Review
The role of Rho GTPases and associated kinases in regulating neurite outgrowth

https://doi.org/10.1016/S1357-2725(01)00167-4Get rights and content

Abstract

Neurones are highly specialised cells that can extend over great distances, enabling the complex networking of the nervous system. We are beginning to understand in detail the molecular mechanisms that control the shape of neurones during development. One family of proteins that are clearly essential are the Rho GTPases which have a pivotal role in regulating the actin cytoskeleton in all cell types. The Rho GTPases are responsible for the activation and downregulation of many downstream kinases. This review discusses individual kinases that are regulated by three members of the Rho GTPases, Rac, Rho and Cdc42 and their function during neurite outgrowth and remodelling.

Introduction

Since the discovery in the mid 1980s and early 1990s that three members of the Rho family of small GTPases (Rac1, RhoA and Cdc42) function to regulate the actin cytoskeleton, many research laboratories have examined if they control the movement and shape of their favourite cell types. Their biological function was initially described in Swiss 3T3 fibroblasts where Rac1 induces the formation of sheet-like structures (lamellipodia), Cdc42 causes the formation of cell surface microspikes (filopodia), while RhoA enhances cell attachment and contractility [1], [2], [3]. The Rho GTPases have since then been shown to control different aspects of the cellular cytoskeleton, largely dependent on the cell types studied. Rac1, RhoA and Cdc42 switch between an active and an inactive state in a highly regulated manner. The active forms function to trigger downstream signalling pathways, many of which are dominated by kinase cascades [4]. The past few years have seen a tremendous amount of new information on the regulation and function of these three proteins in neurones and non-neuronal cells (for recent reviews see, [5], [6], [7], [8], [9], [10]). In neurones it has become largely apparent that Rac1 and Cdc42 induce, while RhoA inhibits neurite outgrowth [6]. However, more work needs to be carried out to understand the relationship between these three proteins in different neurones and whether it is subject to temporal and spatial regulation. In addition to neurite outgrowth, these small GTPases have been shown to control the establishment of neuronal polarity, neuronal migration, growth cone guidance and the function of synapses [6], [7]. Together, the Rho family of small GTPases can regulate every process in a neurone that involves plasticity of the cytoskeleton. This review focuses on studies that have addressed the function of Rac1, RhoA and Cdc42 and downstream kinases in primary neurones both in vitro and in vivo.

Section snippets

The Rho GTPases and neurite outgrowth

A neurone is the highly specialised, basic unit of the functioning nervous system. It transports information to other neurones and non-neuronal cells, enabling the co-ordinated functioning of an organism. Neurones have a highly polarised morphology, with a single long process termed the axon and often many short neurites known as dendrites (Fig. 1). Information is usually received at the dendrites, transported through the cell and delivered further via the axon. For the nervous system to

The p35/Cdk5 kinase

Cdk5 is a member of the cyclin dependent kinase (Cdk) family, which are proline directed serine/threonine kinases that require the association of a regulatory protein for activity [50]. Cdk5 is activated in postmitotic neurones by two highly related proteins, p35 and p39 [51]. The p35 and p39 are to date the only known Cdk5 kinase activators. Whereas, Cdk5 is ubiquitously expressed, p35 and p39 are induced upon cellular differentiation and not detectable in proliferating cells [51]. For a long

Kinases regulated by RhoA

The most studied RhoA effectors are the serine/threonine Rho-associated kinases. Whereas, ROKβ/ROCK1 expression is hardly detectable in brain, ROKα/Rho-kinase/ROCK2/p160ROCK is highly enriched in pyramidal neurones of the cortex and hippocampus and cerebellar Purkinje neurones [91]. Both proteins have an N-terminal kinase domain and C-terminal RhoGTP binding and autoinhibitory domains. In an analogous way to the Pak kinases, Rho associated kinases exist in an autoinhibited state, which is

Kinases regulated by Cdc42

Much less is known about the kinases that function downstream from Cdc42 in neurones. The most studied are Pak1, Pak2 and Pak3, although they have primarily been examined as targets of Rac1, rather than Cdc42. Because both Rac1 and Cdc42 appear to regulate these Pak kinases indistinguishably from each other and Rac1 can be activated by Cdc42, it is hard to separate the effects of these two GTPases on Pak1, Pak2 and Pak3 kinases in vivo. An interesting model has been put forward suggesting that

If only it was this simple

It is becoming increasingly apparent that the Rho GTPases often have opposing roles in regulating the neuronal cytoskeleton and that the same signals that activate one, cause the downregulation of another. Currently, the best examples are Rac1 and RhoA and their differential regulation by the ephrins and semaphorins [37], [38], [40], [41]. Overall, it is clear that these molecules do not work in isolation and signalling pathways that emanate from them converge. Figuring out exactly what kinases

What the future holds

The aim of this review was to sum up and simplify a lot of complex signalling pathways and describe what is known to date on how they may modulate the cytoskeleton in postmitotic, differentiating neurones. Particular emphasis was made on neurite outgrowth and remodelling. It is apparent that the correct wiring of the nervous system is governed by the maintenance of a fine balance between seemingly opposing signals and that localised and transient changes dictate processes such as growth cone

Acknowledgements

Due to space limitations I have not been able to describe all the contributions researchers have made to this field and I apologise to the authors whose work has not been mentioned. I would like to thank Britta Eickholt (King’s College London) for helpful discussions and critically reading this manuscript. Work in the author’s laboratory is supported by the Wellcome Trust, The BBSRC and Glaxo–SmithKline.

References (132)

  • A.J. Ridley et al.

    The small GTP-binding protein Rho regulates the assembly of focal adhesions and actin stress fibers in response to growth factors

    Cell

    (1992)
  • A.J. Ridley et al.

    The small GTP-binding protein Rac regulates growth factor-induced membrane ruffling

    Cell

    (1992)
  • C.D. Nobes et al.

    Rho, Rac, and Cdc42 GTPases regulate the assembly of multimolecular focal complexes associated with actin stress fibers, lamellipodia, and filopodia

    Cell

    (1995)
  • A.J. Ridley, Rho, Oxford University Press, 2000, pp....
  • A. Hall et al.

    Rho GTPases: molecular switches that control the organization and dynamics of the actin cytoskeleton

    Philos. Trans. R. Soc. London B. Biol. Sci.

    (2000)
  • L. Luo

    Rho GTPases in neuronal morphogenesis

    Nat. Rev. Neurosci.

    (2000)
  • B.J. Dickson

    Rho GTPases in growth cone guidance

    Curr. Opin. Neurobiol.

    (2001)
  • D. Bar-Sagi et al.

    Ras and Rho GTPases: a family reunion

    Cell

    (2000)
  • L. Redmond et al.

    The role of Notch and Rho GTPase signaling in the control of dendritic development

    Curr. Opin. Neurobiol.

    (2001)
  • A.A. Schmitz et al.

    Rho GTPases: signaling, migration, and invasion

    Exp. Cell Res.

    (2000)
  • B.K. Mueller

    Growth cone guidance: first steps towards a deeper understanding

    Annu. Rev. Neurosci.

    (1999)
  • F. Nakamura et al.

    Molecular basis of semaphorin-mediated axon guidance

    J. Neurobiol.

    (2000)
  • J.A. Raper

    Semaphorins and their receptors in vertebrates and invertebrates

    Curr. Opin. Neurobiol.

    (2000)
  • C.M. Waterman-Storer et al.

    Positive feedback interactions between microtubule and actin dynamics during cell motility

    Curr. Opin. Cell Biol.

    (1999)
  • N. Kobayashi et al.

    A role of microtubules during the formation of cell processes in neuronal and non-neuronal cells

    Cell Tissue Res.

    (1998)
  • P.W. Baas

    Microtubules and neuronal polarity: lessons from mitosis

    Neuron

    (1999)
  • L. Van Aelst et al.

    Rho GTPases and signaling networks

    Genes Dev.

    (1997)
  • M.R. Alam et al.

    Kalirin, a cytosolic protein with spectrin-like and GDP/GTP exchange factor-like domains that interacts with peptidylglycine alpha-amidating monooxygenase, an integral membrane peptide-processing enzyme

    J. Biol. Chem.

    (1997)
  • P. Billuart et al.

    Oligophrenin-1 encodes a RhoGAP protein involved in X-linked mental retardation

    Nature

    (1998)
  • M.R. Brouns et al.

    p190 RhoGAP is the principal Src substrate in brain and regulates axon outgrowth, guidance and fasciculation

    Nat. Cell Biol.

    (2001)
  • G.G. Habets et al.

    Identification of an invasion-inducing gene, Tiam-1, that encodes a protein with homology to GDP–GTP exchangers for Rho-like proteins

    Cell

    (1994)
  • C. Hall et al.

    alpha2-chimaerin, a Cdc42/Rac1 regulator, is selectively expressed in the rat embryonic nervous system and is involved in neuritogenesis in N1E-115 neuroblastoma cells

    J. Neurosci.

    (2001)
  • Y. Horii et al.

    A novel oncogene, ost, encodes a guanine nucleotide exchange factor that potentially links Rho and Rac signaling pathways

    EMBO J.

    (1994)
  • M. Hoshino et al.

    Identification of the stef gene that encodes a novel guanine nucleotide exchange factor specific for Rac1

    J. Biol. Chem.

    (1999)
  • E. Ehler et al.

    Expression of Tiam-1 in the developing brain suggests a role for the Tiam-1-Rac signaling pathway in cell migration and neurite outgrowth

    Mol. Cell Neurosci.

    (1997)
  • X.M. Ma et al.

    Expression of kalirin, a neuronal GDP/GTP exchange factor of the trio family, in the central nervous system of the adult rat

    J. Comp. Neurol.

    (2001)
  • C.N. Adra et al.

    RhoGDIgamma: a GDP-dissociation inhibitor for Rho proteins with preferential expression in brain and pancreas

    Proc. Natl. Acad. Sci. U.S.A.

    (1997)
  • S.S. Andersen et al.

    Axon formation: a molecular model for the generation of neuronal polarity

    Bioessays

    (2000)
  • F. Bradke et al.

    The role of local actin instability in axon formation

    Science

    (1999)
  • M.L. Ruchhoeft et al.

    The neuronal architecture of Xenopus retinal ganglion cells is sculpted by rho-family GTPases in vivo

    J. Neurosci

    (1999)
  • T.B. Kuhn et al.

    Rac1-dependent actin filament organization in growth cones is necessary for beta1-integrin-mediated advance but not for growth on poly-d-lysine

    J. Neurobiol.

    (1998)
  • L. Luo et al.

    Small GTPases in axon outgrowth

    Perspect. Dev. Neurobiol.

    (1996)
  • T. Lee et al.

    Essential roles of Drosophila RhoA in the regulation of neuroblast proliferation and dendritic but not axonal morphogenesis

    Neuron

    (2000)
  • R. Threadgill et al.

    Regulation of dendritic growth and remodeling by Rho, Rac, and Cdc42

    Neuron

    (1997)
  • D.G. Wilkinson

    Multiple roles of EPH receptors and ephrins in neural development

    Nat. Rev. Neurosci.

    (2001)
  • M. Nakamoto

    Eph receptors and ephrins

    Int. J. Biochem. Cell Biol.

    (2000)
  • S. Wahl et al.

    Ephrin-A5 induces collapse of growth cones by activating Rho and Rho kinase

    J. Cell Biol.

    (2000)
  • S.M. Shamah et al.

    EphA receptors regulate growth cone dynamics through the novel guanine nucleotide exchange factor ephexin

    Cell

    (2001)
  • K.L. Whitford et al.

    Plexin signaling via off-track and Rho family GTPases

    Neuron

    (2001)
  • M.H. Driessens et al.

    Plexin-B semaphorin receptors interact directly with active Rac and regulate the actin cytoskeleton by activating Rho

    Curr. Biol.

    (2001)
  • H. Hu et al.

    Plexin B mediates axon guidance in Drosophila by simultaneously inhibiting active Rac and enhancing RhoA signaling

    Neuron

    (2001)
  • H.G. Vikis et al.

    The semaphorin receptor plexin-B1 specifically interacts with active Rac in a ligand-dependent manner

    Proc. Natl. Acad. Sci. U.S.A.

    (2000)
  • L. Luo et al.

    Distinct morphogenetic functions of similar small GTPases: Drosophila Drac1 is involved in axonal outgrowth and myoblast fusion

    Genes Dev.

    (1994)
  • N. Kaufmann et al.

    Drosophila Rac1 controls motor axon guidance

    Development

    (1998)
  • Z. Li et al.

    Rho GTPases regulate distinct aspects of dendritic arbor growth in Xenopus central neurons in vivo

    Nat. Neurosci.

    (2000)
  • L. Luo et al.

    Differential effects of the Rac GTPase on Purkinje cell axons and dendritic trunks and spines

    Nature

    (1996)
  • I. Arozarena et al.

    Maintenance of CDC42 GDP-bound state by Rho-GDI inhibits MAP kinase activation by the exchange factor Ras-GRF. evidence for Ras-GRF function being inhibited by Cdc42-GDP but unaffected by CDC42-GTP

    J. Biol. Chem.

    (2001)
  • N. Di-Poi et al.

    Mechanism of NADPH oxidase activation by the Rac/Rho-GDI complex

    Biochemistry

    (2001)
  • J.Z. Tsien et al.

    The essential role of hippocampal CA1 NMDA receptor-dependent synaptic plasticity in spatial memory

    Cell

    (1996)
  • M. Meyerson et al.

    A family of human cdc2-related protein kinases

    EMBO J.

    (1992)
  • Cited by (119)

    • The role of Actopaxin in tumor metastasis

      2022, Progress in Biophysics and Molecular Biology
      Citation Excerpt :

      It is not only an important modulator of integrin-induced Rho family signaling transduction to the cytoskeleton, but also an important negative regulator of adhesion-dependent signaling. Besides, CdGAP also works as an inhibitor of lamellipodia formation induced by growth factors (LaLonde et al., 2006; Sun et al., 2004; Nikolic, 2002; Chen et al., 2000; Mehra et al., 2021). CdGAP interacts directly to the amino terminal of Actopaxin through its central domain to regulate integrin-dependent changes in cell morphology and motility (LaLonde et al., 2006; Jenna et al., 2002).

    • An Orchestra of Reovirus Receptors: Still Searching for the Conductor

      2018, Advances in Virus Research
      Citation Excerpt :

      NgR1 lacks a cytoplasmic domain and likely is incapable of intraneuronal signaling without cytoplasmic-domain-containing coreceptors. Under physiological conditions, NgR1 binds myelin-associated ligands and recruits two neural coreceptors to form a tripartite complex that initiates RhoA activation and actin reorganization to prevent axonal outgrowth (Nikolic, 2002). Many other viruses hijack the Rho pathway during entry (Taylor et al., 2011), but it is not known whether reovirus binding to NgR1 initiates signaling events or ligation of coreceptors.

    View all citing articles on Scopus
    View full text