Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
J Endocrinol Invest ; 46(11): 2213-2236, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37378828

ABSTRACT

BACKGROUND: Adaptive thermogenesis represents the main mechanism through which the body generates heat in response to external stimuli, a phenomenon that includes shivering and non-shivering thermogenesis. The non-shivering thermogenesis is mainly exploited by adipose tissue characterized by a brown aspect, which specializes in energy dissipation. A decreased amount of brown adipose tissue has been observed in ageing and chronic illnesses such as obesity, a worldwide health problem characterized by dysfunctional adipose tissue expansion and associated cardiometabolic complications. In the last decades, the discovery of a trans-differentiation mechanism ("browning") within white adipose tissue depots, leading to the generation of brown-like cells, allowed to explore new natural and synthetic compounds able to favour this process and thus enhance thermogenesis with the aim of counteracting obesity. Based on recent findings, brown adipose tissue-activating agents could represent another option in addition to appetite inhibitors and inhibitors of nutrient absorption for obesity treatment. PURPOSE: This review investigates the main molecules involved in the physiological (e.g. incretin hormones) and pharmacological (e.g. ß3-adrenergic receptors agonists, thyroid receptor agonists, farnesoid X receptor agonists, glucagon-like peptide-1, and glucagon receptor agonists) modulation of adaptive thermogenesis and the signalling mechanisms involved.

2.
J Endocrinol Invest ; 45(9): 1663-1671, 2022 Sep.
Article in English | MEDLINE | ID: mdl-35476320

ABSTRACT

PURPOSE: To evaluate if a web-based telemedicine system (the Glucoonline® system) is effective to improve glucose control in insulin-treated patients with type 1 and type 2 diabetes, as compared to standard of care. METHODS: This was a prospective, randomized, controlled trial, carried out at three tertiary referral centers for diabetes in Italy. Adults with insulin-treated type 1 and type 2 diabetes, inadequate glycemic control, and no severe diabetes-related complications and/or comorbidities were eligible for this study. Patients were randomized to either perform telemedicine-assisted (Group A) or standard (Group B) self-monitoring blood glucose (SMBG) for 6 months. In Group A, patients received prompt feedback about their blood glucose levels and therapy suggestions from the study staff via phone/SMS, when appropriate. In Group B, patients had no remote assistance from the study staff between planned visits. RESULTS: 123 patients were included in the final analysis. After 6 months, patients achieved a significant reduction in HbA1c in Group A (-0.38%, p < 0.05) but not in Group B (+ 0.08%, p = 0.53). A significant difference in the percentage of patients with HbA1c < 7% between Group A and Group B was found after 3 months (28.6% vs 11.1%, p = 0.02). Also, fewer patients (p < 0.05) with HbA1c > 8.5% were found in Group A vs Group B, respectively, after both 3 months (14.3% vs 35.2%) and 6 months (21.8% vs 42.9%). CONCLUSIONS: The use of the Glucoonline™ system resulted in improved metabolic control. Telemedicine services have potential to support diabetes self-management and provide the patients with remote, prompt assistance using affordable technological equipment. Trial registration This study was registered at clinicaltrials.gov (NCT01804803) on March 5, 2013.


Subject(s)
Diabetes Mellitus, Type 1 , Diabetes Mellitus, Type 2 , Telemedicine , Adult , Blood Glucose/analysis , Blood Glucose Self-Monitoring/methods , Diamond , Glycated Hemoglobin/analysis , Humans , Hypoglycemic Agents/therapeutic use , Insulin/therapeutic use , Prospective Studies , Telemedicine/methods
3.
J Endocrinol Invest ; 44(12): 2741-2748, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34173961

ABSTRACT

INTRODUCTION: Obstructive sleep apnoea (OSA) is an underdiagnosed condition frequently associated with glycaemic control impairment in patients with type 2 diabetes. AIM: To assess the relationship between glycometabolic parameters and OSA in obese non-diabetic subjects. METHODS: Ninety consecutive subjects (mean age 44.9 ± 12 years, mean BMI 42.1 ± 9 kg/m2) underwent polysomnography and a 2-h oral glucose tolerance test (OGTT). RESULTS: OSA was identified in 75% of subjects, with a higher prevalence of males compared to the group of subjects without OSA (62% vs 32%, p = 0.02). Patients with OSA had comparable BMI (42.8 kg/m2 vs 39.4 kg/m2), a higher average HbA1c (5.8% vs 5.4%, p < 0.001), plasma glucose at 120 min during OGTT (2 h-PG; 123 mg/dl vs 97 mg/dl, p = 0.009) and diastolic blood pressure (81.1 mmHg vs 76.2 mmHg, p = 0.046) than obese subjects without OSA. HbA1c and 2 h-PG were found to be correlated with the apnoea-hypopnoea index (AHI; r = 0.35 and r = 0.42, respectively) and with percent of sleep time with oxyhaemoglobin saturation < 90% (ST90; r = 0.44 and r = 0.39, respectively). Further, in a linear regression model, ST90 and AHI were found to be the main determinants of 2 h-PG (ß = 0.81, p < 0.01 and ß = 0.75, p = 0.02, respectively) after controlling for age, sex, waist circumference, physical activity, and C-reactive protein. Similarly, ST90 and AHI persisted as independent determinants of HbA1c (ß = 0.01, p = 0.01 and ß = 0.01, p = 0.01, respectively). CONCLUSION: Beyond the traditional clinical parameters, the presence of a normal-high value of 2 h-PG and HbA1c should raise suspicion of the presence of OSA in obese subjects.


Subject(s)
Blood Glucose/metabolism , Glycated Hemoglobin/analysis , Hyperglycemia , Obesity , Sleep Apnea, Obstructive , Adult , Body Mass Index , Female , Glucose Tolerance Test/methods , Humans , Hyperglycemia/diagnosis , Hyperglycemia/epidemiology , Hyperglycemia/etiology , Male , Obesity/diagnosis , Obesity/epidemiology , Obesity/metabolism , Obesity/physiopathology , Polysomnography/methods , Postprandial Period , Prevalence , Risk Factors , Severity of Illness Index , Sleep Apnea, Obstructive/blood , Sleep Apnea, Obstructive/diagnosis , Sleep Apnea, Obstructive/epidemiology , Sleep Apnea, Obstructive/physiopathology
4.
J Endocrinol Invest ; 44(5): 921-941, 2021 May.
Article in English | MEDLINE | ID: mdl-33145726

ABSTRACT

The way by which subcutaneous adipose tissue (SAT) expands and undergoes remodeling by storing excess lipids through expansion of adipocytes (hypertrophy) or recruitment of new precursor cells (hyperplasia) impacts the risk of developing cardiometabolic and respiratory diseases. In unhealthy obese subjects, insulin resistance, type 2 diabetes, hypertension, and obstructive sleep apnoea are typically associated with pathologic SAT remodeling characterized by adipocyte hypertrophy, as well as chronic inflammation, hypoxia, increased visceral adipose tissue (VAT), and fatty liver. In contrast, metabolically healthy obese individuals are generally associated with SAT development characterized by the presence of smaller and numerous mature adipocytes, and a lower degree of VAT inflammation and ectopic fat accumulation. The remodeling of SAT and VAT is under genetic regulation and influenced by inherent depot-specific differences of adipose tissue-derived stem cells (ASCs). ASCs have multiple functions such as cell renewal, adipogenic capacity, and angiogenic properties, and secrete a variety of bioactive molecules involved in vascular and extracellular matrix remodeling. Understanding the mechanisms regulating the proliferative and adipogenic capacity of ASCs from SAT and VAT in response to excess calorie intake has become a focus of interest over recent decades. Here, we summarize current knowledge about the biological mechanisms able to foster or impair the recruitment and adipogenic differentiation of ASCs during SAT and VAT development, which regulate body fat distribution and favorable or unfavorable metabolic responses.


Subject(s)
Adipogenesis/physiology , Adipose Tissue , Lipogenesis/genetics , Obesity , Adipocytes/metabolism , Adipose Tissue/metabolism , Adipose Tissue/pathology , Body Fat Distribution , Gene Expression Regulation , Humans , Insulin Resistance , Mesenchymal Stem Cells/metabolism , Obesity/immunology , Obesity/metabolism , Obesity/pathology
5.
Nutr Metab Cardiovasc Dis ; 29(5): 421-431, 2019 05.
Article in English | MEDLINE | ID: mdl-30952574

ABSTRACT

Until recently, in Italy, the use of continuous glucose monitoring (CGM) systems has been limited, but is now rapidly increasing, including the so-called real-time CGM (rtCGM) and the intermittently viewed CGM (iCGM), also called Flash Glucose Monitoring (FGM). These technologies overcome many of the limitations of self-monitoring of blood glucose (SMBG) by fingerprick and allow to go beyond HbA1c to check glucose control in diabetes. However, standardized protocols for applying and interpreting rtCGM and FGM data are lacking. In this paper, we delineate a consensus amongst Italian diabetes physicians on the attributes of rtCGM and FGM technologies, and introduce a consistent approach for their use by Italian healthcare professionals. Most experts consider rtCGM and FGM as two separate categories of interstitial subcutaneous fluid (ISF) sensing technologies, and see them as superior to SMBG. Furthermore, there is strong consensus that rtCGM and FGM reduce hypoglycemia risk, increase the amount of time in the target glucose range and augment treatment satisfaction. However, there is still no agreement on the indication of the FGM for subjects who suffer asymptomatic hypoglycemia. Consensus on the role of education in initiating and optimizing use of rtCGM/FGM and about the interpretation of glucose trends was near unanimous, whereas no consensus was reached on the statement that there are no disadvantages/risks of rtCGM/FGM. Some issues remain in rtCGM/FGM management: a) risk of excessive correction of high or low glucose; b) risk of alert fatigue leading to alert silencing or rtCGM termination; c) allergic reaction to the adhesive keeping rtCGM or FGM sensors in place. The panel almost unanimously agreed that sensor accuracy depends on multiple variables, that alarm setting should be individualized, and that global glycemic profile represent an useful tool in interpreting glucose data. More clinical studies and a wider use of these devices will increase the efficacy and effectiveness of continuous glucose monitoring in Italy.


Subject(s)
Biosensing Techniques/instrumentation , Blood Glucose Self-Monitoring/instrumentation , Blood Glucose/metabolism , Diabetes Mellitus/diagnosis , Extracellular Fluid/metabolism , Wearable Electronic Devices , Biomarkers/blood , Blood Glucose/drug effects , Consensus , Delphi Technique , Diabetes Mellitus/blood , Diabetes Mellitus/drug therapy , Equipment Design , Glycated Hemoglobin/metabolism , Humans , Hypoglycemic Agents/therapeutic use , Italy , Predictive Value of Tests , Reproducibility of Results , Time Factors , Treatment Outcome
6.
J Biol Regul Homeost Agents ; 30(1): 297-302, 2016.
Article in English | MEDLINE | ID: mdl-27049106

ABSTRACT

Increased risk of osteoporosis and its clinical significance in patients with diabetes is controversial. This study aims to increase the data which are available regarding the prevalence of diabetes mellitus in patients affected by fragility fracture in Italy. We retrospectively studied Hospital Discharge Data (HDD) in the Apulian database for the period 2006–2010 to identify a fragility fracture diagnosis in males over 65 years of age and in females over 50. The database was then checked for drug prescriptions to identify those persons who had taken at least one osteoporosis drug. Within this latter group, thanks to hospital admission and prescription records, the subjects affected with diabetes mellitus were identified. Between 2006 and 2010 in Apulia 177,639 patients were hospitalized and diagnosed as having fragility fracture. The greatest number of those fragility fractures were found to be in the 70 to 79 age range (64,917 total; females 56,994, males 7,923). The prevalence of diabetes subjects in Apulia in this period was estimated at 6.5%. In the same region and period 21.1% of subjects affected by diabetes experienced a fragility fracture; in particular, this number was 27% for males and for 20.5% females. This is the first study providing data on the prevalence of fragility fractures and diabetes in the Apulian population. The data confirm that diabetes is a risk factor which influences bone density and risk of fractures and therefore the need of osteoporosis screening and treatment in diabetic patients.


Subject(s)
Diabetes Mellitus/epidemiology , Fractures, Bone/complications , Fractures, Bone/epidemiology , Aged , Aged, 80 and over , Diabetes Mellitus/diagnosis , Female , Humans , Italy/epidemiology , Male , Middle Aged , Prevalence
7.
Nutr Metab Cardiovasc Dis ; 26(4): 333-44, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26947594

ABSTRACT

BACKGROUND AND AIMS: Since treatment with insulin detemir results in a lower weight gain compared to human insulin, we investigated whether detemir is associated with lower ability to promote adipogenesis and/or lipogenesis in human adipose stem cells (ASC). METHODS AND RESULTS: Human ASC isolated from both the subcutaneous and visceral adipose tissues were differentiated for 30 days in the presence of human insulin or insulin detemir. Nile Red and Oil-Red-O staining were used to quantify the rate of ASC conversion to adipocytes and lipid accumulation, respectively. mRNA expression levels of early genes, including Fos and Cebpb, as well as of lipogenic and adipogenic genes, were measured at various phases of differentiation by qRT-PCR. Activation of insulin signaling was assessed by immunoblotting. ASC isolated from subcutaneous and visceral adipose tissue were less differentiated when exposed to insulin detemir compared to human insulin, showing lower rates of adipocyte conversion, reduced triglyceride accumulation, and impaired expression of late-phase adipocyte marker genes, such as Pparg2, Slc2a4, Adipoq, and Cidec. However, no differences in activation of insulin receptor, Akt and Erk and induction of the early genes Fos and Cebpb were observed between insulin detemir and human insulin. CONCLUSION: Insulin detemir displays reduced induction of the Pparg2 adipocyte master gene and diminished effects on adipocyte differentiation and lipogenesis in human subcutaneous and visceral ASC, in spite of normal activation of proximal insulin signaling reactions. These characteristics of insulin detemir may be of potential relevance to its weight-sparing effects observed in the clinical setting.


Subject(s)
Adipocytes/drug effects , Hypoglycemic Agents/pharmacology , Insulin Detemir/pharmacology , Insulin, Long-Acting/pharmacology , Stem Cells/drug effects , Adipocytes/cytology , Adipogenesis/drug effects , Adiponectin/genetics , Adiponectin/metabolism , Adult , Aged , Aged, 80 and over , Apoptosis Regulatory Proteins , CCAAT-Enhancer-Binding Protein-beta/genetics , CCAAT-Enhancer-Binding Protein-beta/metabolism , Cell Differentiation/drug effects , Female , Glucose Transporter Type 4/genetics , Glucose Transporter Type 4/metabolism , Humans , Insulin/metabolism , Intra-Abdominal Fat/cytology , Lipogenesis/drug effects , Male , Middle Aged , PPAR gamma/genetics , PPAR gamma/metabolism , Proteins/genetics , Proteins/metabolism , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction , Stem Cells/metabolism , Subcutaneous Fat/cytology
8.
Acta Diabetol ; 53(3): 403-12, 2016 Jun.
Article in English | MEDLINE | ID: mdl-26429560

ABSTRACT

AIMS: The aim of the study was to evaluate and compare continuous subcutaneous insulin infusion (CSII) use in pediatric and adult age groups. METHODS: Data were collected with a questionnaire sent by e-mail to CSII-experienced Diabetes Centers. The questionnaire assessed: (1) number of CSII-treated patients; (2) patient demographic data and characteristics; (3) structure and organization of Diabetes Centers providing CSII therapy; (4) pump characteristics (conventional pump, sensor-augmented pump); and (5) CSII dropouts. RESULTS: A total of 217 out of 1093 Italian centers participated: 51 pediatric (23.5 %) and 166 (76.5 %) adult centers (AP). Compared to a survey performed in 2005, there was a significant increase in the number of pediatric units when compared to adult units (112 vs 37 %, respectively, p < 0.05). Pediatric age is characterized by a greater concern for quality of life and injections, and a higher dropout rate (10.6 vs 8.9 %) mainly related to pump wearability and site reactions. A complete diabetes-care team is associated with a superior use of technology (fewer dropouts, increased CGM and advanced bolus use) which is, however, still used in a small percentage of patients. CONCLUSIONS: In Italy, the number of CSII-treated pediatric patients (PP) is growing more significantly when compared to adults. Only 60 % of all patients are using advanced functions and 20 % are using CGMs continuously. This confirms the great interest in diabetes technology that is growing in pediatric diabetologists. However, much improvement is warranted in the organization and specialized training of pediatric, adult and transitional facilities.


Subject(s)
Diabetes Mellitus/drug therapy , Insulin Infusion Systems/adverse effects , Adolescent , Adult , Age Factors , Child , Child, Preschool , Female , Humans , Insulin Infusion Systems/psychology , Insulin Infusion Systems/standards , Italy , Male , Patients/psychology , Surveys and Questionnaires
9.
J Intern Med ; 274(2): 176-91, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23565931

ABSTRACT

OBJECTIVES: Poorer control of risk factors for cardiovascular disease (CVD) has been reported in diabetic women, as compared with diabetic men. It has been proposed that this finding is due to gender disparities in treatment intensity. We investigated this hypothesis in a large contemporary cohort of subjects with type 2 diabetes. DESIGN: Observational, cross-sectional study. SUBJECTS AND SETTING: Consecutive patients with type 2 diabetes from the Renal Insufficiency And Cardiovascular Events (RIACE) Italian multicentre study (n = 15 773), attending 19 hospital-based diabetes clinics in 2007-2008. MAIN OUTCOME MEASURES: Traditional CVD risk factors, macro- and microvascular complications and current glucose-, lipid- and blood pressure (BP)-lowering treatments were assessed. RESULTS: Although CVD was more prevalent in men, women showed a less favourable CVD risk profile and worse performance in achieving treatment targets for haemoglobin A1c , LDL, HDL and non-HDL cholesterol, systolic blood pressure (BP) and in particular obesity [body mass index (BMI) and waist circumference], but not for triglycerides and diastolic BP. However, women were more frequently receiving pharmacological treatment for hypertension and to a lesser extent hyperglycaemia and dyslipidaemia than men, and female gender remained an independent predictor of unmet therapeutic targets after adjustment for confounders such as treatments, BMI, duration of diabetes and, except for the systolic BP goal, age. CONCLUSIONS: In women with type 2 diabetes from the RIACE cohort, a more adverse CVD risk profile and a higher likelihood of failing treatment targets, compared with men, were not associated with treatment differences. This suggests that factors other than gender disparities in treatment intensity are responsible.


Subject(s)
Cardiovascular Diseases/epidemiology , Cardiovascular Diseases/therapy , Diabetes Mellitus, Type 2/epidemiology , Diabetes Mellitus, Type 2/therapy , Hypercholesterolemia/epidemiology , Hypertension/epidemiology , Aged , Body Mass Index , Cardiovascular Diseases/diagnosis , Cross-Sectional Studies , Diabetes Mellitus, Type 2/diagnosis , Female , Humans , Hypercholesterolemia/diagnosis , Hyperglycemia/diagnosis , Hyperglycemia/epidemiology , Hypertension/diagnosis , Italy , Male , Middle Aged , Prognosis , Risk Factors , Severity of Illness Index , Sex Factors , Survival Analysis , Treatment Outcome
10.
Endocrinology ; 153(12): 5770-81, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23077074

ABSTRACT

Increased apoptosis of cardiac progenitor cells (CPCs) has been proposed as a mechanism of myocardial damage and dysfunction. Glucagon-like peptide-1 (GLP-1) has been shown to improve heart recovery and function after ischemia and to promote cell survival. The protective effects of GLP-1 on oxidative stress-induced apoptosis were investigated in human CPCs isolated from human heart biopsies. Mesenchymal-type cells were isolated from human heart biopsies, exhibited the marker profile of CPCs, differentiated toward the myocardiocyte, adipocyte, chondrocyte, and osteocyte lineages under appropriate culture conditions, and expressed functional GLP-1 receptors. CPCs were incubated with GLP-1 with or without hydrogen peroxide (H(2)O(2)). Phospho- and total proteins were detected by immunoblotting and immunofluorescence analysis. Gene expression was evaluated by quantitative RT-PCR. The role of the canonical GLP-1 receptor was assessed by using the receptor antagonist exendin(9-39) and receptor-specific silencer small interfering RNAs. Cell apoptosis was quantified by an ELISA assay and by flow cytometry-detected Annexin V. Exposure of CPCs to H(2)O(2) induced a 2-fold increase in cell apoptosis, mediated by activation of the c-Jun N-terminal protein kinase (JNK) pathway. Preincubation of CPCs with GLP-1 avoided H(2)O(2)-triggered JNK phosphorylation and nuclear localization, and protected CPCs from apoptosis. The GLP-1 effects were markedly reduced by coincubation with the receptor antagonist exendin(9-39), small interfering RNA-mediated silencing of the GLP-1 receptor, and pretreatment with the protein kinase A inhibitor H89. In conclusion, activation of GLP-1 receptors prevents oxidative stress-mediated apoptosis in human CPCs by interfering with JNK activation and may represent an important mechanism for the cardioprotective effects of GLP-1.


Subject(s)
Apoptosis , Glucagon-Like Peptide 1/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , Myocardium/cytology , Oxidative Stress , Stem Cells/cytology , Annexin A5/pharmacology , Biopsy , Cell Differentiation , Cell Nucleus/metabolism , Cells, Cultured , Enzyme Activation , Enzyme-Linked Immunosorbent Assay/methods , Flow Cytometry/methods , Humans , Hydrogen Peroxide/metabolism , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , Models, Biological , Peptide Fragments/pharmacology , Phosphorylation , RNA, Small Interfering/metabolism , Signal Transduction
11.
Diabet Med ; 28(5): 560-6, 2011 May.
Article in English | MEDLINE | ID: mdl-21204958

ABSTRACT

AIMS: Postprandial hyperglycaemia in patients with Type 2 diabetes mellitus has been linked to the development of cardiovascular disease. This study compared the effects of mealtime (thrice-daily) nateglinide with once-daily glyburide on postprandial glucose levels in patients with Type 2 diabetes and postprandial hyperglycaemia. METHODS: Patients with Type 2 diabetes aged ≥ 21 years with 2-h postprandial glucose levels ≥ 11.1 mmol/l, HbA(1c) of 6.5-8.5% (48-69 mmol/mol) and BMI of 22-30 kg/m(2) were randomized to 6 weeks' double-blind treatment with nateglinide 120 mg three times daily prior to meals, or glyburide 5 mg once daily before breakfast. The primary endpoint was the baseline-adjusted change in plasma glucose from preprandial (fasting plasma glucose) to 2-h postprandial glucose levels (2-h postprandial glucose excursion) at 6 weeks. RESULTS: Patients were randomized to nateglinide (n = 122) or glyburide (n = 110). The treatment groups were similar in terms of age, gender, BMI, fasting plasma glucose, 2-h postprandial glucose and HbA(1c). At endpoint, nateglinide recipients had significantly greater reductions than those receiving glyburide in both the 2-h (-2.4 vs. -1.6 mmol/l; P = 0.02) and 1-h (-1.7 vs. -0.9 mmol/l; P = 0.016) postprandial glucose excursions. Adverse events, most commonly symptomatic hypoglycaemia, were reported in 26% of recipients of glyburide and 22% of recipients of nateglinide. Episodes of suspected mild hypoglycaemia were reported in 24% of recipients of glyburide and 10% of recipients of nateglinide. CONCLUSIONS: Nateglinide leads to greater reductions in postprandial glucose excursions and is associated with a lower risk of hypoglycaemia than glyburide in this selected population of patients with Type 2 diabetes.


Subject(s)
Cardiovascular Diseases/drug therapy , Cyclohexanes/therapeutic use , Diabetes Mellitus, Type 2/drug therapy , Diabetic Angiopathies/drug therapy , Glyburide/therapeutic use , Hyperglycemia/drug therapy , Phenylalanine/analogs & derivatives , Adult , Aged , Aged, 80 and over , Cardiovascular Diseases/metabolism , Cardiovascular Diseases/prevention & control , Diabetes Mellitus, Type 2/metabolism , Diabetic Angiopathies/metabolism , Diabetic Angiopathies/prevention & control , Double-Blind Method , Fasting , Female , Glycated Hemoglobin/metabolism , Humans , Hyperglycemia/prevention & control , Middle Aged , Nateglinide , Phenylalanine/therapeutic use , Postprandial Period
12.
J Endocrinol Invest ; 34(1): 69-77, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21233599

ABSTRACT

Type 2 diabetes mellitus is increasing in prevalence at alarming rates. Concurrent with its expanding prevalence is the increase in the related risk of morbidity and mortality. Because diabetic patients are prone to cardiovascular disease, treatment strategies should address the cardiovascular risk factors, including blood pressure, lipids, and body weight, in addition to the glycemic aspects of the disease. Newer agents, such as glucagon-like peptide-1 (GLP-1) analogs and dipeptidyl peptidase-4 (DPP-4) inhibitors, have varying degrees of evidence to support their effects on body weight, blood pressure, and lipid levels, beyond glycated hemoglobin reduction. While GLP-1 agonists produce a weight loss, the DPP-4 inhibitors, conversely, appear to have a weight-neutral effect. Substantial evidence demonstrates that both medications produce modest reductions in systolic blood pressure and, in some cases, diastolic blood pressure, and reduce several markers of cardiovascular risk, including C-reactive protein. Moreover, GLP-1 influences endothelial function. The effect of the incretin hormones on serum lipids are either neutral or beneficial, with small, non-significant decreases in LDL cholesterol, increases in HDL cholesterol, and occasionally significant decreases in fasting triglyceride levels. Also, they have positive effects on hepatic steatosis. Although GLP-1 agonists and DPP-4 inhibitors are at present not appropriate for primary treatment of cardiovascular risks factors, the reduction of these parameters is evidently beneficial.


Subject(s)
Combined Modality Therapy/methods , Diabetes Mellitus, Type 2/therapy , Incretins/therapeutic use , Animals , Combined Modality Therapy/trends , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/complications , Glucagon-Like Peptide 1/pharmacology , Health Services Needs and Demand , Humans , Incretins/physiology , Models, Biological , Patient Care Planning/trends
13.
J Endocrinol Invest ; 31(9 Suppl): 79-84, 2008 Sep.
Article in English | MEDLINE | ID: mdl-19020393

ABSTRACT

GH regulates important physiological processes, including somatic growth and development, and carbohydrate and lipid metabolism. GH deficiency and GH replacement therapy exert opposite effects on body composition and fat accumulation, suggesting that GH may directly regulate adipocyte functions. Multiple studies have shown that in tissues previously deprived of GH, short-term stimulation with GH is able to mimic the actions of insulin, including stimulation of amino-acid and glucose transport, and lipogenesis. However, the antagonistic effects of GH on insulin-mediated metabolic responses are well-documented: GH excess in patients with GH-producing pituitary tumors causes hyperinsulinemia, insulin resistance, and even clinical diabetes mellitus. These apparently contradictory effects may be explained at the molecular level by the complex interplay between GH and insulin signaling. In this review, we examine the consequences of acute and chronic effects of GH on visceral fat and on metabolic responses in adipocytes, and discuss experimental data illustrating the integrated crosstalk between GH and insulin.


Subject(s)
Human Growth Hormone/adverse effects , Human Growth Hormone/therapeutic use , Hypopituitarism/drug therapy , Metabolic Diseases/etiology , Human Growth Hormone/deficiency , Humans , Hypoglycemic Agents/therapeutic use , Hypopituitarism/complications , Hypopituitarism/metabolism , Insulin/pharmacology , Insulin Resistance/physiology , Intra-Abdominal Fat/drug effects , Intra-Abdominal Fat/metabolism , Models, Biological , Receptor Cross-Talk/drug effects , Receptor Cross-Talk/physiology
14.
Diabetologia ; 51(1): 155-64, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17960360

ABSTRACT

AIM/HYPOTHESIS: The distinct metabolic properties of visceral and subcutaneous adipocytes may be due to inherent characteristics of the cells that are resident in each fat depot. To test this hypothesis, human adipocytes were differentiated in vitro from precursor stromal cells obtained from visceral and subcutaneous fat depots and analysed for genetic, biochemical and metabolic endpoints. METHODS: Stromal cells were isolated from adipose tissue depots of nondiabetic individuals. mRNA levels of adipocyte-specific proteins were determined by real-time RT-PCR. Insulin signalling was evaluated by immunoblotting with specific antibodies. Glucose transport was measured by a 2-deoxy-glucose uptake assay. Adiponectin secretion in the adipocyte-conditioned medium was determined by a specific RIA. RESULTS: With cell differentiation, mRNA levels of PPARG, C/EBPalpha (also known as CEBPA), AP2 (also known as GTF3A), GLUT4 (also known as SLC2A4) were markedly upregulated, whereas GLUT1 (also known as SLC2A1) mRNA did not change. However, expression of C/EBPalpha, AP2 and adiponectin was higher in subcutaneous than in visceral adipocytes. By contrast, adiponectin was secreted at threefold higher rates by visceral than by subcutaneous adipocytes while visceral adipocytes also showed two- to threefold higher insulin-stimulated glucose uptake. Insulin-induced phosphorylation of the insulin receptor, IRS proteins, Akt and extracellular signal-regulated kinase-1/2 was more rapid and tended to decrease at earlier time-points in visceral than in subcutaneous adipocytes. CONCLUSIONS/INTERPRETATION: Subcutaneous and visceral adipocytes, also when differentiated in vitro from precursor stromal cells, retain differences in gene expression, adiponectin secretion, and insulin action and signalling. Thus, the precursor cells that reside in the visceral and subcutaneous fat depots may already possess inherent and specific metabolic characteristics that will be expressed upon completion of the differentiation programme.


Subject(s)
Adipocytes/metabolism , Adiponectin/metabolism , Gene Expression Profiling , Gene Expression Regulation , Insulin/metabolism , Stromal Cells/cytology , Adipose Tissue/metabolism , Adult , Female , Glucose/metabolism , Humans , In Vitro Techniques , Male , Middle Aged , Radioimmunoassay , Signal Transduction , Stromal Cells/metabolism
15.
J Endocrinol Invest ; 28(3 Suppl): 85-93, 2005.
Article in English | MEDLINE | ID: mdl-16042365

ABSTRACT

DHEA and its sulfate prohormone DHEAS are the most abundant circulating adrenal steroid hormones in humans. DHEA exerts its actions on peripheral target tissues either indirectly, following its conversion to androgens, estrogens or both, or directly, as a steroid hormone interacting with either a nuclear or a membrane receptor. In humans, DHEA shows a characteristic pattern of secretion throughout life. Serum DHEA concentrations decline with advancing age and vary with gender, ethnicity, and environmental factors. Epidemiological studies show an inverse relationship between plasma DHEA(S) levels in men and age-related illnesses, including cardiovascular and metabolic diseases, immune disorders, malignancies, and neurological dysfunction. This has generated great interest on the putative role of DHEA in age-associated illnesses. Administration of DHEA to rats and mice reduces visceral fat accumulation, and improves insulin resistance in experimental models of diet-induced obesity and/or Type 2 diabetes. In addition, recent studies in vitro have shown that DHEA has the capacity to improve endothelial function by increasing nitric oxide (NO) synthesis. Replacement of DHEA in patients with adrenal insufficiency has been shown to exert beneficial effects on well-being, mood, and sexuality. By contrast, in healthy individuals, the physiological age-associated decline in circulating DHEA(S) per se does not justify DHEA supplementation, since the effects of this hormone on metabolic abnormalities, endothelial function in vivo, and cardiovascular events are contradictory. However, these results do not exclude the possibility that DHEA treatment may prove beneficial in specific subgroups of elderly subjects.


Subject(s)
Aging , Dehydroepiandrosterone Sulfate/blood , Adrenal Insufficiency , Affect , Animals , Body Composition/drug effects , Bone Density , Dehydroepiandrosterone Sulfate/administration & dosage , Dehydroepiandrosterone Sulfate/metabolism , Diabetes Mellitus, Type 2 , Endothelium, Vascular/drug effects , Hormone Replacement Therapy , Humans , Insulin Resistance , Male , Neoplasms , Obesity
16.
Acta Physiol Scand ; 183(1): 13-30, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15654917

ABSTRACT

Adipose tissue is now recognized to have a multitude of functions that are of importance in the regulation of energy balance and substrate metabolism. Different hormones, in particular insulin and catecholamines, govern the storage and utilization of energy in the triglyceride depots. In addition, adipocytes produce several different substances with endocrine or paracrine functions, which regulate the overall energetic homeostasis. With excess energy storage, obesity develops, leading to increased risk for type 2 diabetes and cardiovascular disease. The distribution of body fat appears to be even more important than the total amount of fat. Abdominal and, in particular, visceral adiposity is strongly linked to insulin resistance, type 2 diabetes, hypertension and dyslipidaemia, leading to increased risk of cardiovascular disease. The adverse metabolic impact of visceral fat has been attributed to distinct biological properties of adipocytes in this depot compared with other adipose tissue depots. Indeed, regional variations in the metabolic activity of fat cells have been observed. Furthermore, expression studies aiming at defining the unique biological properties of adipose tissues from distinct anatomical sites have identified depot-related differences in the protein content of fat-produced molecules. In this review we wish to summarize important results from the literature and also some recent data from our own work. The main scope is to describe the biological functions of adipose tissue, and to focus on metabolic, hormonal, and signalling differences between fat depots.


Subject(s)
Adipose Tissue/metabolism , Insulin/metabolism , Adipocytes/metabolism , Autonomic Nervous System/metabolism , Catecholamines/metabolism , Diabetes Mellitus, Type 2/metabolism , Glucocorticoids/metabolism , Glucose/metabolism , Humans , Insulin Resistance/physiology , Lipids/biosynthesis , Lipolysis/physiology , Signal Transduction/physiology
17.
Diabetologia ; 47(6): 1020-8, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15170497

ABSTRACT

AIMS/HYPOTHESIS: Type 1 diabetic patients who develop microalbuminuria are clearly disadvantaged in terms of their risk of morbidity and mortality from renal and cardiovascular diseases. It is therefore important to identify potential factors that can predict progression to macroalbuminuria. METHODS: This is a 7-year follow-up study of 352 microalbuminuric Type 1 diabetic patients from 31 European centres. Risk factors at baseline were compared in patients who progressed to macroalbuminuria and in patients who remained microalbuminuric or reverted to normoalbuminuria. Risk factors and albumin excretion rate (AER) were measured centrally. RESULTS: Over 7.3 years, 13.9% of the microalbuminuric patients progressed to macroalbuminuria, 35.5% remained microalbuminuric and 50.6% reverted to normoalbuminuria. Independent baseline risk factors for progression to macroalbuminuria were HbA(1)c (7.9% vs 6.8%, p=0.004), AER (64.4 vs 44.9 microg/min, p=0.0001) and-after adjusting for diabetes duration, HbA(1)c and AER-body weight (72 vs 67 kg, p=0.05). Independent factors associated with regression to normoalbuminuria were diabetes duration (15 vs 18 years, p=0.004), AER (37.2 vs 44.9 microg/min, p=0.0001) and-after adjusting for diabetes duration, HbA(1)c and AER-waist-to-hip ratio (0.83 vs 0.86, p=0.05) and incidence of peripheral neuropathy at baseline (24% vs 38%, p=0.001). Blood pressure and smoking did not emerge as risk factors at baseline for the outcome of microalbuminuria. CONCLUSIONS/INTERPRETATION: A significant fraction of microalbuminuric Type 1 diabetic patients will progress to overt proteinuria. Patients with higher AER values, sub-optimal metabolic control, excess body fat and peripheral neuropathy may carry a particularly high risk of clinical nephropathy requiring aggressive therapeutic intervention.


Subject(s)
Albuminuria/complications , Diabetes Mellitus, Type 1/complications , Disease Progression , Prospective Studies , Risk Factors , Adult , Albuminuria/blood , Albuminuria/drug therapy , Diabetes Mellitus, Type 1/blood , Diabetes Mellitus, Type 1/drug therapy , Diabetic Neuropathies/complications , Diabetic Neuropathies/epidemiology , Diabetic Neuropathies/metabolism , Diabetic Retinopathy/complications , Diabetic Retinopathy/epidemiology , Diabetic Retinopathy/metabolism , Female , Follow-Up Studies , Glycated Hemoglobin/chemistry , Humans , Male , Multivariate Analysis , Patient Selection , Proteinuria/etiology , Proteinuria/physiopathology , Serum Albumin/chemistry , Serum Albumin/metabolism , Time Factors , Triglycerides/blood , gamma-Glutamyltransferase/blood , von Willebrand Factor/chemistry
18.
Diabetes ; 50(12): 2709-20, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11723053

ABSTRACT

The insulin signaling cascade was investigated in rat myocardium in vivo in the presence of streptozocin (STZ)-induced diabetes and after diabetes treatment by islet transplantation under the kidney capsule. The levels of insulin-stimulated tyrosine phosphorylation of the insulin receptor beta-subunit, insulin receptor substrate (IRS)-2, and p52(Shc) were increased in diabetic compared with control heart, whereas tyrosine phosphorylation of IRS-1 was unchanged. The amount of the p85 subunit of phosphatidylinositol 3-kinase (PI 3-kinase) and the level of PI 3-kinase activity associated with IRS-2 were also elevated in diabetes, whereas no changes in IRS-1-associated PI 3-kinase were observed. Insulin-induced phosphorylation of Akt on Thr-308 was increased fivefold in diabetic heart, whereas Akt phosphorylation on Ser-473 was normal. In contrast with Akt phosphorylation, insulin-induced phosphorylation of glycogen synthase kinase (GSK)-3, a major cellular substrate of Akt, was markedly reduced in diabetes. In islet-transplanted rats, the majority of the alterations in insulin-signaling proteins found in diabetic rats were normalized, but insulin stimulation of IRS-2 tyrosine phosphorylation and association with PI 3-kinase was blunted. In conclusion, in the diabetic heart, 1) IRS-1, IRS-2, and p52(Shc) are differently altered, 2) the levels of Akt phosphorylation on Ser-473 and Thr-308, respectively, are not coordinately regulated, and 3) the increased activity of proximal-signaling proteins (i.e., IRS-2 and PI 3-kinase) is not propagated distally to GSK-3. Islet transplantation under the kidney capsule is a potentially effective therapy to correct several diabetes-induced abnormalities of insulin signaling in cardiac muscle but does not restore the responsiveness of all signaling reactions to insulin.


Subject(s)
Adaptor Proteins, Signal Transducing , Adaptor Proteins, Vesicular Transport , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/surgery , Insulin/metabolism , Islets of Langerhans Transplantation , Protein Serine-Threonine Kinases , Signal Transduction , Animals , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Glycogen Synthase Kinase 3 , Glycogen Synthase Kinases , Insulin Receptor Substrate Proteins , Intracellular Signaling Peptides and Proteins , Male , Mitogen-Activated Protein Kinase Kinases/metabolism , Mitogen-Activated Protein Kinases/metabolism , Myocardium/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphoproteins/metabolism , Phosphorylation , Phosphoserine/metabolism , Phosphotyrosine/metabolism , Proteins/metabolism , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , Rats , Rats, Inbred Lew , Receptor, Insulin/metabolism , Shc Signaling Adaptor Proteins , Src Homology 2 Domain-Containing, Transforming Protein 1
19.
Proc Natl Acad Sci U S A ; 97(3): 1125-30, 2000 Feb 01.
Article in English | MEDLINE | ID: mdl-10655495

ABSTRACT

Glucose transport in insulin-regulated tissues is mediated by the GLUT4 and GLUT1 transporters. Using the yeast two-hybrid system, we have cloned the sentrin-conjugating enzyme mUbc9 as a protein that interacts with the GLUT4 COOH-terminal intracellular domain. The mUbc9 enzyme was found to bind directly to GLUT4 and GLUT1 through an 11-aa sequence common to the two transporters and to modify both transporters covalently by conjugation with the mUbc9 substrate, sentrin. Overexpression of mUbc9 in L6 skeletal muscle cells decreased GLUT1 transporter abundance 65%, resulting in decreased basal glucose transport. By contrast, mUbc9 overexpression increased GLUT4 abundance 8-fold, leading to enhanced transport stimulation by insulin. A dominant-negative mUbc9 mutant lacking catalytic activity had effects opposite to those of wild-type mUbc9. The regulation of GLUT4 and GLUT1 was specific, as evidenced by an absence of mUbc9 interaction with or regulation of the GLUT3 transporter isoform in L6 skeletal muscle cells. The mUbc9 sentrin-conjugating enzyme represents a novel regulator of GLUT1 and GLUT4 protein levels with potential importance as a determinant of basal and insulin-stimulated glucose uptake in normal and pathophysiological states.


Subject(s)
Gene Expression Regulation , Ligases/metabolism , Monosaccharide Transport Proteins/metabolism , Muscle Proteins , Muscle, Skeletal/metabolism , Ubiquitin-Conjugating Enzymes , Ubiquitins/metabolism , 3T3 Cells , Amino Acid Sequence , Animals , Biological Transport, Active/drug effects , Glucose/metabolism , Glucose Transporter Type 1 , Glucose Transporter Type 4 , Insulin/pharmacology , Macromolecular Substances , Mice , Molecular Sequence Data , Rats , Recombinant Fusion Proteins/metabolism , SUMO-1 Protein
20.
Diabetes ; 48(4): 801-12, 1999 Apr.
Article in English | MEDLINE | ID: mdl-10102697

ABSTRACT

Insulin-dependent diabetes in rats is characterized by abnormalities of post-binding insulin signaling reactions that are not fully corrected by exogenous insulin therapy. The aim of this study was to investigate the effects of islet transplantation on insulin signaling in skeletal muscle and myocardium of streptozocin (STZ)-induced diabetic rats. Control rats, untreated diabetic rats, and diabetic rats transplanted with syngeneic islets under the kidney capsule were studied. Compared with controls, diabetic rats were characterized by multiple insulin signaling abnormalities in skeletal muscle, which included 1) increased insulin-stimulated tyrosine phosphorylation of the insulin receptor beta-subunit and insulin receptor substrates IRS-1 and IRS-2, 2) increased substrate tyrosine phosphorylation in the basal state, 3) a decreased amount of IRS-1 protein, 4) markedly elevated basal and insulin-stimulated phosphatidylinositol (PI) 3-kinase activity in anti-IRS-1 immunoprecipitates from total tissue extracts, and 5) increased PI 3-kinase activity in low-density microsomes. A similar augmentation of insulin receptor and substrate tyrosine phosphorylation in response to STZ-diabetes was also found in myocardium, although with lower magnitude than that found in skeletal muscle. In addition, STZ-diabetes resulted in decreased IRS-1 and increased IRS-2 protein levels in myocardium. Islet transplantation fully corrected the diabetes-induced changes in protein tyrosine phosphorylation and PI 3-kinase activity and normalized IRS-1 and IRS-2 protein content in both skeletal muscle and myocardium. Thus, insulin delivered into the systemic circulation by pancreatic islets transplanted under the kidney capsule can adequately correct altered insulin signaling mechanisms in insulinopenic diabetes.


Subject(s)
Diabetes Mellitus, Experimental/surgery , Islets of Langerhans Transplantation , Muscle, Skeletal/metabolism , Myocardium/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Receptor, Insulin/metabolism , Tyrosine/metabolism , Animals , Insulin/physiology , Insulin Receptor Substrate Proteins , Intracellular Signaling Peptides and Proteins , Male , Phosphoproteins/metabolism , Phosphorylation , Rats , Rats, Inbred Lew , Signal Transduction/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...