Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
Biochem Biophys Res Commun ; 613: 41-46, 2022 07 12.
Artigo em Inglês | MEDLINE | ID: mdl-35526487

RESUMO

Varicella-zoster virus (VZV) first infects hematopoietic cells, with the infected cells then acting to distribute the virus throughout the body. Sialic acid-binding immunoglobulin-like lectin (Siglec) family molecules recognize sialic acid-containing molecules on the same cell surface, called cis-ligands, or molecules on other cells or soluble agents, called trans-ligands. Among the Siglec family molecules, Siglec-4 and Siglec-7 mediate VZV infection through association with glycoprotein B (gB). As Siglec-7, but not Siglec-4, is expressed on hematopoietic cells such as monocytes, the regulatory mechanism by which Siglec-7 associates with gB is important to our understanding of VZV infection of blood cells. Here, we found that Siglec-7 is required for VZV to infect human primary monocytes. Furthermore, treatment of primary monocytes with sialidase enhanced both VZV gB binding to monocytes and VZV infectivity. Calcium influx in primary monocytes decreased the expression of Siglec-7 cis-ligands and increased VZV infectivity. These results demonstrate that the Siglec-7 cis-ligands present on primary monocytes play an important role in VZV infection through regulation of the interaction between gB and Siglec-7.


Assuntos
Antígenos de Diferenciação Mielomonocítica , Herpesvirus Humano 3 , Lectinas , Monócitos , Antígenos de Diferenciação Mielomonocítica/metabolismo , Herpesvirus Humano 3/fisiologia , Humanos , Lectinas/metabolismo , Ligantes , Monócitos/virologia , Ácido N-Acetilneuramínico , Lectinas Semelhantes a Imunoglobulina de Ligação ao Ácido Siálico , Infecção pelo Vírus da Varicela-Zoster/metabolismo , Infecção pelo Vírus da Varicela-Zoster/virologia
2.
PLoS Pathog ; 16(7): e1008473, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32649716

RESUMO

Herpesviruses are known to encode a number of inhibitors of host cell death, including RIP Homotypic Interaction Motif (RHIM)-containing proteins. Varicella zoster virus (VZV) is a member of the alphaherpesvirus subfamily and is responsible for causing chickenpox and shingles. We have identified a novel viral RHIM in the VZV capsid triplex protein, open reading frame (ORF) 20, that acts as a host cell death inhibitor. Like the human cellular RHIMs in RIPK1 and RIPK3 that stabilise the necrosome in TNF-induced necroptosis, and the viral RHIM in M45 from murine cytomegalovirus that inhibits cell death, the ORF20 RHIM is capable of forming fibrillar functional amyloid complexes. Notably, the ORF20 RHIM forms hybrid amyloid complexes with human ZBP1, a cytoplasmic sensor of viral nucleic acid. Although VZV can inhibit TNF-induced necroptosis, the ORF20 RHIM does not appear to be responsible for this inhibition. In contrast, the ZBP1 pathway is identified as important for VZV infection. Mutation of the ORF20 RHIM renders the virus incapable of efficient spread in ZBP1-expressing HT-29 cells, an effect which can be reversed by the inhibition of caspases. Therefore we conclude that the VZV ORF20 RHIM is important for preventing ZBP1-driven apoptosis during VZV infection, and propose that it mediates this effect by sequestering ZBP1 into decoy amyloid assemblies.


Assuntos
Morte Celular/fisiologia , Herpesvirus Humano 3/metabolismo , Proteínas de Ligação a RNA/metabolismo , Infecção pelo Vírus da Varicela-Zoster/metabolismo , Proteínas Virais/metabolismo , Animais , Humanos , Camundongos
3.
J Virol ; 94(16)2020 07 30.
Artigo em Inglês | MEDLINE | ID: mdl-32493818

RESUMO

The literature on the egress of different herpesviruses after secondary envelopment is contradictory. In this report, we investigated varicella-zoster virus (VZV) egress in a cell line from a child with Pompe disease, a glycogen storage disease caused by a defect in the enzyme required for glycogen digestion. In Pompe cells, both the late autophagy pathway and the mannose-6-phosphate receptor (M6PR) pathway are interrupted. We have postulated that intact autophagic flux is required for higher recoveries of VZV infectivity. To test that hypothesis, we infected Pompe cells and then assessed the VZV infectious cycle. We discovered that the infectious cycle in Pompe cells was remarkably different from that of either fibroblasts or melanoma cells. No large late endosomes filled with VZV particles were observed in Pompe cells; only individual viral particles in small vacuoles were seen. The distribution of the M6PR pathway (trans-Golgi network to late endosomes) was constrained in infected Pompe cells. When cells were analyzed with two different anti-M6PR antibodies, extensive colocalization of the major VZV glycoprotein gE (known to contain M6P residues) and the M6P receptor (M6PR) was documented in the viral highways at the surfaces of non-Pompe cells after maximum-intensity projection of confocal z-stacks, but neither gE nor the M6PR was seen in abundance at the surfaces of infected Pompe cells. Taken together, our results suggested that (i) Pompe cells lack a VZV trafficking pathway within M6PR-positive large endosomes and (ii) most infectious VZV particles in conventional cell substrates are transported via large M6PR-positive vacuoles without degradative xenophagy to the plasma membrane.IMPORTANCE The long-term goal of this research has been to determine why VZV, when grown in cultured cells, invariably is more cell associated and has a lower titer than other alphaherpesviruses, such as herpes simplex virus 1 (HSV1) or pseudorabies virus (PRV). Data from both HSV1 and PRV laboratories have identified a Rab6 secretory pathway for the transport of single enveloped viral particles from the trans-Golgi network within small vacuoles to the plasma membrane. In contrast, after secondary envelopment in fibroblasts or melanoma cells, multiple infectious VZV particles accumulated within large M6PR-positive late endosomes that were not degraded en route to the plasma membrane. We propose that this M6PR pathway is most utilized in VZV infection and least utilized in HSV1 infection, with PRV's usage being closer to HSV1's usage. Supportive data from other VZV, PRV, and HSV1 laboratories about evidence for two egress pathways are included.


Assuntos
Doença de Depósito de Glicogênio Tipo II/metabolismo , Herpesvirus Humano 3/metabolismo , Infecção pelo Vírus da Varicela-Zoster/fisiopatologia , Autofagia/fisiologia , Linhagem Celular , Linhagem Celular Tumoral , Membrana Celular/metabolismo , Varicela/virologia , Endossomos , Exocitose/fisiologia , Herpes Zoster/metabolismo , Herpesvirus Humano 1/metabolismo , Herpesvirus Humano 1/patogenicidade , Herpesvirus Humano 3/patogenicidade , Humanos , Macroautofagia/fisiologia , Receptor IGF Tipo 2/metabolismo , Vacúolos , Infecção pelo Vírus da Varicela-Zoster/metabolismo , Proteínas do Envelope Viral/metabolismo , Vírion , Rede trans-Golgi/metabolismo
4.
J Infect Dis ; 221(7): 1088-1097, 2020 03 16.
Artigo em Inglês | MEDLINE | ID: mdl-31665341

RESUMO

BACKGROUND: Herpes zoster is linked to amyloid-associated diseases, including dementia, macular degeneration, and diabetes mellitus, in epidemiological studies. Thus, we examined whether varicella-zoster virus (VZV)-infected cells produce amyloid. METHODS: Production of intracellular amyloidogenic proteins (amylin, amyloid precursor protein [APP], and amyloid-ß [Aß]) and amyloid, as well as extracellular amylin, Aß, and amyloid, was compared between mock- and VZV-infected quiescent primary human spinal astrocytes (qHA-sps). The ability of supernatant from infected cells to induce amylin or Aß42 aggregation was quantitated. Finally, the amyloidogenic activity of viral peptides was examined. RESULTS: VZV-infected qHA-sps, but not mock-infected qHA-sps, contained intracellular amylin, APP, and/or Aß, and amyloid. No differences in extracellular amylin, Aß40, or Aß42 were detected, yet only supernatant from VZV-infected cells induced amylin aggregation and, to a lesser extent, Aß42 aggregation into amyloid fibrils. VZV glycoprotein B (gB) peptides assembled into fibrils and catalyzed amylin and Aß42 aggregation. CONCLUSIONS: VZV-infected qHA-sps produced intracellular amyloid and their extracellular environment promoted aggregation of cellular peptides into amyloid fibrils that may be due, in part, to VZV gB peptides. These findings suggest that together with host and other environmental factors, VZV infection may increase the toxic amyloid burden and contribute to amyloid-associated disease progression.


Assuntos
Peptídeos beta-Amiloides , Astrócitos , Polipeptídeo Amiloide das Ilhotas Pancreáticas , Infecção pelo Vírus da Varicela-Zoster/metabolismo , Aciclovir/farmacologia , Peptídeos beta-Amiloides/química , Peptídeos beta-Amiloides/metabolismo , Antivirais/farmacologia , Astrócitos/efeitos dos fármacos , Astrócitos/metabolismo , Astrócitos/virologia , Células Cultivadas , Espaço Extracelular/metabolismo , Humanos , Espaço Intracelular/metabolismo , Polipeptídeo Amiloide das Ilhotas Pancreáticas/química , Polipeptídeo Amiloide das Ilhotas Pancreáticas/metabolismo , Proteínas do Envelope Viral/química , Proteínas do Envelope Viral/metabolismo
5.
J Virol ; 93(3)2019 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-30404793

RESUMO

Varicella-zoster virus (VZV) is associated with viremia during primary infection that is presumed to stem from infection of circulating immune cells. While VZV has been shown to be capable of infecting a number of different subsets of circulating immune cells, such as T cells, dendritic cells, and NK cells, less is known about the interaction between VZV and monocytes. Here, we demonstrate that blood-derived human monocytes are permissive to VZV replication in vitro VZV-infected monocytes exhibited each temporal class of VZV gene expression, as evidenced by immunofluorescent staining. VZV virions were observed on the cell surface and viral nucleocapsids were observed in the nucleus of VZV-infected monocytes by scanning electron microscopy. In addition, VZV-infected monocytes were able to transfer infectious virus to human fibroblasts. Infected monocytes displayed impaired dextran-mediated endocytosis, and cell surface immunophenotyping revealed the downregulation of CD14, HLA-DR, CD11b, and the macrophage colony-stimulating factor (M-CSF) receptor. Analysis of the impact of VZV infection on M-CSF-stimulated monocyte-to-macrophage differentiation demonstrated the loss of cell viability, indicating that VZV-infected monocytes were unable to differentiate into viable macrophages. In contrast, macrophages differentiated from monocytes prior to exposure to VZV were highly permissive to infection. This study defines the permissiveness of these myeloid cell types to productive VZV infection and identifies the functional impairment of VZV-infected monocytes.IMPORTANCE Primary VZV infection results in the widespread dissemination of the virus throughout the host. Viral transportation is known to be directly influenced by susceptible immune cells in the circulation. Moreover, infection of immune cells by VZV results in attenuation of the antiviral mechanisms used to control infection and limit spread. Here, we provide evidence that human monocytes, which are highly abundant in the circulation, are permissive to productive VZV infection. Furthermore, monocyte-derived macrophages were also highly permissive to VZV infection, although VZV-infected monocytes were unable to differentiate into macrophages. Exploring the relationships between VZV and permissive immune cells, such as human monocytes and macrophages, elucidates novel immune evasion strategies and provides further insight into the control that VZV has over the immune system.


Assuntos
Diferenciação Celular , Fibroblastos/citologia , Macrófagos/citologia , Monócitos/citologia , Infecção pelo Vírus da Varicela-Zoster/patologia , Vírion , Replicação Viral , Antígenos Virais/metabolismo , Sobrevivência Celular , Células Cultivadas , Endocitose , Fibroblastos/metabolismo , Fibroblastos/virologia , Herpesvirus Humano 3/isolamento & purificação , Humanos , Macrófagos/metabolismo , Macrófagos/virologia , Monócitos/metabolismo , Monócitos/virologia , Infecção pelo Vírus da Varicela-Zoster/metabolismo , Infecção pelo Vírus da Varicela-Zoster/virologia
6.
J Cutan Pathol ; 47(1): 6-11, 2020 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-31441947

RESUMO

BACKGROUND: While usually straightforward, diagnostic features of cutaneous herpes simplex virus and varicella zoster virus infection (HSV/VZV) are not always present in biopsy specimens. Although intuitively the presence of eosinophils may lead the pathologist away from the diagnosis of cutaneous HSV/VZV infection, in our practice we have noted that eosinophils are often encountered in diagnostic specimens. METHODS: To deduce the frequency with which the inflammatory response accompanying cutaneous HSV/VZV infection includes significant numbers of eosinophils, we performed a retrospective review. We included 159 specimens from our database, diagnosed between 2009 and 2017. We determined the number of eosinophils in 10 high-power fields and noted additional histologic factors including presence of follicular involvement, ulceration, and pseudolymphomatous change. RESULTS: Of all included cases, 63% had 0-1 eosinophils, 24% had 2-10 eosinophils, and 13% had more than 10 eosinophils. Statistical analysis did not reveal a significant association between any demographic or histologic features examined and the presence of increased eosinophils. CONCLUSIONS: In this study, more than one-third of biopsy specimens diagnostic of cutaneous HSV/VZV infection had a prominent number of eosinophils. The detection of eosinophils should not be unexpected and should not lessen diagnostic suspicion for cutaneous HSV/VZV infection.


Assuntos
Eosinófilos , Herpes Simples , Herpesvirus Humano 3/metabolismo , Simplexvirus/metabolismo , Pele , Infecção pelo Vírus da Varicela-Zoster , Adolescente , Adulto , Idoso , Biópsia , Criança , Pré-Escolar , Eosinófilos/metabolismo , Eosinófilos/patologia , Eosinófilos/virologia , Feminino , Herpes Simples/metabolismo , Herpes Simples/patologia , Humanos , Lactente , Recém-Nascido , Masculino , Pessoa de Meia-Idade , Pele/metabolismo , Pele/patologia , Pele/virologia , Infecção pelo Vírus da Varicela-Zoster/metabolismo , Infecção pelo Vírus da Varicela-Zoster/patologia
7.
Int J Mol Sci ; 21(8)2020 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-32326627

RESUMO

There is a growing interest in unraveling gene expression mechanisms leading to viral host invasion and infection progression. Current findings reveal that long non-coding RNAs (lncRNAs) are implicated in the regulation of the immune system by influencing gene expression through a wide range of mechanisms. By mining whole-transcriptome shotgun sequencing (RNA-seq) data using machine learning approaches, we detected two lncRNAs (ENSG00000254680 and ENSG00000273149) that are downregulated in a wide range of viral infections and different cell types, including blood monocluclear cells, umbilical vein endothelial cells, and dermal fibroblasts. The efficiency of these two lncRNAs was positively validated in different viral phenotypic scenarios. These two lncRNAs showed a strong downregulation in virus-infected patients when compared to healthy control transcriptomes, indicating that these biomarkers are promising targets for infection diagnosis. To the best of our knowledge, this is the very first study using host lncRNAs biomarkers for the diagnosis of human viral infections.


Assuntos
Células Endoteliais/metabolismo , Fibroblastos/metabolismo , Monócitos/metabolismo , RNA Longo não Codificante/sangue , Viroses/metabolismo , Adulto , Povo Asiático , Biomarcadores/sangue , Biomarcadores/metabolismo , Pré-Escolar , Mineração de Dados , Regulação para Baixo , Células Endoteliais/microbiologia , Infecções por Escherichia coli/genética , Infecções por Escherichia coli/metabolismo , Fibroblastos/microbiologia , Células Endoteliais da Veia Umbilical Humana , Humanos , Influenza Humana/genética , Influenza Humana/metabolismo , Aprendizado de Máquina , México , Monócitos/microbiologia , Monócitos/virologia , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo , RNA-Seq , Infecções por Rotavirus/genética , Infecções por Rotavirus/metabolismo , Infecção pelo Vírus da Varicela-Zoster/genética , Infecção pelo Vírus da Varicela-Zoster/metabolismo , Viroses/genética , População Branca
8.
Artigo em Inglês | MEDLINE | ID: mdl-34759019

RESUMO

BACKGROUND AND OBJECTIVES: Compared with stroke controls, patients with varicella zoster virus (VZV) vasculopathy have increased amyloid in CSF, along with increased amylin (islet amyloid polypeptide [IAPP]) and anti-VZV antibodies. Thus, we examined the gene expression profiles of VZV-infected primary human brain vascular adventitial fibroblasts (HBVAFs), one of the initial arterial cells infected in VZV vasculopathy, to determine whether they are a potential source of amyloid that can disrupt vasculature and potentiate inflammation. METHODS: Mock- and VZV-infected quiescent HBVAFs were harvested at 3 days postinfection. Targeted RNA sequencing of the whole-human transcriptome (BioSpyder Technologies, TempO-Seq) was conducted followed by gene set enrichment and pathway analysis. Selected pathways unique to VZV-infected cells were confirmed by enzyme-linked immunoassays, migration assays, and immunofluorescence analysis (IFA) that included antibodies against amylin and amyloid-beta, as well as amyloid staining by Thioflavin-T. RESULTS: Compared with mock, VZV-infected HBVAFs had significantly enriched gene expression pathways involved in vascular remodeling and vascular diseases; confirmatory studies showed secretion of matrix metalloproteinase-3 and -10, as well increased migration of infected cells and uninfected cells when exposed to conditioned media from VZV-infected cells. In addition, significantly enriched pathways involved in amyloid-associated diseases (diabetes mellitus, amyloidosis, and Alzheimer disease), tauopathy, and progressive neurologic disorder were identified; predicted upstream regulators included amyloid precursor protein, apolipoprotein E, microtubule-associated protein tau, presenilin 1, and IAPP. Confirmatory IFA showed that VZV-infected HBVAFs contained amyloidogenic peptides (amyloid-beta and amylin) and intracellular amyloid. DISCUSSION: Gene expression profiles and pathway enrichment analysis of VZV-infected HBVAFs, as well as phenotypic studies, reveal features of pathologic vascular remodeling (e.g., increased cell migration and changes in the extracellular matrix) that can contribute to cerebrovascular disease. Furthermore, the discovery of amyloid-associated transcriptional pathways and intracellular amyloid deposition in HBVAFs raise the possibility that VZV vasculopathy is an amyloid disease. Amyloid deposition may contribute to cell death and loss of vascular wall integrity, as well as potentiate chronic inflammation in VZV vasculopathy, with disease severity and recurrence determined by the host's ability to clear virus infection and amyloid deposition and by the coexistence of other amyloid-associated diseases (i.e., Alzheimer disease and diabetes mellitus).


Assuntos
Túnica Adventícia , Peptídeos beta-Amiloides/metabolismo , Transtornos Cerebrovasculares , Fibroblastos , Infecção pelo Vírus da Varicela-Zoster , Remodelação Vascular , Túnica Adventícia/citologia , Túnica Adventícia/metabolismo , Túnica Adventícia/patologia , Túnica Adventícia/virologia , Células Cultivadas , Transtornos Cerebrovasculares/metabolismo , Transtornos Cerebrovasculares/patologia , Transtornos Cerebrovasculares/virologia , Fibroblastos/citologia , Fibroblastos/metabolismo , Fibroblastos/patologia , Fibroblastos/virologia , Humanos , Análise de Sequência de RNA , Transcriptoma/fisiologia , Infecção pelo Vírus da Varicela-Zoster/metabolismo , Infecção pelo Vírus da Varicela-Zoster/patologia , Infecção pelo Vírus da Varicela-Zoster/virologia , Remodelação Vascular/fisiologia
9.
Biochem Pharmacol ; 158: 201-206, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30365949

RESUMO

Existing treatments have limited efficacy against severe infection associated with herpes simplex virus (HSV) and herpes zoster virus (VZV), particularly in immunocompromized patients and those with multidermatomal infection. This issue, along with issues regarding drug resistance, support the need for improved therapeutic options. To investigate the antiviral effect of amenamevir, a VZV and HSV helicase-primase inhibitor, in severe infection conditions, mouse models of severe HSV-1 infection were developed by immunosuppression or multidermatomal infection. Mice with cyclosporin-induced immunosuppression and HSV-1 infection via inoculation of a dorsolateral area of skin were orally treated with amenamevir (10-100 mg/kg/day) for different durations (2-5 days). Immunosuppressed mice maintained high skin HSV-1 titers in the absence of treatment. Amenamevir successfully reduced HSV-1 titers at all tested doses in immunosuppressed mice, but required a longer treatment period to avoid a rebound in viral titers due to immunosuppression. To compare the efficacy of amenamevir and valacyclovir, a murine model of multidermatomal HSV-1 infection was generated by scarifying the dorsolateral area of skin in a line and inoculating broadly with HSV-1. The mice were treated with amenamevir or valacyclovir starting on Day 3, 4, or 5 post-infection for 5 days. Although both drugs similarly reduced disease scores when treatment was started on Day 3, amenamevir also reduced disease severity when treatment was initiated on Day 4, whereas valacyclovir did not. Amenamevir was not affected by the host's immune status in terms of effective oral doses and was more efficacious in treating severe cutaneous infection even when treatment initiation was delayed.


Assuntos
Antivirais/uso terapêutico , DNA Helicases/antagonistas & inibidores , DNA Primase/antagonistas & inibidores , Modelos Animais de Doenças , Oxidiazóis/uso terapêutico , Índice de Gravidade de Doença , Infecção pelo Vírus da Varicela-Zoster/tratamento farmacológico , Proteínas Virais/antagonistas & inibidores , Animais , Antivirais/farmacologia , Chlorocebus aethiops , DNA Helicases/metabolismo , DNA Primase/metabolismo , Feminino , Herpesvirus Humano 1/efeitos dos fármacos , Herpesvirus Humano 1/metabolismo , Camundongos , Oxidiazóis/farmacologia , Resultado do Tratamento , Infecção pelo Vírus da Varicela-Zoster/metabolismo , Células Vero , Proteínas Virais/metabolismo
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa