Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Front Pharmacol ; 15: 1302015, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38510652

RESUMO

Background: Elevated levels of somatostatin blunt glucagon counterregulation during hypoglycemia in type 1 diabetes (T1D) and this can be improved using somatostatin receptor 2 (SSTR2) antagonists. Hypoglycemia also occurs in late-stage type 2 diabetes (T2D), particularly when insulin therapy is initiated, but the utility of SSTR2 antagonists in ameliorating hypoglycemia in this disease state is unknown. We examined the efficacy of a single-dose of SSTR2 antagonists in a rodent model of T2D. Methods: High-fat fed (HFF), low dose streptozotocin (STZ, 35 mg/kg)-induced T2D and HFF only, nondiabetic (controls-no STZ) rats were treated with the SSTR2 antagonists ZT-01/PRL-2903 or vehicle (n = 9-11/group) 60 min before an insulin tolerance test (ITT; 2-12 U/kg insulin aspart) or an oral glucose tolerance test (OGTT; 2 g/kg glucose via oral gavage) on separate days. Results: This rodent model of T2D is characterized by higher baseline glucose and HbA1c levels relative to HFF controls. T2D rats also had lower c-peptide levels at baseline and a blunted glucagon counterregulatory response to hypoglycemia when subjected to the ITT. SSTR2 antagonists increased the glucagon response and reduced incidence of hypoglycemia, which was more pronounced with ZT-01 than PRL-2903. ZT-01 treatment in the T2D rats increased glucagon levels above the control response within 60 min of dosing, and values remained elevated during the ITT (glucagon Cmax: 156 ± 50 vs. 77 ± 46 pg/mL, p < 0.01). Hypoglycemia incidence was attenuated with ZT-01 vs. controls (63% vs. 100%) and average time to hypoglycemia onset was also delayed (103.1 ± 24.6 vs. 66.1 ± 23.6 min, p < 0.05). ZT-01 administration at the OGTT onset increased the glucagon response without exacerbating hyperglycemia (2877 ± 806 vs. 2982 ± 781), potentially due to the corresponding increase in c-peptide levels (6251 ± 5463 vs. 14008 ± 5495, p = 0.013). Conclusion: Treatment with SSTR2 antagonists increases glucagon responses in a rat model of T2D and results in less hypoglycemia exposure. Future studies are required to determine the best dosing periods for chronic SSTR2 antagonism treatment in T2D.

2.
Sensors (Basel) ; 23(22)2023 Nov 09.
Artigo em Inglês | MEDLINE | ID: mdl-38005447

RESUMO

The impact of age, sex and body mass index on interstitial glucose levels as measured via continuous glucose monitoring (CGM) during exercise in the healthy population is largely unexplored. We conducted a multivariable generalized estimating equation (GEE) analysis on CGM data (Dexcom G6, 10 days) collected from 119 healthy exercising individuals using CGM with the following specified covariates: age; sex; BMI; exercise type and duration. Females had lower postexercise glycemia as compared with males (92 ± 18 vs. 100 ± 20 mg/dL, p = 0.04) and a greater change in glycemia during exercise from pre- to postexercise (p = 0.001) or from pre-exercise to glucose nadir during exercise (p = 0.009). Younger individuals (i.e., <20 yrs) had higher glucose during exercise as compared with all other age groups (all p < 0.05) and less CGM data in the hypoglycemic range (<70 mg/dL) as compared with those aged 20-39 yrs (p < 0.05). Those who were underweight, based on body mass index (BMI: <18.5 kg/m2), had higher pre-exercise glycemia than the healthy BMI group (104 ± 20 vs. 97 ± 17 mg/dL, p = 0.02) but similar glucose levels after exercise. Resistance exercise was associated with less of a drop in glycemia as compared with aerobic or mixed forms of exercise (p = 0.008) and resulted in a lower percent of time in the hypoglycemic (p = 0.04) or hyperglycemic (glucose > 140 mg/dL) (p = 0.03) ranges. In summary, various factors such as age, sex and exercise type appear to have subtle but potentially important influence on CGM measurements during exercise in healthy individuals.


Assuntos
Hiperglicemia , Hipoglicemia , Masculino , Feminino , Humanos , Glicemia/análise , Índice de Massa Corporal , Automonitorização da Glicemia/métodos , Hipoglicemiantes , Glucose
3.
Diabetes Obes Metab ; 25(6): 1547-1556, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-36734462

RESUMO

AIMS: To examine if glucagon counterregulatory defects exist in a rat model of prediabetes (pre-T2D) and to assess if a selective somatostatin receptor 2 antagonist (SSTR2a), ZT-01, enhances the glucagon response to insulin-induced hypoglycaemia. MATERIALS AND METHODS: Hyperglycaemia was induced in 8- to 9-week-old male, Sprague-Dawley rats via 7 weeks of high-fat diet followed by a single, low-dose intraperitoneal injection of streptozotocin (30 mg/kg). After 2 weeks of basal insulin therapy (0-4 U/d insulin glargine, administered subcutaneously [SC]) to facilitate partial glycaemic recovery and a pre-T2D phenotype, n = 17 pre-T2D and n = 10 normal chow-fed control rats underwent the first of two hypoglycaemic treatment-crossover experiments, separated by a 1-week washout period. On each experimental day, SSTR2a (3 mg/kg ZT-01, SC) or vehicle was administered 1 hour prior to insulin-induced hypoglycaemia (insulin aspart, 6 U/kg, SC). RESULTS: Glucagon counterregulation was marginally reduced with the induction of pre-T2D. Treatment with SSTR2a raised peak plasma glucagon levels and glucagon area under the curve before and after insulin overdose in both and pre-T2D rats. Blood glucose concentration was elevated by 30 minutes after SSTR2a treatment in pre-T2D rats, and hypoglycaemia onset (≤3.9 mmol/L) was delayed by 15 ± 12 minutes compared with vehicle (P < 0.001), despite similar glucose nadirs in the two treatment groups (1.4 ± 0.3 mmol/L). SSTR2a treatment had no effect on blood glucose levels in the control group or on the hypoglycaemia-induced decline in plasma C-peptide levels in either group. CONCLUSIONS: Treatment with an SSTR2a increases glucagon responsiveness and delays the onset of insulin-induced hypoglycaemia in this rat model of pre-T2D where only a modest deficiency in glucagon counterregulation exists.


Assuntos
Diabetes Mellitus Tipo 2 , Hipoglicemia , Estado Pré-Diabético , Masculino , Ratos , Animais , Glucagon , Glicemia , Estado Pré-Diabético/induzido quimicamente , Estado Pré-Diabético/tratamento farmacológico , Ratos Sprague-Dawley , Insulina Aspart , Diabetes Mellitus Tipo 2/tratamento farmacológico
4.
Front Pharmacol ; 14: 1295639, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-38298268

RESUMO

Glucose homeostasis is primarily maintained by pancreatic hormones, insulin and glucagon, with an emerging role for a third islet hormone, somatostatin, in regulating insulin and glucagon responses. Under healthy conditions, somatostatin secreted from pancreatic islet δ-cells inhibits both insulin and glucagon release through somatostatin receptor- induced cAMP-mediated downregulation and paracrine inhibition of ß- and α-cells, respectively. Since glucagon is the body's most important anti-hypoglycemic hormone, and because glucagon counterregulation to hypoglycemia is lost in diabetes, the study of somatostatin biology has led to new investigational medications now in development that may help to restore glucagon counterregulation in type 1 diabetes. This review highlights the normal regulatory role of pancreatic somatostatin signaling in healthy islet function and how the inhibition of somatostatin receptor signaling in pancreatic α-cells may restore normal glucagon counterregulation in diabetes mellitus.

5.
Diabetes Obes Metab ; 24(5): 908-917, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35060297

RESUMO

AIM: To evaluate the pharmacokinetics and efficacy of a novel somatostatin receptor 2 antagonist, ZT-01, to stimulate glucagon release in rats with type 1 diabetes (T1D). METHODS: The pharmacokinetics of ZT-01 and PRL-2903 were assessed following intraperitoneal or subcutaneous dosing at 10 mg/kg. We compared the efficacy of ZT-01 with PRL-2903 to prevent hypoglycaemia during an insulin bolus challenge and under hypoglycaemic clamp conditions. RESULTS: Within 1 hour after intraperitoneal administration, ZT-01 achieved more than 10-fold higher plasma Cmax compared with PRL-2903. Twenty-four hour exposure was 4.7× and 11.3× higher with ZT-01 by the intraperitoneal and subcutaneous routes, respectively. The median time to reach hypoglycaemia of more than 3.0 mmol/L was 60, 70, and 125 minutes following vehicle, PRL-2903, or ZT-01 administration, respectively. Furthermore, rats receiving ZT-01 had significantly higher glucose nadirs following insulin administration compared with PRL-2903- and vehicle-treated rats. During the hypoglycaemic clamp, ZT-01 increased peak glucagon responses by ~4-fold over PRL-2903. CONCLUSIONS: We conclude that ZT-01 may be effective in restoring glucagon responses and preventing the onset of hypoglycaemia in patients with T1D.


Assuntos
Diabetes Mellitus Tipo 1 , Hipoglicemia , Receptores de Somatostatina , Animais , Glicemia , Diabetes Mellitus Tipo 1/tratamento farmacológico , Glucagon , Humanos , Hipoglicemia/induzido quimicamente , Hipoglicemia/prevenção & controle , Insulina , Ratos , Receptores de Somatostatina/antagonistas & inibidores
6.
Endocrinology ; 162(12)2021 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-34477204

RESUMO

Recent antecedent hypoglycemia is a known source of defective glucose counter-regulation in diabetes; the mechanisms perpetuating the cycle of progressive α-cell failure and recurrent hypoglycemia remain unknown. Somatostatin has been shown to suppress the glucagon response to acute hypoglycemia in rodent models of type 1 diabetes. We hypothesized that somatostatin receptor 2 antagonism (SSTR2a) would restore glucagon counterregulation and delay the onset of insulin-induced hypoglycemia in recurrently hypoglycemic, nondiabetic male rats. Healthy, male, Sprague-Dawley rats (n = 39) received bolus injections of insulin (10 U/kg, 8 U/kg, 5 U/kg) on 3 consecutive days to induce hypoglycemia. On day 4, animals were then treated with SSTR2a (10 mg/kg; n = 17) or vehicle (n = 12) 1 hour prior to the induction of hypoglycemia using insulin (5 U/kg). Plasma glucagon level during hypoglycemia was ~30% lower on day 3 (150 ± 75 pg/mL; P < .01), and 68% lower on day 4 in the vehicle group (70 ± 52 pg/mL; P < .001) compared with day 1 (219 ± 99 pg/mL). On day 4, SSTR2a prolonged euglycemia by 25 ± 5 minutes (P < .05) and restored the plasma glucagon response to hypoglycemia. Hepatic glycogen content of SSTR2a-treated rats was 35% lower than vehicle controls after hypoglycemia induction on day 4 (vehicle: 20 ± 7.0 vs SSTR2a: 13 ± 4.4 µmol/g; P < .01). SSTR2a treatment reverses the cumulative glucagon deficit resulting from 3 days of antecedent hypoglycemia in healthy rats. This reversal is associated with decreased hepatic glycogen content and delayed time to hypoglycemic onset. We conclude that recurrent hypoglycemia produces glucagon counterregulatory deficiency in healthy male rats, which can be improved by SSTR2a.


Assuntos
Glucagon/metabolismo , Hipoglicemia/metabolismo , Peptídeos Cíclicos/farmacologia , Animais , Glicemia/efeitos dos fármacos , Glicemia/metabolismo , Glucagon/efeitos dos fármacos , Glucose/metabolismo , Antagonistas de Hormônios/farmacologia , Hipoglicemia/patologia , Glicogênio Hepático/metabolismo , Masculino , Ratos , Ratos Sprague-Dawley , Receptores de Somatostatina/antagonistas & inibidores , Recidiva
7.
Diabetes Care ; 44(2): 610-613, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33328284

RESUMO

OBJECTIVE: Exercising while fasted with type 1 diabetes facilitates weight loss; however, the best strategy to maintain glucose stability remains unclear. RESEARCH DESIGN AND METHODS: Fifteen adults on continuous subcutaneous insulin infusion completed three sessions of fasted walking (120 min at 45% VO2max) in a randomized crossover design: 50% basal rate reduction, set 90 min pre-exercise (-90min50%BRR); usual basal rate with carbohydrate intake of 0.3 g/kg/h (CHO-only); and combined 50% basal rate reduction set at exercise onset with carbohydrate intake of 0.3 g/kg/h (Combo). RESULTS: Combo had a smaller change in glucose (5 ± 47 mg/dL) versus CHO-only (-49 ± 61 mg/dL, P = 0.03) or -90min50%BRR (-34 ± 45 mg/dL). The -90min50%BRR strategy produced higher ß-hydroxybutyrate levels (0.4 ± 0.3 vs. 0.1 ± 0.1 mmol/L) and greater fat oxidation (0.51 ± 0.2 vs. 0.39 ± 0.1 g/min) than CHO-only (both P < 0.05). CONCLUSIONS: All strategies examined produced stable glycemia for fasted exercise, but a 50% basal rate reduction, set 90 min pre-exercise, eliminates carbohydrate needs and enhances fat oxidation better than carbohydrate feeding with or without a basal rate reduction set at exercise onset.


Assuntos
Diabetes Mellitus Tipo 1 , Adulto , Glicemia , Estudos Cross-Over , Diabetes Mellitus Tipo 1/tratamento farmacológico , Carboidratos da Dieta , Exercício Físico , Humanos , Insulina/uso terapêutico , Sistemas de Infusão de Insulina
8.
Stem Cells ; 35(5): 1378-1391, 2017 05.
Artigo em Inglês | MEDLINE | ID: mdl-28233396

RESUMO

We show that the transcriptional corepressor p107 orchestrates a metabolic checkpoint that determines adipocyte lineage fates for non-committed progenitors. p107 accomplishes this when stem cell commitment would normally occur in growth arrested cells. p107-deficient embryonic progenitors are characterized by a metabolic state resembling aerobic glycolysis that is necessary for their pro-thermogenic fate. Indeed, during growth arrest they have a reduced capacity for NADH partitioning between the cytoplasm and mitochondria. Intriguingly, this occurred despite an increase in the capacity for mitochondrial oxidation of non-glucose substrates. The significance of metabolic reprogramming is underscored by the disruption of glycolytic capacities in p107-depleted progenitors that reverted their fates from pro-thermogenic to white adipocytes. Moreover, the manipulation of glycolytic capacity on nonspecified embryonic and adult progenitors forced their beige fat commitment. These innovative findings introduce a new approach to increase pro-thermogenic adipocytes based on simply promoting aerobic glycolysis to manipulate nonspecified progenitor fate decisions. Stem Cells 2017;35:1378-1391.


Assuntos
Adipócitos Marrons/citologia , Adipócitos Brancos/citologia , Pontos de Checagem do Ciclo Celular , Linhagem da Célula , Proteína p107 Retinoblastoma-Like/metabolismo , Aerobiose , Animais , Linhagem Celular , Núcleo Celular/metabolismo , Embrião de Mamíferos/citologia , Técnicas de Silenciamento de Genes , Glicólise , Camundongos Knockout , Modelos Biológicos , Oxirredução , Células-Tronco/citologia , Células-Tronco/metabolismo , Frações Subcelulares/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...