Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 51
Filtrar
1.
Diabetologia ; 66(10): 1925-1942, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37480416

RESUMEN

AIM/HYPOTHESIS: Hyperglycaemia is associated with alpha cell dysfunction, leading to dysregulated glucagon secretion in type 1 and type 2 diabetes; however, the mechanisms involved are still elusive. The nutrient sensor mammalian target of rapamycin complex 1 (mTORC1) plays a major role in the maintenance of alpha cell mass and function. We studied the regulation of alpha cell mTORC1 by nutrients and its role in the development of hyperglucagonaemia in diabetes. METHODS: Alpha cell mTORC1 activity was assessed by immunostaining for phosphorylation of its downstream target, the ribosomal protein S6, and glucagon, followed by confocal microscopy on pancreatic sections and flow cytometry on dispersed human and mouse islets and the alpha cell line, αTC1-6. Metabolomics and metabolic flux were studied by 13C glucose labelling in 2.8 or 16.7 mmol/l glucose followed by LC-MS analysis. To study the role of mTORC1 in mediating hyperglucagonaemia in diabetes, we generated an inducible alpha cell-specific Rptor knockout in the Akita mouse model of diabetes and tested the effects on glucose tolerance by IPGTT and on glucagon secretion. RESULTS: mTORC1 activity was increased in alpha cells from diabetic Akita mice in parallel to the development of hyperglycaemia and hyperglucagonaemia (two- to eightfold increase). Acute exposure of mouse and human islets to amino acids stimulated alpha cell mTORC1 (3.5-fold increase), whereas high glucose concentrations inhibited mTORC1 (1.4-fold decrease). The mTORC1 response to glucose was abolished in human and mouse diabetic alpha cells following prolonged islet exposure to high glucose levels, resulting in sustained activation of mTORC1, along with increased glucagon secretion. Metabolomics and metabolic flux analysis showed that exposure to high glucose levels enhanced glycolysis, glucose oxidation and the synthesis of glucose-derived amino acids. In addition, chronic exposure to high glucose levels increased the expression of Slc7a2 and Slc38a4, which encode amino acid transporters, as well as the levels of branched-chain amino acids and methionine cycle metabolites (~1.3-fold increase for both). Finally, conditional Rptor knockout in alpha cells from adult diabetic mice inhibited mTORC1, thereby inhibiting glucagon secretion (~sixfold decrease) and improving diabetes, despite persistent insulin deficiency. CONCLUSIONS/INTERPRETATION: Alpha cell exposure to hyperglycaemia enhances amino acid synthesis and transport, resulting in sustained activation of mTORC1, thereby increasing glucagon secretion. mTORC1 therefore plays a major role in mediating alpha cell dysfunction in diabetes. DATA AVAILABILITY: All sequencing data are available from the Gene Expression Omnibus (GEO) repository (accession no. GSE154126; https://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE154126 ).


Asunto(s)
Diabetes Mellitus Experimental , Diabetes Mellitus Tipo 2 , Hiperglucemia , Adulto , Humanos , Animales , Glucagón , Diana Mecanicista del Complejo 1 de la Rapamicina , Glucosa , Mamíferos
2.
Bioorg Med Chem ; 87: 117303, 2023 05 03.
Artículo en Inglés | MEDLINE | ID: mdl-37167713

RESUMEN

Since the 1950's, AMP-kinase (AMPK) has been used as a promising target for the development of antidiabetic drugs against Type 2 diabetes mellitus (T2D). Indeed, the canonical antidiabetic drug metformin recruits, at least partially, AMPK activation for its therapeutic effect. Herein we present design and synthesis of 20 novel relatively polar cyclic and acyclic dithioacetals of 2-(Het)arylchroman-6-carbaldehydes, 2-phenyl-1,4-benzodioxane-6-carbaldehyde, and 2-phenylbenzofuran-5-carbaldehyde, which were developed as potential AMPK activators. Three of the synthesized dithioacetals demonstrated significant enhancement (≥70%) of glucose uptake in rat L6 myotubes. Noteworthy, one of the dithioacetals, namely 4-(6-(1,3-dithian-2-yl)chroman-2-yl)pyridine, exhibited high potency comparing to other molecules. It increased the rate of glucose uptake in rat L6 myotubes and augmented insulin secretion from rat INS-1E cells in pharmacological relevant concentrations (up to 2 µM). Both effects were mediated by activation of AMPK. In addition, the compound showed excellent pharmacokinetic profile in healthy mice, including maximal oral bioavailability. Such bifunctionality (increased glucose uptake and insulin secretion) can be used as a starting point for the development of a novel class of antidiabetic drugs with dual activity that is relevant for T2D treatment.


Asunto(s)
Diabetes Mellitus Tipo 2 , Hipoglucemiantes , Ratas , Ratones , Animales , Hipoglucemiantes/farmacología , Hipoglucemiantes/uso terapéutico , Proteínas Quinasas Activadas por AMP , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Glucosa/farmacología , Línea Celular , Fibras Musculares Esqueléticas , Insulina/farmacología
3.
J Cell Sci ; 131(15)2018 08 06.
Artículo en Inglés | MEDLINE | ID: mdl-30002135

RESUMEN

AMPK-mTORC1 signaling senses nutrient availability, thereby regulating autophagy. Surprisingly, we found that, in ß-cells, the AMPK activator 5-amino-4-imidazolecarboxamide ribofuranoside (AICAR) inhibited, rather than stimulated, autophagy. AICAR is an intermediate in the generation of inosine monophosphate, with subsequent conversion to other purine nucleotides. Adenosine regulated autophagy in a concentration-dependent manner: at high concentrations, it mimicked the AICAR effect on autophagy, whereas at low concentrations it stimulated autophagy through its cognate A1 receptor. Adenosine regulation of autophagy was independent of AMPK or mTORC1 activity. Adenosine kinase (ADK) is the principal enzyme for metabolic adenosine clearance. ADK knockdown and pharmacological inhibition of the enzyme markedly stimulated autophagy in an adenosine A1 receptor-dependent manner. High-concentration adenosine increased insulin secretion in a manner sensitive to treatment with the autophagy inducer Tat-beclin1, and inhibition of autophagy augmented secretion. In conclusion, high concentrations of AICAR or adenosine inhibit autophagy, whereas physiological concentrations of adenosine or inhibition of adenosine clearance by ADK stimulate autophagy via the adenosine receptor. Adenosine might thus be an autocrine regulator of autophagy, independent of AMPK-mTORC1 signaling. Adenosine regulates insulin secretion, in part, through modulation of autophagy.


Asunto(s)
Nucleótidos de Adenina/metabolismo , Autofagia/fisiología , Células Secretoras de Insulina/metabolismo , Proteínas Quinasas Activadas por AMP/metabolismo , Adenosina Trifosfato , Animales , Western Blotting , Línea Celular , Técnica del Anticuerpo Fluorescente , Células Hep G2 , Humanos , Insulina/metabolismo , Células Secretoras de Insulina/citología , Masculino , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Ratones , Ratones Endogámicos C57BL , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal
4.
Diabetes Obes Metab ; 20 Suppl 2: 95-103, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-30230182

RESUMEN

ER stress due to proinsulin misfolding has an important role in the pathophysiology of rare forms of permanent neonatal diabetes (PNDM) and probably also of common type 1 (T1D) and type 2 diabetes (T2D). Accumulation of misfolded proinsulin in the ER stimulates the unfolded protein response (UPR) that may eventually lead to apoptosis through a process called the terminal UPR. However, the ß-cell ER has an incredible ability to cope with accumulation of misfolded proteins; therefore, it is not clear whether in common forms of diabetes the accumulation of misfolded proinsulin exceeds the point of no return in which terminal UPR is activated. Many studies showed that the UPR is altered in both T1D and T2D; however, the observed changes in the expression of different UPR markers are inconsistent and it is not clear whether they reflect an adaptive response to stress or indeed mediate the ß-cell dysfunction of diabetes. Herein, we critically review the literature on the effects of proinsulin misfolding and ER stress on ß-cell dysfunction and loss in diabetes with emphasis on ß-cell dynamics, and discuss the gaps in understanding the role of proinsulin misfolding in the pathophysiology of diabetes.


Asunto(s)
Diferenciación Celular , Diabetes Mellitus/etiología , Células Secretoras de Insulina/fisiología , Proinsulina/fisiología , Pliegue de Proteína , Adaptación Fisiológica/fisiología , Animales , Diferenciación Celular/fisiología , Diabetes Mellitus/fisiopatología , Diabetes Mellitus Tipo 1/etiología , Diabetes Mellitus Tipo 1/fisiopatología , Diabetes Mellitus Tipo 2/etiología , Diabetes Mellitus Tipo 2/fisiopatología , Modelos Animales de Enfermedad , Estrés del Retículo Endoplásmico/fisiología , Humanos , Células Secretoras de Insulina/metabolismo , Ratones , Proinsulina/química , Porcinos
5.
Biochim Biophys Acta Biomembr ; 1859(10): 1900-1910, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28648676

RESUMEN

The rate of glucose influx to skeletal muscles is determined primarily by the number of functional units of glucose transporter-4 (GLUT4) in the myotube plasma membrane. The abundance of GLUT4 in the plasma membrane is tightly regulated by insulin or contractile activity, which employ distinct pathways to translocate GLUT4-rich vesicles from intracellular compartments. Various studies have indicated that GLUT4 intrinsic activity is also regulated by conformational changes and/or interactions with membrane components and intracellular proteins in the vicinity of the plasma membrane. Here we show that the non-metabolizable glucose analog 3-O-methyl-d-glucose (MeGlc) augmented the rate of hexose transport into myotubes by increasing GLUT4 intrinsic activity without altering the content of the transporter in the plasma membrane. This effect was not a consequence of ATP depletion or hyperosmolar stress and did not involve Akt/PKB or AMPK signal transduction pathways. MeGlc reduced the inhibitory potency (increased Ki) of indinavir, a selective inhibitor of GLUT4, in a dose-dependent manner. Kinetic analyses indicate that MeGlc induced changes in GLUT4 or GLUT4 complexes within the plasma membrane, which enhanced the hexose transport activity and reduced the potency of indinavir inhibition. Finally, we present a simple kinetic analysis for screening and discovering low molecular weight compounds that augment GLUT4 activity.


Asunto(s)
3-O-Metilglucosa/farmacología , Transportador de Glucosa de Tipo 4/metabolismo , Fibras Musculares Esqueléticas/metabolismo , Adenosina Trifosfato/metabolismo , Animales , Transporte Biológico/efectos de los fármacos , Transporte Biológico/fisiología , Línea Celular , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Glucosa/metabolismo , Insulina/metabolismo , Cinética , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Contracción Muscular/efectos de los fármacos , Contracción Muscular/fisiología , Fibras Musculares Esqueléticas/efectos de los fármacos , Proteínas Musculares/metabolismo , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología
6.
Pharm Res ; 34(12): 2873-2890, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-28983714

RESUMEN

PURPOSE: A series of novel polycyclic aromatic compounds that augment the rate of glucose uptake in L6 myotubes and increase glucose-stimulated insulin secretion from beta-cells were synthesized. Designing these molecules, we have aimed at the two main pathogenic mechanisms of T2D, deficient insulin secretion and diminished glucose clearance. The ultimate purpose of this work was to create a novel antidiabetic drug candidate with bi-functional mode of action. METHODS: All presented compounds were synthesized, and characterized in house. INS-1E cells and L6 myoblasts were used for the experiments. The rate of glucose uptake, mechanism of action, level of insulin secretion and the druggability of the lead compound were studied. RESULTS: The lead compound (6-(1,3-dithiepan-2-yl)-2-phenylchromane), dose- and time-dependently at the low µM range increased the rate of glucose uptake in L6 myotubes and insulin secretion in INS-1E cells. The compound exerted its effects through the activation of the LKB1 (Liver Kinase B1)-AMPK pathway. In vitro metabolic parameters of this lead compound exhibited good druggability. CONCLUSIONS: We anticipate that bi-functionality (increased rate of glucose uptake and augmented insulin secretion) will allow the lead compound to be a starting point for the development of a novel class of antidiabetic drugs.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Cromanos/farmacología , Glucosa/metabolismo , Hipoglucemiantes/farmacología , Células Secretoras de Insulina/efectos de los fármacos , Insulina/metabolismo , Fibras Musculares Esqueléticas/efectos de los fármacos , Animales , Transporte Biológico/efectos de los fármacos , Línea Celular , Células Cultivadas , Cromanos/química , Descubrimiento de Drogas , Activación Enzimática/efectos de los fármacos , Humanos , Hipoglucemiantes/química , Células Secretoras de Insulina/metabolismo , Fibras Musculares Esqueléticas/metabolismo , Ratas
7.
Diabetologia ; 59(7): 1480-1491, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-26831301

RESUMEN

AIMS/HYPOTHESIS: We studied the role of protein degradation pathways in the regulation of insulin production and secretion and hypothesised that autophagy regulates proinsulin degradation, thereby modulating beta cell function. METHODS: Proinsulin localisation in autophagosomes was demonstrated by confocal and electron microscopy. Autophagy was inhibited by knockdown of autophagy-related (ATG) proteins and using the H(+)-ATPase inhibitor bafilomycin-A1. Proinsulin and insulin content and secretion were assessed in static incubations by ELISA and RIA. RESULTS: Confocal and electron microscopy showed proinsulin localised in autophagosomes and lysosomes. Beta-Atg7 (-/-) mice had proinsulin-containing sequestosome 1 (p62 [also known as SQSTM1])(+) aggregates in beta cells, indicating proinsulin is regulated by autophagy in vivo. Short-term bafilomycin-A1 treatment and ATG5/7 knockdown increased steady-state proinsulin and hormone precursor chromogranin A content. ATG5/7 knockdown also increased glucose- and non-fuel-stimulated insulin secretion. Finally, mutated forms of proinsulin that are irreparably misfolded and trapped in the endoplasmic reticulum are more resistant to degradation by autophagy. CONCLUSIONS/INTERPRETATION: In the beta cell, transport-competent secretory peptide precursors, including proinsulin, are regulated by autophagy, whereas efficient clearance of transport-incompetent mutated forms of proinsulin by alternative degradative pathways may be necessary to avoid beta cell proteotoxicity. Reduction of autophagic degradation of proinsulin increases its residency in the secretory pathway, followed by enhanced secretion in response to stimuli.


Asunto(s)
Autofagia/fisiología , Insulina/metabolismo , Animales , Autofagia/genética , Proteína 7 Relacionada con la Autofagia/genética , Proteína 7 Relacionada con la Autofagia/metabolismo , Western Blotting , Línea Celular , Homeostasis/genética , Homeostasis/fisiología , Humanos , Células Secretoras de Insulina/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microscopía Electrónica , Consumo de Oxígeno/genética , Consumo de Oxígeno/fisiología , Interferencia de ARN/fisiología
8.
J Cell Mol Med ; 19(8): 1887-99, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25754218

RESUMEN

Vascular endothelial cell (VEC) senescence is considered an early event in the development of atherosclerotic lesions. Stressful stimuli, in particular oxidative stress, have been linked to premature senescence in the vasculature. Foam cells are a major source of reactive oxygen species and may play a role in the induction of VEC senescence; hence, we investigated their involvement in the induction of VEC senescence in a co-culture transwell system. Primary bovine aortic endothelial cells, exposed to the secretome of THP-1 monocyte-derived foam cells, were analysed for the induction of senescence. Senescence associated ß-galactosidase activity and the expression of p16 and p21 were increased, whereas phosphorylated retinoblastoma protein was reduced. This senescent phenotype was mediated by 4-hydroxnonenal (4-HNE), a lipid peroxidation product secreted from foam cells; scavenging of 4-HNE in the co-culture medium blunted this effect. Furthermore, both foam cells and 4-HNE increased the expression of the pro-oxidant thioredoxin-interacting protein (TXNIP). Molecular manipulation of TXNIP expression confirmed its involvement in foam cell-induced senescence. Previous studies showed that peroxisome proliferator-activated receptor (PPAR)δ was activated by 4-hydroalkenals, such as 4-HNE. Pharmacological interventions supported the involvement of the 4-HNE-PPARδ axis in the induction of TXNIP and VEC senescence. The association of TXNIP with VEC senescence was further supported by immunofluorescent staining of human carotid plaques in which the expression of both TXNIP and p21 was augmented in endothelial cells. Collectively, these findings suggest that foam cell-released 4-HNE activates PPARδ in VEC, leading to increased TXNIP expression and consequently to senescence.


Asunto(s)
Aldehídos/farmacología , Proteínas Portadoras/metabolismo , Senescencia Celular/efectos de los fármacos , Células Endoteliales/metabolismo , Células Espumosas/metabolismo , Animales , Biomarcadores/metabolismo , Bovinos , Línea Celular , Técnicas de Cocultivo , Células Endoteliales/citología , Células Endoteliales/efectos de los fármacos , Técnica del Anticuerpo Fluorescente , Células Espumosas/citología , Células Espumosas/efectos de los fármacos , Depuradores de Radicales Libres/farmacología , Humanos , Peroxidación de Lípido/efectos de los fármacos , Modelos Biológicos , PPAR delta/metabolismo , Placa Aterosclerótica/metabolismo , Placa Aterosclerótica/patología
9.
JCI Insight ; 8(7)2023 04 10.
Artículo en Inglés | MEDLINE | ID: mdl-36809274

RESUMEN

Diabetes is associated with increased risk for kidney disease, heart failure, and mortality. Sodium-glucose cotransporter 2 inhibitors (SGLT2i) prevent these adverse outcomes; however, the mechanisms involved are not clear. We generated a roadmap of the metabolic alterations that occur in different organs in diabetes and in response to SGLT2i. In vivo metabolic labeling with 13C-glucose in normoglycemic and diabetic mice treated with or without dapagliflozin, followed by metabolomics and metabolic flux analyses, showed that, in diabetes, glycolysis and glucose oxidation are impaired in the kidney, liver, and heart. Treatment with dapagliflozin failed to rescue glycolysis. SGLT2 inhibition increased glucose oxidation in all organs; in the kidney, this was associated with modulation of the redox state. Diabetes was associated with altered methionine cycle metabolism, evident by decreased betaine and methionine levels, whereas treatment with SGLT2i increased hepatic betaine along with decreased homocysteine levels. mTORC1 activity was inhibited by SGLT2i along with stimulation of AMPK in both normoglycemic and diabetic animals, possibly explaining the protective effects against kidney, liver, and heart diseases. Collectively, our findings suggest that SGLT2i induces metabolic reprogramming orchestrated by AMPK-mTORC1 signaling with common and distinct effects in various tissues, with implications for diabetes and aging.


Asunto(s)
Diabetes Mellitus Experimental , Inhibidores del Cotransportador de Sodio-Glucosa 2 , Animales , Ratones , Inhibidores del Cotransportador de Sodio-Glucosa 2/farmacología , Diabetes Mellitus Experimental/tratamiento farmacológico , Transportador 2 de Sodio-Glucosa/metabolismo , Proteínas Quinasas Activadas por AMP/metabolismo , Betaína , Glucosa , Sodio/metabolismo , Metionina
10.
J Cell Mol Med ; 16(3): 594-604, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21564514

RESUMEN

We have recently generated lipophilic D-xylose derivatives that increase the rate of glucose uptake in cultured skeletal muscle cells in an AMP-activated protein kinase (AMPK)-dependent manner. The derivative 2,4:3,5-dibenzylidene-D-xylose-diethyl dithioacetal (EH-36) stimulated the rate of glucose transport by increasing the abundance of glucose transporter-4 in the plasma membrane of cultured myotubes. The present study aimed at investigating potential antihyperglycaemic effects of EH-36 in animal models of diabetes. Two animal models were treated subcutaneously with EH-36: streptozotocin-induced diabetes in C57BL/6 mice (a model of insulin-deficient type 1 diabetes), and spontaneously diabetic KKAy mice (Kuo Kondo rats carrying the A(y) yellow obese gene; insulin-resistant type 2 diabetes). The in vivo biodistribution of glucose in control and treated mice was followed with the glucose analogue 2-deoxy-2-[(18) F]-D-glucose; the rate of glucose uptake in excised soleus muscles was measured with [(3) H]-2-deoxy-D-glucose. Pharmacokinetic parameters were determined by non-compartmental analysis of the in vivo data. The effective blood EH-36 concentration in treated animals was 2 µM. It reduced significantly the blood glucose levels in both types of diabetic mice and also corrected the typical compensatory hyperinsulinaemia of KKAy mice. EH-36 markedly increased glucose transport in vivo into skeletal muscle and heart, but not to adipose tissue. This stimulatory effect was mediated by Thr(172) -phosphorylation in AMPK. Biochemical tests in treated animals and acute toxicological examinations showed that EH-36 was well tolerated and not toxic to the mice. These findings indicate that EH-36 is a promising prototype molecule for the development of novel antidiabetic drugs.


Asunto(s)
Acetales/uso terapéutico , Compuestos de Bencilideno/uso terapéutico , Compuestos Bicíclicos Heterocíclicos con Puentes/uso terapéutico , Diabetes Mellitus Experimental/tratamiento farmacológico , Diabetes Mellitus Tipo 1/tratamiento farmacológico , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Hipoglucemiantes/uso terapéutico , Tioglicósidos/uso terapéutico , Quinasas de la Proteína-Quinasa Activada por el AMP , Acetales/administración & dosificación , Animales , Compuestos de Bencilideno/administración & dosificación , Transporte Biológico/efectos de los fármacos , Glucemia/análisis , Compuestos Bicíclicos Heterocíclicos con Puentes/administración & dosificación , Células Cultivadas , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Transportador de Glucosa de Tipo 4/biosíntesis , Corazón/efectos de los fármacos , Hipoglucemiantes/administración & dosificación , Masculino , Ratones , Ratones Endogámicos C57BL , Fibras Musculares Esqueléticas/efectos de los fármacos , Fibras Musculares Esqueléticas/metabolismo , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/metabolismo , Proteínas Quinasas/metabolismo , Ratas , Ratas Wistar , Tioglicósidos/administración & dosificación , Tritio
11.
Diabetes Obes Metab ; 19 Suppl 1: 3, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28880483
12.
Diabetes ; 71(3): 453-469, 2022 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-34862201

RESUMEN

The dynamic regulation of autophagy in ß-cells by cycles of fasting-feeding and its effects on insulin secretion are unknown. In ß-cells, mechanistic target of rapamycin complex 1 (mTORC1) is inhibited while fasting and is rapidly stimulated during refeeding by a single amino acid, leucine, and glucose. Stimulation of mTORC1 by nutrients inhibited the autophagy initiator ULK1 and the transcription factor TFEB, thereby preventing autophagy when ß-cells were continuously exposed to nutrients. Inhibition of mTORC1 by Raptor knockout mimicked the effects of fasting and stimulated autophagy while inhibiting insulin secretion, whereas moderate inhibition of autophagy under these conditions rescued insulin secretion. These results show that mTORC1 regulates insulin secretion through modulation of autophagy under different nutritional situations. In the fasting state, autophagy is regulated in an mTORC1-dependent manner, and its stimulation is required to keep insulin levels low, thereby preventing hypoglycemia. Reciprocally, stimulation of mTORC1 by elevated leucine and glucose, which is common in obesity, may promote hyperinsulinemia by inhibiting autophagy.


Asunto(s)
Autofagia/fisiología , Células Secretoras de Insulina/fisiología , Diana Mecanicista del Complejo 1 de la Rapamicina/fisiología , Animales , Autofagia/efectos de los fármacos , Línea Celular , Ayuno , Glucosa/farmacología , Humanos , Secreción de Insulina/efectos de los fármacos , Secreción de Insulina/fisiología , Leucina/farmacología , Masculino , Diana Mecanicista del Complejo 1 de la Rapamicina/efectos de los fármacos , Diana Mecanicista del Complejo 1 de la Rapamicina/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Periodo Posprandial/fisiología
14.
Cell Rep ; 32(4): 107954, 2020 07 28.
Artículo en Inglés | MEDLINE | ID: mdl-32726619

RESUMEN

Diabetic kidney disease (DKD) increases the risk for mortality and is the leading cause of end-stage renal disease. Treatment with sodium-glucose cotransporter 2 inhibitors (SGLT2i) attenuates the progression of DKD, especially in patients with advanced kidney disease. Herein, we show that in diabetes, mTORC1 activity is increased in renal proximal tubule cells (RPTCs) along with enhanced tubule-interstitial fibrosis; this is prevented by SGLT2i. Constitutive activation of mTORC1 in RPTCs induces renal fibrosis and failure and abolishes the renal-protective effects of SGLT2i in diabetes. On the contrary, partial inhibition of mTORC1 in RPTCs prevents fibrosis and the decline in renal function. Stimulation of mTORC1 in RPTCs turns on a pro-fibrotic program in the renal cortex, whereas its inhibition in diabetes reverses the alterations in gene expression. We suggest that RPTC mTORC1 is a critical node that mediates kidney dysfunction in diabetes and the protective effects of SGLT2i by regulating fibrogenesis.


Asunto(s)
Nefropatías Diabéticas/fisiopatología , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Inhibidores del Cotransportador de Sodio-Glucosa 2/farmacología , Animales , Diabetes Mellitus Tipo 2/metabolismo , Nefropatías Diabéticas/etiología , Humanos , Hipoglucemiantes/farmacología , Riñón/metabolismo , Fallo Renal Crónico/metabolismo , Túbulos Renales Proximales/efectos de los fármacos , Túbulos Renales Proximales/metabolismo , Túbulos Renales Proximales/fisiopatología , Masculino , Diana Mecanicista del Complejo 1 de la Rapamicina/fisiología , Ratones , Inhibidores del Cotransportador de Sodio-Glucosa 2/metabolismo , Porcinos
15.
Biochim Biophys Acta ; 1783(10): 1929-34, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18486624

RESUMEN

Endocrine cells produce large amounts of one or more peptides. The post-translational control of selective production of a single protein is often unknown. We used 3 unrelated approaches to diminish PKCepsilon in rat islets to evaluate its role in preferential glucose-mediated insulin production. Transfection with siRNA (siR-PKCepsilon) or expression of inactive PKCepsilon (PKCepsilon-KD) resulted in a significant reduction in insulin response to glucose (16.7 mmol/l). Glucose stimulation resulted in concentration of PKCepsilon in the perinuclear region, an area known to be rich in ER-Golgi systems, associated with insulin-containing structures. ss'COP1 (RACK2) is the anchoring protein for PKCepsilon. Glucose-stimulated proinsulin production was diminished by 50% in islets expressing PKCepsilon-KD, and 60% in islets expressing RACK2 binding protein (epsilonV1-2); total protein biosynthesis was not affected. In islets expressing epsilonV1-2, a chase period following glucose stimulus resulted in a reduced proinsulin conversion to mature insulin. We propose that PKCepsilon plays a specific role in mediating the glucose-signal into insulin production: binding to ss'COP1 localizes the activated enzyme to the RER where it modulates the shuttling of proinsulin to the TGN. Subsequently the enzyme may be involved in anterograde trafficking of the prohormone or in its processing within the TGN.


Asunto(s)
Glucosa/farmacología , Células Secretoras de Insulina/efectos de los fármacos , Células Secretoras de Insulina/metabolismo , Insulina/biosíntesis , Proteína Quinasa C-epsilon/metabolismo , Animales , Secuencia de Bases , Células Cultivadas , Activación Enzimática , Regulación Enzimológica de la Expresión Génica , Insulina/metabolismo , Secreción de Insulina , Masculino , Proteína Quinasa C-epsilon/genética , Transporte de Proteínas , Ratas , Ratas Wistar , Transducción de Señal
16.
Elife ; 72018 11 09.
Artículo en Inglés | MEDLINE | ID: mdl-30412050

RESUMEN

Unresolved ER stress followed by cell death is recognized as the main cause of a multitude of pathologies including neonatal diabetes. A systematic analysis of the mechanisms of ß-cell loss and dysfunction in Akita mice, in which a mutation in the proinsulin gene causes a severe form of permanent neonatal diabetes, showed no increase in ß-cell apoptosis throughout life. Surprisingly, we found that the main mechanism leading to ß-cell dysfunction is marked impairment of ß-cell growth during the early postnatal life due to transient inhibition of mTORC1, which governs postnatal ß-cell growth and differentiation. Importantly, restoration of mTORC1 activity in neonate ß-cells was sufficient to rescue postnatal ß-cell growth, and to improve diabetes. We propose a scenario for the development of permanent neonatal diabetes, possibly also common forms of diabetes, where early-life events inducing ER stress affect ß-cell mass expansion due to mTOR inhibition.


Asunto(s)
Diabetes Mellitus/genética , Estrés del Retículo Endoplásmico/genética , Diana Mecanicista del Complejo 1 de la Rapamicina/genética , Proinsulina/genética , Animales , Animales Recién Nacidos/genética , Animales Recién Nacidos/crecimiento & desarrollo , Apoptosis/genética , Diabetes Mellitus/patología , Retículo Endoplásmico/genética , Humanos , Células Secretoras de Insulina/patología , Ratones , Mutación , Pliegue de Proteína
17.
Diabetes ; 55(3): 590-9, 2006 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-16505220

RESUMEN

Glucose metabolism affects most major signal pathways in pancreatic beta-cells. Multiple protein kinases, including protein kinase C (PKC) isoenzymes, are involved in these effects; however, their role is poorly defined. Moreover, the dynamics of kinase isoenzyme activation in reference to the biphasic insulin secretion is unknown. In perfused pancreas of Wistar rats, PKCalpha staining was strongly associated with insulin staining, jointly accumulating in the vicinity of the plasma membrane during early first-phase insulin response. The signal declined before the onset of second phase and reappeared during second-phase insulin release as foci, only weekly associated with insulin staining; this signal persisted for at least 15 min after glucose stimulation. In the GK rat, glucose had minimal effect on beta-cell PKCalpha. In control beta-cells, PKCdelta stained as granulated foci with partial association with insulin staining; however, no glucose-dependent translocation was observed. In the GK rat, only minimal staining for PKCdelta was observed, increasing exclusively during early first-phase secretion. In Wistar beta-cells, PKCepsilon concentrated near the nucleus, strongly associated with insulin staining, with dynamics resembling that of biphasic insulin response, but persisting for 15 min after cessation of stimulation. In GK rats, PKCepsilon staining lacked glucose-dependent changes or association with insulin. PKCzeta exhibited bimodal dynamics in control beta-cells: during early first phase, accumulation near the cell membrane was observed, dispersing thereafter. This was followed by a gradual accumulation near the nucleus; 15 min after glucose stimulus, clear PKCzeta staining was observed within the nucleus. In the GK rat, a similar response was only occasionally observed. In control beta-cells, glucose stimulation led to a transient recruitment of PKCtheta, associated with first-phase insulin release, not seen in GK beta-cell. Data from this and related studies support a role for PKCalpha in glucose-induced insulin granule recruitment for exocytosis; a role for PKCepsilon in activation of insulin granules for exocytosis and/or in the glucose-generated time-dependent potentiation signal for insulin release; and a dual function for PKCzeta in initiating insulin release and in a regulatory role in the transcriptional machinery. Furthermore, diminished levels and/or activation of PKCalpha, PKCepsilon, PKCtheta, and PKCzeta could be part of the defective signals downstream to glucose metabolism responsible for the deranged insulin secretion in the GK rat.


Asunto(s)
Diabetes Mellitus Tipo 2/enzimología , Glucosa/farmacología , Células Secretoras de Insulina/enzimología , Isoenzimas/análisis , Proteína Quinasa C/análisis , Animales , Inmunohistoquímica , Insulina/metabolismo , Secreción de Insulina , Masculino , Transporte de Proteínas , Ratas , Ratas Wistar
19.
Med Sci (Paris) ; 23(10): 885-94, 2007 Oct.
Artículo en Francés | MEDLINE | ID: mdl-17937902

RESUMEN

The most common form of diabetes, type 2 diabetes (T2D) is a major Public Health issue which is receiving a great deal of attention both in industrial and public research, in order to develop new and more effective drugs. The hyperglycaemia of T2D is the result of two interdependent defects : decreased biological efficacy of insulin in target tissues (insulin resistance), and a decreased capacity for beta cells to secrete insulin in response to glucose. Furthermore, hyperglycaemia evolves with time and even with rigorous treatment there is a progressive deterioration of glucose homeostasis. Seventy five percent of DT2 patients are obese and show a perturbed lipid profile. beta-cell plasticity is a unique property of these cells to adapt their number and volume (beta-cell mass) and their function to the increased secretory demand linked to insulin resistance. This is well documented in physiological (pregnancy) as well in pathophysiological conditions (obesity, acromegaly). Although the lack of reliable techniques makes it very difficult to document it in humans, this property is likely altered in DT2, mainly as a consequence of the prolonged exposure of islet cells to high plasma levels of glucose and free fatty acids (gluco-lipotoxicity). The mechanisms by which hyperglycaemia and hyperlipidemia exert their deleterious effects on the beta-cell include the generation of Reactive Oxygen Species (ROS) and Reactive Nitrogen Species (RNS) and Advanced Glycosylation End Products (AGE). Altogether the prevailing clinical and experimental data urge us to consider that the pathophysiology of DT2 lies, at least in part, the inability of beta-cells to adapt their functional mass to the prevailing insulin demand. This re-evaluation of the pathophysiology of DT2 stimulates the research of new therapeutic approaches aimed at maintaining and/or restoring the functional beta-cell mass by targeting the mechanisms responsible for its decrease.


Asunto(s)
Diabetes Mellitus Tipo 2/fisiopatología , Islotes Pancreáticos/fisiopatología , Adaptación Fisiológica , Animales , División Celular , Tamaño de la Célula , Diabetes Mellitus Tipo 2/patología , Ácidos Grasos no Esterificados/metabolismo , Glucosa/metabolismo , Productos Finales de Glicación Avanzada/metabolismo , Humanos , Hiperglucemia/fisiopatología , Hiperlipidemias/fisiopatología , Insulina/metabolismo , Resistencia a la Insulina , Secreción de Insulina , Islotes Pancreáticos/metabolismo , Islotes Pancreáticos/patología , Síndrome Metabólico/fisiopatología , Modelos Biológicos , Obesidad/fisiopatología , Estrés Oxidativo , Especies de Nitrógeno Reactivo
20.
Bull Acad Natl Med ; 191(4-5): 941-3; discussion 943, 2007.
Artículo en Francés | MEDLINE | ID: mdl-18225448

RESUMEN

Type 2, non-insulin-dependent diabetes has been increasing exponentially over the past decade and a half and it is estimated that within short it will comprise more than 350 million patients. The pathophysiology of type 2 diabetes is complex, but has two dominating factors, insulin resistance (which is mainly due to obesity and physical inactivity), and deficient insulin production. Indeed, although approximately 80% of type 2 diabetics are obese, 2/3 of overweight or obese persons show normal glucose metabolism. Data accumulated over the past few decades unequivocally indicate that diabetes can not develop in the absence of a major deficiency of insulin secretion. This deficiency is characterised by an early loss of first-phase insulin response to glucose, followed by gradual collapse of the later insulin response as well as of the maximal secretory capacity of the beta-cell. Interestingly, functional modifications in the beta-cell do not present a discontinuity; in fact, some of the characteristics of the diabetic beta-cell function can be found in a fraction of the healthy population. A major challenge has been to answer the question whether the population with decreased insulin secretory capacity represents the substratum from which future diabetics emerge. While many observations suggest that such may indeed be the case, conclusive evidence is still unavailable. The search for the beta-cell molecular mechanisms which prepare the ground for diabetes has been difficult and mainly limited to laboratory models of type 2 diabetes. Greater success has been achieved in elucidating the secondary beta-cell defects elicited once diabetes is established and the beta-cell exposed to chronically elevated glucose and fatty acid levels (so-called gluco-lipotoxicity). The latter reduces the responsiveness of insulin secretion to physiological stimuli, as it impairs the biosynthesis and processing of proinsulin. The leptin resistance of obesity certainly plays a role in this context, since leptin reduces, i.a., the lipid content of the islet. Similarly, the reduced adiponectin levels of obesity favour diminished beta-cell function. Nevertheless, it seems probable that the most important negative factor for the beta-cell in obesity is the inflammatory state. Indeed, several cytokines are deleterious for the beta-cell and may play a role in the pathogenesis of the islet dysfunction of diabetes, as demonstrated by the recent work of Donath and coworkers. We propose the working hypothesis that the "prediabetic" beta-cell in fact is a normal beta-cell whose functional capabilities (insulin secretion and biosynthesis, cell proliferation, resistance to stress...) is at the lower-end of the normal distribution. At times of "reasonable" metabolic requirements, i.e. reasonable energy balance, such a "prediabetic" beta-cell is fully adequate to cover the insulin needs of the organism. Insulin production by the "prediabetic" beta-cell becomes insufficient either when insulin needs become excessive, as is the case in over-nutrition (with or without insulin resistance), or when beta-cell function and adaptation are impaired by "external" factors such as the obesity-related inflammatory cytokines. Thus, deficient beta-cell function is seen as a relative factor against the metabolic background dictated by environmental factors. Type 2 diabetes is a hereditary disease, and many genes have been shown to be linked to diabetes. Our hypothesis is that such genes (or rather their polymorphism) define the range of the functional adaptability of the beta-cell to metabolic demand. This would be an excellent example of gene-environment interaction. Thus, we do not believe that sensu stricto diabetes genes exist. Against the above, optimal diabetes treatment would necessitate--reduction of the metabolic demand on the beta-cell,--support of its function and adaptive capabilities. Several new research avenues, discussed in the present meeting, may open new and improved therapeutic approaches for type 2 diabetes.


Asunto(s)
Diabetes Mellitus Tipo 2/fisiopatología , Glucemia/análisis , Peso Corporal , Diabetes Mellitus Tipo 2/sangre , Diabetes Mellitus Tipo 2/etiología , Diabetes Mellitus Tipo 2/genética , Ácidos Grasos/sangre , Humanos , Insulina/metabolismo , Resistencia a la Insulina , Secreción de Insulina , Células Secretoras de Insulina/fisiología , Leptina/fisiología , Obesidad/fisiopatología , Estado Prediabético/fisiopatología , Proinsulina/biosíntesis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA