You are on page 1of 13

J Mol Neurosci (2009) 37:225–237

DOI 10.1007/s12031-008-9123-1

Quantitative Assessment of Neuronal Differentiation


in Three-dimensional Collagen Gels Using Enhanced Green
Fluorescence Protein Expressing PC12 Pheochromocytoma Cells
Hadar Arien-Zakay & Shimon Lecht & Anat Perets &
Blair Roszell & Peter I. Lelkes & Philip Lazarovici

Received: 27 May 2008 / Accepted: 5 June 2008 / Published online: 16 July 2008
# Humana Press 2008

Abstract There is a paucity of quantitative methods for an ideal model for investigating various aspects of differen-
evaluating the morphological differentiation of neuronal tiation to serve in neural engineering.
cells in a three-dimensional (3-D) system to assist in quality
control of neural tissue engineering constructs for use in Keywords Green fluorescent PC12 cells . NGF .
reparative medicine. Neuronal cells tend to aggregate in the Neuronal differentiation . Three-dimensional collagen gel
3-D scaffolds, hindering the application of two-dimensional
(2-D) morphological methods to quantitate neuronal differ-
entiation. To address this problem, we developed a stable Introduction
transfectant green fluorescence protein (GFP)-PC12 neuronal
cell model, in which the differentiation process in 3-D can be The PC12 cell line has been extensively used in neuroscience
monitored with high sensitivity by fluorescence microscopy. research as a neuronal model for studying neuronal signaling
Under 2-D conditions, the green cells showed collagen (Vaudry et al. 2002) and has become the model of choice for
adherence, round morphology, proliferation properties, ex- the study of neuronal differentiation (Fujita et al. 1989;
pression of the nerve growth factor (NGF) receptors TrkA and Ravni et al. 2006). When treated in conventional two
p75NTR, stimulation of extracellular signal-regulated kinase dimensional (2-D) tissue culture conditions with nanomolar
phosphorylation by NGF and were able to differentiate in a concentrations of nerve growth factor (NGF), PC12 cells
dose-dependent manner upon NGF treatment, like wild-type stop dividing, elaborate neurite processes (outgrowths), and
(wt)-PC12 cells. When grown within 3-D collagen gels, become electrically excitable (Fujita et al. 1989). NGF
upon NGF treatment, the GFP-PC12 cells differentiated, biological effects are mediated by two types of receptors:
expressing long neurite outgrowths. We describe here a new the NGF high-affinity tyrosine kinase (TrkA) receptor
validated method to measure NGF-induced differentiation in (Kaplan and Miller 2000) and the low-affinity neurotrophin
3-D. Having properties similar to those of wt-PC12 and an receptor p75 (p75NTR), a member of the tumor necrosis
ability to grow and differentiate in 3-D structures, these factor receptor superfamily (Chao et al. 1998). Activation
highly visualized GFP-expressing PC12 cells may serve as of these receptors by NGF initiates differentiation of the
cells with a time course of several weeks, characterized by
processes such as proliferation arrest and initiation of the
H. Arien-Zakay : S. Lecht : P. Lazarovici (*) neurite outgrowths in the first 2 days of treatment;
Department of Pharmacology and Experimental Therapeutics, continuous, progressive elongation of the neurites for up
School of Pharmacy, Faculty of Medicine,
to 3 weeks of treatment, including extensive sprouting
The Hebrew University of Jerusalem,
P.O. Box 12065, Jerusalem 91120, Israel (branching) and synapse connections between the neurites;
e-mail: philipl@ekmd.huji.ac.il and generation of neurite networks and ganglia-like cell
organization due to aggregation of neuronal cell bodies
A. Perets : B. Roszell : P. I. Lelkes : P. Lazarovici
(Fujita et al. 1989).
School of Biomedical Engineering, Science and Health Systems,
Drexel University, Lately, PC12 cells have become an important cellular
Philadelphia, PA 19122, USA tool in neural engineering. Their morphological features,
226 J Mol Neurosci (2009) 37:225–237

such as growth, branching, differentiation, and synapse Materials and Methods


formation were found to be influenced by different matrix-
resident biophysical cues such as micro- and nanotopog- Cell Cultures
raphy (Foley et al. 2005; Moxon et al. 2007), including
substrate stiffness (Leach et al. 2007). Furthermore, PC12 PC12 cells (NIH clone, originally provided by Dr. G.
cells were used to explore the microenvironmental adequacy Guroff, NICHD, NIH) were grown in 25 cm2 flasks in
for neuronal cells of new bioengineering materials such as Dulbecco’s modified Eagle’s medium (DMEM) supple-
agarose scaffolds (Yu et al. 1999), peptide scaffolds (Holmes mented with 7% fetal bovine serum (FBS), 7% horse
et al. 2000), neuron–microelectrode interfaces (Bieberich and serum, 20 U/ml penicillin and 20 μg/ml streptomycin, all
Anthony 2004; Moxon et al. 2007), microchannel-containing purchased from Biological Industries (Beit Haemek, Israel).
substrates (Mahoney et al. 2005), and microfabricated Cells were incubated at 37°C in 6% CO2 (Katzir et al.
silicon-based nanostructures (Lopez et al. 2006; Moxon 2002). The cells were detached from the flask mechanically
et al. 2007). and washed twice with phosphate-buffered saline (PBS)
A major issue in neuroengineering is the investigation before reseeding.
of the biocompatibility of scaffolds with neuronal cells, Transduced 293T (human kidney epithelial, ATCC,
aimed at generating a three-dimensional (3-D) neuronal Manassas, VA, USA) cells were cultured in DMEM
constructs for implantation in patients in order to repair containing 400 μg/ml of a neomycin analogue, G418
defective neural pathways. To achieve this goal, it is (Gibco BRL, Rockville, MD, USA), 10% FBS, 20 U/ml
important to use in vitro models in which it is possible to penicillin, and 20 μg/ml streptomycin (Kosaka et al. 2004).
visualize cell growth in the 3-D construct. In general, the All experiments were carried out in a clean room, according
methods used to date include fixation of the neuronal- to ISO7 requirements (10,000 particles/m3) using a P2+
containing scaffolds and histological evaluation of the facility and lentiviral vector generation and transduction
morphology of the fixed cells, allowing temporal “snap- according to NIH biosafety guidelines.
shots” at defined time points, but not a continual real time
monitoring of neuronal development. As an alternative, Lentivirus Production and Transduction
nondestructive fluorescent labeling of the living cells
populating the 3-D construct may be advantageous. Such HIV-1-derived lentiviral vectors were generated by transient
labeling methods must also be compatible with the transfection of a human kidney 293T cell line by a three-
histochemical methods commonly used for routine mor- plasmid expression system. The packaging plasmid encodes
phological analysis (Tohill et al. 2004) and facilitate HIV-1 gag, pol, tat, and rev proteins; the envelope plasmid
noninvasive visualization of neuronal development. encodes the VSV/G glycoprotein gene; and the transfer
The goal of the present study was to generate a vector (TK113) encodes a CMV promoter driving the
fluorescent PC12 cell model to be used in 3-D neuronal expression of GFP (Kosaka et al. 2004). YTK 113 (vector),
engineering in order to address fundamental issues of VSV-G (packaging), and deltaNRF (envelope) plasmids
optimization of neuronal differentiation. In this study, we were transiently transfected into 293T cells using a calcium
generated a stable PC12 cell line expressing the green phosphate transfection method (Kosaka et al. 2004). All
fluorescent protein (GFP) from the jellyfish Aequorea transfections were carried out using 60-mm dishes coated
victoria. GFP does not naturally exist in mammalian with poly-L-lysine where 1.5–2.5×106 293T cells were
species and can be easily detected by UV light and plated in 5 ml of complete medium 24 h before transfection.
fluorescence microscopy. It also has the advantage of One hour before transfection, the medium was replaced
being a “vital dye” whose presence can be monitored in with serum-free DMEM containing antibiotics. The DNA/
living organisms and unfixed tissue (Okabe et al. 1997). It CaCl2 solution was prepared and left at room temperature
has the potential to elucidate nervous system development for 30 min before being added to the cells. The cells were
(Niell and Smith 2004) and to clarify the complex then returned to the 37°C incubator (5% CO2) for 24 h
biological process of neurite outgrowth regulation (Laketa when the medium was replaced with 7 ml of fresh
et al. 2007). In this study, using the GFP-PC12 cells, we complete medium. After 48 h, the medium was removed
defined a new morphological analysis for measurement of and filtered through a 0.45-μm filter to collect the viral
neuronal cell differentiation in a 3-D environment and particles secreted into the medium. The viral titer was
validated this method according to a commonly used adjusted to 4.0×107 IU/ml by dilution.
bioassay for NGF-induced differentiation under 2-D PC12 transduction with the virus was performed as
conditions. previously described for CD34+ human cells (Miyoshi et al.
J Mol Neurosci (2009) 37:225–237 227

1999). Briefly, cells were seeded at a density of 4×105 clone and wild-type cells. GFP expression was estimated by
cells/well in six-well tissue culture plates coated with 10% fluorescence microscopy. Clone adherence was assessed by
poly-L-lysine (Sigma-Aldrich, St. Louis, MO, USA). The monolayer generation; nonadherent cells grew in suspen-
cells were transduced with 1 ml of 4.0×107 IU/ml TK113 sion or partially attached to the dish. The wt-PC12 cultures
viral particles (HIV-based vector from which the GFP contained six different morphotypes (round, monopolar,
reporter gene is constitutively expressed under the control bipolar, tripolar, multipolar, and fibroblast like). The
of the cytomegalovirus promoter) and supplemented with selected GFP-PC12 clones showed a similar heterogeneous
2 ml of PC12 medium. The cells were incubated for 3 days morphology.
post transduction in a 37°C incubator, 6% CO2, at high
humidity. 2-D Cell Growth Conditions

FACS Sorting wt-PC12 or GFP-PC12 cells were seeded on 24-well plates


coated with 200 μg/ml type I rat tail collagen (BD
Flow cytometric analysis of the PC12-GFP/lentivirus- Biosciences) and cultured as described above.
transduced PC12 cell population was performed using a
FACS with fluorescence excitation at 488 nm wavelength 3-D Cell Growth Conditions
(Simons et al. 2006). In brief, the cell culture was
dissociated from the dish by agitation and resuspended in Sandwiched 3-D type I collagen gels at a final concentra-
PBS without magnesium and calcium, supplemented with tion of 1.5 mg/ml solution were constructed as previously
3% FBS and filtered through a 20-μm filter to generate described (Dietrich and Lelkes 2006; Mondrinos et al.
individually dispersed cells. Cell density was adjusted to 2007). Briefly, depending on the final volume required,
20×106 cells/ml. For analysis, 50,000 events were acquired calculated amounts of 10× Dulbecco’s PBS (Mediatech,
on a FACSCanto (Becton-Dickinson, San Jose, CA, USA) Herndon, VA, USA), 1 N NaOH, cell culture grade H2O,
flow cytometer using FACS Diva software (Becton- and type I rat collagen (BD Biosciences) were mixed, and
Dickinson). Evaluation of flow data was performed using the pH was adjusted under sterile conditions to 7.4±0.05.
FlowJo (Tree Star, Ashland, OR, USA). GFP-positive cells To generate the bottom layer of the gel, 400 μl of the
were collected under aseptic conditions in 5 ml FACS collagen solution was added to each well of a 24-well plate
polystyrene tubes (BD Biosciences, Bedford, MA, USA) and allowed to polymerize for 15 min at 37°C. For the
containing 400 μl of FBS. Wild-type PC12 (wt-PC12) cells, second layer, wt-PC12 or GFP-PC12 cells were transferred
at the same density, served as the negative control. into one volume of serum-free medium, which was then
mixed with the collagen solution. A 500-μl volume of this
Clonal Expansion of GFP-positive PC12 Cells suspension was added on top of the first layer. The cells
were evenly distributed by brief agitation of the culture
GFP-positive cells (3.5×105), aseptically sorted by the plates. These were then incubated for 15 min in a
FACS, were returned to culture. After 7 days of culture, hybridization oven (37°C). After complete polymerization
cloning of individual GFP-PC12 expressing cells was of the various gel assemblies, 1 ml of growth medium,
performed by limiting dilution. The cells were then diluted supplemented as described for the 2-D cell culture
to a concentration of 1 cell/100 μl and aliquoted into 480 conditions, was added to the sandwich-like gel assemblies.
wells of five 96-well plates. One day later, a well The cells were then cultured for various periods of time in a
containing a single fluorescent cell was identified by conventional CO2 incubator (6% CO2, 37°C), and the
fluorescence microscopy to ensure that each colony would medium was replaced every 2 days.
be derived from a single cell. To promote proliferation, the
GFP-PC12 cells were grown in PC12 growth medium Cell Proliferation
supplemented with 10% conditioning medium collected
from a confluent wt-PC12 cell culture. The clones were The proliferation of wt-PC12 and GFP-PC12 cells was
monitored every other day and split when the colony estimated daily over a period of up to 14 days using the
reached 50% confluence. After 5 weeks of expansion, the Alamar blue (BD Biosciences) staining method essentially
chosen clones were collected and frozen in PC12 medium as previously described (Nikolaychik et al. 1996). Briefly,
supplemented with 5% dimethylsulfoxide (Sigma-Aldrich). in both 2-D and 3-D cell culture conditions, the wells were
For biological characterization, different parameters were incubated for 4 h with the Alamar blue reagent (10% v/v in
evaluated over a period of several weeks in samples of each complete medium). At the end of the incubation period,
228 J Mol Neurosci (2009) 37:225–237

media samples were collected and their fluorescence Df is a statistical descriptor of fractal space (area and
intensity was evaluated in an ELISA reader at a excitation length) filled by neurite outgrowth (Kirchner et al. 1996). In
of 560 nm and emission of 595 nm at constant gain. Cell our experiments Df ranged from 0.6 to 1.25. According to this
proliferation was calculated as the relative increase in method, the neuronal network of neurite outgrowths is
fluorescence compared with day 0 (control). overlaid with a series of square boxes of decreasing size
(denoted in pixels, p) and the number of boxes (Np) containing
NGF-induced Neuronal Differentiation at least 1 pixel is counted. The negative value of the least-
squares regression slope of the plot of log Np vs log p yields
To assess neuronal differentiation, wt-PC12 or GFP-PC12 Df (Kirchner et al. 1996). The structure was considered fractal
cells were seeded in glass chamber slides (Nunc, Roskilde, if the r2 value of the regression line was >0.95.
Denmark) at a density of 4×105/cm2 on either collagen-
coated surfaces (2-D) or into 3-D collagen gels (1.5 mg/ml). Reverse Transcriptase Polymerase Chain Reaction
One day after seeding, the culture medium was replaced (RT-PCR)
with either the same PC12 medium (control) or with
differentiation medium (PC12 medium supplemented with The total RNA of NGF-treated and -untreated GFP-PC12
50 ng/ml mouse β-subunit, nerve growth factor 2S-mNGF; cells was isolated and genomic DNA was degraded from
Alomone Labs, Jerusalem, Israel). In the 3-D gel, NGF the RNA preparations, using the SV total RNA isolation
diffused through the gel allowing interaction with the cells system (Promega, Madison, WI, USA). A quantity of 1 μg
(Takezawa et al. 2007). The cells were then cultured for up of total RNA was reverse transcribed using the Reverse
to 45 days in a conventional CO2 incubator (6% CO2, 37°C). Transcription System (Promega), according to the manu-
facturer’s instructions. Then, PCR was performed in a final
Neuronal Differentiation Analysis volume of 50 μl containing 5 μg complementary DNA
(cDNA), 50 pmol of upstream sense and downstream sense
To assess neuronal differentiation, neurite outgrowth length primers, and 25 μl of GoTaq® Green Master Mix
was quantitated using the SigmaScanPro 5.0 program as (Promega). The cDNA was amplified by 35 cycles. To
previously described by us (Katzir et al. 2002). Briefly, the generate various cDNA fragments, a Mastercycler gradient
total length of outgrowths from each cell in the region of (Eppendorf, Germany) was programmed as follows: dena-
interest (ROI) was divided by its diameter, thus generating turation of cyclic parameters at 94°C for 1 min, annealing at
an elongation factor, denoted elongation (E) parameter. The 65°C for 1 min, and elongation at 72°C for 2 min (Arien-
experiments were performed at least three times and in Zakay et al. 2007). To identify the various messenger RNA
duplicate. In each well, three ROIs were photographed in (mRNA) transcripts, the following primers were used:
randomly chosen regions. In each ROI, 15–25 cells were
measured, i.e., at least 270 cells were measured for each & β-actin (285 bp, internal control)
treatment. Since neuronal cells in 3-D cultures generate
sense: TCATGAAGTGTGACGTTGACATCCGT-3′
aggregates rendering it difficult to measure the perykarion
antisense: CTTAGAAGCATTTGCGGTGCACGATG-3′
diameter of individual cells, we characterized a fractal
dimension (Df) as a suitable parameter for quantitative & TrkA (571 bp)
measurements of neuronal differentiation. This approach,
sense: CACTAACAGCACATCAAGAGAC-3′
introduced here for the first time for analyzing neuronal
antisense: GAAGACCATGAGCAATGGG-3′
outgrowth, is based on a method utilized for blood capillary
network sprouting measurements (Kirchner et al. 1996; & p75NTR (663 bp)
Lazarovici et al. 2006). ROIs were acquired in 2,560× sense: AGCCAACCAGACCGTGTGTG-3′
1,920 pixel dimensions and with resolution of 300 pixels/ antisense: TTGCAGCTGTTCCACCTCTT-3′
inch. Each image was transformed to 0–255 gray scale, and
a new transparent layer was generated using Photoshop™. All PCR products were analyzed by electrophoresis on
Using a 3-pixel wide digital pencil tool, all outgrowths agarose gel (2%) containing ethidium bromide for UV
were overlaid. The layer containing outgrowth overlays visualization. Wt-PC12-derived mRNA was used as a
(outgrowths network) was saved separately as an 8-bit positive control for validation.
JPEG file. The length and complexity of each outgrowth
networks image were measured using ImageJ software, Western Blot
utilizing fractal box count analysis to facilitate visual
inspection of the neuronal network morphology, and Total cellular protein was isolated using lysis buffer
“skeletonized” to calculate the fractal dimension (Df). The (Clontech, Mountain View, CA, USA). The amount of total
J Mol Neurosci (2009) 37:225–237 229

protein extracted was determined by Bio-Rad protein assay Results


(Bio-Rad, Hercules, CA, USA). Protein samples of 40 μg
were separated by SDS-PAGE (8–10%). The proteins were FACS Sorting Followed by Cloning of Stable Transfectant
electrotransferred (100 V, 1.5 h, 4°C) to nitrocellulose GFP-PC12 Cells
membranes and incubated for 2 h at room temperature (RT)
with 5% nonfat powdered milk in Tris-buffered saline PC12 cells transduced with the lentivirus GFP vector
containing 0.1% Tween 20. Immunodetection of phosphory- underwent sterile separation using the FACS to separate
lated ERK was performed overnight at 4°C using rabbit the population of labeled from the unlabeled cells (Fig. 1a).
monoclonal anti-phospho-p44/42 MAPK (extracellular signal- In a representative flow cytometric analysis of GFP-
regulated kinase—ERK; 1:1,000; Cell Signaling Technology transduced cells, 65.4% of the population was fluorescent
Inc, Danvers, MA, USA) for ERK phosphorylation (Fig. 1a—left, insert). Using appropriate gating, 55.8% of
activity. The membranes were then washed and incubated this population, characterized by very high fluorescence
for 1 h at RT with the secondary antibody/horseradish- intensity (105 fluorescent units) was collected (Fig. 1a—
peroxidase-conjugated goat anti-rabbit antibody (1:10,000, left). The population, representing 84.2% highly intense
Jackson ImmunoResearch, West Grove, PA, USA) and GFP-fluorescent cells (Fig. 1a—right) was re-analyzed.
developed using the ECL reagent (Pierce, Rockford, IL, This cell population was collected aseptically and cultured
USA), allowing visualization of the proteins. Then, the under conventional 2-D culture conditions. Stably trans-
membranes were washed and incubated for 30 min at RT fected GFP clones were selected by limited dilution. Out of
in Restore Western Blot Stripping Buffer (Pierce) and ∼500 clones, four were selected according to the following
reincubated with rabbit monoclonal anti-pan-ERK criteria: (1) ability to proliferate with a mean population
(1:2,000; Invitrogen) antibody to quantify the nonphos- doubling time of 50–80 h, (2) high intensity of GFP
phorylated protein, using a two-step procedure similar to expression, (3) “normal” PC12 morphology, (4) monolayer
that previously described (Takman et al. 2004). Quantita- formation upon adherence to collagen-coated dishes, and
tive analysis of ERK phosphorylation activity was per- (5) “normal” differentiation: neurite outgrowth in response
formed as previously described (Arien-Zakay et al. 2007). to stimulation with NGF. Each of the four clones selected
adhered to collagen and exhibited a morphology similar to
Light and Fluorescence Microscopy that of the wt-PC12 cells as exemplified by one of the
clones (Fig. 1b). Some differences between the four clones
Cell cultures were examined under an inverted Nikon were found in terms of proliferation rate, intensity of GFP
contrast microscope (Eclipse TE-2000-U and Eclipse expression, and the ability to undergo NGF-induced
TS100; Nikon, Melville, NY, USA). All images were differentiation (Table 1). The GFP-PC12-4f clone chosen
acquired digitally with either a Hamamatsu black-and-white for the present study was the one that was most similar in
high-resolution camera or a Nikon Coolpix5000 camera all the relevant criteria to the wt-PC12, expressed high and
and analyzed using Northern Eclipse software (Empix stable GFP intensity (for about 2 years upon freezing and
Imaging Inc., Mississauga, ON, Canada) and SigmaScan reculturing; Fig. 1b), and was characterized by a prolifer-
software (Sigma). GFP fluorescence was quantified with ation rate similar to that of the wt-PC12 (Fig. 1c).
the aid of a confocal laser scanning fluorescence micro-
scope (Olympus 300 IX-70, Japan and Axiovert 200, Zeiss, GFP-PC12 NGF Receptors Expression and Signaling
Göttingen, Germany), using excitation and emission wave-
lengths of 488 and 509 nm, respectively. Pictures were NGF-induced differentiation in the PC12 cells is mediated
acquired with a SensiCam digital camera (PCD, CCD by activation of the NGF receptors: TrkA and p75NTR and
Imaging, Kelhein, Germany) and processed with Image-Pro activation of downstream signaling pathways (Kaplan and
Plus 6.0 (Media Cybernetics®, Silver Spring, MD, USA). Miller 2000). Since GFP-PC12 clones were derived from
individual cells of the parental wt-PC12 cell population and
Statistics considering the large heterogeneity of NGF receptors level
in different neurons, it was extremely important to validate
Each experiment was repeated in at least duplicates three to NGF receptor expression and signaling in the GFP-PC12
four times. Where possible, the data are presented as the cells. This characterization was performed by evaluating the
mean ± SEM as evaluated using the GraphPad InStat 3 level of NGF receptor mRNA expression, NGF-induced
program (GraphPad Software Inc, San Diego, CA, USA). activation of ERK, and NGF-induced neurite outgrowth
Statistically significant differences between the experimen- under 2-D culture conditions.
tal groups were determined by analysis of variance; the mRNA expression levels of both TrkA and p75NTR
results were considered significant when p<0.05. receptors in GFP-PC12 cells were assessed using semi-
230 J Mol Neurosci (2009) 37:225–237

Figure 1 Development and


characterization of stable GFP-
PC12 clonal cells. a FACS
analysis (number of cells with
different fluorescence intensity)
and sorting of GFP-transduced
PC12 cells: left immediately
after transfection; right after 7
days of cell culture and before
cloning by limited dilution. b
Phase contrast and fluorescent
micrographs of wild-type
(wt-PC12) and green fluorescent
protein (GFP)-transduced PC12
cells after several weeks in
culture. c Time course of cell
proliferation (fold increase vs
control—day 0) of GFP-PC12
(gray) in comparison with wt-
PC12 (white) under 2-D condi-
tions, estimated by the Alamar
blue assay. The results represent
the mean ± SEM of triplicate
experiments. *p<0.01 vs day 0

quantitative RT-PCR. The results, normalized to the vs the control, respectively (p<0.01; Fig. 2b). As expected,
expression of β-actin, suggest similar levels of expression in the wt-PC12 cells, treatment with NGF significantly
for both neurotrophin receptors as found in wt-PC12 cells increased the relative phosphorylation of ERK1 and ERK2
(Fig. 2a). Activation of the TrkA receptors by NGF triggers by 11.5- and 12-fold vs the control, respectively (p<0.01;
intracellular signaling cascades, such as the ras-activated, Fig. 2b).
mitogen-activated protein kinase (MAPK) pathway, result- These results clearly indicate a significant similarity in
ing in neuronal differentiation as evidenced by neurite NGF receptor mRNA expression and NGF-induced ERK
outgrowth (Kaplan and Miller 2000). To quantitatively phosphorylation in GFP-PC12 clonal cells compared with
evaluate the NGF-induced TrkA activation in the GFP- that of the parental wt-PC12 cells.
PC12 compared with that in wt-PC12 cells, we measured
the phosphorylated form of the downstream activated Characterization of NGF-induced Differentiation
MAPK member: the extracellular signal-regulated kinase in GFP-PC12 in Comparison with that in wt-PC12 Cells
(Fig. 2b). The basal phosphorylation level of ERK1/2 was in 2-D
very low in both the untreated wt-PC12 and GFP-PC12
cells, while under NGF treatment their phosphorylation was To quantitatively assess NGF-induced differentiation of the
highly elevated in both cell lines. In the GFP-PC12 cells, GFP-PC12 cells under 2-D culture conditions, we used the
treatment with NGF significantly increased the relative same bioassay developed in our laboratory for wt-PC12
phosphorylation of ERK1 and ERK2 by 10.5- and 10-fold cells (Katzir et al. 2002). GFP-PC12 cells at low density
J Mol Neurosci (2009) 37:225–237 231

Table 1 Biological characterization of four different GFP-PC12 clones

PC12 Proliferation GFP expression Morphologyd Adherencee NGF response


clonea (doubling time, h)b (fluorescence intensity)c (% of cells with outgrowths)f

wt-PC12 48 0 Neuronal Monolayer 100


PC12-4c 50–60 103 Neuronal Monolayer 80
PC12-1d 50–60 105–106 Neuronal Monolayer 60
PC12-4d 70–80 104 Heterogeneousg Monolayer 100
PC12-4f 50–60 105 Neuronal Monolayer 100
a
PC12 cells transduced with a GFP-expressing lentiviral vector were isolated by FACS, cloned by limiting dilution into 500 wells, and grown for
1 month. Four clones were isolated and expanded. For biological characterization, samples of each clone and wild-type cells were evaluated for
different parameters over a period of several weeks.
b
Proliferation was estimated, followed daily for 5 days, using the Alamar blue method.
c
GFP expression was estimated by FACS measurement of fluorescence intensity.
d
wt-PC12 cultures contain about six different morphotypes (round cells, monopolar, bipolar, tripolar, multipolar, and a fibroblast-like
morphotype); their presence in the clones indicates plasticity similar to that of the parental cells.
e
Adherence was estimated by flattening of the cells and generation of a monolayer. Nonadherent cells grow in clusters in suspension or are
partially attached to the monolayer.
f
NGF-induced differentiation was evaluated for 3 weeks. The cells were treated with 50 ng/ml NGF, and the cultures were photographed every 3
days by phase contrast and fluorescence microscopy. Elongation of the neurites and formation of the neural networks were estimated as described
in “Materials and Methods”, using the E parameter.
g
This clone spontaneously generates neurite outgrowths.

were treated with 50 ng/ml mouse β-NGF for 7 days or left GFP-PC12 cells extended neurite outgrowths of different
untreated as control. In parallel experiments, wt-PC12 cells lengths (Fig. 3a, lower panel) similar to wt-PC12 cells. A
at the same density were also treated under identical typical dose response of the NGF-induced differentiation is
conditions. Upon NGF treatment, the majority of the presented in Fig. 3b. The similarity of the response in GFP-

Figure 2 Characterization of NGF receptor mRNA transcript expres- mNGF for 30 min or left untreated. Phosphorylation activity was
sion and activity in GFP-PC12 cells compared with that in wild-type evaluated by Western blot, using antibodies directed to the phosphor-
parental cells under 2-D conditions. a mRNA expression of TrkA and ylated (upper) or unphosphorylated (lower) ERK proteins. In the
p75 receptors in relation to mRNA expression of β-actin (internal histograms, the relative phosphorylation (%), calculated as described
control). Total RNA was extracted, RT-PCR was applied to equal in “Materials and Methods”, is presented as: white bars ERK1, gray
amounts of cDNA from each cell type using specific primers (carried bars ERK2. The results represent the mean ± SEM of triplicate
out as described in “Materials and Methods”). b NGF-induced ERK experiments.*p<0.01 vs untreated
activity in the two cell types. The cells were treated with 50 ng/ml
232 J Mol Neurosci (2009) 37:225–237

Figure 3 NGF-induced differentiation in GFP-PC12 cells under 2-D GFP-PC12 cells; white bars wt-PC12 cells. The results represent the
conditions is dose dependent and blocked by K252a. a Phase contrast mean ± SEM of triplicate experiments. *p<0.001 vs untreated
and fluorescent micrographs of GFP-PC12 cells compared with those cultures. c The inhibitory effect of 100 nM K252a, the NGF-TrkA
of wt-PC12 under 2-D cell growth conditions, upon treatment with 50 receptor antagonist, on NGF-induced differentiation of GFP-PC12
ng/ml mNGF (+NGF) or untreated (−NGF). b NGF-induced dose- cells. The results represent the mean ± SEM of triplicate experiments.
dependent differentiation response, measured by elongation of neurite *p<0.001 vs control; **p<0.001 vs NGF
outgrowths (E), as described in “Materials and Methods”; gray bars

PC12 cells vs that in wt-PC12 cells is evident, indicating GFP-PC12 Proliferation in 3-D Collagen Gels
EC50 of 2.9 and 2.6 ng/ml, respectively. A hallmark of
NGF-induced differentiation of the PC12 cells is the To evaluate the proliferation of the GFP-PC12 cells in a 3-D
inhibitory effect induced by K252a, the NGF-TrkA receptor culture venue, the cells were seeded in 3-D type I collagen
antagonist (Koizumi et al. 1988). Therefore, we evaluated gels, and their morphology was evaluated by light and
the ability of K252a to block NGF-induced differentiation fluorescence microscopy (Fig. 4). Due to their intensive,
in GFP-PC12 cells (Fig. 3c). As expected, the data clearly intrinsic (GFP) fluorescence, the GFP-PC12 cells were
indicate that 100 nM K252a inhibited by about 85% the easily detectable in a fluorescence microscope within the
NGF-induced differentiation. 3-D collagen gels (Fig. 4, upper panel). As previously
These results indicate both qualitatively and quantita- reported (Baldwin et al. 1996), in 3-D hydrogels wt-PC12
tively that NGF-induced differentiation of GFP-PC12 is cells attached to the matrix and in a few days form
very similar to that of the wt-PC12 cells. Therefore, GFP- aggregates (Fig. 4, upper panel). This process of aggrega-
PC12 cells provide a new valid model for the visualization tion, which is not observed in 2-D conditions, is probably
and imaging of neurite extension as a surrogate marker of facilitated by the mechanical forces generated by the cells
neuronal differentiation. in 3-D culture. GFP-PC12 cells grown at low density in the
J Mol Neurosci (2009) 37:225–237 233

Figure 4 Phase contrast and fluorescent micrographs of GFP-PC12 cells compared with those of wt-PC12 under 3-D cell growth conditions,
treated with 50 ng/ml mNGF (+NGF) or untreated (−NGF)

3-D collagen gels showed increased proliferation for up to extended very long and dense neurite outgrowths that
21 days (Fig. 5). Although their proliferation in 3-D culture emanated from the 3-D cell aggregates (Fig. 4, lower
conditions was somewhat slower than that of wt-PC12 as panel). Since both the small and the large neurite out-
also observed in 2-D, the increase in cell number with time growths of GFP-PC12 cells were highly visible in the 3-D
was similar to that of wt-PC12. gels, we were also able to measure their length and compare
it with NGF-induced differentiation of GFP-PC12 cells in a
NGF-induced Differentiation of GFP-PC12 Cells in 3-D 2-D environment (Fig. 3). However, due to cell aggregation
Collagen Gels in the 3-D collagen gels (Baldwin et al. 1996), the
estimation of neurite outgrowth, as the ratio between
When grown in collagen gels in the presence of 50 ng/ml neurite length and cell diameter, using the commonly
NGF for up to 14 days, both wt-PC12 and GFP-PC12 cells available 2-D method is not accurate. Furthermore, out-
growths of many neurites in three dimensions made it
difficult to trace them from individual cells. This problem
required the adaptation of a novel measurement assay of
NGF-induced differentiation under 3-D conditions. For this
purpose, we applied NGF-induced differentiation using a
fractal dimension (Df) method used for assessing blood
capillary sprouting and network formation in vitro and in
vivo (Kirchner et al. 1996; Lazarovici et al. 2006). In the
first step, we validated the method by comparing the E and
Df parameters from measurements of the same images upon
2 days of NGF treatment, at the beginning of the
differentiation process (Fig. 6a). A correlation coefficient
of 0.94 of the differentiation effects measured by these two
methods indicates a high similarity in neurite outgrowth
elongation estimated by the fractal dimension method and
the elongation method. Furthermore, the Df parameter was
checked for its ability to reflect a dose-dependent elonga-
Figure 5 Time course of cell proliferation (fold increase vs control— tion of neurite outgrowth (Fig. 6b). By increasing the NGF
day 0) of GFP-PC12 (gray) in comparison with that of wt-PC12
concentration, the differentiation process was enhanced, as
(white) under 3-D conditions, estimated by the Alamar blue assay. The
results represent the mean ± SEM of triplicate experiments. *p<0.01 evident from the parallel change in Df compared with the E
vs day 0; **p<0.05 vs wt-PC12 at the same time point parameter. We then compared NGF-induced differentiation
234 J Mol Neurosci (2009) 37:225–237

Figure 6 Validation of NGF-induced differentiation of GFP-PC12 tion responses, analyzed by Df and E. Upper panel Representative
cells under 2-D conditions by comparing the fractal dimension pictures of NGF-induced neural networks and their representative
parameter (Df) with the elongation parameter (E). E and Df were fractal dimension images. Lower panel Comparison between the dose-
calculated as described in “Materials and Methods”. a The relationship dependent differentiation responses measured using Df and E. The
between Df and E with a correlation coefficient of 0.94 was calculated values represent the mean ± SEM of quadruplicate experiments. *p<
from the regression line. b NGF-induced dose-dependent differentia- 0.01 vs untreated cells

of GFP-PC12 cells cultured in 2-D and 3-D conditions, using Discussion


the Df method. Our data suggest that at 8 days the cells
maintained in 2-D underwent a 3.5-fold more pronounced Neurons, with their long axons and elaborate dendritic
differentiation that those cultured in 3-D (Fig. 7). After arborization, establish the complex circuitry that is essential
15 days of treatment, NGF induced similar degrees of to the proper functioning of the nervous system. A list of
differentiation in both 2-D and 3-D cultures. Extension of structural, molecular, and functional differences between
the culture period to 45 days did not further enhance NGF- axons and dendrites is emerging. However, the mechanisms
induced differentiation of GFP-PC12 cells in 3-D beyond involved in early events of neuronal differentiation, such as
that observed at day 15. neurite initiation and elongation, are less well understood,
mainly because of a lack of quantitative methods to
accurately measure neuronal differentiation. In tissue
engineering research, fluorescent cells derived from GFP-
transgenic rodents or engineered in vitro to express the GFP
transgene have been recently used to elucidate the cell–
microenvironment interaction (Tan et al. 2007), for both
regeneration and repair purposes (Boldrin et al. 2007; Sales
et al. 2007), and to study bone differentiation (Zhou et al.
2006). We, therefore, continued this research approach and
developed a GFP-PC12 cell model suitable for studying
neuronal differentiation in a 3-D microenvironment.
In contrast to some molecular approaches in which the
GFP transgene is targeted to the mitochondria (Sirk et al.
2003) or coupled to a specific intracellular protein (Niell
and Smith 2004), in the present study, the GFP transgene
Figure 7 NGF-induced differentiation of GFP-PC12 cells under 3-D protein accumulated in the cytosol of the green PC12 cells,
(gray) conditions is delayed compared with those under 2-D (white) as previously shown for hematopoietic cells (Miyoshi et al.
conditions as measured by Df. Df was calculated as described in
1999). This enabled both intensive visualization of the
“Materials and Methods”. The values represent the mean ± SEM of
triplicate experiments. *p<0.01 vs untreated cells; **p<0.05 vs 2-D perikarya and the neurite outgrowth cytoplasm of very
conditions at the same time point small neurite branches. These GFP-PC12 cells similar to
J Mol Neurosci (2009) 37:225–237 235

wt-PC12 cells under 2-D conditions proliferated, expressed studies provided preliminary, qualitative evidence that
NGF receptors, and activated the ERK pathway in response neurite outgrowth and branching of PC12 cells was
to NGF. They also showed NGF-induced differentiation, a inhibited with decreasing substrate rigidity (Leach et al.
process inhibited by K252a, the NGF-TrkA receptor 2007). On the other hand, attachment, morphology, and
antagonist. functions of PC12 cells were superior in 3-D compared to
Under 3-D conditions, GFP-PC12 cells also proliferate, 2-D culture conditions (Chia et al. 2005). These observa-
form aggregates (Baldwin et al. 1996), and respond to NGF tions deserve further careful quantitative studies, for which
by neurite outgrowths of different lengths and complexity, the present GFP-P12 cells might be well suited.
again very similar to wt-PC12 cells. Since the techniques In the past, NGF-induced neuronal differentiation, in
for quantifying the NGF-induced differentiation in 2-D particular neurite outgrowth, has been assessed qualitatively
were not applicable for 3-D conditions, we employed and in 3-D cultures using PC12 cells cultured on a variety of
validated a novel approach for measuring neurite outgrowth chemically tailored biomaterials (Pittier et al. 2005; Yu
based on fractal dimension (Df). This method was validated et al. 1999; Holmes et al. 2000) and polymers modified
by the 2-D method in which the elongation parameter (E) is with integrin-binding motifs (Park and Yun 2004). These
measured and used to quantify NGF-induced differentiation studies emphasized the enhanced biocompatibility of these
of GFP-PC12 cells in 2-D by dose response and in 3-D by biomaterials and their prospects for neural regeneration.
time course. At day 8, NGF-induced differentiation of GFP- However, to be suited for clinical neuroregenerative
PC12 cells in 3-D collagen gels was significantly retarded therapy, such designed 3-D matrices will require quantita-
compared to cells cultured in 2-D on collagen-coated tive evaluation of their differentiative properties using novel
surfaces. By contrast after 15 days, neurite extension in analytical tools such as the Df method in conjunction with
both 2-D and 3-D was statistically indistinguishable (Fig. 7). GFP-PC12 cells introduced by this study.
Two plausible explanations for the delayed NGF-induced We expect that this GFP-PC12 cell model will continue
elongation of the neurites in 3-D vs 2-D conditions are to serve in the development of novel bioactive matrices,
proposed: (a) the relative high stiffness rendered by such as electrically conductive polymers for enhanced
collagen gels (1.5 mg/ml), known to generate about 15–17 neuronal differentiation (Guterman et al. 2002; Guo et al.
Pa (Willits and Skornia 2004), inhibited neurite elongation; 2007; Gomez and Schmidt 2007), for studying neuro-
and (b) the process of metaloprotease secretion by the cells, trophin-induced differentiation in/on 3-D polymeric scaf-
known to generate the necessary space in the gel by folds (Levenberg et al. 2005), and for in vitro evaluation of
collagen hydrolysis, required for the elongation of the pharmacologically active microcarriers releasing NGF or
neurite outgrowths, was limited. Indeed, PC12 cells over- other drugs conveying PC12 cells (Tatard et al. 2004). The
expressing the tissue plasminogen activator, which pro- NGF-induced differentiation of GFP neurons, in conjunc-
motes proteolytic degradation of the matrix, grow neurites tion with near-infrared multiphoton microscopy and in vivo
to a greater extent than do control cells (Pittman and laser tomography, should provide novel models for drug
DiBenedetto 1995). screening and high-resolution structural imaging (Schenke-
Since collagen gels and other hydrogel scaffolds are Layland et al. 2006) for measurements of neuronal
relatively translucent, it is feasible to visualize by confocal differentiation in situ.
fluorescent microscopy the morphological differentiation of
GFP-expressing neurons inside the gels and to quantify Acknowledgments This study was supported by grants from the
Stein Family Foundation, Philadelphia, PA (PIL and PL), the
neuritogenesis using the Df method, thus avoiding conven- Nanotechnology Institute of Southeastern Pennsylvania (PIL), and
tional time-consuming histological methods. Over the last the United States–Israel Binational Science Foundation (PL). PL is
decade, the genes for GFP and other color proteins have affiliated with and supported in part by the David R. Bloom Center for
been cloned and used for biological purposes (Suzuki et al. Pharmacy and the Dr. Adolf and Klara Brettler Center for Research in
Molecular Pharmacology and Therapeutics at The Hebrew University
2007), providing an elegant approach which facilitates of Jerusalem, Israel. SL is supported by an “Eshkol” fellowship from
concomitant measurement of multiple proteins involved in The Israel Ministry of Science, Culture and Sport.
the NGF-induced differentiation under 3-D conditions.
Rationally designed matrices for neural engineering and
cell therapy rely on a comprehensive understanding of 3-D
cellular differentiation induced by neurotrophins, such as References
NGF. Under these conditions, both biochemical and matrix
mechanical cues (such as stiffness) will influence neuronal Arien-Zakay, H., Nagler, A., Galski, H., & Lazarovici, P. (2007).
Neuronal conditioning medium and nerve growth factor induce
differentiation. In extending previous work with wt-PC12
neuronal differentiation of collagen-adherent progenitors derived
cells (Saltzman et al. 1992), the present GFP-PC12 cell from human umbilical cord blood. Journal of Molecular
model may be suitable to address this issue. Indeed, several Neuroscience, 32, 179–191. doi:10.1007/s12031-007-0027-2.
236 J Mol Neurosci (2009) 37:225–237

Baldwin, S. P., Krewson, C. E., & Saltzman, W. M. (1996). PC12 cell Laketa, V., Simpson, J. C., Bechtel, S., Wiemann, S., & Pepperkok, R.
aggregation and neurite growth in gels of collagen, laminin and (2007). High-content microscopy identifies new neurite out-
fibronectin. International Journal of Developmental Neuroscience, growth regulators. Molecular Biology of the Cell, 18, 242–252.
14, 351–364. doi:10.1016/0736-5748(96)00018-4. doi:10.1091/mbc.E06-08-0666.
Bieberich, E., & Anthony, G. E. (2004). Neuronal differentiation and Lazarovici, P., Gazit, A., Staniszewska, I., Marcinkiewicz, C., &
synapse formation of PC12 and embryonic stem cells on Lelkes, P. I. (2006). Nerve growth factor (NGF) promotes
interdigitated microelectrode arrays: Contact structures for angiogenesis in the quail chorioallantoic membrane. Endothelium,
neuron-to-electrode signal transmission (NEST). Biosensors & 13, 51–59. doi:10.1080/10623320600669053.
Bioelectronics, 19, 923–931. doi:10.1016/j.bios.2003.08.016. Leach, J. B., Brown, X. Q., Jacot, J. G., Dimilla, P. A., & Wong, J. Y.
Boldrin, L., Elvassore, N., Malerba, A., et al. (2007). Satellite cells (2007). Neurite outgrowth and branching of PC12 cells on very
delivered by micro-patterned scaffolds: A new strategy for cell soft substrates sharply decreases below a threshold of substrate
transplantation in muscle diseases. Tissue Engineering, 13, 253– rigidity. Journal of Neural Engineering, 4, 26–34. doi:10.1088/
262. doi:10.1089/ten.2006.0093. 1741-2560/4/2/003.
Chao, M., Casaccia-Bonnefil, P., Carter, B., Chittka, A., Kong, H., & Levenberg, S., Burdick, J. A., Kraehenbuehl, T., & Langer, R. (2005).
Yoon, S. O. (1998). Neurotrophin receptors: Mediators of life and Neurotrophin-induced differentiation of human embryonic stem
death. Brain Research. Brain Research Reviews, 26, 295–301. cells on three-dimensional polymeric scaffolds. Tissue Engineering,
doi:10.1016/S0165-0173(97)00036-2. 11, 506–512. doi:10.1089/ten.2005.11.506.
Chia, S. M., Lin, P. C., Quek, C. H., et al. (2005). Engineering Lopez, C. A., Fleischman, A. J., Roy, S., & Desai, T. A. (2006).
microenvironment for expansion of sensitive anchorage-dependent Evaluation of silicon nanoporous membranes and ECM-based
mammalian cells. Journal of Biotechnology, 118, 434–447. microenvironments on neurosecretory cells. Biomaterials, 27,
doi:10.1016/j.jbiotec.2005.05.012. 3075–3083. doi:10.1016/j.biomaterials.2005.12.017.
Dietrich, F., & Lelkes, P. I. (2006). Fine-tuning of a three-dimensional Mahoney, M. J., Chen, R. R., Tan, J., & Saltzman, W. M. (2005). The
microcarrier-based angiogenesis assay for the analysis of endo- influence of microchannels on neurite growth and architecture.
thelial–mesenchymal cell co-cultures in fibrin and collagen gels. Biomaterials, 26, 771–778. doi:10.1016/j.biomaterials.2004.03.015.
Angiogenesis, 9, 111–125. doi:10.1007/s10456-006-9037-x. Miyoshi, H., Smith, K. A., Mosier, D. E., Verma, I. M., & Torbett,
Foley, J. D., Grunwald, E. W., Nealey, P. F., & Murphy, C. J. (2005). B. E. (1999). Transduction of human CD34+ cells that mediate
Cooperative modulation of neuritogenesis by PC12 cells by long-term engraftment of NOD/SCID mice by HIV vectors.
topography and nerve growth factor. Biomaterials, 26, 3639– Science, 283, 682–686. doi:10.1126/science.283.5402.682.
3644. doi:10.1016/j.biomaterials.2004.09.048. Mondrinos, M. J., Koutzaki, S., Lelkes, P. I., & Finck, C. M. (2007).
Fujita, K., Lazarovici, P., & Guroff, G. (1989). Regulation of the A tissue-engineered model of fetal distal lung tissue. American
differentiation of PC12 pheochromocytoma cells. Environmental Journal of Physiology. Lung Cellular and Molecular Physiology,
Health Perspectives, 80, 127–142. doi:10.2307/3430738. 293, L639–L650. doi:10.1152/ajplung.00403.2006.
Gomez, N., & Schmidt, C. E. (2007). Nerve growth factor- Moxon, K. A., Hallman, S., Aslani, A., Kalkhoran, N. M., & Lelkes,
immobilized polypyrrole: Bioactive electrically conducting polymer P. I. (2007). Bioactive properties of nanostructured porous silicon
for enhanced neurite extension. Journal of Biomedical Materials for enhancing electrode to neuron interfaces. Journal of Bio-
Research. Part A, 81, 135–149. doi:10.1002/jbm.a.31047. materials Science. Polymer Edition, 18, 1263–1281. doi:10.1163/
Guo, Y., Li, M., Mylonakis, A., et al. (2007). Electroactive oligoani- 156856207782177882.
line-containing self-assembled monolayers for tissue engineering Niell, C. M., & Smith, S. J. (2004). Live optical imaging of nervous
applications. Biomacromolecules, 8, 3025–3034. doi:10.1021/ system development. Annual Review of Physiology, 66, 771–798.
bm070266z. doi:10.1146/annurev.physiol.66.082602.095217.
Guterman, E., Cheng, S., Palouian, K., Bidez, P. R., Lelkes, P. I., & Nikolaychik, V. V., Samet, M. M., & Lelkes, P. I. (1996). A new
Wei, Y. (2002). Peptide-modified electroactive polymers for method for continual quantitation of viable cells on endothelial-
tissue engineering applications. Polymer Preprints, 43, 766–767. ized polyurethanes. Journal of Biomaterials Science. Polymer
Holmes, T. C., de Lacalle, S., Su, X., Liu, G., Rich, A., & Zhang, S. Edition, 7, 881–891. doi:10.1163/156856296X00057.
(2000). Extensive neurite outgrowth and active synapse forma- Okabe, M., Ikawa, M., Kominami, K., Nakanishi, T., & Nishimune, Y.
tion on self-assembling peptide scaffolds. Proceedings of the (1997). ‘Green mice’ as a source of ubiquitous green cells. FEBS
National Academy of Sciences of the United States of America, Letters, 407, 313–319. doi:10.1016/S0014-5793(97)00313-X.
97, 6728–6733. doi:10.1073/pnas.97.12.6728. Park, K. H., & Yun, K. (2004). Immobilization of Arg-Gly-Asp
Kaplan, D. R., & Miller, F. D. (2000). Neurotrophin signal (RGD) sequence in a thermosensitive hydrogel for cell delivery
transduction in the nervous system. Current Opinion in Neuro- using pheochromocytoma cells (PC12). Journal of Bioscience
biology, 10, 381–391. doi:10.1016/S0959-4388(00)00092-1. and Bioengineering, 97, 374–377.
Katzir, I., Shani, J., Regev, K., Shabashov, D., & Lazarovici, P. (2002). Pittier, R., Sauthier, F., Hubbell, J. A., & Hall, H. (2005). Neurite
A quantitative bioassay for nerve growth factor, using PC12 clones extension and in vitro myelination within three-dimensional
expressing different levels of trkA receptors. Journal of Molecular modified fibrin matrices. Journal of Neurobiology, 63, 1–14.
Neuroscience, 18, 251–264. doi:10.1385/JMN:18:3:251. doi:10.1002/neu.20116.
Kirchner, L. M., Schmidt, S. P., & Gruber, B. S. (1996). Quantitation Pittman, R. N., & DiBenedetto, A. J. (1995). PC12 cells over-
of angiogenesis in the chick chorioallantoic membrane model expressing tissue plasminogen activator regenerate neurites to a
using fractal analysis. Microvascular Research, 51, 2–14. greater extent and migrate faster than control cells in complex
doi:10.1006/mvre.1996.0002. extracellular matrix. Journal of Neurochemistry, 64, 566–575.
Koizumi, S., Contreras, M. L., Matsuda, Y., Hama, T., Lazarovici, P., Ravni, A., Bourgault, S., Lebon, A., et al. (2006). The neurotrophic
& Guroff, G. (1988). K-252a: A specific inhibitor of the action of effects of PACAP in PC12 cells: Control by multiple transduction
nerve growth factor on PC 12 cells. The Journal of Neuroscience, pathways. Journal of Neurochemistry, 98, 321–329. doi:10.1111/
8, 715–721. j.1471-4159.2006.03884.x.
Kosaka, Y., Kobayashi, N., Fukazawa, T., et al. (2004). Lentivirus- Sales, V. L., Mettler, B. A., Lopez-Ilasaca, M., Johnson Jr, J. A., &
based gene delivery in mouse embryonic stem cells. Artificial Mayer Jr., J. E. (2007). Endothelial progenitor and mesenchymal
Organs, 28, 271–277. doi:10.1111/j.1525-1594.2004.47297.x. stem cell-derived cells persist in tissue-engineered patch in vivo:
J Mol Neurosci (2009) 37:225–237 237

Application of green and red fluorescent protein-expressing glucose deprivation-induced c-Jun amino-terminal kinase 1 and
retroviral vector. Tissue Engineering, 13, 525–535. doi:10.1089/ stress-activated kinases p38alpha and p38beta activation and
ten.2006.0128. confers neuroprotection in the pheochromocytoma PC12 Model.
Saltzman, W. M., Parkhurst, M. R., Parsons-Wingerter, P., & Zhu, Journal of Molecular Neuroscience, 22, 237–250. doi:10.1385/
W. H. (1992). Three-dimensional cell cultures mimic tissues. JMN:22:3:237.
Annals of the New York Academy of Sciences, 665, 259–273. Tan, W., Vinegoni, C., Norman, J. J., Desai, T. A., & Boppart, S. A.
doi:10.1111/j.1749-6632.1992.tb42590.x. (2007). Imaging cellular responses to mechanical stimuli within
Schenke-Layland, K., Riemann, I., Damour, O., Stock, U. A., & three-dimensional tissue constructs. Microscopy Research and
Konig, K. (2006). Two-photon microscopes and in vivo Technique, 70, 361–371. doi:10.1002/jemt.20420.
multiphoton tomographs—Powerful diagnostic tools for tissue Tatard, V. M., Venier-Julienne, M. C., Benoit, J. P., Menei, P., &
engineering and drug delivery. Advanced Drug Delivery Reviews, Montero-Menei, C. N. (2004). In vivo evaluation of pharmaco-
58, 878–896. doi:10.1016/j.addr.2006.07.004. logically active microcarriers releasing nerve growth factor and
Simons, D. M., Gardner, E. M., & Lelkes, P. I. (2006). Dynamic conveying PC12 cells. Cell Transplantation, 13, 573–583.
culture in a rotating-wall vessel bioreactor differentially inhibits doi:10.3727/000000004783983675.
murine T-lymphocyte activation by mitogenic stimuli upon return Tohill, M. P., Mann, D. J., Mantovani, C. M., Wiberg, M., & Terenghi,
to static conditions in a time-dependent manner. Journal of G. (2004). Green fluorescent protein is a stable morphological
Applied Polymer Science, 100, 1287–1292. doi:10.1152/japp marker for Schwann cell transplants in bioengineered nerve
lphysiol.00887.2005. conduits. Tissue Engineering, 10, 1359–1367.
Sirk, D. P., Zhu, Z., Wadia, J. S., & Mills, L. R. (2003). Flow Vaudry, D., Stork, P. J., Lazarovici, P., & Eiden, L. E. (2002). Signaling
cytometry and GFP: A novel assay for measuring the import and pathways for PC12 cell differentiation: Making the right connec-
turnover of nuclear-encoded mitochondrial proteins in live PC12 tions. Science, 296, 1648–1649. doi:10.1126/science.1071552.
cells. Cytometry. Part A, 56, 15–22. Willits, R. K., & Skornia, S. L. (2004). Effect of collagen gel stiffness
Suzuki, T., Matsuzaki, T., Hagiwara, H., Aoki, T., & Takata, K. on neurite extension. Journal of Biomaterials Science. Polymer
(2007). Recent advances in fluorescent labeling techniques for Edition, 15, 1521–1531. doi:10.1163/1568562042459698.
fluorescence microscopy. Acta Histochemica et Cytochemica, 40, Yu, X., Dillon, G. P., & Bellamkonda, R. B. (1999). A laminin and
131–137. doi:10.1267/ahc.07023. nerve growth factor-laden three-dimensional scaffold for en-
Takezawa, T., Takeuchi, T., Nitani, A., et al. (2007). Collagen vitrigel hanced neurite extension. Tissue Engineering, 5, 291–304.
membrane useful for paracrine assays in vitro and drug delivery doi:10.1089/ten.1999.5.291.
systems in vivo. Journal of Biotechnology, 131, 76–83. Zhou, G., Liu, W., Cui, L., Wang, X., Liu, T., & Cao, Y. (2006).
doi:10.1016/j.jbiotec.2007.05.033. Repair of porcine articular osteochondral defects in non-weight-
Takman, R., Jiang, H., Schaefer, E., Levine, R. A., & Lazarovici, P. bearing areas with autologous bone marrow stromal cells. Tissue
(2004). Nerve growth factor pretreatment attenuates oxygen and Engineering, 12, 3209–3221. doi:10.1089/ten.2006.12.3209.

You might also like