FTY720 Regulates Mitochondria Biogenesis in Dendritic Cells to Prevent Kidney Ischemic Reperfusion Injury

Dendritic cells (DCs) are central in regulating immune responses of kidney ischemia-reperfusion injury (IRI), and strategies to alter DC function may provide new therapeutic opportunities. Sphingosine 1-phosphate (S1P) modulates immunity through binding to its receptors (S1P1-5), and protection from...

Full description

Bibliographic Details
Main Authors: Thomas V. Rousselle, Canan Kuscu, Cem Kuscu, Kailo Schlegel, LiPing Huang, Maria Namwanje, James D. Eason, Liza Makowski, Daniel Maluf, Valeria Mas, Amandeep Bajwa
Format: Article
Language:English
Published: Frontiers Media S.A. 2020-06-01
Series:Frontiers in Immunology
Subjects:
Online Access:https://www.frontiersin.org/article/10.3389/fimmu.2020.01278/full
id doaj-e1a321347247487bb554ae50d64b3edd
record_format Article
collection DOAJ
language English
format Article
sources DOAJ
author Thomas V. Rousselle
Canan Kuscu
Cem Kuscu
Kailo Schlegel
LiPing Huang
Maria Namwanje
James D. Eason
Liza Makowski
Daniel Maluf
Valeria Mas
Amandeep Bajwa
spellingShingle Thomas V. Rousselle
Canan Kuscu
Cem Kuscu
Kailo Schlegel
LiPing Huang
Maria Namwanje
James D. Eason
Liza Makowski
Daniel Maluf
Valeria Mas
Amandeep Bajwa
FTY720 Regulates Mitochondria Biogenesis in Dendritic Cells to Prevent Kidney Ischemic Reperfusion Injury
Frontiers in Immunology
dendritic cell
FTY720
mitochondria
sphingosine-1-phosphate receptor
macrophages
metabolism
author_facet Thomas V. Rousselle
Canan Kuscu
Cem Kuscu
Kailo Schlegel
LiPing Huang
Maria Namwanje
James D. Eason
Liza Makowski
Daniel Maluf
Valeria Mas
Amandeep Bajwa
author_sort Thomas V. Rousselle
title FTY720 Regulates Mitochondria Biogenesis in Dendritic Cells to Prevent Kidney Ischemic Reperfusion Injury
title_short FTY720 Regulates Mitochondria Biogenesis in Dendritic Cells to Prevent Kidney Ischemic Reperfusion Injury
title_full FTY720 Regulates Mitochondria Biogenesis in Dendritic Cells to Prevent Kidney Ischemic Reperfusion Injury
title_fullStr FTY720 Regulates Mitochondria Biogenesis in Dendritic Cells to Prevent Kidney Ischemic Reperfusion Injury
title_full_unstemmed FTY720 Regulates Mitochondria Biogenesis in Dendritic Cells to Prevent Kidney Ischemic Reperfusion Injury
title_sort fty720 regulates mitochondria biogenesis in dendritic cells to prevent kidney ischemic reperfusion injury
publisher Frontiers Media S.A.
series Frontiers in Immunology
issn 1664-3224
publishDate 2020-06-01
description Dendritic cells (DCs) are central in regulating immune responses of kidney ischemia-reperfusion injury (IRI), and strategies to alter DC function may provide new therapeutic opportunities. Sphingosine 1-phosphate (S1P) modulates immunity through binding to its receptors (S1P1-5), and protection from kidney IRI occurs in mice treated with S1PR agonist, FTY720 (FTY). We tested if ex vivo propagation of DCs with FTY could be used as cellular therapy to limit the off-target effects associated with systemic FTY administration in kidney IRI. DCs have the ability of regulate innate and adaptive responses and we posited that treatment of DC with FTY may underlie improvements in kidney IRI. Herein, it was observed that treatment of bone marrow derived dendritic cells (BMDCs) with FTY induced mitochondrial biogenesis, FTY-treated BMDCs (FTY-DCs) showed significantly higher oxygen consumption rate and ATP production compared to vehicle treated BMDCs (Veh-DCs). Adoptive transfer of FTY-DCs to mice 24 h before or 4 h after IRI significantly protected the kidneys from injury compared to mice treated with Veh-DCs. Additionally, allogeneic adoptive transfer of C57BL/6J FTY-DCs into BALB/c mice equally protected the kidneys from IRI. FTY-DCs propagated from S1pr1-deficient DCs derived from CD11cCreS1pr1fl/fl mice as well as blunting mitochondrial oxidation in wildtype (WT) FTY-DCs prior to transfer abrogated the protection observed by FTY-DCs. We queried if DC mitochondrial content alters kidney responses after IRI, a novel but little studied phenomenon shown to be integral to regulation of the immune response. Transfer of mitochondria rich FTY-DCs protects kidneys from IRI as transferred FTY-DCs donated their mitochondria to recipient splenocytes (i.e., macrophages) and prior splenectomy abrogated this protection. Adoptive transfer of FTY-DCs either prior to or after ischemic injury protects kidneys from IRI demonstrating a potent role for donor DC-mitochondria in FTY's efficacy. This is the first evidence, to our knowledge, that DCs have the potential to protect against kidney injury by donating mitochondria to splenic macrophages to alter their bioenergetics thus making them anti-inflammatory. In conclusion, the results support that ex vivo FTY720-induction of the regulatory DC phenotype could have therapeutic relevance that can be preventively infused to reduce acute kidney injury.
topic dendritic cell
FTY720
mitochondria
sphingosine-1-phosphate receptor
macrophages
metabolism
url https://www.frontiersin.org/article/10.3389/fimmu.2020.01278/full
work_keys_str_mv AT thomasvrousselle fty720regulatesmitochondriabiogenesisindendriticcellstopreventkidneyischemicreperfusioninjury
AT canankuscu fty720regulatesmitochondriabiogenesisindendriticcellstopreventkidneyischemicreperfusioninjury
AT cemkuscu fty720regulatesmitochondriabiogenesisindendriticcellstopreventkidneyischemicreperfusioninjury
AT kailoschlegel fty720regulatesmitochondriabiogenesisindendriticcellstopreventkidneyischemicreperfusioninjury
AT lipinghuang fty720regulatesmitochondriabiogenesisindendriticcellstopreventkidneyischemicreperfusioninjury
AT marianamwanje fty720regulatesmitochondriabiogenesisindendriticcellstopreventkidneyischemicreperfusioninjury
AT jamesdeason fty720regulatesmitochondriabiogenesisindendriticcellstopreventkidneyischemicreperfusioninjury
AT lizamakowski fty720regulatesmitochondriabiogenesisindendriticcellstopreventkidneyischemicreperfusioninjury
AT danielmaluf fty720regulatesmitochondriabiogenesisindendriticcellstopreventkidneyischemicreperfusioninjury
AT valeriamas fty720regulatesmitochondriabiogenesisindendriticcellstopreventkidneyischemicreperfusioninjury
AT amandeepbajwa fty720regulatesmitochondriabiogenesisindendriticcellstopreventkidneyischemicreperfusioninjury
_version_ 1724529718962683904
spelling doaj-e1a321347247487bb554ae50d64b3edd2020-11-25T03:41:25ZengFrontiers Media S.A.Frontiers in Immunology1664-32242020-06-011110.3389/fimmu.2020.01278522860FTY720 Regulates Mitochondria Biogenesis in Dendritic Cells to Prevent Kidney Ischemic Reperfusion InjuryThomas V. Rousselle0Canan Kuscu1Cem Kuscu2Kailo Schlegel3LiPing Huang4Maria Namwanje5James D. Eason6Liza Makowski7Daniel Maluf8Valeria Mas9Amandeep Bajwa10Transplant Research Institute, James D. Eason Transplant Institute, Department of Surgery, School of Medicine, University of Tennessee Health Sciences Center, Memphis, TN, United StatesTransplant Research Institute, James D. Eason Transplant Institute, Department of Surgery, School of Medicine, University of Tennessee Health Sciences Center, Memphis, TN, United StatesTransplant Research Institute, James D. Eason Transplant Institute, Department of Surgery, School of Medicine, University of Tennessee Health Sciences Center, Memphis, TN, United StatesDivision of Nephrology and the Center for Immunity, Inflammation and Regenerative Medicine, Department of Medicine, University of Virginia, Charlottesville, VA, United StatesDivision of Nephrology and the Center for Immunity, Inflammation and Regenerative Medicine, Department of Medicine, University of Virginia, Charlottesville, VA, United StatesDepartment of Pediatrics and Genetics, University of Tennessee Health Science Center, Memphis, TN, United StatesTransplant Research Institute, James D. Eason Transplant Institute, Department of Surgery, School of Medicine, University of Tennessee Health Sciences Center, Memphis, TN, United StatesDepartment of Medicine - Division of Hematology and Oncology, College of Medicine, Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Sciences Center, Memphis, TN, United StatesTransplant Research Institute, James D. Eason Transplant Institute, Department of Surgery, School of Medicine, University of Tennessee Health Sciences Center, Memphis, TN, United StatesTransplant Research Institute, James D. Eason Transplant Institute, Department of Surgery, School of Medicine, University of Tennessee Health Sciences Center, Memphis, TN, United StatesTransplant Research Institute, James D. Eason Transplant Institute, Department of Surgery, School of Medicine, University of Tennessee Health Sciences Center, Memphis, TN, United StatesDendritic cells (DCs) are central in regulating immune responses of kidney ischemia-reperfusion injury (IRI), and strategies to alter DC function may provide new therapeutic opportunities. Sphingosine 1-phosphate (S1P) modulates immunity through binding to its receptors (S1P1-5), and protection from kidney IRI occurs in mice treated with S1PR agonist, FTY720 (FTY). We tested if ex vivo propagation of DCs with FTY could be used as cellular therapy to limit the off-target effects associated with systemic FTY administration in kidney IRI. DCs have the ability of regulate innate and adaptive responses and we posited that treatment of DC with FTY may underlie improvements in kidney IRI. Herein, it was observed that treatment of bone marrow derived dendritic cells (BMDCs) with FTY induced mitochondrial biogenesis, FTY-treated BMDCs (FTY-DCs) showed significantly higher oxygen consumption rate and ATP production compared to vehicle treated BMDCs (Veh-DCs). Adoptive transfer of FTY-DCs to mice 24 h before or 4 h after IRI significantly protected the kidneys from injury compared to mice treated with Veh-DCs. Additionally, allogeneic adoptive transfer of C57BL/6J FTY-DCs into BALB/c mice equally protected the kidneys from IRI. FTY-DCs propagated from S1pr1-deficient DCs derived from CD11cCreS1pr1fl/fl mice as well as blunting mitochondrial oxidation in wildtype (WT) FTY-DCs prior to transfer abrogated the protection observed by FTY-DCs. We queried if DC mitochondrial content alters kidney responses after IRI, a novel but little studied phenomenon shown to be integral to regulation of the immune response. Transfer of mitochondria rich FTY-DCs protects kidneys from IRI as transferred FTY-DCs donated their mitochondria to recipient splenocytes (i.e., macrophages) and prior splenectomy abrogated this protection. Adoptive transfer of FTY-DCs either prior to or after ischemic injury protects kidneys from IRI demonstrating a potent role for donor DC-mitochondria in FTY's efficacy. This is the first evidence, to our knowledge, that DCs have the potential to protect against kidney injury by donating mitochondria to splenic macrophages to alter their bioenergetics thus making them anti-inflammatory. In conclusion, the results support that ex vivo FTY720-induction of the regulatory DC phenotype could have therapeutic relevance that can be preventively infused to reduce acute kidney injury.https://www.frontiersin.org/article/10.3389/fimmu.2020.01278/fulldendritic cellFTY720mitochondriasphingosine-1-phosphate receptormacrophagesmetabolism