Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
1.
Cell ; 145(2): 198-211, 2011 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-21496641

RESUMEN

Flap endonuclease (FEN1), essential for DNA replication and repair, removes RNA and DNA 5' flaps. FEN1 5' nuclease superfamily members acting in nucleotide excision repair (XPG), mismatch repair (EXO1), and homologous recombination (GEN1) paradoxically incise structurally distinct bubbles, ends, or Holliday junctions, respectively. Here, structural and functional analyses of human FEN1:DNA complexes show structure-specific, sequence-independent recognition for nicked dsDNA bent 100° with unpaired 3' and 5' flaps. Above the active site, a helical cap over a gateway formed by two helices enforces ssDNA threading and specificity for free 5' ends. Crystallographic analyses of product and substrate complexes reveal that dsDNA binding and bending, the ssDNA gateway, and double-base unpairing flanking the scissile phosphate control precise flap incision by the two-metal-ion active site. Superfamily conserved motifs bind and open dsDNA; direct the target region into the helical gateway, permitting only nonbase-paired oligonucleotides active site access; and support a unified understanding of superfamily substrate specificity.


Asunto(s)
Endonucleasas de ADN Solapado/química , Endonucleasas de ADN Solapado/metabolismo , Secuencia de Aminoácidos , Dominio Catalítico , ADN/metabolismo , Análisis Mutacional de ADN , Exodesoxirribonucleasas/química , Exodesoxirribonucleasas/metabolismo , Humanos , Modelos Moleculares , Datos de Secuencia Molecular , Alineación de Secuencia , Especificidad por Sustrato
2.
J Biol Chem ; 300(6): 107368, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38750793

RESUMEN

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.


Asunto(s)
Hidrolasas Diéster Fosfóricas , Humanos , Hidrolasas Diéster Fosfóricas/metabolismo , Hidrolasas Diéster Fosfóricas/química , Hidrolasas Diéster Fosfóricas/genética , Cristalografía por Rayos X , Biología Computacional/métodos , Motivos de Unión al ARN/genética
3.
Cell ; 133(5): 789-800, 2008 May 30.
Artículo en Inglés | MEDLINE | ID: mdl-18510924

RESUMEN

Mutations in XPD helicase, required for nucleotide excision repair (NER) as part of the transcription/repair complex TFIIH, cause three distinct phenotypes: cancer-prone xeroderma pigmentosum (XP), or aging disorders Cockayne syndrome (CS), and trichothiodystrophy (TTD). To clarify molecular differences underlying these diseases, we determined crystal structures of the XPD catalytic core from Sulfolobus acidocaldarius and measured mutant enzyme activities. Substrate-binding grooves separate adjacent Rad51/RecA-like helicase domains (HD1, HD2) and an arch formed by 4FeS and Arch domains. XP mutations map along the HD1 ATP-binding edge and HD2 DNA-binding channel and impair helicase activity essential for NER. XP/CS mutations both impair helicase activity and likely affect HD2 functional movement. TTD mutants lose or retain helicase activity but map to sites in all four domains expected to cause framework defects impacting TFIIH integrity. These results provide a foundation for understanding disease consequences of mutations in XPD and related 4Fe-4S helicases including FancJ.


Asunto(s)
Proteínas Arqueales/química , Proteínas Arqueales/genética , Sulfolobus acidocaldarius/enzimología , Proteína de la Xerodermia Pigmentosa del Grupo D/química , Proteína de la Xerodermia Pigmentosa del Grupo D/genética , Proteínas Arqueales/metabolismo , Síndrome de Cockayne/genética , Síndrome de Cockayne/metabolismo , Cristalografía por Rayos X , ADN Helicasas/química , ADN Helicasas/genética , ADN Helicasas/metabolismo , Reparación del ADN , Proteínas Hierro-Azufre/química , Proteínas Hierro-Azufre/genética , Modelos Moleculares , Mutación , Estructura Terciaria de Proteína , Homología de Secuencia de Aminoácido , Homología Estructural de Proteína , Síndromes de Tricotiodistrofia/genética , Síndromes de Tricotiodistrofia/metabolismo , Xerodermia Pigmentosa/genética , Xerodermia Pigmentosa/metabolismo , Proteína de la Xerodermia Pigmentosa del Grupo D/metabolismo
4.
Proc Natl Acad Sci U S A ; 117(25): 14127-14138, 2020 06 23.
Artículo en Inglés | MEDLINE | ID: mdl-32522879

RESUMEN

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.


Asunto(s)
Síndrome de Cockayne/genética , Proteínas de Unión al ADN/química , Endonucleasas/química , Proteínas Nucleares/química , Mutación Puntual , Factores de Transcripción/química , Xerodermia Pigmentosa/genética , Sitios de Unión , Secuencia Conservada , ADN/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Endonucleasas/genética , Endonucleasas/metabolismo , Estabilidad de Enzimas , Humanos , Simulación de Dinámica Molecular , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Fenotipo , Unión Proteica , Pliegue de Proteína , Multimerización de Proteína , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
5.
Mol Cell ; 53(1): 7-18, 2014 Jan 09.
Artículo en Inglés | MEDLINE | ID: mdl-24316220

RESUMEN

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.


Asunto(s)
Roturas del ADN de Doble Cadena , Reparación del ADN por Unión de Extremidades , Proteínas de Unión al ADN/metabolismo , Inhibidores Enzimáticos/química , Fase G2 , Reparación del ADN por Recombinación , Línea Celular , Cromatina/genética , Cromatina/metabolismo , Enzimas Reparadoras del ADN/genética , Enzimas Reparadoras del ADN/metabolismo , Proteínas de Unión al ADN/antagonistas & inhibidores , Proteínas de Unión al ADN/genética , Exodesoxirribonucleasas/genética , Exodesoxirribonucleasas/metabolismo , Rayos gamma/efectos adversos , Humanos , Proteína Homóloga de MRE11 , Proteína de Replicación A/genética , Proteína de Replicación A/metabolismo
6.
Mol Cell ; 47(1): 50-60, 2012 Jul 13.
Artículo en Inglés | MEDLINE | ID: mdl-22658721

RESUMEN

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.


Asunto(s)
Transferasas Alquil y Aril/metabolismo , Reparación del ADN , Guanina/análogos & derivados , Proteínas de Schizosaccharomyces pombe/metabolismo , Transferasas Alquil y Aril/química , Transferasas Alquil y Aril/genética , Western Blotting , Cristalografía por Rayos X , Daño del ADN , ADN de Hongos/química , ADN de Hongos/genética , ADN de Hongos/metabolismo , Citometría de Flujo , Fase G1/efectos de los fármacos , Genoma Fúngico/genética , Guanina/química , Guanina/metabolismo , Metilnitronitrosoguanidina/toxicidad , Modelos Moleculares , Mutación , Compuestos de Nitrosourea/toxicidad , Conformación de Ácido Nucleico , Unión Proteica , Estructura Terciaria de Proteína , Schizosaccharomyces/efectos de los fármacos , Schizosaccharomyces/genética , Schizosaccharomyces/metabolismo , Proteínas de Schizosaccharomyces pombe/química , Proteínas de Schizosaccharomyces pombe/genética , Transcripción Genética/genética
7.
Mol Microbiol ; 99(4): 674-85, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26508112

RESUMEN

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.


Asunto(s)
Proteínas Arqueales/química , Proteínas Arqueales/metabolismo , Flagelos/fisiología , Nucleótidos/metabolismo , Sulfolobus acidocaldarius/metabolismo , Adenosina Trifosfatasas/metabolismo , Adenosina Trifosfato/metabolismo , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Proteínas Arqueales/fisiología , Cristalización , Cristalografía por Rayos X , Flagelina/metabolismo , Genes Arqueales , Microscopía Electrónica , Modelos Moleculares , Movimiento , Sulfolobus acidocaldarius/genética
8.
Proc Natl Acad Sci U S A ; 111(23): 8446-51, 2014 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-24912171

RESUMEN

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.


Asunto(s)
Proteínas Bacterianas/metabolismo , Celulosa/metabolismo , Oxigenasas de Función Mixta/metabolismo , Polisacáridos/metabolismo , Secuencia de Aminoácidos , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Dominio Catalítico , Quitina/metabolismo , Cobre/metabolismo , Cristalografía por Rayos X , Espectroscopía de Resonancia por Spin del Electrón , Oxigenasas de Función Mixta/química , Oxigenasas de Función Mixta/genética , Modelos Moleculares , Datos de Secuencia Molecular , Oxidación-Reducción , Unión Proteica , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Homología de Secuencia de Aminoácido , Especificidad por Sustrato , Zinc/metabolismo
9.
EMBO Rep ; 15(5): 601-8, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24714598

RESUMEN

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.


Asunto(s)
Reparación del ADN , Proteínas Nucleares/metabolismo , Proteínas Nucleares/ultraestructura , Nucleosomas/metabolismo , Proteínas Modificadoras Pequeñas Relacionadas con Ubiquitina/metabolismo , Factores de Transcripción/metabolismo , Factores de Transcripción/ultraestructura , Secuencias de Aminoácidos , Animales , Línea Celular , Proteínas Cromosómicas no Histona/metabolismo , Cristalografía por Rayos X , Daño del ADN , Proteínas de Unión al ADN/metabolismo , Técnicas de Inactivación de Genes , Ratones , Proteínas Nucleares/genética , Procesamiento Proteico-Postraduccional , Estructura Terciaria de Proteína , Tamoxifeno/análogos & derivados , Tamoxifeno/farmacología , Telómero/efectos de los fármacos , Telómero/genética , Proteína 2 de Unión a Repeticiones Teloméricas/genética , Factores de Transcripción/genética , Proteína 1 de Unión al Supresor Tumoral P53 , Ubiquitina/metabolismo , Ubiquitina-Proteína Ligasas , Ubiquitinación
10.
Nature ; 459(7248): 808-13, 2009 Jun 11.
Artículo en Inglés | MEDLINE | ID: mdl-19516334

RESUMEN

Alkyltransferase-like proteins (ATLs) share functional motifs with the cancer chemotherapy target O(6)-alkylguanine-DNA alkyltransferase (AGT) and paradoxically protect cells from the biological effects of DNA alkylation damage, despite lacking the reactive cysteine and alkyltransferase activity of AGT. Here we determine Schizosaccharomyces pombe ATL structures without and with damaged DNA containing the endogenous lesion O(6)-methylguanine or cigarette-smoke-derived O(6)-4-(3-pyridyl)-4-oxobutylguanine. These results reveal non-enzymatic DNA nucleotide flipping plus increased DNA distortion and binding pocket size compared to AGT. Our analysis of lesion-binding site conservation identifies new ATLs in sea anemone and ancestral archaea, indicating that ATL interactions are ancestral to present-day repair pathways in all domains of life. Genetic connections to mammalian XPG (also known as ERCC5) and ERCC1 in S. pombe homologues Rad13 and Swi10 and biochemical interactions with Escherichia coli UvrA and UvrC combined with structural results reveal that ATLs sculpt alkylated DNA to create a genetic and structural intersection of base damage processing with nucleotide excision repair.


Asunto(s)
Transferasas Alquil y Aril/química , Transferasas Alquil y Aril/metabolismo , Daño del ADN , Reparación del ADN , Alquilación , Sitios de Unión , Cristalografía por Rayos X , ADN/química , ADN/metabolismo , Guanina/análogos & derivados , Guanina/química , Guanina/metabolismo , Humanos , Modelos Moleculares , Unión Proteica , Conformación Proteica
11.
J Biol Chem ; 288(12): 8445-8455, 2013 Mar 22.
Artículo en Inglés | MEDLINE | ID: mdl-23355472

RESUMEN

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.


Asunto(s)
Proteínas Bacterianas/química , ADN-(Sitio Apurínico o Apirimidínico) Liasa/química , Desoxirribonucleasa IV (Fago T4-Inducido)/química , Thermotoga maritima/enzimología , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Dominio Catalítico , Secuencia Conservada , Cristalografía por Rayos X , ADN/química , ADN-(Sitio Apurínico o Apirimidínico) Liasa/genética , Escherichia coli , Humanos , Simulación de Dinámica Molecular , Mutagénesis Sitio-Dirigida , Conformación de Ácido Nucleico , Unión Proteica , Estructura Secundaria de Proteína , Homología Estructural de Proteína
12.
J Biol Chem ; 287(26): 22295-304, 2012 Jun 22.
Artículo en Inglés | MEDLINE | ID: mdl-22573334

RESUMEN

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.


Asunto(s)
Flavoproteínas/química , Proteínas Luminiscentes/química , Fotoquímica/métodos , Animales , Arabidopsis/metabolismo , Línea Celular , Cristalografía por Rayos X/métodos , Flavoproteínas/metabolismo , Fluorescencia , Genes Reporteros , Haplorrinos , Luz , Modelos Moleculares , Mutagénesis , Oxígeno/química , Fototropinas/química , Conformación Proteica , Espectrofotometría/métodos
13.
J Biol Chem ; 287(15): 12060-9, 2012 Apr 06.
Artículo en Inglés | MEDLINE | ID: mdl-22170053

RESUMEN

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.


Asunto(s)
Desoxirribodipirimidina Fotoliasa/química , Oryza/enzimología , Proteínas de Plantas/química , Secuencias de Aminoácidos , Secuencia de Bases , Sitios de Unión , Dominio Catalítico , Cristalografía por Rayos X , ADN/química , Reparación del ADN , Desoxirribodipirimidina Fotoliasa/genética , Ensayo de Cambio de Movilidad Electroforética , Flavina-Adenina Dinucleótido/química , Enlace de Hidrógeno , Datos de Secuencia Molecular , Conformación de Ácido Nucleico , Oryza/genética , Fosforilación , Filogenia , Proteínas de Plantas/genética , Polimorfismo Genético , Unión Proteica , Homología Estructural de Proteína , Propiedades de Superficie , Rayos Ultravioleta
14.
J Biol Chem ; 287(43): 36322-30, 2012 Oct 19.
Artículo en Inglés | MEDLINE | ID: mdl-22952223

RESUMEN

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.


Asunto(s)
Proteínas Bacterianas/química , Glicósido Hidrolasas/química , Modelos Químicos , Simulación de Dinámica Molecular , Serratia marcescens/enzimología , Dominio Catalítico , Cristalografía por Rayos X , Relación Estructura-Actividad
15.
Nat Commun ; 14(1): 4445, 2023 07 24.
Artículo en Inglés | MEDLINE | ID: mdl-37488098

RESUMEN

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.


Asunto(s)
Neoplasias de la Próstata , Masculino , Humanos , Recombinasa Rad51 , Mutación , Replicación del ADN , Adenosina Trifosfato , Proteínas de Unión al ADN
16.
J Biol Chem ; 286(51): 44254-44265, 2011 Dec 23.
Artículo en Inglés | MEDLINE | ID: mdl-22027840

RESUMEN

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.


Asunto(s)
Proteínas Bacterianas/química , Vacunas Bacterianas/química , Dichelobacter nodosus/química , Proteínas Fimbrias/química , Proteínas Fimbrias/metabolismo , Fimbrias Bacterianas/química , Secuencia de Aminoácidos , Cristalografía por Rayos X/métodos , Modelos Moleculares , Datos de Secuencia Molecular , Polímeros/química , Conformación Proteica , Homología de Secuencia de Aminoácido
17.
Proc Natl Acad Sci U S A ; 106(17): 6962-7, 2009 Apr 28.
Artículo en Inglés | MEDLINE | ID: mdl-19359474

RESUMEN

Homologous flavoproteins from the photolyase (PHR)/cryptochrome (CRY) family use the FAD cofactor in PHRs to catalyze DNA repair and in CRYs to tune the circadian clock and control development. To help address how PHR/CRY members achieve these diverse functions, we determined the crystallographic structure of Arabidopsis thaliana (6-4) PHR (UVR3), which is strikingly (>65%) similar in sequence to human circadian clock CRYs. The structure reveals a substrate-binding cavity specific for the UV-induced DNA lesion, (6-4) photoproduct, and cofactor binding sites different from those of bacterial PHRs and consistent with distinct mechanisms for activities and regulation. Mutational analyses were combined with this prototypic structure for the (6-4) PHR/clock CRY cluster to identify structural and functional motifs: phosphate-binding and Pro-Lys-Leu protrusion motifs constricting access to the substrate-binding cavity above FAD, sulfur loop near the external end of the Trp electron-transfer pathway, and previously undefined C-terminal helix. Our results provide a detailed, unified framework for investigations of (6-4) PHRs and the mammalian CRYs. Conservation of key residues and motifs controlling FAD access and activities suggests that regulation of FAD redox properties and radical stability is essential not only for (6-4) photoproduct DNA repair, but also for circadian clock-regulating CRY functions. The structural and functional results reported here elucidate archetypal relationships within this flavoprotein family and suggest how PHRs and CRYs use local residue and cofactor tuning, rather than larger structural modifications, to achieve their diverse functions encompassing DNA repair, plant growth and development, and circadian clock regulation.


Asunto(s)
Arabidopsis/enzimología , Ritmo Circadiano , Reparación del ADN/genética , ADN de Plantas/metabolismo , Desoxirribodipirimidina Fotoliasa/química , Desoxirribodipirimidina Fotoliasa/metabolismo , Flavoproteínas/metabolismo , Secuencia de Aminoácidos , Animales , Arabidopsis/genética , Proteínas de Arabidopsis , Sitios de Unión , Secuencia Conservada , Criptocromos , Cristalografía por Rayos X , ADN de Plantas/genética , Desoxirribodipirimidina Fotoliasa/genética , Flavoproteínas/genética , Interacciones Hidrofóbicas e Hidrofílicas , Datos de Secuencia Molecular , Fosfatos/química , Fosfatos/metabolismo , Fosforilación , Estructura Terciaria de Proteína , Alineación de Secuencia
18.
J Biol Chem ; 285(41): 31581-9, 2010 Oct 08.
Artículo en Inglés | MEDLINE | ID: mdl-20659888

RESUMEN

Inducible nitric-oxide synthase (iNOS) produces biologically stressful levels of nitric oxide (NO) as a potent mediator of cellular cytotoxicity or signaling. Yet, how this nitrosative stress affects iNOS function in vivo is poorly understood. Here we define two specific non-heme iNOS nitrosation sites discovered by combining UV-visible spectroscopy, chemiluminescence, mass spectrometry, and x-ray crystallography. We detected auto-S-nitrosylation during enzymatic turnover by using chemiluminescence. Selective S-nitrosylation of the ZnS(4) site, which bridges the dimer interface, promoted a dimer-destabilizing order-to-disorder transition. The nitrosated iNOS crystal structure revealed an unexpected N-NO modification on the pterin cofactor. Furthermore, the structurally defined N-NO moiety is solvent-exposed and available to transfer NO to a partner. We investigated glutathione (GSH) as a potential transnitrosation partner because the intracellular GSH concentration is high and NOS can form S-nitrosoglutathione. Our computational results predicted a GSH binding site adjacent to the N-NO-pterin. Moreover, we detected GSH binding to iNOS with saturation transfer difference NMR spectroscopy. Collectively, these observations resolve previous paradoxes regarding this uncommon pterin cofactor in NOS and suggest means for regulating iNOS activity via N-NO-pterin and S-NO-Cys modifications. The iNOS self-nitrosation characterized here appears appropriate to help control NO production in response to cellular conditions.


Asunto(s)
Cisteína/química , Glutatión/química , Óxido Nítrico Sintasa de Tipo II/química , Óxido Nítrico/química , Multimerización de Proteína/fisiología , Pterinas/química , Regulación Alostérica/fisiología , Animales , Cristalografía por Rayos X , Cisteína/metabolismo , Glutatión/metabolismo , Ratones , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa de Tipo II/metabolismo , Resonancia Magnética Nuclear Biomolecular , Oxidación-Reducción , Estructura Cuaternaria de Proteína , Pterinas/metabolismo
19.
Mol Microbiol ; 77(3): 755-70, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20545841

RESUMEN

Type IV pili (T4P) are critical to virulence for Vibrio cholerae and other bacterial pathogens. Among their diverse functions, T4P mediate microcolony formation, which protects the bacteria from host defences and concentrates secreted toxins. The T4P of the two V. cholerae O1 disease biotypes, classical and El Tor, share 81% identity in their TcpA subunits, yet these filaments differ in their interaction patterns as assessed by electron microscopy. To understand the molecular basis for pilus-mediated microcolony formation, we solved a 1.5 A resolution crystal structure of N-terminally truncated El Tor TcpA and compared it with that of classical TcpA. Residues that differ between the two pilins are located on surface-exposed regions of the TcpA subunits. By iteratively changing these non-conserved amino acids in classical TcpA to their respective residues in El Tor TcpA, we identified residues that profoundly affect pilus:pilus interaction patterns and bacterial aggregation. These residues lie on either the protruding d-region of the TcpA subunit or in a cavity between pilin subunits in the pilus filament. Our results support a model whereby pili interact via intercalation of surface protrusions on one filament into depressions between subunits on adjacent filaments as a means to hold V. cholerae cells together in microcolonies.


Asunto(s)
Proteínas Fimbrias/química , Proteínas Fimbrias/metabolismo , Fimbrias Bacterianas/metabolismo , Vibrio cholerae/química , Vibrio cholerae/crecimiento & desarrollo , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Proteínas Fimbrias/genética , Fimbrias Bacterianas/química , Fimbrias Bacterianas/genética , Conformación Molecular , Datos de Secuencia Molecular , Alineación de Secuencia , Vibrio cholerae/genética , Vibrio cholerae/metabolismo
20.
Prog Biophys Mol Biol ; 163: 130-142, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-33115610

RESUMEN

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).


Asunto(s)
Descubrimiento de Drogas , Preparaciones Farmacéuticas , Cristalografía por Rayos X , Daño del ADN , Reparación del ADN
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA