Cargando…

AIF1L regulates actomyosin contractility and filopodial extensions in human podocytes

Podocytes are highly-specialized epithelial cells essentially required for the generation and the maintenance of the kidney filtration barrier. This elementary function is directly based on an elaborated cytoskeletal apparatus establishing a complex network of primary and secondary processes. Here,...

Descripción completa

Detalles Bibliográficos
Autores principales: Yasuda-Yamahara, Mako, Rogg, Manuel, Yamahara, Kosuke, Maier, Jasmin I., Huber, Tobias B., Schell, Christoph
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Public Library of Science 2018
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6042786/
https://www.ncbi.nlm.nih.gov/pubmed/30001384
http://dx.doi.org/10.1371/journal.pone.0200487
_version_ 1783339220093370368
author Yasuda-Yamahara, Mako
Rogg, Manuel
Yamahara, Kosuke
Maier, Jasmin I.
Huber, Tobias B.
Schell, Christoph
author_facet Yasuda-Yamahara, Mako
Rogg, Manuel
Yamahara, Kosuke
Maier, Jasmin I.
Huber, Tobias B.
Schell, Christoph
author_sort Yasuda-Yamahara, Mako
collection PubMed
description Podocytes are highly-specialized epithelial cells essentially required for the generation and the maintenance of the kidney filtration barrier. This elementary function is directly based on an elaborated cytoskeletal apparatus establishing a complex network of primary and secondary processes. Here, we identify the actin-bundling protein allograft-inflammatory-inhibitor 1 like (AIF1L) as a selectively expressed podocyte protein in vivo. We describe the distinct subcellular localization of AIF1L to actin stress fibers, focal adhesion complexes and the nuclear compartment of podocytes in vitro. Genetic deletion of AIF1L in immortalized human podocytes resulted in an increased formation of filopodial extensions and decreased actomyosin contractility. By the use of SILAC based quantitative proteomics analysis we describe the podocyte specific AIF1L interactome and identify several components of the actomyosin machinery such as MYL9 and UNC45A as potential AIF1L interaction partners. Together, these findings indicate an involvement of AIF1L in the stabilization of podocyte morphology by titrating actomyosin contractility and membrane dynamics.
format Online
Article
Text
id pubmed-6042786
institution National Center for Biotechnology Information
language English
publishDate 2018
publisher Public Library of Science
record_format MEDLINE/PubMed
spelling pubmed-60427862018-07-26 AIF1L regulates actomyosin contractility and filopodial extensions in human podocytes Yasuda-Yamahara, Mako Rogg, Manuel Yamahara, Kosuke Maier, Jasmin I. Huber, Tobias B. Schell, Christoph PLoS One Research Article Podocytes are highly-specialized epithelial cells essentially required for the generation and the maintenance of the kidney filtration barrier. This elementary function is directly based on an elaborated cytoskeletal apparatus establishing a complex network of primary and secondary processes. Here, we identify the actin-bundling protein allograft-inflammatory-inhibitor 1 like (AIF1L) as a selectively expressed podocyte protein in vivo. We describe the distinct subcellular localization of AIF1L to actin stress fibers, focal adhesion complexes and the nuclear compartment of podocytes in vitro. Genetic deletion of AIF1L in immortalized human podocytes resulted in an increased formation of filopodial extensions and decreased actomyosin contractility. By the use of SILAC based quantitative proteomics analysis we describe the podocyte specific AIF1L interactome and identify several components of the actomyosin machinery such as MYL9 and UNC45A as potential AIF1L interaction partners. Together, these findings indicate an involvement of AIF1L in the stabilization of podocyte morphology by titrating actomyosin contractility and membrane dynamics. Public Library of Science 2018-07-12 /pmc/articles/PMC6042786/ /pubmed/30001384 http://dx.doi.org/10.1371/journal.pone.0200487 Text en © 2018 Yasuda-Yamahara et al http://creativecommons.org/licenses/by/4.0/ This is an open access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/4.0/) , which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.
spellingShingle Research Article
Yasuda-Yamahara, Mako
Rogg, Manuel
Yamahara, Kosuke
Maier, Jasmin I.
Huber, Tobias B.
Schell, Christoph
AIF1L regulates actomyosin contractility and filopodial extensions in human podocytes
title AIF1L regulates actomyosin contractility and filopodial extensions in human podocytes
title_full AIF1L regulates actomyosin contractility and filopodial extensions in human podocytes
title_fullStr AIF1L regulates actomyosin contractility and filopodial extensions in human podocytes
title_full_unstemmed AIF1L regulates actomyosin contractility and filopodial extensions in human podocytes
title_short AIF1L regulates actomyosin contractility and filopodial extensions in human podocytes
title_sort aif1l regulates actomyosin contractility and filopodial extensions in human podocytes
topic Research Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6042786/
https://www.ncbi.nlm.nih.gov/pubmed/30001384
http://dx.doi.org/10.1371/journal.pone.0200487
work_keys_str_mv AT yasudayamaharamako aif1lregulatesactomyosincontractilityandfilopodialextensionsinhumanpodocytes
AT roggmanuel aif1lregulatesactomyosincontractilityandfilopodialextensionsinhumanpodocytes
AT yamaharakosuke aif1lregulatesactomyosincontractilityandfilopodialextensionsinhumanpodocytes
AT maierjasmini aif1lregulatesactomyosincontractilityandfilopodialextensionsinhumanpodocytes
AT hubertobiasb aif1lregulatesactomyosincontractilityandfilopodialextensionsinhumanpodocytes
AT schellchristoph aif1lregulatesactomyosincontractilityandfilopodialextensionsinhumanpodocytes