Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 224
Filtrar
Más filtros

País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Osteoporos Int ; 32(9): 1837-1848, 2021 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-33594488

RESUMEN

Some studies indicate potential beneficial effects of metformin on body composition and bone. This trial compared metformin + insulin vs placebo + insulin. Metformin treatment had a small but positive effect on bone quality in the peripheral skeleton, reduced weight gain, and resulted in a more beneficial body composition compared with placebo in insulin-treated patients with type 2 diabetes. INTRODUCTION: Glucose-lowering medications affect body composition. We assessed the long-term effects of metformin compared with placebo on whole body bone and body composition measures in patients with type 2 diabetes mellitus. METHODS: This was a sub-study of the Copenhagen Insulin and Metformin Therapy trial, which was a double-blinded randomized placebo-controlled trial assessing 18-month treatment with metformin compared with placebo, in combination with different insulin regimens in patients with type 2 diabetes mellitus (T2DM). The sub-study evaluates the effects on bone mineral content (BMC), density (BMD), and body composition from whole body dual-energy X-ray absorptiometry (DXA) scans which were assessed at baseline and after 18 months. RESULTS: Metformin had a small, but positive, (p < 0.05) effect on subtotal, appendicular, and legs BMC and BMD compared with placebo. After adjustment for sex, age, vitamin D, smoking, BMI, T2DM duration, HbA1c, and insulin dose, the effects on appendicular BMC and BMD persisted (p < 0.05 for both). The changes in appendicular BMC and BMD corresponded approximately to a 0.7% and 0.5% increase in the metformin group and 0.4% and 0.4% decrease in the placebo group, respectively. These effects were mostly driven by an increase in BMC and BMD in the legs and a loss of BMC and BMD in the arms. During 18 months, all participants increased in weight, fat mass (FM), FM%, and lean mass (LM), but decreased in LM%. The metformin group increased less in weight (subtotal weight (weight-head) - 2.4 [- 3.5, - 1.4] kg, p value < 0.001) and FM (- 1.5 [- 2.3, - 0.8] kg, p value < 0.001) and decreased less in LM% (0.6 [0.2, 1.1] %, p value < 0.001) compared with the placebo group. CONCLUSION: Metformin treatment had a small positive effect on BMC and BMD in the peripheral skeleton and reduced weight gain compared with placebo in insulin-treated patients with T2DM.


Asunto(s)
Diabetes Mellitus Tipo 2 , Metformina , Composición Corporal , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Humanos , Insulina , Metformina/uso terapéutico , Sobrepeso
2.
Osteoporos Int ; 29(11): 2517-2526, 2018 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-30027438

RESUMEN

Some antihyperglycemic medications have been found to affect bone metabolism. We assessed the long-term effects of metformin compared with placebo on bone mineral density (BMD) and trabecular bone score (TBS) in patients with type 2 diabetes. Metformin had no significant effect on BMD in the spine and hip or TBS compared with a placebo. INTRODUCTION: Patients with type 2 diabetes mellitus (T2DM) have an increased risk of fractures despite a high bone mass. Some antihyperglycemic medications have been found to affect bone metabolism. We assessed the long-term effects of metformin compared with placebo on bone mineral density (BMD) and trabecular bone score (TBS). METHODS: This was a sub-study of a multicenter, randomized, 18-month placebo-controlled, double-blinded trial with metformin vs. placebo in combination with different insulin regimens (the Copenhagen Insulin and Metformin Therapy trial) in patients with T2DM. BMD in the spine and hip and TBS in the spine were assessed by dual-energy X-ray absorptiometry at baseline and after 18 months follow-up. RESULTS: Four hundred seven patients were included in this sub-study. There were no between-group differences in BMD or TBS. From baseline to 18 months, TBS decreased significantly in both groups (metformin group, - 0.041 [- 0.055, - 0.027]; placebo group - 0.046 [- 0.058, - 0.034]; both p < 0.001). BMD in the spine and total hip did not change significantly from baseline to 18 months. After adjustments for gender, age, vitamin D, smoking, BMI, duration of T2DM, HbA1c, and insulin dose, the TBS between-group differences increased but remained non-significant. HbA1c was negatively associated with TBS (p = 0.009) as was longer duration of diabetes, with the femoral neck BMD (p = 0.003). Body mass index had a positive effect on the hip and femoral neck BMD (p < 0.001, p = 0.045, respectively). CONCLUSIONS: Eighteen months of treatment with metformin had no significant effect on BMD in the spine and hip or TBS in patients with T2DM compared with a placebo. TBS decreased significantly in both groups. TRIAL REGISTRATION: ClinicalTrials.gov (NCT00657943).


Asunto(s)
Densidad Ósea/efectos de los fármacos , Diabetes Mellitus Tipo 2/fisiopatología , Hipoglucemiantes/farmacología , Insulina/farmacología , Metformina/farmacología , Adulto , Anciano , Hueso Esponjoso/efectos de los fármacos , Hueso Esponjoso/fisiopatología , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Método Doble Ciego , Quimioterapia Combinada , Femenino , Cuello Femoral/fisiopatología , Articulación de la Cadera/fisiopatología , Humanos , Hipoglucemiantes/efectos adversos , Hipoglucemiantes/uso terapéutico , Insulina/efectos adversos , Insulina/uso terapéutico , Vértebras Lumbares/fisiopatología , Masculino , Metformina/efectos adversos , Metformina/uso terapéutico , Persona de Mediana Edad , Fracturas Osteoporóticas/inducido químicamente
3.
Clin Endocrinol (Oxf) ; 86(1): 156-159, 2017 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-27649688

RESUMEN

OBJECTIVE: The positive metabolic outcome of Roux-en-Y gastric bypass (RYGB) surgery may involve fibroblast growth factor 21 (FGF21), in both the fasting state and postprandially. We measured the fasting levels of FGF21 before and after bariatric surgery as well as the postprandial FGF21 responses after a glucose load and after a mixed meal. DESIGN: Observational intervention trial. PATIENTS AND MEASUREMENTS: Eight obese, nondiabetic patients underwent RYGB. Plasma FGF21 was measured both before and after surgery on three different days during oral glucose loads (25 g or 50 g glucose) or a mixed meal. Blood samples were taken right before the meal and at 15-min intervals until 90 min and at 150 min and 210 min relative to the start of the meal. RESULTS: Overall, fasting plasma FGF21 did not change significantly before and after surgery (262 ± 71 vs 411 ± 119 pg/ml), but for three subjects, fasting plasma FGF21 increased significantly after surgery. Furthermore, FGF21 levels increased significantly at t = 90 and t = 150 min in response to 50 g glucose, but not after a mixed meal. CONCLUSIONS: In conclusion, the observed increase in postprandial plasma FGF21 in response to glucose and the lack of FGF21 response to a mixed meal may have important implications for the physiologic role of FGF21. The increase in postprandial FGF21 in response to glucose in the early postoperative period may contribute to the metabolic improvements observed after gastric bypass.


Asunto(s)
Factores de Crecimiento de Fibroblastos/sangre , Derivación Gástrica , Obesidad/sangre , Adulto , Femenino , Prueba de Tolerancia a la Glucosa , Humanos , Masculino , Periodo Posprandial
4.
Diabet Med ; 34(12): 1783-1787, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-28782840

RESUMEN

BACKGROUND: Roux-en-Y gastric bypass is associated with an increased risk of postprandial hyperinsulinaemic hypoglycaemia, but the underlying pathophysiology remains poorly understood. We therefore examined the effect of re-routing of nutrient delivery on gut-islet cell crosstalk in a person with severe postprandial hypoglycaemia after Roux-en-Y gastric bypass. CASE REPORT: A person with severe postprandial hypoglycaemia, who underwent surgical reversal of Roux-en-Y gastric bypass, was studied before reversal and at 2 weeks and 3 months after reversal surgery using liquid mixed meal tests and hyperinsulinaemic-euglycaemic clamps. The nadir of postprandial plasma glucose rose from 2.8 mmol/l to 4.1 mmol/l at 2 weeks and to 4.4 mmol/l at 3 months after reversal. Concomitant insulin- and glucagon-like peptide-1 secretion (peak concentrations and area under the curve) clearly decreased after reversal, while concentrations of glucose-dependent insulinotropic polypeptide and ghrelin increased. Insulin clearance declined after reversal, whereas clamp-estimated peripheral insulin sensitivity was unchanged. The person remained without symptoms of hypoglycaemia, but had experienced significant weight gain at 15-month follow-up. DISCUSSION: Accelerated nutrient absorption may be a driving force behind postprandial hyperinsulinaemic hypoglycaemia after Roux-en-Y gastric bypass. Re-routing of nutrients by reversal of the Roux-en-Y gastric bypass diminished postprandial plasma glucose excursions, alleviated postprandial insulin and glucagon-like peptide-1 hypersecretion and eliminated postprandial hypoglycaemia, which emphasizes the importance of altered gut-islet cell crosstalk for glucose metabolism after Roux-en-Y gastric bypass.


Asunto(s)
Derivación Gástrica , Tránsito Gastrointestinal/fisiología , Hipoglucemia/rehabilitación , Hipoglucemia/cirugía , Islotes Pancreáticos/fisiología , Reoperación/rehabilitación , Glucemia/metabolismo , Alimentos , Derivación Gástrica/efectos adversos , Técnica de Clampeo de la Glucosa , Humanos , Hipoglucemia/patología , Hipoglucemia/fisiopatología , Intestinos/fisiología , Intestinos/cirugía , Islotes Pancreáticos/metabolismo , Masculino , Obesidad Mórbida/metabolismo , Obesidad Mórbida/cirugía , Periodo Posprandial , Índice de Severidad de la Enfermedad , Factores de Tiempo , Adulto Joven
5.
Int J Obes (Lond) ; 40(11): 1699-1706, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27434221

RESUMEN

BACKGROUND/OBJECTIVES: Exaggerated postprandial secretion of glucagon-like peptide-1 (GLP-1) and peptide YY (PYY) may explain appetite reduction and weight loss after Roux-en-Y gastric bypass (RYGB), but causality has not been established. We hypothesized that food intake decreases after surgery through combined actions from GLP-1 and PYY. GLP-1 actions can be blocked using the GLP-1 receptor antagonist Exendin 9-39 (Ex-9), whereas PYY actions can be inhibited by the administration of a dipeptidyl peptidase-4 (DPP-4) inhibitor preventing the formation of PYY3-36. SUBJECTS/METHODS: Appetite-regulating gut hormones and appetite ratings during a standard mixed-meal test and effects on subsequent ad libitum food intake were evaluated in two studies: in study 1, nine patients with type 2 diabetes were examined prospectively before and 3 months after RYGB with and without Ex-9. In study 2, 12 RYGB-operated patients were examined in a randomized, placebo-controlled, crossover design on four experimental days with: (1) placebo, (2) Ex-9, (3) the DPP-4 inhibitor, sitagliptin, to reduce formation of PYY3-36 and (4) Ex-9/sitagliptin combined. RESULTS: In study 1, food intake decreased by 35% following RYGB compared with before surgery. Before surgery, GLP-1 receptor blockage increased food intake but no effect was seen postoperatively, whereas PYY secretion was markedly increased. In study 2, combined GLP-1 receptor blockage and DPP-4 inhibitor mediated lowering of PYY3-36 increased food intake by ~20% in RYGB patients, whereas neither GLP-1 receptor blockage nor DPP-4 inhibition alone affected food intake, perhaps because of concomitant marked increases in the unblocked hormone. CONCLUSIONS: Blockade of actions from only one of the two L-cell hormones, GLP-1 and PYY3-36, resulted in concomitant increased secretion of the other, probably explaining the absent effect on food intake on these experimental days. Combined blockade of GLP-1 and PYY actions increased food intake after RYGB, supporting that these hormones have a role in decreased food intake postoperatively.


Asunto(s)
Regulación del Apetito/fisiología , Ingestión de Alimentos/fisiología , Derivación Gástrica , Péptido 1 Similar al Glucagón/metabolismo , Obesidad Mórbida/cirugía , Péptido YY/metabolismo , Apetito/fisiología , Estudios Cruzados , Dinamarca , Diabetes Mellitus Tipo 2/sangre , Diabetes Mellitus Tipo 2/prevención & control , Femenino , Péptido 1 Similar al Glucagón/sangre , Receptor del Péptido 1 Similar al Glucagón/antagonistas & inhibidores , Humanos , Masculino , Obesidad Mórbida/sangre , Fragmentos de Péptidos/uso terapéutico , Péptido YY/sangre , Resultado del Tratamiento , Pérdida de Peso
6.
Int J Obes (Lond) ; 40(2): 281-90, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26303352

RESUMEN

OBJECTIVES: Increased energy expenditure (EE) has been proposed as an important mechanism for weight loss following Roux-en-Y gastric bypass (RYGB). However, this has never been investigated in a controlled setting independent of changes in energy balance. Similarly, only few studies have investigated the effect of RYGB on glycaemic control per se. Here, we investigated the effect of RYGB on EE, appetite, glycaemic control and specific signalling molecules compared with a control group in comparable negative energy balance. SUBJECTS/METHODS: Obese normal glucose-tolerant participants were randomized to receive RYGB after 8 (n=14) or 12 weeks (n=14). The protocol included a visit at week 0 and three visits (weeks 7, 11 and 78) where 24-h EE, appetite and blood parameters were assessed. Participants followed a low-calorie diet from weeks 0-11, with those operated at week 12 serving as a control group for those operated at week 8. RESULTS: Compared with controls, RYGB-operated participants had lower body composition-adjusted 24-h EE and basal EE 3 weeks postoperatively (both P<0.05) but EE parameters at week 78 were not different from preoperative values (week 7). Surgery changed the postprandial response of glucagon-like peptide-1 (GLP-1), peptide YY3-36 (PYY), ghrelin, cholecystokinin, fibroblast growth factor-19 and bile acids (all P<0.05). Particularly, increases in GLP-1, PYY and decreases in ghrelin were associated with decreased appetite. None of HOMA-IR (homeostasis model assessment-estimated insulin resistance), Matsuda index, the insulinogenic index, the disposition index and fasting hepatic insulin clearance were different between the groups, but RYGB operated had lower fasting glucose (P<0.05) and the postprandial glucose profile was shifted to the left (P<0.01). CONCLUSIONS: Our data do not support that EE is increased after RYGB. More likely, RYGB promotes weight loss by reducing appetite, partly mediated by changes in gastrointestinal hormone secretion. Furthermore, we found that the early changes in glycaemic control after RYGB is to a large extent mediated by caloric restriction.


Asunto(s)
Apetito/fisiología , Glucemia/metabolismo , Metabolismo Energético/fisiología , Derivación Gástrica , Ghrelina/metabolismo , Obesidad Mórbida/cirugía , Pérdida de Peso , Adulto , Índice de Masa Corporal , Dinamarca/epidemiología , Femenino , Estudios de Seguimiento , Humanos , Resistencia a la Insulina , Masculino , Obesidad Mórbida/epidemiología , Obesidad Mórbida/metabolismo , Periodo Posprandial , Resultado del Tratamiento
7.
Diabetes Obes Metab ; 18(4): 410-8, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26743775

RESUMEN

AIM: To investigate the dose-response relationship of subcutaneous (s.c.) glucagon administration on plasma glucose and on counter-regulatory hormone responses during s.c. insulin-induced mild hypoglycaemia in patients with type 1 diabetes treated with insulin pumps. METHODS: Eight insulin pump-treated patients completed a blinded, randomized, placebo-controlled study. Hypoglycaemia was induced in the fasting state by an s.c. insulin bolus and, when plasma glucose reached 3.4 mmol/l [95% confidence interval (CI) 3.2-3.5], an s.c. bolus of either 100, 200, 300 µg glucagon or saline was administered. Plasma glucose, counter-regulatory hormones, haemodynamic variables and side effects were measured throughout each study day. Peak plasma glucose level was the primary endpoint. RESULTS: Plasma glucose level increased significantly by a mean (95% CI) of 2.3 (1.7-3.0), 4.2 (3.5-4.8) and 5.0 (4.3-5.6) mmol/l to 6.1 (4.9-7.4), 7.9 (6.4-9.3) and 8.7 (7.8-9.5) vs 3.6 (3.4-3.9) mmol/l (p < 0.001) after the three different glucagon doses as compared with saline, and the increase was neither correlated with weight nor insulin levels. Area under the plasma glucose curve, peak plasma glucose, time to peak plasma glucose and duration of plasma glucose level above baseline were significantly enhanced with increasing glucagon doses; however, these were not significantly different between 200 and 300 µg glucagon. Free fatty acids and heart rates were significantly lower initially after glucagon than after saline injection. Other haemodynamic variables, counter-regulatory hormones and side effects did not differ between interventions. CONCLUSIONS: An s.c. low-dose glucagon bolus effectively restores plasma glucose after insulin overdosing. Further research is needed to investigate whether low-dose glucagon may be an alternative treatment to oral carbohydrate intake for mild hypoglycaemia in patients with type 1 diabetes.


Asunto(s)
Diabetes Mellitus Tipo 1/tratamiento farmacológico , Glucagón/administración & dosificación , Hipoglucemia/prevención & control , Hipoglucemiantes/efectos adversos , Antagonistas de Insulina/administración & dosificación , Insulina Aspart/efectos adversos , Adulto , Anciano , Glucemia/análisis , Péptido C/sangre , Diabetes Mellitus Tipo 1/sangre , Relación Dosis-Respuesta a Droga , Femenino , Glucagón/efectos adversos , Glucagón/farmacocinética , Glucagón/uso terapéutico , Humanos , Hipoglucemia/inducido químicamente , Hipoglucemia/fisiopatología , Hipoglucemiantes/administración & dosificación , Hipoglucemiantes/farmacocinética , Hipoglucemiantes/uso terapéutico , Inyecciones Subcutáneas , Antagonistas de Insulina/efectos adversos , Antagonistas de Insulina/farmacocinética , Antagonistas de Insulina/uso terapéutico , Insulina Aspart/administración & dosificación , Insulina Aspart/farmacocinética , Insulina Aspart/uso terapéutico , Sistemas de Infusión de Insulina , Masculino , Persona de Mediana Edad , Índice de Severidad de la Enfermedad , Método Simple Ciego , Adulto Joven
8.
Int J Clin Pract ; 70(8): 657-67, 2016 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-27384031

RESUMEN

AIMS: To provide a review of the available data and practical use of insulin degludec with insulin aspart (IDegAsp). Premixed insulins provide basal and prandial glucose control; however, they have an intermediate-acting prandial insulin component and do not provide as effective basal coverage as true long-acting insulins, owing to the physicochemical incompatibility of their individual components, coupled with the inflexibility of adjustment. The molecular structure of the co-formulation of IDegAsp, a novel insulin preparation, allows these two molecules to coexist without affecting their individual pharmacodynamic profiles. METHODS: Clinical evidence in phase 2/3 trials of IDegAsp efficacy and safety in type 1 and type 2 diabetes mellitus (T1DM and T2DM) have been assessed and summarised. RESULTS: In people with T2DM, once- and twice-daily dosing provides similar overall glycaemic control (HbA1c ) to current modern insulins, but with lower risk of nocturnal hypoglycaemia. In prior insulin users, glycaemic control was achieved with lower or equal insulin doses vs. other basal+meal-time or premix insulin regimens. In insulin-naïve patients with T2DM, IDegAsp can be started once or twice-daily, based on individual need. People switching from more than once-daily basal or premix insulin therapy can be converted unit-to-unit to once-daily IDegAsp, although this strategy should be assessed by the physician on an individual basis. CONCLUSIONS: IDegAsp offers physicians and people with T2DM a simpler insulin regimen than other available basal-bolus or premix-based insulin regimens, with stable daytime basal coverage, a lower rate of hypoglycaemia and some flexibility in injection timing compared with premix insulins.


Asunto(s)
Diabetes Mellitus Tipo 1/tratamiento farmacológico , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Hipoglucemiantes/uso terapéutico , Insulina Aspart/administración & dosificación , Insulina de Acción Prolongada/administración & dosificación , Glucemia , Ensayos Clínicos Fase II como Asunto , Ensayos Clínicos Fase III como Asunto , Esquema de Medicación , Sustitución de Medicamentos , Humanos , Hipoglucemiantes/efectos adversos , Hipoglucemiantes/farmacología , Insulina Aspart/efectos adversos , Insulina Aspart/farmacología , Insulina de Acción Prolongada/efectos adversos , Insulina de Acción Prolongada/farmacología , Resultado del Tratamiento
9.
Int J Obes (Lond) ; 39(5): 834-41, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25287751

RESUMEN

BACKGROUND: Recent studies indicate that glucagon-like peptide (GLP)-1 inhibits appetite in part through regulation of soluble leptin receptors. Thus, during weight loss maintenance, GLP-1 receptor agonist (GLP-1RA) administration may inhibit weight loss-induced increases in soluble leptin receptors thereby preserving free leptin levels and preventing weight regain. METHODS: In a randomized controlled trial, 52 healthy obese individuals were, after a diet-induced 12% body weight loss, randomized to treatment with or without administration of the GLP-1RA liraglutide (1.2 mg per day). In case of weight gain, low-calorie diet products were allowed to replace up to two meals per day to achieve equal weight maintenance. Glucose tolerance and hormone responses were investigated before and after weight loss and after 52 weeks weight maintenance. Primary end points: increase in soluble leptin receptor plasma levels and decrease in free leptin index after 52 weeks weight loss maintenance. RESULTS: Soluble leptin receptor increase was 59% lower; 2.1±0.7 vs 5.1±0.8 ng ml(-1) (-3.0 (95% confidence interval (CI)=-0.5 to -5.5)), P<0.001 and free leptin index decrease was 43% smaller; -62±15 vs -109±20 (-47 (95% CI=-11 to -83)), P<0.05 with administration of GLP-1RA compared with control group. The 12% weight loss was successfully maintained in both the groups with no significant change in weight after 52 weeks follow-up. The GLP-1RA group had greater weight loss during the weight maintenance period (-2.3 kg (95% CI=-0.6 to -4.0)), and had fewer meal replacements per day compared with the control group (minus one meal per day (95% CI=-0.6 to -1)), P<0.001. Fasting glucose was decreased by an additional -0.2±0.1 mmol l(-1) in the GLP-1RA group in contrast to the control group, where glucose increased 0.3±0.1 mmol l(-1) to the level before weight loss (-0.5mmol l(-1) (95% CI=-0.1 to -0.9)), P<0.005. Meal response of peptide PYY3-36 was higher at week 52 in the GLP-1RA group compared with the control group, P<0.05. CONCLUSIONS: The weight maintaining effect of GLP-1RAs may be mediated by smaller decrease in free leptin and higher PYY3-36 response. Low dose GLP-1RA therapy maintained 12% weight loss for 1 year and may prevent pre-diabetes in obesity.


Asunto(s)
Restricción Calórica , Receptor del Péptido 1 Similar al Glucagón/agonistas , Incretinas/administración & dosificación , Leptina/sangre , Liraglutida/administración & dosificación , Obesidad/tratamiento farmacológico , Estado Prediabético/prevención & control , Pérdida de Peso/efectos de los fármacos , Adulto , Apetito/efectos de los fármacos , Índice de Masa Corporal , Dinamarca , Femenino , Humanos , Masculino , Obesidad/sangre , Obesidad/dietoterapia , Estado Prediabético/sangre , Resultado del Tratamiento
10.
Diabetes Metab Res Rev ; 31(7): 672-9, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25533565

RESUMEN

Intensive insulin treatment in type 1 diabetes reduces the incidence and slows the progression of microvascular and macrovascular complications; however, it is associated with an increased risk of hypoglycaemia and weight gain. In this review, we propose dual-hormone treatment with insulin and glucagon as a method for achieving near normalization of blood glucose levels without increasing hypoglycaemia frequency and weight gain. We briefly summarize glucagon pathophysiology in type 1 diabetes as well as the current applications of glucagon for the treatment of hypoglycaemia. Until now, the use of glucagon has been limited by the need for reconstitution immediately before use, because of instability of the available compounds; however, stabile compounds are soon to be launched and will render long-term intensive dual-hormone treatment in type 1 diabetes possible.


Asunto(s)
Diabetes Mellitus Tipo 1/tratamiento farmacológico , Glucagón/uso terapéutico , Terapia de Reemplazo de Hormonas/métodos , Hormonas/uso terapéutico , Hipoglucemiantes/uso terapéutico , Insulina/uso terapéutico , Quimioterapia Combinada , Humanos , Hipoglucemia/inducido químicamente , Hipoglucemia/prevención & control , Aumento de Peso
11.
Diabetes Obes Metab ; 17(1): 74-81, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25243647

RESUMEN

AIMS: To examine whether 12 weeks of treatment with a dipeptidyl peptidase-4 (DPP-4) inhibitor, sitagliptin, influences the insulin secretion induced by glucose, glucose-dependent insulinotropic polypeptide (GIP) and glucagon-like peptide-1 (GLP-1) during a hyperglycaemic clamp in patients with type 2 diabetes (T2DM). METHODS: A randomized, double-blind, placebo-controlled study was conducted over 12 weeks, during which 25 patients with T2DM completed treatment with either sitagliptin (100 mg once daily) or placebo as add-on therapy to metformin [sitagliptin group (n = 12): mean ± standard error of the mean (s.e.m.) age 54 ± 2.5 years, mean ± s.e.m. HbA1c 7.8 ± 0.2%; placebo group (n = 13): mean ± s.e.m. age: 57 ± 3.0 years, mean ± s.e.m. HbA1c 7.9 ± 0.2 %]. In weeks 1 and 12, the patients underwent three 2-h 15-mM hyperglycaemic clamp experiments with infusion of either saline, GLP-1 or GIP. ß-cell function was evaluated according to first-phase, second-phase, incremental and total insulin and C-peptide responses. RESULTS: In the sitagliptin group, the mean HbA1c concentration was significantly reduced by 0.9% (p = 0.01). The total ß-cell response during GIP infusion improved significantly from week 1 to week 12, both within the sitagliptin group (p = 0.004) and when compared with the placebo group (p = 0.04). The total ß-cell response during GLP-1 infusion was significantly higher (p = 0.001) when compared with saline and GIP infusion, but with no improvement from week 1 to week 12. No significant changes in ß-cell function occurred in the placebo group. CONCLUSIONS: Treatment with the DPP-4 inhibitor sitagliptin over 12 weeks in patients with T2DM partially restored the lost insulinotropic effect of GIP, whereas the preserved insulinotropic effect of GLP-1 was not further improved. A gradual enhancement of the insulinotropic effect of GIP, therefore, possibly contributes to the antidiabetic actions of DPP-4 inhibitors.


Asunto(s)
Diabetes Mellitus Tipo 2/tratamiento farmacológico , Inhibidores de la Dipeptidil-Peptidasa IV/uso terapéutico , Polipéptido Inhibidor Gástrico/metabolismo , Células Secretoras de Insulina/efectos de los fármacos , Insulina/metabolismo , Pirazinas/uso terapéutico , Triazoles/uso terapéutico , Regulación hacia Arriba/efectos de los fármacos , Péptido C/sangre , Diabetes Mellitus Tipo 2/sangre , Diabetes Mellitus Tipo 2/metabolismo , Inhibidores de la Dipeptidil-Peptidasa IV/efectos adversos , Método Doble Ciego , Quimioterapia Combinada/efectos adversos , Femenino , Péptido 1 Similar al Glucagón/metabolismo , Técnica de Clampeo de la Glucosa , Hemoglobina Glucada/análisis , Humanos , Hiperglucemia/prevención & control , Hipoglucemiantes/uso terapéutico , Insulina/sangre , Secreción de Insulina , Células Secretoras de Insulina/metabolismo , Masculino , Metformina/uso terapéutico , Persona de Mediana Edad , Pirazinas/efectos adversos , Fosfato de Sitagliptina , Triazoles/efectos adversos
12.
Diabet Med ; 31(11): 1293-300, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25112609

RESUMEN

AIM: To review and discuss the results from the clinical controlled trials comparing a dipeptidyl peptidase-4 inhibitor with placebo treatment as add-on to insulin treatment with respect to changes in HbA1c , weight, fasting plasma glucose, risk of hypoglycaemia and safety in patients with Type 2 diabetes. METHODS: We searched the MEDLINE and PubMed databases to identify all randomized controlled clinical trials evaluating dipeptidyl peptidase-4 inhibitors as an add-on to insulin in patients with Type 2 diabetes, which were selected for review. The abstracts and posters of the recent annual meetings of the American Diabetes Association and European Association for the Study of Diabetes were hand searched, as were the reference lists of articles identified. RESULTS: Adding a dipeptidyl peptidase-4 inhibitor to insulin treatment resulted in a glucose-lowering effect of ~ 6.6-8.7 mmol/mol (0.60-0.80%) from a baseline HbA1c of 67-78 mmol/mol (8.3-9.3%), without increasing the risk of hypoglycaemia. The dipeptidyl peptidase-4 inhibitor treatment had no effect on body weight or daily dose of insulin. The frequency and severity of adverse events did not differ between dipeptidyl peptidase-4 inhibitor and placebo treatment. CONCLUSION: Adding a dipeptidyl peptidase-4 inhibitor treatment to insulin has a moderate effect on HbA1c , a weight-neutral effect and a good safety profile. The risk of hypoglycaemia is not increased despite a significant improvement in HbA1c .


Asunto(s)
Diabetes Mellitus Tipo 2/tratamiento farmacológico , Inhibidores de la Dipeptidil-Peptidasa IV/uso terapéutico , Hipoglucemiantes/uso terapéutico , Insulina/uso terapéutico , Peso Corporal/efectos de los fármacos , Diabetes Mellitus Tipo 2/sangre , Inhibidores de la Dipeptidil-Peptidasa IV/efectos adversos , Quimioterapia Combinada/efectos adversos , Humanos , Hiperglucemia/prevención & control , Hipoglucemia/inducido químicamente , Hipoglucemia/prevención & control , Hipoglucemiantes/efectos adversos , Insulina/efectos adversos , Ensayos Clínicos Controlados Aleatorios como Asunto
13.
Diabetes Obes Metab ; 16(1): 9-21, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23617798

RESUMEN

The hormone glucagon-like peptide-1 (GLP-1) is released from the gut in response to food intake. It acts as a satiety signal, leading to reduced food intake, and also as a regulator of gastric emptying. Furthermore, GLP-1 functions as an incretin hormone, stimulating insulin release and inhibiting glucagon secretion from the pancreas in response to food ingestion. Evidence suggests that the action or effect of GLP-1 may be impaired in obese subjects, even in those with normal glucose tolerance. GLP-1 impairment may help explain the increased gastric emptying and decreased satiety signalling seen in obesity. Incretin impairment, probably associated with reduced insulinotropic potency of GLP-1, is also characteristic of type 2 diabetes (T2D). Therefore, it is possible that incretin impairment may contribute to the pathophysiological bridge between obesity and T2D. This review summarises current knowledge about the pathophysiology and consequences of GLP-1 and incretin impairment in obesity, and examines the evidence for an incretin-related link between obesity and T2D. It also considers the current literature surrounding the novel use of GLP-1 receptor agonists as a treatment for obesity in patients with normoglycaemia, prediabetes and T2D.


Asunto(s)
Diabetes Mellitus Tipo 2/metabolismo , Péptido 1 Similar al Glucagón/metabolismo , Incretinas/metabolismo , Insulina/metabolismo , Obesidad/metabolismo , Receptores de Glucagón/metabolismo , Glucemia/metabolismo , Diabetes Mellitus Tipo 2/etiología , Vaciamiento Gástrico/efectos de los fármacos , Motilidad Gastrointestinal , Receptor del Péptido 1 Similar al Glucagón , Humanos , Secreción de Insulina , Obesidad/complicaciones , Obesidad/tratamiento farmacológico , Receptores de Glucagón/agonistas , Saciedad/efectos de los fármacos
14.
Int J Obes (Lond) ; 37(11): 1452-9, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23419600

RESUMEN

OBJECTIVE: To identify factors contributing to the variation in weight loss after Roux-en-Y gastric bypass (RYGB). DESIGN: Cross-sectional study of patients with good (excess body mass index lost (EBL) >60%) and poor weight loss response (EBL <50%) >12 months after RYGB and a lean control group matched for age and gender. MATERIALS AND METHODS: Sixteen patients with good weight loss response, 17 patients with poor weight loss response, and eight control subjects were included in the study. Participants underwent dual energy X-ray absorptiometry scan, indirect calorimetry and a 9 h multiple-meal test with measurements of glucose, insulin, total bile acids (TBA), glucagon-like peptide (GLP)-1, peptide YY3-36 (PYY), cholecystokinin (CCK), ghrelin, neurotensin and pancreatic polypeptide (PP) as well as assessment of early dumping and appetite. RESULTS: Suppression of hunger was more pronounced in the good than the poor responders in response to the multiple-meal test (P=0.006). In addition, the good responders had a larger release of GLP-1 (P=0.009) and a greater suppression of ghrelin (P=0.037) during the test, whereas the postprandial secretion of CCK was highest in the poor responders (P=0.005). PYY, neurotensin, PP and TBA release did not differ between the RYGB-operated groups. Compared with control subjects, patients had exaggerated release of GLP-1 (P<0.001), PYY (P=0.008), CCK (P=0.010) and neurotensin (P<0.001). Early dumping was comparable in the good and poor responders, but more pronounced than in controlled subjects. Differences in resting energy expenditure between the three groups were entirely explained by differences in body composition. CONCLUSION: Favorable meal-induced changes in hunger and gut hormone release in patients with good compared with poor weight loss response support the role of gut hormones in the weight loss after RYGB.


Asunto(s)
Regulación del Apetito , Síndrome de Vaciamiento Rápido/metabolismo , Metabolismo Energético , Derivación Gástrica , Obesidad Mórbida/metabolismo , Pérdida de Peso , Absorciometría de Fotón , Ácidos y Sales Biliares/metabolismo , Glucemia/metabolismo , Índice de Masa Corporal , Colecistoquinina/metabolismo , Estudios Transversales , Síndrome de Vaciamiento Rápido/etiología , Femenino , Estudios de Seguimiento , Derivación Gástrica/efectos adversos , Ghrelina/metabolismo , Péptido 1 Similar al Glucagón/metabolismo , Humanos , Masculino , Persona de Mediana Edad , Neurotensina/metabolismo , Obesidad Mórbida/cirugía , Péptido YY/metabolismo , Resultado del Tratamiento
15.
Diabetologia ; 55(2): 443-9, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22009334

RESUMEN

AIMS/HYPOTHESIS: The glucose-lowering drug metformin has been shown to inhibit complex I of the mitochondrial electron transport chain in skeletal muscle. To investigate this effect in vivo we studied skeletal muscle mitochondrial respiratory capacity and content from patients with type 2 diabetes treated with metformin (n = 14) or sulfonylurea (n = 8) and healthy control (n = 18) participants. METHODS: Mitochondrial respiratory capacity was measured ex vivo in permeabilised muscle fibres obtained from the vastus lateralis muscle of all participants. The respiratory response to in vitro titration with metformin was measured in controls. Citrate synthase (CS) activity, and fasting plasma glucose, insulin and HbA(1c) levels were measured and body composition was determined. RESULTS: Participants were matched for age, BMI and percentage body fat. Fasting plasma glucose concentrations were higher (p < 0.05) in those treated with sulfonylureas and metformin than in controls. CS activity was comparable between metformin-treated and control participants, but tended to be lower in those receiving sulfonylureas. Mitochondrial respiratory capacity with substrates for complex I and complex I and II was comparable in the groups, both when estimated per mg of tissue and when normalised to CS activity. In vitro metformin titration demonstrated a dose-dependent inhibitory effect on complex I and II in human skeletal muscle at suprapharmacological concentrations. CONCLUSIONS/INTERPRETATION: Metformin treatment does not inhibit mitochondrial complex I respiration in the electron transport chain in human skeletal muscle of patients with type 2 diabetes when measured ex vivo. Inhibition of complex I and II respiration in controls was demonstrated by metformin titration in vitro at doses well above those observed during metformin treatment.


Asunto(s)
Respiración de la Célula , Diabetes Mellitus Tipo 2/sangre , Complejo I de Transporte de Electrón/metabolismo , Metformina/uso terapéutico , Glucemia/metabolismo , Índice de Masa Corporal , Estudios de Casos y Controles , Citrato (si)-Sintasa/metabolismo , Hemoglobina Glucada/metabolismo , Humanos , Hipoglucemiantes/uso terapéutico , Insulina/metabolismo , Masculino , Persona de Mediana Edad , Modelos Biológicos , Músculo Esquelético/metabolismo , Consumo de Oxígeno , Compuestos de Sulfonilurea/uso terapéutico
16.
Diabetologia ; 55(5): 1406-16, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22286551

RESUMEN

AIMS/HYPOTHESIS: The aim of this study was to evaluate the separate impact of insulin resistance and impaired glucose tolerance (IGT) on the incretin effect. METHODS: Twenty-one healthy glucose-tolerant first-degree relatives of patients with type 2 diabetes underwent a 75 g OGTT, an isoglycaemic i.v. glucose test and a mixed meal to evaluate the incretin effect before and after treatment with dexamethasone to increase insulin resistance. Beta cell glucose sensitivity, beta cell index and fasting proinsulin were measured as indices of beta cell function. RESULTS: After dexamethasone, ten individuals had increased insulin resistance but normal glucose tolerance (NGT), while 11 individuals with an equal increase in insulin resistance developed IGT. In the NGT and IGT groups, the incretin effects were 71 ± 3.2% and 67 ± 4.6% (p = 0.4) before treatment, but decreased significantly in both groups to 58 ± 5.2% and 32 ± 8.8% (p < 0.05 between groups) after treatment. Dexamethasone increased total glucagon-like peptide-1 and glucose-dependent insulinotropic peptide responses to the OGTT. The impaired incretin effect in NGT was observed in the absence of reductions in beta cell glucose sensitivity and beta cell index during i.v. glucose, corrected for insulin resistance, but in parallel with increased proinsulin/C-peptide ratio. CONCLUSION/INTERPRETATION: Insulin resistance and IGT, representing two stages in the path towards diabetes, are associated with differential reductions in the incretin effect seen before the development of IGT and overt type 2 diabetes. The reduction is unrelated to secretion of incretin hormones, but is related to insulin resistance and subtle beta cell defects, and is further aggravated on development of IGT. TRIAL REGISTRATION: ClinicalTrials.gov NCT00784745. FUNDING: This study was supported by a grant from the Novo Nordisk Foundation.


Asunto(s)
Glucemia/efectos de los fármacos , Dexametasona/farmacología , Glucocorticoides/farmacología , Intolerancia a la Glucosa/inducido químicamente , Incretinas/sangre , Resistencia a la Insulina/fisiología , Adulto , Femenino , Intolerancia a la Glucosa/metabolismo , Prueba de Tolerancia a la Glucosa , Humanos , Incretinas/metabolismo , Masculino , Adulto Joven
17.
Diabetologia ; 55(7): 1890-901, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22538359

RESUMEN

Roux-en-Y gastric bypass (RYGB) greatly improves glycaemic control in morbidly obese patients with type 2 diabetes, in many even before significant weight loss. Understanding the responsible mechanisms may contribute to our knowledge of the pathophysiology of type 2 diabetes and help identify new drug targets or improve surgical techniques. This review summarises the present knowledge based on pathophysiological studies published during the last decade. Taken together, two main mechanisms seem to be responsible for the early improvement in glycaemic control after RYGB: (1) an increase in hepatic insulin sensitivity induced, at least in part, by energy restriction and (2) improved beta cell function associated with an exaggerated postprandial glucagon-like peptide 1 secretion owing to the altered transit of nutrients. Later a weight loss induced improvement in peripheral insulin sensitivity follows.


Asunto(s)
Glucemia/metabolismo , Diabetes Mellitus Tipo 2/sangre , Derivación Gástrica , Péptido 1 Similar al Glucagón/sangre , Resistencia a la Insulina , Obesidad Mórbida/cirugía , Diabetes Mellitus Tipo 2/metabolismo , Femenino , Prueba de Tolerancia a la Glucosa , Humanos , Células Secretoras de Insulina/metabolismo , Masculino , Obesidad Mórbida/sangre , Factores de Tiempo , Resultado del Tratamiento
18.
Diabetologia ; 55(1): 105-13, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21953277

RESUMEN

AIMS/HYPOTHESIS: Genome-wide association studies have identified novel WHR and BMI susceptibility loci. The aim of this study was to elucidate if any of these loci had an effect on quantitative measures of glucose homeostasis, including estimates of insulin release and insulin sensitivity in an epidemiological setting. METHODS: By applying an additive genetic model, 14 WHR-associated gene variants and 18 BMI-associated variants were investigated for their relationships with glucose-related metabolic traits in treatment-naive individuals from the population-based Inter99 study sample (n = 6,039). RESULTS: Of the variants associated with BMI, the QPCTL rs2287019 C allele was associated with an increased insulinogenic index of 7.4% per risk allele (p = 4.0 × 10⁻7) and increased disposition index of 5.6% (p = 6.4 × 10⁻5). The LRP1B rs2890652 C allele was associated with insulin resistance, showing a 3.3% increase (p = 0.0011) using the HOMA-insulin resistance (HOMA-IR) index and a 2.2% reduction (p = 0.0014) with the Matsuda index. Of the variants associated with WHR, LYPLAL1/SLC30A10 rs4846567 G allele carriers showed a 5.2% lower HOMA-IR (p = 0.00086) in women, indicating improved insulin sensitivity. Female carriers of the VEGFA rs6905288 A allele were insulin resistant, with a 3.7% increase in HOMA-IR (p = 0.00036) and 4.0% decrease in Matsuda index (p = 2 × 10⁻4). CONCLUSIONS: Our correlative findings from analysing single-locus data suggest that some variation in validated BMI and WHR loci are associated with either increased or decreased insulin sensitivity and thereby potentially with metabolically healthy or metabolically unhealthy subsets of obesity. The results call for testing in larger study samples and for further physiological exploration of the possible metabolic implications of these loci.


Asunto(s)
Aminoaciltransferasas/genética , Lisofosfolipasa/genética , Obesidad/genética , Polimorfismo de Nucleótido Simple , Receptores de LDL/genética , Factor A de Crecimiento Endotelial Vascular/genética , Índice de Masa Corporal , Portador Sano , Estudios Transversales , Dinamarca , Femenino , Estudios de Asociación Genética , Humanos , Hiperinsulinismo/complicaciones , Hiperinsulinismo/genética , Resistencia a la Insulina , Masculino , Persona de Mediana Edad , Modelos Genéticos , Obesidad/complicaciones , Caracteres Sexuales , Relación Cintura-Cadera
19.
Am J Physiol Endocrinol Metab ; 303(1): E122-31, 2012 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-22535748

RESUMEN

Our aim was to study the potential mechanisms responsible for the improvement in glucose control in Type 2 diabetes (T2D) within days after Roux-en-Y gastric bypass (RYGB). Thirteen obese subjects with T2D and twelve matched subjects with normal glucose tolerance (NGT) were examined during a liquid meal before (Pre), 1 wk, 3 mo, and 1 yr after RYGB. Glucose, insulin, C-peptide, glucagon-like peptide-1 (GLP-1), glucose-dependent-insulinotropic polypeptide (GIP), and glucagon concentrations were measured. Insulin resistance (HOMA-IR), ß-cell glucose sensitivity (ß-GS), and disposition index (D(ß-GS): ß-GS × 1/HOMA-IR) were calculated. Within the first week after RYGB, fasting glucose [T2D Pre: 8.8 ± 2.3, 1 wk: 7.0 ± 1.2 (P < 0.001)], and insulin concentrations decreased significantly in both groups. At 129 min, glucose concentrations decreased in T2D [Pre: 11.4 ± 3, 1 wk: 8.2 ± 2 (P = 0.003)] but not in NGT. HOMA-IR decreased by 50% in both groups. ß-GS increased in T2D [Pre: 1.03 ± 0.49, 1 wk: 1.70 ± 1.2, (P = 0.012)] but did not change in NGT. The increase in DI(ß-GS) was 3-fold in T2D and 1.5-fold in NGT. After RYGB, glucagon secretion was increased in response to the meal. GIP secretion was unchanged, while GLP-1 secretion increased more than 10-fold in both groups. The changes induced by RYGB were sustained or further enhanced 3 mo and 1 yr after surgery. Improvement in glycemic control in T2D after RYGB occurs within days after surgery and is associated with increased insulin sensitivity and improved ß-cell function, the latter of which may be explained by dramatic increases in GLP-1 secretion.


Asunto(s)
Diabetes Mellitus Tipo 2/complicaciones , Derivación Gástrica , Resistencia a la Insulina , Células Secretoras de Insulina/metabolismo , Obesidad/metabolismo , Obesidad/cirugía , Adulto , Índice de Masa Corporal , Péptido C/sangre , Péptido C/metabolismo , Femenino , Estudios de Seguimiento , Glucagón/sangre , Glucagón/metabolismo , Péptido 1 Similar al Glucagón/sangre , Péptido 1 Similar al Glucagón/metabolismo , Glucosa/metabolismo , Humanos , Hiperglucemia/prevención & control , Masculino , Persona de Mediana Edad , Obesidad/complicaciones , Obesidad/fisiopatología , Obesidad Mórbida/complicaciones , Obesidad Mórbida/metabolismo , Obesidad Mórbida/fisiopatología , Obesidad Mórbida/cirugía , Periodo Posprandial , Factores de Tiempo
20.
Diabet Med ; 29(4): 479-87, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22050462

RESUMEN

AIM: To explore the putative association of new-onset diabetes and the soluble urokinase plasminogen activator receptor (suPAR), which is a new and stable plasma marker of immune function and low-grade inflammation. This association has been previously suggested by using the less sensitive International Classification of Disease system to detect incident diabetes in the Danish MONICA 10 cohort. METHODS: The Danish National Diabetes Register enabled more accurate identification of incident diabetes during a median follow-up of 13.8 years in the Danish MONICA 10 cohort (n = 2353 generally healthy individuals). The soluble urokinase plasminogen activator receptor was measured by the ELISA method. To fulfil model assumptions, outcome analyses were stratified by age, and further by smoking, owing to the interaction between the soluble urokinase plasminogen activator receptor and smoking on new-onset diabetes (P < 0.0001). RESULTS: New-onset diabetes (n = 182) was associated with increased soluble urokinase plasminogen activator receptor levels (P = 0.013). Among 699 middle-aged (41 and 51 years) and 564 older (61 and 71 years) non-smokers, participants in the upper soluble urokinase plasminogen activator receptor quartile had a sex- and age-adjusted relative risk of 6.01 (95% CI 2.17-16.6, P < 0.0006) and relative risk of 3.25 (95% CI 1.51-6.98, P = 0.0025), respectively, for new-onset diabetes compared with participants in the lowest quartile. This relationship remained significant after additional adjustments for C-reactive protein and leukocytes or fasting glucose and insulin or BMI (P < 0.05). The soluble urokinase plasminogen activator receptor was not related to incident diabetes among smokers (P ≥ 0.85). CONCLUSIONS: In these explorative analyses, the soluble urokinase plasminogen activator receptor associated independently with incident diabetes in non-smokers, supporting an immune origin of Type 2 diabetes. Competing disease risk may explain lack of association among smokers.


Asunto(s)
Diabetes Mellitus Tipo 2/sangre , Mediadores de Inflamación/sangre , Receptores del Activador de Plasminógeno Tipo Uroquinasa/sangre , Adulto , Factores de Edad , Anciano , Biomarcadores/sangre , Estudios de Cohortes , Diabetes Mellitus Tipo 2/inmunología , Diabetes Mellitus Tipo 2/fisiopatología , Femenino , Estudios de Seguimiento , Humanos , Masculino , Persona de Mediana Edad , Pronóstico , Receptores del Activador de Plasminógeno Tipo Uroquinasa/inmunología , Medición de Riesgo , Factores de Riesgo , Factores Sexuales
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA