Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 100
Filtrar
1.
Protein Expr Purif ; 222: 106539, 2024 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-38960013

RESUMO

PF11_0189 is a putative insulin degrading enzyme present in Plasmodium falciparum genome. The catalytic domain of PF11_0189 is about 27 kDa. Substrate specificity study shows PF11_0189 acts upon different types of proteins. The substrate specificity is found to be highest when insulin is used as a substrate. Metal dependency study shows highest dependency of PF11_0189 towards zinc metal for its proteolytic activity. Chelation of zinc metal with EDTA shows complete absence of PF11_0189 activity. Peptide inhibitors, P-70 and P-121 from combinatorial peptide library prepared against PF11_0189 show inhibition with an IC50 value of 4.8 µM and 7.5 µM respectively. A proven natural anti-malarial peptide cyclosporin A shows complete inhibition against PF11_0189 with an IC50 value of 0.75 µM suggesting PF11_0189 as a potential target for peptide inhibitors. The study implicates that PF11_0189 is a zinc metalloprotease involved in catalysis of insulin. The study gives a preliminary insight into the mechanism of complications arising from glucose abnormalities during severe malaria.


Assuntos
Insulisina , Plasmodium falciparum , Proteínas de Protozoários , Plasmodium falciparum/enzimologia , Plasmodium falciparum/genética , Insulisina/genética , Insulisina/química , Insulisina/metabolismo , Proteínas de Protozoários/genética , Proteínas de Protozoários/química , Proteínas de Protozoários/metabolismo , Especificidade por Substrato , Insulina/química , Insulina/metabolismo , Insulina/genética , Zinco/química , Zinco/metabolismo , Genoma de Protozoário , Proteínas Recombinantes/genética , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/isolamento & purificação , Expressão Gênica , Clonagem Molecular , Antimaláricos/química , Antimaláricos/farmacologia , Ciclosporina/química , Ciclosporina/farmacologia
2.
Pharm Res ; 39(4): 611-629, 2022 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-35378698

RESUMO

PURPOSE: This review aims to provide a precise perceptive of the insulin-degrading enzyme (IDE) and its relationship to type 2 diabetes (T2D), Alzheimer's disease (AD), obesity, and cardiovascular diseases. The purpose of the current study was to provide clear idea of treating prevalent diseases such as T2D, and AD by molecular pharmacological therapeutics rather than conventional medicinal therapy. METHODS: To achieve the aims, molecular docking was performed using several softwares such as LIGPLOT+, Python, and Protein-Ligand Interaction Profiler with corresponding tools. RESULTS: The IDE is a large zinc-metalloprotease that breakdown numerous pathophysiologically important extracellular substrates, comprising amyloid ß-protein (Aß) and insulin. Recent studies demonstrated that dysregulation of IDE leads to develop AD and T2D. Specifically, IDE regulates circulating insulin in a variety of organs via a degradation-dependent clearance mechanism. IDE is unique because it was subjected to allosteric activation and mediated via an oligomer structure. CONCLUSION: In this review, we summarised the factors that modulate insulin reformation by IDE and interaction of IDE and some recent reports on IDE inhibitors against AD and T2D. We also highlighted the latest signs of progress of the function of IDE and challenges in advancing IDE- targetted therapies against T2D and AD.


Assuntos
Doença de Alzheimer , Diabetes Mellitus Tipo 2 , Insulisina , Doença de Alzheimer/tratamento farmacológico , Peptídeos beta-Amiloides/metabolismo , Doença Crônica , Diabetes Mellitus Tipo 2/tratamento farmacológico , Diabetes Mellitus Tipo 2/metabolismo , Humanos , Insulina/metabolismo , Insulisina/química , Insulisina/metabolismo , Insulisina/uso terapêutico , Simulação de Acoplamento Molecular
3.
Int J Mol Sci ; 23(3)2022 Feb 03.
Artigo em Inglês | MEDLINE | ID: mdl-35163673

RESUMO

Insulin-degrading enzyme (IDE) is a ubiquitously expressed metallopeptidase that degrades insulin and a large panel of amyloidogenic peptides. IDE is thought to be a potential therapeutic target for type-2 diabetes and neurodegenerative diseases, such as Alzheimer's disease. IDE catalytic chamber, known as a crypt, is formed, so that peptides can be enclosed and degraded. However, the molecular mechanism of the IDE function and peptide recognition, as well as its conformation changes, remains elusive. Our study elucidates IDE structural changes and explains how IDE conformational dynamics is important to modulate the catalytic cycle of IDE. In this aim, a free-substrate IDE crystallographic structure (PDB ID: 2JG4) was used to model a complete structure of IDE. IDE stability and flexibility were studied through molecular dynamics (MD) simulations to witness IDE conformational dynamics switching from a closed to an open state. The description of IDE structural changes was achieved by analysis of the cavity and its expansion over time. Moreover, the quasi-harmonic analysis of the hinge connecting IDE domains and the angles formed over the simulations gave more insights into IDE shifts. Overall, our results could guide toward the use of different approaches to study IDE with different substrates and inhibitors, while taking into account the conformational states resolved in our study.


Assuntos
Insulisina/química , Simulação de Dinâmica Molecular , Humanos , Ligação de Hidrogênio , Termodinâmica , Água/química
4.
Biochemistry ; 60(33): 2519-2523, 2021 08 24.
Artigo em Inglês | MEDLINE | ID: mdl-34342986

RESUMO

Insulin-degrading enzyme (IDE) hydrolyzes monomeric polypeptides, including amyloid-ß (Aß) and HIV-1 p6. It also acts as a nonproteolytic chaperone to prevent Aß polymerization. Here we compare interactions of Aß and non-amyloidogenic p6 with IDE. Although both exhibited similar proteolysis rates, the binding kinetics to an inactive IDE characterized using relaxation-based NMR were remarkably different. IDE and Aß formed a sparsely populated complex with a lifetime of milliseconds in which a short hydrophobic cleavage segment of Aß was anchored to IDE. Strikingly, a second and more stable complex was significantly populated with a subsecond lifetime owing to multiple intermolecular contacts between Aß and IDE. By selectively sequestering Aß in this nonproductive complex, IDE likely increases the critical concentration required for fibrillization. In contrast, IDE and p6 formed a transient, submillisecond complex involving a single anchoring p6 motif. Modulation of intermolecular interactions, thus, allows IDE to differentiate between non-amyloidogenic and amyloidogenic substrates.


Assuntos
Peptídeos beta-Amiloides/química , Insulisina/química , Insulisina/metabolismo , Espectroscopia de Ressonância Magnética/métodos , Chaperonas Moleculares/metabolismo , Produtos do Gene gag do Vírus da Imunodeficiência Humana/química , Peptídeos beta-Amiloides/metabolismo , Cinética , Modelos Químicos , Agregados Proteicos , Dobramento de Proteína , Proteólise , Produtos do Gene gag do Vírus da Imunodeficiência Humana/metabolismo
5.
J Enzyme Inhib Med Chem ; 36(1): 183-187, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-33401948

RESUMO

We have applied a recently developed HPLC-MS enzymatic assay to investigate the cryptic peptides generated by the action of the insulin-degrading enzyme (IDE) on some neuropeptides (NPs) involved in the development of tolerance and dependence to opioids. Particularly, the tested NPs are generated from the NPFF precursor (pro-NPFF (A)): NPFF (FLFQPQRF) and NPAF (AGEGLSSPFWSLAAPQRF). The results show that IDE is able to cleave NPFF and NPAF, generating specific cryptic peptides. As IDE is also responsible for the processing of many other peptides in the brain (amyloid beta protein among the others), we have also performed competitive degradation assays using mixtures of insulin and the above mentioned NPs. Data show that insulin is able to slow down the degradation of both NPs tested, whereas, surprisingly, NPAF is able to accelerate insulin degradation, hinting IDE as the possible link responsible of the mutual influence between insulin and NPs metabolism.


Assuntos
Insulina/química , Insulisina/química , Antagonistas de Entorpecentes/química , Neuropeptídeos/química , Oligopeptídeos/química , Fragmentos de Peptídeos/química , Sequência de Aminoácidos , Analgésicos Opioides/efeitos adversos , Animais , Cromatografia Líquida de Alta Pressão , Tolerância a Medicamentos , Humanos , Insulina/metabolismo , Espectrometria de Massas , Neuropeptídeos/metabolismo , Fragmentos de Peptídeos/análise , Proteólise , Ratos , Proteínas Recombinantes/química , Soluções
6.
Nature ; 511(7507): 94-8, 2014 Jul 03.
Artigo em Inglês | MEDLINE | ID: mdl-24847884

RESUMO

Despite decades of speculation that inhibiting endogenous insulin degradation might treat type-2 diabetes, and the identification of IDE (insulin-degrading enzyme) as a diabetes susceptibility gene, the relationship between the activity of the zinc metalloprotein IDE and glucose homeostasis remains unclear. Although Ide(-/-) mice have elevated insulin levels, they exhibit impaired, rather than improved, glucose tolerance that may arise from compensatory insulin signalling dysfunction. IDE inhibitors that are active in vivo are therefore needed to elucidate IDE's physiological roles and to determine its potential to serve as a target for the treatment of diabetes. Here we report the discovery of a physiologically active IDE inhibitor identified from a DNA-templated macrocycle library. An X-ray structure of the macrocycle bound to IDE reveals that it engages a binding pocket away from the catalytic site, which explains its remarkable selectivity. Treatment of lean and obese mice with this inhibitor shows that IDE regulates the abundance and signalling of glucagon and amylin, in addition to that of insulin. Under physiological conditions that augment insulin and amylin levels, such as oral glucose administration, acute IDE inhibition leads to substantially improved glucose tolerance and slower gastric emptying. These findings demonstrate the feasibility of modulating IDE activity as a new therapeutic strategy to treat type-2 diabetes and expand our understanding of the roles of IDE in glucose and hormone regulation.


Assuntos
Glucagon/metabolismo , Hipoglicemiantes/farmacologia , Insulina/metabolismo , Insulisina/antagonistas & inibidores , Polipeptídeo Amiloide das Ilhotas Pancreáticas/metabolismo , Compostos Macrocíclicos/farmacologia , Animais , Sítios de Ligação , Glicemia/metabolismo , Domínio Catalítico , Diabetes Mellitus Tipo 2/tratamento farmacológico , Diabetes Mellitus Tipo 2/genética , Modelos Animais de Doenças , Esvaziamento Gástrico/efeitos dos fármacos , Predisposição Genética para Doença , Teste de Tolerância a Glucose , Hipoglicemiantes/química , Hipoglicemiantes/uso terapêutico , Insulisina/química , Insulisina/genética , Insulisina/metabolismo , Compostos Macrocíclicos/química , Compostos Macrocíclicos/uso terapêutico , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Modelos Moleculares , Obesidade/tratamento farmacológico , Obesidade/metabolismo , Transdução de Sinais/efeitos dos fármacos , Magreza/tratamento farmacológico , Magreza/metabolismo
7.
Proc Natl Acad Sci U S A ; 114(14): E2826-E2835, 2017 04 04.
Artigo em Inglês | MEDLINE | ID: mdl-28325868

RESUMO

Insulin-degrading enzyme (IDE) hydrolyzes bioactive peptides, including insulin, amylin, and the amyloid ß peptides. Polyanions activate IDE toward some substrates, yet an endogenous polyanion activator has not yet been identified. Here we report that inositol phosphates (InsPs) and phosphatdidylinositol phosphates (PtdInsPs) serve as activators of IDE. InsPs and PtdInsPs interact with the polyanion-binding site located on an inner chamber wall of the enzyme. InsPs activate IDE by up to ∼95-fold, affecting primarily Vmax The extent of activation and binding affinity correlate with the number of phosphate groups on the inositol ring, with phosphate positional effects observed. IDE binds PtdInsPs from solution, immobilized on membranes, or presented in liposomes. Interaction with PtdInsPs, likely PtdIns(3)P, plays a role in localizing IDE to endosomes, where the enzyme reportedly encounters physiological substrates. Thus, InsPs and PtdInsPs can serve as endogenous modulators of IDE activity, as well as regulators of its intracellular spatial distribution.


Assuntos
Endossomos/metabolismo , Fosfatos de Inositol/metabolismo , Insulisina/metabolismo , Fosfatidilinositóis/metabolismo , Androstadienos/farmacologia , Animais , Sítios de Ligação , Células COS , Chlorocebus aethiops , Endossomos/efeitos dos fármacos , Ativação Enzimática , Enzimas Imobilizadas/metabolismo , Concentração de Íons de Hidrogênio , Insulisina/química , Insulisina/genética , Lipossomos/química , Lipossomos/metabolismo , Mutação , Wortmanina
8.
Cell Mol Life Sci ; 75(18): 3441-3456, 2018 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-29594388

RESUMO

The interaction of insulin-degrading enzyme (IDE) with the main intracellular proteasome assemblies (i.e, 30S, 26S and 20S) was analyzed by enzymatic activity, mass spectrometry and native gel electrophoresis. IDE was mainly detected in association with assemblies with at least one free 20S end and biochemical investigations suggest that IDE competes with the 19S in vitro. IDE directly binds the 20S and affects its proteolytic activities in a bimodal fashion, very similar in human and yeast 20S, inhibiting at (IDE) ≤ 30 nM and activating at (IDE) ≥ 30 nM. Only an activating effect is observed in a yeast mutant locked in the "open" conformation (i.e., the α-3ΔN 20S), envisaging a possible role of IDE as modulator of the 20S "open"-"closed" allosteric equilibrium. Protein-protein docking in silico proposes that the interaction between IDE and the 20S could involve the C-term helix of the 20S α-3 subunit which regulates the gate opening of the 20S.


Assuntos
Insulisina/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Regulação Alostérica , Linhagem Celular Tumoral , Cromatografia Líquida de Alta Pressão , Células HEK293 , Humanos , Insulisina/química , Cinética , Simulação de Acoplamento Molecular , Eletroforese em Gel de Poliacrilamida Nativa , Complexo de Endopeptidases do Proteassoma/química , Ligação Proteica , Estrutura Quaternária de Proteína , Estrutura Terciária de Proteína , Espectrometria de Massas em Tandem , Leveduras/metabolismo
9.
Biochemistry ; 57(32): 4903-4914, 2018 08 14.
Artigo em Inglês | MEDLINE | ID: mdl-30004674

RESUMO

Insulin-degrading enzyme (IDE) is a 110 kDa chambered zinc metalloendopeptidase that degrades insulin, amyloid ß, and other intermediate-sized aggregation prone peptides that adopt ß-structures. Structural studies of IDE in complex with multiple physiological substrates have suggested a role for hydrophobic and aromatic residues of the IDE active site in substrate binding and catalysis. Here, we examine functional requirements for conserved hydrophobic and aromatic IDE active site residues that are positioned within 4.5 Å of IDE-bound insulin B chain and amyloid ß peptides in the reported crystal structures for the respective enzyme-substrate complexes. Charge, size, hydrophobicity, aromaticity, and other functional group requirements for substrate binding IDE active site residues were examined through mutational analysis of the recombinant human enzyme and enzyme kinetic studies conducted using native and fluorogenic derivatives of human insulin and amyloid ß peptides. A functional requirement for IDE active site residues F115, A140, F141, Y150, W199, F202, F820, and Y831 was established, and specific contributions of residue charge, size, and hydrophobicity to substrate binding, specificity, and proteolysis were demonstrated. IDE mutant alleles that exhibited enhanced or diminished proteolytic activity toward insulin or amyloid ß peptides and derivative substrates were identified.


Assuntos
Insulisina/química , Insulisina/metabolismo , Peptídeos beta-Amiloides/química , Peptídeos beta-Amiloides/metabolismo , Sítios de Ligação , Catálise , Domínio Catalítico , Humanos , Insulina/metabolismo , Cinética , Estrutura Secundária de Proteína , Especificidade por Substrato
10.
Biochemistry ; 56(1): 228-239, 2017 Jan 10.
Artigo em Inglês | MEDLINE | ID: mdl-27982586

RESUMO

The physiological role of insulin-degrading enzyme (IDE) in the intracytosolic clearance of amyloid ß (Aß) and other amyloid-like peptides supports a hypothesis that human IDE hyperactivation could be therapeutically beneficial for the treatment of late-onset Alzheimer's disease (AD). The major challenge standing in the way of this goal is increasing the specific catalytic activity of IDE against the Aß substrate. There were previous indications that the allosteric mode of IDE activity regulation could potentially provide a highly specific path toward degradation of amyloid-like peptides, while not dramatically affecting activity against other substrates. Recently developed theoretical concepts are used here to explore potential allosteric modulation of the IDE activity as a result of single-residue mutations. Five candidates are selected for experimental follow-up and allosteric free energy calculations: Ser137Ala, Lys396Ala, Asp426Ala, Phe807Ala, and Lys898Ala. Our experiments show that three mutations (Ser137Ala, Phe807Ala, and Lys898Ala) decrease the Km of the Aß substrate. Mutation Lys898Ala results in increased catalytic activity of IDE; on the other hand, Lys364Ala does not change the activity and Asp426Ala diminishes it. Quantifying effects of mutations in terms of allosteric free energy, we show that favorable mutations lead to stabilization of the catalytic sites and other function-relevant distal sites as well as increased dynamics of the IDE-N and IDE-C halves that allow efficient substrate entrance and cleavage. A possibility for intramolecular upregulation of IDE activity against amyloid peptides via allosteric mutations calls for further investigations in this direction. Ultimately, we are hopeful it will lead to the development of IDE-based drugs for the treatment of the late-onset form of AD characterized by an overall impairment of Aß clearance.


Assuntos
Regulação Alostérica , Peptídeos beta-Amiloides/metabolismo , Proteínas Amiloidogênicas/metabolismo , Insulisina/metabolismo , Algoritmos , Doença de Alzheimer/enzimologia , Doença de Alzheimer/genética , Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/química , Proteínas Amiloidogênicas/química , Biocatálise , Domínio Catalítico , Biologia Computacional/métodos , Ensaios Enzimáticos/métodos , Humanos , Insulisina/química , Insulisina/genética , Cinética , Modelos Moleculares , Mutação de Sentido Incorreto , Domínios Proteicos , Especificidade por Substrato , Termodinâmica
11.
Biochim Biophys Acta ; 1860(11 Pt A): 2454-2462, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27443962

RESUMO

Insulin-degrading enzyme (IDE) is a zinc metalloprotease, known to degrade insulin peptide and amyloid-beta (Aß); the key protein involved in Alzheimer's disease (AD). Considering the important role played by IDE in disease progression of AD and type 2 diabetes mellitus (T2DM), we endeavored to identify the Caenorhabditis elegans (C. elegans) IDE orthologous genes and test them for their role in AD related outcomes. We employed bioinformatics, reverse genetics and molecular biology approaches towards identification and functional characterization of putative IDE candidates in C. elegans. Using in-silico analysis we have identified seven C. elegans genes that possess HXXEH motif, an identifying marker of IDE. We further carried out functional analysis of the identified genes in Aß expressing C. elegans strain CL4176 [myo-3/Aß1-42 long 3'-UTR] via studying effect on Aß induced toxicity, cholinergic neuroanatomy, content of acetylcholine/acetylcholine-esterase, extent of reactive oxygen species and expression of FOXO transcription factor DAF-16. Our findings reveal that amongst the identified putative IDE orthologs, a functionally uncharacterized gene C28F5.4 had a profound effect on the tested endpoints. Knocking down C28F5.4 modulated the AD associated conditions by decreasing Aß induced toxicity, severely compromising cholinergic neuroanatomy, reducing expression of acetylcholine-transporter, decreasing acetylcholine content, elevating ROS, with no effect on DAF-16 stress-response protein. These studies provide crucial insight into the structural/functional orthology of IDEs across human and nematode species and further our understanding of the involvement of these proteins and insulin pathway in AD. Further studies could aid in identifying novel drug-targets and in understanding the common modulating factors between AD and T2DM.


Assuntos
Proteínas de Caenorhabditis elegans/genética , Caenorhabditis elegans/genética , Insulisina/genética , Acetilcolina/metabolismo , Doença de Alzheimer/genética , Doença de Alzheimer/metabolismo , Motivos de Aminoácidos , Peptídeos beta-Amiloides/genética , Peptídeos beta-Amiloides/metabolismo , Animais , Caenorhabditis elegans/enzimologia , Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/química , Proteínas de Caenorhabditis elegans/metabolismo , Domínio Catalítico , Insulisina/química , Insulisina/metabolismo , Espécies Reativas de Oxigênio/metabolismo
12.
J Biol Chem ; 290(33): 20044-59, 2015 Aug 14.
Artigo em Inglês | MEDLINE | ID: mdl-26085101

RESUMO

Insulin-degrading enzyme (IDE, insulysin) is the best characterized catabolic enzyme implicated in proteolysis of insulin. Recently, a peptide inhibitor of IDE has been shown to affect levels of insulin, amylin, and glucagon in vivo. However, IDE(-/-) mice display variable phenotypes relating to fasting plasma insulin levels, glucose tolerance, and insulin sensitivity depending on the cohort and age of animals. Here, we interrogated the importance of IDE-mediated catabolism on insulin clearance in vivo. Using a structure-based design, we linked two newly identified ligands binding at unique IDE exosites together to construct a potent series of novel inhibitors. These compounds do not interact with the catalytic zinc of the protease. Because one of these inhibitors (NTE-1) was determined to have pharmacokinetic properties sufficient to sustain plasma levels >50 times its IDE IC50 value, studies in rodents were conducted. In oral glucose tolerance tests with diet-induced obese mice, NTE-1 treatment improved the glucose excursion. Yet in insulin tolerance tests and euglycemic clamp experiments, NTE-1 did not enhance insulin action or increase plasma insulin levels. Importantly, IDE inhibition with NTE-1 did result in elevated plasma amylin levels, suggesting the in vivo role of IDE action on amylin may be more significant than an effect on insulin. Furthermore, using the inhibitors described in this report, we demonstrate that in HEK cells IDE has little impact on insulin clearance. In total, evidence from our studies supports a minimal role for IDE in insulin metabolism in vivo and suggests IDE may be more important in helping regulate amylin clearance.


Assuntos
Inibidores Enzimáticos/farmacologia , Insulina/metabolismo , Insulisina/antagonistas & inibidores , Animais , Sítios de Ligação , Cristalografia por Raios X , Inibidores Enzimáticos/farmacocinética , Células HEK293 , Humanos , Insulisina/química , Modelos Moleculares , Proteólise
13.
J Biol Chem ; 290(30): 18708-20, 2015 Jul 24.
Artigo em Inglês | MEDLINE | ID: mdl-26041774

RESUMO

We describe a novel insulin-degrading enzyme, SidC, that contributes to the proliferation of the human bacterial pathogen Vibrio vulnificus in a mouse model. SidC is phylogenetically distinct from other known insulin-degrading enzymes and is expressed and secreted specifically during host infection. Purified SidC causes a significant decrease in serum insulin levels and an increase in blood glucose levels in mice. A comparison of mice infected with wild type V. vulnificus or an isogenic sidC-deletion strain showed that wild type bacteria proliferated to higher levels. Additionally, hyperglycemia leads to increased proliferation of V. vulnificus in diabetic mice. Consistent with these observations, the sid operon was up-regulated in response to low glucose levels through binding of the cAMP-receptor protein (CRP) complex to a region upstream of the operon. We conclude that glucose levels are important for the survival of V. vulnificus in the host, and that this pathogen uses SidC to actively manipulate host endocrine signals, making the host environment more favorable for bacterial survival and growth.


Assuntos
Proliferação de Células/genética , Interações Hospedeiro-Patógeno/genética , Insulisina/genética , Camundongos Endogâmicos NOD/genética , Vibrio vulnificus/enzimologia , Animais , Glicemia/metabolismo , Diabetes Mellitus Experimental/enzimologia , Diabetes Mellitus Experimental/microbiologia , Diabetes Mellitus Experimental/patologia , Modelos Animais de Doenças , Regulação Bacteriana da Expressão Gênica , Humanos , Insulina/sangue , Insulisina/química , Insulisina/isolamento & purificação , Camundongos , Camundongos Endogâmicos NOD/microbiologia , Vibrioses/genética , Vibrioses/microbiologia , Vibrioses/patologia , Vibrio vulnificus/genética , Vibrio vulnificus/patogenicidade
14.
Phys Chem Chem Phys ; 18(36): 24790-24801, 2016 Sep 14.
Artigo em Inglês | MEDLINE | ID: mdl-27711373

RESUMO

Peptide hydrolysis has been involved in a wide range of biological, biotechnological, and industrial applications. In this perspective, the mechanisms of three distinct peptide bond cleaving enzymes, beta secretase (BACE1), insulin degrading enzyme (IDE), and bovine lens leucine aminopeptidase (BILAP), have been discussed. BACE1 is a catalytic Asp dyad [Asp, Asp-] containing aspartyl protease, while IDE and BILAP are mononuclear [Zn(His, His, Glu)] and binuclear [Zn1(Asp, Glu, Asp)-Zn2(Lys, Glu, Asp, Asp)] core possessing metallopeptidases, respectively. Specifically, enzyme-substrate interactions and the roles of metal ion(s), the ligand environment, second coordination shell residues, and the protein environment in the functioning of these enzymes have been elucidated. This information will be useful to design small inhibitors, activators, and synthetic analogues of these enzymes for biomedical, biotechnological, and industrial applications.


Assuntos
Peptídeos/química , Secretases da Proteína Precursora do Amiloide/química , Secretases da Proteína Precursora do Amiloide/metabolismo , Animais , Biocatálise , Domínio Catalítico , Bovinos , Hidrólise , Insulisina/química , Insulisina/metabolismo , Leucil Aminopeptidase/química , Leucil Aminopeptidase/metabolismo , Ligantes , Peptídeos/metabolismo , Estrutura Terciária de Proteína , Termodinâmica , Zinco/química
15.
Proc Natl Acad Sci U S A ; 110(34): 13827-32, 2013 Aug 20.
Artigo em Inglês | MEDLINE | ID: mdl-23922390

RESUMO

Insulin-degrading enzyme (IDE) selectively degrades the monomer of amyloidogenic peptides and contributes to clearance of amyloid ß (Aß). Thus, IDE retards the progression of Alzheimer's disease. IDE possesses an enclosed catalytic chamber that engulfs and degrades its peptide substrates; however, the molecular mechanism of IDE function, including substrate access to the chamber and recognition, remains elusive. Here, we captured a unique IDE conformation by using a synthetic antibody fragment as a crystallization chaperone. An unexpected displacement of a door subdomain creates an ~18-Å opening to the chamber. This swinging-door mechanism permits the entry of short peptides into the catalytic chamber and disrupts the catalytic site within IDE door subdomain. Given the propensity of amyloidogenic peptides to convert into ß-strands for their polymerization into amyloid fibrils, they also use such ß-strands to stabilize the disrupted catalytic site resided at IDE door subdomain for their degradation by IDE. Thus, action of the swinging door allows IDE to recognize amyloidogenicity by substrate-induced stabilization of the IDE catalytic cleft. Small angle X-ray scattering (SAXS) analysis revealed that IDE exists as a mixture of closed and open states. These open states, which are distinct from the swinging door state, permit entry of larger substrates (e.g., Aß, insulin) to the chamber and are preferred in solution. Mutational studies confirmed the critical roles of the door subdomain and hinge loop joining the N- and C-terminal halves of IDE for catalysis. Together, our data provide insights into the conformational changes of IDE that govern the selective destruction of amyloidogenic peptides.


Assuntos
Proteínas Amiloidogênicas/metabolismo , Insulisina/química , Insulisina/metabolismo , Modelos Moleculares , Conformação Proteica , Proteólise , Domínio Catalítico/genética , Domínio Catalítico/fisiologia , Cristalização , Análise Mutacional de DNA , Escherichia coli , Humanos , Fragmentos Fab das Imunoglobulinas/metabolismo , Insulisina/genética , Mutagênese Sítio-Dirigida , Espalhamento a Baixo Ângulo , Ressonância de Plasmônio de Superfície
16.
J Biol Inorg Chem ; 20(1): 101-108, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25450414

RESUMO

The properties of three novel Platinum(II) compounds toward the insulin-degrading enzyme (IDE) enzymatic activity have been investigated under physiological conditions. The rationale of this study resides on previous observations that these compounds, specifically designed and synthesized by some of us, induce apoptosis in various cancer cell lines, whereas IDE has been proposed as a putative oncogene involved in neuroblastoma onset and progression. Two of these compounds, namely [PtCl(O,O'-acac)(DMSO)] and [Pt(O,O'-acac)(γ-acac)(DMS)], display a modulatory behavior, wherefore activation or inhibition of IDE activity occurs over different concentration ranges (suggesting the existence of two binding sites on the enzyme). On the other hand, [Pt(O,O'-acac)(γ-acac)(DMSO)] shows a typical competitive inhibitory pattern, characterized by a meaningful affinity constant (K i  = 0.95 ± 0.21 µM). Although all three compounds induce cell death in neuroblastoma SHSY5Y cells at concentrations exceeding 2 µM, the two modulators facilitate cells' proliferation at concentrations ≤ 1.5 µM, whereas the competitive inhibitor [Pt(O,O'-acac)(γ-acac)(DMSO)] only shows a pro-apoptotic activity at all investigated concentrations. These features render the [Pt(O,O'-acac)(γ-acac)(DMSO)] a promising "lead compound" for the synthesis of IDE-specific inhibitors (not characterized yet) with therapeutic potentiality.


Assuntos
Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Insulisina/química , Compostos Organoplatínicos/farmacologia , Antineoplásicos/química , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Ensaios de Seleção de Medicamentos Antitumorais , Humanos , Insulisina/antagonistas & inibidores , Cinética , Neuroblastoma/tratamento farmacológico , Compostos Organoplatínicos/química
17.
J Chem Inf Model ; 55(1): 72-83, 2015 Jan 26.
Artigo em Inglês | MEDLINE | ID: mdl-25539133

RESUMO

The ability of the insulin-degrading enzyme (IDE) to degrade amyloid-ß 42 (Aß42), a process regulated by ATP, has been studied as an alternative path in the development of drugs against Alzheimer's disease. In this study, we calculated the potential of mean force for the degradation of Aß42 by IDE in the presence and absence of ATP by umbrella sampling with hybrid quantum mechanics and molecular mechanics (QM/MM) calculations, using the SCC-DFTB QM Hamiltonian and Amber ff99SB force field. Results indicate that the reaction occurs in two steps: The first step is characterized by the formation of the intermediate. The second step is characterized by breaking the peptide bond of the substrate, the latter being the rate-determining step. In our simulations, the activation energy barrier in the absence of ATP is 15 ± 2 kcal mol(-1), which is 7 kcal mol(-1) lower than in the presence of ATP, indicating that the presence of the nucleotide decreases the reaction rate by about 10(5) times.


Assuntos
Trifosfato de Adenosina/metabolismo , Peptídeos beta-Amiloides/metabolismo , Insulisina/metabolismo , Modelos Químicos , Modelos Moleculares , Fragmentos de Peptídeos/metabolismo , Trifosfato de Adenosina/química , Peptídeos beta-Amiloides/química , Insulisina/química , Simulação de Dinâmica Molecular , Fragmentos de Peptídeos/química , Conformação Proteica , Teoria Quântica
18.
Proc Natl Acad Sci U S A ; 109(22): 8523-7, 2012 May 29.
Artigo em Inglês | MEDLINE | ID: mdl-22586115

RESUMO

Peptide hormones and neuropeptides have important roles in physiology and therefore the regulation of these bioactive peptides is of great interest. In some cases proteolysis controls the concentrations and signaling of bioactive peptides, and the peptidases that mediate this biochemistry have proven to be extremely successful drug targets. Due to the lack of any general method to identify these peptidases, however, the role of proteolysis in the regulation of most neuropeptides and peptide hormones is unknown. This limitation prompted us to develop an advanced peptidomics-based strategy to identify the peptidases responsible for the proteolysis of significant bioactive peptides. The application of this approach to calcitonin gene-related peptide (CGRP), a neuropeptide associated with blood pressure and migraine, revealed the endogenous CGRP cleavage sites. This information was then used to biochemically purify the peptidase capable of proteolysis of CGRP at those cleavage sites, which led to the identification of insulin-degrading enzyme (IDE) as a candidate CGRP-degrading enzyme. CGRP had not been identified as an IDE substrate before and we tested the physiological relevance of this interaction by quantitative measurements of CGRP using IDE null (IDE(-/-)) mice. In the absence of IDE, full-length CGRP levels are elevated in vivo, confirming IDE as an endogenous CGRP-degrading enzyme. By linking CGRP and IDE, this strategy uncovers a previously unknown pathway for CGRP regulation and characterizes an additional role for IDE. More generally, this work suggests that this may be an effective general strategy for characterizing these pathways and peptidases moving forward.


Assuntos
Peptídeo Relacionado com Gene de Calcitonina/metabolismo , Insulisina/metabolismo , Fragmentos de Peptídeos/metabolismo , Peptídeos/metabolismo , Sequência de Aminoácidos , Animais , Sítios de Ligação , Peptídeo Relacionado com Gene de Calcitonina/química , Cromatografia Líquida , Feminino , Insulisina/química , Insulisina/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Modelos Moleculares , Dados de Sequência Molecular , Fragmentos de Peptídeos/química , Peptídeos/química , Ligação Proteica , Estrutura Terciária de Proteína , Proteólise , Proteômica , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz , Medula Espinal/química , Medula Espinal/metabolismo , Especificidade por Substrato , Espectrometria de Massas em Tandem
19.
EMBO J ; 29(23): 3952-66, 2010 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-20959807

RESUMO

Macrophage inflammatory protein-1 (MIP-1), MIP-1α (CCL3) and MIP-1ß (CCL4) are chemokines crucial for immune responses towards infection and inflammation. Both MIP-1α and MIP-1ß form high-molecular-weight aggregates. Our crystal structures reveal that MIP-1 aggregation is a polymerization process and human MIP-1α and MIP-1ß form rod-shaped, double-helical polymers. Biophysical analyses and mathematical modelling show that MIP-1 reversibly forms a polydisperse distribution of rod-shaped polymers in solution. Polymerization buries receptor-binding sites of MIP-1α, thus depolymerization mutations enhance MIP-1α to arrest monocytes onto activated human endothelium. However, same depolymerization mutations render MIP-1α ineffective in mouse peritoneal cell recruitment. Mathematical modelling reveals that, for a long-range chemotaxis of MIP-1, polymerization could protect MIP-1 from proteases that selectively degrade monomeric MIP-1. Insulin-degrading enzyme (IDE) is identified as such a protease and decreased expression of IDE leads to elevated MIP-1 levels in microglial cells. Our structural and proteomic studies offer a molecular basis for selective degradation of MIP-1. The regulated MIP-1 polymerization and selective inactivation of MIP-1 monomers by IDE could aid in controlling the MIP-1 chemotactic gradient for immune surveillance.


Assuntos
Quimiocina CCL3/química , Quimiocina CCL3/metabolismo , Quimiocina CCL4/química , Quimiocina CCL4/metabolismo , Insulisina/metabolismo , Sequência de Aminoácidos , Animais , Linhagem Celular , Quimiocina CCL3/genética , Quimiocina CCL3/imunologia , Quimiocina CCL4/genética , Quimiocina CCL4/imunologia , Cristalografia por Raios X , Humanos , Insulisina/química , Proteínas Inflamatórias de Macrófagos/química , Proteínas Inflamatórias de Macrófagos/genética , Proteínas Inflamatórias de Macrófagos/imunologia , Proteínas Inflamatórias de Macrófagos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Modelos Moleculares , Dados de Sequência Molecular , Mutação , Polimerização , Ligação Proteica , Conformação Proteica , Multimerização Proteica
20.
J Chem Inf Model ; 54(5): 1380-90, 2014 May 27.
Artigo em Inglês | MEDLINE | ID: mdl-24697863

RESUMO

Regulation of brain levels of the Amyloid-ß 42 (Aß42) polypeptide by IDE has recently been linked with possible routes for new therapies against Alzheimer's disease (AD). One important aspect is the regulatory mechanism of IDE by ATP, which is an IDE activator in degrading small peptides and an inhibitor in degrading larger peptides, such as Aß42. This relationship was investigated in this study. We present molecular dynamics simulations of Aß42 complexed with IDE, in the absence or presence of either ATP or excess Na(+) and Cl(-) ions. Results suggest a previously unreported inhibition mechanism that depends on charge-induced structural modifications in the active site and interactions simultaneously involving ATP, Aß42, and IDE. Such interactions exist only when both ATP and Aß42 are simultaneously present in the catalytic chamber. This mechanism results in allosteric, noncompetitive inhibition with apparent decrease of substrate affinity, in accordance with experiment.


Assuntos
Trifosfato de Adenosina/farmacologia , Insulisina/antagonistas & inibidores , Simulação de Dinâmica Molecular , Inibidores de Proteases/farmacologia , Trifosfato de Adenosina/metabolismo , Regulação Alostérica/efeitos dos fármacos , Peptídeos beta-Amiloides/química , Peptídeos beta-Amiloides/metabolismo , Domínio Catalítico , Insulisina/química , Insulisina/metabolismo , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/metabolismo , Inibidores de Proteases/metabolismo , Proteólise/efeitos dos fármacos , Termodinâmica , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA