Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
1.
J Invest Dermatol ; 143(12): 2494-2506.e4, 2023 12.
Article in English | MEDLINE | ID: mdl-37236596

ABSTRACT

Skin pigmentation is paused after sun exposure; however, the mechanism behind this pausing is unknown. In this study, we found that the UVB-induced DNA repair system, led by the ataxia telangiectasia mutated (ATM) protein kinase, represses MITF transcriptional activity of pigmentation genes while placing MITF in DNA repair mode, thus directly inhibiting pigment production. Phosphoproteomics analysis revealed ATM to be the most significantly enriched pathway among all UVB-induced DNA repair systems. ATM inhibition in mouse or human skin, either genetically or chemically, induces pigmentation. Upon UVB exposure, MITF transcriptional activation is blocked owing to ATM-dependent phosphorylation of MITF on S414, which modifies MITF activity and interactome toward DNA repair, including binding to TRIM28 and RBBP4. Accordingly, MITF genome occupancy is enriched in sites of high DNA damage that are likely repaired. This suggests that ATM harnesses the pigmentation key activator for the necessary rapid, efficient DNA repair, thus optimizing the chances of the cell surviving. Data are available from ProteomeXchange with the identifier PXD041121.


Subject(s)
Ataxia Telangiectasia , Humans , Animals , Mice , Skin Pigmentation/genetics , DNA Repair , Ataxia Telangiectasia Mutated Proteins/genetics , Ataxia Telangiectasia Mutated Proteins/metabolism , Signal Transduction , DNA Damage , Phosphorylation , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Microphthalmia-Associated Transcription Factor/genetics , Microphthalmia-Associated Transcription Factor/metabolism
2.
Aging Cell ; 22(8): e13869, 2023 08.
Article in English | MEDLINE | ID: mdl-37254625

ABSTRACT

The genetic disorder, ataxia-telangiectasia (A-T), is caused by loss of the homeostatic protein kinase, ATM, and combines genome instability, tissue degeneration, cancer predisposition, and premature aging. Primary fibroblasts from A-T patients exhibit premature senescence when grown at ambient oxygen concentration (21%). Here, we show that reducing oxygen concentration to a physiological level range (3%) dramatically extends the proliferative lifespan of human A-T skin fibroblasts. However, they still undergo senescence earlier than control cells grown under the same conditions and exhibit high genome instability. Comparative RNA-seq analysis of A-T and control fibroblasts cultured at 3% oxygen followed by cluster analysis of differentially expressed genes and functional enrichment analysis, revealed distinct transcriptional dynamics in A-T fibroblasts senescing in physiological oxygen concentration. While some transcriptional patterns were similar to those observed during replicative senescence of control cells, others were unique to the senescing A-T cells. We observed in them a robust activation of interferon-stimulated genes, with undetected expression the interferon genes themselves. This finding suggests an activation of a non-canonical cGAS-STING-mediated pathway, which presumably responds to cytosolic DNA emanating from extranuclear micronuclei detected in these cells. Senescing A-T fibroblasts also exhibited a marked, intriguely complex alteration in the expression of genes associated with extracellular matrix (ECM) remodeling. Notably, many of the induced ECM genes encode senescence-associated secretory phenotype (SASP) factors known for their paracrine pro-fibrotic effects. Our data provide a molecular dimension to the segmental premature aging observed in A-T patients and its associated symptoms, which develop as the patients advance in age.


Subject(s)
Aging, Premature , Ataxia Telangiectasia , Humans , Ataxia Telangiectasia/genetics , Ataxia Telangiectasia/metabolism , Aging, Premature/genetics , Aging, Premature/metabolism , Oxygen/metabolism , Cells, Cultured , Cellular Senescence , Fibroblasts/metabolism , Genomic Instability
3.
Front Pediatr ; 11: 1097779, 2023.
Article in English | MEDLINE | ID: mdl-36937967

ABSTRACT

Objective and aim: Infantile-onset inflammatory bowel disease (IO-IBD), defined as IBD diagnosed at age 2 years or younger, tends to be more severe and refractory to conventional treatment than IBD diagnosed at a later age. However, data about IO-IBD and its long-term follow up are limited. We thus aimed to evaluate the presentation and long-term outcomes of patients with IO-IBD in a retrospective multicenter study. Methods: Medical records of patients diagnosed with IO-IBD in eight medical centers during 2000-2017 with at least 1-year follow up were reviewed. Demographics and disease characteristics at diagnosis including age of onset, disease phenotype and location, surgeries, medical therapy, and comorbid conditions were recorded. Results: Twenty-three patients with IO-IBD (16 males, 70%) were identified and followed for a median (range) of 51.2 (26.0-110.3) months. The mean ages at presentation and at the last follow up were 14 ± 9.8 and 101 ± 77 months, respectively. Six (26%) patients needed ileostomy already at the time of diagnosis and 20 (87%) were treated with corticosteroids. During long-term follow up, remission was achieved in 16 (73%) patients; of whom, 3 (14%) were without medications and 7 (32%) were in remission with the use of 5-aminosalicylic acid only. One patient needed hemicolectomy and one developed a severe EBV related infection. Conclusion: The majority of patients with IO-IBD achieved long-term remission, despite a severe disease presentation at diagnosis. Surgery rate however is high, mainly during the first months from diagnosis.

4.
Otol Neurotol ; 42(5): 666-670, 2021 06 01.
Article in English | MEDLINE | ID: mdl-33967243

ABSTRACT

OBJECTIVE: While COVID-19 symptoms impact rhinology (anosmia) and laryngology (airways), two major disciplines of the otolaryngology armamentarium, the virus has seemed to spare the auditory system. A recent study, however, reported changes in otoacoustic emission (OAE) signals measured in SARS-COV-2 positive patients. We sought to assess the effect of COVID-19 infection on auditory performance in a cohort of recovered SARS-COV-2 patients and controls. To avoid a potential bias of previous audiological dysfunction not related to SARS-COV-2 infection, the study encompasses patients with normal auditory history. We hypothesized that if SARS-COV-2 infection predisposes to hearing loss, we would observe subtle and early audiometric deficits in our cohort in the form of subclinical auditory changes. STUDY DESIGN: Cross-sectional study. SETTING: Tertiary referral center. PATIENTS: The Institutional Review Board approved the study and we recruited participants who had been positive for SARS-COV-2 infection, according to an Reverse Transcription Polymerase Chain Reaction (RT-PCR) test on two nasopharyngeal swabs. The patients included in this study were asymptomatic for the SARS-COV-2 infection and were evaluated following recovery, confirmed by repeated swab testing. The control group comprised healthy individuals matched for age and sex, and with a normal auditory and otologic history. INTERVENTIONS: The eligibility to participate in this study included a normal audiogram, no previous auditory symptoms, normal otoscopy examination with an intact tympanic membrane, and bilateral tympanometry type A. None of our volunteers reported any new auditory symptoms following SARS-COV-2 infection. Ototacoustic emissions (OAE) and auditory brainstem response (ABR) measurements were used to evaluate the auditory function. MAIN OUTCOME MEASURES: OAE and ABR measurements. RESULTS: We have found no significant differences between recovered asymptomatic SARS-COV-2 patients and controls in any of transitory evoked otoacoustic emission (TEOAE), distortion product otoacoustic emissions (DPOAE), or ABR responses. CONCLUSIONS: There is no cochlear dysfunction represented by ABR, TEOAE, and DPOAE responses in recovered COVID-19 asymptomatic patients. Retrocochlear function was also preserved as evident by the ABR responses. A long-term evaluation of a larger cohort of SARS-COV-2 patients will help to identify a possible contribution of SARS-COV-2 infection to recently published anecdotal auditory symptoms associated with COVID-19.


Subject(s)
COVID-19 , SARS-CoV-2 , Cross-Sectional Studies , Evoked Potentials, Auditory, Brain Stem , Humans , Otoacoustic Emissions, Spontaneous
5.
Mol Cell Oncol ; 8(2): 1881395, 2021.
Article in English | MEDLINE | ID: mdl-33860085

ABSTRACT

The DNA damage response is robustly activated by DNA double-strand breaks and controlled by three apical protein kinases of the PI3-kinase-related protein kinase (PIKK) family: ataxia-telangiectasia, mutated (ATM), ataxia-telangiectasia and Rad3-related (ATR) and DNA-dependent protein kinase (DNA-PK). Phosphoproteomic analysis reveals the relative share of these PIKKs in coordinating this network, and compensation by ATR and DNA-PK for ATM absence in the genetic disorder, ataxia-telangiectasia (A-T).

6.
EMBO J ; 40(2): e104400, 2021 01 15.
Article in English | MEDLINE | ID: mdl-33215756

ABSTRACT

The DNA damage response (DDR) is a complex signaling network that relies on cascades of protein phosphorylation, which are initiated by three protein kinases of the family of PI3-kinase-related protein kinases (PIKKs): ATM, ATR, and DNA-PK. ATM is missing or inactivated in the genome instability syndrome, ataxia-telangiectasia (A-T). The relative shares of these PIKKs in the response to genotoxic stress and the functional relationships among them are central questions in the genome stability field. We conducted a comprehensive phosphoproteomic analysis in human wild-type and A-T cells treated with the double-strand break-inducing chemical, neocarzinostatin, and validated the results with the targeted proteomic technique, selected reaction monitoring. We also matched our results with 34 published screens for DDR factors, creating a valuable resource for identifying strong candidates for novel DDR players. We uncovered fine-tuned dynamics between the PIKKs following genotoxic stress, such as DNA-PK-dependent attenuation of ATM. In A-T cells, partial compensation for ATM absence was provided by ATR and DNA-PK, with distinct roles and kinetics. The results highlight intricate relationships between these PIKKs in the DDR.


Subject(s)
DNA Damage/genetics , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation/genetics , Ataxia Telangiectasia Mutated Proteins/genetics , Ataxia Telangiectasia Mutated Proteins/metabolism , Cell Line , Cell Line, Tumor , DNA Breaks, Double-Stranded , DNA Repair/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , HeLa Cells , Humans , Phosphatidylinositol 3-Kinases/genetics , Proteomics/methods , Signal Transduction/genetics
7.
Cell ; 176(3): 505-519.e22, 2019 01 24.
Article in English | MEDLINE | ID: mdl-30612738

ABSTRACT

Genomic instability can be a hallmark of both human genetic disease and cancer. We identify a deleterious UBQLN4 mutation in families with an autosomal recessive syndrome reminiscent of genome instability disorders. UBQLN4 deficiency leads to increased sensitivity to genotoxic stress and delayed DNA double-strand break (DSB) repair. The proteasomal shuttle factor UBQLN4 is phosphorylated by ATM and interacts with ubiquitylated MRE11 to mediate early steps of homologous recombination-mediated DSB repair (HRR). Loss of UBQLN4 leads to chromatin retention of MRE11, promoting non-physiological HRR activity in vitro and in vivo. Conversely, UBQLN4 overexpression represses HRR and favors non-homologous end joining. Moreover, we find UBQLN4 overexpressed in aggressive tumors. In line with an HRR defect in these tumors, UBQLN4 overexpression is associated with PARP1 inhibitor sensitivity. UBQLN4 therefore curtails HRR activity through removal of MRE11 from damaged chromatin and thus offers a therapeutic window for PARP1 inhibitor treatment in UBQLN4-overexpressing tumors.


Subject(s)
Carrier Proteins/genetics , Nuclear Proteins/genetics , Carrier Proteins/metabolism , Chromatin/metabolism , DNA , DNA Breaks, Double-Stranded , DNA Damage/genetics , DNA End-Joining Repair , DNA-Binding Proteins/metabolism , Female , Genomic Instability , Germ-Line Mutation , Homologous Recombination , Humans , MRE11 Homologue Protein/genetics , MRE11 Homologue Protein/metabolism , Male , Neoplasms/genetics , Neoplasms/metabolism , Nuclear Proteins/metabolism , Primary Cell Culture , Recombinational DNA Repair
8.
DNA Repair (Amst) ; 72: 10-17, 2018 12.
Article in English | MEDLINE | ID: mdl-30348496

ABSTRACT

The genome instability syndrome, ataxia-telangiectasia (A-T) is caused by null mutations in the ATM gene, that lead to complete loss or inactivation of the gene's product, the ATM protein kinase. ATM is the primary mobilizer of the cellular response to DNA double-strand breaks (DSBs) - a broad signaling network in which many components are ATM targets. The major clinical feature of A-T is cerebellar atrophy, characterized by relentless loss of Purkinje and granule cells. In Atm-knockout (Atm-KO) mice, complete loss of Atm leads to a very mild neurological phenotype, suggesting that Atm loss is not sufficient to markedly abrogate cerebellar structure and function in this organism. Expression of inactive ("kinase-dead") Atm (AtmKD) in mice leads to embryonic lethality, raising the question of whether conditional expression of AtmKD in the murine nervous system would lead to a more pronounced neurological phenotype than Atm loss. We generated two mouse strains in which AtmKD was conditionally expressed as the sole Atm species: one in the CNS and one specifically in Purkinje cells. Focusing our analysis on Purkinje cells, the dynamics of DSB readouts indicated that DSB repair was delayed longer in the presence of AtmKD compared to Atm loss. However, both strains exhibited normal life span and displayed no gross cerebellar histological abnormalities or significant neurological phenotype. We conclude that the presence of AtmKD is indeed more harmful to DSB repair than Atm loss, but the murine central nervous system can reasonably tolerate the extent of this DSB repair impairment. Greater pressure needs to be exerted on genome stability to obtain a mouse model that recapitulates the severe A-T neurological phenotype.


Subject(s)
Ataxia Telangiectasia Mutated Proteins/deficiency , Ataxia Telangiectasia Mutated Proteins/genetics , Ataxia Telangiectasia/genetics , Cerebellum/metabolism , DNA Breaks, Double-Stranded , DNA Repair/genetics , Phenotype , Animals , Ataxia Telangiectasia/pathology , Cerebellum/pathology , Gene Expression Regulation , Gene Knockout Techniques , Mice , Purkinje Cells/pathology
9.
Mol Cell ; 69(5): 866-878.e7, 2018 03 01.
Article in English | MEDLINE | ID: mdl-29499138

ABSTRACT

Double-strand breaks (DSBs) are critical DNA lesions that robustly activate the elaborate DNA damage response (DDR) network. We identified a critical player in DDR fine-tuning: the E3/E4 ubiquitin ligase UBE4A. UBE4A's recruitment to sites of DNA damage is dependent on primary E3 ligases in the DDR and promotes enhancement and sustainment of K48- and K63-linked ubiquitin chains at these sites. This step is required for timely recruitment of the RAP80 and BRCA1 proteins and proper organization of RAP80- and BRCA1-associated protein complexes at DSB sites. This pathway is essential for optimal end resection at DSBs, and its abrogation leads to upregulation of the highly mutagenic alternative end-joining repair at the expense of error-free homologous recombination repair. Our data uncover a critical regulatory level in the DSB response and underscore the importance of fine-tuning the complex DDR network for accurate and balanced execution of DSB repair.


Subject(s)
BRCA1 Protein/metabolism , Carrier Proteins/metabolism , DNA Breaks, Double-Stranded , Nuclear Proteins/metabolism , Recombinational DNA Repair/physiology , Ubiquitin-Protein Ligases/metabolism , Ubiquitination/physiology , BRCA1 Protein/genetics , Carrier Proteins/genetics , DNA-Binding Proteins , HeLa Cells , Histone Chaperones , Humans , Nuclear Proteins/genetics , Ubiquitin-Protein Ligases/genetics , Ubiquitins/genetics , Ubiquitins/metabolism
10.
Invest New Drugs ; 36(1): 1-9, 2018 02.
Article in English | MEDLINE | ID: mdl-28884410

ABSTRACT

We previously found that the novel histone deacetylase inhibitor (HDACI) butyroyloxymethyl diethylphosphate (AN-7) had greater selectivity against cutaneous T-cell lymphoma (CTCL) than SAHA. AN-7 synergizes with doxorubicin (Dox), an anthracycline antibiotic that induces DNA breaks. This study aimed to elucidate the mechanism underlying the effect of AN-7 on Dox-induced double-strand DNA breaks (DSBs) in CTCL, MyLa and Hut78 cell lines. The following markers/assays were employed: comet assay; western blot of γH2AX and p-KAP1; immunofluorescence of γH2AX nuclear foci; Western blot of repair protein; quantification of DSBs-repair through homologous recombination. DSB induction by Dox was evidenced by an increase in DSB markers, and DSBs-repair, by their subsequent decrease. The addition of AN-7 slightly increased Dox induction of DSBs in MyLa cells with no effect in Hut78 cells. AN-7 inhibited the repair of Dox-induced DSBs, with a more robust effect in Hut78. Treatment with AN-7 followed by Dox reduced the expression of DSB-repair proteins, with direct interference of AN-7 with the homologous recombination repair. AN-7 sensitizes CTCL cell lines to Dox, and when combined with Dox, sustains unrepaired DSBs by suppressing repair protein expression. Our data provide a mechanistic rationale for combining AN-7 with Dox or other DSB inducers as a therapeutic modality in CTCL.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , Butyrates/pharmacology , Doxorubicin/pharmacology , Histone Deacetylase Inhibitors/pharmacology , Organophosphorus Compounds/pharmacology , Prodrugs/pharmacology , Cell Line, Tumor , DNA Breaks, Double-Stranded , DNA Repair/drug effects , Humans , Lymphoma, T-Cell, Cutaneous/drug therapy , Skin Neoplasms/drug therapy
11.
Nucleic Acids Res ; 46(2): 730-747, 2018 01 25.
Article in English | MEDLINE | ID: mdl-29253183

ABSTRACT

The DNA damage response (DDR) is an extensive signaling network that is robustly mobilized by DNA double-strand breaks (DSBs). The primary transducer of the DSB response is the protein kinase, ataxia-telangiectasia, mutated (ATM). Here, we establish nuclear poly(A)-binding protein 1 (PABPN1) as a novel target of ATM and a crucial player in the DSB response. PABPN1 usually functions in regulation of RNA processing and stability. We establish that PABPN1 is recruited to the DDR as a critical regulator of DSB repair. A portion of PABPN1 relocalizes to DSB sites and is phosphorylated on Ser95 in an ATM-dependent manner. PABPN1 depletion sensitizes cells to DSB-inducing agents and prolongs the DSB-induced G2/M cell-cycle arrest, and DSB repair is hampered by PABPN1 depletion or elimination of its phosphorylation site. PABPN1 is required for optimal DSB repair via both nonhomologous end-joining (NHEJ) and homologous recombination repair (HRR), and specifically is essential for efficient DNA-end resection, an initial, key step in HRR. Using mass spectrometry analysis, we capture DNA damage-induced interactions of phospho-PABPN1, including well-established DDR players as well as other RNA metabolizing proteins. Our results uncover a novel ATM-dependent axis in the rapidly growing interface between RNA metabolism and the DDR.


Subject(s)
Ataxia Telangiectasia Mutated Proteins/metabolism , DNA Breaks, Double-Stranded , DNA Repair , Nuclear Proteins/metabolism , Poly(A)-Binding Protein I/metabolism , Ataxia Telangiectasia Mutated Proteins/genetics , Cell Line, Tumor , DNA/genetics , DNA/metabolism , G2 Phase Cell Cycle Checkpoints/genetics , HeLa Cells , Humans , Nuclear Proteins/genetics , Phosphorylation , Poly(A)-Binding Protein I/genetics , Protein Binding , Protein Interaction Maps , RNA Interference
12.
Nucleic Acids Res ; 43(9): 4517-30, 2015 May 19.
Article in English | MEDLINE | ID: mdl-25855810

ABSTRACT

The DNA damage response is vigorously activated by DNA double-strand breaks (DSBs). The chief mobilizer of the DSB response is the ATM protein kinase. We discovered that the COP9 signalosome (CSN) is a crucial player in the DSB response and an ATM target. CSN is a protein complex that regulates the activity of cullin ring ubiquitin ligase (CRL) complexes by removing the ubiquitin-like protein, NEDD8, from their cullin scaffold. We find that the CSN is physically recruited to DSB sites in a neddylation-dependent manner, and is required for timely repair of DSBs, affecting the balance between the two major DSB repair pathways-nonhomologous end-joining and homologous recombination repair (HRR). The CSN is essential for the processivity of deep end-resection-the initial step in HRR. Cullin 4a (CUL4A) is recruited to DSB sites in a CSN- and neddylation-dependent manner, suggesting that CSN partners with CRL4 in this pathway. Furthermore, we found that ATM-mediated phosphorylation of CSN subunit 3 on S410 is critical for proper DSB repair, and that loss of this phosphorylation site alone is sufficient to cause a DDR deficiency phenotype in the mouse. This novel branch of the DSB response thus significantly affects genome stability.


Subject(s)
DNA Breaks, Double-Stranded , DNA Repair , Multiprotein Complexes/metabolism , Peptide Hydrolases/metabolism , Animals , Ataxia Telangiectasia Mutated Proteins/metabolism , COP9 Signalosome Complex , Cell Line , Cells, Cultured , Cullin Proteins/metabolism , Humans , Mice , Nuclear Proteins/metabolism , Protein Kinases/metabolism
14.
Nat Rev Mol Cell Biol ; 14(4): 197-210, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23847781

ABSTRACT

The protein kinase ataxia-telangiectasia mutated (ATM) is best known for its role as an apical activator of the DNA damage response in the face of DNA double-strand breaks (DSBs). Following induction of DSBs, ATM mobilizes one of the most extensive signalling networks that responds to specific stimuli and modifies directly or indirectly a broad range of targets. Although most ATM research has focused on this function, evidence suggests that ATM-mediated phosphorylation has a role in the response to other types of genotoxic stress. Moreover, it has become apparent that ATM is active in other cell signalling pathways involved in maintaining cellular homeostasis.


Subject(s)
Cell Cycle Proteins/physiology , DNA Damage , DNA-Binding Proteins/physiology , Protein Serine-Threonine Kinases/physiology , Tumor Suppressor Proteins/physiology , Animals , Ataxia Telangiectasia Mutated Proteins , Cell Nucleus/enzymology , Homeostasis , Humans , Neurons/enzymology , Oxidation-Reduction , Protein Interaction Maps , Protein Processing, Post-Translational , Protein Stability , Retinal Vessels/enzymology , Signal Transduction , Tumor Suppressor Protein p53/metabolism
15.
Mech Ageing Dev ; 134(10): 496-505, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23583690

ABSTRACT

The cerebellum is exquisitely sensitive to deficiencies in the cellular response to specific DNA lesions. Genetic disorders caused by such deficiencies involve relentless, progressive cerebellar atrophy with striking loss of Purkinje and granule neurons. The reason for the extreme sensitivity of these cells to defective response to certain DNA lesions is unclear. This is particularly true for ataxia-telangiectasia (A-T) - a genomic instability syndrome whose major symptom is cerebellar atrophy. It is important to understand whether the DNA damage response in the cerebellum, particularly in Purkinje neurons, has special characteristics that stem from the unique features of these cells. Murine cerebellar organotypic cultures provide a valuable experimental system for this purpose since they retain the tissue organization for several weeks in culture and appear to provide the delicate Purkinje neurons with a physiological environment close to that in vivo. We have optimized this system and are using it to examine the Atm-mediated DNA damage response (DDR) in the cerebellum, with special emphasis on Purkinje cells. Our results to date, which indicate special chromatin organization in Purkinje cells that affects certain pathways of the DDR, demonstrate the usefulness of cerebellar organotypic cultures for addressing the above questions.


Subject(s)
Chromatin/metabolism , DNA Damage , Genomic Instability , Purkinje Cells/metabolism , Animals , Ataxia Telangiectasia Mutated Proteins/genetics , Ataxia Telangiectasia Mutated Proteins/metabolism , Chromatin/genetics , Chromatin/pathology , Chromobox Protein Homolog 5 , Humans , Mice , Mice, Knockout , Purkinje Cells/pathology , Tissue Culture Techniques
16.
Hum Mol Genet ; 22(14): 2785-94, 2013 Jul 15.
Article in English | MEDLINE | ID: mdl-23515154

ABSTRACT

Familial dysautonomia (FD) is a severe neurodegenerative genetic disorder restricted to the Ashkenazi Jewish population. The most common mutation in FD patients is a T-to-C transition at position 6 of intron 20 of the IKBKAP gene. This mutation causes aberrant skipping of exon 20 in a tissue-specific manner, leading to reduction of the IκB kinase complex-associated protein (IKAP) protein in the nervous system. We established a homozygous humanized mouse strain carrying human exon 20 and its two flanking introns; the 3' intron has the transition observed in the IKBKAP gene of FD patients. Although our FD humanized mouse does not display FD symptoms, the unique, tissue-specific splicing pattern of the IKBKAP in these mice allowed us to evaluate the effect of therapies on gene expression and exon 20 splicing. The FD mice were supplemented with phosphatidylserine (PS), a safe food supplement that increases mRNA and protein levels of IKBKAP in cell lines generated from FD patients. Here we demonstrated that PS treatment increases IKBAKP mRNA and IKAP protein levels in various tissues of FD mice without affecting exon 20 inclusion levels. We also observed that genes associated with transcription regulation and developmental processes were up-regulated in the cerebrum of PS-treated mice. Thus, PS holds promise for the treatment of FD.


Subject(s)
Carrier Proteins/genetics , Dysautonomia, Familial/metabolism , Phosphatidylserines/metabolism , Alternative Splicing , Animals , Carrier Proteins/metabolism , Cell Line , Disease Models, Animal , Dysautonomia, Familial/genetics , Exons , Female , Gene Knock-In Techniques , Humans , Intracellular Signaling Peptides and Proteins , Introns , Male , Mice , Mice, Transgenic , Transcriptional Elongation Factors
17.
Nat Rev Mol Cell Biol ; 14(4): 197-210, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23486281

ABSTRACT

The protein kinase ataxia-telangiectasia mutated (ATM) is best known for its role as an apical activator of the DNA damage response in the face of DNA double-strand breaks (DSBs). Following induction of DSBs, ATM mobilizes one of the most extensive signalling networks that responds to specific stimuli and modifies directly or indirectly a broad range of targets. Although most ATM research has focused on this function, evidence suggests that ATM-mediated phosphorylation has a role in the response to other types of genotoxic stress. Moreover, it has become apparent that ATM is active in other cell signalling pathways involved in maintaining cellular homeostasis.


Subject(s)
Cell Cycle Proteins/metabolism , DNA-Binding Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Tumor Suppressor Proteins/metabolism , Ataxia Telangiectasia Mutated Proteins , DNA Damage , DNA Repair , Homeostasis/genetics , Humans , Models, Genetic , Phosphorylation , Signal Transduction/genetics
18.
J Cell Biol ; 198(3): 273-5, 2012 Aug 06.
Article in English | MEDLINE | ID: mdl-22869592

ABSTRACT

The ataxia telangiectasia mutated (ATM) protein kinase regulates the cellular response to deoxyribonucleic acid (DNA) double-strand breaks by phosphorylating numerous players in the extensive DNA damage response network. Two papers in this issue (Daniel et al. 2012. J. Cell Biol. http://dx.doi.org/10.1083/jcb201204035; Yamamoto et al. 2012. J. Cell Biol. http://dx.doi.org/10.1083/jcb201204098) strikingly show that, in mice, the presence of a catalytically inactive version of ATM is embryonically lethal. This is surprising because mice completely lacking ATM have a much more moderate phenotype. The findings impact on basic cancer research and cancer therapeutics.


Subject(s)
Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Cycle Proteins/physiology , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/physiology , Gene Expression Regulation, Developmental , Gene Expression Regulation, Enzymologic , Mutation , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/physiology , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , Tumor Suppressor Proteins/physiology , Animals , Ataxia Telangiectasia Mutated Proteins , Female , Humans
19.
Cell Cycle ; 10(24): 4300-10, 2011 Dec 15.
Article in English | MEDLINE | ID: mdl-22134242

ABSTRACT

The DNA damage response (DDR) is a complex signaling network that leads to damage repair while modulating numerous cellular processes. DNA double-strand breaks (DSBs), a highly cytotoxic DNA lesion, activate this system most vigorously. The DSB response network is orchestrated by the ATM protein kinase, which phosphorylates key players in its various branches. Proteasome-mediated protein degradation plays an important role in the proteome dynamics following DNA damage induction. Here, we identify the nuclear proteasome activator PA28γ (REGγ; PSME3) as a novel DDR player. PA28γ depletion leads to cellular radiomimetic sensitivity and a marked delay in DSB repair. Specifically, PA28γ deficiency abrogates the balance between the two major DSB repair pathways--nonhomologous end-joining and homologous recombination repair. Furthermore, PA28γ is found to be an ATM target, being recruited to the DNA damage sites and required for rapid accumulation of proteasomes at these sites. Our data reveal a novel ATM-PA28γ-proteasome axis of the DDR that is required for timely coordination of DSB repair.


Subject(s)
Autoantigens/metabolism , Cell Cycle Proteins/metabolism , DNA Breaks, Double-Stranded , DNA Repair/physiology , DNA-Binding Proteins/metabolism , Proteasome Endopeptidase Complex/metabolism , Protein Serine-Threonine Kinases/metabolism , Signal Transduction/physiology , Tumor Suppressor Proteins/metabolism , Ataxia Telangiectasia Mutated Proteins , Cell Line , DNA Repair/genetics , Flow Cytometry , Humans , Immunoblotting , Immunoprecipitation , Proteasome Endopeptidase Complex/deficiency , RNA Interference , RNA, Small Interfering/genetics , Signal Transduction/genetics
20.
Cell Cycle ; 10(2): 308-22, 2011 Jan 15.
Article in English | MEDLINE | ID: mdl-21228624

ABSTRACT

The promyelocytic leukemia (PML) protein is the main structural component of subnuclear domains termed PML nuclear bodies (PML NBs), which are implicated in tumor suppression by regulating apoptosis, cell senescence, and DNA repair. Previously, we demonstrated that ATM kinase can regulate changes in PML NB number in response to DNA double-strand breaks (DSBs). PML NBs make extensive contacts with chromatin and ATM mediates DNA damage-dependent changes in chromatin structure in part by the phosphorylation of the KRAB-associated protein 1 (KAP1) at S824. We now demonstrate that in the absence of DNA damage, reduced KAP1 expression results in a constitutive increase in PML NB number in both human U2-OS cells and normal human diploid fibroblasts. This increase in PML NB number correlated with decreased nuclear lamina-associated heterochromatin and a 30% reduction in chromatin density as observed by electron microscopy, which is reminiscent of DNA damaged chromatin. These changes in chromatin ultrastructure also correlated with increased histone H4 acetylation, and treatment with the HDAC inhibitor TSA failed to further increase PML NB number. Although PML NB number could be restored by complementation with wild-type KAP1, both the loss of KAP1 or complementation with phospho-mutants of KAP1 inhibited the early increase in PML NB number and reduced the fold induction of PML NBs by 25-30% in response to etoposide-induced DNA DSBs. Together these data implicate KAP1-dependent changes in chromatin structure as one possible mechanism by which ATM may regulate PML NB number in response to DNA damage.


Subject(s)
Cell Nucleus Structures/ultrastructure , Chromatin/ultrastructure , Nuclear Proteins/analysis , Repressor Proteins/metabolism , Transcription Factors/analysis , Tumor Suppressor Proteins/analysis , Acetylation , Ataxia Telangiectasia Mutated Proteins , Cell Cycle Proteins/metabolism , Cell Nucleus Structures/chemistry , Cells, Cultured , DNA Damage , DNA-Binding Proteins/metabolism , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylases/chemistry , Histone Deacetylases/metabolism , Histones/metabolism , Humans , Mutation , Nuclear Proteins/chemistry , Phosphorylation , Promyelocytic Leukemia Protein , Protein Serine-Threonine Kinases/metabolism , Repressor Proteins/antagonists & inhibitors , Repressor Proteins/genetics , Transcription Factors/chemistry , Tripartite Motif-Containing Protein 28 , Tumor Suppressor Proteins/chemistry , Tumor Suppressor Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...