Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
PLoS Pathog ; 20(5): e1012261, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38805555

RESUMO

Marek's disease virus (MDV) vaccines were the first vaccines that protected against cancer. The avirulent turkey herpesvirus (HVT) was widely employed and protected billions of chickens from a deadly MDV infection. It is also among the most common vaccine vectors providing protection against a plethora of pathogens. HVT establishes latency in T-cells, allowing the vaccine virus to persist in the host for life. Intriguingly, the HVT genome contains telomeric repeat arrays (TMRs) at both ends; however, their role in the HVT life cycle remains elusive. We have previously shown that similar TMRs in the MDV genome facilitate its integration into host telomeres, which ensures efficient maintenance of the virus genome during latency and tumorigenesis. In this study, we investigated the role of the TMRs in HVT genome integration, latency, and reactivation in vitro and in vivo. Additionally, we examined HVT infection of feather follicles. We generated an HVT mutant lacking both TMRs (vΔTMR) that efficiently replicated in cell culture. We could demonstrate that wild type HVT integrates at the ends of chromosomes containing the telomeres in T-cells, while integration was severely impaired in the absence of the TMRs. To assess the role of TMRs in vivo, we infected one-day-old chickens with HVT or vΔTMR. vΔTMR loads were significantly reduced in the blood and hardly any virus was transported to the feather follicle epithelium where the virus is commonly shed. Strikingly, latency in the spleen and reactivation of the virus were severely impaired in the absence of the TMRs, indicating that the TMRs are crucial for the establishment of latency and reactivation of HVT. Our findings revealed that the TMRs facilitate integration of the HVT genome into host chromosomes, which ensures efficient persistence in the host, reactivation, and transport of the virus to the skin.


Assuntos
Galinhas , Doença de Marek , Telômero , Integração Viral , Latência Viral , Animais , Galinhas/virologia , Telômero/genética , Telômero/virologia , Doença de Marek/virologia , Doença de Marek/imunologia , Doença de Marek/prevenção & controle , Vetores Genéticos , Herpesvirus Meleagrídeo 1/genética , Herpesvirus Meleagrídeo 1/imunologia , Vacinas contra Doença de Marek/imunologia , Vacinas contra Doença de Marek/genética , Genoma Viral , Herpesvirus Galináceo 2/genética , Herpesvirus Galináceo 2/imunologia , Sequências Repetitivas de Ácido Nucleico , Doenças das Aves Domésticas/virologia , Doenças das Aves Domésticas/imunologia , Doenças das Aves Domésticas/prevenção & controle
2.
Genes (Basel) ; 12(10)2021 10 17.
Artigo em Inglês | MEDLINE | ID: mdl-34681024

RESUMO

The avian α-herpesvirus known as Marek's disease virus (MDV) linearly integrates its genomic DNA into host telomeres during infection. The resulting disease, Marek's disease (MD), is characterized by virally-induced lymphomas with high mortality. The temporal dynamics of MDV-positive (MDV+) transformed cells and expansion of MD lymphomas remain targets for further understanding. It also remains to be determined whether specific host chromosomal sites of MDV telomere integration confer an advantage to MDV-transformed cells during tumorigenesis. We applied MDV-specific fluorescence in situ hybridization (MDV FISH) to investigate virus-host cytogenomic interactions within and among a total of 37 gonad lymphomas and neoplastic splenic samples in birds infected with virulent MDV. We also determined single-cell, chromosome-specific MDV integration profiles within and among transformed tissue samples, including multiple samples from the same bird. Most mitotically-dividing cells within neoplastic samples had the cytogenomic phenotype of 'MDV telomere-integrated only', and tissue-specific, temporal changes in phenotype frequencies were detected. Transformed cell populations composing gonad lymphomas exhibited significantly lower diversity, in terms of heterogeneity of MDV integration profiles, at the latest stages of tumorigenesis (>50 days post-infection (dpi)). We further report high interindividual and lower intraindividual variation in MDV integration profiles of lymphoma cells. There was no evidence of integration hotspots into a specific host chromosome(s). Collectively, our data suggests that very few transformed MDV+ T cell populations present earlier in MDV-induced lymphomas (32-50 dpi), survive, and expand to become the dominant clonal population in more advanced MD lymphomas (51-62 dpi) and establish metastatic lymphomas.


Assuntos
Herpesvirus Galináceo 2/genética , Linfoma/genética , Doença de Marek/genética , Doenças das Aves Domésticas/genética , Animais , Carcinogênese/genética , Galinhas/genética , Galinhas/virologia , Herpesvirus Galináceo 2/patogenicidade , Interações Hospedeiro-Patógeno/genética , Hibridização in Situ Fluorescente , Linfoma/etiologia , Linfoma/patologia , Linfoma/virologia , Doença de Marek/complicações , Doença de Marek/patologia , Doença de Marek/virologia , Doenças das Aves Domésticas/virologia , Neoplasias Esplênicas/etiologia , Neoplasias Esplênicas/genética , Neoplasias Esplênicas/patologia , Linfócitos T/virologia , Telômero/genética , Telômero/virologia , Integração Viral/genética
3.
Pediatrics ; 148(3)2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34078749

RESUMO

Multisystem inflammatory syndrome in children (MIS-C) is a serious, sometimes life-threatening late complication of coronavirus disease 2019 (COVID-19) with multiorgan involvement and evidence of immune activation. The pathogenesis of MIS-C is not known, nor is the pathogenesis of the severe organ damage that is the hallmark of MIS-C. Human herpesvirus 6 (HHV-6), the virus responsible for roseola, is a ubiquitous herpesvirus that causes close to universal infection by the age of 3 years. HHV-6 remains latent for life and can be activated during inflammatory states, by other viruses, and by host cell apoptosis. HHV-6 has been associated with end-organ diseases, including hepatitis, carditis, and encephalitis. In addition, ∼1% of people have inherited chromosomally integrated human herpesvirus 6 (iciHHV-6), which is HHV-6 that has been integrated into chromosomal telomeric regions and is transmitted through the germ line. iciHHV-6 can be reactivated and has been associated with altered immune responses. We report here a case of MIS-C in which an initial high HHV-6 DNA polymerase chain reaction viral load assay prompted testing for iciHHV-6, which yielded a positive result. Additional research may be warranted to determine if iciHHV-6 is commonly observed in patients with MIS-C and, if so, whether it may play a part in MIS-C pathogenesis.


Assuntos
COVID-19/virologia , Herpesvirus Humano 6 , Infecções por Roseolovirus/virologia , Síndrome de Resposta Inflamatória Sistêmica/virologia , Teste de Ácido Nucleico para COVID-19 , Criança , DNA Viral/isolamento & purificação , Herpesvirus Humano 6/genética , Herpesvirus Humano 6/isolamento & purificação , Humanos , Masculino , Reação em Cadeia da Polimerase , Telômero/virologia , Carga Viral , Latência Viral
4.
Arch Toxicol ; 95(4): 1517-1520, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33740104

RESUMO

We recently demonstrated a significant shortening of age-adapted telomere length (TL) in lymphocytes of polychlorinated biphenyls (PCB)-exposed individuals. Here, we analyzed TL in individuals of the same PCB-exposed cohort during a 6-year follow-up period, investigating the change in TL between the first and second measurement as a function of time, concentration of PCBs and cytomegalovirus (CMV) infection. The age-adjusted TL of lymphocytes within the cohort of PCB-exposed individuals recovered from a first assessment in 2011 to a second assessment in 2017. Remarkably, if the concentration of lower chlorinated PCBs (LC PCBs) in 2011 was high (≥ 0.055 µg/L), the TL of CMV seropositive individuals remained significantly shortened both compared to age-adjusted controls as well as intra individually. This was confirmed by analysis of covariance as well as by multivariate linear mixed effects models. Since telomeres are responsive to various stress response pathways, including viral infection, we conclude that PCBs could contribute to immune senescence-like phenotypes associated with CMV infections and exacerbate negative aspects associated with the aging of the immune system.


Assuntos
Envelhecimento/imunologia , Infecções por Citomegalovirus/complicações , Bifenilos Policlorados/toxicidade , Telômero/imunologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Estudos de Coortes , Infecções por Citomegalovirus/imunologia , Seguimentos , Humanos , Estudos Longitudinais , Linfócitos/imunologia , Pessoa de Meia-Idade , Telômero/efeitos dos fármacos , Telômero/virologia , Encurtamento do Telômero/imunologia , Fatores de Tempo , Adulto Jovem
5.
PLoS Pathog ; 16(7): e1008683, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32658923

RESUMO

Human herpesvirus 6B (HHV-6B) is a betaherpesvirus capable of integrating its genome into the telomeres of host chromosomes. Until now, the cellular and/or viral proteins facilitating HHV-6B integration have remained elusive. Here we show that a cellular protein, the promyelocytic leukemia protein (PML) that forms nuclear bodies (PML-NBs), associates with the HHV-6B immediate early 1 (IE1) protein at telomeres. We report enhanced levels of SUMOylated IE1 in the presence of PML and have identified a putative SUMO Interacting Motif (SIM) within IE1, essential for its nuclear distribution, overall SUMOylation and association with PML to nuclear bodies. Furthermore, using PML knockout cell lines we made the original observation that PML is required for efficient HHV-6B integration into host chromosomes. Taken together, we could demonstrate that PML-NBs are important for IE1 multiSUMOylation and that PML plays an important role in HHV-6B integration into chromosomes, a strategy developed by this virus to maintain its genome in its host over long periods of time.


Assuntos
Herpesvirus Humano 6/metabolismo , Proteínas Imediatamente Precoces/metabolismo , Fosfoproteínas/metabolismo , Proteína da Leucemia Promielocítica/metabolismo , Infecções por Roseolovirus/metabolismo , Telômero/virologia , Linhagem Celular , Herpesvirus Humano 6/genética , Humanos , Infecções por Roseolovirus/genética , Sumoilação , Latência Viral/genética
6.
Cytogenet Genome Res ; 156(4): 204-214, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30572327

RESUMO

Marek's disease (MD) is an infectious disease characterized by lymphomas and high mortality in susceptible chickens. The causative and ubiquitous alpha-herpesvirus known as MD virus (MDV) integrates into host telomeres during early infection through latency, known to be an important phase for oncogenic transformation. Herein, we sought to determine the influence of vaccination and host genetics on the temporal dynamics of MDV-host genome interactions. We studied integration profiles using 2 MD vaccines that vary in protective efficacy in 2 genetic lines that differ in MD resistance/susceptibility. Virus integration of both oncogenic MDV and vaccine strains was observed in both MD susceptible and resistant birds, however, the lines differed in their dynamic telomere-integration profiles. Notably, the resistant host genotype exhibited a smaller percentage of replicating cells with the virus telomere-integrated only phenotype as compared to the susceptible genotype. Vaccination with Rispens, the most protective MD vaccine, also reduced the establishment of the virus telomere-integrated only phenotype, suggesting a significant role of the phenotype in MD lymphoma development. The effect of Rispens vaccination was most dramatic in the susceptible genotype. These results suggest important connections between vaccinal immunity, MDV telomere integration, virus-induced oncogenesis, and virus-host genome interactions in the context of host genetics and disease susceptibility.


Assuntos
Galinhas/genética , Herpesvirus Galináceo 2/fisiologia , Vacinas contra Doença de Marek/administração & dosagem , Telômero/virologia , Animais , Galinhas/virologia , Resistência à Doença , Genótipo , Herpesvirus Galináceo 2/efeitos dos fármacos , Doença de Marek/prevenção & controle , Doença de Marek/virologia , Vacinas contra Doença de Marek/farmacologia , Doenças das Aves Domésticas/prevenção & controle , Doenças das Aves Domésticas/virologia , Vacinação , Integração Viral/efeitos dos fármacos , Replicação Viral
7.
Viruses ; 10(11)2018 11 21.
Artigo em Inglês | MEDLINE | ID: mdl-30469324

RESUMO

Human herpesvirus-6A and -6B (HHV-6A and -6B) are two closely related betaherpesviruses that infect humans. Upon primary infection they establish a life-long infection termed latency, where the virus genome is integrated into the telomeres of latently infected cells. Intriguingly, HHV-6A/B can integrate into germ cells, leading to individuals with inherited chromosomally-integrated HHV-6 (iciHHV-6), who have the HHV-6 genome in every cell. It is known that telomeric repeats flanking the virus genome are essential for integration; however, the protein factors mediating integration remain enigmatic. We have previously shown that the putative viral integrase U94 is not essential for telomere integration; thus, we set out to assess the contribution of potential viral recombination proteins U41 and U70 towards integration. We could show that U70 enhances dsDNA break repair via a homology-directed mechanism using a reporter cell line. We then engineered cells to produce shRNAs targeting both U41 and U70 to inhibit their expression during infection. Using these cells in our HHV-6A in vitro integration assay, we could show that U41/U70 were dispensable for telomere integration. Furthermore, additional inhibition of the cellular recombinase Rad51 suggested that it was also not essential, indicating that other cellular and/or viral factors must mediate telomere integration.


Assuntos
Herpesvirus Humano 6/fisiologia , Telômero/virologia , Proteínas Virais/metabolismo , Integração Viral , Linhagem Celular , Inativação Gênica , Humanos , Proteínas Virais/genética
8.
Cell Cycle ; 17(17): 2187-2203, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30198385

RESUMO

The population of HIV reservoir in infected person is very small, but extremely long-lived and is a major obstacle for an HIV cure. We previously showed that cells with established HIV latency have deficiencies in DNA damage response (DDR). Here, we investigated ability of HIV-1 to interfere with telomere maintenance, and the effects of targeting telomeres on latently infected cells. Our results show that telomeres are elongated in cultured primary memory CD4 + T cells (TCM) after HIV-1 infection and when virus latency is established. Similarly, much longer telomeres were found in several Jurkat-derived latently infected cell lines, indicating that virus stimulates telomere elongation. Exposing primary CD4+ TCM cells to BRACO19, an agent targeting telomeres, resulted in a higher rate of apoptosis for infected cultures at day 3 post-infection, during HIV-1 latency and for PMA-stimulated cultures with low level of HIV-1 reactivation. Importantly, BRACO19 induced apoptosis in infected cells with potency similar to etoposide and camptothecin, whereas uninfected cells were less affected by BRACO19. We also determined that apoptosis induced by BRACO19 is not caused by telomeres shortening, but is related to formation of gamma-H2AX, implicating DNA damage or uncapping of telomeres, which triggers genome instability. In conclusion, our results indicate that HIV-1 stimulates telomere elongation during latency, suggesting that HIV reservoir has greater capacity for clonal expansion and extended lifespan. Higher rates of apoptosis in response to BRACO19 treatment suggest that HIV reservoirs are more susceptible to targeting telomere maintenance and to inhibitors targeting DDR, which is also involved in stabilizing telomeres.


Assuntos
Linfócitos T CD4-Positivos/virologia , Infecções por HIV/virologia , HIV-1/patogenicidade , Telômero/virologia , Linfócitos T CD4-Positivos/efeitos dos fármacos , Dano ao DNA/imunologia , Infecções por HIV/metabolismo , Humanos , Memória Imunológica/imunologia , Latência Viral/genética , Replicação Viral/imunologia
9.
Adv Exp Med Biol ; 1045: 209-226, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29896669

RESUMO

Upon infection and depending on the infected cell type, human herpesvirus 6A (HHV-6A) and 6B (HHV-6B) can replicate or enter a state of latency. HHV-6A and HHV-6B can integrate their genomes into host chromosomes as one way to establish latency. Viral integration takes place near the subtelomeric/telomeric junction of chromosomes. When HHV-6 infection and integration occur in gametes, the virus can be genetically transmitted. Inherited chromosomally integrated HHV-6 (iciHHV-6)-positive individuals carry one integrated HHV-6 copy per somatic cell. The prevalence of iciHHV-6+ individuals varies between 0.6% and 2%, depending on the geographical region sampled. In this chapter, the mechanisms leading to viral integration and reactivation from latency, as well as some of the biological and medical consequences associated with iciHHV-6, were discussed.


Assuntos
Cromossomos Humanos/virologia , Herpesvirus Humano 6/fisiologia , Infecções por Roseolovirus/virologia , Integração Viral , Animais , DNA Viral/genética , DNA Viral/metabolismo , Herpesvirus Humano 6/genética , Humanos , Telômero/virologia
10.
J Virol ; 91(22)2017 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-28835501

RESUMO

The genomes of human herpesvirus 6A (HHV-6A) and HHV-6B have the capacity to integrate into telomeres, the essential capping structures of chromosomes that play roles in cancer and ageing. About 1% of people worldwide are carriers of chromosomally integrated HHV-6 (ciHHV-6), which is inherited as a genetic trait. Understanding the consequences of integration for the evolution of the viral genome, for the telomere, and for the risk of disease associated with carrier status is hampered by a lack of knowledge about ciHHV-6 genomes. Here, we report an analysis of 28 ciHHV-6 genomes and show that they are significantly divergent from the few modern nonintegrated HHV-6 strains for which complete sequences are currently available. In addition, ciHHV-6B genomes in Europeans are more closely related to each other than to ciHHV-6B genomes from China and Pakistan, suggesting regional variation of the trait. Remarkably, at least one group of European ciHHV-6B carriers has inherited the same ciHHV-6B genome, integrated in the same telomere allele, from a common ancestor estimated to have existed 24,500 ± 10,600 years ago. Despite the antiquity of some, and possibly most, germ line HHV-6 integrations, the majority of ciHHV-6B (95%) and ciHHV-6A (72%) genomes contain a full set of intact viral genes and therefore appear to have the capacity for viral gene expression and full reactivation.IMPORTANCE Inheritance of HHV-6A or HHV-6B integrated into a telomere occurs at a low frequency in most populations studied to date, but its characteristics are poorly understood. However, stratification of ciHHV-6 carriers in modern populations due to common ancestry is an important consideration for genome-wide association studies that aim to identify disease risks for these people. Here, we present full sequence analysis of 28 ciHHV-6 genomes and show that ciHHV-6B in many carriers with European ancestry most likely originated from ancient integration events in a small number of ancestors. We propose that ancient ancestral origins for ciHHV-6A and ciHHV-6B are also likely in other populations. Moreover, despite their antiquity, all of the ciHHV-6 genomes appear to retain the capacity to express viral genes, and most are predicted to be capable of full viral reactivation. These discoveries represent potentially important considerations in immunocompromised patients, in particular in organ transplantation and in stem cell therapy.


Assuntos
Cromossomos Humanos , Genoma Humano , Herpesvirus Humano 6/genética , Característica Quantitativa Herdável , Telômero , Integração Viral/genética , Cromossomos Humanos/genética , Cromossomos Humanos/virologia , Feminino , Estudo de Associação Genômica Ampla , Humanos , Masculino , Telômero/genética , Telômero/virologia
11.
Viruses ; 9(8)2017 08 09.
Artigo em Inglês | MEDLINE | ID: mdl-28792463

RESUMO

The ends of linear genomes, whether viral or cellular, can elicit potent DNA damage and innate immune signals. DNA viruses entering the nucleus share many features with telomeres in their ability to either suppress or co-opt these pathways. Here, we review some of the common mechanisms that viruses and telomeres use to manage the DNA damage and innate immune response pathways. We highlight recent studies on the role of the telomere repeat-containing RNA (TERRA) in response to viral infection. We discuss how TERRA can be activated through a p53-response element embedded in a retrotransposon-like repeat found in human subtelomeres. We consider how TERRA can function as a danger signal when secreted in extracellular vesicles to induce inflammatory cytokines in neighboring cells. These findings suggest that TERRA may be part of the innate immune response to viral infection, and support the hypothesis that telomeres and viruses utilize common mechanisms to maintain genome integrity and regulate innate immunity.


Assuntos
Imunidade Inata , RNA Longo não Codificante/genética , Telômero/fisiologia , Viroses/fisiopatologia , Citocinas/imunologia , Dano ao DNA , Exossomos/fisiologia , Genes p53 , Humanos , Proteínas Nucleares/genética , Fosfoproteínas/genética , RNA não Traduzido/genética , Telômero/virologia , Viroses/imunologia , Viroses/metabolismo , Viroses/virologia
12.
Viruses ; 9(7)2017 07 12.
Artigo em Inglês | MEDLINE | ID: mdl-28704957

RESUMO

Human herpesvirus 6A and 6B, alongside some other herpesviruses, have the striking capacity to integrate into telomeres, the terminal repeated regions of chromosomes. The chromosomally integrated forms, ciHHV-6A and ciHHV-6B, are proposed to be a state of latency and it has been shown that they can both be inherited if integration occurs in the germ line. The first step in full viral reactivation must be the release of the integrated viral genome from the telomere and here we propose various models of this release involving transcription of the viral genome, replication fork collapse, and t-circle mediated release. In this review, we also discuss the relationship between ciHHV-6 and the telomere carrying the insertion, particularly how the presence and subsequent partial or complete release of the ciHHV-6 genome may affect telomere dynamics and the risk of disease.


Assuntos
Cromossomos Humanos/virologia , Genoma Viral , Herpesvirus Humano 6/genética , Telômero/genética , Integração Viral , DNA Viral/genética , Herpesvirus Humano 6/fisiologia , Humanos , Infecções por Roseolovirus/virologia , Telômero/virologia , Vírion/genética , Latência Viral , Replicação Viral/genética
13.
Viruses ; 9(7)2017 06 26.
Artigo em Inglês | MEDLINE | ID: mdl-28672870

RESUMO

Unlike other human herpesviruses, human herpesvirus 6A and 6B (HHV-6A/B) infection can lead to integration of the viral genome in human chromosomes. When integration occurs in germinal cells, the integrated HHV-6A/B genome can be transmitted to 50% of descendants. Such individuals, carrying one copy of the HHV-6A/B genome in every cell, are referred to as having inherited chromosomally-integrated HHV-6A/B (iciHHV-6) and represent approximately 1% of the world's population. Interestingly, HHV-6A/B integrate their genomes in a specific region of the chromosomes known as telomeres. Telomeres are located at chromosomes' ends and play essential roles in chromosomal stability and the long-term proliferative potential of cells. Considering that the integrated HHV-6A/B genome is mostly intact without any gross rearrangements or deletions, integration is likely used for viral maintenance into host cells. Knowing the roles played by telomeres in cellular homeostasis, viral integration in such structure is not likely to be without consequences. At present, the mechanisms and factors involved in HHV-6A/B integration remain poorly defined. In this review, we detail the potential biological and medical impacts of HHV-6A/B integration as well as the possible chromosomal integration and viral excision processes.


Assuntos
Herpesvirus Humano 6/fisiologia , Interações Hospedeiro-Patógeno , Ativação Viral , Integração Viral , Portador Sadio/epidemiologia , Portador Sadio/virologia , Humanos , Infecções por Roseolovirus/epidemiologia , Infecções por Roseolovirus/virologia , Telômero/virologia
14.
J Gen Virol ; 98(2): 266-274, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-28284243

RESUMO

Human herpesvirus 7 (HHV-7) is a betaherpesvirus, and is phylogenetically related to both HHV-6A and HHV-6B. The presence of telomeric repeat sequences at both ends of its genome should make it equally likely to integrate into the human telomere as HHV-6. However, numerous studies have failed to detect germline integration of HHV-7, suggesting an important difference between the HHV-6A/-6B and HHV-7 genomes. In search of possible germline integrated HHV-7, we developed a sensitive and quantitative real-time PCR assay and discovered that primers designed against some parts of the HHV-7 genome can frequently miss HHV-7 positive clinical samples even though they work efficiently in cell-culture-derived HHV-7 positive materials. Using a primer pair against the U90 ORF of HHV-7, we identified a possible case of germline integration of HHV-7 with one copy of viral genome per cell in both peripheral blood cells and hair follicles. Chromosomal integration of HHV-7 in these individuals was confirmed by fluorescence in situ hybridization analysis. Germline integration of HHV-7 was further confirmed by detection of ~2.6 copies of HHV-7 in the hair follicles of one of the parents. Our results shed light on the complex nature of the HHV-7 genome in human-derived materials in comparison to cell-culture-derived materials and show the need for stringent criteria in the selection of primers for epidemiological HHV-7 studies.


Assuntos
Cromossomos Humanos/virologia , Células Germinativas/virologia , Herpesvirus Humano 7/genética , Herpesvirus Humano 7/fisiologia , Infecções por Roseolovirus/virologia , Telômero/virologia , Integração Viral , Adulto , Células Sanguíneas/virologia , Linhagem Celular , Feminino , Genoma Viral , Folículo Piloso/virologia , Humanos , Hibridização in Situ Fluorescente , Masculino , Infecções por Roseolovirus/transmissão
15.
Cell Rep ; 9(6): 2263-78, 2014 Dec 24.
Artigo em Inglês | MEDLINE | ID: mdl-25497088

RESUMO

Telomeres protect the ends of cellular chromosomes. We show here that infection with herpes simplex virus 1 (HSV-1) results in chromosomal structural aberrations at telomeres and the accumulation of telomere dysfunction-induced DNA damage foci (TIFs). At the molecular level, HSV-1 induces transcription of telomere repeat-containing RNA (TERRA), followed by the proteolytic degradation of the telomere protein TPP1 and loss of the telomere repeat DNA signal. The HSV-1-encoded E3 ubiquitin ligase ICP0 is required for TERRA transcription and facilitates TPP1 degradation. Small hairpin RNA (shRNA) depletion of TPP1 increases viral replication, indicating that TPP1 inhibits viral replication. Viral replication protein ICP8 forms foci that coincide with telomeric proteins, and ICP8-null virus failed to degrade telomere DNA signal. These findings suggest that HSV-1 reorganizes telomeres to form ICP8-associated prereplication foci and to promote viral genomic replication.


Assuntos
Herpesvirus Humano 1/fisiologia , Telômero/virologia , Replicação Viral , Linhagem Celular , Aberrações Cromossômicas , Dano ao DNA , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Dipeptidil Peptidases e Tripeptidil Peptidases/genética , Dipeptidil Peptidases e Tripeptidil Peptidases/metabolismo , Herpesvirus Humano 1/metabolismo , Humanos , Proteínas Imediatamente Precoces/genética , Proteínas Imediatamente Precoces/metabolismo , Proteólise , RNA não Traduzido/genética , RNA não Traduzido/metabolismo , Sequências Repetitivas de Ácido Nucleico , Serina Proteases/genética , Serina Proteases/metabolismo , Complexo Shelterina/metabolismo , Telômero/química , Telômero/genética , Proteínas de Ligação a Telômeros/metabolismo , Ubiquitina-Proteína Ligases/genética , Ubiquitina-Proteína Ligases/metabolismo , Proteínas Virais/genética , Proteínas Virais/metabolismo
16.
Curr Opin Virol ; 9: 111-8, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25462442

RESUMO

HHV-6 integrates its genome into telomeres of human chromosomes. Integration can occur in somatic cells or gametes, the latter leading to individuals harboring the HHV-6 genome in every cell. This condition is transmitted to descendants and referred to as inherited chromosomally integrated human herpesvirus 6 (iciHHV-6). Although integration can occur in different chromosomes, it invariably takes place in the telomere region. This integration mechanism represents a way to maintain the virus genome during latency, which is so far unique amongst human herpesviruses. Recent work provides evidence that the integrated HHV-6 genome can be mobilized from the host chromosome, resulting in the onset of disease. Details on required structural determinants, putative integration mechanisms and biological and medical consequences of iciHHV-6 are discussed.


Assuntos
Fenômenos Fisiológicos Celulares , Herpesvirus Humano 6/fisiologia , Infecções por Roseolovirus/virologia , Integração Viral , Latência Viral , Interações Hospedeiro-Patógeno , Humanos , Telômero/virologia , Ativação Viral
17.
Cancer ; 120(17): 2673-83, 2014 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-24840723

RESUMO

BACKGROUND: Telomere maintenance is crucial in carcinogenesis and tumor progression. The results of a previous study from the authors indicated that infection with high-risk human papillomavirus (HR-HPV) types 16, 18, and 58 was a risk factor for esophageal squamous cell carcinoma (ESCC) in the Shantou region of China. In the current study, the authors explored the association between HR-HPV infection, telomere length (TL), and DNA methylation and their significance in the prognosis of patients with ESCC. METHODS: TL and DNA methylation were analyzed by real-time polymerase chain reaction and methylation-specific polymerase chain reaction in 70 cases of ESCC tumor (T) and paired nontumor (NT) tissues and 50 cases of normal esophagus (NE). The prognostic value of TL and DNA methylation in ESCC was analyzed. RESULTS: TL gradually decreased from NE to NT to T tissue. TL in tumor tissue (T-TL) was found to be longer in tissue that was positive for HR-HPV compared with negative tissue and was found to be positively associated with viral load (Spearman correlation, 0.410; P = .037) and integration (represented by the ratio of HR-HPV E2 to E6/E7 genes; P = .01). The DNA methylation ratio of human telomerase reverse transcriptase was more prevalent with long (≥ 0.7) compared with short (< 0.7) T-TL and was positively correlated with T-TL (Spearman correlation, 0.318; P = .007) and HR-HPV integration (P = .036). Furthermore, Cox proportional hazards modeling revealed a high ratio of T-TL to NT-TL (≥ 0.80) as a factor of poor prognosis, independent of other clinicopathologic variables. CONCLUSIONS: HR-HPV infection and integration related to telomere elongation and DNA methylation of human telomerase reverse transcriptase may be a potential biomarker of prognosis in patients with ESCC.


Assuntos
Carcinoma de Células Escamosas/virologia , Neoplasias Esofágicas/virologia , Papillomavirus Humano 16/fisiologia , Papillomavirus Humano 18/fisiologia , Infecções por Papillomavirus/virologia , Homeostase do Telômero , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/mortalidade , Carcinoma de Células Escamosas/patologia , Metilação de DNA , Neoplasias Esofágicas/genética , Neoplasias Esofágicas/mortalidade , Neoplasias Esofágicas/patologia , Carcinoma de Células Escamosas do Esôfago , Feminino , Dosagem de Genes , Interações Hospedeiro-Patógeno , Humanos , Estimativa de Kaplan-Meier , Masculino , Pessoa de Meia-Idade , Infecções por Papillomavirus/genética , Infecções por Papillomavirus/patologia , Prognóstico , Modelos de Riscos Proporcionais , Fatores de Risco , Telomerase/genética , Telômero/genética , Telômero/metabolismo , Telômero/virologia , Proteína 2 de Ligação a Repetições Teloméricas/genética
18.
Virology ; 442(1): 3-11, 2013 Jul 20.
Artigo em Inglês | MEDLINE | ID: mdl-23648233

RESUMO

Human herpesvirus 6B (HHV-6B) is the causative agent of roseola infantum. HHV-6A and 6B can reactivate in immunosuppressed individuals and are linked with severe inflammatory response, organ rejection and central nervous system diseases. About 0.85% of the US and UK population carries an integrated HHV-6 genome in all nucleated cells through germline transmission. We have previously reported that the HHV-6A genome integrated in telomeres of patients suffering from neurological dysfunction and also in telomeres of tissue culture cells. We now report that HHV-6B also integrates in telomeres during latency. Detailed mapping of the integrated viral genomes demonstrates that a single HHV-6 genome integrates and telomere repeats join the left end of the integrated viral genome. When HEK-293 cells carrying integrated HHV-6A were exposed to the histone deacetylase inhibitor Trichostatin A, circularization and/or formation of concatamers were detected and this assay could be used to distinguish between lytic replication and latency.


Assuntos
Mapeamento Cromossômico , Genoma Viral , Herpesvirus Humano 6/genética , Telômero/virologia , Integração Viral , Linhagem Celular , Cromossomos Humanos/virologia , Replicação do DNA , DNA Viral/genética , Feminino , Células HEK293/efeitos dos fármacos , Células HEK293/virologia , Humanos , Ácidos Hidroxâmicos/farmacologia , Masculino , Infecções por Roseolovirus/virologia , Latência Viral
19.
Proc Natl Acad Sci U S A ; 107(12): 5563-8, 2010 Mar 23.
Artigo em Inglês | MEDLINE | ID: mdl-20212114

RESUMO

Previous research has suggested that human herpesvirus-6 (HHV-6) may integrate into host cell chromosomes and be vertically transmitted in the germ line, but the evidence--primarily fluorescence in situ hybridization (FISH)--is indirect. We sought, first, to definitively test these two hypotheses. Peripheral blood mononuclear cells (PBMCs) were isolated from families in which several members, including at least one parent and child, had unusually high copy numbers of HHV-6 DNA per milliliter of blood. FISH confirmed that HHV-6 DNA colocalized with telomeric regions of one allele on chromosomes 17p13.3, 18q23, and 22q13.3, and that the integration site was identical among members of the same family. Integration of the HHV-6 genome into TTAGGG telomere repeats was confirmed by additional methods and sequencing of the integration site. Partial sequencing of the viral genome identified the same integrated HHV-6A strain within members of families, confirming vertical transmission of the viral genome. We next asked whether HHV-6A infection of naïve cell lines could lead to integration. Following infection of naïve Jjhan and HEK-293 cell lines by HHV-6, the virus integrated into telomeres. Reactivation of integrated HHV-6A virus from individuals' PBMCs as well as cell lines was successfully accomplished by compounds known to induce latent herpesvirus replication. Finally, no circular episomal forms were detected even by PCR. Taken together, the data suggest that HHV-6 is unique among human herpesviruses: it specifically and efficiently integrates into telomeres of chromosomes during latency rather than forming episomes, and the integrated viral genome is capable of producing virions.


Assuntos
Cromossomos Humanos/genética , Cromossomos Humanos/virologia , Herpesvirus Humano 6/genética , Herpesvirus Humano 6/patogenicidade , Telômero/genética , Telômero/virologia , Integração Viral/genética , Adulto , Idoso , Idoso de 80 Anos ou mais , Sequência de Bases , Linhagem Celular , Criança , DNA Viral/sangue , DNA Viral/genética , Feminino , Dosagem de Genes , Genoma Viral , Células Germinativas/virologia , Herpesvirus Humano 6/fisiologia , Humanos , Hibridização in Situ Fluorescente , Técnicas In Vitro , Transmissão Vertical de Doenças Infecciosas , Masculino , Pessoa de Meia-Idade , Dados de Sequência Molecular , Plasmídeos/sangue , Plasmídeos/genética , Infecções por Roseolovirus/genética , Infecções por Roseolovirus/transmissão , Infecções por Roseolovirus/virologia , Ativação Viral , Replicação Viral , Adulto Jovem
20.
J Immunol ; 184(7): 3417-23, 2010 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-20176738

RESUMO

Short telomeres of circulating leukocytes are a risk factor for age-related diseases, such as atherosclerosis, but the exact mechanisms generating variations in telomere length are unknown. We hypothesized that induction of differentiated T cells during chronic CMV infection would affect T cell telomere length. To test this, we measured the amount of differentiated T cells and telomere length of lymphocytes during primary CMV infection as well as CMV-seropositive and -seronegative healthy individuals. After primary CMV infection, we observed an increase in highly differentiated cells that coincided with a steep drop in telomere length. Moreover, we found in a cohort of 159 healthy individuals that telomere shortening was more rapid in CMV-seropositive individuals and correlated with the amount of differentiated T cells in both CD4(+) T cells and CD8(+) T cells. Finally, we found that telomere length measured in blood leukocytes is correlated with lymphocyte telomere length. Thus, CMV infection induces a strong decrease in T cell telomere length, which can be explained by changes in the composition of the circulating lymphocyte pool.


Assuntos
Infecções por Citomegalovirus/imunologia , Subpopulações de Linfócitos T/patologia , Linfócitos T/patologia , Telômero/patologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Antivirais/uso terapêutico , Diferenciação Celular/imunologia , Separação Celular , Infecções por Citomegalovirus/genética , Infecções por Citomegalovirus/patologia , DNA Viral/sangue , Citometria de Fluxo , Imunofluorescência , Ganciclovir/uso terapêutico , Humanos , Hibridização in Situ Fluorescente , Transplante de Rim/efeitos adversos , Transplante de Rim/imunologia , Pessoa de Meia-Idade , Reação em Cadeia da Polimerase , Subpopulações de Linfócitos T/imunologia , Linfócitos T/imunologia , Telômero/virologia , Viremia/tratamento farmacológico , Adulto Jovem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA