Springer 2024single Cell Analysis
Springer 2024single Cell Analysis
Single Cell
Analysis
Methods and Protocols
METHODS IN MOLECULAR BIOLOGY
Series Editor
John M. Walker
School of Life and Medical Sciences
University of Hertfordshire
Hatfield, Hertfordshire, UK
Edited by
Miodrag Gužvić
Department of Urology, University Hospital Regensburg, Regensburg, Germany
Editor
Miodrag Gužvić
Department of Urology
University Hospital Regensburg
Regensburg, Germany
This Humana imprint is published by the registered company Springer Science+Business Media, LLC, part of Springer
Nature.
The registered company address is: 1 New York Plaza, New York, NY 10004, U.S.A.
An exciting time lies ahead for cellular biology. We are finally able to move away from
assessing genome, transcriptome, proteome, or metabolome as a mean of a population and
instead focus on the features of single cells. Developments in physics, chemistry, and
molecular biology have pushed the boundaries of what is possible in cellular analysis to
give unprecedented improvements in sensitivity and scale that are allowing us the capability
to approach a full functional appraisal of single cells at a rapid rate.
The speed, sensitivity, accuracy, and scope of existing techniques for single-cell detec-
tion, isolation, and analysis (e.g., FACS, fluorescence microscopy, dielectrophoresis, whole
genome and transcriptome amplification, high-throughput sequencing, etc.) have improved
markedly, broadening research horizons, while complementary techniques in organic spec-
troscopy (e.g., MALDI imaging), inorganic spectroscopy (e.g., ICP-MS), and synchrotron
analysis support this with more detailed information on aspects previously overlooked.
This volume summarizes these techniques, their capabilities, and the type of information
that can be determined, and aims to give an overview of best practice for implementing them
in single-cell analysis in an important and necessary move away from the bulk analysis that is
constraining our boundaries.
Multi-omics analysis of single cells is gaining momentum in recent years. Such
approaches are somewhat underrepresented in this book, and future editions should more
comprehensively cover this important aspect of single-cell analysis. Furthermore, while an
attempt was made to make this volume thematically comprehensive, it is somewhat biased
toward the analysis of single cancer cells. The future editions of this book should broaden its
scope by including protocols on isolation and analysis of single prokaryotic or plant cells.
Still, many thematically overlapping or linked protocols presented in this volume enable
development of complex and complete workflows to isolate and analyze single cells, even
beyond cancer research.
v
Acknowledgments
I am grateful to Rob Hutchinson for support in early phases of preparation of this book, and
to contributors and the publisher for their limitless patience, which I tested many times
while wrestling many obstacles trying to bring this book to see the light of the day.
This book is dedicated to Christoph A. Klein, who introduced me to the multiverse of
single-cell analysis.
vii
Contents
Preface . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . v
Acknowledgments. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . vii
Contributors. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . xi
1 Single Cell Isolation from Surgically Resected Tissue Via Mechanical
Dissociation Using TissueGrinder. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1
Prama Pallavi and Stefan Scheuermann
2 Circulating Tumor Cell Enrichment and Single-Cell Isolation
Combining the CellSearch® and DEPArray™ Systems. . . . . . . . . . . . . . . . . . . . . . . 11
C€a cilia Köstler, Bernhard Polzer, and Barbara Alberter
3 Isolation of Viable Epithelial and Mesenchymal Circulating Tumor Cells
from Breast Cancer Patients . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 43
Justyna Topa, Anna J. Żaczek, and Aleksandra Markiewicz
4 Single-Cell Recovery from Tumor Cell Xenotransplanted Zebrafish
Embryos for the Study of Metastasis-Initiating Cells . . . . . . . . . . . . . . . . . . . . . . . . 53
Pablo Hurtado, Inés Martı́nez-Pena, and Roberto Piñeiro
5 Isolation of Single Circulating Tumor Cells Using VyCAP Puncher System . . . . 65
Thais Pereira-Veiga, Bianca Behrens, Joska J. Broekmaat, Lisa Oomens,
Michiel Stevens, Arjan G. J. Tibbe, Nikolas Stoecklein,
Laura Muinelo-Romay, Roberto Piñeiro, and Clotilde Costa
6 Simultaneous Isolation and Amplification of mRNA and Genomic
DNA of a Single Cell . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 71
Miodrag Gužvić
7 Isolation and Genomic Analysis of Circulating Tumor Cell Clusters
in Cancer Patients. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 101
Carolina Reduzzi, Marta Vismara, Thomas Schamberger, Marco Silvestri,
Rosita Motta, Bernhard M. Polzer, and Vera Cappelletti
8 Establishing Single-Cell Clones from In Vitro-Cultured Circulating
Tumor Cells . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 119
Teng Teng and Min Yu
9 Immunofluorescence Combined with Single-Molecule RNA
Fluorescence In Situ Hybridization for Concurrent Detection
of Proteins and Transcripts in Stress Granules . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 127
Jakub Kochan and Mateusz Wawro
10 Highly Multiplexed and Simultaneous Characterization of Protein
and RNA in Single Cells by Flow or Mass Cytometry Platforms
Using Proximity Ligation Assay for RNA . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 143
Andrew D. Duckworth, Joseph R. Slupsky, and Nagesh Kalakonda
11 Array-Based Comparative Genomic Hybridization for the Detection
of Copy Number Alterations in Single Cells . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 167
Giancarlo Feliciello, Zbigniew Tadeusz Czyz, and Bernhard M. Polzer
ix
x Contents
Index . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 259
Contributors
xi
xii Contributors
Abstract
Primary cells form the basis of modern-day in vitro research analysis tools. Many conventional procedures
for generating single-cell suspensions from solid tissue are neither robust nor reproducible. Here we
describe primary cells isolation from surgically resected tumor tissue via enzyme-free mechanical dissocia-
tion using TissueGrinder, a novel semi-automated benchtop device. The isolated cells can be used for any
downstream biochemical or cell-based analytic assay.
Key words Primary cells, Enzyme-free cell isolation, Single-cell isolation, Mechanical dissociation
1 Introduction
Primary cells isolated from tissue are very important in vitro tools to
answer several complex biological questions. In particular primary
cells from surgically resected tissue can provide insight into cellular
biological activity and hence new insights into disease [1]. Methods
to isolate primary cells from tissues can be broadly divided into two
main categories—explant and tissue dissociation techniques using
mechanical force and enzymatic digestion [2–5]. For more than
50 years, the explant method dominated the field of tissue culture
for obtaining primary cells [6, 7]. In fact, this is one of the simplest
techniques: the tissue is finely minced (1–2 mm3) and placed in a
culture flask [8], then the explant is covered with an appropriate cell
outgrowth medium. Within a week’s time, cells migrate out of the
explants and grow on the culture surface [9]. However, success is
limited by various factors such as long processing times, low yield,
and extensive manual workload, leading to a substantial increase in
the overall time from tissue resection to the generation of a cell line.
Furthermore, not all types of cells are able to migrate out of explant
tissue. The second method, dissociation with enzymes, is not
always desired because enzymatic digestion could affect the
Miodrag Gužvić (ed.), Single Cell Analysis: Methods and Protocols, Methods in Molecular Biology, vol. 2752,
https://doi.org/10.1007/978-1-0716-3621-3_1, © Springer Science+Business Media, LLC, part of Springer Nature 2024
1
2 Prama Pallavi and Stefan Scheuermann
2 Materials
2.1 Tissue Transport 1. Prepare centrifuge tube with 30 mL of DMEM medium sup-
plemented with 100 units/mL of penicillin and 100 μg/mL of
streptomycin.
2. Prepare a transport container with ice, to transport the tube
with medium. Tissue is collected as quickly as possible with a
cool box from surgery.
Single-Cell Isolation Using TissueGrinder 3
3 Methods
3.2 Pre-Cutting of Note: All these steps should be performed under sterile tissue
the Tissue Sample culture bench (see Notes 3 and 4).
1. Wash the tumor tissue with PBS supplemented with 1% Peni-
cillin/Streptomycin solution (Penicillin (10,000 units/mL)
and Streptomycin (10,000 μg/mL) solution)).
2. If the sample is still blood-soaked, then wash with an ammo-
nium chloride solution to lyse erythrocytes.
3. Pre-cut the tissue into small cubes with an edge length of about
1–2 mm with help of a single-use scalpel.
4. Weigh up to 10–400 mg of the pre-cut tissue pieces in a clean
and sterile petri dish.
3.3 Processing with 1. Take the disposable sterile grinding tube sets for the Tissue-
the TissueGrinder Grinder. Assemble by latching the stator into the cell sieve.
Next, wash stator and cell sieve with 1 mL of Dresden Medium
and transfer the cell sieve to the sterile 50 mL centrifuge tube
(see Note 5).
2. Place 10–400 mg of the prepared tissue pieces in the spare
space between the grinding teeth of the rotor (using a 1 mL
pipette) using a pair of sterile tweezers. Add 500 μl of the
Dresden medium and place the stator with cell sieve on the
rotor and close the tube. The detailed assembly of the grinding
unit is shown in Fig. 1.
3. Place the closed tube on a TissueGrinder benchtop port and
select a predefined tissue dissociation program. There are three
different generic programs—soft, medium, and hard—avail-
able for soft (easy to cut), medium (mildly fibroblastic), and
hard tissues (highly fibroblastic and somewhat calcified). In
addition, pre-installed programs for specific tissue types (such
as liver, spleen, heart muscle, etc.) can be selected or custo-
mized for specific user applications. The generic programs
should be selected and executed based on the texture of the
Single-Cell Isolation Using TissueGrinder 5
Fig. 1 Assembly of single-use TissueGrinder sterile grinding set (1) and the tissue dissociation workflow
(2–12). The grinding set consists of a fitting lid to the 50 mL centrifuge tube with a centered hole, the rotor and
the stator both forming the grinding gear, a standard cell strainer with a pore size of 100 μm, and a 50 mL
centrifuge tube the assembled grinding set is locked with the rotor’s specific notch to its counterpart on the
TissueGrinder. The diced tumor tissues, each with edge lengths of 1–2 mm and a total weight of 10–400 mg,
are placed in the inner part of the rotor and the unit is assembled as shown under a cell culture bench. Diced
tissue fragments are processed into single cells by alternating processes of grinding and cutting which is
achieved in the TissueGrinder by clockwise or anticlockwise turning of the rotor. After the dissociation
protocol, the tube is transferred directly to a standard laboratory centrifuge and the generated single-cell
suspension is centrifuged through the 100 μm cell filter. The cell pellet is resuspended and transferred to a
new vessel
Table 1
Steps of three TissueGrinder programs used for processing of tumor
tissues
Table 2
Representative cell counts after processing tissues from anal cancer sample
3.4 Cell Counting 1. Take 10 μl of the cell suspension and mix with 10 μl of the
and Viability (see Trypan blue solution.
Notes 9–12) 2. Load 10 μl of this cell solution on to Luna counting slide and
insert it in the Luna instrument to count the cells.
3. Note down the cell number (Table 2) and viability of the
isolated cells.
3.5 Cell Culture 1. Change the medium of the cells in the six well-plate twice a
week until a confluent cell monolayer is observed.
2. Once a monolayer is observed (Fig. 2), sub-culture the cells
into T25- and T75-flasks with passage ratio of 1:3 (see Notes
13 and 14).
Single-Cell Isolation Using TissueGrinder 7
4 Notes
Acknowledgement
References
1. Lee J-K, Liu Z, Sa JK, Shin S, Wang J, 8. Ruckert F, Aust D, Bohme I, Werner K,
Bordyuh M, Cho HJ, Elliott O, Chu T, Choi Brandt A, Diamandis EP, Krautz C, Hering S,
SW, Rosenbloom DIS, Lee I-H, Shin YJ, Kang Saeger HD, Grutzmann R, Pilarsky C (2012)
HJ, Kim D, Kim SY, Sim M-H, Kim J, Lee T, Five primary human pancreatic adenocarci-
Seo YJ, Shin H, Lee M, Kim SH, Kwon Y-J, Oh noma cell lines established by the outgrowth
J-W, Song M, Kim M, Kong D-S, Choi JW, method. J Surg Res 172(1):29–39. https://
Seol HJ, Lee J-I, Kim ST, Park JO, Kim K-M, doi.org/10.1016/j.jss.2011.04.021
Song S-Y, Lee J-W, Kim H-C, Lee JE, Choi 9. Rückert F, Aust D, Böhme I, Werner K,
MG, Seo SW, Shim YM, Zo JI, Jeong BC, Brandt A, Diamandis EP, Krautz C, Hering S,
Yoon Y, Ryu GH, Kim NKD, Bae JS, Park Saeger HD, Grützmann R, Pilarsky C (2012)
W-Y, Lee J, Verhaak RGW, Iavarone A, Lee J, Five primary human pancreatic adenocarci-
Rabadan R, Nam D-H (2018) Pharmacoge- noma cell lines established by the outgrowth
nomic landscape of patient-derived tumor method. J Surg Res 172(1):29–39. https://
cells informs precision oncology therapy. Nat doi.org/10.1016/j.jss.2011.04.021
Genet 50(10):1399–1411. https://doi.org/ 10. Scheuermann S, Sch€afer A, Langejürgen J, Reis
10.1038/s41588-018-0209-6 C (2019) A step towards enzyme-free tissue
2. Mitra A, Mishra L, Li S (2013) Technologies dissociation. Curr Dir Biomed Eng 5(1):
for deriving primary tumor cells for use in per- 545–548. https://doi.org/10.1515/cdbme-
sonalized cancer therapy. Trends Biotechnol 2019-0137
31(6):347–354. https://doi.org/10.1016/j. 11. Hu P, Zhang W, Xin H, Deng G (2016) Single
tibtech.2013.03.006 cell isolation and analysis. Front Cell Dev Biol
3. Bols NC, Lee LEJ (1991) Technology and uses 4:116. https://doi.org/10.3389/fcell.2016.
of cell cultures from the tissues and organs of 00116
bony fish. Cytotechnology 6(3):163–187. 12. Reichard A, Asosingh K (2019) Best practices
https://doi.org/10.1007/BF00624756 for preparing a single cell suspension from solid
4. Gawad C, Koh W, Quake SR (2016) Single-cell tissues for flow cytometry. Cytometry A 95(2):
genome sequencing: current state of the sci- 219–226. https://doi.org/10.1002/cyto.a.
ence. Nat Rev Genet 17(3):175–188. https:// 23690
doi.org/10.1038/nrg.2015.16 13. Tennant JR (1964) Evaluation of the trypan
5. Esparza-López J, Martı́nez-Aguilar JF, Ibarra- blue technique for determination of cell viabil-
Sánchez MJ (2019) Deriving primary cancer ity. Transplantation 2:685–694. https://doi.
cell cultures for personalized therapy. Rev org/10.1097/00007890-196411000-00001
Investig Clin 71(6):369–380. https://doi. 14. Scheuermann S, Lehmann JM, Mohan RR,
org/10.24875/ric.19002832 Reißfelder C, Rückert F, Langejürgen J, Pallavi
6. Fischer A (1925) Tissue culture; studies in P (2021) TissueGrinder, a novel technology for
experimental morphology and general physiol- rapid generation of patient-derived single cell
ogy of tissue cells in vitro suspensions from solid tumors by mechanical
7. Parker RC, Morgan JF (1950) Methods of tissue dissociation. Front Med 9:721639.
tissue culture. Hoeber Medical Division- https://doi.org/10.3389/fmed2022.721639
Harper & Row, New York
Chapter 2
Abstract
The analysis of circulating tumor cells (CTCs) has shown potential for detection of cancer spread,
prognosis, therapeutic target selection, and monitoring of treatment response. CTCs can be obtained
repeatedly by simple blood draws as so-called “liquid biopsy.” Thus, they can serve as a surrogate material
for primary or metastatic tissue biopsies. In addition, isolation of CTCs provides the possibility to investi-
gate those cells which may hold the (molecular) traits responsible for metastatic progression and ultimately
patient death. As such, CTCs represent a target of utmost importance in cancer research and therapy. In this
chapter, we describe a workflow for the enrichment of CTCs with the FDA-cleared CellSearch® system
followed by the isolation of single CTCs using the DEPArray™ technology enabling further molecular
single-cell analyses.
Key words CTC, CellSearch, DEPArray, Single-cell isolation, Dielectrophoresis, Liquid biopsy
1 Introduction
Miodrag Gužvić (ed.), Single Cell Analysis: Methods and Protocols, Methods in Molecular Biology, vol. 2752,
https://doi.org/10.1007/978-1-0716-3621-3_2, © Springer Science+Business Media, LLC, part of Springer Nature 2024
11
12 €cilia Köstler et al.
Ca
2 Materials
Fig. 1 CellSearch® System consisting of CellSearch® Autoprep System and CellTracks® AnalyzerII. (© by
Menarini Silicon Biosystems SpA)
2.1.3 CellSearch® The CellSearch® Circulating Tumor Cell Kit (see Fig. 2) is used for
Circulating Tumor Cell Kit capturing CTCs of epithelial origin (CD45-, EpCAM+, and cyto-
keratins 8+, 18+, and/or 19+). It contains buffers and reagents for
16 enrichments. Up to its use it has to be stored at 2–8 °C (see
Notes 1 and 2). Once it has been opened, the kit can be used for up
to 30 days for diagnostic purposes (see Note 3).
The kit contains the following reagents:
1. 3.0 mL Anti-EpCAM Ferrofluid (brown cap):
Suspension of 0.022% magnetic particles conjugated to a
mouse monoclonal antibody specific for the cell surface marker
EpCAM present on epithelial cells in a buffer with 0.03%
bovine serum albumin (BSA) and 0.05% ProClin®
300 preservative.
2. 3.0 mL Staining Reagent (white cap):
0.0006% mouse monoclonal antibodies specific to cytoker-
atins conjugated to phycoerythrin (PE) and 0.0012% mouse
14 €cilia Köstler et al.
Ca
Fig. 2 Content of CellSearch® Circulating Tumor Cell (CTC) Kit. (© by Menarini Silicon Biosystems SpA)
2.1.4 CellSearch® The CellSearch® Circulating Tumor Cell Control Kit is used for
Circulating Tumor Cell testing system performance and operator technique. It is sufficient
Control Kit for 24 applications and proves that the system detects both high as
well as low numbers of CTCs. It has to be stored at 2–8 °C and
contains:
CTC Isolation Using DEPArray 15
2.3 (Single)-Cell 1. DEPArray™ Plus for CTC Application Guide and DEPArray™
Isolation with the Instruction.
DEPArray™ Platform 2. DEPArray™ system (see Fig. 3).
16 €cilia Köstler et al.
Ca
3. DEPArray™ cartridges.
4. Buffer for the DEPArray™ system (DABUF).
5. 1.5 mL Protein LoBind tubes.
6. MicroAmp Reaction Tubes with cap 0.2 mL.
7. Micropipettes 5000 μL, 20 μL and related LoRetention filter
tips (see Note 8).
8. Ultrasonic bath with a pulse button.
9. Racks for 1.5 mL and 0.2 mL tubes.
10. Laminar flow hood.
11. Waterproof pen for the unique labeling of the tubes.
3 Methods
3.1 CTC Enrichment Before starting a run, the system must be cleaned (see Note 10).
with the CELLTRACKS® Eight samples can be processed in parallel. Make sure that the
AUTOPREP® System samples fulfill all requirements for the run (see Note 11). The
necessity for a control sample arises whenever patient samples are
processed or when using a new lot number of kit reagents (see Note
12). Make sure to wear the required protective clothes and gloves
for all following steps.
1. Start the CellTracks® Autoprep® System.
2. Bring the CellSearch® Circulating Tumor Cell Kit and one
bottle of the CellSearch® Circulating Tumor Cell Control Kit
to room temperature (see Note 1).
3. Select “Run batch” and enter the password (see Note 13).
4. Make sure, that the system is clean. If necessary, perform the
“Daily Cleaning Procedure” (see Note 10), otherwise go on
with step 4.
5. Take as many CellSearch®Conical Centrifuge Tubes and Caps as
there are samples (including positive control if necessary). Put a
barcode label (if available, see Note 14) on the CellSearch®Co-
nical Centrifuge Tubes for the samples (see Note 15).
6. Mix the CellSave tube by inverting it five times (see Note 16).
7. Remove the cap of the CellSave tube and transfer 7.5 mL of the
blood sample into a CellSearch® Conical Centrifuge Tube. Add
6.5 mL dilution buffer, close it with a cap and mix gently by
inverting five times (see Note 16).
8. Repeat steps 6 and 7 for further samples.
9. Centrifuge the tubes with a horizontal swing-out style rotor at
800 g for 10 min at room temperature. Attention: Set break to
“0” (see Note 17).
10. Prepare the positive control—if necessary—during the centri-
fugation step, otherwise switch to step 13.
11. Put an orange ID label onto the conical tube with the positive
control (see Note 5).
12. Mix the 3 mL control tube gently by vortexing 5 s and invert-
ing the bottle five times (see Note 18). Open the cap and
transfer the complete volume—also from the cap—to a conical
centrifuge tube (see Notes 19 and 20).
13. Open all bottles of the CellSearch® Circulating Tumor Cell Kit
and inspect them (see Note 21).
14. Follow instructions on the screen for choosing the kit, select-
ing a marker and control.
15. After choosing the number of samples, including the control, the
system is “homing,” which means being prepared for the run.
18 €cilia Köstler et al.
Ca
16. Place the rack with the CellSearch® Circulating Tumor Cell Kit
into the CellTracks®AutoPrep® System (see Note 22) and
confirm the kit data with “Next.”
17. Confirm, if results shall be used for patient management (see
Note 23) or not. (If the kit is not valid anymore for in vitro
diagnostics, see Note 3).
18. Put an empty CellSearch® cartridge (without lid) into the
magnetic rack (MAGNEST®) and load it to the system (see
Notes 24 and 25).
19. Repeat step 18 for all remaining MAGNEST®s.
20. Go on with loading the samples and/or control (see Note 26).
If a control is included in the run, follow the loading order in
analogy to the order of the MAGNEST®s which were loaded in
step 18 (see Note 24).
21. Close the door and start the system with “Start.” During the
run, the estimated duration is indicated (see Note 27).
22. When the enrichment is finished, remove the MAGNEST®s
from the instrument. Carefully inspect the content of the
CellSearch® cartridge (see Note 28).
23. Store the MAGNEST®s with the filled and closed CellSearch®
cartridge at room temperature at a dark place in a horizontal
position and analyze them with the CELLTRACKS ANA-
LYZER II® system within the next 24 h (see Note 29). This
is described in Subheading 3.2.
24. Follow the instructions on the screen for removing the empty
sample/control tubes and inspect them (see Note 30). Remove
the kit and close the reagents (see Note 31).
25. Go on with the daily cleaning procedure if no further runs are
planned.
3.2 CTC Counting Keep in mind that enriched samples (also controls) can be scanned
with the CELLTRACKS at the earliest after 20 min of incubation (in a horizontal
ANALYZER II® System position in the dark at room temperature).
1. Switch on the device, computer, and the mercury vapor
lamp (see Note 32). The CELLTRACKS® software starts
automatically.
2. Sign in with the unique password.
3. If it is necessary to perform a system verification (see Note
33), click the “QC test” folder symbol, clean the system
verification cartridge with a fiber-free wipe, open the sam-
ple door, and load it to the sample rack. Push the “Start”
button in the field “System verification” (see Note 34).
After successful verification reload the verification
cartridge.
CTC Isolation Using DEPArray 19
3.3 Sample After enrichment of CTCs with the CellSearch® system, the sample
Preparation for Single- needs to be subjected to a buffer change before applying it to
Cell Isolation with the the DEPArray™ cartridge (see Figs. 4 and 6) for automatic
DEPArray™ System isolation of single CTCs or blood leukocytes or pools thereof.
All following working steps should be performed under a lami-
nar flow hood wearing the required protective clothes and
gloves.
1. Thaw a vial of DABUF.
2. Prepare a 2% BSA in PBS solution (see Note 49).
3. Prepare.
– two aliquots of 325 μL DABUF.
– one labeled 1.5 mL Protein LoBind tube for the
sample.
– 1.5 mL 2% BSA/PBS solution (see step 2).
– one clean 1.5 mL tube for storing the tip.
CTC Isolation Using DEPArray 21
Fig. 4 Backside of the DEPArray™ cartridge with buffer and sample inlet. Picture
adapted from the DepArray™ User Manual. (© by Menarini Silicon Biosystems
SpA)
3.4 Cell Isolation All steps concerning the loading of the DEPArray™ cartridge have
with the DEPArray™ to be performed under a laminar flow hood wearing the required
System protective clothes and gloves.
1. Take the DABUF bottle and incubate them for 10 min in the
ultrasonic bath with the degas function (see Note 56).
2. Start the DEPArray™ system. Select the username in the drop-
down menu to sign in. Enter the password by touching the
“Code” button (see Note 57) and login.
3. Enable the VPN connection in order to have full service for
troubleshooting.
4. Touch the “Sorting,” “CTC,” “UDP fixed CTC” buttons on
the touch screen (see Note 58).
5. Go back to the laminar flow hood with this DEPArray™ car-
tridge. Open cover “1” (see Notes 59 and 60), remove the blue
protective tape, and leave the DEPArray™ cartridge inside the
laminar flow hood for loading.
6. Pipette 2.5 mL of the degassed and solved buffer (step 1) to
the buffer reservoir “B” (see Fig. 4) of the DEPArray™ car-
tridge by using the 5000 μL micropipette (see Note 61).
CTC Isolation Using DEPArray 23
Fig. 5 Populations and Groups panels of the CellBrowser™ with functional buttons. Picture adapted from the
DepArray™ User Manual. (© by Menarini Silicon Biosystems SpA)
20. When the scan is finished, the cell selection workflow can be
started using the “Cell Selection Software” (see Note 71).
21. An automatic selection of cells (see Note 72) is shown in the
“Populations” panel (see Fig. 5).
22. For an overview of the cell list and the plot view in parallel, click
the “Grid and Data Visualization” icon.
23. Select the “Routable Cells” (see Note 73) by clicking on it
(they will be highlighted in blue) and click on the “Scatter
Plot” icon in the “Data Visualization” panel.
24. Select “Mean Intensity PE” for X-axis and “Mean intensity
APC” for Y-axis. Click the “Cross Selection” button to split
the “Routable Cells” selected for PE and APC into the four
subpopulations and move the lines for optimization (see Note
74).
25. Select the PE+/APC- subpopulation (“Q3”) and click the
“Create Table” icon (see Fig. 5) above the cell populations in
the “Populations” panel. A new table, called “[0]Table0 (0),”
is created (see Note 75) and will be filled in with the cells of
interest to be isolated.
26. Sort the cells in the “Grid View” list from highest to lowest
mean intensity of PE by clicking twice in the blue “Signal
Intensity PE” field (see Note 76) on the top of the list.
27. By clicking to the first cell in the “Grid View” list (highlighted
in blue), the fluorescent images of the selected cell are shown
below. Judge the cell by the official cell selection parameters (see
CTC Isolation Using DEPArray 25
Fig. 6 Front side of the DEPArray™ cartridge with the three-chamber system as the heart of the cartridge.
(© by Menarini Silicon Biosystems SpA)
CTC Isolation Using DEPArray 27
3.5 Preparation of Single cells or cell pools are recovered from the DEPArray™ system
Isolated Cells for in a cell number-specific volume (see Note 96). In order to proceed
Molecular Analyses with whole genome amplification (WGA) followed by molecular
analyses, the volume of the recovered single cells or cell pools needs
to be reduced to approx. 1 μL. Volume reduction of the isolated
samples has to be performed under a laminar flow hood wearing the
required protective clothes and gloves.
1. Spin down the 200 μL tubes at room temperature and
14,100 g at room temperature in a fixed rotor centrifuge for
200 μL tubes. The time depends on the number of cells in the
tubes (see Note 97).
2. Proceed with the following steps under a laminar flow hood.
3. If 21–85 cells are recovered in one tube (compare to Note 96),
carefully remove 50 μL of supernatant. Do not touch the
bottom of the tube. Instead, follow the meniscus of the
solution.
4. Add 100 μL of 1× PBS to all tubes (regardless of prior 50 μL
removal of the supernatant) without touching the solution.
28 €cilia Köstler et al.
Ca
4 Notes
19. Tip over the empty and open control bottle and collect remain-
ing drops in the CellSearch®Conical Centrifuge tube for the
control.
20. There is no need for adding dilution buffer and centrifuging
the control tube.
21. Each bottle of the tray should be fixed properly by pushing
them down. To avoid evaporation, remove the caps directly
before the run. An inspection of the solutions in the bottles for
air bubbles is necessary before each run. A clean micropipette
tip can be used to remove them.
22. This is the last possibility to ensure that the reagents are at
room temperature. The system reports if the number of sam-
ples to be processed does not match the number of remaining
samples which can be processed with this kit.
23. Samples which are used for the patient management are always
written in black, on each and every
CELLTRACKS®AUTOPREP® System.
24. Every stained and enriched sample is collected in a single-use
CellSearch® cartridge, which is entered in a magnetic rack, the
so called MAGNEST®. There is no rule specifying the order of
loading samples and controls. However, in order to prevent
confusion, the gray MAGNEST® should be used for the con-
trol and the purple MAGNEST®s for the samples. Make sure
to load control/samples and gray/purple MAGNEST®s
accordingly using the same order. Every MAGNEST® has a
memory chip saving all the information created during the run.
It is deleted before every new run automatically, so the
MAGNEST® can be used continuously.
25. Each CellSearch® cartridge should be empty and clean inside.
The outside can be cleaned with a fiber-free tissue. The appro-
priate number of CellSearch® cartridges (corresponding to the
number of controls/samples) are inserted into the
MAGNEST®s and fixed properly by pushing them down.
MAGNEST®s must be undamaged since this could influence
the recovery of cells and the associated report. After entering a
MAGNEST®, the system shows the barcode of the CellSearch®
cartridge on the screen, closes the door, and moves the
MAGNEST® holder carousel to the next free position. All
filled MAGNEST®s have to be inserted.
26. The samples/controls have to be loaded in a way that the
barcode is directed toward the user and the sample tube is
freely rotatable. If there is no barcode label on the sample
tubes, the sample can be loaded anyway. By pushing “Enter,”
a warning message appears (see also Note 15) and the sample
ID and type can be edited. This information has to be saved.
32 €cilia Köstler et al.
Ca
27. The run takes 2 h 20 min plus 13 min per additional sample.
28. The CellSearch® cartridge must not be removed from the
MAGNEST® for inspection! It should be filled with an
orange-yellow colored solution (if not see Note 30). Be aware
that in rare cases, ferrofluid reagent can aggregate because of
disturbing factors in the blood of some patients. As a conse-
quence, the ferrofluids might mask CTCs during the scan
which could influence the results. Aggregated ferrofluids
close to the plug might have less influence to the results than
aggregates in the middle of the sample. If there are bubbles
inside the CellSearch® cartridge, place the plug onto the Cell-
Search® cartridge opening without pushing it down. While
keeping the plug in its place, knock the MAGNEST® gently
against a hard surface (e.g., bench) to destroy the bubbles.
Then re-seal the CellSearch® cartridge with the plug. If the
CellSearch® cartridge is empty, the associated conical tube
might still be filled with liquid. If this is the case, please seek
advice from the technical support.
29. The closed MAGNEST®s holding the CellSearch® cartridge
should be stored for a minimum of 20 min before scanning in a
horizontal position. This ensures that ferrofluid connected cells
sediment to the same layer for scanning. IVD samples and
controls have to be scanned with the CELLTRACKS ANA-
LYZER II® after 24 h at the latest and are allowed to be
scanned only twice. Note that if samples are scanned more
than twice within 24 h, they lose their IVD status.
30. Every CellSearch®Conical Centrifuge tube has to be inspected
before throwing it away. Ferrofluid accumulations might have
remained inside the tube. The results of such samples should be
considered with care since they might possibly be invalid.
31. Before removing the kit, it is possible to make a print-out with
the information of how many runs can still be performed with
the opened kit. For this, go back to the main menu (“View
Data” ! “Reagent Kit”) and print. For removing the kits,
close the bottles with their uniquely colored caps and store
the kit until next use at 2–8 °C. The dilution buffer has to be
stored at room temperature.
32. The mercury vapor lamp needs to warm up. This takes
approx.15 min. The lamp is switched on either after confirming
an automatically appearing request for warming up the lamp or
by starting it manually by clicking the button “lamp” in the
toolbar. The lamp signal blinks during warm-up phase and
remains green when the lamp is ready. The lamp has a total
run-time of 300 h. In order to save lifetime of the lamp, it is
advisable to switch it off when there are no samples to be
scanned and to avoid repeated switching on and off.
CTC Isolation Using DEPArray 33
many tables have been created (see also Note 79 and the
“CTC-RUO Fixed” application in the user manual).
79. At maximum 10 routing groups can be created. The total
number of cells to be isolated is limited depending on the
number of groups and the total number of cells in the groups
(e.g., with one routing group a maximum of 105 cells can be
selected; with two routing groups 51 cells per group can be
selected, etc.; for more details see user manual). The name of
the groups can be changed by right-click if necessary.
80. The “Cell Panel” consists of the “Sidebar” (selection of groups
and parameters), the “Action Bar” (information about cell
numbers, icons to switch to “Cell Browser” or “Cell Rout-
ing”), and the “Panel Grid” (information and pictures of
selected cells). It is saved automatically and opens by clicking
“Cell Panel” on the top right of the screen in the CellBrow-
ser™ software. Refer to user manual for adaption of the “Cell
Panel.”
81. “Park Routing” (i.e., moving the cells from their place to the
parking chamber of the DEPArray™ cartridge) can be started
by using the “Recovery Manager™” interface. This software
allows to watch the cells moving (due to the incorporated live
camera), to control the exit and recovery of the samples and to
assign samples to the “Recovery Support” layout.
82. It is not necessary but recommended to check if air bubbles are
at the entrance of the parking/exit chamber, which might
block the routing ways. If air bubbles are visible, it is important
to contact the Customer Support before starting the park
routing, otherwise important cells might be lost.
83. It is possible to route all cells within a group (cell pools) or to
select single cells. The selected groups/cells for parking are
labeled with a blue dot.
84. The duration of park routing depends on the number of
selected cells. Both progress and time left are displayed in the
“Park” window.
85. Even if for the “CTC-RUO Fixed” application, the “200 μL-
tube-rack” is preset and recommended for maximum purity of
the samples, another “Recovery Support” (e.g., if more than
33 tubes are needed for one experiment) or other types can be
chosen by clicking the “+” field in the “Exit and Recovery”
section. Choose the appropriate “Recovery Support” from the
drop-down menu and “Add” it. By clicking “Ok” an additional
layout becomes visible in the “Recover Support” section. For
changing the trays during recovery, follow the instructions in
the user manual.
40 €cilia Köstler et al.
Ca
86. “A1” is the defined position for the priming recovery, which is
not to be confused with the cell recoveries. Once selected by
right-click and “Set Priming Recover Position” it is marked in
orange and also displayed in the “Exit and Recovery” section.
87. For collecting the putative CTCs as cell groups or single cells
within the “CTC-RUO Fixed” application, it is recommended
only to use positions in columns 3, 6, 9, and 12 of the “200 μL-
tube-rack” plate layout. This results in a maximum of 33 allow-
able positions (and tubes), “Priming Recovery” included. It is
suggested to use the positions for the tubes starting from
column 3 (A3, B3, C3, etc., then columns 6, 9, and 12, accord-
ingly). The positions selected for the recovery are marked in
blue and are also displayed in the “Exit and Recovery” section.
All cells which have not been selected for recovery yet, stay in
the “Unassigned” group. Refer to user manual for more
detailed information.
88. In order to ensure purity of the samples to be recovered, it is
recommended to set a “Blank Recovery” between the different
cell types by right-clicking the position in the plate layout and
“Set Blank Recover.” The position is marked in blue and is also
displayed in the “Exit and Recovery” section. The “Exit and
Recovery” section also indicates how many drops are used for
filling each tube.
89. The “Recovery Support” is optimized for the 200 μL tubes of
the company Applied Biosystems. If other tubes are to be used, it
is necessary to get confirmation by the customer support.
90. The lid of the tube at position “A1” should be opened and
directed toward “1” (printed on the recovery tray). The same
holds true for the tube in position “A3.” All other tubes should
have their lids directed toward the letters “A”–“H” (printed on
the recovery tray). For a detailed instruction, follow the user
manual.
91. Once washing is started, it is not possible to switch back to the
CellBrowser™ and no more cells can be parked in the
parking area.
92. The process can be stopped by any user regardless of who had
signed in for starting the machine.
93. DEPArray™ cartridges can be used only once.
94. For saving the data, the connection to the backup unit is
obligatory. The backup unit has to be switched on, otherwise
the backup will fail. For more details, follow the user manual.
95. The duration of the backup process depends on the amount of
data to be saved. If a new run is performed the same day, the
backup process can be cancelled by clicking the red button with
the cross at the software screen. It is obligatory to switch off
CTC Isolation Using DEPArray 41
References
1. Miller MC, Doyle GV, Terstappen LW (2010) 5. de Bono JS, Scher HI, Montgomery RB,
Significance of circulating tumor cells detected Parker C, Miller MC, Tissing H, Doyle GV,
by the cellsearch system in patients with meta- Terstappen LW, Pienta KJ, Raghavan D (2008)
static breast colorectal and prostate cancer. J Circulating tumor cells predict survival benefit
Oncol 2010:617421. https://doi.org/10. from treatment in metastatic castration-resistant
1155/2010/617421 prostate cancer. Clin Cancer Res 14(19):
2. Alberter B, Klein CA, Polzer B (2016) Single- 6302–6309. https://doi.org/10.1158/
cell analysis of CTCs with diagnostic precision: 1078-0432.CCR-08-0872
opportunities and challenges for personalized 6. Cohen SJ, Punt CJ, Iannotti N, Saidman BH,
medicine. Expert Rev Mol Diagn 16(1):25–38. Sabbath KD, Gabrail NY, Picus J, Morse M,
https://doi.org/10.1586/14737159.2016. Mitchell E, Miller MC, Doyle GV, Tissing H,
1121099 Terstappen LW, Meropol NJ (2008) Relation-
3. Polzer B, Medoro G, Pasch S, Fontana F, ship of circulating tumor cells to tumor
Zorzino L, Pestka A, Andergassen U, Meier- response, progression-free survival, and overall
Stiegen F, Czyz ZT, Alberter B, Treitschke S, survival in patients with metastatic colorectal
Schamberger T, Sergio M, Bregola G, Doffini A, cancer. J Clin Oncol 26(19):3213–3221.
Gianni S, Calanca A, Signorini G, Bolognesi C, https://doi.org/10.1200/JCO.2007.15.8923
Hartmann A, Fasching PA, Sandri MT, Rack B, 7. Bidard FC, Peeters DJ, Fehm T, Nole F,
Fehm T, Giorgini G, Manaresi N, Klein CA Gisbert-Criado R, Mavroudis D, Grisanti S,
(2014) Molecular profiling of single circulating Generali D, Garcia-Saenz JA, Stebbing J,
tumor cells with diagnostic intention. EMBO Caldas C, Gazzaniga P, Manso L, Zamarchi R,
Mol Med 6(11):1371–1386. https://doi.org/ de Lascoiti AF, De Mattos-Arruda L,
10.15252/emmm.201404033 Ignatiadis M, Lebofsky R, van Laere SJ, Meier-
4. Cristofanilli M, Budd GT, Ellis MJ, Stopeck A, Stiegen F, Sandri MT, Vidal-Martinez J,
Matera J, Miller MC, Reuben JM, Doyle GV, Politaki E, Consoli F, Bottini A, Diaz-Rubio E,
Allard WJ, Terstappen LW, Hayes DF (2004) Krell J, Dawson SJ, Raimondi C, Rutten A,
Circulating tumor cells, disease progression, Janni W, Munzone E, Caranana V, Agelaki S,
and survival in metastatic breast cancer. N Engl Almici C, Dirix L, Solomayer EF, Zorzino L,
J Med 351(8):781–791. https://doi.org/10. Johannes H, Reis-Filho JS, Pantel K, Pierga JY,
1056/NEJMoa040766 Michiels S (2014) Clinical validity of circulating
42 €cilia Köstler et al.
Ca
Abstract
Circulating tumor cells (CTCs) undergoing epithelial-mesenchymal transition (EMT) may exhibit more
aggressive features than epithelial CTCs and are more frequently observed during disease progression.
Therefore, detection and characterization of both epithelial and mesenchymal CTCs in cancer patients are
urgently needed to allow for a better understanding of the metastatic process and more effective treatment.
Here we describe a method for detection and isolation of viable epithelial and mesenchymal CTCs from
peripheral blood of breast cancer patients. The method is based on density gradient centrifugation,
multiplex immunofluorescent staining, and negative anti-CD45 selection. Cells obtained after the proce-
dure are suitable for genomic or transcriptomic profiling, and they can also be isolated by micromanipula-
tion for single-cell analysis.
Key words Breast cancer, Circulating tumor cells, Epithelial-mesenchymal transition, Mesenchymal
phenotype
1 Introduction
Miodrag Gužvić (ed.), Single Cell Analysis: Methods and Protocols, Methods in Molecular Biology, vol. 2752,
https://doi.org/10.1007/978-1-0716-3621-3_3, © Springer Science+Business Media, LLC, part of Springer Nature 2024
43
44 Justyna Topa et al.
and unfavorable tumor characteristics [6, 7], and are more fre-
quently present in the advanced stages of the disease [11]. During
disease progression, cancer cells may change their phenotype into
the most favorable to particular conditions due to EMT and its
reverse process—mesenchymal-epithelial transition (MET)
[12, 13]; activation of EMT might promote the dissemination,
whereas reversing the mesenchymal phenotype might be crucial
for establishing new metastases [14].
Many methods allowing CTCs enrichment, detection, and
isolation were already described [15–17], with a vast majority
relying on epithelial markers (including golden standard in CTCs
detection—CellSearch®), limiting their ability to detect cells under-
going EMT, and further characterization of mesenchymal CTCs.
The method described here involves immunofluorescent (IF) stain-
ing of peripheral blood mononuclear cells (PBMCs) fraction
isolated from BC patient’s blood by density gradient centrifugation
and negative selection of CD45-positive blood cells, which is an
unbiased way of enrichment of CTCs with epithelial and mesen-
chymal features [6, 7]. For IF staining, we used antibodies directed
against common epithelial cell-surface markers, EpCAM and
E-cadherin; as well as MCAM (CD146), a protein present on the
surface of EpCAM-negative BC cell lines [18]. MCAM is associated
with mesenchymal phenotype and increased motility of BC cell
lines [19] and has been shown to improve CTCs detection in BC
patients [20]. As MCAM may be present on circulating endothelial
cells [21], the method includes CD31 (endothelial marker) in the
panel of exclusion markers, next to CD45 (blood cells marker) and
DAPI (dead cells marker). Our approach (see Subheading 2) allows
observation of epithelial markers in the “green” channel, whereas
mesenchymal markers in the “red” channel. All exclusion mar-
kers (CD31, CD45, DAPI) are visible in the “blue” channel. This
setup may be freely modified by the addition of other identified
CTCs markers. Performing IF staining before blood cells depletion
minimizes accidental loss of rare cancer cells during multiple wash-
ing steps. CD45-positive cells negative selection by Dynabeads™
CD45 is one of the most efficient methods for CTCs enrichment
regardless of their phenotype [17, 22] and in combination with IF
staining allows high recovery of living cells (Fig. 1a). The described
method also enables high enrichment of CTCs fraction, as after the
whole procedure about 98.4% of blood cells are depleted (Fig. 1b)
when 5 mL of blood was processed.
2 Materials
2.1 CTCs Enrichment 1. Low bind 1.5 mL tubes (e.g., Ultra High Recovery Tubes,
Starlab, cat. No. E1415-2600 or Protein LoBind Tubes,
Eppendorf, cat. No. 0030108116), 15 mL and 50 mL tubes.
Viable CTCs Isolation Method 45
Fig. 1 (a) The recovery rate of 100 MCF-7 (epithelial) cells and 100 MDA-MB-231 (mesenchymal) cells of BC
cell lines spiked into 5 mL of blood. Recovery rate is shown as mean ± SD from three independent
experiments. (b) Number of nucleated cells after density gradient centrifugation and after the whole procedure
as determined by processing 5 mL of blood from healthy donors (with no diagnosed cancer) samples (n = 6)
3 Methods
3.1 CTCs Enrichment 1. Prepare all buffers and reagents needed during the procedure:
Coating Buffer, 1xPBS, Histopaque® at room temperature,
and additional Coating Buffer and 1× PBS in 4 °C (see Note
3). Pre-coat 2× low-bind 1.5 mL tubes, 4 × 15 mL, and
1 × 50 mL tubes; add 1, 2, and 10 mL of Coating Buffer
(at room temperature) into tubes, respectively, and put on a
roller for 15 min to allow coating walls of the tubes with the
buffer and prevent cells sticking to the tube. After 15 min
remove the solution from the tubes.
2. By venipuncture collect blood sample into K2EDTA-coated
tubes. Discard the tube with the first 1 mL of blood that
might contain contaminating epithelial cells or fibroblasts due
to skin punctuation. Use a second K2EDTA-coated tube to
collect 5 mL of blood for the analysis (see Note 4).
3. To remove platelets, transfer blood into a pre-coated 15 mL
tube and centrifuge at 200 × g, 21 °C for 10 min. Discard the
Viable CTCs Isolation Method 47
top layer of plasma and avoid taking any of the cellular fraction
beneath it (around 2=3 can easily be removed).
4. Refill the remaining sample with 1xPBS (room temperature) to
the volume of 9 mL, gently mix, and carefully layer onto 4 mL
of sterile Histopaque® in the new pre-coated 15 mL tube. Be
careful not to mix the layers of diluted blood and Histopaque®,
as it may affect the separation of cells. Centrifuge the samples at
400 × g, 21 °C for 30 min, with no brake and acceleration (see
Note 5).
5. From that moment carry out all the steps on ice. Collect PBMCs
fraction (see Note 6) into new pre-coated 15 mL tube and refill
to the volume of 10 mL with cold (4 °C) 1xPBS. Centrifuge at
450 × g, 4 °C, 10 min to obtain a cell pellet. Discard the
supernatant and immediately proceed to the next steps of the
procedure.
Table 1
Antibodies mix used for immunofluorescent staininga
Fig. 3 (a) Spiked-in MCF-7 cells positive for epithelial markers (EpCAM and E-cadherin; green), and MDA-MB-
231 positive for MCAM (red) after CTCs isolation procedure. Spiked cells show high viability (DAPI-negative)
and are negative for CD45 and CD31 (blue). (b) Picking of single MCF-7 cell positive for epithelial markers
3.3 Single CTCs Such prepared CTCs-enriched cell suspension may be further pro-
Isolation by cessed as bulk or used for single-cell analyses. For our research,
Micromanipulation single CTCs were isolated by micromanipulation (Fig. 3) described
elsewhere in this volume. Such captured single cells may be further
subjected to genomic and transcriptomic analyses, as demonstrated
in other protocols in this volume.
4 Notes
1. Add FBS under the laminar hood, to ensure the buffer remains
sterile. It will allow longer storage of the Coating Buffer.
100 mL of the buffer is enough to carry out the CTCs isolation
procedure from two 5 mL blood samples.
2. Fluorochromes should be matched to the lasers and filters
available on a particular microscope.
3. About 25 mL of 1xPBS (10 mL at RT and 15 mL at 4 °C) and
50 mL of Coating Buffer (20 mL at RT and 30 mL at 4 °C) are
enough to perform the procedure on one 5 mL blood sample.
Histopaque® may be aliquoted into 4 mL portions, to
reach room temperature faster.
50 Justyna Topa et al.
Acknowledgments
References
1. Sung H, Ferlay J, Siegel RL, Laversanne M, 3. Fallahpour S, Navaneelan T, De P, Borgo A
Soerjomataram I, Jemal A, Bray F (2021) (2017) Breast cancer survival by molecular sub-
Global cancer statistics 2020: GLOBOCAN type: a population-based analysis of cancer reg-
estimates of incidence and mortality worldwide istry data. CMAJ Open 5(3):E734–E739.
for 36 cancers in 185 countries. CA Cancer J https://doi.org/10.9778/cmajo.20170030
Clin 71(3):209–249. https://doi.org/10. 4. Bidard FC, Peeters DJ, Fehm T, Nole F,
3322/caac.21660 Gisbert-Criado R, Mavroudis D, Grisanti S,
2. Dong G, Wang D, Liang X, Gao H, Wang L, Generali D, Garcia-Saenz JA, Stebbing J,
Yu X, Liu J (2014) Factors related to survival Caldas C, Gazzaniga P, Manso L,
rates for breast cancer patients. Int J Clin Exp Zamarchi R, de Lascoiti AF, De Mattos-
Med 7(10):3719–3724 Arruda L, Ignatiadis M, Lebofsky R, van
Laere SJ, Meier-Stiegen F, Sandri MT, Vidal-
Viable CTCs Isolation Method 51
Martinez J, Politaki E, Consoli F, Bottini A, IL-6-induced breast cancer cell EMT and stem-
Diaz-Rubio E, Krell J, Dawson SJ, ness via c-myc activation. Breast Cancer 26(5):
Raimondi C, Rutten A, Janni W, Munzone E, 663–671. https://doi.org/10.1007/s12282-
Caranana V, Agelaki S, Almici C, Dirix L, Solo- 019-00966-3
mayer EF, Zorzino L, Johannes H, Reis-Filho 13. Ocana OH, Corcoles R, Fabra A, Moreno-
JS, Pantel K, Pierga JY, Michiels S (2014) Clin- Bueno G, Acloque H, Vega S, Barrallo-
ical validity of circulating tumour cells in Gimeno A, Cano A, Nieto MA (2012) Meta-
patients with metastatic breast cancer: a pooled static colonization requires the repression of
analysis of individual patient data. Lancet the epithelial-mesenchymal transition inducer
Oncol 15(4):406–414. https://doi.org/10. Prrx1. Cancer Cell 22(6):709–724. https://
1016/S1470-2045(14)70069-5 doi.org/10.1016/j.ccr.2012.10.012
5. Cristofanilli M, Budd GT, Ellis MJ, Stopeck A, 14. Markiewicz A, Topa J, Popeda M, Szade J,
Matera J, Miller MC, Reuben JM, Doyle GV, Skokowski J, Welnicka-Jaskiewicz M, Zaczek
Allard WJ, Terstappen LW, Hayes DF (2004) A (2021) Activation of epithelial-mesenchymal
Circulating tumor cells, disease progression, transition process during breast cancer
and survival in metastatic breast cancer. N progression—the impact of molecular subtype
Engl J Med 351(8):781–791. https://doi. and stromal composition. Acta Biochim Pol
org/10.1056/NEJMoa040766 68(3):385–392. https://doi.org/10.18388/
6. Markiewicz A, Nagel A, Szade J, Majewska H, abp.2020_5719
Skokowski J, Seroczynska B, Stokowy T, 15. Bailey PC, Martin SS (2019) Insights on CTC
Welnicka-Jaskiewicz M, Zaczek AJ (2018) biology and clinical impact emerging from
Aggressive phenotype of cells disseminated via advances in capture technology. Cell 8(6).
hematogenous and lymphatic route in breast https://doi.org/10.3390/cells8060553
cancer patients. Transl Oncol 11(3):722–731. 16. Rushton AJ, Nteliopoulos G, Shaw JA,
https://doi.org/10.1016/j.tranon.2018. Coombes RC (2021) A review of circulating
03.006 tumour cell enrichment technologies. Cancers
7. Markiewicz A, Topa J, Nagel A, Skokowski J, (Basel) 13(5). https://doi.org/10.3390/
Seroczynska B, Stokowy T, Welnicka- cancers13050970
Jaskiewicz M, Zaczek AJ (2019) Spectrum of 17. Topa J, Grešner P, Żaczek AJ, Markiewicz A
epithelial-mesenchymal transition phenotypes (2022) Breast cancer circulating tumor cells
in circulating tumour cells from early breast with mesenchymal features—an unreachable
cancer patients. Cancers (Basel) 11(1). target? Cell Mol Life Sci 79(2):81. https://
https://doi.org/10.3390/cancers11010059 doi.org/10.1007/s00018-021-04064-6
8. Kalluri R, Weinberg RA (2009) The basics of 18. Mostert B, Kraan J, Bolt-de Vries J, van der
epithelial-mesenchymal transition. J Clin Spoel P, Sieuwerts AM, Schutte M, Timmer-
Invest 119(6):1420–1428. https://doi.org/ mans AM, Foekens R, Martens JW, Gratama
10.1172/JCI39104 JW, Foekens JA, Sleijfer S (2011) Detection of
9. Mani SA, Guo W, Liao MJ, Eaton EN, circulating tumor cells in breast cancer may
Ayyanan A, Zhou AY, Brooks M, Reinhard F, improve through enrichment with anti-
Zhang CC, Shipitsin M, Campbell LL, CD146. Breast Cancer Res Treat 127(1):
Polyak K, Brisken C, Yang J, Weinberg RA 33–41. https://doi.org/10.1007/s10549-
(2008) The epithelial-mesenchymal transition 010-0879-y
generates cells with properties of stem cells. 19. Zabouo G, Imbert AM, Jacquemier J,
Cell 133(4):704–715. https://doi.org/10. Finetti P, Moreau T, Esterni B, Birnbaum D,
1016/j.cell.2008.03.027 Bertucci F, Chabannon C (2009) CD146
10. Moreno-Bueno G, Portillo F, Cano A (2008) expression is associated with a poor prognosis
Transcriptional regulation of cell polarity in in human breast tumors and with enhanced
EMT and cancer. Oncogene 27(55): motility in breast cancer cell lines. Breast Can-
6958–6969. https://doi.org/10.1038/onc. cer Res 11(1):R1. https://doi.org/10.1186/
2008.346 bcr2215
11. Zhang S, Wu T, Peng X, Liu J, Liu F, Wu S, 20. Onstenk W, Kraan J, Mostert B, Timmermans
Liu S, Dong Y, Xie S, Ma S (2017) Mesenchy- MM, Charehbili A, Smit VT, Kroep JR, Nortier
mal phenotype of circulating tumor cells is JW, van de Ven S, Heijns JB, Kessels LW, van
associated with distant metastasis in breast can- Laarhoven HW, Bos MM, van de Velde CJ,
cer patients. Cancer Manag Res 9:691–700. Gratama JW, Sieuwerts AM, Martens JW, Foe-
https://doi.org/10.2147/CMAR.S149801 kens JA, Sleijfer S (2015) Improved circulating
12. Gao X, Liu X, Lu Y, Wang Y, Cao W, Liu X, tumor cell detection by a combined EpCAM
Hu H, Wang H (2019) PIM1 is responsible for and MCAM cellsearch enrichment approach in
52 Justyna Topa et al.
patients with breast cancer undergoing neoad- 22. Guo M, Li X, Zhang S, Song H, Zhang W,
juvant chemotherapy. Mol Cancer Ther 14(3): Shang X, Zheng Y, Jiang H, Lv Q, Jiang Y,
8 2 1 – 8 2 7 . h t t p s : // d o i . o r g / 1 0 . 1 1 5 8 / Hao H (2015) Real-time quantitative
1535-7163.MCT-14-0653 RT-PCR detection of circulating tumor cells
21. Strijbos MH, Gratama JW, Kraan J, Lamers from breast cancer patients. Int J Oncol
CH, den Bakker MA, Sleijfer S (2008) Circu- 46(1):281–289. https://doi.org/10.3892/
lating endothelial cells in oncology: pitfalls and ijo.2014.2732
promises. Br J Cancer 98(11):1731–1735.
https://doi.org/10.1038/sj.bjc.6604383
Chapter 4
Abstract
The study of metastasis-competent cells at the single-cell level represents an opportunity to decipher the
molecular mechanisms associated with the metastatic cascade as well as to understand the functional and
molecular heterogeneity of these cells. In this context, preclinical in vivo models of cancer metastasis are
valuable tools to understand the behavior of cancer cells throughout the process. Here we describe a
detailed protocol for the isolation and recovery of individual viable human metastatic cells from zebrafish
embryos xenotransplanted with cancer cells for downstream molecular analysis. We cover the critical steps
for the dissociation of the xenografted zebrafish embryos to generate a single-cell suspension, and the
micromanipulation for their recovery as single cells.
1 Introduction
Miodrag Guzvic (ed.), Single Cell Analysis: Methods and Protocols, Methods in Molecular Biology, vol. 2752,
https://doi.org/10.1007/978-1-0716-3621-3_4, © Springer Science+Business Media, LLC, part of Springer Nature 2024
53
54 Pablo Hurtado et al.
2 Materials
3 Methods
their behavior inside the fish body and allow an easy discrimination
between them and the host cells throughout the process.
Fig. 1 Schematic representation of the protocol for zebrafish embryo dissociation. 1. Zebrafish embryo
dissection. 2. Washing of dissected tissue. 3–4. Enzymatic and mechanical digestion of the tissues.
5. Stopping the enzymatic reaction and pelleting of the cells. 6. Obtaining the single cell suspension
the sample to dislodge the cells. Transfer the tube to the heat
block in between mechanical processing (Fig. 1, step 4) (see
Note 8).
5. Incubate the sample for no longer than 10 min at 37 °C.
6. Prepare 5 mL of cell culture medium with 10% FBS by mixing
in a sterile tube 4.5 mL of medium and 500 μl of FBS.
7. Once the incubation time is finished, add 800 μl of cell culture
medium with 10% of FBS with a micropipette to neutralize the
action of the dissociation solution and avoid cell damage
(Fig. 1, step 5).
8. Transfer the tube to a bench-top centrifuge and spin down the
cells by centrifuging at 700 g for 5 min at room temperature
(Fig. 1, step 5).
9. Carefully remove the supernatant and add 50 μl of PBS + 2%
FBS to the cellular pellet and dislodge it with a micropipette
(Fig. 1, step 6). Keep the cell suspension at room temperature
and avoid putting them in ice.
10. Optional step. A DNA stain for live cells (e.g., Hoechst 33342)
can be added to the cell suspension to visualize the nuclear
morphology and integrity. For use, follow the manufacturer’s
instructions.
11. Visualize cells under the microscope to check for the presence
of fluorescent cells (tumor cells) and their morphology.
3.3 Preparation of 1. Prepare a 5% BSA w/v solution. Weigh 500 mg of BSA and
Collection Chamber for dissolve it in 10 mL of 1× PBS. Mix well until the BSA is fully
a Cellular Suspension dissolved. Filter the solution with a 0.22 μm pore. Add 100 μl
of the 5% BSA solution at every well of a sterile chambered cell
culture slide and remove it quickly (see Note 9).
2. Differentiate within the slide at least two chambers, one as the
picking field and the other one (or others) as the sample field
(Fig. 2, step 1).
3. Add 200 μl of PBS in each picking field (see Note 10).
4. Add 190 μl of PBS in the sample field plus 10 μl of the cell
suspension (Fig. 2, step 1). It is important that the maximum
concentration of cells in the chamber is not superior to 1 × 103
cells/chamber. It will ensure an efficient single-cell isolation
without contaminating cells being carried over (Fig. 3) (see
Note 11).
5. Place the chambered cell culture slide in the fluorescence
microscope specimen holder.
6. Allow cells to set at the bottom of the sample field for a few
minutes.
Single-Cell Recovery of Metastatic Cells from Xenotransplanted Zebrafish 59
Fig. 3 Requirement of cellular density at the sample field for single-cell micromanipulation. Representative
images of a too-high cellular density (a) and an adequate cellular density (b) at sample field
7. Check under the microscope that all cells are sitting at the
bottom of the slide (see Note 12).
Fig. 4 Equipment setup for single-cell micromanipulation. This protocol is described for the use of a
mechanical microinjector and a manual micromanipulator, as shown in the images
3.5 Cell Picking Before beginning with the micromanipulation step, make sure all
the necessary setup for the micromanipulation is previously done
(Fig. 4).
1. Add 1 μl of PBS to the lid of a 0.2 mL sterile PCR tube (see
Note 15). This tube will be used to collect the retrieved cell
from the cell suspension.
2. Use a 20× microscope objective to focus the capillary and for
the aspiration of the cells from the sample field.
3. Locate the cell of interest within the sample field (see Note 16)
and carefully approach the capillary to it making use of the
control knobs. Place the tip of the capillary opposite to the
cell of interest taking the precaution of not pushing or flushing
it away.
4. Clean the area surrounding the cell of interest by flushing away
the contaminating cells with the capillary using the microinjec-
tor control knob. This can be easily done by aspirating and
flushing PBS with the capillary, which will wash away the
possible contamination.
Single-Cell Recovery of Metastatic Cells from Xenotransplanted Zebrafish 61
4 Notes
Acknowledgments
References
Abstract
Tumor heterogeneity has a major role in the development of tumor evasion and resistance to treatments. To
study and understand the intrinsic heterogeneity of cancer cells, the use of single-cell isolation technology
has had a major boost in recent years, gaining ground to bulk analysis in the study of solid tumors. In the
liquid biopsy field, the use of technologies for single-cell analysis has represented a major advance in the
study of the heterogeneity of circulating tumor cells (CTCs), providing relevant information about therapy-
resistant CTCs. However, single-cell analysis of CTCs is still challenging due to the weakness and scarcity of
these cells. In this chapter, we describe a protocol for CTCs isolation at a single-cell level using the VyCAP
Puncher system.
Key words CTC, Liquid biopsy, VyCAP, Puncher, Single cell, Sequencing, CellSearch, Tumor
heterogeneity
1 Introduction
Miodrag Gužvić (ed.), Single Cell Analysis: Methods and Protocols, Methods in Molecular Biology, vol. 2752,
https://doi.org/10.1007/978-1-0716-3621-3_5, © Springer Science+Business Media, LLC, part of Springer Nature 2024
65
66 Thais Pereira-Veiga et al.
Cell
Label and
suspension
enrich CTC
cell suspension
Automated
fluorescent imaging
and selection of
CTCs
Captured single cells
Fig. 1 Workflow scheme for isolation of single CTCs using VyCAP Puncher from a pre-enriched sample by
Cellsearch technology
2 Materials
3 Methods
3.1 Recovering 1. Remove the cartridge from the MAGNEST® cartridge holder
MAGNEST® Cartridge after scanning (see Note 1).
Content 2. Remove the cartridge plug by pushing it upwards from the
back of the cartridge (see Note 2).
3. Homogenize the sample inside the cartridge by pipetting it up
and down inside the cartridge at least 10 times, using a Pasteur
pipette or a gel-loading pipette tip. This procedure will detach
the cells from the cartridge.
4. Transfer the content of the cartridge to a 1.5 mL tube.
5. Using a pipette, add 300 μl of filtered PBS 1× inside the
cartridge by leaning the tip against the front side of the emptied
cartridge (glass side where the barcode is located). Pipette this
68 Thais Pereira-Veiga et al.
fluid up and down for ten times, rinsing the surface. The
removal of the cells can be checked by placing the cartridge
glass slide up under a standard upright fluorescence
microscope.
6. Add the 300 μl of PBS wash into the same 1.5 mL tube that the
sample was previously transferred to. The final sample volume
in the 1.5 mL tube is 600 μl.
3.2 Cell Seeding 1. Pre-wet the VyCAP microwell chip by adding 1 mL of filtered
100% ethanol on top of it. Let the chip sit with the ethanol for
45 min.
2. Connect a disposable filtration unit to the pump and insert the
microwell chip in the slot.
3. Add 1 mL of filtered PBS 1× to the remaining ethanol that is on
top of microwell chip.
4. Set the pressure of the pump station to 10 mbar (see Note 3).
5. Switch on the pump and pull most of the liquid through.
Switch off the pump action when there is only little amount
of fluid left. It is utterly important not to remove all fluid as this
will lead to air entering the microwells, which will limit the
number of available microwells.
6. Rinse the microwell chip by adding another 1 mL of filtered
PBS 1× to the chip and pulling most of the liquid through.
Repeating this step several times will completely remove the
ethanol.
7. Once the microwell chip is rinsed, add 1 mL of filtered PBS 1×
to the system and switch on the pump unit.
8. As soon as a drop of PBS has passed the microwells, add the
600 μl of the sample recovered from the MAGNEST® car-
tridge (see Note 4).
9. Switch off the pump after the whole volume of the sample has
passed.
10. Remove the microwell chip from the filtration station.
11. Wash the back side of the microwell chip with filtered PBS 1×.
12. Dry the back side of the microwell chip gently with a soft tissue
without directly touching the surface of the chip.
3.3 Isolation of CTCs 1. Transfer the microwell chip into the Puncher system to acquire
with the Punching fluorescence images.
System 2. The Puncher system contains the VyCAP Imaging system, an
automated image acquisition software. This system has auto
focus and an automatic filter cube changer that can hold a
maximum of six different filter cubes.
Isolation of Single Circulating Tumor Cells Using VyCAP Puncher System 69
4 Notes
Acknowledgement
References
1. Massague J, Obenauf AC (2016) Metastatic col- 5. Reduzzi C, Motta R, Bertolini G, Miodini P,
onization by circulating tumour cells. Nature Martinetti A, Sottotetti E, Daidone MG, Cap-
529(7586):298–306. https://doi.org/10. pelletti V (2017) Development of a protocol for
1038/nature17038 single-cell analysis of circulating tumor cells in
2. Cristofanilli M, Budd GT, Ellis MJ, Stopeck A, patients with solid tumors. Adv Exp Med Biol
Matera J, Miller MC, Reuben JM, Doyle GV, 994:83–103. https://doi.org/10.1007/978-3-
Allard WJ, Terstappen LW, Hayes DF (2004) 319-55947-6_4
Circulating tumor cells, disease progression, 6. Neumann MH, Schneck H, Decker Y,
and survival in metastatic breast cancer. N Engl Schomer S, Franken A, Endris V, Pfarr N,
J Med 351(8):781–791. https://doi.org/10. Weichert W, Niederacher D, Fehm T, Neubauer
1056/NEJMoa040766 H (2017) Isolation and characterization of cir-
3. Keller L, Pantel K (2019) Unravelling tumour culating tumor cells using a novel workflow
heterogeneity by single-cell profiling of circulat- combining the CellSearch ((R)) system and the
ing tumour cells. Nat Rev Cancer 19(10): CellCelector(). Biotechnol Prog 33(1):
553–567. https://doi.org/10.1038/s41568- 125–132. https://doi.org/10.1002/btpr.2294
019-0180-2 7. Stevens M, Oomens L, Broekmaat J, Weersink J,
4. Di Trapani M, Manaresi N, Medoro G (2018) Abali F, Swennenhuis J, Tibbe A (2018)
DEPArray system: an automatic image-based VyCAP’s puncher technology for single cell
sorter for isolation of pure circulating tumor identification, isolation, and analysis. Cytometry
cells. Cytometry A 93(12):1260–1266. A 93(12):1255–1259. https://doi.org/10.
https://doi.org/10.1002/cyto.a.23687 1002/cyto.a.23631
Chapter 6
Abstract
Many biological or pathological processes are driven by cells difficult to identify or isolate, i.e., rare cells.
Very often, these cells have elusive biology. Therefore, their detailed characterization is of utmost impor-
tance. There are many approaches that allow analysis of few or even many targets within one class of
biomacromolecules/analytes (e.g., DNA, RNA, proteins, etc.) in single cells. However, due to rarity of the
cells of interest, there is a great need to comprehensively analyze multiple analytes within these cells, in other
words to perform multi-omics analysis. In this chapter, I describe a method to isolate, separate, and amplify
total mRNA and genomic DNA of a single cells, using whole transcriptome (WTA) and whole genome
amplification (WGA). These WTA and WGA products enable simultaneous analysis of transcriptome and
genome of a single cell using various downstream high-throughput approaches.
Key words Single-cell analysis, Whole genome amplification, Whole transcriptome analysis, Multi-
omics analysis, Rare cells
1 Introduction
Miodrag Gužvić (ed.), Single Cell Analysis: Methods and Protocols, Methods in Molecular Biology, vol. 2752,
https://doi.org/10.1007/978-1-0716-3621-3_6, © Springer Science+Business Media, LLC, part of Springer Nature 2024
71
72 Miodrag Gužvić
2 Materials
2.1 Isolation of Ideally, this procedure should be performed under laminar flow
mRNA and Genomic bench in a room where no DNA amplification takes place and no
DNA, and Whole amplified DNA is stored. During execution of the protocol, gloves
Transcriptome should be exchanged frequently, especially upon touching anything
Amplification that is outside of the bench (face, chair, drawer handles, etc.). It is
advised not to wear wristwatches, rings, bracelets, etc.
2.1.1 Reagents and Kits All reagents should be molecular biology grade, and DNase and
RNAse free. All dilutions and mixtures should be made with
nuclease-free water. Reagent dilutions made from powder should
be filtered using 0.22 μm filter. All dilutions should be aliquoted to
avoid frequent freeze-thaw cycles.
. mTRAP kit (Active Motif), containing lysis buffer, protease,
streptavidin-coated beads, and biotinylated poly-T gripNA
Probe (peptide nucleic acid, PNA) oligonucleotides. Beads are
stored at 4 °C, the rest is stored at -20 °C. Individual reagents
should be prepared according to manufacturer’s instructions
(lysis buffer and beads are ready to use, protease and PNAs are
lyophilized and need to be dissolved).
. Nuclease-free water.
. Linear polyacrylamide carrier (e.g., AM9520 from Ambion, see
Note 1). Stored at 4 °C.
Single-Cell Genome and Transcriptome Amplification 73
Fig. 1 Schematic representation of WTA procedure (a) and WGA procedure (b). Please note that the
oligonucleotide sequences shown do not have to match those presented in the protocol; the figure serves
to display the principles of individual steps
74 Miodrag Gužvić
Fig. 1 (continued)
Single-Cell Genome and Transcriptome Amplification 75
2.1.2 Plasticware Plasticware in contact with sample should have low nucleic acid
binding properties.
. 0.2 mL domed cap microtubes.
. Microtubes (e.g., 0.5, 1.5, or 2.0 mL) for preparing reagent
mixes.
. 10 μl, 20 μl, 200 μl, and 1000 μl filter-tips.
. 50 mL screw-cap tube.
76 Miodrag Gužvić
2.1.4 Cycling Programs Ideally, dedicated thermal cycler used only for WTA and WGA
should be used, and it should be in the same room. Cycling pro-
grams are shown in Table 1.
2.1.5 Reagent Mixes For buffers, final concentrations of individual components are
given. For reagent mixes, volumes of individual reagents sufficient
for one sample are given; it is recommended to prepare 5–10%
more of the total reagent mix volume than needed for desired
number of samples, to account for pipette errors and “dead”
volumes. Reagent mixes composition are given in Table 2.
2.2 DNA Ideally, this procedure should be performed in the same room and
Precipitation and using the same equipment as for the WTA (see Subheading 2.1).
Whole Genome During execution of the protocol, gloves should be exchanged
Amplification frequently, especially upon touching anything that is outside of
the bench (face, chair, drawer handles, etc.). It is advised not to
wear wrist watches, rings, bracelets, etc.
2.2.1 Reagents and Kits All reagents should be molecular biology grade, and DNase and
RNAse free. All dilutions and mixtures should be made with
nuclease-free water. Reagent dilutions made from powder should
be filtered using 0.22 μm filter. All dilutions should be aliquoted to
avoid frequent freeze-thaw cycles.
. Nuclease-free water.
. 70% ethanol.
. WGA buffer pH 7.5: 100 mM Tris-acetate pH 7.8, 100 mM Mg
(CH3COO)2, 500 mM CH3COOK (see Note 9). Store at -
20 °C.
. 10% Tween 20. Store at -20 °C.
. 10% IGEPAL CA-630. Store at -20 °C.
Single-Cell Genome and Transcriptome Amplification 77
Table 1
Cycling programs used in Subheading 3.1
Table 2
Composition of reagent mixes used in Subheading 3.1
2.2.2 Plasticware Plasticware in contact with sample should have low nucleic acid
binding properties.
. 10 μl, 20 μl, 200 μl, and 1000 μl filter-tips.
. 0.2 mL domed cap microtubes.
. Microtubes (e.g., 0.5, 1.5, or 2.0 mL) for preparing reagent
mixes.
2.2.4 Reagent Mixes For reagent mixes, volumes of individual reagents sufficient for one
sample are given; it is recommended to prepare 5–10% more of the
total reagent mix volume than needed for desired number of sam-
ples, to account for pipette errors and “dead” volumes. Reagent
mixes composition are given in Table 3.
2.2.5 Cycling Programs Ideally, dedicated thermal cycler used only for WTA and WGA
should be used, and it should be in the same room. Cycling pro-
grams are shown in Table 4.
2.3 Control of the This procedure should not be performed in the room where WTA
WTA Quality or WGA takes place, nor the same equipment, plasticware, or
reagents should be used. This procedure can be performed under
the same conditions as any other conventional PCR in your lab.
2.3.1 Reagents and Kits All reagents should be molecular biology grade, and DNase and
RNAse free. All dilutions and mixtures should be made with
nuclease-free water. Reagent dilutions made from powder should
be filtered using 0.22 μm filter. All dilutions should be aliquoted to
avoid frequent freeze-thaw cycles.
80 Miodrag Gužvić
Table 3
Composition of reagent mixes used in Subheading 3.2
Table 4
Cycling programs used in Subheading 3.2
2.3.2 Plasticware Plasticware in contact with sample should have low nucleic acid
binding properties.
. 0.2 mL microtubes.
. Microtubes (e.g., 0.5, 1.5, or 2.0 mL) for preparing reagent
mixes.
. 10 μl, 20 μl, 200 μl, and 1000 μl filter-tips.
2.3.4 Reagent Mixes For reagent mixes, volumes of individual reagents sufficient for one
sample are given; it is recommended to prepare 5–10% more of the
total reagent mix volume than needed for desired number of sam-
ples, to account for pipette errors and “dead” volumes. Reagent
mixes composition are given in Table 5.
2.4 Control of the This procedure should not be performed in the room where WTA
WGA Quality or WGA takes place, nor the same equipment, plasticware, or
reagents should be used. This procedure can be performed under
the same conditions as any other conventional PCR in your lab.
2.4.1 Reagents and Kits All reagents should be molecular biology grade, and DNase and
RNAse free. All dilutions and mixtures should be made with
nuclease-free water. Reagent dilutions made from powder should
be filtered using 0.22 μm filter. All dilutions should be aliquoted to
avoid frequent freeze-thaw cycles.
Single-Cell Genome and Transcriptome Amplification 83
Table 5
Composition of reagent mixes used in Subheading 3.3
Table 6
Cycling programs used in Subheading 3.3
2.4.2 Plasticware Plasticware in contact with sample should have low nucleic acid
binding properties.
. 0.2 mL microtubes.
. Microtubes (e.g., 0.5, 1.5, or 2.0 mL) for preparing reagent
mixes.
. 10 μl, 20 μl, 200 μl, and 1000 μl filter-tips.
2.4.4 Reagent Mixes For reagent mixes, volumes of individual reagents sufficient for one
sample are given; it is recommended to prepare 5–10% more of the
total reagent mix volume than needed for desired number of sam-
ples, to account for pipette errors and “dead” volumes. Reagent
mixes composition are given in Table 7.
Table 7
Composition of reagent mixes used in Subheading 3.4
Table 8
Cycling programs used in Subheading 3.4
3 Methods
3.1 Isolation of The starting point is the sample in 0.2 mL domed cap microtubes
mRNA and Genomic containing one or more cells in 4.4 μl of mTRAP lysis buffer
DNA, and Whole supplemented with 10 ng of E. coli tRNA (see Note 12). The cells
Transcriptome can be freshly isolated or kept at -80 °C.
Amplification 1. Before bringing samples, prepare mixture containing mTRAP
lysis buffer and mTRAP protease mixed in 20:1 ratio. Take 1 μl
of this mixture and mix with 1 μl of mTRAP biotinylated poly-
T PNA oligonucleotides (see Notes 13, 14, and 15).
86 Miodrag Gužvić
A B C D
tip
beads
rack
Fig. 2 Important steps during WTA. (a) Microtubes in 50 mL screw cap tube stuffed with paper wipes (grey). (b)
Position of the pipette tip during bead washing and supernatant removal. (c) Fastening the microtubes on the
stripe of the masking duct tape. (d) Fastening the microtubes using masking duct tape to the vial/bottle of
hybridization oven
2. Bring samples into the laminar flow bench and keep on ice (see
Note 16).
3. Add 2 μl of lysis buffer:protease/PNA mix into the sample (see
Notes 17 and 18). Shortly spin in microcentrifuge.
4. Lyse the sample in thermal cycler, using cycling program LYSIS
(Table 1). See Note 19.
5. Prepare and label two sets of fresh 0.2 mL domed cap micro-
tubes. One set will be used for downstream WTA procedure
and the other for WGA procedure (see Subheading 3.2). See
Note 20.
6. Add 0.8 μl of polyacrylamide carrier into the WGA-set of
0.2 mL tubes from previous step (see Note 21). Put these
microtubes aside.
7. Thoroughly vortex mTRAP streptavidin-conjugated microbe-
ads and shortly spin in microcentrifuge (see Note 22).
8. Once the lysis is done, bring the samples under the laminar flow
bench, shortly spin them, place them on ice, and add 4 μl of
beads into each sample.
9. Stuff some paper towels into 50 mL tube, and put 0.2 micro-
tubes with samples in. Make sure that the microtubes are not
moving (Fig. 2a). Place the 50 mL tube onto the roller and
incubate 45 min at room temperature (see Notes 23 and 24).
10. Shortly before the incubation is done, turn on the hybridiza-
tion oven and set it to 44 °C.
11. Prepare RT-Mix 1 and RT-Mix 2 (without RT enzyme) and
keep them on ice (Table 2). See Note 25.
12. Once the incubation is over, spin the microtubes and place
them on ice.
Single-Cell Genome and Transcriptome Amplification 87
3.3 Checking the This procedure is a multiplex end-point PCR that amplifies three
Quality of WTA fragments of different lengths, mapping to three “housekeeping”
genes (see Note 65).
1. Label enough 0.2 microtubes for WTA samples, positive, and
negative control (see Note 66).
2. Prepare the PCR-mix (Table 5) and distribute 9.0 μl of the mix
to the tubes.
3. Add 1 μl of the WTA product. Shortly spin the tubes, place
them in thermal cycler, and run program WTAQC (Table 6).
90 Miodrag Gužvić
3.4 Checking the This procedure is a multiplex end-point PCR that amplifies four
Quality of WGA fragments of different lengths, located on MseI digested fragments
of different lengths and mapping to different regions of the
genome (see Note 70).
1. Label enough 0.2 microtubes for WGA samples, positive, and
negative control (see Note 71).
2. Prepare the PCR-mix (Table 7) and distribute 9.0 μl of the mix
to the tubes.
3. Add 1 μl of the WGA product. Shortly spin the tubes, place
them in thermal cycler, and run program WGAQC (Table 8).
4. After amplification, perform the fragment analysis using your
preferred way (see Note 67).
5. Note down the number of expected amplified fragments. The
number of bands reflects the quality of the WGA sample (see
Note 72).
4 Notes
Table 9
PCR-mix for WTA reamplification
Table 10
Cycling profile for WTA reamplification
out, and briefly spin the microtube again. If the pellet sticks to
the surface of the tips, just pipet vigorously. At this point,
damaged pellet is better than the lost pellet. Repeat superna-
tant removal, as described in step 4 of Subheading 3.2.
51. Just add ethanol along the microtube wall. Do not mix or
resuspend.
52. These steps serve to wash away buffer residues or other con-
taminating molecules from the DNA pellet.
53. In this step the complete supernatant is removed. Therefore,
pipetting near the pellet cannot be avoided. Just be extra slow
96 Miodrag Gužvić
Table 11
PCR-mix for WGA reamplification
Table 12
Cycling profile for WGA reamplification
References
1. Hay SB, Ferchen K, Chetal K, Grimes HL, Sca-1+/CD34+,- cells--the putative murine
Salomonis N (2018) The human cell atlas lung stem cells. PLoS One 8(12):e83917.
bone marrow single-cell interactive web portal. https://doi.org/10.1371/journal.pone.
Exp Hematol 68:51–61. https://doi.org/10. 0083917
1016/j.exphem.2018.09.004 10. Suzuki T, Asami M, Hoffmann M, Lu X,
2. Gužvić M, Braun B, Ganzer R, Burger M, Gužvić M, Klein CA, Perry ACF (2016) Mice
Nerlich M, Winkler S, Werner-Klein M, Czyz produced by mitotic reprogramming of sperm
ZT, Polzer B, Klein CA (2014) Combined injected into haploid parthenogenotes. Nat
genome and transcriptome analysis of single Commun 7:12676. https://doi.org/10.
disseminated cancer cells from bone marrow 1038/ncomms12676
of prostate cancer patients reveals unexpected 11. Asami M, Lam BYH, Hoffmann M, Suzuki T,
transcriptomes. Cancer Res 74(24): Lu X, Yoshida N, Ma MK, Rainbow K,
7383–7394. https://doi.org/10.1158/ Gužvić M, VerMilyea MD, Yeo GSH, Klein
0008-5472.CAN-14-0934 CA, Perry ACF (2023) A program of successive
3. Lee J, Hyeon DY, Hwang D (2020) Single-cell gene expression in mouse one-cell embryos.
multiomics: technologies and data analysis Cell Rep 42(2):112023. https://doi.org/10.
methods. Exp Mol Med 52(9):1428–1442. 1016/j.celrep.2023.112023
https://doi.org/10.1038/s12276-020- 12. Hartmann CH, Klein CA (2006) Gene expres-
0420-2 sion profiling of single cells on large-scale oli-
4. Klein CA, Schmidt-Kittler O, Schardt JA, gonucleotide arrays. Nucleic Acids Res 34(21):
Pantel K, Speicher MR, Riethmuller G (1999) e143. https://doi.org/10.1093/nar/gkl740
Comparative genomic hybridization, loss of 13. Fuhrmann C, Schmidt-Kittler O, Stoecklein
heterozygosity, and DNA sequence analysis of NH, Petat-Dutter K, Vay C, Bockler K,
single cells. Proc Natl Acad Sci U S A 96(8): Reinhardt R, Ragg T, Klein CA (2008) High-
4494–4499. https://doi.org/10.1073/pnas. resolution array comparative genomic hybridi-
96.8.4494 zation of single micrometastatic tumor cells.
5. Klein CA, Seidl S, Petat-Dutter K, Offner S, Nucleic Acids Res 36(7):e39. https://doi.
Geigl JB, Schmidt-Kittler O, Wendler N, org/10.1093/nar/gkn101
Passlick B, Huber RM, Schlimok G, Baeuerle 14. Czyz ZT, Hoffmann M, Schlimok G, Polzer B,
PA, Riethmuller G (2002) Combined tran- Klein CA (2014) Reliable single cell array CGH
scriptome and genome analysis of single micro- for clinical samples. PLoS One 9(1):e85907.
metastatic cells. Nat Biotechnol 20(4): https://doi.org/10.1371/journal.pone.
3 8 7 – 3 9 2 . h t t p s : // d o i . o r g / 1 0 . 1 0 3 8 / 0085907
nbt0402-387 15. Hoffmann M, Pasch S, Schamberger T,
6. Klein CA, Zohlnhofer D, Petat-Dutter K, Maneck M, Mohlendick B, Schumacher S,
Wendler N (2003) Gene expression analysis of Brockhoff G, Knoefel WT, Izbicki J, Polzer B,
a single or few cells. Curr Protoc Mol Biol Stoecklein NH, Klein CA (2018) Diagnostic
Chapter 25:Unit 25B 28. https://doi.org/10. pathology of early systemic cancer: ERBB2
1002/0471142727.mb25b08s61 gene amplification in single disseminated can-
7. Hahtola S, Burghart E, Jeskanen L, Karenko L, cer cells determines patient survival in operable
Abdel-Rahman WM, Polzer B, Kajanti M, esophageal cancer. Int J Cancer 142(4):
Peltomaki P, Pettersson T, Klein CA, Ranki A 833–843. https://doi.org/10.1002/ijc.
(2008) Clinicopathological characterization 31108
and genomic aberrations in subcutaneous 16. Durst FC, Grujovic A, Ganser I, Hoffmann M,
panniculitis-like T-cell lymphoma. J Invest Ugocsai P, Klein CA, Czyz ZT (2019) Tar-
Dermatol 128(9):2304–2309. https://doi. geted transcript quantification in single
org/10.1038/jid.2008.6 disseminated cancer cells after whole transcrip-
8. Chambers KF, Pearson JF, Pellacani D, Aziz N, tome amplification. PLoS One 14(8):
Gužvić M, Klein CA, Lang SH (2011) Stromal e0216442. https://doi.org/10.1371/journal.
upregulation of lateral epithelial adhesions: pone.0216442
gene expression analysis of signalling pathways 17. Werner-Klein M, Grujovic A, Irlbeck C,
in prostate epithelium. J Biomed Sci 18(1):45. Obradovic M, Hoffmann M, Koerkel-Qu H,
https://doi.org/10.1186/1423-0127-18-45 Lu X, Treitschke S, Kostler C, Botteron C,
9. Hittinger M, Czyz ZT, Huesemann Y, Weidele K, Werno C, Polzer B, Kirsch S,
Maneck M, Botteron C, Kaeufl S, Klein CA, Gužvić M, Warfsmann J, Honarnejad K,
Polzer B (2013) Molecular profiling of single Czyz Z, Feliciello G, Blochberger I,
100 Miodrag Gužvić
Abstract
The role of circulating tumor cell (CTC) clusters in the metastatic dissemination process is gaining
increased attention. Besides homotypic clusters, heterotypic clusters that contain tumor cells admixed
with normal cells are frequently observed in patients with solid tumors. Current methods used for cluster
detection and enumeration do not allow an accurate estimation of the relative fractions of tumor cells. Here
we describe a method for estimating tumor fraction of clusters including isolation and collection of single
clusters, assessment of copy number alterations of single clusters by low-pass whole genome sequencing,
and bioinformatic analysis of sequencing data.
Key words CTC clusters, Micromanipulation, Genomic analysis, Copy number alteration, Tumor
fraction prediction, Low-pass whole genome sequencing
1 Introduction
Miodrag Gužvić (ed.), Single Cell Analysis: Methods and Protocols, Methods in Molecular Biology, vol. 2752,
https://doi.org/10.1007/978-1-0716-3621-3_7, © Springer Science+Business Media, LLC, part of Springer Nature 2024
101
102 Carolina Reduzzi et al.
2 Materials
2.2 Genomic • Ampli1™ WGA Kit (Menarini Silicon Biosystems; see Note 1).
Analysis of CTC • DNA-OFF DNA removal agent (MP Biomedicals).
