Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
Food Funct ; 15(11): 5842-5854, 2024 Jun 04.
Article in English | MEDLINE | ID: mdl-38767145

ABSTRACT

Background: Pure bergamot juice exerts lipid lowering effects in dyslipidemic subjects. It is unknown whether bergamot-based beverages exert similar effects in healthy subjects. Aim: To assess the effects, if any, of a bergamot-based beverage (BBB, bergamot juice ≤25%) on lipid, metabolic and inflammatory biomarkers. Methods: Forty-five healthy subjects were randomised 1 : 1 to BBB intake (400 mL day-1) (55.5%) or control (44.5%) for 12 weeks. Anthropometric (waist circumference, body mass index (BMI)) and clinical (blood pressure) parameters, blood samples (glucose, glycated haemoglobin, insulinemia, lipid profile, liver and renal function, inflammatory biomarkers) and 24-h urine for the analysis of (poly)phenol metabolites were collected at the baseline and at 12 weeks. Intakes of energy, nutrients and food groups were assessed by a 7-day dietary record. Results: Both groups exhibited a time-related significant decrease in total cholesterol (p = 0.02), fasting plasma glucose (p = 0.016), insulin (p = 0.034), BMI (p < 0.001) and waist circumference (p = 0.04), but with no significant between-arm difference. The urinary profile of metabolites from the BBB-derived (poly)phenols well discriminated the two study groups, documenting good compliance in the intervention arm. Notably, urinary bergamot 3-hydroxy-3-methylglutaryl (HMG) -containing flavanones or derived HMG-containing metabolites were not detectable. BBB was well tolerated and no adverse events were recorded. Conclusion: This first randomized controlled trial of BBB consumption in healthy subjects showed no effects of BBB on the cardiometabolic risk profile. BBB consumption is a safe nutritional adjunct in the context of a well balanced diet.


Subject(s)
Biomarkers , Blood Glucose , Lipids , Humans , Male , Female , Adult , Biomarkers/blood , Biomarkers/urine , Middle Aged , Blood Glucose/metabolism , Lipids/blood , Cardiometabolic Risk Factors , Healthy Volunteers , Young Adult , Insulin/blood , Fruit and Vegetable Juices , Body Mass Index , Inflammation , Waist Circumference , Cardiovascular Diseases/prevention & control
2.
Front Endocrinol (Lausanne) ; 13: 972890, 2022.
Article in English | MEDLINE | ID: mdl-36339405

ABSTRACT

Ketogenesis takes place in hepatocyte mitochondria where acetyl-CoA derived from fatty acid catabolism is converted to ketone bodies (KB), namely ß-hydroxybutyrate (ß-OHB), acetoacetate and acetone. KB represent important alternative energy sources under metabolic stress conditions. Ketogenic diets (KDs) are low-carbohydrate, fat-rich eating strategies which have been widely proposed as valid nutritional interventions in several metabolic disorders due to its substantial efficacy in weight loss achievement. Carbohydrate restriction during KD forces the use of FFA, which are subsequently transformed into KB in hepatocytes to provide energy, leading to a significant increase in ketone levels known as "nutritional ketosis". The recent discovery of KB as ligands of G protein-coupled receptors (GPCR) - cellular transducers implicated in a wide range of body functions - has aroused a great interest in understanding whether some of the clinical effects associated to KD consumption might be mediated by the ketone/GPCR axis. Specifically, anti-inflammatory effects associated to KD regimen are presumably due to GPR109A-mediated inhibition of NLRP3 inflammasome by ß-OHB, whilst lipid profile amelioration by KDs could be ascribed to the actions of acetoacetate via GPR43 and of ß-OHB via GPR109A on lipolysis. Thus, this review will focus on the effects of KD-induced nutritional ketosis potentially mediated by specific GPCRs in metabolic and endocrinological disorders. To discriminate the effects of ketone bodies per se, independently of weight loss, only studies comparing ketogenic vs isocaloric non-ketogenic diets will be considered as well as short-term tolerability and safety of KDs.


Subject(s)
Diet, Ketogenic , Ketosis , Humans , Ketone Bodies/metabolism , Acetoacetates , 3-Hydroxybutyric Acid/metabolism , 3-Hydroxybutyric Acid/pharmacology , Receptors, G-Protein-Coupled , Ketones , Carbohydrates , Weight Loss
3.
Cardiovasc Diabetol ; 21(1): 27, 2022 02 17.
Article in English | MEDLINE | ID: mdl-35177077

