Hypoxic human proximal tubular epithelial cells undergo ferroptosis and elicit an NLRP3 inflammasome response in CD1c+ dendritic cells

Abstract Inflammasomes are multiprotein platforms responsible for the release of pro-inflammatory cytokines interleukin (IL)-1β and IL-18. Mouse studies have identified inflammasome activation within dendritic cells (DC) as pivotal for driving tubulointerstitial fibrosis and inflammation, the hallma...

Full description

Bibliographic Details
Main Authors: Kurt T. K. Giuliani, Anca Grivei, Purba Nag, Xiangju Wang, Melissa Rist, Katrina Kildey, Becker Law, Monica S. Ng, Ray Wilkinson, Jacobus Ungerer, Josephine M. Forbes, Helen Healy, Andrew J. Kassianos
Format: Article
Language:English
Published: Nature Publishing Group 2022-08-01
Series:Cell Death and Disease
Online Access:https://doi.org/10.1038/s41419-022-05191-z
_version_ 1811183267838164992
author Kurt T. K. Giuliani
Anca Grivei
Purba Nag
Xiangju Wang
Melissa Rist
Katrina Kildey
Becker Law
Monica S. Ng
Ray Wilkinson
Jacobus Ungerer
Josephine M. Forbes
Helen Healy
Andrew J. Kassianos
author_facet Kurt T. K. Giuliani
Anca Grivei
Purba Nag
Xiangju Wang
Melissa Rist
Katrina Kildey
Becker Law
Monica S. Ng
Ray Wilkinson
Jacobus Ungerer
Josephine M. Forbes
Helen Healy
Andrew J. Kassianos
author_sort Kurt T. K. Giuliani
collection DOAJ
description Abstract Inflammasomes are multiprotein platforms responsible for the release of pro-inflammatory cytokines interleukin (IL)-1β and IL-18. Mouse studies have identified inflammasome activation within dendritic cells (DC) as pivotal for driving tubulointerstitial fibrosis and inflammation, the hallmarks of chronic kidney disease (CKD). However, translation of this work to human CKD remains limited. Here, we examined the complex tubular cell death pathways mediating inflammasome activation in human kidney DC and, thus, CKD progression. Ex vivo patient-derived proximal tubular epithelial cells (PTEC) cultured under hypoxic (1% O2) conditions modelling the CKD microenvironment showed characteristics of ferroptotic cell death, including mitochondrial dysfunction, reductions in the lipid repair enzyme glutathione peroxidase 4 (GPX4) and increases in lipid peroxidation by-product 4-hydroxynonenal (4-HNE) compared with normoxic PTEC. The addition of ferroptosis inhibitor, ferrostatin-1, significantly reduced hypoxic PTEC death. Human CD1c+ DC activated in the presence of hypoxic PTEC displayed significantly increased production of inflammasome-dependent cytokines IL-1β and IL-18. Treatment of co-cultures with VX-765 (caspase-1/4 inhibitor) and MCC950 (NLRP3 inflammasome inhibitor) significantly attenuated IL-1β/IL-18 levels, supporting an NLRP3 inflammasome-dependent DC response. In line with these in vitro findings, in situ immunolabelling of human fibrotic kidney tissue revealed a significant accumulation of tubulointerstitial CD1c+ DC containing active inflammasome (ASC) specks adjacent to ferroptotic PTEC. These data establish ferroptosis as the primary pattern of PTEC necrosis under the hypoxic conditions of CKD. Moreover, this study identifies NLRP3 inflammasome signalling driven by complex tubulointerstitial PTEC-DC interactions as a key checkpoint for therapeutic targeting in human CKD.
first_indexed 2024-04-11T09:45:12Z
format Article
id doaj.art-3f6627c645814f06a5a8676f3b366cb9
institution Directory Open Access Journal
issn 2041-4889
language English
last_indexed 2024-04-11T09:45:12Z
publishDate 2022-08-01
publisher Nature Publishing Group
record_format Article
series Cell Death and Disease
spelling doaj.art-3f6627c645814f06a5a8676f3b366cb92022-12-22T04:31:05ZengNature Publishing GroupCell Death and Disease2041-48892022-08-0113811310.1038/s41419-022-05191-zHypoxic human proximal tubular epithelial cells undergo ferroptosis and elicit an NLRP3 inflammasome response in CD1c+ dendritic cellsKurt T. K. Giuliani0Anca Grivei1Purba Nag2Xiangju Wang3Melissa Rist4Katrina Kildey5Becker Law6Monica S. Ng7Ray Wilkinson8Jacobus Ungerer9Josephine M. Forbes10Helen Healy11Andrew J. Kassianos12Conjoint Internal Medicine Laboratory, Chemical Pathology, Pathology QueenslandConjoint Internal Medicine Laboratory, Chemical Pathology, Pathology QueenslandConjoint Internal Medicine Laboratory, Chemical Pathology, Pathology QueenslandConjoint Internal Medicine Laboratory, Chemical Pathology, Pathology QueenslandConjoint Internal Medicine Laboratory, Chemical Pathology, Pathology QueenslandConjoint Internal Medicine Laboratory, Chemical Pathology, Pathology QueenslandConjoint Internal Medicine Laboratory, Chemical Pathology, Pathology QueenslandConjoint Internal Medicine Laboratory, Chemical Pathology, Pathology QueenslandConjoint Internal Medicine Laboratory, Chemical Pathology, Pathology QueenslandConjoint Internal Medicine Laboratory, Chemical Pathology, Pathology QueenslandFaculty of Medicine, University of QueenslandConjoint Internal Medicine Laboratory, Chemical Pathology, Pathology QueenslandConjoint Internal Medicine Laboratory, Chemical Pathology, Pathology QueenslandAbstract Inflammasomes are multiprotein platforms responsible for the release of pro-inflammatory cytokines interleukin (IL)-1β and IL-18. Mouse studies have identified inflammasome activation within dendritic cells (DC) as pivotal for driving tubulointerstitial fibrosis and inflammation, the hallmarks of chronic kidney disease (CKD). However, translation of this work to human CKD remains limited. Here, we examined the complex tubular cell death pathways mediating inflammasome activation in human kidney DC and, thus, CKD progression. Ex vivo patient-derived proximal tubular epithelial cells (PTEC) cultured under hypoxic (1% O2) conditions modelling the CKD microenvironment showed characteristics of ferroptotic cell death, including mitochondrial dysfunction, reductions in the lipid repair enzyme glutathione peroxidase 4 (GPX4) and increases in lipid peroxidation by-product 4-hydroxynonenal (4-HNE) compared with normoxic PTEC. The addition of ferroptosis inhibitor, ferrostatin-1, significantly reduced hypoxic PTEC death. Human CD1c+ DC activated in the presence of hypoxic PTEC displayed significantly increased production of inflammasome-dependent cytokines IL-1β and IL-18. Treatment of co-cultures with VX-765 (caspase-1/4 inhibitor) and MCC950 (NLRP3 inflammasome inhibitor) significantly attenuated IL-1β/IL-18 levels, supporting an NLRP3 inflammasome-dependent DC response. In line with these in vitro findings, in situ immunolabelling of human fibrotic kidney tissue revealed a significant accumulation of tubulointerstitial CD1c+ DC containing active inflammasome (ASC) specks adjacent to ferroptotic PTEC. These data establish ferroptosis as the primary pattern of PTEC necrosis under the hypoxic conditions of CKD. Moreover, this study identifies NLRP3 inflammasome signalling driven by complex tubulointerstitial PTEC-DC interactions as a key checkpoint for therapeutic targeting in human CKD.https://doi.org/10.1038/s41419-022-05191-z
spellingShingle Kurt T. K. Giuliani
Anca Grivei
Purba Nag
Xiangju Wang
Melissa Rist
Katrina Kildey
Becker Law
Monica S. Ng
Ray Wilkinson
Jacobus Ungerer
Josephine M. Forbes
Helen Healy
Andrew J. Kassianos
Hypoxic human proximal tubular epithelial cells undergo ferroptosis and elicit an NLRP3 inflammasome response in CD1c+ dendritic cells
Cell Death and Disease
title Hypoxic human proximal tubular epithelial cells undergo ferroptosis and elicit an NLRP3 inflammasome response in CD1c+ dendritic cells
title_full Hypoxic human proximal tubular epithelial cells undergo ferroptosis and elicit an NLRP3 inflammasome response in CD1c+ dendritic cells
title_fullStr Hypoxic human proximal tubular epithelial cells undergo ferroptosis and elicit an NLRP3 inflammasome response in CD1c+ dendritic cells
title_full_unstemmed Hypoxic human proximal tubular epithelial cells undergo ferroptosis and elicit an NLRP3 inflammasome response in CD1c+ dendritic cells
title_short Hypoxic human proximal tubular epithelial cells undergo ferroptosis and elicit an NLRP3 inflammasome response in CD1c+ dendritic cells
title_sort hypoxic human proximal tubular epithelial cells undergo ferroptosis and elicit an nlrp3 inflammasome response in cd1c dendritic cells
url https://doi.org/10.1038/s41419-022-05191-z
work_keys_str_mv AT kurttkgiuliani hypoxichumanproximaltubularepithelialcellsundergoferroptosisandelicitannlrp3inflammasomeresponseincd1cdendriticcells
AT ancagrivei hypoxichumanproximaltubularepithelialcellsundergoferroptosisandelicitannlrp3inflammasomeresponseincd1cdendriticcells
AT purbanag hypoxichumanproximaltubularepithelialcellsundergoferroptosisandelicitannlrp3inflammasomeresponseincd1cdendriticcells
AT xiangjuwang hypoxichumanproximaltubularepithelialcellsundergoferroptosisandelicitannlrp3inflammasomeresponseincd1cdendriticcells
AT melissarist hypoxichumanproximaltubularepithelialcellsundergoferroptosisandelicitannlrp3inflammasomeresponseincd1cdendriticcells
AT katrinakildey hypoxichumanproximaltubularepithelialcellsundergoferroptosisandelicitannlrp3inflammasomeresponseincd1cdendriticcells
AT beckerlaw hypoxichumanproximaltubularepithelialcellsundergoferroptosisandelicitannlrp3inflammasomeresponseincd1cdendriticcells
AT monicasng hypoxichumanproximaltubularepithelialcellsundergoferroptosisandelicitannlrp3inflammasomeresponseincd1cdendriticcells
AT raywilkinson hypoxichumanproximaltubularepithelialcellsundergoferroptosisandelicitannlrp3inflammasomeresponseincd1cdendriticcells
AT jacobusungerer hypoxichumanproximaltubularepithelialcellsundergoferroptosisandelicitannlrp3inflammasomeresponseincd1cdendriticcells
AT josephinemforbes hypoxichumanproximaltubularepithelialcellsundergoferroptosisandelicitannlrp3inflammasomeresponseincd1cdendriticcells
AT helenhealy hypoxichumanproximaltubularepithelialcellsundergoferroptosisandelicitannlrp3inflammasomeresponseincd1cdendriticcells
AT andrewjkassianos hypoxichumanproximaltubularepithelialcellsundergoferroptosisandelicitannlrp3inflammasomeresponseincd1cdendriticcells