Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 85
Filtrar
1.
Viruses ; 12(9)2020 09 11.
Artigo em Inglês | MEDLINE | ID: mdl-32932986

RESUMO

Embryonic stem cells (ESC) have the ability to epigenetically silence endogenous and exogenous retroviral sequences. Trim28 plays an important role in establishing this silencing, but less is known about the role other Trim proteins play. The Tif1 family is a sub-group of the Trim family, which possess histone binding ability in addition to the distinctive RING domain. Here, we have examined the interaction between three Tif1 family members, namely Trim24, Trim28 and Trim33, and their function in retroviral silencing. We identify a complex formed in ESC, comprised of these three proteins. We further show that when Trim33 is depleted, the complex collapses and silencing efficiency of both endogenous and exogenous sequences is reduced. Similar transcriptional activation takes place when Trim24 is depleted. Analysis of the H3K9me3 chromatin modification showed a decrease in this repressive mark, following both Trim24 and Trim33 depletion. As Trim28 is an identified binding partner of the H3K9 methyltransferase ESET, this further supports the involvement of Trim28 in the complex. The results presented here suggest that a complex of Tif1 family members, each of which possesses different specificity and efficiency, contributes to the silencing of retroviral sequences in ESC.


Assuntos
Proteínas de Transporte/metabolismo , Células-Tronco Embrionárias/metabolismo , Epigênese Genética/fisiologia , Inativação Gênica , Retroviridae/genética , Retroviridae/fisiologia , Fatores de Transcrição/metabolismo , Animais , Proteínas Reguladoras de Apoptose , Proteínas de Transporte/genética , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/virologia , Retrovirus Endógenos , Células HEK293 , Histonas/metabolismo , Humanos , Vírus da Leucemia Murina , Camundongos , Células NIH 3T3 , Proteínas Nucleares , Ligação Proteica , Fatores de Transcrição/genética , Proteína 28 com Motivo Tripartido/metabolismo
2.
Viruses ; 12(9)2020 09 08.
Artigo em Inglês | MEDLINE | ID: mdl-32911874

RESUMO

Since the global outbreak of SARS-CoV-2 (COVID-19), infections of diverse human organs along with multiple symptoms continue to be reported. However, the susceptibility of the brain to SARS-CoV-2, and the mechanisms underlying neurological infection are still elusive. Here, we utilized human embryonic stem cell-derived brain organoids and monolayer cortical neurons to investigate infection of brain with pseudotyped SARS-CoV-2 viral particles. Spike-containing SARS-CoV-2 pseudovirus infected neural layers within brain organoids. The expression of ACE2, a host cell receptor for SARS-CoV-2, was sustained during the development of brain organoids, especially in the somas of mature neurons, while remaining rare in neural stem cells. However, pseudotyped SARS-CoV-2 was observed in the axon of neurons, which lack ACE2. Neural infectivity of SARS-CoV-2 pseudovirus did not increase in proportion to viral load, but only 10% of neurons were infected. Our findings demonstrate that brain organoids provide a useful model for investigating SARS-CoV-2 entry into the human brain and elucidating the susceptibility of the brain to SARS-CoV-2.


Assuntos
Betacoronavirus/fisiologia , Neurônios/virologia , Organoides/virologia , Prosencéfalo/virologia , Glicoproteína da Espícula de Coronavírus/fisiologia , Enzima de Conversão de Angiotensina 2 , Axônios/enzimologia , Diferenciação Celular , Células Cultivadas , Córtex Cerebral/citologia , Células-Tronco Embrionárias/virologia , Células HEK293 , Humanos , Proteínas do Tecido Nervoso/fisiologia , Células-Tronco Neurais/enzimologia , Células-Tronco Neurais/virologia , Neurônios/enzimologia , Peptidil Dipeptidase A/fisiologia , Prosencéfalo/citologia , Receptores Virais/fisiologia , SARS-CoV-2 , Carga Viral , Tropismo Viral , Internalização do Vírus
3.
Virus Res ; 270: 197646, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31260705

RESUMO

Human cytomegalovirus (HCMV) establishes latency within incompletely differentiated cells of the myeloid lineage. The viral protein UL138 participates in establishing and maintaining this latent state. UL138 has multiple functions during latency that include silencing productive phase viral gene transcription and modulating intracellular protein trafficking. Trafficking and subsequent downregulation of the multidrug resistance-associated protein 1 (MRP1) by UL138 is mediated by one of four Golgi sorting motifs within UL138. Here we investigate whether any of the Golgi sorting motifs of UL138 are required for the establishment and/or maintenance of HCMV latency in model cell systems in vitro. We determined that a mutant UL138 protein lacking an acidic cluster dileucine sorting motif unable to downregulate MRP1, as well as another mutant lacking all four Golgi sorting motifs still silenced viral immediate early (IE) gene expression and prevented progeny virion formation during latency. We conclude that the Golgi sorting motifs are not required for latency establishment or maintenance in model cell systems in vitro.


