Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 49
Filter
Add more filters










Publication year range
1.
Nat Struct Mol Biol ; 2024 Apr 17.
Article in English | MEDLINE | ID: mdl-38632359

ABSTRACT

Current models suggest that DNA double-strand breaks (DSBs) can move to the nuclear periphery for repair. It is unclear to what extent human DSBs display such repositioning. Here we show that the human nuclear envelope localizes to DSBs in a manner depending on DNA damage response (DDR) kinases and cytoplasmic microtubules acetylated by α-tubulin acetyltransferase-1 (ATAT1). These factors collaborate with the linker of nucleoskeleton and cytoskeleton complex (LINC), nuclear pore complex (NPC) protein NUP153, nuclear lamina and kinesins KIF5B and KIF13B to generate DSB-capturing nuclear envelope tubules (dsbNETs). dsbNETs are partly supported by nuclear actin filaments and the circadian factor PER1 and reversed by kinesin KIFC3. Although dsbNETs promote repair and survival, they are also co-opted during poly(ADP-ribose) polymerase (PARP) inhibition to restrain BRCA1-deficient breast cancer cells and are hyper-induced in cells expressing the aging-linked lamin A mutant progerin. In summary, our results advance understanding of nuclear structure-function relationships, uncover a nuclear-cytoplasmic DDR and identify dsbNETs as critical factors in genome organization and stability.

2.
Cell Rep ; 43(3): 113891, 2024 Mar 26.
Article in English | MEDLINE | ID: mdl-38427561

ABSTRACT

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) hinders host gene expression, curbing defenses and licensing viral protein synthesis and virulence. During SARS-CoV-2 infection, the virulence factor non-structural protein 1 (Nsp1) targets the mRNA entry channel of mature cytoplasmic ribosomes, limiting translation. We show that Nsp1 also restrains translation by targeting nucleolar ribosome biogenesis. SARS-CoV-2 infection disrupts 18S and 28S ribosomal RNA (rRNA) processing. Expression of Nsp1 recapitulates the processing defects. Nsp1 abrogates rRNA production without altering the expression of critical processing factors or nucleolar organization. Instead, Nsp1 localizes to the nucleolus, interacting with precursor-rRNA and hindering its maturation separately from the viral protein's role in restricting mature ribosomes. Thus, SARS-CoV-2 Nsp1 limits translation by targeting ribosome biogenesis and mature ribosomes. These findings revise our understanding of how SARS-CoV-2 Nsp1 controls human protein synthesis, suggesting that efforts to counter Nsp1's effect on translation should consider the protein's impact from ribosome manufacturing to mature ribosomes.


Subject(s)
COVID-19 , SARS-CoV-2 , Humans , SARS-CoV-2/genetics , RNA, Ribosomal/metabolism , COVID-19/metabolism , Ribosomes/metabolism , Viral Proteins/metabolism , Viral Nonstructural Proteins/metabolism
3.
Nucleic Acids Res ; 51(19): 10484-10505, 2023 10 27.
Article in English | MEDLINE | ID: mdl-37697435

ABSTRACT

Breast cancer linked with BRCA1/2 mutations commonly recur and resist current therapies, including PARP inhibitors. Given the lack of effective targeted therapies for BRCA1-mutant cancers, we sought to identify novel targets to selectively kill these cancers. Here, we report that loss of RNF8 significantly protects Brca1-mutant mice against mammary tumorigenesis. RNF8 deficiency in human BRCA1-mutant breast cancer cells was found to promote R-loop accumulation and replication fork instability, leading to increased DNA damage, senescence, and synthetic lethality. Mechanistically, RNF8 interacts with XRN2, which is crucial for transcription termination and R-loop resolution. We report that RNF8 ubiquitylates XRN2 to facilitate its recruitment to R-loop-prone genomic loci and that RNF8 deficiency in BRCA1-mutant breast cancer cells decreases XRN2 occupancy at R-loop-prone sites, thereby promoting R-loop accumulation, transcription-replication collisions, excessive genomic instability, and cancer cell death. Collectively, our work identifies a synthetic lethal interaction between RNF8 and BRCA1, which is mediated by a pathological accumulation of R-loops.


Subject(s)
BRCA1 Protein , Breast Neoplasms , Animals , Female , Humans , Mice , BRCA1 Protein/metabolism , BRCA2 Protein/genetics , Breast Neoplasms/genetics , DNA Damage , DNA-Binding Proteins/metabolism , Exoribonucleases/metabolism , Genomic Instability , Neoplasm Recurrence, Local , R-Loop Structures , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Ubiquitination
4.
Nucleic Acids Res ; 51(9): 4341-4362, 2023 05 22.
Article in English | MEDLINE | ID: mdl-36928661

ABSTRACT

BRCA1 mutations are associated with increased breast and ovarian cancer risk. BRCA1-mutant tumors are high-grade, recurrent, and often become resistant to standard therapies. Herein, we performed a targeted CRISPR-Cas9 screen and identified MEPCE, a methylphosphate capping enzyme, as a synthetic lethal interactor of BRCA1. Mechanistically, we demonstrate that depletion of MEPCE in a BRCA1-deficient setting led to dysregulated RNA polymerase II (RNAPII) promoter-proximal pausing, R-loop accumulation, and replication stress, contributing to transcription-replication collisions. These collisions compromise genomic integrity resulting in loss of viability of BRCA1-deficient cells. We also extend these findings to another RNAPII-regulating factor, PAF1. This study identifies a new class of synthetic lethal partners of BRCA1 that exploit the RNAPII pausing regulation and highlight the untapped potential of transcription-replication collision-inducing factors as unique potential therapeutic targets for treating cancers associated with BRCA1 mutations.


Subject(s)
BRCA1 Protein , DNA Replication , Hereditary Breast and Ovarian Cancer Syndrome , Mutation , Transcription, Genetic , Humans , BRCA1 Protein/deficiency , BRCA1 Protein/genetics , DNA Replication/genetics , Hereditary Breast and Ovarian Cancer Syndrome/genetics , Hereditary Breast and Ovarian Cancer Syndrome/pathology , Hereditary Breast and Ovarian Cancer Syndrome/physiopathology , RNA Polymerase II/metabolism , Transcription, Genetic/genetics , Promoter Regions, Genetic , Methyltransferases/deficiency , Methyltransferases/genetics , R-Loop Structures , Cell Death
5.
Nat Commun ; 13(1): 5453, 2022 09 16.
Article in English | MEDLINE | ID: mdl-36114190

ABSTRACT

Survival of motor neuron (SMN) functions in diverse biological pathways via recognition of symmetric dimethylarginine (Rme2s) on proteins by its Tudor domain, and deficiency of SMN leads to spinal muscular atrophy. Here we report a potent and selective antagonist with a 4-iminopyridine scaffold targeting the Tudor domain of SMN. Our structural and mutagenesis studies indicate that both the aromatic ring and imino groups of compound 1 contribute to its selective binding to SMN. Various on-target engagement assays support that compound 1 specifically recognizes SMN in a cellular context and prevents the interaction of SMN with the R1810me2s of RNA polymerase II subunit POLR2A, resulting in transcription termination and R-loop accumulation mimicking SMN depletion. Thus, in addition to the antisense, RNAi and CRISPR/Cas9 techniques, potent SMN antagonists could be used as an efficient tool to understand the biological functions of SMN.


Subject(s)
RNA Polymerase II , SMN Complex Proteins , Humans , Motor Neurons/metabolism , Muscular Atrophy, Spinal/metabolism , RNA Polymerase II/drug effects , RNA Polymerase II/metabolism , SMN Complex Proteins/antagonists & inhibitors , SMN Complex Proteins/drug effects , SMN Complex Proteins/metabolism
6.
STAR Protoc ; 3(4): 101734, 2022 12 16.
Article in English | MEDLINE | ID: mdl-36178790

ABSTRACT

Modulating R-loop triplex nucleic acid structures reveals their roles across the genome. However, common approaches cannot ascribe functions to R-loops in a locus-associated manner. This protocol presents the use of a locus-associated R-loop-modulating system (dubbed LasR), which employs an inducible RNaseH1-EGFP-dCas9 chimaera. We detail the in silico design of sgRNAs and their transfection with the chimaera, and outline steps confirming RNaseH1-EGFP-dCas9 expression, localization, locus-targeted association, and R-loop modulation in cis or trans using immunoblotting, microscopy, and chromatin and DNA-RNA immunoprecipitation. For complete details on the use and execution of this protocol, please refer to Abraham et al. (2020).


Subject(s)
Clustered Regularly Interspaced Short Palindromic Repeats , R-Loop Structures , RNA/genetics , DNA/metabolism , Genome
7.
Trends Genet ; 38(3): 290-304, 2022 03.
Article in English | MEDLINE | ID: mdl-34598804

ABSTRACT

The maintenance of genome stability and cellular homeostasis depends on the temporal and spatial coordination of successive events constituting the classical DNA damage response (DDR). Recent findings suggest close integration and coordination of DDR signaling with specific cellular processes. The mechanisms underlying such coordination remain unclear. We review emerging crosstalk between DNA repair factors, chromatin remodeling, replication, transcription, spatial genome organization, cytoskeletal forces, and liquid-liquid phase separation (LLPS) in mediating DNA repair. We present an overarching DNA repair framework within which these dynamic processes intersect in nuclear space over time. Collectively, this interplay ensures the efficient assembly of DNA repair proteins onto shifting genome structures to preserve genome stability and cell survival.


Subject(s)
Chromatin , DNA Repair , Cell Nucleus/genetics , Cell Nucleus/metabolism , Chromatin/genetics , Chromatin/metabolism , Chromatin Assembly and Disassembly , DNA Damage/genetics , DNA Repair/genetics
8.
Trends Cell Biol ; 31(9): 721-731, 2021 09.
Article in English | MEDLINE | ID: mdl-33902985

ABSTRACT

Microtubules are major cytoskeletal components mediating fundamental cellular processes, including cell division. Recent evidence suggests that microtubules also regulate the nucleus during the cell cycle's interphase stage. Deciphering such roles of microtubules should uncover direct crosstalk between the nucleus and cytoplasm, impacting genome function and organismal health. Here, we review emerging roles for microtubules in interphase genome regulation. We explore how microtubules exert cytoplasmic forces on the nucleus or transport molecular cargo, including DNA, into or within the nucleus. We also describe how microtubules perform these functions by establishing transient or stable connections with nuclear envelope elements. Lastly, we discuss how the regulation of the nucleus by microtubules impacts genome organization and repair. Together, the literature indicates that interphase microtubules are critical regulators of nuclear structure and genome stability.


Subject(s)
Cell Nucleus , Microtubules , Cytoskeleton , Interphase , Nuclear Envelope
9.
J Clin Invest ; 131(3)2021 02 01.
Article in English | MEDLINE | ID: mdl-33529165

ABSTRACT

Germline mutations in BRCA1 and BRCA2 (BRCA1/2) genes considerably increase breast and ovarian cancer risk. Given that tumors with these mutations have elevated genomic instability, they exhibit relative vulnerability to certain chemotherapies and targeted treatments based on poly (ADP-ribose) polymerase (PARP) inhibition. However, the molecular mechanisms that influence cancer risk and therapeutic benefit or resistance remain only partially understood. BRCA1 and BRCA2 have also been implicated in the suppression of R-loops, triple-stranded nucleic acid structures composed of a DNA:RNA hybrid and a displaced ssDNA strand. Here, we report that loss of RNF168, an E3 ubiquitin ligase and DNA double-strand break (DSB) responder, remarkably protected Brca1-mutant mice against mammary tumorigenesis. We demonstrate that RNF168 deficiency resulted in accumulation of R-loops in BRCA1/2-mutant breast and ovarian cancer cells, leading to DSBs, senescence, and subsequent cell death. Using interactome assays, we identified RNF168 interaction with DHX9, a helicase involved in the resolution and removal of R-loops. Mechanistically, RNF168 directly ubiquitylated DHX9 to facilitate its recruitment to R-loop-prone genomic loci. Consequently, loss of RNF168 impaired DHX9 recruitment to R-loops, thereby abrogating its ability to resolve R-loops. The data presented in this study highlight a dependence of BRCA1/2-defective tumors on factors that suppress R-loops and reveal a fundamental RNF168-mediated molecular mechanism that governs cancer development and vulnerability.


Subject(s)
BRCA1 Protein/deficiency , BRCA2 Protein/deficiency , DNA, Neoplasm/metabolism , Genomic Instability , Mammary Neoplasms, Animal/metabolism , Ovarian Neoplasms/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , DNA, Neoplasm/genetics , Female , Genetic Loci , Humans , Mammary Neoplasms, Animal/genetics , Mice , Mice, Knockout , Ovarian Neoplasms/genetics , Ubiquitin-Protein Ligases/genetics
10.
Commun Biol ; 3(1): 773, 2020 12 15.
Article in English | MEDLINE | ID: mdl-33319830

ABSTRACT

Liquid-liquid phase separation (LLPS) has emerged as a central player in the assembly of membraneless compartments termed biomolecular condensates. These compartments are dynamic structures that can condense or dissolve under specific conditions to regulate molecular functions. Such properties allow biomolecular condensates to rapidly respond to changing endogenous or environmental conditions. Here, we review emerging roles for LLPS within the nuclear space, with a specific emphasis on genome organization, expression and repair. Our review highlights the emerging notion that biomolecular condensates regulate the sequential engagement of molecules in multistep biological processes.


Subject(s)
Cell Nucleus/metabolism , Cell Physiological Phenomena , Cell Fractionation , Chemical Fractionation , DNA Repair , Eukaryotic Cells/physiology , Gene Expression Regulation , Genome , Genomics/methods , Heterochromatin/genetics , Heterochromatin/metabolism , Subcellular Fractions
11.
Sci Rep ; 10(1): 16034, 2020 09 29.
Article in English | MEDLINE | ID: mdl-32994470

ABSTRACT

Retrotransposons can represent half of eukaryotic genomes. Retrotransposon dysregulation destabilizes genomes and has been linked to various human diseases. Emerging regulators of retromobility include RNA-DNA hybrid-containing structures known as R-loops. Accumulation of these structures at the transposons of yeast 1 (Ty1) elements has been shown to increase Ty1 retromobility through an unknown mechanism. Here, via a targeted genetic screen, we identified the rnh1Δ rad27Δ yeast mutant, which lacked both the Ty1 inhibitor Rad27 and the RNA-DNA hybrid suppressor Rnh1. The mutant exhibited elevated levels of Ty1 cDNA-associated RNA-DNA hybrids that promoted Ty1 mobility. Moreover, in this rnh1Δ rad27Δ mutant, but not in the double RNase H mutant rnh1Δ rnh201Δ, RNA-DNA hybrids preferentially existed as duplex nucleic acid structures and increased Ty1 mobility in a Rad52-dependent manner. The data indicate that in cells lacking RNA-DNA hybrid and Ty1 repressors, elevated levels of RNA-cDNA hybrids, which are associated with duplex nucleic acid structures, boost Ty1 mobility via a Rad52-dependent mechanism. In contrast, in cells lacking RNA-DNA hybrid repressors alone, elevated levels of RNA-cDNA hybrids, which are associated with triplex nucleic acid structures, boost Ty1 mobility via a Rad52-independent process. We propose that duplex and triplex RNA-DNA hybrids promote transposon mobility via Rad52-dependent or -independent mechanisms.


Subject(s)
R-Loop Structures/genetics , Retroelements/genetics , Retroelements/physiology , DNA, Complementary/genetics , Flap Endonucleases/genetics , Flap Endonucleases/metabolism , RNA/genetics , Rad52 DNA Repair and Recombination Protein/genetics , Rad52 DNA Repair and Recombination Protein/metabolism , Recombination, Genetic/genetics , Ribonuclease H/metabolism , Ribonuclease H/physiology , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism
12.
Nature ; 585(7824): 298-302, 2020 09.
Article in English | MEDLINE | ID: mdl-32669707

ABSTRACT

Proteins are manufactured by ribosomes-macromolecular complexes of protein and RNA molecules that are assembled within major nuclear compartments called nucleoli1,2. Existing models suggest that RNA polymerases I and III (Pol I and Pol III) are the only enzymes that directly mediate the expression of the ribosomal RNA (rRNA) components of ribosomes. Here we show, however, that RNA polymerase II (Pol II) inside human nucleoli operates near genes encoding rRNAs to drive their expression. Pol II, assisted by the neurodegeneration-associated enzyme senataxin, generates a shield comprising triplex nucleic acid structures known as R-loops at intergenic spacers flanking nucleolar rRNA genes. The shield prevents Pol I from producing sense intergenic noncoding RNAs (sincRNAs) that can disrupt nucleolar organization and rRNA expression. These disruptive sincRNAs can be unleashed by Pol II inhibition, senataxin loss, Ewing sarcoma or locus-associated R-loop repression through an experimental system involving the proteins RNaseH1, eGFP and dCas9 (which we refer to as 'red laser'). We reveal a nucleolar Pol-II-dependent mechanism that drives ribosome biogenesis, identify disease-associated disruption of nucleoli by noncoding RNAs, and establish locus-targeted R-loop modulation. Our findings revise theories of labour division between the major RNA polymerases, and identify nucleolar Pol II as a major factor in protein synthesis and nuclear organization, with potential implications for health and disease.


Subject(s)
Cell Nucleolus/enzymology , Cell Nucleolus/genetics , DNA, Ribosomal/genetics , RNA Polymerase II/metabolism , RNA, Untranslated/biosynthesis , RNA, Untranslated/genetics , Ribosomes/metabolism , CRISPR-Associated Protein 9/genetics , CRISPR-Associated Protein 9/metabolism , Cell Line, Tumor , Cell Nucleolus/physiology , DNA Helicases/metabolism , DNA, Intergenic/genetics , Humans , Multifunctional Enzymes/metabolism , Protein Biosynthesis , R-Loop Structures , RNA Helicases/metabolism , RNA Polymerase I/antagonists & inhibitors , RNA Polymerase I/metabolism , Ribonuclease H/metabolism , Ribosomes/chemistry , Ribosomes/genetics , Sarcoma, Ewing/genetics , Sarcoma, Ewing/pathology
13.
Nat Commun ; 11(1): 695, 2020 02 04.
Article in English | MEDLINE | ID: mdl-32019927

ABSTRACT

Cellular processes are influenced by liquid phase separation, but its role in DNA repair is unclear. Here, we show that in Saccharomyces cerevisiae, liquid droplets made up of DNA repair proteins cooperate with different types of DNA damage-inducible intranuclear microtubule filaments (DIMs) to promote the clustering of DNA damage sites and maintain genome stability. Rad52 DNA repair proteins at different DNA damage sites assemble in liquid droplets that fuse into a repair centre droplet via the action of petite DIMs (pti-DIMs). This larger droplet concentrates tubulin and projects short aster-like DIMs (aster-DIMs), which tether the repair centre to longer DIMs mediating the mobilization of damaged DNA to the nuclear periphery for repair. Our findings indicate that cooperation between Rad52 liquid droplets and various types of nuclear filaments promotes the assembly and function of the DNA repair centre.


Subject(s)
DNA Repair , Rad52 DNA Repair and Recombination Protein/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Cell Nucleus/genetics , Cell Nucleus/metabolism , DNA Damage , DNA, Fungal/genetics , Rad52 DNA Repair and Recombination Protein/genetics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics , Tubulin/genetics , Tubulin/metabolism
14.
Trends Cell Biol ; 30(2): 144-156, 2020 02.
Article in English | MEDLINE | ID: mdl-31836279

ABSTRACT

The increased mobility of damaged DNA within the nucleus can promote genome stability and cell survival. New cell biology approaches have indicated that damaged DNA mobility exhibits random and directed movements during DNA repair. Here, we review recent studies that collectively reveal that cooperation between different molecular mechanisms, which underlie increases in the random and directional motion of damaged DNA, can promote genome repair. We also review the latest approaches that can be used to distinguish between random and directed motions of damaged DNA or other biological molecules. Detailed understanding of the mechanisms behind the increased motion of damaged DNA within the nucleus will reveal more of the secrets of genome organization and stability while potentially pointing to novel research and therapeutic tools.


Subject(s)
DNA Damage , DNA Repair , DNA/metabolism , Motion , Chromatin Assembly and Disassembly , Humans , Microtubules/metabolism
15.
Trends Genet ; 35(8): 589-600, 2019 08.
Article in English | MEDLINE | ID: mdl-31155151

ABSTRACT

Genome expression and stability are dependent on biological processes that control repetitive DNA sequences and nuclear compartmentalization. The phase separation of macromolecules has recently emerged as a major player in the control of biological pathways. Here, we summarize recent studies that collectively reveal intersections between phase separation, repetitive DNA elements, and nuclear compartments. These intersections modulate fundamental processes, including gene expression, DNA repair, and cellular lifespan, in the context of health and diseases such as cancer and neurodegeneration.


Subject(s)
Gene Expression Regulation/genetics , Genome/genetics , Neoplasms/genetics , Neurodegenerative Diseases/genetics , Repetitive Sequences, Nucleic Acid/genetics , Aging/genetics , Cell Compartmentation/genetics , DNA Repair/genetics , Genetic Loci/genetics , Humans
16.
Front Cell Dev Biol ; 7: 336, 2019.
Article in English | MEDLINE | ID: mdl-31921848

ABSTRACT

Genetic loci are non-randomly arranged in the nucleus of the cell. This order, which is important to overall genome expression and stability, is maintained by a growing number of factors including the nuclear envelope, various genetic elements and dedicated protein complexes. Here, we review evidence supporting roles for non-coding RNAs (ncRNAs) in the regulation of spatial genome organization and its impact on gene expression and cell survival. Specifically, we discuss how ncRNAs from single-copy and repetitive DNA loci contribute to spatial genome organization by impacting perinuclear chromosome tethering, major nuclear compartments, chromatin looping, and various chromosomal structures. Overall, our analysis of the literature highlights central functions for ncRNAs and their transcription in the modulation of spatial genome organization with connections to human health and disease.

17.
Commun Biol ; 1: 187, 2018.
Article in English | MEDLINE | ID: mdl-30417124

ABSTRACT

Ribosomal DNA (rDNA) repeat instability and protein aggregation are thought to be two major and independent drivers of cellular aging. Pbp1, the yeast ortholog of human ATXN2, maintains rDNA repeat stability and lifespan via suppression of RNA-DNA hybrids. ATXN2 polyglutamine expansion drives neurodegeneration causing spinocerebellar ataxia type 2 and promoting amyotrophic lateral sclerosis. Here, molecular characterization of Pbp1 revealed that its knockout or subjection to disease-modeling polyQ expansion represses Ty1 (Transposons of Yeast) retrotransposons by respectively promoting Trf4-depedendent RNA turnover and Ty1 Gag protein aggregation. This aggregation, but not its impact on retrotransposition, compromises rDNA repeat stability and shortens lifespan by hyper-activating Trf4-dependent turnover of intergenic ncRNA within the repeats. We uncover a function for the conserved Pbp1/ATXN2 proteins in the promotion of retrotransposition, create and describe powerful yeast genetic models of ATXN2-linked neurodegenerative diseases, and connect the major aging mechanisms of rDNA instability and protein aggregation.

18.
Nat Commun ; 9(1): 2567, 2018 07 02.
Article in English | MEDLINE | ID: mdl-29967403

ABSTRACT

Damaged DNA shows increased mobility, which can promote interactions with repair-conducive nuclear pore complexes (NPCs). This apparently random mobility is paradoxically abrogated upon disruption of microtubules or kinesins, factors that typically cooperate to mediate the directional movement of macromolecules. Here, we resolve this paradox by uncovering DNA damage-inducible intranuclear microtubule filaments (DIMs) that mobilize damaged DNA and promote repair. Upon DNA damage, relief of centromeric constraint induces DIMs that cooperate with the Rad9 DNA damage response mediator and Kar3 kinesin motor to capture DNA lesions, which then linearly move along dynamic DIMs. Decreasing and hyper-inducing DIMs respectively abrogates and hyper-activates repair. Accounting for DIM dynamics across cell populations by measuring directional changes of damaged DNA reveals that it exhibits increased non-linear directional behavior in nuclear space. Abrogation of DIM-dependent processes or repair-promoting factors decreases directional behavior. Thus, inducible and dynamic nuclear microtubule filaments directionally mobilize damaged DNA and promote repair.


Subject(s)
Cell Nucleus/metabolism , Chromatin/metabolism , DNA Repair/physiology , Microtubules/metabolism , Saccharomyces cerevisiae/physiology , Cell Cycle Proteins/metabolism , DNA Damage/physiology , Intravital Microscopy , Microtubule-Associated Proteins/metabolism , Saccharomyces cerevisiae Proteins/metabolism
19.
Front Genet ; 9: 157, 2018.
Article in English | MEDLINE | ID: mdl-29741164

ABSTRACT

[This corrects the article on p. 95 in vol. 9, PMID: 29616083.].

20.
Front Genet ; 9: 95, 2018.
Article in English | MEDLINE | ID: mdl-29616083

ABSTRACT

Eukaryotic genomes are non-randomly arranged inside the nucleus. Despite this ordered spatial genome organization, damaged DNA exhibits increased random mobility within nuclear space. This increased random movement is thought to promote DNA repair by facilitating homology search, allowing targeting to repair-conducive nuclear domains, or releasing damage from repair-repressive locations. Recent studies focusing on the relationship between telomeres, DNA repair processes, and nuclear organization have revealed that the disruption of motor proteins or microtubules, which typically mediate the directed motion of cargo, disrupts the random mobility of damaged DNA. These findings define a new biological paradox. Here, I define this as the damaged DNA mobility paradox, describe how it uncovers key gaps in knowledge, and highlight key questions to help guide us toward paradox resolution.

SELECTION OF CITATIONS
SEARCH DETAIL
...