scholarly journals A novel polymeric micelle used for in vivo MR imaging tracking of neural stem cells in acute ischemic stroke

RSC Advances ◽  
2017 ◽  
Vol 7 (25) ◽  
pp. 15041-15052 ◽  
Author(s):  
Liejing Lu ◽  
Yong Wang ◽  
Minghui Cao ◽  
Meiwei Chen ◽  
Bingling Lin ◽  
...  

Novel cationic polymeric micelles based on biodegradable poly(aspartic acid-dimethylethanediamine)–lysine–cholic acid were synthesized for in vivo tracking therapeutic stem cells using MRI.

Molecules ◽  
2016 ◽  
Vol 21 (9) ◽  
pp. 1143 ◽  
Author(s):  
Fang Zhang ◽  
Xiaohui Duan ◽  
Liejing Lu ◽  
Xiang Zhang ◽  
Xiaomei Zhong ◽  
...  

2021 ◽  
Vol 16 (9) ◽  
pp. 1677
Author(s):  
Qiong-Lan Yuan ◽  
Sheng-Jun Wen ◽  
Xi-Min Zheng ◽  
Li-Fen Liu ◽  
Na-Na Li ◽  
...  

2015 ◽  
Vol 2015 ◽  
pp. 1-11 ◽  
Author(s):  
Xiao-Mei Zhong ◽  
Fang Zhang ◽  
Ming Yang ◽  
Xue-Hua Wen ◽  
Xiang Zhang ◽  
...  

Neural stem cells in the adult mammalian brain have a significant level of neurogenesis plasticity. In vivo monitoring of adult endogenous NSCs would be of great benefit to the understanding of the neurogenesis plasticity under normal and pathological conditions. Here we show the feasibility of in vivo targeted MR imaging of endogenous NSCs in adult mouse brain by intraventricular delivery of monoclonal anti-CD15 antibody conjugated superparamagnetic iron oxide nanoparticles. After intraventricular administration of these nanoparticles, the subpopulation of NSCs in the anterior subventricular zone and the beginning of the rostral migratory stream could be in situ labeled and were in vivo visualized with 7.0-T MR imaging during a period from 1 day to 7 days after the injection. Histology confirmed that the injected targeted nanoparticles were specifically bound to CD15 positive cells and their surrounding extracellular matrix. Our results suggest that in vivo targeted MR imaging of endogenous neural stem cells in adult rodent brain could be achieved by using anti-CD15-SPIONs as the molecular probe; and this targeting imaging strategy has the advantage of a rapid in vivo monitoring of the subpopulation of endogenous NSCs in adult brains.


Biomaterials ◽  
2016 ◽  
Vol 77 ◽  
pp. 291-306 ◽  
Author(s):  
Francesca J. Nicholls ◽  
Matthew W. Rotz ◽  
Harmanvir Ghuman ◽  
Keith W. MacRenaris ◽  
Thomas J. Meade ◽  
...  

Stroke ◽  
2013 ◽  
Vol 44 (suppl_1) ◽  
Author(s):  
Yaning Li ◽  
Jun Huang ◽  
Xiaosong He ◽  
Yaohui Tang ◽  
Yifan Lv ◽  
...  

Background and purpose: SDF-1 has double-edged function after ischemic stroke. It can attract both inflammatory cells and neural stem cells (NSC). However, the action of SDF-1 in regulating neural stem cells migration and maturation during post-ischemic stroke is unclear. Here we used adeno-associated virus (AAV) to deliver SDF-1 gene into the peri-infarct area one week after permanent middle cerebral artery occlusion (MCAO) to investigate the effect of SDF-1 on endogenous NSC migration and maturation. Methods and materials: Twenty-four adult ICR male mice received AAV carrying SDF-1 or GFP gene transfer into the peri-infarct area one week after MCAO. Brain atrophy volume, neurobehavioral tests and immunohistochemistry were performed to evaluate the effects of SDF-1 on endogenous NSC migration, maturation and neuronal function repair. Results: SDF-1 was highly expressed in peri-infarct area for at least four weeks after gene transfer. Brain atrophy volume was significantly reduced in AAV-SDF-1 group compared to AAV-GFP gourp ( p <0.05). The results of neurobehavioral tests including neurological deficits and rotarod test paralleled the result of atrophy volume, with neuronal function greatly improved in AAV-SDF-1 group ( p <0.05). Immunohistology showed that the number of nestin and doublecortin positive cells in peri-infarct area was significantly increased in AAV-SDF-1 group in contrast to AAV-GFP group ( p <0.05). In addition, there was no difference in myeloperoxidase positive cells in ischemic peri-infarct area between AAV-SDF-1 and AAV-GFP group ( p >0.05). Conclusion: Our results demonstrated that SDF-1 gene therapy significantly increased NSC migration and maturation without increasing focal inflammatory response; consequently reduced brain atrophy volume and improved the neurological outcomes of animals, suggesting that delayed SDF-1 hyperexpression plays a key role in neurogenesis and neural repair. These in vivo results indicate that SDF-1 holds great potential as a stem cell chemotactic factor in the treatment of ischemic stroke.


2012 ◽  
Vol 5 (1) ◽  
pp. 79-83 ◽  
Author(s):  
Da-Jeong Chang ◽  
Hyeyoung Moon ◽  
Yong Hyun Lee ◽  
Nayeon Lee ◽  
Hong J. Lee ◽  
...  

2008 ◽  
Vol 60 (6) ◽  
pp. 1506-1511 ◽  
Author(s):  
Jesús Ruiz-Cabello ◽  
Piotr Walczak ◽  
Dorota A. Kedziorek ◽  
Vadappuram P. Chacko ◽  
Anna H. Schmieder ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document