Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 50
Filtrar
1.
J Biol Chem ; 297(3): 100995, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34302810

RESUMO

Human immunoglobulin G subclass 3 (IgG3) possesses a uniquely long hinge region that separates its Fab antigen-binding and Fc receptor-binding regions. Owing to this hinge length, the molecular structure of full-length IgG3 remains elusive, and the role of the two conserved Fc glycosylation sites are unknown. To address these issues, we subjected glycosylated and deglycosylated human myeloma IgG3 to multidisciplinary solution structure studies. Using analytical ultracentrifugation, the elongated structure of IgG3 was determined from the reduced sedimentation coefficients s020,w of 5.82 to 6.29 S for both glycosylated and deglycosylated IgG3. X-ray and neutron scattering showed that the Guinier RG values were 6.95 nm for glycosylated IgG3 and were unchanged after deglycosylation, again indicating an elongated structure. The distance distribution function P(r) showed a maximum length of 25 to 28 nm and three distinct maxima. The molecular structure of IgG3 was determined using atomistic modeling based on molecular dynamics simulations of the IgG3 hinge and Monte Carlo simulations to identify physically realistic arrangements of the Fab and Fc regions. This resulted in libraries containing 135,135 and 73,905 glycosylated and deglycosylated IgG3 structures, respectively. Comparisons with the X-ray and neutron scattering curves gave 100 best-fit models for each form of IgG3 that accounted for the experimental scattering curves. These models revealed the first molecular structures for full-length IgG3. The structures exhibited relatively restricted Fab and Fc conformations joined by an extended semirigid hinge, which explains the potent effector functions of IgG3 relative to the other subclasses IgG1, IgG2, and IgG4.


Assuntos
Fragmentos Fab das Imunoglobulinas/química , Imunoglobulina G/química , Mieloma Múltiplo/imunologia , Proteínas do Mieloma/química , Receptores Fc/química , Sequência de Aminoácidos , Cromatografia Líquida/métodos , Glicosilação , Humanos , Espectrometria de Massas/métodos , Simulação de Dinâmica Molecular , Nêutrons , Conformação Proteica , Espalhamento a Baixo Ângulo , Homologia de Sequência de Aminoácidos , Ultracentrifugação/métodos , Difração de Raios X
2.
Nature ; 535(7613): 517-522, 2016 07 28.
Artigo em Inglês | MEDLINE | ID: mdl-27437577

RESUMO

Developmental signals of the Hedgehog (Hh) and Wnt families are transduced across the membrane by Frizzledclass G-protein-coupled receptors (GPCRs) composed of both a heptahelical transmembrane domain (TMD) and an extracellular cysteine-rich domain (CRD). How the large extracellular domains of GPCRs regulate signalling by the TMD is unknown. We present crystal structures of the Hh signal transducer and oncoprotein Smoothened, a GPCR that contains two distinct ligand-binding sites: one in its TMD and one in the CRD. The CRD is stacked a top the TMD, separated by an intervening wedge-like linker domain. Structure-guided mutations show that the interface between the CRD, linker domain and TMD stabilizes the inactive state of Smoothened. Unexpectedly, we find a cholesterol molecule bound to Smoothened in the CRD binding site. Mutations predicted to prevent cholesterol binding impair the ability of Smoothened to transmit native Hh signals. Binding of a clinically used antagonist, vismodegib, to the TMD induces a conformational change that is propagated to the CRD, resulting in loss of cholesterol from the CRD-linker domain-TMD interface. Our results clarify the structural mechanism by which the activity of a GPCR is controlled by ligand-regulated interactions between its extracellular and transmembrane domains.


Assuntos
Espaço Extracelular/metabolismo , Receptores Acoplados a Proteínas G/química , Receptores Acoplados a Proteínas G/metabolismo , Anilidas/química , Anilidas/metabolismo , Anilidas/farmacologia , Antineoplásicos/metabolismo , Antineoplásicos/farmacologia , Sítios de Ligação/genética , Colesterol/metabolismo , Colesterol/farmacologia , Cristalografia por Raios X , Cisteína/química , Cisteína/genética , Cisteína/metabolismo , Proteínas Hedgehog/metabolismo , Humanos , Ligantes , Modelos Moleculares , Ligação Proteica/genética , Estabilidade Proteica/efeitos dos fármacos , Estrutura Terciária de Proteína/efeitos dos fármacos , Estrutura Terciária de Proteína/genética , Piridinas/química , Piridinas/metabolismo , Piridinas/farmacologia , Receptores Acoplados a Proteínas G/antagonistas & inibidores , Receptores Acoplados a Proteínas G/genética , Transdução de Sinais/efeitos dos fármacos , Receptor Smoothened
3.
Biophys J ; 120(9): 1814-1834, 2021 05 04.
Artigo em Inglês | MEDLINE | ID: mdl-33675758

RESUMO

The human immunoglobulin G (IgG) class is the most prevalent antibody in serum, with the IgG1 subclass being the most abundant. IgG1 is composed of two Fab regions connected to a Fc region through a 15-residue hinge peptide. Two glycan chains are conserved in the Fc region in IgG; however, their importance for the structure of intact IgG1 has remained unclear. Here, we subjected glycosylated and deglycosylated monoclonal human IgG1 (designated as A33) to a comparative multidisciplinary structural study of both forms. After deglycosylation using peptide:N-glycosidase F, analytical ultracentrifugation showed that IgG1 remained monomeric and the sedimentation coefficients s020,w of IgG1 decreased from 6.45 S by 0.16-0.27 S. This change was attributed to the reduction in mass after glycan removal. X-ray and neutron scattering revealed changes in the Guinier structural parameters after deglycosylation. Although the radius of gyration (RG) was unchanged, the cross-sectional radius of gyration (RXS-1) increased by 0.1 nm, and the commonly occurring distance peak M2 of the distance distribution curve P(r) increased by 0.4 nm. These changes revealed that the Fab-Fc separation in IgG1 was perturbed after deglycosylation. To explain these changes, atomistic scattering modeling based on Monte Carlo simulations resulted in 123,284 and 119,191 trial structures for glycosylated and deglycosylated IgG1 respectively. From these, 100 x-ray and neutron best-fit models were determined. For these, principal component analyses identified five groups of structural conformations that were different for glycosylated and deglycosylated IgG1. The Fc region in glycosylated IgG1 showed a restricted range of conformations relative to the Fab regions, whereas the Fc region in deglycosylated IgG1 showed a broader conformational spectrum. These more variable Fc conformations account for the loss of binding to the Fcγ receptor in deglycosylated IgG1.


Assuntos
Imunoglobulina G , Receptores de IgG , Estudos Transversais , Humanos , Modelos Moleculares , Polissacarídeos , Conformação Proteica
4.
Nature ; 527(7576): 114-7, 2015 Nov 05.
Artigo em Inglês | MEDLINE | ID: mdl-26503046

RESUMO

Negative-sense RNA viruses, such as influenza, encode large, multidomain RNA-dependent RNA polymerases that can both transcribe and replicate the viral RNA genome. In influenza virus, the polymerase (FluPol) is composed of three polypeptides: PB1, PB2 and PA/P3. PB1 houses the polymerase active site, whereas PB2 and PA/P3 contain, respectively, cap-binding and endonuclease domains required for transcription initiation by cap-snatching. Replication occurs through de novo initiation and involves a complementary RNA intermediate. Currently available structures of the influenza A and B virus polymerases include promoter RNA (the 5' and 3' termini of viral genome segments), showing FluPol in transcription pre-initiation states. Here we report the structure of apo-FluPol from an influenza C virus, solved by X-ray crystallography to 3.9 Å, revealing a new 'closed' conformation. The apo-FluPol forms a compact particle with PB1 at its centre, capped on one face by PB2 and clamped between the two globular domains of P3. Notably, this structure is radically different from those of promoter-bound FluPols. The endonuclease domain of P3 and the domains within the carboxy-terminal two-thirds of PB2 are completely rearranged. The cap-binding site is occluded by PB2, resulting in a conformation that is incompatible with transcription initiation. Thus, our structure captures FluPol in a closed, transcription pre-activation state. This reveals the conformation of newly made apo-FluPol in an infected cell, but may also apply to FluPol in the context of a non-transcribing ribonucleoprotein complex. Comparison of the apo-FluPol structure with those of promoter-bound FluPols allows us to propose a mechanism for FluPol activation. Our study demonstrates the remarkable flexibility of influenza virus RNA polymerase, and aids our understanding of the mechanisms controlling transcription and genome replication.


Assuntos
Gammainfluenzavirus/enzimologia , RNA Polimerase Dependente de RNA/química , Apoenzimas/química , Apoenzimas/metabolismo , Sítios de Ligação , Cristalografia por Raios X , Endonucleases/química , Endonucleases/metabolismo , Ativação Enzimática , Modelos Moleculares , Iniciação Traducional da Cadeia Peptídica , Regiões Promotoras Genéticas/genética , Ligação Proteica , Estrutura Terciária de Proteína , Subunidades Proteicas/química , Subunidades Proteicas/metabolismo , Capuzes de RNA/metabolismo , RNA Viral/biossíntese , RNA Viral/metabolismo , RNA Polimerase Dependente de RNA/metabolismo , Ribonucleoproteínas/química
5.
J Am Chem Soc ; 142(49): 20640-20650, 2020 12 09.
Artigo em Inglês | MEDLINE | ID: mdl-33252237

RESUMO

Controlling the assembly and disassembly of nanoscale protein cages for the capture and internalization of protein or non-proteinaceous components is fundamentally important to a diverse range of bionanotechnological applications. Here, we study the reversible, pressure-induced dissociation of a natural protein nanocage, E. coli bacterioferritin (Bfr), using synchrotron radiation small-angle X-ray scattering (SAXS) and circular dichroism (CD). We demonstrate that hydrostatic pressures of 450 MPa are sufficient to completely dissociate the Bfr 24-mer into protein dimers, and the reversibility and kinetics of the reassembly process can be controlled by selecting appropriate buffer conditions. We also demonstrate that the heme B prosthetic group present at the subunit dimer interface influences the stability and pressure lability of the cage, despite its location being discrete from the interdimer interface that is key to cage assembly. This indicates a major cage-stabilizing role for heme within this family of ferritins.


Assuntos
Proteínas de Bactérias/metabolismo , Grupo dos Citocromos b/metabolismo , Escherichia coli/metabolismo , Ferritinas/metabolismo , Proteínas de Bactérias/química , Dicroísmo Circular , Grupo dos Citocromos b/química , Dimerização , Ferritinas/química , Pressão Hidrostática , Cinética , Espalhamento a Baixo Ângulo , Termodinâmica , Difração de Raios X
6.
J Synchrotron Radiat ; 27(Pt 5): 1438-1446, 2020 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-32876621

RESUMO

B21 is a small-angle X-ray scattering (SAXS) beamline with a bending magnet source in the 3 GeV storage ring at the Diamond Light Source Ltd synchrotron in the UK. The beamline utilizes a double multi-layer monochromator and a toroidal focusing optic to deliver 2 × 1012 photons per second to a 34 × 40 µm (FWHM) focal spot at the in-vacuum Eiger 4M (Dectris) detector. A high-performance liquid chromatography system and a liquid-handling robot make it possible to load solution samples into a temperature-controlled in-vacuum sample cell with a high level of automation. Alternatively, a range of viscous or solid materials may be loaded manually using a range of custom sample cells. A default scattering vector range from 0.0026 to 0.34 Å-1 and low instrument background make B21 convenient for measuring a wide range of biological macromolecules. The beamline has run a full user programme since 2013.

7.
Langmuir ; 36(33): 9649-9657, 2020 08 25.
Artigo em Inglês | MEDLINE | ID: mdl-32202793

RESUMO

Bioproduction of poly(methyl methacrylate) is a fast growing global industry that is limited by cellular toxicity of monomeric methacrylate intermediates to the producer strains. Maintaining high methacrylate concentrations during biofermentation, required by economically viable technologies, challenges bacterial membrane stability and cellular viability. Studying the stability of model lipid membranes in the presence of methacrylates offers unique molecular insights into the mechanisms of methacrylate toxicity, as well as into the fundamental structural bases of membrane assembly. We investigate the structure and stability of model membranes in the presence of high levels of methacrylate esters using solid-state nuclear magnetic resonance (NMR) and small-angle X-ray scattering (SAXS). Wide-line 31P NMR spectroscopy shows that butyl methacrylate (BMA) can be incorporated into the lipid bilayer at concentrations as high as 75 mol % without significantly disrupting membrane integrity and that lipid acyl chain composition can influence membrane tolerance and ability to accommodate BMA. Using high resolution 13C magic angle spinning (MAS) NMR, we show that the presence of 75 mol % BMA lowers the lipid main transition temperature by over 12 degrees, which suggests that BMA intercalates between the lipid chains, causing uncoupling of collective lipid motions that are typically dominated by chain trans-gauche isomerization. Potential uncoupling of the bilayer leaflets to accommodate a separate BMA subphase was not supported by the SAXS experiments, which showed that membrane thickness remained unchanged even at 80% BMA. Reduced X-ray scattering contrast at the polar/apolar interface suggests BMA localization in that region between the lipid molecules.

8.
Genes Dev ; 26(2): 151-62, 2012 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-22279047

RESUMO

SMARCAL1 (SWI/SNF-related, matrix-associated, actin-dependent regulator of chromatin, subfamily A-like1) maintains genome integrity during DNA replication. Here we investigated its mechanism of action. We found that SMARCAL1 travels with elongating replication forks, and its absence leads to MUS81-dependent double-strand break formation. Binding to specific nucleic acid substrates activates SMARCAL1 activity in a reaction that requires its HARP2 (Hep-A-related protein 2) domain. Homology modeling indicates that the HARP domain is similar in structure to the DNA-binding domain of the PUR proteins. Limited proteolysis, small-angle X-ray scattering, and functional assays indicate that the core enzymatic unit consists of the HARP2 and ATPase domains that fold into a stable structure. Surprisingly, SMARCAL1 is capable of binding three-way and four-way Holliday junctions and model replication forks that lack a designed ssDNA region. Furthermore, SMARCAL1 remodels these DNA substrates by promoting branch migration and fork regression. SMARCAL1 mutations that cause Schimke immunoosseous dysplasia or that inactivate the HARP2 domain abrogate these activities. These results suggest that SMARCAL1 continuously surveys replication forks for damage. If damage is present, it remodels the fork to promote repair and restart. Failures in the process lead to activation of an alternative repair mechanism that depends on MUS81-catalyzed cleavage of the damaged fork.


Assuntos
DNA Helicases/metabolismo , Replicação do DNA/fisiologia , DNA Cruciforme/metabolismo , Instabilidade Genômica/fisiologia , Linhagem Celular Tumoral , Quebras de DNA de Cadeia Dupla , DNA Helicases/genética , Replicação do DNA/genética , DNA de Cadeia Simples/metabolismo , Proteínas de Ligação a DNA/metabolismo , Endonucleases/metabolismo , Instabilidade Genômica/genética , Células HEK293 , Humanos , Ligação Proteica , Estrutura Terciária de Proteína , Fase S
9.
J Biol Chem ; 293(33): 12862-12876, 2018 08 17.
Artigo em Inglês | MEDLINE | ID: mdl-29880640

RESUMO

Mitochondrial tRNAs are transcribed as long polycistronic transcripts of precursor tRNAs and undergo posttranscriptional modifications such as endonucleolytic processing and methylation required for their correct structure and function. Among them, 5'-end processing and purine 9 N1-methylation of mitochondrial tRNA are catalyzed by two proteinaceous complexes with overlapping subunit composition. The Mg2+-dependent RNase P complex for 5'-end cleavage comprises the methyltransferase domain-containing protein tRNA methyltransferase 10C, mitochondrial RNase P subunit (TRMT10C/MRPP1), short-chain oxidoreductase hydroxysteroid 17ß-dehydrogenase 10 (HSD17B10/MRPP2), and metallonuclease KIAA0391/MRPP3. An MRPP1-MRPP2 subcomplex also catalyzes the formation of 1-methyladenosine/1-methylguanosine at position 9 using S-adenosyl-l-methionine as methyl donor. However, a lack of structural information has precluded insights into how these complexes methylate and process mitochondrial tRNA. Here, we used a combination of X-ray crystallography, interaction and activity assays, and small angle X-ray scattering (SAXS) to gain structural insight into the two tRNA modification complexes and their components. The MRPP1 N terminus is involved in tRNA binding and monomer-monomer self-interaction, whereas the C-terminal SPOUT fold contains key residues for S-adenosyl-l-methionine binding and N1-methylation. The entirety of MRPP1 interacts with MRPP2 to form the N1-methylation complex, whereas the MRPP1-MRPP2-MRPP3 RNase P complex only assembles in the presence of precursor tRNA. This study proposes low-resolution models of the MRPP1-MRPP2 and MRPP1-MRPP2-MRPP3 complexes that suggest the overall architecture, stoichiometry, and orientation of subunits and tRNA substrates.


Assuntos
3-Hidroxiacil-CoA Desidrogenases/química , Metiltransferases/química , Modelos Moleculares , Complexos Multienzimáticos/química , RNA Mitocondrial/química , RNA de Transferência/química , Ribonuclease P/química , 3-Hidroxiacil-CoA Desidrogenases/metabolismo , Cristalografia por Raios X , Humanos , Metiltransferases/metabolismo , Complexos Multienzimáticos/metabolismo , RNA Mitocondrial/metabolismo , RNA de Transferência/metabolismo , Ribonuclease P/metabolismo , Espalhamento a Baixo Ângulo
10.
Org Biomol Chem ; 17(18): 4543-4553, 2019 05 08.
Artigo em Inglês | MEDLINE | ID: mdl-30994696

RESUMO

We investigate the self-assembly of a palmitoylated (C16-chain at the N terminus) peptide fragment in comparison to the unlipidated peptide EELNRYY, a fragment of the gut hormone peptide PYY3-36. The lipopeptide C16-EELNRYY shows remarkable pH-dependent self-assembly above measured critical aggregation concentrations, forming fibrils at pH 7, but micelles at pH 10. The parent peptide does not show self-assembly behaviour. The lipopeptide forms hydrogels at sufficiently high concentration at pH 7, the dynamic mechanical properties of which were measured. We also show that the tyrosine functionality at the C terminus of EELNRYY can be used to enzymatically produce the pigment melanin. The enzyme tyrosinase oxidises tyrosine into 3,4-dihydroxyphenylalanine (DOPA), DOPA-quinone and further products, eventually forming eumelanin. This is a mechanism of photo-protection in the skin, for this reason controlling tyrosinase activity is a major target for skin care applications and EELNRYY has potential to be developed for such uses.


Assuntos
Lipopeptídeos/química , Melaninas/síntese química , Monofenol Mono-Oxigenase/química , Oligopeptídeos/química , Fragmentos de Peptídeos/química , Peptídeo YY/química , Sequência de Aminoácidos , Corantes Fluorescentes/química , Hidrogéis/química , Concentração de Íons de Hidrogênio , Lipopeptídeos/metabolismo , Micelas , Oligopeptídeos/metabolismo , Fragmentos de Peptídeos/metabolismo , Peptídeo YY/metabolismo , Conformação Proteica em Folha beta , Multimerização Proteica , Pirenos/química , Tirosina/química
11.
Nature ; 496(7446): 477-81, 2013 Apr 25.
Artigo em Inglês | MEDLINE | ID: mdl-23619693

RESUMO

Modern small-angle scattering (SAS) experiments with X-rays or neutrons provide a comprehensive, resolution-limited observation of the thermodynamic state. However, methods for evaluating mass and validating SAS-based models and resolution have been inadequate. Here we define the volume of correlation, Vc, a SAS invariant derived from the scattered intensities that is specific to the structural state of the particle, but independent of concentration and the requirements of a compact, folded particle. We show that Vc defines a ratio, QR, that determines the molecular mass of proteins or RNA ranging from 10 to 1,000 kilodaltons. Furthermore, we propose a statistically robust method for assessing model-data agreements (χ(2)free) akin to cross-validation. Our approach prevents over-fitting of the SAS data and can be used with a newly defined metric, RSAS, for quantitative evaluation of resolution. Together, these metrics (Vc, QR, χ(2)free and RSAS) provide analytical tools for unbiased and accurate macromolecular structural characterizations in solution.


Assuntos
Modelos Moleculares , Espalhamento a Baixo Ângulo , Aldose-Cetose Isomerases/química , Proteínas de Ligação a DNA/química , Modelos Químicos , Conformação Molecular , Peso Molecular , Maleabilidade , RNA Viral/química , Proteínas de Ligação a RNA , Reprodutibilidade dos Testes , Riboswitch/genética , Soluções
12.
Biomacromolecules ; 19(1): 167-177, 2018 01 08.
Artigo em Inglês | MEDLINE | ID: mdl-29195274

RESUMO

We investigate the self-assembly of two telechelic star polymer-peptide conjugates based on poly(ethylene oxide) (PEO) four-arm star polymers capped with oligotyrosine. The conjugates were prepared via N-carboxy anhydride-mediated ring-opening polymerization from PEO star polymer macroinitiators. Self-assembly occurs above a critical aggregation concentration determined via fluorescence probe assays. Peptide conformation was examined using circular dichroism spectroscopy. The structure of self-assembled aggregates was probed using small-angle X-ray scattering and cryogenic transmission electron microscopy. In contrast to previous studies on linear telechelic PEO-oligotyrosine conjugates that show self-assembly into ß-sheet fibrils, the star architecture suppresses fibril formation and micelles are generally observed instead, a small population of fibrils only being observed upon pH adjustment. Hydrogelation is also suppressed by the polymer star architecture. These peptide-functionalized star polymer solutions are cytocompatible at sufficiently low concentration. These systems present tyrosine at high density and may be useful in the development of future enzyme or pH-responsive biomaterials.


Assuntos
Polietilenoglicóis/química , Tirosina/química , Água/química , Linhagem Celular , Cromatografia em Gel , Dicroísmo Circular , Humanos , Hidrogéis/química , Espectroscopia de Ressonância Magnética , Microscopia Eletrônica de Transmissão , Peptídeos/química , Polimerização , Espalhamento a Baixo Ângulo , Soluções , Espectroscopia de Infravermelho com Transformada de Fourier , Difração de Raios X
13.
Mol Cell ; 40(3): 347-8, 2010 Nov 12.
Artigo em Inglês | MEDLINE | ID: mdl-21070960

RESUMO

In this issue of Molecular Cell, Savir and Tlusty (2010) apply signal detection theory to show that homologous recombination machinery is optimally tuned to find homologous DNA sequences within an exceptionally high background of heterologous sequences.

14.
Adv Exp Med Biol ; 1009: 31-45, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29218552

RESUMO

The success of a SAXS experiment for structural investigations depends on two precise measurements, the sample and the buffer background. Buffer matching between the sample and background can be achieved using dialysis methods but in biological SAXS of monodisperse systems, sample preparation is routinely being performed with size exclusion chromatography (SEC). SEC is the most reliable method for SAXS sample preparation as the method not only purifies the sample for SAXS but also almost guarantees ideal buffer matching. Here, I will highlight the use of SEC for SAXS sample preparation and demonstrate using example proteins that SEC purification does not always provide for ideal samples. Scrutiny of the SEC elution peak using quasi-elastic and multi-angle light scattering techniques can reveal hidden features (heterogeneity) of the sample that should be considered during SAXS data analysis. In some cases, sample heterogeneity can be controlled using a small molecule additive and I outline a simple additive screening method for sample preparation.


Assuntos
Cromatografia em Gel/métodos , Proteínas/ultraestrutura , Espalhamento a Baixo Ângulo , Manejo de Espécimes/métodos , Síncrotrons/instrumentação , Difração de Raios X/normas , Soluções Tampão , Cromatografia em Gel/instrumentação , Excipientes/química , Humanos , Fosfatos/química , Agregados Proteicos , Conformação Proteica , Proteínas/química , Sacarose/química , Difração de Raios X/instrumentação , Difração de Raios X/métodos
15.
Proc Natl Acad Sci U S A ; 111(21): 7618-23, 2014 May 27.
Artigo em Inglês | MEDLINE | ID: mdl-24821763

RESUMO

SMARCAL1, a DNA remodeling protein fundamental to genome integrity during replication, is the only gene associated with the developmental disorder Schimke immuno-osseous dysplasia (SIOD). SMARCAL1-deficient cells show collapsed replication forks, S-phase cell cycle arrest, increased chromosomal breaks, hypersensitivity to genotoxic agents, and chromosomal instability. The SMARCAL1 catalytic domain (SMARCAL1(CD)) is composed of an SNF2-type double-stranded DNA motor ATPase fused to a HARP domain of unknown function. The mechanisms by which SMARCAL1 and other DNA translocases repair replication forks are poorly understood, in part because of a lack of structural information on the domains outside of the common ATPase motor. In the present work, we determined the crystal structure of the SMARCAL1 HARP domain and examined its conformation and assembly in solution by small angle X-ray scattering. We report that this domain is conserved with the DNA mismatch and damage recognition domains of MutS/MSH and NER helicase XPB, respectively, as well as with the putative DNA specificity motif of the T4 phage fork regression protein UvsW. Loss of UvsW fork regression activity by deletion of this domain was rescued by its replacement with HARP, establishing the importance of this domain in UvsW and demonstrating a functional complementarity between these structurally homologous domains. Mutation of predicted DNA-binding residues in HARP dramatically reduced fork binding and regression activities of SMARCAL1(CD). Thus, this work has uncovered a conserved substrate recognition domain in DNA repair enzymes that couples ATP-hydrolysis to remodeling of a variety of DNA structures, and provides insight into this domain's role in replication fork stability and genome integrity.


Assuntos
DNA Helicases/química , DNA Helicases/metabolismo , Reparo do DNA/genética , Modelos Moleculares , Ácidos Nucleicos/metabolismo , Trifosfato de Adenosina/metabolismo , Animais , Cromatografia de Afinidade , Cromatografia em Agarose , Cromatografia em Gel , Cromatografia por Troca Iônica , Clonagem Molecular , Cristalização , DNA Helicases/biossíntese , Hidrólise , Funções Verossimilhança , Camundongos , Estrutura Terciária de Proteína , Espalhamento a Baixo Ângulo , Difração de Raios X
16.
Proc Natl Acad Sci U S A ; 111(43): E4568-76, 2014 Oct 28.
Artigo em Inglês | MEDLINE | ID: mdl-25316790

RESUMO

Protein framework alterations in heritable Cu, Zn superoxide dismutase (SOD) mutants cause misassembly and aggregation in cells affected by the motor neuron disease ALS. However, the mechanistic relationship between superoxide dismutase 1 (SOD1) mutations and human disease is controversial, with many hypotheses postulated for the propensity of specific SOD mutants to cause ALS. Here, we experimentally identify distinguishing attributes of ALS mutant SOD proteins that correlate with clinical severity by applying solution biophysical techniques to six ALS mutants at human SOD hotspot glycine 93. A small-angle X-ray scattering (SAXS) assay and other structural methods assessed aggregation propensity by defining the size and shape of fibrillar SOD aggregates after mild biochemical perturbations. Inductively coupled plasma MS quantified metal ion binding stoichiometry, and pulsed dipolar ESR spectroscopy evaluated the Cu(2+) binding site and defined cross-dimer copper-copper distance distributions. Importantly, we find that copper deficiency in these mutants promotes aggregation in a manner strikingly consistent with their clinical severities. G93 mutants seem to properly incorporate metal ions under physiological conditions when assisted by the copper chaperone but release copper under destabilizing conditions more readily than the WT enzyme. Altered intradimer flexibility in ALS mutants may cause differential metal retention and promote distinct aggregation trends observed for mutant proteins in vitro and in ALS patients. Combined biophysical and structural results test and link copper retention to the framework destabilization hypothesis as a unifying general mechanism for both SOD aggregation and ALS disease progression, with implications for disease severity and therapeutic intervention strategies.


Assuntos
Esclerose Lateral Amiotrófica/enzimologia , Esclerose Lateral Amiotrófica/patologia , Mutação/genética , Agregação Patológica de Proteínas/enzimologia , Agregação Patológica de Proteínas/genética , Superóxido Dismutase/genética , Ácidos/metabolismo , Esclerose Lateral Amiotrófica/genética , Cobre/farmacologia , Cristalografia por Raios X , Ácido Edético/farmacologia , Humanos , Modelos Moleculares , Proteínas Mutantes/química , Proteínas Mutantes/genética , Fenótipo , Substâncias Protetoras/farmacologia , Espalhamento a Baixo Ângulo , Soluções , Superóxido Dismutase/química , Superóxido Dismutase-1
17.
EMBO Rep ; 15(5): 601-8, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24714598

RESUMO

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.


Assuntos
Reparo do DNA , Proteínas Nucleares/metabolismo , Proteínas Nucleares/ultraestrutura , Nucleossomos/metabolismo , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/metabolismo , Fatores de Transcrição/metabolismo , Fatores de Transcrição/ultraestrutura , Motivos de Aminoácidos , Animais , Linhagem Celular , Proteínas Cromossômicas não Histona/metabolismo , Cristalografia por Raios X , Dano ao DNA , Proteínas de Ligação a DNA/metabolismo , Técnicas de Inativação de Genes , Camundongos , Proteínas Nucleares/genética , Processamento de Proteína Pós-Traducional , Estrutura Terciária de Proteína , Tamoxifeno/análogos & derivados , Tamoxifeno/farmacologia , Telômero/efeitos dos fármacos , Telômero/genética , Proteína 2 de Ligação a Repetições Teloméricas/genética , Fatores de Transcrição/genética , Proteína 1 de Ligação à Proteína Supressora de Tumor p53 , Ubiquitina/metabolismo , Ubiquitina-Proteína Ligases , Ubiquitinação
18.
Nucleic Acids Res ; 42(2): 906-17, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24150939

RESUMO

The Hop2-Mnd1 complex functions with the DMC1 recombinase in meiotic recombination. Hop2-Mnd1 stabilizes the DMC1-single-stranded DNA (ssDNA) filament and promotes the capture of the double-stranded DNA partner by the recombinase filament to assemble the synaptic complex. Herein, we define the action mechanism of Hop2-Mnd1 in DMC1-mediated recombination. Small angle X-ray scattering analysis and electron microscopy reveal that the heterodimeric Hop2-Mnd1 is a V-shaped molecule. We show that the protein complex harbors three distinct DNA binding sites, and determine their functional relevance. Specifically, the N-terminal double-stranded DNA binding functions of Hop2 and Mnd1 co-operate to mediate synaptic complex assembly, whereas ssDNA binding by the Hop2 C-terminus helps stabilize the DMC1-ssDNA filament. A model of the Hop2-Mnd1-DMC1-ssDNA ensemble is proposed to explain how it mediates homologous DNA pairing in meiotic recombination.


Assuntos
Proteínas de Ciclo Celular/química , Proteínas de Ligação a DNA/química , Recombinação Homóloga , Meiose/genética , Animais , Sítios de Ligação , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , DNA/metabolismo , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Camundongos , Mutação Puntual , Multimerização Proteica , Estrutura Terciária de Proteína , Recombinases/metabolismo
19.
Nucleic Acids Res ; 41(4): 2313-27, 2013 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-23303776

RESUMO

By coupling the protection and organization of single-stranded DNA (ssDNA) with recruitment and alignment of DNA processing factors, replication protein A (RPA) lies at the heart of dynamic multi-protein DNA processing machinery. Nevertheless, how RPA coordinates biochemical functions of its eight domains remains unknown. We examined the structural biochemistry of RPA's DNA-binding activity, combining small-angle X-ray and neutron scattering with all-atom molecular dynamics simulations to investigate the architecture of RPA's DNA-binding core. The scattering data reveal compaction promoted by DNA binding; DNA-free RPA exists in an ensemble of states with inter-domain mobility and becomes progressively more condensed and less dynamic on binding ssDNA. Our results contrast with previous models proposing RPA initially binds ssDNA in a condensed state and becomes more extended as it fully engages the substrate. Moreover, the consensus view that RPA engages ssDNA in initial, intermediate and final stages conflicts with our data revealing that RPA undergoes two (not three) transitions as it binds ssDNA with no evidence for a discrete intermediate state. These results form a framework for understanding how RPA integrates the ssDNA substrate into DNA processing machinery, provides substrate access to its binding partners and promotes the progression and selection of DNA processing pathways.


Assuntos
DNA de Cadeia Simples/química , Proteína de Replicação A/química , DNA de Cadeia Simples/metabolismo , Simulação de Dinâmica Molecular , Difração de Nêutrons , Ligação Proteica , Estrutura Terciária de Proteína , Proteína de Replicação A/metabolismo , Espalhamento a Baixo Ângulo , Difração de Raios X
20.
Nucleic Acids Res ; 40(13): 6070-81, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22492624

RESUMO

X-ray crystallography provides excellent structural data on protein-DNA interfaces, but crystallographic complexes typically contain only small fragments of large DNA molecules. We present a new approach that can use longer DNA substrates and reveal new protein-DNA interactions even in extensively studied systems. Our approach combines rigid-body computational docking with hydrogen/deuterium exchange mass spectrometry (DXMS). DXMS identifies solvent-exposed protein surfaces; docking is used to create a 3-dimensional model of the protein-DNA interaction. We investigated the enzyme uracil-DNA glycosylase (UNG), which detects and cleaves uracil from DNA. UNG was incubated with a 30 bp DNA fragment containing a single uracil, giving the complex with the abasic DNA product. Compared with free UNG, the UNG-DNA complex showed increased solvent protection at the UNG active site and at two regions outside the active site: residues 210-220 and 251-264. Computational docking also identified these two DNA-binding surfaces, but neither shows DNA contact in UNG-DNA crystallographic structures. Our results can be explained by separation of the two DNA strands on one side of the active site. These non-sequence-specific DNA-binding surfaces may aid local uracil search, contribute to binding the abasic DNA product and help present the DNA product to APE-1, the next enzyme on the DNA-repair pathway.


Assuntos
Proteínas de Ligação a DNA/química , DNA/química , Uracila-DNA Glicosidase/química , Algoritmos , Domínio Catalítico , Biologia Computacional/métodos , Cristalografia por Raios X , Medição da Troca de Deutério , Espectrometria de Massas/métodos , Modelos Moleculares
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa