Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 52
Filter
1.
Diabetologia ; 66(10): 1925-1942, 2023 10.
Article in English | MEDLINE | ID: mdl-37480416

ABSTRACT

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 ).


Subject(s)
Diabetes Mellitus, Experimental , Diabetes Mellitus, Type 2 , Hyperglycemia , Adult , Humans , Animals , Glucagon , Mechanistic Target of Rapamycin Complex 1 , Glucose , Mammals
2.
Bioorg Med Chem ; 87: 117303, 2023 05 03.
Article in English | MEDLINE | ID: mdl-37167713

ABSTRACT

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.


Subject(s)
Diabetes Mellitus, Type 2 , Hypoglycemic Agents , Rats , Mice , Animals , Hypoglycemic Agents/pharmacology , Hypoglycemic Agents/therapeutic use , AMP-Activated Protein Kinases , Diabetes Mellitus, Type 2/drug therapy , Glucose/pharmacology , Cell Line , Muscle Fibers, Skeletal , Insulin/pharmacology
3.
JCI Insight ; 8(7)2023 04 10.
Article in English | MEDLINE | ID: mdl-36809274

ABSTRACT

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.


Subject(s)
Diabetes Mellitus, Experimental , Sodium-Glucose Transporter 2 Inhibitors , Animals , Mice , Sodium-Glucose Transporter 2 Inhibitors/pharmacology , Diabetes Mellitus, Experimental/drug therapy , Sodium-Glucose Transporter 2/metabolism , AMP-Activated Protein Kinases/metabolism , Betaine , Glucose , Sodium/metabolism , Methionine
4.
Diabetes ; 71(3): 453-469, 2022 03 01.
Article in English | MEDLINE | ID: mdl-34862201

ABSTRACT

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.


Subject(s)
Autophagy/physiology , Insulin-Secreting Cells/physiology , Mechanistic Target of Rapamycin Complex 1/physiology , Animals , Autophagy/drug effects , Cell Line , Fasting , Glucose/pharmacology , Humans , Insulin Secretion/drug effects , Insulin Secretion/physiology , Leucine/pharmacology , Male , Mechanistic Target of Rapamycin Complex 1/drug effects , Mechanistic Target of Rapamycin Complex 1/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Postprandial Period/physiology
5.
Cell Rep ; 32(4): 107954, 2020 07 28.
Article in English | MEDLINE | ID: mdl-32726619

ABSTRACT

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.


Subject(s)
Diabetic Nephropathies/physiopathology , Mechanistic Target of Rapamycin Complex 1/metabolism , Sodium-Glucose Transporter 2 Inhibitors/pharmacology , Animals , Diabetes Mellitus, Type 2/metabolism , Diabetic Nephropathies/etiology , Humans , Hypoglycemic Agents/pharmacology , Kidney/metabolism , Kidney Failure, Chronic/metabolism , Kidney Tubules, Proximal/drug effects , Kidney Tubules, Proximal/metabolism , Kidney Tubules, Proximal/physiopathology , Male , Mechanistic Target of Rapamycin Complex 1/physiology , Mice , Sodium-Glucose Transporter 2 Inhibitors/metabolism , Swine
6.
Elife ; 72018 11 09.
Article in English | MEDLINE | ID: mdl-30412050

ABSTRACT

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.


Subject(s)
Diabetes Mellitus/genetics , Endoplasmic Reticulum Stress/genetics , Mechanistic Target of Rapamycin Complex 1/genetics , Proinsulin/genetics , Animals , Animals, Newborn/genetics , Animals, Newborn/growth & development , Apoptosis/genetics , Diabetes Mellitus/pathology , Endoplasmic Reticulum/genetics , Humans , Insulin-Secreting Cells/pathology , Mice , Mutation , Protein Folding
7.
Diabetes Obes Metab ; 20 Suppl 2: 95-103, 2018 09.
Article in English | MEDLINE | ID: mdl-30230182

ABSTRACT

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.


Subject(s)
Cell Differentiation , Diabetes Mellitus/etiology , Insulin-Secreting Cells/physiology , Proinsulin/physiology , Protein Folding , Adaptation, Physiological/physiology , Animals , Cell Differentiation/physiology , Diabetes Mellitus/physiopathology , Diabetes Mellitus, Type 1/etiology , Diabetes Mellitus, Type 1/physiopathology , Diabetes Mellitus, Type 2/etiology , Diabetes Mellitus, Type 2/physiopathology , Disease Models, Animal , Endoplasmic Reticulum Stress/physiology , Humans , Insulin-Secreting Cells/metabolism , Mice , Proinsulin/chemistry , Swine
8.
J Cell Sci ; 131(15)2018 08 06.
Article in English | MEDLINE | ID: mdl-30002135

ABSTRACT

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.


Subject(s)
Adenine Nucleotides/metabolism , Autophagy/physiology , Insulin-Secreting Cells/metabolism , AMP-Activated Protein Kinases/metabolism , Adenosine Triphosphate , Animals , Blotting, Western , Cell Line , Fluorescent Antibody Technique , Hep G2 Cells , Humans , Insulin/metabolism , Insulin-Secreting Cells/cytology , Male , Mechanistic Target of Rapamycin Complex 1/metabolism , Mice , Mice, Inbred C57BL , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction
9.
Pharm Res ; 34(12): 2873-2890, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28983714

ABSTRACT

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.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Chromans/pharmacology , Glucose/metabolism , Hypoglycemic Agents/pharmacology , Insulin-Secreting Cells/drug effects , Insulin/metabolism , Muscle Fibers, Skeletal/drug effects , Animals , Biological Transport/drug effects , Cell Line , Cells, Cultured , Chromans/chemistry , Drug Discovery , Enzyme Activation/drug effects , Humans , Hypoglycemic Agents/chemistry , Insulin-Secreting Cells/metabolism , Muscle Fibers, Skeletal/metabolism , Rats
10.
Diabetes Obes Metab ; 19 Suppl 1: 3, 2017 09.
Article in English | MEDLINE | ID: mdl-28880483
11.
Data Brief ; 14: 329-336, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28795110

ABSTRACT

The data presented in this article are related to the research article entitled "Regulation of GLUT4 activity in myotubes by 3-O-methyl-D-glucose" (Shamni et al., 2017) [1]. These data show that the experimental procedures used to analyze the effects of 3-O-methyl-D-glucose (MeGlc) on the rate of hexose transport into myotubes were valid and controlled. The stimulatory effect of MeGlc was limited to glucose transporter 4 (GLUT4) and was independent of ambient glucose and protein synthesis. Cornish-Bowden kinetic analysis of uptake data revealed that MeGlc attenuated indinavir-induced inhibition of hexose transport in a competitive manner.

12.
Biochim Biophys Acta Biomembr ; 1859(10): 1900-1910, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28648676

ABSTRACT

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.


Subject(s)
3-O-Methylglucose/pharmacology , Glucose Transporter Type 4/metabolism , Muscle Fibers, Skeletal/metabolism , Adenosine Triphosphate/metabolism , Animals , Biological Transport/drug effects , Biological Transport/physiology , Cell Line , Cell Membrane/drug effects , Cell Membrane/metabolism , Glucose/metabolism , Insulin/metabolism , Kinetics , Mitogen-Activated Protein Kinases/metabolism , Muscle Contraction/drug effects , Muscle Contraction/physiology , Muscle Fibers, Skeletal/drug effects , Muscle Proteins/metabolism , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Rats , Signal Transduction/drug effects , Signal Transduction/physiology
13.
J Clin Endocrinol Metab ; 101(10): 3592-3599, 2016 10.
Article in English | MEDLINE | ID: mdl-27459537

ABSTRACT

CONTEXT: Type 2 Wolfram syndrome (T2-WFS) is a neuronal and ß-cell degenerative disorder caused by mutations in the CISD2 gene. The mechanisms underlying ß-cell dysfunction in T2-WFS are not known, and treatments that effectively improve diabetes in this context are lacking. OBJECTIVE: Unraveling the mechanisms of ß-cell dysfunction in T2-WFS and the effects of treatment with GLP-1 receptor agonist (GLP-1-RA). DESIGN AND SETTING: A case report and in vitro mechanistic studies. PATIENT AND METHODS: We treated an insulin-dependent T2-WFS patient with the GLP-1-RA exenatide for 9 weeks. An iv glucose/glucagon/arginine stimulation test was performed off-drug before and after intervention. We generated a cellular model of T2-WFS by shRNA knockdown of CISD2 (nutrient-deprivation autophagy factor-1 [NAF-1]) in rat insulinoma cells and studied the mechanisms of ß-cell dysfunction and the effects of GLP-1-RA. RESULTS: Treatment with exenatide resulted in a 70% reduction in daily insulin dose with improved glycemic control, as well as an off-drug 7-fold increase in maximal insulin secretion. NAF-1 repression in INS-1 cells decreased insulin content and glucose-stimulated insulin secretion, while maintaining the response to cAMP, and enhanced the accumulation of labile iron and reactive oxygen species in mitochondria. Remarkably, treatment with GLP-1-RA and/or the iron chelator deferiprone reversed these defects. CONCLUSION: NAF-1 deficiency leads to mitochondrial labile iron accumulation and oxidative stress, which may contribute to ß-cell dysfunction in T2-WFS. Treatment with GLP-1-RA and/or iron chelation improves mitochondrial function and restores ß-cell function. Treatment with GLP-1-RA, probably aided by iron chelation, should be considered in WFS and other forms of diabetes associated with iron dysregulation.


Subject(s)
Aging, Premature/drug therapy , Glucagon-Like Peptide-1 Receptor/agonists , Hearing Loss, Sensorineural/drug therapy , Hypoglycemic Agents/pharmacology , Insulin-Secreting Cells/drug effects , Mitochondria/drug effects , Mitochondrial Diseases/drug therapy , Optic Atrophy/drug therapy , Peptides/pharmacology , Venoms/pharmacology , Animals , Exenatide , Female , Humans , Hypoglycemic Agents/administration & dosage , Peptides/administration & dosage , Rats , Venoms/administration & dosage
15.
Diabetologia ; 59(7): 1480-1491, 2016 07.
Article in English | MEDLINE | ID: mdl-26831301

ABSTRACT

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.


Subject(s)
Autophagy/physiology , Insulin/metabolism , Animals , Autophagy/genetics , Autophagy-Related Protein 7/genetics , Autophagy-Related Protein 7/metabolism , Blotting, Western , Cell Line , Homeostasis/genetics , Homeostasis/physiology , Humans , Insulin-Secreting Cells/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Electron , Oxygen Consumption/genetics , Oxygen Consumption/physiology , RNA Interference/physiology
16.
J Cell Mol Med ; 19(8): 1887-99, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25754218

ABSTRACT

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.


Subject(s)
Aldehydes/pharmacology , Carrier Proteins/metabolism , Cellular Senescence/drug effects , Endothelial Cells/metabolism , Foam Cells/metabolism , Animals , Biomarkers/metabolism , Cattle , Cell Line , Coculture Techniques , Endothelial Cells/cytology , Endothelial Cells/drug effects , Fluorescent Antibody Technique , Foam Cells/cytology , Foam Cells/drug effects , Free Radical Scavengers/pharmacology , Humans , Lipid Peroxidation/drug effects , Models, Biological , PPAR delta/metabolism , Plaque, Atherosclerotic/metabolism , Plaque, Atherosclerotic/pathology
17.
Mol Endocrinol ; 27(10): 1706-23, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23979843

ABSTRACT

Experimental lipotoxicity constitutes a model for ß-cell demise induced by metabolic stress in obesity and type 2 diabetes. Fatty acid excess induces endoplasmic reticulum (ER) stress, which is accompanied by ER morphological changes whose mechanisms and relevance are unknown. We found that the GTPase dynamin-related protein 1 (DRP1), a key regulator of mitochondrial fission, is an ER resident regulating ER morphology in stressed ß-cells. Inhibition of DRP1 activity using a GTP hydrolysis-defective mutant (Ad-K38A) attenuated fatty acid-induced ER expansion and mitochondrial fission. Strikingly, stimulating the key energy-sensor AMP-activated protein kinase (AMPK) increased the phosphorylation at the anti-fission site Serine 637 and largely prevented the alterations in ER and mitochondrial morphology. Expression of a DRP1 mutant resistant to phosphorylation at this position partially prevented the recovery of ER and mitochondrial morphology by AMPK. Fatty acid-induced ER enlargement was associated with proinsulin retention in the ER, together with increased proinsulin/insulin ratio. Stimulation of AMPK prevented these alterations, as well as mitochondrial fragmentation and apoptosis. In summary, DRP1 regulation by AMPK delineates a novel pathway controlling ER and mitochondrial morphology, thereby modulating the response of ß-cells to metabolic stress. DRP1 may thus function as a node integrating signals from stress regulators, such as AMPK, to coordinate organelle shape and function.


Subject(s)
Dynamins/metabolism , Endoplasmic Reticulum Stress , Endoplasmic Reticulum/enzymology , Insulin-Secreting Cells/enzymology , Adenylate Kinase/metabolism , Animals , Apoptosis , Cell Line , Diabetes Mellitus, Type 2/enzymology , Diabetes Mellitus, Type 2/pathology , Endoplasmic Reticulum/pathology , Enzyme Activation , Male , Mice , Mice, Inbred C57BL , Mitochondria/enzymology , Obesity/enzymology , Obesity/pathology , Organelle Shape , Palmitates/pharmacology , Phosphorylation , Unfolded Protein Response
18.
J Med Chem ; 56(13): 5335-50, 2013 Jul 11.
Article in English | MEDLINE | ID: mdl-23750537

ABSTRACT

Adenosine 5'-monophosphate activated protein kinase (AMPK) has emerged as a major potential target for novel antidiabetic drugs. We studied the structure of 2-chloro-5-((Z)-((E)-5-((5-(4,5-dimethyl-2-nitrophenyl)furan-2-yl)methylene)-4-oxothiazolidin-2-ylidene)amino)benzoic acid (PT-1), which attenuates the autoinhibition of the enzyme AMPK, for the design and synthesis of different benzothiazoles with potential antidiabetic activity. We synthesized several structurally related benzothiazole derivatives that increased the rate of glucose uptake in L6 myotubes in an AMPK-dependent manner. One compound, 2-(benzo[d]thiazol-2-ylmethylthio)-6-ethoxybenzo[d]thiazole (34), augmented the rate of glucose uptake up to 2.5-fold compared with vehicle-treated cells and up to 1.1-fold compared to PT-1. Concomitantly, it elevated the abundance of GLUT4 in the plasma membrane of the myotubes and activated AMPK. Subcutaneous administration of 34 to hyperglycemic Kuo Kondo rats carrying the Ay-yellow obese gene (KKAy) mice lowered blood glucose levels toward the normoglycemic range. In accord with its activity, compound 34 showed a high fit value to a pharmacophore model derived from the PT-1.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Benzothiazoles/pharmacology , Hypoglycemic Agents/pharmacology , Muscle Fibers, Skeletal/drug effects , Adenosine Triphosphate/metabolism , Animals , Benzothiazoles/chemical synthesis , Blood Glucose/metabolism , Cell Line , Cell Membrane/drug effects , Cell Membrane/metabolism , Enzyme Activation/drug effects , Glucose/metabolism , Glucose/pharmacokinetics , Glucose Transporter Type 4/metabolism , Hyperglycemia/blood , Hyperglycemia/drug therapy , Hypoglycemic Agents/chemical synthesis , Male , Mice , Models, Chemical , Molecular Structure , Muscle Fibers, Skeletal/metabolism , Rats
19.
Autophagy ; 9(4): 626-8, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23380813

ABSTRACT

Pancreatic ß-cell dysfunction is central in diabetes. The diabetic milieu may impair proinsulin folding, leading to ß-cell endoplasmic reticulum (ER) stress and apoptosis, and thus a worsening of the diabetes. Autophagy is crucial for the well-being of the ß-cell; however, the impact of stimulating autophagy on ß-cell adaptation to ER stress is unknown. We studied the crosstalk between ER stress and autophagy in a rodent model of diabetes, called Akita, in which proinsulin gene mutation leads to protein misfolding and ß-cell demise. We found that proinsulin misfolding stimulates autophagy and, in symmetry, inhibition of autophagy induces ß-cell stress and apoptosis. Under conditions of excessive proinsulin misfolding, stimulation of autophagy by inhibiting MTORC1 alleviates stress and prevents apoptosis. Moreover, treatment of diabetic Akita mice with the MTORC1 inhibitor rapamycin improves diabetes and prevents ß-cell apoptosis. Thus, autophagy is a central adaptive mechanism in ß-cell stress. Stimulation of autophagy may become a novel therapeutic strategy in diabetes.


Subject(s)
Autophagy , Diabetes Mellitus/pathology , Endoplasmic Reticulum Stress , Animals , Diabetes Mellitus/metabolism , Endoplasmic Reticulum/metabolism , Humans , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/pathology , Mice , Models, Biological , Mutation/genetics , Signal Transduction
20.
Diabetes ; 62(4): 1227-37, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23274896

ABSTRACT

Accumulation of misfolded proinsulin in the ß-cell leads to dysfunction induced by endoplasmic reticulum (ER) stress, with diabetes as a consequence. Autophagy helps cellular adaptation to stress via clearance of misfolded proteins and damaged organelles. We studied the effects of proinsulin misfolding on autophagy and the impact of stimulating autophagy on diabetes progression in Akita mice, which carry a mutation in proinsulin, leading to its severe misfolding. Treatment of female diabetic Akita mice with rapamycin improved diabetes, increased pancreatic insulin content, and prevented ß-cell apoptosis. In vitro, autophagic flux was increased in Akita ß-cells. Treatment with rapamycin further stimulated autophagy, evidenced by increased autophagosome formation and enhancement of autophagosome-lysosome fusion. This was associated with attenuation of cellular stress and apoptosis. The mammalian target of rapamycin (mTOR) kinase inhibitor Torin1 mimicked the rapamycin effects on autophagy and stress, indicating that the beneficial effects of rapamycin are indeed mediated via inhibition of mTOR. Finally, inhibition of autophagy exacerbated stress and abolished the anti-ER stress effects of rapamycin. In conclusion, rapamycin reduces ER stress induced by accumulation of misfolded proinsulin, thereby improving diabetes and preventing ß-cell apoptosis. The beneficial effects of rapamycin in this context strictly depend on autophagy; therefore, stimulating autophagy may become a therapeutic approach for diabetes.


Subject(s)
Autophagy/drug effects , Diabetes Mellitus/drug therapy , Endoplasmic Reticulum/physiology , Immunosuppressive Agents/therapeutic use , Sirolimus/therapeutic use , Animals , Autophagy/physiology , Diabetes Mellitus/etiology , Female , Immunosuppressive Agents/pharmacology , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/pathology , Insulin-Secreting Cells/physiology , Male , Mechanistic Target of Rapamycin Complex 1 , Mice , Mice, Inbred C57BL , Mice, Inbred Strains , Multiprotein Complexes , Mutation , Naphthyridines/pharmacology , Proinsulin/chemistry , Proinsulin/genetics , Proinsulin/metabolism , Protein Folding , Proteins/antagonists & inhibitors , Proteins/metabolism , Sirolimus/pharmacology , Stress, Physiological , TOR Serine-Threonine Kinases/antagonists & inhibitors
SELECTION OF CITATIONS
SEARCH DETAIL
...