Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 55
Filter
1.
Arterioscler Thromb Vasc Biol ; 41(12): 3005-3014, 2021 12.
Article in English | MEDLINE | ID: mdl-34706556

ABSTRACT

OBJECTIVE: Functional HDL (high-density lipoprotein) particles that facilitate cholesterol efflux may be cardioprotective. EL (endothelial lipase) hydrolyzes phospholipids promoting catabolism of HDL and subsequent renal excretion. MEDI5884 is a selective, humanized, monoclonal, EL-neutralizing antibody. We sought to determine the safety, pharmacokinetics, and pharmacodynamic effects of multiple doses of MEDI5884 in patients with stable coronary artery disease. Approach and Results: LEGACY was a phase 2a, double-blind, placebo-controlled, parallel-design trial that randomized 132 patients with stable coronary artery disease receiving high-intensity statin therapy to 3 monthly doses of 1 of 5 dose levels of MEDI5884 (50, 100, 200, 350, or 500 mg SC) or matching placebo. The primary end point was the safety and tolerability of MEDI5884 through the end of the study (day 151). Additional end points included change in HDL cholesterol and cholesterol efflux from baseline to day 91, hepatic uptake of cholesterol at day 91, changes in various other lipid parameters. The incidence of adverse events was similar between the placebo and MEDI5884 groups. In a dose-dependent manner, MEDI5884 increased HDL cholesterol up to 51.4% (P<0.0001) and global cholesterol efflux up to 26.2% ([95% CI, 14.3-38.0] P<0.0001). MEDI5884 increased HDL particle number up to 14.4%. At the highest dose tested, an increase in LDL (low-density lipoprotein) cholesterol up to 28.7% (P<0.0001) and apoB (apolipoprotein B) up to 13.1% (P=0.04) was observed with MEDI5884. However, at the potential target doses for future studies, there was no meaningful increase in LDL cholesterol or apoB. CONCLUSIONS: Inhibition of EL by MEDI5884 increases the quantity and quality of functional HDL in patients with stable coronary artery disease on high-intensity statin therapy without an adverse safety signal at the likely dose to be used. These data support further clinical investigation. Registration: URL: https://www.clinicaltrials.gov; Unique identifier: NCT03351738.


Subject(s)
Coronary Artery Disease/drug therapy , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacokinetics , Lipase/antagonists & inhibitors , Aged , Biomarkers/blood , Cholesterol, HDL/blood , Cholesterol, LDL/blood , Coronary Artery Disease/metabolism , Dose-Response Relationship, Drug , Double-Blind Method , Female , Follow-Up Studies , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/administration & dosage , Lipase/immunology , Male , Middle Aged , Retrospective Studies
2.
Mol Cell Proteomics ; 19(6): 971-993, 2020 06.
Article in English | MEDLINE | ID: mdl-32265294

ABSTRACT

The onset of obesity-linked type 2 diabetes (T2D) is marked by an eventual failure in pancreatic ß-cell function and mass that is no longer able to compensate for the inherent insulin resistance and increased metabolic load intrinsic to obesity. However, in a commonly used model of T2D, the db/db mouse, ß-cells have an inbuilt adaptive flexibility enabling them to effectively adjust insulin production rates relative to the metabolic demand. Pancreatic ß-cells from these animals have markedly reduced intracellular insulin stores, yet high rates of (pro)insulin secretion, together with a substantial increase in proinsulin biosynthesis highlighted by expanded rough endoplasmic reticulum and Golgi apparatus. However, when the metabolic overload and/or hyperglycemia is normalized, ß-cells from db/db mice quickly restore their insulin stores and normalize secretory function. This demonstrates the ß-cell's adaptive flexibility and indicates that therapeutic approaches applied to encourage ß-cell rest are capable of restoring endogenous ß-cell function. However, mechanisms that regulate ß-cell adaptive flexibility are essentially unknown. To gain deeper mechanistic insight into the molecular events underlying ß-cell adaptive flexibility in db/db ß-cells, we conducted a combined proteomic and post-translational modification specific proteomic (PTMomics) approach on islets from db/db mice and wild-type controls (WT) with or without prior exposure to normal glucose levels. We identified differential modifications of proteins involved in redox homeostasis, protein refolding, K48-linked deubiquitination, mRNA/protein export, focal adhesion, ERK1/2 signaling, and renin-angiotensin-aldosterone signaling, as well as sialyltransferase activity, associated with ß-cell adaptive flexibility. These proteins are all related to proinsulin biosynthesis and processing, maturation of insulin secretory granules, and vesicular trafficking-core pathways involved in the adaptation of insulin production to meet metabolic demand. Collectively, this study outlines a novel and comprehensive global PTMome signaling map that highlights important molecular mechanisms related to the adaptive flexibility of ß-cell function, providing improved insight into disease pathogenesis of T2D.


Subject(s)
Diabetes Mellitus, Type 2/metabolism , Hyperglycemia/metabolism , Insulin-Secreting Cells/metabolism , Insulin/metabolism , Obesity/metabolism , Proinsulin/biosynthesis , Proteome/metabolism , Animals , Blood Glucose/metabolism , Diabetes Mellitus, Type 2/genetics , Disease Models, Animal , Focal Adhesions , Gene Ontology , Glucose/metabolism , Hyperglycemia/genetics , Insulin Secretion , Insulin-Secreting Cells/pathology , MAP Kinase Signaling System/genetics , Mice , Mice, Inbred C57BL , Obesity/genetics , Phosphoproteins/genetics , Phosphoproteins/metabolism , Proinsulin/metabolism , Protein Interaction Maps , Protein Processing, Post-Translational , Proteome/genetics , Proteomics , Renin-Angiotensin System , Sialic Acids/metabolism , Tandem Mass Spectrometry , Ubiquitination
3.
J Proteome Res ; 18(9): 3245-3258, 2019 09 06.
Article in English | MEDLINE | ID: mdl-31317746

ABSTRACT

For the treatment of patients with prediabetes or diabetes, clinical evidence has emerged that ß-cell function can be restored by glucose-lowering therapeutic strategies. However, little is known about the molecular mechanisms underlying this functional adaptive behavior of the pancreatic ß-cell. This study examines the dynamic changes in protein expression and phosphorylation state associated with (pro)insulin production and secretory pathway function mediated by euglycemia to induce ß-cell rest in obese/diabetic db/db islet ß-cells. Unbiased quantitative profiling of the protein expression and phosphorylation events that occur upon ß-cell adaption during the transition from hyperglycemia to euglycemia was assessed in isolated pancreatic islets from obese diabetic db/db and wild-type (WT) mice using quantitative proteomics and phosphoproteomics together with bioinformatics analysis. Dynamic changes in the expression and phosphorylation of proteins associated with pancreatic ß-cell (pro)insulin production and complementary regulated-secretory pathway regulation were observed in obese diabetic db/db islets in a hyperglycemic environment, relative to WT mouse islets in a normal euglycemic environment, that resolved when isolated db/db islets were exposed to euglycemia for 12 h in vitro. By similarly treating WT islets in parallel, the effects of tissue culture could be mostly eliminated and only those changes associated with resolution by euglycemia were assessed. Among such regulated protein phosphorylation-dependent signaling events were those associated with COPII-coated vesicle-dependent ER exit, ER-to-Golgi trafficking, clathrin-coat disassembly, and a particular association for the luminal Golgi protein kinase, FAM20C, in control of distal secretory pathway trafficking, sorting, and granule biogenesis. Protein expression and especially phosphorylation play key roles in the regulation of (pro)insulin production, correlative secretory pathway trafficking, and the restoration of ß-cell secretory capacity in the adaptive functional ß-cell response to metabolic demand, especially that mediated by glucose.


Subject(s)
Calcium-Binding Proteins/genetics , Diabetes Mellitus, Type 2/drug therapy , Extracellular Matrix Proteins/genetics , Prediabetic State/drug therapy , Proteomics , Animals , Blood Glucose/drug effects , COP-Coated Vesicles/genetics , Diabetes Mellitus, Type 2/blood , Disease Models, Animal , Glucose/metabolism , Golgi Apparatus/drug effects , Humans , Hyperglycemia/drug therapy , Hyperglycemia/genetics , Insulin/biosynthesis , Insulin/genetics , Insulin-Secreting Cells/drug effects , Mice , Mice, Inbred NOD , Obesity/drug therapy , Obesity/genetics , Prediabetic State/blood , Protein Transport/drug effects
4.
J Biol Chem ; 292(5): 1925-1933, 2017 02 03.
Article in English | MEDLINE | ID: mdl-27994053

ABSTRACT

Apelin-36 was discovered as the endogenous ligand for the previously orphan receptor APJ. Apelin-36 has been linked to two major types of biological activities: cardiovascular (stimulation of cardiac contractility and suppression of blood pressure) and metabolic (improving glucose homeostasis and lowering body weight). It has been assumed that both of these activities are modulated through APJ. Here, we demonstrate that the metabolic activity of apelin-36 can be separated from canonical APJ activation. We developed a series of apelin-36 variants in which evolutionarily conserved residues were mutated, and evaluated their ability to modulate glucose homeostasis and body weight in chronic mouse models. We found that apelin-36(L28A) retains full metabolic activity, but is 100-fold impaired in its ability to activate APJ. In contrast to its full metabolic activity, apelin-36(L28A) lost the ability to suppress blood pressure in spontaneously hypertensive rats (SHR). We took advantage of these findings to develop a longer-acting variant of apelin-36 that could modulate glucose homeostasis without impacting blood pressure (or activating APJ). Apelin-36-[L28C(30kDa-PEG)] is 10,000-fold less potent than apelin-36 at activating the APJ receptor but retains its ability to significantly lower blood glucose and improve glucose tolerance in diet-induced obese mice. Apelin-36-[L28C(30kDa-PEG)] provides a starting point for the development of diabetes therapeutics that are devoid of the blood pressure effects associated with canonical APJ activation.


Subject(s)
Adipokines/pharmacology , Blood Glucose/metabolism , Body Weight/drug effects , Intercellular Signaling Peptides and Proteins/pharmacology , Receptors, G-Protein-Coupled/metabolism , Signal Transduction/drug effects , Animals , Apelin , Apelin Receptors , Blood Pressure/drug effects , Mice , Rats , Rats, Inbred SHR
5.
Hepatology ; 65(3): 950-968, 2017 03.
Article in English | MEDLINE | ID: mdl-27880981

ABSTRACT

Because nonalcoholic steatohepatitis (NASH) is associated with impaired liver regeneration, we investigated the effects of G49, a dual glucagon-like peptide-1/glucagon receptor agonist, on NASH and hepatic regeneration. C57Bl/6 mice fed chow or a methionine and choline-deficient (MCD) diet for 1 week were divided into 4 groups: control (chow diet), MCD diet, chow diet plus G49, and M+G49 (MCD diet plus G49). Mice fed a high-fat diet (HFD) for 10 weeks were divided into groups: HFD and H+G49 (HFD plus G49). Following 2 (MCD groups) or 3 (HFD groups) weeks of treatment with G49, partial hepatectomy (PH) was performed, and all mice were maintained on the same treatment schedule for 2 additional weeks. Analysis of liver function, hepatic regeneration, and comprehensive genomic and metabolic profiling were conducted. NASH was ameliorated in the M+G49 group, manifested by reduced inflammation, steatosis, oxidative stress, and apoptosis and increased mitochondrial biogenesis. G49 treatment was also associated with replenishment of intrahepatic glucose due to enhanced gluconeogenesis and reduced glucose use through the pentose phosphate cycle and oxidative metabolism. Following PH, G49 treatment increased survival, restored the cytokine-mediated priming phase, and enhanced the proliferative capacity and hepatic regeneration ratio in mice on the MCD diet. NASH markers remained decreased in M+G49 mice after PH, and glucose use was shifted to the pentose phosphate cycle and oxidative metabolism. G49 administered immediately after PH was also effective at alleviating the pathological changes induced by the MCD diet. Benefits in terms of liver regeneration were also found in mice fed HFD and treated with G49. CONCLUSION: Dual-acting glucagon-like peptide-1/glucagon receptor agonists such as G49 represent a novel therapeutic approach for patients with NASH and particularly those requiring PH. (Hepatology 2017;65:950-968).


Subject(s)
Glucagon-Like Peptide 1/antagonists & inhibitors , Liver Regeneration/drug effects , Non-alcoholic Fatty Liver Disease/drug therapy , Receptors, Glucagon/antagonists & inhibitors , Animals , Biopsy, Needle , Disease Models, Animal , Glucagon-Like Peptide 1/pharmacology , Humans , Immunohistochemistry , Lipid Peroxidation , Liver Regeneration/physiology , Male , Mice , Mice, Inbred C57BL , Non-alcoholic Fatty Liver Disease/pathology , Oxidative Stress , Random Allocation , Receptors, Glucagon/administration & dosage , Treatment Outcome
6.
J Biol Chem ; 287(33): 27407-14, 2012 Aug 10.
Article in English | MEDLINE | ID: mdl-22740691

ABSTRACT

Recent studies suggested that in old mice, beta cells lose their regenerative potential and cannot respond to mitogenic triggers. These studies examined beta cell replication in aged mice under basal conditions and in response to specific stimuli including treatment with the glucagon-like peptide-1 analog exenatide, streptozotocin injection, partial pancreatectomy, and high fat diet. However, it remains possible that the ability to mount a compensatory response of beta cells is retained in old age, but depends on the specific stimulus. Here, we asked whether partial ablation of beta cells in transgenic mice, using doxycycline-inducible expression of diphtheria toxin, triggers a significant compensatory proliferative response in 1-2-year-old animals. Consistent with previous reports, the basal rate of beta cell replication declines dramatically with age, averaging 0.1% in 2-year-old mice. Transient expression of diphtheria toxin in beta cells of old mice resulted in impaired glucose homeostasis and disruption of islet architecture (ratio of beta to alpha cells). Strikingly, the replication rate of surviving beta cells increased 3-fold over basal rate, similarly to the -fold increase in replication rate of beta cells in young transgenic mice. Islet architecture and glucose tolerance slowly normalized, indicating functional significance of compensatory beta cell replication in this setting. Finally, administration of a small molecule glucokinase activator to old mice doubled the frequency of beta cell replication, further showing that old beta cells can respond to the mitogenic trigger of enhanced glycolysis. We conclude that the potential for functionally significant compensatory proliferation of beta cells is retained in old mice, despite a decline in basal replication rate.


Subject(s)
Aging/physiology , Cell Proliferation , Glucose/metabolism , Insulin-Secreting Cells/metabolism , Animals , Diphtheria Toxin/biosynthesis , Diphtheria Toxin/genetics , Enzyme Activators/pharmacology , Gene Expression , Glucokinase/genetics , Glucokinase/metabolism , Glucose/genetics , Glucose Tolerance Test , Insulin-Secreting Cells/cytology , Mice , Mice, Transgenic , Transgenes
7.
Development ; 137(19): 3205-13, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20823063

ABSTRACT

Pancreatic beta cell proliferation has emerged as the principal mechanism for homeostatic maintenance of beta cell mass during adult life. This underscores the importance of understanding the mechanisms of beta cell replication and suggests novel approaches for regenerative therapy to treat diabetes. Here we use an in vivo pulse-chase labeling assay to investigate the replication dynamics of adult mouse beta cells. We find that replicated beta cells are able to re-enter the cell division cycle shortly after mitosis and regain their normal proliferative potential after a short quiescence period of several days. This quiescence period is lengthened with advanced age, but shortened during injury-driven beta cell regeneration and following treatment with a pharmacological activator of glucokinase, providing strong evidence that metabolic demand is a key determinant of cell cycle re-entry. Lastly, we show that cyclin D2, a crucial factor in beta cell replication, is downregulated during cell division, and is slowly upregulated post-mitosis by a glucose-sensitive mechanism. These results demonstrate that beta cells quickly regain their capacity to re-enter the cell cycle post-mitosis and implicate glucose control of cyclin D2 expression in the regulation of this process.


Subject(s)
Cellular Senescence , Glucose/metabolism , Insulin-Secreting Cells/metabolism , Aging , Animals , Cell Cycle , Cell Proliferation , Cyclin D2/metabolism , Insulin-Secreting Cells/cytology , Mice
8.
Pharmaceutics ; 15(10)2023 Oct 09.
Article in English | MEDLINE | ID: mdl-37896196

ABSTRACT

Oral delivery of peptides and biological molecules promises significant benefits to patients as an alternative to daily injections, but the development of these formulations is challenging due to their low bioavailability and high pharmacokinetic variability. Our earlier work focused on the discovery of MEDI7219, a stabilized, lipidated, glucagon-like peptide 1 agonist peptide, and the selection of sodium chenodeoxycholate (Na CDC) and propyl gallate (PG) as permeation enhancer combinations. We hereby describe the development of the MEDI7219 tablet formulations and composition optimization via in vivo studies in dogs. We designed the MEDI7219 immediate-release tablets with the permeation enhancers Na CDC and PG. Immediate-release tablets were coated with an enteric coating that dissolves at pH ≥ 5.5 to target the upper duodenal region of the gastrointestinal tract and sustained-release tablets with a Carbopol bioadhesive polymer were coated with an enteric coating that dissolves at pH ≥ 7.0 to provide a longer presence at the absorption site in the gastrointestinal tract. In addition to immediate- and enteric-coated formulations, we also tested a proprietary delayed release erodible barrier layer tablet (OralogiKTM) to deliver the payload to the target site in the gastrointestinal tract. The design of tablet dosage forms based on the optimization of formulations resulted in up to 10.1% absolute oral bioavailability in dogs with variability as low as 26% for MEDI7219, paving the way for its clinical development.

9.
Diabetes Care ; 46(5): 985-992, 2023 05 01.
Article in English | MEDLINE | ID: mdl-36809463

ABSTRACT

OBJECTIVE: The association between FTO rs9939609 and obesity is modified by physical activity (PA) and/or insulin sensitivity (IS). We aimed to assess whether these modifications are independent, to assess whether PA and/or IS modify the association between rs9939609 and cardiometabolic traits, and to elucidate underlying mechanisms. RESEARCH DESIGN AND METHODS: Genetic association analyses comprised up to 19,585 individuals. PA was self-reported, and IS was defined based on inverted HOMA insulin resistance index. Functional analyses were performed in muscle biopsies from 140 men and in cultured muscle cells. RESULTS: The BMI-increasing effect of the FTO rs9939609 A allele was attenuated by 47% with high PA (ß [SE], -0.32 [0.10] kg/m2, P = 0.0013) and by 51% with high IS (-0.31 [0.09] kg/m2, P = 0.00028). Interestingly, these interactions were essentially independent (PA, -0.20 [0.09] kg/m2, P = 0.023; IS, -0.28 [0.09] kg/m2, P = 0.0011). The rs9939609 A allele was also associated with higher all-cause mortality and certain cardiometabolic outcomes (hazard ratio, 1.07-1.20, P > 0.04), and these effects tended to be weakened by greater PA and IS. Moreover, the rs9939609 A allele was associated with higher expression of FTO in skeletal muscle tissue (0.03 [0.01], P = 0.011), and in skeletal muscle cells, we identified a physical interaction between the FTO promoter and an enhancer region encompassing rs9939609. CONCLUSIONS: Greater PA and IS independently reduced the effect of rs9939609 on obesity. These effects might be mediated through altered expression of FTO in skeletal muscle. Our results indicated that PA and/or other means of increasing insulin sensitivity could counteract FTO-related genetic predisposition to obesity.


Subject(s)
Cardiovascular Diseases , Hyperinsulinism , Insulin Resistance , Male , Humans , Insulin Resistance/genetics , Body Mass Index , Obesity/genetics , Obesity/metabolism , Exercise , Genetic Predisposition to Disease , Insulin/genetics , Insulin, Regular, Human , Polymorphism, Single Nucleotide , Genotype , Alpha-Ketoglutarate-Dependent Dioxygenase FTO/genetics
10.
J Am Heart Assoc ; 12(3): e8145, 2023 02 07.
Article in English | MEDLINE | ID: mdl-36695299

ABSTRACT

Background Lectin-like oxidized low-density lipoprotein (ox-LDL) receptor-1 is a scavenger receptor for oxidized low-density lipoprotein. In adults, higher soluble lectin-like ox-LDL receptor-1 (sLOX-1) levels are associated with cardiovascular disease, type 2 diabetes, and obesity, but a similar link in pediatric overweight/obesity remains uncertain. Methods and Results Analyses were based on the cross-sectional HOLBAEK Study, including 4- to 19-year-olds from an obesity clinic group with body mass index >90th percentile (n=1815) and from a population-based group (n=2039). Fasting plasma levels of sLOX-1 and inflammatory markers were quantified, cardiometabolic risk profiles were assessed, and linear and logistic regression analyses were performed. Pubertal/postpubertal children and adolescents from the obesity clinic group exhibited higher sLOX-1 levels compared with the population (P<0.001). sLOX-1 positively associated with proinflammatory cytokines, matrix metalloproteinases, body mass index SD score, waist SD score, body fat %, plasma alanine aminotransferase, serum high-sensitivity C-reactive protein, plasma low-density lipoprotein cholesterol, triglycerides, systolic and diastolic blood pressure SD score, and inversely associated with plasma high-density lipoprotein cholesterol (all P<0.05). sLOX-1 positively associated with high alanine aminotransferase (odds ratio [OR], 1.16, P=4.1 E-04), insulin resistance (OR, 1.16, P=8.6 E-04), dyslipidemia (OR, 1.25, P=1.8 E-07), and hypertension (OR, 1.12, P=0.02). Conclusions sLOX-1 levels were elevated during and after puberty in children and adolescents with overweight/obesity compared with population-based peers and associated with inflammatory markers and worsened cardiometabolic risk profiles. sLOX-1 may serve as an early marker of cardiometabolic risk and inflammation in pediatric overweight/obesity. Registration The HOLBAEK Study, formerly known as The Danish Childhood Obesity Biobank, ClinicalTrials.gov identifier number NCT00928473, https://clinicaltrials.gov/ct2/show/NCT00928473 (registered June 2009).


Subject(s)
Cardiovascular Diseases , Diabetes Mellitus, Type 2 , Pediatric Obesity , Scavenger Receptors, Class E , Adolescent , Child , Humans , Alanine Transaminase , Biomarkers , Cholesterol , Cross-Sectional Studies , Inflammation/epidemiology , Lipoproteins, LDL , Overweight/epidemiology , Pediatric Obesity/diagnosis , Pediatric Obesity/epidemiology , Scavenger Receptors, Class E/blood
11.
J Biol Chem ; 286(21): 19118-26, 2011 May 27.
Article in English | MEDLINE | ID: mdl-21454522

ABSTRACT

Type 2 diabetes is a global problem, and current ineffective therapeutic strategies pave the way for novel treatments like small molecular activators targeting glucokinase (GCK). GCK activity is fundamental to beta cell and hepatocyte glucose metabolism, and heterozygous activating and inactivating GCK mutations cause hyperinsulinemic hypoglycemia (HH) and maturity onset diabetes of the young (MODY) respectively. Over 600 naturally occurring inactivating mutations have been reported, whereas only 13 activating mutations are documented to date. We report two novel GCK HH mutations (V389L and T103S) at residues where MODY mutations also occur (V389D and T103I). Using recombinant proteins with in vitro assays, we demonstrated that both HH mutants had a greater relative activity index than wild type (6.0 for V389L, 8.4 for T103S, and 1.0 for wild type). This was driven by an increased affinity for glucose (S(0.5), 3.3 ± 0.1 and 3.5 ± 0.1 mm, respectively) versus wild type (7.5 ± 0.1 mm). Correspondingly, the V389D and T103I MODY mutants had markedly reduced relative activity indexes (<0.1). T103I had an altered affinity for glucose (S(0.5), 24.9 ± 0.6 mm), whereas V389D also exhibited a reduced affinity for ATP and decreased catalysis rate (S(0.5), 78.6 ± 4.5 mm; ATP(K(m)), 1.5 ± 0.1 mm; K(cat), 10.3 ± 1.1s(-1)) compared with wild type (ATP(K(m)), 0.4 ± <0.1; K(cat), 62.9 ± 1.2). Both Thr-103 mutants showed reduced inhibition by the endogenous hepatic inhibitor glucokinase regulatory protein. Molecular modeling demonstrated that Thr-103 maps to the allosteric activator site, whereas Val-389 is located remotely to this position and all other previously reported activating mutations, highlighting α-helix 11 as a novel region regulating GCK activity. Our data suggest that pharmacological manipulation of GCK activity at locations distal from the allosteric activator site is possible.


Subject(s)
Glucokinase , Hyperinsulinism , Hypoglycemia , Liver/enzymology , Mutation, Missense , Allosteric Regulation/genetics , Amino Acid Substitution , Child, Preschool , Diabetes Mellitus, Type 2/enzymology , Diabetes Mellitus, Type 2/genetics , Glucokinase/genetics , Glucokinase/metabolism , Humans , Hyperinsulinism/enzymology , Hyperinsulinism/genetics , Hypoglycemia/enzymology , Hypoglycemia/genetics , Infant, Newborn , Male , Protein Structure, Secondary/genetics
12.
J Biol Chem ; 286(45): 39560-72, 2011 Nov 11.
Article in English | MEDLINE | ID: mdl-21921030

ABSTRACT

We performed genome-wide mutagenesis in C57BL/6J mice using N-ethyl-N-nitrosourea to identify mutations causing high blood glucose early in life and to produce new animal models of diabetes. Of a total of 13 new lines confirmed by heritability testing, we identified two semi-dominant pedigrees with novel missense mutations (Gck(K140E) and Gck(P417R)) in the gene encoding glucokinase (Gck), the mammalian glucose sensor that is mutated in human maturity onset diabetes of the young type 2 and the target of emerging anti-hyperglycemic agents that function as glucokinase activators (GKAs). Diabetes phenotype corresponded with genotype (mild-to-severe: Gck(+/+) < Gck(P417R/+), Gck(K140E)(/+) < Gck(P417R/P417R), Gck(P417R/K140E), and Gck(K140E/K140E)) and with the level of expression of GCK in liver. Each mutant was produced as the recombinant enzyme in Escherichia coli, and analysis of k(cat) and tryptophan fluorescence (I(320/360)) during thermal shift unfolding revealed a correlation between thermostability and the severity of hyperglycemia in the whole animal. Disruption of the glucokinase regulatory protein-binding site (GCK(K140E)), but not the ATP binding cassette (GCK(P417R)), prevented inhibition of enzyme activity by glucokinase regulatory protein and corresponded with reduced responsiveness to the GKA drug. Surprisingly, extracts from liver of diabetic GCK mutants inhibited activity of the recombinant enzyme, a property that was also observed in liver extracts from mice with streptozotocin-induced diabetes. These results indicate a relationship between genotype, phenotype, and GKA efficacy. The integration of forward genetic screening and biochemical profiling opens a pathway for preclinical development of mechanism-based diabetes therapies.


Subject(s)
Alkylating Agents/adverse effects , Diabetes Mellitus, Experimental , Enzyme Activators/metabolism , Ethylnitrosourea/adverse effects , Glucokinase , Liver/enzymology , Mutation, Missense , Alkylating Agents/pharmacology , Amino Acid Substitution , Animals , Binding Sites/genetics , Blood Glucose/genetics , Blood Glucose/metabolism , Diabetes Mellitus, Experimental/enzymology , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/pathology , Ethylnitrosourea/pharmacology , Gene Expression Regulation, Enzymologic/drug effects , Gene Expression Regulation, Enzymologic/genetics , Glucokinase/antagonists & inhibitors , Glucokinase/biosynthesis , Glucokinase/genetics , Humans , Hyperglycemia/chemically induced , Hyperglycemia/enzymology , Hyperglycemia/genetics , Liver/pathology , Male , Mice , Mice, Mutant Strains , Organ Specificity , Protein Folding , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
13.
Am J Physiol Endocrinol Metab ; 302(1): E87-E102, 2012 Jan 01.
Article in English | MEDLINE | ID: mdl-21952036

ABSTRACT

It was reported previously that isolated human islets from individuals with type 2 diabetes mellitus (T2DM) show reduced glucose-stimulated insulin release. To assess the possibility that impaired bioenergetics may contribute to this defect, glucose-stimulated respiration (Vo(2)), glucose usage and oxidation, intracellular Ca(2+), and insulin secretion (IS) were measured in pancreatic islets isolated from three healthy and three type 2 diabetic organ donors. Isolated mouse and rat islets were studied for comparison. Islets were exposed to a "staircase" glucose stimulus, whereas IR and Vo(2) were measured. Vo(2) of human islets from normals and diabetics increased sigmoidally from equal baselines of 0.25 nmol/100 islets/min as a function of glucose concentration. Maximal Vo(2) of normal islets at 24 mM glucose was 0.40 ± 0.02 nmol·min(-1)·100 islets(-1), and the glucose S(0.5) was 4.39 ± 0.10 mM. The glucose stimulation of respiration of islets from diabetics was lower, V(max) of 0.32 ± 0.01 nmol·min(-1)·100 islets(-1), and the S(0.5) shifted to 5.43 ± 0.13 mM. Glucose-stimulated IS and the rise of intracellular Ca(2+) were also reduced in diabetic islets. A clinically effective glucokinase activator normalized the defective Vo(2), IR, and free calcium responses during glucose stimulation in islets from type 2 diabetics. The body of data shows that there is a clear relationship between the pancreatic islet energy (ATP) production rate and IS. This relationship was similar for normal human, mouse, and rat islets and the data for all species fitted a single sigmoidal curve. The shared threshold rate for IS was ∼13 pmol·min(-1)·islet(-1). Exendin-4, a GLP-1 analog, shifted the ATP production-IS curve to the left and greatly potentiated IS with an ATP production rate threshold of ∼10 pmol·min(-1)·islet(-1). Our data suggest that impaired ß-cell bioenergetics resulting in greatly reduced ATP production is critical in the molecular pathogenesis of type 2 diabetes mellitus.


Subject(s)
Benzeneacetamides/pharmacology , Diabetes Mellitus, Type 2/metabolism , Enzyme Activators/pharmacology , Glucokinase/metabolism , Glucose/metabolism , Insulin/metabolism , Islets of Langerhans/drug effects , Adult , Animals , Calcium Signaling/drug effects , Cell Respiration/drug effects , Diabetes Mellitus, Type 2/drug therapy , Exenatide , Female , Glucagon-Like Peptide 1/analogs & derivatives , Glucokinase/chemistry , Glycolysis/drug effects , Humans , Hypoglycemic Agents/pharmacology , Insulin Secretion , Islets of Langerhans/metabolism , Male , Mice , Middle Aged , Oxidative Phosphorylation/drug effects , Peptides/pharmacology , Rats , Species Specificity , Tissue Culture Techniques , Venoms/pharmacology
14.
Biochem J ; 440(2): 203-15, 2011 Dec 01.
Article in English | MEDLINE | ID: mdl-21831042

ABSTRACT

GK (glucokinase) is activated by glucose binding to its substrate site, is inhibited by GKRP (GK regulatory protein) and stimulated by GKAs (GK activator drugs). To explore further the mechanisms of these processes we studied pure recombinant human GK (normal enzyme and a selection of 31 mutants) using steady-state kinetics of the enzyme and TF (tryptophan fluorescence). TF studies of the normal binary GK-glucose complex corroborate recent crystallography studies showing that it exists in a closed conformation greatly different from the open conformation of the ligand-free structure, but indistinguishable from the ternary GK-glucose-GKA complex. GKAs did activate and GKRP did inhibit normal GK, whereas its TF was doubled by glucose saturation. However, the enzyme kinetics, GKRP inhibition, TF enhancement by glucose and responsiveness to GKA of the selected mutants varied greatly. Two predominant response patterns were identified accounting for nearly all mutants: (i) GK mutants with a normal or close to normal response to GKA, normally low basal TF (indicating an open conformation), some variability of kinetic parameters (k(cat), glucose S(0.5), h and ATP K(m)), but usually strong GKRP inhibition (13/31); and (ii) GK mutants that are refractory to GKAs, exhibit relatively high basal TF (indicating structural compaction and partial closure), usually show strongly enhanced catalytic activity primarily due to lowering of the glucose S(0.5), but with reduced or no GKRP inhibition in most cases (14/31). These results and those of previous studies are best explained by envisioning a common allosteric regulator region with spatially non-overlapping GKRP- and GKA-binding sites.


Subject(s)
Allosteric Regulation , Glucokinase/metabolism , Carrier Proteins , Fluorescence , Glucokinase/antagonists & inhibitors , Glucokinase/genetics , Glucose/pharmacology , Humans , Kinetics , Point Mutation , Protein Conformation , Tryptophan/chemistry
15.
Skelet Muscle ; 12(1): 26, 2022 12 21.
Article in English | MEDLINE | ID: mdl-36539856

ABSTRACT

BACKGROUND: Sarcopenia is defined as age-related low muscle mass and function, and can also describe the loss of muscle mass in certain medical conditions, such as sarcopenic obesity. Sarcopenic obesity describes loss of muscle and function in obese individuals; however, as sarcopenia is an age-related condition and obesity can occur in any age group, a more accurate term is obesity with low lean muscle mass (OLLMM). Given limited data on OLLMM (particularly in those aged < 65 years), the purpose of this study was to estimate the prevalence of OLLMM in adults aged ≥ 20 years in the USA. METHODS: Data from the National Health and Nutrition Examination Survey (NHANES) 2017-2018 and 1999-2006 were used. OLLMM was defined as an appendicular lean mass, adjusted for body mass index (BMI), cut-off point < 0.789 for males and < 0.512 for females, measured by dual-energy X-ray absorptiometry (DXA). DXA was only measured in individuals 20-59 years old in NHANES 2017-2018; we therefore utilized logistic regression models to predict OLLMM from NHANES 1999-2006 for those aged ≥ 60 years. The prevalence of OLLMM was estimated overall, and by sex, age, race/ethnicity, and clinical subgroup (high BMI, prediabetes, type 2 diabetes mellitus [T2DM], non-alcoholic fatty liver disease [NAFLD] with fibrosis, or post-bariatric surgery). Prevalence estimates were extrapolated to the USA population using NHANES sampling weights. RESULTS: We estimated that, during 2017-2018, 28.7 million or 15.9% of the USA population had OLLMM. The prevalence of OLLMM was greater in older individuals (8.1%, aged 20-59 years vs 28.3%, aged ≥ 60 years), highest (66.6%) in Mexican-American females aged ≥ 60 years, and lowest (2.6%) in non-Hispanic Black males aged 20-59 years. There was a higher prevalence of OLLMM in adults with prediabetes (19.7%), T2DM (34.5%), NAFLD with fibrosis (25.4%), or post-bariatric surgery (21.8%), compared with those without each condition. CONCLUSIONS: Overall, the burden of OLLMM in the USA is substantial, affecting almost 30 million adults. The prevalence of OLLMM increased with age, and among those with prediabetes, T2DM, NAFLD with fibrosis, or post-bariatric surgery. A unified definition of OLLMM will aid diagnosis and treatment strategies.


Subject(s)
Diabetes Mellitus, Type 2 , Non-alcoholic Fatty Liver Disease , Prediabetic State , Sarcopenia , Male , Adult , Female , Humans , Aged , Young Adult , Middle Aged , Sarcopenia/epidemiology , Nutrition Surveys , Non-alcoholic Fatty Liver Disease/complications , Diabetes Mellitus, Type 2/complications , Prevalence , Prediabetic State/complications , Obesity/complications , Obesity/epidemiology , Fibrosis , Muscles , Body Composition
16.
Mol Metab ; 55: 101392, 2022 01.
Article in English | MEDLINE | ID: mdl-34781035

ABSTRACT

OBJECTIVE: Obesity-linked type 2 diabetes (T2D) is a worldwide health concern and many novel approaches are being considered for its treatment and subsequent prevention of serious comorbidities. Co-administration of glucagon like peptide 1 (GLP-1) and peptide YY3-36 (PYY3-36) renders a synergistic decrease in energy intake in obese men. However, mechanistic details of the synergy between these peptide agonists and their effects on metabolic homeostasis remain relatively scarce. METHODS: In this study, we utilized long-acting analogues of GLP-1 and PYY3-36 (via Fc-peptide conjugation) to better characterize the synergistic pharmacological benefits of their co-administration on body weight and glycaemic regulation in obese and diabetic mouse models. Hyperinsulinemic-euglycemic clamps were used to measure weight-independent effects of Fc-PYY3-36 + Fc-GLP-1 on insulin action. Fluorescent light sheet microscopy analysis of whole brain was performed to assess activation of brain regions. RESULTS: Co-administration of long-acting Fc-IgG/peptide conjugates of Fc-GLP-1 and Fc-PYY3-36 (specific for PYY receptor-2 (Y2R)) resulted in profound weight loss, restored glucose homeostasis, and recovered endogenous ß-cell function in two mouse models of obese T2D. Hyperinsulinemic-euglycemic clamps in C57BLKS/J db/db and diet-induced obese Y2R-deficient (Y2RKO) mice indicated Y2R is required for a weight-independent improvement in peripheral insulin sensitivity and enhanced hepatic glycogenesis. Brain cFos staining demonstrated distinct temporal activation of regions of the hypothalamus and hindbrain following Fc-PYY3-36 + Fc-GLP-1R agonist administration. CONCLUSIONS: These results reveal a therapeutic approach for obesity/T2D that improved insulin sensitivity and restored endogenous ß-cell function. These data also highlight the potential association between the gut-brain axis in control of metabolic homeostasis.


Subject(s)
Glucagon-Like Peptide 1/metabolism , Obesity/metabolism , Peptide YY/metabolism , Animals , Blood Glucose/metabolism , Body Weight/drug effects , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Type 2/metabolism , Diet , Eating/drug effects , Energy Intake/drug effects , Energy Metabolism/drug effects , Gastric Bypass , Glucagon-Like Peptide-1 Receptor/metabolism , Hypothalamus , Insulin Resistance/physiology , Insulin-Secreting Cells/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Obese , Obesity/physiopathology , Peptide YY/physiology , Weight Loss
17.
Bioorg Med Chem Lett ; 21(7): 1933-6, 2011 Apr 01.
Article in English | MEDLINE | ID: mdl-21388809

ABSTRACT

The highly potent but modestly selective N-(2-amino-4-methoxy-benzothiazol-7-yl)-N-ethyl-acetamide derivative 2 was selected as the starting point for the design of novel selective A(2B) antagonists, due to its excellent potency, and good drug-like properties. A series of compounds containing nonaromatic amides or ureas of five- or six-membered rings, and also bearing an m-trifluoromethyl-phenyl group (shown to impart superior potency) was prepared and evaluated for their selectivity against the A(2A) and A(1) receptors. This work resulted in the identification of compound 30, with excellent potency and high selectivity against both A(2A) and A(1) receptors.


Subject(s)
Adenosine A2 Receptor Antagonists/chemistry , Adenosine A2 Receptor Antagonists/pharmacology , Benzothiazoles/pharmacology , Drug Discovery , Benzothiazoles/chemistry , Structure-Activity Relationship
18.
Handb Exp Pharmacol ; (203): 357-401, 2011.
Article in English | MEDLINE | ID: mdl-21484579

ABSTRACT

Glucokinase Glucokinase (GK GK ; EC 2.7.1.1.) phosphorylates and regulates glucose metabolism in insulin-producing pancreatic beta-cells, hepatocytes, and certain cells of the endocrine and nervous systems allowing it to play a central role in glucose homeostasis glucose homeostasis . Most importantly, it serves as glucose sensor glucose sensor in pancreatic beta-cells mediating glucose-stimulated insulin biosynthesis and release and it governs the capacity of the liver to convert glucose to glycogen. Activating and inactivating mutations of the glucokinase gene cause autosomal dominant hyperinsulinemic hypoglycemia and hypoinsulinemic hyperglycemia in humans, respectively, illustrating the preeminent role of glucokinase in the regulation of blood glucose and also identifying the enzyme as a potential target for developing antidiabetic drugs antidiabetic drugs . Small molecules called glucokinase activators (GKAs) glucokinase activators (GKAs) which bind to an allosteric activator allosteric activator site of the enzyme have indeed been discovered and hold great promise as new antidiabetic agents. GKAs increase the enzyme's affinity for glucose and also its maximal catalytic rate. Consequently, they stimulate insulin biosynthesis and secretion, enhance hepatic glucose uptake, and augment glucose metabolism and related processes in other glucokinase-expressing cells. Manifestations of these effects, most prominently a lowering of blood glucose, are observed in normal laboratory animals and man but also in animal models of diabetes and patients with type 2 diabetes mellitus (T2DM T2DM ) type 2 diabetes mellitus (T2DM) . These compelling concepts and results sustain a strong R&D effort by many pharmaceutical companies to generate GKAs with characteristics allowing for a novel drug treatment of T2DM.


Subject(s)
Diabetes Mellitus/drug therapy , Enzyme Activators/pharmacology , Glucokinase/metabolism , Hypoglycemic Agents/therapeutic use , Animals , Diabetes Mellitus, Type 2/drug therapy , Enzyme Activators/therapeutic use , Homeostasis/drug effects , Humans , Hyperinsulinism/drug therapy , Hypoglycemic Agents/pharmacology , Receptors, Drug/drug effects
19.
PLoS One ; 16(6): e0252426, 2021.
Article in English | MEDLINE | ID: mdl-34086718

ABSTRACT

Quantification of endogenous biomarkers in clinical studies requires careful evaluation of a number of assay performance parameters. Comparisons of absolute values from several clinical studies can enable retrospective analyses further elucidating the biology of a given biomarker across various study populations. We characterized the performance of a highly multiplex bioanalytical method for quantification of phosphatidylinositols (PI). Hydrophilic interaction chromatography (HILIC) and multiple reaction monitoring (MRM) were employed for targeted multiplex quantification. Odd-chain PI species that are not normally present in human plasma were utilized as surrogate analytes (SA) to assess various assay performance parameters and establish a definitive dynamic linear range for PI lipids. To correct for batch effects, Systematic Error Removal using Random Forest (SERRF) normalization algorithm was employed and used to bridge raw values between two clinical studies, enabling quantitative comparison of their absolute values. A high throughput method was developed, qualified, transferred to an automation platform and applied to sample testing in two clinical trials in healthy volunteers (NCT03001297) and stable Coronary Artery Disease (CAD, NCT03351738) subjects. The method demonstrated acceptable precision and accuracy (±30%) over linear range of 1-1000 nM for SA and 8-fold dilutional linearity for endogenous PI. We determined that mean-adjusted average QC performed best for normalization using SERRF. The comparison of two studies revealed that healthy subject levels of PI are consistently higher across PI species compared to CAD subjects identifying a potential lipid biomarker to be explored in future studies.


Subject(s)
Coronary Artery Disease/blood , Phosphatidylinositols/blood , Chromatography, High Pressure Liquid/methods , Clinical Trials as Topic , Data Interpretation, Statistical , Humans , Software
20.
Sci Rep ; 11(1): 22521, 2021 11 18.
Article in English | MEDLINE | ID: mdl-34795324

ABSTRACT

Peptide therapeutics are increasingly used in the treatment of disease, but their administration by injection reduces patient compliance and convenience, especially for chronic diseases. Thus, oral administration of a peptide therapeutic represents a significant advance in medicine, but is challenged by gastrointestinal instability and ineffective uptake into the circulation. Here, we have used glucagon-like peptide-1 (GLP-1) as a model peptide therapeutic for treating obesity-linked type 2 diabetes, a common chronic disease. We describe a comprehensive multidisciplinary approach leading to the development of MEDI7219, a GLP-1 receptor agonist (GLP-1RA) specifically engineered for oral delivery. Sites of protease/peptidase vulnerabilities in GLP-1 were removed by amino acid substitution and the peptide backbone was bis-lipidated to promote MEDI7219 reversible plasma protein binding without affecting potency. A combination of sodium chenodeoxycholate and propyl gallate was used to enhance bioavailability of MEDI7219 at the site of maximal gastrointestinal absorption, targeted by enteric-coated tablets. This synergistic approach resulted in MEDI7219 bioavailability of ~ 6% in dogs receiving oral tablets. In a dog model of obesity and insulin resistance, MEDI7219 oral tablets significantly decreased food intake, body weight and glucose excursions, validating the approach. This novel approach to the development of MEDI7219 provides a template for the development of other oral peptide therapeutics.


Subject(s)
Chronic Disease , Drug Delivery Systems , Glucagon-Like Peptide-1 Receptor , Peptides , Protein Engineering , Animals , Cricetinae , Humans , Male , Mice , Administration, Oral , Caco-2 Cells , Chemistry, Pharmaceutical/methods , Chenodeoxycholic Acid/administration & dosage , CHO Cells , Chronic Disease/drug therapy , Cricetulus , Diabetes Mellitus, Type 2/drug therapy , Drug Discovery , Glucagon-Like Peptide-1 Receptor/agonists , Insulin-Secreting Cells/cytology , Mice, Inbred C57BL , Peptides/chemistry , Propyl Gallate/administration & dosage , Protein Engineering/methods , Receptors, Glucagon/agonists , Tablets, Enteric-Coated
SELECTION OF CITATIONS
SEARCH DETAIL