ABSTRACT

BACKGROUND: Empagliflozin can curb inflammation and oxidative stress, through sodium-proton exchanger (NHE) inhibition, in a model of lipotoxicity in human myeloid angiogenic cells (MAC), which mediate endothelial repairing processes. Aim of this study is to assess in human MAC whether: (1) Stearic acid (SA) induced inflammation and increase in oxidant stress is accompanied by bioenergetic alterations; (2) empagliflozin anti-lipotoxic action is concomitant with coherent changes in bioenergetic metabolism, possibly via NHE blockade. METHODS: MAC were isolated from peripheral blood of healthy volunteers and incubated in the presence/absence of SA (100 µM for 3 h) with/without empagliflozin (EMPA 100 µM) or amiloride (Ami 100 µM) for 1 h. Cell respiration (oxygen consumption rate OCR) and anaerobic glycolysis (measured as proton production rate) were recorded in real-time by Seahorse technology, and ATP production (anaerobic glycolysis- and oxphos-derived) rates were calculated. RESULTS: SA, at the concentration causing inflammation and increased oxidant stress, altered cell bioenergetics of human MAC, with overall reductions in basal OCR and oxphos-derived ATP production (all p < 0.05), pointing to mitochondrial alterations. EMPA, at the concentration counteracting SA-induced lipotoxicity, both alone and in the presence of SA, caused NHE-independent extensive bioenergetic alterations (from p < 0.05 to p < 0.01), greater than those induced by SA alone. CONCLUSIONS: In human MAC: (1) SA altered cell bioenergetics, concomitantly with inflammation and oxidant stress; (2) EMPA possibly inhibited mitochondrial respiration, (3) the protective effect of EMPA against SA-induced lipotoxicity was unlikely to be mediated through bioenergetic metabolism.


Subject(s)
Benzhydryl Compounds , Glucosides , Benzhydryl Compounds/toxicity , Energy Metabolism , Glucosides/pharmacology , Humans , Sodium/metabolism
4.
Cell Biol Toxicol ; 38(4): 667-678, 2022 08.
Article in English | MEDLINE | ID: mdl-35015170

ABSTRACT

INTRODUCTION: The inflammatory potential of SARS-CoV-2 Spike S1 (Spike) has never been tested in human primary macrophages (MΦ). Different recombinant Spikes might display different effects in vitro, according to protein length and glycosylation, and endotoxin (lipopolysaccharide, LPS) contamination. OBJECTIVES: To assess (1) the effects of different Spikes on human primary MΦ inflammation; (2) whether LPS contamination of recombinant Spike is (con)cause in vitro of increased MΦ inflammation. METHODS: Human primary MΦ were incubated in the presence/absence of several different Spikes (10 nM) or graded concentrations of LPS. Pro-inflammatory marker expression (qPCR and ELISA) and supernatant endotoxin contamination (LAL test) were the main readouts. RESULTS: LPS-free, glycosylated Spike (the form expressed in infected humans) caused no inflammation in human primary MΦ. Two (out of five) Spikes were contaminated with endotoxins ≥ 3 EU/ml and triggered inflammation. A non-contaminated non-glycosylated Spike produced in E. coli induced MΦ inflammation. CONCLUSIONS: Glycosylated Spike per se is not pro-inflammatory for human MΦ, a feature which may be crucial to evade the host innate immunity. In vitro studies with commercially available Spike should be conducted with excruciating attention to potential LPS contamination.


Subject(s)
Endotoxins , Macrophages , Spike Glycoprotein, Coronavirus , COVID-19 , Endotoxins/toxicity , Escherichia coli , Glycosylation , Humans , Inflammation/chemically induced , Inflammation/metabolism , Lipopolysaccharides/toxicity , Macrophages/metabolism , Macrophages/virology , SARS-CoV-2 , Spike Glycoprotein, Coronavirus/metabolism
5.
Eur J Nutr ; 60(3): 1453-1463, 2021 Apr.
Article in English | MEDLINE | ID: mdl-32728879

ABSTRACT

PURPOSE: Coffee is an important source of bioactive compounds, including caffeine, trigonelline, and phenolic compounds. Several studies have highlighted the preventive effects of coffee consumption on major cardiometabolic (CM) diseases, but the impact of different coffee dosages on markers of CM risk in a real-life setting has not been fully understood. This study aimed to investigate the effect of coffee and cocoa-based confectionery containing coffee consumption on several CM risk factors in healthy subjects. METHODS: In a three-arm, crossover, randomized trial, 21 volunteers were assigned to consume in a random order for 1 month: 1 cup of espresso coffee/day, 3 cups of espresso coffee/day, and 1 cup of espresso coffee plus 2 cocoa-based products containing coffee, twice per day. At the last day of each treatment, blood samples were collected and used for the analysis of inflammatory markers, trimethylamine N-oxide, nitric oxide, blood lipids, and markers of glucose/insulin metabolism. Moreover, anthropometric parameters and blood pressure were measured. Finally, food consumption during the interventions was monitored. RESULTS: After 1 month, energy intake did not change among treatments, while significant differences were observed in the intake of saturated fatty acids, sugars, and total carbohydrates. No significant effect on CM markers was observed following neither the consumption of different coffee dosages nor after cocoa-based products containing coffee. CONCLUSIONS: The daily consumption of common dosages of coffee and its substitution with cocoa-based products containing coffee showed no effect on CM risk factors in healthy subjects. TRIAL REGISTRATION NUMBER: Registered at clinicaltrials.gov as NCT03166540, May 21, 2017.


Subject(s)
Cacao , Cardiovascular Diseases , Chocolate , Candy , Cardiovascular Diseases/epidemiology , Cardiovascular Diseases/prevention & control , Coffee , Cross-Over Studies , Humans
6.
Cardiovasc Diabetol ; 19(1): 46, 2020 04 07.
Article in English | MEDLINE | ID: mdl-32264868

ABSTRACT

BACKGROUND: The clear evidence of cardiovascular benefits in cardiovascular outcome trials of sodium-glucose cotransporter 2 inhibitors (SGLT2i) in type 2 diabetes might suggest an effect on atherosclerotic plaque vulnerability and/or thrombosis, in which myeloid angiogenic cells (MAC) and platelets (PLT) are implicated. We tested the effects of SGLT2i on inflammation and oxidant stress in a model of stearic acid (SA)-induced lipotoxicity in MAC and on PLT activation. The possible involvement of the Na+/H+ exchanger (NHE) was also explored. METHOD: MAC and PLT were isolated from peripheral blood of healthy subjects and incubated with/without SGLT2i [empagliflozin (EMPA) and dapagliflozin (DAPA) 1-100 µM] to assess their effects on SA (100 µM)-induced readouts of inflammation, oxidant stress and apoptosis in MAC and on expression of PLT activation markers by flow-cytometry after ADP-stimulation. Potential NHE involvement was tested with amiloride (aspecific NHE inhibitor) or cariporide (NHE1 inhibitor). Differences among culture conditions were identified using one-way ANOVA or Friedman test. RESULTS: NHE isoforms (1,5-9), but not SGLT2 expression, were expressed in MAC and PLT. EMPA and DAPA (100 µM) significantly reduced SA-induced inflammation (IL1ß, TNFα, MCP1), oxidant stress (SOD2, TXN, HO1), but not apoptosis in MAC. EMPA and DAPA (both 1 µM) reduced PLT activation (CD62p and PAC1 expression). SGLT2i effects were mimicked by amiloride, and only partially by cariporide, in MAC, and by both inhibitors in PLT. CONCLUSIONS: EMPA and DAPA ameliorated lipotoxic damage in stearate-treated MAC, and reduced ADP-stimulated PLT activation, potentially via NHE-inhibition, thereby pointing to plaque stabilization and/or thrombosis inhibition as potential mechanism(s) involved in SGLT2i-mediated cardiovascular protection.


Subject(s)
Adenosine Diphosphate/pharmacology , Benzhydryl Compounds/pharmacology , Blood Platelets/drug effects , Endothelial Progenitor Cells/drug effects , Glucosides/pharmacology , Platelet Activation/drug effects , Sodium-Glucose Transporter 2 Inhibitors/pharmacology , Sodium-Glucose Transporter 2/metabolism , Stearic Acids/toxicity , Apoptosis/drug effects , Blood Platelets/metabolism , Blood Platelets/pathology , Cells, Cultured , Endothelial Progenitor Cells/metabolism , Endothelial Progenitor Cells/pathology , Humans , Inflammation Mediators/metabolism , Oxidative Stress/drug effects , Signal Transduction , Sodium-Hydrogen Exchangers/metabolism
8.
Nutrients ; 9(12)2017 Dec 06.
Article in English | MEDLINE | ID: mdl-29211032

ABSTRACT

Myeloid angiogenic cells (MACs) play a key role in endothelial repairing processes and functionality but their activity may be impaired by the lipotoxic effects of some molecules like stearic acid (SA). Among the dietary components potentially able to modulate endothelial function in vivo, (poly)phenolic compounds represent serious candidates. Here, we apply a comprehensive multidisciplinary approach to shed light on the prospects of Bergamot (Citrus bergamia), a citrus fruit rich in flavanones and other phenolic compounds, in the framework of lipotoxicity-induced MACs impairment. The flavanone profile of bergamot juice was characterized and 16 compounds were identified, with a new 3-hydroxy-3-methylglutaryl (HMG) flavanone, isosakuranetin-7-O-neohesperidoside-6″-O-HMG, described for the first time. Then, a pilot bioavailability study was conducted in healthy volunteers to assess the circulating flavanone metabolites in plasma and urine after consumption of bergamot juice. Up to 12 flavanone phase II conjugates (sulfates and glucuronides of hesperetin, naringenin and eriodyctiol) were detected and quantified. Finally, the effect of some of the metabolites identified in vivo, namely hesperetin-7-O-glucuronide, hesperetin-3'-O-glucuronide, naringenin-7-O-glucuronide and naringenin-4'-O-glucuronide, was tested, at physiological concentrations, on gene expression of inflammatory markers and apoptosis in MACs exposed to SA. Under these conditions, naringenin-4'-O-glucuronide and hesperetin-7-O-glucuronide were able to modulate inflammation, while no flavanone glucuronide was effective in curbing stearate-induced lipoapoptosis. These results demonstrate that some flavanone metabolites, derived from the in vivo transformation of bergamot juice phenolics in humans, may mitigate stearate-induced inflammation in MACs.


Subject(s)
Citrus/chemistry , Flavanones/pharmacokinetics , Myeloid Cells/drug effects , Neovascularization, Physiologic/physiology , Adult , Apoptosis , Biological Availability , Cells, Cultured , Chromatography, Liquid/methods , Female , Flavanones/chemistry , Humans , Male , Mass Spectrometry/methods , Molecular Structure , Myeloid Cells/physiology , Phenols/blood , Phenols/urine
9.
Atherosclerosis ; 265: 162-171, 2017 10.
Article in English | MEDLINE | ID: mdl-28892713

ABSTRACT

BACKGROUND AND AIMS: Saturated free fatty acids (SFAs) can induce lipotoxicity in different cells. No studies have investigated the effects of SFA in circulating angiogenic cells (CACs), which play a key role in endothelial repair processes. The aim of the study was to assess the effects of SFAs, specifically stearic acid (SA), on viability and function of CACs and to investigate potential underlying molecular mechanisms. METHODS: CACs were isolated from healthy subjects by established methods. CACs were incubated with BSA-complexed stearate (100 µM) to assess the time course (from 8 to 24 h exposure) of the effects on viability and apoptosis (activation of caspases 3/7), angiogenic function (tube formation assay), pro-inflammatory cytokine (IL-1ß, IL-6, IL-8, MCP-1 and TNFα) gene expression (qPCR) and secretion (ELISA), activation of MAPK (JNK, p38 and Erk1/2) by Western blot and endoplasmic reticulum (ER) stress marker (CHOP, BIP, ATF4, XBP-1 and sXBP-1) gene expression by qPCR. RESULTS: Stearic acid activates effector caspases in CACs in a dose- and time-dependent manner. SA also impairs CAC function and increases pro-inflammatory molecule (IL-1ß, IL-6, IL-8, MCP-1 and TNFα) gene expression and secretion in CACs starting from 3 h of incubation. The activation of JNK by SA mediates pro-inflammatory response, but it may be not necessary for apoptosis. Moreover, SA induces the expression of ER stress markers across the three branches of the ER stress response. CONCLUSIONS: In humans, both function and viability of CACs are exquisitely vulnerable to physiologic concentrations of stearate; lipotoxic impairment of endothelial repair processes may be implicated in vascular damage caused by SFAs.


Subject(s)
Metabolic Syndrome/blood , Monocytes/drug effects , Stearic Acids/adverse effects , Apoptosis/drug effects , Cells, Cultured , Humans , Inflammation/chemically induced , Lipid Metabolism , Metabolic Syndrome/metabolism , Monocytes/physiology , Neovascularization, Physiologic , Stearic Acids/administration & dosage
10.
PLoS One ; 12(8): e0182559, 2017.
Article in English | MEDLINE | ID: mdl-28777829

ABSTRACT

Insulin resistance is considered to be a pathogenetic mechanism in several and diverse diseases (e.g. type 2 diabetes, atherosclerosis) often antedating them in apparently healthy subjects. The aim of this study is to investigate with a microarray based approach whether IR per se is characterized by a specific pattern of gene expression. For this purpose we analyzed the transcriptomic profile of peripheral blood mononuclear cells in two groups (10 subjects each) of healthy individuals, with extreme insulin resistance or sensitivity, matched for BMI, age and gender, selected within the MultiKnowledge Study cohort (n = 148). Data were analyzed with an ad-hoc rank-based classification method. 321 genes composed the gene set distinguishing the insulin resistant and sensitive groups, within which the "Adrenergic signaling in cardiomyocytes" KEGG pathway was significantly represented, suggesting a pattern of increased intracellular cAMP and Ca2+, and apoptosis in the IR group. The same pathway allowed to discriminate between insulin resistance and insulin sensitive subjects with BMI >25, supporting his role as a biomarker of IR. Moreover, ASCM pathway harbored biomarkers able to distinguish healthy and diseased subjects (from publicly available data sets) in IR-related diseases involving excitable cells: type 2 diabetes, chronic heart failure, and Alzheimer's disease. The altered gene expression profile of the ASCM pathway is an early molecular signature of IR and could provide a common molecular pathogenetic platform for IR-related disorders, possibly representing an important aid in the efforts aiming at preventing, early detecting and optimally treating IR-related diseases.


Subject(s)
Alzheimer Disease/genetics , Biomarkers/metabolism , Diabetes Mellitus, Type 2/genetics , Heart Failure/genetics , Insulin Resistance/genetics , Leukocytes, Mononuclear/metabolism , Transcriptome , Adult , Alzheimer Disease/blood , Blood Glucose/metabolism , Case-Control Studies , Diabetes Mellitus, Type 2/blood , Female , Healthy Volunteers , Heart Failure/blood , Humans , Male
11.
Acta Biomed ; 88(2): 172-177, 2017 08 23.
Article in English | MEDLINE | ID: mdl-28845832

ABSTRACT

BACKGROUND AND AIM OF THE STUDY: Physical performance is the result of a complex combination of several factors such as genetic and anthropometric aspects, nutrition and hormonal status. In the past few years many studies have considered the impact of vitamin D on muscular strength and athletic performance.The aim of the present study was to assess the anthropometric measures impacting on physical performance in a group of professional rugby athletes.  As a secondary aim we investigated a possible relationship between baseline vitamin D status and athletic performance status in these subjects. METHODS: All rugby players completed a test-retest reliability study on performance measures, as 70kg jump squat and body weight (BW) jump squat to assess musculoskeletal performance. Additionally at the time point we collected a blood sample of every athletes for the assessment of serum vitamin D. RESULTS: We found that lean mass was an important independent predictor of performance score in 70kg jump squat (p=0.007, R2=0.74) and BW jump squat (p=0.010, R2=0.66) in these well trained athletes. No statistically significant association was present between performance score and serum vitamin D in this specific setting. CONCLUSIONS: We demonstrate a positive interaction between lower limb lean mass and performance score, but we have not been able to identify any statistically significant association between worsening in performance measures and decrease of serum 25 OH Vitamin D.


Subject(s)
Anthropometry , Athletic Performance , Football , Adult , Body Mass Index , Body Weight , Humans , Male , Reproducibility of Results , Vitamin D/analogs & derivatives , Vitamin D/blood
12.
Int J Mol Sci ; 18(7)2017 Jul 12.
Article in English | MEDLINE | ID: mdl-28704936

ABSTRACT

Increased non high-density lipoprotein (HDL)/low-density lipoprotein (LDL) cholesterol levels are independent risk factors for cardiovascular (CV) mortality with no documented threshold. A new combination of nutraceuticals (berberine 200 mg, monacolin K 3 mg, chitosan 10 mg and coenzyme Q 10 mg) with additive lipid-lowering properties has become available. The aim of the study is to test the efficacy of the nutraceutical formulation (one daily) in lowering non-HDL cholesterol vs. placebo at 12 weeks in individuals with non-HDL-cholesterol levels ≥160 mg/dL. 39 subjects (age 52 ± 11 years; 54% females; body mass index 27 ± 4 kg/m²) were randomized (3:1) in a double blind phase II placebo-controlled study. At baseline, 4 and 12 weeks main clinical/biohumoral parameters, pro-inflammatory cytokines, (gut)-hormones, proprotein convertase subtilisin/kexin type 9 (PCSK9) levels and endothelial progenitor cell (EPC) number were assessed. Baseline characteristics were comparable in the two groups. The intervention significantly decreased non-HDL cholesterol (-30 ± 20 mg/dL; p = 0.012), LDL cholesterol (-31 ± 18 mg/dL, p = 0.011) and apolipoprotein (Apo) B (-14 ± 12 mg/dL, p = 0.030) levels compared to the placebo. Pro-inflammatory, hormonal, PCSK9 and EPC levels remained stable throughout the study in both groups. The intervention was well tolerated. Three adverse events occurred: Epstein Barr virus infection, duodenitis and asymptomatic but significant increase in creatine phosphokinase (following intense physical exercise) which required hospitalization. The tested nutraceutical formulation may represent a possible therapeutic strategy in dyslipidemic individuals in primary prevention.


Subject(s)
Berberine/therapeutic use , Biological Products/therapeutic use , Chitosan/therapeutic use , Cholesterol/blood , Dyslipidemias/blood , Dyslipidemias/drug therapy , Adult , Aged , Drug Compounding , Female , Humans , Male , Middle Aged , Proprotein Convertase 9/metabolism
13.
Cardiovasc Diabetol ; 16(1): 27, 2017 02 23.
Article in English | MEDLINE | ID: mdl-28231835

ABSTRACT

BACKGROUND: Fewer circulating endothelial progenitor cells (EPCs) and increased plasma (C-term) stromal cell-derived factor 1α (SDF-1α), a substrate of DPP-4, are biomarkers, and perhaps mediators, of cardiovascular risk and mortality. Short-term/acute treatment with DPP-4 inhibitors improve EPC bioavailability; however, long-term effects of DPP-4i on EPCs bioavailability/plasma (C-term) SDF-1α are unknown. METHODS: Randomized (2:1) open-label trial to compare the effects of vildagliptin (V) (100 mg/day) vs glibenclamide (G) (2.5 mg bid to a maximal dose of 5 mg bid) on circulating EPC levels at 4 and 12 months of treatment in 64 patients with type 2 diabetes in metformin failure. At baseline, and after 4 and 12 months, main clinical/biohumoral parameters, inflammatory biomarkers, concomitant therapies, EPC number (CD34+/CD133+/KDR+/106 cytometric events) and plasma (C-term) SDF-1α (R&D system) were assessed. RESULTS: Baseline characteristics were comparable in the two groups. V and G similarly and significantly (p < 0.0001) improved glucose control. At 12 months, V significantly increased EPC number (p < 0.05) and significantly reduced (C-term) SDF-1α plasma levels (p < 0.01) compared to G, with no differences in inflammatory biomarkers. CONCLUSIONS: V exerts a long-term favorable effect on EPC and (C-term) SDF-1α levels at glucose equipoise, thereby implying a putative beneficial effect on vascular integrity. Trial registration Clinical Trials number: NCT01822548; name: Effect of Vildagliptin vs. Glibenclamide on Circulating Endothelial Progenitor Cell Number Type 2 Diabetes. Registered 28 March, 2013.


Subject(s)
Adamantane/analogs & derivatives , Diabetes Mellitus, Type 2/drug therapy , Dipeptidyl-Peptidase IV Inhibitors/therapeutic use , Endothelial Progenitor Cells/drug effects , Glyburide/therapeutic use , Hypoglycemic Agents/therapeutic use , Nitriles/therapeutic use , Pyrrolidines/therapeutic use , Adamantane/pharmacology , Adamantane/therapeutic use , Aged , Cell Count/methods , Chemokine CXCL12/blood , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/physiopathology , Dipeptidyl-Peptidase IV Inhibitors/pharmacology , Endothelial Progenitor Cells/physiology , Female , Follow-Up Studies , Glyburide/pharmacology , Humans , Hypoglycemic Agents/pharmacology , Male , Middle Aged , Nitriles/pharmacology , Pyrrolidines/pharmacology , Time Factors , Vildagliptin
14.
Molecules ; 21(8)2016 Aug 02.
Article in English | MEDLINE | ID: mdl-27490528

ABSTRACT

The consumption of foodstuffs yielding circulating compounds able to maintain endothelial function by improving nitric oxide (NO) bioavailability can be considered as an effective strategy for cardiovascular disease prevention. This work assessed the in vitro effects of urolithin A, urolithin B, and urolithin B-glucuronide, ellagitannin-derived metabolites of colonic origin, on NO release and endothelial NO synthase (eNOS) activation in primary human aortic endothelial cells (HAECs). Urolithins were tested both individually at 15 µM and as a mixture of 5 µM each, at different time points. The biotransformation of these molecules in cell media due to cell metabolism was also evaluated by UHPLC-MS(n). The mix of urolithins at 5 µM significantly increased nitrite/nitrate levels following 24 h of incubation, while single urolithins at 15 µM did not modify NO bioavailability. Both the mix of urolithins at 5 µM and urolithin B-glucuronide at 15 µM activated eNOS expression. All urolithins underwent metabolic reactions, but these were limited to conjugation with sulfate moieties. This study represents a step forward in the understanding of cardiovascular health benefits of ellagitannin-rich foodstuffs and backs the idea that peripheral cells may contribute to urolithin metabolism.


Subject(s)
Aorta/cytology , Coumarins/pharmacology , Endothelial Cells/drug effects , Nitric Oxide/metabolism , Cells, Cultured , Endothelial Cells/cytology , Gastrointestinal Tract/metabolism , Glucuronides/chemistry , Glucuronides/pharmacology , Humans , Hydrolyzable Tannins/chemistry
15.
Acta Diabetol ; 53(4): 661-7, 2016 Aug.
Article in English | MEDLINE | ID: mdl-27020053

ABSTRACT

AIMS: Individuals with type 2 diabetes show shorter leukocyte telomere length (LTL) compared to people without diabetes. Reduced LTL is associated with increased carotid intima-media thickness (IMT) in healthy subjects. The aim of the study is to assess whether LTL also correlates with IMT in patients with diabetes. METHODS: In a cohort of 104 subjects with type 2 diabetes and atherogenic dyslipidemia, we assessed anthropometric, hemodynamic and metabolic parameters. Common carotid IMT was expressed as the maximum IMT. LTL was assessed by a specific real-time PCR reaction. RESULTS: At univariate analysis, IMT values were positively correlated with age (p < 0.001), previous history of cardiovascular events (p < 0.005), fasting plasma glucose (p < 0.01), HbA1c (p < 0.05) and negatively correlated with LTL (p < 0.05). In a multivariate model, age (p < 0.001) and LTL (p < 0.05) were the only independent predictors of maximum IMT, with an adjusted R (2) of 0.22. CONCLUSIONS: LTL is an independent predictor of subclinical atherosclerosis pointing to a role of LTL as an early marker of vascular burden and cardiovascular disease also in type 2 diabetes.


Subject(s)
Atherosclerosis/genetics , Diabetes Mellitus, Type 2/genetics , Diabetic Angiopathies/genetics , Telomere Shortening , Aged , Atherosclerosis/complications , Biomarkers/blood , Carotid Intima-Media Thickness , Cross-Sectional Studies , Diabetes Mellitus, Type 2/complications , Female , Humans , Leukocytes/cytology , Male , Middle Aged , Real-Time Polymerase Chain Reaction
16.
PLoS One ; 10(3): e0119751, 2015.
Article in English | MEDLINE | ID: mdl-25799240

ABSTRACT

BACKGROUND: Macrophages are a heterogeneous cell population which in response to the cytokine milieu polarize in either classically activated macrophages (M1) or alternatively activated macrophages (M2). This plasticity makes macrophages essential in regulating inflammation, immune response and tissue remodeling and a novel therapeutic target in inflammatory diseases such as atherosclerosis. The aim of the study was to describe the transcriptomic profiles of differently polarized human macrophages to generate new hypotheses on the biological function of the different macrophage subtypes. METHODS AND RESULTS: Polarization of circulating monocytes/macrophages of blood donors was induced in vitro by IFN-γ and LPS (M1), by IL-4 (M2a), and by IL-10 (M2c). Unstimulated cells (RM) served as time controls. Gene expression profile of M1, M2a, M2c and RM was assessed at 6, 12 and 24h after polarization with Whole Human Genome Agilent Microarray technique. When compared to RM, M1 significantly upregulated pathways involved in immunity and inflammation, whereas M2a did the opposite. Conversely, decreased and increased expression of mitochondrial metabolism, consistent with insulin resistant and insulin sensitive patterns, was seen in M1 and M2a, respectively. The time sequence in the expression of some pathways appeared to have some specific bearing on M1 function. Finally, canonical and non-canonical Wnt genes and gene groups, promoting inflammation and tissue remodeling, were upregulated in M2a compared to RM. CONCLUSION: Our data in in vitro polarized human macrophages: 1. confirm and extend known inflammatory and anti-inflammatory gene expression patterns; 2. demonstrate changes in mitochondrial metabolism associated to insulin resistance and insulin sensitivity in M1 and M2a, respectively; 3. highlight the potential relevance of gene expression timing in M1 function; 4. unveil enhanced expression of Wnt pathways in M2a suggesting a potential dual (pro-inflammatory and anti-inflammatory) role of M2a in inflammatory diseases.


Subject(s)
Biomarkers/metabolism , Cytokines/metabolism , Gene Expression Profiling , Macrophage Activation , Macrophages/metabolism , Cell Differentiation , Cells, Cultured , Cytokines/genetics , Humans , Macrophages/cytology , Oligonucleotide Array Sequence Analysis
17.
PLoS One ; 10(3): e0119310, 2015.
Article in English | MEDLINE | ID: mdl-25803285

ABSTRACT

BACKGROUND AND AIM: Sparse evidence suggests a possible link between exposure to airborne nanoparticles (NPs) and cardiovascular (CV) risk, perhaps through mechanisms involving oxidative stress and inflammation. We assessed the effects of TiO2 and Co3O4 NPs in human circulating angiogenic cells (CACs), which take part in vascular endothelium repair/replacement. METHODS: CACs were isolated from healthy donors' buffy coats after culturing lymphomonocytes on fibronectin-coated dishes in endothelial medium for 7 days. CACs were pre-incubated with increasing concentration of TiO2 and Co3O4 (from 1 to 100 µg/ml) to test the effects of NP ­ characterized by Transmission Electron Microscopy ­ on CAC viability, apoptosis (caspase 3/7 activation), function (fibronectin adhesion assay), oxidative stress and inflammatory cytokine gene expression. RESULTS: Neither oxidative stress nor cell death were associated with exposure to TiO2 NP (except at the highest concentration tested), which, however, induced a higher pro-inflammatory effect compared to Co3O4 NPs (p<0.01). Exposure to Co3O4 NPs significantly reduced cell viability (p<0.01) and increased caspase activity (p<0.01), lipid peroxidation end-products (p<0.05) and pro-inflammatory cytokine gene expression (p<0.05 or lower). Notably, CAC functional activity was impaired after exposure to both TiO2 (p<0.05 or lower) and Co3O4 (p<0.01) NPs. CONCLUSIONS: In vitro exposure to TiO2 and Co3O4 NPs exerts detrimental effects on CAC viability and function, possibly mediated by accelerated apoptosis, increased oxidant stress (Co3O4 NPs only) and enhancement of inflammatory pathways (both TiO2 and Co3O4 NPs). Such adverse effects may be relevant for a potential role of exposure to TiO2 and Co3O4 NPs in enhancing CV risk in humans.


Subject(s)
Apoptosis/drug effects , Cobalt/pharmacology , Leukocytes, Mononuclear/drug effects , Neovascularization, Physiologic/drug effects , Oxidative Stress/drug effects , Oxides/pharmacology , Titanium/pharmacology , Cobalt/chemistry , Cobalt/toxicity , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Humans , Leukocytes, Mononuclear/cytology , Metal Nanoparticles/administration & dosage , Metal Nanoparticles/chemistry , Metal Nanoparticles/toxicity , Microscopy, Electron, Transmission , Oxides/chemistry , Oxides/toxicity , Primary Cell Culture , Titanium/chemistry , Titanium/toxicity
18.
Food Funct ; 5(8): 1881-90, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24942553

ABSTRACT

BACKGROUND AND AIMS: Recent data suggest that n-3 PUFA exert beneficial effects on endothelial progenitor cell (EPC) biology. We sought to investigate whether these effects might be mediated by enhanced EPC in vitro function and/or in vivo bioavailability. METHODS AND RESULTS: CACs and late-outgrowth EPCs were isolated from peripheral blood mononuclear cells obtained from 12 donor buffy-coats. The effect of n-3 PUFA (EPA:DHA = 0.9:1.5; 9 µM EPA plus 15 µM DHA) was tested on CAC/EPC viability, function (tube-formation) and pro-inflammatory molecule expression. Circulating EPC (cells positive for CD34, CD133 and kinase insert domain receptor - KDR cell-surface antigens by flow cytometry) number was evaluated in 20 healthy subjects (10 F/10 M, 32 ± 5 years), randomized to receive 4 mackerel or sardine portions per week for 6 weeks followed by a 6 week free-diet period. N-3 PUFA improved CAC and late-outgrowth EPC viability (p < 0.05) and the capacity to form tube-like structures in CACs (+38%; p < 0.05) and late-outgrowth EPCs (+15%; p < 0.05). ICAM-1 expression was reduced in both CACs (p < 0.05) and late-outgrowth EPCs (p < 0.05) and VCAM-1 in late-outgrowth EPCs (p < 0.005). N-3 PUFA significantly decreased TNF-α and MCP-1 expression in CACs and IL-8, TNF-α and MCP-1 in late-outgrowth EPCs (p < 0.05). Circulating EPC number significantly improved after 6 weeks of a fish-enriched diet (p < 0.01) and returned to baseline levels after a 6 week free-diet period (p < 0.01). Plasma EPA levels were independently and positively associated with EPC levels (p < 0.005). CONCLUSION: Our findings support the case of a beneficiary role played by n-3 PUFA in EPC function and bioavailability.


Subject(s)
Docosahexaenoic Acids/blood , Eicosapentaenoic Acid/blood , Endothelial Progenitor Cells/drug effects , Adult , Animals , Apoptosis/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Chemokine CCL2/genetics , Chemokine CCL2/metabolism , Endothelial Progenitor Cells/metabolism , Female , Fishes , Humans , Intercellular Adhesion Molecule-1/genetics , Intercellular Adhesion Molecule-1/metabolism , Interleukin-8/genetics , Interleukin-8/metabolism , Leukocytes, Mononuclear/metabolism , Male , NF-kappa B/genetics , NF-kappa B/metabolism , Seafood , Tumor Necrosis Factor-alpha/metabolism , Vascular Cell Adhesion Molecule-1/genetics , Vascular Cell Adhesion Molecule-1/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism
19.
PLoS One ; 7(11): e48283, 2012.
Article in English | MEDLINE | ID: mdl-23139771

ABSTRACT

BACKGROUND: Evidence suggests that the PPARγ-agonist insulin sensitizer pioglitazone, may provide potential beneficial cardiovascular (CV) effects beyond its anti-hyperglycaemic function. A reduced endothelial progenitor cell (EPC) number is associated with impaired glucose tolerance (IGT) or diabetes, conditions characterised by increased CV risk. AIM: To evaluate whether pioglitazone can provide benefit in vitro in EPCs obtained from IGT subjects. MATERIALS AND METHODS: Early and late-outgrowth EPCs were obtained from peripheral blood mononuclear cells of 14 IGT subjects. The in vitro effect of pioglitazone (10 µM) with/without PPARγ-antagonist GW9662 (1 µM) was assessed on EPC viability, apoptosis, ability to form tubular-like structures and pro-inflammatory molecule expression. RESULTS: Pioglitazone increased early and late-outgrowth EPC viability, with negligible effects on apoptosis. The capacity of EPCs to form tubular-like structures was improved by pioglitazone in early (mean increase 28%; p=0.005) and late-outgrowth (mean increase 30%; p=0.037) EPCs. Pioglitazone reduced ICAM-1 and VCAM-1 adhesion molecule expression in both early (p=0.001 and p=0.012 respectively) and late-outgrowth (p=0.047 and p=0.048, respectively) EPCs. Similarly, pioglitazone reduced TNFα gene and protein expression in both early (p=0.034;p=0.022) and late-outgrowth (p=0.026;p=0.017) EPCs compared to control. These effects were prevented by incubation with the PPARγ-antagonist GW9662. CONCLUSION: Pioglitazone exerts beneficial effects in vitro on EPCs isolated from IGT subjects, supporting the potential implication of pioglitazone as a CV protective agents.


Subject(s)
Endothelial Cells/pathology , Glucose Intolerance/pathology , Glucose Intolerance/physiopathology , Stem Cells/pathology , Thiazolidinediones/pharmacology , Apoptosis/drug effects , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/metabolism , Cell Survival/drug effects , Chemokines/genetics , Chemokines/metabolism , Endothelial Cells/drug effects , Female , Gene Expression Regulation/drug effects , Glucose Intolerance/drug therapy , Glucose Intolerance/genetics , Humans , Inflammation Mediators/metabolism , Male , Middle Aged , NF-kappa B/genetics , NF-kappa B/metabolism , Pioglitazone , Stem Cells/drug effects , Thiazolidinediones/therapeutic use
20.
G Ital Cardiol (Rome) ; 11(10 Suppl 1): 93S-97S, 2010 Oct.
Article in Italian | MEDLINE | ID: mdl-21416835

ABSTRACT

Insulin resistance is associated with a cluster of metabolic and hemodynamic abnormalities that lead to increased cardiovascular morbidity and mortality. In this review the main pathophysiological mechanisms and metabolic consequences of insulin resistance are summarized. The correlation between insulin resistance and cardiovascular disease and the practical utility of the concept of metabolic syndrome as a diagnostic tool are also discussed.


Subject(s)
Cardiovascular Diseases/epidemiology , Diabetes Complications , Insulin Resistance , Metabolic Syndrome , Adipose Tissue/metabolism , Cardiovascular Diseases/etiology , Cardiovascular Diseases/metabolism , Cardiovascular Diseases/mortality , Diabetes Mellitus, Type 2/epidemiology , Female , Follow-Up Studies , Humans , In Vitro Techniques , Liver/metabolism , Longitudinal Studies , Male , Meta-Analysis as Topic , Obesity/metabolism , Randomized Controlled Trials as Topic , Risk Factors , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...