p75NTR is an obligate signaling receptor required for cues that cause sympathetic neuron growth cone collapse

https://doi.org/10.1016/j.mcn.2010.05.015Get rights and content

Abstract

The p75 neurotrophin receptor (p75NTR) is required for the activity of growth cone collapsing factors such as Nogo, MAG, OMgP, and ephrin A. Specifically, p75NTR associates with the Nogo receptor and GPI-linked ephrin A, and unliganded p75NTR mediates the biological effects of those proteins. Here we assess the requirement for p75NTR for the growth cone collapsing responses of semaphorins (Sema) 3A and 3F and ephrin B2 in sympathetic neurons. We show that the ability of Sema 3s or ephrin B2 to collapse growth cones is suppressed in p75NTR−/− sympathetic neurons. Ectopic expression of p75NTR restores the collapsing activity of Sema 3 in p75NTR−/− neurons. Moreover, p75NTR must be bound to its neurotrophin ligands to participate in Sema 3-mediated collapse. Ligand-bound p75NTR participates in Sema 3 and ephrin B2-mediated collapse via the Rho signaling pathway, since inhibition of Rho signaling is sufficient to suppress the effects of Sema 3s and ephrin B2 in p75NTR+/+ but not p75NTR−/− neurons. Our data suggest that in addition to its role as a co-receptor, p75NTR may provide an obligate parallel neurotrophin-activated inhibitory pathway that broadly sensitizes neurons to inhibitory cues.

Introduction

During development, axons are guided by both positive and negative cues that they encounter as they pathfind their way to their targets. These cues bind to receptors on axonal growth cones and newly-formed axons, where they are integrated to culminate in either stabilization or retraction of actin filaments and microtubules, thereby determining the extent and directionality of axonal growth. However, while significant progress has been made in defining the attractive and repulsive cues that are responsible for axon guidance, much less is understood about the way that growth cones and axons integrate the different classes of cues that they encounter.

In the peripheral nervous system, perhaps the best-characterized growth and guidance cues are the neurotrophins. In addition to their well-established roles in peripheral neuron survival and growth (Miller & Kaplan, 2001a, Miller & Kaplan, 2001b, Zhou & Snider, 2006), the neurotrophins regulate peripheral axon pathfinding, target innervation, and pruning (Kohn et al., 1999, Singh et al., 2008). The neurotrophins mediate their positive effects on axonal growth and pathfinding by binding to the Trk neurotrophin receptors, which activate signaling via PI3-kinase and the MEK–ERK pathway to promote axonal growth and growth cone motility (Atwal et al., 2000, Atwal et al., 2003). In contrast, the neurotrophins inhibit axonal growth and cause degeneration of developing axons by binding to the p75 pan-neurotrophin receptor (p75NTR) (Singh et al., 2008), which mediates these effects via Rho-GDI (Yamashita and Tohyama, 2003) and by inhibiting signaling pathways downstream of the Trk receptors (Atwal et al., 2003). In this regard, while neurotrophin binding to p75NTR is sufficient on its own to inhibit axonal growth in some settings (Kohn et al., 1999), p75NTR also acts as an inhibitory co-receptor for other receptors. In particular, the Nogo receptor inhibits growth on myelin substrates by binding to p75NTR and activating the Rho pathway (Yamashita et al., 2005, Yamashita & Tohyama, 2003) and an ephrin A–p75NTR complex reverse signals to mediate retinal axon repulsion during development (Lim et al., 2008). In the latter study, ephrin A mediated its p75NTR-dependent responses via fyn, while both ephrin A and a second ephrin family member, ephrin B2, are thought to primarily signal via RhoA and Cdc42/Rac1, respectively (Cheng et al., 2003, Klein, 2004, Sahin et al., 2005).

These findings raise the intriguing possibility that p75NTR might function as a general and required receptor for growth-inhibitory cues and/or that it might represent an inhibitory signaling system that functions in parallel to sensitize axons to such negative cues. In this regard, two classes of cues well-known for their growth-inhibitory actions in the peripheral and central nervous system are the class 3 semaphorins (Sema 3s), which regulate cell migration, axonal growth and pathfinding, target selection, and axonal pruning (Roth et al., 2009), and B type ephrins which function in axonal guidance and cell migration (Klein, 2004). Sema 3s mediate these effects via a receptor complex comprised of binding subunits, the neuropilins, and signaling components, the Plexin A receptors (He & Tessier-Lavigne, 1997, Takahashi et al., 1999, Tamagnone et al., 1999, Yaron et al., 2005). Interestingly, a number of lines of evidence indicate that the semaphorins interact with the neurotrophins and/or their receptors to regulate peripheral axon growth and guidance. For example, we have previously shown that Sema 3F inhibits Trk signaling to Erk to cause sympathetic neuron growth cone collapse (Atwal et al., 2003) and Ben-Zvi et al. (2007) demonstrated that p75NTR antagonizes Sema 3A to inhibit growth cone collapse in embryonic sensory neurons. Similarly, ephrin B1 can suppress Ras–Erk signaling during neurite retraction mediated by its receptor EphB2 (Elowe et al., 2001).

Here, we have asked whether p75NTR acts in partnership with Sema 3s to inhibit collapse of growth cones, focusing upon peripheral sympathetic neurons. We also asked whether p75NTR is required for the activity of ephrin B2, which we identify as a novel collapsing factor for sympathetic neurons. We show that p75NTR is necessary for Sema 3 and ephrin B2-mediated growth cone collapse. Furthermore, collapse in response to the semaphorins and ephrin B2 requires neurotrophin-mediated activation of a p75NTR–Rho signaling pathway. These findings, together with previous observations that p75NTR signaling is also required for Nogo and ephrin A inhibitory signaling suggest that this receptor is a general co-receptor for ligand–receptor complexes with growth-inhibitory activity.

Section snippets

p75NTR is necessary for Sema 3-mediated collapse of sympathetic neurons

To ask whether p75NTR is required for neuronal responses to semaphorins, we compared the responses of p75NTR+/+ versus p75NTR−/− sympathetic neurons to semaphorins 3A or 3F (Sema 3A or 3F), both of which have been shown to cause growth cone collapse in sympathetic neurons (Atwal et al., 2003). We (Atwal et al., 2003) and others (Yaron et al., 2005) have previously shown that sympathetic neurons of the superior cervical ganglion (SCG) express the Sema 3A/3F receptors Neuropilins 1 and 2, and

Discussion

The data presented here support three major conclusions. First, our data indicate that two cues that collapse sympathetic neuron growth cones, Sema 3A and ephrin B2, require p75NTR to do so, since collapse was largely suppressed in p75NTR−/− sympathetic neurons, and reintroduction of p75NTR was sufficient to rescue collapse in response to Sema 3A. In this regard, while Sema 3A/F has previously been shown to collapse sympathetic neuron growth cones, this is the first report that ephrin B2 is

Analysis of transgenic mice

The Animal Care Committee of the Hospital for Sick Children approved all animal use in accordance with the policies established by the Canadian Council of Animal Care. Mice that were homozygous for a targeted mutation in the Ngfr gene (Lee et al., 1994) in a mixed C129–C57BL6 background were generated and maintained as described previously (Majdan et al., 2001, Singh et al., 2008).

Primary neuron cultures and reagents

Sympathetic neurons were dissected from superior cervical ganglia (SCG) of P1 WT or p75NTR−/− mice and processed as

Acknowledgments

S.N. and D.C.L. were supported by fellowships from the Heart and Stroke Foundation of Canada. This work was supported by a research grant from the Canadian Institute of Health Research (CIHR) to D.R.K. and F.D.M. (MGP-14446). D.R.K. and F.D.M. are Canada Research Chairs and F.D.M. is an H.H.M.I. International Research Scholar. We thank Dr. Phil Barker (McGill University) for the gift of the recombinant adenovirus expressing WT p75NTR and Dr. Tony Pawson (Samuel Lunenfeld Research Institute) for

References (41)

  • T. Takahashi et al.

    Plexin–neuropilin-1 complexes form functional semaphorin-3A receptors

    Cell

    (1999)
  • L. Tamagnone et al.

    Plexins are a large family of receptors for transmembrane, secreted, and GPI-anchored semaphorins in vertebrates

    Cell

    (1999)
  • A.R. Vaillant et al.

    Signaling mechanisms underlying reversible, activity-dependent dendrite formation

    Neuron

    (2002)
  • K.E. Waimey et al.

    Plexin-A3 and plexin-A4 restrict the migration of sympathetic neurons but not their neural crest precursors

    Dev. Biol.

    (2008)
  • A. Yaron et al.

    Differential requirement for Plexin-A3 and -A4 in mediating responses of sensory and sympathetic neurons to distinct class 3 semaphorins

    Neuron

    (2005)
  • H. Aizawa et al.

    Phosphorylation of cofilin by LIM-kinase is necessary for semaphorin 3A-induced growth cone collapse

    Nat. Neurosci.

    (2001)
  • J.K. Atwal et al.

    Semaphorin 3F antagonizes neurotrophin-induced phosphatidylinositol 3-kinase and mitogen-activated protein kinase kinase signaling: a mechanism for growth cone collapse

    J. Neurosci.

    (2003)
  • D.J. Belliveau et al.

    NGF and neurotrophin-3 both activate TrkA on sympathetic neurons but differentially regulate survival and neuritogenesis

    J. Cell Biol.

    (1997)
  • A. Ben-Zvi et al.

    Modulation of semaphorin 3A activity by p75 neurotrophin receptor influences peripheral axon patterning

    J. Neurosci.

    (2007)
  • R.B. Campenot et al.

    Effects of sphingosine, staurosporine, and phorbol ester on neurites of rat sympathetic neurons growing in compartmented cultures

    J. Neurosci.

    (1991)
  • Cited by (31)

    • Local vibration therapy promotes the recovery of nerve function in rats with sciatic nerve injury

      2022, Journal of Integrative Medicine
      Citation Excerpt :

      In addition to chemotaxis of macrophages, digestion of damaged tissues, and regulation of inflammatory mediators, SCs secrete a variety of neurotrophic factors (NTFs), such as NGF, which can be retrogradely transported into the cell body of the effector neuron through endocytosis of the regenerated axon cone to maintain neuronal activity, and regulate axon germination and chemotaxis [47–49]. The expression of P75NTR, a low-affinity receptor of NGF, is upregulated following various injuries and can be combined with various NTFs to regulate different signaling pathways, and mediate cell survival, differentiation, axonal growth and remyelination [50–52]. After PNI, rapid activation of signal pathways at the site of injury induces the expression of related proteins that are involved in regulating cell function and cytoskeletal dynamics [53,54].

    • A small molecule TrkB/TrkC neurotrophin receptor co-activator with distinctive effects on neuronal survival and process outgrowth

      2016, Neuropharmacology
      Citation Excerpt :

      The central nervous system (CNS) undergoes limited repair after injury, due in part to the presence of inhibitors of neurite outgrowth that prevent axonal regeneration, including myelin-associated glycoprotein (MAG) and chondroitin sulfate proteoglycans (CSPGs) (Berry, 1982; McKerracher et al., 1994; Niederost et al., 1999). NTs may or may not facilitate axonal growth in the presence of myelin-component inhibitors (Spencer et al., 2008; Wang et al., 2002; Williams et al., 2005) depending upon the interplay of outgrowth-supporting Trk-mediated signaling and growth cone collapse-promoting p75NTR-mediated signaling, and varying with cell type and physiologic state (Naska et al., 2010). A prior study found that a peptide designed to activate TrkB without effects on p75NTR could reverse MAG inhibition of cerebellar neurite outgrowth, while BDNF did not (Williams et al., 2005), suggesting that isolated stimulation of Trks may support process outgrowth in an inhibitory environment.

    • Identification of Drugs that Regulate Dermal Stem Cells and Enhance Skin Repair

      2016, Stem Cell Reports
      Citation Excerpt :

      Antibodies are described in the Supplemental Experimental Procedures. Secondary SKP spheres were dissociated and seeded at 106 cells/well in 6-well plates (Costar), cultured for 24 hr, treated as indicated, and analyzed on western blots as described (Naska et al., 2010). Densitometry was performed after subtracting the background using ImageJ software.

    • Estrogen and female reproductive tract innervation: Cellular and molecular mechanisms of autonomic neuroplasticity

      2015, Autonomic Neuroscience: Basic and Clinical
      Citation Excerpt :

      Interestingly, Sema3F can antagonize NGF-stimulated TrkA signaling in sympathetic neurons (Atwal et al., 2003). Moreover, p75NTR is required for growth cone collapsing responses of sympathetic neurons to Sema3F; and p75NTR has to be bound to neurotrophins to participate in Sema3-mediated collapse (Naska et al., 2010). This may be relevant because estrogen increases the expression of p75NTR in uterine-projecting sympathetic neurons (Richeri et al., 2005).

    View all citing articles on Scopus
    View full text