Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 127
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Clin Proteomics ; 19(1): 16, 2022 May 19.
Article in English | MEDLINE | ID: mdl-35590248

ABSTRACT

BACKGROUND: Glucagon serves as an important regulatory hormone for regulating blood glucose concentration with tight feedback control exerted by insulin and glucose. There are critical gaps in our understanding of glucagon kinetics, pancreatic α cell function and intra-islet feedback network that are disrupted in type 1 diabetes. This is important for translational research applications of evolving dual-hormone (insulin + glucagon) closed-loop artificial pancreas algorithms and their usage in type 1 diabetes. Thus, it is important to accurately measure glucagon kinetics in vivo and to develop robust models of glucose-insulin-glucagon interplay that could inform next generation of artificial pancreas algorithms. METHODS: Here, we describe the administration of novel 13C15N heavy isotope-containing glucagon tracers-FF glucagon [(Phe 6 13C9,15N; Phe 22 13C9,15N)] and FFLA glucagon [(Phe 6 13C9,15N; Phe 22 13C9,15N; Leu 14 13C6,15N; Ala 19 13C3)] followed by anti-glucagon antibody-based enrichment and LC-MS/MS based-targeted assays using high-resolution mass spectrometry to determine levels of infused glucagon in plasma samples. The optimized assay results were applied for measurement of glucagon turnover in subjects with and without type 1 diabetes infused with isotopically labeled glucagon tracers. RESULTS: The limit of quantitation was found to be 1.56 pg/ml using stable isotope-labeled glucagon as an internal standard. Intra and inter-assay variability was < 6% and < 16%, respectively, for FF glucagon while it was < 5% and < 23%, respectively, for FFLA glucagon. Further, we carried out a novel isotope dilution technique using glucagon tracers for studying glucagon kinetics in type 1 diabetes. CONCLUSIONS: The methods described in this study for simultaneous detection and quantitation of glucagon tracers have clinical utility for investigating glucagon kinetics in vivo in humans.

2.
Mol Pharm ; 19(12): 4612-4624, 2022 12 05.
Article in English | MEDLINE | ID: mdl-36106748

ABSTRACT

Diabetes mellitus (DM), a multifaceted metabolic disorder if not managed properly leads to secondary complications. Diabetic peripheral neuropathy (DPN) is one such complication caused by nerve damage that cannot be reversed but can be delayed. Recently, diabetes patients are using dietary supplements, although there remains a general skepticism about this practice. Curcumin (CUR), one such supplement can help prevent underlying low-grade inflammation in diabetes, but it is plagued by poor oral bioavailability. To better understand the role of bioavailability in clinical outcomes, we have tested double-headed nanosystems containing curcumin (nCUR) on DPN. Because CUR does not influence glucose levels, we have also tested the effects of nCUR combined with long-acting subcutaneous insulin (INS). nCUR with or without INS alleviates DPN at two times lower dose than unformulated CUR, as indicated by qualitative and quantitative analysis of the hind paw, sciatic nerve, spleen, and L4-6 spinal cord. In addition, nCUR and nCUR+INS preserve hind paw nerve axons as evident by the Bielschowsky silver stain and intraepidermal nerve fibers (IENF) density measured by immunofluorescence. The mechanistic studies further corroborated the results, where nCUR or nCUR+INS showed a significant decrease in TUNEL positive cells, mRNA expression of NLRP3, IL-1ß, and macrophage infiltration while preserving nestin and NF200 expression in the sciatic nerve. Together, the data confirms that CUR bioavailability is proportional to clinical outcomes and INS alone may not be one of the solutions for DM. This study highlights the potential of nCUR with or without INS in alleviating DPN and warrants further investigation.


Subject(s)
Curcumin , Diabetes Mellitus, Experimental , Diabetic Neuropathies , Animals , Rats , Curcumin/therapeutic use , Diabetes Mellitus, Experimental/metabolism , Diabetic Neuropathies/drug therapy , Diabetic Neuropathies/metabolism , Insulin , Insulin, Regular, Human , Rats, Sprague-Dawley
3.
Diabetes Obes Metab ; 24(5): 881-890, 2022 05.
Article in English | MEDLINE | ID: mdl-35014156

ABSTRACT

AIM: To evaluate whether short-term treatment with a selective 11ß-Hydroxysteroid dehydrogenase-1 (11ß-HSD1) inhibitor, AZD4017, would block hepatic cortisol production and thereby decrease hepatic fat in patients with nonalcoholic fatty liver disease (NAFLD)/nonalcoholic steatohepatitis (NASH), with or without type 2 diabetes (T2D). MATERIALS AND METHODS: This was a randomized, double-blind, placebo-controlled, phase 2 study conducted at two sites. Key inclusion criteria were the presence of NAFLD or NASH on magnetic resonance imaging (MRI) or recent biopsy positive for NASH. Enrolled patients were randomly assigned (1:1) to AZD4017 or placebo for 12 weeks. Primary outcomes were between-group differences in mean change from baseline to week 12 in liver fat fraction (LFF) and conversion of 13 C cortisone to 13 C cortisol in the liver. RESULTS: A total of 93 patients were randomized; 85 patients completed treatment. The mean (standard deviation [SD]) change in LFF was -0.667 (5.246) and 0.139 (4.323) in the AZD4017 and placebo groups (P = 0.441). For patients with NASH and T2D, the mean (SD) change in LFF was significantly improved in the AZD4017 versus the placebo group (-1.087 [5.374] vs. 1.675 [3.318]; P = 0.033). Conversion of 13 C cortisone to 13 C cortisol was blocked in all patients in the AZD4017 group. There were no significant between-group differences (AZD4017 vs. placebo) in changes in fibrosis, weight, levels of liver enzymes or lipids, or insulin sensitivity. CONCLUSION: Although the study did not meet one of the primary outcomes, AZD4017 blocked the conversion of 13 C cortisone to 13 C cortisol in the liver in all patients who received the drug. In patients with NASH and T2D, AZD4017 improved liver steatosis versus placebo.


Subject(s)
Diabetes Mellitus, Type 2 , Non-alcoholic Fatty Liver Disease , 11-beta-Hydroxysteroid Dehydrogenase Type 1 , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/pathology , Double-Blind Method , Humans , Liver/pathology , Niacinamide/analogs & derivatives , Non-alcoholic Fatty Liver Disease/complications , Non-alcoholic Fatty Liver Disease/drug therapy , Non-alcoholic Fatty Liver Disease/pathology , Piperidines/therapeutic use
4.
Endocr Pract ; 28(5): 528-562, 2022 May.
Article in English | MEDLINE | ID: mdl-35569886

ABSTRACT

OBJECTIVE: To provide evidence-based recommendations regarding the diagnosis and management of nonalcoholic fatty liver disease (NAFLD) and nonalcoholic steatohepatitis (NASH) to endocrinologists, primary care clinicians, health care professionals, and other stakeholders. METHODS: The American Association of Clinical Endocrinology conducted literature searches for relevant articles published from January 1, 2010, to November 15, 2021. A task force of medical experts developed evidence-based guideline recommendations based on a review of clinical evidence, expertise, and informal consensus, according to established American Association of Clinical Endocrinology protocol for guideline development. RECOMMENDATION SUMMARY: This guideline includes 34 evidence-based clinical practice recommendations for the diagnosis and management of persons with NAFLD and/or NASH and contains 385 citations that inform the evidence base. CONCLUSION: NAFLD is a major public health problem that will only worsen in the future, as it is closely linked to the epidemics of obesity and type 2 diabetes mellitus. Given this link, endocrinologists and primary care physicians are in an ideal position to identify persons at risk on to prevent the development of cirrhosis and comorbidities. While no U.S. Food and Drug Administration-approved medications to treat NAFLD are currently available, management can include lifestyle changes that promote an energy deficit leading to weight loss; consideration of weight loss medications, particularly glucagon-like peptide-1 receptor agonists; and bariatric surgery, for persons who have obesity, as well as some diabetes medications, such as pioglitazone and glucagon-like peptide-1 receptor agonists, for those with type 2 diabetes mellitus and NASH. Management should also promote cardiometabolic health and reduce the increased cardiovascular risk associated with this complex disease.


Subject(s)
Diabetes Mellitus, Type 2 , Non-alcoholic Fatty Liver Disease , Diabetes Mellitus, Type 2/drug therapy , Glucagon-Like Peptide-1 Receptor , Humans , Non-alcoholic Fatty Liver Disease/complications , Non-alcoholic Fatty Liver Disease/diagnosis , Non-alcoholic Fatty Liver Disease/therapy , Obesity/complications , Primary Health Care , United States/epidemiology , Weight Loss
5.
Am J Physiol Endocrinol Metab ; 321(1): E122-E129, 2021 07 01.
Article in English | MEDLINE | ID: mdl-33998292

ABSTRACT

Exercise effects (EE) on whole body glucose rate of disappearance (Rd) occur through insulin-independent (IIRd) and insulin-dependent (IDRd) mechanisms. Quantifying these processes in vivo would allow a better understanding of the physiology of glucose regulation. This is of particular importance in individuals with type 1 diabetes (T1D) since such a knowledge may help to improve glucose management. However, such a model is still lacking. Here, we analyzed data from six T1D and six nondiabetic (ND) subjects undergoing a labeled glucose clamp study during, before, and after a 60-min exercise session at 65% V̇o2max on three randomized visits: euglycemia-low insulin, euglycemia-high insulin, and hyperglycemia-low insulin. We tested a set of models, all sharing a single-compartment description of glucose kinetics, but differing in how exercise is assumed to modulate glucose disposal. Model selection was based on parsimony criteria. The best model assumed an exercise-induced immediate effect on IIRd and a delayed effect on IDRd. It predicted that exercise increases IIRd, compared with rest, by 66%-82% and 67%-97% in T1D and ND, respectively, not significantly different between the two groups. Conversely, the exercise effect on IDRd ranged between 81% and 155% in T1D and it was significantly higher than ND, which ranged between 10% and 40%. The exaggerated effect observed in IDRd can explain the higher hypoglycemia risk related to individuals with T1D. This novel exercise model could help in informing safe and effective glucose management during and after exercise in individuals with T1D.NEW & NOTEWORTHY Here, we present a new mathematical model describing the effect of moderate physical activity on insulin-mediated and noninsulin-mediated glucose disposal in subjects with and without diabetes. We believe that this represents a step-forward in the knowledge of type 1 diabetes pathophysiology, and an useful tool to design safe and effective insulin-therapies.


Subject(s)
Blood Glucose/metabolism , Diabetes Mellitus, Type 1/physiopathology , Exercise/physiology , Insulin/administration & dosage , Adolescent , Adult , Blood Glucose/analysis , Diabetes Mellitus, Type 1/blood , Diabetes Mellitus, Type 1/drug therapy , Glucose Clamp Technique , Glycemic Control/methods , Homeostasis/physiology , Humans , Insulin/blood , Models, Theoretical , Oxygen Consumption , Young Adult
6.
Clin Gastroenterol Hepatol ; 19(1): 146-154.e4, 2021 01.
Article in English | MEDLINE | ID: mdl-32360804

ABSTRACT

BACKGROUND & AIMS: Obese patients with nonalcoholic steatohepatitis (NASH) are at risk for cirrhosis if significant weight loss is not achieved. The single fluid-filled intragastric balloon (IGB) induces meaningful weight loss and might be used in NASH treatment. We performed an open-label prospective study to evaluate the effects of IGB placement on metabolic and histologic features of NASH. METHODS: Twenty-one patients with early hepatic fibrosis (81% female; mean age, 54 years; average body mass index, 44 kg/m2) underwent magnetic resonance elastography (MRE) and endoscopic ultrasound with core liver biopsy collection at time IGB placement and removal at a single center from October 2016 through March 2018. The primary outcome measure was the changes in liver histology parameters after IGB, including change in nonalcoholic fatty liver disease activity score (NAS) and fibrosis score. We also evaluated changes in weight, body mass index, waist to hip ratio, aminotransaminases, fasting levels of lipids, fasting glucose, glycosylated hemoglobin, and MRE-detected liver stiffness. RESULTS: Six months after IGB, patients' mean total body weight loss was 11.7% ± 7.7%, with significant reductions in HbA1c (1.3% ± 0.5%) (P = .02). Waist circumference decreased by 14.4 ± 2.2 cm (P = .001). NAS improved in 18 of 20 patients (90%), with a median decrease of 3 points (range, 1-4 points); 16 of 20 patients (80%) had improvements of 2 points or more. Fibrosis improved by 1.17 stages in 15% of patients, and MRE-detected fibrosis improved by 1.5 stages in 10 of 20 patients (50%). Half of patients reached endpoints approved by the Food and Drug Administration of for NASH resolution and fibrosis improvement. Percent total body weight loss did not correlate with reductions in NAS or fibrosis. Other than post-procedural pain (in 5% of patients), no serious adverse events were reported. CONCLUSION: In a prospective study, IGB facilitated significant metabolic and histologic improvements in NASH. IGB appears to be safe and effective for NASH management when combined with a prescribed diet and exercise program. ClinicalTrials.gov no: NCT02880189.


Subject(s)
Gastric Balloon , Non-alcoholic Fatty Liver Disease , Female , Gastric Balloon/adverse effects , Humans , Liver , Male , Middle Aged , Non-alcoholic Fatty Liver Disease/therapy , Prospective Studies , Weight Loss
7.
J Pharmacokinet Pharmacodyn ; 48(2): 225-239, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33394220

ABSTRACT

To shed light on how acute exercise affects blood glucose (BG) concentrations in nondiabetic subjects, we develop a physiological pharmacokinetic/pharmacodynamic model of postprandial glucose dynamics during exercise. We unify several concepts of exercise physiology to derive a multiscale model that includes three important effects of exercise on glucose dynamics: increased endogenous glucose production (EGP), increased glucose uptake in skeletal muscle (SM), and increased glucose delivery to SM by capillary recruitment (i.e. an increase in surface area and blood flow in capillary beds). We compare simulations to experimental observations taken in two cohorts of healthy nondiabetic subjects (resting subjects (n = 12) and exercising subjects (n = 12)) who were each given a mixed-meal tolerance test. Metabolic tracers were used to quantify the glucose flux. Simulations reasonably agree with postprandial measurements of BG concentration and EGP during exercise. Exercise-induced capillary recruitment is predicted to increase glucose transport to SM by 100%, causing hypoglycemia. When recruitment is blunted, as in those with capillary dysfunction, the opposite occurs and higher than expected BG levels are predicted. Model simulations show how three important exercise-induced phenomena interact, impacting BG concentrations. This model describes nondiabetic subjects, but it is a first step to a model that describes glucose dynamics during exercise in those with type 1 diabetes (T1D). Clinicians and engineers can use the insights gained from the model simulations to better understand the connection between exercise and glucose dynamics and ultimately help patients with T1D make more informed insulin dosing decisions around exercise.


Subject(s)
Blood Glucose/analysis , Exercise/physiology , Insulin/metabolism , Models, Biological , Adult , Blood Glucose/metabolism , Computer Simulation , Diabetes Mellitus, Type 1/blood , Diabetes Mellitus, Type 1/metabolism , Healthy Volunteers , Humans , Muscle, Skeletal/metabolism
8.
Am J Physiol Endocrinol Metab ; 316(6): E998-E1011, 2019 06 01.
Article in English | MEDLINE | ID: mdl-30860881

ABSTRACT

Quantification of disturbances in glucose-insulin homeostasis has been the cornerstone of appraising insulin resistance and detecting early-stage diabetes. Metabolic homeostasis arises from feedback and feed-forward interactions among (at least) all four of glucose, insulin, glucagon, and cortisol. Quantifying such tetrapartite interactions in the fasting (endogenously regulated) state overnight could elucidate very early regulatory disruption. In the present study, healthy subjects without diabetes (ND; n = 20) and patients with Type 2 diabetes (T2D; n = 21) were investigated by repeated overnight blood sampling of all four of glucose, insulin, glucagon, and cortisol concentrations. To obviate confounding by hormone-specific disappearance rates, analyses were performed at the level of production (glucose) or secretion (insulin, glucagon, and cortisol) rates estimated by regularized deconvolution analysis. Then, a novel method for quantifying the loss of homeostasis among glucose, insulin, and glucagon (and, when available, cortisol) secretion patterns was developed. Potential early stage prediabetic candidates were identified. The new methodology avoids many of the difficulties encountered in the conventional estimation of insulin-glucose sensitivity or resistance, while incorporating the dynamics of the key coregulators under fasting conditions.


Subject(s)
Blood Glucose/metabolism , Diabetes Mellitus, Type 2/metabolism , Glucagon/metabolism , Hydrocortisone/metabolism , Insulin/metabolism , Prediabetic State/metabolism , Case-Control Studies , Homeostasis , Humans , Insulin Resistance , Insulin Secretion
9.
Am J Physiol Endocrinol Metab ; 317(3): E483-E493, 2019 09 01.
Article in English | MEDLINE | ID: mdl-31265327

ABSTRACT

While the triple tracer isotope dilution method has enabled accurate estimation of carbohydrate turnover after a mixed meal, use of the simple carbohydrate glucose as the carbohydrate source limits its translational applicability to everyday meals that typically contain complex carbohydrates. Hence, utilizing the natural enrichment of [13C]polysaccharide in commercially available grains, we devised a novel tracer method to measure postprandial complex carbohydrate turnover and indices of insulin action and ß-cell function and compared the parameters to those obtained after a simple carbohydrate containing mixed meal. We studied healthy volunteers after either rice (n = 8) or sorghum (n = 8) and glucose (n = 16) containing mixed meals and modified the triple tracer technique to calculate carbohydrate turnover. All meals were matched for calories and macronutrient composition. Rates of meal glucose appearance (2,658 ± 736 vs. 4,487 ± 909 µM·kg-1·2 h-1), endogenous glucose production (-835 ± 283 vs. -1,123 ± 323 µM·kg-1·2 h-1) and glucose disappearance (1,829 ± 807 vs. 3,606 ± 839 µM·kg-1·2 h-1) differed (P < 0.01) between complex and simple carbohydrate containing meals, respectively. Interestingly, there were significant increase in indices of insulin sensitivity (32.5 ± 3.5 vs. 25.6 ± 3.2 10-5 (dl·kg-1·min-2)/pM, P = 0.006) and ß-cell responsivity (disposition index: 1,817 ± 234 vs. 1,236 ± 159 10-14 (dl·kg-1·min-2)/pM, P < 0.005) with complex than simple carbohydrate meals. We present a novel triple tracer approach to estimate postprandial turnover of complex carbohydrate containing mixed meals. We also report higher insulin sensitivity and ß-cell responsivity with complex than with simple carbohydrates in mixed meals of identical calorie and macronutrient compositions in healthy adults.


Subject(s)
Carbohydrate Metabolism/physiology , Dietary Carbohydrates/metabolism , Polysaccharides , Radiopharmaceuticals , Adult , Algorithms , Carbon Isotopes , Female , Glucose/metabolism , Glucose/pharmacokinetics , Healthy Volunteers , Humans , Insulin Resistance , Insulin-Secreting Cells/metabolism , Male , Meals , Oryza , Postprandial Period , Sorghum , Young Adult
10.
Diabetes Obes Metab ; 20(7): 1615-1622, 2018 07.
Article in English | MEDLINE | ID: mdl-29493118

ABSTRACT

AIM: To investigate the mechanisms behind the lower postprandial glucose (PPG) concentrations achieved with fast-acting insulin aspart (faster aspart) than with insulin aspart (IAsp). MATERIALS AND METHODS: In a randomized, double-blind, crossover trial, 41 people with type 1 diabetes received identical subcutaneous single faster aspart and IAsp doses (individualized for each participant), together with a standardized mixed meal (including 75 g carbohydrate labelled with [1-13 C] glucose). PPG turnover was determined by the triple-tracer meal method using continuous, variable [6-3 H] glucose and [6,6-2 H2 ] glucose infusion. RESULTS: Insulin exposure within the first hour was 32% greater with faster aspart than with IAsp (treatment ratio faster aspart/IAsp 1.32 [95% confidence interval {CI} 1.18;1.48]; P < .001), leading to a 0.59-mmol/L non-significantly smaller PPG increment at 1 hour (ΔPG1h ; treatment difference faster aspart-IAsp -0.59 mmol/L [95% CI -1.19; 0.01]; P = .055). The trend towards reduced ΔPG1h with faster aspart was attributable to 12% greater suppression of endogenous glucose production (EGP; treatment ratio 1.12 [95% CI 1.01; 1.25]; P = .040) and 23% higher glucose disappearance (1.23 [95% CI 1.05; 1.45]; P = .012) with faster aspart than with IAsp during the first hour. Suppression of free fatty acid levels during the first hour was 36% greater for faster aspart than for IAsp (1.36 [95% CI 1.01;1.88]; P = .042). CONCLUSIONS: The trend towards improved PPG control with faster aspart vs IAsp in this study was attributable to both greater early suppression of EGP and stimulation of glucose disappearance.


Subject(s)
Blood Glucose/analysis , Diabetes Mellitus, Type 1/drug therapy , Drug Compounding , Gluconeogenesis/drug effects , Hyperglycemia/prevention & control , Hypoglycemic Agents/therapeutic use , Insulin Aspart/therapeutic use , Adult , Carbon Isotopes , Cross-Over Studies , Deuterium , Diabetes Mellitus, Type 1/blood , Diabetes Mellitus, Type 1/metabolism , Double-Blind Method , Fatty Acids, Nonesterified/blood , Female , Follow-Up Studies , Humans , Hypoglycemic Agents/blood , Hypoglycemic Agents/metabolism , Hypoglycemic Agents/pharmacokinetics , Insulin Aspart/blood , Insulin Aspart/metabolism , Insulin Aspart/pharmacokinetics , Male , Middle Aged , Postprandial Period , Tritium
11.
J Pharmacokinet Pharmacodyn ; 45(6): 829-845, 2018 12.
Article in English | MEDLINE | ID: mdl-30392154

ABSTRACT

Our objective is to develop a physiology-based model of insulin kinetics to understand how exercise alters insulin concentrations in those with type 1 diabetes (T1D). We reveal the relationship between the insulin absorption rate ([Formula: see text]) from subcutaneous tissue, the insulin delivery rate ([Formula: see text]) to skeletal muscle, and two physiological parameters that characterize the tissue: the perfusion rate (Q) and the capillary permeability surface area (PS), both of which increase during exercise because of capillary recruitment. We compare model predictions to experimental observations from two pump-wearing T1D cohorts [resting subjects ([Formula: see text]) and exercising subjects ([Formula: see text])] who were each given a mixed-meal tolerance test and a bolus of insulin. Using independently measured values of Q and PS from literature, the model predicts that during exercise insulin concentration increases by 30% in plasma and by 60% in skeletal muscle. Predictions reasonably agree with experimental observations from the two cohorts, without the need for parameter estimation by curve fitting. The insulin kinetics model suggests that the increase in surface area associated with exercise-induced capillary recruitment significantly increases [Formula: see text] and [Formula: see text], which explains why insulin concentrations in plasma and skeletal muscle increase during exercise, ultimately enhancing insulin-dependent glucose uptake. Preventing hypoglycemia is of paramount importance in determining the proper insulin dose during exercise. The presented model provides mechanistic insight into how exercise affects insulin kinetics, which could be useful in guiding the design of decision support systems and artificial pancreas control algorithms.


Subject(s)
Diabetes Mellitus, Type 1/drug therapy , Exercise/physiology , Insulin/pharmacokinetics , Models, Biological , Adult , Algorithms , Blood Glucose/drug effects , Blood Glucose/metabolism , Capillaries/metabolism , Capillary Permeability , Cohort Studies , Decision Support Techniques , Diabetes Mellitus, Type 1/blood , Diabetes Mellitus, Type 1/diagnosis , Female , Glucose Tolerance Test , Humans , Insulin/administration & dosage , Insulin Infusion Systems , Male , Middle Aged , Muscle, Skeletal/blood supply , Muscle, Skeletal/physiology , Pancreas, Artificial
12.
Adv Exp Med Biol ; 1043: 53-64, 2017.
Article in English | MEDLINE | ID: mdl-29224090

ABSTRACT

Sex difference plays a substantial role in the regulation of glucose metabolism in healthy glucose-tolerant humans. The factors which may contribute to the sex-related differences in glucose metabolism include differences in lifestyle (diet and exercise), sex hormones, and body composition. Several epidemiological and observational studies have noted that impaired glucose tolerance is more common in women than men. Some of these studies have attributed this to differences in body composition, while others have attributed impaired insulin sensitivity as a cause of impaired glucose tolerance in women. We studied postprandial glucose metabolism in 120 men and 90 women after ingestion of a mixed meal. Rates of meal glucose appearance, endogenous glucose production, and glucose disappearance were calculated using a novel triple-tracer isotope dilution method. Insulin action and secretion were calculated using validated physiological models. While rate of meal glucose appearance was higher in women than men, rates of glucose disappearance were higher in elderly women than elderly men while young women had lower rates of glucose disappearance than young men. Hence, sex has an impact on postprandial glucose metabolism, and sex differences in carbohydrate metabolism may have important implications for approaches to prevent and manage diabetes in an individual.


Subject(s)
Blood Glucose/metabolism , Insulin-Secreting Cells/metabolism , Insulin/metabolism , Postprandial Period , Age Factors , Aged , Biomarkers/blood , Female , Health Status Disparities , Humans , Insulin/blood , Insulin Secretion , Male , Sex Characteristics , Sex Factors , Time Factors , Young Adult
14.
Am J Physiol Endocrinol Metab ; 309(2): E161-7, 2015 Jul 15.
Article in English | MEDLINE | ID: mdl-25991649

ABSTRACT

Recently, a model was proposed to assess hepatic insulin sensitivity during a meal, i.e., the ability of insulin to suppress glucose production (EGP), SI (P). The model was developed on EGP data obtained from a triple-tracer meal and the tracer-to-tracee clamp technique and validated against the euglycemic hyperinsulinemic clamp. The aim of this study was to assess whether SI (P) can be obtained from plasma concentrations measured after a single-tracer meal by incorporating the above EGP model into the oral glucose minimal model by describing both glucose production and disposal (OMM(PD)). Triple-tracer meal data of two databases (20 healthy and 60 healthy and prediabetic subjects) were used. Virtually model-independent EGP estimates were calculated. OMM(PD) was identified on exogenous and endogenous glucose concentrations, providing indices of SI (P), disposal insulin sensitivity (SI (D)), and EGP. The model fitted the data well, and SI (P) and SI (D) were estimated with precision in both databases (SI (P) = 5.48 ± 0.54 10(-4) dl·kg(-1)·min(-1) per µU/ml and SI (D) = 9.93 ± 2.18 10(-4) dl·kg(-1)·min(-1) per µU/ml in healthy; SI (P) = 5.41 ± 3.55 10(-4) dl·kg(-1)·min(-1) per µU/ml and SI (D) = 5.34 ± 6.17 10(-4) dl·kg(-1)·min(-1) per µU/ml, in healthy and prediabetic subjects). Estimated SI (P) and that derived from the triple-tracer EGP model were very similar on average. Moreover, the time course of EGP normalized to basal EGP (EGPb), and EGP/EGPb agreed with the results obtained using the triple-tracer method. In this study, we have demonstrated that SI (P), SI (D), and EGP/EGPb time course can be estimated reliably from a single-tracer meal protocol in both healthy and prediabetic subjects.


Subject(s)
Indicators and Reagents/administration & dosage , Indicators and Reagents/pharmacokinetics , Insulin Resistance , Liver/metabolism , Models, Biological , Prediabetic State/metabolism , Administration, Oral , Adult , Female , Glucose Clamp Technique , Glucose Tolerance Test , Health , Humans , Insulin/metabolism , Male , Meals , Middle Aged , Radioactive Tracers
15.
Am J Physiol Endocrinol Metab ; 309(5): E474-86, 2015 Sep 01.
Article in English | MEDLINE | ID: mdl-26152766

ABSTRACT

Glucagon use in artificial pancreas for type 1 diabetes (T1D) is being explored for prevention and rescue from hypoglycemia. However, the relationship between glucagon stimulation of endogenous glucose production (EGP) viz., hepatic glucagon sensitivity, and prevailing glucose concentrations has not been examined. To test the hypothesis that glucagon sensitivity is increased at hypoglycemia vs. euglycemia, we studied 29 subjects with T1D randomized to a hypoglycemia or euglycemia clamp. Each subject was studied at three glucagon doses at euglycemia or hypoglycemia, with EGP measured by isotope dilution technique. The peak EGP increments and the integrated EGP response increased with increasing glucagon dose during euglycemia and hypoglycemia. However, the difference in dose response based on glycemia was not significant despite higher catecholamine concentrations in the hypoglycemia group. Knowledge of glucagon's effects on EGP was used to develop an in silico glucagon action model. The model-derived output fitted the obtained data at both euglycemia and hypoglycemia for all glucagon doses tested. Glucagon clearance did not differ between glucagon doses studied in both groups. Therefore, the glucagon controller of a dual hormone control system may not need to adjust glucagon sensitivity, and hence glucagon dosing, based on glucose concentrations during euglycemia and hypoglycemia.


Subject(s)
Blood Glucose/metabolism , Diabetes Mellitus, Type 1/metabolism , Glucagon/pharmacology , Gluconeogenesis/drug effects , Glycogenolysis/drug effects , Hormones/pharmacology , Hypoglycemia/metabolism , Liver/drug effects , Adult , Blood Glucose/drug effects , Computer Simulation , Female , Glucose/metabolism , Glucose Clamp Technique , Humans , Liver/metabolism , Male , Middle Aged , Pancreas, Artificial , Young Adult
16.
Am J Physiol Endocrinol Metab ; 308(12): E1106-15, 2015 Jun 15.
Article in English | MEDLINE | ID: mdl-25898950

ABSTRACT

To determine the effects of exercise on postprandial glucose metabolism and insulin action in type 1 diabetes (T1D), we applied the triple tracer technique to study 16 T1D subjects on insulin pump therapy before, during, and after 75 min of moderate-intensity exercise (50% V̇o2max) that started 120 min after a mixed meal containing 75 g of labeled glucose. Prandial insulin bolus was administered as per each subject's customary insulin/carbohydrate ratio adjusted for meal time meter glucose and the level of physical activity. Basal insulin infusion rates were not altered. There were no episodes of hypoglycemia during the study. Plasma dopamine and norepinephrine concentrations rose during exercise. During exercise, rates of endogenous glucose production rose rapidly to baseline levels despite high circulating insulin and glucose concentrations. Interestingly, plasma insulin concentrations increased during exercise despite no changes in insulin pump infusion rates, implying increased mobilization of insulin from subcutaneous depots. Glucagon concentrations rose before and during exercise. Therapeutic approaches for T1D management during exercise will need to account for its effects on glucose turnover, insulin mobilization, glucagon, and sympathetic response and possibly other blood-borne feedback and afferent reflex mechanisms to improve both hypoglycemia and hyperglycemia.


Subject(s)
Blood Glucose/metabolism , Diabetes Mellitus, Type 1/metabolism , Exercise/physiology , Insulin/metabolism , Postprandial Period , Adolescent , Adult , Aged , Diabetes Mellitus, Type 1/drug therapy , Exercise Test , Female , Gastric Emptying , Humans , Insulin/administration & dosage , Insulin Infusion Systems , Male , Middle Aged , Young Adult
17.
Curr Diab Rep ; 15(6): 605, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25869240

ABSTRACT

Glucose effectiveness (SG) is the ability of glucose per se to stimulate its own uptake and to suppress its own production under basal/constant insulin concentrations. In an individual, glucose tolerance is a function of insulin secretion, insulin action and SG. Under conditions of declining insulin secretion and action (e.g. type 2 diabetes), the degree of SG assumes increasing significance in determining the level of glucose tolerance both in fasted and postprandial states. Although the importance of SG has been recognized for years, mechanisms that contribute to SG are poorly understood. Research data on modulation of SG and its impact in glucose intolerance is limited. In this review, we will focus on the role of SG in the regulation of glucose tolerance, its evaluation, and potential advantages of therapies that can enhance glucose-induced stimulation of glucose uptake and suppression of its own production in conditions of impaired insulin secretion and action.


Subject(s)
Diabetes Mellitus, Type 2/metabolism , Glucose Intolerance/metabolism , Glucose/metabolism , Blood Glucose/metabolism , Diabetes Mellitus, Type 2/physiopathology , Glucose Intolerance/diagnosis , Glucose Intolerance/physiopathology , Homeostasis , Humans , Insulin/metabolism , Models, Biological
18.
Exp Physiol ; 100(1): 69-78, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25557731

ABSTRACT

NEW FINDINGS: What is the central question of this study? Hyperoxia blunts hypoglycaemia counterregulation in healthy adults. We hypothesized that this effect is mediated by the carotid bodies and that: (i) hyperoxia would have no effect on hypoglycaemia counterregulation in carotid body-resected patients; and (ii) carotid body-resected patients would exhibit an impaired counterregulatory response to hypoglycaemia. What is the main finding and its importance? Our data indicate that the effect of hyperoxia on hypoglycaemic counterregulation is mediated by the carotid bodies. However, a relatively normal counterregulatory response to hypoglycaemia in carotid body-resected patients highlights: (i) the potential for long-term adaptations after carotid body resection; and (ii) the importance of redundant mechanisms in mediating hypoglycaemia counterregulation. Hyperoxia reduces hypoglycaemia counterregulation in healthy adults. We hypothesized that this effect is mediated by the carotid bodies and that: (i) hyperoxia would have no effect on hypoglycaemia counterregulation in patients with bilateral carotid body resection; and (ii) carotid body-resected patients would exhibit an impaired counterregulatory response to hypoglycaemia. Five patients (three male and two female) with bilateral carotid body resection for glomus tumours underwent two 180 min hyperinsulinaemic, hypoglycaemic (∼ 3.3 mmol l(-1)) clamps separated by a minimum of 1 week and randomized to either normoxia (21% fractional inspired O2 ) or hyperoxia (100% fractional inspired O2). Ten healthy adults (seven male and three female) served as control subjects. Hypoglycaemia counterregulation in carotid body-resected patients was not significantly altered by hyperoxia (area under the curve expressed as a percentage of the normoxic response: glucose infusion rate, 111 ± 10%; cortisol, 94 ± 6%; glucagon, 107 ± 7%; growth hormone, 92 ± 10%; adrenaline, 89 ± 26%; noradrenaline, 79 ± 15%; main effect of condition, P > 0.05). This is in contrast to previously published results from healthy adults. However, the counterregulatory responses to hypoglycaemia during normoxia were not impaired in carotid body-resected patients when compared with control subjects (main effect of group, P > 0.05). Our data provide further corroborative evidence that the effect of hyperoxia on hypoglycaemic counterregulation is mediated by the carotid bodies. However, relatively normal counterregulatory responses to hypoglycaemia in carotid body-resected patients highlight the importance of redundant mechanisms in mediating hypoglycaemia counterregulation.


Subject(s)
Carotid Body Tumor/surgery , Carotid Body/surgery , Glomus Tumor/surgery , Hyperoxia/physiopathology , Hypoglycemia/physiopathology , Adaptation, Physiological , Adult , Biomarkers/blood , Blood Glucose/metabolism , Carotid Body/physiopathology , Carotid Body Tumor/physiopathology , Female , Glomus Tumor/physiopathology , Humans , Hyperoxia/blood , Hypoglycemia/blood , Insulin/blood , Male , Middle Aged , Minnesota , Time Factors , Young Adult
19.
Clin Auton Res ; 25(4): 243-50, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26141615

ABSTRACT

PURPOSE: Patients with type 1 diabetes mellitus exhibit impairments in autonomic and cardiovascular control which are worsened with acute hypoglycemia--thus increasing the risk of adverse cardiovascular events. Hypoxia, as seen with the common comorbidity of sleep apnea, may lead to further autonomic dysfunction and an increased risk of ventricular arrhythmias. Therefore, we hypothesized that heart rate variability (HRV) and baroreflex sensitivity (BRS) would be reduced during hypoglycemia in adults with type 1 diabetes, with a further decline when combined with hypoxia. METHODS: Subjects with type 1 diabetes (n = 13; HbA1c = 7.5 ± 0.3 %, duration of diabetes = 17 ± 5 yrs) completed two 180 min hyperinsulinemic (2 mU/kg TBW/min), hypoglycemic (~3.3 µmol/mL) clamps separated by a minimum of 1 week and randomized to normoxia (SpO2 ~98 %) or hypoxia (SpO2 ~85 %). Heart rate (electrocardiogram) and blood pressure (finger photoplethysmography) were analyzed at baseline and during the hypoglycemic clamp for measures of HRV and spontaneous cardiac BRS (sCBRS). RESULTS: Hypoglycemia resulted in significant reductions in HRV and sCBRS when compared with baseline levels (main effect of hypoglycemia: p < 0.05). HRV and sCBRS were further impaired during hypoxia (main effect of hypoxia: p < 0.05). CONCLUSIONS: Acute hypoxia worsens hypoglycemia-mediated impairments in autonomic and cardiovascular control in patients with type 1 diabetes and may increase the risk of cardiovascular mortality. These results highlight the potential cumulative dangers of hypoglycemia and hypoxia in this vulnerable population.


Subject(s)
Baroreflex/physiology , Diabetes Mellitus, Type 1/epidemiology , Heart Rate/physiology , Hypoglycemia/epidemiology , Hypoxia/epidemiology , Adult , Diabetes Mellitus, Type 1/diagnosis , Diabetes Mellitus, Type 1/physiopathology , Female , Humans , Hypoglycemia/diagnosis , Hypoglycemia/physiopathology , Hypoxia/diagnosis , Hypoxia/physiopathology , Male , Middle Aged
20.
Diabetologia ; 57(7): 1446-55, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24771091

ABSTRACT

AIMS/HYPOTHESIS: Dysregulation of 11ß-hydroxysteroid dehydrogenase (11ß-HSD) enzyme activities are implicated in the pathogenesis of obesity and insulin resistance. The aim of the study was to determine whether hepatic 11ß-HSD type 1 (11ß-HSD-1) enzyme activity differs in people with and without obesity and type 2 diabetes. METHODS: We measured hepatic 11ß-HSD-1 activity in the overnight fasted state in 20 lean non-diabetic participants (LND), 21 overweight/obese non-diabetic participants (OND) and 20 overweight/obese participants with type 2 diabetes (ODM) using a non-invasive approach. One mg doses of [9,12,12-(2)H3]cortisol (D cortisol) and [4-(13)C]cortisone ([(13)C]cortisone) were ingested, while [1,2,6,7-(3)H]cortisol ([(3)H] cortisol) was infused intravenously to enable concurrent measurements of first-pass hepatic extraction of ingested D cortisol and hepatic conversion of ingested [(13)C]cortisone to C13 cortisol derived from the ingested cortisone (a measure of 11ß-HSD-1 activity in the liver) using an isotope dilution technique. One-way ANOVA models and Kruskal-Wallis tests were used to test the hypothesis. RESULTS: Plasma D cortisol and C13 cortisol concentrations were lower in OND than in LND (p < 0.05) over 6 h of the study. There was no difference (p = 0.15) in C13 and D cortisol concentrations between OND and ODM and between LND and ODM for the same study period. Hepatic conversion of [(13)C]cortisone to C13 cortisol was similar between groups. CONCLUSIONS/INTERPRETATION: Hepatic conversion of [(13)C]cortisone to C13 cortisol did not differ between the groups studied. We conclude that hepatic 11ß-HSD-1 activity is similar in individuals who are overweight/obese or who have type 2 diabetes.


Subject(s)
11-beta-Hydroxysteroid Dehydrogenase Type 1/metabolism , Diabetes Mellitus, Type 2/metabolism , Liver/enzymology , Obesity/enzymology , Adult , Aged , Body Mass Index , Female , Humans , Insulin Resistance , Male , Middle Aged
SELECTION OF CITATIONS
SEARCH DETAIL