Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
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
3.
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
4.
J Am Soc Nephrol ; 29(2): 434-448, 2018 02.
Article in English | MEDLINE | ID: mdl-29030466

ABSTRACT

Altered glucose reabsorption via the facilitative glucose transporter 2 (GLUT2) during diabetes may lead to renal proximal tubule cell (RPTC) injury, inflammation, and interstitial fibrosis. These pathologies are also triggered by activating the cannabinoid-1 receptor (CB1R), which contributes to the development of diabetic nephropathy (DN). However, the link between CB1R and GLUT2 remains to be determined. Here, we show that chronic peripheral CB1R blockade or genetically inactivating CB1Rs in the RPTCs ameliorated diabetes-induced renal structural and functional changes, kidney inflammation, and tubulointerstitial fibrosis in mice. Inhibition of CB1R also downregulated GLUT2 expression, affected the dynamic translocation of GLUT2 to the brush border membrane of RPTCs, and reduced glucose reabsorption. Thus, targeting peripheral CB1R or inhibiting GLUT2 dynamics in RPTCs has the potential to treat and ameliorate DN. These findings may support the rationale for the clinical testing of peripherally restricted CB1R antagonists or the development of novel renal-specific GLUT2 inhibitors against DN.


Subject(s)
Diabetic Nephropathies/genetics , Diabetic Nephropathies/metabolism , Glucose Transporter Type 2/genetics , Glucose Transporter Type 2/metabolism , Kidney Tubules, Proximal/pathology , Receptor, Cannabinoid, CB1/metabolism , Albuminuria/urine , Animals , Biological Transport , Blood Glucose/metabolism , Blood Urea Nitrogen , Creatinine/urine , Diabetic Nephropathies/chemically induced , Dogs , Fibrosis , Glucose/metabolism , Glucose Transporter Type 2/antagonists & inhibitors , Insulin/blood , Islets of Langerhans/pathology , Madin Darby Canine Kidney Cells , Male , Mice , Mice, Knockout , Protein Kinase C beta/metabolism , Pyrazoles/pharmacology , Receptor, Cannabinoid, CB1/antagonists & inhibitors , Receptor, Cannabinoid, CB1/genetics , Streptozocin , Sulfonamides/pharmacology
5.
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
6.
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
7.
Neuroendocrinology ; 103(6): 724-37, 2016.
Article in English | MEDLINE | ID: mdl-26619207

ABSTRACT

BACKGROUND: Everolimus (RAD001), an mTORC1 inhibitor, demonstrated promising, but limited, anticancer effects in neuroendocrine tumors (NETs). Torin1 (a global mTOR inhibitor) and NVP-BEZ235 (a PI3K/mTOR inhibitor) seem to be more effective than RAD001. Autophagy, a degradation pathway that may promote tumor growth, is regulated by mTOR; mTOR inhibition results in stimulation of autophagy. Chloroquine (CQ) inhibits autophagy. AIM: To explore the effect of CQ alone or in combination with RAD001, Torin1 or NVP-BEZ235 on autophagy and on NET cell viability, proliferation and apoptosis. METHODS: The NET cell line BON1 was treated with CQ with or without different mTOR inhibitors. siRNA against ATG5/7 was used to genetically inhibit autophagy. Cellular viability was examined by XTT, proliferation by Ki-67 staining and cell cycles by flow cytometry. Apoptosis was analyzed by Western blotting for cleaved caspase 3 and staining for annexin V; autophagy was evaluated by Western blotting and immunostaining for LC3. RESULTS: RAD001, Torin1, NVP-BEZ235 and CQ all decreased BON1 cell viability. The effect of RAD001 was smaller than that of the other mTOR inhibitors or CQ. Torin1 and NVP-BEZ235 markedly inhibited cell proliferation, without inducing apoptosis. CQ similarly decreased cell proliferation, while robustly increasing apoptosis. Treatment with Torin1 or NVP-BEZ235 together with CQ was additive on viability, without increasing CQ-induced apoptosis. Inhibition of autophagy by ATG5/7 knockdown increased apoptosis in the presence or absence of mTOR inhibitors, mimicking the CQ effects. CONCLUSION: CQ inhibits NET growth by inducing apoptosis and by inhibiting cell proliferation, probably via inhibition of autophagy. CQ may potentiate the antitumor effect of mTOR inhibitors.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Autophagy/drug effects , Chloroquine/pharmacology , Protein Kinase Inhibitors/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival , Everolimus/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Humans , Imidazoles/pharmacology , Ki-67 Antigen/metabolism , Microtubule-Associated Proteins/metabolism , Neuroendocrine Tumors/pathology , Proto-Oncogene Proteins c-akt/metabolism , Quinolines/pharmacology , TOR Serine-Threonine Kinases/antagonists & inhibitors , Time Factors
8.
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
9.
Diabetologia ; 57(8): 1505-16, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24795087

ABSTRACT

Autophagy is a lysosomal degradation pathway recycling intracellular long-lived proteins and damaged organelles, thereby maintaining cellular homeostasis. In addition to inflammatory processes, autophagy has been implicated in the regulation of adipose tissue and beta cell functions. In obesity and type 2 diabetes autophagic activity is modulated in a tissue-dependent manner. In this review we discuss the regulation of autophagy in adipose tissue and beta cells, exemplifying tissue-specific dysregulation of autophagy and its implications for the pathophysiology of obesity and type 2 diabetes. We will highlight common themes and outstanding gaps in our understanding, which need to be addressed before autophagy could be envisioned as a therapeutic target for the treatment of obesity and diabetes.


Subject(s)
Adipose Tissue/metabolism , Autophagy/physiology , Diabetes Mellitus, Type 2/metabolism , Insulin-Secreting Cells/metabolism , Obesity/metabolism , Adipose Tissue/physiopathology , Diabetes Mellitus, Type 2/physiopathology , Humans , Insulin Resistance/physiology , Obesity/physiopathology
10.
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
11.
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
13.
Am J Physiol Endocrinol Metab ; 299(6): E879-86, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20858748

ABSTRACT

The peroxidation of n-3 and n-6 polyunsaturated fatty acids (PUFAs) and of their hydroperoxy metabolites is a complex process. It is initiated by free oxygen radical-induced abstraction of a hydrogen atom from the lipid molecule followed by a series of nonenzymatic reactions that ultimately generate the reactive aldehyde species 4-hydroxyalkenals. The molecule 4-hydroxy-2E-hexenal (4-HHE) is generated by peroxidation of n-3 PUFAs, such as linolenic acid, eicosapentaenoic acid, and docosahexaenoic acid. The aldehyde product 4-hydroxy-2E-nonenal (4-HNE) is the peroxidation product of n-6 PUFAs, such as arachidonic and linoleic acids and their 15-lipoxygenase metabolites, namely 15-hydroperoxyeicosatetraenoic acid (15-HpETE) and 13-hydroperoxyoctadecadienoic acid (13-HpODE). Another reactive peroxidation product is 4-hydroxy-2E,6Z-dodecadienal (4-HDDE), which is derived from 12-hydroperoxyeicosatetraenoic acid (12-HpETE), the 12-lipoxygenase metabolite of arachidonic acid. Hydroxyalkenals, notably 4-HNE, have been implicated in various pathophysiological interactions due to their chemical reactivity and the formation of covalent adducts with macromolecules. The progressive accumulation of these adducts alters normal cell functions that can lead to cell death. The lipophilicity of these aldehydes positively correlates to their chemical reactivity. Nonetheless, at low and noncytotoxic concentrations, these molecules may function as signaling molecules in cells. This has been shown mostly for 4-HNE and to some extent for 4-HHE. The capacity of 4-HDDE to generate such "mixed signals" in cells has received less attention. This review addresses the origin and cellular functions of 4-hydroxyalkernals.


Subject(s)
Fatty Acids, Unsaturated/metabolism , Lipid Peroxidation/physiology , Lipoxygenases/metabolism , Reactive Oxygen Species/metabolism
14.
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
15.
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
16.
Circ Res ; 97(10): 1001-8, 2005 Nov 11.
Article in English | MEDLINE | ID: mdl-16210549

ABSTRACT

Substrate autoregulation of glucose transporter-1 (GLUT-1) mRNA and protein expression provides vascular endothelial and smooth muscle cells a sensitive mechanism to adapt their rate of glucose transport in response to changing glycemic conditions. Hyperglycemia-induced downregulation of glucose transport is particularly important in protecting these cells against an excessive influx of glucose and consequently increased intracellular protein glycation and generation of free radicals; both are detrimental in the development of vascular disease in diabetes. We aimed to investigate the molecular mechanism of high glucose-induced downregulation of GLUT-1 mRNA expression in primary bovine aortic vascular endothelial (VEC) and smooth muscle (VSMC) cell cultures. Using RNA mobility shift, UV cross-linking, and in vitro degradation assays, followed by mass-spectrometric analysis, we identified calreticulin as a specific destabilizing trans-acting factor that binds to a 10-nucleotide cis-acting element (CAE(2181-2190)) in the 3'-untranslated region of GLUT-1 mRNA. Pure calreticulin accelerated the rate of GLUT-1 mRNA-probe degradation in vitro, whereas overexpression of calreticulin in vascular cells decreased significantly the total cell content of GLUT-1 mRNA and protein. The expression of calreticulin was augmented in vascular cells exposed to high glucose in comparison with low-glucose conditions. Similarly, increased expression of calreticulin was observed in aortae of diabetic Psammomys obesus in comparison with normoglycemic controls. These data suggest that CAE(2181-2190)-calreticulin complex, which is formed in VSMC and VEC exposed to hyperglycemic conditions, renders GLUT-1 mRNA susceptible to degradation. This interaction underlies the process of downregulation of glucose transport in vascular cells under high-glucose conditions.


Subject(s)
Calreticulin/physiology , Endothelial Cells/metabolism , Glucose Transporter Type 1/genetics , Hyperglycemia/metabolism , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , RNA Stability , 3' Untranslated Regions/metabolism , Animals , Cattle , Cells, Cultured , Down-Regulation , Muscle, Smooth, Vascular/cytology , Nitric Oxide/biosynthesis , RNA, Messenger/metabolism
17.
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
18.
Mol Biochem Parasitol ; 133(2): 153-62, 2004 Feb.
Article in English | MEDLINE | ID: mdl-14698428

ABSTRACT

Serine proteinase inhibitors (serpins) are irreversible suicide inhibitors of proteinases that regulate a wide range of biological processes, including pathogen evasion of the host defence system. We report the cloning and characterization of a gene encoding a serpin from the protozoan parasite Entamoeba histolytica (Ehserp) that may function in this manner. The protein encoded by Ehserp contains 371 amino acids with a predicted mass of 42.6 kDa. Antibodies to a 42 kDa recombinant Ehserp react specifically with two bands of 42 and 49 kDa in trophozoite extracts. Ehserp has a cytoplasmic localization and is secreted by trophozoites incubated in the presence of mammalian cells, but not by resting trophozoites. A panel of mammalian serine proteinases was screened, but none of them was inhibited by the recombinant Ehserp. In contrast, the 49 kDa Ehserp present in the secretion product (SP) of activated macrophages interacted with human neutrophil cathepsin G to form a complex resistant to sodium dodecyl sulphate. We discuss the nature of the 42 and 49 kDa Ehserp and the possible roles that Ehserp may play in the survival of the parasite inside the host.


Subject(s)
Entamoeba histolytica/genetics , Genes, Protozoan , Serine Proteinase Inhibitors/genetics , Serine Proteinase Inhibitors/metabolism , Serpins/genetics , Serpins/metabolism , Amino Acid Motifs , Amino Acid Sequence , Animals , Base Sequence , CHO Cells , Cathepsin G , Cathepsins/metabolism , Cloning, Molecular , Cricetinae , Entamoeba histolytica/pathogenicity , Gene Expression Regulation , Humans , Isoelectric Point , Jurkat Cells , Molecular Sequence Data , Molecular Weight , Neutrophils/enzymology , Protozoan Proteins/chemistry , Protozoan Proteins/genetics , Protozoan Proteins/metabolism , Protozoan Proteins/pharmacology , Sequence Alignment , Serine Endopeptidases/metabolism , Serine Proteinase Inhibitors/chemistry , Serine Proteinase Inhibitors/isolation & purification , Serpins/chemistry , Serpins/isolation & purification , Substrate Specificity
19.
Free Radic Biol Med ; 65: 978-987, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23973638

ABSTRACT

Peroxidation of polyunsaturated fatty acids is intensified in cells subjected to oxidative stress and results in the generation of various bioactive compounds, of which 4-hydroxyalkenals are prominent. During the progression of type 2 diabetes mellitus, the ensuing hyperglycemia promotes the generation of reactive oxygen species (ROS) that contribute to the development of diabetic complications. It has been suggested that ROS-induced lipid peroxidation and the resulting 4-hydroxyalkenals markedly contribute to the development and progression of these pathologies. Recent findings, however, also suggest that noncytotoxic levels of 4-hydroxyalkenals play important signaling functions in the early phase of diabetes and act as hormetic factors to induce adaptive and protective responses in cells, enabling them to function in the hyperglycemic milieu. Our studies and others' have proposed such regulatory functions for 4-hydroxynonenal and 4-hydroxydodecadienal in insulin secreting ß-cells and vascular endothelial cells, respectively. This review presents and discusses the mechanisms regulating the generation of 4-hydroxyalkenals under high glucose conditions and the molecular interactions underlying the reciprocal transition from hormetic to cytotoxic agents.


Subject(s)
Aldehydes/metabolism , Lipid Peroxidation , Signal Transduction , Animals , Diabetes Mellitus, Type 2/metabolism , Disease Progression , Humans , Oxidative Stress , Reactive Oxygen Species/metabolism
20.
Diabetes ; 60(11): 2830-42, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21896929

ABSTRACT

OBJECTIVE: Previous studies show that polyunsaturated fatty acids (PUFAs) increase the insulin secretory capacity of pancreatic ß-cells. We aimed at identifying PUFA-derived mediators and their cellular targets that are involved in the amplification of insulin release from ß-cells preexposed to high glucose levels. RESEARCH DESIGN AND METHODS: The content of fatty acids in phospholipids of INS-1E ß-cells was determined by lipidomics analysis. High-performance liquid chromatography was used to identify peroxidation products in ß-cell cultures. Static and dynamic glucose-stimulated insulin secretion (GSIS) assays were performed on isolated rat islets and/or INS-1E cells. The function of peroxisome proliferator-activated receptor-δ (PPAR-δ) in regulating insulin secretion was investigated using pharmacological agents and gene expression manipulations. RESULTS: High glucose activated cPLA(2) and, subsequently, the hydrolysis of arachidonic and linoleic acid (AA and LA, respectively) from phospholipids in INS-1E cells. Glucose also increased the level of reactive oxygen species, which promoted the peroxidation of these PUFAs to generate 4-hydroxy-2E-nonenal (4-HNE). The latter mimicked the GSIS-amplifying effect of high glucose preexposure and of the PPAR-δ agonist GW501516 in INS-1E cells and isolated rat islets. These effects were blocked with GSK0660, a selective PPAR-δ antagonist, and the antioxidant N-acetylcysteine or by silencing PPAR-δ expression. High glucose, 4-HNE, and GW501516 also induced luciferase expression in a PPAR-δ-mediated transactivation assay. Cytotoxic effects of 4-HNE were observed only above the physiologically effective concentration range. CONCLUSIONS: Elevated glucose levels augment the release of AA and LA from phospholipids and their peroxidation to 4-HNE in ß-cells. This molecule is an endogenous ligand for PPAR-δ, which amplifies insulin secretion in ß-cells.


Subject(s)
Hyperglycemia/metabolism , Insulin-Secreting Cells/metabolism , Insulin/metabolism , Lipid Peroxidation , PPAR delta/metabolism , Signal Transduction , Aldehydes/adverse effects , Aldehydes/blood , Aldehydes/metabolism , Aldehydes/pharmacology , Animals , Cell Line , Diabetes Mellitus, Type 2/blood , Fatty Acids, Unsaturated/metabolism , Gene Silencing , Gerbillinae , Group IV Phospholipases A2/metabolism , Humans , Insulin Secretion , Insulin-Secreting Cells/drug effects , Islets of Langerhans/drug effects , Islets of Langerhans/metabolism , Lipid Peroxidation/drug effects , Male , PPAR delta/agonists , PPAR delta/antagonists & inhibitors , PPAR delta/genetics , Rats , Rats, Wistar , Reactive Oxygen Species/metabolism , Recombinant Proteins/agonists , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/metabolism , Tissue Culture Techniques
SELECTION OF CITATIONS
SEARCH DETAIL