Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where S Del Guerra is active.

Publication


Featured researches published by S Del Guerra.


Diabetologia | 2008

Epigenetic regulation of PPARGC1A in human type 2 diabetic islets and effect on insulin secretion.

Charlotte Ling; S Del Guerra; R Lupi; Tina Rönn; Charlotte Granhall; Holger Luthman; Pellegrino Masiello; Piero Marchetti; Leif Groop; S. Del Prato

Aims/hypothesisInsulin secretion in pancreatic islets is dependent upon mitochondrial function and production of ATP. The transcriptional coactivator peroxisome proliferator activated receptor gamma coactivator-1 alpha (protein PGC-1α; gene PPARGC1A) is a master regulator of mitochondrial genes and its expression is decreased and related to impaired oxidative phosphorylation in muscle from patients with type 2 diabetes. Whether it plays a similar role in human pancreatic islets is not known. We therefore investigated if PPARGC1A expression is altered in islets from patients with type 2 diabetes and whether this expression is influenced by genetic (PPARGC1A Gly482Ser polymorphism) and epigenetic (DNA methylation) factors. We also tested if experimental downregulation of PPARGC1A expression in human islets influenced insulin secretion.MethodsThe PPARGC1A Gly482Ser polymorphism was genotyped in human pancreatic islets from 48 non-diabetic and 12 type 2 diabetic multi-organ donors and related to PPARGC1A mRNA expression. DNA methylation of the PPARGC1A promoter was analysed in pancreatic islets from ten type 2 diabetic and nine control donors. Isolated human islets were transfected with PPARGC1A silencing RNA (siRNA).ResultsPPARGC1A mRNA expression was reduced by 90% (p < 0.005) and correlated with the reduction in insulin secretion in islets from patients with type 2 diabetes. After downregulation of PPARGC1A expression in human islets by siRNA, insulin secretion was reduced by 41% (p ≤ 0. 01). We were able to ascribe reduced PPARGC1A expression in islets to both genetic and epigenetic factors, i.e. a common PPARGC1A Gly482Ser polymorphism was associated with reduced PPARGC1A mRNA expression (p < 0.00005) and reduced insulin secretion (p < 0.05). In support of an epigenetic influence, the PPARGC1A gene promoter showed a twofold increase in DNA methylation in diabetic islets compared with non-diabetic islets (p < 0.04).Conclusions/interpretationWe have shown for the first time that PPARGC1A might be important in human islet insulin secretion and that expression of PPARGC1A in human islets can be regulated by both genetic and epigenetic factors.


Diabetologia | 2009

Autophagy in human type 2 diabetes pancreatic beta cells

Matilde Masini; Marco Bugliani; R Lupi; S Del Guerra; Ugo Boggi; Franco Filipponi; Lorella Marselli; Pellegrino Masiello; Piero Marchetti

Aims/hypothesisBeta cell loss contributes to type 2 diabetes, with increased apoptosis representing an underlying mechanism. Autophagy, i.e. the physiological degradation of damaged organelles and proteins, may, if altered, be associated with a distinct form of cell death. We studied several features of autophagy in beta cells from type 2 diabetic patients and assessed the role of metabolic perturbation and pharmacological intervention.MethodsPancreatic samples were obtained from organ donors and isolated islets prepared both by collagenase digestion and density gradient centrifugation. Beta cell morphology and morphometry were studied by electron microscopy. Gene expression studies were performed by quantitative RT-PCR.ResultsUsing electron microscopy, we observed more dead beta cells in diabetic (2.24 ± 0.53%) than control (0.66 ± 0.52%) samples (p < 0.01). Massive vacuole overload (suggesting altered autophagy) was associated with 1.18 ± 0.54% dead beta cells in type 2 diabetic samples and with 0.36 ± 0.26% in control samples (p < 0.05). Density volume of autophagic vacuoles and autophagosomes was significantly higher in diabetic beta cells. Unchanged gene expression of beclin-1 and ATG1 (also known as ULK1), and reduced transcription of LAMP2 and cathepsin B and D was observed in type 2 diabetic islets. Exposure of non-diabetic islets to increased NEFA concentration led to a marked increase of vacuole accumulation, together with enhanced beta cell death, which was associated with decreased LAMP2 expression. Metformin ameliorated autophagy alterations in diabetic beta cells and beta cells exposed to NEFA, a process associated with normalisation of LAMP2 expression.Conclusions/interpretationBeta cells in human type 2 diabetes have signs of altered autophagy, which may contribute to loss of beta cell mass. To preserve beta cell mass in diabetic patients, it may be necessary to target multiple cell-death pathways.


Diabetes-metabolism Research and Reviews | 2007

Gliclazide protects human islet beta-cells from apoptosis induced by intermittent high glucose.

S Del Guerra; M Grupillo; Matilde Masini; R Lupi; Marco Bugliani; S Torri; Ugo Boggi; M Del Chiaro; Fabio Vistoli; Franco Mosca; S. Del Prato; Piero Marchetti

Decreased beta‐cell mass, mainly due to apoptosis, is crucial for the development and progression of type 2 diabetes. Chronic exposure to high glucose levels is a probable underlying mechanism, whereas the role of oral anti‐diabetic agents (sulphonylureas in particular) is still unsettled.


Nutrition Metabolism and Cardiovascular Diseases | 2010

G-protein-coupled receptor 40 (GPR40) expression and its regulation in human pancreatic islets: The role of type 2 diabetes and fatty acids

S Del Guerra; Marco Bugliani; V. D'Aleo; S. Del Prato; Ugo Boggi; Franco Mosca; Franco Filipponi; R Lupi

BACKGROUND AND AIMS GPR40 is a membrane-bound receptor paired with medium and long-chain fatty acids (FFA) as endogenous ligands. Its acute activation potentiates insulin secretion from beta cells, whereas prolonged binding might contribute to the deleterious effects of chronic exposure to FFA. Little information is available on the expression of GPR40 and its regulation in human islets (HI). MATERIAL AND METHODS HI were prepared by enzymatic digestion and gradient separation from the pancreas of 20 non-diabetic (Ctrl) and 13 type 2 diabetic (T2DM) multiorgan donors, and functional and molecular studies were then performed. RESULTS By qualitative and quantitative PCR experiments, mRNA expression was shown in HI. Both in T2DM islets and in Ctrl islets pre-exposed for 24 h to 1.0 mmol/l FFA (palmitate:oleate, 2:1), GPR40 mRNA expression was significantly reduced (p<0.01) in the T2DM cells as compared to Ctrl cells. A significant positive correlation was found between glucose-stimulated insulin secretion and GPR40 expression. CONCLUSIONS These results show the expression of GPR40 in human pancreatic islets which are regulated by FFA. The finding that T2DM islets have a lower GPR40 expression, and the correlation of these genes with insulin secretion, raises the possibility of an involvement of GPR40 in human diabetes beta-cell dysfunction.


Diabetes, Obesity and Metabolism | 2008

Effects of exendin-4 on islets from type 2 diabetes patients

R Lupi; R Mancarella; S Del Guerra; Marco Bugliani; S. Del Prato; Ugo Boggi; Franco Mosca; Franco Filipponi; Piero Marchetti

Exendin‐4 is a dipeptidyl peptidase IV (DPP‐IV)‐resistant glucagon‐like peptide1 (GLP‐1) mimetic and its synthetic counterpart, exenatide, is being used in the therapy of type 2 diabetes (T2DM). No information, however, is currently available as for the direct action of exendin‐4 on human T2DM islets. In the present study, we exposed pancreatic islets prepared from non‐diabetic and T2DM subjects to exendin‐4 for 48 h and found that the compound had several, direct beneficial actions on insulin secretion and the expression of genes involved in beta‐cell function and differentiation.


Regulatory Peptides | 2010

The direct effects of GLP-1 and GIP, alone or in combination, on human pancreatic islets

R Lupi; S Del Guerra; V. D'Aleo; Ugo Boggi; Franco Filipponi; Piero Marchetti

GLP-1 and GIP are incretins known to affect beta-cell function and turnover. However, information on the direct actions of these hormones on human islet cells is limited. We tested the effects of acute (45min) or prolonged (2days) exposure to GLP-1 or GIP, alone or in combination, on the function and some molecular features of human islets isolated from non-diabetic and type 2 diabetic multiorgan donors. Acutely, both GLP-1 and, more markedly so, GIP, significantly potentiated glucose-stimulated insulin release, with no apparent synergic action. Some of these effects were observed with type 2 diabetic islets as well. Following prolonged exposure to the incretins, improved insulin secretion was observed, and transcription of insulin, PDX-1 and Bcl-2 was increased in both non-diabetic and diabetic islets, with the combination of GLP-1 and GIP showing more significant effects. Although it is still unclear at what extent these beta-cell direct actions of individual or combined incretins occur in-vivo in humans, nevertheless the results of the present study suggest that enhancing the exposure of pancreatic islets to circulating levels of both incretins may be useful for therapeutical purposes.


Acta Diabetologica | 1997

Effects of glibenclamide and metformin (alone or in combination) on insulin release from isolated human pancreatic islets

R Lupi; Piero Marchetti; R Giannarelli; A Coppelli; C. Tellini; S Del Guerra; M Lorenzetti; M. Carmellini; Franco Mosca; R. Navalesi

Abstract Isolated human pancreatic islets were prepared by collagenase digestion and density gradient purification, and the effects of glibenclamide (0.5 and 5.0 µmol/l) and metformin (20 and 200 µmol/l), alone or in combination, on insulin release were evaluated at varying glucose concentrations. At 3.3 mmol/l glucose level, the addition of 5.0 µmol/l glibenclamide or 5.0 µmol/l glibenclamide plus 200 µmol/l metformin caused a significant increase of insulin release, compared with glucose alone. At 16.7 mmol/l glucose concentration, a significant increase of insulin secretion, compared with glucose alone, was produced by the addition of either 5.0 µmol/l glibenclamide, 200 µmol/l metformin, or both 5.0 µmol/l glibenclamide and 200 µmol/l metformin. The effect of the combination of the two drugs was significantly higher than that with either drug used alone. Thus, glibenclamide was shown to have an insulinotropic effect on human islets at both low and high glucose concentrations, and metformin at high glucose concentrations. A possible synergistic effect of glibenclamide and metformin at high glucose concentrations is also suggested.


Diabetes & Metabolism | 2009

Effects of exposure of human islet beta-cells to normal and high glucose levels with or without gliclazide or glibenclamide.

S Del Guerra; V. D’Aleo; R Lupi; Matilde Masini; Marco Bugliani; Ugo Boggi; Franco Filipponi; Piero Marchetti

AIM To evaluate the effects of exposure to high glucose (HG) levels and sulphonylurea on isolated human islet-cell function, and to investigate some of the mechanisms that might be involved. METHODS Islet cells were isolated, using collagenase digestion and gradient purification, from 13 pancreata from non-diabetic multiorgan donors (age: 61.2+/-11.5 years; gender: 7 men/6 women; body mass index: 25.1+/-2.8kg/m(2)). The cells were then cultured for 5 days with normal glucose (NG) concentrations (5.5mmol/L), or NG and HG (16.7mmol/L) levels (alternating every 24h), with or without the addition of therapeutic concentrations of gliclazide (10micromol/L) or glibenclamide (1.0micromol/L). At the end of incubation, functional (glucose-stimulated insulin secretion), morphological (electron microscopy) and molecular (gene and protein expression) studies were performed. RESULTS Insulin secretion differed significantly between study groups, with marked decreases in the presence of HG plus glibenclamide. Compared with NG, insulin expression decreased significantly with HG, and increased similarly with gliclazide as with glibenclamide. However, exposure to gliclazide, but not glibenclamide, significantly induced expression (at both gene and protein levels) of PDX-1, a fundamental beta-cell differentiation transcription factor, and Ki67, a marker of proliferation. However, gliclazide and glibenclamide did not differ in terms of effects on gene expression of the antiapoptotic molecule Bcl2 (increased significantly with both) and the proapoptotic molecule Bax (decreased significantly with both). CONCLUSION Gliclazide and glibenclamide have different effects on the changes induced by prolonged exposure of human islet cells to high levels of glucose.


Transplantation Proceedings | 2010

Functional and Survival Analysis of Isolated Human Islets

V. D'Aleo; S Del Guerra; G. Gualtierotti; Franco Filipponi; Ugo Boggi; P De Simone; Fabio Vistoli; S. Del Prato; Piero Marchetti; R Lupi

BACKGROUND AND AIMS Pancreatic islet transplantation has become one of the potential treatments for type 1 diabetes. We evaluated functional and viability parameters of isolated islets in relation to donors clinical characteristics and preparation variables. METHODS Islets were isolated from 70 nondiabetic multiorgan donors of overall age of 62.5 +/- 15.9 years. There were 41 men and 29 women. Their mean body mass index (BMI) was 25.62 +/- 3.09 kg/m(2). We evaluated the islet number (IEQ/g pancreatic tissue) insulin release (IR; microU/islet/min) in response to 3.3 (g) or 16.7 (G) mmol/L glucose; calcium flux concentration (CFC); and islet cell viability. RESULTS IEQ was 5249 +/- 1505, with 73.7 +/- 14.96% viable islet cells. IR was 0.03 +/- 0.01 at g and 0.11 +/- 0.06 at G (stimulation index [S] = 3.24 +/- 1.96). CFC was 1.95 +/- 1.03 DeltaRFU. We observed positive correlations between viable cells and IR at g (R(2) = 0.260; P = .013), IR at G (R(2) = 0.165; P = .013), and CFC (R(2) = 0.175; P = .047). A positive correlation was documented between BMI and g (R(2) = 0.245; P = .016) and negative correlations between age with SI (R(2) = 0.188; P = .052) and cold ischemia time with IEQ (R(2) = 0.865; P = .0061). CONCLUSIONS These results showed that quality control of isolated human pancreatic islets allowed assessment of beta-cell function and survival before transplantation, revealing several important variables.


Transplantation Proceedings | 2010

A Common Polymorphism in the Monocyte Chemoattractant Protein-1 (MCP-1) Gene Regulatory Region Influences MCP-1 Expression and Function of Isolated Human Pancreatic Islets

S Del Guerra; V. D'Aleo; G. Gualtierotti; Franco Filipponi; Ugo Boggi; P De Simone; Fabio Vistoli; S. Del Prato; Piero Marchetti; R Lupi

BACKGROUND AND AIMS Islet transplantation is an attractive approach to treat type 1 diabetic patients. However, suboptimal islet engraftment still represents an unsolved problem. It has been shown that human islets release monocyte chemoattractant protein-1 (MCP-1), one of the most powerful macrophage chemokines, which may impair the fate of the transplant. The aim of this study was to evaluate the presence and role of MCP-1 in isolated human islets, including genotyping for a common polymorphism. METHODS Pancreatic islets were isolated by enzymatic digestion and gradient purification from 41 nondiabetic multiorgan donors. We measured MCP-1 mRNA expression by quantitative real- time reverse-transcriptase polymerization chain reaction, analyzed the MCP-1 single nucleotide polymorphism, -2518 G/A (SNP, rs 1024611) and evaluated glucose-stimulated insulin release (IR; microU/islet/min). RESULTS MCP-1 mRNA expression was found in all studied batches of islets. Overall, IR was significantly higher at 16.7 mmol/L than 3.3 mmol/L glucose. We observed a significant negative correlation between MCP-1 mRNA expression and stimulation index (SI). We found that MCP-1 mRNA expression was significantly higher in CC and CT compared with TT genotype groups. Finally, SI was significant lower in the CC with respect to the TT genotype group. CONCLUSIONS These data show that MCP-1 gene expression regulated by the -2518 G/A polymorphism, is correlated with glucose-stimulated insulin release. The study of MCP-1 expression and genotype on isolated islets before transplantation may be useful to understand the inflammatory response after infusion of human islets into patients with type 1 diabetes mellitus.

Collaboration


Dive into the S Del Guerra's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge