2012
DOI: 10.1074/jbc.m112.394965
|View full text |Cite
|
Sign up to set email alerts
|

Rho-associated Coiled-coil Kinase (ROCK) Protein Controls Microtubule Dynamics in a Novel Signaling Pathway That Regulates Cell Migration

Abstract: Background: ROCK regulates microtubule acetylation. Results: ROCK phosphorylation of TPPP1/p25 inhibits the interaction between TPPP1 and HDAC6, resulting in increased HDAC6 deacetylation of microtubules, leading to increased cell motility. Conclusion: ROCK phosphorylation of TPPP1 is a novel signaling pathway that regulates cell migration via increased HDAC6 activity and reduced MT acetylation. Significance: This newly discovered ROCK/TPPP/HDAC6/MT signaling pathway might have important implications for cell … Show more

Help me understand this report

Search citation statements

Order By: Relevance

Paper Sections

Select...
2
1
1
1

Citation Types

7
79
2
1

Year Published

2013
2013
2021
2021

Publication Types

Select...
6
2

Relationship

1
7

Authors

Journals

citations
Cited by 71 publications
(89 citation statements)
references
References 38 publications
7
79
2
1
Order By: Relevance
“…Overexpression of TPPP1 in cells promotes MT polymerization and an increase in MT acetylation, whereas introduction of TPPP1 RNAi reduces MT acetylation [43]. Phosphorylation of TPPP1 by Rho-associated coiled-coil kinase (ROCK) and cyclin-dependent kinase 1 (CDK1) impairs the interaction between TPPP1 and HDAC6, which, in turn, results in increased HDAC6 activity followed by a decrease in cell motility and an increase in cell proliferation, respectively [46,47]. Moreover, a broad spectrum of regulators, which directly interact with and inhibit HDAC6 tubulin deacetylase activity or aggresome formation ability, has been well documented, including paxillin [48], CYLD [49,50], dysferlin [51], Mdp3 [52], p62 [53] and RanBPM [54].…”
Section: Sam68mentioning
confidence: 99%
“…Overexpression of TPPP1 in cells promotes MT polymerization and an increase in MT acetylation, whereas introduction of TPPP1 RNAi reduces MT acetylation [43]. Phosphorylation of TPPP1 by Rho-associated coiled-coil kinase (ROCK) and cyclin-dependent kinase 1 (CDK1) impairs the interaction between TPPP1 and HDAC6, which, in turn, results in increased HDAC6 activity followed by a decrease in cell motility and an increase in cell proliferation, respectively [46,47]. Moreover, a broad spectrum of regulators, which directly interact with and inhibit HDAC6 tubulin deacetylase activity or aggresome formation ability, has been well documented, including paxillin [48], CYLD [49,50], dysferlin [51], Mdp3 [52], p62 [53] and RanBPM [54].…”
Section: Sam68mentioning
confidence: 99%
“…LIMKs are activated through phosphorylation by Rho-associated coiled-coil kinase (ROCK), p21-activated ki-nase 1 (PAK1) and PAK4, downstream effectors of the small GTPases Rho, Rac and Cdc42, respectively [3]- [5]. Interestingly, although it has been reported that LIMK1 and LIMK2 interact with Tubulin Polymerization Promoting Protein 1 (TPPP1) in vitro and in vivo [6]- [8], TPPP1 is not a LIMK substrate in cells [9].…”
Section: Introductionmentioning
confidence: 99%
“…Flag-LIMK1, Flag-LIMK1-DN, Flag-HADC6, myc-cofilin, myc-cofilin S3A, myc-cofilin S3D constructs were transiently transfected into U2OS cells using the Lipofectamine™ 2000 (Life Technologies) transfection reagent according to manufacturer's recommendations. Cells expressing Flag-TPPP1 were generated as previously described [9].…”
Section: Mammalian Cell Culturementioning
confidence: 99%
See 1 more Smart Citation
“…MT dynamics is spatially and temporally regulated by several pathways and MT-interacting proteins (see Table 1): tau, a MT-associated protein (MAP) that stabilizes MTs [Maccioni and Cambiazo, 1995], and katanin, the MT-severing enzyme [Roll-Mecak and McNally, 2010], both interact with the MT lattice; PAR-1 (also known as MARK) phosphorylates classical MAPs and detaches MAPs from MTs [Matenia and Mandelkow, 2009]; +TIPs, the MT plusend-tracking proteins, specifically control the dynamic properties of the MT end [Gouveia and Akhmanova, 2010;Schuyler and Pellman, 2001]; ROCK pathway regulates MT dynamics via phosphorylation of the tubulin polymerization promoting protein 1 (TPPP1/p25) [Schofield et al 2012]; (aPKC)-Aurora A-NDEL1 pathway is crucial for the regulation of MT organization during neurite extension [Mori et al 2009]; Dishevelled (Dvl) pathway and the cooperation of Wnt-Dvl pathways increase MT stability though Gsk3β inhibition and c-Jun N-terminal kinase (JNK) activation [Ciani and Salinas, 2007]. These are only some of the known regulatory mechanisms that contribute to orchestrate MT dynamic remodelling.…”
Section: Microtubule Dynamics Regulationmentioning
confidence: 99%