Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters










Publication year range
1.
Proc Biol Sci ; 291(2023): 20240537, 2024 May.
Article in English | MEDLINE | ID: mdl-38747705

ABSTRACT

The noasaurid ceratosaur Kiyacursor longipes gen. et sp. nov. is described based on a fragmentary skeleton including cervical vertebra, pectoral girdle, humerus and hind limbs from the Lower Cretaceous (Aptian) Ilek Formation at Shestakovo 1 locality in Western Siberia, Russia. This is the first ceratosaur from the Early Cretaceous of Asia, extending the stratigraphic range of Ceratosauria by 40 Myr on that continent. Kiyacursor shares unique hind limb proportions with Elaphrosaurus and Limusaurus, suggesting improved cursorial ability. These taxa show an ostrich-like specialization of the pes, with a large third metatarsal and greatly reduced second metatarsal. By contrast, all other fast running non-avian theropod dinosaurs have an arctometatarsalian pes, with the third metatarsal strongly reduced proximally. The new taxon lived in the Early Cretaceous ecosystem containing a number of other Jurassic relics, such as stem salamanders, protosuchian and shartegosuchid crocodyliforms, tritylodontid synapsids and docodontan mammaliaforms.


Subject(s)
Dinosaurs , Fossils , Animals , Dinosaurs/anatomy & histology , Dinosaurs/classification , Fossils/anatomy & histology , Siberia , Biological Evolution
2.
Nat Microbiol ; 9(6): 1540-1554, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38806670

ABSTRACT

Epstein-Barr virus (EBV) is an aetiologic risk factor for the development of multiple sclerosis (MS). However, the role of EBV-infected B cells in the immunopathology of MS is not well understood. Here we characterized spontaneous lymphoblastoid cell lines (SLCLs) isolated from MS patients and healthy controls (HC) ex vivo to study EBV and host gene expression in the context of an individual's endogenous EBV. SLCLs derived from MS patient B cells during active disease had higher EBV lytic gene expression than SLCLs from MS patients with stable disease or HCs. Host gene expression analysis revealed activation of pathways associated with hypercytokinemia and interferon signalling in MS SLCLs and upregulation of forkhead box protein 1 (FOXP1), which contributes to EBV lytic gene expression. We demonstrate that antiviral approaches targeting EBV replication decreased cytokine production and autologous CD4+ T cell responses in this ex vivo model. These data suggest that dysregulation of intrinsic B cell control of EBV gene expression drives a pro-inflammatory, pathogenic B cell phenotype that can be attenuated by suppressing EBV lytic gene expression.


Subject(s)
B-Lymphocytes , Epstein-Barr Virus Infections , Herpesvirus 4, Human , Multiple Sclerosis , Humans , Herpesvirus 4, Human/genetics , Multiple Sclerosis/virology , Multiple Sclerosis/immunology , Multiple Sclerosis/genetics , Multiple Sclerosis/metabolism , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , B-Lymphocytes/virology , Epstein-Barr Virus Infections/virology , Epstein-Barr Virus Infections/immunology , Epstein-Barr Virus Infections/genetics , Epstein-Barr Virus Infections/complications , Cytokines/metabolism , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/virology , CD4-Positive T-Lymphocytes/metabolism , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Transcriptome , Virus Replication , Gene Expression Regulation, Viral , Cell Line , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Gene Expression Profiling , Adult , Female , Male
3.
Tumour Virus Res ; 15: 200259, 2023 06.
Article in English | MEDLINE | ID: mdl-36863485

ABSTRACT

Kaposi's Sarcoma (KS) is a heterogenous, multifocal vascular malignancy caused by the human herpesvirus 8 (HHV8), also known as Kaposi's Sarcoma-Associated Herpesvirus (KSHV). Here, we show that KS lesions express iNOS/NOS2 broadly throughout KS lesions, with enrichment in LANA positive spindle cells. The iNOS byproduct 3-nitrotyrosine is also enriched in LANA positive tumor cells and colocalizes with a fraction of LANA-nuclear bodies. We show that iNOS is highly expressed in the L1T3/mSLK tumor model of KS. iNOS expression correlated with KSHV lytic cycle gene expression, which was elevated in late-stage tumors (>4 weeks) but to a lesser degree in early stage (1 week) xenografts. Further, we show that L1T3/mSLK tumor growth is sensitive to an inhibitor of nitric oxide, L-NMMA. L-NMMA treatment reduced KSHV gene expression and perturbed cellular gene pathways relating to oxidative phosphorylation and mitochondrial dysfunction. These finding suggest that iNOS is expressed in KSHV infected endothelial-transformed tumor cells in KS, that iNOS expression depends on tumor microenvironment stress conditions, and that iNOS enzymatic activity contributes to KS tumor growth.


Subject(s)
Herpesvirus 8, Human , Sarcoma, Kaposi , Animals , Humans , Mice , Antigens, Viral/genetics , Herpesvirus 8, Human/genetics , omega-N-Methylarginine , Sarcoma, Kaposi/genetics , Tumor Microenvironment
4.
Nat Commun ; 13(1): 5033, 2022 08 26.
Article in English | MEDLINE | ID: mdl-36028493

ABSTRACT

DAXX and ATRX are tumor suppressor proteins that form a histone H3.3 chaperone complex and are frequently mutated in cancers with the alternative lengthening of telomeres (ALT). Here, we show that DAXX and ATRX knock-out (KO) U87-T cells that have acquired ALT-like features have defects in p53 chromatin binding and DNA damage response. RNA-seq analysis revealed that p53 pathway is among the most perturbed. ChIP-seq and ATAC-seq revealed a genome-wide reduction in p53 DNA-binding and corresponding loss of chromatin accessibility at many p53 response elements across the genome. Both DAXX and ATRX null cells showed a depletion of histone H3.3 and accumulation of γH2AX at many p53 sites, including subtelomeres. These findings indicate that loss of DAXX or ATRX can compromise p53 chromatin binding and p53 DNA damage response in ALT-like cells, providing a link between histone composition, chromatin accessibility and tumor suppressor function of p53.


Subject(s)
Chromatin , Histones , Co-Repressor Proteins , DNA Damage , DNA Helicases , Genes, Tumor Suppressor , Molecular Chaperones , Nuclear Proteins , Tumor Suppressor Protein p53 , X-linked Nuclear Protein
5.
J Virol ; 96(7): e0016222, 2022 04 13.
Article in English | MEDLINE | ID: mdl-35319230

ABSTRACT

Human immunodeficiency virus (HIV)-infected macrophages are long-lived cells that sustain persistent virus expression, which is both a barrier to viral eradication and contributor to neurological complications in patients despite antiretroviral therapy (ART). To better understand the regulation of HIV-1 in macrophages, we compared HIV-infected primary human monocyte-derived macrophages (MDM) to acutely infected primary CD4 T cells and Jurkat cells latently infected with HIV (JLAT 8.4). HIV genomes in MDM were actively transcribed despite enrichment with heterochromatin-associated H3K9me3 across the complete HIV genome in combination with elevated activation marks of H3K9ac and H3K27ac at the long terminal repeat (LTR). Macrophage patterns contrasted with JLAT cells, which showed conventional bivalent H3K4me3/H3K27me3, and acutely infected CD4 T cells, which showed an intermediate epigenotype. 5'-Methylcytosine (5mC) was enriched across the HIV genome in latently infected JLAT cells, while 5'-hydroxymethylcytosine (5hmC) was enriched in CD4 cells and MDMs. HIV infection induced multinucleation of MDMs along with DNA damage-associated p53 phosphorylation, as well as loss of TET2 and the nuclear redistribution of 5-hydoxymethylation. Taken together, our findings suggest that HIV induces a unique macrophage nuclear and transcriptional profile, and viral genomes are maintained in a noncanonical bivalent epigenetic state. IMPORTANCE Macrophages serve as a reservoir for long-term persistence and chronic production of HIV. We found an atypical epigenetic control of HIV in macrophages marked by heterochromatic H3K9me3 despite active viral transcription. HIV infection induced changes in macrophage nuclear morphology and epigenetic regulatory factors. These findings may identify new mechanisms to control chronic HIV expression in infected macrophages.


Subject(s)
HIV Infections , HIV-1 , Macrophages , CD4-Positive T-Lymphocytes , Epigenesis, Genetic , Genome, Viral , HIV Infections/genetics , HIV-1/genetics , Humans , Jurkat Cells , Macrophages/virology , Virus Latency/genetics , Virus Replication
6.
J Virol ; 95(13): e0008821, 2021 06 10.
Article in English | MEDLINE | ID: mdl-33883224

ABSTRACT

Epstein-Barr virus (EBV) is a human gammaherpesvirus that is causally associated with various lymphomas and carcinomas. Although EBV is not typically associated with multiple myeloma (MM), it can be found in some B-cell lines derived from MM patients. Here, we analyzed two EBV-positive MM-patient-derived cell lines, IM9 and ARH77, and found defective viral genomes and atypical viral gene expression patterns. We performed transcriptome sequencing to characterize the viral and cellular properties of the two EBV-positive cell lines, compared to the canonical MM cell line 8226. Principal-component analyses indicated that IM9 and ARH77 clustered together and distinct from 8226. Immunological Genome Project analysis designated these cells as stem cell and bone marrow derived. IM9 and ARH77 displayed atypical viral gene expression, including leaky lytic cycle gene expression with an absence of lytic DNA amplification. Genome sequencing revealed that the EBV genomes in ARH77 contain large deletions, while IM9 has copy number losses in multiple EBV loci. Both IM9 and ARH77 showed EBV genome heterogeneity, suggesting cells harboring multiple and variant viral genomes. We identified atypical high-level expression of lytic genes BLRF1 and BLRF2. We demonstrated that short hairpin RNA (shRNA) depletion of BLRF2 altered viral and host gene expression, including a reduction in lytic gene activation and DNA amplification. These findings demonstrate that aberrant viral genomes and lytic gene expression persist in rare B cells derived from MM tumors, and they suggest that EBV may contribute to the etiology of MM. IMPORTANCE EBV is an oncogenic herpesvirus, but its mechanisms of oncogenesis are not fully understood. A role for EBV in MM has not yet been established. We analyzed EBV-positive B-cell lines derived from MM patients and found that the cells harbored defective viral genomes with aberrant viral gene expression patterns and cell gene signatures for bone marrow-derived lymphoid stem cells. These findings suggest that aberrant EBV latent infection may contribute to the etiology of MM.


Subject(s)
B-Lymphocytes/virology , Defective Viruses/genetics , Gene Expression Regulation, Viral/genetics , Herpesvirus 4, Human/genetics , Herpesvirus 4, Human/isolation & purification , Multiple Myeloma/virology , Animals , Cells, Cultured , DNA Copy Number Variations/genetics , Disease Models, Animal , Gene Deletion , Genome, Viral/genetics , Humans , Mice , Mice, SCID , Oxidative Stress/physiology , RNA Interference , RNA, Small Interfering/genetics , Transcriptome/genetics , Virus Activation/genetics
7.
Sci Rep ; 11(1): 3509, 2021 02 10.
Article in English | MEDLINE | ID: mdl-33568696

ABSTRACT

Telomere dysfunction causes chromosomal instability which is associated with many cancers and age-related diseases. The non-coding telomeric repeat-containing RNA (TERRA) forms a structural and regulatory component of the telomere that is implicated in telomere maintenance and chromosomal end protection. The basic N-terminal Gly/Arg-rich (GAR) domain of telomeric repeat-binding factor 2 (TRF2) can bind TERRA but the structural basis and significance of this interaction remains poorly understood. Here, we show that TRF2 GAR recognizes G-quadruplex features of TERRA. We show that small molecules that disrupt the TERRA-TRF2 GAR complex, such as N-methyl mesoporphyrin IX (NMM) or genetic deletion of TRF2 GAR domain, result in the loss of TERRA, and the induction of γH2AX-associated telomeric DNA damage associated with decreased telomere length, and increased telomere aberrations, including telomere fragility. Taken together, our data indicates that the G-quadruplex structure of TERRA is an important recognition element for TRF2 GAR domain and this interaction between TRF2 GAR and TERRA is essential to maintain telomere stability.


Subject(s)
DNA Damage/genetics , RNA/genetics , Telomere/metabolism , Telomeric Repeat Binding Protein 2/genetics , DNA-Binding Proteins/genetics , G-Quadruplexes/drug effects , Humans , Protein Binding/genetics , RNA/metabolism , RNA, Long Noncoding/genetics , Telomeric Repeat Binding Protein 2/metabolism , Transcription Factors/genetics
9.
PLoS Pathog ; 17(1): e1009231, 2021 01.
Article in English | MEDLINE | ID: mdl-33471863

ABSTRACT

Liquid-liquid phase separation (LLPS) can drive formation of diverse and essential macromolecular structures, including those specified by viruses. Kaposi's Sarcoma-Associated Herpesvirus (KSHV) genomes associate with the viral encoded Latency-Associated Nuclear Antigen (LANA) to form stable nuclear bodies (NBs) during latent infection. Here, we show that LANA-NB formation and KSHV genome conformation involves LLPS. Using LLPS disrupting solvents, we show that LANA-NBs are partially disrupted, while DAXX and PML foci are highly resistant. LLPS disruption altered the LANA-dependent KSHV chromosome conformation but did not stimulate lytic reactivation. We found that LANA-NBs undergo major morphological transformation during KSHV lytic reactivation to form LANA-associated replication compartments encompassing KSHV DNA. DAXX colocalizes with the LANA-NBs during latency but is evicted from the LANA-associated lytic replication compartments. These findings indicate the LANA-NBs are dynamic super-molecular nuclear structures that partly depend on LLPS and undergo morphological transitions corresponding to the different modes of viral replication.


Subject(s)
Antigens, Viral/chemistry , Co-Repressor Proteins/metabolism , Genome, Viral/genetics , Herpesvirus 8, Human/genetics , Intranuclear Inclusion Bodies/metabolism , Molecular Chaperones/metabolism , Nuclear Proteins/chemistry , Sarcoma, Kaposi/virology , Antigens, Viral/genetics , Cell Line, Tumor , Herpesvirus 8, Human/physiology , Histones/metabolism , Humans , Inclusion Bodies, Viral/chemistry , Inclusion Bodies, Viral/metabolism , Intranuclear Inclusion Bodies/chemistry , Latent Infection , Liquid-Liquid Extraction , Nuclear Proteins/genetics , Plasmids/genetics , Virus Latency , Virus Replication
10.
Oncotarget ; 11(46): 4224-4242, 2020 Nov 17.
Article in English | MEDLINE | ID: mdl-33245718

ABSTRACT

KSHV-associated cancers have poor prognoses and lack therapeutics that selectively target viral gene functions. We developed a screening campaign to identify known drugs that could be repurposed for the treatment of KSHV-associated cancers. We focused on primary effusion lymphoma (PEL), which has particularly poor treatment outcomes. We developed a luciferase reporter assay to test the ability of drugs to inhibit DNA binding of the KSHV LANA DNA binding domain (DBD). In parallel, we screened drugs for selective inhibition of a KSHV+ PEL cells. While potent hits were identified in each assay, only one hit, Mubritinib, was found to score in both assays. Mubritinib caused PEL cells to undergo cell cycle arrest with accumulation of sub-G1 population and Annexin V. Mubritinib inhibited LANA binding to KSHV terminal repeat (TR) DNA in KSHV+ PEL cells, but did not lead to KSHV lytic cycle reactivation. Mubritinib was originally identified as a receptor tyrosine kinase (RTK) inhibitor selective for HER2/ErbB2. But recent studies have revealed that Mubritinib can also inhibit the electron transport chain (ETC) complex at nanomolar concentrations. We found that other related ETC complex inhibitors (Rotenone and Deguelin) exhibited PEL cell growth inhibition while RTK inhibitors failed. Seahorse analysis demonstrated that Mubritinib selectively inhibits the maximal oxygen consumption (OCR) in PEL cells and metabolomics revealed changes in ATP/ADP and ATP/AMP ratios. These findings indicate that PEL cells are selectively sensitive to ETC complex inhibitors and provide a rationale for repurposing Mubritinib for selective treatment of PEL.

11.
Cell Rep ; 33(6): 108379, 2020 11 10.
Article in English | MEDLINE | ID: mdl-33176153

ABSTRACT

The telomeric shelterin protein telomeric repeat-binding factor 2 (TRF2) recruits origin recognition complex (ORC) proteins, the foundational building blocks of DNA replication origins, to telomeres. We seek to determine whether TRF2-recruited ORC proteins give rise to functional origins in telomere repeat tracts. We find that reduction of telomeric recruitment of ORC2 by expression of an ORC interaction-defective TRF2 mutant significantly reduces telomeric initiation events in human cells. This reduction in initiation events is accompanied by telomere repeat loss, telomere aberrations and dysfunction. We demonstrate that telomeric origins are activated by induced replication stress to provide a key rescue mechanism for completing compromised telomere replication. Importantly, our studies also indicate that the chromatin remodeler SNF2H promotes telomeric initiation events by providing access for ORC2. Collectively, our findings reveal that active recruitment of ORC by TRF2 leads to formation of functional origins, providing an important mechanism for avoiding telomere dysfunction and rescuing challenged telomere replication.


Subject(s)
DNA Replication/genetics , Telomere/metabolism , Telomeric Repeat Binding Protein 2/metabolism , Genomic Instability , Humans
12.
Nat Commun ; 11(1): 877, 2020 02 13.
Article in English | MEDLINE | ID: mdl-32054837

ABSTRACT

Epstein-Barr virus (EBV) genomes persist in latently infected cells as extrachromosomal episomes that attach to host chromosomes through the tethering functions of EBNA1, a viral encoded sequence-specific DNA binding protein. Here we employ circular chromosome conformation capture (4C) analysis to identify genome-wide associations between EBV episomes and host chromosomes. We find that EBV episomes in Burkitt's lymphoma cells preferentially associate with cellular genomic sites containing EBNA1 binding sites enriched with B-cell factors EBF1 and RBP-jK, the repressive histone mark H3K9me3, and AT-rich flanking sequence. These attachment sites correspond to transcriptionally silenced genes with GO enrichment for neuronal function and protein kinase A pathways. Depletion of EBNA1 leads to a transcriptional de-repression of silenced genes and reduction in H3K9me3. EBV attachment sites in lymphoblastoid cells with different latency type show different correlations, suggesting that host chromosome attachment sites are functionally linked to latency type gene expression programs.


Subject(s)
Attachment Sites, Microbiological/genetics , Attachment Sites, Microbiological/physiology , Herpesvirus 4, Human/genetics , Herpesvirus 4, Human/physiology , Host Microbial Interactions/genetics , Burkitt Lymphoma/genetics , Burkitt Lymphoma/virology , Cell Line, Tumor , Chromosomes, Human/genetics , Chromosomes, Human/virology , Epigenesis, Genetic , Epstein-Barr Virus Nuclear Antigens/physiology , Herpesvirus 4, Human/pathogenicity , Host Microbial Interactions/physiology , Humans , Models, Biological , Plasmids/genetics , Virus Latency/genetics , Virus Latency/physiology
13.
EMBO J ; 38(19): e96659, 2019 10 01.
Article in English | MEDLINE | ID: mdl-31454099

ABSTRACT

Loss of the histone H3.3-specific chaperone component ATRX or its partner DAXX frequently occurs in human cancers that employ alternative lengthening of telomeres (ALT) for chromosomal end protection, yet the underlying mechanism remains unclear. Here, we report that ATRX/DAXX does not serve as an immediate repressive switch for ALT. Instead, ATRX or DAXX depletion gradually induces telomere DNA replication dysfunction that activates not only homology-directed DNA repair responses but also cell cycle checkpoint control. Mechanistically, we demonstrate that this process is contingent on ATRX/DAXX histone chaperone function, independently of telomere length. Combined ATAC-seq and telomere chromatin immunoprecipitation studies reveal that ATRX loss provokes progressive telomere decondensation that culminates in the inception of persistent telomere replication dysfunction. We further show that endogenous telomerase activity cannot overcome telomere dysfunction induced by ATRX loss, leaving telomere repair-based ALT as the only viable mechanism for telomere maintenance during immortalization. Together, these findings implicate ALT activation as an adaptive response to ATRX/DAXX loss-induced telomere replication dysfunction.


