNAA oligonucleotides targeting VZVsncRNA. Combinations of LNAA targeting three VZVsncRNA encoded in and near a critical viral regulatory gene were additive, achieving 96 % reduction in virus growth in a cell line. VZV growth was also inhibited by more than 90 % in primary human skin fibroblast cultures by individual and combinations of LNAA to VZVsncRNA. The inhibition by VZVsncRNA was specific and not a consequence of innate immune responses since LNAA to a different VZVsncRNA enhanced VZV growth. Targeted VZVsncRNA lack homologous sequences in the human transcriptome suggesting that LNAA to them would have reduced cytotoxicity if used as therapeutics. These results support further development of oligonucleotides targeting VZVsncRNA as a novel treatment for HZ.


Asunto(s)
Antivirales/farmacología , Herpesvirus Humano 3/efectos de los fármacos , Ácidos Nucleicos/farmacología , ARN Pequeño no Traducido/genética , Fibroblastos , Herpes Zóster/tratamiento farmacológico , Herpesvirus Humano 3/fisiología , Humanos , Oligonucleótidos/síntesis química , ARN Pequeño no Traducido/antagonistas & inhibidores , Replicación Viral/efectos de los fármacos
18.
Exp Dermatol ; 18(4): 409-13, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19054059

RESUMEN

The current standard method for predicting contact allergenicity is the murine local lymph node assay (LLNA). Public objection to the use of animals in testing of cosmetics makes the development of a system that does not use sentient animals highly desirable. The chorioallantoic membrane (CAM) of the chick egg has been extensively used for the growth of normal and transformed mammalian tissues. The CAM is not innervated, and embryos are sacrificed before the development of pain perception. The aim of this study was to determine whether the sensitization phase of contact dermatitis to known cosmetic allergens can be quantified using CAM-engrafted human skin and how these results compare with published EC3 data obtained with the LLNA. We studied six common molecules used in allergen testing and quantified migration of epidermal Langerhans cells (LC) as a measure of their allergic potency. All agents with known allergic potential induced statistically significant migration of LC. The data obtained correlated well with published data for these allergens generated using the LLNA test. The human-skin CAM model therefore has great potential as an inexpensive, non-radioactive, in vivo alternative to the LLNA, which does not require the use of sentient animals. In addition, this system has the advantage of testing the allergic response of human, rather than animal skin.


Asunto(s)
Alérgenos/inmunología , Membrana Corioalantoides/inmunología , Cosméticos/efectos adversos , Dermatitis Alérgica por Contacto/inmunología , Modelos Biológicos , Piel/inmunología , Animales , Movimiento Celular/efectos de los fármacos , Embrión de Pollo , Cosméticos/farmacología , Dermatitis Alérgica por Contacto/etiología , Humanos , Células de Langerhans/patología , Modelos Animales , Valor Predictivo de las Pruebas , Pruebas Cutáneas , Trasplantes
19.
Differentiation ; 76(2): 145-55, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17608731

RESUMEN

Neural precursors have been derived from human embryonic stem cells (hESC) using the bone morphogenetic protein antagonist noggin. These neural precursors can be further differentiated to produce neural cells that express central nervous system (CNS) markers. We have recently shown that naive hESC can be directed to differentiate into peripheral sensory (PS) neuron-like cells and putative neural crest precursors by co-culturing with PA6 stromal cells. In the present study, we examine whether hESC-derived neural precursors (NPC) can differentiate into the peripheral nervous system, as well as CNS cells. As little as 1 week after co-culture with PA6 cells, cells with the molecular characteristics of PS neurons and neural crest are observed in the cultures. With increased time in culture, more PS-like neurons appear, in parallel with a reduction in the neural crest-like cells. These results provide the first evidence that neural precursors derived from hESC have the potential to develop into PS neurons-like as well as CNS-like neuronal cells. About 10% of the cells in NPC-PA6 co-cultures express PS neuron markers after 3 weeks, compared with <1% of hESC cultured on PA6. This enrichment for peripheral neurons makes this an attractive system for generation of peripheral neurons for pathophysiology study and drug development for diseases of the peripheral nervous system such as Familial Dysautonomia and varicella virus infection.


Asunto(s)
Diferenciación Celular , Células Madre Embrionarias/citología , Neuronas Aferentes/citología , Animales , Proteínas Portadoras/metabolismo , Técnicas de Cocultivo , Humanos , Ratones , Nervios Periféricos/citología , Células del Estroma/metabolismo
20.
Front Microbiol ; 10: 1634, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31396173

RESUMEN

The latent state of the human herpesvirus varicella zoster virus (VZV) has remained enigmatic and controversial. While it is well substantiated that VZV persistence is established in neurons after the primary infection (varicella or chickenpox), we know little of the types of neurons harboring latent virus genomes, if all can potentially reactivate, what exactly drives the reactivation process, and the role of immunity in the control of latency. Viral gene expression during latency has been particularly difficult to resolve, although very recent advances indicate that it is more restrictive than was once thought. We do not yet understand how genes expressed in latency function in the maintenance and reactivation processes. Model systems of latency are needed to pursue these questions. This has been especially challenging for VZV because the development of in vivo models of VZV infection has proven difficult. Given that up to one third of the population will clinically reactivate VZV to develop herpes zoster (shingles) and suffer from its common long term problematic sequelae, there is still a need for both in vivo and in vitro model systems. This review will summarize the evolution of models of VZV persistence and address insights that have arisen from the establishment of new in vitro human neuron culture systems that not only harbor a latent state, but permit experimental reactivation and renewed virus production. These models will be discussed in light of the recent data gleaned from the study of VZV latency in human cadaver ganglia.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA