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











Intervalo de año de publicación
2.
J Neuroinflammation ; 20(1): 233, 2023 Oct 11.
Artículo en Inglés | MEDLINE | ID: mdl-37817156

RESUMEN

The insulin-degrading enzyme (IDE) is an evolutionarily conserved zinc-dependent metallopeptidase highly expressed in the brain, where its specific functions remain poorly understood. Besides insulin, IDE is able to cleave many substrates in vitro, including amyloid beta peptides, making this enzyme a candidate pathophysiological link between Alzheimer's disease (AD) and type 2 diabetes (T2D). These antecedents led us to address the impact of IDE absence in hippocampus and olfactory bulb. A specific induction of microgliosis was found in the hippocampus of IDE knockout (IDE-KO) mice, without any effects in neither hippocampal volume nor astrogliosis. Performance on hippocampal-dependent memory tests is influenced by IDE gene dose in 12-month-old mice. Furthermore, a comprehensive characterization of the impact of IDE haploinsufficiency and total deletion in metabolic, behavioral, and molecular parameters in the olfactory bulb, a site of high insulin receptor levels, reveals an unambiguous barcode for IDE-KO mice at that age. Using wildtype and IDE-KO primary microglial cultures, we performed a functional analysis at the cellular level. IDE absence alters microglial responses to environmental signals, resulting in impaired modulation of phenotypic states, with only transitory effects on amyloid-ß management. Collectively, our results reveal previously unknown physiological functions for IDE in microglia that, due to cell-compartment topological reasons, cannot be explained by its enzymatic activity, but instead modulate their multidimensional response to various damaging conditions relevant to aging and AD conditions.


Asunto(s)
Enfermedad de Alzheimer , Diabetes Mellitus Tipo 2 , Insulisina , Ratones , Animales , Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Insulisina/genética , Insulisina/metabolismo , Insulisina/farmacología , Microglía/metabolismo , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Encéfalo/metabolismo , Fenotipo
3.
Diabetes Obes Metab ; 25(11): 3268-3278, 2023 11.
Artículo en Inglés | MEDLINE | ID: mdl-37493025

RESUMEN

AIM: To investigate the use of synthetic preimplantation factor (sPIF) as a potential therapeutic tool for improving glucose-stimulated insulin secretion (GSIS), glucose tolerance and insulin sensitivity in the setting of diabetes. MATERIALS AND METHODS: We used a preclinical murine model of type 2 diabetes (T2D) induced by high-fat diet (HFD) feeding for 12 weeks. Saline or sPIF (1 mg/kg/day) was administered to mice by subcutaneously implanted osmotic mini-pumps for 25 days. Glucose tolerance, circulating insulin and C-peptide levels, and GSIS were assessed. In addition, ß-cells (Min-6) were used to test the effects of sPIF on GSIS and insulin-degrading enzyme (IDE) activity in vitro. The effect of sPIF on GSIS was also tested in human islets. RESULTS: GSIS was enhanced 2-fold by sPIF in human islets ex vivo. Furthermore, continuous administration of sPIF to HFD mice increased circulating levels of insulin and improved glucose tolerance, independently of hepatic insulin clearance. Of note, islets isolated from mice treated with sPIF exhibited restored ß-cell function. Finally, genetic (shRNA-IDE) or pharmacological (6bK) inactivation of IDE in Min-6 abolished sPIF-mediated effects on GSIS, showing that both the protein and its protease activity are required for its action. CONCLUSIONS: We conclude that sPIF is a promising secretagogue for the treatment of T2D.


Asunto(s)
Diabetes Mellitus Tipo 2 , Células Secretoras de Insulina , Insulisina , Islotes Pancreáticos , Ratones , Humanos , Animales , Secreción de Insulina , Insulisina/metabolismo , Insulisina/farmacología , Ratones Obesos , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Diabetes Mellitus Tipo 2/metabolismo , Glucosa/metabolismo , Insulina/metabolismo , Células Secretoras de Insulina/metabolismo , Dieta Alta en Grasa/efectos adversos , Islotes Pancreáticos/metabolismo
4.
Front Endocrinol (Lausanne) ; 13: 922825, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35832432

RESUMEN

The primary cilium is a narrow organelle located at the surface of the cell in contact with the extracellular environment. Once underappreciated, now is thought to efficiently sense external environmental cues and mediate cell-to-cell communication, because many receptors, ion channels, and signaling molecules are highly or differentially expressed in primary cilium. Rare genetic disorders that affect cilia integrity and function, such as Bardet-Biedl syndrome and Alström syndrome, have awoken interest in studying the biology of cilium. In this review, we discuss recent evidence suggesting emerging roles of primary cilium and cilia-mediated signaling pathways in the regulation of pancreatic ß- and α-cell functions, and its implications in regulating glucose homeostasis.


Asunto(s)
Células Secretoras de Glucagón , Insulisina , Cilios , Hormonas Pancreáticas , Transducción de Señal/fisiología
5.
Diabetologia ; 65(8): 1375-1389, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35652923

RESUMEN

AIMS/HYPOTHESIS: Type 2 diabetes is characterised by hyperglucagonaemia and perturbed function of pancreatic glucagon-secreting alpha cells but the molecular mechanisms contributing to these phenotypes are poorly understood. Insulin-degrading enzyme (IDE) is present within all islet cells, mostly in alpha cells, in both mice and humans. Furthermore, IDE can degrade glucagon as well as insulin, suggesting that IDE may play an important role in alpha cell function in vivo. METHODS: We have generated and characterised a novel mouse model with alpha cell-specific deletion of Ide, the A-IDE-KO mouse line. Glucose metabolism and glucagon secretion in vivo was characterised; isolated islets were tested for glucagon and insulin secretion; alpha cell mass, alpha cell proliferation and α-synuclein levels were determined in pancreas sections by immunostaining. RESULTS: Targeted deletion of Ide exclusively in alpha cells triggers hyperglucagonaemia and alpha cell hyperplasia, resulting in elevated constitutive glucagon secretion. The hyperglucagonaemia is attributable in part to dysregulation of glucagon secretion, specifically an impaired ability of IDE-deficient alpha cells to suppress glucagon release in the presence of high glucose or insulin. IDE deficiency also leads to α-synuclein aggregation in alpha cells, which may contribute to impaired glucagon secretion via cytoskeletal dysfunction. We showed further that IDE deficiency triggers impairments in cilia formation, inducing alpha cell hyperplasia and possibly also contributing to dysregulated glucagon secretion and hyperglucagonaemia. CONCLUSIONS/INTERPRETATION: We propose that loss of IDE function in alpha cells contributes to hyperglucagonaemia in type 2 diabetes.


Asunto(s)
Diabetes Mellitus Tipo 2 , Células Secretoras de Glucagón , Células Secretoras de Insulina , Insulisina , Animales , Proliferación Celular/genética , Diabetes Mellitus Tipo 2/metabolismo , Glucagón/metabolismo , Células Secretoras de Glucagón/metabolismo , Hiperplasia/genética , Hiperplasia/metabolismo , Insulina/metabolismo , Células Secretoras de Insulina/metabolismo , Insulisina/genética , Insulisina/metabolismo , Ratones , alfa-Sinucleína/metabolismo
6.
Cells ; 11(2)2022 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-35053342

RESUMEN

The insulin-degrading enzyme (IDE) is a zinc-dependent metalloendopeptidase that belongs to the M16A metalloprotease family. IDE is markedly expressed in the brain, where it is particularly relevant due to its in vitro amyloid beta (Aß)-degrading activity. The subcellular localization of IDE, a paramount aspect to understand how this enzyme can perform its proteolytic functions in vivo, remains highly controversial. In this work, we addressed IDE subcellular localization from an evolutionary perspective. Phylogenetic analyses based on protein sequence and gene and protein structure were performed. An in silico analysis of IDE signal peptide suggests an evolutionary shift in IDE exportation at the prokaryote/eukaryote divide. Subcellular localization experiments in microglia revealed that IDE is mostly cytosolic. Furthermore, IDE associates to membranes by their cytoplasmatic side and further partitions between raft and non-raft domains. When stimulated, microglia change into a secretory active state, produces numerous multivesicular bodies and IDE associates with their membranes. The subsequent inward budding of such membranes internalizes IDE in intraluminal vesicles, which later allows IDE to be exported outside the cells in small extracellular vesicles. We further demonstrate that such an IDE exportation mechanism is regulated by stimuli relevant for microglia in physiological conditions and upon aging and neurodegeneration.


Asunto(s)
Evolución Molecular , Insulisina/metabolismo , Microglía/enzimología , Animales , Línea Celular , Células Cultivadas , Secuencia Conservada , Citosol/metabolismo , Vesículas Extracelulares/metabolismo , Insulisina/ultraestructura , Microdominios de Membrana/metabolismo , Metaloendopeptidasas/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Microglía/ultraestructura , Cuerpos Multivesiculares/metabolismo , Filogenia , Fracciones Subcelulares/metabolismo
7.
Cells ; 10(9)2021 09 16.
Artículo en Inglés | MEDLINE | ID: mdl-34572095

RESUMEN

Insulin-degrading enzyme (IDE) is a highly conserved and ubiquitously expressed Zn2+-metallopeptidase that regulates hepatic insulin sensitivity, albeit its regulation in response to the fasting-to-postprandial transition is poorly understood. In this work, we studied the regulation of IDE mRNA and protein levels as well as its proteolytic activity in the liver, skeletal muscle, and kidneys under fasting (18 h) and refeeding (30 min and 3 h) conditions, in mice fed a standard (SD) or high-fat (HFD) diets. In the liver of mice fed an HFD, fasting reduced IDE protein levels (~30%); whereas refeeding increased its activity (~45%) in both mice fed an SD and HFD. Likewise, IDE protein levels were reduced in the skeletal muscle (~30%) of mice fed an HFD during the fasting state. Circulating lactate concentrations directly correlated with hepatic IDE activity and protein levels. Of note, L-lactate in liver lysates augmented IDE activity in a dose-dependent manner. Additionally, IDE protein levels in liver and muscle tissues, but not its activity, inversely correlated (R2 = 0.3734 and 0.2951, respectively; p < 0.01) with a surrogate marker of insulin resistance (HOMA index). Finally, a multivariate analysis suggests that circulating insulin, glucose, non-esterified fatty acids, and lactate levels might be important in regulating IDE in liver and muscle tissues. Our results highlight that the nutritional regulation of IDE in liver and skeletal muscle is more complex than previously expected in mice, and that fasting/refeeding does not strongly influence the regulation of renal IDE.


Asunto(s)
Ayuno , Conducta Alimentaria , Regulación de la Expresión Génica , Insulina/metabolismo , Insulisina/genética , Insulisina/metabolismo , Animales , Dieta Alta en Grasa , Glucosa/metabolismo , Resistencia a la Insulina , Riñón/metabolismo , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Músculo Esquelético/metabolismo , Especificidad de Órganos , Periodo Posprandial
8.
Mol Metab ; 53: 101306, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34298200

RESUMEN

OBJECTIVES: Restenosis after vessel angioplasty due to dedifferentiation of the vascular smooth muscle cells (VSMCs) limits the success of surgical treatment of vascular occlusions. Type 2 diabetes (T2DM) has a major impact on restenosis, with patients exhibiting more aggressive forms of vascular disease and poorer outcomes after surgery. Kv1.3 channels are critical players in VSMC proliferation. Kv1.3 blockers inhibit VSMCs MEK/ERK signalling and prevent vessel restenosis. We hypothesize that dysregulation of microRNAs (miR) play critical roles in adverse remodelling, contributing to Kv1.3 blockers efficacy in T2DM VSMCs. METHODS AND RESULTS: We used clinically relevant in vivo models of vascular risk factors (VRF) and vessels and VSMCs from T2DM patients. RESUKTS: Human T2DM vessels showed increased remodelling, and changes persisted in culture, with augmented VSMCs migration and proliferation. Moreover, there were downregulation of PI3K/AKT/mTOR and upregulation of MEK/ERK pathways, with increased miR-126 expression. The inhibitory effects of Kv1.3 blockers on remodelling were significantly enhanced in T2DM VSMCs and in VRF model. Finally, miR-126 overexpression confered "diabetic" phenotype to non-T2DM VSMCs by downregulating PI3K/AKT axis. CONCLUSIONS: miR-126 plays crucial roles in T2DM VSMC metabolic memory through activation of MEK/ERK pathway, enhancing the efficacy of Kv1.3 blockers in the prevention of restenosis in T2DM patients.


Asunto(s)
Reestenosis Coronaria/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Epigénesis Genética/genética , Canal de Potasio Kv1.3/metabolismo , MicroARNs/metabolismo , Músculo Liso Vascular/metabolismo , Anciano , Animales , Reestenosis Coronaria/tratamiento farmacológico , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Femenino , Humanos , Canal de Potasio Kv1.3/antagonistas & inhibidores , Masculino , Ratones , MicroARNs/genética , Músculo Liso Vascular/efectos de los fármacos , Bloqueadores de los Canales de Potasio/farmacología
9.
Biomedicines ; 9(1)2021 Jan 17.
Artículo en Inglés | MEDLINE | ID: mdl-33477364

RESUMEN

Insulin-degrading enzyme (IDE) is a highly conserved and ubiquitously expressed metalloprotease that degrades insulin and several other intermediate-size peptides. For many decades, IDE had been assumed to be involved primarily in hepatic insulin clearance, a key process that regulates availability of circulating insulin levels for peripheral tissues. Emerging evidence, however, suggests that IDE has several other important physiological functions relevant to glucose and insulin homeostasis, including the regulation of insulin secretion from pancreatic ß-cells. Investigation of mice with tissue-specific genetic deletion of Ide in the liver and pancreatic ß-cells (L-IDE-KO and B-IDE-KO mice, respectively) has revealed additional roles for IDE in the regulation of hepatic insulin action and sensitivity. In this review, we discuss current knowledge about IDE's function as a regulator of insulin secretion and hepatic insulin sensitivity, both evaluating the classical view of IDE as an insulin protease and also exploring evidence for several non-proteolytic functions. Insulin proteostasis and insulin sensitivity have both been highlighted as targets controlling blood sugar levels in type 2 diabetes, so a clearer understanding the physiological functions of IDE in pancreas and liver could led to the development of novel therapeutics for the treatment of this disease.

10.
Metabolism ; 113: 154352, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32916153

RESUMEN

The insulin-degrading enzyme (IDE) is a metalloendopeptidase with a high affinity for insulin. Human genetic polymorphisms in Ide have been linked to increased risk for T2DM. In mice, hepatic Ide ablation causes glucose intolerance and insulin resistance when mice are fed a regular diet. OBJECTIVE: These studies were undertaken to further investigate its regulatory role in glucose homeostasis and insulin sensitivity in diet-induced obesity. METHODS: To this end, we have compared the metabolic effects of loss versus gain of IDE function in mice fed a high-fat diet (HFD). RESULTS: We demonstrate that loss of IDE function in liver (L-IDE-KO mouse) exacerbates hyperinsulinemia and insulin resistance without changes in insulin clearance but in parallel to an increase in pancreatic ß-cell function. Insulin resistance was associated with increased FoxO1 activation and a ~2-fold increase of GLUT2 protein levels in the liver of HFD-fed mice in response to an intraperitoneal injection of insulin. Conversely, gain of IDE function (adenoviral delivery) improves glucose tolerance and insulin sensitivity, in parallel to a reciprocal ~2-fold reduction in hepatic GLUT2 protein levels. Furthermore, in response to insulin, IDE co-immunoprecipitates with the insulin receptor in liver lysates of mice with adenoviral-mediated liver overexpression of IDE. CONCLUSIONS: We conclude that IDE regulates hepatic insulin action and whole-body glucose metabolism in diet-induced obesity via insulin receptor levels.


Asunto(s)
Dieta Alta en Grasa , Glucosa/metabolismo , Homeostasis , Insulina/metabolismo , Insulisina/metabolismo , Hígado/enzimología , Animales , Hígado/metabolismo , Masculino , Ratones , Ratones Obesos
11.
Mar Drugs ; 18(8)2020 Jul 22.
Artículo en Inglés | MEDLINE | ID: mdl-32708004

RESUMEN

Neurodegenerative diseases are age-related disorders caused by progressive neuronal death in different regions of the nervous system. Neuroinflammation, modulated by glial cells, is a crucial event during the neurodegenerative process; consequently, there is an urgency to find new therapeutic products with anti-glioinflammatory properties. Five new furanocembranolides (1-5), along with leptolide, were isolated from two different extracts of Leptogorgia sp., and compound 6 was obtained from chemical transformation of leptolide. Their structures were determined based on spectroscopic evidence. These seven furanocembranolides were screened in vitro by measuring their ability to modulate interleukin-1ß (IL-1ß) production by microglial BV2 cells after LPS (lipopolysaccharide) stimulation. Leptolide and compounds 3, 4 and 6 exhibited clear anti-inflammatory effects on microglial cells, while compound 2 presented a pro-inflammatory outcome. The in vitro results prompted us to assess anti-glioinflammatory effects of leptolide in vivo in a high-fat diet-induced obese mouse model. Interestingly, leptolide treatment ameliorated both microgliosis and astrogliosis in this animal model. Taken together, our results reveal a promising direct biological effect of furanocembranolides on microglial cells as bioactive anti-inflammatory molecules. Among them, leptolide provides us a feasible therapeutic approach to treat neuroinflammation concomitant with metabolic impairment.


Asunto(s)
Antiinflamatorios/farmacología , Encéfalo/efectos de los fármacos , Hidrocarburos Aromáticos con Puentes/farmacología , Diterpenos/farmacología , Furanos/farmacología , Gliosis/tratamiento farmacológico , Resistencia a la Insulina , Microglía/efectos de los fármacos , Obesidad/complicaciones , Animales , Antozoos/química , Antiinflamatorios/química , Antiinflamatorios/aislamiento & purificación , Encéfalo/metabolismo , Encéfalo/patología , Hidrocarburos Aromáticos con Puentes/química , Hidrocarburos Aromáticos con Puentes/aislamiento & purificación , Línea Celular , Dieta Alta en Grasa , Diterpenos/química , Diterpenos/aislamiento & purificación , Furanos/química , Furanos/aislamiento & purificación , Gliosis/etiología , Gliosis/metabolismo , Gliosis/patología , Interleucina-1beta/metabolismo , Masculino , Ratones Endogámicos C57BL , Microglía/metabolismo , Microglía/patología , Estructura Molecular , Obesidad/metabolismo , Relación Estructura-Actividad
12.
Methods Mol Biol ; 2128: 217-224, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32180196

RESUMEN

Insulin resistance in humans and mice is an important hallmark of metabolic diseases. Therefore, assessment of insulin sensitivity/resistance in animal models provides valuable information in the pathophysiology of diabetes and obesity. Depending on the nature of the information required, we can choose between direct and indirect techniques available for the determination of insulin sensitivity. Thus, the complex hyperinsulinemic-euglycemic glucose clamps and the insulin suppression test assess insulin-mediated glucose utilization under steady-state conditions, whereas less complex methods, such as the insulin tolerance test (ITT), rely on measurements of blood glucose levels in animals subjected to intraperitoneal insulin loading. Finally, surrogated indexes derived from blood glucose and plasma insulin levels are also available for assessment of insulin sensitivity/resistance in vivo. In this chapter, we focus on the intraperitoneal insulin tolerance test (IPITT) protocol for measuring insulin resistance in mice.


Asunto(s)
Automonitorización de la Glucosa Sanguínea/métodos , Glucemia/análisis , Inyecciones Intraperitoneales/métodos , Insulina/administración & dosificación , Insulina/sangre , Animales , Modelos Animales de Enfermedad , Insulina/metabolismo , Resistencia a la Insulina , Ratones , Ratones Endogámicos C57BL , Factores de Tiempo
13.
Methods Mol Biol ; 2128: 291-300, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32180200

RESUMEN

Insulin is a hormone produced and secreted by the ß-cells of the pancreatic islets of Langerhans in response to increased blood glucose levels after a meal. The hormone binds to its receptor located on the plasma membrane triggering an intracellular signaling cascade. This signaling pathway is responsible for the pleiotropic actions of insulin on different tissues, such as regulation of glucose and lipid metabolism, proliferation, and differentiation. Although considerable efforts have been made to understand the molecular mechanism linking the action of the hormone to biological processes, our knowledge is incomplete. Of note, under certain conditions, physiological circulating levels of the hormone are insufficient to properly regulate these processes, a term coined as insulin resistance. The ex vivo analysis of insulin action provides valuable information to decipher intracellular signaling events downstream of the insulin receptor under physiological and pathophysiological conditions. In this chapter, we focus on the analysis of intracellular insulin action ex vivo.


Asunto(s)
Técnicas In Vitro/métodos , Resistencia a la Insulina/fisiología , Insulina/farmacología , Receptor de Insulina/metabolismo , Recolección de Tejidos y Órganos/métodos , Animales , Citosol/metabolismo , Insulina/administración & dosificación , Hígado/citología , Hígado/metabolismo , Proteínas de la Membrana/metabolismo , Ratones , Proteínas Nucleares/metabolismo , Fosforilación , Transporte de Proteínas , Transducción de Señal
14.
Am J Physiol Endocrinol Metab ; 317(5): E805-E819, 2019 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-31479304

RESUMEN

Inhibition of insulin-degrading enzyme (IDE) has been proposed as a possible therapeutic target for type 2 diabetes treatment. However, many aspects of IDE's role in glucose homeostasis need to be clarified. In light of this, new preclinical models are required to elucidate the specific role of this protease in the main tissues related to insulin handling. To address this, here we generated a novel line of mice with selective deletion of the Ide gene within pancreatic beta-cells, B-IDE-KO mice, which have been characterized in terms of multiple metabolic end points, including blood glucose, plasma C-peptide, and intraperitoneal glucose tolerance tests. In addition, glucose-stimulated insulin secretion was quantified in isolated pancreatic islets and beta-cell differentiation markers and insulin secretion machinery were characterized by RT-PCR. Additionally, IDE was genetically and pharmacologically inhibited in INS-1E cells and rodent and human islets, and insulin secretion was assessed. Our results show that, in vivo, life-long deletion of IDE from beta-cells results in increased plasma C-peptide levels. Corroborating these findings, isolated islets from B-IDE-KO mice showed constitutive insulin secretion, a hallmark of beta-cell functional immaturity. Unexpectedly, we found 60% increase in Glut1 (a high-affinity/low-Km glucose transporter), suggesting increased glucose transport into the beta-cell at low glucose levels, which may be related to constitutive insulin secretion. In parallel, IDE inhibition in INS-1E and islet cells resulted in impaired insulin secretion after glucose challenge. We conclude that IDE is required for glucose-stimulated insulin secretion. When IDE is inhibited, insulin secretion machinery is perturbed, causing either inhibition of insulin release at high glucose concentrations or constitutive secretion.


Asunto(s)
Secreción de Insulina/genética , Células Secretoras de Insulina/metabolismo , Insulisina/metabolismo , Animales , Glucemia/metabolismo , Péptido C/sangre , Femenino , Glucosa/farmacología , Prueba de Tolerancia a la Glucosa , Transportador de Glucosa de Tipo 1/metabolismo , Homeostasis , Humanos , Insulisina/genética , Masculino , Ratones , Ratones Noqueados , ARN Interferente Pequeño/farmacología , Ratas
15.
Chemistry ; 25(39): 9287-9294, 2019 Jul 11.
Artículo en Inglés | MEDLINE | ID: mdl-31087418

RESUMEN

The cyclic depsipeptide cereulide toxin it is a very well-known potassium electrogenic ionophore particularly sensitive to pancreatic beta cells. The mechanistic details of its specific activity are unknown. Here, we describe a series of synthetic substituted cereulide potassium ionophores that cause impressive selective activation of glucose-induced insulin secretion in a constitutive manner in rat insulinoma INS1E cells. Our study demonstrates that the different electroneutral K+ transport mechanism exhibited by the anionic mutant depsipeptides when compared with classical electrogenic cereulides can have an important impact of pharmacological value on glucose-stimulated insulin secretion.


Asunto(s)
Depsipéptidos/farmacología , Secreción de Insulina/efectos de los fármacos , Ionóforos/química , Potasio/química , Animales , Transporte Biológico , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Depsipéptidos/síntesis química , Depsipéptidos/química , Glucosa/farmacología , Células Secretoras de Insulina/citología , Células Secretoras de Insulina/efectos de los fármacos , Células Secretoras de Insulina/metabolismo , Microscopía Confocal , Potasio/metabolismo , Ratas
16.
Nutrients ; 12(1)2019 Dec 29.
Artículo en Inglés | MEDLINE | ID: mdl-31905727

RESUMEN

The worldwide epidemics of obesity and diabetes have been linked to increased sugar consumption in humans. Here, we review fructose and glucose metabolism, as well as potential molecular mechanisms by which excessive sugar consumption is associated to metabolic diseases and insulin resistance in humans. To this end, we focus on understanding molecular and cellular mechanisms of fructose and glucose transport and sensing in the intestine, the intracellular signaling effects of dietary sugar metabolism, and its impact on glucose homeostasis in health and disease. Finally, the peripheral and central effects of dietary sugars on the gut-brain axis will be reviewed.


Asunto(s)
Fructosa/metabolismo , Glucosa/metabolismo , Absorción Intestinal , Enfermedades Metabólicas/epidemiología , Animales , Diabetes Mellitus Tipo 2/epidemiología , Diabetes Mellitus Tipo 2/metabolismo , Azúcares de la Dieta/administración & dosificación , Azúcares de la Dieta/metabolismo , Transportador de Glucosa de Tipo 5/metabolismo , Humanos , Resistencia a la Insulina , Intestino Delgado/metabolismo , Hígado/metabolismo , Enfermedades Metabólicas/metabolismo , Enfermedad del Hígado Graso no Alcohólico/epidemiología , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Obesidad/epidemiología , Obesidad/metabolismo , Transportador 1 de Sodio-Glucosa/metabolismo
17.
Metabolism ; 88: 1-11, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30098324

RESUMEN

The role of insulin-degrading enzyme (IDE), a metalloprotease with high affinity for insulin, in insulin clearance remains poorly understood. OBJECTIVE: This study aimed to clarify whether IDE is a major mediator of insulin clearance, and to define its role in the etiology of hepatic insulin resistance. METHODS: We generated mice with liver-specific deletion of Ide (L-IDE-KO) and assessed insulin clearance and action. RESULTS: L-IDE-KO mice exhibited higher (~20%) fasting and non-fasting plasma glucose levels, glucose intolerance and insulin resistance. This phenotype was associated with ~30% lower plasma membrane insulin receptor levels in liver, as well as ~55% reduction in insulin-stimulated phosphorylation of the insulin receptor, and its downstream signaling molecules, AKT1 and AKT2 (reduced by ~40%). In addition, FoxO1 was aberrantly distributed in cellular nuclei, in parallel with up-regulation of the gluconeogenic genes Pck1 and G6pc. Surprisingly, L-IDE-KO mice showed similar plasma insulin levels and hepatic insulin clearance as control mice, despite reduced phosphorylation of the carcinoembryonic antigen-related cell adhesion molecule 1, which upon its insulin-stimulated phosphorylation, promotes receptor-mediated insulin uptake to be degraded. CONCLUSION: IDE is not a rate-limiting regulator of plasma insulin levels in vivo.


Asunto(s)
Prueba de Tolerancia a la Glucosa , Resistencia a la Insulina , Insulina/sangre , Insulisina/metabolismo , Hígado/enzimología , Hígado/fisiopatología , Animales , Gluconeogénesis/genética , Células Secretoras de Insulina/patología , Insulisina/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal , Regulación hacia Arriba
18.
Histol Histopathol ; 33(11): 1167-1180, 2018 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-29726577

RESUMEN

Insulin Degrading Enzyme (IDE) is an endopeptidase that degrades insulin and glucagon. Ide gene has been associated with type-2 diabetes mellitus (DM2). However, the physiological role(s) of IDE in glucose homeostasis and its potential therapeutic benefit remain not completely known. To contribute in the understanding of IDE's role in glucose metabolism, we analyzed IDE protein level in pancreatic islets from two hyperinsulinemic mouse models, db/db and high-fat diet (HFD) mice, as well as in human islets from DM2 patients treated with oral hypoglycemic agents (OHAs) or insulin. IDE protein level was detected by staining and by western-blot. INS1E cells, rat and human islets were treated with insulin and IDE protein level was studied. We have shown for the first time IDE staining in rodent and human tissue, using the proper negative control, IDE null mouse tissue. Our staining indicates that IDE is expressed in both beta- and alpha-cells, with higher expression in alpha-cells. Db/db and HFD mice islets showed increased IDE protein level. Interestingly, human islets from DM2 patients treated with OHAs showed decreased IDE protein level in beta-cells. Meanwhile, islets from insulin-treated DM2 patients showed augmented IDE protein level compared to OHAs patients, pointing to an upregulation of IDE protein level stimulated by insulin. These data correlate nicely with insulin-stimulated upregulation of IDE in cultured INS1E cells, as well as in rat and human islets. In conclusion, our study shows that IDE is expressed in pancreatic beta- and alpha-cells of both rodents and humans, having higher expression in alpha-cells. Furthermore, insulin stimulates IDE protein level in pancreatic beta-cells. These results may have implications in how DM2 patient's treatment affects their beta-cell function.


Asunto(s)
Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Células Secretoras de Glucagón/enzimología , Hipoglucemiantes/farmacología , Células Secretoras de Insulina/enzimología , Insulina/farmacología , Insulisina/biosíntesis , Islotes Pancreáticos/metabolismo , Animales , Células Cultivadas , Diabetes Mellitus Experimental/tratamiento farmacológico , Diabetes Mellitus Experimental/patología , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Diabetes Mellitus Tipo 2/patología , Humanos , Hipoglucemiantes/uso terapéutico , Insulina/uso terapéutico , Ratones , Regulación hacia Arriba
19.
Mar Drugs ; 16(2)2018 Feb 02.
Artículo en Inglés | MEDLINE | ID: mdl-29393907

RESUMEN

Two new chloro-furanocembranolides (1, 2) and two new 1,4-diketo cembranolides (3, 4) were isolated from the crude extract of Leptogorgia sp. together with a new seco-furanocembranolide (5) and the known Z-deoxypukalide (6), rubifolide (7), scabrolide D (8) and epoxylophodione (9). Their structures were determined based on spectroscopic evidence. Four compounds: 1, 2, 7 and 8 were found to activate the proliferation of pancreatic insulin-producing (beta) cells.


Asunto(s)
Antozoos/química , Hidrocarburos Aromáticos con Puentes/farmacología , Furanos/farmacología , Células Secretoras de Insulina/efectos de los fármacos , Animales , Hidrocarburos Aromáticos con Puentes/química , Hidrocarburos Aromáticos con Puentes/aislamiento & purificación , Línea Celular , Proliferación Celular/efectos de los fármacos , Furanos/química , Furanos/aislamiento & purificación , Espectroscopía de Resonancia Magnética , Modelos Moleculares , Estructura Molecular , Oxidación-Reducción , Ratas
20.
Mar Drugs ; 15(9)2017 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-28914811

RESUMEN

Type 2 diabetes (T2DM) is a complex disease linked to pancreatic beta-cell failure and insulin resistance. Current antidiabetic treatment regimens for T2DM include insulin sensitizers and insulin secretagogues. We have previously demonstrated that leptolide, a member of the furanocembranolides family, promotes pancreatic beta-cell proliferation in mice. Considering the beneficial effects of leptolide in diabetic mice, in this study, we aimed to address the capability of leptolide to improve insulin resistance associated with the pathology of obesity. To this end, we tested the hypothesis that leptolide should protect against fatty acid-induced insulin resistance in hepatocytes. In a time-dependent manner, leptolide (0.1 µM) augmented insulin-stimulated phosphorylation of protein kinase B (PKB) by two-fold above vehicle-treated HepG2 cells. In addition, leptolide (0.1 µM) counteracted palmitate-induced insulin resistance by augmenting by four-fold insulin-stimulated phosphorylation of PKB in HepG2 cells. In vivo, acute intraperitoneal administration of leptolide (0.1 mg/kg and 1 mg/kg) improved glucose tolerance and insulin sensitivity in lean mice. Likewise, prolonged leptolide treatment (0.1 mg/kg) in diet-induced obese mice improved insulin sensitivity. These effects were paralleled with an ~50% increased of insulin-stimulated phosphorylation of PKB in liver and skeletal muscle and reduced circulating pro-inflammatory cytokines in obese mice. We concluded that leptolide significantly improves insulin sensitivity in vitro and in obese mice, suggesting that leptolide may be another potential treatment for T2DM.


Asunto(s)
Antozoos , Diterpenos/farmacología , Furanos/farmacología , Hipoglucemiantes/farmacología , Resistencia a la Insulina , Adolescente , Animales , Diabetes Mellitus Tipo 2/complicaciones , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Diabetes Mellitus Tipo 2/patología , Dieta , Diterpenos/uso terapéutico , Furanos/uso terapéutico , Células Hep G2/efectos de los fármacos , Humanos , Hipoglucemiantes/uso terapéutico , Masculino , Ratones , Ratones Endogámicos C57BL , Obesidad Mórbida/complicaciones
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA