Simplifying Genomic Analysis
eBook
Published: September 12, 2024
Credit: Biotools
Pharmacogenomics research is key to the development of precision medicine, focusing on how individual genetic variations affect drug responses to inform clinical decisions.
Real-time PCR (RT-PCR) is widely used in pharmacogenomics research for its sensitivity and accuracy, but it faces challenges including high costs, scale-up issues and the need for constant assay updates.
This eBook explores how microfluidics-based PCR, which has the capability to run multiple reactions simultaneously, is transforming the field to enable more efficient and scalable genomic analysis.
Download this eBook to learn more about:
- The benefits of microfluidics-based PCR
- Considerations for cost-effective analysis in genomics-driven fields
- Challenges and perspectives in evaluating immune function and mechanisms of cell differentiation
MICROFLUIDICS
Simplifying
Genomic Analysis
With Microfluidics Technology
2
4 The Benefits of Microfluidics-Based PCR
Microfluidics-based PCR is revolutionizing how labs approach
genomics analysis.
8 When and Why to Automate
Benefits include improving lab efficiencies and cost savings
for years to come.
10 To Singleplex or Multiplex
What’s the difference?
12 Considerations for Cost-Effective Analysis
in Genomics-Driven Fields
Microfluidics technology empowers the generation of timely
and actionable answers that can be transformative.
16 Challenges and Perspectives in Evaluating Immune
Function and Mechanisms of Cell Differentiation
Experts explain what tools make the most sense and why
they chose what they did.
19 Resources
WHAT’S INSIDE
Contents
Now get the data of
96 plates with just
1
.
standardbio.com/x9
INTRODUCING THE NEW
HIGH-THROUGHPUT
GENOMICS SYSTEM
TM
THEY all want better, more reliable medicines.
YOU need to be confident in your research.
Pharmacogenomics
research simplified with
the power of microfluidics.
VS
96 PLATES
1 CHIP
4 5
The Benefits of
Microfluidics-Based PCR
Microfluidics-based PCR is revolutionizing
how labs approach genomics analysis.
Pharmacogenomics research is rapidly becoming
a critical component of precision medicine and
individualized drug therapy.1
Precision medicine
is built on individual variation in responses to
therapeutics and side effects, which can impact
and inform clinical decisions regarding treatment
strategies. Detecting changes in gene expression
that impact drug reactions at the molecular
level – pharmacogenomics – will be required
to implement such individualized drug therapy
approaches in the future.
Due to its sensitivity, classification accuracy, ease
of operation, and quick processing time, realtime PCR (RT-PCR) is the most popular approach
for pharmacogenomics research.1
Despite these
advantages, there remain some challenges to
using RT-PCR. Perhaps the biggest in the context
of pharmacogenomics research is scale-up and
the associated costs of prep time and reagent
use, which are significant already with smallscale operations. Additional challenges include
modifying assays to include newly discovered
biomarkers, improving data,
and standardizing processes.
Fortunately, there is a way to address these
challenges: microfluidics. Due to the small size of
the reactions they facilitate, microfluidics-based
protocols provide significant cost savings on
reagents and consumables. This capability to run
numerous nanoliter reactions at once allows for
more information to be collected from less precious
sample material. Microfluidics protocols are also
easier to standardize, improve reproducibility, and
require less human intervention, saving personnel
time. In this eBook, we explore how microfluidicsbased PCR is revolutionizing how labs approach
genomics analysis.
What is microfluidics?
Microfluidics is, essentially, the manipulation of
small volumes of fluids (nanoliters or less) within
networks of channels that are tens to hundreds of
microns in diameter. Microfluidic instruments have
low sample and reagent volume requirements and
short analysis times and can facilitate the type of
miniaturization that has led to the rise of lab-on-achip technologies.2 Microfluidic platforms are used
across a broad range of application areas including
pharmacogenomics, sample identification, and
agricultural genomics and are revolutionizing the
way research in these areas is carried out.
