ASN's Mission

To create a world without kidney diseases, the ASN Alliance for Kidney Health elevates care by educating and informing, driving breakthroughs and innovation, and advocating for policies that create transformative changes in kidney medicine throughout the world.

learn more

Contact ASN

1401 H St, NW, Ste 900, Washington, DC 20005

email@asn-online.org

202-640-4660

The Latest on X

Kidney Week

Please note that you are viewing an archived section from 2023 and some content may be unavailable. To unlock all content for 2023, please visit the archives.

Abstract: SA-PO338

Regeneration-Associated Cell-Derived Extracellular Vesicles Preserve Kidney Function After Acute Ischemia Injury

Session Information

Category: Development, Stem Cells, and Regenerative Medicine

  • 600 Development, Stem Cells, and Regenerative Medicine

Authors

  • Salybekov, Amankeldi, Shonan Kamakura Sogo Byoin, Kamakura, Kanagawa, Japan
  • Okamura, Shigeaki, Shonan Kamakura Sogo Byoin, Kamakura, Kanagawa, Japan
  • Hidaka, Sumi, Shonan Kamakura Sogo Byoin, Kamakura, Kanagawa, Japan
  • Ohtake, Takayasu, Shonan Kamakura Sogo Byoin, Kamakura, Kanagawa, Japan
  • Asahara, Takayuki, Shonan Kamakura Sogo Byoin, Kamakura, Kanagawa, Japan
  • Kobayashi, Shuzo, Shonan Kamakura Sogo Byoin, Kamakura, Kanagawa, Japan
Background

Under vasculogenic conditioning, pro-inflammatory cell subsets of peripheral blood mononuclear cells (PBMCs) shift their phenotype to pro-regenerative cells such as vasculogenic endothelial progenitor cells, M2 macrophages, and regulatory T cells, collectively designated as regeneration-associated cells (RACs). In this study, we evaluated the therapeutic efficacy of RAC-derived extracellular vesicles (RACev) compared with a vehicle-treated group using the rat kidney ischemia-reperfusion injury (K-IRI) model.

Methods

Human PBMCs were cultured with defined growth factors for seven and then two days to harvest RACs and RAC-ev, respectively. EV sizes were characterized by nanoparticle tracking analysis. Transmission electron microscopy revealed a bi-layered membrane structure and flow cytometry confirmed the presence of EV-specific markers (CD9, CD63, and Alix).

Results

Notably, systemic injection of RACev significantly decreased serum creatinine and blood urine nitrogen (P < 0.01 and P < 0.005) at day three after the onset of K-IRI than the control group. Histologically, the treatment group showed less fibrosis in the cortex and medullary areas (P < 0.04 and P < 0.01) compared to the control group. CD31 staining confirmed enhanced capillary densities at the cortex and medullary areas in the treatment group compared to the control group (P < 0.003). These beneficial effects were coupled with significant expression of angiogenesis (miR-126-3p/5p, miR-195-3p/5p, miR-146-3p/5p), anti-fibrosis (miR-133a-3p, miR-29b-3p), anti-inflammation (miR-10a-3p, miR-21-3p, miR-24-2p), and anti-apoptosis miRs by RACev. In vivo bioluminescence analysis showed preferential accumulation of RACev in the IR-injured kidney.

Conclusion

In conclusion, systemic transplantation of RACev improved kidney function via protecting from tissue fibrosis, anti-inflammation, and angiogenesis miR delivery to the ischemic tissue.