Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
JCI Insight ; 6(10)2021 05 24.
Article in English | MEDLINE | ID: mdl-34027899

ABSTRACT

GWAS have shown that the common R325W variant of SLC30A8 (ZnT8) increases the risk of type 2 diabetes (T2D). However, ZnT8 haploinsufficiency is protective against T2D in humans, counterintuitive to earlier work in humans and mouse models. Therefore, whether decreasing ZnT8 activity is beneficial or detrimental to ß cell function, especially under conditions of metabolic stress, remains unknown. In order to examine whether the existence of human islet amyloid polypeptide (hIAPP), a coresident of the insulin granule, affects the role of ZnT8 in regulating ß cell function, hIAPP-expressing transgenics were generated with reduced ZnT8 (ZnT8B+/- hIAPP) or null ZnT8 (ZnT8B-/- hIAPP) expression specifically in ß cells. We showed that ZnT8B-/- hIAPP mice on a high-fat diet had intensified amyloid deposition and further impaired glucose tolerance and insulin secretion compared with control, ZnT8B-/-, and hIAPP mice. This can in part be attributed to impaired glucose sensing and islet cell synchronicity. Importantly, ZnT8B+/- hIAPP mice were also glucose intolerant and had reduced insulin secretion and increased amyloid aggregation compared with controls. These data suggest that loss of or reduced ZnT8 activity in ß cells heightened the toxicity induced by hIAPP, leading to impaired ß cell function and glucose homeostasis associated with metabolic stress.


Subject(s)
Amyloidosis/metabolism , Diabetes Mellitus, Type 2/metabolism , Insulin-Secreting Cells/metabolism , Islet Amyloid Polypeptide/metabolism , Zinc Transporter 8 , Animals , Disease Models, Animal , Humans , Islet Amyloid Polypeptide/genetics , Male , Mice , Mice, Transgenic , Zinc Transporter 8/genetics , Zinc Transporter 8/metabolism
2.
Diabetes Obes Metab ; 22(11): 2021-2031, 2020 11.
Article in English | MEDLINE | ID: mdl-32558194

ABSTRACT

AIM: To examine the mechanism of action of γ-aminobutyric acid (GABA) on ß-cell proliferation and investigate if co-treatment with Ly49, a novel GABA type A receptor positive allosteric modulator (GABAA -R PAM), amplifies this effect. METHODS: Human or mouse islets were co-treated for 4-5 days with GABA and selected receptor or cell signalling pathway modulators. Immunofluorescence was used to determine protein co-localization, cell number or proliferation, and islet size. Osmotic minipumps were surgically implanted in mice to assess Ly49 effects on pancreatic ß-cells. RESULTS: Amplification of GABAA -R signalling enhanced GABA-stimulated ß-cell proliferation in cultured mouse islets. Co-treatment of GABA with an inhibitor specific for PI3K, mTORC1/2, or p70S6K, abolished GABA-stimulated ß-cell proliferation in mouse and human islets. Nuclear p-AktSer473 and p-p70S6KThr421/Ser424 expression in pancreatic ß-cells was increased in GABA-treated mice compared with vehicle-treated mice, an effect augmented with GABA and Ly49 co-treatment. Mice co-treated with GABA and Ly49 exhibited enhanced ß-cell area and proliferation compared with GABA-treated mice. Furthermore, S961 injection (an insulin receptor antagonist) resulted in enhanced plasma insulin in GABA and Ly49 co-treated mice compared with GABA-treated mice. Importantly, GABA co-treated with Ly49 increased ß-cell proliferation in human islets providing a potential application for human subjects. CONCLUSIONS: We show that GABA stimulates ß-cell proliferation via the PI3K/mTORC1/p70S6K pathway in both mouse and human islets. Furthermore, we show that Ly49 enhances the ß-cell regenerative effects of GABA, showing potential in the intervention of diabetes.


Subject(s)
Receptors, GABA , Ribosomal Protein S6 Kinases, 70-kDa , Animals , Cell Proliferation , Mechanistic Target of Rapamycin Complex 1 , Mice , gamma-Aminobutyric Acid
3.
J Biol Chem ; 295(29): 9879-9892, 2020 07 17.
Article in English | MEDLINE | ID: mdl-32439805

ABSTRACT

Type 2 diabetes is a chronic metabolic disease characterized by pancreatic ß-cell dysfunction and peripheral insulin resistance. Among individuals with type 2 diabetes, ∼30% exhibit hypomagnesemia. Hypomagnesemia has been linked to insulin resistance through reduced tyrosine kinase activity of the insulin receptor; however, its impact on pancreatic ß-cell function is unknown. In this study, through analysis of several single-cell RNA-sequencing data sets in tandem with quantitative PCR validation in both murine and human islets, we identified NIPAL1 (NIPA-like domain containing 1), encoding a magnesium influx transporter, as an islet-enriched gene. A series of immunofluorescence experiments confirmed NIPAL1's magnesium-dependent expression and that it specifically localizes to the Golgi in Min6-K8 cells, a pancreatic ß-cell-like cell line (mouse insulinoma 6 clone K8). Under varying magnesium concentrations, NIPAL1 knockdown decreased both basal insulin secretion and total insulin content; in contrast, its overexpression increased total insulin content. Although the expression, distribution, and magnesium responsiveness of NIPAL1 in α-TC6 glucagonoma cells (a pancreatic α-cell line) were similar to the observations in Min6-K8 cells, no effect was observed on glucagon secretion in α-TC6 cells under the conditions studied. Overall, these results suggest that NIPAL1 expression is regulated by extracellular magnesium and that down-regulation of this transporter decreases glucose-stimulated insulin secretion and intracellular insulin content, particularly under conditions of hypomagnesemia.


Subject(s)
Cation Transport Proteins/biosynthesis , Insulin Secretion , Insulin-Secreting Cells/metabolism , Magnesium/metabolism , Animals , Cation Transport Proteins/genetics , Cell Line, Tumor , Gene Expression Regulation , Glucagon-Secreting Cells/cytology , Glucagon-Secreting Cells/metabolism , Insulin-Secreting Cells/cytology , Male , Mice
4.
Diabetes Obes Metab ; 21(1): 61-72, 2019 01.
Article in English | MEDLINE | ID: mdl-30062833

ABSTRACT

AIM: Omega-3 fatty acid ethyl ester supplements, available by prescription, are common in the treatment of dyslipidaemia in humans. Recent studies show that 3-carboxy-4-methyl-5-propyl-2-furanpropanoic acid (CMPF), a metabolite formed from fish oil supplementation, was able to prevent and reverse high fat diet (HFD)-induced fatty liver in mice. In the present study, we investigated the underlying molecular mechanisms responsible for CMPF's hepatic lipid-lowering effects. MATERIALS AND METHODS: CD1 male mice were i.p. injected with CMPF (dosage, 6 mg/kg) for 7 days, followed by 5 weeks of a 60% HFD to induce a fatty liver phenotype. Metabolic parameters, liver morphology, lipid content, protein expression and microarray analysis were assessed. We also utilized primary hepatocytes, an in vitro model, to further investigate the direct effects of CMPF on hepatic lipid utilization and biosynthesis. RESULTS: CMPF-treated mice display enhanced hepatic lipid clearance while hepatic lipid storage is prevented, thereby protecting against liver lipid accumulation and development of HFD-induced hepatic insulin resistance. Mechanistically, as CMPF enters the liver, it acts as an allosteric acetyl-coA carboxylase (ACC) inhibitor, which directly induces both fatty acid oxidation and hepatic production of fibroblast growth factor 21 (FGF21). A feed-back loop is initiated by CMPF, which exists between ACC inhibition, fatty acid oxidation and production of FGF21. As a consequence, an adaptive decrease in Insig2/SREBP-1c/FAS protein expression results in priming of the liver to prevent a HFD-induced fatty liver phenotype. CONCLUSION: CMPF is a potential driver of hepatic lipid metabolism, preventing diet-induced hepatic lipid deposition and insulin resistance in the long term.


Subject(s)
Diet, High-Fat/adverse effects , Furans/pharmacology , Insulin Resistance/physiology , Liver , Propionates/pharmacology , Animals , Blood Glucose/drug effects , Body Weight/drug effects , Fatty Liver/metabolism , Fibroblast Growth Factors/metabolism , Lipid Metabolism , Liver/chemistry , Liver/drug effects , Liver/metabolism , Liver/pathology , Male , Mice
5.
FASEB J ; 33(3): 3968-3984, 2019 03.
Article in English | MEDLINE | ID: mdl-30509117

ABSTRACT

γ-Aminobutyric acid (GABA) administration has been shown to increase ß-cell mass, leading to a reversal of type 1 diabetes in mice. Whether GABA has any effect on ß cells of healthy and prediabetic/glucose-intolerant obese mice remains unknown. In the present study, we show that oral GABA administration ( ad libitum) to mice indeed increased pancreatic ß-cell mass, which led to a modest enhancement in insulin secretion and glucose tolerance. However, GABA treatment did not further increase insulin-positive islet area in high fat diet-fed mice and was unable to prevent or reverse glucose intolerance and insulin resistance. Mechanistically, whether in vivo or in vitro, GABA treatment increased ß-cell proliferation. In vitro, the effect was shown to be mediated via the GABAA receptor. Single-cell RNA sequencing analysis revealed that GABA preferentially up-regulated pathways linked to ß-cell proliferation and simultaneously down-regulated those networks required for other processes, including insulin biosynthesis and metabolism. Interestingly, single-cell differential expression analysis revealed GABA treatment gave rise to a distinct subpopulation of ß cells with a unique transcriptional signature, including urocortin 3 ( ucn3), wnt4, and hepacam2. Taken together, this study provides new mechanistic insight into the proliferative nature of GABA but suggests that ß-cell compensation associated with prediabetes overlaps with, and negates, its proliferative effects.-Untereiner, A., Abdo, S., Bhattacharjee, A., Gohil, H., Pourasgari, F., Ibeh, N., Lai, M., Batchuluun, B., Wong, A., Khuu, N., Liu, Y., Al Rijjal, D., Winegarden, N., Virtanen, C., Orser, B. A., Cabrera, O., Varga, G., Rocheleau, J., Dai, F. F., Wheeler, M. B. GABA promotes ß-cell proliferation, but does not overcome impaired glucose homeostasis associated with diet-induced obesity.


Subject(s)
Cell Proliferation , Glucose/metabolism , Insulin-Secreting Cells/metabolism , Obesity/metabolism , Transcriptome , gamma-Aminobutyric Acid/pharmacology , Animals , Cell Line , Cells, Cultured , Diet, High-Fat/adverse effects , Homeostasis , Humans , Insulin/metabolism , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/physiology , Male , Mice , Mice, Inbred C57BL , Obesity/etiology , Receptors, GABA-A/metabolism , Urocortins/metabolism
6.
Diabetes ; 67(5): 885-897, 2018 05.
Article in English | MEDLINE | ID: mdl-29436377

ABSTRACT

Specific circulating metabolites have emerged as important risk factors for the development of diabetes. The acylcarnitines (acylCs) are a family of metabolites known to be elevated in type 2 diabetes (T2D) and linked to peripheral insulin resistance. However, the effect of acylCs on pancreatic ß-cell function is not well understood. Here, we profiled circulating acylCs in two diabetes cohorts: 1) women with gestational diabetes mellitus (GDM) and 2) women with recent GDM who later developed impaired glucose tolerance (IGT), new-onset T2D, or returned to normoglycemia within a 2-year follow-up period. We observed a specific elevation in serum medium-chain (M)-acylCs, particularly hexanoyl- and octanoylcarnitine, among women with GDM and individuals with T2D without alteration in long-chain acylCs. Mice treated with M-acylCs exhibited glucose intolerance, attributed to impaired insulin secretion. Murine and human islets exposed to elevated levels of M-acylCs developed defects in glucose-stimulated insulin secretion and this was directly linked to reduced mitochondrial respiratory capacity and subsequent ability to couple glucose metabolism to insulin secretion. In conclusion, our study reveals that an elevation in circulating M-acylCs is associated with GDM and early stages of T2D onset and that this elevation directly impairs ß-cell function.


Subject(s)
Carnitine/analogs & derivatives , Diabetes Mellitus, Type 2/metabolism , Diabetes, Gestational/metabolism , Glucose Intolerance/metabolism , Insulin-Secreting Cells/metabolism , Adult , Animals , Carnitine/metabolism , Carnitine/pharmacology , Case-Control Studies , Cell Respiration/drug effects , Disease Progression , Female , Humans , Insulin/metabolism , Insulin Secretion , Insulin-Secreting Cells/drug effects , Islets of Langerhans/drug effects , Islets of Langerhans/metabolism , Mice , Postpartum Period , Pregnancy
7.
J Biol Chem ; 290(41): 25045-61, 2015 Oct 09.
Article in English | MEDLINE | ID: mdl-26272612

ABSTRACT

GLP1 activates its receptor, GLP1R, to enhance insulin secretion. The activation and transduction of GLP1R requires complex interactions with a host of accessory proteins, most of which remain largely unknown. In this study, we used membrane-based split ubiquitin yeast two-hybrid assays to identify novel GLP1R interactors in both mouse and human islets. Among these, ATP6ap2 (ATPase H(+)-transporting lysosomal accessory protein 2) was identified in both mouse and human islet screens. ATP6ap2 was shown to be abundant in islets including both alpha and beta cells. When GLP1R and ATP6ap2 were co-expressed in beta cells, GLP1R was shown to directly interact with ATP6ap2, as assessed by co-immunoprecipitation. In INS-1 cells, overexpression of ATP6ap2 did not affect insulin secretion; however, siRNA knockdown decreased both glucose-stimulated and GLP1-induced insulin secretion. Decreases in GLP1-induced insulin secretion were accompanied by attenuated GLP1 stimulated cAMP accumulation. Because ATP6ap2 is a subunit required for V-ATPase assembly of insulin granules, it has been reported to be involved in granule acidification. In accordance with this, we observed impaired insulin granule acidification upon ATP6ap2 knockdown but paradoxically increased proinsulin secretion. Importantly, as a GLP1R interactor, ATP6ap2 was required for GLP1-induced Ca(2+) influx, in part explaining decreased insulin secretion in ATP6ap2 knockdown cells. Taken together, our findings identify a group of proteins that interact with the GLP1R. We further show that one interactor, ATP6ap2, plays a novel dual role in beta cells, modulating both GLP1R signaling and insulin processing to affect insulin secretion.


Subject(s)
Glucagon-Like Peptide-1 Receptor/metabolism , Insulin-Secreting Cells/metabolism , Insulin/metabolism , Proton-Translocating ATPases/metabolism , Receptors, Cell Surface/metabolism , Vacuolar Proton-Translocating ATPases/metabolism , Animals , Biological Transport/drug effects , CHO Cells , Calcium/metabolism , Cricetinae , Cricetulus , Cyclic AMP/metabolism , Gene Knockdown Techniques , Glucagon-Like Peptide 1/pharmacology , Humans , Insulin Secretion , Insulin-Secreting Cells/drug effects , Male , Mice , Protein Binding , Proton-Translocating ATPases/deficiency , Proton-Translocating ATPases/genetics , Receptors, Cell Surface/deficiency , Receptors, Cell Surface/genetics , Vacuolar Proton-Translocating ATPases/deficiency , Vacuolar Proton-Translocating ATPases/genetics
8.
PLoS One ; 10(6): e0129226, 2015.
Article in English | MEDLINE | ID: mdl-26075596

ABSTRACT

Glucagon regulates glucose homeostasis by controlling glycogenolysis and gluconeogenesis in the liver. Exaggerated and dysregulated glucagon secretion can exacerbate hyperglycemia contributing to type 2 diabetes (T2D). Thus, it is important to understand how glucagon receptor (GCGR) activity and signaling is controlled in hepatocytes. To better understand this, we sought to identify proteins that interact with the GCGR to affect ligand-dependent receptor activation. A Flag-tagged human GCGR was recombinantly expressed in Chinese hamster ovary (CHO) cells, and GCGR complexes were isolated by affinity purification (AP). Complexes were then analyzed by mass spectrometry (MS), and protein-GCGR interactions were validated by co-immunoprecipitation (Co-IP) and Western blot. This was followed by studies in primary hepatocytes to assess the effects of each interactor on glucagon-dependent glucose production and intracellular cAMP accumulation, and then in immortalized CHO and liver cell lines to further examine cell signaling. Thirty-three unique interactors were identified from the AP-MS screening of GCGR expressing CHO cells in both glucagon liganded and unliganded states. These studies revealed a particularly robust interaction between GCGR and 5 proteins, further validated by Co-IP, Western blot and qPCR. Overexpression of selected interactors in mouse hepatocytes indicated that two interactors, LDLR and TMED2, significantly enhanced glucagon-stimulated glucose production, while YWHAB inhibited glucose production. This was mirrored with glucagon-stimulated cAMP production, with LDLR and TMED2 enhancing and YWHAB inhibiting cAMP accumulation. To further link these interactors to glucose production, key gluconeogenic genes were assessed. Both LDLR and TMED2 stimulated while YWHAB inhibited PEPCK and G6Pase gene expression. In the present study, we have probed the GCGR interactome and found three novel GCGR interactors that control glucagon-stimulated glucose production by modulating cAMP accumulation and genes that control gluconeogenesis. These interactors may be useful targets to control glucose homeostasis in T2D.


Subject(s)
Liver/metabolism , Protein Interaction Mapping , Protein Interaction Maps , Proteomics , Receptors, Glucagon/agonists , Receptors, Glucagon/metabolism , Animals , CHO Cells , Carrier Proteins , Cell Line , Cricetulus , Cyclic AMP/metabolism , Gene Expression Regulation , Gluconeogenesis/genetics , Glucose/metabolism , Hepatocytes/metabolism , Mice , Protein Binding , Proteomics/methods , Receptors, G-Protein-Coupled , Reproducibility of Results
9.
J Biol Chem ; 290(30): 18757-69, 2015 Jul 24.
Article in English | MEDLINE | ID: mdl-25969539

ABSTRACT

Zinc plays an essential role in the regulation of pancreatic ß cell function, affecting important processes including insulin biosynthesis, glucose-stimulated insulin secretion, and cell viability. Mutations in the zinc efflux transport protein ZnT8 have been linked with both type 1 and type 2 diabetes, further supporting an important role for zinc in glucose homeostasis. However, very little is known about how cytosolic zinc is controlled by zinc influx transporters (ZIPs). In this study, we examined the ß cell and islet ZIP transcriptome and show consistent high expression of ZIP6 (Slc39a6) and ZIP7 (Slc39a7) genes across human and mouse islets and MIN6 ß cells. Modulation of ZIP6 and ZIP7 expression significantly altered cytosolic zinc influx in pancreatic ß cells, indicating an important role for ZIP6 and ZIP7 in regulating cellular zinc homeostasis. Functionally, this dysregulated cytosolic zinc homeostasis led to impaired insulin secretion. In parallel studies, we identified both ZIP6 and ZIP7 as potential interacting proteins with GLP-1R by a membrane yeast two-hybrid assay. Knock-down of ZIP6 but not ZIP7 in MIN6 ß cells impaired the protective effects of GLP-1 on fatty acid-induced cell apoptosis, possibly via reduced activation of the p-ERK pathway. Therefore, our data suggest that ZIP6 and ZIP7 function as two important zinc influx transporters to regulate cytosolic zinc concentrations and insulin secretion in ß cells. In particular, ZIP6 is also capable of directly interacting with GLP-1R to facilitate the protective effect of GLP-1 on ß cell survival.


Subject(s)
Cation Transport Proteins/metabolism , Diabetes Mellitus/genetics , Insulin-Secreting Cells/pathology , Neoplasm Proteins/metabolism , Zinc/metabolism , Animals , Apoptosis , Cation Transport Proteins/biosynthesis , Cation Transport Proteins/genetics , Cytosol/metabolism , Diabetes Mellitus/metabolism , Diabetes Mellitus/pathology , Glucagon-Like Peptide 1/genetics , Glucagon-Like Peptide 1/metabolism , Glucagon-Like Peptide-1 Receptor , Homeostasis , Humans , Insulin/genetics , Insulin/metabolism , Insulin Secretion , Insulin-Secreting Cells/metabolism , MAP Kinase Signaling System/genetics , Mice , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Receptors, Glucagon/genetics , Receptors, Glucagon/metabolism
10.
Mol Cell Proteomics ; 13(11): 3049-62, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25044020

ABSTRACT

Glucagon-like peptide-1 (GLP-1) is an incretin hormone that regulates glucose homeostasis. Because of their direct stimulation of insulin secretion from pancreatic ß cells, GLP-1 receptor (GLP-1R) agonists are now important therapeutic options for the treatment of type 2 diabetes. To better understand the mechanisms that control the insulinotropic actions of GLP-1, affinity purification and mass spectrometry (AP-MS) were employed to uncover potential proteins that functionally interact with the GLP-1R. AP-MS performed on Chinese hamster ovary cells or MIN6 ß cells, both expressing the human GLP-1R, revealed 99 proteins potentially associated with the GLP-1R. Three novel GLP-1R interactors (PGRMC1, Rab5b, and Rab5c) were further validated through co-immunoprecipitation/immunoblotting, fluorescence resonance energy transfer, and immunofluorescence. Functional studies revealed that overexpression of PGRMC1, a novel cell surface receptor that associated with liganded GLP-1R, enhanced GLP-1-induced insulin secretion (GIIS) with the most robust effect. Knockdown of PGRMC1 in ß cells decreased GIIS, indicative of positive interaction with GLP-1R. To gain insight mechanistically, we demonstrated that the cell surface PGRMC1 ligand P4-BSA increased GIIS, whereas its antagonist AG-205 decreased GIIS. It was then found that PGRMC1 increased GLP-1-induced cAMP accumulation. PGRMC1 activation and GIIS induced by P4-BSA could be blocked by inhibition of adenylyl cyclase/EPAC signaling or the EGF receptor-PI3K signal transduction pathway. These data reveal a dual mechanism for PGRMC1-increased GIIS mediated through cAMP and EGF receptor signaling. In conclusion, we identified several novel GLP-1R interacting proteins. PGRMC1 expressed on the cell surface of ß cells was shown to interact with the activated GLP-1R to enhance the insulinotropic actions of GLP-1.


Subject(s)
Glucagon-Like Peptide 1/metabolism , Insulin-Secreting Cells/metabolism , Insulin/metabolism , Membrane Proteins/metabolism , Receptors, Glucagon/metabolism , Receptors, Progesterone/metabolism , Adenylyl Cyclase Inhibitors , Animals , CHO Cells , Cell Line , Cricetinae , Cricetulus , Cyclic AMP/biosynthesis , Cyclic AMP/metabolism , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/metabolism , Glucagon-Like Peptide-1 Receptor , Guanine Nucleotide Exchange Factors/antagonists & inhibitors , Guanine Nucleotide Exchange Factors/metabolism , Humans , Insulin Secretion , Mass Spectrometry , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/genetics , Mice , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Rats , Receptors, Progesterone/antagonists & inhibitors , Receptors, Progesterone/genetics , rab5 GTP-Binding Proteins/metabolism
11.
J Lipid Res ; 55(3): 516-23, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24443560

ABSTRACT

Adipose tissue contains one of the largest reservoirs of cholesterol in the body. Adipocyte dysfunction in obesity is associated with intracellular cholesterol accumulation, and alterations in cholesterol homeostasis have been shown to alter glucose metabolism in cultured adipocytes. ABCA1 plays a major role in cholesterol efflux, suggesting a role for ABCA1 in maintaining cholesterol homeostasis in the adipocyte. However, the impact of adipocyte ABCA1 on adipose tissue function and glucose metabolism is unknown. Our aim was to determine the impact of adipocyte ABCA1 on adipocyte lipid metabolism, body weight, and glucose metabolism in vivo. To address this, we used mice lacking ABCA1 specifically in adipocytes (ABCA1(-ad/-ad)). When fed a high-fat, high-cholesterol diet, ABCA1(-ad/-ad) mice showed increased cholesterol and triglyceride stores in adipose tissue, developed enlarged fat pads, and had increased body weight. Associated with these phenotypic changes, we observed significant changes in the expression of genes involved in cholesterol and glucose homeostasis, including ldlr, abcg1, glut-4, adiponectin, and leptin. ABCA1(-ad/-ad) mice also demonstrated impaired glucose tolerance, lower insulin sensitivity, and decreased insulin secretion. We conclude that ABCA1 in adipocytes influences adipocyte lipid metabolism, body weight, and whole-body glucose homeostasis.


Subject(s)
ATP Binding Cassette Transporter 1/deficiency , Adipocytes/metabolism , Adipose Tissue/metabolism , Blood Glucose/metabolism , Insulin Resistance , Lipids/analysis , ATP Binding Cassette Transporter 1/genetics , Adipocytes/cytology , Adipose Tissue/cytology , Animals , Blotting, Western , Body Weight , Cholesterol/metabolism , Diet, High-Fat , Gene Expression , Glucose/metabolism , Glucose Transporter Type 4/genetics , Glucose Transporter Type 4/metabolism , Homeostasis/genetics , Leptin/genetics , Leptin/metabolism , Lipids/blood , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Nicotinamide Phosphoribosyltransferase/genetics , Nicotinamide Phosphoribosyltransferase/metabolism , Receptors, LDL/genetics , Receptors, LDL/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Triglycerides/metabolism
12.
Arterioscler Thromb Vasc Biol ; 33(12): 2724-32, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24135019

ABSTRACT

OBJECTIVE: The ATP-binding cassette transporter A1 (ABCA1) protein maintains cellular cholesterol homeostasis in several different tissues. In the liver, ABCA1 is crucial for high-density lipoprotein biogenesis, and in the pancreas ABCA1 can regulate insulin secretion. In this study, our aim was to identify novel microRNAs that regulate ABCA1 expression in these tissues. APPROACH AND RESULTS: We combined multiple microRNA prediction programs to identify 8 microRNAs that potentially regulate ABCA1. A luciferase reporter assay demonstrated that 5 of these microRNAs (miR-148, miR-27, miR-144, miR-145, and miR-33a/33b) significantly repressed ABCA1 3'-untranslated region activity with miR-145 resulting in one of the larger decreases. In hepatic HepG2 cells, miR-145 can regulate both ABCA1 protein expression levels and cholesterol efflux function. In murine islets, an increase in miR-145 expression decreased ABCA1 protein expression, increased total islet cholesterol levels, and decreased glucose-stimulated insulin secretion. Inhibiting miR-145 produced the opposite effect of increasing ABCA1 protein levels and improving glucose-stimulated insulin secretion. Finally, increased glucose levels in media significantly decreased miR-145 levels in cultured pancreatic beta cells. These findings suggest that miR-145 is involved in glucose homeostasis and is regulated by glucose concentration. CONCLUSIONS: Our studies demonstrate that miR-145 regulates ABCA1 expression and function, and inhibiting this microRNA represents a novel strategy for increasing ABCA1 expression, promoting high-density lipoprotein biogenesis in the liver, and improving glucose-stimulated insulin secretion in islets.


Subject(s)
ATP Binding Cassette Transporter 1/metabolism , Hepatocytes/metabolism , Islets of Langerhans/metabolism , MicroRNAs/metabolism , 3' Untranslated Regions , ATP Binding Cassette Transporter 1/genetics , Animals , Binding Sites , Cholesterol/metabolism , Gene Expression Regulation , Genes, Reporter , Glucose/metabolism , Hep G2 Cells , Homeostasis , Humans , Insulin/metabolism , Insulin-Secreting Cells/metabolism , Lipoproteins, HDL/metabolism , Mice , Transfection
13.
Can J Physiol Pharmacol ; 90(5): 663-8, 2012 May.
Article in English | MEDLINE | ID: mdl-22530993

ABSTRACT

Dipeptidyl peptidase-4 (DPP-4) inhibitors increase circulating levels of incretin hormones, which can enhance insulin secretion and ß cell function. The aim of this study was to evaluate the effectiveness of MK-626 (a novel DPP-4 inhibitor) to reduce the hyperglycemia and hyperinsulinemia of nonobese type 2 diabetic MKR mice. Twelve to 14-week-old hyperglycemic MKR mice were gavaged daily with MK-626 (3 mg/kg body weight) or vehicle (0.5% methyl cellulose (MC)) for 2 weeks. MK-626-treated mice displayed no change in body weight or adverse reactions, suggesting good tolerance of the drug. Fed blood glucose was significantly reduced over the 2-week experiment; however, it was also reduced in the MC group, suggesting an effect of gavage alone. Fed plasma insulin and glucagon levels and glucose tolerance of MK-626-treated mice were similar to those of MC mice. Therefore, treatment with MK-626 did not correct the prolonged hyperglycemia and impaired glucose tolerance of MKR mice.


Subject(s)
Blood Glucose/drug effects , Dipeptidyl-Peptidase IV Inhibitors/pharmacology , Hyperglycemia/drug therapy , Hyperinsulinism/drug therapy , Insulin/blood , Phenylalanine/analogs & derivatives , Triazoles/pharmacology , Animals , Blood Glucose/metabolism , Body Weight/drug effects , Diabetes Mellitus, Experimental/blood , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/metabolism , Glucagon/blood , Glucagon/metabolism , Glucose/metabolism , Glucose Intolerance/drug therapy , Glucose Intolerance/metabolism , Homeostasis/drug effects , Hyperglycemia/blood , Hyperglycemia/metabolism , Hyperinsulinism/blood , Hyperinsulinism/metabolism , Incretins/blood , Incretins/metabolism , Insulin/metabolism , Insulin Secretion , Islets of Langerhans/drug effects , Islets of Langerhans/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, Obese/blood , Mice, Obese/metabolism , Phenylalanine/pharmacology
14.
Diabetes ; 61(3): 659-64, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22315310

ABSTRACT

Cellular cholesterol homeostasis is important for normal ß-cell function. Disruption of cholesterol transport by decreased function of the ATP-binding cassette (ABC) transporter ABCA1 results in impaired insulin secretion. Mice lacking ß-cell ABCA1 have increased islet expression of ABCG1, another cholesterol transporter implicated in ß-cell function. To determine whether ABCA1 and ABCG1 have complementary roles in ß-cells, mice lacking ABCG1 and ß-cell ABCA1 were generated and glucose tolerance, islet sterol levels, and ß-cell function were assessed. Lack of both ABCG1 and ß-cell ABCA1 resulted in increased fasting glucose levels and a greater impairment in glucose tolerance compared with either ABCG1 deletion or loss of ABCA1 in ß-cells alone. In addition, glucose-stimulated insulin secretion was decreased and sterol accumulation increased in islets lacking both transporters compared with those isolated from knockout mice with each gene alone. Combined deficiency of ABCA1 and ABCG1 also resulted in significant islet inflammation as indicated by increased expression of interleukin-1ß and macrophage infiltration. Thus, lack of both ABCA1 and ABCG1 induces greater defects in ß-cell function than deficiency of either transporter individually. These data suggest that ABCA1 and ABCG1 each make complimentary and important contributions to ß-cell function by maintaining islet cholesterol homeostasis in vivo.


Subject(s)
ATP-Binding Cassette Transporters/physiology , Cholesterol/metabolism , Homeostasis , Inflammation/etiology , Insulin-Secreting Cells/physiology , Islets of Langerhans/metabolism , Lipoproteins/physiology , ATP Binding Cassette Transporter 1 , ATP Binding Cassette Transporter, Subfamily G, Member 1 , Animals , Glucose Intolerance/etiology , Interleukin-1beta/genetics , Macrophages/physiology , Mice , Mice, Inbred C57BL , Transcription Factor CHOP/physiology
15.
Diabetes ; 61(3): 653-8, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22315319

ABSTRACT

Changes in cellular cholesterol affect insulin secretion, and ß-cell-specific deletion or loss-of-function mutations in the cholesterol efflux transporter ATP-binding cassette transporter A1 (ABCA1) result in impaired glucose tolerance and ß-cell dysfunction. Upregulation of ABCA1 expression may therefore be beneficial for the maintenance of normal islet function in diabetes. Studies suggest that microRNA-33a (miR-33a) expression inversely correlates with ABCA1 expression in hepatocytes and macrophages. We examined whether miR-33a regulates ABCA1 expression in pancreatic islets, thereby affecting cholesterol accumulation and insulin secretion. Adenoviral miR-33a overexpression in human or mouse islets reduced ABCA1 expression, decreased glucose-stimulated insulin secretion, and increased cholesterol levels. The miR-33a-induced reduction in insulin secretion was rescued by cholesterol depletion by methyl-ß-cyclodextrin or mevastatin. Inhibition of miR-33a expression in apolipoprotein E knockout islets and ABCA1 overexpression in ß-cell-specific ABCA1 knockout islets rescued normal insulin secretion and reduced islet cholesterol. These findings confirm the critical role of ß-cell ABCA1 in islet cholesterol homeostasis and ß-cell function and highlight modulation of ß-cell miR-33a expression as a means to influence insulin secretion.


Subject(s)
ATP-Binding Cassette Transporters/physiology , Cholesterol/metabolism , Insulin/metabolism , Islets of Langerhans/metabolism , MicroRNAs/physiology , ATP Binding Cassette Transporter 1 , ATP-Binding Cassette Transporters/analysis , ATP-Binding Cassette Transporters/genetics , Animals , Glucose/pharmacology , Humans , Insulin Secretion , Mice , beta-Cyclodextrins/pharmacology
16.
Diabetes ; 60(11): 2710-9, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21984579

ABSTRACT

OBJECTIVE: The role of uncoupling protein 2 (UCP2) in pancreatic ß-cells is highly debated, partly because of the broad tissue distribution of UCP2 and thus limitations of whole-body UCP2 knockout mouse models. To investigate the function of UCP2 in the ß-cell, ß-cell-specific UCP2 knockout mice (UCP2BKO) were generated and characterized. RESEARCH DESIGN AND METHODS: UCP2BKO mice were generated by crossing loxUCP2 mice with mice expressing rat insulin promoter-driven Cre recombinase. Several in vitro and in vivo parameters were measured, including respiration rate, mitochondrial membrane potential, islet ATP content, reactive oxygen species (ROS) levels, glucose-stimulated insulin secretion (GSIS), glucagon secretion, glucose and insulin tolerance, and plasma hormone levels. RESULTS: UCP2BKO ß-cells displayed mildly increased glucose-induced mitochondrial membrane hyperpolarization but unchanged rates of uncoupled respiration and islet ATP content. UCP2BKO islets had elevated intracellular ROS levels that associated with enhanced GSIS. Surprisingly, UCP2BKO mice were glucose-intolerant, showing greater α-cell area, higher islet glucagon content, and aberrant ROS-dependent glucagon secretion under high glucose conditions. CONCLUSIONS: Using a novel ß-cell-specific UCP2KO mouse model, we have shed light on UCP2 function in primary ß-cells. UCP2 does not behave as a classical metabolic uncoupler in the ß-cell, but has a more prominent role in the regulation of intracellular ROS levels that contribute to GSIS amplification. In addition, ß-cell UCP2 contributes to the regulation of intraislet ROS signals that mediate changes in α-cell morphology and glucagon secretion.


Subject(s)
Glucagon-Secreting Cells/metabolism , Glucagon/metabolism , Insulin-Secreting Cells/metabolism , Insulin/metabolism , Ion Channels/physiology , Mitochondrial Proteins/physiology , Reactive Oxygen Species/metabolism , Animals , Genes, Reporter , Glucagon-Secreting Cells/pathology , Glucose Intolerance/genetics , Glucose Intolerance/metabolism , Glucose Intolerance/pathology , Humans , Hyperglycemia/metabolism , Insulin/genetics , Insulin Secretion , Insulin-Secreting Cells/pathology , Ion Channels/genetics , Islets of Langerhans/metabolism , Islets of Langerhans/pathology , Male , Membrane Potential, Mitochondrial , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Mitochondrial Proteins/genetics , Organ Specificity , Promoter Regions, Genetic , Rats , Tissue Culture Techniques , Uncoupling Protein 2
17.
Diabetes ; 60(12): 3186-96, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21998401

ABSTRACT

OBJECTIVE: The ATP-binding cassette transporter A1 (ABCA1) is essential for normal insulin secretion from ß-cells. The aim of this study was to elucidate the mechanisms underlying the impaired insulin secretion in islets lacking ß-cell ABCA1. RESEARCH DESIGN AND METHODS: Calcium imaging, patch clamp, and membrane capacitance were used to assess the effect of ABCA1 deficiency on calcium flux, ion channel function, and exocytosis in islet cells. Electron microscopy was used to analyze ß-cell ultrastructure. The quantity and distribution of proteins involved in insulin-granule exocytosis were also investigated. RESULTS: We show that a lack of ß-cell ABCA1 results in impaired depolarization-induced exocytotic fusion of insulin granules. We observed disturbances in membrane microdomain organization and Golgi and insulin granule morphology in ß-cells as well as elevated fasting plasma proinsulin levels in mice in the absence of ß-cell ABCA1. Acute cholesterol depletion rescued the exocytotic defect in ß-cells lacking ABCA1, indicating that elevated islet cholesterol accumulation directly impairs granule fusion and insulin secretion. CONCLUSIONS: Our data highlight a crucial role of ABCA1 and cellular cholesterol in ß-cells that is necessary for regulated insulin granule fusion events. These data suggest that abnormalities of cholesterol metabolism may contribute to the impaired ß-cell function in diabetes.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Cholesterol/metabolism , Exocytosis/physiology , Insulin-Secreting Cells/metabolism , Insulin/metabolism , Islets of Langerhans/metabolism , ATP Binding Cassette Transporter 1 , ATP-Binding Cassette Transporters/genetics , Animals , Blotting, Western , Calcium/metabolism , Calcium Channels/metabolism , Cell Line , Cell Line, Tumor , Cell Membrane/metabolism , Electrophysiology , Exocytosis/genetics , Glucose Intolerance/genetics , Glucose Intolerance/metabolism , Mice , Mice, Knockout , Microscopy, Electron, Transmission
18.
J Biol Chem ; 285(44): 33623-31, 2010 Oct 29.
Article in English | MEDLINE | ID: mdl-20709750

ABSTRACT

The functional impact of adiponectin on pancreatic beta cells is so far poorly understood. Although adiponectin receptors (AdipoR1/2) were identified, their involvement in adiponectin-induced signaling and other molecules involved is not clearly defined. Therefore, we investigated the role of adiponectin in beta cells and the signaling mediators involved. MIN6 beta cells and mouse islets were stimulated with globular (2.5 µg/ml) or full-length (5 µg/ml) adiponectin under serum starvation, and cell viability, proliferation, apoptosis, insulin gene expression, and secretion were measured. Lysates were subjected to Western blot analysis to determine phosphorylation of AMP-activated protein kinase (AMPK), Akt, or ERK. Functional significance of signaling was confirmed using dominant negative mutants or pharmacological inhibitors. Participation of AdipoRs was assessed by overexpression or siRNA. Adiponectin failed to activate AMPK after 10 min or 1- and 24-h stimulation. ERK was significantly phosphorylated after 24-h treatment with adiponectin, whereas Akt was activated at all time points examined. 24-h stimulation with adiponectin significantly increased cell viability by decreasing cellular apoptosis, and this was prevented by dominant negative Akt, wortmannin (PI3K inhibitor), and U0126 (MEK inhibitor). Moreover, adiponectin regulated insulin gene expression and glucose-stimulated insulin secretion, which was also prevented by wortmannin and U0126 treatment. Interestingly, the data also suggest adiponectin-induced changes in Akt and ERK phosphorylation and caspase-3 may occur independent of the level of AdipoR expression. This study demonstrates a lack of AMPK involvement and implicates Akt and ERK in adiponectin signaling, leading to protection against apoptosis and stimulation of insulin gene expression and secretion in pancreatic beta cells.


Subject(s)
Adiponectin/metabolism , Apoptosis , Gene Expression Regulation , Insulin-Secreting Cells/cytology , Insulin/biosynthesis , Mitogen-Activated Protein Kinase 3/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Animals , Cell Line , Enzyme Inhibitors/pharmacology , Insulin/metabolism , Insulin Secretion , Insulin-Secreting Cells/pathology , Mice , Phosphorylation , RNA, Small Interfering/metabolism , Signal Transduction , Time Factors
19.
J Biol Chem ; 284(44): 30441-52, 2009 Oct 30.
Article in English | MEDLINE | ID: mdl-19690348

ABSTRACT

Voltage-gated eag-related gene (Erg) K(+) channels regulate the electrical activity of many cell types. Data regarding Erg channel expression and function in electrically excitable glucagon and insulin producing cells of the pancreas is limited. In the present study Erg1 mRNA and protein were shown to be highly expressed in human and mouse islets and in alpha-TC6 and Min6 cells alpha- and beta-cell lines, respectively. Whole cell patch clamp recordings demonstrated the functional expression of Erg1 in alpha- and beta-cells, with rBeKm1, an Erg1 antagonist, blocking inward tail currents elicited by a double pulse protocol. Additionally, a small interference RNA approach targeting the kcnh2 gene (Erg1) induced a significant decrease of Erg1 inward tail current in Min6 cells. To investigate further the role of Erg channels in mouse and human islets, ratiometric Fura-2 AM Ca(2+)-imaging experiments were performed on isolated alpha- and beta-cells. Blocking Erg channels with rBeKm1 induced a transient cytoplasmic Ca(2+) increase in both alpha- and beta-cells. This resulted in an increased glucose-dependent insulin secretion, but conversely impaired glucagon secretion under low glucose conditions. Together, these data present Erg1 channels as new mediators of alpha- and beta-cell repolarization. However, antagonism of Erg1 has divergent effects in these cells; to augment glucose-dependent insulin secretion and inhibit low glucose stimulated glucagon secretion.


Subject(s)
Ether-A-Go-Go Potassium Channels/metabolism , Glucagon-Secreting Cells/chemistry , Insulin-Secreting Cells/chemistry , Islets of Langerhans/cytology , Animals , Calcium/metabolism , Glucagon/metabolism , Humans , Insulin/metabolism , Insulin Secretion , Membrane Potentials , Mice , Patch-Clamp Techniques
20.
Proc Natl Acad Sci U S A ; 105(33): 12057-62, 2008 Aug 19.
Article in English | MEDLINE | ID: mdl-18701716

ABSTRACT

In pancreatic beta-cells, uncoupling protein 2 (UCP2) influences mitochondrial oxidative phosphorylation and insulin secretion. Here, we show that alpha-cells express significantly higher levels of UCP2 than do beta-cells. Greater mitochondrial UCP2-related uncoupling was observed in alpha-cells compared with beta-cells and was accompanied by a lower oxidative phosphorylation efficiency (ATP/O). Conversely, reducing UCP2 activity in alpha-cells was associated with higher mitochondrial membrane potential generated by glucose oxidation and with increased ATP synthesis, indicating more efficient metabolic coupling. In vitro, the suppression of UCP2 activity led to reduced glucagon secretion in response to low glucose; however, in vivo, fasting glucagon levels were normal in UCP2(-/-) mice. In addition to its effects on secretion, UCP2 played a cytoprotective role in islets, with UCP2(-/-) alpha-cells being more sensitive to specific death stimuli. In summary, we demonstrate a direct role for UCP2 in maintaining alpha-cell function at the level of glucose metabolism, glucagon secretion, and cytoprotection.


Subject(s)
Glucagon-Secreting Cells/metabolism , Ion Channels/metabolism , Mitochondrial Proteins/metabolism , Adenosine Triphosphate/biosynthesis , Animals , Cell Line , Cell Survival , Glucagon/metabolism , Glucagon-Secreting Cells/cytology , Glucagon-Secreting Cells/drug effects , Insulin-Secreting Cells/metabolism , Ion Channels/deficiency , Ion Channels/genetics , Membrane Potential, Mitochondrial/drug effects , Mice , Mice, Knockout , Mitochondrial Proteins/deficiency , Mitochondrial Proteins/genetics , RNA, Small Interfering/genetics , Uncoupling Protein 2
SELECTION OF CITATIONS
SEARCH DETAIL
...