Rho GTPases regulate PTPμ-mediated nasal neurite outgrowth and temporal repulsion of retinal ganglion cell neurons

https://doi.org/10.1016/j.mcn.2006.11.022Get rights and content

Abstract

Members of the receptor protein tyrosine phosphatase (RPTP) subfamily of cell adhesion molecules (CAMs) mediate neurite outgrowth and growth cone repulsion. PTPμ is a growth permissive substrate for nasal retinal ganglion cell (RGC) neurites and a growth inhibitory substrate for temporal RGCs. In this manuscript, we demonstrate that the distinct PTPμ-dependent phenotypes of nasal outgrowth and temporal repulsion are regulated by Rho GTPases. The role of Rho GTPases in the regulation of nasal outgrowth and temporal repulsion was tested by utilizing dominant negative and constitutively active forms of Rac1, RhoA and Cdc42 in Bonhoeffer stripe assays. Nasal neurite outgrowth on PTPμ was blocked by Cdc42-DN. Temporal repulsion to a PTPμ substrate was substantially reduced by addition of Cdc42-DN. The molecule that regulates the switch between permissive versus repulsive responses to PTPμ is Rac1 for temporal neurons. Inhibition of Rac1 is required for repulsion of temporal neurons. Interestingly, adding Rac1-CA to temporal RGC neurons converted PTPμ-dependent repulsion to a permissive response. In addition, adding exogenous Rac1-DN to nasal neurons induced a phenotype switch from a permissive to repulsive response to PTPμ. Together these data suggest that Cdc42 activity is required for both permissive and repulsive responses to PTPμ. However, the key to PTPμ-dependent repulsion is inhibition of Rac1 activity in temporal RGC neurons.

Introduction

Spatiotemporal patterning of the visual system is a structured event in which retinal ganglion cell (RGC) axons from a particular region of the retina migrate and innervate a specific region of the optic tectum (McLaughlin et al., 2003, O'Leary and McLaughlin, 2005, Thanos and Mey, 2001, van Horck et al., 2004). For example, nasal axons innervate the posterior tectum, while temporal neurons innervate the anterior tectum. Studies have suggested that RGC projection to the tectum is partially due to the graded expression of Eph receptor tyrosine kinases and their ephrin ligands (McLaughlin et al., 2003, O'Leary and McLaughlin, 2005). Therefore, tyrosine phosphorylation is key to the development of the visual system but little is known about the function of tyrosine phosphatases in this system. Receptor tyrosine phosphatases (RPTPs), which catalyze the dephosphorylation of tyrosine residues, are expressed in the nervous system and recent evidence suggests that they may be involved in guiding retinal axons to their targets during development (Brady-Kalnay, 2001, Ensslen-Craig and Brady-Kalnay, 2004). A subfamily of RPTPs, including PTPμ, has cell adhesion molecule-like extracellular segments and intracellular domains with tyrosine phosphatase activity (Brady-Kalnay, 2001, Ensslen-Craig and Brady-Kalnay, 2004, Johnson and Van Vactor, 2003).

The PTPμ-like subfamily of RPTPs includes four members: PTPμ, PTPκ, PTPρ and PCP-2 (Brady-Kalnay, 2001). The PTPμ-like subfamily members contain motifs found in CAMs including a MAM domain, an immunoglobulin (Ig) domain and four FNIII repeats in their extracellular segment. The motifs present in PTPμ suggested that it might function in cell–cell adhesion. We demonstrated that expression of PTPμ induced the aggregation of nonadhesive Sf9 insect cells (Brady-Kalnay et al., 1993, Gebbink et al., 1993). PTPκ and PCP-2 have also been shown to mediate aggregation (Cheng et al., 1997, Sap et al., 1994). These studies demonstrated that the binding is homophilic (i.e. the “ligand” for a transmembrane PTP is an identical PTP molecule on an adjacent cell). Both PTPμ-dependent adhesion and neurite outgrowth are mediated by homophilic binding (Brady-Kalnay and Tonks, 1994, Ensslen-Craig and Brady-Kalnay, 2005). The homophilic binding site resides in the immunoglobulin domain (Brady-Kalnay and Tonks, 1994). The MAM domain of PTPμ also plays a role in cis dimerization (Aricescu et al., 2006, Cismasiu et al., 2004) and cell aggregation by sorting cells expressing PTPμ into distinct aggregates from cells expressing related molecules such as PTPκ (Zondag et al., 1995). Together, these studies demonstrate that PTPμ mediates cell–cell adhesion and suggest that it may transduce signals in response to adhesion that are required for neurite outgrowth.

The juxtamembrane domain of the PTPμ-like enzymes contains a region of homology to the intracellular domain of the cadherins (Brady-Kalnay, 2001). We demonstrated that PTPμ interacts specifically with N-cadherin, E-cadherin and R-cadherin (Brady-Kalnay et al., 1998). Both N- and R-cadherin are expressed in the retina and control retinal histogenesis (Redies, 1997, Redies and Takeichi, 1996). Importantly, we have demonstrated a functional role for the PTPμ/N-cadherin interaction because we found that PTPμ regulates N-cadherin-dependent neurite outgrowth of retinal ganglion cells (Burden-Gulley and Brady-Kalnay, 1999).

PTPμ is expressed in a gradient in both the retina and the tectum (Burden-Gulley et al., 2002). Since the axons of RGCs form the optic nerve and are the sole output from the retina to the brain, the expression of PTPμ on these cells was consistent with a putative role for PTPμ in axonal migration. The ability of retinal ganglion cell axons to grow out of a retinal explant onto a purified protein substrate has been used to study axonal (neurite) growth (Lemmon et al., 1992). To test whether PTPμ promoted neurite outgrowth, we used purified PTPμ as a substrate and demonstrated that it promoted neurite outgrowth from E8 retinal explants (Burden-Gulley and Brady-Kalnay, 1999). More importantly, PTPμ is a permissive substrate for neurite outgrowth from nasal RGC neurons, while it is inhibitory to temporal RGC neurons (Burden-Gulley et al., 2002). The molecular details of the intracellular signals that regulate axonal growth and guidance are not well defined.

It is intriguing that PTPμ is able to mediate both permissive and repulsive responses in RGC neurons. PTPμ is expressed at low levels in nasal neurons and is permissive for nasal RGC neurite outgrowth (Burden-Gulley et al., 2002). PTPμ is expressed at high levels in temporal RGCs and is repulsive to those neurons (Burden-Gulley et al., 2002). Our recent data suggest that the gradient of PTPμ expression in the retina regulates whether PTPμ is a repulsive or permissive guidance cue (Ensslen-Craig and Brady-Kalnay, 2005). We found that expression of exogenous PTPμ in nasal neurites, which increases PTPμ levels to those found in temporal neurons, resulted in a phenotypic switch from a permissive to a repulsive response to a PTPμ substrate (Ensslen-Craig and Brady-Kalnay, 2005). Catalytic activity of PTPμ is necessary for both permissive and repulsive guidance events downstream of PTPμ binding. Since both expression and catalytic activity of PTPμ are required, this implies that PTPμ homophilic binding results in a tyrosine phosphatase-dependent signal that is necessary for PTPμ-mediated nasal outgrowth and temporal repulsion, adding support to the growing body of evidence that RPTP catalytic activity is important for axon guidance (Garrity et al., 1999, Johnson et al., 2001). Most importantly, these results demonstrate a functional significance for the observed gradients in retinotectal expression of PTPμ. In this manuscript, we investigate the molecular mechanisms by which PTPμ regulates both nasal outgrowth and temporal repulsion.

The Rho subfamily of small G-proteins plays a central role in regulating the actin cytoskeleton (Raftopoulou and Hall, 2004, Ridley, 2004). Activation of Cdc42 results in the formation of filopodia while Rac activation induces lamellipodia formation, and Rho activation results in stress fibers in fibroblasts (Nobes and Hall, 1995). Rho GTPases are molecular switches, cycling between GTP-bound “on” and GDP-bound “off” states. This cycle is controlled by GTPase activating proteins (GAPs), guanine nucleotide exchange factors (GEFs) and GDP-dissociation inhibitors (Bishop and Hall, 2000, Kaibuchi et al., 1999).

The role of Rho GTPase activation in the regulation of nasal outgrowth and temporal repulsion was tested by utilizing dominant negative and constitutively active TAT-tagged forms of Rac1, RhoA and Cdc42, which were directly taken up by cells. We prepared alternating stripes of PTPμ and laminin as substrates for single stripe assays and a combination of PTPμ mixed with laminin versus laminin only lanes for the mixed stripe assays. Single stripes measure permissive responses, i.e. the ability to grow or not on one adhesion molecule. Mixed stripes measure repulsive responses because a repulsive substrate-like PTPμ is mixed with a normally permissive cue, such as laminin. The TAT-Rho GTPase fusion proteins were added to the stripe assays and changes in nasal outgrowth or temporal crossing were observed. Nasal neurite outgrowth and temporal repulsion to a PTPμ substrate were substantially reduced by the addition of Cdc42-DN, while inhibition of Rac1 is required for temporal repulsion to PTPμ response. These data suggest that the key to PTPμ-dependent repulsion is inhibition of Rac1 activity in temporal RGC neurons.

Section snippets

Results

We cultured retinal explants from embryonic day 8 chicks in Bonhoeffer stripe assays (Vielmetter et al., 1990, Walter et al., 1987a, Walter et al., 1987b). The Bonhoeffer stripe assay examines the ability of neurons to grow on two protein substrates presented in alternating lanes. A permissive substrate must be present in a lane to observe any neurite outgrowth on that lane (Lemmon et al., 1992). Very few adhesion molecules promote neurite outgrowth of chick retinal ganglion cell (RGC) neurons (

Discussion

In this manuscript, we tested whether the Rho GTPases regulate PTPμ-dependent nasal outgrowth and temporal repulsion. The data presented here indicate that Cdc42 activity is required for both nasal outgrowth and temporal repulsion (Fig. 10), which is consistent with our previous study that demonstrated a requirement of Cdc42 activity for PTPμ-dependent growth cone rearrangement (Rosdahl et al., 2003). In the previous study, the stimulation of E6 retinal cultures with purified PTPμ resulted in a

Culturing of chick retinal explants

Chick embryonic day 8 embryos were staged according to Hamburger and Hamilton (Hamburger and Hamilton, 1951). Retinal explants were prepared as described (Drazba and Lemmon, 1990, Halfter et al., 1983). E8 (stage 32) neural retinas were dissected, flattened on concanavalin-coated nitrocellulose filters and cut into 350 μm wide explants. Explants were placed retinal ganglion side down on substrate-coated dishes and grown in 10% fetal bovine serum (Hyclone, Logan, UT), 2% chick serum 4(Sigma, St.

Acknowledgments

This research was supported by a grant from the National Institutes of Health Grant R01-EY12251 (S.B.K). Additional support was obtained from the Visual Sciences Research Center Core Grant PO-EY11373 from the National Eye Institute. We thank Dr. Steven Dowdy for providing the TAT-Rho GTPase fusion proteins. We thank Carol Luckey for technical support including preparation and purification of the TAT-fusion Rho GTPase proteins and members of the Brady-Kalnay lab for insightful discussions.

References (91)

  • S.E. Ensslen et al.

    PTPmu signaling via PKCdelta is instructive for retinal ganglion cell guidance

    Mol. Cell. Neurosci.

    (2004)
  • S.E. Ensslen-Craig et al.

    Receptor protein tyrosine phosphatases regulate neural development and axon guidance

    Dev. Biol.

    (2004)
  • S.E. Ensslen-Craig et al.

    PTPmu expression and catalytic activity are required for PTPmu-mediated neurite outgrowth and repulsion

    Mol. Cell. Neurosci.

    (2005)
  • J.W. Erickson et al.

    Identification of an actin cytoskeletal complex that includes IQGAP and the Cdc42 GTPase

    J. Biol. Chem.

    (1997)
  • M. Fukata et al.

    Rac1 and Cdc42 capture microtubules through IQGAP1 and CLIP-170

    Cell

    (2002)
  • P.A. Garrity et al.

    Retinal axon target selection in Drosophila is regulated by a receptor protein tyrosine phosphatase

    Neuron

    (1999)
  • M.F.B.G. Gebbink et al.

    Cell-cell adhesion mediated by a receptor-like protein tyrosine phosphatase

    J. Biol. Chem.

    (1993)
  • E. Giniger

    How do Rho family GTPases direct axon growth and guidance? A proposal relating signaling pathways to growth cone mechanics

    Differentiation

    (2002)
  • F. Haj et al.

    Retinotectal ligands for the receptor tyrosine phosphatase CRYPalpha

    Mol. Cell. Neurosci.

    (1999)
  • W. Halfter et al.

    Oriented axon outgrowth from avian embryonic retinae in culture

    Dev. Biol.

    (1983)
  • K. Kaibuchi et al.

    Regulation of cadherin-mediated cell-cell adhesion by the Rho family GTPases

    Curr. Opin. Cell Biol.

    (1999)
  • N.X. Krueger et al.

    The transmembrane tyrosine phosphatase DLAR controls motor axon guidance in Drosophila

    Cell

    (1996)
  • S. Kuroda et al.

    Identification of IQGAP as a putative target for the small GTPases, Cdc42 and Rac1

    J. Biol. Chem.

    (1996)
  • Z. Li et al.

    IQGAP1 and calmodulin modulate E-cadherin function

    J. Biol. Chem.

    (1999)
  • Z. Li et al.

    IQGAP1 promotes neurite outgrowth in a phosphorylation-dependent manner

    J. Biol. Chem.

    (2005)
  • S.C. Mateer et al.

    The mechanism for regulation of the F-actin binding activity of IQGAP1 by calcium/calmodulin

    J. Biol. Chem.

    (2002)
  • T. McLaughlin et al.

    Regulation of axial patterning of the retina and its topographic mapping in the brain

    Curr. Opin. Neurobiol.

    (2003)
  • K. Mikule et al.

    Eicosanoid activation of protein kinase C epsilon: involvement in growth cone repellent signaling

    J. Biol. Chem.

    (2003)
  • M. Nikolic

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

    Int. J. Biochem. Cell Biol.

    (2002)
  • D.D. O'Leary et al.

    Mechanisms of retinotopic map development: Ephs, ephrins, and spontaneous correlated retinal activity

    Prog. Brain Res.

    (2005)
  • J.C. Perron et al.

    Distinct neurite outgrowth signaling pathways converge on ERK activation

    Mol. Cell. Neurosci.

    (1999)
  • P.J. Phillips-Mason et al.

    The receptor protein tyrosine phosphatase PTPμ interacts with IQGAP1

    J. Biol. Chem.

    (2006)
  • M. Raftopoulou et al.

    Cell migration: Rho GTPases lead the way

    Dev. Biol.

    (2004)
  • C. Redies et al.

    Cadherins in the developing central nervous system: an adhesive code for segmental and functional subdivisions

    Dev. Biol.

    (1996)
  • J.A. Rosdahl et al.

    Protein kinase C delta (PKCdelta) is required for protein tyrosine phosphatase mu (PTPmu)-dependent neurite outgrowth

    Mol. Cell. Neurosci.

    (2002)
  • M. Roy et al.

    IQGAP1 binds ERK2 and modulates its activity

    J. Biol. Chem.

    (2004)
  • S.D. Skaper et al.

    Cell signalling cascades regulating neuronal growth-promoting and inhibitory cues

    Prog. Neurobiol.

    (2001)
  • N. Soga et al.

    Rho family GTPases regulate VEGF-stimulated endothelial cell motility

    Exp. Cell Res.

    (2001)
  • Q.L. Sun et al.

    Growth cone steering by receptor tyrosine phosphatase delta defines a distinct class of guidance cue

    Mol. Cell. Neurosci.

    (2000)
  • J.M. Swart-Mataraza et al.

    IQGAP1 is a component of Cdc42 signaling to the cytoskeleton

    J. Biol. Chem.

    (2002)
  • S. Thanos et al.

    Development of the visual system of the chick. II. Mechanisms of axonal guidance

    Brain Res. Rev.

    (2001)
  • J. Tong et al.

    Manipulation of EphB2 regulatory motifs and SH2 binding sites switches MAPK signaling and biological activity

    J. Biol. Chem.

    (2003)
  • F.P. van Horck et al.

    Retinal axon guidance: novel mechanisms for steering

    Curr. Opin. Neurobiol.

    (2004)
  • J. Wang et al.

    Receptor tyrosine phosphatase-delta is a homophilic, neurite-promoting cell adhesion molecule for CNS neurons

    Mol. Cell. Neurosci.

    (1999)
  • L. Weissbach et al.

    Identification of a human rasGAP-related protein containing calmodulin-binding motifs

    J. Biol. Chem.

    (1994)
  • Cited by (10)

    • Regulation of development and cancer by the R2B subfamily of RPTPs and the implications of proteolysis

      2015, Seminars in Cell and Developmental Biology
      Citation Excerpt :

      Tyrosine phosphatase activity of PTPμ is necessary for its regulation of neurite outgrowth [42]. Neurite outgrowth on a purified PTPμ substrate requires signaling via PLCγ1, PKCδ, the Rho GTPases Cdc42 and Rac1, and IQGAP1 [43–48]. Despite its extensive expression in the nervous system, a definitive role for PTPκ during neural development has yet to be demonstrated.

    • Cytoskeleton dynamics in the retina

      2014, Critical Reviews in Eukaryotic Gene Expression
    View all citing articles on Scopus
    View full text