Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 44
Filter
1.
RNA ; 30(8): 955-966, 2024 Jul 16.
Article in English | MEDLINE | ID: mdl-38777382

ABSTRACT

The long noncoding RNA TERRA is transcribed from telomeres in virtually all eukaryotes with linear chromosomes. In humans, TERRA transcription is driven in part by promoters comprising CpG dinucleotide-rich repeats of 29 bp repeats, believed to be present in half of the subtelomeres. Thus far, TERRA expression has been analyzed mainly using molecular biology-based approaches that only generate partial and somehow biased results. Here, we present a novel experimental pipeline to study human TERRA based on long-read sequencing (TERRA ONTseq). By applying TERRA ONTseq to different cell lines, we show that the vast majority of human telomeres produce TERRA and that the cellular levels of TERRA transcripts vary according to their chromosomes of origin. Using TERRA ONTseq, we also identified regions containing TERRA transcription start sites (TSSs) in more than half of human subtelomeres. TERRA TSS regions are generally found immediately downstream from 29 bp repeat-related sequences, which appear to be more widespread than previously estimated. Finally, we isolated a novel TERRA promoter from the highly expressed subtelomere of the long arm of Chromosome 7. With the development of TERRA ONTseq, we provide a refined picture of human TERRA biogenesis and expression and we equip the scientific community with an invaluable tool for future studies.


Subject(s)
Promoter Regions, Genetic , RNA, Long Noncoding , Telomere , Transcription Initiation Site , Transcriptome , Humans , Telomere/genetics , Telomere/metabolism , RNA, Long Noncoding/genetics , High-Throughput Nucleotide Sequencing/methods , Sequence Analysis, RNA/methods
2.
Proc Natl Acad Sci U S A ; 119(39): e2208669119, 2022 09 27.
Article in English | MEDLINE | ID: mdl-36122232

ABSTRACT

A substantial number of human cancers are telomerase-negative and elongate physiologically damaged telomeres through a break-induced replication (BIR)-based mechanism known as alternative lengthening of telomeres (ALT). We recently demonstrated that inhibiting the transcription of the telomeric long noncoding RNA TERRA suppresses telomere damage and ALT features, indicating that telomere transcription is a main trigger of ALT activity. Here we show that experimentally increased TERRA transcription not only increases ALT features, as expected, but also causes rapid loss of telomeric DNA through a pathway that requires the endonuclease Mus81. Our data indicate that the ALT mechanism can endanger telomere integrity if not properly controlled and point to TERRA transcription as a uniquely versatile target for therapy.


Subject(s)
RNA, Long Noncoding , Telomerase , DNA , Endonucleases/metabolism , Humans , Telomerase/genetics , Telomere/genetics , Telomere/metabolism
3.
Nucleic Acids Res ; 50(1): 350-367, 2022 01 11.
Article in English | MEDLINE | ID: mdl-34928380

ABSTRACT

Although the RNA helicase Upf1 has hitherto been examined mostly in relation to its cytoplasmic role in nonsense mediated mRNA decay (NMD), here we report high-throughput ChIP data indicating genome-wide association of Upf1 with active genes in Schizosaccharomyces pombe. This association is RNase sensitive, correlates with Pol II transcription and mRNA expression levels. Changes in Pol II occupancy were detected in a Upf1 deficient (upf1Δ) strain, prevalently at genes showing a high Upf1 relative to Pol II association in wild-type. Additionally, an increased Ser2 Pol II signal was detected at all highly transcribed genes examined by ChIP-qPCR. Furthermore, upf1Δ cells are hypersensitive to the transcription elongation inhibitor 6-azauracil. A significant proportion of the genes associated with Upf1 in wild-type conditions are also mis-regulated in upf1Δ. These data envisage that by operating on the nascent transcript, Upf1 might influence Pol II phosphorylation and transcription.


Subject(s)
RNA Helicases/metabolism , RNA Polymerase II/metabolism , Schizosaccharomyces pombe Proteins/metabolism , Gene Expression Regulation, Fungal , Genome, Fungal , Phosphorylation , RNA Helicases/genetics , RNA Polymerase II/genetics , Schizosaccharomyces , Schizosaccharomyces pombe Proteins/genetics , Transcriptional Activation
4.
RNA ; 27(1): 106-121, 2021 01.
Article in English | MEDLINE | ID: mdl-33127860

ABSTRACT

Telomeric repeat-containing RNA (TERRA) molecules play important roles at telomeres, from heterochromatin regulation to telomerase activity control. In human cells, TERRA is transcribed from subtelomeric promoters located on most chromosome ends and associates with telomeres. The origin of mouse TERRA molecules is, however, unclear, as transcription from the pseudoautosomal PAR locus was recently suggested to account for the vast majority of TERRA in embryonic stem cells (ESC). Here, we confirm the production of TERRA from both the chromosome 18q telomere and the PAR locus in mouse embryonic fibroblasts, ESC, and various mouse cancer and immortalized cell lines, and we identify two novel sources of TERRA on mouse chromosome 2 and X. Using various approaches, we show that PAR-TERRA molecules account for the majority of TERRA transcripts, displaying an increase of two to four orders of magnitude compared to the telomeric 18q transcript. Finally, we present a SILAC-based pull-down screen revealing a large overlap between TERRA-interacting proteins in human and mouse cells, including PRC2 complex subunits, chromatin remodeling factors, DNA replication proteins, Aurora kinases, shelterin complex subunits, Bloom helicase, Coilin, and paraspeckle proteins. Hence, despite originating from distinct genomic regions, mouse and human TERRA are likely to play similar functions in cells.


Subject(s)
Gene Expression Regulation, Neoplastic , Neoplasms/genetics , RNA, Messenger/genetics , Telomere/chemistry , Transcriptome , Animals , Aurora Kinases/genetics , Aurora Kinases/metabolism , Cell Line, Tumor , Chromosomes, Mammalian/chemistry , Chromosomes, Mammalian/metabolism , Computational Biology/methods , Fibroblasts/cytology , Fibroblasts/metabolism , Gene Regulatory Networks , Granulocyte Precursor Cells/cytology , Granulocyte Precursor Cells/metabolism , HeLa Cells , Humans , Mice , Monocytes/cytology , Monocytes/metabolism , Mouse Embryonic Stem Cells/cytology , Mouse Embryonic Stem Cells/metabolism , Neoplasms/metabolism , Neoplasms/pathology , Neurons/cytology , Neurons/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , RNA, Messenger/classification , RNA, Messenger/metabolism , RNA-Binding Proteins/classification , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , RecQ Helicases/genetics , RecQ Helicases/metabolism , Shelterin Complex , Telomere/metabolism , Telomere-Binding Proteins/genetics , Telomere-Binding Proteins/metabolism
5.
Int J Mol Sci ; 23(18)2022 Sep 09.
Article in English | MEDLINE | ID: mdl-36142374

ABSTRACT

RNA-protein interactions drive key cellular pathways such as protein translation, nuclear organization and genome stability maintenance. The human telomeric protein TRF2 binds to the long noncoding RNA TERRA through independent domains, including its N-terminal B domain. We previously demonstrated that TRF2 B domain binding to TERRA supports invasion of TERRA into telomeric double stranded DNA, leading to the formation of telomeric RNA:DNA hybrids. The other telomeric protein TRF1, which also binds to TERRA, suppresses this TRF2-associated activity by preventing TERRA-B domain interactions. Herein, we show that the binding of both TRF1 and TRF2 to TERRA depends on the ability of the latter to form G-quadruplex structures. Moreover, a cluster of arginines within the B domain is largely responsible for its binding to TERRA. On the other side, a patch of glutamates within the N-terminal A domain of TRF1 mainly accounts for the inhibition of TERRA-B domain complex formation. Finally, mouse TRF2 B domain binds to TERRA, similarly to its human counterpart, while mouse TRF1 A domain lacks the inhibitory activity. Our data shed further light on the complex crosstalk between telomeric proteins and RNAs and suggest a lack of functional conservation in mouse.


Subject(s)
RNA, Long Noncoding , Telomeric Repeat Binding Protein 1 , Telomeric Repeat Binding Protein 2 , Animals , DNA/chemistry , Glutamates , Humans , Mice , RNA, Long Noncoding/genetics , Telomere/genetics , Telomere/metabolism , Telomeric Repeat Binding Protein 1/chemistry , Telomeric Repeat Binding Protein 1/genetics , Telomeric Repeat Binding Protein 1/metabolism , Telomeric Repeat Binding Protein 2/genetics
6.
EMBO J ; 34(1): 115-29, 2015 Jan 02.
Article in English | MEDLINE | ID: mdl-25398909

ABSTRACT

The telomeric transcriptome comprises multiple long non-coding RNAs generated by transcription of linear chromosome ends. In a screening performed in Schizosaccharomyces pombe, we identified factors modulating the cellular levels of the telomeric transcriptome. Among these factors, Cay1 is the fission yeast member of the conserved family of Cactins, uncharacterized proteins crucial for cell growth and survival. In cay1∆ mutants, the cellular levels of the telomeric factor Rap1 are drastically diminished due to defects in rap1+ pre-mRNA splicing and Rap1 protein stability. cay1∆ cells accumulate histone H3 acetylated at lysine 9 at telomeres, which become transcriptionally desilenced, are over-elongated by telomerase and cause chromosomal aberrations in the cold. Overexpressing Rap1 in cay1+ deleted cells significantly reverts all telomeric defects. Additionally, cay1∆ mutants accumulate unprocessed Tf2 retrotransposon RNA through Rap1-independent mechanisms. Thus, Cay1 plays crucial roles in cells by ultimately harmonizing expression of transcripts originating from seemingly unrelated genomic loci.


Subject(s)
Chromosomes, Fungal/metabolism , Nuclear Proteins/metabolism , Schizosaccharomyces/metabolism , Telomere/metabolism , Transcription, Genetic/physiology , Chromosome Aberrations , Chromosomes, Fungal/genetics , Gene Deletion , Nuclear Proteins/genetics , Protein Stability , RNA Splicing/physiology , RNA, Fungal/genetics , RNA, Fungal/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Retroelements/physiology , Schizosaccharomyces/genetics , Schizosaccharomyces pombe Proteins/genetics , Schizosaccharomyces pombe Proteins/metabolism , Shelterin Complex , Telomere/genetics , Telomere-Binding Proteins/genetics , Telomere-Binding Proteins/metabolism
7.
J Cell Sci ; 130(4): 767-778, 2017 02 15.
Article in English | MEDLINE | ID: mdl-28062851

ABSTRACT

Cactins constitute a family of eukaryotic proteins broadly conserved from yeast to human and required for fundamental processes such as cell proliferation, genome stability maintenance, organismal development and immune response. Cactin proteins have been found to associate with the spliceosome in several model organisms, nevertheless their molecular functions await elucidation. Here we show that depletion of human cactin leads to premature sister chromatid separation, genome instability and cell proliferation arrest. Moreover, cactin is essential for efficient splicing of thousands of pre-mRNAs, and incomplete splicing of the pre-mRNA of sororin (also known as CDCA5), a cohesin-associated factor, is largely responsible for the aberrant chromatid separation in cactin-depleted cells. Lastly, cactin physically and functionally interacts with the spliceosome-associated factors DHX8 and SRRM2. We propose that cellular complexes comprising cactin, DHX8 and SRRM2 sustain precise chromosome segregation, genome stability and cell proliferation by allowing faithful splicing of specific pre-mRNAs. Our data point to novel pathways of gene expression regulation dependent on cactin, and provide an explanation for the pleiotropic dysfunctions deriving from cactin inactivation in distant eukaryotes.


Subject(s)
Carrier Proteins/metabolism , Chromatids/metabolism , DEAD-box RNA Helicases/metabolism , RNA Precursors/genetics , RNA Splicing Factors/metabolism , RNA Splicing/genetics , RNA-Binding Proteins/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Cell Cycle , Cell Cycle Proteins/metabolism , Cell Nucleus Shape , Cell Proliferation , Genomic Instability , HEK293 Cells , HeLa Cells , Humans , Introns/genetics , Protein Binding , RNA Precursors/metabolism
8.
RNA ; 23(2): 134-141, 2017 02.
Article in English | MEDLINE | ID: mdl-28096443

ABSTRACT

The binding of sequence-specific RNA-interacting proteins, such as the bacteriophage MS2 or PP7 coat proteins, to their corresponding target sequences has been extremely useful and widely used to visualize single mRNAs in vivo. However, introduction of MS2 stem-loops into yeast mRNAs has recently been shown to lead to the accumulation of RNA fragments, suggesting that the loops impair mRNA decay. This result was questioned, because fragment occurrence was mainly assessed using ensemble methods, and their cellular localization and its implications had not been addressed on a single transcript level. Here, we demonstrate that the introduction of either MS2 stem-loops (MS2SL) or PP7 stem-loops (PP7SL) can affect the processing and subcellular localization of mRNA. We use single-molecule fluorescence in situ hybridization (smFISH) to determine the localization of three independent mRNAs tagged with the stem-loop labeling systems in glucose-rich and glucose starvation conditions. Transcripts containing MS2SL or PP7SL display aberrant localization in both the nucleus and the cytoplasm. These defects are most prominent in glucose starvation conditions, with nuclear mRNA processing being altered and stem-loop fragments abnormally enriching in processing bodies (PBs). The mislocalization of SL-containing RNAs is independent of the presence of the MS2 or PP7 coat protein (MCP or PCP).


Subject(s)
Cell Nucleus/metabolism , Cytoplasm/metabolism , Inverted Repeat Sequences , RNA Processing, Post-Transcriptional , RNA, Fungal/metabolism , RNA, Messenger/metabolism , Capsid Proteins/chemistry , Capsid Proteins/genetics , Capsid Proteins/metabolism , Cell Nucleus/drug effects , Cell Nucleus/ultrastructure , Cytoplasm/drug effects , Cytoplasm/ultrastructure , Glucose/metabolism , Glucose/pharmacology , In Situ Hybridization, Fluorescence , Levivirus/chemistry , Plasmids/chemistry , Plasmids/metabolism , RNA Stability , RNA, Fungal/chemistry , RNA, Fungal/genetics , RNA, Messenger/chemistry , RNA, Messenger/genetics , Saccharomyces cerevisiae/drug effects , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Single Molecule Imaging , Staining and Labeling/methods , Transfection
9.
EMBO Rep ; 17(7): 999-1012, 2016 07.
Article in English | MEDLINE | ID: mdl-27154402

ABSTRACT

Telomerase-mediated telomere elongation provides cell populations with the ability to proliferate indefinitely. Telomerase is capable of recognizing and extending the shortest telomeres in cells; nevertheless, how this mechanism is executed remains unclear. Here, we show that, in the fission yeast Schizosaccharomyces pombe, shortened telomeres are highly transcribed into the evolutionarily conserved long noncoding RNA TERRA A fraction of TERRA produced upon telomere shortening is polyadenylated and largely devoid of telomeric repeats, and furthermore, telomerase physically interacts with this polyadenylated TERRA in vivo We also show that experimentally enhanced transcription of a manipulated telomere promotes its association with telomerase and concomitant elongation. Our data represent the first direct evidence that TERRA stimulates telomerase recruitment and activity at chromosome ends in an organism with human-like telomeres.


Subject(s)
DNA-Binding Proteins/metabolism , Schizosaccharomyces/genetics , Schizosaccharomyces/metabolism , Telomerase/metabolism , Telomere Homeostasis , Telomere/genetics , Telomere/metabolism , DNA-Binding Proteins/genetics , Gene Expression Regulation, Fungal , Genome, Fungal , Poly A , Protein Binding , Telomere Shortening , Transcription, Genetic
10.
EMBO J ; 30(19): 4047-58, 2011 Aug 09.
Article in English | MEDLINE | ID: mdl-21829167

ABSTRACT

Eukaryotic up-frameshift 1 (UPF1) is a nucleic acid-dependent ATPase and 5'-to-3' helicase, best characterized for its roles in cytoplasmic RNA quality control. We previously demonstrated that human UPF1 binds to telomeres in vivo and its depletion leads to telomere instability. Here, we show that UPF1 is present at telomeres at least during S and G2/M phases and that UPF1 association with telomeres is stimulated by the phosphoinositide 3-kinase (PI3K)-related protein kinase ataxia telangiectasia mutated and Rad3-related (ATR) and by telomere elongation. UPF1 physically interacts with the telomeric factor TPP1 and with telomerase. Akin to UPF1 binding to telomeres, this latter interaction is mediated by ATR. Moreover, the ATPase activity of UPF1 is required to prevent the telomeric defects observed upon UPF1 depletion, and these defects stem predominantly from inefficient telomere leading-strand replication. Our results portray a scenario where UPF1 orchestrates crucial aspects of telomere biology, including telomere replication and telomere length homeostasis.


Subject(s)
Telomerase/metabolism , Telomere/genetics , Trans-Activators/genetics , Cell Cycle , Cell Line , Cell Nucleus/metabolism , HeLa Cells , Humans , Models, Biological , Proteome , Proteomics , RNA Helicases , Shelterin Complex , Telomerase/genetics , Telomere/ultrastructure , Telomere-Binding Proteins
11.
RNA Biol ; 12(9): 938-41, 2015.
Article in English | MEDLINE | ID: mdl-26158306

ABSTRACT

Alternative Lengthening of Telomeres (ALT) mechanisms allow telomerase-negative immortal cells to buffer replicative telomere shortening. ALT is naturally active in a number of human cancers and might be selected upon telomerase inactivation. ALT is thought to operate through homologous recombination (HR) occurring between telomeric repeats from independent chromosome ends. Indeed, suppression of a number of HR factors impairs ALT cell proliferation. Yet, how HR is initiated at ALT telomeres remains elusive. Mounting evidence suggests that the long noncoding telomeric RNA TERRA renders ALT telomeres recombinogenic by forming RNA:DNA hybrids with the telomeric C-rich strand. TERRA and telomeric hybrids act in concert with a number of other factors, including the RNA endoribonuclease RNaseH1 and the single stranded DNA binding protein RPA. The functional interaction network built upon these different players seems indispensable for ALT telomere maintenance, and digging into the molecular details of this previously unappreciated network might open the way to novel avenues for cancer treatments.


Subject(s)
RNA, Long Noncoding/genetics , Telomere Homeostasis , Homologous Recombination , Humans , Models, Biological , Protein Binding , RNA, Long Noncoding/metabolism , Telomere-Binding Proteins/metabolism , Transcription, Genetic
12.
RNA ; 18(4): 684-93, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22357912

ABSTRACT

Telomeres are nucleoprotein structures capping the physical ends of linear eukaryotic chromosomes. Although largely heterochromatic, telomeres are transcribed into telomeric repeat-containing RNA (TERRA) molecules by RNA polymerase II. The functions associated with telomere transcription and TERRA remain ill defined. Here we show that the transcriptional activity of human telomeres directly regulates their movement during interphase. We find that chemical inhibition of global transcription dampens telomere motion, while global stimulation promotes it. Likewise, when DNA methyltransferase enzymes are deleted to augment telomere transcription, we observe increased telomere movement. Finally, using a cell line engineered with a unique transcriptionally inducible telomere, we show that transcription of one specific telomere stimulates only its own dynamics without overtly affecting its stability or its length. We reveal a new and unforeseen function for telomere transcription as a regulator of telomere motion, and speculate on the intriguing possibility that transcription-dependent telomere motion sustains the maintenance of functional and dysfunctional telomeres.


Subject(s)
Gene Expression Regulation, Neoplastic , Neoplasms/genetics , Telomere , Transcription, Genetic , Base Sequence , Centromere , DNA Modification Methylases/metabolism , DNA Primers , HeLa Cells , Humans , Reverse Transcriptase Polymerase Chain Reaction
13.
Nucleic Acids Res ; 40(7): 2995-3005, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22139915

ABSTRACT

Eukaryotic telomeres are transcribed into telomeric repeat-containing RNA (TERRA). Telomeric transcription has been documented in mammals, birds, zebra fish, plants and budding yeast. Here we show that the chromosome ends of Schizosaccharomyces pombe produce distinct RNA species. As with budding yeast and mammals, S. pombe contains G-rich TERRA molecules and subtelomeric RNA species transcribed in the opposite direction of TERRA (ARRET). Moreover, fission yeast chromosome ends produce two novel RNA species: C-rich telomeric repeat-containing transcripts (ARIA) and subtelomeric transcripts complementary to ARRET (αARRET). RNA polymerase II (RNAPII) associates with pombe chromosome ends in vivo and the telomeric factor Rap1 negatively regulates this association, as well as the cellular accumulation of RNA emanating from chromosome ends. We also show that the RNAPII subunit Rpb7 and the non-canonical poly(A) polymerases Cid12 and Cid14 are involved in the regulation of TERRA, ARIA, ARRET and αARRET transcripts. We confirm the evolutionary conservation of telomere transcription, and reveal intriguing similarities and differences in the composition and regulation of telomeric transcripts among model organisms.


Subject(s)
RNA, Fungal/biosynthesis , Schizosaccharomyces/genetics , Telomere/genetics , Transcriptome , Cell Nucleus/genetics , Chromosomes, Fungal/genetics , Gene Expression Regulation, Fungal , Polyadenylation , Polynucleotide Adenylyltransferase/metabolism , RNA Polymerase II/physiology , RNA, Fungal/analysis , RNA, Fungal/metabolism , Schizosaccharomyces/metabolism , Schizosaccharomyces pombe Proteins/metabolism
14.
FEBS Lett ; 2024 Mar 06.
Article in English | MEDLINE | ID: mdl-38445359

ABSTRACT

Eukaryotic telomeres are transcribed into the long noncoding RNA TERRA. A fraction of TERRA remains associated with telomeres by forming RNA:DNA hybrids dubbed telR-loops. TERRA and telR-loops are essential to promote telomere elongation in human cancer cells that maintain telomeres through a homology-directed repair pathway known as alternative lengthening of telomeres or ALT. However, TERRA and telR-loops compromise telomere integrity and cell viability if their levels are not finely tuned. The study of telomere transcription in ALT cells will enormously expand our understanding of the ALT mechanism and of how genome integrity is maintained. Moreover, telomere transcription, TERRA and telR-loops are likely to become exceptionally suited targets for the development of novel anti-cancer therapies.

15.
Elife ; 112022 02 22.
Article in English | MEDLINE | ID: mdl-35191837

ABSTRACT

DNA oxidation by ten-eleven translocation (TET) family enzymes is essential for epigenetic reprogramming. The conversion of 5-methylcytosine (5mC) into 5-hydroxymethylcytosine (5hmC) initiates developmental and cell-type-specific transcriptional programs through mechanisms that include changes in the chromatin structure. Here, we show that the presence of 5hmC in the transcribed gene promotes the annealing of the nascent RNA to the template DNA strand, leading to the formation of an R-loop. Depletion of TET enzymes reduced global R-loops in the absence of gene expression changes, whereas CRISPR-mediated tethering of TET to an active gene promoted the formation of R-loops. The genome-wide distribution of 5hmC and R-loops shows a positive correlation in mouse and human stem cells and overlap in half of all active genes. Moreover, R-loop resolution leads to differential expression of a subset of genes that are involved in crucial events during stem cell proliferation. Altogether, our data reveal that epigenetic reprogramming via TET activity promotes co-transcriptional R-loop formation, disclosing new mechanisms of gene expression regulation.


Subject(s)
Dioxygenases , R-Loop Structures , 5-Methylcytosine/metabolism , Animals , Cytosine , DNA/metabolism , DNA Methylation , Dioxygenases/genetics , Epigenesis, Genetic , Epigenomics , Humans , Mice
16.
Prog Mol Subcell Biol ; 51: 65-94, 2011.
Article in English | MEDLINE | ID: mdl-21287134

ABSTRACT

Telomeres protect the ends of linear eukaryotic chromosomes from being recognized as DNA double-stranded breaks, thereby maintaining the stability of our genome. The highly heterochromatic nature of telomeres had, for a long time, reinforced the idea that telomeres were transcriptionally silent. Since a few years, however, we know that DNA-dependent RNA polymerase II transcribes telomeric DNA into TElomeric Repeat-containing RNA (TERRA) molecules in a large variety of eukaryotes. In this chapter, we summarize the current knowledge of telomere structure and function and extensively review data accumulated on TERRA biogenesis and regulation. We also discuss putative functions of TERRA in preserving telomere stability and propose future directions for research encompassing this novel and exciting aspect of telomere biology.


Subject(s)
RNA, Long Noncoding , Telomere , Eukaryota/genetics , Humans , RNA Polymerase II/genetics
17.
RNA ; 15(12): 2186-94, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19850908

ABSTRACT

The longstanding dogma that telomeres, the heterochromatic extremities of linear eukaryotic chromosomes, are transcriptionally silent was overturned by the discovery that DNA-dependent RNA polymerase II (RNAPII) transcribes telomeric DNA into telomeric repeat-containing RNA (TERRA). Here, we show that CpG dinucleotide-rich DNA islands, shared among multiple human chromosome ends, promote transcription of TERRA molecules. TERRA promoters sustain cellular expression of reporter genes, are located immediately upstream of TERRA transcription start sites, and are bound by active RNAPII in vivo. Finally, the identified promoter CpG dinucleotides are methylated in vivo, and cytosine methylation negatively regulates TERRA abundance. The existence of subtelomeric promoters, driving TERRA transcription from independent chromosome ends, supports the idea that TERRA exerts fundamental functions in the context of telomere biology.


Subject(s)
CpG Islands , Telomere/genetics , Transcription, Genetic , Cell Line , DNA Methylation , Humans , Promoter Regions, Genetic
18.
Nat Commun ; 12(1): 3760, 2021 06 18.
Article in English | MEDLINE | ID: mdl-34145295

ABSTRACT

Alternative Lengthening of Telomeres (ALT) is a Break-Induced Replication (BIR)-based mechanism elongating telomeres in a subset of human cancer cells. While the notion that spontaneous DNA damage at telomeres is required to initiate ALT, the molecular triggers of this physiological telomere instability are largely unknown. We previously proposed that the telomeric long noncoding RNA TERRA may represent one such trigger; however, given the lack of tools to suppress TERRA transcription in cells, our hypothesis remained speculative. We have developed Transcription Activator-Like Effectors able to rapidly inhibit TERRA transcription from multiple chromosome ends in an ALT cell line. TERRA transcription inhibition decreases marks of DNA replication stress and DNA damage at telomeres and impairs ALT activity and telomere length maintenance. We conclude that TERRA transcription actively destabilizes telomere integrity in ALT cells, thereby triggering BIR and promoting telomere elongation. Our data point to TERRA transcription manipulation as a potentially useful target for therapy.


Subject(s)
RNA, Long Noncoding/genetics , Telomere Homeostasis/genetics , Telomere/genetics , Transcription, Genetic/genetics , Cell Line, Tumor , Chromosome Breakage , DNA Damage/genetics , DNA Replication/genetics , High-Throughput Nucleotide Sequencing , Humans , Neoplasms/genetics
19.
Curr Biol ; 16(4): 433-9, 2006 Feb 21.
Article in English | MEDLINE | ID: mdl-16488880

ABSTRACT

The eukaryotic nonsense-mediated mRNA decay (NMD) pathway degrades mRNAs carrying premature stop codons (PTC). In humans, NMD depends on the RNA- and DNA-dependent 5'-3' helicase UPF1 and six other gene products referred to as SMG1, UPF2, UPF3, EST1A/SMG6, EST1B/SMG5, and EST1C/SMG7. The NMD machinery is also thought to coordinate mRNA nuclear export and translation and to regulate the levels of several physiologic transcripts. Furthermore, in a process named SMD, UPF1 promotes degradation of mRNAs that are bound by Staufen 1. Intriguingly, SMG1 and EST1A/SMG6 function also in DNA repair and telomere maintenance, respectively. Here, we show that UPF1 is also required for genome stability. shRNA-mediated depletion of UPF1 causes human cells to arrest early in S phase, inducing an ATR-dependent DNA-damage response. A fraction of hyperphosphorylated UPF1 associates with chromatin of unperturbed cells, and chromatin association increases in S phase and upon gamma irradiation. ATR phosphorylates UPF1 both in vitro and in vivo, and shRNA-mediated downregulation of ATR diminished the association of UPF1 with chromatin, although it did not affect NMD. Physical interaction of UPF1 with DNA polymerase delta suggests a role for human UPF1 in DNA synthesis during replication or repair.


Subject(s)
Genomic Instability/physiology , RNA Helicases/physiology , S Phase/physiology , Ataxia Telangiectasia Mutated Proteins , Cell Cycle Proteins , Chromatin , DNA Damage , DNA Polymerase III , HeLa Cells , Humans , Phosphorylation , Protein Serine-Threonine Kinases , RNA Stability/physiology
20.
Nucleic Acids Res ; 35(22): 7688-97, 2007.
Article in English | MEDLINE | ID: mdl-17984081

ABSTRACT

The Smg proteins Smg5, Smg6 and Smg7 are involved in nonsense-mediated RNA decay (NMD) in metazoans, but no orthologs have been found in the budding yeast Saccharomyces cerevisiae. Sequence alignments reveal that yeast Ebs1p is similar in structure to the human Smg5-7, with highest homology to Smg7. We demonstrate here that Ebs1p is involved in NMD and behaves similarly to human Smg proteins. Indeed, both loss and overexpression of Ebs1p results in stabilization of NMD targets. However, Ebs1-loss in yeast or Smg7-depletion in human cells only partially disrupts NMD and in the latter, Smg7-depletion is partially compensated for by Smg6. Ebs1p physically interacts with the NMD helicase Upf1p and overexpressed Ebs1p leads to recruitment of Upf1p into cytoplasmic P-bodies. Furthermore, Ebs1p localizes to P-bodies upon glucose starvation along with Upf1p. Overall our findings suggest that NMD is more conserved in evolution than previously thought, and that at least one of the Smg5-7 proteins is conserved in budding yeast.


Subject(s)
Codon, Nonsense , RNA, Messenger/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/genetics , Amino Acid Sequence , Carrier Proteins/chemistry , Gene Deletion , Gene Expression Regulation, Fungal , HeLa Cells , Humans , Molecular Sequence Data , Mutation , Protein Biosynthesis , RNA Helicases/metabolism , Saccharomyces cerevisiae/drug effects , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/genetics , Sequence Alignment , Sirolimus/pharmacology , Telomerase/genetics
SELECTION OF CITATIONS
SEARCH DETAIL