Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Biochem J ; 474(5): 851-864, 2017 02 20.
Artigo em Inglês | MEDLINE | ID: mdl-28049758

RESUMO

Cathepsin K (CatK) is the predominant mammalian bone-degrading protease and thus an ideal target for antiosteoporotic drug development. Rodent models of osteoporosis are preferred due to their close reflection of the human disease and their ease of handling, genetic manipulation and economic affordability. However, large differences in the potency of CatK inhibitors for the mouse/rat vs. the human protease orthologs have made it impossible to use rodent models. This is even more of a problem considering that the most advanced CatK inhibitors, including odanacatib (ODN) and balicatib, failed in human clinical trials due to side effects and rodent models are not available to investigate the mechanism of these failures. Here, we elucidated the structural elements of the potency differences between mouse and human CatK (hCatK) using ODN. We determined and compared the structures of inhibitor-free mouse CatK (mCatK), hCatK and ODN bound to hCatK. Two structural differences were identified and investigated by mutational analysis. Humanizing subsite 2 in mCatK led to a 5-fold improvement of ODN binding, whereas the replacement of Tyr61 in mCatK with Asp resulted in an hCatK with comparable ODN potency. Combining both sites further improved the inhibition of the mCatK variant. Similar results were obtained for balicatib. These findings will allow the generation of transgenic CatK mice that will facilitate the evaluation of CatK inhibitor adverse effects and to explore routes to avoid them.


Assuntos
Benzamidas/química , Compostos de Bifenilo/química , Conservadores da Densidade Óssea/química , Catepsina K/antagonistas & inibidores , Piperazinas/química , Inibidores de Proteases/química , Sequência de Aminoácidos , Animais , Benzamidas/metabolismo , Sítios de Ligação , Compostos de Bifenilo/metabolismo , Conservadores da Densidade Óssea/metabolismo , Catepsina K/química , Catepsina K/genética , Catepsina K/metabolismo , Clonagem Molecular , Cristalografia por Raios X , Escherichia coli/genética , Escherichia coli/metabolismo , Expressão Gênica , Humanos , Cinética , Ligantes , Camundongos , Mutagênese Sítio-Dirigida , Piperazinas/metabolismo , Inibidores de Proteases/metabolismo , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas , Estrutura Secundária de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Alinhamento de Sequência , Homologia Estrutural de Proteína
2.
Nat Chem Biol ; 11(9): 691-6, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26214255

RESUMO

The complex plant flavonol glycoside montbretin A is a potent (Ki = 8 nM) and specific inhibitor of human pancreatic α-amylase with potential as a therapeutic for diabetes and obesity. Controlled degradation studies on montbretin A, coupled with inhibition analyses, identified an essential high-affinity core structure comprising the myricetin and caffeic acid moieties linked via a disaccharide. X-ray structural analyses of the montbretin A-human α-amylase complex confirmed the importance of this core structure and revealed a novel mode of glycosidase inhibition wherein internal π-stacking interactions between the myricetin and caffeic acid organize their ring hydroxyls for optimal hydrogen bonding to the α-amylase catalytic residues D197 and E233. This novel inhibitory motif can be reproduced in a greatly simplified analog, offering potential for new strategies for glycosidase inhibition and therapeutic development.


Assuntos
Desenho de Fármacos , Inibidores Enzimáticos/química , Flavonóis/química , Glicosídeos/química , alfa-Amilases/química , Sítios de Ligação , Ácidos Cafeicos/química , Sequência de Carboidratos , Inibidores Enzimáticos/síntese química , Flavonas/química , Flavonoides/química , Expressão Gênica , Humanos , Ligação de Hidrogênio , Hidrólise , Ligantes , Modelos Moleculares , Dados de Sequência Molecular , Pichia/genética , Pichia/metabolismo , Ligação Proteica , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Trissacarídeos/química , alfa-Amilases/antagonistas & inibidores , alfa-Amilases/genética
3.
Proc Natl Acad Sci U S A ; 111(49): 17474-9, 2014 Dec 09.
Artigo em Inglês | MEDLINE | ID: mdl-25422423

RESUMO

Cathepsin K is the major collagenolytic protease in bone that facilitates physiological as well as pathological bone degradation. Despite its key role in bone remodeling and for being a highly sought-after drug target for the treatment of osteoporosis, the mechanism of collagen fiber degradation by cathepsin K remained elusive. Here, we report the structure of a collagenolytically active cathepsin K protein dimer. Cathepsin K is organized into elongated C-shaped protease dimers that reveal a putative collagen-binding interface aided by glycosaminoglycans. Molecular modeling of collagen binding to the dimer indicates the participation of nonactive site amino acid residues, Q21 and Q92, in collagen unfolding. Mutations at these sites as well as perturbation of the dimer protein-protein interface completely inhibit cathepsin-K-mediated fiber degradation without affecting the hydrolysis of gelatin or synthetic peptide. Using scanning electron microscopy, we demonstrate the specific binding of cathepsin K at the edge of the fibrillar gap region of collagen fibers, which suggest initial cleavage events at the N- and C-terminal ends of tropocollagen molecules. Edman degradation analysis of collagen fiber degradation products revealed those initial cleavage sites. We propose that one cathepsin K molecule binds to collagen-bound glycosaminoglycans at the gap region and recruits a second protease molecule that provides an unfolding and cleavage mechanism for triple helical collagen. Removal of collagen-associated glycosaminoglycans prevents cathepsin K binding and subsequently fiber hydrolysis. Cathepsin K dimer and glycosaminoglycan binding sites represent novel targeting sites for the development of nonactive site-directed second-generation inhibitors of this important drug target.


Assuntos
Catepsina K/química , Colágeno/química , Aminoácidos/química , Sítios de Ligação , Remodelação Óssea , Osso e Ossos/metabolismo , Cristalografia por Raios X , Glicosaminoglicanos/química , Humanos , Hidrólise , Microscopia Eletrônica , Modelos Moleculares , Mutagênese , Osteoporose , Peptídeo Hidrolases/química , Pichia , Desnaturação Proteica , Dobramento de Proteína , Multimerização Proteica , Estrutura Terciária de Proteína , Especificidade por Substrato
4.
Biochemistry ; 55(43): 6000-6009, 2016 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-27756128

RESUMO

Starch provides the major source of caloric intake in many diets. Cleavage of starch into malto-oligosaccharides in the gut is catalyzed by pancreatic α-amylase. These oligosaccharides are then further cleaved by gut wall α-glucosidases to release glucose, which is absorbed into the bloodstream. Potential surface binding sites for starch on the pancreatic amylase, distinct from the active site of the amylase, have been identified through X-ray crystallographic analyses. The role of these sites in the degradation of both starch granules and soluble starch was probed by the generation of a series of surface variants modified at each site to disrupt binding. Kinetic analysis of the binding and/or cleavage of substrates ranging from simple maltotriosides to soluble starch and insoluble starch granules has allowed evaluation of the potential role of each such surface site. In this way, two key surface binding sites, on the same face as the active site, are identified. One site, containing a pair of aromatic residues, is responsible for attachment to starch granules, while a second site featuring a tryptophan residue around which a malto-oligosaccharide wraps is shown to heavily influence soluble starch binding and hydrolysis. These studies provide insights into the mechanisms by which enzymes tackle the degradation of largely insoluble polymers and also present some new approaches to the interrogation of the binding sites involved.


Assuntos
Pâncreas/enzimologia , Amido/metabolismo , alfa-Amilases/metabolismo , Sítios de Ligação , Humanos , Cinética , Mutagênese Sítio-Dirigida , alfa-Amilases/genética
5.
Mol Cell Biochem ; 411(1-2): 373-81, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26547551

RESUMO

Diabetes is an increasingly prevalent disease state with a global impact. It is important that effective and cost-efficient methods be developed to treat this disease state. Zucker diabetic fatty rats, an animal model of type 2 diabetes, were treated with montbretin A (MbA), a selective human pancreatic α-amylase inhibitor, isolated from the corms of the Crocosmia crocosmiiflora plant that may have potential as a glucose-lowering agent. The study purpose was to determine if MbA was an orally effective treatment for diabetes. The effect of MbA was compared to a current clinical treatment modality, acarbose that is associated with gastrointestinal side effects known to affect patient compliance. MbA and acarbose were administered daily in the drinking water. Body weight and fluid intake were measured daily to calculate dose consumption. Plasma glucose levels were determined twice weekly in both the fed and fasted state. At termination samples were collected to assess increased risk of secondary complications related to diabetes and oxidative stress. There was no effect of either MbA or acarbose treatment on insulin levels. Plasma glucose levels were significantly lower following MbA treatment in the ZT group which persisted throughout the study period (day 49: 12.1 ± 1.2 mM). However, while there was an initial decrease in plasma glucose levels in the acarbose-treated fatty group, this effect was not sustained (day 49: 20.6 ± 1.3 mM) through to termination. MbA improved the oxidative status of the fatty diabetic animals as well as attenuated markers for increased risk of cardiovascular complications associated with diabetes. This study demonstrated that, at a lower dose as compared to acarbose (10 mg/kg/day), chronic oral administration of MbA (7.5 mg/kg/day) was an effective glucose-lowering agent in the treatment of type 2 diabetes.


Assuntos
Glicemia/metabolismo , Flavonas/farmacologia , Hipoglicemiantes/farmacologia , Trissacarídeos/farmacologia , Alanina Transaminase/sangue , Animais , Aspartato Aminotransferases/sangue , Masculino , Ratos , Ratos Zucker
6.
J Nat Prod ; 79(8): 1962-70, 2016 08 26.
Artigo em Inglês | MEDLINE | ID: mdl-27498895

RESUMO

Natural products are an important source of novel drug scaffolds. The highly variable and unpredictable timelines associated with isolating novel compounds and elucidating their structures have led to the demise of exploring natural product extract libraries in drug discovery programs. Here we introduce affinity crystallography as a new methodology that significantly shortens the time of the hit to active structure cycle in bioactive natural product discovery research. This affinity crystallography approach is illustrated by using semipure fractions of an actinomycetes culture extract to isolate and identify a cathepsin K inhibitor and to compare the outcome with the traditional assay-guided purification/structural analysis approach. The traditional approach resulted in the identification of the known inhibitor antipain (1) and its new but lower potency dehydration product 2, while the affinity crystallography approach led to the identification of a new high-affinity inhibitor named lichostatinal (3). The structure and potency of lichostatinal (3) was verified by total synthesis and kinetic characterization. To the best of our knowledge, this is the first example of isolating and characterizing a potent enzyme inhibitor from a partially purified crude natural product extract using a protein crystallographic approach.


Assuntos
Produtos Biológicos/farmacologia , Catepsina K/antagonistas & inibidores , Líquens/química , Peptídeos/isolamento & purificação , Peptídeos/farmacologia , Antipaína/química , Antipaína/farmacologia , Produtos Biológicos/síntese química , Produtos Biológicos/química , Colúmbia Britânica , Cristalografia por Raios X , Estrutura Molecular , Ressonância Magnética Nuclear Biomolecular , Peptídeos/química
7.
Biochim Biophys Acta ; 1834(12): 2546-53, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23954305

RESUMO

The citrate synthase (CS) of Escherichia coli is an allosteric hexameric enzyme specifically inhibited by NADH. The crystal structure of wild type (WT) E. coli CS, determined by us previously, has no substrates bound, and part of the active site is in a highly mobile region that is shifted from the position needed for catalysis. The CS of Acetobacter aceti has a similar structure, but has been successfully crystallized with bound substrates: both oxaloacetic acid (OAA) and an analog of acetyl coenzyme A (AcCoA). We engineered a variant of E. coli CS wherein five amino acids in the mobile region have been replaced by those in the A. aceti sequence. The purified enzyme shows unusual kinetics with a low affinity for both substrates. Although the crystal structure without ligands is very similar to that of the WT enzyme (except in the mutated region), complexes are formed with both substrates and the allosteric inhibitor NADH. The complex with OAA in the active site identifies a novel OAA-binding residue, Arg306, which has no functional counterpart in other known CS-OAA complexes. This structure may represent an intermediate in a multi-step substrate binding process where Arg306 changes roles from OAA binding to AcCoA binding. The second complex has the substrate analog, S-carboxymethyl-coenzyme A, in the allosteric NADH-binding site and the AcCoA site is not formed. Additional CS variants unable to bind adenylates at the allosteric site show that this second complex is not a factor in positive allosteric activation of AcCoA binding.


Assuntos
Acetobacter/enzimologia , Acetilcoenzima A/química , Citrato (si)-Sintase/química , Proteínas de Escherichia coli/química , Escherichia coli/enzimologia , NADP/química , Acetobacter/genética , Acetilcoenzima A/genética , Acetilcoenzima A/metabolismo , Regulação Alostérica , Animais , Domínio Catalítico , Citrato (si)-Sintase/genética , Citrato (si)-Sintase/metabolismo , Escherichia coli/genética , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , NADP/genética , NADP/metabolismo , Ligação Proteica , Suínos
8.
Glycobiology ; 23(9): 1075-83, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23735230

RESUMO

Trehalose synthase (TreS) catalyzes the reversible conversion of maltose into trehalose in mycobacteria as one of three biosynthetic pathways to this nonreducing disaccharide. Given the importance of trehalose to survival of mycobacteria, there has been considerable interest in understanding the enzymes involved in its production; indeed the structures of the key enzymes in the other two pathways have already been determined. Herein, we present the first structure of TreS from Mycobacterium smegmatis, thereby providing insights into the catalytic machinery involved in this intriguing intramolecular reaction. This structure, which is of interest both mechanistically and as a potential pharmaceutical target, reveals a narrow and enclosed active site pocket within which intramolecular substrate rearrangements can occur. We also present the structure of a complex of TreS with acarbose, revealing a hitherto unsuspected oligosaccharide-binding site within the C-terminal domain. This may well provide an anchor point for the association of TreS with glycogen, thereby enhancing its role in glycogen biosynthesis and degradation.


Assuntos
Acarbose/metabolismo , Glucosiltransferases/química , Glucosiltransferases/metabolismo , Mycobacterium smegmatis/enzimologia , Acarbose/química , Acarbose/farmacologia , Sequência de Aminoácidos , Biocatálise/efeitos dos fármacos , Domínio Catalítico/efeitos dos fármacos , Glucosiltransferases/antagonistas & inibidores , Dados de Sequência Molecular , Ligação Proteica , Conformação Proteica , Alinhamento de Sequência , Relação Estrutura-Atividade
9.
J Biol Chem ; 286(41): 35601-35609, 2011 Oct 14.
Artigo em Inglês | MEDLINE | ID: mdl-21840994

RESUMO

Trehalose synthase (TreS) catalyzes the reversible interconversion of maltose and trehalose and has been shown recently to function primarily in the mobilization of trehalose as a glycogen precursor. Consequently, the mechanism of this intriguing isomerase is of both academic and potential pharmacological interest. TreS catalyzes the hydrolytic cleavage of α-aryl glucosides as well as α-glucosyl fluoride, thereby allowing facile, continuous assays. Reaction of TreS with 5-fluoroglycosyl fluorides results in the trapping of a covalent glycosyl-enzyme intermediate consistent with TreS being a member of the retaining glycoside hydrolase family 13 enzyme family, thus likely following a two-step, double displacement mechanism. This trapped intermediate was subjected to protease digestion followed by LC-MS/MS analysis, and Asp(230) was thereby identified as the catalytic nucleophile. The isomerization reaction was shown to be an intramolecular process by demonstration of the inability of TreS to incorporate isotope-labeled exogenous glucose into maltose or trehalose consistent with previous studies on other TreS enzymes. The absence of a secondary deuterium kinetic isotope effect and the general independence of k(cat) upon leaving group ability both point to a rate-determining conformational change, likely the opening and closing of the enzyme active site.


Assuntos
Proteínas de Bactérias/química , Glucosiltransferases/química , Mycobacterium smegmatis/enzimologia , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Medição da Troca de Deutério , Glucosiltransferases/genética , Glucosiltransferases/metabolismo , Glicosídeo Hidrolases/química , Glicosídeo Hidrolases/genética , Glicosídeo Hidrolases/metabolismo , Cinética , Mycobacterium smegmatis/genética , Especificidade por Substrato
10.
J Clin Invest ; 130(2): 699-714, 2020 02 03.
Artigo em Inglês | MEDLINE | ID: mdl-31845908

RESUMO

HSP27 is highly expressed in, and supports oncogene addiction of, many cancers. HSP27 phosphorylation is a limiting step for activation of this protein and a target for inhibition, but its highly disordered structure challenges rational structure-guided drug discovery. We performed multistep biochemical, structural, and computational experiments to define a spherical 24-monomer complex composed of 12 HSP27 dimers with a phosphorylation pocket flanked by serine residues between their N-terminal domains. Ivermectin directly binds this pocket to inhibit MAPKAP2-mediated HSP27 phosphorylation and depolymerization, thereby blocking HSP27-regulated survival signaling and client-oncoprotein interactions. Ivermectin potentiated activity of anti-androgen receptor and anti-EGFR drugs in prostate and EGFR/HER2-driven tumor models, respectively, identifying a repurposing approach for cotargeting stress-adaptive responses to overcome resistance to inhibitors of oncogenic pathway signaling.


Assuntos
Proteínas de Choque Térmico , Ivermectina , Chaperonas Moleculares , Neoplasias Experimentais , Receptor ErbB-2 , Células A549 , Animais , Proteínas de Choque Térmico/antagonistas & inibidores , Proteínas de Choque Térmico/química , Proteínas de Choque Térmico/genética , Proteínas de Choque Térmico/metabolismo , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/química , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Ivermectina/química , Ivermectina/farmacologia , Camundongos , Chaperonas Moleculares/antagonistas & inibidores , Chaperonas Moleculares/química , Chaperonas Moleculares/genética , Chaperonas Moleculares/metabolismo , Neoplasias Experimentais/tratamento farmacológico , Neoplasias Experimentais/genética , Neoplasias Experimentais/metabolismo , Neoplasias Experimentais/patologia , Domínios Proteicos , Multimerização Proteica , Proteínas Serina-Treonina Quinases/química , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Receptor ErbB-2/genética , Receptor ErbB-2/metabolismo
11.
Biochemistry ; 48(45): 10752-64, 2009 Nov 17.
Artigo em Inglês | MEDLINE | ID: mdl-19803533

RESUMO

While covalent catalytic intermediates of retaining alpha-transglycosylases have been structurally characterized previously, no such information for a hydrolytic alpha-amylase has been obtained. This study presents a new "in situ" enzymatic elongation methodology that, for the first time, has allowed the isolation and structural characterization of a catalytically competent covalent glycosyl-enzyme intermediate with human pancreatic alpha-amylase. This has been achieved by the use of a 5-fluoro-beta-l-idosyl fluoride "warhead" in conjunction with either alpha-maltotriosyl fluoride or 4'-O-methyl-alpha-maltosyl fluoride as elongation agents. This generates an oligosaccharyl-5-fluoroglycosyl fluoride that then reacts with the free enzyme. The resultant covalent intermediates are extremely stable, with hydrolytic half-lives on the order of 240 h for the trisaccharide complex. In the presence of maltose, however, they undergo turnover via transglycosylation according to a half-life of less than 1 h. Structural studies of intermediate complexes unambiguously show the covalent attachment of a 5-fluoro-alpha-l-idosyl moiety in the chair conformation to the side chain of the catalytic nucleophile D197. The elongated portions of the intermediate complexes are found to bind in the high-affinity -2 and -3 binding subsites, forming extensive hydrogen-bonding interactions. Comparative structural analyses with the related noncovalent complex formed by acarbose highlight the structural rigidity of the enzyme surface during catalysis and the key role that substrate conformational flexibility must play in this process. Taken together, the structural data provide atomic details of several key catalytic steps. The scope of this elongation approach to probe the active sites and catalytic mechanisms of alpha-amylases is further demonstrated through preliminary experiments with porcine pancreatic alpha-amylase.


Assuntos
Inibidores Enzimáticos/farmacologia , Pâncreas/enzimologia , alfa-Amilases/química , Animais , Biocatálise , Sequência de Carboidratos , Cromatografia Líquida de Alta Pressão , Ligação de Hidrogênio , Cinética , Espectrometria de Massas , Dados de Sequência Molecular , Suínos , alfa-Amilases/antagonistas & inibidores
12.
Chem Sci ; 10(48): 11073-11077, 2019 Dec 28.
Artigo em Inglês | MEDLINE | ID: mdl-32206255

RESUMO

Simplified analogues of the potent human amylase inhibitor montbretin A were synthesised and shown to bind tightly, K I = 60 and 70 nM, with improved specificity over medically relevant glycosidases, making them promising candidates for controlling blood glucose. Crystallographic analysis confirmed similar binding modes and identified new active site interactions.

13.
ACS Chem Biol ; 14(8): 1751-1759, 2019 08 16.
Artigo em Inglês | MEDLINE | ID: mdl-31241898

RESUMO

De novo macrocyclic peptides, derived using selection technologies such as phage and mRNA display, present unique and unexpected solutions to challenging biological problems. This is due in part to their unusual folds, which are able to present side chains in ways not available to canonical structures such as α-helices and ß-sheets. Despite much recent interest in these molecules, their folding and binding behavior remains poorly characterized. In this work, we present cocrystallization, docking, and solution NMR structures of three de novo macrocyclic peptides that all bind as competitive inhibitors with single-digit nanomolar Ki to the active site of human pancreatic α-amylase. We show that a short stably folded motif in one of these is nucleated by internal hydrophobic interactions in an otherwise dynamic conformation in solution. Comparison of the solution structures with a target-bound structure from docking indicates that stabilization of the bound conformation is provided through interactions with the target protein after binding. These three structures also reveal a surprising functional convergence to present a motif of a single arginine sandwiched between two aromatic residues in the interactions of the peptide with the key catalytic residues of the enzyme, despite little to no other structural homology. Our results suggest that intramolecular hydrophobic interactions are important for priming binding of small macrocyclic peptides to their target and that high rigidity is not necessary for high affinity.


Assuntos
Inibidores Enzimáticos/metabolismo , alfa-Amilases Pancreáticas/antagonistas & inibidores , alfa-Amilases Pancreáticas/metabolismo , Peptídeos Cíclicos/metabolismo , Domínio Catalítico , Cristalização , Humanos , Simulação de Acoplamento Molecular , alfa-Amilases Pancreáticas/química , Ligação Proteica , Conformação Proteica , Dobramento de Proteína
14.
Chembiochem ; 9(3): 433-8, 2008 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-18214874

RESUMO

Specific inhibitors of human pancreatic alpha-amylase (HPA) have potential as oral agents for the control of blood glucose levels in the treatment of diabetes and obesity. In a search for novel inhibitors, a library of 30 000 crude biological extracts of terrestrial and marine origin has been screened. A number of inhibitory extracts were identified, of which the most potent was subjected to bioassay-guided purification. A family of three glycosylated acyl flavonols, montbretins A-C, was thereby identified and characterized as competitive amylase inhibitors, with K(i) values ranging from 8.1-6100 nM. Competitive inhibition by myricetin, which corresponds to the flavone core, and noncompetitive inhibition by a second fragment, ethyl caffeiate, suggest a binding mode for these inhibitors.


Assuntos
Produtos Biológicos/farmacologia , Inibidores Enzimáticos/farmacologia , Pâncreas/enzimologia , alfa-Amilases/antagonistas & inibidores , Glicosilação , Humanos
15.
J Nutr ; 138(4): 685-92, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18356321

RESUMO

The detailed mechanistic aspects for the final starch digestion process leading to effective alpha-glucogenesis by the 2 mucosal alpha-glucosidases, human sucrase-isomaltase complex (SI) and human maltase-glucoamylase (MGAM), are poorly understood. This is due to the structural complexity and vast variety of starches and their intermediate digestion products, the poorly understood enzyme-substrate interactions occurring during the digestive process, and the limited knowledge of the structure-function properties of SI and MGAM. Here we analyzed the basic catalytic properties of the N-terminal subunit of MGAM (ntMGAM) on the hydrolysis of glucan substrates and compared it with those of human native MGAM isolated by immunochemical methods. In relation to native MGAM, ntMGAM displayed slower activity against maltose to maltopentose (G5) series glucose oligomers, as well as maltodextrins and alpha-limit dextrins, and failed to show the strong substrate inhibitory "brake" effect caused by maltotriose, maltotetrose, and G5 on the native enzyme. In addition, the inhibitory constant for acarbose was 2 orders of magnitude higher for ntMGAM than for native MGAM, suggesting lower affinity and/or fewer binding configurations of the active site in the recombinant enzyme. The results strongly suggested that the C-terminal subunit of MGAM has a greater catalytic efficiency due to a higher affinity for glucan substrates and larger number of binding configurations to its active site. Our results show for the first time, to our knowledge, that the C-terminal subunit of MGAM is responsible for the MGAM peptide's "glucoamylase" activity and is the location of the substrate inhibitory brake. In contrast, the membrane-bound ntMGAM subunit contains the poorly inhibitable "maltase" activity of the internally duplicated enzyme.


Assuntos
Inibidores de Glicosídeo Hidrolases , Subunidades Proteicas/química , Amido/metabolismo , alfa-Glucosidases/química , Acarbose , Catálise , Dextrinas/metabolismo , Humanos , Concentração de Íons de Hidrogênio , Oligossacarídeos/metabolismo , Polissacarídeos/metabolismo , Subunidades Proteicas/metabolismo , Proteínas Recombinantes , Complexo Sacarase-Isomaltase/metabolismo , alfa-Glucosidases/metabolismo
16.
J Pediatr Gastroenterol Nutr ; 45(1): 32-43, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17592362

RESUMO

BACKGROUND: Starches are the major source of dietary glucose in weaned children and adults. However, small intestine alpha-glucogenesis by starch digestion is poorly understood due to substrate structural and chemical complexity, as well as the multiplicity of participating enzymes. Our objective was dissection of luminal and mucosal alpha-glucosidase activities participating in digestion of the soluble starch product maltodextrin (MDx). PATIENTS AND METHODS: Immunoprecipitated assays were performed on biopsy specimens and isolated enterocytes with MDx substrate. RESULTS: Mucosal sucrase-isomaltase (SI) and maltase-glucoamylase (MGAM) contributed 85% of total in vitro alpha-glucogenesis. Recombinant human pancreatic alpha-amylase alone contributed <15% of in vitro alpha-glucogenesis; however, alpha-amylase strongly amplified the mucosal alpha-glucogenic activities by preprocessing of starch to short glucose oligomer substrates. At low glucose oligomer concentrations, MGAM was 10 times more active than SI, but at higher concentrations it experienced substrate inhibition whereas SI was not affected. The in vitro results indicated that MGAM activity is inhibited by alpha-amylase digested starch product "brake" and contributes only 20% of mucosal alpha-glucogenic activity. SI contributes most of the alpha-glucogenic activity at higher oligomer substrate concentrations. CONCLUSIONS: MGAM primes and SI activity sustains and constrains prandial alpha-glucogenesis from starch oligomers at approximately 5% of the uninhibited rate. This coupled mucosal mechanism may contribute to highly efficient glucogenesis from low-starch diets and play a role in meeting the high requirement for glucose during children's brain maturation. The brake could play a constraining role on rates of glucose production from higher-starch diets consumed by an older population at risk for degenerative metabolic disorders.


Assuntos
Enterócitos/metabolismo , Glucana 1,4-alfa-Glucosidase/metabolismo , Glucose/metabolismo , Polissacarídeos/metabolismo , alfa-Glucosidases/metabolismo , Animais , Biópsia , Criança , Digestão , Duodeno/enzimologia , Enterócitos/enzimologia , Humanos , Imunoprecipitação , Mucosa Intestinal/enzimologia , Camundongos , Oligo-1,6-Glucosidase/metabolismo , Amido/metabolismo
17.
Cell Chem Biol ; 24(3): 381-390, 2017 Mar 16.
Artigo em Inglês | MEDLINE | ID: mdl-28262556

RESUMO

Human pancreatic α-amylase (HPA) is responsible for degrading starch to malto-oligosaccharides, thence to glucose, and is therefore an attractive therapeutic target for the treatment of diabetes and obesity. Here we report the discovery of a unique lariat nonapeptide, by means of the RaPID (Random non-standard Peptides Integrated Discovery) system, composed of five amino acids in a head-to-side-chain thioether macrocycle and a further four amino acids in a 310 helical C terminus. This is a potent inhibitor of HPA (Ki = 7 nM) yet exhibits selectivity for the target over other glycosidases tested. Structural studies show that this nonapeptide forms a compact tertiary structure, and illustrate that a general inhibitory motif involving two phenolic groups is often accessed for tight binding of inhibitors to HPA. Furthermore, the work reported here demonstrates the potential of this methodology for the discovery of de novo peptide inhibitors against other glycosidases.


Assuntos
Inibidores Enzimáticos/metabolismo , alfa-Amilases Pancreáticas/metabolismo , Peptídeos/metabolismo , Sequência de Aminoácidos , Animais , Sítios de Ligação , Domínio Catalítico , Cristalografia por Raios X , Inibidores Enzimáticos/química , Humanos , Concentração Inibidora 50 , Cinética , Conformação Molecular , Simulação de Dinâmica Molecular , alfa-Amilases Pancreáticas/antagonistas & inibidores , Biblioteca de Peptídeos , Peptídeos/química , Processamento de Proteína Pós-Traducional , RNA de Transferência/química , RNA de Transferência/metabolismo
18.
FEBS Lett ; 590(8): 1143-51, 2016 04.
Artigo em Inglês | MEDLINE | ID: mdl-27000970

RESUMO

As part of a search for selective, mechanism-based covalent inhibitors of human pancreatic α-amylase we describe the chemoenzymatic synthesis of the disaccharide analog α-glucosyl epi-cyclophellitol, demonstrate its stoichiometric reaction with human pancreatic α-amylase and evaluate the time dependence of its inhibition. X-ray crystallographic analysis of the covalent derivative so formed confirms its reaction at the active site with formation of a covalent bond to the catalytic nucleophile D197. The structure illuminates the interactions with the active site and confirms OH4' on the nonreducing end sugar as a good site for attachment of fluorescent tags in generating probes for localization and quantitation of amylase in vivo.


Assuntos
Cicloexanóis/farmacologia , Dissacarídeos/farmacologia , Compostos de Epóxi/farmacologia , alfa-Amilases Pancreáticas/química , alfa-Amilases Pancreáticas/metabolismo , Domínio Catalítico , Simulação por Computador , Humanos , Ligação de Hidrogênio , Inositol/análogos & derivados , Inositol/química , Inositol/metabolismo , Cinética , Espectrometria de Massas , Modelos Moleculares , Água , Difração de Raios X
19.
ACS Cent Sci ; 2(3): 154-161, 2016 Mar 23.
Artigo em Inglês | MEDLINE | ID: mdl-27066537

RESUMO

Selective inhibitors of human pancreatic α-amylase (HPA) are an effective means of controlling blood sugar levels in the management of diabetes. A high-throughput screen of marine natural product extracts led to the identification of a potent (Ki = 10 pM) peptidic HPA inhibitor, helianthamide, from the Caribbean sea anemone Stichodactyla helianthus. Active helianthamide was produced in Escherichia coli via secretion as a barnase fusion protein. X-ray crystallographic analysis of the complex of helianthamide with porcine pancreatic α-amylase revealed that helianthamide adopts a ß-defensin fold and binds into and across the amylase active site, utilizing a contiguous YIYH inhibitory motif. Helianthamide represents the first of a novel class of glycosidase inhibitors and provides an unusual example of functional malleability of the ß-defensin fold, which is rarely seen outside of its traditional role in antimicrobial peptides.

20.
Protein Sci ; 14(3): 743-55, 2005 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15722449

RESUMO

The mechanism of allosteric activation of alpha-amylase by chloride has been studied through structural and kinetic experiments focusing on the chloride-dependent N298S variant of human pancreatic alpha-amylase (HPA) and a chloride-independent TAKA-amylase. Kinetic analysis of the HPA variant clearly demonstrates the pronounced activating effect of chloride ion binding on reaction rates and its effect on the pH-dependence of catalysis. Structural alterations observed in the N298S variant upon chloride ion binding suggest that the chloride ion plays a variety of roles that serve to promote catalysis. One of these is having a strong influence on the positioning of the acid/base catalyst residue E233. Absence of chloride ion results in multiple conformations for this residue and unexpected enzymatic products. Chloride ion and N298 also appear to stabilize a helical region of polypeptide chain from which projects the flexible substrate binding loop unique to chloride-dependent alpha-amylases. This structural feature also serves to properly orient the catalytically essential residue D300. Comparative analyses show that the chloride-independent alpha-amylases compensate for the absence of bound chloride by substituting a hydrophobic core, altering the manner in which substrate interactions are made and shifting the placement of N298. These evolutionary differences presumably arise in response to alternative operating environments or the advantage gained in a particular product profile. Attempts to engineer chloride-dependence into the chloride-independent TAKA-amylase point out the complexity of this system, and the fact that a multitude of factors play a role in binding chloride ion in the chloride-dependent alpha-amylases.


Assuntos
Cloretos/metabolismo , Pâncreas/enzimologia , alfa-Amilases/química , Acarbose/metabolismo , Sequência de Aminoácidos , Substituição de Aminoácidos , Aspergillus oryzae/enzimologia , Aspergillus oryzae/genética , Ativação Enzimática/fisiologia , Humanos , Ligação de Hidrogênio , Cinética , Mutagênese , Pâncreas/metabolismo , Estrutura Terciária de Proteína , Alinhamento de Sequência , alfa-Amilases/genética , alfa-Amilases/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA