Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Mais filtros

Base de dados
Tipo de documento
Assunto da revista
País de afiliação
Intervalo de ano de publicação
1.
Curr Top Microbiol Immunol ; 438: 135-161, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-35292858

RESUMO

Varicella-zoster virus (VZV) is a human-restricted virus, which raises obstacles to research. The strict human tropism limits knowledge about its pathogenesis and creates challenges for evaluating antiviral treatments and vaccines. The development of humanized mouse models was driven by the need to address these challenges. Here, we summarize the humanized mouse models with xenografts of thymus/liver organoids, skin, dorsal root ganglia, and lung tissues. These models revealed VZV ORFs involved in cell tropism and pathogenesis in differentiated tissues, and made it possible to evaluate antiviral compounds in a mammalian system. Further development of skin organ culture techniques have the added benefit of lower cost and greater speed than mouse models. Human tissues, both in humanized mice and in ex vivo models, will continue to be necessary to study VZV in the tissue microenvironements to which it is adapted.


Assuntos
Herpes Zoster , Herpesvirus Humano 3 , Camundongos , Humanos , Animais , Camundongos SCID , Herpes Zoster/patologia , Xenoenxertos , Modelos Animais de Doenças , Antivirais , Mamíferos
2.
J Virol ; 94(22)2020 10 27.
Artigo em Inglês | MEDLINE | ID: mdl-32878893

RESUMO

The herpesviruses varicella-zoster virus (VZV) and human cytomegalovirus (HCMV) are endemic to humans. VZV causes varicella (chicken pox) and herpes zoster (shingles), while HCMV causes serious disease in immunocompromised patients and neonates. More effective, less toxic antivirals are needed, necessitating better models to study these viruses and evaluate antivirals. Previously, VZV and HCMV models used fetal tissue; here, we developed an adult human skin model to study VZV and HCMV in culture and in vivo While VZV is known to grow in skin, it was unknown whether skin could support an HCMV infection. We used TB40/E HCMV and POka VZV strains to evaluate virus tropism in skin organ culture (SOC) and skin xenograft mouse models. Adult human skin from reduction mammoplasties was prepared for culture on NetWells or mouse implantation. In SOC, VZV infected the epidermis and HCMV infected the dermis. Specifically, HCMV infected fibroblasts, endothelial cells, and hematopoietic cells, with some infected cells able to transfer infection. VZV and HCMV mouse models were developed by subcutaneous transplantation of skin into SCID/beige or athymic nude mice at 2 independent sites. Viruses were inoculated directly into one xenograft, and widespread infection was observed for VZV and HCMV. Notably, we detected VZV- and HCMV-infected cells in the contralateral, uninoculated xenografts, suggesting dissemination from infected xenografts occurred. For the first time, we showed HCMV successfully grows in adult human skin, as does VZV. Thus, this novel system may provide a much-needed preclinical small-animal model for HCMV and VZV and, potentially, other human-restricted viruses.IMPORTANCE Varicella-zoster virus and human cytomegalovirus infect a majority of the global population. While they often cause mild disease, serious illness and complications can arise. Unfortunately, there are few effective drugs to treat these viruses, and many are toxic. To complicate this, these viruses are restricted to replication in human cells and tissues, making them difficult to study in traditional animal models. Current models rely heavily on fetal tissues, can be prohibitively expensive, and are often complicated to generate. While fetal tissue models provide helpful insights, it is necessary to study human viruses in human tissue systems to fully understand these viruses and adequately evaluate novel antivirals. Adult human skin is an appropriate model for these viruses because many target cells are present, including basal keratinocytes, fibroblasts, dendritic cells, and lymphocytes. Skin models, in culture and xenografts in immunodeficient mice, have potential for research on viral pathogenesis, tissue tropism, dissemination, and therapy.


Assuntos
Varicela/virologia , Citomegalovirus/fisiologia , Herpes Zoster/virologia , Herpesvirus Humano 3/fisiologia , Pele/virologia , Animais , Antivirais/farmacologia , Varicela/patologia , Citomegalovirus/efeitos dos fármacos , Modelos Animais de Doenças , Células Endoteliais , Fibroblastos/patologia , Fibroblastos/virologia , Herpes Zoster/patologia , Herpesvirus Humano 3/efeitos dos fármacos , Xenoenxertos , Humanos , Masculino , Camundongos , Camundongos Nus , Camundongos SCID , Técnicas de Cultura de Órgãos , Pele/patologia
3.
J Med Chem ; 66(10): 7038-7053, 2023 05 25.
Artigo em Inglês | MEDLINE | ID: mdl-37140467

RESUMO

Varicella zoster virus (VZV) establishes lifelong infection after primary disease and can reactivate. Several drugs are approved to treat VZV diseases, but new antivirals with greater potency are needed. Previously, we identified ß-l-5-((E)-2-bromovinyl)-1-((2S,4S)-2-(hydroxymethyl)-1,3-(dioxolane-4-yl))uracil (l-BHDU, 1), which had significant anti-VZV activity. In this communication, we report the synthesis and evaluation of numerous l-BHDU prodrugs: amino acid esters (14-26), phosphoramidates (33-34), long-chain lipids (ODE-l-BHDU-MP, 38, and HDP-l-BHDU-MP, 39), and phosphate ester prodrugs (POM-l-BHDU-MP, 41, and POC-l-BHDU-MP, 47). The amino acid ester l-BHDU prodrugs (l-phenylalanine, 16, and l-valine, 17) had a potent antiviral activity with EC50 values of 0.028 and 0.030 µM, respectively. The phosphate ester prodrugs POM-l-BHDU-MP and POC-l-BHDU-MP had a significant anti-VZV activity with EC50 values of 0.035 and 0.034 µM, respectively, and no cellular toxicity (CC50 > 100 µM) was detected. Out of these prodrugs, ODE-l-BHDU-MP (38) and POM-l-BHDU-MP (41) were selected for further evaluation in future studies.


Assuntos
Dioxolanos , Pró-Fármacos , Herpesvirus Humano 3 , Uracila/farmacologia , Uracila/química , Pró-Fármacos/química , Antivirais/química , Aminoácidos , Fosfatos
4.
JID Innov ; 2(4): 100128, 2022 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-35812722

RESUMO

Arboviruses such as flaviviruses and alphaviruses cause a significant human healthcare burden on a global scale. Transmission of these viruses occurs during human blood feeding at the mosquito-skin interface. Not only do pathogen immune evasion strategies influence the initial infection and replication of pathogens delivered, but arthropod salivary factors also influence transmission foci. In vitro cell cultures do not provide an adequate environment to study complex interactions between viral, mosquito, and host factors. To address this need for a whole tissue system, we describe a proof of concept model for arbovirus infection using adult human skin ex vivo with Zika virus (flavivirus) and Mayaro virus (alphavirus). Replication of these viruses in human skin was observed up to 4 days after infection. Egressed viruses could be detected in the culture media as well. Antiviral and proinflammatory genes, including chemoattractant chemokines, were expressed in infected tissue. Immunohistochemical analysis showed the presence of virus in the skin tissue 4 days after infection. This model will be useful to further investigate: (i) the immediate molecular mechanisms of arbovirus infection in human skin, and (ii) the influence of arthropod salivary molecules during initial infection of arboviruses in a more physiologically relevant system.

5.
Viruses ; 14(4)2022 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-35458556

RESUMO

There is a continued need to understand varicella-zoster virus (VZV) pathogenesis and to develop more effective antivirals, as it causes chickenpox and zoster. As a human-restricted alphaherpesvirus, the use of human skin in culture and mice is critical in order to reveal the important VZV genes that are required for pathogenesis but that are not necessarily observed in the cell culture. We previously used VZV-expressing firefly luciferase (fLuc), under the control of the constitutively active SV40 promoter (VZV-BAC-Luc), to measure the VZV spread in the same sample. However, the fLuc expression was independent of viral gene expression and viral DNA replication programs. Here, we developed robust reporter VZV viruses by using bacterial artificial chromosome (BAC) technology, expressing luciferase from VZV-specific promoters. We also identified two spurious mutations in VZV-BAC that were corrected for maximum pathogenesis. VZV with fLuc driven by ORF57 showed superior growth in cells, human skin explants, and skin xenografts in mice. The ORF57-driven luciferase activity had a short half-life in the presence of foscarnet. This background was then used to investigate the roles for ORF36 (thymidine kinase (TK)) and ORF13 (thymidylate synthase (TS)) in skin. The studies reveal that VZV-∆TS had increased sensitivity to brivudine and was highly impaired for skin replication. This is the first report of a phenotype that is associated with the loss of TS.


Assuntos
Antivirais , Herpesvirus Humano 3 , Replicação Viral , Animais , Antivirais/farmacologia , Antivirais/uso terapêutico , Varicela , Replicação do DNA , DNA Viral , Genes Reporter , Herpes Zoster/patologia , Herpesvirus Humano 3/genética , Herpesvirus Humano 3/fisiologia , Humanos , Luciferases/genética , Camundongos , Camundongos SCID , Pele/patologia , Proteínas Virais Reguladoras e Acessórias/genética , Replicação Viral/genética
6.
Sci Rep ; 7(1): 12615, 2017 10 03.
Artigo em Inglês | MEDLINE | ID: mdl-28974743

RESUMO

Pseudomonas aeruginosa is a Gram-negative, opportunistic pathogen that infects immunocompromised and cystic fibrosis patients. Treatment is difficult due to antibiotic resistance, and new antimicrobials are needed to treat infections. The alternative sigma factor 54 (σ54, RpoN), regulates many virulence-associated genes. Thus, we evaluated inhibition of virulence in P. aeruginosa by a designed peptide (RpoN molecular roadblock, RpoN*) which binds specifically to RpoN consensus promoters. We expected that RpoN* binding to its consensus promoter sites would repress gene expression and thus virulence by blocking RpoN and/or other transcription factors. RpoN* reduced transcription of approximately 700 genes as determined by microarray analysis, including genes related to virulence. RpoN* expression significantly reduced motility, protease secretion, pyocyanin and pyoverdine production, rhamnolipid production, and biofilm formation. Given the effectiveness of RpoN* in vitro, we explored its effects in a Caenorhabditis elegans-P. aeruginosa infection model. Expression of RpoN* protected C. elegans in a paralytic killing assay, whereas worms succumbed to paralysis and death in its absence. In a slow killing assay, which mimics establishment and proliferation of an infection, C. elegans survival was prolonged when RpoN* was expressed. Thus, blocking RpoN consensus promoter sites is an effective strategy for abrogation of P. aeruginosa virulence.


Assuntos
Peptídeos/genética , Infecções por Pseudomonas/tratamento farmacológico , Pseudomonas aeruginosa/genética , RNA Polimerase Sigma 54/genética , Animais , Caenorhabditis elegans/genética , Caenorhabditis elegans/microbiologia , Movimento Celular/genética , Proliferação de Células/genética , Modelos Animais de Doenças , Farmacorresistência Bacteriana/genética , Regulação Bacteriana da Expressão Gênica , Glicolipídeos/biossíntese , Glicolipídeos/genética , Humanos , Terapia de Alvo Molecular , Peptídeos/administração & dosagem , Ligação Proteica , Infecções por Pseudomonas/genética , Infecções por Pseudomonas/patologia , Pseudomonas aeruginosa/patogenicidade , RNA Polimerase Sigma 54/administração & dosagem , RNA Polimerase Sigma 54/antagonistas & inibidores , Virulência/genética
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa