Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
Bioinformatics ; 31(7): 1147-9, 2015 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-25433696

RESUMO

UNLABELLED: LigDig is a web server designed to answer questions that previously required several independent queries to diverse data sources. It also performs basic manipulations and analyses of the structures of protein-ligand complexes. The LigDig webserver is modular in design and consists of seven tools, which can be used separately, or via linking the output from one tool to the next, in order to answer more complex questions. Currently, the tools allow a user to: (i) perform a free-text compound search, (ii) search for suitable ligands, particularly inhibitors, of a protein and query their interaction network, (iii) search for the likely function of a ligand, (iv) perform a batch search for compound identifiers, (v) find structures of protein-ligand complexes, (vi) compare three-dimensional structures of ligand binding sites and (vii) prepare coordinate files of protein-ligand complexes for further calculations. AVAILABILITY AND IMPLEMENTATION: LigDig makes use of freely available databases, including ChEMBL, PubChem and SABIO-RK, and software programs, including cytoscape.js, PDB2PQR, ProBiS and Fconv. LigDig can be used by non-experts in bio- and chemoinformatics. LigDig is available at: http://mcm.h-its.org/ligdig. SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Assuntos
Internet , Proteínas/química , Proteínas/metabolismo , Software , Sítios de Ligação , Bases de Dados Factuais , Frutosedifosfatos/metabolismo , Humanos , Ligantes
2.
PLoS Comput Biol ; 9(7): e1003159, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23946717

RESUMO

Pyruvate kinase (PYK) is a critical allosterically regulated enzyme that links glycolysis, the primary energy metabolism, to cellular metabolism. Lactic acid bacteria rely almost exclusively on glycolysis for their energy production under anaerobic conditions, which reinforces the key role of PYK in their metabolism. These organisms are closely related, but have adapted to a huge variety of native environments. They include food-fermenting organisms, important symbionts in the human gut, and antibiotic-resistant pathogens. In contrast to the rather conserved inhibition of PYK by inorganic phosphate, the activation of PYK shows high variability in the type of activating compound between different lactic acid bacteria. System-wide comparative studies of the metabolism of lactic acid bacteria are required to understand the reasons for the diversity of these closely related microorganisms. These require knowledge of the identities of the enzyme modifiers. Here, we predict potential allosteric activators of PYKs from three lactic acid bacteria which are adapted to different native environments. We used protein structure-based molecular modeling and enzyme kinetic modeling to predict and validate potential activators of PYK. Specifically, we compared the electrostatic potential and the binding of phosphate moieties at the allosteric binding sites, and predicted potential allosteric activators by docking. We then made a kinetic model of Lactococcus lactis PYK to relate the activator predictions to the intracellular sugar-phosphate conditions in lactic acid bacteria. This strategy enabled us to predict fructose 1,6-bisphosphate as the sole activator of the Enterococcus faecalis PYK, and to predict that the PYKs from Streptococcus pyogenes and Lactobacillus plantarum show weaker specificity for their allosteric activators, while still having fructose 1,6-bisphosphate play the main activator role in vivo. These differences in the specificity of allosteric activation may reflect adaptation to different environments with different concentrations of activating compounds. The combined computational approach employed can readily be applied to other enzymes.


Assuntos
Lactobacillus/metabolismo , Piruvato Quinase/metabolismo , Regulação Alostérica , Sequência de Aminoácidos , Dados de Sequência Molecular , Piruvato Quinase/química , Homologia de Sequência de Aminoácidos
3.
Proc Natl Acad Sci U S A ; 108(34): E542-9, 2011 Aug 23.
Artigo em Inglês | MEDLINE | ID: mdl-21795601

RESUMO

Human thymidylate synthase is a homodimeric enzyme that plays a key role in DNA synthesis and is a target for several clinically important anticancer drugs that bind to its active site. We have designed peptides to specifically target its dimer interface. Here we show through X-ray diffraction, spectroscopic, kinetic, and calorimetric evidence that the peptides do indeed bind at the interface of the dimeric protein and stabilize its di-inactive form. The "LR" peptide binds at a previously unknown binding site and shows a previously undescribed mechanism for the allosteric inhibition of a homodimeric enzyme. It inhibits the intracellular enzyme in ovarian cancer cells and reduces cellular growth at low micromolar concentrations in both cisplatin-sensitive and -resistant cells without causing protein overexpression. This peptide demonstrates the potential of allosteric inhibition of hTS for overcoming platinum drug resistance in ovarian cancer.


Assuntos
Inibidores Enzimáticos/farmacologia , Terapia de Alvo Molecular , Neoplasias Ovarianas/enzimologia , Peptídeos/metabolismo , Peptídeos/farmacologia , Timidilato Sintase/antagonistas & inibidores , Sequência de Aminoácidos , Sítios de Ligação , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Cisplatino/farmacologia , Cisplatino/uso terapêutico , Cristalografia por Raios X , Desenho de Fármacos , Inibidores Enzimáticos/química , Inibidores Enzimáticos/metabolismo , Inibidores Enzimáticos/uso terapêutico , Feminino , Fluoruracila/farmacologia , Fluoruracila/uso terapêutico , Humanos , Cinética , Modelos Moleculares , Dados de Sequência Molecular , Neoplasias Ovarianas/tratamento farmacológico , Neoplasias Ovarianas/patologia , Peptídeos/química , Peptídeos/uso terapêutico , Ligação Proteica/efeitos dos fármacos , Conformação Proteica , Multimerização Proteica/efeitos dos fármacos , Termodinâmica , Timidilato Sintase/química , Timidilato Sintase/metabolismo
4.
J Chem Inf Model ; 53(5): 1235-52, 2013 May 24.
Artigo em Inglês | MEDLINE | ID: mdl-23621586

RESUMO

We present TRAPP (TRAnsient Pockets in Proteins), a new automated software platform for tracking, analysis, and visualization of binding pocket variations along a protein motion trajectory or within an ensemble of protein structures that may encompass conformational changes ranging from local side chain fluctuations to global backbone motions. TRAPP performs accurate grid-based calculations of the shape and physicochemical characteristics of a binding pocket for each structure and detects the conserved and transient regions of the pocket in an ensemble of protein conformations. It also provides tools for tracing the opening of a particular subpocket and residues that contribute to the binding site. TRAPP thus enables an assessment of the druggability of a disease-related target protein taking its flexibility into account.


Assuntos
Biologia Computacional/métodos , Proteínas/química , Proteínas/metabolismo , Software , Sítios de Ligação , Ligantes , Simulação de Dinâmica Molecular , Análise de Componente Principal
5.
Elife ; 112022 12 07.
Artigo em Inglês | MEDLINE | ID: mdl-36475542

RESUMO

Drugs that target human thymidylate synthase (hTS), a dimeric enzyme, are widely used in anticancer therapy. However, treatment with classical substrate-site-directed TS inhibitors induces over-expression of this protein and development of drug resistance. We thus pursued an alternative strategy that led us to the discovery of TS-dimer destabilizers. These compounds bind at the monomer-monomer interface and shift the dimerization equilibrium of both the recombinant and the intracellular protein toward the inactive monomers. A structural, spectroscopic, and kinetic investigation has provided evidence and quantitative information on the effects of the interaction of these small molecules with hTS. Focusing on the best among them, E7, we have shown that it inhibits hTS in cancer cells and accelerates its proteasomal degradation, thus causing a decrease in the enzyme intracellular level. E7 also showed a superior anticancer profile to fluorouracil in a mouse model of human pancreatic and ovarian cancer. Thus, over sixty years after the discovery of the first TS prodrug inhibitor, fluorouracil, E7 breaks the link between TS inhibition and enhanced expression in response, providing a strategy to fight drug-resistant cancers.


Assuntos
Neoplasias Ovarianas , Timidilato Sintase , Feminino , Animais , Camundongos , Humanos , Sítios de Ligação , Timidilato Sintase/química , Timidilato Sintase/metabolismo , Fluoruracila/farmacologia , Neoplasias Ovarianas/tratamento farmacológico , Inibidores Enzimáticos/farmacologia
6.
Proteins ; 78(1): 135-53, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19768680

RESUMO

A major challenge in drug design is to obtain compounds that bind selectively to their target receptors and do not cause side-effects by binding to other similar receptors. Here, we investigate strategies for applying COMBINE (COMparative BINding Energy) analysis, in conjunction with PIPSA (Protein Interaction Property Similarity Analysis) and ligand docking methods, to address this problem. We evaluate these approaches by application to diverse sets of inhibitors of three structurally related serine proteases of medical relevance: thrombin, trypsin, and urokinase-type plasminogen activator (uPA). We generated target-specific scoring functions (COMBINE models) for the three targets using training sets of ligands with known inhibition constants and structures of their receptor-ligand complexes. These COMBINE models were compared with the PIPSA results and experimental data on receptor selectivity. These scoring functions highlight the ligand-receptor interactions that are particularly important for binding specificity for the different targets. To predict target selectivity in virtual screening, compounds were docked into the three protein binding sites using the program GOLD and the docking solutions were re-ranked with the target-specific scoring functions and computed electrostatic binding free energies. Limits in the accuracy of some of the docking solutions and difficulties in scoring them adversely affected the predictive ability of the target specific scoring functions. Nevertheless, the target-specific scoring functions enabled the selectivity of ligands to thrombin versus trypsin and uPA to be predicted.


Assuntos
Serina Proteases/química , Serina Proteases/metabolismo , Algoritmos , Sequência de Aminoácidos , Humanos , Ligantes , Modelos Moleculares , Dados de Sequência Molecular , Ligação Proteica , Relação Quantitativa Estrutura-Atividade , Alinhamento de Sequência , Termodinâmica , Trombina/química , Trombina/metabolismo , Tripsina/química , Tripsina/metabolismo , Ativador de Plasminogênio Tipo Uroquinase/química , Ativador de Plasminogênio Tipo Uroquinase/metabolismo
7.
J Mol Recognit ; 23(2): 209-19, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-19746440

RESUMO

Given the three-dimensional structure of a protein, how can one find the sites where other molecules might bind to it? Do these sites have the properties necessary for high affinity binding? Is this protein a suitable target for drug design? Here, we discuss recent developments in computational methods to address these and related questions. Geometric methods to identify pockets on protein surfaces have been developed over many years but, with new algorithms, their performance is still improving. Simulation methods show promise in accounting for protein conformational variability to identify transient pockets but lack the ease of use of many of the (rigid) shape-based tools. Sequence and structure comparison approaches are benefiting from the constantly increasing size of sequence and structure databases. Energetic methods can aid identification and characterization of binding pockets, and have undergone recent improvements in the treatment of solvation and hydrophobicity. The "druggability" of a binding site is still difficult to predict with an automated procedure. The methodologies available for this purpose range from simple shape and hydrophobicity scores to computationally demanding free energy simulations.


Assuntos
Desenho de Fármacos , Ligantes , Conformação Proteica , Proteínas/química , Sequência de Aminoácidos , Sítios de Ligação , Bases de Dados de Proteínas , Modelos Moleculares , Dados de Sequência Molecular , Ligação Proteica , Alinhamento de Sequência
8.
J Mol Biol ; 345(2): 211-27, 2005 Jan 14.
Artigo em Inglês | MEDLINE | ID: mdl-15571716

RESUMO

In eukaryotes, many secreted proteins and peptide hormones are excised from larger precursors by calcium-dependent serine proteinases, the proprotein/prohormone convertases (PCs). These PCs cleave their protein substrates very specifically following multiple basic residues. The seven mammalian PCs and their yeast orthologue kexin are multi-domain proteinases consisting of a subtilisin-related catalytic domain, a conserved P-domain and a variable, often cysteine-rich domain, which in some PCs is followed by an additional C-terminal trans-membrane domain and a short cytoplasmic domain. The recently published crystal structures of the soluble mouse furin and yeast kexin ectodomains have revealed the relative arrangement of catalytic and P domains, the exact domain fold and the detailed architecture of the substrate binding clefts. Based on these experimental structures, we now have modelled the structures of the other human/mouse PCs. According to topology and to structure-based sequence comparisons, these other PCs closely resemble furin, with PC4, PACE4 and PC5/6 being more similar, and PC1/3, PC2 and PC7 being less similar to furin. Except for PC1 and PC2, this order of similarity is valid for the catalytic as well as for the P domains, and is almost reversed using kexin as a reference molecule. A similar order results from the number and clustering of negative charges lining the non-prime subsites, explaining the gradually decreasing requirement for basic residues N-terminal to substrate cleavage sites. The preference of the different PCs for distinct substrates seems to be governed by overall charge compensation and matching of the detailed charge distribution pattern.


Assuntos
Furina/química , Pró-Proteína Convertases/química , Proteínas de Saccharomyces cerevisiae/química , Sequência de Aminoácidos , Animais , Sítios de Ligação , Domínio Catalítico , Cristalografia por Raios X , Cisteína/química , Citoplasma/metabolismo , Glicina/química , Humanos , Camundongos , Modelos Moleculares , Dados de Sequência Molecular , Prolina/química , Pró-Proteína Convertases/metabolismo , Ligação Proteica , Conformação Proteica , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Saccharomyces cerevisiae/metabolismo , Homologia de Sequência de Aminoácidos , Especificidade por Substrato
9.
J Med Chem ; 59(16): 7598-616, 2016 08 25.
Artigo em Inglês | MEDLINE | ID: mdl-27411733

RESUMO

Flavonoids represent a potential source of new antitrypanosomatidic leads. Starting from a library of natural products, we combined target-based screening on pteridine reductase 1 with phenotypic screening on Trypanosoma brucei for hit identification. Flavonols were identified as hits, and a library of 16 derivatives was synthesized. Twelve compounds showed EC50 values against T. brucei below 10 µM. Four X-ray crystal structures and docking studies explained the observed structure-activity relationships. Compound 2 (3,6-dihydroxy-2-(3-hydroxyphenyl)-4H-chromen-4-one) was selected for pharmacokinetic studies. Encapsulation of compound 2 in PLGA nanoparticles or cyclodextrins resulted in lower in vitro toxicity when compared to the free compound. Combination studies with methotrexate revealed that compound 13 (3-hydroxy-6-methoxy-2-(4-methoxyphenyl)-4H-chromen-4-one) has the highest synergistic effect at concentration of 1.3 µM, 11.7-fold dose reduction index and no toxicity toward host cells. Our results provide the basis for further chemical modifications aimed at identifying novel antitrypanosomatidic agents showing higher potency toward PTR1 and increased metabolic stability.


Assuntos
Produtos Biológicos/farmacologia , Flavonóis/farmacologia , Tripanossomicidas/farmacologia , Trypanosoma brucei brucei/efeitos dos fármacos , Animais , Produtos Biológicos/síntese química , Produtos Biológicos/química , Linhagem Celular , Relação Dose-Resposta a Droga , Flavonóis/síntese química , Flavonóis/química , Humanos , Macrófagos/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos BALB C , Modelos Moleculares , Estrutura Molecular , Testes de Sensibilidade Parasitária , Relação Estrutura-Atividade , Tripanossomicidas/síntese química , Tripanossomicidas/química
10.
Drug Discov Today Technol ; 1(3): 241-6, 2004 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24981491

RESUMO

The three-dimensional structures of proteins are being solved apace, yet this information is often underused in quantitative structure-activity relationship (QSAR) studies. Here, we describe and compare methods for exploiting protein structures to derive 3D-QSARs. These methods can facilitate molecular design and lead optimization and should increasingly become a standard component of the drug designer's repertoire.:

11.
J Med Chem ; 54(1): 211-21, 2011 Jan 13.
Artigo em Inglês | MEDLINE | ID: mdl-21126022

RESUMO

Folate analogue inhibitors of Leishmania major pteridine reductase (PTR1) are potential antiparasitic drug candidates for combined therapy with dihydrofolate reductase (DHFR) inhibitors. To identify new molecules with specificity for PTR1, we carried out a virtual screening of the Available Chemicals Directory (ACD) database to select compounds that could interact with L. major PTR1 but not with human DHFR. Through two rounds of drug discovery, we successfully identified eighteen drug-like molecules with low micromolar affinities and high in vitro specificity profiles. Their efficacy against Leishmania species was studied in cultured cells of the promastigote stage, using the compounds both alone and in combination with 1 (pyrimethamine; 5-(4-chlorophenyl)-6-ethylpyrimidine-2,4-diamine). Six compounds showed efficacy only in combination. In toxicity tests against human fibroblasts, several compounds showed low toxicity. One compound, 5c (riluzole; 6-(trifluoromethoxy)-1,3-benzothiazol-2-ylamine), a known drug approved for CNS pathologies, was active in combination and is suitable for early preclinical evaluation of its potential for label extension as a PTR1 inhibitor and antiparasitic drug candidate.


Assuntos
Fármacos do Sistema Nervoso Central/química , Modelos Moleculares , Oxirredutases/antagonistas & inibidores , Relação Quantitativa Estrutura-Atividade , Tripanossomicidas/química , Benzotiazóis/síntese química , Benzotiazóis/química , Benzotiazóis/farmacologia , Fármacos do Sistema Nervoso Central/síntese química , Fármacos do Sistema Nervoso Central/farmacologia , Desenho de Fármacos , Sinergismo Farmacológico , Fibroblastos/citologia , Fibroblastos/efeitos dos fármacos , Humanos , Leishmania/efeitos dos fármacos , Leishmania/enzimologia , Oxirredutases/química , Testes de Sensibilidade Parasitária , Pirimetamina/análogos & derivados , Pirimetamina/síntese química , Pirimetamina/química , Pirimetamina/farmacologia , Riluzol/análogos & derivados , Riluzol/síntese química , Riluzol/química , Riluzol/farmacologia , Bibliotecas de Moléculas Pequenas , Tetra-Hidrofolato Desidrogenase/química , Tripanossomicidas/síntese química , Tripanossomicidas/farmacologia
12.
Acta Crystallogr D Biol Crystallogr ; 61(Pt 5): 505-12, 2005 May.
Artigo em Inglês | MEDLINE | ID: mdl-15858259

RESUMO

The mammalian prohormone/proprotein convertase (PC) furin is responsible for the maturation of a great variety of homeostatic but also many pathogenic proteins within the secretory pathway and the endosomal pathway and at the cell surface. Similar to other members of the PC family, furin requires calcium for catalytic activity. In a previous paper, the structural association of the catalytic and the P-domain of furin was shown and data were presented indicating two or three calcium-binding sites. The exact number and the three-dimensional localization of the essential calcium sites within furin have now been determined by collecting X-ray diffraction data on either side of the Ca K absorption edge and by calculating a novel type of double difference map from these anomalous scattering data. Two calcium ions were unambiguously identified: the purely structural Ca-1 also conserved in the bacterial digestive subtilisins and the Ca-2 site specific to PCs and essential for the formation of the P1 specificity-determining S1-binding pocket. In addition, these anomalous diffraction data show that no tightly bound K(+) sites exist in furin.


Assuntos
Cálcio/química , Furina/química , Cristalização , Cristalografia por Raios X , Interpretação Estatística de Dados , Metais/química , Conformação Proteica , Especificidade por Substrato , Subtilisinas/química
13.
Biol Chem ; 386(8): 759-66, 2005 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16201871

RESUMO

Triple-helical collagen IV protomers associate through their N- and C-termini, forming a three-dimensional network that provides basement membranes with mechanical strength. Within this network, the C-terminal non-collagenous (NC1) domains form tight dimeric junctions. Crystallographic analyses of isolated NC1 domains show two trimeric cap-like structures interacting via a large interface. Previously, for NC1 from human placenta type-IV collagen we described covalent alpha1-alpha1 and alpha2-alpha2 crosslinks between Met93 and Lys211 of opposing alpha1(IV) and alpha2(IV) NC1-chains, which further stabilize this interface and explain the occurrence of reduction-insensitive NC1-chain dimers. However, their existence was recently questioned, and we therefore analyzed NC1-domain dimers in more detail by biochemical and protein crystallographic methods. Short-exposure diffraction data show a clear electron density cross-connecting the respective residues, which gradually disappears with prolonged crystal irradiation. Sequence analyses of isolated tryptic peptides derived from denatured NC1 monomers and dimers indicate that only the dimers, but not the monomers, yield these chemically labile cross-linked peptides. These data clearly demonstrate the presence of reduction-resistant, but chemically and radiation-sensitive covalent crosslinks between the side chains of Met93 and Lys211 in human placenta type-IV collagen.


Assuntos
Membrana Basal/metabolismo , Colágeno Tipo IV/química , Reagentes de Ligações Cruzadas/química , Placenta/metabolismo , Sequência de Aminoácidos , Dimerização , Feminino , Humanos , Lisina/química , Metionina/química , Dados de Sequência Molecular , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/metabolismo , Ligação Proteica , Conformação Proteica , Tripsina/metabolismo
15.
Nat Struct Biol ; 10(7): 520-6, 2003 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-12794637

RESUMO

In eukaryotes, many essential secreted proteins and peptide hormones are excised from larger precursors by members of a class of calcium-dependent endoproteinases, the prohormone-proprotein convertases (PCs). Furin, the best-characterized member of the mammalian PC family, has essential functions in embryogenesis and homeostasis but is also implicated in various pathologies such as tumor metastasis, neurodegeneration and various bacterial and viral diseases caused by such pathogens as anthrax and pathogenic Ebola virus strains. Furin cleaves protein precursors with narrow specificity following basic Arg-Xaa-Lys/Arg-Arg-like motifs. The 2.6 A crystal structure of the decanoyl-Arg-Val-Lys-Arg-chloromethylketone (dec-RVKR-cmk)-inhibited mouse furin ectodomain, the first PC structure, reveals an eight-stranded jelly-roll P domain associated with the catalytic domain. Contoured surface loops shape the active site by cleft, thus explaining furin's stringent requirement for arginine at P1 and P4, and lysine at P2 sites by highly charge-complementary pockets. The structure also explains furin's preference for basic residues at P3, P5 and P6 sites. This structure will aid in the rational design of antiviral and antibacterial drugs.


Assuntos
Subtilisinas/metabolismo , Sequência de Aminoácidos , Animais , Domínio Catalítico , Cristalografia por Raios X , Inibidores Enzimáticos/metabolismo , Furina , Camundongos , Modelos Moleculares , Dados de Sequência Molecular , Conformação Proteica , Homologia de Sequência de Aminoácidos , Especificidade por Substrato , Subtilisinas/antagonistas & inibidores , Subtilisinas/química
16.
J Biol Chem ; 279(35): 36788-94, 2004 Aug 27.
Artigo em Inglês | MEDLINE | ID: mdl-15197180

RESUMO

Polyarginine-containing peptides represent potent inhibitors of furin, a mammalian endoprotease that plays an important role in metabolism, activation of pathogenic toxins, and viral proliferation. The therapeutic use of D-polyarginines is especially interesting because they are not cleaved by furin and possess inhibitory potency almost equal to L-polyarginines. In this study we attempted to determine the important elements within polyarginines that contribute to effective inhibition. Structure-function analyses of polyarginine peptides showed that inhibition by polyarginine-containing peptides appeared to depend on the total number of basic charges of the positively charged inhibitors bound to the negatively charged substrate binding pocket; peptide positioning did not appear to be rigorously determined. Screening of L- and D-decapeptide positional scanning combinatorial peptide libraries indicated a preference for basic residues in nearly all positions, similar to previous results with hexapeptide libraries. Length and terminal modification studies showed that the most potent D-polyarginine tested was nona-D-arginine (D9R) amide with a K(i) of 1.3 nm. D9R amide was shown to protect RAW264.7 cells against anthrax toxemia with an IC(50) of 3.7 microm. Because of its high stability, specificity, low toxicity, small molecular weight, and extremely low K(i) against furin, D9R amide or its derivatives may represent promising compounds for therapeutic use.


Assuntos
Arginina/química , Furina/antagonistas & inibidores , Peptídeos/química , Alanina/química , Animais , Antígenos de Bactérias/metabolismo , Toxinas Bacterianas/metabolismo , Células CHO , Cricetinae , Cristalografia por Raios X , DNA Complementar/metabolismo , Relação Dose-Resposta a Droga , Humanos , Hidrólise , Concentração Inibidora 50 , Íons , Cinética , Lisina/química , Camundongos , Modelos Moleculares , Oligopeptídeos/farmacologia , Biblioteca de Peptídeos , Ligação Proteica , Relação Estrutura-Atividade , Especificidade por Substrato
17.
Proc Natl Acad Sci U S A ; 99(10): 6607-12, 2002 May 14.
Artigo em Inglês | MEDLINE | ID: mdl-12011424

RESUMO

Triple-helical collagen IV protomers associate through their N- and C-termini forming a three-dimensional network, which provides basement membranes with an anchoring scaffold and mechanical strength. The noncollagenous (NC1) domain of the C-terminal junction between two adjacent collagen IV protomers from human placenta was crystallized and its 1.9-A structure was solved by multiple anomalous diffraction (MAD) phasing. This hexameric NC1 particle is composed of two trimeric caps, which interact through a large planar interface. Each cap is formed by two alpha 1 fragments and one alpha 2 fragment with a similar previously uncharacterized fold, segmentally arranged around an axial tunnel. Each monomer chain folds into two structurally very similar subdomains, which each contain a finger-like hairpin loop that inserts into a six-stranded beta-sheet of the neighboring subdomain of the same or the adjacent chain. Thus each trimer forms a quite regular, but nonclassical, sixfold propeller. The trimer-trimer interaction is further stabilized by a previously uncharacterized type of covalent cross-link between the side chains of a Met and a Lys residue of the alpha 1 and alpha 2 chains from opposite trimers, explaining previous findings of nonreducible cross-links in NC1. This structure provides insights into NC1-related diseases such as Goodpasture and Alport syndromes.


Assuntos
Colágeno Tipo IV/química , Sequência de Aminoácidos , Colágeno Tipo IV/genética , Cristalografia por Raios X , Feminino , Humanos , Lisina/química , Metionina/química , Modelos Moleculares , Dados de Sequência Molecular , Estrutura Molecular , Placenta/metabolismo , Conformação Proteica , Estrutura Terciária de Proteína , Homologia de Sequência de Aminoácidos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA