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.
Aging (Albany NY) ; 12(11): 10180-10193, 2020 05 26.
Article in English | MEDLINE | ID: mdl-32457253

ABSTRACT

Aging is associated with impaired neovascularization in response to ischemia. MicroRNAs are small noncoding RNAs emerging as key regulators of physiological and pathological processes. Here we investigated the potential role of microRNAs in endothelial cell senescence and age-dependent impairment of neovascularization. Next generation sequencing and qRT-PCR analyses identified miR-130a as a pro-angiogenic microRNA which expression is significantly reduced in old mouse aortic endothelial cells (ECs). Transfection of young ECs with a miR-130a inhibitor leads to accelerated senescence and reduced angiogenic functions. Conversely, forced expression of miR-130a in old ECs reduces senescence and improves angiogenesis. In a mouse model of hindlimb ischemia, intramuscular injection of miR-130a mimic in older mice restores blood flow recovery and vascular densities in ischemic muscles, improves mobility and reduces tissue damage. miR-130a directly targets antiangiogenic homeobox genes MEOX2 and HOXA5. MEOX2 and HOXA5 are significantly increased in the ischemic muscles of aging mice, but forced expression of miR-130a reduces the expression of these factors. miR-130a treatment after ischemia is also associated with increased number and improved functional activities of pro-angiogenic cells (PACs). Forced expression of miR-130a could constitute a novel strategy to improve blood flow recovery and reduce ischemia in older patients with ischemic vascular diseases.


Subject(s)
Aging/genetics , Cellular Senescence/genetics , Endothelium, Vascular/pathology , Ischemia/pathology , MicroRNAs/metabolism , Neovascularization, Physiologic/genetics , Age Factors , Aged , Animals , Aorta/cytology , Cell Movement/drug effects , Cell Movement/genetics , Cellular Senescence/drug effects , Disease Models, Animal , Endothelial Cells/drug effects , Endothelial Cells/pathology , Endothelium, Vascular/cytology , Hindlimb/blood supply , Homeodomain Proteins/genetics , Human Umbilical Vein Endothelial Cells , Humans , Mice , MicroRNAs/agonists , MicroRNAs/antagonists & inhibitors , Neovascularization, Physiologic/drug effects , Primary Cell Culture , Transcription Factors/genetics , Young Adult
2.
Atherosclerosis ; 289: 73-84, 2019 10.
Article in English | MEDLINE | ID: mdl-31479774

ABSTRACT

BACKGROUND AND AIMS: MicroRNA (miR)-146 is a key regulator of inflammation, endothelial activation and atherosclerosis. This study sought to define its potential role for the modulation of ischemia-induced neovascularization in atherosclerotic conditions. METHODS: Next generation sequencing and qRT-PCR analyses were used to compare microRNA expression in the ischemic muscles of hypercholesterolemic ApoE-deficient (ApoE-/-) mice vs. wild type mice, and in HUVECs exposed or not to oxLDL. Neovascularization was investigated in a mouse model of hindlimb ischemia and the functional activities of HUVECs and pro-angiogenic cells (PACs) were assessed in vitro. RESULTS: We found that miR-146b (but not miR-146a) is significantly reduced in the ischemic muscles of ApoE-/- mice, and in HUVECs exposed to oxLDL. Inhibition of miR-146b reduces angiogenesis in vitro, whereas forced expression of miR-146b rescues oxLDL-mediated impairment of endothelial cell proliferation and tube formation. Mechanistically, miR146b directly targets tumor necrosis factor-alpha (TNFa) Receptor Associated Factor 6 (TRAF6) to inhibit inflammation. We found that hypercholesterolemia and oxLDL exposure are associated with higher levels of TRAF6, and increased expression of TNFa. However, forced expression of miR-146b in high cholesterol conditions reduces the expression of these inflammatory factors. In vivo, intramuscular injection of miR-146b mimic reduces ischemic damages and restores blood flow recuperation and capillary density in the ischemic muscles of ApoE-/- mice. Treatment with miR-146b also increases the number and functional activities of pro-angiogenic cells (PACs). CONCLUSIONS: Hypercholesterolemia is associated with reduced expression of miR-146b, which increases TRAF6-dependent inflammation and is associated with poor neovascularization in response to ischemia. Forced expression of miR-146b using a miR mimic could constitute a novel therapeutic strategy to improve ischemia-induced neovascularization in atherosclerotic conditions.


Subject(s)
Hypercholesterolemia/metabolism , Inflammation/metabolism , MicroRNAs/genetics , Neovascularization, Pathologic/metabolism , TNF Receptor-Associated Factor 6/genetics , Animals , Blood Flow Velocity , Cell Movement , Cell Proliferation , Hindlimb/metabolism , Human Umbilical Vein Endothelial Cells , Humans , Intracellular Signaling Peptides and Proteins/genetics , Ischemia/physiopathology , Lipoproteins, LDL/metabolism , Mice , Mice, Knockout, ApoE , MicroRNAs/metabolism , Reactive Oxygen Species/metabolism , Sequence Analysis, RNA , THP-1 Cells
3.
Sci Rep ; 7(1): 14143, 2017 10 26.
Article in English | MEDLINE | ID: mdl-29075011

ABSTRACT

Adverse perinatal conditions can lead to developmental programming of cardiovascular diseases. Prematurely born infants are often exposed to high oxygen levels, which in animal models has been associated with endothelial dysfunction, hypertension, and cardiac remodeling during adulthood. Here we found that adult mice that have been transiently exposed to O2 after birth show defective neovasculariation after hindlimb ischemia, as demonstrated by impaired blood flow recovery, reduced vascular density in ischemic muscles and increased tissue damages. Ischemic muscles isolated from mice exposed to O2 after birth exhibit increased oxidative stress levels and reduced expression of superoxide dismutase 1 (SOD1) and vascular endothelial growth factor (VEGF). Pro-angiogenic cells (PACs) have been shown to have an important role for postnatal neovascularisation. We found that neonatal exposure to O2 is associated with reduced number of PACs in adults. Moreover, the angiogenic activities of both PACs and mature mouse aortic endothelial cells (MAECs) are significantly impaired in mice exposed to hyperoxia after birth. Our results indicate that neonatal exposure to high oxygen levels leads to impaired ischemia-induced neovascularization during adulthood. The mechanism involves deleterious effects on oxidative stress levels and angiogenic signals in ischemic muscles, together with dysfunctional activities of PACs and mature endothelial cells.


Subject(s)
Hindlimb/blood supply , Hyperoxia/physiopathology , Ischemia/physiopathology , Neovascularization, Physiologic/physiology , Animals , Animals, Newborn , Cell Adhesion , Disease Models, Animal , Endothelial Cells/cytology , Endothelial Cells/physiology , Female , Human Umbilical Vein Endothelial Cells , Humans , Mice, Inbred C57BL , Oxygen/adverse effects , Regional Blood Flow , Vascular Endothelial Growth Factor A/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...