Elsevier

Experimental Neurology

Volume 260, October 2014, Pages 19-32
Experimental Neurology

Review
Neuro-immune interactions of neural stem cell transplants: From animal disease models to human trials

https://doi.org/10.1016/j.expneurol.2013.03.009Get rights and content

Abstract

Stem cell technology is a promising branch of regenerative medicine that is aimed at developing new approaches for the treatment of severely debilitating human diseases, including those affecting the central nervous system (CNS).

Despite the increasing understanding of the mechanisms governing their biology, the application of stem cell therapeutics remains challenging. The initial idea that stem cell transplants work in vivo via the replacement of endogenous cells lost or damaged owing to disease has been challenged by accumulating evidence of their therapeutic plasticity. This new concept covers the remarkable immune regulatory and tissue trophic effects that transplanted stem cells exert at the level of the neural microenvironment to promote tissue healing via combination of immune modulatory and tissue protective actions, while retaining predominantly undifferentiated features.

Among a number of promising candidate stem cell sources, neural stem/precursor cells (NPCs) are under extensive investigation with regard to their therapeutic plasticity after transplantation. The significant impact in vivo of experimental NPC therapies in animal models of inflammatory CNS diseases has raised great expectations that these stem cells, or the manipulation of the mechanisms behind their therapeutic impact, could soon be translated to human studies.

This review aims to provide an update on the most recent evidence of therapeutically-relevant neuro-immune interactions following NPC transplants in animal models of multiple sclerosis, cerebral stroke and traumas of the spinal cord, and consideration of the forthcoming challenges related to the early translation of some of these exciting experimental outcomes into clinical medicines.

Introduction

The discovery of adult neurogenesis and the development of protocols that allow in vitro growth and significantly large scale-up of stem and precursor cells of the brain (Reynolds and Weiss, 1992) have fostered the development of innovative therapies aimed at stem cell transplantation for acute and chronic disorders of the nervous system (Cossetti et al., 2012). Motivated by the expectation of achieving CNS repair and/or regeneration via functional neural cell replacement, these studies have demonstrated a potential benefit of neural stem/precursor cell (NPC)-based experimental treatments in animal models of several neurological diseases (Martino et al., 2011). However, mounting evidence suggests that the effects orchestrated by transplanted NPCs are not only associated with the generation of new neurons or glial cells but also that the pathological setting in which these cells are transplanted critically determines the outcome (Cossetti et al., 2012). Cell replacement is therefore only one of the multiple ways in which transplanted NPCs promote tissue repair, and a much more complex therapeutic scenario should be foreseen. The concept of stem cell therapeutic plasticity (Martino and Pluchino, 2006) (or functional multipotency) (Teng et al., 2011) has therefore emerged, as it describes the multiple way(s) grafted NPCs which mediate systemic homeostasis, e.g. by the secretion of tissue trophic factors, as well as interaction with tissue-resident vs. -infiltrating immune cells, at the level of the inflammatory tissue context in which they are either transplanted or to which they migrate after transplantation.

The newest picture is therefore that stem cell therapies, contrary to single-molecule-based pharmaceutical interventions, hold the potential to deliver a complex series of information to a multitude of targets in the diseased microenvironment (Cossetti et al., 2012). While no final mechanisms (or direct evidence) of stem cell-to-host immune system interaction is yet available, a number of studies are now focussing on the cellular signalling that exists between grafted stem cells and endogenous target cells, with the aim of clarifying its physiological or circumstantial nature, and elucidating its molecular signature and therapeutic potential.

Here we will review the most recent evidence of immune modulation following syngeneic NPC transplants in animal models of multiple sclerosis, spinal cord injury and stroke, and discuss the next challenges related to the translation of some of these exciting experimental outcomes into clinical medicines.

Section snippets

Multiple sclerosis

Multiple sclerosis (MS) is a complex, highly debilitating CNS autoimmune disease that constitutes the most common cause of neurological disability in young adults (Compston and Coles, 2002). The main pathological hallmark of MS is the presence of highly heterogeneous, chronic inflammatory and demyelinating perivascular lesions within the CNS (Compston and Coles, 2002, Dyment and Ebers, 2002, Flugel et al., 2001, Lucchinetti et al., 2000, Noseworthy et al., 2000, Wingerchuk et al., 2001). Most

Spinal cord injuries

Spinal cord injuries (SCIs) are devastating and debilitating conditions affecting all regions of the world – predominantly in young adults – which are associated with severe physical, psychological, social and economic burdens on patients and their families [reviewed in Ho et al. (2007) and van den Berg et al. (2010)]. An important premise for the development of effective treatments for SCIs is the precise understanding of the main pathophysiological events following the acute injury and how

Stroke

Clinical recovery after stroke remains very poor despite advances in therapy, and stem cell treatment is considered a promising alternative (Lindvall and Kokaia, 2011). Transplantation of NPCs with different delivery strategies, intraparenchymal (ipc) or icv injection, as well as systemic administration, has been shown to improve clinical signs in experimental stroke models (Liu et al., 2009, Pluchino et al., 2010).

Irrespective of the route of administration, transplanted NPCs migrate towards

Towards clinical trials

Based on the encouraging results collected pre-clinically during the last 5–7 years (Table 1), phase I clinical trials have started to be conducted, both in fatal and non-fatal incurable neurological diseases where the risk/benefit ratio is in theory favourable (Aboody et al., 2011). Besides the unquestionable care regarding the characterisation and manufacture of the medicinal product (Rayment and Williams, 2010), one of the other important hurdles in the design of clinical study for (stem)

Conclusions and future directions

Stem cell-based therapies hold great promise in regenerative neuroscience (Park et al., 2010). The huge advances made over the last few years have enormously deepened our knowledge about the biology of stem cells, leading to global reconsideration of their therapeutic potential and mechanisms of action, as well as their intrinsic limitations (Martino et al., 2011). Importantly, the use of animal models that closely mimic different aspects of human pathologies has also contributed to increasing

Acknowledgments

The authors are grateful to Gianvito Martino for the vision and continuous inspiration, Jayden A. Smith for critically reviewing the article, Paula Francis for proof edits and Letterio Politi for the diffusion weighted image shown in Fig. 1. This work has received support from the National Multiple Sclerosis Society (NMSS, partial grant RG-4001-A1), the Italian Multiple Sclerosis Association (AISM, grant 2010/R/31), the Italian Ministry of Health (GR08-7), Wings for Life, Banca Agricola

References (194)

  • W. Cao et al.

    Leukemia inhibitory factor inhibits T helper 17 cell differentiation and confers treatment effects of neural progenitor cell therapy in autoimmune disease

    Immunity

    (2011)
  • J.T. Chen et al.

    Voxel-based analysis of the evolution of magnetization transfer ratio to quantify remyelination and demyelination with histopathological validation in a multiple sclerosis lesion

    NeuroImage

    (2007)
  • B. Chen et al.

    Neuroprotective effect of grafting GDNF gene-modified neural stem cells on cerebral ischemia in rats

    Brain Res.

    (2009)
  • K. Chu et al.

    Human neural stem cells can migrate, differentiate, and integrate after intravenous transplantation in adult rats with transient forebrain ischemia

    Neurosci. Lett.

    (2003)
  • K. Chu et al.

    Human neural stem cell transplantation reduces spontaneous recurrent seizures following pilocarpine-induced status epilepticus in adult rats

    Brain Res.

    (2004)
  • K. Chu et al.

    Human neural stem cells improve sensorimotor deficits in the adult rat brain with experimental focal ischemia

    Brain Res.

    (2004)
  • M.P. Coleman et al.

    Axon pathology in neurological disease: a neglected therapeutic target

    Trends Neurosci.

    (2002)
  • A. Compston et al.

    Multiple sclerosis

    Lancet

    (2002)
  • D.J. Donnelly et al.

    Inflammation and its role in neuroprotection, axonal regeneration and functional recovery after spinal cord injury

    Exp. Neurol.

    (2008)
  • O. Einstein et al.

    Intraventricular transplantation of neural precursor cell spheres attenuates acute experimental allergic encephalomyelitis

    Mol. Cell. Neurosci.

    (2003)
  • O. Einstein et al.

    Transplanted neural precursor cells reduce brain inflammation to attenuate chronic experimental autoimmune encephalomyelitis

    Exp. Neurol.

    (2006)
  • N. Fainstein et al.

    Neural precursor cells inhibit multiple inflammatory signals

    Mol. Cell. Neurosci.

    (2008)
  • I. Fischer

    Candidate cells for transplantation into the injured CNS

    Prog. Brain Res.

    (2000)
  • A. Flugel et al.

    Migratory activity and functional changes of green fluorescent effector cells before and during experimental autoimmune encephalomyelitis

    Immunity

    (2001)
  • S.S. Han et al.

    Grafted lineage-restricted precursors differentiate exclusively into neurons in the adult spinal cord

    Exp. Neurol.

    (2002)
  • V.K. Harris et al.

    Clinical and pathological effects of intrathecal injection of mesenchymal stem cell-derived neural progenitors in an experimental model of multiple sclerosis

    J. Neurol. Sci.

    (2012)
  • C.E. Hill et al.

    Acute transplantation of glial-restricted precursor cells into spinal cord contusion injuries: survival, differentiation, and effects on lesion environment and axonal regeneration

    Exp. Neurol.

    (2004)
  • C.H. Ho et al.

    Spinal cord injury medicine. 1. Epidemiology and classification

    Arch. Phys. Med. Rehabil.

    (2007)
  • A. Hurtado et al.

    Poly (d, l-lactic acid) macroporous guidance scaffolds seeded with Schwann cells genetically modified to secrete a bi-functional neurotrophin implanted in the completely transected adult rat thoracic spinal cord

    Biomaterials

    (2006)
  • S. Jepson et al.

    LINGO-1, a transmembrane signaling protein, inhibits oligodendrocyte differentiation and myelination through intercellular self-interactions

    J. Biol. Chem.

    (2012)
  • Q. Jiang et al.

    MRI detects white matter reorganization after neural progenitor cell treatment of stroke

    NeuroImage

    (2006)
  • K. Jin et al.

    Effect of human neural precursor cell transplantation on endogenous neurogenesis after focal cerebral ischemia in the rat

    Brain Res.

    (2011)
  • K. Kusano et al.

    Transplanted neural progenitor cells expressing mutant NT3 promote myelination and partial hindlimb recovery in the chronic phase after spinal cord injury

    Biochem. Biophys. Res. Commun.

    (2010)
  • M. Abematsu et al.

    Neurons derived from transplanted neural stem cells restore disrupted neuronal circuitry in a mouse model of spinal cord injury

    J. Clin. Invest.

    (2010)
  • M. Aharonowiz et al.

    Neuroprotective effect of transplanted human embryonic stem cell-derived neural precursors in an animal model of multiple sclerosis

    PLoS One

    (2008)
  • N. Amariglio et al.

    Donor-derived brain tumor following neural stem cell transplantation in an ataxia telangiectasia patient

    PLoS Med.

    (2009)
  • R.H. Andres et al.

    The CCR2/CCL2 interaction mediates the transendothelial recruitment of intravascularly delivered neural stem cells to the ischemic brain

    Stroke

    (2011)
  • R.H. Andres et al.

    Human neural stem cells enhance structural plasticity and axonal transport in the ischaemic brain

    Brain

    (2011)
  • M. Bacigaluppi et al.

    Delayed post-ischaemic neuroprotection following systemic neural stem cell transplantation involves multiple mechanisms

    Brain

    (2009)
  • N.I. Bamber et al.

    Neurotrophins BDNF and NT-3 promote axonal re-entry into the distal host spinal cord through Schwann cell-seeded mini-channels

    Eur. J. Neurosci.

    (2001)
  • F. Barkhof et al.

    Remyelinated lesions in multiple sclerosis: magnetic resonance image appearance

    Arch. Neurol.

    (2003)
  • T. Ben-Hur et al.

    Transplanted multipotential neural precursor cells migrate into the inflamed white matter in response to experimental autoimmune encephalomyelitis

    Glia

    (2003)
  • W.F. Blakemore

    Remyelination of CNS axons by Schwann cells transplanted from the sciatic nerve

    Nature

    (1977)
  • W.F. Blakemore et al.

    Extensive oligodendrocyte remyelination following injection of cultured central nervous system cells into demyelinating lesions in adult central nervous system

    Dev. Neurosci.

    (1988)
  • W.F. Blakemore et al.

    Modelling large areas of demyelination in the rat reveals the potential and possible limitations of transplanted glial cells for remyelination in the CNS

    Glia

    (2002)
  • J.F. Bonner et al.

    Promoting directional axon growth from neural progenitors grafted into the injured spinal cord

    J. Neurosci. Res.

    (2010)
  • J.F. Bonner et al.

    Grafted neural progenitors integrate and restore synaptic connectivity across the injured spinal cord

    J. Neurosci.

    (2011)
  • D. Bottai et al.

    Viability-dependent promoting action of adult neural precursors in spinal cord injury

    Mol. Med.

    (2008)
  • C. Buhnemann et al.

    Neuronal differentiation of transplanted embryonic stem cell-derived precursors in stroke lesions of adult rats

    Brain

    (2006)
  • O. Butovsky et al.

    Morphological aspects of spinal cord autoimmune neuroprotection: colocalization of T cells with B7–2 (CD86) and prevention of cyst formation

    FASEB J.

    (2001)
  • Cited by (65)

    • Mechanisms and repair strategies for white matter degeneration in CNS injury and diseases

      2021, Biochimica et Biophysica Acta - Molecular Basis of Disease
      Citation Excerpt :

      There is myriad of cell sources (e.g. NPCs, OPCs, Schwann cells, mesenchymal stem cells, olfactory ensheathing cells etc.) have been employed to study the benefits of cell transplantation for oligodendrocyte replacement and remyelination in various models of WM injuries. These cells are advantageous due to their established role in development and their contribution to endogenous repair, including immune and trophic modulation [496], remyelination and tissue scaffolding [497,498]. Transplanted cells may contribute towards remyelination after WM degeneration directly or indirectly that leads to improved functional recovery as observed in models of SCI and MS.

    • Combination of In Situ Lcn2 pRNA-RNAi Nanotherapeutics and iNSC Transplantation Ameliorates Experimental SCI in Mice

      2020, Molecular Therapy
      Citation Excerpt :

      When distinguishing between F4/80-positive cells showing an M1-like (pro-inflammatory, induced nitric oxide synthase [iNOS]-expressing) polarization or M2-like (anti-inflammatory, cluster of differentiation 206 [CD206]-expressing) polarization, we observed a moderate reduction of the M1-like subset in both transplanted groups (Figure S2E), with a reciprocal increase in the M2-like subset (Figure S2F). Stem cell transplantation has been shown to stimulate neuroprotection through the secretion of neurotrophic factors.6 The quantification of NeuN-expressing mature neurons revealed that the transplantation of either fGFP-iNSCs or fGFP-NSCs was also able to significantly increase the number of surviving neurons in the ventral horns of the spared tissue surrounding the lesion area (Figure 2E).

    • The therapeutic potential of exogenous adult stem cells for the injured central nervous system

      2020, Handbook of Innovations in Central Nervous System Regenerative Medicine
    View all citing articles on Scopus
    1

    These authors contributed equally to this work.

    View full text