Nogo limits neural plasticity and recovery from injury

https://doi.org/10.1016/j.conb.2014.02.011Get rights and content

Highlights

  • Nogo-A limits neural repair and recovery after adult CNS injuries.

  • NgR1 and S1PR2 are receptors for different domains of Nogo-A.

  • Nogo-A and NgR1 limit experience-dependent neural plasticity.

  • Nogo-A and NgR1 stabilize synaptic, dendritic and axonal anatomy in the adult CNS.

The expression of Nogo-A and the receptor NgR1 limits the recovery of adult mammals from central nervous system injury. Multiple studies have demonstrated efficacy from targeting this pathway for functional recovery and neural repair after spinal cord trauma, ischemic stroke, optic nerve injury and models of multiple sclerosis. Recent molecular studies have added S1PR2 as a receptor for the amino terminal domain of Nogo-A, and have demonstrated shared components for Nogo-A and CSPG signaling as well as novel Nogo antagonists. It has been recognized that neural repair involves plasticity, sprouting and regeneration. A physiologic role for Nogo-A and NgR1 has been documented in the restriction of experience-dependent plasticity with maturity, and the stability of synaptic, dendritic and axonal anatomy.

Introduction

The longitudinal growth of nerve fibers, the regeneration of injured axons and the structural plasticity of axons and dendrites are confined to very short distances and limited spatial dimensions in the adult mammalian central system (CNS). The successful regeneration of adult CNS axons of multiple origins into peripheral nerve grafts placed into spinal cord, brain or optic nerve over centimeter distances emphasized the key role of factors from the local tissue microenvironment in determining the extent of growth [1]. Twenty years ago, specific neurite growth inhibitory factors, many of which were enriched in myelin, were discovered. Nogo-A, the myelin proteins, MAG and OMgp, several semaphorins and ephrins as well as chondroitin sulphate proteoglycans have been identified [2]. For many of these molecules the detailed expression pattern in the adult CNS, the possible interplay between single factors, as well as their in vivo roles in the intact or injured adult CNS are not well characterized yet. More information is available for the membrane protein Nogo-A; some key concepts and the most recent literature on Nogo-A will be summarized in this short review.

Section snippets

Nogo-A interacts with a multisubunit receptor complex

Fragment analyses and binding studies showed that the 1200 aa protein Nogo-A contains more than one growth inhibiting domain (Figure 1). The 66 aa extracellular loop between the two C-terminal intramembrane segments binds to a GPI-linked, LRR-containing membrane protein called NgR1. Signaling is induced by a receptor complex containing the membrane proteins p75 and/or Troy and the LRR-protein Lingo1 [3, 4, 5]. An alternative Nogo-66 receptor is PirB. PirB is expressed at low or undetectable

Nogo-A destabilizes the cytoskeleton and suppresses the cellular growth program

Growth cones collapse and neurite elongation stops upon contact with Nogo-A in a rho/ROCK dependent way. Interestingly, internalization of Nogo-A was shown to be required for these effects in growth cones of hippocampal neurons [16]. Integrin function is also affected by Nogo-A [11], and activation of integrins can overcome the Nogo-A mediated growth inhibition [17]. The actin cytoskeleton-based lamellipodia and filopodia of growth cones are affected at an early stage of the collapse.

Suppression of Nogo-A/Nogo receptor signaling enhances repair after CNS injury

Spinal cord injuries affecting up to half of the spinal cord diameter or strokes destroying only part of the motor cortex often have a good prognosis with substantial spontaneous functional recovery in animals and humans. Compensatory sprouting of intact fiber systems and formation of new circuits including indirect ‘detour’ pathways are currently emerging as important mechanisms underlying these functional recovery processes [22, 23, 24]. The factors which trigger and guide growing fibers,

Nogo-A and NgR1 signaling titrates experience-dependent plasticity

It is abundantly clear that interruption of the Nogo-A to Nogo-66 receptor pathway increases functional recovery after injury by a combination of neuronal plasticity, short range sprouting and axonal regeneration. The observation of anatomical rearrangements far from the injury site and by uninjured pathways raises the question of the natural role of this Nogo pathway for anatomical stability in the adult nervous system (Figure 2).

It is well appreciated that experience-dependent neural

Stability of adult synaptic anatomy is dependent on Nogo-A and NgR1 signaling

A broad range of studies have implicated Nogo-A and NgR1 in determining the formation and stability of synaptic morphology over longer time scales. In hippocampal slices, post-synaptic dendritic spine architecture, as well as axonal length, was found to depend on Nogo-A and NgR1 signaling [20]. Interruption of Nogo-A or NgR1 expression produced more immature appearing dendritic spines. Genetic deletion of NgR1, 2 and 3 demonstrated a key role for these proteins as a brake on synaptogenesis

Behavioral consequences of Nogo-A and NgR1 signaling

Without Nogo-A or NgR1 signaling, adolescent critical periods for experience dependent plasticity remain open and the recovery from CNS injury is enhanced. Are these data associated with altered behavior? One clue may come from the ability of high levels of neuronal excitation to suppress NgR1 levels [62•, 65], perhaps allowing anatomical plasticity to follow periods of hyperexcitation. In a study of mice expressing elevated levels of NgR1 [65], learning was intact but lasting memory formation

Conclusions

The Nogo-A and NgR1 signaling cascade have been studied extensively with regard to neurological trauma. Blockade of these pathways leads to greater functional recovery through a combination of neural plasticity, sprouting and axonal regeneration. The benefits of interrupting Nogo-A or NgR1 are observed in a broad spectrum of preclinical models and are beginning clinical evaluation. Recent molecular studies have demonstrated a signaling pathway for a second domain of Nogo-A that involves the

Conflict of interest

SMS is a co-founder of Axerion Therapeutics, seeking to develop NgR-based and PrP-based therapeutics.

References and recommended reading

Papers of particular interest, published within the period of review, have been highlighted as:

  • • of special interest

  • •• of outstanding interest

Acknowledgements

We acknowledge support from the National Institutes of Health and the Falk Medical Research Trust to SMS, and from the Swiss National Science Foundation and the Christopher and Dana Reeve Foundation to MES.

References (69)

  • M. Nash et al.

    Central nervous system regeneration inhibitors and their intracellular substrates

    Mol Neurobiol

    (2009)
  • M.E. Schwab

    Functions of nogo proteins and their receptors in the nervous system

    Nat Rev Neurosci

    (2010)
  • X. Gou et al.

    Spatio-temporal expression of paired immunoglobulin-like receptor-b in the adult mouse brain after focal cerebral ischaemia

    Brain Injury

    (2013)
  • O. Chivatakarn et al.

    The nogo-66 receptor ngr1 is required only for the acute growth cone-collapsing but not the chronic growth-inhibitory actions of myelin inhibitors

    J Neurosci

    (2007)
  • Y. Sato et al.

    Cartilage acidic protein-1b (lotus), an endogenous nogo receptor antagonist for axon tract formation

    Science

    (2011)
  • T.L. Dickendesher et al.

    Ngr1 and ngr3 are receptors for chondroitin sulfate proteoglycans

    Nat Neurosci

    (2012)
  • F. Hu et al.

    The n-terminal domain of nogo-a inhibits cell adhesion and axonal outgrowth by an integrin-specific mechanism

    J Neurosci

    (2008)
  • A. Kempf et al.

    The sphingolipid receptor s1pr2 is a receptor for nogo-a repressing synaptic plasticity

    PLoS Biol

    (2013)
  • E. Grünewald et al.

    Gpr50 interacts with neuronal nogo-a and affects neurite outgrowth

    Mol Cell Neurosci

    (2009)
  • N. Nakaya et al.

    Olfactomedin 1 interacts with the nogo a receptor complex to regulate axon growth

    J Biol Chem

    (2012)
  • T.L. Stiles et al.

    Ldl receptor-related protein-1 is a sialic-acid-independent receptor for myelin-associated glycoprotein that functions in neurite outgrowth inhibition by mag and cns myelin

    J Cell Sci

    (2013)
  • X. Peng et al.

    Neuronal nogo-a regulates glutamate receptor subunit expression in hippocampal neurons

    J Neurochem

    (2011)
  • L. Montani et al.

    Neuronal nogo-a modulates growth cone motility via rho-gtp/limk1/cofilin in the unlesioned adult nervous system

    J Biol Chem

    (2009)
  • M. Zagrebelsky et al.

    Nogo-a stabilizes the architecture of hippocampal neurons

    J Neurosci

    (2010)
  • L.M. Craveiro et al.

    Neutralization of the membrane protein nogo-a enhances growth and reactive sprouting in established organotypic hippocampal slice cultures

    Eur J Neurosci

    (2008)
  • F.M. Bareyre et al.

    The injured spinal cord spontaneously forms a new intraspinal circuit in adult rats

    Nat Neurosci

    (2004)
  • R. van den Brand et al.

    Restoring voluntary control of locomotion after paralyzing spinal cord injury

    Science

    (2012)
  • K. Liu et al.

    Neuronal intrinsic mechanisms of axon regeneration

    Annu Rev Neurosci

    (2011)
  • Y.K. Pun Raymund et al.

    Excessive activation of mtor in postnatally generated granule cells is sufficient to cause epilepsy

    Neuron

    (2012)
  • D. Akhavan et al.

    Mtor signaling in glioblastoma: lessons learned from bench to bedside

    Neuro-Oncology

    (2010)
  • B. Zorner et al.

    Anti-nogo on the go: from animal models to a clinical trial

    Ann N Y Acad Sci

    (2010)
  • W.B. Cafferty et al.

    Axonal growth therapeutics: regeneration or sprouting or plasticity?

    Trends Neurosci

    (2008)
  • W.B. Cafferty et al.

    Mag and omgp synergize with nogo-a to restrict axonal growth and neurological recovery after spinal cord trauma

    J Neurosci

    (2010)
  • J.K. Lee et al.

    Combined genetic attenuation of myelin and semaphorin-mediated growth inhibition is insufficient to promote serotonergic axon regeneration

    J Neurosci

    (2010)
  • Cited by (286)

    View all citing articles on Scopus
    View full text