Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
Add more filters










Publication year range
1.
J Biol Chem ; 300(6): 107368, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38750793

ABSTRACT

Activating signal co-integrator complex 1 (ASCC1) acts with ASCC-ALKBH3 complex in alkylation damage responses. ASCC1 uniquely combines two evolutionarily ancient domains: nucleotide-binding K-Homology (KH) (associated with regulating splicing, transcriptional, and translation) and two-histidine phosphodiesterase (PDE; associated with hydrolysis of cyclic nucleotide phosphate bonds). Germline mutations link loss of ASCC1 function to spinal muscular atrophy with congenital bone fractures 2 (SMABF2). Herein analysis of The Cancer Genome Atlas (TCGA) suggests ASCC1 RNA overexpression in certain tumors correlates with poor survival, Signatures 29 and 3 mutations, and genetic instability markers. We determined crystal structures of Alvinella pompejana (Ap) ASCC1 and Human (Hs) PDE domain revealing high-resolution details and features conserved over 500 million years of evolution. Extending our understanding of the KH domain Gly-X-X-Gly sequence motif, we define a novel structural Helix-Clasp-Helix (HCH) nucleotide binding motif and show ASCC1 sequence-specific binding to CGCG-containing RNA. The V-shaped PDE nucleotide binding channel has two His-Φ-Ser/Thr-Φ (HXT) motifs (Φ being hydrophobic) positioned to initiate cyclic phosphate bond hydrolysis. A conserved atypical active-site histidine torsion angle implies a novel PDE substrate. Flexible active site loop and arginine-rich domain linker appear regulatory. Small-angle X-ray scattering (SAXS) revealed aligned KH-PDE RNA binding sites with limited flexibility in solution. Quantitative evolutionary bioinformatic analyses of disease and cancer-associated mutations support implied functional roles for RNA binding, phosphodiesterase activity, and regulation. Collective results inform ASCC1's roles in transactivation and alkylation damage responses, its targeting by structure-based inhibitors, and how ASCC1 mutations may impact inherited disease and cancer.


Subject(s)
Phosphoric Diester Hydrolases , Humans , Phosphoric Diester Hydrolases/metabolism , Phosphoric Diester Hydrolases/chemistry , Phosphoric Diester Hydrolases/genetics , Crystallography, X-Ray , Computational Biology/methods , RNA-Binding Motifs/genetics
2.
Nat Commun ; 14(1): 4445, 2023 07 24.
Article in English | MEDLINE | ID: mdl-37488098

ABSTRACT

RAD51C is an enigmatic predisposition gene for breast, ovarian, and prostate cancer. Currently, missing structural and related functional understanding limits patient mutation interpretation to homology-directed repair (HDR) function analysis. Here we report the RAD51C-XRCC3 (CX3) X-ray co-crystal structure with bound ATP analog and define separable RAD51C replication stability roles informed by its three-dimensional structure, assembly, and unappreciated polymerization motif. Mapping of cancer patient mutations as a functional guide confirms ATP-binding matching RAD51 recombinase, yet highlights distinct CX3 interfaces. Analyses of CRISPR/Cas9-edited human cells with RAD51C mutations combined with single-molecule, single-cell and biophysics measurements uncover discrete CX3 regions for DNA replication fork protection, restart and reversal, accomplished by separable functions in DNA binding and implied 5' RAD51 filament capping. Collective findings establish CX3 as a cancer-relevant replication stress response complex, show how HDR-proficient variants could contribute to tumor development, and identify regions to aid functional testing and classification of cancer mutations.


Subject(s)
Prostatic Neoplasms , Male , Humans , Rad51 Recombinase , Mutation , DNA Replication , Adenosine Triphosphate , DNA-Binding Proteins
3.
Prog Biophys Mol Biol ; 163: 143-159, 2021 08.
Article in English | MEDLINE | ID: mdl-33675849

ABSTRACT

Human uracil DNA-glycosylase (UDG) is the prototypic and first identified DNA glycosylase with a vital role in removing deaminated cytosine and incorporated uracil and 5-fluorouracil (5-FU) from DNA. UDG depletion sensitizes cells to high APOBEC3B deaminase and to pemetrexed (PEM) and floxuridine (5-FdU), which are toxic to tumor cells through incorporation of uracil and 5-FU into DNA. To identify small-molecule UDG inhibitors for pre-clinical evaluation, we optimized biochemical screening of a selected diversity collection of >3,000 small-molecules. We found aurintricarboxylic acid (ATA) as an inhibitor of purified UDG at an initial calculated IC50 < 100 nM. Subsequent enzymatic assays confirmed effective ATA inhibition but with an IC50 of 700 nM and showed direct binding to the human UDG with a KD of <700 nM. ATA displays preferential, dose-dependent binding to purified human UDG compared to human 8-oxoguanine DNA glycosylase. ATA did not bind uracil-containing DNA at these concentrations. Yet, combined crystal structure and in silico docking results unveil ATA interactions with the DNA binding channel and uracil-binding pocket in an open, destabilized UDG conformation. Biologically relevant ATA inhibition of UDG was measured in cell lysates from human DLD1 colon cancer cells and in MCF-7 breast cancer cells using a host cell reactivation assay. Collective findings provide proof-of-principle for development of an ATA-based chemotype and "door stopper" strategy targeting inhibitor binding to a destabilized, open pre-catalytic glycosylase conformation that prevents active site closing for functional DNA binding and nucleotide flipping needed to excise altered bases in DNA.


Subject(s)
DNA Repair , Uracil-DNA Glycosidase , Catalytic Domain , Cytidine Deaminase , DNA Damage , Humans , Minor Histocompatibility Antigens , Uracil , Uracil-DNA Glycosidase/genetics , Uracil-DNA Glycosidase/metabolism
4.
Prog Biophys Mol Biol ; 163: 130-142, 2021 08.
Article in English | MEDLINE | ID: mdl-33115610

ABSTRACT

Cancer will directly affect the lives of over one-third of the population. The DNA Damage Response (DDR) is an intricate system involving damage recognition, cell cycle regulation, DNA repair, and ultimately cell fate determination, playing a central role in cancer etiology and therapy. Two primary therapeutic approaches involving DDR targeting include: combinatorial treatments employing anticancer genotoxic agents; and synthetic lethality, exploiting a sporadic DDR defect as a mechanism for cancer-specific therapy. Whereas, many DDR proteins have proven "undruggable", Fragment- and Structure-Based Drug Discovery (FBDD, SBDD) have advanced therapeutic agent identification and development. FBDD has led to 4 (with ∼50 more drugs under preclinical and clinical development), while SBDD is estimated to have contributed to the development of >200, FDA-approved medicines. Protein X-ray crystallography-based fragment library screening, especially for elusive or "undruggable" targets, allows for simultaneous generation of hits plus details of protein-ligand interactions and binding sites (orthosteric or allosteric) that inform chemical tractability, downstream biology, and intellectual property. Using a novel high-throughput crystallography-based fragment library screening platform, we screened five diverse proteins, yielding hit rates of ∼2-8% and crystal structures from ∼1.8 to 3.2 Å. We consider current FBDD/SBDD methods and some exemplary results of efforts to design inhibitors against the DDR nucleases meiotic recombination 11 (MRE11, a.k.a., MRE11A), apurinic/apyrimidinic endonuclease 1 (APE1, a.k.a., APEX1), and flap endonuclease 1 (FEN1).


Subject(s)
Drug Discovery , Pharmaceutical Preparations , Crystallography, X-Ray , DNA Damage , DNA Repair
5.
Proc Natl Acad Sci U S A ; 117(25): 14127-14138, 2020 06 23.
Article in English | MEDLINE | ID: mdl-32522879

ABSTRACT

Xeroderma pigmentosum group G (XPG) protein is both a functional partner in multiple DNA damage responses (DDR) and a pathway coordinator and structure-specific endonuclease in nucleotide excision repair (NER). Different mutations in the XPG gene ERCC5 lead to either of two distinct human diseases: Cancer-prone xeroderma pigmentosum (XP-G) or the fatal neurodevelopmental disorder Cockayne syndrome (XP-G/CS). To address the enigmatic structural mechanism for these differing disease phenotypes and for XPG's role in multiple DDRs, here we determined the crystal structure of human XPG catalytic domain (XPGcat), revealing XPG-specific features for its activities and regulation. Furthermore, XPG DNA binding elements conserved with FEN1 superfamily members enable insights on DNA interactions. Notably, all but one of the known pathogenic point mutations map to XPGcat, and both XP-G and XP-G/CS mutations destabilize XPG and reduce its cellular protein levels. Mapping the distinct mutation classes provides structure-based predictions for disease phenotypes: Residues mutated in XP-G are positioned to reduce local stability and NER activity, whereas residues mutated in XP-G/CS have implied long-range structural defects that would likely disrupt stability of the whole protein, and thus interfere with its functional interactions. Combined data from crystallography, biochemistry, small angle X-ray scattering, and electron microscopy unveil an XPG homodimer that binds, unstacks, and sculpts duplex DNA at internal unpaired regions (bubbles) into strongly bent structures, and suggest how XPG complexes may bind both NER bubble junctions and replication forks. Collective results support XPG scaffolding and DNA sculpting functions in multiple DDR processes to maintain genome stability.


Subject(s)
Cockayne Syndrome/genetics , DNA-Binding Proteins/chemistry , Endonucleases/chemistry , Nuclear Proteins/chemistry , Point Mutation , Transcription Factors/chemistry , Xeroderma Pigmentosum/genetics , Binding Sites , Conserved Sequence , DNA/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Endonucleases/genetics , Endonucleases/metabolism , Enzyme Stability , Humans , Molecular Dynamics Simulation , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Phenotype , Protein Binding , Protein Folding , Protein Multimerization , Transcription Factors/genetics , Transcription Factors/metabolism
6.
Methods Enzymol ; 601: 205-241, 2018.
Article in English | MEDLINE | ID: mdl-29523233

ABSTRACT

For inhibitor design, as in most research, the best system is question dependent. We suggest structurally defined allostery to design specific inhibitors that target regions beyond active sites. We choose systems allowing efficient quality structures with conformational changes as optimal for structure-based design to optimize inhibitors. We maintain that evolutionarily related targets logically provide molecular avatars, where this Sanskrit term for descent includes ideas of functional relationships and of being a physical embodiment of the target's essential features without requiring high sequence identity. Appropriate biochemical and cell assays provide quantitative measurements, and for biomedical impacts, any inhibitor's activity should be validated in human cells. Specificity is effectively shown empirically by testing if mutations blocking target activity remove cellular inhibitor impact. We propose this approach to be superior to experiments testing for lack of cross-reactivity among possible related enzymes, which is a challenging negative experiment. As an exemplary avatar system for protein and DNA allosteric conformational controls, we focus here on developing separation-of-function inhibitors for meiotic recombination 11 nuclease activities. This was achieved not by targeting the active site but rather by geometrically impacting loop motifs analogously to ribosome antibiotics. These loops are neighboring the dimer interface and active site act in sculpting dsDNA and ssDNA into catalytically competent complexes. One of our design constraints is to preserve DNA substrate binding to geometrically block competing enzymes and pathways from the damaged site. We validate our allosteric approach to controlling outcomes in human cells by reversing the radiation sensitivity and genomic instability in BRCA mutant cells.


Subject(s)
Drug Design , MRE11 Homologue Protein/antagonists & inhibitors , Allosteric Regulation , Amino Acid Sequence , Endonucleases/antagonists & inhibitors , Endonucleases/metabolism , Evolution, Molecular , Exonucleases/antagonists & inhibitors , Exonucleases/metabolism , Humans , MRE11 Homologue Protein/genetics , MRE11 Homologue Protein/metabolism , Protein Conformation , Sensitivity and Specificity , Sequence Alignment , Sequence Analysis, Protein
7.
Nat Commun ; 8: 15855, 2017 06 27.
Article in English | MEDLINE | ID: mdl-28653660

ABSTRACT

DNA replication and repair enzyme Flap Endonuclease 1 (FEN1) is vital for genome integrity, and FEN1 mutations arise in multiple cancers. FEN1 precisely cleaves single-stranded (ss) 5'-flaps one nucleotide into duplex (ds) DNA. Yet, how FEN1 selects for but does not incise the ss 5'-flap was enigmatic. Here we combine crystallographic, biochemical and genetic analyses to show that two dsDNA binding sites set the 5'polarity and to reveal unexpected control of the DNA phosphodiester backbone by electrostatic interactions. Via 'phosphate steering', basic residues energetically steer an inverted ss 5'-flap through a gateway over FEN1's active site and shift dsDNA for catalysis. Mutations of these residues cause an 18,000-fold reduction in catalytic rate in vitro and large-scale trinucleotide (GAA)n repeat expansions in vivo, implying failed phosphate-steering promotes an unanticipated lagging-strand template-switch mechanism during replication. Thus, phosphate steering is an unappreciated FEN1 function that enforces 5'-flap specificity and catalysis, preventing genomic instability.


Subject(s)
DNA/genetics , Flap Endonucleases/metabolism , Genomic Instability , Phosphates/metabolism , Amino Acid Sequence , Binding Sites , Catalytic Domain , DNA/chemistry , DNA/metabolism , DNA Repair , DNA Replication , Flap Endonucleases/chemistry , Flap Endonucleases/genetics , Humans , Mutation , Phosphates/chemistry , Sequence Alignment , Substrate Specificity
9.
Mol Microbiol ; 99(4): 674-85, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26508112

ABSTRACT

The motor of the membrane-anchored archaeal motility structure, the archaellum, contains FlaX, FlaI and FlaH. FlaX forms a 30 nm ring structure that acts as a scaffold protein and was shown to interact with the bifunctional ATPase FlaI and FlaH. However, the structure and function of FlaH has been enigmatic. Here we present structural and functional analyses of isolated FlaH and archaellum motor subcomplexes. The FlaH crystal structure reveals a RecA/Rad51 family fold with an ATP bound on a conserved and exposed surface, which presumably forms an oligomerization interface. FlaH does not hydrolyze ATP in vitro, but ATP binding to FlaH is essential for its interaction with FlaI and for archaellum assembly. FlaH interacts with the C-terminus of FlaX, which was earlier shown to be essential for FlaX ring formation and to mediate interaction with FlaI. Electron microscopy reveals that FlaH assembles as a second ring inside the FlaX ring in vitro. Collectively these data reveal central structural mechanisms for FlaH interactions in mediating archaellar assembly: FlaH binding within the FlaX ring and nucleotide-regulated FlaH binding to FlaI form the archaellar basal body core.


Subject(s)
Archaeal Proteins/chemistry , Archaeal Proteins/metabolism , Flagella/physiology , Nucleotides/metabolism , Sulfolobus acidocaldarius/metabolism , Adenosine Triphosphatases/metabolism , Adenosine Triphosphate/metabolism , Amino Acid Motifs , Amino Acid Sequence , Archaeal Proteins/physiology , Crystallization , Crystallography, X-Ray , Flagellin/metabolism , Genes, Archaeal , Microscopy, Electron , Models, Molecular , Movement , Sulfolobus acidocaldarius/genetics
10.
Structure ; 23(5): 863-872, 2015 May 05.
Article in English | MEDLINE | ID: mdl-25865246

ABSTRACT

Archaea employ the archaellum, a type IV pilus-like nanomachine, for swimming motility. In the crenarchaeon Sulfolobus acidocaldarius, the archaellum consists of seven proteins: FlaB/X/G/F/H/I/J. FlaF is conserved and essential for archaellum assembly but no FlaF structures exist. Here, we truncated the FlaF N terminus and solved 1.5-Å and 1.65-Å resolution crystal structures of this monotopic membrane protein. Structures revealed an N-terminal α-helix and an eight-strand ß-sandwich, immunoglobulin-like fold with striking similarity to S-layer proteins. Crystal structures, X-ray scattering, and mutational analyses suggest dimer assembly is needed for in vivo function. The sole cell envelope component of S. acidocaldarius is a paracrystalline S-layer, and FlaF specifically bound to S-layer protein, suggesting that its interaction domain is located in the pseudoperiplasm with its N-terminal helix in the membrane. From these data, FlaF may act as the previously unknown archaellum stator protein that anchors the rotating archaellum to the archaeal cell envelope.


Subject(s)
Archaeal Proteins/chemistry , Archaeal Proteins/metabolism , Membrane Glycoproteins/metabolism , Sulfolobus acidocaldarius/metabolism , Amino Acid Sequence , Archaeal Proteins/genetics , Binding Sites , Cell Membrane/chemistry , Cell Membrane/metabolism , Conserved Sequence , Crystallography, X-Ray , Dimerization , Models, Molecular , Mutation , Protein Structure, Secondary , Sulfolobus acidocaldarius/chemistry , Sulfolobus acidocaldarius/genetics
11.
Proc Natl Acad Sci U S A ; 111(23): 8446-51, 2014 Jun 10.
Article in English | MEDLINE | ID: mdl-24912171

ABSTRACT

For decades, the enzymatic conversion of cellulose was thought to rely on the synergistic action of hydrolytic enzymes, but recent work has shown that lytic polysaccharide monooxygenases (LPMOs) are important contributors to this process. We describe the structural and functional characterization of two functionally coupled cellulose-active LPMOs belonging to auxiliary activity family 10 (AA10) that commonly occur in cellulolytic bacteria. One of these LPMOs cleaves glycosidic bonds by oxidation of the C1 carbon, whereas the other can oxidize both C1 and C4. We thus demonstrate that C4 oxidation is not confined to fungal AA9-type LPMOs. X-ray crystallographic structures were obtained for the enzyme pair from Streptomyces coelicolor, solved at 1.3 Å (ScLPMO10B) and 1.5 Å (CelS2 or ScLPMO10C) resolution. Structural comparisons revealed differences in active site architecture that could relate to the ability to oxidize C4 (and that also seem to apply to AA9-type LPMOs). Despite variation in active site architecture, the two enzymes exhibited similar affinities for Cu(2+) (12-31 nM), redox potentials (242 and 251 mV), and electron paramagnetic resonance spectra, with only the latter clearly different from those of chitin-active AA10-type LPMOs. We conclude that substrate specificity depends not on copper site architecture, but rather on variation in substrate binding and orientation. During cellulose degradation, the members of this LPMO pair act in synergy, indicating different functional roles and providing a rationale for the abundance of these enzymes in biomass-degrading organisms.


Subject(s)
Bacterial Proteins/metabolism , Cellulose/metabolism , Mixed Function Oxygenases/metabolism , Polysaccharides/metabolism , Amino Acid Sequence , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Catalytic Domain , Chitin/metabolism , Copper/metabolism , Crystallography, X-Ray , Electron Spin Resonance Spectroscopy , Mixed Function Oxygenases/chemistry , Mixed Function Oxygenases/genetics , Models, Molecular , Molecular Sequence Data , Oxidation-Reduction , Protein Binding , Protein Structure, Secondary , Protein Structure, Tertiary , Sequence Homology, Amino Acid , Substrate Specificity , Zinc/metabolism
12.
EMBO Rep ; 15(5): 601-8, 2014 May.
Article in English | MEDLINE | ID: mdl-24714598

ABSTRACT

The post-translational modification of DNA repair and checkpoint proteins by ubiquitin and small ubiquitin-like modifier (SUMO) critically orchestrates the DNA damage response (DDR). The ubiquitin ligase RNF4 integrates signaling by SUMO and ubiquitin, through its selective recognition and ubiquitination of SUMO-modified proteins. Here, we define a key new determinant for target discrimination by RNF4, in addition to interaction with SUMO. We identify a nucleosome-targeting motif within the RNF4 RING domain that can bind DNA and thereby enables RNF4 to selectively ubiquitinate nucleosomal histones. Furthermore, RNF4 nucleosome-targeting is crucially required for the repair of TRF2-depleted dysfunctional telomeres by 53BP1-mediated non-homologous end joining.


Subject(s)
DNA Repair , Nuclear Proteins/metabolism , Nuclear Proteins/ultrastructure , Nucleosomes/metabolism , Small Ubiquitin-Related Modifier Proteins/metabolism , Transcription Factors/metabolism , Transcription Factors/ultrastructure , Amino Acid Motifs , Animals , Cell Line , Chromosomal Proteins, Non-Histone/metabolism , Crystallography, X-Ray , DNA Damage , DNA-Binding Proteins/metabolism , Gene Knockout Techniques , Mice , Nuclear Proteins/genetics , Protein Processing, Post-Translational , Protein Structure, Tertiary , Tamoxifen/analogs & derivatives , Tamoxifen/pharmacology , Telomere/drug effects , Telomere/genetics , Telomeric Repeat Binding Protein 2/genetics , Transcription Factors/genetics , Tumor Suppressor p53-Binding Protein 1 , Ubiquitin/metabolism , Ubiquitin-Protein Ligases , Ubiquitination
13.
Mol Cell ; 53(1): 7-18, 2014 Jan 09.
Article in English | MEDLINE | ID: mdl-24316220

ABSTRACT

MRE11 within the MRE11-RAD50-NBS1 (MRN) complex acts in DNA double-strand break repair (DSBR), detection, and signaling; yet, how its endo- and exonuclease activities regulate DSBR by nonhomologous end-joining (NHEJ) versus homologous recombination (HR) remains enigmatic. Here, we employed structure-based design with a focused chemical library to discover specific MRE11 endo- or exonuclease inhibitors. With these inhibitors, we examined repair pathway choice at DSBs generated in G2 following radiation exposure. While nuclease inhibition impairs radiation-induced replication protein A (RPA) chromatin binding, suggesting diminished resection, the inhibitors surprisingly direct different repair outcomes. Endonuclease inhibition promotes NHEJ in lieu of HR, while exonuclease inhibition confers a repair defect. Collectively, the results describe nuclease-specific MRE11 inhibitors, define distinct nuclease roles in DSB repair, and support a mechanism whereby MRE11 endonuclease initiates resection, thereby licensing HR followed by MRE11 exonuclease and EXO1/BLM bidirectional resection toward and away from the DNA end, which commits to HR.


Subject(s)
DNA Breaks, Double-Stranded , DNA End-Joining Repair , DNA-Binding Proteins/metabolism , Enzyme Inhibitors/chemistry , G2 Phase , Recombinational DNA Repair , Cell Line , Chromatin/genetics , Chromatin/metabolism , DNA Repair Enzymes/genetics , DNA Repair Enzymes/metabolism , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/genetics , Exodeoxyribonucleases/genetics , Exodeoxyribonucleases/metabolism , Gamma Rays/adverse effects , Humans , MRE11 Homologue Protein , Replication Protein A/genetics , Replication Protein A/metabolism
14.
J Biol Chem ; 288(12): 8445-8455, 2013 Mar 22.
Article in English | MEDLINE | ID: mdl-23355472

ABSTRACT

Non-coding apurinic/apyrimidinic (AP) sites in DNA form spontaneously and as DNA base excision repair intermediates are the most common toxic and mutagenic in vivo DNA lesion. For repair, AP sites must be processed by 5' AP endonucleases in initial stages of base repair. Human APE1 and bacterial Nfo represent the two conserved 5' AP endonuclease families in the biosphere; they both recognize AP sites and incise the phosphodiester backbone 5' to the lesion, yet they lack similar structures and metal ion requirements. Here, we determined and analyzed crystal structures of a 2.4 Å resolution APE1-DNA product complex with Mg(2+) and a 0.92 Å Nfo with three metal ions. Structural and biochemical comparisons of these two evolutionarily distinct enzymes characterize key APE1 catalytic residues that are potentially functionally similar to Nfo active site components, as further tested and supported by computational analyses. We observe a magnesium-water cluster in the APE1 active site, with only Glu-96 forming the direct protein coordination to the Mg(2+). Despite differences in structure and metal requirements of APE1 and Nfo, comparison of their active site structures surprisingly reveals strong geometric conservation of the catalytic reaction, with APE1 catalytic side chains positioned analogously to Nfo metal positions, suggesting surprising functional equivalence between Nfo metal ions and APE1 residues. The finding that APE1 residues are positioned to substitute for Nfo metal ions is supported by the impact of mutations on activity. Collectively, the results illuminate the activities of residues, metal ions, and active site features for abasic site endonucleases.


Subject(s)
Bacterial Proteins/chemistry , DNA-(Apurinic or Apyrimidinic Site) Lyase/chemistry , Deoxyribonuclease IV (Phage T4-Induced)/chemistry , Thermotoga maritima/enzymology , Amino Acid Sequence , Amino Acid Substitution , Catalytic Domain , Conserved Sequence , Crystallography, X-Ray , DNA/chemistry , DNA-(Apurinic or Apyrimidinic Site) Lyase/genetics , Escherichia coli , Humans , Molecular Dynamics Simulation , Mutagenesis, Site-Directed , Nucleic Acid Conformation , Protein Binding , Protein Structure, Secondary , Structural Homology, Protein
15.
J Biol Chem ; 287(43): 36322-30, 2012 Oct 19.
Article in English | MEDLINE | ID: mdl-22952223

ABSTRACT

Degradation of recalcitrant polysaccharides in nature is typically accomplished by mixtures of processive and nonprocessive glycoside hydrolases (GHs), which exhibit synergistic activity wherein nonprocessive enzymes provide new sites for productive attachment of processive enzymes. GH processivity is typically attributed to active site geometry, but previous work has demonstrated that processivity can be tuned by point mutations or removal of single loops. To gain additional insights into the differences between processive and nonprocessive enzymes that give rise to their synergistic activities, this study reports the crystal structure of the catalytic domain of the GH family 18 nonprocessive endochitinase, ChiC, from Serratia marcescens. This completes the structural characterization of the co-evolved chitinolytic enzymes from this bacterium and enables structural analysis of their complementary functions. The ChiC catalytic module reveals a shallow substrate-binding cleft that lacks aromatic residues vital for processivity, a calcium-binding site not previously seen in GH18 chitinases, and, importantly, a displaced catalytic acid (Glu-141), suggesting flexibility in the catalytic center. Molecular dynamics simulations of two processive chitinases (ChiA and ChiB), the ChiC catalytic module, and an endochitinase from Lactococcus lactis show that the nonprocessive enzymes have more flexible catalytic machineries and that their bound ligands are more solvated and flexible. These three features, which relate to the more dynamic on-off ligand binding processes associated with nonprocessive action, correlate to experimentally measured differences in processivity of the S. marcescens chitinases. These newly defined hallmarks thus appear to be key dynamic metrics in determining processivity in GH enzymes complementing structural insights.


Subject(s)
Bacterial Proteins/chemistry , Glycoside Hydrolases/chemistry , Models, Chemical , Molecular Dynamics Simulation , Serratia marcescens/enzymology , Catalytic Domain , Crystallography, X-Ray , Structure-Activity Relationship
16.
Mol Cell ; 47(1): 50-60, 2012 Jul 13.
Article in English | MEDLINE | ID: mdl-22658721

ABSTRACT

Nucleotide excision repair (NER) has long been known to remove DNA lesions induced by chemical carcinogens, and the molecular mechanism has been partially elucidated. Here we demonstrate that in Schizosaccharomyces pombe a DNA recognition protein, alkyltransferase-like 1 (Atl1), can play a pivotal role in selecting a specific NER pathway, depending on the nature of the DNA modification. The relative ease of dissociation of Atl1 from DNA containing small O(6)-alkylguanines allows accurate completion of global genome repair (GGR), whereas strong Atl1 binding to bulky O(6)-alkylguanines blocks GGR, stalls the transcription machinery, and diverts the damage to transcription-coupled repair. Our findings redraw the initial stages of the NER process in those organisms that express an alkyltransferase-like gene and raise the question of whether or not O(6)-alkylguanine lesions that are poor substrates for the alkyltransferase proteins in higher eukaryotes might, by analogy, signal such lesions for repair by NER.


Subject(s)
Alkyl and Aryl Transferases/metabolism , DNA Repair , Guanine/analogs & derivatives , Schizosaccharomyces pombe Proteins/metabolism , Alkyl and Aryl Transferases/chemistry , Alkyl and Aryl Transferases/genetics , Blotting, Western , Crystallography, X-Ray , DNA Damage , DNA, Fungal/chemistry , DNA, Fungal/genetics , DNA, Fungal/metabolism , Flow Cytometry , G1 Phase/drug effects , Genome, Fungal/genetics , Guanine/chemistry , Guanine/metabolism , Methylnitronitrosoguanidine/toxicity , Models, Molecular , Mutation , Nitrosourea Compounds/toxicity , Nucleic Acid Conformation , Protein Binding , Protein Structure, Tertiary , Schizosaccharomyces/drug effects , Schizosaccharomyces/genetics , Schizosaccharomyces/metabolism , Schizosaccharomyces pombe Proteins/chemistry , Schizosaccharomyces pombe Proteins/genetics , Transcription, Genetic/genetics
17.
J Biol Chem ; 287(26): 22295-304, 2012 Jun 22.
Article in English | MEDLINE | ID: mdl-22573334

ABSTRACT

Fluorescent proteins derived from light, oxygen, or voltage (LOV) domains offer advantages over green fluorescent protein (GFP) from their small size and efficacy under anaerobic conditions. The flavoprotein improved LOV (iLOV) was engineered from the blue light receptor phototropin as a reporter of viral infection. To inform the molecular basis for the improved, photoreversible, fluorescent properties of iLOV, we employed directed evolution and determined five LOV crystallographic structures. Comparative structural analyses between iLOV and its progenitors reveal mutation-induced constraints in the environment of the flavin mononucleotide (FMN) chromophore; in iLOV, the methyl group of Thr-394 "crowds" the FMN isoalloxazine ring, Leu-470 triggers side chain "flipping" of Leu-472, and the terminal FMN phosphate shows increased anchoring. We further engineered iLOV variants that are readily detectable in bacterial and mammalian cells due to order-of-magnitude photostability increases. Structure determination of a resulting representative photostable iLOV (phiLOV) variant reveals additional constraints on the chromophore. Aromatic residues Tyr-401 and Phe-485 in phiLOV sandwich the FMN isoalloxazine ring from both sides, whereas Ser-390 anchors the side chain of FMN-interacting Gln-489 Our combined structural and mutational results reveal that constraining the FMN fluorophore yields improved photochemical properties for iLOV and its new photostable derivative. These findings provide a framework for structural fine-tuning of LOV scaffold proteins to maximize their potential as oxygen-independent fluorescent reporters.


Subject(s)
Flavoproteins/chemistry , Luminescent Proteins/chemistry , Photochemistry/methods , Animals , Arabidopsis/metabolism , Cell Line , Crystallography, X-Ray/methods , Flavoproteins/metabolism , Fluorescence , Genes, Reporter , Haplorhini , Light , Models, Molecular , Mutagenesis , Oxygen/chemistry , Phototropins/chemistry , Protein Conformation , Spectrophotometry/methods
18.
Science ; 335(6075): 1492-6, 2012 Mar 23.
Article in English | MEDLINE | ID: mdl-22323738

ABSTRACT

The recently identified plant photoreceptor UVR8 (UV RESISTANCE LOCUS 8) triggers regulatory changes in gene expression in response to ultraviolet-B (UV-B) light through an unknown mechanism. Here, crystallographic and solution structures of the UVR8 homodimer, together with mutagenesis and far-UV circular dichroism spectroscopy, reveal its mechanisms for UV-B perception and signal transduction. ß-propeller subunits form a remarkable, tryptophan-dominated, dimer interface stitched together by a complex salt-bridge network. Salt-bridging arginines flank the excitonically coupled cross-dimer tryptophan "pyramid" responsible for UV-B sensing. Photoreception reversibly disrupts salt bridges, triggering dimer dissociation and signal initiation. Mutation of a single tryptophan to phenylalanine retunes the photoreceptor to detect UV-C wavelengths. Our analyses establish how UVR8 functions as a photoreceptor without a prosthetic chromophore to promote plant development and survival in sunlight.


Subject(s)
Arabidopsis Proteins/chemistry , Arabidopsis Proteins/metabolism , Chromosomal Proteins, Non-Histone/chemistry , Chromosomal Proteins, Non-Histone/metabolism , Photoreceptors, Plant/chemistry , Photoreceptors, Plant/metabolism , Ultraviolet Rays , Arabidopsis/physiology , Arabidopsis Proteins/genetics , Arginine/chemistry , Chromosomal Proteins, Non-Histone/genetics , Circular Dichroism , Crystallography, X-Ray , Hydrogen Bonding , Hydrogen-Ion Concentration , Light Signal Transduction , Models, Molecular , Mutagenesis , Photoreceptors, Plant/genetics , Protein Conformation , Protein Multimerization , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Tryptophan/chemistry
19.
J Biol Chem ; 287(15): 12060-9, 2012 Apr 06.
Article in English | MEDLINE | ID: mdl-22170053

ABSTRACT

Ozone depletion increases terrestrial solar ultraviolet B (UV-B; 280-315 nm) radiation, intensifying the risks plants face from DNA damage, especially covalent cyclobutane pyrimidine dimers (CPD). Without efficient repair, UV-B destroys genetic integrity, but plant breeding creates rice cultivars with more robust photolyase (PHR) DNA repair activity as an environmental adaptation. So improved strains of Oryza sativa (rice), the staple food for Asia, have expanded rice cultivation worldwide. Efficient light-driven PHR enzymes restore normal pyrimidines to UV-damaged DNA by using blue light via flavin adenine dinucleotide to break pyrimidine dimers. Eukaryotes duplicated the photolyase gene, producing PHRs that gained functions and adopted activities that are distinct from those of prokaryotic PHRs yet are incompletely understood. Many multicellular organisms have two types of PHR: (6-4) PHR, which structurally resembles bacterial CPD PHRs but recognizes different substrates, and Class II CPD PHR, which is remarkably dissimilar in sequence from bacterial PHRs despite their common substrate. To understand the enigmatic DNA repair mechanisms of PHRs in eukaryotic cells, we determined the first crystal structure of a eukaryotic Class II CPD PHR from the rice cultivar Sasanishiki. Our 1.7 Å resolution PHR structure reveals structure-activity relationships in Class II PHRs and tuning for enhanced UV tolerance in plants. Structural comparisons with prokaryotic Class I CPD PHRs identified differences in the binding site for UV-damaged DNA substrate. Convergent evolution of both flavin hydrogen bonding and a Trp electron transfer pathway establish these as critical functional features for PHRs. These results provide a paradigm for light-dependent DNA repair in higher organisms.


Subject(s)
Deoxyribodipyrimidine Photo-Lyase/chemistry , Oryza/enzymology , Plant Proteins/chemistry , Amino Acid Motifs , Base Sequence , Binding Sites , Catalytic Domain , Crystallography, X-Ray , DNA/chemistry , DNA Repair , Deoxyribodipyrimidine Photo-Lyase/genetics , Electrophoretic Mobility Shift Assay , Flavin-Adenine Dinucleotide/chemistry , Hydrogen Bonding , Molecular Sequence Data , Nucleic Acid Conformation , Oryza/genetics , Phosphorylation , Phylogeny , Plant Proteins/genetics , Polymorphism, Genetic , Protein Binding , Structural Homology, Protein , Surface Properties , Ultraviolet Rays
20.
J Biol Chem ; 286(51): 44254-44265, 2011 Dec 23.
Article in English | MEDLINE | ID: mdl-22027840

ABSTRACT

Pilin proteins assemble into Type IV pili (T4P), surface-displayed bacterial filaments with virulence functions including motility, attachment, transformation, immune escape, and colony formation. However, challenges in crystallizing full-length fiber-forming and membrane protein pilins leave unanswered questions regarding pilin structures, assembly, functions, and vaccine potential. Here we report pilin structures of full-length DnFimA from the sheep pathogen Dichelobacter nodosus and FtPilE from the human pathogen Francisella tularensis at 2.3 and 1 Å resolution, respectively. The DnFimA structure reveals an extended kinked N-terminal α-helix, an unusual centrally located disulfide, conserved subdomains, and assembled epitopes informing serogroup vaccines. An interaction between the conserved Glu-5 carboxyl oxygen and the N-terminal amine of an adjacent subunit in the crystallographic dimer is consistent with the hypothesis of a salt bridge between these groups driving T4P assembly. The FtPilE structure identifies an authentic Type IV pilin and provides a framework for understanding the role of T4P in F. tularensis virulence. Combined results define a unified pilin architecture, specialized subdomain roles in pilus assembly and function, and potential therapeutic targets.


Subject(s)
Bacterial Proteins/chemistry , Bacterial Vaccines/chemistry , Dichelobacter nodosus/chemistry , Fimbriae Proteins/chemistry , Fimbriae Proteins/metabolism , Fimbriae, Bacterial/chemistry , Amino Acid Sequence , Crystallography, X-Ray/methods , Models, Molecular , Molecular Sequence Data , Polymers/chemistry , Protein Conformation , Sequence Homology, Amino Acid
SELECTION OF CITATIONS
SEARCH DETAIL
...