Clusters: WGA, QC, and
• Thermal Cycler (Applied Biosystems GeneAmp 9700 or supe-
Low-Pass CNA (See
rior; see Note 2).
Subheading 3.2)
• Set of dedicated micropipettes (Gilson).
2.2.1 Whole Genome
• PCR microcentrifuge tube 0.2 mL (Amplitube™ thin wall reac-
Amplification (WGA)
tion tube 200 μL flat snap cap, Starlab).
• Dual-filter/barrier tips (Eppendorf).
• Mini Centrifuge suitable for 0.2 mL PCR tubes.
• Laminar flow hood.
• -20 °C storage freezer.
• Vortex mixer.
• See Notes 3 and 4.
2.2.3 LowPass Copy • Ampli1™ LowPass Kit for Ion Torrent (Menarini Silicon Bio-
Number Alteration (LpCNA) systems; see Note 6).
Sequencing • DNA OFF DNA removal agent (MP Biomedicals).
• Thermal Cycler (Applied Biosystems GeneAmp 9700 or supe-
rior; see Note 2).
• Ethanol Bio Ultra, for molecular biology, ≥99.8%.
• Nuclease-free water (Molecular Biology Grade).
• Low TE: 10 mM Tris–HCl, pH 8.0, 0.1 mM EDTA).
• 0.2 mL PCR tubes (Axygen Maxymum Recovery thin wall PCR
tubes with flat cap)
• Aerosol-resistant tips and pipettes range from 1 to 1000 μL.
• SPRI select reagent (Beckman Coulter).
104 Carolina Reduzzi et al.
3 Methods
Fig. 1 The pictures show a cluster (dotted white circle) before and after cell detachment with the microma-
nipulator. The red circles indicate two single cells detached from the cluster
14. Focus the picking field and move the picking capillary by using
the Patch Man joystick into the middle of the visible field.
Lower the capillary to the surface of the picking field and
flush out the FBS (not all) using the air pump. Lift the capillary
out of the picking field (see Note 15).
15. Take 1 μL of PBS from the picking field and transfer into a
0.2 mL PCR tube containing 2 μL lysis buffer from the WGA
kit (see Subheading 3.2) and store it on ice until the end of the
isolation procedure. This refers to picking (negative) control.
16. Focus a target CTC cluster, lower the picking capillary in front
of the cluster. Try to remove the cluster by pushing or scratch-
ing it with the glass capillary. Be careful and do not destroy the
cells/cluster (Fig. 1) (see Note 16).
17. Aspirate the target cluster into the glass capillary, lift the capil-
lary out of the sample slide, focus the picking field, and lower
the glass capillary to the surface (see Note 17).
18. Take a 10 μL pipette including 10 μL pipette tip. Flush the
picked cluster out of the capillary and collect it with the pipette
within 1 μL (see Note 18).
19. Transfer the isolated cluster into a 0.2 mL PCR tube contain-
ing 2 μL of the lysis buffer from the WGA kit prepared in step 9
and store on ice until the end of the isolation procedure, and
then continue with the WGA procedure (see Note 19).
3.2 Genomic The Ampli1™ WGA method was originally designed to amplify
Analysis of CTC DNA from a single cell, but is also suitable for amplification of CTC
Clusters clusters whose cell number ranges from 2 cells to 1000. Due to the
extreme sensitivity of the WGA, it is important to avoid contami-
3.2.1 Whole Genome nation by exogenous DNA during the CTC-picking step.
Amplification (WGA) The Ampli1™ WGA method is based on a ligation-mediated
adaptor linker PCR technique, which assures a balanced amplifica-
tion due to restriction enzyme-promoted fragmentation of the
genome. The workflow for Ampli1™ WGA includes five steps
that may be executed in 1 day.
All reagents necessary to successfully execute the WGA proce-
dure are provided in the Ampli1™ WGA Kit.
1. Prepare control samples.
It is strictly recommended to process the following con-
trols along with samples in each run of Ampli1™ WGA
procedure:
• Two no-cell controls: by adding 1 μL of Ampli1™ Water.
• One positive control: by adding 1 μL of purified DNA
(1 ng/μL) extracted from a cell line. For this step use a
dedicated and decontaminated 10 μL micropipette. It is
108 Carolina Reduzzi et al.
3.2.2 Quality Control (QC) The Ampli1™QC kit contains reagents for a PCR-based assay
of WGA Products useful for the qualitative evaluation of the obtained Ampli1™WGA
products (for human-origin samples). It predicts the suitability of
each sample for downstream analyses and is based on a multiplex
PCR for four markers. Positivity of at least two markers predicts
successful genome-wide analyses to detect copy number alterations
(CNA).
Store the Ampli1™ QC Kit at -15/-20 °C. Transfer the tube
containing the enzyme to ice just prior to use. Other kit compo-
nents should be thawed, stored on ice, and briefly vortexed before
use (see Note 22).
1. It is recommended to prepare the following controls for
Ampli1™ QC reaction:
• Blank control: by adding 1 μL of PCR water.
• Positive control: by adding 1 μL of 100 ng/μL human
genomic DNA.
2. Prepare the reaction mixture according to the instructions of
the Ampli1™QC kit. Add 1 μL of Ampli1™WGA product,
prepare a blank and a positive control, and incubate in the
thermal cycler as indicated.
3. For the qualitative evaluation of PCR products, we load 1 μL of
the final product on the Agilent DNA1000 chip on the Bioa-
nalyzer Agilent 2100.
3.2.3 Low Pass CNA Ampli1™ LowPass Kit is designed to generate multiplexed,
Sequencing sequencing-ready libraries in a streamlined single-day protocol, to
detect chromosomal aneuploidies and CNA, by low-pass whole
genome sequencing using the Ion PGM or the Ion S5 systems
(see Note 6). Libraries are created through a single-reaction step
Isolation and Genomic Analysis of CTCs 109
3.3 Bioinformatic 1. Open Torrent Suite software and perform the pre-processing
Analysis and steps on raw sequences as indicated by the manufacturer:
Interpretation of • Quality check.
Sequencing Data
• Alignment to hg19 (TMAP aligner).
3.3.1 Pre-processing and 2. Obtain alignment file (.bam) from Torrent Suite. Exclude
Alignment (See Note 31) samples not passed quality control and/or samples with aligned
reads counts lower than 400,000.
3.3.2 Alignment Quality 1. Perform alignment quality control using FastqQC with stan-
Control dard settings. Do not consider “Per base sequence content,”
“Per sequence GC content,” “Sequence length distribution,”
110 Carolina Reduzzi et al.
3.3.3 Copy Number 1. Run ichorCNA on alignment file (.bam) with the following
Alteration Analysis and settings (see Note 32):
Tumor Fraction Estimation • Window = 1 Mb.
• Ploidy = 2.
• estimatePloidy = TRUE.
• estimateNormal = TRUE.
• normalPanel = TRUE.
• normal = c(0.1, 0.2, 0.3, 0.4, 0.5, 0.6, 0.7, 0.8, 0.9).
2. Retrieve the Copy number alteration calls for each segmented
genomic region (1 Mb) and estimated tumor fraction
(TF) from <sampleID>.seg.txt and < sampleID>.params.txt
files, respectively (see Note 33).
3. Do not consider chr19 during the evaluation of CNA profile
(see Note 34). Classify as unclear the CNA profiles showing
one of the following features:
• Normal profile but only 1 genomic region with amplifica-
tion/deletion lower than 125 Mb.
• Normal profile but sum of amplification/deletion of differ-
ent genomic regions lower than 375 Mb.
Classify all CNA profiles with alterations above these
thresholds as aberrant.
4. Classify samples as Normal, Tumor, or Unclear based on cri-
teria shown in the following table (see Notes 35 and 36).
Please see Fig. 2 for an example of two clusters containing
cells with aberrant DANA and one with normal diploid cells
(Table 1).
4 Notes
Fig. 2 CNA profiles obtained with low-pass WGS from 3 CTC clusters: two with aberrant CNA profiles (upper)
and one with normal CNA profile (bottom). Color codes refer to 1 copy, 2 copies, 3 copies, and more than
4 copies for each single genomic region
Table 1
CNA classification of isolated clusters
placed on the table. Then place your elbow on the box and lean
your forearm against the microscope. Caution: do not touch
any moving parts such as the microscope table with your
forearm.
19. This is a possible stopping point. Cells can be stored at -20 °C
until WGA.
20. This step can be done in parallel with STEP 1/STEP 2A by
using a different thermal cycler. Otherwise, the two steps must
be done subsequently in the same thermal cycler storing the
samples at +2–8 °C. This step is a pre-preparation of a compo-
nent for the reaction mix of STEP 3.
21. Once the program is ended, remove the tunes and store them
at -20 °C in a freezer located in a separate lab space dedicated
to downstream analysis of the amplified products.
22. Ampli1™ QC Kit is a POST WGA amplification kit. The
reaction must be prepared in the PCR working area, and
NOT in WGA working area. Pipettes for WGA should not be
used for this step. To save time all incubation steps of the
protocol should be pre-programmed on the thermal cycler.
23. In order to prevent any contamination, it is strongly recom-
mended to wear powder-free nitrile gloves for all protocol
steps, use only freshly opened plastic ware, dedicate a separate
working area, and use separate set pipettes for pre-PCR and for
library preparations. We recommend using a laminar-flow
hood and cleaning the top with DNA-off, sterile water, and
ethanol 70° before starting. To save time, all incubation steps
of the protocol should be pre-programmed on the thermal
cycler. Before starting, clean the laminar flow hood, prepare
the magnet, prepare the requested volume of ethanol 80%, the
dedicated pipettes, the LowTE buffer, the labeled 0.2 mL
tubes, and do not forget to vortex the beads making sure to
obtain a homogeneous solution. We suggest processing
12 samples in each single experiment.
24. To compensate for volume losses during pipetting, it is sug-
gested to include a 5% volume excess in each reaction mix.
When handling the barcodes, open one tube at time to prevent
any cross-contamination. Touch the side of tube with the
pipette tip and slowly dispense reagent on the side to form a
droplet.
25. Be very careful during removal of the supernatant to avoid
touching the beads loaded with the Ampli1™ WGA fragments.
Before discarding the supernatant make sure that there are no
beads in the tip. Should the pipette tip contain beads, carefully
discharge the supernatant back into its tube and wait a few
minutes to allow the optimal capture of the beads by the
magnet before removing the supernatant. For the first washing
Isolation and Genomic Analysis of CTCs 115
References
1. Castro-Giner F, Aceto N (2020) Tracking can- (2021) Circulating tumor cell clusters are fre-
cer progression: from circulating tumor cells to quently detected in women with early-stage
metastasis. Genome Med 12(1):31. https:// breast cancer. Cancers (Basel) 13(10).
doi.org/10.1186/s13073-020-00728-3 https://doi.org/10.3390/cancers13102356
2. Aceto N (2020) Bring along your friends: 8. Okonechnikov K, Conesa A, Garcia-Alcalde F
homotypic and heterotypic circulating tumor (2016) Qualimap 2: advanced multi-sample
cell clustering to accelerate metastasis. Biom J quality control for high-throughput sequenc-
43(1):18–23. https://doi.org/10.1016/j.bj. ing data. Bioinformatics 32(2):292–294.
2019.11.002 https://doi.org/10.1093/bioinformatics/
3. Heeke S, Mograbi B, Alix-Panabieres C, Hof- btv566
man P (2019) Never travel alone: the crosstalk 9. Adalsteinsson VA, Ha G, Freeman SS,
of circulating tumor cells and the blood micro- Choudhury AD, Stover DG, Parsons HA,
environment. Cell 8(7). https://doi.org/10. Gydush G, Reed SC, Rotem D, Rhoades J,
3390/cells8070714 Loginov D, Livitz D, Rosebrock D,
4. Schuster E, Taftaf R, Reduzzi C, Albert MK, Leshchiner I, Kim J, Stewart C,
Romero-Calvo I, Liu H (2021) Better Rosenberg M, Francis JM, Zhang CZ,
together: circulating tumor cell clustering in Cohen O, Oh C, Ding H, Polak P, Lloyd M,
metastatic cancer. Trends Cancer 7(11): Mahmud S, Helvie K, Merrill MS, Santiago
1020–1032. https://doi.org/10.1016/j. RA, O’Connor EP, Jeong SH, Leeson R,
trecan.2021.07.001 Barry RM, Kramkowski JF, Zhang Z,
5. Pineiro R, Martinez-Pena I, Lopez-Lopez R Polacek L, Lohr JG, Schleicher M,
(2020) Relevance of CTC clusters in breast Lipscomb E, Saltzman A, Oliver NM,
cancer metastasis. Adv Exp Med Biol 1220: Marini L, Waks AG, Harshman LC, Tolaney
93–115. https://doi.org/10.1007/978-3- SM, Van Allen EM, Winer EP, Lin NU,
030-35805-1_7 Nakabayashi M, Taplin ME, Johannessen CM,
Garraway LA, Golub TR, Boehm JS, Wagle N,
6. Silvestri M, Reduzzi C, Feliciello G, Getz G, Love JC, Meyerson M (2017) Scalable
Vismara M, Schamberger T, Kostler C, whole-exome sequencing of cell-free DNA
Motta R, Calza S, Ferraris C, Vingiani A, reveals high concordance with metastatic
Pruneri G, Daidone MG, Klein CA, Polzer B, tumors. Nat Commun 8(1):1324. https://
Cappelletti V (2021) Detection of Genomically doi.org/10.1038/s41467-017-00965-y
aberrant cells within circulating tumor micro-
emboli (CTMs) isolated from early-stage breast 10. Zhao M, Wang Q, Wang Q, Jia P, Zhao Z
cancer patients. Cancers (Basel) 13(6). https:// (2013) Computational tools for copy number
doi.org/10.3390/cancers13061409 variation (CNV) detection using next-
generation sequencing data: features and per-
7. Reduzzi C, Di Cosimo S, Gerratana L, spectives. BMC Bioinform 14(Suppl 11).
Motta R, Martinetti A, Vingiani A, https://doi.org/10.1186/1471-2105-14-
D’Amico P, Zhang Y, Vismara M, S11-S1
Depretto C, Scaperrotta G, Folli S, Pruneri G,
Cristofanilli M, Daidone MG, Cappelletti V
Chapter 8
Abstract
Cancer is a common health problem with more than 90% of deaths due to metastases. Circulating tumor
cells (CTCs) contain precursors that can initiate metastases. However, CTCs are rare, heterogeneous, and
difficult to expand in culture. We have previously created CTC-derived cell lines from stage IV breast cancer
patients. These CTC lines were used to establish single-cell CTC clones using flow cytometry cell sorting.
Key words Circulating tumor cells, Single-cell clones, CTC line culture, Breast cancer, Heterogeneity
1 Introduction
Miodrag Gužvić (ed.), Single Cell Analysis: Methods and Protocols, Methods in Molecular Biology, vol. 2752,
https://doi.org/10.1007/978-1-0716-3621-3_8, © Springer Science+Business Media, LLC, part of Springer Nature 2024
119
120 Teng Teng and Min Yu
2 Materials
2.2 CTC Culture RPMI 1640 (with phenol red) supplemented with: 20 ng/mL
Recombinant Human EGF, 20 ng/mL Recombinant Human
FGF-basic, 0.02% v/v B27 Supplement, 10 mg/mL Gentamicin,
Establishing Single-Cell Clones from In Vitro-Cultured Circulating Tumor Cells 121
a b c
Sorting Single CTC culture Clone expansion
Single CTC
Single CTC
suspended Cells reach
80% confluence
collection
Single CTC Separate to
Settle down several 96-wells
PBS+1%BSA
Single CTC
stay at bottom
space
Collect to
6-wells
Fig. 1 Illustration of the protocol. (a)-(c) show the method of establishing single cell clones from in vitro
cultured circulating tumor cells: (a) flow cytometry (Subheading 3.1); (b) single CTC culture (Subheading 3.2);
and (c) establishment of single-cell clones (Subheading 3.2)
2.3 Incubator Cells were cultured in a humid 37 C incubator with 5% CO2 and
Conditions for Single- 4% O2. Culture media was changed every 3–4 days.
Clone Cell Culture
122 Teng Teng and Min Yu
3 Methods
3.1 Flow Cytometry 1. Take the CTCs cultured in suspension in ultra-low attachment
plates and check the cell conditions under the microscope.
Generally, cells cluster in small clumps and stay in the center
of the well, and media looks clean and clear. Under the micro-
scope, CTCs should look healthy, round, clean, and clear with a
distinguishable cytoplasmic membrane, allowing light to pass
through and with minimal debris in the media (Fig. 2h).
2. Transfer CTCs to a 1.7 mL sterile Eppendorf tube and centri-
fuge for 10–12 s (speed around 1200r cf., recommended bench
mini-centrifuge; see Note 2). The cell pellet should be visible
(see Note 3). Remove the supernatant slowly using a pipet.
3. Wash the cell pellet using cold sterile 1% BSA in PBS buffer,
pipet up and down to mix, and centrifuge for 10–12 s (speed
around 1200r cf., recommended bench mini-centrifuge). The
cell pellet should be visible. Remove the supernatant slowly
using a pipet (optional; see Note 4).
4. Add 1 mL of cold sterile 1% BSA in PBS buffer to Eppendorf
tube, pipet up and down carefully about 100 times to fully mix,
and separate cells from each other (This step will affect the
sorting result.).
5. Remove the bubbles at the very top.
6. Transfer 5 μL to another Eppendorf tube, add 5 μL of Trypan
blue, mix, check under the microscope using a hemocytometer,
and ensure that the majority of cells are alive, healthy, and
single cells.
7. Add cold 1% BSA in PBS buffer to Eppendorf tube and make
the final volume 1 mL.
8. Add 7-AAD, which stains dead cells. Keep on ice, ready for
sorting (see Note 5).
9. Add DRAQ5, which stains viable cells (optional; see Note 5).
10. Sort single cells into GravityTRAP™ ULA Plate 96 well (Per-
kinElmer) that was pre-filled with CTC media 90–100 uL/
well, 1 cell per well (see Notes 6–9).
3.2 CTC Culture 1. After flow cytometry sorting, culture cells in a humid 37 °C
incubator with 5% CO2 and 4% O2 for 2–3 h. This will allow
the single cells to settle to the bottom of the well.
Establishing Single-Cell Clones from In Vitro-Cultured Circulating Tumor Cells 123
Fig. 2 Example images of an established single CTC clone at different time points. (a)–(h) show a typical cell
expansion from single BRx-68 CTC under the microscope. Magnification times is 5× for (a)–(g), and 10× for
(h). A single BRx-68 CTC in the well (a) immediately after sorting, and after being in culture for (b) 6 days, (c)
18 days, (d) 39 days, (e) 51 days, (f) 60 days, (g) 69 days. (h) A BRx-68 single clone line being moved to a
24-well plate
4 Notes
Acknowledgments
Foundation, the Pew Charitable Trusts, and the Alexander & Mar-
garet Stewart Trust (Pew-Stewart Scholar for Cancer Research) to
M.Y. We are grateful to Ms. C. Lytal for assisting with manuscript
editing.
References
1. Allemani C, Weir HK, Carreira H, Cancer Res 12(14 Pt 1):4218–4224. https://
Harewood R, Spika D, Wang XS, Bannon F, doi.org/10.1158/1078-0432.CCR-05-2821
Ahn JV, Johnson CJ, Bonaventure A, Marcos- 7. Heller G, McCormack R, Kheoh T, Molina A,
Gragera R, Stiller C, Azevedo e Silva G, Chen Smith MR, Dreicer R, Saad F, de Wit R, Aftab
WQ, Ogunbiyi OJ, Rachet B, Soeberg MJ, DT, Hirmand M, Limon A, Fizazi K,
You H, Matsuda T, Bielska-Lasota M, Fleisher M, de Bono JS, Scher HI (2018) Cir-
Storm H, Tucker TC, Coleman MP, Group culating tumor cell number as a response mea-
CW (2015) Global surveillance of cancer sur- sure of prolonged survival for metastatic
vival 1995–2009: analysis of individual data for castration-resistant prostate cancer: a compari-
25,676,887 patients from 279 population- son with prostate-specific antigen across five
based registries in 67 countries (CONCORD- randomized phase III clinical trials. J Clin
2). Lancet 385(9972):977–1010. https://doi. Oncol 36(6):572–580. https://doi.org/10.
org/10.1016/S0140-6736(14)62038-9 1200/JCO.2017.75.2998
2. Weigelt B, Peterse JL, van’t Veer LJ (2005) 8. Khoo BL, Grenci G, Lim YB, Lee SC, Han J,
Breast cancer metastasis: markers and models. Lim CT (2018) Expansion of patient-derived
Nat Rev Cancer 5(8):591–602. https://doi. circulating tumor cells from liquid biopsies
org/10.1038/nrc1670 using a CTC microfluidic culture device. Nat
3. Yadavalli S, Jayaram S, Manda SS, Madugundu Protoc 13(1):34–58. https://doi.org/10.
AK, Nayakanti DS, Tan TZ, Bhat R, 1038/nprot.2017.125
Rangarajan A, Chatterjee A, Gowda H, Thiery 9. Stott SL, Lee RJ, Nagrath S, Yu M, Miyamoto
JP, Kumar P (2017) Data-driven discovery of DT, Ulkus L, Inserra EJ, Ulman M, Springer S,
extravasation pathway in circulating tumor Nakamura Z, Moore AL, Tsukrov DI, Kemp-
cells. Sci Rep 7:43710. https://doi.org/10. ner ME, Dahl DM, Wu CL, Iafrate AJ, Smith
1038/srep43710 MR, Tompkins RG, Sequist LV, Toner M,
4. Aceto N, Bardia A, Miyamoto DT, Donaldson Haber DA, Maheswaran S (2010) Isolation
MC, Wittner BS, Spencer JA, Yu M, Pely A, and characterization of circulating tumor cells
Engstrom A, Zhu H, Brannigan BW, Kapur R, from patients with localized and metastatic
Stott SL, Shioda T, Ramaswamy S, Ting DT, prostate cancer. Sci Transl Med 2(25):25ra23.
Lin CP, Toner M, Haber DA, Maheswaran S https://doi.org/10.1126/scitranslmed.
(2014) Circulating tumor cell clusters are oli- 3000403
goclonal precursors of breast cancer metastasis. 10. Hodgkinson CL, Morrow CJ, Li Y, Metcalf
Cell 158(5):1110–1122. https://doi.org/10. RL, Rothwell DG, Trapani F, Polanski R,
1016/j.cell.2014.07.013 Burt DJ, Simpson KL, Morris K, Pepper SD,
5. Baccelli I, Schneeweiss A, Riethdorf S, Nonaka D, Greystoke A, Kelly P, Bola B, Krebs
Stenzinger A, Schillert A, Vogel V, Klein C, MG, Antonello J, Ayub M, Faulkner S,
Saini M, Bauerle T, Wallwiener M, Holland- Priest L, Carter L, Tate C, Miller CJ,
Letz T, Hofner T, Sprick M, Scharpff M, Blackhall F, Brady G, Dive C (2014) Tumori-
Marme F, Sinn HP, Pantel K, Weichert W, genicity and genetic profiling of circulating
Trumpp A (2013) Identification of a popula- tumor cells in small-cell lung cancer. Nat Med
tion of blood circulating tumor cells from 20(8):897–903. https://doi.org/10.1038/
breast cancer patients that initiates metastasis nm.3600
in a xenograft assay. Nat Biotechnol 31(6): 11. USFDA (2008) CallSearch-circulating tumor
539–544. https://doi.org/10.1038/nbt. cell kit: K073338
2576 12. Karabacak NM, Spuhler PS, Fachin F, Lim EJ,
6. Hayes DF, Cristofanilli M, Budd GT, Ellis MJ, Pai V, Ozkumur E, Martel JM, Kojic N,
Stopeck A, Miller MC, Matera J, Allard WJ, Smith K, Chen PI, Yang J, Hwang H,
Doyle GV, Terstappen LW (2006) Circulating Morgan B, Trautwein J, Barber TA, Stott SL,
tumor cells at each follow-up time point during Maheswaran S, Kapur R, Haber DA, Toner M
therapy of metastatic breast cancer patients pre- (2014) Microfluidic, marker-free isolation of
dict progression-free and overall survival. Clin
126 Teng Teng and Min Yu
circulating tumor cells from blood samples. 16. Yu M, Bardia A, Aceto N, Bersani F, Madden
Nat Protoc 9(3):694–710. https://doi.org/ MW, Donaldson MC, Desai R, Zhu H,
10.1038/nprot.2014.044 Comaills V, Zheng Z, Wittner BS, Stojanov P,
13. Sarioglu AF, Aceto N, Kojic N, Donaldson Brachtel E, Sgroi D, Kapur R, Shioda T, Ting
MC, Zeinali M, Hamza B, Engstrom A, DT, Ramaswamy S, Getz G, Iafrate AJ,
Zhu H, Sundaresan TK, Miyamoto DT, Benes C, Toner M, Maheswaran S, Haber DA
Luo X, Bardia A, Wittner BS, Ramaswamy S, (2014) Cancer therapy. Ex vivo culture of cir-
Shioda T, Ting DT, Stott SL, Kapur R, culating breast tumor cells for individualized
Maheswaran S, Haber DA, Toner M (2015) A testing of drug susceptibility. Science
microfluidic device for label-free, physical cap- 345(6193):216–220. https://doi.org/10.
ture of circulating tumor cell clusters. Nat 1126/science.1253533
Methods 12(7):685–691. https://doi.org/ 17. De Mattos-Arruda L, Cortes J, Santarpia L,
10.1038/nmeth.3404 Vivancos A, Tabernero J, Reis-Filho JS, Seoane
14. Fachin F, Spuhler P, Martel-Foley JM, Edd JF, J (2013) Circulating tumour cells and cell-free
Barber TA, Walsh J, Karabacak M, Pai V, Yu M, DNA as tools for managing breast cancer. Nat
Smith K, Hwang H, Yang J, Shah S, Rev Clin Oncol 10(7):377–389. https://doi.
Yarmush R, Sequist LV, Stott SL, org/10.1038/nrclinonc.2013.80
Maheswaran S, Haber DA, Kapur R, Toner M 18. Messaritakis I, Stoltidis D, Kotsakis A, Dermit-
(2017) Monolithic chip for high-throughput zaki EK, Koinis F, Lagoudaki E,
blood cell depletion to sort rare circulating Koutsopoulos A, Politaki E, Apostolaki S,
tumor cells. Sci Rep 7(1):10936. https://doi. Souglakos J, Georgoulias V (2017) TTF-1-
org/10.1038/s41598-017-11119-x and/or CD56-positive circulating tumor cells
15. Chen L, Peng M, Li N, Song Q, Yao Y, Xu B, in patients with small cell lung cancer (SCLC).
Liu H, Ruan P (2018) Combined use of Sci Rep 7:45351. https://doi.org/10.1038/
EpCAM and FRalpha enables the high- srep45351
efficiency capture of circulating tumor cells in 19. Hunter KW, Amin R, Deasy S, Ha NH, Wake-
non-small cell lung cancer. Sci Rep 8(1):1188. field L (2018) Genetic insights into the morass
https://doi.org/10.1038/s41598-018- of metastatic heterogeneity. Nat Rev Cancer
19391-1 18(4):211–223. https://doi.org/10.1038/
nrc.2017.126
Chapter 9
Abstract
Immunofluorescence (IF) microscopy is arguably one of the most commonly used methods for studying
structure and composition of stress granules (SGs). While in most cases standard IF protocols are sufficient
to visualize protein components of SGs, concurrent detection of proteins and transcripts in stress granules
requires more sophisticated and problematic approaches. Here we present a well-established, simple,
robust, and fluorescent protein-compatible method for simultaneous detection of proteins and transcripts
in individual stress granules using combination of IF and single-molecule RNA fluorescence in situ
hybridization (smRNA FISH).
Key words Microscopy, Immunofluorescence, smRNA FISH, Stress granules, RNA, lncRNA, Single-
cell analysis
1 Introduction
Miodrag Gužvić (ed.), Single Cell Analysis: Methods and Protocols, Methods in Molecular Biology, vol. 2752,
https://doi.org/10.1007/978-1-0716-3621-3_9, © Springer Science+Business Media, LLC, part of Springer Nature 2024
127
128 Jakub Kochan and Mateusz Wawro
2 Materials
Read this section carefully and ensure to have all reagents prepared
before beginning the procedure. Also make sure to read the
detailed protocol and respective notes in advance. Observe caution
not to introduce RNase contamination into your reagents and
Protein and mRNA Detection Using Simultaneous IF and RNA-FISH 129
Fig. 1 Formation of stress granules is a result of subcellular re-localization of specific components rather than
change in their cellular level. (a) Western blot analysis of extracts from HeLa cells, control or sodium arsenite-
treated (500 μM, 30 and 60 min), using anti-G3BP1 (upper panel), anti-phospho-eIF2α (Ser51, middle panel),
and anti-lamin (loading control, lower panel) antibodies. While no change in total G3BP level is detected upon
treatment with sodium arsenite, observed phosphorylation of eIF2α clearly reveals exposure of cells to stress
conditions. (b) Immunofluorescence-based approach allows observation of stress granules formed upon
treatment of the cells with sodium arsenite. Images of HeLa cells with visualized G3BP1, a key marker of
stress granules (red) in control and sodium arsenite-treated cells. Blue: DAPI (nuclei). Scale bar: 10 μm
Fig. 2 The humidifying chamber assembly. Components used to build the humidifying chamber are presented
in the left picture: (1) paper towels, (2) 50 mL conical tube screws, (3) parafilm, (4) distilled water, (5) glass
plate, and (6) glass Petri dish. Right image presents a typical view of an assembled chamber (a – a glass plate
with piece of parafilm placed on top (stage for coverslips), (b) – an orientation mark on the parafilm used for
proper alignment of coverslips)
2.4 smRNA FISH 1. 20× SSC buffer: 3 M sodium chloride, 300 mM sodium citrate.
We use Ambion 20× SSC solution (Invitrogen). Store at
RT. 20× SSC can be also easily prepared in house (see Note 8).
2. 2× SSC buffer. Prepare in a 50 mL sterile, RNase-free conical
tube. For 50 mL mix 5 mL 20× SSC and 45 sterile, RNase-free
water. Store at RT.
3. Wash buffer: 2× SSC, 10% (v/v) formamide. Prepare in a
50 mL sterile, RNase-free conical tube. For 50 mL, mix 5 mL
20× SSC, 5 mL formamide, and add sterile, RNase-free water
to 50 mL (see Note 9). Sterile filter through a 0.2 μm syringe
filter. Store at RT.
132 Jakub Kochan and Mateusz Wawro
Fig. 3 Specificity verification of used probe blend targeting lncRNA NORAD. (a) Detection of NORAD (red) using
smRNA FISH in wild-type HeLa cells (left panel), mock-transfected (middle panel), and HeLaiKD cells with
induced NORAD knock-down (right panel). Blue: DAPI (nuclei). Scale bar: 10 μm (b) Quantification of the
smRNA FISH experiments shown in (a). Twenty-five cells were analyzed for each condition. (C) qPCR analysis
of NORAD expression in mock-transfected and HeLaiKD cells with induced NORAD knock-down. Bars: average
from two independent experiments with two technical replicates. Error bars: 95% CI
3 Methods
3.1 Plating and 1. Using tweezers place sterile glass coverslips in a multi-well cell
Treatment of the Cells culture plate and seed the cells at the desired density suitable for
single-cell imaging and analysis. Usually 50,000–60,000 cells
per well of 12-well plate in 1 mL of medium should be suffi-
cient. In general, it is not recommended to let the cells reach
100% confluence (i.e., monolayer) at the time of fixation (see
Note 11).
2. Place the cells in a dedicated incubator for 24–48 h to allow
proper attachment and accommodation. If needed, change the
medium at desired time point or treat the cells according to
your experimental requirements.
3. Induce stress granule formation (see Note 12) and proceed
immediately to subsequent steps.
Fig. 4 Picture of the bent needle attached to a syringe used to manipulate coverslips. Zooms present the tip of
the bent needle in more details
3.4 Single-Molecule 1. Prepare fresh smRNA FISH wash buffer and thaw a fresh
RNA Fluorescence In aliquot of hybridization buffer.
Situ Hybridization 2. Remove PBS and equilibrate the specimens by incubation in
smRNA FISH wash buffer (1 mL/well) for 5–10 min at RT.
3. During specimen equilibration, prepare probe blend working
solution. Dilute the probe blend stock in hybridization buffer.
When calculating the volume of the probe, you need to follow a
similar routine as for the primary and secondary antibodies.
Take 50 μl of probe droplet per coverslip and add 50 μl excess
to compensate for liquid losses.
4. Equilibrate the humidifying chamber to 37 °C and prepare a
glass plate with a piece of parafilm placed on it (use the same
chamber construction as in previous steps).
5. Place droplets of probe blend (50 μl/coverslip) on the parafilm.
6. Use tweezers and a bent needle (yet another new one) attached
to a syringe to place coverslips (cells toward the droplet) onto the
probe blend droplets (see Note 18).
7. Hybridize the cells with probes at 37 °C overnight in the dark.
8. Following the overnight hybridization, gently transfer the spe-
cimens into a fresh, sterile cell culture plate wells filled with
smRNA FISH wash buffer prewarmed to 37 °C (rotate the
Protein and mRNA Detection Using Simultaneous IF and RNA-FISH 137
4 Notes
Fig. 6 Comparison of smRNA FISH images (maximum intensity projections) before (left panel) and after
deconvolution (right panel). Scale bar: 10 μm
Fig. 7 Hyperthermia and sodium arsenite induce stress granule formation and lead to sequestration of lncRNA
NORAD in stress granules. IF-smRNA FISH images of HeLa cells stained to visualize NORAD (red) and G3BP1, a
key marker of SGs (green), during two different types of stresses (hyperthermia and sodium arsenite). Blue:
DAPI (nuclei). Scale bars: 10 μm (upper panels), 1 μm (lower panels)
Acknowledgement
References
1. Panas MD, Ivanov P, Anderson P (2016) Franzmann TM (2020) RNA-induced confor-
Mechanistic insights into mammalian stress mational switching and clustering of G3BP
granule dynamics. J Cell Biol 215(3): drive stress granule assembly by condensation.
313–323. https://doi.org/10.1083/jcb. Cell 181(2):346–361 e317. https://doi.org/
201609081 10.1016/j.cell.2020.03.049
2. Protter DSW, Parker R (2016) Principles and 7. Sanders DW, Kedersha N, Lee DSW, Strom
properties of stress granules. Trends Cell Biol AR, Drake V, Riback JA, Bracha D, Eeftens
26(9):668–679. https://doi.org/10.1016/j. JM, Iwanicki A, Wang A, Wei MT,
tcb.2016.05.004 Whitney G, Lyons SM, Anderson P, Jacobs
3. Molliex A, Temirov J, Lee J, Coughlin M, WM, Ivanov P, Brangwynne CP (2020) Com-
Kanagaraj AP, Kim HJ, Mittag T, Taylor JP peting protein-RNA interaction networks con-
(2015) Phase separation by low complexity trol multiphase intracellular organization. Cell
domains promotes stress granule assembly and 181(2):306–324 e328. https://doi.org/10.
drives pathological fibrillization. Cell 163(1): 1016/j.cell.2020.03.050
123–133. https://doi.org/10.1016/j.cell. 8. Yang P, Mathieu C, Kolaitis RM, Zhang P,
2015.09.015 Messing J, Yurtsever U, Yang Z, Wu J, Li Y,
4. Perez-Pepe M, Fernandez-Alvarez AJ, Boccac- Pan Q, Yu J, Martin EW, Mittag T, Kim HJ,
cio GL (2018) Life and work of stress granules Taylor JP (2020) G3BP1 is a tunable switch
and processing bodies: new insights into their that triggers phase separation to assemble stress
formation and function. Biochemistry 57(17): granules. Cell 181(2):325–345 e328. https://
2488–2498. https://doi.org/10.1021/acs. doi.org/10.1016/j.cell.2020.03.046
biochem.8b00025 9. Khong A, Matheny T, Jain S, Mitchell SF,
5. Van Treeck B, Parker R (2019) Principles of Wheeler JR, Parker R (2017) The stress gran-
stress granules revealed by imaging approaches. ule transcriptome reveals principles of mRNA
Cold Spring Harb Perspect Biol 11(2). accumulation in stress granules. Mol Cell
https://doi.org/10.1101/cshperspect. 68(4):808–820 e805. https://doi.org/10.
a033068 1016/j.molcel.2017.10.015
6. Guillen-Boixet J, Kopach A, Holehouse AS, 10. Kochan J, Wawro M, Kasza A (2015) Simulta-
Wittmann S, Jahnel M, Schlussler R, Kim K, neous detection of mRNA and protein in single
Trussina I, Wang J, Mateju D, Poser I, cells using immunofluorescence-combined sin-
Maharana S, Ruer-Gruss M, Richter D, gle-molecule RNA FISH. Biotechniques
Zhang X, Chang YT, Guck J, Honigmann A, 59(4):209–212, 214, 216 passim. https://
Mahamid J, Hyman AA, Pappu RV, Alberti S, doi.org/10.2144/000114340
Chapter 10
Abstract
In situ hybridization of oligonucleotide probes to intracellular RNA allows quantification of predefined
gene transcripts within millions of single cells using cytometry platforms. Previous methods have been
hindered by the number of RNA that can be analyzed simultaneously. Here we describe a method called
proximity ligation assay for RNA (PLAYR) that permits highly multiplexed RNA analysis that can be
combined with antibody staining. Potentially any number of RNA combined with antigen can be analyzed
together, being limited only by the number of analytes that can be measured simultaneously.
Key words Transcriptome analysis, Flow cytometry, Single-cell analysis, Single-cell multi-omics, In
situ hybridization, Mass spectrometry, Proteome analysis, Proximity ligation assay for RNA (PLAYR)
1 Introduction
Miodrag Gužvić (ed.), Single Cell Analysis: Methods and Protocols, Methods in Molecular Biology, vol. 2752,
https://doi.org/10.1007/978-1-0716-3621-3_10, © Springer Science+Business Media, LLC, part of Springer Nature 2024
143
144 Andrew D. Duckworth et al.
A
Probe1
5’-NNNNNNNNNNNNNNNNNNNAAAAAAAAAANNNNNNNNNGACACTCTT-3’
Probe2
5’-NNNNNNNNNNNNNNNNNNNAAAAAAAAAAGACGCTAATNNNNNNNNN-3’
RNA-binding sequence Linker backbone- insert-
binding binding
sequence sequence
B
Backbone
Insert
…CTGCGATTA NNNNNNNNNTTTNNNNNNNNN CTGTGAGAA…
GACGCTAATNNNNNNNNN NNNNNNNNNGACACTCTT RCA begins
AAAAAAAAAA
AAAAAAAAAA
here
Probe2 Probe1
NNNNNNNNNNNNNNNNNNN NNNNNNNNNNNNNNNNNNN
5’ –NNNNNNNNNNNNNNNNNNNNNNNNNNNNNNNNNNNNNNNNNNNNNNNNNN-3’ RNA
Fig. 1 Overview of PLAYR probe design and cognate interactions between oligonucleotides within the PLAYR
systems. (a) Overview of the PLAYR probe design, with each sequence component shaded differently. (b)
Diagram showing sequence interactions between different oligonucleotides during PLAYR. “N” represents
nucleotides that vary depending upon the target (RNA-binding sequence) and the PLAYR system (insert-
binding sequence) used
Table 1
Example of a 6-choose-3 LCB scheme using Cadmium isotopes
Cadmium Isotopes
106 110 111 113 114 116
Barcode 1 x x x
2 x x x
3 x x x
4 x x x
5 x x x
6 x x x
7 x x x
8 x x x
9 x x x
10 x x x
11 x x x
12 x x x
13 x x x
14 x x x
15 x x x
16 x x x
17 x x x
18 x x x
19 x x x
20 x x x
An antibody (or antibodies) that target constituently and highly expressed antigen(s) are
labeled with different cadmium isotopes and combined in sets of three (x’s) to produce
LCBs. Cadmium isotopes can be substituted for other metal isotopes and can be tailored
around your panel design. If antibodies target the same epitope and their staining signal
is weak due to competition for antigen binding, barcoding may perform better with just
two isotopes per barcode (e.g., a 7-choose-2 scheme provides 21 barcodes)
(continued)
Table 2
148
(continued)
Table 3
List of 5′ PLAYR probe sequences that target constituently expressed RNA in human cells
Probe
Gene pair 5′ Probe1 5′ Probe2
GAPDH 1 TGATGGCAACAATATCCACTTT GTTAAAAGCAGCCCTGGTGA
2 TGGAAGATGGTGATGGGATT ATTGATGACAAGCTTCCCGT
3 TGGACTCCACGACGTACTCA CATCGCCCCACTTGATTTT
4 CCTGCTTCACCACCTTCTTG TCATCATATTTGGCAGGTTTTT
ACTB 1 TGCCAATGGTGATGACCTG GTCAGGCAGCTCGTAGCTCT
2 TGTTGGCGTACAGGTCTTTG ATGTCCACGTCACACTTCATG
3 CGTCATACTCCTGCTTGCTG ATCTGCTGGAAGGTGGACAG
4 TGTTTTCTGCGCAAGTTAGGT TCAAGAAAGGGTGTAACGCA
RPL27 1 GTTTCATGAACTTGCCCA GGCCCTACCAGCAAAAAGGAAAG
TTTCGG
2 GCGATCTGAGGTGCCATCATCAA TCTTCACGATGACAGCTTTGCG
T TC
3 TCTCTTGGCGATCTTCTTC GCTGTCACTTTGCGGGGGTAG
TTGCC
4 TCTTCAAACTTGACCTTGGCC GCCTTGCGTTTAAGAGCAGGATC
TCC T
5 AGCATCTAAAACCGCAGTTTC ACCACTTGTTCTTGCCTGTCTTG
TGG T
Table shows predesigned 5′ sequences for PLAYR probe pairs targeting the indicated “housekeeping” transcripts. To
complete the PLAYR probe, the 3′ Probe1 or 2 sequence from the PLAYR systems table (Table 2; any system may be
chosen) needs to be added to their 3′ end
Time(min)
0
PMA+Iono
30
60
120
240
360
RNA1 RNA2 RNA3 RNA4 RNA5 RNA6 RNA7 ACTB
B C RNA1 RNA2
RNA1 RNA2
qPCR (a.u)
Fig. 2 Validation of PLAYR probe specificity. B-cell line MAVER1 treated with 100 nM phorbol 12-myristate
13-acetate (PMA) + 1 μM ionomycin (PMA + Iono) for up to 6 h was examined by PLAYR (mass cytometry) and
qPCR for expression changes in eight RNAs (including ACTB). (a) Overlayed histograms showing single-cell
expression for each of the eight transcripts at each timepoint. (b) Comparison of mean PLAYR signal (right-
hand y axis) with quantification by qPCR (left-hand y axis) for 4 RNAs shown in (a) over the time course with
PMA + Iono. (c) Linear regression of data shown in (b) showing concordance of relative signal for PLAYR
(y axis) and qPCR (x axis) at each timepoint; gray shading indicates 95% confidence intervals
last step of PLAYR where DDP is added. After RNA and internal
antigen staining, cells should be run immediately for analysis by
flow cytometry. In contrast, cells can be left in fixative and analyzed
up to 48 h later using mass cytometry. Instructions on how to
process samples for analysis by mass cytometry are described in
Methods Subheading 3.4 of the protocol. The protocol has the
potential to be adapted for non-suspension cells for quantification
of RNA in tissues or adherent cells using microscopy.
2 Materials
All buffers are made and stored at room temperature (RT) unless
stated otherwise. Filter tips should be used for all mass cytometry
and PLAYR methods. All reagents should be RNase and DNase
152 Andrew D. Duckworth et al.
Fig. 3 Schematic of PLAYR staining procedure. Schematic overview of the PLAYR assay, with the top panel
showing the stages (1–5) for sample preparation and the bottom panel illustrating the stages (6–10) for RNA
staining with PLAYR. Stages 1–4 are all optional and, if not required, can be excluded without affecting the
detection of RNA with PLAYR. For PLAYR-CYTOF, live cell barcoding can be performed first (stage 1), allowing
multiple samples to be pooled into one tube. Dead cells can then be stained at RT (stage 2) followed by surface
antigen staining on ice (stage 3). Covalent crosslinking of surface antibodies using BS3 reagent is then
performed (stage 4) on ice to help maintain antigen binding during the remaining stages of PLAYR. Cells are
then fixed in PFA and permeabilized with methanol in stage 5. Once in methanol, cells can be stored at -80 °C
allowing the assay to be paused. Stages 6–10 illustrate the sequential steps of oligonucleotide binding and
amplification used by PLAYR. Internal antibody staining can be included in the final stage alongside incubation
with DDPs
2.3 LCB for PLAYR- 1. Metal-conjugated antibodies for LCB: Dilute antibodies in
CyTOF (Optional) CSB to make 100 μL barcoding solution per 3 × 106 cells
stained (see Note 3). Antibody solutions can be made 2 h
before use and stored at 4 °C. See Table 1 for an example of
preparing an LCB scheme for PBMC using cadmium-
conjugated anti-CD45 antibodies.
2.4 Staining to 1. Dead cell exclusion reagent: For mass cytometry we use Cell-
Identify Dead Cells ID cisplatin (Fluidigm, cat. no. 201064). For flow cytometry
(Optional) we use Live/Dead Fixable Dead Cell Stain Kits diluted 1:1000
in RT PBS (Invitrogen). Reagents should be diluted 1:1000
immediately before use in RT PBS.
2.5 Surface Antibody 1. Metal- (for mass cytometry [19]; Fluidigm) or fluorophore-
Staining (Optional) conjugated (for flow cytometry) antibodies for surface antigen
phenotyping (see Note 2): Dilute antibodies in CSB to make
100 μL of antigen phenotyping solution per 3 × 106 cells
stained (see Note 3). Antibody cocktails can be made up to
2 h before use and stored at 4 °C.
2. 5 mM Bis(sulfosuccinimidyl) suberate (BS3 crosslinker): Dis-
solve BS3 crosslinker in ice-cold PBS (see Note 4).
2.6 Cell Fixation and 1. Ice-cold 1.6% Paraformaldehyde (PFA): Dilute 16% PFA using
Permeabilization ice-cold PBS and place on ice. 1.6% PFA can be made up to
1 day before use.
2. Ice-cold methanol.
12. PLAYR Phi29 buffer wash: 1× Phi29 buffer (as supplied by the
manufacturer) diluted in ddH2O. Make fresh up to 20 min
before use in Subheading 3.3, Step 20.
13. PLAYR Phi29 amplification mix: 1× Phi29 buffer (as supplied
by the manufacturer), 5 U/mL Phi29 DNA polymerase, and
800 μM dNTP. Dilute in RNase-free ddH2O. Make fresh up to
20 min before use in Subheading 3.3, Step 20.
14. PLAYR mass cytometry detection probe buffer: 10 nM of each
metal-labeled DDP diluted in cell staining buffer. Use this
buffer if performing analysis by mass cytometry. If internal
antigens are also being analyzed, an appropriate concentration
of metal-labeled antibodies should be added to this buffer.
Make fresh up to 2 h before use in Subheading 3.3, Step 24.
15. PLAYR flow cytometry detection probe buffer: 1 nM of each
fluorescent-labeled DDP diluted in CSB. Use this buffer if
performing analysis by flow cytometry. If internal antigens are
also being analyzed, an appropriate concentration of
fluorescent-labeled antibodies should be added to this buffer.
Make fresh up to 2 h before use in Subheading 3.3, Step 24.
2.9 Software 1. Fluidigm CyTOF system control software for mass cytometry
(www.fluidigm.com/software).
2. NxT software (Thermofisher; cat. no. A25554) for the Attune
flow cytometer (or equivalent).
3. Software appropriate for analysis of multiparametric mass cyto-
metry data files (e.g., Cytobank (https://www.cytobank.org/),
Cytosplore (https://www.cytosplore.org/), etc.).
4. R software (https://www.r-project.org/) for PLAYR probe
design.
5. PLAYRDesign software to design PLAYR probes for your
RNA of interest (detailed instructions for installation can be
found at https://github.com/nolanlab/PLAYRDesign).
156 Andrew D. Duckworth et al.
3 Methods
3.1 Labeling DDPs 1. Calculate or make a note of the starting number of nmol for
with Metal Isotopes for each detection probe as this will be required during calculations
Mass Cytometry in step 18.
2. Add 500 mM TCEP to the 250 μM DDP to a final concentra-
tion of 50 mM (1 in 10) and incubate at RT for 30 min.
3. If required, raise the volume to a minimum of 200 μL using
ddH2O and then add sodium acetate to a final concentration of
300 mM. Mix by vortexing.
4. Add three volumes of ice-cold 100% ethanol and mix by vortex.
5. Place the tube at -20 °C overnight to allow precipitation.
Alternatively, precipitation and longer-term storage can be
performed at -80 °C for up to 6 months.
6. From the Maxpar X8 Antibody Labeling Kit, centrifuge the X8
polymer tube 4000rcf/30 s/RT) to ensure the reagent is at the
bottom.
7. Dissolve the polymer in 95 μL of L-Buffer by pipetting.
8. Add 5 μL of lanthanide metal solution and mix by pipetting.
9. Incubate the polymer-metal mix at 37 °C for 30 min.
10. While the polymer-metal mix is incubating (step 9), centrifuge
the precipitated DDP from step 5 at 14,000rcf/4 °C/30 min.
11. Add 200 μL of L-Buffer to a 3 kDa MWCO filter.
12. After the incubation in step 9, add the metal-loaded polymer
to the filter containing the 200 μL L-Buffer.
13. Centrifuge metal-conjugated polymer at 12,000rcf/RT/
30 min (see Note 11).
14. Once the centrifuge step is completed from step 10, carefully
remove supernatant from DDP pellet. Resuspend in 400 μL
ice-cold 75% ethanol and centrifuge at 14,000rcf/4 °C/
10 min.
15. Retrieve 3 kDa filter from centrifuge (from step 13) and
discard flow through. Add 300 μL of C-Buffer to the filter.
Centrifuge at 12,000rcf/RT/30 min; leave at RT while com-
pleting detection probe purification.
16. Repeat step 14 to wash the DDP pellet a second time.
17. After the final wash of the DDP in step 16, remove as much
ethanol as possible (a brief centrifuge step (14,000rcf/4 °C/
30 s) may help to pool residual ethanol for removal). Air-dry
the pellet (the tube can be placed at 40 °C on a heat block to
decrease drying time, see Note 12).
Highly Multiplexed and Simultaneous Characterization of Protein and RNA in. . . 157
3.2 Surface Antibody LCB for PLAYR-CyTOF, staining to identify dead cells, and sur-
Staining, Dead Cell face antibody staining are all independent staining procedures and,
Exclusion, and if not required, each section can be excluded without affecting one
Preparing Cells for another or the detection of RNA by PLAYR. To minimize changes
PLAYR in transcript expression during surface antibody staining, cells
158 Andrew D. Duckworth et al.
should be kept cold (between 1 °C and 4 °C) for all steps unless
stated otherwise. All incubation steps for flow cytometry during
and post binding of fluorophore-conjugated reagents should be
performed in the dark to reduce quenching.
1. Aliquot 3 × 106 cells into an Eppendorf tube on ice for each
sample (see Notes 13 and 14).
2. Pellet cells by centrifugation at 500rcf/4 °C/3 min. (See Notes
15 and 16.)
3. Wash in ice-cold PBS by centrifugation at 500rcf/4 °C/3 min.
LCB for PLAYR-CyTOF (Optional)
4. Resuspend each sample in its appropriate LCB antibody solu-
tion (3 × 106 cells per 100 μL) and incubate on ice for 30 min.
Resuspend cells by agitation after 15 min.
5. Pellet cells by centrifugation at 500rcf/4 °C/3 min.
6. Wash pellet twice in 200 μL ice-cold CSB.
7. Resuspend in 200 μL ice-cold PBS and pool barcoded samples
together into one tube.
8. Centrifuge at 500rcf/4 °C/5 min and remove supernatant.
Staining to Identify Dead Cells (Optional)
9. Resuspend cells in 1 mL dead cell exclusion reagent per 107
cells.
10. Incubate at RT for 5 min.
11. Add 4× volume of ice-cold CSB to quench unreacted dead cell
exclusion dye and place tubes on ice.
12. Pellet cells by centrifugation at 500rcf/4 °C/5 min and
remove supernatant.
Surface Antibody Staining (Optional)
13. Resuspend cells in surface antibody cocktail and incubate on
ice for 30 min. Resuspend cells by agitation after 15 min.
14. Wash cells twice in ice-cold CSB.
15. Wash cells in ice-cold PBS.
16. Resuspend cells in 5 mM BS3 crosslinker (1 mL per 107 cells)
and incubate on ice for 30 min (see Note 4). Resuspend cells
by agitation after 15 min.
17. Pellet cells by centrifugation at 500rcf/4 °C/3 min and
remove supernatant.
Cell Fixation and Permeabilization
18. Resuspend cells in ice-cold 1.6% PFA (1 mL per 3 × 106 cells)
and incubate on ice for 10 min.
Highly Multiplexed and Simultaneous Characterization of Protein and RNA in. . . 159
4 Notes
20. Once cells are removed from methanol RNA degradation can
set in. Speed in processing during the PBSTR+ wash step and
fast transfer into the PLAYR probe hybridization buffer is
essential to maintain RNA integrity.
21. Due to the small volumes used, ensure that all wash buffer is
removed from the pellet before the addition of PLAYR hybri-
dization buffer to the cells in Subheading 3.3 Step 6.
22. Cool on ice to avoid denaturing RNasin in the PLAYR hybri-
dization buffer.
23. Incubation temperatures of PLAYR hybridization buffer and
post hybridization wash can be reduced to 37 °C to help
maintain surface antibody binding. Specificity for your specific
PLAYR probes should be confirmed while using these milder
conditions.
24. The RCA times using Phi29 can be made longer to increase the
signal from the assay. However, we have found that longer
RCA times (past the 2 h and 4 h suggested) do not increase
sensitivity of the assay (i.e., negative cells remain negative). As
mentioned in the introduction, the best way to increase sensi-
tivity is by increasing the number of probe pairs used to target
the RNA.
25. PLAYR-CyTOF signals will slowly depreciate the longer you
leave the samples at 4 °C. We recommend running the samples
within 48 h of finishing the assay to achieve maximum signal.
26. Cell concentrations may be adjusted depending upon how
readily they clog the mass cytometer. Larger or more sticky
cells may require running at lower concentrations.
27. Vigorously shake the EQ Four Element Calibration Bead bot-
tle for 30 s to make sure the contents are evenly resuspended
before adding to the sample.
References
1. Bauman JG, Bayer JA, van Dekken H (1990) 3. Lalli E, Gibellini D, Santi S, Facchini A (1992)
Fluorescent in-situ hybridization to detect cel- In situ hybridization in suspension and flow
lular RNA by flow cytometry and confocal cytometry as a tool for the study of gene
microscopy. J Microsc 157(Pt 1):73–81. expression. Anal Biochem 207(2):298–303.
https://doi.org/10.1111/j.1365-2818.1990. https://doi.org/10.1016/0003-2697(92)
tb02948.x 90015-y
2. Porichis F, Hart MG, Griesbeck M, Everett 4. Patterson BK, Till M, Otto P, Goolsby C, Fur-
HL, Hassan M, Baxter AE, Lindqvist M, Miller tado MR, McBride LJ, Wolinsky SM (1993)
SM, Soghoian DZ, Kavanagh DG, Reynolds S, Detection of HIV-1 DNA and messenger
Norris B, Mordecai SK, Nguyen Q, Lai C, RNA in individual cells by PCR-driven in situ
Kaufmann DE (2014) High-throughput hybridization and flow cytometry. Science
detection of miRNAs and gene-specific 260(5110):976–979. https://doi.org/10.
mRNA at the single-cell level by flow cytome- 1126/science.8493534
try. Nat Commun 5:5641. https://doi.org/ 5. Belloc F, Durrieu F (1994) Detection of
10.1038/ncomms6641 mRNA species by flow cytometry. Methods
Highly Multiplexed and Simultaneous Characterization of Protein and RNA in. . . 165
Abstract
Comprehensive genome-wide analyses of single cells represent an important tool for clinical applications,
such as pre-implantation diagnostic and prenatal diagnosis, as well as for cancer research purpose. For the
latter, studies of tumor heterogeneity, circulating tumor cells (CTCs), and disseminated cancer cells
(DCCs) require the analysis of single-cell genomes. Here we describe a reliable and robust array-based
comparative genomic hybridization (aCGH) protocol based on Ampli 1™ whole genome amplification
that allows the detection of copy number alterations (CNAs) in single cancer cells as small as 100 kb.
Key words aCGH, Single cells, PCR labeling, Array hybridization, CNAs
1 Introduction
Miodrag Gužvić (ed.), Single Cell Analysis: Methods and Protocols, Methods in Molecular Biology, vol. 2752,
https://doi.org/10.1007/978-1-0716-3621-3_11, © Springer Science+Business Media, LLC, part of Springer Nature 2024
167
168 Giancarlo Feliciello et al.
2 Materials
2.1 Reagents In this protocol high-quality reagents are necessary to execute the
aCGH procedure. Utilization of the materials included in the kits,
reagents, or instrumentations indicated maximizes the likelihood of
a successful experiment. If using third-party materials, the user is
recommended to prepare all solutions using ultrapure water
(HPLC purified, DNAse-free) and PCR analytical grade reagents.
Store all reagents and enzymes according to the guidelines
specified by the manufacturer. Reagent expired or subjected to
improper storage may compromise the outcome of the aCGH
experiment. Always store the reagents separately from the DNA
samples and avoid repeated freeze-thaw cycles of solutions contain-
ing DNA or enzymes. When preparing frozen reagent solutions
thaw completely, mix briefly on a vortex mixer and spin in a micro-
centrifuge to collect the contents off the wall and lid, and pre-chill
to 4 °C or on ice until use.
2.2 Laboratory Setup To assure optimal condition for the aCGH, it is recommended to
perform all steps under a dedicated vertical laminar flow hood.
Make sure that all the pipetting aids are calibrated and handled
properly. Always wear powder-free laboratory gloves and use dedi-
cated solutions and pipettes with nuclease-free aerosol-resistant tips
to avoid contamination of reagents with nucleases and cross-
contaminations of samples. Maintain a clean working area by wip-
ing surfaces with Ethanol 80% or bleaching solutions (e.g., DNA-
Zap™ or DNA AWAY*) and by UV-irradiation of PCR bench, tips,
tubes, and tube racks.
Array-Based Comparative Genomic Hybridization for the Detection of Copy. . . 169
2.3 Safety Notes Cyanine reagents are considered hazardous and may be harmful if
swallowed. Avoid contact with eyes, skin, and clothing.
2× HI-RPM Hybridization Buffers are considered hazardous
and may be harmful if swallowed. Avoid contact with eyes, skin, and
clothing.
Acetonitrile is considered toxic to blood, kidneys, liver, lungs,
gastrointestinal tract, central nervous system, skin, eyes, and upper
respiratory tract. Avoid contact with eyes, skin, and clothing.
Always refer to the Material Safety Data Sheet (MSDS) of your
specific country.
Carefully follow all waste disposal regulations when disposing
waste materials.
2.4 DNA Templates aCGH application uses a “two-color” process to measure DNA
copy number alterations in a Test Sample relative to a Reference
Sample.
1. Test Sample: The chapter describes the recommendations to
label and hybridize amplified DNA from tumor cells, in partic-
ular single DCCs and CTCs, but its spectrum of application is
wider and may include other applications where the amount of
input DNA material is very limited. Therefore, the method is
also well suited for the analysis of cell pools (≥2 cells), speci-
mens extracted from FFPE tissue, as well as cells or DNA
material collected with the use of various isolation procedures
(e.g., micromanipulators mounted on microscopes, the
DEParray™ System). A fundamental prerequisite to the use
of this protocol is the amplification with Ampli 1™ WGA Kit
that has been already described in detail in this book series
[17]. Moreover, aCGH of single cells demands for samples
with high-quality amplified DNA. Always assess the Genomic
Integrity Index (GII) of the samples before proceeding to
expensive molecular assays [18]. The use of DNA samples
that do not fulfill these quality criteria may result in the failure
of the labeling reactions or low quality of the aCGH results
(high background noise) that may impair the reliable detection
of copy number alterations.
2. Reference Sample: A pool of peripheral blood lymphocytes
isolated from a healthy male/female donor and amplified with
Ampli 1™ WGA Kit. Sex-matched or sex-mismatched controls
can be used in different settings. If the gender of the sample
donor is known, selection of reference DNA is based on the
gender of the donor of the test sample. Sex-matched designs
are more commonly used when the technology is well estab-
lished and may offer better sensitivity for detecting gains or
losses of regions on the X and Y chromosomes.
170 Giancarlo Feliciello et al.
2.5 Fluorescent 1. Expand Long Template PCR System (Roche) containing: 10×
Labeling of Amplified Expand Long Template Buffer 1, 2, and 3 and Expand Long
DNA Template Enzyme Mix (5 U/μL).
2. Bovine Serum Albumin from bovine serum, 20 mg/mL
(Roche).
3. Fluorescent-labeled nucleotides 25 nmol each
(GE-Healthcare): Cy3-dCTP and Cy3-dUTP for the Reference
Sample, Cy5-dCTP and Cy5-dUTP for the Test Sample.
4. 9/10 mixes Deoxynucleotide triphosphate-dNTPs 100 mM
each (VWR International): 10 μL dATP, 10 μL dGTP, 9 μL
dCTP, 9 μL dTTP, and 62 μL of H2O.
5. Lib1 primer 5′- TAGTGGGATTCCTGCTGTCAGT -3′
(20 μM): 20 μL of Lib1 (100 μM) and 80 μL of H2O.
3 Methods
3.1 PCR-Based Purpose In this step Test and Reference samples are differentially
Labeling Using labeled with dUTPs and dCTPs nucleotides conjugated to different
Incorporation of Dye- Cyanine dyes, Cy5 and Cy3 that have a red and green fluorescence,
Conjugated dNTPs and respectively. Cycle after cycle, the polymerase will incorporate the
Tru1I Digestion fluorescent nucleotides into the newly synthetized DNA strand
generating amplicons marked with two different colors. The result-
ing PCR products are then subjected to digestion with Tru1I
restriction endonuclease to cleave off the PCR-adaptors prior to
the aCGH hybridization, thereby avoiding unspecific cross-
hybridization of Test sample with Reference sample and oligonucle-
otide probes on the array.
Duration
1. Hands-on time: 45 min.
2. PCR program: 1 h 25 min.
3. Digestion program: 3 h.
172 Giancarlo Feliciello et al.
DNA Labeling
Hands-on: 30 min Hybridization
PCR: 1h Hands-on: 30 min
Denaturation : 3 min
Reagents
Renaturation: 30 min
DNA
Incubation: 24 h
Expand Long Template PCR
System Reagents
Cy5 dUTP/dCTP Cot1 DNA
Cy3 dUTP/dCTP Oligo aCGH/ChIP-on –chip
Lib1 Primer Hybridization kit
9/10 dNTPs Mix Tween20
BSA Igepal
Nuclease-free Water Output
Output DNA Hybridized on array
Cy5 and Cy3 labeled DNA
Fig. 1 Overview of the single cell aCGH workflow described in this chapter
Array-Based Comparative Genomic Hybridization for the Detection of Copy. . . 173
Table 1
Cy5/Cy3 labeling master mix
Table 2
PCR program for DNA labeling
Table 3
DNA digestion mix
Reagent Volume
Buffer R (10×) 5.6 μL
Tru1I 1.0 μL
Table 4
Thermal profile of DNA digestion
Temperature Time
65 °C 3h
4 °C 1
3.2 Purification of Purpose In this step the excess labeled nucleotides that have not
the PCR/Digestion been incorporated, as well as PCR and digestion reagents, are
Product and removed by column purification. Additionally, the cleaved
Measurement of the PCR-adaptors generated after digestion are also discarded in the
DNA Yield and purification process. Only when the samples are purified the mea-
Incorporation Rate surement can take place on the Nanodrop instrument.
Duration
1. Hands-on time: 30 min.
2. Purification: 30 min.
3. Quantification: 10 min.
Procedure
1. Assemble the Amicon Ultra 0.5 column as described by the
manufacturer.
2. Pool the resulting products of the same sample and fill up the
volume to 480 μL with H2O. Centrifuge at 14,000 RCF for
10 min at room temperature.
Array-Based Comparative Genomic Hybridization for the Detection of Copy. . . 175
3.3 Hybridization Purpose During this step the labeled genomic DNA from the Test
and the Reference samples are mixed in equal amounts and
co-hybridized to an array containing immobilized 60-mers of
nucleotides. Cot-1 DNA is used to reduce the cross-hybridization
of repetitive DNA sequences to the array preventing the nonspecific
binding of a labeled probe of interest to the repetitive DNA
sequence (see Note 7).
176 Giancarlo Feliciello et al.
Table 5
Hybridization mix
Reagent Volume
Cot1-DNA (1 μg/μL) 5.0 μL
Agilent blocking reagent (10×) 12.0 μL
HI-RPM hybridization buffer (2×) 60.0 μL
Tween20–25% (v/v) 5.2 μL
Igepal – 25% 5.2 μL
Total 87.4 μL
Duration
1. Hands-on time: 15–20 min (see Note 8).
2. Processing time of the Thermomixer 1: 3 min.
3. Processing time of the Thermomixer 2: 30 min.
4. Processing time of the Agilent Microarray Oven: 24 h.
Procedure
. Step 1: Denaturation /Renaturation of the hybridization
solution.
1. Turn on Thermomixer 1 and 2 and set the following para-
meters: 95 °C for 3 min at 350 rpm and 37 °C for 30 min at
350 rpm, respectively.
2. Turn on the Agilent Microarray Oven and set the temperature
to 65 °C (see Note 9).
3. Thaw the necessary reagents and the desired amount of Test
and Reference samples at room temperature (see Note 10).
4. Assemble the Hybridization Mix as indicated in Table 5 (see
Note 11).
5. In a new labeled 1.5 mL Eppendorf tube, mix Test and Refer-
ence samples (19 μL each) (see Note 12).
6. Add 87.4 μL of the Hybridization Mix into the premixed
samples for a total of 125.4 μL.
7. Mix gently by pipetting slowly up and down (see Note 11) and
quickly spin to drive contents to the bottom of the
reaction tube.
8. Denature the samples in Thermomixer 1: transfer the samples
and incubate at 95 °C for 3 min at 350 rpm (see Note 13).
9. Renature the samples in Thermomixer 2: transfer the samples
swiftly from Thermomixer 1 into Thermomixer 2 and incubate
at 37 °C for 30 min at 350 rpm (see Note 14).
Array-Based Comparative Genomic Hybridization for the Detection of Copy. . . 177
10. Remove the samples from the Thermomixer 2 and spin 1 min
at 6000 RCF in a minicentrifuge.
11. Samples are ready and must be hybridized immediately. Keep
the samples at 37 °C until the Hybridization starts (see Note
15).
3.4 Washing Purpose Washing steps are meant to remove the excess of unbound
probes or probes loosely attached to non-complementary DNA
while leaving the matched probe target intact. Additionally, they
clean the corner spots of the main grid – necessary to position
correctly the grid template – from contaminating materials (i.e.,
dust or dirty particles).
Duration
1. Hands-on time: 5 min.
2. Disassembly in Agilent Oligo aCGH/ChIP-on-Chip Wash
Buffer 1: 1 min.
3. Agilent Oligo aCGH/ChIP-on-Chip Wash Buffer 1, wash step
1: 2 min 30 s.
4. Agilent Oligo aCGH/ChIP-on-Chip Wash Buffer 1, wash step
2: 2 min 30 s.
5. Agilent Oligo aCGH/ChIP-on-Chip Wash Buffer 2, wash step
1: 30 s.
6. Agilent Oligo aCGH/ChIP-on-Chip Wash Buffer 2, wash step
2: 30 s.
7. Acetonitrile wash: 5 s.
Procedure
. Step 1: Disassembly of the Agilent Hybridization Assembly.
2. Transfer the aCGH slide into a new slide-staining jar filled with
Agilent Oligo aCGH/ChIP-on-Chip Wash Buffer 1 and wash
on the horizontal oscillator at room temperature for 2 min and
30 s, 120 rpm (see Note 30).
3. Invert the aCGH slide and place it back again in the same slide-
staining jar, wash on the horizontal oscillator at room temper-
ature for 2 min and 30 s, 120 rpm (see Note 31).
4. Shortly before the end of the wash prepare the slide-staining jar
and the Agilent Oligo aCGH/ChIP-on-Chip Wash Buffer
2 that have been equilibrated overnight at 37 °C.
5. Transfer the aCGH slide into the slide-staining jar filled with
Agilent Oligo aCGH/ChIP-on-Chip Wash Buffer 2 and wash
on the horizontal oscillator at 37 °C for 30 s, 120 rpm (see
Note 27).
6. Invert the aCGH slide and place it back again in the same slide-
staining jar, wash on the horizontal oscillator at 37 °C for 30 s,
120 rpm.
7. Dip the aCGH slide shortly (3–5 s) into the slide-staining dish
filled with Acetonitrile.
8. Using a KimTech tissue, wipe gently the remaining drops on
the side of the slide (see Note 32).
9. Place the aCGH slide in the Slide Holder with the “Agilent”
side of the slide facing up.
10. Place the Ozone barrier slide on top of the aCGH slide (see
Note 33).
11. Close the Slide Holder and push on the tab end until you hear
it click.
12. The aCGH slide is now ready for scanning (see Note 34).
3.5 Scanning and Purpose The aCGH slides are scanned into image files using a
Extraction specific microarray scanner. During this operation the spot inten-
sities for the two colors are measured and a .tif image, necessary for
the data extraction, is generated. In the Feature extraction software,
the fluorescence data are translated into log ratio, allowing the
identification of aberrations in the Test Sample.
Duration
1. Hands-on time: 5 min.
2. Scanning time: Depending on the number of aCGH slides
analyzed.
3. Extraction time: Depending on the number of aCGH slides
analyzed.
180 Giancarlo Feliciello et al.
Procedure
. Step 1: Scanning.
4 Notes
33. The second stage where ozone can cause a problem occurs
when the slide is being scanned. The increased air circulation
at the open end of the slide holder (left side) causes more
ozone to contact the Cy5 thereby inducing a visible degrada-
tion gradient. To prevent degradation during scanning we
recommend using a barrier slide, which sits over the microar-
ray. This seals the array in a small chamber and limits the extent
of ozone degradation.
34. Scanning the slides immediately after this step is highly recom-
mended to minimize the reduction in the fluorescence signal
intensity due to environmental oxidants and possible photo-
bleaching. In case immediate scanning is not possible keep the
slides in black light-sealed boxes (you can use the boxes the
array slides come in) or slide-holding boxes wrapped in
aluminum foil.
References
1. Pinkel D, Segraves R, Sudar D, Clark S, disseminated cancer cell--the unit of selection.
Poole I, Kowbel D, Collins C, Kuo WL, Adv Cancer Res 89:35–67
Chen C, Zhai Y, Dairkee SH, Ljung BM, 7. Riethdorf S, Wikman H, Pantel K (2008)
Gray JW, Albertson DG (1998) High resolu- Review: biological relevance of disseminated
tion analysis of DNA copy number variation tumor cells in cancer patients. Int J Cancer
using comparative genomic hybridization to 123(9):1991–2006. https://doi.org/10.
microarrays. Nat Genet 20(2):207–211. 1002/ijc.23825
https://doi.org/10.1038/2524 8. Cohen SJ, Punt CJ, Iannotti N, Saidman BH,
2. Solinas-Toldo S, Lampel S, Stilgenbauer S, Sabbath KD, Gabrail NY, Picus J, Morse M,
Nickolenko J, Benner A, Dohner H, Mitchell E, Miller MC, Doyle GV, Tissing H,
Cremer T, Lichter P (1997) Matrix-based Terstappen LW, Meropol NJ (2008) Relation-
comparative genomic hybridization: biochips ship of circulating tumor cells to tumor
to screen for genomic imbalances. Genes Chro- response, progression-free survival, and overall
mosomes Cancer 20(4):399–407 survival in patients with metastatic colorectal
3. Fiorentino F (2012) Array comparative geno- cancer. J Clin Oncol 26(19):3213–3221.
mic hybridization: its role in preimplantation https://doi.org/10.1200/JCO.2007.15.
genetic diagnosis. Curr Opin Obstet Gynecol 8923
24(4):203–209. https://doi.org/10.1097/ 9. Klein CA, Schmidt-Kittler O, Schardt JA,
GCO.0b013e328355854d Pantel K, Speicher MR, Riethmuller G (1999)
4. Munne S (2012) Preimplantation genetic diag- Comparative genomic hybridization, loss of
nosis for aneuploidy and translocations using heterozygosity, and DNA sequence analysis of
array comparative genomic hybridization. Curr single cells. Proc Natl Acad Sci U S A 96(8):
Genomics 13(6):463–470. https://doi.org/ 4494–4499
10.2174/138920212802510457 10. Greshock J, Feng B, Nogueira C, Ivanova E,
5. Fiorentino F, Caiazzo F, Napolitano S, Perna I, Nathanson K, Protopopov A, Weber
Spizzichino L, Bono S, Sessa M, Nuccitelli A, BL, Chin L (2007) A comparison of DNA copy
Biricik A, Gordon A, Rizzo G, Baldi M (2011) number profiling platforms. Cancer Res
Introducing array comparative genomic hybri- 67(21):10173–10180. https://doi.org/10.
dization into routine prenatal diagnosis prac- 1158/0008-5472.CAN-07-2102
tice: a prospective study on over 1000 11. Haraksingh RR, Abyzov A, Gerstein M, Urban
consecutive clinical cases. Prenat Diagn AE, Snyder M (2011) Genome-wide mapping
31(13):1270–1282. https://doi.org/10. of copy number variation in humans: compara-
1002/pd.2884 tive analysis of high resolution array platforms.
6. Klein CA (2003) The systemic progression of PLoS One 6(11):e27859. https://doi.org/10.
human cancer: a focus on the individual 1371/journal.pone.0027859
Array-Based Comparative Genomic Hybridization for the Detection of Copy. . . 187
Abstract
Micro RNAs represent important post-transcriptional regulators in health and are involved in the onset of
many diseases. Therefore, the further characterization of physiological miRNA functions is an important
basic research question, and miRNAs even have high potential as biomarkers both for prognosis and
diagnosis. In order to exploit this potential, it is mandatory to accurately quantify the miRNA expression
not only in bulk but also on the single-cell level. Here, we describe a protocol, which facilitates miRNA
sequencing library preparation of very low input samples, single cells, and even clinical samples such as
circulating tumor cells. The protocol can be combined with different single-cell isolation methods (e.g.,
micromanipulation and FACS sorting). After cell lysis, sequencing adapters are ligated to the miRNAs,
other ncRNA species, and adapter dimers are reduced by exonuclease digest, the miRNA library is reverse
transcribed, amplified, and purified. Furthermore, quality controls are described to select only high-quality
samples for sequencing.
Key words miRNA, small RNA, ncRNA, Single-cell sequencing, NGS, CTC
1 Introduction
Miodrag Gužvić (ed.), Single Cell Analysis: Methods and Protocols, Methods in Molecular Biology, vol. 2752,
https://doi.org/10.1007/978-1-0716-3621-3_12, © Springer Science+Business Media, LLC, part of Springer Nature 2024
189
190 Sarah M. Hücker and Stefan Kirsch
2 Materials
2.2 Reagents If not stated differentially, the reagents are stored at -20 °C and
thawed on ice.
1. 1% Triton X-100 (Sigma-Aldrich) in nuclease-free water, store
at RT.
miRNA Sequencing Library Preparation 191
rRNA
2. 5.8S rRNA removal
block
5‘ ad 3‘ ad
6. Reverse transcription
cDNA
i5 i7
i5 i7
7. PCR amplification
Table 1
Names and sequences of the required oligonucleotides
3 Methods
3.2 Single Cell 1. Harvest cells and resuspend cell pellet in PBS (see Notes 3 and
Isolation 4).
2. Pick single cells by micromanipulation (see Notes 5 and 6).
3. Aspirate single cell into 1 μL PBS, pipet into lysis buffer, and
centrifuge down.
4. Store the tubes with the isolated cell on dried ice while isolating
additional single cells.
5. Long-term storage of isolated cells in lysis buffer is possible at
-80 °C.
Table 2
Program of the first PCR
Table 3
Program of the indexing PCR
21. Mix indexing PCR mix, pipet 23.5 μL into a fresh 0.2 mL
reaction tube, and spin down.
22. Mix the sample after first PCR (step 19) and pipet 1 μL each
into the indexing PCR mix.
23. Pipet 0.5 μL of 100 μM Index RPI oligo into the indexing PCR
mix. Use a different Index RPI oligo for every sample (see
Note 9).
24. Mix sample, spin down, and perform the PCR program shown
in Table 3 in a thermo cycler.
3.5 Library Quality 1. Determine the dsDNA concentration using the Qubit™ 1×
Control dsDNA HS Assay Kit: Prepare standards (10 μL standard +
190 μL Qubit working solution) and samples (1 μL sam-
ple + 199 μL Qubit working solution), vortex, incubate for
2 min in the dark, and measure dsDNA concentration
(Note 14).
2. If the DNA concentration is higher than 2 ng/μL, dilute 1 μL
sample to 2 ng/μL with water. If the DNA concentration is
lower than 2 ng/μL, just use 1 μL undiluted sample.
3. Determine the fragment length distribution on a Bioanalyzer
using a High Sensitivity DNA Kit. Pipet 1 μL of (diluted)
sample on the chip (see Notes 15–17).
4 Notes
Fig. 2 Example Bioanalyzer profiles of three miRNA libraries. (a) This library shows the expected fragment
length distribution with the main product at about 150 bp. (b) This library contains a large amount of adapter
dimers visible by the main product at about 120 bp. (c) The main product is larger than 150 bp indicating
contamination by larger RNA classes
References
1. Cai Y, Yu X, Hu S, Yu J (2009) A brief review 6. Fuchs RT, Sun Z, Zhuang F, Robb GB (2015)
on the mechanisms of miRNA regulation. Bias in ligation-based small RNA sequencing
Genomics Proteomics Bioinformatics 7(4): library construction is determined by adaptor
147–154. https://doi.org/10.1016/S1672- and RNA structure. PLoS One 10(5):
0229(08)60044-3 e0126049. https://doi.org/10.1371/journal.
2. Ludwig N, Leidinger P, Becker K, Backes C, pone.0126049
Fehlmann T, Pallasch C, Rheinheimer S, 7. Giraldez MD, Spengler RM, Etheridge A,
Meder B, Stahler C, Meese E, Keller A (2016) Godoy PM, Barczak AJ, Srinivasan S, De Hoff
Distribution of miRNA expression across PL, Tanriverdi K, Courtright A, Lu S,
human tissues. Nucleic Acids Res 44(8): Khoory J, Rubio R, Baxter D, Driedonks
3865–3877. https://doi.org/10.1093/nar/ TAP, Buermans HPJ, Nolte-’t Hoen ENM,
gkw116 Jiang H, Wang K, Ghiran I, Wang YE, Van
3. Di Leva G, Garofalo M, Croce CM (2014) Keuren-Jensen K, Freedman JE, Woodruff
MicroRNAs in cancer. Annu Rev Pathol 9: PG, Laurent LC, Erle DJ, Galas DJ, Tewari M
287–314. https://doi.org/10.1146/annurev- (2018) Comprehensive multi-center assess-
pathol-012513-104715 ment of small RNA-seq methods for quantita-
4. Horita M, Farquharson C, Stephen LA (2021) tive miRNA profiling. Nat Biotechnol 36(8):
The role of miR-29 family in disease. J Cell 746–757. https://doi.org/10.1038/nbt.
Biochem 122(7):696–715. https://doi.org/ 4183
10.1002/jcb.29896 8. Wright C, Rajpurohit A, Burke EE, Williams C,
5. Baran-Gale J, Kurtz CL, Erdos MR, Sison C, Collado-Torres L, Kimos M, Brandon NJ,
Young A, Fannin EE, Chines PS, Sethupathy P Cross AJ, Jaffe AE, Weinberger DR, Shin JH
(2015) Addressing bias in small RNA library (2019) Comprehensive assessment of multiple
preparation for sequencing: a new protocol biases in small RNA sequencing reveals signifi-
recovers microRNAs that evade capture by cur- cant differences in the performance of widely
rent methods. Front Genet 6:352. https://doi. used methods. BMC Genomics 20(1):513.
org/10.3389/fgene.2015.00352 https://doi.org/10.1186/s12864-019-
5870-3
miRNA Sequencing Library Preparation 199
Abstract
In this new era of precision medicine, characterization of single-cell subpopulations to better understand
disease etiology is paramount. It is thus an opportune time to explore techniques that allow molecular
analysis of single cells and to better understand the basis of pathogenesis of diseases like cancer. Single-cell
western blotting is one such method that allows analysis of single cells at the protein level. In contrast to
traditional western blotting, which relies heavily on bulk analysis of lysates generated from tissues and is
often indicative of the population average, this technique allows analysis of lysates from single-cell sub-
populations thereby providing a glimpse into cell heterogeneity. The method entails the use of a chip
containing 30 μm thick photoactivated polyacrylamide gel spotted with nearly 6400 microwells. Single cells
loaded on the chip are captured in the microwells by passive gravity and are then lysed and electrophoresed
using the MILO™ single-cell western platform. This method forgoes the use of transfer of proteins on a
PVDF and a nitrocellulose membrane, as performed in traditional western blotting, and all other steps
including probing of primary and fluorescent secondary antibodies against the protein of interest are
performed directly on the chip. The proteins of interest can then be visualized by scanning a chip with
the use of a microarray scanner. The entire procedure can be performed in as less as 4–6 h, and thus this
method provides several advantages over traditional western blotting.
Key words Single-cell western blot, Immunoblot, Precision medicine, Tumor heterogeneity, Cell
heterogeneity
1 Introduction
Miodrag Gužvić (ed.), Single Cell Analysis: Methods and Protocols, Methods in Molecular Biology, vol. 2752,
https://doi.org/10.1007/978-1-0716-3621-3_13, © Springer Science+Business Media, LLC, part of Springer Nature 2024
201
202 Prashant V. Thakkar
Fig. 1 Schematic of single-cell western blot protocol. Steps a–e highlight different steps in the single-cell
western blot protocol. (a) The scWest chip is divided into 16 different grids (displayed over two rows and eight
columns). (b) Each grid contains 400 microwells thus giving us a total of 6400 microwells per chip. (c) The
chip is rehydrated with 1× Suspension buffer and a single-cell suspension (approximately 100,000 cells) is
loaded on the cells. (d) The cells are allowed to settle and nearly 30% of the total microwells are occupied with
cells. The excess number of cells is then washed off. (e) The cells in the microwells are then lysed and
electrophoresed in the same step using the MILO™ single-cell western platform. The chip is washed twice
with 1× wash buffer followed by one rinse with deionized water. (f) The chip is then probed with primary
antibodies against the protein of interest followed by fluorescent secondary antibody. (g) The chip is then
scanned using a microarray scanner and the resulting image can be analyzed using the Scout software.
(Figure adapted from [12])
2 Materials
3 Methods
3.1 Preparation of 1. The 10× suspension buffer and 5× wash buffer are provided
Buffers and Reagents with the standard scWest kit.
2. Prepare 1× suspension buffer and 1× wash buffer using the
above-mentioned 10× and 5× stock solutions respectively
using deionized water or molecular grade water [10].
3.2 Priming and 1. Take a scWest chip and place it in the center of a clean 10 cm
Rehydrating petri dish with the gel-side facing up. It is important that the
scWest Chips dish is clean and sterile since any dust particles that settle on the
chip can lead to a high background during the imaging steps.
Touching the gel surface of the chip must be avoided since that
can add to more dust particles on the chip.
2. Add 10 mL of 1× suspension buffer to the petri dish to
completely cover the chip.
3. Incubate the chip for at least 10 min, with slow shaking
(<100 rpm) on an orbital shaker before use, so that the chip
is completely rehydrated. Alternatively, the chip can also simply
be allowed to stand in 1× suspension buffer, although, it is
important to make sure that the chip is completely immersed.
4. The chip can be kept in the 1× suspension buffer until the
single-cell suspension is ready to be loaded on the chip.
4. Gently tap the flask to detach all the cells and neutralize the
trypsin with at least equal volume of growth medium contain-
ing 10% FBS.
5. Count cells using hemocytometer or other cell counting equip-
ment and prepare a 5 mL suspension of 10,000 to 100,000
cells/mL in growth medium with 1× suspension buffer.
6. Centrifuge cells at 300 × g for 5 min. Remove the growth
medium and wash the cells once with 5 mL of 1× PBS (without
MgCl2 and CaCl2).
7. Centrifuge cells at 300 × g for 5 min. Remove 1× PBS solution
and resuspend cells in 5 mL of 1× suspension buffer. The
single-cell suspension is now ready for use on the single-cell
western blot.
3.4 Loading scWest 1. Remove the scWest chip from the petri dish containing 1×
Chips with Single-Cell suspension buffer and tilt it on one side to remove excess
Suspension liquid. A paper tissue can be placed underneath to help drain
off the excess liquid ensuring that paper tissue does not touch
the top of the gel. Place the chip in a new and clean 10 cm petri
dish. For selecting appropriate scWest chips, see Note 1.
2. Load 1 mL (approximately 100,000 cells) of the single-cell
suspension dropwise on the scWest chip [10]. Ensure that all
areas of the chip are fully covered (Fig. 1c). For using lower
number of cells, see Note 2.
3. Allow the cells to settle for 5–20 min. This step allows the cells
to enter and settle in the wells (Fig. 1d). For optimizing
appropriate settling time for different cells, see Note 3.
4. Once the cells are allowed to settle for the desired time, tilt the
slide to drain off the liquid and remove the uncaptured cells
from the top of the slide. To efficiently remove all the cells from
the top of the chip, tilt the petri dish containing the chip at a
45° angle and carefully run the 1× suspension buffer from top
to bottom using a 1000 uL pipette.
5. At this step, the chip can be viewed under a bright field micro-
scope to visualize settling of the cells in microwells. Immerse
the slide back into the 10 cm petri dish containing 1× suspen-
sion buffer. This chip is now ready to be run on MILO.
3.5 Running scWest 1. Turn on the MILO instrument and use the touch screen to
Chip Using MILO™ create optimized user settings. Most typically the conditions
Instrument listed below can serve as a good reference point [10]. Further
optimization may be required for determining optimal
conditions.
Lysis time: 10–15 s.
Electrophoresis run time: 60–90 s.
206 Prashant V. Thakkar
scWest chip
Trough
Fig. 2 Methodology to load the chip in the MILO™ instrument to ensure efficient lysis: 300 μL of lysis buffer is
loaded on the shorter end of the trough in the electrophoresis. Tilt the chip at a 45° angle and align the shorter
end of the chip to the shorter end of the trough where lysis buffer was added. Gradually lay the chip from the
other end so that the chip now lays flat and perfectly aligned in the trough. This will cause the lysis buffer to
spread throughout the bottom surface of the chip
3.6 Probing scWest 1. Dilute the primary antibody to the final concentration of
Chips with Primary 0.2 μg/μL to 0.05 μg/μL using the “antibody diluent 2”
and Fluorescent solution provided in the kit [10].
Secondary Antibodies The protocol is designed to use as little as a total volume of
80 μL of the antibody at a dilution of 1:5 to 1:20, that is, for a 1:
20 dilution add 4 μL of the antibody with 76 μL of the “antibody
diluent 2” solution. Most commercially available antibodies
should work just fine within this concentration range. This con-
centration range can serve as a good reference point, although
further optimization may be required. Primary antibodies of two
different species can also be multiplexed in the same step. Thus, to
make an antibody solution of two different primary antibodies at
1:20 dilution each, add 4 μL of each of the antibodies with 72 μL
of the antibody diluent. Alternatively, the antibodies against two
different proteins of interest, even though from two different
species, can also be probed sequentially.
2. A special probing chamber (available through ProteinSimple)
[10] is used for antibody incubation. Pipette 80 μL of antibody
solution on one end of the probing chamber and place the chip
with gel side facing down so that the antibody solution spreads
evenly and covers the entire gel surface of the chip. To ensure
that the antibody covers the entire gel surface, the chip should
be held steadily on one end where the antibody solution was
loaded and gradually lowering the chip from the other end
(Fig. 3).
3. Avoid bubbles since no antibody signal will be detected in the
spot of the bubble. Adjust the chip to eliminate the bubbles
if any.
4. Incubate the chip with primary antibody at room temperature
for 1 h.
208 Prashant V. Thakkar
scWest chip
Probing chamber
80 ml antibody
solution loaded on
one end the
probing chamber
Fig. 3 Methodology to load the chip on antibody probing chamber while probing
it with primary and fluorescent secondary antibodies: Pipette 80 μL of secondary
antibody solution on one end of the probing chamber and chip is placed with gel
side facing down so that the antibody solution spreads evenly and covers the
entire gel surface of the chip. The chip should be held steadily on one end where
the antibody solution was loaded and gradually lowering the chip from the
other end
5. At the end of 1-h incubation, carefully lift the chip from one
end while holding the other end so as not to tear the gel. Invert
it with the gel side facing up in a 10 cm petri dish.
6. Wash it three times with 1× wash buffer, while gently shaking
on an orbital shaker for 10 min each.
7. At the end of the third wash, prepare the fluorescent secondary
antibody solution using the “Antibody diluent” at 1:20 to 1:40
dilution [10].
Similar to primary antibody incubation, secondary antibo-
dies against primary antibodies from two different species can be
multiplexed. It is important that the two secondary antibodies
against the two different primary antibodies fluoresce in differ-
ent spectral channels to ensure specificity of the signal.
8. For secondary antibody incubation, pipette 80 μL of secondary
antibody solution on one end of the probing chamber and
place the chip with gel side facing down so that the antibody
solution spreads evenly and covers the entire gel surface of the
chip. To ensure that the antibody covers the entire gel surface,
the chip should be lowered slowly keeping one end steadily
rested on the probing chamber and gradually lowering the chip
from the other end (Fig. 3).
9. Incubate the secondary antibody solution for 1 h, this time
covering it with either a black box or aluminum foil so as to
protect it from light.
Single-Cell Western Blot 209
10. At the end of the 1-h incubation, carefully lift the chip from
one end while holding the other end so as not to tear the gel.
Invert it to with the gel side facing up in a 10 cm petri dish.
11. Wash it three times with 1× wash buffer, while gently shaking
on an orbital shaker for 15 min each this time also protecting
from light [10].
12. After the last wash, proceed to incubating primary antibody for
the next protein interest. If all the proteins of interest have
been probed with their respective primary and secondary anti-
bodies, air dry the chip in the dark for about 40–45 min.
13. The chip is now ready for imaging.
3.7 Imaging of 1. scWest chip can be scanned using many of the recommended
scWest Chips fluorescence microarray scanners recommended by Protein-
Simple [10]. In case where the microarray scanner is not avail-
able, see Note 5 for the use of Zeiss spinning disc microscope
for scanning images.
2. We have personally used GenePix4000B from Molecular
Devices [11], which in our experience is very user-friendly to
use. However, this instrument can only allow for scanning of
up to two different fluorescent channels (excitation wave-
lengths of 532 nm and 635 nm with emission wavelengths in
greenish yellow to red/far red regions of the spectrum)
whereas other microarray scanners can allow up to four or
even five different channels thus allowing for probing of more
proteins per chip.
3. To start scanning the slide using the GenePix4000B, open the
door and lift the slide holder. Push the slide clamp to place the
slide in its position with gel side facing down. Lower the slide
holder back to its original position and close the door.
4. Close the instrument door and start the “Genepix pro image
analysis software” on the computer attached to the instrument.
5. Once the software interface is open, click on “Hardware set-
tings” to select the wavelength the chip would be scanned
at. For each wavelength, set the scan pixel resolution to 5 μm
and adjust the PMT gain to maximize the brightness of the
signal without saturating the detector. Change the power of
each laser to 100%.
6. Once all the settings are set, one can hit preview scan to get a
quick preview of image the scan would generate. If the preview
scan looks good, one can then hit “Data Scan” for scanning the
chip using both wavelengths of “Single wavelength scan”
option to scan the chip only using one wavelength.
7. The instrument allows simultaneous scans at two wavelengths
and scans the standard scWest chip (approximately
210 Prashant V. Thakkar
3.8 Data Analysis 1. The images generated using microarray scanners for any given
Using Scout Software chip are saved as separate file per channel. These images are
saved in .tiff format, and this format can be easily used for
analysis using Scout software. Export/Add all the images for
a given chip from “File” menu in the Scout software [10].
2. The software requires that each exported image be registered.
To do this, select the first block of the first row and the eight
block in the second row (i.e., two diagonally opposite grids).
Then click on the first well in the first block and the last well in
the last block. This step helps ensure that the image alignment
is correct and any misalignment can be corrected [10].
3. Following step 2, the software will then automatically detect
the lanes with a positive signal/peaks. This detection can be
then manually inspected, and these parameters can be adjusted
to define the true signal under “Scan properties” table [10].
4. Once all the peaks are detected, go to the peak table to identify
and name the peak of interest as the appropriate molecular
weight size peak.
5. The software can then provide statistics such as total number of
microwells occupied, total number of wells that are positive in
each spectral channel, etc. A representative image of the results
obtained from this single-cell western blot method is presented
in Fig. 4.
3.9 Limitations Although this technique shows incredible promise and is huge
advancement in allowing molecular analysis of proteins at the level
of single cells, it is not without limitations. Some of these limita-
tions are highlighted below:
1. The method currently does not incorporate the use of molecu-
lar weight markers that can be run simultaneously with the
sample. This limitation then requires one to probe for one of
the housekeeping genes such as Tubulin or GAPDH to be used
as a size marker. One can then predict the size of the protein of
interest based on the size and migration of the housekeeping
gene. In addition, since one has to always probe for a
housekeeping gene, it expends one of the fluorescent channels,
leaving one less channel that can be used for probing an addi-
tional protein of interest.
2. Although migration of protein occurs over a short distance of
900 μm, and while the migration of proteins of different
molecular sizes follows a linear range in that short distance,
Single-Cell Western Blot 211
4 Notes
microwells with more than one cell per microwell, that is,
doublets and triplets per microwell [10].
4. Use of reducing agents to completely denature proteins
The lysis buffer provided in the kit currently does not
contain any reducing agents. Furthermore, since the lysis step
is performed directly on the chip once cells are loaded in the
microwells, there is no boiling step, which may result in incom-
plete denaturation of proteins as well as incomplete dissocia-
tion of oligomers. Reducing agents can be easily incorporated
in the protocol by using 1× SDS buffer for lysis of cells in the
microwells. Although this buffer is highly ionic, the conduc-
tance of 1× SDS buffer is higher compared to the lysis buffer
provided in the kit. Hence, the voltage should be reduced from
240 V to 150 V and run time should be increased by approxi-
mately 50–60%. Alternatively, 10 mM TCEP (tris-(2-carbox-
yethyl)phosphine) (pH 7.0) can be added to the lysis buffer
provided to the kit. This is important since the increased con-
ductance by the lysis buffer can lead to heating up of the gel,
which may in turn affect the resolution.
5. Use of Zeiss spinning disc microscope for scanning scWest chips
In cases where microarray scanners are hard to find or are
not available in one’s institution, we have also successfully
employed Zeiss spinning disc microscope where one can scan
the entire chip in “tiles” and then “stitch” all the images to
create a single image of the entire chip. This image can then be
exported to the Scout software for further analysis.
References
1. Ashley EA, Butte AJ, Wheeler MT, Chen R, Bhattacharjee A, Eichler EE, Bamshad M,
Klein TE, Dewey FE, Dudley JT, Ormond Nickerson DA, Shendure J (2009) Targeted
KE, Pavlovic A, Morgan AA, Pushkarev D, capture and massively parallel sequencing of
Neff NF, Hudgins L, Gong L, Hodges LM, 12 human exomes. Nature 461(7261):
Berlin DS, Thorn CF, Sangkuhl K, Hebert JM, 2 7 2 – 2 7 6 . h t t p s : // d o i . o r g / 1 0 . 1 0 3 8 /
Woon M, Sagreiya H, Whaley R, Knowles JW, nature08250
Chou MF, Thakuria JV, Rosenbaum AM, Zar- 4. Ashley EA (2016) Towards precision medicine.
anek AW, Church GM, Greely HT, Quake SR, Nature reviews. Genetics 17(9):507–522.
Altman RB (2010) Clinical assessment incor- https://doi.org/10.1038/nrg.2016.86
porating a personal genome. Lancet 5. McGranahan N, Swanton C (2017) Clonal
375(9725):1525–1535. https://doi.org/10. heterogeneity and tumor evolution: past, pres-
1016/S0140-6736(10)60452-7 ent, and the future. Cell 168(4):613–628.
2. Choi M, Scholl UI, Ji W, Liu T, Tikhonova IR, https://doi.org/10.1016/j.cell.2017.01.018
Zumbo P, Nayir A, Bakkaloglu A, Ozen S, 6. Ignatiadis M, Dawson SJ (2014) Circulating
Sanjad S, Nelson-Williams C, Farhi A, tumor cells and circulating tumor DNA for
Mane S, Lifton RP (2009) Genetic diagnosis precision medicine: dream or reality? Ann
by whole exome capture and massively parallel Oncol 25(12):2304–2313. https://doi.org/
DNA sequencing. Proc Natl Acad Sci U S A 10.1093/annonc/mdu480
106(45):19096–19101. https://doi.org/10.
1073/pnas.0910672106 7. Yuan GC, Cai L, Elowitz M, Enver T, Fan G,
Guo G, Irizarry R, Kharchenko P, Kim J,
3. Ng SB, Turner EH, Robertson PD, Flygare Orkin S, Quackenbush J, Saadatpour A,
SD, Bigham AW, Lee C, Shaffer T, Wong M, Schroeder T, Shivdasani R, Tirosh I (2017)
214 Prashant V. Thakkar
Challenges and emerging directions in single- 10. MILO™ user guide from ProteinSimple.
cell analysis. Genome Biol 18(1):84. https:// https://www.proteinsimple.com/documents/
doi.org/10.1186/s13059-017-1218-y Milo_User_Guide_R5.pdf
8. Zhu S, Qing T, Zheng Y, Jin L, Shi L (2017) 11. Genepix4000B Microarray scanner user guide.
Advances in single-cell RNA sequencing and its http://mdc.custhelp.com/euf/assets/con
applications in cancer research. Oncotarget tent/GenePix_4000B_UserGuide.pdf
8(32):53763–53779. https://doi.org/10. 12. Hughes AJ, Spelke DP, Xu Z, Kang CC, Schaf-
18632/oncotarget.17893 fer DV, Herr AE (2014) Single-cell western
9. Haque A, Engel J, Teichmann SA, Lonnberg T blotting. Nat Methods 11(7):749–755.
(2017) A practical guide to single-cell RNA-- https://doi.org/10.1038/nmeth.2992
sequencing for biomedical research and clinical
applications. Genome Med 9(1):75. https://
doi.org/10.1186/s13073-017-0467-4
Chapter 14
Abstract
Laser ablation inductively coupled plasma-mass spectrometry (LA-ICP-MS) is a well-established and
sensitive analytical technique, which provides high-resolution imaging of endogenous elements, element
tagged-markers, metal-containing nanoparticles, and metallodrugs within cells. Here we describe a proto-
col for imaging the subcellular distribution of platinum within A549 cells, following their incubation with
the platinum-based anticancer agent, Oxaliplatin. We outline the essential steps in sample preparation and
instrumental setup and discuss how the current generation of low-dispersion instruments facilitates new
approaches to data acquisition and image processing. The protocol described herein can be easily adapted
for other cell lines and metal-containing labeling agents.
1 Introduction
Miodrag Gužvić (ed.), Single Cell Analysis: Methods and Protocols, Methods in Molecular Biology, vol. 2752,
https://doi.org/10.1007/978-1-0716-3621-3_14, © Springer Science+Business Media, LLC, part of Springer Nature 2024
215
216 Amy J. Managh and Calum J. Greenhalgh
the protein distribution, DNA content, cell volume, and cell cycle
phase of individual cells [6, 7]. LA-ICP-MS has also been used to
determine nanoparticle and drug uptake by cells. Studies have
reported both quantitative determination of Au nanoparticle
uptake by whole cells [8] and subcellular imaging of their localiza-
tion to different cellular compartments [9]. Furthermore, the tech-
nique may be vital for monitoring the fate of rare cell populations
used in cell-based treatments. In a 2013 study, individual regu-
latory T cells that were labeled with the MRI contrast agent Omnis-
can were successfully identified in mouse peritoneal lavage samples
at 10 days post-administration [10]. Subsequent publications have
also reported LA-ICP-MS identification of individual therapeutic
cells within ex vivo tissue sections in similar cell tracking experi-
ments [11, 12].
Despite the success of the above applications, LA-ICP-MS has
not yet gained full traction within the molecular biology commu-
nity. This is mainly due to inefficiencies associated with the previous
generation of instruments, in particular in the interface between the
LA and ICP-MS units, which resulted in broadening of signals and
hence lengthy analysis times compared to conventional imaging
methods. In recent years a number of new interfaces have been
developed to combat this problem [13–15]. These new technolo-
gies provide vastly improved aerosol transport characteristics, lead-
ing to faster signal responses, higher absolute sensitivity, and
improved signal-to-noise ratios. Importantly, these new develop-
ments are now starting to reach the market. In this protocol we
describe a method for qualitative single-cell analysis and imaging of
platinum distribution in Oxaliplatin spiked A549 cells, using one of
these new laser ablation platforms. We focus on how the improved
aerosol transport characteristics have created an opportunity to
move away from the traditional “rastered” method of imaging,
toward an approach that preserves maximum image resolution for
individual cell analysis.
2 Materials
3 Methods
3.1 Preparation of Cell culture and labeling conditions will vary depending on the
Cells for Analysis nature of the cell line being studied. The instructions below are
based on the culture of A549 cells, which are an adherent human
lung adenocarcinoma cell line, and their labeling with the antican-
cer drug, Oxaliplatin. For other cell lines and labeling approaches,
you should follow your usual procedures for that cell line, and then
proceed to step 9.
1. Seed cells at an initial density of 1 × 106 cells in 5 mL of
complete media. Incubate at 37 °C in 5% CO2 overnight.
2. Add 50 μM Oxaliplatin to the culture media and leave to
incubate for a further 24 h.
3. Aspirate the used media. Add 5 mL PBS to the flask (see Note
2) and gently swirl for 1 min ensuring the entire surface is
covered. Aspirate the PBS into a waste container. Repeat 2–4
times (see Note 3).
4. Add 1 mL of warm trypsin-EDTA solution to the flask and
incubate at 37 °C for 3 min.
5. Inspect the flask under a microscope to check that the cells have
successfully detached from the surface (see Note 4), then add
4 mL of complete media to quench the trypsin.
6. Set aside a 100 μL aliquot of the cell suspension for cell
counting.
7. Transfer the remaining cells into a 15 mL falcon tube and
centrifuge at 400 rpm for 5 min.
218 Amy J. Managh and Calum J. Greenhalgh
3.2 Setting Up the 1. Insert the slide into the laser ablation chamber and secure it in
LA-ICP-MS System place (see Note 8). Ensure that the chamber is appropriately
sealed afterwards by fastening the screws on the outside of the
chamber (finger-tight).
2. Connect the inlet of the laser ablation chamber to a helium
supply and connect the outlet of the chamber to the ICP-MS
torch, using suitable tubing, e.g., Tygon. Allow for the intro-
duction of an argon make-up flow between the ablation cham-
ber and the ICP torch (see Note 9).
3. If available, connect a trigger cable between SyncOut on the
laser ablation system and the external trigger in port on the
mass spectrometer. This is to synchronize the firing of the laser
with the acquisition cycle of the mass spectrometer.
4. Purge the chamber with helium for around 10 min by selecting
the purge button in the laser software. While the instrument is
purging it is useful to move the inner cup around the chamber
to aid removal of residual air from the corners of the chamber.
This can be done using the x,y controls.
5. Light the plasma (see Note 10). Once the plasma has lit set the
helium flow through the laser ablation chamber to around
1.4 L/min. It is useful to do this at a low ramp rate of around
20 mL/min in order to avoid extinguishing the plasma. Allow
the plasma conditions to stabilize for approximately an hour
before performing any analysis.
6. A sample to cup distance of 0.2–0.3 mm is optimal for efficient
material transport out of the chamber. To set this use the z
LA-ICP-MS of Single Cells 219
Fig. 1 Left image: Microscopic image showing A549 cells that were dosed with 50 μM Oxaliplatin. The
Cytospun cells are spaced an adequate distance for single-cell targeting. Right: LA-ICP-MS signal for the
analysis of the Pt in the cell on the far left. Data points in raw counts (shown as horizontal lines) are spaced at
1 ms intervals allowing the full peak profile to be visualized
3.3 LA-ICP-MS Here, analyzing the platinum content of whole cells is used to
Analysis of Pt Content qualitatively assess the heterogeneity in Oxaliplatin uptake across
in Whole Cells the cell population.
1. In the LA software select the spot tool, and then left-click on
the center of one of the cells of interest.
2. Right-click the above spot and amend its properties. To ensure
the entire cell is ablated, set a spot size that is at least 5–10 μm
larger than the diameter of cells of interest (for this analysis a
spot size of 60 μm was used). Set the repetition frequency as
220 Amy J. Managh and Calum J. Greenhalgh
3.4 LA-ICP-MS Here, subcellular imaging is used to qualitatively assess the distri-
Imaging of Pt bution of platinum within individual cells, in order to assess
Distribution Across whether the drug is reaching its target site of action (binding to
Cells DNA in the nucleus).
1. In the LA software, use the line tool to draw a single horizontal
or vertical line that is long enough to span the region of interest
at its widest point. If necessary, this line can be subsequently
resized or repositioned to the edge of the region of interest,
using the Edit Endpoints function.
2. Right-click the above line and amend its properties. Choose a
laser beam diameter that is smaller than the dimensions of the
cells of interest (in this work a 2 μm spot size was used). Set the
highest possible repetition frequency that enables baseline res-
olution between the peaks from consecutive ablation events
(in this work 20 Hz was used). Select a scan rate that is
LA-ICP-MS of Single Cells 221
3.5 Generation of LA- 1. The data output from the imaging experiment will consist of a
ICP-MS Images series of files, each file containing the data for one ablated line.
Assuming sufficient instrument sensitivity, the data will consist
of peaks separated by background (or close to background)
222 Amy J. Managh and Calum J. Greenhalgh
Fig. 2 Schematic depiction of the data processing approach required for a pixel-by-pixel imaging experiment.
The instrumental conditions are set such that every laser shot samples an adjacent, non-overlapping area and
the signal is allowed to wash out before the next location is sampled. In this example, data points in raw
counts (shown as horizontal lines) are spaced at 10 ms intervals in the ICP-MS method, allowing 5 data points
to be collected per laser shot (laser at 20 Hz). Markers are then placed between each laser shot and the data
between the markers is summed to give a value for each pixel in the final image
Fig. 3 Left image: Microscopic image of a clump of A549 cells, which were dosed with 50 μM Oxaliplatin.
Right: LA-ICP-MS image showing the distribution of Pt across the cells. Cells were imaged using a 2 μm laser
beam, with a repetition frequency of 20 Hz and a scan speed of 40 μm/s. It appears that Pt is preferentially
accumulated in the nucleus of these cells
4 Notes
15. The peaks can be manually located within the Excel data or this
can be automated through the use of in-house Excel macros or
Matlab scripts. We use a custom app, the LA-ICP-MS Image
Tool, to find the peaks in our LA-ICP-MS data [16]. Once the
peak areas have been found, the %RSD of the data can be
calculated to provide information on the variability in label
uptake across the cell population. We find that plotting a
histogram of the integrated signals is a useful way of visually
displaying this information in publications and conference pre-
sentations. For an example histogram, see reference [10].
16. Some modern systems have updates that allow the option of
sending a trigger pulse before every laser shot. This approach
may simplify your data processing. It is advisable to speak to
your relevant LA and ICP-MS service engineers to discuss
whether this is a possible option for your instrumentation.
References
1. Giesen C, Wang HA, Schapiro D, Zivanovic N, ablation inductively coupled plasma mass spec-
Jacobs A, Hattendorf B, Schuffler PJ, trometry. J Anal Atom Spectrom 26(11):
Grolimund D, Buhmann JM, Brandt S, 2160–2165. https://doi.org/10.1039/
Varga Z, Wild PJ, Gunther D, Bodenmiller B c1ja10227c
(2014) Highly multiplexed imaging of tumor 6. Herrmann AJ, Techritz S, Jakubowski N,
tissues with subcellular resolution by mass Haase A, Luch A, Panne U, Mueller L (2017)
cytometry. Nat Methods 11(4):417–422. A simple metal staining procedure for identifi-
https://doi.org/10.1038/nmeth.2869 cation and visualization of single cells by
2. Theiner S, Van Malderen SJM, Van Acker T, LA-ICP-MS. Analyst 142(10):1703–1710.
Legin A, Keppler BK, Vanhaecke F, Koellen- https://doi.org/10.1039/c6an02638a
sperger G (2017) Fast high-resolution laser 7. Mueller L, Herrmann AJ, Techritz S, Panne U,
ablation-inductively coupled plasma mass spec- Jakubowski N (2017) Quantitative characteri-
trometry imaging of the distribution of zation of single cells by use of immunocyto-
platinum-based anticancer compounds in mul- chemistry combined with multiplex
ticellular tumor spheroids. Anal Chem 89(23): LA-ICP-MS. Anal Bioanal Chem 409(14):
12641–12645. https://doi.org/10.1021/acs. 3667–3676. https://doi.org/10.1007/
analchem.7b02681 s00216-017-0310-1
3. de Vega RG, Fernandez-Sanchez ML, 8. Wang M, Zheng LN, Wang B, Chen HQ, Zhao
Pisonero J, Eiro N, Vizoso FJ, Sanz-Medel A YL, Chai ZF, Reid HJ, Sharp BL, Feng WY
(2017) Quantitative bioimaging of Ca, Fe, Cu (2014) Quantitative analysis of gold nanopar-
and Zn in breast cancer tissues by ticles in single cells by laser ablation inductively
LA-ICP-MS. J Anal Atom Spectrom 32(3): coupled plasma-mass spectrometry. Anal Chem
6 7 1 – 6 7 7 . h t t p s : // d o i . o r g / 1 0 . 1 0 3 9 / 86(20):10252–10256. https://doi.org/10.
c6ja00390g 1021/ac502438n
4. Willmes M, Glessner JJG, Carleton SA, Gerrity 9. Drescher D, Giesen C, Traub H, Panne U,
PC, Hobbs JA (2016) Sr-87/Sr-86 isotope Kneipp J, Jakubowski N (2012) Quantitative
ratio analysis by laser ablation MC-ICP-MS in imaging of gold and silver nanoparticles in sin-
scales, spines, and fin rays as a nonlethal alter- gle eukaryotic cells by laser ablation ICP-MS.
native to otoliths for reconstructing fish life Anal Chem 84(22):9684–9688. https://doi.
history. Can J Fish Aquat Sci 73(12): org/10.1021/ac302639c
1852–1860. https://doi.org/10.1139/cjfas- 10. Managh AJ, Edwards SL, Bushell A, Wood KJ,
2016-0103 Geissler EK, Hutchinson JA, Hutchinson RW,
5. Giesen C, Waentig L, Mairinger T, Drescher D, Reid HJ, Sharp BL (2013) Single cell tracking
Kneipp J, Roos PH, Panne U, Jakubowski N of gadolinium labeled CD4(+) T cells by laser
(2011) Iodine as an elemental marker for imag- ablation inductively coupled plasma mass
ing of single cells and tissue sections by laser
226 Amy J. Managh and Calum J. Greenhalgh
Abstract
Cells exhibit diverse morphologic phenotypes, biophysical and functional properties, and gene expression
patterns. Understanding how these features are interrelated at the level of single cells has been challenging
due to the lack of techniques for multimodal profiling of individual cells. We recently developed Patch-seq,
a technique that combines whole-cell patch clamp recording, immunohistochemistry, and single-cell
RNA-sequencing (scRNA-seq) to comprehensively profile single cells. Here we present a detailed step-
by-step protocol for obtaining high-quality morphological, electrophysiological, and transcriptomic data
from single cells. Patch-seq enables researchers to explore the rich, multidimensional phenotypic variability
among cells and to directly correlate gene expression with phenotype at the level of single cells.
1 Introduction
Miodrag Gužvić (ed.), Single Cell Analysis: Methods and Protocols, Methods in Molecular Biology, vol. 2752,
https://doi.org/10.1007/978-1-0716-3621-3_15, © Springer Science+Business Media, LLC, part of Springer Nature 2024
227
228 Cathryn R. Cadwell and Andreas S. Tolias
2 Materials
Fig. 1 Schematic of Patch-seq technique. (Adapted from [10] with permission from Springer)
Fig. 2 Custom equipment used in Patch-seq. (a) 1 mL syringe with 0.2 μm syringe filter and tapered pipette
tip, used for backfilling glass pipettes. (b) Patch pipette marked at approximately 0.3 μL volume. (c) Positive
pressure device used to eject pipette contents into PCR tube after aspirating cell contents. (d) Staining
chamber used for immunohistochemistry. (Reproduced from [10] with permission from Springer)
3 Methods
3.1 Electro- 1. Prepare all work surfaces and equipment that may come in
physiology and Single- contact with RNase-free solutions by cleaning with RNase
Cell RNA Sample Zap, followed by 70% (vol/vol) ethanol (see Note 3). Any
Collection equipment that may have come into contact with post-PCR
products (tube holders, pipettes) should also be cleaned thor-
oughly with DNA-OFF before RNase Zap. Items can be
cleaned the day before the experiment.
2. Thaw an aliquot of RNase-free internal solution stock, and add
recombinant RNase inhibitor to a final concentration of 1 U/μ
L and vortex well. Addition of RNase inhibitor should increase
the osmolarity of the internal solution to the desired range of
315–320 mOsm. Adjust the osmolarity of the ACSF to be
18–20 mOsm lower than the internal solution, adding small
amounts of sucrose as needed to increase the osmolarity of the
ACSF (see Note 4).
234 Cathryn R. Cadwell and Andreas S. Tolias
Fig. 3 Ideal patching approach for aspiration of cell contents. (a) The target cell is approached from the side
rather than the top to provide the most direct path for cell contents to enter the pipette. As the cell slowly
swells, it may be helpful to move the pipette back a few microns to release the tension on the membrane and
prevent current leakage. During aspiration of the cell contents, the cell is continuously monitored to ensure
that no extracellular contents are entering the pipette. Following aspiration, the pipette is slowly pulled away
from the cell membrane, leaving the shrunken cell body intact in the tissue slice to preserve axodendritic
morphology. (b) Examples of collapse of the cell body after aspiration of cell contents, visualized using
two-photon imaging (2PI). Scale bars, 10 μm. (c) Examples of collapse of the cell body after aspiration of cell
contents, visualized using differential interference contrast (DIC) imaging. Scale bar, 10 μm. (Reproduced from
[10] with permission from Springer)
3.2 cDNA Library 1. Remove any remaining bubbles by gently tapping the side of
Preparation and each tube. Spin down the samples at 700 g for 10 s at 4 °C and
Sequencing place back on ice.
2. Denature the RNA by incubating the samples in a thermal
cycler for 3 min at 72 °C.
3. Add 5.7 μL of RT mix to each tube and pipette up and down
four times to mix. Gently tap each tube as needed to remove
bubbles and spin down at 700 g for 10 s at 4 °C. Immediately
return to ice.
4. Incubate the samples in a thermal cycler according to the
following program: step 1: 42 °C for 90 min; step 2: 50 °C
for 2 min; step 3: 42 °C for 2 min (repeat steps 2–3 ten times);
step 4: 70 °C for 10 min; step 5: Hold at 4 °C (see Note 13).
5. Add 15 μL of PCR mix to each tube, vortex to mix, and spin
down at 700 g for 10 s at 4 °C.
6. Incubate the samples in a thermal cycler according to the
following program: step 1: 98 °C for 3 min; step 2: 98 °C
for 20 s; step 3: 67 °C for 15 s; step 4: 72 °C for 6 min (repeat
steps 2–4 eighteen times); step 5: 72 °C for 5 min; step 6:
Hold at 4 °C (see Notes 14 and 15).
7. Allow the Axygen AxyPrep mag PCR beads to come to room
temperature for 15 min. Vortex well.
8. Add 17.5 μL of beads to each sample and mix thoroughly by
pipetting up and down at least 10 times.
9. Transfer samples to a 96-well plate, cover with lid, and incubate
for 8 min at room temperature.
10. Place the 96-well plate on the magnetic stand and incubate for
an additional 5 min.
11. Carefully remove the clear solution without disturbing the
beads.
12. Wash the beads two times with 80% ethanol for 30 s each time.
Carefully remove the ethanol without touching the beads after
each wash.
13. Cover the plate with a lid and leave at room temperature until
the beads are completely dry and small cracks appear on the
surface (typically 5–10 min).
14. Take the plate off the magnetic stand, add 17.5 μL of Buffer EB
to each well, and pipette up and down ~10 times to resuspend
the beads.
15. Incubate the plate off the magnetic stand for 2 min.
16. Place the plate back on the magnetic stand and incubate for an
additional 2–3 min. Without touching the beads, collect 15 μL
Patch-seq of Single Cells 237
Fig. 4 Quality check of full-length single-cell cDNA libraries. Bioanalyzer profiles of single-cell amplified cDNA
from several cells from a typical experiment (a) showing variable cDNA yield, as well as a negative control
from the same experiment that consisted of ERCC spike-in RNA only. Profiles from degraded (b) or
contaminated (c) samples are also shown. The low molecular weight fragments in (c) were traced back to
bacterial contamination of a reagent. (Reproduced from [10] with permission from Springer)
of the clear liquid from each well into a new 0.2 mL PCR tube
(see Note 16).
17. Check the concentration and size distribution of the full-
length cDNA library on an Agilent Bioanalyzer using the
Agilent High Sensitivity DNA Kit according to the manufac-
turer’s instructions (Fig. 4; see Note 17).
18. Dilute each sample to 50 pg/μL.
19. Add 4 μL of tagmentation mix to 6 μL of each sample.
Vortex well.
238 Cathryn R. Cadwell and Andreas S. Tolias
Fig. 6 Post-sequencing quality control. The quality of the samples can be assessed by analyzing the
sequencing depth (a), number of detected genes (b), the maximum spearman correlation between each cell
and all other cells (c), and fraction of reads mapped to exons, introns, and intergenic segments (d). For this
data set approximately 10% of the samples were discarded post-sequencing based on pre-determined quality
control criteria (dashed lines in (a) and (b) represent the cutoffs below which samples were discarded). (e)
Histogram of mean normalized expression across cells for all genes in the example dataset. Genes with a
mean expression of less than one (dashed line) can be excluded to minimize the impact of gene dropout on
downstream analyses. (Reproduced from [10] with permission from Springer)
3.3 Immunohisto- 1. After 10–14 days of fixation, transfer the tissue slices to a
chemistry and staining chamber with mesh-bottom wells (Fig. 1d, see Note
Morphological 20).
Recovery 2. Carefully remove any trace of PFA and wash the slices three
times with 0.01 M PBS. Cover and leave on the rotator for
10 min at room temperature during each wash.
3. Remove the PBS and submerge slices in the 3% (vol/vol)
H2O2. Cover and leave on the rotator at room temperature
for 30 min.
4. Remove the 3% (vol/vol) H2O2 and wash the slices three times
with 0.01 M PBS. Cover and leave on the rotator for 10 min at
room temperature during each wash.
5. Remove the PBS and add the A/B mix. Cover and store
overnight at 4 °C.
6. Remove the A/B mix and wash the slices four times with
0.01 M PBS. Cover and leave on the rotator for 10 min at
room temperature during each wash.
7. Remove the PBS and add the DAB mix. Leave on the rotator at
room temperature for 7–8 min.
8. Remove the DAB mix and wash the slices three times with
0.01 M PBS. Cover and leave on the rotator for 10 min at
room temperature during each wash.
9. Mount the slices on microscope slides in Mowiol mounting
medium and cover with a glass coverslip (see Note 21).
10. Visualize cell morphology under a light microscope (Fig. 7, see
Note 22).
240 Cathryn R. Cadwell and Andreas S. Tolias
Fig. 7 Direct morphological recovery of Patch-seq cells. Examples of two excitatory (a, c) and two inhibitory
(b, d) neurons with successful characterization of morphologic and electrophysiological profiles as well as
acquisition of full-length cDNA. Scale bars for histochemical staining, 50 μm. Scale bars for electrophysiologi-
cal traces, 100 ms and 50 mV/500 pA. (Adapted from [10] with permission from Springer)
4 Notes
10. Be cautious to apply the suction gently to avoid losing the seal.
Use a large (20–30 mL), new syringe that is partially with-
drawn, and ensure smooth contact between the rubber piston
and syringe wall. If the cell is lost (i.e., there is a sudden increase
in the amount of current required to hold the cell at -70 mV),
immediately discontinue suction and remove the pipette. The
RNA may still be salvageable, but endogenous RNases from
the extracellular space will degrade it quickly. If a substantial
amount of extracellular material has entered the pipette, the
sample should be discarded.
11. If attempting to preserve cell morphology, first move the
pipette back slowly from the cell and allow the membrane to
slowly retract from the pipette before removing the pipette
from the tissue.
12. The fixed tissue slice can be stored at 4 °C for 10–14 days
before proceeding with immunohistochemistry.
13. Samples can be left in the PCR machine overnight while run-
ning the RT reaction.
14. PCR Products can be stored at -20 or -80 °C for up to
6 months before proceeding.
15. Amplified cDNA should be handled and stored in a separate
post-PCR area away from the pre-PCR samples to prevent
cross-contamination of new samples with amplified cDNA.
16. Purified full-length cDNA can be stored at -20 or -80 °C for
up to 6 months before proceeding with tagmentation and
sequencing.
17. The full-length cDNA should have a broad peak at
1500–2000 bp and very few fragments <500 bp.
18. The mean length of fragments in the final sequencing library
should be <500 bp.
19. A complete overview of possible analytical approaches is
beyond the scope of this chapter. In addition to the repertoire
of scRNA-seq analytical tools, additional resources specific to
analyzing Patch-seq data sets include [15–17].
20. If a specialized staining chamber is not available, staining can
be performed in a 24-well plate by changing the solution in
each well separately.
21. The microscope slides can be stored indefinitely at room tem-
perature, protected from light.
22. Staining quality may continue to improve for 48–72 h after
mounting on slides.
Patch-seq of Single Cells 243
References
1. Picelli S, Bjorklund AK, Faridani OR, 10. Cadwell CR, Scala F, Li S, Livrizzi G, Shen S,
Sagasser S, Winberg G, Sandberg R (2013) Sandberg R, Jiang X, Tolias AS (2017) Multi-
Smart-seq2 for sensitive full-length transcrip- modal profiling of single-cell morphology,
tome profiling in single cells. Nat Methods electrophysiology, and gene expression using
10(11):1096–1098. https://doi.org/10. Patch-seq. Nat Protoc 12(12):2531–2553.
1038/nmeth.2639 https://doi.org/10.1038/nprot.2017.120
2. Tang F, Barbacioru C, Wang Y, Nordman E, 11. Scala F, Kobak D, Bernabucci M, Bernaerts Y,
Lee C, Xu N, Wang X, Bodeau J, Tuch BB, Cadwell CR, Castro JR, Hartmanis L, Jiang X,
Siddiqui A, Lao K, Surani MA (2009) mRNA- Laturnus S, Miranda E, Mulherkar S, Tan ZH,
Seq whole-transcriptome analysis of a single Yao Z, Zeng H, Sandberg R, Berens P, Tolias
cell. Nat Methods 6(5):377–382. https://doi. AS (2021) Phenotypic variation of transcrip-
org/10.1038/nmeth.1315 tomic cell types in mouse motor cortex. Nature
3. Eberwine J, Yeh H, Miyashiro K, Cao Y, Nair S, 598(7879):144–150. https://doi.org/10.
Finnell R, Zettel M, Coleman P (1992) Analy- 1038/s41586-020-2907-3
sis of gene expression in single live neurons. 12. Scala F, Kobak D, Shan S, Bernaerts Y,
Proc Natl Acad Sci U S A 89(7):3010–3014. Laturnus S, Cadwell CR, Hartmanis L,
https://doi.org/10.1073/pnas.89.7.3010 Froudarakis E, Castro JR, Tan ZH,
4. Sucher NJ, Deitcher DL (1995) PCR and Papadopoulos S, Patel SS, Sandberg R,
patch-clamp analysis of single neurons. Neuron Berens P, Jiang X, Tolias AS (2019) Layer 4 of
14(6):1095–1100. https://doi.org/10.1016/ mouse neocortex differs in cell types and circuit
0896-6273(95)90257-0 organization between sensory areas. Nat Com-
5. Sucher NJ, Deitcher DL, Baro DJ, Warrick mun 10(1):4174. https://doi.org/10.1038/
RM, Guenther E (2000) Genes and channels: s41467-019-12058-z
patch/voltage-clamp analysis and single-cell 13. Picelli S, Bjorklund AK, Reinius B, Sagasser S,
RT-PCR. Cell Tissue Res 302(3):295–307. Winberg G, Sandberg R (2014) Tn5 transpo-
https://doi.org/10.1007/s004410000289 sase and tagmentation procedures for massively
6. Subkhankulova T, Yano K, Robinson HP, Live- scaled sequencing projects. Genome Res
sey FJ (2010) Grouping and classifying 24(12):2033–2040. https://doi.org/10.
electrophysiologically-defined classes of neo- 1101/gr.177881.114
cortical neurons by single cell, whole-genome 14. Jiang X, Shen S, Cadwell CR, Berens P, Sinz F,
expression profiling. Front Mol Neurosci 3:10. Ecker AS, Patel S, Tolias AS (2015) Principles
https://doi.org/10.3389/fnmol.2010.00010 of connectivity among morphologically
7. Qiu S, Luo S, Evgrafov O, Li R, Schroth GP, defined cell types in adult neocortex. Science
Levitt P, Knowles JA, Wang K (2012) Single- 350(6264):aac9462. https://doi.org/10.
neuron RNA-Seq: technical feasibility and 1126/science.aac9462
reproducibility. Front Genet 3:124. https:// 15. Kobak D, Berens P (2019) The art of using
doi.org/10.3389/fgene.2012.00124 t-SNE for single-cell transcriptomics. Nat
8. Cadwell CR, Palasantza A, Jiang X, Berens P, Commun 10(1):5416. https://doi.org/10.
Deng Q, Yilmaz M, Reimer J, Shen S, 1038/s41467-019-13056-x
Bethge M, Tolias KF, Sandberg R, Tolias AS 16. Kobak D, Bernaerts Y, Weis MA, Scala F, Tolias
(2016) Electrophysiological, transcriptomic AS, Berens P (2021) Sparse reduced-rank
and morphologic profiling of single neurons regression for exploratory visualisation of
using patch-seq. Nat Biotechnol 34(2): paired multivariate data. J R Stat Soc Ser C
199–203. https://doi.org/10.1038/nbt. Appl Stat 70(4):980–1000. https://doi.org/
3445 10.1111/rssc.12494
9. Fuzik J, Zeisel A, Mate Z, Calvigioni D, 17. Tripathy SJ, Toker L, Bomkamp C, Mancarci
Yanagawa Y, Szabo G, Linnarsson S, Harkany BO, Belmadani M, Pavlidis P (2018) Assessing
T (2016) Integration of electrophysiological transcriptome quality in patch-seq datasets.
recordings with single-cell RNA-seq data iden- Front Mol Neurosci 11:363. https://doi.org/
tifies neuronal subtypes. Nat Biotechnol 34(2): 10.3389/fnmol.2018.00363
175–183. https://doi.org/10.1038/nbt.
3443
Chapter 16
Abstract
During the last decade a wide range of single-cell and single-nucleus next-generation sequencing techni-
ques have been developed, which revolutionized detection of rare cell populations, enabling creation of
comprehensive cell atlases of complex organs and tissues. State-of-the-art methods do not only allow
classical transcriptomics of individual cells but also comprise a number of epigenetic approaches, including
assessment of chromatin accessibility by single-nucleus Assay for Transposase Accessible Chromatin ATAC-
seq (snATAC-seq). The snATAC-seq assay detects “open chromatin,” a term for low nucleosome occu-
pancy of genomic regions, which is a prerequisite for effective transcription factor binding. Information
about open chromatin at the single-nucleus level helps to recognize epigenetic changes, sometimes before
transcription of respective genes occurs. snATAC-seq detects cellular heterogeneity in otherwise still
transcriptionally and/or morphologically homogeneous cell populations. Chromatin accessibility assays
may be used to detect epigenetic changes in cardiac lineages during heart development, chromatin
landscape changes during aging, and epigenetic alterations in heart diseases. Here, we provide an optimized
protocol for snATAC-seq of murine hearts. We describe isolation of single nuclei from snap-frozen hearts,
provide hints for preparation of libraries suitable for snATAC-seq next-generation sequencing (NGS) using
the Chromium 10× platform, and give general recommendations for downstream analysis using conven-
tional bioinformatic pipelines and packages. The protocol should serve as a beginner’s guide to generate
high-quality snATAC-seq datasets and to perform chromatin accessibility analysis of individual heart-
derived cell nuclei.
Key words Heart, Single-cell, Single-nucleus ATAC-seq, Transposase, Sequencing, Chromatin acces-
sibility, NGS, Chromium 10x™
1 Introduction
Miodrag Gužvić (ed.), Single Cell Analysis: Methods and Protocols, Methods in Molecular Biology, vol. 2752,
https://doi.org/10.1007/978-1-0716-3621-3_16, © Springer Science+Business Media, LLC, part of Springer Nature 2024
245
246 Michail Yekelchyk et al.
2 Materials
See Table 1 for the list of suggested equipment, and Table 2 for the
list of necessary chemicals, reagents, and enzymes.
snATAC-seq of Individual Heart Cells 247
Table 1
Laboratory equipment
Equipment Manufacturer
GentleMACS dissociator, M tubes Miltenyi Biotec, Germany
40 μm cell strainer No preference
Centrifuge (for 15 mL conical tubes and for 1.5 mL tubes) No preference
15 mL conical tubes, 1.5 mL tubes, 0.2 mL tubes No preference
Fluorescent-activated cell sorting (FACS) machine No preference
Chromium Controller & Next GEM Accessory Kit 120223 or 1000202, 10xGenomics, USA
Polymerase Chain Reaction (PCR) thermocycler No preference
Magnetic separator for SPRI bead cleanup No preference
Qubit measurement device and HS DNA kit Q32851, Thermo, USA
Capillary electrophoresis instrument Agilent, USA or PerkinElmer, USA
Next Generation Sequencer Illumina, USA
Table 2
Chemicals and enzymes
Final concentration in
Chemicals and enzymes the protocol Manufacturer
Dulbecco’s phosphate-buffered saline (DPBS) – Gibco / Thermo Fisher
Scientific, USA
Anti-PCM-1 antibody 1:1000 HPA023374, Sigma-
Aldrich, Germany
Anti-rabbit secondary antibody conjugated to 1:1000 A11011, Life
Alexa 594 Technologies
Chromium Next GEM Chip H Single Cell Kit – 1000162, 10xGenomics,
USA
Chromium Next GEM Single Cell ATAC – 1000176, 10xGenomics,
Library & Gel Bead Kit v1.1 USA
Single Index Kit N Set A – 1000212, 10xGenomics,
USA
Nuclease-free water – Elut
Elution Buffer (EB) – Qiagen, Netherlands
Solid Phase Reversible Immobilization (SPRI) – AC-60050, MagBio,
magnetic beads Switzerland
High-75 NextSeq500 cartridge – 20024906, Illumina, USA
248 Michail Yekelchyk et al.
Buffer Recipes
1. Lysis buffer: 5 mM CaCl2, 3 mM Mg(CH3COO)2, 2 mM
EDTA, 0.5 mM Ethylene glycol-bis(β-aminoethyl ether)-N,
N,N′,N′-tetraacetic acid (EGTA), and 10 mM Tris-HCl
(pH 8).
2. Triton buffer I: 0.4% Triton X-100 solution: 40 μL Triton
X-100 in 10 mL PBS.
3. Triton buffer II: 0.2% Triton X-100 solution: 20 μL Triton
X-100 in 10 mL PBS.
4. Sucrose buffer: 1 M sucrose, 3 mM Mg(CH3COO)2, 10 mM
Tris-HCl (pH 8).
5. Staining buffer: DPBS, 1% BSA, 0.2% Igepal CA-630,
1 mM EDTA.
6. DAPI staining solution: 2 ng/mL DAPI in nuclease-free water.
7. Tagmentation Mix: prepared in accordance with the Next
GEM scATAC-seq kit manual (contains ATAC Buffer B
(PN2000193) and ATAC Enzyme (PN2000123 or
PN2000138)).
8. Barcoding Mastermix: prepared in accordance with the Next
GEM scATAC-seq kit manual (contains scATAC Gel Beads
v.1.1 (PN2000210), Reducing Agent B (PN2000087), Bar-
coding Reagent B (PN2000194), Barcoding Enzyme
(PN2000125 or PN2000139), Partitioning Oil
(PN2000190), and 10× controller accessory parts as per
manual).
9. Elution solution: prepared in accordance with the Next GEM
scATAC-seq kit manual (contains Buffer EB, 0.1% Tween
20 (10 μL Tween 20 in 10 mL PBS), and Reducing Agent B
(PN2000087)).
3 Methods
3.1 A Brief Protocol The following protocol describes isolation of nuclei from murine
for Cardiac Nuclei hearts and optional identification of nuclei with PCM1 (pericen-
Isolation triolar material 1), which specifically labels cardiomyocyte nuclei
[9]. Combined with fluorescence-activated cell sorting (FACS),
this approach not only allows purification of all nuclei from hearts
for snATAC-seq but also separation of cardiomyocytes nuclei from
non-cardiomyocytes, which has advantages for some applications.
1. Sacrifice a mouse following approved procedures and dissect
the heart. Place the heart into ice-cold PBS buffer to remove
residual blood (see Note 1). Snap-freeze the heart in liquid
snATAC-seq of Individual Heart Cells 249
3.2 Brief Description The following protocol briefly describes the snATAC-seq library
of the Chromium Next preparation using the Chromium 10x™ kit and controller. For a
GEM Single-Cell ATAC detailed protocol, refer to the manufacturer’s instructions
Protocol [10, 11]. Here, we provide tips for library preparation from cardiac
nuclei.
1. The total number of nuclei isolated from whole murine hearts
is way too high for direct loading onto the Chromium 10×
snATAC-seq cartridge. The nuclei suspension should be
diluted. Depending on the efficiency of nuclei isolation, con-
secutive dilution steps may be necessary. The cartridge allows
250 Michail Yekelchyk et al.
3.3 NGS Library To check quality (QC) and size of the final snATAC-seq library,
Quality Assessment measure concentration and size distribution. We suggest to use the
Qubit ™ HS DNA kit for concentration measurements (normal
range for such NGS libraries is 1–50 ng/μL). Capillary electropho-
resis should be employed to determine size distribution of the
library (Agilent Bioanalyzer, Agilent Fragment-Analyzer, PerkinEl-
mer Lab-Chip, or similar). We do not recommend using agarose gel
electrophoresis, since its sensitivity and resolution are not sufficient
for this purpose. Often snATAC-seq libraries have a “wavy struc-
ture” of size distribution, reflecting the nucleosome pattern
(Fig. 1). Although a “wavy structure” is a good sign, it is not
mandatory before moving forward and may depend on the cell
type and state of chromatin (see Note 4).
3.4 Sequencing of Sequencing should be performed using the Illumina platform. For
the snATAC-seq example, Illumina NextSeq500 and “High-75” cartridges can be
Libraries used, which allows to obtain 400–500 M of sequencing reads,
accommodating one snATAC-seq library (up to 10k data points)
(see Note 5). The following setup should be used for NextSeq500
(paired-end sequencing; bp):
Read 1–34;
Read 2–34;
Index 1–8;
Index 2–16.
3.5 Description of 1. After sequencing, the acquired BCL output folder is used for
the Conventional demultiplexing of sequencing reads. Demultiplexing is done
Bioinformatical using the “cellranger-atac mkfastq” pipeline, which is part of
Analysis Steps the Cell Ranger ATAC analysis pipeline developed by
10xGenomics [11].
2. After the sequencing reads are demultiplexed, read mapping,
peak detection, and count are accomplished via the “cellranger-
atac count” pipeline (see Note 6).
3. (Optional) To combine multiple snATAC-seq runs, the “cell-
ranger-atac aggr” pipeline can be used. Note, that individual
runs should be first demultiplexed and counted individually (see
Note 7).
4. After the counting (and aggregating, if applicable) is done, we
recommend to continue the analysis in the R programming
environment. We suggest to use “Signac” and “Seurat” R
packages as a basis [12–14].
5. Counts and metadata are imported into R using the
“Read10x_h5” function. The metadata and the fragments
252 Michail Yekelchyk et al.
Fig. 1 Quality control of snATAC-seq libraries. A “wavy” pattern of the snATAC-seq library is a good indicator
of quality. A “wavy” pattern reflects nucleosome occupancy, which may vary among different cell types. In
general, a consistent “wavy” pattern is observed in cells with tight chromatin organization (e.g., various stem
cell populations). Some fully differentiated somatic cells may have a less well-defined, or even absent “wavy”
pattern
Fig. 2 Identification of different cell types in snATAC-seq datasets. (a) Heatmap showing chromatin accessi-
bility (“gene activity”) of top marker genes in different cell types of the heart. (b) The scatter plots overlay
marker gene accessibility (“gene activity”) in different colors over UMAP clustering (continuous scale)
snATAC-seq of Individual Heart Cells 253
Fig. 3 Visualization of differentially accessible genes in snATAC-seq datasets. (a) Knowledge about marker
genes accessibility profiles and their distribution across nuclei in UMAP clusters allows identification of
clusters corresponding to different cell types. (b) Example of DAG visualization as a color overlay over UMAP
clustering. (c) Visualization of chromatin accessibility at a gene of interest. Coverage plots are presented that
show distribution of raw snATAC-seq reads across the genomic region in different UMAP clusters. (d) GSEA of
DAGs between cardiomyocyte nuclei and all other nuclei (Reactome database)
files are read from the respective .csv and .tsv.gz files from the
output folder of “cellranger-atac count” (or “cellranger-atac
aggr” if applicable) pipeline.
6. The Seurat-class object is created using “CreateSeuratObject”
function of the Signac/Seurat packages. The respective peak
and gene annotations are acquired from the “EnsDb.Mmuscu-
lus.v79” (use the latest available version) package. The follow-
ing QC includes the detection of nucleosome signals,
transcription starting sites (TSS) enrichment, percentages of
reads in the peaks, and others [15]. Based on QC evaluation,
outliers are removed from the dataset.
254 Michail Yekelchyk et al.
4 Notes
Acknowledgments
References
A H
A549 cell..............................................216, 217, 219, 222 Heart................................ 4, 26, 246, 248, 249, 252–254
Array comparative genomic hybridization Heterogeneity................................... 54, 66, 71, 120, 202
(aCGH).................. 167–172, 177–181, 184, 185
I
B
Imaging.................................................68, 132, 133, 137,
Breast cancer (BC) .................................. 43–45, 102, 120 141, 204, 207–210, 215, 216, 220–222, 234
Immunoblot .................................................................. 202
C Immunofluorescence (IF)....................................... 44, 46,
Cell heterogeneity ................................................ 120, 202 48, 50, 66, 128, 130–136
Cell type................................ 7, 26, 28, 29, 40, 189, 197, Inductively coupled plasma-mass spectrometry
227, 228, 246, 250, 252, 253 (ICP-MS) ...... 215, 216, 218, 220–222, 224, 225
CellSearch ................................ 11–15, 17–21, 28, 30–33, In situ hybridization ............................................ 127–141
35, 36, 44, 65–67, 101, 168
L
Chromatin accessibility .......................245, 246, 252, 253
Chromium 10x™................................................. 202, 249 Laser ablation .............................................. 215, 216, 218
Circulating tumor cell (CTC) .................................11–15, Liquid biopsies ..................................................... 119, 202
17–19, 21, 25, 26, 28–30, 32, 36, 38, 40, 43, 44, LncRNAs ....................................128, 132, 138, 144, 190
46–50, 65–68, 84, 101–109, 111, 112, 116, Low-pass whole genome sequencing........................... 108
119–124, 167–169, 181, 190, 202
Circulating tumor cell (CTC) clusters ................ 101–117 M
Copy number alteration (CNA)......................... 102–104, Mass spectrometry ............................................... 215–225
108–111, 116, 167–186 Mechanical dissociation ................................................ 2, 8
CTC line culture .................................................. 120, 122 Mesenchymal phenotype ................................................ 44
Metal ..................................................................... 156–157
D
Metals ................................ 144–146, 152, 156, 161, 204
DEParray ........................................................66, 168, 169 Metastasis...........................................................43, 53, 54,
Dielectrophoresis (DEP) ................................................ 12 65, 71, 102, 119, 120
Dissociation ................................ 1–8, 54, 55, 57, 58, 213 Microarray hybridization .............................................. 171
Micromanipulation ...........................................48, 54, 55,
E 59, 60, 190, 193, 197
Electrophysiology................................................. 227–242 Microscopy ......................8, 56, 128, 132, 137, 151, 234
Epithelial-mesenchymal transition (EMT) ..............43, 44 Morphology..........................................34, 38, 58, 62, 66,
233, 234, 237, 242
F
N
Flow cytometry ...................................... 8, 120–122, 124,
149–151, 153, 155, 158, 160, 161, 163, 197 Next generation sequencing (NGS) ...........................109,
116, 246, 250
G
O
Genomic analysis ................................. 103–104, 106–109
Oxaliplatin ...........................................216, 217, 219, 222
Miodrag Gužvić (ed.), Single Cell Analysis: Methods and Protocols, Methods in Molecular Biology, vol. 2752,
https://doi.org/10.1007/978-1-0716-3621-3, © Springer Science+Business Media, LLC, part of Springer Nature 2024
259
SINGLE CELL ANALYSIS: METHODS AND PROTOCOLS
260 Index
P Single cell nucleus ATAC-seq.............................. 245–256
Single-cell RNA-sequencing (scRNA-seq) ........ 228, 242,
Patch clamp .......................................................... 227, 228 256
Patch-seq .............................................228, 229, 240, 242 Single-cell suspension .............................. 2, 5, 8, 57, 190,
PCR labeling ................................................112, 171–174 196, 203–205
Precision medicine ........................................................ 201 Single cell western blot ........................................ 201–213
Primary cells .................................................................. 1, 2 Single-molecule RNA fluorescence in situ hybridization
Proximity ligation assay for RNA (PLAYR) ....... 143–164 (smRNA FISH) ....................................... 128, 131,
Puncher......................................................................66–69 132, 134, 136–138
Stress granule (SG) .................... 127–129, 133, 138, 140
R
RNA ..................................................54, 71, 91, 143–164, T
189–197, 202, 227, 229–231, 233, 234, 237, Transcriptome analysis .................................................... 71
240, 242 Transcriptomic ................................................... 8, 48, 227
Transposase........................................................... 232, 246
S
Tumor fraction prediction............................................ 102
Sequencing .............................................................. 12, 66,
71, 72, 90, 91, 98, 102–104, 108–110, 116, 117, V
143, 189–197, 227–229, 232, 234, 237, 238, VyCAP .......................................................................66–68
240, 242, 250, 253
Single cell......................................... 2, 12, 48, 57, 66, 71, W
128, 143, 167, 190, 202, 227, 247
Single cell analysis ............... 8, 48, 66, 71, 181, 202, 216 Whole-cell recording..................................................... 227
Single cell clones .................................................. 120, 121
Z
Single cell isolation.................................. 12, 19, 193, 197
Single cell ICP-MS........................................................ 219 Zebrafish .......................................................53–57, 61, 62
Single cell multi-omics.................................................... 71