Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
EMBO Rep ; 25(2): 876-901, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38177925

RESUMO

FANCJ, a DNA helicase linked to Fanconi anemia and frequently mutated in cancers, counteracts replication stress by dismantling unconventional DNA secondary structures (such as G-quadruplexes) that occur at the DNA replication fork in certain sequence contexts. However, how FANCJ is recruited to the replisome is unknown. Here, we report that FANCJ directly binds to AND-1 (the vertebrate ortholog of budding yeast Ctf4), a homo-trimeric protein adaptor that connects the CDC45/MCM2-7/GINS replicative DNA helicase with DNA polymerase α and several other factors at DNA replication forks. The interaction between FANCJ and AND-1 requires the integrity of an evolutionarily conserved Ctf4-interacting protein (CIP) box located between the FANCJ helicase motifs IV and V. Disruption of the CIP box significantly reduces FANCJ association with the replisome, causing enhanced DNA damage, decreased replication fork recovery and fork asymmetry in cells unchallenged or treated with Pyridostatin, a G-quadruplex-binder, or Mitomycin C, a DNA inter-strand cross-linking agent. Cancer-relevant FANCJ CIP box variants display reduced AND-1-binding and enhanced DNA damage, a finding that suggests their potential role in cancer predisposition.


Assuntos
DNA , Neoplasias , Humanos , DNA/química , Replicação do DNA , Instabilidade Genômica , Proteínas de Manutenção de Minicromossomo
2.
J Cell Physiol ; 236(8): 5664-5675, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-33432587

RESUMO

Warsaw breakage syndrome (WABS), is caused by biallelic mutations of DDX11, a gene coding a DNA helicase. We have recently reported two affected sisters, compound heterozygous for a missense (p.Leu836Pro) and a frameshift (p.Lys303Glufs*22) variant. By investigating the pathogenic mechanism, we demonstrate the inability of the DDX11 p.Leu836Pro mutant to unwind forked DNA substrates, while retaining DNA binding activity. We observed the accumulation of patient-derived cells at the G2/M phase and increased chromosomal fragmentation after mitomycin C treatment. The phenotype partially overlaps with features of the Fanconi anemia cells, which shows not only genomic instability but also defective mitochondria. This prompted us to examine mitochondrial functionality in WABS cells and revealed an altered aerobic metabolism. This opens the door to the further elucidation of the molecular and cellular basis of an impaired mitochondrial phenotype and sheds light on this fundamental process in cell physiology and the pathogenesis of these diseases.


Assuntos
DNA Helicases/genética , Anemia de Fanconi/genética , Instabilidade Genômica/genética , Síndrome de Kearns-Sayre/metabolismo , Miopatias Mitocondriais/metabolismo , Anormalidades Múltiplas/genética , RNA Helicases DEAD-box/genética , DNA Helicases/metabolismo , Anemia de Fanconi/metabolismo , Genômica , Humanos , Síndrome de Kearns-Sayre/genética , Miopatias Mitocondriais/genética , Mutação/genética
3.
J Biol Chem ; 294(11): 3947-3956, 2019 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-30655288

RESUMO

Human DNA polymerase δ is essential for DNA replication and acts in conjunction with the processivity factor proliferating cell nuclear antigen (PCNA). In addition to its catalytic subunit (p125), pol δ comprises three regulatory subunits (p50, p68, and p12). PCNA interacts with all of these subunits, but only the interaction with p68 has been structurally characterized. Here, we report solution NMR-, isothermal calorimetry-, and X-ray crystallography-based analyses of the p12-PCNA interaction, which takes part in the modulation of the rate and fidelity of DNA synthesis by pol δ. We show that p12 binds with micromolar affinity to the classical PIP-binding pocket of PCNA via a highly atypical PIP box located at the p12 N terminus. Unlike the canonical PIP box of p68, the PIP box of p12 lacks the conserved glutamine; binds through a 2-fork plug made of an isoleucine and a tyrosine residue at +3 and +8 positions, respectively; and is stabilized by an aspartate at +6 position, which creates a network of intramolecular hydrogen bonds. These findings add to growing evidence that PCNA can bind a diverse range of protein sequences that may be broadly grouped as PIP-like motifs as has been previously suggested.


Assuntos
DNA Polimerase III/metabolismo , Antígeno Nuclear de Célula em Proliferação/metabolismo , Motivos de Aminoácidos , Calorimetria , Domínio Catalítico , DNA Polimerase III/química , DNA Polimerase III/isolamento & purificação , Humanos , Modelos Moleculares , Ressonância Magnética Nuclear Biomolecular , Antígeno Nuclear de Célula em Proliferação/química , Antígeno Nuclear de Célula em Proliferação/isolamento & purificação
4.
Nucleic Acids Res ; 46(18): 9816-9828, 2018 10 12.
Artigo em Inglês | MEDLINE | ID: mdl-30102405

RESUMO

p15PAF is an oncogenic intrinsically disordered protein that regulates DNA replication and lesion bypass by interacting with the human sliding clamp PCNA. In the absence of DNA, p15PAF traverses the PCNA ring via an extended PIP-box that contacts the sliding surface. Here, we probed the atomic-scale structure of p15PAF-PCNA-DNA ternary complexes. Crystallography and MD simulations show that, when p15PAF occupies two subunits of the PCNA homotrimer, DNA within the ring channel binds the unoccupied subunit. The structure of PCNA-bound p15PAF in the absence and presence of DNA is invariant, and solution NMR confirms that DNA does not displace p15PAF from the ring wall. Thus, p15PAF reduces the available sliding surfaces of PCNA, and may function as a belt that fastens the DNA to the clamp during synthesis by the replicative polymerase (pol δ). This constraint, however, may need to be released for efficient DNA lesion bypass by the translesion synthesis polymerase (pol η). Accordingly, our biochemical data show that p15PAF impairs primer synthesis by pol η-PCNA holoenzyme against both damaged and normal DNA templates. In light of our findings, we discuss the possible mechanistic roles of p15PAF in DNA replication and suppression of DNA lesion bypass.


Assuntos
Proteínas de Transporte/química , DNA/química , Proteínas Intrinsicamente Desordenadas/química , Antígeno Nuclear de Célula em Proliferação/química , Proteínas de Transporte/genética , Cristalografia por Raios X , DNA/genética , DNA Polimerase III/química , DNA Polimerase III/genética , Replicação do DNA/genética , Proteínas de Ligação a DNA , DNA Polimerase Dirigida por DNA/química , DNA Polimerase Dirigida por DNA/genética , Escherichia coli/genética , Holoenzimas/química , Holoenzimas/genética , Humanos , Proteínas Intrinsicamente Desordenadas/genética , Espectroscopia de Ressonância Magnética , Complexos Multiproteicos/química , Complexos Multiproteicos/genética , Antígeno Nuclear de Célula em Proliferação/genética
5.
Int J Mol Sci ; 21(3)2020 Jan 31.
Artigo em Inglês | MEDLINE | ID: mdl-32023808

RESUMO

The human zinc transporter 8 (hZnT8) plays important roles in the storage of insulin in the secretory vesicles of pancreatic ß cells. hZnT8 consists of a transmembrane domain, with its N- and C-termini protruding into the cytoplasm. Interestingly, the exchange of arginine to tryptophan at position 325 in the C-terminal domain (CTD) increases the risk of developing type 2 diabetes mellitus (T2D). In the present study, the CTDs of hZnT8 (the wild-type (WT) and its disease risk variant (R325W)) were expressed, purified, and characterized in their native forms by biophysical techniques. The data reveal that the CTDs form tetramers which are stabilized by zinc binding, and exhibit negligible differences in their secondary structure content and zinc-binding affinities in solution. These findings provide the basis for conducting further structural studies aimed at unravelling the molecular mechanism underlying the increased susceptibility to develop T2D, which is modulated by the disease risk variant.


Assuntos
Substituição de Aminoácidos , Diabetes Mellitus Tipo 2/genética , Transportador 8 de Zinco/química , Transportador 8 de Zinco/metabolismo , Zinco/metabolismo , Arginina/metabolismo , Cristalografia por Raios X , Humanos , Modelos Moleculares , Domínios Proteicos , Multimerização Proteica , Estrutura Secundária de Proteína , Espalhamento a Baixo Ângulo , Triptofano/metabolismo , Difração de Raios X , Transportador 8 de Zinco/genética
6.
PLoS Comput Biol ; 14(8): e1006295, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-30071012

RESUMO

Several channels, ranging from TRP receptors to Gap junctions, allow the exchange of small organic solute across cell membrane. However, very little is known about the molecular mechanism of their permeation. Cyclic Nucleotide Gated (CNG) channels, despite their homology with K+ channels and in contrast with them, allow the passage of larger methylated and ethylated ammonium ions like dimethylammonium (DMA) and ethylammonium (EA). We combined electrophysiology and molecular dynamics simulations to examine how DMA interacts with the pore and permeates through it. Due to the presence of hydrophobic groups, DMA enters easily in the channel and, unlike the alkali cations, does not need to cross any barrier. We also show that while the crystal structure is consistent with the presence of a single DMA ion at full occupancy, the channel is able to conduct a sizable current of DMA ions only when two ions are present inside the channel. Moreover, the second DMA ion dramatically changes the free energy landscape, destabilizing the crystallographic binding site and lowering by almost 25 kJ/mol the binding affinity between DMA and the channel. Based on the results of the simulation the experimental electron density maps can be re-interpreted with the presence of a second ion at lower occupancy. In this mechanism the flexibility of the channel plays a key role, extending the classical multi-ion permeation paradigm in which conductance is enhanced by the plain interaction between the ions.


Assuntos
Canais de Cátion Regulados por Nucleotídeos Cíclicos/metabolismo , Proteínas de Transporte de Cátions Orgânicos/fisiologia , Animais , Fenômenos Biofísicos , Cátions/metabolismo , Simulação por Computador , Canais de Cátion Regulados por Nucleotídeos Cíclicos/fisiologia , Dimetilaminas/metabolismo , Junções Comunicantes/metabolismo , Potenciais da Membrana/fisiologia , Simulação de Dinâmica Molecular , Oócitos/fisiologia , Compostos de Amônio Quaternário/metabolismo , Sódio/metabolismo , Xenopus laevis
7.
J Biol Chem ; 292(10): 4176-4184, 2017 03 10.
Artigo em Inglês | MEDLINE | ID: mdl-27998982

RESUMO

RecQ helicases are essential in the maintenance of genome stability. Five paralogues (RecQ1, Bloom, Werner, RecQ4, and RecQ5) are found in human cells, with distinct but overlapping roles. Mutations in human RecQ4 give rise to three distinct genetic disorders (Rothmund-Thomson, RAPADILINO, and Baller-Gerold syndromes), characterized by genetic instability, growth deficiency, and predisposition to cancer. Previous studies suggested that RecQ4 was unique because it did not seem to contain a RecQ C-terminal region (RQC) found in the other RecQ paralogues; such a region consists of a zinc domain and a winged helix domain and plays an important role in enzyme activity. However, our recent bioinformatic analysis identified in RecQ4 a putative RQC. To experimentally confirm this hypothesis, we report the purification and characterization of the catalytic core of human RecQ4. Inductively coupled plasma-atomic emission spectrometry detected the unusual presence of two zinc clusters within the zinc domain, consistent with the bioinformatic prediction. Analysis of site-directed mutants, targeting key RQC residues (putative zinc ligands and the aromatic residue predicted to be at the tip of the winged helix ß-hairpin), showed a decrease in DNA binding, unwinding, and annealing, as expected for a functional RQC domain. Low resolution structural information obtained by small angle X-ray scattering data suggests that RecQ4 interacts with DNA in a manner similar to RecQ1, whereas the winged helix domain may assume alternative conformations, as seen in the bacterial enzymes. These combined results experimentally confirm the presence of a functional RQC domain in human RecQ4.


Assuntos
DNA/metabolismo , Proteínas Mutantes/química , Proteínas Mutantes/metabolismo , RecQ Helicases/química , RecQ Helicases/metabolismo , Adenosina Trifosfatases/metabolismo , Biologia Computacional , Cristalografia por Raios X , DNA/química , DNA/genética , Bases de Dados de Proteínas , Humanos , Mutagênese Sítio-Dirigida , Proteínas Mutantes/genética , Mutação/genética , Ligação Proteica , Domínios Proteicos , Estrutura Terciária de Proteína , RecQ Helicases/genética
8.
Proc Natl Acad Sci U S A ; 112(27): E3619-28, 2015 Jul 07.
Artigo em Inglês | MEDLINE | ID: mdl-26100907

RESUMO

Cyclic nucleotide-gated (CNG) ion channels, despite a significant homology with the highly selective K(+) channels, do not discriminate among monovalent alkali cations and are permeable also to several organic cations. We combined electrophysiology, molecular dynamics (MD) simulations, and X-ray crystallography to demonstrate that the pore of CNG channels is highly flexible. When a CNG mimic is crystallized in the presence of a variety of monovalent cations, including Na(+), Cs(+), and dimethylammonium (DMA(+)), the side chain of Glu66 in the selectivity filter shows multiple conformations and the diameter of the pore changes significantly. MD simulations indicate that Glu66 and the prolines in the outer vestibule undergo large fluctuations, which are modulated by the ionic species and the voltage. This flexibility underlies the coupling between gating and permeation and the poor ionic selectivity of CNG channels.


Assuntos
Canais de Cátion Regulados por Nucleotídeos Cíclicos/química , Canais de Cátion Regulados por Nucleotídeos Cíclicos/metabolismo , Ativação do Canal Iônico/fisiologia , Conformação Proteica , Sequência de Aminoácidos , Animais , Proteínas de Bactérias/química , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Cátions Monovalentes/metabolismo , Bovinos , Césio/metabolismo , Cristalografia por Raios X , Canais de Cátion Regulados por Nucleotídeos Cíclicos/genética , Feminino , Ativação do Canal Iônico/genética , Transporte de Íons/efeitos dos fármacos , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/fisiologia , Simulação de Dinâmica Molecular , Dados de Sequência Molecular , Proteínas Mutantes/química , Proteínas Mutantes/metabolismo , Mutação de Sentido Incorreto , Oócitos/metabolismo , Oócitos/fisiologia , Técnicas de Patch-Clamp , Homologia de Sequência de Aminoácidos , Sódio/metabolismo , Xenopus laevis
9.
Chromosoma ; 122(1-2): 47-53, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23412083

RESUMO

The CMG (Cdc45-MCM-GINS) complex is the eukaryotic replicative helicase, the enzyme that unwinds double-stranded DNA at replication forks. All three components of the CMG complex are essential for its function, but only in the case of MCM, the molecular motor that harnesses the energy of ATP hydrolysis to catalyse strand separation, is that function clear. Here, we review current knowledge of the three-dimensional structure of the CMG complex and its components and highlight recent advances in our understanding of its evolutionary origins.


Assuntos
Proteínas de Ciclo Celular/química , DNA Helicases/química , Replicação do DNA/genética , Proteínas de Ligação a DNA/química , Proteína 1 de Manutenção de Minicromossomo/química , Proteínas de Ciclo Celular/genética , Proteínas Cromossômicas não Histona/química , Proteínas Cromossômicas não Histona/genética , DNA/genética , DNA Helicases/genética , Proteínas de Ligação a DNA/genética , Evolução Molecular , Humanos , Proteína 1 de Manutenção de Minicromossomo/genética , Conformação Proteica
10.
J Biol Chem ; 287(6): 4121-8, 2012 Feb 03.
Artigo em Inglês | MEDLINE | ID: mdl-22147708

RESUMO

Cdc45 is an essential protein conserved in all eukaryotes and is involved both in the initiation of DNA replication and the progression of the replication fork. With GINS, Cdc45 is an essential cofactor of the Mcm2-7 replicative helicase complex. Despite its importance, no detailed information is available on either the structure or the biochemistry of the protein. Intriguingly, whereas homologues of both GINS and Mcm proteins have been described in Archaea, no counterpart for Cdc45 is known. Herein we report a bioinformatic analysis that shows a weak but significant relationship among eukaryotic Cdc45 proteins and a large family of phosphoesterases that has been described as the DHH family, including inorganic pyrophosphatases and RecJ ssDNA exonucleases. These enzymes catalyze the hydrolysis of phosphodiester bonds via a mechanism involving two Mn(2+) ions. Only a subset of the amino acids that coordinates Mn(2+) is conserved in Cdc45. We report biochemical and structural data on the recombinant human Cdc45 protein, consistent with the proposed DHH family affiliation. Like the RecJ exonucleases, the human Cdc45 protein is able to bind single-stranded, but not double-stranded DNA. Small angle x-ray scattering data are consistent with a model compatible with the crystallographic structure of the RecJ/DHH family members.


Assuntos
Proteínas de Bactérias/genética , Proteínas de Ciclo Celular/genética , Replicação do DNA/fisiologia , Evolução Molecular , Exodesoxirribonucleases/genética , Modelos Moleculares , Diester Fosfórico Hidrolases/genética , Proteínas de Bactérias/química , Proteínas de Bactérias/metabolismo , Catálise , Proteínas de Ciclo Celular/química , Proteínas de Ciclo Celular/metabolismo , DNA de Cadeia Simples/química , DNA de Cadeia Simples/genética , DNA de Cadeia Simples/metabolismo , Exodesoxirribonucleases/química , Exodesoxirribonucleases/metabolismo , Humanos , Manganês/química , Manganês/metabolismo , Diester Fosfórico Hidrolases/química , Diester Fosfórico Hidrolases/metabolismo , Homologia de Sequência de Aminoácidos , Relação Estrutura-Atividade
11.
Adv Exp Med Biol ; 767: 75-95, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23161007

RESUMO

Hexameric helicases are responsible for many biological processes, ranging from DNA replication in various life domains to DNA repair, transcriptional regulation and RNA metabolism, and encompass superfamilies 3-6 (SF3-6).To harness the chemical energy from ATP hydrolysis for mechanical work, hexameric helicases have a conserved core engine, called ASCE, that belongs to a subdivision of the P-loop NTPases. Some of the ring helicases (SF4 and SF5) use a variant of ASCE known as RecA-like, while some (SF3 and SF6) use another variant known as AAA+ fold. The NTP-binding sites are located at the interface between monomers and include amino-acid residues coming from neighbouring subunits, providing a mean for small structural changes within the ATP-binding site to be amplified into large inter-subunit movement.The ring structure has a central channel which encircles the nucleic acid. The topological link between the protein and the nucleic acid substrate increases the stability and processivity of the enzyme. This is probably the reason why within cellular systems the critical step of unwinding dsDNA ahead of the replication fork seems to be almost invariably carried out by a toroidal helicase, whether in bacteria, archaea or eukaryotes, as well as in some viruses.Over the last few years, a large number of biochemical, biophysical and structural data have thrown new light onto the architecture and function of these remarkable machines. Although the evidence is still limited to a couple of systems, biochemical and structural results suggest that motors based on RecA and AAA+ folds have converged on similar mechanisms to couple ATP-driven conformational changes to movement along nucleic acids.


Assuntos
DNA Helicases , Replicação do DNA , Trifosfato de Adenosina/metabolismo , Sítios de Ligação , DNA/química , DNA Helicases/química , Nucleosídeo-Trifosfatase , Estrutura Terciária de Proteína
12.
Sci Rep ; 13(1): 12610, 2023 08 03.
Artigo em Inglês | MEDLINE | ID: mdl-37537265

RESUMO

Despite numerous reports on the interactions of G-quadruplexes (G4s) with helicases, systematic analysis addressing the selectivity and specificity of each helicase towards a variety of G4 topologies are scarce. Among the helicases able to unwind G4s are those containing an iron-sulphur (FeS) cluster, including both the bacterial DinG (found in E. coli and several pathogenic bacteria) and the medically important eukaryotic homologues (XPD, FancJ, DDX11 and RTEL1). We carried out a detailed study of the interactions between the E. coli DinG and a variety of G4s, by employing physicochemical and biochemical methodologies. A series of G4-rich sequences from different genomic locations (promoter and telomeric regions), able to form unimolecular G4 structures with diverse topologies, were analyzed (c-KIT1, KRAS, c-MYC, BCL2, Tel23, T30695, Zic1). DinG binds to most of the investigated G4s with little discrimination, while it exhibits a clear degree of unwinding specificity towards different G4 topologies. Whereas previous reports suggested that DinG was active only on bimolecular G4s, here we show that it is also able to bind to and resolve the more physiologically relevant unimolecular G4s. In addition, when the G4 structures were stabilized by ligands (Pyridostatin, PhenDC3, BRACO-19 or Netropsin), the DinG unwinding activity decreased and in most cases was abolished, with a pattern that is not simply explained by a change in binding affinity. Overall, these results have important implications for the biochemistry of helicases, strongly suggesting that when analysing the G4 unwinding property of an enzyme, it is necessary to investigate a variety of G4 substrates.


Assuntos
Escherichia coli , Quadruplex G , Regiões Promotoras Genéticas
13.
EMBO J ; 27(16): 2250-8, 2008 Aug 20.
Artigo em Inglês | MEDLINE | ID: mdl-18650940

RESUMO

The eukaryotic MCM2-7 complex is recruited at origins of replication during the G1 phase and acts as the main helicase at the replication fork during the S phase of the cell cycle. To characterize the interplay between the MCM helicase and DNA prior to the melting of the double helix, we determined the structure of an archaeal MCM orthologue bound to a 5.6-kb double-stranded DNA segment, using cryo-electron microscopy. DNA wraps around the N-terminal face of a single hexameric ring. This interaction requires a conformational change within the outer belt of the MCM N-terminal domain, exposing a previously unrecognized helix-turn-helix DNA-binding motif. Our findings provide novel insights into the role of the MCM complex during the initiation step of DNA replication.


Assuntos
Microscopia Crioeletrônica , DNA Helicases/química , DNA Helicases/ultraestrutura , DNA/ultraestrutura , Methanobacteriaceae/enzimologia , Sítios de Ligação , Modelos Moleculares , Proteínas Mutantes/ultraestrutura , Ligação Proteica , Estrutura Terciária de Proteína
14.
Crit Rev Biochem Mol Biol ; 44(5): 326-42, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19780640

RESUMO

The eukaryotic MCM2-7 complex is recruited onto origins of replication during the G1 phase of the cell cycle and acts as the main helicase at the replication fork during the S phase. Over the last few years a number of structural reports on MCM proteins using both electron microscopy and protein crystallography have been published. The crystal structures of two (almost) full-length archaeal homologs provide the first atomic pictures of a MCM helicase. However one of the structures is at low resolution and the other is of an inactive MCM. Moreover, both proteins are monomeric in the crystal, whereas the activity of the complex is critically dependent on oligomerization. Lower resolution structures derived from electron microscopy studies are therefore crucial to complement the crystallographic analysis and to assemble the multimeric complex that is active in the cell. A critical analysis of all the structural results elucidates the potential conformational changes and dynamic behavior of MCM helicase to provide a first insight into the gamut of molecular configurations adopted during the processes of DNA melting and unwinding.


Assuntos
Ciclo Celular , DNA Helicases/química , DNA Helicases/metabolismo , Proteínas de Domínio MADS/química , Proteínas de Domínio MADS/metabolismo , Animais , DNA/química , DNA/metabolismo , DNA Helicases/genética , Humanos , Ligação Proteica , Conformação Proteica
15.
Structure ; 17(2): 211-22, 2009 Feb 13.
Artigo em Inglês | MEDLINE | ID: mdl-19217392

RESUMO

The minichromosome maintenance (MCM) proteins, members of the AAA+ (ATPase associated with diverse cellular activities) superfamily, are believed to constitute the replicative helicase in eukaryotic and archaeal species. Here, we present the 1.9 A resolution crystal structure of a monomeric MCM homolog from Methanopyrus kandleri, the first crystallographic structure of a full-length MCM. We also present an 18 A cryo-electron microscopy reconstruction of the hexameric MCM from Methanothermobacter thermautotrophicus, and fit the atomic resolution crystal structure into the reconstruction in order to generate an atomic model for the oligomeric assembly. These structural data reveal a distinct active site topology consisting of a unique arrangement of critical determinants. The structures also provide a molecular framework for understanding the functional contributions of trans-acting elements that facilitate intersubunit crosstalk in response to DNA binding and ATP hydrolysis.


Assuntos
Proteínas Arqueais/química , DNA Helicases/química , Sequência de Aminoácidos , Animais , Archaea/enzimologia , Proteínas Arqueais/metabolismo , Domínio Catalítico , DNA Helicases/metabolismo , Replicação do DNA/fisiologia , Células Eucarióticas/enzimologia , Humanos , Componente 2 do Complexo de Manutenção de Minicromossomo , Modelos Moleculares , Dados de Sequência Molecular , Proteínas Nucleares/química , Proteínas Nucleares/metabolismo , Conformação Proteica , Multimerização Proteica , Homologia de Sequência de Aminoácidos
16.
Materials (Basel) ; 14(3)2021 Feb 02.
Artigo em Inglês | MEDLINE | ID: mdl-33540751

RESUMO

The MCM (minichromosome maintenance) protein complex forms an hexameric ring and has a key role in the replication machinery of Eukaryotes and Archaea, where it functions as the replicative helicase opening up the DNA double helix ahead of the polymerases. Here, we present a study of the interaction between DNA and the archaeal MCM complex from Methanothermobacter thermautotrophicus by means of atomic force microscopy (AFM) single molecule imaging. We first optimized the protocol (surface treatment and buffer conditions) to obtain AFM images of surface-equilibrated DNA molecules before and after the interaction with the protein complex. We discriminated between two modes of interaction, one in which the protein induces a sharp bend in the DNA, and one where there is no bending. We found that the presence of the MCM complex also affects the DNA contour length. A possible interpretation of the observed behavior is that in one case the hexameric ring encircles the dsDNA, while in the other the nucleic acid wraps on the outside of the ring, undergoing a change of direction. We confirmed this topographical assignment by testing two mutants, one affecting the N-terminal ß-hairpins projecting towards the central channel, and thus preventing DNA loading, the other lacking an external subdomain and thus preventing wrapping. The statistical analysis of the distribution of the protein complexes between the two modes, together with the dissection of the changes of DNA contour length and binding angle upon interaction, for the wild type and the two mutants, is consistent with the hypothesis. We discuss the results in view of the various modes of nucleic acid interactions that have been proposed for both archaeal and eukaryotic MCM complexes.

17.
Mol Genet Genomic Med ; 7(5): e639, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30924321

RESUMO

BACKGROUND: Warsaw Breakage Syndrome (WABS) is an ultra rare cohesinopathy caused by biallelic mutation of DDX11 gene. It is clinically characterized by pre and postnatal growth delay, microcephaly, hearing loss with cochlear hypoplasia, skin color abnormalities, and dysmorphisms. METHODS: Mutational screening and functional analyses (protein expression and 3D-modeling) were performed in order to investigate the presence and pathogenicity of DDX11 variant identified in our patients. RESULTS: We report the clinical history of two sisters affected by WABS with a pathological mytomicin C test carrying compound heterozygous mutations (c.2507T > C / c.907_920del) of the DDX11 gene. The pathogenicity of this variant was confirmed in the light of a bioinformatic study and protein three-dimensional modeling, as well as expression analysis. CONCLUSION: These findings further extend the clinical and molecular knowledge about the WABS showing a possible mild phenotype without major malformations or intellectual disability.


Assuntos
Anormalidades Múltiplas/genética , Manchas Café com Leite/genética , RNA Helicases DEAD-box/genética , DNA Helicases/genética , Perda Auditiva Neurossensorial/genética , Fenótipo , Anormalidades Múltiplas/patologia , Manchas Café com Leite/patologia , Linhagem Celular , Células Cultivadas , Criança , Pré-Escolar , Feminino , Perda Auditiva Neurossensorial/patologia , Humanos , Mutação , Síndrome
18.
Cells ; 8(3)2019 03 16.
Artigo em Inglês | MEDLINE | ID: mdl-30884854

RESUMO

Members of the tripartite motif family of E3 ubiquitin ligases are characterized by the presence of a conserved N-terminal module composed of a RING domain followed by one or two B-box domains, a coiled-coil and a variable C-terminal region. The RING and B-box are both Zn-binding domains but, while the RING is found in a large number of proteins, the B-box is exclusive to the tripartite motif (TRIM) family members in metazoans. Whereas the RING has been extensively characterized and shown to possess intrinsic E3 ligase catalytic activity, much less is known about the role of the B-box domains. In this study, we adopted an in vitro approach using recombinant point- and deletion-mutants to characterize the contribution of the TRIM32 Zn-binding domains to the activity of this E3 ligase that is altered in a genetic form of muscular dystrophy. We found that the RING domain is crucial for E3 ligase activity and E2 specificity, whereas a complete B-box domain is involved in chain assembly rate modulation. Further, in vitro, the RING domain is necessary to modulate TRIM32 oligomerization, whereas, in cells, both the RING and B-box cooperate to specify TRIM32 subcellular localization, which if altered may impact the pathogenesis of diseases.


Assuntos
Distrofia Muscular do Cíngulo dos Membros/genética , Mutação/genética , Fatores de Transcrição/química , Fatores de Transcrição/genética , Proteínas com Motivo Tripartido/química , Proteínas com Motivo Tripartido/genética , Ubiquitina-Proteína Ligases/química , Ubiquitina-Proteína Ligases/genética , Zinco/metabolismo , Animais , Biocatálise , Linhagem Celular , Humanos , Camundongos , Proteínas Mutantes/metabolismo , Domínios Proteicos , Multimerização Proteica , Fatores de Transcrição/metabolismo , Proteínas com Motivo Tripartido/metabolismo , Ubiquitina-Proteína Ligases/metabolismo
19.
Nucleic Acids Res ; 34(20): 5829-38, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-17062628

RESUMO

The MCM complex from the archaeon Methanother-mobacter thermautotrophicus is a model for the eukaryotic MCM2-7 helicase. We present electron-microscopy single-particle reconstructions of a DNA treated M.thermautotrophicus MCM sample and a ADP.AlF(x) treated sample, respectively assembling as double hexamers and double heptamers. The electron-density maps display an unexpected asymmetry between the two rings, suggesting that large conformational changes can occur within the complex. The structure of the MCM N-terminal domain, as well as the AAA+ and the C-terminal HTH dom-ains of ZraR can be fitted into the reconstructions. Distinct configurations can be modelled for the AAA+ and the HTH domains, suggesting the nature of the conformational change within the complex. The pre-sensor 1 and the helix 2 insertions, important for the activity, can be located pointing towards the centre of the channel in the presence of DNA. We propose a mechanistic model for the helicase activity, based on a ligand-controlled rotation of the AAA+ subunits.


Assuntos
Proteínas Arqueais/química , DNA Helicases/química , Methanobacteriaceae/enzimologia , Modelos Moleculares , Adenosina Trifosfatases/química , Proteínas Arqueais/metabolismo , Proteínas Arqueais/ultraestrutura , DNA/química , DNA Helicases/metabolismo , DNA Helicases/ultraestrutura , Processamento de Imagem Assistida por Computador , Estrutura Terciária de Proteína
20.
Genes (Basel) ; 9(11)2018 Nov 21.
Artigo em Inglês | MEDLINE | ID: mdl-30469382

RESUMO

DDX11/ChlR1 (Chl1 in yeast) is a DNA helicase involved in sister chromatid cohesion and in DNA repair pathways. The protein belongs to the family of the iron⁻sulphur cluster containing DNA helicases, whose deficiencies have been linked to a number of diseases affecting genome stability. Mutations of human DDX11 are indeed associated with the rare genetic disorder named Warsaw breakage syndrome, showing both chromosomal breakages and chromatid cohesion defects. Moreover, growing evidence of a potential role in oncogenesis further emphasizes the clinical relevance of DDX11. Here, we illustrate the biochemical and structural features of DDX11 and how it cooperates with multiple protein partners in the cell, acting at the interface of DNA replication/repair/recombination and sister chromatid cohesion to preserve genome stability.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA