Cargando…

Tubular epithelial cell-to-macrophage communication forms a negative feedback loop via extracellular vesicle transfer to promote renal inflammation and apoptosis in diabetic nephropathy

Background: Macrophage infiltration around lipotoxic tubular epithelial cells (TECs) is a hallmark of diabetic nephropathy (DN). However, how these two types of cells communicate remains obscure. We previously demonstrated that LRG1 was elevated in the process of kidney injury. Here, we demonstrated...

Descripción completa

Detalles Bibliográficos
Autores principales: Jiang, Wen-juan, Xu, Chuan-ting, Du, Chang-lin, Dong, Jia-hui, Xu, Song-bing, Hu, Bing-feng, Feng, Rui, Zang, Dan-dan, Meng, Xiao-ming, Huang, Cheng, Li, Jun, Ma, Tao-tao
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Ivyspring International Publisher 2022
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8690920/
https://www.ncbi.nlm.nih.gov/pubmed/34987648
http://dx.doi.org/10.7150/thno.63735
_version_ 1784618703971680256
author Jiang, Wen-juan
Xu, Chuan-ting
Du, Chang-lin
Dong, Jia-hui
Xu, Song-bing
Hu, Bing-feng
Feng, Rui
Zang, Dan-dan
Meng, Xiao-ming
Huang, Cheng
Li, Jun
Ma, Tao-tao
author_facet Jiang, Wen-juan
Xu, Chuan-ting
Du, Chang-lin
Dong, Jia-hui
Xu, Song-bing
Hu, Bing-feng
Feng, Rui
Zang, Dan-dan
Meng, Xiao-ming
Huang, Cheng
Li, Jun
Ma, Tao-tao
author_sort Jiang, Wen-juan
collection PubMed
description Background: Macrophage infiltration around lipotoxic tubular epithelial cells (TECs) is a hallmark of diabetic nephropathy (DN). However, how these two types of cells communicate remains obscure. We previously demonstrated that LRG1 was elevated in the process of kidney injury. Here, we demonstrated that macrophage-derived, LRG1-enriched extracellular vesicles (EVs) exacerbated DN. Methods: We induced an experimental T2DM mouse model with a HFD diet for four months. Renal primary epithelial cells and macrophage-derived EVs were isolated from T2D mice by differential ultracentrifugation. To investigate whether lipotoxic TEC-derived EV (EV(e)) activate macrophages, mouse bone marrow-derived macrophages (BMDMs) were incubated with EV(e). To investigate whether activated macrophage-derived EVs (EV(m)) induce lipotoxic TEC apoptosis, EV(m) were cocultured with primary renal tubular epithelial cells. Subsequently, we evaluated the effect of LRG1 in EV(e) by investigating the apoptosis mechanism. Results: We demonstrated that incubation of primary TECs of DN or HK-2 mTECs with lysophosphatidyl choline (LPC) increased the release of EV(e). Interestingly, TEC-derived EV(e) activated an inflammatory phenotype in macrophages and induced the release of macrophage-derived EV(m). Furthermore, EV(m) could induce apoptosis in TECs injured by LPC. Importantly, we found that leucine-rich α-2-glycoprotein 1 (LRG1)-enriched EV(e) activated macrophages via a TGFβR1-dependent process and that tumor necrosis factor-related apoptosis-inducing ligand (TRAIL)-enriched EV(m) induced apoptosis in injured TECs via a death receptor 5 (DR5)-dependent process. Conclusion: Our findings indicated a novel cell communication mechanism between tubular epithelial cells and macrophages in DN, which could be a potential therapeutic target.
format Online
Article
Text
id pubmed-8690920
institution National Center for Biotechnology Information
language English
publishDate 2022
publisher Ivyspring International Publisher
record_format MEDLINE/PubMed
spelling pubmed-86909202022-01-04 Tubular epithelial cell-to-macrophage communication forms a negative feedback loop via extracellular vesicle transfer to promote renal inflammation and apoptosis in diabetic nephropathy Jiang, Wen-juan Xu, Chuan-ting Du, Chang-lin Dong, Jia-hui Xu, Song-bing Hu, Bing-feng Feng, Rui Zang, Dan-dan Meng, Xiao-ming Huang, Cheng Li, Jun Ma, Tao-tao Theranostics Research Paper Background: Macrophage infiltration around lipotoxic tubular epithelial cells (TECs) is a hallmark of diabetic nephropathy (DN). However, how these two types of cells communicate remains obscure. We previously demonstrated that LRG1 was elevated in the process of kidney injury. Here, we demonstrated that macrophage-derived, LRG1-enriched extracellular vesicles (EVs) exacerbated DN. Methods: We induced an experimental T2DM mouse model with a HFD diet for four months. Renal primary epithelial cells and macrophage-derived EVs were isolated from T2D mice by differential ultracentrifugation. To investigate whether lipotoxic TEC-derived EV (EV(e)) activate macrophages, mouse bone marrow-derived macrophages (BMDMs) were incubated with EV(e). To investigate whether activated macrophage-derived EVs (EV(m)) induce lipotoxic TEC apoptosis, EV(m) were cocultured with primary renal tubular epithelial cells. Subsequently, we evaluated the effect of LRG1 in EV(e) by investigating the apoptosis mechanism. Results: We demonstrated that incubation of primary TECs of DN or HK-2 mTECs with lysophosphatidyl choline (LPC) increased the release of EV(e). Interestingly, TEC-derived EV(e) activated an inflammatory phenotype in macrophages and induced the release of macrophage-derived EV(m). Furthermore, EV(m) could induce apoptosis in TECs injured by LPC. Importantly, we found that leucine-rich α-2-glycoprotein 1 (LRG1)-enriched EV(e) activated macrophages via a TGFβR1-dependent process and that tumor necrosis factor-related apoptosis-inducing ligand (TRAIL)-enriched EV(m) induced apoptosis in injured TECs via a death receptor 5 (DR5)-dependent process. Conclusion: Our findings indicated a novel cell communication mechanism between tubular epithelial cells and macrophages in DN, which could be a potential therapeutic target. Ivyspring International Publisher 2022-01-01 /pmc/articles/PMC8690920/ /pubmed/34987648 http://dx.doi.org/10.7150/thno.63735 Text en © The author(s) https://creativecommons.org/licenses/by/4.0/This is an open access article distributed under the terms of the Creative Commons Attribution License (https://creativecommons.org/licenses/by/4.0/). See http://ivyspring.com/terms for full terms and conditions.
spellingShingle Research Paper
Jiang, Wen-juan
Xu, Chuan-ting
Du, Chang-lin
Dong, Jia-hui
Xu, Song-bing
Hu, Bing-feng
Feng, Rui
Zang, Dan-dan
Meng, Xiao-ming
Huang, Cheng
Li, Jun
Ma, Tao-tao
Tubular epithelial cell-to-macrophage communication forms a negative feedback loop via extracellular vesicle transfer to promote renal inflammation and apoptosis in diabetic nephropathy
title Tubular epithelial cell-to-macrophage communication forms a negative feedback loop via extracellular vesicle transfer to promote renal inflammation and apoptosis in diabetic nephropathy
title_full Tubular epithelial cell-to-macrophage communication forms a negative feedback loop via extracellular vesicle transfer to promote renal inflammation and apoptosis in diabetic nephropathy
title_fullStr Tubular epithelial cell-to-macrophage communication forms a negative feedback loop via extracellular vesicle transfer to promote renal inflammation and apoptosis in diabetic nephropathy
title_full_unstemmed Tubular epithelial cell-to-macrophage communication forms a negative feedback loop via extracellular vesicle transfer to promote renal inflammation and apoptosis in diabetic nephropathy
title_short Tubular epithelial cell-to-macrophage communication forms a negative feedback loop via extracellular vesicle transfer to promote renal inflammation and apoptosis in diabetic nephropathy
title_sort tubular epithelial cell-to-macrophage communication forms a negative feedback loop via extracellular vesicle transfer to promote renal inflammation and apoptosis in diabetic nephropathy
topic Research Paper
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8690920/
https://www.ncbi.nlm.nih.gov/pubmed/34987648
http://dx.doi.org/10.7150/thno.63735
work_keys_str_mv AT jiangwenjuan tubularepithelialcelltomacrophagecommunicationformsanegativefeedbackloopviaextracellularvesicletransfertopromoterenalinflammationandapoptosisindiabeticnephropathy
AT xuchuanting tubularepithelialcelltomacrophagecommunicationformsanegativefeedbackloopviaextracellularvesicletransfertopromoterenalinflammationandapoptosisindiabeticnephropathy
AT duchanglin tubularepithelialcelltomacrophagecommunicationformsanegativefeedbackloopviaextracellularvesicletransfertopromoterenalinflammationandapoptosisindiabeticnephropathy
AT dongjiahui tubularepithelialcelltomacrophagecommunicationformsanegativefeedbackloopviaextracellularvesicletransfertopromoterenalinflammationandapoptosisindiabeticnephropathy
AT xusongbing tubularepithelialcelltomacrophagecommunicationformsanegativefeedbackloopviaextracellularvesicletransfertopromoterenalinflammationandapoptosisindiabeticnephropathy
AT hubingfeng tubularepithelialcelltomacrophagecommunicationformsanegativefeedbackloopviaextracellularvesicletransfertopromoterenalinflammationandapoptosisindiabeticnephropathy
AT fengrui tubularepithelialcelltomacrophagecommunicationformsanegativefeedbackloopviaextracellularvesicletransfertopromoterenalinflammationandapoptosisindiabeticnephropathy
AT zangdandan tubularepithelialcelltomacrophagecommunicationformsanegativefeedbackloopviaextracellularvesicletransfertopromoterenalinflammationandapoptosisindiabeticnephropathy
AT mengxiaoming tubularepithelialcelltomacrophagecommunicationformsanegativefeedbackloopviaextracellularvesicletransfertopromoterenalinflammationandapoptosisindiabeticnephropathy
AT huangcheng tubularepithelialcelltomacrophagecommunicationformsanegativefeedbackloopviaextracellularvesicletransfertopromoterenalinflammationandapoptosisindiabeticnephropathy
AT lijun tubularepithelialcelltomacrophagecommunicationformsanegativefeedbackloopviaextracellularvesicletransfertopromoterenalinflammationandapoptosisindiabeticnephropathy
AT mataotao tubularepithelialcelltomacrophagecommunicationformsanegativefeedbackloopviaextracellularvesicletransfertopromoterenalinflammationandapoptosisindiabeticnephropathy