Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 84
Filter
1.
J Endocrinol Invest ; 2024 Apr 29.
Article in English | MEDLINE | ID: mdl-38683498

ABSTRACT

PURPOSE: To evaluate the variables influencing the therapeutic choice toward oral versus subcutaneous semaglutide in a cohort of diabetic subjects. METHODS: We retrospectively collected data of 292 patients followed at the Diabetes Unit of the University Hospital of Siena and the Hospital of Grosseto, who were prescribed oral (n = 115) or subcutaneous (n = 177) semaglutide between October 2021 and October 2022. RESULTS: Oral semaglutide was preferentially prescribed in older subjects with longer disease duration in replacement of other antidiabetic drugs, while subcutaneous semaglutide was preferentially prescribed in add-on to metformin in subjects with higher body weight and BMI. After 6 months, both formulations significantly improved glycemic control and body weight, however injectable semaglutide showed a greater efficacy on A1c levels, weight loss, BMI and waist circumference reduction. No differences were found in terms of adverse events. CONCLUSION: In our experience, injectable semaglutide is preferred in patients with excess weight and shorter disease duration, while the oral formulation was used later and especially after therapeutic failure of previous therapies. Follow-up data indicate similar tolerability and efficacy of both formulations, despite subcutaneous semaglutide demonstrated greater efficacy.

3.
J Endocrinol Invest ; 46(12): 2583-2599, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37286863

ABSTRACT

PURPOSE/METHODS: The determination of tumour biomarkers is paramount to advancing personalized medicine, more so in rare tumours like medullary thyroid carcinoma (MTC), whose diagnosis is still challenging. The aim of this study was to identify non-invasive circulating biomarkers in MTC. To achieve this goal, paired MTC tissue and plasma extracellular vesicle samples were collected from multiple centres and microRNA (miRNA) expression levels were evaluated. RESULTS: The samples from a discovery cohort of 23 MTC patients were analysed using miRNA arrays. Lasso logistic regression analysis resulted in the identification of a set of circulating miRNAs as diagnostic biomarkers. Among them, miR-26b-5p and miR-451a, were highly expressed and their expression decreased during follow-up in disease-free patients in the discovery cohort. Circulating miR-26b-5p and miR-451a were validated using droplet digital PCR in a second independent cohort of 12 MTC patients. CONCLUSION: This study allowed the identification and validation of a signature of two circulating miRNAs, miR-26b-5p and miR-451a, in two independent cohorts reporting a significant diagnostic performance for MTC. The results of this study offer advancements in molecular diagnosis of MTC proposing a novel non-invasive tool to use in precision medicine.


Subject(s)
Circulating MicroRNA , MicroRNAs , Thyroid Neoplasms , Humans , MicroRNAs/genetics , Thyroid Neoplasms/diagnosis , Thyroid Neoplasms/genetics , Thyroid Neoplasms/pathology , Biomarkers , Biomarkers, Tumor/metabolism
4.
Cytokine ; 141: 155455, 2021 05.
Article in English | MEDLINE | ID: mdl-33548798

ABSTRACT

BACKGROUND: Severe acute respiratory syndrome caused by novel coronavirus 2 (SARS-CoV-2) emerged in Wuhan (China) in December 2019. Here we evaluated a panel of biomarkers to phenotype patients and to define the role of immuno-inflammatory mediators as biomarkers of severity. MATERIALS AND METHODS: Serum samples were obtained from 24 COVID-19 patients on admission to hospital, before any treatment or infusion of intravenous steroids or invasive ventilation. KL-6 IL-6 and C-peptide were measured by chemiluminescent enzyme immunoassay. IL-6 assay was validated for accuracy and precision. The validity of variables used to distinguish severe from mild-to-moderate patients was assessed by areas under curves (AUC) of the receiver operating characteristic (ROC) and logistic regression was performed to combine parameters of the two groups. RESULTS: In the severe group, IL-6, CRP and KL-6 concentrations were significantly higher than in mild-to-moderate patients. KL-6, IL-6 and CRP concentrations were directly correlated with each other. ROC curve analysis of the logistic regression model including IL-6, KL-6 and CRP showed the best performance with an AUC of 0.95. CONCLUSIONS: Besides corroborating previous reports of over-expression of IL-6 in severe COVID-19 patients requiring mechanical ventilation, analytical determination of other mediators showed that IL-6 concentrations were correlated with those of KL-6 and CRP. The combination of these three prognostic bioindicators made it possible to distinguish severe COVID-19 patients with poor prognosis from mild-to-moderate patients.


Subject(s)
Biomarkers/blood , COVID-19/blood , COVID-19/immunology , Cytokines/blood , Pandemics , SARS-CoV-2 , Aged , C-Peptide/blood , C-Reactive Protein/metabolism , COVID-19/epidemiology , Case-Control Studies , Female , Humans , Inflammation Mediators/blood , Interleukin-6/blood , Italy/epidemiology , Male , Middle Aged , Mucin-1/blood , Prognosis , Severity of Illness Index
5.
Diabetes Metab ; 43(6): 512-520, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28499695

ABSTRACT

AIM: SGLT2 inhibitors reduce renal glucose uptake through an insulin-independent mechanism. They also increase glucagon concentration, although the extent to which this is due to a direct effect on pancreatic alpha cells remains unclear. METHODS: In the present work, αTC1 cells treated with the SGLT2 inhibitor dapagliflozin (Dapa) were analyzed for glucose transporters, molecular mediators of hormone secretion, glucagon and GLP-1 release, and the effects of somatostatin. Data were validated in murine and human pancreatic islets. RESULTS: SLC5A2 (the SGLT2-encoding gene) was nearly undetectable in αTC1 cells, not even by a digital PCR technique using different probes. In contrast, SLC5A1 (the SGLT1-encoding gene) was constitutively abundant in αTC1 cells and in islets, and increased with Dapa. This was associated with greater glucagon release, preceded by increased expression of preproglucagon and HNF4α. Looking at the candidate intracellular signalling pathway, reduced PASK and increased AMPK-α2 expression were also detected. GLUT1 and GLUT2, as well as regulators of glucagon release and alpha-cell phenotype (chromogranin A, paired box 6, proprotein convertase 1/2, synaptophysin), were unaffected by Dapa, as were GLP-1 receptor expression and GLP-1 release. Low glucose did not influence the stimulatory effect of Dapa on glucagon release, but was instead almost fully reverted by SLC5A1 silencing. When the effect of Dapa on AMPK and PASK, emerging regulators of lipid and glucose metabolism, was tested, upregulated AMPK-α2 appeared to be involved in molecular signalling. CONCLUSION: Our study has shown that, in αTC1 cells, Dapa acutely upregulates SGLT1 expression and increases glucagon release through an SGLT1-dependent mechanism, with SGLT2 expression virtually undetectable. These results suggest the involvement of SGLT1 in modulating glucagon increases following SGLT2 inhibition.


Subject(s)
Benzhydryl Compounds/pharmacology , Glucagon-Secreting Cells/drug effects , Glucagon/metabolism , Glucosides/pharmacology , Sodium-Glucose Transporter 1/metabolism , Sodium-Glucose Transporter 2 Inhibitors , Sodium-Glucose Transporter 2/metabolism , Animals , Cell Line , Cells, Cultured , Gene Silencing , Glucagon-Like Peptide 1/metabolism , Glucagon-Secreting Cells/metabolism , Glucose/metabolism , Humans , Mice , Signal Processing, Computer-Assisted , Sodium-Glucose Transporter 1/genetics , Somatostatin
6.
J Endocrinol Invest ; 40(6): 591-610, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28213644

ABSTRACT

Diabetes is a complex, multifactorial group of metabolic diseases characterized by chronic hyperglycaemia due to pancreatic beta-cell dysfunction and/or loss. It is characterized by an asymptomatic and highly variable prodromic phase, which renders diabetes mellitus difficult to be predicted with sufficient accuracy. Despite several efforts in the identification and standardization of newly trustable. Biomarkers able to predict and follow-up diabetes and to specifically subtype its different forms, few of them have proven of clinical utility. Recently, a new class of endogenous non-coding small RNAs, namely microRNAs, have been indicated as putative biomarkers, being released by cells and tissues and found in a cell-free circulating form in many biological fluids, including serum and/or plasma. MicroRNAs have been initially identified as promising biomarkers in cancer, and nowadays their application has been extended to other diseases, including diabetes. Although an increasing number of studies focused on the evaluation of circulating microRNAs in diabetes, few reproducibly identified microRNAs as biomarkers for disease prediction or follow-up. Technological problems as well as the need to obtain highly standardized operating procedures and methods are still an issue in such research field. In this review, we comprehensively resume the main and most recent findings on circulating microRNAs, and their possible use as biomarkers to predict and follow-up diabetes and its complications, as well as the methodological challenges to standardize accurate operating procedures for their analysis.


Subject(s)
Biomarkers/blood , Circulating MicroRNA/genetics , Diabetes Mellitus/classification , Diabetes Mellitus/diagnosis , Circulating MicroRNA/blood , Diabetes Mellitus/blood , Diabetes Mellitus/genetics , Humans
7.
Diabetes Obes Metab ; 17(7): 689-98, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25846577

ABSTRACT

AIMS: To evaluate third-line thiazolidinedione (TZD) or glimepiride therapy in patients inadequately controlled on metformin + exenatide twice daily, and third-line exenatide twice daily in patients inadequately controlled on metformin + glimepiride. METHODS: In this randomized, open-label, multicentre trial, 144 patients with type 2 diabetes inadequately controlled [glycated haemoglobin (HbA1c) >9% (75 mmol/mol) after 3 months' treatment or >7% (53 mmol/mol) at two consecutive visits 3 months apart, after 6 months' treatment] on metformin + exenatide twice daily were re-randomized to add-on TZD or glimepiride, and 166 patients inadequately controlled on metformin + glimepiride received add-on exenatide twice daily. Changes in HbA1c, body mass index (BMI), lipids, hypoglycaemia and vital signs were evaluated. RESULTS: The median duration of triple therapy was ∼2 years. In patients inadequately controlled on metformin + exenatide twice daily, add-on TZD decreased HbA1c levels significantly better than add-on glimepiride: 130-week difference 0.48% [95% confidence interval (CI) 0.19-0.77] or 5.2 mmol/mol (95% CI 2.1-8.4; p = 0.001), but with significantly increased BMI and systolic blood pressure. The ratio of documented symptomatic (blood glucose ≤70 mg/dl [3.9 mmol/l]) hypoglycaemia rates for add-on glimepiride to add-on TZD was 8.48 (p < 0.0001). Add-on exenatide twice daily after metformin + glimepiride significantly reduced HbA1c levels: mean [standard deviation (s.d.)] change from baseline -0.35 (0.89)% [-3.8 (9.7) mmol/mol] and BMI: mean (s.d.) change from baseline -0.82 (1.9) kg/m(2) at 130 weeks, with a slightly increased rate of documented symptomatic hypoglycaemia from metformin + glimepiride (ratio 1.49). CONCLUSIONS: TZD, but not glimepiride, was an effective and well tolerated third-line therapy in patients without glycaemic control after long-term therapy with metformin + exenatide twice daily. Exenatide twice daily was an effective and well tolerated third-line therapy in patients inadequately controlled on metformin + glimepiride.


Subject(s)
Diabetes Mellitus, Type 2/drug therapy , Hypoglycemic Agents/administration & dosage , Metformin/administration & dosage , Peptides/administration & dosage , Sulfonylurea Compounds/administration & dosage , Thiazolidinediones/administration & dosage , Venoms/administration & dosage , Adult , Aged , Blood Glucose/analysis , Body Mass Index , Diabetes Mellitus, Type 2/blood , Drug Administration Schedule , Drug Therapy, Combination , Europe , Exenatide , Female , Glycated Hemoglobin/analysis , Humans , Hypoglycemia/chemically induced , Lipids/blood , Male , Middle Aged , Prospective Studies , Treatment Failure
8.
Diabetologia ; 55(12): 3262-72, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22965295

ABSTRACT

AIMS/HYPOTHESIS: Glucagon-like peptide 1 (GLP-1) is a major incretin, mainly produced by the intestinal L cells, with beneficial actions on pancreatic beta cells. However, while in vivo only very small amounts of GLP-1 reach the pancreas in bioactive form, some observations indicate that GLP-1 may also be produced in the islets. We performed comprehensive morphological, functional and molecular studies to evaluate the presence and various features of a local GLP-1 system in human pancreatic islet cells, including those from type 2 diabetic patients. METHODS: The presence of insulin, glucagon, GLP-1, proconvertase (PC) 1/3 and PC2 was determined in human pancreas by immunohistochemistry with confocal microscopy. Islets were isolated from non-diabetic and type 2 diabetic donors. GLP-1 protein abundance was evaluated by immunoblotting and matrix-assisted laser desorption-ionisation-time of flight (MALDI-TOF) mass spectrometry. Single alpha and beta cell suspensions were obtained by enzymatic dissociation and FACS sorting. Glucagon and GLP-1 release were measured in response to nutrients. RESULTS: Confocal microscopy showed the presence of GLP-1-like and PC1/3 immunoreactivity in subsets of alpha cells, whereas GLP-1 was not observed in beta cells. The presence of GLP-1 in isolated islets was confirmed by immunoblotting, followed by mass spectrometry. Isolated islets and alpha (but not beta) cell fractions released GLP-1, which was regulated by glucose and arginine. PC1/3 (also known as PCSK1) gene expression was shown in alpha cells. GLP-1 release was significantly higher from type 2 diabetic than from non-diabetic isolated islets. CONCLUSIONS/INTERPRETATION: We have shown the presence of a functionally competent GLP-1 system in human pancreatic islets, which resides in alpha cells and might be modulated by type 2 diabetes.


Subject(s)
Diabetes Mellitus, Type 2/metabolism , Glucagon-Like Peptide 1/metabolism , Glucagon-Secreting Cells/metabolism , Glucagon/metabolism , Insulin/metabolism , Female , Humans , Immunohistochemistry , Male , Mass Spectrometry , Middle Aged , Pancreas/metabolism
9.
Clin Exp Immunol ; 168(1): 24-9, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22385233

ABSTRACT

Type 1 diabetes mellitus (T1DM) is a multi-factorial immune-mediated disease characterized by the autoimmune destruction of insulin-producing pancreatic islet beta cells in genetically susceptible individuals. Epidemiological evidence has also documented the constant rise in the incidence of T1DM worldwide, with viral infections representing one of the candidate environmental risk factors identified by several independent studies. In fact, epidemiological data showed that T1DM incidence increases after epidemics due to enteroviruses and that enteroviral RNA can be detected in the blood of >50% of T1DM patients at the time of disease onset. Furthermore, both in-vitro and ex-vivo studies have shown that viruses can infect pancreatic beta cells with consequent effects ranging from functional damage to cell death.


Subject(s)
Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/virology , Enterovirus Infections/immunology , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/virology , Animals , Diabetes Mellitus, Type 1/epidemiology , Enterovirus/immunology , Enterovirus/pathogenicity , Enterovirus Infections/complications , Enterovirus Infections/virology , Humans , Insulin-Secreting Cells/immunology , Mice , Nitric Oxide Synthase Type II/biosynthesis , Nitric Oxide Synthase Type II/metabolism , RNA, Viral/blood , Rats
10.
Acta Diabetol ; 49 Suppl 1: S123-31, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22249339

ABSTRACT

Post-transplant diabetes mellitus represents an important complication of prolonged immunosuppressive treatment after solid organ transplantation. The immunosuppressive toxicity, responsible for a persistent impairment of glucose metabolism in pancreatic islet-transplanted patients, is mainly attributed to calcineurin inhibitors and steroids, while other immunosuppressive molecules (azathioprine and mycophenolic acid, MPA) are considered not to have a toxic effect. In the present study, in vitro effects of MPA have been investigated in mouse beta-cell lines (ßTC-1 and ßTC-6) and in purified human pancreatic islets. ßTC-1, ßTC-6, and human pancreatic islets were exposed to various concentrations of MPA for different times. Consequently, we evaluated the viability, the induction of apoptosis, the glucose-stimulated insulin secretion, and the expression of ß-cell function genes (Isl1, Pax6, Glut-2, glucokinase) and apoptosis-related genes (Bax and Bcl2). ßTC-1, ßTC-6, and human islets treated, respectively, for 48 and 72 h with 15-30 nM MPA showed altered islet architecture, as compared with control cells. We observed for ßTC-1 and ßTC-6 almost 70% reduction in cell viability; three to sixfold induction of TUNEL/apoptotic-positive cells quantified by FACS analysis. A twofold increase in apoptotic cells was observed in human islets after MPA exposure associated with strong inhibition of glucose-stimulated insulin secretion. Furthermore, we showed significant down-regulation of gene expression of molecules involved in ß-cell function and increase rate between Bax/Bcl2. Our data demonstrate that MPA has an in vitro diabetogenic effect interfering at multiple levels with survival and function of murine and human pancreatic ß-cells.


Subject(s)
Gene Expression/drug effects , Immunosuppressive Agents/pharmacology , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/drug effects , Mycophenolic Acid/pharmacology , Adolescent , Adult , Animals , Apoptosis/drug effects , Cell Line , Cell Survival/drug effects , Cells, Cultured , Eye Proteins/genetics , Eye Proteins/metabolism , Female , Glucose Transporter Type 2/genetics , Glucose Transporter Type 2/metabolism , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Insulin/metabolism , Insulin Secretion , Insulin-Secreting Cells/metabolism , Islets of Langerhans/cytology , Islets of Langerhans/drug effects , Male , Mice , Middle Aged , PAX6 Transcription Factor , Paired Box Transcription Factors/genetics , Paired Box Transcription Factors/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Young Adult
12.
Clin Exp Immunol ; 166(3): 317-24, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22059988

ABSTRACT

Highly sensitive and specific radioimmunoassays have been validated for autoantibodies reacting with the four major autoantigens identified so far in autoimmune diabetes. However, the analysis of this large number of autoantigens has increased the costs and time necessary for complete autoantibody screenings. Our aim was to demonstrate that it is possible to detect the immunoreactivity against a combination of four different autoantigens by a single assay, this representing a rapid, low-cost first approach to evaluate humoral autoimmunity in diabetes. By using this novel multi-autoantigen radioimmunoassay (MAA), in subsequent steps we analysed 830 sera, 476 of known and 354 of unknown diabetes-specific immunoreactivity, collected from various groups of individuals including type 1 and type 2 diabetes patients, autoantibody-positive patients with a clinical diagnosis of type 2 diabetes (LADA), prediabetic subjects, individuals at risk to develop autoimmune diabetes, siblings of type 1 diabetic patients, coeliac patients and healthy control subjects. All sera reacting with one or more of the four autoantigens by single assays also resulted positive with MAA, as well as eight of 24 type 1 diabetic patients classified initially as autoantibody-negative at disease onset based on single autoantibody assays. In addition, MAA showed 92% sensitivity and 99% specificity by analysing 140 blinded sera from type 1 diabetic patients and control subjects provided in the 2010 Diabetes Autoantibody Standardization Program. MAA is the first combined method also able to evaluate, in addition to glutamic acid decarboxylase (GAD) and tyrosine phosphatase (IA)-2, insulin and islet beta-cell zinc cation efflux transporter (ZnT8) autoantibodies. It appears to be particularly appropriate as a first-line approach for large-scale population-based screenings of anti-islet autoimmunity.


Subject(s)
Autoantibodies/blood , Diabetes Mellitus, Type 1/diagnosis , Radioimmunoassay/methods , Adult , Aged , Autoantibodies/immunology , Autoantigens/immunology , Cation Transport Proteins/immunology , Diabetes Mellitus, Type 1/immunology , Female , Glutamate Decarboxylase/immunology , High-Throughput Screening Assays/methods , Humans , Insulin/immunology , Male , Middle Aged , Protein Tyrosine Phosphatases/immunology , Sensitivity and Specificity
13.
Diabetes Metab Res Rev ; 27(8): 755-60, 2011 Nov.
Article in English | MEDLINE | ID: mdl-22069255

ABSTRACT

BACKGROUND: Hedgehog pathway plays an important role during pancreas development, when its inactivation is crucial to assure expression of pancreatic marker genes involved in the organ formation and to assure an appropriate organogenesis. Patched1 (Ptch1) is a transmembrane receptor of hedgehog pathway which has a key role in this process. In fact, heterozygous Ptch1 mutant (ptc+/-) mice are affected by an impaired glucose tolerance accompanied by reduced islet function. In the light that the cell distribution of Ptch1 receptor within the endocrine pancreas has not yet been established, we aimed at identifying the pancreatic endocrine cell subset(s) expressing such molecule. METHODS: Double immunostaining for Ptch1 and pancreatic hormones insulin, glucagon and somatostatin on pancreatic paraffin sections of C57BL/6J mice and human non-diabetic multiorgan donors was performed and analysed using confocal microscopy. In addition, diabetes was experimentally induced in mice by intraperitoneal injection of streptozotocin. Quantitative real-time polymerase chain reaction after laser-capture microdissection of different islets from frozen pancreatic murine tissue sections was also performed. RESULTS: Ptch1 receptor was detected only in somatostatin-positive delta cells both in mice and in human pancreas; in mice its expression was not affected by streptozotocin treatment. A significant increase of Ptch1 mRNA expression levels in the islet periphery versus the islet core was observed by quantitative real-time polymerase chain reaction, in accord with immunohistochemical observations. CONCLUSION: Our data show a delta-cell-specific expression of Ptch1 receptor in murine and human pancreas.


Subject(s)
Islets of Langerhans/metabolism , Receptors, Cell Surface/biosynthesis , Somatostatin-Secreting Cells/metabolism , Animals , Humans , Islets of Langerhans/cytology , Male , Mice , Mice, Inbred C57BL , Patched Receptors , Patched-1 Receptor
14.
Transplant Proc ; 43(1): 330-2, 2011.
Article in English | MEDLINE | ID: mdl-21335216

ABSTRACT

Type 1 diabetes (T1D) is a polygenic disorder where loci within the human leukocyte antigen (HLA) account for most of the genetic susceptibility. Nongenetic factors, most likely environmental, are also involved in the pathogenesis of the disease, resulting in T-cell-mediated autoimmune attack against pancreatic beta cells. Although our understanding of the natural history of T1D has significantly improved during the last decades, the pathogenesis of the disease remains elusive as are successful strategies for primary intervention. Interesting findings are expected from the emerging field of microRNAs (miRNAs), a family of endogenous small noncoding RNA molecules that regulate gene expression. They play a key role in post-transcriptional regulation by selectively binding complementary messenger RNAs, thus affecting translation. miRNAs affect key biological processes including cell proliferation, differentiation, development, and metabolism. In addition, miRNAs are also involved in the regulation of the immune system and insulin secretion. Interestingly, miRNAs have been identified in both normal and pathological conditions, functioning as predictive markers in certain human diseases. Herein, we have discussed the potential application of this new field to T1D. Research in this area may help to identify variations in genes coding for selected miRNAs that may contribute to diabetes susceptibility. In addition, mechanistic studies on the role of miRNAs in the modulation of the immune system may elucidate important regulatory mechanisms, identifying potential therapeutic targets to ameliorate responses to islet transplantation.


Subject(s)
Diabetes Mellitus, Type 1/therapy , Islets of Langerhans Transplantation , MicroRNAs , Diabetes Mellitus, Type 1/immunology , Humans , Immunosuppressive Agents/administration & dosage
16.
Nutr Metab Cardiovasc Dis ; 20(9): 683-90, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20934862

ABSTRACT

Diabetes and osteoporosis are common and complex disorders with an enormous health burden that can be often associated especially in middle-age and elderly individuals. Although there is raising awareness of the higher fractures rates among patients with type 1 (DM1) and 2 (DM2) diabetes, there are few data available on the pathogenetic mechanisms responsible for this increased risk. Importantly, several experimental and clinical observations suggest that bone abnormalities associated with diabetes may differ, at least in part, from those associated with senile or post-menopausal osteoporosis. This implies that specific preventive and therapeutic strategies have to be developed and tested to prevent fractures in DM1 and DM2 patients. It is also likely that shared (i.e. due to glucose-toxicity) as well as different (i.e. due to insulin levels or other hormones) mechanisms may be associated with bone fragility in DM1 and DM2. Moreover, the hypothesis of an endocrine role of the skeleton in the regulation of glucose metabolism and insulin sensitivity has been recently proposed by experimental observations. This review summarizes the recent clinical and experimental advances on glucose tolerance, bone fragility and osteoporosis associated with diabetes.


Subject(s)
Bone and Bones/physiopathology , Diabetes Mellitus, Type 1/complications , Diabetes Mellitus, Type 2/complications , Osteoporosis/complications , Aged , Bone Density , Bone Remodeling , Bone and Bones/drug effects , Diabetes Mellitus, Type 1/physiopathology , Diabetes Mellitus, Type 2/physiopathology , Female , Fractures, Bone/complications , Fractures, Bone/epidemiology , Fractures, Bone/prevention & control , Humans , Hyperglycemia/complications , Hypoglycemic Agents/therapeutic use , Male , Middle Aged , Osteoporosis/epidemiology , Risk Factors , Tensile Strength
17.
Clin Exp Immunol ; 162(3): 407-14, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20819089

ABSTRACT

Recent data show that regulatory cells with transforming growth factor (TGF)-ß1-dependent activity are able to restore self-tolerance in overtly diabetic non-obese diabetic (NOD) mice. Thus, TGF-ß1 seems to have a relevant role in protection from autoimmune diabetes. Our aim was to investigate the possible significance of serum TGF-ß1 measurement in the natural history of diabetes in NOD mice, as well as in children positive for at least one islet-related antibody. Serum TGF-ß1 (both total and active) was measured by enzyme-linked immunosorbent assay at monthly intervals in 26 NOD mice during the spontaneous development of diabetes and, on a yearly basis, in nine siblings of patients with type 1 diabetes (T1D) with a follow-up of 4 years. Diabetes appeared between the 12th week of age and the end of the study period (36 weeks) in 17 mice. TGF-ß1 serum level variations occurred in the prediabetic period in both NOD mice and humans and diabetes diagnosis followed a continuing reduction of active TGF-ß1 (aTGF-ß1) serum levels. In mice, aTGF-ß1 serum levels measured at 4 weeks of age correlated positively with severity of insulitis, and negatively with percentage of insulin-positive cells. Our findings suggest that in NOD mice serum TGF-ß1 levels during the natural history of the diabetes reflect the course of islet inflammation. The measurement of aTGF-ß1 in islet-related antibody-positive subjects may provide insights into the natural history of prediabetic phase of T1D.


Subject(s)
Diabetes Mellitus, Type 1/diagnosis , Pancreas/pathology , Transforming Growth Factor beta1/blood , Adolescent , Animals , Autoantibodies/blood , Child , Child, Preschool , Diabetes Mellitus, Type 1/blood , Diabetes Mellitus, Type 1/physiopathology , Disease Progression , Female , Follow-Up Studies , Humans , Inflammation , Male , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Pancreas/immunology
18.
Diabetologia ; 53(7): 1395-405, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20369226

ABSTRACT

AIMS/HYPOTHESIS: Beta cell failure is a crucial component in the pathogenesis of type 2 diabetes. One of the proposed mechanisms of beta cell failure is local inflammation, but the presence of pancreatic islet inflammation in type 2 diabetes and the mechanisms involved remain under debate. METHODS: Chemokine and cytokine expression was studied by microarray analysis of laser-capture microdissected islets from pancreases obtained from ten non-diabetic and ten type 2 diabetic donors, and by real-time PCR of human islets exposed to oleate or palmitate at 6 or 28 mmol/l glucose. The cellular source of the chemokines was analysed by immunofluorescence of pancreatic sections from individuals without diabetes and with type 2 diabetes. RESULTS: Microarray analysis of laser-capture microdissected beta cells showed increased chemokine and cytokine expression in type 2 diabetes compared with non-diabetic controls. The inflammatory response in type 2 diabetes was mimicked by exposure of non-diabetic human islets to palmitate, but not to oleate or high glucose, leading to the induction of IL-1beta, TNF-alpha, IL-6, IL-8, chemokine (C-X-C motif) ligand 1 (CXCL1) and chemokine (C-C motif) ligand 2 (CCL2). Interference with IL-1beta signalling abolished palmitate-induced cytokine and chemokine expression but failed to prevent lipotoxic human islet cell death. Palmitate activated nuclear factor kappaB (NF-kappaB) in human pancreatic beta and non-beta cells, and chemically induced endoplasmic reticulum stress caused cytokine expression and NF-kappaB activation similar to that occurring with palmitate. CONCLUSIONS/INTERPRETATION: Saturated-fatty-acid-induced NF-kappaB activation and endoplasmic reticulum stress may contribute to IL-1beta production and mild islet inflammation in type 2 diabetes. This inflammatory process does not contribute to lipotoxicity ex vivo, but may lead to local chemokine release.


Subject(s)
Chemokine CCL2/metabolism , Diabetes Mellitus, Type 2/metabolism , Insulin-Secreting Cells/metabolism , Islets of Langerhans/drug effects , Islets of Langerhans/metabolism , Palmitates/pharmacology , Aged , Cell Line , Chemokine CXCL1 , Enzyme-Linked Immunosorbent Assay , Female , Fluorescent Antibody Technique , Humans , In Vitro Techniques , Interleukin-6/metabolism , Interleukin-8/metabolism , Male , Middle Aged , NF-kappa B/metabolism , Oligonucleotide Array Sequence Analysis , Polymerase Chain Reaction , Radioimmunoassay , Tumor Necrosis Factor-alpha/metabolism
19.
Clin Exp Immunol ; 159(3): 338-43, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20059481

ABSTRACT

Type 1 diabetes results from a T cell-mediated destruction of insulin-producing pancreatic beta cells. Little is known on local factors contributing to migration of T cells to pancreatic tissue. We recently demonstrated evidence of viral infection in beta cells in several recent-onset type 1 diabetes patients. Islet inflammation was analysed in a series of new- or recent-onset type 1 diabetic patients and non-diabetic control subjects. Autoimmune T cell reactivity was studied in lymphocytes derived from pancreas-draining lymph nodes of one recent-onset type 1 diabetes patient in partial clinical remission. Insulitic lesions were characterized by presence of beta cells, elevated levels of the chemokine CXCL10 and infiltration of lymphocytes expressing the corresponding chemokine receptor CXCR3 in all pancreatic lesions of type 1 diabetes patients, regardless of enterovirus infection of beta cells. CXCR3 and CXCL10 were undetectable in pancreata of non-diabetic control subjects. T cells isolated from draining lymph nodes of a recent-onset patient with virally infected beta cells and in clinical remission reacted with multiple islet autoantigens and displayed a mixed interferon (IFN)-gamma/interleukin (IL)-10 cytokine pattern. Our data point to CXCL10 as an important cytokine in distressed islets that may contribute to inflammation leading to insulitis and beta cell destruction, regardless of local viral infection. We demonstrate further pro- and anti-inflammatory islet autoreactivity, indicating that different adaptive and innate immune responses may contribute to insulitis and beta cell destruction.


Subject(s)
Chemokine CXCL10/immunology , Diabetes Mellitus, Type 1/immunology , Insulin-Secreting Cells/immunology , Receptors, CXCR3/immunology , Adolescent , Adult , Child, Preschool , Diabetes Mellitus, Type 1/pathology , Diabetes Mellitus, Type 1/therapy , Diabetes Mellitus, Type 1/virology , Enterovirus/immunology , Enterovirus Infections/immunology , Enterovirus Infections/pathology , Enterovirus Infections/therapy , Female , Gene Expression Regulation/immunology , Humans , Immunity, Cellular , Inflammation/immunology , Inflammation/therapy , Inflammation/virology , Insulin-Secreting Cells/pathology , Insulin-Secreting Cells/virology , Interferon-gamma/immunology , Interleukin-10/immunology , Lymph Nodes/immunology , Lymph Nodes/pathology , Male , Retrospective Studies , T-Lymphocytes/immunology , T-Lymphocytes/pathology
20.
Diabetes Obes Metab ; 11(6): 589-95, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19515179

ABSTRACT

AIM: To compare the tolerability and efficacy of vildagliptin to pioglitazone as add-on therapy in patients with type 2 diabetes inadequately controlled with metformin monotherapy over 1-year duration. METHODS: This 52-week, multicentre, randomized, active-controlled study compared vildagliptin (50 mg b.i.d., n = 295) and pioglitazone (30 mg daily, n = 281) in patients with inadequate glycaemic control [haemoglobin A1c (HbA(1c)) 7.5-11%] receiving a stable dose of metformin (>or=1500 mg). The primary objective was to demonstrate non-inferiority of vildagliptin at 24 weeks in the change in HbA(1c) from baseline. The objective of the additional 28 weeks of the study was to assess long-term safety, while also assessing mean change from baseline to study end in HbA(1c), fasting plasma glucose and body weight. RESULTS: When added to a stable dose of metformin (mean baseline dose approximately 2 g/day), the non-inferiority of HbA(1c) lowering of vildagliptin to pioglitazone over 24 weeks was established at the non-inferiority margin of 0.3% (between-group difference = 0.1%). During the remaining 28 weeks, comparable HbA(1c) decreases were recorded in both groups. Overall adverse event (AE) rates were similar in both groups, as was the occurrence of peripheral oedema. Hypoglycaemia occurred rarely in both groups. Serious AEs occurred more frequently with pioglitazone group. While mean body weight increased significantly in the pioglitazone group (+2.6 kg) from baseline, there was no significant weight gain with vildagliptin (+0.2 kg). CONCLUSIONS: When added to metformin, vildagliptin demonstrates favourable safety and tolerability over 1 year. Vildagliptin provided additional HbA(1c) lowering to that achieved with metformin alone and comparable to that achieved with pioglitazone, with only pioglitazone causing weight gain.


Subject(s)
Adamantane/analogs & derivatives , Diabetes Mellitus, Type 2/drug therapy , Dipeptidyl-Peptidase IV Inhibitors/therapeutic use , Hypoglycemic Agents/therapeutic use , Metformin/therapeutic use , Nitriles/therapeutic use , Pyrrolidines/therapeutic use , Thiazolidinediones/therapeutic use , Adamantane/adverse effects , Adamantane/therapeutic use , Adolescent , Adult , Aged , Blood Glucose , Body Weight , Dipeptidyl-Peptidase IV Inhibitors/adverse effects , Drug Therapy, Combination , Edema/chemically induced , Fasting , Female , Glycated Hemoglobin/metabolism , Humans , Hypoglycemia/chemically induced , Hypoglycemic Agents/adverse effects , Male , Metformin/adverse effects , Middle Aged , Nitriles/adverse effects , Pioglitazone , Pyrrolidines/adverse effects , Thiazolidinediones/adverse effects , Treatment Outcome , Vildagliptin , Weight Gain/drug effects , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL