Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 60
Filter
1.
Annu Rev Biochem ; 89: 103-133, 2020 06 20.
Article in English | MEDLINE | ID: mdl-32176524

ABSTRACT

Cells confront DNA damage in every cell cycle. Among the most deleterious types of DNA damage are DNA double-strand breaks (DSBs), which can cause cell lethality if unrepaired or cancers if improperly repaired. In response to DNA DSBs, cells activate a complex DNA damage checkpoint (DDC) response that arrests the cell cycle, reprograms gene expression, and mobilizes DNA repair factors to prevent the inheritance of unrepaired and broken chromosomes. Here we examine the DDC, induced by DNA DSBs, in the budding yeast model system and in mammals.


Subject(s)
Cell Cycle Checkpoints/genetics , DNA End-Joining Repair , DNA/genetics , Recombinational DNA Repair , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae/genetics , Animals , Ataxia Telangiectasia Mutated Proteins/chemistry , Ataxia Telangiectasia Mutated Proteins/genetics , Ataxia Telangiectasia Mutated Proteins/metabolism , Checkpoint Kinase 1/genetics , Checkpoint Kinase 1/metabolism , Checkpoint Kinase 2/genetics , Checkpoint Kinase 2/metabolism , DNA/chemistry , DNA/metabolism , DNA Breaks, Double-Stranded , Humans , Models, Molecular , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Protein Structure, Secondary , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
2.
Nat Rev Mol Cell Biol ; 22(12): 796-814, 2021 12.
Article in English | MEDLINE | ID: mdl-34429537

ABSTRACT

The protein kinase ataxia telangiectasia mutated (ATM) is a master regulator of double-strand DNA break (DSB) signalling and stress responses. For three decades, ATM has been investigated extensively to elucidate its roles in the DNA damage response (DDR) and in the pathogenesis of ataxia telangiectasia (A-T), a human neurodegenerative disease caused by loss of ATM. Although hundreds of proteins have been identified as ATM phosphorylation targets and many important roles for this kinase have been identified, it is still unclear how ATM deficiency leads to the early-onset cerebellar degeneration that is common in all individuals with A-T. Recent studies suggest the existence of links between ATM deficiency and other cerebellum-specific neurological disorders, as well as the existence of broader similarities with more common neurodegenerative disorders. In this Review, we discuss recent structural insights into ATM regulation, and possible aetiologies of A-T phenotypes, including reactive oxygen species, mitochondrial dysfunction, alterations in transcription, R-loop metabolism and alternative splicing, defects in cellular proteostasis and metabolism, and potential pathogenic roles for hyper-poly(ADP-ribosyl)ation.


Subject(s)
Ataxia Telangiectasia Mutated Proteins/metabolism , Ataxia Telangiectasia/metabolism , Neurodegenerative Diseases/metabolism , Ataxia Telangiectasia/genetics , Ataxia Telangiectasia/pathology , Ataxia Telangiectasia Mutated Proteins/chemistry , Ataxia Telangiectasia Mutated Proteins/deficiency , DNA Repair , Homeostasis , Humans , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/pathology , Oxidation-Reduction , Phosphorylation , Poly(ADP-ribose) Polymerases/metabolism , RNA/metabolism
3.
Mol Cell ; 81(6): 1231-1245.e8, 2021 03 18.
Article in English | MEDLINE | ID: mdl-33503405

ABSTRACT

ATR checkpoint signaling is crucial for cellular responses to DNA replication impediments. Using an optogenetic platform, we show that TopBP1, the main activator of ATR, self-assembles extensively to yield micrometer-sized condensates. These opto-TopBP1 condensates are functional entities organized in tightly packed clusters of spherical nano-particles. TopBP1 condensates are reversible, occasionally fuse, and co-localize with TopBP1 partner proteins. We provide evidence that TopBP1 condensation is a molecular switch that amplifies ATR activity to phosphorylate checkpoint kinase 1 (Chk1) and slow down replication forks. Single amino acid substitutions of key residues in the intrinsically disordered ATR activation domain disrupt TopBP1 condensation and consequently ATR/Chk1 signaling. In physiologic salt concentration and pH, purified TopBP1 undergoes liquid-liquid phase separation in vitro. We propose that the actuation mechanism of ATR signaling is the assembly of TopBP1 condensates driven by highly regulated multivalent and cooperative interactions.


Subject(s)
Ataxia Telangiectasia Mutated Proteins , Carrier Proteins , Cell Nucleus , DNA-Binding Proteins , Mutation, Missense , Nuclear Proteins , Signal Transduction , Amino Acid Substitution , Animals , Ataxia Telangiectasia Mutated Proteins/chemistry , Ataxia Telangiectasia Mutated Proteins/genetics , Ataxia Telangiectasia Mutated Proteins/metabolism , Carrier Proteins/chemistry , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Nucleus/chemistry , Cell Nucleus/genetics , Cell Nucleus/metabolism , Checkpoint Kinase 1/chemistry , Checkpoint Kinase 1/genetics , Checkpoint Kinase 1/metabolism , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , HeLa Cells , Humans , Nuclear Proteins/chemistry , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Sf9 Cells , Spodoptera
4.
Mol Cell ; 66(6): 801-817, 2017 Jun 15.
Article in English | MEDLINE | ID: mdl-28622525

ABSTRACT

In vertebrate cells, the DNA damage response is controlled by three related kinases: ATM, ATR, and DNA-PK. It has been 20 years since the cloning of ATR, the last of the three to be identified. During this time, our understanding of how these kinases regulate DNA repair and associated events has grown profoundly, although major questions remain unanswered. Here, we provide a historical perspective of their discovery and discuss their established functions in sensing and responding to genotoxic stress. We also highlight what is known regarding their structural similarities and common mechanisms of regulation, as well as emerging non-canonical roles and how our knowledge of ATM, ATR, and DNA-PK is being translated to benefit human health.


Subject(s)
Ataxia Telangiectasia Mutated Proteins/metabolism , Calcium-Binding Proteins/metabolism , Cell Nucleus/enzymology , DNA Damage , DNA Repair , Animals , Antineoplastic Agents/therapeutic use , Ataxia Telangiectasia Mutated Proteins/antagonists & inhibitors , Ataxia Telangiectasia Mutated Proteins/chemistry , Ataxia Telangiectasia Mutated Proteins/history , Calcium-Binding Proteins/antagonists & inhibitors , Calcium-Binding Proteins/chemistry , Calcium-Binding Proteins/history , DNA Repair/drug effects , Enzyme Activation , History, 20th Century , History, 21st Century , Humans , Models, Molecular , Neoplasms/drug therapy , Neoplasms/enzymology , Neoplasms/genetics , Neoplasms/pathology , Phosphorylation , Protein Conformation , Protein Kinase Inhibitors/therapeutic use , Signal Transduction , Structure-Activity Relationship , Tumor Suppressor Protein p53/metabolism
5.
Proc Natl Acad Sci U S A ; 118(23)2021 06 08.
Article in English | MEDLINE | ID: mdl-34074749

ABSTRACT

Ataxia telangiectasia and Rad3 related (ATR) activation after replication stress involves a cascade of reactions, including replication protein A (RPA) complex loading onto single-stranded DNA and ATR activator loading onto chromatin. The contribution of histone modifications to ATR activation, however, is unclear. Here, we report that H3K14 trimethylation responds to replication stress by enhancing ATR activation. First, we confirmed that H3K14 monomethylation, dimethylation, and trimethylation all exist in mammalian cells, and that both SUV39H1 and SETD2 methyltransferases can catalyze H3K14 trimethylation in vivo and in vitro. Interestingly, SETD2-mediated H3K14 trimethylation markedly increases in response to replication stress induced with hydroxyurea, a replication stress inducer. Under these conditions, SETD2-mediated H3K14me3 recruited the RPA complex to chromatin via a direct interaction with RPA70. The increase in H3K14me3 levels was abolished, and RPA loading was attenuated when SETD2 was depleted or H3K14 was mutated. Rather, the cells were sensitive to replication stress such that the replication forks failed to restart, and cell-cycle progression was delayed. These findings help us understand how H3K14 trimethylation links replication stress with ATR activation.


Subject(s)
Ataxia Telangiectasia Mutated Proteins/metabolism , DNA Replication , DNA/biosynthesis , Histone-Lysine N-Methyltransferase/metabolism , Histones/metabolism , Replication Protein A/metabolism , Animals , Ataxia Telangiectasia Mutated Proteins/chemistry , Ataxia Telangiectasia Mutated Proteins/genetics , DNA/chemistry , DNA/genetics , Histone-Lysine N-Methyltransferase/chemistry , Histone-Lysine N-Methyltransferase/genetics , Histones/chemistry , Histones/genetics , Humans , Methylation , Methyltransferases/chemistry , Methyltransferases/genetics , Methyltransferases/metabolism , Replication Protein A/chemistry , Replication Protein A/genetics , Repressor Proteins/chemistry , Repressor Proteins/genetics , Repressor Proteins/metabolism
6.
Mol Cell ; 60(1): 35-46, 2015 Oct 01.
Article in English | MEDLINE | ID: mdl-26387736

ABSTRACT

ATR, a PI3K-like protein kinase, plays a key role in regulating DNA damage responses. Its nuclear checkpoint kinase function is well documented, but little is known about its function outside the nucleus. Here we report that ATR has an antiapoptotic activity at mitochondria in response to UV damage, and this activity is independent of its hallmark checkpoint/kinase activity and partner ATRIP. ATR contains a BH3-like domain that allows ATR-tBid interaction at mitochondria, suppressing cytochrome c release and apoptosis. This mitochondrial activity of ATR is downregulated by Pin1 that isomerizes ATR from cis-isomer to trans-isomer at the phosphorylated Ser428-Pro429 motif. However, UV inactivates Pin1 via DAPK1, stabilizing the pro-survival cis-isomeric ATR. In contrast, nuclear ATR remains in the trans-isoform disregarding UV. This cytoplasmic response of ATR may provide a mechanism for the observed antiapoptotic role of ATR in suppressing carcinogenesis and its inhibition in sensitizing anticancer agents for killing of cancer cells.


Subject(s)
BH3 Interacting Domain Death Agonist Protein/metabolism , Mitochondria/radiation effects , Peptidylprolyl Isomerase/metabolism , Apoptosis , Ataxia Telangiectasia Mutated Proteins/chemistry , Ataxia Telangiectasia Mutated Proteins/metabolism , Binding Sites , Cell Line, Tumor , Cytochromes c/metabolism , DNA Damage , Gene Expression Regulation , HCT116 Cells , HEK293 Cells , Humans , Mitochondria/genetics , NIMA-Interacting Peptidylprolyl Isomerase , Protein Conformation , bcl-2-Associated X Protein/metabolism
7.
J Biol Chem ; 296: 100112, 2021.
Article in English | MEDLINE | ID: mdl-33434574

ABSTRACT

Proper repair of damaged DNA is critical for the maintenance of genome stability. A complex composed of Integrator subunit 3 (Ints3), single-stranded DNA-binding protein 1 (SSB1), and SSB-interacting protein 1 (SSBIP1) is required for efficient homologous recombination-dependent repair of double-strand breaks (DSBs) and ataxia-telangiectasia mutated (ATM)-dependent signaling pathways. It is known that in this complex the Ints3 N-terminal domain scaffolds SSB1 and SSBIP1. However, the molecular basis for the function of the Ints3 C-terminal domain remains unclear. Here, we present the crystal structure of the Ints3 C-terminal domain, uncovering a HEAT-repeat superhelical fold. Using structure and mutation analysis, we show that the C-terminal domain exists as a stable dimer. A basic groove and a cluster of conserved residues on two opposite sides of the dimer bind single-stranded RNA/DNA (ssRNA/ssDNA) and Integrator complex subunit 6 (Ints6), respectively. Dimerization is required for nucleic acid binding, but not for Ints6 binding. Additionally, in vitro experiments using HEK 293T cells demonstrate that Ints6 interaction is critical for maintaining SSB1 protein level. Taken together, our findings establish the structural basis of a multifunctional Ints3 C-terminal module, allowing us to propose a novel mode of nucleic acid recognition by helical repeat protein and paving the way for future mechanistic studies.


Subject(s)
Ataxia Telangiectasia Mutated Proteins/chemistry , Ataxia Telangiectasia Mutated Proteins/metabolism , RNA-Binding Proteins/chemistry , RNA-Binding Proteins/metabolism , Tumor Suppressor Proteins/chemistry , Tumor Suppressor Proteins/metabolism , DNA Breaks, Double-Stranded , HEK293 Cells , Humans , Protein Binding , Protein Multimerization , Protein Structure, Secondary , Protein Structure, Tertiary , Proteolysis
8.
Mol Cell ; 54(6): 999-1011, 2014 Jun 19.
Article in English | MEDLINE | ID: mdl-24950377

ABSTRACT

The melanocortin 1 receptor (MC1R), which signals through cAMP, is a melanocytic transmembrane receptor involved in pigmentation, adaptive tanning, and melanoma resistance. We report MC1R-mediated or pharmacologically-induced cAMP signaling promotes nucleotide excision repair (NER) in a cAMP-dependent protein kinase A (PKA)-dependent manner. PKA directly phosphorylates ataxia telangiectasia and Rad3-related protein (ATR) at Ser435, which actively recruits the key NER protein xeroderma pigmentosum complementation group A (XPA) to sites of nuclear UV photodamage, accelerating clearance of UV-induced photolesions and reducing mutagenesis. Loss of Ser435 within ATR prevents PKA-mediated ATR phosphorylation, disrupts ATR-XPA binding, delays recruitment of XPA to UV-damaged DNA, and elevates UV-induced mutagenesis. This study mechanistically links cAMP-PKA signaling to NER and illustrates potential benefits of cAMP pharmacological rescue to reduce UV mutagenesis in MC1R-defective, melanoma-susceptible individuals.


Subject(s)
DNA Damage , Receptor, Melanocortin, Type 1/genetics , Xeroderma Pigmentosum Group A Protein/metabolism , Animals , Ataxia Telangiectasia Mutated Proteins/chemistry , Ataxia Telangiectasia Mutated Proteins/genetics , Cell Line, Tumor , Cyclic AMP-Dependent Protein Kinases/genetics , DNA Repair/genetics , DNA-Binding Proteins/genetics , HEK293 Cells , Humans , MCF-7 Cells , Mice , Mice, Inbred C57BL , Mutagenesis/radiation effects , Phosphorylation/radiation effects , Pigmentation/genetics , Protein Processing, Post-Translational/genetics , Protein Processing, Post-Translational/radiation effects , RNA Interference , RNA, Small Interfering , Signal Transduction/genetics , Signal Transduction/radiation effects , Ultraviolet Rays , Xeroderma Pigmentosum Group A Protein/genetics
9.
Biochem J ; 478(1): 135-156, 2021 01 15.
Article in English | MEDLINE | ID: mdl-33439989

ABSTRACT

Genomic integrity is most threatened by double-strand breaks, which, if left unrepaired, lead to carcinogenesis or cell death. The cell generates a network of protein-protein signaling interactions that emanate from the DNA damage which are now recognized as a rich basis for anti-cancer therapy development. Deciphering the structures of signaling proteins has been an uphill task owing to their large size and complex domain organization. Recent advances in mammalian protein expression/purification and cryo-EM-based structure determination have led to significant progress in our understanding of these large multidomain proteins. This review is an overview of the structural principles that underlie some of the key signaling proteins that function at the double-strand break site. We also discuss some plausible ideas that could be considered for future structural approaches to visualize and build a more complete understanding of protein dynamics at the break site.


Subject(s)
DNA Breaks, Double-Stranded , DNA Repair/genetics , Signal Transduction/genetics , Acid Anhydride Hydrolases/chemistry , Acid Anhydride Hydrolases/metabolism , Animals , Ataxia Telangiectasia Mutated Proteins/chemistry , Ataxia Telangiectasia Mutated Proteins/metabolism , Cell Cycle Proteins/chemistry , Cell Cycle Proteins/metabolism , DNA Damage/genetics , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/metabolism , Humans , MRE11 Homologue Protein/chemistry , MRE11 Homologue Protein/metabolism , Nuclear Proteins/chemistry , Nuclear Proteins/metabolism , Protein Processing, Post-Translational/genetics , Tumor Suppressor Proteins/chemistry , Tumor Suppressor Proteins/metabolism , Tumor Suppressor p53-Binding Protein 1/chemistry , Tumor Suppressor p53-Binding Protein 1/metabolism , Ubiquitin-Protein Ligases/chemistry , Ubiquitin-Protein Ligases/metabolism
10.
Int J Mol Sci ; 23(1)2022 Jan 04.
Article in English | MEDLINE | ID: mdl-35008949

ABSTRACT

Ataxia-telangiectasia mutated (ATM) functions as a key initiator and coordinator of DNA damage and cellular stress responses. ATM signaling pathways contain many downstream targets that regulate multiple important cellular processes, including DNA damage repair, apoptosis, cell cycle arrest, oxidative sensing, and proliferation. Over the past few decades, associations between germline ATM pathogenic variants and cancer risk have been reported, particularly for breast and pancreatic cancers. In addition, given that ATM plays a critical role in repairing double-strand breaks, inhibiting other DNA repair pathways could be a synthetic lethal approach. Based on this rationale, several DNA damage response inhibitors are currently being tested in ATM-deficient cancers. In this review, we discuss the current knowledge related to the structure of the ATM gene, function of ATM kinase, clinical significance of ATM germline pathogenic variants in patients with hereditary cancers, and ongoing efforts to target ATM for the benefit of cancer patients.


Subject(s)
Ataxia Telangiectasia Mutated Proteins/genetics , Ataxia Telangiectasia Mutated Proteins/metabolism , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Genetic Predisposition to Disease , Neoplasms/etiology , Neoplasms/metabolism , Animals , Apoptosis/genetics , Ataxia Telangiectasia Mutated Proteins/chemistry , Cell Cycle/genetics , DNA Breaks, Double-Stranded , DNA Damage , DNA Repair , Disease Management , Gene Expression Regulation , Germ-Line Mutation , Humans , Neoplasms/diagnosis , Neoplasms/therapy , Oxidation-Reduction , Oxidative Stress , Precision Medicine , Signal Transduction
11.
Cell Mol Life Sci ; 77(1): 3-18, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31748913

ABSTRACT

Homologous recombination (HR) is a pathway to faithfully repair DNA double-strand breaks (DSBs). At the core of this pathway is a DNA recombinase, which, as a nucleoprotein filament on ssDNA, pairs with homologous DNA as a template to repair the damaged site. In eukaryotes Rad51 is the recombinase capable of carrying out essential steps including strand invasion, homology search on the sister chromatid and strand exchange. Importantly, a tightly regulated process involving many protein factors has evolved to ensure proper localisation of this DNA repair machinery and its correct timing within the cell cycle. Dysregulation of any of the proteins involved can result in unchecked DNA damage, leading to uncontrolled cell division and cancer. Indeed, many are tumour suppressors and are key targets in the development of new cancer therapies. Over the past 40 years, our structural and mechanistic understanding of homologous recombination has steadily increased with notable recent advancements due to the advances in single particle cryo electron microscopy. These have resulted in higher resolution structural models of the signalling proteins ATM (ataxia telangiectasia mutated), and ATR (ataxia telangiectasia and Rad3-related protein), along with various structures of Rad51. However, structural information of the other major players involved, such as BRCA1 (breast cancer type 1 susceptibility protein) and BRCA2 (breast cancer type 2 susceptibility protein), has been limited to crystal structures of isolated domains and low-resolution electron microscopy reconstructions of the full-length proteins. Here we summarise the current structural understanding of homologous recombination, focusing on key proteins in recruitment and signalling events as well as the mediators for the Rad51 recombinase.


Subject(s)
DNA Damage , Protein Interaction Maps , Recombinational DNA Repair , Animals , Ataxia Telangiectasia Mutated Proteins/chemistry , Ataxia Telangiectasia Mutated Proteins/metabolism , BRCA1 Protein/chemistry , BRCA1 Protein/metabolism , BRCA2 Protein/chemistry , BRCA2 Protein/metabolism , DNA/chemistry , DNA/genetics , Humans , Models, Molecular , Protein Conformation , Rad51 Recombinase/chemistry , Rad51 Recombinase/metabolism
12.
J Biol Chem ; 294(17): 7098-7112, 2019 04 26.
Article in English | MEDLINE | ID: mdl-30867195

ABSTRACT

The Ser/Thr protein kinase ataxia telangiectasia mutated (ATM) plays an important role in the DNA damage response, signaling in response to redox signals, the control of metabolic processes, and mitochondrial homeostasis. ATM localizes to the nucleus and at the plasma membrane, mitochondria, peroxisomes, and other cytoplasmic vesicular structures. It has been shown that the C-terminal FATC domain of human ATM (hATMfatc) can interact with a range of membrane mimetics and may thereby act as a membrane-anchoring unit. Here, NMR structural and 15N relaxation data, NMR data using spin-labeled micelles, and MD simulations of micelle-associated hATMfatc revealed that it binds the micelle by a dynamic assembly of three helices with many residues of hATMfatc located in the headgroup region. We observed that none of the three helices penetrates the micelle deeply or makes significant tertiary contacts to the other helices. NMR-monitored interaction experiments with hATMfatc variants in which two conserved aromatic residues (Phe3049 and Trp3052) were either individually or both replaced by alanine disclosed that the double substitution does not abrogate the interaction with micelles and bicelles at the high concentrations at which these aggregates are typically used, but impairs interactions with small unilamellar vesicles, usually used at much lower lipid concentrations and considered a better mimetic for natural membranes. We conclude that the observed dynamic structure of micelle-associated hATMfatc may enable it to interact with differently composed membranes or membrane-associated interaction partners and thereby regulate ATM's kinase activity. Moreover, the FATC domain of ATM may function as a membrane-anchoring unit for other biomolecules.


Subject(s)
Ataxia Telangiectasia Mutated Proteins/chemistry , Nuclear Magnetic Resonance, Biomolecular/methods , Ataxia Telangiectasia Mutated Proteins/metabolism , Cell Membrane/metabolism , Humans , Micelles , Molecular Dynamics Simulation , Protein Binding , Protein Conformation , Protein Domains
13.
J Biol Chem ; 294(21): 8395-8402, 2019 05 24.
Article in English | MEDLINE | ID: mdl-30940728

ABSTRACT

DNA damage response Ser/Thr kinases, including ataxia telangiectasia-mutated (ATM) and Rad3-related (ATR), control cell cycle progression, DNA repair, and apoptosis. ATR is activated by ETAA1 activator of ATR kinase (ETAA1) or DNA topoisomerase II binding protein 1 (TOPBP1). Both ETAA1 and TOPBP1 contain experimentally defined ATR activation domains (AADs) that are mostly unstructured and have minimal sequence similarity. A tryptophan residue in both AADs is required for ATR activation, but the other features of these domains and the mechanism by which they activate ATR are unknown. In this study, using bioinformatic analyses, kinase assays, co-immunoprecipitation, and immunofluorescence measures of signaling, we more specifically defined the TOPBP1 and ETAA1 AADs and identified additional features of the AADs needed for ATR activation. We found that both ETAA1 and TOPBP1 contain a predicted coiled-coil motif that is required for ATR activation in vitro and in cells. Mutation of the predicted coiled coils does not alter AAD oligomerization but does impair binding of the AADs to ATR. These results suggest that TOPBP1 and ETAA1 activate ATR using similar motifs and mechanisms.


Subject(s)
Antigens, Surface , Ataxia Telangiectasia Mutated Proteins , Carrier Proteins , DNA-Binding Proteins , Nuclear Proteins , Amino Acid Motifs , Antigens, Surface/chemistry , Antigens, Surface/genetics , Antigens, Surface/metabolism , Ataxia Telangiectasia Mutated Proteins/chemistry , Ataxia Telangiectasia Mutated Proteins/genetics , Ataxia Telangiectasia Mutated Proteins/metabolism , Carrier Proteins/chemistry , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Line, Tumor , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Enzyme Activation , HEK293 Cells , Humans , Nuclear Proteins/chemistry , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Protein Domains
14.
Biochemistry ; 57(47): 6592-6603, 2018 11 27.
Article in English | MEDLINE | ID: mdl-30380295

ABSTRACT

DNA damage response (DDR) pathways form an integral part of the body's repair machinery, and ATR (ataxia-telangiectasia and Rad3-related kinase) protein is one of the key mediators in the DDR pathway that helps in maintaining genomic integrity. A growing body of evidence suggests that inhibition of ATR can help sensitize tumor cells to combinatorial treatment. However, specific ATR kinase inhibitors have largely remained elusive until now. Despite much interest in the protein for more than a decade, there has been little characterization of only the kinase domain, an essential target site for a variety of ATR inhibitors. Here, we report our findings for the bacterial expression, purification, and biological characterization of this potentially important recombinant kinase domain, which could further be considered for structure elucidation studies. Introduction of a solubility partner, i.e., maltose binding protein (MBP), at the N-terminus of the ATR kinase domain generated a soluble form of the protein, i.e., MBP-tagged hATR kinase domain (MBP-ATR-6X His), which was found to be catalytically active, as assessed by substrate p53 Ser-15 phosphorylation (EPPLSQEAFADLWKK). Our results also highlight the prospect of utilization of the overexpressed recombinant ATR kinase domain in characterization of kinase domain specific inhibitors.


Subject(s)
Ataxia Telangiectasia Mutated Proteins/metabolism , Factor Xa/metabolism , Maltose-Binding Proteins/metabolism , Recombinant Fusion Proteins/metabolism , Serine/metabolism , Tumor Suppressor Protein p53/metabolism , Ataxia Telangiectasia Mutated Proteins/chemistry , Catalysis , Humans , Phosphorylation , Serine/chemistry , Tumor Suppressor Protein p53/chemistry
15.
J Med Genet ; 54(11): 732-741, 2017 11.
Article in English | MEDLINE | ID: mdl-28779002

ABSTRACT

BACKGROUND: Breast cancer (BC) is the most common malignancy in women and has a major heritable component. The risks associated with most rare susceptibility variants are not well estimated. To better characterise the contribution of variants in ATM, CHEK2, PALB2 and XRCC2, we sequenced their coding regions in 13 087 BC cases and 5488 controls from East Anglia, UK. METHODS: Gene coding regions were enriched via PCR, sequenced, variant called and filtered for quality. ORs for BC risk were estimated separately for carriers of truncating variants and of rare missense variants, which were further subdivided by functional domain and pathogenicity as predicted by four in silico algorithms. RESULTS: Truncating variants in PALB2 (OR=4.69, 95% CI 2.27 to 9.68), ATM (OR=3.26; 95% CI 1.82 to 6.46) and CHEK2 (OR=3.11; 95% CI 2.15 to 4.69), but not XRCC2 (OR=0.94; 95% CI 0.26 to 4.19) were associated with increased BC risk. Truncating variants in ATM and CHEK2 were more strongly associated with risk of oestrogen receptor (ER)-positive than ER-negative disease, while those in PALB2 were associated with similar risks for both subtypes. There was also some evidence that missense variants in ATM, CHEK2 and PALB2 may contribute to BC risk, but larger studies are necessary to quantify the magnitude of this effect. CONCLUSIONS: Truncating variants in PALB2 are associated with a higher risk of BC than those in ATM or CHEK2. A substantial risk of BC due to truncating XRCC2 variants can be excluded.


Subject(s)
Ataxia Telangiectasia Mutated Proteins/genetics , Breast Neoplasms/genetics , Checkpoint Kinase 2/genetics , DNA-Binding Proteins/genetics , Fanconi Anemia Complementation Group N Protein/genetics , Ataxia Telangiectasia Mutated Proteins/chemistry , Checkpoint Kinase 2/chemistry , DNA-Binding Proteins/chemistry , Fanconi Anemia Complementation Group N Protein/chemistry , Female , Genetic Predisposition to Disease , Genetic Variation , Humans , Sequence Analysis, Protein
16.
Molecules ; 23(5)2018 Apr 24.
Article in English | MEDLINE | ID: mdl-29695073

ABSTRACT

Targeting DNA damage and response (DDR) pathway has become an attractive approach in cancer therapy. The key mediators involved in this pathway are ataxia telangiectasia-mutated kinase (ATM) and ataxia telangiectasia-mutated, Rad3-related kinase (ATR). These kinases induce cell cycle arrest in response to chemo- and radio-therapy and facilitate DNA repair via their major downstream targets. Targeting ATP-binding site of these kinases is currently under study. Torin2 is a second generation ATP competitive mTOR kinase inhibitor (EC50 = 250 pmol/L) with better pharmacokinetic profile. Torin2 also exhibits potent biochemical and cellular activity against ATM (EC50 = 28 nmol/L) and ATR (EC50 = 35 nmol/L) kinases. In this study, eight new Torin2 analogs were designed and synthesized through multistep synthesis. All the synthesized compounds were characterized by NMR and mass analysis. The newly synthesized analogs were evaluated for their anti-cancer activity via CellTiter-Glo® assay. Additionally, compounds 13 and 14 also showed significant inhibition for ATR and mTOR substrates, i.e., p-Chk1 Ser 317 and p70 S6K Thr 389, respectively. Compounds 13 and 14 displayed promising anti-cancer activity with HCT-116 cell lines in the preliminary study. Further, a comparative model of ATR kinase was generated using the SWISS-MODEL server and validated using PROCHECK, ProSA analysis. Synthesized compounds were docked into the ATP-binding site to understand the binding modes and for the rational design of new inhibitors.


Subject(s)
Chemistry Techniques, Synthetic , Drug Design , Molecular Docking Simulation , Naphthyridines/chemistry , Naphthyridines/pharmacology , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Amino Acid Sequence , Ataxia Telangiectasia Mutated Proteins/antagonists & inhibitors , Ataxia Telangiectasia Mutated Proteins/chemistry , Binding Sites , Cell Line , Humans , Molecular Dynamics Simulation , Molecular Structure , Naphthyridines/chemical synthesis , Protein Binding , Protein Kinase Inhibitors/chemical synthesis , Reproducibility of Results , Structure-Activity Relationship , TOR Serine-Threonine Kinases/antagonists & inhibitors , TOR Serine-Threonine Kinases/chemistry
17.
J Biol Chem ; 291(26): 13436-47, 2016 Jun 24.
Article in English | MEDLINE | ID: mdl-27129217

ABSTRACT

The phosphatidylinositol 3-kinase-related protein kinases are key regulators controlling a wide range of cellular events. The yeast Tel1 and Mec1·Ddc2 complex (ATM and ATR-ATRIP in humans) play pivotal roles in DNA replication, DNA damage signaling, and repair. Here, we present the first structural insight for dimers of Mec1·Ddc2 and Tel1 using single-particle electron microscopy. Both kinases reveal a head to head dimer with one major dimeric interface through the N-terminal HEAT (named after Huntingtin, elongation factor 3, protein phosphatase 2A, and yeast kinase TOR1) repeat. Their dimeric interface is significantly distinct from the interface of mTOR complex 1 dimer, which oligomerizes through two spatially separate interfaces. We also observe different structural organizations of kinase domains of Mec1 and Tel1. The kinase domains in the Mec1·Ddc2 dimer are located in close proximity to each other. However, in the Tel1 dimer they are fully separated, providing potential access of substrates to this kinase, even in its dimeric form.


Subject(s)
Ataxia Telangiectasia Mutated Proteins/chemistry , Protein Multimerization , Ataxia Telangiectasia Mutated Proteins/genetics , Humans , Protein Domains , Protein Structure, Quaternary , Structural Homology, Protein
18.
J Proteome Res ; 12(10): 4302-15, 2013 Oct 04.
Article in English | MEDLINE | ID: mdl-23964713

ABSTRACT

Reactive oxygen species (ROS) play an important role in normal biological functions and pathological processes. ROS is one of the driving forces for oxidizing proteins, especially on cysteine thiols. The labile, transient, and dynamic nature of oxidative modifications poses enormous technical challenges for both accurate modification site determination and quantitation of cysteine thiols. The present study describes a mass spectrometry-based approach that allows effective discovery and quantification of irreversible cysteine modifications. The utilization of a long reverse phase column provides high-resolution chromatography to separate different forms of modified cysteine thiols from protein complexes or cell lysates. This Fourier transform mass spectrometry (FT-MS) approach enabled detection and quantitation of ataxia telangiectasia mutated (ATM) complex cysteine sulfoxidation states using Skyline MS1 filtering. When we applied the long column ultra high pressure liquid chromatography (UPLC)-MS/MS analysis, 61 and 44 peptides from cell lysates and cells were identified with cysteine modifications in response to in vitro and in vivo H2O2 oxidation, respectively. Long column ultra high pressure liquid chromatography pseudo selected reaction monitoring (UPLC-pSRM) was then developed to monitor the oxidative level of cysteine thiols in cell lysate under varying concentrations of H2O2 treatment. From UPLC-pSRM analysis, the dynamic conversion of sulfinic (S-O2H) and sulfonic acid (S-O3H) was observed within nucleoside diphosphate kinase (Nm23-H1) and heat shock 70 kDa protein 8 (Hsc70). These methods are suitable for proteome-wide studies, providing a highly sensitive, straightforward approach to identify proteins containing redox-sensitive cysteine thiols in biological systems.


Subject(s)
Cysteine/metabolism , Proteome/metabolism , Amino Acid Sequence , Ataxia Telangiectasia Mutated Proteins/chemistry , Ataxia Telangiectasia Mutated Proteins/isolation & purification , Ataxia Telangiectasia Mutated Proteins/metabolism , Chromatography, Affinity , Chromatography, High Pressure Liquid , HEK293 Cells , Humans , Hydrogen Peroxide/metabolism , Hydrogen Peroxide/pharmacology , Molecular Sequence Data , Oxidants/metabolism , Oxidants/pharmacology , Oxidation-Reduction , Protein-Arginine N-Methyltransferases/chemistry , Protein-Arginine N-Methyltransferases/isolation & purification , Protein-Arginine N-Methyltransferases/metabolism , Proteome/chemistry , Proteome/isolation & purification , Reference Standards , Sulfoxides/isolation & purification , Sulfoxides/metabolism , Tandem Mass Spectrometry/standards
19.
Elife ; 112022 01 25.
Article in English | MEDLINE | ID: mdl-35076389

ABSTRACT

DNA double-strand breaks (DSBs) can lead to mutations, chromosomal rearrangements, genome instability, and cancer. Central to the sensing of DSBs is the ATM (Ataxia-telangiectasia mutated) kinase, which belongs to the phosphatidylinositol 3-kinase-related protein kinase (PIKK) family. In response to DSBs, ATM is activated by the MRN (Mre11-Rad50-Nbs1) protein complex through a poorly understood process that also requires double-stranded DNA. Previous studies indicate that the FxF/Y motif of Nbs1 directly binds to ATM, and is required to retain active ATM at sites of DNA damage. Here, we report the 2.5 Å resolution cryo-EM structures of human ATM and its complex with the Nbs1 FxF/Y motif. In keeping with previous structures of ATM and its yeast homolog Tel1, the dimeric human ATM kinase adopts a symmetric, butterfly-shaped structure. The conformation of the ATM kinase domain is most similar to the inactive states of other PIKKs, suggesting that activation may involve an analogous realigning of the N and C lobes along with relieving the blockage of the substrate-binding site. We also show that the Nbs1 FxF/Y motif binds to a conserved hydrophobic cleft within the Spiral domain of ATM, suggesting an allosteric mechanism of activation. We evaluate the importance of these structural findings with mutagenesis and biochemical assays.


Subject(s)
Ataxia Telangiectasia Mutated Proteins/chemistry , Ataxia Telangiectasia Mutated Proteins/genetics , Cell Cycle Proteins/metabolism , DNA Breaks, Double-Stranded , Nuclear Proteins/metabolism , HEK293 Cells , Humans , Mutation/genetics , Phosphatidylinositol 3-Kinase/metabolism , Phosphorylation , Protein Binding
20.
Genes (Basel) ; 12(6)2021 05 30.
Article in English | MEDLINE | ID: mdl-34070860

ABSTRACT

ATM is among of the most critical initiators and coordinators of the DNA-damage response. ATM canonical and non-canonical signaling pathways involve hundreds of downstream targets that control many important cellular processes such as DNA damage repair, apoptosis, cell cycle arrest, metabolism, proliferation, oxidative sensing, among others. Of note, ATM is often considered a major tumor suppressor because of its ability to induce apoptosis and cell cycle arrest. However, in some advanced stage tumor cells, ATM signaling is increased and confers remarkable advantages for cancer cell survival, resistance to radiation and chemotherapy, biosynthesis, proliferation, and metastasis. This review focuses on addressing major characteristics, signaling pathways and especially the diverse roles of ATM in cellular homeostasis and cancer development.


Subject(s)
Ataxia Telangiectasia Mutated Proteins/genetics , Carcinogenesis/genetics , Animals , Ataxia Telangiectasia Mutated Proteins/chemistry , Ataxia Telangiectasia Mutated Proteins/metabolism , Carcinogenesis/metabolism , DNA Repair , Humans , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL