Retinal regeneration mechanisms linked to multiple cancer molecules: A therapeutic conundrum

https://doi.org/10.1016/j.preteyeres.2016.08.003Get rights and content

Abstract

Over the last decade, a large number of research articles have been published demonstrating regeneration and/or neuroprotection of retinal ganglion cells following manipulation of specific genetic and molecular targets. Interestingly, of the targets that have been identified to promote repair following visual system damage, many are genes known to be mutated in different types of cancer. This review 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 similar mechanisms may improve axonal regeneration in the optic nerve.

Introduction

In recent years there has been an exponential increase in the number of publications reporting cell survival and/or axon regeneration in models of optic nerve injury, for example where intraocular pressure is elevated to induce retinal ganglion cell (RGC) loss or following localised optic nerve crush. Numerous factors with diverse physiological functions, including mammalian target of rapamycin (mTOR), kruppel-like family (KLF) transcription factors, suppressor of cytokine signalling 3 (SOCS3) and cytokines (ciliary neurotrophic factor (CNTF)/leukemia inhibitory factor (LIF)), have been suggested to play a key role in RGC survival and/or axon regeneration. Proposed mechanisms include effects on the availability of neurotrophic factors, activation of intrinsic survival/growth mechanisms, modulation of axonal transport and glia-dependent support as well as inhibition of apoptotic mechanisms in RGCs. Interestingly, many of the mechanisms that have been suggested to promote either survival and/or axon regeneration in the damaged visual system are directly or indirectly associated with cancer, including malignancies that affect the eye and visual pathways.

Cancer can be defined as the loss of normal cellular growth control where unregulated proliferation of cells result in malignant pathologies (De Potter et al., 2002). In contrast, failure of neuronal regeneration can be considered as an intrinsic inability to re-activate the normal growth mechanisms that occur during development. In this sense, cancer and regenerative failure could be viewed as different sides of the same coin. The central nervous system (CNS) can be stimulated to regenerate by manipulation of a variety of cancer-associated molecular signalling pathways and excellent reviews have been published on this subject recently (Benowitz et al., 2015, He and Jin, 2016, Kaplan et al., 2015, Smith et al., 2015). Of the identified regeneration-associated genes, many are tumour suppressors, including phosphatase and tensin homolog (PTEN (Park et al., 2008)) or oncogenes such as MYC (Buchser et al., 2010, Pomerantz and Blau, 2013). In this review, we will focus on cancer genes and signalling mechanisms that have been proposed to underlie both malignancy and regeneration in the mature nervous system. We will review several signalling pathways, including PTEN/mTOR pathway, phosphatidylinositol-3-kinase (PI3K), cytokine and inflammatory factors including SOCS-3, trophic factors (such as CNTF), anti-apoptotic factors (including Bcl-2), KLF, transcription factors (including signal transducer and activator of transcription-3 (STAT-3)) as well as additional proposed mechanisms that are known to be involved in cancer pathology but that have also been implicated in axon regeneration. The optic nerve crush model has been extensively utilized to evaluate the impact of modulating different signalling factors on axon regeneration in a physiological setting and has been important in defining signalling mechanisms involved in promoting axon regeneration in the adult mammalian CNS. A major question is whether manipulation of genes involved in cancer can be considered as a feasible mechanism to promote axon regeneration in a clinical setting, where the potential for oncogenesis is a concern. However, it is conceivable that the post-mitotic nature of neurons in the mature visual system may limit this risk of malignancy. In this review we will also consider how oncogenic risk might be mitigated through careful selection and application of the pharmacological or molecular tools used to manipulate cancer-associated pathways.

Section snippets

Signalling mechanisms involved in cancer and axon regeneration

In this section we will focus on cancer genes and signalling mechanisms that have been proposed to underlie both malignancy and regeneration in the mature nervous system.

Less explored potential targets

Manipulating cancer-associated genes to promote repair and recovery in the damaged CNS may appear counter-intuitive, but it is worth noting that nature uses these growth mechanisms for precisely this purpose in other organisms while avoiding oncogenesis. Indeed some non-mammalian species, namely zebrafish, xenopus and other anamniotes, retain the capacity to regenerate CNS axons well into the adult years (Turner and Delaney, 1979). Several factors, discussed in this section, have been

Approaches to reducing cancer risk when promoting regeneration

Neuronal regenerative therapies that target cancer –associated molecular pathways raise a serious concern regarding oncogenesis, although conceivably this risk may be limited by the post-mitotic nature of the mature visual system. Genes may function differently in a post-mitotic cellular context, thereby allowing the modulation of tumour suppressors without enacting oncogenic mechanisms. Thus it is plausible that modulation of tumour suppressors, if targeted in a spatially and temporally

Beyond axonal growth: challenges for repair

In order to promote repair and recovery following CNS damage, neurons must survive and then extend regenerating axons, which must then traverse the hostile inhibitory CNS environment and find appropriate synaptic targets to allow functional circuits to be re-established. However manipulating factors which promote axonal growth may not be sufficient to restore functions such as useful vision. In addition, the therapeutic time window for axonal regeneration will need to be established and the

Conclusion and future directions

It is clear that where most extensive axon regeneration has been observed in the adult CNS to date it has resulted from manipulation or indirect regulation of cancer associated pathways (see Table 1 for summary of selected factors discussed here). Many of these studies have exclusively focused on the regenerative potential of manipulated cancer associated genes but it is clear that the long term oncogenic risk will need to be assessed if such therapies can be considered as a realistic option

References (138)

  • S.T. Hanea et al.

    Preparation of embryonic retinal explants to study CNS neurite growth

    Exp. Eye Res.

    (2016)
  • L.S. Harrington et al.

    Restraining PI3K: mTOR signalling goes back to the membrane

    Trends Biochem. Sci.

    (2005)
  • Z. He et al.

    Intrinsic control of axon regeneration

    Neuron

    (2016)
  • K. Hiwatashi et al.

    Suppression of SOCS3 in macrophages prevents cancer metastasis by modifying macrophage phase and MCP2/CCL8 induction

    Cancer Lett.

    (2011)
  • P. Icard et al.

    The cancer tumor: a metabolic parasite?

    Bull. Cancer

    (2013)
  • S.W. Lapan et al.

    Transcriptome analysis of the planarian eye identifies ovo as a specific regulator of eye regeneration

    Cell Rep.

    (2012)
  • M. Lesina et al.

    Stat3/Socs3 activation by IL-6 transsignaling promotes progression of pancreatic intraepithelial neoplasia and development of pancreatic cancer

    Cancer Cell

    (2011)
  • B. Li et al.

    Involvement of Rho/ROCK signalling in small cell lung cancer migration through human brain microvascular endothelial cells

    FEBS Lett.

    (2006)
  • S.T. Mehta et al.

    Hyperactivated Stat3 boosts axon regeneration in the CNS

    Exp. Neurol.

    (2016)
  • D. Metcalf

    The granulocyte-macrophage colony stimulating factors

    Cell

    (1985)
  • R.M. Mohammad et al.

    Broad targeting of resistance to apoptosis in cancer

    Semin. Cancer Biol.

    (2015)
  • P.P. Monnier et al.

    The Rho/ROCK pathway mediates neurite growth-inhibitory activity associated with the chondroitin sulfate proteoglycans of the CNS glial scar

    Mol. Cell. Neurosci.

    (2003)
  • K. Pruitt et al.

    Ras and Rho regulation of the cell cycle and oncogenesis

    Cancer Lett.

    (2001)
  • T. Anastassiadis et al.

    Comprehensive assay of kinase catalytic activity reveals features of kinase inhibitor selectivity

    Nat. Biotechnol.

    (2011)
  • A. Apara et al.

    Molecular mechanisms of the suppression of axon regeneration by KLF transcription factors

    Neural Regen. Res.

    (2014)
  • W.S. Beane et al.

    Inhibition of planar cell polarity extends neural growth during regeneration, homeostasis, and development

    Stem Cells Dev.

    (2012)
  • Y. Bei et al.

    RhoA/Rho-kinase activation promotes lung fibrosis in an animal model of systemic sclerosis

    Exp. Lung Res.

    (2016)
  • L.I. Benowitz et al.

    Reaching the brain: advances in optic nerve regeneration

    Exp. Neurol.

    (2015)
  • M.D. Benson et al.

    Ephrin-B3 is a myelin-based inhibitor of neurite outgrowth

    Proc. Natl. Acad. Sci. U. S. A.

    (2005)
  • J. Bertrand et al.

    Application of Rho antagonist to neuronal cell bodies promotes neurite growth in compartmented cultures and regeneration of retinal ganglion cell axons in the optic nerve of adult rats

    J. Neurosci. Off. J. Soc. Neurosci.

    (2005)
  • M.G. Blackmore et al.

    Kruppel-like Factor 7 engineered for transcriptional activation promotes axon regeneration in the adult corticospinal tract

    Proc. Natl. Acad. Sci. U. S. A.

    (2012)
  • S.D. Boomkamp et al.

    The development of a rat in vitro model of spinal cord injury demonstrating the additive effects of Rho and ROCK inhibitors on neurite outgrowth and myelination

    Glia

    (2012)
  • J. Bostrom et al.

    Mutation of the PTEN (MMAC1) tumor suppressor gene in a subset of glioblastomas but not in meningiomas with loss of chromosome arm 10q

    Cancer Res.

    (1998)
  • W.J. Buchser et al.

    Kinase/phosphatase overexpression reveals pathways regulating hippocampal neuron morphology

    Mol. Syst. Biol.

    (2010)
  • A. Charest et al.

    ROS fusion tyrosine kinase activates a SH2 domain-containing phosphatase-2/phosphatidylinositol 3-kinase/mammalian target of rapamycin signaling axis to form glioblastoma in mice

    Cancer Res.

    (2006)
  • D.F. Chen et al.

    Bcl-2 promotes regeneration of severed axons in mammalian CNS

    Nature

    (1997)
  • H. Chen et al.

    Epidermal growth factor receptor in adult retinal neurons of rat, mouse, and human

    J. Comp. Neurol.

    (2007)
  • Z.Y. Chen et al.

    Role of Rho/ROCK in the migration of vascular smooth muscle cells

    Sheng li ke xue jin zhan [Prog. Physiol.]

    (2013)
  • V.T. Chin et al.

    Rho-associated kinase signalling and the cancer microenvironment: novel biological implications and therapeutic opportunities

    Expert Rev. Mol. Med.

    (2015)
  • K.J. Christie et al.

    PTEN inhibition to facilitate intrinsic regenerative outgrowth of adult peripheral axons

    J. Neurosci. Off. J. Soc. Neurosci.

    (2010)
  • S. de Lima et al.

    Full-length axon regeneration in the adult mouse optic nerve and partial recovery of simple visual behaviors

    Proc. Natl. Acad. Sci. U. S. A.

    (2012)
  • P. De Potter et al.

    Ocular manifestations of cancer

    J. Fr. d'ophtalmologie

    (2002)
  • H. Diekmann et al.

    Active mechanistic target of rapamycin plays an ancillary rather than essential role in zebrafish CNS axon regeneration

    Front. Cell. Neurosci.

    (2015)
  • F. Elsaeidi et al.

    Jak/Stat signaling stimulates zebrafish optic nerve regeneration and overcomes the inhibitory actions of Socs3 and Sfpq

    J. Neurosci. Off. J. Soc. Neurosci.

    (2014)
  • R. Endersby et al.

    PTEN signaling in brain: neuropathology and tumorigenesis

    Oncogene

    (2008)
  • J.W. Fawcett

    The glial response to injury and its role in the inhibition of CNS repair

    Adv. Exp. Med. Biol.

    (2006)
  • D. Fischer et al.

    Switching mature retinal ganglion cells to a robust growth state in vivo: gene expression and synergy with RhoA inactivation

    J. Neurosci. Off. J. Soc. Neurosci.

    (2004)
  • S.H. Friend et al.

    A human DNA segment with properties of the gene that predisposes to retinoblastoma and osteosarcoma

    Nature

    (1986)
  • M. Gallardo et al.

    Aberrant hnRNP K expression: all roads lead to cancer

    Cell Cycle

    (2016)
  • P. Gaub et al.

    The histone acetyltransferase p300 promotes intrinsic axonal regeneration

    Brain J. Neurol.

    (2011)
  • Cited by (0)

    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%.

    View full text