Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.762
Filter
Add more filters

Publication year range
1.
Annu Rev Biochem ; 89: 159-187, 2020 06 20.
Article in English | MEDLINE | ID: mdl-32176523

ABSTRACT

This review focuses on imaging DNA and single RNA molecules in living cells to define eukaryotic functional organization and dynamic processes. The latest advances in technologies to visualize individual DNA loci and RNAs in real time are discussed. Single-molecule fluorescence microscopy provides the spatial and temporal resolution to reveal mechanisms regulating fundamental cell functions. Novel insights into the regulation of nuclear architecture, transcription, posttranscriptional RNA processing, and RNA localization provided by multicolor fluorescence microscopy are reviewed. A perspective on the future use of live imaging technologies and overcoming their current limitations is provided.


Subject(s)
Cell Nucleus/ultrastructure , Chromatin/ultrastructure , DNA/ultrastructure , Gene Expression Regulation , RNA, Messenger/ultrastructure , RNA, Small Untranslated/ultrastructure , Animals , Cell Nucleus/genetics , Cell Nucleus/metabolism , Chromatin/metabolism , DNA/genetics , DNA/metabolism , DNA Replication , Eukaryotic Cells/metabolism , Eukaryotic Cells/ultrastructure , Humans , Microscopy, Fluorescence , Protein Biosynthesis , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Untranslated/genetics , RNA, Small Untranslated/metabolism , Single Molecule Imaging/instrumentation , Single Molecule Imaging/methods , Staining and Labeling/methods , Telomere/metabolism , Telomere/ultrastructure , Transcription, Genetic
2.
Nature ; 609(7929): 1048-1055, 2022 09.
Article in English | MEDLINE | ID: mdl-36104563

ABSTRACT

Telomeres, the ends of eukaryotic chromosomes, play pivotal parts in ageing and cancer and are targets of DNA damage and the DNA damage response1-5. Little is known about the structure of telomeric chromatin at the molecular level. Here we used negative stain electron microscopy and single-molecule magnetic tweezers to characterize 3-kbp-long telomeric chromatin fibres. We also obtained the cryogenic electron microscopy structure of the condensed telomeric tetranucleosome and its dinucleosome unit. The structure displayed close stacking of nucleosomes with a columnar arrangement, and an unusually short nucleosome repeat  length that comprised about 132 bp DNA wound in a continuous superhelix around histone octamers. This columnar structure is primarily stabilized by the H2A carboxy-terminal and histone amino-terminal tails in a synergistic manner. The columnar conformation results in exposure of the DNA helix, which may make it susceptible to both DNA damage and the DNA damage response. The conformation also exists in an alternative open state, in which one nucleosome is unstacked and flipped out, which exposes the acidic patch of the histone surface. The structural features revealed in this work suggest mechanisms by which protein factors involved in telomere maintenance can access telomeric chromatin in its compact form.


Subject(s)
Chromatin , DNA , Histones , Molecular Conformation , Telomere , Chromatin/chemistry , Chromatin/genetics , Chromatin/ultrastructure , DNA/chemistry , DNA/metabolism , DNA/ultrastructure , DNA Damage , Histones/chemistry , Histones/metabolism , Histones/ultrastructure , Humans , Microscopy, Electron , Nucleosomes/chemistry , Nucleosomes/genetics , Nucleosomes/ultrastructure , Single Molecule Imaging , Telomere/chemistry , Telomere/genetics , Telomere/ultrastructure
3.
Nature ; 593(7859): 454-459, 2021 05.
Article in English | MEDLINE | ID: mdl-33981033

ABSTRACT

Telomerase is unique among the reverse transcriptases in containing a noncoding RNA (known as telomerase RNA (TER)) that includes a short template that is used for the processive synthesis of G-rich telomeric DNA repeats at the 3' ends of most eukaryotic chromosomes1. Telomerase maintains genomic integrity, and its activity or dysregulation are critical determinants of human longevity, stem cell renewal and cancer progression2,3. Previous cryo-electron microscopy structures have established the general architecture, protein components and stoichiometries of Tetrahymena and human telomerase, but our understandings of the details of DNA-protein and RNA-protein interactions and of the mechanisms and recruitment involved remain limited4-6. Here we report cryo-electron microscopy structures of active Tetrahymena telomerase with telomeric DNA at different steps of nucleotide addition. Interactions between telomerase reverse transcriptase (TERT), TER and DNA reveal the structural basis of the determination of the 5' and 3' template boundaries, handling of the template-DNA duplex and separation of the product strand during nucleotide addition. The structure and binding interface between TERT and telomerase protein p50 (a homologue of human TPP17,8) define conserved interactions that are required for telomerase activation and recruitment to telomeres. Telomerase La-related protein p65 remodels several regions of TER, bridging the 5' and 3' ends and the conserved pseudoknot to facilitate assembly of the TERT-TER catalytic core.


Subject(s)
Cryoelectron Microscopy , Telomerase/chemistry , Telomerase/metabolism , Telomere/metabolism , Tetrahymena thermophila/enzymology , Amino Acid Motifs , Binding Sites , DNA/chemistry , DNA/metabolism , DNA/ultrastructure , Humans , Models, Molecular , Nucleotides , Protein Binding , RNA/chemistry , RNA/metabolism , RNA/ultrastructure , Ribonucleoproteins/chemistry , Ribonucleoproteins/metabolism , Ribonucleoproteins/ultrastructure , Shelterin Complex/chemistry , Shelterin Complex/metabolism , Telomerase/ultrastructure , Telomere/genetics , Telomere/ultrastructure , Telomere-Binding Proteins/chemistry , Telomere-Binding Proteins/metabolism , Templates, Genetic , Tetrahymena thermophila/ultrastructure
4.
Nature ; 593(7859): 449-453, 2021 05.
Article in English | MEDLINE | ID: mdl-33883742

ABSTRACT

Telomerase adds telomeric repeats at chromosome ends to compensate for the telomere loss that is caused by incomplete genome end replication1. In humans, telomerase is upregulated during embryogenesis and in cancers, and mutations that compromise the function of telomerase result in disease2. A previous structure of human telomerase at a resolution of 8 Å revealed a vertebrate-specific composition and architecture3, comprising a catalytic core that is flexibly tethered to an H and ACA (hereafter, H/ACA) box ribonucleoprotein (RNP) lobe by telomerase RNA. High-resolution structural information is necessary to develop treatments that can effectively modulate telomerase activity as a therapeutic approach against cancers and disease. Here we used cryo-electron microscopy to determine the structure of human telomerase holoenzyme bound to telomeric DNA at sub-4 Å resolution, which reveals crucial DNA- and RNA-binding interfaces in the active site of telomerase as well as the locations of mutations that alter telomerase activity. We identified a histone H2A-H2B dimer within the holoenzyme that was bound to an essential telomerase RNA motif, which suggests a role for histones in the folding and function of telomerase RNA. Furthermore, this structure of a eukaryotic H/ACA RNP reveals the molecular recognition of conserved RNA and protein motifs, as well as interactions that are crucial for understanding the molecular pathology of many mutations that cause disease. Our findings provide the structural details of the assembly and active site of human telomerase, which paves the way for the development of therapeutic agents that target this enzyme.


Subject(s)
Cryoelectron Microscopy , DNA/chemistry , DNA/ultrastructure , Telomerase/chemistry , Telomerase/ultrastructure , Telomere , Binding Sites , Catalytic Domain , DNA/genetics , DNA/metabolism , Histones/chemistry , Histones/metabolism , Holoenzymes/chemistry , Holoenzymes/metabolism , Holoenzymes/ultrastructure , Humans , Models, Molecular , Mutation , Nucleic Acid Conformation , Nucleotide Motifs , Protein Subunits/chemistry , Protein Subunits/metabolism , RNA/chemistry , RNA/metabolism , RNA/ultrastructure , Ribonucleoproteins/chemistry , Ribonucleoproteins/genetics , Ribonucleoproteins/metabolism , Ribonucleoproteins/ultrastructure , Telomerase/metabolism , Telomere/genetics , Telomere/metabolism , Telomere/ultrastructure
5.
Mol Cell ; 75(1): 131-144.e3, 2019 07 11.
Article in English | MEDLINE | ID: mdl-31204167

ABSTRACT

In Saccharomyces cerevisiae, dicentric chromosomes stemming from telomere fusions preferentially break at the fusion. This process restores a normal karyotype and protects chromosomes from the detrimental consequences of accidental fusions. Here, we address the molecular basis of this rescue pathway. We observe that tandem arrays tightly bound by the telomere factor Rap1 or a heterologous high-affinity DNA binding factor are sufficient to establish breakage hotspots, mimicking telomere fusions within dicentrics. We also show that condensins generate forces sufficient to rapidly refold dicentrics prior to breakage by cytokinesis and are essential to the preferential breakage at telomere fusions. Thus, the rescue of fused telomeres results from a condensin- and Rap1-driven chromosome folding that favors fusion entrapment where abscission takes place. Because a close spacing between the DNA-bound Rap1 molecules is essential to this process, Rap1 may act by stalling condensins.


Subject(s)
Adenosine Triphosphatases/genetics , Chromosomes, Fungal/metabolism , DNA, Fungal/genetics , DNA-Binding Proteins/genetics , Multiprotein Complexes/genetics , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae/genetics , Telomere-Binding Proteins/genetics , Telomere/metabolism , Transcription Factors/genetics , Adenosine Triphosphatases/metabolism , Chromosome Breakpoints , Chromosomes, Fungal/ultrastructure , Cytokinesis/genetics , DNA, Fungal/metabolism , DNA-Binding Proteins/metabolism , Gene Expression , Karyotype , Models, Genetic , Multiprotein Complexes/metabolism , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae/ultrastructure , Saccharomyces cerevisiae Proteins/metabolism , Shelterin Complex , Telomere/ultrastructure , Telomere-Binding Proteins/metabolism , Transcription Factors/metabolism
6.
Mol Cell ; 71(4): 510-525.e6, 2018 08 16.
Article in English | MEDLINE | ID: mdl-30033372

ABSTRACT

Telomeres regulate DNA damage response (DDR) and DNA repair activity at chromosome ends. How telomere macromolecular structure contributes to ATM regulation and its potential dissociation from control over non-homologous end joining (NHEJ)-dependent telomere fusion is of central importance to telomere-dependent cell aging and tumor suppression. Using super-resolution microscopy, we identify that ATM activation at mammalian telomeres with reduced TRF2 or at human telomeres during mitotic arrest occurs specifically with a structural change from telomere loops (t-loops) to linearized telomeres. Additionally, we find the TRFH domain of TRF2 regulates t-loop formation while suppressing ATM activity. Notably, we demonstrate that ATM activation and telomere linearity occur separately from telomere fusion via NHEJ and that linear DDR-positive telomeres can remain resistant to fusion, even during an extended G1 arrest, when NHEJ is most active. Collectively, these results suggest t-loops act as conformational switches that specifically regulate ATM activation independent of telomere mechanisms to inhibit NHEJ.


Subject(s)
Ataxia Telangiectasia Mutated Proteins/genetics , DNA End-Joining Repair , Telomere/metabolism , Telomeric Repeat Binding Protein 2/genetics , Animals , Ataxia Telangiectasia Mutated Proteins/metabolism , Cell Line , Cell Line, Tumor , DNA Damage , Fibroblasts/cytology , Fibroblasts/metabolism , G1 Phase Cell Cycle Checkpoints/genetics , HEK293 Cells , HeLa Cells , Humans , Mice , Mitosis , Protein Domains , Telomere/ultrastructure , Telomeric Repeat Binding Protein 2/chemistry , Telomeric Repeat Binding Protein 2/metabolism
7.
Genes Dev ; 31(6): 578-589, 2017 03 15.
Article in English | MEDLINE | ID: mdl-28381412

ABSTRACT

Telomeres are protected by shelterin, a six-subunit protein complex that represses the DNA damage response (DDR) at chromosome ends. Extensive data suggest that TRF2 in shelterin remodels telomeres into the t-loop structure, thereby hiding telomere ends from double-stranded break repair and ATM signaling, whereas POT1 represses ATR signaling by excluding RPA. An alternative protection mechanism was suggested recently by which shelterin subunits TRF1, TRF2, and TIN2 mediate telomeric chromatin compaction, which was proposed to minimize access of DDR factors. We performed superresolution imaging of telomeres in mouse cells after conditional deletion of TRF1, TRF2, or both, the latter of which results in the complete loss of shelterin. Upon removal of TRF1 or TRF2, we observed only minor changes in the telomere volume in most of our experiments. Upon codeletion of TRF1 and TRF2, the telomere volume increased by varying amounts, but even those samples exhibiting small changes in telomere volume showed DDR at nearly all telomeres. Upon shelterin removal, telomeres underwent 53BP1-dependent clustering, potentially explaining at least in part the apparent increase in telomere volume. Furthermore, chromatin accessibility, as determined by ATAC-seq (assay for transposase-accessible chromatin [ATAC] with high-throughput sequencing), was not substantially altered by shelterin removal. These results suggest that the DDR induced by shelterin removal does not require substantial telomere decompaction.


Subject(s)
DNA Damage , Telomere/ultrastructure , Telomeric Repeat Binding Protein 1/physiology , Telomeric Repeat Binding Protein 2/physiology , Animals , Cells, Cultured , Chromatin/physiology , Mice , Microscopy, Fluorescence , Tumor Suppressor p53-Binding Protein 1/physiology
8.
Genes Dev ; 31(6): 567-577, 2017 Mar 15.
Article in English | MEDLINE | ID: mdl-28381410

ABSTRACT

Telomeres are specialized nucleoprotein structures that protect chromosome ends from DNA damage response (DDR) and DNA rearrangements. The telomeric shelterin protein TRF2 suppresses the DDR, and this function has been attributed to its abilities to trigger t-loop formation or prevent massive decompaction and loss of density of telomeric chromatin. Here, we applied stochastic optical reconstruction microscopy (STORM) to measure the sizes and shapes of functional human telomeres of different lengths and dysfunctional telomeres that elicit a DDR. Telomeres have an ovoid appearance with considerable plasticity in shape. Examination of many telomeres demonstrated that depletion of TRF2, TRF1, or both affected the sizes of only a small subset of telomeres. Costaining of telomeres with DDR markers further revealed that the majority of DDR signaling telomeres retained a normal size. Thus, DDR signaling at telomeres does not require decompaction. We propose that telomeres are monitored by the DDR machinery in the absence of telomere expansion and that the DDR is triggered by changes at the molecular level in structure and protein composition.


Subject(s)
DNA Damage , Telomere/ultrastructure , Chromatin/physiology , Fluorescent Antibody Technique , HeLa Cells , Humans , In Situ Hybridization, Fluorescence , Microscopy, Fluorescence , Telomeric Repeat Binding Protein 1/analysis , Telomeric Repeat Binding Protein 1/immunology , Telomeric Repeat Binding Protein 1/physiology , Telomeric Repeat Binding Protein 2/physiology
9.
Nucleic Acids Res ; 50(9): 5047-5063, 2022 05 20.
Article in English | MEDLINE | ID: mdl-35489064

ABSTRACT

Telomeres, the ends of linear chromosomes, are composed of repetitive DNA sequences, histones and a protein complex called shelterin. How DNA is packaged at telomeres is an outstanding question in the field with significant implications for human health and disease. Here, we studied the architecture of telomeres and their spatial association with other chromatin domains in different cell types using correlative light and electron microscopy. To this end, the shelterin protein TRF1 or TRF2 was fused in tandem to eGFP and the peroxidase APEX2, which provided a selective and electron-dense label to interrogate telomere organization by transmission electron microscopy, electron tomography and scanning electron microscopy. Together, our work reveals, for the first time, ultrastructural insight into telomere architecture. We show that telomeres are composed of a dense and highly compacted mesh of chromatin fibres. In addition, we identify marked differences in telomere size, shape and chromatin compaction between cancer and non-cancer cells and show that telomeres are in direct contact with other heterochromatin regions. Our work resolves the internal architecture of telomeres with unprecedented resolution and advances our understanding of how telomeres are organized in situ.


Subject(s)
Telomere/ultrastructure , Humans , Microscopy, Electron , Shelterin Complex , Telomere/genetics , Telomere/metabolism , Telomeric Repeat Binding Protein 1/metabolism , Telomeric Repeat Binding Protein 2/metabolism
10.
Nat Methods ; 17(7): 734-740, 2020 07.
Article in English | MEDLINE | ID: mdl-32541853

ABSTRACT

An outstanding challenge in single-molecule localization microscopy is the accurate and precise localization of individual point emitters in three dimensions in densely labeled samples. One established approach for three-dimensional single-molecule localization is point-spread-function (PSF) engineering, in which the PSF is engineered to vary distinctively with emitter depth using additional optical elements. However, images of dense emitters, which are desirable for improving temporal resolution, pose a challenge for algorithmic localization of engineered PSFs, due to lateral overlap of the emitter PSFs. Here we train a neural network to localize multiple emitters with densely overlapping Tetrapod PSFs over a large axial range. We then use the network to design the optimal PSF for the multi-emitter case. We demonstrate our approach experimentally with super-resolution reconstructions of mitochondria and volumetric imaging of fluorescently labeled telomeres in cells. Our approach, DeepSTORM3D, enables the study of biological processes in whole cells at timescales that are rarely explored in localization microscopy.


Subject(s)
Deep Learning , Imaging, Three-Dimensional/methods , Single Molecule Imaging/methods , Biological Phenomena , Neural Networks, Computer , Telomere/ultrastructure
11.
Cell ; 132(5): 745-57, 2008 Mar 07.
Article in English | MEDLINE | ID: mdl-18329362

ABSTRACT

Single-strand extensions of the G strand of telomeres are known to be critical for chromosome-end protection and length regulation. Here, we report that in C. elegans, chromosome termini possess 3' G-strand overhangs as well as 5' C-strand overhangs. C tails are as abundant as G tails and are generated by a well-regulated process. These two classes of overhangs are bound by two single-stranded DNA binding proteins, CeOB1 and CeOB2, which exhibit specificity for G-rich or C-rich telomeric DNA. Strains of worms deleted for CeOB1 have elongated telomeres as well as extended G tails, whereas CeOB2 deficiency leads to telomere-length heterogeneity. Both CeOB1 and CeOB2 contain OB (oligo-saccharide/oligo-nucleotide binding) folds, which exhibit structural similarity to the second and first OB folds of the mammalian telomere binding protein hPOT1, respectively. Our results suggest that C. elegans telomere homeostasis relies on a novel mechanism that involves 5' and 3' single-stranded termini.


Subject(s)
Caenorhabditis elegans/genetics , DNA-Binding Proteins/metabolism , Telomere/metabolism , Animals , Animals, Genetically Modified , Caenorhabditis elegans/metabolism , Cell Line , DNA, Helminth/metabolism , DNA, Single-Stranded/metabolism , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , Embryo, Nonmammalian/metabolism , Humans , Structural Homology, Protein , Telomere/chemistry , Telomere/ultrastructure
12.
Nucleic Acids Res ; 49(2): 760-775, 2021 01 25.
Article in English | MEDLINE | ID: mdl-33347580

ABSTRACT

Chromosome stability is primarily determined by telomere length. TRF1 is the core subunit of shelterin that plays a critical role in telomere organization and replication. However, the dynamics of TRF1 in scenarios of telomere-processing activities remain elusive. Using single-molecule magnetic tweezers, we here investigated the dynamics of TRF1 upon organizing a human telomere and the protein-DNA interactions at a moving telomeric fork. We first developed a method to obtain telomeres from human cells for directly measuring the telomere length by single-molecule force spectroscopy. Next, we examined the compaction and decompaction of a telomere by TRF1 dimers. TRF1 dissociates from a compacted telomere with heterogenous loops in ∼20 s. We also found a negative correlation between the number of telomeric loops and loop sizes. We further characterized the dynamics of TRF1 at a telomeric DNA fork. With binding energies of 11 kBT, TRF1 can modulate the forward and backward steps of DNA fork movements by 2-9 s at a critical force of F1/2, temporarily maintaining the telomeric fork open. Our results shed light on the mechanisms of how TRF1 organizes human telomeres and facilitates the efficient replication of telomeric DNA. Our work will help future research on the chemical biology of telomeres and shelterin-targeted drug discovery.


Subject(s)
Micromanipulation/methods , Telomere/ultrastructure , Telomeric Repeat Binding Protein 1/metabolism , Biotinylation , Digoxigenin , Humans , Inverted Repeat Sequences , K562 Cells , Magnets , Shelterin Complex , Single Molecule Imaging , Telomere/chemistry , Telomere-Binding Proteins/chemistry , Telomere-Binding Proteins/physiology
13.
BMC Cancer ; 22(1): 137, 2022 Feb 03.
Article in English | MEDLINE | ID: mdl-35114947

ABSTRACT

BACKGROUND: Telomeres are protective structures at chromosome ends which shorten gradually with increasing age. In chronic lymphocytic leukemia (CLL), short telomeres have been associated with unfavorable disease outcome, but the link between clonal evolution and telomere shortening remains unresolved. METHODS: We investigated relative telomere length (RTL) in a well-characterized cohort of 198 CLL patients by qPCR and focused in detail on a subgroup 26 patients who underwent clonal evolution of TP53 mutations (evolTP53). In the evolTP53 subgroup we explored factors influencing clonal evolution and corresponding changes in telomere length through measurements of telomerase expression, lymphocyte doubling time, and BCR signaling activity. RESULTS: At baseline, RTL of the evolTP53 patients was scattered across the entire RTL spectrum observed in our CLL cohort. RTL changed in the follow-up samples of 16/26 (62%) evolTP53 cases, inclining to reach intermediate RTL values, i.e., longer telomeres shortened compared to baseline while shorter ones prolonged. For the first time we show that TP53 clonal shifts are linked to RTL change, including unexpected RTL prolongation. We further investigated parameters associated with RTL changes. Unstable telomeres were significantly more frequent among younger patients (P = 0.032). Shorter telomeres were associated with decreased activity of the B-cell receptor signaling components p-ERK1/2, p-ZAP-70/SYK, and p-NFκB (P = 0.04, P = 0.01, and P = 0.02, respectively). CONCLUSIONS: Our study revealed that changes of telomere length reflect evolution in leukemic subclone proportion, and are associated with specific clinico-biological features of the explored cohort.


Subject(s)
Clonal Evolution/genetics , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Telomere/ultrastructure , Tumor Suppressor Protein p53/genetics , Female , Humans , Male , Middle Aged , Mutation , Proto-Oncogene Proteins c-bcr/metabolism , Signal Transduction , Telomerase/genetics
14.
Nat Chem Biol ; 16(7): 801-809, 2020 07.
Article in English | MEDLINE | ID: mdl-32066968

ABSTRACT

Telomere maintenance by telomerase is essential for continuous proliferation of human cells and is vital for the survival of stem cells and 90% of cancer cells. To compensate for telomeric DNA lost during DNA replication, telomerase processively adds GGTTAG repeats to chromosome ends by copying the template region within its RNA subunit. Between repeat additions, the RNA template must be recycled. How telomerase remains associated with substrate DNA during this critical translocation step remains unknown. Using a single-molecule telomerase activity assay utilizing high-resolution optical tweezers, we demonstrate that stable substrate DNA binding at an anchor site within telomerase facilitates the processive synthesis of telomeric repeats. The product DNA synthesized by telomerase can be recaptured by the anchor site or fold into G-quadruplex structures. Our results provide detailed mechanistic insights into telomerase catalysis, a process of critical importance in aging and cancer.


Subject(s)
DNA/metabolism , G-Quadruplexes , RNA/metabolism , Telomerase/metabolism , Telomere/enzymology , Biocatalysis , DNA/genetics , DNA Replication , Gene Expression , HEK293 Cells , Humans , Optical Tweezers , RNA/genetics , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Telomerase/genetics , Telomere/ultrastructure
15.
Nucleic Acids Res ; 48(16): 9195-9203, 2020 09 18.
Article in English | MEDLINE | ID: mdl-32810236

ABSTRACT

G-quadruplex (GQ) is formed at various regions of DNA, including telomeres of chromosomes and regulatory regions of oncogenes. Since GQ is important in both gene regulation and genome instability, the biological and medical implications of this abnormal DNA structure have been intensively studied. Its formation mechanisms, however, are not clearly understood yet. We report single-molecule fluorescence experiments to monitor the cotranscriptional GQ formation coupled with R-loop formation using T7 RNA polymerase. The GQ is formed very rarely per single-round transcription. R-loop formation precedes and facilitates GQ formation. Once formed, some GQs are extremely stable, resistant even to RNase H treatment, and accumulate in multiple-round transcription conditions. On the other hand, GQ existing in the non-template strand promotes the R-loop formation in the next rounds of transcription. Our study clearly shows the existence of a positive feedback mechanism of GQ and R-loop formations, which may possibly contribute to gene regulation and genome instability.


Subject(s)
DNA/ultrastructure , G-Quadruplexes , R-Loop Structures/genetics , Single Molecule Imaging/methods , DNA-Directed RNA Polymerases/ultrastructure , Fluorescence , Fluorescence Resonance Energy Transfer , Humans , Telomere/ultrastructure , Viral Proteins/ultrastructure
16.
Nucleic Acids Res ; 48(21): 11942-11957, 2020 12 02.
Article in English | MEDLINE | ID: mdl-33137181

ABSTRACT

Genomic DNA and cellular RNAs can form a variety of non-B secondary structures, including G-quadruplex (G4) and R-loops. G4s are constituted by stacked guanine tetrads held together by Hoogsteen hydrogen bonds and can form at key regulatory sites of eukaryote genomes and transcripts, including gene promoters, untranslated exon regions and telomeres. R-loops are 3-stranded structures wherein the two strands of a DNA duplex are melted and one of them is annealed to an RNA. Specific G4 binders are intensively investigated to discover new effective anticancer drugs based on a common rationale, i.e.: the selective inhibition of oncogene expression or specific impairment of telomere maintenance. However, despite the high number of known G4 binders, such a selective molecular activity has not been fully established and several published data point to a different mode of action. We will review published data that address the close structural interplay between G4s and R-loops in vitro and in vivo, and how these interactions can have functional consequences in relation to G4 binder activity. We propose that R-loops can play a previously-underestimated role in G4 binder action, in relation to DNA damage induction, telomere maintenance, genome and epigenome instability and alterations of gene expression programs.


Subject(s)
DNA/chemistry , G-Quadruplexes , Genome, Human , R-Loop Structures , RNA/chemistry , Aminoquinolines/chemistry , Aminoquinolines/pharmacology , Base Pairing , DNA/genetics , DNA/metabolism , G-Quadruplexes/drug effects , Genomic Instability , Guanine/chemistry , Guanine/metabolism , Humans , Hydrogen Bonding , Ligands , Models, Molecular , Picolinic Acids/chemistry , Picolinic Acids/pharmacology , Promoter Regions, Genetic , R-Loop Structures/drug effects , RNA/genetics , RNA/metabolism , Telomere/drug effects , Telomere/metabolism , Telomere/ultrastructure , Telomere Homeostasis
17.
Proc Natl Acad Sci U S A ; 116(17): 8350-8359, 2019 04 23.
Article in English | MEDLINE | ID: mdl-30944218

ABSTRACT

G-quadruplexes (GQs) can adopt diverse structures and are functionally implicated in transcription, replication, translation, and maintenance of telomere. Their conformational diversity under physiological levels of mechanical stress, however, is poorly understood. We used single-molecule fluorescence-force spectroscopy that combines fluorescence resonance energy transfer with optical tweezers to measure human telomeric sequences under tension. Abrupt GQ unfolding with K+ in solution occurred at as many as four discrete levels of force. Added to an ultrastable state and a gradually unfolding state, there were six mechanically distinct structures. Extreme mechanical diversity was also observed with Na+, although GQs were mechanically weaker. Our ability to detect small conformational changes at low forces enabled the determination of refolding forces of about 2 pN. Refolding was rapid and stochastically redistributed molecules to mechanically distinct states. A single guanine-to-thymine substitution mutant required much higher ion concentrations to display GQ-like unfolding and refolded via intermediates, contrary to the wild type. Contradicting an earlier proposal, truncation to three hexanucleotide repeats resulted in a single-stranded DNA-like mechanical behavior under all conditions, indicating that at least four repeats are required to form mechanically stable structures.


Subject(s)
DNA/ultrastructure , Fluorescence Resonance Energy Transfer/methods , G-Quadruplexes , Telomere/ultrastructure , DNA/chemistry , Guanine/chemistry , Humans , Optical Tweezers , Repetitive Sequences, Nucleic Acid , Telomere/chemistry , Thymine/chemistry
18.
Proc Natl Acad Sci U S A ; 116(30): 15122-15127, 2019 07 23.
Article in English | MEDLINE | ID: mdl-31285335

ABSTRACT

Telomere shortening to a critical length can trigger aging and shorter life spans in mice and humans by a mechanism that involves induction of a persistent DNA damage response at chromosome ends and loss of cellular viability. However, whether telomere length is a universal determinant of species longevity is not known. To determine whether telomere shortening can be a single parameter to predict species longevities, here we measured in parallel the telomere length of a wide variety of species (birds and mammals) with very different life spans and body sizes, including mouse (Mus musculus), goat (Capra hircus), Audouin's gull (Larus audouinii), reindeer (Rangifer tarandus), griffon vulture (Gyps fulvus), bottlenose dolphin (Tursiops truncatus), American flamingo (Phoenicopterus ruber), and Sumatran elephant (Elephas maximus sumatranus). We found that the telomere shortening rate, but not the initial telomere length alone, is a powerful predictor of species life span. These results support the notion that critical telomere shortening and the consequent onset of telomeric DNA damage and cellular senescence are a general determinant of species life span.


Subject(s)
Longevity/genetics , Telomere Shortening , Telomere/ultrastructure , Animals , Bottle-Nosed Dolphin/genetics , Cellular Senescence , Charadriiformes/genetics , Elephants/genetics , Falconiformes/genetics , Goats/genetics , Humans , Mice , Regression Analysis , Reindeer/genetics , Species Specificity
19.
Int J Sports Med ; 43(1): 29-33, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34256387

ABSTRACT

Lower SIRT1 and insulin resistance are associated with accelerated telomere shortening. This study investigated whether the lifestyle of master athletes can attenuate these age-related changes and thereby slow aging. We compared insulin, SIRT1, and telomere length in highly trained male master athletes (n=52; aged 49.9±7.2 yrs) and age-matched non-athletes (n=19; aged 47.3±8.9 yrs). This is a cross-sectional study, in which all data were collected in one visit. Overnight fasted SIRT1 and insulin levels in whole blood were assessed using commercial kits. Relative telomere length was determined in leukocytes through qPCR analyses. Master athletes had higher SIRT1, lower insulin, and longer telomere length than age-matched non-athletes (p<0.05 for all). Insulin was inversely associated with SIRT1 (r=-0.38; p=0.001). Telomere length correlated positively with SIRT1 (r=0.65; p=0.001), whereas telomere length and insulin were not correlated (r=0.03; p=0.87). In conclusion, master athletes have higher SIRT1, lower insulin, and longer telomeres than age-matched non-athletes. Furthermore, SIRT1 was negatively associated with insulin and positively associated with telomere length. These findings suggest that in this sample of middle-aged participants reduced insulin, increased SIRT1 activity, and attenuation of biological aging are connected.


Subject(s)
Athletes , Insulin/blood , Longevity , Sirtuin 1 , Telomere/ultrastructure , Adult , Aging , Cross-Sectional Studies , Humans , Leukocytes , Male , Middle Aged , Sirtuin 1/genetics
20.
Annu Rev Genet ; 47: 563-99, 2013.
Article in English | MEDLINE | ID: mdl-24050176

ABSTRACT

Meiotic recombination is essential for fertility in most sexually reproducing species. This process also creates new combinations of alleles and has important consequences for genome evolution. Meiotic recombination is initiated by the formation of DNA double-strand breaks (DSBs), which are repaired by homologous recombination. DSBs are catalyzed by the evolutionarily conserved SPO11 protein, assisted by several other factors. Some of them are absolutely required, whereas others are needed only for full levels of DSB formation and may participate in the regulation of DSB timing and frequency as well as the coordination between DSB formation and repair. The sites where DSBs occur are not randomly distributed in the genome, and remarkably distinct strategies have emerged to control their localization in different species. Here, I review the recent advances in the components required for DSB formation and localization in the various model organisms in which these studies have been performed.


Subject(s)
Eukaryota/genetics , Homologous Recombination , Meiosis/genetics , Animals , Centromere/genetics , Centromere/ultrastructure , Chromatin/genetics , Chromatin/ultrastructure , Conserved Sequence , Crossing Over, Genetic , DNA Breaks, Double-Stranded , DNA Repair , DNA Topoisomerases, Type II/genetics , DNA Topoisomerases, Type II/physiology , DNA-Binding Proteins/physiology , Endodeoxyribonucleases/physiology , Gene Duplication , Humans , Models, Genetic , Protein Isoforms/physiology , Protein Structure, Tertiary , Species Specificity , Tandem Repeat Sequences , Telomere/genetics , Telomere/ultrastructure
SELECTION OF CITATIONS
SEARCH DETAIL