Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Aging Cell ; 20(11): e13484, 2021 11.
Article in English | MEDLINE | ID: mdl-34612580

ABSTRACT

Werner syndrome (WS) is an accelerated aging disorder characterized by genomic instability, which is caused by WRN protein deficiency. WRN participates in DNA metabolism including DNA repair. In a previous report, we showed that WRN protein is recruited to laser-induced DNA double-strand break (DSB) sites during various stages of the cell cycle with similar intensities, supporting that WRN participates in both non-homologous end joining (NHEJ) and homologous recombination (HR). Here, we demonstrate that the phosphorylation of WRN by CDK2 on serine residue 426 is critical for WRN to make its DSB repair pathway choice between NHEJ and HR. Cells expressing WRN engineered to mimic the unphosphorylated or phosphorylation state at serine 426 showed abnormal DSB recruitment, altered RPA interaction, strand annealing, and DSB repair activities. The CDK2 phosphorylation on serine 426 stabilizes WRN's affinity for RPA, likely increasing its long-range resection at the end of DNA strands, which is a crucial step for HR. Collectively, the data shown here demonstrate that a CDK2-dependent phosphorylation of WRN regulates DSB repair pathway choice and cell cycle participation.


Subject(s)
Cyclin-Dependent Kinase 2/metabolism , DNA Breaks, Double-Stranded/radiation effects , DNA End-Joining Repair/genetics , Homologous Recombination , Signal Transduction/genetics , Werner Syndrome Helicase/metabolism , Cell Cycle/genetics , Cell Line, Tumor , Cyclin-Dependent Kinase 2/genetics , DNA/metabolism , HEK293 Cells , Humans , Phosphorylation/genetics , Replication Protein A/metabolism , Serine/metabolism , Transfection , Werner Syndrome/genetics , Werner Syndrome/metabolism , Werner Syndrome Helicase/genetics
2.
Clin Cancer Res ; 25(18): 5584-5594, 2019 09 15.
Article in English | MEDLINE | ID: mdl-31196855

ABSTRACT

PURPOSE: Somatic inactivating mutations in ARID1A, a component of the SWI/SNF chromatin remodeling complex, are detected in various types of human malignancies. Loss of ARID1A compromises DNA damage repair. The induced DNA damage burden may increase reliance on PARP-dependent DNA repair of cancer cells to maintain genome integrity and render susceptibility to PARP inhibitor therapy.Experimental Design: Isogenic ARID1A-/- and wild-type cell lines were used for assessing DNA damage response, DNA compactness, and profiling global serine/threonine phosphoproteomic in vivo. A panel of inhibitors targeting DNA repair pathways was screened for a synergistic antitumor effect with irradiation in ARID1A-/- tumors. RESULTS: ARID1A-deficient endometrial cells exhibit sustained levels in DNA damage response, a result further supported by in vivo phosphoproteomic analysis. Our results show that ARID1A is essential for establishing an open chromatin state upon DNA damage, a process required for recruitment of 53BP1 and RIF1, key mediators of non-homologous end-joining (NHEJ) machinery, to DNA lesions. The inability of ARID1A-/- cells to mount NHEJ repair results in a partial cytotoxic response to radiation. Small-molecule compound screens revealed that PARP inhibitors act synergistically with radiation to potentiate cytotoxicity in ARID1A-/- cells. Combination treatment with low-dose radiation and olaparib greatly improved antitumor efficacy, resulting in long-term remission in mice bearing ARID1A-deficient tumors. CONCLUSIONS: ARID1A-deficient cells acquire high sensitivity to PARP inhibition after exposure to exogenously induced DNA breaks such as ionizing radiation. Our findings suggest a novel biologically informed strategy for treating ARID1A-deficient malignancies.


Subject(s)
DNA-Binding Proteins/deficiency , Drug Resistance, Neoplasm/genetics , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Radiation Tolerance/genetics , Transcription Factors/deficiency , Animals , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cell Survival/drug effects , DNA Breaks, Double-Stranded , DNA Damage , DNA End-Joining Repair , DNA Repair , Disease Models, Animal , Gene Knockdown Techniques , Humans , Mice , Mice, Transgenic , Models, Biological
3.
Nat Commun ; 8(1): 2039, 2017 12 11.
Article in English | MEDLINE | ID: mdl-29229926

ABSTRACT

Pathway choice within DNA double-strand break (DSB) repair is a tightly regulated process to maintain genome integrity. RECQL4, deficient in Rothmund-Thomson Syndrome, promotes the two major DSB repair pathways, non-homologous end joining (NHEJ) and homologous recombination (HR). Here we report that RECQL4 promotes and coordinates NHEJ and HR in different cell cycle phases. RECQL4 interacts with Ku70 to promote NHEJ in G1 when overall cyclin-dependent kinase (CDK) activity is low. During S/G2 phases, CDK1 and CDK2 (CDK1/2) phosphorylate RECQL4 on serines 89 and 251, enhancing MRE11/RECQL4 interaction and RECQL4 recruitment to DSBs. After phosphorylation, RECQL4 is ubiquitinated by the DDB1-CUL4A E3 ubiquitin ligase, which facilitates its accumulation at DSBs. Phosphorylation of RECQL4 stimulates its helicase activity, promotes DNA end resection, increases HR and cell survival after ionizing radiation, and prevents cellular senescence. Collectively, we propose that RECQL4 modulates the pathway choice of NHEJ and HR in a cell cycle-dependent manner.


Subject(s)
Cell Cycle , DNA Breaks, Double-Stranded , DNA End-Joining Repair , RecQ Helicases/metabolism , Recombinational DNA Repair , Ubiquitination , Cell Line, Tumor , Cullin Proteins/genetics , Cullin Proteins/metabolism , Cyclin-Dependent Kinases/genetics , Cyclin-Dependent Kinases/metabolism , HEK293 Cells , Humans , Ku Autoantigen/genetics , Ku Autoantigen/metabolism , Phosphorylation , Protein Binding , RNA Interference , RecQ Helicases/genetics
4.
F1000Res ; 6: 1779, 2017.
Article in English | MEDLINE | ID: mdl-29043077

ABSTRACT

Aging, the universal phenomenon, affects human health and is the primary risk factor for major disease pathologies. Progeroid diseases, which mimic aging at an accelerated rate, have provided cues in understanding the hallmarks of aging. Mutations in DNA repair genes as well as in telomerase subunits are known to cause progeroid syndromes. Werner syndrome (WS), which is characterized by accelerated aging, is an autosomal-recessive genetic disorder. Hallmarks that define the aging process include genomic instability, telomere attrition, epigenetic alterations, loss of proteostasis, deregulation of nutrient sensing, mitochondrial dysfunction, cellular senescence, stem cell exhaustion, and altered intercellular communication. WS recapitulates these hallmarks of aging and shows increased incidence and early onset of specific cancers. Genome integrity and stability ensure the normal functioning of the cell and are mainly guarded by the DNA repair machinery and telomeres. WRN, being a RecQ helicase, protects genome stability by regulating DNA repair pathways and telomeres. Recent advances in WS research have elucidated WRN's role in DNA repair pathway choice regulation, telomere maintenance, resolution of complex DNA structures, epigenetic regulation, and stem cell maintenance.

5.
Nat Commun ; 7: 13785, 2016 12 06.
Article in English | MEDLINE | ID: mdl-27922005

ABSTRACT

Werner syndrome (WS) is an accelerated ageing disorder with genomic instability caused by WRN protein deficiency. Many features seen in WS can be explained by the diverse functions of WRN in DNA metabolism. However, the origin of the large genomic deletions and telomere fusions are not yet understood. Here, we report that WRN regulates the pathway choice between classical (c)- and alternative (alt)-nonhomologous end joining (NHEJ) during DNA double-strand break (DSB) repair. It promotes c-NHEJ via helicase and exonuclease activities and inhibits alt-NHEJ using non-enzymatic functions. When WRN is recruited to the DSBs it suppresses the recruitment of MRE11 and CtIP, and protects the DSBs from 5' end resection. Moreover, knockdown of Wrn, alone or in combination with Trf2 in mouse embryonic fibroblasts results in increased telomere fusions, which were ablated by Ctip knockdown. We show that WRN regulates alt-NHEJ and shields DSBs from MRE11/CtIP-mediated resection to prevent large deletions and telomere fusions.


Subject(s)
DNA Breaks, Double-Stranded , DNA End-Joining Repair , DNA Repair , Werner Syndrome Helicase/metabolism , Animals , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Line , Cell Line, Tumor , Endodeoxyribonucleases , Humans , MRE11 Homologue Protein/genetics , MRE11 Homologue Protein/metabolism , Mice , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , RNA Interference , Telomere/genetics , Telomere/metabolism , Werner Syndrome/genetics , Werner Syndrome/metabolism , Werner Syndrome/pathology , Werner Syndrome Helicase/genetics
6.
Cell Metab ; 24(4): 566-581, 2016 10 11.
Article in English | MEDLINE | ID: mdl-27732836

ABSTRACT

Ataxia telangiectasia (A-T) is a rare autosomal recessive disease characterized by progressive neurodegeneration and cerebellar ataxia. A-T is causally linked to defects in ATM, a master regulator of the response to and repair of DNA double-strand breaks. The molecular basis of cerebellar atrophy and neurodegeneration in A-T patients is unclear. Here we report and examine the significance of increased PARylation, low NAD+, and mitochondrial dysfunction in ATM-deficient neurons, mice, and worms. Treatments that replenish intracellular NAD+ reduce the severity of A-T neuropathology, normalize neuromuscular function, delay memory loss, and extend lifespan in both animal models. Mechanistically, treatments that increase intracellular NAD+ also stimulate neuronal DNA repair and improve mitochondrial quality via mitophagy. This work links two major theories on aging, DNA damage accumulation, and mitochondrial dysfunction through nuclear DNA damage-induced nuclear-mitochondrial signaling, and demonstrates that they are important pathophysiological determinants in premature aging of A-T, pointing to therapeutic interventions.


Subject(s)
Ataxia Telangiectasia/pathology , DNA Repair/drug effects , Health , Longevity/drug effects , Mitophagy/drug effects , NAD/pharmacology , Animals , Ataxia Telangiectasia Mutated Proteins/deficiency , Ataxia Telangiectasia Mutated Proteins/metabolism , Behavior, Animal , Caenorhabditis elegans/metabolism , Caenorhabditis elegans/ultrastructure , Cells, Cultured , Disease Models, Animal , Gene Knockdown Techniques , Homeostasis/drug effects , Metabolomics , Mice , Neurons/drug effects , Neurons/metabolism , Phenotype , Phthalazines/pharmacology , Piperazines/pharmacology , Proteomics , Rats, Sprague-Dawley , Signal Transduction/drug effects , Sirtuin 1/metabolism
7.
Cell Rep ; 16(1): 161-173, 2016 06 28.
Article in English | MEDLINE | ID: mdl-27320928

ABSTRACT

The RecQ helicase RECQL4, mutated in Rothmund-Thomson syndrome, regulates genome stability, aging, and cancer. Here, we identify a crucial role for RECQL4 in DNA end resection, which is the initial and an essential step of homologous recombination (HR)-dependent DNA double-strand break repair (DSBR). Depletion of RECQL4 severely reduces HR-mediated repair and 5' end resection in vivo. RECQL4 physically interacts with MRE11-RAD50-NBS1 (MRN), which senses DSBs and initiates DNA end resection with CtIP. The MRE11 exonuclease regulates the retention of RECQL4 at laser-induced DSBs. RECQL4 also directly interacts with CtIP via its N-terminal domain and promotes CtIP recruitment to the MRN complex at DSBs. Moreover, inactivation of RECQL4's helicase activity impairs DNA end processing and HR-dependent DSBR without affecting its interaction with MRE11 and CtIP, suggesting an important role for RECQL4's unwinding activity in the process. Thus, we report that RECQL4 is an important participant in HR-dependent DSBR.


Subject(s)
DNA Breaks, Double-Stranded , DNA End-Joining Repair , RecQ Helicases/metabolism , Carrier Proteins , Cell Line, Tumor , DNA Repair Enzymes/metabolism , Exodeoxyribonucleases/metabolism , Humans , MRE11 Homologue Protein/metabolism , Nuclear Proteins , Recombinational DNA Repair , Replication Protein A/metabolism , Repressor Proteins
8.
Oncotarget ; 7(12): 13269-84, 2016 Mar 22.
Article in English | MEDLINE | ID: mdl-26959889

ABSTRACT

Werner syndrome protein (WRN) is a RecQ helicase that participates in DNA repair, genome stability and cellular senescence. The five human RecQ helicases, RECQL1, Bloom, WRN, RECQL4 and RECQL5 play critical roles in DNA repair and cell survival after treatment with the anticancer drug camptothecin (CPT). CPT derivatives are widely used in cancer chemotherapy to inhibit topoisomerase I and generate DNA double-strand breaks during replication. Here we studied the effects of CPT on the stability and expression dynamics of human RecQ helicases. In the cells treated with CPT, we observed distinct effects on WRN compared to other human RecQ helicases. CPT altered the cellular localization of WRN and induced its degradation by a ubiquitin-mediated proteasome pathway. WRN knockdown cells as well as CPT treated cells became senescent and stained positive for senescence-associated ß-galactosidase at a higher frequency compared to control cells. However, the senescent phenotype was attenuated by ectopic expression of WRN suggesting functional implication of WRN degradation in CPT treated cells. Approximately 5-23% of breast cancer tumors are known to respond to CPT-based chemotherapy. Interestingly, we found that the extent of CPT-induced WRN degradation correlates with increasing sensitivity of breast cancer cells to CPT. The abundance of WRN decreased in CPT-treated sensitive cells; however, WRN remained relatively stable in CPT-resistant breast cancer cells. In a large clinical cohort of breast cancer patients, we find that WRN and topoisomerase I expression correlate with an aggressive tumor phenotype and poor prognosis. Our novel observations suggest that WRN abundance along with CPT-induced degradation could be a promising strategy for personalizing CPT-based cancer chemotherapeutic regimens.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Breast Neoplasms/drug therapy , Camptothecin/pharmacology , DNA Topoisomerases, Type I/metabolism , Drug Resistance, Neoplasm , Gene Expression Regulation, Neoplastic/drug effects , Werner Syndrome Helicase/metabolism , Apoptosis , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Proliferation , DNA Topoisomerases, Type I/genetics , Female , Humans , Prognosis , Survival Rate , Tumor Cells, Cultured , Werner Syndrome Helicase/genetics
9.
Mol Cell Biol ; 35(23): 3974-89, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26391948

ABSTRACT

Poly(ADP-ribose) (PAR) polymerase 1 (PARP1) catalyzes the poly(ADP-ribosyl)ation (PARylation) of proteins, a posttranslational modification which forms the nucleic acid-like polymer PAR. PARP1 and PAR are integral players in the early DNA damage response, since PARylation orchestrates the recruitment of repair proteins to sites of damage. Human RecQ helicases are DNA unwinding proteins that are critical responders to DNA damage, but how their recruitment and activities are regulated by PARPs and PAR is poorly understood. Here we report that all human RecQ helicases interact with PAR noncovalently. Furthermore, we define the effects that PARP1, PARylated PARP1, and PAR have on RECQL5 and WRN, using both in vitro and in vivo assays. We show that PARylation is involved in the recruitment of RECQL5 and WRN to laser-induced DNA damage and that RECQL5 and WRN have differential responses to PARylated PARP1 and PAR. Furthermore, we show that the loss of RECQL5 or WRN resulted in increased sensitivity to PARP inhibition. In conclusion, our results demonstrate that PARP1 and PAR actively, and in some instances differentially, regulate the activities and cellular localization of RECQL5 and WRN, suggesting that PARylation acts as a fine-tuning mechanism to coordinate their functions in time and space during the genotoxic stress response.


Subject(s)
Exodeoxyribonucleases/metabolism , Poly Adenosine Diphosphate Ribose/metabolism , Poly(ADP-ribose) Polymerases/metabolism , Protein Interaction Maps , RecQ Helicases/metabolism , Adenosine Triphosphatases/metabolism , DNA Breaks, Double-Stranded , DNA Repair , Enzyme Activation/drug effects , HEK293 Cells , HeLa Cells , Humans , Poly (ADP-Ribose) Polymerase-1 , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Werner Syndrome Helicase
10.
PLoS One ; 9(12): e114155, 2014.
Article in English | MEDLINE | ID: mdl-25531437

ABSTRACT

Extracellular Tat (eTat) plays an important role in HIV-1 pathogenesis. The presence of anti-Tat antibodies is negatively correlated with disease progression, hence making Tat a potential vaccine candidate. The cytotoxicity and moderate immunogenicity of Tat however remain impediments for developing Tat-based vaccines. Here, we report a novel strategy to concurrently enhance the immunogenicity and safety profile of Tat. The grafting of universal helper T-lymphocyte (HTL) epitopes, Pan DR Epitope (PADRE) and Pol711 into the cysteine rich domain (CRD) and the basic domain (BD) abolished the transactivation potential of the Tat protein. The HTL-Tat proteins elicited a significantly higher titer of antibodies as compared to the wild-type Tat in BALB/c mice. While the N-terminal epitope remained immunodominant in HTL-Tat immunizations, an additional epitope in exon-2 was recognized with comparable magnitude suggesting a broader immune recognition. Additionally, the HTL-Tat proteins induced cross-reactive antibodies of high avidity that efficiently neutralized exogenous Tat, thus blocking the activation of a Tat-defective provirus. With advantages such as presentation of multiple B-cell epitopes, enhanced antibody response and importantly, transactivation-deficient Tat protein, this approach has potential application for the generation of Tat-based HIV/AIDS vaccines.


Subject(s)
Epitopes, T-Lymphocyte/immunology , HIV-1/immunology , Safety , T-Lymphocytes, Helper-Inducer/immunology , tat Gene Products, Human Immunodeficiency Virus/immunology , AIDS Vaccines/adverse effects , AIDS Vaccines/immunology , Animals , Antibodies, Viral/immunology , Cell Line , Engineering , Epitopes, B-Lymphocyte/immunology , Extracellular Space/metabolism , Female , Genetic Vectors/genetics , Immunization , Mice , Protein Structure, Tertiary , Th1 Cells/immunology , Th2 Cells/immunology , Transcriptional Activation/immunology , tat Gene Products, Human Immunodeficiency Virus/chemistry , tat Gene Products, Human Immunodeficiency Virus/genetics , tat Gene Products, Human Immunodeficiency Virus/metabolism
11.
Crit Rev Biochem Mol Biol ; 49(6): 463-72, 2014.
Article in English | MEDLINE | ID: mdl-25048400

ABSTRACT

DNA double-strand breaks are highly toxic DNA lesions that cause genomic instability, if not efficiently repaired. RecQ helicases are a family of highly conserved proteins that maintain genomic stability through their important roles in several DNA repair pathways, including DNA double-strand break repair. Double-strand breaks can be repaired by homologous recombination (HR) using sister chromatids as templates to facilitate precise DNA repair, or by an HR-independent mechanism known as non-homologous end-joining (NHEJ) (error-prone). NHEJ is a non-templated DNA repair process, in which DNA termini are directly ligated. Canonical NHEJ requires DNA-PKcs and Ku70/80, while alternative NHEJ pathways are DNA-PKcs and Ku70/80 independent. This review discusses the role of RecQ helicases in NHEJ, alternative (or back-up) NHEJ (B-NHEJ) and microhomology-mediated end-joining (MMEJ) in V(D)J recombination, class switch recombination and telomere maintenance.


Subject(s)
DNA End-Joining Repair , RecQ Helicases/metabolism , Aging , Animals , DNA/genetics , DNA/metabolism , Humans , Neoplasms/genetics , Neoplasms/metabolism , Telomere/genetics , Telomere/metabolism , V(D)J Recombination
12.
Carcinogenesis ; 35(11): 2415-24, 2014 Nov.
Article in English | MEDLINE | ID: mdl-24942867

ABSTRACT

RECQL4, a member of the RecQ helicase family, is a multifunctional participant in DNA metabolism. RECQL4 protein participates in several functions both in the nucleus and in the cytoplasm of the cell, and mutations in human RECQL4 are associated with three genetic disorders: Rothmund-Thomson, RAPADILINO and Baller-Gerold syndromes. We previously reported that RECQL4 is recruited to laser-induced DNA double-strand breaks (DSB). Here, we have characterized the functional roles of RECQL4 in the non-homologous end joining (NHEJ) pathway of DSB repair. In an in vitro NHEJ assay that depends on the activity of DNA-dependent protein kinase (DNA-PK), extracts from RECQL4 knockdown cells display reduced end-joining activity on DNA substrates with cohesive and non-cohesive ends. Depletion of RECQL4 also reduced the end joining activity on a GFP reporter plasmid in vivo. Knockdown of RECQL4 increased the sensitivity of cells to γ-irradiation and resulted in accumulation of 53BP1 foci after irradiation, indicating defects in the processing of DSB. We find that RECQL4 interacts with the Ku70/Ku80 heterodimer, part of the DNA-PK complex, via its N-terminal domain. Further, RECQL4 stimulates higher order DNA binding of Ku70/Ku80 to a blunt end DNA substrate. Taken together, these results implicate that RECQL4 participates in the NHEJ pathway of DSB repair via a functional interaction with the Ku70/Ku80 complex. This is the first study to provide both in vitro and in vivo evidence for a role of a RecQ helicase in NHEJ.


Subject(s)
Antigens, Nuclear/genetics , DNA End-Joining Repair/genetics , DNA-Binding Proteins/genetics , RecQ Helicases/genetics , Rothmund-Thomson Syndrome/genetics , DNA Breaks, Double-Stranded/radiation effects , DNA-Activated Protein Kinase/genetics , Gamma Rays , Gene Knockdown Techniques , HeLa Cells , Humans , Intracellular Signaling Peptides and Proteins/genetics , Ku Autoantigen , Radiation Tolerance/genetics , RecQ Helicases/antagonists & inhibitors , Rothmund-Thomson Syndrome/pathology , Tumor Suppressor p53-Binding Protein 1
13.
Neurobiol Aging ; 35(6): 1293-300, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24485507

ABSTRACT

Alzheimer's disease (AD) is a senile dementia with increased incidence in older subjects (age >65 years). One of the earliest markers of AD is oxidative DNA damage. Recently, it has been reported that preclinical AD patient brains show elevated levels of oxidative damage in both nuclear and mitochondrial nucleic acids. Moreover, different oxidative lesions in mitochondrial DNA are between 5- and 10-fold higher than in nuclear DNA in both control and AD postmortem brains. We previously showed that there is a significant loss of base excision repair (BER) components in whole tissue extracts of AD and mild cognitive impairment subjects relative to matched control subjects. However, comprehensive analysis of specific steps in BER levels in mitochondrial extracts of AD patient brains is not available. In this study, we mainly investigated various components of BER in mitochondrial extracts of AD and matched control postmortem brain samples. We found that the 5-hydroxyuracil incision and ligase activities are significantly lower in AD brains, whereas the uracil incision, abasic site cleavage, and deoxyribonucleotide triphosphate incorporation activities are normal in these samples.


Subject(s)
Alzheimer Disease/genetics , DNA Damage/genetics , DNA Repair/genetics , DNA, Mitochondrial/genetics , Mitochondria/genetics , Aged , Aged, 80 and over , Brain/metabolism , Brain/ultrastructure , DNA Ligases/metabolism , Dideoxynucleotides/metabolism , Humans , Middle Aged , Mitochondria/metabolism , Mitochondria/pathology , Oxidative Stress/genetics , Uracil/analogs & derivatives , Uracil/metabolism
14.
Mol Cell Biol ; 31(23): 4832-43, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21969602

ABSTRACT

Nuclear factor 90 (NF90), an RNA-binding protein implicated in the regulation of gene expression, exists as a heterodimeric complex with NF45. We previously reported that depletion of the NF90/NF45 complex results in a multinucleated phenotype. Time-lapse microscopy revealed that binucleated cells arise by incomplete abscission of progeny cells followed by fusion. Multinucleate cells arose through aberrant division of binucleated cells and displayed abnormal metaphase plates and anaphase chromatin bridges suggestive of DNA repair defects. NF90 and NF45 are known to interact with the DNA-dependent protein kinase (DNA-PK), which is involved in telomere maintenance and DNA repair by nonhomologous end joining (NHEJ). We hypothesized that NF90 modulates the activity of DNA-PK. In an in vitro NHEJ assay system, DNA end joining was reduced by NF90/NF45 immunodepletion or by RNA digestion to an extent similar to that for catalytic subunit DNA-PKcs immunodepletion. In vivo, NF90/NF45-depleted cells displayed increased γ-histone 2A.X foci, indicative of an accumulation of double-strand DNA breaks (DSBs), and increased sensitivity to ionizing radiation consistent with decreased DSB repair. Further, NF90/NF45 knockdown reduced end-joining activity in vivo. These results identify the NF90/NF45 complex as a regulator of DNA damage repair mediated by DNA-PK and suggest that structured RNA may modulate this process.


Subject(s)
DNA Breaks, Double-Stranded , DNA End-Joining Repair , Multiprotein Complexes/metabolism , Nuclear Factor 45 Protein/metabolism , Nuclear Factor 90 Proteins/metabolism , Antigens, Nuclear/metabolism , Cell Fusion , Cell Nucleus/metabolism , DNA/metabolism , DNA/radiation effects , DNA-Activated Protein Kinase/metabolism , DNA-Binding Proteins/metabolism , Enzyme Assays , Gene Knockdown Techniques , HeLa Cells , Humans , Immunoprecipitation , Ku Autoantigen , Microscopy, Confocal , Microscopy, Fluorescence , Nuclear Factor 45 Protein/genetics , Nuclear Factor 90 Proteins/genetics , Nuclear Proteins/metabolism , RNA Interference , Time-Lapse Imaging
15.
J Mol Biol ; 410(5): 917-32, 2011 Jul 29.
Article in English | MEDLINE | ID: mdl-21763496

ABSTRACT

Human immunodeficiency virus (HIV) exploits cellular proteins during its replicative cycle and latent infection. The positive transcription elongation factor b (P-TEFb) is a key cellular transcription factor critical for these viral processes and is a drug target. During viral replication, P-TEFb is recruited via interactions of its cyclin T1 subunit with the HIV Tat (transactivator of transcription) protein and TAR (transactivation response) element. Through RNA silencing and over-expression experiments, we discovered that nuclear factor 90 (NF90), a cellular RNA binding protein, regulates P-TEFb expression. NF90 depletion reduced cyclin T1 protein levels by inhibiting translation initiation. Regulation was mediated by the 3' untranslated region of cyclin T1 mRNA independently of microRNAs. Cyclin T1 induction is involved in the escape of HIV-1 from latency. We show that the activation of viral replication by phorbol ester in latently infected monocytic cells requires the posttranscriptional induction of NF90 and cyclin T1, implicating NF90 in protein kinase C signaling pathways. This investigation reveals a novel mechanism of cyclin T1 regulation and establishes NF90 as a regulator of HIV-1 replication during both productive infection and induction from latency.


Subject(s)
Cyclin T/genetics , HIV-1/physiology , Protein Biosynthesis , Virus Latency/physiology , Virus Replication/physiology , 3' Untranslated Regions/genetics , Cyclin T/metabolism , Cyclin-Dependent Kinase 9/genetics , Gene Expression Regulation, Viral , Gene Knockdown Techniques , HIV Core Protein p24/metabolism , HIV-1/genetics , HeLa Cells , Humans , Nuclear Factor 90 Proteins/metabolism , Positive Transcriptional Elongation Factor B/metabolism , Promoter Regions, Genetic/genetics , Transcription, Genetic , Virus Latency/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...