Sunteți pe pagina 1din 19

Source Journal of Neurological Disorders and Therapies

Review

Neural Tissue Engineering: Combating Alzheimer’s disease


Luke C. Gordon1,2*

Received Date: 01-05-2018,


Alzheimer’s disease is the most common neurodegenerative disorder, with prevalence accumulating
alarmingly. An effective disease modifying therapy remains elusive, as current clinical treatments
Accepted Date: 22-05-2018
only deliver minimal, symptomatic relief whilst cellular death continues unperturbed. Pathologically,
Published Date: 04-06-2018
Alzheimer’s disease is characterized by extracellular amyloidal proteins and intracellular
Editor: R M Damian Holsinger neurofibrillary tangles. However, the intricacies of the neuropathological mechanisms are not
http://sourcejournals.com/ explicitly defined. The collective absence of understanding regarding disease progression is the
probable reason for the lack of preventative measures or effective treatments. Tissue engineering
solutions show promise in the mitigation of Alzheimer’s disease utilizing a myriad of experimental
techniques that are not centric around the pathophysiology of the disease, but rather the regeneration
of neuronal tissue to improve functionality. As disease mechanisms are still unconfirmed with
oppositional theories still dominating literature, tissue engineering solutions are hugely diverse in
regard to their targets of treatment, modality of delivery and time of intervention. This review will
aim to investigate the current landscape of putative tissue engineering solutions, evaluate the efficacy
of present symptomatic treatments, and propose challenges obstructing future applications specific
to clinical translation in Alzheimer’s disease.
Keywords: Alzheimer’s disease, tissue engineering, neurodegeneration, stem cells, scaffol
Anatomy and Pathophysiology of Alzheimer’s
disease Aβ is a peptide of 40 or 42 amino acids resultant from
systematic cleavage of amyloid precursor protein (APP)
The central nervous system (CNS) comprises of the by β- and γ-secretases. Monomeric amyloid present
brain and spinal cord, which are actively protected by during brain development is considered as
the blood-brain barrier (BBB) from a multitude of neuroprotective due to its quantifiable presence in brain
biomolecules [1]. Neural tissue consists of and cerebral spinal fluid (CSF) samples of healthy
predominantly two cell types: neural and glial cells, each populations, and the alleged supportive role it has in
with their own complex generative processes [2]. neuronal development [6]. Rather, it is hypothesized that
Neurons are the fundamental unit in the brain’s electrical an oligomeric derivate of amyloid is the neurotoxic
circuitry, capable of transmitting sensory and motor form6. Aβ accumulates predominantly in the
signals responsible for the detection of external stimuli hippocampus and entorhinal cortex [7] to form
and generation of movement in muscular entities oligomers and dense core plaques that are collectively
respectively. Neuronal cell bodies and glia are synaptotoxic to neuronal transmissions [8]. These
susceptible to a myriad of neurodegenerative conditions depositions of Aβ constituents are hypothesized to halter
capable of unsettling brain architecture and eliciting action potentials in regions primarily responsible for
subsequent abnormal functionality [3]. memory consolidation [8,9], development [9], learning
[8,9], and deterrence of synaptic plasticity [9,10]. Thus,
Aβ accumulation can be accredited with provoking
The most common neurodegenerative condition is symptomatic hallmarks including short-term memory
Alzheimer’s disease (AD) [4]. Clinically, AD symptoms loss, difficulty with language, disorientation, mood
arise from the loss of cholinergic neurons resulting in swings, loss of motivation, inability to manage self-care
progressive cognitive deterioration [2]. Histologically, and societal withdrawal which are analogous with these
AD is characterized by the deposition of amyloid beta brain centres [9]. These symptoms can accelerate with
(Aβ) plaques and tau-immunoreactive neurofibrillary progression of the disease leaving patients completely
tangles (NFT), which concurrently shape diagnostic dependent [9].
criterion for the continuum that differentiates ‘normal’
aging from AD dementia [5].
Despite amyloid deposition being a well-established
indicator and contributor of AD within literature (i.e.
1
Department of Physiology F13, University of Sydney, “The amyloid cascade hypothesis” [11]), NFT
Sydney, NSW 2006, Australia aggregation has been shown to more tightly dictate the
2
Biomedical Engineering J13, School of Aerospace, degree of dementia impairment in comparison to Aβ
Mechanical and Mechatronic Engineering, The University of [12-14]. NFTs are comprised of hyper-phosphorylated
Sydney, NSW 2006, Australia, E-mail:
lgor3244@uni.sydney.edu.au

© 2018 Gordon LC.


This open access article is distributed under a Creative Commons Attribution (CC-BY) 3.0 license. S J Neurol Disord & Ther, 2018, 1-4 | 1
© 2018 Gordon LC et al. licensee Source Journals. This is an open access article is properly cited and
distributed under the terms and conditions of creative commons attribution license which permits unrestricted use,
distribution and reproduction in any medium.

forms of the microtubule-associated tau protein. Tau resultant from cardiac arrest has even been demonstrated as
protein is abundant in CNS neurons as it is primarily a novel biomarker to predict neurological outcome in
required to modulate stability of axonal microtubules patients [36], the accuracy of which outperforms clinical
and thus is considered functionally innate. Deregulation information currently collected (age, time to return of
of modulatory protein kinases and phosphatases leads to spontaneous circulation and bystander cardiopulmonary
hyperphosphorylation of tau which aggressively resuscitation) and serum neuron-specific enolase [37], a
aggregate to form intermediate bodies of paired helical novel biomarker currently recommended in multiple
filaments (PHFs) approximately 10nm in diameter [5]. treatment guidelines.
PHFs will further pair to construct an intracellular
helical tridimensional conformation. The resultant
aggregate will displace the nucleus towards the The correlation between CVD and AD-related biomolecule
periphery of the soma distending the neuron and elevations advocates that cardiac dysfunction may play a
eventuating in cellular apoptosis [15]. Once the cell has larger role in AD pathogenesis that previous postulated. For
perished the extraneuronal “ghost” tangle or NFT is this reason, treatments which improve the vascular faction of
formed [5]. NFTs are originally reported in the the neurovascular unit (NVU; the intimate anatomical
entorhinal territory of the brain in the case of AD, and relationship in the brain between neurons, astrocytes,
as the disease progresses will undergo a non-random myocytes, pericytes, extracellular matrix components and
degenerative pathway throughout the cerebral cortex endothelial cells of the BBB) may improve neurological
and brainstem [16]. outcome in AD patients.

To support these two biomolecules as possibly Despite the knowledge regarding potential biomolecules and
independent or additive contributors of AD, transgenic microvascular dysfunction that may potentiate AD, the
mutations in both APP and tau have been found to overall aetiology remains unknown. Despite current
generate dementing illness in animal models [17-19]. emergent treatments primarily aimed at minimising these
The debate presently continues on the primary biomolecular depositions, it should be noted there remains a
biomolecule ensuing AD [20] meaning that great lack of knowledge regarding the roles of these factors within
scepticism should be used when evaluating treatments the pathological processes of AD.
that are based on these pathophysiological profiles.
Current Treatments for Alzheimer’s disease
In addition to these primary biomolecules, microvascular
dysfunction has also been linked to neurodegeneration and
AD. Comorbidity of cardiovascular disease (CVD) and AD Protracted life expectancy in recent decades has yielded a
has been highlighted suggesting an association between significant escalation in the prevalence of AD, the most
these two complex age-associated diseases [21,22]. The link common age-related neurodegenerative disease. The
between these two disease states is suggested to be due to population is aging alarmingly with figures demonstrating
shared risk factors including hypertension [23–25], diabetes that the worldwide fraction of individuals >60 years is
mellitus [26–28], Apolipoprotein E genotype [29,30] and projected to rise from 11.7% in 2013 to 21.1% by 2050,
high cholesterol [31]. placing an onus on researchers to address age-corelated
dementing illness, such as AD [38]. AD is an affliction
affecting approximately 47 million people worldwide with
While these risk factors are primarily responsible for cardiac an estimated global cost of US$818 billion. Current
malady, it has been suggested that the resultant CVD from projections estimate this number of sufferers will be in
these factors can lead to impaired Aβ and NFT clearance in excess of 131 million individuals by 2050 [39]. By the end
the brain due to reduced cerebral blood flow [32,33]. While of 2018 the financial burden will be in excess 1 trillion [39].
a clear causation relationship between CVD and AD is Due to this significantly increasing prevalence rate and
absent, evidence is mounting to suggest CVD may play a associated cost, a viable intervention to mitigate this terminal
dominant role in the generation or propagation of dementia and debilitating disease is critically required. To date, no
and AD. Increased Aβ deposits have been found in the brains clinical treatment exists that has had the ability to
of coronary artery disease subjects (75%) while subjects not reproducibility halter AD pathology.
suffering from CVD-related disease rarely demonstrated
AD-like Aβ deposits (12%) [34]. Hypoxic events due to
cardiac arrest have additionally been shown to generate time- The functional consequences of AD are further exacerbated
dependent elevations in serum Aβ42, with most patients by the brain’s ineptitude to regenerate neuronal tissue
seeing a 7-fold increase following a 10-hour (h) period as following chronic neurodegenerative conditions [40]. This is
compared to 1h post-cardiac arrest. The magnitude of dissimilar to other bodily systems which are able to self-heal
elevation was further shown to correlate with clinical following injury [40]. Thus, a treatment for AD must include
outcome [32]. The effects of hypoxia from cardiac arrest is the capability of stimulating remedial healing. Furthermore,
not confined to Aβ, elevations in tau have also been reported complex inflammatory responses to AD routinely include
24h following cardiac arrest [35]. The serum tau elevation glial scarring, microglial infiltration, astrocyte proliferation
and mobilization of inflammatory cytokines. These

© 2018 Gordon LC.


This open access article is distributed under a Creative Commons Attribution (CC-BY) 3.0 license. S J Neurol Disord & Ther, 2018, 1-4 | 2
© 2018 Gordon LC et al. licensee Source Journals. This is an open access article is properly cited and
distributed under the terms and conditions of creative commons attribution license which permits unrestricted use,
distribution and reproduction in any medium.

collectively affect injury endpoints of the disease and vary defined by a mini-mental state examination score (MMSE)
patient-to-patient [41,42]. of 5 to 17), due to only minute effect sizes within clinical
trials [46]. As behavioral deficits are normally recognized at
later stages of disease progression [47,48], it is hypothesized
This attenuated neuronal regeneration in conjunction with that for the majority of severe AD cases, CI interventions are
glial scarring and an improper inflammatory response limits unable to mitigate already established neurodegeneration.
the ability of CNS neurons to repopulate during AD and Patients using CI interventions have also been reported to
results in cyclical damage. Current interventions are unable experience significant adverse events compared to placebo
to halter this degenerative process leading to admission of in clinical trialling including nausea, vomiting, diarrhea,
AD patients into palliative care with negligible hope of anorexia, headache and abdominal pain occasionally
recovery. This further exacerbates the emotional and resulting in trial withdrawal. Furthermore, patient outcomes
financial toll placed upon patients and family [38]. Current are highly variable and predictions of drug success are
interventions include Cholinesterase Inhibitors (CI), N- impossible prior to treatment, with sub-groups deemed
methyl-D-aspartate antagonists (NMDA-I), combination commonly as ‘non-responders’ [46]. With significant
therapies, antidepressants and anti-psychotics. These variability, null treatment sub-groups, small effect sizes and
methods remain symptomatic for the treatment of AD as they significant adverse events, CI intervention efficacy remains
merely attempt to neutralize neurotransmitter disturbances in question as a viable treatment for AD and efforts are
resultant from the disease, whilst the underlying biochemical continuing to offer a superior alternative.
mechanisms ensue [43].

N-Methyl-D-Aspartate Antagonists
Cholinesterase Inhibitors

A non-competitive, moderate–affinity NMDA receptor


Three CIs are currently marketed (donepezil, rivastigmine, antagonist, memantine is suggested to improve neuronal
galantamine) which combat the loss of acetylcholine (ACh) survival through reduced excitotoxicity. Excessive
neurons of the basal forebrain. This disease hallmark occurs glutamatergic activation, partly through activation of NMDA
relatively early in the progression of AD and results in a receptors, results in neurotoxicity, occasioning cell death
deficiency of enzymatic function expediting memory loss [43,49]. Memantine exerts its therapeutic effects by blocking
and deterioration of cognitive faculties [44]. glutamatergic NMDA receptors, which results in
normalization of the glutamate system that in turn
ameliorates cognitive and memory deficits [49]. Memantine
Ordinarily ACh is released from presynaptic neuronal
has comparatively high tolerability and low adverse event
terminals, into the synaptic cleft and binds to post-synaptic
profiles due to its low affinity and rapid off-rate kinetics.
muscarinic and nicotinic receptors. However,
This aids in the preservation of the physiological function of
Acetylcholinesterases (AChE) present on the post-synaptic
the receptor [49,50]. Unlike other NMDA antagonists, the
neuron are responsible for modulation of ACh levels and
relatively fast off-rate kinetics of memantine prevents its
actively terminate signal transduction of ACh via hydrolysis.
accumulation in the neuronal channel as well as interference
A choline molecule resultant from this process is then
with normal synaptic transmission, which is essential when
recycled into the pre-synaptic neuron and ACh is regenerated
moderating glutamatergic pathways [51].
via combination with acetyl-CoA due to the efforts of
choline acetyltransferase [45].
Memantine is an increasingly effective antagonist during
conditions of high glutamate concentrations or over-
The majority of CIs will bind irreversibly to AChE,
activation of NMDA receptors, whilst relatively inactive in
immobilizing and preventing its binding to post-synaptic
normal neurological conditions [52]. In AD, blocking of
receptors [45]. Some clinically marketed irreversible CIs
NMDA receptors with memantine also mitigates Aβ-induced
(including Trichlorfon) are also available, but this class of
degeneration of cholinergic neurons in the nucleus basalis of
pharmaceutical is associated with increased toxicity risk
Meynert and hippocampal regions in rat [53-55].
[45]. CIs reduce cholinesterase activity responsible for the
Furthermore, Memantine exhibits an ability to minimize
lysing of choline-esters including the fundamental
levels of secreted APP and Aβ in multiple models of
neurotransmitter, ACh. CIs through this mechanism increase
transgenic AD mice [56,57]. Via these two additive
endogenous cholinergic transmission across the synaptic
mechanisms, memantine is suggested to elicit
cleft and optimize neuronal action potentials improving
neuroprotection in AD. Clinical trials have established that
memory retention, cognitive function and activities of daily
memantine can significantly moderate clinical deterioration
living45. Regrettably, CIs don’t halter the relentless
in AD, however like CIs, only provoke small effect sizes on
cholinergic neuronal damage but rather delay cognitive
cognition, mood and behaviour [58-61] without affecting
decline by enhancing efficiency of neurotransmission across
pathophysiology.
synapses of surviving ACh neurons [45]. As a result, CI
treatments have been shown to have tapering effects as AD
pathophysiology ensues [45,46]. Moreover, CI treatment has
been shown to be ineffective in patients with severe AD (as

This journal is © The Source Journal of Neurological Disorders and Therapies 2018 S J Neurol Disord & Ther, 2017, 1-4 | 3
© 2018 Gordon LC et al. licensee Source Journals. This is an open access article is properly cited and
distributed under the terms and conditions of creative commons attribution license which permits unrestricted use,
distribution and reproduction in any medium.

Combined Therapy of NMDA-I and CI using MMSE) were insignificant at 24 weeks and even 52
weeks of treatment failed to establish a dose-dependent
result [70]. Results of this double blind, placebo-controlled
Clinical trial evidence has demonstrated significant benefit trial are indicative of amyloid not being the centric pathology
in cognition and language for moderate to severe AD patients of AD and this is well established in literature [71,72].
following combined treatment of NMDA-I (memantine) and Despite this, phase III trials of aducanumab are underway in
CI (donepezil) compared to memantine alone [43,62-64]. order to determine efficacy of amyloid-modulatory
However, this effect is not reproducible in mild to moderate amelioration of AD.
groups of AD, thus benefit is only existent in the latter stages
of AD and effect sizes are minimal.
In summary, all of the aforementioned current treatments are
merely symptomatic and offer only weak beneficial effects
Alleviation of Psychological and Behavioural Symptoms for cognition or the behavioural and psychological
Commonly Associated with Alzheimer’s Disease symptoms of AD. Early disease modifying interventions are
necessary to ease AD pathogenesis and improve long-term
outcomes for patients. As delay in treatment is associated
Compounding significant memory and cognitive deficits are
with a non reversible pathogenic advancement [69], these
the debilitating non-cognitive neuropsychiatric symptoms of
disease modification treatments need to coincide with
AD. These include (with their respective prevalence in AD
prompt diagnosis and exhibit the capability of slowing or
patients) apathy (72%), psychosis (38%), anxiety and
halting pathophysiology, neither of which are achieved with
depression (59%), dysphoria (40%), aberrant motor function
present-day medications or protocols.
(38%), aggression and belligerence (64%) and hallucinations
(10%) [65]. Whilst anti-dementia drugs such as CIs and
NMDA-Is have been shown to mitigate some behavioural Tissue Engineering Strategies for Alzheimer’s Disease
symptoms [46,66,67], they have tapering efficacy in severe
cases. The adoption of clinically accepted antipsychotics and
antidepressants are then employed as a final effort to rescue Tissue engineering is an emergent technology to aid in the
quality of life during the late stages of disease. fight against AD. Tissue engineering for AD aims to
Antidepressants trialled for AD include serotonin and regenerate neural tissue mitigating further degeneration
noradrenaline reuptake inhibitors (SNRIs), selective rather than merely abetting symptoms. Tissue engineering
serotonin reuptake inhibitors (SSRIs) and tricyclic agents. encompasses a multidisciplinary field, which culminates
SSRIs have been demonstrated as the most effective engineering and medical sciences in the development of
antidepressant to treat comorbid depression in AD patients biological substitutes to reinstate, conserve or improve tissue
[65] and have demonstrated efficacy in decreasing functionality impacted by disease or injury [49]. The human
depressive episodes, behavioural disturbances and brain is arguably the most elaborate organ in regards to
improving activities of daily life. However cognitive decline composition, circuitry and the myriad of largely
has remained unaltered by these treatments [65,68]. undetermined functions. This makes the brain a difficult
Antipsychotic use in dementia is contentious, as this class of target for tissue engineering endeavors. This review will
medication is associated with higher risk of mortality, analyze current tissue engineering methodologies including
pneumonia, cerebrovascular morbidity and accelerated stem cells, scaffolds and growth factors. This review will
cognitive decline as compared to anti-dementia treatments also evaluate pertinent attempts to create neuronal tissue
[65]. models with tissue engineering strategies including brain
microplatforms and ex vivo organotypic cultures. The
culmination of these will be employed in coming years to
Disease Modifying Treatments: Interference of Amyloid improve clinical outcomes for patients with AD. Whilst
β Deposition microvascular dysfunction has been suggested to
demonstrate a role in AD pathophysiology, this review will
not encompass vascular tissue engineering but rather focus
Amyloid-modifying approaches are currently occupying the
on techniques of synthesising and supporting neuronal tissue
majority of clinical trial endeavours for AD. Early trial
only.
results with amyloid depletion drugs are disappointing, with
the majority of patients gaining little to no functional
recovery even when administration precedes symptoms or Stem Cells
within the early stages of the disease. Clinical trials have
even reported damaging effects. AN1792, the first Aβ
vaccination attempt, resulted in 6% of phase 2 clinical trial Cell-based therapies for AD aim to replace or promote
patients exhibiting evidence of meningoencephalitis, a lethal survival of damaged neuronal cells. This is facilitated by the
inflammation of the bran and meninges [69]. Conversely, a transplantation of multiple stem cell modalities including
more recent attempt using aducanumab, a monoclonal mesenchymal stromal cells (MSCs) [73,74], neural stem
antibody for a conformational epitope found on Aβ, has cells [75], embryonic stem cells (ESCs) [76,77] and induced
elicited significant reduction in amyloid deposition, however pluripotent stem cells (iPSCs) [77]. These cells enable the
measures of functional and cognitive recovery (as measured regeneration of damaged host tissue via direct cell

© 2018 Gordon LC.


This open access article is distributed under a Creative Commons Attribution (CC-BY) 3.0 license. S J Neurol Disord & Ther, 2018, 1-4 | 4
© 2018 Gordon LC et al. licensee Source Journals. This is an open access article is properly cited and
distributed under the terms and conditions of creative commons attribution license which permits unrestricted use,
distribution and reproduction in any medium.

replacement or indirect secretion factors stimulating [72]. Unfortunately, these cells are rare in the adult brain,
neuroprotection or neurogenesis [78,79]. Stem cells are require intrusive surgical procedures for harvest and
capable of self-renewal and differentiation into dedicated proliferate only to a limited capacity in current culturing
progenitor cells decided by the environment where they mediums [96,97]. They are located in the lateral walls of the
preside. For AD, stem cells need to differentiate and mature ventricles and the dentate gyrus of the hippocampus [98,99].
into neural stem cells, specifically cholinergic neurons of the NSC treatments for AD have several requirements to ensure
nucleus basalis [80] or hippocampal region [81]. In addition success following transplantation. For example, transplanted
to the replacement of injured neurons, stem cells can stem cells must survive in the adult brain once injected,
stimulate neural precursors, promote structural migrate from a focal injection site to populate the targeted
neuroplasticity, inhibit proinflammatory cytokines, suppress regions, differentiate with high fidelity to produce the
neuronal apoptosis, and generate various growth factors required sub-type of neuron and form suitable synaptic
[72,82]. Presently, stem cell research remains in its infancy interfaces [100]. For AD, as opposed to other
for AD. Identification of the ideal stem cell type and the most neurodegenerative conditions like Parkinson’s disease,
appropriate route of administration for this lesioned tissue is widespread throughout multiple regions
neurodegenerative disease are required before tissue making treatment with NSCs extremely complex. In order to
engineering can offer a ‘cure’. combat this, the hippocampal, entorhinal cortex and basal
forebrain can be targeted, as they are the most profoundly
affected. Blurton-Jones and colleagues found that bilateral
Neural tissue engineering methodology is being trialled transplantation of NSC into the hippocampus of aged 3xTg-
extensively in CNS disorders including stroke [83,84], AD mice (which exhibit both amyloid and tau pathology)
traumatic brain injury (TBI) [85-87], Huntington’s disease was able to rescue learning and memory compared to non-
(HD) [88-90], Parkinson’s disease (PD) [91-94] and treated transgenic animals [101]. The proposed mechanism
Alzheimer’s disease (AD) [94,95]. was a robust enhancement in hippocampal synaptic density
facilitated by brain-derived neurotrophic factor (BDNF).
BDNF is implicated in the differentiation of new neurons and
For most methodologies, the neural stem cell (NSC) is
synapses, vital for learning, memory and higher-order
pivotal to the success of these techniques. NSCs are
thinking [85], and is secreted by NSCs [101]. Thus, Blurton-
multipotent, immature, uncommitted cells that exist
Jones and colleagues suggested that NSC-derived BDNF via
predominantly in the brain during developmental stages and
implantation was able to intensify endogenous synaptic
less prolifically in the adult nervous system [72]. NSCs will
connectivity and has potential to improve memory and
differentiate into neurons, astrocytes and oligodentrocytes,
cognition.
which collectively support the complex circuitry of the CNS

Table 1 - Advantages and limitations of stem cells subtypes for applications in Alzheimer's disease

This journal is © The Source Journal of Neurological Disorders and Therapies 2018 S J Neurol Disord & Ther, 2017, 1-4 | 5
© 2018 Gordon LC et al. licensee Source Journals. This is an open access article is properly cited and
distributed under the terms and conditions of creative commons attribution license which permits unrestricted use,
distribution and reproduction in any medium.

with AD remain unclear (see Table 1 for summary). Despite


NSCs have also been demonstrated to be neuroprotective for this, stem cell injections do have strengthening evidence of
cortical neurons in transgenic P301S mice [102,103], a mouse efficacy signifying its potential in future treatments for AD.
model of AD that displays overt tau pathology and neuronal Animal models continue to make up the majority of evidence
loss [104]. The putative mechanism of action is via to ratify tissue engineering therapies, leaving multiple concerns
neurotrophic up-regulation of glial cell-derived neurotrophic regarding ethics and safety in humans. Until improvements in
factor (GDNF) and activity-dependent neuroprotective protein cell survival and long-term results are observed, extrapolation
(ADNP) in conjunction with NSC differentiation to astrocytes cannot be conducted in humans.
[104]. It is thus suggested that NSC-derived neurotrophic
factors mitigate neurodegeneration in murine models of AD. Scaffolds
Trials of human NSCs and identification of a clinical grade
NSC source are required before clinical trials can proceed Stem cell transplantation demonstrates efficacy for AD animal
[100]. MSCs are multipotent stromal stem cells derived models, however poor cell survival remains a significant
predominantly from bone marrow, adipose tissue and challenge as host immunity cells attempt to eradicate injected
umbilical cord blood [105]. Literature dictates that MSCs have cells [119,120]. Autologous cells, such as iPSCs, whilst they
been able to differentiate into neural cells whilst also reducing do recruit an immune response, are comparably minimal to
Aβ aggregates in vitro and in vivo and could possess allogenic and xenogeneic sources [119]. A sizeable
therapeutic benefit for AD [105,106]. Aβ aggregate clearance immunological barrier remains in the path of clinical trials for
is attributed to MSC-induced microglial cell activation stem cell transplantation, as functional rescue of
[107,108]. Immunologically, microglia are suggested to neurodegenerative conditions will not arise if the majority of
regulate growth factors, which can stimulate neurogenesis and injected cells cannot survive to regenerate. Literature routinely
cytokines that modulate neuroinflammation [88]. This reports <1% cell survival for conventional stem cell
synergistic approach of neurogenesis and neuroprotection transplantations [118,119]. Engraftment is a requirement of
could be beneficial for patients with AD and has been lone stem cell injections to ensure proliferation; otherwise the
demonstrated to increase hippocampal neurogenesis in APP prognosis of cell survival will be dire. An additional challenge
transgenic mice following intracerebral injection [109]. includes spatial organization of injected cells as the cells
become frequently dissociated. To combat these tribulations,
ESCs boast pluripotency, the ability to self-renew indefinitely employment of micro-scaffolding can generate cells in three-
and an aptitude to differentiate into virtually all cell types of dimensional configurations which will provide guidance for
the CNS [110]. Due to their high proliferative abilities, these tissue regeneration, structural support, engraftment and
cells are being used in experiments as inexhaustible sources of ultimately cell survival.
neurons to test candidate drugs in neurodegenerative diseases
[110]. Nistor and colleagues found that human neuronal Recent advancements by Carlson and colleagues demonstrated
progenitor cells could be generated in large quantities and high successful grafting of injected scaffold-supported
purity from ESCs [111]. This progenitor population was shown reprogrammed neuronal networks into hippocampal brain
to elicit neuron-specific markers and could differentiate into slices and mice brain striatum in vivo using an iPSC cell source
various neuronal subtypes including cholinergic and [121]. The scaffold utilized an electrospun 100μm synthetic
dopaminergic cells [111]. However, translation of ESCs for poly (desaminotyrosyl tyrosine ethyl ester carbonate) (pDTEc)
clinical therapies must be conducted with extreme caution; the polymer fibre, which was injected with seeded IPSCs capable
persistence of non-differentiated cells from this cell source can of differentiation into human induced neuronal (iN) cells.
be tumorigenic due to genetic instability or malignant Differentiation of IPSCs into iN cells was completed using
transformation when cultured [112,113]. ectopic expression factors (Brn2, Ascl1, and Myt1l) and single
transcription factors (Ascl1, NeuroD1 or Ngn2) [122-125].
Recently developed iPSC technology involves the Literature describes pDTEc as the leading candidate polymer
reprogramming of adult somatic cells to gain self-renewal and from all tyrosine-derived polycarbonates [126], is supportive
pluripotency capabilities similar to ESCs [114-116]. As iPSCs of pluripotent stem cell cultures when microscaled and is
can be derived autologously and are able to proliferate biocompatible. Results of micro-scaffolding injections were
indefinitely, somatic cells from a patient can be a sufficient cell promising. Neurite outgrowth was shown to improve 3.5-fold
source for transplantation therapy. Furthermore, this in hippocampal brain slices and cell survival had a 38-fold
autologous source can prevent immunological rejection issues improvement during transplantation into mice striatal tissue in
and quashes the ethical issues raised from ESC use [117,118]. vivo using the scaffolds. This was comparative to isolated cells
In recent years, research has utilised iPSCs derived from AD alone [121].
patients to model the neurodegenerative disease and produce
patient-specific pathogenic species, potentially removing the Scaffold fibre diameter was an important consideration as
need for animal and human testing [117]. diameter changes induced profound effects upon cellular
proliferation. Thick scaffolds of 3.23 μm permitted greater
Despite the considerable interest and research using stem cell scaffold permeability to cellular infiltration, whilst the thinner
technology to mitigate AD, knowledge of the ideal stem cell fibre scaffolds (1.25 μm) were comparatively impermeable.
source capable of targeting the multifocal lesions associated This was attributed to the decreased void space between the

© 2018 Gordon LC.


This open access article is distributed under a Creative Commons Attribution (CC-BY) 3.0 license. S J Neurol Disord & Ther, 2018, 1-4 | 6
© 2018 Gordon LC et al. licensee Source Journals. This is an open access article is properly cited and
distributed under the terms and conditions of creative commons attribution license which permits unrestricted use,
distribution and reproduction in any medium.

thicker 3.23 μm scaffold fibres [121]. To improve induction of Current clinical trials are being conducted to evaluate nasal
neuronal cells, the addition of doxycycline to the cellular application of insulin. This delivery method has insulin enter
scaffold stimulated the loss of undifferentiated morphology, the brain directly and induces minimal effects on peripheral
the development of early, bipolar neuronal cells and generated glucose levels, which can lead to insulin desensitization [136].
robust expression of βIII-tubulin and microtubule-associated A randomized, double blinded and placebo-controlled phase II
protein 2 (MAP2). These proteins are almost exclusively trial of 104 patients demonstrated that nasal applications for 4
neuronal cytoskeletal proteins [121,127] demonstrating that months of 20IU and 40IU of insulin improved memory, general
the iPSCs had indeed differentiated into iNs. cognition and the ability to conduct day-to-day tasks, which
were also correlated with important biomarkers of AD
It has been recently found that teratoma formation is a including Aβ42 level and tau/Aβ42 ratios [146]. This is
significant risk associated with injections of isolated reinforced in various other clinical trials using intranasal
pluripotent stem cells [121]. This risk is theoretically reduced delivered anti-diabetic drugs [147,149] and is currently
with the use of scaffolding materials. Colonies of dissociated recruiting for phase III clinical trials [150].
iNs with intercellular distances of <6.4μm reportedly succumb
to motility-induced aggregation, inability to differentiate, BDNF is responsible for the longevity and functionality of the
asymmetrical colonies, subsequent improper proliferation of entorhinal cortical and hippocampal neurons [151,152]. It has
cells [128] and subsequent increased risk of teratoma formation been demonstrated that declining BDNF occurs in early stages
[121]. Micro-scaffolding suggestively reduces teratoma- of AD, via signalling through the TrkB receptor [153]. BDNF
formation propensity via establishing a third substrate itself has incompetent pharmacokinetics for systemic
dimension, constraining migration paths of cells and dictates a administration as it has a brief half-life in the plasma and poor
minimal intercellular distance resultant from the scaffold’s BBB penetrance [153].
large surface area. This combination can improve the
assortment of cells and thus reduce teratoma formation. Various methods are employed to modulate BDNF levels
including transplantation of NSCs, lentivirus gene transfer
The completed scaffold with these configurations, increased [154], attachment of BDNF to a monoclonal antibody capable
excitability and exhibited robust ability to fire action potentials of BBB transportation [155] or nasal administration of a BDNF
throughout the micro-scaffold construct, demonstrated by mimetic [156]. As mentioned above, Blurton-Jones, et al.
patch-clamp electrophysiology, when seeded with iN cells ex exhibited the beneficial effects of NSC implantation, which is
vivo compared to injected cells alone [121]. When injected in resultant from increased BDNF levels. Furthermore, Nagahara
vivo, cell survival was estimated at 7.58% utilizing the injected and colleagues, successfully evaluated neuroprotection from
scaffold-seeded iN cells compared to 0.11% when injected increased BDNF in various animal models [154]. Amyloid-
with dissociated iN cells alone. This demonstrated efficacy had transgenic mice upon BDNF gene delivery were able to reverse
no overt change in inflammatory response and also synaptic loss, partially stabilize abnormal gene expression and
demonstrated some ingrowth of endogenous tissues into the restore learning and memory. It should also be noted that these
scaffolds. This experimentation depicts the beneficial outcomes were all independent of amyloid plaque load. For
implications of scaffolding in neuronal tissue. However, this is aged rats, BDNF infusions mitigated cognitive decline and
novel data with minimal evidence at present. Scaffolding has entorhinal neuronal death, which occurred in control-aged rats.
not reached trial stages but demonstrates exciting potential for Finally, in an aged primate model with bilateral radiofrequency
the treatment of AD with stem cell sources, as currently lesions, BDNF-lentiviral vectors ameliorated age-related
dissociated stem cell methodology cannot generate the cognitive decline and neuronal atrophy, which was
required confluence of cells. demonstrated in sham-control experiments [154]. These
models demonstrated neuroprotective effects on fundamental
Growth Factors circuitry involved in AD, indicating success of therapeutic
BDNF delivery.
Various growth factors are being tested in an attempt to
ameliorate AD pathogenesis. Neurogenesis in adults is dictated Studies have recently demonstrated the pivotal role that NGF
by several physiological and pathological factors resulting plays in aging and the occurrence of age-related abnormalities
from the proliferation of NSCs. Up-regulatory factors of such as AD. NGF has demonstrated regenerative and
neurogenesis include estrogen [129,130], antidepressants neuroprotective effects on cholinergic neurons of the basal
[131,132] and growth factors, most notably are BDNF [133], forebrain in various animal models [157-159], highlighting the
insulin growth factor I (IGF-1) [134-136], incretins [136] and protein as a candidate dementia treatment. Uncertainty remains
nerve growth factor (NGF) [137]. on the role NGF could play in mitigating the pathogenesis of
AD. NGF levels in the serum, CSF, hippocampal and parietal
Type 2 diabetes mellitus (T2DM) has been identified as a risk cortices remain similar in AD and normal-aged patients [160].
factor for AD, suggesting that insulin-signalling impairment Conversely, NGF levels in the plasma and dentate gyrus have
could be an initiator or accelerator to this disease [136-138] and been reported as abnormally high for AD patients [161].
this is reinforced in various epidemiology studies [139-170]. Contrasting evidence is also found in preclinical studies, with
Insulin receptors are expressed on neurons which dictate intracerebroventricular administration of Aβ in mice models
dendritic sprouting, neuronal stem cell activation, cell growth, being inductive of reduced NGF protein expression, which is
repair and neuroprotection via oxidative stress [143-146]. suggested to elicit cognitive impairment [162]. Despite the

This journal is © The Source Journal of Neurological Disorders and Therapies 2018 S J Neurol Disord & Ther, 2017, 1-4 | 7
© 2018 Gordon LC et al. licensee Source Journals. This is an open access article is properly cited and
distributed under the terms and conditions of creative commons attribution license which permits unrestricted use,
distribution and reproduction in any medium.

convoluted data on NGF’s complex role in AD, Ferreira, et al. neurotoxic but also demonstrates the ability for in vitro
conducted an interesting phase I clinical trial in 2015 [159]. Six neurodegenerative disease modelling using tissue-engineering
patients were implanted with encapsulated NGF in the basal methodologies. These techniques mitigate the numerous costly
forebrain for 12 months. Results were encouraging with lower and time-consuming attributes of animal testing which still
levels of Aβ in CSF, however there was evidence of brain remains the benchmark for neurodegenerative studies.
atrophy accompanied by increased NFT and tau levels. A
second phase I clinical trial by Tuszynski and colleagues Ex vivo culturing Models of Alzheimer’s Disease: Past to
investigated 10 patients with early stage AD using both ex vivo the Present
and in vivo NGF gene therapy techniques [163]. NGF resultant
trophic responses were demonstrated including axonal While the majority of evidence described in this review has
sprouting toward local sources of NGF and cholinergic cell been related to in vivo stem cell transplantations due to the
hypertrophy. As trophic responses, like those aforementioned, importance of various physiological processes affecting cell
have been shown to ameliorate cognitive deficits in a range of survival, ex vivo models have also been utilised for the
animal models of neurodegeneration [164-166], phase II purposes of investigating stem cell tissue engineering
clinical trials of NGF gene therapy in AD is underway to strategies. The first reports of cell cultures capable of observing
translate these findings to humans [167]. neuronal cells were in 1907. Harrison published an account
wherein frog ectoderm nerve cells were removed and placed in
Brain Microplatforms a drop of lymph for observation [182]. During this time,
Harrison observed neuronal cell aggregation and the branching
AD pathophysiology is highly complex, with multiple of filaments. It was concluded that this development was an
hypotheses of its synthesis. Testing to better understand AD is example of self-differentiation. He also observed nerve fibres
conducted in animal-based and in vitro models but there are which were capable of growing outwards at approximately 15-
significant limitations. Animal testing exhibits high costs, low- 60μm/h [183]. Upon termination of the filaments, an amoeboid
throughput, intensive labour regimes, and experimental end was created which was observed to extend further fibres.
variations. Conversely, in vitro models currently present These were postulated to be surveying the media around the
simplified systems, which often lead to bias or false results nerve cell.
[168]. Brain microplatforms aim to be biomimetic of the 3D
structure, abundant vasculature, BBB and cerebrospinal fluid In 1911, Shorey took this concept further, creating a culture
(CSF) to better emulate the natural tissue in vitro. These medium that was successful in culturing neuroblasts taken
heterogeneously cellular 3D environments aim to improve the from a Necturus, a genus of salamander. Utilising a medium
current 2D monolayer in vitro cultures by using concave containing water, peptone, NaCl, beef extract and gelatin,
microwells, which can produce large quantities of microscopic Shorey was able to produce nerve fibres from late stage
spherical tissue of unique type, shape and size. Developments neuroblasts taken from the medullary canal [184]. The
recently in microfluidics and microelectromechanical systems constituents of the tissue culture media were found to be
have established an ability to emulate in vivo conditions using important, claiming that the beef extract (a metabolic product
in vitro methodology, which previously could not be replicated of muscle) was required for axonal growth. The complexities
[169]. of culturing nerve cells from undifferentiated stem cells and
neuroblasts continued to be explored further.
Specifically for AD, techniques to imitate the disease using
microplatforms include propagation of Aβ or tau through During this period of the early twentieth century, various
neuronal networks and axonal transfer [170-174], Aβ or tau conditions were deemed necessary to ensure successful
resultant toxicity [175-178] and glial cell function [179,180]. culturing of nerve tissues. The first was rigorous asepsis, as the
These models are additionally viable for drug testing and culture medium routinely contained plasma and embryotic
disease modelling to accelerate understanding of this material, it was an exceptional environment for the
debilitating condition. Park, et al. were able to produce a 3D colonization of bacteria [183]. Another feature previously
culture-based microfluidic chip with neurospheroids and a overlooked, was the mechanical features of the culture. In
constant flow of interstitial fluid [181]. Neurospheroid cells recent years, mechanical features such as the elasticity of
were generated by neural progenitor cells, which were isolated culturing matrices, have been fully appreciated as capable of
from rat embryonic cerebral cortical regions. The cells were altering stem cell lineages [185]. In the 1930s, the mechanical
then suspended and micropipetted into concave wells and features and ultrastructures of the media were likewise found
cultured for 10 days to generate multiple almost identical to be important; for example various authors demonstrated that
neurospheroids. By replicating the pathophysiology of AD free floating tissues prompted the cessation of growth and
with the addition of Aβ to the culturing substrate, the authors subsequent degeneration of cell populations [183,186,187]. In
were able to use various immunofluorescent stains to illustrate 1934, Weiss further investigated the effects of mechanical
the interaction of Aβ with the neural tissues architecture. It was forces on the orientation of nerve processes specifically. Prior
demonstrated that thioflavin S immunoflourescence (specific to this knowledge, electrical currents were suggested to be the
marker for Aβ) was increased, showing successful adherence main orientating factor for nerve processes [188], a finding that
of Aβ to the neurospheres and β-III tubulin expression was has been quoted extensively within literature [189–191].
reduced indicative of a decline of neuronal density within the Simply, by applying a small electrical field (eg 125 mW/mm)
neurospheres. This supports the hypothesis that Aβ is to cultured neurons the speed and orientation of nerve fibres

© 2018 Gordon LC.


This open access article is distributed under a Creative Commons Attribution (CC-BY) 3.0 license. S J Neurol Disord & Ther, 2018, 1-4 | 8
© 2018 Gordon LC et al. licensee Source Journals. This is an open access article is properly cited and
distributed under the terms and conditions of creative commons attribution license which permits unrestricted use,
distribution and reproduction in any medium.

would be modulated. Many conflicting reports have been cholinergic neuronal population from a single source to model
published since with findings disputing [192–194] and AD for tissue engineering practices is far too simplified to be
advocating [189,190,195,196] an electrically induced accurately compared to in vivo models. Despite this,
orientation effect for nerve fibre sprouting. organotypic cultures of the late twentieth century have made
milestones in the mission to close the gap between in vivo and
The mechanical and electrical hypotheses explained above ex vivo models of the brain.
demonstrate the complexities of deriving accurate ex vivo
models of neurological environments. In the early 1940s, Organotypic cultures, which are defined as explanted tissues
Roger Sperry added further perplexing evidence for the that continue to develop in an ex vivo environment, have been
required conditions of neuron populations in vitro. He found developed in order to study the neurodegeneration of
that neurons in culture had sensitivities to chemical gradients cholinergic neurons as in AD. In 1983, Keller, et al. reported
which could ensue regenerative processes. Sperry’s work built the presence of cholinergic neurons and nerve fibres in
upon various reports from other groups that lower vertebrates organotypic cultures derived from the septum and
demonstrated an ability to recover vision after a transection of hippocampus of rat brains [205] which had previously been
the optic nerve [197–199]. Sperry conducted a variety of required to be studied in situ [206,207]. Gähwiler, et al. further
experiments on the retinotectal system of the newt and frog to characterized this model in 1990, demonstrating the first
investigate how the transection of this system could be neuronal culture supported with NGF. He established that NGF
reversed. In a newt, he demonstrated that if the eye was rotated was capable of enhancing the number of AChE-positive
180° and transected the newt, once the optical nerve had neurons and the activities of AChE and choline
regenerated, would behave as though the visual world had been acetyltransferase, enzymes that are responsible for the
respectively revolved [200]. For example, if food was breakdown and synthesis of ACh respectively. NGF is an
presented above the newt, it twisted its head to look down. example of a revolutionary step for neuronal in vitro
Similarly in frogs, Sperry demonstrated that uncrossing the experimentation [208] and has demonstrated reproducibly, the
optic nerves by transecting them and allowing for ipsilateral ability to neuroprotect cholinergic neurons in organotypic
regeneration would manifest left-right visual reversal [201]. brain slice cultures. For example, Humpel and colleagues have
The influences of mechanical guidance upon nerve fibre demonstrated an ability to increase the number of neurons per
regeneration were rejected as Sperry postulated that the nerves cultured brain slice by 10-fold by adding 10ng/ml of NGF to a
would have taken systematic paths through the fibre, which culture of cholinergic neurons of the nucleus basalis of
would have preserved a linear hierarchy between the transected Meynert [209,210].
fibres, however this was not the case. Using silver stained
sections of the optical nerve, he found that the fibres, once At present, many groups have utilized ex vivo organotypic slice
regenerated, were highly disordered describing the scene as a models to study many aspects of AD. Such cultures have been
bird’s nest. As the fibres did not act according to a mechanical used to study AD-related cytochemical changes [211,212],
cue for the regenerative process, he proposed that the optical apoptotic cell death following Aβ-induced neurotoxicity [213]
fibres according to their origins in the retina, followed and tau phosphorylation [214]. Whilst ex vivo models such as
chemical cues when re-establishing retinotectal connections. those described are useful, they remain primarily a tool to
Following the theories of embryology, Sperry hypothesized investigate mechanistic pathways, which must still be further
that throughout neuronal differentiation, retinal ganglion cells ratified in in vivo animal models. A difficult question that still
and tectal cells acquired two categories of cytochemical labels. remains is whether slices derived from postnatal donors,
The first label denotes the neuron type as either retinal ganglion cultured for a matter of months can be used to represent mature
or tectal, whilst the second denotes a positional label denoting adult situations such as those attributable to AD patients? With
the topographical position of the retinal ganglion cell within culturing protocols becoming more advanced with time, the
the retina. In this postulation, known as the chemoaffinity gap is closing between neuronal in vivo and ex vivo models but
theory, he suggested that along the length of the optical appropriate testing prior to clinical trials cannot rely entirely
pathway existed cytochemical gradients which dictated the on ex vivo models. Despite this, ex vivo organotypic slice
suitable brachial connections in the optic tract [202]. models have some advantages over in vivo models at the
present, most notably they have higher throughput and the
With evidence for more than a century that neurons exhibit ability to co-culture various cell types in the brain [210].
sensitivity to chemical, mechanical and electrical
environmental factors, to date there is no ex vivo neuronal As demonstrated by the historical development of neuronal
models for tissue engineering purposes which are capable of cultures, difficulties of neuron survival and the unknown
refabricating analogous in vivo settings. To add a further influences affecting brain cells have clouded the potential of
dimension of complexity, within each of the brain regions simulating accurate in vivo environments. Despite this, as more
specifically affected in AD, the impacted cholinergic neurons is learnt about the intricacies of the physiological environment
are intrinsically different when harvested from different of AD, ex vivo models and the tissue engineering techniques
affected regions (e.g. septal-diagonal band region, nucleus used to create them will exponentially advance. This will
basalis magnocellularis region, striatum, the pontomesechalic hopefully lead to appropriate ex vivo neuronal cultures to
tegmentum region of the brainstem and the spinal cord) of the accelerate research of tissue engineering strategies in the fight
brain in terms of volume, fibre outgrowth, innervation patterns against AD and other neurodegenerative conditions.
and morphology [203,204]. For this reason, creating a

This journal is © The Source Journal of Neurological Disorders and Therapies 2018 S J Neurol Disord & Ther, 2017, 1-4 | 9
© 2018 Gordon LC et al. licensee Source Journals. This is an open access article is properly cited and
distributed under the terms and conditions of creative commons attribution license which permits unrestricted use,
distribution and reproduction in any medium.

Challenges Facing Tissue Engineering Solutions for immunosuppressants. High doses are normally required to
Alzheimer’s Disease elicit increased engraftment and can lead to toxicity as
demonstrated in both human [224,225] and animal models
Each of the mentioned tissue engineering applications [226,227]. Additionally studies show that immunosuppressive
designed for the treatment of AD pose unique challenges. As drugs can further augment various cell populations, their
this is a new therapeutic paradigm, tissue engineering proliferative profiles and ultimately their cell survival [228-
outcomes are difficult to predict especially when translating 231]. Comprehensive analysis of immunological reactions in
animal results to human clinical trials. Predictability is further response to transplantation of candidate cell sources needs to
impaired in AD-specific tissue engineering solutions as they be conducted when profiling and estimating engraftment of
act upon mechanisms which are poorly understood. Common cells. If required, the interaction of stem cell populations with
complications to all tissue engineering methodologies include co-administered immunosuppressants should be additionally
mitigation of immune responses, ethical considerations studied.
surrounding animal and human testing, regulatory obstacles,
cost of development and commercialization. This review An additional complication is fibrotic scar formation. This
highlights below the challenges at the pinnacle of each tissue intermediate stage of the regeneration can lead to a haltering in
engineering strategy and evaluates potential solutions for their the regenerative tissue if cells are exposed to imperfect micro-
future. environments [232]. In the AD brain, glial scars are supposed
to escalate Aβ genesis [233], which further progresses the
Stem Cells: Ensuring Cellular Survival disease state. It is for this reason that competent stem cell
infiltration is required so scarred tissue can be replaced
Stem cell transplantation demonstrates great promise through subsequent to neurodegenerative insult234. Upon replacement,
two mechanisms. First, transplantation can delay the onset of the functional and electrical characteristics should mimic that
symptoms and directly regenerate tissue. Second, stem cell of innate tissue.
grafts can facilitate trophic support for the survival and
propagation of endogenous cells whilst positively modulating In summary, the most pertinent problem with dissociated stem
inflammatory responses [215]. cell solutions in AD is the profound death of injected cells,
leading to minimal levels of cell rescue. While methods do
Post-transplantation cellular death remains a significant barrier exist to alleviate this, including the use of
to stem cell transplantation as animal models routinely immunosuppressants, they can lead to serious consequences
demonstrated poor cell survival rates of <1% for dissociated including toxicity. Improvement of cell survival has been
cells [216,217]. Disruptions of local immune signaling from shown in scaffolding techniques utilized in neural tissue
transplantations reduce cellular survival but can additionally engineering, which could hold validity in future AD tissue
exacerbate disease progression via aggravation of engineering solutions.
inflammatory responses [218,219]. For dissociated stem cell
transplants, anti-inflammatory drugs may provide respite Seeded Scaffolds: Minimizing Tissue Damage
[220], however this can be dampening to important
inflammatory responses. For example, acute inflammation has Seeded scaffolds for neural engineering include the challenges
been shown to elicit hormesis (i.e. tissue is protected in the listed above including; cell survival, engraftment and
short-term by neuroprotective effects) [221] while chronic immunological responses but the introduction of polymeric
inflammation is explicitly damaging. Differentiating this structures elicits new issues. These include the
threshold between acute and chronic responses is difficult. biodegradability, biocompatibility, electro-activity and cell-
Stem cells, namely MSCs, have been shown to be material interactions of the scaffold and ensuring mode of
unpredictable immunomodulators of a range of inflammatory implantation does not damage neural tissue.
cytokines. In a model of TBI in mice, transplanted MSCs were
shown to down-regulate pro-inflammatory cytokines Biodegradability is vital as prolonged exposure to scaffolding
interleukin-6 (IL-6) and anti-inflammatory IL-10 mRNA post- constructs can lead to chronic inflammatory reactions [235].
injection and increase IL-6 mRNA [222]. These non- Materials, like pDTEc have tunable hydrophobicity and
synchronous responses to stem cell transplantation are degradation profiles, which is optimal as they can be tailored
commonly reported [222]. These complex immunological to the treatment regime. By allowing degradation of the
reactions to stem cell transplantation are not fully understood scaffold, regenerated cells can produce their own extracellular
and have greatly varied implications for patients. To mitigate matrix [236].
these problems of unpredictability, withdrawal from allogenic
sources should be considered. Adoption of autologous cellular Biocompatibility is fundamental to the success of the
sources, such as iPSCs, will improve immunological disarray treatment. The scaffold and collapsed constituents should
and improve host response predictability [223]. generate negligible immune reactions to allow healing, post-
treatment. Cells both seeded and innate should ultimately
Engraftment is highly dependent on host-immunity responses migrate through the scaffold and proliferate before laying
to injections, as without cellular attachment, cell survival is down new matrix [236]. Structurally for the brain, soft
improbable. Strategies to increase engraftment and cell scaffolds have been suggested to be mimetic of innate brain
survival predominately involve the use of

© 2018 Gordon LC.


This open access article is distributed under a Creative Commons Attribution (CC-BY) 3.0 license. S J Neurol Disord & Ther, 2018, 1-4 | 10
© 2018 Gordon LC et al. licensee Source Journals. This is an open access article is properly cited and
distributed under the terms and conditions of creative commons attribution license which permits unrestricted use,
distribution and reproduction in any medium.

tissue and this favors the proliferation of neural stem cells CNS using hydrogels”. J Biomed Mater Res Part B Appl
instead of glial cells [236]. Biomater vol 87; no. 1, pp. 251–63.
4 Jeffrey Cummings, Paul S. Aisen, Bruno DuBois, Lutz
A new consideration only now being addressed in neural Frölich, Clifford R. JackJr, et al. (2016) “Drug
engineering is electro-activity of scaffolds. The use of development in Alzheimer’s disease: the path to 2025”.
graphene as a surface scaffold modification is currently being Alzheimers Res Ther vol. 8.
trialed in preclinical studies of rats. This has been shown to 5 Serrano-Pozo A, Frosch MP, Masliah E, Hyman BT
improve electrical interactions whilst actually reducing (2011) “Neuropathological Alterations in Alzheimer
microglial recruitment compared to non-graphene structures Disease”. Cold Spring Harb Perspect Med vol. 1; no. 1.
[237]. 6 Giuffrida ML, Caraci F, Pignataro B, Cataldo S, De
Bona P, et al. (Aug.2009) “Beta-amyloid monomers are
The generation of brain mimetic scaffolds will occupy future neuroprotective”. J Neurosci vol. 29; no. 34, pp. 10582–
endeavors of the technology in attempts to remedy lesions 87.
generated in AD and other neurodegenerative conditions. 7 Reilly JF, Games D, Rydel RE, Freedman S, Schenk D,
et al. (2003) “Amyloid deposition in the hippocampus
Growth Factors: Overcoming the Brains Defences to Elicit and entorhinal cortex: quantitative analysis of a
Trophic Responses transgenic mouse model”. Proc Natl Acad Sci USA vol.
100; no. 8, pp. 4837–42.
A number of growth factors, namely neurotrophins are 8 Reitz C (2012) “Alzheimer’s Disease and the Amyloid
inherently poor at transporting past the BBB. To combat this Cascade Hypothesis: A Critical Review”. Int J
issue, monoclonal antibodies can be developed to move these Alzheimers Dis vol. 2012;
neurotrophins across the BBB via receptor–mediated transport. 9 Carl A Gold, Andrew E Budson (Dec.2008) “Memory
Investigation is also underway to optimize transport through loss in Alzheimer’s disease: implications for
the BBB utilizing a variety of nanoparticles. A study in 2009 development of therapeutics”. Expert Rev Neurother
demonstrated this ability using poly(butyl cyanoacrylate) vol. 8; no. 12, pp. 1879–91.
nanoparticles which were able to act as a carrier following 10 Armstrong RA (Sep. 2014) “A critical analysis of the
intravenous injection238. Future application of growth factors ‘amyloid cascade hypothesis’ ”. Folia Neuropathol vol.
must either use this nanoparticle method or the use of stem cell- 52; no. 3, pp. 211–25.
induced neurotrophins as described above to produce clinical 11 Hardy JA, Higgins GA (Apr. 1992) “Alzheimer’s
effect. disease: the amyloid cascade hypothesis”. Science vol.
256; no. 5054, pp. 184–85.
Conclusion 12 Daniel P. Perlm (2010) “Neuropathology of Alzheimer’s
Disease”. Mt Sinai J Med vol. 77; no. 1, pp. 32–42.
13 Arriagada PV, Growdon JH, Hedley-Whyte ET, Hyman
Without cure or even an adequate disease modifying treatment
BT (1992) “Neurofibrillary tangles but not senile
for AD, literature is generating traction in the field of tissue
plaques parallel duration and severity of Alzheimer’s
engineering. Whilst this body of evidence is its infancy for
disease”. Neurology. vol. 4; no. 3 Pt 1, pp. 631–39.
applications within this highly intricate and undetermined
14 Bierer LM, Hof PR, Purohit DP, Carlin L, Schmeidler J
condition, current preclinical evidence is promising with
and Davis KL, et al. (1995) “Neocortical neurofibrillary
treatments on the precipice of clinical trials. Each tissue
tangles correlate with dementia severity in Alzheimer’s
engineering technology has unique challenges that must be
disease”. Arch Neurol vol. 52; no. 1, pp. 81–8.
overcome, but for each successful treatment, thorough
15 Braak E, Braak H, Mandelkow EM (1994) “A sequence
preclinical testing is required to ensure safety during human
of cytoskeleton changes related to the formation of
translation. The brain’s inability to regenerate healthy
neurofibrillary tangles and neuropil threads”. Acta
functional tissue leaves currently diagnosed AD patients
Neuropathol vol. 87; no. 6, pp. 554–67.
without hope. Tissue engineering aims to create a future in
16 Braak H, Braak E, Strothjohann M (1994) “Abnormally
which impending cognitive decline does not plague the future
phosphorylated tau protein related to the formation of
of AD patients.
neurofibrillary tangles and neuropil threads in the
cerebral cortex of sheep and goat”. Neurosci Lett vol.
References 171; no. 1–2, pp. 1–4.
17 Bales KR, Dodart JC, DeMattos RB, Holtzman DM,
1 Roux F, Couraud PO (2005) “Rat brain endothelial cell Paul SM (2002) “Apolipoprotein E, amyloid, and
lines for the study of blood-brain barrier permeability
and transport functions”. Cell Mol Neurobiol volume 25; Alzheimer disease”. Mol Interv vol. 2; no. 6, pp. 363–
no. 1, pp. 41–58. 75, 339.
2 Willerth SM (2011) “Neural tissue engineering using 18 DeMattos RB, Cirrito JR, Parsadanian M, May PC,
embryonic and induced pluripotent stem cells”. Stem O’Dell MA, et al. (2004) “ApoE and Clusterin
Cell Res Ther vol. 2; no. 2, p. 17. Cooperatively Suppress Aβ Levels and Deposition”.
3 Nisbet DR, Crompton KE, Horne MK, Finkelstein DI, Neuron. vol. 41; no. 2, pp. 193–202.
Forsythe JS (2008) “Neural tissue engineering of the 19 Duff K (2001) “Transgenic mouse models of
Alzheimer’s disease: phenotype and mechanisms of
pathogenesis”. Biochem Soc Symp. no. 67, pp. 195–202.

This journal is © The Source Journal of Neurological Disorders and Therapies 2018 S J Neurol Disord & Ther, 2017, 1-4 | 11
© 2018 Gordon LC et al. licensee Source Journals. This is an open access article is properly cited and
distributed under the terms and conditions of creative commons attribution license which permits unrestricted use,
distribution and reproduction in any medium.

20 Small SA, Duff K (2008) “Linking Abeta and tau in late- cardiac arrest: Results of a pilot study”. Resuscitation
onset Alzheimer’s disease: a dual pathway hypothesis”. vol. 84; no. 3, pp. 351–56.
Neuron. vol. 60; no. 4, pp. 534–42. 36 Mattsson N, Zetterberg H, Nielsen N, Blennow K,
21 Qiu C, Winblad B, Marengoni A, Klarin I, Fastbom J Dankiewicz J, et al. (2017) “Serum tau and neurological
and Fratiglioni L (2006) “Heart failure and risk of outcome in cardiac arrest”. Ann Neurol vol. 82; no. 5,
dementia and Alzheimer disease: a population-based pp. 665–75.
cohort study”. Arch Intern Med. vol. 166; no. 9, pp. 37 Stammet P, Collignon O, Hassager C, Wise MP,
1003–8. Hovdenes J, et al. (2015) “Neuron-Specific Enolase as a
22 Rusanen M, Kivipelto M, Leavalhti E, Lakktikainen T, Predictor of Death or Poor Neurological Outcome After
Tuomilehto J, et al. (2014) “Heart diseases and long- Out-of-Hospital Cardiac Arrest and Targeted
term risk of dementia and Alzheimer’s disease: a Temperature Management at 33°C and 36°C”. J Am
population-based CAIDE study.” J Alzheimers Dis vol. Coll Cardiol vol. 65; no. 19, pp. 2104–14.
42; no. 1, pp. 183–91. 38 Sander M, Oxlund B, Jespersen A, Krasnik A,
23 Thorin E (2015) “Hypertension and Alzheimer Disease: Mortensen EL, et al. (2015) “The challenges of human
Another Brick in the Wall of Awareness”. Hypertension population ageing”. Age Ageing vol. 44; no. 2, pp. 185–
vol. 65; no. 1, pp. 36–8. 7.
24 Nelson L, Gard P, Tabet N (2014) “Hypertension and 39 Prince Martin, Comas-Herrera, Adelina, Knapp, Martin,
inflammation in Alzheimer’s disease: close partners in et al. (2016) “World Alzheimer Report 2016”.
disease development and progression!”. J Alzheimers Alzheimer’s disease International.
Dis vol. 41; no. 2, pp. 331–43. 40 Huebner EA, Strittmatter SM (2009) “Axon
25 Carnevale D, Perrotta M, Lembo G, Trimarco B (2016) Regeneration in the Peripheral and Central Nervous
“Pathophysiological Links Among Hypertension and Systems”. Results Probl Cell Differ vol. 48; pp. 339–51.
Alzheimer’s Disease”. High Blood Press. Cardiovasc 41 Lucas SM, Rothwell NJ, Gibson RM (2006) “The role
Prev. vol. 23; no. 1, pp. 3–7. of inflammation in CNS injury and disease”. Br J
26 Akter K, Lanza EA, Martin SA, Myronyuk N, Rua M Pharmacol vol. 147; no. Suppl 1, pp. S232–S40.
(2011) “Diabetes mellitus and Alzheimer’s disease: 42 Zindler E, Zipp F (2010) “Neuronal injury in chronic
shared pathology and treatment?”. Br J Clin Pharmacol CNS inflammation,” Best Pract Res Clin Anaesthesiol
vol. 71; no. 3, pp. 365–76. vol. 24; no. 4, pp. 551–62.
27 Mario Barbagallo, L. J. Dominguez (2014) “Type 2 43 Konstantina G. Yiannopoulou, Sokratis G.
diabetes mellitus and Alzheimer’s disease.” World J Papageorgiou(2013) “Current and future treatments for
Diabetes. vol. 5; no. 6, pp. 889–93. Alzheimer’s disease”. Ther Adv Neurol Disord vol. 6;
28 Chin-Chou Huang, Chia-Min Chung, Hsin-Bang Leu, no. 1, pp. 19–33.
Ling-Yu Lin, Chun-Chih Chiu, et al. (2014) “Diabetes 44 Cummings JL, Back C (1998) “The cholinergic
Mellitus and the Risk of Alzheimer’s Disease: A hypothesis of neuropsychiatric symptoms in
Nationwide Population-Based Study”. PLoS ONE. vol. Alzheimer’s disease”. Am J Geriatr Psychiatry vol. 6;
9; no. 1. no. 2 Suppl 1, pp. S64–78.
29 Kanekiyo T, Xu H, Bu G (2014) “ApoE and Aβ in 45 Mirjana B Čolović, Danijela Z Krstić, Tamara D
Alzheimer’s Disease: Accidental Encounters or Lazarević-Pašti, Aleksandra M Bondžić, Vesna M Vasić
Partners?”. Neuron vol. 81; no. 4, pp. 740–54. (2013) “Acetylcholinesterase Inhibitors: Pharmacology
30 Kim J, Basak JM, Holtzman DM (2009) “The Role of and Toxicology”. Curr Neuropharmacol vol. 11; no. 3,
Apolipoprotein E in Alzheimer’s Disease”. Neuron vol. pp. 315–35.
63; no. 3, pp. 287–303. 46 Birks J (2006) “Cholinesterase inhibitors for
31 Puglielli L, Tanzi RE, Kovacs DM (2003) “Alzheimer’s Alzheimer’s disease”. in Cochrane Database of
disease: the cholesterol connection”. Nat Neurosci vol. Systematic Reviews.
6; no. 4, pp. 345–51. 47 Michael A. Boss (2000) “Diagnostic approaches to
32 Wang J, Gu BJ, Masters CL, Wang YJ (2017) “A Alzheimer’s disease.” Biochimica et Biophysica Acta
systemic view of Alzheimer disease — insights from (BBA) - Molecular Basis of Disease vol. 1502; no. 1, pp.
amyloid-β metabolism beyond the brain”. Nat Rev 188–200.
Neurol vol. 13; no. 10, pp. 612–23. 48 DeFina PA, Moser RS, Glenn M, Lichtenstein JD, Fellus
33 Henrik Zetterberg, Erik Mortberg, Linan Song, Lei J (2013) “Alzheimer’s Disease Clinical and Research
Chang, Gali K. Provuncher (2011), et al. “Hypoxia due Update for Health Care Practitioners”. Journal of Aging
to cardiac arrest induces a time-dependent increase in Research vol. 2013; p. e207178.
serum amyloid β levels in humans”. PloS One vol. 6; no. 49 Olivares D, Deshpande VK, Shi Y, Lahiri DK, Greig
12, p. e28263. NH, et al. (2012) “N-methyl D-aspartate (NMDA)
34 Sparks DL, Hunsaker JC, Scheff SW, Kryscio RJ, receptor antagonists and memantine treatment for
Henson JL (1990) “Cortical senile plaques in coronary Alzheimer’s disease, vascular dementia and Parkinson’s
artery disease, aging and Alzheimer’s disease”. disease”. Curr Alzheimer Res vol. 9, no. 6, pp. 746–58.
Neurobiol Aging vol. 11; no. 6, pp. 601–07. 50 Lipton SA (2004) “Paradigm shift in NMDA receptor
35 Randall J, Mortberg E, Provuncher GK, Fournier DR, antagonist drug development: molecular mechanism of
Duffy DC, et al. (2013) “Tau proteins in serum predict uncompetitive inhibition by memantine in the treatment
neurological outcome after hypoxic brain injury from

© 2018 Gordon LC.


This open access article is distributed under a Creative Commons Attribution (CC-BY) 3.0 license. S J Neurol Disord & Ther, 2018, 1-4 | 12
© 2018 Gordon LC et al. licensee Source Journals. This is an open access article is properly cited and
distributed under the terms and conditions of creative commons attribution license which permits unrestricted use,
distribution and reproduction in any medium.

of Alzheimer’s disease and other neurologic disorders”. stable donepezil treatment”. Alzheimer Dis Assoc
J Alzheimers Dis vol. 6; no. 6 Suppl, pp. S61–74. Disord vol. 20; no. 4, pp. 263–8.
51 Lipton SA (2005) “The molecular basis of memantine 63 Robert Howard, Rupert McShane, F.R.C.Psych, James
action in Alzheimer’s disease and other neurologic Lindesay, Craig Ritchie, et al. (2012) “Donepezil and
disorders: low-affinity, uncompetitive antagonism.” memantine for moderate-to-severe Alzheimer’s
Curr Alzheimer Res vol. 2; no. 2, pp. 155–165. disease”. N Engl J Med vol. 366; no. 10, pp. 893–903.
52 Parsons CG, Danysz W, Quack G (1999) “Memantine is 64 PN Tariot, Farlow MR, Grossberg GT, Graha, SM,
a clinically well tolerated N-methyl-D-aspartate McDonald S and GergeI I, et al. (2004) “Memantine
(NMDA) receptor antagonist--a review of preclinical treatment in patients with moderate to severe Alzheimer
data”. Neuropharmacology vol. 38; no. 6, pp. 735–67. disease already receiving donepezil: a randomized
53 Nyakas C, Granic I, Halmy LG, Banerjee P, Luiten PG controlled trial”. JAMA vol. 291; no. 3, pp. 317–24.
(2011) “The basal forebrain cholinergic system in aging 65 Zec RF, Burkett NR (2008) “Non-pharmacological and
and dementia. Rescuing cholinergic neurons from pharmacological treatment of the cognitive and
neurotoxic amyloid-β42 with memantine”. Behav Brain behavioral symptoms of Alzheimer disease”.
Res vol. 221; no. 2, pp. 594–603. NeuroRehabilitation vol. 23; no. 5, pp. 425–38.
54 Wenk GL, Danysz W, Mobley SL (1995) “MK-801, 66 Maidment ID, Fox CG, Boustani M, Rodriguez J, Brown
memantine and amantadine show neuroprotective RC, and Katona CL (2008) “Efficacy of memantine on
activity in the nucleus basalis magnocellularis”. Eur J behavioral and psychological symptoms related to
Pharmacol vol. 293; no. 3, pp. 267–70. dementia: a systematic meta-analysis”. Ann
55 Miguel-Hidalgo JJ, Alvarez XA, Cacabelos R, Quack Pharmacother vol. 42; no. 1, pp. 32–8.
G(2002) “Neuroprotection by memantine against 67 Farlow M (2002) “A clinical overview of cholinesterase
neurodegeneration induced by beta-amyloid(1-40)”. inhibitors in Alzheimer’s disease.” Int Psychogeriatr vol.
Brain Res vol. 958; no. 1, pp. 210–21. 14; Suppl 1, pp. 93–126.
56 Ray B, Banerjee PK, Greig NH, Lahiri DK (2010) 68 Lyketsos CG, DelCampo L, Steinberg M, Miles Q,
“Memantine treatment decreases levels of secreted Steele CD, et al. (2003) “Treating depression in
Alzheimer’s amyloid precursor protein (APP) and Alzheimer disease: efficacy and safety of sertraline
amyloid beta (A beta) peptide in the human therapy, and the benefits of depression reduction: the
neuroblastoma cells”. Neurosci. Lett vol. 470, no. 1, pp. DIADS,” Arch Gen Psychiatry vol. 60; no. 7, pp. 737–
1–5. 46.
57 Alley GM, Bailey JA, Chen D, Ray B, Puli LK and 69 Gilman S, Koller M, Black RS, Jenkins L, Griffith SG,
Tanila H (2010), et al. “Memantine lowers amyloid-beta et al. (2005) “Clinical effects of Abeta immunization
peptide levels in neuronal cultures and in APP/PS1 (AN1792) in patients with AD in an interrupted trial”.
transgenic mice”. J Neurosci Res vol. 88, no. 1, pp. 143– Neurology vol. 64; no. 9, pp. 1553–62.
54. 70 Sevigny J, Chiao P, Bussière T, Weinreb PH, Williams
58 Grossberg GT, Pejović V, Miller ML, Graham SM L, et al. (2016) “The antibody aducanumab reduces Aβ
(2009) “Memantine therapy of behavioral symptoms in plaques in Alzheimer’s disease”. Nature vol. 537; no.
community-dwelling patients with moderate to severe 7618, pp. 50–6.
Alzheimer’s disease”. Dement Geriatr Cogn Disord vol. 71 Morris GP, Clark IA, Vissel B (2014) “Inconsistencies
27, no. 2, pp. 164–72. and Controversies Surrounding the Amyloid Hypothesis
59 Agüera-Ortiz LF, Sanchez Ortiz C, Duran Alonso JC, of Alzheimer’s Disease”. Acta Neuropathol Commun
Garcia Lopez MT, Garzon Maldonado F and Gomez vol. 2.
Camello A, et al. Grupo de Investigadores del Estudio 72 Rajalingham Sakthiswary, Azman Ali Raymond (2012)
MEMORY (2010) “[Memantine in the pharmacologic “Stem cell therapy in neurodegenerative diseases.”
treatment of moderately severe to severe Alzheimer’s Neural Regen Res vol. 7; no. 23, pp. 1822–31.
disease in Spain (MEMORY study)]”. Rev Neurol vol. 73 Kode JA, Mukherjee S, Joglekar MV, Hardikar AA
51; no. 9, pp. 525–34. (2009) “Mesenchymal stem cells: immunobiology and
60 Peskind ER, Potkin SG, Pomara N, Ott BR, Graham SM role in immunomodulation and tissue regeneration”.
and Olin JT, et al. (2006) “Memantine treatment in mild Cytotherapy vol. 11; no. 4, pp. 377–91.
to moderate Alzheimer disease: a 24-week randomized, 74 Bang OY, Lee JS, Lee PH, Lee G (2005) “Autologous
controlled trial”. Am J Geriatr Psychiatry vol. 14; no. 8, mesenchymal stem cell transplantation in stroke
pp. 704–15. patients.” Ann. Neurol vol. 57; no. 6, pp. 874–82.
61 van Dyck CH, Tariot PN, Meyers B, Malca Resnick E, 75 Kokaia Z, Martino G, Schwartz M, Lindvall O (2012)
Memantine MEM-MD-01 Study Group (2007) “A 24- “Cross-talk between neural stem cells and immune cells:
week randomized, controlled trial of memantine in the key to better brain repair?”. Nat Neurosci vol. 15; no.
patients with moderate-to-severe Alzheimer disease”. 8, pp. 1078–87.
Alzheimer Dis Assoc Disord, vol. 21, no. 2, pp. 136–43. 76 Lerou PH, Daley GQ (2005) “Therapeutic potential of
62 Feldman HH, Schmitt FA, Olin JT, Memantine MEM- embryonic stem cells”. Blood Rev vol. 19; no. 6, pp.
MD-02 Study Group (2006) “Activities of daily living in 321–31.
moderate-to-severe Alzheimer disease: an analysis of 77 Willerth SM (2011) “Neural tissue engineering using
the treatment effects of memantine in patients receiving embryonic and induced pluripotent stem cells”. Stem
Cell Res Ther vol. 2; no. 2, p. 17.

This journal is © The Source Journal of Neurological Disorders and Therapies 2018 S J Neurol Disord & Ther, 2017, 1-4 | 13
© 2018 Gordon LC et al. licensee Source Journals. This is an open access article is properly cited and
distributed under the terms and conditions of creative commons attribution license which permits unrestricted use,
distribution and reproduction in any medium.

78 Tam RY, Fuehrmann T, Mitrousis N, Shoichet MS Outcome in Parkinsonian Rats”. Stem Cells Transl Med
(2014) “Regenerative Therapies for Central Nervous vol. 4; no. 1, pp. 31–43.
System Diseases: a Biomaterials Approach.” 92 Tony Tung-Yin Lee, Cheng-Fang Tsai, Tsung-Hsun
Neuropsychopharmacology vol. 39; no. 1, pp. 169–88. Hsieh, Jia-Jin Jason Chen, Yu-Chih Wang, et al. (2012)
79 Bliss TM, Andres RH, Steinberg GK (2010) “Ectopic Pregnancy-Derived Human Trophoblastic
“Optimizing the success of cell transplantation therapy Stem Cells Regenerate Dopaminergic Nigrostriatal
for stroke”. Neurobiology of Disease vol. 37; no. 2, pp. Pathway to Treat Parkinsonian Rats”. PLoS One vol. 7;
275–83. no. 12.
80 Cundiff PE, Anderson SA (2011) “Impact of induced 93 Chen LW, Kuang F, Wei LC, Ding YX, Yung KK, et al.
pluripotent stem cells on the study of central nervous (2011) “Potential application of induced pluripotent
system disease. [Review]”. Current Opinion in Genetics stem cells in cell replacement therapy for Parkinson’s
& Development vol. 21; no. 3, pp. 354–61. disease”. CNS Neurol Disord Drug Targets vol. 10; no.
81 Blurton-Jones M, Kitazawa M, Martinez-Coria H, 4, pp. 449–58.
Castello NA, Müller FJ, et al. (2009) “Neural stem cells 94 Wang Q, Matsumoto Y, Shindo T, Miyake K, Shindo A,
improve cognition via BDNF in a transgenic model of et al. (2006) “Neural stem cells transplantation in cortex
Alzheimer disease”. Proc Natl Acad Sci U.S.A. vol. 106; in a mouse model of Alzheimer’s disease”. J Med Invest
no. 32, pp. 13594–99. vol. 53; no. 1–2, pp. 61–9.
82 M.E Abdel-Salam, Omar (2011) “Stem cell therapy for 95 Moghadam FH, Alaie H, Karbalaie K, Tanhaei S, Nasr
Alzheimer’s disease,” CNS Neurol Disord Drug Targets Esfahani MH, et al. (2009) “Transplantation of primed
vol. 10; no. 4, pp. 459–85. or unprimed mouse embryonic stem cell-derived neural
83 Zhang H, Sun F, Wang J, Xie L, Yang C, et al. (2017) precursor cells improves cognitive function in
“Combining Injectable Plasma Scaffold with Alzheimerian rats”. Differentiation vol. 78; no. 2–3, pp.
Mesenchymal Stem/Stromal Cells for Repairing Infarct 59–68.
Cavity after Ischemic Stroke.” Aging Dis vol. 8; no. 2, 96 Mitchell KE, Weiss ML, Mitchell BM, Martin P, Davis
pp. 203–14. D, et al. (2003) “Matrix cells from Wharton’s jelly form
84 Wei L, Fraser JL, Lu ZY, Hu X, Yu SP (2012) neurons and glia”. Stem Cells vol. 21; no. 1, pp. 50–60.
“Transplantation of hypoxia preconditioned bone 97 Sanluis-Verdes A, Sanluis-Verdes N, Manso-Revilla
marrow mesenchymal stem cells enhances angiogenesis MJ, Castro-Castro AM, Pombo-Otero J, et al. (2017)
and neurogenesis after cerebral ischemia in rats”. “Tissue engineering for neurodegenerative diseases
Neurobiol. Dis vol. 46; no. 3, pp. 635–45. using human amniotic membrane and umbilical cord”.
85 Hoane MR, Becerra GD, Shank JE, Tatko L, Pak ES, et Cell & Tissue Banking vol. 18; no. 1, pp. 1–15.
al. (2004) “Transplantation of neuronal and glial 98 Reynolds BA, Weiss S (1992) “Generation of neurons
precursors dramatically improves sensorimotor function and astrocytes from isolated cells of the adult
but not cognitive function in the traumatically injured mammalian central nervous system”. Science vol. 255,
brain,” J. Neurotrauma vol. 21; no. 2, pp. 163–74. no. 5052, pp. 1707–10.
86 Anwarul Hasan, George Deeb, Rahaf Rahal, Khairallah 99 Clarke L, van der Kooy D (2011) “The adult mouse
Atwi, Stefania Mondello, et al. (2017) “Mesenchymal dentate gyrus contains populations of committed
Stem Cells in the Treatment of Traumatic Brain Injury”. progenitor cells that are distinct from subependymal
Front Neurol vol. 8; p. 28. zone neural stem cells”. Stem Cells vol. 29; no. 9, pp.
87 Rolfe A, Sun D (2015) “Stem Cell Therapy in Brain 1448–58.
Trauma: Implications for Repair and Regeneration of 100 Marsh SE, Blurton-Jones M (2017) “Neural stem cell
Injured Brain in Experimental TBI Models”. in Brain therapy for neurodegenerative disorders: The role of
Neurotrauma: Molecular, Neuropsychological, and neurotrophic support”. Neurochemistry International.
Rehabilitation Aspects, F. H. Kobeissy, Ed. Boca Raton 101 Blurton-Jones M, Kitazawa M, Martinez-Coria H,
(FL): CRC Press/Taylor & Francis. Castello NA, Müller FJ, et al. (2009) “Neural stem cells
88 Song Jihwan, Soon-Tae Lee, Wonsuk Kang, Jung-Eun improve cognition via BDNF in a transgenic model of
Park, Kon Chu, et al. (2007) “Human embryonic stem Alzheimer disease”. PNAS vol. 106; no. 32, pp. 13594–
cell-derived neural precursor transplants attenuate 99.
apomorphine-induced rotational behavior in rats with 102 Acheson A, Conover JC, Fandl JP, DeChiara TM,
unilateral quinolinic acid lesions”. Neuroscience Letters Russell M, et al. (1995) “A BDNF autocrine loop in
vol. 423; no. 1, pp. 58–61. adult sensory neurons prevents cell death”. Nature vol.
89 Stephen B. Dunnett, Anne E. Rosser (2007) “Stem cell 374; no. 6521, pp. 450–53.
transplantation for Huntington’s disease,” Experimental 103 Hampton DW, Webber DJ, Bilican B, Goedert M,
Neurology vol. 203; no. 2, pp. 279–92. Spillantini MG, et al. (2010) “Cell-mediated
90 Willian L Fodor (2003) “Tissue engineering and cell neuroprotection in a mouse model of human
based therapies, from the bench to the clinic: The tauopathy”. J. Neurosci vol. 30; no. 30, pp. 9973–83.
potential to replace, repair and regenerate”. Reprod Biol 104 Tincer G, Mashkaryan V, Bhattarai P, Kizil C (2016)
Endocrinol vol. 1; p. 102. “Neural stem/progenitor cells in Alzheimer’s disease”.
91 Müller J, Ossig C, Greiner JF, Hauser S, Fauser M, et al. Yale J Biol Med vol. 89; no. 1, pp. 23–35.
(2015) “Intrastriatal Transplantation of Adult Human 105 Turgeman G (2015) “The therapeutic potential of
Neural Crest-Derived Stem Cells Improves Functional mesenchymal stem cells in Alzheimer’s disease:

© 2018 Gordon LC.


This open access article is distributed under a Creative Commons Attribution (CC-BY) 3.0 license. S J Neurol Disord & Ther, 2018, 1-4 | 14
© 2018 Gordon LC et al. licensee Source Journals. This is an open access article is properly cited and
distributed under the terms and conditions of creative commons attribution license which permits unrestricted use,
distribution and reproduction in any medium.

converging mechanisms”. Neural Regen Res vol. 10; no. 119 Anderson AJ, Haus DL, Hooshmand MJ, Perez H,
5, pp. 698–99. Sontag CJ, et al. (2011) “Achieving stable human stem
106 Yuji S. Takeda, Qiaobing Xu (2015) “Neuronal cell engraftment and survival in the CNS: is the future of
Differentiation of Human Mesenchymal Stem Cells regenerative medicine immunodeficient?”. Regen Med
Using Exosomes Derived from Differentiating Neuronal vol. 6; no. 3, pp. 367–406.
Cells”. PLOS ONE vol. 10; no. 8, p. e0135111. 120 Kriks S, Shim JW, Piao J, Ganat YM, Wakeman DR,
107 Lee JK, Jin HK, Bae JS (2009) “Bone marrow- et al. (2011) “Dopamine neurons derived from human
derived mesenchymal stem cells reduce brain amyloid- ES cells efficiently engraft in animal models of
beta deposition and accelerate the activation of Parkinson’s disease”. Nature vol. 480; no. 7378, pp.
microglia in an acutely induced Alzheimer’s disease 547–51.
mouse model”. Neurosci. Lett vol. 450; no. 2, pp. 136– 121 Aaron L. Carlson, Neal K. Bennett, Nicola L.
41. Francis, Apoorva Halikere Apoorva Halikere, Stephen
108 Lee JK, Schuchman EH, Jin HK, Bae JS (2012) Clarke, et al. (2016) “Generation and transplantation of
“Soluble CCL5 derived from bone marrow-derived reprogrammed human neurons in the brain using 3D
mesenchymal stem cells and activated by amyloid β microtopographic scaffolds”. Nature Communications
ameliorates Alzheimer’s disease in mice by recruiting vol. 7; p. 10862.
bone marrow-induced microglia immune responses”. 122 Eva C. Thoma, Erhard Wischmeyer, Nils Offen,
Stem Cells vol. 30; no. 7, pp. 1544–55. Katja Maurus, Anna-Leena Sirén, et al. (2012) “Ectopic
109 Yan Y, Ma T, Gong K, Ao Q, Zhang X, et al. (2014) expression of neurogenin 2 alone is sufficient to induce
“Adipose-derived mesenchymal stem cell differentiation of embryonic stem cells into mature
transplantation promotes adult neurogenesis in the neurons”. PLoS ONE vol. 7; no. 6, p. e38651.
brains of Alzheimer’s disease mice”. Neural Regen Res 123 Zhang Y, Pak C, Han Y, Ahlenius H, Zhang Z, et al.
vol. 9; no. 8, pp. 798–805. (2013) “Rapid single-step induction of functional
110 Malgrange B, Borgs L, Grobarczyk B, Purnelle A, neurons from human pluripotent stem cells”. Neuron
Ernst P, et al.(2011) “Using human pluripotent stem vol. 78; no. 5, pp. 785–98.
cells to untangle neurodegenerative disease 124 Bourke SL, Kohn J (2003) “Polymers derived from
mechanisms”. Cell. Mol. Life Sci vol. 68; no. 4, pp. 635– the amino acid L-tyrosine: polycarbonates, polyarylates
49. and copolymers with poly(ethylene glycol)”. Adv. Drug
111 Gabriel Nistor, Monica M. Siegenthaler, Stephane N. Deliv. Rev. vol. 55; no. 4, pp. 447–66.
Poirier, Sharyn Rossi, Aleksandra J. Poole, et al. (2011) 125 Carlson AL, Florek CA, Kim JJ, Neubauer T, Moore
“Derivation of high purity neuronal progenitors from JC, et al. (2012) “Microfibrous substrate geometry as a
human embryonic stem cells”. PLoS ONE vol. 6; no. 6, critical trigger for organization, self-renewal, and
p. e20692. differentiation of human embryonic stem cells within
112 Lefort N, Feyeux M, Bas C, Féraud O, Bennaceur- synthetic 3-dimensional microenvironments”. FASEB J.
Griscelli A, et al. (2008) “Human embryonic stem cells vol. 26,;no. 8, pp. 3240–51.
reveal recurrent genomic instability at 20q11.21”. Nat. 126 Hooper KA, Macon ND, Kohn J (1998)
Biotechnol vol. 26, no. 12; pp. 1364–66. “Comparative histological evaluation of new tyrosine-
113 Schwarz SC, Schwarz J (2010) “Translation of stem derived polymers and poly (L-lactic acid) as a function
cell therapy for neurological diseases”. Transl Res vol. of polymer degradation”. J Biomed Mater Res vol. 41;
156; no. 3, pp. 155–60. no. 3, pp. 443–54.
114 Takahashi K, Tanabe K, Ohnuki M, Narita M, 127 Vinores SA, HermanMM, Katsetos CD, May EE,
Ichisaka T, et al. (2007) “Induction of pluripotent stem Frankfurter A (1994) “Neuron-associated class III beta-
cells from adult human fibroblasts by defined factors”. tubulin, tau, and MAP2 in the D-283 Med cell line and
Cell vol. 131; no. 5, pp. 861–72. in primary explants of human medulloblastoma”.
115 Yu J, Vodyanik MA, Smuga-Otto K, Antosiewicz- Histochem J vol. 26; no. 8, pp. 678–85.
Bourget J, Frane JL, et al. (2007) “Induced pluripotent 128 Li L, Wang BH, Wang S, Moalim-Nour L, Mohib K,
stem cell lines derived from human somatic cells”. et al. (2010) “Individual Cell Movement, Asymmetric
Science vol. 318; no. 5858, pp. 1917–20. Colony Expansion, Rho-Associated Kinase, and E-
116 Takahashi K, Yamanaka S (2006) “Induction of Cadherin Impact the Clonogenicity of Human
pluripotent stem cells from mouse embryonic and adult Embryonic Stem Cells”. Biophys J vol. 98; no. 11, pp.
fibroblast cultures by defined factors”. Cell vol. 126; no. 2442–51.
4, pp. 663–76. 129 Brännvall K, Korhonen L, Lindholm D (2002)
117 Yang J, Li S, He XB, Cheng C, Le W (2016) “Estrogen-receptor-dependent regulation of neural stem
“Induced pluripotent stem cells in Alzheimer’s disease: cell proliferation and differentiation”. Mol Cell Neurosci
applications for disease modeling and cell-replacement vol. 21; no. 3, pp. 512–20.
therapy”. Molecular Neurodegeneration. vol. 11, p. 39. 130 Perez-Martin M, Azcoitia, Trejo JL, Sierra A,
118 Bellin M, Marchetto MC, Gage FH, Mummery CL Garcia-Segura LM (2003) “An antagonist of estrogen
(2012) “Induced pluripotent stem cells: the new receptors blocks the induction of adult neurogenesis by
patient?”. Nat. Rev. Mol. Cell Biol vol. 13; no. 11, pp. insulin-like growth factor-I in the dentate gyrus of adult
713–26. female rat”. Eur J Neurosci vol. 18; no. 4, pp. 923–30.

This journal is © The Source Journal of Neurological Disorders and Therapies 2018 S J Neurol Disord & Ther, 2017, 1-4 | 15
© 2018 Gordon LC et al. licensee Source Journals. This is an open access article is properly cited and
distributed under the terms and conditions of creative commons attribution license which permits unrestricted use,
distribution and reproduction in any medium.

131 Malberg JE, Eisch AJ, Nestler EJ, Duman RS (2000) disease: a review of basic research and clinical
“Chronic antidepressant treatment increases evidence”. CNS Drugs vol. 27; no. 7, pp. 505–14.
neurogenesis in adult rat hippocampus”. J Neurosci vol. 147 Craft S, Baker LD, Montine TJ, Minoshima S,
20; no. 24, pp. 9104–10. Watson GS, et al. (2012) “Intranasal insulin therapy for
132 Manev H, Uz T, Smalheiser NR, Manev R (2001) Alzheimer disease and amnestic mild cognitive
“Antidepressants alter cell proliferation in the adult impairment: a pilot clinical trial”. Arch Neurol vol. 69;
brain in vivo and in neural cultures in vitro”. Eur J no. 1, pp. 29–38.
Pharmacol vol. 411; no. 1–2, pp. 67–70. 148 Luchsinger JA, Perez T, Chang H, Mehta P, Steffener
133 Zigova T, Pencea V, Wiegand SJ, Luskin MB (1998) J, et al. (2016) “Metformin in Amnestic Mild Cognitive
“Intraventricular administration of BDNF increases the Impairment: Results of a Pilot Randomized Placebo
number of newly generated neurons in the adult Controlled Clinical Trial”. J Alzheimers Dis vol. 51; no.
olfactory bulb”. Mol Cel Neuroscil vol. 11; no. 4, pp. 2, pp. 501–14.
234–45. 149 Claxton A, Baker LD, Hanson A, Trittschuh EH,
134 Anderson MF, Aberg MA, Nilsson M, Eriksson PS Cholerton, et al. (2015) “Long-acting intranasal insulin
(2002) “Insulin-like growth factor-I and neurogenesis in detemir improves cognition for adults with mild
the adult mammalian brain”. Brain Res Dev Brain Res cognitive impairment or early-stage Alzheimer’s disease
vol. 134; no. 1–2, pp. 115–22. dementia”. J Alzheimers Dis vol. 44; no. 3, pp. 897–906.
135 Trejo JL, Carro E, Torres-Aleman I (2001) 150 N I on Aging (2014) “Study of Nasal Insulin to Fight
“Circulating insulin-like growth factor I mediates Forgetfulness (SNIFF)”. National Institute on Aging 03-
exercise-induced increases in the number of new Feb-2014.
neurons in the adult hippocampus”. J Neurosci vol. 21; 151 Conner JM, Lauterborn JC, Yan Q, Gall CM, Varon
no. 5, pp. 1628–34. S (1997) “Distribution of brain-derived neurotrophic
136 Hölscher C (2014) “Insulin, incretins and other factor (BDNF) protein and mRNA in the normal adult
growth factors as potential novel treatments for rat CNS: evidence for anterograde axonal transport”. J
Alzheimer’s and Parkinson’s diseases”. Biochemical Neurosci vol. 17; no. 7, pp. 2295–13.
Society Transactions vol. 42; no. 2, pp. 593–99. 152 Yan Q, Rosenfeld RD,, Hawkins N, Lopez OT, et al.
137 Tuszynski MH, Yang JH, Barba D, U HS, Bakay RA, (1997) “Expression of brain-derived neurotrophic factor
et al. (2015) “Nerve Growth Factor Gene Therapy: protein in the adult rat central nervous system”.
Activation of Neuronal Responses in Alzheimer Neuroscience vol. 78; no. 2, pp. 431–48.
Disease”. JAMA Neurol vol. 72; no. 10, pp. 1139–47. 153 Tanila H (2017) “The role of BDNF in Alzheimer’s
138 Aviles-Olmos I, Limousin P, Lees A, Foltynie T disease”. Neurobiology of Disease vol. 97; Part B, pp.
(2013) “Parkinson’s disease, insulin resistance and 114–18.
novel agents of neuroprotection”. Brain vol. 136; no. Pt 154 Nagahara AH, Merrill D, Coppola G, Tsukada S,
2, pp. 374–84. Schroeder BE, et al. (2009) “Neuroprotective effects of
139 Strachan MW (2005) “Insulin and cognitive function brain-derived neurotrophic factor in rodent and primate
in humans: experimental data and therapeutic models of Alzheimer’s disease”. Nat Med vol. 15; no. 3,
considerations”. Biochemical Society Transactions vol. pp. 331–337.
33; no. 5, pp. 1037–40. 155 Boado RJ, Zhang Y, Zhang Y, Pardridge WM (2007)
140 Haan MN (2006) “Therapy Insight: type 2 diabetes “Genetic engineering, expression, and activity of a
mellitus and the risk of late-onset Alzheimer’s disease”. fusion protein of a human neurotrophin and a molecular
Nat Clin Pract Neurol vol. 2; no. 3, pp. 159–66. Trojan horse for delivery across the human blood–brain
141 Luchsinger JA, Tang MX, Shea S, Mayeux R (2004) barrier”. Biotechnol. Bioeng vol. 97; no. 6, pp. 1376–86.
“Hyperinsulinemia and risk of Alzheimer disease,” 156 Stephen M. Massa, Tao Yang, Youmei Xie, Jian Shi,
Neurology vol. 63; no. 7, pp. 1187–92. Mehmet Bilgen, et al. (2010) “Small molecule BDNF
142 Ristow M (2004) “Neurodegenerative disorders mimetics activate TrkB signaling and prevent neuronal
associated with diabetes mellitus”. J Mol Med vol. 82; degeneration in rodents”. J Clin Invest vol. 120; no. 5,
no. 8, pp. 510–29. pp. 1774–85.
143 Stockhorst U, de Fries D, Steingrueber HJ, 157 F. Hefti (1986) “Nerve Growth Factor Promotes
ScherbaumWA (2004) “Insulin and the CNS: effects on Survival of Septal Cholinergic Neurons After Injury”. in
food intake, memory, and endocrine parameters and the Alzheimer’s and Parkinson’s Disease, A. Fisher, I.
role of intranasal insulin administration in humans”. Hanin, and C. Lachman, Eds. Springer US. pp. 615–25.
Physiol Behav vol. 83; no. 1, pp. 47–54. 158 Conner JM, Franks KM, Titterness AK, Russell K,
144 Hoyer S, (2004) “Glucose metabolism and insulin Merrill DA, et al. (2009) “NGF is essential for
receptor signal transduction in Alzheimer disease”. Eur hippocampal plasticity and learning,” J Neurosci vol. 29;
J Pharmacol vol. 490; no. 1–3, pp. 115–25. no. 35, pp. 10883–889.
145 van Dam PS, Aleman A (2004) “Insulin-like growth 159 Ferreira D, Westman E, Eyjolfsdottir H, Almqvist P,
factor-I, cognition and brain aging”. Eur J Pharmacol Lind G, et al. (2015) “Brain changes in Alzheimer’s
vol. 490,; no. 1–3, pp. 87–95. disease patients with implanted encapsulated cells
146 Jessica Freiherr, Manfred Hallschmid, William H. releasing nerve growth factor”. J Alzheimers Dis vol. 43;
Frey II, Yvonne F. Brünner, Colin D. Chapman, et al. no. 3, pp. 1059–72.
(2013) “Intranasal insulin as a treatment for Alzheimer’s

© 2018 Gordon LC.


This open access article is distributed under a Creative Commons Attribution (CC-BY) 3.0 license. S J Neurol Disord & Ther, 2018, 1-4 | 16
© 2018 Gordon LC et al. licensee Source Journals. This is an open access article is properly cited and
distributed under the terms and conditions of creative commons attribution license which permits unrestricted use,
distribution and reproduction in any medium.

160 Murase K, Nabeshima T, Robitaille Y, Quirion R, connections along axonal membranes”. Ann Neurol vol.
Ogawa M, et al. (1993) “NGF level of is not decreased 75; no. 1, pp. 88–97.
in the serum, brain-spinal fluid, hippocampus, or parietal 174 Simon Dujardin, Lecolle K, Raphaëlle Caillierez,
cortex of individuals with Alzheimer’s disease”. Séverine Bégard, Nadège Zommer, et al. (2014)
Biochem Biophys Res Commun vol. 193; no. 1, pp. “Neuron-to-neuron wild-type Tau protein transfer
198–203. through a trans-synaptic mechanism: relevance to
161 Faria MC, Gonçalves GS, Rocha NP, Moraes E, sporadic tauopathies”. Acta Neuropathologica
Bicalho MA, et al. (2014) “Increased plasma levels of Communications vol. 2; p. 14.
BDNF and inflammatory markers in Alzheimer’s 175 Poon WW, Blurton-Jones M, Tu CH, Feinberg L,
disease”. J Psychiatr Res vol. 53; pp. 166–72. Chabrier MA, et al. (2011) “β-Amyloid impairs axonal
162 Ji C, Song C, Zuo P (2011) “The mechanism of BDNF retrograde trafficking”. Neurobiology of Aging
memory impairment induced by Aβ chronic vol. 32; no. 5, pp. 821–33.
administration involves imbalance between cytokines 176 Kunze A, Meissner R, Brando S, Renaud P (2011)
and neurotrophins in the rat hippocampus”. Curr “Co-pathological connected primary neurons in a
Alzheimer Res vol. 8; no. 4, pp. 410–20. microfluidic device for Alzheimer studies”.
163 Tuszynski MH, Yang JH, Barba D, U HS, U HS, et Biotechnology and Bioengineering vol. 108; no. 9, pp.
al. (2015) “Nerve Growth Factor Gene Therapy 2241–45.
Activates Neuronal Responses in Alzheimer’s Disease”. 177 Choi YJ, Chae S, Kim JH, Barald KF, Park JY, et al.
JAMA Neurol vol. 72; no. 10, pp. 1139–47. (2013) “Neurotoxic amyloid beta oligomeric assemblies
164 Fischer W, Wictorin K, Björklund A, Williams LR, recreated in microfluidic platform with interstitial level
Varon S, et al. (1987) “Amelioration of cholinergic of slow flow”. Scientific Reports, vol. 3; p. 1921.
neuron atrophy and spatial memory impairment in aged 178 Heinz-Georg Jahnkea, Annett Braesigk, Till GA
rats by nerve growth factor”. Nature vol. 329; no. 6134, Mack, Sarah Pönick, Frank Striggow, et al. (2012)
pp. 65–68. “Impedance spectroscopy based measurement system
165 MH Tuszynski, FH Gage (1995) “Bridging grafts for quantitative and label-free real-time monitoring of
and transient nerve growth factor infusions promote tauopathy in hippocampal slice culture”. Biosensors and
long-term central nervous system neuronal rescue and Bioelectronics vol. 32; no. 1, pp. 250–58.
partial functional recovery”. Proc Natl Acad Sci U.S.A., 179 Bianco F, Tonna N, Lovchik RD, Mastrangelo R,
vol. 92; no. 10, pp. 4621–25. Morini R, et al. (2012) “Overflow Microfluidic
166 Markowska AL, Koliatsos VE, Breckler SJ, Price Networks: Application to the Biochemical Analysis of
DL, Olton DS (1994) “Human nerve growth factor Brain Cell Interactions in Complex Neuroinflammatory
improves spatial memory in aged but not in young rats”. Scenarios”. Anal Chem vol. 84; no. 22, pp. 9833–40.
J Neurosci vol. 14; no. 8, pp. 4815–24. 180 Avaid Hai, Joseph Shappir, Micha E Spira (2010)
167 Tanila H (2017) “Randomized, Controlled Study “In-cell recordings by extracellular microelectrodes”.
Evaluating CERE-110 in Subjects With Mild to Nat Meth vol. 7; no. 3, pp. 200–202.
Moderate Alzheimer’s Disease. 181 Park J, Lee BK, Jeong GS, Hyun JK, Lee CJ, Lee SH
168 YoonYoungYi, JiSooPark, JaehoLim, Sang- (2014) “Three-dimensional brain-on-a-chip with an
HoonLee (2015) “Central Nervous System and its interstitial level of flow and its application as an in vitro
Disease Models on a Chip”. Trends in Biotechnology model of Alzheimer’s disease”. Lab Chip vol. 15; no. 1,
vol. 33; no. 12, pp. 762–76. pp. 141–50.
169 van der Meer, van den Berg A (2012) “Organs-on- 182 Rose G. Harrison, M. J. Greenman, Franklin P. Mall,
chips: breaking the in vitro impasse”. Integr Biol (Camb) C. M. Jackson (1907) “Observations of the living
vol. 4; no. 5, pp. 461–70. developing nerve fiber”. Anat Rec 1: 116–28.
170 Deleglise B, Magnifico S, Duplus E, Vaur P, 183 W. R. Ashby (1937) “Tissue Culture Methods in the
Soubeyre V, et al. (2014) “β-amyloid induces a dying- Study of the Nervous System: A Review”. J Neurol
back process and remote trans-synaptic alterations in a Psychopathol vol. 17; no. 68, pp. 322–27.
microfluidic-based reconstructed neuronal network”. 184 Marian L. Shorey (1911) “A study of the
Acta Neuropathologica Communications vol. 2; p. 145. differentiation of neuroblasts in artificial culture media”.
171 Stoothoff W, Jones PB, Spires-Jones T, Joyner D, J Exp Zool vol. 10; no. 1, pp. 85–93.
Chhabra E, et al. (2009) “Differential effect of three- 185 Engler AJ, Sen S, Sweeney HL, Discher DE (2006)
repeat and four-repeat tau on mitochondrial axonal “Matrix Elasticity Directs Stem Cell Lineage
transport”. Journal of Neurochemistry vol. 111; no. 2, Specification”. Cell vol. 126; no. 4, pp. 677–89.
pp. 417–27. 186 Leo Loeb (1921) “Amœboid Movement, Tissue
172 Hill AJ, Jones NA, Williams CM, Stephens GJ, Formation and Consistency of Protoplasm”. Science vol.
Whalley BJ (2010) “Development of multi-electrode 53; no. 1368, pp. 261–62.
array screening for anticonvulsants in acute rat brain 187 Uhlenhuth E (1915) “The form of the epithelial cells
slices”. Journal of Neuroscience Methods vol. 185; no. in cultures of frog skin, and its relation to the consistency
2, pp. 246–56. of the medium”. J Exp Med vol. 22; no. 1, pp. 76–10.
173 Song HL, Shim S, Kim DH, Won SH, Joo S, et al. 188 Sven Ingvar (1920) “Reaction of cells to the galvanic
(2014) “β-Amyloid is transmitted via neuronal current in tissue cultures”. Proc Soc. Exp Biol Med vol.
17; no. 8, pp. 198–99.

This journal is © The Source Journal of Neurological Disorders and Therapies 2018 S J Neurol Disord & Ther, 2017, 1-4 | 17
© 2018 Gordon LC et al. licensee Source Journals. This is an open access article is properly cited and
distributed under the terms and conditions of creative commons attribution license which permits unrestricted use,
distribution and reproduction in any medium.

189 Sisken BF, Smith SD (1975) “The effects of minute 206 Matthews DA, Nadler JV, Lynch GS, Cotman CW
direct electrical currents on cultured chick embryo (1974) “Development of cholinergic innervation in the
trigeminal ganglia”. J Embryol Exp Morphol vol. 33; no. hippocampal formation of the rat: I. Histochemical
1, pp. 29–41. demonstration of acetylcholinesterase activity”. Dev
190 Gordon Marsh, HW Beams (1946) “In vitro control Biol vol. 36; no. 1, pp. 130–41.
of growing check nerve fibers by applied electric 207 Matthews DA, Nadler JV, Lynch GS, Cotman CW
currents”. J Cell Comp Physiol vol. 27; no. 3, pp. 139– (1974) “Development of cholinergic innervation in the
57. hippocampal formation of the rat: I. Histochemical
191 Jaffe LF, Poo MM (1979) “Neurites grow faster demonstration of acetylcholinesterase activity”. Dev
towards the cathode than the anode in a steady field”. J Biol vol. 36; no. 1, pp. 130–41.
Exp Zool vol. 209; no. 1, pp. 115–28. 208 Levi-Montalcini R, Dal Toso R, della Valle F, Skaper
192 Paul Weiss (1934) “In vitro experiments on the SD, Leon A (1995) “Update of the NGF saga”. J Neurol
factors determining the course of the outgrowing nerve Sci vol. 130; no. 2, pp. 119–27.
fiber”. J Exp Zool vol. 68: 393–448. 209 Weis C, Marksteiner J, Humpel C (2001) “Nerve
193 G Levi (1934) Explantation, besonders die Struktur growth factor and glial cell line-derived neurotrophic
und die biologischen Eigenschaften der in vitro factor restore the cholinergic neuronal phenotype in
gezüchteten Zellen und Gewebe. Julius Springer. organotypic brain slices of the basal nucleus of
194 SC Williams (1936) “A study of the reactions of Meynert”. Neuroscience, vol. 102, no. 1, pp. 129–138.
growing embryonic nerve fibers to the passage of direct 210 Humpel C (2015) “Organotypic brain slice cultures:
electric current through the surrounding medium,” Anat A review”. Neuroscience vol. 305; pp. 86–98.
Rec, vol. 64, no. suppl 3, 56–7. 211 Suh EC, Jung YJ, Kim YA, Park EM, Lee KE (2008)
195 A Karssen and B Sager (1934) “Sur l’influence du “A beta 25-35 induces presynaptic changes in
courant électrique sur la croissance des neuroblastes in organotypic hippocampal slice cultures”.
vitro,” Arch Exp Zellforsch 16: 255–59. Neurotoxicology vol. 29; no. 4, pp. 691–699.
196 G Grosse, G Lindner, P Schneider (1969) “Der 212 Frozza RL, Horn AP, Hoppe JB, Simão F, Gerhardt
Einfluss des elektrischem Feldes auf in vitro kultiviertes D, et al. (2009) “A comparative study of beta-amyloid
Nervengewebe,” Z Mikrosk-Anat Forsch, 80: 260–67. peptides Abeta1-42 and Abeta25-35 toxicity in
197 LS Stone (1944) “Functional Polarization in Retinal organotypic hippocampal slice cultures”. Neurochem.
Development and Its Reestablishment in Regenerating Res vol. 34; no. 2, pp. 295–303.
Retinæ of Rotated Grafted Eyes.,” Proc. Soc. Exp. Biol. 213 Allen YS, Devanathan PH, Owen GP (1995)
Med., 57: 13–4. “Neurotoxicity of beta-amyloid protein: cytochemical
198 LS Stone and NT Ussher (1927) “Return of Vision changes and apoptotic cell death investigated in
and Other Observations in Replanted Amphibian Eyes.,” organotypic cultures”. Clin Exp Pharmacol Physiol vol.
Proc. Soc. Exp. Biol. Med., 25: 213–15. 22; no. 5, pp. 370–71.
199 DN Beers (1929) “Return of Vision and Other 214 Jaya R P Prasanthi, Tyler Larson, Jared Schommer,
Observations in Transplanted Amphibian Eyes”. Proc. Othman Ghribi (2011) “Silencing GADD153/CHOP
Soc. Exp Biol Med vol. 26, no. 6, pp. 477–79. gene expression protects against Alzheimer’s disease-
200 R. W. Sperry (1943) “Effect of 180 degree rotation like pathology induced by 27-hydroxycholesterol in
of the retinal field on visuomotor coordination”. J Exp rabbit hippocampus”. PloS One vol. 6; no. 10, p. e26420.
Zool vol. 92: 263–79.
201 R. W. Sperry (1945) “Restoration of vision after 215 Hofer HR, Tuan RS (2016) “Secreted trophic factors
crossing of optic nerves and after contralateral of mesenchymal stem cells support neurovascular and
transplantation of eye”. J Neurophysiol vol. 8; no. 1, pp. musculoskeletal therapies”. Stem Cell Research &
15–28. Therapy vol. 7; p. 131.
202 Meyer RL (1998) “Roger Sperry and his 216 Kriks S, Shim JW, Piao J, Ganat YM, Wakeman DR,
chemoaffinity hypothesis”. Neuropsychologia vol. 36; et al. “Dopamine neurons derived from human ES cells
no. 10, pp. 957–80. efficiently engraft in animal models of Parkinson’s
203 Deborah J. Clarke, Ola G. Nilsson, Patrik Brundin, disease”. Nature vol. 480; no. 7378, pp. 547–51.
Anders Björklund (1990) “Synaptic connections formed 217 Yamauchi T, Kuroda Y, Morita T, Shichinohe H,
by grafts of different types of cholinergic neurons in the Houkin K, et al. (2015) “Therapeutic effects of human
host hippocampus”. Exp.Neurol vol. 107; no. 1, pp. 11– multilineage-differentiating stress enduring (MUSE)
22. cell transplantation into infarct brain of mice”. PLoS
204 Nilsson OG, Clarke DJ, Brundin P, Björklund A ONE vol. 10,; no. 3, p. e0116009.
(1988) “Comparison of growth and reinnervation 218 Pamela A Carpentier, Theo D Palmer (2009)
properties of cholinergic neurons from different brain “Immune Influence on Adult Neural Stem Cell
regions grafted to the hippocampus”. J Comp Neurol Regulation and Function”. Neuron, vol. 64; no. 1, pp.
vol. 268; no. 2, pp. 204–22. 79–92.
205 Keller F, Rimvall K, Waser PG (1983) “Choline 219 Michael F Azari, Louisa Mathias, Ezgi Ozturk,
acetyltransferase in organotypic cultures of rat septum David S Cram, Richard L Boyd, et al. (2010)
and hippocampus”. Neurosci Lett vol. 42, no. 3, pp. 273– “Mesenchymal Stem Cells for Treatment of CNS
78.

© 2018 Gordon LC.


This open access article is distributed under a Creative Commons Attribution (CC-BY) 3.0 license. S J Neurol Disord & Ther, 2018, 1-4 | 18
© 2018 Gordon LC et al. licensee Source Journals. This is an open access article is properly cited and
distributed under the terms and conditions of creative commons attribution license which permits unrestricted use,
distribution and reproduction in any medium.

Injury”. Curr Neuropharmacol vol. 8, no. 4; pp. 316–23. osteogenic differentiation in bone marrow-derived
mesenchymal stem cells”. J Orthop Sci vol. 12; no. 1,
220 Mashkouri S, Crowley MG, Liska MG, Corey S, pp. 83–88.
Borlongan CV (2010) “Utilizing pharmacotherapy and 230 Bin Wang, Zhifeng Xiao, Bing Chen, Jin Han, Yuan
mesenchymal stem cell therapy to reduce inflammation Gao, et al. (2008) “Nogo-66 promotes the differentiation
following traumatic brain injury”. Neural Regen Res of neural progenitors into astroglial lineage cells through
vol. 11; no. 9, pp. 1379–84. mTOR-STAT3 pathway”. PLoS ONE vol. 3; no. 3, p.
221 Lozano D, Gonzales-Portillo GS, Acosta S, de la e1856.
Pena I, Tajiri N, et al. (2015) “Neuroinflammatory 231 Reekmans K, Praet J, Daans J, Reumers V, Pauwels
responses to traumatic brain injury: etiology, clinical P , et al. (2012) “Current Challenges for the
consequences, and therapeutic opportunities”. Advancement of Neural Stem Cell Biology and
Neuropsychiatr Dis Treat vol. 11; pp. 97–106. Transplantation Research”. Stem Cell Rev and Rep vol.
222 Layla T Galindo, Thais R. M. Filippo, Patricia 8; no. 1, pp. 262–78.
Semedo, Carolina B. Ariza, Caroline M. Moreira, et al. 232 Rolls A, Shechter R, Schwartz M (2009) “The bright
(2011) “Mesenchymal Stem Cell Therapy Modulates the side of the glial scar in CNS repair”. Nat. Rev. Neurosci
Inflammatory Response in Experimental Traumatic vol. 10; no. 3, pp. 235–41.
Brain Injury”. Neurology Research International vol. 233 Nagele RG, Wegiel J, Venkataraman V, Imaki H,
2011; p. e564089. Wang KC, et al. (2004) “Contribution of glial cells to the
223 Lakkis FG, Lechler RI (2013) “Origin and Biology development of amyloid plaques in Alzheimer’s
of the Allogeneic Response”. Cold Spring Harb Perspect disease”. Neurobiology of Aging vol. 25; no. 5, pp. 663–
Med vol. 3; no. 8, p. a014993. 74.
224 de Mattos AM, Olyaei AJ, Bennett WM (2000) 234 Alberto Serrano-Pozo, Teresa Gómez-Isla, John H.
“Nephrotoxicity of immunosuppressive drugs: long- Growdon, Matthew P. Frosch, Bradley T. Hyman (2013)
term consequences and challenges for the future”. Am J “A Phenotypic Change But Not Proliferation Underlies
Kidney Dis vol. 35; no. 2, pp. 333–46. Glial Responses in Alzheimer Disease,” Am J Pathol
225 Wu Q, Marescaux C, Wolff V, Jeung MY, Kessler R, vol. 182; no. 6, pp. 2332–44.
et al. (2010) “Tacrolimus-associated posterior reversible 235 Taraballi F, Corradetti B, Minardi S, Powel S,
encephalopathy syndrome after solid organ Cabrera F, et al. (2016) “Biomimetic collagenous
transplantation”. Eur Neurol vol. 64; no. 3, pp. 169–77. scaffold to tune inflammation by targeting
226 Yamauchi A, Oishi R, Kataoka Y, et al. (2004) macrophages”. J Tissue Eng vol. 7.
“Tacrolimus-induced neurotoxicity and nephrotoxicity 236 Fergal J.O'Brien (2011) “Biomaterials & scaffolds
is ameliorated by administration in the dark phase in for tissue engineering”. Materials Today vol. 14; no. 3,
rats”. Cell. Mol. Neurobiol vol. 24; no. 5, pp. 695–704. pp. 88–95.
227 Kochi S, Takanaga H, Matsuo H, Ohtani H, Naito M, 237 Kun Zhou, Sepideh Motamed, George A. Thouas,
et al. (2000) “Induction of apoptosis in mouse brain Claude C. Bernard, Dan Li, et al. (2016) “Graphene
capillary endothelial cells by cyclosporin A and Functionalized Scaffolds Reduce the Inflammatory
tacrolimus”. Life Sci vol. 66; no. 23, pp. 2255–60. Response and Supports Endogenous Neuroblast
228 Song LH, Pan W, Yu YH, Quarles LD, Zhou HH, et Migration when Implanted in the Adult Brain”. PLOS
al. (2006) “Resveratrol prevents CsA inhibition of ONE vol. 11; no. 3, p. e0151589.
proliferation and osteoblastic differentiation of mouse 238 Kurakhmaeva KB, Djindjikhashvili IA, Petrov VE,
bone marrow-derived mesenchymal stem cells through Balabanyan VU, Voronina T, et al. (2009) “Brain
an ER/NO/cGMP pathway”. Toxicol In Vitro vol. 20; targeting of nerve growth factor using poly(butyl
no. 6, pp. 915–22. cyanoacrylate) nanoparticles”. J Drug Target vol. 17; no.
8, pp. 564–74.
229 Isomoto S, Hattori K, Ohgushi H, Nakajima H,
Tanaka Y, et al. (2007) “Rapamycin as an inhibitor of

sourcejournals.com/submit-manuscript/
Submit your next manuscript to Source Journals and take
full advantage of (or) mail to sjndt@sourcejournals.com
 Convenient online submission Thorough peer review
 No space constraints or colour figure charges
 Immediate publication on acceptance
 Research which is freely available for redistribution

Submit your manuscript at

This journal is © The Source Journal of Neurological Disorders and Therapies 2018 S J Neurol Disord & Ther, 2017, 1-4 | 19

S-ar putea să vă placă și