Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
Mol Cell Endocrinol ; 556: 111740, 2022 10 01.
Article in English | MEDLINE | ID: mdl-35932980

ABSTRACT

Dysregulation of adipose tissue involves increased cellular hypoxia, ER stress, and inflammation and altered adipokine production, contributing to the aetiology of obesity-related diseases including type 2 diabetes and cardiovascular disease. This study aimed to investigate the effects of Vitamin C supplementation on these processes in primary human preadipocytes and adipocytes. Treatment of preadipocytes and adipocytes with the proinflammatory cytokine TNFα and palmitic acid (PA), to mimic the obesogenic milieu, significantly increased markers of hypoxia, ER stress and inflammation and reduced secretion of high molecular weight (HMW) adiponectin. Importantly, Vitamin C abolished TNFα+PA induced hypoxia and significantly reduced the increases in ER stress and inflammation in both cell types. Vitamin C also significantly increased the secretion of HMW adiponectin from adipocytes. These findings indicate that Vitamin C can reduce obesity-associated cellular stress and thus provide a rationale for future investigations.


Subject(s)
Diabetes Mellitus, Type 2 , Tumor Necrosis Factor-alpha , Adipocytes/metabolism , Adiponectin/metabolism , Ascorbic Acid/metabolism , Ascorbic Acid/pharmacology , Diabetes Mellitus, Type 2/metabolism , Humans , Hypoxia/metabolism , Inflammation/metabolism , Obesity/metabolism , Tumor Necrosis Factor-alpha/pharmacology
2.
FASEB Bioadv ; 3(1): 36-48, 2021 Jan.
Article in English | MEDLINE | ID: mdl-33490882

ABSTRACT

Chronic metabolic acidosis plays a role in cachexia by enhancing total proteolysis in skeletal muscle. Glucocorticoid also triggers proteolysis and plays a permissive role in the effect of acidosis. The System A amino acid transporter SNAT2/SLC38A2 is ubiquitously expressed in mammalian cells including muscle, performing Na+-dependent active import of neutral amino acids, and is strongly inhibited by low pH. Exposure of rat skeletal muscle cell line L6-G8C5 to low pH rapidly inhibits SNAT2 transport activity and enhances total proteolysis rate. Pharmacological inhibition or silencing of SNAT2 also enhances proteolysis. This study tests the hypothesis that the glucocorticoid dexamethasone (DEX), like low pH, inhibits SNAT2 activity in L6-G8C5 myotubes, thus contributing to total proteolysis. Incubation with 500 nM DEX for 4 h reduced the System A amino acid transport rate to half the rate in control cultures. This inhibition depended on glucocorticoid receptor-mediated gene transcription, but SNAT2 mRNA levels were unaffected by DEX. In contrast, the SNAT2 protein assessed by immunoblotting was significantly depleted. The co-inhibitory effects of DEX and low pH on System A transport activity were additive in stimulating total proteolysis. In keeping with this mechanism, DEX's inhibitory effect on SNAT2 transport activity was significantly blunted by the proteasome inhibitor MG132. Proof of principle was achieved in similar experiments using recombinant expression of a GFP-tagged SNAT2 fusion protein in HEK293A cells. It is concluded that DEX acutely depletes the SNAT2 transporter protein, at least partly through proteasome-dependent degradation of this functionally important transporter.

3.
Biochem J ; 475(5): 923-929, 2018 03 09.
Article in English | MEDLINE | ID: mdl-29463644

ABSTRACT

Inositol-requiring enzyme 1 alpha (IRE1α) is an endoplasmic reticulum (ER)-transmembrane endonuclease that is activated in response to ER stress as part of the unfolded protein response (UPR). Chronic activation of the UPR has been implicated in the pathogenesis of many common diseases including diabetes, cancer, and neurological pathologies such as Huntington's and Alzheimer's disease. 7-Hydroxy-4-methyl-2-oxo-2H-chromene-8-carbaldehyde (4µ8C) is widely used as a specific inhibitor of IRE1α ribonuclease activity (IC50 of 6.89 µM in cultured cells). However, in this paper, we demonstrate that 4µ8C acts as a potent reactive oxygen species (ROS) scavenger, both in a cell-free assay and in cultured cells, at concentrations lower than that widely used to inhibit IRE1α activity. In vitro we show that, 4µ8C effectively decreases xanthine/xanthine oxidase catalysed superoxide production with an IC50 of 0.2 µM whereas in cultured endothelial and clonal pancreatic ß-cells, 4µ8C inhibits angiotensin II-induced ROS production with IC50 values of 1.92 and 0.29 µM, respectively. In light of this discovery, conclusions reached using 4µ8C as an inhibitor of IRE1α should be carefully evaluated. However, this unexpected off-target effect of 4µ8C may prove therapeutically advantageous for the treatment of pathologies that are thought to be caused by, or exacerbated by, both oxidative and ER stress such as endothelial dysfunction and/or diabetes.


