Sei sulla pagina 1di 15

REVIEWS

Intrinsic mechanisms of neuronal


axon regeneration
Marcus Mahar    and Valeria Cavalli*
Abstract | Permanent disabilities following CNS injuries result from the failure of injured axons to
regenerate and rebuild functional connections with their original targets. By contrast, injury to
peripheral nerves is followed by robust regeneration, which can lead to recovery of sensory and
motor functions. This regenerative response requires the induction of widespread transcriptional
and epigenetic changes in injured neurons. Considerable progress has been made in recent years
in understanding how peripheral axon injury elicits these widespread changes through the
coordinated actions of transcription factors, epigenetic modifiers and, to a lesser extent,
microRNAs. Although many questions remain about the interplay between these mechanisms,
these new findings provide important insights into the pivotal role of coordinated gene
expression and chromatin remodelling in the neuronal response to injury.

Transcription factors Inhibitory factors present in the CNS tissue environ­ regenerative systems5,8. During neuronal development,
Proteins that activate or ment and the poor intrinsic regenerative capacity of axon growth competence decreases as neurons form
repress the expression of genes adult CNS neurons are obstacles to axon regeneration synapses to transmit information and integrate into
by binding to DNA sequence and functional recovery1–4. Aberrant synaptogenesis, functional neuronal circuits. The pro-​regenerative cel­
motifs proximal to a gene’s
transcription start site or
in which injured CNS axons at the lesion site remain lular changes after axon injury are thought to recapitu­
interacting enhancer regions. engaged in a synaptic function or maintenance state late some of the developmental processes that occur
rather than switching to a regenerative state, has also before synapse formation, thus switching the neuron
Epigenetic modifiers emerged as a barrier to axonal regrowth4,5. The failure from an active, electrically transmitting state back to
Proteins that post-​translationally
of damaged adult CNS axons to regrow results in perma­ an electrically silent, growth-​competent state4,9. Using
modify either DNA or histones,
which affects DNA compaction
nent disabilities for individuals with spinal cord injury peripheral sensory neurons and model organisms,
and accessibility for protein or stroke. As such, control of central axon regeneration such as Caenorhabditis elegans and zebrafish, many
binding. is both a major unmet medical need and an unsolved intrinsic signalling pathways that trigger the regenera­
problem in neurobiology. By contrast, functional reco­ tive response have been identified. An overview of the
MicroRNAs
(miRNAs). Single-​stranded
very can often occur following injury in the peripheral various model organisms and injury paradigms used
RNA molecules of 20–23 nervous system (PNS) because peripheral neurons retain to study axon regeneration is given in Box  1. More
nucleotides in length, the ability to self-​repair and reactivate intrinsic growth recently, studies have implicated the activity of vari­
generated endogenously from programmes after injury. However, severe insults to ous transcription factors, epigenetic modifiers of DNA
a single-​stranded hairpin
peripheral nerves can also lead to permanent neurologi­ methylation and chromatin structure, and microRNAs
precursor, which act as
post-​transcriptional inhibitors
cal deficits, including failure of reinnervation and the (miRNAs) that together activate the pro-​regenerative
in association with the development of chronic pain. In those cases, recovery is transcriptional programme.
RNA-​induced silencing slow and limited even when nerve grafts provide a bridge In this Review, we describe the most recent studies
complex (RISC). to promote axon regeneration6,7. To solve these problems that have focused on understanding how injured neu­
and promote functional recovery in the adult mamma­ rons revert to a growth-​competent state. We review
lian CNS and PNS, a major focus of research has been what is currently known about the early injury signals
Department of Neuroscience, the identification of the molecular mechanisms that elicited at the site of injury in regeneration-​competent
Hope Center for Neurological trigger a regenerative response and enable meaningful, axons and then focus on the intracellular communica­
Disorders and Center of long-​distance axon regeneration. tion mechanisms required to change the genetic and
Regenerative Medicine, Two primary approaches have been used to under­ epigenetic programme of injured neurons in order to
Washington University School
of Medicine, St Louis, MO,
stand the cell-​intrinsic mechanisms underlying axon allow axon regeneration to occur. Other aspects of axon
USA. regeneration: studying the mechanisms governing the regeneration, which include the roles of the cellular
*e-​mail: cavalli@wustl.edu developmental decline of axon growth competency in environment, axon–soma signalling and growth cone
https://doi.org/10.1038/ the CNS and studying the injury-​induced activation of formation and dynamics have been extensively covered
s41583-018-0001-8 a pro-​regenerative programme in the PNS and other in recent reviews4,5,8,10–13 and will not be discussed here.

Nature Reviews | Neuroscience


© 2018 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
Reviews

Box 1 | Models used to study intrinsic axon regeneration mechanisms


Mammalian sensory neurons with cell bodies in the dorsal root ganglia share unique features of both CNs and peripheral
nervous system neurons because they project both a peripheral axonal branch into peripheral nerves and a central axonal
branch through the dorsal root into the spinal cord. whereas injury to the peripheral axon elicits a regenerative response,
injury to the central axon fails to do so51,123. the activation of a pro-​regenerative programme following peripheral
axon injury is illustrated by the conditioning injury paradigm, in which a sensory neuron exposed to a prior
peripheral nerve lesion exhibits a dramatic improvement in peripheral axon regeneration following subsequent injury
when compared with that of a naive neuron104,213,214. this paradigm has also been used to reveal that activation of a
pro-​regenerative programme by a prior peripheral nerve lesion can partially overcome the inhibitory environment of
the CNs to improve regeneration of the central axonal branch after injury215.
spinal cord contusion injuries or partial spinal cord transections have also been used to study ascending sensory axon
regeneration in the CNs31,166. additionally, the regeneration of descending fibres can be examined in these models, and
studies have shown that different neuronal subtypes possess different regenerative capacities216.
another widely used model of CNs injury is a crush injury to the optic nerve, the axons of which project from retinal
ganglion cells (rGCs) in the retina and synapse in the superior colliculus. injury to the optic nerve initiates both
regenerative and apoptotic responses, with apoptosis as the ultimately dominant response66. Genetic manipulation of
rGCs has been shown to be capable of enhancing both survival and regeneration93.
axon injury responses have also been investigated in non-​mammalian vertebrates and invertebrates. the ability of
vertebrate zebrafish to fully recover following complete spinal cord transection has provided unique insights into the role
of non-​neuronal cells in the regenerative response17. the nematode Caenorhabditis elegans provides several unique
advantages, including optical transparency, the possibility of genetic manipulation and the availability of methods to
axotomize single axons with a laser14. a number of different injury paradigms have also been used in the larval and adult
nervous system of the fruitfly Drosophila melanogaster, which also enables the power of live imaging to be combined with
genetic manipulation16. Other animals used to study injury responses include Aplysia, which has been instrumental in
demonstrating the existence of positive injury signals217, lamprey, which possess both regenerating and non-​regenerating
CNs neurons17, and the axolotl, which can also regenerate its spinal cord following complete transection218. More details
for each experimental model can be found in ref.8.

This Review largely focuses on studies on mice, as One multifaceted signalling molecule downstream
recent reviews have covered axon regeneration in other of calcium is the second messenger cAMP. In rat dorsal
model systems including C. elegans14,15, Drosophila root ganglion (DRG) neurons, cAMP levels correlate
melanogaster16 and fish17. with growth capacity32. Generated by the calcium-​
dependent enzyme adenylyl cyclase after peripheral
Injury signalling injury, cAMP affects the activity of several important
In regeneration-​competent neurons, injury signalling proteins, including dual leucine zipper-​bearing kinase
occurs in two distinct temporal phases: a rapid phase (DLK; also known as MAP3K12), a highly conserved
that is dictated by a retrograde calcium wave that prop­ pro-​regenerative kinase21,33–35, and cAMP-​responsive
agates into the soma18–21 and a slow phase characterized element-​binding protein 1 (CREB1), an important pro-​
by the retrograde transport of signalling molecules by regenerative transcription factor32,36,37. Although cAMP
motor proteins10,22. induction is important for the activity of several pro-​
regenerative pathways, induction of the pro-​regenerative
Rapid injury signals. In regeneration-​competent neu­ programme requires additional factors: cAMP stimula­
rons, axonal membrane rupture combined with the tion activates only a fraction of injury-​induced genes
opening of voltage-​dependent sodium channels and in peripheral sensory neurons and is not sufficient
the inversion of Na+/Ca2+ exchange pumps promote to promote growth38.
an influx of calcium ions at the injury site10. A calcium Another set of genes that are rapidly activated by
wave is propagated to the soma by the actions of calcium-​dependent cAMP signalling and may contri­
L-​type voltage-​gated calcium channels (VGCCs) and the bute immediately following injury are immediate early
release of calcium from the endoplasmic reticulum via genes39, which are important for neuronal activity-​
ryanodine receptors10,18,20,21,23–25. The intracellular calcium induced responses and plasticity40. However, because
reaches millimolar concentrations in both the axon and few studies focus on early post-​injury time points,
soma after injury19 and plays several key roles in the ini­ the contribution of these genes (with the exception
tial stages of regeneration, including roles in membrane of the transcription factor AP-1 (JUN)) to axon regen­
resealing26,27, growth cone assembly11, translocation and eration remains largely unstudied. In fact, many neu­
activation of specific epigenetic modifiers28, local protein ronal activity-​induced molecules are common to various
synthesis29 and activation of additional signalling mole­ axon regenerative responses: these responses include
Motor proteins cules and transcription factors10. However, proper regu­ the calcium-​d ependent nuclear export of histone
Proteins, such as kinesin, lation of this intracellular calcium is required, as excess deacetylase 5 (HDAC5)20,41 and the activation of CREB1
dynein and myosin, that use concentrations can lead to cell death30 and the activity (refs36,42,43), CREB-​binding protein (CBP; also known as
either the microtubule or the of specific VGCCs is inhibitory to regeneration24,31. CREBBP)40,44–46, serum response factor (SRF)47–49 and
actin cytoskeleton for
movement by converting
Although the influence of calcium in regenerating neu­ mitogen-​activated protein (MAP) kinases49–51. A better
chemical energy into rons is widespread, this Review focuses on its effects on understanding of both the similarities and differences
mechanical force. signalling pathways, transcription and epigenetics (Fig. 1). between the neuronal responses to activity and those

www.nature.com/nrn
© 2018 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
Reviews

to injury may provide key insights into the early post-​ In uninjured, mature neurons, STAT3 mRNA is local­
injury events that set the stage for later pro-​regenerative ized to the axon, and the protein remains in an inactive
transcriptional changes. state localized to the cytoplasm57. Upon injury, axonal
STAT3 mRNA is locally translated near the injury site.
Delayed injury signals. The slower phase of injury Both Janus kinases (JAKs) and MAPKKs phosphorylate
signalling is mediated by retrograde transport of sev­ STAT3: JAKs phosphorylate Y705 and MAPKKs phos­
eral injury-​responsive signalling proteins, including phorylate S727 (ref.67). Although it is known that JAKs
extracellular-​signal-related kinases (ERKs)52, DLK, are activated via membrane glycoprotein 130 (gp130;
JUN N-​terminal kinase (JNK)53–56 and the transcription also known as IL6Rβ) signalling68, it is unclear which
factor signal transducer and activator of transcription 3 ligands are responsible for gp130 activation and their
(STAT3)57. Following axon injury, ERK1 and ERK2 are source. Several studies have suggested that interleukin 6
phosphorylated and associate with both vimentin and (IL-6)69–71 and CC-​chemokine ligand 2 (CCL2) produced
locally translated importin-​β in a calcium-​dependent by macrophages72 are responsible for gp130 activation in
manner. This association allows for both binding to DRG neurons, whereas astrocytic expression of ciliary
dynein for retrograde transport and protection from neurotrophic factor (CNTF)73 and leukaemia inhibitory
dephosphorylation52,58. Several lines of evidence have factor (LIF)74 activate gp130 in RGCs. pSTAT3 is retro­
demonstrated the importance of ERK1 and/or ERK2 gradely transported by the molecular motor dynein to
activation in the regenerative response. Studies have the cell soma57. In RGCs, it was recently demonstrated
shown that pharmacological inhibition of dual specific­ that the phosphorylation site serves as a localization sig­
ity MAP kinase kinase 1 (MAPKK1) or MAPKK2 (MAP nal for STAT3, with phosphorylation of Y705 directing
kinase kinases (MAPKKs) that are upstream of ERK) it to the nucleus (where it influences transcription) and
or the genetic deletion of vimentin significantly reduces phosphorylation of S727 directing it to the mitochon­
peripheral axon regeneration52. Furthermore, constitu­ dria (leading to increased ATP synthesis)67. STAT3 phos­
tive activation of serine/threonine-​protein kinase B-​raf phorylation at Y705 also plays a role in increasing the
(BRAF; a MAPKK kinase (MAPKKK) that is upstream interaction of STAT3 with stathmin75, which antagonizes
of MAPKK1 and MAPKK2) is sufficient to promote the microtubule destabilizing activity of stathmin76 and
robust axon regrowth into the adult spinal cord59. rescues axon degeneration in a mouse model of motor
Interestingly, the DLK–JNK pathway also appears neuron disease77.
to function in a calcium-​dependent manner. Studies In summary, axon injury leads to the activation of
in multiple organisms have revealed that DLK is a several signalling pathways and, in many cases, local
key sensor of local injury that informs the cell soma generation and activation of transcription factors or
about axonal injury35,56,60,61. Pharmacologically induced epigenetic modifiers78 that convey specific post-​injury
microtubule instability activates DLK60 as does cAMP cellular changes from the site of axon injury to the soma
(via protein kinase A (PKA)) in both D. melanogaster (Fig. 1). Injury location along the length of the axon
and mammals34 (Fig. 1). Downstream of DLK signalling, is also a key determinant of the cell body response:
JNK is both activated and retrogradely transported to proximal, but not distal, axon injury leads to severe loss
the nucleus, where it activates the transcription factor of rubrospinal neurons and facial motor neurons79,80.
JUN53–55,62. DLK activity is also necessary for the imme­ Similarly, RGC loss is more severe and more rapid when
diate retrograde transport of another injury signalling the optic nerve is injured close to the eye than when it
protein, phosphorylated STAT3 (pSTAT3), but is dispen­ is injured far from the eye81,82. These studies suggest
sable for the transport of both JNK and STAT3 after the that the availability of trophic support from exogenous
early injury stage63. Local alterations in the microtubule sources and, possibly, intracellular mechanisms that can
cytoskeleton elicited by injury, particularly an increase sense length83 contribute to the relationship between the
in tyrosinated α-​tubulin, facilitate the retrograde trans­ cellular response and the injury location.
port of these injury signals64. These studies demonstrate
the connection between the microtubule cytoskeleton Pathways involved in translation. Beyond the classi­
in sensing axon injury and the mechanisms by which cally defined injury signals, which are activated in
information about the injury is transmitted to the soma. the axon and travel back to the soma, other signalling
Although axon injury activates several pathways pathways have recently been shown to be involved in
in an analogous manner in diverse types of neurons, the control of axon regeneration. Exposure of neurons
Cofactor
contextual cues that are specific to those neuronal pop­ to growth-​limiting molecules, such as myelin-​derived
Protein that binds with other ulations can result in a dramatically different interpre­ Nogo, myelin-​associated glycoprotein (MAG) and
proteins to form heteromeric tation of the injury signals. Studies in retinal ganglion chondroitin sulfate proteoglycans (CSPGs), activates
complexes that alter or cells (RGCs) suggest that cofactor interactions are one the enzyme poly(ADP-​ribose) polymerase 1 (PARP1),
enhance the function of its
mechanism of specifying the cellular response following causing the accumulation of poly(ADP-​ribose), which
binding partners.
DLK activation65. Whereas DLK signalling in peripheral limits axon growth84. Pharmacological inhibition or
Poly(ADP-​ribose) neurons is necessary for the activation of several pro-​ genetic deletion of PARP1 facilitates axon regeneration
A negatively charged polymer regenerative pathways63, in RGCs it promotes cell death66. over non-​permissive substrates in cultured cortical and
of ADP-​ribose that can be In the latter context, DLK interacts with leucine zipper-​ DRG neurons by a mechanism that is independent of
added to proteins. Poly(ADP-​
ribose) represents a unique
bearing kinase (LZK; also known as MAP3K13), another histone poly(ADP-​ribosylation) (PARylation) or the
post-​translational modification MAPKKK, to activate a set of transcription factors that activation of regeneration-​associated genes (RAGs)84.
that regulates protein function. leads to RGC death after optic nerve injury65. This system is conserved across species, as removal of

Nature Reviews | Neuroscience


© 2018 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
Reviews

a Transcription factors
Satellite
glial cell Epigenetic modifiers
Signalling molecules
Soma
Transport and cytoskeletal molecules
Nucleus
Me Me Me

Pro-regenerative genes

TET3

Ca2+
PRKD1
Central branch Peripheral branch VGCC Injury

Axon
Calcium backpropagation
Translocation
DLK PKA cAMP
XBP1
Stress ERK
Importin-β mRNA:
Retrograde transport • STAT3
ER
• Filamin A
Filamin A • Importin-β

P P
STAT3
Retrograde transport

Cytokines

b
GFAP KCNJ10

Y705
P
STAT3
Ac Ac ?
TF
TF TF
Pro-regenerative genes
XBP1 TET3

KAT2B
HDAC3
HDAC5 Injury

Retrograde transport
Injury signals DLK
(DLK, JNK and JIP3)
ERK Anterograde transport
Vimentin Dynein–dynactin Tyrosinated
Importin-β tubulin
S727
P Acetylated tubulin
STAT3 PRKD1
HDAC5 Filamin A

Mitochondrion Kinesin
Deacetylated
tubulin
Microtubule

www.nature.com/nrn
© 2018 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
Reviews

◀ Fig. 1 | signalling events following peripheral axon injury. a | Early injury signalling regeneration and appears to have a role that is conserved
events. Following injury , calcium rapidly flows into the damaged axon and is actively in C. elegans, D. melanogaster and mice89–91. Another
propagated to the cell soma by voltage-​gated calcium channels (VGCCs) and the release type of stress response, involving glucocorticoid release
of calcium by internal stores in the endoplasmic reticulum (ER). The release of stored and increased expression of glucocorticoid receptors
calcium also triggers cellular stress pathways in the axon, resulting in the splicing of
in sensory neurons, is also integral to the regenerative
X-​box-binding protein 1 (XBP1) and its translocation to the nucleus91. The increase in
calcium activates pro-​regenerative pathways at the site of injury and in the cell soma.
response after nerve injury71.
For example, near the injury site, calcium activates cAMP, which in turn activates protein Mammalian target of rapamycin (mTOR) is a key
kinase A (PKA). PKA then activates dual leucine zipper-​bearing kinase (DLK), a key molecule involved in another very well-​established path­
mediator of later regenerative events34. Similarly , calcium at the injury site facilitates way that can be manipulated to promote regeneration92.
both the local translation of signal transducer and activator of transcription 3 (STAT3)57, mTOR is activated following peripheral but not central
importin-​β58 and filamin A247, and the association of extracellular-​signal-related kinase axon injury, and genetic activation of mTOR through
(ERK) with importin-​β, which allows them to be retrogradely transported52. The the deletion of upstream negative regulators, including
backpropagation of calcium to the cell soma promotes the translocation of serine/ phosphatase and tensin homologue (PTEN), hamartin
threonine-​protein kinase D1 (PRKD1) to the nucleus, which promotes histone (TSC1) and tuberin (TSC2), increases axon regenera­
deacetylase 5 (HDAC5) nuclear export20. The calcium backpropagation also facilitates
tion in both the PNS and CNS93–96. However, both the
the expression of the methylcytosine dioxygenase TET3, which promotes post-​injury
demethylation of DNA28. In addition to the actions of calcium, other pathways are
upstream and downstream mechanisms involved in
triggered by the injury: in the axon, the transcription factor STAT3 is phosphorylated, mTOR function in axon regeneration are not well under­
probably through the activity of cytokines, and is retrogradely transported to the nucleus stood. Crosstalk between mTOR and STAT3 signalling
to activate regenerative programmes57. b | Late injury signalling events. In the axon, DLK pathways97, as well as negative feedback by the mTOR
enables the retrograde transport of several injury signalling proteins, including JUN downstream target ribosomal protein S6 kinase-​β1
N-​terminal kinase (JNK), JNK-​interacting protein 3 (JIP3) and DLK itself, to the nucleus53,54,63. (S6K1)98, suggest that a deeper understanding of this
This process is also facilitated by an increase in tyrosinated tubulin near the injury site64. master regulator of axon growth is needed.
Phosphorylated STAT3 can localize either to mitochondria (when phosphorylated at The relevance of cell-​specific injury responses is
S727) to increase ATP synthesis or to the nucleus (when phosphorylated at Y705) to emerging with the observation that in RGCs, PTEN
influence transcription of pro-​regenerative genes67. ERK , which is protected by vimentin
deletion increases regeneration almost exclusively in
during its retrograde transport, causes the histone acetyltransferase KAT2B to translocate
into the nucleus52,166. Conversely , HDAC5 and HDAC3 are exported from the nucleus in
the α-​RGC subtype99. Whether this is a result of an
response to PRKD1 nuclear translocation20 and HDAC5 is transported to the growth epigenetic barrier (see below and Box 2) that is pres­
cone, where it interacts with filamin A and PRKD1 to deacetylate microtubules167,247. The ent in other RGC subtypes remains to be determined.
changes in subcellular location of these histone modifiers, along with the calcium-​ Cell-​type-specific responses may also result from other
dependent increase in TET3 (ref.28), cause a transition from repressive DNA and histone mechanisms, such as post-​transcriptional modification
methylation (Me) into permissive histone acetylation (Ac). This transition likely results in a of RNA, that affect gene expression (and are referred to
relaxation of the DNA around pro-​regenerative genes, allowing binding by pro-​ as epitranscriptomic mechanisms)100. Because RNA modi­
regenerative transcription factors and expression of pro-​regenerative genes20,28,179. fications were proposed to affect activity-​induced RNA
Additional signalling may also be provided by surrounding satellite glial cells, which editing and localization in the brain101 and are known
respond to axon injury by downregulating the ATP-​dependent inwardly rectifying
to affect cortical development102, it is likely that RNA
potassium channel Kir4.1 (KCNJ10)242 and upregulating glial fibrillary acidic protein
(GFAP)231. The molecular mechanisms by which satellite glial cells can sense a distant
modifications have an important role in axon regenera­
axon injury remain largely unknown (indicated by the question mark). P, phosphorylation. tion. Indeed, nerve injury was recently shown to induce
an increase in N6-methyladenosine, the most abundant
modification of mRNA, in transcripts encoding many
poly(ADP-​ribose) from proteins by poly(ADP-​ribose) RAGs, and deletion of the enzymes responsible for
RNA processing glycohydrolases (PARGs) enhances axon regeneration in this epitranscriptomic mark was shown to reduce axon
The process by which an RNA
molecule translated from DNA
C. elegans neurons85. Additionally, PARP is locally acti­ regeneration in the PNS103.
undergoes modifications, vated in the injured optic nerve84; however, inhibition
including 5′ capping, 3′ of PARP is insufficient to enhance axonal regeneration Transcriptional changes
polyadenylation, splicing and in either the optic nerve crush injury or thoracic spinal In the two decades following the discovery that peri­
methylation, before the RNA is
cord injury mouse models86. This is consistent with the pheral axon regeneration is transcription-​dependent104,
translated into a protein.
fact that downstream of Nogo, MAG and CSPGs, other an increasing focus has been put on uncovering and
Unfolded protein response pathways cause growth cone collapse87. Although PARP understanding the roles of axon injury-​induced genes105.
A cellular stress response that inhibitors alone may not be sufficient to promote reco­ More recently, advances in next-​generation sequencing
is triggered by an excess of very, it will be interesting to see whether these pathways have enabled many studies to characterize the post-​
unfolded or misfolded proteins
in the endoplasmic reticulum.
can synergize in combinatorial approaches. injury transcriptional changes. These studies have iden­
RNA processing has also been shown to control axon tified more than 1,000 differentially expressed genes
Epitranscriptomic regenerative capacity. The RNA processing enzymes and uncovered many novel regulators of axon regene­
mechanisms RNA 3´-terminal phosphate cyclases (RTCA and ration106. One group of proteins that have received par­
Post-​transcriptional RNA
RTCB) act to inhibit axon regeneration after nerve ticular attention in these studies is transcription factors.
modifications that regulate
mRNA half-​life or translation or injury in C. elegans and D. melanogaster88,89. By con­ With their ability to bind regulatory DNA elements, such
otherwise alter biological trast, protein archease, an RNA ligase cofactor that as promoters and enhancers, transcription factors can
processes. functions downstream of RTCA, serves as a pro-​ influence the expression of large sets of genes in a cell-​
regeneration factor89. These RNA processing enzymes type-specific or context-​specific manner. Additionally,
Next-​generation sequencing
High-​throughput parallel
are involved in the unconventional mRNA splicing of exogenous expression of certain sets of transcription fac­
sequencing of either DNA or X-​box-binding protein 1 (XBP1), a stress sensor involved tors can instruct cells to undergo complex cellular changes,
RNA. in the unfolded protein response. XBP1 is required for such as reprogramming from one cell type to another107.

Nature Reviews | Neuroscience


© 2018 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
Reviews

Box 2 | epigenetic modifiers and transcription factors in the regulation of gene expression
Gene expression is regulated by the coordinated action of epigenetic modifiers and transcription factors219. epigenetic
mechanisms are defined as covalent chemical modifications of DNa and histones, which play an important role in
defining gene expression through the modulation of chromatin structure and function. DNa can be modified by
methylation (resulting in 5-methylcytosine marks) and demethylation pathways, which may produce intermediate
products (such as 5-hydroxymethylcytosine)220. whereas DNa methylation had been thought to be static in cells after
mitosis, it is now clear that this is a very dynamic modification of the DNa in both peripheral nervous system and CNs
neurons221. DNa is also packaged around histone octamers, which have tails that can be post-​translationally modified in
various ways, including through acetylation, methylation and phosphorylation165. these modifications are performed by
enzymes called ‘writers’, are removed by another class of enzymes called ‘erasers’ and serve as recognition sites for
proteins called ‘readers’. the genomic specificity of writers and erasers is largely dictated by the proteins with which they
complex222. readers and transcription factors recognize post-​translational motifs or sequence motifs, respectively, and
these motifs serve as binding sites for the complex. in the case of transcription factors, the presence of DNa methylation
can alter their cognate motif and therefore affect where they bind222,223. Cumulatively, these modifications affect the
overall compaction and packaging of DNa around histones, which alter the ability of transcriptional machinery to bind
DNa224. additional layers of gene regulation are provided by the packaging of chromatin, which is regulated by histone
occupancy and can either facilitate or block transcription factor binding and the initiation of transcription, and the
association of DNa into topologically associated domains in which regions of DNa are tethered together spatially224,225.

As such, understanding which transcription factors the histone acetyltransferase p300 (p300 HAT) to pro­
are responsible for inducing the post-​injury transcrip­ mote the expression of several pro-​regenerative target
tional changes that promote regeneration in peripheral genes119. Indeed, transcriptional regulation of these
neurons may enable CNS regeneration to be boosted genes by pSMAD1 is necessary for sciatic nerve regene­
through combinatorial manipulation. ration119, and pSMAD1 activation by bone morpho­
genetic protein 2 (BMP2) and/or BMP4 is sufficient
Transcription factors activated by injury signalling. As to promote axon growth in DRG neurons in vitro117.
previously discussed, many transcription factors are acti­ One interesting recent study also demonstrated that
vated directly downstream of injury signalling pathways. activation of SMAD2 and/or SMAD3 by activin signal­
For example, JNK signalling leads to the activation of ling after axon injury in a wild-​derived inbred mouse
JUN and increased expression of the cAMP-​dependent strain (CAST/Ei mice)120 confers a greater regenerative
transcription factor ATF3 (refs55,108,109). In DRG neurons, potential after peripheral and central injury when com­
JUN activation occurs in response to peripheral, but not pared with other previously derived mouse strains121.
central, axon injury51. Interestingly, JUN and ATF3 are Accordingly, exogenous activin is sufficient to increase
sufficient to promote regeneration only under certain peripheral regeneration in poorly regenerating C57Bl/6
cellular conditions. Although overexpression of ATF3 mice and was shown to modestly increase optic nerve
can promote peripheral nerve regeneration109, it fails to regeneration121.
do so in several models of CNS injury110,111. Conversely, Although these studies clearly demonstrate the
JUN overexpression can promote regeneration in cul­ importance of SMADs in the pro-​regenerative response,
tured cortical110,112,113 and DRG neurons114; however, as little is known about either the genes downstream of
described above, its activation downstream of DLK also SMAD transcriptional regulation or the partners with
promotes cell death in RGCs66. Whereas neuronal JUN which the SMAD transcription factors bind. Because
is clearly necessary for peripheral axon regeneration115, SMADs have a low binding affinity for DNA, binding
JNK-​mediated phosphorylation of JUN appears to not generally requires the recruitment of additional tran­
be required for its full function in facial nerve regenera­ scription factors or epigenetic modifiers122. Several bind­
tion116. Importantly, co-​expression of ATF3 and JUN can ing partners have been previously identified for SMAD2
cooperate to improve regeneration in DRG neurons to a and SMAD3, including pro-​regenerative transcription
greater extent than can be achieved by the expression of factors and epigenetic modifiers such as JUN, ATF3,
either transcription factor alone114. However, this coop­ cellular tumour antigen p53 (TP53), p300 HAT and
erative effect does not occur in cortical neurons110. These the histone acetyltransferase KAT2B122. Whether these
results make it clear that the extent to which genetic interactions occur in neurons after injury and affect
manipulation can impact regeneration is strongly influ­ transcriptional regulation is yet to be determined.
enced by cellular context and the competency of the As with the previously discussed transcription
neurons to respond (Box 3). factors, STAT3 is specifically activated in response to
Several members of the SMAD family of transcrip­ peripheral axon injury but not spinal cord or dorsal root
tion factors are also involved in peripheral axon regen­ injury123. This injury-​induced activation is necessary for
eration. Activated downstream of the injury-​induced the initiation of growth but not for axonal elongation:
phosphatidylinositol 3-kinase (PI3K)–glycogen syn­ mice with genetic deletion of STAT3 exhibit a delayed
thase kinase 3 (GSK3) signalling pathway, SMAD regenerative response in peripheral nerves124. The role
family member 1 (SMAD1) is both phosphorylated of STAT3 in initiating axon regeneration was further
and upregulated after peripheral injury, leading to its demonstrated in studies in which overexpression of
accumulation in the nucleus117,118. Once in the nucleus, STAT3 increased outgrowth and collateral sprouting
phosphorylated SMAD1 (pSMAD1) interacts with of central DRG branches after a dorsal column lesion124.

www.nature.com/nrn
© 2018 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
Reviews

Box 3 | cellular context and neuronal competency for axon regeneration Identifying combinations of pro-​regenerative tran-
scription factors. In addition to the previously high­
Genetic manipulations that have focused on overexpression of regeneration-​ lighted transcription factors, several others — namely
associated genes or transcription factors have yielded limited success114,143,146,148. this Myc proto-​o ncogene protein (MYC) 137, hypoxia-​
lack of success could be because more than one factor is necessary to fully stimulate inducible factor 1α (HIF1α)138, CREB1 (refs36,113), SOX11
the growth process; however, another explanation is that the cellular context
(refs139–143), TP53 (refs46,144), SRF48,145 and XBP1 (refs90,91) —
dictates the competency of the neurons to respond to a given genetic manipulation.
indeed, multiple factors influence the dynamics of chromatin interactions during have been identified to have roles in axon regeneration
development 201,226
, and each cell type is likely to have a specific, unique chromatin (see Table 1 and refs146,147). As little is known about the
organization. in the context of cellular reprogramming, the accessibility of the mechanisms by which these transcription factors are
chromatin to a given transcription factor dictates the effectiveness of the conversion227. activated after injury, their regulation during devel­
Differences in chromatin accessibility may also lead to different outcomes, as genomic opment or their downstream targets, they will not be
regions that are differentially bound by given transcription factors or differences in further discussed in detail here. However, as this list
signalling pathways may lead to different cellular responses, such as the cell death and continues to grow, it has become increasingly clear
regeneration that are promoted by transcription factor sOX11 (ref.143) and dual leucine that new methods are needed to identify those factors that
zipper bearing kinase (DLK)65, respectively. as such, therapies that address both cellular work cooperatively to establish the pro-​regenerative
competency and the manipulation of gene expression may be necessary.
gene regulatory network. Although interactions between
the known pro-​regenerative transcription factors have
STAT3 overexpression also enhances remodelling of been extensively studied in other cell types, it is criti­
lesioned corticospinal tract (CST) fibres and induces cal to understand their interactions in the context of
de novo formation of collaterals from unlesioned CST axon injury. This need is further highlighted by recent
fibres, which promotes functional recovery after spinal studies in which combinatorial co-​expression of a few
cord injury125. Indeed, overexpression of a hyperacti­ of these transcription factors had limited to no ability to
vated form of STAT3 can also modestly promote optic boost regeneration above single-​factor overexpression in
nerve regeneration67,126. Accordingly, inhibition of JAK2, poorly regenerating neurons110,148.
an upstream activator of STAT3, reduces regenerative So how do we identify combinations of transcrip­
growth both in vitro and in vivo127, whereas activa­ tion factors that are capable of reprogramming CNS
tion of the JAK–STAT pathway using cytokines such as neurons to a regenerative state without altering cell
CNTF can increase RGC survival and modestly pro­ identity? This is a question that is quickly becoming
mote regeneration128,129. The limited extent to which central to ongoing attempts to design therapies for
these cytokines can increase regeneration is thought CNS injury. Several lessons can be learned from pro­
to be due to the injury-​induced increase in suppressor tocols developed to convert cells into different types
of cytokine signalling 3 (SOCS3) expression, a potent and to engineer cell identity. First, these studies have
JAK–STAT inhibitor. Indeed, genetic deletion of SOCS3 shown that core transcription factors important for the
significantly increased regeneration when combined establishment of a particular cellular identity, such as
with expression of CNTF and knockout of the tumour the famous OSKM (OCT4, SOX2, KLF4 and MYC)
suppressor PTEN129,130. factors in induced pluripotent stem cells (iPSCs)149,150,
cooperatively bind enhancers and promoters to regulate
Developmentally regulated transcription factors. the transcription of downstream gene targets151,152. In the
Although many of the previously discussed transcrip­ context of regenerating neurons, core pro-​regenerative
tion factors have been discovered in the context of adult transcription factors would be likely to both repress
peripheral axon injury, the decline in axon growth com­ genes associated with mature neurons engaged in syn­
petency in the CNS following synapse formation has also aptic transmission and activate those associated with
proved useful in discovering regeneration-​associated growth and survival. Importantly, studies have also
transcription factors. In particular, many of these studies shown that each of the core transcription factors regu­
have focused on the Krueppel-​like factor (KLF) family of lates the expression of the others, reinforcing cell fate152.
transcription factors, which have been shown to regulate Whereas one recent study identified a core network of
Cytokines
Originally defined as immune the growth competency of RGCs and other CNS neu­ hub transcription factors involved in peripheral regene­
system proteins, these proteins rons131–133. Interestingly, examination of sequence sim­ ration114, their downstream targets and whether they
are now known to be released ilarity among KLF family members showed that those regulate the expression of the other transcription factors
by most cells and are that share a high degree of sequence similarity also have in the context of regeneration are still unclear. Similarly,
important in regulating
intercellular communication,
similar effects on regeneration131. For example, over­ it is unlikely that the transcription factors that drive the
cell function and cell survival. expression of KLF4, the expression of which increases pro-​regenerative network are activated simultaneously.
during development in RGCs, potently inhibits axon As such, examining how the pro-​regenerative gene reg­
Induced pluripotent stem regeneration in RGCs, whereas its deletion promotes ulatory network develops temporally will be critical
cells
axon regeneration in vivo after optic nerve injury131. for future efforts to recapitulate this process. Finally,
(iPSCs). Cells created from
differentiated cell types (for Similarly, deletion of KLF9, the expression of which reprogramming studies have shown that a successful
example, fibroblasts) that are also increases during development, promotes long-​ transition between cellular states requires epigenetic
reprogrammed by a cocktail distance optic nerve regeneration in a JNK3-dependent reorganization153. In the context of cellular reprogram­
of transcription factors (or manner134. Conversely, both KLF6 (refs131,135) and KLF7 ming, pioneer transcription factors, which can bind
other approaches) back to a
(refs131,132), which are developmentally downregulated, regions of compacted chromatin and recruit epigenetic
pluripotent state and are
capable of differentiating into promote growth when overexpressed in CNS neurons modifiers, play a substantial role in inducing these
all three germ layers. in both mammals and zebrafish136 (Table 1). changes and are necessary for successful conversion153–155.

Nature Reviews | Neuroscience


© 2018 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
Reviews

Table 1 | Pro-​regenerative transcription factors and their effects on axon regeneration


Transcription factor Necessary for sufficient for sufficient for
regeneration in PNs? regeneration in the PNs? regeneration in the cNs?
ATF3 Yes108 Yes109,114,148 No109,110,148
CREB1 Unknown Yes36 Yes36,113
HIF1α Yes 138
Yes 138
Unknown
JUN Yes 115,116
Yes 114
Yes110,112,113
KLF6 Unknown Unknown Yes131,135
KLF7 Unknown Unknown Yes131,132
MYC Unknown Unknown Yes137
SMAD1 Yes117,118,249 No118 Unknown
SMAD2 Unknown Unknown Unknown
SMAD3 Unknown Unknown Unknown
SOX11 Yes 140
Yes 141
Yes142,143
SRF Yes48 Yes48 Yes145
STAT3 Yes 124
Unknown Yes67,125,126,250
TP53 Unknown Yes144 Yes46,144
XBP1 Yes 91
Yes 91
Unknown
Several studies have manipulated transcription factors to alter the regenerative potential of neurons both in vitro and in vivo.
Experiments in which the transcription factor has been knocked down via short hairpin RNA or small interfering RNA or genetically
deleted have shown that several are necessary for PNS regeneration. Conversely , overexpression of various transcription factors
has demonstrated their ability to boost regeneration in either the peripheral nervous system (PNS) or CNS. ATF3, cAMP-​dependent
transcription factor ATF3; CREB1, cAMP-​responsive element-​binding protein 1; HIF1α, hypoxia-​inducible factor 1α;
JUN, transcription factor AP-1; KLF, Krueppel-​like factor ; MYC, Myc proto-​oncogene protein; SMAD, SMAD family member ; SOX11,
transcription factor SOX11; SRF, serum response factor ; STAT3 signal transducer and activator of transcription 3; TP53, cellular
tumour antigen p53; XBP1, X-​box-binding protein 1.

It will be interesting to see whether one or more pio­ contrast, SOX11 expression kills α-​RGCs, the cellu­
neer factors play a similar role in post-​injury epigenetic lar subtype that preferentially regenerates following
changes and whether they are required for inducing treatments such as PTEN deletion143. SOX11 expres­
long-​distance regeneration in CNS neurons. However, sion also promotes CST regeneration after spinal
it is also possible that even the addition of pioneer tran­ cord injury but interferes with functional recovery142.
scription factors is insufficient to reprogramme CNS Careful evaluation of the resulting phenotype follow­
neurons to grow owing to the presence of a growth-​ ing transcription factor manipulation will therefore be
repressive chromatin landscape in these cells153,156. necessary to evaluate any potential therapies as they
Indeed, a decrease in promoter accessibility during should promote both survival and regeneration while
cortical development has been observed110, revealing a maintaining the competency to respond and innervate
potential intrinsic constraint to axon growth and regen­ appropriate targets.
eration. This constraint may cause a failure reminiscent In DRG neurons, differences in the regenerative
of the low efficiency observed in cell reprogramming, response have even been observed between subtypes of
in which repressive chromatin remains a barrier to neurons. Single-​cell RNA sequencing (RNAseq) ana­
deterministic reprogramming153. Detailed analysis of lysis of injured DRG neurons revealed heterogeneous
the chromatin accessibility in neurons with differential transcriptomic responses159, which suggest that various
regenerative capacities will therefore be instrumental subtypes of DRG neurons utilize different gene regula­
in engineering and developing new strategies to direct tory networks to control regeneration, cell death and
axon regeneration. Indeed, recent technical develop­ neuropathic pain. These studies emphasize the need to
ments now enable researchers to study transcription and investigate molecular mechanisms of axonal injury and
chromatin accessibility in a cell-​type-specific manner157 regeneration in single cells. With the advent of various
and have been applied to show that neuronal activity single-​cell sequencing techniques, including RNAseq
modifies the chromatin accessibility landscape in the and assay for transposase-​accessible chromatin using
adult brain158. sequencing (ATACseq), these studies are now possible.
Another limitation of therapeutic strategies in which Although single-​cell analysis is not suitable for all stud­
the overexpression of transcription factors is used to ies, future transcriptomic and epigenetic analyses should
potentiate regeneration is that the response to a given utilize other available methods such as fluorescence
transcription factor may vary according to the cell activated cell sorting (FACS)160, laser capture microdis­
type in which it is overexpressed. For example, it was section (LCM)161, translating ribosome affinity purifica­
recently shown that SOX11, a known pro-​regenerative tion (TRAP)162 and isolation of nuclei tagged in specific
transcription factor in the PNS139, can reprogramme cell types (INTACT)157 to analyse defined populations
adult non-​α-RGCs to a growth-​competent state. By of neurons.

www.nature.com/nrn
© 2018 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
Reviews

Epigenetic integration in the nucleus overexpression of pro-​regenerative transcription factors


The term ‘epigenetics’ broadly encompasses the chro­ to promote CNS regeneration is yet to be determined.
matin modifications and changes in DNA structure that Calcium influx, protein kinase C (PKC) signalling
affect gene expression but are not directly encoded in and ERK signalling have been linked to changes in
the DNA sequence. Epigenetic mechanisms that can histone acetylation in response to injury20,166; however,
induce changes in gene expression include the covalent other signalling pathways also affect chromatin modi­
modification of histones and DNA, which can influence fications171. Histone modifiers have been identified as
chromatin 3D structure and function. An overview of downstream effectors of signal transduction pathways
different types of epigenetic modifiers and how they in other systems, some of which are known to be acti­
cooperate with each other and with transcription fac­ vated by axon injury. For example, whereas the phos­
tors is given in Box 2. Because epigenetic regulators can phorylation of H3S28 and H3S10 occurs downstream
affect transcription on a genome-​wide scale, yet do so of MAPK, PI3K, JAK–STAT and cAMP–CREB signal­
in a cell-​type-specific and context-​specific manner, ling, H3K18 acetylation functions downstream of PI3K
they represent interesting targets to promote neural signalling172. Similarly, changes in cellular metabolism
repair. Whereas the role of epigenetics in regulating can influence epigenetics and thus transcription173. As
CNS gene expression during development163 or neural the transition to a pro-​regenerative state is likely to be
activity is being actively investigated164, its role in axon metabolically demanding, the differential ability of cell
regeneration is only emerging. types to cope with this stress may contribute to their
ability to successfully regenerate.
Histone modifications. Although there are well over
200 identified histone modifications165, some of the best DNA modifications. One of the best-​studied and most
characterized involve acetylation of lysine residues in the stable epigenetic DNA modifications is the generation
amino-​terminal tail of histones. The addition of these of 5-methylcytosine (5mC), an epigenetic mark pro­
modifications, which are dynamically added by his­ duced when a methyl group is covalently added to cyto­
tone acetyltransferases (HATs) and removed by histone sine nucleotides within the DNA. This modification,
deacetylases (HDACs), helps to establish a more relaxed which is primarily deposited at CpG dinucleotides, is
chromatin conformation that is readily bound by tran­ dynamically regulated during neuronal development
scriptional machinery. Following axon injury, there is a and in response to various stimuli163,164. Three DNA
global accumulation of both H3K9 and H3K14 acetyl­ (cytosine-5)-methyltransferase (DNMT) enzymes —
ation20,166, and H4 acetylation119 in peripheral neurons, DNMT1, DNMT3A and DNMT3B — are responsible for
which may, in part, be caused by two recently discovered catalysing this reaction. All three isoforms are expressed
mechanisms. First, downstream of the injury-​induced in DRG sensory neurons; however, DNMT3A may also be
calcium influx, serine/threonine-​protein kinase D1 expressed in satellite glial cells in the DRG174,175. Generally
(PRKD1, also known as PKCμ) is phosphorylated, associated with transcriptional repression, DNA methyl­
causing it to translocate to the nucleus20. The increase ation can be removed by methylcytosine dioxygenases
in nuclear PRKD1 then causes HDAC5 to be exported (TET proteins) to activate transcription. TET proteins can­
from the nucleus20. In the axon tip, HDAC5 deacetylates not directly remove methyl groups from DNA but instead
microtubules, thereby increasing growth cone dynamics sequentially oxidize 5mC to 5-hydroxymethylcytosine
and promoting axon growth20,167. Similarly, there is evi­ (5hmC), 5-formylcytosine (5fC) and 5-carboxylcytosine
dence that HDAC3 is also exported from the nucleus to (5caC). Both 5fC and 5caC can then be targeted by thy­
some extent20. Activation of ERK signalling also causes mine DNA glycosylase-​mediated base excision repair,
epigenetic changes in sensory neurons by stimulating leaving behind an unmethylated cytosine176. Although
the accumulation of KAT2B in the nucleus, in which it originally viewed as an intermediate in this demethy­
binds and acetylates histones at several pro-​regenerative lation process, 5hmC is now known to be a stable DNA
genes166. However, in contrast to peripheral injury, CNS modification in the nervous system177.
injury fails to stimulate both the nuclear export of Conflicting results have arisen regarding the impor­
HDAC5 and the nuclear import of KAT2B, which may tance and dynamics of DNA methylation after periph­
explain the lack of an observable increase in histone eral axon injury. In one study, the methylation of CpG
acetylation or transcriptional changes following these islands, the regions of CG-​rich DNA typically located
injuries119,166. Interestingly, in injured RGCs, HDAC3 is near transcription start sites, was assayed following
imported into the nucleus, where it functions to repress either sciatic or dorsal column injury166. Few regions
gene expression and promote apoptosis168,169. The lack were found to be differentially methylated following
of a pro-​regenerative epigenetic response following sciatic injury, and comparisons between sciatic and
CNS injury may explain the failure of overexpressed dorsal column injuries also revealed few differences in
pro-​regenerative transcription factors to produce long-​ methylation166. However, in another study, the admin­
distance CNS regeneration. Notably, both exogenous istration of folate, which increased DNA methylation
expression of KAT2B166 and the use of HDAC inhibi­ globally in the spinal cord, increased regeneration of
tors170 are sufficient to modestly promote regeneration spinal neurons following combined spinal cord and
after spinal cord injury, and overexpression of p300 HAT peripheral nerve injury, and folate receptor-​α (FRα),
can promote some regeneration following optic nerve was shown to be necessary for peripheral regenera­
crush45. However, whether the manipulation of these tion178. Whether these effects of increased DNA methy­
epigenetic pathways can act synergistically with the lation are neuron-​specific remains to be determined.

Nature Reviews | Neuroscience


© 2018 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
Reviews

Upstream Role of microRNAs in injured neurons


a Before injury enhancer Despite their ability to regulate more than 60% of the
human transcriptome, the importance of miRNAs in
axon regeneration is only just beginning to be under­
5mC Me 5mC stood180. For these small (21–23 nucleotide) non-​coding
H3K9Me DNMTs H3K9Me H3K9Me RNAs to become active, they must first undergo a series
of processing steps181. At least one step of this biogene­
H3K9Me H3K9Me H3K9Me sis pathway has been identified as being necessary for
HDACs peripheral nerve regeneration as mice lacking one of
5mC the key synthesis enzymes, Dicer, exhibit both reduced
axon growth in vitro and reduced functional recovery in
vivo182,183. However, whether other proteins involved
Promoter Pro-regenerative gene in the miRNA biogenesis process are also necessary for
regeneration is yet to be determined. A potential area of
b interest will be to determine whether the endoplasmic
reticulum stress pathway and the miRNA biogene­sis
pathway, which are known to regulate each other, do so
5hmC p300HAT MYC ATF3 5hmC in the context of axon regeneration184,185. This is possible
CBP TET3 given that the endoplasmic reticulum stress response
A
KAT2B KAT2B AA promotes regeneration of sensory neurons91,186 and
H3K9Ac H3K9Ac survival of RGCs187.
mRNA
CBP Similar to other genes and non-​coding RNAs, miRNAs
Y705
P p300HAT are expressed in a tissue-​d ependent and context-​
STAT3 JUN TP53 RNAPII
dependent manner. For example, several miRNAs have
key roles in neural development and are expressed in
a temporally and spatially specific manner that corre­
Transcription factors Epigenetic modifiers
sponds to these roles188. As such, recent studies have
Fig. 2 | Model of epigenetic activation of pro-​regenerative genes in response to sought to determine the roles of pro-​regenerative
axon injury. a | When dorsal root ganglion neurons have formed synapses and are miRNAs189–193 (reviewed in ref. 176). Together, these
electrically active, pro-​regenerative gene bodies, promoters and enhancers are studies have identified a variety of miRNAs that exhibit
maintained in a transcriptionally repressed state through DNA methylation by DNA upregulated expression in the DRG following sciatic
(cytosine-5)-methyltransferases (DNMTs), histone methylation by yet unknown histone injury in mice and are capable of increasing regeneration
methyltransferases and deacetylation by histone deacetylases (HDACs)20,28,179.  in vitro, including miR-431, miR-222 and miR-21, and
b | Following injury , the nuclear export of HDACs20 and import of the histone have implicated several downstream genes as negative
acetyltransferase (HAT) KAT2B166 causes an increase in acetylation of histone H3K9 regulators of regeneration (including Kremen1 and Spry2
(H3K9Ac), whereas the upregulation of the methylcytosine dioxygenase TET3 causes the (refs190,192,193)). Notably, the expression of miR-222 pro­
conversion of repressive 5-methylcytosine (5mC) into 5-hydroxymethylcytosine (5hmC)28.
motes neurite outgrowth in cultured DRG neurons, in
Similarly , CREB-​binding protein (CBP) and p300 HAT, HATs that frequently bind to both
enhancers and promoters, associate with cellular tumour antigen p53 (TP53) to increase part by silencing PTEN192, consistent with the observed
H3K9Ac in pro-​regenerative gene promoters144,248. The turnover of transcriptionally decrease in PTEN levels in DRGs follow­ing periphe­
repressive marks into activating marks likely causes a relaxation in the chromatin, which ral nerve injury96. One recent study in CAST/Ei mice,
should allow pro-​regenerative transcription factors — such as Myc proto-​oncogene which have an increased neuronal regenerative capacity,
protein (MYC), cAMP-​dependent transcription factor ATF3, transcription factor AP-1 demonstrated the importance of miRNAs that are
(JUN) and signal transducer and activator of transcription 3 (STAT3) — to bind regulatory downregulated following periphe ­ral injury in this
elements and influence the transcription of downstream pro-​regenerative genes. regeneration194. miRNA sequencing was used to iden­
H3K9Me, methylation of histone HeK9; P, phosphorylation; RNAPII, RNA polymerase II. tify miR-7048-3p as an miRNA that is downregulated
after injury. This downregulation causes an increase
Another study reported widespread changes in 5hmC in expression of its target achaete-​scute homologue 1
after both sciatic nerve crush and dorsal column tran­ (ASCL1), a transcription factor that is capable of modestly
section in mice 179. Several pro-​regenerative genes, promoting growth194.
including Atf3, Smad1 and Bdnf, acquired 5hmC marks Despite a recent focus on utilizing miRNAs as ther­
after peripheral injury, but not after central injury179. apeutic targets, they have yet to be manipulated to
This finding is consistent with the finding that TET3 improve axon regeneration in vivo. This is largely due
is upregulated28,179 in response to peripheral injury and to general issues surrounding the in vivo manipulation
is necessary for proper regeneration and behavioural of miRNAs195. Another difficulty is the lack of con­
recovery28. Although these studies focused on 5hmC, sensus as to which miRNAs are important for regene­
the dynamic regulation of this mark strongly suggests ration. Indeed, published lists of differentially expressed
that 5mC is also being dynamically regulated either miRNAs tend to have little overlap with one another, even
through demethylation or through conversion into in studies with identical post-​injury time points190,192,193.
stable 5hmC. Future studies using methods with greater A few experimental reasons may explain this hetero­
genome coverage and cell-​type specificity will be able geneity, including the use of different species, different
to more accurately identify regions of differential injury paradigms and the limitations of microarrays.
methylation after injury (Fig. 2). The use of the whole DRG, a tissue with a notoriously

www.nature.com/nrn
© 2018 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
Reviews

Box 4 | contribution of glial cells to peripheral nerve regeneration how injured neurons can reprogramme themselves to
regrow their axons while maintaining their competency
in the mouse peripheral nervous system, the intrinsic mechanisms regulating axon to form synapses and how they can stop growth once
regeneration have traditionally been assumed to involve changes that occur within the regeneration is complete is still not understood. Indeed,
neuron. However, satellite glial cells (sGCs), the main type of glial cells in sensory studies focused on the temporal regulation of regenerat­
ganglia, ensheathe sensory neuron soma (Fig. 1). each sensory neuron and its
ing neurons will help us to understand how this process
ensheathing sGCs thus form a distinct functional and morphological unit 228–230
. sGCs
are similar to astrocytes in the CNs as they have the capacity to buffer the extracellular is initiated after injury, maintained during elongation
environment via potassium and calcium channels and express the intermediate and successfully shut down upon target reinnervation.
filament glial fibrillary acidic protein (GFaP). recent studies have provided evidence Going beyond transcriptomics to understand the use
that distant nerve injury induces morphological and molecular changes in sGCs, of regulatory elements, such as enhancers and promot­
suggesting that sGCs can recognize and respond to this stimulus. these changes ers, and the influence of genome topology in the context
include an increase in GFaP expression231–236, extracellular-​signal-related kinase of axon regeneration will also be important. The cell-​
(erK) phosphorylation237,238, an increase in sGC coupling via gap junctions239–241, type-specific use of enhancer elements and the organi­
downregulation of the atP-​dependent inwardly rectifying potassium channel Kir4.1 zation of topologically associated domains are likely to
(ref.242) and proliferation231,243,244. although little is known about how sGCs respond to
be instrumental in determining the competency of cells
axon injury, early spontaneous neuronal activity and direct communication between
233
to respond properly to axon injury198–202.
the neuronal soma and sGCs231 were proposed to underlie sGC activation. indeed,
robust communication between neurons and sGCs is critical for neuronal excitability Whereas the majority of the pathways discussed in
and nociception245, and future studies will reveal whether sGCs, similarly to the repair this Review are highly conserved, others exist only
schwann cells246, participate in the neuronal response to peripheral nerve lesion and in higher mammals or humans. For example, ARMCX1
control regenerative outcomes. is a mammalian-​specific gene that controls mitochon­
drial transport during neuronal repair203. Increased
mitochondrial transport to injured axons is required
Genome topology
heterogeneous population of cells, as an input for for regeneration203,204, and CSPGs can prevent mitocho­
The 3D DNA structure that miRNA profiling could also have a profound impact on ndria from localizing to the growth cone205. There are
dictates its accessibility to these studies: in addition to neurons and satellite glial also human-​specific mRNA splice variants and human-​
binding by proteins such as cells (Box 4), macrophages are known to migrate into the specific genes in the nervous system that may play a
transcription factors and
DRG after injury196,197. Furthermore, the heterogeneity of role in the regenerative response206–208. To determine
epigenetic modifiers.
injury responses in different sensory neuron subtypes159 whether findings in animal model systems are predic­
is also likely to have influenced these miRNA profiles. It tive of the injury responses of human neurons, it will
will be important to continue to examine how different be important to determine whether the epigenomic and
neuronal subtypes respond to injury and how the regu­ transcriptional pathways identified in animal models
lation of translation in different populations of sensory also function in human neurons. For example, in the
neurons relates to their functional recovery. developing retina, most epigenetic changes are con­
served from mice to humans209; however, whether the
Conclusion and future directions epigenetic injury responses are conserved in humans
Although considerable progress has been made in is unknown.
understanding the mechanisms underlying peripheral Finally, age is another key consideration that is highly
nerve regeneration and how these can be manipulated to relevant to the development of therapeutic strategies. As
promote regeneration in the CNS, there are many chal­ axon injury in humans can result from a variety of insults
lenges to overcome in order to develop therapies that at any age, it is important to note that regeneration
achieve complete axon regrowth and functional recov­ tends to be increasingly limited with age210,211. Indeed,
ery. Bioinformatics approaches to integrate discoveries epigenetic profiling revealed that promoter accessibility
related to regenerative capacity across multiple laborato­ decreases with cortical maturation110, and methylation of
ries could help to drive drug discovery, but such studies histone H3K27, a mark that correlates with closed chro­
remain difficult to interpret because of the lack of cell-​ matin, increases with age in the brain in medium spiny
type-specific data. Although a recent study identified neurons212. Thus, manipulating epigenetic machinery
pathways and small molecules that can enhance growth will likely be critical in efforts to improve regenerative
of injured neurons114, the effects observed were small, outcomes and may need to be tailored according to age.
and future multilevel bioinformatics analyses will ben­
Published online xx xx xxxx
efit from novel neuron-​specific data sets. Furthermore,

1. Liu, K., Tedeschi, A., Park, K. K. & He, Z. Neuronal central nervous system: lessons from neuronal 10. Rishal, I. & Fainzilber, M. Axon-​soma communication
intrinsic mechanisms of axon regeneration. development. Dev. Dyn. 247, 18–23 (2017). in neuronal injury. Nat. Rev. Neurosci. 15, 32–42
|Annu. Rev. Neurosci. 34, 131–152 (2011). 6. Wood, M. D., Kemp, S. W., Weber, C., Borschel, G. H. (2014).
2. Di Giovanni, S. Molecular targets for axon & Gordon, T. Outcome measures of peripheral 11. Bradke, F., Fawcett, J. W. & Spira, M. E. Assembly
regeneration: focus on the intrinsic pathways. nerve regeneration. Ann. Anat. 193, 321–333 of a new growth cone after axotomy: the precursor to
Expert Opin. Ther. Targets 13, 1387–1398 (2011). axon regeneration. Nat. Rev. Neurosci. 13, 183–193
(2009). 7. Saheb-​Al-Zamani, M. et al. Limited regeneration in (2012).
3. Schwab, M. E. & Strittmatter, S. M. Nogo limits long acellular nerve allografts is associated with 12. Cho, Y. & Cavalli, V. HDAC signaling in neuronal
neural plasticity and recovery from injury. Curr. Opin. increased Schwann cell senescence. Exp. Neurol. 247, development and axon regeneration. Curr. Opin.
Neurobiol. 27, 53–60 (2014). 165–177 (2013). Neurobiol. 27, 118–126 (2014).
4. Tedeschi, A. & Bradke, F. Spatial and temporal 8. He, Z. & Jin, Y. Intrinsic control of axon regeneration. 13. Lu, Y., Belin, S. & He, Z. Signaling regulations of
arrangement of neuronal intrinsic and extrinsic Neuron 90, 437–451 (2016). neuronal regenerative ability. Curr. Opin. Neurobiol.
mechanisms controlling axon regeneration. 9. Quadrato, G. & Di Giovanni, S. Waking up the sleepers: 27, 135–142 (2014).
Curr. Opin. Neurobiol. 42, 118–127 (2017). shared transcriptional pathways in axonal regeneration 14. Byrne, A. B. & Hammarlund, M. Axon regeneration in
5. Kaplan, A., Bueno, M., Hua, L. & Fournier, A. E. and neurogenesis. Cell. Mol. Life Sci. 70, 993–1007 C. elegans: worming our way to mechanisms of axon
Maximizing functional axon repair in the injured (2013). regeneration. Exp. Neurol. 287, 300–309 (2017).

Nature Reviews | Neuroscience


© 2018 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
Reviews

15. Chisholm, A. D., Hutter, H., Jin, Y. & Wadsworth, W. G. 36. Gao, Y. et al. Activated CREB is sufficient to overcome This study shows that activation of BRAF
The genetics of axon guidance and axon regeneration inhibitors in myelin and promote spinal axon synergizes with PTEN knockout to promote optic
in Caenorhabditis elegans. Genetics 204, 849–882 regeneration in vivo. Neuron 44, 609–621 (2004). nerve regeneration.
(2016). 37. Ma, T. C. & Willis, D. E. What makes a RAG regeneration 60. Valakh, V., Walker, L. J., Skeath, J. B. & DiAntonio, A.
16. Hao, Y. & Collins, C. Intrinsic mechanisms for axon associated? Front. Mol. Neurosci. 8, 43 (2015). Loss of the spectraplakin short stop activates the DLK
regeneration: insights from injured axons in 38. Blesch, A. et al. Conditioning lesions before or after injury response pathway in Drosophila. J. Neurosci.
Drosophila. Curr. Opin. Genet. Dev. 44, 84–91 spinal cord injury recruit broad genetic mechanisms 33, 17863–17873 (2013).
(2017). that sustain axonal regeneration: superiority to 61. Yan, D., Wu, Z., Chisholm, A. D. & Jin, Y. The DLK-1
17. Rasmussen, J. P. & Sagasti, A. Learning to swim, camp-​mediated effects. Exp. Neurol. 235, 162–173 kinase promotes mRNA stability and local translation
again: axon regeneration in fish. Exp. Neurol. 287, (2012). in C. elegans synapses and axon regeneration.
318–330 (2017). 39. Perez-​Cadahia, B., Drobic, B. & Davie, J. R. Activation Cell 138, 1005–1018 (2009).
18. Ziv, N. E. & Spira, M. E. Spatiotemporal distribution of and function of immediate-​early genes in the nervous 62. Kenney, A. M. & Kocsis, J. D. Peripheral axotomy
Ca2+ following axotomy and throughout the recovery system. Biochem. Cell Biol. 89, 61–73 (2011). induces long-​term c-​Jun amino-​terminal kinase-1
process of cultured Aplysia neurons. Eur. J. Neurosci. 40. West, A. E. & Greenberg, M. E. Neuronal activation and activator protein-1 binding activity by
5, 657–668 (1993). activity-​regulated gene transcription in synapse c-​Jun and junD in adult rat dorsal root ganglia in vivo.
19. Ziv, N. E. & Spira, M. E. Axotomy induces a transient development and cognitive function. Cold Spring Harb. J. Neurosci. 18, 1318–1328 (1998).
and localized elevation of the free intracellular calcium Perspect. Biol. 3, a005744 (2011). 63. Shin, J. et al. Dual leucine zipper kinase is required for
concentration to the millimolar range. J. Neurophysiol. 41. Chawla, S., Vanhoutte, P., Arnold, F. J., Huang, C. L. & retrograde injury signaling and axonal regeneration.
74, 2625–2637 (1995). Bading, H. Neuronal activity-​dependent Neuron 74, 1015–1022 (2012).
20. Cho, Y., Sloutsky, R., Naegle, K. M. & Cavalli, V. nucleocytoplasmic shuttling of HDAC4 and HDAC5. 64. Song, W., Cho, Y., Watt, D. & Cavalli, V. Tubulin-​tyrosine
Injury-​induced HDAC5 nuclear export is essential for J. Neurochem. 85, 151–159 (2003). ligase (TTL)-mediated increase in tyrosinated α-​tubulin
axon regeneration. Cell 155, 894–908 (2013). 42. Lonze, B. E. & Ginty, D. D. Function and regulation in injured axons is required for retrograde injury
This study demonstrates that, following axon of CREB family transcription factors in the nervous signaling and axon regeneration. J. Biol. Chem. 290,
injury, a calcium wave triggers epigenetic changes system. Neuron 35, 605–623 (2002). 14765–14775 (2015).
through the nuclear export of HDAC5 to promote 43. Sheng, M., Thompson, M. A. & Greenberg, M. E. 65. Welsbie, D. S. et al. Enhanced functional genomic
expression of pro-​regenerative genes. CREB: a Ca(2+)-regulated transcription factor screening identifies novel mediators of dual leucine
21. Ghosh-​Roy, A., Wu, Z., Goncharov, A., Jin, Y. & phosphorylated by calmodulin-​dependent kinases. zipper kinase-​dependent injury signaling in neurons.
Chisholm, A. D. Calcium and cyclic AMP promote Science 252, 1427–1430 (1991). Neuron 94, 1142–1154.e6 (2017).
axonal regeneration in Caenorhabditis elegans and 44. Chrivia, J. C. et al. Phosphorylated CREB binds This study identifies novel members of the DLK
require DLK-1 kinase. J. Neurosci. 30, 3175–3183 specifically to the nuclear protein CBP. Nature 365, injury signalling pathway in RGCs and demonstrates
(2010). 855–859 (1993). that LZK cooperates with DLK to promote RGC cell
22. Saito, A. & Cavalli, V. Signaling over distances. 45. Gaub, P. et al. The histone acetyltransferase p300 death in response to axon injury.
Mol. Cell. Proteomics 15, 382–393 (2016). promotes intrinsic axonal regeneration. Brain 134, 66. Watkins, T. A. et al. DLK initiates a transcriptional
23. Kulbatski, I., Cook, D. J. & Tator, C. H. Calcium entry 2134–2148 (2011). program that couples apoptotic and regenerative
through L-​type calcium channels is essential for 46. Tedeschi, A., Nguyen, T., Puttagunta, R., Gaub, P. & responses to axonal injury. Proc. Natl Acad. Sci. USA
neurite regeneration in cultured sympathetic neurons. Di Giovanni, S. A p53-CBP/p300 transcription module 110, 4039–4044 (2013).
J. Neurotrauma 21, 357–374 (2004). is required for GAP-43 expression, axon outgrowth, 67. Luo, X. et al. Enhanced transcriptional activity and
24. Enes, J. et al. Electrical activity suppresses axon and regeneration. Cell Death Differ 16, 543–554 mitochondrial localization of STAT3 co-​induce axon
growth through Ca(v)1.2 channels in adult primary (2009). regrowth in the adult central nervous system.
sensory neurons. Curr. Biol. 20, 1154–1164 (2010). 47. Knoll, B. Serum response factor mediated gene Cell Rep. 15, 398–410 (2016).
25. Sun, L. et al. Neuronal regeneration in C. elegans activity in physiological and pathological processes of 68. Zigmond, R. E. gp130 cytokines are positive signals
requires subcellular calcium release by ryanodine neuronal motility. Front. Mol. Neurosci. 4, 49 (2011). triggering changes in gene expression and axon
receptor channels and can be enhanced by 48. Stern, S. et al. The transcription factor serum response outgrowth in peripheral neurons following injury.
optogenetic stimulation. J. Neurosci. 34, factor stimulates axon regeneration through cytoplasmic Front. Mol. Neurosci. 4, 62 (2011).
15947–15956 (2014). localization and cofilin interaction. J. Neurosci. 33, 69. Zhong, J., Dietzel, I. D., Wahle, P., Kopf, M. &
26. Tuck, E. & Cavalli, V. Roles of membrane trafficking in 18836–18848 (2013). Heumann, R. Sensory impairments and delayed
nerve repair and regeneration. Commun. Integr. Biol. 49. Xia, Z., Dudek, H., Miranti, C. K. & Greenberg, M. E. regeneration of sensory axons in interleukin-6-deficient
3, 209–214 (2010). Calcium influx via the NMDA receptor induces mice. J. Neurosci. 19, 4305–4313 (1999).
27. McNeil, P. L. & Kirchhausen, T. An emergency immediate early gene transcription by a MAP 70. Cafferty, W. B. et al. Conditioning injury-​induced spinal
response team for membrane repair. Nat. Rev. Mol. kinase/ERK-​dependent mechanism. J. Neurosci. 16, axon regeneration fails in interleukin-6 knock-​out mice.
Cell Biol. 6, 499–505 (2005). 5425–5436 (1996). J. Neurosci. 24, 4432–4443 (2004).
28. Weng, Y. L. et al. An intrinsic epigenetic barrier for 50. Chierzi, S., Ratto, G. M., Verma, P. & Fawcett, J. W. 71. Lerch, J. K. et al. Stress increases peripheral axon
functional axon regeneration. Neuron 94, 337–346. The ability of axons to regenerate their growth cones growth and regeneration through glucocorticoid
e6 (2017). depends on axonal type and age, and is regulated by receptor-​dependent transcriptional programs. eNeuro
This study reveals that DNA is actively calcium, cAMP and ERK. Eur. J. Neurosci. 21, https://doi.org/10.1523/ENEURO.0246-17.2017
demethylated at the promoters of RAGs, which is 2051–2062 (2005). (2017).
required to promote functional axonal regeneration 51. Broude, E., McAtee, M., Kelley, M. S. & Bregman, B. S. 72. Niemi, J. P., DeFrancesco-​Lisowitz, A., Cregg, J. M.,
of peripheral sensory neurons. c-​Jun expression in adult rat dorsal root ganglion Howarth, M. & Zigmond, R. E. Overexpression of the
29. Perry, R. B. & Fainzilber, M. Local translation in neurons: differential response after central or monocyte chemokine CCL2 in dorsal root ganglion
neuronal processes — in vivo tests of a “heretical peripheral axotomy. Exp. Neurol. 148, 367–377 neurons causes a conditioning-​like increase in neurite
hypothesis”. Dev. Neurobiol. 74, 210–217 (2014). (1997). outgrowth and does so via a STAT3 dependent
30. Stirling, D. P. & Stys, P. K. Mechanisms of axonal 52. Perlson, E. et al. Vimentin-​dependent spatial mechanism. Exp. Neurol. 275, 25–37 (2016).
injury: internodal nanocomplexes and calcium translocation of an activated MAP kinase in injured 73. Muller, A., Hauk, T. G. & Fischer, D. Astrocyte-​derived
deregulation. Trends Mol. Med. 16, 160–170 nerve. Neuron 45, 715–726 (2005). CNTF switches mature RGCs to a regenerative state
(2010). 53. Cavalli, V., Kujala, P., Klumperman, J. & Goldstein, L. S. following inflammatory stimulation. Brain 130,
31. Tedeschi, A. et al. The calcium channel subunit Sunday Driver links axonal transport to damage 3308–3320 (2007).
Alpha2delta2 suppresses axon regeneration in the signaling. J. Cell Biol. 168, 775–787 (2005). 74. Leibinger, M., Andreadaki, A., Diekmann, H. &
adult CNS. Neuron 92, 419–434 (2016). 54. Drerup, C. M. & Nechiporuk, A. V. JNK-​interacting Fischer, D. Neuronal STAT3 activation is essential for
This study shows that the calcium channel subunit protein 3 mediates the retrograde transport of CNTF- and inflammatory stimulation-​induced CNS
VGCC-​α2/δ2 (CACNA2D2) is developmentally activated c-​Jun N-​terminal kinase and lysosomes. axon regeneration. Cell Death Dis. 4, e805 (2013).
upregulated in DRG neurons, decreasing neuronal PLoS Genet. 9, e1003303 (2013). 75. Verma, N. K. et al. STAT3-stathmin interactions
growth capacity, and that its inhibition with the 55. Lindwall, C. & Kanje, M. Retrograde axonal transport control microtubule dynamics in migrating T-​cells.
existing drug pregabalin increases regenerative of JNK signaling molecules influence injury induced J. Biol. Chem. 284, 12349–12362 (2009).
capacity. nuclear changes in p-​c-Jun and ATF3 in adult rat 76. Ng, D. C. et al. Stat3 regulates microtubules by
32. Cai, D. et al. Neuronal cyclic AMP controls the sensory neurons. Mol. Cell Neurosci. 29, 269–282 antagonizing the depolymerization activity of
developmental loss in ability of axons to regenerate. (2005). stathmin. J. Cell Biol. 172, 245–257 (2006).
J. Neurosci. 21, 4731–4739 (2001). 56. Xiong, X. et al. Protein turnover of the Wallenda/DLK 77. Selvaraj, B. T., Frank, N., Bender, F. L., Asan, E. &
33. Udina, E. et al. Electrical stimulation of intact kinase regulates a retrograde response to axonal Sendtner, M. Local axonal function of STAT3 rescues
peripheral sensory axons in rats promotes outgrowth injury. J. Cell Biol. 191, 211–223 (2010). axon degeneration in the pmn model of motoneuron
of their central projections. Exp. Neurol. 210, 57. Ben-​Yaakov, K. et al. Axonal transcription factors disease. J. Cell Biol. 199, 437–451 (2012).
238–247 (2008). signal retrogradely in lesioned peripheral nerve. 78. Twiss, J. L. & Merianda, T. T. Old dogs with new tricks:
34. Hao, Y. et al. An evolutionarily conserved mechanism EMBO J. 31, 1350–1363 (2012). intra-​axonal translation of nuclear proteins. Neural
for cAMP elicited axonal regeneration involves direct This study shows that STAT3 is locally translated in Regen. Res. 10, 1560–1562 (2015).
activation of the dual leucine zipper kinase DLK. eLife the injured axon and retrogradely transported to 79. Liu, P. H., Tsai, H. Y., Chung, Y. W., Wang, Y. J. &
5, e14048 (2016). the nucleus to promote sensory neuron survival. Tseng, G. F. The proximity of the lesion to cell bodies
This study demonstrates that the cAMP–PKA 58. Hanz, S. et al. Axoplasmic importins enable determines the free radical risk induced in rat
pathway activates the DLK pathway, which is retrograde injury signaling in lesioned nerve. Neuron rubrospinal neurons subjected to axonal injury.
necessary for axon regeneration. 40, 1095–1104 (2003). Anat. Embryol. (Berl.) 207, 439–451 (2004).
35. Hammarlund, M., Nix, P., Hauth, L., Jorgensen, E. M. 59. O’Donovan, K. J. et al. B-​RAF kinase drives 80. Liu, P. H., Yang, L. H., Wang, T. Y., Wang, Y. J. &
& Bastiani, M. Axon regeneration requires a developmental axon growth and promotes axon Tseng, G. F. Proximity of lesioning determines
conserved MAP kinase pathway. Science 323, regeneration in the injured mature CNS. J. Exp. Med. response of facial motoneurons to peripheral axotomy.
802–806 (2009). 211, 801–814 (2014). J. Neurotrauma 23, 1857–1873 (2006).

www.nature.com/nrn
© 2018 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
Reviews

81. Villegas-​Perez, M. P., Vidal-​Sanz, M., Rasminsky, M., 104. Smith, D. S. & Skene, J. H. A transcription-​dependent 126. Mehta, S. T., Luo, X., Park, K. K., Bixby, J. L. &
Bray, G. M. & Aguayo, A. J. Rapid and protracted switch controls competence of adult neurons for Lemmon, V. P. Hyperactivated Stat3 boosts axon
phases of retinal ganglion cell loss follow axotomy in distinct modes of axon growth. J. Neurosci. 17, regeneration in the CNS. Exp. Neurol. 280,
the optic nerve of adult rats. J. Neurobiol. 24, 23–36 646–658 (1997). 115–120 (2016).
(1993). This study establishes that successful axon 127. Qiu, J., Cafferty, W. B., McMahon, S. B. &
82. Berkelaar, M., Clarke, D. B., Wang, Y. C., Bray, G. M. & regeneration requires a transcription-​dependent Thompson, S. W. Conditioning injury-​induced spinal
Aguayo, A. J. Axotomy results in delayed death and phase. axon regeneration requires signal transducer and
apoptosis of retinal ganglion cells in adult rats. 105. Moore, D. L. & Goldberg, J. L. Multiple transcription activator of transcription 3 activation. J. Neurosci. 25,
J. Neurosci. 14, 4368–4374 (1994). factor families regulate axon growth and regeneration. 1645–1653 (2005).
83. Rishal, I. et al. A motor-​driven mechanism for Dev. Neurobiol. 71, 1186–1211 (2011). 128. Lingor, P. et al. ROCK inhibition and CNTF interact on
cell-​length sensing. Cell Rep. 1, 608–616 (2012). 106. Blackmore, M. G. Molecular control of axon growth: intrinsic signalling pathways and differentially regulate
84. Brochier, C., Jones, J. I., Willis, D. E. & Langley, B. insights from comparative gene profiling and survival and regeneration in retinal ganglion cells.
Poly(ADP-​ribose) polymerase 1 is a novel target to high-​throughput screening. Int. Rev. Neurobiol. 105, Brain 131, 250–263 (2008).
promote axonal regeneration. Proc. Natl Acad. Sci. 39–70 (2012). 129. Smith, P. D. et al. SOCS3 deletion promotes optic
USA 112, 15220–15225 (2015). 107. Xu, J., Du, Y. & Deng, H. Direct lineage reprogramming: nerve regeneration in vivo. Neuron 64, 617–623
85. Byrne, A. B. et al. Inhibiting poly(ADP-​ribosylation) strategies, mechanisms, and applications. Cell Stem Cell (2009).
improves axon regeneration. eLife 5, e12734 (2016). 16, 119–134 (2015). 130. Sun, F. et al. Sustained axon regeneration induced
This study demonstrates that PARylation is a 108. Gey, M. et al. Atf3 mutant mice show reduced axon by co-​deletion of PTEN and SOCS3. Nature 480,
conserved regulator of axon regeneration that regeneration and impaired regeneration-​associated 372–375 (2011).
functions downstream of DLK signalling. gene induction after peripheral nerve injury. Open Biol. 131. Moore, D. L. et al. KLF family members regulate
86. Wang, X. et al. Inhibition of poly-​ADP-ribosylation fails 6, 160091 (2016). intrinsic axon regeneration ability. Science 326,
to increase axonal regeneration or improve functional 109. Seijffers, R., Mills, C. D. & Woolf, C. J. ATF3 increases 298–301 (2009).
recovery after adult mammalian CNS injury. eNeuro the intrinsic growth state of DRG neurons to enhance This study establishes that the developmentally
https://doi.org/10.1523/ENEURO.0270-16.2016 peripheral nerve regeneration. J. Neurosci. 27, regulated KLF family of transcription factors
(2016). 7911–7920 (2007). regulates axon regeneration.
87. Yiu, G. & He, Z. Glial inhibition of CNS axon 110. Venkatesh, I., Simpson, M. T., Coley, D. M. & Blackmore, 132. Blackmore, M. G. et al. Kruppel-​like Factor 7
regeneration. Nat. Rev. Neurosci. 7, 617–627 M. G. Epigenetic profiling reveals a developmental engineered for transcriptional activation promotes
(2006). decrease in promoter accessibility during cortical axon regeneration in the adult corticospinal tract.
88. Kosmaczewski, S. G. et al. RNA ligation in neurons by maturation in vivo. Neuroepigenetics 8, 19–26 (2016). Proc. Natl Acad. Sci. USA 109, 7517–7522 (2012).
RtcB inhibits axon regeneration. Proc. Natl Acad. Sci. This study finds that, although there is an increase 133. Moore, D. L., Apara, A. & Goldberg, J. L. Kruppel-​like
USA 112, 8451–8456 (2015). in H3K27me3 at RAG promoters in cortical neurons transcription factors in the nervous system: novel
89. Song, Y. et al. Regulation of axon regeneration by the during development, manipulation of HDACs or players in neurite outgrowth and axon regeneration.
RNA repair and splicing pathway. Nat. Neurosci. 18, HATs does not further promote regeneration. Mol. Cell. Neurosci. 47, 233–243 (2011).
817–825 (2015). 111. Seijffers, R., Allchorne, A. J. & Woolf, C. J. The 134. Apara, A. et al. KLF9 and JNK3 interact to suppress
90. Nix, P. et al. Axon regeneration genes identified transcription factor ATF-3 promotes neurite outgrowth. axon regeneration in the adult CNS. J. Neurosci. 37,
by RNAi screening in C. elegans. J. Neurosci. 34, Mol. Cell. Neurosci. 32, 143–154 (2006). 9632–9644 (2017).
629–645 (2014). 112. Lerch, J. K., Martinez-​Ondaro, Y. R., Bixby, J. L. & 135. Blackmore, M. G. et al. High content screening of
91. Onate, M. et al. Activation of the unfolded protein Lemmon, V. P. cJun promotes CNS axon growth. cortical neurons identifies novel regulators of axon
response promotes axonal regeneration after Mol. Cell. Neurosci. 59, 97–105 (2014). growth. Mol. Cell. Neurosci. 44, 43–54 (2010).
peripheral nerve injury. Sci. Rep. 6, 21709 (2016). 113. Simpson, M. T. et al. The tumor suppressor HHEX 136. Veldman, M. B., Bemben, M. A., Thompson, R. C. &
92. Berry, M., Ahmed, Z., Morgan-​Warren, P., Fulton, D. & inhibits axon growth when prematurely expressed in Goldman, D. Gene expression analysis of zebrafish
Logan, A. Prospects for mTOR-​mediated functional developing central nervous system neurons. Mol. Cell. retinal ganglion cells during optic nerve regeneration
repair after central nervous system trauma. Neurobiol. Neurosci. 68, 272–283 (2015). identifies KLF6a and KLF7a as important regulators of
Dis. 85, 99–110 (2016). 114. Chandran, V. et al. A systems-​level analysis of the axon regeneration. Dev. Biol. 312, 596–612 (2007).
93. Park, K. K. et al. Promoting axon regeneration in the peripheral nerve intrinsic axonal growth program. 137. Belin, S. et al. Injury-​induced decline of intrinsic
adult CNS by modulation of the PTEN/mTOR pathway. Neuron 89, 956–970 (2016). regenerative ability revealed by quantitative proteomics.
Science 322, 963–966 (2008). This study uses a multilevel bioinformatics approach Neuron 86, 1000–1014 (2015).
94. Abe, N., Borson, S. H., Gambello, M. J., Wang, F. & to identify pharmacological compounds that 138. Cho, Y. et al. Activating injury-​responsive genes with
Cavalli, V. Mammalian target of rapamycin (mTOR) enhance axon regeneration. hypoxia enhances axon regeneration through neuronal
activation increases axonal growth capacity of injured 115. Raivich, G. et al. The AP-1 transcription factor c-​Jun is HIF-1alpha. Neuron 88, 720–734 (2015).
peripheral nerves. J. Biol. Chem. 285, 28034–28043 required for efficient axonal regeneration. Neuron 43, 139. Jankowski, M. P. et al. Sox11 transcription factor
(2010). 57–67 (2004). modulates peripheral nerve regeneration in adult
95. Liu, K. et al. PTEN deletion enhances the regenerative 116. Ruff, C. A. et al. Neuronal c-​Jun is required for mice. Brain Res. 1256, 43–54 (2009).
ability of adult corticospinal neurons. Nat. Neurosci. successful axonal regeneration, but the effects of 140. Jankowski, M. P., Cornuet, P. K., McIlwrath, S.,
13, 1075–1081 (2010). phosphorylation of its N-​terminus are moderate. Koerber, H. R. & Albers, K. M. SRY-​box containing
96. Christie, K. J., Webber, C. A., Martinez, J. A., Singh, B. J. Neurochem. 121, 607–618 (2012). gene 11 (Sox11) transcription factor is required for
& Zochodne, D. W. PTEN inhibition to facilitate 117. Zou, H., Ho, C., Wong, K. & Tessier-​Lavigne, M. neuron survival and neurite growth. Neuroscience
intrinsic regenerative outgrowth of adult peripheral Axotomy-​induced Smad1 activation promotes axonal 143, 501–514 (2006).
axons. J. Neurosci. 30, 9306–9315 (2010). growth in adult sensory neurons. J. Neurosci. 29, 141. Jing, X., Wang, T., Huang, S., Glorioso, J. C. &
97. Chen, W. et al. Rapamycin-​resistant mTOR activity is 7116–7123 (2009). Albers, K. M. The transcription factor Sox11 promotes
required for sensory axon regeneration induced by a 118. Saijilafu et al. PI3K-​GSK3 signalling regulates nerve regeneration through activation of the
conditioning lesion. eNeuro https://doi.org/10.1523/ mammalian axon regeneration by inducing the regeneration-​associated gene Sprr1a. Exp. Neurol.
ENEURO.0358-16.2016 (2016). expression of Smad1. Nat. Commun. 4, 2690 (2013). 233, 221–232 (2012).
98. Al-​Ali, H. et al. The mTOR substrate S6 kinase 1 119. Finelli, M. J., Wong, J. K. & Zou, H. Epigenetic 142. Wang, Z., Reynolds, A., Kirry, A., Nienhaus, C. &
(S6K1) is a negative regulator of axon regeneration regulation of sensory axon regeneration after spinal Blackmore, M. G. Overexpression of Sox11 promotes
and a potential drug target for central nervous system cord injury. J. Neurosci. 33, 19664–19676 (2013). corticospinal tract regeneration after spinal injury
injury. J. Neurosci. 37, 7079–7095 (2017). This study reveals that histone-​modifying enzymes while interfering with functional recovery. J. Neurosci.
This study finds that treatment with a specific work together with SMAD1 to facilitate an increase 35, 3139–3145 (2015).
S6K1 inhibitor promotes cortical spinal tract in histone H4 acetylation and transcription of RAGs 143. Norsworthy, M. W. et al. Sox11 expression promotes
regeneration and behavioural recovery. and to promote axon regeneration. regeneration of some retinal ganglion cell types but
99. Duan, X. et al. Subtype-​specific regeneration of 120. Tedeschi, A., Omura, T. & Costigan, M. CNS repair and kills others. Neuron 94, 1112–1120.e4 (2017).
retinal ganglion cells following axotomy: effects of axon regeneration: using genetic variation to determine This study reveals that expression of SOX11
osteopontin and mTOR signaling. Neuron 85, mechanisms. Exp. Neurol. 287, 409–422 (2017). reactivates an axon growth programme and
1244–1256 (2015). 121. Omura, T. et al. Robust axonal regeneration occurs promotes axon regeneration in a subset of adult
100. Roundtree, I. A., Evans, M. E., Pan, T. & He, C. in the injured CAST/Ei mouse CNS. Neuron 86, RGCs but kills other types of RGCs.
Dynamic RNA modifications in gene expression 1215–1227 (2015). 144. Di Giovanni, S. et al. The tumor suppressor protein
regulation. Cell 169, 1187–1200 (2017). 122. Hill, C. S. Transcriptional control by the SMADs. p53 is required for neurite outgrowth and axon
101. Leighton, L. J. et al. Experience-​dependent neural Cold Spring Harb. Perspect. Biol. 8, a022079 regeneration. EMBO J. 25, 4084–4096 (2006).
plasticity, learning, and memory in the era of (2016). 145. Stern, S. & Knoll, B. CNS axon regeneration inhibitors
epitranscriptomics. Genes Brain Behav. https://doi. 123. Schwaiger, F. W. et al. Peripheral but not central stimulate an immediate early gene response via MAP
org/10.1111/gbb.12426 (2017). axotomy induces changes in Janus kinases (JAK) and kinase-​SRF signaling. Mol. Brain 7, 86 (2014).
102. Yoon, K. J. et al. Temporal control of mammalian signal transducers and activators of transcription 146. Venkatesh, I. & Blackmore, M. G. Selecting optimal
cortical neurogenesis by m(6)A methylation. Cell 171, (STAT). Eur. J. Neurosci. 12, 1165–1176 (2000). combinations of transcription factors to promote axon
877–889.e17 (2017). 124. Bareyre, F. M. et al. In vivo imaging reveals a regeneration: why mechanisms matter. Neurosci. Lett.
103. Weng, Y. L. et al. Epitranscriptomic m(6)A regulation phase-​specific role of STAT3 during central and 652, 64–73 (2017).
of axon regeneration in the adult mammalian nervous peripheral nervous system axon regeneration. 147. Fagoe, N. D., van Heest, J. & Verhaagen, J. Spinal cord
system. Neuron 97, 313–325.e6 (2018). Proc. Natl Acad. Sci. USA 108, 6282–6287 injury and the neuron-​intrinsic regeneration-​associated
This study uncovers an epitranscriptomic (2011). gene program. Neuromolecular Med. 16, 799–813
mechanism in which axon injury elevates 125. Lang, C., Bradley, P. M., Jacobi, A., Kerschensteiner, (2014).
N6-methyladenosine levels in regeneration-​ M. & Bareyre, F. M. STAT3 promotes corticospinal 148. Fagoe, N. D., Attwell, C. L., Kouwenhoven, D.,
associated transcripts, which is essential for remodelling and functional recovery after spinal cord Verhaagen, J. & Mason, M. R. Overexpression of ATF3
functional peripheral axon regeneration. injury. EMBO Rep. 14, 931–937 (2013). or the combination of ATF3, c-​Jun, STAT3 and Smad1

Nature Reviews | Neuroscience


© 2018 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
Reviews

promotes regeneration of the central axon branch 173. Lu, C. & Thompson, C. B. Metabolic regulation of 199. Ali, T., Renkawitz, R. & Bartkuhn, M. Insulators and
of sensory neurons but without synergistic effects. epigenetics. Cell Metab. 16, 9–17 (2012). domains of gene expression. Curr. Opin. Genet. Dev.
Hum. Mol. Genet. 24, 6788–6800 (2015). 174. Pollema-​Mays, S. L., Centeno, M. V., Apkarian, A. V. & 37, 17–26 (2016).
149. Takahashi, K. & Yamanaka, S. Induction of pluripotent Martina, M. Expression of DNA methyltransferases in 200. Kim, T. K. et al. Widespread transcription at neuronal
stem cells from mouse embryonic and adult fibroblast adult dorsal root ganglia is cell-​type specific and up activity-​regulated enhancers. Nature 465, 182–187
cultures by defined factors. Cell 126, 663–676 (2006). regulated in a rodent model of neuropathic pain. (2010).
150. Takahashi, K. & Yamanaka, S. A decade of transcription Front. Cell. Neurosci. 8, 217 (2014). 201. Bonev, B. & Cavalli, G. Organization and function
factor-​mediated reprogramming to pluripotency. 175. Zhao, J. Y. et al. DNA methyltransferase DNMT3a of the 3D genome. Nat. Rev. Genet. 17, 661–678
Nat. Rev. Mol. Cell Biol. 17, 183–193 (2016). contributes to neuropathic pain by repressing (2016).
151. Chronis, C. et al. Cooperative binding of transcription Kcna2 in primary afferent neurons. Nat. Commun. 8, 202. Ong, C. T. & Corces, V. G. Enhancer function: new
factors orchestrates reprogramming. Cell 168, 14712 (2017). insights into the regulation of tissue-​specific gene
442–459.e20 (2017). 176. Weng, Y. L., Joseph, J., An, R., Song, H. & Ming, G. L. expression. Nat. Rev. Genet. 12, 283–293 (2011).
152. Lee, T. I. & Young, R. A. Transcriptional regulation and Epigenetic regulation of axonal regenerative capacity. 203. Cartoni, R. et al. The mammalian-​specific protein
its misregulation in disease. Cell 152, 1237–1251 Epigenomics 8, 1429–1442 (2016). Armcx1 regulates mitochondrial transport during
(2013). 177. Bachman, M. et al. 5-Hydroxymethylcytosine is a axon regeneration. Neuron 92, 1294–1307 (2016).
153. Guo, C. & Morris, S. A. Engineering cell identity: predominantly stable DNA modification. Nat. Chem. 204. Han, S. M., Baig, H. S. & Hammarlund, M.
establishing new gene regulatory and chromatin 6, 1049–1055 (2014). Mitochondria localize to injured axons to support
landscapes. Curr. Opin. Genet. Dev. 46, 50–57 178. Iskandar, B. J. et al. Folate regulation of axonal regeneration. Neuron 92, 1308–1323 (2016).
(2017). regeneration in the rodent central nervous system 205. Sainath, R. et al. Chondroitin sulfate proteoglycans
154. Iwafuchi-​Doi, M. & Zaret, K. S. Pioneer transcription through DNA methylation. J. Clin. Invest. 120, negatively regulate the positioning of mitochondria
factors in cell reprogramming. Genes Dev. 28, 1603–1616 (2010). and endoplasmic reticulum to distal axons.
2679–2692 (2014). 179. Loh, Y. E. et al. Comprehensive mapping of Dev. Neurobiol. 77, 1351–1370 (2017).
155. Zaret, K. S. & Mango, S. E. Pioneer transcription 5-hydroxymethylcytosine epigenetic dynamics in axon 206. Seo, J., Singh, N. N., Ottesen, E. W., Lee, B. M. &
factors, chromatin dynamics, and cell fate control. regeneration. Epigenetics 12, 77–92 (2017). Singh, R. N. A novel human-​specific splice isoform
Curr. Opin. Genet. Dev. 37, 76–81 (2016). 180. Ghibaudi, M., Boido, M. & Vercelli, A. Functional alters the critical C-​terminus of Survival Motor
156. Gifford, C. A. & Meissner, A. Epigenetic obstacles integration of complex miRNA networks in central Neuron protein. Sci. Rep 6, 30778 (2016).
encountered by transcription factors: reprogramming and peripheral lesion and axonal regeneration. 207. Li, M. et al. A human-​specific AS3MT isoform and
against all odds. Curr. Opin. Genet. Dev. 22, 409–415 Prog. Neurobiol. 158, 69–93 (2017). BORCS7 are molecular risk factors in the 10q24.32
(2012). 181. Yates, L. A., Norbury, C. J. & Gilbert, R. J. The long schizophrenia-​associated locus. Nat. Med. 22,
157. Mo, A. et al. Epigenomic signatures of neuronal and short of microRNA. Cell 153, 516–519 (2013). 649–656 (2016).
diversity in the mammalian brain. Neuron 86, 182. Wu, D., Raafat, A., Pak, E., Clemens, S. & Murashov, 208. Stahl, P. D. & Wainszelbaum, M. J. Human-​specific
1369–1384 (2015). A. K. Dicer-​microRNA pathway is critical for peripheral genes may offer a unique window into human cell
158. Su, Y. et al. Neuronal activity modifies the chromatin nerve regeneration and functional recovery in vivo and signaling. Sci Signal. 2, pe59 (2009).
accessibility landscape in the adult brain. Nat. Neurosci. regenerative axonogenesis in vitro. Exp. Neurol. 233, 209. Aldiri, I. et al. The dynamic epigenetic landscape
20, 476–483 (2017). 555–565 (2012). of the retina during development, reprogramming,
159. Hu, G. et al. Single-​cell RNA-​seq reveals distinct injury 183. Wu, D., Raafat, M., Pak, E., Hammond, S. & and tumorigenesis. Neuron 94, 550–568.e10 (2017).
responses in different types of DRG sensory neurons. Murashov, A. K. MicroRNA machinery responds to 210. Geoffroy, C. G., Meves, J. M. & Zheng, B. The age
Sci. Rep. 6, 31851 (2016). peripheral nerve lesion in an injury-​regulated pattern. factor in axonal repair after spinal cord injury: a focus
This study uses single-​cell RNAseq to reveal Neuroscience 190, 386–397 (2011). on neuron-​intrinsic mechanisms. Neurosci. Lett. 652,
distinctive and sustained heterogeneity of 184. Emde, A. & Hornstein, E. miRNAs at the interface of 41–49 (2017).
transcriptomic responses to axon injury at the cellular stress and disease. EMBO J. 33, 1428–1437 211. Painter, M. W. et al. Diminished Schwann cell repair
single-​neuron level. (2014). responses underlie age-​associated impaired axonal
160. Arlotta, P. et al. Neuronal subtype-​specific genes that 185. Maurel, M. & Chevet, E. Endoplasmic reticulum stress regeneration. Neuron 83, 331–343 (2014).
control corticospinal motor neuron development signaling: the microRNA connection. Am. J. Physiol. 212. von Schimmelmann, M. et al. Polycomb repressive
in vivo. Neuron 45, 207–221 (2005). Cell Physiol. 304, C1117–C1126 (2013). complex 2 (PRC2) silences genes responsible for
161. Nichterwitz, S., Benitez, J. A., Hoogstraaten, R., Deng, 186. Ying, Z. et al. The unfolded protein response and neurodegeneration. Nat. Neurosci. 19, 1321–1330
Q. & Hedlund, E. LCM-​Seq: a method for spatial cholesterol biosynthesis link Luman/CREB3 to (2016).
transcriptomic profiling using laser capture regenerative axon growth in sensory neurons. 213. Richardson, P. M. & Issa, V. M. Peripheral injury
microdissection coupled with PolyA-​based RNA J. Neurosci. 35, 14557–14570 (2015). enhances central regeneration of primary sensory
sequencing. Methods Mol. Biol. 1649, 95–110 187. Hu, Y. et al. Differential effects of unfolded protein neurones. Nature 309, 791–793 (1984).
(2018). response pathways on axon injury-​induced death 214. McQuarrie, I. G. & Grafstein, B. Axon outgrowth
162. Doyle, J. P. et al. Application of a translational profiling of retinal ganglion cells. Neuron 73, 445–452 enhanced by a previous nerve injury. Arch. Neurol. 29,
approach for the comparative analysis of CNS cell (2012). 53–55 (1973).
types. Cell 135, 749–762 (2008). 188. Sun, A. X., Crabtree, G. R. & Yoo, A. S. MicroRNAs: 215. Neumann, S. & Woolf, C. J. Regeneration of dorsal
163. Yao, B. et al. Epigenetic mechanisms in neurogenesis. regulators of neuronal fate. Curr. Opin. Cell Biol. 25, column fibers into and beyond the lesion site following
Nat. Rev. Neurosci. 17, 537–549 (2016). 215–221 (2013). adult spinal cord injury. Neuron 23, 83–91 (1999).
164. Cholewa-​Waclaw, J. et al. The role of epigenetic 189. Motti, D. et al. Identification of miRNAs involved in 216. Tuszynski, M. H. & Steward, O. Concepts and methods
mechanisms in the regulation of gene expression in DRG neurite outgrowth and their putative targets. for the study of axonal regeneration in the CNS.
the nervous system. J. Neurosci. 36, 11427–11434 FEBS Lett. 591, 2091–2105 (2017). Neuron 74, 777–791 (2012).
(2016). 190. Strickland, I. T. et al. Axotomy-​induced miR-21 217. Ambron, R. T. & Walters, E. T. Priming events and
165. Zhao, Y. & Garcia, B. A. Comprehensive catalog of promotes axon growth in adult dorsal root ganglion retrograde injury signals. A new perspective on the
currently documented histone modifications. Cold neurons. PLOS ONE 6, e23423 (2011). cellular and molecular biology of nerve regeneration.
Spring Harb. Perspect. Biol. 7, a025064 (2015). 191. Zhang, H. Y. et al. MicroRNAs 144, 145, and 214 are Mol. Neurobiol. 13, 61–79 (1996).
166. Puttagunta, R. et al. PCAF-​dependent epigenetic down-​regulated in primary neurons responding to 218. Tazaki, A., Tanaka, E. M. & Fei, J. Salamander spinal
changes promote axonal regeneration in the central sciatic nerve transection. Brain Res. 1383, 62–70 cord regeneration: the ultimate positive control in
nervous system. Nat. Commun. 5, 3527 (2014). (2011). vertebrate spinal cord regeneration. Dev. Biol. 432,
This study shows that KAT2B functions 192. Zhou, S. et al. microRNA-222 targeting PTEN 63–71 (2017).
downstream of ERK signalling to increase histone promotes neurite outgrowth from adult dorsal root 219. Graff, J., Kim, D., Dobbin, M. M. & Tsai, L. H.
H3 acetylation at the promoters of key RAGs ganglion neurons following sciatic nerve transection. Epigenetic regulation of gene expression in
following peripheral but not central axonal injury. PLOS ONE 7, e44768 (2012). physiological and pathological brain processes.
167. Cho, Y. & Cavalli, V. HDAC5 is a novel injury-​regulated 193. Wu, D. & Murashov, A. K. MicroRNA-431 regulates Physiol. Rev. 91, 603–649 (2011).
tubulin deacetylase controlling axon regeneration. axon regeneration in mature sensory neurons by 220. Wang, Z., Tang, B., He, Y. & Jin, P. DNA methylation
EMBO J. 31, 3063–3078 (2012). targeting the Wnt antagonist Kremen1. Front. Mol. dynamics in neurogenesis. Epigenomics 8, 401–414
168. Pelzel, H. R., Schlamp, C. L. & Nickells, R. W. Histone Neurosci 6, 35 (2013). (2016).
H4 deacetylation plays a critical role in early gene 194. Lisi, V. et al. Enhanced neuronal regeneration in the 221. Guo, J. U. et al. Neuronal activity modifies the DNA
silencing during neuronal apoptosis. BMC Neurosci. CAST/Ei mouse strain is linked to expression of methylation landscape in the adult brain. Nat. Neurosci.
11, 62 (2010). differentiation markers after injury. Cell Rep. 20, 14, 1345–1351 (2011).
169. Schmitt, H. M., Pelzel, H. R., Schlamp, C. L. & Nickells, 1136–1147 (2017). 222. Calo, E. & Wysocka, J. Modification of enhancer
R. W. Histone deacetylase 3 (HDAC3) plays an 195. Li, Z. & Rana, T. M. Therapeutic targeting of microRNAs: chromatin: what, how, and why? Mol. Cell 49,
important role in retinal ganglion cell death after acute current status and future challenges. Nat. Rev. Drug 825–837 (2013).
optic nerve injury. Mol. Neurodegener. 9, 39 (2014). Discov. 13, 622–638 (2014). 223. Zhu, H., Wang, G. & Qian, J. Transcription factors
170. Lv, L., Han, X., Sun, Y., Wang, X. & Dong, Q. Valproic 196. Niemi, J. P. et al. A critical role for macrophages as readers and effectors of DNA methylation.
acid improves locomotion in vivo after SCI and axonal near axotomized neuronal cell bodies in stimulating Nat. Rev. Genet. 17, 551–565 (2016).
growth of neurons in vitro. Exp. Neurol. 233, nerve regeneration. J. Neurosci. 33, 16236–16248 224. Gonzalez-​Sandoval, A. & Gasser, S. M. On TADs
783–790 (2012). (2013). and LADs: spatial control over gene expression.
171. Riccio, A. Dynamic epigenetic regulation in neurons: 197. Kwon, M. J. et al. Contribution of macrophages to Trends Genet. 32, 485–495 (2016).
enzymes, stimuli and signaling pathways. enhanced regenerative capacity of dorsal root ganglia 225. Ong, C. T. & Corces, V. G. CTCF: an architectural
Nat. Neurosci. 13, 1330–1337 (2010). sensory neurons by conditioning injury. J. Neurosci. protein bridging genome topology and function.
172. Bhat, N., Park, J., Zoghbi, H. Y., Arthur, J. S. & 33, 15095–15108 (2013). Nat. Rev. Genet. 15, 234–246 (2014).
Zaret, K. S. The chromatin modifier MSK1/2 suppresses 198. Ciabrelli, F. & Cavalli, G. Chromatin-​driven behavior of 226. Bonev, B. et al. Multiscale 3D genome rewiring during
endocrine cell fates during mouse pancreatic topologically associating domains. J. Mol. Biol. 427, mouse neural development. Cell 171, 557–572.e24
development. PLOS ONE 11, e0166703 (2016). 608–625 (2015). (2017).

www.nature.com/nrn
© 2018 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
Reviews

This study performs an ultra-​high-resolution Hi-​C 237. Obata, K. & Noguchi, K. MAPK activation in 248. Gaub, P. et al. HDAC inhibition promotes neuronal
mapping of mouse neural differentiation and nociceptive neurons and pain hypersensitivity. Life Sci. outgrowth and counteracts growth cone collapse
provides insights into the factors that influence the 74, 2643–2653 (2004). through CBP/p300 and P/CAF-​dependent p53
dynamics of chromatin interactions during neuronal 238. Mikuzuki, L. et al. Phenotypic change in trigeminal acetylation. Cell Death Differ. 17, 1392–1408
development. ganglion neurons associated with satellite cell (2010).
227. Soufi, A. & Zaret, K. S. Understanding impediments to activation via extracellular signal-​regulated kinase 249. Parikh, P. et al. Regeneration of axons in injured
cellular conversion to pluripotency by assessing the phosphorylation is involved in lingual neuropathic spinal cord by activation of bone morphogenetic
earliest events in ectopic transcription factor binding pain. Eur. J. Neurosci. 46, 2190–2202 (2017). protein/Smad1 signaling pathway in adult
to the genome. Cell Cycle 12, 1487–1491 (2013). 239. Pannese, E. Perikaryal surface specializations of neurons. Proc. Natl Acad. Sci. USA 108, E99–E107
228. Valentin, G. in Muller’s Archiv fur Anatomie, neurons in sensory ganglia. Int. Rev. Cytol. 220, (2011).
Physiologie und wissenschaftliche Medicin (ed. Müller, 1–34 (2002). 250. Smith, R. P. et al. Transcriptional profiling of intrinsic
J.) 139–164. (Veit et Comp, Berlin, 1839). 240. Hanani, M., Huang, T. Y., Cherkas, P. S., Ledda, M. & PNS factors in the postnatal mouse. Mol. Cell. Neurosci.
229. Pannese, E. Number and structure of perisomatic Pannese, E. Glial cell plasticity in sensory ganglia 46, 32–44 (2011).
satellite cells of spinal ganglia under normal induced by nerve damage. Neuroscience 114,
conditions or during axon regeneration and neuronal 279–283 (2002). Acknowledgements
hypertrophy. Z. Zellforsch. Mikrosk. Anat. 63, 241. Hanani, M. Satellite glial cells in sensory ganglia: The authors’ research on these topics has been generously
568–592 (1964). from form to function. Brain Res. Brain Res. Rev. 48, supported by the US National Institute of Health grants
230. Pannese, E. The structure of the perineuronal sheath 457–476 (2005). NS096034, NS082446 and NS099603, the University of
of satellite glial cells (SGCs) in sensory ganglia. Neuron 242. Vit, J. P., Ohara, P. T., Bhargava, A., Kelley, K. & Missouri Spinal Cord Injury Research Program and a Philip
Glia Biol. 6, 3–10 (2010). Jasmin, L. Silencing the Kir4.1 potassium channel and Sima K. Needleman Doctoral Fellowship. The authors
231. Christie, K. et al. Intraganglionic interactions between subunit in satellite glial cells of the rat trigeminal thank H. Gabel for helpful comments and critical reading of
satellite cells and adult sensory neurons. Mol. Cell. ganglion results in pain-​like behavior in the absence of the manuscript. The authors thank the Cavalli laboratory
Neurosci. 67, 1–12 (2015). nerve injury. J. Neurosci. 28, 4161–4171 (2008). members for their helpful comments on the manuscript. The
232. Fenzi, F., Benedetti, M. D., Moretto, G. & Rizzuto, N. 243. Humbertson, A. Jr., Zimmermann, E. & Leedy, M. A authors apologize to those whose studies could not be cited
Glial cell and macrophage reactions in rat spinal chronological study of mitotic activity in satellite owing to space limitation.
ganglion after peripheral nerve lesions: an cell hyperplasia associated with chromatolytic
immunocytochemical and morphometric study. neurons. Z. Zellforsch. Mikrosk. Anat. 100, 507–515 Author contributions
Arch. Ital. Biol. 139, 357–365 (2001). (1969). M.M. and V.C. researched data for the article, made substantial
233. Xie, W., Strong, J. A. & Zhang, J. M. Early blockade of 244. Donegan, M., Kernisant, M., Cua, C., Jasmin, L. & contributions to discussions of the content, wrote the article
injured primary sensory afferents reduces glial cell Ohara, P. T. Satellite glial cell proliferation in the and reviewed and/or edited the manuscript before submission.
activation in two rat neuropathic pain models. trigeminal ganglia after chronic constriction injury of
Neuroscience 160, 847–857 (2009). the infraorbital nerve. Glia 61, 2000–2008 (2013). Competing interests
234. Zhang, H. et al. Altered functional properties of 245. Huang, L. Y., Gu, Y. & Chen, Y. Communication The authors declare no competing interests.
satellite glial cells in compressed spinal ganglia. Glia between neuronal somata and satellite glial cells in
57, 1588–1599 (2009). sensory ganglia. Glia 61, 1571–1581 (2013). Publisher’s note
235. Xu, M., Aita, M. & Chavkin, C. Partial infraorbital 246. Jessen, K. R. & Mirsky, R. The repair Schwann cell and Springer Nature remains neutral with regard to jurisdictional
nerve ligation as a model of trigeminal nerve injury its function in regenerating nerves. J. Physiol. 594, claims in published maps and institutional affiliations.
in the mouse: behavioral, neural, and glial reactions. 3521–3531 (2016).
J. Pain 9, 1036–1048 (2008). 247. Cho, Y., Park, D. & Cavalli, V. Filamin A is required Reviewer information
236. Warwick, R. A. & Hanani, M. The contribution of in injured axons for HDAC5 activity and axon Nature Reviews Neuroscience thanks S. Di Giovanni, J. Twiss
satellite glial cells to chemotherapy-​induced regeneration. J. Biol. Chem. 290, 22759–22770 and the other anonymous reviewer(s), for their contribution
neuropathic pain. Eur. J. Pain 17, 571–580 (2013). (2015). to the peer review of this work.

Nature Reviews | Neuroscience


© 2018 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.

Potrebbero piacerti anche