Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Ann Surg ; 275(2): e453-e462, 2022 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-32487804

RESUMO

OBJECTIVE: Acute Pancreatitis (AP) is sudden onset pancreas inflammation that causes systemic injury with a wide and markedly heterogeneous range of clinical consequences. Here, we hypothesized that this observed clinical diversity corresponds to diversity in molecular subtypes that can be identified in clinical and multiomics data. SUMMARY BACKGROUND DATA: Observational cohort study. n = 57 for the discovery cohort (clinical, transcriptomics, proteomics, and metabolomics data) and n = 312 for the validation cohort (clinical and metabolomics data). METHODS: We integrated coincident transcriptomics, proteomics, and metabolomics data at serial time points between admission to hospital and up to 48 hours after recruitment from a cohort of patients presenting with acute pancreatitis. We systematically evaluated 4 different metrics for patient similarity using unbiased mathematical, biological, and clinical measures of internal and external validity.We next compared the AP molecular endotypes with previous descriptions of endotypes in a critically ill population with acute respiratory distress syndrome (ARDS). RESULTS: Our results identify 4 distinct and stable AP molecular endotypes. We validated our findings in a second independent cohort of patients with AP.We observed that 2 endotypes in AP recapitulate disease endotypes previously reported in ARDS. CONCLUSIONS: Our results show that molecular endotypes exist in AP and reflect biological patterns that are also present in ARDS, suggesting that generalizable patterns exist in diverse presentations of critical illness.


Assuntos
Pancreatite/classificação , Pancreatite/diagnóstico , Estudos de Coortes , Humanos , Metabolômica , Proteômica
2.
Am J Physiol Endocrinol Metab ; 314(6): E605-E619, 2018 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-28655718

RESUMO

In striated muscle, eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA) have differential effects on the metabolism of glucose and differential effects on the metabolism of protein. We have shown that, despite similar incorporation, treatment of C2C12 myotubes (CM) with EPA but not DHA improves glucose uptake and protein accretion. We hypothesized that these differential effects of EPA and DHA may be due to divergent shifts in lipidomic profiles leading to altered proteomic profiles. We therefore carried out an assessment of the impact of treating CM with EPA and DHA on lipidomic and proteomic profiles. Fatty acid methyl esters (FAME) analysis revealed that both EPA and DHA led to similar but substantials changes in fatty acid profiles with the exception of arachidonic acid, which was decreased only by DHA, and docosapentanoic acid (DPA), which was increased only by EPA treatment. Global lipidomic analysis showed that EPA and DHA induced large alterations in the cellular lipid profiles and in particular, the phospholipid classes. Subsequent targeted analysis confirmed that the most differentially regulated species were phosphatidylcholines and phosphatidylethanolamines containing long-chain fatty acids with five (EPA treatment) or six (DHA treatment) double bonds. As these are typically membrane-associated lipid species we hypothesized that these treatments differentially altered the membrane-associated proteome. Stable isotope labeling by amino acids in cell culture (SILAC)-based proteomics of the membrane fraction revealed significant divergence in the effects of EPA and DHA on the membrane-associated proteome. We conclude that the EPA-specific increase in polyunsaturated long-chain fatty acids in the phospholipid fraction is associated with an altered membrane-associated proteome and these may be critical events in the metabolic remodeling induced by EPA treatment.


Assuntos
Ácidos Docosa-Hexaenoicos/farmacologia , Ácido Eicosapentaenoico/farmacologia , Glucose/metabolismo , Metabolismo dos Lipídeos/efeitos dos fármacos , Proteínas de Membrana/efeitos dos fármacos , Músculo Esquelético/efeitos dos fármacos , Proteoma/efeitos dos fármacos , Animais , Metabolismo dos Carboidratos/efeitos dos fármacos , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Células Cultivadas , Ácido Eicosapentaenoico/análogos & derivados , Ácidos Graxos/metabolismo , Proteínas de Membrana/metabolismo , Camundongos , Músculo Esquelético/metabolismo , Proteoma/metabolismo , Triglicerídeos/metabolismo
3.
Proc Natl Acad Sci U S A ; 110(33): 13600-5, 2013 Aug 13.
Artigo em Inglês | MEDLINE | ID: mdl-23904481

RESUMO

Biofilms represent the predominant mode of microbial growth in the natural environment. Bacillus subtilis is a ubiquitous Gram-positive soil bacterium that functions as an effective plant growth-promoting agent. The biofilm matrix is composed of an exopolysaccharide and an amyloid fiber-forming protein, TasA, and assembles with the aid of a small secreted protein, BslA. Here we show that natively synthesized and secreted BslA forms surface layers around the biofilm. Biophysical analysis demonstrates that BslA can self-assemble at interfaces, forming an elastic film. Molecular function is revealed from analysis of the crystal structure of BslA, which consists of an Ig-type fold with the addition of an unusual, extremely hydrophobic "cap" region. A combination of in vivo biofilm formation and in vitro biophysical analysis demonstrates that the central hydrophobic residues of the cap are essential to allow a hydrophobic, nonwetting biofilm to form as they control the surface activity of the BslA protein. The hydrophobic cap exhibits physiochemical properties remarkably similar to the hydrophobic surface found in fungal hydrophobins; thus, BslA is a structurally defined bacterial hydrophobin. We suggest that biofilms formed by other species of bacteria may have evolved similar mechanisms to provide protection to the resident bacterial community.


Assuntos
Bacillus subtilis/metabolismo , Proteínas de Bactérias/química , Biofilmes , Modelos Moleculares , Conformação Proteica , Proteínas de Bactérias/metabolismo , Biofísica , Imunofluorescência , Interações Hidrofóbicas e Hidrofílicas , Microscopia Confocal
4.
Glycobiology ; 23(5): 536-45, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23300007

RESUMO

Sialyltransferases (STs) play essential roles in signaling and in the cellular recognition processes of mammalian cells by selectively installing cell-surface sialic acids in an appropriate manner both temporally and organ-specifically. The availability of the first three-dimensional structure of a mammalian (GT29) sialyltransferase has, for the first time, allowed quantitative structure/function analyses to be performed, thereby providing reliable insights into the roles of key active site amino acids. Kinetic analyses of mutants of ST3Gal-I, in conjunction with structural studies, have confirmed the mechanistic roles of His302 and His319 as general acid and base catalysts, respectively, and have quantitated other interactions with the cytosine monophosphate-N-acetyl ß-neuraminic acid donor substrate. The contributions of side chains that provide key interactions with the acceptor substrate, defining its specificity, have also been quantitated. Particularly important transition-state interactions of 2.5 and 2.7 kcal mol(-1) are found between the acceptor axial 4-hydroxyl and the conserved side chains of Gln108 and Tyr269, respectively. These results provide a basis for the engineering of mammalian STs to accommodate non-natural substrate analogs that should prove valuable as chemical biological probes of sialyltransferase function.


Assuntos
Mutação de Sentido Incorreto , Sialiltransferases/genética , Sialiltransferases/metabolismo , Sequência de Aminoácidos , Animais , Domínio Catalítico , Ácido N-Acetilneuramínico do Monofosfato de Citidina/metabolismo , Cinética , Dados de Sequência Molecular , Ligação Proteica , Sialiltransferases/química , Especificidade por Substrato , Suínos , beta-Galactosídeo alfa-2,3-Sialiltransferase
5.
Acta Crystallogr D Biol Crystallogr ; 68(Pt 8): 1019-29, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22868768

RESUMO

Glucosamine-6-phosphate N-acetyltransferase 1 (GNA1) produces GlcNAc-6-phosphate from GlcN-6-phosphate and acetyl coenzyme A. Early mercury-labelling experiments implicated a conserved cysteine in the reaction mechanism, whereas recent structural data appear to support a mechanism in which this cysteine plays no role. Here, two crystal structures of Caenorhabditis elegans GNA1 are reported, revealing an unusual covalent complex between this cysteine and the coenzyme A product. Mass-spectrometric and reduction studies showed that this inactive covalent complex can be reactivated through reduction, yet mutagenesis of the cysteine supports a previously reported bi-bi mechanism. The data unify the apparently contradictory earlier reports on the role of a cysteine in the GNA1 active site.


Assuntos
Caenorhabditis elegans/enzimologia , Coenzima A/química , Glucosamina 6-Fosfato N-Acetiltransferase/química , Animais , Domínio Catalítico , Clonagem Molecular , Sequência Conservada , Cristalografia por Raios X/métodos , Cisteína/química , Cinética , Espectrometria de Massas/métodos , Modelos Moleculares , Conformação Molecular , Mutação , Oxigênio/química , Ligação Proteica
6.
BMC Res Notes ; 12(1): 494, 2019 Aug 08.
Artigo em Inglês | MEDLINE | ID: mdl-31395095

RESUMO

OBJECTIVES: Glyphosate (N-phosphonomethyl glycine) and its commercial herbicide formulations have been shown to exert toxicity via various mechanisms. It has been asserted that glyphosate substitutes for glycine in polypeptide chains leading to protein misfolding and toxicity. However, as no direct evidence exists for glycine to glyphosate substitution in proteins, including in mammalian organisms, we tested this claim by conducting a proteomics analysis of MDA-MB-231 human breast cancer cells grown in the presence of 100 mg/L glyphosate for 6 days. Protein extracts from three treated and three untreated cell cultures were analysed as one TMT-6plex labelled sample, to highlight a specific pattern (+/+/+/-/-/-) of reporter intensities for peptides bearing true glyphosate treatment induced-post translational modifications as well as allowing an investigation of the total proteome. RESULTS: Comparative statistical analysis of global proteome changes between glyphosate treated and non-treated samples did not show significant differences. Crucially, filtering of data to focus analysis on peptides potentially bearing glycine for glyphosate replacement revealed that the TMT reporter intensity pattern of all candidates showed conclusively that they are all false discoveries, with none displaying the expected TMT pattern for such a substitution. Thus, the assertion that glyphosate substitutes for glycine in protein polypeptide chains is incorrect.


Assuntos
Glicina/análogos & derivados , Glicina/metabolismo , Herbicidas/química , Proteínas de Neoplasias/metabolismo , Processamento de Proteína Pós-Traducional , Proteoma/metabolismo , Linhagem Celular Tumoral , Expressão Gênica , Glicina/química , Glicina-tRNA Ligase/química , Glicina-tRNA Ligase/genética , Glicina-tRNA Ligase/metabolismo , Herbicidas/metabolismo , Humanos , Modelos Moleculares , Proteínas de Neoplasias/genética , Proteoma/genética , Glifosato
7.
Front Microbiol ; 9: 2026, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30233517

RESUMO

Bacterial surface polysaccharides play significant roles in fitness and virulence. In Gram-negative bacteria such as Escherichia coli, major surface polysaccharides are lipopolysaccharide (LPS) and capsule, representing O- and K-antigens, respectively. There are multiple combinations of O:K types, many of which are well-characterized and can be related to ecotype or pathotype. In this investigation, we have identified a novel O:K permutation resulting through a process of major genome reorganization in a clade of E. coli. A multidrug-resistant, extended-spectrum ß-lactamase (ESBL)-producing strain - E. coli 26561 - represented a prototype of strains combining a locus variant of O89 and group 1 capsular polysaccharide. Specifically, the variant O89 locus in this strain was truncated at gnd, flanked by insertion sequences and located between nfsB and ybdK and we apply the term O89m for this variant. The prototype lacked colanic acid and O-antigen loci between yegH and hisI with this tandem polysaccharide locus being replaced with a group 1 capsule (G1C) which, rather than being a recognized E. coli capsule type, this locus matched to Klebsiella K10 capsule type. A genomic survey identified more than 200 E. coli strains which possessed the O89m locus variant with one of a variety of G1C types. Isolates from our collection with the combination of O89m and G1C all displayed a mucoid phenotype and E. coli 26561 was unusual in exhibiting a mucoviscous phenotype more recognized as a characteristic among Klebsiella strains. Despite the locus truncation and novel location, all O89m:G1C strains examined showed a ladder pattern typifying smooth LPS and also showed high molecular weight, alcian blue-staining polysaccharide in cellular and/or extra-cellular fractions. Expression of both O-antigen and capsule biosynthesis loci were confirmed in prototype strain 26561 through quantitative proteome analysis. Further in silico exploration of more than 200 E. coli strains possessing the O89m:G1C combination identified a very high prevalence of multidrug resistance (MDR) - 85% possessed resistance to three or more antibiotic classes and a high proportion (58%) of these carried ESBL and/or carbapenemase. The increasing isolation of O89m:G1C isolates from extra-intestinal infection sites suggests that these represents an emergent clade of invasive, MDR E. coli.

8.
Chem Biol ; 12(9): 973-80, 2005 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16183021

RESUMO

Family 18 chitinases play key roles in a range of pathogenic organisms and are overexpressed in the asthmatic lung. By screening a library of marketed drug molecules, we have identified methylxanthine derivatives as possible inhibitor leads. These derivatives, theophylline, caffeine, and pentoxifylline, are used therapeutically as antiinflammatory agents, with pleiotropic mechanisms of action. Here it is shown that they are also competitive inhibitors against a fungal family 18 chitinase, with pentoxifylline being the most potent (K(i) of 37 microM). Crystallographic analysis of chitinase-inhibitor complexes revealed specific interactions with the active site, mimicking the reaction intermediate analog, allosamidin. Mutagenesis identified the key active site residues, conserved in mammalian chitinases, which contribute to inhibitor affinity. Enzyme assays also revealed that these methylxanthines are active against human chitinases.


Assuntos
Quitinases/antagonistas & inibidores , Inibidores Enzimáticos/farmacologia , Xantinas/farmacologia , Quitinases/metabolismo , Inibidores Enzimáticos/química , Inibidores Enzimáticos/metabolismo , Modelos Moleculares , Estrutura Molecular , Ligação Proteica , Xantinas/química , Xantinas/metabolismo
9.
Chem Biol ; 12(1): 65-76, 2005 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-15664516

RESUMO

Family 18 chitinases play key roles in organisms ranging from bacteria to man. There is a need for specific, potent inhibitors to probe the function of these chitinases in different organisms. Such molecules could also provide leads for the development of chemotherapeuticals with fungicidal, insecticidal, or anti-inflammatory potential. Recently, two natural product peptides, argifin and argadin, have been characterized, which structurally mimic chitinase-chitooligosaccharide interactions and inhibit a bacterial chitinase in the nM-mM range. Here, we show that these inhibitors also act on human and Aspergillus fumigatus chitinases. The structures of these enzymes in complex with argifin and argadin, together with mutagenesis, fluorescence, and enzymology, reveal that subtle changes in the binding site dramatically affect affinity and selectivity. The data show that it may be possible to develop specific chitinase inhibitors based on the argifin/argadin scaffolds.


Assuntos
Aspergillus fumigatus/enzimologia , Bactérias/enzimologia , Quitinases/antagonistas & inibidores , Quitinases/química , Inibidores Enzimáticos/farmacologia , Peptídeos Cíclicos/farmacologia , Aspergillus fumigatus/efeitos dos fármacos , Bactérias/efeitos dos fármacos , Ligação Competitiva , Carboidratos/química , Carboidratos/farmacologia , Quitinases/classificação , Clonagem Molecular , Desenho de Fármacos , Inibidores Enzimáticos/química , Humanos , Ligação de Hidrogênio , Cinética , Masculino , Mimetismo Molecular , Dados de Sequência Molecular , Estrutura Molecular , Peptídeos Cíclicos/química , Estrutura Terciária de Proteína , Relação Estrutura-Atividade , Especificidade por Substrato
10.
Biochim Biophys Acta ; 1696(1): 103-11, 2004 Jan 14.
Artigo em Inglês | MEDLINE | ID: mdl-14726210

RESUMO

Catalysis by ChiB, a family 18 chitinase from Serratia marcescens, involves a conformational change of Asp142 which is part of a characteristic D(140)XD(142)XE(144) sequence motif. In the free enzyme Asp142 points towards Asp140, whereas it rotates towards the catalytic acid, Glu144, upon ligand binding. Mutation of Asp142 to Asn reduced k(cat) and affinity for allosamidin, a competitive inhibitor. The X-ray structure of the D142N mutant showed that Asn142 points towards Glu144 in the absence of a ligand. The active site also showed other structural adjustments (Tyr10, Ser93) that had previously been observed in the wild-type enzyme upon substrate binding. The X-ray structure of a complex of D142N with allosamidin, a pseudotrisaccharide competitive inhibitor, was essentially identical to that of the wild-type enzyme in complex with the same compound. Thus, the reduced allosamidin affinity in the mutant is not caused by structural changes but solely by the loss of electrostatic interactions with Asp142. The importance of electrostatics was further confirmed by the pH dependence of catalysis and allosamidin inhibition. The pH-dependent apparent affinities for allosamidin were not correlated with k(cat), indicating that it is probably better to view the inhibitor as a mimic of the oxazolinium ion reaction intermediate than as a transition state analogue.


Assuntos
Acetilglucosamina/análogos & derivados , Acetilglucosamina/química , Quitinases/química , Serratia marcescens/enzimologia , Trissacarídeos/química , Ligação Competitiva , Catálise , Quitinases/antagonistas & inibidores , Quitinases/genética , Inibidores Enzimáticos/química , Concentração de Íons de Hidrogênio , Cinética , Modelos Moleculares , Mutagênese Sítio-Dirigida , Mutação , Serratia marcescens/genética
11.
Open Biol ; 3(10): 130021, 2013 Oct 02.
Artigo em Inglês | MEDLINE | ID: mdl-24088714

RESUMO

The dynamic modification of proteins by O-linked N-acetylglucosamine (O-GlcNAc) is an essential posttranslational modification present in higher eukaryotes. Removal of O-GlcNAc is catalysed by O-GlcNAcase, a multi-domain enzyme that has been reported to be bifunctional, possessing both glycoside hydrolase and histone acetyltransferase (AT) activity. Insights into the mechanism, protein substrate recognition and inhibition of the hydrolase domain of human OGA (hOGA) have been obtained via the use of the structures of bacterial homologues. However, the molecular basis of AT activity of OGA, which has only been reported in vitro, is not presently understood. Here, we describe the crystal structure of a putative acetyltransferase (OgpAT) that we identified in the genome of the marine bacterium Oceanicola granulosus, showing homology to the hOGA C-terminal AT domain (hOGA-AT). The structure of OgpAT in complex with acetyl coenzyme A (AcCoA) reveals that, by homology modelling, hOGA-AT adopts a variant AT fold with a unique loop creating a deep tunnel. The structures, together with mutagenesis and surface plasmon resonance data, reveal that while the bacterial OgpAT binds AcCoA, the hOGA-AT does not, as explained by the lack of key residues normally required to bind AcCoA. Thus, the C-terminal domain of hOGA is a catalytically incompetent 'pseudo'-AT.


Assuntos
Acetilglucosamina/metabolismo , Histona Acetiltransferases/química , Histona Acetiltransferases/genética , Rhodobacteraceae/enzimologia , beta-N-Acetil-Hexosaminidases/química , Acetilcoenzima A/química , Acetilcoenzima A/metabolismo , Acetilglucosamina/química , Sequência de Aminoácidos , Proteínas de Bactérias/química , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Sítios de Ligação , Domínio Catalítico , Cristalografia por Raios X , Histona Acetiltransferases/metabolismo , Humanos , Modelos Moleculares , Dados de Sequência Molecular , Enzimas Multifuncionais/química , Enzimas Multifuncionais/genética , Enzimas Multifuncionais/metabolismo , Óperon , Ligação Proteica , Dobramento de Proteína , Processamento de Proteína Pós-Traducional , Estrutura Secundária de Proteína , Rhodobacteraceae/genética , Alinhamento de Sequência , Especificidade por Substrato , Ressonância de Plasmônio de Superfície , beta-N-Acetil-Hexosaminidases/genética , beta-N-Acetil-Hexosaminidases/metabolismo
12.
ACS Med Chem Lett ; 2(6): 428-32, 2011 Jun 09.
Artigo em Inglês | MEDLINE | ID: mdl-24900325

RESUMO

Chitinases of the GH18 family play important roles in a variety of pathogenic organisms and have also been shown to be involved in human asthma progression, making these enzymes potential drug targets. While a number of potent GH18 chitinase inhibitors have been described, in general, these compounds suffer from limited synthetic accessibility or unfavorable medicinal-chemical properties, making them poor starting points for the development of chitinase-targeted drugs. Exploiting available structural data, we have rationally designed bisdionin C, a submicromolar inhibitor of GH18 enzymes, that possesses desirable druglike properties and tractable chemical synthesis. A crystallographic structure of a chitinase-bisdionin C complex shows the two aromatic systems of the ligand interacting with two conserved tryptophan residues exposed in the active site cleft of the enzyme, while at the same time forming extensive hydrogen-bonding interactions with the catalytic machinery. The observed mode of binding, together with inhibition data, suggests that bisdionin C presents an attractive starting point for the development of specific inhibitors of bacterial-type, but not plant-type, GH 18 chitinases.

13.
Nat Struct Mol Biol ; 16(11): 1186-8, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19820709

RESUMO

Mammalian cell surfaces are modified by complex arrays of glycoproteins, glycolipids and polysaccharides, many of which terminate in sialic acid and have central roles in essential processes including cell recognition, adhesion and immunogenicity. Sialylation of glycoconjugates is performed by a set of sequence-related enzymes known as sialyltransferases (STs). Here we present the crystal structure of a mammalian ST, porcine ST3Gal-I, providing a structural basis for understanding the mechanism and specificity of these enzymes and for the design of selective inhibitors.


Assuntos
Sialiltransferases/química , Animais , Domínio Catalítico , Cristalografia por Raios X , Humanos , Modelos Moleculares , Ácido N-Acetilneuramínico/metabolismo , Estrutura Secundária de Proteína , Sialiltransferases/genética , Sialiltransferases/metabolismo , Especificidade por Substrato , Suínos , beta-Galactosídeo alfa-2,3-Sialiltransferase
14.
EMBO J ; 25(7): 1569-78, 2006 Apr 05.
Artigo em Inglês | MEDLINE | ID: mdl-16541109

RESUMO

O-linked N-acetylglucosamine (O-GlcNAc) modification of specific serines/threonines on intracellular proteins in higher eukaryotes has been shown to directly regulate important processes such as the cell cycle, insulin sensitivity and transcription. The structure, molecular mechanisms of catalysis, protein substrate recognition/specificity of the eukaryotic O-GlcNAc transferase and hydrolase are largely unknown. Here we describe the crystal structure, enzymology and in vitro activity on human substrates of Clostridium perfringens NagJ, a close homologue of human O-GlcNAcase (OGA), representing the first family 84 glycoside hydrolase structure. The structure reveals a deep active site pocket highly conserved with the human enzyme, compatible with binding of O-GlcNAcylated peptides. Together with mutagenesis data, the structure supports a variant of the substrate-assisted catalytic mechanism, involving two aspartic acids and an unusually positioned tyrosine. Insights into recognition of substrate come from a complex with the transition state mimic O-(2-acetamido-2-deoxy-D-glucopyranosylidene)amino-N-phenylcarbamate (Ki=5.4 nM). Strikingly, the enzyme is inhibited by the pseudosubstrate peptide Ala-Cys(-S-GlcNAc)-Ala, and has OGA activity against O-GlcNAcylated human proteins, suggesting that the enzyme is a suitable model for further studies into the function of human OGA.


Assuntos
Acetilglucosamina/análogos & derivados , Acetilglucosamina/química , Acetilglucosaminidase/química , Histona Acetiltransferases/química , Complexos Multienzimáticos/química , Células 3T3 , Acetilglucosamina/metabolismo , Acetilglucosaminidase/genética , Acetilglucosaminidase/metabolismo , Sequência de Aminoácidos , Animais , Sítios de Ligação , Catálise , Clostridium perfringens/enzimologia , Cristalografia por Raios X , Glicosilação , Histona Acetiltransferases/genética , Histona Acetiltransferases/metabolismo , Humanos , Hidrólise , Camundongos , Modelos Moleculares , Dados de Sequência Molecular , Complexos Multienzimáticos/genética , Complexos Multienzimáticos/metabolismo , Mutação , Oximas/química , Fenilcarbamatos/química , Fosforilação , Conformação Proteica , Homologia de Sequência de Aminoácidos , Estreptozocina/química , Especificidade por Substrato , beta-N-Acetil-Hexosaminidases/química
15.
J Biol Chem ; 281(37): 27278-85, 2006 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-16844689

RESUMO

Family 18 chitinases play key roles in the life cycles of a variety of organisms ranging from bacteria to man. Very recently it has been shown that one of the mammalian chitinases is highly overexpressed in the asthmatic lung and contributes to the pathogenic process through recruitment of inflammatory cells. Although several potent natural product chitinase inhibitors have been identified, their chemotherapeutic potential or their use as cell biological tools is limited due to their size, complex chemistry, and limited availability. We describe a virtual screening-based approach to identification of a novel, purine-based, chitinase inhibitor. This inhibitor acts in the low micromolar (Ki=2.8+/-0.2 microM) range in a competitive mode. Dissection of the binding mode by x-ray crystallography reveals that the compound, which consists of two linked caffeine moieties, binds in the active site through extensive and not previously observed stacking interactions with conserved, solvent exposed tryptophans. Such exposed aromatics are also present in the structures of many other carbohydrate processing enzymes. The compound exhibits favorable chemical properties and is likely to be useful as a general scaffold for development of pan-family 18 chitinase inhibitors.


Assuntos
Bioquímica/métodos , Quitinases/antagonistas & inibidores , Inibidores Enzimáticos/química , Algoritmos , Aspergillus fumigatus/enzimologia , Sítios de Ligação , Cafeína/química , Quitinases/química , Biologia Computacional/métodos , Cristalografia por Raios X , Desenho de Fármacos , Inibidores Enzimáticos/farmacologia , Escherichia coli/metabolismo , Modelos Químicos , Modelos Moleculares , Triptofano/química
16.
J Biol Chem ; 278(22): 20110-6, 2003 May 30.
Artigo em Inglês | MEDLINE | ID: mdl-12639956

RESUMO

The pseudotrisaccharide allosamidin is a potent family 18 chitinase inhibitor with demonstrated biological activity against insects, fungi, and the Plasmodium falciparum life cycle. The synthesis and biological properties of several derivatives have been reported. The structural interactions of allosamidin with several family 18 chitinases have been determined by x-ray crystallography previously. Here, a high resolution structure of chitotriosidase, the human macrophage chitinase, in complex with allosamidin is presented. In addition, complexes of the allosamidin derivatives demethylallosamidin, methylallosamidin, and glucoallosamidin B are described, together with their inhibitory properties. Similar to other chitinases, inhibition of the human chitinase by allosamidin derivatives lacking a methyl group is 10-fold stronger, and smaller effects are observed for the methyl and C3 epimer derivatives. The structures explain the effects on inhibition in terms of altered hydrogen bonding and hydrophobic interactions, together with displaced water molecules. The data reported here represent a first step toward structure-based design of specific allosamidin derivatives.


Assuntos
Acetilglucosamina/análogos & derivados , Acetilglucosamina/química , Quitinases/metabolismo , Macrófagos/enzimologia , Trissacarídeos/química , Acetilglucosamina/metabolismo , Sequência de Aminoácidos , Cristalografia por Raios X , Humanos , Modelos Moleculares , Dados de Sequência Molecular , Conformação Proteica , Homologia de Sequência de Aminoácidos , Trissacarídeos/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA