AIF1L regulates actomyosin contractility and filopodial extensions in human podocytes

Standard

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.

in: PLOS ONE, Jahrgang 13, Nr. 7, 2018, S. e0200487.

Publikationen: SCORING: Beitrag in Fachzeitschrift/ZeitungSCORING: ZeitschriftenaufsatzForschungBegutachtung

Harvard

Yasuda-Yamahara, M, Rogg, M, Yamahara, K, Maier, JI, Huber, TB & Schell, C 2018, 'AIF1L regulates actomyosin contractility and filopodial extensions in human podocytes', PLOS ONE, Jg. 13, Nr. 7, S. e0200487. https://doi.org/10.1371/journal.pone.0200487

APA

Yasuda-Yamahara, M., Rogg, M., Yamahara, K., Maier, J. I., Huber, T. B., & Schell, C. (2018). AIF1L regulates actomyosin contractility and filopodial extensions in human podocytes. PLOS ONE, 13(7), e0200487. https://doi.org/10.1371/journal.pone.0200487

Vancouver

Bibtex

@article{ea711b20f10d4eacbff66b69cac979f1,
title = "AIF1L regulates actomyosin contractility and filopodial extensions in human podocytes",
abstract = "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.",
keywords = "Actomyosin, Calcium-Binding Proteins, Cells, Cultured, Focal Adhesions, Humans, Intracellular Signaling Peptides and Proteins, Microfilament Proteins, Myosin Light Chains, Podocytes, Pseudopodia, Stress Fibers, Journal Article, Research Support, Non-U.S. Gov't",
author = "Mako Yasuda-Yamahara and Manuel Rogg and Kosuke Yamahara and Maier, {Jasmin I} and Huber, {Tobias B} and Christoph Schell",
year = "2018",
doi = "10.1371/journal.pone.0200487",
language = "English",
volume = "13",
pages = "e0200487",
journal = "PLOS ONE",
issn = "1932-6203",
publisher = "Public Library of Science",
number = "7",

}

RIS

TY - JOUR

T1 - AIF1L regulates actomyosin contractility and filopodial extensions in human podocytes

AU - Yasuda-Yamahara, Mako

AU - Rogg, Manuel

AU - Yamahara, Kosuke

AU - Maier, Jasmin I

AU - Huber, Tobias B

AU - Schell, Christoph

PY - 2018

Y1 - 2018

N2 - 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.

AB - 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.

KW - Actomyosin

KW - Calcium-Binding Proteins

KW - Cells, Cultured

KW - Focal Adhesions

KW - Humans

KW - Intracellular Signaling Peptides and Proteins

KW - Microfilament Proteins

KW - Myosin Light Chains

KW - Podocytes

KW - Pseudopodia

KW - Stress Fibers

KW - Journal Article

KW - Research Support, Non-U.S. Gov't

U2 - 10.1371/journal.pone.0200487

DO - 10.1371/journal.pone.0200487

M3 - SCORING: Journal article

C2 - 30001384

VL - 13

SP - e0200487

JO - PLOS ONE

JF - PLOS ONE

SN - 1932-6203

IS - 7

ER -