Subject(s)
Antioxidants/pharmacology , Endoribonucleases/antagonists & inhibitors , Hymecromone/analogs & derivatives , Protein Serine-Threonine Kinases/antagonists & inhibitors , Animals , Cells, Cultured , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum Stress/drug effects , Hymecromone/pharmacology , Mice , Reactive Oxygen Species/metabolism , Ribonucleases/antagonists & inhibitors , Unfolded Protein Response/drug effects
4.
Sci Rep ; 8(1): 1818, 2018 01 29.
Article in English | MEDLINE | ID: mdl-29379034

ABSTRACT

Endoplasmic reticulum (ER) stress has been implicated in the development of hypertension 3 through the induction of endothelial impairment. As 3',4'-dihydroxyflavonol (DiOHF) 4 reduces vascular injury caused by ischaemia/reperfusion or diabetes, and flavonols have been demonstrated to attenuate ER stress, we investigated whether DiOHF can protect mice from ER stress-induced endothelial dysfunction. Male C57BLK/6 J mice were injected with tunicamycin to induce ER stress in the presence or absence of either DiOHF or tauroursodeoxycholic acid (TUDCA), an inhibitor of ER stress. Tunicamycin elevated blood pressure and impaired endothelium-dependent relaxation. Moreover, in aortae there was evidence of ER stress, oxidative stress and reduced NO production. This was coincident with increased NOX2 expression and reduced phosphorylation of endothelial nitric oxide synthase (eNOS) on Ser1176. Importantly, the effects of tunicamycin were significantly ameliorated by DiOHF or TUDCA. DiOHF also inhibited tunicamycin-induced ER stress and apoptosis in cultured human endothelial cells (HUVEC). These results provide evidence that ER stress is likely an important initiator of endothelial dysfunction through the induction of oxidative stress and a reduction in NO synthesis and that DiOHF directly protects against ER stress- induced injury. DiOHF may be useful to prevent ER and oxidative stress to preserve endothelial function, for example in hypertension.


Subject(s)
Apoptosis/drug effects , Endoplasmic Reticulum Stress/drug effects , Endothelium, Vascular/drug effects , Flavonols/pharmacology , Vascular Diseases/drug therapy , Animals , Aorta/drug effects , Aorta/metabolism , Cells, Cultured , Endothelium, Vascular/metabolism , Human Umbilical Vein Endothelial Cells , Humans , Male , Mice , Mice, Inbred C57BL , Nitric Oxide/metabolism , Nitric Oxide Synthase Type III/metabolism , Oxidative Stress/drug effects , Tunicamycin/pharmacology , Vascular Diseases/metabolism
5.
Endocrinology ; 158(10): 3162-3173, 2017 10 01.
Article in English | MEDLINE | ID: mdl-28938442

ABSTRACT

The metabolic syndrome is associated with an increase in the activation of the renin angiotensin system, whose inhibition reduces the incidence of new-onset diabetes. Importantly, angiotensin II (AngII), independently of its vasoconstrictor action, causes ß-cell inflammation and dysfunction, which may be an early step in the development of type 2 diabetes. The aim of this study was to determine how AngII causes ß-cell dysfunction. Islets of Langerhans were isolated from C57BL/6J mice that had been infused with AngII in the presence or absence of taurine-conjugated ursodeoxycholic acid (TUDCA) and effects on endoplasmic reticulum (ER) stress, inflammation, and ß-cell function determined. The mechanism of action of AngII was further investigated using isolated murine islets and clonal ß cells. We show that AngII triggers ER stress, an increase in the messenger RNA expression of proinflammatory cytokines, and promotes ß-cell dysfunction in murine islets of Langerhans both in vivo and ex vivo. These effects were significantly attenuated by TUDCA, an inhibitor of ER stress. We also show that AngII-induced ER stress is required for the increased expression of proinflammatory cytokines and is caused by reactive oxygen species and IP3 receptor activation. These data reveal that the induction of ER stress is critical for AngII-induced ß-cell dysfunction and indicates how therapies that promote ER homeostasis may be beneficial in the prevention of type 2 diabetes.


Subject(s)
Angiotensin II/pharmacology , Endoplasmic Reticulum Stress/physiology , Inflammation/physiopathology , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/physiology , Animals , Cell Line, Tumor , Cytokines/genetics , Diabetes Mellitus, Type 2/etiology , Diabetes Mellitus, Type 2/prevention & control , Endoplasmic Reticulum Stress/drug effects , Endoribonucleases/antagonists & inhibitors , Endoribonucleases/genetics , Endoribonucleases/physiology , Gene Expression/drug effects , Gene Knockdown Techniques , Glucose/pharmacology , Inositol 1,4,5-Trisphosphate Receptors/physiology , Insulinoma , Islets of Langerhans/drug effects , Islets of Langerhans/physiopathology , Male , Mice , Mice, Inbred C57BL , Pancreatic Neoplasms , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/physiology , RNA, Small Interfering , Reactive Oxygen Species/metabolism , Renin-Angiotensin System/physiology , Taurine/pharmacology , Ursodeoxycholic Acid/pharmacology , eIF-2 Kinase/antagonists & inhibitors , eIF-2 Kinase/physiology
6.
Biochimie ; 138: 156-167, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28478109

ABSTRACT

Endoplasmic reticulum (ER) stress and the activation of the unfolded protein response (UPR) have been implicated in the pathogenesis of many common human diseases. Integral to the UPR and an important determinant in cell fate is the expression of the pro-apoptotic transcription factor C/EBP homologous protein (CHOP). This is promoted by activating transcription factor 4 (ATF4) whose expression is rapidly up-regulated in response to ER stress through an eIF2α phosphorylation-dependent increase in protein synthesis. Our data demonstrates that this ER stress-induced increase in ATF4 and CHOP expression is initiated by an increase in Atf4 and Chop mRNA, which is also dependent upon eIF2α phosphorylation. Despite being dependent on eIF2α phosphorylation, we provide evidence that these increases in Atf4 and Chop mRNA expression may occur independently of de novo protein synthesis. Moreover, we show that ER stress-induced Chop mRNA expression is exacerbated by Sirtuin-1 (SIRT1) inhibition indicating that changes in the energy status of the cell may play an important role in its regulation. This work highlights and extends previous findings, and provides important new insights into the mechanism of ER stress-induced expression of Atf4 and Chop mRNA that clearly warrants further investigation.


Subject(s)
Activating Transcription Factor 4/genetics , Endoplasmic Reticulum Stress , Eukaryotic Initiation Factor-2/metabolism , Signal Transduction , Sirtuin 1 , Transcription Factor CHOP/genetics , Animals , Mice , Phosphorylation , Protein Biosynthesis , Unfolded Protein Response , Up-Regulation
7.
PLoS One ; 12(4): e0173676, 2017.
Article in English | MEDLINE | ID: mdl-28422956

ABSTRACT

Autophagy is a catabolic mechanism to degrade cellular components to maintain cellular energy levels during starvation, a condition where PPARα may be activated. Here we report a reduced autophagic capacity in the liver following chronic activation of PPARα with fenofibrate (FB) in mice. Chronic administration of the PPARα agonist FB substantially reduced the levels of multiple autophagy proteins in the liver (Atg3, Agt4B, Atg5, Atg7 and beclin 1) which were associated with a decrease in the light chain LC3II/LC3I ratio and the accumulation of p62. This was concomitant with an increase in the expression of lipogenic proteins mSREBP1c, ACC, FAS and SCD1. These effects of FB were completely abolished in PPARα-/- mice but remained intact in mice with global deletion of FGF21, a key downstream mediator for PPARα-induced effects. Further studies showed that decreased the content of autophagy proteins by FB was associated with a significant reduction in the level of FoxO1, a transcriptional regulator of autophagic proteins, which occurred independently of both mTOR and Akt. These findings suggest that chronic stimulation of PPARα may suppress the autophagy capacity in the liver as a result of reduced content of a number of autophagy-associated proteins independent of FGF21.


Subject(s)
Autophagy/drug effects , Fenofibrate/pharmacology , Gene Expression Regulation/drug effects , Liver/drug effects , PPAR alpha/agonists , Animals , Autophagy/genetics , Autophagy-Related Protein 5/antagonists & inhibitors , Autophagy-Related Protein 5/genetics , Autophagy-Related Protein 5/metabolism , Autophagy-Related Protein 7/antagonists & inhibitors , Autophagy-Related Protein 7/genetics , Autophagy-Related Protein 7/metabolism , Autophagy-Related Proteins/antagonists & inhibitors , Autophagy-Related Proteins/genetics , Autophagy-Related Proteins/metabolism , Beclin-1/genetics , Beclin-1/metabolism , Blood Glucose/metabolism , Cysteine Endopeptidases/genetics , Cysteine Endopeptidases/metabolism , Fibroblast Growth Factors/genetics , Fibroblast Growth Factors/metabolism , Forkhead Box Protein O1/genetics , Forkhead Box Protein O1/metabolism , Liver/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , PPAR alpha/genetics , PPAR alpha/metabolism , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Sequestosome-1 Protein/genetics , Sequestosome-1 Protein/metabolism , Signal Transduction , Stearoyl-CoA Desaturase/genetics , Stearoyl-CoA Desaturase/metabolism , Sterol Regulatory Element Binding Protein 1/genetics , Sterol Regulatory Element Binding Protein 1/metabolism , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism , Triglycerides/metabolism , Ubiquitin-Conjugating Enzymes/antagonists & inhibitors , Ubiquitin-Conjugating Enzymes/genetics , Ubiquitin-Conjugating Enzymes/metabolism , fas Receptor/genetics , fas Receptor/metabolism
8.
Diabetes ; 65(6): 1472-80, 2016 06.
Article in English | MEDLINE | ID: mdl-27222391

ABSTRACT

Endoplasmic reticulum (ER) stress caused by perturbations in ER homeostasis activates an adaptive response termed the unfolded protein response (UPR) whose function is to resolve ER stress. If unsuccessful, the UPR initiates a proapoptotic program to eliminate the malfunctioning cells from the organism. It is the activation of this proapoptotic UPR in pancreatic ß-cells that has been implicated in the onset of type 2 diabetes and thus, in this context, is considered a maladaptive response. However, there is growing evidence that ß-cell death in type 2 diabetes may not be caused by a maladaptive UPR but by the inhibition of the adaptive UPR. In this review, we discuss the evidence for a role of the UPR in ß-cell dysfunction and death in the development of type 2 diabetes and ask the following question: Is ß-cell dysfunction the result of a maladaptive UPR or a failure of the UPR to adequately adapt? The answer to this question is critically important in defining potential therapeutic strategies for the treatment and prevention of type 2 diabetes. In addition, we discuss the potential role of the adaptive UPR in staving off type 2 diabetes by enhancing ß-cell mass and function in response to insulin resistance.


Subject(s)
Diabetes Mellitus, Type 2/physiopathology , Endoplasmic Reticulum Stress/physiology , Insulin Resistance , Insulin-Secreting Cells/physiology , Unfolded Protein Response/physiology , Adaptation, Physiological , Cell Death , Homeostasis , Humans , Insulin-Secreting Cells/metabolism , eIF-2 Kinase/physiology
9.
FASEB J ; 30(7): 2549-56, 2016 07.
Article in English | MEDLINE | ID: mdl-27022162

ABSTRACT

The accumulation of unfolded proteins within the endoplasmic reticulum (ER) causes ER stress and activation of unfolded protein response (UPR). This response can trigger ER-associated degradation and autophagy, which clear unfolded proteins and restore protein homeostasis. Recently, it has become clear that ubiquitination plays an important role in the regulation of autophagy. In the present study, we investigated how the E3 ubiquitin ligase neural precursor cell-expressed, developmentally down-regulated protein 4-2 (Nedd4-2) interacts with ER stress and autophagy. In mice, we found that an increase in the expression of Nedd4-2, which was concomitant with the activation of the UPR and autophagy, was caused by a prolonged high-fructose and high-fat diet that induces ER stress in the liver. Pharmacologic induction of ER stress also led to an increase in Nedd4-2 expression in cultured cells, which was coincident with UPR and autophagy activation. The inhibition of inositol-requiring enzyme 1 significantly suppressed Nedd4-2 expression. Moreover, increased Nedd4-2 expression in vivo was closely associated with the activation of inositol-requiring enzyme 1 and increased expression of the spliced form of X-box binding protein 1. Furthermore, knockdown of Nedd4-2 in cultured cells suppressed both basal autophagy and ER stress-induced autophagy, whereas overexpression of Nedd4-2-induced autophagy. Taken together, our findings provide evidence that Nedd4-2 is up-regulated in response to ER stress by the spliced form of X-box binding protein 1 and that this is important in the induction of an appropriate autophagic response.-Wang, H. Sun, R.-Q., Camera, D., Zeng, X.-Y., Jo, E., Chan, S. M. H., Herbert, T. P., Molero, J. C., Ye, J.-M. Endoplasmic reticulum stress up-regulates Nedd4-2 to induce autophagy.


Subject(s)
Autophagy/physiology , Endoplasmic Reticulum/physiology , Endosomal Sorting Complexes Required for Transport/metabolism , Gene Expression Regulation/physiology , Ubiquitin-Protein Ligases/metabolism , Up-Regulation/physiology , Animals , Endosomal Sorting Complexes Required for Transport/genetics , Fibroblasts/metabolism , HEK293 Cells , HeLa Cells , Humans , Liver/metabolism , Male , Mice , Nedd4 Ubiquitin Protein Ligases , Ubiquitin-Protein Ligases/genetics , X-Box Binding Protein 1/genetics , X-Box Binding Protein 1/metabolism
10.
J Mol Endocrinol ; 56(3): 175-87, 2016 04.
Article in English | MEDLINE | ID: mdl-26647387

ABSTRACT

The branched-chain amino acids (BCAA) leucine, isoleucine and valine, are essential amino acids that play a critical role in cellular signalling and metabolism. They acutely stimulate insulin secretion and activate the regulatory serine/threonine kinase mammalian target of rapamycin complex 1 (mTORC1), a kinase that promotes increased ß-cell mass and function. The effects of BCAA on cellular function are dependent on their active transport into the mammalian cells via amino acid transporters and thus the expression and activity of these transporters likely influence ß-cell signalling and function. In this report, we show that the System-L transporters are required for BCAA uptake into clonal ß-cell lines and pancreatic islets, and that these are essential for signalling to mTORC1. Further investigation revealed that the System-L amino acid transporter 1 (LAT1) is abundantly expressed in the islets, and that knockdown of LAT1 using siRNA inhibits mTORC1 signalling, leucine-stimulated insulin secretion and islet cell proliferation. In summary, we show that the LAT1 is required for regulating ß-cell signalling and function in islets and thus may be a novel pharmacological/nutritional target for the treatment and prevention of type 2 diabetes.


Subject(s)
Amino Acid Transport System L/metabolism , Insulin-Secreting Cells/metabolism , Signal Transduction , Amino Acid Transport System L/genetics , Animals , Cell Line, Tumor , Cell Proliferation , Gene Expression , Insulin/metabolism , Islets of Langerhans/metabolism , Large Neutral Amino Acid-Transporter 1/metabolism , Leucine/metabolism , Male , Mechanistic Target of Rapamycin Complex 1 , Multiprotein Complexes/metabolism , Rats , TOR Serine-Threonine Kinases/metabolism
11.
J Mol Endocrinol ; 53(1): 105-15, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24994913

ABSTRACT

Glucagon-like peptide 1 receptor (GLP1R) agonists, such as exendin-4, potentiate glucose-stimulated insulin secretion and are currently used in the management of type 2 diabetes. Interestingly, GLP1R agonists also have the ability to augment ß-cell mass. In this report, we provide evidence that in the presence of glucose, exendin-4 stimulates rodent islet cell DNA replication via the activation of ribosomal protein S6 kinase 1 (S6K1) and that this is mediated by the protein kinase B (PKB)-dependent activation of mTOR complex 1 (mTORC1). We show that activation of this pathway is caused by the autocrine or paracrine activation of the IGF1 receptor (IGF1R), as siRNA-mediated knockdown of the IGF1R effectively blocked exendin-4-stimulated PKB and mTORC1 activation. In contrast, pharmacological inactivation of the epidermal growth factor receptor has no discernible effect on exendin-4-stimulated PKB or mTORC1 activation. Therefore, we conclude that GLP1R agonists stimulate ß-cell proliferation via the PKB-dependent stimulation of mTORC1/S6K1 whose activation is mediated through the autocrine/paracrine activation of the IGF1R. This work provides a better understanding of the molecular basis of GLP1 agonist-induced ß-cell proliferation which could potentially be exploited in the identification of novel drug targets that increase ß-cell mass.


Subject(s)
Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/metabolism , Multiprotein Complexes/metabolism , Peptides/pharmacology , Receptor, IGF Type 1/metabolism , Ribosomal Protein S6 Kinases/metabolism , TOR Serine-Threonine Kinases/metabolism , Venoms/pharmacology , Animals , Cell Line , Cell Proliferation/drug effects , DNA Replication/drug effects , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/pathology , Exenatide , Gene Knockdown Techniques , Glucagon-Like Peptide-1 Receptor , Hypoglycemic Agents/pharmacology , Insulin-Secreting Cells/cytology , Male , Mechanistic Target of Rapamycin Complex 1 , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Wistar , Receptor, IGF Type 1/antagonists & inhibitors , Receptor, IGF Type 1/genetics , Receptors, Glucagon/agonists , Signal Transduction/drug effects
12.
Antioxid Redox Signal ; 20(14): 2091-106, 2014 May 10.
Article in English | MEDLINE | ID: mdl-24053669

ABSTRACT

AIMS: Age-related macular degeneration (AMD), a major cause of legal blindness in the elderly, is associated with genetic and environmental risk factors, such as cigarette smoking. Recent evidence shows that cigarette smoke (CS) that contains high levels of potent oxidants preferably targets retinal pigment epithelium (RPE) leading to oxidative damage and apoptosis; however, the mechanisms are poorly understood. The present study aimed to investigate the role of endoplasmic reticulum (ER) stress and the unfolded protein response (UPR) in CS-related RPE apoptosis. RESULTS: ER stress and proapoptotic gene C/EBP homologous protein (CHOP) were induced in the RPE/choroid complex from mice exposed to CS for 2 weeks and in human RPE cells treated with hydroquinone, a potent oxidant found at high concentrations in CS. Suppressing ER stress or inhibiting CHOP activation by pharmacological chaperones or genetic approaches attenuated hydroquinone-induced RPE cell apoptosis. In contrast to enhanced CHOP activation, protein level of active X-box binding protein 1 (XBP1), a major regulator of the adaptive UPR, was reduced in hydroquinone-treated cells. Conditional knockout of XBP1 gene in the RPE resulted in caspase-12 activation, increased CHOP expression, and decreased antiapoptotic gene Bcl-2. Furthermore, XBP1-deficient RPE cells are more sensitive to oxidative damage induced by hydroquinone or NaIO3, a CS-unrelated chemical oxidant. Conversely, overexpressing XBP1 protected RPE cells and attenuated oxidative stress-induced RPE apoptosis. INNOVATION AND CONCLUSION: These findings provide strong evidence suggesting an important role of ER stress and the UPR in CS-related oxidative injury of RPE cells. Thus, the modulation of the UPR signaling may provide a promising target for the treatment of AMD.


Subject(s)
Endoplasmic Reticulum Stress , Retinal Pigment Epithelium/metabolism , Retinal Pigment Epithelium/pathology , Unfolded Protein Response , Animals , Apoptosis/drug effects , DNA-Binding Proteins/metabolism , Endoplasmic Reticulum Stress/drug effects , Female , Humans , Hydroquinones/pharmacology , Macular Degeneration/metabolism , Macular Degeneration/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Oxidation-Reduction/drug effects , Regulatory Factor X Transcription Factors , Retinal Pigment Epithelium/drug effects , Smoking/adverse effects , Structure-Activity Relationship , Transcription Factor CHOP/metabolism , Transcription Factors/metabolism , Unfolded Protein Response/drug effects , X-Box Binding Protein 1
13.
PLoS One ; 7(3): e33004, 2012.
Article in English | MEDLINE | ID: mdl-22412973

ABSTRACT

Glucagon like peptide-1 (GLP-1) is released from intestinal L-cells in response to nutrient ingestion and acts upon pancreatic ß-cells potentiating glucose-stimulated insulin secretion and stimulating ß-cell proliferation, differentiation, survival and gene transcription. These effects are mediated through the activation of multiple signal transduction pathways including the extracellular regulated kinase (ERK) pathway. We have previously reported that GLP-1 activates ERK through a mechanism dependent upon the influx of extracellular Ca(2+) through L-type voltage gated Ca(2+) channels (VGCC). However, the mechanism by which L-type VGCCs couple to the ERK signalling pathway in pancreatic ß-cells is poorly understood. In this report, we characterise the relationship between L-type VGCC mediated changes in intracellular Ca(2+) concentration ([Ca(2+)](i)) and the activation of ERK, and demonstrate that the sustained activation of ERK (up to 30 min) in response to GLP-1 requires the continual activation of the L-type VGCC yet does not require a sustained increase in global [Ca(2+)](i) or Ca(2+) efflux from the endoplasmic reticulum. Moreover, sustained elevation of [Ca(2+)](i) induced by ionomycin is insufficient to stimulate the prolonged activation of ERK. Using the cell permeant Ca(2+) chelators, EGTA-AM and BAPTA-AM, to determine the spatial dynamics of L-type VGCC-dependent Ca(2+) signalling to ERK, we provide evidence that a sustained increase in Ca(2+) within the microdomain of the L-type VGCC is sufficient for signalling to ERK and that this plays an important role in GLP-1- stimulated ERK activation.


Subject(s)
Calcium Channels, L-Type/metabolism , Calcium/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Glucagon-Like Peptide 1/metabolism , Animals , Cell Line , Enzyme Activation/drug effects , Glucagon-Like Peptide 1/pharmacology , Mice , Signal Transduction/drug effects
14.
Acta Diabetol ; 49(4): 277-89, 2012 Aug.
Article in English | MEDLINE | ID: mdl-21833779

ABSTRACT

Muscarinic acetylcholine receptor (mAChR) activation of pancreatic ß-cells elevates intracellular Ca(2+) and potentiates glucose-stimulated insulin secretion. In addition, it activates a number of signaling molecules, including ERK1/2, whose activation has been shown to play an important role in regulating pancreatic ß-cell function and mass. The aim of this work was to determine how mAChR activation elevates intracellular Ca(2+) concentration ([Ca(2+)]( i )) and activates ERK1/2 in the pancreatic ß-cell line MIN6. We demonstrate that agonist-stimulated ERK1/2 activation is dependent on the activation of phospholipase C and an elevation in [Ca(2+)]( i ), but is independent of the activation of diacylglycerol-dependent protein kinase C isoenzymes. Using a pharmacological approach, we provide evidence that agonist-induced increases in [Ca(2+)]( i ) and ERK activity require (1) IP(3) receptor-mediated mobilization of Ca(2+) from the endoplasmic reticulum, (2) influx of extracellular Ca(2+) through store-operated channels, (3) closure of K(ATP) channels, and (4) Ca(2+) entry via L-type voltage-operated Ca(2+) channels. Moreover, this Ca(2+)-dependent activation of ERK is mediated via both Ras-dependent and Ras-independent mechanisms. In summary, this study provides important insights into the multifactorial signaling mechanisms linking mAChR activation to increases in [Ca(2+)]( i ) and ERK activity.


Subject(s)
Calcium/metabolism , Insulin-Secreting Cells/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Receptors, Muscarinic/physiology , Carbachol/pharmacology , Cell Line , Cholinergic Agonists/pharmacology , Enzyme Activation/physiology , Inositol 1,4,5-Trisphosphate Receptors/physiology , Mitogen-Activated Protein Kinase 3/metabolism , Phosphorylation , Signal Transduction , Transfection , Type C Phospholipases/antagonists & inhibitors , Type C Phospholipases/metabolism
15.
Cell Mol Life Sci ; 69(8): 1289-304, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22068611

ABSTRACT

Type-2 diabetes mellitus (T2DM) is a disorder that is characterized by high blood glucose concentration in the context of insulin resistance and/or relative insulin deficiency. It causes metabolic changes that lead to the damage and functional impairment of organs and tissues resulting in increased morbidity and mortality. It is this form of diabetes whose prevalence is increasing at an alarming rate due to the 'obesity epidemic', as obesity is a key risk factor in the development of insulin resistance. However, the majority of individuals who have insulin resistance do not develop diabetes due to a compensatory increase in insulin secretion in response to an increase in insulin demand. This adaptive response is sustained by an increase in both ß-cell function and mass. Importantly, there is increasing evidence that the Serine/Threonine kinase mammalian target of rapamycin (mTOR) plays a key role in the regulation of ß-cell mass and therefore likely plays a critical role in ß-cell adaptation. Therefore, the primary focus of this review is to summarize our current understanding of the role of mTOR in stimulating pancreatic ß-cell mass and thus, in the prevention of type-2 diabetes.


Subject(s)
Diabetes Mellitus, Type 2/metabolism , Insulin-Secreting Cells/metabolism , TOR Serine-Threonine Kinases/metabolism , Animals , Diabetes Mellitus, Type 2/genetics , Humans , Obesity/metabolism , TOR Serine-Threonine Kinases/genetics
16.
Cell Signal ; 23(12): 1927-35, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21763421

ABSTRACT

cAMP and mTOR signalling pathways control a number of critical cellular processes including metabolism, protein synthesis, proliferation and cell survival and therefore understanding the signalling events which integrate these two signalling pathways is of particular interest. In this study, we show that the pharmacological elevation of [cAMP](i) in mouse embryonic fibroblasts (MEFs) and human embryonic kidney 293 (HEK293) cells inhibits mTORC1 activation via a PKA-dependent mechanism. Although the inhibitory effect of cAMP on mTOR could be mediated by impinging on signalling cascades (i.e. PKB, MAPK and AMPK) that inhibit TSC1/2, an upstream negative regulator of mTORC1, we show that cAMP inhibits mTORC1 in TSC2 knockout (TSC2(-/-)) MEFs. We also show that cAMP inhibits insulin and amino acid-stimulated mTORC1 activation independently of Rheb, Rag GTPases, TSC2, PKB, MAPK and AMPK, indicating that cAMP may act independently of known regulatory inputs into mTOR. Moreover, we show that the prolonged elevation in [cAMP](i) can also inhibit mTORC2. We provide evidence that this cAMP-dependent inhibition of mTORC1/2 is caused by the dissociation of mTORC1 and 2 and a reduction in mTOR catalytic activity, as determined by its auto-phosphorylation on Ser2481. Taken together, these results provide an important insight into how cAMP signals to mTOR and down-regulates its activity, which may lead to the identification of novel drug targets to inhibit mTOR that could be used for the treatment and prevention of human diseases such as cancer.


Subject(s)
Cyclic AMP/physiology , Proteins/metabolism , Signal Transduction , Transcription Factors/metabolism , 1-Methyl-3-isobutylxanthine/pharmacology , Adaptor Proteins, Signal Transducing , Adenylate Kinase/metabolism , Amino Acids/pharmacology , Amino Acids/physiology , Animals , Carrier Proteins/metabolism , Cell Cycle Proteins , Cell Line , Colforsin/pharmacology , Cyclic AMP/chemistry , Cyclic AMP/metabolism , Eukaryotic Initiation Factors , Gene Deletion , Gene Knockout Techniques , Humans , Insulin/pharmacology , Insulin/physiology , Mechanistic Target of Rapamycin Complex 1 , Mice , Monomeric GTP-Binding Proteins/genetics , Monomeric GTP-Binding Proteins/metabolism , Multiprotein Complexes , Neuropeptides/genetics , Neuropeptides/metabolism , Phosphodiesterase Inhibitors/pharmacology , Phosphoproteins/metabolism , Phosphorylation , Protein Binding , Protein Kinases/metabolism , Proteins/agonists , Proteins/antagonists & inhibitors , Ras Homolog Enriched in Brain Protein , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Ribosomal Protein S6 Kinases, 90-kDa/metabolism , TOR Serine-Threonine Kinases , Transcription Factors/agonists , Transcription Factors/antagonists & inhibitors , Tuberous Sclerosis Complex 2 Protein , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
17.
Mol Endocrinol ; 25(2): 315-26, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21193559

ABSTRACT

Protein kinase R-like ER kinase (PERK) is activated at physiologically low glucose concentrations in pancreatic ß-cells. However, the molecular mechanisms by which PERK is activated under these conditions and its role in ß-cell function are poorly understood. In this report, we investigated, in dispersed rat islets of Langerhans and mouse insulinoma-6 (MIN6) cells, the relationship between extracellular glucose concentration, the free endoplasmic reticulum (ER) calcium concentration ([Ca(2+)](ER)) measured directly using an ER targeted fluorescence resonance energy transfer-based calcium sensor, and the activation of PERK. We found that a decrease in glucose concentration leads to a concentration-dependent reduction in [Ca(2+)](ER) that parallels the activation of PERK and the phosphorylation of its substrate eukaryotic initiation factor-2α. We provide evidence that this decrease in [Ca(2+)](ER) is caused by a decrease in sarcoplasmic/ER Ca(2+)-ATPase pump activity mediated by a reduction in the energy status of the cell. Importantly, we also report that PERK-dependent eukaryotic initiation factor-2α phosphorylation at low glucose concentration plays a significant role in 1) the regulation of both proinsulin and global protein synthesis, 2) cell viability, and 3) conferring preemptive cytoprotection against ER stress. Taken together, these results provide evidence that a decrease in the ATP/energy status of the cell in response to a decrease in glucose concentration results in sarcoplasmic/ER Ca(2+)-ATPase pump inhibition, the efflux of Ca(2+) from the ER, and the activation of PERK, which plays an important role in both pancreatic ß-cell function and survival.


Subject(s)
Calcium/metabolism , Endoplasmic Reticulum/metabolism , Glucose/metabolism , Insulin-Secreting Cells/metabolism , Sarcoplasmic Reticulum Calcium-Transporting ATPases/antagonists & inhibitors , eIF-2 Kinase/metabolism , Animals , Cell Line, Tumor , Cell Survival , Cells, Cultured , Enzyme Activation , Eukaryotic Initiation Factor-2/metabolism , Flow Cytometry , Fluorescence Resonance Energy Transfer , Insulin-Secreting Cells/cytology , Islets of Langerhans/metabolism , Male , Mice , Phosphorylation , Proinsulin/biosynthesis , Protein Biosynthesis , Rats , Rats, Wistar , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism
18.
J Mol Biol ; 389(3): 480-94, 2009 Jun 12.
Article in English | MEDLINE | ID: mdl-19376132

ABSTRACT

Ribosomal protein S6 (rpS6) is phosphorylated in vivo by isoforms of p70 S6 protein kinase and p90 ribosomal S6 kinase, and there is good evidence that it plays a positive role in controlling pancreatic beta-cell size and function. In this report, we demonstrate in the pancreatic beta-cell line MIN6 (mouse insulinoma cell line 6) and islets of Langerhans that agents which stimulate increases in cAMP, such as glucagon-like peptide-1 and forskolin, lead to the phosphorylation of rpS6 at Ser235/Ser236 independently of the activation of the currently known in vivo rpS6 kinases via a pathway that is sensitive to inhibitors of cAMP-dependent protein kinase [protein kinase A (PKA)]. This cAMP-dependent rpS6 kinase activity is also sensitive to PKI in vitro, and PKA exclusively phosphorylates recombinant rpS6 on Ser235/Ser236 in vitro. With these data taken together, we conclude that PKA can phosphorylate rpS6 exclusively at Ser235/Ser236 in vivo in pancreatic beta-cells, thus providing a potentially important link between cAMP signalling and the regulation of protein synthesis. Lastly, we provide evidence that PKA is also likely to phosphorylate rpS6 on Ser235/Ser236 in vivo in a number of other mammalian cell types.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/metabolism , Insulin-Secreting Cells/enzymology , Ribosomal Protein S6 Kinases/metabolism , Animals , Cell Line, Tumor , Colforsin/pharmacology , Glucagon-Like Peptide 1/pharmacology , Mice , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Phosphorylation , Rats , Ribosomal Protein S6 Kinases/genetics , Serine/genetics , Serine/metabolism
19.
J Am Soc Nephrol ; 19(11): 2119-29, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18650482

ABSTRACT

Insulin resistance is a major cause of muscle wasting in patients with ESRD. Uremic metabolic acidosis impairs insulin signaling, which normally suppresses proteolysis. The low pH may inhibit the SNAT2 l-Glutamine (L-Gln) transporter, which controls protein synthesis via amino acid-dependent insulin signaling through mammalian target of rapamycin (mTOR). Whether SNAT2 also regulates signaling to pathways that control proteolysis is unknown. In this study, inhibition of SNAT2 with the selective competitive substrate methylaminoisobutyrate or metabolic acidosis (pH 7.1) depleted intracellular L-Gln and stimulated proteolysis in cultured L6 myotubes. At pH 7.1, inhibition of the proteasome led to greater depletion of L-Gln, indicating that amino acids liberated by proteolysis sustain L-Gln levels when SNAT2 is inhibited by acidosis. Acidosis shifted the dose-response curve for suppression of proteolysis by insulin to the right, confirming that acid increases proteolysis by inducing insulin resistance. Blocking mTOR or phosphatidylinositol-3-kinase (PI3K) increased proteolysis, indicating that both signaling pathways are involved in its regulation. When both mTOR and PI3K were inhibited, methylaminoisobutyrate or acidosis did not stimulate proteolysis further. Moreover, partial silencing of SNAT2 expression in myotubes and myoblasts with small interfering RNA stimulated proteolysis and impaired insulin signaling through PI3K. In conclusion, SNAT2 not only regulates mTOR but also regulates proteolysis through PI3K and provides a link among acidosis, insulin resistance, and protein wasting in skeletal muscle cells.


Subject(s)
Acidosis/metabolism , Amino Acid Transport System A/antagonists & inhibitors , Muscle, Skeletal/metabolism , Amino Acid Transport System A/genetics , Amino Acid Transport System A/metabolism , Animals , Base Sequence , Carrier Proteins/metabolism , Cell Line , Glutamine/metabolism , Humans , Hydrogen-Ion Concentration , Insulin Resistance , Myoblasts, Skeletal/metabolism , Peptide Hydrolases/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Proto-Oncogene Proteins c-akt/metabolism , RNA, Small Interfering/genetics , Rats , Signal Transduction , TOR Serine-Threonine Kinases
20.
Biochem J ; 410(3): 485-93, 2008 Mar 15.
Article in English | MEDLINE | ID: mdl-18052927

ABSTRACT

In the present study, we demonstrate that, in pancreatic beta-cells, eIF2alpha (eukaryotic initiation factor 2alpha) phosphorylation in response to a decrease in glucose concentration is primarily mediated by the activation of PERK [PKR (protein kinase RNA activated)-like endoplasmic reticulum kinase]. We provide evidence that this increase in PERK activity is evoked by a decrease in the energy status of the cell via a potentially novel mechanism that is independent of IRE1 (inositol requiring enzyme 1) activation and the accumulation of unfolded nascent proteins within the endoplasmic reticulum. The inhibition of eIF2alpha phosphorylation in glucose-deprived cells by the overexpression of dominant-negative PERK or an N-terminal truncation mutant of GADD34 (growth-arrest and DNA-damage-inducible protein 34) leads to a 53% increase in the rate of total protein synthesis. Polysome analysis revealed that this coincides with an increase in the amplitude but not the number of ribosomes per mRNA, indicating that eIF2alpha dephosphorylation mobilizes hitherto untranslated mRNAs on to polysomes. In summary, we show that PERK is activated at low glucose concentrations in response to a decrease in energy status and that this plays an important role in glucose-regulated protein synthesis in pancreatic beta-cells.


Subject(s)
Energy Metabolism , Eukaryotic Initiation Factor-2/metabolism , Islets of Langerhans/metabolism , Protein Biosynthesis , eIF-2 Kinase/metabolism , Adenosine Triphosphate/metabolism , Animals , Base Sequence , Blotting, Western , DNA Primers , Electrophoresis, Polyacrylamide Gel , Enzyme Activation , Gene Silencing , Glucose/metabolism , Islets of Langerhans/cytology , Mice , Phosphorylation , Polymerase Chain Reaction , RNA, Small Interfering , eIF-2 Kinase/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...