Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 119
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Appetite ; 193: 107160, 2024 02 01.
Article in English | MEDLINE | ID: mdl-38101518

ABSTRACT

We have shown insulin resistance is associated with the choice of sugar-sweetened over monk fruit sweetened yogurt. This study extends this research by assessing the association between insulin resistance and reinforcing value for sugar versus monk fruit-sweetened yogurt, and testing the hypothesis that this effect is moderated by greater blood glucose response in people with insulin resistance. Eighteen people with overweight/obesity (BMI = 35.8 kg/m2, range 26.2-48.5) with varying degrees of insulin resistance (Homeostatic Model Assessment of Insulin Resistance (HOMA-IR) = 2.6, range of 0.6-8.0) had blood glucose measured for 2 h after a sugar challenge. Over six days, they consumed, in a double-blind fashion, novel flavored-colored sugar or monk fruit-sweetened yogurts, and the reinforcing value of sugar or monk fruit-sweetened yogurts and delay discounting (DD) were measured. HOMA-IR (r = 0.62, p = .006) and insulin (r = 0.51, p = .03) were related to the reinforcing value of sugar-sweetened, but not monk fruit-sweetened yogurt (r = -0.07, -0.10, respectively). The blood glucose area under the curve moderated the relationship between HOMA-IR and the reinforcing value of sugar-sweetened yogurt (p = .02). People with greater HOMA-IR and greater blood glucose excursions responded the most for sugar-sweetened yogurt. These results extend previous research and confirm the hypothesis that individual differences in response to sugar may activate brain reward centers and condition people to prefer high-sugar foods. DD was related to sugar reinforcement (r = -0.46, p = .03), consistent with the idea that those with high sugar reinforcement desire immediate gratification, and DD moderated the relationship between HOMA-IR and the reinforcing value of sugar-sweetened yogurt (p < .001). Research should test whether reducing insulin resistance would permit people with insulin resistance to choose lower-sugar foods.


Subject(s)
Glucose , Insulin Resistance , Humans , Blood Glucose , Sugars , Yogurt , Beverages , Insulin
2.
Am J Physiol Endocrinol Metab ; 325(4): E406-E411, 2023 10 01.
Article in English | MEDLINE | ID: mdl-37672250

ABSTRACT

Obesity is associated with hypertension. However, the mechanisms involved are not fully understood. Therefore, we investigated the relationship between obesity and vasoactive mediators. In this cross-sectional study, blood pressure (BP) and vasoactive mediators of hypertension are compared among 135 adults in the nonobese, obese, and morbidly obese body mass index (BMI) ranges (BMI ≤27, 30-40, and >40 kg/m2, respectively). Angiotensinogen, angiotensin II, renin, aldosterone, endothelin-1 (ET-1), neprilysin, atrial natriuretic peptide (ANP), brain natriuretic peptide (BNP), cyclic guanosine monophosphate (cGMP), and cyclic adenosine monophosphate (cAMP) levels were measured and their relationship to BP, BMI, race, and gender were investigated. Systolic and diastolic BP (SBP and DSP) were significantly higher in subjects with obesity and morbid obesity compared with nonobese. Angiotensin II, ET-1, and neprilysin were significantly higher in subjects with morbid obesity while BNP was lower. Levels of angiotensinogen, renin, aldosterone, ANP, cGMP, and cAMP did not differ between the groups. BMI was positively related to SBP, DBP, angiotensin II, ET-1, and neprilysin, and inversely related to cGMP and BNP. Age, male gender, and African-American race were associated with higher SBP. SBP was positively related to angiotensin II and ET-1 and inversely related to aldosterone, renin, and cGMP. On multivariate analyses, age, BMI, gender, and race were the main determinants of SBP, and excluding these variables, angiotensin II, aldosterone, renin, and ET-1 accounted for 21.1% ability to predict SBP. Obesity, especially morbid obesity, is associated with higher BP, higher angiotensin II and ET-1 (vasoconstrictors), and lower levels BNP and cGMP (vasodilators). SBP variability can be partly explained by angiotensin II, aldosterone, renin, and ET-1.NEW & NOTEWORTHY Our data show that obesity, especially morbid obesity, is associated with higher blood pressure levels and increases angiotensin II and endotherlin-1 (ET-1) (vasoconstrictors) and lower levels BNP and cGMP (vasodilators) and that systolic blood pressure variability can be partly explained by levels of angiotensin II, aldosterone, renin, and ET-1. The effect of these mediators on blood pressure is in addition to the effects of other known factors related to age, male gender, and AA race.


Subject(s)
Hypertension , Obesity, Morbid , Adult , Humans , Male , Renin , Angiotensinogen/pharmacology , Renin-Angiotensin System , Aldosterone/pharmacology , Angiotensin II/pharmacology , Cross-Sectional Studies , Neprilysin , Hypertension/complications , Blood Pressure , Vasodilator Agents , Vasoconstrictor Agents
3.
Psychosom Med ; 85(3): 289-293, 2023 04 01.
Article in English | MEDLINE | ID: mdl-36799726

ABSTRACT

OBJECTIVE: Insulin resistance is associated with elevated activation of food reward, which should be associated with an increased reinforcing value of food. Research has also shown that sugar is a macronutrient strongly associated with reward and reinforcing value of food. This research is designed to assess whether insulin resistance is associated with a stronger preference for sugar-sweetened, thus elevating blood glucose responses in obese people with varying degrees of insulin resistance. METHODS: Thirteen people with obesity (body mass index, 39.1 kg/m 2 ; range, 30.0-45.1 kg/m 2 ) with varying degrees of insulin resistance (Homeostatic Model Assessment of Insulin Resistance, 5.2; range, 0.7-11.6) consumed novel flavored-colored yogurts that were sweetened with either sugar or monkfruit daily for 6 days to assess whether when given the choice of sugar-sweetened versus monkfruit-sweetened yogurts to consume, participants preferred sugar-sweetened yogurts. RESULTS: Participants consumed a greater amount ( p = .009) and percentage ( p = .04) of sugar-sweetened yogurt earned than monkfruit-sweetened yogurt. The percent of sugar-sweetened versus monkfruit-sweetened yogurt consumed in relationship to amount earned was related to insulin resistance ( r = 0.64, p = .019), glycated hemoglobin ( r = 0.61, p = .027), insulin ( r = 0.58, p = .007), and glucose ( r = 0.56, p = .048). CONCLUSIONS: Insulin resistance is associated with preference for sugar-sweetened foods in participants with obesity, which may make it hard to make dietary changes. Research is needed to assess whether treatments that improve insulin resistance also change the preference for sugar-sweetened or high-glycemic-index foods.


Subject(s)
Insulin Resistance , Humans , Insulin Resistance/physiology , Glycated Hemoglobin , Pilot Projects , Sugars , Yogurt , Obesity
5.
Diabetes Obes Metab ; 24(3): 365-376, 2022 03.
Article in English | MEDLINE | ID: mdl-34779091

ABSTRACT

Based on global estimates, almost 10% of adults have diabetes, of whom 40% are estimated to also have chronic kidney disease (CKD). Almost 2 decades ago, treatments targeting the renin-angiotensin system (RAS) were shown to slow the progression of kidney disease. More recently, studies have reported the additive benefits of antihyperglycaemic sodium-glucose co-transporter-2 inhibitors in combination with RAS inhibitors on both CKD progression and cardiovascular outcomes. However, these recent data also showed that patients continue to progress to kidney failure or die from kidney- or cardiovascular-related causes. Therefore, new agents are needed to address this continuing risk. Overactivation of the mineralocorticoid (MR) receptor contributes to kidney inflammation and fibrosis, suggesting that it is an appropriate treatment target in patients with diabetes and CKD. Novel, selective non-steroidal MR antagonists are being studied in these patients, and the results of two large recently completed clinical trials have shown that one such treatment, finerenone, significantly reduces CKD progression and cardiovascular events compared with standard of care. This review summarizes the pathogenic mechanisms of CKD in type 2 diabetes and examines the potential benefit of novel disease-modifying agents that target inflammatory and fibrotic factors in these patients.


Subject(s)
Diabetes Mellitus, Type 2 , Diabetic Nephropathies , Renal Insufficiency, Chronic , Sodium-Glucose Transporter 2 Inhibitors , Adult , Diabetes Mellitus, Type 2/chemically induced , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/drug therapy , Diabetic Nephropathies/drug therapy , Diabetic Nephropathies/etiology , Diabetic Nephropathies/prevention & control , Humans , Kidney , Mineralocorticoid Receptor Antagonists/therapeutic use , Renal Insufficiency, Chronic/chemically induced , Renal Insufficiency, Chronic/complications , Renal Insufficiency, Chronic/drug therapy , Sodium-Glucose Transporter 2 Inhibitors/therapeutic use
7.
Diabetes Obes Metab ; 23(1): 18-28, 2021 01.
Article in English | MEDLINE | ID: mdl-32991053

ABSTRACT

The role of testosterone in improving sexual symptoms in men with hypogonadism is well known. However, recent studies indicate that testosterone plays an important role in several metabolic functions in males. Multiple PubMed searches were conducted with the use of the terms testosterone, insulin sensitivity, obesity, type 2 diabetes, anaemia, bone density, osteoporosis, fat mass, lean mass and body composition. This narrative review is focused on detailing the mechanisms that underlie the metabolic aspects of testosterone therapy in humans. Testosterone enhances insulin sensitivity in obese men with hypogonadism by decreasing fat mass, increasing lean mass, decreasing free fatty acids and suppressing inflammation. At a cellular level, testosterone increases the expression of insulin receptor ß subunit, insulin receptor substrate-1, protein kinase B and glucose transporter type 4 in adipose tissue and adenosine 5'-monophosphate-activated protein kinase expression and activity in skeletal muscle. Observational studies show that long-term therapy with testosterone prevents progression from prediabetes to diabetes and improves HbA1c. Testosterone increases skeletal muscle satellite cell activator, fibroblast growth factor-2 and decreases expression of the muscle growth suppressors, myostatin and myogenic regulatory factor 4. Testosterone increases haematocrit by suppressing hepcidin and increasing expression of ferroportin along with that of transferrin receptor and plasma transferrin concentrations. Testosterone also increases serum osteocalcin concentrations, which may account for its anabolic actions on bone. In conclusion, testosterone exerts a series of potent metabolic effects, which include insulin sensitization, maintenance and growth of the skeletal muscle, suppression of adipose tissue growth and maintenance of erythropoiesis and haematocrit.


Subject(s)
Diabetes Mellitus, Type 2 , Hypogonadism , Insulin Resistance , Body Composition , Diabetes Mellitus, Type 2/drug therapy , Humans , Hypogonadism/drug therapy , Male , Testosterone
8.
Diabetes Obes Metab ; 23(7): 1614-1623, 2021 07.
Article in English | MEDLINE | ID: mdl-33729664

ABSTRACT

AIM: To investigate the mechanisms underlying improvements in blood pressure (BP) and congestive heart failure outcomes following treatment with dapagliflozin, a sodium-glucose co-transporter-2 inhibitor. RESEARCH DESIGN AND METHODS: A total of 52 patients with type 2 diabetes (T2D) with an HbA1c of less than 8% participated in this prospective, double-blind and placebo-controlled study. Patients were randomized (1:1) to either dapagliflozin 10 mg daily or placebo for 12 weeks. Half the patients were also monitored for 6 h following their first dose for acute effects on BP. Blood and urine samples were collected and levels of angiotensinogen, angiotensin II, renin, aldosterone, endothelin-1, atrial natriuretic peptide (ANP), brain natriuretic peptide, cyclic adenosine monophosphate, cyclic guanosine monophosphate (cGMP) and neprilysin were measured. The expression of angiotensin-converting enzyme, guanylate cyclase and phosphodiesterase 5 (PDE5) was measured in circulating mononuclear cells (MNC). RESULTS: A total of 24 and 23 patients receiving dapagliflozin and placebo, respectively, completed the 12-week study. Systolic BP decreased significantly, compared with placebo, both after single-dose (by 7 ± 3 mmHg) and 12-week (by 7 ± 2 mmHg) treatment with dapagliflozin. Dapagliflozin suppressed angiotensin II and angiotensinogen (by 10.5 ± 2.1 and 1.45 ± 0.42 µg/mL, respectively) and increased ANP and cGMP (by 34 ± 11 and 29 ± 11 pmol/mL, respectively) compared with the placebo group. cGMP levels also increased acutely following a single dose of dapagliflozin. Dapagliflozin also suppressed PDE5 expression by 26% ± 11% in MNC. There were no changes observed in the other vasoactive mediators investigated. CONCLUSIONS: Dapagliflozin administration in T2D resulted in both acute and chronic reduction in systolic BP, a reduction in vasoconstrictors and an increase in vasodilators. These changes may potentially contribute to its antihypertensive effects and its benefits in congestive cardiac failure.


Subject(s)
Diabetes Mellitus, Type 2 , Benzhydryl Compounds , Blood Glucose , Blood Pressure , Diabetes Mellitus, Type 2/drug therapy , Double-Blind Method , Glucosides/therapeutic use , Humans , Prospective Studies
9.
Diabetes Metab Res Rev ; 36(6): e3312, 2020 09.
Article in English | MEDLINE | ID: mdl-32191386

ABSTRACT

BACKGROUND: We examined the hearing function in adults with and without type 1 diabetes (T1D) to investigate whether an association exists between hearing loss and duration of diabetes, haemoglobin A1C level, diabetes complications and levels of select serum and urinary biomarkers. METHODS: We measured pure tone audiometry (PTA) thresholds; serum levels of C-reactive protein (CRP), vascular endothelial growth factor (VEGF), soluble receptors for advanced glycation end-product (sRAGE); and urinary isoprostane in 30 adults with T1D (age 43.8 ± 11.4 years). We also measured PTA thresholds in 11 adults without diabetes (age 53 ± 5.5 years). RESULTS: 63.3% of adults with T1D had high-frequency hearing loss. Among adults with T1D, those with hearing loss were older (48.2 vs 36.2 years old, P < .01), had a longer duration of diabetes (30.7 vs 21.2 years, P = .02), a greater prevalence of peripheral neuropathy (57.9 vs 9.1%, P = .02) and significantly lower median levels of sRAGE (1054.27 vs 1306.83 pg/mL, P = .03) compared to those with normal hearing. Adults with T1D between the ages of 40 and 60 years old, who had diabetes for ≥35 years, had significantly higher PTA thresholds at both 500and 8000 Hz than age-matched adults without diabetes. CONCLUSIONS: A significant proportion of adults with T1D have high-frequency hearing loss before age of 60 that is positively associated with age, duration of diabetes and presence of peripheral neuropathy. Our results are in support of previous studies suggesting a potential protective role of sRAGE against AGE toxicity and diabetes complications.


Subject(s)
Biomarkers/blood , Diabetes Complications/diagnosis , Diabetes Mellitus, Type 1/complications , Hearing Loss/diagnosis , Receptor for Advanced Glycation End Products/blood , Vascular Diseases/diagnosis , Adult , Blood Glucose/analysis , Case-Control Studies , Diabetes Complications/blood , Diabetes Complications/etiology , Female , Follow-Up Studies , Glycated Hemoglobin/analysis , Hearing Loss/blood , Hearing Loss/etiology , Humans , Male , Middle Aged , Prognosis , Vascular Diseases/blood , Vascular Diseases/etiology , Young Adult
10.
Diabetes Obes Metab ; 22(10): 1742-1752, 2020 10.
Article in English | MEDLINE | ID: mdl-32424935

ABSTRACT

AIM: To investigate the effects of liraglutide treatment on glycaemic control and adipose tissue metabolism in overweight and obese people with type 1 diabetes (T1DM). RESEARCH DESIGN AND METHODS: A total of 84 adult overweight and obese patients with T1DM, with no detectable C-peptide, were randomized (1:1) to either placebo or 1.8 mg/d liraglutide for 6 months. Blood samples were collected at 0, 12 and 26 weeks. Subcutaneous adipose tissue biopsies, a high-calorie high-fat meal challenge test, continuous glucose monitoring, dual-energy X-ray absorptiometry and MRI were performed before and at the end of treatment. RESULTS: In all, 37 and 27 patients who received liraglutide and placebo, respectively, completed the study. Glycated haemoglobin fell by 0.41 ± 0.18% (4.5±1.4 mmol/mol) from baseline after liraglutide treatment (P = 0.001), and by 0.29 ± 0.19% (3.1±2.0 mmol/mol) compared to placebo (P = 0.1). There was no increase in hypoglycaemia, while the time spent in normal glycaemia increased (P = 0.015) and time spent in hyperglycaemia decreased (P = 0.019). Body weight fell significantly in the liraglutide group, mostly in the form of fat mass loss (including visceral fat), with no change in lean mass. Systolic blood pressure (SBP) also fell after liraglutide treatment. Liraglutide also caused a significant increase in the expression of adipose tissue triglyceride lipase, carnitine palmitoyl transferase-1, peroxisome proliferator-activated receptor (PPAR)α, PPARδ, uncoupling protein-2 and type 2 iodothyronine deiodinase in the adipose tissue. CONCLUSIONS: Liraglutide improves glycaemia, reduces adiposity and SBP. Liraglutide also stimulates mechanisms involved with an increase in lipid oxidation and thermogenesis, while conserving lean body mass.


Subject(s)
Diabetes Mellitus, Type 1 , Diabetes Mellitus, Type 2 , Adult , Blood Glucose , Blood Glucose Self-Monitoring , Diabetes Mellitus, Type 1/complications , Diabetes Mellitus, Type 1/drug therapy , Double-Blind Method , Glycated Hemoglobin/analysis , Humans , Hypoglycemic Agents/therapeutic use , Liraglutide/therapeutic use , Obesity/complications , Overweight/complications , Overweight/drug therapy , Treatment Outcome , Weight Loss
11.
Diabetes Obes Metab ; 21(4): 1054-1057, 2019 04.
Article in English | MEDLINE | ID: mdl-30536789

ABSTRACT

We aimed to test the hypothesis that addition of glucagon-like peptide-1 receptor agonists (GLP-1RAs) to insulin in C-peptide-positive patients with type 1 diabetes (T1D) will result in a reduction in glycated haemoglobin (HbA1c) with reduced insulin requirements and a rise in C-peptide concentrations. We conducted a retrospective analysis of 11 normal-weight patients with T1D consecutively treated with a GLP-1RA in addition to insulin. Paired t tests were used to compare the changes in HbA1c, insulin doses, body weight, body mass index, and C-peptide concentrations prior to and 12 ± 1 weeks after GLP-1RA therapy. At the end of 12 ± 1 weeks of GLP-1RA therapy, HbA1c fell from 10.74 ± 0.96% (95 ± 10.5 mmol/mol) to 7.4 ± 0.58% (58 ± 6.3mmol/mol) (P < 0.01), body weight fell from 71 ± 2.0 to 69 ± 2 kg (P = 0.06), and total insulin dose was reduced by 64% from 33 ± 6 to 11 ± 5 units (P < 0.01). Five out of 10 patients did not require any insulin. C-peptide concentrations increased significantly from 0.43 ± 0.09 ng/ml (0.14 ± 0.02 nmol/L) to 1.42 ± 0.42ng/ml (0.47 ± 0.13 nmol/L) (P = 0.01). Addition of GLP-1RA therapy to insulin in normal-weight patients with T1D led to a reduction in HbA1c with reduced insulin requirements, a 3.5-fold increase in C-peptide concentrations and freedom from insulin therapy in 50% of patients who tolerated the GLP-1RA therapy over a period of 12 ± 1 weeks.


Subject(s)
Diabetes Mellitus, Type 1/drug therapy , Glucagon-Like Peptide-1 Receptor/agonists , Glucagon-Like Peptides/analogs & derivatives , Hypoglycemic Agents/therapeutic use , Immunoglobulin Fc Fragments/therapeutic use , Insulin/therapeutic use , Liraglutide/therapeutic use , Recombinant Fusion Proteins/therapeutic use , Adult , Autoantibodies/immunology , Body Weight , C-Peptide/metabolism , Deprescriptions , Diabetes Mellitus, Type 1/metabolism , Drug Therapy, Combination , Female , Glucagon-Like Peptides/therapeutic use , Glutamate Decarboxylase/immunology , Glycated Hemoglobin/metabolism , Humans , Male , Middle Aged , Retrospective Studies , Treatment Outcome , Weight Loss
12.
Diabetes Obes Metab ; 20 Suppl 1: 59-67, 2018 02.
Article in English | MEDLINE | ID: mdl-29364583

ABSTRACT

While the use of incretins, including GLP-1 receptor agonists and PDD-IV inhibitors, is well established in the treatment of type 2 diabetes, many other aspects of these agents are yet to be discovered and utilized for their potential clinical benefit. These include the potential role of GLP-1 receptor agonists in the induction of weight loss, blood pressure reduction, anti-inflammatory and nephro- and cardio-protective actions. Their potential benefit in type 1 diabetes is also being investigated. This review will attempt to comprehensively describe novel discoveries in the field of incretin pathophysiology and pharmacology beyond their classical role in the treatment of type 2 diabetes.


Subject(s)
Dipeptidyl-Peptidase IV Inhibitors/therapeutic use , Drug Repositioning , Glucagon-Like Peptide-1 Receptor/agonists , Hypoglycemic Agents/therapeutic use , Incretins/pharmacology , Diabetes Mellitus, Type 1/drug therapy , Humans , Weight Loss/drug effects
13.
Diabetes Obes Metab ; 20(8): 2029-2033, 2018 08.
Article in English | MEDLINE | ID: mdl-29652095

ABSTRACT

The aim of this study was to determine if weight loss following Roux-en-Y gastric bypass (RYGB) surgery in morbidly obese patients is associated with a decrease in plasma concentrations of neprilysin, mediators of the renin angiotensin system (RAS), catecholamines and endothelin-1, and also with an increase in the concentrations of vasodilators. Fasting blood samples were obtained from 15 patients with morbid obesity and diabetes prior to and 6 months after RYGB surgery. Circulating levels of neprilysin, vasoconstrictors, vasodilators, and the mRNA expression of related genes in circulating mononuclear cells (MNC) were measured. Six months after RYGB surgery the concentrations of neprilysin, angiotensinogen, angiotensin II, renin and endothelin-1 fell significantly by 27 ±16%, 22 ±10%, 22 ±8%, 35 ±13% and 17 ±6% (P < .05 for all), respectively, while ANP concentrations increased significantly by 24 ±13%. There was no significant change in aldosterone, BNP, cAMP or cGMP concentrations, or angiotensin converting enzyme (ACE) expression. These changes may contribute to the reduction of congestive cardiac failure and blood pressure risks after RYGB surgery.


Subject(s)
Bariatric Surgery/adverse effects , Endothelin-1/blood , Heart Failure/prevention & control , Hypertension/prevention & control , Neprilysin/blood , Obesity, Morbid/surgery , Renin-Angiotensin System , Body Mass Index , Diabetes Mellitus, Type 2/complications , Diabetic Angiopathies/epidemiology , Diabetic Angiopathies/prevention & control , Diabetic Cardiomyopathies/epidemiology , Diabetic Cardiomyopathies/prevention & control , Female , Glycated Hemoglobin/analysis , Heart Failure/complications , Heart Failure/epidemiology , Heart Failure/etiology , Humans , Hypertension/complications , Hypertension/epidemiology , Hypertension/etiology , Insulin Resistance , Male , Middle Aged , Obesity, Morbid/complications , Obesity, Morbid/metabolism , Obesity, Morbid/physiopathology , Postoperative Period , Prospective Studies , Risk , Weight Loss
14.
Diabetes Obes Metab ; 19(5): 729-733, 2017 05.
Article in English | MEDLINE | ID: mdl-27891769

ABSTRACT

In view of the known vasodilatory effects of glucagon-like peptide-1 and exenatide, we investigated the effects of exenatide on vasoactive factors. We analysed blood samples and mononuclear cells (MNCs) from a previous study, collected after a single dose and 12 weeks of exenatide or placebo treatment in a series of 24 patients with type 2 diabetes mellitus. After exenatide treatment, plasma concentrations of atrial natriuretic peptide, cyclic guanyl monophosphate (cGMP) and cyclic adenyl monophosphate increased significantly at 12 weeks. Plasma cGMP and adenylate cyclase expression in MNCs increased significantly after a single dose. Angiotensinogen concentration fell significantly 2 hours after a single dose and at 12 weeks, while renin and angiotensin II levels fell significantly only after a single dose and not after 12 weeks of treatment. Exenatide also suppressed the plasma concentration of transforming growth factor-ß and the expression of P311 in MNCs at 12 weeks. Thus, exenatide induces an increase in a series of vasodilators, while suppressing the renin-angiotensin system. These changes may contribute to the overall vasodilatory effect of exenatide.


Subject(s)
Antihypertensive Agents/therapeutic use , Atrial Natriuretic Factor/agonists , Gene Expression Regulation/drug effects , Glucagon-Like Peptide 1/agonists , Leukocytes, Mononuclear/drug effects , Nerve Tissue Proteins/antagonists & inhibitors , Oncogene Proteins/antagonists & inhibitors , Peptides/therapeutic use , Venoms/therapeutic use , Adenylyl Cyclases/chemistry , Adenylyl Cyclases/genetics , Adenylyl Cyclases/metabolism , Angiotensinogen/antagonists & inhibitors , Angiotensinogen/blood , Anti-Obesity Agents/therapeutic use , Atrial Natriuretic Factor/blood , Blood Pressure/drug effects , Cyclic AMP/agonists , Cyclic AMP/blood , Cyclic GMP/agonists , Cyclic GMP/blood , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/immunology , Diabetes Mellitus, Type 2/metabolism , Exenatide , Glucagon-Like Peptide 1/metabolism , Humans , Hypoglycemic Agents/therapeutic use , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Obesity/blood , Obesity/drug therapy , Obesity/immunology , Obesity/metabolism , Oncogene Proteins/genetics , Oncogene Proteins/metabolism , Renin-Angiotensin System/drug effects , Reproducibility of Results , Single-Blind Method , Transforming Growth Factor beta/antagonists & inhibitors , Transforming Growth Factor beta/blood
15.
Diabetes Obes Metab ; 19(9): 1306-1311, 2017 09.
Article in English | MEDLINE | ID: mdl-28304146

ABSTRACT

In view of the occurrence of diabetic ketoacidosis associated with the use of sodium-glucose transport protein-2 inhibitors in patients with type 1 diabetes (T1DM) and the relative absence of this complication in patients treated with liraglutide in spite of reductions in insulin doses, we investigated the effect of liraglutide on ketogenesis. Twenty-six patients with inadequately controlled T1DM were randomly divided into 2 groups of 13 patients each. After an overnight fast, patients were injected, subcutaneously, with either liraglutide 1.8 mg or with placebo. They were maintained on their basal insulin infusion and were followed up in our clinical research unit for 5 hours. The patients injected with placebo maintained their glucose and glucagon concentrations without an increase, but there was a significant increase in free fatty acids (FFA), acetoacetate and ß-hydoxybutyrate concentrations. In contrast, liraglutide significantly reduced the increase in FFA, and totally prevented the increase in acetoacetate and ß-hydroxybutyrate concentrations while suppressing glucagon and ghrelin concentrations. Thus, a single dose of liraglutide is acutely inhibitory to ketogenesis.


Subject(s)
Diabetes Mellitus, Type 1/drug therapy , Glucagon-Like Peptide-1 Receptor/agonists , Glucagon/antagonists & inhibitors , Hypoglycemic Agents/therapeutic use , Ketone Bodies/antagonists & inhibitors , Lipolysis/drug effects , Liraglutide/therapeutic use , Adult , Diabetes Mellitus, Type 1/blood , Diabetes Mellitus, Type 1/metabolism , Double-Blind Method , Drug Resistance , Drug Therapy, Combination , Fatty Acids, Nonesterified/antagonists & inhibitors , Fatty Acids, Nonesterified/blood , Female , Ghrelin/antagonists & inhibitors , Ghrelin/blood , Glucagon/blood , Glucagon-Like Peptide-1 Receptor/metabolism , Humans , Hypoglycemic Agents/administration & dosage , Injections, Subcutaneous , Insulin/administration & dosage , Insulin/therapeutic use , Insulin Infusion Systems , Ketone Bodies/biosynthesis , Ketone Bodies/blood , Liraglutide/administration & dosage , Male , Middle Aged
16.
Clin Endocrinol (Oxf) ; 85(5): 772-780, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27292585

ABSTRACT

CONTEXT: As the syndrome of hypogonadotropic hypogonadism (HH) is associated with anaemia and the administration of testosterone restores haematocrit to normal, we investigated the potential underlying mechanisms. DESIGN: Randomized, double-blind, placebo-controlled trial. METHODS: We measured basal serum concentrations of erythropoietin, iron, iron binding capacity, transferrin (saturated and unsaturated), ferritin and hepcidin and the expression of ferroportin and transferrin receptor (TR) in peripheral blood mononuclear cells (MNC) of 94 men with type 2 diabetes. Forty-four men had HH (defined as subnormal free testosterone along with low or normal LH concentrations) while 50 were eugonadal. Men with HH were randomized to testosterone or placebo treatment every 2 weeks for 15 weeks. Blood samples were collected at baseline, 3 and 15 weeks after starting treatment. Twenty men in testosterone group and 14 men in placebo group completed the study. RESULTS: Haematocrit levels were lower in men with HH (41·1 ± 3·9% vs 43·8 ± 3·4%, P = 0·001). There were no differences in plasma concentrations of hepcidin, ferritin, erythropoietin, transferrin or iron, or in the expression of ferroportin or TR in MNC among HH and eugonadal men. Haematocrit increased to 45·3 ± 4·5%, hepcidin decreased by 28 ± 7% and erythropoietin increased by 21 ± 7% after testosterone therapy (P < 0·05). There was no significant change in ferritin concentrations, but transferrin concentration increased while transferrin saturation and iron concentrations decreased (P < 0·05). Ferroportin and TR mRNA expression in MNC increased by 70 ± 13% and 43 ± 10%, respectively (P < 0·01), after testosterone therapy. CONCLUSIONS: The increase in haematocrit following testosterone therapy is associated with an increase in erythropoietin, the suppression of hepcidin, and an increase in the expression of ferroportin and TR.


Subject(s)
Diabetes Mellitus, Type 2/drug therapy , Ferritins/drug effects , Hepcidins/drug effects , Hypogonadism/drug therapy , Iron/metabolism , Testosterone/pharmacology , Adult , Aged , Cation Transport Proteins/biosynthesis , Cation Transport Proteins/blood , Diabetes Mellitus, Type 2/blood , Double-Blind Method , Erythropoietin/blood , Ferritins/blood , Hematocrit , Hepcidins/blood , Humans , Hypogonadism/blood , Iron/blood , Leukocytes, Mononuclear/metabolism , Male , Middle Aged , Receptors, Transferrin/biosynthesis , Receptors, Transferrin/blood
17.
Am J Physiol Endocrinol Metab ; 308(3): E185-91, 2015 Feb 01.
Article in English | MEDLINE | ID: mdl-25406260

ABSTRACT

This study was conducted to investigate whether a high-fat/high-carbohydrate (HFHC) meal induces an increase in plasma concentrations of glucagon, dipeptidyl peptidase-IV (DPP-IV), and CD26 expression in mononuclear cells (MNC) while reducing insulin, C-peptide, proinsulin, GIP, and GLP-1 concentrations. Ten healthy normal subjects were given either a 910-calorie HFHC meal or an American Heart Association (AHA) meal rich in fruit and fiber during the first visit and the other meal during the second visit in crossover design. Blood samples were collected at baseline and at 15, 30, 45, 60, 75, 90, 120, 180, and 300 min following the meal. There was a significantly greater increase in glucose concentrations and lower increase in postprandial insulin, C-peptide, and proinsulin concentrations and lower insulin/glucose ratios following the HFHC meal. HFHC meal intake induced marked increases in plasma glucagon and DPP-IV concentrations and an increase in CD26 mRNA expression in MNC compared with the AHA meal. In addition, the HFHC meal induced a reduction in GIP and peak GLP-1 secretion compared with the AHA meal. This was associated with a significantly greater increase in oxidative stress and proinflammatory mediators including, ROS generation, TNFα, and IL-1ß mRNA expression and plasma concentrations of TBARS, FFA, and LPS. We conclude that the proinflammatory HFHC meals result in lower insulin, C-peptide, proinsulin, and GIP secretion in association with higher plasma glucagon and DPP-IV concentrations and CD26 expression in MNC compared with the AHA meal.


Subject(s)
Diet, High-Fat , Dietary Fiber/administration & dosage , Fruit , Glucagon/blood , Incretins/blood , Insulin/metabolism , Adult , Dietary Fats/administration & dosage , Down-Regulation/drug effects , Female , Humans , Insulin/blood , Insulin Secretion , Male , Meals , Middle Aged , Up-Regulation/drug effects , Young Adult
18.
World J Diabetes ; 15(2): 133-136, 2024 Feb 15.
Article in English | MEDLINE | ID: mdl-38464377

ABSTRACT

Type 1 diabetes (T1D) is a chronic autoimmune condition that destroys insulin-producing beta cells in the pancreas, leading to insulin deficiency and hyper-glycemia. The management of T1D primarily focuses on exogenous insulin replacement to control blood glucose levels. However, this approach does not address the underlying autoimmune process or prevent the progressive loss of beta cells. Recent research has explored the potential of glucagon-like peptide-1 receptor agonists (GLP-1RAs) as a novel intervention to modify the disease course and delay the onset of T1D. GLP-1RAs are medications initially developed for treating type 2 diabetes. They exert their effects by enhancing glucose-dependent insulin secretion, suppressing glucagon secretion, and slowing gastric emptying. Emerging evidence suggests that GLP-1RAs may also benefit the treatment of newly diagnosed patients with T1D. This article aims to highlight the potential of GLP-1RAs as an intervention to delay the onset of T1D, possibly through their potential immunomodulatory and anti-inflammatory effects and preservation of beta-cells. This article aims to explore the potential of shifting the paradigm of T1D management from reactive insulin replacement to proactive disease modification, which should open new avenues for preventing and treating T1D, improving the quality of life and long-term outcomes for individuals at risk of T1D.

19.
J Investig Med ; 72(5): 487-491, 2024 06.
Article in English | MEDLINE | ID: mdl-38644376

ABSTRACT

Oral calorie intake causes an acute and transient decline in serum testosterone concentrations. It is not known whether this decline occurs in men on testosterone therapy. In this study, we evaluated the change in testosterone concentrations following oral glucose ingestion in hypogonadal men before and after treatment with testosterone therapy. This is a secondary analysis of samples previously collected from a study of hypogonadal men with type 2 diabetes who received testosterone therapy. Study participants (n = 14) ingested 75 grams of oral glucose, and blood samples were collected over 2 h. The test was repeated after 23 weeks of intramuscular testosterone therapy. The mean age and body mass index of study volunteers were 53 ± 8 years and 38 ± 7 kg/m2, respectively. Following glucose intake, testosterone concentrations fell significantly prior to testosterone therapy (week 0, p = 0.04). The nadir of testosterone concentration was at 1 h, followed by recovery to baseline by 2 h. In contrast, there was no change in testosterone concentrations at week 23. The change in serum testosterone concentrations at 60 min was significantly more at week 0 than week 23 (-11 ± 10% vs 0 ± 16%, p = 0.05). We conclude that oral glucose intake has no impact on testosterone concentrations in men on testosterone therapy. Endocrinology societies should consider clarifying in their recommendations that fasting testosterone concentrations are required for the diagnosis of hypogonadism, but not for monitoring testosterone therapy.


Subject(s)
Glucose , Testosterone , Humans , Testosterone/blood , Male , Middle Aged , Glucose/metabolism , Hypogonadism/drug therapy , Hypogonadism/blood , Administration, Oral , Adult
20.
Am J Physiol Endocrinol Metab ; 304(8): E810-8, 2013 Apr 15.
Article in English | MEDLINE | ID: mdl-23403945

ABSTRACT

The purpose of this study was to determine whether an insulin infusion exerts an anti-inflammatory effect and whether the infusion of small amounts of glucose results in oxidative and inflammatory stress in patients with type 1 diabetes. Ten patients with type 1 diabetes were infused with either 2 U/h of insulin with 100 ml 5% dextrose/h to or just dextrose (100 ml/h) or physiological saline (100 ml/h) for 4 h after an overnight fast on three separate days. Blood samples were collected at 0, 2, 4, and 6 h. Insulin with glucose infusion led to the maintenance of euglycemia and a significant suppression of reactive oxygen species (ROS) generation, p47(phox) expression, Toll-like receptor (TLR)-4, TLR-2, TLR-1, CD14, high-mobility group-B1 (HMGB1), p38 mitogen-activated protein (MAP) kinase, c-Jun NH2-terminal kinase (JNK)-1, and platelet/endothelial cell adhesion molecule expression and a fall in serum concentrations of C-reactive protein, HMGB1, and rapid upon activation T cell expressed and secreted. Glucose infusion led to an increase in plasma glucose concentration from 115 (fasting) to 215 (at 4 and 6 h) mg/dl and to an increase in ROS generation, the expression of TLR-4, TLR-2, TLR-1, HMGB1, p38 MAP kinase, and JNK-1, and plasma concentrations of HMGB1. While insulin reduces indexes of oxidative and inflammatory stress in patients with type 1 diabetes, even small amounts of glucose (20 g over 4 h) induce oxidative and inflammatory stress. These effects are reflected in TLR, p38 MAP kinase, and HMGB1 expression. The induction of significant oxidative and inflammatory stress by small amounts of glucose in patients with type 1 diabetes may have important pathophysiological and therapeutic implications.


Subject(s)
Diabetes Mellitus, Type 1/drug therapy , Glucose/administration & dosage , HMGB1 Protein/metabolism , Inflammation/drug therapy , Insulin/administration & dosage , Toll-Like Receptors/metabolism , Adult , Blood Glucose/drug effects , Blood Glucose/metabolism , Cross-Over Studies , Diabetes Mellitus, Type 1/blood , Diabetes Mellitus, Type 1/immunology , Fatty Acids, Nonesterified/blood , Female , Gene Expression/drug effects , Gene Expression/immunology , HMGB1 Protein/genetics , Humans , Hypoglycemic Agents/administration & dosage , Hypoglycemic Agents/blood , Inflammation/blood , Inflammation/immunology , Infusions, Intravenous , Insulin/blood , JNK Mitogen-Activated Protein Kinases/genetics , JNK Mitogen-Activated Protein Kinases/metabolism , Lipopolysaccharide Receptors/genetics , Lipopolysaccharide Receptors/metabolism , Male , Middle Aged , Oxidative Stress/drug effects , Oxidative Stress/immunology , Toll-Like Receptor 1/genetics , Toll-Like Receptor 1/metabolism , Toll-Like Receptor 2/genetics , Toll-Like Receptor 2/metabolism , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/metabolism , Toll-Like Receptors/genetics , p38 Mitogen-Activated Protein Kinases/genetics , p38 Mitogen-Activated Protein Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL