Advancing Neurodegenerative Disease Research With iPSCs
eBook
Published: October 24, 2025
Credit: Sartorius
The growing burden of neurodegenerative diseases has intensified the need for translational models that replicate human biology more accurately. Traditional approaches often fail to predict clinical outcomes due to poor relevance and reproducibility.
Induced pluripotent stem cells (iPSCs) now offer a transformative path toward modeling complex disease mechanisms and testing potential therapeutics in human-relevant systems.
This eBook explores the latest advances in iPSC-derived models, highlighting how they bridge preclinical and clinical research to accelerate discovery in neurodegenerative disease research.
Download this eBook to discover:
- How iPSCs enhance disease modeling accuracy and therapeutic prediction
- Methods for integrating complex cell models and live-cell analysis
- Insights into future applications of iPSC-based neuroscience research
The Progress and
Promise of iPSCs
in Disease Modeling
Simplifying Progress
The Burden of Gradual Decay
Neurodegenerative diseases represent a group of
conditions marked by the progressive decline of neuron
structure and function. These diseases are particularly
harrowing as they lead to a gradual deterioration of motor
skills, memory, and cognitive abilities, profoundly affecting
patient lives.
Alzheimer’s and Parkinson’s diseases are the most common
neurodegenerative disorders, each presenting unique
challenges. Alzheimer’s disease gradually erodes memory
and cognitive function, while Parkinson’s disease is known
for its motor symptoms, such as tremors and stiffness. The
burden of these neurodegenerative disorders is immense,
not only in terms of the number of people affected, but also
in the impact on families and the healthcare system. The
Alzheimer’s Association estimates that 6.7 million Americans
aged 65 and older live with Alzheimer’s dementia. In
contrast, Parkinson’s disease affects nearly one million
people in the United States, according to the Parkinson’s
Foundation.
Current treatment options for neurodegenerative diseases
are limited. Medications approved for symptom
management provide only temporary relief and do not
address the underlying disease progression. For example,
cholinesterase inhibitors in Alzheimer’s may temporarily
improve memory and judgment, but they do not prevent
neuronal death. Similarly, dopamine precursors can alleviate
symptoms in Parkinson’s but do not stop motor loss and
cellular deterioration. This underscores the urgent need for
therapies that target the pathogenesis of these diseases.
Barriers to Progress
One of the major hurdles in developing such therapies is
the restrictive nature of the blood-brain barrier (BBB). The
BBB serves as a protective shield, preventing nearly 99% of
substances from entering the brain. While this is crucial for
brain health, it also poses a significant challenge for drug
delivery, as most therapeutic agents cannot cross the BBB
to reach their targets in the central nervous system.
Moreover, the lack of accurate translational models that
mimic human neurodegenerative diseases complicates
research. Primary neuronal cultures, acquired postmortem,
are restricted by availability, a short lifespan, and
meticulous handling protocols. Immortalized cell lines,
while easier to maintain, often lack the complex behaviors
of true neurons, are prone to genetic changes, and do not
replicate the brain’s three-dimensional structure.
Alternatively, animal models can be poor predictors of
treatment response in humans, leading to a high failure
rate in clinical trials. Researchers are addressing these
limitations by embracing advanced cell culture systems.
Exploring New Dimensions in Neuroscience
with Advanced Cell Models
Table of Contents
Application Note
Solutions for the Culturing, Maintaining
and Characterization of Induced
Pluripotent Stem Cells
Daryl Cole, PhD — Scientist - Sartorius
Kirsty McBain — Scientist - Sartorius
Nicola Bevan — Manager - Sartorius
Introduction
From drug discovery to organoid modeling of disease, stem
cells are increasingly being used in research as a vital tool
for scientific investigation. The current trend away from
animal models and the push to more relevant systems for
simulating the human body require flexible and specific
tools to achieve this goal. Induced Pluripotent Stem Cells
(iPSCs) are produced from normal tissue, through the
forced expression of key transcription factors1, providing a
limitless supply of these precious cells for research and
development. Due to the very specialized nature of these
cells, their maintenance and culture is more intensive than
most cell lines. For this reason, it is important that solutions
for the culture and maintenance of these cell types are
readily and widely available. Characterization of stem cells
can be difficult and unreliable, depending on the
methodology used, which is why it is important to develop
robust techniques for monitoring stem cells throughout
culture and experimental testing. If conditions are not
optimal during the maintenance of iPSCs, their pluripotency
can be lost.
Reproducibility is highly prized in research and automated
solutions can provide high levels of consistency in method
and data generation. The CellCelector Flex is an
automated platform for targeted cell identification and
picking that is not only highly accurate, but also very
gentle on cells, providing an ideal solution when working
with delicate iPSCs. The Incucyte® Live-Cell Analysis
platform automates the imaging processes of iPSC
workflows, allowing cells to be monitored over time to
analyze changes in morphology and colony formation from
within the incubator. This limits the disturbance to
precious iPSC culture plates, but also enables real-time
tracking of cell growth and health metrics.
Key words or phrases:
Induced Pluripotent Stem Cells, iPSCs, Pluripotency,
Stem Cells, CellCelector Flex, Incucyte, iQue, Cell
Culture, iPSC Characterization, iPSC R&D
Find out more: www.sartorius.com/ipscs
Further characterization of iPSCs can be performed on
the iQue®3 High-Throughput Cytometry Platform,
investigating changes in expression of pluripotency
markers integral to maintaining stemness, providing an
overview of the status of iPSCs.
Many traditional methods for culturing, monitoring and
characterizing iPSCs can:
1. Be inconsistent and unreliable, resulting in seeded
populations with high levels of heterogeneity, cell death
and differentiation
2. Require regular disturbance of culture plates to
monitor growth and confluency, with no integrated
options for analysis
3. Demand large volumes of precious sample for analysis,
resulting in less material for downstream applications
4. Necessitate the use of a variety of techniques to measure
multiple characteristics
This application note discusses the novel solutions
provided by Sartorius platforms for the culture,
maintenance, and characterization of iPSCs, during
research and development.
Methods
The following methods outline a flexible, in-depth
workflow for growth and characterization of iPSCs using
multiple Sartorius platforms.
Cell Culture and Maintenance
Picking and seeding iPSCs
Individual cells and colonies were picked using the
CellCelector Flex with the Adherent Colony Picking Module
and seeded into tissue culture plates for further expansion
and downstream processing. Images were taken prior to and
post picking to monitor and record the effects of colony
manipulation using the CellCelector Flex. Propidium Iodide
(PI) staining was undertaken on iPSC colonies after seeding
by adding PI at a concentration of 500 nM and incubating
for 3 minutes, rinsing twice with PBS and resuspending in
growth medium (mTESR Plus) for imaging.
Figure 1. Schematic showcasing the use of Sartorius platforms in iPSC culture.
Using the three Sartorius instruments, CellCelector Flex, Incucyte® and iQue®3, iPSCs can be picked and seeded, pluripotency tested,
and growth and confluency monitored.
Picking and Isolating cells
CellCelector Flex Incucyte® Live-Cell
Analysis Platform
iQue®3 High-Throughput
Screening Cytometer
Monitoring, morphology,
confluency and growth
Characterization of
marker expression
Thawing and Culturing iPSCs
Cells (ATCC-DYS0100 cells derived from human foreskin
fibroblasts) were thawed and plated onto Vitronectin XF™
(1:25 dilution in CellAdhere™ Dilution Buffer) precoated
6-well plates at a seeding density of 1x106 cells/well in 1 mL
growth medium (mTESR™ Plus) supplemented with
Y-27632 (ROCK inhibitor, 10 μM) and incubated at 37°C.
iPSCs were monitored using the Incucyte® system to
assess confluency, colony formation, and general cell
morphology and health. The confluence of colonies was
analyzed using the integrated Incucyte® AI confluence
Characterization and Monitoring of Pluripotency
Pluripotency Characterization: iQue®
iPSCs were dissociated to single cells during passage and at
specified timepoints using Gentle Cell Dissociation Reagent.
Single cell suspensions were stained with cell surface marker
antibodies (in PBS + 2% FBS) for one non-pluripotent marker,
SSEA-1, and two pluripotency markers, SSEA-4 and TRA-1-
60, in addition to the iQue® Membrane Integrity (B/Red)
Dye, for viability analysis. Cells were seeded at 2x104 cells/well
in a V-bottom 96-well plate and stained with the cocktail of
Monitoring Pluripotency and Cell Health: Incucyte®
During the experiments, iPSCs were monitored for
changes in morphology and confluency using the
Incucyte® Live-Cell Analysis platform. Cultured iPSCs
lines were monitored by high definition (HD) phase
contrast at 4-hour intervals using a repeating scan
schedule at 10X. Nuclear to cytoplasmic ratios were
Intracellular and Surface Marker Studies
iPSC and control THP-1 cells were seeded at 2x104 cells/
well in a V-bottom 96-well plate and fixed, permeabilized
and stained according to the protocol found in the
following tech note: Intracellular Staining Assay for iQue®
Platform. Pluripotency markers, SSEA-4, TRA-1-60, Oct
Results
Developing workflows for the culture and
characterization of stem cells such as iPSCs is vital in
producing consistent, reproducible and robust data.
Using the Sartorius platforms showcased here (Figure
1), we can highlight the benefits of the approaches
described for culturing iPSCs that are healthy and
pluripotent while monitoring and characterizing these
stem cells for key markers of health and stemness.
software algorithm. Passages were performed every 3-4
days at approximately 60-70% confluence using Gentle
Cell Dissociation Reagent and replated at 1x105 cells/well.
Medium changes were performed daily during the week,
while double volume medium changes were performed on
Friday to account for no medium changes over the
weekend. For the non-optimized iPSC culture, cells were
grown as above except using RPMI 1640 medium
supplemented with 10% FBS, L-glutamine 2 mM,
Penicillin/Streptomycin 100 μg/mL.
antibodies described (RT in the dark for 30 minutes). To wash
plates, PBS + 2% FBS (100 μl) was added, prior to
centrifugation (300 x g, 5 minutes), then aspirated. Plates
were shaken (3000 rpm, 60 seconds) and the samples
resuspended in PBS + 2% FBS (20 μL), prior to being
analyzed on the iQue®3. Analysis of data was performed
using the iQue Forecyt® software after compensation had
been optimized for each of the antibodies.
calculated by staining iPSC nuclei using the Incucyte®
Nuclight Rapid Red Dye (1:1000) and measuring the
cytoplasmic area (confluence mask) and the nuclear
area (fluorescence mask) using basic masking to
quantify pluripotency/normal iPSC morphology.
3/4 and Sox-2 were analyzed, while SSEA-1 expression was
used as a marker for non-pluripotency. Analysis was
performed on the iQue Forecyt® software after
compensation had been optimized for each of the
antibodies.
Picking iPSCs Using the CellCelector Flex Is Fast, Gentle
and Reliable
It is important when working with any cell system, but
notably stem cells such as iPSCs, to maintain good cell
health. The data here highlights the delicate, gentle picking
and seeding capability of the CellCelector Flex. When
stained with Propidium Iodide (PI), a stain that indicates cell
death, manual manipulation of iPSCs produces an
Figure 2. Picking iPSCs using the CellCelector Flex is accurate, fast, gentle and reliable.
Micrographs taken using the CellCelector platform highlighting iPSC colonies selected by the system. (A) Manually and (B) CellCelector picked
and seeded iPSC colony stained with propidium iodide (PI) to identify cell death. (C) Micrograph depicting an area of differentiation in a stem cell
colony prior to picking with the CellCelector. (D) The same area of the culture plate shown in (C) after removal. (E) Micrograph of a large iPSC
colony grown on a feeder layer, prior to picking a section of pluripotent cells. The bottom right of the colony has indications of spontaneous
differentiation. (F) The colony in (E) after picking using the CellCelector Flex, the area of pluripotent cells targeted by the machine has been
collected for further culture. Scale bar equals 500 μm.
increased number of PI positive cells when compared to
the CellCelector Flex, indicative of fewer healthy cells
(Figure 2A). The CellCelector Flex colony also has less
debris and more tightly defined borders (Figure 2B).
The flexibility and power of the CellCelector Flex is
exemplified by its capabilities, it is able to pick single iPSCs
or whole iPSC colonies from a tissue culture plate. This
provides the opportunity to select ideal colonies from
cultures on a standard plate for further propagation.
Additionally, portions of colonies can be selected for further
culture. This is useful if a portion of the colony
spontaneously differentiates. Differentiated sections can be
removed or pluripotent sections can be picked for
passaging or analysis (Figure 2C-F).
Propidium iodide Propidium iodide
C. Differentiated Section
E. Pluripotent Section
A. Manual B. CellCelector Flex
D. Removal of Section
F. Removal of Section
Figure 3. Monitoring morphology and pluripotent potential during iPSC culture.
Incucyte® images of iPSCs grown under optimized (mTESR Plus) and non-optimized (RPMI)
conditions. (A, D) Fluorescent images of iPSCs stained with Nuclight Rapid Red Dye comparing
nuclear density between conditions. (B, E) Phase contrast images of the same iPSCs showing
morphological differences between the two variables. (C, F) Analysis masking on the Incucyte®
depicting confluency and nuclear masking that can be used to determine the nuclear/
cytoplasm ratio illustrated in (G). Scale bar equals 400 μm.
total nuclei area =nuclear/cytoplasm ratio
total cytoplasmic area
Monitoring Morphology and Pluripotent Potential
During iPSC Culture
The CellCelector Flex can be used within the same
workflow as another Sartorius platform, the Incucyte® Live-
Cell Analysis platform. This system provides tools for
monitoring cells during culture within the incubator, so
changes in morphology can be recorded and analyzed
without requiring removal of culture plates. In the following
case, losses in morphological indicators of pluripotency can
be observed, recorded, and subsequent analysis can be
performed to quantify these changes.
Incucyte® images of iPSCs after 2 days in culture, show a
marked difference in morphology between the optimized
and non-optimized culture conditions. iPSCs grown in
optimized conditions form tightly packed colonies with
clearly defined edges, that ‘glow’ under phase images
(Figure 3B), by contrast, non-optimized iPSCs are much
more spread out and no longer form tightly packed
colonies, they are beginning to resemble fibroblast cells
(Figure 3E). Nuclear staining using Incucyte® Nuclight
Rapid Red Dye also highlights the separation of the cells
when grown in non-optimized conditions (Figure 3D),
nuclei are much more spread out and lose the tight
distribution found in optimized conditions (Figure 3A).
Quantification of these morphological differences was
performed using the Incucyte® Adherent Cell-by-Cell
scan at 10X magnification and nuclear and cytoplasm
area measurements were made using the Basic Analyzer
and AI Confluence analysis (micrographs in Figure 3C, F)
using the following equation to provide a nuclear/
cytoplasm ratio, a standard measurement used when
studying iPSCs.
The graph in Figure 3G illustrates the reduction in this ratio
in the non-optimized conditions, from 0.6 to 0.4. The more
iPSC like, and thus pluripotent, a cell is, the higher the
nuclear/cytoplasm ratio.
A
D
B
E
C
F
Nuclear/Cytoplasm Ratio
1.0
0.2
0.4
0.6
0.8
0.0
Optimized Non-optimized
G.
6
Figure 4. Changes in iPSC marker expression
analyzed with the iQue®3.
Bar graphs of data collected in iQue Forecyt®
software of iPSCs grown for (A) 2 days and (B) 4
days in optimized (mTESR Plus) and nonoptimized
(RPMI) media to induce
‘differentiation’. Marker expression of SSEA-1
(non-pluripotent marker), SSEA-4 (pluripotent
marker), TRA-1-60 (pluripotent marker) and
‘Pluripotent’ (SSEA-1 negative, SSEA-4/
TRA-1-60 positive) shown (± SEM, n=4). (C) Dot
plots showing SSEA-1 and ‘Pluripotent’ marker
raw data as presented in the iQue Forecyt®
software of iPSCs grown under optimized and
non-optimized conditions for 2 days (n=4).
NCCIT and THP-1 are control cell lines for
pluripotent marker expression and nonpluripotent
marker expression, respectively.
profile over the time course of these studies was observed
(95 ± 0.4% for pluripotent markers and less than 1.8 ± 0.5%
for SSEA-1). (Figure 4A, B). In addition, the increase in nonpluripotent
marker SSEA-1 expression (57.5 ± 0.7%) is clear
as early as 2 days post treatment (Figure 4A) and remains
high throughout culture.
In Figure 4C, (dot plots taken directly from iQue Forecyt®
software) there is a clear shift in SSEA-1 expression between
the optimized (1.63% SSEA-1 positive) and non-optimized
conditions (57.5 % SSEA-1 positive) (upper two dot plots).
The lower plots further illustrate the shift away from
pluripotent marker expression in the non-optimized
conditions, where the optimized iPSCs present a compact
population in the upper right quadrant of the plot (SSEA-
4+, TRA-1-60+) while the non-optimized iPSCs present a
much more spread population shifting into the TRA-1-60
negative portion of the plot.
Percentage Expression
of Live Cells
THP-1 NCCIT
Changes in iPSC Marker Expression Analyzed
with the iQue®3 High-Throughput Cytometry Platform
To investigate further the losses in pluripotency in iPSCs
when cultured in non-optimal conditions, surface marker
expression of specific pluripotency markers can be
analyzed with the iQue® 3, requiring as little as 10 μL per
sample.
iPSCs grown in non-optimized conditions show rapid loss
of pluripotency marker expression compared to optimized
conditions (Figure 4). This indicates a loss in pluripotency
correlating with the data collected on the Incucyte®
platform (Figure 3). After 2 days in culture (Figure 4A),
analysis of non-optimized conditions shows a decrease in
expression of pluripotency markers SSEA-4 (97.3 ± 0.8%),
TRA-1-60 (89.8 ± 0.9%), and the pluripotent population
(34.6 ± 0.3%), with a further decrease after 4 days of
treatment (SSEA-4 63.4 ± 2.9%, TRA-1-60 58.9 ± 2.9%,
pluripotent population 19.3 ± 3.0%) when compared with
optimized conditions (Figure 4B). In contrast, for the
optimized iPSCs, no marked differences in expression
A.
100
50
0
iPSC
Optimized
iPSC
Non-optimized
Percentage Expression
of Live Cells
THP-1 NCCIT
B.
100
50
0
iPSC
Optimized
iPSC
Non-optimized
SSC-H
SSEA-4 (RL 1-H)
SSEA-4 (RL 1-H)
SSC-H
107
107
0.49%
0.11%
15.96%
2.55%
99.40%
0.00%
80.64%
0.85%
Region Set 2 107 Region Set 3
107
105
105
105
104
103
105
106
106
106
106
SSEA-1 (BL1-H)
Optimized SSEA-1 negative
Optimized
Non-optimized SSEA-1 negative
Non-optimized
TRA-1-60 (BL2-H) TRA-1-60 (BL2-H)
SSEA-1 (BL1-H)
105
105
105
105
106
106 107
106
106
103
103
103
103
104
104
104
104
102
102
C.
SSEA-1
SSEA-4
TRA-1-60
Pluripotent
7
Figure 5. Surface and intracellular marker staining provides solutions for high throughput cellular characterization.
SSEA-1 was used as a marker of normal, non-pluripotent cells, while SSEA-4, TRA-1-60, Sox 2, and Oct 3/4 were all used to characterize
pluripotent cells. (A) Histograms and dot plots created in the Forecyt software system for iQue®3, showing the expression of various surface and
intracellular markers in iPSC and control cells (n=4). (B) Heatmap from iQue Forecyt ® illustrating the expression of the same markers, representing
the plate map and expression profile per well. (C) Bar graph showing marker expression data in 3rd party software (± SEM, n=4).
Surface and Intracellular Marker Staining Provides
Solutions for High-Throughput Cellular Characterization
Using the iQue®3 to monitor intracellular markers in
addition to surface markers further characterizes the
pluripotency of cells.
Using THP-1 cells as a non-pluripotent control, iPSCs were
fixed, permeabilized and stained for the surface markers
SSEA-1, SSEA-4 and TRA-1-60, in addition to the
intracellular markers Oct 3/4 and Sox 2 (Figure 5). Dot plot
data taken directly from iQue Forecyt® software, clearly
show the expression of pluripotency markers SSEA-4, TRA-
1-60, Oct 3/4 and Sox 2 in iPSC cells (black) and the nonpluripotent
marker, SSEA-1, only expressed in the THP-1
control cell line (yellow) (Figure 5A). The heatmap in Figure
5B illustrates this expression pattern in a plate view
configuration, where black is high expression and yellow is
low expression, exemplifying the flexibility of data
presentation in the iQue Forecyt® software. Analysis of this
data as a bar graph in Figure 5C further highlights the
contrasting expression profiles of the two cell types. The
ability to characterize a range of marker expression in cell
lines, including iPSCs, via a flexible multiplexed workflow,
exemplifies the power and utility of Sartorius platforms
such as the iQue®3 High-Throughput Cytometry Platform.
SSEA-1
SSEA-4
TRA-1-60
Sox 2
Oct 3/4 100
0
B. THP-1 iPSC
Events
100
90
80
70
60
50
40
30
20
10
0
100
90
80
70
60
50
40
30
20
10
0
SSEA-1 (BL1-H)
102 103
Live Live Live Live Live
Live Live Live Live Live
104 105 106
SSEA-1 (BL1-H)
102 103 104 105 106 107
SSEA-4 (RL1-H)
102 103 104 105 106 107
TRA-1-60 (BL2-H)
102 103 104 105 106 107
Sox 2 (VL1-H)
102 103 104 105 106 107
Oct 3/4 (VL2-H)
102 103 104 105 106 107
SSEA-1 (RL1-H)
102 103 104 105 106
TRA-1-60 (BL2-H)
102 103 104 105 106
Sox 2 (VL1-H)
102 103 104 105 106
Oct 3/4 (VL2-H)
102 103 104 105 106
140
120
100
80
60
40
20
0
SSC-H Events
107
106
105
104
103
SSC-H
107
106
105
104
103
SSC-H
107
106
105
104
103
SSC-H
107
106
105
104
103
SSC-H
107
106
105
104
103
140
120
100
80
60
40
20
0
Events
140
120
100
80
60
40
20
0
Events
Samples THP-1
Samples iPSC
SSEA-1 pos SSEA-4 pos TRA-1-60 pos Sox 2* Oct 3/4*
A.
Percentage Expression
of Live Cells
SSEA-1 SSEA-4
100
50
0
Surface Markers Intracellular Markers
TRA-1-60 Sox 2 Oct 3/4
THP-1
iPSC
C.
8
Specifications subject to change without notice. ©2023 All rights reserved. All names of Sartorius products are registered trademarks and the property of Sartorius AG and/or one of its affiliated companies.
Culture-Maintain-characterize-iPSCs-App-Note-2307-en-L-Sartorius Status: 07 | 2023
Conclusions
iPSCs are increasingly used in many areas of research,
requiring specific conditions for optimal growth, to
maintain pluripotency, viability, and propagation
potential. These requirements are often expensive and
methods for monitoring iPSC status can be complex and
time intensive, requiring multiple complicated
techniques and solutions.
Using various Sartorius platforms throughout an iPSC
culture workflow, we have shown how we can successfully
pick and seed iPSCs, monitor their morphological status
and characterize their pluripotency using the
CellCelector Flex, the Incucyte® Live-Cell Analysis
platform and the iQue®3 High-Throughput Cytometry
Platformr. The key advantages of using this combined
workflow over conventional methods are:
1. Consistency and reliability, the CellCelector Flex can
reproducibly pick specific iPSC colonies for further
testing or culture, maintaining high levels of cell health
2. The ability to monitor delicate iPSC line culture
morphology and growth characteristics without
removing plates from the incubator
3. Minimal sample volumes required to characterize
precious cell types, with minimal attrition for
downstream requirements
4. Multiplexing experiments providing flexibility for the
characterization of multiple metrics, such as surface
and intracellular marker expression, using the same
platform
The data presented here showcases the advantages of
using a streamlined workflow combining multiple
Sartorius systems for the culture, monitoring and
characterization of iPSCs for several applications from
drug development, disease modeling and clinical
therapy research.
References
1. Takahashi K, Yamanaka S. (2022) Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures
by defined factors. Cell. 2006 Aug 25;126(4):663-76. doi: 10.1016/j.cell.2006.07.024. Epub 2006 Aug 10. PMID: 16904174.
North America
Phone +1 734 769 1600
Europe
Phone +44 1763 227400
Email:
Japan
Phone +81 3 6478 5202
China
Phone +86 21 6878 2300
Rest of Asia Pacific and other
countries around the world:
Email: lps.opm.na@sartorius.com Phone +65 6872 3966
orderhandling.lps.ne@sartorius.com
Email: sartoriusap@sartorius.com
For questions, email: AskAScientist@sartorius.com
Find out more: www.sartorius.com/ique
Find out more: www.sartorius.com/incucyte
Neurodegenerative diseases, which include Alzheimer’s
disease and related dementias, Parkinson’s disease, and
motor neuron diseases such as amyotrophic lateral
sclerosis (ALS) and spinal muscular atrophy, significantly
impact individuals and society. Developing effective
treatments for these conditions hinges on access to
translational models that help identify drug candidates
with a high likelihood of success in clinical trials. This article
offers an overview of the role of complex cell models in
neurodegenerative disease research, as well as advanced
live-cell techniques for obtaining essential real-time
insights from these systems.
The Necessity of Complex Cell Models
for Disease Modeling
Traditional disease modeling has relied heavily on the use
of immortalized cell lines, such as HeLa and HEK293 cells,
and primary cells derived from animal or human tissues.
While these models have facilitated many scientific
breakthroughs, they inherently lack the complexity of
human disease states.
Complex cell models, such as iPSCs and 3D cultures,
represent a paradigm shift in disease modeling.1 iPSCs,
with their capacity to differentiate into any cell type, offer a
renewable source of patient-specific cells that retain the
donor’s genetic information. This allows for the modeling of
diseases with a genetic component in a patient-specific
way, providing insights into the personalized nature of
disease progression and response to treatment. iPSCs also
enable the study of rare diseases for which patient samples
are scarce, broadening the scope of research and potential
therapeutic interventions.
Three-dimensional (3D) cell models, including organoids
and spheroids, further enhance the capabilities of iPSCs by
providing a scaffold for the development of tissue-like
structures. These models replicate the architecture and
multicellular complexity of organs, including the formation
of gradients for oxygen, nutrients, and signaling molecules,
which are essential for understanding disease progression
and the efficacy of therapeutics.
Advancing Neurodegenerative Disease Research:
The Critical Role of Complex Cell Models
and Live-Cell Techniques
Tina Shahian, PhD — Content Writer - Sartorius
Predictive Cellular Models
for Neurodegenerative Diseases
The use of iPSCs in neurodegenerative disease modeling
enables detailed exploration of disease mechanisms with
unprecedented precision. iPSC-derived neurons can
display key Alzheimer’s features, including amyloid-beta
peptide aggregation and tau hyperphosphorylation,
providing a dynamic system for studying disease
progression. Additionally, iPSCs allow for the study of
neuronal loss and synaptic dysfunction, crucial to
understanding neurodegeneration.
Beyond their role in modeling disease pathology, iPSCs are
transforming drug discovery and development. iPSCderived
neural cells enable high-throughput screening to
pinpoint potential therapeutics and assess drug toxicity
and efficacy, thus fast-tracking the journey from laboratory
research to clinical application.
Stem cell-derived 3D cell culture techniques have
significantly improved our capacity to model
neurodegenerative diseases.2,3 For Alzheimer’s disease, 3D
brain organoids enable the observation of amyloid plaque
formation and neurofibrillary tangles, providing a more
accurate platform for investigating disease mechanisms
and evaluating drug candidates. These 3D models also
enhance the study of non-neuronal factors in
neurodegeneration, including microglial involvement in
inflammation and the influence of astrocytes on neuronal
health.4
Pros and Cons of Traditional Techniques for Cell Analysis
Traditional cell biology techniques like flow cytometry and
high-content imaging (HCI) have been pivotal in studying
cellular processes and disease states. Flow cytometry
analyzes individual cells, yielding detailed data on cell
health, size, and marker expression through fluorophores
and labeled antibodies. HCI enables the visualization and
analysis of static cell populations, capturing heterogeneity
and allowing for single-cell level subset analysis.
However, these methods have limitations, especially with
complex cell models. The preparatory and labeling
processes required for flow cytometry and HCI can alter
the cellular environment, potentially introducing artifacts
that affect experimental results. Moreover, these
techniques usually provide snapshots at single time points,
missing the dynamic cellular changes over time that are
crucial for understanding disease progression.
Real-Time Monitoring of Live Cellular Behavior
Live-cell analysis overcomes many of these limitations by
enabling continuous monitoring. This method is especially
useful for complex models, providing real-time insights into
living cells’ behavior and function without disrupting their
natural state. For example, live-cell analysis can observe
the growth and maturation of iPSC-derived neurons or the
development of neural networks in 3D brain organoids,
offering a glimpse into these processes.
Technologies like the Incucyte® Live-Cell Analysis System
(Sartorius) are tailored for live-cell analysis, allowing for
automated imaging and analysis within a cell incubator’s
controlled environment. Its non-invasive approach permits
the detection of gradual cellular changes in morphology,
function, and interactions, crucial for studying diseases with
slow cellular progression. Additionally, the system’s
capability to simultaneously monitor multiple parameters,
such as cell morphology and marker expression, is invaluable
for understanding complex cellular behaviors like
phagocytosis.
Numerous studies have illustrated the utility of live-cell
analysis in neuroscience research, including one by Jessica
Tilman’s group at Axol Bioscience Ltd. demonstrating
disease-specific functional impairments in ALS cells.5 Their
work revealed morphological and functional differences
between healthy and iPSC-derived ALS cells, with ALS motor
neurons showing disorganized structures and erratic firing
patterns, and ALS microglia demonstrating reduced
phagocytosis. They used the Incucyte® system to measure
spontaneous neuronal activity and microglial phagocytosis,
highlighting the value of real-time monitoring of cellular
behavior.
Conclusion
The integration of advanced cell models with live-cell
analysis is a critical development in neurodegenerative
disease research. This non-destructive approach allows for
the continuous collection of vital data, enhancing our
References
1. Langhans SA. Three-dimensional in vitro cell culture models in drug discovery and drug repositioning. Front Pharmacol. 9, 6
(2018). https://doi.org/10.3389/fphar.2018.00006
2. Lee HK, Velazquez Sanchez C, Chen M, Morin PJ, Wells JM, Hanlon EB, Xia W. Three-dimensional human neuro-spheroid
model of Alzheimer’s disease based on differentiated induced pluripotent stem cells. PLoS ONE. 11, e0163072 (2016). https://
doi.org/10.1371/journal.pone.0163072
3. Park J, Wetzel I, Marriott I, Dréau D, D’Avanzo C, Kim DY, Tanzi RE, Cho H. A 3D human triculture system modeling
neurodegeneration and neuroinflammation in Alzheimer’s disease. Nat Neurosci. 21, 941–951 (2018). https://doi.org/10.1038/
s41593-018-0175-4
4. Park J, Wetzel I, Marriott I, Dréau D, D’Avanzo C, Kim DY, Tanzi RE, Cho H. A 3D human triculture system modeling
neurodegeneration and neuroinflammation in Alzheimer’s disease. Nat Neurosci. 21, 941–951 (2018). https://doi.org/10.1038/
s41593-018-0175-4
5. Tilman J. iPSC-derived motor neurons and microglia from ALS background display disease phenotype. Axol Bioscience Ltd.
(2023, August 31). Available from: https://www.sartorius.com/en/products/live-cell-imaging-analysis/live-cell-analysisresources/
ipsc-derived-motor-neurons-and-microglia-white-paper
White Paper
August 31, 2023
Keywords or phrases:
ALS, Neurodegenerative, iPSC, Immunocytochemistry,
Neuronal Activity, Phagocytosis, Motor Neurons,
Microglia, Live-Cell Analysis
iPSC-Derived Motor Neurons and Microglia
From ALS Background Display Disease
Phenotype
Jessica Tilman
Axol Bioscience Ltd, Roslin Innovation Centre, Charnock Bradley Building, Easter Bush Campus, Easter Bush EH25 9RG UK
Correspondence
Email: AskAScientist@sartorius.com
Abstract
Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disorder caused by the degeneration of motor neurons, and ultimately
results in death. Despite the fatal nature of this disease, there is no current cure or effective treatment available. The clear
overlap between ALS and Frontotemporal Dementia (FTD) has led to the belief that it is not only disruption in motor neurons
which drives ALS symptoms, but other cell types such as microglia, the main immune cell found in the brain, may also play a role.
To investigate this, fibroblasts taken from a healthy individual and an ALS patient carrying a C9orf72 hexanucleotide expansion
(GGGGCC >145) were reprogrammed to Induced Pluripotent Stem Cells (iPSCs), and then differentiated to motor neurons and
microglia alongside controls. The cells were then assessed for differences in morphology and functional activity, measured via
electrophysiology, spontaneous neural activity, and microglial phagocytosis. Diseased iPSC-derived motor neurons exhibited less
organized morphology compared to control, as well as less synchronized firing patterns and hyperexcitability. ALS patient-derived
microglia demonstrated a decreased capability for phagocytosis, particularly when cryopreserved. Taken together, these results
demonstrate clear morphological and functional differences in differentiated cells derived from a C9orf72-positive ALS patient,
compared to healthy control cells which can therefore be used as a robust model for C9orf72 ALS research and drug discovery.
For further information, visit www.sartorius.com
2
Amyotrophic lateral sclerosis (ALS) is a neurodegenerative
disorder caused by the degeneration of motor neurons,
and ultimately results in death on average 2–3 years after
onset of symptoms. 5–10% of ALS cases have a familial
background with an autosomal dominant inheritance
pattern. Of these, mutations in C9orf72 account for the
highest number of cases1. Sporadic ALS accounts for the
remaining cases, with a high degree of them containing
identifiable genetic causes. There is a clear overlap between
ALS and FTD with approximately 5% of ALS patients showing
overt characteristics of FTD, and up to 50% showing more
subtle similarities2. This observation has led to the belief
that it is not only disruption in motor neurons which drives
ALS symptoms, but other cell types play an important role
too. Microglia are the main immune cell found in the brain,
with key roles in maintaining neuronal homeostasis as well as
driving inflammatory responses3. Changes in inflammatory
response and phagocytosis have been heavily implicated
in neurodegeneration and FTD, and more recently have
been shown to be defective in ALS. C9orf72 mutation has
been shown to drive an exaggerated immune response
in microglia and lead to impaired phagocytosis and
excitotoxicity in motor neurons3,4.
ALS has an incidence rate of 2:100,000 per year, and
despite the fatal nature of this disease, there is no current
cure or effective treatment available1. There has been
significant progress in the field with greater understanding
of the genetics, pathology, and biomarkers of ALS, but
development of novel and effective therapies has in part
been hindered by a lack of relevant and translational
pre-clinical models. Understanding how different cell
types become impaired and drive the development and
progression of ALS is crucial in developing novel and
effective treatments.
Use of relevant pre-clinical models is essential in not only
understanding disease onset and progression, but also in
gaining insight into the functionality of potential new treatments.
ALS is a heterogeneous condition, and as such one
treatment plan is unlikely to work for all patients. Mouse
models are limited in this respect, as in most cases they rely
on genetic modification or silencing of a key gene and would
therefore only apply to patients with the same mutation.
iPSC-derived models provide a relevant human platform
which is reproducible and scalable. More importantly, the
ability to reprogram samples directly from affected individuals
allows for patient stratification based on not only genetic
factors, but also on defined symptoms. This is invaluable in
determining which treatments may work more effectively for
subpopulations of patients with ALS and drive more effective
clinical trials.
An additional benefit of using iPSC-derived cells is the
ability to differentiate these cells towards different lineages
and generate several cell types which all contribute
to development of ALS. Motor neurons harbouring a
C9orf72 mutation are known to be hyperexcitable and to
become dysfunctional in ALS. However, C9orf72 is most
highly expressed in myeloid cells, implying an important
contribution from these cells. Using iPSC-derived motor
neurons and microglia from the same patient background
enables for the study of these cells in isolation as well as in
co-culture.
Method and Assays
axoLines™ iPSCs:
Fibroblasts taken from a healthy individual and an ALS
patient carrying a C9orf72 hexanucleotide expansion
(GGGGCC >145) were reprogrammed to iPSCs using
Sendai virus or episomal vector.
axoCells™ iPSC-Derived Motor Neurons:
To generate mature motor neurons, iPSCs were differentiated
to axoCells motor neuron progenitor cells (ax0078
and ax0074). Progenitor cells were generated in monolayer
using small molecule induction methods. Progenitors were
then matured to motor neurons for up to 20 days using the
Axol Bioscience methodology to include neural media with
the addition of Axol Bioscience motor neuron accelerator
(ax0072 and ax0179). Motor neurons were characterized by
immunocytochemistry, showing expression of HB9, LIM3,
OLIG2, and TUJ1 by day 104 of maturation.
axoCells™ iPSC-Derived Microglia:
iPSCs were differentiated to macrophage-precursors by
mesoderm induction via the formation of embryoid bodies.
Precursor cells were characterised by flow cytometry for key
macrophage|monocyte markers (CD14, CD11b, and CD16)
before maturing the cells to microglia for a further 7 days
using a cocktail of growth factors. At day 7 cells were either
used for assays or cryopreserved. Upon thaw, cryopreserved
microglia were plated on cell culture plates coated with
Surebond XF and Concanavalin A and matured for a further
7 days before performing assays. Fresh and cryopreserved
microglia were characterized by immunocytochemistry for
expression of key microglia markers TMEM119, P2RY12,
CX3CR1, and Iba1.
Immunocytochemistry:
Cells were fixed using 4% PFA for 10 minutes and washed in
phosphate buffered saline (PBS). Cells were permeabilised
using 1% Triton X and blocked in buffer containing Tween
20 and Donkey serum. Cells were then stained with primary
antibodies for 2 hours at room temperature (RT) and washed
twice using PBS-Tween. Secondary antibodies conjugated
to FITC or AF-594 were then added for a further 2 hour
incubation at RT. All wells were treated with ProLong™ Gold
Antifade reagent containing DAPI and imaged using a Leica
microscope.
Introduction
3
Multi-Electrode Array (MEA):
Motor neuron progenitor cells were seeded at 150,000
cells/cm2 in 48 well Axion CytoView MEA plates coated with
vitronectin. Neurons were matured for 10 days, and MEA
recordings of spontaneous firing taken throughout this
process using an Axion Maestro Pro system. Recordings were
analyzed to provide information on neuronal activity, burst
firing, and synchronicity.
Spontaneous Neuronal Activity (SNA):
On day two of maturation of motor neurons, cells were
transfected with Incucyte® Neuroburst Orange Lentivirus
(Sartorius, Cat. no. 4736), a lentiviral based live-cell neuronal
labeling reagent driven by a synapsin promoter, leading to
the long term expression of an orange fluorescent calcium
indicator. Motor neurons were imaged daily up to day 21 of
maturation. Images were acquired using an Incucyte® S3
Live-Cell Analysis System configured with an Orange/NIR
optical module and integrated Incucyte® Neuronal Activity
Analysis Software Module was used to determine metrics of
spontaneous neuronal activity.
Phagocytosis:
Myelin basic protein (MBP) was labeled with pHrodo® Orange
Cell Labeling Dye for Incucyte® (Sartorius, Cat. no.: 4766 )
following the manufacturer’s specifications. Labeled MBP
was used as bait for microglia. Briefly, microglia were seeded
in 384 well plates and matured for 7 days. On day 7, bait was
added to each well and imaged using an Incucyte® S3 Live-
Cell Analysis System for up to 24 hours. Cytochalasin D was
added to control wells 30 minutes prior to addition of bait.
iPSC-Derived Motor Neuron Morphology
axoCells™ iPSC-derived motor neurons undergo quality
control by staining for key markers such as CHAT, TUJ1, and
HB9
(not shown) to ensure maturity (Figure 1). While these
markers are present in both control and ALS motor neurons,
the morphology of the neuronal networks shows differences
between the two, indicating a disease driven phenotype.
Control motor neurons form large clusters of cell bodies
connected by strong cabling networks formed by neurites.
In comparison, ALS motor neurons form many smaller
clusters across the culture with a more disordered network
formation. This could drive the hyperexcitability observed
in ALS motor neuron cultures and explain in part the loss of
synchronicity in this population.
Phase contrast Blue: DAPI
Figure 1: Mature axoCells™ iPSC-derived motor neurons (control and ALS) immunocytochemistry. Cells were matured for 20 days and stained for markers
of motor neurons. DAPI was used as a nuclear dye.
Ax0078 (control)
Ax0074 (ALS C9orf72)
Green: HB9 Red: TUJ1 Merge
4
Figure 2: MEA recordings of day 10 mature motor neurons from healthy and ALS patients. Traces of activity detected for each cell line (top) and raster plot
(bottom) demonstrating the difference in firing frequency and synchronicity.
axoCells™ Motor Neuron (control-derived)
Motor Neuron MEA Sodium Spike Profile
axoCells™ Motor Neuron (ALS-derived)
Motor Neuron MEA Sodium Spike Profile
Time (sec)
Synchronized clustered firing
100 200 300 400 500 600
There are several methods that can be used to assess
neuronal function in vitro, one of which is using a multi
electrode array (MEA) system. The functional behavior
of neuronal networks can be captured to investigate electrical
activity by measuring spontaneous action potentials (field
potentials). Using this system, electrical activity can be
compared between healthy and disease motor neurons to
identify patterns of firing which contribute to the disease
phenotype. In this way, a ‘disease phenotype’ can be defined
and used to test potential drug candidates that may lead
to a return to normal firing in motor neurons from a disease
background.
Another method to assess neuronal function is to interrogate
the function of the whole well, as opposed to MEA in which
a certain number of electrodes provide the information for
the cell networks directly on them. Spontaneous neuronal
activity can be measured using the Incucyte® Neuronal
Activity Assay, wherein cells are monitored daily over the
maturation process. Cells are transfected with Incucyte®
Neuroburst Orange Lentivirus, a genetically-encoded
calcium sensor, resulting in long term non-perturbing
expression in neuronal cell types.
Using integrated software, calcium flux can then be kinetically
measured as a readout for spontaneous neuronal activity
and enabling assessment of burst rate, burst duration, and
synchronicity (mean correlation) to investigate functional
differences between cell lines. Data obtained from healthyand
ALS-derived motor neurons shows that control neurons
have a regular and synchronized firing pattern while ALS
neurons fire more frequently and with reduced synchronicity
(Figure 3 A). ALS motor neurons exhibited significantly
higher burst rate compared to the control line, along with
lower burst duration and lower mean correlation (hence less
synchronized burst firing) (Figure 3 B).
Synchronized clustered firing
Time (sec)
100 150 200 250 300 350 400 450 500 550
Motor neurons from control and ALS patient backgrounds
were assessed using an Axion MEA system and demonstrated
a clear disease phenotype (Figure 2). Control neurons
exhibited regular and synchronized firing while ALS
neurons showed more random clustered firing. This can
be observed in the field potential traces (top) and in the
raster plot (bottom). This disease phenotype is in line with
the physiological dysfunction observed in neurons from
ALS patients, including hyperexcitability and a loss of
synchronicity within the cell population.
axoCells™ ALS Derived Motor Neurons Display Hyperexcitability Across Several
Assay Platforms
5
Time (sec)
Intensity (OCU)
18
0 10 20 30 40 50 60 70 80 90
16
14
12
10
8
6
4
2
0
Time (sec)
Intensity (OCU)
0 10 20 30 40 50 60 70 80 90
2
10
3
4
5
6
7
8
9
10
A) axoCells™ Motor Neuron axoCells™ Motor Neuron
B) Mean Burst Rate
Burst rate (1/min)
6
ax0074 (ALS)
ax0078 (cont)
4
2
0
*
Mean correlation
1.2
ax0074 (ALS)
ax0078 (cont)
0.8
0.4
0
**
1.0
0.6
0.2
Burst duration (sec)
15
ax0074 (ALS)
ax0078 (cont)
10
05
0
**
Mean Correlation Mean Burst Duration
Figure 3: Spontaneous neuronal activity assay on day 18 motor neurons. Calcium signalling was monitored and analysed using the Incucyte® Neuronal
Activity Software Module. A) Burst patterns for control and ALS motor neurons. B) Neuronal activity analysis for calcium signalling. Statistical significance
was assessed using a non-parametric T-test, *p<0.05, **p<0.01.
iPSC-derived microglia provide a platform to investigate how
the immune response contributes to neurodegeneration and
thus can be used for disease modeling and drug discovery.
axoCells™ iPSC-derived microglia show expression of key
markers by immunocytochemistry, including TMEM119,
Iba1, P2RY12, and CX3CR1. Using iPSC-derived microglia, a
clear difference could be observed in the capability of these
cells to uptake myelin basic protein (MBP) with significantly
higher phagocytosis in healthy cells compared to those from
an ALS patient (Figure 4).
In addition, ALS microglia were more sensitive to the
cryopreservation process, with the difference between
healthy and disease cells exacerbated in thawed cells.
Together this data indicate that ALS microglia are affected by
the C9orf72 expansion in ALS with decreased phagocytosis
and potentially becoming more sensitive to stresses such as
cryopreservation.
ALS Disease Phenotype is Observed in Microglia Phagocytosis
North America
Sartorius Corporation
565 Johnson Avenue
Bohemia, NY 11716
USA
Phone +1 734 769 1600
Email: orders.US07@sartorius.com
Europe
Sartorius UK Ltd.
Longmead Business Centre
Blenheim Road
Epsom
Surrey, KT19 9QQ
United Kingdom
Phone +44 1763 227400
Email: eurordersUK03@sartorius.com
For further information, visit
www.sartorius.com
Axol Bioscience Ltd
Roslin Innovation Centre,
Charnock Bradley Building,
Easter Bush Campus, Easter
Bush EH25 9RG UK
Asia Pacific
Sartorius Japan K.K
4th Floor, Daiwa Shinagawa North Bldg.
1-8-11, Kita-Shinagawa 1-chome
Shinagawa-Ku
Tokyo 140-0001
Japan
Phone +813 6478 5202
Email: eurordersUS07@sartorius.com
Specifications subject to change without notice. ©2023. All rights reserved.
All names of Sartorius products are registered trademarks and the property of Sartorius AG and | or one of its affiliated companies. IPSC-Derived-Motor-Neurons-Whitepaper-en-Sartorius.
Status: 10 | 2023
Figure 4: axoCells™ iPSC-derived microglia characterized by A) immunocytochemistry for key markers (representative images); B) phagocytosis of
pHrodo® for Incucyte® labeled Myelin Basic Protein (MBP) was assessed on an Incucyte® Live-Cell Analysis System up to 24h using fresh and cryopreserved
microglia. Cytochalasin D (10 μM) was used as a control. Statistical significance was assessed using a one-way ANOVA, **p<0.01, ***p<0.001, ****p<0.001.
Summary & Outlook
iPSC-derived cells provide a valuable tool for disease
modelling, with the ability to use cells directly from affected
patients, as well as the potential to drive these cells towards
multiple lineages. In this study, axoCells™ motor neurons
and microglia from the same healthy and ALS axoLines™
iPSCs were shown to exhibit disease phenotypes which can
be quantified and used for future drug discovery purposes.
Further characterization of both cell types could elucidate
additional functional dysregulation compared to healthy
cells which may provide valuable insight into future potential
treatments. In addition to understanding disease phenotype
for each cell type, the potential to develop a co-culture
system would add value in determining how these cells
interact and whether both microglia and motor neurons
from ALS patients could drive a more pronounced disease
phenotype.
References
1. Mead, R.J., Shan, N., Reiser, H.J. et al. Amyotrophic lateral
sclerosis: a neurodegenerative disorder poised for successful
therapeutic translation. Nat Rev Drug Discov 22, 185–212
(2023). https://doi.org/10.1038/s41573-022-00612-2
2. Poulomi Banerjee et al. Cell-autonomous immune dysfunction
driven by disrupted autophagy in C9orf72-ALS
iPSC-derived microglia contributes to neurodegeneration.
Sci. Adv.9,eabq0651(2023).DOI:10.1126/sciadv.abq0651
3. J. Brettschneider, J. B. Toledo, V. M. van Deerlin, L. Elman,
L. McCluskey, V. M.-Y. Lee, J. Q. Trojanowski, Microglial
activation correlates with disease progression and upper
motor neuron clinical symptoms in amyotrophic lateral
sclerosis. PLOS ONE 7, e39216 (2012).
4. O. Dols-Icardo, V. Montal, S. Sirisi, G. López-Pernas, L.
Cervera-Carles, M. Querol-Vilaseca, L. Muñoz, O. Belbin,
D. Alcolea, L. Molina-Porcel, J. Pegueroles, J. Turón-Sans,
R. Blesa, A. Lleó, J. Fortea, R. Rojas-García, J. Clarimón,
Motor cortex transcriptome reveals microglial key events in
amyotrophic lateral sclerosis.
A)
Red: TMEM119
Blue: DAPI
Red: P2RY12
Blue: DAPI
Green: Iba1
Blue: DAPII
Red: CX3CR1
Blue: DAPI
Integrated total intensity
0.8
Healthy
ALS (C9orf72)
0.4
0.0
**
Cyto D 10μM
****
0.6
0.2
B) MBP 24h (fresh)
Relative Phagocytosis
0.5
Healthy
ALS (C9orf72)
0.3
0.0
***
Cyto D
***
0.4
0.2
0.1
MBP 24h (Cryopreserved)
Note: axoCells™ and axoLines™ are registered trademarks of Axol Bioscience
iPSCs in Focus:
Translational Strategies for
Reproducible Organoid Research
Effective analysis of 3D cell models can be challenging. The Incucyte® Live-cell Analysis
System is a turnkey solution that automatically monitors and quantifies cell formation,
growth, and health in real time directly inside the incubator. Multiple assays are available
for real-time visualization and label-free objective quantification to optimize organoid
and spheroid culture conditions.
Simplifying Progress
Specifications subject to change without notice. © 2024. All rights reserved. Incucyte and all names of Sartorius products
are registered trademarks and the property of Sartorius AG.
Application Note
December 01, 2022
Keywords or phrases:
Neuroscience, Neuronal activity, Neuronal Function,
GECI, Calcium flux, iPSC, Cell Analysis Systems
Find out more: www.sartorius.com/incucyte
Long-Term Live-Cell Visualization and
Quantification of Neuronal Activity
S. L. Alcantara¹, J. Trigg¹, J. Rauch², L. Oupicka², N. Holtz², E. Endsley², T. Dale¹*
¹ Sartorius UK Ltd., Royston, Hertfordshire, UK
² Sartorius Corporation, 300 West Morgan Road, Ann Arbor, MI 48108 USA
* With thanks to T. Campbell**, T. Jackson, A. Overland, J. Brown, T. G. Garay
** Talisman-therapeutics.com
Introduction
A major impediment to studying diseases affecting the human nervous system is the ability to monitor, analyze,
and quantify the activity of neuronal cell populations that accurately represent human phenotypes. These
limitations are the consequence of minimal access to cells from human patient tissue, as well as a lack of purposebuilt
instrumentation enabling functional measurements from neuronal cells at sufficient throughput to permit
full phenotypic characterizations. With the advent of cellular reprogramming technologies, there has been
abundant research toward protocol development to differentiate human induced pluripotent stem cells (hiPSCs)
into multiple cell populations found in the brain (e.g. neuronal, glial, immunological, etc.). This has resulted in the
generation of many different neuronal cell models (e.g. dopaminergic, GABAergic, glutamatergic, peripheral, etc.),
most of which remain poorly characterized. This imparts a requirement to better understand in vitro cellular
models and identify means by which they could be refined. The Incucyte® Live-Cell Analysis System technology,
methodology, and applications described within this application note were designed with these issues in mind.
That is, to provide researchers with a set of automated tools in order to facilitate the evaluation, characterization,
and validation of complex neuronal models.
Susana Lopez Alcantara — Scientist – Sartorius
Jasmine Trigg — Scientist – Sartorius
John Rauch — Manager of Cell Assays – Sartorius
Libby Oupicka — Scientist – Sartorius
Nevine Holtz, PhD — Manager of Advanced Algorithm Development – Sartorius
Eric Endsley — Head of Technology Translation – Sartorius
Tim Dale — Head of LPS Applications – Sartorius
With thanks to:
T. Campbell — Talisman-therapeutics.com
T. Jackson, A. Overland, J. Brown, T. G. Garay
Burst Rate
Correlation
Mean Correlation Mean Burst Rate
Time (days)
1.00
0.75
0.50
0.25
0.00
9/min
6
3
0
0 5 10 15 20
Figure 1: Incucyte® Neuronal Activity Assay Workflow
The Incucyte® Neuronal Activity Assay allows for measurements of long-term synaptic activity from neuronal cell models in physiologically relevant
conditions. The assay provides an end-to-end solution consisting of reagents, protocols, instrumentation, and software for a user-friendly workflow.
2
Assay Principle
While measuring morphological features of neurons
(e.g. neurite outgrowth) can provide insight into their
structure, neuronal activity assays provide a more
exquisite and sophisticated understanding of how
neurons function, form synaptic connections with other
neurons, and how they respond to their environment.
In this application note, we describe an integrated
solution for long-term neuronal activity measurements
based on Incucyte® Neuroburst Orange Lentivirus,
a neuronal specific, live-cell genetically-encoded calcium
indicator (GECI) and the Incucyte® Live-Cell Analysis
Systems configured with either an Orange/NIR or a
Green/Orange/NIR Optical Module. Along with
integrated analysis tools provided by the Incucyte®
Neuronal Activity Analysis Software Module, this enables
automated quantification of calcium oscillations and
morphological monitoring of thousands of functional
neurons within a culture over long periods of time—days,
weeks, and months (Figure 1). This approach provides
researchers the opportunity to better understand how
and when network connections are made between cells
in culture, and how the environmental context (e.g. drug
treatments, stromal cells, media formulations, etc.)
can alter their behavior.
The Incucyte® Live-Cell Analysis System user interface is
designed to visualize neuronal activity within each well of a
96-well plate. Each scan consists of a 30–180 second
“Stare Mode” capture of cellular activity at a rate of three
frames per second. Each “Stare Mode”-acquired movie is
distilled into a single range image to allow for simple
viewing (Figure 2B). This image represents the range of
intensities that are detected from each cell within the
culture over the specified scan time. Using this image,
automated image segmentation tools are used to identify
active objects (cells) within each well (Figure 2C). Based
on the changing fluorescent intensity of each individual
cell, intensity traces are displayed for every active cell in
the culture (Figure 2D). Scanning is typically completed
once every 24 hours. As shown in these sample traces of
iPSC-derived iCell® GlutaNeurons (Cellular Dynamics),
the activity within these cultures can significantly change
from day-to-day as the network matures, in this case with
minimal activity at Day 4, a gradual increase at Day 7, and
highly synchronous activity visualized at Day 12 and Day 17.
Once these data are collected, several automated metrics
are calculated for each well and at each scan time,
allowing for simple visualization of changing metrics over
the full time-course of the experiment (Table 1).
1. Prepare assay plate
Infect neurons in mono- or
co-culture with Incucyte®
Neuroburst Orange
Lentivirus.
2. Acquire movies
Capture short-term,
calcium-flux kinetics
using Incucyte® Stare
Mode acquisition.
3. Analyze active cells
Quantify burst rate of active
neurons in every movie and
visually assess morphology
with phase-contrast images.
4. Conduct chronic studies
Continuously interrogate
dynamic changes in the same
population of cells in individual
wells over weeks or months.
Active Objects=1363
Mean Correlation=0.493
Active Objects=118
Mean Correlation=0.0161
Active Objects=2003
Mean Correlation=0.864
Active Objects=2028
Mean Correlation=0.828
Intensity (OCU) Intensity (OCU) Intensity (OCU)
Intensity (OCU)
0
0
0 0
60
60
60 60
120
120
120 120
180
180
180 180
Time (sec)
Time (sec)
Time (sec) Time (sec)
4.8
4.8
4.8 4.8
3.6
3.6
3.6 3.6
2.4
2.4
2.4 2.4
1.2
1.2
1.2 1.2
0
0
0 0
Figure 2: Incucyte® Neuronal Activity Assay Protocol
and Purpose-built Software
Quick guide workflow of Incucyte® Neuroburst Orange Lentivirus
infection protocol (A). The Incucyte® Neuronal Activity Analysis
Software Module user interface is capable of displaying object traces,
viewing movies, and longitudinal data of neuronal activity from each
well (B). Fluorescent range image and automated segmentation mask
of each active object represents a snapshot of activity over the
complete scan (C). An example of iCell® GlutaNeuron calcium traces
from each 3 minute scan indicate changing neuronal activity
(fluorescence intensity) over 17 days in culture (D).
B. C.
D.
Fluorescent Range
Day 4
Day 12
Active Object Mask
Day 7
Day 19
3
Coat plate with
matrix of choice and
incubate at ambient
temperature
overnight.
Plate Incucyte®
rCortical Neurons.
Plate Incucyte®
rAstrocytes.
1. Remove Incucyte® Neuroburst
Orange Lentivirus.
2. Add Uridine + 5- Fluoro-2’-
deoxyuridine.
3. Start Neuronal Activity scanning.
Add Incucyte®
Neuroburst Orange
Lentivirus.
1. DIV: -1 2. DIV: 0 3. DIV: 0 + 2 hr 4. DIV: 2 5. DIV: 3
A. Quick Guide: Incucyte® Neuroburst Orange Lentivirus
Neurons
per well
Table 1: Neuronal Activity Analysis Metrics
Figure 3: Functional Activity of Primary Neurons.
Primary rat forebrain neurons were seeded at 40K (rows A and B), 20K
(rows C and D), 10K (rows E and F), and 5K (rows G and H) cells/well.
All densities of neurons were plated in a co-culture with primary rat
astrocytes seeded at 15K cells/well and transduced with the Incucyte®
Neuroburst Orange Lentivirus. 96-well vessel view of the range image
over the course of the scan provides a snapshot of active wells at each
time point (A). Summary traces of fluorescence intensity across all
active objects for the 96-well plate at Day 12 provide an overview of
activity and display metrics of bursting intensity, active object number
and mean correlation (B). 96-well throughput with high kinetic
reproducibility over 12 days in culture (C).
A. B. Summary Trace Activity at Day 12
C. Number of Active Objects over 12 Days
1
A
B
C
D
E
F
G
H
2 3 4 5 6 7 8 9 10 11 12
4
Metric Description
Active Object Count
(1/image)
The number of objects (cells/cell clusters) that burst at least once above the Minimum Burst threshold
over the total scan time.
Mean Intensity (OCU) The mean intensity of an object over the total scan time. All objects within the image are calculated
individually, then values are averaged.
Mean Correlation Every object is compared to every other object in the image to generate a value between -1 and 1, with 0
being completely random and 1 being highly synchronized. This is a measure of network connectivity.
Mean Burst Duration (sec) The duration of each calcium burst over the total scan time is calculated individually, then values are averaged.
Mean Burst Rate (1/min) The number of calcium bursts over the total scan time divided by the scan time in min.
Mean Burst Strength (OCU) The area under each calcium burst divided by the duration of that burst is calculated individually, then
values are averaged.
Results
Optimization of Incucyte® Neuroburst Orange Lentivirus
Primary rat neurons (E18) in co-culture with primary rat astrocytes
represent a well-tested model for studying neuronal
activity. In this experiment, E18 rat forebrain neurons were
plated at decreasing cell densities (5–40K/well) in co-culture
with a fixed number of rat astrocytes (15K per well). As
visualized in Figure 3A, fluorescence intensity within the
range image strongly correlates with cell density, with the
highest amount of activity observed at 40K neurons/well.
The range image also provides the researcher with a
qualitative assessment of morphology, toxicity, and
transduction efficiency. Summary Ca2+ traces of neuronal
activity provide a quantitative assessment of activity within
each well, and density of neurons tested was optimal for
visualization of neuronal activity within each scan (Figure
3B) and detection of active objects over the full 12-day
time course (Figure 3C).
5
Functional Profiling of Different iPSC-derived Neurons
Using the Incucyte® Live-Cell Analysis System and the
Incucyte® Neuroburst Orange Lentivirus, we evaluated
four different types of iPSC-derived neurons over
30–50 days in culture. These included iCell® GlutaNeurons
(Figure 4A), iCell® GABANeurons (Figure 4B), iCell®
DopaNeurons (Figure 4C) co-cultured with primary rat
astrocytes, as well as CNS.4U neurons (Figure 4D). iCell®
GlutaNeurons, described as human glutamatergicenriched
cortical neurons derived from iPSCs, displayed a
rapid induction of calcium burst activity in >1500 cells that
became highly correlated within 10 days of co-culture.
iCell® GABANeurons, characterized as a culture of >95%
pure population of GABAergic (inhibitory) neurons, also
displayed a rapid increase in the number of cells with
calcium burst activity within the first week of co-culture.
However, iCell® GABANeurons did not display significant
correlation at any time-point tested, in line with their
inhibitory phenotype. A closer examination of cellular
activity at Day 14, displayed as object traces over the full
3 minute scan (Figure 4A and 4B), supports the
observation of a significant number of active cells in both
the iCell® Gluta- and GABANeurons; the former displaying
higher calcium burst intensity and synchronicity when
compared to the latter. Interestingly, the kinetics of iCell®
DopaNeuron activity was strikingly similar to iCell®
GlutaNeurons, illustrating a very rapid induction of highly
active, highly correlated networks within the first 10 days of
culture. Ncardia® CNS.4U cells represent an in vitro coculture
model of hiPSC-derived neurons and astrocytes.
These cells showed significant activity from nearly 1200
cells within the first week of culture and an increase in
correlated activity (network connectivity) at approximately
Day 34 in culture, reaching a correlation of 0.7 at Day 45
when the experiment was terminated.
0
0 0
0 0
0
Active
Object #
Correlation
Active
Object #
Correlation
Active
Object #
Correlation
Active
Object #
Correlation
30
100 120 140 160 180 100 120 140 160 180
30 30
10 30
40 40
10 10
5 10
20 20
5 5
15 5
60 80 60 80
15 15
20 15
20 20
25 20
25 25
25
Days
Time (sec) Time (sec)
Days Days
Days
1.0
0.8
0.6
0.4
0.2
0.0
2.8
2
1
0
2.8
2
1
0
1.0
0.8
0.6
0.4
0.2
0.0
1.0
0.8
0.6
0.4
0.2
0.0
1.0
0.8
0.6
0.4
0.2
0.0
2000
1500
1000
500
0
3000
2000
1000
0
1500
1000
500
0
1000
800
600
400
200
0
Figure 4: Functional Activity of
Different iPSC-derived
Neurons
iCell® GlutaNeurons, iCell® GABANeurons,
iCell® DopaNeurons (Cellular Dynamics
International) and CNS.4U neurons
(Ncardia®) were all seeded at 20K cells/well.
iCell® GlutaNeurons, iCell® GABANeurons
and iCell® DopaNeurons were also plated
with a co-culture of rat astrocytes seeded at
15K cells/well. Neurons were infected with
Incucyte® Neuroburst Orange Lentivirus,
and Active Object Number and Mean
Correlation were quantified for up to
45 days. Example calcium oscillation traces
and kinetic graphs of activity metrics over
time for iCell® GlutaNeurons (A) and iCell®
GABANeurons (B). Mean Correlation and
Active Object Count were quantified for
iCell® DopaNeurons (25 days) (C) and
CNS.4U neurons (45 days) (D). Data points
represent Mean ± SEM.
A. iCell® GlutaNeurons
C. iCell® DopaNeurons
Mean Correlation Intensity (OCU)
Intensity (OCU)
Mean Correlation
Mean Correlation Mean Correlation
Active Objects Active Objects
Active Objects Active Objects
B. iCell® GABANeurons
D. CNS.4U
Figure 5: Quantifying Pharmacological Neurotoxic Effects of Chemotherapeutic Taxol ®
Rat cortical neurons seeded at 30K cells/well were co-cultured with rat astrocytes seeded at 15K cells/well and transduced with Incucyte®
Neuroburst Orange or Neurolight Orange Lentivirus at Day 3 in culture. Live-cell analysis measurements were made each day using the Incucyte®
Live-Cell Analysis System. After 11 days, neural networks had fully formed and stabilized. Taxol® or vehicle control was then added and cultures
were monitored for an additional 11 days. Time courses of neurite development (A) and neuronal activity (B) prior to, and after the addition of,
control or increasing concentrations of Taxol® are shown. Potency (IC50 values) plotted against time post-treatment for neuronal activity (grey)
and neurite length (orange) (C). Data is expressed as % neurite length or active object count, normalized to the pre-treatment value. Data points
represent Mean ± SEM. Neuronal activity summary traces at pre-treatment and at 5, 10, and 15 days post-treatment display decreased activity
levels over the course of the experiment (D).
D.
Pre-treatment Drug: Day 5 Drug: Day 10 Drug: Day 15
10-6 M
10-7 M
10-8 M
10-9 M
10-10 M
10-11 M
10-12 M
Vehicle
0 7 14 21 0 7 14 21 2 4 6 8 10
Days Days Days
120
80
40
0
120
80
40
0
Neurite Length
Neuronal Activity
1000
100
10
A. B. C.
Normalized Neurite Length
Normalized Active Objects
IC50 (pM)
6
Insights Into Structure-Function Quantification
Peripheral neuropathies are a common side effect of
chemotherapeutic drugs such as paclitaxel (Taxol®)
and are associated with numbness and loss of sensory
function. To study potential neurotoxic effects, primary
rat cortical neurons were first co-cultured with primary
rat astrocytes for 11 days, allowing the cultures to
mature and stabilize. Baseline measurements of activity
and morphology were made each day using Incucyte®
Neuroburst Orange and Incucyte® Neurolight Orange
Lentivirus respectively (Figure 5). At Day 11, cultures
were treated with a range of concentrations of Taxol®.
Activity and morphology were monitored for a further
11 days in culture. Figure 5 illustrates that by Day 21, at
sub-nanomolar (<10-⁹ M) concentrations of Taxol® only
small changes in neurite length were observed, while a
reduction in neuronal activity occurred (Figure 5C).
Individual well traces indicated both concentrationand
time-dependent responses of neuronal activity
following Taxol® treatment as shown in Figure 5D.
Stem Cell Development Workflow
The development of advanced human cell-based models,
such as human iPSC-derived neurons and glia in monoculture
and co-culture, that are species- and -disease
relevant, have increased in recent years. Live-cell analysis
provides insights into these translational models and
enables the opportunity to identify novel pharmacological
treatments for neurodegenerative diseases.
Robust methods are needed when developing human
iPSC-derived models. This includes the characterization
of the cells and quality control (QC) of the final model.
Differentiation from Human iPSCs – Phase Images
Neural Maintenance Media
16-Day Neural Differentiation of iPSCs
Using Proprietary Methods (Based on Shi
et al., 2012 Nature Protocols 7:1836-46)
Monitor Neural Differentiation with Phase Microscope Day 30 Day 60
Transcriptomic Profiling Transcriptomic Profiling
Proteomic Profiling by
ICC Functional QC
Talisman Workflow
Transcriptomic Profiling Neurite Outgrowth
Late Born Upper ICC
Layer Neurons
and Ostrocytes
Day 60+
Early Born
Deep Layer
Neurons
Day 25
Cortical
Rosettes
Day 18
iPSCs
Day 0 Day 12
Figure 6: Differentiation of Human iPSC-Derived Neurons Workflow
Neuronal Activity
Intensity (OCU)
Time (sec)
0 30 60 90 120
6
5
43
2
1
0
7
Figure 6 illustrates the differentiation and characterisation
workflow developed at Talisman Therapeutics for use with
human iPSC-derived neurons. This workflow describes
where the use of both neurite outgrowth and neuronal
activity, as quantified on the Incucyte® Live-Cell Analysis
System can be integrated into the workflow alongside
other techniques. Once matured, these neurons are shown
to form active networks suitable to test novel treatments
for disease.
Glia Modulation
As opposed to the traditional view that brain function
results exclusively from neuronal activity, it is now widely
accepted as a more coordinated perspective involving
both neurons and glia. Astrocytes are regarded as active
partners in brain activity via bidirectional communication,
orchestrated at the tripartite synapse, composed of the
neuronal pre- and post-synapses and their close interaction
with the surrounded astroglia.
To investigate the effect of astrocytes in the signalling
response of neurons via measurements of calcium oscillations,
a humanized live-cell model of neuronal activity was
developed in collaboration with Talisman Therapeutics.
Recent advances in hiPSC offer a powerful in vitro model
strategy for the study of both healthy and disease stages
of the human nervous system. Non-perturbing neurite
outgrowth measurements can be performed in monoor
co-culture (post-infection with Incucyte® Neurolight
Orange Lentivirus) via automatic segmentation of timelapse
imaging using the Incucyte® Neurotrack Analysis
Software Module. Cell bodies and neurites are discriminated
and kinetically quantified. Co-cultures include interactions
observed in more complex systems that monocultures
are unable to capture. When co-cultured with astrocytes,
neurons developed greater and more branched neurites
compared to monocultures (Figure 7). The functional profile
of co-cultures also differed of that of monocultures, the
former showing greater active objects (1/image), burst
duration (sec), and lower burst rate (1/min), at a similar
correlation, indicating greater network stability in the
presence of glial cells. The presence of astrocytes impacts
neuronal architecture and network activity. Network activity
in neuron-astrocyte co-cultures differs from that seen in
neuronal monocultures, displaying a reduced frequency of
longer-lasting calcium events, characteristic of a more
mature neuronal network through the development
of a network’s ability to fire trains of action potentials.
Neurite Development
Time (h)
160
120
80
40
0
MC
CC
0 48 96 144 192 240
Neurite length (mm/mm²)
Time (h)
4
3
2
1
0
0 48 96 144 192 240
Branch Points (x10³/mm²)
Neural Activity
1000
750
500
250
0
Active Objects (1/Image)
MC
CC
10
8
6
4
2
0
Burst Rate (1/image)
1.0
0.8
0.6
0.4
0.2
0
ActiveMean Correlation
8
6
4
2
0
Burst Duration (sec)
Intensity (OCU)
Time (sec)
0 30 60 90 120
5
4
3
2
1
0
Mono-Culture
Intensity (OCU)
Time (sec)
0 30 60 90 120
5
4
3
2
1
0
Co-Culture
Figure 7: iPSC Neurons Co-cultured With Mature Astrocytes Yield Greater Outgrowth and Branching and Neuronal
Network Activity Is Modified.
Network activity in neuron-astrocyte co-culture differs from monocultures, displaying a reduced frequency of longer-lasting burst rates
(characteristic of a more mature neuronal network through the development of a network’s ability to fire trains of APs). n = 12. Traces and bar
charts are 23 days post-infection.
8
9
Conclusions
In this application note, we present data to support the
use of the Incucyte® Live-Cell Analysis System configured
with an Orange/NIR or Green/Orange/NIR Optical
Module to characterize and refine different neuronal
phenotypes and their maturation for modeling their
function in vitro. This single live-cell imaging platform, in
combination with the Incucyte® Neuroburst Orange
Lentivirus, allows users to assess calcium flux kinetics and
continuously monitor morphology of neuronal
populations long-term using non-perturbing reagents,
validated protocols that are cell-sparing, and a built-in,
guided interface for non-experts provided by the
Incucyte® Neuronal Activity Analysis Software Module.
The system can be used within the operator’s own
incubator under physiological conditions and may be used
with a variety of neuronal cell types (such as primary
neurons and iPSC-derived models).
As described above, this system and reagents can provide
valuable, “real world” kinetic insights into neuronal network
activity and connectivity in neurological models that might
be missed by traditional end-point analysis.
Knowing when iPSC-derived neurons become functionally
active, how to optimize their activity, and gaining insight into
the synaptic connectivity of cultured neurons has eluded
neuroscience researchers. Using predominantly GABA and
Gluta iPSC-derived neuronal models, we have shown how the
Incucyte® Live-Cell Analysis System provides a means to
study a variety of cell models using relevant quantitative
metrics (e.g. neurite length and cellular activity). Additionally,
these disease-relevant humanized models allow us to
investigate pharmacological modulation. Lastly, the
robustness and throughput provided by the Incucyte® Live-
Cell Analysis System enables researchers to focus on isolating
variables in order to improve iPSC-derived neuronal model
development.
The Incucyte® Live-Cell Analysis System provides a
complete end-to-end solution for the characterization of
neuronal phenotypes and their maturation, not only for
neuronal cell function, but also provides important
information for a wide variety of neurological questions
that may be missed by other methods.
Application Note
Optimization of iPSC Culture Protocol Using
High Quality Growth Factors and Cytokines
Daryl Cole, Amber Ward, Jasmine Trigg, Nicola Bevan
Sartorius UK Ltd, Royston, Hertfordshire, UK
Correspondence
Email: AskAScientist@sartorius.com
Abstract
Cytokines and growth factors constitute a diverse group of proteins essential for intercellular communication. Two cytokines
that are a key focus for in vitro cell culture are fibroblast growth factor 2 (FGF-2) and transforming growth factor β1 (TGF-β1).
These cytokines are used in precise concentrations for maintaining induced pluripotent stem cells (iPSCs) in their pluripotent
state. However, achieving the desired efficacy of these cytokines and growth factors in an in vitro setting has been
challenging due to issues like thermostability, leading to a short half-life and necessitating frequent media changes, which are
resource intensive.
In this application note, we present data on the utilization and optimization of Sartorius Research Use Only (RUO) Cytokines
and Growth Factors for iPSC culture. Our goal is to sustain pluripotency and proliferative potential while reducing the need
for daily media replenishment. We assess this by analyzing crucial cell surface markers, morphological indicators, and growth
patterns using advanced tools like the iQue®3 High-Throughput Screening Cytometer and the Incucyte® Live-Cell Analysis
System, showcasing the practicality of Sartorius RUO Growth Factors and Cytokines in iPSC culture maintenance.
November 1, 2023
Keywords or phrases:
Research Use Only (RUO), Cytokines, Growth Factors,
iPSC, Cell Culture, Pluripotency, Live-Cell Analysis,
Advanced Flow Cytometry, Immunocytochemistry
Daryle Cole, PhD — Scientist – Sartorius
Daryle Cole, PhD — Field Application Specialist – Sartorius
Jasmine Trigg — Scientist – Sartorius
Nicola Bevan — Scientist – Sartorius
2
Introduction
Cytokines are a large and diverse family of proteins secreted
by cells for intercellular communication. By binding to
extracellular receptors, they can induce an intracellular
cascade of signaling to initiate a biological response.
The induction and co-ordination of a range of processes
are attributed to the correct signal intensity and timing of
cytokine release, from embryonic development to growth
and wound healing. One group of cytokines, classified as
growth factors, are responsible for the growth of a specific
tissue by stimulating proliferation and differentiation.
The importance of these growth factors can be seen
in diseases where their strict regulation is lost, such as
rheumatoid arthritis, multiple sclerosis, and COVID-19.¹,²,³
There is often a specific growth factor that drives the
growth and differentiation of a defined tissue, for example
epidermal growth factor (EGF) enhances osteogenesis,
whilst fibroblast growth factor (FGF) and vascular endothelial
growth factor (VEGF) stimulate angiogenesis.⁴,⁵
Since these growth factors are required for maintaining
cell health in vivo, it is essential that cells cultured in vitro
also have access to physiological levels of growth factors.
Two cytokines that are a key focus for in vitro cell culture
are fibroblast growth factor 2 (FGF-2) and transforming
growth factor β1 (TGF-β1 PLUS). FGF-2 is mitogenic,
playing a significant role in physiological processes such
as embryonic development, tissue repair, and cell growth,
as well as pathological processes such as tumor growth,
perfusion, and invasion.⁶ Similarly, TGF-β1 PLUS plays a key
role in differentiation and oncogenesis and is associated with
Alzheimer’s disease, but is also secreted by leukocytes as a
key component of coordinating the immune response.⁷
Induced pluripotent stem cells (iPSCs) are an in vitro cell
model that require precise concentrations of these growth
factors to maintain their pluripotent properties. iPSCs start
as somatic cells that undergo a reprogramming process,
using cytokines to alter their cell fate and enable the cell
to gain pluripotent properties.⁸ Through further exposure
to growth factors that mimic the in vivo differentiation
process, theoretically these iPSCs can then differentiate
into any somatic cell type. iPSCs are valuable for studying
the differentiation process as they do not face the same
ethical concerns of protocols using embryonic stem cells.
The pluripotent qualities of a cell can be determined by
investigating cell surface markers, such as stage-specific
embryonic antigen (SSEA)-1, SSEA-4, TRA-1-60, and
intracellular markers such as SOX2 and OCT-4.⁹
Despite their importance in cell culture maintenance,
translating the efficacy of growth factors into an in vitro
environment has been challenging. Sensitive cell types, such
as iPSCs, require highly pure growth factors and cytokines as
even trace amounts of endotoxins and other impurities can
impact their genetic stability and differentiation potential.
While recombinant growth factors and cytokines are free
of contaminating animal proteins, thermostability can be
an issue, resulting in a short half-life. This means growth
factors in the cell culture media degrade faster, necessitating
regular media changing; both a time and resource-intensive
process.¹⁰ Furthermore, many growth factors become
compromised after three freeze-thaw cycles, complicating
their use in culture.¹¹
Sartorius Research Use Only (RUO) Growth Factors
and Cytokines are produced using recombinant human
DNA technology and do not contain any animal-derived
contaminants. They are also manufactured to the highest
quality standards to ensure purity and low endotoxicity.
In this application note, we demonstrate optimization of
culture conditions to maintain a pluripotent phenotype
and the proliferative capability of iPSCs using Sartorius
RUO Growth Factors and Cytokines, including thermostable
varieties. We also demonstrate the utility of the iQue®3
High-Throughput Cytometry Platform and Incucyte® Live-
Cell Analysis System for analyzing cell surface markers, cell
growth, and cell morphology.
Methods and Materials
Cell Culture
iPSCs were passaged using Accutase® to lift cells twice
per week in vitronectin (10 μg/mL) coated 6-well plates
at a density of 100 K/well. NutriStem® hPSC XF (Cat. No.
05-100-1A) pre-conditioned iPSCs (ATCC 1019) were
cultured in NutriStem® hPSC XF media, whereas iPSC 1019
cells were cultured in commercial media. After passaging,
cells were reseeded in media containing 10 μM ROCK
inhibitor (Y-27632), which was removed after 24 hours.
Growth Factors for Standard iPSC Culture
To optimize the concentration of RUO Recombinant
Human TGF-β1 PLUS Protein (Cat. No. CYK-0050-0010)
and Recombinant Human FGF-2 Protein 154 aa (Cat. No.
CYK-0100-0023) to be added to the growth factor-free
media, several concentrations of each growth factor were
tested. iPSCs were seeded in vitronectin (10 μg/mL) coated
24-well plates (100 K/well). Pluripotency was determined at
3 and 6 days post-seeding. A concentration range of FGF-2
was tested (3.13 – 200 ng/mL) in combination with 2 ng/mL
TGF-β1 PLUS in all conditions. Similarly, a concentration
range of TGF-β1 PLUS was tested (0.13 – 8 ng/mL) with
100 ng/mL FGF-2 in all conditions. NutriStem® hPSC XF
GF-free (Cat. No. 06-5100-01-1A), commercial media,
and RPMI 1640 were used as control media formulations.
THP-1 cells were used as a non-pluripotent biological
control. The cells were analyzed for marker expression using
the iQue® Advanced Flow Cytometry Platform.
Generating Concentration Response Curves
iPSCs were seeded at 20 K/well in vitronectin coated 24-well
plates and monitored over 6 days in the Incucyte® Live-
Cell Analysis System. The cells were fed daily with 500 μL
media containing a concentration range of TGF-β1 PLUS
and FGF-2. NutriStem® hPSC XF GF-free, NutriStem®
hPSC XF, commercial media, and RPMI 1640 were used
as controls. The cells were then lifted using Accutase® and
reseeded in a 96-well V-bottom plate, centrifuged at 300 g
for 5 minutes and washed twice with PBS + 5% FBS before
being treated with SSEA-1-FITC (1:50), SSEA-4-APC
(1:800), TRA-1-60-PE (1:25), and iQue® B/Red Viability Dye
(1:50) and incubated at room temperature in the dark for
30 minutes. The plate was then spun at 300 g for 5 minutes
before analyzing on the iQue®3 High-Throughput Cytometry
Platform.
FGF-2-G3 Long Term Comparison
Recombinant FGF2-G3 protein (Cat. No. CYK-0100-0002)
is an enhanced, thermostable version of FGF-2 designed to
enable longer duration between feeds. Cells were seeded
at a density of 20 K/well in vitronectin (10 μg/mL) coated
24-well plates. These cells were treated with a concentration
range of FGF2-G3 in combination with 2 ng/mL of TGF-β1
PLUS, without subsequent refeeds. NutriStem® hPSC XF
GF-free, stabilized commercial media (enabling longer
duration between feeds), and RPMI 1640 were used as
control media formulations. THP-1 cells were used as a
non-pluripotent biological control. After 6 days, the cells
were analyzed on the iQue®3 platform. Cells were
monitored using the Incucyte® Live-Cell Analysis System for
the duration of the experiment to investigate growth
dynamics and morphological changes.
Immunocytochemistry (ICC)
iPSCs were seeded at 2 K/well in a 96-well plate in ROCK
inhibitor (1:1000). After 24 hours, ROCK inhibitor was
removed and 100 μL NutriStem® XF was added to each
well. After a further 24 hours, a concentration range of
TGF-β1 PLUS was added alongside controls of NutriStem®
hPSC XF, NutriStem® hPSC XF GF-free, commercial media,
commercial media plus, and RPMI 1640. After 5 days, cells
were washed, fixed, permeabilized, and blocked. Cells were
then immunostained for markers of pluripotency (SSEA-4
Alexa Fluor™ 647, OCT-4 Alexa Fluor™ 555, and SOX2 Alexa
Fluor™ 488; ThermoFisher A24881) and imaged in PBS.
Phase and fluorescence images were acquired using the
Incucyte® system and analyzed using integrated software.
Results
Ensuring iPSCs are cultured optimally is essential for
successfully maintaining the pluripotency, health, and
longevity of these highly valuable cells. The advantage of
using iPSCs in research is in their differentiation capability,
where disparate tissue types can be derived from a
single cell. Due to their complex nature, the requirement
for specific culture conditions necessitates the use of a
variety of supplements in culture media to grow highly
pluripotent iPSCs.
The following data highlights the efficacy of Sartorius RUO
Growth Factors and Cytokines as supplements in media for
the successful culture of iPSCs.
Note: iPSCs were cultured in NutriStem® hPSC XF GF-free (NutriStem®-GF) supplemented with concentration ranges of Sartorius RUO FGF-2 and
TGF-β1 PLUS. Cells were harvested after 3 and 6 days and analyzed on the iQue®3 High-Throughput Screening Cytometer. (A) Pluripotent (SSEA-1 -,
SSEA-4 +, TRA-1-60 +) and (B) Non-pluripotent (SSEA-1 +) expression profile analysis of iPSCs supplemented with FGF-2. (C) Pluripotent and (D) Nonpluripotent
expression profile analysis of iPSCs supplemented with TGF-β1 PLUS. Data presented as mean ± SEM, n = 3.
Figure 1: Quantification of iPSC Marker Expression During Stem Cell Maintenance.
NutriStem®-GF
FGF-2 50 ng/mL
FGF-2 100 ng/mL
FGF-2 200 ng/mL
Commercial media
RPMI
THP-1
0
50
100
Pluripotent
% positive of live cells
A
FGF-2 and TGF-β1 PLUS Supplementation Maintains
Pluripotency in iPSCs
We grew iPSCs in NutriStem® hPSC XF GF-free with or
without supplementation with FGF-2 and TGF-β1 PLUS
over 3 and 6 days with daily feeds. Using the iQue®3 High-
Throughput Screening Cytometer, we analyzed changes in
cell surface pluripotency marker expression associated with
optimized growth conditions. The capabilities of the iQue®3
allow for multiple markers to be analyzed in the same sample
well, greatly increasing the speed of data collection and
saving precious sample. Comparisons of pluripotency
(SSEA-1 −, SSEA-4 +, TRA-1-60 +) and non-pluripotency
(SSEA-1 +) marker expression profiles
(Figure 1) highlight a decrease in pluripotency marker
expression when iPSCs are cultured in NutriStem® XF
GF-free for 6 days.
NutriStem®-GF
TGF-ß1 PLUS 2 ng/mL
TGF-ß1 PLUS 4 ng/mL
TGF-ß1 PLUS 8 ng/mL
Commercial media
RPMI
THP-1
0
50
100
Pluripotent
% positive of live cells
C
NutriStem®-GF
FGF-2 50 ng/mL
FGF-2 100 ng/mL
FGF-2 200 ng/mL
Commercial media
RPMI
THP-1
0
50
100
Non-pluripotent
% positive of live cells
B
NutriStem®-GF
TGF-ß1 PLUS 2 ng /mL
TGF-ß1 PLUS 4 ng/mL
TGF-ß1 PLUS 8 ng/mL
Commercial media
RPMI
THP-1
0
50
100
Non-pluripotent
% positive of live cells
D
3 days 6 days
3 days 6 days
3 days 6 days
3 days 6 days
4
Results
Ensuring iPSCs are cultured optimally is essential for
successfully maintaining the pluripotency, health, and
longevity of these highly valuable cells. The advantage of
using iPSCs in research is in their differentiation capability,
where disparate tissue types can be derived from a
single cell. Due to their complex nature, the requirement
for specific culture conditions necessitates the use of a
variety of supplements in culture media to grow highly
pluripotent iPSCs.
The following data highlights the efficacy of Sartorius RUO
Growth Factors and Cytokines as supplements in media for
the successful culture of iPSCs.
Note: iPSCs were cultured in NutriStem® hPSC XF GF-free (NutriStem®-GF) supplemented with concentration ranges of Sartorius RUO FGF-2 and
TGF-β1 PLUS. Cells were harvested after 3 and 6 days and analyzed on the iQue®3 High-Throughput Screening Cytometer. (A) Pluripotent (SSEA-1 -,
SSEA-4 +, TRA-1-60 +) and (B) Non-pluripotent (SSEA-1 +) expression profile analysis of iPSCs supplemented with FGF-2. (C) Pluripotent and (D) Nonpluripotent
expression profile analysis of iPSCs supplemented with TGF-β1 PLUS. Data presented as mean ± SEM, n = 3.
Figure 1: Quantification of iPSC Marker Expression During Stem Cell Maintenance.
NutriStem®-GF
FGF-2 50 ng/mL
FGF-2 100 ng/mL
FGF-2 200 ng/mL
Commercial media
RPMI
THP-1
0
50
100
Pluripotent
% positive of live cells
A
FGF-2 and TGF-β1 PLUS Supplementation Maintains
Pluripotency in iPSCs
We grew iPSCs in NutriStem® hPSC XF GF-free with or
without supplementation with FGF-2 and TGF-β1 PLUS
over 3 and 6 days with daily feeds. Using the iQue®3 High-
Throughput Screening Cytometer, we analyzed changes in
cell surface pluripotency marker expression associated with
optimized growth conditions. The capabilities of the iQue®3
allow for multiple markers to be analyzed in the same sample
well, greatly increasing the speed of data collection and
saving precious sample. Comparisons of pluripotency
(SSEA-1 −, SSEA-4 +, TRA-1-60 +) and non-pluripotency
(SSEA-1 +) marker expression profiles
(Figure 1) highlight a decrease in pluripotency marker
expression when iPSCs are cultured in NutriStem® XF
GF-free for 6 days.
NutriStem®-GF
TGF-ß1 PLUS 2 ng/mL
TGF-ß1 PLUS 4 ng/mL
TGF-ß1 PLUS 8 ng/mL
Commercial media
RPMI
THP-1
0
50
100
Pluripotent
% positive of live cells
C
NutriStem®-GF
FGF-2 50 ng/mL
FGF-2 100 ng/mL
FGF-2 200 ng/mL
Commercial media
RPMI
THP-1
0
50
100
Non-pluripotent
% positive of live cells
B
NutriStem®-GF
TGF-ß1 PLUS 2 ng /mL
TGF-ß1 PLUS 4 ng/mL
TGF-ß1 PLUS 8 ng/mL
Commercial media
RPMI
THP-1
0
50
100
Non-pluripotent
% positive of live cells
D
3 days 6 days
3 days 6 days
3 days 6 days
3 days 6 days
5
In previous experiments, iPSCs cultured in RPMI presented a
loss in pluripotency marker expression and a gain in SSEA-1
expression, so this condition was used as a non-optimized
control. High expression of SSEA-1 combined with low
levels of pluripotency marker expression were seen in
non-pluripotent iPSC controls; grown in RPMI, or the THP-1
non-pluripotent immortalized cell line. Supplementation
with FGF-2 (50 - 200 ng/mL) showed a marked,
concentration-dependent increase in pluripotency marker
expression (Figure 1A), while SSEA-1 expression remained
constant (Figure 1B). We observed a similar trend when
supplementing with TGF-β1 PLUS; pluripotency marker
expression maintained at 6 days in cells supplemented
with concentrations as low as 2 ng/mL (Figure 1C). SSEA-1
expression also remains low throughout the study timeline
(Figure 1D).
iPSCs Supplemented with FGF-2 and TGF-β1 PLUS
Display Pluripotent Morphological Phenotypes
In addition to phenotypic marker analysis using the iQue®3
High-Throughput Screening Cytometer, it is also important
to assess the morphological characteristics of iPSCs to
provide more insight into the condition and growth of
cultures. Using the Incucyte® Live-Cell Analysis System, we
were able to compare different media for their impact on
the growth and morphology of the live iPSCs directly from
the incubator, without perturbing the cells (Figure 2). Highdefinition
phase contrast images of iPSCs grown in
NutriStem®
XF–GF show low confluency in the plate, with visible
spontaneous differentiation. Following addition of FGF-2
or TGF-β1 PLUS, iPSC morphology changed, with higher
confluency, increased colony density, and no spontaneous
differentiation. The morphology was comparable to cells
grown in NutriStem® XF. The RPMI control illustrated that
the morphological presentation associated with a loss of
pluripotency; the colony density and compactness was
reduced, and the cellular cytoplasmic area was increased.
Figure 2: Morphological Analysis of iPSCs During Stem Cell Maintenance.
Note: iPSCs were cultured in NutriStem® XF–GF supplemented with a range of concentrations of Sartorius RUO Recombinant Human FGF-2 and
TGF-β1 PLUS. Cells were monitored over 6 days and images acquired using the Incucyte® Live-Cell Analysis System. Representative 10X phase contrast
images taken at 2 days and 10 hours post-treatment.
200 ng/mL FGF-2 NutriStem® XF-GF Commercial Media
2 ng/mL TGF-ß1 PLUS NutriStem® XF RPMI
Growth Rate and Confluency of iPSCs Supplemented With
FGF-2 and TGF-β1 PLUS is Concentration-Dependent
The acquisition of kinetic data on the Incucyte® Live-Cell
Analysis System enabled easy quantification of iPSC
growth via confluency and phase object area analysis
(Figure 3). Here we showed the difference in iPSC growth
when cultured with varying concentrations of FGF-2 and
TGF-β1 PLUS (Figure 3). The data shows that in higher
concentrations of FGF-2 (50 – 200 ng/mL), iPSC growth
is similar with a plateau at 3 days, which remains stable
(Figure 3A). In contrast, data for lower FGF-2 concentrations
(3.13 – 25 ng/mL), show a dramatic drop in confluency post
three days, indicating a lack of growth and potential cell
death. Analysis of average phase object area (size of iPSC
colonies) over time provided more granular indication of
iPSC growth patterns.
Note: iPSCs were cultured in NutriStem® XF–GF supplemented with a range of concentrations of Sartorius RUO Recombinant Human FGF-2 and
TGF-β1 PLUS. Cells were monitored over 6 days and analyzed using the Incucyte® Live-Cell Analysis System. Confluency analysis of iPSCs supplemented
with (A) FGF-2 at decreasing concentrations and (C) TGF-β1 PLUS at decreasing concentrations. Phase Area Object Average analysis at 5 days for
(B) FGF-2 and (D) TGF-β1 PLUS.
Figure 3: Growth Analysis of iPSCs During Stem Cell Maintenance.
0
40
100
Phase Object Confluence (%)
80
60
20
1
A
2 3 4 5
Days
NutriStem® XF
FGF-2 200 ng/mL
FGF-2 100 ng/mL
FGF-2 50 ng/mL
FGF-2 25 ng/mL
FGF-2 12.5 ng/mL
FGF-2 6.25 ng/mL
FGF-2 3.13 ng/mL
NutriStem® XF-GF
For example, lower concentrations of FGF-2
(3.13–25 ng/mL) produced iPSCs that form smaller
colonies after 5 days, compared to higher concentrations
of supplemented FGF-2 (25–200 ng/mL) (Figure 3B).
TGF-β1 PLUS supplementation at a range of concentrations
(Figure 3C), had minimal impact on the growth of iPSCs,
indicating this protein is not as critical as FGF-2 for iPSC
growth at this concentration range. Analysis of average
phase object area, however, revealed that with decreasing
concentrations of TGF-β1 PLUS, iPSCs form smaller colonies
over time. Although growth is minimally affected, colony size
varies depending on TGF-β1 PLUS concentration (Figure
3D). Based on this data, we recommend the concentrations
of 100 ng/mL FGF-2 and 2 ng/mL TGF-β1 PLUS for optimal
growth and maintenance of iPSCs.
0
40
100
Phase Object Confluence (%)
80
60
20
1
C
2 3 4 5
Days
NutriStem® XF
TGF-1 PLUS 8 ng/mL
TGF-1 PLUS 4 ng/mL
TGF-1 PLUS 2 ng/mL
TGF-1 PLUS 1 ng/mL
TGF-1 PLUS 0.5 ng/mL
TGF-1 PLUS 0.25 ng/mL
TGF-1 PLUS 0.13 ng/mL
NutriStem® XF-GF
NutriStem® XF-GF
FGF-2 3.13 ng/mL
Phase Area Object Average (m²)
NutriStem® XF
B
FGF-2 6.25 ng/mL
FGF-2 12.5 ng/mL
FGF-2 25 ng/mL
FGF-2 50 ng/mL
FGF-2 100 ng/mL
FGF-2 200 ng/mL
0
5×106
1×106
1.5×106
2×106
2.5×106
NutriStem® XF-GF
TGF-ß1 PLUS 0.13 ng/mL
Phase Area Object Average (m²)
NutriStem® XF
D
TGF-ß1 PLUS 0.25 ng/mL
TGF-ß1 PLUS 0.5ng/mL
TGF-ß1 PLUS 1ng/mL
TGF-ß1 PLUS 2 ng/mL
TGF-ß1 PLUS 4 ng/mL
TGF-ß1 PLUS 8ng/mL
0
5×106
1×106
1.5×106
2×106
2.5×106
6
Growth Rate and Confluency of iPSCs Supplemented With
FGF-2 and TGF-β1 PLUS is Concentration-Dependent
The acquisition of kinetic data on the Incucyte® Live-Cell
Analysis System enabled easy quantification of iPSC
growth via confluency and phase object area analysis
(Figure 3). Here we showed the difference in iPSC growth
when cultured with varying concentrations of FGF-2 and
TGF-β1 PLUS (Figure 3). The data shows that in higher
concentrations of FGF-2 (50 – 200 ng/mL), iPSC growth
is similar with a plateau at 3 days, which remains stable
(Figure 3A). In contrast, data for lower FGF-2 concentrations
(3.13 – 25 ng/mL), show a dramatic drop in confluency post
three days, indicating a lack of growth and potential cell
death. Analysis of average phase object area (size of iPSC
colonies) over time provided more granular indication of
iPSC growth patterns.
Note: iPSCs were cultured in NutriStem® XF–GF supplemented with a range of concentrations of Sartorius RUO Recombinant Human FGF-2 and
TGF-β1 PLUS. Cells were monitored over 6 days and analyzed using the Incucyte® Live-Cell Analysis System. Confluency analysis of iPSCs supplemented
with (A) FGF-2 at decreasing concentrations and (C) TGF-β1 PLUS at decreasing concentrations. Phase Area Object Average analysis at 5 days for
(B) FGF-2 and (D) TGF-β1 PLUS.
Figure 3: Growth Analysis of iPSCs During Stem Cell Maintenance.
0
40
100
Phase Object Confluence (%)
80
60
20
1
A
2 3 4 5
Days
NutriStem® XF
FGF-2 200 ng/mL
FGF-2 100 ng/mL
FGF-2 50 ng/mL
FGF-2 25 ng/mL
FGF-2 12.5 ng/mL
FGF-2 6.25 ng/mL
FGF-2 3.13 ng/mL
NutriStem® XF-GF
For example, lower concentrations of FGF-2
(3.13–25 ng/mL) produced iPSCs that form smaller
colonies after 5 days, compared to higher concentrations
of supplemented FGF-2 (25–200 ng/mL) (Figure 3B).
TGF-β1 PLUS supplementation at a range of concentrations
(Figure 3C), had minimal impact on the growth of iPSCs,
indicating this protein is not as critical as FGF-2 for iPSC
growth at this concentration range. Analysis of average
phase object area, however, revealed that with decreasing
concentrations of TGF-β1 PLUS, iPSCs form smaller colonies
over time. Although growth is minimally affected, colony size
varies depending on TGF-β1 PLUS concentration (Figure
3D). Based on this data, we recommend the concentrations
of 100 ng/mL FGF-2 and 2 ng/mL TGF-β1 PLUS for optimal
growth and maintenance of iPSCs.
0
40
100
Phase Object Confluence (%)
80
60
20
1
C
2 3 4 5
Days
NutriStem® XF
TGF-1 PLUS 8 ng/mL
TGF-1 PLUS 4 ng/mL
TGF-1 PLUS 2 ng/mL
TGF-1 PLUS 1 ng/mL
TGF-1 PLUS 0.5 ng/mL
TGF-1 PLUS 0.25 ng/mL
TGF-1 PLUS 0.13 ng/mL
NutriStem® XF-GF
NutriStem® XF-GF
FGF-2 3.13 ng/mL
Phase Area Object Average (m²)
NutriStem® XF
B
FGF-2 6.25 ng/mL
FGF-2 12.5 ng/mL
FGF-2 25 ng/mL
FGF-2 50 ng/mL
FGF-2 100 ng/mL
FGF-2 200 ng/mL
0
5×106
1×106
1.5×106
2×106
2.5×106
NutriStem® XF-GF
TGF-ß1 PLUS 0.13ng/mL
Phase Area Object Average (m²)
NutriStem® XF
D
TGF-ß1 PLUS 0.25 ng/mL
TGF-ß1 PLUS 0.5 ng/mL
TGF-ß1 PLUS 1ng/mL
TGF-ß1 PLUS 2 ng/mL
TGF-ß1 PLUS 4 ng/mL
TGF-ß1 PLUS 8ng/mL
0
5×106
1×106
1.5×106
2×106
2.5×106
Pluripotency marker expression was higher in all cells treated
with FGF2-G3 compared to all other treatment types, with
a consistent level across the concentration range (~70%)
outperforming an alternative commercial media (Figure 4A).
Comparisons of SSEA-1 expression show very low levels in
iPSCs cultured with FGF2-G3 from 50–200 ng/mL, lower
than NutriStem® hPSC XF GF-free and NutriStem hPSC
XF (Figure 4B). Control cells grown in RPMI, or THP-1 cell
controls, presented high expression of SSEA-1 and low levels
of pluripotency marker expression as seen previously. This
data indicates that thermostable Sartorius RUO FGF2-G3
maintains function in culture for longer periods, mitigating
the need for daily media changes.
Note: iPSCs were cultured in NutriStem® XF–GF supplemented with thermostable FGF2-G3 at a range of concentrations for 5 days without media
changes and pluripotency markers were analyzed on the iQue®3 High-Throughput Screening Cytometer on day 6. (A) Pluripotent (SSEA-1, SSEA-4 +,
TRA-1-60 +) and (B) Non-pluripotent (SSEA-1 +) marker expression profile. Stabilized CM (Stabilized Commercial Media enabling extended duration
between feeds). Representative data from one of 3 experiments. Data presented as mean ± SEM, n = 3.
NutriStem® XF-GF
FGF2-G3 50 ng/mL
FGF2-G3 100 ng/mL
FGF2-G3 200 ng/mL
Stabilized CM
RPMI
THP-1
0
20
40
60
80
100
Use of Thermostable FGF2-G3 Facilitates iPSC Culture
Without Daily Feeding
iPSCs are high maintenance and require daily media
refreshing to maintain a pluripotent phenotype.
Thermostable growth factors enable reduced media
change frequency due to increased compound stability,
which eliminates the need for inconvenient weekend media
changes. To test this, we grew iPSCs in NutriStem® hPSC
XF GF-free with supplementation (2 ng/mL TGF-β1 PLUS,
varying concentrations of FGF2-G3) for 5 days without
media changes and analyzed marker expression on the
iQue®3 platform at day 6. The Sartorius RUO FGF2-G3 is
an enhanced, thermostable version of FGF-2 designed to
enable longer duration between feeds.
Figure 4: Long Term iPSC Culture Without Media Change.
A Pluripotent
% positive of live cells
NutriStem® XF-GF
FGF2-G3 50 ng/mL
FGF2-G3 100 ng/mL
FGF2-G3 200 ng/mL
Stabilized CM
RPMI
THP-1
0
20
40
60
80
100
B Non-pluripotent
% positive of live cells
Morphological Analysis of iPSCs Supplemented With
FGF2-G3
We monitored the iPSCs supplemented with thermostable
FGF2-G3 on the Incucyte® Live-Cell Analysis System
over the 5-day study to investigate morphological effects.
Representative images of cells cultured in FGF2-G3 PLUS
supplemented NutriStem® hPSC XF GF-free showed
formation of very tightly condensed colonies with defined
edges and minimal cell death (Figure 5A). While iPSCs grown
in NutriStem® hPSC XF GF-free without supplementation
presented reduced growth with more cell death (Figure 5B).
Cells grown in commercial media produced very tight
colonies with defined edges, but with some spontaneous
differentiation (data not shown).
Analysis of the Incucyte® images for iPSCs grown in
FGF2-G3 supplemented media for 5 days without daily
media refeeds highlighted changes in growth compared
to alternative media types.
Note: iPSCs were cultured in NutriStem® hPSC XF GF-free and
supplemented with FGF2-G3. Cells were monitored over 5 days and
imaged and analyzed on the Incucyte® Live-Cell Analysis System.
Representative 10X phase contrast images taken at 2 days and 10 hours
post treatment of iPSCs treated with (A) NutriStem® hPSC XF GF-free
supplemented with 200 ng/mL FGF2-G3 and (B) NutriStem® hPSC XF
GF-free. (C) Confluency analysis of iPSCs supplemented with FGF2-G3 at
decreasing concentrations.
Figure 5: Morphological Visualization and Long-Term Growth Analysis of iPSCs Supplemented With FGF2-G3
Without Media Changes.
For example, iPSCs grown in NutriStem® hPSC XF GF-free
supplemented with FGF2-G3 exhibited increased growth
over NutriStem® hPSC XF and NutriStem® hPSC XF
GF-free cultured iPSCs (Figure 5C). NutriStem® hPSC
XF outperformed NutriStem® hPSC XF GF-free, but the
differences were less marked at the end of the time course,
suggesting degradation of key growth factors, such as
FGF-2. Culturing iPSCs in alternative commercial media
designed for weekend feeding provided similar growth to
FGF2-G3 supplementation, however, these cells plateaued
sooner, suggesting exhaustion of growth factors required for
continued growth (data not shown).
Interestingly, lower concentrations of FGF2-G3
supplementation (50 and 100 ng/mL) achieved the same
effect as the highest concentration (200 ng/mL) in analyses
on both the iQue®3 and Incucyte® platforms, suggesting
that the potency of this growth factor allows for lower
dosing, thus saving reagent.
0
8
Phase Object Confluence
Normalized to pre-treatment
6
4
2
1
C
2 3 4 5
Days
FGF2-G3 200 ng/mL
FGF2-G3 100 ng/mL
FGF2-G3 50 ng/mL
NutriStem® XF-GF
NutriStem® XF
A 200 ng/mL FGF2-G3 B NutriStem® XF-GF
0
8
Phase Object Confluence
Normalized to pre-treatment
6
4
2
1
C
2 3 4 5
Days
FGF2-G3 200 ng/mL
FGF2-G3 100 ng/mL
FGF2-G3 50 ng/mL
NutriStem® XF-GF
NutriStem® XF
Determining Pluripotency Marker Expression in iPSCs
Using Immunocytochemistry
To gain further insight about the efficacy of Sartorius RUO
Growth Factors and Cytokines, we cultured iPSCs in RPMI
and NutriStem® hPSC XF GF-free supplemented with
100 ng/mL FGF-2 and 2 ng/mL TGF-β1 PLUS followed
by staining to observe expression of key markers for
pluripotency in different culture conditions. Our data showed
that when supplemented with Sartorius RUO Growth Factors
and Cytokines, iPSCs expressed high levels of pluripotency
markers: the cell surface marker SSEA-4, and sub-cellular
markers SOX2 and OCT-4. However, cells grown in RPMI
displayed much lower levels of expression, as expected
(Figure 6A). The Incucyte® Live-Cell Analysis System
facilitated our investigation of the marker expression pattern
in iPSCs grown under different conditions.
SSEA-4 expression, for example, highlights the differences
in distribution in pluripotent iPSCs grown with FGF-2 and
TGF-β1 PLUS compared to RPMI. Low levels of expression
were evident in RPMI cultured iPSCs, while a high level of
expression throughout the surface of the cell was clearly
visible in growth factor supplemented iPSCs.
Quantification of fluorescence area normalized to phase
area to account for differences in cell area revealed global
differences in marker expression across each condition
(Figure 6B). SSEA-4 expression was approximately 30%
higher in supplemented iPSCs compared to RPMI, while
OCT-4 and SOX2 expression was approximately 10% higher
in supplemented iPSCs. These data support previous analysis
of marker expression on the iQue®3 High-Throughput
Cytometry Platform, indicating an increased level of
pluripotency in iPSCs supplemented with Sartorius RUO
Growth Factors and Cytokines compared to other media
conditions.
Figure 6: Immunocytochemistry Analysis of Pluripotency Marker Expression in iPSCs Supplemented
With FGF-2 and TGF-β1 PLUS.
Note: iPSCs were cultured in NutriStem® hPSC XF GF-free and
supplemented with 100 ng/mL FGF-2 and 2 ng/mL TGF-β1 PLUS or RPMI
for 2 days. Cells were fixed then stained for SSEA-4, OCT-4 and SOX2 and
imaged and analyzed on the Incucyte® Live-Cell Analysis System.
(A) Representative 20X fluorescence images taken of iPSCs in the
indicated conditions. (B) Analysis of Incucyte® images comparing the
percentage of fluorescence area normalized to phase area for each marker
under each condition. Representative data from one of 3 experiments.
Data presented as mean ± SEM, n = 3.
A SSEA-4 OCT-4
100 ng/mL FGF-2 +
2 ng/mL TGF-ß1 PLUS
SOX2 Merged
SSEA-4 OCT-4
RPMI
SOX2 Merged
SSEA-4 OCT-4 SOX2
0
10
20
30
40
50
60
B
Fluorescence Area/Phase Area (%)
NutriStem® XF + Supplements RPMI
Specifications subject to change without notice.
©2023. All rights reserved. All names of Sartorius products are registered trademarks and the property of Sartorius AG and/or one of its affiliated companies.
Filename: Optimization-of-iPSC-Culture-Protocol-RUO-Cytokines-Application-Note-en-Sartorius
Status: 10 | 2023
North America
Sartorius Corporation
300 West Morgan Road
Ann Arbor, Michigan 48108
USA
Phone +1 734 769 1600
Email: orders.US07@sartorius.com
Europe
Sartorius UK Ltd.
Longmead Business Centre
Blenheim Road
Epsom
Surrey, KT19 9QQ
United Kingdom
Phone +44 1763 227400
Email: euorders.UK03@sartorius.com
For further information, visit
www.sartorius.com
Asia Pacific
Sartorius Japan K.K.
4th Floor, Daiwa Shinagawa North Bldg.
1-8-11, Kita-Shinagawa 1-chome
Shinagawa-Ku
Tokyo 140-0001
Japan
Phone +81 3 6478 5202
Email: orders.US07@sartorius.com
Conclusion
iPSCs are rapidly becoming the cell type of choice for a
multitude of research and clinical fields; they are highly
proliferative and can be differentiated into all somatic tissues
in the human body. However, the requirements for successful
culture of these cells are both resource and time-intensive.
It is also of paramount importance to maintain pluripotency
during culture and finding the correct media formulation
is essential to achieving this goal. The data shown here
demonstrate the efficacy of Sartorius RUO Growth Factors
and Cytokines as supplements in iPSC media formulations
to preserve pluripotency, support growth, and increase the
interval between feeding, helping to remove the requirement
of daily media exchanges. Sartorius RUO Growth Factors
and Cytokines are free from animal-derived contaminants,
are manufactured to the highest quality standards ensuring
purity and low endotoxicity, providing excellent solutions
to challenging cell culture conditions. In addition, using the
iQue®3 High-Throughput Cytometry Platform in
conjunction with the Incucyte® Live-Cell Analysis System,
allows simple monitoring and quantification of growth and
pluripotent phenotype of iPSC cells during cell culture and
media formulation development.
References
1. Rosengren, S., Corr, M. and Boyle, D.L. (2010) ‘Plateletderived
growth factor and transforming growth factor
beta synergistically potentiate inflammatory mediator
synthesis by fibroblast-like synoviocytes’, Arthritis
Research & Therapy, 12(2). doi:10.1186/ar2981.
2. Lu, H. et al. (2021) ‘Growth factors and their roles in
multiple sclerosis risk’, Frontiers in Immunology, 12.
doi:10.3389/fimmu.2021.768682.
3. Ilias, I. et al. (2021) ‘Covid-19 and growth hormone/
insulin-like growth factor 1: Study in critically and noncritically
ill patients’, Frontiers in Endocrinology, 12.
doi:10.3389/fendo.2021.644055.
4. Platt, M.O. et al. (2009) ‘Sustained epidermal growth
factor receptor levels and activation by tethered
ligand binding enhances osteogenic differentiation of
multi-potent marrow stromal cells’, Journal of Cellular
Physiology, 221(2), pp. 306–317. doi:10.1002/jcp.21854.
5. Kovacevic, I., Hoffmeister, M. and Oess, S.
(2015) ‘Fibroblast growth factor signaling in
vascular development’, Endothelial Signaling
in Development and Disease, pp. 93–114.
doi:10.1007/978-1-4939-2907-8_4.
6. Itoh, N. et al. (2016) ‘Roles of FGF signals in heart
development, health, and disease’, Frontiers in
Cell and Developmental Biology, 4. doi:10.3389/
fcell.2016.00110.
7. Zhang, X., Huang, W.-J. and Chen, W.-W. (2016)
‘TGF-β1 factor in the cerebrovascular diseases of
Alzheimer’s disease’, European Review for Medical and
Pharmacological Sciences, 20(24), pp. 5178–5185.
8. Kogut, I. et al. (2018) ‘High-efficiency RNA-based
reprogramming of human primary fibroblasts’,
Nature Communications, 9(1). doi:10.1038/
s41467-018-03190-3.
9. Song, Y. et al. (2021) ‘Generation and characterization
of a human IPSC line HECI001-A from a healthy adult
donor pancreata’, Stem Cell Research, 56, p. 102541.
10. Liu, C. et al. (2021) ‘Cytokines: From clinical significance
to quantification’, Advanced Science, 8(15), p. 2004433.
doi:10.1002/advs.202004433.
11. Zhou, X. et al. (2010) ‘Conceptual and methodological
issues relevant to cytokine and inflammatory marker
measurements in clinical research’, Current Opinion in
Clinical Nutrition and Metabolic Care, 13(5), pp. 541–
547. doi:10.1097/mco.0b013e32833cf3bc.
Elevate Your Research With
High-Performance Media and Reagents
for Stem Cell Growth and Cultivation
Achieve consistent, scalable results in both 2D and 3D stem cell cultures using Sartorius’
range of high-quality growth factors, cytokines, transfection reagents, and animal-free
cell culture media. Our products meet stringent quality benchmarks, enhancing cell
health and easing upkeep, paving the way for a transition toward manufacturing.
Simplifying Progress
Specifications subject to change without notice. © 2024. All rights reserved. Incucyte and all names of Sartorius products
are registered trademarks and the property of Sartorius AG.
Specifications subject to change without notice. ©2024 All rights reserved. All names of Sartorius products are registered trademarks and the property of Sartorius AG and/or one of its affiliated companies.
Stem-cells-in-disease-modeling+research-ebook-2403-en-Sartorius Status: 03 | 2024
North America
Phone +1 734 769 1600
Europe
Phone +44 1763 227400
Email:
Japan
Phone +81 3 6478 5202
China
Phone +86 21 6878 2300
Rest of Asia Pacific and other
countries around the world:
Phone +65 6872 3966
Brought to you by
Download the eBook for FREE Now!
Information you provide will be shared with the sponsors for this content. Technology Networks or its sponsors may contact you to offer you content or products based on your interest in this topic. You may opt-out at any time.
Experiencing issues viewing the form? Click here to access an alternate version