Abstract

Dendritic cells (DCs) are unique immune cells that can link innate and adaptive immune responses and Immunometabolism greatly impacts their phenotype. Rapamycin is a macrolide compound that has immunosuppressant functions and is used to prevent graft loss in kidney transplantation. The current study evaluated the therapeutic potential of ex-vivo rapamycin treated DCs to protect kidneys in a mouse model of acute kidney injury (AKI). For the rapamycin single (S) treatment (Rapa-S-DC), Veh-DCs were treated with rapamycin (10 ng/mL) for 1 h before LPS. In contrast, rapamycin multiple (M) treatment (Rapa-M-DC) were exposed to 3 treatments over 7 days. Only multiple ex-vivo rapamycin treatments of DCs induced a persistent reprogramming of mitochondrial metabolism. These DCs had 18-fold more mitochondria, had almost 4-fold higher oxygen consumption rates, and produced more ATP compared to Veh-DCs (Veh treated control DCs). Pathway analysis showed IL10 signaling as a major contributing pathway to the altered immunophenotype after Rapamycin treatment compared to vehicle with significantly lower cytokines Tnfa, Il1b, and Il6, while regulators of mitochondrial content Pgc1a, Tfam, and Ho1 remained elevated. Critically, adoptive transfer of rapamycin-treated DCs to WT recipients 24 h before bilateral kidney ischemia significantly protected the kidneys from injury with a significant 3-fold improvement in kidney function. Last, the infusion of DCs containing higher mitochondria numbers (treated ex-vivo with healthy isolated mitochondria (10 µg/mL) one day before) also partially protected the kidneys from IRI. These studies demonstrate that pre-emptive infusion of ex-vivo reprogrammed DCs that have higher mitochondria content has therapeutic capacity to induce an anti-inflammatory regulatory phenotype to protect kidneys from injury.

Highlights

  • The pathobiology of acute kidney injury is multifactorial and involves intricate interactions between renal parenchymal cells and immune cells [1,2]

  • LPS treated Dendritic cells (DCs) were used for RNASeq or labeled with various mitochondrial dyes

  • In the current study we demonstrated protection from kidney ischemia-reperfusion injury (IRI) induced by adoptive transfer of rapamycin-treated DCs alternatively regulates

Read more

Summary

Introduction

The pathobiology of acute kidney injury is multifactorial and involves intricate interactions between renal parenchymal cells and immune cells [1,2]. DCs are one of the primary cell types involved in innate immunity and are essential in providing protection against pathogens. DCs are located extensively throughout all tissues, with a subset of DCs (CD11c+ DCs) residing in the renal parenchyma. These CD11c+ DCs play a pathogenic role in worsening renal injury following ischemia-reperfusion injury (IRI) [8]. IRI is defined as injury resulting from the reintroduction of oxygenated blood to an organ after a period of ischemia and is often seen in myocardial infarction, stroke, and solid organ transplantation

Objectives
Methods
Results
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