The focal adhesion-associated proteins DOCK5 and GIT2 comprise a rheostat in control of epithelial invasion

Research output: Contribution to journalJournal articleResearchpeer-review

Standard

The focal adhesion-associated proteins DOCK5 and GIT2 comprise a rheostat in control of epithelial invasion. / Frank, Scott R; Köllmann, C P; van Lidth de Jeude, J F; Thiagarajah, J R; Engelholm, L H; Frödin, M; Hansen, S H.

In: Oncogene, Vol. 36, No. 13, 30.03.2017, p. 1816-1828.

Research output: Contribution to journalJournal articleResearchpeer-review

Harvard

Frank, SR, Köllmann, CP, van Lidth de Jeude, JF, Thiagarajah, JR, Engelholm, LH, Frödin, M & Hansen, SH 2017, 'The focal adhesion-associated proteins DOCK5 and GIT2 comprise a rheostat in control of epithelial invasion', Oncogene, vol. 36, no. 13, pp. 1816-1828. https://doi.org/10.1038/onc.2016.345

APA

Frank, S. R., Köllmann, C. P., van Lidth de Jeude, J. F., Thiagarajah, J. R., Engelholm, L. H., Frödin, M., & Hansen, S. H. (2017). The focal adhesion-associated proteins DOCK5 and GIT2 comprise a rheostat in control of epithelial invasion. Oncogene, 36(13), 1816-1828. https://doi.org/10.1038/onc.2016.345

Vancouver

Frank SR, Köllmann CP, van Lidth de Jeude JF, Thiagarajah JR, Engelholm LH, Frödin M et al. The focal adhesion-associated proteins DOCK5 and GIT2 comprise a rheostat in control of epithelial invasion. Oncogene. 2017 Mar 30;36(13):1816-1828. https://doi.org/10.1038/onc.2016.345

Author

Frank, Scott R ; Köllmann, C P ; van Lidth de Jeude, J F ; Thiagarajah, J R ; Engelholm, L H ; Frödin, M ; Hansen, S H. / The focal adhesion-associated proteins DOCK5 and GIT2 comprise a rheostat in control of epithelial invasion. In: Oncogene. 2017 ; Vol. 36, No. 13. pp. 1816-1828.

Bibtex

@article{650f10b56c784a269ef185b1d35d02ad,
title = "The focal adhesion-associated proteins DOCK5 and GIT2 comprise a rheostat in control of epithelial invasion",
abstract = "DOCK proteins are guanine nucleotide exchange factors for Rac and Cdc42 GTPases. DOCK1 is the founding member of the family and acts downstream of integrins via the canonical Crk-p130Cas complex to activate Rac GTPases in numerous contexts. In contrast, DOCK5, which possesses the greatest similarity to DOCK1, remains sparingly studied. Here we establish that DOCK5 has a non-redundant role in regulating motile and invasive capacities of epithelial cells. DOCK1 is constitutively associated with sites of integrin attachment termed focal adhesions (FAs). In contrast, we demonstrate that DOCK5 recruitment to FAs in Hela cells is restricted by GIT2, an established regulator of FA signaling. We determine that GIT2 is targeted to FAs in response to Rho-ROCK signaling and actomyosin contractility. Accordingly, inhibition of ROCK activity or MLC function promotes enrichment of DOCK5 in membrane protrusions and nascent cell-substratum adhesions. We further demonstrate that GIT2 inhibits the interaction of DOCK5 with Crk. Moreover, we show that depletion of GIT2 promotes DOCK5-dependent activation of the Crk-p130Cas signaling cascade to promote Rac1-mediated lamellipodial protrusion and FA turnover. The antagonism between GIT2 and DOCK5 extends to non-transformed MCF10A mammary epithelial cells, with DOCK5 'dialing-up' and GIT2 'dialing-down' invasiveness. Finally, we determine that DOCK5 inhibition attenuates invasion and metastasis of MDA-MB-231 cells and prolongs life span of mice injected with these cells. Collectively, our work identifies DOCK5 as a key regulator of epithelial invasion and metastasis, and demonstrates that suppression of DOCK5 by GIT2 represents a previously unappreciated mechanism for coordination of Rho and Rac GTPases.Oncogene advance online publication, 26 September 2016; doi:10.1038/onc.2016.345.",
author = "Frank, {Scott R} and K{\"o}llmann, {C P} and {van Lidth de Jeude}, {J F} and Thiagarajah, {J R} and Engelholm, {L H} and M Fr{\"o}din and Hansen, {S H}",
year = "2017",
month = mar,
day = "30",
doi = "10.1038/onc.2016.345",
language = "English",
volume = "36",
pages = "1816--1828",
journal = "Oncogene",
issn = "0950-9232",
publisher = "nature publishing group",
number = "13",

}

RIS

TY - JOUR

T1 - The focal adhesion-associated proteins DOCK5 and GIT2 comprise a rheostat in control of epithelial invasion

AU - Frank, Scott R

AU - Köllmann, C P

AU - van Lidth de Jeude, J F

AU - Thiagarajah, J R

AU - Engelholm, L H

AU - Frödin, M

AU - Hansen, S H

PY - 2017/3/30

Y1 - 2017/3/30

N2 - DOCK proteins are guanine nucleotide exchange factors for Rac and Cdc42 GTPases. DOCK1 is the founding member of the family and acts downstream of integrins via the canonical Crk-p130Cas complex to activate Rac GTPases in numerous contexts. In contrast, DOCK5, which possesses the greatest similarity to DOCK1, remains sparingly studied. Here we establish that DOCK5 has a non-redundant role in regulating motile and invasive capacities of epithelial cells. DOCK1 is constitutively associated with sites of integrin attachment termed focal adhesions (FAs). In contrast, we demonstrate that DOCK5 recruitment to FAs in Hela cells is restricted by GIT2, an established regulator of FA signaling. We determine that GIT2 is targeted to FAs in response to Rho-ROCK signaling and actomyosin contractility. Accordingly, inhibition of ROCK activity or MLC function promotes enrichment of DOCK5 in membrane protrusions and nascent cell-substratum adhesions. We further demonstrate that GIT2 inhibits the interaction of DOCK5 with Crk. Moreover, we show that depletion of GIT2 promotes DOCK5-dependent activation of the Crk-p130Cas signaling cascade to promote Rac1-mediated lamellipodial protrusion and FA turnover. The antagonism between GIT2 and DOCK5 extends to non-transformed MCF10A mammary epithelial cells, with DOCK5 'dialing-up' and GIT2 'dialing-down' invasiveness. Finally, we determine that DOCK5 inhibition attenuates invasion and metastasis of MDA-MB-231 cells and prolongs life span of mice injected with these cells. Collectively, our work identifies DOCK5 as a key regulator of epithelial invasion and metastasis, and demonstrates that suppression of DOCK5 by GIT2 represents a previously unappreciated mechanism for coordination of Rho and Rac GTPases.Oncogene advance online publication, 26 September 2016; doi:10.1038/onc.2016.345.

AB - DOCK proteins are guanine nucleotide exchange factors for Rac and Cdc42 GTPases. DOCK1 is the founding member of the family and acts downstream of integrins via the canonical Crk-p130Cas complex to activate Rac GTPases in numerous contexts. In contrast, DOCK5, which possesses the greatest similarity to DOCK1, remains sparingly studied. Here we establish that DOCK5 has a non-redundant role in regulating motile and invasive capacities of epithelial cells. DOCK1 is constitutively associated with sites of integrin attachment termed focal adhesions (FAs). In contrast, we demonstrate that DOCK5 recruitment to FAs in Hela cells is restricted by GIT2, an established regulator of FA signaling. We determine that GIT2 is targeted to FAs in response to Rho-ROCK signaling and actomyosin contractility. Accordingly, inhibition of ROCK activity or MLC function promotes enrichment of DOCK5 in membrane protrusions and nascent cell-substratum adhesions. We further demonstrate that GIT2 inhibits the interaction of DOCK5 with Crk. Moreover, we show that depletion of GIT2 promotes DOCK5-dependent activation of the Crk-p130Cas signaling cascade to promote Rac1-mediated lamellipodial protrusion and FA turnover. The antagonism between GIT2 and DOCK5 extends to non-transformed MCF10A mammary epithelial cells, with DOCK5 'dialing-up' and GIT2 'dialing-down' invasiveness. Finally, we determine that DOCK5 inhibition attenuates invasion and metastasis of MDA-MB-231 cells and prolongs life span of mice injected with these cells. Collectively, our work identifies DOCK5 as a key regulator of epithelial invasion and metastasis, and demonstrates that suppression of DOCK5 by GIT2 represents a previously unappreciated mechanism for coordination of Rho and Rac GTPases.Oncogene advance online publication, 26 September 2016; doi:10.1038/onc.2016.345.

U2 - 10.1038/onc.2016.345

DO - 10.1038/onc.2016.345

M3 - Journal article

C2 - 27669437

VL - 36

SP - 1816

EP - 1828

JO - Oncogene

JF - Oncogene

SN - 0950-9232

IS - 13

ER -

ID: 179349833