Subject(s)
Co-Repressor Proteins/genetics , Molecular Chaperones/genetics , Telomere Homeostasis , Telomere/metabolism , X-linked Nuclear Protein/genetics , Cell Line , DNA Repair , Gene Deletion , HEK293 Cells , Humans , Telomerase/metabolism
14.
Oncotarget ; 10(38): 3581-3591, 2019 Jun 04.
Article in English | MEDLINE | ID: mdl-31217894

ABSTRACT

Subtelomeric transcription and chromatin can have a significant impact on telomere repeat maintenance and chromosome stability. We have previously found that tumor suppressor protein p53 (TP53) can bind to retrotransposon-like elements in a majority of human subtelomeres to regulate TERRA transcription and telomeric histone acetylation in response to DNA damage. TP53 also prevents the accumulation of γH2AX DNA-damage signaling at telomeres. We now show that the inherited TP53 polymorphism Pro47Ser (hereafter S47), which is enriched in populations of African descent, is associated with elevated marks of telomere dysfunction. We found that human and mouse cells carrying the S47 variant show increased γH2AX DNA-damage signals at telomeres, as well as reduced TERRA transcription and subtelomeric histone acetylation in response to DNA damage stress. Cell-lines containing inducible genes for P47 or S47 versions of p53, as well mouse embryo fibroblasts (MEFs) reconstituted with human p53, showed elevated telomere-induced DNA damage foci and metaphase telomere signal loss in cells with S47. Human lymphoblastoid cell lines (LCLs) derived from individuals homozygous for S47, show increased accumulation of subtelomeric γH2AX and unstable telomere repeats in response to DNA damage relative to age matched LCLs homozygous for P47. Furthermore, LCLs with S47 had reduced replicative lifespan. These studies indicate that the naturally occurring S47 variant of p53 can affect telomeric chromatin, telomere repeat stability, and replicative capacity. We discuss the potential evolutionary significance of the S47 variant to African populations with respect to telomere regulation and the implications for inherited health disparities.

15.
PLoS Pathog ; 15(1): e1007489, 2019 01.
Article in English | MEDLINE | ID: mdl-30682185

ABSTRACT

The molecular basis for the formation of functional, higher-ordered macro-molecular domains is not completely known. The Kaposi's Sarcoma-Associated Herpesvirus (KSHV) genome forms a super-molecular domain structure during latent infection that is strictly dependent on the DNA binding of the viral nuclear antigen LANA to the viral terminal repeats (TR). LANA is known to form oligomeric structures that have been implicated in viral episome maintenance. In this study, we show that the LANA oligomerization interface is required for the formation of higher-order nuclear bodies that partially colocalize with DAXX, EZH2, H3K27me3, and ORC2 but not with PML. These nuclear bodies assemble at the periphery of condensed cellular chromosomes during mitotic cell division. We demonstrate that the LANA oligomerization interface contributes to the cooperative DNA binding at the viral TR and the recruitment of ORC to the viral episome. Oligomerization mutants failed to auto-regulate LANA/ORF73 transcription, and this correlated with the loss of a chromosome conformational DNA-loop between the TR and LANA promoter. Viral genomes with LANA oligomerization mutants were subject to genome rearrangements including the loss of subgenomic DNA. Our data suggests that LANA oligomerization drives stable binding to the TR and formation of an epigenetically stable chromatin architecture resulting in higher-order LANA nuclear bodies important for viral genome integrity and long-term episome persistence.


Subject(s)
Antigens, Viral/metabolism , Herpesvirus 8, Human/metabolism , Herpesvirus 8, Human/physiology , Nuclear Proteins/metabolism , Adaptor Proteins, Signal Transducing , Antigens, Viral/genetics , Cell Line , Cell Nucleus/metabolism , Chromatin/metabolism , Chromosomes/metabolism , Co-Repressor Proteins , DNA Replication , DNA, Viral/genetics , Enhancer of Zeste Homolog 2 Protein , Genome, Viral , Herpesvirus 8, Human/genetics , Humans , Intranuclear Inclusion Bodies/metabolism , Molecular Chaperones , Nuclear Proteins/genetics , Origin Recognition Complex , Terminal Repeat Sequences , Virus Latency/genetics
16.
Nat Commun ; 8(1): 2114, 2017 12 13.
Article in English | MEDLINE | ID: mdl-29235471

ABSTRACT

Telomere repeat DNA forms a nucleo-protein structure that can obstruct chromosomal DNA replication, especially under conditions of replication stress. Transcription of telomere repeats can initiate at subtelomeric CTCF-binding sites to generate telomere repeat-encoding RNA (TERRA), but the role of transcription, CTCF, and TERRA in telomere replication is not known. Here, we have used CRISPR/Cas9 gene editing to mutate CTCF-binding sites at the putative start site of TERRA transcripts for a class of subtelomeres. Under replication stress, telomeres lacking CTCF-driven TERRA exhibit sister-telomere loss and upon entry into mitosis, exhibit the formation of ultra-fine anaphase bridges and micronuclei. Importantly, these phenotypes could be rescued by the forced transcription of TERRA independent of CTCF binding. Our findings indicate that subtelomeric CTCF facilitates telomeric DNA replication by promoting TERRA transcription. Our findings also demonstrate that CTCF-driven TERRA transcription acts in cis to facilitate telomere repeat replication and chromosome stability.


Subject(s)
CCCTC-Binding Factor/genetics , DNA Replication , DNA-Binding Proteins/genetics , Telomere/genetics , Transcription Factors/genetics , Transcription, Genetic , Base Sequence , CCCTC-Binding Factor/metabolism , CRISPR-Cas Systems , DNA-Binding Proteins/metabolism , Gene Editing/methods , HCT116 Cells , Humans , Mutation , Protein Binding , Telomere/metabolism , Transcription Factors/metabolism
17.
Sci Rep ; 7: 39861, 2017 01 03.
Article in English | MEDLINE | ID: mdl-28045091

ABSTRACT

CTCF is an essential chromatin regulator implicated in important nuclear processes including in nuclear organization and transcription. Herpes Simplex Virus-1 (HSV-1) is a ubiquitous human pathogen, which enters productive infection in human epithelial and many other cell types. CTCF is known to bind several sites in the HSV-1 genome during latency and reactivation, but its function has not been defined. Here, we report that CTCF interacts extensively with the HSV-1 DNA during lytic infection by ChIP-seq, and its knockdown results in the reduction of viral transcription, viral genome copy number and virus yield. CTCF knockdown led to increased H3K9me3 and H3K27me3, and a reduction of RNA pol II occupancy on viral genes. Importantly, ChIP-seq analysis revealed that there is a higher level of CTD Ser2P modified RNA Pol II near CTCF peaks relative to the Ser5P form in the viral genome. Consistent with this, CTCF knockdown reduced the Ser2P but increased Ser5P modified forms of RNA Pol II on viral genes. These results suggest that CTCF promotes HSV-1 lytic transcription by facilitating the elongation of RNA Pol II and preventing silenced chromatin on the viral genome.


Subject(s)
CCCTC-Binding Factor/metabolism , Genome, Viral , Herpesvirus 1, Human/genetics , RNA, Viral/genetics , Virus Replication , Animals , Chlorocebus aethiops , Chromatin Assembly and Disassembly , HEK293 Cells , HeLa Cells , Herpesvirus 1, Human/metabolism , Herpesvirus 1, Human/pathogenicity , Histones/genetics , Histones/metabolism , Humans , Protein Binding , RNA Polymerase II/metabolism , RNA, Viral/metabolism , Transcription, Genetic , Vero Cells
18.
Nat Commun ; 7: 12707, 2016 09 01.
Article in English | MEDLINE | ID: mdl-27581705

ABSTRACT

The histone H3.3 chaperone DAXX is implicated in formation of heterochromatin and transcription silencing, especially for newly infecting DNA virus genomes entering the nucleus. Epstein-Barr virus (EBV) can efficiently establish stable latent infection as a chromatinized episome in the nucleus of infected cells. The EBV tegument BNRF1 is a DAXX-interacting protein required for the establishment of selective viral gene expression during latency. Here we report the structure of BNRF1 DAXX-interaction domain (DID) in complex with DAXX histone-binding domain (HBD) and histones H3.3-H4. BNRF1 DID contacts DAXX HBD and histones through non-conserved loops. The BNRF1-DAXX interface is responsible for BNRF1 localization to PML-nuclear bodies typically associated with host-antiviral resistance and transcriptional repression. Paradoxically, the interface is also required for selective transcription activation of viral latent cycle genes required for driving B-cell proliferation. These findings reveal molecular details of virus reprogramming of an antiviral histone chaperone to promote viral latency and cellular immortalization.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Herpesvirus 4, Human/genetics , Histone Chaperones/metabolism , Histones/metabolism , Nuclear Proteins/metabolism , Viral Envelope Proteins/metabolism , B-Lymphocytes/immunology , Cell Line , Cell Nucleus/metabolism , Cell Proliferation/genetics , Chromatin Assembly and Disassembly/genetics , Co-Repressor Proteins , HEK293 Cells , Humans , Molecular Chaperones , Protein Binding/genetics , Protein Domains , Virus Latency/genetics
19.
EMBO J ; 35(2): 193-207, 2016 Jan 18.
Article in English | MEDLINE | ID: mdl-26658110

ABSTRACT

Telomeres and tumor suppressor protein TP53 (p53) function in genome protection, but a direct role of p53 at telomeres has not yet been described. Here, we have identified non-canonical p53-binding sites within the human subtelomeres that suppress the accumulation of DNA damage at telomeric repeat DNA. These non-canonical subtelomeric p53-binding sites conferred transcription enhancer-like functions that include an increase in local histone H3K9 and H3K27 acetylation and stimulation of subtelomeric transcripts, including telomere repeat-containing RNA (TERRA). p53 suppressed formation of telomere-associated γH2AX and prevented telomere DNA degradation in response to DNA damage stress. Our findings indicate that p53 provides a direct chromatin-associated protection to human telomeres, as well as other fragile genomic sites. We propose that p53-associated chromatin modifications enhance local DNA repair or protection to provide a previously unrecognized tumor suppressor function of p53.


Subject(s)
Carrier Proteins/metabolism , DNA Damage/genetics , Telomere/metabolism , Tumor Suppressor Protein p53/metabolism , Carrier Proteins/genetics , HCT116 Cells , Humans , Protein Binding , Telomere/genetics , Tumor Suppressor Protein p53/genetics
20.
Cell Rep ; 9(6): 2263-78, 2014 Dec 24.
Article in English | MEDLINE | ID: mdl-25497088

ABSTRACT

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.


Subject(s)
Herpesvirus 1, Human/physiology , Telomere/virology , Virus Replication , Cell Line , Chromosome Aberrations , DNA Damage , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Dipeptidyl-Peptidases and Tripeptidyl-Peptidases/genetics , Dipeptidyl-Peptidases and Tripeptidyl-Peptidases/metabolism , Herpesvirus 1, Human/metabolism , Humans , Immediate-Early Proteins/genetics , Immediate-Early Proteins/metabolism , Proteolysis , RNA, Untranslated/genetics , RNA, Untranslated/metabolism , Repetitive Sequences, Nucleic Acid , Serine Proteases/genetics , Serine Proteases/metabolism , Shelterin Complex/metabolism , Telomere/chemistry , Telomere/genetics , Telomere-Binding Proteins/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Viral Proteins/genetics , Viral Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...