Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 3 de 3
Filter
Add more filters










Database
Language
Publication year range
1.
Sci Rep ; 7: 44584, 2017 03 15.
Article in English | MEDLINE | ID: mdl-28294194

ABSTRACT

Diabetic Nephropathy (DN) is the leading cause of end-stage renal disease. Preclinical and experimental studies show that PDE5 inhibitors (PDE5is) exert protective effects in DN improving perivascular inflammation. Using a mouse model of diabetic kidney injury we investigated the protective proprieties of PDE5is on renal hemodynamics and the molecular mechanisms involved. PDE5i treatment prevented the development of DN-related hypertension (P < 0.001), the increase of urine albumin creatinine ratio (P < 0.01), the fall in glomerular filtration rate (P < 0.001), and improved renal resistive index (P < 0.001) and kidney microcirculation. Moreover PDE5i attenuated the rise of nephropathy biomarkers, soluble urokinase-type plasminogen activator receptor, suPAR and neutrophil gelatinase-associated lipocalin, NGAL. In treated animals, blood vessel perfusion was improved and vascular leakage reduced, suggesting preserved renal endothelium integrity, as confirmed by higher capillary density, number of CD31+ cells and pericyte coverage. Analysis of the mechanisms involved revealed the induction of bone morphogenetic protein-7 (BMP7) expression, a critical regulator of angiogenesis and kidney homeostasis, through a PDE5i-dependent downregulation of miR-22. In conclusion PDE5i slows the progression of DN in mice, improving hemodynamic parameters and vessel integrity. Regulation of miR-22/BMP7, an unknown mechanism of PDE5is in nephrovascular protection, might represent a novel therapeutic option for treatment of diabetic complications.


Subject(s)
Bone Morphogenetic Protein 7/genetics , Cyclic Nucleotide Phosphodiesterases, Type 5/genetics , Diabetic Nephropathies/genetics , MicroRNAs/genetics , Phosphodiesterase 5 Inhibitors/administration & dosage , Albumins/metabolism , Animals , Biomarkers/blood , Creatinine/urine , Cyclic Nucleotide Phosphodiesterases, Type 5/drug effects , Diabetic Nephropathies/complications , Diabetic Nephropathies/pathology , Gene Expression Regulation/drug effects , Glomerular Filtration Rate/drug effects , Hemodynamics/drug effects , Humans , Hypertension/complications , Hypertension/drug therapy , Hypertension/genetics , Hypertension/pathology , Kidney/drug effects , Kidney/pathology , Kidney Failure, Chronic/drug therapy , Kidney Failure, Chronic/genetics , Kidney Failure, Chronic/pathology , Male , Mice , Receptors, Urokinase Plasminogen Activator/genetics
2.
J Clin Endocrinol Metab ; 101(4): 1525-34, 2016 04.
Article in English | MEDLINE | ID: mdl-26964730

ABSTRACT

CONTEXT: Visceral adiposity plays a significant role in cardiovascular risk. PDE5 inhibitors (PDE5i) can improve cardiac function and insulin sensitivity in type 2 diabetes patients. OBJECTIVE: To investigate whether PDE5i affect visceral adipose tissue (VAT), specifically epicardial fat (epicardial adipose tissue [EAT]), and what mechanism is involved, using microarray-based profiling of pharmacologically modulated microRNA (miRNAs). DESIGN: Randomized, double-blind, placebo-controlled study in type 2 diabetes. PATIENTS AND INTERVENTION: A total of 59 diabetic patients were randomized to receive 100-mg/d sildenafil or placebo for 12 weeks. Fat biopsies were collected in a subgroup of patients. In a parallel protocol, db/db mice were randomized to 12 weeks of sildenafil or vehicle, and VAT was collected. MAIN OUTCOME AND MEASURES: Anthropometric and metabolic parameters, EAT quantification through cardiac magnetic resonance imaging, array of 2005 circulating miRNAs, quantitative PCR, and flow cytometry of VAT. RESULTS: Compared with placebo, sildenafil reduced waist circumference (P = .024) and EAT (P = .045). Microarray analysis identified some miRNAs differentially regulated by sildenafil, including down-regulation of miR-22-3p, confirmed by real-time quantitative PCR (P < .001). Sildenafil's modulation of miR-22-3p expression was confirmed in vitro in HL1 cardiomyocytes. Up-regulation of SIRT1, a known target of miR-22-3p, was found in both serum and sc fat in sildenafil-treated subjects. Compared with vehicle, 12-week sildenafil treatment down-regulated miR-22-3p and up-regulated Sirtuin1 (SIRT1) gene expression in VAT from db/db mice, shifting adipose tissue cell composition toward a less inflamed profile. CONCLUSIONS: Treatment with PDE5i in humans and murine models of diabetes improves VAT, targeting SIRT1 through a modulation of miR-22-3p expression.


Subject(s)
Adiposity/drug effects , Cyclic Nucleotide Phosphodiesterases, Type 5/chemistry , Diabetes Mellitus, Type 2/physiopathology , MicroRNAs/genetics , Obesity, Abdominal/drug therapy , Phosphodiesterase 5 Inhibitors/pharmacology , Sildenafil Citrate/pharmacology , Sirtuin 1/metabolism , Adiposity/genetics , Adult , Aged , Animals , Double-Blind Method , Flow Cytometry , Humans , Male , Mice , Mice, Obese , Middle Aged , Obesity, Abdominal/metabolism , Obesity, Abdominal/pathology , Real-Time Polymerase Chain Reaction , Sirtuin 1/genetics
3.
PLoS One ; 10(5): e0126580, 2015.
Article in English | MEDLINE | ID: mdl-25961566

ABSTRACT

Diabetes mellitus is characterized by changes in endothelial cells that alter monocyte recruitment, increase classic (M1-type) tissue macrophage infiltration and lead to self-sustained inflammation. Our and other groups recently showed that chronic inhibition of phosphodiesterase-5 (PDE5i) affects circulating cytokine levels in patients with diabetes; whether PDE5i also affects circulating monocytes and tissue inflammatory cell infiltration remains to be established. Using murine streptozotocin (STZ)-induced diabetes and in human vitro cell-cell adhesion models we show that chronic hyperglycemia induces changes in myeloid and endothelial cells that alter monocyte recruitment and lead to self-sustained inflammation. Continuous PDE5i with sildenafil (SILD) expanded tissue anti-inflammatory TIE2-expressing monocytes (TEMs), which are known to limit inflammation and promote tissue repair. Specifically, SILD: 1) normalizes the frequency of circulating pro-inflammatory monocytes triggered by hyperglycemia (53.7 ± 7.9% of CD11b+Gr-1+ cells in STZ vs. 30.4 ± 8.3% in STZ+SILD and 27.1 ± 1.6% in CTRL, P<0.01); 2) prevents STZ-induced tissue inflammatory infiltration (4-fold increase in F4/80+ macrophages in diabetic vs. control mice) by increasing renal and heart anti-inflammatory TEMs (30.9 ± 3.6% in STZ+SILD vs. 6.9 ± 2.7% in STZ, P <0.01, and 11.6 ± 2.9% in CTRL mice); 3) reduces vascular inflammatory proteins (iNOS, COX2, VCAM-1) promoting tissue protection; 4) lowers monocyte adhesion to human endothelial cells in vitro through the TIE2 receptor. All these changes occurred independently from changes of glycemic status. In summary, we demonstrate that circulating renal and cardiac TEMs are defective in chronic hyperglycemia and that SILD normalizes their levels by facilitating the shift from classic (M1-like) to alternative (M2-like)/TEM macrophage polarization. Restoration of tissue TEMs with PDE5i could represent an additional pharmacological tool to prevent end-organ diabetic complications.


Subject(s)
Cyclic Nucleotide Phosphodiesterases, Type 5/genetics , Diabetes Mellitus, Experimental/drug therapy , Hyperglycemia/drug therapy , Phosphodiesterase 5 Inhibitors/pharmacology , Protective Agents/pharmacology , Sildenafil Citrate/pharmacology , Animals , Cell Adhesion/drug effects , Cell Communication/drug effects , Cell Count , Coculture Techniques , Cyclic Nucleotide Phosphodiesterases, Type 5/metabolism , Cyclooxygenase 2/genetics , Cyclooxygenase 2/metabolism , Diabetes Mellitus, Experimental/chemically induced , Diabetes Mellitus, Experimental/enzymology , Diabetes Mellitus, Experimental/pathology , Endothelial Cells/drug effects , Endothelial Cells/enzymology , Endothelial Cells/pathology , Gene Expression Regulation , Hyperglycemia/chemically induced , Hyperglycemia/enzymology , Hyperglycemia/pathology , Macrophages/drug effects , Macrophages/enzymology , Macrophages/pathology , Male , Mice , Monocytes/drug effects , Monocytes/enzymology , Monocytes/pathology , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/metabolism , Receptor, TIE-2/genetics , Receptor, TIE-2/metabolism , Signal Transduction , Streptozocin , Vascular Cell Adhesion Molecule-1/genetics , Vascular Cell Adhesion Molecule-1/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...