Assuntos
Citomegalovirus/genética , Complexo de Golgi , Transporte Proteico , Proteínas Virais/genética , Latência Viral/genética , Motivos de Aminoácidos , Infecções por Citomegalovirus/virologia , Células-Tronco Embrionárias/virologia , Genes Precoces , Humanos , Proteínas Associadas à Resistência a Múltiplos Medicamentos/genética , Mutação , Células THP-1
4.
Genes (Basel) ; 10(3)2019 03 20.
Artigo em Inglês | MEDLINE | ID: mdl-30897824

RESUMO

The discovery of mammalian pluripotent embryonic stem cells (ESC) has revolutionised cell research and regenerative medicine. More recently discovered chicken ESC (cESC), though less intensively studied, are increasingly popular as vaccine substrates due to a dearth of avian cell lines. Information on the comparative performance of cESC with common vaccine viruses is limited. Using RNA-sequencing, we compared cESC transcriptional programmes elicited by stimulation with chicken type I interferon or infection with vaccine viruses routinely propagated in primary chicken embryo fibroblasts (CEF). We used poxviruses (fowlpox virus (FWPV) FP9, canarypox virus (CNPV), and modified vaccinia virus Ankara (MVA)) and a birnavirus (infectious bursal disease virus (IBDV) PBG98). Interferon-stimulated genes (ISGs) were induced in cESC to levels comparable to those in CEF and immortalised chicken fibroblast DF-1 cells. cESC are permissive (with distinct host transcriptional responses) to MVA, FP9, and CNPV but, surprisingly, not to PBG98. MVA, CNPV, and FP9 suppressed innate immune responses, while PBG98 induced a subset of ISGs. Dysregulation of signalling pathways (i.e., NFκB, TRAF) was observed, which might affect immune responses and viral replication. In conclusion, we show that cESC are an attractive alternative substrate to study and propagate poxvirus recombinant vaccine vectors.


Assuntos
Células-Tronco Embrionárias/virologia , Perfilação da Expressão Gênica/veterinária , Redes Reguladoras de Genes , Poxviridae/imunologia , Animais , Células Cultivadas , Embrião de Galinha , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/imunologia , Regulação da Expressão Gênica , Interferon Tipo I/imunologia , Poxviridae/classificação , Análise de Sequência de RNA/veterinária , Especificidade da Espécie , Vacinas Virais/classificação , Vacinas Virais/imunologia
5.
Semin Cancer Biol ; 53: 17-30, 2018 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-30317035

RESUMO

Cancer incidence and mortality, metastasis, drug resistance and recurrence are still the critical issues of oncological diseases. In this scenario, increasing scientific evidences demonstrate that the activation of human endogenous retroviruses (HERVs) is involved in the aggressiveness of tumors such as melanoma, breast, germ cell, renal, ovarian, liver and haematological cancers. In their dynamic regulation, HERVs have also proved to be important determinants of pluripotency in human embryonic stem cells (ESC) and of the reprogramming process of induced pluripotent stem cells (iPSCs). In many types of tumors, essential characteristics of aggressiveness have been associated with the achievement of stemness features, often accompanied with the identification of defined subpopulations, termed cancer stem cells (CSCs), which possess stem cell-like properties and sustain tumorigenesis. Indeed, CSCs show high self-renewal capacity with a peculiar potential in tumor initiation, progression, metastasis, heterogeneity, recurrence, radiotherapy and drug resistance. However, HERVs role in CSCs biology is still not fully elucidated. In this regard, CD133 is a widely recognized marker of CSCs, and our group demonstrated, for the first time, the requirement of HERV-K activation to expand and maintain a CD133+ melanoma cell subpopulation with stemness features in response to microenvironmental modifications. The review will discuss HERVs expression as cancer hallmark, with particular focus on their role in the regulation of cancer stemness features and the potential involvement as targets for therapy.


Assuntos
Retrovirus Endógenos/genética , Neoplasias/genética , Células-Tronco Neoplásicas/metabolismo , Ativação Viral/genética , Transformação Celular Neoplásica/genética , Células-Tronco Embrionárias/metabolismo , Células-Tronco Embrionárias/virologia , Regulação Neoplásica da Expressão Gênica , Regulação Viral da Expressão Gênica , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Células-Tronco Pluripotentes Induzidas/virologia , Neoplasias/patologia , Neoplasias/virologia
6.
PLoS One ; 13(7): e0200086, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29985932

RESUMO

Zika virus (ZIKV) drew worldwide attention when a recent epidemic was linked to fetal microcephaly. Here we used human embryonic stem cell derived trophoblasts as a model for primitive placental trophoblast to test the hypothesis that there are differences in how the two genetically distinct ZIKV lineages, African (AF) and Asian (AS), target the human placenta. Upon infection with three AF (ib-H30656, SEN/1984/41525-DAK, and MR-766) and three AS (FSS13025, MexI-44, and PANcdc259249) ZIKV strains, we observed that severe placental cell lysis was only induced after infection with AF strains, while viral replication rates remained similar between both lineages. Differences in cytopathic effects (CPE) were not observed in Vero cells, indicating that the AF strains were not inherently superior at cell lysis. Taken together, we propose that infection with AF strains of ZIKV early in pregnancy would likely result in pregnancy loss, rather than allow further fetal development with accompanying brain damage. Our results also suggest that the long term laboratory-adapted MR-766 strain does not behave aberrantly in cell culture relative to other AF lineage strains.


Assuntos
Efeito Citopatogênico Viral , Trofoblastos/virologia , Infecção por Zika virus/virologia , Zika virus/genética , Zika virus/patogenicidade , Animais , Técnicas de Cultura de Células , Linhagem Celular Tumoral , Chlorocebus aethiops , Células-Tronco Embrionárias/virologia , Humanos , Especificidade da Espécie , Células Vero , Replicação Viral
7.
Dev Comp Immunol ; 84: 307-314, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29524446

RESUMO

The antiviral effects of beta-glucan, an immunostimulatory agent were studied in zebrafish both in vitro and in vivo. Here we show that zebrafish ZF4 cells as well as whole fish primed with yeast ß-glucan zymosan exhibited increased cytokine expression and elevated response to spring viremia of carp virus (SVCV) infection. In vitro, previous treatment of ß-glucan enhanced ZF4 cell viability against SVCV infection which is associated to the activation of interferon signaling pathway and inflammatory cytokines gene expression. In vivo, the SVCV-infected fish primed with ß-glucan had a higher survival rate (≈73%) than the control SVCV-infected group (≈33%). Additionally, up-regulation of the expression of a set of genes involved in innate immune response was detected in zebrafish intraperitoneally injected of ß-glucan: il1b, il6, il8, il10 and tnfa transcripts showed increased expression that appear to be rapid (2 days) but not long-lived (less than 2 weeks). The present study is, to our knowledge, the first to combine cell culture and in vivo approaches to describe host response to ß-glucan stimulation and viral infection in zebrafish.


Assuntos
Antivirais/metabolismo , Células-Tronco Embrionárias/fisiologia , Doenças dos Peixes/imunologia , Infecções por Rhabdoviridae/imunologia , Rhabdoviridae/imunologia , Leveduras/metabolismo , Peixe-Zebra/imunologia , Zimosan/metabolismo , beta-Glucanas/metabolismo , Animais , Linhagem Celular , Citocinas/genética , Citocinas/metabolismo , Células-Tronco Embrionárias/virologia , Proteínas de Peixes/metabolismo , Regulação da Expressão Gênica , Imunidade Inata/genética , Transdução de Sinais
8.
Dev Cell ; 44(6): 752-761.e4, 2018 03 26.
Artigo em Inglês | MEDLINE | ID: mdl-29587145

RESUMO

Basal cells (BCs) are p63-expressing multipotent progenitors of skin, tracheoesophageal and urinary tracts. p63 is abundant in developing airways; however, it remains largely unclear how embryonic p63+ cells contribute to the developing and postnatal respiratory tract epithelium, and ultimately how they relate to adult BCs. Using lineage-tracing and functional approaches in vivo, we show that p63+ cells arising from the lung primordium are initially multipotent progenitors of airway and alveolar lineages but later become restricted proximally to generate the tracheal adult stem cell pool. In intrapulmonary airways, these cells are maintained immature to adulthood in bronchi, establishing a rare p63+Krt5- progenitor cell population that responds to H1N1 virus-induced severe injury. Intriguingly, this pool includes a CC10 lineage-labeled p63+Krt5- cell subpopulation required for a full H1N1-response. These data elucidate key aspects in the establishment of regionally distinct adult stem cell pools in the respiratory system, potentially with relevance to other organs.


Assuntos
Linhagem da Célula , Células-Tronco Embrionárias/citologia , Vírus da Influenza A Subtipo H1N1/fisiologia , Pulmão/citologia , Fosfoproteínas/fisiologia , Mucosa Respiratória/citologia , Células-Tronco/citologia , Transativadores/fisiologia , Animais , Diferenciação Celular , Células Cultivadas , Células-Tronco Embrionárias/metabolismo , Células-Tronco Embrionárias/virologia , Feminino , Pulmão/metabolismo , Pulmão/virologia , Masculino , Camundongos , Camundongos Knockout , Mucosa Respiratória/metabolismo , Mucosa Respiratória/virologia , Células-Tronco/metabolismo , Células-Tronco/virologia , Traqueia/citologia , Traqueia/metabolismo , Traqueia/virologia
9.
Cell Rep ; 21(2): 517-532, 2017 Oct 10.
Artigo em Inglês | MEDLINE | ID: mdl-29020636

RESUMO

The human cerebral cortex possesses distinct structural and functional features that are not found in the lower species traditionally used to model brain development and disease. Accordingly, considerable attention has been placed on the development of methods to direct pluripotent stem cells to form human brain-like structures termed organoids. However, many organoid differentiation protocols are inefficient and display marked variability in their ability to recapitulate the three-dimensional architecture and course of neurogenesis in the developing human brain. Here, we describe optimized organoid culture methods that efficiently and reliably produce cortical and basal ganglia structures similar to those in the human fetal brain in vivo. Neurons within the organoids are functional and exhibit network-like activities. We further demonstrate the utility of this organoid system for modeling the teratogenic effects of Zika virus on the developing brain and identifying more susceptibility receptors and therapeutic compounds that can mitigate its destructive actions.


Assuntos
Antirretrovirais/farmacologia , Córtex Cerebral/citologia , Avaliação Pré-Clínica de Medicamentos/métodos , Organoides/virologia , Cultura Primária de Células/métodos , Zika virus/efeitos dos fármacos , Linhagem Celular , Córtex Cerebral/virologia , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Células-Tronco Embrionárias/virologia , Humanos , Neurônios/citologia , Neurônios/metabolismo , Neurônios/virologia , Organoides/citologia , Organoides/metabolismo , Receptores Proteína Tirosina Quinases/metabolismo , Fator de Transcrição STAT3/metabolismo , c-Mer Tirosina Quinase/metabolismo
10.
PLoS One ; 12(4): e0175259, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28406989

RESUMO

Marek's disease virus is the etiological agent of a major lymphoproliferative disorder in poultry and the prototype of the Mardivirus genus. Primary avian somatic cells are currently used for virus replication and vaccine production, but they are largely refractory to any genetic modification compatible with the preservation of intact viral susceptibility. We explored the concept of induction of viral replication permissiveness in an established pluripotent chicken embryonic stem cell-line (cES) in order to derive a new fully susceptible cell-line. Chicken ES cells were not permissive for Mardivirus infection, but as soon as differentiation was triggered, replication of Marek's disease virus was detected. From a panel of cyto-differentiating agents, hexamethylene bis (acetamide) (HMBA) was found to be the most efficient regarding the induction of permissiveness. These initial findings prompted us to analyse the effect of HMBA on gene expression, to derive a new mesenchymal cell line, the so-called ESCDL-1, and monitor its susceptibility for Mardivirus replication. All Mardiviruses tested so far replicated equally well on primary embryonic skin cells and on ESCDL-1, and the latter showed no variation related to its passage number in its permissiveness for virus infection. Viral morphogenesis studies confirmed efficient multiplication with, as in other in vitro models, no extra-cellular virus production. We could show that ESCDL-1 can be transfected to express a transgene and subsequently cloned without any loss in permissiveness. Consequently, ESCDL-1 was genetically modified to complement viral gene deletions thus yielding stable trans-complementing cell lines. We herein claim that derivation of stable differentiated cell-lines from cES cell lines might be an alternative solution to the cultivation of primary cells for virology studies.


Assuntos
Células-Tronco Embrionárias/virologia , Mardivirus/fisiologia , Replicação Viral/fisiologia , Acetamidas/farmacologia , Animais , Linhagem Celular , Embrião de Galinha , Galinhas , Células-Tronco Embrionárias/metabolismo , Doença de Marek/metabolismo , Replicação Viral/efeitos dos fármacos
11.
Proc Natl Acad Sci U S A ; 114(9): E1587-E1596, 2017 02 28.
Artigo em Inglês | MEDLINE | ID: mdl-28193876

RESUMO

Infection of pregnant women by Asian lineage strains of Zika virus (ZIKV) has been linked to brain abnormalities in their infants, yet it is uncertain when during pregnancy the human conceptus is most vulnerable to the virus. We have examined two models to study susceptibility of human placental trophoblast to ZIKV: cytotrophoblast and syncytiotrophoblast derived from placental villi at term and colonies of trophoblast differentiated from embryonic stem cells (ESC). The latter appear to be analogous to the primitive placenta formed during implantation. The cells from term placentas, which resist infection, do not express genes encoding most attachment factors implicated in ZIKV entry but do express many genes associated with antiviral defense. By contrast, the ESC-derived trophoblasts possess a wide range of attachment factors for ZIKV entry and lack components of a robust antiviral response system. These cells, particularly areas of syncytiotrophoblast within the colonies, quickly become infected, produce infectious virus and undergo lysis within 48 h after exposure to low titers (multiplicity of infection > 0.07) of an African lineage strain (MR766 Uganda: ZIKVU) considered to be benign with regards to effects on fetal development. Unexpectedly, lytic effects required significantly higher titers of the presumed more virulent FSS13025 Cambodia (ZIKVC). Our data suggest that the developing fetus might be most vulnerable to ZIKV early in the first trimester before a protective zone of mature villous trophoblast has been established. Additionally, MR766 is highly trophic toward primitive trophoblast, which may put the early conceptus of an infected mother at high risk for destruction.


Assuntos
Placenta/virologia , Trofoblastos/virologia , Infecção por Zika virus/virologia , Zika virus/patogenicidade , Camboja , Células Cultivadas , Células-Tronco Embrionárias/virologia , Feminino , Humanos , Gravidez , Primeiro Trimestre da Gravidez/fisiologia , Uganda
12.
J Hepatol ; 66(3): 494-503, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-27746336

RESUMO

BACKGROUND & AIMS: One major obstacle of hepatitis B virus (HBV) research is the lack of efficient cell culture system permissive for viral infection and replication. The aim of our study was to establish a robust HBV infection model by using hepatocyte-like cells (HLCs) derived from human pluripotent stem cells. METHODS: HLCs were differentiated from human embryonic stem cells and induced pluripotent stem cells. Maturation of hepatocyte functions was determined. After HBV infection, total viral DNA, cccDNA, total viral RNA, pgRNA, HBeAg and HBsAg were measured. RESULTS: More than 90% of the HLCs expressed strong signals of human hepatocyte markers, like albumin, as well as known host factors required for HBV infection, suggesting that these cells possessed key features of mature hepatocytes. Notably, HLCs expressed the viral receptor sodium-taurocholate cotransporting polypeptide more stably than primary human hepatocytes (PHHs). HLCs supported robust infection and some spreading of HBV. Finally, by using this model, we identified two host-targeting agents, genistin and PA452, as novel antivirals. CONCLUSIONS: Stem cell-derived HLCs fully support HBV infection. This novel HLC HBV infection model offers a unique opportunity to advance our understanding of the molecular details of the HBV life cycle; to further characterize virus-host interactions and to define new targets for HBV curative treatment. LAY SUMMARY: Our study used human pluripotent stem cells to develop hepatocyte-like cells (HLCs) capable of expressing hepatocyte markers and host factors important for HBV infection. These cells fully support HBV infection and virus-host interactions, allowing for the identification of two novel antiviral agents. Thus, stem cell-derived HLCs provide a highly physiologically relevant system to advance our understanding of viral life cycle and provide a new tool for antiviral drug screening and development.


Assuntos
Hepatite B/virologia , Hepatócitos/virologia , Células-Tronco/virologia , Antivirais/farmacologia , Diferenciação Celular , Linhagem Celular , Células Cultivadas , Descoberta de Drogas , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/virologia , Hepatite B/tratamento farmacológico , Hepatite B/patologia , Vírus da Hepatite B/patogenicidade , Vírus da Hepatite B/fisiologia , Hepatócitos/citologia , Interações Hospedeiro-Patógeno , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/virologia , Modelos Biológicos , Células-Tronco/citologia , Replicação Viral
13.
Cancer Lett ; 381(1): 176-93, 2016 10 10.
Artigo em Inglês | MEDLINE | ID: mdl-27497790

RESUMO

Somatic mutations and chromosome instability are hallmarks of genomic aberrations in cancer cells. Aneuploidies represent common manifestations of chromosome instability, which is frequently observed in human embryos and malignant solid tumors. Activation of human endogenous retroviruses (HERV)-derived loci is documented in preimplantation human embryos, hESC, and multiple types of human malignancies. It remains unknown whether the HERV activation may highlight a common molecular pathway contributing to the frequent occurrence of chromosome instability in the early stages of human embryonic development and the emergence of genomic aberrations in cancer. Single cell RNA sequencing analysis of human preimplantation embryos reveals activation of specific LTR7/HERVH loci during the transition from the oocytes to zygotes and identifies HERVH network signatures associated with the aneuploidy in human embryos. The correlation patterns' analysis links transcriptome signatures of the HERVH network activation of the in vivo matured human oocytes with gene expression profiles of clinical samples of prostate tumors supporting the existence of a cancer progression pathway from putative precursor lesions (prostatic intraepithelial neoplasia) to localized and metastatic prostate cancers. Tracking signatures of HERVH networks' activation in tumor samples from cancer patients with known long-term therapy outcomes enabled patients' stratification into sub-groups with markedly distinct likelihoods of therapy failure and death from cancer. Genome-wide analyses of human-specific genetic elements of stem cell-associated retroviruses (SCARs)-regulated networks in 12,093 clinical tumor samples across 29 cancer types revealed pan-cancer genomic signatures of clinically-lethal therapy resistant disease defined by the presence of somatic non-silent mutations (SNMs), gene-level copy number changes, and transcripts and proteins' expression of SCARs-regulated host genes. More than 73% of all cancer deaths occurred in patients whose tumors harbor the SNMs' signatures. Linear regression analysis of the cancer intractability in the United States population demonstrated that organ-specific cancer death rates are directly correlated with the percentages of patients whose tumors harbor the SNMs' signatures. Present analyses suggest that awakening of SCARs-regulated stemness networks in differentiated cells is associated with development of diverse spectrum of genomic aberrations in multiple types of clinically lethal malignant tumors contributing to emergence of therapy-resistant cancer phenotypes.


Assuntos
Blastocisto/virologia , Transformação Celular Viral/genética , Células-Tronco Embrionárias/virologia , Retrovirus Endógenos/genética , Genômica/métodos , Neoplasias/genética , Neoplasias/virologia , Células-Tronco Neoplásicas/virologia , Ativação Viral/genética , Instabilidade Cromossômica , Resistencia a Medicamentos Antineoplásicos/genética , Feminino , Perfilação da Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento , Regulação Neoplásica da Expressão Gênica , Regulação Viral da Expressão Gênica , Predisposição Genética para Doença , Estudo de Associação Genômica Ampla , Interações Hospedeiro-Patógeno , Humanos , Estimativa de Kaplan-Meier , Modelos Lineares , Masculino , Mutação , Neoplasias/patologia , Neoplasias/terapia , Células-Tronco Neoplásicas/patologia , Fenótipo , Prognóstico , Neoplasia Prostática Intraepitelial/genética , Neoplasia Prostática Intraepitelial/virologia , Neoplasias da Próstata/genética , Neoplasias da Próstata/virologia , Fatores de Risco , Sequências Repetidas Terminais
14.
Protein Cell ; 7(9): 651-61, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27430948

RESUMO

Virus infection consists of entry, synthesis of macromolecular components, virus assembly and release. Understanding of the mechanisms underlying each event is necessary for the intervention of virus infection in human healthcare and agriculture. Here we report the visualization of Singapore grouper iridovirus (SGIV) assembly in the medaka haploid embryonic stem (ES) cell line HX1. SGIV is a highly infectious DNA virus that causes a massive loss in marine aquaculture. Ectopic expression of VP88GFP, a fusion between green fluorescent protein and the envelope protein VP088, did not compromise the ES cell properties and susceptibility to SGIV infection. Although VP88GFP disperses evenly in the cytoplasm of non-infected cells, it undergoes aggregation and redistribution in SGIV-infected cells. Real-time visualization revealed multiple key stages of VP88GFP redistribution and the dynamics of viral assembly site (VAS). Specifically, VP88GFP entry into and condensation in the VAS occurred within a 6-h duration, a similar duration was observed also for the release of VP88GFP-containing SGIV out of the cell. Taken together, VP088 is an excellent marker for visualizing the SGIV infection process. Our results provide new insight into macromolecular component recruitment and SGIV assembly.


Assuntos
Células-Tronco Embrionárias/virologia , Doenças dos Peixes/virologia , Iridoviridae/fisiologia , Proteínas Virais/metabolismo , Montagem de Vírus/fisiologia , Animais , Linhagem Celular , Células-Tronco Embrionárias/metabolismo , Células-Tronco Embrionárias/patologia , Doenças dos Peixes/genética , Doenças dos Peixes/metabolismo , Humanos , Oryzias , Proteínas Virais/genética
15.
Cancer Lett ; 376(2): 347-59, 2016 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-27084523

RESUMO

Recent reports revealed consistent activation of specific endogenous retroviral elements in human preimplantation embryos and embryonic stem cells. Activity of stem cell associated retroviruses (SCARs) has been implicated in seeding thousands of human-specific regulatory sequences in the hESC genome. Activation of specific SCARs has been demonstrated in patients diagnosed with multiple types of cancer, autoimmune diseases, and neurodegenerative disorders, and appears associated with clinically lethal therapy resistant death-from-cancer phenotypes in a sub-set of cancer patients diagnosed with different types of malignant tumors. A hallmark feature of human-specific SCAR integration sites is deletions of ancestral DNA. Analysis of human-specific genetic loci of SCARs' stemness networks in tumor samples of TCGA cohorts representing 29 cancer types suggests that this approach may facilitate identification of pan-cancer genomic signatures of clinically-lethal disease defined by the presence of somatic non-silent mutations, gene-level copy number changes, and transcripts and proteins' expression of SCAR-regulated host genes. Present analyses indicate that multiple lines of strong circumstantial evidence support the hypothesis that activation of SCARs' networks may play an important role in cancer progression and metastasis, perhaps contributing to the emergence of clinically-lethal therapy-resistant death-from-cancer phenotypes.


Assuntos
Resistencia a Medicamentos Antineoplásicos , Células-Tronco Embrionárias/virologia , Neoplasias/virologia , Células-Tronco Neoplásicas/virologia , Retroviridae/patogenicidade , Ativação Viral , Animais , Antineoplásicos/uso terapêutico , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo , Linhagem da Célula , Resistencia a Medicamentos Antineoplásicos/genética , Células-Tronco Embrionárias/efeitos dos fármacos , Células-Tronco Embrionárias/metabolismo , Regulação Neoplásica da Expressão Gênica , Regulação Viral da Expressão Gênica , Interações Hospedeiro-Patógeno , Humanos , Neoplasias/tratamento farmacológico , Neoplasias/genética , Neoplasias/metabolismo , Células-Tronco Neoplásicas/efeitos dos fármacos , Células-Tronco Neoplásicas/metabolismo , Fenótipo , Prognóstico , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo , Retroviridae/genética , Retroviridae/metabolismo
16.
Antiviral Res ; 129: 67-73, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-26875788

RESUMO

Human cytomegalovirus (HCMV) is the leading cause of congenital infections. Symptomatic newborns present with a range of sequelae including disorders of the CNS such as visual impairment, microcephaly, mental retardation and hearing loss. HCMV congenital infection causes gross changes in brain morphology and disturbances in glial and neuronal distribution, number and migration. In these studies, we have evaluated the effectiveness of the antiviral maribavir in inhibiting HCMV infections of ES cell-derived neuronal progenitor cells (NPC). We used EZ-spheres generated from H9 ES cells which are pre-rosette NPCs that retain long-term potential to differentiate into diverse central and peripheral neural lineages following directed differentiation. Our results demonstrate that the maribavir disrupts HCMV replication and viral yield in undifferentiated EZ-sphere-derived NPCs. In addition, we observed that maribavir limits HCMV replication and reduces the percentage of infected cells during differentiation of NPCs. Finally, early steps in differentiation are maintained during infection by treating with maribavir, likely an indirect effect resulting from decreased viral spread. Future studies of NPC proliferation and differentiation during infection treated with maribavir could provide the impetus for studying maribavir as an antiviral agent for congenital HCMV disease.


Assuntos
Antivirais/farmacologia , Células-Tronco Neurais/efeitos dos fármacos , Células-Tronco Neurais/virologia , Neurogênese/efeitos dos fármacos , Benzimidazóis/farmacologia , Linhagem Celular , Células-Tronco Embrionárias/efeitos dos fármacos , Células-Tronco Embrionárias/virologia , Humanos , Células-Tronco Neurais/fisiologia , Neurônios/citologia , Ribonucleosídeos/farmacologia , Replicação Viral/efeitos dos fármacos
17.
APMIS ; 124(1-2): 4-10, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26818257

RESUMO

The integration of proviral DNA into host chromosomal DNA as an obligatory step in the replication cycle of retroviruses is a natural event of genetic recombination between virus and host. When integration occurs in cells of the germ line, it results in mendelian inheritance of viral sequences that we call endogenous retroviruses (ERV) and HERV for humans. HERVs and host often establish a symbiotic relationship, especially in the placenta and in pluripotent embryonic stem cells, but HERVs occasionally have deleterious consequences for the host. This special issue of APMIS features the fascinating relationships between HERV and humans in health and disease.


Assuntos
Retrovirus Endógenos/genética , Retrovirus Endógenos/fisiologia , Genoma Humano , Células-Tronco Embrionárias/fisiologia , Células-Tronco Embrionárias/virologia , Retrovirus Endógenos/patogenicidade , Feminino , Genoma Viral , Interações Hospedeiro-Patógeno , Humanos , Placenta/fisiologia , Placenta/virologia , Gravidez , RNA Viral/genética
18.
Cell ; 163(1): 230-45, 2015 Sep 24.
Artigo em Inglês | MEDLINE | ID: mdl-26365490

RESUMO

Embryonic stem cells (ESCs) repress the expression of exogenous proviruses and endogenous retroviruses (ERVs). Here, we systematically dissected the cellular factors involved in provirus repression in embryonic carcinomas (ECs) and ESCs by a genome-wide siRNA screen. Histone chaperones (Chaf1a/b), sumoylation factors (Sumo2/Ube2i/Sae1/Uba2/Senp6), and chromatin modifiers (Trim28/Eset/Atf7ip) are key determinants that establish provirus silencing. RNA-seq analysis uncovered the roles of Chaf1a/b and sumoylation modifiers in the repression of ERVs. ChIP-seq analysis demonstrates direct recruitment of Chaf1a and Sumo2 to ERVs. Chaf1a reinforces transcriptional repression via its interaction with members of the NuRD complex (Kdm1a, Hdac1/2) and Eset, while Sumo2 orchestrates the provirus repressive function of the canonical Zfp809/Trim28/Eset machinery by sumoylation of Trim28. Our study reports a genome-wide atlas of functional nodes that mediate proviral silencing in ESCs and illuminates the comprehensive, interconnected, and multi-layered genetic and epigenetic mechanisms by which ESCs repress retroviruses within the genome.


Assuntos
Células-Tronco Embrionárias/virologia , Retrovirus Endógenos/genética , Provírus/genética , Animais , Fator 1 de Modelagem da Cromatina/genética , Fator 1 de Modelagem da Cromatina/metabolismo , Células-Tronco de Carcinoma Embrionário/virologia , Epigênese Genética , Camundongos , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/metabolismo
19.
Cell ; 163(1): 30-2, 2015 Sep 24.
Artigo em Inglês | MEDLINE | ID: mdl-26406368

RESUMO

Retroviral restriction is a complex phenomenon that, despite remarkable recent progress, is far from being well understood. In this Preview, we introduce an insightful study by Yang et al. that represents the first attempt to identify the global determinants of retroviral repression in pluripotent mammalian cells.


Assuntos
Células-Tronco Embrionárias/virologia , Retrovirus Endógenos/genética , Provírus/genética , Animais
20.
PLoS Pathog ; 11(6): e1004885, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-26042814

RESUMO

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


Assuntos
Herpesvirus Humano 3/fisiologia , Células-Tronco Neurais/virologia , Neurônios/virologia , Ativação Viral/fisiologia , Latência Viral/fisiologia , Células-Tronco Embrionárias/virologia , Herpes Zoster/virologia , Humanos , Hibridização In Situ , Técnicas In Vitro , Reação em Cadeia da Polimerase
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA