Abstract

BackgroundThe therapeutic effect of mesenchymal stem cells (MSCs) from human adipose tissue on renal interstitial fibrosis has been demonstrated by several groups. However, the way to enhance the renoprotective effect of adipose-derived mesenchymal stem cells (AMSCs) and the possible mechanisms are still unclear. The present study aimed to determine whether glial cell line-derived neurotrophic factor (GDNF)-modified AMSCs hold an enhanced protective effect on renal fibrosis.MethodsAMSCs were isolated and purified for culture. The gene GDNF has been constructed to transfect into AMSCs. The ability of GFP-AMSCs and GDNF-AMSCs supernatants to promote tube formation of endothelial cells, repair damaged endothelial cell junctions, and improve endothelial cell function was compared by using tube formation assay, immunofluorescence techniques, and vascular ring assay, respectively. Furthermore, HE and Masson staining were used to observe the histological morphology of the kidney in vivo. Peritubular capillary changes were detected and analyzed by fluorescence microangiography (FMA). Meanwhile, the hypoxia, oxidative stress, fibrotic markers, and PI3K/Akt pathway proteins were measured by western blot or qRT-PCR technics.ResultsCompared with GFP-AMSCs only, GDNF-AMSCs could enhance the repair of injured endothelial cells and promote angiogenesis through secreting more growth factors in the supernatant of GDNF-AMSC culture media demonstrated in vitro studies. Studies in vivo, unilateral ureteral obstruction (UUO)-induced mice were injected with transfected AMSCs through their tail veins. We showed that enhanced homing of AMSCs was observed in the GDNF-AMSC group compared with the GFP-AMSC group. The animals treated with GDNF-AMSCs exhibited an improvement of capillary rarefaction and fibrosis induced by obstructed kidney compared with the GFP-AMSC group. Furthermore, we reported that GDNF-AMSCs protect renal tissues against microvascular injuries via activation of the PI3K/Akt signaling pathway. Therefore, GDNF-AMSCs further ameliorated the tissue hypoxia, suppressed oxidative stress, and finally inhibited endothelial to mesenchymal transition noting by decreased coexpression of endothelial cell (CD31) and myofibroblast (a-SMA) markers.ConclusionCollectively, our data indicated that the GDNF gene enhances the ability of AMSCs in improving renal microcirculation through PI3K/Akt/eNOS signaling pathway and afterward inhibit the EndMT process and kidney fibrogenesis, which should have a vast of implications in designing future remedies for chronic kidney disease (CKD) treatment.

Highlights

  • The therapeutic effect of mesenchymal stem cells (MSCs) from human adipose tissue on renal interstitial fibrosis has been demonstrated by several groups

  • Collectively, our data indicated that the glial cell line-derived neurotrophic factor (GDNF) gene enhances the ability of adipose-derived mesenchymal stem cells (AMSCs) in improving renal microcirculation through Phosphoinositide 3 kinase (PI3K)/Akt/endothelial nitric oxide (NO) synthase (eNOS) signaling pathway and afterward inhibit the endothelial-to-mesenchymal transition (EndMT) process and kidney fibrogenesis, which should have a vast of implications in designing future remedies for chronic kidney disease (CKD) treatment

  • AMSCs at the third passage were transfected with adenovirus vectors at the most appropriate multiplicity of infection (MOI) = 20 following standard procedures

Read more

Summary

Introduction

The therapeutic effect of mesenchymal stem cells (MSCs) from human adipose tissue on renal interstitial fibrosis has been demonstrated by several groups. The way to enhance the renoprotective effect of adipose-derived mesenchymal stem cells (AMSCs) and the possible mechanisms are still unclear. The characteristics of renal fibrosis are hypoxia, oxidative stress, inflammatory cell infiltration, fibroblast activation, and extracellular matrix accumulation. These mentioned characteristics are the common pathogenic pathway of progressive CKD renal disease [2,3,4]. The loss of capillaries results in the imbalance of local tissue oxygen demand and supply, which causes renal fibrosis and further leads to a series of events, such as the accumulation of reactive oxygen metabolites and EndMT

Methods
Results
Discussion
Conclusion
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call