The benefits of microfluidics
for RT-PCR
Automated microfluidics-based protocols
yield several benefits for RT-PCR and NGS
library preparation, particularly when using
them for research applications that require
scalability, flexibility, and accuracy, such as
pharmacogenomics:
• Scalability (For example, the Standard BioTools™
microfluidics-based RT-PCR system requires
only about 2 µL of sample for 384 targets,
facilitating over 9,000 reactions at once.)
• Flexible, easy-to-adjust protocols and
experimental plans
• Small sample and reagent requirements,
preserving samples and reducing reagent costs
• Multiple integrated fluidic circuit (IFC) formats
to accommodate different combinations of
samples or assays with the ability to quickly
modify or add to an assay
• Ease of use and standardization capabilities
• Closed system that is reliable, eliminates
many individual pipetting steps, and has low
contamination risk
These benefits together increase the power
of real-time PCR based on microfluidics for
applications such as pharmacogenomics by
yielding more data and deeper insights with less
effort. In this eBook, we’ll dig deeper into some
of the critical advantages made possible through
microfluidics technology.
Less effort
Automation is a critical aspect of flexible, scalable,
and accurate microfluidics-based technologies.
Contrary to popular opinion, automation isn’t just
for labs processing thousands of samples regularly.
It yields several advantages for any lab, regardless
of size, such as improved reproducibility with, and
because of, less manual effort. In Chapter 2 of
this eBook, we discuss the benefits automation
provides to any lab, whether small- or large-scale,
and provide real-world examples of benefits
experienced by research groups utilizing Standard
BioTools microfluidics technology. Less time and
effort needed from personnel leads to significant
cost savings, which we also explore.
CHAPTER 1
6
More data and deeper insights
A major benefit provided by automated
microfluidics-based technologies is the deeper
insights they enable. Multiplexing has long been
used to extract more data per sample from
experiments, but it suffers from uneven amplification
across different targets, an issue that is addressed
using singleplex reactions. With microfluidics
technology, samples and reagents can be loaded
separately and then automatically combined in a
pairwise manner within the closed system to create
individual singleplex reactions in a multiplex format.
In Chapter 3 of this eBook, we dig deeper into
the benefits provided by singleplex simplicity at
multiplex throughput.
Concluding remarks
Pharmacogenomics research is a critical
research area enabling precision medicine and
personalized therapeutics. Doing it at scale is
the only way to identify the unique interactions
between host genetics and environment that
impact how individuals respond (or don’t) to
specific therapeutic interventions and schedules.
But scaling RT-PCR, the major biomolecular tool
enabling pharmacogenomics, has been
a challenge.
In this introduction, we provided a small sampling
of the efficiency, time and personnel savings,
scalability, and flexibility that microfluidics
technology can bring to RT-PCR. Download and
read the entire eBook to learn more about how
microfluidics technology can make your RTPCR protocols work harder and better for you,
facilitating safety and efficacy studies for improved
disease management.
References
1. Zheng, L-J. et al. “Clinical application and importance of
one-step human CYP2C19 genotype detection.” Journal of
International Medical Research 46 (2018): 4,965–73.
2. Tarn, M.D. and Pamme, N. “Microfluidics” chapter, Reference
Module in Chemistry, Molecular Sciences and Engineering.
Elsevier (2013):1–7.
Do More With Less | standardbio.com/x9
Deep insights with nanoscale genomics.
The only genomics system for real-time PCR and next-generation sequencing
library preparation to support discovery through screening.
THE NEW
HIGH-THROUGHPUT
GENOMICS SYSTEM
INCREASED
PRODUCTIVITY
Integrated reaction
setup for streamlined
workflows generates
up to 46,080
datapoints per
8-hour shift and up
to 384 barcoded
libraries per day.
DEEPER
INSIGHTS
Easily add or
remove assays and
simultaneously detect
up to 96 targets with
singleplex simplicity.
Design libraries for
dual coverage to
enhance performance.
OPTIMIZED
RESOURCES
Sustainably generate
up to 9,216 datapoints
with 96x savings
compared with
traditional methods.
EFFICIENT
OPERATION
Compact instrument
with walk-away
automation produces
PCR data in as little
as 2 hours and
NGS-ready libraries in
approximately 8 hours.
8 9
When and Why to Automate
Benefits include improving lab efficiencies
and cost savings for years to come.
By Steve Kain
To automate or not to automate
Over the past several years, I’ve worked with
researchers in a variety of lab environments. These
include both individual labs and genomics core
labs, both in academia and commercial settings.
When considering automation systems for
workflows such as genotyping or gene expression
by real-time PCR, the most common driver for
adoption is sample number. Researchers often feel
that unless they are routinely processing hundreds
to thousands of genomic samples, investments in
an automation system do not make sense.
Consideration of sample volumes is important,
and no doubt automation can save money at high
volumes of samples, but these are not the most
important reasons to consider automating your
genomics workflows.
First and foremost, automation leads to
more robust assay performance with
improved reproducibility.
The length of many genomics workflows requires
several small-volume pipetting steps, mixing,
incubations, and transfer steps. Each of these
steps can introduce considerable variance,
leading to questionable results and the need for
assay replicates to minimize variable findings.
When automation systems are employed, such
processing steps are standardized and lead to
less variability.
For example, one of the more commonly used
assays with Standard BioTools microfluidics
technology is genotyping performed on the
Biomark™ X system with a 96.96 Dynamic Array™.
This system can generate 9,216 datapoints from
96 samples and 96 individual PCR reactions
run in parallel. The equivalent experiment using
conventional systems requires preparing 96 x 96-
well plates.
Imagine the variability that could be introduced
between reactions due to manual processing at
this scale, not to mention the additional time and
costs incurred. In contrast, the Dynamic Array is a
closed system that automatically runs singleplex
reactions at the same time.
Many of the processing steps are fully automated
on Biomark X™, including thermocycling and data
acquisition. Because of the exacting standards
for the design and manufacture of the Biomark
X system components, you can be assured of
great reproducibility between samples, runs,
and laboratory operators. Definitely consider
sample volume when you think about automating
genomic workflows, but understand that the most
important reason is to improve the reproducibility
of your assays and hence the reliability of your
experimental results.
Invest in automation to save money
in the long run
For core lab directors, your most precious and
expensive resource likely leaves the lab at the end
of every day – your staff.
In order to reduce overhead and save money,
core labs should focus on a new metric popular
in startup companies. It’s known as revenue per
employee. If your average revenue per employee
is lower than your expense, then you can focus
on maximizing ways to increase that revenue
by implementing processes that make your
employees more productive. One such example
is to invest in automation solutions to get the most
out of your labor costs.
As one example, customers who have implemented
the Advanta™ RNA-Seq NGS Library Prep Kit
together with the Juno™ system can perform costefficient NGS library prep due to reduced reagent
expenses and lowered labor costs.
In comparison to standard manual processing,
the automated library prep method can save
almost $200,000 USD for the preparation of
5,000 RNA-seq libraries. In addition, the intuitive
interface and streamlined operations of Juno mean
labs are able to minimize time spent by staff in
preparing libraries.
Because the majority of the processing steps
are automated, the hands-on time for library
preparation is significantly reduced and requires
less-skilled operators. This latter benefit also helps
to ease the burden of training new people when
lab staff turns over. Lastly, by investing in quality
instrumentation such as Juno, you will have lab
automation that is built to last, thereby improving
lab efficiencies and cost savings for years to come.
About the author
Steve Kain, PhD, combines his scientific knowledge with
commercial experience in genomics, cell biology, drug
discovery, next-generation DNA sequencing, and botanical
science and testing services.
2
CHAPTER
10 11
To Singleplex or Multiplex
What’s the difference?
Several key factors must be considered when
designing a real-time PCR experiment, including
how many targets you intend to detect. Many
researchers opt for multiplex detection chemistries
due to its ability to provide more data per sample
with minimal added cost, but you need enough
targets for it to make sense. When getting ready
to start your real-time PCR experiment, one of the
first key decisions is assay design, and you need to
decide whether singleplex or multiplex detection
is the best fit. These two detection methods
involve different assay design and setup, each with
its own set of advantages and disadvantages.
Why multiplex
In multiplex PCR, multiple target sequences are
amplified in a single reaction. In real-time PCR,
these target sequences are commonly detected
using probes that have different dye labels.
Multiplex assays are attractive because they can
help you save time, cost, and most importantly,
sample. However, multiplexing has its drawbacks.
Primer design plays a crucial role in multiplex
success, and often multiple rounds of optimization
are needed to determine an appropriate primer
concentration. Primers and unintended PCR
products or artifacts may compete in amplification,
resulting in uneven amplification for different
targets. In fact, it’s common practice to perform
singleplex PCR in order to amplify loci that
multiplexing has failed to amplify.
Once optimized, multiplex assays can also be
difficult to modify. Given the time and effort
required to design a successful assay with a
particular combination of compatible primer pairs,
redesigning easily becomes a headache. Changes
in routine assays are inevitable to advance any
system, whether for screening or testing, but the
ability to adapt quickly to these changes is not
afforded with multiplexed reactions.
Why singleplex
Singleplex PCR is easier than multiplexing. Only
one target is amplified per reaction, so your assay
is easier to design and implement due to the
absence of potential competition during PCR.
However, many researchers find singleplexing
limiting, since using several reactions to detect
multiple targets can mean higher costs in materials
and labor and more sample used per reaction.
The ability to scale an assay is also challenging
when using singleplex reactions. Since each
reaction must be isolated in an individual well,
you can only scale to the number of reactions
often done in 96- or 384-well plates that can be
completed in a day. And when you consider the
need for more reagents every time you add more
reactions, cost can quickly become an issue.
Multiplex with singleplex
simplicity with Standard BioTools
microfluidics technology
Standard BioTools microfluidics technology
assembles your PCR reactions for you in an
automated, miniaturized fashion. The integrated
fluidic circuits (IFCs) have nanoscale reaction
volumes, so you use less of your precious sample
and reagent. Samples and assays are loaded
into the IFC separately and then automatically
combined in a pairwise manner within the closed
system of the IFC to create individual singleplex
reactions, with up to 192 singleplex reactions
per sample (depending on IFC format). You can
add, remove, or replace assays on demand and
scale throughput without changing technologies,
allowing up to 96 individual samples and controls
to be interrogated with up to 96 individual assays
with the same dye label, for a total of up to 9,216
individual reactions per run. Think of it as multiplex
throughput with singleplex simplicity.
Learn more about microfluidics technology >
3
CHAPTER
12 13
The end result of any genomic investigation is
discovering the role that genetics plays in health,
disease, and potential treatment. In order for basic,
translational, and clinical researchers to learn about
differences and changes in an individual’s genetic
makeup, they must identify, measure, and compare
genomic features or regulatory and functional
elements within the genome.
While there are a variety of technologies and
tools used in genomic analysis, progress and
improvement in high-throughput and cost-effective
methods are inconsistent depending on the
application and the accuracy of data needed.
Get big answers with
micro-sized technology
Comprehensive and versatile genomic analysis
with microfluidics technology empowers the
generation of timely and actionable answers
that can transform all genomics studies.
Proven to enable high-complexity analysis with
unprecedented proficiency, microfluidics-based
solutions streamline workflows for applications
demanding sensitivity and broad range,
including genotyping, gene expression, sample
identification, copy number variation, and NGS
library preparation.
Simplifying workflows through nanoscale
automation maximizes efficiency and provides
the flexibility to scale projects with increased
data output. This allows you to adjust your
experimental plan to match your needs and
interests with microfluidics-based PCR and NGS
library preparation.
Here are just some of the areas in which the
life sciences community is using microfluidics
technology to accelerate and advance
genomics knowledge.
The promise of personalized
medicine with pharmacogenomics
Drug development and efficacy testing can be
improved by more comprehensively understanding
the effects of drugs and their mechanism
of action. Pharmacogenomic research and
pharmacodynamic studies are a key element of
personalized and precision medicine, significant in
understanding response to medications.
Research has shown that a pharmacogenomic
profile can be used to guide care and reduce
the incidence of adverse drug reactions. These
studies can be accelerated using microfluidicsbased real-time PCR to test multiple targets with
multiple interactions at once. This provides a more
complete overview of how a target molecule
functions when binding to an active site on an
enzyme or interacting with cell surface signaling
proteins that can disrupt downstream signaling.
Microfluidics technology facilitates
pharmacogenomics studies with a single highthroughput workflow that consolidates several
assays into one experiment without the need for
multiplexing. This allows labs to customize content,
avoiding the limitation of fixed-format panels, and
cut sample and reagent use by 100x.
Case study
In a 2021 paper, a group from the University of
Minnesota describes developing and implementing
a cost-effective in-house pharmacogenomic
testing research program at a major academic
health system. Pharmacogenomics testing in
clinical research presents a major opportunity
for improving therapeutic outcomes and could
become a significant part of precision medicine.
However, this value is limited by successful
implementation of actionable testing.
This work outlines the development and technical
validation of an in-house SNP targeting multiplex
PCR-based assay on the 96.96 Dynamic Array
used with a Standard BioTools IFC Controller HX,
FC1™ cycler, and Biomark HD.
Principles and methods of sample
identification to support integrative
genomic analysis
Single-nucleotide polymorphism (SNP) genotyping
methods are broadly used for sample identification
and quality control in a biobank facility, core lab, or
sequencing center. Genotyping with a dedicated
set of SNP assays for genetic sex determination,
DNA quality assessment, or a SNP fingerprint can
confirm sample integrity and origin.
Biobanks and genomics centers must overcome
several challenges to ensure that they provide
the highest-quality, correct samples for research
projects. Sample mix-ups, contamination, or
handling errors can occur before or after samples
enter the molecular laboratory or storage
facility. Processing misidentified, poor-quality,
or contaminated samples may lead to incorrect
interpretation of results.
Implementing a standard genotyping workflow to
confirm the identity and quality of each sample
before analysis represents an ideal solution to
maximize the integrity of study results.
The Advanta
Pharmacogenomics Assay:
• Supports extraction-free and extracted
buccal swabs
• Uses a single workflow for SNPs
and CNVs
• Contains a core panel of actionable
targets with flexibility to add
custom content
• Combines analysis software with
interpreted results
Considerations for
Cost-Effective Analysis
in Genomics-Driven Fields
Microfluidics technology empowers the generation of timely
and actionable answers that can be transformative.
4
CHAPTER
14 15
Microfluidics-based PCR offers the quality needed
for confidence in sample identification data.
The Advanta Sample ID Genotyping Panel is a
96-SNP assay enabling laboratories to generate
a sample-specific genetic fingerprint and quality
assessment from research specimens throughout
the sample journey.
Cancer genomics for
precision oncology
Personalized approaches for cancer treatments
are challenged by the individual nature of cancer
itself, presenting differently in each patient.
The identification of predictive biomarkers and
signatures could help direct which patients would
benefit from a therapy or accurately assess the risk
of recurrence and progression.
Microfluidics-based gene-expression assays can
aid in the research for therapy decision-making
by identifying suitable biomarker candidates. The
capability to scale this kind of discovery while
reducing the volume of reactions supports the
active search for effective biomarkers without the
barriers of cost and time.
Case study
In a 2022 paper in OncoImmunology, “Innate
lymphoid cells: NK and cytotoxic ILC3 subsets
infiltrate metastatic breast cancer lymph nodes,”
Institut de Recherche Saint-Louis scientists
investigated the impact of the local tumor
environment on innate immune response in the
lymph nodes of breast cancer patients. Innate
lymphoid cells (ILCs), including cytotoxic natural
killer (NK) cells and helper-type ILCs, are important
regulators of tissue immune homeostasis.
Through flow cytometry, cytokine release assays,
and qPCR (using Juno and Biomark HD), they
characterized the ILC populations that were
infiltrating lymph nodes. Data showed that the
local tumor microenvironment inhibited NK cell
functions, but cytokine stimulation restored their
functionality. Results suggest a consideration of
combination immunotherapies
for improved efficacy.
References
1. Mroz, P. et al. “Development and implementation of
in-house pharmacogenomic testing program at a major
academic health system.” Frontiers in Genetics 12 (2021):
712602.
2. Rethacker, L. et al. “Innate lymphoid cells: NK and cytotoxic
ILC3 subsets infiltrate metastatic breast cancer lymph
nodes.” OncoImmunology 11 (2022): 2057396.
What is molecular
sample identification?
This proactive sample identification
system is also known as sample ID
or DNA fingerprinting. Panels of DNA
markers are analyzed for each sample,
generating a unique genotype or
fingerprint. The genotype obtained is an
indelible, nontransferable identifier for
the sample. Carefully selected markers
can provide additional information,
such as assessment of sample
quality, genetic gender identity, and
population prediction.
How is it used?
DNA samples can be fingerprinted
(identified) prior to distribution.
Sample identity can be confirmed.
• Compare genetic sex to reported sex.
• Compare genotypes from distribution
sample to accession genotypes.
Low-quality DNA issues can be identified.
• Low genotype calling rate
• Sample contamination
As emerging therapies reveal new biomarkers
and expand the need for samples, the costs
and labor required to complete this important
research work also rise. The Advanta IO Gene
Expression Assay, used with the Biomark systems,
was developed in collaboration with leading
researchers in the biopharmaceutical industry to
provide the right balance of biomarker breadth,
assay flexibility, and workflow efficiency, providing
a reliable, sensitive, and cost-effective toolset
for identifying gene-expression signatures from
immune and cancer cells.
The panel consists of 170 genes, including
markers for:
• Immune cell identification
• Immune and cancer cell function
• Immune regulation and cell fate
• Checkpoint therapy response
Optimized for formalin-fixed, paraffin-embedded
and fresh frozen tumor samples, the assay uses
TaqMan® chemistry to sensitively measure gene
expression, with five reference genes serving as
analysis controls. Each reaction is miniaturized
to nanoliter volume and controlled using precise
automation to empower accurate and costeffective qPCR analysis across a large dynamic
range. Since assay introduction into the IFC is
under user control, researchers also have the
flexibility to add up to
17 new assays or exchange gene assays within
the panel to achieve experimental goals – all
without affecting the original panel content,
protocol, or workflow.
16 17
When designing a study, many factors must be
considered that, when put together, empower the
generation of valuable data that can be turned into
actionable insights.
A well-designed study can provide the framework
to ensure scientific integrity and credibility of data.
We’ve talked with a group of experts in the fields
of infectious disease, immunology, and molecular
genetics about their philosophies on how to
approach a research question, what tools make the
most sense, and why they chose what they did.
When you thought about designing
your research study on the immune
response to infection, how were you
able to test so many samples within a
set timeframe and budget?
Leisha McGrath, a PhD student at the Marine and Freshwater
Research Centre in Ireland, focused her graduate research
on the presence of antimicrobial peptides in Atlantic salmon.
Knowledge of the initial immune response in fish infected
with amoebic gill disease could potentially offer a method to
detect onset as climate change increases risk of disease.
The system we used was very beneficial for us, and
the setup was ideal – very straightforward, very
easy to follow up, and much faster than we had
been expecting. We actually started setting up an
overwhelming number of qPCR reactions manually
in tubes, working out the number of tissues we
needed to test, how many replicates would be
ideal for this type of experiment, and what the cost
would be in running so many reactions.
The center had just installed a Biomark HD system,
and the team was told they would be able to do
all their reactions on one microfluidic array instead
of multiples of tubes. This was very appealing. We
thought that would be a much better and simpler
approach. Plus, we didn’t have to go back and
do multiple rounds of qPCR, which in itself would
introduce more variation and more issues.
I suppose it’s difficult to visualize it before you
see the system work for yourself. But once you
perform the experiment and get the readout, you
understand that compared to a standard reaction
volume or standard tube, it is such an advantage to
be able to stretch everything out a bit further.
I think what we’re showing with this study is that
the Biomark system enables simple setup across a
range of experiments, empowering you to screen
bigger populations detection, and study different
infections or even support vaccine research or
understanding response to treatments. You could
do it all.
How did you choose the right
approach to RNA analysis in your
hypoxia-on-a-chip research, given
the small size of your experimental
platform and minimal cellular
material obtained?
Scott Magness, PhD, is an Associate Professor and the
Founder and Director of the Center for Gastrointestinal
Biology and Disease Advanced Analytics Core at the
University of North Carolina School of Medicine and is
interested in understanding the genetic mechanisms that
control stem cell maintenance and differentiation.
For us, the Advanta RNA-seq workflow was vital to
our studies because we were challenged with only
being able to retrieve a small number of cells from
our model for subsequent analysis. The culture
chamber in the gut-on-a-chip system is very small
– the entire five-well platform is smaller than a
microscope slide – and we only get about 50,000
cells that generate a confluent monolayer. This
translates to small amounts of cellular material and
small amounts of harvested total RNA to use in the
library prep protocol.
Conventional RNA-seq systems simply cannot
achieve what we need on this system. Also, in
order to perform all technical and biological
replicates necessary for these experiments,
conventional systems become very expensive. The
Advanta system was a game changer, enabling
us to use these microdevices and characterize as
many transcriptional responses as we can while
managing costs.
Our data quality from our samples with low cell
numbers was excellent, providing a really nice
overview of what was happening to these stem
cells during a hypoxic event. For example, we
obtained >30 million reads per sample, 98.5%
aligned to the human genome reference and
80.2% aligned to the transcriptome reference. This
level of clean data helped us immensely in pulling
out differential gene-expression patterns that we
could use to generate hypotheses for how the
cells were responding to hypoxia over time.
Read Scott’s interview to see how he developed a
hypoxia-on-a-chip environment.
Why is scalability and cost-efficiency
so important when DNA fingerprinting
samples at the genetics biorepository?
Kelly Nudelman, PhD, Assistant Research Professor,
and Tae-Hwi (Linus) Schwantes-An, PhD, Assistant
Research Professor, highlight the importance of sample
identification and quality control at the Indiana University
Genetics Biobank.
One of our goals is to ensure that all of our
samples have been quality-controlled to the
highest standard. Using DNA fingerprinting to
assess samples prior to accession and distribution
can save time and resources. Researchers who
receive samples from the biorepository need to
have confidence that the samples they receive
have been correctly identified from the sample
source and will yield interpretable data from their
planned downstream analyses.
Challenges and Perspectives
in Evaluating Immune
Function and Mechanisms
of Cell Differentiation
5
CHAPTER
18 19
The Standard BioTools workflow was chosen
because of its scalability and cost-efficiency and
the small quantity of DNA needed to test samples.
We found that many alternative workflows are
either high-throughput/high-cost or low-throughput/
low-cost. What makes the Standard BioTools
microfluidics-based workflow unique is that it is
high-throughput at a cost that is scalable to our
process. It saves our customers and our lab time
and resources.
For example, as the field of neurodegenerative
disease grows, new targets of interest will be
identified. Being able to customize the assay
allows us to adapt to how the field is changing, and
this is something we can do using the Standard
BioTools workflow. Let’s say a new variant is
discovered, and everyone wants to know the
status for that particular variant in her or his
sample, we’ll be able to easily make a modification
to our panel, adding the new variant quickly.
What can you reveal with single-cell
versus bulk analysis and how can
you obtain the level of sensitivity
needed to detect a wider range of
gene expression?
Christophe Lancrin, PhD, Research Group Leader at the
European Molecular Biology Laboratory, is focused on
discovering an intermediate cell population with endothelial
and hematopoietic characteristics co-expressing seven
essential transcription factors at the single-cell level using
C1™ and Biomark HD.
Single-cell analysis is powerful. When you work
in bulk, you miss information. Working with single
cells is definitely a big plus but you need to use the
right technology for the right question. Although
bulk transcriptomics can reveal crucial overall gene
correlations between semi-stable cellular states, it
cannot resolve subtler gene interactions occurring
in complex transitional states. In addition, using a
bulk approach makes it difficult to infer the direct
consequences on the transcriptional landscape
upon which these transcription factors are acting.
These limitations can be overcome by the use of
single-cell approaches.
We needed the C1 full-length mRNA sequencing
technology because of its sensitivity. Not only were
we detecting transcription factors, but we also had
to detect a range of gene-expression levels. We
required higher-quality sequencing data to perform
these challenging analyses.
Become an Expert: SPARK E-Seminar Series
Genomics workflows and applications for
the new Standard BioTools Biomark X
A Single Workflow for Genotyping and
Copy Number Variation in Pharmacogenomic
Testing Application Note
Standard BioTools Microfluidics Videos The PCR Advantage: How the Biomark
HD System Enhances the TruGraf GeneExpression Test for Kidney Rejection
Brochure: X9 High-Throughput
Genomics System
Resources
6
CHAPTER
Unleashing tools to accelerate
breakthroughs in human health™
For Research Use Only. Not for use in diagnostic procedures.
Information in this publication is subject to change without notice. Patent and License Information: www.standardbio.com/legal/notices. Trademarks: Standard BioTools, the
Standard BioTools logo, Fluidigm, the Fluidigm logo, "Unleashing tools to accelerate breakthroughs in human health", Advanta, Biomark, Biomark X, C1, D3, Dynamic Array, FC1,
Juno and X9 are trademarks and/or registered trademarks of Standard BioTools Inc. (f.k.a. Fluidigm Corporation) or its affiliates in the United States and/or other countries. All
other trademarks are the sole property of their respective owners. ©2023 Standard BioTools Inc. All rights reserved. 04/2023
FLDM-01132 Rev 01
SALES
North America | +1 650 266 6170 | info-us@standardbio.com
Europe/Middle East/Africa/Russia | +33 1 60 92 42 40 | info-europe@standardbio.com
Latin America | +1 650 266 6170 | info-latinamerica@standardbio.com
Japan | +81 3 3662 2150 | info-japan@standardbio.com
China (excluding Hong Kong/Macau) | +86 21 3255 8368 | info-china@standardbio.com
All other Asia-Pacific countries/India/Australia | +1 650 266 6170 | info-asia@standardbio.com
CORPORATE HEADQUARTERS
2 Tower Place, Suite 2000
South San Francisco, CA 94080 USA
Toll-free: 866 359 4354 in the US and Canada
Fax: 650 871 7152
standardbio.com
We are dedicated to supporting your research. At Standard BioTools, we meticulously
work to improve and update our product offerings to better serve your needs.
Brought to you by
Download the eBook for FREE Now!
Information you provide will be shared with the sponsors for this content.
Technology Networks or its sponsors may contact you to offer you content or products based on your interest in this topic. You may opt-out at any time.
Experiencing issues viewing the form? Click here to access an alternate version