Anda di halaman 1dari 13

Progress in Retinal and Eye Research 56 (2017) 19e31

Contents lists available at ScienceDirect

Progress in Retinal and Eye Research


journal homepage: www.elsevier.com/locate/prer

Retinal regeneration mechanisms linked to multiple cancer


molecules: A therapeutic conundrum
Amanda Barber a, 1, Kyle Farmer b, 1, Keith R. Martin a, c, d, 1, Patrice D. Smith a, b, *, 1
a
John van Geest Centre for Brain Repair, University of Cambridge, UK
b
Department of Neuroscience, Carleton University, Ottawa, Ontario, Canada
c
Medical Research Council e Wellcome Trust Cambridge Stem Cell Institute, Cambridge, UK
d
Cambridge NIHR Biomedical Research Centre, Cambridge, UK

a r t i c l e i n f o a b s t r a c t

Article history: Over the last decade, a large number of research articles have been published demonstrating regener-
Received 27 October 2015 ation and/or neuroprotection of retinal ganglion cells following manipulation of specic genetic and
Received in revised form molecular targets. Interestingly, of the targets that have been identied to promote repair following
6 March 2016
visual system damage, many are genes known to be mutated in different types of cancer. This review
Accepted 7 March 2016
Available online 30 August 2016
explores recent literature on the potential for modulating cancer genes as a therapeutic strategy for
visual system repair and looks at the potential clinical challenges associated with implementing this type
of therapy. We also discuss signalling mechanisms that have been implicated in cancer and consider how
Keywords:
Retinal ganglion cells
similar mechanisms may improve axonal regeneration in the optic nerve.
Axon regeneration 2016 Elsevier Ltd. All rights reserved.
Cancer genes
PTEN
Visual system repair mechanisms

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 20
2. Signalling mechanisms involved in cancer and axon regeneration . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 20
2.1. PI3K/PTEN/mTOR/Akt signalling . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 20
2.2. Cytokine signalling and inflammatory mechanisms . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 22
2.3. Anti-apoptotic factors and their role in cancer biology and implications for optic nerve regeneration . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 23
2.4. Kruppel-like family (KLF) of transcription factors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 24
2.5. Rho/ROCK pathway . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 24
2.6. Haematopoietically Expressed Homeobox (HHEX) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 26
2.7. Epidermal growth factor receptor (EGFR) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 26

Abbreviations: AAV, Adeno-associated virus; CNS, Central Nervous System; CNTF, ciliary neurotrophic factor; DLK, dual leucine zipper kinase; DRG, dorsal root ganglion;
EGFR, Epidermal growth factor receptor; EMT, epithelial-mesenchymal transition; GAP-43, Growth associated protein 43; GBM, glioblastoma multiforme; GM-CSF, gran-
ulocyte macrophage-colony stimulating factor; gp130, glycoprotein 130; HCC, Hepatocellular carcinoma cells; HHEX, Hematopoietically Expressed Homeobox; hnRNP K,
heterogeneous nuclear ribonucleoprotein K; IL-6, Interleukin-6; Jak/STAT, Janus kinase-signal transduce and activator of transcription; KLF, Kruppel-like Family; LIF, Leukemia
inhibitory factor; mTOR, mammalian target of rapamycin; NF-M, medium neurolament; PI3K, phosphatidylinositol-3-kinase; PTEN, phosphatase and tensin homolog; Rb,
retinoblastoma protein; RGC, retinal ganglion cell; SOCS-3, suppressor of cytokine signalling 3; Sfpq, Splicing factor proline- and glutamine-rich protein; STAT-3, signal
transducer and activator of transcription-3; VEGF, Vascular endothelial growth factor.
* Corresponding author. Department of Neuroscience, Carleton University, Ottawa, Ontario, Canada.
E-mail address: Patrice.Smith@carleton.ca (P.D. Smith).
1
Percentage of work contributed by each author in the production of the manuscript is as follows: Patrice D. Smith: 50%, Amanda Barber: 30%, Kyle Farmer: 10%, Keith
Martin: 10%.

http://dx.doi.org/10.1016/j.preteyeres.2016.08.003
1350-9462/ 2016 Elsevier Ltd. All rights reserved.
20 A. Barber et al. / Progress in Retinal and Eye Research 56 (2017) 19e31

3. Less explored potential targets . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 26


3.1. Heterogeneous nuclear ribonucleoprotein (hnRNP) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 27
3.2. Rb/E2F family . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 27
4. Approaches to reducing cancer risk when promoting regeneration . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 27
5. Beyond axonal growth: challenges for repair . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 28
6. Conclusion and future directions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 29
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 29

1. Introduction the adult mammalian CNS. A major question is whether manipu-


lation of genes involved in cancer can be considered as a feasible
In recent years there has been an exponential increase in the mechanism to promote axon regeneration in a clinical setting,
number of publications reporting cell survival and/or axon regen- where the potential for oncogenesis is a concern. However, it is
eration in models of optic nerve injury, for example where intra- conceivable that the post-mitotic nature of neurons in the mature
ocular pressure is elevated to induce retinal ganglion cell (RGC) loss visual system may limit this risk of malignancy. In this review we
or following localised optic nerve crush. Numerous factors with will also consider how oncogenic risk might be mitigated through
diverse physiological functions, including mammalian target of careful selection and application of the pharmacological or mo-
rapamycin (mTOR), kruppel-like family (KLF) transcription factors, lecular tools used to manipulate cancer-associated pathways.
suppressor of cytokine signalling 3 (SOCS3) and cytokines (ciliary
neurotrophic factor (CNTF)/leukemia inhibitory factor (LIF)), have 2. Signalling mechanisms involved in cancer and axon
been suggested to play a key role in RGC survival and/or axon regeneration
regeneration. Proposed mechanisms include effects on the avail-
ability of neurotrophic factors, activation of intrinsic survival/ In this section we will focus on cancer genes and signalling
growth mechanisms, modulation of axonal transport and glia- mechanisms that have been proposed to underlie both malignancy
dependent support as well as inhibition of apoptotic mechanisms and regeneration in the mature nervous system.
in RGCs. Interestingly, many of the mechanisms that have been
suggested to promote either survival and/or axon regeneration in 2.1. PI3K/PTEN/mTOR/Akt signalling
the damaged visual system are directly or indirectly associated with
cancer, including malignancies that affect the eye and visual Many human cancers, including some tumours of the brain and
pathways. visual system, result from specic genetic alterations and there is
Cancer can be dened as the loss of normal cellular growth evidence that deletions in a particular region of chromosome 10 are
control where unregulated proliferation of cells result in malignant often involved (Kon et al., 1998; Li et al., 1997). As an example,
pathologies (De Potter et al., 2002). In contrast, failure of neuronal gliomas are the most common solid tumours in the CNS (brain,
regeneration can be considered as an intrinsic inability to re- spinal cord and visual system) and more than half of glioblastomas
activate the normal growth mechanisms that occur during devel- develop as a consequence of chromosome 10q loss (Bostrom et al.,
opment. In this sense, cancer and regenerative failure could be 1998). PTEN, a known tumour suppressor has been identied as a
viewed as different sides of the same coin. The central nervous candidate gene on chromosome 10q which is mutated in many
system (CNS) can be stimulated to regenerate by manipulation of a cancers and responsible for a large proportion of glioblastomas in
variety of cancer-associated molecular signalling pathways and the nervous system (Li et al., 1997).
excellent reviews have been published on this subject recently The PTEN protein is a 403 amino acid protein that is evolu-
(Benowitz et al., 2015; He and Jin, 2016; Kaplan et al., 2015; Smith tionarily conserved across species and through its many functional
et al., 2015). Of the identied regeneration-associated genes, many domains contributes to the regulation of a myriad of cellular
are tumour suppressors, including phosphatase and tensin homo- functions including cell survival, cellular growth, migration, pro-
log (PTEN (Park et al., 2008)) or oncogenes such as MYC (Buchser liferation, cellular homeostasis and metabolism (Fig. 1). PTEN
et al., 2010; Pomerantz and Blau, 2013). In this review, we will controls a number of complex cellular events at a transcriptional
focus on cancer genes and signalling mechanisms that have been level but also operates through post-translational modications of
proposed to underlie both malignancy and regeneration in the target proteins. Most well-known is its role as a regulator of the
mature nervous system. We will review several signalling path- PI3K pathway (Weng et al., 2001), which is mediated by the lipid
ways, including PTEN/mTOR pathway, phosphatidylinositol-3- phosphatase activity of PTEN that antagonises PI3K function in
kinase (PI3K), cytokine and inammatory factors including SOCS- many different physiological systems, including the brain. Inhibi-
3, trophic factors (such as CNTF), anti-apoptotic factors (including tion of PI3K, and downstream signalling via Akt, mTOR and S6 ki-
Bcl-2), KLF, transcription factors (including signal transducer and nase activity, results in arrested protein translation and cellular
activator of transcription-3 (STAT-3)) as well as additional proposed growth (Gao et al., 2000). Unsurprisingly, the phosphatase function
mechanisms that are known to be involved in cancer pathology but of PTEN is critical to its role as a tumour suppressor and loss of PTEN
that have also been implicated in axon regeneration. The optic function is known to play a role in initiating tumour pathogenesis
nerve crush model has been extensively utilized to evaluate the (Weng et al., 1999). Loss of the homeostatic and metabolic effects of
impact of modulating different signalling factors on axon regener- PTEN may also contribute to tumour formation (Cordero-Espinoza
ation in a physiological setting and has been important in dening and Hagen, 2013; Icard and Lincet, 2013). In addition, PTEN is
signalling mechanisms involved in promoting axon regeneration in intimately involved in regulating other tumour suppressor
A. Barber et al. / Progress in Retinal and Eye Research 56 (2017) 19e31 21

molecules and as such loss of PTEN function could amplify the loss (Song et al., 2012b), C. elegans GABA motor neurons (Byrne et al.,
of other tumour suppressors, potentiating the loss of control of 2014), zebrash dorsal root ganglion (DRG) neurons (Abe et al.,
cellular growth. Overall, loss of PTEN function may result in 2010) and blastemal cells following n amputation (Hirose et al.,
disruption of cell growth mechanisms, angiogenesis, cell prolifer- 2014). The role of PTEN and mTOR signalling in axon regenera-
ation and altered migration as well as dysregulation of other tion is functionally conserved across several highly divergent spe-
tumour suppressor pathways. cies suggest that axon regeneration and the mechanisms that
PTEN is a potent suppressor of axon regeneration and several regulate it are of ancient origin.
groups have demonstrated enhanced axon regeneration of RGCs Previous work has shown that conditional gene deletion using
(Park et al., 2008), mammalian peripheral sensory (sciatic) neuro- adeno-associated virus (AAV) mediated Cre deletion of oxed PTEN
nes (Christie et al., 2010) and corticospinal neurones (Liu et al., in adult mouse RGCs is not only effective in promoting cell survival
2010) following deletion or knockdown of PTEN, an effect mostly but also initiates long distance axon growth in vivo (de Lima et al.,
mediated by enhancing mTOR signalling. PTEN deletion/enhanced 2012; Park et al., 2008). Recently, Duan and colleagues demon-
mTOR signalling has been shown to be important for axon regen- strated that PTEN deletion specically promotes regeneration of a-
eration in a number regeneration-competent species; examples RGCs, a subtype of RGCs that preferentially survive post-axotomy
include Drosophila class IV dendritic arborisation (da) neurons and show high mTOR signalling activity compared to other RGC

Fig. 1. PTEN is one of the most widely studied pathways in both retinal regeneration and cancer elds. PTEN is a 403 amino acid phosphatase that acts as a primary regulator of the
PI3k/Akt/mTOR signalling pathways. The amino terminus consists of a membrane binding domain that interacts with phosphoinositides. A core phosphatase domain is important in
facilitating the phosphatase activity of the protein. The C-terminal contains a C2 domain which is important in anchoring PTEN to specic protein targets as well as the plasma
membrane. The C-terminal domain is post-translationally regulated through phosphorylation events and contains binding sites for proteins containing PDZ-domains (A). The
expected folding pattern of the protein is represented in (B). PTEN blocks the conversion of PIP2 into PIP3 thereby preventing the activation of Akt. With feedback control by PTEN,
Akt regulates various intracellular events including cell survival, regeneration and cell death processes (C).
22 A. Barber et al. / Progress in Retinal and Eye Research 56 (2017) 19e31

subtypes (Duan et al., 2015). Further studies have shown successful in the C-terminal domain of PTEN as several nonsense mutations
suppression of PTEN in adult wild type mouse corticospinal tract have been described in human tumours in the C-terminus of PTEN
axons using an RNAi (Zukor et al., 2013). This study demonstrated outside of the traditionally studied phosphatase domains (Tamura
that an shRNA-assisted approach to stimulating axon regeneration et al., 1999a, 1999b).
of corticospinal tract axons following spinal cord injury recapitu- In summary, the pro-regenerative role of inhibiting PTEN func-
lated the ndings of an earlier study by the same group (Liu et al., tion is now well established in the visual system through the use of
2010) whereby PTEN gene deletion resulted in axon regeneration experimental animal models, particularly the optic nerve crush
up to 2 mm beyond the lesion site. Geoffroy and colleagues recently injury model in mice. In addition, there is now ample evidence that
revealed an age-dependent decline in axon regeneration induced activating inammation (see below), combined with cAMP treat-
by PTEN deletion in adult mammalian CNS neurons (corticospinal ment (Kurimoto et al., 2010) as well as genetic ablation of SOCS3
and rubrospinal neurons) (Geoffroy et al., 2016). Interestingly, PTEN (Sun et al., 2011) may enhance the regeneration mediated by PTEN
deletion enhanced the intrinsic growth ability by increasing mTOR inhibition. However, there is a conceivable oncogenic risk associ-
activity in both young and aged CNS neurons, where neuronal soma ated with manipulation of PTEN, a risk that could be associated
size and axonal growth proximal to the injury site was similar, with the mTOR pathway itself or with non-mTOR-dependent,
leading the authors to attribute the age-dependent decline in PTEN-related activities. Perhaps oncogenic risk could be mini-
regenerative ability to extrinsic inuences at and around the injury mised by developing an mTOR-specic method to facilitate axon
site (Geoffroy et al., 2016). Whether the same relationship holds regeneration.
true for RGCs remain to be explored.
Although the oncogenic potential of PTEN inhibition has not 2.2. Cytokine signalling and inammatory mechanisms
been explored in regeneration studies, the involvement of PTEN in
several visual system and brain cancers suggests caution should be Cytokines are inammatory messengers that play a critical role
adopted when manipulating PTEN for regenerative purposes. in modulation of a variety of biological functions in the nervous
Indeed, haploinsufciency of PTEN has been identied in many system. Advances in the eld of neuro-immunology have stim-
tumours, including several which affect the visual system. ulated efforts to understand the role of immune modulators in
Approximately 70% of glioblastoma multiforme (GBM) cases show nervous system function and several immunologically relevant
loss of chromosome 10q (Bostrom et al., 1998), although it is signalling cascades have been identied over the past two decades.
possible that other genes on 10q could also be involved in patho- There are several examples of cytokines that promote axon
genesis of GBM. Clinical ndings suggest that alterations in the regeneration but also participate in signalling cascades relevant to
PTEN gene (resulting in PTEN protein deciency) may contribute to cancer.
multiple aspects of glioma pathology and may play a critical role in Inammation, either as a result of lens injury or administration
determining prognosis and therapeutic response in patients of pro-inammatory agents such as zymosan, is known to promote
suffering from this and other forms of cancer. Visual system gli- survival and regeneration of injured RGCs, an effect mediated by
omas, including retinoblastoma, are relatively common and both activated macrophages and retinal astrocytes (Leibinger et al.,
concerted research efforts have gone into identifying relevant 2009). Fischer and colleagues, demonstrated that upon inamma-
pathogenic mechanisms. Interestingly, many of the molecules that tory stimulation, CNTF, LIF, and interleukin-6 (IL-6) are upregulated
have been implicated in axon regeneration processes in the visual and released by retinal astrocytes and have pro-regenerative effects
system have also been suggested to play a role in retinoblastoma. on injured neurones via a signal transducer and activator of tran-
Indeed, PTEN has been previously shown to be involved in the scription 3 (STAT3) dependent mechanism (Leibinger et al., 2013;
control of retinoblastoma protein (Paramio et al., 1999), which is Muller et al., 2007). CNTF, IL-6 and LIF exert their effects by bind-
critical in the development of retinoblastoma cancers; both spo- ing to receptors and forming a complex with glycoprotein 130
radic and inherited forms of retinoblastoma have been linked to (gp130) (Heinrich et al., 2003; Leibinger et al., 2013). This complex
PTEN (Zochodne, 2014). In addition to its proposed function in the in turn activates the Jak/STAT (Janus kinase-signal transducer and
visual system, PTEN has also been linked to brain tumours activator of transcription) pathway via phosphorylation of the
(Endersby and Baker, 2008; Haas-Kogan and Stokoe, 2008). This STAT3 transcription factor which initiates gene expression
supports the critical nature of PTEN function in regulating (Heinrich et al., 2003). The gp130 complex can also trigger activa-
cancerous growth in the CNS. The structural components of PTEN tion of the PI3K/mTOR pathway by phosphorylation of the cancer
are critical to its function in cellular growth (Fig. 1); in addition, the associated protein tyrosine phosphatase, SHP-2 (Charest et al.,
downstream cascades that are involved in regulating PTEN function 2006; Hirano, 1998; Nicholson et al., 2000), supporting a role for
can be PI3K-dependent or independent (Simpson and Parsons, this pathway in cellular growth programs. SOCS-3 is a known Jak/
2001; Weng et al., 2001). To elaborate further on the PI3k inde- STAT signalling repressor and the ability of RGCs to respond to
pendent activity of PTEN it must be considered that the PTEN CNTF, and thus regenerative ability, is greatly enhanced by SOCS3
protein is composed of 5 functional domains. Indeed, there are over deletion (Qin et al., 2013; Smith et al., 2009). Furthermore, RGC
3000 reported mutations of the PTEN gene reported in the COSMIC axon regeneration can be observed in RGCs with constitutively
(catalogue of somatic mutations in cancer) online database (http:// active STAT3 (Pernet et al., 2013). Recent work has demonstrated
cancer.sanger.ac.uk/cosmic) reecting the varied functions of PTEN extensive axon regeneration following transduction of RGCs with
and its diverse role in cancer. PTEN acts as both a lipid phosphatase, an AAV construct expressing constitutively active Stat3CA fused
and is associated with mTOR pathway activation; it also acts as a with viral active domain (VP16), which is known to hyperactivate
protein phosphatase when associated with focal adhesion kinase transcription factors by recruiting transcriptional co-factors to the
(FAK) and plays a role in integrin signalling (Tamura et al., 1998, DNA binding domain (Hirai et al., 2010; Mehta et al., 2016). Inter-
1999b). Importantly, it is known to be involved in facilitating estingly, in zebrash, a species that retains regenerative ability in
tumour cells invasive and metastatic behaviours (Tamura et al., adulthood, Jak/STAT signalling driven in part by CNTF in the pres-
1998, 1999a). Indeed, the functions of PTEN have been expanded ence of SOCS3 (Smith et al., 2009) is important for robust optic
signicantly in recent years to include phosphatase-independent nerve regeneration (Elsaeidi et al., 2014; Smith et al., 2009). In
activities that have been elegantly reviewed by (Song et al., addition, LIF has been shown to be upregulated in zebrash RGCs 3
2012a). In addition, there may be potential unexplored functions days after optic nerve injury and is immediately followed by STAT3
A. Barber et al. / Progress in Retinal and Eye Research 56 (2017) 19e31 23

activation (Ogai et al., 2014). including Bcl-2, p53 and caspase 3, has been implicated in onco-
Inammation is known to play a pivotal role in cancer initiation genesis (Sitorus et al., 2009). While these molecules have been
and malignant progression, and Jak/STAT signalling has been studied in the context of retinal cancers, it is of note that they have
demonstrated to play a key role in the initiation of malignant also been modulated in models of retinal disease and have shown
transformation and cancer progression of lymphopoiesis and promise as potential targets for therapeutic intervention. As an
gastric adenomas (Jenkins et al., 2005; Yin et al., 2015). Indeed, example, Bcl-2 overexpressing mice show improved survival and
enhanced expression of the IL-6 cytokine superfamily, including axon regeneration (Chen et al., 1997). Additionally, a supportive role
CNTF and LIF, has been demonstrated to be upregulated in human for Bcl-2 has been established in the retina (Huang et al., 2003).
brain tumours (Lilja et al., 2001). LIF has been shown to be over- Although it must be noted that the mood stabilizer, lithium, has
expressed in several types of cancers and has been associated with multiple mechanisms of action; in these experiments, lithium was
the progression of several malignancies, including rhabdomyosar- used to induce expression of Bcl-2, which in turn resulted in sig-
coma (cancer in striated muscle tissue), choriocarcinoma (cancer in nicant improvement in RGC survival as well as a regenerative
tissue that normally would become the placenta) and melanoma (Li effect on RGC axons (Huang et al., 2003). While normal retinal
et al., 2014). As an inhibitory regulator of the Jak/STAT pathway, tissue expresses low levels of Bcl-2, it was noted that specic
SOCS3 can be described to have an anti-tumour effect and reduced concentrations of lithium enhanced Bcl-2 expression and improved
activity of SOCS3 has been associated with several cancers. For RGC survival and axon regeneration. Intriguingly, and supporting
example, hypermethylation has been attributed to silencing of the idea that cancer-causing agents could be benecial to the
SOCS3 in human lung cancer (He et al., 2003). In addition, mouse damaged visual system, previous work has demonstrated that
studies involving conditional SOCS3 knockout in either gastroin- lithium treatment enhances the proliferation of potentially regen-
testinal epithelial cells (Inagaki-Ohara et al., 2014) or pancreatic erative stem cells in the retina but is also associated with the
cells (Lesina et al., 2011) demonstrated an increase in tumour for- development of retinoblastoma (Silva et al., 2010). Studies exam-
mation. Although the tumour suppressor role of SOCS3 appears ining the oncogenic risk of Bcl-2 manipulation in RGC axon
clear, several studies indicate a varying role for SOCS3 depending regeneration experiments are lacking.
on the type of cancer. For example, SOCS3 knockdown in prostate Similarly, p53 has been repeatedly shown to be a major
cancer resulted in enhanced apoptotic cell death (Puhr et al., 2009). contributor to cell death processes in the visual system with several
In addition, the number of lung and liver metastatic tumour nod- reports indicating the potential benet of reducing levels of p53 in
ules was reduced in macrophage specic SOCS3 conditional the damaged visual pathway (Park et al., 2008; Wilson et al., 2013).
knockout mice compared to wild-type animals (Hiwatashi et al., The role of p53 in visual system repair remains controversial; our
2011). These studies indicate that the effect of SOCS3 dysregula- work suggests that conditional deletion of p53 promotes RGC sur-
tion can have varying effects depending on the tissue type. Indeed, vival, but is not sufcient to enhance axon regeneration in vivo
the long term effect of SOCS3 dysregulation within the visual sys- (Park et al., 2008). P53 is a critical modulator of a variety of nervous
tem is unknown, highlighting the need for cautious manipulation system functions and has an established role in growth inhibition
of SOCS3 signalling pathways. and DNA repair, where post-translational modication of the pro-
Recent work has revealed the potential benet of manipulating tein is critical to this latter function (reviewed in (Vousden and
granulocyte macrophage colony stimulating factor (GM-CSF) on Prives, 2009)). The pro-apoptotic functions of p53 have also been
neurite growth in vitro (Hanea et al., 2016; Legacy et al., 2013). In actively investigated in both cancerous and non-cancerous tissue
support of the current theme regarding the regenerative impact of types, with many studies supporting the role of p53 in cell death
cancer-related molecules, GM-CSF is a hematopoietic cytokine that mechanisms in the nervous system. The activation of p53 is highly
is involved in regulating cancerous growth in several contexts regulated and relies upon post-translational modication that fa-
(Metcalf, 1985a,b). In particular, GM-CSF has the ability to regulate cilitates its role as a transcription factor, with molecular targets of
the proliferation of myeloid leukemia cells (Metcalf, 1985a,b). Taken diverse functions (Dai and Gu, 2010). While a possible role for
together, the accumulating evidence points to a distinct role for dysregulation of apoptotic molecules in cancer is well established,
cytokine signalling in the regulating both axon regeneration and the potential modulation of these factors as a possible means of
cancer. promoting repair of the damaged visual system deserves further
investigation. However the challenge remains in how to maximize
2.3. Anti-apoptotic factors and their role in cancer biology and regeneration without increasing cancer risk.
implications for optic nerve regeneration Promoting cell death/apoptosis is not always detrimental to the
visual system. As an example, recent work suggests that modu-
Apoptosis is a form of programmed cell death which occurs lating mixed-lineage dual leucine zipper kinase (DLK) initiates a
naturally in the nervous system and throughout the body. This cell death program which appears to be important in promoting
process is involved in regulating the number of cells in the devel- axon regeneration in the damaged visual system (Watkins et al.,
oping nervous system and indeed of all developing tissue, but also 2013). In this regard, DLK was found to initiate a transcriptional
has a role in facilitating the removal of old and/or dying neurons. program which coupled the process of apoptosis to improvements
Therapies that target the destruction of cancerous cells generally in axon regenerative ability in mice. While the mechanisms
trigger some form of cell death, including the induction of involved are not absolutely clear, evidence does point to a distinct
apoptosis related mechanisms to eliminate cancerous cells. The role for DLK in regulating regeneration of RGC axons (Watkins et al.,
dysregulation of apoptotic mechanisms has been suggested as a 2013). A dramatic increase in expression of DLK was seen following
possible avenue to generate malignant cells (Mohammad et al., optic nerve crush injury (Watkins et al., 2013). The transcriptional
2015) where the uncontrolled proliferation caused by abnormal targets of DLK are not well known, but there are many genes that
apoptotic mechanisms results in cancer cell survival. In some in- have been identied as possible response elements for DLK. The
stances, this dysregulation of apoptosis could also contribute to paradoxical induction of a presumed pro-apoptotic molecule, DLK,
worse prognosis following therapeutic intervention. There are with activation of regeneration associated genes emphasizes the
many factors that orchestrate the apoptotic signalling cascade and complexity of the problem that is to be targeted in promoting repair
many potential molecular abnormalities can result from abnormal of the damaged visual system. Nonetheless, again here the theme of
function of these factors. Loss of several anti-apoptotic factors, utilizing molecules implicated in oncogenesis remains. Indeed, DLK
24 A. Barber et al. / Progress in Retinal and Eye Research 56 (2017) 19e31

is considered a tumour suppressor and has been previously linked highlighting that KLF6 is essential for HCC cells to evade cell death
with cancer development (Wu et al., 2010), although it has not yet (Sirach et al., 2007). Studies to examine how KLF6 exerts its pro-
been identied in retinal cancers. The possible pro-apoptotic regenerative effect would be interesting, especially to identify
function of DLK in malignant and non-malignant tissue has been whether axon growth is associated with enhanced p53 expression.
suggested and supports the possible utilization of this molecule as a Expression of KLF7, which has been demonstrated to support axon
possible therapeutic target for cancer. growth, has been associated with a poor prognosis of paediatric
acute lymphoblastic leukemia and is used as a biomarker for
2.4. Kruppel-like family (KLF) of transcription factors therapy resistance and relapse (Schuettpelz et al., 2012). Although
the mechanism behind this phenomenon is not clear, it has been
The Krppel-like family of transcription factors (KLFs) are a set suggested that enforced expression of KLF7 resulted in upregula-
of zinc nger DNA binding proteins that regulate gene expression. tion of pro-survival members of the Bcl2 family, whose expression
In mammals there are 17 genes in the KLF family expressed in has been associated with chemotherapy resistance (Schuettpelz
different tissues performing a variety of cellular functions from et al., 2012). It would be interesting to examine whether the pro-
proliferation to apoptosis, differentiation to migration and plurip- regenerative effects of KLF7 were accompanied by enhanced sur-
otency. Many KLF members are expressed in neurons and recently, vival of neurones and mediated by Bcl2.
it has been shown that specic KLF members can regulate intrinsic The role of KLFs in cancer are varied and although the long term
axon growth ability of RGCs and other types of CNS neurons (Apara effect of KLF manipulation within a regenerative context has not
and Goldberg, 2014; Moore et al., 2009), where different members been explored, the lack of associated KLFs to visual system related
of the same KLF family have been shown to have opposing effects malignancies may suggest the oncogenic risk is low. Instead, the
on axon growth ability. The planarian atworm can regenerate any cancer literature may offer us clues as to how KLF manipulation
missing body parts, and KLF has been shown to be required for may promote axon regeneration.
wound regeneration (Scimone et al., 2014) including photoreceptor
neuron regeneration (Lapan and Reddien, 2012). In mammalian 2.5. Rho/ROCK pathway
development the growth potential of axons diminishes with
maturation, a phenomenon that is associated with increased ROCK (Rho-associated protein kinase) is the downstream sig-
expression of KLF4 and KLF9. Indeed, when these factors are nalling protein of the small GTPase Rho, both of which form the
overexpressed in cell culture, neuronal axon growth potential is Rho/ROCK (Rho/Rho-associated coiled-coil containing protein ki-
inhibited (Moore et al., 2009). Furthermore, studies performed by nase) pathway, an important signal transduction system within the
Moore et al. (2009) and Qin et al. (2013) demonstrated that axon CNS. Most commonly known for regulating the cytoskeleton and
regeneration following optic nerve crush injury can be promoted by playing an essential role in cell migration, growth and development
genetic deletion or knock-down of KLF4 in adult mice, an effect that (Chen and Yao, 2013), the Rho/ROCK pathway is also important in
was mediated by the Jak/STAT3 pathway (Moore et al., 2009; Qin regulating visual system function by synthesizing extracellular
et al., 2013). matrix components, critical for the normal function of the retina
Conversely, KLF6 and KLF7 expression declines during the same (Watanabe, 2010). Furthermore, Rho/ROCK mechanisms have been
developmental time window where axon growth ability declines implicated in maintenance of intraocular pressure, with the
illustrating the heterogeneity in function of the KLF family. development of ophthalmic solution to treat glaucoma based upon
Furthermore, KLF6 and KLF7 expression have been shown to pro- Rho/ROCK pathway inhibition (Kaneko et al., 2016; Wang et al.,
mote axon growth both in culture and in the regenerating optic 2013). The role of the Rho/ROCK pathway in regeneration-
nerve of zebrash (Moore et al., 2009; Veldman et al., 2007). In competent species has not been examined in great detail. Previ-
addition, Blackmore et al. (2012) demonstrated that in vivo acti- ous work has shown that the Rho/ROCK pathway is required for
vation of KLF7 enhanced axon regeneration in the spinal cord interkinetic nuclear migration of proliferating Muller glia that
(Blackmore et al., 2012). drives regeneration of photoreceptors in zebrash, a process that
Over the past few years evidence has emerged that KLF dysre- does not translate to mammals (Lahne et al., 2015). The most
gulation plays a role in human cancers, reviewed in detail by informative study is offered by Beane and colleagues, who
Limame et al. (2014). Of the KLF members discussed above, KLF4, demonstrated a role for ROCK in planarian atworm regeneration
whose inhibition promoted axon growth, has been described both (Beane et al., 2012), where inhibition of ROCK by RNAi led to
as a tumour suppressor and oncogene. However, there is a clear role excessive growth of optic neurons well after control animals had
for KLF4 in cancer related epithelial-mesenchymal transition nished their regeneration process. This suggests a role for the Rho/
(EMT), a process that involves a shift in cellular polarity enabling ROCK pathway as a tissue growth limiting mechanism to reduce
cancer cells to invade the surrounding stroma. KLF4 has been production and replacement of cells and prevent uncontrolled
documented to be a potent inducer of epithelial differentiation in growth. Although the long term effects of Rho/ROCK inhibition in
EMT (Limame et al., 2014). KLF9, whose down regulation promotes mammalian regeneration has not been examined, the Beane study
axon regeneration, surprisingly has been shown to positively may indicate that long term inhibition is undesirable (Beane et al.,
regulate p53 expression and has been identied as a tumour sup- 2012).
pressor in glioblastomas, colorectal tumours and hepatocellular The potential role of the Rho/ROCK pathway in CNS regenera-
carcinoma (Sun et al., 2014). KLF6, whose expression results in tion is reviewed extensively previously (Liu et al., 2015; Wang
enhanced axon growth, has been described as a tumour suppressor et al., 2013). The adult CNS offers a hostile environment to
in prostate (Narla et al., 2001), hepatic (Zhenzhen et al., 2012) and regenerating axons, through the expression of myelin-derived
gastric cancer (Zhang et al., 2010) among others. Three splice var- inhibitory molecules (Nogo, OMgp, MAG), chondroitin sulphate
iants of the KLF6 gene have been documented which antagonise the proteoglycans that form components of the glia scar (Fawcett,
tumour suppressor function of wild type KLF6 (Narla et al., 2005). 2006; Smith et al., 2015) and inhibitory guidance cues such as
However, the role of KLF6 in cancer is controversial. KLF6 has been semaphorins and ephrins (Benson et al., 2005). These extracellular
shown to be inactive in many solid tumours yet in vitro silencing of factors exerts their regeneration inhibitory effects through acti-
KLF6 in hepatocellular carcinoma cells (HCC) induced apoptosis by vation of their specic surface receptors on axons (eg Nogo re-
upregulating p53 and inhibiting pro-apoptotic Bcl-xL expression, ceptor, paired immunoglobulin-like receptor, p75 neurotrophin
A. Barber et al. / Progress in Retinal and Eye Research 56 (2017) 19e31 25

receptor, Lingo-1) but their signalling converges by activating the et al., 2012) also boosted the regenerative effects. Y-27632 alone
Rho/ROCK pathway which modulates the actin cytoskeleton and has also been shown to increase myelination in an in vitro injury
results in growth cone collapse and inhibition of neurite out- spinal cord model (Boomkamp et al., 2012), a phenomenon that
growths. Indeed, RhoA, ROCK1 and ROCK2 have been shown to be has been shown to be important in optic nerve regeneration (Bei
expressed at the optic nerve head where they may act to inhibit et al., 2016), however, whether Y-27632 can promote re-
axon regeneration (Goldhagen et al., 2012). Inhibition of the Rho/ myelination of the optic nerve, or in in vivo models, remains to
ROCK signalling pathway has thus received much attention in be addressed.
efforts to promote regeneration of injured axons. Previous The Rho/ROCK signalling pathway is very well established in
research has demonstrated that through targeting of the small oncogenesis (Pruitt and Der, 2001) and the ways in which it con-
GTPase Rho, regeneration of RGCs can be enhanced in vitro tributes to the oncogenic process are numerous and only briey
(Monnier et al., 2003) and in adult rodents following optic nerve reviewed here (and illustrated in Fig. 2). For a more detailed ac-
lesion by overcoming myelin inhibition (Bertrand et al., 2005; count, comprehensive reviews can be found from (Chin et al., 2015;
Fischer et al., 2004). The most commonly known inhibitor of the Rath and Olson, 2012). As mentioned above, the Rho/ROCK
Rho/ROCK pathway is Y-27632 which acts by blocking the ATP- pathway is central to regulation of the actinomycin cytoskeleton
binding site on the catalytic domain of ROCK. (Zohrabian et al., and thus to cell motility, a function that is essential for cancer cell
2009). Lingor and colleagues demonstrated that Y-27632, along migration and invasion. Indeed aberrant activation of this pathway
with other ROCK inhibitors Fasudil (HA-1077) and Dimethylfasudil is well established in the literature as a potential precursor to
(H-1152), promoted neurite outgrowth of RGCs cultured on migration and invasion of tumour cells in vitro and in vivo (Itoh
inhibitory chondroitin sulphate proteoglycan substrate (Lingor et al., 1999; Li et al., 2006). In addition, the Rho/ROCK pathway is
et al., 2007). However, it is worth mentioning that these widely critical for tumour-associated angiogenesis, which is a key char-
used ROCK inhibitors can have multiple targets and can inhibit a acteristic of tumour progression and is stimulated by the intra-
number of other kinases at high doses (Anastassiadis et al., 2011). tumour hypoxic environment and largely controlled by vascular
Y-27632 also stimulated RGC axon regeneration in the rat optic endothelial growth factor (VEGF)-mediated Rho/ROCK activation
nerve crush in vivo model in a dose dependent manner (Lingor (Chin et al., 2015). Rho/ROCK signalling also plays a role in loos-
et al., 2007) and in adult cats (Sagawa et al., 2007). Interestingly ening the endothelial intercellular junctions, a process required for
combined ROCK inhibition with increased CNTF results in additive the regulation and development of new endothelial capillaries (Liu
effects on neurite outgrowth extension in vitro (Ahmed et al., and Senger, 2004).
2009) and on RGC axon regeneration following optic nerve crush Many studies have demonstrated the therapeutic value of
in adults rats (Lingor et al., 2008). Similarly co-administration of Y- inhibiting the Rho/ROCK pathway in cancer models, including
27632 with cAMP (Ahmed et al., 2009) and erythropoietin (Tan reduced cellular proliferation, invasion and angiogenesis in vitro

Fig. 2. The Rho/ROCK pathway is a critical regulatory cascade that is primarily involved in regulating cytoskeletal structure and maintenance, while playing a critical role during
early development. Importantly, the Rho/ROCK pathway has also been found to be a key regulator of PTEN signalling mechanisms. As such, this pathway is often studied in the
context of both regeneration and oncogenesis. Y-27632 has several kinase targets; it is an effective inhibitor of ROCK signalling.
26 A. Barber et al. / Progress in Retinal and Eye Research 56 (2017) 19e31

and reduced tumour growth and metastasis in vivo which are unlocking the role of oncogenes and tumour suppressors could be
reviewed extensively by Chin et al. (2015) and too numerous to effective in facilitating axon growth in the adult CNS, including the
discuss in detail in this review (Chin et al., 2015). visual system.
In summary, considering the varied role of the Rho/ROCK
pathway in cell migration and adhesion it is of no surprise that it 2.7. Epidermal growth factor receptor (EGFR)
has an important role in both axon regeneration and oncogenesis.
What we can learn from the cancer literature is that there are The EGFR gene mutations are frequently seen in lung cancers,
numerous mechanisms by which the Rho/ROCK pathway can particularly in adenocarcinomas (Midha et al., 2015). EGFR is well
contribute to the oncogenic process, unsurprising considering the established to be involved in the pathogenesis of cancer, particu-
plethora of affectors and effectors of this pathway. The specic larly in the lung, although there is currently little evidence sup-
details of how the Rho/ROCK operates in oncogenesis are more porting a role for EGFR in cancers of the visual system. However,
thoroughly understood in comparison to the axon regeneration EGFR has been suggested to play a critical role in neuronal function
literature. It would be interesting to have a clearer picture of which throughout the CNS. EGFR is expressed in the retina, where it
upstream affectors and downstream effectors of the Rho/ROCK regulates neuronal as well as glial precursor proliferation and dif-
pathway account for the regeneration effects. Rho/ROCK dysregu- ferentiation (Chen et al., 2007). The function of EGFR in mature
lation promotes later stage cancer processes and whether neurons is not well dened, but there is evidence suggesting a
manipulation of this pathway is enough to cause cancer is unclear. possible role in regulation of RGC survival and axon regeneration
The oncogenic risk of Rho/ROCK pathway inhibition as a method to (Berry et al., 2011). EGFR transactivation has been suggested to play
stimulate axon regeneration may therefore be minimal, but when a key role in inhibition of CNS axon growth. In this regard, small
used in combinatorial therapies such as PTEN gene deletion etc, the molecule inhibitors of EGFR promote neurite growth on inhibitory
oncogenic risk could conceivably be considerably elevated. Indeed substrates in vitro and axon regeneration after optic nerve injury
little is known about the role of Rho/ROCK in regeneration in vivo (Koprivica et al., 2005). However, the role of EGFR in the
competent species, although there is evidence that points to the visual system remains unclear (Berry et al., 2011; Koprivica et al.,
Rho/ROCK pathway acting as a regenerating tissue growth brake 2005). A proposed role for EGFR in regulating RGC viability in a
that may minimize oncogenic risk. model of retinopathy has been suggested by Hewing et al. (2013),
where the authors showed that pharmacological EGFR inhibition
2.6. Haematopoietically Expressed Homeobox (HHEX) protects RGCs from oxygen-induced retinopathy in mice (Hewing
et al., 2013). However, it must be considered that inhibitors of
Recently, Blackmore and colleagues tested a large number of EGFR can have dose dependent off-targets effects, including ERBB4
transcription factors that have previously been implicated in and other kinases, as such these effects may be a combined result of
regulating tumour suppressive functions in neuronal cells and multiple receptor systems and not solely EGFR. Although the evi-
evaluated the impact of modulating these factors on neurite growth dence remains controversial, the theme holds true for EGFR which
(Simpson et al., 2015). A functional screen identied several genes represents another oncogenic molecule that could be targeted to
that were considered tumour suppressor or oncogenic in regulation promote repair of the damaged visual system.
of neurite growth. Of the molecules identied, the authors carried
out follow-up experiments to determine the impact of modulating 3. Less explored potential targets
the CNS expressing tumour suppressor, HHEX (Su et al., 2012), on
axon growth ability. This work demonstrated that overexpression Manipulating cancer-associated genes to promote repair and
of HHEX reduced axon growth ability, supporting a role for HHEX in recovery in the damaged CNS may appear counter-intuitive, but it is
limiting axon growth in the CNS (Simpson et al., 2015). These data worth noting that nature uses these growth mechanisms for pre-
again support the theme and hypothesis that repurposing and cisely this purpose in other organisms while avoiding oncogenesis.

Fig. 3. Throughout this review we have discussed various genes, receptors, ligands, and messenger systems, and how they contribute to either regenerative and/or oncogenic
processes as individual contributing factors. Importantly, the majority of the factors discussed act on complimentary systems and can be combined into a comprehensive pathway
with signicant crosstalk among signalling factors. When taken together one can easily see how these factors interact with each other and all to cell survival, regeneration as well as
cellular proliferation.
A. Barber et al. / Progress in Retinal and Eye Research 56 (2017) 19e31 27

Indeed some non-mammalian species, namely zebrash, xenopus both by serotype tropism and by the use of a cell specic promoter.
and other anamniotes, retain the capacity to regenerate CNS axons However non-viral approaches such as siRNA and small molecule
well into the adult years (Turner and Delaney, 1979). Several factors, or peptide compounds could also be of particular importance
discussed in this section, have been identied to contribute to the where the temporal effect is limited by their degradation rate.
regenerative process in these models that have yet to be explored in Small molecule inhibitors of tumour suppressor genes or activators
mammalian regeneration. We will also discuss other unexplored of oncogenes could be feasible therapeutically as they can be
targets generated by mammalian research. delivered locally to the eye, where the retinal blood barrier may
restrict spatial delivery avoiding unwanted systemic effects.
3.1. Heterogeneous nuclear ribonucleoprotein (hnRNP) As mentioned previously, in other regeneration competent
model systems cancer-associated genes have been identied to
Both growth associated protein 43 (GAP-43) and medium neu- promote repair and recovery in the damaged CNS while avoiding
rolament (NF-M) are needed to establish the growth cone and oncogenic risk. Studies examining how cancer-associated genes
facilitate axon elongation and thus are important for axon regen- and pathways are used in regeneration competent model systems
eration (Jacobson et al., 1986). Expression of both is tightly may inform us of how to manipulate these genes/pathways safely
controlled by transcriptional and post-transcriptional methods. in the mammalian system. For example PTEN/mTOR activity has
RNA-binding proteins regulate the translation of specic RNAs. been associated with many cancers and has been demonstrated to
RNA-binding protein hnRNP K directly binds both GAP-43 and NF- facilitate neuronal axon regeneration in rodents (Park et al., 2008;
M RNAs exporting them from the nucleus for translation and has Smith et al., 2009; Sun et al., 2011). mTOR activity has also been
been shown to be essential in Xenopus optic axon regeneration (Liu shown to be important for regenerating tissues in the zebrash,
et al., 2012). The role of hnRNP K in mammalian axon regeneration specically in DRG neurons (Abe et al., 2010), blastemal cells
has yet to be explored but caution must be adopted as it this RNA following n amputation (Hirose et al., 2014), and RGCs post optic
binding protein has been described as both a tumour suppressor nerve injury (Diekmann et al., 2015). Interestingly, all tissues
and oncogene (Gallardo et al., 2016). exhibit a transient burst of mTOR activity followed by rapid down
regulation. Transient mTOR activity can be observed in zebrash
3.2. Rb/E2F family RGCs up to 4 days post optic nerve injury correlating with RGC
axonal growth, an effect that was compromised by mTOR inhibition
The retinoblastoma protein (Rb1) is involved in regulating cell by rapamycin (Diekmann et al., 2015). It has been suggested that
cycle progression and suppresses E2F transcriptional activity to activation of the mTOR pathway post axotomy in zebrash is tightly
promote its cellular functions. Rb phosphorylation regulates and regulated by expression of LIF (Diekmann et al., 2015; Ogai et al.,
inhibits its binding to E2F. Rb1 is well characterized as a gene that is 2014) and by its autoregulatory negative feedback mechanism
frequently deleted in retinoblastoma, which is seen in young chil- where activated mTOR itself strongly represses the PI3K/AKT
dren (Friend et al., 1986). The mechanisms utilized by Rb are not pathway (Harrington et al., 2005). LIF also shows a similar time
clearly understood, and currently there is no clear evidence sup- course of expression as mTOR activity in the zebrash axotomy
porting a role for RB in optic nerve regeneration, in contrast to model, with an early transient peak of expression followed by a
PTEN, where PTEN decient neurons are effective at mounting a quick down regulation (Ogai et al., 2014). Such tight spatial regu-
regenerative response in the visual system (Park et al., 2008; Smith lation of both mTOR activity and LIF expression may help to subvert
et al., 2009; Sun et al., 2011). Interestingly, co-deletion of both PTEN uncontrolled cellular growth and thus oncogenic risk.
and Rb results in the development of retinoblastoma (Xie et al., In addition, Jak/STAT signalling regulated in part by CNTF drives
2015). Furthermore, the lysine acetyltransferase p300, which is optic nerve regeneration in the zebrash even in the presence of
also linked to retinoblastoma has also been implicated in enhancing SOC3 (Elsaeidi et al., 2014). Interestingly, SOCS3a and splicing factor
regenerative axonal growth (Gaub et al., 2011). Overexpression of proline- and glutamine-rich (Sfpq) proteins known to inhibit the
p300, which is normally involved in development of RGCs, plays a regenerative response, were upregulated following optic nerve
key role in promoting axon regeneration following optic nerve lesion and regulated by Jak/STAT signalling (Elsaeidi et al., 2014).
damage (Gaub et al., 2011). Together, these data support a possible Knockdown of these proteins enhanced optic nerve regeneration,
role for retinoblastoma genes in regulating axon regeneration as can also be observed in the mammalian system. Most intrigu-
mechanisms in the adult visual system, but further investigation is ingly, zebrash were able to overcome the inhibitory effects of
necessary to clearly dene this potential role. SOCS3a and Sfpq but inducing IL-6 cytokines expression, including
CNTF, IL-11 and Clcf1/Crlf1a, in RGCs to stimulate Jak/Stat signalling
4. Approaches to reducing cancer risk when promoting leading to robust axon regeneration. This suggests that expression
regeneration of SOCS3 may act as an inhibitory mechanism to prevent uncon-
trolled and aberrant axonal growth. These studies indicate that it is
Neuronal regenerative therapies that target cancer eassociated possible to overcome Jak/STAT signalling suppression by SOCS3
molecular pathways raise a serious concern regarding oncogenesis, without genetic deletion and that perhaps a combinatorial
although conceivably this risk may be limited by the post-mitotic approach using several cytokines may be of use to drive Jak/STAT
nature of the mature visual system. Genes may function differ- mediated axon regeneration. An alternative approach is to adopt
ently in a post-mitotic cellular context, thereby allowing the the use of designer growth factors to induce a stronger and more
modulation of tumour suppressors without enacting oncogenic specic regeneration effect than natural growth factors. A good
mechanisms. Thus it is plausible that modulation of tumour sup- example of such an approach was recently reported by Leibinger
pressors, if targeted in a spatially and temporally directed manner, et al. (2016), whereby a designer cytokine hyper-IL-6 (hIL-6) was
could be benecial to repair and recovery of the CNS, including the delivered virally to directly activate the gp130 receptor (Leibinger
visual system. There are multiple options available that could et al., 2016). The authors demonstrated stronger activation of JAK/
facilitate the modulation of cancer molecules to promote axon STAT3 and PI3K/Akt/mTOR signalling pathways, which was
regeneration in a clinically safer manner. For example viral- accompanied by increased neurite growth of cultured RGCs and
mediated options have been consistently suggested, where the vi- DRGs and enhanced axon regeneration in the optic nerve crush
rus is deemed replication decient and expression can be limited mouse model compared to CNTF and IL-6. This study highlights the
28 A. Barber et al. / Progress in Retinal and Eye Research 56 (2017) 19e31

Table 1
The role of various factors in retinal regeneration and/or carcinogenesis. The table summarises selected sources referenced for each factor proposed to be involved in regulating
both axon regeneration and cancer mechanisms.

Factor Retinal regeneration sources Cancer sources

Bcl-2 Chen et al., 1997; Huang et al., 2003 Schuettpelz et al., 2012; Sitorus et al., 2009
DLK Watkins et al., 2013; Wu et al., 2010
EGFR Berry et al., 2011; Chen et al., 2007; Hewing et al., 2013; Koprivica et al., Midha et al., 2015
2005
GM-CSF Hanea et al., 2016; Legacy et al., 2013 Metcalf, 1985a,b
HHEX Simpson et al., 2015 Su et al., 2012
hnRNP K Liu et al., 2012 Gallardo et al., 2016
JAK/STAT Elsaeidi et al., 2014; Moore et al., 2009; Smith et al., 2009; Qin et al., 2013 Jenkins et al., 2005; Yin et al., 2015
KLF Apara and Goldberg, 2014; Lapan and Reddien, 2012; Moore et al., 2009; Limame et al., 2014
Scimone et al., 2014
KLF4 Moore et al., 2009; Qin et al., 2013 Limame et al., 2014
KLF6 Moore et al., 2009; Veldman et al., 2007 Narla et al., 2001, 2005; Zhang et al., 2010; Zhenzhen et al., 2012
KLF7 Blackmore et al., 2012; Moore et al., 2009; Veldman et al., 2007 Schuettpelz et al., 2012;
KLF9 Moore et al., 2009 Sun et al., 2014
LIF Diekmann et al., 2015; Leibinger et al., 2009; Muller et al., 2007; Ogai et al., Li et al., 2014
2014
mTOR Abe et al., 2010; Charest et al., 2006; Diekmann et al., 2015; Hirano, 1998; Park et al., 2008
Hirose et al., 2014; Nicholson et al., 2000; Ogai et al., 2014; Park et al., 2008
MYC Belin et al., 2015 Weiss, 1982
p53 Dai and Gu, 2010; Park et al., 2008; Vousden and Prives, 2009; Wilson et al., Sirach et al., 2007; Sitorus et al., 2009; Sun et al., 2014
2013
PTEN Abe et al., 2010; Byrne et al., 2014; Christie et al., 2010; de Lima et al., 2012; Bostrom et al., 1998; Cordero-Espinoza and Hagen, 2013; Endersby and
Duan et al., 2015; Geoffroy et al., 2016; Hirose et al., 2014; Liu et al., 2010; Baker, 2008; Haas-Kogan and Stokoe, 2008; Icard and Lincet 2013; Li et al.,
Park et al., 2008; Smith et al., 2009; Song et al., 2012a,b; Sun et al., 2011; 1997; Paramio et al., 1999; Weng et al., 1999; Xie et al., 2015; Zochodne,
Zukor et al., 2013 2014
Rb1 Park et al., 2008; Smith et al., 2009; Sun et al., 2011 Friend et al., 1986; Xie et al., 2015
Rho Bertrand et al., 2005; Chen and Yao, 2013; Goldhagen et al., 2012; Kaneko Chin et al., 2015; Itoh et al., 1999; Li et al., 2006; Liu and Senger, 2004; Pruitt
et al., 2016; Lahne et al., 2015; Monnier et al., 2003; Wang et al., 2013; and Der, 2001; Rath and Olson, 2012
Watanabe, 2010
ROCK Ahmed et al., 2009; Beane et al., 2012; Bertrand et al., 2005; Boomkamp Chin et al., 2015; Itoh et al., 1999; Li et al., 2006; Liu and Senger, 2004; Pruitt
et al., 2012; Bei et al., 2016; Chen and Yao, 2013; Kaneko et al., 2016; Lahne and Der, 2001; Rath and Olson, 2012
et al., 2015; Lingor et al., 2008; Liu et al., 2015; Monnier et al., 2003; Tan
et al., 2012; Wang et al., 2013; Watanabe, 2010; Zohrabian et al., 2009
SOCS-3 Elsaeidi et al., 2014; Smith et al., 2009; Sun et al., 2011; Qin et al., 2013 He et al., 2003; Hiwatashi et al., 2011; Inagaki-Ohara et al., 2014; Lesina
et al., 2011; Puhr et al., 2009

importance of understanding the expression proles of growth appropriate synaptic targets to allow functional circuits to be re-
factor receptors. Specically, both CNTF and IL-6 must rst bind to established. However manipulating factors which promote axonal
their non-signalling a-receptors, which in turn acts to recruit growth may not be sufcient to restore functions such as useful
further signalling receptor subunits: gp130/LIFR in the case of CNTF vision. In addition, the therapeutic time window for axonal
and a gp130 homodimer in the case of IL-6 (Heinrich et al., 2003). regeneration will need to be established and the extent of target
Low expression of the a-receptor subunits rather than gp130 itself CNS remodelling post injury should be considered when evaluating
results in a limited response to CNTF or IL-6 (Heinrich et al., 2003), re-connectivity of visual input. We have previously shown that
indeed in the case of optic nerve crush injury the CNTF a-receptor manipulation of some cancer-related genes, such as PTEN and
subunits is actually downregulated (Miotke et al., 2007). In this SOCS3, promotes both neuron survival and axon regeneration
instance, direct activation of the gp130 receptor subunit would be in vivo (Park et al., 2008; Smith et al., 2009). Others have also shown
favourable and the designer hIL-6, that consists of the covalently enhanced axonal regeneration by manipulating other cancer-
linked bioactive parts of IL-6 and soluble IL-6a, does exactly this related mechanisms, either with (Belin et al., 2015) or without
(Peters et al., 1998). The development of designer growth factors (Moore et al., 2009) enhanced cell survival. Our previous work also
may offer a method to specically and more effectively activate found evidence of dissociation between cell survival mechanisms
specic elements of the relevant signalling pathways involved in and axon regeneration mechanisms in the visual system in certain
axon regeneration. contexts. We have shown that p53, a well-known tumour sup-
As an alternative strategy, it is conceivable that toxin or drug pressor, when conditionally knocked out in RGCs, promotes neuron
sensitivity could be engineered into cells as part of a regenerative survival but has no effect on axon regeneration (Park et al., 2008).
therapy to enable cells to be selectively eliminated should they Together, these results support the claim that combinatorial ap-
escape cell cycle control. Conditional gene expression systems, proaches to protection and regeneration may be necessary to
where genes can be turned on or off using drugs such as tamoxifen facilitate recovery following CNS injury, especially given the po-
or tetracycline, have been widely used in neuroscience research and tential molecular crosstalk among the different factors (Fig. 3).
could potentially form part of a risk reduction strategy for neural Indeed, some of the cancer regulatory molecules that have been
regeneration treatments (Schonig et al., 2012). discussed throughout this review, show dramatic impact on not
only axon regeneration, but in most cases cell survival as well. The
5. Beyond axonal growth: challenges for repair possibility to harness the modulation of oncogenes and tumour
suppressor molecules toward promoting both cell survival and
In order to promote repair and recovery following CNS damage, axon regeneration in the post-mitotic neurons of the retina is of
neurons must survive and then extend regenerating axons, which particular interest and value as we strive to identify molecular
must then traverse the hostile inhibitory CNS environment and nd avenues to promote repair of the damaged visual system.
A. Barber et al. / Progress in Retinal and Eye Research 56 (2017) 19e31 29

6. Conclusion and future directions 81, 561e573.


Charest, A., Wilker, E.W., McLaughlin, M.E., Lane, K., Gowda, R., Coven, S.,
McMahon, K., Kovach, S., Feng, Y., Yaffe, M.B., Jacks, T., Housman, D., 2006. ROS
It is clear that where most extensive axon regeneration has been fusion tyrosine kinase activates a SH2 domain-containing phosphatase-2/
observed in the adult CNS to date it has resulted from manipulation phosphatidylinositol 3-kinase/mammalian target of rapamycin signaling axis to
or indirect regulation of cancer associated pathways (see Table 1 for form glioblastoma in mice. Cancer Res. 66, 7473e7481.
Chen, D.F., Schneider, G.E., Martinou, J.C., Tonegawa, S., 1997. Bcl-2 promotes
summary of selected factors discussed here). Many of these studies regeneration of severed axons in mammalian CNS. Nature 385, 434e439.
have exclusively focused on the regenerative potential of manipu- Chen, H., Liu, B., Neufeld, A.H., 2007. Epidermal growth factor receptor in adult
lated cancer associated genes but it is clear that the long term retinal neurons of rat, mouse, and human. J. Comp. Neurol. 500, 299e310.
Chen, Z.Y., Yao, W.J., 2013. Role of Rho/ROCK in the migration of vascular smooth
oncogenic risk will need to be assessed if such therapies can be muscle cells. Sheng li ke xue jin zhan [Prog. Physiol.] 44, 269e274.
considered as a realistic option for patients. Several considerations Chin, V.T., Nagrial, A.M., Chou, A., Biankin, A.V., Gill, A.J., Timpson, P., Pajic, M., 2015.
must be taken into account to effectively implement this type of Rho-associated kinase signalling and the cancer microenvironment: novel
biological implications and therapeutic opportunities. Expert Rev. Mol. Med. 17,
therapeutic approach. Certainly cancer gene manipulation will e17.
require consideration of both spatial and temporal constraints to Christie, K.J., Webber, C.A., Martinez, J.A., Singh, B., Zochodne, D.W., 2010. PTEN
reduce the possibility of oncogenesis in the adult CNS. inhibition to facilitate intrinsic regenerative outgrowth of adult peripheral
axons. J. Neurosci. Off. J. Soc. Neurosci. 30, 9306e9315.
Even in the event that there is no therapeutically appropriate Cordero-Espinoza, L., Hagen, T., 2013. Increased concentrations of fructose 2,6-
avenue to implement cancer gene therapy as a mechanism to bisphosphate contribute to the Warburg effect in phosphatase and tensin ho-
promote regeneration, there is tremendous value in the under- molog (PTEN)-decient cells. J. Biol. Chem. 288, 36020e36028.
Dai, C., Gu, W., 2010. p53 post-translational modication: deregulated in tumori-
standing gained from studying the role of these molecules in the
genesis. Trends Mol. Med. 16, 528e536.
CNS. The knowledge gained could provide important insights into de Lima, S., Koriyama, Y., Kurimoto, T., Oliveira, J.T., Yin, Y., Li, Y., Gilbert, H.Y.,
the cellular and molecular mechanisms required to promote axon Fagiolini, M., Martinez, A.M., Benowitz, L., 2012. Full-length axon regeneration
regeneration, thereby driving additional research into which mo- in the adult mouse optic nerve and partial recovery of simple visual behaviors.
Proc. Natl. Acad. Sci. U. S. A. 109, 9149e9154.
lecular targets are critical to promoting regeneration in the adult De Potter, P., Disneur, D., Levecq, L., Snyers, B., 2002. Ocular manifestations of
CNS. cancer. J. Fr. d'ophtalmologie 25, 194e202.
Diekmann, H., Kalbhen, P., Fischer, D., 2015. Active mechanistic target of rapamycin
plays an ancillary rather than essential role in zebrash CNS axon regeneration.
References Front. Cell. Neurosci. 9, 251.
Duan, X., Qiao, M., Bei, F., Kim, I.J., He, Z., Sanes, J.R., 2015. Subtype-specic
Abe, N., Borson, S.H., Gambello, M.J., Wang, F., Cavalli, V., 2010. Mammalian target of regeneration of retinal ganglion cells following axotomy: effects of osteopontin
rapamycin (mTOR) activation increases axonal growth capacity of injured pe- and mTOR signaling. Neuron 85, 1244e1256.
ripheral nerves. J. Biol. Chem. 285, 28034e28043. Elsaeidi, F., Bemben, M.A., Zhao, X.F., Goldman, D., 2014. Jak/Stat signaling stimu-
Ahmed, Z., Berry, M., Logan, A., 2009. ROCK inhibition promotes adult retinal lates zebrash optic nerve regeneration and overcomes the inhibitory actions of
ganglion cell neurite outgrowth only in the presence of growth promoting Socs3 and Sfpq. J. Neurosci. Off. J. Soc. Neurosci. 34, 2632e2644.
factors. Mol. Cell. Neurosci. 42, 128e133. Endersby, R., Baker, S.J., 2008. PTEN signaling in brain: neuropathology and
Anastassiadis, T., Deacon, S.W., Devarajan, K., Ma, H., Peterson, J.R., 2011. Compre- tumorigenesis. Oncogene 27, 5416e5430.
hensive assay of kinase catalytic activity reveals features of kinase inhibitor Fawcett, J.W., 2006. The glial response to injury and its role in the inhibition of CNS
selectivity. Nat. Biotechnol. 29, 1039e1045. repair. Adv. Exp. Med. Biol. 557, 11e24.
Apara, A., Goldberg, J.L., 2014. Molecular mechanisms of the suppression of axon Fischer, D., Petkova, V., Thanos, S., Benowitz, L.I., 2004. Switching mature retinal
regeneration by KLF transcription factors. Neural Regen. Res. 9, 1418e1421. ganglion cells to a robust growth state in vivo: gene expression and synergy
Beane, W.S., Tseng, A.S., Morokuma, J., Lemire, J.M., Levin, M., 2012. Inhibition of with RhoA inactivation. J. Neurosci. Off. J. Soc. Neurosci. 24, 8726e8740.
planar cell polarity extends neural growth during regeneration, homeostasis, Friend, S.H., Bernards, R., Rogelj, S., Weinberg, R.A., Rapaport, J.M., Albert, D.M.,
and development. Stem Cells Dev. 21, 2085e2094. Dryja, T.P., 1986. A human DNA segment with properties of the gene that pre-
Bei, Y., Hua-Huy, T., Nicco, C., Duong-Quy, S., Le-Dong, N.N., Tiev, K.P., Chereau, C., disposes to retinoblastoma and osteosarcoma. Nature 323, 643e646.
Batteux, F., Dinh-Xuan, A.T., 2016. RhoA/Rho-kinase activation promotes lung Gallardo, M., Hornbaker, M.J., Zhang, X., Hu, P., Bueso-Ramos, C., Post, S.M., 2016.
brosis in an animal model of systemic sclerosis. Exp. Lung Res. 42, 44e55. Aberrant hnRNP K expression: all roads lead to cancer. Cell Cycle 15, 1552e1557.
Belin, S., Nawabi, H., Wang, C., Tang, S., Latremoliere, A., Warren, P., Schorle, H., Gao, X., Neufeld, T.P., Pan, D., 2000. Drosophila PTEN regulates cell growth and
Uncu, C., Woolf, C.J., He, Z., Steen, J.A., 2015. Injury-induced decline of intrinsic proliferation through PI3K-dependent and -independent pathways. Dev. Biol.
regenerative ability revealed by quantitative proteomics. Neuron 86, 221, 404e418.
1000e1014. Gaub, P., Joshi, Y., Wuttke, A., Naumann, U., Schnichels, S., Heiduschka, P., Di
Benowitz, L.I., He, Z., Goldberg, J.L., 2015. Reaching the brain: advances in optic Giovanni, S., 2011. The histone acetyltransferase p300 promotes intrinsic axonal
nerve regeneration. Exp. Neurol. 30141e30142 pii: S0014-4886. regeneration. Brain J. Neurol. 134, 2134e2148.
Benson, M.D., Romero, M.I., Lush, M.E., Lu, Q.R., Henkemeyer, M., Parada, L.F., 2005. Geoffroy, C.G., Hilton, B.J., Tetzlaff, W., Zheng, B., 2016. Evidence for an age-
Ephrin-B3 is a myelin-based inhibitor of neurite outgrowth. Proc. Natl. Acad. dependent decline in axon regeneration in the adult mammalian central ner-
Sci. U. S. A. 102, 10694e10699. vous system. Cell Rep. 15, 238e246.
Berry, M., Ahmed, Z., Douglas, M.R., Logan, A., 2011. Epidermal growth factor re- Goldhagen, B., Proia, A.D., Epstein, D.L., Rao, P.V., 2012. Elevated levels of RhoA in
ceptor antagonists and CNS axon regeneration: mechanisms and controversies. the optic nerve head of human eyes with glaucoma. J. Glaucoma 21, 530e538.
Brain Res. Bull. 84, 289e299. Haas-Kogan, D., Stokoe, D., 2008. PTEN in brain tumors. Expert Rev. Neurother. 8,
Bertrand, J., Winton, M.J., Rodriguez-Hernandez, N., Campenot, R.B., McKerracher, L., 599e610.
2005. Application of Rho antagonist to neuronal cell bodies promotes neurite Hanea, S.T., Shanmugalingam, U., Fournier, A.E., Smith, P.D., 2016. Preparation of
growth in compartmented cultures and regeneration of retinal ganglion cell embryonic retinal explants to study CNS neurite growth. Exp. Eye Res. 146,
axons in the optic nerve of adult rats. J. Neurosci. Off. J. Soc. Neurosci. 25, 304e312.
1113e1121. Harrington, L.S., Findlay, G.M., Lamb, R.F., 2005. Restraining PI3K: mTOR signalling
Blackmore, M.G., Wang, Z., Lerch, J.K., Motti, D., Zhang, Y.P., Shields, C.B., Lee, J.K., goes back to the membrane. Trends Biochem. Sci. 30, 35e42.
Goldberg, J.L., Lemmon, V.P., Bixby, J.L., 2012. Kruppel-like Factor 7 engineered He, B., You, L., Uematsu, K., Zang, K., Xu, Z., Lee, A.Y., Costello, J.F., McCormick, F.,
for transcriptional activation promotes axon regeneration in the adult cortico- Jablons, D.M., 2003. SOCS-3 is frequently silenced by hypermethylation and
spinal tract. Proc. Natl. Acad. Sci. U. S. A. 109, 7517e7522. suppresses cell growth in human lung cancer. Proc. Natl. Acad. Sci. U. S. A. 100,
Boomkamp, S.D., Riehle, M.O., Wood, J., Olson, M.F., Barnett, S.C., 2012. The devel- 14133e14138.
opment of a rat in vitro model of spinal cord injury demonstrating the additive He, Z., Jin, Y., 2016. Intrinsic control of axon regeneration. Neuron 90, 437e451.
effects of Rho and ROCK inhibitors on neurite outgrowth and myelination. Glia Heinrich, P.C., Behrmann, I., Haan, S., Hermanns, H.M., Muller-Newen, G., Schaper, F.,
60, 441e456. 2003. Principles of interleukin (IL)-6-type cytokine signalling and its regulation.
Bostrom, J., Cobbers, J.M., Wolter, M., Tabatabai, G., Weber, R.G., Lichter, P., Biochem. J. 374, 1e20.
Collins, V.P., Reifenberger, G., 1998. Mutation of the PTEN (MMAC1) tumor Hewing, N.J., Weskamp, G., Vermaat, J., Farage, E., Glomski, K., Swendeman, S.,
suppressor gene in a subset of glioblastomas but not in meningiomas with loss Chan, R.V., Chiang, M.F., Khokha, R., Anand-Apte, B., Blobel, C.P., 2013. Intra-
of chromosome arm 10q. Cancer Res. 58, 29e33. vitreal injection of TIMP3 or the EGFR inhibitor erlotinib offers protection from
Buchser, W.J., Slepak, T.I., Gutierrez-Arenas, O., Bixby, J.L., Lemmon, V.P., 2010. Ki- oxygen-induced retinopathy in mice. Investig. Ophthalmol. Vis. Sci. 54,
nase/phosphatase overexpression reveals pathways regulating hippocampal 864e870.
neuron morphology. Mol. Syst. Biol. 6, 391. Hirai, H., Tani, T., Kikyo, N., 2010. Structure and functions of powerful trans-
Byrne, A.B., Walradt, T., Gardner, K.E., Hubbert, A., Reinke, V., Hammarlund, M., activators: VP16, MyoD and FoxA. Int. J. Dev. Biol. 54, 1589e1596.
2014. Insulin/IGF1 signaling inhibits age-dependent axon regeneration. Neuron Hirano, T., 1998. Interleukin 6 and its receptor: ten years later. Int. Rev. Immunol. 16,
30 A. Barber et al. / Progress in Retinal and Eye Research 56 (2017) 19e31

249e284. 29e48.
Hirose, K., Shiomi, T., Hozumi, S., Kikuchi, Y., 2014. Mechanistic target of rapamycin Lingor, P., Teusch, N., Schwarz, K., Mueller, R., Mack, H., Bahr, M., Mueller, B.K., 2007.
complex 1 signaling regulates cell proliferation, cell survival, and differentiation Inhibition of Rho kinase (ROCK) increases neurite outgrowth on chondroitin
in regenerating zebrash ns. BMC Dev. Biol. 14, 42. sulphate proteoglycan in vitro and axonal regeneration in the adult optic nerve
Hiwatashi, K., Tamiya, T., Hasegawa, E., Fukaya, T., Hashimoto, M., Kakoi, K., in vivo. J. Neurochem. 103, 181e189.
Kashiwagi, I., Kimura, A., Inoue, N., Morita, R., Yasukawa, H., Yoshimura, A., 2011. Lingor, P., Tonges, L., Pieper, N., Bermel, C., Barski, E., Planchamp, V., Bahr, M., 2008.
Suppression of SOCS3 in macrophages prevents cancer metastasis by modifying ROCK inhibition and CNTF interact on intrinsic signalling pathways and
macrophage phase and MCP2/CCL8 induction. Cancer Lett. 308, 172e180. differentially regulate survival and regeneration in retinal ganglion cells. Brain J.
Huang, X., Wu, D.Y., Chen, G., Manji, H., Chen, D.F., 2003. Support of retinal ganglion Neurol. 131, 250e263.
cell survival and axon regeneration by lithium through a Bcl-2-dependent Liu, J., Gao, H.Y., Wang, X.F., 2015. The role of the Rho/ROCK signaling pathway in
mechanism. Investig. Ophthalmol. Vis. Sci. 44, 347e354. inhibiting axonal regeneration in the central nervous system. Neural Regen. Res.
Icard, P., Lincet, H., 2013. The cancer tumor: a metabolic parasite? Bull. Cancer 100, 10, 1892e1896.
427e433. Liu, K., Lu, Y., Lee, J.K., Samara, R., Willenberg, R., Sears-Kraxberger, I., Tedeschi, A.,
Inagaki-Ohara, K., Mayuzumi, H., Kato, S., Minokoshi, Y., Otsubo, T., Kawamura, Y.I., Park, K.K., Jin, D., Cai, B., Xu, B., Connolly, L., Steward, O., Zheng, B., He, Z., 2010.
Dohi, T., Matsuzaki, G., Yoshimura, A., 2014. Enhancement of leptin receptor PTEN deletion enhances the regenerative ability of adult corticospinal neurons.
signaling by SOCS3 deciency induces development of gastric tumors in mice. Nat. Neurosci. 13, 1075e1081.
Oncogene 33, 74e84. Liu, Y., Senger, D.R., 2004. Matrix-specic activation of Src and Rho initiates capil-
Itoh, K., Yoshioka, K., Akedo, H., Uehata, M., Ishizaki, T., Narumiya, S., 1999. An lary morphogenesis of endothelial cells. FASEB J. Off. Publ. Fed. Am. Soc. Exp.
essential part for Rho-associated kinase in the transcellular invasion of tumor Biol. 18, 457e468.
cells. Nat. Med. 5, 221e225. Liu, Y., Yu, H., Deaton, S.K., Szaro, B.G., 2012. Heterogeneous nuclear ribonucleo-
Jacobson, R.D., Virag, I., Skene, J.H., 1986. A protein associated with axon growth, protein K, an RNA-binding protein, is required for optic axon regeneration in
GAP-43, is widely distributed and developmentally regulated in rat CNS. Xenopus laevis. J. Neurosci. Off. J. Soc. Neurosci. 32, 3563e3574.
J. Neurosci. Off. J. Soc. Neurosci. 6, 1843e1855. Mehta, S.T., Luo, X., Park, K.K., Bixby, J.L., Lemmon, V.P., 2016. Hyperactivated Stat3
Jenkins, B.J., Grail, D., Nheu, T., Najdovska, M., Wang, B., Waring, P., Inglese, M., boosts axon regeneration in the CNS. Exp. Neurol. 280, 115e120.
McLoughlin, R.M., Jones, S.A., Topley, N., Baumann, H., Judd, L.M., Giraud, A.S., Metcalf, D., 1985a. The granulocyte-macrophage colony-stimulating factors. Science
Boussioutas, A., Zhu, H.J., Ernst, M., 2005. Hyperactivation of Stat3 in gp130 229, 16e22.
mutant mice promotes gastric hyperproliferation and desensitizes TGF-beta Metcalf, D., 1985b. The granulocyte-macrophage colony stimulating factors. Cell 43,
signaling. Nat. Med. 11, 845e852. 5e6.
Kaneko, Y., Ohta, M., Inoue, T., Mizuno, K., Isobe, T., Tanabe, S., Tanihara, H., 2016. Midha, A., Dearden, S., McCormack, R., 2015. EGFR mutation incidence in non-
Effects of K-115 (Ripasudil), a novel ROCK inhibitor, on trabecular meshwork small-cell lung cancer of adenocarcinoma histology: a systematic review and
and Schlemm's canal endothelial cells. Sci. Rep. 6, 19640. global map by ethnicity (mutMapII). Am. J. Cancer Res. 5, 2892e2911.
Kaplan, A., Ong Tone, S., Fournier, A.E., 2015. Extrinsic and intrinsic regulation of Miotke, J.A., MacLennan, A.J., Meyer, R.L., 2007. Immunohistochemical localization
axon regeneration at a crossroads. Front. Mol. Neurosci. 8, 27. of CNTFRalpha in adult mouse retina and optic nerve following intraorbital
Kon, H., Sonoda, Y., Kumabe, T., Yoshimoto, T., Sekiya, T., Murakami, Y., 1998. nerve crush: evidence for the axonal loss of a trophic factor receptor after
Structural and functional evidence for the presence of tumor suppressor genes injury. J. Comp. Neurol. 500, 384e400.
on the short arm of chromosome 10 in human gliomas. Oncogene 16, 257e263. Mohammad, R.M., Muqbil, I., Lowe, L., Yedjou, C., Hsu, H.Y., Lin, L.T., Siegelin, M.D.,
Koprivica, V., Cho, K.S., Park, J.B., Yiu, G., Atwal, J., Gore, B., Kim, J.A., Lin, E., Tessier- Fimognari, C., Kumar, N.B., Dou, Q.P., Yang, H., Samadi, A.K., Russo, G.L.,
Lavigne, M., Chen, D.F., He, Z., 2005. EGFR activation mediates inhibition of axon Spagnuolo, C., Ray, S.K., Chakrabarti, M., Morre, J.D., Coley, H.M., Honoki, K.,
regeneration by myelin and chondroitin sulfate proteoglycans. Science 310, Fujii, H., Georgakilas, A.G., Amedei, A., Niccolai, E., Amin, A., Ashraf, S.S.,
106e110. Helferich, W.G., Yang, X., Boosani, C.S., Guha, G., Bhakta, D., Ciriolo, M.R.,
Kurimoto, T., Yin, Y., Omura, K., Gilbert, H.Y., Kim, D., Cen, L.P., Moko, L., Kugler, S., Aquilano, K., Chen, S., Mohammed, S.I., Keith, W.N., Bilsland, A., Halicka, D.,
Benowitz, L.I., 2010. Long-distance axon regeneration in the mature optic nerve: Nowsheen, S., Azmi, A.S., 2015. Broad targeting of resistance to apoptosis in
contributions of oncomodulin, cAMP, and pten gene deletion. J. Neurosci. Off. J. cancer. Semin. Cancer Biol. (35 Suppl), S78eS103.
Soc. Neurosci. 30, 15654e15663. Monnier, P.P., Sierra, A., Schwab, J.M., Henke-Fahle, S., Mueller, B.K., 2003. The Rho/
Lahne, M., Li, J., Marton, R.M., Hyde, D.R., 2015. Actin-Cytoskeleton- and Rock- ROCK pathway mediates neurite growth-inhibitory activity associated with the
mediated INM are required for photoreceptor regeneration in the adult chondroitin sulfate proteoglycans of the CNS glial scar. Mol. Cell. Neurosci. 22,
Zebrash retina. J. Neurosci. Off. J. Soc. Neurosci. 35, 15612e15634. 319e330.
Lapan, S.W., Reddien, P.W., 2012. Transcriptome analysis of the planarian eye Moore, D.L., Blackmore, M.G., Hu, Y., Kaestner, K.H., Bixby, J.L., Lemmon, V.P.,
identies ovo as a specic regulator of eye regeneration. Cell Rep. 2, 294e307. Goldberg, J.L., 2009. KLF family members regulate intrinsic axon regeneration
Legacy, J., Hanea, S., Theoret, J., Smith, P.D., 2013. Granulocyte macrophage colony- ability. Science 326, 298e301.
stimulating factor promotes regeneration of retinal ganglion cells in vitro Muller, A., Hauk, T.G., Fischer, D., 2007. Astrocyte-derived CNTF switches mature
through a mammalian target of rapamycin-dependent mechanism. J. Neurosci. RGCs to a regenerative state following inammatory stimulation. Brain J.
Res. 91, 771e779. Neurol. 130, 3308e3320.
Leibinger, M., Andreadaki, A., Gobrecht, P., Levin, E., Diekmann, H., Fischer, D., 2016. Narla, G., DiFeo, A., Yao, S., Banno, A., Hod, E., Reeves, H.L., Qiao, R.F., Camacho-
Boosting central nervous system axon regeneration by circumventing limita- Vanegas, O., Levine, A., Kirschenbaum, A., Chan, A.M., Friedman, S.L.,
tions of natural cytokine signaling. Mol. Mol. Ther. J. Am. Soc. Gene Ther. http:// Martignetti, J.A., 2005. Targeted inhibition of the KLF6 splice variant, KLF6 SV1,
dx.doi.org/10.1038/mt.2016.102. suppresses prostate cancer cell growth and spread. Cancer Res. 65, 5761e5768.
Leibinger, M., Muller, A., Andreadaki, A., Hauk, T.G., Kirsch, M., Fischer, D., 2009. Narla, G., Heath, K.E., Reeves, H.L., Li, D., Giono, L.E., Kimmelman, A.C.,
Neuroprotective and axon growth-promoting effects following inammatory Glucksman, M.J., Narla, J., Eng, F.J., Chan, A.M., Ferrari, A.C., Martignetti, J.A.,
stimulation on mature retinal ganglion cells in mice depend on ciliary neuro- Friedman, S.L., 2001. KLF6, a candidate tumor suppressor gene mutated in
trophic factor and leukemia inhibitory factor. J. Neurosci. Off. J. Soc. Neurosci. prostate cancer. Science 294, 2563e2566.
29, 14334e14341. Nicholson, S.E., De Souza, D., Fabri, L.J., Corbin, J., Willson, T.A., Zhang, J.G., Silva, A.,
Leibinger, M., Muller, A., Gobrecht, P., Diekmann, H., Andreadaki, A., Fischer, D., Asimakis, M., Farley, A., Nash, A.D., Metcalf, D., Hilton, D.J., Nicola, N.A., Baca, M.,
2013. Interleukin-6 contributes to CNS axon regeneration upon inammatory 2000. Suppressor of cytokine signaling-3 preferentially binds to the SHP-2-
stimulation. Cell Death Dis. 4, e609. binding site on the shared cytokine receptor subunit gp130. Proc. Natl. Acad.
Lesina, M., Kurkowski, M.U., Ludes, K., Rose-John, S., Treiber, M., Kloppel, G., Sci. U. S. A. 97, 6493e6498.
Yoshimura, A., Reindl, W., Sipos, B., Akira, S., Schmid, R.M., Algul, H., 2011. Stat3/ Ogai, K., Kuwana, A., Hisano, S., Nagashima, M., Koriyama, Y., Sugitani, K.,
Socs3 activation by IL-6 transsignaling promotes progression of pancreatic Mawatari, K., Nakashima, H., Kato, S., 2014. Upregulation of leukemia inhibitory
intraepithelial neoplasia and development of pancreatic cancer. Cancer Cell 19, factor (LIF) during the early stage of optic nerve regeneration in zebrash. PLoS
456e469. One 9, e106010.
Li, B., Zhao, W.D., Tan, Z.M., Fang, W.G., Zhu, L., Chen, Y.H., 2006. Involvement of Paramio, J.M., Navarro, M., Segrelles, C., Gomez-Casero, E., Jorcano, J.L., 1999. PTEN
Rho/ROCK signalling in small cell lung cancer migration through human brain tumour suppressor is linked to the cell cycle control through the retinoblas-
microvascular endothelial cells. FEBS Lett. 580, 4252e4260. toma protein. Oncogene 18, 7462e7468.
Li, J., Yen, C., Liaw, D., Podsypanina, K., Bose, S., Wang, S.I., Puc, J., Miliaresis, C., Park, K.K., Liu, K., Hu, Y., Smith, P.D., Wang, C., Cai, B., Xu, B., Connolly, L., Kramvis, I.,
Rodgers, L., McCombie, R., Bigner, S.H., Giovanella, B.C., Ittmann, M., Tycko, B., Sahin, M., He, Z., 2008. Promoting axon regeneration in the adult CNS by
Hibshoosh, H., Wigler, M.H., Parsons, R., 1997. PTEN, a putative protein tyrosine modulation of the PTEN/mTOR pathway. Science 322, 963e966.
phosphatase gene mutated in human brain, breast, and prostate cancer. Science Pernet, V., Joly, S., Jordi, N., Dalkara, D., Guzik-Kornacka, A., Flannery, J.G.,
275, 1943e1947. Schwab, M.E., 2013. Misguidance and modulation of axonal regeneration by
Li, X., Yang, Q., Yu, H., Wu, L., Zhao, Y., Zhang, C., Yue, X., Liu, Z., Wu, H., Haffty, B.G., Stat3 and Rho/ROCK signaling in the transparent optic nerve. Cell Death Dis. 4,
Feng, Z., Hu, W., 2014. LIF promotes tumorigenesis and metastasis of breast e734.
cancer through the AKT-mTOR pathway. Oncotarget 5, 788e801. Peters, M., Blinn, G., Solem, F., Fischer, M., Meyer zum Buschenfelde, K.H., Rose-
Lilja, A., Nordborg, C., Brun, A., Salford, L.G., Aman, P., 2001. Expression of the IL-6 John, S., 1998. In vivo and in vitro activities of the gp130-stimulating designer
family cytokines in human brain tumors. Int. J. Oncol. 19, 495e499. cytokine Hyper-IL-6. J. Immunol. 161, 3575e3581.
Limame, R., Op de Beeck, K., Lardon, F., De Wever, O., Pauwels, P., 2014. Kruppel-like Pomerantz, J.H., Blau, H.M., 2013. Tumor suppressors: enhancers or suppressors of
factors in cancer progression: three ngers on the steering wheel. Oncotarget 5, regeneration? Development 140, 2502e2512.
A. Barber et al. / Progress in Retinal and Eye Research 56 (2017) 19e31 31

Pruitt, K., Der, C.J., 2001. Ras and Rho regulation of the cell cycle and oncogenesis. inhibition of cell invasion, migration, and growth: differential involvement of
Cancer Lett. 171, 1e10. focal adhesion kinase and p130Cas. Cancer Res. 59, 442e449.
Puhr, M., Santer, F.R., Neuwirt, H., Susani, M., Nemeth, J.A., Hobisch, A., Kenner, L., Tamura, M., Gu, J., Tran, H., Yamada, K.M., 1999b. PTEN gene and integrin signaling
Culig, Z., 2009. Down-regulation of suppressor of cytokine signaling-3 causes in cancer. J. Natl. Cancer Inst. 91, 1820e1828.
prostate cancer cell death through activation of the extrinsic and intrinsic Tan, H., Zhong, Y., Shen, X., Cheng, Y., Jiao, Q., Deng, L., 2012. Erythropoietin pro-
apoptosis pathways. Cancer Res. 69, 7375e7384. motes axonal regeneration after optic nerve crush in vivo by inhibition of RhoA/
Qin, S., Zou, Y., Zhang, C.L., 2013. Cross-talk between KLF4 and STAT3 regulates axon ROCK signaling pathway. Neuropharmacology 63, 1182e1190.
regeneration. Nat. Commun. 4, 2633. Turner, J.E., Delaney, R.K., 1979. Retinal ganglion cell response to axotomy and nerve
Rath, N., Olson, M.F., 2012. Rho-associated kinases in tumorigenesis: re-considering growth factor in the regenerating visual system of the newt (Notophthalmus
ROCK inhibition for cancer therapy. EMBO Rep. 13, 900e908. viridescens): an ultrastructural morphometric analysis. Brain Res. 171, 197e212.
Sagawa, H., Terasaki, H., Nakamura, M., Ichikawa, M., Yata, T., Tokita, Y., Veldman, M.B., Bemben, M.A., Thompson, R.C., Goldman, D., 2007. Gene expression
Watanabe, M., 2007. A novel ROCK inhibitor, Y-39983, promotes regeneration of analysis of zebrash retinal ganglion cells during optic nerve regeneration
crushed axons of retinal ganglion cells into the optic nerve of adult cats. Exp. identies KLF6a and KLF7a as important regulators of axon regeneration. Dev.
Neurol. 205, 230e240. Biol. 312, 596e612.
Schonig, K., Weber, T., Frommig, A., Wendler, L., Pesold, B., Djandji, D., Bujard, H., Vousden, K.H., Prives, C., 2009. Blinded by the light: the growing complexity of p53.
Bartsch, D., 2012. Conditional gene expression systems in the transgenic rat Cell 137, 413e431.
brain. BMC Biol. 10, 77. Wang, J., Liu, X., Zhong, Y., 2013. Rho/Rho-associated kinase pathway in glaucoma
Schuettpelz, L.G., Gopalan, P.K., Giuste, F.O., Romine, M.P., van Os, R., Link, D.C., 2012. (review). Int. J. Oncol. 43, 1357e1367.
Kruppel-like factor 7 overexpression suppresses hematopoietic stem and pro- Watanabe, M., 2010. Regeneration of optic nerve bers of adult mammals. Dev.
genitor cell function. Blood 120, 2981e2989. Growth Differ. 52, 567e576.
Scimone, M.L., Kravarik, K.M., Lapan, S.W., Reddien, P.W., 2014. Neoblast speciali- Watkins, T.A., Wang, B., Huntwork-Rodriguez, S., Yang, J., Jiang, Z., Eastham-
zation in regeneration of the planarian Schmidtea mediterranea. Stem Cell Rep. Anderson, J., Modrusan, Z., Kaminker, J.S., Tessier-Lavigne, M., Lewcock, J.W.,
3, 339e352. 2013. DLK initiates a transcriptional program that couples apoptotic and
Silva, A.K., Yi, H., Hayes, S.H., Seigel, G.M., Hackam, A.S., 2010. Lithium chloride regenerative responses to axonal injury. Proc. Natl. Acad. Sci. U. S. A. 110,
regulates the proliferation of stem-like cells in retinoblastoma cell lines: a 4039e4044.
potential role for the canonical Wnt signaling pathway. Mol. Vis. 16, 36e45. Weiss, R., 1982. The myc oncogene in man and birds. Nature 299, 9e10.
Simpson, L., Parsons, R., 2001. PTEN: life as a tumor suppressor. Exp. Cell Res. 264, Weng, L., Brown, J., Eng, C., 2001. PTEN induces apoptosis and cell cycle arrest
29e41. through phosphoinositol-3-kinase/Akt-dependent and -independent pathways.
Simpson, M.T., Venkatesh, I., Callif, B.L., Thiel, L.K., Coley, D.M., Winsor, K.N., Hum. Mol. Genet. 10, 237e242.
Wang, Z., Kramer, A.A., Lerch, J.K., Blackmore, M.G., 2015. The tumor suppressor Weng, L.P., Smith, W.M., Dahia, P.L., Ziebold, U., Gil, E., Lees, J.A., Eng, C., 1999. PTEN
HHEX inhibits axon growth when prematurely expressed in developing central suppresses breast cancer cell growth by phosphatase activity-dependent G1
nervous system neurons. Mol. Cell. Neurosci. 68, 272e283. arrest followed by cell death. Cancer Res. 59, 5808e5814.
Sirach, E., Bureau, C., Peron, J.M., Pradayrol, L., Vinel, J.P., Buscail, L., Cordelier, P., Wilson, A.M., Morquette, B., Abdouh, M., Unsain, N., Barker, P.A., Feinstein, E.,
2007. KLF6 transcription factor protects hepatocellular carcinoma-derived cells Bernier, G., Di Polo, A., 2013. ASPP1/2 regulate p53-dependent death of retinal
from apoptosis. Cell Death Differ. 14, 1202e1210. ganglion cells through PUMA and Fas/CD95 activation in vivo. J. Neurosci. Off. J.
Sitorus, R.S., Gumay, S., van der Valk, P., 2009. The apoptotic paradox in retino- Soc. Neurosci. 33, 2205e2216.
blastoma. Ann. N. Y. Acad. Sci. 1171, 77e86. Wu, Y., Yan, Q., Zuo, J., Saiyin, H., Jiang, W., Qiao, S., Yu, L., 2010. Link of Dlk/ZIP
Smith, P.D., Coulson-Thomas, V.J., Foscarin, S., Kwok, J.C., Fawcett, J.W., 2015. GAG- kinase to cell apoptosis and tumor suppression. Biochem. Biophys. Res. Com-
ing with the neuron: the role of glycosaminoglycan patterning in the central mun. 392, 510e515.
nervous system. Exp. Neurol. 274, 100e114. Xie, C., Lu, H., Nomura, A., Hanse, E.A., Forster, C.L., Parker, J.B., Linden, M.A.,
Smith, P.D., Sun, F., Park, K.K., Cai, B., Wang, C., Kuwako, K., Martinez-Carrasco, I., Karasch, C., Hallstrom, T.C., 2015. Co-deleting Pten with Rb in retinal progenitor
Connolly, L., He, Z., 2009. SOCS3 deletion promotes optic nerve regeneration cells in mice results in fully penetrant bilateral retinoblastomas. Mol. Cancer 14,
in vivo. Neuron 64, 617e623. 93.
Song, M.S., Salmena, L., Pandol, P.P., 2012a. The functions and regulation of the Yin, Y., Liu, W., Dai, Y., 2015. SOCS3 and its role in associated diseases. Hum.
PTEN tumour suppressor. Nat. Rev. Mol. Cell Biol. 13, 283e296. Immunol. 76, 775e780.
Song, Y., Ori-McKenney, K.M., Zheng, Y., Han, C., Jan, L.Y., Jan, Y.N., 2012b. Regen- Zhang, Q., Tan, X.P., Yuan, Y.S., Hu, C.M., He, C.H., Wang, W.Z., Li, J.C., Zhao, Q.,
eration of Drosophila sensory neuron axons and dendrites is regulated by the Liu, N.Z., 2010. Decreased expression of KLF6 and its signicance in gastric
Akt pathway involving Pten and microRNA bantam. Genes Dev. 26, 1612e1625. carcinoma. Med. Oncol. 27, 1295e1302.
Su, J., You, P., Zhao, J.P., Zhang, S.L., Song, S.H., Fu, Z.R., Ye, L.W., Zi, X.Y., Xie, D.F., Zhenzhen, Z., De'an, T., Limin, X., Wei, Y., Min, L., 2012. New candidate tumor-
Zhu, M.H., Hu, Y.P., 2012. A potential role for the homeoprotein Hhex in he- suppressor gene KLF6 and its splice variant KLF6 SV2 counterbalancing
patocellular carcinoma progression. Med. Oncol. 29, 1059e1067. expression in primary hepatocarcinoma. Hepato-Gastroenterology 59,
Sun, F., Park, K.K., Belin, S., Wang, D., Lu, T., Chen, G., Zhang, K., Yeung, C., Feng, G., 473e476.
Yankner, B.A., He, Z., 2011. Sustained axon regeneration induced by co-deletion Zochodne, D.W., 2014. Neurons and tumor suppressors. ACS Chem. Neurosci. 5,
of PTEN and SOCS3. Nature 480, 372e375. 618e620.
Sun, J., Wang, B., Liu, Y., Zhang, L., Ma, A., Yang, Z., Ji, Y., Liu, Y., 2014. Transcription Zohrabian, V.M., Forzani, B., Chau, Z., Murali, R., Jhanwar-Uniyal, M., 2009. Rho/
factor KLF9 suppresses the growth of hepatocellular carcinoma cells in vivo and ROCK and MAPK signaling pathways are involved in glioblastoma cell migration
positively regulates p53 expression. Cancer Lett. 355, 25e33. and proliferation. Anticancer Res. 29, 119e123.
Tamura, M., Gu, J., Matsumoto, K., Aota, S., Parsons, R., Yamada, K.M., 1998. Inhibi- Zukor, K., Belin, S., Wang, C., Keelan, N., Wang, X., He, Z., 2013. Short hairpin RNA
tion of cell migration, spreading, and focal adhesions by tumor suppressor against PTEN enhances regenerative growth of corticospinal tract axons after
PTEN. Science 280, 1614e1617. spinal cord injury. J. Neurosci. Off. J. Soc. Neurosci. 33, 15350e15361.
Tamura, M., Gu, J., Takino, T., Yamada, K.M., 1999a. Tumor suppressor PTEN

Anda mungkin juga menyukai