Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 86
Filter
1.
Poult Sci ; 103(8): 103898, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38936216

ABSTRACT

Exosome-mediated horizontal and vertical transmission of subgroup J avian leukosis virus (ALV-J) in poultry flocks can lead to growth inhibition and severe immunosuppression. However, there are few reports on the early infection of chicken embryonic stem cells (cESCs) with ALV-J. In this study, we confirmed that early infection with ALV-J can accelerate the differentiation of cESCs and promote the secretion of exosomes. To investigate the modulation strategy of ALV-J in cESCs, circRNA sequencing was performed for further analysis. A total of 305 differentially expressed circRNAs (DECs) were obtained, including 71 upregulated DECs. Circ-CCDC7 was found to be the most upregulated DEC and was assessed by qRT-PCR, with the result consistent with the result of circRNA-seq. Based on qRT-PCR, gga-miR-6568-3p was found to be the target of the top 3 DECs, including circ-CCDC7, and the stem cell marker gene Pax7 was identified as the target gene of gga-miR-6568-3p. This study demonstrated that exosomal circ-CCDC7/gga-miR-6568-3p/Pax7 accelerates the differentiation of cESCs after early infection with ALV-J.


Subject(s)
Avian Leukosis Virus , Avian Leukosis , Cell Differentiation , Chickens , Exosomes , MicroRNAs , RNA, Circular , Animals , Avian Leukosis Virus/physiology , Exosomes/metabolism , Exosomes/virology , Exosomes/genetics , RNA, Circular/genetics , RNA, Circular/metabolism , Avian Leukosis/virology , MicroRNAs/genetics , MicroRNAs/metabolism , Poultry Diseases/virology , Poultry Diseases/genetics , Embryonic Stem Cells/virology , Embryonic Stem Cells/physiology , Chick Embryo , Avian Proteins/genetics , Avian Proteins/metabolism
2.
Viruses ; 12(9)2020 09 11.
Article in English | MEDLINE | ID: mdl-32932986

ABSTRACT

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.


Subject(s)
Carrier Proteins/metabolism , Embryonic Stem Cells/metabolism , Epigenesis, Genetic/physiology , Gene Silencing , Retroviridae/genetics , Retroviridae/physiology , Transcription Factors/metabolism , Animals , Apoptosis Regulatory Proteins , Carrier Proteins/genetics , Embryonic Stem Cells/cytology , Embryonic Stem Cells/virology , Endogenous Retroviruses , HEK293 Cells , Histones/metabolism , Humans , Leukemia Virus, Murine , Mice , NIH 3T3 Cells , Nuclear Proteins , Protein Binding , Transcription Factors/genetics , Tripartite Motif-Containing Protein 28/metabolism
3.
Viruses ; 12(9)2020 09 08.
Article in English | MEDLINE | ID: mdl-32911874

ABSTRACT

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.


Subject(s)
Betacoronavirus/physiology , Neurons/virology , Organoids/virology , Prosencephalon/virology , Spike Glycoprotein, Coronavirus/physiology , Angiotensin-Converting Enzyme 2 , Axons/enzymology , Cell Differentiation , Cells, Cultured , Cerebral Cortex/cytology , Embryonic Stem Cells/virology , HEK293 Cells , Humans , Nerve Tissue Proteins/physiology , Neural Stem Cells/enzymology , Neural Stem Cells/virology , Neurons/enzymology , Peptidyl-Dipeptidase A/physiology , Prosencephalon/cytology , Receptors, Virus/physiology , SARS-CoV-2 , Viral Load , Viral Tropism , Virus Internalization
4.
Virus Res ; 270: 197646, 2019 09.
Article in English | MEDLINE | ID: mdl-31260705

ABSTRACT

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.


Subject(s)
Cytomegalovirus/genetics , Golgi Apparatus , Protein Transport , Viral Proteins/genetics , Virus Latency/genetics , Amino Acid Motifs , Cytomegalovirus Infections/virology , Embryonic Stem Cells/virology , Genes, Immediate-Early , Humans , Multidrug Resistance-Associated Proteins/genetics , Mutation , THP-1 Cells
5.
Genes (Basel) ; 10(3)2019 03 20.
Article in English | MEDLINE | ID: mdl-30897824

ABSTRACT

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.


Subject(s)
Embryonic Stem Cells/virology , Gene Expression Profiling/veterinary , Gene Regulatory Networks , Poxviridae/immunology , Animals , Cells, Cultured , Chick Embryo , Embryonic Stem Cells/cytology , Embryonic Stem Cells/immunology , Gene Expression Regulation , Interferon Type I/immunology , Poxviridae/classification , Sequence Analysis, RNA/veterinary , Species Specificity , Viral Vaccines/classification , Viral Vaccines/immunology
6.
Semin Cancer Biol ; 53: 17-30, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30317035

ABSTRACT

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.


Subject(s)
Endogenous Retroviruses/genetics , Neoplasms/genetics , Neoplastic Stem Cells/metabolism , Virus Activation/genetics , Cell Transformation, Neoplastic/genetics , Embryonic Stem Cells/metabolism , Embryonic Stem Cells/virology , Gene Expression Regulation, Neoplastic , Gene Expression Regulation, Viral , Humans , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/virology , Neoplasms/pathology , Neoplasms/virology
7.
PLoS One ; 13(7): e0200086, 2018.
Article in English | MEDLINE | ID: mdl-29985932

ABSTRACT

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.


Subject(s)
Cytopathogenic Effect, Viral , Trophoblasts/virology , Zika Virus Infection/virology , Zika Virus/genetics , Zika Virus/pathogenicity , Animals , Cell Culture Techniques , Cell Line, Tumor , Chlorocebus aethiops , Embryonic Stem Cells/virology , Humans , Species Specificity , Vero Cells , Virus Replication
8.
Dev Cell ; 44(6): 752-761.e4, 2018 03 26.
Article in English | MEDLINE | ID: mdl-29587145

ABSTRACT

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.


Subject(s)
Cell Lineage , Embryonic Stem Cells/cytology , Influenza A Virus, H1N1 Subtype/physiology , Lung/cytology , Phosphoproteins/physiology , Respiratory Mucosa/cytology , Stem Cells/cytology , Trans-Activators/physiology , Animals , Cell Differentiation , Cells, Cultured , Embryonic Stem Cells/metabolism , Embryonic Stem Cells/virology , Female , Lung/metabolism , Lung/virology , Male , Mice , Mice, Knockout , Respiratory Mucosa/metabolism , Respiratory Mucosa/virology , Stem Cells/metabolism , Stem Cells/virology , Trachea/cytology , Trachea/metabolism , Trachea/virology
9.
Dev Comp Immunol ; 84: 307-314, 2018 07.
Article in English | MEDLINE | ID: mdl-29524446

ABSTRACT

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.


Subject(s)
Antiviral Agents/metabolism , Embryonic Stem Cells/physiology , Fish Diseases/immunology , Rhabdoviridae Infections/immunology , Rhabdoviridae/immunology , Yeasts/metabolism , Zebrafish/immunology , Zymosan/metabolism , beta-Glucans/metabolism , Animals , Cell Line , Cytokines/genetics , Cytokines/metabolism , Embryonic Stem Cells/virology , Fish Proteins/metabolism , Gene Expression Regulation , Immunity, Innate/genetics , Signal Transduction
10.
Cell Rep ; 21(2): 517-532, 2017 Oct 10.
Article in English | MEDLINE | ID: mdl-29020636

ABSTRACT

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.


Subject(s)
Anti-Retroviral Agents/pharmacology , Cerebral Cortex/cytology , Drug Evaluation, Preclinical/methods , Organoids/virology , Primary Cell Culture/methods , Zika Virus/drug effects , Cell Line , Cerebral Cortex/virology , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Embryonic Stem Cells/virology , Humans , Neurons/cytology , Neurons/metabolism , Neurons/virology , Organoids/cytology , Organoids/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , STAT3 Transcription Factor/metabolism , c-Mer Tyrosine Kinase/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL