Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Methods Mol Biol ; 2235: 139-153, 2021.
Article in English | MEDLINE | ID: mdl-33576975

ABSTRACT

MicroRNAs (miRNAs) are expressed in all cell types, including pericytes, and play essential roles in vascular development, homeostasis, and disease. Manipulation of pericytes with miRNA mimics and inhibitors represents an essential tool to study the role of pericytes in vascular development and regeneration and to better understand the therapeutic potential of miRNA manipulation in pericytes. Here we describe methods for manipulating pericyte function by using miRNA mimics and inhibitors. We also describe methods to assess pericyte function (proliferation and migration) after manipulation with miRNAs and explain how miRNA gene targets can be identified and validated in pericytes after manipulation with miRNA.


Subject(s)
Cloning, Molecular/methods , MicroRNAs/genetics , Pericytes/metabolism , Animals , Gene Expression Regulation/genetics , Humans , MicroRNAs/physiology , Pericytes/physiology , Transfection/methods , Transformation, Genetic/genetics
2.
JCI Insight ; 5(14)2020 07 23.
Article in English | MEDLINE | ID: mdl-32544097

ABSTRACT

Following myocardial infarction (MI), the adult heart has minimal regenerative potential. Conversely, the neonatal heart can undergo extensive regeneration, and neovascularization capacity was hypothesized to contribute to this difference. Here, we demonstrate the higher angiogenic potential of neonatal compared with adult mouse cardiac endothelial cells (MCECs) in vitro and use this difference to identify candidate microRNAs (miRs) regulating cardiac angiogenesis after MI. miR expression profiling revealed miR-96 and miR-183 upregulation in adult compared with neonatal MCECs. Their overexpression decreased the angiogenic potential of neonatal MCECs in vitro and prevented scar resolution and neovascularization in neonatal mice after MI. Inversely, their inhibition improved the angiogenic potential of adult MCECs, and miR-96/miR-183-KO mice had increased peri-infarct neovascularization. In silico analyses identified anillin (ANLN) as a direct target of miR-96 and miR-183. In agreement, Anln expression declined following their overexpression and increased after their inhibition in vitro. Moreover, ANLN expression inversely correlated with miR-96 expression and age in cardiac ECs of cardiovascular patients. In vivo, ANLN+ vessels were enriched in the peri-infarct area of miR-96/miR-183-KO mice. These findings identify miR-96 and miR-183 as regulators of neovascularization following MI and miR-regulated genes, such as anillin, as potential therapeutic targets for cardiovascular disease.


Subject(s)
MicroRNAs/genetics , Microfilament Proteins/genetics , Myocardial Infarction/genetics , Animals , Cell Proliferation/genetics , Disease Models, Animal , Endothelial Cells/metabolism , Gene Expression Regulation/genetics , Heart/growth & development , Heart/physiopathology , Humans , Mice , Mice, Knockout , Myocardial Infarction/drug therapy , Myocardial Infarction/pathology , Neovascularization, Physiologic/genetics
3.
Ageing Res Rev ; 35: 301-311, 2017 May.
Article in English | MEDLINE | ID: mdl-27829173

ABSTRACT

Sirtuin 6 (SIRT6) is a member of the sirtuin family NAD+-dependent deacetylases with multiple roles in controlling organism homeostasis, lifespan, and diseases. Due to its complex and opposite functional roles, this sirtuin is considered a two-edged sword in health and disease. Indeed, SIRT6 improves longevity, similarly to the founding yeast member, silent information regulator-2 (Sir2), and modulates genome stability, telomere integrity, transcription, and DNA repair. Its deficiency is associated with chronic inflammation, diabetes, cardiac hypertrophy, obesity, liver dysfunction, muscle/adipocyte disorders, and cancer. Besides, pieces of evidence showed that SIRT6 is a promoter of specific oncogenic pathways, thus disclosing its dual role regarding cancer development. Collectively, these findings suggest that multiple mechanisms, to date not entirely known, underlie the intriguing roles of SIRT6. Here we provide an overview of the current molecular mechanisms through which SIRT6 controls cancer and heart diseases, and describe its recent implications in the atherosclerotic plaque development.


Subject(s)
Cardiovascular Diseases/metabolism , Homeostasis/physiology , Longevity/physiology , Neoplasms/metabolism , Sirtuins/metabolism , DNA Repair/physiology , Humans , Telomere Homeostasis/physiology
4.
Theriogenology ; 88: 236-243, 2017 Jan 15.
Article in English | MEDLINE | ID: mdl-27769574

ABSTRACT

The aim of this study was to evaluate the effect of carnitine supplementation of semen extender on fertility parameters of frozen-thawed buffalo sperm. Buffalo semen was cryopreserved in BioXcell containing 0 (control group), 2.5 and 7.5-mM carnitine. After thawing, viability, motility, membrane integrity and capacitation status (assessed by localization of phosphotyrosine-containing proteins and chlortetracycline, chlortetracycline assay) were evaluated. Furthermore, total antioxidant capacity, reactive oxygen species (ROS) and lipid peroxidation levels, as well as adenosine triphosphate (ATP) content and phospholipids concentration were assessed. Finally, in vitro-fertilizing ability was evaluated after heterologous IVF. An increased post-thawing sperm motility and membrane integrity were recorded in both treated groups compared with the control (44.4 ± 3.5, 53.1 ± 3.9, and 52.5 ± 3.6%; P < 0.05 and 48.44 ± 0.69, 55.19 ± 0.54, 59.63 ± 0.30%; P < 0.01 with 0, 2.5-mM, and 7.5-mM carnitine, respectively). Supplementation of carnitine to the freezing extender decreased (P < 0.01) the percentage of sperm displaying fluorescence at both equatorial and anterior acrosomal regions (pattern EA), corresponding to high capacitation level, compared with the control (30.3 ± 3.8, 18.8 ± 2.8, and 7.2 ± 2.9%, respectively, with 0, 2.5-mM, and 7.5-mM carnitine). In agreement with this, carnitine also decreased (P < 0.01) the percentage of sperm displaying chlortetracycline pattern B (capacitated sperm) (63.8 ± 1.8, 46.8 ± 2.2, and 37.2 ± 1.8%, respectively with 0, 2.5-, and 7.5-mM carnitine). Interestingly, carnitine increased total antioxidant capacity and ATP content of buffalo frozen-thawed sperm (1.32 ± 0.02, 1.34 ± 0.01, 1.37 ± 0.01 mM/L and 4.1 ± 0.1, 5.3 ± 0.1 and 8.2 ± 0.4 nM × 108 sperm; P < 0.01, respectively, with 0, 2.5- and 7.5-mM carnitine). Intracellular ROS decreased in carnitine-treated sperm compared with the control, as indicated by dihydroethidium (DHE) values (0.22 ± 0.01, 0.18 ± 0.01, and 0.14 ± 0.0 µM/100 µL dihydroethidium, respectively, with 0, 2.5-, and 7.5-mM carnitine; P < 0.01), whereas lipid peroxidation levels (on average 30.5 ± 0.3 nmol/mL MDA) and phospholipids concentration (on average 0.14 ± 0.00 µg/120 × 106 sperm) were unaffected. Despite the improved sperm quality, the percentage of normospermic penetration after IVF was not influenced (on average 53.5 ± 1.8). In conclusion, enrichment of extender with carnitine improved buffalo sperm quality by increasing ATP generation and modulating ROS production, without affecting in vitro fertilizing ability.


Subject(s)
Buffaloes , Carnitine/pharmacology , Freezing , Semen Preservation/veterinary , Sperm Capacitation/physiology , Spermatozoa/drug effects , Animals , Cell Survival , Cryopreservation/veterinary , Male , Oxidative Stress , Sperm Motility/drug effects , Spermatozoa/physiology
5.
J Diabetes Complications ; 31(2): 295-303, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27771154

ABSTRACT

AIMS: Glucagon like peptide 1 (GLP-1) analogues and dipeptidyl peptidase IV (DPP-4) inhibitors reduce atherosclerosis progression in type 2 diabetes mellitus (T2DM) patients and are associated with morphological and compositional characteristics of stable plaque phenotype. GLP-1 promotes the secretion of adiponectin which exerts anti-inflammatory effects through the adaptor protein PH domain and leucine zipper containing 1 (APPL1). The potential role of APPL1 expression in the evolution of atherosclerotic plaque in TDM2 patients has not previously evaluated. METHODS: The effect of incretin therapy in the regulation of adiponectin/APPL1 signaling was evaluated both on carotid plaques of asymptomatic diabetic (n=71) and non-diabetic patients (n=52), and through in vitro experiments on endothelial cell (EC). RESULTS: Atherosclerotic plaques of T2DM patients showed lower adiponectin and APPL1 levels compared with non-diabetic patients, along with higher oxidative stress, tumor necrosis factor-α (TNF-α), vimentin, and matrix metalloproteinase-9 (MMP-9) levels. Among T2DM subjects, current incretin-users presented higher APPL1 and adiponectin content compared with never incretin-users. Similarly, in vitro observations on endothelial cells co-treated with high-glucose (25mM) and GLP-1 (100nM) showed a greater APPL1 protein expression compared with high-glucose treatment alone. CONCLUSIONS: Our findings suggest a potential role of adiponectin/APPL1 signaling in mediating the effect of incretin in the prevention of atherosclerosis progression or plaque vulnerability in T2DM.


Subject(s)
Adaptor Proteins, Signal Transducing/agonists , Adiponectin/metabolism , Diabetes Mellitus, Type 2/drug therapy , Diabetic Angiopathies/prevention & control , Incretins/therapeutic use , Plaque, Atherosclerotic/prevention & control , Signal Transduction/drug effects , Adaptor Proteins, Signal Transducing/metabolism , Aged , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Antioxidants/pharmacology , Antioxidants/therapeutic use , Carotid Stenosis/complications , Carotid Stenosis/epidemiology , Carotid Stenosis/prevention & control , Carotid Stenosis/surgery , Cells, Cultured , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/pathology , Diabetic Angiopathies/epidemiology , Diabetic Angiopathies/pathology , Diabetic Angiopathies/surgery , Endarterectomy, Carotid , Endothelium, Vascular/drug effects , Endothelium, Vascular/immunology , Endothelium, Vascular/metabolism , Endothelium, Vascular/pathology , Female , Glucagon-Like Peptide 1/metabolism , Humans , Incretins/pharmacology , Italy/epidemiology , Male , Oxidative Stress/drug effects , Plaque, Atherosclerotic/complications , Plaque, Atherosclerotic/epidemiology , Plaque, Atherosclerotic/pathology , Risk Factors , Secondary Prevention
6.
Free Radic Biol Med ; 96: 211-22, 2016 07.
Article in English | MEDLINE | ID: mdl-27101740

ABSTRACT

Ergothioneine (Egt), the betaine of 2-mercapto-L-histidine, is a dietary antioxidant protecting against many diseases, including cardiovascular disease (CVD), through a redox mechanism different from alkylthiols. Here, experiments were designed to evaluate the mechanisms underlying the beneficial effect of Egt against hyperglycaemia-induced senescence in endothelial cells. To this end, cells were incubated with increasing concentrations of Egt (0.01-1.00mM) for 12h followed by incubation for 48h with high-glucose (25mM). Cell evaluation indicated that viability was not affected by mM concentrations of Egt and that the high-glucose cytotoxicity was prevented with the highest efficacy at 0.5mM Egt. The cytoprotective effect of Egt was paralleled by reduced ROS production, cell senescence, and, interestingly, the formation of hercynine (EH), a betaine we recently found to be produced during the Egt oxidation pathway. Notably, the Egt beneficial effect was exerted through the upregulation of sirtuin 1 (SIRT1) and sirtuin 6 (SIRT6) expression and the downregulation of p66Shc and NF-κB. SIRT1 activity inhibition and SIRT6 gene silencing by small interfering RNA abolished the protective effect of Egt against the high-glucose-induced endothelial senescence. These data provide the first evidence of the Egt ability to interfere with endothelial senescence linked to hyperglycaemia through the regulation of SIRT1 and SIRT6 signaling, thus further strengthening the already assessed role of these two histone deacetylases in type 2 diabetes.


Subject(s)
Diabetes Mellitus, Type 2/drug therapy , Ergothioneine/administration & dosage , Hyperglycemia/drug therapy , Sirtuin 1/genetics , Sirtuins/genetics , Antioxidants/metabolism , Betaine/analogs & derivatives , Betaine/metabolism , Cellular Senescence/drug effects , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/metabolism , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Gene Expression Regulation/drug effects , Glucose/metabolism , Histidine/analogs & derivatives , Histidine/metabolism , Humans , Hyperglycemia/genetics , Hyperglycemia/metabolism , Oxidative Stress/drug effects , Src Homology 2 Domain-Containing, Transforming Protein 1/genetics
8.
Biochim Biophys Acta ; 1852(7): 1311-22, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25766107

ABSTRACT

Silent information regulator-2 (Sir-2) proteins, or sirtuins, are a highly conserved protein family of histone deacetylases that promote longevity by mediating many of the beneficial effects of calorie restriction which extends life span and reduces the incidence of cancer, cardiovascular disease (CVD), and diabetes. Here, we review the role of sirtuins (SIRT1-7) in vascular homeostasis and diseases by providing an update on the latest knowledge about their roles in endothelial damage and vascular repair mechanisms. Among all sirtuins, in the light of the numerous functions reported on SIRT1 in the vascular system, herein we discuss its roles not only in the control of endothelial cells (EC) functionality but also in other cell types beyond EC, including endothelial progenitor cells (EPC), smooth muscle cells (SMC), and immune cells. Furthermore, we also provide an update on the growing field of compounds under clinical evaluation for the modulation of SIRT1 which, at the state of the art, represents the most promising target for the development of novel drugs against CVD, especially when concomitant with type 2 diabetes.


Subject(s)
Sirtuins/metabolism , Vascular Diseases/metabolism , Animals , Humans , Signal Transduction , Sirtuins/classification , Sirtuins/genetics , Vascular Diseases/drug therapy , Vascular Diseases/enzymology
SELECTION OF CITATIONS
SEARCH DETAIL
...