The kinesin KIF9 and reggie/flotillin proteins regulate matrix degradation by macrophage podosomes.

Standard

The kinesin KIF9 and reggie/flotillin proteins regulate matrix degradation by macrophage podosomes. / Cornfine, Susanne; Himmel, Mirko; Kopp, Petra; El-Azzouzi, Karim; Wiesner, Christiane; Krüger, Marcus; Rudel, Thomas; Linder, Stefan.

in: MOL BIOL CELL, Jahrgang 22, Nr. 2, 2, 2011, S. 202-215.

Publikationen: SCORING: Beitrag in Fachzeitschrift/ZeitungSCORING: ZeitschriftenaufsatzForschungBegutachtung

Harvard

Cornfine, S, Himmel, M, Kopp, P, El-Azzouzi, K, Wiesner, C, Krüger, M, Rudel, T & Linder, S 2011, 'The kinesin KIF9 and reggie/flotillin proteins regulate matrix degradation by macrophage podosomes.', MOL BIOL CELL, Jg. 22, Nr. 2, 2, S. 202-215. <http://www.ncbi.nlm.nih.gov/pubmed/21119006?dopt=Citation>

APA

Vancouver

Cornfine S, Himmel M, Kopp P, El-Azzouzi K, Wiesner C, Krüger M et al. The kinesin KIF9 and reggie/flotillin proteins regulate matrix degradation by macrophage podosomes. MOL BIOL CELL. 2011;22(2):202-215. 2.

Bibtex

@article{fcf70a192304471c9ede824abbdece1c,
title = "The kinesin KIF9 and reggie/flotillin proteins regulate matrix degradation by macrophage podosomes.",
abstract = "Podosomes are actin-based matrix contacts in a variety of cell types, most notably monocytic cells, and are characterized by their ability to lyse extracellular matrix material. Besides their dependence on actin regulation, podosomes are also influenced by microtubules and microtubule-dependent transport processes. Here we describe a novel role for KIF9, a previously little-characterized member of the kinesin motor family, in the regulation of podosomes in primary human macrophages. We find that small interfering RNA (siRNA)/short-hairpin RNA-induced knockdown of KIF9 significantly affects both numbers and matrix degradation of podosomes. Overexpression and microinjection experiments reveal that the unique C-terminal region of KIF9 is crucial for these effects, presumably through binding of specific interactors. Indeed, we further identify reggie-1/flotillin-2, a signaling mediator between intracellular vesicles and the cell periphery, as an interactor of the KIF9 C-terminus. Reggie-1 dynamically colocalizes with KIF9 in living cells, and, consistent with KIF9-mediated effects, siRNA-induced knockdown of reggies/flotillins significantly impairs matrix degradation by podosomes. In sum, we identify the kinesin KIF9 and reggie/flotillin proteins as novel regulators of macrophage podosomes and show that their interaction is critical for the matrix-degrading ability of these structures.",
author = "Susanne Cornfine and Mirko Himmel and Petra Kopp and Karim El-Azzouzi and Christiane Wiesner and Marcus Kr{\"u}ger and Thomas Rudel and Stefan Linder",
year = "2011",
language = "Deutsch",
volume = "22",
pages = "202--215",
journal = "MOL BIOL CELL",
issn = "1059-1524",
publisher = "American Society for Cell Biology",
number = "2",

}

RIS

TY - JOUR

T1 - The kinesin KIF9 and reggie/flotillin proteins regulate matrix degradation by macrophage podosomes.

AU - Cornfine, Susanne

AU - Himmel, Mirko

AU - Kopp, Petra

AU - El-Azzouzi, Karim

AU - Wiesner, Christiane

AU - Krüger, Marcus

AU - Rudel, Thomas

AU - Linder, Stefan

PY - 2011

Y1 - 2011

N2 - Podosomes are actin-based matrix contacts in a variety of cell types, most notably monocytic cells, and are characterized by their ability to lyse extracellular matrix material. Besides their dependence on actin regulation, podosomes are also influenced by microtubules and microtubule-dependent transport processes. Here we describe a novel role for KIF9, a previously little-characterized member of the kinesin motor family, in the regulation of podosomes in primary human macrophages. We find that small interfering RNA (siRNA)/short-hairpin RNA-induced knockdown of KIF9 significantly affects both numbers and matrix degradation of podosomes. Overexpression and microinjection experiments reveal that the unique C-terminal region of KIF9 is crucial for these effects, presumably through binding of specific interactors. Indeed, we further identify reggie-1/flotillin-2, a signaling mediator between intracellular vesicles and the cell periphery, as an interactor of the KIF9 C-terminus. Reggie-1 dynamically colocalizes with KIF9 in living cells, and, consistent with KIF9-mediated effects, siRNA-induced knockdown of reggies/flotillins significantly impairs matrix degradation by podosomes. In sum, we identify the kinesin KIF9 and reggie/flotillin proteins as novel regulators of macrophage podosomes and show that their interaction is critical for the matrix-degrading ability of these structures.

AB - Podosomes are actin-based matrix contacts in a variety of cell types, most notably monocytic cells, and are characterized by their ability to lyse extracellular matrix material. Besides their dependence on actin regulation, podosomes are also influenced by microtubules and microtubule-dependent transport processes. Here we describe a novel role for KIF9, a previously little-characterized member of the kinesin motor family, in the regulation of podosomes in primary human macrophages. We find that small interfering RNA (siRNA)/short-hairpin RNA-induced knockdown of KIF9 significantly affects both numbers and matrix degradation of podosomes. Overexpression and microinjection experiments reveal that the unique C-terminal region of KIF9 is crucial for these effects, presumably through binding of specific interactors. Indeed, we further identify reggie-1/flotillin-2, a signaling mediator between intracellular vesicles and the cell periphery, as an interactor of the KIF9 C-terminus. Reggie-1 dynamically colocalizes with KIF9 in living cells, and, consistent with KIF9-mediated effects, siRNA-induced knockdown of reggies/flotillins significantly impairs matrix degradation by podosomes. In sum, we identify the kinesin KIF9 and reggie/flotillin proteins as novel regulators of macrophage podosomes and show that their interaction is critical for the matrix-degrading ability of these structures.

M3 - SCORING: Zeitschriftenaufsatz

VL - 22

SP - 202

EP - 215

JO - MOL BIOL CELL

JF - MOL BIOL CELL

SN - 1059-1524

IS - 2

M1 - 2

ER -