GENERATION AND TRANSPLANTATION NEURONES 3D PDF


À propos / Télécharger Lire le document
Nom original: GENERATION AND TRANSPLANTATION NEURONES 3D.pdf
Titre: Generation and transplantation of reprogrammed human neurons in the brain using 3D microtopographic scaffolds
Auteur: Aaron L. Carlson

Ce fichier au format PDF 1.6 a été généré par Arbortext Advanced Print Publisher 9.0.226/W / Acrobat Distiller 8.1.0 (Windows), et a été envoyé sur Fichier-PDF le 25/03/2016 à 19:07, depuis l'adresse IP 92.102.x.x. La présente page de téléchargement du fichier a été vue 765 fois.
Taille du document: 2.3 Mo (10 pages).
Confidentialité: fichier public





Aperçu du document


ARTICLE
Received 28 Jun 2015 | Accepted 27 Jan 2016 | Published 17 Mar 2016

DOI: 10.1038/ncomms10862

OPEN

Generation and transplantation of reprogrammed
human neurons in the brain using 3D
microtopographic scaffolds
Aaron L. Carlson1,*, Neal K. Bennett1,*, Nicola L. Francis1, Apoorva Halikere2,3, Stephen Clarke4,
Jennifer C. Moore5, Ronald P. Hart4,5, Kenneth Paradiso4, Marius Wernig6, Joachim Kohn7, Zhiping P. Pang2,3
& Prabhas V. Moghe1,8

Cell replacement therapy with human pluripotent stem cell-derived neurons has the potential
to ameliorate neurodegenerative dysfunction and central nervous system injuries, but
reprogrammed neurons are dissociated and spatially disorganized during transplantation,
rendering poor cell survival, functionality and engraftment in vivo. Here, we present the design
of three-dimensional (3D) microtopographic scaffolds, using tunable electrospun
microfibrous polymeric substrates that promote in situ stem cell neuronal reprogramming,
neural network establishment and support neuronal engraftment into the brain.
Scaffold-supported, reprogrammed neuronal networks were successfully grafted into
organotypic hippocampal brain slices, showing an B3.5-fold improvement in neurite
outgrowth and increased action potential firing relative to injected isolated cells.
Transplantation of scaffold-supported neuronal networks into mouse brain striatum improved
survival B38-fold at the injection site relative to injected isolated cells, and allowed delivery
of multiple neuronal subtypes. Thus, 3D microscale biomaterials represent a promising
platform for the transplantation of therapeutic human neurons with broad neuro-regenerative
relevance.

1 Department of Biomedical Engineering, Rutgers University, 599 Taylor Road, Piscataway, New Jersey 08854, USA. 2 Department of Neuroscience and Cell
Biology, Rutgers Robert Wood Johnson Medical School, 89 French Street, New Brunswick, New Jersey 08854, USA. 3 Child Health Institute of New Jersey,
Rutgers Robert Wood Johnson Medical School, 89 French Street, New Brunswick, New Jersey 08854, USA. 4 Department of Cell Biology and Neuroscience,
Rutgers University, 604 Allison Road, Piscataway, New Jersey 08854, USA. 5 Human Genetics Institute of New Jersey, 145 Bevier Road, Piscataway, New
Jersey 08854, USA. 6 Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, California 94305, USA.
7 Department of Chemistry and Chemical Biology, New Jersey Center for Biomaterials, 145 Bevier Road, Piscataway, New Jersey 08854, USA. 8 Department
of Chemical and Biochemical Engineering, Rutgers University, 98 Brett Road, Piscataway, New Jersey 08854, USA. * These authors contributed equally to this
work. Correspondence and requests for materials should be addressed to Z.P.P. (email: pangzh@rwjms.rutgers.edu) or to P.V.M. (email: moghe@rutgers.edu).

NATURE COMMUNICATIONS | 7:10862 | DOI: 10.1038/ncomms10862 | www.nature.com/naturecommunications

1

ARTICLE

NATURE COMMUNICATIONS | DOI: 10.1038/ncomms10862

N

eurodegenerative diseases and traumatic brain injuries
result in a loss of functional neurons in the central
nervous system (CNS) and are responsible for substantial
deterioration in quality of life. Although cell transplantation
therapies have shown some promise towards functional
recovery in animal models, the efficacy of these therapies
has been limited by poor cell survival rates1–3. Human induced
pluripotent stem (iPS) cells have recently emerged as a promising
renewable source of expandable patient-specific cells that can be
used to generate human neurons4–6. These iPS cell-derived
neurons are a potentially invaluable tool as a cell source for the
in vivo treatment of neurodegenerative diseases and traumatic
CNS injury7,8.
While many neuronal differentiation protocols have been
established, a robust protocol was recently advanced for the
accelerated production of human neuronal cells from iPS cells,
called induced neuronal (iN) cells, by the direct conversion of iPS
cells using ectopic expression of sets (Brn2, Ascl1, Myt1l) or single
transcription factors (Ascl1, NeuroD1 or Ngn2) (refs 9–12).
Direct conversion yields improved conversion efficiency and
accelerates maturation relative to standard differentiation
protocols. However, effective approaches to generate these cells
in three-dimensional (3D) configurations and to deliver
these cells therapeutically in vivo have yet to be established.
Cell replacement therapies of matured neurons in the brain have
conventionally been limited to injection of dissociated cells2,13.
An alternative approach using biomaterial scaffolds can provide
structural support to cells during transplantation, which could
improve cell engraftment and survival. In addition, many types of
cells behave differently when cultured in two-dimensional (2D)
versus 3D substrates14,15, leading to the development of 3D
biomaterials that better mimic aspects of the in vivo cellular
microenvironment16. In particular, microscale fibrous substrates
enhance several neural cell behaviours in vitro, including neurite
outgrowth17, neuronal maturation18 and neuronal differentiation.
These results motivate the interfacing of microscale fibrous
scaffolds with human induced neuronal cells for in vitro and
in vivo applications. The concept of using 3D biomaterials to
support transplantation has been used with synthetic hydrogels19,
microparticles20 and natural protein matrices21,22 to transplant
neural progenitor cells into the brain, however, these sorts
of scaffolds are not conducive to extended culture or maturation
of cells in vitro, and yield limited control of the cell
microenvironment once transplanted.
In this study, we advance the concept of designing electrospun
synthetic polymer fibres to support the neuronal reprogramming
of iPS cells within a 3D environment via the ectopic expression
of NeuroD1. We demonstrated that fine-tuning the fibrous
architecture enhances neuronal maturation and functionality
in vitro, and also reduces the population of residual unconverted
cells. Finally, we investigated the role of micron-scale fibrous
scaffolds as injectable transplantation vehicles for adherent
networks of iNs, and their ability to enhance the survival
and engraftment of these neurons in murine brain tissue ex vivo
and in vivo.
Results
Induction of neurons via single defined transcription factors.
We initially identified that of the four transcription factors (TFs),
that is, Brn2, Ascl1, Myt1L and NeuroD1, used by Pang et al.12 to
reprogram human fibroblasts to neurons, only Ascl1 and
NeuroD1 could individually induce neuronal conversion of iPS
cells, as also reported elsewhere10. Early neurons induced by
NeuroD1 expression exhibited complex neuronal morphologies
and express mature neuronal markers, so all subsequent studies
2

were done using NeuroD1 as the single TF for generating iN cells.
We transduced human iPS cells with lentiviruses encoding
tetracycline-inducible, tetOn-NeuroD1, rtTA, and, in selected
cases, tetOn-EGFP, and maintained them as undifferentiated iPS
cells in mTeSR medium for multiple passages in the absence of
doxycycline (dox; Supplementary Fig. 1a). We refer to these cells
as ‘RN-iPS’ cells, since these cells were transduced with rtTA and
NeuroD1. RN-iPS cells maintain expression of pluripotency
markers SSEA-4 and Oct-4 over multiple passages in the absence
of dox, indicating that they remain undifferentiated after
transduction (Supplementary Fig. 1a).
To assess neuronal induction from RN-iPS cells, dense cultures
of RN-iPS cells were seeded and dox was added 24 h after plating
(Supplementary Fig. 1b). Early stage iN cells expressed neuronal
marker bIII-tubulin and residual undifferentiated iPS cells
expressed Oct-4 (Supplementary Fig. 1c). Dox addition rapidly
induces the loss of undifferentiated morphology and the
acquisition of bipolar, early neuronal morphologies in a subset
of cells within 48 h (Supplementary Fig. 1c–e). Dox also
rapidly induces EGFP expression in control cells transduced with
tetOn-EGFP. By contrast, cells transduced with only tetOn-EGFP
and rtTA (lacking tetOn-NeuroD1) show strong GFP
fluorescence but maintain undifferentiated hPSC morphology
upon dox addition (Supplementary Fig. 1e).
Despite the rapid, dox-induced neuronal conversion of a subset
of RN-iPS cells, the remaining undifferentiated cells proliferated
and rapidly overtook iN cultures. To address this, cells
were replated 4 days post dox treatment, which established
more uniform human neuronal cultures and eliminated many
undifferentiated cells (Fig. 1a). The replating process may disrupt
the cell–cell contacts necessary for undifferentiated iPS cell
survival. After replating, enriched iN cells could be maintained
for 4 weeks or longer before characterization.
As expected, human iN cells robustly express bIII-tubulin and
microtubule-associated protein 2 (MAP2) by day 12–14 after dox
addition (Fig. 1b). To identify the neuronal subtypes generated by
this procedure, iN cells were replated 4 days after dox addition
onto glial cell monolayers as reported elsewhere23.
Immunocytochemistry on 28 day iNs post replating revealed that
most cells expressed glutamate vesicular transporter VGLUT1,
indicating that these cells are predominantly excitatory
glutamatergic neurons (Fig. 1c). Occasional cells expressing
markers of other neuronal subtypes were also observed,
including inhibitory GABAergic neurons (expressing vesicular
GABA transporter) and dopaminergic neurons (expressing
tyrosine hydroxylase; Fig. 1c). Most cells also robustly expressed
the pre-synaptic protein synaptophysin (Fig. 1d). Finally, patchclamp electrophysiology demonstrated that iN cells are electrically
active and express functional voltage-dependent Na þ channels as
well as voltage-dependent K þ channels as revealed by whole-cell
current recordings (Fig. 1e). Strikingly, these human iN cells were
capable of firing repetitive action potentials (Fig. 1f). Taken
together, these data indicate the robust generation of functional
neuronal cells via NeuroD1 overexpression.
Generation of functional neurons on 3D electrospun fibres.
Next, we investigated whether NeuroD1 expression would similarly induce neuronal conversion and maturation within model
3D electrospun substrates. We constructed fibrous substrates by
electrospinning poly(desaminotyrosyl tyrosine ethyl ester carbonate) (pDTEc), into two architectures, which will be referred to as
‘thin’ and ‘thick’ fibre substrates with average fibre diameters of
1.25±0.05 mm and 3.23±0.06 mm, respectively24 (Fig. 2a–d).
pDTEc is the lead candidate polymer from a combinatorial
library of tyrosine-derived polycarbonates25, as it effectively

NATURE COMMUNICATIONS | 7:10862 | DOI: 10.1038/ncomms10862 | www.nature.com/naturecommunications

ARTICLE

NATURE COMMUNICATIONS | DOI: 10.1038/ncomms10862

a
Dox
Accutase
passage

Accutase passage
to PDL/laminin

d(–1) d0
d4 d5
mTeSR
N2M+dox
+Y

b

d12–28
NBM+dox

βIII-tubulin

MAP2

Merge+DAPI

+10 mV
VGLUT1 MAP2 DAPI

VGAT βIII-tubulin DAPI

40 ms

500 pA

e

c

TH MAP2 DAPI
–50 mV

d

–65 mV

30 mV

f

200 ms
+25 pA

Synaptophysin

βIII-tubulin

Merge+DAPI

–15 pA

Figure 1 | Characterization of neuronal conversion and maturation in human induced-neuronal cells. (a) Schematic of long-term iN reprogramming to
functional neurons. (b) After 12 days of conversion, iN cells express bIII-tubulin and MAP2. (c) Further differentiation on glial cells for 28 days allows
specification of several neuronal subtypes, including vesicular GABA transporter-expressing GABAergic neurons and tyrosine hydroxylase-expressing
dopaminergic neurons, in addition to the predominantly glutamate vescular transporter-expressing glutamatergic neurons. (d) Day 28 iN cells also
extensively express mature neuronal makers such as the synaptic vesicle protein synaptophysin. (e) Whole-cell current recordings demonstrate that
iN cells after 14 days culture in the absence of glia are predominantly electrically active with functional voltage-dependent Na þ channels, as well as
voltage-dependent K þ channels, and (f) fire repetitive induced-action potentials (n ¼ 27/27). Scale bar, 50 mm. Inset: Scale bar, 10 mm.

supports pluripotent stem cell culture when fabricated into
microscale fibrous substrates24, and is biocompatible26. Similarly,
our results with 3D polymeric substrates compared with 2D
polymeric substrates suggest that the fibrous architecture governs
the longer term cellular behaviours observed in contrast to the
polymer composition that plays a role on early interfacial
phenomena. The thick fibre scaffolds are volumetrically
permeable to cellular infiltration, whereas the thin fibre
scaffolds are relatively impermeable, due to decreased void
space between fibres. Without additional material modifications,
it is difficult to produce equal fibre sizes with variable porosity, as
both properties are simultaneously modulated when altering
electrospinning parameters. We hypothesize that cell permeable,
thick fibre substrates will support improved iN maturation and
functionality by promoting enhanced 3D organization and cell–
cell contacts relative to less permeable, thin fibre substrates and
2D controls.
Human iN cells were generated within 3D constructs by
treating RN-iPS cells, cultured on 2D tissue-culture plates, with
dox for 4 days, followed by replating onto 3D electrospun
substrates or 2D controls, according to the time course schematic
shown in Fig. 2e. Human iN cells on 3D electrospun fibres
showed complex morphology with extensive neurite outgrowth
and expressed bIII-tubulin, MAP2 and synaptophysin after 12
days of reprogramming, similarly to 2D cultures (Fig. 2f,g). In
addition, electrophysiological recordings revealed iN cells in
electrospun substrates fired action potentials, demonstrating the
derivation of functional iN cells (Fig. 2h,i).
Effect of 3D scaffold architecture on human iN conversion.
Next, we examined whether the fibre architecture could be tuned

to enhance human iN maturation, as we and others have shown
that geometric cues can influence both human iPS cell and neural
cell behaviours24,27. RN-iPS cells were treated with dox for 4 days,
then replated onto 3D fibrous substrates or 2D controls, including
2D polymer-coated controls, for an additional 8 days of culture.
Immunocytochemistry for proliferation marker Ki67 and Oct-4
revealed that significantly more proliferative and pluripotent cells
were retained when iN cells were replated onto 2D substrates
compared with 3D fibrous substrates (Po0.05, one-way
ANOVA; Fig. 3a,c, Supplementary Figs 2,3). This suggests that
the fibrous architectures may be able to selectively reduce the
presence of residual proliferative and pluripotent iPS cells.
Human iN cells in all conditions expressed extensive
bIII-tubulin-positive processes, along with robust MAP2
expression (Fig. 3a, Supplementary Fig. 2). Significantly greater
numbers of human iN cells expressed MAP2 in thick fibre
substrates relative to 2D controls (Po0.0001, one-way ANOVA)
and thin fibre substrates (Po0.05, one-way ANOVA), indicating
accelerated maturation (Fig. 3b). qRT–PCR also revealed
increasing trends in expression of several neuronal genes in 3D
substrates relative to 2D controls, including bIII-tubulin, MAP2,
synapsin 1 and VGLUT1, though not statistically significant
(Supplementary Fig. 4). Most importantly, calcium imaging to
identify the fraction of cells that respond to a field electrical
stimulation indicated that thick fibre substrates yielded a high
degree of activity, namely, 470% electrically active cells by day
12 of culture (Fig. 3d–e), which was significantly greater than
those on the thin fibre substrates (Po0.05, one-way ANOVA).
Inhibition of E-cadherin-dependent cell–cell contacts markedly
reduced neuronal outgrowth in 2D, and decreased activity
measured by calcium imaging for iNs on 2D and 3D thick fibre
substrates but not thin fibre substrates (Supplementary Fig. 5).

NATURE COMMUNICATIONS | 7:10862 | DOI: 10.1038/ncomms10862 | www.nature.com/naturecommunications

3

ARTICLE

NATURE COMMUNICATIONS | DOI: 10.1038/ncomms10862

Thick fibres

c

f

βIII-tubulin

MAP2

DAPI Merge+substrate

βIII-tubulin

Synaptophysin

DAPI Merge+substrate

g

d

i

e

Dox Accutase passage
to substrates
Accutase
passage
d(–1) d0
mTeSR
+Y

d4
N2M+dox

d5

40 ms

500 pA

h

30 mV

b

Thin fibres

–65 mV

a

200 ms

+10 mV

+50 pA

–80 mV

–30 pA

D10–14
NBM+dox

Figure 2 | Characterization of electrospun polymer fibres and validation for support of iN differentiation. (a,b) Scanning electron microscopy and
(c,d) reflectance images of 2D fibrous and 3D electrospun pDTEc fibres, with substantially variable fibre architectures and porosities that respectively do
not allow and allow cellular infiltration. (e) Schematic of RN-iPS cell reprogramming on 3D electrospun fibres. (f,g) RN-iPS reprogramming was carried out
on 3D electrospun fibres, demonstrating that generation of bIII-tubulin and MAP2 positive iN cells on 3D electrospun fibres after 12 days proceeds similarly
to 2D controls shown in Fig. 1. (h) Whole-cell current recordings demonstrate that iN cells cultured for 10 days on 3D electrospun fibres, 14 days total were
predominantly electrically active with functional voltage-dependent Na þ channels, as well as voltage-dependent K þ channels, and (i) fire repetitive
induced action potentials (n ¼ 18/20). Scale bar, 50 mm (c–d,f). Scale bar, 10 mm (g).

This indicates that 3D microfibrous architectures establish
neuronal networks with enhanced cell–cell contacts and influence
both functional and phenotypic maturity of iN cellular networks.
Microscale scaffolds support outgrowth and survival in brain.
Next, transplantable constructs were designed to deliver human
iN cells into the brain for regenerative therapies. The large mats
of electrospun fibres (0.3–2 cm diameter discs) conventionally
fabricated for in vitro studies cannot be easily transplanted into
the CNS. To allow for injection in vivo, 100 mm square ‘microscale
scaffolds’ that could be injected through a 21-gauge needle were
created by downscaling thick fibre electrospun substrates with a
Vibratome. Human iN cells were seeded in suspension onto
microscale scaffolds analogous to our previous studies, which
resulted in efficient population of microscale scaffolds with iN
cells. Cells in microscale scaffolds matured into bIII-tubulin and
MAP2-expressing neuronal cells, similarly to cells on macroscale
fibrous substrates (Supplementary Fig. 6). The average number of
live human iN cells in each scaffold was 83±13 (n ¼ 19;
Supplementary Fig. 7).
The ability of microscale electrospun scaffolds to promote
human iN cell survival and engraftment was first assessed using
an ex vivo model consisting of organotypic hippocampal slice
cultures from NOD-SCID IL2Rgc null mice (Fig. 4a). Human iN
cells on 4–5 microscale scaffolds were injected into hippocampal
slices, alongside equivalent numbers of dissociated cells on
paired slices, and engraftment and functionality was assessed.
Immunocytochemistry revealed that 3 days after transplantation,
injected scaffold-supported iN cells had average neurite lengths of
831±169 mm, which was significantly greater (Po0.0001,
one-way ANOVA) than those of injected dissociated iNs, which
had neurite lengths of 241±42 mm (Fig. 4b–e). Electrophysiological recordings from human iN cells after 3 weeks indicated that
both dissociated and scaffold-supported iN cells fire action
potentials in response to current injection (Fig. 4f,g), with
scaffold-seeded iN cells displaying enhanced excitability in
4

response to injected currents (Fig. 4h). In addition, both modes
of transplanted cells had mature Na þ channel expression
(Fig. 4i). This suggests that microscale scaffolds enhance
engraftment and functionality of transplanted iN cells.
Next, human iN cell survival was assessed after transplantation
of scaffold-supported or dissociated cells into the mouse striatum
in vivo. Three weeks after transplantation (Fig. 5a), immunocytochemistry of a 1.8 mm  1.8 mm field including the injection
site (Fig. 5b–e) revealed an average survival rate of 5.74±3.16%
based on injected scaffold-seeded iN cells, a 38-fold improvement
(Po0.05, one-way ANOVA) compared with an average survival
rate of 0.15±0.15% of 100,000 injected dissociated iN cells
(Fig. 5g). The survival rate of dissociated iN cell controls was
comparable to that reported by Zhang et al.10 When magnitude
of injected cells were matched at B1,000 cells, quantification of
surviving cells yielded an average of 7.58±4.60% of injected
scaffold-seeded iN cells compared with 0±0% out of 1,000
injected dissociated iN cells. The scaffold-seeded iNs maintained
neurite length (35±8 mm) comparable to those of viable,
dissociated iNs (39±15 mm) in the experiment when number of
injected cells was unmatched. No overt difference in
inflammatory response was detected between injection modes,
and some ingrowth of host tissue into scaffolds was
observed (Supplementary Fig. 8). Surviving transplanted iNs
expressed neuronal cell-adhesion molecule CD56, bIII-tubulin
and synaptophysin (Fig. 5f, Supplementary Fig. 9). Post-synaptic
density protein 95 (PSD95, depicted in blue, with downwardpointing arrows) was detected adjacent or co-localized to
transplanted GFP-labelled and synaptophysin-expressing iN
neurite terminals, suggestive of synaptic integration with host
tissue (Fig. 5f; Supplementary Movie 1). Notably, when
microscale scaffolds were used to transplant multiple subtypes
of neurons, we observed retention of neurons of distinct
specificity in close apposition in vivo (Fig. 5i,j). Similar
differences in survival were also seen when glutamatergic and
dopamine iNs (Supplementary Fig. 10) were co-transplanted
either dissociated or on microscaffolds. In all in vivo experiments,

NATURE COMMUNICATIONS | 7:10862 | DOI: 10.1038/ncomms10862 | www.nature.com/naturecommunications

ARTICLE

NATURE COMMUNICATIONS | DOI: 10.1038/ncomms10862

a

b

c

*

d

Thick fibres

Thin fibres

2D control

Before stim During stim

After stim

Percentage of Ki67+ cells

25

NS
.

20
15
10

NS

5

po 2D
ly
m
3D er
3D thi
n
th
ic
k

0

2D

Ki67 MAP2 DAPI

NS

e
1.0
Fraction of electrically active cells

3D substrate
thick fibres

*

80
70
60
50
40
30
20
10
0

po 2D
ly
m
3D er
3D thin
th
ic
k

3D substrate
thin fibres

2D polymer

2D

2D glass

Percentage of MAP2+ cells

***

0.9
0.8
0.7
0.6
0.5

*

0.4
0.3
0.2
0.1
0.0
2D

3D 3D
Thin Thick

Figure 3 | Comparison of neuronal selection and maturation in 2D and 3D
substrates. (a) Human iN populations robustly express MAP2 in 2D
and 3D conditions, while populations of unconverted, proliferative
Ki67-expressing iPS cells persist in iN populations plated in 2D conditions.
Scale bar, 100 mm. (b) Quantification reveals an enhancement of maturation
as assessed by MAP2 expression in 3D electrospun thick fibres relative to
thin fibres, as well as in both 3D fibrous conditions relative to 2D
conditions. n ¼ 3, *Po0.05, ***Po0.001 by one-way ANOVA. (c)
Quantification reveals an enhancement of neuronal selection, as assessed
by Ki67 expression, fewer residual proliferative cells remained in iNs
replated into 3D fibrous conditions relative to 2D conditions. Scale bar,
100 mm. n ¼ 3; *Po0.05, by one-way ANOVA. (d) Heat map images from
calcium recordings of human iN cells before (left), during (middle) and after
field electrical stimulation). Scale bar, 75 mm. (e) Quantification of the
fraction of cells that respond to electrical stimulation with a substantial
increase in fluorescence intensity reveals highly active cell populations
in 2D and 3D thick fibre substrates relative to cells in the thin fibre
substrates. n ¼ 3; *Po0.05 by one-way ANOVA, all error bars presented
as mean±1 s.d.

some human iNs were observed to migrate off scaffolds, and
minimal scaffold degradation was observed in the 1–3 weeks
post-transplantation.
Discussion
Cell therapies for treatment of CNS brain injury and disease are
challenged by poor cell survival, engraftment and retention after
transplantation both into the brain and spinal cord, with cell
survival ofo1% routinely reported28. The objective of this study
was to advance a new integrated biomaterials-based paradigm to
both rapidly reprogram pluripotent stem cells to neurons and
efficiently deliver enriched, organized neuronal networks to the
brain. To achieve this, we engineered human iPS cells with the

neuronal TF, NeuroD1, and demonstrated efficient and rapid
neuronal conversion in 3D fibrous substrates fabricated from
synthetic polymers that were geometrically tuned to accelerate
neuronal maturation and network establishment. Utilizing this
system, we report that 3D microfibrous substrates can guide
many important characteristics of human iN cells derived from
iPS cells that are relevant for in vivo applications, including
(i) in vitro maturation, electrical activity and purification;
(ii) neurite outgrowth, survival and electrical activity after
transplantation into ex vivo brain tissue and (iii) survival and
engraftment after transplantation in vivo into the striatum. These
results highlight the promise of biomaterials-based delivery as a
platform for reprogramming and regenerative cell sourcing in the
CNS and as a potential model that can be further adapted and
evaluated for therapeutic efficacy.
Our primary hypothesis was that microfibrous substrates with
thick fibres and interfibre spacing would provide a 3D microniche
for rapid, high-purity conversion and accelerated maturation of
iPS cell-derived iNs, and that such 3D substrate-supported
neuronal networks would exhibit improved levels of retention
and engraftment following transplantation into the brain. In vitro,
thick fibre substrates enhanced human iNs maturation and
excitability, while selectively excluding residual undifferentiated
iPS cells. Both of these phenomena arise from controlled cell
confinement and 3D juxtacrine signalling organization. While the
enhanced MAP2 expression in iN cells in thick fibre substrates is
consistent with previously reported results18,29–31, the concurrent
increase in excitability indicates active both phenotypic and
functional maturation. We propose that the thick fibre substrates
drive iN confinement24, leading to accelerated maturation due to
increased engagement of neural cell-adhesion molecules
such as L1 or N-cadherin, which have known roles in neural
development32–34 and neuritogenesis29,31. In contrast, the thin
fibre substrates fail to support neuronal infiltration and
aggregation, resulting in the diminished excitability relative to
more 3D, thick fibre substrates.
For effective scaffolds that can form reprogrammed neuronal
networks, a key feature is that the proliferation of undifferentiated
iPS cells should be suppressed. The geometry we have identified
for electrospun scaffolds limits the proportion of undifferentiated
iPS cells. Undifferentiated human ES and iPS cells require
media supplementation with the rho-associated kinase (ROCK)
inhibitor Y-27632 to survive dissociation to single cells35. In the
absence of Y-27632, the degree to which dissociated iPS cells can
re-establish E-cadherin-mediated cell–cell contacts that have been
demonstrated to be required for survival and proliferation36
is severely diminished when seeded onto electrospun fibrous
substrates. Isolated pluripotent stem cells have been observed to
form viable colonies largely based on motility-induced
aggregation rather than single cell clonal expansion when
intercellular distances were less than 6.4 mm (ref. 37). The large
surface area, added substrate dimension and constrained
migration paths of the electrospun fibres effectively increase
intercellular distances, which would limit motility-induced
aggregation and could explain the observed increase in
differentiation of iPS cells and loss of residual proliferative cells
on microfibrous substrates. This reduction in residual
undifferentiated iPS cells could be used to supplement other
methods for eliminating the risk of teratoma formation in vivo
due to the presence of undifferentiated cells, however, additional
studies are required to assess the tumorigenic potential of these
cells once delivered in vivo.
After establishing a substrate geometry that accelerated iN
maturation, the ability of these substrates to promote engraftment
of human iN cells into the CNS was first probed in an ex vivo
organotypic brain slice model. Delivery of iN cells on microscale

NATURE COMMUNICATIONS | 7:10862 | DOI: 10.1038/ncomms10862 | www.nature.com/naturecommunications

5

ARTICLE

NATURE COMMUNICATIONS | DOI: 10.1038/ncomms10862

f

Dissociated excitatory iNs

iNs seeded on scaffold
20 mV

a

100 ms
200 pA

Dissociated iNs

Cortical–striatal brain
slices

100 ms

Ex-vivo co-culture

iN-seeded scaffolds
Dissociated
Scaffold

40

h
Number of action
potentials

50

30
20

1,800

1,600

1,400

1,200

800

1,000

600

400

0

0

10
200

Frequency (%)

b

d

70 pA

70 pA

–20 pA

–20 pA

Scaffold-iN cells
6

Dissociated-iN cells

4
2
0
10

Neurite length (μm)

i

1,500

***

20

30
40
50
Current injected (pA)

Dissociated excitatory iNs

60

70

iNs seeded on scaffold
1 nA

e
Average neurite
length (μm)

c

200 pA
20 mV

g

1,000

100 ms
500

Dissociated

Scaffold

90 mV

90 mV

–100 mV

–100 mV

Figure 4 | iNs supported by scaffolds support outgrowth and survival ex vivo. (a) Hundred-micrometre edge scaffolds were cut from electrospun fibres
before seeding with iNs, followed by injection onto ex vivo cultured mouse pup brain slices. (b–e) Neurite length was found to be significantly enhanced in
transplanted scaffold-supported iNs when comparing dissociated GFP-labelled iNs (b) with iNs seeded on scaffolds (c) injected onto mouse ex vivo brain
slices (n ¼ 8 brain slices for each transplantation mode). (f–h) Both scaffold-supported iNs (n ¼ 6) and dissociated iNs (n ¼ 5) fire action potentials in
response to current injection 14 days after transplantation onto mouse ex vivo brain slices, however, scaffold-supported iNs displayed enhanced excitability
relative to dissociated iNs. Both modes of transplanted iNs were observed to have mature sodium channel expression (i). ***Po0.0001 by one-way
ANOVA. Scale bar, 20 mm. All error bars presented as mean±1 s.d.

scaffolds markedly improved both neurite outgrowth and
electrical functionality after transplantation compared with the
effects of injected isolated cell suspensions. This suggests that
cell retention and survival are enhanced because transplantation
of iN cells on microscale scaffolds preserves cell–cell contacts
while avoiding the need for dissociation. Dissociation of cells
before injection likely leads to increased anoikis38,39, contributing
to diminished engraftment after transplantation. The
improvement in electrical activity after transplantation of iN
cells in scaffolds is promising, and suggests that these benefits
may translate in vivo.
Transplantation of iN cells in scaffolds into the mouse striatum
showed that the percentage of viable cells after 3 weeks was an
order-of-magnitude greater than that relative to injection of
isolated single cells. This increased survival is likely indicative of
decreased anoikis or apoptosis that otherwise arises from
enzymatic disruption of cell–cell and cell–matrix interactions38,39.
These results are promising particularly in light of recent studies
investigating the effects of human iPS cell-derived neurons on
treating CNS disorders that have shown efficacy in vivo even
with low levels of cell survival1,10, suggesting that increased
cell survival could lead to amplified levels of efficacy of these cell
therapeutics. In addition, host neuron post-synaptic markers were
detected adjacent to pre-synaptic marker-expressing transplanted
neurons, suggesting that single-factor reprogrammed neurons are
capable of functionally integrating into host tissue. Interestingly,
6

some authors have found that extensive lesions can actually
enhance neuronal graft survival and integration, as denervated
targets may provide a positive stimulatory effect to new
neurons40,41, suggesting that even greater cell survival may be
possible when applying this scaffold system in a
neurodegenerative disease or injury context.
Finally, the ability for 3D microscale scaffolds to support
co-culture and concurrent transplantation of neuronal networks
consisting of neurons of multiple subtypes was evaluated.
Transplantation of intact networks of mixed subtype neuron
populations could be an invaluable tool for treating complex
brain disorders or injuries that affect multiple subtypes, or for
priming transplanted neurons with pre-established synaptic
inputs. For example, attempts for transplanting dopamine
neurons derived from fetal brain tissue42,43 or derived from
human iPS cells1,44 have been made and yield amelioration of
Parkinson’s Disease; with the paradigm we proposed one can
envision a model to graft mini-neurocircuitry composed of
excitatory-dopaminergic neurons into the host brain. We
hypothesize that proper excitation from the mini-neurocircuitry
could produce more profound amelioration of locomotive deficits
in Parkinson’s Disease.
Induced human neurons offer a rich cell source for a variety of
in vitro and in vivo applications in modelling and treating CNS
diseases and injuries. Our prototype epitomizes a biomaterial
device for subtype specific neuronal reprogramming and

NATURE COMMUNICATIONS | 7:10862 | DOI: 10.1038/ncomms10862 | www.nature.com/naturecommunications

ARTICLE

NATURE COMMUNICATIONS | DOI: 10.1038/ncomms10862

a

Dissociated iNs

Scaffoldsupported
cell treatment

In-vivo transplantation

Dissociated
cell
treatment

b

c

x–z plane

iN-seeded scaffolds

f

y–z plane

d

e

y
x
GFP synaptophysin

*

14

h

10

PSD95

i

60

12

2

Unmatched

Dissociated

Scaffold

Dissociated

0

Matched,
1,000 cells

30
20

j

10
0
Dissociated

4

40

Scaffold

6

Neurite length (µm)

50

8

Scaffold

Percentage of
surviving cells of total

g

*

Figure 5 | iNs supported by scaffolds support outgrowth and survival in vivo. (a) iN-seeded scaffolds were injected into mouse striatum and
compared with injected dissociated cells. GFP-expressing surviving iNs were found 3 weeks post transplantation in mouse striatum, both for dissociated iNs
(b) and with iN-seeded microscaffolds (c–e), located in the vicinity of microscaffolds (indicated with dashed line). Scale bar, 25 mm; 100 mm for b,c and
d,e, respectively. (f) Post-synaptic density protein 95 (PSD-95, blue, indicated using downward-pointing arrows) was detected adjacent to transplanted
GFP-labelled iN neurite terminals, which co-localized with synaptophysin (red, with red þ green or yellow regions indicated with upward-pointing arrows),
suggestive of synaptic integration with host tissue. Scale bar, 5 mm. (g) Quantification of surviving cells yielded an average survival rate of 5.74±3.16% out
of an average 802±95.8 injected scaffold-seeded iN cells (n ¼ 3), compared with an average survival rate of 0.15±0.15% out of 100,000 injected
dissociated iN cells, or 7.58±4.60% of injected scaffold-seeded iN cells compared with 0±0% out of 1,000 injected dissociated iN cells, for a similarmagnitude number of injected cell comparison (n ¼ 3). *Po0.05 by one-way ANOVA, all error bars presented as mean±1 s.d. There was no significant
difference in neurite length between the two modes of transplanted cells (h). GFP-expressing NeuroD1 iNs and RFP-expressing dopamine neurons were
preserved in close proximity when co-transplanted on microscaffolds (indicated with dashed line) (j), while only some sparse dissociated and transplanted
iNs survived (i) 1 week post transplantation into mouse striatum. Scale bar, 25 mm.

NATURE COMMUNICATIONS | 7:10862 | DOI: 10.1038/ncomms10862 | www.nature.com/naturecommunications

7

ARTICLE

NATURE COMMUNICATIONS | DOI: 10.1038/ncomms10862

transplantation strategy that encourages transplant survival and
engraftment. Similar prototypes could be engineered to apply
human neuronal cells of varying subtypes for targeted treatment
of a wide range of CNS disorders.
Methods
Lentivirus production. The lentiviral constructs rtTA-FUW, Tet-O-FUW
NeuroD1, Tet-O-FUW EGFP, Tet-O-FUW Ascl1, Tet-O-FUW Nurr1, Tet-O-FUW
Lmx1a, Tet-O-FUW Pitx3, Tet-O-FUW EN1, Tet-O-FUW Foxa2, and the
packaging plasmids pMDL.g/pRRE, pMD.2 (VSVg), and RSV-REV were
constructed by the Marius Wernig laboratory at Stanford University. Lentivirus
particles were generated by first transfecting 293FT cells by the calcium phosphate
precipitation method with each of the three packaging plasmids and one of the
lentiviral plasmids in standard 293FT medium, which consists of DMEM
supplemented with 10% fetal bovine serum, 1% non-essential amino acids (all from
Life Technologies, Carlsbad, CA) and 1% penicillin/streptomycin (Lonza,
Walkersville, PA)45. Medium is replaced after 24 h and medium containing
viral particles is collected at 48 and 72 h. Viral particles are concentrated
200  by ultracentrifugation at 25,000 r.p.m. for 2 h at 4 °C, and stored at  80 °C
until use.

iPS cell culture and neuronal conversion. Human iPS cells were obtained from
the Rutgers University RUCDR Infinite Biologics. iPS cells were derived from
human foreskin fibroblasts by retroviral infection with Oct-4, Sox2, Klf4 and
c-Myc46. Human iPS cells were cultured on Matrigel-treated dishes in mTeSR-1
(ref. 24). iPS cells were passaged with dispase every 5–7 days with manual removal
of spontaneously differentiated areas with fire-polished glass pipets. iPS cells from
passage 15–35 were used in these studies. iPS cells were passaged as single cells with
Accutase (Stem Cell Technologies) plated at high density (8  104 cells per cm2) in
mTeSR-1 supplemented with 5 mM Y-27632 (R&D Systems, Minneapolis, MN)35.
The following day, medium was replaced with mTeSR-1 with 5 mM Y-27632 and
2 mM polybrene (Sigma-Aldrich, St Louis, MO) and concentrated virus was added
(rtTA-FUW, Tet-O-FUW NeuroD1, and in some experiments, Tet-O-FUW
EGFP). The following day, medium was replaced with mTeSR-1 and cells were
returned to normal culture for up to eight additional passages. To induce neuronal
conversion, infected iPSCs (termed iPSC-RNs) were passaged with Accutase and
Y-27632, and the following day switched to an N2-based conversion medium
(N2M) consisting of DMEM/F12, 2 mM L-glutamine, 1  N2 supplement,
1  non-essential amino acids, 1% penicillin/streptomycin and 2 mg ml  1
doxycycline, which induces expression of the NeuroD1 and EGFP constructs.
Medium was replaced daily, and cells were replated on day 3–5 onto 2D or 3D
substrates. One day after replating, medium was switched to NBM, consisting of
neurobasal medium, 2 mM L-glutamine, 1  B27 supplement without Vitamin A
(Life Technologies), 0.2 mM ascorbic acid, 1 mM cAMP (Sigma-Aldrich),
2 mg ml  1 doxycycline and 10 ng ml  1 each of brain-derived neurotrophic factor,
Glial cell-line derived neurotrophic factor and neurotrophin-3 (Peprotech).
Medium was replaced every 2–3 days for the duration of the experiments.
To generate dopamine iNs for co-culture studies, 2.5  105 cells per six-well
plate were plated, and the following day medium was replaced with mTeSR-1 with
5 mM Y-27632, 2 mM polybrene, and concentrated virus was added (rtTA-FUW,
Ascl1, Nurr1, Lmx1a and, in some experiments, Tet-O-FUW RFP). After three
days, medium was replaced with Neurobasal media (Life Technologies) and
concentrated virus (Pitx3, EN1, Foxa2). Four days after initial infection, the
dopamine iNs would be co-seeded onto microscale scaffolds.

Electrospun fibrous substrate fabrication. Fibrous substrates and 2D polymer
film controls were fabricated from tyrosine-derived polycarbonate pDTEc via
electrospinning and spin-coating, respectively24. These polymers come from a
combinatorial library of polymers with tunable hydrophobicity and degradation
properties. pDTEc was dissolved in 1,1,1,3,3,3-Hexafluoro-2-propanol
(Sigma-Aldrich) at 9%, or 18% weight by volume for thin and thick fibre
configurations respectively. For the 9% weight by volume solution, 5% N,Ndimethylformamide was added to prevent beading of fibres. Polymer solutions
were electrospun from a þ 18 kV spinneret to a  6 kV flat plate collector at
3 ml h  1 over a distance of 30 cm, resulting in fibre mats 250–500-mm thick.
Surface morphology was observed on an AMRAY 1830 I scanning electron
microscope and fibre diameter was quantified by measuring 100 individual fibres
from each fibrous substrate using NIH-ImageJ software (http://rsb.info.nih.gov/ij/).
Quantitative data are presented as mean±95% confidence interval. Control
2D polymer films were prepared using a 1% polymer solution dissolved in
tetrahydrofuran, which was then spin-coated onto glass coverslips at 4,000 r.p.m.
for 30 s (ref. 47). For in vitro studies, electrospun fibres were sterilized by ultraviolet
treatment, oxygen plasma treated for hydration, and adsorbed with 10 mg ml  1
poly-D-lysine in HEPES buffer (pH 8.4) and 4 mg ml  1 laminin in PBS before
seeding dissociated iNs 4 days after initiating neuronal conversion with
N2M þ dox.
8

Microscale scaffold preparation and neuronal seeding. Microscale fibrous
scaffolds were prepared by cutting oxygen plasma treated and hydrated electrospun
fibre mats into 100 mm squares with a McIlwain tissue chopper (Vibratome),
and sterilized and coated with poly-D-lysine and laminin as described above. On
day 4 of neuronal conversion, iN cells were dissociated with Accutase and
resuspended in N2M with dox at 5 million cells per ml. The scaffolds and iN
cell suspension were mixed in a 48-well non-TCPS plate for 1 h with occasional
gentle agitation, followed by three washes to remove unattached cells. Scaffolds
containing iN cells were cultured for 1 day in N2M, then cultured in NBM until
used for ex vivo or in vivo experiments. For experiments examining co-cultures of
dopamine iNs and NeuroD1 iNs, dissociated dopamine and NeuroD1 iNs were
dissociated, resuspended and combined 1:1 ratios to a concentration of 5 million
total cells per ml.
Ex vivo slice preparation and scaffold transplantation. Organotypic mouse brain
slice cultures were prepared by isolating the cortico-striatal area of 6-day-old C57BL/
6 mouse brains and cutting into 300 mm slices using a vibratome (Leica Microsystems) in ice-cold sucrose solution (204 mM sucrose, 26.2 mM NaHCO3, 11 mM
glucose, 2.5 mM KCl, 2 mM MgSO4, 1 mM NaH2PO4, 0.5 mM CaCl2 saturated with
95% O2 and 5% CO2) (ref. 48). Slices were transferred onto Millicell organotypic cell
culture inserts (Millipore) in a six-well tissue-culture plate and cultured in
Neurobasal A medium (Life Technologies) supplemented with 1 mg ml  1 insulin,
0.5 mM ascorbic acid, 25% horse serum, and 1% penicillin/streptomycin. Mouse
brain slices were cultured for 2 days before iN cell transplantation. Two days after
preparing brain slices, dissociated and scaffold-seeded GFP þ iN cells (day 6
post-conversion) were transplanted onto the surface of the cortico-striatal slices
using a pipette. Medium was changed every 2–3 days and slices were fixed in 4%
paraformaldehyde after 7 days for immunohistochemistry to quantify neurite outgrowth, or cultured 14 days before electrophysiology studies.
In vivo cell injection and scaffold transplantation. All animal experiments were
carried out according to the Rutgers University Policy on Animal Welfare and were
approved by the Institutional Animal Care and Use Committee (IACUC) at Rutgers University Robert Wood Johnson Medical School. Male, 5-week-old NODSCID IL2Rgc null mice (20–35 g; Jackson Laboratory) were anaesthetised with
isoflurane (induction at 4% and maintained at 0.5–1% inhalation) and injected
with a total volume of 10 ml, containing either 1  103, 1  105 dissociated GFP þ
iN cells or B10 scaffolds seeded with GFP þ iN cells (B85 cells per scaffold) at day
6 after initiating neuronal conversion, resuspended in ice-cold MEM (Life Technologies). Cells or cells on scaffolds were injected stereotactically into the striatum
using a 100 ml gastight Hamilton syringe and 21G Hamilton needle. Bilateral
injections were made at the following coordinates (in mm): AP, 0.5 (from bregma);
ML,  2.0; DV,  3.0 (from dura). Mice were killed and processed for immunohistochemistry 3 weeks after transplantation for quantification of human iN cells
within the graft by visualization of the human-specific nuclei antibody HuNu
(Millipore) GFP, and RFP in the case of multiple-subtype co-transplantation (see
Supplementary Table 1 for a detailed list of primary antibodies). Neurite outgrowth
was quantified by measurement of GFP þ neurites.
Electrophysiology. Electrophysiolocial recordings were performed at prescribed
time intervals from iN cells in 2D or 3D configurations in whole-cell mode using a
Multiclamp 700B amplifier12,45. The bath solution contained 140 mM NaCl, 5 mM
KCl, 2 mM MgCl1, 2 mM CaCl2, 10 mM HEPES and 10 mM glucose, at pH 7.4. The
pipette solution for whole-cell voltage-dependent current recordings and for
current-clamp experiments contained 10 mM KCl, 123 mM K-gluconate, 1 mM
MgCl2, 10 mM HEPES, 4 mM glucose, 1 mM EGTA, 0.1 mM CaCl2, 1 mM K2ATP,
0.2 mM Na4GTP, pH 7.2. Membrane resting potentials were kept in the range of
 65 to  70 mV, with step currents injected to elicit action potentials.
Immunocytochemistry. Neuronal phenotypes were characterized by
immunocytochemistry for 2D and 3D substrates. Cells were fixed with 4% paraformaldehyde for 30 min at room temperature, followed by three washes with PBS.
Cells were simultaneously blocked and permeabilized in blocking buffer consisting
of PBS supplemented with 5% normal goat serum (MP Biomedicals), 1% bovine
serum albumin (Sigma-Aldrich), and 0.1% Triton X-100 (Sigma-Aldrich) for 1 h at
room temperature. Primary antibodies were incubated in blocking buffer at 4 °C
overnight, followed by three 15-min PBS washes. Fluorophore-conjugated secondary antibodies (Alexa Fluor 488, 594, or 647, Life Technologies) were incubated
in blocking buffer for 1 h at room temperature, followed by three 15-min washes
and at least one wash of 1–2 h. Samples were counter-stained with 40 ,6-Diamidino2-phenylindole dihydrochloride (Sigma-Aldrich) to visualize nuclei and mounted
with Prolong Gold Anti-Fade reagent (Life Technologies) before imaging. Samples
were imaged on a Leica SP2 laser scanning confocal microscope using 10  dry or
63  immersion objectives. Images of cells on 3D electrospun fibres are presented
as maximum intensity projections of z-stacks, unless otherwise stated. Image
quantification was performed via custom ImageJ macros and manual counting. A
complete listing of the primary antibodies used in this study can be found in
Supplementary Table 1.

NATURE COMMUNICATIONS | 7:10862 | DOI: 10.1038/ncomms10862 | www.nature.com/naturecommunications

ARTICLE

NATURE COMMUNICATIONS | DOI: 10.1038/ncomms10862

Calcium imaging. iN cells were labelled with 3 mM Fluo-4 AM calcium indicator
dye for 30 min at room temperature in extracellular bath solution (147 mM NaCl,
5 mM KCl, 2 mM CaCl2, 2 mM MgCl2, 10 mM HEPES and 10 uM Glucose, pH 7.4
and osmolarity 290–300 mOsm) containing 0.02% pluronic F-127. After incubation, cells were rinsed twice with bath solution and incubated for an additional
30 min at room temperature to allow complete de-esterification of the Fluo-4
AM dye before imaging. Time-lapse imaging was performed on a Leica SP2 confocal microscope at a resolution of 512  512 pixels and a time resolution of
1 Hz. Image analysis was performed using custom ImageJ macros and MATLAB
routines for semi-automated ROI selection and peak detection to quantify
the fraction of cells that were electrically active. Cells were considered to be electrically active if they showed a spike in fluorescence following application of an
electrical stimulus. The electrical stimulus was applied using a function generator
(Global Specialties) and consisted of a 5 s stimulus at 7.5 V cm  1 with 5 ms square
pulses at 40 Hz.
Quantitative real-time polymerase chain reaction. Total RNA was extracted
from iPS cells cultured in 2D or 3D using the RNEasy kit (Qiagen, Valencia, CA)
according to the manufacturer’s instructions, including treatment with RNase-free
DNase to remove genomic DNA. A high-capacity cDNA reverse transcription kit
(Applied Biosystems, Foster City, CA) containing random primers was used to
reverse transcribe 200 ng total RNA from each sample to cDNA. Taqman gene 194
expression master mix and Taqman gene-expression assays (Applied Biosystems)
were used for template amplification of 10 ng cDNA per reaction. Quantitative
real-time polymerase chain reaction (qRT–PCR) was carried out on a 7500 Fast
Real-Time PCR instrument (Applied Biosystems). Relative gene expression was
calculated using the DDCT method, normalizing to GAPDH as the endogenous
control and undifferentiated IPS cells in standard 2D culture (on Matrigel-coated
dishes in mTeSR media) after 7 days as the reference sample. Taqman
gene-expression assays were used in these studies, listed in Supplementary
Table 2.
Statistical analysis. All data are presented as mean±s.d. Statistical significance is
evaluated by single-factor ANOVA and a Tukey’s post hoc test, with Po0.05
considered statistically significant.

References
1. Kriks, S. et al. Dopamine neurons derived from human ES cells efficiently
engraft in animal models of Parkinson’s disease. Nature 480, 547–551 (2011).
2. Yamauchi, T. et al. Therapeutic effects of human multilineage-differentiating
stress enduring (MUSE) cell transplantation into infarct brain of mice. PLoS
ONE 10, e0116009 (2015).
3. Anderson, A. J. et al. Achieving stable human stem cell engraftment and
survival in the CNS: is the future of regenerative medicine immunodeficient?
Regen. Med. 6, 367–406 (2011).
4. Dimos, J. T. et al. Induced pluripotent stem cells generated from patients with
ALS can be differentiated into motor neurons. Science 321, 1218–1221 (2008).
5. Nguyen, H. N. et al. LRRK2 mutant iPSC-derived DA neurons demonstrate
increased susceptibility to oxidative stress. Cell Stem Cell 8, 267–280 (2011).
6. Tran, N. N., Ladran, I. G. & Brennand, K. J. Modeling schizophrenia using
induced pluripotent stem cell-derived and fibroblast-induced neurons.
Schizophr. Bull. 39, 4–10 (2013).
7. Hibaoui, Y. & Feki, A. Human pluripotent stem cells: applications and
challenges in neurological diseases. Front. Physiol. 3, 267 (2012).
8. Burns, T. C. & Steinberg, G. K. Stem cells and stroke: opportunities, challenges
and strategies. Expert. Opin. Biol. Ther. 11, 447–461 (2011).
9. Thoma, E. C. et al. Ectopic expression of neurogenin 2 alone is sufficient to
induce differentiation of embryonic stem cells into mature neurons. PLoS ONE
7, e38651 (2012).
10. Zhang, Y. et al. Rapid single-step induction of functional neurons from human
pluripotent stem cells. Neuron 78, 785–798 (2013).
11. Chanda, S. et al. Generation of induced neuronal cells by the single
reprogramming factor ASCL1. Stem Cell Rep. 3, 282–296 (2014).
12. Pang, Z. P. et al. Induction of human neuronal cells by defined transcription
factors. Nature 476, 220–223 (2011).
13. Han, F. et al. Human induced pluripotent stem cell-derived neurons improve
motor asymmetry in a 6-hydroxydopamine-induced rat model of Parkinson’s
disease. Cytotherapy 17, 665–679 (2015).
14. Cukierman, E., Pankov, R., Stevens, D. R. & Yamada, K. M. Taking cell-matrix
adhesions to the third dimension. Science 294, 1708–1712 (2001).
15. Doyle, A. D., Wang, F. W., Matsumoto, K. & Yamada, K. M. One-dimensional
topography underlies three-dimensional fibrillar cell migration. J. Cell Biol. 184,
481–490 (2009).
16. Martino, S., D’Angelo, F., Armentano, I., Kenny, J. M. & Orlacchio, A. Stem
cell-biomaterial interactions for regenerative medicine. Biotechnol. Adv. 30,
338–351 (2012).
17. Nisbet, D. R. et al. Interaction of embryonic cortical neurons on nanofibrous
scaffolds for neural tissue engineering. J. Neural. Eng. 4, 35–41 (2007).

18. Gertz, C. C. et al. Accelerated neuritogenesis and maturation of primary spinal
motor neurons in response to nanofibers. Dev. Neurobiol. 70, 589–603 (2010).
19. Cheng, T. Y., Chen, M. H., Chang, W. H., Huang, M. Y. & Wang, T. W. Neural
stem cells encapsulated in a functionalized self-assembling peptide hydrogel for
brain tissue engineering. Biomaterials 34, 2005–2016 (2013).
20. Bible, E. et al. Neo-vascularization of the stroke cavity by implantation of
human neural stem cells on VEGF-releasing PLGA microparticles. Biomaterials
33, 7435–7446 (2012).
21. Yasuda, H. et al. Effect of biodegradable fibrin scaffold on survival, migration,
and differentiation of transplanted bone marrow stromal cells after cortical
injury in rats. J. Neurosurg. 112, 336–344 (2010).
22. Ma, F. et al. Accelerating proliferation of neural stem/progenitor cells in
collagen sponges immobilized with engineered basic fibroblast growth factor for
nervous system tissue engineering. Biomacromolecules 15, 1062–1068 (2014).
23. Skaper, S. D., Argentini, C. & Barbierato, M. Culture of neonatal rodent
microglia, astrocytes, and oligodendrocytes from cortex and spinal cord.
Methods Mol. Biol. 846, 67–77 (2012).
24. Carlson, A. L. et al. Microfibrous substrate geometry as a critical trigger for
organization, self-renewal, and differentiation of human embryonic stem cells
within synthetic 3-dimensional microenvironments. FASEB J 26, 3240–3251
(2012).
25. Bourke, S. L. & Kohn, J. Polymers derived from the amino acid -tyrosine:
polycarbonates, polyarylates and copolymers with poly(ethylene glycol). Adv.
Drug. Deliv. Rev. 55, 447–466 (2003).
26. Hooper, K. A., Macon, N. D. & Kohn, J. Comparative histological evaluation of
new tyrosine-derived polymers and poly (L-lactic acid) as a function of polymer
degradation. J. Biomed. Mater. Res. 41, 443–454 (1998).
27. Christopherson, G. T., Song, H. & Mao, H.-Q. The influence of fiber diameter
of electrospun substrates on neural stem cell differentiation and proliferation.
Biomaterials 30, 556–564 (2009).
28. Espuny-Camacho, I. et al. Pyramidal neurons derived from human pluripotent
stem cells integrate efficiently into mouse brain circuits in vivo. Neuron 77,
440–456 (2013).
29. Cherry, J. F., Bennett, N. K., Schachner, M. & Moghe, P. V. Engineered
N-cadherin and L1 biomimetic substrates concertedly promote neuronal
differentiation, neurite extension and neuroprotection of human neural stem
cells. Acta. Biomater. 10, 4113–4126 (2014).
30. Landers, J. et al. Carbon nanotube composites as multifunctional substrates for
in situ actuation of differentiation of human neural stem cells. Adv. Healthc.
Mater. 3, 1745–1752 (2014).
31. Cherry, J. F. et al. Oriented, multimeric biointerfaces of the L1 cell adhesion
molecule: an approach to enhance neuronal and neural stem cell functions on
2-D and 3-D polymer substrates. Biointerphases 7, 22 (2012).
32. Bruses, J. L. N-cadherin signaling in synapse formation and neuronal
physiology. Mol. Neurobiol. 33, 237–252 (2006).
33. Tan, Z. J., Peng, Y., Song, H. L., Zheng, J. J. & Yu, X. N-cadherin-dependent
neuron-neuron interaction is required for the maintenance of activity-induced
dendrite growth. Proc. Natl Acad. Sci. USA 107, 9873–9878 (2010).
34. Piccoli, G., Rutishauser, U. & Bruses, J. L. N-cadherin juxtamembrane domain
modulates voltage-gated Ca2 þ current via RhoA GTPase and Rho-associated
kinase. J. Neurosci. 24, 10918–10923 (2004).
35. Watanabe, K. et al. A ROCK inhibitor permits survival of dissociated human
embryonic stem cells. Nat. Biotech. 25, 681–686 (2007).
36. Xu, Y. et al. Revealing a core signaling regulatory mechanism for pluripotent
stem cell survival and self-renewal by small molecules. Proc. Natl Acad. Sci.
USA 107, 8129–8134 (2010).
37. Li, L. et al. Individual cell movement, asymmetric colony expansion,
rho-associated kinase, and E-cadherin impact the clonogenicity of human
embryonic stem cells. Biophys. J. 98, 2442–2451 (2010).
38. Marchionini, D. M. et al. Interference with anoikis-induced cell death of
dopamine neurons: implications for augmenting embryonic graft survival in a
rat model of Parkinson’s disease. J. Comp. Neurol. 464, 172–179 (2003).
39. Frisch, S. M. & Screaton, R. A. Anoikis mechanisms. Curr. Opin. Cell Biol. 13,
555–562 (2001).
40. Kirik, D., Winkler, C. & Bjorklund, A. Growth and functional efficacy of
intrastriatal nigral transplants depend on the extent of nigrostriatal
degeneration. J. Neurosci. 21, 2889–2896 (2001).
41. Nikkhah, G. et al. Microtransplantation of dopaminergic cell suspensions:
further characterization and optimization of grafting parameters. Cell
Transplant. 18, 119–133 (2009).
42. Studer, L. et al. Enhanced proliferation, survival, and dopaminergic differentiation
of CNS precursors in lowered oxygen. J. Neurosci. 20, 7377–7383 (2000).
43. Studer, L., Tabar, V. & McKay, R. D. Transplantation of expanded
mesencephalic precursors leads to recovery in parkinsonian rats. Nat. Neurosci.
1, 290–295 (1998).
44. Wernig, M. et al. Neurons derived from reprogrammed fibroblasts functionally
integrate into the fetal brain and improve symptoms of rats with Parkinson’s
disease. Proc. Natl Acad. Sci. USA 105, 5856–5861 (2008).

NATURE COMMUNICATIONS | 7:10862 | DOI: 10.1038/ncomms10862 | www.nature.com/naturecommunications

9

ARTICLE

NATURE COMMUNICATIONS | DOI: 10.1038/ncomms10862

45. Vierbuchen, T. et al. Direct conversion of fibroblasts to functional neurons by
defined factors. Nature 463, 1035–1041 (2010).
46. Takahashi, K. et al. Induction of pluripotent stem cells from adult human
fibroblasts by defined factors. Cell 131, 861–872 (2007).
47. Treiser, M. D. et al. Cytoskeleton-based forecasting of stem cell lineage fates.
Proc. Natl Acad. Sci. USA 107, 610–615 (2009).
48. Citri, A. et al. Calcium binding to PICK1 is essential for the intracellular
retention of AMPA receptors underlying long-term depression. J. Neurosci. 30,
16437–16452 (2010).

Acknowledgements
Support from various funding sources is gratefully acknowledged, including NIH
RESBIO: Integrated Resource for Polymeric Biomaterials P41 EB001046 (P.V.M., J.K.),
NSF IGERT on Stem Cell Science and Engineering, DGE 0801620 (A.L.C., A.H., P.V.M.),
Exploratory Research Grant from NJSCR and Stem Cell Core Grant from NJCST
(P.V.M., R.P.H.), NJSCR Fellowship (A.L.C.), NIH T32 EB005583 on Translational
Regenerative Medicine (N.L.F., J.K., P.V.M.), NIH 5R00NS051401 (K.P), NIH NIAAA
F31AA024033 (A.H.), NIAAA R01 AA023797 (Z.P.P.), and NIH NIDA DA035594
and DA03968 (Z.P.P., R.P.H.).

Author contributions
A.L.C., N.K.B., N.L.F., R.P.H., K.P., Z.P.P. and P.V.M. conceived the study and designed
the experiments. A.L.C., N.K.B., N.L.F., A.H., S.C. and J.C.M. performed the experiments.

10

A.L.C., N.K.B., N.L.F., A.H., S.C. and K.P. analysed the data. A.L.C., N.K.B., N.L.F.,
M.W., J.K., Z.P.P. and P.M. wrote the manuscript.

Additional information
Supplementary Information accompanies this paper at http://www.nature.com/
naturecommunications
Competing financial interests: The authors declare no competing financial
interests.
Reprints and permission information is available online at http://npg.nature.com/
reprintsandpermissions/
How to cite this article: Carlson, A. L. et al. Generation and transplantation of
reprogrammed human neurons in the brain using 3D microtopographic scaffolds.
Nat. Commun. 7:10862 doi: 10.1038/ncomms10862 (2016).
This work is licensed under a Creative Commons Attribution 4.0
International License. The images or other third party material in this
article are included in the article’s Creative Commons license, unless indicated otherwise
in the credit line; if the material is not included under the Creative Commons license,
users will need to obtain permission from the license holder to reproduce the material.
To view a copy of this license, visit http://creativecommons.org/licenses/by/4.0/

NATURE COMMUNICATIONS | 7:10862 | DOI: 10.1038/ncomms10862 | www.nature.com/naturecommunications


Aperçu du document GENERATION AND TRANSPLANTATION NEURONES 3D.pdf - page 1/10

 
GENERATION AND TRANSPLANTATION NEURONES 3D.pdf - page 2/10
GENERATION AND TRANSPLANTATION NEURONES 3D.pdf - page 3/10
GENERATION AND TRANSPLANTATION NEURONES 3D.pdf - page 4/10
GENERATION AND TRANSPLANTATION NEURONES 3D.pdf - page 5/10
GENERATION AND TRANSPLANTATION NEURONES 3D.pdf - page 6/10
 







Télécharger le fichier (PDF)





Documents récents du même auteur


HENRI   19 juillet 2016
IDEE   19 juillet 2016
CAFE   19 juillet 2016
PEAU   19 juillet 2016
NOVLANGUE   19 juillet 2016
AVC   19 juillet 2016
DAMIEN   19 juillet 2016
JET LAG   19 juillet 2016
A Putative Human Pheromon1   19 juillet 2016
JORDI LLORENS   19 juillet 2016
reactive and proactive aggression   17 juillet 2016
Holy Warriors Exploring the Psychological Processe   17 juillet 2016
baillement orl (1)   6 juillet 2016
Bienfait de la randonnée   6 juin 2016
pleine lune et sommeil   21 avril 2016
Past experience   18 avril 2016
Musiq&Parf LaCelle 05-03-2016   6 mars 2016
HFEA approval for new   27 février 2016
CRISP CHINE 2   27 février 2016
IGAS VALPROATE   24 février 2016

Sur le même sujet..







Ce fichier a été mis en ligne par un utilisateur du site Fichier PDF. Identifiant unique du document: 00411691.
⚠️  Signaler un contenu illicite
Pour plus d'informations sur notre politique de lutte contre la diffusion illicite de contenus protégés par droit d'auteur, consultez notre page dédiée.