Background Image
Table of Contents Table of Contents
Previous Page  16 / 48 Next Page
Information
Show Menu
Previous Page 16 / 48 Next Page
Page Background

REVIEW

SA JOURNAL OF DIABETES & VASCULAR DISEASE

14

VOLUME 12 NUMBER 1 • JULY 2015

recombinant proteins injected into cells via polyarginine anchors,

which has coined the nomenclature ‘piPSCs’ – protein-induced

pluripotent stem cells;

16

and small-compound mimicking, which

raises reprogramming efficiency

.17

iPS cells offer the advantage of being: easily and non-invasively

harvested; useful tools for drug development; models for disease

processes

in vitro

; and a source of autologous cells for transplantation

due to a lower risk of immunorejection. Disadvantages include a

propensity towards tumorigenesis and a lack of long-term data on

stability and safety.

18

Peripheral and central injury models

Progress on the utility of cell-based therapies for neuropathic pain

research is dependent upon the application of appropriate experi-

mental animal models of peripheral and central nerve lesions.

There are two broad groups of experimental animal models of

neuropathic pain: those that localise the lesion e.g. dorsal root

ganglion lesion, peripheral nerve lesion, spinal cord lesion, and

dorsal and ventral root lesion; and those that describe the type

of lesion e.g. transection, tumour cell invasion or laser radiation,

cryoneurolysis, crush, stimulation of perineuronal inflammation,

and tight or loose ligature.

19

However, no single animal model

entirely recaptures the full range of neuropathic pain mechanisms.

Inanimalmodelstheassessmentandquantificationofneuropathic

pain by direct evaluation is not feasible. Rather subjectively, most

data obtained using animal models have relied on the use of evoked

pain-related behaviours such as withdrawal responses as surrogate

markers for neuropathic pain.

20

Leading on from this, complexities

regarding assessing neuropathic pain in animal models are further

exemplified by attempts to extrapolate and identify relevant markers

for spontaneous pain. This is particularly problematic for patients

with neuropathic pain but rather difficult to measure in rodents.

Surrogate indicators include: changes in general innate behaviours

such as locomotion, burrowing, digging, excessive grooming

and nesting;

21

and more complicated paradigms using Pavlovian

conditioning methods such as conditioned place preference and

aversion.

22

Notwithstanding these methods, laboratory tools for

objectively assessing neuropathic pain in animal models are available

and utilise two surrogate markers, namely: thermal hyperalgesia

using the acetone test; and mechano-allodynia using the von Frey

test.

19

Evidence on the utility of ES cells for the treatment of

neuropathic pain

ES cells have been used to treat neuropathic pain by regenerating

GABAergic interneurons with restoration of the inhibitory tone in

the dorsal horn of the spinal cord, the lack of which would other-

wise contribute to the hyperexcitability that underlies allodynia

and hyperalgesia. Evidence in support of this used mouse ES

cell-derived MGE cortical inhibitory precursor cells, which were

transplanted into a mouse model of peripheral sciatic nerve injury.

23

Using ES cell-derived MGE cells that expressed GFP under the

control of the Gad1 promoter, it was demonstrated that the ES

cell-derived MGE grafts adapted and thrived in the novel spinal

cord environment and migrated throughout the ipsilateral dorsal

and ventral horns. Within two weeks of transplantation the grafted

cells showed immunocytochemical evidence of differentiation

towards a neuronal phenotype (NeuN+) and likewise demonstrated

immunoreactivity for markers of cortical GABAergic interneurons,

namely GABA, neuropeptide Y, parvalbumin and somatostatin.

Furthermore, the transplants structurally integrated into host spinal

cord circuitry as evidenced by neurite outgrowth, sprouting, path-

finding andsynapse formation with host primary afferent and post-

synaptic neurons.

The grafts targeted and influenced a range of spinal cord

neurons including projection neurons of lamina I that normally

receive nociceptive stimuli. There was a close temporal relationship

between the improvement in mechanical allodynia and integration

of the grafted cells, which indicated the latter was instrumental

for recovery. There was no correlation between the number of

transplanted cells and observed anti-nociceptive effects, which

suggests there may be a threshold above which the number of

grafted cells is of less significance in attaining analgesia. There was

a differential effect observed for ES cell-derived MGE transplants

with efficacy demonstrated specifically for neuropathic pain, which

was not matched in a model of inflammatory pain in response to

formalin-induced tissue injury. This suggests MGE grafts are disease

rather than symptom modifying.

The strength of Braz

et al.

’s study is based on the novel observation

that ES cell-derived MGE grafts restored anti-nociceptive inhibitory

GABAergic neurotransmission by structural integration into host

spinal cord neuronal circuits. This contrasts with previous simplistic

mechanisms of achieving the same by studies that merely focused

on releasing GABA using adenoviral and HSV vectors expressing

the GABA synthesising enzyme GAD65 in neuropathic models of

trigeminal neuralgia

24

and spinal nerve ligation.

25

Leading on from

this, the conclusion of Braz

et al.

’s study has been corroborated by

other groups, which increases its reliability.

26

Braz

et al.

’s study failed to account for the anti-nociceptive

summative effects of other co-existent endogenous inhibitory

pathways, which may have confounded the cause and effect

relationship between the independent and dependent variables. It

was presumed the anti-nociceptive effects observed post ES cell-

derived MGE transplantation was GABA mediated, as evidenced

by normalisation of GAD65 mRNA levels in the peripheral nerve

injury model, which is normally associated with low GAD65 mRNA

levels. However, the inhibitory neurotransmitters glycine and

serotonin, which co-exist in some spinal cord GABAergic neurons,

may have accounted for the inhibition and anti-allodynic effects

recorded.

27

Additionally, evidence suggests that following nerve

injury, activation of microglia results in a BDNF-mediated shift in the

chloride gradient of projection neurons in lamina I and deep in the

dorsal horn, which leads to GABAergic inputs becoming somewhat

paradoxically excitatory and pro-nociceptive.

28,29

The results from

Mackie, De Koninck and Price’s studies do not corroborate the

functional integrative mechanism postulated by Braz

et al.

’s study.

The findings of Braz

et al.

’s study are further limited in their

long-term application; they only provide data on the utility of ES

cell-derived MGE grafts for the treatment of neuropathic pain for

28 days post-transplantation. Accordingly the study was unable

to determine whether: the anti-nociceptive effects observed were

sustained; if tolerance developed; and if the experimental models

showed delayed anti-allodynic effects or developed an analgesic

phenotype characterised by mechanical thresholds greater than the

baseline.

A separate study used a modified retinoic acid protocol to induce

differentiation of mouse ES cells into neural and glial precursors.

These were used for transplantation into a mouse model of

central neuropathic pain induced secondary to excitotoxic spinal