Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 45
Filter
Add more filters










Publication year range
1.
Orphanet J Rare Dis ; 16(1): 410, 2021 10 02.
Article in English | MEDLINE | ID: mdl-34600590

ABSTRACT

BACKGROUND: Abnormalities of the immune system are rarely reported in patients affected by RASopathies. Aim of the current study was to investigate the prevalence of immune system dysfunction in a cohort of patients affected by RASopathies. STUDY DESIGN: A group of 69 patients was enrolled: 60 at the Federico II University, Naples, 7 at University Magna Graecia of Catanzaro, 2 at "Scuola Medica Salernitana", Salerno. An age- and sex-matched control group was also enrolled. Autoimmune disorders were investigated according to international consensus criteria. Immune framework was also evaluated by immunoglobulin levels, CD3, CD4, CD8, CD19, CD56 lymphocyte subpopulations, autoantibodies levels and panel of inflammatory molecules, in both patients and controls. RESULTS: Frequent upper respiratory tract infections were recorded in 2 patients; pneumonia, psoriasis and alopecia in single patients. Low IgA levels were detected in 8/44 patients (18.18%), low CD8 T cells in 13/35 patients (37.14%). Anti-tg and anti-TPO antibodies were detected in 3/24 patients (12.5%), anti r-TSH in 2 cases (8.33%), all in euthyroidism. Serum IgA and CD8 levels were significantly lower in patients than in controls (p 0.00685; p 0.000656 respectively). All tested patients showed increased inflammatory molecules compared to controls. These findings may anticipate the detection of overt autoimmune disease. CONCLUSIONS: Patients affected by RASopathies are at risk to develop autoimmune disorders. Routine screening for autoimmunity is recommended in patients with RASopathy.


Subject(s)
Autoimmune Diseases , Immunity, Cellular , Antigens, CD19 , Autoimmunity , Humans
2.
Platelets ; 29(5): 463-467, 2018 Jul.
Article in English | MEDLINE | ID: mdl-28635382

ABSTRACT

Platelet-derived factors are biomaterials that might accelerate healing process in oral, maxillofacial, and several other applications. Release of specific factors by platelet concentrates is critical to achieving a successful outcome. Here, we have shown that platelet-rich fibrin (PRF) clots were beneficial sources of leukocytes, which may directly affect the release of chemokines and growth factors. When compared with the standard leukocyte-PRF (L-PRF), the experimental low-force modified procedure [defined as advanced-PRF (A-PRF)] entrapped the same content of viable leukocytes, released a similar amount of inflammatory cytokines, but secreted 3-, 1.6-, 3-, and 1.2-fold higher levels of Eotaxin, CCL5, platelet-derived growth factor (PDGF) and vascular endothelial growth factor (VEGF), respectively. A leukocyte-free scaffold, such as plasma rich in growth factors (PRGF), released only platelet-specific factors and, in particular, the F3 fraction, the richest in growth factors, secreted higher amount of CCL5 and PDGF compared to F1 and F2 fractions. In conclusion, different procedures and leukocyte content affect cytokine, chemokines, and growth factor release from platelet derivatives, which may be helpful in different clinical settings.


Subject(s)
Blood Platelets/metabolism , Chemokines/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Leukocytes/metabolism , Adult , Female , Humans , Male , Platelet-Rich Plasma/metabolism
3.
Int J Obes (Lond) ; 40(6): 929-37, 2016 06.
Article in English | MEDLINE | ID: mdl-26980478

ABSTRACT

BACKGROUND/OBJECTIVES: The genomic bases of the adipose tissue abnormalities induced by chronic positive calorie excess have been only partially elucidated. We adopted a genome-wide approach to directly test whether long-term high-fat diet (HFD) exposure affects the DNA methylation profile of the mouse adipose tissue and to identify the functional consequences of these changes. SUBJECTS/METHODS: We have used epididymal fat of mice fed either high-fat (HFD) or regular chow (STD) diet for 5 months and performed genome-wide DNA methylation analyses by methylated DNA immunoprecipitation sequencing (MeDIP-seq). Mouse Homeobox (Hox) Gene DNA Methylation PCR, RT-qPCR and bisulphite sequencing analyses were then performed. RESULTS: Mice fed the HFD progressively expanded their adipose mass accompanied by a significant decrease in glucose tolerance (P<0.001) and insulin sensitivity (P<0.05). MeDIP-seq data analysis revealed a uniform distribution of differentially methylated regions (DMR) through the entire adipocyte genome, with a higher number of hypermethylated regions in HFD mice (P<0.005). This different methylation profile was accompanied by increased expression of the Dnmt3a DNA methyltransferase (Dnmt; P<0.05) and the methyl-CpG-binding domain protein Mbd3 (P<0.05) genes in HFD mice. Gene ontology analysis revealed that, in the HFD-treated mice, the Hox family of development genes was highly enriched in differentially methylated genes (P=0.008). To validate this finding, Hoxa5, which is implicated in fat tissue differentiation and remodeling, has been selected and analyzed by bisulphite sequencing, confirming hypermethylation in the adipose tissue from the HFD mice. Hoxa5 hypermethylation was associated with downregulation of Hoxa5 mRNA and protein expression. Feeding animals previously exposed to the HFD with a standard chow diet for two further months improved the metabolic phenotype of the animals, accompanied by return of Hoxa5 methylation and expression levels (P<0.05) to values similar to those of the control mice maintained under standard chow. CONCLUSIONS: HFD induces adipose tissue abnormalities accompanied by epigenetic changes at the Hoxa5 adipose tissue remodeling gene.


Subject(s)
Adipose Tissue/metabolism , DNA Methylation , Diet, High-Fat , Down-Regulation , Homeodomain Proteins/genetics , Phosphoproteins/genetics , Transcription, Genetic , Animals , Disease Models, Animal , Epigenesis, Genetic , Immunohistochemistry , Male , Mice , Mice, Inbred C57BL , Obesity/genetics , Transcription Factors
4.
J Endocrinol Invest ; 39(3): 259-63, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26105974

ABSTRACT

In the last decades, many reports have focused the attention on deleterious effects of novel environmental chemical compounds, including bisphenol A (BPA), on human health. BPA, a common and widely chemical contaminant acting as endocrine disruptor, accumulates in adipose tissue and may affect adipocyte metabolic and inflammatory functions. BPA, at low chronic doses, is now considered as an obesogen compound, and might contribute to the rise of metabolic syndrome, visceral adiposity and diabetes epidemics. The BPA worldwide presence in the environment is responsible for chronic exposure during vulnerable periods, such as foetal and neonatal life. The BPA source of contamination can occur via food, beverage, wastewater, air, dust and soil. BPA, as lipophilic compound, may accumulate into the adipose tissue already during foetal life and may affect adulthood health, through adverse effects on the growth and development of organs and tissues. Thus, based on several studies, it would be crucial to consider further actions aimed to refine risk assessment at least in vulnerable population, such as foetuses, infants and young children, to prevent metabolic diseases and obesity.


Subject(s)
Air Pollutants, Occupational/adverse effects , Benzhydryl Compounds/adverse effects , Environmental Exposure/adverse effects , Metabolic Syndrome/etiology , Phenols/adverse effects , Humans , Metabolic Syndrome/epidemiology , Risk Assessment , Vulnerable Populations
5.
Carbohydr Polym ; 131: 407-14, 2015 Oct 20.
Article in English | MEDLINE | ID: mdl-26256201

ABSTRACT

In this paper we propose polysaccharide hydrogels combining alginate (ALG) and hyaluronan (HA) as biofunctional platform for dermal wound repair. Hydrogels produced by internal gelation were homogeneous and easy to handle. Rheological evaluation of gelation kinetics of ALG/HA mixtures at different ratios allowed understanding the HA effect on ALG cross-linking process. Disk-shaped hydrogels, at different ALG/HA ratio, were characterized for morphology, homogeneity and mechanical properties. Results suggest that, although the presence of HA does significantly slow down gelation kinetics, the concentration of cross-links reached at the end of gelation is scarcely affected. The in vitro activity of ALG/HA dressings was tested on adipose derived multipotent adult stem cells (Ad-MSC) and an immortalized keratinocyte cell line (HaCaT). Hydrogels did not interfere with cell viability in both cells lines, but significantly promoted gap closure in a scratch assay at early (1 day) and late (5 days) stages as compared to hydrogels made of ALG alone (p<0.01 and 0.001 for Ad-MSC and HaCaT, respectively). In vivo wound healing studies, conducted on a rat model of excised wound indicated that after 5 days ALG/HA hydrogels significantly promoted wound closure as compared to ALG ones (p<0.001). Overall results demonstrate that the integration of HA in a physically cross-linked ALG hydrogel can be a versatile strategy to promote wound healing that can be easily translated in a clinical setting.


Subject(s)
Alginates/pharmacology , Hyaluronic Acid/pharmacology , Hydrogels/pharmacology , Wound Healing/drug effects , Animals , Cell Line , Cell Movement/drug effects , Cell Survival/drug effects , Cross-Linking Reagents/pharmacology , Disease Models, Animal , Glucuronic Acid/pharmacology , Hexuronic Acids/pharmacology , Humans , Male , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/drug effects , Rats, Wistar , Rheology/drug effects
6.
PPAR Res ; 2014: 537865, 2014.
Article in English | MEDLINE | ID: mdl-24790595

ABSTRACT

The nuclear receptor PPAR γ is a key regulator of adipogenesis, and alterations of its function are associated with different pathological processes related to metabolic syndrome. We recently identified two PPARG transcripts encoding dominant negative PPAR γ isoforms. The existence of different PPARG variants suggests that alternative splicing is crucial to modulate PPAR γ function, underlying some underestimated aspects of its regulation. Here we investigate PPARG expression in different tissues and cells affected in metabolic syndrome and, in particular, during adipocyte differentiation of human mesenchymal stem cells. We defined the transcript-specific expression pattern of PPARG variants encoding both canonical and dominant negative isoforms and identified a novel PPARG transcript, γ 1ORF4. Our analysis indicated that, during adipogenesis, the transcription of alternative PPARG variants is regulated in a time-specific manner through differential usage of distinct promoters. In addition, our analysis describes-for the first time-the differential contribution of three ORF4 variants to this process, suggesting a still unexplored role for these dominant negative isoforms during adipogenesis. Therefore, our results highlight crucial aspects of PPARG regulation, suggesting the need of further investigation to rule out the differential impact of all PPARG transcripts in both physiologic and pathologic conditions, such as metabolism-related disorders.

7.
Platelets ; 25(4): 252-6, 2014.
Article in English | MEDLINE | ID: mdl-23855408

ABSTRACT

Abstract Platelet derivatives are commonly used in wound healing and tissue regeneration. Different procedures of platelet preparation may differentially affect growth factor release and cell growth. Preparation of platelet-rich fibrin (PRF) is accompanied by release of growth factors, including platelet-derived growth factor (PDGF), vascular endothelial growth factor (VEGF) and transforming growth factor ß1 (TGFß1), and several cytokines. When compared with the standard procedure for platelet-rich plasma (PRP), PRF released 2-fold less PDGF, but >15-fold and >2-fold VEGF and TGFß1, respectively. Also, the release of several cytokines (IL-4, IL-6, IL-8, IL-10, IFNγ, MIP-1α, MIP-1ß and TNFα) was significantly increased in PRF-conditioned medium (CM), compared to PRP-CM. Incubation of both human skin fibroblasts and human umbilical vein endothelial cells (HUVECs) with PRF-derived membrane (mPRF) or with PRF-CM enhanced cell proliferation by >2-fold (p<0.05). Interestingly, PRP elicited fibroblast growth at a higher extent compared to PRF. At variance, PRF effect on HUVEC growth was significantly greater than that of PRP, consistent with a higher concentration of VEGF in the PRF-CM. Thus, the procedure of PRP preparation leads to a larger release of PDGF, as a possible result of platelet degranulation, while PRF enhances the release of proangiogenic factors.


Subject(s)
Blood Platelets/physiology , Intercellular Signaling Peptides and Proteins/metabolism , Platelet-Rich Plasma , Adult , Cell Proliferation/drug effects , Cytokines/metabolism , Female , Fibroblasts/drug effects , Fibroblasts/metabolism , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Intercellular Signaling Peptides and Proteins/pharmacology , Male , Young Adult
8.
Vet Parasitol ; 193(1-3): 252-5, 2013 Mar 31.
Article in English | MEDLINE | ID: mdl-23312871

ABSTRACT

Protozoal merozoites were identified in the cerebrospinal fluid of two sheep with neurological disease in the UK. Polymerase chain reaction (PCR) identified the merozoites as Sarcocystis capracanis, a common protozoal pathogen of goats. This is the first report of this species infecting sheep and may represent an aberrant infection with sheep acting as dead end hosts, or alternatively could indicate that sheep are able to act as intermediate hosts for S. capracanis, widening the previously reported host range of this pathogen. It is possible that S. capracanis is a previously unrecognised cause of ovine protozoal meningoencephalitis (OPM) in the UK.


Subject(s)
Sarcocystis/isolation & purification , Sarcocystosis/veterinary , Sheep Diseases/parasitology , Animals , Female , Male , Sarcocystosis/cerebrospinal fluid , Sarcocystosis/parasitology , Sheep , Sheep Diseases/cerebrospinal fluid , Sheep Diseases/epidemiology
9.
Diabetologia ; 55(10): 2811-2822, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22798065

ABSTRACT

AIMS/HYPOTHESIS: Type 2 diabetes and obesity are associated with increased risk of site-specific cancers. We have investigated whether metabolic alterations at the level of adipose-derived differentiating cells may affect specific phenotypes of breast cancer cells. METHODS: Growth profiles of breast cancer cell lines were evaluated in co-cultures with differentiated adipocytes or their precursor cells and upon treatment with adipocyte conditioned media. Production and release of cytokines and growth factors were assessed by real-time RT-PCR and multiplex-based ELISA assays. RESULTS: Co-cultures with either differentiated mouse 3T3-L1 or human mammary adipocytes increased viability of MCF-7 cells to a greater extent, when compared with their undifferentiated precursors. Adipocytes cultured in 25 mmol/l glucose were twofold more effective in promoting cell growth, compared with those grown in 5.5 mmol/l glucose, and activated mitogenic pathways in MCF-7 cells. Growth-promoting action was also enhanced when adipocytes were incubated in the presence of palmitate or oleate. Interestingly, 3T3-L1 and human adipocytes released higher amounts of keratinocyte-derived chemokine/IL-8, the protein 'regulated upon activation, normally T expressed, and secreted' (RANTES), and IGF-1, compared with their precursor cells. Their levels were reduced upon incubation with low glucose and enhanced by fatty acids. Moreover, both undifferentiated cells and differentiated adipocytes from obese individuals displayed about twofold higher IGF-1 release and MCF-7 cell growth induction than lean individuals. Finally, inhibition of the IGF-1 pathway almost completely prevented the growth-promoting effect of adipocytes on breast cancer cells. CONCLUSIONS/INTERPRETATION: IGF-1 release by adipocytes is regulated by glucose and fatty acids and may contribute to the control of cancer cell growth in obese individuals.


Subject(s)
Adipocytes/metabolism , Breast Neoplasms/pathology , Cell Proliferation/drug effects , Glucose/pharmacology , Insulin-Like Growth Factor I/metabolism , Oleic Acid/pharmacology , Palmitates/pharmacology , Adenocarcinoma/pathology , Adipocytes/drug effects , Adipocytes/pathology , Adult , Aged , Cell Communication/physiology , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/physiology , Chemokine CCL5/metabolism , Coculture Techniques , Female , Humans , Interleukin-8/metabolism , MCF-7 Cells , Middle Aged , Obesity/metabolism , Obesity/pathology , Signal Transduction/physiology
10.
Cell Death Differ ; 19(7): 1127-38, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22281705

ABSTRACT

TGF-beta1 has been shown to induce autophagy in certain cells but whether and how this action is exerted in muscle and whether this activity relates to TGF-beta1 control of muscle cell differentiation remains unknown. Here, we show that expression of the autophagy-promoting protein phosphoprotein enriched in diabetes/phosphoprotein enriched in astrocytes (PED/PEA-15) progressively declines during L6 and C2C12 skeletal muscle cell differentiation. PED/PEA-15 underwent rapid induction upon TGF-beta1 exposure of L6 and C2C12 myoblasts, accompanied by impaired differentiation into mature myotubes. TGF-beta1 also induced autophagy in the L6 and C2C12 cells through a PP2A/FoxO1-mediated mechanism. Both the TGF-beta1 effect on differentiation and that on autophagy were blocked by specific PED/PEA-15 ShRNAs. Myoblasts stably overexpressing PED/PEA-15 did not differentiate and showed markedly enhanced autophagy. In these same cells, the autophagy inhibitor 3-methyladenine rescued TGF-beta1 effect on both autophagy and myogenesis, indicating that PED/PEA-15 mediates TGF-beta1 effects in muscle. Muscles from transgenic mice overexpressing PED/PEA-15 featured a significant number of atrophic fibers, accompanied by increased light chain 3 (LC3)II to LC3I ratio and reduced PP2A/FoxO1 phosphorylation. Interestingly, these mice showed significantly impaired locomotor activity compared with their non-transgenic littermates. TGF-beta1 causes transcriptional upregulation of the autophagy-promoting gene PED/PEA-15, which in turn is capable to induce atrophic responses in skeletal muscle in vivo.


Subject(s)
Autophagy/drug effects , Muscle, Skeletal/cytology , Phosphoproteins/metabolism , Transforming Growth Factor beta1/pharmacology , Adenine/analogs & derivatives , Adenine/pharmacology , Animals , Apoptosis Regulatory Proteins , Astrocytes/cytology , Astrocytes/metabolism , Carboxylic Ester Hydrolases/metabolism , Cell Line , Forkhead Box Protein O1 , Forkhead Transcription Factors/metabolism , Mice , Mice, Transgenic , Microtubule-Associated Proteins/metabolism , Muscle Development , Muscle, Skeletal/metabolism , Myoblasts/cytology , Myoblasts/metabolism , Phosphoproteins/antagonists & inhibitors , Phosphoproteins/genetics , Phosphorylation , RNA Interference , RNA, Small Interfering/metabolism
11.
J Endocrinol Invest ; 33(7): 446-50, 2010.
Article in English | MEDLINE | ID: mdl-20671408

ABSTRACT

BACKGROUND: The cellular abundance of the phosphoprotein enriched in diabetes (PED/PEA-15), a 15 kDa protein related to insulin resistance (IR), is increased in women with polycystic ovary syndrome (PCOS). AIM: To investigate whether metformin (MET) has additive effects on PED/PEA-15 protein levels. MATERIAL/SUBJECTS AND METHODS: This is an open label, prospective clinical study over 6 months. Ten hyperandrogenic obese PCOS women [age: 24.6+/-1.6 yr; body mass index (BMI): 30.7+/-1.2 kg/m(2)] were treated with MET (1250 mg/day). Ten age- and BMI-matched normo-androgenic women were used as controls. Outcome measures are: PED/PEA-15 protein levels, fasting plasma glucose and insulin (FPI), reciprocal index of homeostasis model assessment of insulin resistance (1/HOMA-IR); quantitative insulin sensitivity check index (QUICKI); wholebody insulin sensitivity index (ISI); SHBG; total testosterone; free androgen index (FAI). RESULTS: At baseline FPI and PED/PEA- 15 protein levels were higher, while 1/HOMA-IR, QUICKI, and ISI were lower (p<0.001) in MET group than in controls. After treatment, independently of body weight and hyperandrogenism, FPI, and PED/PEA-15 protein levels decreased (p=0.001 and 0.004, respectively), while, 1/HOMA-IR, QUICKI, and ISI increased (p<0.001). PED/PEA-15 protein levels correlated significantly with ISI either before (r=0.636; p=0.048), and after treatment (r=0.758; p=0.011). CONCLUSIONS: PED/PEA-15 protein levels reduced after a short course of treatment with MET in a group hyperandrogenic obese PCOS women. This effect was independent of body weight and hyperandrogenism, and correlated with ISI, thus adding a further benefit to obese PCOS women.


Subject(s)
Insulin Resistance/physiology , Intracellular Signaling Peptides and Proteins/metabolism , Metformin/therapeutic use , Phosphoproteins/metabolism , Polycystic Ovary Syndrome/drug therapy , Adult , Androgens/blood , Apoptosis Regulatory Proteins , Blood Glucose/metabolism , Female , Humans , Insulin/blood , Obesity/blood , Polycystic Ovary Syndrome/physiopathology , Sex Hormone-Binding Globulin/metabolism
12.
Diabetologia ; 53(7): 1482-92, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20396999

ABSTRACT

AIMS/HYPOTHESIS: Overexpression of PED (also known as PEA15) determines insulin resistance and impaired insulin secretion and may contribute to progression toward type 2 diabetes. Recently, we found that the transcription factor hepatocyte nuclear factor (HNF)-4alpha binds to PED promoter and represses its transcription. However, the molecular details responsible for regulation of PED gene remain unclear. METHODS: Here we used gain and loss of function approaches to investigate the hypothesis that HNF-4alpha controls chromatin remodelling at the PED promoter in human cell lines. RESULTS: HNF-4alpha production and binding induce chromatin remodelling at the -250 to 50 region of PED, indicating that remodelling is limited to two nucleosomes located at the proximal promoter. Chromatin immunoprecipitation assays also revealed concomitant HNF-4alpha-induced deacetylation of histone H3 at Lys9 and Lys14, and increased dimethylation of histone H3 at Lys9. The latter was followed by reduction of histone H3 Lys4 dimethylation. HNF-4alpha was also shown to target the histone deacetylase complex associated with silencing mediator of retinoic acid and thyroid hormone receptor, both at the PED promoter, and at GRB14 and USP21 regulatory regions, leading to a reduction of mRNA levels. Moreover, HNF-4alpha silencing and PED overexpression were accompanied by a significant reduction of hepatic glycogen content. CONCLUSIONS/INTERPRETATION: These results show that HNF-4alpha serves as a scaffold protein for histone deacetylase activities, thereby inhibiting liver expression of genes including PED. Dysregulation of these mechanisms may lead to upregulation of the PED gene in type 2 diabetes.


Subject(s)
Epigenesis, Genetic/physiology , Hepatocyte Nuclear Factor 4/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Phosphoproteins/metabolism , Acetylation , Animals , Apoptosis Regulatory Proteins , Blotting, Western , Chromatin Assembly and Disassembly/genetics , Chromatin Assembly and Disassembly/physiology , Chromatin Immunoprecipitation , Epigenesis, Genetic/genetics , Hep G2 Cells , Hepatocyte Nuclear Factor 4/genetics , Histones/metabolism , Humans , Intracellular Signaling Peptides and Proteins/genetics , Mice , Mice, Transgenic , Nucleosomes/genetics , Phosphoproteins/genetics , Promoter Regions, Genetic/genetics , Reverse Transcriptase Polymerase Chain Reaction
13.
Diabetologia ; 53(5): 955-65, 2010 May.
Article in English | MEDLINE | ID: mdl-20165829

ABSTRACT

AIMS/HYPOTHESIS: Glucosamine, generated during hyperglycaemia, causes insulin resistance in different cells. Here we sought to evaluate the possible role of endoplasmic reticulum (ER) stress in the induction of insulin resistance by glucosamine in skeletal muscle cells. METHODS: Real-time RT-PCR analysis, 2-deoxy-D: -glucose (2-DG) uptake and western blot analysis were carried out in rat and human muscle cell lines. RESULTS: In both rat and human myotubes, glucosamine treatment caused a significant increase in the expression of the ER stress markers immunoglobulin heavy chain-binding protein/glucose-regulated protein 78 kDa (BIP/GRP78 [also known as HSPA5]), X-box binding protein-1 (XBP1) and activating transcription factor 6 (ATF6). In addition, glucosamine impaired insulin-stimulated 2-DG uptake in both rat and human myotubes. Interestingly, pretreatment of both rat and human myotubes with the chemical chaperones 4-phenylbutyric acid (PBA) or tauroursodeoxycholic acid (TUDCA), completely prevented the effect of glucosamine on both ER stress induction and insulin-induced glucose uptake. In both rat and human myotubes, glucosamine treatment reduced mRNA and protein levels of the gene encoding GLUT4 and mRNA levels of the main regulators of the gene encoding GLUT4 (myocyte enhancer factor 2 a [MEF2A] and peroxisome proliferator-activated receptor-gamma coactivator 1alpha [PGC1alpha]). Again, PBA or TUDCA pretreatment prevented glucosamine-induced inhibition of GLUT4 (also known as SLC2A4), MEF2A and PGC1alpha (also known as PPARGC1A). Finally, we showed that overproduction of ATF6 is sufficient to inhibit the expression of genes GLUT4, MEF2A and PGC1alpha and that ATF6 silencing with a specific small interfering RNA is sufficient to completely prevent glucosamine-induced inhibition of GLUT4, MEF2A and PGC1alpha in skeletal muscle cells. CONCLUSIONS/INTERPRETATION: In this work we show that glucosamine-induced ER stress causes insulin resistance in both human and rat myotubes and impairs GLUT4 production and insulin-induced glucose uptake via an ATF6-dependent decrease of the GLUT4 regulators MEF2A and PGC1alpha.


Subject(s)
Activating Transcription Factor 6/metabolism , Endoplasmic Reticulum/metabolism , Glucosamine/metabolism , Glucose Transporter Type 4/metabolism , Muscle Fibers, Skeletal/metabolism , Activating Transcription Factor 6/genetics , Analysis of Variance , Animals , Blotting, Western , Cell Line , Cells, Cultured , Chromatin Immunoprecipitation , Dose-Response Relationship, Drug , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum Chaperone BiP , Glucosamine/pharmacology , Glucose/metabolism , Glucose/pharmacology , Glucose Transporter Type 4/genetics , Heat-Shock Proteins/genetics , Heat-Shock Proteins/metabolism , Humans , Insulin/metabolism , Insulin/pharmacology , Insulin Resistance/physiology , MADS Domain Proteins/genetics , MADS Domain Proteins/metabolism , MEF2 Transcription Factors , Middle Aged , Molecular Chaperones/genetics , Molecular Chaperones/metabolism , Muscle Fibers, Skeletal/cytology , Muscle Fibers, Skeletal/drug effects , Myogenic Regulatory Factors/genetics , Myogenic Regulatory Factors/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Rats , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factors/genetics , Transcription Factors/metabolism
14.
Diabetologia ; 52(12): 2642-52, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19789852

ABSTRACT

AIMS/HYPOTHESIS: Overproduction of phosphoprotein enriched in diabetes (PED, also known as phosphoprotein enriched in astrocytes-15 [PEA-15]) is a common feature of type 2 diabetes and impairs insulin action in cultured cells and in mice. Nevertheless, the potential role of PED in diabetic complications is still unknown. METHODS: We studied the effect of PED overproduction and depletion on kidney function in animal and cellular models. RESULTS: Transgenic mice overexpressing PED (PEDTg) featured age-dependent increases of plasma creatinine levels and urinary volume, accompanied by expansion of the mesangial area, compared with wild-type littermates. Serum and kidney levels of TGF-beta1 were also higher in 6- and 9-month-old PEDTg. Overexpression of PED in human kidney 2 cells significantly increased TGF-beta1 levels, SMAD family members (SMAD)2/3 phosphorylation and fibronectin production. Opposite results were obtained following genetic silencing of PED in human kidney 2 cells by antisense oligonucleotides. Inhibition of phospholipase D and protein kinase C-beta by 2-butanol and LY373196 respectively reduced TGF-beta1, SMAD2/3 phosphorylation and fibronectin production. Moreover, inhibition of TGF-beta1 receptor activity and SMAD2/3 production by SB431542 and antisense oligonucleotides respectively reduced fibronectin secretion by about 50%. TGF-beta1 circulating levels were significantly reduced in Ped knockout mice and positively correlated with PED content in peripheral blood leucocytes of type 2 diabetic patients. CONCLUSIONS/INTERPRETATION: These data indicate that PED regulates fibronectin production via phospholipase D/protein kinase C-beta and TGF-beta1/SMAD pathways in kidney cells. Raised PED levels may therefore contribute to the abnormal accumulation of extracellular matrix and renal dysfunction in diabetes.


Subject(s)
Protein Kinase C/genetics , Transforming Growth Factor beta1/genetics , Actins/genetics , Animals , Astrocytes/metabolism , Blood Pressure , DNA Primers , Diabetes Mellitus, Type 1/complications , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/physiopathology , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/physiopathology , Diabetic Nephropathies/epidemiology , Fatty Acids, Nonesterified/blood , Fibronectins/genetics , Gene Expression Regulation , Heart Rate , Humans , Insulin/blood , Kidney/physiology , Kidney Failure, Chronic/etiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Phenotype , Phosphoproteins/biosynthesis , Phosphoproteins/genetics , Protein Kinase C beta , Reverse Transcriptase Polymerase Chain Reaction , Smad2 Protein/genetics , Up-Regulation
16.
Radiat Prot Dosimetry ; 123(1): 74-82, 2007.
Article in English | MEDLINE | ID: mdl-16849378

ABSTRACT

In recent years, many cases of contamination of metal scraps by unwanted radioactive materials have occurred. Moreover, international organisations are evaluating the possibility to re-use or to recycle metals coming from nuclear power plants. The metal recycling industry has started to worry about radiation exposure of workers that could be in contact with contaminated metals during each manufacturing phase. Risks are strongly dependent on the radiation source features. The aim of this study is to perform risk assessment for workers involved in chemical pickling of steel coils. Monte Carlo simulations have been performed, using the MCNP package and considering coils contaminated with (60)Co, (137)Cs, (241)Am and (226)Ra. Under the most conservative conditions (coil contaminated with 1.0 kBq g(-1) of (60)Co), the dose assessment results lower than the European dose limit for the population (1 mSv y(-1)), considering a maximum number of 10 contaminated coils handled per year. The only exception concerns the case of (241)Am, for which internal contamination could be non- negligible and should be verified in the specific cases. In every case, radiation exposure risk for people standing at 50 m from the coil is widely <1 mSv y(-1).


Subject(s)
Equipment Contamination/prevention & control , Metallurgy/methods , Occupational Exposure/adverse effects , Radiation Injuries/prevention & control , Radioisotopes/analysis , Body Burden , Humans , Metals/analysis , Nuclear Reactors , Power Plants , Radiation Monitoring , Radiation Protection , Relative Biological Effectiveness , Risk Assessment , Risk Factors
17.
Diabetologia ; 49(12): 3058-66, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17021921

ABSTRACT

AIMS/HYPOTHESIS: Overexpression of the gene encoding phosphoprotein enriched in astrocytes 15 (PEA15), also known as phosphoprotein enriched in diabetes (PED), causes insulin resistance and diabetes in transgenic mice and has been observed in type 2 diabetic individuals. The aim of this study was to investigate whether PEA15 overexpression occurs in individuals at high risk of diabetes and whether it is associated with specific type 2 diabetes subphenotypes. SUBJECTS AND METHODS: We analysed PEA15 expression in euglycaemic first-degree relatives (FDR) of type 2 diabetic subjects. RESULTS: The expression of PEA15 in peripheral blood leucocytes (PBLs) paralleled that in fat and skeletal muscle tissues. In PBLs from the FDR, PEA15 expression was two-fold higher than in euglycaemic individuals with no family history of diabetes (control subjects), both at the protein and the mRNA level (p < 0.001). The expression of PEA15 was comparable in FDR and type 2 diabetic subjects and in each group close to one-third of the subjects expressed PEA15 levels more than 2 SD higher than the mean of control subjects. Subjects with IFG with at least one type 2 diabetes-affected FDR also overexpressed PEA15 (p < 0.05). In all the groups analysed, PEA15 expression was independent of sex and unrelated to age, BMI, waist circumference, systolic and diastolic BP, and fasting cholesterol, triacylglycerol and glucose levels. However, in euglycaemic FDR of type 2 diabetic subjects, PEA15 expression was inversely correlated with insulin sensitivity (r = -557, p = 0.01). CONCLUSIONS/INTERPRETATION: We conclude that PEA15 overexpression represents a common defect in FDR of patients with type 2 diabetes and is correlated with reduced insulin sensitivity in these individuals.


Subject(s)
Diabetes Mellitus, Type 2/genetics , Gene Expression Regulation , Insulin Resistance/genetics , Intracellular Signaling Peptides and Proteins/genetics , Phosphoproteins/genetics , Adult , Apoptosis Regulatory Proteins , Blood Glucose/metabolism , DNA Primers , Diabetes Mellitus, Type 2/physiopathology , Family , Female , Humans , Male , Phosphoproteins/metabolism , RNA/genetics , RNA/isolation & purification , Reverse Transcriptase Polymerase Chain Reaction
18.
J Biol Chem ; 276(48): 45088-97, 2001 Nov 30.
Article in English | MEDLINE | ID: mdl-11577086

ABSTRACT

In L6 skeletal muscle cells and immortalized hepatocytes, insulin induced a 2-fold increase in the activity of the pyruvate dehydrogenase (PDH) complex. This effect was almost completely blocked by the protein kinase C (PKC) delta inhibitor Rottlerin and by PKCdelta antisense oligonucleotides. At variance, overexpression of wild-type PKCdelta or of an active PKCdelta mutant induced PDH complex activity in both L6 and liver cells. Insulin stimulation of the activity of the PDH complex was accompanied by a 2.5-fold increase in PDH phosphatases 1 and 2 (PDP1/2) activity with no change in the activity of PDH kinase. PKCdelta antisense blocked insulin activation of PDP1/2, the same as with PDH. In insulin-exposed cells, PDP1/2 activation was paralleled by activation and mitochondrial translocation of PKCdelta, as revealed by cell subfractionation and confocal microscopy studies. The mitochondrial translocation of PKCdelta, like its activation, was prevented by Rottlerin. In extracts from insulin-stimulated cells, PKCdelta co-precipitated with PDP1/2. PKCdelta also bound to PDP1/2 in overlay blots, suggesting that direct PKCdelta-PDP interaction may occur in vivo as well. In intact cells, insulin exposure determined PDP1/2 phosphorylation, which was specifically prevented by PKCdelta antisense. PKCdelta also phosphorylated PDP in vitro, followed by PDP1/2 activation. Thus, in muscle and liver cells, insulin causes activation and mitochondrial translocation of PKCdelta, accompanied by PDP phosphorylation and activation. These events are necessary for insulin activation of the PDH complex in these cells.


Subject(s)
Insulin/metabolism , Isoenzymes/chemistry , Isoenzymes/metabolism , Liver/enzymology , Muscles/enzymology , Protein Kinase C/chemistry , Protein Kinase C/metabolism , Pyruvate Dehydrogenase Complex/metabolism , Animals , Cell Line , Enzyme Activation , Enzyme Inhibitors/pharmacology , Humans , Liver/cytology , Microscopy, Fluorescence , Muscle, Skeletal/enzymology , Muscles/cytology , Mutation , Phosphorylation , Precipitin Tests , Protein Binding , Protein Isoforms , Protein Kinase C-delta , Protein Transport , Rats , Time Factors , Transfection
19.
J Biol Chem ; 276(40): 37109-19, 2001 Oct 05.
Article in English | MEDLINE | ID: mdl-11481324

ABSTRACT

We have investigated glycogen synthase (GS) activation in L6hIR cells expressing a peptide corresponding to the kinase regulatory loop binding domain of insulin receptor substrate-2 (IRS-2) (KRLB). In several clones of these cells (B2, F4), insulin-dependent binding of the KRLB to insulin receptors was accompanied by a block of IRS-2, but not IRS-1, phosphorylation, and insulin receptor binding. GS activation by insulin was also inhibited by >70% in these cells (p < 0.001). The impairment of GS activation was paralleled by a similarly sized inhibition of glycogen synthase kinase 3 alpha (GSK3 alpha) and GSK3 beta inactivation by insulin with no change in protein phosphatase 1 activity. PDK1 (a phosphatidylinositol trisphosphate-dependent kinase) and Akt/protein kinase B (PKB) activation by insulin showed no difference in B2, F4, and in control L6hIR cells. At variance, insulin did not activate PKC zeta in B2 and F4 cells. In L6hIR, inhibition of PKC zeta activity by either a PKC zeta antisense or a dominant negative mutant also reduced by 75% insulin inactivation of GSK3 alpha and -beta (p < 0.001) and insulin stimulation of GS (p < 0.002), similar to Akt/PKB inhibition. In L6hIR, insulin induced protein kinase C zeta (PKC zeta) co-precipitation with GSK3 alpha and beta. PKC zeta also phosphorylated GSK3 alpha and -beta. Alone, these events did not significantly affect GSK3 alpha and -beta activities. Inhibition of PKC zeta activity, however, reduced Akt/PKB phosphorylation of the key serine sites on GSK3 alpha and -beta by >80% (p < 0.001) and prevented full GSK3 inactivation by insulin. Thus, IRS-2, not IRS-1, signals insulin activation of GS in the L6hIR skeletal muscle cells. In these cells, insulin inhibition of GSK3 alpha and -beta requires dual phosphorylation by both Akt/PKB and PKC zeta.


Subject(s)
Muscle, Skeletal/enzymology , Phosphoproteins/metabolism , Protein Kinase C/metabolism , Protein Serine-Threonine Kinases , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cells, Cultured , Enzyme Activation , Glycogen Synthase Kinase 3 , Glycogen Synthase Kinases , Humans , Insulin/physiology , Insulin Receptor Substrate Proteins , Intracellular Signaling Peptides and Proteins , Muscle, Skeletal/cytology , Muscle, Skeletal/metabolism , Peptides/metabolism , Phosphorylation , Protein Structure, Tertiary , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , Signal Transduction , Viral Proteins/metabolism
20.
J Endocrinol Invest ; 24(6): 460-7, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11434672

ABSTRACT

Insulin action on target tissues is mediated by specific tyrosine kinase receptors. Upon ligand binding insulin receptors autophosphorylate and phosphorylate intracellular substrates on tyrosine residues. These early events of insulin action are followed by the activation of a number of enzymes, including protein kinase C (PKC). At least 14 PKC isoforms have been identified and cloned to date. PKCs appear to play dual roles in insulin signaling. For instance, they are involved in transduction of specific insulin signals but also contribute to the generation of insulin resistance. In this article, we will analyze the experimental evidence addressing the mechanism by which insulin might activate individual PKC isoforms as well as the role of single PKCs in insulin-induced bioeffects.


Subject(s)
Insulin/pharmacology , Isoenzymes/physiology , Protein Kinase C/physiology , Animals , Enzyme Activation/drug effects , Humans , Phosphorylation , Receptor, Insulin/metabolism , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...