Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 343
Filter
1.
J Mol Biol ; 435(24): 168353, 2023 12 15.
Article in English | MEDLINE | ID: mdl-37935254

ABSTRACT

The Y-family DNA polymerases - Pol ι, Pol η, Pol κ and Rev1 - are most well-known for their roles in the DNA damage tolerance pathway of translesion synthesis (TLS). They function to overcome replication barriers by bypassing DNA damage lesions that cannot be normally replicated, allowing replication forks to continue without stalling. In this work, we demonstrate a novel interaction between each Y-family polymerase and the nucleotide excision repair (NER) proteins, RAD23A and RAD23B. We initially focus on the interaction between RAD23A and Pol ι, and through a series of biochemical, cell-based, and structural assays, find that the RAD23A ubiquitin-binding domains (UBA1 and UBA2) interact with separate sites within the Pol ι catalytic domain. While this interaction involves the ubiquitin-binding cleft of UBA2, Pol ι interacts with a distinct surface on UBA1. We further find that mutating or deleting either UBA domain disrupts the RAD23A-Pol ι interaction, demonstrating that both interactions are necessary for stable binding. We also provide evidence that both RAD23 proteins interact with Pol ι in a similar manner, as well as with each of the Y-family polymerases. These results shed light on the interplay between the different functions of the RAD23 proteins and reveal novel binding partners for the Y-family TLS polymerases.


Subject(s)
DNA Repair Enzymes , DNA-Binding Proteins , DNA-Directed DNA Polymerase , DNA Damage , DNA Polymerase iota/chemistry , DNA Repair , DNA Replication , DNA-Directed DNA Polymerase/metabolism , Ubiquitins/chemistry , DNA-Binding Proteins/chemistry , DNA Repair Enzymes/chemistry
2.
Biomol NMR Assign ; 17(2): 287-291, 2023 12.
Article in English | MEDLINE | ID: mdl-37812322

ABSTRACT

hHR23a (human homolog of Rad23 a) functions in nucleotide excision repair and proteasome-mediated protein degradation. It contains an N-terminal ubiquitin-like (UBL) domain, an xeroderma pigmentosum C (XPC)-binding domain, and a ubiquitin-associated (UBA) domain preceding and following the XPC-binding domain. Each of the four structural domains are connected by flexible linker regions. We report in this NMR study, the 1H, 15N and 13C resonance assignments for the backbone and sidechain atoms of the hHR23a full-length protein with BioMagResBank accession number 52059. Assignments are 97% and 87% for the backbone (NH, N, C', Cα, and Hα) and sidechain atoms of the hHR23a structured regions. The secondary structural elements predicted from the NMR data fit well to the hHR23a NMR structure. The assignments described in this manuscript can be used to apply NMR for studies of hHR23a with its binding partners.


Subject(s)
DNA Repair Enzymes , Proteasome Endopeptidase Complex , Humans , DNA Repair Enzymes/chemistry , Protein Structure, Tertiary , DNA-Binding Proteins/chemistry , Nuclear Magnetic Resonance, Biomolecular , Ubiquitin/metabolism
3.
FEBS Lett ; 597(13): 1770-1778, 2023 07.
Article in English | MEDLINE | ID: mdl-36914375

ABSTRACT

Human MutT homolog 1 (MTH1), also known as Nudix-type motif 1 (NUDT1), hydrolyzes 8-oxo-dGTP and 2-oxo-dATP with broad substrate recognition and has attracted attention in anticancer therapeutics. Previous studies on MTH1 have proposed that the exchange of the protonation state between Asp119 and Asp120 is essential for the broad substrate recognition of MTH1. To understand the relationship between protonation states and substrate binding, we determined the crystal structures of MTH1 at pH 7.7-9.7. With increasing pH, MTH1 gradually loses its substrate-binding ability, indicating that Asp119 is deprotonated at pH 8.0-9.1 in 8-oxo-dGTP recognition and Asp120 is deprotonated at pH 8.6-9.7 in 2-oxo-dATP recognition. These results confirm that MTH1 recognizes 8-oxo-dGTP and 2-oxo-dATP by exchanging the protonation state between Asp119 and Asp120 with higher pKa .


Subject(s)
Phosphoric Monoester Hydrolases , Pyrophosphatases , Humans , Pyrophosphatases/chemistry , Pyrophosphatases/metabolism , Phosphoric Monoester Hydrolases/chemistry , DNA Repair Enzymes/chemistry , DNA Repair Enzymes/metabolism , Nudix Hydrolases
4.
Chembiochem ; 24(13): e202200756, 2023 07 03.
Article in English | MEDLINE | ID: mdl-36917742

ABSTRACT

We report a two-step validation approach to evaluate the suitability of metal-binding groups for targeting DNA damage-repair metalloenzymes using model enzyme SNM1A. A fragment-based screening approach was first used to identify metal-binding fragments suitable for targeting the enzyme. Effective fragments were then incorporated into oligonucleotides using the copper-catalysed azide-alkyne cycloaddition reaction. These modified oligonucleotides were recognised by SNM1A at >1000-fold lower concentrations than their fragment counterparts. The exonuclease SNM1A is a key enzyme involved in the repair of interstrand crosslinks, a highly cytotoxic form of DNA damage. However, SNM1A and other enzymes of this class are poorly understood, as there is a lack of tools available to facilitate their study. Our novel approach of incorporating functional fragments into oligonucleotides is broadly applicable to generating modified oligonucleotide structures with high affinity for DNA damage-repair enzymes.


Subject(s)
Cell Cycle Proteins , Exodeoxyribonucleases , Exodeoxyribonucleases/metabolism , DNA Repair Enzymes/chemistry , DNA Repair Enzymes/metabolism , DNA Repair , Oligonucleotides/chemistry
5.
Orphanet J Rare Dis ; 17(1): 121, 2022 03 05.
Article in English | MEDLINE | ID: mdl-35248096

ABSTRACT

BACKGROUND: Cockayne syndrome (CS) is a rare autosomal recessive disorder caused by mutations in ERCC6/CSB or ERCC8/CSA that participate in the transcription-coupled nucleotide excision repair (TC-NER) of UV-induced DNA damage. CS patients display a large heterogeneity of clinical symptoms and severities, the reason of which is not fully understood, and that cannot be anticipated in the diagnostic phase. In addition, little data is available for affected siblings, and this disease is largely undiagnosed in North Africa. METHODS: We report here the clinical description as well as genetic and functional characterization of eight Tunisian CS patients, including siblings. These patients, who belonged to six unrelated families, underwent complete clinical examination and biochemical analyses. Sanger sequencing was performed for the recurrent mutation in five families, and targeted gene sequencing was done for one patient of the sixth family. We also performed Recovery RNA Synthesis (RRS) to confirm the functional impairment of DNA repair in patient-derived fibroblasts. RESULTS: Six out of eight patients carried a homozygous indel mutation (c.598_600delinsAA) in exon 7 of ERCC8, and displayed a variable clinical spectrum including between siblings sharing the same mutation. The other two patients were siblings who carried a homozygous splice-site variant in ERCC8 (c.843+1G>C). This last pair presented more severe clinical manifestations, which are rarely associated with CSA mutations, leading to gastrostomy and hepatic damage. Impaired TC-NER was confirmed by RRS in six tested patients. CONCLUSIONS: This study provides the first deep characterization of case series of CS patients carrying CSA mutations in North Africa. These mutations have been described only in this region and in the Middle-East. We also provide the largest characterization of multiple unrelated patients, as well as siblings, carrying the same mutation, providing a framework for dissecting elusive genotype-phenotype correlations in CS.


Subject(s)
Cockayne Syndrome , Cockayne Syndrome/diagnosis , Cockayne Syndrome/genetics , DNA Repair/genetics , DNA Repair Enzymes/chemistry , DNA Repair Enzymes/genetics , DNA Repair Enzymes/metabolism , Homozygote , Humans , Mutation/genetics , Poly-ADP-Ribose Binding Proteins/genetics , Siblings , Transcription Factors/genetics
6.
ChemMedChem ; 17(5): e202100603, 2022 03 04.
Article in English | MEDLINE | ID: mdl-34905656

ABSTRACT

Certain cancers exhibit upregulation of DNA interstrand crosslink repair pathways, which contributes to resistance to crosslinking chemotherapy drugs and poor prognoses. Inhibition of enzymes implicated in interstrand crosslink repair is therefore a promising strategy for improving the efficacy of cancer treatment. One such target enzyme is SNM1A, a zinc co-ordinating 5'-3' exonuclease. Previous studies have demonstrated the feasibility of inhibiting SNM1A using modified nucleosides appended with zinc-binding groups. In this work, we sought to develop more effective SNM1A inhibitors by exploiting interactions with the phosphate-binding pocket adjacent to the enzyme's active site, in addition to the catalytic zinc ions. A series of nucleoside derivatives bearing phosphate moieties at the 5'-position, as well as zinc-binding groups at the 3'-position, were prepared and tested in gel-electrophoresis and real-time fluorescence assays. As well as investigating novel zinc-binding groups, we found that incorporation of a 5'-phosphate dramatically increased the potency of the inhibitors.


Subject(s)
Exodeoxyribonucleases , Nucleosides , DNA Repair , DNA Repair Enzymes/chemistry , DNA Repair Enzymes/metabolism , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Exodeoxyribonucleases/chemistry , Exodeoxyribonucleases/metabolism , Nucleosides/pharmacology , Phosphates , Phosphorylation , Zinc/pharmacology
7.
Eur J Hum Genet ; 30(1): 101-110, 2022 01.
Article in English | MEDLINE | ID: mdl-34697416

ABSTRACT

Biallelic PNKP variants cause heterogeneous disorders ranging from neurodevelopmental disorder with microcephaly/seizures to adult-onset Charcot-Marie-Tooth disease. To date, only postnatal descriptions exist. We present the first prenatal diagnosis of PNKP-related primary microcephaly. Pathological examination of a male fetus in the 18th gestational week revealed micrencephaly with extracerebral malformations and thus presumed syndromic microcephaly. A recessive disorder was suspected because of previous pregnancy termination for similar abnormalities. Prenatal trio-exome sequencing identified compound heterozygosity for the PNKP variants c.498G>A, p.[(=),0?] and c.302C>T, p.(Pro101Leu). Segregation confirmed both variants in the sister fetus. Through RNA analyses, we characterized exon 4 skipping affecting the PNKP forkhead-associated (FHA) and phosphatase domains (p.Leu67_Lys166del) as the predominant effect of the paternal c.498G>A variant. We retrospectively investigated two unrelated individuals diagnosed with biallelic PNKP-variants to compare prenatal/postnatal phenotypes. Both carry the splice donor variant c.1029+2T>C in trans with a variant in the FHA domain (c.311T>C, p.(Leu104Pro); c.151G>C, p.(Val51Leu)). RNA-seq showed complex splicing for c.1029+2T>C and c.151G>C. Structural modeling revealed significant clustering of missense variants in the FHA domain with variants generating structural damage. Our clinical description extends the PNKP-continuum to the prenatal stage. Investigating possible PNKP-variant effects using RNA and structural modeling, we highlight the mutational complexity and exemplify a PNKP-variant characterization framework.


Subject(s)
DNA Repair Enzymes/genetics , Microcephaly/genetics , Phosphotransferases (Alcohol Group Acceptor)/genetics , Adult , DNA Repair Enzymes/chemistry , Female , Fetus/abnormalities , Humans , Male , Microcephaly/diagnosis , Mutation, Missense , Phenotype , Phosphotransferases (Alcohol Group Acceptor)/chemistry , Prenatal Diagnosis , Protein Domains , RNA Splicing
8.
Nat Commun ; 12(1): 6864, 2021 11 25.
Article in English | MEDLINE | ID: mdl-34824204

ABSTRACT

HIV-1 Vpr is a prototypic member of a large family of structurally related lentiviral virulence factors that antagonize various aspects of innate antiviral immunity. It subverts host cell DNA repair and protein degradation machineries by binding and inhibiting specific post-replication repair enzymes, linking them via the DCAF1 substrate adaptor to the Cullin 4 RING E3 ligase (CRL4DCAF1). HIV-1 Vpr also binds to the multi-domain protein hHR23A, which interacts with the nucleotide excision repair protein XPC and shuttles ubiquitinated proteins to the proteasome. Here, we report the atomic resolution structure of Vpr in complex with the C-terminal half of hHR23A, containing the XPC-binding (XPCB) and ubiquitin-associated (UBA2) domains. The XPCB and UBA2 domains bind to different sides of Vpr's 3-helix-bundle structure, with UBA2 interacting with the α2 and α3 helices of Vpr, while the XPCB domain contacts the opposite side of Vpr's α3 helix. The structure as well as biochemical results reveal that hHR23A and DCAF1 use overlapping binding surfaces on Vpr, even though the two proteins exhibit entirely different three-dimensional structures. Our findings show that Vpr independently targets hHR23A- and DCAF1- dependent pathways and highlight HIV-1 Vpr as a versatile module that interferes with DNA repair and protein degradation pathways.


Subject(s)
DNA Repair Enzymes/chemistry , DNA-Binding Proteins/chemistry , HIV-1/chemistry , vpr Gene Products, Human Immunodeficiency Virus/chemistry , Crystallography, X-Ray , DNA Repair , DNA Repair Enzymes/metabolism , DNA-Binding Proteins/metabolism , HIV-1/metabolism , Host-Pathogen Interactions , Humans , Nuclear Magnetic Resonance, Biomolecular , Protein Binding , Protein Interaction Domains and Motifs , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/metabolism , Protein Structure, Secondary , Ubiquitin-Protein Ligases/chemistry , Ubiquitin-Protein Ligases/metabolism , vpr Gene Products, Human Immunodeficiency Virus/metabolism
9.
Nature ; 598(7880): 368-372, 2021 10.
Article in English | MEDLINE | ID: mdl-34526721

ABSTRACT

Transcription-coupled DNA repair removes bulky DNA lesions from the genome1,2 and protects cells against ultraviolet (UV) irradiation3. Transcription-coupled DNA repair begins when RNA polymerase II (Pol II) stalls at a DNA lesion and recruits the Cockayne syndrome protein CSB, the E3 ubiquitin ligase, CRL4CSA and UV-stimulated scaffold protein A (UVSSA)3. Here we provide five high-resolution structures of Pol II transcription complexes containing human transcription-coupled DNA repair factors and the elongation factors PAF1 complex (PAF) and SPT6. Together with biochemical and published3,4 data, the structures provide a model for transcription-repair coupling. Stalling of Pol II at a DNA lesion triggers replacement of the elongation factor DSIF by CSB, which binds to PAF and moves upstream DNA to SPT6. The resulting elongation complex, ECTCR, uses the CSA-stimulated translocase activity of CSB to pull on upstream DNA and push Pol II forward. If the lesion cannot be bypassed, CRL4CSA spans over the Pol II clamp and ubiquitylates the RPB1 residue K1268, enabling recruitment of TFIIH to UVSSA and DNA repair. Conformational changes in CRL4CSA lead to ubiquitylation of CSB and to release of transcription-coupled DNA repair factors before transcription may continue over repaired DNA.


Subject(s)
Cryoelectron Microscopy , DNA Repair , Multiprotein Complexes/chemistry , Multiprotein Complexes/ultrastructure , RNA Polymerase II/chemistry , RNA Polymerase II/ultrastructure , Transcription, Genetic , Carrier Proteins/chemistry , Carrier Proteins/metabolism , Carrier Proteins/ultrastructure , DNA Helicases/chemistry , DNA Helicases/metabolism , DNA Helicases/ultrastructure , DNA Repair Enzymes/chemistry , DNA Repair Enzymes/metabolism , DNA Repair Enzymes/ultrastructure , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/ultrastructure , Humans , Models, Molecular , Multiprotein Complexes/metabolism , Poly-ADP-Ribose Binding Proteins/chemistry , Poly-ADP-Ribose Binding Proteins/metabolism , Poly-ADP-Ribose Binding Proteins/ultrastructure , RNA Polymerase II/metabolism , Transcription Elongation, Genetic , Transcription Factor TFIIH/chemistry , Transcription Factor TFIIH/metabolism , Transcription Factor TFIIH/ultrastructure , Transcription Factors/chemistry , Transcription Factors/metabolism , Transcription Factors/ultrastructure , Ubiquitin-Protein Ligases/chemistry , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Protein Ligases/ultrastructure , Ubiquitination
10.
Biomolecules ; 11(7)2021 07 10.
Article in English | MEDLINE | ID: mdl-34356632

ABSTRACT

Ubiquitin (Ub) specifically interacts with the Ub-associating domain (UBA) in a proteasomal shuttle factor, while the latter is involved in either proteasomal targeting or self-assembly coacervation. PINK1 phosphorylates Ub at S65 and makes Ub alternate between C-terminally relaxed (pUbRL) and retracted conformations (pUbRT). Using NMR spectroscopy, we show that pUbRL but not pUbRT preferentially interacts with the UBA from two proteasomal shuttle factors Ubqln2 and Rad23A. Yet discriminatorily, Ubqln2-UBA binds to pUb more tightly than Rad23A does and selectively enriches pUbRL upon complex formation. Further, we determine the solution structure of the complex between Ubqln2-UBA and pUbRL and uncover the thermodynamic basis for the stronger interaction. NMR kinetics analysis at different timescales further suggests an indued-fit binding mechanism for pUb-UBA interaction. Notably, at a relatively low saturation level, the dissociation rate of the UBA-pUbRL complex is comparable with the exchange rate between pUbRL and pUbRT. Thus, a kinetic constraint would dictate the interaction between Ub and UBA, thus fine-tuning the functional state of the proteasomal shuttle factors.


Subject(s)
Adaptor Proteins, Signal Transducing/chemistry , Autophagy-Related Proteins/chemistry , DNA Repair Enzymes/chemistry , DNA-Binding Proteins/chemistry , Protein Kinases/chemistry , Ubiquitin/chemistry , Adaptor Proteins, Signal Transducing/metabolism , Autophagy-Related Proteins/metabolism , DNA Repair Enzymes/metabolism , DNA-Binding Proteins/metabolism , Humans , Nuclear Magnetic Resonance, Biomolecular , Protein Binding , Protein Domains , Protein Kinases/metabolism , Thermodynamics , Ubiquitin/metabolism
11.
Protein Sci ; 30(11): 2187-2205, 2021 11.
Article in English | MEDLINE | ID: mdl-34420242

ABSTRACT

The seven xeroderma pigmentosum proteins (XPps), XPA-XPG, coordinate the nucleotide excision repair (NER) pathway, promoting the excision of DNA lesions caused by exposition to ionizing radiation, majorly from ultraviolet light. Significant efforts are made to investigate NER since mutations in any of the seven XPps may cause the xeroderma pigmentosum and trichothiodystrophy diseases. However, these proteins collaborate with other pivotal players in all known NER steps to accurately exert their purposes. Therefore, in the old and ever-evolving field of DNA repair, it is imperative to reexamine and describe their structures to understand NER properly. This work provides an up-to-date review of the protein structural aspects of the closest partners that directly interact and influence XPps: RAD23B, CETN2, DDB1, RPA (RPA70, 32, and 14), p8 (GTF2H5), and ERCC1. Structurally and functionally vital domains, regions, and critical residues are reexamined, providing structural lessons and perspectives about these indispensable proteins in the NER and other DNA repair pathways. By gathering all data related to the major human xeroderma pigmentosum-interacting proteins, this review will aid newcomers on the subject and guide structural and functional future studies.


Subject(s)
DNA Repair Enzymes , DNA Repair , DNA-Binding Proteins , DNA , Mutation , Xeroderma Pigmentosum , DNA/chemistry , DNA/genetics , DNA/metabolism , DNA Repair Enzymes/chemistry , DNA Repair Enzymes/genetics , DNA Repair Enzymes/metabolism , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Humans , Xeroderma Pigmentosum/genetics , Xeroderma Pigmentosum/metabolism
12.
J Biol Chem ; 297(1): 100862, 2021 07.
Article in English | MEDLINE | ID: mdl-34116057

ABSTRACT

The Elongin complex was originally identified as an RNA polymerase II (RNAPII) elongation factor and subsequently as the substrate recognition component of a Cullin-RING E3 ubiquitin ligase. More recent evidence indicates that the Elongin ubiquitin ligase assembles with the Cockayne syndrome B helicase (CSB) in response to DNA damage and can target stalled polymerases for ubiquitylation and removal from the genome. In this report, we present evidence that the CSB-Elongin ubiquitin ligase pathway has roles beyond the DNA damage response in the activation of RNAPII-mediated transcription. We observed that assembly of the CSB-Elongin ubiquitin ligase is induced not just by DNA damage, but also by a variety of signals that activate RNAPII-mediated transcription, including endoplasmic reticulum (ER) stress, amino acid starvation, retinoic acid, glucocorticoids, and doxycycline treatment of cells carrying several copies of a doxycycline-inducible reporter. Using glucocorticoid receptor (GR)-regulated genes as a model, we showed that glucocorticoid-induced transcription is accompanied by rapid recruitment of CSB and the Elongin ubiquitin ligase to target genes in a step that depends upon the presence of transcribing RNAPII on those genes. Consistent with the idea that the CSB-Elongin pathway plays a direct role in GR-regulated transcription, mouse cells lacking the Elongin subunit Elongin A exhibit delays in both RNAPII accumulation on and dismissal from target genes following glucocorticoid addition and withdrawal, respectively. Taken together, our findings bring to light a new role for the CSB-Elongin pathway in RNAPII-mediated transcription.


Subject(s)
DNA Helicases/genetics , DNA Repair Enzymes/genetics , Elongin/genetics , Poly-ADP-Ribose Binding Proteins/genetics , RNA Polymerase II/genetics , Ubiquitin-Protein Ligases/genetics , Animals , Cockayne Syndrome/enzymology , Cockayne Syndrome/genetics , DNA Helicases/chemistry , DNA Helicases/ultrastructure , DNA Repair/genetics , DNA Repair Enzymes/chemistry , DNA Repair Enzymes/ultrastructure , Elongin/chemistry , Elongin/ultrastructure , Humans , Mice , Multiprotein Complexes/chemistry , Multiprotein Complexes/genetics , Multiprotein Complexes/ultrastructure , Poly-ADP-Ribose Binding Proteins/chemistry , Poly-ADP-Ribose Binding Proteins/ultrastructure , RNA Polymerase II/chemistry , Receptors, Glucocorticoid/chemistry , Receptors, Glucocorticoid/genetics , Ubiquitin/chemistry , Ubiquitin/genetics , Ubiquitin-Protein Ligases/chemistry , Ubiquitin-Protein Ligases/ultrastructure , Ubiquitination/genetics
13.
Protein Sci ; 30(9): 1735-1756, 2021 09.
Article in English | MEDLINE | ID: mdl-34056803

ABSTRACT

Evolutionary selection ensures specificity and efficiency in dynamic metastable macromolecular machines that repair DNA damage without releasing toxic and mutagenic intermediates. Here we examine non-homologous end joining (NHEJ) as the primary conserved DNA double-strand break (DSB) repair process in human cells. NHEJ has exemplary key roles in networks determining the development, outcome of cancer treatments by DSB-inducing agents, generation of antibody and T-cell receptor diversity, and innate immune response for RNA viruses. We determine mechanistic insights into NHEJ structural biochemistry focusing upon advanced small angle X-ray scattering (SAXS) results combined with X-ray crystallography (MX) and cryo-electron microscopy (cryo-EM). SAXS coupled to atomic structures enables integrated structural biology for objective quantitative assessment of conformational ensembles and assemblies in solution, intra-molecular distances, structural similarity, functional disorder, conformational switching, and flexibility. Importantly, NHEJ complexes in solution undergo larger allosteric transitions than seen in their cryo-EM or MX structures. In the long-range synaptic complex, X-ray repair cross-complementing 4 (XRCC4) plus XRCC4-like-factor (XLF) form a flexible bridge and linchpin for DNA ends bound to KU heterodimer (Ku70/80) and DNA-PKcs (DNA-dependent protein kinase catalytic subunit). Upon binding two DNA ends, auto-phosphorylation opens DNA-PKcs dimer licensing NHEJ via concerted conformational transformations of XLF-XRCC4, XLF-Ku80, and LigIVBRCT -Ku70 interfaces. Integrated structures reveal multifunctional roles for disordered linkers and modular dynamic interfaces promoting DSB end processing and alignment into the short-range complex for ligation by LigIV. Integrated findings define dynamic assemblies fundamental to designing separation-of-function mutants and allosteric inhibitors targeting conformational transitions in multifunctional complexes.


Subject(s)
DNA Ligase ATP/chemistry , DNA Repair Enzymes/chemistry , DNA, Neoplasm/chemistry , DNA-Activated Protein Kinase/chemistry , DNA-Binding Proteins/chemistry , Ku Autoantigen/chemistry , Neoplasms/genetics , Binding Sites , DNA Breaks, Double-Stranded , DNA Ligase ATP/genetics , DNA Ligase ATP/metabolism , DNA Repair Enzymes/genetics , DNA Repair Enzymes/metabolism , DNA, Neoplasm/genetics , DNA, Neoplasm/metabolism , DNA-Activated Protein Kinase/genetics , DNA-Activated Protein Kinase/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Gene Expression Regulation, Neoplastic , Genomic Instability , Humans , Kinetics , Ku Autoantigen/genetics , Ku Autoantigen/metabolism , Models, Molecular , Neoplasms/metabolism , Neoplasms/pathology , Protein Binding , Protein Conformation , Protein Interaction Domains and Motifs , Substrate Specificity
14.
Cells ; 10(4)2021 04 10.
Article in English | MEDLINE | ID: mdl-33920220

ABSTRACT

Cockayne syndrome (CS) is a DNA repair syndrome characterized by a broad spectrum of clinical manifestations such as neurodegeneration, premature aging, developmental impairment, photosensitivity and other symptoms. Mutations in Cockayne syndrome protein B (CSB) are present in the vast majority of CS patients and in other DNA repair-related pathologies. In the literature, the role of CSB in different DNA repair pathways has been highlighted, however, new CSB functions have been identified in DNA transcription, mitochondrial biology, telomere maintenance and p53 regulation. Herein, we present an overview of identified structural elements and processes that impact on CSB activity and its post-translational modifications, known to balance the different roles of the protein not only during normal conditions but most importantly in stress situations. Moreover, since CSB has been found to be overexpressed in a number of different tumors, its role in cancer is presented and possible therapeutic targeting is discussed.


Subject(s)
Cockayne Syndrome/genetics , DNA Helicases/genetics , DNA Repair Enzymes/genetics , Neoplasms/genetics , Poly-ADP-Ribose Binding Proteins/genetics , Animals , Cockayne Syndrome/metabolism , DNA Damage , DNA Helicases/chemistry , DNA Helicases/metabolism , DNA Repair , DNA Repair Enzymes/chemistry , DNA Repair Enzymes/metabolism , Gene Expression Regulation, Neoplastic , Humans , Models, Molecular , Mutation , Neoplasms/metabolism , Poly-ADP-Ribose Binding Proteins/chemistry , Poly-ADP-Ribose Binding Proteins/metabolism , Protein Conformation , Protein Processing, Post-Translational
15.
Int J Mol Sci ; 22(7)2021 Mar 25.
Article in English | MEDLINE | ID: mdl-33806087

ABSTRACT

Cockayne syndrome group B protein (CSB), a member of the SWI/SNF superfamily, resides in an elongating RNA polymerase II (RNAPII) complex and regulates transcription elongation. CSB contains a C-terminal winged helix domain (WHD) that binds to ubiquitin and plays an important role in DNA repair. However, little is known about the role of the CSB-WHD in transcription regulation. Here, we report that CSB is dependent upon its WHD to regulate RNAPII abundance at promoter proximal pause (PPP) sites of several actively transcribed genes, a key step in the regulation of transcription elongation. We show that two ubiquitin binding-defective mutations in the CSB-WHD, which impair CSB's ability to promote cell survival in response to treatment with cisplatin, have little impact on its ability to stimulate RNAPII occupancy at PPP sites. In addition, we demonstrate that two cancer-associated CSB mutations, which are located on the opposite side of the CSB-WHD away from its ubiquitin-binding pocket, impair CSB's ability to promote RNAPII occupancy at PPP sites. Taken together, these results suggest that CSB promotes RNAPII association with PPP sites in a manner requiring the CSB-WHD but independent of its ubiquitin-binding activity. These results further imply that CSB-mediated RNAPII occupancy at PPP sites is mechanistically separable from CSB-mediated repair of cisplatin-induced DNA damage.


Subject(s)
DNA Helicases/metabolism , DNA Repair Enzymes/metabolism , DNA Repair , Gene Expression Regulation , Mutation , Neoplasms/genetics , Poly-ADP-Ribose Binding Proteins/metabolism , RNA Polymerase II/metabolism , Cell Line, Tumor , Cell Survival , Cisplatin/adverse effects , Cisplatin/pharmacology , DNA Damage/drug effects , DNA Helicases/chemistry , DNA Repair Enzymes/chemistry , Humans , Poly-ADP-Ribose Binding Proteins/chemistry , Promoter Regions, Genetic , Protein Binding , Transcription Factors/metabolism , Ubiquitin/metabolism
16.
Ann Nucl Med ; 35(4): 493-503, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33532992

ABSTRACT

OBJECTIVE: Isocitrate dehydrogenase (IDH) mutation, telomerase reverse transcriptase (TERT) promoter mutation and O6-methylguanine-DNA methyltransferase (MGMT) promoter methylation status are diagnostic, prognostic, predictive and therapeutic biomarkers for primary diffuse gliomas, and this study aimed to explore the relationship between choline (CHO) positron emission tomography (PET) parameters and these molecular alterations. METHODS: Twenty-eight patients who were histopathologically diagnosed with primary diffuse glioma and underwent presurgical CHO PET/CT were retrospectively analyzed, and IDH, TERT and MGMT alterations were examined. The volume of interest (VOI) was semiautomatically defined based on standardized uptake value (SUV) thresholds, and 5 traditional CHO parameters, namely, SUVmax, SUVmean, metabolic tumor volume (MTV), total lesion CHO uptake (TLC) and tumor-to-normal contralateral cortex activity ratio (T/N ratio), were calculated. Wilcoxon rank-sum tests and receiver operating characteristic (ROC) curves were applied to evaluate the differences and performances of the CHO parameters, and their capability to stratify patient prognosis was also evaluated. RESULTS: All 5 parameters were significantly higher in IDH-wildtype gliomas than in IDH-mutant gliomas (p = 0.0001-0.037), and SUVmax, SUVmean, TLC and the T/N ratio exhibited good performances in distinguishing the IDH status (areas under the ROC curve (AUCs) 0.856-0.918, accuracies 0.857-0.893) as well as stratifying patient prognosis. Although the differences and performances of the traditional parameters in distinguishing diverse TERT and MGMT statuses were moderate in the whole population, the T/N ratio and TLC displayed certain predictive value in discriminating the TERT status in the IDH-mutant and IDH-wildtype subgroups (p = 0.028-0.048, AUCs 0.857-0.860, accuracies 0.800-0.917, respectively). CONCLUSIONS: Traditional CHO PET parameters are capable of distinguishing IDH but not TERT or MGMT alterations in the whole population. In accordance with the clinical understanding of TERT promoter mutations, the T/N ratio and TLC can also discriminate the TERT status in IDH subgroups.


Subject(s)
Biomarkers, Tumor/analysis , Choline/analysis , DNA Modification Methylases/chemistry , DNA Repair Enzymes/chemistry , Glioma/diagnostic imaging , Isocitrate Dehydrogenase/chemistry , Telomerase/chemistry , Tumor Suppressor Proteins/chemistry , Adult , Aged , DNA Modification Methylases/genetics , DNA Repair Enzymes/genetics , Female , Humans , Isocitrate Dehydrogenase/genetics , Male , Middle Aged , Mutation , Positron Emission Tomography Computed Tomography , Prognosis , Promoter Regions, Genetic , Retrospective Studies , Telomerase/genetics , Telomerase/metabolism , Tumor Suppressor Proteins/genetics
17.
Nucleic Acids Res ; 49(5): 2418-2434, 2021 03 18.
Article in English | MEDLINE | ID: mdl-33590097

ABSTRACT

Cockayne syndrome (CS) is a segmental premature aging syndrome caused primarily by defects in the CSA or CSB genes. In addition to premature aging, CS patients typically exhibit microcephaly, progressive mental and sensorial retardation and cutaneous photosensitivity. Defects in the CSB gene were initially thought to primarily impair transcription-coupled nucleotide excision repair (TC-NER), predicting a relatively consistent phenotype among CS patients. In contrast, the phenotypes of CS patients are pleiotropic and variable. The latter is consistent with recent work that implicates CSB in multiple cellular systems and pathways, including DNA base excision repair, interstrand cross-link repair, transcription, chromatin remodeling, RNAPII processing, nucleolin regulation, rDNA transcription, redox homeostasis, and mitochondrial function. The discovery of additional functions for CSB could potentially explain the many clinical phenotypes of CSB patients. This review focuses on the diverse roles played by CSB in cellular pathways that enhance genome stability, providing insight into the molecular features of this complex premature aging disease.


Subject(s)
DNA Helicases/physiology , DNA Repair Enzymes/physiology , Poly-ADP-Ribose Binding Proteins/physiology , Chromatin Assembly and Disassembly , DNA Breaks, Double-Stranded , DNA Helicases/chemistry , DNA Repair , DNA Repair Enzymes/chemistry , DNA, Ribosomal/biosynthesis , Gene Expression Regulation , Humans , Mitochondria/genetics , Mitochondria/metabolism , Poly-ADP-Ribose Binding Proteins/chemistry , RNA Polymerase II/metabolism , Transcription, Genetic
18.
Int J Mol Sci ; 22(3)2021 Jan 28.
Article in English | MEDLINE | ID: mdl-33525366

ABSTRACT

MTH1 is an enzyme that hydrolyzes 8-oxo-dGTP, which is an oxidatively damaged nucleobase, into 8-oxo-dGMP in nucleotide pools to prevent its mis-incorporation into genomic DNA. Selective and potent MTH1-binding molecules have potential as biological tools and drug candidates. We recently developed 8-halogenated 7-deaza-dGTP as an 8-oxo-dGTP mimic and found that it was not hydrolyzed, but inhibited enzyme activity. To further increase MTH1 binding, we herein designed and synthesized 7,8-dihalogenated 7-deaza-dG derivatives. We successfully synthesized multiple derivatives, including substituted nucleosides and nucleotides, using 7-deaza-dG as a starting material. Evaluations of the inhibition of MTH1 activity revealed the strong inhibitory effects on enzyme activity of the 7,8-dihalogenated 7-deaza-dG derivatives, particularly 7,8-dibromo 7-daza-dGTP. Based on the results obtained on kinetic parameters and from computational docking simulating studies, these nucleotide analogs interacted with the active site of MTH1 and competitively inhibited the substrate 8-oxodGTP. Therefore, novel properties of repair enzymes in cells may be elucidated using new compounds.


Subject(s)
DNA Repair Enzymes/chemistry , Deoxyguanine Nucleotides/chemistry , Deoxyguanine Nucleotides/chemical synthesis , Drug Design , Phosphoric Monoester Hydrolases/chemistry , Binding Sites , DNA Damage , DNA Repair Enzymes/antagonists & inhibitors , DNA Repair Enzymes/metabolism , Deoxyguanine Nucleotides/metabolism , Deoxyguanine Nucleotides/pharmacology , Enzyme Assays , Halogenation , Humans , Hydrolysis , Kinetics , Molecular Docking Simulation , Molecular Mimicry , Oxidative Stress , Phosphoric Monoester Hydrolases/antagonists & inhibitors , Phosphoric Monoester Hydrolases/metabolism , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Structure-Activity Relationship , Substrate Specificity
19.
Nucleic Acids Res ; 49(2): 1023-1032, 2021 01 25.
Article in English | MEDLINE | ID: mdl-33367848

ABSTRACT

Pseudomonas putida MPE exemplifies a novel clade of manganese-dependent single-strand DNA endonuclease within the binuclear metallophosphoesterase superfamily. MPE is encoded within a widely conserved DNA repair operon. Via structure-guided mutagenesis, we identify His113 and His81 as essential for DNA nuclease activity, albeit inessential for hydrolysis of bis-p-nitrophenylphosphate. We propose that His113 contacts the scissile phosphodiester and serves as a general acid catalyst to expel the OH leaving group of the product strand. We find that MPE cleaves the 3' and 5' single-strands of tailed duplex DNAs and that MPE can sense and incise duplexes at sites of short mismatch bulges and opposite a nick. We show that MPE is an ambidextrous phosphodiesterase capable of hydrolyzing the ssDNA backbone in either orientation to generate a mixture of 3'-OH and 3'-PO4 cleavage products. The directionality of phosphodiester hydrolysis is dictated by the orientation of the water nucleophile vis-à-vis the OH leaving group, which must be near apical for the reaction to proceed. We propose that the MPE active site and metal-bound water nucleophile are invariant and the enzyme can bind the ssDNA productively in opposite orientations.


Subject(s)
Bacterial Proteins/metabolism , DNA Repair Enzymes/metabolism , DNA, Single-Stranded/metabolism , Deoxyribonuclease I/metabolism , Pseudomonas putida/enzymology , Bacterial Proteins/chemistry , Base Pairing , Catalytic Domain , DNA Mismatch Repair , DNA Repair , DNA Repair Enzymes/chemistry , Deoxyribonuclease I/chemistry , Histidine/chemistry , Hydrolysis , Manganese/chemistry , Models, Molecular , Nitrophenols/metabolism , Phosphates/chemistry , Protein Binding , Protein Conformation , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Structure-Activity Relationship , Water
20.
J Proteomics ; 234: 104083, 2021 03 15.
Article in English | MEDLINE | ID: mdl-33373718

ABSTRACT

Using high-throughput BioPlex assays, we determined that six fractions from the venom of Conus nux inhibit the adhesion of various recombinant PfEMP-1 protein domains (PF08_0106 CIDR1α3.1, PF11_0521 DBL2ß3, and PFL0030c DBL3X and DBL5e) to their corresponding receptors (CD36, ICAM-1, and CSA, respectively). The protein domain-receptor interactions permit P. falciparum-infected erythrocytes (IE) to evade elimination in the spleen by adhering to the microvasculature in various organs including the placenta. The sequences for the main components of the fractions, determined by tandem mass spectrometry, yielded four T-superfamily conotoxins, one (CC-Loop-CC) with I-IV, II-III connectivity and three (CC-Loop-CXaaC) with a I-III, II-IV connectivity. The 3D structure for one of the latter, NuxVA = GCCPAPLTCHCVIY, revealed a novel scaffold defined by double turns forming a hairpin-like structure stabilized by the two disulfide bonds. Two other main fraction components were a miniM conotoxin, and a O2-superfamily conotoxin with cysteine framework VI/VII. This study is the first one of its kind suggesting the use of conotoxins for developing pharmacological tools for anti-adhesion adjunct therapy against malaria. Similarly, mitigation of emerging diseases like AIDS and COVID-19, can also benefit from conotoxins as inhibitors of protein-protein interactions as treatment. BIOLOGICAL SIGNIFICANCE: Among the 850+ species of cone snail species there are hundreds of thousands of diverse venom exopeptides that have been selected throughout several million years of evolution to capture prey and deter predators. They do so by targeting several surface proteins present in target excitable cells. This immense biomolecular library of conopeptides can be explored for potential use as therapeutic leads against persistent and emerging diseases affecting non-excitable systems. We aim to expand the pharmacological reach of conotoxins/conopeptides by revealing their in vitro capacity to disrupt protein-protein and protein-polysaccharide interactions that directly contribute to pathology of Plasmodium falciparum malaria. This is significant for severe forms of malaria, which might be deadly even after treated with current parasite-killing drugs because of persistent cytoadhesion of P. falciparum infected erythrocytes even when parasites within red blood cells are dead. Anti-adhesion adjunct drugs would de-sequester or prevent additional sequestration of infected erythrocytes and may significantly improve survival of malaria patients. These results provide a lead for further investigations into conotoxins and other venom peptides as potential candidates for anti-adhesion or blockade-therapies. This study is the first of its kind and it suggests that conotoxins can be developed as pharmacological tools for anti-adhesion adjunct therapy against malaria. Similarly, mitigation of emerging diseases like AIDS and COVID-19, can also benefit from conotoxins as potential inhibitors of protein-protein interactions as treatment.


Subject(s)
CD36 Antigens , DNA Repair Enzymes , Erythrocytes , Intercellular Adhesion Molecule-1 , Mollusk Venoms , Plasmodium falciparum , Transcription Factors , Animals , CD36 Antigens/chemistry , CD36 Antigens/metabolism , COVID-19 , Conus Snail , DNA Repair Enzymes/chemistry , DNA Repair Enzymes/metabolism , Erythrocytes/chemistry , Erythrocytes/metabolism , Erythrocytes/parasitology , Humans , Intercellular Adhesion Molecule-1/chemistry , Intercellular Adhesion Molecule-1/metabolism , Mollusk Venoms/chemistry , Mollusk Venoms/pharmacology , Plasmodium falciparum/chemistry , Plasmodium falciparum/metabolism , Protein Domains , Protozoan Proteins , SARS-CoV-2 , Transcription Factors/chemistry , Transcription Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL