Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Proc Natl Acad Sci U S A ; 119(20): e2121499119, 2022 05 17.
Artigo em Inglês | MEDLINE | ID: mdl-35537048

RESUMO

As the global elderly population grows, it is socioeconomically and medically critical to provide diverse and effective means of mitigating the impact of aging on human health. Previous studies showed that the adeno-associated virus (AAV) vector induced overexpression of certain proteins, which can suppress or reverse the effects of aging in animal models. In our study, we sought to determine whether the high-capacity cytomegalovirus vector (CMV) can be an effective and safe gene delivery method for two such protective factors: telomerase reverse transcriptase (TERT) and follistatin (FST). We found that the mouse cytomegalovirus (MCMV) carrying exogenous TERT or FST (MCMVTERT or MCMVFST) extended median lifespan by 41.4% and 32.5%, respectively. We report CMV being used successfully as both an intranasal and injectable gene therapy system to extend longevity. Specifically, this treatment significantly improved glucose tolerance, physical performance, as well as preventing body mass loss and alopecia. Further, telomere shortening associated with aging was ameliorated by TERT and mitochondrial structure deterioration was halted in both treatments. Intranasal and injectable preparations performed equally well in safely and efficiently delivering gene therapy to multiple organs, with long-lasting benefits and without carcinogenicity or unwanted side effects. Translating this research to humans could have significant benefits associated with quality of life and an increased health span.


Assuntos
Infecções por Citomegalovirus , Terapia Genética , Expectativa de Vida , Telomerase , Administração por Inalação , Animais , Folistatina/genética , Terapia Genética/efeitos adversos , Terapia Genética/métodos , Vetores Genéticos/genética , Injeções Intraperitoneais , Camundongos , Modelos Animais , Neoplasias , Telomerase/genética , Telomerase/metabolismo
2.
Nat Commun ; 13(1): 824, 2022 02 11.
Artigo em Inglês | MEDLINE | ID: mdl-35149692

RESUMO

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


Assuntos
Vacina contra Varicela/imunologia , Varicela/imunologia , Pele/imunologia , Vacinas Atenuadas/imunologia , Animais , Linhagem Celular , Varicela/prevenção & controle , Feminino , Fibroblastos , Cobaias , Herpes Zoster/virologia , Herpesvirus Humano 3 , Humanos , Imunogenicidade da Vacina , Pulmão , Masculino , Camundongos , Neurônios/patologia , Coelhos , Ratos , Pele/patologia , Vacinação , Vacinas Virais
3.
J Med Virol ; 88(8): 1417-26, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27249069

RESUMO

Human cytomegalovirus (HCMV) attenuated strains, Towne, and AD169, differ from prototypic pathogenic strains, such as Toledo, in that they are missing a ∼15-kb segment in the UL/b' region. In contrast to the attenuated strains, Toledo can replicate in human tissue implants in SCID (SCID-hu) mice. Thus, this model provides a unique in vivo system to study the mechanism of viral pathogenesis. Twenty-two ORFs have been annotated in the UL/b' region, including tissue-tropic genes encoded in a pentameric gH/gl complex. To differentiate the role of the pentameric gH/gl complex versus the functions of other ORFs in the 15-kb region in supporting viral growth in vivo, a series of recombinant viral strains were constructed and their ability to replicate in SCID-hu mice was tested. The mutations in the Towne and AD169 strains were repaired to restore their pentameric gH/gl complex and it was found that these changes did not rescue their inability to replicate in the SCID-hu mice. Subsequently four deletion viruses (D1, D2, D3, and D4) in the 15-kb region from the Toledo strain were created. It was demonstrated that D2 and D3 were able to grow in SCID-hu mice, while D1 and D4 were not viable. Interestingly, co-infection of the implant with the D1 and D4 viruses could compensate their respective growth defect in vivo. The results demonstrated that rescuing viral epithelial tropism is not sufficient to revert the attenuation phenotype of AD169 or Towne, and pathogenic genes are located in the segments missing in D1 and D4 viruses. J. Med. Virol. 88:1417-1426, 2016. © 2016 Wiley Periodicals, Inc.


Assuntos
Citomegalovirus/genética , Citomegalovirus/fisiologia , Mutação , Proteínas Virais/genética , Proteínas Virais/metabolismo , Animais , Linhagem Celular , Coinfecção , Citomegalovirus/patogenicidade , Infecções por Citomegalovirus/virologia , Deleção de Genes , Genoma Viral , Humanos , Camundongos , Camundongos SCID , Camundongos Transgênicos , Fases de Leitura Aberta , Replicação Viral
4.
Lab Anim (NY) ; 45(4): 140-2, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-27003353

RESUMO

In a continuing effort to better understand the transmission and persistence of chronic wasting disease in wild populations of cervids, Colorado State University, Fort Collins houses two species of deer indoors to study the pathogenesis of chronic wasting disease. Here we report key aspects regarding the husbandry and medication of Reeves' muntjac and white-tailed deer in captivity for research purposes.


Assuntos
Experimentação Animal , Criação de Animais Domésticos/métodos , Cervos , Doença de Emaciação Crônica , Animais , Modelos Animais de Doenças , Feminino , Abrigo para Animais , Masculino
5.
J Gen Virol ; 96(11): 3444-3455, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26358706

RESUMO

The presence of disease-associated prions in tissues and bodily fluids of chronic wasting disease (CWD)-infected cervids has received much investigation, yet little is known about mother-to-offspring transmission of CWD. Our previous work demonstrated that mother-to-offspring transmission is efficient in an experimental setting. To address the question of relevance in a naturally exposed free-ranging population, we assessed maternal and fetal tissues derived from 19 elk dam-calf pairs collected from free-ranging Rocky Mountain elk from north-central Colorado, a known CWD endemic region. Conventional immunohistochemistry identified three of 19 CWD-positive dams, whereas a more sensitive assay [serial protein misfolding cyclic amplification (sPMCA)] detected CWD prion seeding activity (PrPCWD) in 15 of 19 dams. PrPCWD distribution in tissues was widespread, and included the central nervous system (CNS), lymphoreticular system, and reproductive, secretory, excretory and adipose tissues. Interestingly, five of 15 sPMCA-positive dams showed no evidence of PrPCWD in either CNS or lymphoreticular system, sites typically assessed in diagnosing CWD. Analysis of fetal tissues harvested from the 15 sPMCA-positive dams revealed PrPCWD in 80 % of fetuses (12 of 15), regardless of gestational stage. These findings demonstrated that PrPCWD is more abundant in peripheral tissues of CWD-exposed elk than current diagnostic methods suggest, and that transmission of prions from mother to offspring may contribute to the efficient transmission of CWD in naturally exposed cervid populations.


Assuntos
Animais Selvagens/metabolismo , Cervos/metabolismo , Transmissão Vertical de Doenças Infecciosas , Doenças Priônicas/transmissão , Príons/metabolismo , Doença de Emaciação Crônica/transmissão , Animais , Sistema Nervoso Central/metabolismo , Colorado , Cervos/embriologia , Feminino , Masculino , Doenças Priônicas/embriologia , Doenças Priônicas/metabolismo , Príons/genética , Distribuição Tecidual , Doença de Emaciação Crônica/embriologia , Doença de Emaciação Crônica/metabolismo
6.
J Virol ; 88(14): 7973-86, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24807720

RESUMO

The varicella-zoster virus (VZV) open reading frame 54 (ORF54) gene encodes an 87-kDa monomer that oligomerizes to form the VZV portal protein, pORF54. pORF54 was hypothesized to perform a function similar to that of a previously described herpes simplex virus 1 (HSV-1) homolog, pUL6. pUL6 and the associated viral terminase are required for processing of concatemeric viral DNA and packaging of individual viral genomes into preformed capsids. In this report, we describe two VZV bacterial artificial chromosome (BAC) constructs with ORF54 gene deletions, Δ54L (full ORF deletion) and Δ54S (partial internal deletion). The full deletion of ORF54 likely disrupted essential adjacent genes (ORF53 and ORF55) and therefore could not be complemented on an ORF54-expressing cell line (ARPE54). In contrast, Δ54S was successfully propagated in ARPE54 cells but failed to replicate in parental, noncomplementing ARPE19 cells. Transmission electron microscopy confirmed the presence of only empty VZV capsids in Δ54S-infected ARPE19 cell nuclei. Similar to the HSV-1 genome, the VZV genome is composed of a unique long region (UL) and a unique short region (US) flanked by inverted repeats. DNA from cells infected with parental VZV (VZVLUC strain) contained the predicted UL and US termini, whereas cells infected with Δ54S contained neither. This result demonstrates that Δ54S is not able to process and package viral DNA, thus making pORF54 an excellent chemotherapeutic target. In addition, the utility of BAC constructs Δ54L and Δ54S as tools for the isolation of site-directed ORF54 mutants was demonstrated by recombineering single-nucleotide changes within ORF54 that conferred resistance to VZV-specific portal protein inhibitors. Importance: Antivirals with novel mechanisms of action would provide additional therapeutic options to treat human herpesvirus infections. Proteins involved in the herpesviral DNA encapsidation process have become promising antiviral targets. Previously, we described a series of N-α-methylbenzyl-N'-aryl thiourea analogs that target the VZV portal protein (pORF54) and prevent viral replication in vitro. To better understand the mechanism of action of these compounds, it is important to define the structural and functional characteristics of the VZV portal protein. In contrast to HSV, no VZV mutants have been described for any of the seven essential DNA encapsidation genes. The VZV ORF54 deletion mutant described in this study represents the first VZV encapsidation mutant reported to date. We demonstrate that the deletion mutant can serve as a platform for the isolation of portal mutants via recombineering and provide a strategy for more in-depth studies of VZV portal structure and function.


Assuntos
DNA Viral/metabolismo , Herpesvirus Humano 3/fisiologia , Proteínas Virais/metabolismo , Montagem de Vírus , Capsídeo/ultraestrutura , Linhagem Celular , Deleção de Genes , Teste de Complementação Genética , Herpesvirus Humano 3/genética , Herpesvirus Humano 3/ultraestrutura , Humanos , Microscopia Eletrônica de Transmissão , Proteínas Virais/genética
7.
J Neurovirol ; 18(6): 462-70, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22918852

RESUMO

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.


Assuntos
Herpesvirus Humano 3/fisiologia , Neurônios/metabolismo , Neurônios/virologia , Proteínas Virais/metabolismo , Latência Viral , Transporte Axonal , Capsídeo/metabolismo , Diferenciação Celular , Células Cultivadas , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Genes Reporter , Proteínas de Fluorescência Verde , Humanos , Cinética , Técnicas Analíticas Microfluídicas , Modelos Biológicos , Neurônios/ultraestrutura , Fases de Leitura Aberta , Imagem com Lapso de Tempo , Proteínas Virais/genética , Replicação Viral
8.
J Virol ; 86(16): 8614-24, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22674980

RESUMO

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.


Assuntos
Herpesvirus Humano 3/patogenicidade , Neurônios/virologia , Proteínas Virais/metabolismo , Fatores de Virulência/metabolismo , Animais , Modelos Animais de Doenças , Deleção de Genes , Herpes Zoster/patologia , Herpes Zoster/virologia , Herpesvirus Humano 3/genética , Humanos , Camundongos , Técnicas de Cultura de Órgãos , Proteínas Virais/genética , Virulência , Fatores de Virulência/genética
9.
PLoS Pathog ; 6: e1000971, 2010 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-20617166

RESUMO

The Varicella Zoster Virus (VZV) is a ubiquitous human alpha-herpesvirus that is the causative agent of chicken pox and shingles. Although an attenuated VZV vaccine (v-Oka) has been widely used in children in the United States, chicken pox outbreaks are still seen, and the shingles vaccine only reduces the risk of shingles by 50%. Therefore, VZV still remains an important public health concern. Knowledge of VZV replication and pathogenesis remains limited due to its highly cell-associated nature in cultured cells, the difficulty of generating recombinant viruses, and VZV's almost exclusive tropism for human cells and tissues. In order to circumvent these hurdles, we cloned the entire VZV (p-Oka) genome into a bacterial artificial chromosome that included a dual-reporter system (GFP and luciferase reporter genes). We used PCR-based mutagenesis and the homologous recombination system in the E. coli to individually delete each of the genome's 70 unique ORFs. The collection of viral mutants obtained was systematically examined both in MeWo cells and in cultured human fetal skin organ samples. We use our genome-wide deletion library to provide novel functional annotations to 51% of the VZV proteome. We found 44 out of 70 VZV ORFs to be essential for viral replication. Among the 26 non-essential ORF deletion mutants, eight have discernable growth defects in MeWo. Interestingly, four ORFs were found to be required for viral replication in skin organ cultures, but not in MeWo cells, suggesting their potential roles as skin tropism factors. One of the genes (ORF7) has never been described as a skin tropic factor. The global profiling of the VZV genome gives further insights into the replication and pathogenesis of this virus, which can lead to improved prevention and therapy of chicken pox and shingles.


Assuntos
Herpesvirus Humano 3/genética , Pele/virologia , Proteínas Virais/fisiologia , Linhagem Celular Tumoral , Cromossomos Artificiais Bacterianos , Genoma Viral , Estudo de Associação Genômica Ampla , Herpesvirus Humano 3/imunologia , Herpesvirus Humano 3/fisiologia , Humanos , Pele/imunologia , Vacinas Atenuadas , Proteínas Virais/genética , Replicação Viral/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA