Elsevier

Neuroscience Letters

Volume 652, 23 June 2017, Pages 56-63
Neuroscience Letters

Review article
The role of cAMP and its downstream targets in neurite growth in the adult nervous system

https://doi.org/10.1016/j.neulet.2016.12.033Get rights and content

Abstract

Injured neurons in the adult mammalian central nervous system (CNS) have a very limited capacity for axonal regeneration and neurite outgrowth. This inability to grow new axons or to regrow injured axons is due to the presence of molecules that inhibit axonal growth, and age related changes in the neuron’s innate growth capabilities. Available levels of cAMP are thought to have an important role in linking both of these factors. Elevated levels of cAMP in the developing nervous system are important for the guidance and stability of growth cones. As the nervous system matures, cAMP levels decline and the growth promoting effects of cAMP diminish. It has frequently been demonstrated that increasing neuronal cAMP can enhance neurite growth and regeneration. Some methods used to increase cAMP include administration of cAMP agonists, conditioning lesions, or electrical stimulation. Furthermore, it has been proposed that multiple stages of cAMP induced growth exist, one directly caused by its downstream effector Protein Kinase A (PKA) and one caused by the eventual upregulation of gene transcription.

Although the role cAMP in promoting axon growth is well accepted, the downstream pathways that mediate cAMP-mediated axonal growth are less clear. This is partly because various key studies that explored the link between PKA and axonal outgrowth relied on the PKA inhibitors KT5720 and H89. More recent studies have shown that both of these drugs are less specific than initially thought and can inhibit a number of other signalling molecules including the Exchange Protein Activated by cAMP (EPAC). Consequently, it has recently been shown that a number of intracellular signalling pathways previously attributed to PKA can now be attributed solely to activation of EPAC specific pathways, or the simultaneous co-activation of PKA and EPAC specific pathways. These new studies open the door to new potential treatments for repairing the injured spinal cord.

Introduction

Ever since David and Aguayo [1] first demonstrated that injured central nervous system (CNS) axons can grow into a peripheral nerve graft, a major question has been: why do CNS axons not regenerate within the injured CNS? Three streams of research tackled this question. One stream examined whether the lack of CNS regeneration was due to an inhibitory environment. This research later identified inhibitory factors for neurite outgrowth including myelin associated inhibitors [2], and chondroitin sulfate proteoglycans (CSPGs) in the glial scar and the perineuronal net [3], [4]. The second stream examined whether reduced CNS regeneration was due to a lack of neurotrophic support resulting in the neurons not having the ability to regrow, as proposed by Aguayo himself [1]. The third stream examined the question of whether age related changes might influence the decline in neuronal growth abilities [5], [6]. These streams of research led to three main treatment strategies. Some laboratories attempted to neutralize neurite growth inhibitors (e.g., MAG [7] or Nogo [8], [9];, and CSPGs [10], [11]), while others augmented neurotrophic support by adding growth factors like brain-derived neurotrophic factor (BDNF), neurotrophin-3 (NT-3), or glial cell-derived neurotrophic factor (GDNF), to the injured spinal cord [12], [13], [14], [15], [16] (Fig. 1). Alternatively, some groups attempted to modulate intracellular signalling pathways with the idea to recreate conditions of the developing CNS in order to allow injured neurons to regrow [5], [6], [12], [17], [18]. As a result of all of this research, it is now well accepted that a combination of all of three factors (growth inhibitors, in sufficient of growth promoting factors and developmental changes) are responsible for the limited neurite outgrowth and repair after a spinal cord injury (SCI) in adult mammals.

With many intrinsic and extrinsic factors converging to prevent regeneration (growth at the tip of lesioned axons) and plasticity (e.g., growth of new collaterals) after SCI, there is a major effort to identify whether there is a common cell signalling pathway responsible for inhibiting neurite growth. This effort has resulted in the knowledge that both the intrinsic and extrinsic factors inhibiting axonal growth after SCI seem to converge on a number of signalling pathways associated with cAMP, calcium/calmodulin, Ras homolog gene family member A (RhoA), and the mechanistic target of rapamycin (mTOR). A number of recent reviews have focused on roles of RhoA [18], [19] and mTOR [6], [20] in axon growth and regeneration; this review will focus on cAMP and its downstream effectors. Specifically, we will discuss the relationship between cAMP and neurite outgrowth in the adult CNS, recently identified cAMP cell signalling pathways linked to axonal growth after SCI, and possible treatment strategies aimed to activate relevant cAMP dependent pathways.

Section snippets

The role of cAMP in neurite growth

In the embryonic CNS, cAMP levels are high and are linked to the extensive axonal growth that occurs during development [5]. It has been shown that not long after birth, cAMP levels in dorsal root ganglia (DRG) neurons rapidly decline in parallel with the reduced regenerative capacity of their CNS axons [5], [21]. Similarly, blocking protein kinase A (PKA), a downstream effector of cAMP, using the PKA antagonists KT5720 and Rp-cAMPS in cultured rat DRG and retinal ganglion (RG) neurons resulted

Neurotrophins and cAMP

Neurotrophins are important signalling molecules for cell survival and neurite outgrowth. They can promote axon growth even in adult neurons exposed to myelin-associated inhibitors. For example, cerebellar and DRG neurons plated with the neurotrophins BDNF or neurite growth factor (NGF) prior to exposure to MAG and/or myelin, have increased neurite outgrowth compared to neurons without neurotrophins [22]. Dougherty et al. [23] further explored neurotrophin expression in the spinal cord and

Elevating cAMP levels to promote neurite outgrowth

With the knowledge that elevated cAMP levels could promote neurite outgrowth in the inhibitory environment of the injured adult mammalian CNS, it was a logical next step to target cAMP and other parts of its signalling pathway to promote regeneration. Neumann and colleagues [36] demonstrated the pro-regenerative effect of high cAMP levels by injecting a cAMP analog (db-cAMP) in DRGs in vivo, thus activating downstream effectors including PKA, and CREB (see Fig. 1) and promoting regeneration of

Raising cAMP using electrical stimulation

The importance of cAMP in the regulation of neurite outgrowth and myelin-associated inhibition is illustrated even further by the finding that cAMP levels drop in the spinal cord, brainstem, and cortex following SCI [39], consistent with lack of axonal regeneration. A possible reason for the post injury decline in cAMP is the disuse of affected limbs and the associated inactivity of neurons within the involved circuitry. This is supported by the finding that directly following unilateral SCI in

cAMP signalling

High neuronal cAMP levels are linked to increased neurite outgrowth in both permissive and inhibitory environments, but the mechanisms by which cAMP increases neuronal growth have yet to be fully understood. A well-known downstream effector of cAMP is PKA (Fig. 1). Various studies have shown that blocking PKA using KT5720 eliminates the growth promoting effects of increased cAMP levels [5], [12], [17], [37], [54], [55] (Fig. 1). Qiu et al. [37], showed that there are two phases of regeneration,

Exchange protein activated by cAMP

EPAC is activated by cAMP independently of PKA (Fig. 1) and likely a key factor in cAMP induced neurite outgrowth, as well as many other aspects of cAMP signalling in other areas of the body that had been previously thought to be the result of PKA activation [64]. Two isoforms have been described, EPAC1 and EPAC2, with varying expression levels and tissue distributions throughout development [65]. EPAC1 is highly expressed in embryonic nerve cells while EPAC2 is expressed mostly in developed

Conclusion

Numerous studies suggest that increasing cAMP levels can increase neurite outgrowth and could be effective as both short and long-term treatment option for SCI. However, due to cAMP being an important and ubiquitous signalling molecule throughout the body, direct activation of cAMP would likely cause numerous adverse side effects. Thus, the understanding of how to specifically activate downstream targets is an important step in the development of clinically relevant treatments. As more accurate

References (71)

  • K.D. Dougherty et al.

    Brain-derived neurotrophic factor in astrocytes, oligodendrocytes, and microglia/macrophages after spinal cord injury

    Neurobiol. Dis.

    (2000)
  • T.D. Hernandez et al.

    Seizures and recovery from experimental brain damage

    Exp. Neurol.

    (1988)
  • E. Kunkel-Bagden

    Methods to assess the development and recovery of locomotor function after spinal cord injury in rats

    Exp. Neurol.

    (1993)
  • A. Krajacic et al.

    Training-induced plasticity in rats with cervical spinal cord injury: effects and side effects

    Behav. Brain Res.

    (2010)
  • B.I. Awad et al.

    Potential role of growth factors in the management of spinal cord injury

    World Neurosurg.

    (2015)
  • A.R. Harvey et al.

    Neurotrophic factors for spinal cord repair: which, where, how and when to apply, and for what period of time?

    Brain Res.

    (2015)
  • N. Weishaupt et al.

    BDNF: The career of a multifaceted neurotrophin in spinal cord injury

    Exp. Neurol.

    (2012)
  • S. Neumann et al.

    Regeneration of sensory axons within the injured spinal cord induced by intraganglionic cAMP elevation

    Neuron

    (2002)
  • J. Qiu et al.

    Spinal axon regeneration induced by elevation of cyclic AMP

    Neuron

    (2002)
  • S. Neumann et al.

    Regeneration of dorsal column fibers into and beyond the lesion site following adult spinal cord injury

    Neuron

    (1999)
  • T. Gordon

    Electrical stimulation to enhance axon regeneration after peripheral nerve injuries in animal models and humans

    Neurotherapeutics

    (2016)
  • N.M. Geremia et al.

    Electrical stimulation promotes sensory neuron regeneration and growth-associated gene expression

    Exp. Neurol.

    (2007)
  • E. Udina et al.

    Electrical stimulation of intact peripheral sensory axons in rats promotes outgrowth of their central projections

    Exp. Neurol.

    (2008)
  • D. Cai et al.

    Prior exposure to neurotrophins blocks inhibition of axonal regeneration by MAG and myelin via a cAMP-dependent mechanism

    Neuron

    (1999)
  • G. Ming et al.

    CAMP-dependent growth cone guidance by netrin-1

    Neuron

    (1997)
  • T. Ye et al.

    Cyclin-dependent kinase 5 in axon growth and regeneration, in

    Int. Rev. Neurobiol. Elsevier

    (2012)
  • M.M. Siddiq et al.

    Metallothionein-I/II promotes axonal regeneration in the central nervous system

    J. Biol. Chem.

    (2015)
  • M. Domeniconi et al.

    Myelin-associated glycoprotein interacts with the Nogo66 receptor to inhibit neurite outgrowth

    Neuron

    (2002)
  • A.J. Murray et al.

    Epac mediates cyclic AMP-dependent axon growth, guidance and regeneration

    Mol. Cell. Neurosci.

    (2008)
  • A.G. Peace et al.

    New perspectives in cyclic AMP-mediated axon growth and guidance: the emerging epoch of Epac

    Brain Res. Bull.

    (2011)
  • D. Wei et al.

    Inhibiting cortical protein kinase a in spinal cord injured rats enhances efficacy of rehabilitative training

    Exp. Neurol.

    (2016)
  • S. David et al.

    Axonal elongation into peripheral nervous system bridges after central nervous system injury in adult rats

    Science

    (1981)
  • M.E. Schwab et al.

    Degeneration and regeneration of axons in the lesioned spinal cord

    Physiol. Rev.

    (1996)
  • V.J. Tom et al.

    Studies on the development and behavior of the dystrophic growth cone, the hallmark of regeneration failure, in an in vitro model of the glial scar and after spinal cord injury

    J. Neurosci.

    (2004)
  • D. Cai et al.

    Neuronal cyclic AMP controls the developmental loss in ability of axons to regenerate

    J. Neurosci.

    (2001)
  • Cited by (67)

    View all citing articles on Scopus
    View full text