biocompare_ebook-advances-in-cell-biology-research (2)

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 27

Contents

4 Novel Tools Advancing Cell Biology Research


Enabling products and technologies help answer new questions
and solve problems.

6 Discovery of Potential Soluble Epoxide


Hydrolase Inhibitors Using a High-Throughput
Screening Assay
sEH inhibitors are promising for treating inflammatory and
cardiovascular conditions.

14 Improved Live Cell Imaging Applications


Novel technology is an effective way for researchers to utilize
live cell imaging in their experiments.

22 Cryopreserved PBMCs Retain Phenotype


and Function
Viable cryopreserved PBMCs offer a convenient alternative to fresh PBMCs.

2
Produced with Support from

Biocompare Tel: 800-637-1277


395 Oyster Point Blvd., Suite 300 info@biocompare.com
South San Francisco, CA 94080 www.biocompare.com
Novel Tools Advancing
Cell Biology Research
Enabling products and technologies help answer new questions and
solve problems.

Cell biology research spans a broad range of areas The next article in this eBook hones in on an au-
including cell structure and function, cell develop- tomated high-throughput screening platform re-
ment, and cells and disease. There are a multitude cently developed to identify small molecule inhib-
of tools, technologies, and techniques available for itors. This automated HTS assay is shown to be an
investigating each of these topics, and in this eBook, efficient way to screen compound libraries against
we look at some of those resources and how they a specific target. In addition, with this automated
are being used to answer new questions, solve per- format, multiple assays can be easily modified to
plexing problems, and help researchers attain ex- encompass a wide range of target enzymes and
perimental success. compound library combinations.

High-throughput screening Live-cell imaging

High-throughput technologies have accelerated Advances in microscopy are helping scientists dis-
the discovery of small molecule drug candidates cover more than ever before at the cellular and mo-
by enabling the screening of libraries with millions lecular levels. One important “dimension” in mod-
of compounds to test their activity against selected ern microscopy is time. Being able to observe cell
targets. Yet because binding does not always pre- behavior over a continuous time frame can be use-
dict success, other parameters are now being con- ful, for example when observing responses to novel
sidered. The most commonly asked question these drug candidates. By contrast, getting a “snapshot”
days is “which compound can enter the relevant view at a single endpoint does not always convey
cell type, bind to the target, and revert the cell to a the whole story in terms of cellular response and
healthy phenotype?”. kinetics. That’s where live cell imaging comes in.

4
Live cell imaging tools have evolved significantly cancer research, as well as advanced therapies. In
and are becoming more accessible than ever be- vitro PBMC studies are crucial in cell biology and
fore. These technologies are not only of great inter- immunology research. PBMCs are also used in in
est to scientists working in the pharmaceutical and vivo studies, translational research, as well as dis-
biotech industries, they can be of great value when ease modeling and toxicity screening.
applied to drug discovery and development.
Frozen (cryopreserved) PBMCs offer many advan-
The third chapter in this eBook reviews the ef- tages over fresh PBMCs, enabling researchers to
fectiveness of live cell imaging for monitoring plan their experiments ahead of time with ready-
3D multicellular models, studying subcellular lo- to-use cells that can be stored in the freezer until
calization, and investigating cytotoxic activity of needed. One concern with frozen cells is the po-
immune cells. tential loss of viability, phenotype, or function re-
sulting from cryopreservation. In the final chapter
Cryopreserved peripheral blood in this eBook, we look at the characterization of
mononuclear cells frozen PBMCs as well as a direct comparison of T
cell activation responses through measurement of
Peripheral blood mononuclear cells, or PBMCs, are expression levels of T cell activation markers. The
peripheral blood cells that have one round nucleus, data shown suggest that functional, viable cryo-
including lymphocytes (T cells, B cells, and natural preserved PBMCs offer a convenient alternative to
killer cells), monocytes, and macrophages. They are fresh PBMCs in cell-based assays and immunolog-
used in drug discovery, diagnostic development, ical studies.

5
Discovery of Potential Soluble
Epoxide Hydrolase Inhibitors
Using a High-Throughput
Screening Assay
sEH inhibitors are a promising strategy for treating inflammatory
and cardiovascular conditions.

Key Features Introduction


• Soluble epoxide hydrolase (sEH) inhibitors Epoxyeicosatrienoic acids (EETs) are cytochrome
are a promising strategy for the treatment of P450 (CYP450) metabolites of arachidonic acid
inflammatory and cardiovascular conditions. formed by CYP450 epoxygenases and include four
regioisomers: 5(6)-, 8(9)-, 11(12)-, and 14(15)-EET.1
• We have developed an automated high-
EETs have been shown to have a wide range of
throughput screening (HTS) platform to
anti-inflammatory and cardioprotective effects.2
identify small molecule inhibitors that
However, EETs are rapidly hydrolyzed by soluble
enables testing of a large number of
epoxide hydrolase (sEH) to their respective dihy-
compounds in days instead of weeks using
droxyeicosatrienoic acids (DiHETrEs), a conversion
minimal sample volumes.
that is coincident with significant loss of their bio-
• We screened all 235 compounds in Cayman’s logical activity. Accordingly, the use of sEH inhib-
Anti-Inflammatory Screening Library and itors to increase EET accumulation is a promising
identified 36 compounds as potential sEH strategy for the treatment of inflammatory and car-
inhibitors using this approach. diovascular conditions.

• This HTS platform accelerates drug discovery. Here, a high-throughput screen (HTS) is de-
It can be adapted to many other enzyme scribed utilizing a modified version of Cayman’s
activity assays and performed using Cayman Soluble Epoxide Hydrolase Inhibitor Screening
or client-provided compound libraries. Assay Kit adapted for automation and increased

6
SIMPLE &
EFFECTIVE
ASSAYS
Explore more than 650 robust biochemical
and cell-based assay kits to measure:
· Cell viability
· Receptor activation
· Cellular metabolism
· Mitochondrial function
· Enzyme activity
· Oxidative stress
· Transcription factor activation
· Biomarker levels
www.caymanchem.com/assays

HAVE A CUSTOM PROJECT?


Contact our team of experts for assay design,
development, validation, automation, and manufacturing.
We can build quantitative assays on a variety of analysis
and detection platforms to suit your specific needs.
www.caymanchem.com/bioanalysis
throughput. By automating this assay, data that compounds supplied as 10 mM stock solutions in
would normally take 1-2 weeks to generate and DMSO in three 96-well Matrix™ tube racks. Com-
analyze are obtained over the course of three pounds were reformatted to a single, mapped 384-
days. Additionally, the use of automation drasti- well source plate for HTS. From the source plate,
cally improves reproducibility and reduces both seven barcode-labeled dilution plates were pro-
inter- and intra-assay variability. For this screen, duced, each containing vehicle, positive control,
Cayman’s Anti-Inflammatory Screening Library, and 10 pt ½ log concentration-response curves for
which consists of 235 biologically active and struc- 32 compounds. An additional dilution plate was
turally diverse anti-inflammatory compounds, prepared as described above with 16 compounds.
was evaluated. This screening library was chosen DMSO was used as the vehicle (negative) control,
due to the relevance of sEH as a therapeutic target and the sEH inhibitor AUDA was used as the pos-
itive control.
for inflammation.
Automated HTS Assays
Methods
A fully automated platform consisting of three ro-
Compound Library and Controls botic instruments was used for this HTS assay (Fig-
ure 1). All liquid handling steps were performed
An HTS of sEH inhibitors was performed in the using the Agilent Bravo liquid handler equipped
HTS facility at Cayman Chemical. Cayman’s An- with a 384 ST pipetting head. Assay and dilution
ti-Inflammatory Screening Library contains 235 plates were stored in separate stackers and placed

Figure 1. HTS assay workflow and reaction conditions. The Agilent Bravo automated liquid handler performs all liquid transfer steps in
the HTS assay. The platform is integrated with an Agilent BenchCel 4R microplate handler to enable automated microplate handling to
the BioTek Synergy Neo2 multimode microplate reader.

8
on the pipetting deck of the Agilent Bravo liquid where σp is the standard deviation of the positive
handler using the Agilent BenchCel 4R microplate control (AUDA), σn is the standard deviation of the
handler as required. Assay plates were transferred negative control (DMSO), µp is the mean of the pos-
from the deck of the Agilent Bravo liquid handler itive control, and µn is the mean of the negative
to the stage of a BioTek Synergy Neo2 multimode control. Assays with Z’-factors between 0.5 and 1
microplate reader for data acquisition. are considered excellent assays.
Test compounds were assayed in duplicate in black, Data Analysis
barcode-labeled 384-well non-binding plates. First,
human recombinant sEH was diluted in sEH assay Enzymatic rates were determined by calculating
buffer, and 39.5 µl of this solution was transferred the increase in fluorescence versus time from the
to each well of the assay plate. Next, 0.5 µl of test slope of the linear portion of the reaction. Maxi-
compound was transferred from the dilution plate mum fluorescence was obtained at 20 minutes.
to the assay plate. The reaction components were Percent activity values for test compounds were
preincubated for 5 minutes at room temperature. calculated based on wells containing the vehi-
Enzymatic reactions were initiated by the addi- cle control (100% activity) and wells containing
tion of 10 µl of the sEH substrate PHOME. Assay the positive control (sEH inhibitor AUDA). IC50 val-
plates were placed immediately on a BioTek Syn- ues were calculated using GraphPad Prism with a
ergy Neo2 multimode microplate reader set to four-parameter non-linear regression curve fit.
25°C using the Agilent BenchCel 4R microplate Some compounds exhibited background fluores-
handler. Kinetic measurements were obtained ev- cent properties at high concentrations. This was
ery 3 minutes for a total of 20 minutes using a 360 determined by plotting compound fluorescence at
nm excitation filter and 460 nm emission filter (360 the start of the assay versus compound concentra-
excitation/460 emission). Following completion, tion. Data were omitted in instances where com-
assay and dilution plates were stored in separate pound fluorescence caused significant interference
stackers. Enzyme and substrate stocks, as well as with the assay or data analysis. Data were retained
inhibitor and substrate pipet tips, were refreshed in instances where compound background fluo-
between plates. rescence caused no significant impact to the sEH
Quality Control inhibitory rate.

The Z’-factor is a widely used metric to evalu- Assay Performance


ate an assay’s suitability for HTS applications.3 It
measures assay robustness by determining the Representative vehicle and positive control re-
separation between the positive and negative sponses are shown in Figure 2. The average Z’-fac-
controls. The Z’-factor was calculated using the tor for all assays was 0.60, indicating robust as-
following equation: say performance. We achieved a throughput of
20 compounds/hour using the automated HTS
method described in the Methods section (data
not shown).

9
compounds in Figure 3. An IC50 value of 10 µM was
Results & Discussion
used as the baseline value to compare all com-
pounds. IC50 values below 10 µM were considered
IC50 values for sEH inhibition obtained in the HTS
potential sEH inhibitors whereas compounds with
assay are shown for all 235 anti-inflammatory IC50 values above 10 µM were considered unlikely
sEH inhibitors. 10 µM was chosen in order to cap-
ture weaker secondary target activity, as would be
anticipated with compounds with known primary
targets. In total, 36 anti-inflammatory compounds
were discovered with sEH IC50 values below 10 µM
in this HTS.

Six hit compounds that exhibited potent IC50 values


and/or clear concentration-response curves are
shown in Figure 4. The main anti-inflammatory tar-
gets, primary target potency, and sEH IC50 values of
these six hit compounds are summarized in Table 1.
Figure 2. Scatterplot showing the vehicle control (solid circles;
DMSO) and positive control (solid squares; AUDA) samples for
SC-75741, an NF-κB inhibitor, and trans-AUCB, a
the Z’-factor calculation of a representative 384-well plate in known sEH inhibitor, were identified as potent sEH
the sEH inhibitor HTS assay. Each point represents an individu- inhibitors in this HTS assay.4,5 Both compounds in-
al well on a single plate (n = 32 for each control). The solid lines hibited sEH with potency that exceeded the range
represent the mean fluorescence rates of the positive and neg- of concentrations tested (10 nM-10 µM). Hence, IC50
ative controls. The dashed lines indicate 3 standard deviations values for these compounds are indicated as <10
above or below the mean. The Z’-factor calculated for this was
nM. Given these potencies, these compounds are
data was 0.60, indicating excellent assay performance.
prime candidates for secondary screening.

Figure 3. Scatterplot of IC50 values for test compounds. The baseline value used to compare all compounds was 10 μM (-5 Log (M)). Com-
pounds with IC50 values less than 10 μM were considered potential sEH inhibitors, whereas compounds with IC50 values greater than 10
μM were considered unlikely inhibitors. Compounds circled in red exhibited potent IC50 values and/or clear concentration-response curves.

10
Figure 4. Concentration-response curves of select hit compounds. All data are plotted as mean ± standard deviation of two technical
replicates. The Z’-factor values for assay plates containing these compounds were >0.5. Compounds marked with a triple asterisk (***)
exhibited background fluorescent properties at high concentrations.

11
Table 1. Comparison of IC50 values for primary target and sEH of six hit compounds identified in this automated HTS assay.
*Data is shown for IC50 values unless otherwise indicated.

Ciclesonide is a prodrug of the glucocorticoid taglandins (PGs) from arachidonic acid, a family of
receptor agonist desisobutyryl ciclesonide.6 Glu- lipid signaling molecules with multiple inflamma-
cocorticoids are amongst the most well-known tory functions. Using dual inhibitors of COX-2 and
anti-inflammatory compounds. Despite cicle- sEH, potentially like DuP-697, to halt PG production
sonide displaying background fluorescence at concurrent with increasing tissue EET accumula-
high concentrations, we were able to determine tion is an intriguing approach for the treatment of
that ciclesonide has an IC50 value of 100 nM for inflammatory conditions.
the inhibition of sEH. Ciclesonide has limited
solubility in aqueous solutions. The higher com- LCK Inhibitor inhibits the Src-family non-receptor
pound concentrations used in this HTS assay protein tyrosine kinase LCK.9,10 LCK is expressed by
may have approached the limit of its aqueous T cells and mediates IL-2 production, a hallmark of
solubility, which could have contributed to high T cell activation, in response to T cell receptor sig-
background fluorescence. naling. Here, we discovered that LCK Inhibitor also
inhibits sEH activity (IC50 = 6,070 nM). Thus, it could
SCH 79797, an antagonist of proteinase-activated be of interest for future studies to determine if LCK
receptor 1 (PAR1), was found to have sEH inhibitory Inhibitor increases EET levels and if this is a viable
activity in this screen (IC50 = 372 nM).7 SCH 79797 strategy to dampen aberrant T cell activity.
inhibits PAR1 activation on vascular smooth mus-
cle cells and endothelial cells as well as α-throm-
bin-induced platelet aggregation. Whether or not
Conclusions
the sEH inhibitory activity of SCH 79797 contrib-
We have established an automated HTS platform
utes in part to these cardioprotective effects could
to identify small molecule inhibitors. This auto-
be explored in follow-up studies.
mated HTS assay is an efficient way to screen com-
DuP-697, a selective COX-2 inhibitor, was discov- pound libraries against a specific target. Given the
ered to be a potential sEH inhibitor (IC50 = 813 throughput, this assay could easily be applied to a
nM).8 This finding is especially intriguing given the larger compound library to increase the probabil-
extensive role of COX-2 in inflammation. COX-2 is ity for hits. With this automated format, multiple
primarily responsible for the production of pros- assays can be easily modified to encompass a wide

12
range of target enzymes and compound library 6. Belvisi, M.G., Bundschuh, D.S., Stoeck, M., et al.
combinations. This automated HTS assay is appli- Preclinical profile of ciclesonide, a novel corticosteroid
cable to basic research as well as drug discovery. for the treatment of asthma. J. Pharmacol. Exp. Ther.
314(2), 568-574 (2005).
References 7. Ahn, H.S., Foster, C., Boykow, G., et al. Inhibition of
cellular action of thrombin by N3-cyclopropyl-7-{[4-
1. Lai, J. and Chen, C. The role of epoxyeicosatrienoic
(1-methylethyl)phenyl]methyl}-7H-pyrrolo[3,2-f ]
acids in cardiac remodeling. Front. Physiol. 12, 642470
quinazoline-1,3-diamine (SCH 79797), a nonpeptide
(2021).
thrombin receptor antagonist. Biochem. Pharmacol.
2. Spector, A.A. and Kim, H.-Y. Cytochrome P450 60(10), 1425-1434 (2000).
epoxygenase pathway of polyunsaturated fatty acid
metabolism. Biochim. Biophys. Acta 1851(4), 356- 365 8. Kargman, S., Wong, E., Greig, G.M., et al. Mechanism
(2015). of selective inhibition of human prostaglandin G/H
synthase-1 and -2 in intact cells. Biochem. Pharmacol.
3. Zhang, J.-H., Chung, T.D.Y., and Oldenburg, K.R. A 52(7), 1113-1125 (1996).
simple statistical parameter for use in evaluation and
validation of high throughput screening assays. J. 9. Burchat, A.F., Calderwood, D.J., Hirst, G.C., et al.
Biomol. Screen. 4(2), 67-73 (1999). Pyrrolo[2,3-d]pyrimidines containing an extended
5-substituent as potent and selective inhibitors of lck
4. Leban, J., Baierl, M., Mies, J., et al. A novel class of II. Bioorg. Med. Chem. Lett. 10(19), 2171-2174 (2000).
potent NF-κB signaling inhibitors. Bioorg. Med. Chem.
Lett. 17(21), 5858-5862 (2007). 10. Arnold, L.D., Calderwood, D.J., Dixon, R.W., et al.
Pyrrolo[2,3-d]pyrimidines containing an extended
5. Hwang, S.H., Wagner, K.M., Morisseau, C., et al. 5-substituent as potent and selective inhibitors of lck
Synthesis and structure-activity relationship studies
I. Bioorg. Med. Chem. Lett. 10(19), 2167-2170 (2000).
of urea-containing pyrazoles as dual inhibitors of
cyclooxygenase-2 and soluble epoxide hydrolase. J. 11. Cayman Chemical Company. Unpublished internal
Med. Chem. 54(8), 3037-3050 (2011). company data. (2000).

Additional Resource
High-Throughput Screening

13
Improved Live Cell
Imaging Applications
Novel technology is an effective way for researchers to utilize live cell
imaging in their experiments.

To determine the cell type and number suitable


Introduction
for spheroid formation, two types of cells at vari-
A live cell imaging method that continuously cap- ous concentrations prepared by a FACSCOPE B cell
tures the images of cells over time has recently counter were seeded in a round bottom 96 well
been applied in various research and development plate to generate spheroids and then they were
settings. The purpose of this study was to check the observed using Celloger® Mini Plus.
effectiveness of the live cell imaging system on the As a result of comparing two types of cells, MDA-
following applications. MB-231, a human breast cancer cell line, and Hela
• Monitoring 3D multicellular model cell, a human cervical cancer cell line, the area oc-
cupied by spheroids was very different in the two
• Subcellular localization cells despite the same number of cells forming the
spheroids. Hela cells occupied a smaller area and ef-
• Cytotoxic activity of immune cell
fectively generated spheroids, expecting to be more
Topic 1. Monitoring 3D multicellular strongly aggregated than MDA-MB-231 (Figure 1).
model: Spheroid formation and screening
When spheroids are formed by aggregation of
3D (Three-dimensional) culture is one of the cell cells themselves, the degree of aggregation varies
culture methods that could overcome the limita- depending on the cell type, number of cells, and
tions of 2D culture by realizing an environment sim- culture method; thereby affecting the spheroid for-
ilar to an actual tissue. 3D culture without scaffold mation.2 If the changes in the spheroid area can be
lets cells form aggregation by simply increasing determined through real-time imaging, we could
cell-to-cell contact. A spheroid, the simplest struc- know when the spheroid formation is completed
ture formed by scaffold-free 3D culture, mimics a (the time when there are no more changes in area),
solid tumor in vivo and exhibits similar properties and select cells that aggregate more efficiently,
in terms of growth kinetics and metabolic rate.1,2 such as Hela cells. (Figure 2).

In this experiment, we tried to confirm the an- As a result of checking the spheroid area through
ti-cancer effect of nocodazole on the spheroid. time-dependent imaging after treatment with

14
Figure 1. Seeding concentration-dependent spheroid formation in two cell lines. Image captured by Celloger® Mini Plus (4X optics) when
MDA-MB-231 (upper) and Hela cells (lower) were seeded with 10,000, 5,000, and 1,000 cells per well in a round bottom 96-well plate and
then cultured for 26 hours.

nocodazole, an anti-cancer drug, it was found that the effect of drug appears is possible only through
the spheroid size continued to increase regardless real-time imaging. In the case of nocodazole treat-
of drug treatment until 18 hours. Spheroids not ment, the effect of the drug appeared after 18
treated with nocodazole continued to increase hours, which is presumably because it takes time
even after 18 hours, while in the case of nocodazole for apoptosis to begin after undergoing abnormal
treatment, it was found that it gradually decreased cell division. More details on apoptosis by noco-
after 18 hours (Figure 3). Identifying the time when dazole will be introduced in Topic 2.

Figure 2. Dynamics of cell coverage during spheroid formation. Both MDA-MB-231 and Hela cells, seeded at 5,000 cells per well, were
captured every 15min by Celloger® Mini plus (4X optics) and the area covered by a spheroid was estimated using Celloger® Mini Plus anal-
ysis app. A. Cell images showing the analyzed cell coverage in yellow. Images were captured immediately and 25 hours after seeding.
B. Changes in the cell coverage (average, N=3) during spheroid formation.

16
Topic 2. Subcellular localization: and as they undergo prophase stage, they gradu-
Observing the travel of nucleus in mitotic ally are condensed and the fluorescence becomes
arrested by an anti-cancer agent more intense compared to interphase (Figure 4A,
after 45 min). After passing through metaphase, in
To observe changes in intracellular structure and which the chromosomes are aligned in the center
protein movement, it is usually necessary to ex- of the cell (Figure 4A, 75 min~90 min), they are sep-
press a target protein fused with a fluorescent pro- arated into two sister chromatids, and two daugh-
tein. We conducted transfection with the plasmid ter cells are finally generated in telophase (Figure
containing the corresponding gene. Cells stably ex- 4A, after 105 min). On the other hand, after treat-
pressed green fluorescent protein fused histone 2B ment with nocodazole,4 which is known to induce
(H2B-GFP), which visualizes the dynamics of chro- cell cycle arrest, metaphase was formed later (de-
mosomal structure during cell cycle progression.3 layed) than the ones not treated with nocodazole
We monitored real-time cells to observe nuclear (Figure 4B, 165 min). The nucleus in metaphase did
changes that could predict cell cycle. As shown in not divide into two and became untangled state
Figure 4, the chromosomes are loosely distributed (Figure 4B, 255 min~270 min), and DNA eventually
throughout the interphase stage (Figure 4A, 0 min), fragmented resulting in apoptosis (Figure 4B, 1170
min). In the process of cell division, chromosomes
bind to the mitotic spindle, a structure composed
of microtubules, and are pulled to both ends to be
divided. In the case of nocodazole, it is known to
disturb microtubule dynamics in cell division.4 In
addition, it is possible to observe the form of chro-
mosomes condensed in metaphase, anaphase, and
telophase during cell division without fluorescence
labeling through high-resolution bright-field im-
aging of the Celloger (Figure 5).

Topic 3. Cytotoxic activity of immune cell:


NK-killing assay
Natural killer (NK) cells play a critical role in prima-
ry defense by recognizing pathogens, virus-infect-
ed cells, or tumor cells as target and killing them.
Because of this role, they are in the limelight in
Figure 3. Increase in spheroid area (average, N=3. Error cancer immunotherapy as a specialized immune
bar=standard error) via time. The lower left image is a effector cell that responds to abnormal cells.5 We
spheroid at 18 hours after nocodazole treatment outlined performed an assay to evaluate the killing ability
with red line. The image on the right is a spheroid at 26 of NK cells through Celloger® Mini Plus imaging
hours, and the area reduced compared to the image at 18 and used NK 92 cell, a lymphoblast derived from
hours is marked in yellow. Images used for analysis were
blood as an effector NK cells. K562 cells, a cell line
captured every 30min by Celloger® Mini Plus (4X optics).
derived from human chronic myelogenous leu-

17
Figure 4. Timelapse images of H2B-GFP transfected Hela cells with or without nocodazole . A. Control cells were not treated with nocodazole.
B. Cells were treated with 62.5 nM nocodazole. In each panel, upper images are brightfield images and lower images are fluorescence images.

kemia, were used as a target cell and visualized


through cytoplasmic staining with CellTracker™
Green CMFDA Dye. When the coculture was car-
ried out by increasing the ratio of effector NK92
while fixing the target K562 concentration, it was
confirmed through real-time imaging that the
higher the NK cell ratio, the more efficient the
target K562 apoptosis was induced, thereby re-
ducing the number of fluorescent cells (Figure 6).
Since the receptor of NK cells recognizes and bind
to the target antigen, the NK cells are observed
Figure 5. Image of Hela cells during mitosis taken by
as aggregated with the target cells. It is presumed
Celloger Nano (4X optics)
that the higher the concentration of NK cells, the

18
larger the aggregated size and the higher the immune cells such as T cells and NK cells, measures
probability of killing the tumor cells.6 the amount of 51Cr released when target cells die.
Chromium release assay, a well-known and com- This is difficult to screen over time because there
monly used method for quantifying cytotoxicity by is not only a risk of radiation exposure, but the su-

Figure6.Extinctioneffectofk562cellsdependingontheconcentrationofNK-92cellsNK92cells(effector)wereincreasedinproportiontoK562
cells (target) stained with CellTracker™ Green CMFDA, and cocultured. Bright field and green fluorescence channel images were captured
with Celloger® Mini Plus (4X optics) every hour for up to 20 hours. A. Bright field image tile merged with green fluorescence channel images.
B. Green fluorescence image tile.

19
pernatant containing 51Cr must also be collected it includes a stitching function that can obtain an
and measured repeatedly.7 On the other hand, the image of a larger area (Figure 7).
real-time imaging using Celloger makes it easy to
observe that fluorescence-stained target cells de- References
crease due to apoptosis, thereby allowing quan-
tification of cytotoxic activity of immune cells. In 1. Li, Grace N., et al. Tissue Engineering, 2007, 13.5:
addition, it can increase measurement accuracy as 1035-1047.

2. Han, Se Jik; KWON, Sangwoo; KIM, Kyung Sook.


Cancer Cell International, 2021, 21.1: 1-19.

3. Kanda, Teru; Sullivan, Kevin F.; WAHL, Geoffrey M.


Current Biology, 1998, 8.7: 377-385.

4. Jordan, Mary Ann; Thrower, Douglas; Wilson, Leslie.


Journal of Cell Science, 1992, 102.3: 401-416.

5. Vivier, Eric, et al. Science, 2011, 331.6013: 44-49.

Figure 7. Screen for stitching with Celloger® Mini Plus anal- 6. Prager, Isabel; Watzl, Carsten. Journal of Leukocyte
ysis app after imaging one well with 3X3 tile in Celloger® Biology, 2019, 105.6: 1319-1329
Mini Plus
7. Karimi, Mobin A., et al. PloS One, 2014, 9.2: e89357.

20
Explore, Learn, Decide
www.biocompare.com

Changing the Way You Research


Find out why more than 3 million scientists around the globe rely on
Biocompare to learn about and find the right products for their research.
Visit us today at biocompare.com

Bench Tips Product Reviews

includes over 7 million products from 400 suppliers


Featured Articles Citations

Review a product to become a Biocompare Product


Investigator and receive an Amazon Gift Card

395 Oyster Point Blvd #300


For More Information Visit us at Biocompare.com South San Francisco, CA 94080
(650) 637–1277
Cryopreserved PBMCs Retain
Phenotype and Function
Viable cryopreserved PBMCs offer a convenient alternative
to fresh PBMCs.

Frozen (cryopreserved) peripheral blood mono- • CD14 (monocyte marker)


nuclear cells (PBMCs) offer many advantages over
• CD56 (NK cell marker)
fresh PBMCs, enabling researchers to plan their
experiments ahead of time with ready-to-use cells • CD4 (T helper subset)
that can be stored in the freezer until needed. One
concern with frozen cells is the potential loss of vi- • CD8 (T cell subset)
ability, phenotype, or function resulting from cryo- • CD19 (B cells)
preservation. We report here the characterization
of frozen PBMCs from BioIVT as well as direct com- Cryopreserved PBMCs from three individual
parison of T cell activation responses through mea- healthy donors were used for all experiments. Rep-
surement of expression levels of well-established resentative data are shown in Figure 1. Viability
T cell activation markers (CD25 and CD69). Data and composition results are summarized in Table 1.
suggest these frozen PBMCs maintained viabili- Average cell viability was determined to be 91.8%
ty and phenotype during cryopreservation. While (Figure 1A). The phenotypic composition of each
stunted, T cells exhibited appropriate activation cell population, expressed as a percentage of the
responses upon anti-CD3 and anti-CD28 antibody gated cell population (see Figure 1B), aligned with
stimulation. Functional, viable cryopreserved PB- expected values. Results suggest these frozen PB-
MCs offer a convenient alternative to fresh PBMCs MCs maintained viability and phenotype during
in cell-based assays and immunological studies. cryopreservation and thawing.

Frozen PMBC characterization PBMCs and T cell activation


Frozen PBMCs were first assessed for viability us- Peripheral blood mononuclear cells consist of lym-
ing propidium iodide staining and characterized phocytes (T cells, B cells, NK cells) and monocytes.
by flow cytometry using fluorescent antibodies These cells give selective responses to the immune
against the following protein markers: system and are key players in immunity. T cells are

22
Cell Monitoring,
EVOLVED.
The Millicell® DCI
Digital Cell Imager Small footprint USB and Wi-Fi
with sleek design image transfer

Assess your cell cultures with ease. Our


Two-click
Millicell® DCI instrument enables more efficient
image capture
execution of the repetitive daily techniques
associated with cell passaging. Quickly and
objectively assess confluency, morphology, and
growth trends for more consistent cell cultures.

Quickly determine
Web-based confluency and
app for estimate total
data analysis, cell count
Taking confluency sorting, and
archiving
to a new level.
To learn more, visit
MillicellDCI.com

© 2021 Merck KGaA, Darmstadt, Germany


and/or its affiliates. All Rights Reserved.
MilliporeSigma, the vibrant M, Millipore,
and Millicell are trademarks of Merck KGaA,
Darmstadt, Germany or its affiliates. All other
trademarks are the property of their respective
owners. Detailed information on trademarks is
available via publicly accessible resources.
35890 06/2021

The life science business


of Merck KGaA, Darmstadt,
Germany operates as
MilliporeSigma in the
U.S. and Canada.
Table 1. Viability and composition of frozen PBMCs. All cell
populations were selected by gating on lymphocytes and monocytes
by size and CD45+ expression as shown in Figure 1B. T cells were
further selected using a combined gate of CD4+ and/or CD8+. Frozen
PBMCs from three individual healthy donors were used. Results
indicate the average value and standard deviation as determined by
flow cytometry.
Averaged result
Viability 91.8% ± 2.2%
Population composition
Monocytes 32.0% ± 8.8%
NK cells 4.4% ± 2.0%
B cells 10.1% ± 1.6%
T cells (CD4 subset) 32.9% ± 4.1%
T cells (CD8 subset) 18.0% ± 4.0%

Figure 2. Activated T cells display increased CD25 expression


Figure 1.A) Frozen PBMC viability as determined by
and transiently increased CD69 expression.
propidium iodide staining and flow cytometry. Calculat-
ed cell viability (averaged from three samples) was 91.8%.
B) Phenotypic analysis of frozen PBMCs. CD45+ cells were
analyzed for CD14, CD19, and CD8/CD4 expression to de-
CD69 cell surface markers can therefore be used as
termine the composition of monocytes, B cells, and T cells measures of T cell activation and function.
respectively. For NK cells, CD56 staining was used. Tabu-
lated results are shown in Table 1. To determine whether these frozen PBMCs re-
tained their function, we used flow cytometry to
measure changes in CD25 and CD69 following
activated through extracellular signals that are me-
stimulation with anti-CD3 and anti-CD28 antibod-
diated mainly by T cell receptor (TCR) complexes.
ies. Experiments were performed using both fresh
It has been shown that certain agonists such as and frozen PBMCs.
antibodies to CD3 and CD28 can trigger TCR sig-
naling observable through changes in cell surface CD25 expression
marker expression similar to actual activation of
TCR via antigenic stimulation. Activation of T cells PBMC samples were processed in three separate
results in increased CD25 expression and transient- experiments. For each experiment, cell surface
ly increased CD69 expression (Figure 2). CD25 and marker expression was compared using two sam-

24
ples of fresh cells from different donors and two tivation with anti-CD3 and anti-CD28 antibodies.
samples of frozen cells from different donors. Cells Fresh cells exhibited a larger initial increase in CD25
were activated using RPMI(+) culture media con- expression compared to frozen cells post-activa-
taining anti-CD3 (3 µg/mL) and anti-CD28 (1 µg/ tion, though a robust response was still observed
mL) antibodies. RPMI(+) media without activation with frozen cells on Days 1 and 3. Longer incuba-
antibodies was used as a baseline control. Cells tions resulted in a larger increase in CD25 expres-
were stained using fluorescently labeled anti-CD25 sion for both fresh and frozen cells.
antibody at the indicated time points post-activa-
tion and analyzed the same day by flow cytometry
(Figure 3). For analysis, cells were gated by size for
CD69 expression
the lymphocyte population using forward scatter
Experiments and gating strategy were run as de-
area and side scatter area. Cells were then select-
scribed for CD25, using fluorescently labeled an-
ed by gating on lymphocytes using CD45+ expres-
sion and gating on T cells using a combined gate ti-CD69 antibody instead of fluorescent anti-CD25
of CD4+ and/or CD8+. The resulting cells were con- antibody. Results are shown in Figure 4. As ex-
sidered T cells of either CD4+ or CD8+ lineage. pected, both fresh and frozen PBMCs showed an
increase in T cell CD69 expression following acti-
As expected, both fresh and frozen PBMCs showed vation with anti-CD3 and anti-CD28 antibodies.
an increase in T cell CD25 expression following ac-

Figure 3. Expression of CD25 at 24 hours (A) or 72 hours


(B) post-activation, with donor matched no-activation Figure 4. Expression of CD69 at 24 hours (A) or 72 hours (B)
control samples for each timepoint. A-B) Representative post-activation, with donor matched no-activation control sam-
histograms, with cells shown gated on CD45+ WBCs and ples for each timepoint. A-B) Representative histograms, with
either CD4+ or CD8+ for T cells. C) Change in percent of cells shown gated on CD45+ WBCs and either CD4+ or CD8+ for
cell population expressing CD25, calculated as the differ- T cells. C) Change in percent of cell population expressing CD69,
ence between donor match activation and no-activation calculated as the difference between donor match activation
samples (fresh PBMCs, n = 2; frozen PBMCs, n = 4). D) Table and no-activation samples (fresh PBMCs, n = 2; frozen PBMCs, n
depicting the values graphed in C. = 4). D) Table depicting the values graphed in C.

25
Fresh cells again exhibited a larger initial increase type, and function after cryopreservation, offering
in CD69 expression compared to frozen cells a convenient, ready-to-use alternative to fresh cells
post-activation, though a detectable response for most applications.
was still observed with frozen PBMCs. For CD69,
a significantly larger response was observed one Materials and methods
day post-activation compared to three days post
activation for both fresh and frozen cells. This Materials
aligns with the expected transient response of the
• Frozen PBMCs from BioIVT
CD69 cell surface marker.
(#HUMANPBMC-0001993)

Frozen PBMC function summary • Fresh PBMCs (AllCells®)

Functional results are summarized in Figure 5. Pat- • Mouse anti-human CD3, monoclonal
terns of changes in CD25 and CD69 marker expres- (#SAB4700041)
sion matched expected responses for both fresh • Mouse anti-human CD28, monoclonal
and frozen PBMC samples. Frozen PBMCs showed (Calbiochem® #217669-100UG)
weaker initial activation compared to fresh PBMCs
as measured by changes in CD25 and CD69 mark- • Anti-CD25-FITC (#SAB47000363)
er expression. Despite this difference, detectable
• Anti-CD69-FITC (BD Biosciences #555530)
responses were observed with frozen cells at both
24 hours and 72 hours post-activation. These data • Anti-CD14-FITC (#FCMAB181F)
suggest these frozen PBMCs retain viability, pheno-
• Anti-CD56-APC (#SAB4700450)

• Anti-CD4-APC (#MABF416)

• Anti-CD8-PE (#MABF1680)

• Anti-CD19-FITC (#FCMAB184F)

• Propidium iodide (#537060)

• RPMI(+) medium: RPMI containing 10% FBS,


1% Pen/Strep, 1% L-Glutamine

• Phosphate buffered saline (PBS)

Figure 5. Summary of data comparing activation of fresh • FACS buffer: 95% PBS and 5% RPMI(+)
and frozen PBMCs as measured by changes in CD25 and
CD69 marker expression. Though a weaker response was
Cell thawing and treatment
observed with frozen cells, the data suggest these cryopre-
1. Thaw frozen vials of PBMCs in the 37 °C
served PBMCs retain their function.
water bath for 2-5 minutes

26
2. Add RPMI(+) medium to both frozen and 4. Spin down the microcentrifuge tubes in a
fresh PBMCs centrifuge at 300 x g for 5 minutes. Make
sure cells formulate a pellet. If no visible
3. Pellet cells at 300 x g for 5 minutes pellet, repeat spin of cells.
4. Remove supernatant 5. Remove supernatant to 50 µL line on
microcentrifuge tube. Resuspend cells with
5. Resuspend cells in RPMI(+) medium
light pipetting.
6. Count cells on hemocytometer and plate
6. Add in the fluorophore-labeled antibody
100,000 live cells in a 200 µL volume
against the desired cell surface marker and
per well into a 96-well plate. Note that incubate in the dark for 40 minutes at room
samples will be resuspended in RPMI(+) temperature.
media containing anti-CD3 (3 µg/mL final
concentration) and anti-CD28 (1 µg/mL 7. Repeat steps 3-5.
final concentration) for stimulation and 8. Resuspend cells in 250 µL of FACS buffer
activation response, or only RPMI(+) media and mix by gently pipetting. Stain cells for
alone for non-activated samples. viability if desired.
Cell staining and gating on 9. Transfer the full volume (~300 µL) from
flow cytometer each microcentrifuge tube into a 96-well
U bottom plate for analysis using a Guava®
1. For all wash steps, use FACS buffer.
easyCyte™ 8HT Flow Cytometer.
2. Begin with samples in a 96-well plate View our full listing of normal PBMCs, disease state
as described in the “Cell thawing and PBMCs, and immune cell subsets from BioIVT. For
treatment” section. Transfer the whole more information about our PMBCs and Immune
volume of each well (~200 µL) into individual Cell Subsets, please visit www.sigmaaldrich.com/
microcentrifuge tubes. bioivt.
3. Add 1 mL FACS buffer to each MilliporeSigma is the U.S. and Canada Life Science
microcentrifuge tube. business of Merck KGaA, Darmstadt, Germany.

27

You might also like