Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 77
Filter
Add more filters










Publication year range
1.
J Biol Chem ; 300(5): 107265, 2024 May.
Article in English | MEDLINE | ID: mdl-38582452

ABSTRACT

Histidine kinases are key bacterial sensors that recognize diverse environmental stimuli. While mechanisms of phosphorylation and phosphotransfer by cytoplasmic kinase domains are relatively well-characterized, the ways in which extracytoplasmic sensor domains regulate activation remain mysterious. The Cpx envelope stress response is a conserved Gram-negative two-component system which is controlled by the sensor kinase CpxA. We report the structure of the Escherichia coli CpxA sensor domain (CpxA-SD) as a globular Per-ARNT-Sim (PAS)-like fold highly similar to that of Vibrio parahaemolyticus CpxA as determined by X-ray crystallography. Because sensor kinase dimerization is important for signaling, we used AlphaFold2 to model CpxA-SD in the context of its connected transmembrane domains, which yielded a novel dimer of PAS domains possessing a distinct dimer organization compared to previously characterized sensor domains. Gain of function cpxA∗ alleles map to the dimer interface, and mutation of other residues in this region also leads to constitutive activation. CpxA activation can be suppressed by mutations that restore inter-monomer interactions, suggesting that inhibitory interactions between CpxA-SD monomers are the major point of control for CpxA activation and signaling. Searching through hundreds of structural homologs revealed the sensor domain of Pseudomonas aeruginosa sensor kinase PfeS as the only PAS structure in the same novel dimer orientation as CpxA, suggesting that our dimer orientation may be utilized by other extracytoplasmic PAS domains. Overall, our findings provide insight into the diversity of the organization of PAS sensory domains and how they regulate sensor kinase activation.


Subject(s)
Escherichia coli Proteins , Escherichia coli , Histidine Kinase , Protein Domains , Protein Multimerization , Bacterial Proteins/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Crystallography, X-Ray , Escherichia coli/enzymology , Escherichia coli/genetics , Escherichia coli Proteins/metabolism , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/genetics , Histidine Kinase/metabolism , Histidine Kinase/chemistry , Histidine Kinase/genetics , Models, Molecular , Signal Transduction , Vibrio parahaemolyticus/enzymology , Vibrio parahaemolyticus/genetics
2.
J Biol Chem ; 300(3): 105709, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38309501

ABSTRACT

Double-strand breaks (DSBs) are DNA lesions that pose a significant threat to genomic stability. The repair of DSBs by the homologous recombination (HR) pathway is preceded by DNA end resection, the 5' to 3' nucleolytic degradation of DNA away from the DSB. We and others previously identified a role for RNF138, a really interesting new gene finger E3 ubiquitin ligase, in stimulating DNA end resection and HR. Yet, little is known about how RNF138's function is regulated in the context of DSB repair. Here, we show that RNF138 is phosphorylated at residue T27 by cyclin-dependent kinase (CDK) activity during the S and G2 phases of the cell cycle. We also observe that RNF138 is ubiquitylated constitutively, with ubiquitylation occurring in part on residue K158 and rising during the S/G2 phases. Interestingly, RNF138 ubiquitylation decreases upon genotoxic stress. By mutating RNF138 at residues T27, K158, and the previously identified S124 ataxia telangiectasia mutated phosphorylation site (Han et al., 2016, ref. 22), we find that post-translational modifications at all three positions mediate DSB repair. Cells expressing the T27A, K158R, and S124A variants of RNF138 are impaired in DNA end resection, HR activity, and are more sensitive to ionizing radiation compared to those expressing wildtype RNF138. Our findings shed more light on how RNF138 activity is controlled by the cell during HR.


Subject(s)
DNA Breaks, Double-Stranded , DNA End-Joining Repair , Ubiquitin-Protein Ligases , Homologous Recombination , Phosphorylation , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Ubiquitination , Humans , HEK293 Cells
3.
J Biol Chem ; 299(11): 105341, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37832873

ABSTRACT

The emergence of severe acute respiratory syndrome coronavirus 2, the causative agent of coronavirus disease 2019, has resulted in the largest pandemic in recent history. Current therapeutic strategies to mitigate this disease have focused on the development of vaccines and on drugs that inhibit the viral 3CL protease or RNA-dependent RNA polymerase enzymes. A less-explored and potentially complementary drug target is Nsp15, a uracil-specific RNA endonuclease that shields coronaviruses and other nidoviruses from mammalian innate immune defenses. Here, we perform a high-throughput screen of over 100,000 small molecules to identify Nsp15 inhibitors. We characterize the potency, mechanism, selectivity, and predicted binding mode of five lead compounds. We show that one of these, IPA-3, is an irreversible inhibitor that might act via covalent modification of Cys residues within Nsp15. Moreover, we demonstrate that three of these inhibitors (hexachlorophene, IPA-3, and CID5675221) block severe acute respiratory syndrome coronavirus 2 replication in cells at subtoxic doses. This study provides a pipeline for the identification of Nsp15 inhibitors and pinpoints lead compounds for further development against coronavirus disease 2019 and related coronavirus infections.


Subject(s)
Antiviral Agents , Endoribonucleases , SARS-CoV-2 , Viral Nonstructural Proteins , Antiviral Agents/pharmacology , Endoribonucleases/antagonists & inhibitors , SARS-CoV-2/drug effects , SARS-CoV-2/enzymology , Viral Nonstructural Proteins/antagonists & inhibitors , Virus Replication/drug effects
4.
Acta Crystallogr D Struct Biol ; 79(Pt 10): 953-955, 2023 Oct 01.
Article in English | MEDLINE | ID: mdl-37712437

ABSTRACT

Michael James is remembered.

5.
Biochim Biophys Acta Biomembr ; 1865(1): 184069, 2023 01 01.
Article in English | MEDLINE | ID: mdl-36216097

ABSTRACT

With the advent of modern detectors and robust structure solution pipeline, cryogenic electron microscopy has recently proved to be game changer in structural biology. Membrane proteins are challenging targets for structural biologists. This minireview focuses a membrane embedded triglyceride synthesizing machine, DGAT1. Decades of research had built the foundational knowledge on this enzyme's activity. However, recently solved cryo-EM structures of this enzyme, in apo and bound form, has provided critical mechanistic insights. The flipping of the catalytic histidine is critical of enzyme catalysis. The structures explain why the enzyme has preference to long fatty acyl chains over the short forms.


Subject(s)
Diacylglycerol O-Acyltransferase , Histidine , Diacylglycerol O-Acyltransferase/chemistry , Diacylglycerol O-Acyltransferase/metabolism , Triglycerides/metabolism
6.
Diabetes ; 72(1): 126-134, 2023 01 01.
Article in English | MEDLINE | ID: mdl-36256885

ABSTRACT

Despite significant progress in understanding the pathogenesis of type 2 diabetes (T2D), the condition remains difficult to manage. Hence, new therapeutic options targeting unique mechanisms of action are required. We have previously observed that elevated skeletal muscle succinyl CoA:3-ketoacid CoA transferase (SCOT) activity, the rate-limiting enzyme of ketone oxidation, contributes to the hyperglycemia characterizing obesity and T2D. Moreover, we identified that the typical antipsychotic agent pimozide is a SCOT inhibitor that can alleviate obesity-induced hyperglycemia. We now extend those observations here, using computer-assisted in silico modeling and in vivo pharmacology studies that highlight SCOT as a noncanonical target shared among the diphenylbutylpiperidine (DPBP) drug class, which includes penfluridol and fluspirilene. All three DPBPs tested (pimozide, penfluridol, and fluspirilene) improved glycemia in obese mice. While the canonical target of the DPBPs is the dopamine 2 receptor, studies in obese mice demonstrated that acute or chronic treatment with a structurally unrelated antipsychotic dopamine 2 receptor antagonist, lurasidone, was devoid of glucose-lowering actions. We further observed that the DPBPs improved glycemia in a SCOT-dependent manner in skeletal muscle, suggesting that this older class of antipsychotic agents may have utility in being repurposed for the treatment of T2D.


Subject(s)
Antipsychotic Agents , Diabetes Mellitus, Type 2 , Hyperglycemia , Animals , Mice , Antipsychotic Agents/pharmacology , Antipsychotic Agents/therapeutic use , Coenzyme A-Transferases , Diabetes Mellitus, Type 2/drug therapy , Dopamine , Fluspirilene/pharmacology , Hyperglycemia/drug therapy , Mice, Obese , Penfluridol/pharmacology , Pimozide/pharmacology , Receptors, Dopamine/metabolism
7.
Nat Commun ; 13(1): 7076, 2022 11 18.
Article in English | MEDLINE | ID: mdl-36400772

ABSTRACT

The ProQ/FinO family of RNA binding proteins mediate sRNA-directed gene regulation throughout gram-negative bacteria. Here, we investigate the structural basis for RNA recognition by ProQ/FinO proteins, through the crystal structure of the ProQ/FinO domain of the Legionella pneumophila DNA uptake regulator, RocC, bound to the transcriptional terminator of its primary partner, the sRNA RocR. The structure reveals specific recognition of the 3' nucleotide of the terminator by a conserved pocket involving a ß-turn-α-helix motif, while the hairpin portion of the terminator is recognized by a conserved α-helical N-cap motif. Structure-guided mutagenesis reveals key RNA contact residues that are critical for RocC/RocR to repress the uptake of environmental DNA in L. pneumophila. Structural analysis and RNA binding studies reveal that other ProQ/FinO domains also recognize related transcriptional terminators with different specificities for the length of the 3' ssRNA tail.


Subject(s)
RNA, Small Untranslated , RNA-Binding Proteins , RNA-Binding Proteins/metabolism , RNA, Small Untranslated/genetics
8.
Front Immunol ; 13: 906687, 2022.
Article in English | MEDLINE | ID: mdl-35784352

ABSTRACT

Dexamethasone may reduce mortality in COVID-19 patients. Whether dexamethasone or endogenous glucocorticoids, such as cortisol, biochemically interact with SARS-CoV-2 spike 1 protein (S1), or its cellular receptor ACE2, is unknown. Using molecular dynamics (MD) simulations and binding energy calculations, we identified 162 druggable pockets in various conformational states of S1 and all possible binding pockets for cortisol and dexamethasone. Through biochemical binding studies, we confirmed that cortisol and dexamethasone bind to S1. Limited proteolysis and mass spectrometry analyses validated several MD identified binding pockets for cortisol and dexamethasone on S1. Interaction assays indicated that cortisol and dexamethasone separately and cooperatively disrupt S1 interaction with ACE2, through direct binding to S1, without affecting ACE2 catalytic activity. Cortisol disrupted the binding of the mutant S1 Beta variant (E484K, K417N, N501Y) to ACE2. Delta and Omicron variants are mutated in or near identified cortisol-binding pockets in S1, which may affect cortisol binding to them. In the presence of cortisol, we find increased inhibition of S1 binding to ACE2 by an anti-SARS-CoV-2 S1 human chimeric monoclonal antibody against the receptor binding domain. Whether glucocorticoid/S1 direct interaction is an innate defence mechanism that may have contributed to mild or asymptomatic SARS-CoV-2 infection deserves further investigation.


Subject(s)
Angiotensin-Converting Enzyme 2 , COVID-19 Drug Treatment , Antibodies, Viral , Dexamethasone/pharmacology , Glucocorticoids/pharmacology , Humans , Hydrocortisone , Peptidyl-Dipeptidase A/metabolism , SARS-CoV-2
9.
Sci Rep ; 12(1): 5386, 2022 03 30.
Article in English | MEDLINE | ID: mdl-35354845

ABSTRACT

Polynucleotide Kinase-Phosphatase (PNKP) is a bifunctional enzyme that possesses both DNA 3'-phosphatase and DNA 5'-kinase activities, which are required for processing termini of single- and double-strand breaks generated by reactive oxygen species (ROS), ionizing radiation and topoisomerase I poisons. Even though PNKP is central to DNA repair, there have been no reports linking PNKP mutations in a Microcephaly, Seizures, and Developmental Delay (MSCZ) patient to cancer. Here, we characterized the biochemical significance of 2 germ-line point mutations in the PNKP gene of a 3-year old male with MSCZ who presented with a high-grade brain tumor (glioblastoma multiforme) within the cerebellum. Functional and biochemical studies demonstrated these PNKP mutations significantly diminished DNA kinase/phosphatase activities, altered its cellular distribution, caused defective repair of DNA single/double stranded breaks, and were associated with a higher propensity for oncogenic transformation. Our findings indicate that specific PNKP mutations may contribute to tumor initiation within susceptible cells in the CNS by limiting DNA damage repair and increasing rates of spontaneous mutations resulting in pediatric glioma associated driver mutations such as ATRX and TP53.


Subject(s)
Brain Neoplasms , Microcephaly , Brain Neoplasms/genetics , Child , Child, Preschool , DNA Repair/genetics , DNA Repair Enzymes/metabolism , Humans , Male , Microcephaly/genetics , Mutation , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Seizures/genetics
10.
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
11.
Biomol NMR Assign ; 15(1): 61-64, 2021 04.
Article in English | MEDLINE | ID: mdl-33179208

ABSTRACT

In prokaryotic species, gene expression is commonly regulated by small, non-coding RNAs (sRNAs). In the gram-negative bacterium Legionella pneumophila, the regulatory, trans-acting sRNA molecule RocR base pairs with a complementary sequence in the 5'-untranslated region of mRNAs encoding for proteins in the bacterial DNA uptake system, thereby controlling natural competence. Sense-antisense duplexing of RocR with targeted mRNAs is mediated by the recently described RNA chaperone RocC. RocC contains a 12 kDa FinO-domain, which acts as sRNA binding platform, along with an extended C-terminal segment that is predicted to be mostly disordered but appears to be required for repression of bacterial competence. In this work we assigned backbone and side chain 1H, 13C, and 15N chemical shifts of RocC's FinO-domain by solution NMR spectroscopy. The chemical shift data for this protein indicate a mixed α/ß fold that is reminiscent of FinO from Escherichia coli. Our NMR resonance assignments provide the basis for a comprehensive analysis of RocC's chaperoning mechanism on a structural level.


Subject(s)
Nuclear Magnetic Resonance, Biomolecular , RNA, Small Untranslated , Escherichia coli Proteins , Legionella pneumophila
12.
Methods Mol Biol ; 2106: 1-18, 2020.
Article in English | MEDLINE | ID: mdl-31889248

ABSTRACT

The FinO family of proteins constitutes a group of RNA chaperones that interacts with small RNAs (sRNAs) to regulate gene expression in many bacterial species. Here we describe detailed protocols for the biochemical analysis of the RNA chaperone activity of these proteins. Methods are described for preparation of RNA, RNA 5' end labeling with radioisotope and modified EMSA protocols to test the ability of these proteins to catalyze RNA strand exchange and RNA duplex formation.


Subject(s)
Electrophoretic Mobility Shift Assay/methods , Escherichia coli Proteins/chemistry , Molecular Chaperones/chemistry , Molecular Probe Techniques , RNA, Small Untranslated/chemistry , RNA-Binding Proteins/chemistry , Repressor Proteins/chemistry , Escherichia coli Proteins/metabolism , Molecular Chaperones/metabolism , RNA Stability , RNA, Small Untranslated/metabolism , RNA-Binding Proteins/metabolism , Repressor Proteins/metabolism
13.
Structure ; 27(10): 1485-1496.e4, 2019 10 01.
Article in English | MEDLINE | ID: mdl-31402222

ABSTRACT

ASPP (apoptosis-stimulating proteins of p53) proteins bind PP-1c (protein phosphatase 1) and regulate p53 impacting cancer cell growth and apoptosis. Here we determine the crystal structure of the oncogenic ASPP protein, iASPP, bound to PP-1c. The structure reveals a 1:1 complex that relies on interactions of the iASPP SILK and RVxF motifs with PP-1c, plus interactions of the PP-1c PxxPxR motif with the iASPP SH3 domain. Small-angle X-ray scattering analyses suggest that the crystal structure undergoes slow interconversion with more extended conformations in solution. We show that iASPP, and the tumor suppressor ASPP2, enhance the catalytic activity of PP-1c against the small-molecule substrate, pNPP as well as p53. The combined results suggest that PxxPxR binding to iASPP SH3 domain is critical for complex formation, and that the modular ASPP-PP-1c interface provides dynamic flexibility that enables functional binding and dephosphorylation of p53 and other diverse protein substrates.


Subject(s)
Apoptosis Regulatory Proteins/chemistry , Apoptosis Regulatory Proteins/metabolism , Intracellular Signaling Peptides and Proteins/chemistry , Intracellular Signaling Peptides and Proteins/metabolism , Protein Phosphatase 1/metabolism , Repressor Proteins/chemistry , Repressor Proteins/metabolism , Amino Acid Motifs , Aniline Compounds/metabolism , Binding Sites , Biocatalysis , Crystallography, X-Ray , Humans , Models, Molecular , Organophosphorus Compounds/metabolism , Protein Binding , Protein Conformation , Protein Phosphatase 1/chemistry , Scattering, Small Angle , Tumor Suppressor Protein p53/metabolism , X-Ray Diffraction
14.
J Bacteriol ; 201(1)2019 01 01.
Article in English | MEDLINE | ID: mdl-30322855

ABSTRACT

The F plasmid tra operon encodes most of the proteins required for bacterial conjugation. TraJ and ArcA are known activators of the tra operon promoter PY, which is subject to H-NS-mediated silencing. Donor ability and promoter activity assays indicated that PY is inactivated by silencers and requires both TraJ and ArcA for activation to support efficient F conjugation. The observed low-level, ArcA-independent F conjugation is caused by tra expression from upstream alternative promoters. Electrophoretic mobility shift assays showed that TraJ alone weakly binds to PY regulatory DNA; however, TraJ binding is significantly enhanced by ArcA binding to the same DNA, indicating cooperativity of the two proteins. Analysis of binding affinities between ArcA and various DNA fragments in the PY regulatory region defined a 22-bp tandem repeat sequence (from -76 to -55 of PY) sufficient for optimal ArcA binding, which is immediately upstream of the predicted TraJ-binding site (from -54 to -34). Deletion analysis of the PY promoter in strains deficient in TraJ, ArcA, and/or H-NS determined that sequences upstream of -103 are required by silencers including H-NS for PY silencing, whereas sequences downstream of -77 are targeted by TraJ and ArcA for activation. TraJ and ArcA appear not only to counteract PY silencers but also to directly activate PY in a cooperative manner. Our data reveal the cooperativity of TraJ and ArcA during PY activation and provide insights into the regulatory circuit controlling F-family plasmid-mediated bacterial conjugation.IMPORTANCE Conjugation is a major mechanism for dissemination of antibiotic resistance and virulence among bacterial populations. The tra operon in the F family of conjugative plasmids encodes most of the proteins involved in bacterial conjugation. This work reveals that activation of tra operon transcription requires two proteins, TraJ and ArcA, to bind cooperatively to adjacent sites immediately upstream of the major tra promoter PY The interaction of TraJ and ArcA with the tra operon not only relieves PY from silencers but also directly activates it. These findings provide insights into the regulatory circuit of the F-family plasmid-mediated bacterial conjugation.


Subject(s)
Bacterial Outer Membrane Proteins/metabolism , Conjugation, Genetic , Escherichia coli Proteins/metabolism , Escherichia coli/genetics , F Factor , Gene Expression Regulation, Bacterial , Operon , Repressor Proteins/metabolism , Bacterial Outer Membrane Proteins/genetics , DNA, Bacterial/metabolism , Escherichia coli/enzymology , Escherichia coli Proteins/genetics , Gene Deletion , Promoter Regions, Genetic , Protein Binding , Protein Interaction Mapping , Repressor Proteins/genetics
15.
J Biol Chem ; 294(2): 520-530, 2019 01 11.
Article in English | MEDLINE | ID: mdl-30446622

ABSTRACT

Polynucleotide kinase/phosphatase (PNKP) and X-ray repair cross-complementing 1 (XRCC1) are key proteins in the single-strand DNA break repair pathway. Phosphorylated XRCC1 stimulates PNKP by binding to its forkhead-associated (FHA) domain, whereas nonphosphorylated XRCC1 stimulates PNKP by interacting with the PNKP catalytic domain. Here, we have further studied the interactions between these two proteins, including two variants of XRCC1 (R194W and R280H) arising from single-nucleotide polymorphisms (SNPs) that have been associated with elevated cancer risk in some reports. We observed that interaction of the PNKP FHA domain with phosphorylated XRCC1 extends beyond the immediate, well-characterized phosphorylated region of XRCC1 (residues 515-526). We also found that an XRCC1 fragment, comprising residues 166-436, binds tightly to PNKP and DNA and efficiently activates PNKP's kinase activity. However, interaction of either of the SNP-derived variants of this fragment with PNKP was considerably weaker, and their stimulation of PNKP was severely reduced, although they still could bind DNA effectively. Laser microirradiation revealed reduced recruitment of PNKP to damaged DNA in cells expressing either XRCC1 variant compared with PNKP recruitment in cells expressing WT XRCC1 even though WT and variant XRCC1s were equally efficient at localizing to the damaged DNA. These findings suggest that the elevated risk of cancer associated with these XRCC1 SNPs reported in some studies may be due in part to the reduced ability of these XRCC1 variants to recruit PNKP to damaged DNA.


Subject(s)
DNA Repair Enzymes/metabolism , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Polymorphism, Single Nucleotide , Protein Interaction Domains and Motifs , X-ray Repair Cross Complementing Protein 1/genetics , X-ray Repair Cross Complementing Protein 1/metabolism , Animals , CHO Cells , Cricetulus , DNA Damage , DNA Repair Enzymes/chemistry , Phosphotransferases (Alcohol Group Acceptor)/chemistry , Protein Interaction Maps , X-ray Repair Cross Complementing Protein 1/chemistry
16.
Structure ; 25(10): 1582-1588.e3, 2017 10 03.
Article in English | MEDLINE | ID: mdl-28919440

ABSTRACT

Topoisomerase IIß binding protein 1 (TopBP1) is a critical protein-protein interaction hub in DNA replication checkpoint control. It was proposed that TopBP1 BRCT5 interacts with Bloom syndrome helicase (BLM) to regulate genome stability through either phospho-Ser304 or phospho-Ser338 of BLM. Here we show that TopBP1 BRCT5 specifically interacts with the BLM region surrounding pSer304, not pSer338. Our crystal structure of TopBP1 BRCT4/5 bound to BLM reveals recognition of pSer304 by a conserved pSer-binding pocket, and interactions between an FVPP motif N-terminal to pSer304 and a hydrophobic groove on BRCT5. This interaction utilizes the same surface of BRCT5 that recognizes the DNA damage mediator, MDC1; however the binding orientations of MDC1 and BLM are reversed. While the MDC1 interactions are largely electrostatic, the interaction with BLM has higher affinity and relies on a mix of electrostatics and hydrophobicity. We suggest that similar evolutionarily conserved interactions may govern interactions between TopBP1 and 53BP1.


Subject(s)
DNA-Binding Proteins/chemistry , DNA-Binding Proteins/metabolism , RecQ Helicases/chemistry , RecQ Helicases/metabolism , Animals , Binding Sites , Carrier Proteins/chemistry , Carrier Proteins/metabolism , Crystallography, X-Ray , Humans , Mice , Models, Molecular , Nuclear Proteins/chemistry , Nuclear Proteins/metabolism , Phosphorylation , Protein Conformation , Serine/metabolism , Trans-Activators/metabolism
17.
Nucleic Acids Res ; 45(10): 6238-6251, 2017 Jun 02.
Article in English | MEDLINE | ID: mdl-28453785

ABSTRACT

Non-homologous end joining (NHEJ) repairs DNA double strand breaks in non-cycling eukaryotic cells. NHEJ relies on polynucleotide kinase/phosphatase (PNKP), which generates 5΄-phosphate/3΄-hydroxyl DNA termini that are critical for ligation by the NHEJ DNA ligase, LigIV. PNKP and LigIV require the NHEJ scaffolding protein, XRCC4. The PNKP FHA domain binds to the CK2-phosphorylated XRCC4 C-terminal tail, while LigIV uses its tandem BRCT repeats to bind the XRCC4 coiled-coil. Yet, the assembled PNKP-XRCC4-LigIV complex remains uncharacterized. Here, we report purification and characterization of a recombinant PNKP-XRCC4-LigIV complex. We show that the stable binding of PNKP in this complex requires XRCC4 phosphorylation and that only one PNKP protomer binds per XRCC4 dimer. Small angle X-ray scattering (SAXS) reveals a flexible multi-state complex that suggests that both the PNKP FHA and catalytic domains contact the XRCC4 coiled-coil and LigIV BRCT repeats. Hydrogen-deuterium exchange indicates protection of a surface on the PNKP phosphatase domain that may contact XRCC4-LigIV. A mutation on this surface (E326K) causes the hereditary neuro-developmental disorder, MCSZ. This mutation impairs PNKP recruitment to damaged DNA in human cells and provides a possible disease mechanism. Together, this work unveils multipoint contacts between PNKP and XRCC4-LigIV that regulate PNKP recruitment and activity within NHEJ.


Subject(s)
DNA End-Joining Repair/physiology , DNA Ligase ATP/physiology , DNA Repair Enzymes/physiology , DNA-Binding Proteins/physiology , Phosphotransferases (Alcohol Group Acceptor)/physiology , Catalytic Domain , DNA Damage , DNA Ligase ATP/chemistry , DNA Repair Enzymes/chemistry , DNA Repair Enzymes/deficiency , DNA Repair Enzymes/genetics , DNA-Binding Proteins/chemistry , Deuterium/metabolism , Developmental Disabilities/genetics , Humans , Mass Spectrometry , Microcephaly/genetics , Models, Molecular , Multiprotein Complexes , Mutation, Missense , Phosphorylation , Phosphotransferases (Alcohol Group Acceptor)/chemistry , Phosphotransferases (Alcohol Group Acceptor)/deficiency , Phosphotransferases (Alcohol Group Acceptor)/genetics , Point Mutation , Protein Binding , Protein Conformation , Protein Interaction Mapping , Protein Processing, Post-Translational , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Scattering, Small Angle , Seizures/genetics , Syndrome , X-Ray Diffraction
18.
Biochemistry ; 56(12): 1737-1745, 2017 03 28.
Article in English | MEDLINE | ID: mdl-28276686

ABSTRACT

Polynucleotide kinase/phosphatase (PNKP) is a DNA strand break repair enzyme that uses separate 5' kinase and 3' phosphatase active sites to convert damaged 5'-hydroxyl/3'-phosphate strand termini to ligatable 5'-phosphate/3'-hydroxyl ends. The phosphatase active site has received particular attention as a target of inhibition in cancer therapy development. The phosphatase domain dephosphorylates a range of single- and double-stranded substrates; however, structural studies have shown that the phosphatase catalytic cleft can bind only single-stranded substrates. Here we use a catalytically inactive but structurally intact phosphatase mutant to probe interactions between PNKP and a variety of single- and double-stranded DNA substrates using an electrophoretic mobility shift assay. This work indicates that the phosphatase domain binds 3'-phosphorylated single-stranded DNAs in a manner that is highly dependent on the presence of the 3'-phosphate. Double-stranded substrate binding, in contrast, is not as dependent on the 3'-phosphate. Experiments comparing blunt-end, 3'-overhanging, and frayed-end substrates indicate that the predicted loss of energy due to base pair disruption upon binding of the phosphatase active site is likely balanced by favorable interactions between the liberated complementary strand and PNKP. Comparison of the effects on substrate binding of mutations within the phosphatase active site cleft with mutations in surrounding positively charged surfaces suggests that the surrounding surfaces are important for binding to double-stranded substrates. We further show that while fluorescence polarization methods can detect specific binding of single-stranded DNAs with the phosphatase domain, this method does not detect specific interactions between the PNKP phosphatase and double-stranded substrates.


Subject(s)
DNA Repair , DNA, Single-Stranded/chemistry , DNA/chemistry , Phosphates/chemistry , Phosphotransferases (Alcohol Group Acceptor)/chemistry , Animals , Binding Sites , Catalytic Domain , Cloning, Molecular , DNA/genetics , DNA/metabolism , DNA Damage , DNA, Single-Stranded/genetics , DNA, Single-Stranded/metabolism , Electrophoretic Mobility Shift Assay , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , Mice , Molecular Docking Simulation , Mutation , Phosphates/metabolism , Phosphotransferases (Alcohol Group Acceptor)/genetics , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Protein Binding , Protein Domains , Protein Structure, Secondary , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Structure-Activity Relationship , Substrate Specificity
19.
Trends Microbiol ; 25(4): 247-249, 2017 04.
Article in English | MEDLINE | ID: mdl-28189381

ABSTRACT

The stability and function of regulatory small RNAs (sRNAs) often require a specialized RNA-binding protein called an RNA chaperone. Recent findings show that proteins containing a ProQ/FinO domain constitute a new class of RNA chaperones that could play key roles in post-transcriptional gene regulation throughout bacterial species.


Subject(s)
Escherichia coli/genetics , Molecular Chaperones/genetics , Protein Domains/genetics , RNA, Bacterial/genetics , RNA, Small Untranslated/genetics , Escherichia coli/metabolism , Escherichia coli Proteins/genetics , RNA-Binding Proteins/genetics , Repressor Proteins/genetics , Salmonella enterica/genetics , Salmonella enterica/metabolism
20.
Mech Ageing Dev ; 161(Pt A): 130-140, 2017 01.
Article in English | MEDLINE | ID: mdl-27470939

ABSTRACT

The termini of DNA strand breaks induced by reactive oxygen species or by abortive DNA metabolic intermediates require processing to enable subsequent gap filling and ligation to proceed. The three proteins, tyrosyl DNA-phosphodiesterase 1 (TDP1), aprataxin (APTX) and polynucleotide kinase/phosphatase (PNKP) each act on a discrete set of modified strand-break termini. Recently, a series of neurodegenerative and neurodevelopmental disorders have been associated with mutations in the genes coding for these proteins. Mutations in TDP1 and APTX have been linked to Spinocerebellar ataxia with axonal neuropathy (SCAN1) and Ataxia-ocular motor apraxia 1 (AOA1), respectively, while mutations in PNKP are considered to be responsible for Microcephaly with seizures (MCSZ) and Ataxia-ocular motor apraxia 4 (AOA4). Here we present an overview of the mechanisms of these proteins and how their impairment may give rise to their respective disorders.


Subject(s)
DNA Breaks, Double-Stranded , DNA Repair Enzymes , DNA-Binding Proteins , Heredodegenerative Disorders, Nervous System , Mutation , Neurodevelopmental Disorders , Nuclear Proteins , Phosphoric Diester Hydrolases , Phosphotransferases (Alcohol Group Acceptor) , DNA Repair Enzymes/genetics , DNA Repair Enzymes/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Heredodegenerative Disorders, Nervous System/genetics , Heredodegenerative Disorders, Nervous System/metabolism , Humans , Neurodevelopmental Disorders/genetics , Neurodevelopmental Disorders/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Phosphoric Diester Hydrolases/genetics , Phosphoric Diester Hydrolases/metabolism , Phosphotransferases (Alcohol Group Acceptor)/genetics , Phosphotransferases (Alcohol Group Acceptor)/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...