Imperial College London

Professor Costanza Emanueli

Faculty of MedicineNational Heart & Lung Institute

Chair in Cardiovascular Science
 
 
 
//

Contact

 

c.emanueli Website

 
 
//

Location

 

434ICTEM buildingHammersmith Campus

//

Summary

 

Publications

Citation

BibTex format

@article{Besnier:2018:10.1016/j.ymthe.2018.06.003,
author = {Besnier, M and Gasparino, S and Vono, R and Sangalli, E and Facoetti, A and Bollati, V and Cantone, L and Zaccagnini, G and Maimone, B and Fuschi, P and Da, Silva D and Schiavulli, M and Aday, S and Caputo, M and Madeddu, P and Emanueli, C and Martelli, F and Spinetti, G},
doi = {10.1016/j.ymthe.2018.06.003},
journal = {Molecular Therapy},
pages = {1694--1705},
title = {miR-210 enhances the therapeutic potential of bone-marrow-derived circulating proangiogenic cells in the setting of limb ischemia},
url = {http://dx.doi.org/10.1016/j.ymthe.2018.06.003},
volume = {26},
year = {2018}
}

RIS format (EndNote, RefMan)

TY  - JOUR
AB - Therapies based on circulating proangiogenic cells (PACs) have shown promise in ischemic disease models but require further optimization to reach the bedside. Ischemia-associated hypoxia robustly increases microRNA-210 (miR-210) expression in several cell types, including endothelial cells (ECs). In ECs, miR-210 represses EphrinA3 (EFNA3), inducing proangiogenic responses. This study provides new mechanistic evidences for a role of miR-210 in PACs. PACs were obtained from either adult peripheral blood or cord blood. miR-210 expression was modulated with either an inhibitory complementary oligonucleotide (anti-miR-210) or a miRNA mimic (pre-miR-210). Scramble and absence of transfection served as controls. As expected, hypoxia increased miR-210 in PACs. In vivo, migration toward and adhesion to the ischemic endothelium facilitate the proangiogenic actions of transplanted PACs. In vitro, PAC migration toward SDF-1α/CXCL12 was impaired by anti-miR-210 and enhanced by pre-miR-210. Moreover, pre-miR-210 increased PAC adhesion to ECs and supported angiogenic responses in co-cultured ECs. These responses were not associated with changes in extracellular miR-210 and were abrogated by lentivirus-mediated EFNA3 overexpression. Finally, ex-vivo pre-miR-210 transfection predisposed PACs to induce post-ischemic therapeutic neovascularization and blood flow recovery in an immunodeficient mouse limb ischemia model. In conclusion, miR-210 modulates PAC functions and improves their therapeutic potential in limb ischemia.
AU - Besnier,M
AU - Gasparino,S
AU - Vono,R
AU - Sangalli,E
AU - Facoetti,A
AU - Bollati,V
AU - Cantone,L
AU - Zaccagnini,G
AU - Maimone,B
AU - Fuschi,P
AU - Da,Silva D
AU - Schiavulli,M
AU - Aday,S
AU - Caputo,M
AU - Madeddu,P
AU - Emanueli,C
AU - Martelli,F
AU - Spinetti,G
DO - 10.1016/j.ymthe.2018.06.003
EP - 1705
PY - 2018///
SN - 1525-0016
SP - 1694
TI - miR-210 enhances the therapeutic potential of bone-marrow-derived circulating proangiogenic cells in the setting of limb ischemia
T2 - Molecular Therapy
UR - http://dx.doi.org/10.1016/j.ymthe.2018.06.003
UR - http://gateway.webofknowledge.com/gateway/Gateway.cgi?GWVersion=2&SrcApp=PARTNER_APP&SrcAuth=LinksAMR&KeyUT=WOS:000439688600010&DestLinkType=FullRecord&DestApp=ALL_WOS&UsrCustomerID=1ba7043ffcc86c417c072aa74d649202
UR - http://hdl.handle.net/10044/1/62067
VL - 26
ER -