Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
FASEB J ; 35(5): e21374, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33835493

RESUMO

Inhibition of insulin-degrading enzyme (IDE) is a possible target for treating diabetes. However, it has not yet evolved into a medical intervention, mainly because most developed inhibitors target the zinc in IDE's catalytic site, potentially causing toxicity to other essential metalloproteases. Since IDE is a cellular receptor for the varicella-zoster virus (VZV), we constructed a VZV-based inhibitor. We computationally characterized its interaction site with IDE showing that the peptide specifically binds inside IDE's central cavity, however, not in close proximity to the zinc ion. We confirmed the peptide's effective inhibition on IDE activity in vitro and showed its efficacy in ameliorating insulin-related defects in types 1 and 2 diabetes mouse models. In addition, we suggest that inhibition of IDE may ameliorate the pro-inflammatory profile of CD4+ T-cells toward insulin. Together, we propose a potential role of a designed VZV-derived peptide to serve as a selectively-targeted and as an efficient diabetes therapy.


Assuntos
Diabetes Mellitus Experimental/terapia , Diabetes Mellitus Tipo 1/terapia , Diabetes Mellitus Tipo 2/terapia , Insulina/metabolismo , Insulisina/antagonistas & inibidores , Fragmentos de Peptídeos/administração & dosagem , Proteínas do Envelope Viral/metabolismo , Animais , Linfócitos T CD4-Positivos/imunologia , Diabetes Mellitus Experimental/etiologia , Diabetes Mellitus Experimental/patologia , Diabetes Mellitus Tipo 1/etiologia , Diabetes Mellitus Tipo 1/patologia , Diabetes Mellitus Tipo 2/etiologia , Diabetes Mellitus Tipo 2/patologia , Inibidores Enzimáticos/administração & dosagem , Feminino , Herpesvirus Humano 3/fisiologia , Insulisina/genética , Insulisina/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NOD , Camundongos Knockout
2.
Molecules ; 25(10)2020 May 16.
Artigo em Inglês | MEDLINE | ID: mdl-32429462

RESUMO

Alzheimer's disease (AD) is the most frequent type of dementia affecting memory, thinking and behaviour. The major hallmark of the disease is pathological neurodegeneration due to abnormal aggregation of Amyloid beta (Aß) peptides generated by ß- and γ-secretases via amyloidogenic pathway. Purpose of the current study was to evaluate the effects of theasaponin E1 on the inhibition of Aß producing ß-, γ-secretases (BACE1, PS1 and NCT) and acetylcholinesterase and activation of the non-amyloidogenic APP processing α-secretase (ADAM10). Additionally, theasaponin E1 effects on Aß degrading and clearing proteins neprilysin and insulin degrading enzyme (IDE). The effect of theasaponin E1 on these crucial enzymes was investigated by RT-PCR, ELISA, western blotting and fluorometric assays using mouse neuroblastoma cells (SweAPP N2a). theasaponin E1 was extracted and purified from green tea seed extract via HPLC, and N2a cells were treated with different concentrations for 24 h. Gene and protein expression in the cells were measured to determine the effects of activation and/or inhibition of theasaponin E1 on ß- and γ-secretases, neprilysin and IDE. Results demonstrated that theasaponin E1 significantly reduced Aß concentration by activation of the α-secretase and neprilysin. The activities of ß- and γ-secretase were reduced in a dose-dependent manner due to downregulation of BACE1, presenilin, and nicastrin. Similarly, theasaponin E1 significantly reduced the activity of acetylcholinesterase. Overall, from the results it is concluded that green tea seed extracted saponin E1 possess therapeutic significance as a neuroprotective natural product recommended for the treatment of Alzheimer's disease.


Assuntos
Camellia sinensis/química , Regulação da Expressão Gênica/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Fármacos Neuroprotetores/farmacologia , Ácido Oleanólico/análogos & derivados , Saponinas/farmacologia , Proteína ADAM10/genética , Proteína ADAM10/metabolismo , Acetilcolinesterase/genética , Acetilcolinesterase/metabolismo , Secretases da Proteína Precursora do Amiloide/antagonistas & inibidores , Secretases da Proteína Precursora do Amiloide/genética , Secretases da Proteína Precursora do Amiloide/metabolismo , Peptídeos beta-Amiloides/antagonistas & inibidores , Peptídeos beta-Amiloides/genética , Peptídeos beta-Amiloides/metabolismo , Animais , Ácido Aspártico Endopeptidases/antagonistas & inibidores , Ácido Aspártico Endopeptidases/genética , Ácido Aspártico Endopeptidases/metabolismo , Linhagem Celular Tumoral , Relação Dose-Resposta a Droga , Insulisina/antagonistas & inibidores , Insulisina/genética , Insulisina/metabolismo , Glicoproteínas de Membrana/antagonistas & inibidores , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Proteínas de Membrana/agonistas , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos , Neprilisina/antagonistas & inibidores , Neprilisina/genética , Neprilisina/metabolismo , Neurônios/metabolismo , Neurônios/patologia , Fármacos Neuroprotetores/isolamento & purificação , Ácido Oleanólico/isolamento & purificação , Ácido Oleanólico/farmacologia , Extratos Vegetais/química , Presenilinas/antagonistas & inibidores , Presenilinas/genética , Presenilinas/metabolismo , Saponinas/isolamento & purificação , Sementes/química , Chá/química
3.
Int J Mol Sci ; 20(18)2019 Sep 10.
Artigo em Inglês | MEDLINE | ID: mdl-31509943

RESUMO

Insulin-degrading enzyme (IDE) was applied to catalyze hydrolysis of Nociceptin/Orphanin 1-16 (OFQ/N) to show the involvement of the enzyme in degradation of neuropeptides engaged in pain transmission. Moreover, IDE degradative action towards insulin (Ins) was inhibited by the OFQ/N fragments, suggesting a possible regulatory mechanism in the central nervous system. It has been found that OFQ/N and Ins affect each other degradation by IDE, although in a different manner. Indeed, while the digestion of OFQ/N is significantly affected by the presence of Ins, the kinetic profile of the Ins hydrolysis is not affected by the presence of OFQ/N. However, the main hydrolytic fragments of OFQ/N produced by IDE exert inhibitory activity towards the IDE-mediated Ins degradation. Here, we present the results indicating that, besides Ins, IDE cleaves neuropeptides and their released fragments act as inhibitors of IDE activity toward Ins. Having in mind that IDE is present in the brain, which also contains Ins receptors, it cannot be excluded that this enzyme indirectly participates in neural communication of pain signals and that neuropeptides involved in pain transmission may contribute to the regulation of IDE activity. Finally, preliminary results on the metabolism of OFQ/N, carried out in the rat spinal cord homogenate in the presence of various inhibitors specific for different classes of proteases, show that OFQ/N proteolysis in rat spinal cord could be due, besides IDE, also to a cysteine protease not yet identified.


Assuntos
Insulina/metabolismo , Insulisina/metabolismo , Peptídeos Opioides/metabolismo , Medula Espinal/metabolismo , Sequência de Aminoácidos , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Cromatografia Líquida/métodos , Insulina/química , Insulisina/antagonistas & inibidores , Espectrometria de Massas/métodos , Neuropeptídeos/química , Neuropeptídeos/metabolismo , Neuropeptídeos/farmacologia , Peptídeos Opioides/química , Dor/prevenção & controle , Medição da Dor/métodos , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/farmacologia , Ratos , Receptor de Insulina/metabolismo , Medula Espinal/efeitos dos fármacos , Nociceptina
4.
Hypertension ; 72(4): 909-917, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-30354710

RESUMO

SDF-1α (stromal cell-derived factor-1α) is a CXCR4-receptor agonist and DPP4 (dipeptidyl peptidase 4) substrate. SDF-1α, particularly when combined with sitagliptin to block the metabolism of SDF-1α by DPP4, stimulates proliferation of cardiac fibroblasts via the CXCR4 receptor; this effect is greater in cells from spontaneously hypertensive rats versus Wistar-Kyoto normotensive rats. Emerging evidence indicates that ubiquitin(1-76) exists in plasma and is a potent CXCR4-receptor agonist. Therefore, we hypothesized that ubiquitin(1-76), similar to SDF-1α, should increase proliferation of cardiac fibroblasts. Contrary to our working hypothesis, ubiquitin(1-76) did not stimulate cardiac fibroblast proliferation, yet unexpectedly antagonized the proproliferative effects of SDF-1α combined with sitagliptin. In this regard, ubiquitin(1-76) was more potent in spontaneously hypertensive versus Wistar-Kyoto cells. In the presence of 6bk (selective inhibitor of insulin-degrading enzyme [IDE]; an enzyme known to convert ubiquitin(1-76) to ubiquitin(1-74)), ubiquitin(1-76) no longer antagonized the proproliferative effects of SDF-1α/sitagliptin. Ubiquitin(1-74) also antagonized the proproliferative effects of SDF-1α/sitagliptin, and this effect of ubiquitin(1-74) was not blocked by 6bk and was >10-fold more potent compared with ubiquitin(1-76). Neither ubiquitin(1-76) nor ubiquitin(1-74) inhibited the proproliferative effects of the non-CXCR4 receptor agonist neuropeptide Y (activates Y1 receptors). Cardiac fibroblasts expressed IDE mRNA, protein, and activity and converted ubiquitin(1-76) to ubiquitin(1-74). Spontaneously hypertensive fibroblasts expressed greater IDE activity. Extracellular ubiquitin(1-76) blocks the proproliferative effects of SDF-1α/sitagliptin via its conversion by IDE to ubiquitin(1-74), a potent CXCR4 antagonist. Thus, IDE inhibitors, particularly when combined with DPP4 inhibitors or hypertension, could increase the risk of cardiac fibrosis.


Assuntos
Proliferação de Células , Quimiocina CXCL12/metabolismo , Fibroblastos , Hipertensão/metabolismo , Insulisina , Miocárdio/patologia , Receptores CXCR4 , Animais , Pressão Sanguínea/fisiologia , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/fisiologia , Células Cultivadas , Dipeptidil Peptidase 4/metabolismo , Inibidores da Dipeptidil Peptidase IV/farmacologia , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Fibrose , Insulisina/antagonistas & inibidores , Insulisina/metabolismo , Neuropeptídeo Y/metabolismo , Ratos , Ratos Endogâmicos SHR , Ratos Endogâmicos WKY , Receptores CXCR4/agonistas , Receptores CXCR4/metabolismo , Transdução de Sinais , Fosfato de Sitagliptina/farmacologia , Ubiquitina/metabolismo
5.
PLoS One ; 13(2): e0193101, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29447281

RESUMO

Insulin-degrading enzyme (IDE) is an atypical zinc-metalloendopeptidase that hydrolyzes insulin and other intermediate-sized peptide hormones, many of which are implicated in skin health and wound healing. Pharmacological inhibitors of IDE administered internally have been shown to slow the breakdown of insulin and thereby potentiate insulin action. Given the importance of insulin and other IDE substrates for a variety of dermatological processes, pharmacological inhibitors of IDE suitable for topical applications would be expected to hold significant therapeutic and cosmetic potential. Existing IDE inhibitors, however, are prohibitively expensive, difficult to synthesize and of undetermined toxicity. Here we used phage display to discover novel peptidic inhibitors of IDE, which were subsequently characterized in vitro and in cell culture assays. Among several peptide sequences tested, a cyclic dodecapeptide dubbed P12-3A was found to potently inhibit the degradation of insulin (Ki = 2.5 ± 0.31 µM) and other substrates by IDE, while also being resistant to degradation, stable in biological milieu, and highly selective for IDE. In cell culture, P12-3A was shown to potentiate several insulin-induced processes, including the transcription, translation and secretion of alpha-1 type I collagen in primary murine skin fibroblasts, and the migration of keratinocytes in a scratch wound migration assay. By virtue of its potency, stability, specificity for IDE, low cost of synthesis, and demonstrated ability to potentiate insulin-induced processes involved in wound healing and skin health, P12-3A holds significant therapeutic and cosmetic potential for topical applications.


Assuntos
Inibidores Enzimáticos/farmacologia , Fibroblastos/efeitos dos fármacos , Insulisina/antagonistas & inibidores , Peptídeos/farmacologia , Animais , Técnicas de Visualização da Superfície Celular , Células Cultivadas , Fibroblastos/enzimologia , Camundongos
6.
Endocrinology ; 157(9): 3462-8, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27404391

RESUMO

Islet amyloid deposition in human type 2 diabetes results in ß-cell loss. These amyloid deposits contain the unique amyloidogenic peptide human islet amyloid polypeptide (hIAPP), which is also a known substrate of the protease insulin-degrading enzyme (IDE). Whereas IDE inhibition has recently been demonstrated to improve glucose metabolism in mice, inhibiting it has also been shown to increase cell death when synthetic hIAPP is applied exogenously to a ß-cell line. Thus, we wanted to determine whether a similar deleterious effect is observed when hIAPP is endogenously produced and secreted from islets. To address this issue, we cultured hIAPP transgenic mouse islets that have the propensity to form amyloid for 48 and 144 hours in 16.7 mM glucose in the presence and absence of the IDE inhibitor 1. At neither time interval did IDE inhibition increase amyloid formation or ß-cell loss. Thus, the inhibition of IDE may represent an approach to improve glucose metabolism in human type 2 diabetes, without inducing amyloid deposition and its deleterious effects.


Assuntos
Amiloide/metabolismo , Insulisina/antagonistas & inibidores , Polipeptídeo Amiloide das Ilhotas Pancreáticas/metabolismo , Ilhotas Pancreáticas/metabolismo , Animais , Apoptose , Diabetes Mellitus Tipo 2/metabolismo , Feminino , Humanos , Insulina/metabolismo , Insulisina/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Camundongos Transgênicos
7.
Thromb Haemost ; 115(5): 1010-24, 2016 05 02.
Artigo em Inglês | MEDLINE | ID: mdl-26842698

RESUMO

PreImplantation factor (PIF) is a 15-amino acid peptide endogenously secreted by viable embryos, regulating/enabling maternal (host) acceptance/tolerance to the "invading" embryo (allograft) all-while preserving maternal immunity to fight infections. Such attributes make PIF a potential therapeutic agent for chronic inflammatory diseases. We investigated whether PIF's immunomodulatory properties prevent progression of atherosclerosis in the hyper-cholesterolaemic ApoE-deficient murine model. Male, high-fat diet fed, ApoE-deficient (ApoE-/-) mice were administered either PBS, scrambled PIF (0.3-3 mg/kg) or PIF (0.3-3 mg/kg) for seven weeks. After treatment, PIF (3 mg/kg)-treated ApoE-/- mice displayed significantly reduced atherosclerosis lesion burden in the aortic sinus and aortic arch, without any effect on lipid profile. PIF also caused a significant reduction in infiltration of macrophages, decreased expression of pro-inflammatory adhesion molecules, cytokines and chemokines in the plaque, and reduced circulating IFN-γ levels. PIF preferentially binds to monocytes/neutrophils. In vitro, PIF attenuated monocyte migration (MCP-1-induced chemotaxis assay) and in vivo in LPS peritonitis model. Also PIF prevented leukocyte extravasation (peritonitis thioglycollate-induced model), demonstrating that PIF exerts its effect in part by modulation of monocyte function. Inhibition of the potassium channel KCNAB3 (Kv1.3) and of the insulin degrading enzyme (IDE) was demonstrated as potential mechanism of PIF's immunomodulatory effects. In conclusion, PIF regulates/lowers inflammation and prevents atherosclerosis development without affecting circulating lipids. Overall our findings establish PIF as a strong immunomodulatory drug candidate for atherosclerosis therapy.


Assuntos
Aterosclerose/prevenção & controle , Fatores Imunológicos/farmacologia , Peptídeos/farmacologia , Animais , Apolipoproteínas E/deficiência , Aterosclerose/patologia , Aterosclerose/fisiopatologia , Linfócitos T CD4-Positivos/efeitos dos fármacos , Linfócitos T CD4-Positivos/imunologia , Linhagem Celular , Dieta Hiperlipídica/efeitos adversos , Humanos , Mediadores da Inflamação/metabolismo , Insulisina/antagonistas & inibidores , Insulisina/genética , Canal de Potássio Kv1.3/antagonistas & inibidores , Canal de Potássio Kv1.3/genética , Lipídeos/sangue , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Monócitos/efeitos dos fármacos , Placa Aterosclerótica/prevenção & controle , RNA Interferente Pequeno/genética
8.
ACS Chem Biol ; 10(12): 2716-24, 2015 Dec 18.
Artigo em Inglês | MEDLINE | ID: mdl-26398879

RESUMO

Many therapeutically important enzymes are present in multiple cellular compartments, where they can carry out markedly different functions; thus, there is a need for pharmacological strategies to selectively manipulate distinct pools of target enzymes. Insulin-degrading enzyme (IDE) is a thiol-sensitive zinc-metallopeptidase that hydrolyzes diverse peptide substrates in both the cytosol and the extracellular space, but current genetic and pharmacological approaches are incapable of selectively inhibiting the protease in specific subcellular compartments. Here, we describe the discovery, characterization, and kinetics-based optimization of potent benzoisothiazolone-based inhibitors that, by virtue of a unique quasi-irreversible mode of inhibition, exclusively inhibit extracellular IDE. The mechanism of inhibition involves nucleophilic attack by a specific active-site thiol of the enzyme on the inhibitors, which bear an isothiazolone ring that undergoes irreversible ring opening with the formation of a disulfide bond. Notably, binding of the inhibitors is reversible under reducing conditions, thus restricting inhibition to IDE present in the extracellular space. The identified inhibitors are highly potent (IC50(app) = 63 nM), nontoxic at concentrations up to 100 µM, and appear to preferentially target a specific cysteine residue within IDE. These novel inhibitors represent powerful new tools for clarifying the physiological and pathophysiological roles of this poorly understood protease, and their unusual mechanism of action should be applicable to other therapeutic targets.


Assuntos
Citosol/química , Sistemas de Liberação de Medicamentos , Inibidores Enzimáticos/química , Espaço Extracelular/enzimologia , Insulisina/antagonistas & inibidores , Compostos de Sulfidrila/farmacologia , Simulação por Computador , Avaliação Pré-Clínica de Medicamentos , Inibidores Enzimáticos/síntese química , Inibidores Enzimáticos/farmacologia , Concentração Inibidora 50 , Antagonistas da Insulina/farmacologia , Insulisina/química , Modelos Biológicos , Estrutura Molecular , Relação Estrutura-Atividade , Compostos de Sulfidrila/química
9.
Cell Cycle ; 14(14): 2293-300, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25945652

RESUMO

Diabetes mellitus type 2 (T2DM), insulin therapy, and hyperinsulinemia are independent risk factors of liver cancer. Recently, the use of a novel inhibitor of insulin degrading enzyme (IDE) was proposed as a new therapeutic strategy in T2DM. However, IDE inhibition might stimulate liver cell proliferation via increased intracellular insulin concentration. The aim of this study was to characterize effects of inhibition of IDE activity in HepG2 hepatoma cells and to analyze liver specific expression of IDE in subjects with T2DM. HepG2 cells were treated with 10 nM insulin for 24 h with or without inhibition of IDE activity using IDE RNAi, and cell transcriptome and proliferation rate were analyzed. Human liver samples (n = 22) were used for the gene expression profiling by microarrays. In HepG2 cells, IDE knockdown changed expression of genes involved in cell cycle and apoptosis pathways. Proliferation rate was lower in IDE knockdown cells than in controls. Microarray analysis revealed the decrease of hepatic IDE expression in subjects with T2DM accompanied by the downregulation of the p53-dependent genes FAS and CCNG2, but not by the upregulation of proliferation markers MKI67, MCM2 and PCNA. Similar results were found in the liver microarray dataset from GEO Profiles database. In conclusion, IDE expression is decreased in liver of subjects with T2DM which is accompanied by the dysregulation of p53 pathway. Prolonged use of IDE inhibitors for T2DM treatment should be carefully tested in animal studies regarding its potential effect on hepatic tumorigenesis.


Assuntos
Proliferação de Células/efeitos dos fármacos , Insulina/farmacologia , Insulisina/metabolismo , Fígado/metabolismo , Adulto , Idoso , Apoptose/efeitos dos fármacos , Estudos de Coortes , Ciclina G2/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/patologia , Feminino , Perfilação da Expressão Gênica , Células Hep G2 , Humanos , Insulisina/antagonistas & inibidores , Insulisina/genética , Antígeno Ki-67/metabolismo , Masculino , Pessoa de Meia-Idade , Componente 2 do Complexo de Manutenção de Minicromossomo/metabolismo , Antígeno Nuclear de Célula em Proliferação/metabolismo , Interferência de RNA , Transcriptoma/efeitos dos fármacos , Receptor fas/metabolismo
10.
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
11.
Cell Metab ; 20(2): 201-3, 2014 Aug 05.
Artigo em Inglês | MEDLINE | ID: mdl-25100059

RESUMO

The insulin-degrading enzyme (IDE) has been identified as a type 2 diabetes and Alzheimer's disease susceptibility gene, though its physiological function remains unclear. Maianti et al. (2014) now propose that an IDE inhibitor may be a promising therapeutic strategy for type 2 diabetes.


Assuntos
Glucagon/metabolismo , Hipoglicemiantes/farmacologia , Insulina/metabolismo , Insulisina/antagonistas & inibidores , Polipeptídeo Amiloide das Ilhotas Pancreáticas/metabolismo , Compostos Macrocíclicos/farmacologia , Animais , Masculino
12.
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
13.
PLoS One ; 9(4): e95399, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24740421

RESUMO

Insulin degrading enzyme (IDE) is a potential drug target in the treatment of type 2 diabetes (T2D). IDE controls circulating insulin through a degradation-dependent clearance mechanism in multiple tissues. However, there is not sufficient information about IDE regulation in obesity. In this study, we test obesity-associated factors and pioglitazone in the regulation of IDE in diet-induced obese (DIO) C57BL/6 mice. The enzyme activity and protein level of IDE were increased in the liver of DIO mice. Pioglitazone (10 mg/kg/day) administration for 2 months significantly enhanced the enzyme activity (75%), protein (180%) and mRNA (100%) of IDE in DIO mice. The pioglitazone-induced changes were coupled with 50% reduction in fasting insulin and 20% reduction in fasting blood glucose. The mechanism of IDE regulation in liver was investigated in the mouse hepatoma cell line (Hepa 1c1c7 cells), in which pioglitazone (5 µM) increased IDE protein and mRNA in a time-dependent manner in an 8 h study. Free fatty acid (palmitate 300 µM) induced IDE protein, but reduced the mRNA. Glucagon induced, and TNF-α decreased IDE protein. Insulin did not exhibit any activity in the same condition. In summary, pioglitazone, FFA and glucagon directly increased, but TNF-α decreased the IDE activity in hepatocytes. The results suggest that IDE activity is regulated in liver by multiple factors in obesity and pioglitazone may induce IDE activity in the control of T2D.


Assuntos
Hipoglicemiantes/farmacologia , Insulisina/genética , Fígado/efeitos dos fármacos , Obesidade/tratamento farmacológico , RNA Mensageiro/genética , Tiazolidinedionas/farmacologia , Animais , Glicemia/metabolismo , Linhagem Celular Tumoral , Dieta Hiperlipídica/efeitos adversos , Expressão Gênica/efeitos dos fármacos , Glucagon/farmacologia , Hepatócitos/citologia , Hepatócitos/efeitos dos fármacos , Hepatócitos/metabolismo , Insulina/metabolismo , Insulina/farmacologia , Insulisina/antagonistas & inibidores , Insulisina/metabolismo , Fígado/metabolismo , Fígado/patologia , Camundongos , Camundongos Endogâmicos C57BL , Obesidade/etiologia , Obesidade/genética , Obesidade/patologia , Ácido Palmítico/farmacologia , Pioglitazona , RNA Mensageiro/metabolismo , Fator de Necrose Tumoral alfa/farmacologia
14.
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
15.
J Med Chem ; 56(6): 2246-55, 2013 Mar 28.
Artigo em Inglês | MEDLINE | ID: mdl-23437776

RESUMO

Insulin-degrading enzyme (IDE) is an atypical zinc-metallopeptidase that degrades insulin and the amyloid ß-protein and is strongly implicated in the pathogenesis of diabetes and Alzheimer's disease. We recently developed the first effective inhibitors of IDE, peptide hydroxamates that, while highly potent and selective, are relatively large (MW > 740) and difficult to synthesize. We present here a facile synthetic route that yields enantiomerically pure derivatives comparable in potency to the parent compounds. Through the generation of truncated variants, we identified a compound with significantly reduced size (MW = 455.5) that nonetheless retains good potency (ki = 78 ± 11 nM) and selectivity for IDE. Notably, the potency of these inhibitors was found to vary as much as 60-fold in a substrate-specific manner, an unexpected finding for active site-directed inhibitors. Collectively, our findings demonstrate that potent, small-molecule IDE inhibitors can be developed that, in certain instances, can be highly substrate selective.


Assuntos
Desenho de Fármacos , Ácidos Hidroxâmicos/química , Ácidos Hidroxâmicos/farmacologia , Insulisina/antagonistas & inibidores , Insulisina/metabolismo , Peptídeos/química , Inibidores Enzimáticos/síntese química , Inibidores Enzimáticos/química , Inibidores Enzimáticos/metabolismo , Inibidores Enzimáticos/farmacologia , Humanos , Ácidos Hidroxâmicos/síntese química , Ácidos Hidroxâmicos/metabolismo , Insulisina/química , Simulação de Acoplamento Molecular , Conformação Proteica , Estereoisomerismo , Especificidade por Substrato
16.
Biochem Biophys Res Commun ; 402(4): 762-6, 2010 Nov 26.
Artigo em Inglês | MEDLINE | ID: mdl-21036154

RESUMO

Subjects with the metabolic syndrome (insulin resistance, glucose intolerance, dyslipidemia, hypertension, etc.) have a relative increase in abdominal fat tissue compared to normal individuals and obesity has also been shown to be associated with a decrease in insulin clearance. The majority of the clearance of insulin is due to the action of insulin-degrading enzyme (IDE) and IDE is present throughout all tissues. Since abdominal fat is increased in obesity we hypothesized that IDE may be altered in the different fat depots. Adipocytes were isolated from fat samples obtained from subjects during elective abdominal surgery. Fat samples were taken from subcutaneous (SQ) and visceral (VIS) sites. Insulin metabolism was compared in adipocytes isolated from SQ and VIS fat tissue. Adipocytes from the VIS site degraded more insulin that those from SQ fat tissue. Inhibitors of cathepsins B and D has no effect on the degradation of insulin, while bacitracin, an inhibitor of IDE, inhibited degradation by approx. 33% in both SQ and VIS adipocytes. These data show that insulin metabolism is relatively greater in VIS than in SQ fat tissue and potentially due to IDE.


Assuntos
Gordura Abdominal/metabolismo , Adipócitos/metabolismo , Tecido Adiposo/metabolismo , Insulina/metabolismo , Tela Subcutânea/metabolismo , Gordura Abdominal/citologia , Tecido Adiposo/citologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Catepsina B/antagonistas & inibidores , Catepsina B/metabolismo , Catepsina D/antagonistas & inibidores , Catepsina D/metabolismo , Feminino , Humanos , Insulisina/antagonistas & inibidores , Insulisina/metabolismo , Masculino , Síndrome Metabólica/metabolismo , Pessoa de Meia-Idade
17.
Metabolism ; 58(8): 1047-9, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19423142

RESUMO

There is evidence in animal studies that free fatty acids (FFA) can decrease protein degradation, but the exact mechanism is not known. We have shown that FFA can inhibit proteasome activity in vitro by interacting with insulin-degrading enzyme. Here we show that FFA can also inhibit the proteasome in whole cells. HepG2 cells were treated with various FFA, and proteasome activity was measured using a cell-permeable substrate for the chymotrypsin-like activity. Octanoic acid, a medium-chain fatty acid, did not affect proteasome activity. However, oleic and linoleic acids inhibited the chymotrypsin-like activity up to 80%, with approximate IC50s of 80 and 40 micromol/L, respectively. Insulin also inhibited but was not additive with the FFA, suggesting that they work through the same mechanism. These results show that the proteasome can be inhibited by FFA in whole cells and suggest that insulin-degrading enzyme may mediate this effect. This mechanism may be applicable to whole animals and represents a means to integrate hormonal and nutrient signals on the control of protein degradation.


Assuntos
Ácidos Graxos não Esterificados/metabolismo , Insulisina/antagonistas & inibidores , Insulisina/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Linhagem Celular Tumoral , Quimotripsina/metabolismo , Ácidos Graxos não Esterificados/farmacologia , Humanos , Concentração Inibidora 50 , Ácido Linoleico/metabolismo , Ácido Oleico/metabolismo , Tripsina/metabolismo
18.
Diabetologia ; 52(8): 1656-64, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19396426

RESUMO

AIMS/HYPOTHESIS: Hepatic insulin degradation decreases in type 2 diabetes. Insulin-degrading enzyme (IDE) plays a key role in insulin degradation and its gene is located in a diabetes-associated chromosomal region. We hypothesised that IDE may be regulated by insulin and/or glucose in a liver cell model. To validate the observed regulation of IDE in vivo, we analysed biopsies of human adipose tissue during different clamp experiments in men. METHODS: Human hepatoma HepG2 cells were incubated in normal (1 g/l) or high (4.5 g/l) glucose medium and treated with insulin for 24 h. Catalytic activity, mRNA and protein levels of IDE were assessed. IDE mRNA levels were measured in biopsies of human subcutaneous adipose tissue before and at 240 min of hyperinsulinaemic, euglycaemic and hyperglycaemic clamps. RESULTS: In HepG2 cells, insulin increased IDE activity under normal glucose conditions with no change in IDE mRNA or protein levels. Under conditions of high glucose, insulin increased mRNA levels of IDE without changes in IDE activity. Both in normal and high glucose medium, insulin increased levels of the catalytically more active 15a IDE isoform compared with the 15b isoform. In subcutaneous adipose tissue, IDE mRNA levels were not significantly upregulated after euglycaemic or hyperglycaemic clamps. CONCLUSIONS/INTERPRETATION: Insulin increases IDE activity in HepG2 cells in normal but not in high glucose conditions. This disturbance cannot be explained by corresponding alterations in IDE protein levels or IDE splicing. The loss of insulin-induced regulation of IDE activity under hyperglycaemia may contribute to the reduced insulin extraction and peripheral hyperinsulinaemia in type 2 diabetes.


Assuntos
Carcinoma Hepatocelular/enzimologia , Glucose/farmacologia , Insulina/farmacologia , Insulisina/metabolismo , Tecido Adiposo/citologia , Biópsia , Western Blotting , Linhagem Celular Tumoral , Primers do DNA , Ativação Enzimática , Humanos , Hipoxantina Fosforribosiltransferase/genética , Insulisina/antagonistas & inibidores , Insulisina/genética , Neoplasias Hepáticas/enzimologia , RNA Ribossômico/genética , RNA Ribossômico/isolamento & purificação , Reação em Cadeia da Polimerase Via Transcriptase Reversa
19.
Biochemistry ; 47(48): 12822-34, 2008 Dec 02.
Artigo em Inglês | MEDLINE | ID: mdl-18986166

RESUMO

Insulin degrading enzyme (IDE) utilizes a large catalytic chamber to selectively bind and degrade peptide substrates such as insulin and amyloid beta (Abeta). Tight interactions with substrates occur at an exosite located approximately 30 A away from the catalytic center that anchors the N-terminus of substrates to facilitate binding and subsequent cleavages at the catalytic site. However, IDE also degrades peptide substrates that are too short to occupy both the catalytic site and the exosite simultaneously. Here, we use kinins as a model system to address the kinetics and regulation of human IDE with short peptides. IDE specifically degrades bradykinin and kallidin at the Pro/Phe site. A 1.9 A crystal structure of bradykinin-bound IDE reveals the binding of bradykinin to the exosite and not to the catalytic site. In agreement with observed high K(m) values, this suggests low affinity of bradykinin for IDE. This structure also provides the molecular basis on how the binding of short peptides at the exosite could regulate substrate recognition. We also found that human IDE is potently inhibited by physiologically relevant concentrations of S-nitrosylation and oxidation agents. Cysteine-directed modifications play a key role, since an IDE mutant devoid of all 13 cysteines is insensitive to the inhibition by S-nitrosoglutathione, hydrogen peroxide, or N-ethylmaleimide. Specifically, cysteine 819 of human IDE is located inside the catalytic chamber pointing toward an extended hydrophobic pocket and is critical for the inactivation. Thiol-directed modification of this residue likely causes local structural perturbation to reduce substrate binding and catalysis.


Assuntos
Cisteína/metabolismo , Insulisina/química , Insulisina/metabolismo , Peptídeos/química , Peptídeos/metabolismo , Alquilação , Substituição de Aminoácidos , Domínio Catalítico , Sequência Conservada , Cristalografia por Raios X , Inibidores Enzimáticos/farmacologia , Humanos , Insulisina/antagonistas & inibidores , Insulisina/genética , Calidina/metabolismo , Maleimidas/química , Maleimidas/metabolismo , Modelos Moleculares , Oxirredução , Processamento de Proteína Pós-Traducional , Especificidade por Substrato , Compostos de Sulfidrila/metabolismo
20.
Curr Alzheimer Res ; 5(2): 212-24, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18393806

RESUMO

The steady state concentration of the Alzheimer's amyloid-beta peptide in the brain represents a balance between its biosynthesis from the transmembrane amyloid precursor protein (APP), its oligomerisation into neurotoxic and stable species and its degradation by a variety of amyloid-degrading enzymes, principally metallopeptidases. These include, among others, neprilysin (NEP) and its homologue endothelin-converting enzyme (ECE), insulysin (IDE), angiotensin-converting enzyme (ACE) and matrix metalloproteinase-9 (MMP-9). In addition, the serine proteinase, plasmin, may participate in extracellular metabolism of the amyloid peptide under regulation of the plasminogen-activator inhibitor. These various amyloid-degrading enzymes have distinct subcellular localizations, and differential responses to aging, oxidative stress and pharmacological agents and their upregulation may provide a novel and viable therapeutic strategy for prevention and treatment of Alzheimer's disease. Potential approaches to manipulate expression levels of the key amyloid-degrading enzymes are highlighted.


Assuntos
Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/enzimologia , Amiloide/metabolismo , Inibidores Enzimáticos/uso terapêutico , Animais , Ácido Aspártico Endopeptidases/metabolismo , Enzimas Conversoras de Endotelina , Fibrinolisina/antagonistas & inibidores , Fibrinolisina/metabolismo , Humanos , Insulisina/antagonistas & inibidores , Insulisina/metabolismo , Metaloproteinase 9 da Matriz/metabolismo , Inibidores de Metaloproteinases de Matriz , Metaloendopeptidases/metabolismo , Neprilisina/antagonistas & inibidores , Neprilisina/metabolismo , Peptidil Dipeptidase A/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA