42 results on '"Lamers MH"'
Search Results
2. The AMR Accelerator: from individual organizations to efficient antibiotic development partnerships.
- Author
-
Fernow J, Olliver M, Couet W, Lagrange S, Lamers MH, Olesen OF, Orrling K, Pieren M, Sloan DJ, Vaquero JJ, Miles TJ, and Karlén A
- Abstract
Competing Interests: M.P. is employed by BioVersys AG and holds stocks in the company. T.J.M. is employed by GSK and owns shares in the company.
- Published
- 2025
- Full Text
- View/download PDF
3. The RecA-NT homology motif in ImuB mediates the interaction with ImuA' which is essential for DNA damage-induced mutagenesis.
- Author
-
Santos JA, Timinskas K, Ramudzuli AA, Lamers MH, Venclovas Č, Warner DF, and Gessner SJ
- Abstract
The mycobacterial mutasome - comprising ImuA', ImuB, and DnaE2 - has been implicated in DNA damage-induced mutagenesis in Mycobacterium tuberculosis. ImuB, which is predicted to enable mutasome function via its interaction with the β clamp, is a catalytically inactive Y-family DNA polymerase. Like some other members of the Y-family, ImuB features a recently identified amino acid motif with homology to the RecA N-terminus (RecA-NT). Given the role of RecA-NT in RecA oligomerization, we hypothesized that ImuB RecA-NT mediates the interaction with ImuA', a RecA homolog of unknown function. Here, we constructed a panel of imuB alleles in which the RecA-NT was removed, or mutated. Our results indicate that RecA-NT is critical for the interaction of ImuB with ImuA'. A region downstream of RecA-NT, ImuB-C, appears to stabilize the ImuB-ImuA' interaction, but its removal does not prevent complex formation. In contrast, replacing two hydrophobic residues of RecA-NT, L378 and V383, disrupts the ImuA'-ImuB interaction. To our knowledge, this is the first experimental evidence suggesting a role for RecA-NT in mediating the interaction between a Y-family member and a RecA homolog., Competing Interests: CONFLICT OF INTEREST The authors declare that they have no competing interests., (Copyright © 2024 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
4. Recurrent CLTC::SYK fusions and CSF1R mutations in juvenile xanthogranuloma of soft tissue.
- Author
-
Kemps PG, Baelde HJ, Vorderman RHP, Stelloo E, Swennenhuis JF, Szuhai K, Lamers MH, Kenkhuis B, Al-Hussaini M, Briaire-de Bruijn IH, Lam SW, Bovée JVMG, Cleven AHG, Verdijk RM, van Noesel CJM, van Dijk MR, Scheijde-Vermeulen MA, Bruggink AH, van Laar JAM, de Vries ACH, Tissing WJE, van den Bos C, von Deimling A, van Wezel T, van Halteren AGS, and Hogendoorn PCW
- Subjects
- Humans, Male, Female, Child, Child, Preschool, Adult, Infant, Adolescent, Oncogene Proteins, Fusion genetics, Young Adult, Antigens, Differentiation, B-Lymphocyte, Histocompatibility Antigens Class II, Receptor, Macrophage Colony-Stimulating Factor, Xanthogranuloma, Juvenile genetics, Xanthogranuloma, Juvenile pathology, Xanthogranuloma, Juvenile diagnosis, Receptors, Granulocyte-Macrophage Colony-Stimulating Factor genetics, Mutation, Syk Kinase genetics, Syk Kinase metabolism
- Abstract
Abstract: Juvenile xanthogranuloma (JXG) is a histiocytic neoplasm that usually presents in the skin. Rarely, extracutaneous localizations occur; the genetic drivers of this clinical variant of JXG remain incompletely characterized. We present detailed clinicopathologic and molecular data of 16 children with extracutaneous JXG and 5 adults with xanthogranulomas confined to the central nervous system (CNS) or soft tissue. Tissue samples were obtained through the Dutch Nationwide Pathology Databank and analyzed with an innovative sequencing technique capable of detecting both small genomic variants and gene rearrangements. Targetable kinase alterations were detected in 16 of 16 children and 1 of 5 adults. Alterations included CLTC::SYK fusions in 6 children and CSF1R mutations in 7 others; all below 2 years of age with soft tissue tumors. One child had a CSF1R mutation and MRC1::PDGFRB fusion. Most were treated surgically, although spontaneous regression occurred in 1 of 6 with CLTC::SYK and 2 of 7 with CSF1R mutations, underscoring that treatment is not always necessary. Tumors with CLTC::SYK fusions generally lacked Touton giant cells but exhibited many other histologic features of JXG and concordant methylation profiles. Using multispectral immunofluorescence, phosphorylated-spleen tyrosine kinase expression was localized to CD163+ histiocytes; tumors with CLTC::SYK fusions also demonstrated mTOR activation, cyclin D1 expression, and variable phosphorylated-extracellular signal-regulated kinase expression. BRAFV600E was detected in 1 child and 1 adult with CNS-xanthogranulomas; both responded to BRAF inhibition. Finally, a TPM3::NTRK1 fusion or MAP2K1 deletion was detected in 2 children with systemic JXG who experienced spontaneous disease regression. This study advances the molecular understanding of histiocytic neoplasms and may guide diagnostics and clinical management., (© 2024 American Society of Hematology. Published by Elsevier Inc. All rights are reserved, including those for text and data mining, AI training, and similar technologies.)
- Published
- 2024
- Full Text
- View/download PDF
5. A recurrent NTRK1 tyrosine kinase domain mutation pair is characteristic in a subset of dedifferentiated liposarcomas.
- Author
-
Lippai Z, Péterfia B, Papp G, Dezső K, Bedics G, Pápai Z, Lamers MH, Kuin RC, Szuhai K, and Sápi Z
- Subjects
- Humans, Mutation, Oncogene Proteins, Fusion genetics, Receptor, trkA genetics, Liposarcoma genetics, Sarcoma genetics, Soft Tissue Neoplasms genetics
- Abstract
Introduction: Dedifferentiated liposarcoma (DDLPS) is a common form of liposarcoma with challenging treatment modalities. Pan-TRK immunopositivity can be often observed without NTRK gene fusion in soft tissue sarcomas with myogenic differentiation. Expression and the role of NTRK in DDLPS are under-studied. We sought to identify activating mutations of the NTRK genes., Materials and Methods: 131 DDLPS patients were selected for pan-TRK immunohistochemistry and positive cases were analyzed by Sanger sequencing for NTRK1, NTRK2 and NTRK3 genes. Functional assays were performed using a lentiviral transduction system to study the effect of NTRK variants in fibroblast, immortalized fibroblast, and dedifferentiated liposarcoma cell lines., Results: Out of the 131 DDLPS cases, 75 immunohistochemical staining positive cases, 46 were successfully Sanger sequenced. A recurrent somatic mutation pair in cis position (NGS) of the NTRK1 c.1810C>T (p.H604Y) and c.1838G>T (p.G613V) was identified in six cases (13%) that have never been reported in DDLPS. NTRK fusions were excluded in all six cases by FISH and NGS. The phospho-AKT immunopositivity among the six mutated cases suggested downstream activation of the NTRK signaling pathway. Functional assays showed no transforming effects, but resistance to first- and second-line TRK inhibitors of the p.G613V and p.H604Y variant., Conclusions: We detected (de novo/somatic) missense mutation variants in cis position of the NTRK1 gene in a subset of DDLPS indicating modifying mutations that may contribute to tumorigenesis in a subset of DDLPS. These variants beget resistance to TRK inhibitors indicating an interesting biomarker for other studies with TRK inhibitors., Competing Interests: Declaration of Competing Interest The authors have no conflicts of interest to declare., (Copyright © 2024 The Authors. Published by Elsevier Ltd.. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
6. A four-point molecular handover during Okazaki maturation.
- Author
-
Botto MM, Borsellini A, and Lamers MH
- Subjects
- DNA Replication, RNA metabolism, DNA Ligases genetics, DNA Ligases metabolism, DNA Ligase ATP metabolism, Endonucleases metabolism, DNA Polymerase III genetics, DNA Polymerase III metabolism, DNA chemistry
- Abstract
DNA replication introduces thousands of RNA primers into the lagging strand that need to be removed for replication to be completed. In Escherichia coli when the replicative DNA polymerase Pol IIIα terminates at a previously synthesized RNA primer, DNA Pol I takes over and continues DNA synthesis while displacing the downstream RNA primer. The displaced primer is subsequently excised by an endonuclease, followed by the sealing of the nick by a DNA ligase. Yet how the sequential actions of Pol IIIα, Pol I polymerase, Pol I endonuclease and DNA ligase are coordinated is poorly defined. Here we show that each enzymatic activity prepares the DNA substrate for the next activity, creating an efficient four-point molecular handover. The cryogenic-electron microscopy structure of Pol I bound to a DNA substrate with both an upstream and downstream primer reveals how it displaces the primer in a manner analogous to the monomeric helicases. Moreover, we find that in addition to its flap-directed nuclease activity, the endonuclease domain of Pol I also specifically cuts at the RNA-DNA junction, thus marking the end of the RNA primer and creating a 5' end that is a suitable substrate for the ligase activity of LigA once all RNA has been removed., (© 2023. The Author(s), under exclusive licence to Springer Nature America, Inc.)
- Published
- 2023
- Full Text
- View/download PDF
7. Investigating the composition and recruitment of the mycobacterial ImuA'-ImuB-DnaE2 mutasome.
- Author
-
Gessner S, Martin ZA, Reiche MA, Santos JA, Dinkele R, Ramudzuli A, Dhar N, de Wet TJ, Anoosheh S, Lang DM, Aaron J, Chew TL, Herrmann J, Müller R, McKinney JD, Woodgate R, Mizrahi V, Venclovas Č, Lamers MH, and Warner DF
- Subjects
- Mutagenesis, DNA Repair, Antitubercular Agents pharmacology, Bacterial Proteins chemistry, Mycobacterium tuberculosis genetics
- Abstract
A DNA damage-inducible mutagenic gene cassette has been implicated in the emergence of drug resistance in Mycobacterium tuberculosis during anti-tuberculosis (TB) chemotherapy. However, the molecular composition and operation of the encoded 'mycobacterial mutasome' - minimally comprising DnaE2 polymerase and ImuA' and ImuB accessory proteins - remain elusive. Following exposure of mycobacteria to DNA damaging agents, we observe that DnaE2 and ImuB co-localize with the DNA polymerase III β subunit (β clamp) in distinct intracellular foci. Notably, genetic inactivation of the mutasome in an imuB
AAAAGG mutant containing a disrupted β clamp-binding motif abolishes ImuB-β clamp focus formation, a phenotype recapitulated pharmacologically by treating bacilli with griselimycin and in biochemical assays in which this β clamp-binding antibiotic collapses pre-formed ImuB-β clamp complexes. These observations establish the essentiality of the ImuB-β clamp interaction for mutagenic DNA repair in mycobacteria, identifying the mutasome as target for adjunctive therapeutics designed to protect anti-TB drugs against emerging resistance., Competing Interests: SG, ZM, MR, JS, RD, AR, ND, Td, SA, DL, JA, TC, JH, RM, JM, RW, VM, ČV, ML, DW No competing interests declared- Published
- 2023
- Full Text
- View/download PDF
8. RecA-NT homology motif in ImuB is essential for mycobacterial ImuA'-ImuB protein interaction and mutasome function.
- Author
-
Santos JA, Timinskas K, Lamers MH, Venclovas Č, Warner DF, and Gessner SJ
- Abstract
The mycobacterial mutasome - minimally comprising ImuA', ImuB, and DnaE2 proteins - has been implicated in DNA damage-induced mutagenesis in Mycobacterium tuberculosis . ImuB, predicted to enable mutasome function via its interaction with the β clamp, is a catalytically inactive member of the Y-family of DNA polymerases. Like other members of the Y family, ImuB features a recently identified amino acid motif with homology to the RecA-N-terminus (RecA-NT). In RecA, the motif mediates oligomerization of RecA monomers into RecA filaments. Given the role of ImuB in the mycobacterial mutasome, we hypothesized that the ImuB RecA-NT motif might mediate its interaction with ImuA', a RecA homolog of unknown function. To investigate this possibility, we constructed a panel of imuB alleles in which RecA-NT was removed, or mutated. Results from microbiological and biochemical assays indicate that RecA-NT is critical for the interaction of ImuB with ImuA'. A region downstream of RecA-NT (ImuB-C) also appears to stabilize the ImuB-ImuA' interaction, but its removal does not prevent complex formation. In contrast, replacing two key hydrophobic residues of RecA-NT, L378 and V383, is sufficient to disrupt ImuA'-ImuB interaction. To our knowledge, this constitutes the first experimental evidence showing the role of the RecA-NT motif in mediating the interaction between a Y-family member and a RecA homolog., Competing Interests: CONFLICT OF INTEREST The authors declare that they have no competing interests.
- Published
- 2023
- Full Text
- View/download PDF
9. High-Throughput Exonuclease Assay Based on the Fluorescent Base Analogue 2-Aminopurine.
- Author
-
Botto MM, Murthy S, and Lamers MH
- Abstract
Exonucleases are essential enzymes that remove nucleotides from free DNA ends during DNA replication, DNA repair, and telomere maintenance. Due to their essential role, they are potential targets for novel anticancer and antimicrobial drugs but have so far been little exploited. Here, we present a simple and versatile real-time exonuclease assay based on 2-aminopurine, an intrinsically fluorescent nucleotide that is quenched by neighboring bases when embedded in DNA. We show that our assay is applicable to different eukaryotic and bacterial exonucleases acting on both 3' and 5' DNA ends over a wide range of protein activities and suitable for a high-throughput inhibitor screening campaign. Using our assay, we discover a novel inhibitor of the Mycobacterium tuberculosis PHP-exonuclease that is part of the replicative DNA polymerase DnaE1. Hence, our novel assay will be a useful tool for high-throughput screening for novel exonuclease inhibitors that may interfere with DNA replication or DNA maintenance., Competing Interests: The authors declare no competing financial interest., (© 2023 The Authors. Published by American Chemical Society.)
- Published
- 2023
- Full Text
- View/download PDF
10. MutL binds to 3' resected DNA ends and blocks DNA polymerase access.
- Author
-
Borsellini A, Lebbink JHG, and Lamers MH
- Subjects
- DNA metabolism, DNA-Directed DNA Polymerase metabolism, Escherichia coli genetics, Escherichia coli metabolism, DNA Mismatch Repair, Escherichia coli Proteins metabolism, MutL Proteins genetics
- Abstract
DNA mismatch repair removes mis-incorporated bases after DNA replication and reduces the error rate a 100-1000-fold. After recognition of a mismatch, a large section of up to a thousand nucleotides is removed from the daughter strand followed by re-synthesis. How these opposite activities are coordinated is poorly understood. Here we show that the Escherichia coli MutL protein binds to the 3' end of the resected strand and blocks access of Pol I and Pol III. The cryo-EM structure of an 85-kDa MutL-DNA complex, determined to 3.7 Å resolution, reveals a unique DNA binding mode that positions MutL at the 3' end of a primer-template, but not at a 5' resected DNA end or a blunt DNA end. Hence, our work reveals a novel role for MutL in the final stages of mismatch repair by preventing premature DNA synthesis during removal of the mismatched strand., (© The Author(s) 2022. Published by Oxford University Press on behalf of Nucleic Acids Research.)
- Published
- 2022
- Full Text
- View/download PDF
11. DNA-Dependent Binding of Nargenicin to DnaE1 Inhibits Replication in Mycobacterium tuberculosis .
- Author
-
Chengalroyen MD, Mason MK, Borsellini A, Tassoni R, Abrahams GL, Lynch S, Ahn YM, Ambler J, Young K, Crowley BM, Olsen DB, Warner DF, Barry Iii CE, Boshoff HIM, Lamers MH, and Mizrahi V
- Subjects
- Anti-Bacterial Agents pharmacology, Cryoelectron Microscopy, DNA-Directed DNA Polymerase, Humans, Mycobacterium tuberculosis genetics, Tuberculosis drug therapy, Tuberculosis microbiology
- Abstract
Natural products provide a rich source of potential antimicrobials for treating infectious diseases for which drug resistance has emerged. Foremost among these diseases is tuberculosis. Assessment of the antimycobacterial activity of nargenicin, a natural product that targets the replicative DNA polymerase of Staphylococcus aureus , revealed that it is a bactericidal genotoxin that induces a DNA damage response in Mycobacterium tuberculosis ( Mtb ) and inhibits growth by blocking the replicative DNA polymerase, DnaE1. Cryo-electron microscopy revealed that binding of nargenicin to Mtb DnaE1 requires the DNA substrate such that nargenicin is wedged between the terminal base pair and the polymerase and occupies the position of both the incoming nucleotide and templating base. Comparative analysis across three bacterial species suggests that the activity of nargenicin is partly attributable to the DNA binding affinity of the replicative polymerase. This work has laid the foundation for target-led drug discovery efforts focused on Mtb DnaE1.
- Published
- 2022
- Full Text
- View/download PDF
12. Cryogenic electron microscopy structures reveal how ATP and DNA binding in MutS coordinates sequential steps of DNA mismatch repair.
- Author
-
Borsellini A, Kunetsky V, Friedhoff P, and Lamers MH
- Subjects
- Adenosine Diphosphate metabolism, Catalytic Domain, Escherichia coli, Hydrolysis, Models, Molecular, Protein Binding, Protein Multimerization, Adenosine Triphosphate metabolism, Cryoelectron Microscopy, DNA metabolism, DNA Mismatch Repair, Escherichia coli Proteins metabolism, Escherichia coli Proteins ultrastructure, MutS DNA Mismatch-Binding Protein metabolism, MutS DNA Mismatch-Binding Protein ultrastructure
- Abstract
DNA mismatch repair detects and corrects mismatches introduced during DNA replication. The protein MutS scans for mismatches and coordinates the repair cascade. During this process, MutS undergoes multiple conformational changes in response to ATP binding, hydrolysis and release, but how ATP induces the various MutS conformations is incompletely understood. Here we present four cryogenic electron microscopy structures of Escherichia coli MutS at sequential stages of the ATP hydrolysis cycle that reveal how ATP binding and hydrolysis induce closing and opening of the MutS dimer, respectively. Biophysical analysis demonstrates how DNA binding modulates the ATPase cycle by prevention of hydrolysis during scanning and mismatch binding, while preventing ADP release in the sliding clamp state. Nucleotide release is achieved when MutS encounters single-stranded DNA that is produced during removal of the daughter strand. The combination of ATP binding and hydrolysis and its modulation by DNA enables MutS to adopt the different conformations needed to coordinate the sequential steps of the mismatch repair cascade., (© 2022. The Author(s), under exclusive licence to Springer Nature America, Inc.)
- Published
- 2022
- Full Text
- View/download PDF
13. The selection process of licensing a DNA mismatch for repair.
- Author
-
Fernandez-Leiro R, Bhairosing-Kok D, Kunetsky V, Laffeber C, Winterwerp HH, Groothuizen F, Fish A, Lebbink JHG, Friedhoff P, Sixma TK, and Lamers MH
- Subjects
- Cryoelectron Microscopy, DNA genetics, DNA Mismatch Repair genetics, DNA Repair genetics, DNA Replication genetics, Escherichia coli genetics, Escherichia coli ultrastructure, Escherichia coli Proteins genetics, MutL Proteins genetics, MutS DNA Mismatch-Binding Protein genetics, DNA ultrastructure, Escherichia coli Proteins ultrastructure, MutL Proteins ultrastructure, MutS DNA Mismatch-Binding Protein ultrastructure, Protein Conformation
- Abstract
DNA mismatch repair detects and removes mismatches from DNA by a conserved mechanism, reducing the error rate of DNA replication by 100- to 1,000-fold. In this process, MutS homologs scan DNA, recognize mismatches and initiate repair. How the MutS homologs selectively license repair of a mismatch among millions of matched base pairs is not understood. Here we present four cryo-EM structures of Escherichia coli MutS that provide snapshots, from scanning homoduplex DNA to mismatch binding and MutL activation via an intermediate state. During scanning, the homoduplex DNA forms a steric block that prevents MutS from transitioning into the MutL-bound clamp state, which can only be overcome through kinking of the DNA at a mismatch. Structural asymmetry in all four structures indicates a division of labor between the two MutS monomers. Together, these structures reveal how a small conformational change from the homoduplex- to heteroduplex-bound MutS acts as a licensing step that triggers a dramatic conformational change that enables MutL binding and initiation of the repair cascade.
- Published
- 2021
- Full Text
- View/download PDF
14. Novel Antibiotics Targeting Bacterial Replicative DNA Polymerases.
- Author
-
Santos JA and Lamers MH
- Abstract
Multidrug resistance is a worldwide problem that is an increasing threat to global health. Therefore, the development of new antibiotics that inhibit novel targets is of great urgency. Some of the most successful antibiotics inhibit RNA transcription, RNA translation, and DNA replication. Transcription and translation are inhibited by directly targeting the RNA polymerase or ribosome, respectively. DNA replication, in contrast, is inhibited indirectly through targeting of DNA gyrases, and there are currently no antibiotics that inhibit DNA replication by directly targeting the replisome. This contrasts with antiviral therapies where the viral replicases are extensively targeted. In the last two decades there has been a steady increase in the number of compounds that target the bacterial replisome. In particular a variety of inhibitors of the bacterial replicative polymerases PolC and DnaE have been described, with one of the DNA polymerase inhibitors entering clinical trials for the first time. In this review we will discuss past and current work on inhibition of DNA replication, and the potential of bacterial DNA polymerase inhibitors in particular as attractive targets for a new generation of antibiotics.
- Published
- 2020
- Full Text
- View/download PDF
15. Polymerization and editing modes of a high-fidelity DNA polymerase are linked by a well-defined path.
- Author
-
Dodd T, Botto M, Paul F, Fernandez-Leiro R, Lamers MH, and Ivanov I
- Subjects
- DNA chemistry, DNA Polymerase III metabolism, DNA Primers, DNA-Directed DNA Polymerase chemistry, Escherichia coli metabolism, Exonucleases metabolism, Kinetics, Models, Molecular, Protein Conformation, DNA metabolism, DNA Replication physiology, DNA-Directed DNA Polymerase metabolism, Polymerization
- Abstract
Proofreading by replicative DNA polymerases is a fundamental mechanism ensuring DNA replication fidelity. In proofreading, mis-incorporated nucleotides are excised through the 3'-5' exonuclease activity of the DNA polymerase holoenzyme. The exonuclease site is distal from the polymerization site, imposing stringent structural and kinetic requirements for efficient primer strand transfer. Yet, the molecular mechanism of this transfer is not known. Here we employ molecular simulations using recent cryo-EM structures and biochemical analyses to delineate an optimal free energy path connecting the polymerization and exonuclease states of E. coli replicative DNA polymerase Pol III. We identify structures for all intermediates, in which the transitioning primer strand is stabilized by conserved Pol III residues along the fingers, thumb and exonuclease domains. We demonstrate switching kinetics on a tens of milliseconds timescale and unveil a complete pol-to-exo switching mechanism, validated by targeted mutational experiments.
- Published
- 2020
- Full Text
- View/download PDF
16. Reduced structural flexibility for an exonuclease deficient DNA polymerase III mutant.
- Author
-
Gahlon HL, Walker AR, Cisneros GA, Lamers MH, and Rueda DS
- Subjects
- Base Pair Mismatch, DNA chemistry, DNA genetics, DNA Polymerase III chemistry, DNA Polymerase III genetics, Escherichia coli chemistry, Exonucleases chemistry, Exonucleases genetics, Fluorescence Resonance Energy Transfer methods, Kinetics, Molecular Dynamics Simulation, Protein Binding, Protein Conformation, Protein Subunits, DNA metabolism, DNA Polymerase III metabolism, Exonucleases metabolism
- Abstract
DNA synthesis, carried out by DNA polymerases, requires balancing speed and accuracy for faithful replication of the genome. High fidelity DNA polymerases contain a 3'-5' exonuclease domain that can remove misincorporated nucleotides on the 3' end of the primer strand, a process called proofreading. The E. coli replicative polymerase, DNA polymerase III, has spatially separated (∼55 Å apart) polymerase and exonuclease subunits. Here, we report on the dynamics of E. coli DNA polymerase III proofreading in the presence of its processivity factor, the β2-sliding clamp, at varying base pair termini using single-molecule FRET. We find that the binding kinetics do not depend on the base identity at the termini, indicating a tolerance for DNA mismatches. Further, our single-molecule data and MD simulations show two previously unobserved features: (1) DNA Polymerase III is a highly dynamic protein that adopts multiple conformational states while bound to DNA with matched or mismatched ends, and (2) an exonuclease-deficient DNA polymerase III has reduced conformational flexibility. Overall, our single-molecule experiments provide high time-resolution insight into a mechanism that ensures high fidelity DNA replication to maintain genome integrity.
- Published
- 2018
- Full Text
- View/download PDF
17. Single-molecule studies contrast ordered DNA replication with stochastic translesion synthesis.
- Author
-
Zhao G, Gleave ES, and Lamers MH
- Subjects
- Single Molecule Imaging, DNA Polymerase II metabolism, DNA Polymerase III metabolism, DNA Polymerase beta metabolism, DNA Replication, DNA, Bacterial biosynthesis, Escherichia coli enzymology, Escherichia coli genetics
- Abstract
High fidelity replicative DNA polymerases are unable to synthesize past DNA adducts that result from diverse chemicals, reactive oxygen species or UV light. To bypass these replication blocks, cells utilize specialized translesion DNA polymerases that are intrinsically error prone and associated with mutagenesis, drug resistance, and cancer. How untimely access of translesion polymerases to DNA is prevented is poorly understood. Here we use co-localization single-molecule spectroscopy (CoSMoS) to follow the exchange of the E. coli replicative DNA polymerase Pol IIIcore with the translesion polymerases Pol II and Pol IV. We find that in contrast to the toolbelt model, the replicative and translesion polymerases do not form a stable complex on one clamp but alternate their binding. Furthermore, while the loading of clamp and Pol IIIcore is highly organized, the exchange with the translesion polymerases is stochastic and is not determined by lesion-recognition but instead a concentration-dependent competition between the polymerases.
- Published
- 2017
- Full Text
- View/download PDF
18. High-fidelity DNA replication in Mycobacterium tuberculosis relies on a trinuclear zinc center.
- Author
-
Baños-Mateos S, van Roon AM, Lang UF, Maslen SL, Skehel JM, and Lamers MH
- Subjects
- Cryoelectron Microscopy, Deoxyribonuclease IV (Phage T4-Induced), Exodeoxyribonucleases ultrastructure, Molecular Structure, Zinc isolation & purification, DNA Replication, Exodeoxyribonucleases metabolism, Mycobacterium tuberculosis enzymology
- Abstract
High-fidelity DNA replication depends on a proofreading 3'-5' exonuclease that is associated with the replicative DNA polymerase. The replicative DNA polymerase DnaE1 from the major pathogen Mycobacterium tuberculosis (Mtb) uses its intrinsic PHP-exonuclease that is distinct from the canonical DEDD exonucleases found in the Escherichia coli and eukaryotic replisomes. The mechanism of the PHP-exonuclease is not known. Here, we present the crystal structure of the Mtb DnaE1 polymerase. The PHP-exonuclease has a trinuclear zinc center, coordinated by nine conserved residues. Cryo-EM analysis reveals the entry path of the primer strand in the PHP-exonuclease active site. Furthermore, the PHP-exonuclease shows a striking similarity to E. coli endonuclease IV, which provides clues regarding the mechanism of action. Altogether, this work provides important insights into the PHP-exonuclease and reveals unique properties that make it an attractive target for novel anti-mycobacterial drugs.The polymerase and histidinol phosphatase (PHP) domain in the DNA polymerase DnaE1 is essential for mycobacterial high-fidelity DNA replication. Here, the authors determine the DnaE1 crystal structure, which reveals the PHP-exonuclease mechanism that can be exploited for antibiotic development.
- Published
- 2017
- Full Text
- View/download PDF
19. DNA Replication in Mycobacterium tuberculosis.
- Author
-
Ditse Z, Lamers MH, and Warner DF
- Subjects
- Animals, Humans, Adaptation, Biological, DNA Replication, Mutation, Mycobacterium tuberculosis genetics, Mycobacterium tuberculosis growth & development
- Abstract
Faithful replication and maintenance of the genome are essential to the ability of any organism to survive and propagate. For an obligate pathogen such as Mycobacterium tuberculosis that has to complete successive cycles of transmission, infection, and disease in order to retain a foothold in the human population, this requires that genome replication and maintenance must be accomplished under the metabolic, immune, and antibiotic stresses encountered during passage through variable host environments. Comparative genomic analyses have established that chromosomal mutations enable M. tuberculosis to adapt to these stresses: the emergence of drug-resistant isolates provides direct evidence of this capacity, so too the well-documented genetic diversity among M. tuberculosis lineages across geographic loci, as well as the microvariation within individual patients that is increasingly observed as whole-genome sequencing methodologies are applied to clinical samples and tuberculosis (TB) disease models. However, the precise mutagenic mechanisms responsible for M. tuberculosis evolution and adaptation are poorly understood. Here, we summarize current knowledge of the machinery responsible for DNA replication in M. tuberculosis, and discuss the potential contribution of the expanded complement of mycobacterial DNA polymerases to mutagenesis. We also consider briefly the possible role of DNA replication-in particular, its regulation and coordination with cell division-in the ability of M. tuberculosis to withstand antibacterial stresses, including host immune effectors and antibiotics, through the generation at the population level of a tolerant state, or through the formation of a subpopulation of persister bacilli-both of which might be relevant to the emergence and fixation of genetic drug resistance.
- Published
- 2017
- Full Text
- View/download PDF
20. Self-correcting mismatches during high-fidelity DNA replication.
- Author
-
Fernandez-Leiro R, Conrad J, Yang JC, Freund SM, Scheres SH, and Lamers MH
- Subjects
- Base Pairing, Base Sequence, Catalytic Domain, Cryoelectron Microscopy, DNA Mismatch Repair, DNA, Bacterial chemistry, DNA, Bacterial genetics, Escherichia coli enzymology, Escherichia coli genetics, Nucleic Acid Conformation, Protein Binding, Protein Conformation, alpha-Helical, Protein Structure, Quaternary, Protein Subunits chemistry, DNA Polymerase III chemistry, DNA Replication
- Abstract
Faithful DNA replication is essential to all forms of life and depends on the action of 3'-5' exonucleases that remove misincorporated nucleotides from the newly synthesized strand. However, how the DNA is transferred from the polymerase to the exonuclease active site is not known. Here we present the cryo-EM structure of the editing mode of the catalytic core of the Escherichia coli replisome, revealing a dramatic distortion of the DNA whereby the polymerase thumb domain acts as a wedge that separates the two DNA strands. Importantly, NMR analysis of the DNA substrate shows that the presence of a mismatch increases the fraying of the DNA, thus enabling it to reach the exonuclease active site. Therefore the mismatch corrects itself, whereas the exonuclease subunit plays a passive role. Hence, our work provides unique insights into high-fidelity replication and establishes a new paradigm for the correction of misincorporated nucleotides., Competing Interests: The authors declare no competing financial interests.
- Published
- 2017
- Full Text
- View/download PDF
21. cryo-EM structures of the E. coli replicative DNA polymerase reveal its dynamic interactions with the DNA sliding clamp, exonuclease and τ .
- Author
-
Fernandez-Leiro R, Conrad J, Scheres SH, and Lamers MH
- Abstract
The replicative DNA polymerase PolIIIα from Escherichia coli is a uniquely fast and processive enzyme. For its activity it relies on the DNA sliding clamp β, the proofreading exonuclease ε and the C-terminal domain of the clamp loader subunit τ. Due to the dynamic nature of the four-protein complex it has long been refractory to structural characterization. Here we present the 8 Å resolution cryo-electron microscopy structures of DNA-bound and DNA-free states of the PolIII-clamp-exonuclease-τ
c complex. The structures show how the polymerase is tethered to the DNA through multiple contacts with the clamp and exonuclease. A novel contact between the polymerase and clamp is made in the DNA bound state, facilitated by a large movement of the polymerase tail domain and τc . These structures provide crucial insights into the organization of the catalytic core of the replisome and form an important step towards determining the structure of the complete holoenzyme.- Published
- 2015
- Full Text
- View/download PDF
22. Structural characterization of the principal mRNA-export factor Mex67-Mtr2 from Chaetomium thermophilum.
- Author
-
Aibara S, Valkov E, Lamers MH, Dimitrova L, Hurt E, and Stewart M
- Subjects
- Crystallography, X-Ray methods, Membrane Transport Proteins metabolism, Nuclear Proteins metabolism, Nucleocytoplasmic Transport Proteins metabolism, Protein Binding physiology, Protein Structure, Secondary, Protein Structure, Tertiary, RNA, Messenger metabolism, RNA-Binding Proteins metabolism, Saccharomyces cerevisiae Proteins metabolism, Scattering, Small Angle, Chaetomium, Membrane Transport Proteins chemistry, Nuclear Proteins chemistry, Nucleocytoplasmic Transport Proteins chemistry, RNA, Messenger chemistry, RNA-Binding Proteins chemistry, Saccharomyces cerevisiae Proteins chemistry
- Abstract
Members of the Mex67-Mtr2/NXF-NXT1 family are the principal mediators of the nuclear export of mRNA. Mex67/NXF1 has a modular structure based on four domains (RRM, LRR, NTF2-like and UBA) that are thought to be present across species, although the level of sequence conservation between organisms, especially in lower eukaryotes, is low. Here, the crystal structures of these domains from the thermophilic fungus Chaetomium thermophilum are presented together with small-angle X-ray scattering (SAXS) and in vitro RNA-binding data that indicate that, not withstanding the limited sequence conservation between different NXF family members, the molecules retain similar structural and RNA-binding properties. Moreover, the resolution of crystal structures obtained with the C. thermophilum domains was often higher than that obtained previously and, when combined with solution and biochemical studies, provided insight into the structural organization, self-association and RNA-binding properties of Mex67-Mtr2 that facilitate mRNA nuclear export.
- Published
- 2015
- Full Text
- View/download PDF
23. DNA replication fidelity in Mycobacterium tuberculosis is mediated by an ancestral prokaryotic proofreader.
- Author
-
Rock JM, Lang UF, Chase MR, Ford CB, Gerrick ER, Gawande R, Coscolla M, Gagneux S, Fortune SM, and Lamers MH
- Subjects
- Amino Acid Sequence, Antitubercular Agents pharmacology, DNA, Bacterial genetics, Drug Resistance, Bacterial, Microbial Sensitivity Tests, Molecular Sequence Data, Mycobacterium tuberculosis drug effects, Mycobacterium tuberculosis genetics, Phenotype, Phylogeny, Polymorphism, Single Nucleotide, Bacterial Proteins genetics, DNA Polymerase III genetics, DNA Replication, Mycobacterium tuberculosis enzymology
- Abstract
The DNA replication machinery is an important target for antibiotic development in increasingly drug-resistant bacteria, including Mycobacterium tuberculosis. Although blocking DNA replication leads to cell death, disrupting the processes used to ensure replication fidelity can accelerate mutation and the evolution of drug resistance. In Escherichia coli, the proofreading subunit of the replisome, the ɛ exonuclease, is essential for high-fidelity DNA replication; however, we find that the corresponding subunit is completely dispensable in M. tuberculosis. Rather, the mycobacterial replicative polymerase DnaE1 itself encodes an editing function that proofreads DNA replication, mediated by an intrinsic 3'-5' exonuclease activity within its PHP domain. Inactivation of the DnaE1 PHP domain increases the mutation rate by more than 3,000-fold. Moreover, phylogenetic analysis of DNA replication proofreading in the bacterial kingdom suggests that E. coli is a phylogenetic outlier and that PHP domain-mediated proofreading is widely conserved and indeed may be the ancestral prokaryotic proofreader.
- Published
- 2015
- Full Text
- View/download PDF
24. Aminoadamantanes versus other antiviral drugs for chronic hepatitis C.
- Author
-
Lamers MH, Broekman M, Drenth JP, and Gluud C
- Subjects
- Humans, Interferon-alpha therapeutic use, Interferon-gamma therapeutic use, Mycophenolic Acid analogs & derivatives, Mycophenolic Acid therapeutic use, Randomized Controlled Trials as Topic, Ribavirin therapeutic use, Amantadine therapeutic use, Antiviral Agents therapeutic use, Hepatitis C, Chronic drug therapy
- Abstract
Background: Hepatitis C virus infection affects around 3% of the world population or approximately 160 million people. A variable proportion (5% to 40%) of the infected people develop clinical symptoms. Hence, hepatitis C virus is a leading cause of liver-related morbidity and mortality with hepatic fibrosis, end-stage liver cirrhosis, and hepatocellular carcinoma as the dominant clinical sequelae. Combination therapy with pegylated (peg) interferon-alpha and ribavirin achieves sustained virological response (that is, undetectable hepatitis C virus RNA in serum by sensitivity testing six months after the end of treatment) in approximately 40% to 80% of treated patients, depending on viral genotype. Recently, a new class of drugs have emerged for hepatitis C infection, the direct acting antivirals, which in combination with standard therapy or alone can lead to sustained virological response in 80% or more of treated patients. Aminoadamantanes, mostly amantadine, are antiviral drugs used for the treatment of patients with chronic hepatitis C. We have previously systematically reviewed amantadine versus placebo or no intervention and found no significant effects of the amantadine on all-cause mortality or liver-related morbidity and on adverse events in patients with hepatitis C. Overall, we did not observe a significant effect of amantadine on sustained virological response. In this review, we systematically review aminoadamantanes versus other antiviral drugs., Objectives: To assess the beneficial and harmful effects of aminoadamantanes versus other antiviral drugs for patients with chronic hepatitis C virus infection by conducting a systematic review with meta-analyses and trial sequential analyses of randomised clinical trials., Search Methods: The Cochrane Hepato-Biliary Group Controlled Trials Register (1996 to December 2013), the Cochrane Central Register of Controlled Trials (CENTRAL) (Issue 11 of 12, 2013), MEDLINE (1946 to December 2013), EMBASE (1974 to December 2013), Science Citation Index EXPANDED (1900 to December 2013), the WHO International Clinical Trials Registry Platform (www.who.int/ictrp), Google Scholar, and Eudrapharm up to December 2013. Furthermore, full text searches were conducted until December 2013., Selection Criteria: Randomised clinical trials assessing aminoadamantanes in participants with chronic hepatitis C virus infection., Data Collection and Analysis: Two authors independently extracted data. RevMan Analysis was used for statistical analysis of dichotomous data using risk ratio (RR) with 95% confidence intervals (CI). Methodological domains were used to assess the risk of systematic errors ('bias'). We used trial sequential analysis to assess risk of random errors ('play of chance')., Main Results: Six randomised clinical trials with 581 participants with chronic hepatitis C were included. All trials had high risk of bias. The included trials compared amantadine versus other antiviral drugs: ribavirin, mycophenolate mofetil, interferon-alpha, or interferon-gamma. Standard antiviral therapy (interferon-alpha, interferon-alpha plus ribavirin, or peg interferon alpha) was administered equally to the intervention and the control groups in five trials, depending on when the trial was conducted. Four trials compared amantadine versus ribavirin. There were no deaths or liver-related morbidity in the two intervention groups (0/216 (0%) versus 0/211 (0%); 4 trials; very low quality of the evidence). The lower estimated risk for (serious) adverse events leading to treatment discontinuation with amantadine was imprecise (RR 0.56, 95% CI 0.27 to 1.16; based on 10/216 (5%) versus 18/211 (9%) participants in 4 trials; very low quality of the evidence). There were more participants with failure of sustained virological response in the amantadine group than in the ribavirin group (206/216 (96%) versus 176/211 (84%); RR 1.14, 95% CI 1.07 to 1.22, 4 trials; low quality of the evidence). Amantadine versus ribavirin more often failed to achieve end-of follow-up biochemical response (41/46 (89%) versus 31/46 (67%); RR 1.31, 95% CI 1.05 to 1.63; 2 trials; very low quality of the evidence). One trial compared amantadine versus mycophenolate mofetil. There were no significant differences between the two treatment groups, except that amantadine was inferior to mycophenolate mofetil regarding the outcome failure to achieve end-of treatment virological response (low quality of evidence). One trial each compared amantadine versus interferon-alpha or interferon-gamma. Both comparisons showed no significant differences in the treatment outcomes (very low quality of the evidence). The observed effects could be due to real effects, systematic errors (bias), or random errors (play of chance). This possible influence on the observed effect by play of chance is due to the fact that trial sequential analyses could not confirm our findings. We were not able to perform meta-analyses on failure of histological improvement and quality of life due to lack of valid data in all trial comparisons., Authors' Conclusions: This systematic review has identified evidence of very low quality for the key outcomes of all-cause mortality or liver-related morbidity and adverse events in people with chronic hepatitis C when treated with amantadine compared with ribavirin, mycophenolate, interferon-alpha, or interferon-gamma. The timeframe for measuring the composite outcome was insufficient in the included trials. There was low quality evidence that amantadine led to more participants who failed to achieve sustained virological response compared with ribavirin. This observation may be real or caused by systematic errors (bias), but it does not seem to be caused by random error (play of chance). Due to the low quality of the evidence, we are unable to determine definitively whether amantadine is less effective than other antivirals in patients with chronic hepatitis C. As it appears less likely that future trials assessing amantadine or potentially other aminoadamantanes for patients with chronic hepatitis C would show strong benefits, it is probably better to focus on the assessments of other direct acting antiviral drugs. We found no evidence assessing other aminoadamantanes in randomised clinical trials in order to recommend or refute their use.
- Published
- 2014
- Full Text
- View/download PDF
25. Aminoadamantanes for chronic hepatitis C.
- Author
-
Lamers MH, Broekman M, Drenth JP, and Gluud C
- Subjects
- Adult, Drug Therapy, Combination methods, Hepatitis C, Chronic virology, Humans, Interferon-alpha therapeutic use, Polyethylene Glycols therapeutic use, Randomized Controlled Trials as Topic, Recombinant Proteins therapeutic use, Ribavirin therapeutic use, Treatment Outcome, Amantadine therapeutic use, Antiviral Agents therapeutic use, Hepatitis C, Chronic drug therapy
- Abstract
Background: Around 3% of the world's population (approximately 160 million people) are chronically infected with hepatitis C virus. The proportion of infected people who develop clinical symptoms varies between 5% and 40%. Combination therapy with pegylated interferon-alpha plus ribavirin eradicates the virus from the blood six months after treatment (sustained virological response) in approximately 40% to 80% of infected patients, depending on the viral genotype. New antiviral agents, such as boceprevir and telaprevir, in combination with standard therapy, can increase sustained virological response in genotype 1 infected patients to at least 70%. There is therefore an unmet need for drugs that can achieve a higher proportion of sustained virological response. Aminoadamantanes are antiviral drugs used for treatment of patients with chronic hepatitis C., Objectives: To assess the beneficial and harmful effects of aminoadamantanes for patients with chronic hepatitis C infection by conducting a systematic review with meta-analyses of randomised clinical trials, as well as trial sequential analyses., Search Methods: We conducted electronic searches of the Cochrane Hepato-Biliary Group Controlled Trials Register (1996 to December 2013), the Cochrane Central Register of Controlled Trials (CENTRAL) 2013, Issue 11 of 12 (1995 to December 2013), MEDLINE (1946 to December 2013), EMBASE (1974 to December 2013), Science Citation Index EXPANDED (1900 to December 2013), the WHO International Clinical Trials Registry Platform (www.who.int/ictrp), Google Scholar, and Eudrapharm up to December 2013 and checked the reference lists of identified publications., Selection Criteria: Randomised clinical trials assessing aminoadamantanes in patients with chronic hepatitis C infection., Data Collection and Analysis: Two authors independently extracted data. We assessed for risks of systematic errors ('bias') using the 'Risk of bias' tool. We analysed dichotomous data with risk ratio (RR) and continuous data with mean difference (MD) or standardised mean difference (SMD), both with 95% confidence intervals (CI). We used trial sequential analysis to assess the risk of random errors ('play of chance'). We assessed quality using the GRADE system., Main Results: We included 41 randomised clinical trials with 6193 patients with chronic hepatitis C. All trials had high risk of bias. All included trials compared amantadine versus placebo or no intervention. Standard antiviral therapy was administered equally to the intervention and the control groups in 40 trials. The standard antiviral therapy, which was administered to both intervention groups, was interferon-alpha, interferon-alpha plus ribavirin, and peg interferon-alpha plus ribavirin, depending on the time when the trial was conducted.When we meta-analysed all trials together, the overall results demonstrated no significant effects of amantadine, when compared with placebo or no intervention, on our all-cause mortality or liver-related morbidity composite outcome (5/2353 (0.2%) versus 6/2264 (0.3%); RR 0.90, 95% CI 0.38 to 2.17; I² = 0%; 32 trials; very low quality). There was also no significant effect on adverse events (288/2869 (10%) versus 293/2777 (11%); RR 0.98, 95% CI 0.84 to 1.14; I² = 0%; 35 trials; moderate quality). We used both fixed-effect and random-effects meta-analyses. Amantadine, when compared with placebo or no intervention, did not significantly influence the number of patients who failed to achieve a sustained virological response (1821/2861 (64%) versus 1737/2721 (64%); RR 0.98, 95% CI 0.95 to 1.02; I² = 35%; 35 trials; moderate quality). However, in the subgroup using interferon plus ribavirin, amantadine decreased the number of patients who failed to achieve a sustained virological response (422/666 (63%) versus 447/628 (71%); RR 0.89, 95% CI 0.83 to 0.96; I² = 41%; 11 trials; low quality). Similar results were found for failure to achieve an end of treatment virological response. Amantadine, when compared with placebo or no intervention, significantly decreased the number of patients without normalisation of alanine aminotransferase (ALT) serum levels at the end of treatment (671/1141 (59%) versus 732/1100 (67%); RR 0.88, 95% CI 0.83 to 0.94; I² = 47%; 19 trials; low quality). Amantadine, when compared with placebo or no intervention, did not significantly influence the end of follow-up biochemical response (1133/1896 (60%) versus 1151/1848 (62%); RR 0.95, 95% CI 0.91 to 1.00; I² = 49%; 21 trials; low quality).The observed beneficial effects could be true effects but could also be due to both systematic errors (bias) and random errors (play of chance). The latter is due to the fact that trial sequential analyses could not confirm or refute our findings. We were not able to perform meta-analyses for failure of histological improvement or quality of life due to a lack of valid data., Authors' Conclusions: This systematic review does not demonstrate any significant effects of amantadine on all-cause mortality or liver-related morbidity composite outcome and on adverse events in patients with hepatitis C; however, the median trial duration was 12 months, with a median follow-up of six months, which is not long enough to assess the composite outcome sufficiently. Overall, we did not see an effect of amantadine on failure to achieve a sustained virological response. Subgroup analyses demonstrated that the combination of amantadine plus interferon-alpha and ribavirin seems to increase the number of patients achieving a sustained virological response. This finding may be caused by both systematic errors (bias) and risks of random errors (play of chance), but it could also be real. Based on the results of the overall evidence, it appears less likely that future trials assessing amantadine for patients with chronic hepatitis C will show strong benefits. Therefore, it is probably advisable to wait for the results of trials assessing other direct-acting antiviral drugs. In the absence of convincing evidence of benefit, the use of amantadine is justified in the context of randomised clinical trials assessing the effects of combination therapy. We found a lack of evidence on other aminoadamantanes than amantadine.
- Published
- 2014
- Full Text
- View/download PDF
26. Hekate: software suite for the mass spectrometric analysis and three-dimensional visualization of cross-linked protein samples.
- Author
-
Holding AN, Lamers MH, Stephens E, and Skehel JM
- Subjects
- Amino Acid Sequence, Animals, Cattle, Cross-Linking Reagents chemistry, Escherichia coli chemistry, Escherichia coli enzymology, Molecular Sequence Data, Cytochromes c chemistry, DNA Polymerase III chemistry, Escherichia coli Proteins chemistry, Mass Spectrometry statistics & numerical data, Models, Molecular, Software
- Abstract
Chemical cross-linking of proteins combined with mass spectrometry provides an attractive and novel method for the analysis of native protein structures and protein complexes. Analysis of the data however is complex. Only a small number of cross-linked peptides are produced during sample preparation and must be identified against a background of more abundant native peptides. To facilitate the search and identification of cross-linked peptides, we have developed a novel software suite, named Hekate. Hekate is a suite of tools that address the challenges involved in analyzing protein cross-linking experiments when combined with mass spectrometry. The software is an integrated pipeline for the automation of the data analysis workflow and provides a novel scoring system based on principles of linear peptide analysis. In addition, it provides a tool for the visualization of identified cross-links using three-dimensional models, which is particularly useful when combining chemical cross-linking with other structural techniques. Hekate was validated by the comparative analysis of cytochrome c (bovine heart) against previously reported data. Further validation was carried out on known structural elements of DNA polymerase III, the catalytic α-subunit of the Escherichia coli DNA replisome along with new insight into the previously uncharacterized C-terminal domain of the protein.
- Published
- 2013
- Full Text
- View/download PDF
27. Treatment of hepatitis C monoinfection in adults--Dutch national guidelines.
- Author
-
Lamers MH, Broekman MM, Boucher CA, Brouwer JT, Burger DM, van Hoek B, Hoepelman AI, de Knegt RJ, Reesink HW, and Drenth JP
- Subjects
- Adult, Antiviral Agents adverse effects, Drug Interactions, Genotype, Humans, Interferon alpha-2, Interferon-alpha therapeutic use, Netherlands, Oligopeptides adverse effects, Polyethylene Glycols therapeutic use, Proline adverse effects, Proline therapeutic use, Recombinant Proteins therapeutic use, Ribavirin therapeutic use, Antiviral Agents therapeutic use, Hepacivirus genetics, Hepatitis C, Chronic drug therapy, Oligopeptides therapeutic use, Proline analogs & derivatives
- Abstract
In this new Dutch guideline for hepatitis C virus infection we provide recommendations for the management of hepatitis C infection. Until 2012 the standard for treatment consisted of pegylated interferon alpha (peg-IFNa) and ribavirin. The advent of first-generation direct antiviral agents such as boceprevir and telaprevir has changed the concept of treatment of adult chronic hepatitis C genotype 1 infected patients. There are three benefits of boceprevir and telaprevir. They increase the likelihood of cure in 1) naive genotype 1 patients and 2) in patients who did not respond to earlier treatment with peg-IFNa and ribavirin, while 3) allowing shortening of treatment duration from 48 weeks to 24 or 28 weeks, which is possible in 40-60% of non-cirrhotic naive (boceprevir and telaprevir) and relapsing patients (telaprevir). The use of boceprevir and telaprevir is associated with multiple side effects and awareness of these side effects is needed to guide the patient through the treatment process. This guideline, formulated on behalf of The Netherlands Association of Hepato-gastroenterologists, The Netherlands Association of Internal Medicine, and The Dutch Association for the Study of Liver Disease, serves as a manual for physicians for the management and treatment of acute and chronic hepatitis C virus monoinfection in adults.
- Published
- 2013
28. A structural role for the PHP domain in E. coli DNA polymerase III.
- Author
-
Barros T, Guenther J, Kelch B, Anaya J, Prabhakar A, O'Donnell M, Kuriyan J, and Lamers MH
- Subjects
- Amino Acid Sequence, Binding Sites, Catalytic Domain, Crystallography, X-Ray, DNA Polymerase III genetics, DNA Polymerase III metabolism, Escherichia coli Proteins genetics, Escherichia coli Proteins metabolism, Metals metabolism, Molecular Sequence Data, Mutation, Protein Stability, Protein Structure, Tertiary, Sequence Alignment, DNA Polymerase III chemistry, Escherichia coli enzymology, Escherichia coli Proteins chemistry
- Abstract
Background: In addition to the core catalytic machinery, bacterial replicative DNA polymerases contain a Polymerase and Histidinol Phosphatase (PHP) domain whose function is not entirely understood. The PHP domains of some bacterial replicases are active metal-dependent nucleases that may play a role in proofreading. In E. coli DNA polymerase III, however, the PHP domain has lost several metal-coordinating residues and is likely to be catalytically inactive., Results: Genomic searches show that the loss of metal-coordinating residues in polymerase PHP domains is likely to have coevolved with the presence of a separate proofreading exonuclease that works with the polymerase. Although the E. coli Pol III PHP domain has lost metal-coordinating residues, the structure of the domain has been conserved to a remarkable degree when compared to that of metal-binding PHP domains. This is demonstrated by our ability to restore metal binding with only three point mutations, as confirmed by the metal-bound crystal structure of this mutant determined at 2.9 Å resolution. We also show that Pol III, a large multi-domain protein, unfolds cooperatively and that mutations in the degenerate metal-binding site of the PHP domain decrease the overall stability of Pol III and reduce its activity., Conclusions: While the presence of a PHP domain in replicative bacterial polymerases is strictly conserved, its ability to coordinate metals and to perform proofreading exonuclease activity is not, suggesting additional non-enzymatic roles for the domain. Our results show that the PHP domain is a major structural element in Pol III and its integrity modulates both the stability and activity of the polymerase.
- Published
- 2013
- Full Text
- View/download PDF
29. Architecture of the Pol III-clamp-exonuclease complex reveals key roles of the exonuclease subunit in processive DNA synthesis and repair.
- Author
-
Toste Rêgo A, Holding AN, Kent H, and Lamers MH
- Subjects
- DNA Polymerase III chemistry, DNA Polymerase III genetics, DNA Polymerase III physiology, DNA Replication genetics, DNA Replication physiology, DNA-Directed DNA Polymerase metabolism, DNA-Directed DNA Polymerase physiology, Escherichia coli genetics, Escherichia coli Proteins genetics, Exodeoxyribonucleases chemistry, Exodeoxyribonucleases genetics, Exodeoxyribonucleases physiology, Models, Biological, Models, Molecular, Multienzyme Complexes metabolism, Multienzyme Complexes physiology, Protein Binding physiology, Protein Structure, Quaternary, Protein Subunits, DNA biosynthesis, DNA Polymerase III metabolism, DNA Polymerase beta metabolism, DNA Repair genetics, DNA-Directed DNA Polymerase chemistry, Escherichia coli enzymology, Escherichia coli Proteins metabolism, Exodeoxyribonucleases metabolism, Multienzyme Complexes chemistry
- Abstract
DNA polymerase III (Pol III) is the catalytic α subunit of the bacterial DNA Polymerase III holoenzyme. To reach maximum activity, Pol III binds to the DNA sliding clamp β and the exonuclease ε that provide processivity and proofreading, respectively. Here, we characterize the architecture of the Pol III-clamp-exonuclease complex by chemical crosslinking combined with mass spectrometry and biochemical methods, providing the first structural view of the trimeric complex. Our analysis reveals that the exonuclease is sandwiched between the polymerase and clamp and enhances the binding between the two proteins by providing a second, indirect, interaction between the polymerase and clamp. In addition, we show that the exonuclease binds the clamp via the canonical binding pocket and thus prevents binding of the translesion DNA polymerase IV to the clamp, providing a novel insight into the mechanism by which the replication machinery can switch between replication, proofreading, and translesion synthesis.
- Published
- 2013
- Full Text
- View/download PDF
30. ESCRT-III binding protein MITD1 is involved in cytokinesis and has an unanticipated PLD fold that binds membranes.
- Author
-
Hadders MA, Agromayor M, Obita T, Perisic O, Caballe A, Kloc M, Lamers MH, Williams RL, and Martin-Serrano J
- Subjects
- Crystallography, X-Ray, HeLa Cells, Humans, Membrane Proteins chemistry, Membrane Proteins metabolism, Microtubule-Associated Proteins chemistry, Microtubule-Associated Proteins metabolism, Models, Molecular, Protein Binding, Protein Folding, Cytokinesis physiology, Endosomal Sorting Complexes Required for Transport metabolism, Membrane Proteins physiology, Microtubule-Associated Proteins physiology, Phospholipase D metabolism
- Abstract
The endosomal sorting complexes required for transport (ESCRT) proteins have a critical function in abscission, the final separation of the daughter cells during cytokinesis. Here, we describe the structure and function of a previously uncharacterized ESCRT-III interacting protein, MIT-domain containing protein 1 (MITD1). Crystal structures of MITD1 reveal a dimer, with a microtubule-interacting and trafficking (MIT) domain at the N terminus and a unique, unanticipated phospholipase D-like (PLD) domain at the C terminus that binds membranes. We show that the MIT domain binds to a subset of ESCRT-III subunits and that this interaction mediates MITD1 recruitment to the midbody during cytokinesis. Depletion of MITD1 causes a distinct cytokinetic phenotype consistent with destabilization of the midbody and abscission failure. These results suggest a model whereby MITD1 coordinates the activity of ESCRT-III during abscission with earlier events in the final stages of cell division.
- Published
- 2012
- Full Text
- View/download PDF
31. Hepatitis C virus infection management in 2012.
- Author
-
Van Gulick JJ, Lamers MH, and Drenth JP
- Subjects
- Antiviral Agents adverse effects, Drug Therapy, Combination, Hepatitis C diagnosis, Hepatitis C epidemiology, Humans, Patient Selection, Practice Guidelines as Topic, Risk Assessment, Risk Factors, Treatment Outcome, Viral Load, Antiviral Agents therapeutic use, Hepatitis C drug therapy
- Abstract
Hepatitis C virus (HCV) is the most common infectious cause of chronic liver disease in Europe. With the introduction of interferon based therapy in combination with ribavirin treatment of chronic HCV has become feasible. This therapy has become the standard of care for patients with HCV and depending on the HCV genotype treatment is successful in 40-70% of patients. In the recent years a new class of drugs have emerged that changed the landscape of HCV treatment. These direct antiviral agents inhibit the NS3/N4A serine protease of HCV. Prototypes are telaprevir and boceprevir and they specifically exert antiviral activity against genotype 1 HCV. A series of landmark trials has paved the way for introduction of these agents, and they have documented a great improvement in the care of genotype 1 HCV patients. Telaprevir and boceprevir are given in combination with pegylated interferon and ribavirin and are useful for treatment naive as well as treatment experienced patients. The clinician should be aware of these developments as they have implications for side effect management, and drug-drug interactions. Finally, strategic use of these agents comes with stopping rules and require close monitoring of the HCV viral load.
- Published
- 2012
32. Interferon-α for patients with chronic hepatitis delta: a systematic review of randomized clinical trials.
- Author
-
Lamers MH, Kirgiz ÖÖ, Heidrich B, Wedemeyer H, and Drenth JP
- Subjects
- Alanine Transaminase blood, Antiviral Agents administration & dosage, Antiviral Agents therapeutic use, Databases, Factual, Dose-Response Relationship, Drug, Drug Therapy, Combination methods, Hepatitis D, Chronic genetics, Hepatitis D, Chronic virology, Hepatitis Delta Virus pathogenicity, Humans, Interferon-alpha administration & dosage, Lamivudine administration & dosage, Lamivudine therapeutic use, RNA, Viral genetics, Randomized Controlled Trials as Topic, Ribavirin administration & dosage, Ribavirin therapeutic use, Risk Factors, Treatment Outcome, Hepatitis D, Chronic drug therapy, Interferon-alpha therapeutic use, RNA, Viral analysis
- Abstract
Background: Hepatitis delta virus (HDV) infection therapy is unclear. This systematic analysis aimed to clarify the evidence on the efficacy of interferon (IFN)-α-based therapy in HDV., Methods: We performed a systematic search on electronic databases including MEDLINE (1970 to January 2011), Web of Science, The Cochrane Library and ClinicalTrials.gov. Randomized clinical trials (RCTs) comparing IFN-α-based therapy with either another drug, placebo or no intervention were included. We excluded paediatric studies. We calculated relative risks (RRs) for comparison of treatment options on the primary outcome measure, which was defined as undetectable levels of HDV RNA and normal alanine aminotransferase at end of treatment (EOT; 1 year)., Results: Nine RCTs were included. Seven trials evaluated the treatment with IFN-α (n=132). The remaining two trials evaluated treatment with pegylated (PEG)-IFN-α (n=45). We found that 1-year treatment with high-dose IFN-α achieved better primary outcome rates than with PEG-IFN-α (RR=4.14, 95% CI 1.00, 17.14). Data for 1-year treatment with low-dose IFN-α compared with PEG-IFN-α were similar (RR=2.83, 95% CI 0.65, 12.40), as were low-dose IFN-α versus high-dose IFN-α (RR=0.68, 95% CI 0.31, 1.50). High-dose IFN-α and PEG-IFN-α reached similar HDV RNA suppression 24 weeks after EOT (RR=1.00, 95% CI 0.51, 1.97). None of the 55 patients assigned to no intervention obtained undetectable levels of HDV RNA and only one patient achieved normalization of alanine aminotransferase level., Conclusions: Based on available RCTs, 1-year high-dose IFN-α monotherapy appears to be more effective than PEG-IFN-α for treatment of HDV patients, with efficacy rates of approximately 30%. There is a lack of head-to-head comparisons. Combination therapies and longer treatment duration need to be investigated.
- Published
- 2012
- Full Text
- View/download PDF
33. Mechanism for activation of the EGF receptor catalytic domain by the juxtamembrane segment.
- Author
-
Jura N, Endres NF, Engel K, Deindl S, Das R, Lamers MH, Wemmer DE, Zhang X, and Kuriyan J
- Subjects
- Amino Acid Sequence, Crystallography, X-Ray, Dimerization, ErbB Receptors metabolism, Models, Molecular, Molecular Sequence Data, Protein Structure, Tertiary, Sequence Alignment, Cell Membrane metabolism, ErbB Receptors chemistry
- Abstract
Signaling by the epidermal growth factor receptor requires an allosteric interaction between the kinase domains of two receptors, whereby one activates the other. We show that the intracellular juxtamembrane segment of the receptor, known to potentiate kinase activity, is able to dimerize the kinase domains. The C-terminal half of the juxtamembrane segment latches the activated kinase domain to the activator, and the N-terminal half of this segment further potentiates dimerization, most likely by forming an antiparallel helical dimer that engages the transmembrane helices of the activated receptor. Our data are consistent with a mechanism in which the extracellular domains block the intrinsic ability of the transmembrane and cytoplasmic domains to dimerize and activate, with ligand binding releasing this block. The formation of the activating juxtamembrane latch is prevented by the C-terminal tails in a structure of an inactive kinase domain dimer, suggesting how alternative dimers can prevent ligand-independent activation.
- Published
- 2009
- Full Text
- View/download PDF
34. Current clinical care compared with new Dutch guidelines for hepatitis C treatment.
- Author
-
Slavenburg S, Lamers MH, Roomer R, de Knegt RJ, van Oijen MG, and Drenth JP
- Subjects
- Antiviral Agents administration & dosage, Genotype, Hepacivirus genetics, Humans, Netherlands, Ribavirin administration & dosage, Societies, Medical, Surveys and Questionnaires, Viral Load, Guideline Adherence statistics & numerical data, Hepatitis C therapy, Practice Guidelines as Topic, Practice Patterns, Physicians' statistics & numerical data
- Abstract
Background: Recently, the Dutch Association of Gastroenterology and Hepatology issued new guidelines for the treatment of chronic hepatitis C virus (HCV). These guidelines reflect the current standard of care. Before these guidelines were published and implemented we (1) studied the current clinical care of HCV patients among Dutch physicians, and (2) identified areas for future refinement in the current treatment., Methods: We conducted a non-targeted survey among Dutch medical specialists in Gastroenterology, Hepatology and Internal Medicine who actively treat HCV patients. The questionnaire contained items about facility, duration and dosing of treatment, and side effect management using clinical vignettes followed by short questions., Results: We received 49 questionnaires from treating HCV specialists. The majority (65%) of respondents treat HCV patients during regular outpatient clinics, while 35% treat these patients in a separate setting dedicated to the care of HCV patients. The majority of physicians follow the stipulated dosage regimens of pegylated interferon (88%) and ribavirin (83%). A minority (13%) exceed the advised dosage of ribavirin. Side effects such as neutropenia are mostly managed by decreasing the interferon dosage (42%). Some 35% of physicians reduce ribavirin if haemoglobin levels drop below 5.4 mmol/l, and 41% initiate erythropoietin treatment., Conclusion: Dutch clinical practice reflects the recently issued HCV guidelines. An important area of refinement in treatment of HCV is the management of side effects.
- Published
- 2009
35. Structure and regulatory mechanism of Aquifex aeolicus NtrC4: variability and evolution in bacterial transcriptional regulation.
- Author
-
Batchelor JD, Doucleff M, Lee CJ, Matsubara K, De Carlo S, Heideker J, Lamers MH, Pelton JG, and Wemmer DE
- Subjects
- Adenosine Triphosphate metabolism, Amino Acid Sequence, Bacterial Proteins ultrastructure, Computational Biology, Crystallography, X-Ray, DNA, Bacterial metabolism, Dimerization, Hydrolysis, Magnetic Resonance Spectroscopy, Molecular Sequence Data, Protein Binding, Protein Structure, Quaternary, Protein Structure, Secondary, Protein Structure, Tertiary, Sequence Alignment, Sequence Homology, Amino Acid, Solutions, Trans-Activators chemistry, Bacteria genetics, Bacterial Proteins chemistry, Bacterial Proteins genetics, Evolution, Molecular, Gene Expression Regulation, Bacterial, Transcription, Genetic
- Abstract
Genetic changes lead gradually to altered protein function, making deduction of the molecular basis for activity from a sequence difficult. Comparative studies provide insights into the functional consequences of specific changes. Here we present structural and biochemical studies of NtrC4, a sigma-54 activator from Aquifex aeolicus, and compare it with NtrC1 (a paralog) and NtrC (a homolog from Salmonella enterica) to provide insight into how a substantial change in regulatory mechanism may have occurred. Activity assays show that assembly of NtrC4's active oligomer is repressed by the N-terminal receiver domain, and that BeF3- addition (mimicking phosphorylation) removes this repression. Observation of assembly without activation for NtrC4 indicates that it is much less strongly repressed than NtrC1. The crystal structure of the unactivated receiver-ATPase domain combination shows a partially disrupted interface. NMR structures of the regulatory domain show that its activation mechanism is very similar to that of NtrC1. The crystal structure of the NtrC4 DNA-binding domain shows that it is dimeric and more similar in structure to NtrC than NtrC1. Electron microscope images of the ATPase-DNA-binding domain combination show formation of oligomeric rings. Sequence alignments provide insights into the distribution of activation mechanisms in this family of proteins.
- Published
- 2008
- Full Text
- View/download PDF
36. A consensus view of DNA binding by the C family of replicative DNA polymerases.
- Author
-
Lamers MH and O'Donnell M
- Subjects
- Amino Acid Motifs physiology, Bacterial Proteins metabolism, Catalytic Domain physiology, Crystallography, X-Ray, DNA-Directed DNA Polymerase metabolism, Genome, Bacterial physiology, Protein Structure, Quaternary physiology, Protein Structure, Tertiary physiology, Structure-Activity Relationship, Bacillaceae enzymology, Bacterial Proteins chemistry, DNA-Directed DNA Polymerase chemistry
- Published
- 2008
- Full Text
- View/download PDF
37. The crystal structure of the catalytic domain of a eukaryotic guanylate cyclase.
- Author
-
Winger JA, Derbyshire ER, Lamers MH, Marletta MA, and Kuriyan J
- Subjects
- Adenylyl Cyclases chemistry, Adenylyl Cyclases genetics, Adenylyl Cyclases metabolism, Animals, Base Sequence, Chlamydomonas reinhardtii chemistry, Crystallography, X-Ray, Dimerization, Enzyme Activation, Guanylate Cyclase genetics, Guanylate Cyclase metabolism, Models, Molecular, Molecular Sequence Data, Protein Conformation, Protein Structure, Tertiary, Sequence Alignment, Catalytic Domain, Guanylate Cyclase chemistry
- Abstract
Background: Soluble guanylate cyclases generate cyclic GMP when bound to nitric oxide, thereby linking nitric oxide levels to the control of processes such as vascular homeostasis and neurotransmission. The guanylate cyclase catalytic module, for which no structure has been determined at present, is a class III nucleotide cyclase domain that is also found in mammalian membrane-bound guanylate and adenylate cyclases., Results: We have determined the crystal structure of the catalytic domain of a soluble guanylate cyclase from the green algae Chlamydomonas reinhardtii at 2.55 A resolution, and show that it is a dimeric molecule., Conclusion: Comparison of the structure of the guanylate cyclase domain with the known structures of adenylate cyclases confirms the close similarity in architecture between these two enzymes, as expected from their sequence similarity. The comparison also suggests that the crystallized guanylate cyclase is in an inactive conformation, and the structure provides indications as to how activation might occur. We demonstrate that the two active sites in the dimer exhibit positive cooperativity, with a Hill coefficient of approximately 1.5. Positive cooperativity has also been observed in the homodimeric mammalian membrane-bound guanylate cyclases. The structure described here provides a reliable model for functional analysis of mammalian guanylate cyclases, which are closely related in sequence.
- Published
- 2008
- Full Text
- View/download PDF
38. Crystal structure of the catalytic alpha subunit of E. coli replicative DNA polymerase III.
- Author
-
Lamers MH, Georgescu RE, Lee SG, O'Donnell M, and Kuriyan J
- Subjects
- Bacterial Proteins chemistry, Binding Sites, Catalytic Domain, Crystallography, X-Ray, DNA chemistry, DNA metabolism, DNA Polymerase I chemistry, DNA Polymerase III metabolism, DNA Polymerase beta metabolism, Escherichia coli Proteins metabolism, Exodeoxyribonucleases chemistry, Models, Molecular, Protein Binding, Protein Folding, Pyrophosphatases chemistry, Structural Homology, Protein, DNA Polymerase III chemistry, DNA Polymerase beta chemistry, Escherichia coli Proteins chemistry
- Abstract
Bacterial replicative DNA polymerases such as Polymerase III (Pol III) share no sequence similarity with other polymerases. The crystal structure, determined at 2.3 A resolution, of a large fragment of Pol III (residues 1-917), reveals a unique chain fold with localized similarity in the catalytic domain to DNA polymerase beta and related nucleotidyltransferases. The structure of Pol III is strikingly different from those of members of the canonical DNA polymerase families, which include eukaryotic replicative polymerases, suggesting that the DNA replication machinery in bacteria arose independently. A structural element near the active site in Pol III that is not present in nucleotidyltransferases but which resembles an element at the active sites of some canonical DNA polymerases suggests that, at a more distant level, all DNA polymerases may share a common ancestor. The structure also suggests a model for interaction of Pol III with the sliding clamp and DNA.
- Published
- 2006
- Full Text
- View/download PDF
39. ATP increases the affinity between MutS ATPase domains. Implications for ATP hydrolysis and conformational changes.
- Author
-
Lamers MH, Georgijevic D, Lebbink JH, Winterwerp HH, Agianian B, de Wind N, and Sixma TK
- Subjects
- Amino Acid Substitution, Crystallography, X-Ray, Escherichia coli Proteins, Models, Molecular, MutS DNA Mismatch-Binding Protein, Mutagenesis, Site-Directed, Protein Conformation, Recombinant Proteins chemistry, Recombinant Proteins metabolism, Adenosine Triphosphatases chemistry, Adenosine Triphosphatases metabolism, Adenosine Triphosphate analogs & derivatives, Adenosine Triphosphate metabolism, Bacterial Proteins chemistry, Bacterial Proteins metabolism, Base Pair Mismatch genetics, DNA-Binding Proteins chemistry, DNA-Binding Proteins metabolism, Escherichia coli enzymology, Escherichia coli genetics
- Abstract
MutS is the key protein of the Escherichia coli DNA mismatch repair system. It recognizes mispaired and unpaired bases and has intrinsic ATPase activity. ATP binding after mismatch recognition by MutS serves as a switch that enables MutL binding and the subsequent initiation of mismatch repair. However, the mechanism of this switch is poorly understood. We have investigated the effects of ATP binding on the MutS structure. Crystallographic studies of ATP-soaked crystals of MutS show a trapped intermediate, with ATP in the nucleotide-binding site. Local rearrangements of several residues around the nucleotide-binding site suggest a movement of the two ATPase domains of the MutS dimer toward each other. Analytical ultracentrifugation experiments confirm such a rearrangement, showing increased affinity between the ATPase domains upon ATP binding and decreased affinity in the presence of ADP. Mutations of specific residues in the nucleotide-binding domain reduce the dimer affinity of the ATPase domains. In addition, ATP-induced release of DNA is strongly reduced in these mutants, suggesting that the two activities are coupled. Hence, it seems plausible that modulation of the affinity between ATPase domains is the driving force for conformational changes in the MutS dimer. These changes are driven by distinct amino acids in the nucleotide-binding site and form the basis for long-range interactions between the ATPase domains and DNA-binding domains and subsequent binding of MutL and initiation of mismatch repair.
- Published
- 2004
- Full Text
- View/download PDF
40. Structures of Escherichia coli DNA mismatch repair enzyme MutS in complex with different mismatches: a common recognition mode for diverse substrates.
- Author
-
Natrajan G, Lamers MH, Enzlin JH, Winterwerp HH, Perrakis A, and Sixma TK
- Subjects
- Adenosine Triphosphatases metabolism, Bacterial Proteins metabolism, Binding Sites, DNA genetics, DNA metabolism, DNA-Binding Proteins metabolism, Escherichia coli enzymology, Escherichia coli Proteins, Glutamic Acid chemistry, Glutamic Acid metabolism, Hydrogen Bonding, Models, Molecular, MutS DNA Mismatch-Binding Protein, Phenylalanine chemistry, Phenylalanine metabolism, Protein Conformation, Protein Structure, Tertiary, Substrate Specificity, Adenosine Triphosphatases chemistry, Bacterial Proteins chemistry, Base Pair Mismatch, DNA chemistry, DNA-Binding Proteins chemistry
- Abstract
We have refined a series of isomorphous crystal structures of the Escherichia coli DNA mismatch repair enzyme MutS in complex with G:T, A:A, C:A and G:G mismatches and also with a single unpaired thymidine. In all these structures, the DNA is kinked by approximately 60 degrees upon protein binding. Two residues widely conserved in the MutS family are involved in mismatch recognition. The phenylalanine, Phe 36, is seen stacking on one of the mismatched bases. The same base is also seen forming a hydrogen bond to the glutamate Glu 38. This hydrogen bond involves the N7 if the base stacking on Phe 36 is a purine and the N3 if it is a pyrimidine (thymine). Thus, MutS uses a common binding mode to recognize a wide range of mismatches.
- Published
- 2003
- Full Text
- View/download PDF
41. The alternating ATPase domains of MutS control DNA mismatch repair.
- Author
-
Lamers MH, Winterwerp HH, and Sixma TK
- Subjects
- Adenosine Triphosphatases chemistry, Adenosine Triphosphatases genetics, Adenosine Triphosphate metabolism, Adenylyl Imidodiphosphate metabolism, Amino Acid Sequence, Animals, Crystallography, X-Ray, DNA Repair, Dimerization, Escherichia coli genetics, Escherichia coli Proteins chemistry, Escherichia coli Proteins genetics, Humans, Models, Molecular, Molecular Sequence Data, MutS DNA Mismatch-Binding Protein, Nucleotides metabolism, Phenotype, Protein Binding, Protein Conformation, Protein Structure, Tertiary, Sequence Alignment, Vanadates metabolism, Adenosine Triphosphatases metabolism, Bacterial Proteins, Base Pair Mismatch, DNA-Binding Proteins, Escherichia coli metabolism, Escherichia coli Proteins metabolism
- Abstract
DNA mismatch repair is an essential safeguard of genomic integrity by removing base mispairings that may arise from DNA polymerase errors or from homologous recombination between DNA strands. In Escherichia coli, the MutS enzyme recognizes mismatches and initiates repair. MutS has an intrinsic ATPase activity crucial for its function, but which is poorly understood. We show here that within the MutS homodimer, the two chemically identical ATPase sites have different affinities for ADP, and the two sites alternate in ATP hydrolysis. A single residue, Arg697, located at the interface of the two ATPase domains, controls the asymmetry. When mutated, the asymmetry is lost and mismatch repair in vivo is impaired. We propose that asymmetry of the ATPase domains is an essential feature of mismatch repair that controls the timing of the different steps in the repair cascade.
- Published
- 2003
- Full Text
- View/download PDF
42. The crystal structure of DNA mismatch repair protein MutS binding to a G x T mismatch.
- Author
-
Lamers MH, Perrakis A, Enzlin JH, Winterwerp HH, de Wind N, and Sixma TK
- Subjects
- Adenosine Triphosphatases metabolism, Bacterial Proteins genetics, Binding Sites, Colorectal Neoplasms, Hereditary Nonpolyposis genetics, Crystallography, X-Ray, DNA, Bacterial metabolism, DNA-Binding Proteins metabolism, Dimerization, Escherichia coli chemistry, Escherichia coli metabolism, Guanine metabolism, Humans, Hydrolysis, Models, Molecular, MutS DNA Mismatch-Binding Protein, MutS Homolog 2 Protein, Mutation, Nucleic Acid Conformation, Protein Conformation, Proto-Oncogene Proteins genetics, Proto-Oncogene Proteins physiology, Thymine metabolism, Bacterial Proteins physiology, Base Pair Mismatch, DNA Repair, DNA, Bacterial chemistry, DNA-Binding Proteins chemistry, Escherichia coli Proteins
- Abstract
DNA mismatch repair ensures genomic integrity on DNA replication. Recognition of a DNA mismatch by a dimeric MutS protein initiates a cascade of reactions and results in repair of the newly synthesized strand; however, details of the molecular mechanism remain controversial. Here we present the crystal structure at 2.2 A of MutS from Escherichia coli bound to a G x T mismatch. The two MutS monomers have different conformations and form a heterodimer at the structural level. Only one monomer recognizes the mismatch specifically and has ADP bound. Mismatch recognition occurs by extensive minor groove interactions causing unusual base pairing and kinking of the DNA. Nonspecific major groove DNA-binding domains from both monomers embrace the DNA in a clamp-like structure. The interleaved nucleotide-binding sites are located far from the DNA. Mutations in human MutS alpha (MSH2/MSH6) that lead to hereditary predisposition for cancer, such as hereditary non-polyposis colorectal cancer, can be mapped to this crystal structure.
- Published
- 2000
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.